1
|
Nguyen TNA, Huang PS, Chu PY, Hsieh CH, Wu MH. Recent Progress in Enhanced Cancer Diagnosis, Prognosis, and Monitoring Using a Combined Analysis of the Number of Circulating Tumor Cells (CTCs) and Other Clinical Parameters. Cancers (Basel) 2023; 15:5372. [PMID: 38001632 PMCID: PMC10670359 DOI: 10.3390/cancers15225372] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/05/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Analysis of circulating tumor cells (CTCs) holds promise to diagnose cancer or monitor its development. Among the methods, counting CTC numbers in blood samples could be the simplest way to implement it. Nevertheless, its clinical utility has not yet been fully accepted. The reasons could be due to the rarity and heterogeneity of CTCs in blood samples that could lead to misleading results from assays only based on single CTC counts. To address this issue, a feasible direction is to combine the CTC counts with other clinical data for analysis. Recent studies have demonstrated the use of this new strategy for early detection and prognosis evaluation of cancers, or even for the distinguishment of cancers with different stages. Overall, this approach could pave a new path to improve the technical problems in the clinical applications of CTC counting techniques. In this review, the information relevant to CTCs, including their characteristics, clinical use of CTC counting, and technologies for CTC enrichment, were first introduced. This was followed by discussing the challenges and new perspectives of CTC counting techniques for clinical applications. Finally, the advantages and the recent progress in combining CTC counts with other clinical parameters for clinical applications have been discussed.
Collapse
Affiliation(s)
- Thi Ngoc Anh Nguyen
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan City 33302, Taiwan; (T.N.A.N.); (P.-S.H.); (P.-Y.C.)
| | - Po-Shuan Huang
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan City 33302, Taiwan; (T.N.A.N.); (P.-S.H.); (P.-Y.C.)
| | - Po-Yu Chu
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan City 33302, Taiwan; (T.N.A.N.); (P.-S.H.); (P.-Y.C.)
| | - Chia-Hsun Hsieh
- Division of Hematology-Oncology, Department of Internal Medicine, New Taipei City Municipal TuCheng Hospital, New Taipei City 23652, Taiwan;
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33302, Taiwan
| | - Min-Hsien Wu
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan City 33302, Taiwan; (T.N.A.N.); (P.-S.H.); (P.-Y.C.)
- Division of Hematology-Oncology, Department of Internal Medicine, New Taipei City Municipal TuCheng Hospital, New Taipei City 23652, Taiwan;
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33302, Taiwan
| |
Collapse
|
2
|
Clack K, Soda N, Kasetsirikul S, Mahmudunnabi RG, Nguyen NT, Shiddiky MJA. Toward Personalized Nanomedicine: The Critical Evaluation of Micro and Nanodevices for Cancer Biomarker Analysis in Liquid Biopsy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205856. [PMID: 36631277 DOI: 10.1002/smll.202205856] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/20/2022] [Indexed: 06/17/2023]
Abstract
Liquid biopsy for the analysis of circulating cancer biomarkers (CBs) is a major advancement toward the early detection of cancer. In comparison to tissue biopsy techniques, liquid biopsy is relatively painless, offering multiple sampling opportunities across easily accessible bodily fluids such as blood, urine, and saliva. Liquid biopsy is also relatively inexpensive and simple, avoiding the requirement for specialized laboratory equipment or trained medical staff. Major advances in the field of liquid biopsy are attributed largely to developments in nanotechnology and microfabrication that enables the creation of highly precise chip-based platforms. These devices can overcome detection limitations of an individual biomarker by detecting multiple markers simultaneously on the same chip, or by featuring integrated and combined target separation techniques. In this review, the major advances in the field of portable and semi-portable micro, nano, and multiplexed platforms for CB detection for the early diagnosis of cancer are highlighted. A comparative discussion is also provided, noting merits and drawbacks of the platforms, especially in terms of portability. Finally, key challenges toward device portability and possible solutions, as well as discussing the future direction of the field are highlighted.
Collapse
Affiliation(s)
- Kimberley Clack
- School of Environment and Science (ESC), Griffith University, Nathan Campus, Nathan, QLD, 4111, Australia
- Queensland Micro and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD, 4111, Australia
| | - Narshone Soda
- Queensland Micro and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD, 4111, Australia
| | - Surasak Kasetsirikul
- Queensland Micro and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD, 4111, Australia
| | - Rabbee G Mahmudunnabi
- School of Environment and Science (ESC), Griffith University, Nathan Campus, Nathan, QLD, 4111, Australia
- Queensland Micro and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD, 4111, Australia
| | - Nam-Trung Nguyen
- Queensland Micro and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD, 4111, Australia
| | - Muhammad J A Shiddiky
- School of Environment and Science (ESC), Griffith University, Nathan Campus, Nathan, QLD, 4111, Australia
- Queensland Micro and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD, 4111, Australia
| |
Collapse
|
3
|
Zhu J, Tan Z, Zhang J, An M, Khaykin VM, Cuneo KC, Parikh ND, Lubman DM. Sequential Method for Analysis of CTCs and Exosomes from the Same Sample of Patient Blood. ACS OMEGA 2022; 7:37581-37588. [PMID: 36312392 PMCID: PMC9609053 DOI: 10.1021/acsomega.2c04428] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/29/2022] [Indexed: 06/16/2023]
Abstract
Circulating tumor cells (CTCs) and exosomes, both released from the primary tumor into peripheral blood, are a promising source of cancer biomarkers. They are detectable in the blood and carry a large diversity of biological molecules, which can be used for the diagnosis and monitoring of minimally invasive cancers. However, due to their intrinsic differences in counts, size, and molecular contents, studies have focused on only one type of vesicle. Herein, we have developed an integrated system to sequentially isolate CTCs and exosomes from a single patient blood sample for further profiling and analysis. The CTCs are isolated using a commercial filtration method and then the remaining blood is processed using multiple cycles of ultracentrifugation to isolate the exosomes. The method uses two available technologies where the eluent from CTC isolation is usually discarded and interfaces them, so that the eluent can be interfaced to exosome isolation methods. The CTCs are identified based on fluorescence staining of their surface markers, while the exosomes are analyzed using transmission electron microscopy, nanosight tracking analysis, and mass spec proteomic analysis. This analysis showed CTCs detected by their surface markers for metastatic hepatocellular carcinoma (HCC), while essentially none were detected for cirrhosis. The exosome analysis resulted in the identification of ∼500-1000 exosome proteins per sample confirmed by detection of exosome surface markers CD9, CD63, CD81, and TSG101 in addition to proteins related to cancer progression. Proteins enriched in HCC exosomes were shown to be involved in the immune response, metastasis, and proliferation.
Collapse
Affiliation(s)
- Jianhui Zhu
- Department
of Surgery, The University of Michigan, 1150 West Medical Center Drive, Ann Arbor, Michigan 48109, United States
| | - Zhijing Tan
- Department
of Surgery, The University of Michigan, 1150 West Medical Center Drive, Ann Arbor, Michigan 48109, United States
| | - Jie Zhang
- Department
of Surgery, The University of Michigan, 1150 West Medical Center Drive, Ann Arbor, Michigan 48109, United States
| | - Mingrui An
- Department
of Surgery, The University of Michigan, 1150 West Medical Center Drive, Ann Arbor, Michigan 48109, United States
| | - Valerie M. Khaykin
- Division
of Gastroenterology and Hepatology, University
of Michigan Medical Center, Ann
Arbor, Michigan 48109, United States
| | - Kyle C. Cuneo
- Department
of Radiation Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Neehar D. Parikh
- Division
of Gastroenterology and Hepatology, University
of Michigan Medical Center, Ann
Arbor, Michigan 48109, United States
| | - David M. Lubman
- Department
of Surgery, The University of Michigan, 1150 West Medical Center Drive, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
4
|
Iovanna J. Implementing biological markers as a tool to guide clinical care of patients with pancreatic cancer. Transl Oncol 2020; 14:100965. [PMID: 33248412 PMCID: PMC7704461 DOI: 10.1016/j.tranon.2020.100965] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/04/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
A major obstacle for the effective treatment of PDAC is its molecular heterogeneity. Stratification of PDAC using markers highly specific, reproducible, sensitive, easily measurable and inexpensive is necessary. At the early stages, clinician’s priority lies in rapid diagnosis, so that the patient receives surgery without delay. At advanced disease stages, priority is to determine the tumor subtype and select a suitable effective treatment.
A major obstacle for the effective treatment of pancreatic ductal adenocarcinoma (PDAC) is its molecular heterogeneity, reflected by the diverse clinical outcomes and responses to therapies that occur. The tumors of patients with PDAC must therefore be closely examined and classified before treatment initiation in order to predict the natural evolution of the disease and the response to therapy. To stratify patients, it is absolutely necessary to identify biological markers that are highly specific and reproducible, and easily measurable by inexpensive sensitive techniques. Several promising strategies to find biomarkers are already available or under development, such as the use of liquid biopsies to detect circulating tumor cells, circulating free DNA, methylated DNA, circulating RNA, and exosomes and extracellular vesicles, as well as immunological markers and molecular markers. Such biomarkers are capable of classifying patients with PDAC and predicting their therapeutic sensitivity. Interestingly, developing chemograms using primary cell lines or organoids and analyzing the resulting high-throughput data via artificial intelligence would be highly beneficial to patients. How can exploiting these biomarkers benefit patients with resectable, borderline resectable, locally advanced, and metastatic PDAC? In fact, the utility of these biomarkers depends on the patient's clinical situation. At the early stages of the disease, the clinician's priority lies in rapid diagnosis, so that the patient receives surgery without delay; at advanced disease stages, where therapeutic possibilities are severely limited, the priority is to determine the PDAC tumor subtype so as to estimate the clinical outcome and select a suitable effective treatment.
Collapse
Affiliation(s)
- Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, 163 Avenue de Luminy, 13288 Marseille, France.
| |
Collapse
|
5
|
Rivera-Báez L, Lohse I, Lin E, Raghavan S, Owen S, Harouaka R, Herman K, Mehta G, Lawrence TS, Morgan MA, Cuneo KC, Nagrath S. Expansion of Circulating Tumor Cells from Patients with Locally Advanced Pancreatic Cancer Enable Patient Derived Xenografts and Functional Studies for Personalized Medicine. Cancers (Basel) 2020; 12:cancers12041011. [PMID: 32326109 PMCID: PMC7225920 DOI: 10.3390/cancers12041011] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/03/2020] [Accepted: 04/14/2020] [Indexed: 12/22/2022] Open
Abstract
Improvement in pancreatic cancer treatment represents an urgent medical goal that has been hampered by the lack of predictive biomarkers. Circulating Tumor Cells (CTCs) may be able to overcome this issue by allowing the monitoring of therapeutic response and tumor aggressiveness through ex vivo expansion. The successful expansion of CTCs is challenging, due to their low numbers in blood and the high abundance of blood cells. Here, we explored the utility of pancreatic CTC cultures as a preclinical model for treatment response. CTCs were isolated from ten patients with locally advanced pancreatic cancer using the Labyrinth, a biomarker independent, size based, inertial microfluidic separation device. Three patient-derived CTC samples were successfully expanded in adherent and spheroid cultures. Molecular and functional characterization was performed on the expanded CTC lines. CTC lines exhibited KRAS mutations, consistent with pancreatic cancers. Additionally, we evaluated take rate and metastatic potential in vivo and examined the utility of CTC lines for cytotoxicity assays. Patient derived expanded CTCs successfully generated patient derived xenograft (PDX) models with a 100% take rate. Our results demonstrate that CTC cultures are possible and provide a valuable resource for translational pancreatic cancer research, while also providing meaningful insight into the development of distant metastasis, as well as treatment resistance.
Collapse
Affiliation(s)
- Lianette Rivera-Báez
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48105, USA; (L.R.-B.); (E.L.); (S.O.)
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48105, USA;
| | - Ines Lohse
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (I.L.); (K.H.); (M.A.M.)
| | - Eric Lin
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48105, USA; (L.R.-B.); (E.L.); (S.O.)
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48105, USA;
| | - Shreya Raghavan
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Sarah Owen
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48105, USA; (L.R.-B.); (E.L.); (S.O.)
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48105, USA;
| | - Ramdane Harouaka
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109, USA; (R.H.); (T.S.L.)
| | - Kirk Herman
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (I.L.); (K.H.); (M.A.M.)
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109, USA; (R.H.); (T.S.L.)
| | - Geeta Mehta
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48105, USA;
| | - Theodore S. Lawrence
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109, USA; (R.H.); (T.S.L.)
| | - Meredith A. Morgan
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (I.L.); (K.H.); (M.A.M.)
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109, USA; (R.H.); (T.S.L.)
| | - Kyle C. Cuneo
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (I.L.); (K.H.); (M.A.M.)
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109, USA; (R.H.); (T.S.L.)
- Veterans Administration Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
- Correspondence: (K.C.C.); (S.N.)
| | - Sunitha Nagrath
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48105, USA; (L.R.-B.); (E.L.); (S.O.)
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48105, USA;
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109, USA; (R.H.); (T.S.L.)
- Correspondence: (K.C.C.); (S.N.)
| |
Collapse
|
6
|
Chen S, Tauber G, Langsenlehner T, Schmölzer LM, Pötscher M, Riethdorf S, Kuske A, Leitinger G, Kashofer K, Czyż ZT, Polzer B, Pantel K, Sedlmayr P, Kroneis T, El-Heliebi A. In Vivo Detection of Circulating Tumor Cells in High-Risk Non-Metastatic Prostate Cancer Patients Undergoing Radiotherapy. Cancers (Basel) 2019; 11:E933. [PMID: 31277254 PMCID: PMC6678903 DOI: 10.3390/cancers11070933] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 06/20/2019] [Accepted: 06/28/2019] [Indexed: 12/13/2022] Open
Abstract
High-risk non-metastatic prostate cancer (PCa) has the potential to progress into lethal disease. Treatment options are manifold but, given a lack of surrogate biomarkers, it remains unclear which treatment offers the best results. Several studies have reported circulating tumor cells (CTCs) to be a prognostic biomarker in metastatic PCa. However, few reports on CTCs in high-risk non-metastatic PCa are available. Herein, we evaluated CTC detection in high-risk non-metastatic PCa patients using the in vivo CellCollector CANCER01 (DC01) and CellSearch system. CTC counts were analyzed and compared before and after radiotherapy (two sampling time points) in 51 high-risk non-metastatic PCa patients and were further compared according to isolation technique; further, CTC counts were correlated to clinical features. Use of DC01 resulted in a significantly higher percentage of CTC-positive samples compared to CellSearch (33.7% vs. 18.6%; p = 0.024) and yielded significantly higher CTC numbers (range: 0-15 vs. 0-5; p = 0.006). Matched pair analysis of samples between two sampling time points showed no difference in CTC counts determined by both techniques. CTC counts were not correlated with clinicopathological features. In vivo enrichment using DC01 has the potential to detect CTC at a higher efficiency compared to CellSearch, suggesting that CTC is a suitable biomarker in high-risk non-metastatic PCa.
Collapse
Affiliation(s)
- Shukun Chen
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Gerlinde Tauber
- Department of Therapeutic Radiology and Oncology, Medical University of Graz, 8036 Graz, Austria
| | - Tanja Langsenlehner
- Department of Therapeutic Radiology and Oncology, Medical University of Graz, 8036 Graz, Austria
| | - Linda Maria Schmölzer
- Department of Therapeutic Radiology and Oncology, Medical University of Graz, 8036 Graz, Austria
| | - Michaela Pötscher
- Department of Therapeutic Radiology and Oncology, Medical University of Graz, 8036 Graz, Austria
| | - Sabine Riethdorf
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Andra Kuske
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Gerd Leitinger
- Department of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Karl Kashofer
- Diagnostic and Research Institute of Pathology, Medical University Graz, 8036 Graz, Austria
| | - Zbigniew T Czyż
- Division Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, 93053 Regensburg, Germany
| | - Bernhard Polzer
- Division Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, 93053 Regensburg, Germany
| | - Klaus Pantel
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Peter Sedlmayr
- Department of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Thomas Kroneis
- Department of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria.
| | - Amin El-Heliebi
- Department of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria
- Center for Biomarker Research, CBmed, 8010 Graz, Austria
| |
Collapse
|
7
|
|
8
|
Agnoletto C, Corrà F, Minotti L, Baldassari F, Crudele F, Cook WJJ, Di Leva G, d'Adamo AP, Gasparini P, Volinia S. Heterogeneity in Circulating Tumor Cells: The Relevance of the Stem-Cell Subset. Cancers (Basel) 2019; 11:cancers11040483. [PMID: 30959764 PMCID: PMC6521045 DOI: 10.3390/cancers11040483] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/16/2019] [Accepted: 03/30/2019] [Indexed: 12/20/2022] Open
Abstract
The release of circulating tumor cells (CTCs) into vasculature is an early event in the metastatic process. The analysis of CTCs in patients has recently received widespread attention because of its clinical implications, particularly for precision medicine. Accumulated evidence documents a large heterogeneity in CTCs across patients. Currently, the most accepted view is that tumor cells with an intermediate phenotype between epithelial and mesenchymal have the highest plasticity. Indeed, the existence of a meta-stable or partial epithelial–mesenchymal transition (EMT) cell state, with both epithelial and mesenchymal features, can be easily reconciled with the concept of a highly plastic stem-like state. A close connection between EMT and cancer stem cells (CSC) traits, with enhanced metastatic competence and drug resistance, has also been described. Accordingly, a subset of CTCs consisting of CSC, present a stemness profile, are able to survive chemotherapy, and generate metastases after xenotransplantation in immunodeficient mice. In the present review, we discuss the current evidence connecting CTCs, EMT, and stemness. An improved understanding of the CTC/EMT/CSC connections may uncover novel therapeutic targets, irrespective of the tumor type, since most cancers seem to harbor a pool of CSCs, and disclose important mechanisms underlying tumorigenicity.
Collapse
Affiliation(s)
- Chiara Agnoletto
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Fabio Corrà
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Linda Minotti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Federica Baldassari
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Francesca Crudele
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | | | - Gianpiero Di Leva
- School of Environment and Life Sciences, University of Salford, Salford M5 4WT, UK.
| | - Adamo Pio d'Adamo
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy.
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
| | - Paolo Gasparini
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy.
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
| | - Stefano Volinia
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
9
|
Parisi C, Markou A, Strati A, Kasimir-Bauer S, Lianidou ES. Development and Validation of Multiplex Liquid Bead Array Assay for the Simultaneous Expression of 14 Genes in Circulating Tumor Cells. Anal Chem 2019; 91:3443-3451. [PMID: 30663875 DOI: 10.1021/acs.analchem.8b04975] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Liquid biopsy, based on the molecular information extracted from circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA), offers the possibility to characterize the evolution of a solid tumor in real time and is highly important for diagnostic and therapeutic purposes. The aim of the present study was the development and validation of a novel liquid bead array methodology for the molecular characterization of CTCs and its application in breast cancer. In the present study we developed and evaluated a multiplex polymerase chain reaction (PCR)-coupled liquid bead array (MLBA) assay for studying simultaneously the expression of 14 genes in CTCs. The 14-gene MLBA assay is characterized by high analytical specificity, sensitivity, and reproducibility. The analytical performance of the 14-gene MLBA assay was compared with a commercially available test (AdnaTest BreastCancer, Qiagen, Germany) and our previously described multiplex quantitative reverse transcription PCR (RT-qPCR) assays. The developed assay has the potential to be further expanded in order to include up to 100 gene targets. The assay is highly specific for each target gene and is not affected by the numerous primers and probes used for multiplexing; hence, it constitutes a sample-, cost-, and time-saving analysis.
Collapse
Affiliation(s)
- Cleo Parisi
- Analysis of Circulating Tumor Cells Lab, Laboratory of Analytical Chemistry, Department of Chemistry , University of Athens , Athens 15771 , Greece
| | - Athina Markou
- Analysis of Circulating Tumor Cells Lab, Laboratory of Analytical Chemistry, Department of Chemistry , University of Athens , Athens 15771 , Greece
| | - Areti Strati
- Analysis of Circulating Tumor Cells Lab, Laboratory of Analytical Chemistry, Department of Chemistry , University of Athens , Athens 15771 , Greece
| | - Sabine Kasimir-Bauer
- Department of Gynecology and Obstetrics , University Hospital of Essen, University of Duisburg-Essen , D-45122 Essen , Germany
| | - Evi S Lianidou
- Analysis of Circulating Tumor Cells Lab, Laboratory of Analytical Chemistry, Department of Chemistry , University of Athens , Athens 15771 , Greece
| |
Collapse
|
10
|
Ozbey A, Karimzadehkhouei M, Kocaturk NM, Bilir SE, Kutlu O, Gozuacik D, Kosar A. Inertial focusing of cancer cell lines in curvilinear microchannels. MICRO AND NANO ENGINEERING 2019. [DOI: 10.1016/j.mne.2019.01.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
11
|
Wang H, Stoecklein NH, Lin PP, Gires O. Circulating and disseminated tumor cells: diagnostic tools and therapeutic targets in motion. Oncotarget 2018; 8:1884-1912. [PMID: 27683128 PMCID: PMC5352105 DOI: 10.18632/oncotarget.12242] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 09/20/2016] [Indexed: 12/16/2022] Open
Abstract
Enumeration of circulating tumor cells (CTCs) in peripheral blood with the gold standard CellSearchTM has proven prognostic value for tumor recurrence and progression of metastatic disease. Therefore, the further molecular characterization of isolated CTCs might have clinical relevance as liquid biopsy for therapeutic decision-making and to monitor disease progression. The direct analysis of systemic cancer appears particularly important in view of the known disparity in expression of therapeutic targets as well as epithelial-to-mesenchymal transition (EMT)-based heterogeneity between primary and systemic tumor cells, which all substantially complicate monitoring and therapeutic targeting at present. Since CTCs are the potential precursor cells of metastasis, their in-depth molecular profiling should also provide a useful resource for target discovery. The present review will discuss the use of systemically spread cancer cells as liquid biopsy and focus on potential target antigens.
Collapse
Affiliation(s)
- Hongxia Wang
- Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Nikolas H Stoecklein
- Department of General, Visceral and Pediatric Surgery, Medical Faculty, University Hospital of the Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | | | - Olivier Gires
- Department of Otorhinolaryngology, Head and Neck Surgery, Grosshadern Medical Center, Ludwig-Maximilians-University of Munich, Munich, Germany.,Clinical Cooperation Group Personalized Radiotherapy of Head and Neck Tumors, Helmholtz, Germany
| |
Collapse
|
12
|
Wu CP, Wu P, Zhao HF, Liu WL, Li WP. Clinical Applications of and Challenges in Single-Cell Analysis of Circulating Tumor Cells. DNA Cell Biol 2018; 37:78-89. [PMID: 29265876 DOI: 10.1089/dna.2017.3981] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Chang-peng Wu
- Department of Neurosurgery, Shenzhen Second People's Hospital, Clinical Medicine College of Anhui Medical University, Shenzhen, China
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Peng Wu
- The Affiliated Luohu Hospital of Shenzhen University, Shenzhen Luohu Hospital Group Department of Urology, Shenzhen, China
| | - Hua-fu Zhao
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Department of Neurosurgery/Neuro-oncology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wen-lan Liu
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Wei-ping Li
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| |
Collapse
|
13
|
Jan YJ, Chen JF, Zhu Y, Lu YT, Chen SH, Chung H, Smalley M, Huang YW, Dong J, Chen LC, Yu HH, Tomlinson JS, Hou S, Agopian VG, Posadas EM, Tseng HR. NanoVelcro rare-cell assays for detection and characterization of circulating tumor cells. Adv Drug Deliv Rev 2018; 125:78-93. [PMID: 29551650 PMCID: PMC5993593 DOI: 10.1016/j.addr.2018.03.006] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 03/08/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022]
Abstract
Circulating tumor cells (CTCs) are cancer cells shredded from either a primary tumor or a metastatic site and circulate in the blood as the potential cellular origin of metastasis. By detecting and analyzing CTCs, we will be able to noninvasively monitor disease progression in individual cancer patients and obtain insightful information for assessing disease status, thus realizing the concept of "tumor liquid biopsy". However, it is technically challenging to identify CTCs in patient blood samples because of the extremely low abundance of CTCs among a large number of hematologic cells. In order to address this challenge, our research team at UCLA pioneered a unique concept of "NanoVelcro" cell-affinity substrates, in which CTC capture agent-coated nanostructured substrates were utilized to immobilize CTCs with remarkable efficiency. Four generations of NanoVelcro CTC assays have been developed over the past decade for a variety of clinical utilities. The 1st-gen NanoVelcro Chips, composed of a silicon nanowire substrate (SiNS) and an overlaid microfluidic chaotic mixer, were created for CTC enumeration. The 2nd-gen NanoVelcro Chips (i.e., NanoVelcro-LMD), based on polymer nanosubstrates, were developed for single-CTC isolation in conjunction with the use of the laser microdissection (LMD) technique. By grafting thermoresponsive polymer brushes onto SiNS, the 3rd-gen Thermoresponsive NanoVelcro Chips have demonstrated the capture and release of CTCs at 37 and 4 °C respectively, thereby allowing for rapid CTC purification while maintaining cell viability and molecular integrity. Fabricated with boronic acid-grafted conducting polymer-based nanomaterial on chip surface, the 4th-gen NanoVelcro Chips (Sweet chip) were able to purify CTCs with well-preserved RNA transcripts, which could be used for downstream analysis of several cancer specific RNA biomarkers. In this review article, we will summarize the development of the four generations of NanoVelcro CTC assays, and the clinical applications of each generation of devices.
Collapse
Affiliation(s)
- Yu Jen Jan
- Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jie-Fu Chen
- Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yazhen Zhu
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yi-Tsung Lu
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Szu Hao Chen
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Howard Chung
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Matthew Smalley
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA; CytoLumina Technologies Corp., Los Angeles, CA, USA
| | - Yen-Wen Huang
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA; CytoLumina Technologies Corp., Los Angeles, CA, USA
| | - Jiantong Dong
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Li-Ching Chen
- Department of Obstetrics and Gynecology, Cathay General Hospital, Taipei, Taiwan
| | - Hsiao-Hua Yu
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - James S Tomlinson
- Department of Surgery, University of California, Los Angeles, Los Angeles, CA, USA; Center for Pancreatic Disease, University of California, Los Angeles, Los Angeles, CA, USA; Department of Surgery, Greater Los Angeles Veteran's Affairs Administration, Los Angeles, CA, USA
| | - Shuang Hou
- Department of Surgery, University of California, Los Angeles, Los Angeles, CA, USA
| | - Vatche G Agopian
- Department of Surgery, University of California, Los Angeles, Los Angeles, CA, USA; Liver Transplantation and Hepatobiliary Surgery, University of California, Los Angeles, Los Angeles, CA, USA
| | - Edwin M Posadas
- Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hsian-Rong Tseng
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
14
|
Chinen LTD, Abdallah EA, Braun AC, Flores BDCTDCP, Corassa M, Sanches SM, Fanelli MF. Circulating Tumor Cells as Cancer Biomarkers in the Clinic. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 994:1-41. [PMID: 28560666 DOI: 10.1007/978-3-319-55947-6_1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It is believed that the development of metastatic cancer requires the presence of circulating tumor cells (CTCs) , which are found in a patient's circulation as rare abnormal cells comingled with billions of the normal red and white blood cells. The systems developed for detection of CTCs have brought progress to cancer treatment. The molecular characterization of CTCs can aid in the development of new drugs, and their presence during treatment can help clinicians determine the prognosis of the patient. Studies have been carried out in patients early in the disease course, with only primary tumors, and the role of CTCs in prognosis seems to be as important as it is in patients with metastatic disease. The published studies on CTCs have focused on their prognostic significance, their utility in real-time monitoring of therapies, the identification of therapeutic and resistance targets, and understanding the process of metastasis . The analysis of CTCs during the early stages, as a "liquid biopsy," helps to monitor patients at different points in the disease course, including minimal residual disease, providing valuable information about the very early assessment of treatment effectiveness. Finally, CTCs can be used to screen patients with family histories of cancer or with diseases that can lead to the development of cancer. With standard protocols, this easily obtained and practical tool can be used to prevent the growth and spread of cancer. In this chapter, we review some important aspects of CTCs , surveying the disease aspects where these cells have been investigated.
Collapse
Affiliation(s)
| | - Emne Ali Abdallah
- International Research Center, A. C. Camargo Cancer Center, Rua Taguá 440, São Paulo, SP, 01508-010, Brazil
| | - Alexcia Camila Braun
- International Research Center, A. C. Camargo Cancer Center, Rua Taguá 440, São Paulo, SP, 01508-010, Brazil
| | | | - Marcelo Corassa
- Department of Medical Oncology, A. C. Camargo Cancer Center, Rua Professor Antônio Prudente, São Paulo, SP, 01509-010, Brazil
| | - Solange Moraes Sanches
- Department of Medical Oncology, A. C. Camargo Cancer Center, Rua Professor Antônio Prudente, São Paulo, SP, 01509-010, Brazil
| | - Marcello Ferretti Fanelli
- Department of Medical Oncology, A. C. Camargo Cancer Center, Rua Professor Antônio Prudente, São Paulo, SP, 01509-010, Brazil
| |
Collapse
|
15
|
Minimal residual disease in melanoma: circulating melanoma cells and predictive role of MCAM/MUC18/MelCAM/CD146. Cell Death Discov 2017; 3:17005. [PMID: 28280601 PMCID: PMC5337524 DOI: 10.1038/cddiscovery.2017.5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/20/2016] [Accepted: 01/01/2017] [Indexed: 12/11/2022] Open
Abstract
Circulating tumour cells (CTCs), identified in numerous cancers including melanoma, are unquestionably considered valuable and useful as diagnostic and prognostic markers. They can be detected at all melanoma stages and may persist long after treatment. A crucial step in metastatic processes is the intravascular invasion of neoplastic cells as circulating melanoma cells (CMCs). Only a small percentage of these released cells are efficient and capable of colonizing with a strong metastatic potential. CMCs' ability to survive in circulation express a variety of genes with continuous changes of signal pathways and proteins to escape immune surveillance. This makes it difficult to detect them; therefore, specific isolation, enrichment and characterization of CMC population could be useful to monitor disease status and patient clinical outcome. Overall and disease-free survival have been correlated with the presence of CMCs. Specific melanoma antigens, in particular MCAM (MUC18/MelCAM/CD146), could be a potentially useful tool to isolate CMCs as well as be a prognostic, predictive biomarker. These are the areas reviewed in the article.
Collapse
|
16
|
De Souza LM, Robertson BM, Robertson GP. Future of circulating tumor cells in the melanoma clinical and research laboratory settings. Cancer Lett 2017; 392:60-70. [PMID: 28163189 DOI: 10.1016/j.canlet.2017.01.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 01/12/2017] [Accepted: 01/13/2017] [Indexed: 12/24/2022]
Abstract
Circulating tumor cells (CTC) have become a field of interest for oncologists based on the premise that they constitute the underpinning for metastatic dissemination. The lethal nature of cancer is no longer attributed to solid tumor formation, but rather to the process of metastasis; shifting the focus of current studies towards the isolation and identification of metastatic progenitors, such as CTCs. CTCs originate from primary tumor masses that undergo morphologic and genetic alterations, which involve the release of mesenchymal-like cancer cells into the bloodstream, capable of invading nearby tissues for secondary tumor development. Cancerous cells contained in the primary tumor mass acquire the motile mesenchymal phenotype as a result of the Epithelial-to-Mesenchymal Transition, where substantial variations in protein expression and signaling pathways take place. CTCs that migrate from the primary tumor, intravasate into the systemic vasculature, are transported through the bloodstream, and invade tissues and organs suitable for secondary tumor development. While only a limited number of CTCs are viable in the bloodstream, their ability to elude the immune system, evade apoptosis and successfully metastasize at secondary tumor sites, makes CTCs promising candidates for unraveling the triggers that initiates the metastatic process. In this article, these subjects are explored in greater depth to elucidate the potential use of CTCs in the detection, disease staging and management of metastatic melanoma.
Collapse
Affiliation(s)
- Luisa M De Souza
- The Pennsylvania State University College of Medicine, Departments of Pharmacology, 500 University Drive, Hershey, PA 17033, USA.
| | - Bailey M Robertson
- The Pennsylvania State University College of Medicine, Departments of Pharmacology, 500 University Drive, Hershey, PA 17033, USA
| | - Gavin P Robertson
- The Pennsylvania State University College of Medicine, Departments of Pharmacology, 500 University Drive, Hershey, PA 17033, USA; Pathology, 500 University Drive, Hershey, PA 17033, USA; Dermatology, 500 University Drive, Hershey, PA 17033, USA; Surgery, 500 University Drive, Hershey, PA 17033, USA; The Melanoma and Skin Cancer Center, 500 University Drive, Hershey, PA 17033, USA; The Melanoma Therapeutics Program, 500 University Drive, Hershey, PA 17033, USA.
| |
Collapse
|
17
|
Circulating tumor cell detection in hepatocellular carcinoma based on karyoplasmic ratios using imaging flow cytometry. Sci Rep 2016; 6:39808. [PMID: 28009002 PMCID: PMC5180239 DOI: 10.1038/srep39808] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/25/2016] [Indexed: 12/13/2022] Open
Abstract
Circulating tumor cells (CTCs) originate from tumor tissues and are associated with cancer prognosis. However, existing technologies for CTC detection are limited owing to a lack of specific or accurate biomarkers. Here, we developed a new method for CTC detection based on the karyoplasmic ratio, without biomarkers. Consecutive patients with liver cancer or non-cancer liver diseases were recruited. CTCs in blood samples were analyzed by imaging flow cytometry based on the karyoplasmic ratio as well as EpCAM and CD45. Microvascular invasion (MVI), tumor recurrence, and survival were recorded for all patients. A total of 56.2 ± 23.8/100,000 cells with high karyoplasmic ratios (HKR cells) were detected in cancer patients, which was higher than the number of HKR cells in the non-cancer group (7.6 ± 2.2/100,000). There was also a difference in HKR cells between liver cancer patients with and without MVI. Based on a receiver operating characteristic curve analysis, the threshold was 21.8 HKR cells per 100,000 peripheral blood mononuclear cells, and the area under the curve was higher than those of traditional methods (e.g., CD45 and EpCAM staining). These results indicate that the new CTC detection method was more sensitive and reliable than existing methods. Accordingly, it may improve clinical CTC detection.
Collapse
|
18
|
Heck MM, Thalgott M, Schmid SC, Oh WK, Gong Y, Wang L, Zhu J, Seitz AK, Porst D, Höppner M, Retz M, Gschwend JE, Nawroth R. A 2-Gene Panel Derived From Prostate Cancer-Enhanced Transcripts in Whole Blood Is Prognostic for Survival and Predicts Treatment Benefit in Metastatic Castration-Resistant Prostate Cancer. Prostate 2016; 76:1160-8. [PMID: 27198487 DOI: 10.1002/pros.23202] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 04/26/2016] [Indexed: 11/07/2022]
Abstract
BACKGROUND To determine a prognostic model derived from prostate cancer-enhanced transcripts in whole blood of castration-resistant prostate cancer (CRPC) patients and explore its applicability as a surrogate of treatment response. METHODS Six out of twenty-three selected transcripts were identified as specific for detection of metastatic prostate cancer cells in peripheral blood using quantitative polymerase chain reaction (qPCR). Their prognostic value was explored in whole blood samples of a training cohort (n = 22 CRPC patients, New York, USA). A resulting 2-gene panel (2GP) including KLK2 and TMPRSS2 was validated in an independent cohort with pre- and post-treatment blood draws after 9-16 weeks of systemic treament (n = 86 CRPC patients, Munich, Germany). Overall survival (OS), prostate-specific antigen progression-free survival (PSA-PFS), and clinical PFS were analyzed. Kaplan-Meier and cox regression analyses were performed. RESULTS An unfavorable 2GP (≥1 marker positive) identified patients with poor survival (median OS 10.0 months [95%CI 5.7-14.2] vs. not reached; P = 0.023). This was validated in an independent cohort at pre-treatment (median OS 7.8 [95%CI 6.5-9.2] vs. 17.3 months [95%CI 10.7-23.8]; P = 0.004) and post-treatment blood draw (median OS 5.0 [95%CI 0.0-10.0] vs. 18.0 months [95%CI 9.5-26.6]; P = 0.003). The 2GP independently predicted OS on multivariate analysis (hazard ratio 2.1 [95%CI 1.1-4.0]; P = 0.034) and performed better than PSA decline at correlation with OS. Conversion to favorable 2GP during treatment correlated with improved OS (7.8 to 20.9 months), PSA-PFS (2.8 to 12.0 months), and clinical PFS (4.6 to 8.0 months). CONCLUSIONS The established 2GP is prognostic for survival at pre- and post-treatment blood draw in CRPC patients and conversion to favorable 2GP predicts treatment benefit. Prostate 76:1160-1168, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Matthias M Heck
- Department of Urology, Klinikum rechts der Isar, Technical University of Munich, München, Germany
| | - Mark Thalgott
- Department of Urology, Klinikum rechts der Isar, Technical University of Munich, München, Germany
| | - Sebastian C Schmid
- Department of Urology, Klinikum rechts der Isar, Technical University of Munich, München, Germany
| | - William K Oh
- Department of Hematology/Oncology, Mount Sinai Hospital, The Tisch Cancer Institute, New York, New York
| | - Yixuan Gong
- Department of Hematology/Oncology, Mount Sinai Hospital, The Tisch Cancer Institute, New York, New York
| | - Li Wang
- Genetic and Genomic Sciences, The Tisch Cancer Institute, New York, New York
| | - Jun Zhu
- Genetic and Genomic Sciences, The Tisch Cancer Institute, New York, New York
| | - Anna-Katharina Seitz
- Department of Urology, Klinikum rechts der Isar, Technical University of Munich, München, Germany
| | - Desiree Porst
- Department of Urology, Klinikum rechts der Isar, Technical University of Munich, München, Germany
| | - Michael Höppner
- Department of Urology, Klinikum rechts der Isar, Technical University of Munich, München, Germany
| | - Margitta Retz
- Department of Urology, Klinikum rechts der Isar, Technical University of Munich, München, Germany
| | - Jürgen E Gschwend
- Department of Urology, Klinikum rechts der Isar, Technical University of Munich, München, Germany
| | - Roman Nawroth
- Department of Urology, Klinikum rechts der Isar, Technical University of Munich, München, Germany
| |
Collapse
|
19
|
Chen JF, Zhu Y, Lu YT, Hodara E, Hou S, Agopian VG, Tomlinson JS, Posadas EM, Tseng HR. Clinical Applications of NanoVelcro Rare-Cell Assays for Detection and Characterization of Circulating Tumor Cells. Theranostics 2016; 6:1425-39. [PMID: 27375790 PMCID: PMC4924510 DOI: 10.7150/thno.15359] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 05/06/2016] [Indexed: 12/22/2022] Open
Abstract
Liquid biopsy of tumor through isolation of circulating tumor cells (CTCs) allows non-invasive, repetitive, and systemic sampling of disease. Although detecting and enumerating CTCs is of prognostic significance in metastatic cancer, it is conceivable that performing molecular and functional characterization on CTCs will reveal unprecedented insight into the pathogenic mechanisms driving lethal disease. Nanomaterial-embedded cancer diagnostic platforms, i.e., NanoVelcro CTC Assays represent a unique rare-cell sorting method that enables detection isolation, and characterization of CTCs in peripheral blood, providing an opportunity to noninvasively monitor disease progression in individual cancer patients. Over the past decade, a series of NanoVelcro CTC Assays has been demonstrated for exploring the full potential of CTCs as a clinical biomarker, including CTC enumeration, phenotyping, genotyping and expression profiling. In this review article, the authors will briefly introduce the development of three generations of NanoVelcro CTC Assays, and highlight the clinical applications of each generation for various types of solid cancers, including prostate cancer, pancreatic cancer, lung cancer, and melanoma.
Collapse
Affiliation(s)
- Jie-Fu Chen
- 1. Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Yazhen Zhu
- 2. Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, California, USA;; 3. Department of Pathology, Guangdong Provincial Hospital of TCM, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yi-Tsung Lu
- 1. Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Elisabeth Hodara
- 1. Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Shuang Hou
- 3. Department of Pathology, Guangdong Provincial Hospital of TCM, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Vatche G Agopian
- 4. Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA;; 5. Liver Transplantation and Hepatobiliary Surgery, University of California, Los Angeles, Los Angeles, California, USA
| | - James S Tomlinson
- 4. Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA;; 6. Center for Pancreatic Disease, University of California, Los Angeles, Los Angeles, California, USA;; 7. Department of Surgery Greater Los Angeles Veteran's Affairs Administration, Los Angeles, California, USA
| | - Edwin M Posadas
- 1. Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Hsian-Rong Tseng
- 2. Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
20
|
Ferreira MM, Ramani VC, Jeffrey SS. Circulating tumor cell technologies †. Mol Oncol 2016; 10:374-94. [PMID: 26897752 PMCID: PMC5528969 DOI: 10.1016/j.molonc.2016.01.007] [Citation(s) in RCA: 359] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 01/16/2016] [Accepted: 01/19/2016] [Indexed: 02/08/2023] Open
Abstract
Circulating tumor cells, a component of the “liquid biopsy”, hold great potential to transform the current landscape of cancer therapy. A key challenge to unlocking the clinical utility of CTCs lies in the ability to detect and isolate these rare cells using methods amenable to downstream characterization and other applications. In this review, we will provide an overview of current technologies used to detect and capture CTCs with brief insights into the workings of individual technologies. We focus on the strategies employed by different platforms and discuss the advantages of each. As our understanding of CTC biology matures, CTC technologies will need to evolve, and we discuss some of the present challenges facing the field in light of recent data encompassing epithelial‐to‐mesenchymal transition, tumor‐initiating cells, and CTC clusters. We present a comprehensive overview of CTC detection and capture technologies. We provide a conceptual description of strategies used in different technologies. We highlight the key features of individual technologies. We discuss CTC technology performance in the context of clinical studies.
Collapse
Affiliation(s)
- Meghaan M Ferreira
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Vishnu C Ramani
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Stefanie S Jeffrey
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
21
|
Hoshiba T, Nikaido M, Yagi S, Konno I, Yoshihiro A, Tanaka M. Blood-compatible poly (2-methoxyethyl acrylate) for the adhesion and proliferation of lung cancer cells toward the isolation and analysis of circulating tumor cells. J BIOACT COMPAT POL 2016. [DOI: 10.1177/0883911515618976] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Circulating tumor cells have received attention for their role in cancer diagnosis and the decision on which chemotherapeutic course to take. For these purposes, the isolation of circulating tumor cells has been important. Previously, we reported that non-blood cells can adhere on blood-compatible polymer substrates, such as poly(2-methoxyethyl acrylate) and poly(tetrahydrofurfuryl acrylate). In this study, we examined whether blood-compatible poly(2-methoxyethyl acrylate) and poly(tetrahydrofurfuryl acrylate) allow the adhesion and growth of A549 lung cancer cells for isolating circulating tumor cells by adhesion-mediated manner to diagnose metastatic cancer and to decide on the chemotherapeutic course. A549 cells can adhere on poly(2-methoxyethyl acrylate) and poly(tetrahydrofurfuryl acrylate) substrates via an integrin-dependent mechanism after 1 h of incubation, suggesting that blood-compatible poly(2-methoxyethyl acrylate) and poly(tetrahydrofurfuryl acrylate) substrates possess the ability to capture circulating tumor cells selectively from peripheral blood. After 1 day of culture, A549 cells started to spread on poly(2-methoxyethyl acrylate) and poly(tetrahydrofurfuryl acrylate) substrates. A549 can also grow on poly(2-methoxyethyl acrylate) and poly(tetrahydrofurfuryl acrylate) substrates. Additionally, the chemoresistance of A549 cells against 5-fluorouracil on poly(2-methoxyethyl acrylate) and poly(tetrahydrofurfuryl acrylate) substrates was similar to that on the conventional cell culture substrate, tissue culture polystyrene. These results indicate that circulating tumor cells can be cultured on poly(2-methoxyethyl acrylate) and poly(tetrahydrofurfuryl acrylate) substrates after they are isolated from peripheral blood, and poly(2-methoxyethyl acrylate) and poly(tetrahydrofurfuryl acrylate) substrates can be used as circulating tumor cell culture substrates for screening anti-cancer drugs. Therefore, poly(2-methoxyethyl acrylate) and poly(tetrahydrofurfuryl acrylate) substrates might be able to be applied to the development of a new device for a circulating tumor cell–based diagnosis of metastatic cancer and a personalized medicine approach regarding the decision of which chemotherapeutic course should be taken.
Collapse
Affiliation(s)
- Takashi Hoshiba
- Frontier Center for Organic Materials, Yamagata University, Yamagata, Japan
- International Center for Materials Nanoarchitectonics, National Institute for Materials Science, Tsukuba, Japan
| | - Mayo Nikaido
- Graduate School of Science and Engineering, Yamagata University, Yamagata, Japan
| | - Satomi Yagi
- Graduate School of Science and Engineering, Yamagata University, Yamagata, Japan
| | - Iku Konno
- Department of Biochemical Engineering, Yamagata University, Yamagata, Japan
| | - Ayano Yoshihiro
- Department of Biochemical Engineering, Yamagata University, Yamagata, Japan
| | - Masaru Tanaka
- Frontier Center for Organic Materials, Yamagata University, Yamagata, Japan
- Institute for Materials Chemistry and Engineering, Kyushu University, Fukuoka, Japan
| |
Collapse
|
22
|
Hoshiba T, Orui T, Endo C, Sato K, Yoshihiro A, Minagawa Y, Tanaka M. Adhesion-based simple capture and recovery of circulating tumor cells using a blood-compatible and thermo-responsive polymer-coated substrate. RSC Adv 2016. [DOI: 10.1039/c6ra15229e] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Circulating tumor cells (CTCs) have been a focus of study for metastatic cancer diagnostics, in in vitro anti-cancer drug screening to decide the chemotherapeutic course, and cancer biology research.
Collapse
Affiliation(s)
- Takashi Hoshiba
- Frontier Center for Organic Materials
- Yamagata University
- Yonezawa
- Japan
- International Center for Materials Nanoarchitectonics
| | - Toshihiko Orui
- Graduate School of Science and Engineering
- Yamagata University
- Yonezawa
- Japan
| | - Chiho Endo
- Graduate School of Science and Engineering
- Yamagata University
- Yonezawa
- Japan
| | - Kazuhiro Sato
- Graduate School of Science and Engineering
- Yamagata University
- Yonezawa
- Japan
| | - Ayano Yoshihiro
- Department of Biochemical Engineering
- Yamagata University
- Yonezawa
- Japan
| | | | - Masaru Tanaka
- Frontier Center for Organic Materials
- Yamagata University
- Yonezawa
- Japan
- Institute for Materials Chemistry and Engineering
| |
Collapse
|
23
|
Strohm EM, Kolios MC. Classification of blood cells and tumor cells using label-free ultrasound and photoacoustics. Cytometry A 2015; 87:741-9. [DOI: 10.1002/cyto.a.22698] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 03/05/2015] [Accepted: 05/04/2015] [Indexed: 12/15/2022]
Affiliation(s)
- Eric M. Strohm
- Department of Physics; Ryerson University; Toronto Canada
| | | |
Collapse
|
24
|
Lu NN, Xie M, Wang J, Lv SW, Yi JS, Dong WG, Huang WH. Biotin-triggered decomposable immunomagnetic beads for capture and release of circulating tumor cells. ACS APPLIED MATERIALS & INTERFACES 2015; 7:8817-26. [PMID: 25853336 DOI: 10.1021/acsami.5b01397] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Isolation of rare, pure, and viable circulating tumor cells (CTCs) provides a significant insight in early cancer diagnosis, and release of captured CTCs without damage for ex vivo culture may offer an opportunity for personalized cancer therapy. In this work, we described a biotin-triggered decomposable immunomagnetic system, in which peptide-tagged antibody designed by chemical conjugation was specifically immobilized on engineered protein-coated magnetic beads. The interaction between peptide and engineered protein can be reversibly destroyed by biotin treatment, making capture and release of CTCs possible. Furthermore, the peptide could mediate multiple antibodies' coimmobilization on engineered protein-coated magnetic beads, by which capture efficiency for CTCs was obviously improved. Quantitative results showed that 70% of captured cells could be released by biotin addition, and 85% of released cells remained viable. In addition, 79% of cancer cells spiked in human whole blood were captured and could also be successfully released for culture. Finally, immunomagnetic beads simultaneously loaded with anti-EpCAM, anti-HER2, and anti-EGFR were successfully applied to isolate and detect CTCs in 17 cancer patients' peripheral blood samples, and 2-215 CTCs were identified with high purity. These results suggest that our method is reliable and has great potential in CTC detection for CTC-based molecular profiling, diagnosis, and therapy.
Collapse
Affiliation(s)
- Ning-Ning Lu
- †Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Min Xie
- †Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Jing Wang
- ‡Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Song-Wei Lv
- †Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Jia-Sheng Yi
- ‡Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wei-Guo Dong
- ‡Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wei-Hua Huang
- †Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| |
Collapse
|
25
|
Mayo C, Ortega FG, Giménez-Capitán A, Molina-Vila MA, Serrano MJ, Viteri S, Costa C, Gascó A, Bertran-Alamillo J, Karachaliou N, Lorente JA, Tarón M, Rosell R. CK-coated magnetic-based beads as a tool to isolate circulating tumor cells (CTCs) in human tumors. Transl Lung Cancer Res 2015; 2:65-71. [PMID: 25806217 DOI: 10.3978/j.issn.2218-6751.2013.02.06] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 02/25/2013] [Indexed: 01/01/2023]
Abstract
Circulating tumor cells (CTCs) can be detected in the blood of many cancer patients and play a key role in metastasis. In addition, after the development of technologies with the necessary sensitivity and reproducibility, the diagnostic potential of these cells is being actively explored. Recently, the U.S. Food and Drug Administration has approved the CellSearch(®) System, based on magnetic beads coated with epithelial cell-adhesion molecule (EpCAM) antibody. Despite its usefulness, this system can miss CTCs that lose epithelial antigens due to the epithelial-mesenchymal transition and, in the case of advanced NSCLC, CTCs positivity can be demonstrated only in 30-50% of patients. In an effort to overcome these drawbacks, new methods are being developed. In this study, we have evaluated CK-coated beads as a system to isolate CTCs from lung cancer patients in the clinical setting, and have evaluated if they can be a useful source of material for genetic testing. We were able to identify CTCs in 17 of the 30 patients included in the study (57%), with a range of 1 to 7 cells. In two of them, we found only CTCs with an EMT pattern. CTC positivity seemed to correlate with the clinical history of the malignancy. CTCs could be detected in more than 80% of stage III-IV lung cancer patients at presentation or in blood samples taken immediately after surgery. The percentage dropped to 13% in patients responding to chemotherapy or TKIs, raising again to 57% after tumor progression. Finally, we tested the CTCs isolated from 8 patients for EGFR and k-ras mutations, but gene amplification was successful only in the 3 patients with 4 or more CTCs.
Collapse
Affiliation(s)
- Clara Mayo
- Breakthrought Cancer Unit, Laboratory of Oncology, Pangaea Biotech, Dexeus University Hospital, Barcelona, Spain
| | - Francisco G Ortega
- GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, Granada, Spain
| | - Ana Giménez-Capitán
- Breakthrought Cancer Unit, Laboratory of Oncology, Pangaea Biotech, Dexeus University Hospital, Barcelona, Spain
| | - Miguel A Molina-Vila
- Breakthrought Cancer Unit, Laboratory of Oncology, Pangaea Biotech, Dexeus University Hospital, Barcelona, Spain
| | - Maria José Serrano
- GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, Granada, Spain
| | - Santiago Viteri
- Breakthrought Cancer Unit, Laboratory of Oncology, Pangaea Biotech, Dexeus University Hospital, Barcelona, Spain
| | - Carlota Costa
- Breakthrought Cancer Unit, Laboratory of Oncology, Pangaea Biotech, Dexeus University Hospital, Barcelona, Spain
| | - Amaya Gascó
- Breakthrought Cancer Unit, Laboratory of Oncology, Pangaea Biotech, Dexeus University Hospital, Barcelona, Spain
| | - Jordi Bertran-Alamillo
- Breakthrought Cancer Unit, Laboratory of Oncology, Pangaea Biotech, Dexeus University Hospital, Barcelona, Spain
| | - Niki Karachaliou
- Breakthrought Cancer Unit, Laboratory of Oncology, Pangaea Biotech, Dexeus University Hospital, Barcelona, Spain
| | - José Antonio Lorente
- GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, Granada, Spain
| | - Miquel Tarón
- Breakthrought Cancer Unit, Laboratory of Oncology, Pangaea Biotech, Dexeus University Hospital, Barcelona, Spain ; ; Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Rafael Rosell
- Breakthrought Cancer Unit, Laboratory of Oncology, Pangaea Biotech, Dexeus University Hospital, Barcelona, Spain ; ; Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona, Spain
| |
Collapse
|
26
|
Yu L, Shi Z, Gao L, Li C. Mitigated reactive oxygen species generation leads to an improvement of cell proliferation on poly[glycidyl methacrylate-co-poly(ethylene glycol) methacrylate] functionalized polydimethylsiloxane surfaces. J Biomed Mater Res A 2015; 103:2987-97. [DOI: 10.1002/jbm.a.35432] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 12/19/2014] [Accepted: 02/13/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Ling Yu
- Faculty of Materials & Energy; Institute for Clean Energy & Advanced Materials, Southwest University; Chongqing 400715 China
- Chongqing Key Laboratory for Advanced Materials and Technologies of Clean Energies; Chongqing 400715 China
- Chongqing Engineering Research Center for Rapid Diagnosis of Fatal Diseases; Chongqing 400715 China
| | - ZhuanZhuan Shi
- Faculty of Materials & Energy; Institute for Clean Energy & Advanced Materials, Southwest University; Chongqing 400715 China
- Chongqing Key Laboratory for Advanced Materials and Technologies of Clean Energies; Chongqing 400715 China
- Chongqing Engineering Research Center for Rapid Diagnosis of Fatal Diseases; Chongqing 400715 China
| | - LiXia Gao
- Faculty of Materials & Energy; Institute for Clean Energy & Advanced Materials, Southwest University; Chongqing 400715 China
- Chongqing Key Laboratory for Advanced Materials and Technologies of Clean Energies; Chongqing 400715 China
- Chongqing Engineering Research Center for Rapid Diagnosis of Fatal Diseases; Chongqing 400715 China
| | - ChangMing Li
- Faculty of Materials & Energy; Institute for Clean Energy & Advanced Materials, Southwest University; Chongqing 400715 China
- Chongqing Key Laboratory for Advanced Materials and Technologies of Clean Energies; Chongqing 400715 China
- Chongqing Engineering Research Center for Rapid Diagnosis of Fatal Diseases; Chongqing 400715 China
| |
Collapse
|
27
|
Sfakianakis S, Bei ES, Zervakis M, Vassou D, Kafetzopoulos D. On the identification of circulating tumor cells in breast cancer. IEEE J Biomed Health Inform 2015; 18:773-82. [PMID: 24808221 DOI: 10.1109/jbhi.2013.2295262] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Breast cancer is a highly heterogeneous disease and very common among western women. The main cause of death is not the primary tumor but its metastases at distant sites, such as lymph nodes and other organs (preferentially lung, liver, and bones). The study of circulating tumor cells (CTCs) in peripheral blood resulting from tumor cell invasion and intravascular filtration highlights their crucial role concerning tumor aggressiveness and metastasis. Genomic research regarding CTCs monitoring for breast cancer is limited due to the lack of indicative genes for their detection and isolation. Instead of direct CTC detection, in our study, we focus on the identification of factors in peripheral blood that can indirectly reveal the presence of such cells. Using selected publicly available breast cancer and peripheral blood microarray datasets, we follow a two-step elimination procedure for the identification of several discriminant factors. Our procedure facilitates the identification of major genes involved in breast cancer pathology, which are also indicative of CTCs presence.
Collapse
|
28
|
Mumford BS, Robertson GP. Circulating melanoma cells in the diagnosis and monitoring of melanoma: an appraisal of clinical potential. Mol Diagn Ther 2014; 18:175-83. [PMID: 24297151 DOI: 10.1007/s40291-013-0071-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Circulating melanoma cells (CMCs) are thought to be the foundation for metastatic disease, which makes this cancer especially lethal. Cancer cells contained in the primary tumor undergo genotypic and phenotypic changes leading to an epithelial-to-mesenchymal transition, during which numerous changes occur in signaling pathways and proteins in the cells. CMCs are then shed off or migrate from the primary tumor and intravasate the vasculature system. A few CMCs are able to survive in the circulation through expression of a variety of genes and also by evading immune system recognition to establish metastases at distant sites after extravasating from the vessels. The presence of CMCs in the blood of a melanoma patient can be used for disease staging, predicting metastasis development, and evaluating the efficacy of therapeutic agents. Overall survival and disease-free duration can also be correlated with the presence of CMCs. Finally, analysis of CMCs for druggable therapeutic gene targets could lead to the development of personalized treatment regimens to prevent metastasis. Thus, the study of CMCs shows promise for the detection, staging, and monitoring of disease treatment, as well as for determination of prognosis and predicting overall disease-free survival. These are the areas reviewed in this article.
Collapse
Affiliation(s)
- Brigid S Mumford
- Department of Pharmacology, Penn State College of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
| | | |
Collapse
|
29
|
Assessment of EGFR mutations in circulating tumor cell preparations from NSCLC patients by next generation sequencing: toward a real-time liquid biopsy for treatment. PLoS One 2014; 9:e103883. [PMID: 25137181 PMCID: PMC4138040 DOI: 10.1371/journal.pone.0103883] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 07/08/2014] [Indexed: 01/18/2023] Open
Abstract
Introduction Assessment of EGFR mutation in non-small cell lung cancer (NSCLC) patients is mandatory for optimization of pharmacologic treatment. In this respect, mutation analysis of circulating tumor cells (CTCs) may be desirable since they may provide real-time information on patient's disease status. Experimental Design Blood samples were collected from 37 patients enrolled in the TRIGGER study, a prospective phase II multi-center trial of erlotinib treatment in advanced NSCLC patients with activating EGFR mutations in tumor tissue. 10 CTC preparations from breast cancer patients without EGFR mutations in their primary tumors and 12 blood samples from healthy subjects were analyzed as negative controls. CTC preparations, obtained by the Veridex CellSearch System, were subjected to ultra-deep next generation sequencing (NGS) on the Roche 454 GS junior platform. Results CTCs fulfilling all Veridex criteria were present in 41% of the patients examined, ranging in number between 1 and 29. In addition to validated CTCs, potential neoplastic elements were seen in 33 cases. These included cells not fulfilling all Veridex criteria (also known as “suspicious objects”) found in 5 (13%) of 37 cases, and isolated or clustered large naked nuclei with irregular shape observed in 33 (89%) cases. EGFR mutations were identified by NGS in CTC preparations of 31 (84%) patients, corresponding to those present in matching tumor tissue. Twenty-five (96%) of 26 deletions at exon 19 and 6 (55%) of 11 mutations at exon 21 were detectable (P = 0.005). In 4 (13%) cases, multiple EGFR mutations, suggesting CTC heterogeneity, were documented. No mutations were found in control samples. Conclusions We report for the first time that the CellSearch System coupled with NGS is a very sensitive and specific diagnostic tool for EGFR mutation analysis in CTC preparations with potential clinical impact.
Collapse
|
30
|
Identification of novel markers that outperform EpCAM in quantifying circulating tumor cells. Cell Oncol (Dordr) 2014; 37:235-43. [PMID: 25001871 DOI: 10.1007/s13402-014-0178-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2014] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Circulating tumor cells (CTCs) can be used to predict the spread of cancer to distant sites, to monitor the clinical response to therapy and to predict patient survival. The currently used EpCAM antibody-mediated identification of CTCs may lead to false negative results due to the low level or absence of EpCAM expression in several types of cancer, thus provoking a need to identify novel CTC markers. METHODS The Cancer Cell Line Encyclopedia (CCLE) microarray dataset, storing 18,915 gene expression profiles across 967 cancer cell lines derived from 25 primary sites, was systematically analyzed. The results obtained were cross-validated using an independent microarray dataset generated from 1,911 clinical cancer specimens derived from 15 different cancers. RESULTS Through bioinformatics analyses we identified, categorized and prioritized three classes of novel markers: pan-CTC markers (n = 45), EpCAM((-/low)) CTC markers (n = 16) and single cancer type-specific markers (n = 74). The pan-CTC markers were significantly, uniformly and constitutively over-expressed in most cancer types, except in cancers of hematopoietic and lymphoid origin. The EpCAM((-/low)) CTC markers were over-expressed in cancers with low or undetectable EpCAM expression levels. Among these, 22 markers were validated in an independent microarray dataset. In addition, 74 markers that were over-expressed in only single cancer types were categorized. CONCLUSIONS The combined use of these novel markers in conjunction with cancer type-specific markers should be able to quantify CTCs that are not captured by EpCAM antibodies, and to enhance the sensitivity and specificity of CTC detection among admixtures containing leucocytes or other types of contaminants.
Collapse
|
31
|
Nel I, David P, Gerken GGH, Schlaak JF, Hoffmann AC. Role of circulating tumor cells and cancer stem cells in hepatocellular carcinoma. Hepatol Int 2014. [PMID: 26202635 DOI: 10.1007/s12072-014-9539-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Circulating tumor cells (CTC) and cancer stem cells (CSC) have been proposed as tools for detection and characterization of disease and individualization of therapy in patients with many solid tumors. Several automated and semi-automated techniques for identification and isolation of these cells from blood have been proposed and reviewed mostly focusing on their feasibility. In this mini review we summarize the recent relevant literature on this topic and discuss the clinical usability of measuring CTC and CSC in peripheral blood in patients with hepatocellular carcinoma (HCC). Besides literature, the basis for this evaluation was the authors' experience with treating HCC and research experience on CSC and CTC. Few original reports and reviews have been published focusing on CTC and CSC in HCC. Though HCC is one of the five most common malignancies worldwide only recently these cells have come into focus for detection and characterization of this disease that is characterized by high plasticity and malignancy. A focused and prospective validation of the clinical usability of detecting these cells in HCC is still needed, but results seem promising that they may add great benefit for early detection and individualization of therapy.
Collapse
Affiliation(s)
- Ivonne Nel
- Department of Medical Oncology, Molecular Oncology Risk-Profile Evaluation, West German Cancer Center, University Hospital of Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Paul David
- Department of Medical Oncology, Molecular Oncology Risk-Profile Evaluation, West German Cancer Center, University Hospital of Essen, Hufelandstrasse 55, 45147, Essen, Germany
- Institute of Liver and Biliary Sciences (ILBS), Vasant Kunj, New Delhi, India
| | - Guido G H Gerken
- Department of Gastroenterology and Hepatology, University Hospital of Essen, Essen, Germany
| | - Joerg F Schlaak
- Department of Gastroenterology and Hepatology, University Hospital of Essen, Essen, Germany
- Department of Internal Medicine, Evangelisches Krankenhaus Duisburg-Nord, Duisburg, Germany
| | - Andreas-Claudius Hoffmann
- Department of Medical Oncology, Molecular Oncology Risk-Profile Evaluation, West German Cancer Center, University Hospital of Essen, Hufelandstrasse 55, 45147, Essen, Germany.
| |
Collapse
|
32
|
Tryfonidis K, Kafousi M, Perraki M, Apostolaki S, Agelaki S, Georgoulias V, Stathopoulos E, Mavroudis D. Detection of circulating cytokeratin-19 mRNA-positive cells in the blood and the mitotic index of the primary tumor have independent prognostic value in early breast cancer. Clin Breast Cancer 2014; 14:442-50. [PMID: 24958324 DOI: 10.1016/j.clbc.2014.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 04/07/2014] [Accepted: 04/23/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND Previous studies showed that molecular detection of CK-19 mRNA in peripheral blood and the mitotic index of primary tumors have prognostic value in early breast cancer. The aim of this study was to assess the association between these variables. PATIENTS AND METHODS The primary tumors of 223 operable breast cancer patients (92 premenopausal and 131 postmenopausal) were evaluated for the MAI classified as either ≤ 5 per 10, 6 to 10 per 10 and > 10 per 10 or < 10 per 10 and ≥ 10 per 10 mitoses per high power field using a standardized protocol previously reported. Peripheral blood was also collected before and after the end of adjuvant chemotherapy for detection of CK-19 mRNA-positive cells using reverse transcription polymerase chain reaction previously described. RESULTS After a median follow-up of 118 months, 75 patients (33.6%) experienced disease relapse and 56 (25.1%) died of breast cancer. MAI was strongly associated with disease-free survival (DFS) and overall survival (OS) (P < .001 for DFS and OS together). Detecting CK-19 mRNA-positive cells in the peripheral blood before but not after adjuvant chemotherapy was associated with marginally worse DFS (P = .055) and OS (P = .059). Cox regression analysis revealed that MAI and CK-19 mRNA-positive cell detection before adjuvant chemotherapy were independent variables associated with decreased DFS (P < .001 and P = .038, respectively) and OS (P < .001 and P = .029, respectively). There was no significant interaction between MAI and detection of CK-19 mRNA-positive cells. CONCLUSION MAI of the primary tumor and detection of CK-19 mRNA-positive cells in the blood before adjuvant chemotherapy in early breast cancer patients are 2 independent prognostic factors associated with clinical outcome.
Collapse
Affiliation(s)
- Konstantinos Tryfonidis
- Department of Medical Oncology, University General Hospital of Heraklion, Heraklion, Crete, Greece
| | - Maria Kafousi
- Department of Pathology, University General Hospital of Heraklion, Heraklion, Crete, Greece
| | - Maria Perraki
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Stella Apostolaki
- Department of Pathology, University General Hospital of Heraklion, Heraklion, Crete, Greece
| | - Sofia Agelaki
- Department of Medical Oncology, University General Hospital of Heraklion, Heraklion, Crete, Greece; Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Vassilis Georgoulias
- Department of Medical Oncology, University General Hospital of Heraklion, Heraklion, Crete, Greece; Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | | | - Dimitris Mavroudis
- Department of Medical Oncology, University General Hospital of Heraklion, Heraklion, Crete, Greece; Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Heraklion, Crete, Greece.
| |
Collapse
|
33
|
Lowes LE, Allan AL. Recent advances in the molecular characterization of circulating tumor cells. Cancers (Basel) 2014; 6:595-624. [PMID: 24633084 PMCID: PMC3980613 DOI: 10.3390/cancers6010595] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 01/28/2014] [Accepted: 02/20/2014] [Indexed: 12/16/2022] Open
Abstract
Although circulating tumor cells (CTCs) were first observed over a century ago, lack of sensitive methodology precluded detailed study of these cells until recently. However, technological advances have now facilitated the identification, enumeration, and characterization of CTCs using a variety of methods. The majority of evidence supporting the use of CTCs in clinical decision-making has been related to enumeration using the CellSearch® system and correlation with prognosis. Growing evidence also suggests that CTC monitoring can provide an early indication of patient treatment response based on comparison of CTC levels before and after therapy. However, perhaps the greatest potential that CTCs hold for oncology lies at the level of molecular characterization. Clinical treatment decisions may be more effective if they are based on molecular characteristics of metastatic cells rather than on those of the primary tumor alone. Molecular characterization of CTCs (which can be repeatedly isolated in a minimally invasive fashion) provides the opportunity for a "real-time liquid biopsy" that allows assessment of genetic drift, investigation of molecular disease evolution, and identification of actionable genomic characteristics. This review focuses on recent advances in this area, including approaches involving immunophenotyping, fluorescence in situ hybridization (FISH), multiplex RT-PCR, microarray, and genomic sequencing.
Collapse
Affiliation(s)
- Lori E Lowes
- London Regional Cancer Program, London Health Sciences Centre, London, ON N6A 4L6, Canada.
| | - Alison L Allan
- London Regional Cancer Program, London Health Sciences Centre, London, ON N6A 4L6, Canada.
| |
Collapse
|
34
|
Bossmann SH, Troyer DL. Point-of-care routine rapid screening: the future of cancer diagnosis? Expert Rev Mol Diagn 2014; 13:107-9. [DOI: 10.1586/erm.13.3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
35
|
Diagnostic leukapheresis enables reliable detection of circulating tumor cells of nonmetastatic cancer patients. Proc Natl Acad Sci U S A 2013; 110:16580-5. [PMID: 24065821 DOI: 10.1073/pnas.1313594110] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Circulating tumor cells (CTCs) are promising biomarkers for diagnosis and therapy in systemic cancer. However, their infrequent and unreliable detection, especially in nonmetastatic cancer, currently impedes the clinical use of CTCs. Because leukapheresis (LA) targets peripheral blood mononuclear cells, which have a similar density to CTCs, and usually involves processing the whole circulating blood, we tested whether LA could substantially increase CTC detection in operable cancer patients. Therefore, we screened LA products generated from up to 25 L of blood per patient in two independent studies, and found that CTCs can be detected in more than 90% of nonmetastatic breast cancer patients. Interestingly, complete white blood cell sampling enabled determining an upper level for total CTC numbers of about 100,000 cells (median, 7,500 CTCs) per patient and identified a correlation of CTC numbers with anatomic disease spread. We further show that diagnostic leukapheresis can be easily combined with the US Food and Drug Administration-approved CellSearch system for standardized enumeration of CTCs. Direct comparison with 7.5 mL of blood revealed a significantly higher CTC frequency in matched LA samples. Finally, genomic single-cell profiling disclosed highly aberrant CTCs as therapy-escaping variants in breast cancer. In conclusion, LA is a clinically safe method that enabled a reliable detection of CTCs at high frequency even in nonmetastatic cancer patients, and might facilitate the routine clinical use of CTCs as in the sense of a liquid biopsy. Combined with technologies for single-cell molecular genetics or cell biology, it may significantly improve prediction of therapy response and monitoring of early systemic cancer.
Collapse
|
36
|
Kim MS, Kim J, Lee W, Cho SJ, Oh JM, Lee JY, Baek S, Kim YJ, Sim TS, Lee HJ, Jung GE, Kim SI, Park JM, Oh JH, Gurel O, Lee SS, Lee JG. A trachea-inspired bifurcated microfilter capturing viable circulating tumor cells via altered biophysical properties as measured by atomic force microscopy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2013; 9:3103-3110. [PMID: 23401221 DOI: 10.1002/smll.201202317] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 10/15/2012] [Indexed: 06/01/2023]
Abstract
Circulating tumor cells (CTCs), though exceedingly rare in the blood, are nonetheless becoming increasingly important in cancer diagnostics. Despite this keen interest and the growing number of potential clinical applications, there has been limited success in developing a CTC isolation platform that simultaneously optimizes recovery rates, purity, and cell compatibility. Herein, a novel tracheal carina-inspired bifurcated (TRAB) microfilter system is reported, which uses an optimal filter gap size satisfying both 100% theoretical recovery rate and purity, as determined by biomechanical analysis and fluid-structure interaction (FSI) simulations. Biomechanical properties are also used to clearly discriminate between cancer cells and leukocytes, whereby cancer cells are selectively bound to melamine microbeads, which increase the size and stiffness of these cells. Nanoindentation experiments are conducted to measure the stiffness of leukocytes as compared to the microbead-conjugated cancer cells, with these parameters then being used in FSI analyses to optimize the filter gap size. The simulation results show that given a flow rate of 100 μL min(-1), an 8 μm filter gap optimizes the recovery rate and purity. MCF-7 breast cancer cells with solid microbeads are spiked into 3 mL of whole blood and, by using this flow rate along with the optimized microfilter dimensions, the cell mixture passes through the TRAB filter, which achieves a recovery rate of 93% and purity of 59%. Regarding cell compatibility, it is verified that the isolation procedure does not adversely affect cell viability, thus also confirming that the re-collected cancer cells can be cultured for up to 8 days. This work demonstrates a CTC isolation technology platform that optimizes high recovery rates and cell purity while also providing a framework for functional cell studies, potentially enabling even more sensitive and specific cancer diagnostics.
Collapse
Affiliation(s)
- Minseok S Kim
- POCT In Vitro Diagnostics Group, R&D Solution Group, Samsung Advanced Institute of Technology (SAIT), San 14, Nongseo-dong, Giheung-gu, Yongin-si, Gyeonggi-do, Korea.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Lianidou ES, Markou A, Strati A. Molecular characterization of circulating tumor cells in breast cancer: challenges and promises for individualized cancer treatment. Cancer Metastasis Rev 2013; 31:663-71. [PMID: 22692478 DOI: 10.1007/s10555-012-9366-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Blood testing using Circulating Tumor Cells (CTCs) has emerged as one of the hottest fields in cancer diagnosis. Research on CTCs present nowadays a challenge, as these cells are well defined targets for understanding tumour biology and improving cancer treatment. The presence of tumor cells in patient's bone marrow or peripheral blood is an early indicator of metastasis and may signal tumor spread sooner than clinical symptoms appear and imaging results confirm a poor prognosis. CTC enumeration can serve as a "liquid biopsy" and an early marker to assess response to systemic therapy. Definition of biomarkers based on comprehensive characterization of CTCs has a strong potential to be translated to individualized targeted treatments and spare breast cancer patients unnecessary and ineffective therapies but also to reduce the costs for the health system and to downsize the extent and length of clinical studies. In this review, we briefly summarize recent studies on the molecular characterization of circulating tumor cells in breast cancer and discuss challenges and promises of CTCs for individualized cancer treatment.
Collapse
Affiliation(s)
- Evi S Lianidou
- Analysis of Circulating Tumor Cells Lab, Laboratory of Analytical Chemistry, Department of Chemistry, University of Athens, 15771 Athens, Greece.
| | | | | |
Collapse
|
38
|
Hosokawa M, Yoshikawa T, Negishi R, Yoshino T, Koh Y, Kenmotsu H, Naito T, Takahashi T, Yamamoto N, Kikuhara Y, Kanbara H, Tanaka T, Yamaguchi K, Matsunaga T. Microcavity array system for size-based enrichment of circulating tumor cells from the blood of patients with small-cell lung cancer. Anal Chem 2013; 85:5692-8. [PMID: 23706033 DOI: 10.1021/ac400167x] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this study, we present a method for efficient enrichment of small-sized circulating tumor cells (CTCs) such as those found in the blood of small-cell lung cancer (SCLC) patients using a microcavity array (MCA) system. To enrich CTCs from whole blood, a microfabricated nickel filter with a rectangular MCA (10(4) cavities/filter) was integrated with a miniaturized device, allowing for the isolation of tumor cells based on differences in size and deformability between tumor and blood cells. The shape and porosity of the MCA were optimized to efficiently capture small tumor cells on the microcavities under low flow resistance conditions, while allowing other blood cells to effectively pass through. Under optimized conditions, approximately 80% of SCLC (NCI-H69 and NCI-H82) cells spiked in 1 mL of whole blood were successfully recovered. In clinical samples, CTCs were detectable in 16 of 16 SCLC patients. In addition, the number of leukocytes captured on the rectangular MCA was significantly lower than that on the circular MCA (p < 0.001), suggesting that the use of the rectangular MCA diminishes a considerable number of carryover leukocytes. Therefore, our system has potential as a tool for the detection of CTCs in small cell-type tumors and detailed molecular analyses of CTCs.
Collapse
Affiliation(s)
- Masahito Hosokawa
- Division of Biotechnology and Life Science, Institute of Engineering, Tokyo University of Agriculture and Technology, Koganei, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Bayer CL, Joshi PP, Emelianov SY. Photoacoustic imaging: a potential tool to detect early indicators of metastasis. Expert Rev Med Devices 2013; 10:125-34. [PMID: 23278229 DOI: 10.1586/erd.12.62] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The metastasis of cancer is a multistage process involving complex biological interactions and difficult to predict outcomes. Accurate assessment of the extent of metastasis is critical for clinical practice; unfortunately, medical imaging methods capable of identifying the early stages of invasion and metastasis are lacking. Photoacoustic imaging is capable of providing noninvasive, real-time imaging of significant anatomical and physiological changes. indicating the progression of cancer invasion and metastasis. Preclinically, photoacoustic methods have been used to image lymphatic anatomy, including the sentinel lymph nodes, to identify circulating tumor cells within vasculature and to detect micrometastases. Progress has begun toward the development of clinically applicable photoacoustic imaging systems to assist with the determination of cancer stage and likelihood of metastatic invasion.
Collapse
Affiliation(s)
- Carolyn L Bayer
- Department of Biomedical Engineering, The University of Texas at Austin, 1 University Station, Austin, TX 78712, USA.
| | | | | |
Collapse
|
40
|
Cassatella MC, Zorzino L, Sandri MT. Single circulating tumor cell profiling: a new perspective for targeted therapy? Future Oncol 2013; 8:1253-6. [PMID: 23130926 DOI: 10.2217/fon.12.119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Evaluation of: Powell AA, Talasaz AH, Zhang H et al. Single cell profiling of circulating tumor cells: transcriptional heterogeneity and diversity from breast cancer cell lines. PLoS ONE 7(5), e33788 (2012). Circulating tumor cells (CTCs) may represent a possible useful tool to better define the prognosis of patients. The presence of CTCs can help to predict an increased risk for disease relapse, and they might be an early marker for treatment efficacy that could help in deciding treatment continuation. Cancer metastasis occurs when cells, shed from the primary tumor, enter the circulation and begin to grow in distant locations around the body. In metastatic stages, shed cells may differ from those of the primary tumor, as the tumor phenotype can change during the course of the disease. It is important to identify relevant targets expressed on these cells to provide clinical information on therapy choice, efficacy and drug resistance. Many efforts are now devoted to the characterization of the single cell. This article focuses on the possibility of profiling single CTCs in patients with breast cancer.
Collapse
Affiliation(s)
- M C Cassatella
- Laboratory Medicine Division, European Institute of Oncology, Via Ripamonti 435, 20141 Milan, Italy
| | | | | |
Collapse
|
41
|
Shen Q, Xu L, Zhao L, Wu D, Fan Y, Zhou Y, OuYang WH, Xu X, Zhang Z, Song M, Lee T, Garcia MA, Xiong B, Hou S, Tseng HR, Fang X. Specific capture and release of circulating tumor cells using aptamer-modified nanosubstrates. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2013; 25:2368-73. [PMID: 23495071 PMCID: PMC3786685 DOI: 10.1002/adma.201300082] [Citation(s) in RCA: 228] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Indexed: 05/19/2023]
Affiliation(s)
- Qinglin Shen
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan, Hubei, 430071 (P. R. China); Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Building 114, Los Angeles, CA 90095-1770 (USA), Web: http://tseng-lab.com
| | - Li Xu
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Science, Beiyi Street 2#, Zhongguancun, Beijing, 100190 (P.R. China)
| | - Libo Zhao
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Building 114, Los Angeles, CA 90095-1770 (USA), Web: http://tseng-lab.com Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Science, Beiyi Street 2#, Zhongguancun, Beijing, 100190 (P.R. China)
| | - Dongxia Wu
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Building 114, Los Angeles, CA 90095-1770 (USA), Web: http://tseng-lab.com
| | - Yunshan Fan
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Building 114, Los Angeles, CA 90095-1770 (USA), Web: http://tseng-lab.com
| | - Yiliang Zhou
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Building 114, Los Angeles, CA 90095-1770 (USA), Web: http://tseng-lab.com
| | - Wei-Han OuYang
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Building 114, Los Angeles, CA 90095-1770 (USA), Web: http://tseng-lab.com
| | - Xiaochun Xu
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Building 114, Los Angeles, CA 90095-1770 (USA), Web: http://tseng-lab.com
| | - Zhen Zhang
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Science, Beiyi Street 2#, Zhongguancun, Beijing, 100190 (P.R. China)
| | - Min Song
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Building 114, Los Angeles, CA 90095-1770 (USA), Web: http://tseng-lab.com
| | - Tom Lee
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Building 114, Los Angeles, CA 90095-1770 (USA), Web: http://tseng-lab.com
| | - Mitch A. Garcia
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Building 114, Los Angeles, CA 90095-1770 (USA), Web: http://tseng-lab.com
| | - Bin Xiong
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan, Hubei, 430071 (P. R. China)
| | - Shuang Hou
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Building 114, Los Angeles, CA 90095-1770 (USA), Web: http://tseng-lab.com
| | - Hsian-Rong Tseng
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Building 114, Los Angeles, CA 90095-1770 (USA), Web: http://tseng-lab.com
| | - Xiaohong Fang
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Science, Beiyi Street 2#, Zhongguancun, Beijing, 100190 (P.R. China)
| |
Collapse
|
42
|
Hou S, Zhao L, Shen Q, Yu J, Ng C, Kong X, Wu D, Song M, Shi X, Xu X, OuYang WH, He R, Zhao XZ, Lee T, Brunicardi FC, Garcia MA, Ribas A, Lo RS, Tseng HR. Polymer nanofiber-embedded microchips for detection, isolation, and molecular analysis of single circulating melanoma cells. Angew Chem Int Ed Engl 2013; 52:3379-83. [PMID: 23436302 PMCID: PMC3807678 DOI: 10.1002/anie.201208452] [Citation(s) in RCA: 181] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2012] [Revised: 12/20/2012] [Indexed: 12/19/2022]
Affiliation(s)
- Shuang Hou
- Department of Molecular and Medical Pharmacology, Crump Institute
for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI),
University of California, Los Angeles, 570 Westwood Plaza, Building 114, Los
Angeles, CA 90095-1770, USA, Web: http://www.tseng-lab.com
| | - Libo Zhao
- Department of Molecular and Medical Pharmacology, Crump Institute
for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI),
University of California, Los Angeles, 570 Westwood Plaza, Building 114, Los
Angeles, CA 90095-1770, USA
| | - Qinglin Shen
- Department of Molecular and Medical Pharmacology, Crump Institute
for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI),
University of California, Los Angeles, 570 Westwood Plaza, Building 114, Los
Angeles, CA 90095-1770, USA
- Department of Applied Physics and Department of Oncology Surgery,
Wuhan University, Wuhan, PRC
| | - Juehua Yu
- Department of Surgery, University of California, Los Angeles
| | - Charles Ng
- Division of Hematology and Oncology, Department of Medicine,
Department of Surgery, and Department of Molecular and Medical Pharmacology,
University of California, Los Angeles
| | - Xiangju Kong
- Division of Dermatology, Department of Medicine, University of
California, Los Angeles
| | - Dongxia Wu
- Department of Molecular and Medical Pharmacology, Crump Institute
for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI),
University of California, Los Angeles, 570 Westwood Plaza, Building 114, Los
Angeles, CA 90095-1770, USA
| | - Min Song
- Department of Molecular and Medical Pharmacology, Crump Institute
for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI),
University of California, Los Angeles, 570 Westwood Plaza, Building 114, Los
Angeles, CA 90095-1770, USA
| | - Xiaohong Shi
- Department of Molecular and Medical Pharmacology, Crump Institute
for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI),
University of California, Los Angeles, 570 Westwood Plaza, Building 114, Los
Angeles, CA 90095-1770, USA
| | - Xiaochun Xu
- CytoLumina Technologies Corp., 21038 Commerce Point Dr., Walnut,
CA 91789, USA
| | - Wei-Han OuYang
- CytoLumina Technologies Corp., 21038 Commerce Point Dr., Walnut,
CA 91789, USA
| | - Rongxian He
- Department of Applied Physics and Department of Oncology Surgery,
Wuhan University, Wuhan, PRC
| | - Xing-Zhong Zhao
- Department of Applied Physics and Department of Oncology Surgery,
Wuhan University, Wuhan, PRC
| | - Tom Lee
- Department of Molecular and Medical Pharmacology, Crump Institute
for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI),
University of California, Los Angeles, 570 Westwood Plaza, Building 114, Los
Angeles, CA 90095-1770, USA
| | | | - Mitch André Garcia
- CytoLumina Technologies Corp., 21038 Commerce Point Dr., Walnut,
CA 91789, USA
| | - Antoni Ribas
- Division of Hematology and Oncology, Department of Medicine,
Department of Surgery, and Department of Molecular and Medical Pharmacology,
University of California, Los Angeles
| | - Roger S. Lo
- Division of Dermatology, Department of Medicine, University of
California, Los Angeles
| | - Hsian-Rong Tseng
- Department of Molecular and Medical Pharmacology, Crump Institute
for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI),
University of California, Los Angeles, 570 Westwood Plaza, Building 114, Los
Angeles, CA 90095-1770, USA
| |
Collapse
|
43
|
Hou S, Zhao L, Shen Q, Yu J, Ng C, Kong X, Wu D, Song M, Shi X, Xu X, OuYang WH, He R, Zhao XZ, Lee T, Brunicardi FC, Garcia MA, Ribas A, Lo RS, Tseng HR. Polymer Nanofiber-Embedded Microchips for Detection, Isolation, and Molecular Analysis of Single Circulating Melanoma Cells. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201208452] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
44
|
Loutherback K, D'Silva J, Liu L, Wu A, Austin RH, Sturm JC. Deterministic separation of cancer cells from blood at 10 mL/min. AIP ADVANCES 2012; 2:42107. [PMID: 23112922 PMCID: PMC3477176 DOI: 10.1063/1.4758131] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2012] [Accepted: 09/21/2012] [Indexed: 05/07/2023]
Abstract
Circulating tumor cells (CTCs) and circulating clusters of cancer and stromal cells have been identified in the blood of patients with malignant cancer and can be used as a diagnostic for disease severity, assess the efficacy of different treatment strategies and possibly determine the eventual location of metastatic invasions for possible treatment. There is thus a critical need to isolate, propagate and characterize viable CTCs and clusters of cancer cells with their associated stroma cells. Here, we present a microfluidic device for mL/min flow rate, continuous-flow capture of viable CTCs from blood using deterministic lateral displacement (DLD) arrays. We show here that a DLD array device can isolate CTCs from blood with capture efficiency greater than 85% CTCs at volumetric flow rates of up to 10 mL/min with no effect on cell viability.
Collapse
Affiliation(s)
- Kevin Loutherback
- Princeton Institute for the Science and Technology of Materials (PRISM), Princeton Universtiy, Princeton, NJ, USA ; Department of Electrical Engineering, Princeton University, Princeton, NJ, USA
| | | | | | | | | | | |
Collapse
|
45
|
Yu C, Shiozawa Y, Taichman RS, McCauley LK, Pienta K, Keller E. Prostate cancer and parasitism of the bone hematopoietic stem cell niche. Crit Rev Eukaryot Gene Expr 2012; 22:131-48. [PMID: 22856431 DOI: 10.1615/critreveukargeneexpr.v22.i2.50] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
A subpopulation of men that appear cured of prostate cancer (PCa) develop bone metastases many years after prostatectomy. This observation indicates that PCa cells were present outside of the prostate at the time of prostatectomy and remained dormant. Several lines of evidence indicate that there are disseminated tumor cells (DTCs) in the bone marrow at the time of prostatectomy. DTCs parasitize the bone microenvironment, where they derive support and impact the microenvironment itself. These DTCs appear to be a heterogeneous population of PCa cells; however, some of them appear to have some aspects of a cancer stem cell (CSC) phenotype as they can develop into clinically detectable metastases. The concept of CSC is controversial; however, several markers of CSC have been identified for PCa, which may represent cells of either basal or luminal origin. These DTCs have now been shown to compete for the hematopoietic stem cell niche in bone, where they may be placed in a dormant state. Interaction with a variety of host factors, including cytokine and cells, may impact the metastatic development and progression, including the dormant state. For example, myeloid cells have been shown to impact both the premetastatic niche and established tumors. Understanding the concepts of how PCa successfully parasitizes the bone microenvironment is paramount toward identifying therapeutic candidates to prevent or diminish PCa bone metastases.
Collapse
Affiliation(s)
- Chunyan Yu
- Department of Urology, School of Medicine, University of Michigan, Ann Arbor, MI 48109-0940, USA
| | | | | | | | | | | |
Collapse
|
46
|
Abstract
Our understanding of the role of lymph nodes (LN) in the metastasization process (MET) is marginal. Positive LNs (pLN) are the most important prognostic factor and lymph node dissection (LND) is still standard practice in primary treatment. However, up to now, there is almost no evidence that elective LND has a survival benefit. Based on many clinical and experimental findings, we propose that tumor foci in regional LN are incapable of metastasization and can therefore not infiltrate further LN and organs. Available data demonstrate a very early infiltration of MET capable tumor cells from the primary tumor into regional LN, and thereafter an increased probability of subsequent LN infiltrations. Disparate growth rates of the first versus subsequent infiltrating tumors as well as the asymptotic growth and prognosis of large tumor foci in LN explain many clinical observations for solid tumors. The consequence of the hypothesis "pLN do not metastasize" would impact clinical treatment and research and contribute to understanding the mounting evidence against LND.
Collapse
Affiliation(s)
- Jutta Engel
- Ludwig-Maximilians-University, Clinic Großhadern, Munich, Germany
| | | | | |
Collapse
|
47
|
Burdick MM, Henson KA, Delgadillo LF, Choi YE, Goetz DJ, Tees DFJ, Benencia F. Expression of E-selectin ligands on circulating tumor cells: cross-regulation with cancer stem cell regulatory pathways? Front Oncol 2012; 2:103. [PMID: 22934288 PMCID: PMC3422812 DOI: 10.3389/fonc.2012.00103] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 08/02/2012] [Indexed: 12/19/2022] Open
Abstract
Although significant progress has been made in the fight against cancer, successful treatment strategies have yet to be developed to combat those tumors that have metastasized to distant organs. Poor characterization of the molecular mechanisms of cancer spread is a major impediment to designing predictive diagnostics and effective clinical interventions against late stage disease. In hematogenous metastasis, it is widely suspected that circulating tumor cells (CTCs) express specific adhesion molecules that actively initiate contact with the vascular endothelium lining the vessel walls of the target organ. This "tethering" is mediated by ligands expressed by CTCs that bind to E-selectin expressed by endothelial cells. However, it is currently unknown whether expression of functional E-selectin ligands on CTCs is related to cancer stem cell regulatory or maintenance pathways, particularly epithelial-to-mesenchymal transition and the reverse, mesenchymal-to-epithelial transition. In this hypothesis and theory article, we explore the potential roles of these mechanisms on the dynamic regulation of selectin ligands mediating CTC trafficking during metastasis.
Collapse
Affiliation(s)
- Monica M Burdick
- Department of Chemical and Biomolecular Engineering, Russ College of Engineering and Technology, Ohio University Athens, OH, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Barbazán J, Alonso-Alconada L, Muinelo-Romay L, Vieito M, Abalo A, Alonso-Nocelo M, Candamio S, Gallardo E, Fernández B, Abdulkader I, de Los Ángeles Casares M, Gómez-Tato A, López-López R, Abal M. Molecular characterization of circulating tumor cells in human metastatic colorectal cancer. PLoS One 2012; 7:e40476. [PMID: 22811761 PMCID: PMC3397799 DOI: 10.1371/journal.pone.0040476] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 06/08/2012] [Indexed: 12/29/2022] Open
Abstract
Metastatic colorectal cancer (mCRC) relies on the detachment of aggressive malignant cells from the primary tumor into the bloodstream and, concordantly, the presence of these Circulating Tumor Cells (CTC) is associated with a poor prognosis. In this work, the molecular characterization of CTC from mCRC patients was approached, with the aim of understanding their biology and improving their clinical utility in the management of colorectal cancer patients. For this, EpCAM-based immunoisolation of CTC was combined with whole transcriptome amplification and hybridization onto cDNA microarrays. Gene expression data from mCRC patients, once the background of unspecific immunoisolation from a group of controls had been subtracted, resulted in 410 genes that characterized the CTC population. Bioinformatics were used for the biological interpretation of the data, revealing that CTC are characterized by genes related to cell movement and adhesion, cell death and proliferation, and cell signalling and interaction. RTqPCR on an independent series of mCRC patients and controls was used for the validation of a number of genes related to the main cellular functions characterizing the CTC population. Comparison between primary carcinomas and lung and liver metastases further involved the CTC-genes in the promotion of metastasis. Moreover, the correlation of CTC-gene expression with clinical parameters demonstrated detection and prognosis significance. In conclusion, the molecular characterization of CTC from mCRC patients and the identification of diagnostic and prognostic biomarkers represent an innovative and promising approach in the clinical management of this type of patients.
Collapse
Affiliation(s)
- Jorge Barbazán
- Translational Laboratory, Medical Oncology Department, Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Santana SM, Liu H, Bander NH, Gleghorn JP, Kirby BJ. Immunocapture of prostate cancer cells by use of anti-PSMA antibodies in microdevices. Biomed Microdevices 2012; 14:401-7. [PMID: 22143878 DOI: 10.1007/s10544-011-9616-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Patients suffering from cancer can shed tumor cells into the bloodstream, leading to one of the most important mechanisms of metastasis. As such, the capture of these cells is of great interest. Circulating tumor cells are typically extracted from circulation through positive selection with the epithelial cell-adhesion molecule (EpCAM), leading to currently unknown biases when cells are undergoing epithelial-to-mesenchymal transition. For prostate cancer, prostate-specific membrane antigen (PSMA) presents a compelling target for immunocapture, as PSMA levels increase in higher-grade cancers and metastatic disease and are specific to the prostate epithelium. This study uses monoclonal antibodies J591 and J415-antibodies that are highly specific for intact extracellular domains of PSMA on live cells-in microfluidic devices for the capture of LNCaPs, a PSMA-expressing immortalized prostate cancer cell line, over a range of concentrations and shear stresses relevant to immunocapture. Our results show that J591 outperforms J415 and a mix of the two for prostate cancer capture, and that capture performance saturates following incubation with antibody concentrations of 10 micrograms per milliliter.
Collapse
Affiliation(s)
- Steven M Santana
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, 245 Upson Hall, Ithaca, NY 14853, USA.
| | | | | | | | | |
Collapse
|
50
|
|