201
|
Xu Y, Wang X. Fluid and cell behaviors along a 3D printed alginate/gelatin/fibrin channel. Biotechnol Bioeng 2015; 112:1683-95. [PMID: 25727058 DOI: 10.1002/bit.25579] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 02/17/2015] [Indexed: 02/06/2023]
Abstract
Three-dimensional (3D) cell manipulation is available with the integration of microfluidic technology and rapid prototyping techniques. High-Fidelity (Hi-Fi) constructs hold enormous therapeutic potential for organ manufacturing and regenerative medicine. In the present paper we introduced a quasi-three-dimensional (Q3D) model with parallel biocompatible alginate/gelatin/fibrin hurdles. The behaviors of fluids and cells along the microfluidic channels with various widths were studied. Cells inside the newly designed microfluidic channels attached and grew well. Morphological changes of adipose-derived stem cells (ADSCs) in both two-dimensional (2D) and 3D milieu were found on the printed constructs. Endothelialization occurred with the co-cultures of ADSCs and hepatocytes. This study provides insights into the interactions among fluids, cells and biomaterials, the behaviors of fluids and cells along the microfluidic channels, and the applications of Q3D techniques.
Collapse
Affiliation(s)
- Yufan Xu
- Department of Mechanical Engineering, Key Laboratory for Advanced Materials Processing Technology, Ministry of Education & Center of Organ Manufacturing, Tsinghua University, Beijing, 100084, P.R. China
| | - Xiaohong Wang
- Department of Mechanical Engineering, Key Laboratory for Advanced Materials Processing Technology, Ministry of Education & Center of Organ Manufacturing, Tsinghua University, Beijing, 100084, P.R. China. .,State Key Laboratory of Materials Processing and Die & Mould Technology, Huazhong University of Science and Technology, Wuhan, 430074, P.R. China.
| |
Collapse
|
202
|
Xue P, Wu Y, Guo J, Kang Y. Highly efficient capture and harvest of circulating tumor cells on a microfluidic chip integrated with herringbone and micropost arrays. Biomed Microdevices 2015; 17:39. [DOI: 10.1007/s10544-015-9945-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
203
|
Chen P, Huang YY, Hoshino K, Zhang JX. Microscale magnetic field modulation for enhanced capture and distribution of rare circulating tumor cells. Sci Rep 2015; 5:8745. [PMID: 25735563 PMCID: PMC4348664 DOI: 10.1038/srep08745] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 02/02/2015] [Indexed: 01/01/2023] Open
Abstract
Immunomagnetic assay combines the powers of the magnetic separation and biomarker recognition and has been an effective tool to perform rare Circulating Tumor Cells detection. Key factors associated with immunomagnetic assay include the capture rate, which indicates the sensitivity of the system, and distributions of target cells after capture, which impact the cell integrity and other biological properties that are critical to downstream analyses. Here we present a theoretical framework and technical approach to implement a microscale magnetic immunoassay through modulating local magnetic field towards enhanced capture and distribution of rare cancer cells. Through the design of a two-dimensional micromagnet array, we characterize the magnetic field generation and quantify the impact of the micromagnets on rare cell separation. Good agreement is achieved between the theory and experiments using a human colon cancer cell line (COLO205) as the capture targets.
Collapse
Affiliation(s)
- Peng Chen
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Yu-Yen Huang
- Thayer School of Engineering, Dartmouth College, NH 03755, USA
| | - Kazunori Hoshino
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - John X.J. Zhang
- Thayer School of Engineering, Dartmouth College, NH 03755, USA
| |
Collapse
|
204
|
Ma Y, Hao S, Wang S, Zhao Y, Lim B, Lei M, Spector DJ, El-Deiry WS, Zheng SY, Zhu J. A Combinatory Strategy for Detection of Live CTCs Using Microfiltration and a New Telomerase-Selective Adenovirus. Mol Cancer Ther 2015; 14:835-43. [PMID: 25589497 DOI: 10.1158/1535-7163.mct-14-0693] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 12/29/2014] [Indexed: 01/12/2023]
Abstract
Circulating tumor cells (CTC) have become an important biomarker for early cancer diagnosis, prognosis, and treatment monitoring. Recently, a replication-competent recombinant adenovirus driven by a human telomerase gene (hTERT) promoter was shown to detect live CTCs in blood samples of patients with cancer. Here, we report a new class of adenoviruses containing regulatory elements that repress the hTERT gene in normal cells. Compared with the virus with only the hTERT core promoter, the new viruses showed better selectivity for replication in cancer cells than in normal cells. In particular, Ad5GTSe, containing three extra copies of a repressor element, displayed a superior tropism for cancer cells among leukocytes and was thus selected for CTC detection in blood samples. To further improve the efficiency and specificity of CTC identification, we tested a combinatory strategy of microfiltration enrichment using flexible micro spring arrays and Ad5GTSe imaging. Our experiments showed that this method efficiently detected both cancer cells spiked into healthy blood and potential CTCs in blood samples of patients with breast and pancreatic cancer, demonstrating its potential as a highly sensitive and reliable system for detection and capture of CTCs of different tumor types.
Collapse
Affiliation(s)
- Yanchun Ma
- College of Life Science, Northwest A&F University, Taicheng Road, Yangling, Shaanxi, China. Department of C&M Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Sijie Hao
- Department of C&M Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Shuwen Wang
- Department of C&M Physiology, Penn State College of Medicine, Hershey, Pennsylvania. Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington
| | - Yuanjun Zhao
- Department of C&M Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Bora Lim
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington
| | - Ming Lei
- College of Life Science, Northwest A&F University, Taicheng Road, Yangling, Shaanxi, China
| | - David J Spector
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Wafik S El-Deiry
- Division of Hematology-Oncology, Penn State Hershey Cancer Institute, Hershey, Pennsylvania
| | - Si-Yang Zheng
- Micro & Nano Integrated Biosystem Laboratory, Department of Biomedical Engineering and Materials Research Institute, Pennsylvania State University, University Park, Pennsylvania
| | - Jiyue Zhu
- Department of C&M Physiology, Penn State College of Medicine, Hershey, Pennsylvania. Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington.
| |
Collapse
|
205
|
Pang L, Shen S, Ma C, Ma T, Zhang R, Tian C, Zhao L, Liu W, Wang J. Deformability and size-based cancer cell separation using an integrated microfluidic device. Analyst 2015; 140:7335-46. [DOI: 10.1039/c5an00799b] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We present an integrated microfluidic device for cell separation based on the cell size and deformability by combining the microstructure-constricted filtration and pneumatic microvalves.
Collapse
Affiliation(s)
- Long Pang
- Colleges of Veterinary Medicine and Science
- Northwest A&F University
- Yangling
- P. R. China
| | - Shaofei Shen
- Colleges of Veterinary Medicine and Science
- Northwest A&F University
- Yangling
- P. R. China
| | - Chao Ma
- Colleges of Veterinary Medicine and Science
- Northwest A&F University
- Yangling
- P. R. China
| | - Tongtong Ma
- Colleges of Veterinary Medicine and Science
- Northwest A&F University
- Yangling
- P. R. China
| | - Rui Zhang
- Department of Biochemistry & Biophysics
- Texas A&M University College Station
- USA
| | - Chang Tian
- Colleges of Veterinary Medicine and Science
- Northwest A&F University
- Yangling
- P. R. China
| | - Lei Zhao
- Colleges of Veterinary Medicine and Science
- Northwest A&F University
- Yangling
- P. R. China
| | - Wenming Liu
- Colleges of Veterinary Medicine and Science
- Northwest A&F University
- Yangling
- P. R. China
| | - Jinyi Wang
- Colleges of Veterinary Medicine and Science
- Northwest A&F University
- Yangling
- P. R. China
| |
Collapse
|
206
|
Lee SW, Hyun KA, Kim SI, Kang JY, Jung HI. Continuous enrichment of circulating tumor cells using a microfluidic lateral flow filtration chip. J Chromatogr A 2015; 1377:100-5. [DOI: 10.1016/j.chroma.2014.12.037] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 12/08/2014] [Accepted: 12/10/2014] [Indexed: 10/24/2022]
|
207
|
Wu W, Trinh KTL, Lee NY. Flow-through polymerase chain reaction inside a seamless 3D helical microreactor fabricated utilizing a silicone tube and a paraffin mold. Analyst 2015; 140:1416-20. [DOI: 10.1039/c4an01675k] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Seamless 3D helical silicone tube microreactors were fabricated for performing flow-through PCR employing a single hot plate and a portable micropump.
Collapse
Affiliation(s)
- Wenming Wu
- Department of BioNano Technology
- Gachon University
- Seongnam-si
- Korea
| | | | - Nae Yoon Lee
- Department of BioNano Technology
- Gachon University
- Seongnam-si
- Korea
- Gachon Medical Research Institute
| |
Collapse
|
208
|
Diéguez L, Winter MA, Pocock KJ, Bremmell KE, Thierry B. Efficient microfluidic negative enrichment of circulating tumor cells in blood using roughened PDMS. Analyst 2015; 140:3565-72. [DOI: 10.1039/c4an01768d] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Depletion of >99.7% WBCs enabling tumor cell recovery from blood with nano-rough PDMS microfluidic negative enrichment devices functionalised with anti-CD45.
Collapse
Affiliation(s)
- L. Diéguez
- Ian Wark Research Institute
- University of South Australia
- Australia
| | - M. A. Winter
- Ian Wark Research Institute
- University of South Australia
- Australia
| | - K. J. Pocock
- Ian Wark Research Institute
- University of South Australia
- Australia
| | - K. E. Bremmell
- School of Pharmacy and Medical Sciences
- University of South Australia
- Adelaide
- Australia
| | - B. Thierry
- Ian Wark Research Institute
- University of South Australia
- Australia
| |
Collapse
|
209
|
Toss A, Mu Z, Fernandez S, Cristofanilli M. CTC enumeration and characterization: moving toward personalized medicine. ANNALS OF TRANSLATIONAL MEDICINE 2014; 2:108. [PMID: 25489582 DOI: 10.3978/j.issn.2305-5839.2014.09.06] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 08/28/2014] [Indexed: 12/27/2022]
Abstract
The primary cause of tumor-related death in breast cancer (BC) is still represented by distant metastasization. The dissemination of tumor cells from the primary tumor to distant sites through bloodstream cannot be early detected by standard imaging methods. The enumeration of circulating tumor cells (CTCs) represents an effective prognostic and predictive biomarker, which is able to monitor efficacy of adjuvant therapies, detect early development of (micro)metastases and at last, assess therapeutic responses of advanced disease earlier than traditional imaging methods. Moreover, since repeated tissue biopsies are invasive, costly and not always feasible, the assessment of tumor characteristics on CTCs, by a peripheral blood sample as a 'liquid biopsy', represents an attractive opportunity. The implementation of molecular and genomic characterization of CTCs could contribute to improve the treatment selection and thus, to move toward more personalized treatments. This review describes the current state of the art on CTC detection strategies, the evidence to demonstrate their clinical validity, and their potential impact for both future clinical trial design and, decision-making process in our daily practice.
Collapse
Affiliation(s)
- Angela Toss
- 1 Department of Oncology, Haematology and Respiratory Diseases, University of Modena and Reggio Emilia, Modena, Italy ; 2 Department of Medical Oncology, Thomas Jefferson University & Kimmel Cancer Center, Philadelphia, PA, USA
| | - Zhaomei Mu
- 1 Department of Oncology, Haematology and Respiratory Diseases, University of Modena and Reggio Emilia, Modena, Italy ; 2 Department of Medical Oncology, Thomas Jefferson University & Kimmel Cancer Center, Philadelphia, PA, USA
| | - Sandra Fernandez
- 1 Department of Oncology, Haematology and Respiratory Diseases, University of Modena and Reggio Emilia, Modena, Italy ; 2 Department of Medical Oncology, Thomas Jefferson University & Kimmel Cancer Center, Philadelphia, PA, USA
| | - Massimo Cristofanilli
- 1 Department of Oncology, Haematology and Respiratory Diseases, University of Modena and Reggio Emilia, Modena, Italy ; 2 Department of Medical Oncology, Thomas Jefferson University & Kimmel Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
210
|
Zhou MD, Hao S, Williams AJ, Harouaka RA, Schrand B, Rawal S, Ao Z, Brennaman R, Gilboa E, Lu B, Wang S, Zhu J, Datar R, Cote R, Tai YC, Zheng SY. Separable bilayer microfiltration device for viable label-free enrichment of circulating tumour cells. Sci Rep 2014; 4:7392. [PMID: 25487434 PMCID: PMC4260227 DOI: 10.1038/srep07392] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 11/20/2014] [Indexed: 01/18/2023] Open
Abstract
The analysis of circulating tumour cells (CTCs) in cancer patients could provide important information for therapeutic management. Enrichment of viable CTCs could permit performance of functional analyses on CTCs to broaden understanding of metastatic disease. However, this has not been widely accomplished. Addressing this challenge, we present a separable bilayer (SB) microfilter for viable size-based CTC capture. Unlike other single-layer CTC microfilters, the precise gap between the two layers and the architecture of pore alignment result in drastic reduction in mechanical stress on CTCs, capturing them viably. Using multiple cancer cell lines spiked in healthy donor blood, the SB microfilter demonstrated high capture efficiency (78-83%), high retention of cell viability (71-74%), high tumour cell enrichment against leukocytes (1.7-2 × 10(3)), and widespread ability to establish cultures post-capture (100% of cell lines tested). In a metastatic mouse model, SB microfilters successfully enriched viable mouse CTCs from 0.4-0.6 mL whole mouse blood samples and established in vitro cultures for further genetic and functional analysis. Our preliminary studies reflect the efficacy of the SB microfilter device to efficiently and reliably enrich viable CTCs in animal model studies, constituting an exciting technology for new insights in cancer research.
Collapse
Affiliation(s)
- Ming-Da Zhou
- Micro & Nano Integrated Biosystem (MINIBio) Laboratory, Department of Biomedical Engineering and Materials Research Institute, Pennsylvania State University, University Park, PA 16802, U.S.A.
| | - Sijie Hao
- Micro & Nano Integrated Biosystem (MINIBio) Laboratory, Department of Biomedical Engineering and Materials Research Institute, Pennsylvania State University, University Park, PA 16802, U.S.A.
| | - Anthony J. Williams
- Department of Pathology, University of Miami – Miller School of Medicine, Miami, FL 33136, U.S.A.
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami – Miller School of Medicine, Miami, FL 33136, U.S.A.
- Dr John T Macdonald Foundation Biomedical Nanotechnology Institute, University of Miami – Miller School of Medicine, Miami, FL 33136, U.S.A.
| | - Ramdane A. Harouaka
- Micro & Nano Integrated Biosystem (MINIBio) Laboratory, Department of Biomedical Engineering and Materials Research Institute, Pennsylvania State University, University Park, PA 16802, U.S.A.
| | - Brett Schrand
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami – Miller School of Medicine, Miami, FL 33136, U.S.A.
- Department of Microbiology and Immunology, University of Miami – Miller School of Medicine, Miami, FL 33136, U.S.A.
| | - Siddarth Rawal
- Department of Pathology, University of Miami – Miller School of Medicine, Miami, FL 33136, U.S.A.
- Dr John T Macdonald Foundation Biomedical Nanotechnology Institute, University of Miami – Miller School of Medicine, Miami, FL 33136, U.S.A.
| | - Zheng Ao
- Department of Pathology, University of Miami – Miller School of Medicine, Miami, FL 33136, U.S.A.
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami – Miller School of Medicine, Miami, FL 33136, U.S.A.
- Dr John T Macdonald Foundation Biomedical Nanotechnology Institute, University of Miami – Miller School of Medicine, Miami, FL 33136, U.S.A.
| | - Randall Brennaman
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami – Miller School of Medicine, Miami, FL 33136, U.S.A.
- Department of Microbiology and Immunology, University of Miami – Miller School of Medicine, Miami, FL 33136, U.S.A.
| | - Eli Gilboa
- Department of Microbiology and Immunology, University of Miami – Miller School of Medicine, Miami, FL 33136, U.S.A.
| | - Bo Lu
- Caltech Micromachining Laboratory, California Institute of Technology, MC 136-93, Pasadena, CA 91125, U.S.A.
| | - Shuwen Wang
- Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, WA 99210, U.S.A
| | - Jiyue Zhu
- Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, WA 99210, U.S.A
| | - Ram Datar
- Department of Pathology, University of Miami – Miller School of Medicine, Miami, FL 33136, U.S.A.
- Dr John T Macdonald Foundation Biomedical Nanotechnology Institute, University of Miami – Miller School of Medicine, Miami, FL 33136, U.S.A.
| | - Richard Cote
- Department of Pathology, University of Miami – Miller School of Medicine, Miami, FL 33136, U.S.A.
- Dr John T Macdonald Foundation Biomedical Nanotechnology Institute, University of Miami – Miller School of Medicine, Miami, FL 33136, U.S.A.
| | - Yu-Chong Tai
- Caltech Micromachining Laboratory, California Institute of Technology, MC 136-93, Pasadena, CA 91125, U.S.A.
| | - Si-Yang Zheng
- Micro & Nano Integrated Biosystem (MINIBio) Laboratory, Department of Biomedical Engineering and Materials Research Institute, Pennsylvania State University, University Park, PA 16802, U.S.A.
| |
Collapse
|
211
|
Mego M, Reuben JM. Prognostic and Predictive Role of Circulating Tumor Cells in Breast Cancer. CURRENT BREAST CANCER REPORTS 2014. [DOI: 10.1007/s12609-014-0164-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
212
|
Lee A, Park J, Lim M, Sunkara V, Kim SY, Kim GH, Kim MH, Cho YK. All-in-one centrifugal microfluidic device for size-selective circulating tumor cell isolation with high purity. Anal Chem 2014; 86:11349-11356. [PMID: 25317565 DOI: 10.1021/ac5035049] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Circulating tumor cells (CTCs) have gained increasing attention owing to their roles in cancer recurrence and progression. Due to the rarity of CTCs in the bloodstream, an enrichment process is essential for effective target cell characterization. However, in a typical pressure-driven microfluidic system, the enrichment process generally requires complicated equipment and long processing times. Furthermore, the commonly used immunoaffinity-based positive selection method is limited, as its recovery rate relies on EpCAM expression of target CTCs, which shows heterogeneity among cell types. Here, we propose a centrifugal-force-based size-selective CTC isolation platform that can isolate and enumerate CTCs from whole blood within 30 s with high purity. The device was validated using the MCF-7 breast cancer cell line spiked in phosphate-buffered saline and whole blood, and an average capture efficiency of 61% was achieved, which is typical for size-based filtration. The capture efficiency for whole blood samples varied from 44% to 84% under various flow conditions and dilution factors. Under the optimized operating conditions, a few hundred white blood cells per 1 mL of whole blood were captured, representing a 20-fold decrease compared to those obtained using a commercialized size-based CTC isolation device. In clinical validation, normalized CTC counts varied from 10 to 60 per 7.5 mL of blood from gastric and lung cancer patients, yielding a detection rate of 50% and 38%, respectively. Overall, our CTC isolation device enables rapid and label-free isolation of CTCs with high purity, which should greatly improve downstream molecular analyses of captured CTCs.
Collapse
Affiliation(s)
- Ada Lee
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST) , UNIST-gil 50, Ulsan, 689-798, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
213
|
Kim TH, Yoon HJ, Stella P, Nagrath S. Cascaded spiral microfluidic device for deterministic and high purity continuous separation of circulating tumor cells. BIOMICROFLUIDICS 2014; 8:064117. [PMID: 25553193 PMCID: PMC4257960 DOI: 10.1063/1.4903501] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 11/24/2014] [Indexed: 05/03/2023]
Abstract
Inertial microfluidics is an emerging class of technologies developed to separate circulating tumor cells (CTCs). However, defining design parameters and flow conditions for optimal operation remains nondeterministic due to incomplete understanding of the mechanics, which has led to challenges in designing efficient systems. Here, we perform a parametric study of the inertial focusing effects observed in low aspect ratio curvilinear microchannels and utilize the results to demonstrate the isolation of CTCs with high purity. First, we systematically vary parameters including the channel height, width, and radius of curvature over a wide range of flow velocities to analyze its effect on size dependent differential focusing and migration behaviors of binary (10 μm and 20 μm) particles. Second, we use these results to identify optimal flow regimes to achieve maximum separation in various channel configurations and establish design guidelines to readily provide information for developing spiral channels tailored to potentially arbitrary flow conditions that yield a desired equilibrium position for optimal size based CTC separation. Finally, we describe a fully integrated, sheath-less cascaded spiral microfluidic device to continuously isolate CTCs. Human breast cancer epithelial cells were successfully extracted from leukocytes, achieving 86.76% recovery, 97.91% depletion rate, and sustaining high viability upon collection to demonstrate the versatility of the device. Importantly, this device was designed without the cumbersome trail-and-error optimization process that has hindered the development of designing such inertial microfluidic systems.
Collapse
Affiliation(s)
| | | | - Philip Stella
- Department of Chemical and Biomolecular Engineering, University of Notre Dame , Notre Dame, Indiana 46556, USA
| | | |
Collapse
|
214
|
Song Y, Li M, Pan X, Wang Q, Li D. Size-based cell sorting with a resistive pulse sensor and an electromagnetic pump in a microfluidic chip. Electrophoresis 2014; 36:398-404. [DOI: 10.1002/elps.201400292] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 08/03/2014] [Accepted: 08/11/2014] [Indexed: 12/24/2022]
Affiliation(s)
- Yongxin Song
- Department of Marine Engineering; Dalian Maritime University; Dalian P. R. China
| | - Mengqi Li
- Department of Marine Engineering; Dalian Maritime University; Dalian P. R. China
| | - Xinxiang Pan
- Department of Marine Engineering; Dalian Maritime University; Dalian P. R. China
| | - Qi Wang
- Department of Respiratory Medicine; The second affiliated hospital of Dalian Medical University; Dalian P. R. China
| | - Dongqing Li
- Department of Mechanical and Mechatronics Engineering; University of Waterloo; Waterloo Ontario Canada
| |
Collapse
|
215
|
Cup-shaped superparamagnetic hemispheres for size-selective cell filtration. Sci Rep 2014; 4:6362. [PMID: 25219418 PMCID: PMC4163672 DOI: 10.1038/srep06362] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 08/07/2014] [Indexed: 12/23/2022] Open
Abstract
We propose a new method of size separation of cells exploiting precisely size-controlled hemispherical superparamagnetic microparticles. A three-layered structure of a 2-nm nickel layer inserted between 15-nm silicon dioxide layers was formed on polystyrene cast spheres by vapor deposition. The polystyrene was then removed by burning and the hemispherical superparamagnetic microparticles, “magcups”, were obtained. The standard target cells (CCRF-CEM, 12 ± 2 μm) were mixed with a set of different sizes of the fabricated magcups, and we confirmed that the cells were captured in the magcups having cavities larger than 15 μm in diameter, and then gathered by magnetic force. The collected cells were grown in a culture medium without any damage. The results suggest that this method is quick, simple and non-invasive size separation of target cells.
Collapse
|
216
|
Yabusaki M, Sato J, Kohyama A, Kojima T, Nobuoka D, Yoshikawa T, Sawada Y, Murakami K, Gohda K, Okegawa T, Nakamura M, Takamatsu K, Ito M, Kaneko K, Nakatsura T. Detection and preliminary evaluation of circulating tumor cells in the peripheral blood of patients with eight types of cancer using a telomerase-specific adenovirus. Oncol Rep 2014; 32:1772-8. [PMID: 25176113 PMCID: PMC4203327 DOI: 10.3892/or.2014.3436] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 07/02/2014] [Indexed: 01/23/2023] Open
Abstract
We developed a detection method for circulating tumor cells (CTCs) using the telomerase-specific adenovirus OBP-401. This recombinant virus has a telomerase promoter at the 5′-end of the viral genome and GFP at the 3′-end. To date, CTC enumeration using OBP-401 has shown prognostic impact for gastric and small cell lung cancer patients. In the present study, peripheral blood samples from patients with eight types of cancer, including some cancers previously untested with OBP-401 (i.e., esophagus, pancreas, and prostate cancers) were subjected to this method in order to evaluate its versatility. It was recently discovered that some white blood cells (WBCs) false-positively react with OBP-401. Although anti-CD45 antibodies can absorb these adverse cells from peripheral blood, the simplicity of the OBP-401 method would be diminished by the introduction of antibody treatment. Therefore, we evaluated another approach to minimize the false positivity of WBCs. Seven anti-CD antibodies were employed to stain the species of WBCs that false-positively reacted with OBP-401. We revealed that the false-positively reacted WBCs were monocytes in the peripheral blood of both healthy subjects and cancer patients. Based on a size distribution analysis of the GFP-positive monocytes, the size criterion for CTCs using OBP-401 was defined to be a cellular diameter >8.4 μm. In total, 43% of 86 cancer patients examined in the present study were CTC-positive using this definition. CTCs were enumerated from peripheral blood samples collected from patients with each of the eight types of cancer; the detectability of CTCs for esophagus, pancreas and prostate cancers by the OBP-401 method was confirmed for the first time in the present study. However, no clear correlation between CTC positivity and the clinical characteristics of patients with any type of cancer was observed because of the small number of patients with each type of cancer. An additional clinical study will be conducted to confirm the clinical meaning of CTCs enumerated by OBP-401.
Collapse
Affiliation(s)
- Mina Yabusaki
- Central Research Laboratories, Sysmex Corporation, Kobe, Hyogo, Japan
| | - Jun Sato
- Central Research Laboratories, Sysmex Corporation, Kobe, Hyogo, Japan
| | - Atsushi Kohyama
- Division of Colorectal and Pelvic Surgery, National Cancer Center Hospital East, Kashiwanoha, Kashiwa, Chiba, Japan
| | - Takashi Kojima
- Department of Gastroenterology, Endoscopy Division, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Daisuke Nobuoka
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| | - Toshiaki Yoshikawa
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| | - Yu Sawada
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| | | | - Keigo Gohda
- Central Research Laboratories, Sysmex Corporation, Kobe, Hyogo, Japan
| | - Takatsugu Okegawa
- Department of Urology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Masaru Nakamura
- Department of Obstetrics and Gynecology, Tokyo Dental College, Ichikawa General Hospital, Ichikawa, Chiba, Japan
| | - Kiyoshi Takamatsu
- Department of Obstetrics and Gynecology, Tokyo Dental College, Ichikawa General Hospital, Ichikawa, Chiba, Japan
| | - Masaaki Ito
- Division of Colorectal and Pelvic Surgery, National Cancer Center Hospital East, Kashiwanoha, Kashiwa, Chiba, Japan
| | - Kazuhiro Kaneko
- Department of Gastroenterology, Endoscopy Division, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Tetsuya Nakatsura
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| |
Collapse
|
217
|
Kim H, Terazono H, Nakamura Y, Sakai K, Hattori A, Odaka M, Girault M, Arao T, Nishio K, Miyagi Y, Yasuda K. Development of on-chip multi-imaging flow cytometry for identification of imaging biomarkers of clustered circulating tumor cells. PLoS One 2014; 9:e104372. [PMID: 25140522 PMCID: PMC4139271 DOI: 10.1371/journal.pone.0104372] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 07/10/2014] [Indexed: 01/10/2023] Open
Abstract
An on-chip multi-imaging flow cytometry system has been developed to obtain morphometric parameters of cell clusters such as cell number, perimeter, total cross-sectional area, number of nuclei and size of clusters as “imaging biomarkers”, with simultaneous acquisition and analysis of both bright-field (BF) and fluorescent (FL) images at 200 frames per second (fps); by using this system, we examined the effectiveness of using imaging biomarkers for the identification of clustered circulating tumor cells (CTCs). Sample blood of rats in which a prostate cancer cell line (MAT-LyLu) had been pre-implanted was applied to a microchannel on a disposable microchip after staining the nuclei using fluorescent dye for their visualization, and the acquired images were measured and compared with those of healthy rats. In terms of the results, clustered cells having (1) cell area larger than 200 µm2 and (2) nucleus area larger than 90 µm2 were specifically observed in cancer cell-implanted blood, but were not observed in healthy rats. In addition, (3) clusters having more than 3 nuclei were specific for cancer-implanted blood and (4) a ratio between the actual perimeter and the perimeter calculated from the obtained area, which reflects a shape distorted from ideal roundness, of less than 0.90 was specific for all clusters having more than 3 nuclei and was also specific for cancer-implanted blood. The collected clusters larger than 300 µm2 were examined by quantitative gene copy number assay, and were identified as being CTCs. These results indicate the usefulness of the imaging biomarkers for characterizing clusters, and all of the four examined imaging biomarkers—cluster area, nuclei area, nuclei number, and ratio of perimeter—can identify clustered CTCs in blood with the same level of preciseness using multi-imaging cytometry.
Collapse
Affiliation(s)
- Hyonchol Kim
- Kanagawa Academy of Science and Technology, Takatsu, Kawasaki, Japan
| | - Hideyuki Terazono
- Kanagawa Academy of Science and Technology, Takatsu, Kawasaki, Japan
- Department of Biomedical Information, Division of Biosystems, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Chiyoda, Tokyo, Japan
| | - Yoshiyasu Nakamura
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Asahi-ku, Yokohama, Japan
| | - Kazuko Sakai
- Department of Genome Biology, School of Medicine, Kinki University, Osaka-Sayama, Osaka, Japan
| | - Akihiro Hattori
- Kanagawa Academy of Science and Technology, Takatsu, Kawasaki, Japan
| | - Masao Odaka
- Kanagawa Academy of Science and Technology, Takatsu, Kawasaki, Japan
| | - Mathias Girault
- Kanagawa Academy of Science and Technology, Takatsu, Kawasaki, Japan
| | - Tokuzo Arao
- Department of Genome Biology, School of Medicine, Kinki University, Osaka-Sayama, Osaka, Japan
| | - Kazuto Nishio
- Department of Genome Biology, School of Medicine, Kinki University, Osaka-Sayama, Osaka, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Asahi-ku, Yokohama, Japan
| | - Kenji Yasuda
- Kanagawa Academy of Science and Technology, Takatsu, Kawasaki, Japan
- Department of Biomedical Information, Division of Biosystems, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Chiyoda, Tokyo, Japan
- * E-mail:
| |
Collapse
|
218
|
Microfluidic device with integrated microfilter of conical-shaped holes for high efficiency and high purity capture of circulating tumor cells. Sci Rep 2014; 4:6052. [PMID: 25116599 PMCID: PMC7365311 DOI: 10.1038/srep06052] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 07/25/2014] [Indexed: 02/06/2023] Open
Abstract
Capture of circulating tumor cells (CTCs) from peripheral blood of cancer patients has major implications for metastatic detection and therapy analyses. Here we demonstrated a microfluidic device for high efficiency and high purity capture of CTCs. The key novelty of this approach lies on the integration of a microfilter with conical-shaped holes and a micro-injector with cross-flow components for size dependent capture of tumor cells without significant retention of non-tumor cells. Under conditions of constant flow rate, tumor cells spiked into phosphate buffered saline could be recovered and then cultured for further analyses. When tumor cells were spiked in blood of healthy donors, they could also be recovered at high efficiency and high clearance efficiency of white blood cells. When the same device was used for clinical validation, CTCs could be detected in blood samples of cancer patients but not in that of healthy donors. Finally, the capture efficiency of tumor cells is cell-type dependent but the hole size of the filter should be more closely correlated to the nuclei size of the tumor cells. Together with the advantage of easy operation, low-cost and high potential of integration, this approach offers unprecedented opportunities for metastatic detection and cancer treatment monitoring.
Collapse
|
219
|
Beattie W, Qin X, Wang L, Ma H. Clog-free cell filtration using resettable cell traps. LAB ON A CHIP 2014; 14:2657-2665. [PMID: 24710608 DOI: 10.1039/c4lc00306c] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The separation of cells by filtration through microstructured constrictions is limited by clogging and adsorption, which reduce selectivity and prevent the extraction of separated cells. To address this key challenge, we developed a mechanism for simply and reliably adjusting the cross-section of a microfluidic channel to selectively capture cells based on a combination of size and deformability. After a brief holding period, trapped cells can then be released back into flow, and if necessary, extracted for subsequent analysis. Periodically clearing filter constrictions of separated cells greatly improves selectivity and throughput, and minimizes adsorption of cells to the filter microstructure. This mechanism is capable of discriminating cell-sized polystyrene microspheres with <1 μm resolution. Rare cancer cells doped into leukocytes can be enriched ~1800× with ~90% yield despite a significant overlap in size between these cell types. An important characteristic of this process is that contaminant leukocytes are captured by non-specific adsorption and not mechanical constraint, enabling repeated filtration to improve performance. The throughput of this mechanism is 900,000 cells per hour for 32 multiplexed microchannels, or ~1,200,000 cells cm⁻² h⁻¹ on a per area basis, which exceeds existing micropore filtration mechanisms by a factor of 20.
Collapse
Affiliation(s)
- William Beattie
- Department of Mechanical Engineering, University of British Columbia, 2054-6250 Applied Science Lane, Vancouver, BC, Canada V6T 1Z4.
| | | | | | | |
Collapse
|
220
|
Galletti G, Portella L, Tagawa ST, Kirby BJ, Giannakakou P, Nanus DM. Circulating tumor cells in prostate cancer diagnosis and monitoring: an appraisal of clinical potential. Mol Diagn Ther 2014; 18:389-402. [PMID: 24809501 PMCID: PMC4149177 DOI: 10.1007/s40291-014-0101-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Circulating tumor cells (CTCs) have emerged as a viable solution to the lack of tumor tissue availability for patients with a variety of solid tumors, including prostate cancer. Different approaches have been used to capture this tumor cell population and several of these techniques have been used to assess the potential role of CTCs as a biological marker to predict treatment efficacy and clinical outcome. CTCs are now considered a strong tool to understand the molecular characteristics of prostate cancer, and to be used and analyzed as a 'liquid biopsy' in the attempt to grasp the biological portrait of the disease in the individual patient.
Collapse
Affiliation(s)
- Giuseppe Galletti
- Division of Hematology and Medical Oncology and the Weill Cornell Cancer Center, Weill Cornell Medical College, New York, USA
| | - Luigi Portella
- Division of Hematology and Medical Oncology and the Weill Cornell Cancer Center, Weill Cornell Medical College, New York, USA
| | - Scott T. Tagawa
- Division of Hematology and Medical Oncology and the Weill Cornell Cancer Center, Weill Cornell Medical College, New York, USA
| | - Brian J. Kirby
- Division of Hematology and Medical Oncology and the Weill Cornell Cancer Center, Weill Cornell Medical College, New York, USA
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - Paraskevi Giannakakou
- Division of Hematology and Medical Oncology and the Weill Cornell Cancer Center, Weill Cornell Medical College, New York, USA
| | - David M. Nanus
- Division of Hematology and Medical Oncology and the Weill Cornell Cancer Center, Weill Cornell Medical College, New York, USA
- Division of Hematology and Medical Oncology and the Weill Cornell Cancer Center, Weill Cornell Medical College, 1305 York Avenue, Room 741, New York, NY 10021, USA
| |
Collapse
|
221
|
Li X, Chen W, Liu G, Lu W, Fu J. Continuous-flow microfluidic blood cell sorting for unprocessed whole blood using surface-micromachined microfiltration membranes. LAB ON A CHIP 2014; 14:2565-75. [PMID: 24895109 PMCID: PMC4106416 DOI: 10.1039/c4lc00350k] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
White blood cells (WBCs) constitute about 0.1% of the blood cells, yet they play a critical role in innate and adaptive immune responses against pathogenic infections, allergic conditions, and malignancies and thus contain rich information about the immune status of the body. Rapid isolation of WBCs directly from whole blood is a prerequisite for any integrated immunoassay platform designed for examining WBC phenotypes and functions; however, such functionality is still challenging for blood-on-a-chip systems, as existing microfluidic cell sorting techniques are inadequate for efficiently processing unprocessed whole blood on chip with concurrent high throughput and cell purity. Herein we report a microfluidic chip for continuous-flow isolation and sorting of WBCs from whole blood with high throughput and separation efficiency. The microfluidic cell sorting chip leveraged the crossflow filtration scheme in conjunction with a surface-micromachined poly(dimethylsiloxane) (PDMS) microfiltration membrane (PMM) with high porosity. With a sample throughput of 1 mL h(-1), the microfluidic cell sorting chip could recover 27.4 ± 4.9% WBCs with a purity of 93.5 ± 0.5%. By virtue of its separation efficiency, ease of sample recovery, and high throughput enabled by its continuous-flow operation, the microfluidic cell sorting chip holds promise as an upstream component for blood sample preparation and analysis in integrated blood-on-a-chip systems.
Collapse
Affiliation(s)
- Xiang Li
- Integrated Biosystems and Biomechanics Laboratory, University of Michigan, Ann Arbor, Michigan 48109, USA.
| | | | | | | | | |
Collapse
|
222
|
Zhang Z, Xu J, Hong B, Chen X. The effects of 3D channel geometry on CTC passing pressure--towards deformability-based cancer cell separation. LAB ON A CHIP 2014; 14:2576-84. [PMID: 24895079 DOI: 10.1039/c4lc00301b] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Various lab on a chip devices have been developed recently to detect and separate circulating tumour cells (CTCs) for early stage cancer detection. Because CTCs are extremely rare in the blood, next generation CTC microfilters aim at significant improvement in both efficiency and throughput. CTC microfilters based on cell deformability seem to be a promising direction. In the present research, we study a CTC passing event through a micro-filtering channel with various 3D geometries. The pressure signatures for different types of cells passing through different channels are characterized numerically. Specifically, five kinds of cross-sections, circular, square, triangular and two kinds of rectangular channels with aspect ratios of 2 and 5, are studied in this work. The total pressures for cells passing through the channels are calculated and reveal different behaviour from what is predicted by the static surface tension model. Among all five cross-sections studied, the circular cross-section features the highest critical pressure and thus is most suitable for high efficiency CTC separation. The square filtering channel provides the second largest critical pressure, and the triangular cross-section provides the least critical pressure among these three cross-sections. All these three cross-sections are better than the rectangular channels with aspect ratios of 2 and 5. For the rectangular channel, a high aspect ratio channel may lead to cell splitting at high speed, which will result in a periodic pressure signature. Our findings will provide valuable information for the design of next generation CTC microfilters.
Collapse
Affiliation(s)
- Zhifeng Zhang
- Mechanical Engineering, Department of ENCS, Washington State University, Vancouver, 98686, WA, USA
| | | | | | | |
Collapse
|
223
|
Clinical validation of an ultra high-throughput spiral microfluidics for the detection and enrichment of viable circulating tumor cells. PLoS One 2014; 9:e99409. [PMID: 24999991 PMCID: PMC4085042 DOI: 10.1371/journal.pone.0099409] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 05/14/2014] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Circulating tumor cells (CTCs) are cancer cells that can be isolated via liquid biopsy from blood and can be phenotypically and genetically characterized to provide critical information for guiding cancer treatment. Current analysis of CTCs is hindered by the throughput, selectivity and specificity of devices or assays used in CTC detection and isolation. METHODOLOGY/PRINCIPAL FINDINGS Here, we enriched and characterized putative CTCs from blood samples of patients with both advanced stage metastatic breast and lung cancers using a novel multiplexed spiral microfluidic chip. This system detected putative CTCs under high sensitivity (100%, n = 56) (Breast cancer samples: 12-1275 CTCs/ml; Lung cancer samples: 10-1535 CTCs/ml) rapidly from clinically relevant blood volumes (7.5 ml under 5 min). Blood samples were completely separated into plasma, CTCs and PBMCs components and each fraction were characterized with immunophenotyping (Pan-cytokeratin/CD45, CD44/CD24, EpCAM), fluorescence in-situ hybridization (FISH) (EML4-ALK) or targeted somatic mutation analysis. We used an ultra-sensitive mass spectrometry based system to highlight the presence of an EGFR-activating mutation in both isolated CTCs and plasma cell-free DNA (cf-DNA), and demonstrate concordance with the original tumor-biopsy samples. CONCLUSIONS/SIGNIFICANCE We have clinically validated our multiplexed microfluidic chip for the ultra high-throughput, low-cost and label-free enrichment of CTCs. Retrieved cells were unlabeled and viable, enabling potential propagation and real-time downstream analysis using next generation sequencing (NGS) or proteomic analysis.
Collapse
|
224
|
|
225
|
Sun W, Huang T, Li G, Shen W, Zhu J, Jin Q, Zhao J, Jia C, Zhang Z. The advantage of circulating tumor cells over serum carcinoembryonic antigen for predicting treatment responses in rectal cancer. Future Oncol 2014; 9:1489-500. [PMID: 24106900 DOI: 10.2217/fon.13.91] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
AIM The objective of this study was to investigate the clinical significance of circulating tumor cells (CTCs) on the evaluation and prediction of treatment responses in rectal cancer patients compared with serum carcinoembryonic antigen (CEA). MATERIALS & METHODS Both CTCs and CEA levels of 103 rectal cancer patients (66 with stage II-III and 37 with recurrence or metastasis) were analyzed before and after chemoradiotherapy. CTCs were detected using EpCAM magnetic bead-based enrichment combined with cytometric identification. RESULTS CTCs were detected in all patients while no tumor cells were found in healthy controls. CTC levels in metastatic patients were significantly higher than those with recurrence or stage II-III rectal cancer. There is a close relationship between CTC levels and treatment outcomes but serum CEA did not have any correlation. CONCLUSION CTCs are promising markers for the evaluation and prediction of treatment responses in rectal cancer patients, superior to the conventional tumor marker CEA.
Collapse
Affiliation(s)
- Wenjie Sun
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai 200032, China
| | | | | | | | | | | | | | | | | |
Collapse
|
226
|
Ito H, Inoue H, Kimura S, Ohmori T, Ishikawa F, Gohda K, Sato J. Prognostic impact of the number of viable circulating cells with high telomerase activity in gastric cancer patients: a prospective study. Int J Oncol 2014; 45:227-34. [PMID: 24788213 DOI: 10.3892/ijo.2014.2409] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Accepted: 04/08/2014] [Indexed: 12/26/2022] Open
Abstract
The identification of circulating tumor cells (CTCs) in peripheral blood is a useful approach to estimate prognosis, monitor disease progression and measure treatment effects in several types of malignancies. We have previously used OBP-401, a telomerase-specific, replication-selective, oncolytic adenoviral agent carrying the green fluorescent protein (GFP) gene. GFP-positive cells (GFP+ cells) were counted under a fluorescence microscope. Our results showed that the number of at least 7.735 µm in diameter GFP+ cells (L-GFP+ cells) in the peripheral blood was a significant marker of prognosis in gastric cancer patients. However, tumor cells undergoing epithelial-mesenchymal transition (EMT) have been reported to be smaller in size than cells without EMT features; thus, CTCs undergoing EMT may escape detection with this technique. Therefore, in this study, we analyzed the relationship between patient outcome and the number of GFP+ cells of any size. We obtained peripheral blood samples from 65 patients with gastric cancer. After infection of OBP-401, GFP+ cells were counted and measured. The relationship between the number of GFP+ cells and surgical outcome was analyzed. The median follow-up period of the surviving patients was 36 months. A significant difference in overall survival was found between patients with 0-5 and patients with ≥6 L-GFP+ cells. No clear relationship was established between the number of small-sized GFP+ cells and patient prognosis. The number of L-GFP+ cells was significantly related to overall survival in patients with gastric cancer. The detection of L-GFP+ cells using OBP-401 may be a useful prognostic marker in gastric cancer.
Collapse
Affiliation(s)
- Hiroaki Ito
- Digestive Disease Center, Showa University Northern Yokohama Hospital, Tsuzuki-ku, Yokohama 224-8503, Japan
| | - Haruhiro Inoue
- Digestive Disease Center, Showa University Northern Yokohama Hospital, Tsuzuki-ku, Yokohama 224-8503, Japan
| | - Satoshi Kimura
- Department of Laboratory Medicine and Central Clinical Laboratory, Showa University Northern Yokohama Hospital, Tsuzuki-ku, Yokohama 224-8503, Japan
| | - Tohru Ohmori
- Institute of Molecular Oncology, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Fumihiro Ishikawa
- Department of Cancer Cell Biology, Showa University School of Pharmacy, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Keigo Gohda
- Central Research Laboratories, Sysmex Corporation, Nishi-ku, Kobe 651-2271, Japan
| | - Jun Sato
- Central Research Laboratories, Sysmex Corporation, Nishi-ku, Kobe 651-2271, Japan
| |
Collapse
|
227
|
Polymeric microfluidic devices exhibiting sufficient capture of cancer cell line for isolation of circulating tumor cells. Biomed Microdevices 2014; 15:611-616. [PMID: 23666489 DOI: 10.1007/s10544-013-9775-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Here, we developed polymeric microfluidic devices for the isolation of circulating tumor cells. The devices, with more than 30,000 microposts in the channel, were produced successfully by a UV light-curing process lasting 3 min. The device surface was coated with anti-epithelial cell adhesion molecule antibody by just contacting the antibody solution, and a flow system including the device was established to send a cell suspension through it. We carried out flow tests for evaluation of the device's ability to capture tumor cells using an esophageal cancer cell line, KYSE220, dispersed in phosphate-buffered saline or mononuclear cell separation from whole blood. After the suspension flowed through the chip, many cells were seen to be captured on the microposts coated with the antibody, whereas there were few cells in the device without the antibody. Owing to the transparency of the device, we could observe the intact and the stained cells captured on the microposts by transmitted light microscopy and phase contrast microscopy, in addition to fluorescent microscopy, which required fluorescence labeling. Cell capture efficiencies (i.e., recovery rates of the flowing cancer cells by capture with the microfluidic device) were measured. The resulting values were 0.88 and 0.95 for cell suspension in phosphate-buffered saline, and 0.85 for the suspension in the mononuclear cell separation, suggesting the sufficiency of this device for the isolation of circulating tumor cells. Therefore, our device may be useful for research and treatments that rely on investigation of circulating tumor cells in the blood of cancer patients.
Collapse
|
228
|
Abstract
More than two decades ago, microfluidics began to show its impact in biological research. Since then, the field of microfluidics has evolving rapidly. Cancer is one of the leading causes of death worldwide. Microfluidics holds great promise in cancer diagnosis and also serves as an emerging tool for understanding cancer biology. Microfluidics can be valuable for cancer investigation due to its high sensitivity, high throughput, less material-consumption, low cost, and enhanced spatio-temporal control. The physical laws on microscale offer an advantage enabling the control of physics, biology, chemistry and physiology at cellular level. Furthermore, microfluidic based platforms are portable and can be easily designed for point-of-care diagnostics. Developing and applying the state of the art microfluidic technologies to address the unmet challenges in cancer can expand the horizons of not only fundamental biology but also the management of disease and patient care. Despite the various microfluidic technologies available in the field, few have been tested clinically, which can be attributed to the various challenges existing in bridging the gap between the emerging technology and real world applications. We present a review of role of microfluidics in cancer research, including the history, recent advances and future directions to explore where the field stand currently in addressing complex clinical challenges and future of it. This review identifies four critical areas in cancer research, in which microfluidics can change the current paradigm. These include cancer cell isolation, molecular diagnostics, tumor biology and high-throughput screening for therapeutics. In addition, some of our lab's current research is presented in the corresponding sections.
Collapse
Affiliation(s)
- Zhuo Zhang
- Department of Chemical Engineering, University of Michigan, 2300 Hayward Street, Ann Arbor, MI, 48109, USA
| | - Sunitha Nagrath
- Department of Chemical Engineering, University of Michigan, 2300 Hayward Street, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
229
|
Gach PC, Attayek PJ, Whittlesey RL, Yeh JJ, Allbritton NL. Micropallet arrays for the capture, isolation and culture of circulating tumor cells from whole blood of mice engrafted with primary human pancreatic adenocarcinoma. Biosens Bioelectron 2014; 54:476-83. [PMID: 24316450 PMCID: PMC3947965 DOI: 10.1016/j.bios.2013.11.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Revised: 10/30/2013] [Accepted: 11/06/2013] [Indexed: 11/20/2022]
Abstract
Circulating tumor cells (CTCs) are important biomarkers of cancer progression and metastatic potential. The rarity of CTCs in peripheral blood has driven the development of technologies to isolate these tumor cells with high specificity; however, there are limited techniques available for isolating target CTCs following enumeration. A strategy is described to capture and isolate viable tumor cells from whole blood using an array of releasable microstructures termed micropallets. Specific capture of nucleated cells or cells expressing epithelial cell adhesion molecules (EpCAM) was achieved by functionalizing micropallet surfaces with either fibronectin, Matrigel or anti-EpCAM antibody. Surface grafting of poly(acrylic acid) followed by covalent binding of protein A/G enabled efficient capture of EpCAM antibody on the micropallet surface. MCF-7 cells, a human breast adenocarcinoma, were retained on the array surface with 90±8% efficiency when using an anti-EpCAM-coated array. To demonstrate the efficiency of tumor cell retention on micropallet arrays in the presence of blood, MCF-7 cells were mixed into whole blood and added to small arrays (71 mm(2)) coated with fibronectin, Matrigel or anti-EpCAM. These approaches achieved MCF-7 cell capture from ≤10 µL of whole blood with efficiencies greater than 85%. Furthermore, MCF-7 cells intermixed with 1 mL blood and loaded onto large arrays (7171 mm(2)) were captured with high efficiencies (≥97%), could be isolated from the array by a laser-based approach and were demonstrated to yield a high rate of colony formation (≥85%) after removal from the array. Clinical utility of this technology was shown through the capture, isolation and successful culture of CTCs from the blood of mice engrafted with primary human pancreatic tumors. Direct capture and isolation of living tumor cells from blood followed by analysis or culture will be a valuable tool for cancer cell characterization.
Collapse
Affiliation(s)
- Philip C Gach
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599, United States
| | - Peter J Attayek
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC 27599, United States; Joint Department of Biomedical Engineering, North Carolina State University, Raleigh, NC 27695, United States
| | - Rebecca L Whittlesey
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, United States
| | - Jen Jen Yeh
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, United States; Department of Surgery, Division of surgical Oncology, University of North Carolina, Chapel Hill, NC 27599, United States; Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27599, United States
| | - Nancy L Allbritton
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599, United States; Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC 27599, United States; Joint Department of Biomedical Engineering, North Carolina State University, Raleigh, NC 27695, United States; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, United States; Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27599, United States.
| |
Collapse
|
230
|
Yoon HJ, Kozminsky M, Nagrath S. Emerging role of nanomaterials in circulating tumor cell isolation and analysis. ACS NANO 2014; 8:1995-2017. [PMID: 24601556 PMCID: PMC4004319 DOI: 10.1021/nn5004277] [Citation(s) in RCA: 191] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Circulating tumor cells (CTCs) are low frequency cells found in the bloodstream after having been shed from a primary tumor. These cells are research targets because of the information they may potentially provide about both an individual cancer as well as the mechanisms through which cancer spreads in the process of metastasis. Established technologies exist for CTC isolation, but the recent progress and future of this field lie in nanomaterials. In this review, we provide perspective into historical CTC capture as well as current research being conducted, emphasizing the significance of the materials being used to fabricate these devices. The modern investigation into CTCs initially featured techniques that have since been commercialized. A major innovation in the field was the development of a microfluidic capture device, first fabricated in silicon and followed up with glass and thermopolymer devices. We then specifically highlight the technologies incorporating magnetic nanoparticles, carbon nanotubes, nanowires, nanopillars, nanofibers, and nanoroughened surfaces, graphene oxide and their fabrication methods. The nanoscale provides a new set of tools that has the potential to overcome current limitations associated with CTC capture and analysis. We believe the current trajectory of the field is in the direction of nanomaterials, allowing the improvements necessary to further CTC research.
Collapse
|
231
|
Costa C, Abal M, López-López R, Muinelo-Romay L. Biosensors for the detection of circulating tumour cells. SENSORS 2014; 14:4856-75. [PMID: 24618729 PMCID: PMC4003971 DOI: 10.3390/s140304856] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 01/28/2014] [Accepted: 02/28/2014] [Indexed: 12/14/2022]
Abstract
Metastasis is the cause of most cancer deaths. Circulating tumour cells (CTCs) are cells released from the primary tumour into the bloodstream that are considered the main promoters of metastasis. Therefore, these cells are targets for understanding tumour biology and improving clinical management of the disease. Several techniques have emerged in recent years to isolate, detect, and characterise CTCs. As CTCs are a rare event, their study requires multidisciplinary considerations of both biological and physical properties. In addition, as isolation of viable cells may give further insights into metastatic development, cell recovery must be done with minimal cell damage. The ideal system for CTCs analysis must include maximum efficiency of detection in real time. In this sense, new approaches used to enrich CTCs from clinical samples have provided an important improvement in cell recovery. However, this progress should be accompanied by more efficient strategies of cell quantification. A range of biosensor platforms are being introduced into the technology for CTCs quantification with promising results. This review provides an update on recent progress in CTCs identification using different approaches based on sensor signaling.
Collapse
Affiliation(s)
- Clotilde Costa
- Translational Medical Oncology, Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), Trav. Choupana s/n 15706 Santiago de Compostela, Spain.
| | - Miguel Abal
- Translational Medical Oncology, Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), Trav. Choupana s/n 15706 Santiago de Compostela, Spain.
| | - Rafael López-López
- Translational Medical Oncology, Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), Trav. Choupana s/n 15706 Santiago de Compostela, Spain.
| | - Laura Muinelo-Romay
- Unity of CTCs analysis Translational Medical Oncology, Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), Trav. Choupana s/n 15706 Santiago de Compostela, Spain.
| |
Collapse
|
232
|
Friedlander TW, Fong L. The end of the beginning: circulating tumor cells as a biomarker in castration-resistant prostate cancer. J Clin Oncol 2014; 32:1104-6. [PMID: 24616311 DOI: 10.1200/jco.2013.54.7307] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Terence W Friedlander
- Helen Diller Family Comprehensive Cancer Center at the University of California, San Francisco, San Francisco, CA
| | | |
Collapse
|
233
|
Chen Y, Li S, Gu Y, Li P, Ding X, Wang L, McCoy JP, Levine SJ, Huang TJ. Continuous enrichment of low-abundance cell samples using standing surface acoustic waves (SSAW). LAB ON A CHIP 2014; 14:924-30. [PMID: 24413889 PMCID: PMC4688895 DOI: 10.1039/c3lc51001h] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Cell enrichment is a powerful tool in a variety of cellular studies, especially in applications with low-abundance cell types. In this work, we developed a standing surface acoustic wave (SSAW) based microfluidic device for non-contact, continuous cell enrichment. With a pair of parallel interdigital transducers (IDT) deposited on a piezoelectric substrate, a one-dimensional SSAW field was established along disposable micro-tubing channels, generating numerous pressure nodes (and thus numerous cell-enrichment regions). Our method is able to concentrate highly diluted blood cells by more than 100 fold with a recovery efficiency of up to 99%. Such highly effective cell enrichment was achieved without using sheath flow. The SSAW-based technique presented here is simple, bio-compatible, label-free, and sheath-flow-free. With these advantages, it could be valuable for many biomedical applications.
Collapse
Affiliation(s)
- Yuchao Chen
- Department of Engineering Science and Mechanics, The Pennsylvania, State University, University Park, PA 16802, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
234
|
Chen Y, Li P, Huang PH, Xie Y, Mai JD, Wang L, Nguyen NT, Huang TJ. Rare cell isolation and analysis in microfluidics. LAB ON A CHIP 2014; 14:626-45. [PMID: 24406985 PMCID: PMC3991782 DOI: 10.1039/c3lc90136j] [Citation(s) in RCA: 206] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Rare cells are low-abundance cells in a much larger population of background cells. Conventional benchtop techniques have limited capabilities to isolate and analyze rare cells because of their generally low selectivity and significant sample loss. Recent rapid advances in microfluidics have been providing robust solutions to the challenges in the isolation and analysis of rare cells. In addition to the apparent performance enhancements resulting in higher efficiencies and sensitivity levels, microfluidics provides other advanced features such as simpler handling of small sample volumes and multiplexing capabilities for high-throughput processing. All of these advantages make microfluidics an excellent platform to deal with the transport, isolation, and analysis of rare cells. Various cellular biomarkers, including physical properties, dielectric properties, as well as immunoaffinities, have been explored for isolating rare cells. In this Focus article, we discuss the design considerations of representative microfluidic devices for rare cell isolation and analysis. Examples from recently published works are discussed to highlight the advantages and limitations of the different techniques. Various applications of these techniques are then introduced. Finally, a perspective on the development trends and promising research directions in this field are proposed.
Collapse
Affiliation(s)
- Yuchao Chen
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA
| | - Peng Li
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA
| | - Po-Hsun Huang
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA
| | - Yuliang Xie
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - John D. Mai
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, PR China
| | - Lin Wang
- Ascent Bio-Nano Technologies Inc., State College, PA 16801, USA
| | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology Centre, Griffith University, 170 Kessels Road, Brisbane 4111, Australia
| | - Tony Jun Huang
- Fax: 814-865-9974; Tel: 814-863-4209; Fax: 61-(0)7-3735-8021; Tel: 61-(0)7-3735-3921;
| |
Collapse
|
235
|
Yusa A, Toneri M, Masuda T, Ito S, Yamamoto S, Okochi M, Kondo N, Iwata H, Yatabe Y, Ichinosawa Y, Kinuta S, Kondo E, Honda H, Arai F, Nakanishi H. Development of a new rapid isolation device for circulating tumor cells (CTCs) using 3D palladium filter and its application for genetic analysis. PLoS One 2014; 9:e88821. [PMID: 24523941 PMCID: PMC3921253 DOI: 10.1371/journal.pone.0088821] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 01/12/2014] [Indexed: 11/18/2022] Open
Abstract
Circulating tumor cells (CTCs) in the blood of patients with epithelial malignancies provide a promising and minimally invasive source for early detection of metastasis, monitoring of therapeutic effects and basic research addressing the mechanism of metastasis. In this study, we developed a new filtration-based, sensitive CTC isolation device. This device consists of a 3-dimensional (3D) palladium (Pd) filter with an 8 µm-sized pore in the lower layer and a 30 µm-sized pocket in the upper layer to trap CTCs on a filter micro-fabricated by precise lithography plus electroforming process. This is a simple pump-less device driven by gravity flow and can enrich CTCs from whole blood within 20 min. After on-device staining of CTCs for 30 min, the filter cassette was removed from the device, fixed in a cassette holder and set up on the upright fluorescence microscope. Enumeration and isolation of CTCs for subsequent genetic analysis from the beginning were completed within 1.5 hr and 2 hr, respectively. Cell spike experiments demonstrated that the recovery rate of tumor cells from blood by this Pd filter device was more than 85%. Single living tumor cells were efficiently isolated from these spiked tumor cells by a micromanipulator, and KRAS mutation, HER2 gene amplification and overexpression, for example, were successfully detected from such isolated single tumor cells. Sequential analysis of blood from mice bearing metastasis revealed that CTC increased with progression of metastasis. Furthermore, a significant increase in the number of CTCs from the blood of patients with metastatic breast cancer was observed compared with patients without metastasis and healthy volunteers. These results suggest that this new 3D Pd filter-based device would be a useful tool for the rapid, cost effective and sensitive detection, enumeration, isolation and genetic analysis of CTCs from peripheral blood in both preclinical and clinical settings.
Collapse
Affiliation(s)
- Akiko Yusa
- Aichi Science and Technology Foundation, Knowledge Hub Aichi, Priority Research Projects, Japan
- Department of Micro-Nano Systems Engineering, Graduate School of Engineering, Nagoya University, Japan
- Division of Oncological Pathology, Aichi Cancer Center Research Institute, Japan
| | - Makoto Toneri
- Department of Gastrointestinal Surgery, Aichi Cancer Center Central Hospital, Japan
- Division of Oncological Pathology, Aichi Cancer Center Research Institute, Japan
| | - Taisuke Masuda
- Department of Micro-Nano Systems Engineering, Graduate School of Engineering, Nagoya University, Japan
| | - Seiji Ito
- Department of Gastrointestinal Surgery, Aichi Cancer Center Central Hospital, Japan
| | - Shuhei Yamamoto
- Department of Biotechnogloy, Graduate School of Engineering, Nagoya University, Japan
| | - Mina Okochi
- Department of Biotechnogloy, Graduate School of Engineering, Nagoya University, Japan
| | - Naoto Kondo
- Department of Breast Oncology, Aichi Cancer Center Central Hospital, Japan
| | - Hiroji Iwata
- Department of Breast Oncology, Aichi Cancer Center Central Hospital, Japan
| | - Yasushi Yatabe
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center Central Hospital, Japan
| | | | | | - Eisaku Kondo
- Division of Oncological Pathology, Aichi Cancer Center Research Institute, Japan
| | - Hiroyuki Honda
- Department of Biotechnogloy, Graduate School of Engineering, Nagoya University, Japan
| | - Fumihito Arai
- Department of Micro-Nano Systems Engineering, Graduate School of Engineering, Nagoya University, Japan
| | - Hayao Nakanishi
- Division of Oncological Pathology, Aichi Cancer Center Research Institute, Japan
- * E-mail:
| |
Collapse
|
236
|
Song Y, Huang YY, Liu X, Zhang X, Ferrari M, Qin L. Point-of-care technologies for molecular diagnostics using a drop of blood. Trends Biotechnol 2014; 32:132-9. [PMID: 24525172 DOI: 10.1016/j.tibtech.2014.01.003] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 01/07/2014] [Accepted: 01/08/2014] [Indexed: 11/19/2022]
Abstract
Molecular diagnostics is crucial for prevention, identification, and treatment of disease. Traditional technologies for molecular diagnostics using blood are limited to laboratory use because they rely on sample purification and sophisticated instruments, are labor and time intensive, expensive, and require highly trained operators. This review discusses the frontiers of point-of-care (POC) diagnostic technologies using a drop of blood obtained from a finger prick. These technologies, including emerging biotechnologies, nanotechnologies, and microfluidics, hold the potential for rapid, accurate, and inexpensive disease diagnostics.
Collapse
Affiliation(s)
- Yujun Song
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Yu-Yen Huang
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78758, USA
| | - Xuewu Liu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Xiaojing Zhang
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78758, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Lidong Qin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY 10065, USA.
| |
Collapse
|
237
|
Park S, Ang RR, Duffy SP, Bazov J, Chi KN, Black PC, Ma H. Morphological differences between circulating tumor cells from prostate cancer patients and cultured prostate cancer cells. PLoS One 2014; 9:e85264. [PMID: 24416373 PMCID: PMC3885705 DOI: 10.1371/journal.pone.0085264] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Accepted: 11/25/2013] [Indexed: 12/01/2022] Open
Abstract
Circulating tumor cell (CTC) enumeration promises to be an important predictor of clinical outcome for a range of cancers. Established CTC enumeration methods primarily rely on affinity capture of cell surface antigens, and have been criticized for underestimation of CTC numbers due to antigenic bias. Emerging CTC capture strategies typically distinguish these cells based on their assumed biomechanical characteristics, which are often validated using cultured cancer cells. In this study, we developed a software tool to investigate the morphological properties of CTCs from patients with castrate resistant prostate cancer and cultured prostate cancer cells in order to establish whether the latter is an appropriate model for the former. We isolated both CTCs and cultured cancer cells from whole blood using the CellSearch® system and examined various cytomorphological characteristics. In contrast with cultured cancer cells, CTCs enriched by CellSearch® system were found to have significantly smaller size, larger nuclear-cytoplasmic ratio, and more elongated shape. These CTCs were also found to exhibit significantly more variability than cultured cancer cells in nuclear-cytoplasmic ratio and shape profile.
Collapse
Affiliation(s)
- Sunyoung Park
- Department of Mechanical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Richard R. Ang
- Department of Mechanical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Simon P. Duffy
- Department of Mechanical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Biology, Kwantlen Polytechnic University, Surrey, British Columbia, Canada
| | - Jenny Bazov
- Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Kim N. Chi
- Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, British Columbia, Canada
- BC Cancer Agency, Vancouver Cancer Centre, Vancouver, British Columbia, Canada
- Department of Urologic Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Peter C. Black
- BC Cancer Agency, Vancouver Cancer Centre, Vancouver, British Columbia, Canada
- Department of Urologic Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hongshen Ma
- Department of Mechanical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
- Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, British Columbia, Canada
- Department of Urologic Science, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
238
|
Alix-Panabières C, Pantel K. Technologies for detection of circulating tumor cells: facts and vision. LAB ON A CHIP 2014; 14:57-62. [PMID: 24145967 DOI: 10.1039/c3lc50644d] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Hematogeneous tumor cell dissemination is a key step in cancer progression. The detection of CTCs in the peripheral blood of patients with solid epithelial tumors (e.g., breast, prostate, lung and colon cancer) holds great promise, and many exciting technologies have been developed over the past years. However, the detection and molecular characterization of circulating tumor cells (CTCs) remain technically challenging. The identification and characterization of CTCs require extremely sensitive and specific analytical methods, which are usually a combination of complex enrichment and detection procedures. CTCs occur at very low concentrations of one tumor cell in the background of millions of normal blood cells and the epithelial-mesenchymal plasticity of CTCs can hamper their detection by the epithelial markers used in current CTC assays. In the present review, we summarize current methods for the enrichment and detection of CTCs and discuss the key challenges and perspectives of CTC analyses within the context of improved clinical management of cancer patients.
Collapse
Affiliation(s)
- Catherine Alix-Panabières
- University Medical Centre, Saint-Eloi Hospital, Department of Cellular and Tissue Biopathology of Tumors, Laboratory of Rare Human Circulating Cells (LCCRH), Montpellier, France
| | | |
Collapse
|
239
|
Hyun KA, Jung HI. Advances and critical concerns with the microfluidic enrichments of circulating tumor cells. LAB ON A CHIP 2014; 14:45-56. [PMID: 23982141 DOI: 10.1039/c3lc50582k] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Over the past two decades, circulating tumor cells (CTCs) have been widely recognized for their importance in clinical trials. While most enrichment methods for these cells have been conducted through the batch process due to their rarity in blood and the need for large sample volumes, the batch process leads to unavoidable cell loss. Given the heterogenetic features of CTCs, this cell loss may limit the validity of research that relies on the isolation of CTCs; such research includes cancer prognosis, diagnosis of minimal residual diseases, assessment of tumor sensitivity to anticancer drugs, and the personalization of anticancer therapies. Recent advances in microfluidic approaches have made it possible to enrich CTCs with a small degree of cell loss. In this review, we highlight several microfluidic-based positive and negative enrichment methods that are the subject of considerable research interest (e.g. EpCAM-dependent assay and EpCAM-independent assay) and suggest a microfluidic-based single cell analysis platform for the down-stream analysis of CTCs. We also discuss critical concerns and future directions for research.
Collapse
Affiliation(s)
- Kyung-A Hyun
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-no Seodaemun-gu, Seoul 120-752, South Korea.
| | | |
Collapse
|
240
|
Jin C, McFaul SM, Duffy SP, Deng X, Tavassoli P, Black PC, Ma H. Technologies for label-free separation of circulating tumor cells: from historical foundations to recent developments. LAB ON A CHIP 2014; 14:32-44. [PMID: 23963515 DOI: 10.1039/c3lc50625h] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Circulating tumor cells (CTCs) are malignant cells shed into the bloodstream from a tumor that have the potential to establish metastases in different anatomical sites. The separation and subsequent characterization of these cells is emerging as an important tool for both biomarker discovery and the elucidation of mechanisms of metastasis. Established methods for separating CTCs rely on biochemical markers of epithelial cells that are known to be unreliable because of epithelial-to-mesenchymal transition, which reduces expression for epithelial markers. Emerging label-free separation methods based on the biophysical and biomechanical properties of CTCs have the potential to address this key shortcoming and present greater flexibility in the subsequent characterization of these cells. In this review we first present what is known about the biophysical and biomechanical properties of CTCs from historical studies and recent research. We then review biophysical label-free technologies that have been developed for CTC separation, including techniques based on filtration, hydrodynamic chromatography, and dielectrophoresis. Finally, we evaluate these separation methods and discuss requirements for subsequent characterization of CTCs.
Collapse
Affiliation(s)
- Chao Jin
- Department of Mechanical Engineering, University of British Columbia, 2054-6250 Applied Science Lane, Vancouver, BC, Canada V6T 1Z4.
| | | | | | | | | | | | | |
Collapse
|
241
|
|
242
|
Samlowski WE, McGregor JR, Samlowski ST, Tharkar S, Shen S, Bentz JS. Growth of Circulating Tumor Cell-Derived Colonies from Peripheral Blood of Melanoma Patients: Preliminary Characterization of Colony Composition. Health (London) 2014. [DOI: 10.4236/health.2014.612181] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
243
|
Friedlander TW, Premasekharan G, Paris PL. Looking back, to the future of circulating tumor cells. Pharmacol Ther 2013; 142:271-80. [PMID: 24362084 DOI: 10.1016/j.pharmthera.2013.12.011] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 11/27/2013] [Indexed: 12/12/2022]
Abstract
Detection and analysis of circulating tumor cells (CTCs) from patients with metastatic malignancies have become active areas of research in recent years. CTC enumeration has already proven useful in establishing prognosis for patients with metastatic breast, colon, and prostate cancer. More recently, studies are going beyond enumeration, exploring the CTCs as a means to better understand the mechanisms of tumorigenesis, invasion, and metastasis and the value of CTC characterization for prognosis and tailoring of treatment. Analysis of CTC subpopulations, for example, is highlighting the importance of the epithelial to mesenchymal transition (EMT), a process which may be crucial for allowing tumors to invade into and grow at sites distant from the original tumor site. Similarly, the detection of CTCs expressing markers of stemness may also have important implications for treatment resistance. Genomic analysis of CTC and CTC subpopulations may allow for selection of novel therapeutic targets to combat treatment resistance. CTCs become a particularly valuable biospecimen resource when tissue biopsies are unavailable or not feasible and liquid biopsies allow for serial monitoring. Lastly, cultures of patient-derived CTCs may allow for an evaluation of therapeutic strategies performed ex vivo and in real time. This review article will focus on these developments, starting with the CTC pathogenesis, going on to discuss the different platforms available for CTC isolation and their use to date in these arenas, then will explore multiple topics including the existing data concerning CTC subpopulations and their clinical relevance, genomic characterization, and lastly, avenues for future research.
Collapse
Affiliation(s)
- Terence W Friedlander
- Division of Hematology & Medical Oncology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, United States.
| | - Gayatri Premasekharan
- Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, United States
| | - Pamela L Paris
- Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, United States
| |
Collapse
|
244
|
Abstract
The metastatic dissemination and spread of malignant circulating tumor cells (CTCs) accounts for more than 90% of cancer-related deaths. CTCs detach from a primary tumor, travel through the circulatory system, and then invade and proliferate in distant organs. The detection of CTCs from blood has been established for prognostic monitoring and is predictive of patient outcome. Analysis of CTCs could enable the means for early detection and screening in cancer, as well as provide diagnostic access to tumor tissues in a minimally invasive way. The fundamental challenge with analyzing CTCs is the fact that they occur at extremely low concentrations in blood, on the order of one out of a billion cells. Various technologies have been proposed to isolate CTCs for enrichment. Here we focus on antigen-independent approaches that are not limited by specific capture antibodies. Intrinsic physical properties of CTCs, including cell size, deformability, and electrical properties, are reviewed, and technologies developed to exploit them for enrichment from blood are summarized. Physical enrichment technologies are of particular interest as they have the potential to increase yield and enable the analysis of rare CTC phenotypes that may not be otherwise obtained.
Collapse
Affiliation(s)
- Ramdane A. Harouaka
- Micro & Nano Integrated Biosystem (MINIBio) Laboratory, Department of Bioengineering and Materials Research Institute, Pennsylvania State University, University Park, PA 16802, U.S.A
- Penn State Hershey Cancer Institute, Hershey, PA 17033, U.S.A
| | - Merisa Nisic
- Micro & Nano Integrated Biosystem (MINIBio) Laboratory, Department of Bioengineering and Materials Research Institute, Pennsylvania State University, University Park, PA 16802, U.S.A
- Penn State Hershey Cancer Institute, Hershey, PA 17033, U.S.A
| | - Si-Yang Zheng
- Micro & Nano Integrated Biosystem (MINIBio) Laboratory, Department of Bioengineering and Materials Research Institute, Pennsylvania State University, University Park, PA 16802, U.S.A
- Penn State Hershey Cancer Institute, Hershey, PA 17033, U.S.A
| |
Collapse
|
245
|
Harouaka RA, Zhou MD, Yeh YT, Khan WJ, Das A, Liu X, Christ CC, Dicker DT, Baney TS, Kaifi JT, Belani CP, Truica CI, El-Deiry WS, Allerton JP, Zheng SY. Flexible micro spring array device for high-throughput enrichment of viable circulating tumor cells. Clin Chem 2013; 60:323-33. [PMID: 24132944 DOI: 10.1373/clinchem.2013.206805] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND The dissemination of circulating tumor cells (CTCs) that cause metastases in distant organs accounts for the majority of cancer-related deaths. CTCs have been established as a cancer biomarker of known prognostic value. The enrichment of viable CTCs for ex vivo analysis could further improve cancer diagnosis and guide treatment selection. We designed a new flexible micro spring array (FMSA) device for the enrichment of viable CTCs independent of antigen expression. METHODS Unlike previous microfiltration devices, flexible structures at the micro scale minimize cell damage to preserve viability, while maximizing throughput to allow rapid enrichment directly from whole blood with no need for sample preprocessing. Device performance with respect to capture efficiency, enrichment against leukocytes, viability, and proliferability was characterized. CTCs and CTC microclusters were enriched from clinical samples obtained from breast, lung, and colorectal cancer patients. RESULTS The FMSA device enriched tumor cells with 90% capture efficiency, higher than 10(4) enrichment, and better than 80% viability from 7.5-mL whole blood samples in <10 min on a 0.5-cm(2) device. The FMSA detected at least 1 CTC in 16 out of 21 clinical samples (approximately 76%) compared to 4 out of 18 (approximately 22%) detected with the commercial CellSearch® system. There was no incidence of clogging in over 100 tested fresh whole blood samples. CONCLUSIONS The FMSA device provides a versatile platform capable of viable enrichment and analysis of CTCs from clinically relevant volumes of whole blood.
Collapse
Affiliation(s)
- Ramdane A Harouaka
- Micro & Nano Integrated Biosystem (MINIBio) Laboratory, Department of Biomedical Engineering and Materials Research Institute, Pennsylvania State University, University Park, PA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
246
|
Harouaka R, Kang Z, Zheng SY, Cao L. Circulating tumor cells: advances in isolation and analysis, and challenges for clinical applications. Pharmacol Ther 2013; 141:209-21. [PMID: 24134902 DOI: 10.1016/j.pharmthera.2013.10.004] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 10/08/2013] [Indexed: 12/28/2022]
Abstract
Circulating tumor cells (CTCs) are rare cancer cells released from tumors into the bloodstream that are thought to have a key role in cancer metastasis. The presence of CTCs has been associated with worse prognosis in several major cancer types, including breast, prostate and colorectal cancer. There is considerable interest in CTC research and technologies for their potential use as cancer biomarkers that may enhance cancer diagnosis and prognosis, facilitate drug development, and improve the treatment of cancer patients. This review provides an update on recent progress in CTC isolation and molecular characterization technologies. Furthermore, the review covers significant advances and limitations in the clinical applications of CTC-based assays for cancer prognosis, response to anti-cancer therapies, and exploratory studies in biomarkers predictive of sensitivity and resistance to cancer therapies.
Collapse
Affiliation(s)
- Ramdane Harouaka
- Department of Bioengineering, Penn State University, University Park, PA, USA
| | - Zhigang Kang
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Si-Yang Zheng
- Department of Bioengineering, Penn State University, University Park, PA, USA
| | - Liang Cao
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
247
|
Svobodova Z, Kucerova J, Autebert J, Horak D, Bruckova L, Viovy JL, Bilkova Z. Application of an improved magnetic immunosorbent in an Ephesia chip designed for circulating tumor cell capture. Electrophoresis 2013; 35:323-9. [DOI: 10.1002/elps.201300196] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 05/31/2013] [Accepted: 06/01/2013] [Indexed: 01/09/2023]
Affiliation(s)
- Zuzana Svobodova
- Department of Biological and Biochemical Sciences; Faculty of Chemical Technology, University of Pardubice; Pardubice Czech Republic
| | - Jana Kucerova
- Department of Biological and Biochemical Sciences; Faculty of Chemical Technology, University of Pardubice; Pardubice Czech Republic
| | - Julien Autebert
- Macromolecules and Microsystems in Biology and Medicine; Institute Curie; Paris France
| | - Daniel Horak
- Institute of Macromolecular Chemistry; Academy of Sciences of the Czech Republic; Prague Czech Republic
| | - Lenka Bruckova
- Department of Biological and Biochemical Sciences; Faculty of Chemical Technology, University of Pardubice; Pardubice Czech Republic
| | - Jean-Louis Viovy
- Macromolecules and Microsystems in Biology and Medicine; Institute Curie; Paris France
| | - Zuzana Bilkova
- Department of Biological and Biochemical Sciences; Faculty of Chemical Technology, University of Pardubice; Pardubice Czech Republic
| |
Collapse
|
248
|
El-Heliebi A, Kroneis T, Zöhrer E, Haybaeck J, Fischereder K, Kampel-Kettner K, Zigeuner R, Pock H, Riedl R, Stauber R, Geigl JB, Huppertz B, Sedlmayr P, Lackner C. Are morphological criteria sufficient for the identification of circulating tumor cells in renal cancer? J Transl Med 2013; 11:214. [PMID: 24044779 PMCID: PMC3848446 DOI: 10.1186/1479-5876-11-214] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 09/13/2013] [Indexed: 12/22/2022] Open
Abstract
Background Single circulating tumor cells (CTCs) or circulating tumor microemboli (CTMs) are potential biomarkers of renal cell cancer (RCC), however studies of CTCs/CTMs in RCC are limited. In this pilot study we aimed to evaluate a novel blood filtration technique suited for cytomorphological classification, immunocytochemical and molecular characterization of filtered, so called circulating non-hematologic cells (CNHCs) - putative CTCs/CTMs - in patients with RCC. Methods Blood of 40 patients with renal tumors was subjected to ScreenCell® filtration. CNHCs were classified according to cytomorphological criteria. Immunocytochemical analysis was performed with antibodies against CD45, CD31 and carbonic anhydrase IX (CAIX, a RCC marker). DNA of selected CNHCs and respective primary tumors was analysed by array-CGH. Results CNHC-clusters with malignant or uncertain malignant cytomorphological features - putative CTMs - were negative for CD45, positive for CD31, while only 6% were CAIX positive. Array-CGH revealed that 83% of malignant and uncertain malignant cells did represent with a balanced genome whereas 17% presented genomic DNA imbalances which did not match the aberrations of the primary tumors. Putative single CTCs were negative for CD45, 33% were positive for CD31 and 56% were positive for CAIX. Conclusions The majority of CNHC-clusters, putative CTMs, retrieved by ScreenCell® filtration may be of endothelial origin. Morphological criteria seem to be insufficient to distinguish malignant from non-malignant cells in renal cancer.
Collapse
Affiliation(s)
- Amin El-Heliebi
- Institute of Pathology, Medical University of Graz, Auenbruggerplatz 25, Graz, 8036, Austria.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
249
|
Liu Z, Zhang W, Huang F, Feng H, Shu W, Xu X, Chen Y. High throughput capture of circulating tumor cells using an integrated microfluidic system. Biosens Bioelectron 2013; 47:113-9. [DOI: 10.1016/j.bios.2013.03.017] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 03/04/2013] [Accepted: 03/08/2013] [Indexed: 12/12/2022]
|
250
|
Conteduca V, Zamarchi R, Rossi E, Condelli V, Troiani L, Aieta M. Circulating tumor cells: utopia or reality? Future Oncol 2013; 9:1337-52. [DOI: 10.2217/fon.13.101] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Circulating tumor cells (CTCs) could be considered a sign of tumor aggressiveness, but highly sensitive and specific methods of CTC detection are necessary owing to the rarity and heterogeneity of CTCs in peripheral blood. This review summarizes recent studies on tumor biology, with particular attention to the metastatic cascade, and the molecular characterization and clinical significance of CTCs. Recent technological approaches to enrich and detect these cells and challenges of CTCs for individualized cancer treatment are also discussed. This review also provides an insight into the positive and negative features of the future potential applications of CTC detection, which sometimes remains still a ‘utopia’, but its actual utility remains among the fastest growing research fields in oncology.
Collapse
Affiliation(s)
- Vincenza Conteduca
- Department of Medical Oncology, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Via Piero Maroncelli 40, 47014 Meldola (FC), Italy
| | | | - Elisabetta Rossi
- Department of Surgery, Oncology & Gastroenterology, Oncology Section, University of Padova, Italy
| | - Valentina Condelli
- Centro di Riferimento Oncologico della Basilicata IRCCS, Rionero in Vulture, Italy
| | - Laura Troiani
- Centro di Riferimento Oncologico della Basilicata IRCCS, Rionero in Vulture, Italy
| | - Michele Aieta
- Centro di Riferimento Oncologico della Basilicata IRCCS, Rionero in Vulture, Italy
| |
Collapse
|