701
|
|
702
|
Hart CD, Galardi F, De Luca F, Pestrin M, Di Leo A. Circulating Tumour Cells as Liquid Biopsy in Breast Cancer—Advancing from Prognostic to Predictive Potential. CURRENT BREAST CANCER REPORTS 2015. [DOI: 10.1007/s12609-014-0177-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
703
|
Ligaba SB, Khurana A, Graham G, Krawczyk E, Jablonski S, Petricoin EF, Glazer RI, Upadhyay G. Multifactorial analysis of conditional reprogramming of human keratinocytes. PLoS One 2015; 10:e0116755. [PMID: 25714835 PMCID: PMC4340869 DOI: 10.1371/journal.pone.0116755] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 12/12/2014] [Indexed: 02/07/2023] Open
Abstract
Co-culture of human primary epithelial cells with irradiated 3T3 fibroblast feeder cells (J2 cells) and the Rho kinase inhibitor Y-27632 (Y) allows for the unrestricted growth of cells of epithelial origin by the process termed conditional reprogramming. To better understand the nature of the signaling processes associated with conditionally reprogrammed cells, the effect of the two critical components of the co-culture conditions, J2 cells and Y, on the growth of human foreskin keratinocytes (HFKs) was evaluated by gene expression profiling, reverse-phase protein arrays and siRNA screening. J2 cells and Y acted cooperatively to down-regulate differentiation, and upregulate proliferation and cell adhesion, including increased pT308Akt and pERK, and reduced TGF-β pathway signaling. These findings establish a mechanistic basis for the unlimited growth potential of human epithelial cells that will be invaluable to assess the effect of genetic changes in pathologic tissues and their response to therapeutic agents.
Collapse
Affiliation(s)
- Segni B. Ligaba
- Department of Pathology, Georgetown University, Washington, DC, 20007, United States of America
| | - Anikita Khurana
- Department of Pathology, Georgetown University, Washington, DC, 20007, United States of America
| | - Garrett Graham
- Department of Oncology, Georgetown University, Washington, DC, 20007, United States of America
| | - Ewa Krawczyk
- Department of Pathology, Georgetown University, Washington, DC, 20007, United States of America
| | - Sandra Jablonski
- Department of Oncology, Georgetown University, Washington, DC, 20007, United States of America
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20007, United States of America
| | - Emanuel F. Petricoin
- Department of Molecular and Microbiology, George Mason University, Manassas, Virginia, 20110, United States of America
| | - Robert I. Glazer
- Department of Oncology, Georgetown University, Washington, DC, 20007, United States of America
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20007, United States of America
| | - Geeta Upadhyay
- Department of Pathology, Georgetown University, Washington, DC, 20007, United States of America
- Department of Oncology, Georgetown University, Washington, DC, 20007, United States of America
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20007, United States of America
- * E-mail:
| |
Collapse
|
704
|
Nanobiotechnology for the Therapeutic Targeting of Cancer Cells in Blood. Cell Mol Bioeng 2015; 8:137-150. [PMID: 25798204 PMCID: PMC4361771 DOI: 10.1007/s12195-015-0381-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Accepted: 01/19/2015] [Indexed: 12/11/2022] Open
Abstract
During metastasis, circulating tumor cells migrate away from a primary tumor via the blood circulation to form secondary tumors in distant organs. Mounting evidence from clinical observations indicates that the number of circulating tumor cells (CTCs) in the blood correlates with the progression of solid tumors before and during chemotherapy. Beyond the well-established role of CTCs as a fluid biopsy, however, the field of targeting CTCs for the prevention or reduction of metastases has just emerged. Conventional cancer therapeutics have a relatively short circulation time in the blood which may render the killing of CTCs inefficient due to reduced exposure of CTCs to drugs. Nevertheless, over the past few decades, the development of nanoparticles and nanoformulations to improve the half-life and release profile of drugs in circulation has rejuvenated certain traditional medicines in the emerging field of CTC neutralization. This review focuses on how the principles of nanomedicine may be applied to target CTCs. Moreover, inspired by the interactions between CTCs and host cells in the blood circulation, novel biomimetic approaches for targeted drug delivery are presented.
Collapse
|
705
|
Nanobiotechnology for the Therapeutic Targeting of Cancer Cells in Blood. Cell Mol Bioeng 2015. [DOI: 10.1007/s12195-015-0378-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
706
|
Xie J, Dong H, Chen H, Zhao R, Sinko PJ, Shen W, Wang J, Lu Y, Yang X, Xie F, Jia L. Exploring cancer metastasis prevention strategy: interrupting adhesion of cancer cells to vascular endothelia of potential metastatic tissues by antibody-coated nanomaterial. J Nanobiotechnology 2015; 13:9. [PMID: 25643843 PMCID: PMC4320453 DOI: 10.1186/s12951-015-0072-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 01/23/2015] [Indexed: 12/13/2022] Open
Abstract
Background Cancer metastasis caused by circulating tumor cells (CTCs) accounts for 90% cancer-related death worldwide. Blocking the circulation of CTCs in bloodstream and their hetero-adhesion to vascular endothelia of the distant metastatic organs may prevent cancer metastasis. Nanomaterial-based intervention with adhesion between CTCs and endothelia has not been reported. Driven by the novel idea that multivalent conjugation of EpCAM and Slex antibodies to dendrimer surface may enhance the capacity and specificity of the nanomaterial conjugates for capturing and down-regulating colorectal CTCs, we conjugated the dendrimer nanomaterial with the EpCAM and Slex antibodies, and examined the capacity of the dual antibody-coated nanomaterial for their roles in interrupting CTCs-related cancer metastasis. Results The antibody-coated nanomaterial was synthesized and characterized. The conjugates specifically bound and captured colon cancer cells SW620. The conjugate inhibited the cells’ viability and their adhesion to fibronectin (Fn)-coated substrate or human umbilical vein endothelial cells (HUVECs) in a concentration-dependent manner. In comparison with SW480 and LoVo cell lines, the activity and adhesion of SW620 to Fn-coated substrate and HUVECs were more specifically inhibited by the dual antibody conjugate because of the higher levels of EpCAM and Slex on SW620 cell surface. The hetero-adhesion between SW620 and Fn-coated substrate, or HUVECs was inhibited by about 60-70%. The dual conjugate showed the inhibition capacity more significant than its corresponding single antibody conjugates. Conclusions The present study provides the new evidence that coating nanomaterials with more than one antibody against CTCs may effectively interfere with the interaction between SW620 and HUVECs. Electronic supplementary material The online version of this article (doi:10.1186/s12951-015-0072-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jingjing Xie
- Cancer Metastasis Alert and Prevention Center, and Biopharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 523 Industry Road, Science Building, 3FL, Fuzhou, Fujian, 350002, China.
| | - Haiyan Dong
- Cancer Metastasis Alert and Prevention Center, and Biopharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 523 Industry Road, Science Building, 3FL, Fuzhou, Fujian, 350002, China.
| | - Hongning Chen
- Cancer Metastasis Alert and Prevention Center, and Biopharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 523 Industry Road, Science Building, 3FL, Fuzhou, Fujian, 350002, China.
| | - Rongli Zhao
- Cancer Metastasis Alert and Prevention Center, and Biopharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 523 Industry Road, Science Building, 3FL, Fuzhou, Fujian, 350002, China.
| | - Patrick J Sinko
- Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA.
| | - Weiyu Shen
- Cancer Metastasis Alert and Prevention Center, and Biopharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 523 Industry Road, Science Building, 3FL, Fuzhou, Fujian, 350002, China.
| | - Jichuang Wang
- Cancer Metastasis Alert and Prevention Center, and Biopharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 523 Industry Road, Science Building, 3FL, Fuzhou, Fujian, 350002, China.
| | - Yusheng Lu
- Cancer Metastasis Alert and Prevention Center, and Biopharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 523 Industry Road, Science Building, 3FL, Fuzhou, Fujian, 350002, China.
| | - Xiang Yang
- Cancer Metastasis Alert and Prevention Center, and Biopharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 523 Industry Road, Science Building, 3FL, Fuzhou, Fujian, 350002, China.
| | - Fangwei Xie
- Department of Medicine Oncology, East Hospital of Xiamen University, Fuzhou, 350004, China.
| | - Lee Jia
- Cancer Metastasis Alert and Prevention Center, and Biopharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 523 Industry Road, Science Building, 3FL, Fuzhou, Fujian, 350002, China.
| |
Collapse
|
707
|
Beije N, Jager A, Sleijfer S. Circulating tumor cell enumeration by the CellSearch system: The clinician’s guide to breast cancer treatment? Cancer Treat Rev 2015; 41:144-50. [DOI: 10.1016/j.ctrv.2014.12.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 12/11/2014] [Accepted: 12/12/2014] [Indexed: 02/02/2023]
|
708
|
Ke Z, Lin M, Chen JF, Choi JS, Zhang Y, Fong A, Liang AJ, Chen SF, Li Q, Fang W, Zhang P, Garcia MA, Lee T, Song M, Lin HA, Zhao H, Luo SC, Hou S, Yu HH, Tseng HR. Programming thermoresponsiveness of NanoVelcro substrates enables effective purification of circulating tumor cells in lung cancer patients. ACS NANO 2015; 9:62-70. [PMID: 25495128 PMCID: PMC4310634 DOI: 10.1021/nn5056282] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Unlike tumor biopsies that can be constrained by problems such as sampling bias, circulating tumor cells (CTCs) are regarded as the "liquid biopsy" of the tumor, providing convenient access to all disease sites, including primary tumor and fatal metastases. Although enumerating CTCs is of prognostic significance in solid tumors, it is conceivable that performing molecular and functional analyses on CTCs will reveal much significant insight into tumor biology to guide proper therapeutic intervention. We developed the Thermoresponsive NanoVelcro CTC purification system that can be digitally programmed to achieve an optimal performance for purifying CTCs from non-small cell lung cancer (NSCLC) patients. The performance of this unique CTC purification system was optimized by systematically modulating surface chemistry, flow rates, and heating/cooling cycles. By applying a physiologically endurable stimulation (i.e., temperature between 4 and 37 °C), the mild operational parameters allow minimum disruption to CTCs' viability and molecular integrity. Subsequently, we were able to successfully demonstrate culture expansion and mutational analysis of the CTCs purified by this CTC purification system. Most excitingly, we adopted the combined use of the Thermoresponsive NanoVelcro system with downstream mutational analysis to monitor the disease evolution of an index NSCLC patient, highlighting its translational value in managing NSCLC.
Collapse
Affiliation(s)
- Zunfu Ke
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
- Address correspondence to , , ,
| | - Millicent Lin
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Jie-Fu Chen
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Jin-sil Choi
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Yang Zhang
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Anna Fong
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - An-Jou Liang
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Shang-Fu Chen
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Qingyu Li
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Wenfeng Fang
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Pingshan Zhang
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Mitch A. Garcia
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Tom Lee
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Min Song
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Hsing-An Lin
- Responsive Organic Materials Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Haichao Zhao
- Responsive Organic Materials Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Shyh-Chyang Luo
- Responsive Organic Materials Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
- Department of Materials Science and Engineering, National Cheng Kung University, Tainan 70101, Taiwan
| | - Shuang Hou
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
- Address correspondence to , , ,
| | - Hsiao-hua Yu
- Responsive Organic Materials Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
- Institute of Chemistry, Academia Sinica, 128 Academia Road Sec. 2, Nankang, Taipei 115, Taiwan
- Address correspondence to , , ,
| | - Hsian-Rong Tseng
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
- Address correspondence to , , ,
| |
Collapse
|
709
|
Vela I, Chen Y. Prostate cancer organoids: a potential new tool for testing drug sensitivity. Expert Rev Anticancer Ther 2015; 15:261-3. [PMID: 25603995 DOI: 10.1586/14737140.2015.1003046] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Recent technical advances have enabled for the first time, reliable in vitro culture of prostate cancer samples as prostate cancer organoids. This breakthrough provides the significant possibility of high throughput drug screening covering the spectrum of prostate cancer phenotypes seen clinically. These advances will enable precision medicine to become a reality, allowing patient samples to be screened for effective therapeutics ex vivo, with tailoring of treatments specific to that individual. This will hopefully lead to enhanced clinical outcomes, avoid morbidity due to ineffective therapies and improve the quality of life in men with advanced prostate cancer.
Collapse
Affiliation(s)
- Ian Vela
- Department of Surgery, Urology Service, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | | |
Collapse
|
710
|
Adorno-Cruz V, Kibria G, Liu X, Doherty M, Junk DJ, Guan D, Hubert C, Venere M, Mulkearns-Hubert E, Sinyuk M, Alvarado A, Caplan AI, Rich J, Gerson SL, Lathia J, Liu H. Cancer stem cells: targeting the roots of cancer, seeds of metastasis, and sources of therapy resistance. Cancer Res 2015; 75:924-9. [PMID: 25604264 DOI: 10.1158/0008-5472.can-14-3225] [Citation(s) in RCA: 192] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
With the goal to remove the roots of cancer, eliminate metastatic seeds, and overcome therapy resistance, the 2014 inaugural International Cancer Stem Cell (CSC) Conference at Cleveland, OH, convened together over 320 investigators, including 55 invited world-class speakers, 25 short oral presenters, and 100 poster presenters, to gain an in-depth understanding of CSCs and explore therapeutic opportunities targeting CSCs. The meeting enabled intriguing discussions on several topics including: genetics and epigenetics; cancer origin and evolution; microenvironment and exosomes; metabolism and inflammation; metastasis and therapy resistance; single cell and heterogeneity; plasticity and reprogramming; as well as other new concepts. Reports of clinical trials targeting CSCs emphasized the urgent need for strategically designing combinational CSC-targeting therapies against cancer.
Collapse
Affiliation(s)
- Valery Adorno-Cruz
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Golam Kibria
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Xia Liu
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Mary Doherty
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Damian J Junk
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Dongyin Guan
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Chris Hubert
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Monica Venere
- Department of Cancer Biology, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Erin Mulkearns-Hubert
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Maksim Sinyuk
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Alvaro Alvarado
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Arnold I Caplan
- Department of Biology, Case Western Reserve University School of Medicine, Cleveland, Ohio. National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Jeremy Rich
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio. National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio. Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Stanton L Gerson
- National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio. Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio. Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Justin Lathia
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio. National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio. Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Huiping Liu
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio. National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio. Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio.
| |
Collapse
|
711
|
Viraka Nellore BP, Kanchanapally R, Pramanik A, Sinha SS, Chavva SR, Hamme A, Ray PC. Aptamer-conjugated graphene oxide membranes for highly efficient capture and accurate identification of multiple types of circulating tumor cells. Bioconjug Chem 2015; 26:235-42. [PMID: 25565372 PMCID: PMC4578366 DOI: 10.1021/bc500503e] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Tumor metastasis is responsible for 1 in 4 deaths in the United States. Though it has been well-documented over past two decades that circulating tumor cells (CTCs) in blood can be used as a biomarker for metastatic cancer, there are enormous challenges in capturing and identifying CTCs with sufficient sensitivity and specificity. Because of the heterogeneous expression of CTC markers, it is now well understood that a single CTC marker is insufficient to capture all CTCs from the blood. Driven by the clear need, this study reports for the first time highly efficient capture and accurate identification of multiple types of CTCs from infected blood using aptamer-modified porous graphene oxide membranes. The results demonstrate that dye-modified S6, A9, and YJ-1 aptamers attached to 20-40 μm porous garphene oxide membranes are capable of capturing multiple types of tumor cells (SKBR3 breast cancer cells, LNCaP prostate cancer cells, and SW-948 colon cancer cells) selectively and simultaneously from infected blood. Our result shows that the capture efficiency of graphene oxide membranes is ~95% for multiple types of tumor cells; for each tumor concentration, 10 cells are present per milliliter of blood sample. The selectivity of our assay for capturing targeted tumor cells has been demonstrated using membranes without an antibody. Blood infected with different cells also has been used to demonstrate the targeted tumor cell capturing ability of aptamer-conjugated membranes. Our data also demonstrate that accurate analysis of multiple types of captured CTCs can be performed using multicolor fluorescence imaging. Aptamer-conjugated membranes reported here have good potential for the early diagnosis of diseases that are currently being detected by means of cell capture technologies.
Collapse
Affiliation(s)
- Bhanu Priya Viraka Nellore
- Department of Chemistry and Biochemistry, Jackson State University , Jackson, Mississippi 39217, United States
| | | | | | | | | | | | | |
Collapse
|
712
|
Cayrefourcq L, Mazard T, Joosse S, Solassol J, Ramos J, Assenat E, Schumacher U, Costes V, Maudelonde T, Pantel K, Alix-Panabières C. Establishment and characterization of a cell line from human circulating colon cancer cells. Cancer Res 2015; 75:892-901. [PMID: 25592149 DOI: 10.1158/0008-5472.can-14-2613] [Citation(s) in RCA: 301] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Circulating tumor cells (CTC) in blood are promising new biomarkers potentially useful for prognostic prediction and monitoring of therapies in patients with solid tumors including colon cancer. Moreover, CTC research opens a new avenue for understanding the biology of metastasis in patients with cancer. However, an in-depth investigation of CTCs is hampered by the very low number of these cells, especially in the blood of patients with colorectal cancer. Thus, the establishment of cell cultures and permanent cell lines from CTCs has become the most challenging task over the past year. Here, we describe, for the first time, the establishment of cell cultures and a permanent cell line from CTCs of one patient with colon cancer. The cell line designated CTC-MCC-41 has been cultured for more than one year, and the cells have been characterized at the genome, transcriptome, proteome, and secretome levels. This thorough analysis showed that CTC-MCC-41 cells resemble characteristics of the original tumor cells in the patient with colon cancer and display a stable phenotype characterized by an intermediate epithelial/mesenchymal phenotype, stem cell-like properties, and an osteomimetic signature, indicating a bone marrow origin. Functional studies showed that CTC-MCC-41 cells induced rapidly in vitro endothelial cell tube formation and in vivo tumors after xenografting in immunodeficient mice. The establishment of this first colon cancer CTC line allows now a wealth of functional studies on the biology of CTCs as well as in vitro and in vivo drug testing.
Collapse
Affiliation(s)
- Laure Cayrefourcq
- Laboratory of Rare Human Circulating Cells (LCCRH), Department of Cellular and Tissular Biopathology of Tumors, University Medical Centre, Montpellier, France. EA2415 - Help for Personalized Decision: Methodological Aspects, University Institute of Clinical Research, Montpellier University, Montpellier, France
| | - Thibault Mazard
- Department of Medical Oncology, University Medical Centre, Montpellier, France
| | - Simon Joosse
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jérôme Solassol
- Laboratory of Hormonal and Cell Biology, Department of Cellular and Tissular Biopathology of Tumors, University Medical Centre, Montpellier, France
| | - Jeanne Ramos
- Laboratory of Pathology, Department of Cellular and Tissular Biopathology of Tumors, University Medical Centre, Montpellier, France
| | - Eric Assenat
- Department of Medical Oncology, University Medical Centre, Montpellier, France
| | - Udo Schumacher
- Department of Anatomy und Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Valérie Costes
- Laboratory of Pathology, Department of Cellular and Tissular Biopathology of Tumors, University Medical Centre, Montpellier, France
| | - Thierry Maudelonde
- EA2415 - Help for Personalized Decision: Methodological Aspects, University Institute of Clinical Research, Montpellier University, Montpellier, France. Laboratory of Hormonal and Cell Biology, Department of Cellular and Tissular Biopathology of Tumors, University Medical Centre, Montpellier, France
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), Department of Cellular and Tissular Biopathology of Tumors, University Medical Centre, Montpellier, France. EA2415 - Help for Personalized Decision: Methodological Aspects, University Institute of Clinical Research, Montpellier University, Montpellier, France.
| |
Collapse
|
713
|
Burke JE, Williams RL. Synergy in activating class I PI3Ks. Trends Biochem Sci 2015; 40:88-100. [PMID: 25573003 DOI: 10.1016/j.tibs.2014.12.003] [Citation(s) in RCA: 156] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 12/03/2014] [Accepted: 12/05/2014] [Indexed: 12/20/2022]
Abstract
The class I phosphoinositide 3-kinases (PI3Ks) are lipid kinases that transduce a host of cellular signals and regulate a broad range of essential functions including growth, proliferation, and migration. As such, PI3Ks have pivotal roles in diseases such as cancer, diabetes, primary immune disorders, and inflammation. These enzymes are activated downstream of numerous activating stimuli including receptor tyrosine kinases, G protein-coupled receptors (GPCRs), and the Ras superfamily of small G proteins. A major challenge is to decipher how each PI3K isoform is able to successfully synergize these inputs into their intended signaling function. This article highlights recent progress in characterizing the molecular mechanisms of PI3K isoform-specific activation pathways, as well as novel roles for PI3Ks in human diseases, specifically cancer and immune diseases.
Collapse
Affiliation(s)
- John E Burke
- Department of Biochemistry and Microbiology, University of Victoria, 3800 Finnerty Drive, Victoria BC, V8P 5C2, Canada.
| | - Roger L Williams
- Medical Research Council (MRC) Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| |
Collapse
|
714
|
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
|
715
|
Dokmanovic M, Wu WJ. Monitoring Trastuzumab Resistance and Cardiotoxicity: A Tale of Personalized Medicine. Adv Clin Chem 2015; 70:95-130. [PMID: 26231486 DOI: 10.1016/bs.acc.2015.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
While approval of trastuzumab, a recombinant monoclonal antibody directed against HER2, along with a diagnostic kit to detect breast cancers which are positive for HER2 overexpression, has advanced a new era of stratified and personalized medicine, it also created several challenges to our scientific and clinical practice. These problems include trastuzumab resistance and trastuzumab-induced cardiotoxicity. In this review, we will summarize data from the literature regarding mechanisms of trastuzumab resistance and trastuzumab-induced cardiotoxicity and present some promising model systems that may advance our understanding of these mechanisms. Our discussion will include development of circulating tumor cells and circulating tumor DNA for monitoring tumor burden, of patient-derived xenograft models for preclinical testing of novel therapies, and of novel therapeutic strategies for trastuzumab-resistance and possible integration of these strategies in the design of co-clinical studies for testing in relevant patient subpopulations.
Collapse
|
716
|
J. McGrail D, 1 School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332-0100;, S. Patel K, N. Khambhati N, Pithadia K, R. Dawson M. Utilizing temporal variations in chemotherapeutic response to improve breast cancer treatment efficacy. AIMS BIOENGINEERING 2015. [DOI: 10.3934/bioeng.2015.4.310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
717
|
Circulating Tumor Cells: Who is the Killer? CANCER MICROENVIRONMENT 2014; 7:161-76. [PMID: 25527469 PMCID: PMC4275541 DOI: 10.1007/s12307-014-0164-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 11/27/2014] [Indexed: 01/05/2023]
Abstract
This article is a critical note on the subject of Circulating Tumor Cells (CTC). It takes into account the tumor identity of Circulating Tumor Cells as cancer seeds in transit from primary to secondary soils, rather than as a “biomarker”, and considers the help this field could bring to cancer patients. It is not meant to duplicate information already available in a large number of reviews, but to stimulate considerations, further studies and development helping the clinical use of tumor cells isolated from blood as a modern personalized, non-invasive, predictive test to improve cancer patients’ life. The analysis of CTC challenges, methodological bias and critical issues points out to the need of referring to tumor cells extracted from blood without any bias and identified by cytopathological diagnosis as Circulating Cancer Cells (CCC). Finally, this article highlights recent developments and identifies burning questions which should be addressed to improve our understanding of the domain of CCC and their potential to change the clinical practice.
Collapse
|
718
|
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
|
719
|
HSP90 empowers evolution of resistance to hormonal therapy in human breast cancer models. Proc Natl Acad Sci U S A 2014; 111:18297-302. [PMID: 25489079 DOI: 10.1073/pnas.1421323111] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The efficacy of hormonal therapies for advanced estrogen receptor-positive breast cancers is limited by the nearly inevitable development of acquired resistance. Efforts to block the emergence of resistance have met with limited success, largely because the mechanisms underlying it are so varied and complex. Here, we investigate a new strategy aimed at the very processes by which cancers evolve resistance. From yeast to vertebrates, heat shock protein 90 (HSP90) plays a unique role among molecular chaperones by promoting the evolution of heritable new traits. It does so by regulating the folding of a diverse portfolio of metastable client proteins, many of which mediate adaptive responses that allow organisms to adapt and thrive in the face of diverse challenges, including those posed by drugs. Guided by our previous work in pathogenic fungi, in which very modest HSP90 inhibition impairs resistance to mechanistically diverse antifungals, we examined the effect of similarly modest HSP90 inhibition on the emergence of resistance to antiestrogens in breast cancer models. Even though this degree of inhibition fell below the threshold for proteotoxic activation of the heat-shock response and had no overt anticancer activity on its own, it dramatically impaired the emergence of resistance to hormone antagonists both in cell culture and in mice. Our findings strongly support the clinical testing of combined hormone antagonist-low-level HSP90 inhibitor regimens in the treatment of metastatic estrogen receptor-positive breast cancer. At a broader level, they also provide promising proof of principle for a generalizable strategy to combat the pervasive problem of rapidly emerging resistance to molecularly targeted therapeutics.
Collapse
|
720
|
Hoffman RM. Orthotopic mouse models of tumor metastasis expressing fluorescent reporters produce imageable circulating tumor cells. CANCER MICROENVIRONMENT 2014; 7:133-8. [PMID: 25417145 DOI: 10.1007/s12307-014-0154-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 09/15/2014] [Indexed: 02/05/2023]
Abstract
Circulating tumor cells (CTC) are of high importance, since they are potential metastatic precursors and are readily available for prognostic analysis and treatment testing. In this review, we demonstrate the great power that green fluorescent protein (GFP) labeling and orthotopic mouse models of cancer confer to the study of CTCs for isolation and characterization, including metastatic testing in mice and the chick embryo as well as drug response testing in vitro. We also describe a facile method to label patient CTCs ex vivo using a telomerase-expressing GFP-containing adenovirus that will allow the CTC studies described in this review to be translated clinically.
Collapse
Affiliation(s)
- Robert M Hoffman
- AntiCancer, Inc., 7917 Ostrow Street, San Diego, CA, 92111, USA,
| |
Collapse
|
721
|
La culture ex vivo de cellules tumorales circulantes : un réel espoir de thérapeutique personnalisée? Bull Cancer 2014. [DOI: 10.1684/bdc.2014.2045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
722
|
Lin M, Chen JF, Lu YT, Zhang Y, Song J, Hou S, Ke Z, Tseng HR. Nanostructure embedded microchips for detection, isolation, and characterization of circulating tumor cells. Acc Chem Res 2014; 47:2941-50. [PMID: 25111636 PMCID: PMC4204926 DOI: 10.1021/ar5001617] [Citation(s) in RCA: 172] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Circulating
tumor cells (CTCs) are cancer cells that break away
from either a primary tumor or a metastatic site and circulate in
the peripheral blood as the cellular origin of metastasis. With their
role as a “tumor liquid biopsy”, CTCs provide convenient
access to all disease sites, including that of the primary tumor and
the site of fatal metastases. It is conceivable that detecting and
analyzing CTCs will provide insightful information in assessing the
disease status without the flaws and limitations encountered in performing
conventional tumor biopsies. However, identifying CTCs in patient
blood samples is technically challenging due to the extremely low
abundance of CTCs among a large number of hematologic cells. To address
this unmet need, there have been significant research endeavors, especially
in the fields of chemistry, materials science, and bioengineering,
devoted to developing CTC detection, isolation, and characterization
technologies. Inspired by the nanoscale interactions observed
in the tissue microenvironment,
our research team at UCLA pioneered a unique concept of “NanoVelcro”
cell-affinity substrates, in which CTC capture agent-coated nanostructured
substrates were utilized to immobilize CTCs with high efficiency.
The working mechanism of NanoVelcro cell-affinity substrates mimics
that of Velcro: when the two fabric strips of a Velcro fastener are
pressed together, tangling between the hairy surfaces on two strips
leads to strong binding. Through continuous evolution, three generations
(gens) of NanoVelcro CTC chips have been established to achieve different
clinical utilities. The first-gen NanoVelcro chip, composed of a silicon
nanowire substrate (SiNS) and an overlaid microfluidic chaotic mixer,
was created for CTC enumeration. Side-by-side analytical validation
studies using clinical blood samples suggested that the sensitivity
of first-gen NanoVelcro chip outperforms that of FDA-approved CellSearch.
In conjunction with the use of the laser microdissection (LMD) technique,
second-gen NanoVelcro chips (i.e., NanoVelcro-LMD), based on polymer
nanosubstrates, were developed for single-CTC isolation. The individually
isolated CTCs can be subjected to single-CTC genotyping (e.g., Sanger
sequencing and next-generation sequencing, NGS) to verify the CTC’s
role as tumor liquid biopsy. Created by grafting of thermoresponsive
polymer brushes onto SiNS, third-gen NanoVelcro chips (i.e., Thermoresponsive
NanoVelcro) have demonstrated the capture and release of CTCs at 37
and 4 °C, respectively. The temperature-dependent conformational
changes of polymer brushes can effectively alter the accessibility
of the capture agent on SiNS, allowing for rapid CTC purification
with desired viability and molecular integrity. This Account
summarizes the continuous evolution of NanoVelcro
CTC assays from the emergence of the original idea all the way to
their applications in cancer research. We envision that NanoVelcro
CTC assays will lead the way for powerful and cost-efficient diagnostic
platforms for researchers to better understand underlying disease
mechanisms and for physicians to monitor real-time disease progression.
Collapse
Affiliation(s)
- Millicent Lin
- Department
of Pathology, The First Affiliated hospital of Sun Yat-sen University, Guangzhou, 510080 Guangdong, People’s Republic of China
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Jie-Fu Chen
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Yi-Tsung Lu
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Yang Zhang
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Jinzhao Song
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Shuang Hou
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Zunfu Ke
- Department
of Pathology, The First Affiliated hospital of Sun Yat-sen University, Guangzhou, 510080 Guangdong, People’s Republic of China
| | - Hsian-Rong Tseng
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| |
Collapse
|
723
|
Ignatiadis M, Dawson SJ. Circulating tumor cells and circulating tumor DNA for precision medicine: dream or reality? Ann Oncol 2014; 25:2304-2313. [PMID: 25336116 DOI: 10.1093/annonc/mdu480] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Next-generation sequencing studies have provided further evidence to support the notion that cancer is a disease characterized by Darwinian evolution. Today, we often fail to capture this evolution and treatment decisions, even in the metastatic setting, are often based on analysis of primary tumor diagnosed years ago. Currently, this is considered a major reason for treatment failures in cancer care. Recent technological advances in the detection and characterization of circulating tumor cells and circulating tumor DNA might address this and allow for treatment tailoring based on real-time monitoring of tumor evolution. In this review, we summarize the most important recent findings in the field, focusing on challenges and opportunities in moving these tools forward in clinical practice.
Collapse
Affiliation(s)
- M Ignatiadis
- Department of Medical Oncology and Breast Cancer Translational Research Laboratory, J. C. Heuson, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.
| | - S-J Dawson
- Division of Cancer Medicine; Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| |
Collapse
|
724
|
Giuliano M, Giordano A, Jackson S, De Giorgi U, Mego M, Cohen EN, Gao H, Anfossi S, Handy BC, Ueno NT, Alvarez RH, De Placido S, Valero V, Hortobagyi GN, Reuben JM, Cristofanilli M. Circulating tumor cells as early predictors of metastatic spread in breast cancer patients with limited metastatic dissemination. Breast Cancer Res 2014; 16:440. [PMID: 25223629 PMCID: PMC4303121 DOI: 10.1186/s13058-014-0440-8] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 09/01/2014] [Indexed: 12/24/2022] Open
Abstract
Introduction Traditional factors currently used for prognostic stratification do not always adequately predict treatment response and disease evolution in advanced breast cancer patients. Therefore, the use of blood-based markers, such as circulating tumor cells (CTCs), represents a promising complementary strategy for disease monitoring. In this retrospective study, we explored the role of CTC counts as predictors of disease evolution in breast cancer patients with limited metastatic dissemination. Methods A total of 492 advanced breast cancer patients who had a CTC count assessed by CellSearch prior to starting a new line of systemic therapy were eligible for this analysis. Using the threshold of 5 CTCs/7.5 ml of blood, pretreatment CTC counts were correlated in the overall population with metastatic site distribution, evaluated at baseline and at the time of treatment failure, using Fisher’s exact test. Time to visceral progression and time to the development of new metastatic lesions and sites were estimated in patients with nonvisceral metastases and with single-site metastatic disease, respectively, by the Kaplan-Meier method. Survival times were compared between groups according to pretreatment CTC count by logrank test. Results In the overall population, a pretreatment level ≥5 CTCs/7.5 ml was associated with an increased baseline number of metastatic sites compared with <5 CTCs/7.5 ml (P = 0.0077). At the time of treatment failure, patients with ≥5 CTCs/7.5 ml more frequently developed new metastatic lesions and sites compared with those with <5 CTCs/7.5 ml (development of new lesions: P = 0.0002; development of new sites: P = 0.0031). Among patients with disease originally confined to nonvisceral sites, ≥5 CTCs/7.5 ml was associated with remarkably shorter time to visceral metastases (P = 0.0021) and overall survival (P = 0.0006) compared with <5 CTCs/7.5 ml. In patients with single-site metastatic disease, ≥5 CTCs/7.5 ml was associated with a significant reduction of the time to development of new metastatic sites (P = 0.0051) and new lesions (P = 0.0002) and with worse overall survival (P = 0.0101). Conclusion Our results suggest that baseline CTC counts can be used as an early predictor of metastatic potential in breast cancer patients with limited metastatic dissemination. Electronic supplementary material The online version of this article (doi:10.1186/s13058-014-0440-8) contains supplementary material, which is available to authorized users.
Collapse
|
725
|
|
726
|
|
727
|
Lianidou ES. Molecular characterization of circulating tumor cells: Holy Grail for personalized cancer treatment? Clin Chem 2014; 60:1249-51. [PMID: 25142245 DOI: 10.1373/clinchem.2014.230144] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Evi S Lianidou
- Analysis of Circulating Tumor Cells (ACTC) Lab, Laboratory of Analytical Chemistry, Department of Chemistry, University of Athens, Athens, Greece.
| |
Collapse
|
728
|
Roving tumour cells tracked down. Nature 2014. [DOI: 10.1038/511266c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|