51
|
León-Mateos L, Vieito M, Anido U, López López R, Muinelo Romay L. Clinical Application of Circulating Tumour Cells in Prostate Cancer: From Bench to Bedside and Back. Int J Mol Sci 2016; 17:E1580. [PMID: 27657044 PMCID: PMC5037845 DOI: 10.3390/ijms17091580] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 09/05/2016] [Accepted: 09/09/2016] [Indexed: 01/19/2023] Open
Abstract
Prostate cancer is the most common cancer in men worldwide. To improve future drug development and patient management, surrogate biomarkers associated with relevant outcomes are required. Circulating tumour cells (CTCs) are tumour cells that can enter the circulatory system, and are principally responsible for the development of metastasis at distant sites. In recent years, interest in detecting CTCs as a surrogate biomarker has ghiiukjrown. Clinical studies have revealed that high levels of CTCs in the blood correlate with disease progression in patients with prostate cancer; however, their predictive value for monitoring therapeutic response is less clear. Despite the important progress in CTC clinical development, there are critical requirements for the implementation of their analysis as a routine oncology tool. The goal of the present review is to provide an update on the advances in the clinical validation of CTCs as a surrogate biomarker and to discuss the principal obstacles and main challenges to their inclusion in clinical practice.
Collapse
Affiliation(s)
- Luis León-Mateos
- Axencia Galega de Coñecemento en Saúde (ACIS), SERGAS, Avda, Fernando de Casa Novoa, Santiago de Compostela 15707, Spain.
| | - María Vieito
- London Regional Cancer Program, London Health Sciences Centre, London, ON N6A 4L6, Canada.
| | - Urbano Anido
- Translational Medical Oncology/Liquid Biopsy Analysis Unit, Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela 15706, Spain.
| | - Rafael López López
- Translational Medical Oncology/Liquid Biopsy Analysis Unit, Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela 15706, Spain.
| | - Laura Muinelo Romay
- Translational Medical Oncology/Liquid Biopsy Analysis Unit, Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela 15706, Spain.
| |
Collapse
|
52
|
Nagrath S, Jack RM, Sahai V, Simeone DM. Opportunities and Challenges for Pancreatic Circulating Tumor Cells. Gastroenterology 2016; 151:412-26. [PMID: 27339829 DOI: 10.1053/j.gastro.2016.05.052] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 05/25/2016] [Accepted: 06/07/2016] [Indexed: 12/30/2022]
Abstract
Sensitive and reproducible platforms have been developed for detection, isolation, and enrichment of circulating tumor cells (CTCs)-rare cells that enter the blood from solid tumors, including those of the breast, prostate gland, lung, pancreas, and colon. These might be used as biomarkers in diagnosis or determination of prognosis. CTCs are no longer simply detected and quantified; they are now used in ex vivo studies of anticancer agents and early detection. We review what we have recently learned about CTCs from pancreatic tumors, describing advances in their isolation and analysis and challenges to their clinical utility. We summarize technologies used to isolate CTCs from blood samples of patients with pancreatic cancer, including immunoaffinity and label-free physical attribute-based capture. We explain methods of CTC analysis and how findings from these studies might be used to detect cancer at earlier stages, monitor disease progression, and determine prognosis. We review studies that have expanded CTCs for testing of anticancer agents and how these approaches might be used to personalize treatment. Advances in the detection, isolation, and analysis of CTCs have increased our understanding of the dissemination and progression of pancreatic cancer. However, standardization of methodologies and prospective studies are needed for this emerging technology to have a significant effect on clinical care.
Collapse
Affiliation(s)
- Sunitha Nagrath
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan; Biointerfaces Program, University of Michigan, Ann Arbor, Michigan; Translational Oncology Program, University of Michigan, Ann Arbor, Michigan.
| | - Rhonda M Jack
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan; Biointerfaces Program, University of Michigan, Ann Arbor, Michigan; Translational Oncology Program, University of Michigan, Ann Arbor, Michigan
| | - Vaibhav Sahai
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan; Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Diane M Simeone
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan; Department of Surgery, University of Michigan, Ann Arbor, Michigan; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
53
|
Abstract
FUNDAMENTAL BASIS OF METASTATIC PROCESS Metastatic process is described as a "dissemination of neoplastic cells in a distant secondary site, in which cells proliferate to develop a mass of cells partially differentiated". The vast majority of death in solid cancers is the consequence of metastasis development which lead to vital organ dysfunction. In the present review, either recent discoveries or controversial subjects associated with metastasis process will be discussed. Indeed epithelia-mesenchymal transition (EMT), circulating tumor cells, tumor dormancy, colonization in distant organ and cancer stem cells are tackled.
Collapse
Affiliation(s)
- Jean-Marc Pascussi
- UMR 5203, Inserm U1191, Université Montpellier, Institut de Génomique Fonctionnelle, Montpellier, France
| | - Julie Giraud
- UMR 5203, Inserm U1191, Université Montpellier, Institut de Génomique Fonctionnelle, Montpellier, France
| | - Emmanuelle Samalin
- Institut Régional du Cancer de Montpellier (ICM), Val d'Aurelle, Montpellier, France
| | - Fanny Grillet
- UMR 5203, Inserm U1191, Université Montpellier, Institut de Génomique Fonctionnelle, Montpellier, France
| | - Julie Pannequin
- UMR 5203, Inserm U1191, Université Montpellier, Institut de Génomique Fonctionnelle, Montpellier, France.
| |
Collapse
|
54
|
Brouwer A, De Laere B, Peeters D, Peeters M, Salgado R, Dirix L, Van Laere S. Evaluation and consequences of heterogeneity in the circulating tumor cell compartment. Oncotarget 2016; 7:48625-48643. [PMID: 26980749 PMCID: PMC5217044 DOI: 10.18632/oncotarget.8015] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 02/18/2016] [Indexed: 02/06/2023] Open
Abstract
A growing understanding of the molecular biology of cancer and the identification of specific aberrations driving cancer evolution have led to the development of various targeted agents. Therapeutic decisions concerning these drugs are often guided by single biopsies of the primary tumor. Yet, it is well known that tumors can exhibit significant heterogeneity and change over time as a result of selective pressure. Circulating tumor cells (CTCs) are shed from various tumor sites and are thought to represent the molecular landscape of a patient's overall tumor burden. Moreover, a minimal-invasive liquid biopsy facilitates monitoring of clonal evolution during therapy pressure and disease progression in real-time. While more information becomes available regarding heterogeneity among CTCs, comparison between these studies is needed. In this review, we focus on the genomic and transcriptional heterogeneity found in the CTC compartment, and its significance for clinical decision making.
Collapse
Affiliation(s)
- Anja Brouwer
- Center for Oncological Research (CORE), University of Antwerp, Antwerp, Belgium
- Department of Oncology, Antwerp University Hospital, Antwerp, Belgium
| | - Bram De Laere
- Center for Oncological Research (CORE), University of Antwerp, Antwerp, Belgium
| | - Dieter Peeters
- Center for Oncological Research (CORE), University of Antwerp, Antwerp, Belgium
- Department of Pathology, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
| | - Marc Peeters
- Center for Oncological Research (CORE), University of Antwerp, Antwerp, Belgium
- Department of Oncology, Antwerp University Hospital, Antwerp, Belgium
| | - Roberto Salgado
- Center for Oncological Research (CORE), University of Antwerp, Antwerp, Belgium
- Department of Pathology, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
- Breast Cancer Translational Research Laboratory, Jules Bordet Institute, Brussels, Belgium
| | - Luc Dirix
- Center for Oncological Research (CORE), University of Antwerp, Antwerp, Belgium
- Department of Oncology, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
| | - Steven Van Laere
- Center for Oncological Research (CORE), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
55
|
Abstract
Isolation and analysis of cancer cells from body fluids have significant implications in diagnosis and therapeutic treatment of cancers. Circulating tumor cells (CTCs) are cancer cells circulating in the peripheral blood or spreading iatrogenically into blood vessels, which is an early step in the cascade of events leading to cancer metastasis. Therefore, CTCs can be used for diagnosing for therapeutic treatment, prognosing a given anticancer intervention, and estimating the risk of metastatic relapse. However, isolation of CTCs is a significant technological challenge due to their rarity and low recovery rate using traditional purification techniques. Recently microfluidic devices represent a promising platform for isolating cancer cells with high efficiency in processing complex cellular fluids, with simplicity, sensitivity, and throughput. This review summarizes recent methods of CTC isolation and analysis, as well as their applications in clinical studies.
Collapse
Affiliation(s)
- J Zhang
- University of Florida, Gainesville, FL, United States
| | - K Chen
- University of Florida, Gainesville, FL, United States
| | - Z H Fan
- University of Florida, Gainesville, FL, United States.
| |
Collapse
|
56
|
Abstract
Cell-to-cell variation and heterogeneity are fundamental and intrinsic characteristics of stem cell populations, but these differences are masked when bulk cells are used for omic analysis. Single-cell sequencing technologies serve as powerful tools to dissect cellular heterogeneity comprehensively and to identify distinct phenotypic cell types, even within a 'homogeneous' stem cell population. These technologies, including single-cell genome, epigenome, and transcriptome sequencing technologies, have been developing rapidly in recent years. The application of these methods to different types of stem cells, including pluripotent stem cells and tissue-specific stem cells, has led to exciting new findings in the stem cell field. In this review, we discuss the recent progress as well as future perspectives in the methodologies and applications of single-cell omic sequencing technologies.
Collapse
Affiliation(s)
- Lu Wen
- Biodynamic Optical Imaging Center, College of Life Sciences, Peking University, Beijing, 100871, China.
| | - Fuchou Tang
- Biodynamic Optical Imaging Center, College of Life Sciences, Peking University, Beijing, 100871, China. .,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
57
|
Abstract
Single-cell RNA-sequencing (scRNA-seq) has emerged as a revolutionary tool that allows us to address scientific questions that eluded examination just a few years ago. With the advantages of scRNA-seq come computational challenges that are just beginning to be addressed. In this article, we highlight the computational methods available for the design and analysis of scRNA-seq experiments, their advantages and disadvantages in various settings, the open questions for which novel methods are needed, and expected future developments in this exciting area.
Collapse
Affiliation(s)
- Rhonda Bacher
- Department of Statistics, University of Wisconsin, Madison, WI, 53706, USA
| | - Christina Kendziorski
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI, 53726, USA.
| |
Collapse
|
58
|
Cho WJ, Oliveira DSM, Najy AJ, Mainetti LE, Aoun HD, Cher ML, Heath E, Kim HRC, Bonfil RD. Gene expression analysis of bone metastasis and circulating tumor cells from metastatic castrate-resistant prostate cancer patients. J Transl Med 2016; 14:72. [PMID: 26975354 PMCID: PMC4791970 DOI: 10.1186/s12967-016-0829-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 03/05/2016] [Indexed: 01/15/2023] Open
Abstract
Background Characterization of genes linked to bone metastasis is critical for identification of novel prognostic or predictive biomarkers and potential therapeutic targets in metastatic castrate-resistant prostate cancer (mCRPC). Although bone marrow core biopsies (BMBx) can be obtained for gene profiling, the procedure itself is invasive and uncommon practice in mCRPC patients. Conversely, circulating tumor cells (CTCs), which are likely to stem from bone metastases, can be isolated from blood. The goals of this exploratory study were to establish a sensitive methodology to analyze gene expression in BMBx and CTCs, and to determine whether the presence or absence of detectable gene expression is concordant in matching samples from mCRPC patients. Methods The CellSearch® platform was used to enrich and enumerate CTCs. Low numbers of PC3 prostate cancer (PCa) cells were spiked into normal blood to assess cell recovery rate. RNA extracted from recovered PC3 cells was amplified using an Eberwine-based procedure to obtain antisense mRNA (aRNA), and assess the linearity of the RNA amplification method. In this pilot study, RNAs extracted from CTCs and PCa cells microdissected from formalin-fixed paraffin-embedded BMBx, were amplified to obtain aRNA and assess the expression of eight genes functionally relevant to PCa bone metastasis using RT-PCR. Results RNAs were successfully extracted from as few as 1–5 PCa cells in blood samples. The relative expression levels of reference genes were maintained after RNA amplification. The integrity of the amplified RNA was also demonstrated by RT-PCR analysis using primer sets that target the 5′-end, middle, and 3′-end of reference mRNA. We found that in 21 out of 28 comparisons, the presence or absence of detectable gene expression in CTCs and PCa cells microdissected from single bone lesions of the same patients was concordant. Conclusions This exploratory analysis suggests that aRNA amplification through in vitro transcription may be useful as a method to detect gene expression in small numbers of CTCs and tumor cells microdissected from bone metastatic lesions. In some cases, gene expression in CTCs and BMBxs was not concordant, raising questions about using CTC gene expression to make clinical decisions. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-0829-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Won Jin Cho
- Department of Urology, Wayne State University School of Medicine and Karmanos Cancer Institute, 540 E. Canfield, Scott Hall # 9105, Detroit, MI, 4820, USA
| | - Daniel S M Oliveira
- Department of Urology, Wayne State University School of Medicine and Karmanos Cancer Institute, 540 E. Canfield, Scott Hall # 9105, Detroit, MI, 4820, USA
| | - Abdo J Najy
- Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, USA
| | - Leandro E Mainetti
- Department of Urology, Wayne State University School of Medicine and Karmanos Cancer Institute, 540 E. Canfield, Scott Hall # 9105, Detroit, MI, 4820, USA
| | - Hussein D Aoun
- Department of Radiology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, USA
| | - Michael L Cher
- Department of Urology, Wayne State University School of Medicine and Karmanos Cancer Institute, 540 E. Canfield, Scott Hall # 9105, Detroit, MI, 4820, USA.,Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, USA
| | - Elisabeth Heath
- Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, USA
| | - Hyeong-Reh C Kim
- Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, USA
| | - R Daniel Bonfil
- Department of Urology, Wayne State University School of Medicine and Karmanos Cancer Institute, 540 E. Canfield, Scott Hall # 9105, Detroit, MI, 4820, USA. .,Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, USA. .,Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, USA.
| |
Collapse
|
59
|
Ferreira MM, Ramani VC, Jeffrey SS. Circulating tumor cell technologies †. Mol Oncol 2016; 10:374-94. [PMID: 26897752 PMCID: PMC5528969 DOI: 10.1016/j.molonc.2016.01.007] [Citation(s) in RCA: 359] [Impact Index Per Article: 44.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 01/16/2016] [Accepted: 01/19/2016] [Indexed: 02/08/2023] Open
Abstract
Circulating tumor cells, a component of the “liquid biopsy”, hold great potential to transform the current landscape of cancer therapy. A key challenge to unlocking the clinical utility of CTCs lies in the ability to detect and isolate these rare cells using methods amenable to downstream characterization and other applications. In this review, we will provide an overview of current technologies used to detect and capture CTCs with brief insights into the workings of individual technologies. We focus on the strategies employed by different platforms and discuss the advantages of each. As our understanding of CTC biology matures, CTC technologies will need to evolve, and we discuss some of the present challenges facing the field in light of recent data encompassing epithelial‐to‐mesenchymal transition, tumor‐initiating cells, and CTC clusters. We present a comprehensive overview of CTC detection and capture technologies. We provide a conceptual description of strategies used in different technologies. We highlight the key features of individual technologies. We discuss CTC technology performance in the context of clinical studies.
Collapse
Affiliation(s)
- Meghaan M Ferreira
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Vishnu C Ramani
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Stefanie S Jeffrey
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
60
|
Court CM, Ankeny JS, Sho S, Tomlinson JS. Circulating Tumor Cells in Gastrointestinal Cancer: Current Practices and Future Directions. Cancer Treat Res 2016; 168:345-376. [PMID: 29206383 DOI: 10.1007/978-3-319-34244-3_17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
GI cancers are the leading cause of cancer-related death worldwide primarily due to a combination of late presentation and aggressive biology. The lack of adequate biomarkers for screening, diagnosis, staging, and prognosis confounds clinical decision-making and delays potentially effective therapies. Circulating tumor cells (CTCs) are a new biomarker with particular promise in GI cancers, potentially offering clinicians and researchers real-time access to tumor tissue in a reliable, safe, and cost-effective manner. Preliminary studies have investigated the potential clinical utility of CTCs for all GI cancer types with promising results. Furthermore, advances in single cell analytics have been successfully applied to CTCs, allowing for exciting new clinical and research applications. In this chapter, we will review the current state of CTC research in GI cancers as well as the potential future applications that are currently being developed.
Collapse
|
61
|
Salvianti F, Pazzagli M, Pinzani P. Single circulating tumor cell sequencing as an advanced tool in cancer management. Expert Rev Mol Diagn 2015; 16:51-63. [PMID: 26560087 DOI: 10.1586/14737159.2016.1116942] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Circulating tumor cells (CTCs) shed by the primary tumor and metastases are considered a real-time 'liquid biopsy', reflecting the disease complexity that evolves during progression, showing in its late stages different genetic, epigenetic and expression features. Consequently, heterogeneity and development of characteristic features upon disease progression are the two main goals that emerging technologies should account for in view of a clinical application. Single-cell analysis, now possible due to technological advances, may help elucidate tumor heterogeneity at the CTC level. This review focuses on the necessary steps for the analysis of CTCs at the single-cell level. A concise overview is given on the alternative methods referring in particular to studies on the mutational status of single CTCs from cancer patients.
Collapse
Affiliation(s)
- Francesca Salvianti
- a Department of Clinical, Experimental and Biomedical Sciences , University of Florence , Firenze , Italy
| | - Mario Pazzagli
- a Department of Clinical, Experimental and Biomedical Sciences , University of Florence , Firenze , Italy
| | - Pamela Pinzani
- a Department of Clinical, Experimental and Biomedical Sciences , University of Florence , Firenze , Italy
| |
Collapse
|
62
|
Circulating tumor cells in breast cancer--current status and perspectives. Crit Rev Oncol Hematol 2015; 97:22-9. [PMID: 26563820 DOI: 10.1016/j.critrevonc.2015.10.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 09/12/2015] [Accepted: 10/27/2015] [Indexed: 12/15/2022] Open
Abstract
The phenomenon of tumor cell dissemination through the blood stream has been known since the 19th century. Circulating tumor cells (CTCs) may be detected in peripheral blood of patients with breast cancer and may serve as a surrogate marker for minimal residual disease. Prognostic relevance of CTCs has already been demonstrated in early and metastatic breast cancer and commercially available detection systems are currently employed in various clinical trials. Since peripheral blood is an easily accessible compartment, serial reevaluation of CTCs is possible and may contribute to better therapy monitoring. Another potential of CTCs lies in the characterization of tumor cells. Expression profiles may differ between CTCs and primary tumor, which may result in different responses to treatment. Assessment of molecular features of CTCs may be an important step for the optimization of adjuvant and metastatic systemic therapy.
Collapse
|
63
|
Vlaeminck-Guillem V. When Prostate Cancer Circulates in the Bloodstream. Diagnostics (Basel) 2015; 5:428-74. [PMID: 26854164 PMCID: PMC4728468 DOI: 10.3390/diagnostics5040428] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 10/14/2015] [Accepted: 10/22/2015] [Indexed: 12/14/2022] Open
Abstract
Management of patients with prostate cancer is currently based on imperfect clinical, biological, radiological and pathological evaluation. Prostate cancer aggressiveness, including metastatic potential, remains difficult to accurately estimate. In an attempt to better adapt therapeutics to an individual (personalized medicine), reliable evaluation of the intrinsic molecular biology of the tumor is warranted, and particularly for all tumor sites (primary tumors and secondary sites) at any time of the disease progression. As a consequence of their natural tendency to grow (passive invasion) or as a consequence of an active blood vessel invasion by metastase-initiating cells, tumors shed various materials into the bloodstream. Major efforts have been recently made to develop powerful and accurate methods able to detect, quantify and/or analyze all these circulating tumor materials: circulating tumors cells, disseminating tumor cells, extracellular vesicles (including exosomes), nucleic acids, etc. The aim of this review is to summarize current knowledge about these circulating tumor materials and their applications in translational research.
Collapse
Affiliation(s)
- Virginie Vlaeminck-Guillem
- Cancer Research Centre of Lyon, U1052 INSERM, CNRS 5286, Léon Bérard Centre, Lyon I University, 28 rue Laennec, Lyon 69008, France.
- Medical Unit of Molecular Oncology and Transfer, Department of Biochemistry and Molecular Biology, University Hospital of Lyon-Sud, Hospices Civils of Lyon, Lyon 69008, France.
| |
Collapse
|
64
|
Jokerst JV, Chen Z, Xu L, Nolley R, Chang E, Mitchell B, Brooks JD, Gambhir SS. A Magnetic Bead-Based Sensor for the Quantification of Multiple Prostate Cancer Biomarkers. PLoS One 2015; 10:e0139484. [PMID: 26421725 PMCID: PMC4589536 DOI: 10.1371/journal.pone.0139484] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 09/13/2015] [Indexed: 12/20/2022] Open
Abstract
Novel biomarker assays and upgraded analytical tools are urgently needed to accurately discriminate benign prostatic hypertrophy (BPH) from prostate cancer (CaP). To address this unmet clinical need, we report a piezeoelectric/magnetic bead-based assay to quantitate prostate specific antigen (PSA; free and total), prostatic acid phosphatase, carbonic anhydrase 1 (CA1), osteonectin, IL-6 soluble receptor (IL-6sr), and spondin-2. We used the sensor to measure these seven proteins in serum samples from 120 benign prostate hypertrophy patients and 100 Gleason score 6 and 7 CaP using serum samples previously collected and banked. The results were analyzed with receiver operator characteristic curve analysis. There were significant differences between BPH and CaP patients in the PSA, CA1, and spondin-2 assays. The highest AUC discrimination was achieved with a spondin-2 OR free/total PSA operation—the area under the curve was 0.84 with a p value below 10−6. Some of these data seem to contradict previous reports and highlight the importance of sample selection and proper assay building in the development of biomarker measurement schemes. This bead-based system offers important advantages in assay building including low cost, high throughput, and rapid identification of an optimal matched antibody pair.
Collapse
Affiliation(s)
- Jesse V. Jokerst
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California, United States of America
| | - Zuxiong Chen
- Department of Urology, Stanford University, Stanford, California, United States of America
| | - Lingyun Xu
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California, United States of America
| | - Rosalie Nolley
- Department of Urology, Stanford University, Stanford, California, United States of America
| | - Edwin Chang
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California, United States of America
| | - Breeana Mitchell
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California, United States of America
| | - James D. Brooks
- Department of Urology, Stanford University, Stanford, California, United States of America
- * E-mail: (JDB); (SSG)
| | - Sanjiv S. Gambhir
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California, United States of America
- Bioengineering, Materials Science & Engineering, Bio-X, Stanford University, Stanford, California, United States of America
- * E-mail: (JDB); (SSG)
| |
Collapse
|
65
|
Azevedo AS, Follain G, Patthabhiraman S, Harlepp S, Goetz JG. Metastasis of circulating tumor cells: favorable soil or suitable biomechanics, or both? Cell Adh Migr 2015; 9:345-56. [PMID: 26312653 DOI: 10.1080/19336918.2015.1059563] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Metastasis is the end product of a multistep process where cancer cells disseminate and home themselves in distant organs. Tumor cell extravasation is a rare, inefficient and transient event in nature and makes its studies very difficult. Noteworthy, little is known about how cancer cells arrest, adhere and pass through the endothelium of capillaries. Moreover, the key events driving metastatic growth in specific organs are not well understood. Thus, although metastasis is the leading cause of cancer-related death, how cancer cells acquire their abilities to colonize distant organs and why they do so in specific locations remain central questions in the understanding of this deadly disease. In this review, we would like to confront 2 concepts explaining the efficiency and location of metastatic secondary tumors. While the "seed and soil" hypothesis states that metastasis occurs at sites where the local microenvironment is favorable, the "mechanical" concept argues that metastatic seeding occurs at sites of optimal flow patterns. In addition, recent evidence suggests that the primary event driving tumor cell arrest before extravasation is mostly controlled by blood circulation patterns as well as mechanical cues during the process of extravasation. In conclusion, the organ tropism displayed by cancer cells during metastatic colonization is a multi-step process, which is regulated by the delivery and survival of circulating tumor cells (CTCs) through blood circulation, the ability of these CTCs to adhere and cross the physical barrier imposed by the endothelium and finally by the suitability of the soil to favor growth of secondary tumors.
Collapse
Affiliation(s)
- Ana Sofia Azevedo
- a Inserm U1109; MN3T ; Strasbourg , France.,b Université de Strasbourg ; Strasbourg , France.,c LabEx Medalis; Université de Strasbourg ; Strasbourg , France.,d Fédération de Médecine Translationnelle de Strasbourg (FMTS) ; Strasbourg , France
| | - Gautier Follain
- a Inserm U1109; MN3T ; Strasbourg , France.,b Université de Strasbourg ; Strasbourg , France.,c LabEx Medalis; Université de Strasbourg ; Strasbourg , France.,d Fédération de Médecine Translationnelle de Strasbourg (FMTS) ; Strasbourg , France
| | - Shankar Patthabhiraman
- a Inserm U1109; MN3T ; Strasbourg , France.,b Université de Strasbourg ; Strasbourg , France.,c LabEx Medalis; Université de Strasbourg ; Strasbourg , France.,d Fédération de Médecine Translationnelle de Strasbourg (FMTS) ; Strasbourg , France
| | - Sébastien Harlepp
- b Université de Strasbourg ; Strasbourg , France.,e IPCMS UMR7504 ; Strasbourg , France.,f LabEx NIE; Université de Strasbourg ; Strasbourg , France
| | - Jacky G Goetz
- a Inserm U1109; MN3T ; Strasbourg , France.,b Université de Strasbourg ; Strasbourg , France.,c LabEx Medalis; Université de Strasbourg ; Strasbourg , France.,d Fédération de Médecine Translationnelle de Strasbourg (FMTS) ; Strasbourg , France
| |
Collapse
|
66
|
Morimoto A, Mogami T, Watanabe M, Iijima K, Akiyama Y, Katayama K, Futami T, Yamamoto N, Sawada T, Koizumi F, Koh Y. High-Density Dielectrophoretic Microwell Array for Detection, Capture, and Single-Cell Analysis of Rare Tumor Cells in Peripheral Blood. PLoS One 2015; 10:e0130418. [PMID: 26107884 PMCID: PMC4480363 DOI: 10.1371/journal.pone.0130418] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 05/20/2015] [Indexed: 12/17/2022] Open
Abstract
Development of a reliable platform and workflow to detect and capture a small number of mutation-bearing circulating tumor cells (CTCs) from a blood sample is necessary for the development of noninvasive cancer diagnosis. In this preclinical study, we aimed to develop a capture system for molecular characterization of single CTCs based on high-density dielectrophoretic microwell array technology. Spike-in experiments using lung cancer cell lines were conducted. The microwell array was used to capture spiked cancer cells, and captured single cells were subjected to whole genome amplification followed by sequencing. A high detection rate (70.2%–90.0%) and excellent linear performance (R2 = 0.8189–0.9999) were noted between the observed and expected numbers of tumor cells. The detection rate was markedly higher than that obtained using the CellSearch system in a blinded manner, suggesting the superior sensitivity of our system in detecting EpCAM− tumor cells. Isolation of single captured tumor cells, followed by detection of EGFR mutations, was achieved using Sanger sequencing. Using a microwell array, we established an efficient and convenient platform for the capture and characterization of single CTCs. The results of a proof-of-principle preclinical study indicated that this platform has potential for the molecular characterization of captured CTCs from patients.
Collapse
Affiliation(s)
- Atsushi Morimoto
- Life Science Research Laboratory, Tosoh Corporation, Kanagawa, Japan
| | - Toshifumi Mogami
- Life Science Research Laboratory, Tosoh Corporation, Kanagawa, Japan
| | - Masaru Watanabe
- Third Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan
| | - Kazuki Iijima
- Life Science Research Laboratory, Tosoh Corporation, Kanagawa, Japan
| | - Yasuyuki Akiyama
- Life Science Research Laboratory, Tosoh Corporation, Kanagawa, Japan
| | - Koji Katayama
- Life Science Research Laboratory, Tosoh Corporation, Kanagawa, Japan
| | - Toru Futami
- Life Science Research Laboratory, Tosoh Corporation, Kanagawa, Japan
| | - Nobuyuki Yamamoto
- Third Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan
| | - Takeshi Sawada
- Shien-Lab, National Cancer Center Hospital, Tokyo, Japan
| | | | - Yasuhiro Koh
- Third Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
67
|
Li J, Gregory SG, Garcia-Blanco MA, Armstrong AJ. Using circulating tumor cells to inform on prostate cancer biology and clinical utility. Crit Rev Clin Lab Sci 2015; 52:191-210. [PMID: 26079252 DOI: 10.3109/10408363.2015.1023430] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Substantial advances in the molecular biology of prostate cancer have led to the approval of multiple new systemic agents to treat men with metastatic castration-resistant prostate cancer (mCRPC). These treatments encompass androgen receptor directed therapies, immunotherapies, bone targeting radiopharmaceuticals and cytotoxic chemotherapies. There is, however, great heterogeneity in the degree of patient benefit with these agents, thus fueling the need to develop predictive biomarkers that are able to rationally guide therapy. Circulating tumor cells (CTCs) have the potential to provide an assessment of tumor-specific biomarkers through a non-invasive, repeatable "liquid biopsy" of a patient's cancer at a given point in time. CTCs have been extensively studied in men with mCRPC, where CTC enumeration using the Cellsearch® method has been validated and FDA approved to be used in conjunction with other clinical parameters as a prognostic biomarker in metastatic prostate cancer. In addition to enumeration, more sophisticated molecular profiling of CTCs is now feasible and may provide more clinical utility as it may reflect tumor evolution within an individual particularly under the pressure of systemic therapies. Here, we review technologies used to detect and characterize CTCs, and the potential biological and clinical utility of CTC molecular profiling in men with metastatic prostate cancer.
Collapse
Affiliation(s)
- Jing Li
- a Duke Cancer Institute, Duke University Medical Center , Durham , NC , USA
| | | | | | | |
Collapse
|
68
|
Abstract
Molecular characterization of circulating tumor cells (CTCs) found in the blood of cancer patients offers the potential to provide new insights into the biology of cancer metastasis. However, since they are rare and difficult to isolate, the molecular nature of CTCs remains poorly understood. In this paper, we reviewed a decade's worth of scientific literature (2003-2013) describing efforts on isolation and genomic analysis of CTCs. The limited number of CTC genomic studies we found attested to the infancy of this field of study. These initial reports, however, provide an important framework for future comprehensive exploration of CTC biology. For CTCs to be broadly accepted as therapeutic targets and biomarkers of metastatic spread, further in-depth molecular characterization is warranted.
Collapse
|
69
|
Yap TA, Lorente D, Omlin A, Olmos D, de Bono JS. Circulating tumor cells: a multifunctional biomarker. Clin Cancer Res 2015; 20:2553-68. [PMID: 24831278 DOI: 10.1158/1078-0432.ccr-13-2664] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
One of the most promising developments in translational cancer medicine has been the emergence of circulating tumor cells (CTC) as a minimally invasive multifunctional biomarker. CTCs in peripheral blood originate from solid tumors and are involved in the process of hematogenous metastatic spread to distant sites for the establishment of secondary foci of disease. The emergence of modern CTC technologies has enabled serial assessments to be undertaken at multiple time points along a patient's cancer journey for pharmacodynamic (PD), prognostic, predictive, and intermediate endpoint biomarker studies. Despite the promise of CTCs as multifunctional biomarkers, there are still numerous challenges that hinder their incorporation into standard clinical practice. This review discusses the key technical aspects of CTC technologies, including the importance of assay validation and clinical qualification, and compares existing and novel CTC enrichment platforms. This article discusses the utility of CTCs as a multifunctional biomarker and focuses on the potential of CTCs as PD endpoints either directly via the molecular characterization of specific markers or indirectly through CTC enumeration. We propose strategies for incorporating CTCs as PD biomarkers in translational clinical trials, such as the Pharmacological Audit Trail. We also discuss issues relating to intrapatient heterogeneity and the challenges associated with isolating CTCs undergoing epithelial-mesenchymal transition, as well as apoptotic and small CTCs. Finally, we envision the future promise of CTCs for the selection and monitoring of antitumor precision therapies, including applications in single CTC phenotypic and genomic profiling and CTC-derived xenografts, and discuss the promises and limitations of such approaches. See ALL articles in this CCR focus section, "Progress in pharmacodynamic endpoints."
Collapse
Affiliation(s)
- Timothy A Yap
- Authors' Affiliations: Division of Clinical Studies, The Institute of Cancer Research; Drug Development Unit, Royal Marsden NHS Foundation Trust, Sutton, Surrey, United Kingdom; Kantonsspital St. Gallen, Department of Medical Oncology, Gallen, Switzerland; and Spanish National Cancer Research Centre, Madrid, SpainAuthors' Affiliations: Division of Clinical Studies, The Institute of Cancer Research; Drug Development Unit, Royal Marsden NHS Foundation Trust, Sutton, Surrey, United Kingdom; Kantonsspital St. Gallen, Department of Medical Oncology, Gallen, Switzerland; and Spanish National Cancer Research Centre, Madrid, Spain
| | - David Lorente
- Authors' Affiliations: Division of Clinical Studies, The Institute of Cancer Research; Drug Development Unit, Royal Marsden NHS Foundation Trust, Sutton, Surrey, United Kingdom; Kantonsspital St. Gallen, Department of Medical Oncology, Gallen, Switzerland; and Spanish National Cancer Research Centre, Madrid, SpainAuthors' Affiliations: Division of Clinical Studies, The Institute of Cancer Research; Drug Development Unit, Royal Marsden NHS Foundation Trust, Sutton, Surrey, United Kingdom; Kantonsspital St. Gallen, Department of Medical Oncology, Gallen, Switzerland; and Spanish National Cancer Research Centre, Madrid, Spain
| | - Aurelius Omlin
- Authors' Affiliations: Division of Clinical Studies, The Institute of Cancer Research; Drug Development Unit, Royal Marsden NHS Foundation Trust, Sutton, Surrey, United Kingdom; Kantonsspital St. Gallen, Department of Medical Oncology, Gallen, Switzerland; and Spanish National Cancer Research Centre, Madrid, Spain
| | - David Olmos
- Authors' Affiliations: Division of Clinical Studies, The Institute of Cancer Research; Drug Development Unit, Royal Marsden NHS Foundation Trust, Sutton, Surrey, United Kingdom; Kantonsspital St. Gallen, Department of Medical Oncology, Gallen, Switzerland; and Spanish National Cancer Research Centre, Madrid, Spain
| | - Johann S de Bono
- Authors' Affiliations: Division of Clinical Studies, The Institute of Cancer Research; Drug Development Unit, Royal Marsden NHS Foundation Trust, Sutton, Surrey, United Kingdom; Kantonsspital St. Gallen, Department of Medical Oncology, Gallen, Switzerland; and Spanish National Cancer Research Centre, Madrid, SpainAuthors' Affiliations: Division of Clinical Studies, The Institute of Cancer Research; Drug Development Unit, Royal Marsden NHS Foundation Trust, Sutton, Surrey, United Kingdom; Kantonsspital St. Gallen, Department of Medical Oncology, Gallen, Switzerland; and Spanish National Cancer Research Centre, Madrid, Spain
| |
Collapse
|
70
|
Smith JP, Huang C, Kirby BJ. Enhancing sensitivity and specificity in rare cell capture microdevices with dielectrophoresis. BIOMICROFLUIDICS 2015; 9:014116. [PMID: 25759749 PMCID: PMC4327920 DOI: 10.1063/1.4908049] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 02/02/2015] [Indexed: 05/11/2023]
Abstract
The capture and subsequent analysis of rare cells, such as circulating tumor cells from a peripheral blood sample, has the potential to advance our understanding and treatment of a wide range of diseases. There is a particular need for high purity (i.e., high specificity) techniques to isolate these cells, reducing the time and cost required for single-cell genetic analyses by decreasing the number of contaminating cells analyzed. Previous work has shown that antibody-based immunocapture can be combined with dielectrophoresis (DEP) to differentially isolate cancer cells from leukocytes in a characterization device. Here, we build on that work by developing numerical simulations that identify microfluidic obstacle array geometries where DEP-immunocapture can be used to maximize the capture of target rare cells, while minimizing the capture of contaminating cells. We consider geometries with electrodes offset from the array and parallel to the fluid flow, maximizing the magnitude of the resulting electric field at the obstacles' leading and trailing edges, and minimizing it at the obstacles' shoulders. This configuration attracts cells with a positive DEP (pDEP) response to the leading edge, where the shear stress is low and residence time is long, resulting in a high capture probability; although these cells are also repelled from the shoulder region, the high local fluid velocity at the shoulder minimizes the impact on the overall transport and capture. Likewise, cells undergoing negative DEP (nDEP) are repelled from regions of high capture probability and attracted to regions where capture is unlikely. These simulations predict that DEP can be used to reduce the probability of capturing contaminating peripheral blood mononuclear cells (using nDEP) from 0.16 to 0.01 while simultaneously increasing the capture of several pancreatic cancer cell lines from 0.03-0.10 to 0.14-0.55, laying the groundwork for the experimental study of hybrid DEP-immunocapture obstacle array microdevices.
Collapse
Affiliation(s)
- James P Smith
- Sibley School of Mechanical and Aerospace Engineering, Cornell University , Ithaca, New York 14853, USA
| | - Chao Huang
- Department of Biomedical Engineering, Cornell University , Ithaca, New York 14853, USA
| | | |
Collapse
|
71
|
Fina E, Callari M, Reduzzi C, D'Aiuto F, Mariani G, Generali D, Pierotti MA, Daidone MG, Cappelletti V. Gene expression profiling of circulating tumor cells in breast cancer. Clin Chem 2014; 61:278-89. [PMID: 25411184 DOI: 10.1373/clinchem.2014.229476] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Determining the transcriptional profile of circulating tumor cells (CTCs) may allow the acquisition of clinically relevant information while overcoming tumor heterogeneity-related biases associated with use of tissue samples for biomarker assessment. However, such molecular characterization is challenging because CTCs are rare and outnumbered by blood cells. METHODS Here, we describe a technical protocol to measure the expression of >29 000 genes in CTCs captured from whole blood with magnetic beads linked with antibodies against epithelial cell adhesion molecule (EpCAM) and the carcinoma-associated mucin, MUC1, designed to be used for CTC characterization in clinical samples. Low numbers of cells (5-200) from the MCF7 and MDA-MB-468 breast cancer cell lines were spiked in healthy donor blood samples and isolated with the AdnaTest EMT-1/Stem CellSelect kit. Gene expression profiles (GEPs) were obtained with the WG-DASL HT assay and compared with GEPs obtained from RNA isolated from cultured cell lines and unspiked samples. RESULTS GEPs from samples containing 25 or more spiked cells correlated (r = 0.95) with cognate 100-ng RNA input samples, clustered separately from blood control samples, and allowed MCF7 and MDA-MB-468 cells to be distinguished. GEPs with comparable technical quality were also obtained in a preliminary series of clinical samples. CONCLUSIONS Our approach allows technically reliable GEPs to be obtained from isolated CTCs for the acquisition of biologically useful information. It is reproducible and suitable for application in prospective studies to assess the clinical utility of CTC GEPs, provided that >25 CTCs can be isolated.
Collapse
Affiliation(s)
- Emanuela Fina
- Biomarkers Unit, Department of Experimental Oncology and Molecular Medicine
| | - Maurizio Callari
- Biomarkers Unit, Department of Experimental Oncology and Molecular Medicine
| | - Carolina Reduzzi
- Biomarkers Unit, Department of Experimental Oncology and Molecular Medicine
| | - Francesca D'Aiuto
- Biomarkers Unit, Department of Experimental Oncology and Molecular Medicine
| | | | - Daniele Generali
- U.O. Multidisciplinare di Patologia Mammaria, U.S. Terapia Molecolare e Farmacogenomica, A.O. Istituti Ospitalieri di Cremona, Cremona, Italy
| | - Marco A Pierotti
- Scientific Directorate, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maria G Daidone
- Biomarkers Unit, Department of Experimental Oncology and Molecular Medicine,
| | - Vera Cappelletti
- Biomarkers Unit, Department of Experimental Oncology and Molecular Medicine
| |
Collapse
|
72
|
Single-cell analyses of transcriptional heterogeneity during drug tolerance transition in cancer cells by RNA sequencing. Proc Natl Acad Sci U S A 2014; 111:E4726-35. [PMID: 25339441 DOI: 10.1073/pnas.1404656111] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The acute cellular response to stress generates a subpopulation of reversibly stress-tolerant cells under conditions that are lethal to the majority of the population. Stress tolerance is attributed to heterogeneity of gene expression within the population to ensure survival of a minority. We performed whole transcriptome sequencing analyses of metastatic human breast cancer cells subjected to the chemotherapeutic agent paclitaxel at the single-cell and population levels. Here we show that specific transcriptional programs are enacted within untreated, stressed, and drug-tolerant cell groups while generating high heterogeneity between single cells within and between groups. We further demonstrate that drug-tolerant cells contain specific RNA variants residing in genes involved in microtubule organization and stabilization, as well as cell adhesion and cell surface signaling. In addition, the gene expression profile of drug-tolerant cells is similar to that of untreated cells within a few doublings. Thus, single-cell analyses reveal the dynamics of the stress response in terms of cell-specific RNA variants driving heterogeneity, the survival of a minority population through generation of specific RNA variants, and the efficient reconversion of stress-tolerant cells back to normalcy.
Collapse
|
73
|
Single-cell sequencing technologies: current and future. J Genet Genomics 2014; 41:513-28. [PMID: 25438696 DOI: 10.1016/j.jgg.2014.09.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 09/01/2014] [Accepted: 09/16/2014] [Indexed: 12/15/2022]
Abstract
Intensively developed in the last few years, single-cell sequencing technologies now present numerous advantages over traditional sequencing methods for solving the problems of biological heterogeneity and low quantities of available biological materials. The application of single-cell sequencing technologies has profoundly changed our understanding of a series of biological phenomena, including gene transcription, embryo development, and carcinogenesis. However, before single-cell sequencing technologies can be used extensively, researchers face the serious challenge of overcoming inherent issues of high amplification bias, low accuracy and reproducibility. Here, we simply summarize the techniques used for single-cell isolation, and review the current technologies used in single-cell genomic, transcriptomic, and epigenomic sequencing. We discuss the merits, defects, and scope of application of single-cell sequencing technologies and then speculate on the direction of future developments.
Collapse
|
74
|
Single-cell RNA sequencing identifies extracellular matrix gene expression by pancreatic circulating tumor cells. Cell Rep 2014. [PMID: 25242334 DOI: 10.1016/j.celrep.2014.08.029.single-cell] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Circulating tumor cells (CTCs) are shed from primary tumors into the bloodstream, mediating the hematogenous spread of cancer to distant organs. To define their composition, we compared genome-wide expression profiles of CTCs with matched primary tumors in a mouse model of pancreatic cancer, isolating individual CTCs using epitope-independent microfluidic capture, followed by single-cell RNA sequencing. CTCs clustered separately from primary tumors and tumor-derived cell lines, showing low-proliferative signatures, enrichment for the stem-cell-associated gene Aldh1a2, biphenotypic expression of epithelial and mesenchymal markers, and expression of Igfbp5, a gene transcript enriched at the epithelial-stromal interface. Mouse as well as human pancreatic CTCs exhibit a very high expression of stromal-derived extracellular matrix (ECM) proteins, including SPARC, whose knockdown in cancer cells suppresses cell migration and invasiveness. The aberrant expression by CTCs of stromal ECM genes points to their contribution of microenvironmental signals for the spread of cancer to distant organs.
Collapse
|
75
|
Ting DT, Wittner BS, Ligorio M, Vincent Jordan N, Shah AM, Miyamoto DT, Aceto N, Bersani F, Brannigan BW, Xega K, Ciciliano JC, Zhu H, MacKenzie OC, Trautwein J, Arora KS, Shahid M, Ellis HL, Qu N, Bardeesy N, Rivera MN, Deshpande V, Ferrone CR, Kapur R, Ramaswamy S, Shioda T, Toner M, Maheswaran S, Haber DA. Single-cell RNA sequencing identifies extracellular matrix gene expression by pancreatic circulating tumor cells. Cell Rep 2014; 8:1905-1918. [PMID: 25242334 PMCID: PMC4230325 DOI: 10.1016/j.celrep.2014.08.029] [Citation(s) in RCA: 372] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 07/16/2014] [Accepted: 08/13/2014] [Indexed: 12/27/2022] Open
Abstract
Circulating tumor cells (CTCs) are shed from primary tumors into the bloodstream, mediating the hematogenous spread of cancer to distant organs. To define their composition, we compared genome-wide expression profiles of CTCs with matched primary tumors in a mouse model of pancreatic cancer, isolating individual CTCs using epitope-independent microfluidic capture, followed by single-cell RNA sequencing. CTCs clustered separately from primary tumors and tumor-derived cell lines, showing low-proliferative signatures, enrichment for the stem-cell-associated gene Aldh1a2, biphenotypic expression of epithelial and mesenchymal markers, and expression of Igfbp5, a gene transcript enriched at the epithelial-stromal interface. Mouse as well as human pancreatic CTCs exhibit a very high expression of stromal-derived extracellular matrix (ECM) proteins, including SPARC, whose knockdown in cancer cells suppresses cell migration and invasiveness. The aberrant expression by CTCs of stromal ECM genes points to their contribution of microenvironmental signals for the spread of cancer to distant organs.
Collapse
Affiliation(s)
- David T Ting
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Ben S Wittner
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Matteo Ligorio
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Surgery, Harvard Medical School, Boston, MA 02114, USA; Department of Health Sciences, University of Genoa, 16126 Genoa, Italy
| | - Nicole Vincent Jordan
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Ajay M Shah
- Center for Engineering in Medicine, Harvard Medical School, Boston, MA 02114, USA; Department of Surgery, Harvard Medical School, Boston, MA 02114, USA
| | - David T Miyamoto
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Radiation Oncology, Harvard Medical School, Boston, MA 02114, USA
| | - Nicola Aceto
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Francesca Bersani
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Brian W Brannigan
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Kristina Xega
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Jordan C Ciciliano
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Huili Zhu
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Olivia C MacKenzie
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Julie Trautwein
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Kshitij S Arora
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Surgery, Harvard Medical School, Boston, MA 02114, USA; Department of Pathology, Harvard Medical School, Boston, MA 02114, USA
| | - Mohammad Shahid
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Surgery, Harvard Medical School, Boston, MA 02114, USA; Department of Pathology, Harvard Medical School, Boston, MA 02114, USA
| | - Haley L Ellis
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Na Qu
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Nabeel Bardeesy
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Miguel N Rivera
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Pathology, Harvard Medical School, Boston, MA 02114, USA
| | - Vikram Deshpande
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Pathology, Harvard Medical School, Boston, MA 02114, USA
| | - Cristina R Ferrone
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Surgery, Harvard Medical School, Boston, MA 02114, USA
| | - Ravi Kapur
- Center for Engineering in Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Sridhar Ramaswamy
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Toshi Shioda
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Mehmet Toner
- Center for Engineering in Medicine, Harvard Medical School, Boston, MA 02114, USA; Department of Surgery, Harvard Medical School, Boston, MA 02114, USA
| | - Shyamala Maheswaran
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Surgery, Harvard Medical School, Boston, MA 02114, USA.
| | - Daniel A Haber
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
76
|
Shtivelman E, Beer TM, Evans CP. Molecular pathways and targets in prostate cancer. Oncotarget 2014; 5:7217-59. [PMID: 25277175 PMCID: PMC4202120 DOI: 10.18632/oncotarget.2406] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 08/28/2014] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer co-opts a unique set of cellular pathways in its initiation and progression. The heterogeneity of prostate cancers is evident at earlier stages, and has led to rigorous efforts to stratify the localized prostate cancers, so that progression to advanced stages could be predicted based upon salient features of the early disease. The deregulated androgen receptor signaling is undeniably most important in the progression of the majority of prostate tumors. It is perhaps because of the primacy of the androgen receptor governed transcriptional program in prostate epithelium cells that once this program is corrupted, the consequences of the ensuing changes in activity are pleotropic and could contribute to malignancy in multiple ways. Following localized surgical and radiation therapies, 20-40% of patients will relapse and progress, and will be treated with androgen deprivation therapies. The successful development of the new agents that inhibit androgen signaling has changed the progression free survival in hormone resistant disease, but this has not changed the almost ubiquitous development of truly resistant phenotypes in advanced prostate cancer. This review summarizes the current understanding of the molecular pathways involved in localized and metastatic prostate cancer, with an emphasis on the clinical implications of the new knowledge.
Collapse
Affiliation(s)
| | - Tomasz M. Beer
- Oregon Health & Science University, Knight Cancer Institute, Portland, OR
| | - Christopher P. Evans
- Department of Urology and Comprehensive Cancer Center, University of California Davis, Davis, CA
| |
Collapse
|
77
|
Clinical utility of circulating tumour cell detection in non-small-cell lung cancer. Curr Treat Options Oncol 2014; 14:610-22. [PMID: 23996475 DOI: 10.1007/s11864-013-0253-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OPINION STATEMENT Recent years have witnessed increased interest in the detection of circulating tumour cells (CTCs) for diagnosis, monitoring, and treatment decision making in patients with cancer. Factors that have led to accelerated research in this field include advances in technologies for examination of intact CTCs, personalised medicine with treatment selection according to molecular characteristics, and continued lack of understanding of the biology of treatment resistance and metastasis. CTCs offer promise as a surrogate for tissue where there is insufficient tissue for molecular analysis and where there is a requirement to serially monitor molecular changes in cancer cells through treatment or on progression. In patients with either small cell or non-small cell lung cancer (NSCLC), there is evidence that CTC number is prognostic and that CTCs counted before and after treatment mirror treatment response. In patients with molecularly defined subtypes of NSCLC, CTCs demonstrate the same molecular changes as the cancer cells of the tumour. However, CTCs are not quite ready for "primetime" in the lung cancer clinic. There are still more questions than answers with respect to the optimal technologies for their detection and analysis, their biological significance, and their clinical utility. Despite this the current pace of progress in CTC technology development seems set to make "liquid biopsies" a clinical reality within the next decade. For the everyday clinician and clinical trialist, it will be important to maintain knowledge of the strengths and weaknesses of the technologies and evolving evidence base for CTCs as a routinely used diagnostic tool.
Collapse
|
78
|
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: 3.6] [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
|
79
|
Liu N, Liu L, Pan X. Single-cell analysis of the transcriptome and its application in the characterization of stem cells and early embryos. Cell Mol Life Sci 2014; 71:2707-15. [PMID: 24652479 PMCID: PMC11113295 DOI: 10.1007/s00018-014-1601-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 01/16/2014] [Accepted: 02/03/2014] [Indexed: 12/17/2022]
Abstract
Cellular heterogeneity within a cell population is a common phenomenon in multicellular organisms, tissues, cultured cells, and even FACS-sorted subpopulations. Important information may be masked if the cells are studied as a mass. Transcriptome profiling is a parameter that has been intensively studied, and relatively easier to address than protein composition. To understand the basis and importance of heterogeneity and stochastic aspects of the cell function and its mechanisms, it is essential to examine transcriptomes of a panel of single cells. High-throughput technologies, starting from microarrays and now RNA-seq, provide a full view of the expression of transcriptomes but are limited by the amount of RNA for analysis. Recently, several new approaches for amplification and sequencing the transcriptome of single cells or a limited low number of cells have been developed and applied. In this review, we summarize these major strategies, such as PCR-based methods, IVT-based methods, phi29-DNA polymerase-based methods, and several other methods, including their principles, characteristics, advantages, and limitations, with representative applications in cancer stem cells, early development, and embryonic stem cells. The prospects for development of future technology and application of transcriptome analysis in a single cell are also discussed.
Collapse
Affiliation(s)
- Na Liu
- State Key Laboratory of Medicinal Chemical Biology, Department of Cell Biology and Genetics, College of Life Science, Nankai University, Tianjin, 300071, China,
| | | | | |
Collapse
|
80
|
Huang C, Smith JP, Saha TN, Rhim AD, Kirby BJ. Characterization of microfluidic shear-dependent epithelial cell adhesion molecule immunocapture and enrichment of pancreatic cancer cells from blood cells with dielectrophoresis. BIOMICROFLUIDICS 2014; 8:044107. [PMID: 25379092 PMCID: PMC4189216 DOI: 10.1063/1.4890466] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 07/07/2014] [Indexed: 05/02/2023]
Abstract
Current microfluidic techniques for isolating circulating tumor cells (CTCs) from cancer patient blood are limited by low capture purity, and dielectrophoresis (DEP) has the potential to complement existing immunocapture techniques to improve capture performance. We present a hybrid DEP and immunocapture Hele-Shaw flow cell to characterize DEP's effects on immunocapture of pancreatic cancer cells (Capan-1, PANC-1, and BxPC-3) and peripheral blood mononuclear cells (PBMCs) with an anti-EpCAM (epithelial cell adhesion molecule) antibody. By carefully specifying the applied electric field frequency, we demonstrate that pancreatic cancer cells are attracted to immunocapture surfaces by positive DEP whereas PBMCs are repelled by negative DEP. Using an exponential capture model to interpret our capture data, we show that immunocapture performance is dependent on the applied DEP force sign and magnitude, cell surface EpCAM expression level, and shear stress experienced by cells flowing in the capture device. Our work suggests that DEP can not only repel contaminating blood cells but also enhance capture of cancer cell populations that are less likely to be captured by traditional immunocapture methods. This combination of DEP and immunocapture techniques to potentially increase CTC capture purity can facilitate subsequent biological analyses of captured CTCs and research on cancer metastasis and drug therapies.
Collapse
Affiliation(s)
- Chao Huang
- Department of Biomedical Engineering, Cornell University , Ithaca, New York 14853, USA
| | - James P Smith
- Sibley School of Mechanical and Aerospace Engineering, Cornell University , Ithaca, New York 14853, USA
| | - Trisha N Saha
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School , Ann Arbor, Michigan 48109, USA
| | - Andrew D Rhim
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School , Ann Arbor, Michigan 48109, USA
| | | |
Collapse
|
81
|
Chan CML, Au TCC, Chan ATC, Ma BBY, Tsui NBY, Ng SSM, Hui EP, Chan LWC, Ho WS, Yung BYM, Wong SCC. Advanced technologies for studying circulating tumor cells at the protein level. Expert Rev Proteomics 2014; 10:579-89. [PMID: 24206230 DOI: 10.1586/14789450.2013.858021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Metastasis is the main cause of cancer death. As the tumor progresses, cells from the primary tumor site are shed into the bloodstream as circulating tumor cells (CTCs). Eventually, these cells colonize other organs and form distant metastases. It is therefore imperative that we gain a better understanding of the biological characteristics of CTCs for development of novel treatment modalities to minimize metastasis-associated cancer deaths. In recent years, rapid developments in technologies for the study of CTCs have taken place. We now have a variety of tools for the isolation and examination of CTCs which were not available before. This review introduces some commonly used protein markers in CTC investigations and summarizes a few advanced technologies which have been successfully applied for studying CTC biology at the protein level.
Collapse
Affiliation(s)
- Charles Ming Lok Chan
- Department of Clinical Oncology, State Key Laboratory in Oncology in South China, Sir Y K Pao Centre for Cancer, Hong Kong Cancer Institute and Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Watanabe M, Serizawa M, Sawada T, Takeda K, Takahashi T, Yamamoto N, Koizumi F, Koh Y. A novel flow cytometry-based cell capture platform for the detection, capture and molecular characterization of rare tumor cells in blood. J Transl Med 2014; 12:143. [PMID: 24886394 PMCID: PMC4053587 DOI: 10.1186/1479-5876-12-143] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 05/12/2014] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Personalized cancer treatment relies on the accurate detection of actionable genomic aberrations in tumor cells. Circulating tumor cells (CTCs) could provide an alternative genetic resource for diagnosis; however, the technical difficulties in isolating and analyzing rare CTCs have limited progress to date. In this preclinical study, we aimed to develop an improved capture system for molecular characterization of CTCs based on a novel cell sorting technology. METHODS We developed a cell capture platform using On-chip Sort (On-Chip Biotechnologies), a novel bench-top cell sorter equipped with a disposable microfluidic chip. Spike-in experiments comprising a series of lung cancer cell lines with varying epithelial cell adhesion molecule (EpCAM) expression levels were conducted to assess the capture and purification efficiency of the platform. Samples were negatively enriched using anti-CD45-coated magnetic beads to remove white blood cells, followed by sample fixation and labeling. The enriched and labeled samples were then sorted by On-chip Sort based on cytokeratin, vimentin, and CD45 expression. Captured cells were immediately subjected to whole genome amplification followed by mutation analysis using deep targeted sequencing, and copy number analysis using quantitative polymerase chain reaction (qPCR). RESULTS Spike-in experiments revealed an excellent overall mean capture rate of 70.9%. A 100% success rate in the detection of EGFR, KRAS and BRAF mutations from captured cells was achieved using pyrosequencing and deep sequencing. The mutant variant detection rates were markedly higher than those obtained with the CellSearch profile kit. qPCR analysis of amplified DNA demonstrated reproducible detection of copy number changes of the EGFR in captured tumor cells. CONCLUSIONS Using a novel cell sorter, we established an efficient and convenient platform for the capture of CTCs. Results of a proof-of-principle preclinical study indicated that this platform has potential for the molecular characterization of captured CTCs from patients.
Collapse
Affiliation(s)
- Masaru Watanabe
- Drug Discovery and Development Division, Shizuoka Cancer Center Research Institute, Shizuoka, Japan
- Third Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan
| | - Masakuni Serizawa
- Drug Discovery and Development Division, Shizuoka Cancer Center Research Institute, Shizuoka, Japan
| | - Takeshi Sawada
- Shien-Lab, National Cancer Center Hospital, Tokyo, Japan
| | | | - Toshiaki Takahashi
- Division of Thoracic Oncology, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Nobuyuki Yamamoto
- Third Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan
- Division of Thoracic Oncology, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | | | - Yasuhiro Koh
- Drug Discovery and Development Division, Shizuoka Cancer Center Research Institute, Shizuoka, Japan
- Third Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
83
|
Abstract
The availability of new therapeutic options for the treatment of metastatic castration-resistant prostate cancer (mCRPC) has heightened the importance of monitoring and assessing treatment response. Accordingly, there is an unmet clinical need for reliable biomarkers that can be used to guide therapy. Circulating tumour cells (CTCs) are rare cells that are shed from primary and metastatic tumour deposits into the peripheral circulation, and represent a means of performing noninvasive tumour sampling. Indeed, enumeration of CTCs before and after therapy has shown that CTC burden correlates with prognosis in patients with mCRPC. Moreover, studies have demonstrated the potential of molecular analysis of CTCs in monitoring and predicting response to therapy in patients. This Review describes the challenges associated with monitoring treatment response in mCRPC, and the advancements in CTC-analysis technologies applied to such assessments and, ultimately, guiding prostate cancer treatment.
Collapse
|
84
|
Haber DA, Velculescu VE. Blood-based analyses of cancer: circulating tumor cells and circulating tumor DNA. Cancer Discov 2014; 4:650-61. [PMID: 24801577 DOI: 10.1158/2159-8290.cd-13-1014] [Citation(s) in RCA: 528] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
UNLABELLED The ability to study nonhematologic cancers through noninvasive sampling of blood is one of the most exciting and rapidly advancing fields in cancer diagnostics. This has been driven both by major technologic advances, including the isolation of intact cancer cells and the analysis of cancer cell-derived DNA from blood samples, and by the increasing application of molecularly driven therapeutics, which rely on such accurate and timely measurements of critical biomarkers. Moreover, the dramatic efficacy of these potent cancer therapies drives the selection for additional genetic changes as tumors acquire drug resistance, necessitating repeated sampling of cancer cells to adjust therapy in response to tumor evolution. Together, these advanced noninvasive diagnostic capabilities and their applications in guiding precision cancer therapies are poised to change the ways in which we select and monitor cancer treatments. SIGNIFICANCE Recent advances in technologies to analyze circulating tumor cells and circulating tumor DNA are setting the stage for real-time, noninvasive monitoring of cancer and providing novel insights into cancer evolution, invasion, and metastasis.
Collapse
Affiliation(s)
- Daniel A Haber
- Authors' Affiliations:Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts; Howard Hughes Medical Institute, Chevy Chase; and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MarylandAuthors' Affiliations:Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts; Howard Hughes Medical Institute, Chevy Chase; and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Victor E Velculescu
- Authors' Affiliations:Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts; Howard Hughes Medical Institute, Chevy Chase; and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
85
|
Lohr JG, Adalsteinsson VA, Cibulskis K, Choudhury AD, Rosenberg M, Cruz-Gordillo P, Francis J, Zhang CZ, Shalek AK, Satija R, Trombetta JT, Lu D, Tallapragada N, Tahirova N, Kim S, Blumenstiel B, Sougnez C, Lowe A, Wong B, Auclair D, Van Allen EM, Nakabayashi M, Lis RT, Lee GSM, Li T, Chabot MS, Ly A, Taplin ME, Clancy TE, Loda M, Regev A, Meyerson M, Hahn WC, Kantoff PW, Golub TR, Getz G, Boehm JS, Love JC. Whole-exome sequencing of circulating tumor cells provides a window into metastatic prostate cancer. Nat Biotechnol 2014; 32:479-84. [PMID: 24752078 PMCID: PMC4034575 DOI: 10.1038/nbt.2892] [Citation(s) in RCA: 416] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 03/30/2014] [Indexed: 02/06/2023]
Abstract
Comprehensive analyses of cancer genomes promise to inform prognoses and precise cancer treatments. A major barrier, however, is inaccessibility of metastatic tissue. A potential solution is to characterize circulating tumor cells (CTCs), but this requires overcoming the challenges of isolating rare cells and sequencing low-input material. Here we report an integrated process to isolate, qualify and sequence whole exomes of CTCs with high fidelity using a census-based sequencing strategy. Power calculations suggest that mapping of >99.995% of the standard exome is possible in CTCs. We validated our process in two patients with prostate cancer, including one for whom we sequenced CTCs, a lymph node metastasis and nine cores of the primary tumor. Fifty-one of 73 CTC mutations (70%) were present in matched tissue. Moreover, we identified 10 early trunk and 56 metastatic trunk mutations in the non-CTC tumor samples and found 90% and 73% of these mutations, respectively, in CTC exomes. This study establishes a foundation for CTC genomics in the clinic.
Collapse
Affiliation(s)
- Jens G. Lohr
- The Eli and Edythe Broad Institute, Cambridge, Massachusetts 02412, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Viktor A. Adalsteinsson
- The Eli and Edythe Broad Institute, Cambridge, Massachusetts 02412, USA
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, 77 Massachusetts Ave., Bldg. 76-231, Cambridge, Massachusetts 02139, USA
| | | | - Atish D. Choudhury
- The Eli and Edythe Broad Institute, Cambridge, Massachusetts 02412, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Mara Rosenberg
- The Eli and Edythe Broad Institute, Cambridge, Massachusetts 02412, USA
| | | | - Joshua Francis
- The Eli and Edythe Broad Institute, Cambridge, Massachusetts 02412, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Cheng-Zhong Zhang
- The Eli and Edythe Broad Institute, Cambridge, Massachusetts 02412, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Alex K. Shalek
- Department of Chemistry and Chemical Biology and Department of Physics, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Rahul Satija
- The Eli and Edythe Broad Institute, Cambridge, Massachusetts 02412, USA
| | - John T. Trombetta
- The Eli and Edythe Broad Institute, Cambridge, Massachusetts 02412, USA
| | - Diana Lu
- The Eli and Edythe Broad Institute, Cambridge, Massachusetts 02412, USA
| | - Naren Tallapragada
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, 77 Massachusetts Ave., Bldg. 76-231, Cambridge, Massachusetts 02139, USA
| | - Narmin Tahirova
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, 77 Massachusetts Ave., Bldg. 76-231, Cambridge, Massachusetts 02139, USA
| | - Sora Kim
- The Eli and Edythe Broad Institute, Cambridge, Massachusetts 02412, USA
| | | | - Carrie Sougnez
- The Eli and Edythe Broad Institute, Cambridge, Massachusetts 02412, USA
| | - Alarice Lowe
- Brigham and Women’s Hospital, Boston, Massachusetts 02115, USA
| | - Bang Wong
- The Eli and Edythe Broad Institute, Cambridge, Massachusetts 02412, USA
| | - Daniel Auclair
- The Eli and Edythe Broad Institute, Cambridge, Massachusetts 02412, USA
| | - Eliezer M. Van Allen
- The Eli and Edythe Broad Institute, Cambridge, Massachusetts 02412, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Mari Nakabayashi
- Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Rosina T. Lis
- Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Gwo-Shu M. Lee
- Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Tiantian Li
- Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | | | - Amy Ly
- Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Mary-Ellen Taplin
- Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Thomas E. Clancy
- Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Harvard Medical School, Boston, Massachusetts 02115, USA
- Brigham and Women’s Hospital, Boston, Massachusetts 02115, USA
| | - Massimo Loda
- The Eli and Edythe Broad Institute, Cambridge, Massachusetts 02412, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Harvard Medical School, Boston, Massachusetts 02115, USA
- Brigham and Women’s Hospital, Boston, Massachusetts 02115, USA
| | - Aviv Regev
- The Eli and Edythe Broad Institute, Cambridge, Massachusetts 02412, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
| | - Matthew Meyerson
- The Eli and Edythe Broad Institute, Cambridge, Massachusetts 02412, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Harvard Medical School, Boston, Massachusetts 02115, USA
| | - William C. Hahn
- The Eli and Edythe Broad Institute, Cambridge, Massachusetts 02412, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Harvard Medical School, Boston, Massachusetts 02115, USA
- Brigham and Women’s Hospital, Boston, Massachusetts 02115, USA
| | - Philip W. Kantoff
- Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Todd R. Golub
- The Eli and Edythe Broad Institute, Cambridge, Massachusetts 02412, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Harvard Medical School, Boston, Massachusetts 02115, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
| | - Gad Getz
- The Eli and Edythe Broad Institute, Cambridge, Massachusetts 02412, USA
- Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Jesse S. Boehm
- The Eli and Edythe Broad Institute, Cambridge, Massachusetts 02412, USA
| | - J. Christopher Love
- The Eli and Edythe Broad Institute, Cambridge, Massachusetts 02412, USA
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, 77 Massachusetts Ave., Bldg. 76-231, Cambridge, Massachusetts 02139, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
86
|
Adalsteinsson VA, Love JC. Towards Engineered Processes for Sequencing-Based Analysis of Single Circulating Tumor Cells. Curr Opin Chem Eng 2014; 4:97-104. [PMID: 24839591 DOI: 10.1016/j.coche.2014.01.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Sequencing-based analysis of single circulating tumor cells (CTCs) has the potential to revolutionize our understanding of metastatic cancer and improve clinical care. Technologies exist to enrich, identify, recover, and sequence single cells, but to enable systematic routine analysis of single CTCs from a range of cancer patients, there is a need to establish processes that efficiently integrate these specific operations. Such engineered processes should address challenges associated with the yield and viability of enriched CTCs, the robust identification of candidate single CTCs with minimal degradation of DNA, the bias in whole-genome amplification, and the efficient handling of candidate single CTCs or their amplified DNA products. Advances in methods for single-cell analysis and nanoscale technologies suggest opportunities to overcome these challenges, and could create integrated platforms that perform several of the unit operations together. Ultimately, technologies should be selected or adapted for optimal performance and compatibility in an integrated process.
Collapse
Affiliation(s)
- Viktor A Adalsteinsson
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA ; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA ; The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02412, USA
| | - J Christopher Love
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA ; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA ; The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02412, USA
| |
Collapse
|
87
|
Tsujiura M, Ichikawa D, Konishi H, Komatsu S, Shiozaki A, Otsuji E. Liquid biopsy of gastric cancer patients: Circulating tumor cells and cell-free nucleic acids. World J Gastroenterol 2014; 20:3265-3286. [PMID: 24696609 PMCID: PMC3964398 DOI: 10.3748/wjg.v20.i12.3265] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 12/27/2013] [Accepted: 02/20/2014] [Indexed: 02/06/2023] Open
Abstract
To improve the clinical outcomes of cancer patients, early detection and accurate monitoring of diseases are necessary. Numerous genetic and epigenetic alterations contribute to oncogenesis and cancer progression, and analyses of these changes have been increasingly utilized for diagnostic, prognostic and therapeutic purposes in malignant diseases including gastric cancer (GC). Surgical and/or biopsy specimens are generally used to understand the tumor-associated alterations; however, those approaches cannot always be performed because of their invasive characteristics and may fail to reflect current tumor dynamics and drug sensitivities, which may change during the therapeutic process. Therefore, the importance of developing a non-invasive biomarker with the ability to monitor real-time tumor dynamics should be emphasized. This concept, so called “liquid biopsy”, would provide an ideal therapeutic strategy for an individual cancer patient and would facilitate the development of “tailor-made” cancer management programs. In the blood of cancer patients, the presence and potent utilities of circulating tumor cells (CTCs) and cell-free nucleic acids (cfNAs) such as DNA, mRNA and microRNA have been recognized, and their clinical relevance is attracting considerable attention. In this review, we discuss recent developments in this research field as well as the relevance and future perspectives of CTCs and cfNAs in cancer patients, especially focusing on GC.
Collapse
|
88
|
Krebs MG, Metcalf RL, Carter L, Brady G, Blackhall FH, Dive C. Molecular analysis of circulating tumour cells-biology and biomarkers. Nat Rev Clin Oncol 2014; 11:129-44. [PMID: 24445517 DOI: 10.1038/nrclinonc.2013.253] [Citation(s) in RCA: 464] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Growing evidence for intratumour heterogeneity informs us that single-site biopsies fall short of revealing the complete genomic landscape of a tumour. With an expanding repertoire of targeted agents entering the clinic, screening tumours for genomic aberrations is increasingly important, as is interrogating the tumours for resistance mechanisms upon disease progression. Multiple biopsies separated spatially and temporally are impractical, uncomfortable for the patient and not without risk. Here, we describe how circulating tumour cells (CTCs), captured from a minimally invasive blood test-and readily amenable to serial sampling-have the potential to inform intratumour heterogeneity and tumour evolution, although it remains to be determined how useful this will be in the clinic. Technologies for detecting and isolating CTCs include the validated CellSearch(®) system, but other technologies are gaining prominence. We also discuss how recent CTC discoveries map to mechanisms of haematological spread, previously described in preclinical models, including evidence for epithelial-mesenchymal transition, collective cell migration and cells with tumour-initiating capacity within the circulation. Advances in single-cell molecular analysis are enhancing our ability to explore mechanisms of metastasis, and the combination of CTC and cell-free DNA assays are anticipated to provide invaluable blood-borne biomarkers for real-time patient monitoring and treatment stratification.
Collapse
Affiliation(s)
- Matthew G Krebs
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester and Manchester Cancer Research Centre, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Robert L Metcalf
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester and Manchester Cancer Research Centre, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Louise Carter
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester and Manchester Cancer Research Centre, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Ged Brady
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester and Manchester Cancer Research Centre, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Fiona H Blackhall
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester and Manchester Cancer Research Centre, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Caroline Dive
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester and Manchester Cancer Research Centre, 550 Wilmslow Road, Manchester M20 4BX, UK
| |
Collapse
|
89
|
Single cell analysis of cancer genomes. Curr Opin Genet Dev 2014; 24:82-91. [PMID: 24531336 DOI: 10.1016/j.gde.2013.12.004] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Accepted: 12/15/2013] [Indexed: 12/19/2022]
Abstract
Genomic studies have provided key insights into how cancers develop, evolve, metastasize and respond to treatment. Cancers result from an interplay between mutation, selection and clonal expansions. In solid tumours, this Darwinian competition between subclones is also influenced by topological factors. Recent advances have made it possible to study cancers at the single cell level. These methods represent important tools to dissect cancer evolution and provide the potential to considerably change both cancer research and clinical practice. Here we discuss state-of-the-art methods for the isolation of a single cell, whole-genome and whole-transcriptome amplification of the cell's nucleic acids, as well as microarray and massively parallel sequencing analysis of such amplification products. We discuss the strengths and the limitations of the techniques, and explore single-cell methodologies for future cancer research, as well as diagnosis and treatment of the disease.
Collapse
|
90
|
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: 5.6] [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
|
91
|
Circulating tumor cells in prostate cancer. Cancers (Basel) 2013; 5:1676-90. [PMID: 24305656 PMCID: PMC3875960 DOI: 10.3390/cancers5041676] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 11/20/2013] [Accepted: 11/25/2013] [Indexed: 12/31/2022] Open
Abstract
Circulating tumor cells (CTCs) can provide a non-invasive, repeatable snapshot of an individual patient's tumor. In prostate cancer, CTC enumeration has been extensively studied and validated as a prognostic tool and has received FDA clearance for use in monitoring advanced disease. More recently, CTC analysis has been shifting from enumeration to more sophisticated molecular characterization of captured cells, which serve as a "liquid biopsy" of the tumor, reflecting molecular changes in an individual's malignancy over time. Here we will review the main CTC studies in advanced and localized prostate cancer, highlighting the important gains as well as the challenges posed by various approaches, and their implications for advancing prostate cancer management.
Collapse
|
92
|
Marinov GK, Williams BA, McCue K, Schroth GP, Gertz J, Myers RM, Wold BJ. From single-cell to cell-pool transcriptomes: stochasticity in gene expression and RNA splicing. Genome Res 2013; 24:496-510. [PMID: 24299736 PMCID: PMC3941114 DOI: 10.1101/gr.161034.113] [Citation(s) in RCA: 338] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Single-cell RNA-seq mammalian transcriptome studies are at an early stage in uncovering cell-to-cell variation in gene expression, transcript processing and editing, and regulatory module activity. Despite great progress recently, substantial challenges remain, including discriminating biological variation from technical noise. Here we apply the SMART-seq single-cell RNA-seq protocol to study the reference lymphoblastoid cell line GM12878. By using spike-in quantification standards, we estimate the absolute number of RNA molecules per cell for each gene and find significant variation in total mRNA content: between 50,000 and 300,000 transcripts per cell. We directly measure technical stochasticity by a pool/split design and find that there are significant differences in expression between individual cells, over and above technical variation. Specific gene coexpression modules were preferentially expressed in subsets of individual cells, including one enriched for mRNA processing and splicing factors. We assess cell-to-cell variation in alternative splicing and allelic bias and report evidence of significant differences in splice site usage that exceed splice variation in the pool/split comparison. Finally, we show that transcriptomes from small pools of 30–100 cells approach the information content and reproducibility of contemporary RNA-seq from large amounts of input material. Together, our results define an experimental and computational path forward for analyzing gene expression in rare cell types and cell states.
Collapse
Affiliation(s)
- Georgi K Marinov
- Division of Biology, California Institute of Technology, Pasadena, California 91125, USA
| | | | | | | | | | | | | |
Collapse
|
93
|
Sclafani F, Smyth E, Cunningham D, Chau I, Turner A, Watkins D. A pilot study assessing the incidence and clinical significance of circulating tumor cells in esophagogastric cancers. Clin Colorectal Cancer 2013; 13:94-9. [PMID: 24332356 DOI: 10.1016/j.clcc.2013.11.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 10/10/2013] [Accepted: 11/08/2013] [Indexed: 02/08/2023]
Abstract
BACKGROUND Circulating tumor cells (CTCs) have been found to be of clinical utility in predicting response to treatment and prognosis in several malignancies. Less is known of the prevalence and clinical relevance of CTCs in esophagogastric adenocarcinoma, with the available data arising from heterogeneous patient populations using varied detection methods. PATIENTS AND METHODS A pilot study was undertaken to assess the prevalence of CTCs in patients with advanced esophageal or gastric adenocarcinoma. Patients were eligible if they had advanced disease and either had received no prior therapy or had progressed after prior chemotherapy. Blood samples for CTC analysis were obtained at baseline and during the course of treatment. The CellSearch immunomagnetic CTC detection platform was used. RESULTS Twenty-two patients with metastatic esophageal or gastric adenocarcinoma were enrolled. Eighteen received first-line EOX (epirubicin/oxaliplatin/capecitabine) chemotherapy (± panitumumab) and had baseline samples suitable for CTC analysis. At baseline, ≥ 2 CTCs were detected in 8 patients (44%). Overall tumor response rate was 60% in patients with < 2 CTCs and 37.5% in patients with ≥ 2 CTCs. Median progression-free and overall survival were 6.1 and 10.5 months and 5.2 and 6.1 months in the groups of patients with < 2 CTCs and ≥ 2 CTCs, respectively. The study was prematurely discontinued, owing to the withdrawal of commercial support. CONCLUSION The incidence of CTCs in locally advanced or metastatic esophagogastric cancer may be clinically relevant. Investigation of the potential clinical utility of CTCs is warranted in a larger cohort of patients with esophagogastric cancer.
Collapse
Affiliation(s)
- Francesco Sclafani
- Department of Medicine, Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom.
| | - Elizabeth Smyth
- Department of Medicine, Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - David Cunningham
- Department of Medicine, Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Ian Chau
- Department of Medicine, Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Andrea Turner
- Department of Medicine, Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - David Watkins
- Department of Medicine, Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| |
Collapse
|
94
|
Kin C, Kidess E, Poultsides GA, Visser BC, Jeffrey SS. Colorectal cancer diagnostics: biomarkers, cell-free DNA, circulating tumor cells and defining heterogeneous populations by single-cell analysis. Expert Rev Mol Diagn 2013; 13:581-99. [PMID: 23895128 DOI: 10.1586/14737159.2013.811896] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Reliable biomarkers are needed to guide treatment of colorectal cancer, as well as for surveillance to detect recurrence and monitor therapeutic response. In this review, the authors discuss the use of various biomarkers in addition to serum carcinoembryonic antigen, the current surveillance method for metastatic recurrence after resection. The clinical relevance of mutations including microsatellite instability, KRAS, BRAF and SMAD4 is addressed. The role of circulating tumor cells and cell-free DNA with regards to their implementation into clinical use is discussed, as well as how single-cell analysis may fit into a monitoring program. The detection and characterization of circulating tumor cells and cell-free DNA in colorectal cancer patients will not only improve the understanding of the development of metastasis, but may also supplant the use of other biomarkers.
Collapse
Affiliation(s)
- Cindy Kin
- Department of Surgery, Stanford University School of Medicine, CA, USA.
| | | | | | | | | |
Collapse
|
95
|
Becker TM, Caixeiro NJ, Lim SH, Tognela A, Kienzle N, Scott KF, Spring KJ, de Souza P. New frontiers in circulating tumor cell analysis: A reference guide for biomolecular profiling toward translational clinical use. Int J Cancer 2013; 134:2523-33. [PMID: 24122526 DOI: 10.1002/ijc.28516] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 09/04/2013] [Accepted: 09/16/2013] [Indexed: 12/14/2022]
Abstract
Circulating tumor cells (CTCs) are now routinely isolated from blood, and measurement of CTC concentrations appears to correlate well with survival in patients with cancer. Interrogation of the molecular profile of CTCs for expression of protein biomarkers, genetic variants and gene expression provides opportunities to use this information to guide personalized treatment, monitor therapy and detect emerging resistance. However, successful application of profiling techniques requires analyses that deliver a reliable and clinically relevant representation of a patient's cancer as it changes with time. Here, we comprehensively review the current knowledge of therapeutically relevant biomarkers in isolated CTCs obtained by fluorescence imaging and genomic profiling approaches. The reviewed data support the notion that molecular profiling of CTCs will provide a reliable representation or surrogate index of tumor burden. Large-scale translational trials, many currently in progress, will provide critical data to progress CTC analysis toward wider clinical use in personalized treatment.
Collapse
Affiliation(s)
- Therese M Becker
- Ingham Institute for Applied Medical Research, Medical Oncology, Liverpool, New South Wales, Australia; University of New South Wales, School of Medicine, Sydney, New South Wales, Australia
| | | | | | | | | | | | | | | |
Collapse
|
96
|
Li J, Yin ZF. Molecular characterization of circulating tumor cells and individualized cancer diagnosis and therapy. Shijie Huaren Xiaohua Zazhi 2013; 21:2679-2684. [DOI: 10.11569/wcjd.v21.i26.2679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The heterogeneity of cancer cells involved in oncogenesis and metastasis has always been the key factor that impedes tumor diagnosis and treatment (especially traditional chemotherapies). In recent years, molecular characterization of tumors and accordingly implementation of individualized treatment targeting specific molecular markers have become a hotspot for cancer research. As a link between the primary tumor and metastases, circulating tumor cells (CTCs) provide a window into tumor biology and the metastatic cascade. With their real-time, non-invasive and repeatable access, CTCs are excellent resources of tumor specimens. Molecular characterization of CTCs is of great significance for tumor molecular analysis and individualized treatment. Here we review the recent progress in molecular characterization of CTCs and individualized cancer diagnosis and therapy.
Collapse
|
97
|
Esmaeilsabzali H, Beischlag TV, Cox ME, Parameswaran AM, Park EJ. Detection and isolation of circulating tumor cells: principles and methods. Biotechnol Adv 2013; 31:1063-84. [PMID: 23999357 DOI: 10.1016/j.biotechadv.2013.08.016] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 07/24/2013] [Accepted: 08/19/2013] [Indexed: 12/17/2022]
Abstract
Efforts to improve the clinical management of several cancers include finding better methods for the quantitative and qualitative analysis of circulating tumor cells (CTCs). However, detection and isolation of CTCs from the blood circulation is not a trivial task given their scarcity and the lack of reliable markers to identify these cells. With a variety of emerging technologies, a thorough review of the exploited principles and techniques as well as the trends observed in the development of these technologies can assist researchers to recognize the potential improvements and alternative approaches. To help better understand the related biological concepts, a simplified framework explaining cancer formation and its spread to other organs as well as how CTCs contribute to this process has been presented first. Then, based on their basic working-principles, the existing methods for detection and isolation of CTCs have been classified and reviewed as nucleic acid-based, physical properties-based and antibody-based methods. The review of literature suggests that antibody-based methods, particularly in conjunction with a microfluidic lab-on-a-chip setting, offer the highest overall performance for detection and isolation of CTCs. Further biological and engineering-related research is required to improve the existing methods. These include finding more specific markers for CTCs as well as enhancing the throughput, sensitivity, and analytic functionality of current devices.
Collapse
Affiliation(s)
- Hadi Esmaeilsabzali
- School of Mechatronic Systems Engineering, Simon Fraser University, 250-13450 102nd Avenue, Surrey, V3T 0A3, BC, Canada; Faculty of Health Sciences, Simon Fraser University, 8888 University Drive, Burnaby, V5A 1S6, BC, Canada; School of Engineering Science, Simon Fraser University, 8888 University Drive, Burnaby, V5A 1S6, BC, Canada
| | | | | | | | | |
Collapse
|
98
|
Morris MJ, Autio KA, Basch EM, Danila DC, Larson S, Scher HI. Monitoring the clinical outcomes in advanced prostate cancer: what imaging modalities and other markers are reliable? Semin Oncol 2013; 40:375-92. [PMID: 23806501 DOI: 10.1053/j.seminoncol.2013.04.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Effective patient care and efficient drug development require accurate tools to assess treatment effects. For metastatic castration-resistant prostate cancer (mCRPC), response biomarkers have historically been poorly reproducible, inaccurate, inconsistently applied, or only loosely associated with tangible clinical benefits such as survival. However, the field of response assessments for prostate cancer is maturing, in compliance with a rigorous process defined by analytic validation, clinical validation, and clinical qualification. For example, bone imaging with technetium-99m scintigraphy has historically been poorly used in prostate cancer clinical trials and routine patient care, and frequently has led to poor decision-making. However, contemporary clinical trial consensus criteria (Prostate Cancer Working Group 2 [PCWG2]) have standardized the definition of progression on bone scintigraphy and the clinical trials endpoint of radiographic progression-free survival (rPFS). A validated bone scan interpretation form captures the relevant data elements. rPFS and the forms have been undergoing prospective testing in multiple phase III studies. The first of these trials demonstrated a high degree of reproducibility and correlation with overall survival, and rPFS was used by the US Food and Drug Administration (FDA) for approval of abiraterone in chemotherapy-naïve mCRPC. Circulating tumor cells (CTC) are another class of assays with significant promise as response-indicator biomarkers. CTC enumeration has undergone analytic validation and has been FDA-cleared for monitoring patients with prostate cancer in conjunction with other clinical methods. It is not yet a surrogate for survival. Patient-reported outcomes (PROs) are direct indicators of patient benefit. The assays to measure PROs must undergo each of the steps of biomarker development, and are increasingly being standardized and used as clinical trial endpoints. In this review, we critically assess each of these classes of novel biomarkers--imaging, CTC, and PROs--in regard to the quality of data supporting their use to monitor clinical outcomes in advanced prostate cancer.
Collapse
Affiliation(s)
- Michael J Morris
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA.
| | | | | | | | | | | |
Collapse
|
99
|
Welty CJ, Coleman I, Coleman R, Lakely B, Xia J, Chen S, Gulati R, Larson SR, Lange PH, Montgomery B, Nelson PS, Vessella RL, Morrissey C. Single cell transcriptomic analysis of prostate cancer cells. BMC Mol Biol 2013; 14:6. [PMID: 23414343 PMCID: PMC3599075 DOI: 10.1186/1471-2199-14-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 02/11/2013] [Indexed: 01/24/2023] Open
Abstract
Background The ability to interrogate circulating tumor cells (CTC) and disseminated tumor cells (DTC) is restricted by the small number detected and isolated (typically <10). To determine if a commercially available technology could provide a transcriptomic profile of a single prostate cancer (PCa) cell, we clonally selected and cultured a single passage of cell cycle synchronized C4-2B PCa cells. Ten sets of single, 5-, or 10-cells were isolated using a micromanipulator under direct visualization with an inverted microscope. Additionally, two groups of 10 individual DTC, each isolated from bone marrow of 2 patients with metastatic PCa were obtained. RNA was amplified using the WT-Ovation™ One-Direct Amplification System. The amplified material was hybridized on a 44K Whole Human Gene Expression Microarray. A high stringency threshold, a mean Alexa Fluor® 3 signal intensity above 300, was used for gene detection. Relative expression levels were validated for select genes using real-time PCR (RT-qPCR). Results Using this approach, 22,410, 20,423, and 17,009 probes were positive on the arrays from 10-cell pools, 5-cell pools, and single-cells, respectively. The sensitivity and specificity of gene detection on the single-cell analyses were 0.739 and 0.972 respectively when compared to 10-cell pools, and 0.814 and 0.979 respectively when compared to 5-cell pools, demonstrating a low false positive rate. Among 10,000 randomly selected pairs of genes, the Pearson correlation coefficient was 0.875 between the single-cell and 5-cell pools and 0.783 between the single-cell and 10-cell pools. As expected, abundant transcripts in the 5- and 10-cell samples were detected by RT-qPCR in the single-cell isolates, while lower abundance messages were not. Using the same stringency, 16,039 probes were positive on the patient single-cell arrays. Cluster analysis showed that all 10 DTC grouped together within each patient. Conclusions A transcriptomic profile can be reliably obtained from a single cell using commercially available technology. As expected, fewer amplified genes are detected from a single-cell sample than from pooled-cell samples, however this method can be used to reliably obtain a transcriptomic profile from DTC isolated from the bone marrow of patients with PCa.
Collapse
|