401
|
|
402
|
Alva A, Friedlander T, Clark M, Huebner T, Daignault S, Hussain M, Lee C, Hafez K, Hollenbeck B, Weizer A, Premasekharan G, Tran T, Fu C, Ionescu-Zanetti C, Schwartz M, Fan A, Paris P. Circulating Tumor Cells as Potential Biomarkers in Bladder Cancer. J Urol 2015; 194:790-8. [PMID: 25912492 DOI: 10.1016/j.juro.2015.02.2951] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2015] [Indexed: 02/08/2023]
Abstract
PURPOSE We explored the diagnostic use of circulating tumor cells in patients with neoadjuvant bladder cancer using enumeration and next generation sequencing. MATERIALS AND METHODS A total of 20 patients with bladder cancer who were eligible for cisplatin based neoadjuvant chemotherapy were enrolled in an institutional review board approved study. Subjects underwent blood draws at baseline and after 1 cycle of chemotherapy. A total of 11 patients with metastatic bladder cancer and 13 healthy donors were analyzed for comparison. Samples were enriched for circulating tumor cells using the novel IsoFlux™ System microfluidic collection device. Circulating tumor cell counts were analyzed for repeatability and compared with Food and Drug Administration cleared circulating tumor cells. Circulating tumor cells were also analyzed for mutational status using next generation sequencing. RESULTS Median circulating tumor cell counts were 13 at baseline and 5 at followup in the neoadjuvant group, 29 in the metastatic group and 2 in the healthy group. The concordance of circulating tumor cell levels, defined as low-fewer than 10, medium-11 to 30 and high-greater than 30, across replicate tubes was 100% in 15 preparations. In matched samples the IsoFlux test showed 10 or more circulating tumor cells in 4 of 9 samples (44%) while CellSearch® showed 0 of 9 (0%). At cystectomy 4 months after baseline all 3 patients (100%) with medium/high circulating tumor cell levels at baseline and followup had unfavorable pathological stage disease (T1-T4 or N+). Next generation sequencing analysis showed somatic variant detection in 4 of 8 patients using a targeted cancer panel. All 8 cases (100%) had a medium/high circulating tumor cell level with a circulating tumor cell fraction of greater than 5% purity. CONCLUSIONS This study demonstrates a potential role for circulating tumor cell assays in the management of bladder cancer. The IsoFlux method of circulating tumor cell detection shows increased sensitivity compared with CellSearch. A next generation sequencing assay is presented with sufficient sensitivity to detect genomic alterations in circulating tumor cells.
Collapse
Affiliation(s)
- Ajjai Alva
- Division of Hematology and Oncology, Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan.
| | - Terence Friedlander
- Division of Hematology and Medical Oncology, Helen Diller Family Comprehensive Cancer Center, University of California-San Francisco, San Francisco, California
| | - Melanie Clark
- Division of Hematology and Oncology, Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Tamara Huebner
- Division of Hematology and Oncology, Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Stephanie Daignault
- Division of Hematology and Oncology, Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Maha Hussain
- Division of Hematology and Oncology, Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Cheryl Lee
- Department of Urology, University of Michigan, Ann Arbor, Michigan
| | - Khaled Hafez
- Department of Urology, University of Michigan, Ann Arbor, Michigan
| | - Brent Hollenbeck
- Department of Urology, University of Michigan, Ann Arbor, Michigan
| | - Alon Weizer
- Department of Urology, University of Michigan, Ann Arbor, Michigan
| | - Gayatri Premasekharan
- Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California-San Francisco, San Francisco, California
| | - Tony Tran
- Fluxion Biosciences, San Francisco, California
| | | | | | | | - Andrea Fan
- Fluxion Biosciences, San Francisco, California
| | - Pamela Paris
- Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California-San Francisco, San Francisco, California
| |
Collapse
|
403
|
Calibrating genomic and allelic coverage bias in single-cell sequencing. Nat Commun 2015; 6:6822. [PMID: 25879913 DOI: 10.1038/ncomms7822] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 03/03/2015] [Indexed: 02/07/2023] Open
Abstract
Artifacts introduced in whole-genome amplification (WGA) make it difficult to derive accurate genomic information from single-cell genomes and require different analytical strategies from bulk genome analysis. Here, we describe statistical methods to quantitatively assess the amplification bias resulting from whole-genome amplification of single-cell genomic DNA. Analysis of single-cell DNA libraries generated by different technologies revealed universal features of the genome coverage bias predominantly generated at the amplicon level (1-10 kb). The magnitude of coverage bias can be accurately calibrated from low-pass sequencing (∼0.1 × ) to predict the depth-of-coverage yield of single-cell DNA libraries sequenced at arbitrary depths. We further provide a benchmark comparison of single-cell libraries generated by multi-strand displacement amplification (MDA) and multiple annealing and looping-based amplification cycles (MALBAC). Finally, we develop statistical models to calibrate allelic bias in single-cell whole-genome amplification and demonstrate a census-based strategy for efficient and accurate variant detection from low-input biopsy samples.
Collapse
|
404
|
Baslan T, Kendall J, Ward B, Cox H, Leotta A, Rodgers L, Riggs M, D'Italia S, Sun G, Yong M, Miskimen K, Gilmore H, Saborowski M, Dimitrova N, Krasnitz A, Harris L, Wigler M, Hicks J. Optimizing sparse sequencing of single cells for highly multiplex copy number profiling. Genome Res 2015; 25:714-24. [PMID: 25858951 PMCID: PMC4417119 DOI: 10.1101/gr.188060.114] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 03/19/2015] [Indexed: 12/12/2022]
Abstract
Genome-wide analysis at the level of single cells has recently emerged as a powerful tool to dissect genome heterogeneity in cancer, neurobiology, and development. To be truly transformative, single-cell approaches must affordably accommodate large numbers of single cells. This is feasible in the case of copy number variation (CNV), because CNV determination requires only sparse sequence coverage. We have used a combination of bioinformatic and molecular approaches to optimize single-cell DNA amplification and library preparation for highly multiplexed sequencing, yielding a method that can produce genome-wide CNV profiles of up to a hundred individual cells on a single lane of an Illumina HiSeq instrument. We apply the method to human cancer cell lines and biopsied cancer tissue, thereby illustrating its efficiency, reproducibility, and power to reveal underlying genetic heterogeneity and clonal phylogeny. The capacity of the method to facilitate the rapid profiling of hundreds to thousands of single-cell genomes represents a key step in making single-cell profiling an easily accessible tool for studying cell lineage.
Collapse
Affiliation(s)
- Timour Baslan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA; Department of Molecular and Cellular Biology, Stony Brook University, Stony Brook, New York 11790, USA
| | - Jude Kendall
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Brian Ward
- Sigma-Aldrich Research Technology, Saint Louis, Missouri 63103, USA
| | - Hilary Cox
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Anthony Leotta
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Linda Rodgers
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Michael Riggs
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Sean D'Italia
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Guoli Sun
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Mao Yong
- Phillips Research North America, Biomedical Informatics, Briarcliff Manor, New York 10510, USA
| | - Kristy Miskimen
- Division of Hematology/Oncology, Department of Medicine, Case Western Reserve School of Medicine, Cleveland, Ohio 44106, USA
| | - Hannah Gilmore
- Department of Pathology, University Hospitals Case Medical Center and Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Michael Saborowski
- Clinic for Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Nevenka Dimitrova
- Phillips Research North America, Biomedical Informatics, Briarcliff Manor, New York 10510, USA
| | | | - Lyndsay Harris
- Division of Hematology/Oncology, Department of Medicine, Case Western Reserve School of Medicine, Cleveland, Ohio 44106, USA; Seidman Cancer Center, University Hospitals of Case Western, Cleveland, Ohio 44106, USA
| | - Michael Wigler
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - James Hicks
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| |
Collapse
|
405
|
|
406
|
Leung ML, Wang Y, Waters J, Navin NE. SNES: single nucleus exome sequencing. Genome Biol 2015; 16:55. [PMID: 25853327 PMCID: PMC4373516 DOI: 10.1186/s13059-015-0616-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 02/18/2015] [Indexed: 01/17/2023] Open
Abstract
Single-cell genome sequencing methods are challenged by poor physical coverage and high error rates, making it difficult to distinguish real biological variants from technical artifacts. To address this problem, we developed a method called SNES that combines flow-sorting of single G1/0 or G2/M nuclei, time-limited multiple-displacement-amplification, exome capture, and next-generation sequencing to generate high coverage (96%) data from single human cells. We validated our method in a fibroblast cell line, and show low allelic dropout and false-positive error rates, resulting in high detection efficiencies for single nucleotide variants (92%) and indels (85%) in single cells.
Collapse
|
407
|
Abstract
Advances in next-generation sequencing and bioinformatics have led to an unprecedented view of the cancer genome and its evolution. Genomic studies have demonstrated the complex and heterogeneous clonal landscape of tumors of different origins and the potential impact of intratumor heterogeneity on treatment response and resistance, cancer progression, and the risk of disease relapse. However, the significance of subclonal mutations, in particular mutations in driver genes, and their evolution through time and their dynamics in response to cancer therapies, is yet to be determined. The necessary tools are now available to prospectively determine whether clonal heterogeneity can be used as a biomarker of clinical outcome and to what extent subclonal somatic alterations might influence clinical outcome. Studies that use longitudinal tissue sampling, integrating both genomic and clinical data, have the potential to reveal the subclonal composition and track the evolution of tumors to address these questions and to begin to define the breadth of genetic diversity in different tumor types and its relevance to patient outcome. Such studies may provide further evidence for drug-resistance mechanisms informing combinatorial, adaptive, and tumor immune therapies placed within the context of tumor evolution.
Collapse
Affiliation(s)
- Mariam Jamal-Hanjani
- UCL Cancer Institute, Paul O'Gorman Building, London, United Kingdom. Cancer Research UK London Research institute, London, United Kingdom
| | - Sergio A Quezada
- UCL Cancer Institute, Paul O'Gorman Building, London, United Kingdom
| | - James Larkin
- Department of Oncology, Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Charles Swanton
- UCL Cancer Institute, Paul O'Gorman Building, London, United Kingdom. Cancer Research UK London Research institute, London, United Kingdom.
| |
Collapse
|
408
|
Chen S, Sanjana NE, Zheng K, Shalem O, Lee K, Shi X, Scott DA, Song J, Pan JQ, Weissleder R, Lee H, Zhang F, Sharp PA. Genome-wide CRISPR screen in a mouse model of tumor growth and metastasis. Cell 2015; 160:1246-60. [PMID: 25748654 PMCID: PMC4380877 DOI: 10.1016/j.cell.2015.02.038] [Citation(s) in RCA: 670] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 02/03/2015] [Accepted: 02/18/2015] [Indexed: 12/14/2022]
Abstract
Genetic screens are powerful tools for identifying genes responsible for diverse phenotypes. Here we describe a genome-wide CRISPR/Cas9-mediated loss-of-function screen in tumor growth and metastasis. We mutagenized a non-metastatic mouse cancer cell line using a genome-scale library with 67,405 single-guide RNAs (sgRNAs). The mutant cell pool rapidly generates metastases when transplanted into immunocompromised mice. Enriched sgRNAs in lung metastases and late-stage primary tumors were found to target a small set of genes, suggesting that specific loss-of-function mutations drive tumor growth and metastasis. Individual sgRNAs and a small pool of 624 sgRNAs targeting the top-scoring genes from the primary screen dramatically accelerate metastasis. In all of these experiments, the effect of mutations on primary tumor growth positively correlates with the development of metastases. Our study demonstrates Cas9-based screening as a robust method to systematically assay gene phenotypes in cancer evolution in vivo.
Collapse
Affiliation(s)
- Sidi Chen
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Neville E Sanjana
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA; McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kaijie Zheng
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
| | - Ophir Shalem
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
| | - Kyungheon Lee
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Xi Shi
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
| | - David A Scott
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
| | - Jun Song
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jen Q Pan
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Hakho Lee
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Feng Zhang
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA; McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Phillip A Sharp
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
409
|
Nanobiotechnology for the Therapeutic Targeting of Cancer Cells in Blood. Cell Mol Bioeng 2015; 8:137-150. [PMID: 25798204 PMCID: PMC4361771 DOI: 10.1007/s12195-015-0381-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Accepted: 01/19/2015] [Indexed: 12/11/2022] Open
Abstract
During metastasis, circulating tumor cells migrate away from a primary tumor via the blood circulation to form secondary tumors in distant organs. Mounting evidence from clinical observations indicates that the number of circulating tumor cells (CTCs) in the blood correlates with the progression of solid tumors before and during chemotherapy. Beyond the well-established role of CTCs as a fluid biopsy, however, the field of targeting CTCs for the prevention or reduction of metastases has just emerged. Conventional cancer therapeutics have a relatively short circulation time in the blood which may render the killing of CTCs inefficient due to reduced exposure of CTCs to drugs. Nevertheless, over the past few decades, the development of nanoparticles and nanoformulations to improve the half-life and release profile of drugs in circulation has rejuvenated certain traditional medicines in the emerging field of CTC neutralization. This review focuses on how the principles of nanomedicine may be applied to target CTCs. Moreover, inspired by the interactions between CTCs and host cells in the blood circulation, novel biomimetic approaches for targeted drug delivery are presented.
Collapse
|
410
|
Ng CKY, Schultheis AM, Bidard FC, Weigelt B, Reis-Filho JS. Breast cancer genomics from microarrays to massively parallel sequencing: paradigms and new insights. J Natl Cancer Inst 2015; 107:djv015. [PMID: 25713166 DOI: 10.1093/jnci/djv015] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Rapid advancements in massively parallel sequencing methods have enabled the analysis of breast cancer genomes at an unprecedented resolution, which have revealed the remarkable heterogeneity of the disease. As a result, we now accept that despite originating in the breast, estrogen receptor (ER)-positive and ER-negative breast cancers are completely different diseases at the molecular level. It has become apparent that there are very few highly recurrently mutated genes such as TP53, PIK3CA, and GATA3, that no two breast cancers display an identical repertoire of somatic genetic alterations at base-pair resolution and that there might not be a single highly recurrently mutated gene that defines each of the "intrinsic" subtypes of breast cancer (ie, basal-like, HER2-enriched, luminal A, and luminal B). Breast cancer heterogeneity, however, extends beyond the diversity between tumors. There is burgeoning evidence to demonstrate that at least some primary breast cancers are composed of multiple, genetically diverse clones at diagnosis and that metastatic lesions may differ in their repertoire of somatic genetic alterations when compared with their respective primary tumors. Several biological phenomena may shape the reported intratumor genetic heterogeneity observed in breast cancers, including the different mutational processes and multiple types of genomic instability. Harnessing the emerging concepts of the diversity of breast cancer genomes and the phenomenon of intratumor genetic heterogeneity will be essential for the development of optimal methods for diagnosis, disease monitoring, and the matching of patients to the drugs that would benefit them the most.
Collapse
Affiliation(s)
- Charlotte K Y Ng
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY (CKYN, AMS, BW, JSRF); Department of Medical Oncology, SIRIC, Institut Curie, Paris, France (FCB); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY (JSRF)
| | - Anne M Schultheis
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY (CKYN, AMS, BW, JSRF); Department of Medical Oncology, SIRIC, Institut Curie, Paris, France (FCB); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY (JSRF)
| | - Francois-Clement Bidard
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY (CKYN, AMS, BW, JSRF); Department of Medical Oncology, SIRIC, Institut Curie, Paris, France (FCB); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY (JSRF)
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY (CKYN, AMS, BW, JSRF); Department of Medical Oncology, SIRIC, Institut Curie, Paris, France (FCB); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY (JSRF).
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY (CKYN, AMS, BW, JSRF); Department of Medical Oncology, SIRIC, Institut Curie, Paris, France (FCB); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY (JSRF).
| |
Collapse
|
411
|
Affiliation(s)
| | - Klaus Pantel
- Institute of Tumor Biology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| |
Collapse
|
412
|
Nanobiotechnology for the Therapeutic Targeting of Cancer Cells in Blood. Cell Mol Bioeng 2015. [DOI: 10.1007/s12195-015-0378-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
413
|
Todorova K, Metodiev MV, Metodieva G, Zasheva D, Mincheff M, Hayrabedyan S. miR-204 is dysregulated in metastatic prostate cancer in vitro. Mol Carcinog 2015; 55:131-47. [PMID: 25630658 DOI: 10.1002/mc.22263] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Revised: 11/03/2015] [Accepted: 11/07/2015] [Indexed: 02/04/2023]
Abstract
During cancer progression, the genome instability incurred rearrangement could possibly turn some of the tumor suppressor micro-RNAs into pro-oncogenic ones. We aimed to investigate miR-204 in the context of prostate cancer progression using a cell line model of different levels of genome instability (LNCaP, PC3, VCaP and NCI H660), as demonstrated by the availability of ERG fusion. We studied the effect of miR-204 modulation on master transcription factors important for lineage development, cell differentiation and prostate cancer bone marrow metastasis. We followed c-MYB, ETS1 and RUNX2 transcript and protein expression and the miR-204 affected global proteome. We further investigated if these transcription factors exert an effect on miR-204 expression (qPCR, luciferase reporter assay) by silencing them using esiRNA. We found dualistic miR-204 effects, either acting as a tumor suppressor on c-MYB, or as an oncomiR on ETS1. RUNX2 and ETS1 regulation by miR-204 was ERG fusion dependent, demonstrating regulatory circuitry disruption in advanced metastatic models. miR-204 also differentially affected mRNA splicing and protein stability. miR-204 levels were found dependent on cancer hypermethylation and supported by positive feedback induced by all three transcription factors. In this regulatory circuitry among miR-204, c-MYB, RUNX2 and ETS1, the c-MYB was found to induce all three other members, but its expression was differentially affected by the methylation status in lymph node vs. bone metastasis. We demonstrate that not only tumor suppressor micro-RNA loss, but also significant genome rearrangement-driven regulatory loop perturbations play a role in the advanced cancer progression, conferring better pro-survival and metastatic potential.
Collapse
Affiliation(s)
- Krassimira Todorova
- Institute of Biology and Immunology of Reproduction at Bulgarian Academy of Sciences, Sofia, Bulgaria
| | | | | | - Diana Zasheva
- Institute of Biology and Immunology of Reproduction at Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Milcho Mincheff
- Cellular and Gene Therapy Ward, National Specialized Hematology Hospital, Sofia, Bulgaria
| | - Soren Hayrabedyan
- Institute of Biology and Immunology of Reproduction at Bulgarian Academy of Sciences, Sofia, Bulgaria
| |
Collapse
|
414
|
Ke Z, Lin M, Chen JF, Choi JS, Zhang Y, Fong A, Liang AJ, Chen SF, Li Q, Fang W, Zhang P, Garcia MA, Lee T, Song M, Lin HA, Zhao H, Luo SC, Hou S, Yu HH, Tseng HR. Programming thermoresponsiveness of NanoVelcro substrates enables effective purification of circulating tumor cells in lung cancer patients. ACS NANO 2015; 9:62-70. [PMID: 25495128 PMCID: PMC4310634 DOI: 10.1021/nn5056282] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Unlike tumor biopsies that can be constrained by problems such as sampling bias, circulating tumor cells (CTCs) are regarded as the "liquid biopsy" of the tumor, providing convenient access to all disease sites, including primary tumor and fatal metastases. Although enumerating CTCs is of prognostic significance in solid tumors, it is conceivable that performing molecular and functional analyses on CTCs will reveal much significant insight into tumor biology to guide proper therapeutic intervention. We developed the Thermoresponsive NanoVelcro CTC purification system that can be digitally programmed to achieve an optimal performance for purifying CTCs from non-small cell lung cancer (NSCLC) patients. The performance of this unique CTC purification system was optimized by systematically modulating surface chemistry, flow rates, and heating/cooling cycles. By applying a physiologically endurable stimulation (i.e., temperature between 4 and 37 °C), the mild operational parameters allow minimum disruption to CTCs' viability and molecular integrity. Subsequently, we were able to successfully demonstrate culture expansion and mutational analysis of the CTCs purified by this CTC purification system. Most excitingly, we adopted the combined use of the Thermoresponsive NanoVelcro system with downstream mutational analysis to monitor the disease evolution of an index NSCLC patient, highlighting its translational value in managing NSCLC.
Collapse
Affiliation(s)
- Zunfu Ke
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
- Address correspondence to , , ,
| | - Millicent Lin
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Jie-Fu Chen
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Jin-sil Choi
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Yang Zhang
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Anna Fong
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - An-Jou Liang
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Shang-Fu Chen
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Qingyu Li
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Wenfeng Fang
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Pingshan Zhang
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Mitch A. Garcia
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Tom Lee
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Min Song
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Hsing-An Lin
- Responsive Organic Materials Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Haichao Zhao
- Responsive Organic Materials Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Shyh-Chyang Luo
- Responsive Organic Materials Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
- Department of Materials Science and Engineering, National Cheng Kung University, Tainan 70101, Taiwan
| | - Shuang Hou
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
- Address correspondence to , , ,
| | - Hsiao-hua Yu
- Responsive Organic Materials Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
- Institute of Chemistry, Academia Sinica, 128 Academia Road Sec. 2, Nankang, Taipei 115, Taiwan
- Address correspondence to , , ,
| | - Hsian-Rong Tseng
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California 90095, United States
- Address correspondence to , , ,
| |
Collapse
|
415
|
Viraka Nellore BP, Kanchanapally R, Pramanik A, Sinha SS, Chavva SR, Hamme A, Ray PC. Aptamer-conjugated graphene oxide membranes for highly efficient capture and accurate identification of multiple types of circulating tumor cells. Bioconjug Chem 2015; 26:235-42. [PMID: 25565372 PMCID: PMC4578366 DOI: 10.1021/bc500503e] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Tumor metastasis is responsible for 1 in 4 deaths in the United States. Though it has been well-documented over past two decades that circulating tumor cells (CTCs) in blood can be used as a biomarker for metastatic cancer, there are enormous challenges in capturing and identifying CTCs with sufficient sensitivity and specificity. Because of the heterogeneous expression of CTC markers, it is now well understood that a single CTC marker is insufficient to capture all CTCs from the blood. Driven by the clear need, this study reports for the first time highly efficient capture and accurate identification of multiple types of CTCs from infected blood using aptamer-modified porous graphene oxide membranes. The results demonstrate that dye-modified S6, A9, and YJ-1 aptamers attached to 20-40 μm porous garphene oxide membranes are capable of capturing multiple types of tumor cells (SKBR3 breast cancer cells, LNCaP prostate cancer cells, and SW-948 colon cancer cells) selectively and simultaneously from infected blood. Our result shows that the capture efficiency of graphene oxide membranes is ~95% for multiple types of tumor cells; for each tumor concentration, 10 cells are present per milliliter of blood sample. The selectivity of our assay for capturing targeted tumor cells has been demonstrated using membranes without an antibody. Blood infected with different cells also has been used to demonstrate the targeted tumor cell capturing ability of aptamer-conjugated membranes. Our data also demonstrate that accurate analysis of multiple types of captured CTCs can be performed using multicolor fluorescence imaging. Aptamer-conjugated membranes reported here have good potential for the early diagnosis of diseases that are currently being detected by means of cell capture technologies.
Collapse
Affiliation(s)
- Bhanu Priya Viraka Nellore
- Department of Chemistry and Biochemistry, Jackson State University , Jackson, Mississippi 39217, United States
| | | | | | | | | | | | | |
Collapse
|
416
|
Orsola A, Bellmunt J. The “Artificial” Docetaxel Space: The Evolving Treatment Paradigm of Metastatic Castration-Resistant Prostate Cancer. Eur Urol 2015; 67:30-32. [DOI: 10.1016/j.eururo.2014.07.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 07/18/2014] [Indexed: 11/24/2022]
|
417
|
Reyes EE, VanderWeele DJ, Isikbay M, Duggan R, Campanile A, Stadler WM, Vander Griend DJ, Szmulewitz RZ. Quantitative characterization of androgen receptor protein expression and cellular localization in circulating tumor cells from patients with metastatic castration-resistant prostate cancer. J Transl Med 2014; 12:313. [PMID: 25424879 PMCID: PMC4252013 DOI: 10.1186/s12967-014-0313-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 10/27/2014] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Many current therapies for metastatic castration-resistant prostate cancer (mCRPC) are aimed at AR signaling; however, resistance to these therapies is inevitable. To personalize CRPC therapy in an individual with clinical progression despite maximal AR signaling blockade, it is important to characterize the status of AR activity within their cancer. Biopsies of bone metastases are invasive and frequently fail to yield sufficient tissue for further study. Evaluation of circulating tumor cells (CTCs) offers an alternative, minimally invasive mechanism to characterize and study late-stage disease. The goal of this study was to evaluate the utility of CTC interrogation with respect to the AR as a potential novel therapeutic biomarker in patients with mCRPC. METHODS Fifteen mL of whole blood was collected from patients with progressive, metastatic mCRPC, the mononuclear cell portion was isolated, and fluorescence-activated cell sorting (FACS) was used to isolate and evaluate CTCs. A novel protocol was optimized to use ImageStreamX to quantitatively analyze AR expression and subcellular localization within CTCs. Co-expression of AR and the proliferation marker Ki67 was also determined using ImageStreamX. RESULTS We found inter-patient and intra-patient heterogeneity in expression and localization of AR. Increased AR expression and nuclear localization are associated with elevated co-expression of Ki-67, consistent with the continued role for AR in castration-resistant disease. Despite intra-patient heterogeneity, CTCs from patients with prior exposure to abiraterone had increased AR expression compared to CTCs from patients who were abiraterone-naïve. CONCLUSIONS As our toolbox for targeting AR function expands, our ability to evaluate AR expression and function within tumor samples from patients with late-stage disease will likely be a critical component of the personalized management of advanced prostate cancer. AR expression and nuclear localization varies within patients and between patients; however it remains associated with markers of proliferation. This supports a molecularly diverse AR-centric pathobiology imparting castration-resistance.
Collapse
Affiliation(s)
- Edwin E Reyes
- Department of Surgery, University of Chicago, Chicago, IL, USA.
- Committee on Immunology, University of Chicago, Chicago, IL, USA.
| | | | - Masis Isikbay
- Department of Surgery, University of Chicago, Chicago, IL, USA.
| | - Ryan Duggan
- Flow Cytometry Facility, University of Chicago, Chicago, IL, USA.
| | - Alexa Campanile
- Department of Medicine, University of Chicago, Chicago, IL, USA.
| | - Walter M Stadler
- Department of Surgery, University of Chicago, Chicago, IL, USA.
- Department of Medicine, University of Chicago, Chicago, IL, USA.
| | | | | |
Collapse
|
418
|
Bone marrow as a reservoir for disseminated tumor cells: a special source for liquid biopsy in cancer patients. BONEKEY REPORTS 2014; 3:584. [PMID: 25419458 DOI: 10.1038/bonekey.2014.79] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 09/03/2014] [Indexed: 12/13/2022]
Abstract
Besides circulating tumor cells, disseminated tumor cells (DTCs) in bone marrow (BM) might be used as a 'liquid biopsy' to obtain information helpful to steer therapies in individual patients. Moreover, the molecular characterization of DTCs may provide important insight into the biology of cancer metastasis. BM is a frequent site of metastasis in breast, prostate and lung cancer, and it might represent a sanctuary site for DTCs derived from various additional types of epithelial tumors. Highly sensitive and specific immunocytological and molecular methods enable the detection of DTCs in BM of cancer patients at the single-cell level years before the occurrence of metastases. This information might be useful to assess individual prognosis and stratify patients at risk to systemic adjuvant anti-cancer therapies. Although most data on the prognostic value of DTCs are available for breast cancer, several single institution studies including patients with colon, lung, prostate, esophageal, gastric, pancreatic, ovarian and head and neck carcinomas have also documented an association between the presence of DTCs at primary surgery and subsequent metastatic relapse. Most DTCs are in a dormant (that is, non-proliferative) stage, frequently express HER2 and display a cancer stem cell and immune escape phenotype. Here, we summarize the current knowledge about specific biological properties of DTCs in BM, and discuss the clinical relevance of DTC detection in cancer patients with regard to an improved individualized therapeutic management. This will stimulate further technical developments that may make BM sampling more acceptable for the clinical management of patients with solid tumors.
Collapse
|
419
|
Zheng F, Cheng Y, Wang J, Lu J, Zhang B, Zhao Y, Gu Z. Aptamer-functionalized barcode particles for the capture and detection of multiple types of circulating tumor cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2014; 26:7333-8. [PMID: 25251012 DOI: 10.1002/adma.201403530] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Indexed: 05/21/2023]
Abstract
Aptamer-functionalized barcode particles are employed to capture and detect various types of circulating tumor cells (CTCs). The particles are spherical colloidal crystal clusters, and the reflection properties that arise from their structures are how their codes are evaluated. Aptamer functionalization (with TD05, Sgc8, and Sgd5) make the particles interact with specific CTC types; dendrimers are used to amplify the effect of the aptamers, allowing for increased sensitivity, reliability, and specificity in CTC capture, detection, and subsequent release.
Collapse
Affiliation(s)
- Fuyin Zheng
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, 210096, China
| | | | | | | | | | | | | |
Collapse
|
420
|
Lin M, Chen JF, Lu YT, Zhang Y, Song J, Hou S, Ke Z, Tseng HR. Nanostructure embedded microchips for detection, isolation, and characterization of circulating tumor cells. Acc Chem Res 2014; 47:2941-50. [PMID: 25111636 PMCID: PMC4204926 DOI: 10.1021/ar5001617] [Citation(s) in RCA: 171] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Circulating
tumor cells (CTCs) are cancer cells that break away
from either a primary tumor or a metastatic site and circulate in
the peripheral blood as the cellular origin of metastasis. With their
role as a “tumor liquid biopsy”, CTCs provide convenient
access to all disease sites, including that of the primary tumor and
the site of fatal metastases. It is conceivable that detecting and
analyzing CTCs will provide insightful information in assessing the
disease status without the flaws and limitations encountered in performing
conventional tumor biopsies. However, identifying CTCs in patient
blood samples is technically challenging due to the extremely low
abundance of CTCs among a large number of hematologic cells. To address
this unmet need, there have been significant research endeavors, especially
in the fields of chemistry, materials science, and bioengineering,
devoted to developing CTC detection, isolation, and characterization
technologies. Inspired by the nanoscale interactions observed
in the tissue microenvironment,
our research team at UCLA pioneered a unique concept of “NanoVelcro”
cell-affinity substrates, in which CTC capture agent-coated nanostructured
substrates were utilized to immobilize CTCs with high efficiency.
The working mechanism of NanoVelcro cell-affinity substrates mimics
that of Velcro: when the two fabric strips of a Velcro fastener are
pressed together, tangling between the hairy surfaces on two strips
leads to strong binding. Through continuous evolution, three generations
(gens) of NanoVelcro CTC chips have been established to achieve different
clinical utilities. The first-gen NanoVelcro chip, composed of a silicon
nanowire substrate (SiNS) and an overlaid microfluidic chaotic mixer,
was created for CTC enumeration. Side-by-side analytical validation
studies using clinical blood samples suggested that the sensitivity
of first-gen NanoVelcro chip outperforms that of FDA-approved CellSearch.
In conjunction with the use of the laser microdissection (LMD) technique,
second-gen NanoVelcro chips (i.e., NanoVelcro-LMD), based on polymer
nanosubstrates, were developed for single-CTC isolation. The individually
isolated CTCs can be subjected to single-CTC genotyping (e.g., Sanger
sequencing and next-generation sequencing, NGS) to verify the CTC’s
role as tumor liquid biopsy. Created by grafting of thermoresponsive
polymer brushes onto SiNS, third-gen NanoVelcro chips (i.e., Thermoresponsive
NanoVelcro) have demonstrated the capture and release of CTCs at 37
and 4 °C, respectively. The temperature-dependent conformational
changes of polymer brushes can effectively alter the accessibility
of the capture agent on SiNS, allowing for rapid CTC purification
with desired viability and molecular integrity. This Account
summarizes the continuous evolution of NanoVelcro
CTC assays from the emergence of the original idea all the way to
their applications in cancer research. We envision that NanoVelcro
CTC assays will lead the way for powerful and cost-efficient diagnostic
platforms for researchers to better understand underlying disease
mechanisms and for physicians to monitor real-time disease progression.
Collapse
Affiliation(s)
- Millicent Lin
- Department
of Pathology, The First Affiliated hospital of Sun Yat-sen University, Guangzhou, 510080 Guangdong, People’s Republic of China
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Jie-Fu Chen
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Yi-Tsung Lu
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Yang Zhang
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Jinzhao Song
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Shuang Hou
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Zunfu Ke
- Department
of Pathology, The First Affiliated hospital of Sun Yat-sen University, Guangzhou, 510080 Guangdong, People’s Republic of China
| | - Hsian-Rong Tseng
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| |
Collapse
|
421
|
Ignatiadis M, Dawson SJ. Circulating tumor cells and circulating tumor DNA for precision medicine: dream or reality? Ann Oncol 2014; 25:2304-2313. [PMID: 25336116 DOI: 10.1093/annonc/mdu480] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Next-generation sequencing studies have provided further evidence to support the notion that cancer is a disease characterized by Darwinian evolution. Today, we often fail to capture this evolution and treatment decisions, even in the metastatic setting, are often based on analysis of primary tumor diagnosed years ago. Currently, this is considered a major reason for treatment failures in cancer care. Recent technological advances in the detection and characterization of circulating tumor cells and circulating tumor DNA might address this and allow for treatment tailoring based on real-time monitoring of tumor evolution. In this review, we summarize the most important recent findings in the field, focusing on challenges and opportunities in moving these tools forward in clinical practice.
Collapse
Affiliation(s)
- M Ignatiadis
- Department of Medical Oncology and Breast Cancer Translational Research Laboratory, J. C. Heuson, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.
| | - S-J Dawson
- Division of Cancer Medicine; Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| |
Collapse
|
422
|
Lin E, Chien J, Ong FS, Fan JB. Challenges and opportunities for next-generation sequencing in companion diagnostics. Expert Rev Mol Diagn 2014; 15:193-209. [DOI: 10.1586/14737159.2015.961916] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
423
|
Abstract
During the past ten years, circulating tumour cells (CTCs) have received enormous attention as new biomarkers and the subject of basic research. Although CTCs are already used in numerous clinical trials, their clinical utility is still under investigation. Many issues regarding the detection and characterization of CTCs remain unknown. In this Opinion article, we propose a conceptual framework of CTC assays and point out current challenges of CTC research, which might structure this dynamic field of translational cancer research.
Collapse
Affiliation(s)
- Catherine Alix-Panabières
- 1] University Medical Centre, Saint-Eloi Hospital, Institute of Medicine Regenerative &Biotherapy, Department of Cellular and Tissular Biopathology of Tumors, Laboratory of Rare Human Circulating Cells, 80 Avenue Augustin Fliche 34295 Montpellier Cedex 5, Montpellier, France. [2] University Institute of Clinical Research UM1 - EA2415 - Epidemiology, Biostatistics &Public Health, 641, Avenue du Doyen Gaston GIRAUD 34093 Montpellier Cedex 5, Montpellier, France
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center, Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| |
Collapse
|
424
|
Mateo J, Gerlinger M, Rodrigues DN, de Bono JS. The promise of circulating tumor cell analysis in cancer management. Genome Biol 2014. [PMID: 25222379 DOI: 10.1186/s13059‐014‐0448‐5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Enumeration and molecular characterization of circulating tumor cells isolated from peripheral blood of patients with cancer can aid selection of targeted therapy for patients, monitoring of response to therapies and optimization of drug development, while also providing valuable information about intratumoral heterogeneity.
Collapse
|
425
|
Mateo J, Gerlinger M, Rodrigues DN, de Bono JS. The promise of circulating tumor cell analysis in cancer management. Genome Biol 2014; 15:448. [PMID: 25222379 PMCID: PMC4281949 DOI: 10.1186/s13059-014-0448-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Enumeration and molecular characterization of circulating tumor cells isolated from peripheral blood of patients with cancer can aid selection of targeted therapy for patients, monitoring of response to therapies and optimization of drug development, while also providing valuable information about intratumoral heterogeneity.
Collapse
Affiliation(s)
- Joaquin Mateo
- />Division of Cancer Therapeutics and Division of Clinical Studies, The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey, SM2 5NG UK
- />Drug Development Unit, The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT UK
| | - Marco Gerlinger
- />Centre for Evolution and Cancer, The Institute of Cancer Research, 123 Old Brompton Road, London, SW7 3RP UK
- />Gastrointestinal Cancer Unit, Department of Medicine, The Royal Marsden NHS Foundation Trust, Fulham Road, London, SW3 6JJ UK
| | - Daniel Nava Rodrigues
- />Division of Cancer Therapeutics and Division of Clinical Studies, The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey, SM2 5NG UK
- />Drug Development Unit, The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT UK
| | - Johann S de Bono
- />Division of Cancer Therapeutics and Division of Clinical Studies, The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey, SM2 5NG UK
- />Drug Development Unit, The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT UK
| |
Collapse
|
426
|
Abstract
The study of single cancer cells has transformed from qualitative microscopic images to quantitative genomic datasets. This paradigm shift has been fueled by the development of single-cell sequencing technologies, which provide a powerful new approach to study complex biological processes in human cancers.
Collapse
|
427
|
Lianidou ES. Molecular characterization of circulating tumor cells: Holy Grail for personalized cancer treatment? Clin Chem 2014; 60:1249-51. [PMID: 25142245 DOI: 10.1373/clinchem.2014.230144] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Evi S Lianidou
- Analysis of Circulating Tumor Cells (ACTC) Lab, Laboratory of Analytical Chemistry, Department of Chemistry, University of Athens, Athens, Greece.
| |
Collapse
|
428
|
Ten years of next-generation sequencing technology. Trends Genet 2014; 30:418-26. [PMID: 25108476 DOI: 10.1016/j.tig.2014.07.001] [Citation(s) in RCA: 901] [Impact Index Per Article: 81.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 07/08/2014] [Accepted: 07/09/2014] [Indexed: 02/06/2023]
Abstract
Ten years ago next-generation sequencing (NGS) technologies appeared on the market. During the past decade, tremendous progress has been made in terms of speed, read length, and throughput, along with a sharp reduction in per-base cost. Together, these advances democratized NGS and paved the way for the development of a large number of novel NGS applications in basic science as well as in translational research areas such as clinical diagnostics, agrigenomics, and forensic science. Here we provide an overview of the evolution of NGS and discuss the most significant improvements in sequencing technologies and library preparation protocols. We also explore the current landscape of NGS applications and provide a perspective for future developments.
Collapse
|
429
|
Yao X, Williamson C, Adalsteinsson VA, D'Agostino RS, Fitton T, Smaroff GG, William RT, Wittrup KD, Love JC. Tumor cells are dislodged into the pulmonary vein during lobectomy. J Thorac Cardiovasc Surg 2014; 148:3224-31.e1-5. [PMID: 25172322 DOI: 10.1016/j.jtcvs.2014.06.074] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 05/29/2014] [Accepted: 06/13/2014] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Intraoperative tumor shedding may facilitate tumor dissemination. In earlier studies, shed tumor cells were defined primarily by cytomorphological examination, and normal epithelial cells could not always be distinguished from tumor cells. We sought to accurately identify tumor cells using single-cell sequencing and determine whether these cells were mobilized into the circulation during pulmonary lobectomy. METHODS Forty-two blood samples collected from the tumor-draining pulmonary vein at the end of lobectomy procedures were analyzed. Arrays of nanowells were used to enumerate and retrieve single EpCAM(+) cells. Targeted sequencing of 10 to 15 cells and nested polymerase chain reaction of single cells detected somatic mutations in shed epithelial cells consistent with patient-matched tumor but not normal tissue. RESULTS The mean number of EpCAM(+) cells in video-assisted thoracoscopy (VATS) lobectomy (no wedge) specimens (n = 16) was 165 (median, 115; range, 0-509) but sampling cells from 3 patients indicated that only 0% to 38% of the EpCAM(+) cells were tumor cells. The mean number of EpCAM(+) cells in VATS lobectomy (wedge) specimens (n = 12) was 1128 (median, 197; range, 47-9406) and all of the EpCAM(+) cells were normal epithelial cells in 2 patients sampled. The mean number of EpCAM(+) cells in thoracotomy specimens (n = 14) was 238 (median, 22; range, 9-2920) and 0% to 50% of total EpCAM(+) cells were tumor cells based on 4 patients sampled. CONCLUSIONS Surgery mobilizes tumor cells into the pulmonary vein, along with many normal epithelial cells. EpCAM alone cannot differentiate between normal and tumor cells. On the other hand, single-cell genetic approaches with patient-matched normal and tumor tissues can accurately quantify the number of shed tumor cells.
Collapse
Affiliation(s)
- Xiaosai Yao
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Mass; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Mass
| | - Christina Williamson
- Department of Thoracic and Cardiovascular Surgery, Lahey Hospital and Medical Center, Burlington, Mass
| | - Viktor A Adalsteinsson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Mass; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Mass; Broad Institute of MIT and Harvard, Cambridge, Mass
| | - Richard S D'Agostino
- Department of Thoracic and Cardiovascular Surgery, Lahey Hospital and Medical Center, Burlington, Mass
| | - Torin Fitton
- Department of Thoracic and Cardiovascular Surgery, Lahey Hospital and Medical Center, Burlington, Mass
| | - Gregory G Smaroff
- Department of Thoracic and Cardiovascular Surgery, Lahey Hospital and Medical Center, Burlington, Mass
| | - Robert T William
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Mass
| | - K Dane Wittrup
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Mass; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Mass; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Mass
| | - J Christopher Love
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Mass; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Mass; Broad Institute of MIT and Harvard, Cambridge, Mass.
| |
Collapse
|
430
|
Single cell mutational analysis of PIK3CA in circulating tumor cells and metastases in breast cancer reveals heterogeneity, discordance, and mutation persistence in cultured disseminated tumor cells from bone marrow. BMC Cancer 2014; 14:456. [PMID: 24947048 PMCID: PMC4071027 DOI: 10.1186/1471-2407-14-456] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 05/28/2014] [Indexed: 12/30/2022] Open
Abstract
Background Therapeutic decisions in cancer are generally guided by molecular biomarkers or, for some newer therapeutics, primary tumor genotype. However, because biomarkers or genotypes may change as new metastases emerge, circulating tumor cells (CTCs) from blood are being investigated for a role in guiding real-time drug selection during disease progression, expecting that CTCs will comprehensively represent the full spectrum of genomic changes in metastases. However, information is limited regarding mutational heterogeneity among CTCs and metastases in breast cancer as discerned by single cell analysis. The presence of disseminated tumor cells (DTCs) in bone marrow also carry prognostic significance in breast cancer, but with variability between CTC and DTC detection. Here we analyze a series of single tumor cells, CTCs, and DTCs for PIK3CA mutations and report CTC and corresponding metastatic genotypes. Methods We used the MagSweeper, an immunomagnetic separation device, to capture live single tumor cells from breast cancer patients’ primary and metastatic tissues, blood, and bone marrow. Single cells were screened for mutations in exons 9 and 20 of the PIK3CA gene. Captured DTCs grown in cell culture were also sequenced for PIK3CA mutations. Results Among 242 individual tumor cells isolated from 17 patients and tested for mutations, 48 mutated tumor cells were identified in three patients. Single cell analyses revealed mutational heterogeneity among CTCs and tumor cells in tissues. In a patient followed serially, there was mutational discordance between CTCs, DTCs, and metastases, and among CTCs isolated at different time points. DTCs from this patient propagated in vitro contained a PIK3CA mutation, which was maintained despite morphological changes during 21 days of cell culture. Conclusions Single cell analysis of CTCs can demonstrate genotypic heterogeneity, changes over time, and discordance from DTCs and distant metastases. We present a cautionary case showing that CTCs from any single blood draw do not always reflect metastatic genotype, and that CTC and DTC analyses may provide independent clinical information. Isolated DTCs remain viable and can be propagated in culture while maintaining their original mutational status, potentially serving as a future resource for investigating new drug therapies.
Collapse
|