1601
|
Sarangi S, Mosulpuria K, Higgins MJ, Bardia A. The Evolving Role of Circulating Tumor Cells in the Personalized Management of Breast Cancer: from Enumeration to Molecular Characterization. CURRENT BREAST CANCER REPORTS 2014; 6:146-153. [PMID: 25431637 DOI: 10.1007/s12609-014-0149-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Circulating Tumor cells (CTCs) represent tumor cells in the blood stream dislodged from the primary tumor. The presence of CTCs in the bloodstream provides a unique opportunity to sample cancer tissue by means of a relatively less-invasive "liquid biopsy." Over the past decade, there has been a tremendous increase in the amount of research examining the potential clinical utility of CTCs in the management of cancer. A number of techniques to refine the sensitivity and range of CTC assays are also in development. In this article, we review the recent developments in the current and potential clinical applications of CTCs in breast cancer. CTC enumeration already has an established role as a prognostic biomarker in metastatic breast cancer, while molecular characterization of CTCs can serve as a potential predictive biomarker for therapy selection, pharmacodynamic evaluation, and identification of novel actionable targets for novel therapies. The role of CTCs in breast cancer screening and detection of recurrence is currently limited. Further development in techniques will be pivotal in enhancing the broad applicability of CTCs and advancing the field of personalized breast cancer therapy.
Collapse
Affiliation(s)
- Sasmit Sarangi
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | | | - Michaela J Higgins
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - Aditya Bardia
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| |
Collapse
|
1602
|
Prevention of Bone Metastases in Breast Cancer Patients. Therapeutic Perspectives. J Clin Med 2014; 3:521-36. [PMID: 26237389 PMCID: PMC4449698 DOI: 10.3390/jcm3020521] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Revised: 03/03/2014] [Accepted: 03/14/2014] [Indexed: 01/13/2023] Open
Abstract
One in four breast cancer patients is at risk of developing bone metastases in her life time. The early prevention of bone metastases is a crucial challenge. It has been suggested that the use of zoledronic acid (ZOL) in the adjuvant setting may reduce the persistence of disseminated tumor cells and thereby might improve outcome, specifically in a population of patients with a low estrogen microenvironment. More recently, the results of a large meta-analysis from 41 randomized trials comparing a bisphosphonate (BP) to placebo or to an open control have been presented at the 2013 San Antonio Breast Cancer Meeting. Data on 17,016 patients confirm that adjuvant BPs, irrespective of the type of treatment or the treatment schedule and formulation (oral or intra-venously (IV)), significantly reduced bone recurrences and improved breast cancer survival in postmenopausal women. No advantage was seen in premenopausal women. BPs are soon likely to become integrated into standard practice. Published data on the mechanisms involved in tumor cell seeding from the primary site, in homing to bone tissues and in the reactivation of dormant tumor cells will be reviewed; these might offer new ideas for innovative combination strategies.
Collapse
|
1603
|
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: 541] [Impact Index Per Article: 49.2] [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
|
1604
|
Kruglyak KM, Lin E, Ong FS. Next-generation sequencing in precision oncology: challenges and opportunities. Expert Rev Mol Diagn 2014; 14:635-7. [PMID: 24791810 DOI: 10.1586/14737159.2014.916213] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
High throughput gene sequencing is transforming the utilization of genomics in patient care by providing physicians with a powerful tool to aid the diagnosis and management of disease, particularly in precision oncology. As next-generation sequencing (NGS)-based diagnostic assays are developed, significant hurdles such as assessing tumor heterogeneity, characterizing 'driver' and 'passenger' mutations, typing molecular signatures of individual cancers and determining limits of detection pose significant challenges for clinical laboratories and downstream bioinformatics analyses. Despite these challenges, NGS has the potential to affect all facets of cancer treatment, including early detection and diagnosis through cancer screening in at-risk populations and assessing therapeutic efficacy by detection of circulating tumor DNA via noninvasive blood draws. As the utilization of NGS in precision oncology matures, NGS-based laboratory tests could be used throughout the evolution of cancer in patients and allow for cancers to be monitored and managed as a chronic disease, rather than an acute condition.
Collapse
|
1605
|
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: 417] [Impact Index Per Article: 37.9] [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
|
1606
|
|
1607
|
Shtivelman E, Davies MA, Hwu P, Yang J, Lotem M, Oren M, Flaherty KT, Fisher DE. Pathways and therapeutic targets in melanoma. Oncotarget 2014; 5:1701-52. [PMID: 24743024 PMCID: PMC4039128 DOI: 10.18632/oncotarget.1892] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 04/07/2014] [Indexed: 02/07/2023] Open
Abstract
This review aims to summarize the current knowledge of molecular pathways and their clinical relevance in melanoma. Metastatic melanoma was a grim diagnosis, but in recent years tremendous advances have been made in treatments. Chemotherapy provided little benefit in these patients, but development of targeted and new immune approaches made radical changes in prognosis. This would not have happened without remarkable advances in understanding the biology of disease and tremendous progress in the genomic (and other "omics") scale analyses of tumors. The big problems facing the field are no longer focused exclusively on the development of new treatment modalities, though this is a very busy area of clinical research. The focus shifted now to understanding and overcoming resistance to targeted therapies, and understanding the underlying causes of the heterogeneous responses to immune therapy.
Collapse
Affiliation(s)
| | | | - Patrick Hwu
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James Yang
- National Cancer Institute, NIH, Washington DC, USA
| | - Michal Lotem
- Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Moshe Oren
- The Weizmann Institute of Science, Rehovot, Israel
| | | | - David E. Fisher
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| |
Collapse
|
1608
|
Newman AM, Bratman SV, To J, Wynne JF, Eclov NCW, Modlin LA, Liu CL, Neal JW, Wakelee HA, Merritt RE, Shrager JB, Loo BW, Alizadeh AA, Diehn M. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat Med 2014; 20:548-54. [PMID: 24705333 PMCID: PMC4016134 DOI: 10.1038/nm.3519] [Citation(s) in RCA: 1592] [Impact Index Per Article: 144.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 11/06/2013] [Indexed: 02/06/2023]
Abstract
Circulating tumor DNA (ctDNA) represents a promising biomarker for noninvasive assessment of cancer burden, but existing methods have insufficient sensitivity or patient coverage for broad clinical applicability. Here we introduce CAncer Personalized Profiling by deep Sequencing (CAPP-Seq), an economical and ultrasensitive method for quantifying ctDNA. We implemented CAPP-Seq for non-small cell lung cancer (NSCLC) with a design covering multiple classes of somatic alterations that identified mutations in >95% of tumors. We detected ctDNA in 100% of stage II–IV and 50% of stage I NSCLC patients, with 96% specificity for mutant allele fractions down to ~0.02%. Levels of ctDNA significantly correlated with tumor volume, distinguished between residual disease and treatment-related imaging changes, and provided earlier response assessment than radiographic approaches. Finally, we explored biopsy-free tumor screening and genotyping with CAPP-Seq. We envision that CAPP-Seq could be routinely applied clinically to detect and monitor diverse malignancies, thus facilitating personalized cancer therapy.
Collapse
Affiliation(s)
- Aaron M Newman
- 1] Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA. [2] Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California, USA. [3]
| | - Scott V Bratman
- 1] Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA. [2] Department of Radiation Oncology, Stanford University, Stanford, California, USA. [3]
| | - Jacqueline To
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
| | - Jacob F Wynne
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
| | - Neville C W Eclov
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
| | - Leslie A Modlin
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
| | - Chih Long Liu
- 1] Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA. [2] Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | - Joel W Neal
- Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | - Heather A Wakelee
- Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | - Robert E Merritt
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Stanford School of Medicine, Stanford University, Stanford, California, USA
| | - Joseph B Shrager
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Stanford School of Medicine, Stanford University, Stanford, California, USA
| | - Billy W Loo
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
| | - Ash A Alizadeh
- 1] Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA. [2] Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California, USA. [3] Division of Hematology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | - Maximilian Diehn
- 1] Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA. [2] Department of Radiation Oncology, Stanford University, Stanford, California, USA. [3] Stanford Cancer Institute, Stanford University, Stanford, California, USA
| |
Collapse
|
1609
|
Bidard FC, Peeters DJ, Fehm T, Nolé F, Gisbert-Criado R, Mavroudis D, Grisanti S, Generali D, Garcia-Saenz JA, Stebbing J, Caldas C, Gazzaniga P, Manso L, Zamarchi R, de Lascoiti AF, De Mattos-Arruda L, Ignatiadis M, Lebofsky R, van Laere SJ, Meier-Stiegen F, Sandri MT, Vidal-Martinez J, Politaki E, Consoli F, Bottini A, Diaz-Rubio E, Krell J, Dawson SJ, Raimondi C, Rutten A, Janni W, Munzone E, Carañana V, Agelaki S, Almici C, Dirix L, Solomayer EF, Zorzino L, Johannes H, Reis-Filho JS, Pantel K, Pierga JY, Michiels S. Clinical validity of circulating tumour cells in patients with metastatic breast cancer: a pooled analysis of individual patient data. Lancet Oncol 2014; 15:406-14. [PMID: 24636208 DOI: 10.1016/s1470-2045(14)70069-5] [Citation(s) in RCA: 611] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND We aimed to assess the clinical validity of circulating tumour cell (CTC) quantification for prognostication of patients with metastatic breast cancer by undertaking a pooled analysis of individual patient data. METHODS We contacted 51 European centres and asked them to provide reported and unreported anonymised data for individual patients with metastatic breast cancer who participated in studies between January, 2003, and July, 2012. Eligible studies had participants starting a new line of therapy, data for progression-free survival or overall survival, or both, and CTC quantification by the CellSearch method at baseline (before start of new treatment). We used Cox regression models, stratified by study, to establish the association between CTC count and progression-free survival and overall survival. We used the landmark method to assess the prognostic value of CTC and serum marker changes during treatment. We assessed the added value of CTCs or serum markers to prognostic clinicopathological models in a resampling procedure using likelihood ratio (LR) χ(2) statistics. FINDINGS 17 centres provided data for 1944 eligible patients from 20 studies. 911 patients (46·9%) had a CTC count of 5 per 7·5 mL or higher at baseline, which was associated with decreased progression-free survival (hazard ratio [HR] 1·92, 95% CI 1·73-2·14, p<0·0001) and overall survival (HR 2·78, 95% CI 2·42-3·19, p<0·0001) compared with patients with a CTC count of less than 5 per 7·5 mL at baseline. Increased CTC counts 3-5 weeks after start of treatment, adjusted for CTC count at baseline, were associated with shortened progression-free survival (HR 1·85, 95% CI 1·48-2·32, p<0·0001) and overall survival (HR 2·26, 95% CI 1·68-3·03) as were increased CTC counts after 6-8 weeks (progression-free survival HR 2·20, 95% CI 1·66-2·90, p<0·0001; overall survival HR 2·91, 95% CI 2·01-4·23, p<0·0001). Survival prediction was significantly improved by addition of baseline CTC count to the clinicopathological models (progression-free survival LR 38·4, 95% CI 21·9-60·3, p<0·0001; overall survival LR 64·9, 95% CI 41·3-93·4, p<0·0001). This model was further improved by addition of CTC change at 3-5 weeks (progression-free survival LR 8·2, 95% CI 0·78-20·4, p=0·004; overall survival LR 11·5, 95% CI 2·6-25·1, p=0·0007) and at 6-8 weeks (progression-free survival LR 15·3, 95% CI 5·2-28·3; overall survival LR 14·6, 95% CI 4·0-30·6; both p<0·0001). Carcinoembryonic antigen and cancer antigen 15-3 concentrations at baseline and during therapy did not add significant information to the best baseline model. INTERPRETATION These data confirm the independent prognostic effect of CTC count on progression-free survival and overall survival. CTC count also improves the prognostication of metastatic breast cancer when added to full clinicopathological predictive models, whereas serum tumour markers do not. FUNDING Janssen Diagnostics, the Nuovo-Soldati foundation for cancer research.
Collapse
Affiliation(s)
- François-Clément Bidard
- Department of Medical Oncology and SIRIC, Institut Curie, Paris, France; Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Dieter J Peeters
- Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Antwerp, Belgium; University of Antwerp, Antwerp, Belgium
| | - Tanja Fehm
- Department of Gynecology and Obstetrics, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Franco Nolé
- Division of Medical Senology, European Institute of Oncology, Milan, Italy
| | | | | | - Salvatore Grisanti
- Department of Transfusion Medicine, Laboratory for Stem Cells Manipulation and Cryopreservation, AO Spedali Civili di Brescia, Brescia, Italy
| | | | - Jose A Garcia-Saenz
- Department of Oncology, Hospital Clinico San Carlos, Department of Medicine, University Complutense of Madrid, Spain
| | - Justin Stebbing
- Imperial College and Imperial College Healthcare NHS Trust, London, UK
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, UK; Cambridge Experimental Cancer Medicine Centre, Addenbrooke's Hospital, Cambridge University Hospital NHS Foundation Trust, and NIHR Cambridge Biomedical Research Centre, Cambridge, UK; Cambridge Breast Unit, Addenbrooke's Hospital, Cambridge University Hospital NHS Foundation Trust, and NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Paola Gazzaniga
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | | | | | | | - Leticia De Mattos-Arruda
- Val d'Hebron Institute of Oncology, Val d'Hebron University Hospital, and Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Michail Ignatiadis
- Department of Medical Oncology and Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Ronald Lebofsky
- Department of Medical Oncology and SIRIC, Institut Curie, Paris, France
| | - Steven J van Laere
- Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Antwerp, Belgium; University of Antwerp, Antwerp, Belgium; Department of Oncology, KU-Leuven, Leuven, Belgium
| | - Franziska Meier-Stiegen
- Department of Gynecology and Obstetrics, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Maria-Teresa Sandri
- Division of Laboratory Medicine, European Institute of Oncology, Milan, Italy
| | | | | | - Francesca Consoli
- Department of Transfusion Medicine, Laboratory for Stem Cells Manipulation and Cryopreservation, AO Spedali Civili di Brescia, Brescia, Italy
| | | | - Eduardo Diaz-Rubio
- Department of Oncology, Hospital Clinico San Carlos, Department of Medicine, University Complutense of Madrid, Spain
| | - Jonathan Krell
- Imperial College and Imperial College Healthcare NHS Trust, London, UK
| | - Sarah-Jane Dawson
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Cristina Raimondi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Annemie Rutten
- Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
| | | | - Elisabetta Munzone
- Division of Medical Senology, European Institute of Oncology, Milan, Italy
| | | | | | - Camillo Almici
- Department of Transfusion Medicine, Laboratory for Stem Cells Manipulation and Cryopreservation, AO Spedali Civili di Brescia, Brescia, Italy
| | - Luc Dirix
- Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Antwerp, Belgium; University of Antwerp, Antwerp, Belgium
| | | | - Laura Zorzino
- Division of Laboratory Medicine, European Institute of Oncology, Milan, Italy
| | - Helene Johannes
- International Drug Development Institute, Louvain-La-Neuve, Belgium
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Klaus Pantel
- Department of Tumor Biology, Center of Experimental Medicine, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jean-Yves Pierga
- Department of Medical Oncology and SIRIC, Institut Curie, Paris, France; Université Paris Descartes, Paris, France.
| | - Stefan Michiels
- Service de Biostatistique et d'Epidémiologie, Gustave Roussy, Université Paris-Sud, Villejuif, France
| |
Collapse
|
1610
|
Roth A, Khattra J, Yap D, Wan A, Laks E, Biele J, Ha G, Aparicio S, Bouchard-Côté A, Shah SP. PyClone: statistical inference of clonal population structure in cancer. Nat Methods 2014; 11:396-8. [PMID: 24633410 PMCID: PMC4864026 DOI: 10.1038/nmeth.2883] [Citation(s) in RCA: 707] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 01/31/2014] [Indexed: 12/25/2022]
Abstract
We introduce PyClone, a statistical model for inference of clonal population structures in cancers. PyClone is a Bayesian clustering method for grouping sets of deeply sequenced somatic mutations into putative clonal clusters while estimating their cellular prevalences and accounting for allelic imbalances introduced by segmental copy-number changes and normal-cell contamination. Single-cell sequencing validation demonstrates PyClone's accuracy.
Collapse
Affiliation(s)
- Andrew Roth
- Bioinformatics Graduate Program, University Of British Columbia, Vancouver, Canada
- Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
| | - Jaswinder Khattra
- Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
| | - Damian Yap
- Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
| | - Adrian Wan
- Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
| | - Emma Laks
- Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
| | - Justina Biele
- Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
| | - Gavin Ha
- Bioinformatics Graduate Program, University Of British Columbia, Vancouver, Canada
- Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
| | - Samuel Aparicio
- Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada
| | | | - Sohrab P. Shah
- Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada
| |
Collapse
|
1611
|
Eroglu Z, Fielder O, Somlo G. Analysis of circulating tumor cells in breast cancer. J Natl Compr Canc Netw 2014; 11:977-85. [PMID: 23946176 DOI: 10.6004/jnccn.2013.0118] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Over the past few decades, substantial progress has been made in the diagnosis and treatment of breast cancer. Early identification of relapsed and metastatic disease has been a primary focus of ongoing research. Circulating tumor cells (CTCs) are implicated as harbingers of metastases. With advances in detection technologies, CTCs offer the option for real-time liquid biopsies. Methods to identify CTCs in the bloodstream by physical or biochemical properties, although feasible, still require improvements to promote widespread, reproducible use. Sufficient data support enumeration and assessment of changes in the number of CTCs as prognostic indicators, but controversy around their predictive utility for selecting treatments remains. As the technology to detect CTCs and characterize their heterogeneous molecular profile evolves, additional information will likely be obtained to guide targeted and individualized therapies.
Collapse
Affiliation(s)
- Zeynep Eroglu
- From Departments of aMedical Oncology and Therapeutic Research and bHematology and Hematopoietic Cell Transplantation, City of Hope Cancer Center, Duarte, California
| | | | | |
Collapse
|
1612
|
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: 52] [Impact Index Per Article: 4.7] [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
|
1613
|
Mouliere F, El Messaoudi S, Pang D, Dritschilo A, Thierry AR. Multi-marker analysis of circulating cell-free DNA toward personalized medicine for colorectal cancer. Mol Oncol 2014; 8:927-41. [PMID: 24698732 DOI: 10.1016/j.molonc.2014.02.005] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 01/27/2014] [Accepted: 02/12/2014] [Indexed: 12/14/2022] Open
Abstract
Development of a Q-PCR-based assay for the high-performance analysis of circulating cell-free DNA (ccfDNA) requires good knowledge of its structure and size. In this work, we present the first visual determination of ccfDNA by Atomic Force Microscopy (AFM) on plasma samples from colorectal cancer (CRC) patients and healthy donors. In addition to the examination of fragment size distribution profile as performed by Q-PCR, this analysis confirms that ccfDNA is highly fragmented and that more than 80% of ccfDNA fragments in CRC plasma are below 145 bp. We adapted an Allele-Specific Blocker (ASB) Q-PCR to small ccfDNA fragments to determine simultaneously the total ccfDNA concentration, the presence of point mutation, the proportion of mutated allele, and a ccfDNA integrity index. The data validated analytically these four parameters in 124 CRC clinical samples and 71 healthy individuals. The multi-marker method, termed Intplex, enables sensitive and specific non-invasive analysis of tumor ccfDNA, which has great potential in terms of cost, quality control, and easy implementation in every clinical center laboratory.
Collapse
Affiliation(s)
- Florent Mouliere
- Institut de Recherche en Cancérologie de Montpellier, U896 Inserm, Montpellier, France
| | - Safia El Messaoudi
- Institut de Recherche en Cancérologie de Montpellier, U896 Inserm, Montpellier, France
| | - Dalong Pang
- Department of Radiation Oncology, Lombardi Cancer Center, Georgetown University, Washington DC, USA
| | - Anatoly Dritschilo
- Department of Radiation Oncology, Lombardi Cancer Center, Georgetown University, Washington DC, USA
| | - Alain R Thierry
- Institut de Recherche en Cancérologie de Montpellier, U896 Inserm, Montpellier, France.
| |
Collapse
|
1614
|
Siravegna G, Bardelli A. Minimal Residual Disease in Breast Cancer: In Blood Veritas. Clin Cancer Res 2014; 20:2505-7. [DOI: 10.1158/1078-0432.ccr-14-0370] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
1615
|
Vassy JL, Lautenbach DM, McLaughlin HM, Kong SW, Christensen KD, Krier J, Kohane IS, Feuerman LZ, Blumenthal-Barby J, Roberts JS, Lehmann LS, Ho CY, Ubel PA, MacRae CA, Seidman CE, Murray MF, McGuire AL, Rehm HL, Green RC. The MedSeq Project: a randomized trial of integrating whole genome sequencing into clinical medicine. Trials 2014; 15:85. [PMID: 24645908 PMCID: PMC4113228 DOI: 10.1186/1745-6215-15-85] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 02/28/2014] [Indexed: 11/28/2022] Open
Abstract
Background Whole genome sequencing (WGS) is already being used in certain clinical and research settings, but its impact on patient well-being, health-care utilization, and clinical decision-making remains largely unstudied. It is also unknown how best to communicate sequencing results to physicians and patients to improve health. We describe the design of the MedSeq Project: the first randomized trials of WGS in clinical care. Methods/Design This pair of randomized controlled trials compares WGS to standard of care in two clinical contexts: (a) disease-specific genomic medicine in a cardiomyopathy clinic and (b) general genomic medicine in primary care. We are recruiting 8 to 12 cardiologists, 8 to 12 primary care physicians, and approximately 200 of their patients. Patient participants in both the cardiology and primary care trials are randomly assigned to receive a family history assessment with or without WGS. Our laboratory delivers a genome report to physician participants that balances the needs to enhance understandability of genomic information and to convey its complexity. We provide an educational curriculum for physician participants and offer them a hotline to genetics professionals for guidance in interpreting and managing their patients’ genome reports. Using varied data sources, including surveys, semi-structured interviews, and review of clinical data, we measure the attitudes, behaviors and outcomes of physician and patient participants at multiple time points before and after the disclosure of these results. Discussion The impact of emerging sequencing technologies on patient care is unclear. We have designed a process of interpreting WGS results and delivering them to physicians in a way that anticipates how we envision genomic medicine will evolve in the near future. That is, our WGS report provides clinically relevant information while communicating the complexity and uncertainty of WGS results to physicians and, through physicians, to their patients. This project will not only illuminate the impact of integrating genomic medicine into the clinical care of patients but also inform the design of future studies. Trial registration ClinicalTrials.gov identifier
NCT01736566
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Robert C Green
- Genomes2People and Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Broad Institute and Harvard Medical School, 41 Avenue Louis Pasteur, Suite 301, 02115 Boston, MA, USA.
| | | |
Collapse
|
1616
|
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: 467] [Impact Index Per Article: 42.5] [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
|
1617
|
Song H, Nan Y, Cheng XW. Circulating cf-DNA: A promising, noninvasive tool for assessment of early cardio-metabolic risk. Atherosclerosis 2014; 233:307-9. [DOI: 10.1016/j.atherosclerosis.2013.11.081] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 11/12/2013] [Indexed: 11/30/2022]
|
1618
|
Tang C, Gao H, Li X, Liu Y, Li J, Qin H, Wang W, Qu L, An J, Yang S, Liu X. Different treatment orders achieved similar clinical results: a retrospective study for retreatment of epidermal growth factor receptor tyrosine kinase inhibitors in 120 patients with non-small-cell lung cancer. J Cancer Res Clin Oncol 2014; 140:427-33. [PMID: 24401995 DOI: 10.1007/s00432-014-1582-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 01/03/2014] [Indexed: 01/31/2023]
Abstract
BACKGROUND It was reported the retreatment of epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) may bring benefit to non-small-cell lung cancer (NSCLC) patients who benefited previously. Nevertheless, the treatment order in most of the prior literature was gefitinib (G) to erlotinib (E), and little was known about whether other treatment order may also bring benefit to the patients. METHODS One hundred and twenty NSCLC patients who received EGFR-TKIs treatment twice were enrolled in this study. The safety and effectiveness of the second EGFR-TKIs administration, as well as the influencing factors that contribute to this process, were analyzed retrospectively. RESULTS Forty-nine (40.8%) patients were retreated with same kind of EGFR-TKIs: 30 (25%) were G and 19 (15.8%) were E. Seventy-one (59.2%) patients switched to another kind: 55 (45.8%) were G to E and 16 (13.4%) were the reverse. Notably, no differences in clinical benefits were found among the four different treatment orders. For the second administration, the adverse effects of all patients were generally classified as grade I-II and the 1-year survival rate reached 32.5%. The objective response rate, disease control rate, median progression-free survival (PFS), and overall survival was 10.0% (12/120), 52.5% (63/120), 2.3 (95% CI 1.5-3.0) months and 8.0 (95% CI 7.0-8.5) months, respectively. The univariate and multivariate analyses revealed that those patients who benefited from prior EGFR-TKIs were easier to get benefit from the second administration, and the strongest beneficial indicators of the retreatment were PFS of the initial EGFR-TKIs (≥6 months, HR 0.611, 95% CI 0.354-0.901, P = 0.0076) and time interval between the two EGFR-TKIs treatment (≥4 months, HR 0.529, 95% CI 0.328-0.852, P = 0.0088). CONCLUSION Those patients who benefited from prior EGFR-TKIs were easier to get benefit from the second administration. A time interval of ≥4 months may improve the retreatment, but differences in clinical benefit were not found among different treatment orders. If the retrospective result could be validated further in the future, it would be helpful for rational administration of EGFR-TKIs.
Collapse
Affiliation(s)
- Chuanhao Tang
- Department of Lung Cancer, Affiliated Hospital of Academy of Military Medical Sciences, No. 8 Dongdajie, Beijing, 100071, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1619
|
Personalized ovarian cancer disease surveillance and detection of candidate therapeutic drug target in circulating tumor DNA. Neoplasia 2014. [PMID: 24563622 DOI: 10.1593/neo.131900] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Retrospective studies have demonstrated that nearly 50% of patients with ovarian cancer with normal cancer antigen 125 (CA125) levels have persistent disease; however, prospectively distinguishing between patients is currently impossible. Here, we demonstrate that for one patient, with the first reported fibroblast growth factor receptor 2 (FGFR2) fusion transcript in ovarian cancer, circulating tumor DNA (ctDNA) is a more sensitive and specific biomarker than CA125, and it can also inform on a candidate therapeutic. For a 4-year period, during which the patient underwent primary debulking surgery and chemotherapy, tumor recurrences, and multiple chemotherapeutic regimens, blood samples were longitudinally collected and stored. Whereas postsurgical CA125 levels were elevated only three times for 28 measurements, the FGFR2 fusion ctDNA biomarker was readily detectable by quantitative real-time reverse transcription-polymerase chain reaction (PCR) in all of these same blood samples and in the tumor recurrences. Given the persistence of the FGFR2 fusion, we treated tumor cells derived from this patient and others with the FGFR2 inhibitor BGJ398. Only tumor cells derived from this patient were sensitive to FGFR2 inhibitor treatment. Using the same methodologic approach, we demonstrate in a second patient with a different fusion that PCR and agarose gel electrophoresis can also be used to identify tumor-specific DNA in the circulation. Taken together, we demonstrate that a relatively inexpensive, PCR-based ctDNA surveillance assay can outperform CA125 in identifying occult disease.
Collapse
|
1620
|
[Personalized medicine and breast cancer: anticipatory medicine, prognostic evaluation and therapeutic targeting]. Bull Cancer 2014; 100:1295-310. [PMID: 24225763 DOI: 10.1684/bdc.2013.1856] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Breast cancer is now considered as a large collection of distinct biological entities, the management of which is increasingly personalized. Personalized medicine - defined as a medicine, which uses molecular profiles, notably genetic profiles, from patients and/or tumors to tailor therapeutic decisions - is now introduced in the management of breast cancer at any stages: screening and prevention of hereditary forms, prognostic and predictive evaluation of early breast cancer, and, more recently, novel clinical trials in advanced breast cancer, where genetic characterization of tumor tissue based on genomics, including next-generation sequencing tools, is used to drive specific therapeutic targeting.
Collapse
|
1621
|
Chen P, Huang YY, Hoshino K, Zhang X. Multiscale immunomagnetic enrichment of circulating tumor cells: from tubes to microchips. LAB ON A CHIP 2014; 14:446-58. [PMID: 24292816 DOI: 10.1039/c3lc51107c] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
We review the rare cancer cell sorting technologies, with a focus on multiscale immunomagnetic approaches. Starting from the conventional magnetic activated cell sorting system, we derive the scaling laws of immunomagnetic assay and justify the recent trend of using downscaled systems for CTC studies. Furthermore, we introduce recent work on combining the immunomagnetic assay with microfluidic technology for enhanced separation. We summarize different types of in-channel micro-magnetic structures that can further increase the local magnetic field without lowering the system throughput. Related design concepts, principles, and microfabrication techniques are presented and evaluated.
Collapse
Affiliation(s)
- Peng Chen
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, USA.
| | | | | | | |
Collapse
|
1622
|
Beaver JA, Jelovac D, Balukrishna S, Cochran R, Croessmann S, Zabransky DJ, Wong HY, Toro PV, Cidado J, Blair BG, Chu D, Burns T, Higgins MJ, Stearns V, Jacobs L, Habibi M, Lange J, Hurley PJ, Lauring J, VanDenBerg D, Kessler J, Jeter S, Samuels ML, Maar D, Cope L, Cimino-Mathews A, Argani P, Wolff AC, Park BH. Detection of cancer DNA in plasma of patients with early-stage breast cancer. Clin Cancer Res 2014; 20:2643-2650. [PMID: 24504125 DOI: 10.1158/1078-0432.ccr-13-2933] [Citation(s) in RCA: 301] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE Detecting circulating plasma tumor DNA (ptDNA) in patients with early-stage cancer has the potential to change how oncologists recommend systemic therapies for solid tumors after surgery. Droplet digital polymerase chain reaction (ddPCR) is a novel sensitive and specific platform for mutation detection. EXPERIMENTAL DESIGN In this prospective study, primary breast tumors and matched pre- and postsurgery blood samples were collected from patients with early-stage breast cancer (n = 29). Tumors (n = 30) were analyzed by Sanger sequencing for common PIK3CA mutations, and DNA from these tumors and matched plasma were then analyzed for PIK3CA mutations using ddPCR. RESULTS Sequencing of tumors identified seven PIK3CA exon 20 mutations (H1047R) and three exon 9 mutations (E545K). Analysis of tumors by ddPCR confirmed these mutations and identified five additional mutations. Presurgery plasma samples (n = 29) were then analyzed for PIK3CA mutations using ddPCR. Of the 15 PIK3CA mutations detected in tumors by ddPCR, 14 of the corresponding mutations were detected in presurgical ptDNA, whereas no mutations were found in plasma from patients with PIK3CA wild-type tumors (sensitivity 93.3%, specificity 100%). Ten patients with mutation-positive ptDNA presurgery had ddPCR analysis of postsurgery plasma, with five patients having detectable ptDNA postsurgery. CONCLUSIONS This prospective study demonstrates accurate mutation detection in tumor tissues using ddPCR, and that ptDNA can be detected in blood before and after surgery in patients with early-stage breast cancer. Future studies can now address whether ptDNA detected after surgery identifies patients at risk for recurrence, which could guide chemotherapy decisions for individual patients.
Collapse
Affiliation(s)
- Julia A Beaver
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287
| | - Danijela Jelovac
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287
| | | | - Rory Cochran
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287
| | - Sarah Croessmann
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287
| | - Daniel J Zabransky
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287
| | - Hong Yuen Wong
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287
| | - Patricia Valda Toro
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287
| | - Justin Cidado
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287
| | - Brian G Blair
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287
| | - David Chu
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287
| | - Timothy Burns
- University of Pittsburgh Hillman Cancer Center, Pittsburgh, PA 15213-1863
| | | | - Vered Stearns
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287
| | - Lisa Jacobs
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287
| | - Mehran Habibi
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287
| | - Julie Lange
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287
| | - Paula J Hurley
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287
| | - Josh Lauring
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287
| | - Dustin VanDenBerg
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287
| | - Jill Kessler
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287
| | - Stacie Jeter
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287
| | | | - Dianna Maar
- Bio-Rad Laboratories, Digital Biology Center, Pleasanton, CA 94566
| | - Leslie Cope
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287
| | | | - Pedram Argani
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287
| | - Antonio C Wolff
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287
| | - Ben H Park
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287
| |
Collapse
|
1623
|
Watson PH, Nussbeck SY, Carter C, O'Donoghue S, Cheah S, Matzke LAM, Barnes RO, Bartlett J, Carpenter J, Grizzle WE, Johnston RN, Mes-Masson AM, Murphy L, Sexton K, Shepherd L, Simeon-Dubach D, Zeps N, Schacter B. A framework for biobank sustainability. Biopreserv Biobank 2014; 12:60-8. [PMID: 24620771 PMCID: PMC4150367 DOI: 10.1089/bio.2013.0064] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Each year funding agencies and academic institutions spend millions of dollars and euros on biobanking. All funding providers assume that after initial investments biobanks should be able to operate sustainably. However the topic of sustainability is challenging for the discipline of biobanking for several major reasons: the diversity in the biobanking landscape, the different purposes of biobanks, the fact that biobanks are dissimilar to other research infrastructures and the absence of universally understood or applicable value metrics for funders and other stakeholders. In this article our aim is to delineate a framework to allow more effective discussion and action around approaches for improving biobank sustainability. The term sustainability is often used to mean fiscally self-sustaining, but this restricted definition is not sufficient for biobanking. Instead we propose that biobank sustainability should be considered within a framework of three dimensions - financial, operational, and social. In each dimension, areas of focus or elements are identified that may allow different types of biobanks to distinguish and evaluate the relevance, likelihood, and impact of each element, as well as the risks to the biobank of failure to address them. Examples of practical solutions, tools and strategies to address biobank sustainability are also discussed.
Collapse
Affiliation(s)
- Peter H Watson
- 1 University of British Columbia Office of Biobank Education and Research , Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1624
|
Antiviral signaling protein MITA acts as a tumor suppressor in breast cancer by regulating NF-κB induced cell death. Biochim Biophys Acta Mol Basis Dis 2014; 1842:144-53. [DOI: 10.1016/j.bbadis.2013.11.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Revised: 10/26/2013] [Accepted: 11/07/2013] [Indexed: 12/18/2022]
|
1625
|
Wan L, Pantel K, Kang Y. Tumor metastasis: moving new biological insights into the clinic. Nat Med 2014; 19:1450-64. [PMID: 24202397 DOI: 10.1038/nm.3391] [Citation(s) in RCA: 598] [Impact Index Per Article: 54.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 10/04/2013] [Indexed: 02/07/2023]
Abstract
As the culprit behind most cancer-related deaths, metastasis is the ultimate challenge in our effort to fight cancer as a life-threatening disease. The explosive growth of metastasis research in the past decade has yielded an unprecedented wealth of information about the tumor-intrinsic and tumor-extrinsic mechanisms that dictate metastatic behaviors, the molecular and cellular basis underlying the distinct courses of metastatic progression in different cancers and what renders metastatic cancer refractory to available therapies. However, integration of such new knowledge into an improved, metastasis-oriented oncological drug development strategy is needed to thwart the development of metastatic disease at every stage of progression.
Collapse
Affiliation(s)
- Liling Wan
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | | | | |
Collapse
|
1626
|
Abstract
Genotyping tumor tissue in search of somatic genetic alterations for actionable information has become routine practice in clinical oncology. Although these sequence alterations are highly informative, sampling tumor tissue has significant inherent limitations; tumor tissue is a single snapshot in time, is subject to selection bias resulting from tumor heterogeneity, and can be difficult to obtain. Cell-free fragments of DNA are shed into the bloodstream by cells undergoing apoptosis or necrosis, and the load of circulating cell-free DNA (cfDNA) correlates with tumor staging and prognosis. Moreover, recent advances in the sensitivity and accuracy of DNA analysis have allowed for genotyping of cfDNA for somatic genomic alterations found in tumors. The ability to detect and quantify tumor mutations has proven effective in tracking tumor dynamics in real time as well as serving as a liquid biopsy that can be used for a variety of clinical and investigational applications not previously possible.
Collapse
Affiliation(s)
- Luis A Diaz
- Luis A. Diaz Jr, Swim Across America Laboratory and Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of Medicine, Baltimore, MD; and Alberto Bardelli, Institute for Cancer Research and Treatment at Candiolo, University of Torino, Candiolo, and the Fondazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milan, Italy
| | | |
Collapse
|
1627
|
Oxnard GR, Paweletz CP, Kuang Y, Mach SL, O'Connell A, Messineo MM, Luke JJ, Butaney M, Kirschmeier P, Jackman DM, Jänne PA. Noninvasive detection of response and resistance in EGFR-mutant lung cancer using quantitative next-generation genotyping of cell-free plasma DNA. Clin Cancer Res 2014; 20:1698-1705. [PMID: 24429876 DOI: 10.1158/1078-0432.ccr-13-2482] [Citation(s) in RCA: 628] [Impact Index Per Article: 57.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSE Tumor genotyping using cell-free plasma DNA (cfDNA) has the potential to allow noninvasive assessment of tumor biology, yet many existing assays are cumbersome and vulnerable to false-positive results. We sought to determine whether droplet digital PCR (ddPCR) of cfDNA would allow highly specific and quantitative assessment of tumor genotype. EXPERIMENTAL DESIGN ddPCR assays for EGFR, KRAS, and BRAF mutations were developed using plasma collected from patients with advanced lung cancer or melanoma of a known tumor genotype. Sensitivity and specificity were determined using cancers with nonoverlapping genotypes as positive and negative controls. Serial assessment of response and resistance was studied in patients with EGFR-mutant lung cancer on a prospective trial of erlotinib. RESULTS We identified a reference range for EGFR L858R and exon 19 deletions in specimens from KRAS-mutant lung cancer, allowing identification of candidate thresholds with high sensitivity and 100% specificity. Received operative characteristic curve analysis of four assays demonstrated an area under the curve in the range of 0.80 to 0.94. Sensitivity improved in specimens with optimal cfDNA concentrations. Serial plasma genotyping of EGFR-mutant lung cancer on erlotinib demonstrated pretreatment detection of EGFR mutations, complete plasma response in most cases, and increasing levels of EGFR T790M emerging before objective progression. CONCLUSIONS Noninvasive genotyping of cfDNA using ddPCR demonstrates assay qualities that could allow effective translation into a clinical diagnostic. Serial quantification of plasma genotype allows noninvasive assessment of response and resistance, including detection of resistance mutations up to 16 weeks before radiographic progression.
Collapse
Affiliation(s)
- Geoffrey R Oxnard
- Department of Medical Oncology, Brigham and Women's Hospital & Harvard Medical School Boston, MA.,Department of Medicine, Brigham and Women's Hospital & Harvard Medical School Boston, MA
| | - Cloud P Paweletz
- Department of Medical Oncology, Brigham and Women's Hospital & Harvard Medical School Boston, MA.,Belfer Institute for Applied Cancer Science, Dana-Farber Cancer Institute, Brigham and Women's Hospital & Harvard Medical School Boston, MA
| | - Yanan Kuang
- Department of Medical Oncology, Brigham and Women's Hospital & Harvard Medical School Boston, MA.,Belfer Institute for Applied Cancer Science, Dana-Farber Cancer Institute, Brigham and Women's Hospital & Harvard Medical School Boston, MA
| | - Stacy L Mach
- Department of Medical Oncology, Brigham and Women's Hospital & Harvard Medical School Boston, MA
| | - Allison O'Connell
- Department of Medical Oncology, Brigham and Women's Hospital & Harvard Medical School Boston, MA.,Belfer Institute for Applied Cancer Science, Dana-Farber Cancer Institute, Brigham and Women's Hospital & Harvard Medical School Boston, MA
| | - Melissa M Messineo
- Department of Medical Oncology, Brigham and Women's Hospital & Harvard Medical School Boston, MA.,Belfer Institute for Applied Cancer Science, Dana-Farber Cancer Institute, Brigham and Women's Hospital & Harvard Medical School Boston, MA
| | - Jason J Luke
- Department of Medical Oncology, Brigham and Women's Hospital & Harvard Medical School Boston, MA.,Department of Medicine, Brigham and Women's Hospital & Harvard Medical School Boston, MA
| | - Mohit Butaney
- Department of Medical Oncology, Brigham and Women's Hospital & Harvard Medical School Boston, MA
| | - Paul Kirschmeier
- Department of Medical Oncology, Brigham and Women's Hospital & Harvard Medical School Boston, MA.,Belfer Institute for Applied Cancer Science, Dana-Farber Cancer Institute, Brigham and Women's Hospital & Harvard Medical School Boston, MA
| | - David M Jackman
- Department of Medical Oncology, Brigham and Women's Hospital & Harvard Medical School Boston, MA.,Department of Medicine, Brigham and Women's Hospital & Harvard Medical School Boston, MA
| | - Pasi A Jänne
- Department of Medical Oncology, Brigham and Women's Hospital & Harvard Medical School Boston, MA.,Belfer Institute for Applied Cancer Science, Dana-Farber Cancer Institute, Brigham and Women's Hospital & Harvard Medical School Boston, MA.,Department of Medicine, Brigham and Women's Hospital & Harvard Medical School Boston, MA
| |
Collapse
|
1628
|
Abstract
For a decade, the technologies behind DNA sequencing have improved rapidly in cost reduction and speed. Sequencing in large populations of cancer patients is leading to dramatic advances in our understanding of the cancer genome. The wide variety of cancer types sequenced and analyzed using these technologies has revealed many novel fundamental genetic mechanisms driving cancer initiation, progression, and maintenance. We have deepened our understanding of the signaling pathways, demonstrating disruption in epigenetic regulation and destabilization of the splicing machinery. The molecular mechanisms of resistance to targeted therapies are being elucidated for the first time. The translation of genome-scale variation into clinically actionable information is still in its infancy; nevertheless, insights from sequencing studies have led to the discovery of a variety of novel diagnostic biomarkers and therapeutic targets. Here, we review recent advances in cancer genomics and discuss what the new findings have taught us about cancer biology and, more importantly, how these new findings guide more effective diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Linghua Wang
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030
| | - David A. Wheeler
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
1629
|
Breast cancer in 2013: Genomics, drug approval, and optimal treatment duration. Nat Rev Clin Oncol 2014; 11:71-2. [PMID: 24419301 DOI: 10.1038/nrclinonc.2013.250] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
1630
|
Affiliation(s)
- Costanza Paoletti
- Breast Oncology Program of the Comprehensive Cancer Center, and the Department of Internal Medicine, University of Michigan Health and Hospital System, Ann Arbor, Michigan 48109;
| | - Daniel F. Hayes
- Breast Oncology Program of the Comprehensive Cancer Center, and the Department of Internal Medicine, University of Michigan Health and Hospital System, Ann Arbor, Michigan 48109;
| |
Collapse
|
1631
|
McCarthy JJ, McLeod HL, Ginsburg GS. Genomic medicine: a decade of successes, challenges, and opportunities. Sci Transl Med 2014; 5:189sr4. [PMID: 23761042 DOI: 10.1126/scitranslmed.3005785] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Genomic medicine--an aspirational term 10 years ago--is gaining momentum across the entire clinical continuum from risk assessment in healthy individuals to genome-guided treatment in patients with complex diseases. We review the latest achievements in genome research and their impact on medicine, primarily in the past decade. In most cases, genomic medicine tools remain in the realm of research, but some tools are crossing over into clinical application, where they have the potential to markedly alter the clinical care of patients. In this State of the Art Review, we highlight notable examples including the use of next-generation sequencing in cancer pharmacogenomics, in the diagnosis of rare disorders, and in the tracking of infectious disease outbreaks. We also discuss progress in dissecting the molecular basis of common diseases, the role of the host microbiome, the identification of drug response biomarkers, and the repurposing of drugs. The significant challenges of implementing genomic medicine are examined, along with the innovative solutions being sought. These challenges include the difficulty in establishing clinical validity and utility of tests, how to increase awareness and promote their uptake by clinicians, a changing regulatory and coverage landscape, the need for education, and addressing the ethical aspects of genomics for patients and society. Finally, we consider the future of genomics in medicine and offer a glimpse of the forces shaping genomic medicine, such as fundamental shifts in how we define disease, how medicine is delivered to patients, and how consumers are managing their own health and affecting change.
Collapse
Affiliation(s)
- Jeanette J McCarthy
- Institute for Genome Sciences & Policy, Duke University, Durham, NC 27708, USA
| | | | | |
Collapse
|
1632
|
Patel JD, Krilov L, Adams S, Aghajanian C, Basch E, Brose MS, Carroll WL, de Lima M, Gilbert MR, Kris MG, Marshall JL, Masters GA, O'Day SJ, Polite B, Schwartz GK, Sharma S, Thompson I, Vogelzang NJ, Roth BJ. Clinical Cancer Advances 2013: Annual Report on Progress Against Cancer From the American Society of Clinical Oncology. J Clin Oncol 2014; 32:129-60. [DOI: 10.1200/jco.2013.53.7076] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
A MESSAGE FROM ASCO'S PRESIDENTSince its founding in 1964, the American Society of Clinical Oncology (ASCO) has been committed to improving cancer outcomes through research and the delivery of quality care. Research is the bedrock of discovering better treatments—providing hope to the millions of individuals who face a cancer diagnosis each year.The studies featured in “Clinical Cancer Advances 2013: Annual Report on Progress Against Cancer From the American Society of Clinical Oncology” represent the invaluable contributions of thousands of patients who participate in clinical trials and the scientists who conduct basic and clinical research. The insights described in this report, such as how cancers hide from the immune system and why cancers may become resistant to targeted drugs, enable us to envision a future in which cancer will be even more controllable and preventable.The scientific process is thoughtful, deliberate, and sometimes slow, but each advance, while helping patients, now also points toward new research questions and unexplored opportunities. Both dramatic and subtle breakthroughs occur so that progress against cancer typically builds over many years. Success requires vision, persistence, and a long-term commitment to supporting cancer research and training.Our nation's longstanding investment in federally funded cancer research has contributed significantly to a growing array of effective new treatments and a much deeper understanding of the drivers of cancer. But despite this progress, our position as a world leader in advancing medical knowledge and our ability to attract the most promising and talented investigators are now threatened by an acute problem: Federal funding for cancer research has steadily eroded over the past decade, and only 15% of the ever-shrinking budget is actually spent on clinical trials. This dismal reality threatens the pace of progress against cancer and undermines our ability to address the continuing needs of our patients.Despite this extremely challenging economic environment, we continue to make progress. Maintaining and accelerating that progress require that we keep our eyes on the future and pursue a path that builds on the stunning successes of the past. We must continue to show our policymakers the successes in cancer survival and quality of life (QOL) they have enabled, emphasizing the need to sustain our national investment in the remarkably productive US cancer research enterprise.We must also look to innovative methods for transforming how we care for—and learn from—patients with cancer. Consider, for example, that fewer than 5% of adult patients with cancer currently participate in clinical trials. What if we were able to draw lessons from the other 95%? This possibility led ASCO this year to launch CancerLinQ, a groundbreaking health information technology initiative that will provide physicians with access to vast quantities of clinical data about real-world patients and help achieve higher quality, higher value cancer care.As you read the following pages, I hope our collective progress against cancer over the past year inspires you. More importantly, I hope the pride you feel motivates you to help us accelerate the pace of scientific advancement.Clifford A. Hudis, MD, FACPPresidentAmerican Society of Clinical Oncology
Collapse
Affiliation(s)
- Jyoti D. Patel
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Lada Krilov
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Sylvia Adams
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Carol Aghajanian
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Ethan Basch
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Marcia S. Brose
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - William L. Carroll
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Marcos de Lima
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Mark R. Gilbert
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Mark G. Kris
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - John L. Marshall
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Gregory A. Masters
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Steven J. O'Day
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Blasé Polite
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Gary K. Schwartz
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Sunil Sharma
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Ian Thompson
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Nicholas J. Vogelzang
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Bruce J. Roth
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| |
Collapse
|
1633
|
Turker I, Uyeturk U, Sonmez OU, Oksuzoglu B, Helvaci K, Arslan UY, Budakoglu B, Alkis N, Aksoy S, Zengin N. Detection of circulating tumor cells in breast cancer patients: prognostic predictive role. Asian Pac J Cancer Prev 2014; 14:1601-7. [PMID: 23679243 DOI: 10.7314/apjcp.2013.14.3.1601] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
A determination of circulating tumor cell (CTC) effectiveness for prediction of progression-free survival (PFS) and overall survival (OS) was conducted as an adjunct to standard treatment of care in breast cancer management. Between November 2008 and March 2009, 22 metastatic and 12 early stage breast carcinoma patients, admitted to Ankara Oncology Training and Research Hospital, were included in this prospective trial. Patients' characteristics, treatment schedules and survival data were evaluated. CTC was detected twice by CellSearch method before and 9-12 weeks after the initiation of chemotherapy. A cut-off value equal or greater than 5 cells per 7.5 ml blood sample was considered positive. All patients were female. Median ages were 48.0 (range: 29-65) and 52.5 (range: 35-66) in early stage and metastatic subgroups, respectively. CTC was positive in 3 (13.6%) patients before chemotherapy and 6 (27.3%) patients during chemotherapy in the metastatic subgroup whereas positive in only one patient in the early stage subgroup before and during chemotherapy. The median follow-up was 22.0 (range: 21-23) and 19.0 (range: 5-23) months in the early stage and metastatic groups, respectively. In the metastatic group, both median PFS and OS were significantly shorter in any time CTC positive patients compared to CTC negative patients (PFS: 4.0 vs 14.0 months, Log-Rank p=0.013; and OS: 8.0 months vs. 20.5 months, Log-Rank p<0.001). OS was affected from multiple visceral metastatic sites (p=0.055) and higher grade (p=0.044) besides CTC positivity (log rank p<0.001). Radiological response of chemotherapy was also correlated with better survival (p<0.001). As a result, CTC positivity was confirmed as a prospective marker even in a small patient population, in this single center study. Measurement of CTC by CellSearch method in metastatic breast carcinoma cases may allow indications of early risk of relapse or death with even as few as two measurements during a chemotherapy program, but this finding should be confirmed with prospective trials in larger study populations.
Collapse
Affiliation(s)
- Ibrahim Turker
- Medical Oncology Department, Ankara Oncology Training and Research Hospital, Ankara, Turkey.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1634
|
Shupe MP, Graham LJ, Schneble EJ, Flynt FL, Clemenshaw MN, Kirkpatrick AD, Stojadinovic A, Peoples GE, Shumway NM. Future directions for monitoring treatment responses in breast cancer. J Cancer 2014; 5:69-78. [PMID: 24396499 PMCID: PMC3881222 DOI: 10.7150/jca.7048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Accepted: 07/24/2013] [Indexed: 11/05/2022] Open
Abstract
In the prior review, we outlined the current standard of care for monitoring treatment responses in breast cancer and discussed the many challenges associated with these strategies. We described the challenges faced in common clinical settings such as the adjuvant setting, neoadjuvant setting, and the metastatic setting. In this review, we will expand upon future directions meant to overcome several of these current challenges. We will also explore several new and promising methods under investigation to enhance how we monitor treatment responses in breast cancer. Furthermore, we will highlight several new technologies and techniques for monitoring breast cancer treatment in the adjuvant, neoadjuvant and metastatic setting.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Alexander Stojadinovic
- 5. Department of Surgical Oncology, San Antonio Military Medical Center (SAMMC), Brooke Army Medical Center, 3551 Roger Brooke Dr., Ft. Sam Houston, TX 78234, USA
| | - George E Peoples
- 6. Department of Surgical Oncology, Walter Reid National Military Medical Center, 8901 Rockville Pike, Bethesda, MD 20889, USA
| | | |
Collapse
|
1635
|
Gu J, Taylor CR, Phil D. Practicing pathology in the era of big data and personalized medicine. Appl Immunohistochem Mol Morphol 2014; 22:1-9. [PMID: 24326463 PMCID: PMC4206549 DOI: 10.1097/pai.0000000000000022] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The traditional task of the pathologist is to assist physicians in making the correct diagnosis of diseases at the earliest possible stage to effectuate the optimal treatment strategy for each individual patient. In this respect surgical pathology (the traditional tissue diagnosis) is but a tool. It is not, of itself, the purpose of pathology practice; and change is in the air. This January 2014 issue of Applied Immunohistochemistry and Molecular Morphology (AIMM) embraces that change by the incorporation of the agenda and content of the journal Diagnostic Molecular Morphology (DMP). Over a decade ago AIMM introduced and promoted the concept of "molecular morphology," and has sought to publish molecular studies that correlate with the morphologic features that continue to define cancer and many diseases. That intent is now reinforced and extended by the merger with DMP, as a logical and timely response to the growing impact of a wide range of genetic and molecular technologies that are beginning to reshape the way in which pathology is practiced. The use of molecular and genomic techniques already demonstrates clear value in the diagnosis of disease, with treatment tailored specifically to individual patients. Personalized medicine is the future, and personalized medicine demands personalized pathology. The need for integration of the flood of new molecular data, with surgical pathology, digital pathology, and the full range of pathology data in the electronic medical record has never been greater. This review describes the possible impact of these pressures upon the discipline of pathology, and examines possible outcomes. There is a sense of excitement and adventure. Active adaption and innovation are required. The new AIMM, incorporating DMP, seeks to position itself for a central role in this process.
Collapse
Affiliation(s)
- Jiang Gu
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | | | - D Phil
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| |
Collapse
|
1636
|
Weaver JMJ, Ross-Innes CS, Fitzgerald RC. The '-omics' revolution and oesophageal adenocarcinoma. Nat Rev Gastroenterol Hepatol 2014; 11:19-27. [PMID: 23982683 DOI: 10.1038/nrgastro.2013.150] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Oesophageal adenocarcinoma (OAC) is the eighth most common cancer type worldwide with a dismal 5-year survival. Barrett oesophagus, the replacement of the normal squamous epithelia with glandular cells, is the first step in the pathway towards OAC. Although most patients with OAC present de novo, the presence of the easily detectable OAC precursor lesion, Barrett oesophagus, enables the possibility of early detection of high-risk patients who are more likely to progress. Currently, identification of high-risk patients depends on histopathological assessment of dysplasia with no regards to molecular pathogenesis. In the future, screening and risk stratification initiatives for Barrett oesophagus that incorporate molecular profiles might permit improved early diagnosis and intervention strategies with the possibility of preventing OAC. For the majority of patients presenting de novo at an advanced stage, combining so-called -omics datasets with current clinical staging algorithms might enable OACs to be better classified according to distinct molecular programmes, thereby leading to better targeted treatment strategies as well as cancer monitoring regimes. This Review discusses how the latest advances in -omics technologies have improved our understanding of the development and biology of OAC, and how this development might alter patient management in the future.
Collapse
Affiliation(s)
- Jamie M J Weaver
- MRC Cancer Cell Unit, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, UK
| | - Caryn S Ross-Innes
- MRC Cancer Cell Unit, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, UK
| | - Rebecca C Fitzgerald
- MRC Cancer Cell Unit, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, UK
| |
Collapse
|
1637
|
Wong HY, Park BH. Plasma tumor DNA: on your markers, get set, go! ANNALS OF TRANSLATIONAL MEDICINE 2014; 2:2. [PMID: 25332978 PMCID: PMC4200658 DOI: 10.3978/j.issn.2305-5839.2013.06.07] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 06/24/2013] [Indexed: 12/13/2022]
Abstract
Metastatic breast cancer is incurable, yet highly treatable with endocrine, HER2 directed and chemotherapies improving survival for many patients. Successful treatment depends on the ability to monitor disease burden and response to therapies. Recently, a proof of principle study has shown that plasma tumor DNA (ptDNA) can be used as a reliable breast cancer biomarker in metastatic disease, due to its sensitivity and wide dynamic range. ptDNA more accurately reflects changes in response to therapies, and absolute levels of ptDNA demonstrate prognostic significance. Thus, ptDNA as a liquid biopsy shows great promise in the clinical management of metastatic breast cancer though further technical challenges and larger confirmatory studies are needed.
Collapse
|
1638
|
Shaw JA, Stebbing J. Circulating free DNA in the management of breast cancer. ANNALS OF TRANSLATIONAL MEDICINE 2014; 2:3. [PMID: 25332979 PMCID: PMC4200656 DOI: 10.3978/j.issn.2305-5839.2013.06.06] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 06/24/2013] [Indexed: 01/05/2023]
Abstract
Circulating cell-free DNA (cfDNA) is now under investigation as a "liquid biopsy" in the real time management of cancer. In The New England Journal of Medicine Dawson et al. reported a proof of concept investigation of tumour specific alterations in cfDNA and demonstrate that this fraction termed "circulating tumour DNA" (ctDNA) shows greater correlation with changes in tumour burden than two other circulating biomarkers (CA 15-3 and circulating tumor cell counts) in individuals with metastatic breast cancer receiving therapy.
Collapse
|
1639
|
Lee SH, Sim SH, Kim JY, Cha S, Song A. Application of cancer genomics to solve unmet clinical needs. Genomics Inform 2013; 11:174-9. [PMID: 24465227 PMCID: PMC3897843 DOI: 10.5808/gi.2013.11.4.174] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 11/12/2013] [Accepted: 11/15/2013] [Indexed: 12/12/2022] Open
Abstract
The large amount of data on cancer genome research has contributed to our understanding of cancer biology. Indeed, the genomics approach has a strong advantage for analyzing multi-factorial and complicated problems, such as cancer. It is time to think about the actual usage of cancer genomics in the clinical field. The clinical cancer field has lots of unmet needs in the management of cancer patients, which has been defined in the pre-genomic era. Unmet clinical needs are not well known to bioinformaticians and even non-clinician cancer scientists. A personalized approach in the clinical field will bring potential additional challenges to cancer genomics, because most data to now have been population-based rather than individual-based. We can maximize the use of cancer genomics in the clinical field if cancer scientists, bioinformaticians, and clinicians think and work together in solving unmet clinical needs. In this review, we present one imaginary case of a cancer patient, with which we can think about unmet clinical needs to solve with cancer genomics in the diagnosis, prediction of prognosis, monitoring the status of cancer, and personalized treatment decision.
Collapse
Affiliation(s)
- Se-Hoon Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 110-799, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Sung Hoon Sim
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Ji-Yeon Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Soojin Cha
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Ahnah Song
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 110-799, Korea
| |
Collapse
|
1640
|
Ni X, Zhuo M, Su Z, Duan J, Gao Y, Wang Z, Zong C, Bai H, Chapman AR, Zhao J, Xu L, An T, Ma Q, Wang Y, Wu M, Sun Y, Wang S, Li Z, Yang X, Yong J, Su XD, Lu Y, Bai F, Xie XS, Wang J. Reproducible copy number variation patterns among single circulating tumor cells of lung cancer patients. Proc Natl Acad Sci U S A 2013; 110:21083-21088. [PMID: 24324171 PMCID: PMC3876226 DOI: 10.1073/pnas.1320659110] [Citation(s) in RCA: 322] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Circulating tumor cells (CTCs) enter peripheral blood from primary tumors and seed metastases. The genome sequencing of CTCs could offer noninvasive prognosis or even diagnosis, but has been hampered by low single-cell genome coverage of scarce CTCs. Here, we report the use of the recently developed multiple annealing and looping-based amplification cycles for whole-genome amplification of single CTCs from lung cancer patients. We observed characteristic cancer-associated single-nucleotide variations and insertions/deletions in exomes of CTCs. These mutations provided information needed for individualized therapy, such as drug resistance and phenotypic transition, but were heterogeneous from cell to cell. In contrast, every CTC from an individual patient, regardless of the cancer subtypes, exhibited reproducible copy number variation (CNV) patterns, similar to those of the metastatic tumor of the same patient. Interestingly, different patients with the same lung cancer adenocarcinoma (ADC) shared similar CNV patterns in their CTCs. Even more interestingly, patients of small-cell lung cancer have CNV patterns distinctly different from those of ADC patients. Our finding suggests that CNVs at certain genomic loci are selected for the metastasis of cancer. The reproducibility of cancer-specific CNVs offers potential for CTC-based cancer diagnostics.
Collapse
Affiliation(s)
- Xiaohui Ni
- Biodynamic Optical Imaging Center, School of Life Sciences, Peking University, Beijing 100871, China
- Department of Chemistry and Chemical Biology
| | | | - Zhe Su
- Biodynamic Optical Imaging Center, School of Life Sciences, Peking University, Beijing 100871, China
| | | | - Yan Gao
- Biodynamic Optical Imaging Center, School of Life Sciences, Peking University, Beijing 100871, China
| | - Zhijie Wang
- Departments of Thoracic Medical Oncology and
| | | | - Hua Bai
- Departments of Thoracic Medical Oncology and
| | - Alec R. Chapman
- Department of Chemistry and Chemical Biology
- Program in Biophysics, Harvard University, Cambridge, MA 02138; and
| | - Jun Zhao
- Departments of Thoracic Medical Oncology and
| | - Liya Xu
- Biodynamic Optical Imaging Center, School of Life Sciences, Peking University, Beijing 100871, China
| | - Tongtong An
- Departments of Thoracic Medical Oncology and
| | - Qi Ma
- Biodynamic Optical Imaging Center, School of Life Sciences, Peking University, Beijing 100871, China
| | - Yuyan Wang
- Departments of Thoracic Medical Oncology and
| | - Meina Wu
- Departments of Thoracic Medical Oncology and
| | | | | | | | | | - Jun Yong
- Department of Chemistry and Chemical Biology
| | - Xiao-Dong Su
- Biodynamic Optical Imaging Center, School of Life Sciences, Peking University, Beijing 100871, China
| | - Youyong Lu
- Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Fan Bai
- Biodynamic Optical Imaging Center, School of Life Sciences, Peking University, Beijing 100871, China
| | - X. Sunney Xie
- Biodynamic Optical Imaging Center, School of Life Sciences, Peking University, Beijing 100871, China
- Department of Chemistry and Chemical Biology
| | - Jie Wang
- Departments of Thoracic Medical Oncology and
| |
Collapse
|
1641
|
Friedlander TW, Premasekharan G, Paris PL. Looking back, to the future of circulating tumor cells. Pharmacol Ther 2013; 142:271-80. [PMID: 24362084 DOI: 10.1016/j.pharmthera.2013.12.011] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 11/27/2013] [Indexed: 12/12/2022]
Abstract
Detection and analysis of circulating tumor cells (CTCs) from patients with metastatic malignancies have become active areas of research in recent years. CTC enumeration has already proven useful in establishing prognosis for patients with metastatic breast, colon, and prostate cancer. More recently, studies are going beyond enumeration, exploring the CTCs as a means to better understand the mechanisms of tumorigenesis, invasion, and metastasis and the value of CTC characterization for prognosis and tailoring of treatment. Analysis of CTC subpopulations, for example, is highlighting the importance of the epithelial to mesenchymal transition (EMT), a process which may be crucial for allowing tumors to invade into and grow at sites distant from the original tumor site. Similarly, the detection of CTCs expressing markers of stemness may also have important implications for treatment resistance. Genomic analysis of CTC and CTC subpopulations may allow for selection of novel therapeutic targets to combat treatment resistance. CTCs become a particularly valuable biospecimen resource when tissue biopsies are unavailable or not feasible and liquid biopsies allow for serial monitoring. Lastly, cultures of patient-derived CTCs may allow for an evaluation of therapeutic strategies performed ex vivo and in real time. This review article will focus on these developments, starting with the CTC pathogenesis, going on to discuss the different platforms available for CTC isolation and their use to date in these arenas, then will explore multiple topics including the existing data concerning CTC subpopulations and their clinical relevance, genomic characterization, and lastly, avenues for future research.
Collapse
Affiliation(s)
- Terence W Friedlander
- Division of Hematology & Medical Oncology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, United States.
| | - Gayatri Premasekharan
- Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, United States
| | - Pamela L Paris
- Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, United States
| |
Collapse
|
1642
|
Capovilla M. [Cellular and molecular mechanisms of carcinogenic side effects and resistance to BRAF inhibitors in metastatic melanoma with BRAFV600 mutation: state of the knowledge]. Ann Pathol 2013; 33:375-85. [PMID: 24331719 DOI: 10.1016/j.annpat.2013.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 09/04/2013] [Indexed: 01/07/2023]
Abstract
Cutaneous melanoma is a malignant tumor with a high metastatic potential. If an early treatment is associated with a favorable outcome, the prognosis of metastatic melanoma remains poor. Advances in molecular characterization of cancers, notably the discovery of BRAF gene mutations in metastatic melanoma, allowed to the recent development of targeted therapies against mutated BRAF protein. Despite high tumor response rates observed in clinical trials, these new drugs are associated with frequent secondary tumor resistance occurrence and paradoxical carcinogenic side effects. The cellular and molecular mechanisms of these carcinogenic side effects and secondary resistance are not yet fully elucidated and are actually intensely studied. This review of the literature focus on the mechanisms of these carcinogenic side effects and on the tumor resistance associated with anti-BRAF targeted therapies.
Collapse
MESH Headings
- Antineoplastic Agents/adverse effects
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Carcinoma, Squamous Cell/chemically induced
- Cell Transformation, Neoplastic/drug effects
- Drug Resistance, Neoplasm/genetics
- Enzyme Activation/drug effects
- Epigenesis, Genetic
- Gene Expression Regulation, Neoplastic
- Genes, ras
- Humans
- Indoles/adverse effects
- Indoles/pharmacology
- Indoles/therapeutic use
- Intercellular Signaling Peptides and Proteins/metabolism
- Keratoacanthoma/chemically induced
- Leukemia/chemically induced
- MAP Kinase Signaling System/drug effects
- Melanoma/chemically induced
- Melanoma/drug therapy
- Melanoma/genetics
- Melanoma/immunology
- Melanoma/secondary
- Models, Biological
- Molecular Targeted Therapy
- Mutation, Missense
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/genetics
- Neoplasm Proteins/physiology
- Neoplasms, Second Primary/chemically induced
- Neoplastic Stem Cells/enzymology
- Nevus, Pigmented/enzymology
- Nevus, Pigmented/pathology
- Point Mutation
- Protein Kinase Inhibitors/adverse effects
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Proto-Oncogene Proteins B-raf/antagonists & inhibitors
- Proto-Oncogene Proteins B-raf/genetics
- Proto-Oncogene Proteins B-raf/physiology
- Proto-Oncogene Proteins c-raf/biosynthesis
- Proto-Oncogene Proteins c-raf/physiology
- Skin Neoplasms/chemically induced
- Sulfonamides/adverse effects
- Sulfonamides/pharmacology
- Sulfonamides/therapeutic use
- Tumor Microenvironment
- Vemurafenib
Collapse
Affiliation(s)
- Mathieu Capovilla
- Service de pathologie, centre François-Baclesse, 3, avenue Général-Harris, BP 5026, 14076 Caen cedex 05, France.
| |
Collapse
|
1643
|
Li-Pook-Than J, Snyder M. iPOP goes the world: integrated personalized Omics profiling and the road toward improved health care. ACTA ACUST UNITED AC 2013; 20:660-6. [PMID: 23706632 DOI: 10.1016/j.chembiol.2013.05.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 05/03/2013] [Accepted: 05/09/2013] [Indexed: 10/26/2022]
Abstract
The health of an individual depends upon their DNA as well as upon environmental factors (environome or exposome). It is expected that although the genome is the blueprint of an individual, its analysis with that of the other omes such as the DNA methylome, the transcriptome, proteome, and metabolome will further provide a dynamic assessment of the physiology and health state of an individual. This review will help to categorize the current progress of omics analyses and how omics integration can be used for medical research. We believe that integrative personal omics profiling (iPOP) is a stepping stone to a new road to personalized health care and may improve disease risk assessment, accuracy of diagnosis, disease monitoring, targeted treatments, and understanding the biological processes of disease states for their prevention.
Collapse
Affiliation(s)
- Jennifer Li-Pook-Than
- Department of Genetics, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | | |
Collapse
|
1644
|
Patel A, Schwab R, Liu YT, Bafna V. Amplification and thrifty single-molecule sequencing of recurrent somatic structural variations. Genome Res 2013; 24:318-28. [PMID: 24307551 PMCID: PMC3912422 DOI: 10.1101/gr.161497.113] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Deletion of tumor-suppressor genes as well as other genomic rearrangements pervade cancer genomes across numerous types of solid tumor and hematologic malignancies. However, even for a specific rearrangement, the breakpoints may vary between individuals, such as the recurrent CDKN2A deletion. Characterizing the exact breakpoints for structural variants (SVs) is useful for designating patient-specific tumor biomarkers. We propose AmBre (Amplification of Breakpoints), a method to target SV breakpoints occurring in samples composed of heterogeneous tumor and germline DNA. Additionally, AmBre validates SVs called by whole-exome/genome sequencing and hybridization arrays. AmBre involves a PCR-based approach to amplify the DNA segment containing an SV's breakpoint and then confirms breakpoints using sequencing by Pacific Biosciences RS. To amplify breakpoints with PCR, primers tiling specified target regions are carefully selected with a simulated annealing algorithm to minimize off-target amplification and maximize efficiency at capturing all possible breakpoints within the target regions. To confirm correct amplification and obtain breakpoints, PCR amplicons are combined without barcoding and simultaneously long-read sequenced using a single SMRT cell. Our algorithm efficiently separates reads based on breakpoints. Each read group supporting the same breakpoint corresponds with an amplicon and a consensus amplicon sequence is called. AmBre was used to discover CDKN2A deletion breakpoints in cancer cell lines: A549, CEM, Detroit562, MOLT4, MCF7, and T98G. Also, we successfully assayed RUNX1–RUNX1T1 reciprocal translocations by finding both breakpoints in the Kasumi-1 cell line. AmBre successfully targets SVs where DNA harboring the breakpoints are present in 1:1000 mixtures.
Collapse
Affiliation(s)
- Anand Patel
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, California 92093, USA
| | | | | | | |
Collapse
|
1645
|
Martinez P, McGranahan N, Birkbak NJ, Gerlinger M, Swanton C. Computational optimisation of targeted DNA sequencing for cancer detection. Sci Rep 2013; 3:3309. [PMID: 24296834 PMCID: PMC6506443 DOI: 10.1038/srep03309] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 11/06/2013] [Indexed: 12/13/2022] Open
Abstract
Despite recent progress thanks to next-generation sequencing technologies, personalised cancer medicine is still hampered by intra-tumour heterogeneity and drug resistance. As most patients with advanced metastatic disease face poor survival, there is need to improve early diagnosis. Analysing circulating tumour DNA (ctDNA) might represent a non-invasive method to detect mutations in patients, facilitating early detection. In this article, we define reduced gene panels from publicly available datasets as a first step to assess and optimise the potential of targeted ctDNA scans for early tumour detection. Dividing 4,467 samples into one discovery and two independent validation cohorts, we show that up to 76% of 10 cancer types harbour at least one mutation in a panel of only 25 genes, with high sensitivity across most tumour types. Our analyses demonstrate that targeting “hotspot” regions would introduce biases towards in-frame mutations and would compromise the reproducibility of tumour detection.
Collapse
Affiliation(s)
- Pierre Martinez
- Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | | | | | | | | |
Collapse
|
1646
|
Abstract
Next generation sequencing (NGS), massively parallel or deep sequencing are related terms that describe a DNA sequencing technology which has revolutionised genomic research. Using NGS an entire human genome can be sequenced within a single day. In contrast, the previous Sanger sequencing technology, used to decipher the human genome, required over a decade to deliver the final draft. Although in genome research NGS has mostly superseded conventional Sanger sequencing, it has not yet translated into routine clinical practice. The aim of this article is to review the potential applications of NGS in paediatrics.
Collapse
Affiliation(s)
- Sam Behjati
- Cancer Genome Project, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, , Hinxton, Cambridgeshire, UK
| | | |
Collapse
|
1647
|
Palacios DA, Miyake M, Rosser CJ. Research Highlights: Highlights from the latest articles in biomarkers in medicine. Biomark Med 2013. [DOI: 10.2217/bmm.13.98] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Diego Aguilar Palacios
- College of Medicine, University of Central Florida, 6900 Lake Nona Blvd, Orlando, FL 32827, USA
| | - Makito Miyake
- College of Medicine, University of Central Florida, 6900 Lake Nona Blvd, Orlando, FL 32827, USA
| | - Charles J Rosser
- College of Medicine, University of Central Florida, 6900 Lake Nona Blvd, Orlando, FL 32827, USA
| |
Collapse
|
1648
|
Rački N, Morisset D, Gutierrez-Aguirre I, Ravnikar M. One-step RT-droplet digital PCR: a breakthrough in the quantification of waterborne RNA viruses. Anal Bioanal Chem 2013; 406:661-7. [PMID: 24276251 PMCID: PMC3892107 DOI: 10.1007/s00216-013-7476-y] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 10/25/2013] [Accepted: 10/30/2013] [Indexed: 01/13/2023]
Abstract
Water contamination by viruses has an increasing worldwide impact on human health, and has led to requirements for accurate and quantitative molecular tools. Here, we report the first one-step reverse-transcription droplet digital PCR-based absolute quantification of a RNA virus (rotavirus) in different types of surface water samples. This quantification method proved to be more precise and more tolerant to inhibitory substances than the benchmarking reverse-transcription real-time PCR (RT-qPCR), and needs no standard curve. This new tool is fully amenable for the quantification of viruses in the particularly low concentrations usually found in water samples.
Collapse
Affiliation(s)
- Nejc Rački
- Department of Biotechnology and Systems Biology, National Institute of Biology, Vecna pot 111, 1000, Ljubljana, Slovenia
| | | | | | | |
Collapse
|
1649
|
Koboldt DC, Steinberg KM, Larson DE, Wilson RK, Mardis ER. The next-generation sequencing revolution and its impact on genomics. Cell 2013; 155:27-38. [PMID: 24074859 DOI: 10.1016/j.cell.2013.09.006] [Citation(s) in RCA: 628] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Indexed: 02/07/2023]
Abstract
Genomics is a relatively new scientific discipline, having DNA sequencing as its core technology. As technology has improved the cost and scale of genome characterization over sequencing's 40-year history, the scope of inquiry has commensurately broadened. Massively parallel sequencing has proven revolutionary, shifting the paradigm of genomics to address biological questions at a genome-wide scale. Sequencing now empowers clinical diagnostics and other aspects of medical care, including disease risk, therapeutic identification, and prenatal testing. This Review explores the current state of genomics in the massively parallel sequencing era.
Collapse
Affiliation(s)
- Daniel C Koboldt
- The Genome Institute, School of Medicine, Washington University, St. Louis, MO 63108, USA
| | | | | | | | | |
Collapse
|
1650
|
Abstract
The natural history of breast diseases has changed overtime and successive therapeutic strategies have been adapted accordingly. Recently, biological findings on geno- and phenotypic characteristics of tumor cells offer new basis for the development of treatments that target homogenous and various subtypes of breast cancer. Unfortunately, traditional clinical research tools are not in phase with rapid changes in both biological knowledge of the disease and new targeted agents. New methodological approaches are urgently needed to validate such changes and improvements in prognosis.
Collapse
|