201
|
Frenel JS, Carreira S, Goodall J, Roda D, Perez-Lopez R, Tunariu N, Riisnaes R, Miranda S, Figueiredo I, Nava-Rodrigues D, Smith A, Leux C, Garcia-Murillas I, Ferraldeschi R, Lorente D, Mateo J, Ong M, Yap TA, Banerji U, Gasi Tandefelt D, Turner N, Attard G, de Bono JS. Serial Next-Generation Sequencing of Circulating Cell-Free DNA Evaluating Tumor Clone Response To Molecularly Targeted Drug Administration. Clin Cancer Res 2015; 21:4586-96. [PMID: 26085511 PMCID: PMC4580992 DOI: 10.1158/1078-0432.ccr-15-0584] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 05/26/2015] [Indexed: 12/25/2022]
Abstract
PURPOSE We evaluated whether next-generation sequencing (NGS) of circulating cell-free DNA (cfDNA) could be used for patient selection and as a tumor clone response biomarker in patients with advanced cancers participating in early-phase clinical trials of targeted drugs. EXPERIMENTAL DESIGN Plasma samples from patients with known tumor mutations who completed at least two courses of investigational targeted therapy were collected monthly, until disease progression. NGS was performed sequentially on the Ion Torrent PGM platform. RESULTS cfDNA was extracted from 39 patients with various tumor types. Treatments administered targeted mainly the PI3K-AKT-mTOR pathway (n = 28) or MEK (n = 7). Overall, 159 plasma samples were sequenced with a mean sequencing coverage achieved of 1,685X across experiments. At trial initiation (C1D1), 23 of 39 (59%) patients had at least one mutation identified in cfDNA (mean 2, range 1-5). Out of the 44 mutations identified at C1D1, TP53, PIK3CA and KRAS were the top 3 mutated genes identified, with 18 (41%), 9 (20%), 8 (18%) different mutations, respectively. Out of these 23 patients, 13 received a targeted drug matching their tumor profile. For the 23 patients with cfDNA mutation at C1D1, the monitoring of mutation allele frequency (AF) in consecutive plasma samples during treatment with targeted drugs demonstrated potential treatment associated clonal responses. Longitudinal monitoring of cfDNA samples with multiple mutations indicated the presence of separate clones behaving discordantly. Molecular changes at cfDNA mutation level were associated with time to disease progression by RECIST criteria. CONCLUSIONS Targeted NGS of cfDNA has potential clinical utility to monitor the delivery of targeted therapies.
Collapse
Affiliation(s)
- Jean Sebastien Frenel
- The Institute of Cancer Research and the Royal Marsden Hospital, Sutton, Surrey, London, United Kingdom. Institut de Cancerologie de l'Ouest, Nantes-Saint Herblain, France
| | - Suzanne Carreira
- The Institute of Cancer Research, Sutton, Surrey, London, United Kingdom
| | - Jane Goodall
- The Institute of Cancer Research, Sutton, Surrey, London, United Kingdom
| | - Desam Roda
- The Institute of Cancer Research and the Royal Marsden Hospital, Sutton, Surrey, London, United Kingdom
| | - Raquel Perez-Lopez
- The Institute of Cancer Research and the Royal Marsden Hospital, Sutton, Surrey, London, United Kingdom
| | - Nina Tunariu
- The Institute of Cancer Research and the Royal Marsden Hospital, Sutton, Surrey, London, United Kingdom
| | - Ruth Riisnaes
- The Institute of Cancer Research, Sutton, Surrey, London, United Kingdom
| | - Susana Miranda
- The Institute of Cancer Research, Sutton, Surrey, London, United Kingdom
| | - Ines Figueiredo
- The Institute of Cancer Research, Sutton, Surrey, London, United Kingdom
| | | | - Alan Smith
- The Institute of Cancer Research and the Royal Marsden Hospital, Sutton, Surrey, London, United Kingdom
| | - Christophe Leux
- Département de Santé Publique, CHU de Nantes, Nantes Cedex 1, France
| | - Isaac Garcia-Murillas
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, United Kingdom
| | - Roberta Ferraldeschi
- The Institute of Cancer Research and the Royal Marsden Hospital, Sutton, Surrey, London, United Kingdom
| | - David Lorente
- The Institute of Cancer Research and the Royal Marsden Hospital, Sutton, Surrey, London, United Kingdom
| | - Joaquin Mateo
- The Institute of Cancer Research and the Royal Marsden Hospital, Sutton, Surrey, London, United Kingdom
| | - Michael Ong
- The Institute of Cancer Research and the Royal Marsden Hospital, Sutton, Surrey, London, United Kingdom
| | - Timothy A Yap
- The Institute of Cancer Research and the Royal Marsden Hospital, Sutton, Surrey, London, United Kingdom
| | - Udai Banerji
- The Institute of Cancer Research and the Royal Marsden Hospital, Sutton, Surrey, London, United Kingdom
| | | | - Nick Turner
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, United Kingdom
| | - Gerhardt Attard
- The Institute of Cancer Research and the Royal Marsden Hospital, Sutton, Surrey, London, United Kingdom
| | - Johann S de Bono
- The Institute of Cancer Research and the Royal Marsden Hospital, Sutton, Surrey, London, United Kingdom.
| |
Collapse
|
202
|
Abstract
Currently, gliomas are diagnosed by neuroimaging, and refined diagnosis requires resection or biopsy to obtain tumour tissue for histopathological classification and grading. Blood-derived biomarkers, therefore, would be useful as minimally invasive markers that could support diagnosis and enable monitoring of tumour growth and response to treatment. Such circulating biomarkers could distinguish true progression from therapy-associated changes such as radiation necrosis, and help evaluate the persistence or disappearance of a therapeutic target, such as an oncoprotein or a targetable gene mutation, after targeted therapy. Unlike for other tumours, circulating biomarkers for gliomas are still being defined and are not yet in use in clinical practice. Circulating tumour DNA (ctDNA) isolated from plasma has been shown to reflect the mutational status of glioblastoma, and extracellular vesicles (EVs) containing ctDNA, microRNA and proteins function as rapidly adapting reservoirs for glioma biomarkers such as typical DNA mutations, regulatory microRNAs and oncoproteins. Ideally, circulating tumour cells could enable profiling of the whole-tumour genome, but they are difficult to detect and can reflect only a single cell type of the heterogeneous tumour composition, whereas EVs reflect the complex heterogeneity of the whole tumour, as well as its adaptations to therapy. Although all categories of potential blood-derived biomarkers need to be developed further, findings from other tumour types suggest that EVs are the most promising biomarkers.
Collapse
|
203
|
Lheureux S, Karakasis K, Kohn EC, Oza AM. Ovarian cancer treatment: The end of empiricism? Cancer 2015; 121:3203-11. [PMID: 26096019 PMCID: PMC4744728 DOI: 10.1002/cncr.29481] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 04/12/2015] [Accepted: 05/05/2015] [Indexed: 01/06/2023]
Abstract
The diagnosis, investigation, and management of ovarian cancer are in a state of flux-balancing ever rapid advances in our understanding of its biology with 3 decades of clinical trials. Clinical trials that started with empirically driven selections have evolved in an evidence-informed manner to gradually improve outcome. Has this improved understanding of the biology and associated calls to action led to appropriate changes in therapy? In this review, the authors discuss incorporating emerging data on biology, combinations, dose, and scheduling of new and existing agents with patient preferences in the management of women with ovarian cancer.
Collapse
Affiliation(s)
- Stephanie Lheureux
- Division of Medical Oncology and Hematology, Bras Family Drug Development ProgramPrincess Margaret Cancer CenterTorontoOntarioCanada
| | - Katherine Karakasis
- Division of Medical Oncology and Hematology, Bras Family Drug Development ProgramPrincess Margaret Cancer CenterTorontoOntarioCanada
| | - Elise C. Kohn
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and DiagnosisNational Cancer InstituteBethesdaMaryland
| | - Amit M. Oza
- Division of Medical Oncology and Hematology, Bras Family Drug Development ProgramPrincess Margaret Cancer CenterTorontoOntarioCanada
| |
Collapse
|
204
|
Le Tourneau C, Delord JP, Gonçalves A, Gavoille C, Dubot C, Isambert N, Campone M, Trédan O, Massiani MA, Mauborgne C, Armanet S, Servant N, Bièche I, Bernard V, Gentien D, Jezequel P, Attignon V, Boyault S, Vincent-Salomon A, Servois V, Sablin MP, Kamal M, Paoletti X. Molecularly targeted therapy based on tumour molecular profiling versus conventional therapy for advanced cancer (SHIVA): a multicentre, open-label, proof-of-concept, randomised, controlled phase 2 trial. Lancet Oncol 2015; 16:1324-34. [PMID: 26342236 DOI: 10.1016/s1470-2045(15)00188-6] [Citation(s) in RCA: 796] [Impact Index Per Article: 79.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 07/16/2015] [Accepted: 07/17/2015] [Indexed: 01/05/2023]
Abstract
BACKGROUND Molecularly targeted agents have been reported to have anti-tumour activity for patients whose tumours harbour the matching molecular alteration. These results have led to increased off-label use of molecularly targeted agents on the basis of identified molecular alterations. We assessed the efficacy of several molecularly targeted agents marketed in France, which were chosen on the basis of tumour molecular profiling but used outside their indications, in patients with advanced cancer for whom standard-of-care therapy had failed. METHODS The open-label, randomised, controlled phase 2 SHIVA trial was done at eight French academic centres. We included adult patients with any kind of metastatic solid tumour refractory to standard of care, provided they had an Eastern Cooperative Oncology Group performance status of 0 or 1, disease that was accessible for a biopsy or resection of a metastatic site, and at least one measurable lesion. The molecular profile of each patient's tumour was established with a mandatory biopsy of a metastatic tumour and large-scale genomic testing. We only included patients for whom a molecular alteration was identified within one of three molecular pathways (hormone receptor, PI3K/AKT/mTOR, RAF/MEK), which could be matched to one of ten regimens including 11 available molecularly targeted agents (erlotinib, lapatinib plus trastuzumab, sorafenib, imatinib, dasatinib, vemurafenib, everolimus, abiraterone, letrozole, tamoxifen). We randomly assigned these patients (1:1) to receive a matched molecularly targeted agent (experimental group) or treatment at physician's choice (control group) by central block randomisation (blocks of size six). Randomisation was done centrally with a web-based response system and was stratified according to the Royal Marsden Hospital prognostic score (0 or 1 vs 2 or 3) and the altered molecular pathway. Clinicians and patients were not masked to treatment allocation. Treatments in both groups were given in accordance with the approved product information and standard practice protocols at each institution and were continued until evidence of disease progression. The primary endpoint was progression-free survival in the intention-to-treat population, which was not assessed by independent central review. We assessed safety in any patients who received at least one dose of their assigned treatment. This trial is registered with ClinicalTrials.gov, number NCT01771458. FINDINGS Between Oct 4, 2012, and July 11, 2014, we screened 741 patients with any tumour type. 293 (40%) patients had at least one molecular alteration matching one of the 10 available regimens. At the time of data cutoff, Jan 20, 2015, 195 (26%) patients had been randomly assigned, with 99 in the experimental group and 96 in the control group. All patients in the experimental group started treatment, as did 92 in the control group. Two patients in the control group received a molecularly targeted agent: both were included in their assigned group for efficacy analyses, the patient who received an agent that was allowed in the experimental group was included in the experimental group for the purposes of safety analyses, while the other patient, who received a molecularly targeted agent and chemotherapy, was kept in the control group for safety analyses. Median follow-up was 11·3 months (IQR 5·8-11·6) in the experimental group and 11·3 months (8·1-11·6) in the control group at the time of the primary analysis of progression-free survival. Median progression-free survival was 2·3 months (95% CI 1·7-3·8) in the experimental group versus 2·0 months (1·8-2·1) in the control group (hazard ratio 0·88, 95% CI 0·65-1·19, p=0·41). In the safety population, 43 (43%) of 100 patients treated with a molecularly targeted agent and 32 (35%) of 91 patients treated with cytotoxic chemotherapy had grade 3-4 adverse events (p=0·30). INTERPRETATION The use of molecularly targeted agents outside their indications does not improve progression-free survival compared with treatment at physician's choice in heavily pretreated patients with cancer. Off-label use of molecularly targeted agents should be discouraged, but enrolment in clinical trials should be encouraged to assess predictive biomarkers of efficacy.
Collapse
Affiliation(s)
- Christophe Le Tourneau
- Department of Medical Oncology, Institut Curie, Paris & Saint-Cloud, France; EA7285: Risques cliniques et sécurité en santé des femmes et en santé périnatale, Versailles Saint-Quentin-en Yvelines University, Montigny-le-Bretonneux, France.
| | - Jean-Pierre Delord
- Department of Medical Oncology, Institut Claudius Régaud, Toulouse, France
| | - Anthony Gonçalves
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Céline Gavoille
- Department of Medical Oncology, Centre Alexis Vautrin, Nancy, France
| | - Coraline Dubot
- Department of Medical Oncology, Institut Curie, Saint-Cloud, France
| | - Nicolas Isambert
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
| | - Mario Campone
- Department of Medical Oncology, Centre René Gauducheau, Nantes, France
| | - Olivier Trédan
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | | | | | | | | | - Ivan Bièche
- Pharmacogenomics Unit, Institut Curie, Paris, France
| | | | - David Gentien
- Translational Research Department, Institut Curie, Paris, France
| | | | | | | | | | | | | | - Maud Kamal
- Department of Medical Oncoology, Institut Curie, Paris, France
| | - Xavier Paoletti
- Department of Biostatistics, Institut Curie, Paris, France; Unité INSERM U900, Paris, France
| | | |
Collapse
|
205
|
Tinhofer I, Niehr F, Konschak R, Liebs S, Munz M, Stenzinger A, Weichert W, Keilholz U, Budach V. Next-generation sequencing: hype and hope for development of personalized radiation therapy? Radiat Oncol 2015; 10:183. [PMID: 26316159 PMCID: PMC4554356 DOI: 10.1186/s13014-015-0481-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/07/2015] [Indexed: 01/18/2023] Open
Abstract
The introduction of next-generation sequencing (NGS) in the field of cancer research has boosted worldwide efforts of genome-wide personalized oncology aiming at identifying predictive biomarkers and novel actionable targets. Despite considerable progress in understanding the molecular biology of distinct cancer entities by the use of this revolutionary technology and despite contemporaneous innovations in drug development, translation of NGS findings into improved concepts for cancer treatment remains a challenge. The aim of this article is to describe shortly the NGS platforms for DNA sequencing and in more detail key achievements and unresolved hurdles. A special focus will be given on potential clinical applications of this innovative technique in the field of radiation oncology.
Collapse
Affiliation(s)
- Ingeborg Tinhofer
- Department of Radiooncology and Radiotherapy, Charité University Hospital Berlin, Translational Radiation Oncology Research Laboratory, Charitéplatz 1, 10117, Berlin, Germany. .,German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK) partner site, Heidelberg, Germany.
| | - Franziska Niehr
- Department of Radiooncology and Radiotherapy, Charité University Hospital Berlin, Translational Radiation Oncology Research Laboratory, Charitéplatz 1, 10117, Berlin, Germany.,German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK) partner site, Heidelberg, Germany
| | - Robert Konschak
- Department of Radiooncology and Radiotherapy, Charité University Hospital Berlin, Translational Radiation Oncology Research Laboratory, Charitéplatz 1, 10117, Berlin, Germany.,German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK) partner site, Heidelberg, Germany
| | - Sandra Liebs
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK) partner site, Heidelberg, Germany
| | - Matthias Munz
- Group for Computational Modelling in Medicine, Institute for Theoretical Biology, Humboldt Universität, Berlin, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, University Hospital and National Center for Tumor Diseases, Heidelberg, Germany
| | - Wilko Weichert
- Institute of Pathology, University Hospital and National Center for Tumor Diseases, Heidelberg, Germany.,Institute of Pathology, University Hospital and National Center for Tumor Diseases, Heidelberg, Germany
| | - Ulrich Keilholz
- Charité Comprehensive Cancer Center, Charité University Hospital, Berlin, Germany
| | - Volker Budach
- Department of Radiooncology and Radiotherapy, Charité University Hospital Berlin, Translational Radiation Oncology Research Laboratory, Charitéplatz 1, 10117, Berlin, Germany.,German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK) partner site, Heidelberg, Germany
| |
Collapse
|
206
|
Ashton-Prolla P, Goldim JR, Vairo FPE, da Silveira Matte U, Sequeiros J. Genomic analysis in the clinic: benefits and challenges for health care professionals and patients in Brazil. J Community Genet 2015; 6:275-83. [PMID: 26040235 PMCID: PMC4524873 DOI: 10.1007/s12687-015-0238-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 05/13/2015] [Indexed: 12/20/2022] Open
Abstract
Despite significant advances in the diagnosis and treatment of genetic diseases in the last two decades, there is still a significant proportion where a causative mutation cannot be identified and a definitive genetic diagnosis remains elusive. New genome-wide or high-throughput multiple gene tests have brought new hope to the field, since they can offer fast, cost-effective and comprehensive analysis of genetic variation. This is particularly interesting in disorders with high genetic heterogeneity. There are, however, limitations and concerns regarding the implementation of genomic analysis in everyday clinical practice, including some particular to emerging and developing economies, as Brazil. They include the limited number of actionable genetic variants known to date, difficulties in determining the clinical validity and utility of novel variants, growth of direct-to-consumer genetic testing using a genomic approach and lack of proper training of health care professionals to adequately request, interpret and use genetic information. Despite all these concerns and limitations, the availability of genomic tests has grown at an extremely rapid pace and commercially available services include initiatives in almost all areas of clinical genetics, including newborn and carrier screening. We discuss the benefits and limitations of genomic testing, as well as the ethical implications and the challenges for genetic education and enough available and qualified health care professionals, to ensure the adequate process of informed consent, meaningful interpretation and use of genomic data and definition of a clear regulatory framework in the particular context of Brazil.
Collapse
Affiliation(s)
- Patrícia Ashton-Prolla
- Serviço de Genetica Medica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil,
| | | | | | | | | |
Collapse
|
207
|
Comprehensive evaluation of methods to isolate, quantify, and characterize circulating cell-free DNA from small volumes of plasma. Anal Bioanal Chem 2015; 407:6873-8. [DOI: 10.1007/s00216-015-8846-4] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 06/03/2015] [Accepted: 06/10/2015] [Indexed: 12/21/2022]
|
208
|
Zill OA, Greene C, Sebisanovic D, Siew LM, Leng J, Vu M, Hendifar AE, Wang Z, Atreya CE, Kelley RK, Van Loon K, Ko AH, Tempero MA, Bivona TG, Munster PN, Talasaz A, Collisson EA. Cell-Free DNA Next-Generation Sequencing in Pancreatobiliary Carcinomas. Cancer Discov 2015; 5:1040-8. [PMID: 26109333 DOI: 10.1158/2159-8290.cd-15-0274] [Citation(s) in RCA: 187] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 06/22/2015] [Indexed: 02/06/2023]
Abstract
UNLABELLED Patients with pancreatic and biliary carcinomas lack personalized treatment options, in part because biopsies are often inadequate for molecular characterization. Cell-free DNA (cfDNA) sequencing may enable a precision oncology approach in this setting. We attempted to prospectively analyze 54 genes in tumor and cfDNA for 26 patients. Tumor sequencing failed in 9 patients (35%). In the remaining 17, 90.3% (95% confidence interval, 73.1%-97.5%) of mutations detected in tumor biopsies were also detected in cfDNA. The diagnostic accuracy of cfDNA sequencing was 97.7%, with 92.3% average sensitivity and 100% specificity across five informative genes. Changes in cfDNA correlated well with tumor marker dynamics in serial sampling (r = 0.93). We demonstrate that cfDNA sequencing is feasible, accurate, and sensitive in identifying tumor-derived mutations without prior knowledge of tumor genotype or the abundance of circulating tumor DNA. cfDNA sequencing should be considered in pancreatobiliary cancer trials where tissue sampling is unsafe, infeasible, or otherwise unsuccessful. SIGNIFICANCE Precision medicine efforts in biliary and pancreatic cancers have been frustrated by difficulties in obtaining adequate tumor tissue for next-generation sequencing. cfDNA sequencing reliably and accurately detects tumor-derived mutations, paving the way for precision oncology approaches in these deadly diseases.
Collapse
Affiliation(s)
- Oliver A Zill
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California. Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California. Guardant Health, Inc., Redwood City, California
| | - Claire Greene
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California. Department of Medicine, University of California, San Francisco, California
| | | | | | - Jim Leng
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California. Department of Medicine, University of California, San Francisco, California
| | - Mary Vu
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | | | - Zhen Wang
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Chloe E Atreya
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California. Department of Medicine, University of California, San Francisco, California
| | - Robin K Kelley
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California. Department of Medicine, University of California, San Francisco, California
| | - Katherine Van Loon
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California. Department of Medicine, University of California, San Francisco, California
| | - Andrew H Ko
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California. Department of Medicine, University of California, San Francisco, California
| | - Margaret A Tempero
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California. Department of Medicine, University of California, San Francisco, California
| | - Trever G Bivona
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California. Department of Medicine, University of California, San Francisco, California
| | - Pamela N Munster
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California. Department of Medicine, University of California, San Francisco, California
| | | | - Eric A Collisson
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California. Department of Medicine, University of California, San Francisco, California.
| |
Collapse
|
209
|
Zill OA, Greene C, Sebisanovic D, Siew LM, Leng J, Vu M, Hendifar AE, Wang Z, Atreya CE, Kelley RK, Van Loon K, Ko AH, Tempero MA, Bivona TG, Munster PN, Talasaz A, Collisson EA. Cell-Free DNA Next-Generation Sequencing in Pancreatobiliary Carcinomas. Cancer Discov 2015. [PMID: 26109333 DOI: 10.1158/2159-8290.cd-15-0274.cell-free] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
UNLABELLED Patients with pancreatic and biliary carcinomas lack personalized treatment options, in part because biopsies are often inadequate for molecular characterization. Cell-free DNA (cfDNA) sequencing may enable a precision oncology approach in this setting. We attempted to prospectively analyze 54 genes in tumor and cfDNA for 26 patients. Tumor sequencing failed in 9 patients (35%). In the remaining 17, 90.3% (95% confidence interval, 73.1%-97.5%) of mutations detected in tumor biopsies were also detected in cfDNA. The diagnostic accuracy of cfDNA sequencing was 97.7%, with 92.3% average sensitivity and 100% specificity across five informative genes. Changes in cfDNA correlated well with tumor marker dynamics in serial sampling (r = 0.93). We demonstrate that cfDNA sequencing is feasible, accurate, and sensitive in identifying tumor-derived mutations without prior knowledge of tumor genotype or the abundance of circulating tumor DNA. cfDNA sequencing should be considered in pancreatobiliary cancer trials where tissue sampling is unsafe, infeasible, or otherwise unsuccessful. SIGNIFICANCE Precision medicine efforts in biliary and pancreatic cancers have been frustrated by difficulties in obtaining adequate tumor tissue for next-generation sequencing. cfDNA sequencing reliably and accurately detects tumor-derived mutations, paving the way for precision oncology approaches in these deadly diseases.
Collapse
Affiliation(s)
- Oliver A Zill
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California. Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California. Guardant Health, Inc., Redwood City, California
| | - Claire Greene
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California. Department of Medicine, University of California, San Francisco, California
| | | | | | - Jim Leng
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California. Department of Medicine, University of California, San Francisco, California
| | - Mary Vu
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | | | - Zhen Wang
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Chloe E Atreya
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California. Department of Medicine, University of California, San Francisco, California
| | - Robin K Kelley
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California. Department of Medicine, University of California, San Francisco, California
| | - Katherine Van Loon
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California. Department of Medicine, University of California, San Francisco, California
| | - Andrew H Ko
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California. Department of Medicine, University of California, San Francisco, California
| | - Margaret A Tempero
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California. Department of Medicine, University of California, San Francisco, California
| | - Trever G Bivona
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California. Department of Medicine, University of California, San Francisco, California
| | - Pamela N Munster
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California. Department of Medicine, University of California, San Francisco, California
| | | | - Eric A Collisson
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California. Department of Medicine, University of California, San Francisco, California.
| |
Collapse
|
210
|
Francis G, Stein S. Circulating Cell-Free Tumour DNA in the Management of Cancer. Int J Mol Sci 2015; 16:14122-42. [PMID: 26101870 PMCID: PMC4490543 DOI: 10.3390/ijms160614122] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 04/23/2015] [Accepted: 05/26/2015] [Indexed: 12/22/2022] Open
Abstract
With the development of new sensitive molecular techniques, circulating cell-free tumour DNA containing mutations can be identified in the plasma of cancer patients. The applications of this technology may result in significant changes to the care and management of cancer patients. Whilst, currently, these "liquid biopsies" are used to supplement the histological diagnosis of cancer and metastatic disease, in the future these assays may replace the need for invasive procedures. Applications include the monitoring of tumour burden, the monitoring of minimal residual disease, monitoring of tumour heterogeneity, monitoring of molecular resistance and early diagnosis of tumours and metastatic disease.
Collapse
Affiliation(s)
- Glenn Francis
- Director Pathology, Genomics for Life, Herston 4006, Australia.
- School of Medicine, Griffith University, Gold Coast 4215, Australia.
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia 4067, Australia.
| | - Sandra Stein
- Laboratory Director, Genomics for Life, Herston 4006, Australia.
| |
Collapse
|
211
|
Lebofsky R, Decraene C, Bernard V, Kamal M, Blin A, Leroy Q, Rio Frio T, Pierron G, Callens C, Bieche I, Saliou A, Madic J, Rouleau E, Bidard FC, Lantz O, Stern MH, Le Tourneau C, Pierga JY. Circulating tumor DNA as a non-invasive substitute to metastasis biopsy for tumor genotyping and personalized medicine in a prospective trial across all tumor types. Mol Oncol 2015. [PMID: 25579085 DOI: 10.1016/j.molonc.2014.12.003] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cell-free tumor DNA (ctDNA) has the potential to enable non-invasive diagnostic tests for personalized medicine in providing similar molecular information as that derived from invasive tumor biopsies. The histology-independent phase II SHIVA trial matches patients with targeted therapeutics based on previous screening of multiple somatic mutations using metastatic biopsies. To evaluate the utility of ctDNA in this trial, as an ancillary study we performed de novo detection of somatic mutations using plasma DNA compared to metastasis biopsies in 34 patients covering 18 different tumor types, scanning 46 genes and more than 6800 COSMIC mutations with a multiplexed next-generation sequencing panel. In 27 patients, 28 of 29 mutations identified in metastasis biopsies (97%) were detected in matched ctDNA. Among these 27 patients, one additional mutation was found in ctDNA only. In the seven other patients, mutation detection from metastasis biopsy failed due to inadequate biopsy material, but was successful in all plasma DNA samples providing three more potential actionable mutations. These results suggest that ctDNA analysis is a potential alternative and/or replacement to analyses using costly, harmful and lengthy tissue biopsies of metastasis, irrespective of cancer type and metastatic site, for multiplexed mutation detection in selecting personalized therapies based on the patient's tumor genetic content.
Collapse
Affiliation(s)
- Ronald Lebofsky
- Circulating Cancer Biomarkers Lab, SiRIC, Translational Research Department, Institut Curie, Paris, France
| | - Charles Decraene
- Circulating Cancer Biomarkers Lab, SiRIC, Translational Research Department, Institut Curie, Paris, France; CNRS UMR144, Institut Curie, Paris, France
| | | | - Maud Kamal
- Department of Medical Oncology, Institut Curie, Paris and Saint-Cloud, France
| | - Anthony Blin
- ICGex NGS Platform, Institut Curie, Paris, France
| | | | | | | | | | - Ivan Bieche
- Oncogenetic Laboratory, Institut Curie, Paris, France
| | - Adrien Saliou
- Circulating Cancer Biomarkers Lab, SiRIC, Translational Research Department, Institut Curie, Paris, France
| | - Jordan Madic
- Circulating Cancer Biomarkers Lab, SiRIC, Translational Research Department, Institut Curie, Paris, France
| | | | - François-Clément Bidard
- Circulating Cancer Biomarkers Lab, SiRIC, Translational Research Department, Institut Curie, Paris, France; Department of Medical Oncology, Institut Curie, Paris and Saint-Cloud, France
| | - Olivier Lantz
- INSERM U932, Institut Curie, Paris, France; CIC-BT-507, Institut Curie, Paris, France
| | | | - Christophe Le Tourneau
- Department of Medical Oncology, Institut Curie, Paris and Saint-Cloud, France; INSERM U900, Institut Curie, Paris, France
| | - Jean-Yves Pierga
- Circulating Cancer Biomarkers Lab, SiRIC, Translational Research Department, Institut Curie, Paris, France; Department of Medical Oncology, Institut Curie, Paris and Saint-Cloud, France; University Paris Descartes, Paris, France.
| |
Collapse
|
212
|
Lebofsky R, Decraene C, Bernard V, Kamal M, Blin A, Leroy Q, Rio Frio T, Pierron G, Callens C, Bieche I, Saliou A, Madic J, Rouleau E, Bidard FC, Lantz O, Stern MH, Le Tourneau C, Pierga JY. Circulating tumor DNA as a non-invasive substitute to metastasis biopsy for tumor genotyping and personalized medicine in a prospective trial across all tumor types. Mol Oncol 2014; 9:783-90. [PMID: 25579085 DOI: 10.1016/j.molonc.2014.12.003] [Citation(s) in RCA: 202] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 11/26/2014] [Accepted: 12/10/2014] [Indexed: 12/18/2022] Open
Abstract
Cell-free tumor DNA (ctDNA) has the potential to enable non-invasive diagnostic tests for personalized medicine in providing similar molecular information as that derived from invasive tumor biopsies. The histology-independent phase II SHIVA trial matches patients with targeted therapeutics based on previous screening of multiple somatic mutations using metastatic biopsies. To evaluate the utility of ctDNA in this trial, as an ancillary study we performed de novo detection of somatic mutations using plasma DNA compared to metastasis biopsies in 34 patients covering 18 different tumor types, scanning 46 genes and more than 6800 COSMIC mutations with a multiplexed next-generation sequencing panel. In 27 patients, 28 of 29 mutations identified in metastasis biopsies (97%) were detected in matched ctDNA. Among these 27 patients, one additional mutation was found in ctDNA only. In the seven other patients, mutation detection from metastasis biopsy failed due to inadequate biopsy material, but was successful in all plasma DNA samples providing three more potential actionable mutations. These results suggest that ctDNA analysis is a potential alternative and/or replacement to analyses using costly, harmful and lengthy tissue biopsies of metastasis, irrespective of cancer type and metastatic site, for multiplexed mutation detection in selecting personalized therapies based on the patient's tumor genetic content.
Collapse
Affiliation(s)
- Ronald Lebofsky
- Circulating Cancer Biomarkers Lab, SiRIC, Translational Research Department, Institut Curie, Paris, France
| | - Charles Decraene
- Circulating Cancer Biomarkers Lab, SiRIC, Translational Research Department, Institut Curie, Paris, France; CNRS UMR144, Institut Curie, Paris, France
| | | | - Maud Kamal
- Department of Medical Oncology, Institut Curie, Paris and Saint-Cloud, France
| | - Anthony Blin
- ICGex NGS Platform, Institut Curie, Paris, France
| | | | | | | | | | - Ivan Bieche
- Oncogenetic Laboratory, Institut Curie, Paris, France
| | - Adrien Saliou
- Circulating Cancer Biomarkers Lab, SiRIC, Translational Research Department, Institut Curie, Paris, France
| | - Jordan Madic
- Circulating Cancer Biomarkers Lab, SiRIC, Translational Research Department, Institut Curie, Paris, France
| | | | - François-Clément Bidard
- Circulating Cancer Biomarkers Lab, SiRIC, Translational Research Department, Institut Curie, Paris, France; Department of Medical Oncology, Institut Curie, Paris and Saint-Cloud, France
| | - Olivier Lantz
- INSERM U932, Institut Curie, Paris, France; CIC-BT-507, Institut Curie, Paris, France
| | | | - Christophe Le Tourneau
- Department of Medical Oncology, Institut Curie, Paris and Saint-Cloud, France; INSERM U900, Institut Curie, Paris, France
| | - Jean-Yves Pierga
- Circulating Cancer Biomarkers Lab, SiRIC, Translational Research Department, Institut Curie, Paris, France; Department of Medical Oncology, Institut Curie, Paris and Saint-Cloud, France; University Paris Descartes, Paris, France.
| |
Collapse
|