1101
|
Graham D, Lipman G, Sehgal V, Lovat LB. Monitoring the premalignant potential of Barrett's oesophagus'. Frontline Gastroenterol 2016; 7:316-322. [PMID: 27761232 PMCID: PMC5036243 DOI: 10.1136/flgastro-2016-100712] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 04/15/2016] [Indexed: 02/04/2023] Open
Abstract
The landscape for patients with Barrett's oesophagus (BE) has changed significantly in the last decade. Research and new guidelines have helped gastroenterologists to better identify those patients with BE who are particularly at risk of developing oesophageal adenocarcinoma. In parallel, developments in endoscopic image enhancement technology and optical biopsy techniques have improved our ability to detect high-risk lesions. Once these lesions have been identified, the improvements in minimally invasive endoscopic therapies has meant that these patients can potentially be cured of early cancer and high-risk dysplastic lesions without the need for surgery, which still has a significant morbidity and mortality. The importance of reaching an accurate diagnosis of BE remains of paramount importance. More work is needed, however. The vast majority of those undergoing surveillance for their BE do not progress towards cancer and thus undergo a regular invasive procedure, which may impact on their psychological and physical well-being while incurring significant cost to the health service. New work that explores cheaper endoscopic or non-invasive ways to identify the at-risk individual provides exciting avenues for research. In future, the diagnosis and monitoring of patients with BE could move away from hospitals and into primary care.
Collapse
Affiliation(s)
- David Graham
- Division of Surgery and Interventional Science, University College London, London, UK,Gastrointestinal Unit, University College Hospital, London, UK
| | - Gideon Lipman
- Division of Surgery and Interventional Science, University College London, London, UK,Gastrointestinal Unit, University College Hospital, London, UK
| | - Vinay Sehgal
- Division of Surgery and Interventional Science, University College London, London, UK,Gastrointestinal Unit, University College Hospital, London, UK
| | - Laurence B Lovat
- Division of Surgery and Interventional Science, University College London, London, UK,Gastrointestinal Unit, University College Hospital, London, UK
| |
Collapse
|
1102
|
Pérez-Callejo D, Romero A, Provencio M, Torrente M. Liquid biopsy based biomarkers in non-small cell lung cancer for diagnosis and treatment monitoring. Transl Lung Cancer Res 2016; 5:455-465. [PMID: 27826527 DOI: 10.21037/tlcr.2016.10.07] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Advances in the knowledge of the biology of non-small cell lung cancer (NSCLC) have revealed molecular information used for systemic cancer therapy targeting metastatic disease, with an important impact on patients overall survival (OS) and quality of life. However, a biopsy of overt metastases is an invasive procedure limited to certain locations and not easily acceptable in the clinic. Moreover, a single biopsy cannot reflect the clonal heterogeneity of the tumor. The analysis of peripheral blood samples of cancer patients represents a new source of cancer-derived material, known as liquid biopsy, and its components can be obtained from almost all body fluids. These components have shown to reflect characteristics of the status of both the primary and metastatic diseases, helping the clinicians to move towards a personalized medicine. The present review focuses on the liquid biopsy components: circulating tumor cells (CTCS), circulating free DNA (cfDNA), exosomes and tumor-educated platelets (TEP); the isolation technologies used and their potential use for non-invasive screening, early diagnosis, prognosis, response to treatment and real time monitoring of the disease, in NSCLC patients.
Collapse
Affiliation(s)
- David Pérez-Callejo
- Department of Medical Oncology, Puerta de Hierro Majadahonda University Hospital, Madrid, Spain
| | - Atocha Romero
- Department of Medical Oncology, Puerta de Hierro Majadahonda University Hospital, Madrid, Spain
| | - Mariano Provencio
- Department of Medical Oncology, Puerta de Hierro Majadahonda University Hospital, Madrid, Spain
| | - María Torrente
- Department of Medical Oncology, Puerta de Hierro Majadahonda University Hospital, Madrid, Spain
| |
Collapse
|
1103
|
Lehrach H. Omics approaches to individual variation: modeling networks and the virtual patient. DIALOGUES IN CLINICAL NEUROSCIENCE 2016. [PMID: 27757060 PMCID: PMC5067143 DOI: 10.31887/dcns.2016.18.3/hlehrach] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Every human is unique. We differ in our genomes, environment, behavior, disease history, and past and current medical treatment—a complex catalog of differences that often leads to variations in the way each of us responds to a particular therapy. We argue here that true personalization of drug therapies will rely on “virtual patient” models based on a detailed characterization of the individual patient by molecular, imaging, and sensor techniques. The models will be based, wherever possible, on the molecular mechanisms of disease processes and drug action but can also expand to hybrid models including statistics/machine learning/artificial intelligence-based elements trained on available data to address therapeutic areas or therapies for which insufficient information on mechanisms is available. Depending on the disease, its mechanisms, and the therapy, virtual patient models can be implemented at a fairly high level of abstraction, with molecular models representing cells, cell types, or organs relevant to the clinical question, interacting not only with each other but also the environment. In the future, “virtual patient/in-silico self” models may not only become a central element of our health care system, reducing otherwise unavoidable mistakes and unnecessary costs, but also act as “guardian angels” accompanying us through life to protect us against dangers and to help us to deal intelligently with our own health and wellness.
Collapse
|
1104
|
Aguado C, Giménez-Capitán A, Karachaliou N, Pérez-Rosado A, Viteri S, Morales-Espinosa D, Rosell R. Fusion gene and splice variant analyses in liquid biopsies of lung cancer patients. Transl Lung Cancer Res 2016; 5:525-531. [PMID: 27826534 DOI: 10.21037/tlcr.2016.09.02] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Obtaining a biopsy of solid tumors requires invasive procedures that strongly limit patient compliance. In contrast, a blood extraction is safe, can be performed at many time points during the course disease and encourages appropriate therapy modifications, potentially improving the patient's clinical outcome and quality of life. Fusion of the tyrosine kinase genes anaplastic lymphoma kinase (ALK), C-ROS oncogen 1 (ROS 1), rearranged during transfection (RET) and neurotrophic tyrosine kinase 1 (NTRK1) occur in 1-5% of lung adenocarcinomas and constitute therapeutic targets for tyrosine kinase inhibitors. In addition, a MET splicing variant of exon 14, has been reported in 2-4% of lung adenocarcinoma and recent studies suggests that targeted therapies inhibiting MET signaling would be beneficial for patients with this alteration. In this review, we will summarize the new techniques recently developed to detect ALK, RET, ROS and NTRK1 fusions and MET exon 14 splicing variant in liquid biopsy using plasma, serum, circulating tumor cells (CTCs), platelets and exosomes as starting material.
Collapse
Affiliation(s)
- Cristina Aguado
- Pangaea Biotech, Laboratory of Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Ana Giménez-Capitán
- Pangaea Biotech, Laboratory of Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Niki Karachaliou
- Pangaea Biotech, Laboratory of Oncology, Quirón Dexeus University Hospital, Barcelona, Spain;; Dr Rosell Oncology Institute, Quirón Dexeus University Hospital, Barcelona, Spain;; University Hospital Sagrat Cor Quirónsalud group, Barcelona, Spain
| | - Ana Pérez-Rosado
- Pangaea Biotech, Laboratory of Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Santiago Viteri
- Dr Rosell Oncology Institute, Quirón Dexeus University Hospital, Barcelona, Spain
| | | | - Rafael Rosell
- Pangaea Biotech, Laboratory of Oncology, Quirón Dexeus University Hospital, Barcelona, Spain;; Dr Rosell Oncology Institute, Quirón Dexeus University Hospital, Barcelona, Spain;; Cancer Biology & Precision Medicine Program, Catalan Institute of Oncology, Germans Trias i Pujol Health Sciences Institute and Hospital, Badalona, Spain;; Autonomous University of Barcelona (UAB), Campus Can Ruti, Badalona, Spain;; Molecular Oncology Research (MORe) Foundation, Barcelona, Spain
| |
Collapse
|
1105
|
Sonnenblick A, Pondé N, Piccart M. Metastatic breast cancer: The Odyssey of personalization. Mol Oncol 2016; 10:1147-59. [PMID: 27430154 PMCID: PMC5423195 DOI: 10.1016/j.molonc.2016.07.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 06/27/2016] [Accepted: 07/05/2016] [Indexed: 12/31/2022] Open
Abstract
Metastatic breast cancer is the most frequent cause of cancer death for women worldwide. In the last 15 years, a large number of new agents have entered clinical use, a result of the dramatic increase in our understanding of the molecular underpinnings of metastatic breast cancer. However, while these agents have led to better outcomes, they are also at the root cause of increasing financial pressure on healthcare systems. Moreover, decision making in an era where every year new agents are added to the therapeutic armamentarium has also become a significant challenge for medical oncologists. In the present article, we will provide an ample review on the most recent developments in the field of treatment of the different subtypes of metastatic breast cancer with a critical discussion on the slow progress made in identifying response biomarkers. New hopes in the form of ctDNA monitoring and functional imaging will be presented.
Collapse
Affiliation(s)
- A Sonnenblick
- Department of Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Boulevard de Waterloo 125, B 1000 Brussels, Belgium; Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - N Pondé
- Department of Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Boulevard de Waterloo 125, B 1000 Brussels, Belgium
| | - M Piccart
- Department of Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Boulevard de Waterloo 125, B 1000 Brussels, Belgium.
| |
Collapse
|
1106
|
Calabuig-Fariñas S, Jantus-Lewintre E, Herreros-Pomares A, Camps C. Circulating tumor cells versus circulating tumor DNA in lung cancer-which one will win? Transl Lung Cancer Res 2016; 5:466-482. [PMID: 27826528 DOI: 10.21037/tlcr.2016.10.02] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Liquid biopsies appear to be a reliable alternative to conventional biopsies that can provide both precise molecular data useful for improving the clinical management of lung cancer patients as well as a less invasive way of monitoring tumor behavior. These advances are supported by important biotechnological developments in the fields of circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA). Analysis of CTCs and ctDNA may be useful in treatment selection, for response monitoring, and in studying resistance mechanisms. This review focuses on the most recent technological achievements and the most relevant clinical applications for lung cancer patients in the CTC and ctDNA fields, highlighting those that are already (or are close to) being implemented in daily clinical practice.
Collapse
Affiliation(s)
- Silvia Calabuig-Fariñas
- Molecular Oncology Laboratory, General University Hospital Research Foundation, University General Hospital of Valencia, Valencia, Spain;; Department of Pathology, Universitat de València, Valencia, Spain
| | - Eloísa Jantus-Lewintre
- Molecular Oncology Laboratory, General University Hospital Research Foundation, University General Hospital of Valencia, Valencia, Spain;; Department of Biotechnology, Universitat Politècnica de València, Valencia, Spain
| | - Alejandro Herreros-Pomares
- Molecular Oncology Laboratory, General University Hospital Research Foundation, University General Hospital of Valencia, Valencia, Spain;; Department of Biotechnology, Universitat Politècnica de València, Valencia, Spain
| | - Carlos Camps
- Molecular Oncology Laboratory, General University Hospital Research Foundation, University General Hospital of Valencia, Valencia, Spain;; Department of Medicine, Universitat de València, Valencia, Spain;; Department of Medical Oncology, University General Hospital of Valencia, Valencia, Spain
| |
Collapse
|
1107
|
Malapelle U, Pisapia P, Rocco D, Smeraglio R, di Spirito M, Bellevicine C, Troncone G. Next generation sequencing techniques in liquid biopsy: focus on non-small cell lung cancer patients. Transl Lung Cancer Res 2016; 5:505-510. [PMID: 27826531 DOI: 10.21037/tlcr.2016.10.08] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The advent of genomic based personalized medicine has led to multiple advances in the molecular characterization of many tumor types, such as non-small cell lung cancer (NSCLC). NSCLC is diagnosed in most cases on small tissue samples that may be not always sufficient for EGFR mutational assessment to select patients for first and second generations' tyrosine kinase inhibitors (TKIs) therapy. In patients without tissue availability at presentation, the analysis of cell free DNA (cfDNA) derived from liquid biopsy samples, in particular from plasma, represent an established alternative to provide EGFR mutational testing for treatment decision making. In addition, a new paradigm for TKIs resistance management was recently approved by Food and Drug Administration, supporting the liquid biopsy based genotyping prior to tissue based genotyping for the detection of T790M mutation to select patients for third generation TKIs. In these settings, real time PCR (RT-PCR) and digital PCR 'targeted' methods, which detect known mutations by specific probes, have extensively been adopted. Taking into account the restricted reference range and the limited multiplexing power of these targeted methods, the performance of liquid biopsy analyses may be further improved by next generation sequencing (NGS). While most tissue based NGS genotyping is well established, liquid biopsy NGS application is challenging, requiring a careful validation of the whole process, from blood collection to variant calling. Here we review this evolving field, highlighting those methodological points that are crucial to accurately select NSCLC patients for TKIs treatment administration by NGS on cfDNA.
Collapse
Affiliation(s)
- Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Pasquale Pisapia
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Danilo Rocco
- Oncology Unit, A.O.R.N. Vincenzo Monaldi - Ospedale dei Colli, Naples, Italy
| | - Riccardo Smeraglio
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Maria di Spirito
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Claudio Bellevicine
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Giancarlo Troncone
- Department of Public Health, University of Naples Federico II, Naples, Italy
| |
Collapse
|
1108
|
Affiliation(s)
- Smadar Geva
- Institute of Oncology, Davidoff Cancer Center, Rabin Medical Center, Petach Tikva, Israel; ; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Laila C Roisman
- Institute of Oncology, Davidoff Cancer Center, Rabin Medical Center, Petach Tikva, Israel
| | - Nir Peled
- Institute of Oncology, Davidoff Cancer Center, Rabin Medical Center, Petach Tikva, Israel; ; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
1109
|
Pietri E, Conteduca V, Andreis D, Massa I, Melegari E, Sarti S, Cecconetto L, Schirone A, Bravaccini S, Serra P, Fedeli A, Maltoni R, Amadori D, De Giorgi U, Rocca A. Androgen receptor signaling pathways as a target for breast cancer treatment. Endocr Relat Cancer 2016; 23:R485-98. [PMID: 27528625 DOI: 10.1530/erc-16-0190] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 08/12/2016] [Indexed: 12/23/2022]
Abstract
The androgen receptor (AR) is a ligand-dependent transcription factor, and its effects on breast range from physiological pubertal development and age-related modifications to cancer onset and proliferation. The prevalence of AR in early breast cancer is around 60%, and AR is more frequently expressed in ER-positive than in ER-negative tumors. We offer an overview of AR signaling pathways in different breast cancer subtypes, providing evidence that its oncogenic role is likely to be different in distinct biological and clinical scenarios. In particular, in ER-positive breast cancer, AR signaling often antagonizes the growth stimulatory effect of ER signaling; in triple-negative breast cancer (TNBC), AR seems to drive tumor progression (at least in luminal AR subtype of TNBC with a gene expression profile mimicking luminal subtypes despite being negative to ER and enriched in AR expression); in HER2-positive breast cancer, in the absence of ER expression, AR signaling has a proliferative role. These data represent the rationale for AR-targeting treatment as a potentially new target therapy in breast cancer subset using androgen agonists in some AR-positive/ER-positive tumors, AR antagonists in triple-negative/AR-positive tumors and in combination with anti-HER2 agents or with other signaling pathways inhibitors (including PI3K/MYC/ERK) in HER2-positive/AR-positive tumors. Only the ongoing and future prospective clinical trials will allow us to establish which agents are the best option in every specific condition, keeping in mind that there is evidence of opposite androgens and AR agonist/antagonist drug effects on cell proliferation particularly in AR-positive/ER-positive tumors.
Collapse
Affiliation(s)
- Elisabetta Pietri
- Department of Medical OncologyIstituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Forlì-Cesena, Italy
| | - Vincenza Conteduca
- Department of Medical OncologyIstituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Forlì-Cesena, Italy
| | - Daniele Andreis
- Unit of Biostatistics and Clinical TrialsIstituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Forlì-Cesena, Italy
| | - Ilaria Massa
- Unit of Biostatistics and Clinical TrialsIstituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Forlì-Cesena, Italy
| | - Elisabetta Melegari
- Department of Medical OncologyIstituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Forlì-Cesena, Italy
| | - Samanta Sarti
- Department of Medical OncologyIstituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Forlì-Cesena, Italy
| | - Lorenzo Cecconetto
- Department of Medical OncologyIstituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Forlì-Cesena, Italy
| | - Alessio Schirone
- Department of Medical OncologyIstituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Forlì-Cesena, Italy
| | - Sara Bravaccini
- Biosciences LaboratoryIstituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Forlì-Cesena, Italy
| | - Patrizia Serra
- Unit of Biostatistics and Clinical TrialsIstituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Forlì-Cesena, Italy
| | - Anna Fedeli
- Department of Medical OncologyIstituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Forlì-Cesena, Italy
| | - Roberta Maltoni
- Department of Medical OncologyIstituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Forlì-Cesena, Italy
| | - Dino Amadori
- Department of Medical OncologyIstituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Forlì-Cesena, Italy
| | - Ugo De Giorgi
- Department of Medical OncologyIstituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Forlì-Cesena, Italy
| | - Andrea Rocca
- Department of Medical OncologyIstituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Forlì-Cesena, Italy
| |
Collapse
|
1110
|
Bioluminescence Microscopy as a Method to Measure Single Cell Androgen Receptor Activity Heterogeneous Responses to Antiandrogens. Sci Rep 2016; 6:33968. [PMID: 27678181 PMCID: PMC5039635 DOI: 10.1038/srep33968] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 09/06/2016] [Indexed: 12/14/2022] Open
Abstract
Cancer cell heterogeneity is well-documented. Therefore, techniques to monitor single cell heterogeneous responses to treatment are needed. We developed a highly translational and quantitative bioluminescence microscopy method to measure single cell androgen receptor (AR) activity modulation by antiandrogens from fluid biopsies. We showed that this assay can detect heterogeneous cellular response to drug treatment and that the sum of single cell AR activity can mirror the response in the whole cell population. This method may thus be used to monitor heterogeneous dynamic treatment responses in cancer cells.
Collapse
|
1111
|
Affiliation(s)
- Marius Ilié
- Laboratory of Clinical and Experimental Pathology and Liquid Biopsy Laboratory, Pasteur Hospital, University of Nice Sophia Antipolis, Nice, France;; Institute for Research on Cancer and Ageing, Nice (IRCAN), Inserm U1081/UMR CNRS 7284, Team 3, Antoine Lacassagne Cancer Center, Nice, France;; Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, Nice, France;; University Hospital Federation OncoAge, CHU de Nice, University of Nice Sophia Antipolis, Nice, France
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology and Liquid Biopsy Laboratory, Pasteur Hospital, University of Nice Sophia Antipolis, Nice, France;; Institute for Research on Cancer and Ageing, Nice (IRCAN), Inserm U1081/UMR CNRS 7284, Team 3, Antoine Lacassagne Cancer Center, Nice, France;; Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, Nice, France;; University Hospital Federation OncoAge, CHU de Nice, University of Nice Sophia Antipolis, Nice, France
| |
Collapse
|
1112
|
Kaisaki PJ, Cutts A, Popitsch N, Camps C, Pentony MM, Wilson G, Page S, Kaur K, Vavoulis D, Henderson S, Gupta A, Middleton MR, Karydis I, Talbot DC, Schuh A, Taylor JC. Targeted Next-Generation Sequencing of Plasma DNA from Cancer Patients: Factors Influencing Consistency with Tumour DNA and Prospective Investigation of Its Utility for Diagnosis. PLoS One 2016; 11:e0162809. [PMID: 27626278 PMCID: PMC5023174 DOI: 10.1371/journal.pone.0162809] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 08/29/2016] [Indexed: 12/16/2022] Open
Abstract
Use of circulating tumour DNA (ctDNA) as a liquid biopsy has been proposed for potential identification and monitoring of solid tumours. We investigate a next-generation sequencing approach for mutation detection in ctDNA in two related studies using a targeted panel. The first study was retrospective, using blood samples taken from melanoma patients at diverse timepoints before or after treatment, aiming to evaluate correlation between mutations identified in biopsy and ctDNA, and to acquire a first impression of influencing factors. We found good concordance between ctDNA and tumour mutations of melanoma patients when blood samples were collected within one year of biopsy or before treatment. In contrast, when ctDNA was sequenced after targeted treatment in melanoma, mutations were no longer found in 9 out of 10 patients, suggesting the method might be useful for detecting treatment response. Building on these findings, we focused the second study on ctDNA obtained before biopsy in lung patients, i.e. when a tentative diagnosis of lung cancer had been made, but no treatment had started. The main objective of this prospective study was to evaluate use of ctDNA in diagnosis, investigating the concordance of biopsy and ctDNA-derived mutation detection. Here we also found positive correlation between diagnostic lung biopsy results and pre-biopsy ctDNA sequencing, providing support for using ctDNA as a cost-effective, non-invasive solution when the tumour is inaccessible or when biopsy poses significant risk to the patient.
Collapse
Affiliation(s)
- Pamela J. Kaisaki
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- National Institute for Health Research Oxford Biomedical Research Centre (NIHR Oxford BRC), Oxford, United Kingdom
- * E-mail:
| | - Anthony Cutts
- Biomedical Research Centre/National Health Service Translational Molecular Diagnostics Centre, Oxford University Hospitals, John Radcliffe Hospital, Oxford, United Kingdom
| | - Niko Popitsch
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- National Institute for Health Research Oxford Biomedical Research Centre (NIHR Oxford BRC), Oxford, United Kingdom
| | - Carme Camps
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- National Institute for Health Research Oxford Biomedical Research Centre (NIHR Oxford BRC), Oxford, United Kingdom
| | - Melissa M. Pentony
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- National Institute for Health Research Oxford Biomedical Research Centre (NIHR Oxford BRC), Oxford, United Kingdom
| | - Gareth Wilson
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- National Institute for Health Research Oxford Biomedical Research Centre (NIHR Oxford BRC), Oxford, United Kingdom
| | - Suzanne Page
- Biomedical Research Centre/National Health Service Translational Molecular Diagnostics Centre, Oxford University Hospitals, John Radcliffe Hospital, Oxford, United Kingdom
| | - Kulvinder Kaur
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- National Institute for Health Research Oxford Biomedical Research Centre (NIHR Oxford BRC), Oxford, United Kingdom
| | - Dimitris Vavoulis
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- National Institute for Health Research Oxford Biomedical Research Centre (NIHR Oxford BRC), Oxford, United Kingdom
- Biomedical Research Centre/National Health Service Translational Molecular Diagnostics Centre, Oxford University Hospitals, John Radcliffe Hospital, Oxford, United Kingdom
| | - Shirley Henderson
- Biomedical Research Centre/National Health Service Translational Molecular Diagnostics Centre, Oxford University Hospitals, John Radcliffe Hospital, Oxford, United Kingdom
| | - Avinash Gupta
- National Institute for Health Research Oxford Biomedical Research Centre (NIHR Oxford BRC), Oxford, United Kingdom
- Department of Oncology, University of Oxford, Churchill Hospital, Oxford, United Kingdom
| | - Mark R. Middleton
- National Institute for Health Research Oxford Biomedical Research Centre (NIHR Oxford BRC), Oxford, United Kingdom
- Department of Oncology, University of Oxford, Churchill Hospital, Oxford, United Kingdom
| | - Ioannis Karydis
- National Institute for Health Research Oxford Biomedical Research Centre (NIHR Oxford BRC), Oxford, United Kingdom
- Department of Oncology, University of Oxford, Churchill Hospital, Oxford, United Kingdom
| | - Denis C. Talbot
- Department of Oncology, University of Oxford, Churchill Hospital, Oxford, United Kingdom
| | - Anna Schuh
- Biomedical Research Centre/National Health Service Translational Molecular Diagnostics Centre, Oxford University Hospitals, John Radcliffe Hospital, Oxford, United Kingdom
| | - Jenny C. Taylor
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- National Institute for Health Research Oxford Biomedical Research Centre (NIHR Oxford BRC), Oxford, United Kingdom
| |
Collapse
|
1113
|
Carpinetti P, Donnard E, Bettoni F, Asprino P, Koyama F, Rozanski A, Sabbaga J, Habr-Gama A, Parmigiani RB, Galante PAF, Perez RO, Camargo AA. The use of personalized biomarkers and liquid biopsies to monitor treatment response and disease recurrence in locally advanced rectal cancer after neoadjuvant chemoradiation. Oncotarget 2016; 6:38360-71. [PMID: 26451609 PMCID: PMC4742005 DOI: 10.18632/oncotarget.5256] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 09/24/2015] [Indexed: 12/18/2022] Open
Abstract
Neoadjuvant chemoradiotherapy (nCRT) followed by surgery is the mainstay treatment for locally advanced rectal cancer. Variable degrees of tumor regression are observed after nCRT and alternative treatment strategies, including close surveillance without immediate surgery, have been investigated to spare patients with complete tumor regression from potentially adverse outcomes of radical surgery. However, clinical and radiological assessment of response does not allow accurate identification of patients with complete response. In addition, surveillance for recurrence is similarly important for these patients, as early detection of recurrence allows salvage resections and adjuvant interventions. We report the use of liquid biopsies and personalized biomarkers for monitoring treatment response to nCRT and detecting residual disease and recurrence in patients with rectal cancer. We sequenced the whole-genome of four rectal tumors to identify patient-specific chromosomal rearrangements that were used to monitor circulating tumor DNA (ctDNA) in liquid biopsies collected at diagnosis and during nCRT and follow-up. We compared ctDNA levels to clinical, radiological and pathological response to nCRT. Our results indicate that personalized biomarkers and liquid biopsies may not be sensitive for the detection of microscopic residual disease. However, it can be efficiently used to monitor treatment response to nCRT and detect disease recurrence, preceding increases in CEA levels and radiological diagnosis. Similar good results were observed when assessing tumor response to systemic therapy and disease progression. Our study supports the use of personalized biomarkers and liquid biopsies to tailor the management of rectal cancer patients, however, replication in a larger cohort is necessary to introduce this strategy into clinical practice.
Collapse
Affiliation(s)
- Paola Carpinetti
- Ludwig Institute for Cancer Research, São Paulo, SP, Brazil.,Centro de Oncologia Molecular Hospital Sírio Libanês, São Paulo, SP, Brazil.,Programa de Pós Graduação em Bioquímica, Instituto de Química, Universidade de São Paulo, SP, Brazil
| | - Elisa Donnard
- Ludwig Institute for Cancer Research, São Paulo, SP, Brazil.,Centro de Oncologia Molecular Hospital Sírio Libanês, São Paulo, SP, Brazil.,Programa de Pós Graduação em Bioquímica, Instituto de Química, Universidade de São Paulo, SP, Brazil
| | - Fabiana Bettoni
- Centro de Oncologia Molecular Hospital Sírio Libanês, São Paulo, SP, Brazil
| | - Paula Asprino
- Centro de Oncologia Molecular Hospital Sírio Libanês, São Paulo, SP, Brazil
| | | | - Andrei Rozanski
- Centro de Oncologia Molecular Hospital Sírio Libanês, São Paulo, SP, Brazil
| | - Jorge Sabbaga
- Centro de Oncologia Clínica, Hospital Sírio Libanês, São Paulo, SP, Brazil
| | - Angelita Habr-Gama
- Angelita & Joaquim Gama Institute, São Paulo, SP, Brazil.,University of São Paulo, School of Medicine, São Paulo, SP, Brazil
| | | | - Pedro A F Galante
- Centro de Oncologia Molecular Hospital Sírio Libanês, São Paulo, SP, Brazil
| | - Rodrigo O Perez
- Ludwig Institute for Cancer Research, São Paulo, SP, Brazil.,Angelita & Joaquim Gama Institute, São Paulo, SP, Brazil.,University of São Paulo, School of Medicine, São Paulo, SP, Brazil
| | - Anamaria A Camargo
- Ludwig Institute for Cancer Research, São Paulo, SP, Brazil.,Centro de Oncologia Molecular Hospital Sírio Libanês, São Paulo, SP, Brazil
| |
Collapse
|
1114
|
Kristiansen MK, Niemann I, Lindegaard JC, Christiansen M, Joergensen MW, Vogel I, Lildballe DL, Sunde L. Cell-free DNA in pregnancy with choriocarcinoma and coexistent live fetus: A case report. Medicine (Baltimore) 2016; 95:e4721. [PMID: 27631219 PMCID: PMC5402562 DOI: 10.1097/md.0000000000004721] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND This case report describes the use of analysis of cell-free DNA in the blood of a patient with a pregnancy with one live fetus and a choriocarcinoma diagnosed at 22 weeks of gestation. RESULTS The result of the analysis of 16 microsatellite loci on 14 chromosomes in the cell-free DNA in plasma was consistent with the result of the analysis of a tumor biopsy indicating biparental diploid origin of the genome. The DNA markers were discordant with the markers of the placenta indicating two separate conceptions. CONCLUSION Our results indicate that analysis of cell-free DNA in plasma allows determination of the origin of a choriocarcinoma without tissue biopsy, even in the presence of a co-existent pregnancy.
Collapse
Affiliation(s)
- Mona Kjaerboel Kristiansen
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus N
- Correspondence: Mona Kristiansen, Department of Clinical Genetics, Aarhus University Hospital, Brendstrupgaardsvej 21 C, Skejby, DK-8200 Aarhus N, Denmark (e-mail: )
| | - Isa Niemann
- Department of Gynecology and Obstetrics, Aarhus University Hospital, Aarhus N
| | | | | | - Mette Warming Joergensen
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus N
- Institute of Pathology, Aarhus University Hospital, Aarhus C
| | - Ida Vogel
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus N
| | | | - Lone Sunde
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus N
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| |
Collapse
|
1115
|
Waqar SN, Bonomi PD, Govindan R, Hirsch FR, Riely GJ, Papadimitrakopoulou V, Kazandjian D, Khozin S, Larkins E, Dickson DJ, Malik S, Horn L, Ferris A, Shaw AT, Jänne PA, Mok TS, Herbst R, Keegan P, Pazdur R, Blumenthal GM. Clinician Perspectives on Current Issues in Lung Cancer Drug Development. J Thorac Oncol 2016; 11:1387-96. [PMID: 27401214 PMCID: PMC5131641 DOI: 10.1016/j.jtho.2016.05.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 04/26/2016] [Accepted: 05/08/2016] [Indexed: 12/26/2022]
Abstract
Recent advances in molecularly targeted therapy and immunotherapy offer a glimmer of hope for potentially realizing the dream of personalized therapy for lung cancer. This article highlights current questions in clinical trial design, enrollment strategies and patient focused drug development, with particular emphasis on unique issues in trials of targeted therapy and immunotherapy.
Collapse
Affiliation(s)
- Saiama N. Waqar
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | | | - Ramaswamy Govindan
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | | | | | | | | | - Sean Khozin
- U.S. Food and Drug Administration, Silver Spring, MD
| | - Erin Larkins
- U.S. Food and Drug Administration, Silver Spring, MD
| | | | | | - Leora Horn
- Vanderbilt University Medical Center, Nashville, TN
| | | | - Alice T. Shaw
- Massachusetts General Hospital Cancer Center, Boston, MA
| | | | | | | | | | | | | |
Collapse
|
1116
|
Liquid Biopsies. MED GENET-BERLIN 2016. [DOI: 10.1007/s11825-016-0090-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Zusammenfassung
„Liquid Biopsy“ bezieht sich in der Onkologie auf die Untersuchung von zirkulierenden Tumorzellen (engl.: circulating tumor cells [CTCs]) oder zirkulierender Tumor-DNA (engl.: circulating tumor DNA [ctDNA]), um nicht invasiv aus dem peripheren Blut Charakteristika über das Tumorgenom von Krebspatienten zu erfassen. Den Liquid Biopsies wird ein immenses Potenzial für zukünftige Entwicklungen in der personalisierten Medizin und dem Einsatz zielgerichteter Therapien eingeräumt. In dieser Übersichtsarbeit zeigen wir, dass aber noch viele offene Fragen angegangen werden müssen, bevor Liquid Biopsies für die klinische Routine eingesetzt werden können. Eine besondere Herausforderung liegt darin, dass Ergebnisse je nach gewähltem Untersuchungsverfahren eine Tragweite haben können, die weit über die ursprünglich beabsichtigte Untersuchung des Tumorgenoms hinausgeht. All diese Fragen können nur in einem interdisziplinären Ansatz unter Beteiligung der Humangenetik, Onkologie, Pathologie, Bioinformatik und Bioethik angegangen und beantwortet werden.
Collapse
|
1117
|
Vu BT, Tan Le D, Van Pham P. Liquid biopsies: tumour diagnosis and treatment monitoring. BIOMEDICAL RESEARCH AND THERAPY 2016. [DOI: 10.7603/s40730-016-0035-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
1118
|
Ulz P, Thallinger GG, Auer M, Graf R, Kashofer K, Jahn SW, Abete L, Pristauz G, Petru E, Geigl JB, Heitzer E, Speicher MR. Inferring expressed genes by whole-genome sequencing of plasma DNA. Nat Genet 2016; 48:1273-8. [DOI: 10.1038/ng.3648] [Citation(s) in RCA: 212] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 07/22/2016] [Indexed: 12/14/2022]
|
1119
|
Clark P. Serum markers for hepatocellular carcinoma. Clin Liver Dis (Hoboken) 2016; 8:29-33. [PMID: 31041059 PMCID: PMC6490191 DOI: 10.1002/cld.565] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 04/25/2016] [Accepted: 05/06/2016] [Indexed: 02/04/2023] Open
Affiliation(s)
- Paul Clark
- School of MedicineUniversity of QueenslandQueenslandAustralia,Cancer Control UnitQIMR‐Berghofer Medical Research InstituteQueenslandAustralia,Department of GastroenterologyPrincess Alexandra HospitalQueenslandAustralia
| |
Collapse
|
1120
|
Pathologists and liquid biopsies: to be or not to be? Virchows Arch 2016; 469:601-609. [PMID: 27553354 DOI: 10.1007/s00428-016-2004-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 06/25/2016] [Accepted: 08/09/2016] [Indexed: 12/19/2022]
Abstract
Recently, the advent of therapies targeting genomic alterations has improved the care of patients with certain types of cancer. While molecular targets were initially detected in nucleic acid samples extracted from tumor tissue, detection of nucleic acids in circulating blood has allowed the development of what has become known as liquid biopsies, which provide a complementary and alternative sample source allowing identification of genomic alterations that might be addressed by targeted therapy. Consequently, liquid biopsies might rapidly revolutionize oncology practice in allowing administration of more effective treatments. Liquid biopsies also provide an approach towards short-term monitoring of metastatic cancer patients to evaluate efficacy of treatment and/or early detection of secondary mutations responsible for resistance to treatment. In this context, pathologists, who have already been required in recent years to take interest in the domain of molecular pathology of cancer, now face new challenges. The attitude of pathologists to and level of involvement in the practice of liquid biopsies, including mastering the methods employed in molecular analysis of blood samples, need close attention. Regardless of the level of involvement of pathologists in this new field, it is mandatory that oncologists, biologists, geneticists, and pathologists work together to coordinate the pre-analytical, analytical, and post-analytical phases of molecular assessment of tissue and liquid samples of individual cancer patients. The challenges include (1) implementation of effective and efficient procedures for reception and analysis of liquid and tissue samples for histopathological and molecular evaluation and (2) assuring short turn-around times to facilitate rapid optimization of individual patient treatment. In this paper, we will review the following: (1) recent data concerning the concept of liquid biopsies in oncology and its development for patient care, (2) advantages and limitations of molecular analyses performed on blood samples compared to those performed on tissue samples, and (3) short-term challenges facing pathologists in dealing with liquid biopsies of cancer patients and new strategies to early detect metastatic tumor cell clones.
Collapse
|
1121
|
Hao Y, Zhang P, Xuei X, Nakshatri H, Edenberg HJ, Li L, Liu Y. Statistical modeling for sensitive detection of low-frequency single nucleotide variants. BMC Genomics 2016; 17 Suppl 7:514. [PMID: 27556804 PMCID: PMC5001245 DOI: 10.1186/s12864-016-2905-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background Sensitive detection of low-frequency single nucleotide variants carries great significance in many applications. In cancer genetics research, tumor biopsies are a mixture of normal and tumor cells from various subpopulations due to tumor heterogeneity. Thus the frequencies of somatic variants from a subpopulation tend to be low. Liquid biopsies, which monitor circulating tumor DNA in blood to detect metastatic potential, also face the challenge of detecting low-frequency variants due to the small percentage of the circulating tumor DNA in blood. Moreover, in population genetics research, although pooled sequencing of a large number of individuals is cost-effective, pooling dilutes the signals of variants from any individual. Detection of low frequency variants is difficult and can be cofounded by sequencing artifacts. Existing methods are limited in sensitivity and mainly focus on frequencies around 2 % to 5 %; most fail to consider differential sequencing artifacts. Results We aimed to push down the frequency detection limit close to the position specific sequencing error rates by modeling the observed erroneous read counts with respect to genomic sequence contexts. 4 distributions suitable for count data modeling (using generalized linear models) were extensively characterized in terms of their goodness-of-fit as well as the performances on real sequencing data benchmarks, which were specifically designed for testing detection of low-frequency variants; two sequencing technologies with significantly different chemistry mechanisms were used to explore systematic errors. We found the zero-inflated negative binomial distribution generalized linear mode is superior to the other models tested, and the advantage is most evident at 0.5 % to 1 % range. This method is also generalizable to different sequencing technologies. Under standard sequencing protocols and depth given in the testing benchmarks, 95.3 % recall and 79.9 % precision for Ion Proton data, 95.6 % recall and 97.0 % precision for Illumina MiSeq data were achieved for SNVs with frequency > = 1 %, while the detection limit is around 0.5 %. Conclusions Our method enables sensitive detection of low-frequency single nucleotide variants across different sequencing platforms and will facilitate research and clinical applications such as pooled sequencing, cancer early detection, prognostic assessment, metastatic monitoring, and relapses or acquired resistance identification. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2905-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yangyang Hao
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Pengyue Zhang
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Xiaoling Xuei
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Harikrishna Nakshatri
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Howard J Edenberg
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Lang Li
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
1122
|
Yuan J. Circulating protein and antibody biomarker for personalized cancer immunotherapy. J Immunother Cancer 2016; 4:46. [PMID: 27532021 PMCID: PMC4986378 DOI: 10.1186/s40425-016-0150-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 07/18/2016] [Indexed: 11/10/2022] Open
Abstract
Immune checkpoint blockade therapies are revolutionizing standard cancer treatments. Immune checkpoint inhibitors likely function to enhance the tumor specific antigen response in order to achieve favorable clinical outcomes. Thus, continuous efforts to identify the common tumor-specific antigens are essential for the broad clinical application of these therapies. Several immunoproteomics approaches have been used in order to screen for this specificity. In a recent article from Jhaveri and colleagues published in the February issue of Cancer Immunology Research, antibody biomarkers were screened in pancreatic cancer patients who received allogeneic, granulocyte-macrophage colony stimulating factor-secreting pancreatic cancer vaccine (GVAX) by using a serum antibody-based SILAC immunoprecipitation (SASI) approach. Using this assay, several new tumor antigens (MYPT1, PSMC5 and TRFR) were identified that were found to have significantly different expression in tumors compared with normal tissue. Moreover, patients with detectable antibodies showed improved disease-free survival after GVAX therapy. These targets need to be further validated to determine the full spectrum of tumor antigen immunogencity and their potential clinical application. In addition to antibodies, circulating protein, DNA and RNA in peripheral blood are under clinical investigation as liquid biopsies and have the potential to provide guidance for future personalized cancer immunotherapy.
Collapse
Affiliation(s)
- Jianda Yuan
- Oncology Clinical Research, Merck Research Laboratories, Rahway, 07065 NJ USA
| |
Collapse
|
1123
|
Neueste technologische Entwicklungen für die Analyse von zirkulierender Tumor-DNA. MED GENET-BERLIN 2016. [DOI: 10.1007/s11825-016-0089-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Zusammenfassung
Die Analyse von zirkulierender Tumor-DNA, zusammen mit der Analyse von zirkulierenden Tumorzellen auch oft Liquid Biopsy genannt, ist ein sich rasch entwickelndes Feld in der medizinischen Forschung. Obwohl es von der Entdeckung der zellfreien DNA bis hin zur Erkenntnis, dass sie sich als Biomarker eignet, Jahrzehnte gedauert hat, wurde der klinische Nutzen der ctDNA hinsichtlich der Überwachung des Therapieansprechens, der Identifizierung von Resistenzmechanismen und neu aufkommenden Therapiezielen sowie der Detektion von minimaler Resterkrankung mittlerweile in unzähligen Studien bewiesen.
Aufgrund der hohen Variabilität, mit der ctDNA in der Zirkulation vorkommt, sowie der starken Fragmentierung, stellt die ctDNA aber einen schwierigen Analyten dar. In den letzten Jahren haben erhebliche technologische Fortschritte dazu beigetragen, dass eine Routineanwendung der ctDNA-Analysen tatsächlich realisierbar wird, sofern eine Reihe von regulatorischen Hürden überwunden wird.
Collapse
|
1124
|
Biosensors for liquid biopsy: circulating nucleic acids to diagnose and treat cancer. Anal Bioanal Chem 2016; 408:7255-64. [PMID: 27497966 DOI: 10.1007/s00216-016-9806-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 07/05/2016] [Accepted: 07/18/2016] [Indexed: 01/05/2023]
Abstract
The detection of cancer biomarkers freely circulating in blood offers new opportunities for cancer early diagnosis, patient follow-up, and therapy efficacy assessment based on liquid biopsy. In particular, circulating cell-free nucleic acids released from tumor cells have recently attracted great attention also because they become detectable in blood before the appearance of other circulating biomarkers, such as circulating tumor cells. The detection of circulating nucleic acids poses several technical challenges that arise from their low concentration and relatively small size. Here, possibilities offered by innovative biosensing approaches for the detection of circulating DNA in peripheral blood and blood-derived products such as plasma and serum blood are discussed. Different transduction principles are used to detect circulating DNAs and great advantages are derived from the combined use of nanostructured materials.
Collapse
|
1125
|
Ma Y, Luk A, Young FP, Lynch D, Chua W, Balakrishnar B, de Souza P, Becker TM. Droplet Digital PCR Based Androgen Receptor Variant 7 (AR-V7) Detection from Prostate Cancer Patient Blood Biopsies. Int J Mol Sci 2016; 17:E1264. [PMID: 27527157 PMCID: PMC5000662 DOI: 10.3390/ijms17081264] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 07/25/2016] [Accepted: 07/27/2016] [Indexed: 11/26/2022] Open
Abstract
Androgen receptor splice variant V7 (AR-V7) was recently identified as a valuable predictive biomarker in metastatic castrate-resistant prostate cancer. Here, we report a new, sensitive and accurate screen for AR-V7 mRNA expression directly from circulating tumor cells (CTCs): We combined EpCAM-based immunomagnetic CTC isolation using the IsoFlux microfluidic platform with droplet digital polymerase chain reaction (ddPCR) to analyze total AR and AR-V7 expression from prostate cancer patients CTCs. We demonstrate that AR-V7 is reliably detectable in enriched CTC samples with as little as five CTCs, even considering tumor heterogeneity, and confirm detection of AR-V7 in CTC samples from advanced prostate cancer (PCa) patients with AR-V7 detection limited to castrate resistant disease status in our sample set. Sensitive molecular analyses of circulating tumor cells (CTCs) or circulating tumor nucleic acids present exciting strategies to detect biomarkers, such as AR-V7 from non-invasive blood samples, so-called blood biopsies.
Collapse
Affiliation(s)
- Yafeng Ma
- Centre for Circulating Tumor Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, 1 Campbell St., Liverpool, NSW 2170, Australia.
| | - Alison Luk
- Centre for Circulating Tumor Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, 1 Campbell St., Liverpool, NSW 2170, Australia.
| | - Francis P Young
- Centre for Circulating Tumor Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, 1 Campbell St., Liverpool, NSW 2170, Australia.
- South Western Clinical School, University of New South Wales, Goulburn St., Liverpool, NSW 2170, Australia.
| | - David Lynch
- Centre for Circulating Tumor Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, 1 Campbell St., Liverpool, NSW 2170, Australia.
- Western Sydney University Clinical School, Elizabeth St., Liverpool, NSW 2170, Australia.
| | - Wei Chua
- Department of Medical Oncology, Liverpool Hospital, Elizabeth St & Goulburn St., Liverpool, NSW 2170, Australia.
| | - Bavanthi Balakrishnar
- Department of Medical Oncology, Liverpool Hospital, Elizabeth St & Goulburn St., Liverpool, NSW 2170, Australia.
| | - Paul de Souza
- Centre for Circulating Tumor Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, 1 Campbell St., Liverpool, NSW 2170, Australia.
- South Western Clinical School, University of New South Wales, Goulburn St., Liverpool, NSW 2170, Australia.
- Western Sydney University Clinical School, Elizabeth St., Liverpool, NSW 2170, Australia.
- Department of Medical Oncology, Liverpool Hospital, Elizabeth St & Goulburn St., Liverpool, NSW 2170, Australia.
| | - Therese M Becker
- Centre for Circulating Tumor Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, 1 Campbell St., Liverpool, NSW 2170, Australia.
- South Western Clinical School, University of New South Wales, Goulburn St., Liverpool, NSW 2170, Australia.
- Western Sydney University Clinical School, Elizabeth St., Liverpool, NSW 2170, Australia.
| |
Collapse
|
1126
|
Doherty M, Metcalfe T, Guardino E, Peters E, Ramage L. Precision medicine and oncology: an overview of the opportunities presented by next-generation sequencing and big data and the challenges posed to conventional drug development and regulatory approval pathways. Ann Oncol 2016; 27:1644-6. [DOI: 10.1093/annonc/mdw165] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
1127
|
Urinary circulating DNA detection for dynamic tracking of EGFR mutations for NSCLC patients treated with EGFR-TKIs. Clin Transl Oncol 2016; 19:332-340. [PMID: 27468867 DOI: 10.1007/s12094-016-1534-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 07/20/2016] [Indexed: 10/21/2022]
Abstract
PURPOSE Changes in EGFR profiles of non small cell lung cancer (NSCLC) patients correlates to clinical outcome. Extracting quality tumor tissue remains a challenge for molecular profiling. Our study aims to ascertain the clinical relevance of urinary cell free DNA as an alternative tumor material source. METHODS 150 patients with activating EGFR mutation and received EGFR-TKIs were recruited to participate in the serial monitoring study. Matched primary tumor samples were taken together with blood and urine specimens before the initiation of TKIs. The EGFR mutation testing was performed and quantified using ddPCR. For serial time point measurements, urine and blood samples were extracted at 1-month intervals for duration of 9 months. RESULTS Urinary ctDNA yielded a close agreement of 88 % on EGFR mutation status when compared to primary tissue at baseline. Almost all samples detected via urine specimens were uncovered in plasma samples. Analysis of urinary cell free DNA at different time points showed a strong correlation to treatment efficacy. Interestingly, a secondary EGFR mutation T790M was detected for 53 % of the patients during monitoring. The results were corroborated with the plasma ctDNA analysis. The T790M+ group had a reduced median survival when compared to the wildtype group. CONCLUSION Urinary cell free DNA may be a potential alternative to conventional primary tissue based EGFR mutation testing. Our findings showed that the assay sensitivity was comparable to results from blood plasma. Urinary samples being noninvasive and readily available have clinical utility for monitoring of EGFR TKI treatment.
Collapse
|
1128
|
Macedo JE. New era of epidermal growth factor receptor-tyrosine kinase inhibitors for lung cancer. World J Respirol 2016; 6:57-62. [DOI: 10.5320/wjr.v6.i2.57] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 06/29/2016] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is the leading cause of death globally, besides recent advances in its management; it maintains a low 5-year survival rate of 15%. The discovery of epidermal growth factor receptor (EGFR) activating mutations and the introduction of its tyrosine kinase inhibitors (TKIs) have expanded the treatment options for patients with non-small cell lung cancer. Nowadays, EGFR mutation testing is now a common routine for newly diagnosed lung cancer. First generation TKIs developed, erlotinib and gefitinib, were reversible ones. After a median of 14 mo, eventually all EGFR mutated patients develop resistance to reversible TKIs. Afatinib, dacomitinib and neratinib, second generation inhibitors, are selective and irreversible TKIs. Finally, third generation phase I clinical trials were performed, with lower toxicity profiles, and targeting with more precision the driving clone of this heterogeneous disease.
Collapse
|
1129
|
He K, Chen D, Ruan H, Li X, Tong J, Xu X, Zhang L, Yu J. BRAFV600E-dependent Mcl-1 stabilization leads to everolimus resistance in colon cancer cells. Oncotarget 2016; 7:47699-47710. [PMID: 27351224 PMCID: PMC5216972 DOI: 10.18632/oncotarget.10277] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 06/09/2016] [Indexed: 01/06/2023] Open
Abstract
mTOR activation is commonly caused by oncogenic mutations in RAS/RAF/MAPK and PI3K/AKT pathways, and promotes cancer progression and therapeutic resistance. However, mTOR inhibitors show limited single agent efficacy in patients. mTOR inhibitors suppress tumor cell growth and angiogenesis, and have recently been shown to induce death receptor/FADD-dependent apoptosis in colon cancers. Using a panel of BRAF V600E and WT colorectal cancer cell lines and in vitro selected resistant culture, and xenograft models, we demonstrate here that BRAFV600E confers resistance to mTOR inhibitors. Everolimus treatment disrupts the S6K1-IRS-2/PI3K negative feedback loop, leading to BRAF V600E-dependent activation of ERK and Mcl-1 stabilization in colon cancer cells, which in turn blocks the crosstalk from the death receptor to mitochondria. Co-treatment with inhibitors to Mcl-1, PI3K, RAF or MEK restores mTOR inhibitor-induced apoptosis by antagonizing Mcl-1 or abrogating ERK activation in BRAFV600E cells. Our findings provide a rationale for genotype-guided patient stratification and potential drug combinations to prevent or mitigate undesired activation of survival pathways induced by mTOR inhibitors.
Collapse
Affiliation(s)
- Kan He
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Dongshi Chen
- University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Hang Ruan
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Xiangyun Li
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- The Third Military Medical University Daping Hospital, Daping, Yu Zhong District, Chongqing 400042, P.R. China
| | - Jingshan Tong
- University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Xiang Xu
- The Third Military Medical University Daping Hospital, Daping, Yu Zhong District, Chongqing 400042, P.R. China
| | - Lin Zhang
- University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jian Yu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
1130
|
Li Z, Guo X, Tang L, Peng L, Chen M, Luo X, Wang S, Xiao Z, Deng Z, Dai L, Xia K, Wang J. Methylation analysis of plasma cell-free DNA for breast cancer early detection using bisulfite next-generation sequencing. Tumour Biol 2016; 37:13111-13119. [PMID: 27449045 DOI: 10.1007/s13277-016-5190-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 07/13/2016] [Indexed: 12/15/2022] Open
Abstract
Circulating cell-free DNA (cfDNA) has been considered as a potential biomarker for non-invasive cancer detection. To evaluate the methylation levels of six candidate genes (EGFR, GREM1, PDGFRB, PPM1E, SOX17, and WRN) in plasma cfDNA as biomarkers for breast cancer early detection, quantitative analysis of the promoter methylation of these genes from 86 breast cancer patients and 67 healthy controls was performed by using microfluidic-PCR-based target enrichment and next-generation bisulfite sequencing technology. The predictive performance of different logistic models based on methylation status of candidate genes was investigated by means of the area under the ROC curve (AUC) and odds ratio (OR) analysis. Results revealed that EGFR, PPM1E, and 8 gene-specific CpG sites showed significantly hypermethylation in cancer patients' plasma and significantly associated with breast cancer (OR ranging from 2.51 to 9.88). The AUC values for these biomarkers were ranging from 0.66 to 0.75. Combinations of multiple hypermethylated genes or CpG sites substantially improved the predictive performance for breast cancer detection. Our study demonstrated the feasibility of quantitative measurement of candidate gene methylation in cfDNA by using microfluidic-PCR-based target enrichment and bisulfite next-generation sequencing, which is worthy of further validation and potentially benefits a broad range of applications in clinical oncology practice. Quantitative analysis of methylation pattern of plasma cfDNA by next-generation sequencing might be a valuable non-invasive tool for early detection of breast cancer.
Collapse
Affiliation(s)
- Zibo Li
- The State Key Laboratory of Medical Genetics and School of Life Sciences, Central South University, 172 Tongzipo Road, Changsha, Hunan, 410013, China
| | - Xinwu Guo
- Sanway Gene Technology Inc., Changsha, Hunan, 410205, China
| | - Lili Tang
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Limin Peng
- Sanway Gene Technology Inc., Changsha, Hunan, 410205, China
| | - Ming Chen
- Sanway Gene Technology Inc., Changsha, Hunan, 410205, China
| | - Xipeng Luo
- Sanway Gene Technology Inc., Changsha, Hunan, 410205, China
| | - Shouman Wang
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Zhi Xiao
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Zhongping Deng
- Sanway Gene Technology Inc., Changsha, Hunan, 410205, China
- Research Center for Technologies in Nucleic Acid-Based Diagnostics, Changsha, Hunan, 410205, China
- Research Center for Technologies in Nucleic Acid-Based Diagnostics and Therapeutics, Changsha, Hunan, 410205, China
| | - Lizhong Dai
- Sanway Gene Technology Inc., Changsha, Hunan, 410205, China
- Research Center for Technologies in Nucleic Acid-Based Diagnostics, Changsha, Hunan, 410205, China
- Research Center for Technologies in Nucleic Acid-Based Diagnostics and Therapeutics, Changsha, Hunan, 410205, China
| | - Kun Xia
- The State Key Laboratory of Medical Genetics and School of Life Sciences, Central South University, 172 Tongzipo Road, Changsha, Hunan, 410013, China
| | - Jun Wang
- The State Key Laboratory of Medical Genetics and School of Life Sciences, Central South University, 172 Tongzipo Road, Changsha, Hunan, 410013, China.
| |
Collapse
|
1131
|
The "Liquid Biopsy": the Role of Circulating DNA and RNA in Central Nervous System Tumors. Curr Neurol Neurosci Rep 2016; 16:25. [PMID: 26838352 DOI: 10.1007/s11910-016-0629-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The detection of tumor-derived circulating nucleic acids in patients with cancer, known as the "liquid biopsy," has expanded from use in plasma to other bodily fluids in an increasing number of malignancies. Circulating nucleic acids could be of particular use in central nervous system tumors as biopsy carries a 5-7 % risk of major morbidity. This application presents unique challenges that have limited the use of cell-free DNA and RNA in the diagnosis and monitoring of CNS tumors. Recent work suggests that cerebrospinal fluid may be a useful source of CNS tumor-derived circulating nucleic acids. In this review, we discuss the available data and future outlook on the use of the liquid biopsy for CNS tumors.
Collapse
|
1132
|
Alcaide M, Yu S, Bushell K, Fornika D, Nielsen JS, Nelson BH, Mann KK, Assouline S, Johnson NA, Morin RD. Multiplex Droplet Digital PCR Quantification of Recurrent Somatic Mutations in Diffuse Large B-Cell and Follicular Lymphoma. Clin Chem 2016; 62:1238-47. [PMID: 27440511 DOI: 10.1373/clinchem.2016.255315] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/23/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND A plethora of options to detect mutations in tumor-derived DNA currently exist but each suffers limitations in analytical sensitivity, cost, or scalability. Droplet digital PCR (ddPCR) is an appealing technology for detecting the presence of specific mutations based on a priori knowledge and can be applied to tumor biopsies, including formalin-fixed paraffin embedded (FFPE) tissues. More recently, ddPCR has gained popularity in its utility in quantifying circulating tumor DNA. METHODS We have developed a suite of novel ddPCR assays for detecting recurrent mutations that are prevalent in common B-cell non-Hodgkin lymphomas (NHLs), including diffuse large B-cell lymphoma, follicular lymphoma, and lymphoplasmacytic lymphoma. These assays allowed the differentiation and counting of mutant and wild-type molecules using one single hydrolysis probe. We also implemented multiplexing that allowed the simultaneous detection of distinct mutations and an "inverted" ddPCR assay design, based on employing probes matching wild-type alleles, capable of detecting the presence of multiple single nucleotide polymorphisms. RESULTS The assays successfully detected and quantified somatic mutations commonly affecting enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) (Y641) and signal transducer and activator of transcription 6 (STAT6) (D419) hotspots in fresh tumor, FFPE, and liquid biopsies. The "inverted" ddPCR approach effectively reported any single nucleotide variant affecting either of these 2 hotspots as well. Finally, we could effectively multiplex hydrolysis probes targeting 2 additional lymphoma-related hotspots: myeloid differentiation primary response 88 (MYD88; L265P) and cyclin D3 (CCND3; I290R). CONCLUSIONS Our suite of ddPCR assays provides sufficient analytical sensitivity and specificity for either the invasive or noninvasive detection of multiple recurrent somatic mutations in B-cell NHLs.
Collapse
Affiliation(s)
- Miguel Alcaide
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Stephen Yu
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Kevin Bushell
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Daniel Fornika
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Julie S Nielsen
- Deeley Research Centre, BC Cancer Agency, Victoria, BC, Canada
| | - Brad H Nelson
- Deeley Research Centre, BC Cancer Agency, Victoria, BC, Canada
| | - Koren K Mann
- Department of Medicine, Jewish General Hospital, Montreal, Quebec, Canada
| | - Sarit Assouline
- Department of Medicine, Jewish General Hospital, Montreal, Quebec, Canada
| | - Nathalie A Johnson
- Department of Medicine, Jewish General Hospital, Montreal, Quebec, Canada
| | - Ryan D Morin
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada; Genome Sciences Centre, BC Cancer Agency, Vancouver, BC, Canada.
| |
Collapse
|
1133
|
Imamura T, Komatsu S, Ichikawa D, Kawaguchi T, Miyamae M, Okajima W, Ohashi T, Arita T, Konishi H, Shiozaki A, Morimura R, Ikoma H, Okamoto K, Otsuji E. Liquid biopsy in patients with pancreatic cancer: Circulating tumor cells and cell-free nucleic acids. World J Gastroenterol 2016; 22:5627-5641. [PMID: 27433079 PMCID: PMC4932201 DOI: 10.3748/wjg.v22.i25.5627] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 05/25/2016] [Accepted: 06/15/2016] [Indexed: 02/06/2023] Open
Abstract
Despite recent advances in surgical techniques and perioperative management, the prognosis of pancreatic cancer (PCa) remains extremely poor. To provide optimal treatment for each patient with Pca, superior biomarkers are urgently needed in all phases of management from early detection to staging, treatment monitoring, and prognosis. In the blood of patients with cancer, circulating tumor cells (CTCs) and cell-free nucleic acids (cfNAs), such as DNA, mRNA, and noncoding RNA have been recognized. In the recent years, their presence in the blood has encouraged researchers to investigate their potential use as novel blood biomarkers, and numerous studies have demonstrated their potential clinical utility as a biomarker for certain types of cancer. This concept, called “liquid biopsy” has been focused on as a less invasive, alternative approach to cancer tissue biopsy for obtaining genetic and epigenetic aberrations that contribute to oncogenesis and cancer progression. In this article, we review the available literature on CTCs and cfNAs in patients with cancer, particularly focusing on PCa, and discuss future perspectives in this field.
Collapse
|
1134
|
Khoo BL, Grenci G, Jing T, Lim YB, Lee SC, Thiery JP, Han J, Lim CT. Liquid biopsy and therapeutic response: Circulating tumor cell cultures for evaluation of anticancer treatment. SCIENCE ADVANCES 2016; 2:e1600274. [PMID: 27453941 PMCID: PMC4956185 DOI: 10.1126/sciadv.1600274] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 06/14/2016] [Indexed: 05/08/2023]
Abstract
The lack of a robust anticancer drug screening system to monitor patients during treatment delays realization of personalized treatment. We demonstrate an efficient approach to evaluate drug response using patient-derived circulating tumor cell (CTC) cultures obtained from liquid biopsy. Custom microfabricated tapered microwells were integrated with microfluidics to allow robust formation of CTC clusters without pre-enrichment and subsequent drug screening in situ. Rapid feedback after 2 weeks promotes immediate intervention upon detection of drug resistance or tolerance. The procedure was clinically validated with blood samples (n = 73) from 55 patients with early-stage, newly diagnosed, locally advanced, or refractory metastatic breast cancer. Twenty-four of these samples were used for drug evaluation. Cluster formation potential correlated inversely with increased drug concentration and therapeutic treatment. This new and robust liquid biopsy technique can potentially evaluate patient prognosis with CTC clusters during treatment and provide a noninvasive and inexpensive assessment that can guide drug discovery development or therapeutic choices for personalized treatment.
Collapse
Affiliation(s)
- Bee Luan Khoo
- BioSystems and Micromechanics (BioSyM) IRG, Singapore-MIT Alliance for Research and Technology (SMART) Centre, Singapore 138602, Singapore
| | - Gianluca Grenci
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Tengyang Jing
- BioSystems and Micromechanics (BioSyM) IRG, Singapore-MIT Alliance for Research and Technology (SMART) Centre, Singapore 138602, Singapore
- Department of Biomedical Engineering, National University of Singapore, 7 Engineering Drive 1, Singapore 117574, Singapore
| | - Ying Bena Lim
- Department of Biomedical Engineering, National University of Singapore, 7 Engineering Drive 1, Singapore 117574, Singapore
| | - Soo Chin Lee
- Department of Hematology-Oncology, National University Cancer Institute, National University Hospital, Singapore 119074, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Jean Paul Thiery
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Jongyoon Han
- BioSystems and Micromechanics (BioSyM) IRG, Singapore-MIT Alliance for Research and Technology (SMART) Centre, Singapore 138602, Singapore
- Department of Electrical Engineering and Computer Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Chwee Teck Lim
- BioSystems and Micromechanics (BioSyM) IRG, Singapore-MIT Alliance for Research and Technology (SMART) Centre, Singapore 138602, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
- Department of Biomedical Engineering, National University of Singapore, 7 Engineering Drive 1, Singapore 117574, Singapore
- Department of Mechanical Engineering, National University of Singapore, Singapore 117575, Singapore
- Corresponding author.
| |
Collapse
|
1135
|
Liao W, Yang H, Xu H, Wang Y, Ge P, Ren J, Xu W, Lu X, Sang X, Zhong S, Zhang H, Mao Y. Noninvasive detection of tumor-associated mutations from circulating cell-free DNA in hepatocellular carcinoma patients by targeted deep sequencing. Oncotarget 2016; 7:40481-40490. [PMID: 27248174 PMCID: PMC5130021 DOI: 10.18632/oncotarget.9629] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 05/02/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Detection of circulating cell-free DNA (cfDNA) has potential clinical value for assessing tumor biology in patients with hepatocellular carcinoma (HCC), yet many traditional assays lack robustness. This study was the first to apply a high-throughput sequencing platform to detect tumor-associated mutations in HCC from circulating tumor-derived DNA (ctDNA) and to evaluate the utility and feasibility of this approach. METHODS Using the MiSeq™ system, plasma and matched tumor DNA samples were analyzed for hotspot mutations in the TERT, CTNNB1, and TP53 genes that had been verified as the most prevalent mutations in HCC. We compared tumor and plasma data and prospectively investigated the association between significant mutations detected in ctDNA and the patients' clinical outcomes. RESULTS In 41 patients, we detected tumor-associated mutations for HCC in 8 (19.5%) plasma samples. Among them, one showed a tumor-associated mutation in ctDNA but not in the tumor tissue which we used to detect. We also found that ctDNA with mutations could be detected more easily in patients who suffered vascular invasion (P=0.041) and predicted a shorter recurrence-free survival time (P<0.001). There was no relationship between detectable mutations and concentration of cfDNA (P=0.818). CONCLUSIONS The results of our study suggest that tumor-associated mutations detected in plasma are associated with vascular invasion and might be used to predict a shorter recurrence-free survival time for HCC patients. This kind of biomarker can overcome the limitations of tumor heterogeneity. Moreover, the diagnostic performance is improved if multiple mutations in different genes are combined.
Collapse
Affiliation(s)
- Wenjun Liao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Huayu Yang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Haifeng Xu
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Yanan Wang
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Collaborative Innovation Center for Cancer Medicine, Institute of Basic Medical Sciences and School of Basic Medicine, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Penglei Ge
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Jinjun Ren
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Xu
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Xin Lu
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Xinting Sang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Shouxian Zhong
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Hongbing Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Collaborative Innovation Center for Cancer Medicine, Institute of Basic Medical Sciences and School of Basic Medicine, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
1136
|
Olsen JA, Kenna LA, Tipon RC, Spelios MG, Stecker MM, Akirav EM. A Minimally-invasive Blood-derived Biomarker of Oligodendrocyte Cell-loss in Multiple Sclerosis. EBioMedicine 2016; 10:227-35. [PMID: 27381476 PMCID: PMC5006601 DOI: 10.1016/j.ebiom.2016.06.031] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/15/2016] [Accepted: 06/23/2016] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis (MS) is a neurodegenerative disease of the central nervous system (CNS). Minimally invasive biomarkers of MS are required for disease diagnosis and treatment. Differentially methylated circulating-free DNA (cfDNA) is a useful biomarker for disease diagnosis and prognosis, and may offer to be a viable approach for understanding MS. Here, methylation-specific primers and quantitative real-time PCR were used to study methylation patterns of the myelin oligodendrocyte glycoprotein (MOG) gene, which is expressed primarily in myelin-producing oligodendrocytes (ODCs). MOG-DNA was demethylated in O4+ ODCs in mice and in DNA from human oligodendrocyte precursor cells (OPCs) when compared with other cell types. In the cuprizone-fed mouse model of demyelination, ODC derived demethylated MOG cfDNA was increased in serum and was associated with tissue-wide demyelination, demonstrating the utility of demethylated MOG cfDNA as a biomarker of ODC death. Collected sera from patients with active (symptomatic) relapsing-remitting MS (RRMS) demonstrated a higher signature of demethylated MOG cfDNA when compared with patients with inactive disease and healthy controls. Taken together, these results offer a minimally invasive approach to measuring ODC death in the blood of MS patients that may be used to monitor disease progression. Currently, there are no molecular biomarkers of multiple sclerosis (MS). A minimally invasive assay for measuring oligodendrocyte (ODC) cell loss in relapsing-remitting MS is described. DNA methylation of the myelin oligodendrocyte glycoprotein gene is used as a measure of ODC loss in the blood.
Multiple sclerosis (MS) is a neurodegenerative disease of the central nervous system. Currently, there are no molecular biomarkers of MS, thereby limiting disease diagnosis, prognosis, and assessment of new clinical interventions. Myelin oligodendrocyte glycoprotein (MOG) is expressed solely by oligodendrocytes (ODCs) as an integral part of the myelin sheath. This report describes a minimally invasive biomarker assay for measuring ODC-derived DNA in the blood of MS patients. It describes the presence of unique DNA methylation patterns in the MOG gene in ODCs, which is used to design methylation-specific primers. Analysis of sera from patients with active relapsing-remitting MS shows an increase in levels of ODC-derived circulating free DNA when compared with inactive disease and healthy controls.
Collapse
Affiliation(s)
- John A Olsen
- Research Institute, Islet Biology, Winthrop-University Hospital, Mineola, NY, USA
| | - Lauren A Kenna
- Research Institute, Islet Biology, Winthrop-University Hospital, Mineola, NY, USA
| | - Regine C Tipon
- Research Institute, Islet Biology, Winthrop-University Hospital, Mineola, NY, USA
| | - Michael G Spelios
- Research Institute, Islet Biology, Winthrop-University Hospital, Mineola, NY, USA
| | - Mark M Stecker
- Department of Neuroscience, Winthrop-University Hospital Mineola, NY, USA
| | - Eitan M Akirav
- Research Institute, Islet Biology, Winthrop-University Hospital, Mineola, NY, USA; Stony Brook University School of Medicine, Stony Brook, NY, USA.
| |
Collapse
|
1137
|
Salvi S, Casadio V, Conteduca V, Lolli C, Gurioli G, Martignano F, Schepisi G, Testoni S, Scarpi E, Amadori D, Calistri D, Attard G, Giorgi UD. Circulating AR copy number and outcome to enzalutamide in docetaxel-treated metastatic castration-resistant prostate cancer. Oncotarget 2016; 7:37839-37845. [PMID: 27191887 PMCID: PMC5122353 DOI: 10.18632/oncotarget.9341] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 04/27/2016] [Indexed: 12/11/2022] Open
Abstract
In the present study, we aimed to evaluate the association of circulating AR copy number (CN) and outcome in a cohort of patients with advanced castration-resistant prostate cancer (CRPC) treated with enzalutamide after docetaxel. Fifty-nine CRPC patients were evaluated. AR CN was analyzed with real-time and digital PCR in the serum collected at starting of treatment. Progressive disease was defined on the basis of Prostate Cancer Working Group 2 criteria. AR CN gain was found in 21 of 59 (36%) patients. Median baseline PSA, alkaline phosphatase and lactate dehydrogenase levels were higher in the AR CN gained group (p = 0.007, p = 0.003, p = 0.0009, respectively). Median PFS of patients with AR CN gain was 2.4 (95%CI: 1.9-3.2) vs. 4.0 months (95%CI: 3.0-6.5) of those with no gain (p = 0.0004). Median OS of patients with AR CN gain was 6.1 (95%CI: 3.4-8.6) vs. 14.1 months (95%CI: 8.2-20.5) of those with no gain (p = 0.0003). At multivariate analysis, PSA decline ≥ 50% and AR CN showed a significant association with PFS (p = 0.008 and p = 0.002, respectively) and OS (p = 0.009 and p = 0.001, respectively). These findings indicate that the detection of circulating AR CN gain is a promising non-invasive biomarker for outcome prediction to enzalutamide treatment in CRPC patients.
Collapse
Affiliation(s)
- Samanta Salvi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Valentina Casadio
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Vincenza Conteduca
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Cristian Lolli
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Giorgia Gurioli
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Filippo Martignano
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
- University of Florence, Florence, Italy
| | - Giuseppe Schepisi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Sara Testoni
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Emanuela Scarpi
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Dino Amadori
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Daniele Calistri
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Gerhardt Attard
- The Institute of Cancer Research and The Royal Marsden, London, UK
| | - Ugo De Giorgi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| |
Collapse
|
1138
|
Zheng D, Chen H. [Advances in Liquid Biopsy and its Clinical Application in the Diagnosis
and Treatment of Non-small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2016; 19:394-8. [PMID: 27335304 PMCID: PMC6015208 DOI: 10.3779/j.issn.1009-3419.2016.06.19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
随着近几年科学技术的进步,液体活检技术也有了长足的发展,并在肿瘤的早期诊断及后期治疗中扮演着越来越重要的角色。相比于传统的组织活检,液体活检以其独有的无创性、便捷性、高重复性等特点在临床上得到更多的青睐,在未来有着巨大的发展潜力。本文重点探讨了循环肿瘤细胞(circulating tumor cells, CTCs)和循环肿瘤DNA(circulating tumor DNA, ctDNA),作为液体活检最重要的两个检测对象,其历史、生物学特性,检测手段,局限性及其在非小细胞肺癌诊治中的应用。
Collapse
Affiliation(s)
- Difan Zheng
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Haiquan Chen
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| |
Collapse
|
1139
|
Takahashi H, Kagara N, Tanei T, Naoi Y, Shimoda M, Shimomura A, Shimazu K, Kim SJ, Noguchi S. Correlation of Methylated Circulating Tumor DNA With Response to Neoadjuvant Chemotherapy in Breast Cancer Patients. Clin Breast Cancer 2016; 17:61-69.e3. [PMID: 27395416 DOI: 10.1016/j.clbc.2016.06.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 05/19/2016] [Accepted: 06/08/2016] [Indexed: 12/18/2022]
Abstract
BACKGROUND Circulating tumor DNA (ctDNA) is known to harbor tumor-specific genetic or epigenetic alterations. In the present study, the correlation of ctDNA with tumor response to neoadjuvant chemotherapy (NAC) was evaluated in primary breast cancer patients. PATIENTS AND METHODS Plasma samples were obtained from 87 primary breast cancer patients (stage II-III) before and after NAC, as well as 1 year after surgery. Methylated ctDNA (met-ctDNA) was determined by one-step methylation-specific PCR (OS-MSP) for the promoter region of RASSF1A. RESULTS The positivity (23.0%, 20/87) of met-ctDNA before NAC was significantly (P < .05) higher than that of carcinoembryonic antigen (CEA) (8.6%) and cancer-associated antigen (CA) 15-3 (7.4%). In the patients with positive met-ctDNA before NAC, met-ctDNA significantly decreased after NAC in those with disease that responded to therapy (P = .006), but not in patients whose disease did not respond to therapy. Met-ctDNA after NAC was found to be significantly (P = .008) correlated to the extent of residual tumor burden. Of the 7 patients who showed an increase in met-ctDNA at 1 year after surgery, 3 developed recurrence. CONCLUSION Met-ctDNA is a more sensitive marker than CEA and CA15-3, and it might be useful in monitoring the clinical tumor response to NAC. In addition, the potential use of met-ctDNA as a tumor marker for monitoring postoperative recurrence has been suggested.
Collapse
Affiliation(s)
- Hiroyo Takahashi
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Naofumi Kagara
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Osaka, Japan.
| | - Tomonori Tanei
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasuto Naoi
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masafumi Shimoda
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Atsushi Shimomura
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kenzo Shimazu
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Seung Jin Kim
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shinzaburo Noguchi
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
1140
|
Xia Y, Huang CC, Dittmar R, Du M, Wang Y, Liu H, Shenoy N, Wang L, Kohli M. Copy number variations in urine cell free DNA as biomarkers in advanced prostate cancer. Oncotarget 2016; 7:35818-35831. [PMID: 27127882 PMCID: PMC5094965 DOI: 10.18632/oncotarget.9027] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 04/16/2016] [Indexed: 01/14/2023] Open
Abstract
Genetic profiling of urine cell free DNA (cfDNA) has not been evaluated in advanced prostate cancer. We performed whole genome sequencing of urine cfDNAs to identify tumor-associated copy number variations in urine before and after initiating androgen deprivation therapy in HSPC stage and docetaxel chemotherapy in CRPC stage. A log2 ratio-based copy number analysis detected common genomic abnormalities in prostate cancer including AR amplification in 5/10 CRPC patients. Other abnormalities identified included TMPRSS2-ERG fusion, PTEN gene deletion, NOTCH1 locus amplification along with genomic amplifications at 8q24.3, 9q34.3, 11p15.5 and 14q11.2, and deletions at 4q35.2, 5q31.3, 7q36.3, 12q24.33, and 16p11.2. By comparing copy number between pre- and post-treatment, we found significant copy number changes in 34 genomic loci. To estimate the somatic tumor DNA fraction in urine cfDNAs, we developed a Urine Genomic Abnormality (UGA) score algorithm that summed the top ten most significant segments with copy number changes. The UGA scores correlated with tumor burden and the change in UGA score after stage-specific therapies reflected disease progression status and overall survival. The study demonstrates the potential clinical utility of urine cfDNAs in predicting treatment response and monitoring disease progression.
Collapse
Affiliation(s)
- Yun Xia
- Department of General Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Pathology and MCW Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Chiang-Ching Huang
- Joseph J. Zilber School of Public Health, University of Wisconsin, Milwaukee, WI, USA
| | - Rachel Dittmar
- Department of Pathology and MCW Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Meijun Du
- Department of Pathology and MCW Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yuan Wang
- Department of Pathology and MCW Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hongyan Liu
- Department of Pathology and MCW Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Niraj Shenoy
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Liang Wang
- Department of Pathology and MCW Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Manish Kohli
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
1141
|
Touat M, Duran-Peña A, Alentorn A, Lacroix L, Massard C, Idbaih A. Emerging circulating biomarkers in glioblastoma: promises and challenges. Expert Rev Mol Diagn 2016; 15:1311-23. [PMID: 26394701 DOI: 10.1586/14737159.2015.1087315] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Glioblastoma (GBM) is the most common and devastating primary malignant brain tumor in adults. The past few years have seen major progress in our understanding of the molecular basis of GBM. These advances, which have contributed to the development of novel targeted therapies, will change the paradigms in GBM therapy from disease-based to individually tailored molecular target-based treatment. No validated circulating biomarkers have yet been integrated into clinical practice for GBM. There is thus a critical need to implement minimally invasive clinical tests enabling molecular stratification and prognosis assessment, as well as the prediction and monitoring of treatment response. After examination of data from recent studies exploring several categories of tumor-associated biomarkers (circulating tumor cells, extracellular vesicles, nucleic acids and oncometabolites) identified in the blood, cerebrospinal fluid and urine, this article discusses the challenges and prospects for the development of circulating biomarkers in GBM.
Collapse
Affiliation(s)
- Mehdi Touat
- a 1 Inserm U981, Université Paris Sud, Gustave Roussy, F-94805 Villejuif, France.,b 2 Département d'innovations thérapeutiques précoces, Gustave Roussy, F-94805 Villejuif, France
| | - Alberto Duran-Peña
- c 3 AP-HP, Hôpital Universitaire la Pitié Salpêtrière, Service de Neurologie 2-Mazarin, F-75013, Paris, France
| | - Agusti Alentorn
- c 3 AP-HP, Hôpital Universitaire la Pitié Salpêtrière, Service de Neurologie 2-Mazarin, F-75013, Paris, France.,d 4 Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
| | - Ludovic Lacroix
- a 1 Inserm U981, Université Paris Sud, Gustave Roussy, F-94805 Villejuif, France.,e 5 Département de biologie médicale et de pathologie, Gustave Roussy, F-94805 Villejuif, France.,f 6 Laboratoire de recherche translationnelle et centre de ressources biologiques, Gustave Roussy, F-94805 Villejuif, France
| | - Christophe Massard
- a 1 Inserm U981, Université Paris Sud, Gustave Roussy, F-94805 Villejuif, France.,b 2 Département d'innovations thérapeutiques précoces, Gustave Roussy, F-94805 Villejuif, France
| | - Ahmed Idbaih
- c 3 AP-HP, Hôpital Universitaire la Pitié Salpêtrière, Service de Neurologie 2-Mazarin, F-75013, Paris, France.,d 4 Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
| |
Collapse
|
1142
|
Chabon JJ, Simmons AD, Lovejoy AF, Esfahani MS, Newman AM, Haringsma HJ, Kurtz DM, Stehr H, Scherer F, Karlovich CA, Harding TC, Durkin KA, Otterson GA, Purcell WT, Camidge DR, Goldman JW, Sequist LV, Piotrowska Z, Wakelee HA, Neal JW, Alizadeh AA, Diehn M. Circulating tumour DNA profiling reveals heterogeneity of EGFR inhibitor resistance mechanisms in lung cancer patients. Nat Commun 2016; 7:11815. [PMID: 27283993 PMCID: PMC4906406 DOI: 10.1038/ncomms11815] [Citation(s) in RCA: 488] [Impact Index Per Article: 54.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 05/03/2016] [Indexed: 12/20/2022] Open
Abstract
Circulating tumour DNA (ctDNA) analysis facilitates studies of tumour heterogeneity. Here we employ CAPP-Seq ctDNA analysis to study resistance mechanisms in 43 non-small cell lung cancer (NSCLC) patients treated with the third-generation epidermal growth factor receptor (EGFR) inhibitor rociletinib. We observe multiple resistance mechanisms in 46% of patients after treatment with first-line inhibitors, indicating frequent intra-patient heterogeneity. Rociletinib resistance recurrently involves MET, EGFR, PIK3CA, ERRB2, KRAS and RB1. We describe a novel EGFR L798I mutation and find that EGFR C797S, which arises in ∼33% of patients after osimertinib treatment, occurs in <3% after rociletinib. Increased MET copy number is the most frequent rociletinib resistance mechanism in this cohort and patients with multiple pre-existing mechanisms (T790M and MET) experience inferior responses. Similarly, rociletinib-resistant xenografts develop MET amplification that can be overcome with the MET inhibitor crizotinib. These results underscore the importance of tumour heterogeneity in NSCLC and the utility of ctDNA-based resistance mechanism assessment.
Collapse
Affiliation(s)
- Jacob J. Chabon
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, California 94305, USA
| | | | - Alexander F. Lovejoy
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, California 94305, USA
| | - Mohammad S. Esfahani
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, California 94305, USA
| | - Aaron M. Newman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, California 94305, USA
| | | | - David M. Kurtz
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California 94305, USA
- Department of Bioengineering, Stanford University, Stanford, California 94305, USA
| | - Henning Stehr
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, California 94305, USA
| | - Florian Scherer
- Stanford Cancer Institute, Stanford University, Stanford, California 94305, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California 94305, USA
| | | | | | - Kathleen A. Durkin
- Molecular Graphics and Computation Facility, College of Chemistry, University of California, Berkeley, California 94720, USA
| | | | - W. Thomas Purcell
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - D. Ross Camidge
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Jonathan W. Goldman
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Lecia V. Sequist
- Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Zofia Piotrowska
- Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Heather A. Wakelee
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California 94305, USA
| | - Joel W. Neal
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California 94305, USA
| | - Ash A. Alizadeh
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, California 94305, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California 94305, USA
- Division of Hematology, Department of Medicine, Stanford University, Stanford, California 94305, USA
| | - Maximilian Diehn
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, California 94305, USA
- Department of Radiation Oncology, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
1143
|
Yanagita M, Redig AJ, Paweletz CP, Dahlberg SE, O'Connell A, Feeney N, Taibi M, Boucher D, Oxnard GR, Johnson BE, Costa DB, Jackman DM, Jänne PA. A Prospective Evaluation of Circulating Tumor Cells and Cell-Free DNA in EGFR-Mutant Non-Small Cell Lung Cancer Patients Treated with Erlotinib on a Phase II Trial. Clin Cancer Res 2016; 22:6010-6020. [PMID: 27281561 DOI: 10.1158/1078-0432.ccr-16-0909] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 05/21/2016] [Accepted: 05/23/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Genotype-directed therapy is the standard of care for advanced non-small cell lung cancer (NSCLC), but obtaining tumor tissue for genotyping remains a challenge. Circulating tumor cell (CTC) or cell-free DNA (cfDNA) analysis may allow for noninvasive evaluation. This prospective trial evaluated CTCs and cfDNA in EGFR-mutant NSCLC patients treated with erlotinib until progression. EXPERIMENTAL DESIGN EGFR-mutant NSCLC patients were enrolled in a phase II trial of erlotinib. Blood was collected at baseline, every 2 months on study, and at disease progression. Plasma genotyping was performed by droplet digital PCR for EGFR19del, L858R, and T790M. CTCs were isolated by CellSave, enumerated, and analyzed by immunofluorescence for CD45 and pan-cytokeratin and EGFR and MET FISH were also performed. Rebiopsy was performed at disease progression. RESULTS Sixty patients were enrolled; 44 patients discontinued therapy for disease progression. Rebiopsy occurred in 35 of 44 patients (80%), with paired CTC/cfDNA analysis in 41 of 44 samples at baseline and 36 of 44 samples at progression. T790M was identified in 23 of 35 (66%) tissue biopsies and 9 of 39 (23%) cfDNA samples. CTC analysis at progression identified MET amplification in 3 samples in which tissue analysis could not be performed. cfDNA analysis identified T790M in 2 samples in which rebiopsy was not possible. At diagnosis, high levels of cfDNA but not high levels of CTCs correlated with progression-free survival. CONCLUSIONS cfDNA and CTCs are complementary, noninvasive assays for evaluation of acquired resistance to first-line EGFR TKIs and may expand the number of patients in whom actionable genetic information can be obtained at acquired resistance. Serial cfDNA monitoring may offer greater clinical utility than serial monitoring of CTCs. Clin Cancer Res; 22(24); 6010-20. ©2016 AACR.
Collapse
Affiliation(s)
- Masahiko Yanagita
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Amanda J Redig
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Cloud P Paweletz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Belfer Center for Applied Cancer Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Suzanne E Dahlberg
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Allison O'Connell
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Nora Feeney
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Myriam Taibi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - David Boucher
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Geoffrey R Oxnard
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Bruce E Johnson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Daniel B Costa
- Harvard Medical School, Boston, Massachusetts.,Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - David M Jackman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Pasi A Jänne
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Harvard Medical School, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Belfer Center for Applied Cancer Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
1144
|
Corti D, Kearns JD. Promises and pitfalls for recombinant oligoclonal antibodies-based therapeutics in cancer and infectious disease. Curr Opin Immunol 2016; 40:51-61. [PMID: 26995095 PMCID: PMC7127534 DOI: 10.1016/j.coi.2016.03.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Revised: 02/29/2016] [Accepted: 03/01/2016] [Indexed: 02/08/2023]
Abstract
Monoclonal antibodies (mAbs) have revolutionized the diagnosis and treatment of many human diseases and the application of combinations of mAbs has demonstrated improved therapeutic activity in both preclinical and clinical testing. Combinations of antibodies have several advantages such as the capacities to target multiple and mutating antigens in complex pathogens and to engage varied epitopes on multiple disease-related antigens (e.g. receptors) to overcome heterogeneity and plasticity. Oligoclonal antibodies are an emerging therapeutic format in which a novel antibody combination is developed as a single drug product. Here, we will provide historical context on the use of oligoclonal antibodies in oncology and infectious diseases and will highlight practical considerations related to their preclinical and clinical development programs.
Collapse
Affiliation(s)
| | - Jeffrey D Kearns
- Merrimack Pharmaceuticals, Inc., One Kendall Square, Suite B7201, Cambridge, MA 02139, USA.
| |
Collapse
|
1145
|
Zhou J, Huang A, Yang XR. Liquid Biopsy and its Potential for Management of Hepatocellular Carcinoma. J Gastrointest Cancer 2016; 47:157-167. [PMID: 26969471 DOI: 10.1007/s12029-016-9801-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE We summarized the recent findings of liquid biopsy in cancer field and discussed its potential utility in hepatocellular carcinoma. METHODS Literature published in MEDLINE, EMBASE, and Science Direct electronic databases was searched and reviewed. RESULTS Liquid biopsy specially referred to the detection of nucleic acids (circulating cell-free DNA, cfDNA) and circulating tumor cells (CTCs) in the blood of cancer patients. Compared to conventional single-site sampling or biopsy method, liquid biopsy had the advantages such as non-invasiveness, dynamic monitoring, and the most important of all, overcoming the limit of spatial and temporal heterogeneity. The genomic information of cancer could be profiled by genotyping cfDNA/CTC and subsequently applied to make molecular classification, targeted therapy guidance, and unveil drug resistance mechanisms. The serial sampling feature of liquid biopsy made it possible to monitor treatment response in a real-time manner and predict tumor metastasis/recurrence in advance. CONCLUSIONS Liquid biopsy is a non-invasive, dynamic, and informative sampling method with important clinical translational significance in cancer research and practice. Much work needs to be done before it is used in the management of HCC.
Collapse
Affiliation(s)
- Jian Zhou
- Liver Surgery Department, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 20032, China.
| | - Ao Huang
- Liver Surgery Department, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 20032, China
| | - Xin-Rong Yang
- Liver Surgery Department, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 20032, China
| |
Collapse
|
1146
|
Vargas DY, Kramer FR, Tyagi S, Marras SAE. Multiplex Real-Time PCR Assays that Measure the Abundance of Extremely Rare Mutations Associated with Cancer. PLoS One 2016; 11:e0156546. [PMID: 27244445 PMCID: PMC4887114 DOI: 10.1371/journal.pone.0156546] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/16/2016] [Indexed: 12/18/2022] Open
Abstract
We describe the use of “SuperSelective” primers that enable the detection and quantitation of somatic mutations whose presence relates to cancer diagnosis, prognosis, and therapy, in real-time PCR assays that can potentially analyze rare DNA fragments present in blood samples (liquid biopsies). The design of these deoxyribonucleotide primers incorporates both a relatively long “5' anchor sequence” that hybridizes strongly to target DNA fragments, and a very short, physically and functionally separate, “3' foot sequence” that is perfectly complementary to the mutant target sequence, but mismatches the wild-type sequence. As few as ten mutant fragments can reliably be detected in the presence of 1,000,000 wild-type fragments, even when the difference between the mutant and the wild type is only a single nucleotide polymorphism. Multiplex PCR assays employing a set of SuperSelective primers, and a corresponding set of differently colored molecular beacon probes, can be used in situations where the different mutations, though occurring in different cells, are located in the same codon. These non-symmetric real-time multiplex PCR assays contain limited concentrations of each SuperSelective primer, thereby enabling the simultaneous determination of each mutation’s abundance by comparing its threshold value to the threshold value of a reference gene present in the sample.
Collapse
Affiliation(s)
- Diana Y. Vargas
- Public Health Research Institute, Rutgers University, Newark, New Jersey, United States of America
| | - Fred Russell Kramer
- Public Health Research Institute, Rutgers University, Newark, New Jersey, United States of America
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
- * E-mail:
| | - Sanjay Tyagi
- Public Health Research Institute, Rutgers University, Newark, New Jersey, United States of America
- Department of Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Salvatore A. E. Marras
- Public Health Research Institute, Rutgers University, Newark, New Jersey, United States of America
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| |
Collapse
|
1147
|
He M, Zeng Y. Microfluidic Exosome Analysis toward Liquid Biopsy for Cancer. ACTA ACUST UNITED AC 2016; 21:599-608. [PMID: 27215792 DOI: 10.1177/2211068216651035] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Indexed: 12/20/2022]
Abstract
Assessment of a tumor's molecular makeup using biofluid samples, known as liquid biopsy, is a prominent research topic in precision medicine for cancer, due to its noninvasive property allowing repeat sampling for monitoring molecular changes of tumors over time. Circulating exosomes recently have been recognized as promising tumor surrogates because they deliver enriched biomarkers, such as proteins, RNAs, and DNA. However, purification and characterization of these exosomes are technically challenging. Microfluidic lab-on-a-chip technology effectively addresses these challenges owing to its inherent advantages in integration and automation of multiple functional modules, enhancing sensing performance, and expediting analysis processes. In this article, we review the state-of-the-art development of microfluidic technologies for exosome isolation and molecular characterization with emphasis on their applications toward liquid biopsy-based analysis of cancer. Finally, we share our perspectives on current challenges and future directions of microfluidic exosome analysis.
Collapse
Affiliation(s)
- Mei He
- Department of Biological and Agricultural Engineering, College of Engineering, Kansas State University, Manhattan, KS, USA
| | - Yong Zeng
- Department of Chemistry, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
1148
|
Endzeliņš E, Melne V, Kalniņa Z, Lietuvietis V, Riekstiņa U, Llorente A, Linē A. Diagnostic, prognostic and predictive value of cell-free miRNAs in prostate cancer: a systematic review. Mol Cancer 2016; 15:41. [PMID: 27189160 PMCID: PMC4870749 DOI: 10.1186/s12943-016-0523-5] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 05/12/2016] [Indexed: 01/01/2023] Open
Abstract
Prostate cancer, the second most frequently diagnosed cancer in males worldwide, is estimated to be diagnosed in 1.1 million men per year. Introduction of PSA testing substantially improved early detection of prostate cancer, however it also led to overdiagnosis and subsequent overtreatment of patients with an indolent disease. Treatment outcome and management of prostate cancer could be improved by the development of non-invasive biomarker assays that aid in increasing the sensitivity and specificity of prostate cancer screening, help to distinguish aggressive from indolent disease and guide therapeutic decisions. Prostate cancer cells release miRNAs into the bloodstream, where they exist incorporated into ribonucleoprotein complexes or extracellular vesicles. Later, cell-free miRNAs have been found in various other biofluids. The initial RNA sequencing studies suggested that most of the circulating cell-free miRNAs in healthy individuals are derived from blood cells, while specific disease-associated miRNA signatures may appear in the circulation of patients affected with various diseases, including cancer. This raised a hope that cell-free miRNAs may serve as non-invasive biomarkers for prostate cancer. Indeed, a number of cell-free miRNAs that potentially may serve as diagnostic, prognostic or predictive biomarkers have been discovered in blood or other biofluids of prostate cancer patients and need to be validated in appropriately designed longitudinal studies and clinical trials. In this review, we systematically summarise studies investigating cell-free miRNAs in biofluids of prostate cancer patients and discuss the utility of the identified biomarkers in various clinical scenarios. Furthermore, we discuss the possible mechanisms of miRNA release into biofluids and outline the biological questions and technical challenges that have arisen from these studies.
Collapse
Affiliation(s)
- Edgars Endzeliņš
- Latvian Biomedical Research and Study Centre, Ratsupites Str 1, k-1, LV-1067, Riga, Latvia
| | - Vita Melne
- Latvian Biomedical Research and Study Centre, Ratsupites Str 1, k-1, LV-1067, Riga, Latvia.,Riga Stradiņš University, Dzirciema Str 16, Riga, LV-1007, Latvia
| | - Zane Kalniņa
- Latvian Biomedical Research and Study Centre, Ratsupites Str 1, k-1, LV-1067, Riga, Latvia
| | - Vilnis Lietuvietis
- Latvian Biomedical Research and Study Centre, Ratsupites Str 1, k-1, LV-1067, Riga, Latvia.,Riga Stradiņš University, Dzirciema Str 16, Riga, LV-1007, Latvia
| | - Una Riekstiņa
- Faculty of Medicine, University of Latvia, 19 Raina blvd., Riga, LV-1586, Latvia
| | - Alicia Llorente
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, 0379, Oslo, Norway
| | - Aija Linē
- Latvian Biomedical Research and Study Centre, Ratsupites Str 1, k-1, LV-1067, Riga, Latvia.
| |
Collapse
|
1149
|
Schwaederle M, Husain H, Fanta PT, Piccioni DE, Kesari S, Schwab RB, Patel SP, Harismendy O, Ikeda M, Parker BA, Kurzrock R. Use of Liquid Biopsies in Clinical Oncology: Pilot Experience in 168 Patients. Clin Cancer Res 2016; 22:5497-5505. [DOI: 10.1158/1078-0432.ccr-16-0318] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 03/29/2016] [Accepted: 05/02/2016] [Indexed: 11/16/2022]
|
1150
|
Detecting Primary KIT Mutations in Presurgical Plasma of Patients with Gastrointestinal Stromal Tumor. Mol Diagn Ther 2016; 20:347-51. [DOI: 10.1007/s40291-016-0203-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|