1201
|
Malczewska A, Kidd M, Matar S, Kos-Kudla B, Modlin IM. A Comprehensive Assessment of the Role of miRNAs as Biomarkers in Gastroenteropancreatic Neuroendocrine Tumors. Neuroendocrinology 2018; 107:73-90. [PMID: 29566385 DOI: 10.1159/000487326] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 01/25/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND/AIMS A key issue in neuroendocrine neoplasia management is the identification of blood signatures that specifically define the activity of a cancer or local tumor microenvironment. MicroRNAs (miRNAs) may represent such a candidate. To evaluate their clinical utility as biomarkers in gastroenteropancreatic neuroendocrine tumors (GEP-NETs), we assessed their expression in tissue and blood. METHODS A systematic review of PubMed was undertaken to identify studies investigating miRNAs in GEP-NETs and their utility as blood or tissue biomarkers. RESULTS Twenty-two studies using a range of methodologies with different normalization protocols were identified: tumor - gastric NET type 1 (n = 1 study: MiR-222, regulates p27KIP1), pancreatic (n = 6: MiR-21 [inflammatory marker, oncogene] and MiR-144 [PI3K/AKT signaling], both up- and downregulated depending on the method), small intestinal (n = 7: no consistent signature), and colorectal (n = 3: no consistent signature); blood - gastric NET type 1 (n = 1: MiR-222), pancreatic (n = 3: MiR-21), and small intestinal (n = 3: no consistent signature). The studies all included heterogeneous cohorts, were insufficiently powered, and utilized different methodologies, and age- and gender-matched controls were not used. Different miRNA isolation methods and detection protocols resulted in inconsistent expression comparing tumor and blood. A scientific discrepancy was the downregulated expression of some circulating candidates compared to tissue levels, suggesting methodological issues or physiological responses to the tumor. Both are of concern in defining the biometrics of a marker. CONCLUSIONS A potential biomarker for GEP-NETs included MiR-21 (small bowel and pancreas), but this epithelial tumor marker requires prospective validation. Overall, significant scientific investigation remains to identify and demonstrate neuroendocrine specificity and to validate candidate miRNA biomarkers.
Collapse
Affiliation(s)
- Anna Malczewska
- Department of Endocrinology and Neuroendocrine Tumors, Medical University of Silesia, Katowice, Poland
| | - Mark Kidd
- Wren Laboratories, Branford, Connecticut, USA
| | - Somer Matar
- Wren Laboratories, Branford, Connecticut, USA
| | - Beata Kos-Kudla
- Department of Endocrinology and Neuroendocrine Tumors, Medical University of Silesia, Katowice, Poland
| | - Irvin M Modlin
- Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
1202
|
Calvo AR, Ibarra GH, Vibat CRT, Singh VM. Detecting an ALK Rearrangement via Liquid Biopsy Enabled a Targeted Therapy-based Approach for Treating a Patient with Advanced Non-small Cell Lung Cancer. ACTA ACUST UNITED AC 2018. [DOI: 10.17925/ohr.2018.14.1.38] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Initial diagnostic biopsy procedures often yield insufficient tissue for molecular testing, and invasive surgical biopsies can be associated with significant cost as well as risk to the patient. Liquid biopsy offers an alternative and economical means for molecular characterization of tumors via a simple peripheral blood draw. This case report describes the ability of liquid biopsy to detect an ALK translocation where tissue analysis by fluorescence in situ hybridization was negative for the genetic alteration. Identification of an ALK rearrangement in circulating tumor cells from a blood specimen led to sequential targeted therapies that included crizotinib followed by alectinib. The patient demonstrated outstanding clinical response during treatment with each of the prescribed ALK inhibitors. This case demonstrates the clinical utility of Biocept’s liquid biopsy to detect actionable biomarkers by surveying the systemic landscape of a patient’s disease where identification of the same genetic drivers may be missed in analyses of heterogeneous tumor tissue.
Collapse
Affiliation(s)
- Alejandro R Calvo
- Loma Linda University School of Medicine, KMC Campus and Kettering Cancer Center, Kettering, Ohio, US
| | | | | | | |
Collapse
|
1203
|
Castro-Giner F, Scheidmann MC, Aceto N. Beyond Enumeration: Functional and Computational Analysis of Circulating Tumor Cells to Investigate Cancer Metastasis. Front Med (Lausanne) 2018; 5:34. [PMID: 29520361 PMCID: PMC5827555 DOI: 10.3389/fmed.2018.00034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Circulating tumor cells (CTCs) are defined as those cells that detach from a cancerous lesion and enter the bloodstream. While generally most CTCs are subjected to high shear stress, anoikis signals, and immune attack in the circulatory system, few are able to survive and reach a distant organ in a viable state, possibly leading to metastasis formation. A large number of studies, both prospective and retrospective, have highlighted the association between CTC abundance and bad prognosis in patients with various cancer types. Yet, beyond CTC enumeration, much less is known about the distinction between metastatic and nonmetastatic CTCs, namely those features that enable only some CTCs to survive and seed a cancerous lesion at a distant site. In addition, critical aspects such as CTC heterogeneity, mechanisms that trigger CTC intravasation and extravasation, as well as vulnerabilities of metastatic CTCs subpopulations are poorly understood. In this short review, we highlight recent studies that successfully adopted functional and computational analysis to gain insights into CTC biology. We also discuss approaches to overcome challenges that are associated with CTC isolation, molecular and computational analysis, and speculate regarding few open questions that currently frame the CTC research field.
Collapse
Affiliation(s)
- Francesc Castro-Giner
- Department of Biomedicine, Cancer Metastasis Laboratory, University of Basel, University Hospital Basel, Basel, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Manuel C. Scheidmann
- Department of Biomedicine, Cancer Metastasis Laboratory, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Nicola Aceto
- Department of Biomedicine, Cancer Metastasis Laboratory, University of Basel, University Hospital Basel, Basel, Switzerland
- *Correspondence: Nicola Aceto,
| |
Collapse
|
1204
|
Wauters E, Vansteenkiste J. Will liquid biopsies become our fluid transition to personalized immunotherapy? Ann Oncol 2018; 29:11-13. [PMID: 29165546 DOI: 10.1093/annonc/mdx751] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- E Wauters
- Respiratory Oncology Unit (Pneumology), University Hospital KU Leuven, Leuven, Belgium.,Leuven Lung Cancer Group, University Hospital KU Leuven, Leuven, Belgium.,Laboratory of Pneumology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - J Vansteenkiste
- Respiratory Oncology Unit (Pneumology), University Hospital KU Leuven, Leuven, Belgium.,Leuven Lung Cancer Group, University Hospital KU Leuven, Leuven, Belgium.,Laboratory of Pneumology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| |
Collapse
|
1205
|
Golan T, Milella M, Ackerstein A, Berger R. The changing face of clinical trials in the personalized medicine and immuno-oncology era: report from the international congress on clinical trials in Oncology & Hemato-Oncology (ICTO 2017). JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:192. [PMID: 29282151 PMCID: PMC5745625 DOI: 10.1186/s13046-017-0668-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 12/18/2017] [Indexed: 01/10/2023]
Abstract
In the past decade, the oncology community has witnessed major advances in the understanding of cancer biology and major breakthroughs in several different therapeutic areas, from solid tumors to hematological malignancies; moreover, the advent of effective immunotherapy approaches, such as immune-checkpoint blockade, is revolutionizing treatment algorithms in almost all oncology disease areas. As knowledge evolves and new weapons emerge in the “war against cancer”, clinical and translational research need to adapt to a rapidly changing environment to effectively translate novel concepts into sustainable and accessible therapeutic options for cancer patients. With this in mind, translational cancer researchers, oncology professionals, treatment experts, CRO and industry leaders, as well as patient representatives gathered in London, 16-17 March 2017, for The International Congress on Clinical Trials in Oncology and Hemato-Oncology (ICTO2017), to discuss the changing face of oncology clinical trials in the new era of personalized medicine and immuno-oncology. A wide range of topics, including clinical trial design in immuno-oncology, biomarker-oriented drug development paths, statistical design and endpoint selection, challenges in the design and conduct of personalized medicine clinical trials, risk-based monitoring, financing and reimbursement, as well as best operational practices, were discussed in an open, highly interactive format, favoring networking among all relevant stakeholders. The most relevant data, approaches and issues emerged and discussed during the conference are summarized in this report.
Collapse
Affiliation(s)
- Talia Golan
- Oncology Institute, Sheba Medical Center, Emek HaEla St 1, Tel Hashomer, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michele Milella
- Division of Medical Oncology 1, Regina Elena National Cancer Institute, via Elio Chianesi 53, 00144, Rome, Italy.
| | - Aliza Ackerstein
- Oncology Institute, Sheba Medical Center, Emek HaEla St 1, Tel Hashomer, Ramat Gan, Israel
| | - Ranaan Berger
- Oncology Institute, Sheba Medical Center, Emek HaEla St 1, Tel Hashomer, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
1206
|
Volckmar AL, Sültmann H, Riediger A, Fioretos T, Schirmacher P, Endris V, Stenzinger A, Dietz S. A field guide for cancer diagnostics using cell-free DNA: From principles to practice and clinical applications. Genes Chromosomes Cancer 2017; 57:123-139. [DOI: 10.1002/gcc.22517] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 11/30/2017] [Accepted: 12/01/2017] [Indexed: 12/16/2022] Open
Affiliation(s)
- Anna-Lena Volckmar
- Institute of Pathology, University Hospital Heidelberg; Heidelberg Germany
| | - Holger Sültmann
- Division of Cancer Genome Research; German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK); Heidelberg Germany
| | - Anja Riediger
- Division of Cancer Genome Research; German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK); Heidelberg Germany
| | - Thoas Fioretos
- Department of Clinical Genetics; Lund University; Lund Sweden
- Department of Clinical Genetics; University and Regional Laboratories; Region Skåne Lund Sweden
| | - Peter Schirmacher
- Institute of Pathology, University Hospital Heidelberg; Heidelberg Germany
| | - Volker Endris
- Institute of Pathology, University Hospital Heidelberg; Heidelberg Germany
| | - Albrecht Stenzinger
- Institute of Pathology, University Hospital Heidelberg; Heidelberg Germany
- German Cancer Consortium (DKTK), Partner Site Heidelberg, and German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Steffen Dietz
- Division of Cancer Genome Research; German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK); Heidelberg Germany
| |
Collapse
|
1207
|
Wang Z, Wang X, Yuan J, Zhang X, Zhou J, Lu M, Liu D, Li J, Shen L. Survival Benefit of Palliative Local Treatments and Efficacy of Different Pharmacotherapies in Colorectal Cancer With Lung Metastasis: Results From a Large Retrospective Study. Clin Colorectal Cancer 2017; 17:e233-e255. [PMID: 29305209 DOI: 10.1016/j.clcc.2017.12.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 12/07/2017] [Indexed: 01/04/2023]
Abstract
BACKGROUND For most colorectal cancer patients with initial lung metastasis (LM), the only suitable treatments are palliative, including palliative local therapy and pharmacotherapy. We investigated the role of palliative local treatments in prolonging survival and the efficacy of different pharmacotherapies. PATIENTS AND METHODS After performing a medical record review of 2233 patients with metastatic colorectal cancer, 684 were identified as having LM. Their clinicopathologic characteristics, treatment patterns, and outcomes were analyzed retrospectively. RESULTS For nonresectable initial LM, patients receiving palliative local therapy had significantly longer median progression-free survival (PFS) and overall survival (OS) than those treated with pharmacotherapy alone: PFS 16.1 months versus 7.4 months (P < .001) and OS 51.8 months versus 23.8 months (P < .001), respectively. Cox multivariate analysis confirmed the survival benefit induced by palliative local therapy. Chemonaive patients receiving single-agent fluoropyrimidine had shorter PFS and longer OS compared to oxaliplatin- or irinotecan-based doublets when used as first-line treatment (PFS 4.8, 7.4, and 7.3 months; and OS 28.7, 21.2, and 20.1 months, respectively); however, these differences were not statistically significant. The addition of targeted agents to cytotoxic drugs prolonged PFS (10.5 vs. 7.2 months, P = .005) but not OS (27.8 vs. 21.2 months, P = .454). Carcinoembryonic antigen level, LM-associated symptoms, extrapulmonary disease, and histopathologic type were independent pretreatment prognostic factors. CONCLUSION Local treatments of LM may confer a survival benefit in the palliative setting. First-line single-agent fluoropyrimidine may be used in patients with good prognosis.
Collapse
Affiliation(s)
- Zhenghang Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xicheng Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jiajia Yuan
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jun Zhou
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Ming Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Dan Liu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China.
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|
1208
|
Tseng JS, Yang TY, Chen KC, Hsu KH, Huang YH, Su KY, Yu SL, Chang GC. Intercalated Treatment Following Rebiopsy Is Associated with a Shorter Progression-Free Survival of Osimertinib Treatment. Cancer Res Treat 2017; 50:1164-1174. [PMID: 29228521 PMCID: PMC6192904 DOI: 10.4143/crt.2017.460] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 12/08/2017] [Indexed: 12/18/2022] Open
Abstract
Purpose Epidermal growth factor receptor (EGFR) T790M mutation serves as an important predictor of osimertinib efficacy. However, little is known about how it works among patients with various timings of T790M emergence and treatment. Materials and Methods Advanced EGFR-mutant lung adenocarcinoma patients with positive T790M mutation in tumor were retrospectively enrolled and observed to determine the outcomes of osimertinib treatment. We evaluated the association between patients’ characteristics and the efficacy of osimertinib treatment, particularly with respect to the timing of T790M emergence and osimertinib prescription. Results A total of 91 patients were enrolled, including 14 (15.4%) with primary and 77 (84.6%) with acquired T790M mutation. The objective response rate and disease controlratewere 60.9% and 85.1%, respectively. The median progression-free survival (PFS) and overall survival were 11.5 months (95% confidence interval [CI], 9.0 to 14.0) and 30.4 months (95% CI, 11.3 to 49.5), respectively. There was no significant difference in response rate and PFS between primary and acquired T790M populations. In the acquired T790M subgroup, patientswho received osimertinib after T790M had been confirmed by rebiopsy had a longer PFS than those with intercalated treatments between rebiopsy and osimertinib prescription (14.0 months [95% CI, 9.0 to 18.9] vs. 7.2 months [95% CI, 3.7 to 10.8]; adjusted hazard ratio, 0.48 [95% CI, 0.24 to 0.98; p=0.043]). Rebiopsy timing did not influence the outcome. Conclusion Osimertinib prescription with intercalated treatment following rebiopsy but not the timing of T790M emergence influenced the treatment outcome. We suggest that it is better to start osimertinib treatment once T790M mutation has been confirmed by biopsy.
Collapse
Affiliation(s)
- Jeng-Sen Tseng
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Tsung-Ying Yang
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Kun-Chieh Chen
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Kuo-Hsuan Hsu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan.,Division of Critical Care and Respiratory Therapy, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yen-Hsiang Huang
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Kang-Yi Su
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Sung-Liang Yu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Center of Genomic Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Pathology and Graduate Institute of Pathology, National Taiwan University College of Medicine, Taipei, Taiwan.,Center for Optoelectronic Biomedicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Gee-Chen Chang
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Comprehensive Cancer Center, Taichung Veterans General Hospital, Taichung, Taiwan
| |
Collapse
|
1209
|
Affiliation(s)
- Erik Sahai
- Tumour Cell Biology Laboratory, Francis Crick Institute
| | - Charles Swanton
- Translational Cancer Therapeutics Laboratory, Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| |
Collapse
|
1210
|
Mason J, Blyth B, MacManus MP, Martin OA. Treatment for non-small-cell lung cancer and circulating tumor cells. Lung Cancer Manag 2017; 6:129-139. [PMID: 30643579 PMCID: PMC6310303 DOI: 10.2217/lmt-2017-0019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/02/2018] [Indexed: 12/25/2022] Open
Abstract
Surgery is the main curative therapy for patients with localized non-small-cell lung cancer while radiotherapy (RT), alone or with concurrent platinum-based chemotherapy, remains the primary curative modality for locoregionally advanced non-small-cell lung cancer. The risk of distant metastasis is high after curative-intent treatment, largely attributable to the presence of undetected micrometastases, but which could also be related to treatment-related increases in circulating tumor cells (CTCs). CTC mobilization by RT or systemic therapies might either reflect efficient tumor destruction with improved prognosis, or might promote metastasis and thus represent a potential therapeutic target. RT may induce prometastatic biological alterations in CTC at the cellular level, which are detectable by 'liquid biopsies', though their rarity represents a major challenge. Improved methods of isolation and ex vivo propagation will be essential for the future of CTC research.
Collapse
Affiliation(s)
- Joel Mason
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
- Research Division, Peter MacCallum Cancer Center, Melbourne, Australia
- Department of Pathology, The University of Melbourne, Melbourne, Australia
| | - Benjamin Blyth
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
- Research Division, Peter MacCallum Cancer Center, Melbourne, Australia
| | - Michael P MacManus
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
- Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Olga A Martin
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
- Research Division, Peter MacCallum Cancer Center, Melbourne, Australia
- Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
1211
|
Liu F, Vermesh O, Mani V, Ge TJ, Madsen SJ, Sabour A, Hsu EC, Gowrishankar G, Kanada M, Jokerst JV, Sierra RG, Chang E, Lau K, Sridhar K, Bermudez A, Pitteri SJ, Stoyanova T, Sinclair R, Nair VS, Gambhir SS, Demirci U. The Exosome Total Isolation Chip. ACS NANO 2017; 11:10712-10723. [PMID: 29090896 PMCID: PMC5983373 DOI: 10.1021/acsnano.7b04878] [Citation(s) in RCA: 250] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Circulating tumor-derived extracellular vesicles (EVs) have emerged as a promising source for identifying cancer biomarkers for early cancer detection. However, the clinical utility of EVs has thus far been limited by the fact that most EV isolation methods are tedious, nonstandardized, and require bulky instrumentation such as ultracentrifugation (UC). Here, we report a size-based EV isolation tool called ExoTIC (exosome total isolation chip), which is simple, easy-to-use, modular, and facilitates high-yield and high-purity EV isolation from biofluids. ExoTIC achieves an EV yield ∼4-1000-fold higher than that with UC, and EV-derived protein and microRNA levels are well-correlated between the two methods. Moreover, we demonstrate that ExoTIC is a modular platform that can sort a heterogeneous population of cancer cell line EVs based on size. Further, we utilize ExoTIC to isolate EVs from cancer patient clinical samples, including plasma, urine, and lavage, demonstrating the device's broad applicability to cancers and other diseases. Finally, the ability of ExoTIC to efficiently isolate EVs from small sample volumes opens up avenues for preclinical studies in small animal tumor models and for point-of-care EV-based clinical testing from fingerprick quantities (10-100 μL) of blood.
Collapse
Affiliation(s)
- Fei Liu
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- School of Ophthalmology & Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
- Wenzhou Institute of Biomaterials and Engineering, Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| | - Ophir Vermesh
- Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Molecular Imaging Program at Stanford, Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
| | - Vigneshwaran Mani
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
| | - Tianjia J. Ge
- Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Molecular Imaging Program at Stanford, Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
| | - Steven J. Madsen
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94304, United States
| | - Andrew Sabour
- Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Molecular Imaging Program at Stanford, Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
| | - En-Chi Hsu
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
| | - Gayatri Gowrishankar
- Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Molecular Imaging Program at Stanford, Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
| | - Masamitsu Kanada
- Molecular Imaging Program at Stanford, Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Department of Pediatrics, School of Medicine, Stanford University, Stanford, California 94304, United States
- Department of Pharmacology & Toxicology, and Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, Michigan 48824, United States
| | - Jesse V. Jokerst
- Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Molecular Imaging Program at Stanford, Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, United States
| | - Raymond G. Sierra
- Stanford PULSE Institute, SLAC National Accelerator Lab, Menlo Park, California 94025, United States
- Hard X-ray Department, LCLS, SLAC National Accelerator Lab, Menlo Park, California 94025, United States
| | - Edwin Chang
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Molecular Imaging Program at Stanford, Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
| | - Kenneth Lau
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
| | - Kaushik Sridhar
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
| | - Abel Bermudez
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
| | - Sharon J. Pitteri
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
| | - Tanya Stoyanova
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
| | - Robert Sinclair
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94304, United States
| | - Viswam S. Nair
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Department of Medicine, Stanford University, Stanford, California 94304, United States
| | - Sanjiv S. Gambhir
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Molecular Imaging Program at Stanford, Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94304, United States
- Department of Bioengineering, Stanford University, Stanford, California 94304, United States
- Corresponding Authors: ,
| | - Utkan Demirci
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Department of Radiology, School of Medicine, Stanford University, Stanford, California 94304, United States
- Corresponding Authors: ,
| |
Collapse
|
1212
|
Chen L, Fu W, Zheng L, Liu Z, Liang G. Recent Progress of Small-Molecule Epidermal Growth Factor Receptor (EGFR) Inhibitors against C797S Resistance in Non-Small-Cell Lung Cancer. J Med Chem 2017; 61:4290-4300. [PMID: 29136465 DOI: 10.1021/acs.jmedchem.7b01310] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The epidermal growth factor receptor (EGFR) has been a particular interest for drug development for treatment of non-small-cell lung cancer (NSCLC). The current third-generation EGFR small-molecule inhibitors, especially osimertinib, are at the forefront clinically for treatment of patients with NSCLC. However, a high percentage of these treated patients developed a tertiary cystein-797 to serine-790 (C797S) mutation in the EGFR kinase domain. This C797S mutation is thought to induce resistance to all current irreversible EGFR TKIs. In this Miniperspective, we present key mechanisms of resistance in response to third-generation EGFR TKIs, and emerging reports on novel EGFR TKIs to combat the resistance. Specifically, we analyze the allosteric and ATP-competitive inhibitors in terms of drug discovery, binding mechanism, and their potency and selectivity against EGFR harboring C797S mutations. Lastly, we provide some perspectives on new challenges and future directions in this field.
Collapse
Affiliation(s)
- Lingfeng Chen
- Chemical Biology Research Center at School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China.,School of Chemical Engineering , Nanjing University of Science and Technology , Nanjing , Jiangsu 210094 , China
| | - Weitao Fu
- Chemical Biology Research Center at School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China
| | - Lulu Zheng
- Chemical Biology Research Center at School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China
| | - Zhiguo Liu
- Chemical Biology Research Center at School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China
| | - Guang Liang
- Chemical Biology Research Center at School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China.,School of Chemical Engineering , Nanjing University of Science and Technology , Nanjing , Jiangsu 210094 , China
| |
Collapse
|
1213
|
Macías M, Alegre E, Díaz-Lagares A, Patiño A, Pérez-Gracia JL, Sanmamed M, López-López R, Varo N, González A. Liquid Biopsy: From Basic Research to Clinical Practice. Adv Clin Chem 2017; 83:73-119. [PMID: 29304904 DOI: 10.1016/bs.acc.2017.10.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Liquid biopsy refers to the molecular analysis in biological fluids of nucleic acids, subcellular structures, especially exosomes, and, in the context of cancer, circulating tumor cells. In the last 10 years, there has been an intensive research in liquid biopsy to achieve a less invasive and more precise personalized medicine. Molecular assessment of these circulating biomarkers can complement or even surrogate tissue biopsy. Because of this research, liquid biopsy has been introduced in clinical practice, especially in oncology, prenatal screening, and transplantation. Here we review the biology, methodological approaches, and clinical applications of the main biomarkers involved in liquid biopsy.
Collapse
Affiliation(s)
| | - Estibaliz Alegre
- Clínica Universidad de Navarra, Pamplona, Spain; The Health Research Institute of Navarra (IDISNA), Pamplona, Spain
| | - Angel Díaz-Lagares
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), University Clinical Hospital of Santiago (CHUS), CIBERONC, Santiago de Compostela, Spain; Roche-CHUS Joint Unit, University Clinical Hospital of Santiago (CHUS), Santiago de Compostela, Spain
| | - Ana Patiño
- Clínica Universidad de Navarra, Pamplona, Spain; The Health Research Institute of Navarra (IDISNA), Pamplona, Spain
| | - Jose L Pérez-Gracia
- Clínica Universidad de Navarra, Pamplona, Spain; The Health Research Institute of Navarra (IDISNA), Pamplona, Spain
| | - Miguel Sanmamed
- Yale University School of Medicine, New Haven, CT, United States
| | - Rafael López-López
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), University Clinical Hospital of Santiago (CHUS), CIBERONC, Santiago de Compostela, Spain; Roche-CHUS Joint Unit, University Clinical Hospital of Santiago (CHUS), Santiago de Compostela, Spain
| | - Nerea Varo
- Clínica Universidad de Navarra, Pamplona, Spain; The Health Research Institute of Navarra (IDISNA), Pamplona, Spain
| | - Alvaro González
- Clínica Universidad de Navarra, Pamplona, Spain; The Health Research Institute of Navarra (IDISNA), Pamplona, Spain.
| |
Collapse
|
1214
|
Pando A, Reagan JL, Quesenberry P, Fast LD. Extracellular vesicles in leukemia. Leuk Res 2017; 64:52-60. [PMID: 29190514 DOI: 10.1016/j.leukres.2017.11.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 11/12/2017] [Accepted: 11/21/2017] [Indexed: 12/21/2022]
Abstract
Extracellular vesicles (EV) are nano-sized membrane enclosed vehicles that are involved in cell-to-cell communication and carry cargo that is representative of the parent cell. Recent studies have highlighted the significant roles leukemia EVs play in tumor progression, and ways in which they can lead to treatment evasion, thus meriting further investigation. Leukemia EVs are involved in crosstalk between the leukemia cell and its surroundings, transforming it into a cancer favorable microenvironment. Due to the diverse biological content found in leukemia EVs, they have an assortment of effects on the cells they interact with and can be harnessed as candidates for diagnostic and therapeutic treatments. This review focuses on EVs in the context of leukemia and the means by which they modulate their microenvironment, hematopoiesis, and the immune system to facilitate malignancy. We will also address current and prospective EV-based therapeutics.
Collapse
Affiliation(s)
- Alejandro Pando
- Division of Hematology and Oncology, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - John L Reagan
- Division of Hematology and Oncology, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Peter Quesenberry
- Division of Hematology and Oncology, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Loren D Fast
- Division of Hematology and Oncology, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI, USA.
| |
Collapse
|
1215
|
Filosso PL, Kidd M, Roffinella M, Lewczuk A, Chung KM, Kolasinska-Cwikla A, Cwikla J, Lowczak A, Doboszynska A, Malczewska A, Catalano M, Zunino V, Boita M, Arvat E, Cristofori R, Guerrera F, Oliaro A, Tesselaar M, Buikhuisen W, Kos-Kudla B, Papotti M, Bodei L, Drozdov I, Modlin I. The utility of blood neuroendocrine gene transcript measurement in the diagnosis of bronchopulmonary neuroendocrine tumours and as a tool to evaluate surgical resection and disease progression†. Eur J Cardiothorac Surg 2017; 53:631-639. [DOI: 10.1093/ejcts/ezx386] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 09/30/2017] [Indexed: 01/04/2023] Open
Affiliation(s)
- Pier Luigi Filosso
- Department of Thoracic Surgery and Department of Oncology, University of Torino, Torino, Italy
| | - Mark Kidd
- Wren Laboratories, Branford, CT, USA
| | - Matteo Roffinella
- Department of Thoracic Surgery and Department of Oncology, University of Torino, Torino, Italy
| | - Anna Lewczuk
- Department of Endocrinology, Medical University of Gdansk, Gdansk, Poland
| | | | | | - Jaroslaw Cwikla
- Department of Radiology, University of Warmia and Mazury, Olsztyn, Poland
| | - Anna Lowczak
- Department of Radiology, University of Warmia and Mazury, Olsztyn, Poland
| | - Anna Doboszynska
- Department of Radiology, University of Warmia and Mazury, Olsztyn, Poland
| | - Anna Malczewska
- Department of Endocrinology, Medical University of Silesia, Katowice, Poland
| | - Maria Catalano
- Department of Thoracic Surgery and Department of Oncology, University of Torino, Torino, Italy
| | - Valentina Zunino
- Department of Thoracic Surgery and Department of Oncology, University of Torino, Torino, Italy
| | - Monica Boita
- Department of Thoracic Surgery and Department of Oncology, University of Torino, Torino, Italy
| | - Emanuela Arvat
- Department of Thoracic Surgery and Department of Oncology, University of Torino, Torino, Italy
| | - Riccardo Cristofori
- Department of Thoracic Surgery and Department of Oncology, University of Torino, Torino, Italy
| | - Francesco Guerrera
- Department of Thoracic Surgery and Department of Oncology, University of Torino, Torino, Italy
| | - Alberto Oliaro
- Department of Thoracic Surgery and Department of Oncology, University of Torino, Torino, Italy
| | - Margot Tesselaar
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Wieneke Buikhuisen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Beata Kos-Kudla
- Department of Endocrinology, Medical University of Silesia, Katowice, Poland
| | - Mauro Papotti
- Department of Thoracic Surgery and Department of Oncology, University of Torino, Torino, Italy
| | - Lisa Bodei
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Irvin Modlin
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
1216
|
Postel M, Roosen A, Laurent-Puig P, Taly V, Wang-Renault SF. Droplet-based digital PCR and next generation sequencing for monitoring circulating tumor DNA: a cancer diagnostic perspective. Expert Rev Mol Diagn 2017; 18:7-17. [PMID: 29115895 DOI: 10.1080/14737159.2018.1400384] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Early detection of cancers through the analysis of ctDNA could have a significant impact on morbidity and mortality of cancer patients. However, using ctDNA for early cancer diagnosis is challenging partly due to the low amount of tumor DNA released in the circulation and its dilution within DNA originating from non-tumor cells. Development of new technologies such as droplet-based digital PCR (ddPCR) or optimized next generation sequencing (NGS) has greatly improved the sensitivity, specificity and precision for the detection of rare sequences. Areas covered: This paper will focus on the potential application of ddPCR and optimized NGS to detect ctDNA for detection of cancer recurrence and minimal residual disease as well as early diagnosis of cancer patients. Expert commentary: Compared to tumor tissue biopsies, blood-based ctDNA analyses are minimally invasive and accessible for regular follow-up of cancer patients. They are also described as a better picture of patients' pathology allowing to highlight both tumor heterogeneity and multiple tumor sites. After a brief introduction on the application of the follow-up of ctDNA using genetic or epigenetic biomarkers for prognosis and surveillance of cancer patients, potential perspectives of using ctDNA for early diagnosis of cancers will be presented.
Collapse
Affiliation(s)
- Mathilde Postel
- a INSERM UMR-S1147, CNRS SNC5014; Paris Descartes University, Equipe labellisée Ligue Nationale contre le cancer , Paris , France
| | - Alice Roosen
- a INSERM UMR-S1147, CNRS SNC5014; Paris Descartes University, Equipe labellisée Ligue Nationale contre le cancer , Paris , France
| | - Pierre Laurent-Puig
- a INSERM UMR-S1147, CNRS SNC5014; Paris Descartes University, Equipe labellisée Ligue Nationale contre le cancer , Paris , France.,b Department of Biology , European Georges Pompidou Hospital, AP-HP , Paris , France
| | - Valerie Taly
- a INSERM UMR-S1147, CNRS SNC5014; Paris Descartes University, Equipe labellisée Ligue Nationale contre le cancer , Paris , France
| | - Shu-Fang Wang-Renault
- a INSERM UMR-S1147, CNRS SNC5014; Paris Descartes University, Equipe labellisée Ligue Nationale contre le cancer , Paris , France
| |
Collapse
|
1217
|
D'Agata R, Giuffrida MC, Spoto G. Peptide Nucleic Acid-Based Biosensors for Cancer Diagnosis. Molecules 2017; 22:E1951. [PMID: 29137122 PMCID: PMC6150339 DOI: 10.3390/molecules22111951] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 11/06/2017] [Accepted: 11/09/2017] [Indexed: 12/18/2022] Open
Abstract
The monitoring of DNA and RNA biomarkers freely circulating in the blood constitutes the basis of innovative cancer detection methods based on liquid biopsy. Such methods are expected to provide new opportunities for a better understanding of cancer disease at the molecular level, thus contributing to improved patient outcomes. Advanced biosensors can advance possibilities for cancer-related nucleic acid biomarkers detection. In this context, peptide nucleic acids (PNAs) play an important role in the fabrication of highly sensitive biosensors. This review provides an overview of recently described PNA-based biosensors for cancer biomarker detection. One of the most striking features of the described detection approaches is represented by the possibility to detect target nucleic acids at the ultra-low concentration with the capability to identify single-base mutations.
Collapse
Affiliation(s)
- Roberta D'Agata
- Dipartimento di Scienze Chimiche, Università di Catania, Viale Andrea Doria 6, I-95125 Catania, Italy.
| | - Maria Chiara Giuffrida
- Consorzio Interuniversitario "Istituto Nazionale di Biostrutture e Biosistemi", c/o Dipartimento di Scienze Chimiche, Università di Catania, Viale Andrea Doria 6, I-95125 Catania, Italy.
| | - Giuseppe Spoto
- Dipartimento di Scienze Chimiche, Università di Catania, Viale Andrea Doria 6, I-95125 Catania, Italy.
- Consorzio Interuniversitario "Istituto Nazionale di Biostrutture e Biosistemi", c/o Dipartimento di Scienze Chimiche, Università di Catania, Viale Andrea Doria 6, I-95125 Catania, Italy.
| |
Collapse
|
1218
|
Liquid Biopsy and Therapeutic Targets: Present and Future Issues in Thoracic Oncology. Cancers (Basel) 2017; 9:cancers9110154. [PMID: 29125548 PMCID: PMC5704172 DOI: 10.3390/cancers9110154] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 11/06/2017] [Accepted: 11/08/2017] [Indexed: 02/06/2023] Open
Abstract
The practice of liquid biopsy (LB) has revolutionized the care of patients with metastatic lung cancer. Many oncologists now use this approach in daily practice, applying precise procedures for the detection of activating or resistance mutations in EGFR. These tests are performed with plasma DNA and have been approved as companion diagnostic test for patients treated with tyrosine kinase inhibitors. ALK is another important target in lung cancer since it leads to treatment of patients who are positive for a rearrangement in ALK identified with tumor tissue. By analogy with EGFR, LB for detection of genomic alterations in ALK (rearrangements or mutations) has been rapidly adopted in the clinic. However, this promising approach has some limitations and has not yet been disseminated as much as the blood test targeting EGFR. In addition to these two therapeutic targets LB can be used for evaluation of the genomic status of other genes of interest of patients with lung cancer (ROS1, RET, NTRK MET, BRAF, HER2, etc.). LB can be performed to evaluate a specific target or for a more or less complex panel of genes. Considering the number of potential targets for clinical trials, techniques of next-generation sequencing of circulating DNA are on the rise. This review will provide an update on the contribution of LB to care of patients with metastatic lung cancer, including the present limits of this approach, and will consider certain perspectives.
Collapse
|
1219
|
Sefrioui D, Beaussire L, Perdrix A, Clatot F, Michel P, Frebourg T, Di Fiore F, Sarafan-Vasseur N. Direct circulating tumor DNA detection from unpurified plasma using a digital PCR platform. Clin Biochem 2017. [DOI: 10.1016/j.clinbiochem.2017.06.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
1220
|
Chung JH, Pavlick D, Hartmaier R, Schrock AB, Young L, Forcier B, Ye P, Levin MK, Goldberg M, Burris H, Gay LM, Hoffman AD, Stephens PJ, Frampton GM, Lipson DM, Nguyen DM, Ganesan S, Park BH, Vahdat LT, Leyland-Jones B, Mughal TI, Pusztai L, O'Shaughnessy J, Miller VA, Ross JS, Ali SM. Hybrid capture-based genomic profiling of circulating tumor DNA from patients with estrogen receptor-positive metastatic breast cancer. Ann Oncol 2017; 28:2866-2873. [PMID: 28945887 PMCID: PMC5834148 DOI: 10.1093/annonc/mdx490] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Genomic changes that occur in breast cancer during the course of disease have been informed by sequencing of primary and metastatic tumor tissue. For patients with relapsed and metastatic disease, evolution of the breast cancer genome highlights the importance of using a recent sample for genomic profiling to guide clinical decision-making. Obtaining a metastatic tissue biopsy can be challenging, and analysis of circulating tumor DNA (ctDNA) from blood may provide a minimally invasive alternative. PATIENTS AND METHODS Hybrid capture-based genomic profiling was carried out on ctDNA from 254 female patients with estrogen receptor-positive breast cancer. Peripheral blood samples were submitted by clinicians in the course of routine clinical care between May 2016 and March 2017. Sequencing of 62 genes was carried out to a median unique coverage depth of 7503×. Genomic alterations (GAs) in ctDNA were evaluated and compared with matched tissue samples and genomic datasets of tissue from breast cancer. RESULTS At least 1 GA was reported in 78% of samples. Frequently altered genes were TP53 (38%), ESR1 (31%) and PIK3CA (31%). Temporally matched ctDNA and tissue samples were available for 14 patients; 89% of mutations detected in tissue were also detected in ctDNA. Diverse ESR1 GAs including mutation, rearrangement and amplification, were observed. Multiple concurrent ESR1 GAs were observed in 40% of ESR1-altered cases, suggesting polyclonal origin; ESR1 compound mutations were also observed in two cases. ESR1-altered cases harbored co-occurring GAs in PIK3CA (35%), FGFR1 (16%), ERBB2 (8%), BRCA1/2 (5%), and AKT1 (4%). CONCLUSIONS GAs relevant to relapsed/metastatic breast cancer management were identified, including diverse ESR1 GAs. Genomic profiling of ctDNA demonstrated sensitive detection of mutations found in tissue. Detection of amplifications was associated with ctDNA fraction. Genomic profiling of ctDNA may provide a complementary and possibly alternative approach to tissue-based genomic testing for patients with estrogen receptor-positive metastatic breast cancer.
Collapse
Affiliation(s)
- J H Chung
- Foundation Medicine, Inc., Cambridge.
| | - D Pavlick
- Foundation Medicine, Inc., Cambridge
| | | | | | - L Young
- Foundation Medicine, Inc., Cambridge
| | - B Forcier
- Foundation Medicine, Inc., Cambridge
| | - P Ye
- Avera Cancer Institute, Sioux Falls
| | - M K Levin
- Baylor University Medical Center, Texas Oncology, US Oncology, Dallas
| | | | - H Burris
- Sarah Cannon Research Institute, Nashville
| | - L M Gay
- Foundation Medicine, Inc., Cambridge
| | | | | | | | | | - D M Nguyen
- Sutter Medical Group of the Redwoods, Santa Rosa
| | - S Ganesan
- Division of Medical Oncology, Department of Medicine, Rutgers Cancer Institute of New Jersey, New Brunswick
| | - B H Park
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore
| | - L T Vahdat
- Weill Cornell Breast Center, Weill Cornell Medicine, New York
| | | | - T I Mughal
- Foundation Medicine, Inc., Cambridge; Tufts University Medical Center, Boston
| | - L Pusztai
- Department of Breast Medical Oncology, Yale University, Yale Cancer Center, New Haven
| | - J O'Shaughnessy
- Baylor University Medical Center, Texas Oncology, US Oncology, Dallas
| | | | - J S Ross
- Foundation Medicine, Inc., Cambridge; Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, USA. mailto:
| | - S M Ali
- Foundation Medicine, Inc., Cambridge
| |
Collapse
|
1221
|
Renaux-Petel M, Charbonnier F, Théry JC, Fermey P, Lienard G, Bou J, Coutant S, Vezain M, Kasper E, Fourneaux S, Manase S, Blanluet M, Leheup B, Mansuy L, Champigneulle J, Chappé C, Longy M, Sévenet N, Paillerets BBD, Guerrini-Rousseau L, Brugières L, Caron O, Sabourin JC, Tournier I, Baert-Desurmont S, Frébourg T, Bougeard G. Contribution of de novo and mosaic TP53 mutations to Li-Fraumeni syndrome. J Med Genet 2017; 55:173-180. [DOI: 10.1136/jmedgenet-2017-104976] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 09/28/2017] [Accepted: 10/09/2017] [Indexed: 01/06/2023]
Abstract
BackgroundDevelopment of tumours such as adrenocortical carcinomas (ACC), choroid plexus tumours (CPT) or female breast cancers before age 31 or multiple primary cancers belonging to the Li-Fraumeni (LFS) spectrum is, independently of the familial history, highly suggestive of a germline TP53 mutation. The aim of this study was to determine the contribution of de novo and mosaic mutations to LFS.Methods and resultsAmong 328 unrelated patients harbouring a germline TP53 mutation identified by Sanger sequencing and/or QMPSF, we could show that the mutations had occurred de novo in 40 cases, without detectable parental age effect. Sanger sequencing revealed two mosaic mutations in a child with ACC and in an unaffected father of a child with medulloblastoma. Re-analysis of blood DNA by next-generation sequencing, performed at a depth above 500X, from 108 patients suggestive of LFS without detectable TP53 mutations, allowed us to identify 6 additional cases of mosaic TP53 mutations, in 2/49 children with ACC, 2/21 children with CPT, in 1/31 women with breast cancer before age 31 and in a patient who developed an osteosarcoma at age 12, a breast carcinoma and a breast sarcoma at age 35.ConclusionsThis study performed on a large series of TP53 mutation carriers allows estimating the contribution to LFS of de novo mutations to at least 14% (48/336) and suggests that approximately one-fifth of these de novo mutations occur during embryonic development. Considering the medical impact of TP53 mutation identification, medical laboratories in charge of TP53 testing should ensure the detection of mosaic mutations.
Collapse
|
1222
|
|
1223
|
Jiang T, Li X, Wang J, Su C, Han W, Zhao C, Wu F, Gao G, Li W, Chen X, Li J, Zhou F, Zhao J, Cai W, Zhang H, Du B, Zhang J, Ren S, Zhou C, Yu H, Hirsch FR. Mutational Landscape of cfDNA Identifies Distinct Molecular Features Associated With Therapeutic Response to First-Line Platinum-Based Doublet Chemotherapy in Patients with Advanced NSCLC. Theranostics 2017; 7:4753-4762. [PMID: 29187901 PMCID: PMC5706097 DOI: 10.7150/thno.21687] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/13/2017] [Indexed: 01/15/2023] Open
Abstract
Rationale To investigate whether the mutational landscape of circulating cell-free DNA (cfDNA) could predict and dynamically monitor the response to first-line platinum-based chemotherapy in patients with advanced non-small-cell lung cancer (NSCLC). Methods Eligible patients were included and blood samples were collected from a phase III trial. Both cfDNA fragments and fragmented genomic DNA were extracted for enrichment in a 1.15M size panel covering exon regions of 1,086 genes. Molecular mutational burden (MMB) was calculated to investigate the relationship between molecular features of cfDNA and response to chemotherapy. Results In total, 52 eligible cases were enrolled and their blood samples were prospectively collected at baseline, every cycle of chemotherapy and time of disease progression. At baseline, alterations of 17 genes were found. Patients with partial response (PR) had significantly lower baseline MMB of these genes than those patients with either stable disease (SD) (P = 0.0006) or progression disease (PD) (P = 0.0074). Further analysis revealed that the mutational landscape of cfDNA from pretreatment blood samples were distinctly different among patients with PR vs. SD/PD. For patients with baseline TP53 mutation, those with PR experienced a significant reduction in MMB whereas patients with SD or PD experienced an increase after two, three or four cycles of chemotherapy. Furthermore, patients with low MMB had superior response rate and significantly longer progression-free survival than those with high MMB. Conclusion This study indicated that the mutational landscape of cfDNA has potential clinical value to predict the therapeutic response to first-line platinum-based doublet chemotherapy in NSCLC patients. At the single gene level, dynamic change of molecular mutational burden of TP53 is valuable to monitor efficacy (and, therefore, might aid in early recognition of resistance and relapse) in patients harboring this mutation at baseline.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, P.R. China
| | - Xuefei Li
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Jianfei Wang
- Beijing Genecast Biotechnology Co., Beijing, P.R. China
| | - Chunxia Su
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, P.R. China
| | - Wenbo Han
- Beijing Genecast Biotechnology Co., Beijing, P.R. China
| | - Chao Zhao
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Fengying Wu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, P.R. China
| | - Guanghui Gao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, P.R. China
| | - Wei Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, P.R. China
| | - Xiaoxia Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, P.R. China
| | - Jiayu Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, P.R. China
| | - Fei Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, P.R. China
| | - Jing Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, P.R. China
| | - Weijing Cai
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, P.R. China
| | - Henghui Zhang
- Beijing Genecast Biotechnology Co., Beijing, P.R. China
| | - Bo Du
- Beijing Genecast Biotechnology Co., Beijing, P.R. China
| | - Jun Zhang
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Shengxiang Ren
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, P.R. China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, P.R. China
| | - Hui Yu
- Department of Medicine, Division of Medical Oncology, University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO, USA
| | - Fred R. Hirsch
- Department of Medicine, Division of Medical Oncology, University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
1224
|
The potential of liquid biopsies for the early detection of cancer. NPJ Precis Oncol 2017; 1:36. [PMID: 29872715 PMCID: PMC5871864 DOI: 10.1038/s41698-017-0039-5] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/25/2017] [Accepted: 09/25/2017] [Indexed: 02/07/2023] Open
Abstract
Precision medicine refers to the choosing of targeted therapies based on genetic data. Due to the increasing availability of data from large-scale tumor genome sequencing projects, genome-driven oncology may have enormous potential to change the clinical management of patients with cancer. To this end, components of tumors, which are shed into the circulation, i.e., circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), or extracellular vesicles, are increasingly being used for monitoring tumor genomes. A growing number of publications have documented that these “liquid biopsies” are informative regarding response to given therapies, are capable of detecting relapse with lead time compared to standard measures, and reveal mechanisms of resistance. However, the majority of published studies relate to advanced tumor stages and the use of liquid biopsies for detection of very early malignant disease stages is less well documented. In early disease stages, strategies for analysis are in principle relatively similar to advanced stages. However, at these early stages, several factors pose particular difficulties and challenges, including the lower frequency and volume of aberrations, potentially confounding phenomena such as clonal expansions of non-tumorous tissues or the accumulation of cancer-associated mutations with age, and the incomplete insight into driver alterations. Here we discuss biology, technical complexities and clinical significance for early cancer detection and their impact on precision oncology.
Collapse
|
1225
|
Mithraprabhu S, Spencer A. Circulating tumour DNA analysis in multiple myeloma. Oncotarget 2017; 8:90610-90611. [PMID: 29207579 PMCID: PMC5710860 DOI: 10.18632/oncotarget.21595] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Indexed: 12/12/2022] Open
Affiliation(s)
- Sridurga Mithraprabhu
- Andrew Spencer: Australian Centre for Blood Diseases, Monash University, Malignant Haematology & Stem Cell Transplantation, Alfred Hospital and Department of Clinical Haematology, Monash University, Melbourne, Australia
| | - Andrew Spencer
- Andrew Spencer: Australian Centre for Blood Diseases, Monash University, Malignant Haematology & Stem Cell Transplantation, Alfred Hospital and Department of Clinical Haematology, Monash University, Melbourne, Australia
| |
Collapse
|
1226
|
Davalos V, Martinez-Cardus A, Esteller M. The Epigenomic Revolution in Breast Cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2163-2174. [DOI: 10.1016/j.ajpath.2017.07.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/06/2017] [Accepted: 07/06/2017] [Indexed: 02/09/2023]
|
1227
|
Piton N, Lamy A, Sabourin JC. Séquençage des tumeurs : évolutions et révolutions. Cancer Radiother 2017; 21:580-583. [DOI: 10.1016/j.canrad.2017.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 07/22/2017] [Indexed: 11/26/2022]
|
1228
|
Manicone M, Poggiana C, Facchinetti A, Zamarchi R. Critical issues in the clinical application of liquid biopsy in non-small cell lung cancer. J Thorac Dis 2017; 9:S1346-S1358. [PMID: 29184673 DOI: 10.21037/jtd.2017.07.28] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Current therapeutic options for non-small cell lung cancer (NSCLC) patients are chemotherapy and targeted therapy directed mainly against epidermal growth factor receptor (EGFR) mutations and anaplastic lymphoma kinase (ALK) rearrangements. Targeted therapy relies on the availability of tumor biopsies for molecular profiling at diagnosis and to longitudinally monitor treatment response and resistance development. Unfortunately, tumor biopsy might be invasive, recover poor material of suboptimal quality, and cause sample bias due to tumor heterogeneity. Many studies have illustrated the potential of liquid biopsy as minimal invasive approach to respond to the urgent need for real time monitoring, stratification, and personalized optimized treatment in NSCLC patients. In principle, the liquid biopsy could provide the genetic landscape of primary and metastatic cancerous lesions, detecting "druggable" genomic alterations or associated with treatment resistance. Moreover, it would guarantee the prognostic/predictive biomarkers evaluation in patients for whom biopsies are inaccessible or difficult to repeat. At this regard, the prognostic value of circulating tumor cells (CTCs) in NSCLC patients has been largely investigated, but still their clinical utility as tumor biomarker is hampered by the lack of a consensus on the criteria necessary and sufficient to define them and on the standard operating procedures (SOPs) for their assessment. This review will summarize current developments on liquid biopsy in NSCLC, addressing the technology issues that contribute to the poor ability to track CTCs in the blood of NSCLC patients, thus limiting their extensive use in the clinical practice, and analyzing the solutions adopted to overcome such limits, on the road towards the clinical validation.
Collapse
Affiliation(s)
| | | | - Antonella Facchinetti
- IOV-IRCCS, Padova, Italy.,Department of Surgery, Oncology and Gastroenterology, Oncology Section, University of Padova, Padova, Italy
| | | |
Collapse
|
1229
|
Duréndez-Sáez E, Azkárate A, Meri M, Calabuig-Fariñas S, Aguilar-Gallardo C, Blasco A, Jantus-Lewintre E, Camps C. New insights in non-small-cell lung cancer: circulating tumor cells and cell-free DNA. J Thorac Dis 2017; 9:S1332-S1345. [PMID: 29184672 DOI: 10.21037/jtd.2017.06.112] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Lung cancer is the second most frequent tumor and the leading cause of death by cancer in both men and women. Increasing knowledge about the cancer genome and tumor environment has led to a new setting in which morphological and molecular characterization is needed to treat patients in the most personalized way in order to achieve better outcomes. Since tumor products can be detected in body fluids, the liquid biopsy, particularly, peripheral blood, has emerged as a new source for lung cancer biomarker's analysis. A variety of tumor components can be used for this purpose. Among them, circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) should be especially considered. Different detection methods for both CTCs and ctDNA have been and are being developed to improve the sensitivity and specificity of these tests. This would lead to better characterization and would solve some clinical doubts at different disease evolution times, e.g., intratumoral or temporal heterogeneity, difficulty in the obtaining a tumor sample, etc., and would also avoid the side effects of very expensive and complicated tumor obtaining interventions. CTCs and ctDNA are useful in different lung cancer settings. Their value has been shown for the early diagnosis, prognosis, prediction of treatment efficacy, monitoring responses and early detection of lung cancer relapse. CTCs have still not been validated for use in clinical settings in non-small-cell lung cancer (NSCLC), while ctDNA has been approved by the Food and Drug Administration (FDA) and European Medical Association (EMA), and the main clinical guidelines used for detect different epidermal growth factor receptor (EGFR) mutations and the monitoring and treatment choice of mutated patients with tyrosine kinase inhibitors (TKIs). This review, describes how ctDNA seem to be winning the race against CTCs from the laboratory bench to clinical practice due to easier obtaining methods, manipulation and its implementation into clinical practice.
Collapse
Affiliation(s)
- Elena Duréndez-Sáez
- Molecular Oncology Laboratory, Fundación Hospital General Universitario de Valencia, Valencia, Spain
| | - Aitor Azkárate
- Department of Oncology, University Hospital Son Espases, Palma de Mallorca, Spain
| | - Marina Meri
- Department of Medical Oncology, Hospital General Universitario de Valencia, Valencia, Spain
| | - Silvia Calabuig-Fariñas
- Molecular Oncology Laboratory, Fundación Hospital General Universitario de Valencia, Valencia, Spain.,Department of Pathology, Universitat de València, Valencia, Spain.,CIBERONC, Valencia, Spain
| | | | - Ana Blasco
- Department of Medical Oncology, Hospital General Universitario de Valencia, Valencia, Spain.,CIBERONC, Valencia, Spain
| | - Eloisa Jantus-Lewintre
- Molecular Oncology Laboratory, Fundación Hospital General Universitario de Valencia, Valencia, Spain.,CIBERONC, Valencia, Spain.,Department of Biotechnology, Universitat Politècnica de València, Valencia, Spain
| | - Carlos Camps
- Molecular Oncology Laboratory, Fundación Hospital General Universitario de Valencia, Valencia, Spain.,Department of Medical Oncology, Hospital General Universitario de Valencia, Valencia, Spain.,CIBERONC, Valencia, Spain.,Department of Medicine, Universitat de València, Valencia, Spain
| |
Collapse
|
1230
|
Abstract
INTRODUCTION Minimally invasive methods will augment the clinical approach for establishing the diagnosis or monitoring treatment response of central nervous system tumors. Liquid biopsy by blood or cerebrospinal fluid sampling holds promise in this regard. Areas covered: In this literature review, the authors highlight recent studies describing the analysis of circulating tumor cells, cell free nucleic acids, and extracellular vesicles as strategies to accomplish liquid biopsy in glioblastoma and metastatic tumors. The authors then discuss the continued efforts to improve signal detection, standardize the liquid biopsy handling and preparation, develop platforms for clinical application, and establish a role for liquid biopsies in personalized medicine. Expert commentary: As the technologies used to analyze these biomarkers continue to evolve, we propose that there is a future potential to precisely diagnose and monitor treatment response with liquid biopsies.
Collapse
Affiliation(s)
- Ganesh M. Shankar
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - Leonora Balaj
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - Shannon L. Stott
- Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Brian Nahed
- Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Bob S. Carter
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
1231
|
Sahengbieke S, Wang J, Li X, Wang Y, Lai M, Wu J. Circulating cell-free high mobility group AT-hook 2 mRNA as a detection marker in the serum of colorectal cancer patients. J Clin Lab Anal 2017; 32:e22332. [PMID: 28948632 DOI: 10.1002/jcla.22332] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 09/01/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Detection of circulating cell-free mRNA serves as noninvasive tools for cancer diagnosis. As an oncofetal protein, HMGA2 (high mobility group AT-hook 2) is upregulated in colorectal cancer (CRC) tissues. However, it is not clear whether the increased levels of circulating cell-free HMGA2 mRNA functions as potential biomarkers for improved diagnosis of CRC. METHODS To assess its clinical significance in diagnosis and prediction, we evaluated serum levels of circulating HMGA2 mRNA in CRC patients and in healthy controls. In this study, 83 CRC patients and 11 normal controls were enrolled in this study. We used real-time quantitative reverse transcription-PCR to evaluate the plasma mRNA levels of HMGA2 and analyze the correlation between their expression and clinicopathologic characteristics. RESULTS We found that the levels of HMGA2 mRNA were significantly higher in CRC patients compared with healthy volunteers. The patients with right-sided CRC, colon cancer, positive nerve infiltration, positive vascular invasion, negative microsatellite instability (MSI), and increasing in serum carbohydrate antigen (CA) 199 had higher levels of plasma HMGA2 mRNA. A strong positive correlation between circulating cell-free HMGA2 mRNA and CA199 level in serum was found in our study. Furthermore, statistical analysis revealed that levels of HMGA2 mRNA in plasma and in tumors were strictly correlated. CONCLUSIONS Collectively, our data suggested that cell-free HMGA2 mRNA in plasma might function as a novel diagnostic marker for CRC.
Collapse
Affiliation(s)
- Sana Sahengbieke
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jian Wang
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiangwei Li
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yuhong Wang
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Maode Lai
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jingjing Wu
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
1232
|
Ramón Y Cajal S, Capdevila C, Hernandez-Losa J, De Mattos-Arruda L, Ghosh A, Lorent J, Larsson O, Aasen T, Postovit LM, Topisirovic I. Cancer as an ecomolecular disease and a neoplastic consortium. Biochim Biophys Acta Rev Cancer 2017; 1868:484-499. [PMID: 28947238 DOI: 10.1016/j.bbcan.2017.09.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 09/19/2017] [Accepted: 09/20/2017] [Indexed: 12/26/2022]
Abstract
Current anticancer paradigms largely target driver mutations considered integral for cancer cell survival and tumor progression. Although initially successful, many of these strategies are unable to overcome the tremendous heterogeneity that characterizes advanced tumors, resulting in the emergence of resistant disease. Cancer is a rapidly evolving, multifactorial disease that accumulates numerous genetic and epigenetic alterations. This results in wide phenotypic and molecular heterogeneity within the tumor, the complexity of which is further amplified through specific interactions between cancer cells and the tumor microenvironment. In this context, cancer may be perceived as an "ecomolecular" disease that involves cooperation between several neoplastic clones and their interactions with immune cells, stromal fibroblasts, and other cell types present in the microenvironment. This collaboration is mediated by a variety of secreted factors. Cancer is therefore analogous to complex ecosystems such as microbial consortia. In the present article, we comment on the current paradigms and perspectives guiding the development of cancer diagnostics and therapeutics and the potential application of systems biology to untangle the complexity of neoplasia. In our opinion, conceptualization of neoplasia as an ecomolecular disease is warranted. Advances in knowledge pertinent to the complexity and dynamics of interactions within the cancer ecosystem are likely to improve understanding of tumor etiology, pathogenesis, and progression. This knowledge is anticipated to facilitate the design of new and more effective therapeutic approaches that target the tumor ecosystem in its entirety.
Collapse
Affiliation(s)
- Santiago Ramón Y Cajal
- Translational Molecular Pathology, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain; Pathology Department, Vall d'Hebron Hospital, 08035 Barcelona, Spain; Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Spain.
| | - Claudia Capdevila
- Translational Molecular Pathology, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Javier Hernandez-Losa
- Translational Molecular Pathology, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain; Pathology Department, Vall d'Hebron Hospital, 08035 Barcelona, Spain; Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Spain
| | - Leticia De Mattos-Arruda
- Translational Molecular Pathology, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Abhishek Ghosh
- Lady Davis Institute, JGH, SMBD, Gerald-Bronfman Department of Oncology, McGill University QC, Montreal H3T 1E2, Canada
| | - Julie Lorent
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, 171 65 Solna, Sweden
| | - Ola Larsson
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, 171 65 Solna, Sweden
| | - Trond Aasen
- Translational Molecular Pathology, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain; Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Spain
| | - Lynne-Marie Postovit
- Cancer Research Institute of Northern Alberta Department of Oncology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Ivan Topisirovic
- Lady Davis Institute, JGH, SMBD, Gerald-Bronfman Department of Oncology, McGill University QC, Montreal H3T 1E2, Canada
| |
Collapse
|
1233
|
Singh AP, Li S, Cheng H. Circulating DNA in EGFR-mutated lung cancer. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:379. [PMID: 29057239 DOI: 10.21037/atm.2017.07.10] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Circulating tumor DNA (ctDNA) consists of short double stranded DNA fragments that are released by tumors including non-small cell lung cancer (NSCLC). With the identification of driver mutations in the epidermal growth factor receptor (EGFR) gene and development of targeted tyrosine kinase inhibitors (TKIs), the clinical outcome of NSCLC patients in this subgroup has improved tremendously. The gold standard to assess EGFR mutation is through tissue biopsy, which can be limited by difficulty in accessing the tumor, inability of patients to tolerate invasive procedures, insufficient sample for molecular testing and inability to capture intratumoral heterogeneity. The great need for rapid and accurate identification of activating EGFR mutations in NSCLC patients paves the road for ctDNA technology. Studies have demonstrated ctDNA to be a reliable complement to tumor genotyping. Platforms like digital polymerase chain reaction (PCR) and next-generation sequencing based analyses have made it possible to identify EGFR mutations in plasma with high sensitivity and specificity. This article will provide an overview on ctDNA in the context of EGFR mutated NSCLC, especially its emerging applications in diagnosis, disease surveillance, treatment monitoring and detection of resistance mechanisms.
Collapse
Affiliation(s)
- Aditi P Singh
- Department of Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - Shenduo Li
- Department of Medicine, Jacobi Medical Center, Bronx, NY, USA
| | - Haiying Cheng
- Department of Oncology, Montefiore Medical Center, Bronx, NY, USA
| |
Collapse
|
1234
|
Fiala C, Diamandis EP. Circulating tumor DNA for personalized lung cancer monitoring. BMC Med 2017; 15:157. [PMID: 28814291 PMCID: PMC5559853 DOI: 10.1186/s12916-017-0921-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 08/01/2017] [Indexed: 11/15/2022] Open
Abstract
Advances in deep sequencing technology have led to developments in personalized medicine. Here, we describe the implications of a recent investigation that sequenced ctDNA from the plasma of non-small cell lung cancer patients to develop personalized ctDNA tests. These 'liquid biopsies' have shown promise in monitoring tumor growth and response to treatment, providing a timely overview of mutations present in the tumor. We discuss the advantages of this budding approach, as well as its challenges and drawbacks, while also providing areas for further investigation and an outlook for the future.
Collapse
Affiliation(s)
- Clare Fiala
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, 60 Murray St. Box 32, Floor 6, Rm L6-201, Toronto, ON, MST 3L9, Canada
| | - Eleftherios P Diamandis
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, 60 Murray St. Box 32, Floor 6, Rm L6-201, Toronto, ON, MST 3L9, Canada. .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada. .,Department of Clinical Biochemistry, University Health Network, 60 Murray St. Box 32, Floor 6, Rm L6-201, Toronto, ON, MST 3L9, Canada.
| |
Collapse
|
1235
|
ALK Status Assessment with Liquid Biopsies of Lung Cancer Patients. Cancers (Basel) 2017; 9:cancers9080106. [PMID: 28805673 PMCID: PMC5575609 DOI: 10.3390/cancers9080106] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 08/06/2017] [Accepted: 08/10/2017] [Indexed: 12/22/2022] Open
Abstract
Patients with advanced stage non-small cell lung carcinoma (NSCLC) harboring an anaplastic lymphoma kinase ALK gene rearrangement, detected from a tissue sample, can benefit from targeted ALK inhibitor treatment. However, while treatment is initially effective in most cases, relapse or progression occurs due to different resistance mechanisms including mutations in the tyrosine kinase domain of echinoderm microtubule-associated protein-like 4 (EML44)-ALK. The liquid biopsy concept has recently radically changed the clinical care of NSCLC patients, in particular for those harboring an epidermal growth factor receptor (EGFR) gene mutation. Therefore, liquid biopsy is an alternative or complementary method to tissue biopsy for the detection of some resistance mutations in EGFR arising during tyrosine kinase inhibitor treatment. Moreover, in some frail patients, or if the tumor lesion is not accessible to a tissue biopsy, a liquid biopsy can also detect some activating mutations in EGFR on initial assessment. Recent studies have evaluated the possibility of also using a liquid biopsy approach to detect an ALK rearrangement and/or the emergence during inhibitor treatment of some resistance mutations in ALK. These assessments can be performed by studying circulating tumor cells by fluorescent in situ hybridization and by immunocytochemistry and/or after the isolation of RNA from plasma samples, free or associated with platelets. Thus, the liquid biopsy may be a complementary or sometimes alternative method for the assessment of the ALK status in certain NSCLC patients, as well as a non-invasive approach for early detection of ALK mutations. In this review, we highlight the current data concerning the role of the liquid biopsy for the ALK status assessment for NSCLC patients, and we compare the different approaches for this evaluation from blood samples.
Collapse
|
1236
|
Clinical value of ctDNA in upper-GI cancers: A systematic review and meta-analysis. Biochim Biophys Acta Rev Cancer 2017; 1868:394-403. [PMID: 28801248 DOI: 10.1016/j.bbcan.2017.08.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/05/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND The recent expanding technical possibilities to detect tumor derived mutations in blood, so-called circulating tumor DNA (ctDNA), has rapidly increased the interest in liquid biopsies. This review and meta-analysis explores the clinical value of ctDNA in malignancies of the upper gastro-intestinal tract. METHODS PubMed, Cochrane and Embase databases were searched to identify studies reporting the diagnostic, prognostic or predictive value of ctDNA in patients with esophageal, gastric and pancreatic cancer, until January 2017. The diagnostic accuracy and, using random-effect pair-wise meta-analyses, the prognostic value of ctDNA was assessed. RESULTS A total of 34 studies met the inclusion criteria. For esophageal and gastric cancer, amplification of oncogenes in blood, such as HER2 and MYC, can be relevant for diagnostic purposes, and to predict treatment response in certain patient subpopulations. Given the limited number of studies assessing the role of ctDNA in esophageal and gastric cancer, the meta-analysis estimated the diagnostic accuracy and predictive value of ctDNA in pancreatic cancer only (n=10). The pooled sensitivity and specificity of ctDNA as a diagnostic tool in pancreatic cancer were 28% and 95%, respectively. Patients with pancreatic cancer and detectable ctDNA demonstrated a worse overall survival compared to patients with undetectable ctDNA (HR 1.92, 95% confidence interval (CI) 1.15-3.22, p=0.01). CONCLUSION The presence of ctDNA is significantly associated with a poor prognosis in patients with pancreatic cancer. The use of ctDNA in clinical practice is promising, although standardization of sequencing techniques and further development of high-sensitive detection methods is needed.
Collapse
|
1237
|
Moreno L, Pearson ADJ, Paoletti X, Jimenez I, Geoerger B, Kearns PR, Zwaan CM, Doz F, Baruchel A, Vormoor J, Casanova M, Pfister SM, Morland B, Vassal G. Early phase clinical trials of anticancer agents in children and adolescents - an ITCC perspective. Nat Rev Clin Oncol 2017; 14:497-507. [PMID: 28508875 DOI: 10.1038/nrclinonc.2017.59] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In the past decade, the landscape of drug development in oncology has evolved dramatically; however, this paradigm shift remains to be adopted in early phase clinical trial designs for studies of molecularly targeted agents and immunotherapeutic agents in paediatric malignancies. In drug development, prioritization of drugs on the basis of knowledge of tumour biology, molecular 'drivers' of disease and a drug's mechanism of action, and therapeutic unmet needs are key elements; these aspects are relevant to early phase paediatric trials, in which molecular profiling is strongly encouraged. Herein, we describe the strategy of the Innovative Therapies for Children with Cancer (ITCC) Consortium, which advocates for the adoption of trial designs that enable uninterrupted patient recruitment, the extrapolation from studies in adults when possible, and the inclusion of expansion cohorts. If a drug has neither serious dose-related toxicities nor a narrow therapeutic index, then studies should generally be started at the adult recommended phase II dose corrected for body surface area, and act as dose-confirmation studies. The use of adaptive trial designs will enable drugs with promising activity to progress rapidly to randomized studies and, therefore, will substantially accelerate drug development for children and adolescents with cancer.
Collapse
Affiliation(s)
- Lucas Moreno
- Paediatric Phase I-II Clinical Trials Unit, Paediatric Haematology &Oncology, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Andrew D J Pearson
- Paediatric Drug Development, Children and Young People's Unit, The Royal Marsden NHS Foundation Trust, Sutton, UK; and at the Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, Sutton, UK
| | - Xavier Paoletti
- Biostatistics and Epidemiology, INSERM U1018, Gustave Roussy, Paris, France
| | - Irene Jimenez
- Department of Paediatric, Adolescents and Young Adults Oncology, Institut Curie; and at the University Paris Descartes, Paris, France
| | - Birgit Geoerger
- Department of Paediatric and Adolescent Oncology, CNRS UMR 8203 Vectorology and Anticancer Treatments, Gustave Roussy, University Paris-Sud, Villejuif, France
| | - Pamela R Kearns
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - C Michel Zwaan
- Department of Paediatric Oncology/Haematology, Erasmus MC/Sophia Children's Hospital, Rotterdam, Netherlands
| | - Francois Doz
- Department of Paediatric, Adolescents and Young Adults Oncology, Institut Curie; and at the University Paris Descartes, Paris, France
| | - Andre Baruchel
- Department of Paediatric Haematology, Hôpital Robert Debré, AP-HP; and at the University Paris Diderot, Paris, France
| | - Josef Vormoor
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University; and at the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Michela Casanova
- Paediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Stefan M Pfister
- German Cancer Research Center (DKFZ); German Cancer Consortium (DKTK); and at the Heidelberg University Hospital, Heidelberg, Germany
| | - Bruce Morland
- Department of Paediatric Oncology, Birmingham Children's Hospital, Birmingham, UK
| | - Gilles Vassal
- Department of Clinical Research, Gustave Roussy, Paris-Sud University, Paris, France
| |
Collapse
|
1238
|
Overcoming Oncogenic Mediated Tumor Immunity in Prostate Cancer. Int J Mol Sci 2017; 18:ijms18071542. [PMID: 28714919 PMCID: PMC5536030 DOI: 10.3390/ijms18071542] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 06/26/2017] [Accepted: 06/29/2017] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy is being tested intensively in clinical trials for prostate cancer; it includes immune checkpoint inhibition, prostate specific antigen (PSA) vaccines and dendritic cell-based strategies. Despite increasing evidence for clinical responses, the consensus of multiple trials is that prostate cancers are poorly responsive to immunotherapy. Prostate cancer has a high degree of pathological and genetic heterogeneity compared to other cancer types, which may account for immunotherapeutic resistance. This hypothesis also implies that select types of prostate tumors may be differentially responsive to immune-based strategies and that the clinical stage, pathological grade and underlying genetic landscape may be important criteria in identifying tumors that respond to immune therapies. One strategy is to target oncogenic driver pathways in combination with immunotherapies with the goal of overcoming tumor immunity and broadening the number of patients achieving a clinical response. In this analysis, we address the hypothesis that driver oncogenic signaling pathways regulate cancer progression, tumor immunity and resistance to current immune therapeutics in prostate cancer. We propose that increased responsiveness may be achieved through the combined use of immunotherapies and inhibitors targeting tumor cell autonomous pathways that contribute towards anti-tumor immunity in patients with prostate cancer.
Collapse
|
1239
|
|
1240
|
Hicks JK, Saller J, Wang E, Boyle T, Gray JE. Cell-free circulating tumor DNA supplementing tissue biopsies for identification of targetable mutations: Implications for precision medicine and considerations for reconciling results. Lung Cancer 2017; 111:135-138. [PMID: 28838384 DOI: 10.1016/j.lungcan.2017.06.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 06/21/2017] [Accepted: 06/22/2017] [Indexed: 10/19/2022]
Abstract
Cell-free circulating tumor DNA (ctDNA) next-generation sequencing (NGS) is emerging as a noninvasive technique for detecting targetable mutations. We describe two lung adenocarcinoma cases that show the clinical utility of supplementing tumor biopsy molecular interrogation with ctDNA NGS. For both cases, ctDNA NGS identified actionable mutations that were previously not reported by molecular interrogation of tissue. Explanations are provided for the observed differences between ctDNA and tumor biopsy genomic results along with considerations for reconciling findings. Case 1 consisted of a patient with multiple lesions in the left and right lung that was initially suspected to be related to malignancy. A tumor biopsy was positive for EGFR-mutated lung cancer. ctDNA NGS reported an activating KRAS mutation, which was unexpected given the rare occurrence of EGFR/KRAS co-mutations. Radiologic imaging and ctDNA NGS resulted in the diagnoses of synchronous EGFR-mutated left lung cancer and KRAS-mutated right lung cancer. The second case describes a patient who was negative for RET rearrangements by tissue interrogation, but positive for a RET-KIF5B fusion by ctDNA NGS. Further tissue analysis demonstrated heterogeneity was the cause of differing results. We demonstrate that supplementing tumor biopsies with ctDNA NGS has a crucial role in patient care. Understanding the causes of differing ctDNA and tumor biopsy genomic results is essential for reconciling findings and application to precision medicine management.
Collapse
Affiliation(s)
- J Kevin Hicks
- DeBartolo Family Personalized Medicine Institute, Department of Individualized Cancer Management, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - James Saller
- DeBartolo Family Personalized Medicine Institute, Department of Individualized Cancer Management, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Emilie Wang
- Department of Internal Medicine, University of South Florida, Tampa, FL, USA
| | - Theresa Boyle
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jhanelle E Gray
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| |
Collapse
|
1241
|
Pedrol E, Garcia-Algar M, Massons J, Nazarenus M, Guerrini L, Martínez J, Rodenas A, Fernandez-Carrascal A, Aguiló M, Estevez LG, Calvo I, Olano-Daza A, Garcia-Rico E, Díaz F, Alvarez-Puebla RA. Optofluidic device for the quantification of circulating tumor cells in breast cancer. Sci Rep 2017. [PMID: 28623262 PMCID: PMC5473916 DOI: 10.1038/s41598-017-04033-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Metastatic cancer patients require a continuous monitoring during the sequential treatment cycles to carefully evaluate their disease evolution. Repetition of biopsies is very invasive and not always feasible. Herein, we design and demonstrate a 3D-flow focusing microfluidic device, where all optics are integrated into the chip, for the fluorescence quantification of CTCs in real samples. To test the chip performance, two cell membrane targets, the epithelial cell adhesion molecule, EpCAM, and the receptor tyrosine-protein kinase, HER2, are selected. The efficiency of the platform is demonstrated on cell lines and in a variety of healthy donors and metastatic-breast cancer patients.
Collapse
Affiliation(s)
- Eric Pedrol
- Física i Cristal•lografia de Materials i Nanomaterials and EmaS. Universitat Rovira i Virgili, Carrer Marcel•lí Domingo 1, 43007, Tarragona, Spain
| | - Manuel Garcia-Algar
- Departamento de Química Física e Inorgánica and EmaS, Universitat Rovira i Virgili, Carrer Marcel•lí Domingo 1, 43007, Tarragona, Spain
| | - Jaume Massons
- Física i Cristal•lografia de Materials i Nanomaterials and EmaS. Universitat Rovira i Virgili, Carrer Marcel•lí Domingo 1, 43007, Tarragona, Spain
| | - Moritz Nazarenus
- Departamento de Química Física e Inorgánica and EmaS, Universitat Rovira i Virgili, Carrer Marcel•lí Domingo 1, 43007, Tarragona, Spain
| | - Luca Guerrini
- Departamento de Química Física e Inorgánica and EmaS, Universitat Rovira i Virgili, Carrer Marcel•lí Domingo 1, 43007, Tarragona, Spain
| | - Javier Martínez
- Física i Cristal•lografia de Materials i Nanomaterials and EmaS. Universitat Rovira i Virgili, Carrer Marcel•lí Domingo 1, 43007, Tarragona, Spain
| | - Airan Rodenas
- Física i Cristal•lografia de Materials i Nanomaterials and EmaS. Universitat Rovira i Virgili, Carrer Marcel•lí Domingo 1, 43007, Tarragona, Spain
| | - Ana Fernandez-Carrascal
- Departamento de Química Física e Inorgánica and EmaS, Universitat Rovira i Virgili, Carrer Marcel•lí Domingo 1, 43007, Tarragona, Spain
| | - Magdalena Aguiló
- Física i Cristal•lografia de Materials i Nanomaterials and EmaS. Universitat Rovira i Virgili, Carrer Marcel•lí Domingo 1, 43007, Tarragona, Spain
| | - Laura G Estevez
- Fundacion de Investigacion HM Hospitales, San Bernardo 101, 28015, Madrid, Spain.,Centro Integral Oncologico Clara Campal (CIOCC), Oña 10, 28050, Madrid, Spain
| | - Isabel Calvo
- Fundacion de Investigacion HM Hospitales, San Bernardo 101, 28015, Madrid, Spain.,Centro Integral Oncologico Clara Campal (CIOCC), Oña 10, 28050, Madrid, Spain
| | - Ana Olano-Daza
- Department of Medical Oncology, Hospital Universitario HM Torrelodones, Castillo de Olivares s/n, 28250, Torrelodones, Spain
| | - Eduardo Garcia-Rico
- Fundacion de Investigacion HM Hospitales, San Bernardo 101, 28015, Madrid, Spain. .,Centro Integral Oncologico Clara Campal (CIOCC), Oña 10, 28050, Madrid, Spain. .,Department of Medical Oncology, Hospital Universitario HM Torrelodones, Castillo de Olivares s/n, 28250, Torrelodones, Spain. .,School of Medicine, San Pablo CEU, Calle Julián Romea, 18, 28003, Madrid, Spain.
| | - Francesc Díaz
- Física i Cristal•lografia de Materials i Nanomaterials and EmaS. Universitat Rovira i Virgili, Carrer Marcel•lí Domingo 1, 43007, Tarragona, Spain.
| | - Ramon A Alvarez-Puebla
- Departamento de Química Física e Inorgánica and EmaS, Universitat Rovira i Virgili, Carrer Marcel•lí Domingo 1, 43007, Tarragona, Spain. .,ICREA, Passeig Lluís Companys 23, 08010, Barcelona, Spain.
| |
Collapse
|
1242
|
Ahmed Z, Gravel S. Intratumor Heterogeneity and Circulating Tumor Cell Clusters. Mol Biol Evol 2017; 35:2135-2144. [PMID: 29897504 DOI: 10.1093/molbev/msy115] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 05/23/2018] [Indexed: 12/19/2022] Open
Abstract
Genetic diversity plays a central role in tumor progression, metastasis, and resistance to treatment. Experiments are shedding light on this diversity at ever finer scales, but interpretation is challenging. Using recent progress in numerical models, we simulate macroscopic tumors to investigate the interplay between growth dynamics, microscopic composition, and circulating tumor cell cluster diversity. We find that modest differences in growth parameters can profoundly change microscopic diversity. Simple outwards expansion leads to spatially segregated clones and low diversity, as expected. However, a modest cell turnover can result in an increased number of divisions and mixing among clones resulting in increased microscopic diversity in the tumor core. Using simulations to estimate power to detect such spatial trends, we find that multiregion sequencing data from contemporary studies is marginally powered to detect the predicted effects. Slightly larger samples, improved detection of rare variants, or sequencing of smaller biopsies or circulating tumor cell clusters would allow one to distinguish between leading models of tumor evolution. The genetic composition of circulating tumor cell clusters, which can be obtained from non-invasive blood draws, is therefore informative about tumor evolution and its metastatic potential.
Collapse
Affiliation(s)
- Zafarali Ahmed
- School of Computer Science, McGill University, Montréeal, QC, Canada
| | - Simon Gravel
- Department of Human Genetics, McGill University, Montréeal, QC, Canada.,McGill University and Genome Québec Innovation Centre, Montréal, QC, Canada
| |
Collapse
|
1243
|
Prostate Cancer Stem Cell Markers Drive Progression, Therapeutic Resistance, and Bone Metastasis. Stem Cells Int 2017; 2017:8629234. [PMID: 28690641 PMCID: PMC5485361 DOI: 10.1155/2017/8629234] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 03/29/2017] [Accepted: 05/03/2017] [Indexed: 02/07/2023] Open
Abstract
Metastatic or recurrent tumors are the primary cause of cancer-related death. For prostate cancer, patients diagnosed with local disease have a 99% 5-year survival rate; however, this 5-year survival rate drops to 28% in patients with metastatic disease. This dramatic decline in survival has driven interest in discovering new markers able to identify tumors likely to recur and in developing new methods to prevent metastases from occurring. Biomarker discovery for aggressive tumor cells includes attempts to identify cancer stem cells (CSCs). CSCs are defined as tumor cells capable of self-renewal and regenerating the entire tumor heterogeneity. Thus, it is hypothesized that CSCs may drive primary tumor aggressiveness, metastatic colonization, and therapeutic relapse. The ability to identify these cells in the primary tumor or circulation would provide prognostic information capable of driving prostate cancer treatment decisions. Further, the ability to target these CSCs could prevent tumor metastasis and relapse after therapy allowing for prostate cancer to finally be cured. Here, we will review potential CSC markers and highlight evidence that describes how cells expressing each marker may drive prostate cancer progression, metastatic colonization and growth, tumor recurrence, and resistance to treatment.
Collapse
|
1244
|
Ades F, Tryfonidis K, Zardavas D. The past and future of breast cancer treatment-from the papyrus to individualised treatment approaches. Ecancermedicalscience 2017; 11:746. [PMID: 28690677 PMCID: PMC5481194 DOI: 10.3332/ecancer.2017.746] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Indexed: 12/16/2022] Open
Abstract
Cancer is one of the oldest diseases ever described, since ancient Egypt there have always been attempts to treat and cure this illness. The growing body of knowledge about breast cancer biology and improvements in surgical and medical treatments has been built over time with contributions from many talented and enthusiastic physicians and researchers. Medical advances have changed the approach from a previously incurable condition, into a surgical disease. Further improvements in cancer biology have allowed the development of systemic treatments, hormonal therapies, and targeted drugs. The description of the molecular intrinsic subtypes of breast cancer clarified the understanding of breast cancer as a group of heterogeneous diseases, associated with different clinical outcomes, and therapeutic opportunities. This paper reviews how breast cancer treatment has improved since the earliest descriptions, in ancient times, and how future approaches, such as gene signatures, molecular profiling, and liquid biopsies, aim to further develop individualised treatments and improve treatment outcomes.
Collapse
Affiliation(s)
- Felipe Ades
- Hospital Albert Einstein, Avenida Albert Einstein, 627 - Morumbi, São Paulo - SP, 05652-900 Brazil
| | - Konstantinos Tryfonidis
- European Organisation for Research and Treatment of Cancer, Avenue E. Mounier 83/11, 1200 Brussels, Belgium
| | - Dimitrios Zardavas
- Breast International Group (BIG), Boulevard de Waterloo 76, Brussels 1000, Belgium
| |
Collapse
|
1245
|
Caivano A, La Rocca F, Laurenzana I, Trino S, De Luca L, Lamorte D, Del Vecchio L, Musto P. Extracellular Vesicles in Hematological Malignancies: From Biology to Therapy. Int J Mol Sci 2017; 18:E1183. [PMID: 28574430 PMCID: PMC5486006 DOI: 10.3390/ijms18061183] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 05/29/2017] [Accepted: 05/30/2017] [Indexed: 12/22/2022] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous group of particles, between 15 nanometers and 10 microns in diameter, released by almost all cell types in physiological and pathological conditions, including tumors. EVs have recently emerged as particularly interesting informative vehicles, so that they could be considered a true "cell biopsy". Indeed, EV cargo, including proteins, lipids, and nucleic acids, generally reflects the nature and status of the origin cells. In some cases, EVs are enriched of peculiar molecular cargo, thus suggesting at least a degree of specific cellular packaging. EVs are identified as important and critical players in intercellular communications in short and long distance interplays. Here, we examine the physiological role of EVs and their activity in cross-talk between bone marrow microenvironment and neoplastic cells in hematological malignancies (HMs). In these diseases, HM EVs can modify tumor and bone marrow microenvironment, making the latter "stronger" in supporting malignancy, inducing drug resistance, and suppressing the immune system. Moreover, EVs are abundant in biologic fluids and protect their molecular cargo against degradation. For these and other "natural" characteristics, EVs could be potential biomarkers in a context of HM liquid biopsy and therapeutic tools. These aspects will be also analyzed in this review.
Collapse
Affiliation(s)
- Antonella Caivano
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 858028 Rionero in Vulture, Italy.
| | - Francesco La Rocca
- Laboratory of Clinical Research and Advanced Diagnostics, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Ilaria Laurenzana
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 858028 Rionero in Vulture, Italy.
| | - Stefania Trino
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 858028 Rionero in Vulture, Italy.
| | - Luciana De Luca
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 858028 Rionero in Vulture, Italy.
| | - Daniela Lamorte
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 858028 Rionero in Vulture, Italy.
| | - Luigi Del Vecchio
- CEINGE-Biotecnologie Avanzate scarl, Federico II University, 80138 Naples, Italy.
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, 80138 Naples, Italy.
| | - Pellegrino Musto
- Scientific Direction, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| |
Collapse
|
1246
|
Siravegna G, Sartore-Bianchi A, Mussolin B, Cassingena A, Amatu A, Novara L, Buscarino M, Corti G, Crisafulli G, Bartolini A, Tosi F, Erlander M, Di Nicolantonio F, Siena S, Bardelli A. Tracking aCAD-ALK gene rearrangement in urine and blood of a colorectal cancer patient treated with an ALK inhibitor. Ann Oncol 2017; 28:1302-1308. [DOI: 10.1093/annonc/mdx095] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
1247
|
El Bairi K, Amrani M, Kandhro AH, Afqir S. Prediction of therapy response in ovarian cancer: Where are we now? Crit Rev Clin Lab Sci 2017; 54:233-266. [PMID: 28443762 DOI: 10.1080/10408363.2017.1313190] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Therapy resistance is a major challenge in the management of ovarian cancer (OC). Advances in detection and new technology validation have led to the emergence of biomarkers that can predict responses to available therapies. It is important to identify predictive biomarkers to select resistant and sensitive patients in order to reduce important toxicities, to reduce costs and to increase survival. The discovery of predictive and prognostic biomarkers for monitoring therapy is a developing field and provides promising perspectives in the era of personalized medicine. This review article will discuss the biology of OC with a focus on targetable pathways; current therapies; mechanisms of resistance; predictive biomarkers for chemotherapy, antiangiogenic and DNA-targeted therapies, and optimal cytoreductive surgery; and the emergence of liquid biopsy using recent studies from the Medline database and ClinicalTrials.gov.
Collapse
Affiliation(s)
- Khalid El Bairi
- a Faculty of Medicine and Pharmacy , Mohamed Ist University , Oujda , Morocco
| | - Mariam Amrani
- b Equipe de Recherche ONCOGYMA, Faculty of Medicine, Pathology Department , National Institute of Oncology, Université Mohamed V , Rabat , Morocco
| | - Abdul Hafeez Kandhro
- c Department of Biochemistry , Healthcare Molecular and Diagnostic Laboratory , Hyderabad , Pakistan
| | - Said Afqir
- d Department of Medical Oncology , Mohamed VI University Hospital , Oujda , Morocco
| |
Collapse
|
1248
|
Puccini A, Berger MD, Zhang W, Lenz HJ. What We Know About Stage II and III Colon Cancer: It's Still Not Enough. Target Oncol 2017; 12:265-275. [PMID: 28504299 PMCID: PMC7489295 DOI: 10.1007/s11523-017-0494-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The introduction of oxaliplatin as adjuvant treatment for stage III colon cancer in 2004 has been the last practice changing progress in adjuvant treatment for patients with early colon cancer. Since then, many prognostic and predictive biomarkers have been studied, but only DNA mismatch repair status has been validated as having an important prognostic value. Accordingly, TNM and clinical-pathological patterns, such as pT4 lesions and lymph node sampling <12 nodes, are the main factors that guide physicians' choice regarding adjuvant treatment. More recently, many biomarkers showed promising results: POLE, ErbB2, CDX2, SMAD4, BRAF and KRAS. In addition to these, immune-contexture, molecular classification, and gene signatures could become new ways to better classify colon cancer patients with more discriminatory power than TNM. The aim of this review is to report the state-of-the-art of prognostic and predictive factors in the adjuvant setting and which of these could modify clinical practice and maybe replace TNM classification.
Collapse
Affiliation(s)
- Alberto Puccini
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Suite 3456, Los Angeles, CA, 90033, USA
- Medical Oncology Unit, IRCCS AOU San Martino - IST, Genoa, Italy
| | - Martin D Berger
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Suite 3456, Los Angeles, CA, 90033, USA
| | - Wu Zhang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Suite 3456, Los Angeles, CA, 90033, USA
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Suite 3456, Los Angeles, CA, 90033, USA.
| |
Collapse
|
1249
|
Changes of Microrna Levels in Plasma of Patients with Rectal Cancer during Chemoradiotherapy. Int J Mol Sci 2017; 18:ijms18061140. [PMID: 28554991 PMCID: PMC5485964 DOI: 10.3390/ijms18061140] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 05/19/2017] [Accepted: 05/22/2017] [Indexed: 12/15/2022] Open
Abstract
Since the response to chemoradiotherapy in patients with locally advanced rectal cancer is heterogeneous, valid biomarkers are needed to monitor tumor response. Circulating microRNAs are promising candidates, however analyses of circulating microRNAs in rectal cancer are still rare. 111 patients with rectal cancer and 46 age-matched normal controls were enrolled. The expression levels of 30 microRNAs were analyzed in 17 pre-treatment patients’ plasma samples. Differentially regulated microRNAs were validated in 94 independent patients. For 52 of the 94 patients a paired comparison between pre-treatment and post-treatment samples was performed. miR-17, miR-18b, miR-20a, miR-31, and miR-193a_3p, were significantly downregulated in pre-treatment plasma samples of patients with rectal cancer (p < 0.05). miR-29c, miR-30c, and miR-195 showed a trend of differential regulation. After validation, miR-31 and miR-30c were significantly deregulated by a decrease of expression. In 52 patients expression analyses of the 8 microRNAs in matched pre-treatment and post-treatment samples showed a significant decrease for all microRNAs (p < 0.05) after treatment. Expression levels of miR-31 and miR-30c could serve as valid biomarkers if validated in a prospective study. Plasma microRNA expression levels do not necessarily represent miRNA expression levels in tumor tissue. Also, expression levels of microRNAs change during multimodal therapy.
Collapse
|
1250
|
|