51
|
Wang X, Yang M, Liu Q, Yang S, Geng X, Yang Y, Fa H, Wang Y, Hou C. An Ultrasensitive Electrochemical DNA Biosensor Based on Carboxylated Multi-walled Carbon Nanotube/Molybdenum Disulfide Composites for KRAS Gene Detection. ANAL SCI 2018; 35:441-448. [PMID: 30606912 DOI: 10.2116/analsci.18p518] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In this paper, an ultrasensitive electrochemical biosensor based on carboxylated multi-walled carbon nanotube/molybdenum disulfide composites (MWCNTs-COOH/MoS2) for the detection of KRAS gene is described. An easy, low-cost method, named one-step hydrothermal, was used for the synthesize of MWCNTs-COOH/MoS2 nanocomposites, and scanning electronic microscopy (SEM), high resolution transmission electron microscopy (HRTEM), Fourier transform infrared spectroscopy (FT-IR), X-ray photoelectron spectroscopy (XPS) and X-ray diffraction (XRD) were used for characterizing the prepared composites. Furthermore, cyclic voltammetry (CV) and differential pulse voltammetry (DPV) were employed for an electrochemical performance study of this biosensor. Under optimal conditions, the detection limit of target DNA achieved down to 3 fM (S/N = 3) with high sensitivity; the linear range with the logarithm of the concentrations of target DNA varied from 1.0 × 10-14 to 1.0 × 10-7 M. Finally, the practicality of our proposed sensor was verified by a determination of the KRAS gene in human serum samples with good accuracy and high precision due to the excellent conductivity and large active surface area of the MWCNTs-COOH/MoS2 nanocomposites. This proposed biosensor thus provides a practical method for the rapid and sensitive analysis of gene detection.
Collapse
Affiliation(s)
- Xiaojing Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University
| | - Mei Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University
| | - Qingyan Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University
| | - Siyi Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University
| | - Xintong Geng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University
| | - Yixia Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University
| | - Huanbao Fa
- College of Chemistry and Chemical Engineering, Chongqing University
| | - Yongzhong Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University
| | - Changjun Hou
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University
| |
Collapse
|
52
|
Park C, Park H, Lee HJ, Lee HS, Park KH, Choi CH, Na S. Double amplified colorimetric detection of DNA using gold nanoparticles, enzymes and a catalytic hairpin assembly. Mikrochim Acta 2018; 186:34. [PMID: 30564970 DOI: 10.1007/s00604-018-3154-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/07/2018] [Indexed: 02/07/2023]
Abstract
The authors describe an isothermal and ultrasensitive colorimetric DNA assay that consists of two amplification stages using enzymes and a catalytic hairpin assembly (CHA). The first step consists in the selective amplification of DNA using Klenow fragment and nicking enzyme. The second step consists in the amplification of the optical signal by using a catalytic hairpin assembly. After two amplification steps, the DNA reaction induces the aggregation of the red gold nanoparticles to give a blue color shift. The degree of aggregation can be quantified by measurement of the ratio of the UV-vis absorbances of the solutions at 620 and 524 nm which are the wavelengths of the aggregated gold nanoparticles and bare gold nanoparticles. The detection limit is as low as 3.1 fM. Due to the use of a specific enzyme, only the desired DNAs will be detected. The method can be applied to the determination of DNA of various lengths. Despite the presence of large amounts of wildtype DNA, it can readily detect a target DNA. Conceivably, the technique has a large potential because of its high sensitivity and selectivity. Graphical abstract Schematic presentation of DNA detection using gold nanoparticles (AuNP), enzymes and catalytic hairpin assembly (CHA). Effective DNA detection is achieved through the aggregation of AuNPs which is caused by DNA amplification using enzymes and signal amplification using CHA.
Collapse
Affiliation(s)
- Chanho Park
- Department of Mechanical Engineering, Korea University, Seoul, 136-701, Republic of Korea
| | - Hyunjun Park
- Department of Mechanical Engineering, Korea University, Seoul, 136-701, Republic of Korea
| | - Hye Jin Lee
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University, Seoul, 136-701, South Korea
| | - Hye Sun Lee
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University, Seoul, 136-701, South Korea
| | - Kyong Hwa Park
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University, Seoul, 136-701, South Korea
| | - Chang-Hwan Choi
- Department of Mechanical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Sungsoo Na
- Department of Mechanical Engineering, Korea University, Seoul, 136-701, Republic of Korea.
| |
Collapse
|
53
|
Hamard C, Mignard X, Pecuchet N, Mathiot N, Blons H, Laurent-Puig P, Leroy K, Lupo A, Chapron J, Giraud F, Arrondeau J, Goldwasser F, Alifano M, Damotte D, Wislez M. [IHC, FISH, CISH, NGS in non-small cell lung cancer: What changes in the biomarker era?]. REVUE DE PNEUMOLOGIE CLINIQUE 2018; 74:327-338. [PMID: 30343945 DOI: 10.1016/j.pneumo.2018.09.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Lung cancer is the leading cause of cancer deaths in France, with about 30,000 deaths per year. The overwhelming majority (90 %) are tobacco-related. The prognosis is dark but great therapeutic advances have been made with the development of targeted therapies first and then immunotherapy afterwards. These medications are conditioned to the expression of biomarkers that require specific tools in routine to measure them. We will detail in this chapter several techniques of anatomopathology, cytogenetics and molecular biology necessary for the detection of biomarkers in lung cancers, and their applications in thoracic oncology in 2018.
Collapse
Affiliation(s)
- C Hamard
- Service de pneumologie, hôpital Cochin, Assistance publique-Hôpitaux de Paris, 27, rue du Faubourg-St-Jacques, 75014 Paris, France; Inserm UMRS1138, équipe « cancer, immune control and escape », centre de recherche des Cordeliers, Paris Descartes université, 75006 Paris, France
| | - X Mignard
- GRC n(o) 04, Theranoscan, Sorbonne université, hôpital Tenon, AP-HP, 75020 Paris, France
| | - N Pecuchet
- Inserm UMRS 1147, université Paris Descartes-Sorbonne Paris Cité, hôpital européen Georges-Pompidou, Assistance publique-Hôpitaux de Paris, 75015 Paris, France
| | - N Mathiot
- Service de pneumologie, hôpital Cochin, Assistance publique-Hôpitaux de Paris, 27, rue du Faubourg-St-Jacques, 75014 Paris, France
| | - H Blons
- Inserm UMRS 1147, université Paris Descartes-Sorbonne Paris Cité, hôpital européen Georges-Pompidou, Assistance publique-Hôpitaux de Paris, 75015 Paris, France
| | - P Laurent-Puig
- Inserm UMRS 1147, université Paris Descartes-Sorbonne Paris Cité, hôpital européen Georges-Pompidou, Assistance publique-Hôpitaux de Paris, 75015 Paris, France
| | - K Leroy
- Inserm UMRS1138, équipe « cancer, immune control and escape », centre de recherche des Cordeliers, Paris Descartes université, 75006 Paris, France
| | - A Lupo
- Inserm UMRS1138, équipe « cancer, immune control and escape », centre de recherche des Cordeliers, Paris Descartes université, 75006 Paris, France; Service d'anatomie-pathologique, hôpital Cochin, Assistance publique-Hôpitaux de Paris, 75014 Paris, France
| | - J Chapron
- Service de pneumologie, hôpital Cochin, Assistance publique-Hôpitaux de Paris, 27, rue du Faubourg-St-Jacques, 75014 Paris, France
| | - F Giraud
- Service de pneumologie, hôpital Cochin, Assistance publique-Hôpitaux de Paris, 27, rue du Faubourg-St-Jacques, 75014 Paris, France
| | - J Arrondeau
- Service d'oncologie, hôpital Cochin, Assistance publique-Hôpitaux de Paris, 75014 Paris, France
| | - F Goldwasser
- Inserm UMRS1138, équipe « cancer, immune control and escape », centre de recherche des Cordeliers, Paris Descartes université, 75006 Paris, France; Service d'oncologie, hôpital Cochin, Assistance publique-Hôpitaux de Paris, 75014 Paris, France
| | - M Alifano
- Inserm UMRS1138, équipe « cancer, immune control and escape », centre de recherche des Cordeliers, Paris Descartes université, 75006 Paris, France; Service de chirurgie thoracique, hôpital Cochin, Assistance publique-Hôpitaux de Paris, 75014 Paris, France
| | - D Damotte
- Inserm UMRS1138, équipe « cancer, immune control and escape », centre de recherche des Cordeliers, Paris Descartes université, 75006 Paris, France; Service d'anatomie-pathologique, hôpital Cochin, Assistance publique-Hôpitaux de Paris, 75014 Paris, France
| | - M Wislez
- Service de pneumologie, hôpital Cochin, Assistance publique-Hôpitaux de Paris, 27, rue du Faubourg-St-Jacques, 75014 Paris, France; Inserm UMRS1138, équipe « cancer, immune control and escape », centre de recherche des Cordeliers, Paris Descartes université, 75006 Paris, France.
| |
Collapse
|
54
|
Moore DA, Balbi K, Ingham A, Arkenau HT, Bennett P. Analysis of a large cohort of non-small cell lung cancers submitted for somatic variant analysis demonstrates that targeted next-generation sequencing is fit for purpose as a molecular diagnostic assay in routine practice. J Clin Pathol 2018; 71:1001-1006. [PMID: 30054375 DOI: 10.1136/jclinpath-2018-205319] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/04/2018] [Accepted: 07/09/2018] [Indexed: 01/07/2023]
Abstract
AIMS Targeted next-generation sequencing (tNGS) is increasingly being adopted as an alternative to single gene testing in some centres. Our aim was to assess the overall fitness and utility of tNGS as a routine clinical test in non-small cell lung cancer (NSCLC). METHODS All NSCLC cases submitted to a single laboratory for tNGS analysis over a 3-year period were included. Rejection/failure rates and turnaround times were calculated. For reportable cases, data relating to observed genetic changes likely to be driving tumour growth and/or contributing to therapeutic resistance were extracted. The impact of varied referral site practices (tissue processing and sample format submitted) on analytical outcomes was also considered. RESULTS A total of 2796 cases were submitted, of which 217 (7.8%) were rejected and 131 (5.1%) failed. The median turnaround time was seven working days. Of 2448 reported cases, KRAS, EGFR or other recognised driver mutations were observed in 35%, 17% and 5.4%, respectively. Of the remaining cases, 3.5% demonstrated significant incidental evidence of gene amplification. In 15% of EGFR-driven cases, evidence of an EGFR tyrosine kinase inhibitor resistance mechanism was observed. Potential concerns around the provision of slides or precut 'rolls' only (cf, formalin fixed paraffin embedded (FFPE) tissue blocks) as standard practice by certain referral sites were identified. CONCLUSIONS A tNGS panel approach is practically achievable, with acceptable success rates and turnaround times, in the context of a routine clinical service. Furthermore, it provides additional clinically and analytically relevant information, which is not available from single gene testing alone.
Collapse
Affiliation(s)
- David Allan Moore
- Sarah Cannon Molecular Diagnostics, London, UK .,Department of Pathology, UCL Cancer Institute, London, UK
| | - Kevin Balbi
- Sarah Cannon Molecular Diagnostics, London, UK
| | | | | | | |
Collapse
|
55
|
Naito Y, Takahashi H, Shitara K, Okamoto W, Bando H, Kuwata T, Kuboki Y, Matsumoto S, Miki I, Yamanaka T, Watanabe A, Kojima M. Feasibility study of cancer genome alterations identified by next generation sequencing: ABC study. Jpn J Clin Oncol 2018; 48:559-564. [PMID: 29659903 PMCID: PMC5974784 DOI: 10.1093/jjco/hyy052] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/05/2018] [Indexed: 01/08/2023] Open
Abstract
Background To confirm the feasibility and explore the clinical applicability of amplicon sequencing by next generation sequencing (NGS) of biopsy samples from patients with advanced solid tumors, we conducted a prospective study. Methods Patients with unresectable, advanced, or recurrent solid tumors were included. Key eligibility criteria were as follows: 20 years or older, any planned systemic therapy, adequate lesion for biopsy, and written informed consent. Samples were fixed in 10% buffered formalin and embedded in paraffin. Cancer-derived DNA was extracted, and amplicon sequencing was performed using Ion AmpliseqTM Cancer Hotspot Panel version 1.0 or version 2.0 by central vendor. We evaluated the success rate of sequencing, and the proportion of the patients with actionable mutations. We organized an expert panel to share the results of targeted sequence, make annotations and reports, and discuss concomitant ethical/legal/social issues. Results A total of 232 patients were included, and 208 were successfully analyzed (success rate of 89.7%). The biopsy procedures were safe, with only one case of Grade 3 vasovagal reaction. The proportion of actionable/druggable mutations was 38.9% (81/208), which was not significantly different between the cancer panel version 1.0 and version 2.0 (P = 0.476). Expert panel could discuss the findings and make sufficient reports. Conclusions We confirmed the feasibility of NGS-based amplicon sequencing using biopsy samples, making the basis for nationwide genome screening for cancer patients using biopsy samples. Our results suggest that focused panel may be sufficient to detect major mutations.
Collapse
Affiliation(s)
- Yoichi Naito
- National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Hideaki Takahashi
- National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Kohei Shitara
- National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Wataru Okamoto
- National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Hideaki Bando
- National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Takeshi Kuwata
- National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Yasutoshi Kuboki
- National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Shingo Matsumoto
- National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Izumi Miki
- National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | | | | | - Motohiro Kojima
- National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| |
Collapse
|
56
|
Lüsebrink J, Pieper M, Tillmann RL, Brockmann M, Schildgen O, Schildgen V. Pre-clinical validation of a next generation sequencing testing panel. Exp Mol Pathol 2018; 104:170-174. [DOI: 10.1016/j.yexmp.2018.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 03/09/2018] [Accepted: 04/02/2018] [Indexed: 12/13/2022]
|
57
|
Wu M, Pan X, Xu Y, Wu S, Wu X, Chen B. Methodological comparison of the allele refractory mutation system and direct sequencing for detecting EGFR mutations in NSCLC, and the association of EGFR mutations with patient characteristics. Oncol Lett 2018; 16:1087-1094. [PMID: 30061936 DOI: 10.3892/ol.2018.8775] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 11/02/2017] [Indexed: 12/30/2022] Open
Abstract
Gefitinib is effective for patients with non-small cell lung cancer (NSCLC) with a mutation in the epidermal growth factor receptor (EGFR) gene, which makes the detection of EGFR mutations a critical step prior to determining a treatment schedule. Therefore, the present study determined the EGFR mutation status in patients with NSCLC using an allele refractory mutation system (ARMS) and analyzed the detection ratio for different specimen types. A total of 1,596 NSCLS samples were collected and EGFR gene mutations were detected on exons 18-21 using ARMS and direct sequencing. The concordance of two methods reached 89.21%, with a total mutation rate of 45.55% (727/1,596), in which the mutation rate in lung adenocarcinoma samples was markedly increased compared with squamous cell carcinoma (51.77 vs. 8.68%). In patients with lung adenocarcinoma, EGFR mutations were more frequent in female patients than male patients (65.53 vs. 39.80%, P<0.01); there was no observable difference depending on age. Similar results were obtained for squamous cell carcinoma. In the present study, certain rare mutations were also identified; these may be subjects for further study. The impact of different sample types on the consistency between the methods was determined to be insignificant. ARMS is a more applicable approach for large-scale clinical detection than direct sequencing, and we hypothesize that ARMS may replace direct sequencing if the drawbacks of ARMS, including its narrow detection range, can be amended.
Collapse
Affiliation(s)
- Minmin Wu
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Xiaodong Pan
- Department of Transplantation Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yaya Xu
- Department of Transplantation Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Siying Wu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Xiuling Wu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Bicheng Chen
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
58
|
D'Haene N, Fontanges Q, De Nève N, Blanchard O, Melendez B, Delos M, Dehou MF, Maris C, Nagy N, Rousseau E, Vandenhove J, Gilles A, De Prez C, Verset L, Van Craynest MP, Demetter P, Van Laethem JL, Salmon I, Le Mercier M. Clinical application of targeted next-generation sequencing for colorectal cancer patients: a multicentric Belgian experience. Oncotarget 2018; 9:20761-20768. [PMID: 29755687 PMCID: PMC5945518 DOI: 10.18632/oncotarget.25099] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 03/17/2018] [Indexed: 01/14/2023] Open
Abstract
International guidelines made RAS (KRAS and NRAS) status a prerequisite for the use of anti-EGFR agents for metastatic colorectal cancer (CRC) patients. Daily, new data emerges on the theranostic and prognostic role of molecular biomarkers; this is a strong incentive for a validated, sensitive, and broadly available molecular screening test. Next-generation sequencing (NGS) has begun to supplant other technologies for genomic profiling. We report here our 2 years of clinical practice using NGS results to guide therapeutic decisions. The Ion Torrent AmpliSeq colon/lung cancer panel, which allows mutation detection in 22 cancer-related genes, was prospectively used in clinical practice (BELAC ISO 15189 accredited method). The DNA of 741 formalin-fixed paraffin-embedded CRC tissues, including primary tumors and metastasis, was obtained from 14 different Belgian institutions and subjected to targeted NGS. Of the tumors tested, 98% (727) were successfully sequenced and 89% (650) harbored at least one mutation. KRAS, BRAF and NRAS mutations were found in 335 (46%), 78 (11%) and 32 (4%) samples, respectively. These mutation frequencies were consistent with those reported in public databases. Moreover, mutations and amplifications in potentially actionable genes were identified in 464 samples (64%), including mutations in PIK3CA (14%), ERBB2 (0.4%), AKT1 (0.6%), and MAP2K1 (0.1%), as well as amplifications of ERBB2 (0.3%) and EGFR (0.3%). The median turnaround time between reception of the sample in the laboratory and report release was 8 calendar days. Overall, the AmpliSeq colon/lung cancer panel was successfully applied in daily practice and provided reliable clinically relevant information for CRC patients.
Collapse
Affiliation(s)
- Nicky D'Haene
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Quitterie Fontanges
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Nancy De Nève
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Oriane Blanchard
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Barbara Melendez
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Monique Delos
- Department of Pathology, CHU UCL Namur, Yvoir, Belgium
| | | | - Calliope Maris
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium.,Department of Pathology, Braine l´Alleud Waterloo Hospital, Braine l´Alleud, Belgium
| | - Nathalie Nagy
- Department of Pathology, Charleroi University Hospital, Charleroi, Belgium
| | | | | | - André Gilles
- Department of Pathology, EPICURA Hospital, Frameries, Belgium
| | - Carine De Prez
- Department of Pathology, Brugmann University Hospital, Brussels, Belgium
| | - Laurine Verset
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium.,CurePath, Jumet, Belgium
| | | | - Pieter Demetter
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean-Luc Van Laethem
- Department of Oncology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Isabelle Salmon
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Marie Le Mercier
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
59
|
Lindeman NI, Cagle PT, Aisner DL, Arcila ME, Beasley MB, Bernicker EH, Colasacco C, Dacic S, Hirsch FR, Kerr K, Kwiatkowski DJ, Ladanyi M, Nowak JA, Sholl L, Temple-Smolkin R, Solomon B, Souter LH, Thunnissen E, Tsao MS, Ventura CB, Wynes MW, Yatabe Y. Updated Molecular Testing Guideline for the Selection of Lung Cancer Patients for Treatment With Targeted Tyrosine Kinase Inhibitors: Guideline From the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology. Arch Pathol Lab Med 2018; 142:321-346. [PMID: 29355391 DOI: 10.5858/arpa.2017-0388-cp] [Citation(s) in RCA: 573] [Impact Index Per Article: 81.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
CONTEXT - In 2013, an evidence-based guideline was published by the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology to set standards for the molecular analysis of lung cancers to guide treatment decisions with targeted inhibitors. New evidence has prompted an evaluation of additional laboratory technologies, targetable genes, patient populations, and tumor types for testing. OBJECTIVE - To systematically review and update the 2013 guideline to affirm its validity; to assess the evidence of new genetic discoveries, technologies, and therapies; and to issue an evidence-based update. DESIGN - The College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology convened an expert panel to develop an evidence-based guideline to help define the key questions and literature search terms, review abstracts and full articles, and draft recommendations. RESULTS - Eighteen new recommendations were drafted. The panel also updated 3 recommendations from the 2013 guideline. CONCLUSIONS - The 2013 guideline was largely reaffirmed with updated recommendations to allow testing of cytology samples, require improved assay sensitivity, and recommend against the use of immunohistochemistry for EGFR testing. Key new recommendations include ROS1 testing for all adenocarcinoma patients; the inclusion of additional genes ( ERBB2, MET, BRAF, KRAS, and RET) for laboratories that perform next-generation sequencing panels; immunohistochemistry as an alternative to fluorescence in situ hybridization for ALK and/or ROS1 testing; use of 5% sensitivity assays for EGFR T790M mutations in patients with secondary resistance to EGFR inhibitors; and the use of cell-free DNA to "rule in" targetable mutations when tissue is limited or hard to obtain.
Collapse
Affiliation(s)
- Neal I Lindeman
- From the Departments of Pathology (Drs Lindeman and Sholl) and Medicine (Dr Kwiatkowski), Brigham and Women's Hospital, Boston, Massachusetts; the Cancer Center (Dr Bernicker) and the Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas (Dr Cagle); the Department of Pathology, University of Colorado School of Medicine, Denver (Dr Aisner); the Diagnostic and Molecular Pathology Laboratory (Dr Arcila) and the Molecular Diagnostics Service (Dr Ladanyi), Memorial Sloan Kettering Cancer Center, New York, New York; the Department of Pathology & Medicine, Pulmonary, Critical Care and Sleep Medicine, New York, New York (Dr Beasley); the Pathology and Laboratory Quality Center, College of American Pathologists, Northfield, Illinois (Mss Colasacco and Ventura); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Medicine and Pathology, University of Colorado, Denver (Dr Hirsch); the Department of Pathology, University of Aberdeen, Aberdeen, Scotland (Dr Kerr); the Department of Molecular Pathology, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Clinical and Scientific Affairs Division, Association for Molecular Pathology, Bethesda, Maryland (Dr Temple-Smolkin); the Molecular Therapeutics and Biomarkers Laboratory, Peter Maccallum Cancer Center, Melbourne, Australia (Dr Solomon); the Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands (Dr Thunnissen); the Department of Laboratory Medicine and Pathobiology, Princess Margaret Cancer Center, Toronto, Ontario, Canada (Dr Tsao); Scientific Affairs, International Association for the Study of Lung Cancer, Aurora, Colorado (Dr Wynes); and the Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan (Dr Yatabe). Dr Souter is in private practice in Wellanport, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Lindeman NI, Cagle PT, Aisner DL, Arcila ME, Beasley MB, Bernicker EH, Colasacco C, Dacic S, Hirsch FR, Kerr K, Kwiatkowski DJ, Ladanyi M, Nowak JA, Sholl L, Temple-Smolkin R, Solomon B, Souter LH, Thunnissen E, Tsao MS, Ventura CB, Wynes MW, Yatabe Y. Updated Molecular Testing Guideline for the Selection of Lung Cancer Patients for Treatment With Targeted Tyrosine Kinase Inhibitors: Guideline From the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology. J Thorac Oncol 2018; 13:323-358. [PMID: 29396253 DOI: 10.1016/j.jtho.2017.12.001] [Citation(s) in RCA: 357] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2017] [Indexed: 12/15/2022]
Abstract
CONTEXT In 2013, an evidence-based guideline was published by the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology to set standards for the molecular analysis of lung cancers to guide treatment decisions with targeted inhibitors. New evidence has prompted an evaluation of additional laboratory technologies, targetable genes, patient populations, and tumor types for testing. OBJECTIVE To systematically review and update the 2013 guideline to affirm its validity; to assess the evidence of new genetic discoveries, technologies, and therapies; and to issue an evidence-based update. DESIGN The College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology convened an expert panel to develop an evidence-based guideline to help define the key questions and literature search terms, review abstracts and full articles, and draft recommendations. RESULTS Eighteen new recommendations were drafted. The panel also updated 3 recommendations from the 2013 guideline. CONCLUSIONS The 2013 guideline was largely reaffirmed with updated recommendations to allow testing of cytology samples, require improved assay sensitivity, and recommend against the use of immunohistochemistry for EGFR testing. Key new recommendations include ROS1 testing for all adenocarcinoma patients; the inclusion of additional genes (ERBB2, MET, BRAF, KRAS, and RET) for laboratories that perform next-generation sequencing panels; immunohistochemistry as an alternative to fluorescence in situ hybridization for ALK and/or ROS1 testing; use of 5% sensitivity assays for EGFR T790M mutations in patients with secondary resistance to EGFR inhibitors; and the use of cell-free DNA to "rule in" targetable mutations when tissue is limited or hard to obtain.
Collapse
Affiliation(s)
- Neal I Lindeman
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts.
| | - Philip T Cagle
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas
| | - Dara L Aisner
- Department of Pathology, University of Colorado School of Medicine, Denver, New York
| | - Maria E Arcila
- Diagnostic and Molecular Pathology Laboratory, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mary Beth Beasley
- Department of Pathology & Medicine, Pulmonary, Critical Care and Sleep Medicine, New York, New York
| | | | - Carol Colasacco
- Pathology and Laboratory Quality Center, College of American Pathologists, Northfield, Illinois
| | - Sanja Dacic
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Fred R Hirsch
- Department of Medicine and Pathology, University of Colorado, Denver, New York
| | - Keith Kerr
- Department of Pathology, University of Aberdeen, Aberdeen, Scotland
| | | | - Marc Ladanyi
- Molecular Diagnostics Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jan A Nowak
- Department of Molecular Pathology, Roswell Park Cancer Institute, Buffalo, New York
| | - Lynette Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Robyn Temple-Smolkin
- Clinical and Scientific Affairs Division, Association for Molecular Pathology, Bethesda, Maryland
| | - Benjamin Solomon
- Molecular Therapeutics and Biomarkers Laboratory, Peter Maccallum Cancer Center, Melbourne, Australia
| | | | - Erik Thunnissen
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | - Ming S Tsao
- Department of Laboratory Medicine and Pathobiology, Princess Margaret Cancer Center, Toronto, Ontario, Canada
| | - Christina B Ventura
- Pathology and Laboratory Quality Center, College of American Pathologists, Northfield, Illinois
| | - Murry W Wynes
- Scientific Affairs, International Association for the Study of Lung Cancer, Aurora, Colorado
| | - Yasushi Yatabe
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| |
Collapse
|
61
|
Lindeman NI, Cagle PT, Aisner DL, Arcila ME, Beasley MB, Bernicker EH, Colasacco C, Dacic S, Hirsch FR, Kerr K, Kwiatkowski DJ, Ladanyi M, Nowak JA, Sholl L, Temple-Smolkin R, Solomon B, Souter LH, Thunnissen E, Tsao MS, Ventura CB, Wynes MW, Yatabe Y. Updated Molecular Testing Guideline for the Selection of Lung Cancer Patients for Treatment With Targeted Tyrosine Kinase Inhibitors: Guideline From the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology. J Mol Diagn 2018; 20:129-159. [PMID: 29398453 DOI: 10.1016/j.jmoldx.2017.11.004] [Citation(s) in RCA: 248] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2017] [Indexed: 02/07/2023] Open
Abstract
CONTEXT In 2013, an evidence-based guideline was published by the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology to set standards for the molecular analysis of lung cancers to guide treatment decisions with targeted inhibitors. New evidence has prompted an evaluation of additional laboratory technologies, targetable genes, patient populations, and tumor types for testing. OBJECTIVE To systematically review and update the 2013 guideline to affirm its validity; to assess the evidence of new genetic discoveries, technologies, and therapies; and to issue an evidence-based update. DESIGN The College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology convened an expert panel to develop an evidence-based guideline to help define the key questions and literature search terms, review abstracts and full articles, and draft recommendations. RESULTS Eighteen new recommendations were drafted. The panel also updated 3 recommendations from the 2013 guideline. CONCLUSIONS The 2013 guideline was largely reaffirmed with updated recommendations to allow testing of cytology samples, require improved assay sensitivity, and recommend against the use of immunohistochemistry for EGFR testing. Key new recommendations include ROS1 testing for all adenocarcinoma patients; the inclusion of additional genes (ERBB2, MET, BRAF, KRAS, and RET) for laboratories that perform next-generation sequencing panels; immunohistochemistry as an alternative to fluorescence in situ hybridization for ALK and/or ROS1 testing; use of 5% sensitivity assays for EGFR T790M mutations in patients with secondary resistance to EGFR inhibitors; and the use of cell-free DNA to "rule in" targetable mutations when tissue is limited or hard to obtain.
Collapse
Affiliation(s)
- Neal I Lindeman
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts.
| | - Philip T Cagle
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas
| | - Dara L Aisner
- Department of Pathology, University of Colorado School of Medicine, Denver, Colorado
| | - Maria E Arcila
- Diagnostic and Molecular Pathology Laboratory, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mary Beth Beasley
- Department of Pathology & Medicine, Pulmonary, Critical Care and Sleep Medicine, New York, New York
| | - Eric H Bernicker
- Cancer Research Program, Houston Methodist Research Institute, Houston, Texas
| | - Carol Colasacco
- Pathology and Laboratory Quality Center, College of American Pathologists, Northfield, Illinois
| | - Sanja Dacic
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Fred R Hirsch
- Department of Medicine and Pathology, University of Colorado, Denver, Colorado
| | - Keith Kerr
- Department of Pathology, University of Aberdeen, Aberdeen, Scotland
| | | | - Marc Ladanyi
- Molecular Diagnostics Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jan A Nowak
- Department of Molecular Pathology, Roswell Park Cancer Institute, Buffalo, New York
| | - Lynette Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Robyn Temple-Smolkin
- Clinical and Scientific Affairs Division, Association for Molecular Pathology, Bethesda, Maryland
| | - Benjamin Solomon
- Molecular Therapeutics and Biomarkers Laboratory, Peter Maccallum Cancer Center, Melbourne, Australia
| | | | - Erik Thunnissen
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Ming S Tsao
- Department of Laboratory Medicine and Pathobiology, Princess Margaret Cancer Center, Toronto, Ontario, Canada
| | - Christina B Ventura
- Pathology and Laboratory Quality Center, College of American Pathologists, Northfield, Illinois
| | - Murry W Wynes
- Scientific Affairs, International Association for the Study of Lung Cancer, Aurora, Colorado
| | - Yasushi Yatabe
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| |
Collapse
|
62
|
Bennett CW, Berchem G, Kim YJ, El-Khoury V. Cell-free DNA and next-generation sequencing in the service of personalized medicine for lung cancer. Oncotarget 2018; 7:71013-71035. [PMID: 27589834 PMCID: PMC5342606 DOI: 10.18632/oncotarget.11717] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 08/11/2016] [Indexed: 12/13/2022] Open
Abstract
Personalized medicine has emerged as the future of cancer care to ensure that patients receive individualized treatment specific to their needs. In order to provide such care, molecular techniques that enable oncologists to diagnose, treat, and monitor tumors are necessary. In the field of lung cancer, cell free DNA (cfDNA) shows great potential as a less invasive liquid biopsy technique, and next-generation sequencing (NGS) is a promising tool for analysis of tumor mutations. In this review, we outline the evolution of cfDNA and NGS and discuss the progress of using them in a clinical setting for patients with lung cancer. We also present an analysis of the role of cfDNA as a liquid biopsy technique and NGS as an analytical tool in studying EGFR and MET, two frequently mutated genes in lung cancer. Ultimately, we hope that using cfDNA and NGS for cancer diagnosis and treatment will become standard for patients with lung cancer and across the field of oncology.
Collapse
Affiliation(s)
- Catherine W Bennett
- Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg
| | - Guy Berchem
- Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg.,Centre Hospitalier de Luxembourg, L-1210 Luxembourg, Luxembourg
| | - Yeoun Jin Kim
- Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg
| | - Victoria El-Khoury
- Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg
| |
Collapse
|
63
|
Tang JH, Chia D. Liquid Biopsies in the Screening of Oncogenic Mutations in NSCLC and its Application in Targeted Therapy. Crit Rev Oncog 2018; 20:357-71. [PMID: 27279235 DOI: 10.1615/critrevoncog.v20.i5-6.90] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Non-small cell lung cancer (NSCLC) still dominates cancer-related deaths in America. Despite this, new discoveries and advancements in technology are helping with the detection and treatment of NSCLC. The discovery of circulating tumor DNA in blood and other biofluids is essential for the creation of a DNA biomarker. Limitations in technology and sequencing have stunted assay development, but with recent advancements in the next-generation sequencing, droplet digital PCR, and EFIRM, the detection of mutations in biofluids has become possible with reasonable sensitivity and specificity. These methods have been applied to the detection of mutations in NSCLC by measuring the levels of circulating tumor DNA. ALK fusion genes along with mutations in EGFR and KRAS have been shown to correlate to tumor size and metastasis. These methods allow for noninvasive, affordable, and efficient diagnoses of oncogenic mutations that overcome the issues of traditional biopsies. These issues include tumor heterogeneity and early detection of cancers with asymptomatic early stages. Early detection and treatment remain the best way to ensure survival. This review aims to describe these new technologies along with their application in mutation detection in NSCLC in order to proactively utilize targeted anticancer therapy.
Collapse
Affiliation(s)
- Jason H Tang
- Department of Pathology, UCLA David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - David Chia
- Department of Pathology, UCLA David Geffen School of Medicine at UCLA, Los Angeles, CA
| |
Collapse
|
64
|
Kotelnikova EA, Pyatnitskiy M, Paleeva A, Kremenetskaya O, Vinogradov D. Practical aspects of NGS-based pathways analysis for personalized cancer science and medicine. Oncotarget 2018; 7:52493-52516. [PMID: 27191992 PMCID: PMC5239569 DOI: 10.18632/oncotarget.9370] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 04/18/2016] [Indexed: 12/17/2022] Open
Abstract
Nowadays, the personalized approach to health care and cancer care in particular is becoming more and more popular and is taking an important place in the translational medicine paradigm. In some cases, detection of the patient-specific individual mutations that point to a targeted therapy has already become a routine practice for clinical oncologists. Wider panels of genetic markers are also on the market which cover a greater number of possible oncogenes including those with lower reliability of resulting medical conclusions. In light of the large availability of high-throughput technologies, it is very tempting to use complete patient-specific New Generation Sequencing (NGS) or other "omics" data for cancer treatment guidance. However, there are still no gold standard methods and protocols to evaluate them. Here we will discuss the clinical utility of each of the data types and describe a systems biology approach adapted for single patient measurements. We will try to summarize the current state of the field focusing on the clinically relevant case-studies and practical aspects of data processing.
Collapse
Affiliation(s)
- Ekaterina A Kotelnikova
- Personal Biomedicine, Moscow, Russia.,A. A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia.,Institute Biomedical Research August Pi Sunyer (IDIBAPS), Hospital Clinic of Barcelona, Barcelona, Spain
| | - Mikhail Pyatnitskiy
- Personal Biomedicine, Moscow, Russia.,Orekhovich Institute of Biomedical Chemistry, Moscow, Russia.,Pirogov Russian National Research Medical University, Moscow, Russia
| | | | - Olga Kremenetskaya
- Personal Biomedicine, Moscow, Russia.,Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Moscow, Russia
| | - Dmitriy Vinogradov
- Personal Biomedicine, Moscow, Russia.,A. A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia.,Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
65
|
Shi Y, Zhang W, Ye Y, Cheng Y, Han L, Liu P, Zhao W, Tong Z, Yu J. Benefit of everolimus as a monotherapy for a refractory breast cancer patient bearing multiple genetic mutations in the PI3K/AKT/mTOR signaling pathway. Cancer Biol Med 2018; 15:314-321. [PMID: 30197799 PMCID: PMC6121052 DOI: 10.20892/j.issn.2095-3941.2017.0188] [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] [Indexed: 12/16/2022] Open
Abstract
A postmenopausal patient with a diagnosis of estrogen receptor (ER) (+), progesterone receptor (PR) (+), and human epidermal growth factor receptor-2 (HER2) (-) breast cancer was reported. The patient refused surgery and was resistant to conventional chemotherapy regimens. Computed tomography and the circulating tumor cell test indicated that the patient’s tumor burden increased rapidly even after several chemotherapy sessions. Multiple genetic aberrances in the phosphatidylinositol3-kinases (PI3K) signaling pathway were detected via next-generation sequencing (NGS)-based liquid biopsy, including a p. G1007R missense mutation in exon 21 of PIK3CA (33.61%), a p.L70fs frameshift mutation in exon 3 of phosphatase and tension homolog deleted on chromosome ten (PTEN) (49.14%), and a p. D1542Y missense mutation in exon 32 of mammalian target of rapamycin (mTOR) (1.66%). Therefore, only the mTOR inhibitor everolimus was administered to the patient. Partial remission (PR) was observed after 2 months, and sustained stable disease (SD) was observed after a year and a half. Subsequent sequencing showed that the mutation ratio of PIK3CA decreased to 4.17%, and that the PTEN and mTOR mutations disappeared, which revealed the significant curative effect of everolimus. We report the first case of successful monotherapy treatment using everolimus in a patient with advanced breast cancer bearing mutations in genes involved in the PI3K/ARK/mTOR signaling pathway. The success of this case highlights the invaluable clinical contribution of NGS-based liquid biopsy, as it successfully provided an optimal therapeutic target for the patient with advanced breast cancer.
Collapse
Affiliation(s)
- Yehui Shi
- Medical Oncology Department of Breast Cancer
| | - Wenwen Zhang
- Cancer Molecular Diagnostics Core.,Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center forCancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin300060, China
| | | | | | - Lei Han
- Cancer Molecular Diagnostics Core
| | | | | | | | - Jinpu Yu
- Cancer Molecular Diagnostics Core.,Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center forCancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin300060, China
| |
Collapse
|
66
|
Youssef O, Knuuttila A, Piirilä P, Böhling T, Sarhadi V, Knuutila S. Presence of cancer-associated mutations in exhaled breath condensates of healthy individuals by next generation sequencing. Oncotarget 2017; 8:18166-18176. [PMID: 28199989 PMCID: PMC5392316 DOI: 10.18632/oncotarget.15233] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 01/16/2017] [Indexed: 01/29/2023] Open
Abstract
Exhaled breath condensate (EBC) is a non-invasive source that can be used for studying different genetic alterations occurring in lung tissue. However, the low yield of DNA available from EBC has hampered the more detailed mutation analysis by conventional methods. We applied the more sensitive amplicon-based next generation sequencing (NGS) to identify cancer related mutations in DNA isolated from EBC. In order to apply any method for the purpose of mutation screening in cancer patients, it is important to clarify the incidence of these mutations in healthy individuals. Therefore, we studied mutations in hotspot regions of 22 cancer genes of 20 healthy, mainly non-smoker individuals, using AmpliSeq colon and lung cancer panel and sequenced on Ion PGM. In 15 individuals, we detected 35 missense mutations in TP53, KRAS, NRAS, SMAD4, MET, CTNNB1, PTEN, BRAF, DDR2, EGFR, PIK3CA, NOTCH1, FBXW7, FGFR3, and ERBB2: these have been earlier reported in different tumor tissues. Additionally, 106 novel mutations not reported previously were also detected. One healthy non-smoker subject had a KRAS G12D mutation in EBC DNA. Our results demonstrate that DNA from EBC of healthy subjects can reveal mutations that could represent very early neoplastic changes or alternatively a normal process of apoptosis eliminating damaged cells with mutations or altered genetic material. Further assessment is needed to determine if NGS analysis of EBC could be a screening method for high risk individuals such as smokers, where it could be applied in the early diagnosis of lung cancer and monitoring treatment efficacy.
Collapse
Affiliation(s)
- Omar Youssef
- Faculty of Medicine, Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Aija Knuuttila
- Department of Pulmonary Medicine, University of Helsinki and Helsinki University Hospital, Heart and Lung Center, Helsinki, Finland
| | - Päivi Piirilä
- Unit of Clinical Physiology, HUS-Medical Imaging Center, Helsinki University Hospital and Helsinki University, Helsinki, Finland
| | - Tom Böhling
- Department of Pathology, University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Virinder Sarhadi
- Faculty of Medicine, Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Sakari Knuutila
- Faculty of Medicine, Department of Pathology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
67
|
Darwanto A, Hein AM, Strauss S, Kong Y, Sheridan A, Richards D, Lader E, Ngowe M, Pelletier T, Adams D, Ricker A, Patel N, Kühne A, Hughes S, Shiffman D, Zimmermann D, Te Kaat K, Rothmann T. Use of the QIAGEN GeneReader NGS system for detection of KRAS mutations, validated by the QIAGEN Therascreen PCR kit and alternative NGS platform. BMC Cancer 2017; 17:358. [PMID: 28532404 PMCID: PMC5441096 DOI: 10.1186/s12885-017-3328-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 05/05/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The detection of somatic mutations in primary tumors is critical for the understanding of cancer evolution and targeting therapy. Multiple technologies have been developed to enable the detection of such mutations. Next generation sequencing (NGS) is a new platform that is gradually becoming the technology of choice for genotyping cancer samples, owing to its ability to simultaneously interrogate many genomic loci at massively high efficiency and increasingly lower cost. However, multiple barriers still exist for its broader adoption in clinical research practice, such as fragmented workflow and complex bioinformatics analysis and interpretation. METHODS We performed validation of the QIAGEN GeneReader NGS System using the QIAact Actionable Insights Tumor Panel, focusing on clinically meaningful mutations by using DNA extracted from formalin-fixed paraffin-embedded (FFPE) colorectal tissue with known KRAS mutations. The performance of the GeneReader was evaluated and compared to data generated from alternative technologies (PCR and pyrosequencing) as well as an alternative NGS platform. The results were further confirmed with Sanger sequencing. RESULTS The data generated from the GeneReader achieved 100% concordance with reference technologies. Furthermore, the GeneReader workflow provides a truly integrated workflow, eliminating artifacts resulting from routine sample preparation; and providing up-to-date interpretation of test results. CONCLUSION The GeneReader NGS system offers an effective and efficient method to identify somatic (KRAS) cancer mutations.
Collapse
Affiliation(s)
- Agus Darwanto
- QIAGEN Waltham, 35 Gatehouse Dr, Waltham, MA, 02451, USA.,Novartis Institutes for BioMedical Research, Cambridge, MA, 02139, USA
| | | | - Sascha Strauss
- QIAGEN GmbH, QIAGEN Strasse 1, 40724, Hilden, Nordrhein-Westfalen, Germany
| | - Yi Kong
- QIAGEN Redwood City, 1700 Seaport Blvd, Redwood, CA, 94063, USA
| | | | - Dan Richards
- QIAGEN Redwood City, 1700 Seaport Blvd, Redwood, CA, 94063, USA
| | - Eric Lader
- QIAGEN Frederick, 6951 Executive Way, Frederick, MD, 21703, USA
| | - Monika Ngowe
- QIAGEN Waltham, 35 Gatehouse Dr, Waltham, MA, 02451, USA.,T2 Biosystems, Lexington, MA, 02421, USA
| | | | - Danielle Adams
- QIAGEN Waltham, 35 Gatehouse Dr, Waltham, MA, 02451, USA.,Macherey-Nigel, Bethlehem, PA, 18020, USA
| | - Austin Ricker
- QIAGEN Waltham, 35 Gatehouse Dr, Waltham, MA, 02451, USA
| | - Nishit Patel
- QIAGEN Waltham, 35 Gatehouse Dr, Waltham, MA, 02451, USA
| | - Andreas Kühne
- QIAGEN GmbH, QIAGEN Strasse 1, 40724, Hilden, Nordrhein-Westfalen, Germany
| | - Simon Hughes
- QIAGEN Manchester, Skelton House Lloyd Street North, Manchester, M15 6SH, UK
| | - Dan Shiffman
- QIAGEN Redwood City, 1700 Seaport Blvd, Redwood, CA, 94063, USA
| | - Dirk Zimmermann
- QIAGEN GmbH, QIAGEN Strasse 1, 40724, Hilden, Nordrhein-Westfalen, Germany
| | - Kai Te Kaat
- QIAGEN GmbH, QIAGEN Strasse 1, 40724, Hilden, Nordrhein-Westfalen, Germany
| | - Thomas Rothmann
- QIAGEN GmbH, QIAGEN Strasse 1, 40724, Hilden, Nordrhein-Westfalen, Germany.
| |
Collapse
|
68
|
DiBardino DM, Rawson DW, Saqi A, Heymann JJ, Pagan CA, Bulman WA. Next-generation sequencing of non-small cell lung cancer using a customized, targeted sequencing panel: Emphasis on small biopsy and cytology. Cytojournal 2017; 14:7. [PMID: 28413430 PMCID: PMC5379858 DOI: 10.4103/1742-6413.202602] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 09/08/2016] [Indexed: 02/02/2023] Open
Abstract
Background: Next-generation sequencing (NGS) with a multi-gene panel is now available for patients with lung adenocarcinoma, but the performance characteristics and clinical utility of this testing are not well-described. We present the results of an extended 467 gene panel in a series of advanced, highly selected nonsmall cell lung cancer (NSCLC) patients using a range of specimens, including predominantly small biopsy and cytology specimens. Materials and Methods: A retrospective review of 22 NSCLC biopsies sent for NGS using an extended gene panel from January 2014 to July 2015. The customized NGS panel sequences 467 cancer-associated genes with exonic and intronic sequences obtained from purified tumor DNA. Genomic alterations, patient characteristics, and success of testing were determined. Results: The majority of samples tested were metastatic lung adenocarcinoma on final pathology. Of the 22 specimens tested, 5 (22.7%) were surgical resections and 17 (77.3%) were small biopsy and cytology specimens. Twenty-one (95%) of the specimens were adequate for full sequencing and yielded a total of 204 genomic alterations (average 8.9 per tumor), of which 17 (average 0.81 per tumor) were actionable and/or clinically relevant. Genomic alterations were found most commonly in the TP53, EGFR, EPHB1, MLL3, APC, SETD2, KRAS, DNMT3A, RB1, CDKN2A, ARID1A, EP300, KDM6B, RAD50, STK11, and BRCA2 genes. Conclusions: NGS using a comprehensive gene panel was performed successfully in 95% of all NSCLC cases in this series, including 94% small biopsy and cytology specimens and 100% surgical resections. This custom assay was performed on a range of tumor specimens and demonstrates that small specimens are able to provide a similar depth of information as larger ones. As many patients present at an advanced stage and only small specimens are obtained, the information these provide has the potential for guiding treatment in highly selected patients with advanced lung adenocarcinoma.
Collapse
Affiliation(s)
- David M DiBardino
- Address: Division of Pulmonology, Allergy, Immunology and Critical Care, Section of Interventional Pulmonology, University of Pennsylvania, Philadelphia, PA, USA
| | - David W Rawson
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Anjali Saqi
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Jonas J Heymann
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Carlos A Pagan
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - William A Bulman
- Division of Pulmonary, Allergy and Critical Care Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| |
Collapse
|
69
|
Asor E, Stav MY, Simon E, Fahoum I, Sabo E, Ben-Izhak O, Hershkovitz D. Risk for molecular contamination of tissue samples evaluated for targeted anti-cancer therapy. PLoS One 2017; 12:e0173760. [PMID: 28288198 PMCID: PMC5348008 DOI: 10.1371/journal.pone.0173760] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 02/27/2017] [Indexed: 01/26/2023] Open
Abstract
With the increasing usage of sensitive PCR technology for pharmacogenetics, cross contamination becomes a significant concern. Researchers employed techniques which basically include replacing laboratory equipment after each sample preparation; however, there are no recommended guidelines. In the present work we wanted to evaluate the risk of cross contamination during tissue processing using the routine precaution measures. Twenty-one surgical samples of lung adenocarcinoma were used, of which 7 contained EGFR exon 19 mutation, 7 contained EGFR exon 21 mutation (p.L858R) and 7 were EGFR wild-type. The samples were ordered by alternating the mutation group to maximize the potential for cross contamination and underwent tissue sectioning and de-paraffinization. The entire process was performed using the same tools. Following DNA extraction all samples underwent PCR amplification and were scrutinized for small fractions of EGFR mutation using deep sequencing with the Ion torrent PGM technology. Twenty samples yielded results. The fraction of mutated copies was 41 ± 23% (range 11–66) for the cases with known exon 19 mutation and 48±24% (range 0–65) for the cases with known exon 21 mutations. No in-frame exon 19 deletion mutations were identified in the wild-type (WT) and exon 21 groups. The fraction of EGFR exon 21 (codon 858) mutations was 0.018±0.014% (range 0–0.05%) in the WT and exon 19 groups, which was not statistically different than the background sequencing artifact noise for the same base-pair alteration (p = 0.21). Our results suggest that standard precautions are sufficient for molecular pathology diagnosis of surgical samples and are not associated with increased risk of cross contamination.
Collapse
Affiliation(s)
- Eyal Asor
- B. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Institute of Pathology, Rambam Health Care Campus, Haifa, Israel
| | - Michael Y. Stav
- Institute of Pathology, Rambam Health Care Campus, Haifa, Israel
| | - Einav Simon
- B. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ibrahim Fahoum
- Institute of Pathology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Edmond Sabo
- B. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Institute of Pathology, Rambam Health Care Campus, Haifa, Israel
| | - Ofer Ben-Izhak
- B. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Institute of Pathology, Rambam Health Care Campus, Haifa, Israel
| | - Dov Hershkovitz
- Institute of Pathology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- * E-mail:
| |
Collapse
|
70
|
Lu YQ, Lu KH. Advancements in next-generation sequencing for diagnosis and treatment of non-small-cell lung cancer. Chronic Dis Transl Med 2017; 3:1-7. [PMID: 29063051 PMCID: PMC5627693 DOI: 10.1016/j.cdtm.2017.02.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Indexed: 01/01/2023] Open
Affiliation(s)
- Ying-Qiang Lu
- Department of Clinical Medicine, The First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Kai-Hua Lu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
71
|
Brait M, Izumchenko E, Kagohara LT, Long S, Wysocki PT, Faherty B, Fertig EJ, Khor TO, Bruckheimer E, Baia G, Ciznadija D, Sloma I, Ben-Zvi I, Paz K, Sidransky D. Comparative mutational landscape analysis of patient-derived tumour xenografts. Br J Cancer 2017; 116:515-523. [PMID: 28118322 PMCID: PMC5318980 DOI: 10.1038/bjc.2016.450] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 11/23/2016] [Accepted: 12/14/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Screening of patients for cancer-driving mutations is now used for cancer prognosis, remission scoring and treatment selection. Although recently emerged targeted next-generation sequencing-based approaches offer promising diagnostic capabilities, there are still limitations. There is a pressing clinical need for a well-validated, rapid, cost-effective mutation profiling system in patient specimens. Given their speed and cost-effectiveness, quantitative PCR mutation detection techniques are well suited for the clinical environment. The qBiomarker mutation PCR array has high sensitivity and shorter turnaround times compared with other methods. However, a direct comparison with existing viable alternatives are required to assess its true potential and limitations. METHODS In this study, we evaluated a panel of 117 patient-derived tumour xenografts by the qBiomarker array and compared with other methods for mutation detection, including Ion AmpliSeq sequencing, whole-exome sequencing and droplet digital PCR. RESULTS Our broad analysis demonstrates that the qBiomarker's performance is on par with that of other labour-intensive and expensive methods of cancer mutation detection of frequently altered cancer-associated genes, and provides a foundation for supporting its consideration as an option for molecular diagnostics. CONCLUSIONS This large-scale direct comparison and validation of currently available mutation detection approaches is extremely relevant for the current scenario of precision medicine and will lead to informed choice of screening methodologies, especially in lower budget conditions or time frame limitations.
Collapse
Affiliation(s)
- Mariana Brait
- Department of Otolaryngology and Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Evgeny Izumchenko
- Department of Otolaryngology and Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Luciane T Kagohara
- Department of Otolaryngology and Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Samuel Long
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Piotr T Wysocki
- Department of Otolaryngology and Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Brian Faherty
- Department of Otolaryngology and Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Elana J Fertig
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Tin Oo Khor
- Champions Oncology, Baltimore, MD 21205, USA
| | | | - Gilson Baia
- Champions Oncology, Baltimore, MD 21205, USA
| | | | - Ido Sloma
- Champions Oncology, Baltimore, MD 21205, USA
| | - Ido Ben-Zvi
- Champions Oncology, Baltimore, MD 21205, USA
| | - Keren Paz
- Champions Oncology, Baltimore, MD 21205, USA
| | - David Sidransky
- Department of Otolaryngology and Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
72
|
Jang K, Choi J, Park C, Na S. Label-free and high-sensitive detection of Kirsten rat sarcoma viral oncogene homolog and epidermal growth factor receptor mutation using Kelvin probe force microscopy. Biosens Bioelectron 2017; 87:222-228. [DOI: 10.1016/j.bios.2016.08.045] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 08/05/2016] [Accepted: 08/15/2016] [Indexed: 12/21/2022]
|
73
|
Mäki-Nevala S, Sarhadi VK, Knuuttila A, Scheinin I, Ellonen P, Lagström S, Rönty M, Kettunen E, Husgafvel-Pursiainen K, Wolff H, Knuutila S. Driver Gene and Novel Mutations in Asbestos-Exposed Lung Adenocarcinoma and Malignant Mesothelioma Detected by Exome Sequencing. Lung 2016; 194:125-35. [PMID: 26463840 DOI: 10.1007/s00408-015-9814-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 09/27/2015] [Indexed: 12/30/2022]
Abstract
BACKGROUND Asbestos is a carcinogen linked to malignant mesothelioma (MM) and lung cancer. Some gene aberrations related to asbestos exposure are recognized, but many associated mutations remain obscure. We performed exome sequencing to determine the association of previously known mutations (driver gene mutations) with asbestos and to identify novel mutations related to asbestos exposure in lung adenocarcinoma (LAC) and MM. METHODS Exome sequencing was performed on DNA from 47 tumor tissues of MM (21) and LAC (26) patients, 27 of whom had been asbestos-exposed (18 MM, 9 LAC). In addition, 9 normal lung/blood samples of LAC were sequenced. Novel mutations identified from exome data were validated by amplicon-based deep sequencing. Driver gene mutations in BRAF, EGFR, ERBB2, HRAS, KRAS, MET, NRAS, PIK3CA, STK11, and ephrin receptor genes (EPHA1-8, 10 and EPHB1-4, 6) were studied for both LAC and MM, and in BAP1, CUL1, CDKN2A, and NF2 for MM. RESULTS In asbestos-exposed MM patients, previously non-described NF2 frameshift mutation (one) and BAP1 mutations (four) were detected. Exome data mining revealed some genes potentially associated with asbestos exposure, such as MRPL1 and SDK1. BAP1 and COPG1 mutations were seen exclusively in MM. Pathogenic KRAS mutations were common in LAC patients (42 %), both in non-exposed (n = 5) and exposed patients (n = 6). Pathogenic BRAF mutations were found in two LACs. CONCLUSION BAP1 mutations occurred in asbestos-exposed MM. MRPL1, SDK1, SEMA5B, and INPP4A could possibly serve as candidate genes for alterations associated with asbestos exposure. KRAS mutations in LAC were not associated with asbestos exposure.
Collapse
|
74
|
Asaka S, Yoshizawa A, Matsuda K, Yamaguchi A, Yamamoto H, Shiina T, Nakata R, Ogawa K, Zhang M, Honda T. A novel, rapid point-of-care test for lung cancer patients to detect epidermal growth factor receptor gene mutations by using real-time droplet-PCR and fresh liquid cytology specimens. Oncol Rep 2016; 37:1020-1026. [PMID: 27922678 DOI: 10.3892/or.2016.5287] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 07/08/2016] [Indexed: 11/06/2022] Open
Abstract
Epidermal growth factor receptor gene (EGFR) mutations are associated with response to tyrosine kinase inhibitors (TKIs) in patients with non-small cell lung cancer (NSCLC). We developed a novel, rapid EGFR mutation assay using a real-time droplet-polymerase chain reaction machine (EGFR d-PCR assay). The purpose of this study was to validate the performance of the EGFR d-PCR assay using fresh liquid cytology specimens. We analyzed three major EGFR mutations (L858R in exon 21, E746_A750del in exon 19 and T790M in exon 20) in 80 fresh liquid cytology specimens of adenocarcinoma (ADC) or NSCLC-not otherwise specified (NOS) via the EGFR d-PCR assay and conventional real-time PCR assay using the therascreen® EGFR RGQ PCR kit (Therascreen assay). In addition, we performed sensitivity assays using cell lines with EGFR mutations. The EGFR d-PCR assay detected 16 L858Rs, 8 E746_A750dels and 1 T790M mutation and the Therascreen assay detected 16 L858Rs, 11 deletions in exon 19 and 1 T790M mutation. The results were concordant between the two assays. The reaction time of the EGFR d-PCR assay was 8 min and 10 sec, but that of the Therascreen assay was 1 h and 45 min. Sensitivity, as assessed by the detection limit of the EGFR d-PCR assay was 0.5, 0.05 and 0.5% for L858R, E746_A750del and T790M, respectively. The EGFR d-PCR assay markedly reduced the detection time of major EGFR mutations with high sensitivity compared with the conventional Therascreen assay and is expected to expedite EGFR-TKI therapy for lung cancer patients, especially those in advanced stages.
Collapse
Affiliation(s)
- Shiho Asaka
- Department of Laboratory Medicine, Shinshu University Hospital, Matsumoto, Nagano 390-8621, Japan
| | - Akihiko Yoshizawa
- Department of Laboratory Medicine, Shinshu University Hospital, Matsumoto, Nagano 390-8621, Japan
| | - Kazuyuki Matsuda
- Department of Laboratory Medicine, Shinshu University Hospital, Matsumoto, Nagano 390-8621, Japan
| | - Akemi Yamaguchi
- Graduate School of Science and Technology, Shinshu University, Wakasato, Nagano 380-8553, Japan
| | - Hiroshi Yamamoto
- First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Nagano 390-8621, Japan
| | - Takayuki Shiina
- Department of Thoracic Surgery, Shinshu University Hospital, Matsumoto, Nagano 390-8621, Japan
| | - Rie Nakata
- Department of Laboratory Medicine, Shinshu University Hospital, Matsumoto, Nagano 390-8621, Japan
| | - Kaoru Ogawa
- Shinshu University School of Medicine, Matsumoto, Nagano 390-8621, Japan
| | - Meng Zhang
- Department of Thoracic Surgery, Shinshu University Hospital, Matsumoto, Nagano 390-8621, Japan
| | - Takayuki Honda
- Department of Laboratory Medicine, Shinshu University Hospital, Matsumoto, Nagano 390-8621, Japan
| |
Collapse
|
75
|
Yang Y, Yin W, He W, Jiang C, Zhou X, Song X, Zhu J, Fei K, Cao W, Jiang G. Phenotype-genotype correlation in multiple primary lung cancer patients in China. Sci Rep 2016; 6:36177. [PMID: 27796337 PMCID: PMC5087074 DOI: 10.1038/srep36177] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 10/12/2016] [Indexed: 11/09/2022] Open
Abstract
Due to recent advances in high-resolution detection technology, multiple primary lung cancer (MPLC) is becoming an increasingly common diagnosis. However, the genotype-phenotype correlations in MPLC patients have not yet been assessed. In this study, we analyzed the clinical and pathological data for 129 consecutive MPLC patients who received curative surgery at the Tongji University Shanghai Pulmonary Hospital, China. We have screened 129 patients in the present study and found mutations in EGFR, BRAF, ROS1 and KRAS genes, as well as the rearrangement of the EML4-ALK gene in 113 patients. The mean patient age was 59.9 (25-78) years old and 41 patients were males (31.8%). Among the total patients, 123 (95.4%) had two primary lesions, 5 (3.9%) had three primary lesions, and 1 (0.8%) had four primary lesions. In 38.8% of the patients, all lesions were located on only one side of the body. Most of the detected mutations (98 patients) were in the EGFR gene. The patients exhibited significant differences in the EGFR mutation, age at diagnosis, and foci location.
Collapse
Affiliation(s)
- Yang Yang
- Department of Thoracic Surgery, Tongji University Shanghai Pulmonary Hospital, Shanghai 200433, China
| | - Wei Yin
- Key Laboratory of Oral Biomedical Engineering of Ministry of Education, Hospital and School of Stomatology, Wuhan University, Wuhan 430079, China
| | - Wenxin He
- Department of Thoracic Surgery, Tongji University Shanghai Pulmonary Hospital, Shanghai 200433, China
| | - Chao Jiang
- Department of Thoracic Surgery, Tongji University Shanghai Pulmonary Hospital, Shanghai 200433, China
| | - Xiao Zhou
- Department of Thoracic Surgery, Tongji University Shanghai Pulmonary Hospital, Shanghai 200433, China
| | - Xiao Song
- Department of Thoracic Surgery, Tongji University Shanghai Pulmonary Hospital, Shanghai 200433, China
| | - Junjie Zhu
- Department of Thoracic Surgery, Tongji University Shanghai Pulmonary Hospital, Shanghai 200433, China
| | - Ke Fei
- Department of Thoracic Surgery, Tongji University Shanghai Pulmonary Hospital, Shanghai 200433, China
| | - Weijun Cao
- Department of Respiratory, Tongji University Shanghai Pulmonary Hospital, Shanghai 200433, China
| | - Gening Jiang
- Department of Thoracic Surgery, Tongji University Shanghai Pulmonary Hospital, Shanghai 200433, China
| |
Collapse
|
76
|
Arreaza G, Qiu P, Pang L, Albright A, Hong LZ, Marton MJ, Levitan D. Pre-Analytical Considerations for Successful Next-Generation Sequencing (NGS): Challenges and Opportunities for Formalin-Fixed and Paraffin-Embedded Tumor Tissue (FFPE) Samples. Int J Mol Sci 2016; 17:ijms17091579. [PMID: 27657050 PMCID: PMC5037844 DOI: 10.3390/ijms17091579] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 09/08/2016] [Accepted: 09/13/2016] [Indexed: 12/11/2022] Open
Abstract
In cancer drug discovery, it is important to investigate the genetic determinants of response or resistance to cancer therapy as well as factors that contribute to adverse events in the course of clinical trials. Despite the emergence of new technologies and the ability to measure more diverse analytes (e.g., circulating tumor cell (CTC), circulating tumor DNA (ctDNA), etc.), tumor tissue is still the most common and reliable source for biomarker investigation. Because of its worldwide use and ability to preserve samples for many decades at ambient temperature, formalin-fixed, paraffin-embedded tumor tissue (FFPE) is likely to be the preferred choice for tissue preservation in clinical practice for the foreseeable future. Multiple analyses are routinely performed on the same FFPE samples (such as Immunohistochemistry (IHC), in situ hybridization, RNAseq, DNAseq, TILseq, Methyl-Seq, etc.). Thus, specimen prioritization and optimization of the isolation of analytes is critical to ensure successful completion of each assay. FFPE is notorious for producing suboptimal DNA quality and low DNA yield. However, commercial vendors tend to request higher DNA sample mass than what is actually required for downstream assays, which restricts the breadth of biomarker work that can be performed. We evaluated multiple genomics service laboratories to assess the current state of NGS pre-analytical processing of FFPE. Significant differences in pre-analytical capabilities were observed. Key aspects are highlighted and recommendations are made to improve the current practice in translational research.
Collapse
Affiliation(s)
- Gladys Arreaza
- Translational Medicine, Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ 07033, USA.
| | - Ping Qiu
- Translational Medicine, Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ 07033, USA.
| | - Ling Pang
- Translational Medicine, Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ 07033, USA.
| | - Andrew Albright
- Translational Medicine, Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ 07033, USA.
| | - Lewis Z Hong
- Translational Biomarkers, Merck Research Laboratories, Merck Sharp & Dohme, Singapore 609927, Singapore.
| | - Matthew J Marton
- Translational Medicine, Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ 07033, USA.
| | - Diane Levitan
- Translational Medicine, Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ 07033, USA.
| |
Collapse
|
77
|
Mäki-Nevala S, Sarhadi VK, Rönty M, Kettunen E, Husgafvel-Pursiainen K, Wolff H, Knuuttila A, Knuutila S. Hot spot mutations in Finnish non-small cell lung cancers. Lung Cancer 2016; 99:102-10. [DOI: 10.1016/j.lungcan.2016.06.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 06/07/2016] [Accepted: 06/27/2016] [Indexed: 12/22/2022]
|
78
|
Mack E, Stabla K, Riera-Knorrenschild J, Moll R, Neubauer A, Brendel C. A rational two-step approach to KRAS mutation testing in colorectal cancer using high resolution melting analysis and pyrosequencing. BMC Cancer 2016; 16:585. [PMID: 27485514 PMCID: PMC4971616 DOI: 10.1186/s12885-016-2589-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 07/20/2016] [Indexed: 01/03/2023] Open
Abstract
Background KRAS mutation testing is mandatory in the management of metastatic colorectal cancer prior to treatment with anti-EGFR antibodies as patients whose tumors express mutant KRAS do not benefit from these agents. Although the U.S. Food and Drug Administration has recently approved two in-vitro diagnostics kits for determination of KRAS status, there is generally no consensus on the preferred method and new tests are continuously being developed. Most of these techniques focus on the hotspot mutations at codons 12 and 13 of the KRAS gene. Methods We describe a two-step approach to KRAS codon 12/13 mutation testing involving high resolution melting analysis (HRM) followed by pyrosequencing using the Therascreen KRAS Pyro kit (Qiagen) of only those samples that are not clearly identified as KRAS wildtype or mutant by HRM. First, we determined KRAS status in a panel of 61 colorectal cancer samples using both methods to compare technical performance and concordance of results. Subsequently, we evaluated practicability and costs of our concept in an independent set of 120 colorectal cancer samples in a routine diagnostic setting. Results HRM and pyrosequencing appeared to be equally sensitive, allowing for clear detection of mutant alleles at a mutant allele frequency ≥12.5 %. Pyrosequencing yielded more exploitable results due to lower input requirements and a lower rate of analysis failures. KRAS codon 12/13 status was called concordantly for 98.2 % (56/57) of all samples that could be successfully analysed by both methods and 100 % (19/19) of samples that were identified mutant by HRM. Reviewing the actual effort and expenses for KRAS mutation testing in our laboratory revealed, that the selective use of pyrosequencing for only those samples that could not be analysed by HRM increased the fraction of valid results from 87.5 % for HRM alone to 99.2 % (119/120) while allowing for a net reduction of operational costs of >75 % compared to pyrosequencing alone. Conclusions Combination of HRM and pyrosequencing in a two-step diagnostic procedure constitutes a reliable and economic analysis platform for KRAS mutation testing in colorectal cancer in a clinical setting. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2589-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elisabeth Mack
- Klinik für Hämatologie, Onkologie und Immunologie, Universitätsklinikum Gießen und Marburg, Standort Marburg, Philipps-Universität Marburg, Baldingerstraße, Marburg, Germany
| | - Kathleen Stabla
- Klinik für Hämatologie, Onkologie und Immunologie, Universitätsklinikum Gießen und Marburg, Standort Marburg, Philipps-Universität Marburg, Baldingerstraße, Marburg, Germany
| | - Jorge Riera-Knorrenschild
- Klinik für Hämatologie, Onkologie und Immunologie, Universitätsklinikum Gießen und Marburg, Standort Marburg, Philipps-Universität Marburg, Baldingerstraße, Marburg, Germany
| | - Roland Moll
- Institut für Pathologie, Universitätsklinikum Gießen und Marburg, Standort Marburg, Philipps-Universität Marburg, Baldingerstraße, Marburg, Germany
| | - Andreas Neubauer
- Klinik für Hämatologie, Onkologie und Immunologie, Universitätsklinikum Gießen und Marburg, Standort Marburg, Philipps-Universität Marburg, Baldingerstraße, Marburg, Germany
| | - Cornelia Brendel
- Klinik für Hämatologie, Onkologie und Immunologie, Universitätsklinikum Gießen und Marburg, Standort Marburg, Philipps-Universität Marburg, Baldingerstraße, Marburg, Germany.
| |
Collapse
|
79
|
Shaw WH, Lin Q, Muhammad ZZBR, Lee JJ, Khong WX, Ng OT, Tan EL, Li P. Identification of HIV Mutation as Diagnostic Biomarker through Next Generation Sequencing. J Clin Diagn Res 2016; 10:DC04-8. [PMID: 27630839 DOI: 10.7860/jcdr/2016/19760.8140] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 05/31/2016] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Current clinical detection of Human immunodeficiency virus 1 (HIV-1) is used to target viral genes and proteins. However, the immunoassay, such as viral culture or Polymerase Chain Reaction (PCR), lacks accuracy in the diagnosis, as these conventional assays rely on the stable genome and HIV-1 is a highly-mutated virus. Next generation sequencing (NGS) promises to be transformative for the practice of infectious disease, and the rapidly reducing cost and processing time mean that this will become a feasible technology in diagnostic and research laboratories in the near future. The technology offers the superior sensitivity to detect the pathogenic viruses, including unknown and unexpected strains. AIM To leverage the NGS technology in order to improve current HIV-1 diagnosis and genotyping methods. MATERIALS AND METHODS Ten blood samples were collected from HIV-1 infected patients which were diagnosed by RT PCR at Singapore Communicable Disease Centre, Tan Tock Seng Hospital from October 2014 to March 2015. Viral RNAs were extracted from blood plasma and reversed into cDNA. The HIV-1 cDNA samples were cleaned up using a PCR purification kit and the sequencing library was prepared and identified through MiSeq. RESULTS Two common mutations were observed in all ten samples. The common mutations were identified at genome locations 1908 and 2104 as missense and silent mutations respectively, conferring S37N and S3S found on aspartic protease and reverse transcriptase subunits. CONCLUSION The common mutations identified in this study were not previously reported, therefore suggesting the potential for them to be used for identification of viral infection, disease transmission and drug resistance. This was especially the case for, missense mutation S37N which could cause an amino acid change in viral proteases thus reducing the binding affinity of some protease inhibitors. Thus, the unique common mutations identified in this study could be used as diagnostic biomarkers to indicate the origin of infection as being from Singapore.
Collapse
Affiliation(s)
- Wen Hui Shaw
- Student, Centre for Biomedical and Life Sciences, Singapore Polytechnic , 500 Dover Road, Singapore
| | - Qianqian Lin
- Student, Centre for Biomedical and Life Sciences, Singapore Polytechnic , 500 Dover Road, Singapore
| | | | - Jia Jun Lee
- Research Assistant, Department of Infectious Diseases, Tan Tock Seng Hospital , Singapore
| | - Wei Xin Khong
- Scientific Officer, Department of Infectious Diseases, Tan Tock Seng Hospital , Singapore
| | - Oon Tek Ng
- Consultant, Department of Infectious Diseases, Tan Tock Seng Hospital , Singapore
| | - Eng Lee Tan
- Centre Director, Department of Paediatrics, University Children's Medical Institute, National University Hospital , Singapore, 119074 and Centre for Biomedical and Life Sciences, Singapore Polytechnic, 500 Dover Road, Singapore
| | - Peng Li
- Project Leader, Centre for Biomedical and Life Sciences, Singapore Polytechnic , 500 Dover Road, Singapore
| |
Collapse
|
80
|
Diagnostic Accuracy of PIK3CA Mutation Detection by Circulating Free DNA in Breast Cancer: A Meta-Analysis of Diagnostic Test Accuracy. PLoS One 2016; 11:e0158143. [PMID: 27336598 PMCID: PMC4918940 DOI: 10.1371/journal.pone.0158143] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 06/11/2016] [Indexed: 12/18/2022] Open
Abstract
Mutation of p110 alpha-catalytic subunit of phosphatidylinositol 3-kinase (PIK3CA) has high predictive and prognostic values for breast cancer. Hence, there has been a marked interest in detecting and monitoring PIK3CA genotype with non-invasive technique, such as circulating free DNA (cfDNA). However, the diagnostic accuracy of PIK3CA genotyping by cfDNA is still a problem of controversy. Here, we conducted the first meta-analysis to evaluate overall diagnostic performance of cfDNA for PIK3CA mutation detection. Literature search was performed in Pubmed, Embase and Cochrane Central Register of Controlled Trials databases. Seven cohorts from five studies with 247 patients were included. The pooled sensitivity, specificity, positive and negative likelihood ratio, diagnostic odds ratio and area under summary receiver operating characteristic curve were calculated for accuracy evaluation. The pooled sensitivity and specificity were 0.86 (95% confidence interval [CI] 0.32–0.99) and 0.98 (95% CI 0.86–1.00), respectively; the pooled positive and negative likelihood ratio were 42.8 (95% CI 5.1–356.9) and 0.14 (95% CI 0.02–1.34), respectively; diagnostic odds ratio for evaluating the overall diagnostic performance was 300 (95% CI 8–11867); area under summary receiver operating characteristic curve reached 0.99 (95% CI 0.97–0.99). Subgroup analysis with metastatic breast cancer revealed remarkable improvement in diagnostic performance (sensitivity: 0.86–0.91; specificity: 0.98; diagnostic odds ratio: 300–428). This meta-analysis proved that detecting PIK3CA gene mutation by cfDNA has high diagnostic accuracy in breast cancer, especially for metastatic breast cancer. It may serve as a reliable non-invasive assay for detecting and monitoring PIK3CA mutation status in order to deliver personalized and precise treatment.
Collapse
|
81
|
Kruglyak KM, Lin E, Ong FS. Next-Generation Sequencing and Applications to the Diagnosis and Treatment of Lung Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 890:123-36. [PMID: 26703802 DOI: 10.1007/978-3-319-24932-2_7] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cancer is a genetic disease characterized by uncontrolled growth of abnormal cells. Over time, somatic mutations accumulate in the cells of an individual due to replication errors, chromosome segregation errors, or DNA damage. When not caught by traditional mechanisms, these somatic mutations can lead to cellular proliferation, the hallmark of cancer. Lung cancer is the leading cause of cancer-related mortality in the United States, accounting for approximately 160,000 deaths annually. Five year survival rates for lung cancer remain low (<50 %) for all stages, with even worse prognosis (<15 %) in late stage cases. Technological advances, including advances in next-generation sequencing (NGS), offer the vision of personalized medicine or precision oncology, wherein an individual's treatment can be based on his or her individual molecular profile, rather than on historical population-based medicine. Towards this end, NGS has already been used to identify new biomarker candidates for the early diagnosis of lung cancer and is increasingly used to guide personalized treatment decisions. In this review we will provide a high-level overview of NGS technology and summarize its application to the diagnosis and treatment of lung cancer. We will also describe how NGS can drive advances that bring us closer to precision oncology and discuss some of the technical challenges that will need to be overcome in order to realize this ultimate goal.
Collapse
Affiliation(s)
| | - Erick Lin
- Medical Affairs, Ambry Genetics, Inc., Aliso Viejo, CA, USA
| | - Frank S Ong
- Medical Affairs and Clinical Development, NantHealth, LLC, Culver City, CA, USA.
| |
Collapse
|
82
|
Froyen G, Broekmans A, Hillen F, Pat K, Achten R, Mebis J, Rummens JL, Willemse J, Maes B. Validation and Application of a Custom-Designed Targeted Next-Generation Sequencing Panel for the Diagnostic Mutational Profiling of Solid Tumors. PLoS One 2016; 11:e0154038. [PMID: 27101000 PMCID: PMC4839685 DOI: 10.1371/journal.pone.0154038] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 04/07/2016] [Indexed: 01/15/2023] Open
Abstract
The inevitable switch from standard molecular methods to next-generation sequencing for the molecular profiling of tumors is challenging for most diagnostic laboratories. However, fixed validation criteria for diagnostic accreditation are not in place because of the great variability in methods and aims. Here, we describe the validation of a custom panel of hotspots in 24 genes for the detection of somatic mutations in non-small cell lung carcinoma, colorectal carcinoma and malignant melanoma starting from FFPE sections, using 14, 36 and 5 cases, respectively. The targeted hotspots were selected for their present or future clinical relevance in solid tumor types. The target regions were enriched with the TruSeq approach starting from limited amounts of DNA. Cost effective sequencing of 12 pooled libraries was done using a micro flow cell on the MiSeq and subsequent data analysis with MiSeqReporter and VariantStudio. The entire workflow was diagnostically validated showing a robust performance with maximal sensitivity and specificity using as thresholds a variant allele frequency >5% and a minimal amplicon coverage of 300. We implemented this method through the analysis of 150 routine diagnostic samples and identified clinically relevant mutations in 16 genes including KRAS (32%), TP53 (32%), BRAF (12%), APC (11%), EGFR (8%) and NRAS (5%). Importantly, the highest success rate was obtained when using also the low quality DNA samples. In conclusion, we provide a workflow for the validation of targeted NGS by a custom-designed pan-solid tumor panel in a molecular diagnostic lab and demonstrate its robustness in a clinical setting.
Collapse
Affiliation(s)
- Guy Froyen
- Department of Clinical Biology, Jessa Hospital, Hasselt, Belgium
- * E-mail:
| | - An Broekmans
- Department of Clinical Biology, Jessa Hospital, Hasselt, Belgium
| | - Femke Hillen
- Department of Clinical Biology, Jessa Hospital, Hasselt, Belgium
| | - Karin Pat
- Department of Pneumology, Jessa Hospital, Hasselt, Belgium
| | - Ruth Achten
- Department of Pathology, Jessa Hospital, Hasselt, Belgium
| | - Jeroen Mebis
- Department of Medical Oncology, Jessa Hospital, Hasselt, Belgium
| | - Jean-Luc Rummens
- Department of Clinical Biology, Jessa Hospital, Hasselt, Belgium
| | - Johan Willemse
- Department of Clinical Biology, Jessa Hospital, Hasselt, Belgium
- Department of Clinical Biology, AZ Turnhout, Turnhout, Belgium
| | - Brigitte Maes
- Department of Clinical Biology, Jessa Hospital, Hasselt, Belgium
| |
Collapse
|
83
|
Deciphering intra-tumor heterogeneity of lung adenocarcinoma confirms that dominant, branching, and private gene mutations occur within individual tumor nodules. Virchows Arch 2016; 468:651-62. [DOI: 10.1007/s00428-016-1931-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Revised: 01/09/2016] [Accepted: 03/21/2016] [Indexed: 01/17/2023]
|
84
|
Increase EGFR Mutations Detection Rate in Lung Adenocarcinoma by Real-Time PCR Screening Followed by Direct Sequencing. Appl Immunohistochem Mol Morphol 2016; 23:343-8. [PMID: 25961746 DOI: 10.1097/pdm.0000000000000037] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Recently, a number of small-molecule tyrosine kinase inhibitors (TKIs) have been developed to target the ATP-binding cleft of the epidermal growth factor receptor (EGFR). The presence of EGFR mutations in non-small cell lung cancer (NSCLC) correlates with the responsiveness to TKIs. Therefore, the identification of EGFR mutations before the administration of TKIs of NSCLC has become important. The aim of the present study was to investigate the occurrence of EGFR mutations in the southern Taiwanese population with NSCLC using a combination of real-time polymerase chain reaction (PCR) kit and direct sequencing. METHODS In the present study, DNAs were extracted from 249 cases of formalin-fixed, paraffin-embedded NSCLC samples for clinical EGFR mutational analysis by real-time PCR kit and direct sequencing. RESULTS The results showed that the frequency of EGFR mutations is 63% in the southern Taiwanese population. Most of the EGFR mutations are located at exons 19 and 21. In addition, we indicated that a combination of real-time PCR kit and direct sequencing increases the rate of mutation by 4%. Direct sequencing revealed 9 EGFR mutations including 6 reported EGFR mutations and 3 novel EGFR mutations. CONCLUSIONS In the present study, we have demonstrated that a combination of real-time PCR kit and direct sequencing increases the detection rate of EGFR mutations. Therefore, our proposed EGFR mutation detection strategy could be applied in clinical settings. In addition, our results indicated the prevalence of EGFR mutational status in the southern Taiwanese population.
Collapse
|
85
|
Plönes T, Engel-Riedel W, Stoelben E, Limmroth C, Schildgen O, Schildgen V. Molecular Pathology and Personalized Medicine: The Dawn of a New Era in Companion Diagnostics-Practical Considerations about Companion Diagnostics for Non-Small-Cell-Lung-Cancer. J Pers Med 2016; 6:jpm6010003. [PMID: 26784235 PMCID: PMC4810382 DOI: 10.3390/jpm6010003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 01/12/2016] [Accepted: 01/13/2016] [Indexed: 01/04/2023] Open
Abstract
Companion diagnostics (CDx) have become a major tool in molecular pathology and assist in therapy decisions in an increasing number of various cancers. Particularly, the developments in lung cancer have been most impressing in the last decade and consequently lung cancer mutation testing and molecular profiling has become a major business of diagnostic laboratories. However, it has become difficult to decide which biomarkers are currently relevant for therapy decisions, as many of the new biomarkers are not yet approved as therapy targets, remain in the status of clinical studies, or still have not left the experimental phase. The current review is focussed on those markers that do have current therapy implications, practical implications arising from the respective companion diagnostics, and thus is focused on daily practice.
Collapse
Affiliation(s)
- Till Plönes
- Lungclinic Merheim, Department of Thoracic Surgery, Lung Clinic Cologne, Kliniken der Stadt Köln gGmbH, Cologne Merheim Hospital, Faculty of Health/School of Medicine, Witten/Herdecke, Ostmerheimerstrasse 200, 51109 Köln, Germany.
| | - Walburga Engel-Riedel
- Lungclinic Merheim, Department of Thoracic Surgery, Lung Clinic Cologne, Kliniken der Stadt Köln gGmbH, Cologne Merheim Hospital, Faculty of Health/School of Medicine, Witten/Herdecke, Ostmerheimerstrasse 200, 51109 Köln, Germany.
| | - Erich Stoelben
- Lungclinic Merheim, Department of Thoracic Surgery, Lung Clinic Cologne, Kliniken der Stadt Köln gGmbH, Cologne Merheim Hospital, Faculty of Health/School of Medicine, Witten/Herdecke, Ostmerheimerstrasse 200, 51109 Köln, Germany.
| | - Christina Limmroth
- Clinics for Internal Medicine Holweide, Hospital of Cologne, Neufelder Str. 34, 51067 Köln, Germany.
| | - Oliver Schildgen
- Institute for Pathology, Hospital of Cologne, Private University Witten/Herdecke, Ostmerheimerstrasse 200, 51109 Köln, Germany.
| | - Verena Schildgen
- Institute for Pathology, Hospital of Cologne, Private University Witten/Herdecke, Ostmerheimerstrasse 200, 51109 Köln, Germany.
| |
Collapse
|
86
|
Dickson DJ, Pfeifer JD. Real-world data in the molecular era-finding the reality in the real world. Clin Pharmacol Ther 2016; 99:186-97. [PMID: 26565654 DOI: 10.1002/cpt.300] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 11/10/2015] [Indexed: 01/06/2023]
Abstract
Real-world data (RWD) promises to provide a pivotal element to the understanding of personalized medicine. However, without true representation (or the reality) of the patient-disease biosystem and its molecular contributors, RWD may hamper rather than help this advancement. In this review article, we discuss RWD vs. clinical reality and the disconnects that exist currently (emphasizing molecular medicine), and methods of closing the gaps between RWD and reality.
Collapse
Affiliation(s)
- D J Dickson
- Molecular Evidence Development Consortium, Rexburg, Idaho, USA
| | - J D Pfeifer
- Department of Pathology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
87
|
Wang W, Jiang X, Song Z, Zhang Y. Patients harboring EGFR mutation after primary resistance to crizotinib and response to EGFR-tyrosine kinase inhibitor. Onco Targets Ther 2016; 9:211-5. [PMID: 26811689 PMCID: PMC4712969 DOI: 10.2147/ott.s97100] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Anaplastic lymphoma kinase (ALK) rearrangement lung cancer responds to ALK tyrosine kinase inhibitors. It is known that many cases ultimately acquired resistance to crizotinib. However, a case of primary resistance is rare. We present a case of harboring exon 19 deletion in epidermal growth factor receptor in ALK rearranged lung adenocarcinoma, who experienced a partial tumor response to icotinib after failure with crizotinib therapy and chemotherapy. Considering the partial response, we conclude that it is important to find the cause of resistance to crizotinib. We detected gene mutations with plasma by the next-generation sequencing; the next-generation sequencing demonstrates an attractive system to identify mutations improving the outcome of patients with a deadly disease.
Collapse
Affiliation(s)
- Wenxian Wang
- Department of Chemotherapy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China
| | - Xiaowen Jiang
- Department of Chemotherapy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China
| | - Zhengbo Song
- Department of Chemotherapy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China; Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology, Hangzhou, Zhejiang, People's Republic of China
| | - Yiping Zhang
- Department of Chemotherapy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China; Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
88
|
Gao J, Wu H, Wang L, Zhang H, Duan H, Lu J, Liang Z. Validation of targeted next-generation sequencing for RAS mutation detection in FFPE colorectal cancer tissues: comparison with Sanger sequencing and ARMS-Scorpion real-time PCR. BMJ Open 2016; 6:e009532. [PMID: 26747035 PMCID: PMC4716245 DOI: 10.1136/bmjopen-2015-009532] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE To validate the targeted next-generation sequencing (NGS) platform-Ion Torrent PGM for KRAS exon 2 and expanded RAS mutations detection in formalin-fixed paraffin-embedded (FFPE) colorectal cancer (CRC) specimens, with comparison of Sanger sequencing and ARMS-Scorpion real-time PCR. SETTING Beijing, China. PARTICIPANTS 51 archived FFPE CRC samples (36 men, 15 women) were retrospectively randomly selected and then checked by an experienced pathologist for sequencing based on histological confirmation of CRC and availability of sufficient tissue. METHODS RAS mutations were detected in the 51 FFPE CRC samples by PGM analysis, Sanger sequencing and the Therascreen KRAS assay, respectively. Agreement among the 3 methods was assessed. Assay sensitivity was further determined by sequencing serially diluted DNA from FFPE cell lines with known mutation statuses. RESULTS 13 of 51 (25.5%) cases had a mutation in KRAS exon 2, as determined by PGM analysis. PGM analysis showed 100% (51/51) concordance with Sanger sequencing (κ=1.000, 95% CI 1 to 1) and 98.04% (50/51) agreement with the Therascreen assay (κ=0.947, 95% CI 0.844 to 1) for detecting KRAS exon 2 mutations, respectively. The only discrepant case harboured a KRAS exon 2 mutation (c.37G>T) that was not covered by the Therascreen kit. The dilution series experiment results showed that PGM was able to detect KRAS mutations at a frequency of as low as 1%. Importantly, RAS mutations other than KRAS exon 2 mutations were also detected in 10 samples by PGM. Furthermore, mutations in other CRC-related genes could be simultaneously detected in a single test by PGM. CONCLUSIONS The targeted NGS platform is specific and sensitive for KRAS exon 2 mutation detection and is appropriate for use in routine clinical testing. Moreover, it is sample saving and cost-efficient and time-efficient, and has great potential for clinical application to expand testing to include mutations in RAS and other CRC-related genes.
Collapse
Affiliation(s)
- Jie Gao
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, China
| | - Huanwen Wu
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, China
| | - Li Wang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, China
| | - Hui Zhang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, China
| | - Huanli Duan
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, China
| | - Junliang Lu
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, China
| | - Zhiyong Liang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, China
| |
Collapse
|
89
|
Pécuchet N, Legras A, Laurent-Puig P, Blons H. Place du NGS (Next Generation Sequencing) et de l’ADN tumoral circulant dans le testing moléculaire des cancers bronchiques. Ann Pathol 2016; 36:80-93. [DOI: 10.1016/j.annpat.2015.11.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 11/12/2015] [Indexed: 02/05/2023]
|
90
|
Emerging Biomarkers in Personalized Therapy of Lung Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 890:25-36. [DOI: 10.1007/978-3-319-24932-2_2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
91
|
D’Haene N, Le Mercier M, De Nève N, Blanchard O, Delaunoy M, El Housni H, Dessars B, Heimann P, Remmelink M, Demetter P, Tejpar S, Salmon I. Clinical Validation of Targeted Next Generation Sequencing for Colon and Lung Cancers. PLoS One 2015; 10:e0138245. [PMID: 26366557 PMCID: PMC4569137 DOI: 10.1371/journal.pone.0138245] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 08/27/2015] [Indexed: 11/18/2022] Open
Abstract
Objective Recently, Next Generation Sequencing (NGS) has begun to supplant other technologies for gene mutation testing that is now required for targeted therapies. However, transfer of NGS technology to clinical daily practice requires validation. Methods We validated the Ion Torrent AmpliSeq Colon and Lung cancer panel interrogating 1850 hotspots in 22 genes using the Ion Torrent Personal Genome Machine. First, we used commercial reference standards that carry mutations at defined allelic frequency (AF). Then, 51 colorectal adenocarcinomas (CRC) and 39 non small cell lung carcinomas (NSCLC) were retrospectively analyzed. Results Sensitivity and accuracy for detecting variants at an AF >4% was 100% for commercial reference standards. Among the 90 cases, 89 (98.9%) were successfully sequenced. Among the 86 samples for which NGS and the reference test were both informative, 83 showed concordant results between NGS and the reference test; i.e. KRAS and BRAF for CRC and EGFR for NSCLC, with the 3 discordant cases each characterized by an AF <10%. Conclusions Overall, the AmpliSeq colon/lung cancer panel was specific and sensitive for mutation analysis of gene panels and can be incorporated into clinical daily practice.
Collapse
Affiliation(s)
- Nicky D’Haene
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Marie Le Mercier
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Nancy De Nève
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Oriane Blanchard
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Mélanie Delaunoy
- Department of Genetics, Erasme Hospital,Université Libre de Bruxelles, Brussels, Belgium
| | - Hakim El Housni
- Department of Genetics, Erasme Hospital,Université Libre de Bruxelles, Brussels, Belgium
| | - Barbara Dessars
- Department of Genetics, Erasme Hospital,Université Libre de Bruxelles, Brussels, Belgium
| | - Pierre Heimann
- Department of Genetics, Erasme Hospital,Université Libre de Bruxelles, Brussels, Belgium
| | - Myriam Remmelink
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Pieter Demetter
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Sabine Tejpar
- Department of Oncology, University Hospital Leuven, Leuven, Belgium
| | - Isabelle Salmon
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
- * E-mail:
| |
Collapse
|
92
|
Moens LNJ, Falk-Sörqvist E, Ljungström V, Mattsson J, Sundström M, La Fleur L, Mathot L, Micke P, Nilsson M, Botling J. HaloPlex Targeted Resequencing for Mutation Detection in Clinical Formalin-Fixed, Paraffin-Embedded Tumor Samples. J Mol Diagn 2015; 17:729-39. [PMID: 26354930 DOI: 10.1016/j.jmoldx.2015.06.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 05/29/2015] [Accepted: 06/17/2015] [Indexed: 12/13/2022] Open
Abstract
In recent years, the advent of massively parallel next-generation sequencing technologies has enabled substantial advances in the study of human diseases. Combined with targeted DNA enrichment methods, high sequence coverage can be obtained for different genes simultaneously at a reduced cost per sample, creating unique opportunities for clinical cancer diagnostics. However, the formalin-fixed, paraffin-embedded (FFPE) process of tissue samples, routinely used in pathology departments, results in DNA fragmentation and nucleotide modifications that introduce a number of technical challenges for downstream biomolecular analyses. We evaluated the HaloPlex target enrichment system for somatic mutation detection in 80 tissue fractions derived from 20 clinical cancer cases with paired tumor and normal tissue available in both FFPE and fresh-frozen format. Several modifications to the standard method were introduced, including a reduced target fragment length and two strand capturing. We found that FFPE material can be used for HaloPlex-based target enrichment and next-generation sequencing, even when starting from small amounts of DNA. By specifically capturing both strands for each target fragment, we were able to reduce the number of false-positive errors caused by FFPE-induced artifacts and lower the detection limit for somatic mutations. We believe that the HaloPlex method presented here will be broadly applicable as a tool for somatic mutation detection in clinical cancer settings.
Collapse
Affiliation(s)
- Lotte N J Moens
- Department of Immunology Genetics and Pathology, Uppsala University, Science for Life Laboratory, Uppsala, Sweden
| | - Elin Falk-Sörqvist
- Department of Immunology Genetics and Pathology, Uppsala University, Science for Life Laboratory, Uppsala, Sweden
| | - Viktor Ljungström
- Department of Immunology Genetics and Pathology, Uppsala University, Science for Life Laboratory, Uppsala, Sweden
| | - Johanna Mattsson
- Department of Immunology Genetics and Pathology, Uppsala University, Science for Life Laboratory, Uppsala, Sweden
| | - Magnus Sundström
- Department of Immunology Genetics and Pathology, Uppsala University, Science for Life Laboratory, Uppsala, Sweden
| | - Linnéa La Fleur
- Department of Immunology Genetics and Pathology, Uppsala University, Science for Life Laboratory, Uppsala, Sweden
| | - Lucy Mathot
- Department of Immunology Genetics and Pathology, Uppsala University, Science for Life Laboratory, Uppsala, Sweden
| | - Patrick Micke
- Department of Immunology Genetics and Pathology, Uppsala University, Science for Life Laboratory, Uppsala, Sweden
| | - Mats Nilsson
- Department of Immunology Genetics and Pathology, Uppsala University, Science for Life Laboratory, Uppsala, Sweden; Department of Biochemistry and Biophysics, Stockholm University, Science for Life Laboratory, Stockholm, Sweden.
| | - Johan Botling
- Department of Immunology Genetics and Pathology, Uppsala University, Science for Life Laboratory, Uppsala, Sweden.
| |
Collapse
|
93
|
Deeb KK, Hohman CM, Risch NF, Metzger DJ, Starostik P. Routine Clinical Mutation Profiling of Non–Small Cell Lung Cancer Using Next-Generation Sequencing. Arch Pathol Lab Med 2015; 139:913-21. [DOI: 10.5858/arpa.2014-0095-oa] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Context
The availability of massive, parallel-sequencing technologies makes possible efficient, simultaneous detection of driver and druggable mutations in cancer.
Objective
To develop an amplicon-based, next-generation sequencing, mutation-detection assay for lung cancer using the 454 GS Junior (Roche Applied Science, Indianapolis, Indiana) platform.
Design
Fusion primers incorporating target sequence, 454 adaptors, and multiplex identifiers were designed to generate 35 amplicons (median length 246 base pairs) covering 8.9 kilobases of mutational hotspots in AKT1, BRAF, EGFR, ERBB2, HRAS, KRAS, NRAS, PIK3CA, and MAP2K1 genes and all exons of the PTEN gene.
Results
The assay was validated on 23 formalin-fixed, paraffin-embedded lung cancer specimens. A minimum number of reads was consistently achieved with overall median read depth of 529× per amplicon. In total, 25 point mutations and 4 insertions/deletions (indels) with a frequency of 5.5% to 93.1% mutant alleles were detected. All EGFR, ERBB2, KRAS, PIK3CA, KRAS, and PTEN mutations, as detected by next-generation sequencing, were confirmed by pyrosequencing, with the exception of 3 point mutations in a tumor sample with low mutant-allele burden (below the pyrosequencing limit of detection).
Conclusions
The GS Junior–based, targeted, resequencing assay for a focused set of non–small cell lung cancer driver genes allows for quick and sensitive detection of point mutations and indels for the most relevant therapeutic genes in this type of cancer.
Collapse
Affiliation(s)
| | | | | | | | - Petr Starostik
- From the Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York. Dr Deeb is now with the University Hospitals Case Medical Center, Cleveland, Ohio; Mr Risch is now with Life Technologies, Grand Island, New York; and Dr Starostik is now with the Department of Pathology, Immunology and Laboratory Medicine, College of Medicine University of Florida, Gainesv
| |
Collapse
|
94
|
Combined Targeted DNA Sequencing in Non-Small Cell Lung Cancer (NSCLC) Using UNCseq and NGScopy, and RNA Sequencing Using UNCqeR for the Detection of Genetic Aberrations in NSCLC. PLoS One 2015; 10:e0129280. [PMID: 26076459 PMCID: PMC4468211 DOI: 10.1371/journal.pone.0129280] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 05/06/2015] [Indexed: 01/21/2023] Open
Abstract
The recent FDA approval of the MiSeqDx platform provides a unique opportunity to develop targeted next generation sequencing (NGS) panels for human disease, including cancer. We have developed a scalable, targeted panel-based assay termed UNCseq, which involves a NGS panel of over 200 cancer-associated genes and a standardized downstream bioinformatics pipeline for detection of single nucleotide variations (SNV) as well as small insertions and deletions (indel). In addition, we developed a novel algorithm, NGScopy, designed for samples with sparse sequencing coverage to detect large-scale copy number variations (CNV), similar to human SNP Array 6.0 as well as small-scale intragenic CNV. Overall, we applied this assay to 100 snap-frozen lung cancer specimens lacking same-patient germline DNA (07–0120 tissue cohort) and validated our results against Sanger sequencing, SNP Array, and our recently published integrated DNA-seq/RNA-seq assay, UNCqeR, where RNA-seq of same-patient tumor specimens confirmed SNV detected by DNA-seq, if RNA-seq coverage depth was adequate. In addition, we applied the UNCseq assay on an independent lung cancer tumor tissue collection with available same-patient germline DNA (11–1115 tissue cohort) and confirmed mutations using assays performed in a CLIA-certified laboratory. We conclude that UNCseq can identify SNV, indel, and CNV in tumor specimens lacking germline DNA in a cost-efficient fashion.
Collapse
|
95
|
Di Paolo A, Polillo M, Lastella M, Bocci G, Del Re M, Danesi R. Methods: for studying pharmacogenetic profiles of combination chemotherapeutic drugs. Expert Opin Drug Metab Toxicol 2015; 11:1253-67. [PMID: 26037261 DOI: 10.1517/17425255.2015.1053460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Molecular and genetic analysis of tumors and individuals has led to patient-centered therapies, through the discovery and identification of genetic markers predictive of drug efficacy and tolerability. Present therapies often include a combination of synergic drugs, each of them directed against different targets. Therefore, the pharmacogenetic profiling of tumor masses and patients is becoming a challenge, and several questions may arise when planning a translational study. AREAS COVERED The review presents the different techniques used to stratify oncology patients and to tailor antineoplastic treatments according to individual pharmacogenetic profiling. The advantages of these methodologies are discussed as well as current limits. EXPERT OPINION Facing the rapid technological evolution for genetic analyses, the most pressing issues are the choice of appropriate strategies (i.e., from gene candidate up to next-generation sequencing) and the possibility to replicate study results for their final validation. It is likely that the latter will be the major obstacle in the future. However, the present landscape is opening up new possibilities, overcoming those hurdles that have limited result translation into clinical settings for years.
Collapse
Affiliation(s)
- Antonello Di Paolo
- University of Pisa, Department of Clinical and Experimental Medicine, Via Roma 55, 56126 Pisa , Italy +39 050 2218755 ; +39 050 2218758 ;
| | | | | | | | | | | |
Collapse
|
96
|
Quinn AM, Hickson N, Adaway M, Priest L, Jaeger E, Udar N, Keeling C, Kamieniorz M, Dive C, Wallace A, Byers RJ, Newman WG, Nonaka D, Blackhall FH. Diagnostic Mutation Profiling and Validation of Non-Small-Cell Lung Cancer Small Biopsy Samples using a High Throughput Platform. J Thorac Oncol 2015; 10:784-792. [PMID: 25634010 DOI: 10.1097/jto.0000000000000473] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND A single platform designed for the synchronous screening of multiple mutations can potentially enable molecular profiling in samples of limited tumor tissue. This approach is ideal for the assessment of advanced non-small-cell lung cancer (NSCLC) diagnostic specimens, which often comprise small biopsies. Therefore, we aimed in this study to validate the mass spectrometry-based Sequenom LungCarta panel and MassARRAY platform using DNA extracted from a single 5 μM formalin-fixed paraffin-embedded tissue section. METHODS Mutations, including those with an equivocal spectrum, detected in 90 cases of NSCLC (72 lung biopsies, 13 metastatic tissue biopsies, three resections, and two cytology samples) were validated by a combination of standard sequencing techniques, immunohistochemical staining for p53 protein, and next-generation sequencing with the TruSight Tumor panel. RESULTS Fifty-five mutations were diagnosed in 47 cases (52%) in the following genes: TP53 (22), KRAS (15), EGFR (5), MET (3), PIK3CA (3), STK11 (2), NRF-2 (2), EPHA5 (1), EPHA3 (1), and MAP2K1 (1). Of the 90 samples, one failed testing due to poor quality DNA. An additional 7 TP53 mutations were detected by next-generation sequencing, which facilitated the interpretation of p53 immunohistochemistry but required 5 × 10 μM tumor sections per sample tested. CONCLUSIONS The LungCarta panel is a sensitive method of screening for multiple alterations (214 mutations across 26 genes) and which optimizes the use of limited amounts of tumor DNA isolated from small specimens.
Collapse
Affiliation(s)
- Anne Marie Quinn
- Manchester Centre for Genomic Medicine, Institute of Human Development, University of Manchester and St. Mary's Hospital, Manchester, UK.
| | - Nicholas Hickson
- Manchester Centre for Genomic Medicine, Institute of Human Development, University of Manchester and St. Mary's Hospital, Manchester, UK
| | - Megan Adaway
- Manchester Centre for Genomic Medicine, Institute of Human Development, University of Manchester and St. Mary's Hospital, Manchester, UK
| | - Lynsey Priest
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester Cancer Research Centre, University of Manchester, Manchester, UK
| | | | | | | | | | - Caroline Dive
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester Cancer Research Centre, University of Manchester, Manchester, UK
| | - Andrew Wallace
- Manchester Centre for Genomic Medicine, Institute of Human Development, University of Manchester and St. Mary's Hospital, Manchester, UK
| | | | - William G Newman
- Manchester Centre for Genomic Medicine, Institute of Human Development, University of Manchester and St. Mary's Hospital, Manchester, UK
| | | | - Fiona H Blackhall
- The Christie NHS Foundation Trust, Manchester, UK; Medical Oncology, Institute of Cancer Studies, University of Manchester, Manchester, UK
| |
Collapse
|
97
|
Heuckmann JM, Thomas RK. A new generation of cancer genome diagnostics for routine clinical use: overcoming the roadblocks to personalized cancer medicine. Ann Oncol 2015; 26:1830-1837. [PMID: 25899787 DOI: 10.1093/annonc/mdv184] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 04/07/2015] [Indexed: 12/22/2022] Open
Abstract
The identification of 'druggable' kinase gene alterations has revolutionized cancer treatment in the last decade by providing new and successfully targetable drug targets. Thus, genotyping tumors for matching the right patients with the right drugs have become a clinical routine. Today, advances in sequencing technology and computational genome analyses enable the discovery of a constantly growing number of genome alterations relevant for clinical decision making. As a consequence, several technological approaches have emerged in order to deal with these rapidly increasing demands for clinical cancer genome analyses. Here, we describe challenges on the path to the broad introduction of diagnostic cancer genome analyses and the technologies that can be applied to overcome them. We define three generations of molecular diagnostics that are in clinical use. The latest generation of these approaches involves deep and thus, highly sensitive sequencing of all therapeutically relevant types of genome alterations-mutations, copy number alterations and rearrangements/fusions-in a single assay. Such approaches therefore have substantial advantages (less time and less tissue required) over PCR-based methods that typically have to be combined with fluorescence in situ hybridization for detection of gene amplifications and fusions. Since these new technologies work reliably on routine diagnostic formalin-fixed, paraffin-embedded specimens, they can help expedite the broad introduction of personalized cancer therapy into the clinic by providing comprehensive, sensitive and accurate cancer genome diagnoses in 'real-time'.
Collapse
Affiliation(s)
| | - R K Thomas
- Department of Translational Genomics, Medical Faculty, University of Cologne, Cologne, Germany.
| |
Collapse
|
98
|
Dudley JC, Gurda GT, Tseng LH, Anderson DA, Chen G, Taube JM, Gocke CD, Eshleman JR, Lin MT. Tumor cellularity as a quality assurance measure for accurate clinical detection of BRAF mutations in melanoma. Mol Diagn Ther 2015; 18:409-18. [PMID: 24604154 DOI: 10.1007/s40291-014-0091-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND Detection of BRAF mutations is an established standard of care to predict small-molecule inhibitor (vemurafenib) response in metastatic melanoma. Molecular assays should be designed to detect not only the most common p.V600E mutation, but also p.V600K and other non-p.V600E mutations. OBJECTIVE The purpose of this study was to assess if tumor cellularity can function as a quality assurance (QA) measure in molecular diagnostics. Potential causes of discrepancy between the observed and predicted mutant allele percentage were also explored. METHODS We correlated pathologist-generated estimates of tumor cellularity versus mutant allele percentage via pyrosequencing as a QA measure for BRAF mutation detection in formalin-fixed, paraffin-embedded melanoma specimens. RESULTS BRAF mutations were seen in 27/62 (44 %) specimens, with 93 % p.V600E and 7 % non-p.V600E. Correlation between p.V600E mutant percentage and tumor cellularity was poor-moderate (r = -0.02; p = 0.8), primarily because six samples showed a low p.V600E signal despite high tumor cellularity. A QA investigation revealed that our initial pyrosequencing assay showed a false positive, weak p.V600E signal in specimens with a p.V600K mutation. A redesigned assay detected BRAF mutations in 50/131 (38 %) specimens, including 30 % non-p.V600E. This revised assay showed strong correlation between p.V600E BRAF mutant percentage and tumor cellularity (r = 0.76; p ≤ 0.01). Re-evaluation of the previously discordant samples by the revised assay confirmed a high level of p.V600K mutation in five specimens. CONCLUSIONS Pathologists play important roles in molecular diagnostics, beyond identification of correct cells for testing. Accurate evaluation of tumor cellularity not only ensures sufficient material for required analytic sensitivity, but also provides an independent QA measure of the molecular assays.
Collapse
Affiliation(s)
- Jonathan C Dudley
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Park SB202, 600 North Wolfe St., Baltimore, MD, 21287, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Hastings RJ, Bown N, Tibiletti MG, Debiec-Rychter M, Vanni R, Espinet B, van Roy N, Roberts P, van den Berg-de-Ruiter E, Bernheim A, Schoumans J, Chatters S, Zemanova Z, Stevens-Kroef M, Simons A, Heim S, Salido M, Ylstra B, Betts DR. Guidelines for cytogenetic investigations in tumours. Eur J Hum Genet 2015; 24:6-13. [PMID: 25804401 DOI: 10.1038/ejhg.2015.35] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 01/19/2015] [Accepted: 02/03/2015] [Indexed: 12/31/2022] Open
Affiliation(s)
- Rosalind J Hastings
- Women's Centre, John Radcliffe Hospital, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Nick Bown
- Northern Genetics Service, Institute of Genomic Medicine, Central Parkway, Newcastle, UK
| | - Maria G Tibiletti
- UO Anatomia Patologica, Ospedale di Circolo-Polo Universitario, Varese, Italy
| | - Maria Debiec-Rychter
- Laboratory for Genetics of Malignant Disorders, Department of Human Genetics, University Hospital Gasthuisberg, UZ Leuven, Leuven, Belgium
| | - Roberta Vanni
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, Italy
| | - Blanca Espinet
- Laboratori de Citogenètica Molecular, Servei de Patologia, Hospital del Mar, Barcelona, Spain
| | - Nadine van Roy
- Centre for Medical Genetics, University Hospital Ghent, Ghent, Belgium
| | - Paul Roberts
- Cytogenetics Department, St James's Hospital, Leeds, UK
| | - Eva van den Berg-de-Ruiter
- Department of Genetics, University of Groningen, University Medical Centre Groningen, RB Groningen, The Netherlands
| | - Alain Bernheim
- Génétique des tumeurs (INSERM U985), Laboratoire de Cytogénétique, Pathologie Moléculaire Gustave Roussy, Paris-Villejuif, France
| | - Jacqueline Schoumans
- Cancer Cytogenetic Unit, Lausanne University Hospital, CHUV, Lausanne, Switzerland
| | - Steve Chatters
- Haematology, Cellular and Molecular Diagnostic Service, Great Ormond St Hospital, London, UK
| | - Zuzana Zemanova
- Center of Oncocytogenetics, Institute of Clinical Biochemistry and Laboratory Diagnostics, General University Hospital and First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Marian Stevens-Kroef
- Radboud University Nijmegen Medical Centre Department of Human Genetics, HB Nijmegen, The Netherlands
| | - Annet Simons
- Radboud University Nijmegen Medical Centre Department of Human Genetics, HB Nijmegen, The Netherlands
| | - Sverre Heim
- Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Marta Salido
- Laboratori de Citogenètica Molecular, Servei de Patologia, Hospital del Mar, Barcelona, Spain
| | - Bauke Ylstra
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, MB Amsterdam, The Netherlands
| | - David R Betts
- Department of Clinical Genetics, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland
| | | | | |
Collapse
|
100
|
Takahashi Y, Akahane T, Sawada T, Ikeda H, Tempaku A, Yamauchi S, Nishihara H, Tanaka S, Nitta K, Ide W, Hashimoto I, Kamada H. Adult classical glioblastoma with a BRAF V600E mutation. World J Surg Oncol 2015; 13:100. [PMID: 25885250 PMCID: PMC4358908 DOI: 10.1186/s12957-015-0521-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 02/23/2015] [Indexed: 11/29/2022] Open
Abstract
The B-Raf proto-oncogene serine/threonine kinase (B-Raf) is a member of the Raf kinase family. The BRAF V600E mutation occurs frequently in certain brain tumors such as pleomorphic xanthoastrocytoma, ganglioglioma, and pilocytic astrocytoma, and less frequently in epithelioid and giant cell glioblastoma. BRAF V600E mutation in these cases has been canonically detected using Sanger sequencing or immunohistochemistry but not with next-generation sequencing (NGS). Moreover, to our knowledge, there is no detailed report of the BRAF V600E mutation in an adult glioblastoma with classical histologic features (c-GBM). Therefore, we performed NGS analysis to determine the mutational status of BRAF of 13 glioblastomas (GBMs) (11 primary and 2 secondary cases) and detected one tumor harboring the BRAF V600E mutation. We report here the detection of the BRAF V600E mutation in a patient with c-GBM and describe the patient’s clinical course as well as the results of histopathological analysis.
Collapse
Affiliation(s)
- Yoshinobu Takahashi
- Department of Neurosurgery, Hokuto Hospital, 7-5, Inada, Obihiro, Hokkaido, 080-0039, Japan. .,Department of Biology and Genetics, Laboratory of Cancer Medical Science, Hokuto Hospital, 7-5, Inada, Obihiro, Hokkaido, 080-0039, Japan.
| | - Toshiaki Akahane
- Department of Pathology, Hokuto Hospital, 7-5, Inada, Obihiro, Hokkaido, 080-0039, Japan. .,Department of Biology and Genetics, Laboratory of Cancer Medical Science, Hokuto Hospital, 7-5, Inada, Obihiro, Hokkaido, 080-0039, Japan.
| | - Takahiro Sawada
- Department of Biology and Genetics, Laboratory of Cancer Medical Science, Hokuto Hospital, 7-5, Inada, Obihiro, Hokkaido, 080-0039, Japan.
| | - Hidetoshi Ikeda
- Department of Neurosurgery, Hokuto Hospital, 7-5, Inada, Obihiro, Hokkaido, 080-0039, Japan.
| | - Akira Tempaku
- Department of Neurosurgery, Hokuto Hospital, 7-5, Inada, Obihiro, Hokkaido, 080-0039, Japan. .,Department of Biology and Genetics, Laboratory of Cancer Medical Science, Hokuto Hospital, 7-5, Inada, Obihiro, Hokkaido, 080-0039, Japan.
| | - Shigeru Yamauchi
- Department of Neurosurgery, Hokuto Hospital, 7-5, Inada, Obihiro, Hokkaido, 080-0039, Japan.
| | - Hiroshi Nishihara
- Department of Pathology, Hokuto Hospital, 7-5, Inada, Obihiro, Hokkaido, 080-0039, Japan. .,Department of Biology and Genetics, Laboratory of Cancer Medical Science, Hokuto Hospital, 7-5, Inada, Obihiro, Hokkaido, 080-0039, Japan. .,Department of Translational Pathology, Hokkaido University Graduate School of Medicine, N15, W7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan.
| | - Shinya Tanaka
- Department of Translational Pathology, Hokkaido University Graduate School of Medicine, N15, W7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan.
| | - Kazumi Nitta
- Department of Neurosurgery, Hokuto Hospital, 7-5, Inada, Obihiro, Hokkaido, 080-0039, Japan.
| | - Wataru Ide
- Department of Neurosurgery, Hokuto Hospital, 7-5, Inada, Obihiro, Hokkaido, 080-0039, Japan.
| | - Ikuo Hashimoto
- Department of Neurosurgery, Hokuto Hospital, 7-5, Inada, Obihiro, Hokkaido, 080-0039, Japan.
| | - Hajime Kamada
- Department of Neurosurgery, Hokuto Hospital, 7-5, Inada, Obihiro, Hokkaido, 080-0039, Japan.
| |
Collapse
|