101
|
Velizheva NP, Rechsteiner MP, Wong CE, Zhong Q, Rössle M, Bode B, Moch H, Soltermann A, Wild PJ, Tischler V. Cytology smears as excellent starting material for next-generation sequencing-based molecular testing of patients with adenocarcinoma of the lung. Cancer Cytopathol 2016; 125:30-40. [DOI: 10.1002/cncy.21771] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 07/08/2016] [Accepted: 07/27/2016] [Indexed: 11/10/2022]
Affiliation(s)
| | | | - Christine E. Wong
- Institute of Surgical Pathology; University Hospital Zurich; Zurich Switzerland
| | - Qing Zhong
- Institute of Surgical Pathology; University Hospital Zurich; Zurich Switzerland
| | - Matthias Rössle
- Cantonal Hospital Graubunden, Institute of Pathology; Chur Switzerland
| | - Beata Bode
- Institute of Surgical Pathology; University Hospital Zurich; Zurich Switzerland
| | - Holger Moch
- Institute of Surgical Pathology; University Hospital Zurich; Zurich Switzerland
| | - Alex Soltermann
- Institute of Surgical Pathology; University Hospital Zurich; Zurich Switzerland
| | - Peter J. Wild
- Institute of Surgical Pathology; University Hospital Zurich; Zurich Switzerland
| | - Verena Tischler
- Institute of Surgical Pathology; University Hospital Zurich; Zurich Switzerland
| |
Collapse
|
102
|
Technical and US regulatory issues in triaging material for the molecular laboratory. Cancer Cytopathol 2016; 125:83-90. [DOI: 10.1002/cncy.21774] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 07/24/2016] [Accepted: 07/27/2016] [Indexed: 12/31/2022]
|
103
|
Hiley CT, Le Quesne J, Santis G, Sharpe R, de Castro DG, Middleton G, Swanton C. Challenges in molecular testing in non-small-cell lung cancer patients with advanced disease. Lancet 2016; 388:1002-11. [PMID: 27598680 DOI: 10.1016/s0140-6736(16)31340-x] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 07/15/2016] [Accepted: 07/25/2016] [Indexed: 12/18/2022]
Abstract
Lung cancer diagnostics have progressed greatly in the previous decade. Development of molecular testing to identify an increasing number of potentially clinically actionable genetic variants, using smaller samples obtained via minimally invasive techniques, is a huge challenge. Tumour heterogeneity and cancer evolution in response to therapy means that repeat biopsies or circulating biomarkers are likely to be increasingly useful to adapt treatment as resistance develops. We highlight some of the current challenges faced in clinical practice for molecular testing of EGFR, ALK, and new biomarkers such as PDL1. Implementation of next generation sequencing platforms for molecular diagnostics in non-small-cell lung cancer is increasingly common, allowing testing of multiple genetic variants from a single sample. The use of next generation sequencing to recruit for molecularly stratified clinical trials is discussed in the context of the UK Stratified Medicine Programme and The UK National Lung Matrix Trial.
Collapse
Affiliation(s)
- Crispin T Hiley
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK; Division of Cancer Studies, King's College London, London, UK
| | - John Le Quesne
- Department of Cancer Studies, University of Leicester, Leicester, UK
| | - George Santis
- Department of Respiratory Medicine and Allergy, King's College London, UK
| | | | - David Gonzalez de Castro
- Centre for Molecular Pathology, Royal Marsden Hospital, Sutton, UK; School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast, UK
| | - Gary Middleton
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK; University Hospital Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Birmingham, UK
| | - Charles Swanton
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK.
| |
Collapse
|
104
|
Layfield LJ, Roy-Chowdhuri S, Baloch Z, Ehya H, Geisinger K, Hsiao SJ, Lin O, Lindeman NI, Roh M, Schmitt F, Sidiropoulos N, VanderLaan PA. Utilization of ancillary studies in the cytologic diagnosis of respiratory lesions: The papanicolaou society of cytopathology consensus recommendations for respiratory cytology. Diagn Cytopathol 2016; 44:1000-1009. [PMID: 27561242 DOI: 10.1002/dc.23549] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 07/06/2016] [Accepted: 07/14/2016] [Indexed: 12/14/2022]
Abstract
The Papanicolaou Society of Cytopathology has developed a set of guidelines for respiratory cytology including indications for sputum examination, bronchial washings and brushings, CT-guided FNA and endobronchial ultrasound guided fine needle aspiration (EBUS-FNA), as well as recommendations for classification and criteria, ancillary testing and post-cytologic diagnosis management and follow-up. All recommendation documents are based on the expertise of committee members, an extensive literature review, and feedback from presentations at national and international conferences. The guideline documents selectively present the results of these discussions. The present document summarizes recommendations for ancillary testing of cytologic samples. Ancillary testing including microbiologic, immunocytochemical, flow cytometric, and molecular testing, including next-generation sequencing are discussed. Diagn. Cytopathol. 2016;44:1000-1009. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lester J Layfield
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, Missouri
| | - Sinchita Roy-Chowdhuri
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zubair Baloch
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hormoz Ehya
- Department of Pathology, Fox Chase Cancer Center, Temple University, Philadelphia, Pennsylvania
| | - Kim Geisinger
- Department of Pathology, University of Mississippi, Jackson, Mississippi
| | - Susan J Hsiao
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Oscar Lin
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Neal I Lindeman
- Department of Pathology, Harvard Medical School, Boston, Massachusetts
| | - Michael Roh
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Fernando Schmitt
- Département de Médecine, Laboratoire National de Santé, Dudelange, Luxembourg
| | - Nikoletta Sidiropoulos
- Department of Pathology and Laboratory Medicine, The University of Vermont, Burlington, Vermont
| | - Paul A VanderLaan
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| |
Collapse
|
105
|
Peluso AL, Ieni A, Mignogna C, Zeppa P. Lymph Node Fine-Needle Cytology: Beyond Flow Cytometry. Acta Cytol 2016; 60:372-384. [PMID: 27560152 DOI: 10.1159/000447734] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 06/20/2016] [Indexed: 12/16/2022]
Abstract
Lymph node (LN) fine-needle cytology (FNC) coupled with flow cytometry immunophenotyping provides relevant information for the diagnosis of non-Hodgkin lymphoma (NHL). Numerous studies have shown FNC samples to be suitable for different molecular procedures; in this review, some of the molecular procedures most commonly employed for NHL are briefly described and evaluated in this perspective. Fluorescence in situ hybridization and chromogenic in situ hybridization are briefly described. Polymerase chain reaction (PCR)-based assays are used to identify and quantify mutations and translocations, namely immunoglobulin (IGH) and T-cell receptor rearrangements by clonality testing and IGVH somatic hypermutations either by Sanger sequencing, single-strand conformational polymorphisms or RT-PCR strategies. High-throughput technologies (HTT) encompass numerous and different diagnostic tools that share the capacity of multiple molecular investigation and sample processing in a fast and reproducible manner. HTT includes gene expression profiling, comparative genomic hybridization, single-nucleotide polymorphism arrays and next-generation sequencing technologies. A brief description of these tools and their potential application to LN FNC is reported. The challenge for FNC will be to achieve new knowledge and apply new technologies to FNC, exploiting its own basic qualities.
Collapse
Affiliation(s)
- Anna Lucia Peluso
- Department of Medicine and Surgery, University of Salerno, Baronissi, Italy
| | | | | | | |
Collapse
|
106
|
Jin M, Wakely PE. Endoscopic/Endobronchial Ultrasound-Guided Fine Needle Aspiration and Ancillary Techniques, Particularly Flow Cytometry, in Diagnosing Deep-Seated Lymphomas. Acta Cytol 2016; 60:326-335. [PMID: 27414717 DOI: 10.1159/000447253] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 05/24/2016] [Indexed: 12/14/2022]
Abstract
Evaluation of deep-seated lymphomas by fine-needle aspiration (FNA) can be challenging due to their reduced accessibility. Controversy remains as to whether FNA and ancillary techniques can be used to diagnose deep-seated lymphomas reliably and sufficiently for clinical management. Most published studies are favorable that endobronchial ultrasound (EBUS)/endoscopic ultrasound (EUS)-FNA plays an important role in the diagnosis of deep-seated lymphomas. The addition of ancillary techniques, particularly flow cytometry, increases diagnostic yield. While subclassification is possible in a reasonable proportion of cases, the reported rates of successful subclassification are lower than those for lymphoma detection/diagnosis. The diagnostic limitation exists for Hodgkin's lymphoma, grading of follicular lymphoma, and some T-cell lymphomas. The role of FNA in deep-seated lymphomas is much better established for recurrent than primary disease. It remains unclear whether the use of large-sized-needle FNA or a combination of core needle biopsy and FNA improves subclassification. It is important for cytopathologists to have considerable understanding of the WHO lymphoma classification and develop a collaborative working relationship with hematopathologists and oncologists. As EUS/EBUS-FNA techniques advance and sophisticated molecular techniques such as next- generation sequencing become possible, the role of FNA in the diagnosis of deep-seated lymphomas will possibly increase.
Collapse
Affiliation(s)
- Ming Jin
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | | |
Collapse
|
107
|
Ozluk Y, Firat P, Yegen G, Hocaoglu J, Tas S, Yilmazbayhan D. EGFR mutation testing using archival-stained smears in non-small cell lung carcinoma. Cytopathology 2016; 28:35-45. [PMID: 27389045 DOI: 10.1111/cyt.12357] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2016] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) have shown benefits regarding progression-free and overall survival in patients whose tumours show EGFR mutations. Most patients' lung cancer is metastatic when detected. Small tissue samples and cytological materials are widely used in diagnosis. The aim of the present study was to compare the EGFR mutation analysis results between cytology, small biopsies and resections. METHODS Archival material for EGFR testing was reviewed. Cell blocks and/or stained smears and tissue blocks were used where appropriate. The tumour cell count and percentage were recorded as well as the DNA content. The influence of TTF-1 immunoreactivity on EGFR testing was also investigated. RESULTS The study cohort included 300 unpaired specimens of 84 resections, 83 small biopsies and 133 cytological materials. EGFR mutation rates did not differ significantly for cytology, small biopsy and resections (P > 0.05). The higher tumour cell percentage in FNAs than in exfoliative cytology did not affect the EGFR mutation status. EGFR mutation rates were similar when either slides or cell blocks were used. Cytology slides revealed a higher tumour cell content and DNA concentration than the cell blocks. May-Grünwald-Giemsa (MGG)-stained smears had higher rates of the EGFR mutation than the Papanicolaou (Pap)-stained slides (P < 0.05). Tumours with negative immunoreactivity for TTF-1 are less likely to have an EGFR mutation (P < 0.05). CONCLUSIONS Cytological materials can be used successfully for mutation analysis in lung cancer.
Collapse
Affiliation(s)
- Y Ozluk
- Department of Pathology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - P Firat
- Department of Pathology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - G Yegen
- Department of Pathology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - J Hocaoglu
- Department of Pathology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - S Tas
- Department of Pathology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - D Yilmazbayhan
- Department of Pathology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
108
|
Roy-Chowdhuri S, Stewart J. Preanalytic Variables in Cytology: Lessons Learned From Next-Generation Sequencing—The MD Anderson Experience. Arch Pathol Lab Med 2016; 140:1191-1199. [DOI: 10.5858/arpa.2016-0117-ra] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Context.—
As our understanding of genomic alterations underlying solid tumor malignancies continues to evolve, molecular testing of tumor samples is increasingly used to guide therapeutic management. Next-generation sequencing (NGS) provides a novel platform for the simultaneous screening of multiple genes using small amounts of DNA. Several recent studies have described NGS mutational analysis using cytologic specimens. The cytopathologist's role in specimen assessment and triaging is critical to effectively implementing NGS in routine cytology practice.
Objectives.—
To review the NGS experience and a variety of preanalytic factors that affect NGS success rates of cytologic specimens at our institution.
Data Sources.—
To evaluate cytology specimen adequacy rates for NGS, we reviewed a 14-month period of image-guided fine-needle aspiration and core needle biopsies used for testing. In addition, we reviewed data from our previously published studies to evaluate preanalytic factors affecting NGS success in these specimens.
Conclusions.—
Identifying factors that affect NGS success rates in cytology specimens is crucial for a better understanding of specimen adequacy requirements and for proper use of limited-volume tissue samples. In our practice, which uses direct smears as well as cell block sections, NGS success rates in core needle biopsy and fine-needle aspiration samples are comparable. The chance of successful testing is further increased by procuring concurrent fine-needle aspiration and core needle biopsy samples. The type of glass slides used for direct smears and the method of tissue extraction affect our DNA yield. Validating a DNA input for cytology samples that is lower than that recommended by the manufacturer has significantly increased our NGS success rate.
Collapse
Affiliation(s)
- Sinchita Roy-Chowdhuri
- From the Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston
| | | |
Collapse
|
109
|
DiBardino DM, Saqi A, Elvin JA, Greenbowe J, Suh JH, Miller VA, Ali SM, Stoopler M, Bulman WA. Yield and Clinical Utility of Next-Generation Sequencing in Selected Patients With Lung Adenocarcinoma. Clin Lung Cancer 2016; 17:517-522.e3. [PMID: 27378171 DOI: 10.1016/j.cllc.2016.05.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 05/27/2016] [Accepted: 05/31/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND Next-generation sequencing is available for assessing genomic alterations in non-small-cell lung cancer (NSCLC), although the performance characteristics and clinical utility has not been well characterized. This technique can be used to sequence hundreds of known cancer-associated genes. Our aim was to investigate the diagnostic success and clinically relevant results of extensive sequencing in NSCLC patients. PATIENTS AND METHODS A case series of 49 NSCLC patients was used to determine the success of extended next-generation sequencing, record genomic alterations, and evaluate clinical utility. Data were collected in a retrospective review. Sequencing was performed using a hybridization capture of 3320 exons from 236 cancer-related genes and 47 introns of 19 genes applied to ≥50 ng of DNA and sequenced to high, uniform coverage of 622 times. RESULTS Sequencing was successful in 29 of 32 (91%) surgical/excisional specimens, and 12 of 17 (71%) nonsurgical specimens including an endoscopic forceps biopsy, core needle biopsies, fine-needle aspirates, and effusion cytologies. All 5 transthoracic core needle biopsies failed. A total of 179 genomic alterations (average 4.37 per tumor) were found. A total of 63 were clinically relevant (average 1.54 per tumor). The most frequently mutated genes were tumor protein p53, cyclin-dependent kinase inhibitor 2A, megalencephalic leukoencephalopathy with subcortical cysts 1, rapamycin-insensitive companion of mammalian target of rapamycin, epithelial growth factor receptor, SWI/SNF Related, Matrix Associated, Actin Dependent Regulator Of Chromatin, Subfamily A, Member 4, cyclin-dependent kinase inhibitor 2B, phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit α, Kirsten rat sarcoma viral oncogene homolog, Erb-B2 receptor tyrosine kinase 2, Serine/Threonine Kinase 11, and NK2 Homeobox 1. Sequencing results led to a change in management in 7 of 49 cases (14.3%). CONCLUSION Extended next-generation sequencing was performed successfully in 41 (83.7%) cases of NSCLC using a range of pathology specimens. Testing had the potential to affect treatment decisions in selected patients.
Collapse
Affiliation(s)
- David M DiBardino
- Division of Pulmonary, Allergy and Critical Care Medicine, Columbia University College of Physicians and Surgeons, New York, NY.
| | - Anjali Saqi
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY
| | | | | | | | | | | | - Mark Stoopler
- Division of Hematology/Oncology, Columbia University College of Physicians and Surgeons, New York, NY
| | - William A Bulman
- Division of Pulmonary, Allergy and Critical Care Medicine, Columbia University College of Physicians and Surgeons, New York, NY
| |
Collapse
|
110
|
Abstract
INTRODUCTION Chronic granulomatous disease (CGD) is a primary immunodeficiency characterized by recurrent, life-threatening bacterial and fungal infections of the skin, the airways, the lymph nodes, the liver, the brain and the bones. Frequently found pathogens are Staphylococcus aureus, Aspergillus species, Klebsiella species, Burkholderia cepacia, Serratia marcescens and Salmonella species. SOURCES OF DATA CGD is a rare (∼1:250 000 individuals) disease caused by mutations in any one of the five components of the NADPH oxidase in phagocytic leucocytes. This enzyme generates superoxide and is essential for intracellular killing of pathogens by phagocytes. AREAS OF AGREEMENT CGD patients suffer not only from life-threatening infections, but also from excessive inflammatory reactions. AREAS OF CONTROVERSY Neither the cause of these inflammatory reactions nor the way to treat them is clear. AREAS TIMELY FOR DEVELOPING RESEARCH Patient selection for and timing of bone marrow transplantation along with gene therapy.
Collapse
Affiliation(s)
- Dirk Roos
- Department of Blood Cell Research, Sanquin Blood Supply Organisation, Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
111
|
|
112
|
Roy-Chowdhuri S, Aisner DL, Allen TC, Beasley MB, Borczuk A, Cagle PT, Capelozzi V, Dacic S, da Cunha Santos G, Hariri LP, Kerr KM, Lantuejoul S, Mino-Kenudson M, Moreira A, Raparia K, Rekhtman N, Sholl L, Thunnissen E, Tsao MS, Vivero M, Yatabe Y. Biomarker Testing in Lung Carcinoma Cytology Specimens: A Perspective From Members of the Pulmonary Pathology Society. Arch Pathol Lab Med 2016; 140:1267-1272. [PMID: 27081878 DOI: 10.5858/arpa.2016-0091-sa] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The advent of targeted therapy in lung cancer has heralded a paradigm shift in the practice of cytopathology with the need for accurately subtyping lung carcinoma, as well as providing adequate material for molecular studies, to help guide clinical and therapeutic decisions. The variety and versatility of cytologic-specimen preparations offer significant advantages to molecular testing; however, they frequently remain underused. Therefore, evaluating the utility and adequacy of cytologic specimens is critical, not only from a lung cancer diagnosis standpoint but also for the myriad ancillary studies that are necessary to provide appropriate clinical management. A large fraction of lung cancers are diagnosed by aspiration or exfoliative cytology specimens, and thus, optimizing strategies to triage and best use the tissue for diagnosis and biomarker studies forms a critical component of lung cancer management. This review focuses on the opportunities and challenges of using cytologic specimens for molecular diagnosis of lung cancer and the role of cytopathology in the molecular era.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Yasushi Yatabe
- From the Department of Pathology, University of Texas MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); the Department of Pathology, University of Colorado Cancer Center, Denver (Dr Aisner); the Department of Pathology, University of Texas Medical Branch, Galveston (Dr Allen); the Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York (Dr Beasley); the Department of Pathology, Weill Cornell Medical College, New York (Drs Borczuk and Cagle); the Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas (Dr Cagle); the Department of Pathology, University of Sao Paulo Medical School, Sao Paulo, Brazil (Dr Capelozzi); the Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Pathology, University Health Network, Princess Margaret Cancer Centre, and the University of Toronto, Toronto, Ontario, Canada (Drs da Cunha Santos and Tsao); the Department of Pathology, Massachusetts General Hospital, and Harvard Medical School, Boston (Drs Hariri and Mino-Kenudson); the Department of Pathology, Aberdeen University Medical School, and Aberdeen Royal Infirmary, Foresterhill, Aberdeen, Scotland, United Kingdom (Dr Kerr); the Department of Biopathology, Centre Léon Bérard, Lyon, and J Fourier University, Institut National de la Santé et de la Recherche Médicale-Institut Albert Bonniot, Grenoble, France (Dr Lantuejoul); the Department of Pathology, New York University, New York (Dr Moreira); the Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois (Dr Raparia); the Department of Pathology, Memorial Sloan Kettering Cancer Center, New York (Dr Rekhtman); the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston (Drs Sholl and Vivero); the Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands (Dr Thunnissen); and the Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan (Dr Yatabe)
| |
Collapse
|
113
|
Zhang N, Liu H, Yue G, Zhang Y, You J, Wang H. Molecular Heterogeneity of Ewing Sarcoma as Detected by Ion Torrent Sequencing. PLoS One 2016; 11:e0153546. [PMID: 27077911 PMCID: PMC4831808 DOI: 10.1371/journal.pone.0153546] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 03/31/2016] [Indexed: 12/26/2022] Open
Abstract
Ewing sarcoma (ES) is the second most common malignant bone and soft tissue tumor in children and adolescents. Despite advances in comprehensive treatment, patients with ES metastases still suffer poor outcomes, thus, emphasizing the need for detailed genetic profiles of ES patients to identify suitable molecular biomarkers for improved prognosis and development of effective and targeted therapies. In this study, the next generation sequencing Ion AmpliSeq™ Cancer Hotspot Panel v2 was used to identify cancer-related gene mutations in the tissue samples from 20 ES patients. This platform targeted 207 amplicons of 2800 loci in 50 cancer-related genes. Among the 20 tissue specimens, 62 nonsynonymous hotspot mutations were identified in 26 cancer-related genes, revealing the molecular heterogeneity of ES. Among these, five novel mutations in cancer-related genes (KDR, STK11, MLH1, KRAS, and PTPN11) were detected in ES, and these mutations were confirmed with traditional Sanger sequencing. ES patients with KDR, STK11, and MLH1 mutations had higher Ki-67 proliferation indices than the ES patients lacking such mutations. Notably, more than half of the ES patients harbored one or two possible ‘druggable’ mutations that have been previously linked to a clinical cancer treatment option. Our results provided the foundation to not only elucidate possible mechanisms involved in ES pathogenesis but also indicated the utility of Ion Torrent sequencing as a sensitive and cost-effective tool to screen key oncogenes and tumor suppressors in order to develop personalized therapy for ES patients.
Collapse
Affiliation(s)
- Nana Zhang
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.,Department of Pathology, Peking University Third Hospital, Beijing, China
| | - Haijing Liu
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.,Department of Pathology, Peking University Third Hospital, Beijing, China
| | - Guanjun Yue
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.,Department of Pathology, Peking University Third Hospital, Beijing, China
| | - Yan Zhang
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.,Department of Pathology, Peking University Third Hospital, Beijing, China
| | - Jiangfeng You
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.,Department of Pathology, Peking University Third Hospital, Beijing, China
| | - Hua Wang
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.,Department of Pathology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
114
|
Building a Robust Tumor Profiling Program: Synergy between Next-Generation Sequencing and Targeted Single-Gene Testing. PLoS One 2016; 11:e0152851. [PMID: 27043212 PMCID: PMC4820127 DOI: 10.1371/journal.pone.0152851] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 03/21/2016] [Indexed: 12/25/2022] Open
Abstract
Next-generation sequencing (NGS) is a powerful platform for identifying cancer mutations. Routine clinical adoption of NGS requires optimized quality control metrics to ensure accurate results. To assess the robustness of our clinical NGS pipeline, we analyzed the results of 304 solid tumor and hematologic malignancy specimens tested simultaneously by NGS and one or more targeted single-gene tests (EGFR, KRAS, BRAF, NPM1, FLT3, and JAK2). For samples that passed our validated tumor percentage and DNA quality and quantity thresholds, there was perfect concordance between NGS and targeted single-gene tests with the exception of two FLT3 internal tandem duplications that fell below the stringent pre-established reporting threshold but were readily detected by manual inspection. In addition, NGS identified clinically significant mutations not covered by single-gene tests. These findings confirm NGS as a reliable platform for routine clinical use when appropriate quality control metrics, such as tumor percentage and DNA quality cutoffs, are in place. Based on our findings, we suggest a simple workflow that should facilitate adoption of clinical oncologic NGS services at other institutions.
Collapse
|
115
|
Dong L, Wang W, Li A, Kansal R, Chen Y, Chen H, Li X. Clinical Next Generation Sequencing for Precision Medicine in Cancer. Curr Genomics 2016; 16:253-63. [PMID: 27006629 PMCID: PMC4765520 DOI: 10.2174/1389202915666150511205313] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Revised: 05/05/2015] [Accepted: 05/08/2015] [Indexed: 12/23/2022] Open
Abstract
Rapid adoption of next generation sequencing (NGS) in genomic medicine has been driven
by low cost, high throughput sequencing and rapid advances in our understanding of the genetic bases
of human diseases. Today, the NGS method has dominated sequencing space in genomic research, and
quickly entered clinical practice. Because unique features of NGS perfectly meet the clinical reality
(need to do more with less), the NGS technology is becoming a driving force to realize the dream of precision medicine.
This article describes the strengths of NGS, NGS panels used in precision medicine, current applications of NGS in cytology,
and its challenges and future directions for routine clinical use.
Collapse
Affiliation(s)
- Ling Dong
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, 650 Charles Young Dr., Los Angeles, CA 90095, USA
| | - Wanheng Wang
- Shanxi Guoxin Caregeno Medical Laboratories (MedLab), 7 Jiahua St., Taiyuan, Shanxi Province, China 030006
| | - Alvin Li
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, 650 Charles Young Dr., Los Angeles, CA 90095, USA
| | - Rina Kansal
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, 650 Charles Young Dr., Los Angeles, CA 90095, USA
| | - Yuhan Chen
- Heilongjiang University of Chinese Medicine, No. 24 Heping Road, Harbin, China 150040
| | - Hong Chen
- Qiqihaer First Hospital, No.30, Gongyuan Road, Qiqihar, Heilongjiang Province, China 161005
| | - Xinmin Li
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, 650 Charles Young Dr., Los Angeles, CA 90095, USA
| |
Collapse
|
116
|
Treece AL, Montgomery ND, Patel NM, Civalier CJ, Dodd LG, Gulley ML, Booker JK, Weck KE. FNA smears as a potential source of DNA for targeted next-generation sequencing of lung adenocarcinomas. Cancer Cytopathol 2016; 124:406-14. [PMID: 26882436 DOI: 10.1002/cncy.21699] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 01/04/2016] [Accepted: 01/04/2016] [Indexed: 11/12/2022]
Abstract
BACKGROUND Diff-Quik-stained fine-needle aspiration (FNA) smears and touch preparations from biopsies represent alternative specimens for molecular testing when cell block or biopsy material is insufficient. This study describes the use of these samples for targeted next-generation sequencing (NGS) of primary and metastatic lung adenocarcinoma and reports the DNA quality and success rates of FNA smears versus other specimens from 1 year of clinical use. METHODS A validation set of 10 slides from 9 patients with prior clinical epidermal growth factor receptor (EGFR) Sanger sequencing and KRAS pyrosequencing (5 KRAS-positive/EGFR-negative and 4 KRAS-negative/EGFR-negative) underwent DNA extraction, quality assessment, and targeted NGS. Subsequently, lung adenocarcinoma specimens submitted for NGS solid tumor mutation panel testing in 1 calendar year (60 biopsies, 57 resections, 33 FNA cell blocks, 12 FNA smears, and 10 body fluid cell blocks) were reviewed for specimen adequacy, sequencing success, and DNA quality. RESULTS All 10 validation samples met the DNA quality threshold (delta Ct threshold < 8; range, -2.2 to 4.9) and yielded 0.5 to 22 μg of DNA. The KRAS and EGFR mutation status from FNA smears according to NGS was concordant with previous clinical testing for all 10 samples. In the 1-year review, FNA smears were 100% successful, and this suggested a performance equivalent to or better than the performance of established specimen types, including FNA cell blocks. DNA quality according to ΔCt was significantly better with FNA smears versus biopsies, resections, and FNA cell blocks. CONCLUSIONS FNA smears of lung adenocarcinomas are high-quality alternative specimens for a targeted NGS panel with a high success rate in clinical practice. Cancer Cytopathol 2016;124:406-14. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- Amanda L Treece
- Pathology and Laboratory Medicine, Children's Hospital Colorado, Aurora, Colorado
| | - Nathan D Montgomery
- Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Nirali M Patel
- Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Chris J Civalier
- Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Leslie G Dodd
- Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Margaret L Gulley
- Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jessica K Booker
- Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Karen E Weck
- Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
117
|
Han HS, Magliocco AM. Molecular Testing and the Pathologist's Role in Clinical Trials of Breast Cancer. Clin Breast Cancer 2016; 16:166-79. [PMID: 27103546 DOI: 10.1016/j.clbc.2016.02.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 01/11/2016] [Accepted: 02/03/2016] [Indexed: 01/26/2023]
Abstract
Molecular characterization of breast cancer is pivotal for identifying new molecular targets and determining the appropriate treatment choices. Advances in molecular profiling technology have given greater insight into this heterogeneous disease, over and above hormone receptor and human epidermal growth factor receptor 2 status. Agents targeting recently characterized molecular biomarkers are under clinical development; the success of these targeted agents is likely to depend on identifying the patient population most likely to benefit. Therefore, clinical trials of breast cancer often require prescreening for, or stratification by, relevant molecular markers or exploratory analyses of biomarkers that can predict or monitor the response to treatment. Consequently, the role of the pathologist has become increasingly important. The key considerations for pathologists include tissue availability, ownership of archival tissue, type of diagnostic/biomarker test required, method of sample processing, concordance between different tests and testing centers, and tumor heterogeneity. In the present review, we explore how pathology is used in current clinical trials of breast cancer and describe the various technologies available for molecular testing. Furthermore, the factors required for the successful application of pathology in clinical trials of breast cancer and the issues that can arise and how these can be circumvented are discussed.
Collapse
Affiliation(s)
- Hyo Sook Han
- Department of Women's Oncology, Moffitt Cancer Center, Tampa, FL
| | | |
Collapse
|
118
|
Goswami RS, Luthra R, Singh RR, Patel KP, Routbort MJ, Aldape KD, Yao H, Dang HD, Barkoh BA, Manekia J, Medeiros LJ, Roy-Chowdhuri S, Stewart J, Broaddus RR, Chen H. Identification of Factors Affecting the Success of Next-Generation Sequencing Testing in Solid Tumors. Am J Clin Pathol 2016; 145:222-37. [PMID: 27124905 DOI: 10.1093/ajcp/aqv023] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Clinical laboratories are rapidly implementing next-generation sequencing (NGS) tests for mutation analysis, but there are few guidelines regarding sample quality for successful results. METHODS We aimed to establish tissue quality parameters for successful NGS in solid tumors and to improve NGS performance. RESULTS Analysis of 614 clinical cases tested in 2013 using a 50-gene hotspot mutation panel identified the major cause for unsuccessful NGS analysis was DNA less than 10 ng (91%, 67/74) associated with extremely small and low cellularity samples. High success rates were associated with resection procedures (333/342, 97%) and biopsied tumor larger than 10 mm(2) (77/77, 100%). NGS can be successfully performed on bone specimens processed with formic acid-based decalcification procedures (8/11, 73%). Tumor type and paraffin block age did not affect success. We demonstrated that NGS can be carried out on samples with less than 10 ng DNA. Analysis of 408 cases tested in 2014 using an optimized workflow showed improved NGS success rates from 88% to 95% (387/408) with pronounced improvement among tiny (<10 mm(2)) samples (from 76% to 94%) as well as cytology samples (from 58% to 87%). CONCLUSIONS Identifying preanalytical tissue factors allows us to improve NGS performance and to successfully test tumors obtained from minimally invasive procedures.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hui Yao
- Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston
| | | | | | | | | | | | | | | | | |
Collapse
|
119
|
Ballester LY, Luthra R, Kanagal-Shamanna R, Singh RR. Advances in clinical next-generation sequencing: target enrichment and sequencing technologies. Expert Rev Mol Diagn 2016; 16:357-72. [PMID: 26680590 DOI: 10.1586/14737159.2016.1133298] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The huge parallel sequencing capabilities of next generation sequencing technologies have made them the tools of choice to characterize genomic aberrations for research and diagnostic purposes. For clinical applications, screening the whole genome or exome is challenging owing to the large genomic area to be sequenced, associated costs, complexity of data, and lack of known clinical significance of all genes. Consequently, routine screening involves limited markers with established clinical relevance. This process, referred to as targeted genome sequencing, requires selective enrichment of the genomic areas comprising these markers via one of several primer or probe-based enrichment strategies, followed by sequencing of the enriched genomic areas. Here, the authors review current target enrichment approaches and next generation sequencing platforms, focusing on the underlying principles, capabilities, and limitations of each technology along with validation and implementation for clinical testing.
Collapse
Affiliation(s)
- Leomar Y Ballester
- a Department of Pathology and Immunology , Baylor College of Medicine , Houston , TX , USA
| | - Rajyalakshmi Luthra
- b Department of Hematopathology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Rashmi Kanagal-Shamanna
- b Department of Hematopathology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Rajesh R Singh
- b Department of Hematopathology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|
120
|
Lih CJ, Sims DJ, Harrington RD, Polley EC, Zhao Y, Mehaffey MG, Forbes TD, Das B, Walsh WD, Datta V, Harper KN, Bouk CH, Rubinstein LV, Simon RM, Conley BA, Chen AP, Kummar S, Doroshow JH, Williams PM. Analytical Validation and Application of a Targeted Next-Generation Sequencing Mutation-Detection Assay for Use in Treatment Assignment in the NCI-MPACT Trial. J Mol Diagn 2016; 18:51-67. [PMID: 26602013 PMCID: PMC5812775 DOI: 10.1016/j.jmoldx.2015.07.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 07/08/2015] [Accepted: 07/22/2015] [Indexed: 12/21/2022] Open
Abstract
Robust and analytically validated assays are essential for clinical studies. We outline an analytical validation study of a targeted next-generation sequencing mutation-detection assay used for patient selection in the National Cancer Institute Molecular Profiling-Based Assignment of Cancer Therapy (NCI-MPACT) trial (NCT01827384). Using DNA samples from normal or tumor cell lines and xenografts with known variants, we assessed the sensitivity, specificity, and reproducibility of the NCI-MPACT assay in five variant types: single-nucleotide variants (SNVs), SNVs at homopolymeric (HP) regions (≥3 identical bases), small insertions/deletions (indels), large indels (gap ≥4 bp), and indels at HP regions. The assay achieved sensitivities of 100% for 64 SNVs, nine SNVs at HP regions, and 11 large indels, 83.33% for six indels, and 93.33% for 15 indels at HP regions. Zero false positives (100% specificity) were found in 380 actionable mutation loci in 96 runs of haplotype map cells. Reproducibility analysis showed 96.3% to 100% intraoperator and 98.1% to 100% interoperator mean concordance in detected variants and 100% reproducibility in treatment selection. To date, 38 tumors have been screened, 34 passed preanalytical quality control, and 18 had actionable mutations for treatment assignment. The NCI-MPACT assay is well suited for its intended investigational use and can serve as a template for developing next-generation sequencing assays for other cancer clinical trial applications.
Collapse
Affiliation(s)
- Chih-Jian Lih
- Molecular Characterization and Clinical Assay Development Laboratory, Leidos Biomedical Research Inc., and Frederick National Laboratory for Cancer Research, Frederick, Maryland.
| | - David J Sims
- Molecular Characterization and Clinical Assay Development Laboratory, Leidos Biomedical Research Inc., and Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Robin D Harrington
- Molecular Characterization and Clinical Assay Development Laboratory, Leidos Biomedical Research Inc., and Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Eric C Polley
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Yingdong Zhao
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Michele G Mehaffey
- Molecular Characterization and Clinical Assay Development Laboratory, Leidos Biomedical Research Inc., and Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Thomas D Forbes
- Molecular Characterization and Clinical Assay Development Laboratory, Leidos Biomedical Research Inc., and Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Biswajit Das
- Molecular Characterization and Clinical Assay Development Laboratory, Leidos Biomedical Research Inc., and Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - William D Walsh
- Molecular Characterization and Clinical Assay Development Laboratory, Leidos Biomedical Research Inc., and Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Vivekananda Datta
- Molecular Characterization and Clinical Assay Development Laboratory, Leidos Biomedical Research Inc., and Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Kneshay N Harper
- Molecular Characterization and Clinical Assay Development Laboratory, Leidos Biomedical Research Inc., and Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Courtney H Bouk
- Molecular Characterization and Clinical Assay Development Laboratory, Leidos Biomedical Research Inc., and Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Lawrence V Rubinstein
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Richard M Simon
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Barbara A Conley
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Alice P Chen
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Shivaani Kummar
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Paul M Williams
- Molecular Characterization and Clinical Assay Development Laboratory, Leidos Biomedical Research Inc., and Frederick National Laboratory for Cancer Research, Frederick, Maryland; Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
121
|
Mahon S. Management of Individuals With a Mutation in the Ataxia Telangiectasia Mutated Gene. Oncol Nurs Forum 2016; 43:114-7. [DOI: 10.1188/16.onf.114-117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
122
|
Wei S, Lieberman D, Morrissette JJD, Baloch ZW, Roth DB, McGrath C. Using "residual" FNA rinse and body fluid specimens for next-generation sequencing: An institutional experience. Cancer Cytopathol 2015; 124:324-9. [PMID: 26682952 DOI: 10.1002/cncy.21666] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 11/10/2015] [Accepted: 11/11/2015] [Indexed: 01/17/2023]
Abstract
BACKGROUND Tissue specimens are typically considered optimal for molecular testing; however, in the current era of personalized medicine, cytopathology specimens are increasingly recognized as potential sources for molecular testing. This is often accomplished by using cell block specimens and/or fine-needle aspiration (FNA) smear preparations. In this study, the authors investigated the feasibility, performance, and quality of "residual" FNA rinse and body effusion fluids used for next-generation sequencing (NGS). METHODS Sequence data were generated from 17 malignancies in 16 patients from 13 FNA (10 lymph nodes, 1 lung, and 2 bone lesions) and 4 effusion (3 pleural and 1 pericardial) specimens. Malignancies included carcinomas (lung, breast, ovarian, and unknown primary), melanoma, and myeloma. Paired NGS testing was performed in 7 patients who had surgical biopsy or cell block specimens available. Routinely processed residual FNA rinse material and body fluids were used for DNA extraction and NGS (targeted gene panel). RESULTS NGS was successfully performed on all 17 specimens. A significant amount of DNA was obtained from the residual FNA rinse (176.3 ng/μL) compared with the paired cell block slides (10.6 ng/μL). Two of the 10 lung adenocarcinomas (20%) demonstrated epidermal growth factor receptor (EGFR) mutations, including 1 leucine-to-arginine substitution at codon 858 (L858R) in exon 21 and 1 codon 2235_2249 deletion (resulting in an in-frame deletion of 5 amino acids from position 746 to 750 [glutamic acid, leucine, arginine, glutamic acid, and alanine]; E746_A750del) in exon 19. Three KRAS [Kirsten rat sarcoma viral oncogene homolog] mutations, 1 BRAF (v-Raf murine sarcoma viral oncogene homolog B1) mutation, and 1 NRAS (neuroblastoma RAS viral oncogene homolog) mutation were identified in the remaining lung adenocarcinomas. Patients who underwent paired testing demonstrated 100% concordant mutations. CONCLUSIONS Targeted NGS can be performed on residual FNA rinse and body fluid specimens. This approach is particularly important when a paucicellular cell block or biopsy specimen is encountered. Cancer Cytopathol 2016;124:324-29. © 2015 American Cancer Society.
Collapse
Affiliation(s)
- Shuanzeng Wei
- Department of Pathology and Laboratory Medicine, Center for Personalized Diagnostics, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - David Lieberman
- Department of Pathology and Laboratory Medicine, Center for Personalized Diagnostics, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jennifer J D Morrissette
- Department of Pathology and Laboratory Medicine, Center for Personalized Diagnostics, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Zubair W Baloch
- Department of Pathology and Laboratory Medicine, Center for Personalized Diagnostics, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - David B Roth
- Department of Pathology and Laboratory Medicine, Center for Personalized Diagnostics, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Cindy McGrath
- Department of Pathology and Laboratory Medicine, Center for Personalized Diagnostics, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
123
|
Roy-Chowdhuri S, Chow CW, Kane MK, Yao H, Wistuba II, Krishnamurthy S, Stewart J, Staerkel G. Optimizing the DNA yield for molecular analysis from cytologic preparations. Cancer Cytopathol 2015; 124:254-60. [PMID: 26630358 DOI: 10.1002/cncy.21664] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 11/04/2015] [Accepted: 11/05/2015] [Indexed: 01/21/2023]
Abstract
BACKGROUND Cytology smears and cytospin preparations are increasingly being used for molecular testing. With these limited samples, optimizing tissue extraction to maximize the DNA yield is, therefore, critical. This study examined 2 common methods of tissue extraction and compared DNA yields from different types of glass slides. METHODS The H226 lung cancer cell line and 5 clinical samples of cellular effusions were used to prepare Diff-Quik-stained cytospins on 4 types of glass slides: fully frosted (FF), nonfrosted (NF), positively charged (PC), and silane-coated (SC). Tissue extraction was performed by either scalpel-blade scraping or cell lifting with the Pinpoint Slide DNA Isolation System (Zymo Research). DNA was extracted with the QIAamp DNA Mini Kit (Qiagen) and was quantified with the Quant-iT PicoGreen Kit (Life Technologies). RESULTS The DNA yield in cell-line cytospins was significantly lower from FF slides versus NF, PC, and SC slides with both scraping and cell-lifting methods. In addition, scraping yielded significantly more DNA than cell lifting (P = .005). DNA yields from 5 clinical effusion cases with FF and NF slides showed results similar to the results for cell-line samples, with scraping consistently yielding more DNA than cell lifting and with NF slides outperforming FF slides. CONCLUSIONS Optimizing the DNA yield extracted from cytology specimens maximizes the chances of successful molecular testing and is critical in cases of low or marginal cellularity. This study demonstrates the following: 1) scraping yields more DNA than cell lifting, and 2) NF slides yield more DNA than FF slides.
Collapse
Affiliation(s)
- Sinchita Roy-Chowdhuri
- Cytopathology Section, Department of Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chi-Wan Chow
- Department of Translational Molecular Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mary K Kane
- Cytopathology Section, Department of Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hui Yao
- Department of Translational Molecular Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Savitri Krishnamurthy
- Cytopathology Section, Department of Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John Stewart
- Cytopathology Section, Department of Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gregg Staerkel
- Cytopathology Section, Department of Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
124
|
Pusztaszeri MP, García JJ, Faquin WC. Salivary gland FNA: New markers and new opportunities for improved diagnosis. Cancer Cytopathol 2015; 124:307-16. [DOI: 10.1002/cncy.21649] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 10/21/2015] [Accepted: 10/22/2015] [Indexed: 01/11/2023]
Affiliation(s)
| | - Joaquín J. García
- Department of Laboratory Medicine and Pathology; Mayo Clinic; Rochester New York
| | - William C. Faquin
- Department of Pathology; Massachusetts General Hospital; Boston Massachusetts
- Harvard Medical School; Boston Massachusetts
| |
Collapse
|
125
|
Roy-Chowdhuri S, de Melo Gagliato D, Routbort MJ, Patel KP, Singh RR, Broaddus R, Lazar AJ, Sahin A, Alvarez RH, Moulder S, Wheler JJ, Janku F, Gonzalez-Angulo AM, Chavez-MacGregor M, Valero V, Ueno NT, Mills G, Mendelsohn J, Yao H, Aldape K, Luthra R, Meric-Bernstam F. Multigene clinical mutational profiling of breast carcinoma using next-generation sequencing. Am J Clin Pathol 2015; 144:713-21. [PMID: 26486734 DOI: 10.1309/ajcpwdeqycyc92jq] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVES The advent of next-generation sequencing (NGS) platforms in the realm of clinical molecular diagnostics provides multigene mutational profiling through massively parallel sequencing. METHODS We analyzed 415 breast carcinoma samples from 354 patients using NGS in known hotspots of 46 commonly known cancer-causing genes. RESULTS A total of 281 somatic nonsynonymous mutations were detected in 62.1% of patients. TP53 was most frequently mutated (38.8%), followed by PIK3CA (31.7%), AKT1 (6%), and ATM (3.9%), with other mutations detected at a lower frequency. When stratified into clinically relevant therapeutic groups (estrogen receptor [ER]/progesterone receptor [PR]+ human epidermal growth factor receptor 2 [HER2]-, ER/PR+HER2+, ER/PR-HER2+, ER/PR/HER2-), each group showed distinct mutational profiles. The ER/PR+HER2- tumors (n = 132) showed the highest frequency of PIK3CA mutations (38%), while the triple-negative tumors (n = 64) had a significantly higher number of TP53 mutations (62%). Of the 61 patients tested for both primary and metastatic tumors, concordant results were seen in 47 (77%) patients, while 13 patients showed additional mutations in the metastasis. CONCLUSIONS Our results indicate that breast cancers may harbor potentially actionable mutations for targeted therapeutics. Therefore, NGS-based mutational profiling can provide useful information that can guide targeted cancer therapy.
Collapse
|
126
|
Piqueret-Stephan L, Marcaillou C, Reyes C, Honoré A, Letexier M, Gentien D, Droin N, Lacroix L, Scoazec JY, Vielh P. Massively parallel DNA sequencing from routinely processed cytological smears. Cancer Cytopathol 2015; 124:241-53. [DOI: 10.1002/cncy.21639] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 10/03/2015] [Accepted: 10/05/2015] [Indexed: 11/07/2022]
Affiliation(s)
| | | | - Cécile Reyes
- Genomic Platform, Translational Research Department; Institut Curie; Paris France
| | - Aurélie Honoré
- Analyse Moléculaire, Modélisation et Imagerie de la maladie Cancéreuse (AMMICa) CNRS Unité Mixte de Service 3655, INSERM US23, Paris Sud University), Translational Research Laboratory; Gustave Roussy Villejuif France
| | | | - David Gentien
- Genomic Platform, Translational Research Department; Institut Curie; Paris France
| | - Nathalie Droin
- Gustave Roussy; Villejuif France
- Unit 1009, INSERM; Villejuif France
| | - Ludovic Lacroix
- Analyse Moléculaire, Modélisation et Imagerie de la maladie Cancéreuse (AMMICa) CNRS Unité Mixte de Service 3655, INSERM US23, Paris Sud University), Translational Research Laboratory; Gustave Roussy Villejuif France
- Department of Biopathology; Gustave Roussy Villejuif France
| | - Jean-Yves Scoazec
- Analyse Moléculaire, Modélisation et Imagerie de la maladie Cancéreuse (AMMICa) CNRS Unité Mixte de Service 3655, INSERM US23, Paris Sud University), Translational Research Laboratory; Gustave Roussy Villejuif France
- Department of Biopathology; Gustave Roussy Villejuif France
| | - Philippe Vielh
- Gustave Roussy; Villejuif France
- Unité Mixte de Recherche 981, INSERM; Villejuif France
- Analyse Moléculaire, Modélisation et Imagerie de la maladie Cancéreuse (AMMICa) CNRS Unité Mixte de Service 3655, INSERM US23, Paris Sud University), Translational Research Laboratory; Gustave Roussy Villejuif France
- Department of Biopathology; Gustave Roussy Villejuif France
| |
Collapse
|
127
|
Abstract
Personalized therapy concepts in which the active agent is adapted to genetic alterations in the tumor of the patient, have in recent years led to a paradigm shift in oncology. A comprehensive molecular diagnostic tumor characterization is therefore essential before initiating therapy in order to select the optimal therapy for the patient. The continuously increasing number of genetic alterations which can be treated and known resistance mechanisms together with limited availability of test material represents a completely new challenge for molecular diagnostics. The possibility of being able to determine mutations, translocations and changes in the number of copies not only from paraffin-embedded tumor tissue but also from cytological material and even circulating tumor DNA, substantially extends the diagnostic options.
Collapse
|
128
|
Luthra R, Chen H, Roy-Chowdhuri S, Singh RR. Next-Generation Sequencing in Clinical Molecular Diagnostics of Cancer: Advantages and Challenges. Cancers (Basel) 2015; 7:2023-36. [PMID: 26473927 PMCID: PMC4695874 DOI: 10.3390/cancers7040874] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 09/21/2015] [Accepted: 10/01/2015] [Indexed: 11/16/2022] Open
Abstract
The application of next-generation sequencing (NGS) to characterize cancer genomes has resulted in the discovery of numerous genetic markers. Consequently, the number of markers that warrant routine screening in molecular diagnostic laboratories, often from limited tumor material, has increased. This increased demand has been difficult to manage by traditional low- and/or medium-throughput sequencing platforms. Massively parallel sequencing capabilities of NGS provide a much-needed alternative for mutation screening in multiple genes with a single low investment of DNA. However, implementation of NGS technologies, most of which are for research use only (RUO), in a diagnostic laboratory, needs extensive validation in order to establish Clinical Laboratory Improvement Amendments (CLIA) and College of American Pathologists (CAP)-compliant performance characteristics. Here, we have reviewed approaches for validation of NGS technology for routine screening of tumors. We discuss the criteria for selecting gene markers to include in the NGS panel and the deciding factors for selecting target capture approaches and sequencing platforms. We also discuss challenges in result reporting, storage and retrieval of the voluminous sequencing data and the future potential of clinical NGS.
Collapse
Affiliation(s)
- Rajyalakshmi Luthra
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 8515 Fannin Street, Houston, TX 77054, USA.
| | - Hui Chen
- Department of Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX-77030, USA.
| | - Sinchita Roy-Chowdhuri
- Department of Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX-77030, USA.
| | - R Rajesh Singh
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 8515 Fannin Street, Houston, TX 77054, USA.
| |
Collapse
|
129
|
Vigliar E, Malapelle U, de Luca C, Bellevicine C, Troncone G. Challenges and opportunities of next-generation sequencing: a cytopathologist's perspective. Cytopathology 2015; 26:271-83. [DOI: 10.1111/cyt.12265] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2015] [Indexed: 01/14/2023]
Affiliation(s)
- E. Vigliar
- Pathology Division; Department of Public Health; University of Naples Federico II; Naples Italy
| | - U. Malapelle
- Pathology Division; Department of Public Health; University of Naples Federico II; Naples Italy
| | - C. de Luca
- Pathology Division; Department of Public Health; University of Naples Federico II; Naples Italy
| | - C. Bellevicine
- Pathology Division; Department of Public Health; University of Naples Federico II; Naples Italy
| | - G. Troncone
- Pathology Division; Department of Public Health; University of Naples Federico II; Naples Italy
| |
Collapse
|
130
|
Dumur CI, Almenara JA, Powers CN, Ferreira-Gonzalez A. Quality control material for the detection of somatic mutations in fixed clinical specimens by next-generation sequencing. Diagn Pathol 2015; 10:169. [PMID: 26376646 PMCID: PMC4573924 DOI: 10.1186/s13000-015-0403-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 08/28/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Targeted next generation sequencing (NGS) technology to assess the mutational status of multiple genes on formalin-fixed, paraffin embedded (FFPE) tumors is rapidly being adopted in clinical settings, where quality control (QC) practices are required. Establishing reliable FFPE QC materials for NGS can be challenging and/or expensive. Here, we established a reliable and cost-effective FFPE QC material for routine utilization in the Ion AmpliSeq™ Cancer Hotspot Panel v2 (CHP2) assay. METHODS The performance characteristics of the CHP2 assay were determined by sequencing various cell line mixtures and 55 different FFPE tumors on the Ion Torrent PGM platform. A FFPE QC material was prepared from a mixture of cell lines derived from different cancers, comprising single nucleotide variants and small deletions on actionable genes at different allelic frequencies. RESULTS The CHP2 assay performed with high precision and sensitivity when custom variant calling pipeline parameters where established. In addition, all expected somatic variants in the QC material were consistently called at variant frequencies ranging from 9.1 % (CV = 11.1 %) to 37.9 % (CV = 2.8 %). CONCLUSIONS The availability of a reliable and cost-effective QC material is instrumental in assessing the performance of this or any targeted NGS assay that detects somatic variants in fixed solid tumor specimens.
Collapse
Affiliation(s)
- Catherine I Dumur
- Department of Pathology, Virginia Commonwealth University, Clinical Support Center, Room 247, 403 North 13th Street, Richmond, VA, 23298, USA.
| | - Jorge A Almenara
- Department of Pathology, Virginia Commonwealth University, Clinical Support Center, Room 247, 403 North 13th Street, Richmond, VA, 23298, USA.
| | - Celeste N Powers
- Department of Pathology, Virginia Commonwealth University, Clinical Support Center, Room 247, 403 North 13th Street, Richmond, VA, 23298, USA.
| | - Andrea Ferreira-Gonzalez
- Department of Pathology, Virginia Commonwealth University, Clinical Support Center, Room 247, 403 North 13th Street, Richmond, VA, 23298, USA.
| |
Collapse
|
131
|
Courtade-Saïdi M, Fleury Feith J. [Pre-analytical quality in fluid samples cytopathology: Results of a survey from the French Society of Clinical Cytology]. Ann Pathol 2015; 35:378-86. [PMID: 26372499 DOI: 10.1016/j.annpat.2015.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 08/05/2015] [Accepted: 08/05/2015] [Indexed: 11/18/2022]
Abstract
INTRODUCTION The pre-analytical step includes sample collection, preparation, transportation and storage in the pathology unit where the diagnosis is performed. The pathologist ensures that pre-analytical conditions are in line with expectations. The lack of standardization for handling cytological samples makes this pre-analytical step difficult to harmonize. Moreover, this step depends on the nature of the sample: fresh liquid or fixed material, air-dried smears, liquid-based cytology. The aim of the study was to review the different practices in French structures of pathology on the pre-analytical phase concerning cytological fluids such as broncho-alveolar lavage (BALF), serous fluids and urine. METHODS A survey was conducted on the basis of the pre-analytical chapter of the ISO 15189 and sent to 191 French pathological structures (105 public and 86 private). RESULTS Fifty-six laboratories replied to the survey. Ninety-five per cent have a computerized management system and 70% a manual on sample handling. The general instructions requested for the patients and sample identification were highly correctly filled with a short time routing and additional tests prescription. By contrast, information are variable concerning the clinical information requested and the type of tubes for collecting fluids and the volumes required as well as the actions taken in case of non-conformity. For the specific items concerning BALF, serous fluids and urine, this survey has shown a great heterogeneity according to sample collection, fixation and of clinical information. CONCLUSION This survey demonstrates that the pre-analytical quality for BALF, serous fluids and urine is not optimal and that some corrections of the practices are recommended with a standardization of numerous steps in order to increase the reproducibility of additional tests such as immunocytochemistry, cytogenetic and molecular biology. Some recommendations have been written.
Collapse
Affiliation(s)
- Monique Courtade-Saïdi
- Département d'anatomie et cytologie pathologiques, institut universitaire du cancer de Toulouse Oncopole, 1, avenue Irène-Joliot-Curie, 31059 Toulouse cedex 09, France.
| | - Jocelyne Fleury Feith
- Service d'anatomie et cytologie pathologiques, hôpital Tenon, CHUG Paris-Est, AP-HP, 4, rue de la Chine, 75020 Paris, France
| |
Collapse
|
132
|
Pusztaszeri MP, Krane JF, Faquin WC. BRAF testing and thyroid FNA. Cancer Cytopathol 2015; 123:689-95. [PMID: 26335367 DOI: 10.1002/cncy.21614] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 08/18/2015] [Indexed: 12/17/2022]
Affiliation(s)
- Marc P Pusztaszeri
- Department of Pathology, Geneva University Hospital, Geneva, Switzerland
| | - Jeffrey F Krane
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - William C Faquin
- Harvard Medical School, Boston, Massachusetts.,Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
133
|
Analysis of Pre-Analytic Factors Affecting the Success of Clinical Next-Generation Sequencing of Solid Organ Malignancies. Cancers (Basel) 2015; 7:1699-715. [PMID: 26343728 PMCID: PMC4586792 DOI: 10.3390/cancers7030859] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 08/20/2015] [Accepted: 08/21/2015] [Indexed: 01/28/2023] Open
Abstract
Application of next-generation sequencing (NGS) technology to routine clinical practice has enabled characterization of personalized cancer genomes to identify patients likely to have a response to targeted therapy. The proper selection of tumor sample for downstream NGS based mutational analysis is critical to generate accurate results and to guide therapeutic intervention. However, multiple pre-analytic factors come into play in determining the success of NGS testing. In this review, we discuss pre-analytic requirements for AmpliSeq PCR-based sequencing using Ion Torrent Personal Genome Machine (PGM) (Life Technologies), a NGS sequencing platform that is often used by clinical laboratories for sequencing solid tumors because of its low input DNA requirement from formalin fixed and paraffin embedded tissue. The success of NGS mutational analysis is affected not only by the input DNA quantity but also by several other factors, including the specimen type, the DNA quality, and the tumor cellularity. Here, we review tissue requirements for solid tumor NGS based mutational analysis, including procedure types, tissue types, tumor volume and fraction, decalcification, and treatment effects.
Collapse
|
134
|
Towards a Next-Generation Sequencing Diagnostic Service for Tumour Genotyping: A Comparison of Panels and Platforms. BIOMED RESEARCH INTERNATIONAL 2015; 2015:478017. [PMID: 26351634 PMCID: PMC4553307 DOI: 10.1155/2015/478017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 08/10/2015] [Indexed: 11/23/2022]
Abstract
Detection of clinically actionable mutations in diagnostic tumour specimens aids in the selection of targeted therapeutics. With an ever increasing number of clinically significant mutations identified, tumour genetic diagnostics is moving from single to multigene analysis. As it is still not feasible for routine diagnostic laboratories to perform sequencing of the entire cancer genome, our approach was to undertake targeted mutation detection. To optimise our diagnostic workflow, we evaluated three target enrichment strategies using two next-generation sequencing (NGS) platforms (Illumina MiSeq and Ion PGM). The target enrichment strategies were Fluidigm Access Array custom amplicon panel including 13 genes (MiSeq sequencing), the Oxford Gene Technologies (OGT) SureSeq Solid Tumour hybridisation panel including 60 genes (MiSeq sequencing), and an Ion AmpliSeq Cancer Hotspot Panel including 50 genes (Ion PGM sequencing). DNA extracted from formalin-fixed paraffin-embedded (FFPE) blocks of eight previously characterised cancer cell lines was tested using the three panels. Matching genomic DNA from fresh cultures of these cell lines was also tested using the custom Fluidigm panel and the OGT SureSeq Solid Tumour panel. Each panel allowed mutation detection of core cancer genes including KRAS, BRAF, and EGFR. Our results indicate that the panels enable accurate variant detection despite sequencing from FFPE DNA.
Collapse
|
135
|
Lee HB, Joung JG, Kim J, Lee KM, Ryu HS, Lee HO, Moon HG, Park WY, Noh DY, Han W. The use of FNA samples for whole-exome sequencing and detection of somatic mutations in breast cancer surgical specimens. Cancer Cytopathol 2015; 123:669-77. [PMID: 26265110 DOI: 10.1002/cncy.21599] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 07/04/2015] [Accepted: 07/07/2015] [Indexed: 01/12/2023]
Abstract
BACKGROUND The availability of suitable biospecimens is critical to the success of advanced genomic analyses. The objective of this study was to assess the sensitivity of fine-needle aspiration (FNA) compared with gross surgical sampling (GSS) from surgical specimens for the detection of somatic mutations in breast cancer using whole-exome sequencing (WES). METHODS DNA was extracted from paired GSS tissues and FNA samples of surgically resected breast cancer from 12 patients and was used for WES. Sanger sequencing was performed to validate selected somatic mutations. Tumor purity was calculated for each sample using sequencing data. RESULTS There was no difference in the total amount of DNA extracted from GSS tissues and FNA samples. WES was successfully performed for all 12 pairs of samples. The median number of somatic mutations identified in individual samples was higher in FNA samples than in GSS tissues (39.5 vs 18.5; P = .036). The somatic mutation profiles from both sampling methods were well correlated for samples that had GSS tissues with high tumor content, as indicated by hematoxylin and eosin staining. Nineteen mutations that were identified exclusively in FNA samples were subjected to Sanger sequencing, and 13 of those mutations (68.4%) were validated. The mean estimated tumor purity was higher in FNA samples than in GSS tissues (55.87% vs 25.76%), and FNA samples were estimated to have a consistently higher proportion of malignant cells. CONCLUSIONS The current results suggest that FNA is feasible for the collection of tumor samples sufficient for WES analysis and that the higher purity obtained using this method may make it more reliable for genomic studies.
Collapse
Affiliation(s)
- Han-Byoel Lee
- Department of Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Je-Gun Joung
- Samsung Genome Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Jisun Kim
- Department of Surgery, Asan Medical Center, Seoul, Republic of Korea
| | - Kyung-Min Lee
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Han Suk Ryu
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hae-Ock Lee
- Samsung Genome Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Hyeong-Gon Moon
- Department of Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, Republic of Korea.,Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Dong-Young Noh
- Department of Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Wonshik Han
- Department of Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
136
|
Roy-Chowdhuri S, Goswami RS, Chen H, Patel KP, Routbort MJ, Singh RR, Broaddus RR, Barkoh BA, Manekia J, Yao H, Medeiros LJ, Staerkel G, Luthra R, Stewart J. Factors affecting the success of next-generation sequencing in cytology specimens. Cancer Cytopathol 2015; 123:659-68. [PMID: 26230354 DOI: 10.1002/cncy.21597] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 06/22/2015] [Accepted: 06/23/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND The use of cytology specimens for next-generation sequencing (NGS) is particularly challenging because of the unconventional substrate of smears and the often limited sample volume. An analysis of factors affecting NGS testing in cytologic samples may help to increase the frequency of successful testing. METHODS This study reviewed variables associated with all in-house cytology cases (n = 207) that were analyzed by NGS with the Ion Torrent platform during a 10-month interval. A statistical analysis was performed to measure the effects of the DNA input threshold, specimen preparation, slide type, tumor fraction, DNA yield, and cytopathologist bias. RESULTS One hundred sixty-four of 207 cases (79%) were successfully sequenced by NGS; 43 (21%) failed because of either a low DNA yield or a template/library preparation failure. The median estimated tumor fraction and DNA concentration for the successfully sequenced cases were 70% and 2.5 ng/μL, respectively, whereas they were 60% and 0.2 ng/μL, respectively, for NGS failures. Cell block sections were tested in 91 cases, and smears were used in 116 cases. NGS success positively correlated with the DNA yield but not the tumor fraction. Cell block preparations showed a higher success rate than smears. Frosted-tip slides yielded significantly more DNA than fully frosted slides. Lowering the input DNA concentration below the manufacturer's recommended threshold of 10 ng (>0.85 ng/μL) resulted in a marked increase in the NGS success rate from 58.6% to 89.8%. CONCLUSIONS The failure of NGS with cytology samples is usually a result of suboptimal DNA due to multiple pre-analytical factors. Knowledge of these factors will allow better selection of cytology material for mutational analysis.
Collapse
Affiliation(s)
- Sinchita Roy-Chowdhuri
- Department of Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rashmi S Goswami
- Department of Hematopathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hui Chen
- Department of Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Keyur P Patel
- Department of Hematopathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mark J Routbort
- Department of Hematopathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rajesh R Singh
- Department of Hematopathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Russell R Broaddus
- Department of Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bedia A Barkoh
- Department of Hematopathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jawad Manekia
- Department of Hematopathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hui Yao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - L Jeffrey Medeiros
- Department of Hematopathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gregg Staerkel
- Department of Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rajyalakshmi Luthra
- Department of Hematopathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John Stewart
- Department of Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
137
|
Roh MH. The Utilization of Cytologic Fine-Needle Aspirates of Lung Cancer for Molecular Diagnostic Testing. J Pathol Transl Med 2015; 49:300-9. [PMID: 26076721 PMCID: PMC4508567 DOI: 10.4132/jptm.2015.06.16] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 06/10/2015] [Accepted: 06/16/2015] [Indexed: 01/10/2023] Open
Abstract
In this era of precision medicine, our understanding and knowledge of the molecular landscape associated with lung cancer pathogenesis continues to evolve. This information is being increasingly exploited to treat advanced stage lung cancer patients with tailored, targeted therapy. During the management of these patients, minimally invasive procedures to obtain samples for tissue diagnoses are desirable. Cytologic fine-needle aspirates are often utilized for this purpose and are important not only for rendering diagnoses to subtype patients’ lung cancers, but also for ascertaining molecular diagnostic information for treatment purposes. Thus, cytologic fine-needle aspirates must be utilized and triaged judiciously to achieve both objectives. In this review, strategies in utilizing fine-needle aspirates will be discussed in the context of our current understanding of the clinically actionable molecular aberrations underlying non-small cell lung cancer and the molecular assays applied to these samples in order to obtain treatment-relevant molecular diagnostic information.
Collapse
Affiliation(s)
- Michael H Roh
- Department of Pathology, University of Michigan Health System, Ann Arbor, MI, USA
| |
Collapse
|
138
|
Wang M, Escudero-Ibarz L, Moody S, Zeng N, Clipson A, Huang Y, Xue X, Grigoropoulos NF, Barrans S, Worrillow L, Forshew T, Su J, Firth A, Martin H, Jack A, Brugger K, Du MQ. Somatic Mutation Screening Using Archival Formalin-Fixed, Paraffin-Embedded Tissues by Fluidigm Multiplex PCR and Illumina Sequencing. J Mol Diagn 2015; 17:521-32. [PMID: 26165823 DOI: 10.1016/j.jmoldx.2015.04.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 04/02/2015] [Accepted: 04/27/2015] [Indexed: 01/03/2023] Open
Abstract
High-throughput somatic mutation screening using FFPE tissues is a major challenge because of a lack of established methods and validated variant calling algorithms. We aimed to develop a targeted sequencing protocol by Fluidigm multiplex PCR and Illumina sequencing and to establish a companion variant calling algorithm. The experimental protocol and variant calling algorithm were first developed and optimized against a series of somatic mutations (147 substitutions, 12 indels ranging from 1 to 33 bp) in seven genes, previously detected by Sanger sequencing of DNA from 163 FFPE lymphoma biopsy specimens. The optimized experimental protocol and variant calling algorithm were further ascertained in two separate experiments by including the seven genes as a part of larger gene panels (22 or 13 genes) using FFPE and high-molecular-weight lymphoma DNAs, respectively. We found that most false-positive variants were due to DNA degradation, deamination, and Taq polymerase errors, but they were nonreproducible and could be efficiently eliminated by duplicate experiments. A small fraction of false-positive variants appeared in duplicate, but they were at low alternative allele frequencies and could be separated from mutations when appropriate threshold value was used. In conclusion, we established a robust practical approach for high-throughput mutation screening using archival FFPE tissues.
Collapse
Affiliation(s)
- Ming Wang
- Division of Molecular Histopathology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Leire Escudero-Ibarz
- Division of Molecular Histopathology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Sarah Moody
- Division of Molecular Histopathology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Naiyan Zeng
- Division of Molecular Histopathology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Alexandra Clipson
- Division of Molecular Histopathology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Yuanxue Huang
- Division of Molecular Histopathology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Xuemin Xue
- Division of Molecular Histopathology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Nicholas F Grigoropoulos
- Division of Molecular Histopathology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Sharon Barrans
- Haematological Malignancy Diagnostic Service, St. James's Institute of Oncology, Leeds, United Kingdom
| | - Lisa Worrillow
- Haematological Malignancy Diagnostic Service, St. James's Institute of Oncology, Leeds, United Kingdom
| | - Tim Forshew
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Jing Su
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Andrew Firth
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Howard Martin
- Department of Molecular Genetics, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Andrew Jack
- Haematological Malignancy Diagnostic Service, St. James's Institute of Oncology, Leeds, United Kingdom
| | - Kim Brugger
- Department of Molecular Genetics, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Ming-Qing Du
- Division of Molecular Histopathology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
139
|
Pusztaszeri MP, Faquin WC. Update in salivary gland cytopathology: Recent molecular advances and diagnostic applications. Semin Diagn Pathol 2015; 32:264-74. [DOI: 10.1053/j.semdp.2014.12.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
140
|
Next-generation sequencing for molecular diagnosis of lung adenocarcinoma specimens obtained by fine needle aspiration cytology. Sci Rep 2015; 5:11317. [PMID: 26066407 PMCID: PMC4464150 DOI: 10.1038/srep11317] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 05/20/2015] [Indexed: 12/16/2022] Open
Abstract
Identification of multi-gene variations has led to the development of new targeted therapies in lung adenocarcinoma patients, and identification of an appropriate patient population with a reliable screening method is the key to the overall success of tumor targeted therapies. In this study, we used the Ion Torrent next-generation sequencing (NGS) technique to screen for mutations in 89 cases of lung adenocarcinoma metastatic lymph node specimens obtained by fine-needle aspiration cytology (FNAC). Of the 89 specimens, 30 (34%) were found to harbor epidermal growth factor receptor (EGFR) kinase domain mutations. Seven (8%) samples harbored KRAS mutations, and three (3%) samples had BRAF mutations involving exon 11 (G469A) and exon 15 (V600E). Eight (9%) samples harbored PIK3CA mutations. One (1%) sample had a HRAS G12C mutation. Thirty-two (36%) samples (36%) harbored TP53 mutations. Other genes including APC, ATM, MET, PTPN11, GNAS, HRAS, RB1, SMAD4 and STK11 were found each in one case. Our study has demonstrated that NGS using the Ion Torrent technology is a useful tool for gene mutation screening in lung adenocarcinoma metastatic lymph node specimens obtained by FNAC, and may promote the development of new targeted therapies in lung adenocarcinoma patients.
Collapse
|
141
|
Al-Kateb H, Nguyen TT, Steger-May K, Pfeifer JD. Identification of major factors associated with failed clinical molecular oncology testing performed by next generation sequencing (NGS). Mol Oncol 2015; 9:1737-43. [PMID: 26071350 DOI: 10.1016/j.molonc.2015.05.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 05/05/2015] [Accepted: 05/07/2015] [Indexed: 12/21/2022] Open
Abstract
PURPOSE DNA analysis by NGS has become important to direct the clinical care of cancer patients. However, NGS is not successful in all cases, and the factors responsible for test failures have not been systematically evaluated. MATERIALS AND METHODS A series of 1528 solid and hematolymphoid tumor specimens was tested by an NGS comprehensive cancer panel during 2012-2014. DNA was extracted and 2×101 bp paired-end sequence reads were generated on cancer-related genes utilizing Illumina HiSeq and MiSeq platforms. RESULTS Testing was unsuccessful in 343 (22.5%) specimens. The failure was due to insufficient tissue (INST) in 223/343 (65%) cases, insufficient DNA (INS-DNA) in 99/343 (28.9%) cases, and failed library (FL) in 21/343 (6.1%) cases. 87/99 (88%) of the INS-DNA cases had below 10 ng DNA available for testing. Factors associated with INST and INS-DNA failures were site of biopsy (SOB) and type of biopsy (TOB) (both p < 0.0001), and clinical setting of biopsy (CSB, initial diagnosis or recurrence) (p < 0.0001). Factors common to INST and FL were age of specimen (p ≤ 0.006) and tumor viability (p ≤ 0.05). Factors common to INS-DNA and FL were DNA purity and DNA degradation (all p ≤ 0.005). In multivariate analysis, common predictors for INST and INS-DNA included CSB (p = 0.048 and p < 0.0001) and TOB (both p ≤ 0.003), respectively. SOB (p = 0.004) and number of cores (p = 0.001) were specific for INS-DNA, whereas TOB and DNA degradation were associated with FL (p = 0.04 and 0.02, respectively). CONCLUSIONS Pre-analytical causes (INST and INS-DNA) accounted for about 90% of all failed cases; independent of test design. Clinical setting; site and type of biopsy; and number of cores used for testing all correlated with failure. Accounting for these factors at the time of tissue biopsy acquisition could improve the analytic success rate.
Collapse
Affiliation(s)
- Hussam Al-Kateb
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, USA.
| | - TuDung T Nguyen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, USA
| | - Karen Steger-May
- Division of Biostatistics, Washington University School of Medicine, St. Louis, USA
| | - John D Pfeifer
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, USA
| |
Collapse
|
142
|
Abstract
Background Biliary cancers are highly aggressive tumors that are often diagnosed an advanced disease stage and have a poor outcome with systemic therapy. Recent efforts towards molecular characterization have identified a subset of biliary patients that have HER2/neu amplification or mutation. HER2/neu amplification is associated with response to HER2/neu-directed therapy in breast and gastric cancers. However, the efficacy of HER2/neu-targeted therapy in biliary cancers is unknown. Patients and methods We retrospectively reviewed cases of advanced gallbladder cancer and cholangiocarcinoma with HER2/neu genetic aberrations or protein overexpression who received HER2/neu-directed therapy between 2007 and 2014. Clinical data were retrieved from medical records, and imaging studies were independently reviewed. Results Nine patients with gallbladder cancer and five patients with cholangiocarcinoma had received HER2/neu-directed therapy (trastuzumab, lapatinib, or pertuzumab) during the study period. In the gallbladder cancer group, HER2/neu gene amplification or overexpression was detected in eight cases. These patients experienced disease stability (n = 3), partial response (n = 4), or complete response (n = 1) with HER2/neu-directed therapy. One patient had HER2/neu mutation and experienced a mixed response after lapatinib therapy. The duration of response varied from 8+ to 168 weeks (median 40 weeks), and three patients are still on therapy. One patient developed HER2/neu amplification as a secondary event after FGFR-directed therapy for FGF3-TACC3 gene fusion. The cholangiocarcinoma cases treated in this series had a higher proportion of HER2/neu mutations, and no radiological responses were seen in these patients despite HER2/neu-directed therapy. Conclusions HER2/neu blockade is a promising treatment strategy for gallbladder cancer patients with gene amplification and deserves further exploration in a multi-center study.
Collapse
|
143
|
Oh SY, Lee HT. Efficiency of EGFR mutation analysis for small microdissected cytological specimens using multitech DNA extraction solution. Cancer Cytopathol 2015; 123:401-12. [PMID: 25964184 DOI: 10.1002/cncy.21550] [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] [Received: 02/24/2015] [Revised: 03/23/2015] [Accepted: 03/31/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND The microdissection method has greatly facilitated the isolation of pure cell populations for accurate analysis of mutations. However, the absence of coverslips in these preparations leads to poor resolution of cellular morphological features. In the current study, the authors developed the MultiTech DNA extraction solution to improve the visualization of cell morphology for microdissection and tested it for the preservation of morphological properties of cells, quality of DNA, and ability to detect mutations. METHODS A total of 121 cytological samples, including fine-needle aspirates, sputum, pleural fluid, and bronchial washings, were selected from hospital archives. DNA extracted from microdissected cells was evaluated by epidermal growth factor receptor (EGFR) mutation analysis using pyrosequencing, Sanger sequencing, and peptide nucleic acid (PNA)-mediated real-time polymerase chain reaction clamping. Morphological features of cells as well as DNA quality and quantity were analyzed in several cytological samples to assess the performance of the MultiTech DNA extraction solution. The results were compared with previous EGFR mutation tests. RESULTS The MultiTech DNA extraction solution improved the morphology of archived stained cells before microdissection and provided a higher DNA yield than the commercial QIAamp DNA Mini Kit in samples containing a minimal number of cells (25-50 cells). The authors were able to detect identical EGFR mutations by using different analysis platforms and consistently identified these mutations in samples comprising as few as 25 microdissected cells. CONCLUSIONS The MultiTech DNA extraction solution is a reliable medium that improves the resolution of cell morphology during microdissection. It was particularly useful in EGFR mutations of samples containing a small number of cells.
Collapse
Affiliation(s)
- Seo Young Oh
- Department of Animal Biotechnology, College of Animal Bioscience and Technology, Konkuk University, Seoul, Korea
| | - Hoon Taek Lee
- Department of Animal Biotechnology, College of Animal Bioscience and Technology, Konkuk University, Seoul, Korea
| |
Collapse
|
144
|
Cheng DT, Mitchell TN, Zehir A, Shah RH, Benayed R, Syed A, Chandramohan R, Liu ZY, Won HH, Scott SN, Brannon AR, O'Reilly C, Sadowska J, Casanova J, Yannes A, Hechtman JF, Yao J, Song W, Ross DS, Oultache A, Dogan S, Borsu L, Hameed M, Nafa K, Arcila ME, Ladanyi M, Berger MF. Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT): A Hybridization Capture-Based Next-Generation Sequencing Clinical Assay for Solid Tumor Molecular Oncology. J Mol Diagn 2015; 17:251-64. [PMID: 25801821 DOI: 10.1016/j.jmoldx.2014.12.006] [Citation(s) in RCA: 1542] [Impact Index Per Article: 171.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 11/20/2014] [Accepted: 12/18/2014] [Indexed: 01/17/2023] Open
Abstract
The identification of specific genetic alterations as key oncogenic drivers and the development of targeted therapies are together transforming clinical oncology and creating a pressing need for increased breadth and throughput of clinical genotyping. Next-generation sequencing assays allow the efficient and unbiased detection of clinically actionable mutations. To enable precision oncology in patients with solid tumors, we developed Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT), a hybridization capture-based next-generation sequencing assay for targeted deep sequencing of all exons and selected introns of 341 key cancer genes in formalin-fixed, paraffin-embedded tumors. Barcoded libraries from patient-matched tumor and normal samples were captured, sequenced, and subjected to a custom analysis pipeline to identify somatic mutations. Sensitivity, specificity, reproducibility of MSK-IMPACT were assessed through extensive analytical validation. We tested 284 tumor samples with previously known point mutations and insertions/deletions in 47 exons of 19 cancer genes. All known variants were accurately detected, and there was high reproducibility of inter- and intrarun replicates. The detection limit for low-frequency variants was approximately 2% for hotspot mutations and 5% for nonhotspot mutations. Copy number alterations and structural rearrangements were also reliably detected. MSK-IMPACT profiles oncogenic DNA alterations in clinical solid tumor samples with high accuracy and sensitivity. Paired analysis of tumors and patient-matched normal samples enables unambiguous detection of somatic mutations to guide treatment decisions.
Collapse
Affiliation(s)
- Donavan T Cheng
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Talia N Mitchell
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ahmet Zehir
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ronak H Shah
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ryma Benayed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Aijazuddin Syed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Raghu Chandramohan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Zhen Yu Liu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Helen H Won
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sasinya N Scott
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - A Rose Brannon
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Catherine O'Reilly
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Justyna Sadowska
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jacklyn Casanova
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Angela Yannes
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jaclyn F Hechtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jinjuan Yao
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Wei Song
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Dara S Ross
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alifya Oultache
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Snjezana Dogan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Laetitia Borsu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Meera Hameed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Khedoudja Nafa
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maria E Arcila
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael F Berger
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
145
|
Gan C, Love C, Beshay V, Macrae F, Fox S, Waring P, Taylor G. Applicability of next generation sequencing technology in microsatellite instability testing. Genes (Basel) 2015; 6:46-59. [PMID: 25685876 PMCID: PMC4377833 DOI: 10.3390/genes6010046] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 01/27/2015] [Indexed: 11/16/2022] Open
Abstract
Microsatellite instability (MSI) is a useful marker for risk assessment, prediction of chemotherapy responsiveness and prognosis in patients with colorectal cancer. Here, we describe a next generation sequencing approach for MSI testing using the MiSeq platform. Different from other MSI capturing strategies that are based on targeted gene capture, we utilize “deep resequencing”, where we focus the sequencing on only the microsatellite regions of interest. We sequenced a series of 44 colorectal tumours with normal controls for five MSI loci (BAT25, BAT26, BAT34c4, D18S55, D5S346) and a second series of six colorectal tumours (no control) with two mononucleotide loci (BAT25, BAT26). In the first series, we were able to determine 17 MSI-High, 1 MSI-Low and 26 microsatellite stable (MSS) tumours. In the second series, there were three MSI-High and three MSS tumours. Although there was some variation within individual markers, this NGS method produced the same overall MSI status for each tumour, as obtained with the traditional multiplex PCR-based method.
Collapse
Affiliation(s)
- Chun Gan
- Department of Pathology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria 3052, Australia.
- Department of Colorectal Medicine and Genetics, Familial Cancer Clinic, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia.
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia.
| | - Clare Love
- Department of Pathology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria 3052, Australia.
| | - Victoria Beshay
- Department of Pathology and Sir Peter MacCallum Department of Oncology, University of Melbourne, East Melbourne, Victoria 3002, Australia.
| | - Finlay Macrae
- Department of Colorectal Medicine and Genetics, Familial Cancer Clinic, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia.
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia.
| | - Stephen Fox
- Department of Pathology and Sir Peter MacCallum Department of Oncology, University of Melbourne, East Melbourne, Victoria 3002, Australia.
| | - Paul Waring
- Department of Pathology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria 3052, Australia.
| | - Graham Taylor
- Department of Pathology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria 3052, Australia.
| |
Collapse
|
146
|
Li-Chang HH, Kasaian K, Ng Y, Lum A, Kong E, Lim H, Jones SJ, Huntsman DG, Schaeffer DF, Yip S. Retrospective review using targeted deep sequencing reveals mutational differences between gastroesophageal junction and gastric carcinomas. BMC Cancer 2015; 15:32. [PMID: 25656989 PMCID: PMC4322811 DOI: 10.1186/s12885-015-1021-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 01/14/2015] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Adenocarcinomas of both the gastroesophageal junction and stomach are molecularly complex, but differ with respect to epidemiology, etiology and survival. There are few data directly comparing the frequencies of single nucleotide mutations in cancer-related genes between the two sites. Sequencing of targeted gene panels may be useful in uncovering multiple genomic aberrations using a single test. METHODS DNA from 92 gastroesophageal junction and 75 gastric adenocarcinoma resection specimens was extracted from formalin-fixed paraffin-embedded tissue. Targeted deep sequencing of 46 cancer-related genes was performed through emulsion PCR followed by semiconductor-based sequencing. Gastroesophageal junction and gastric carcinomas were contrasted with respect to mutational profiles, immunohistochemistry and in situ hybridization, as well as corresponding clinicopathologic data. RESULTS Gastroesophageal junction carcinomas were associated with younger age, more frequent intestinal-type histology, more frequent p53 overexpression, and worse disease-free survival on multivariable analysis. Among all cases, 145 mutations were detected in 31 genes. TP53 mutations were the most common abnormality detected, and were more common in gastroesophageal junction carcinomas (42% vs. 27%, p = 0.036). Mutations in the Wnt pathway components APC and CTNNB1 were more common among gastric carcinomas (16% vs. 3%, p = 0.006), and gastric carcinomas were more likely to have ≥3 driver mutations detected (11% vs. 2%, p = 0.044). Twenty percent of cases had potentially actionable mutations identified. R132H and R132C missense mutations in the IDH1 gene were observed, and are the first reported mutations of their kind in gastric carcinoma. CONCLUSIONS Panel sequencing of routine pathology material can yield mutational information on several driver genes, including some for which targeted therapies are available. Differing rates of mutations and clinicopathologic differences support a distinction between adenocarcinomas that arise in the gastroesophageal junction and those that arise in the stomach proper.
Collapse
Affiliation(s)
- Hector H Li-Chang
- University of British Columbia, Vancouver, Canada.
- Division of Anatomic Pathology, Department of Pathology and Laboratory Medicine, Vancouver General Hospital, 855 12 Ave W, Vancouver, BC, V5Z 1 M9, Canada.
- Department of Molecular Oncology, British Columbia Cancer Agency, Vancouver, Canada.
| | - Katayoon Kasaian
- University of British Columbia, Vancouver, Canada.
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, Canada.
| | - Ying Ng
- Centre for Translational and Applied Genomics, British Columbia Cancer Agency, Vancouver, Canada.
| | - Amy Lum
- Centre for Translational and Applied Genomics, British Columbia Cancer Agency, Vancouver, Canada.
| | - Esther Kong
- Centre for Translational and Applied Genomics, British Columbia Cancer Agency, Vancouver, Canada.
| | - Howard Lim
- University of British Columbia, Vancouver, Canada.
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, Canada.
| | - Steven Jm Jones
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, Canada.
| | - David G Huntsman
- University of British Columbia, Vancouver, Canada.
- Division of Anatomic Pathology, Department of Pathology and Laboratory Medicine, Vancouver General Hospital, 855 12 Ave W, Vancouver, BC, V5Z 1 M9, Canada.
- Department of Molecular Oncology, British Columbia Cancer Agency, Vancouver, Canada.
- Centre for Translational and Applied Genomics, British Columbia Cancer Agency, Vancouver, Canada.
| | - David F Schaeffer
- University of British Columbia, Vancouver, Canada.
- Division of Anatomic Pathology, Department of Pathology and Laboratory Medicine, Vancouver General Hospital, 855 12 Ave W, Vancouver, BC, V5Z 1 M9, Canada.
| | - Stephen Yip
- University of British Columbia, Vancouver, Canada.
- Division of Anatomic Pathology, Department of Pathology and Laboratory Medicine, Vancouver General Hospital, 855 12 Ave W, Vancouver, BC, V5Z 1 M9, Canada.
- Centre for Translational and Applied Genomics, British Columbia Cancer Agency, Vancouver, Canada.
| |
Collapse
|
147
|
Abstract
The thyroid parafollicular cell, or commonly named "C-cell," functions in serum calcium homeostasis. Elevations in serum calcium trigger release of calcitonin from the C-cell, which in turn functions to inhibit absorption of calcium by the intestine, resorption of bone by the osteoclast, and reabsorption of calcium by renal tubular cells. Oncogenic transformation of the thyroid C-cell is thought to progress through a hyperplastic process prior to malignancy with increasing levels of serum calcitonin serving as a biomarker for tumor burden. The discovery that multiple endocrine neoplasia type 2 is caused by activating mutations of the RET gene serves to highlight the RET-RAS-MAPK signaling pathway in both initiation and progression of medullary thyroid carcinoma (MTC). Thyroid C-cells are known to express RET at high levels relative to most cell types; therefore, aberrant activation of this receptor is targeted primarily to the C-cell, providing one possible cause of tissue-specific oncogenesis. The role of RET signaling in normal C-cell function is unknown though calcitonin gene transcription appears to be sensitive to RET activation. Beyond RET, the modeling of oncogenesis in animals and screening of human tumors for candidate gene mutations have uncovered mutation of RAS family members and inactivation of Rb1 regulatory pathway as potential mediators of C-cell transformation. A growing understanding of how RET interacts with these pathways, both in normal C-cell function and during oncogenic transformation, will help in the development of novel molecular-targeted therapies.
Collapse
Affiliation(s)
- Gilbert J Cote
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1461, Houston, TX, 77030, USA.
| | - Elizabeth G Grubbs
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1484, Houston, TX, 77030, USA
| | - Marie-Claude Hofmann
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1461, Houston, TX, 77030, USA
| |
Collapse
|
148
|
Dumur CI, Kraft AO. Next-generation sequencing and the cytopathologist. Cancer Cytopathol 2014; 123:69-70. [PMID: 25557465 DOI: 10.1002/cncy.21515] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 12/10/2014] [Indexed: 11/07/2022]
|
149
|
Churi CR, Shroff R, Wang Y, Rashid A, Kang HC, Weatherly J, Zuo M, Zinner R, Hong D, Meric-Bernstam F, Janku F, Crane CH, Mishra L, Vauthey JN, Wolff RA, Mills G, Javle M. Mutation profiling in cholangiocarcinoma: prognostic and therapeutic implications. PLoS One 2014; 9:e115383. [PMID: 25536104 PMCID: PMC4275227 DOI: 10.1371/journal.pone.0115383] [Citation(s) in RCA: 338] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 11/21/2014] [Indexed: 02/07/2023] Open
Abstract
Background Cholangiocarcinoma (CCA) is clinically heterogeneous; intra and extrahepatic CCA have diverse clinical presentations. Next generation sequencing (NGS) technology may identify the genetic differences between these entities and identify molecular subgroups for targeted therapeutics. Methods We describe successful NGS-based testing of 75 CCA patients along with the prognostic and therapeutic implications of findings. Mutation profiling was performed using either a) NGS panel of hotspot regions in 46 cancer-related genes using a 318-chip on Ion PGM Sequencer or b) Illumina HiSeq 2000 sequencing platform for 3,769 exons of 236 cancer-related genes plus 47 introns from 19 genes to an average depth of 1000X. Clinical data was abstracted and correlated with clinical outcome. Patients with targetable mutations were referred to appropriate clinical trials. Results There were significant differences between intrahepatic (n = 55) and extrahepatic CCA (n = 20) in regard to the nature and frequency of the genetic aberrations (GAs). IDH1 and DNA repair gene alterations occurred more frequently in intrahepatic CCA, while ERBB2 GAs occurred in the extrahepatic group. Commonly occurring GAs in intrahepatic CCA were TP53 (35%), KRAS (24%), ARID1A (20%), IDH1 (18%), MCL1 (16%) and PBRM1 (11%). Most frequent GAs in extrahepatic CCA (n = 20) were TP53 (45%), KRAS (40%), ERBB2 (25%), SMAD4 (25%), FBXW7 (15%) and CDKN2A (15%). In intrahepatic CCA, KRAS, TP53 or MAPK/mTOR GAs were significantly associated with a worse prognosis while FGFR GAs correlated with a relatively indolent disease course. IDH1 GAs did not have any prognostic significance. GAs in the chromatin modulating genes, BAP1 and PBRM1 were associated with bone metastases and worse survival in extrahepatic CCA. Radiologic responses and clinical benefit was noted with EGFR, FGFR, C-met, B-RAF and MEK inhibitors. Conclusion There are significant genetic differences between intra and extrahepatic CCA. NGS can potentially identify disease subsets with distinct prognostic and therapeutic implications.
Collapse
Affiliation(s)
- Chaitanya R. Churi
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Rachna Shroff
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Ying Wang
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Asif Rashid
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - HyunSeon C. Kang
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Jacqueline Weatherly
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Mingxin Zuo
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Ralph Zinner
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - David Hong
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Funda Meric-Bernstam
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Filip Janku
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Christopher H. Crane
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Lopa Mishra
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Jean-Nicholas Vauthey
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Robert A. Wolff
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Gordon Mills
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Milind Javle
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
150
|
Le Mercier M, D'Haene N, De Nève N, Blanchard O, Degand C, Rorive S, Salmon I. Next-generation sequencing improves the diagnosis of thyroid FNA specimens with indeterminate cytology. Histopathology 2014; 66:215-24. [PMID: 24834793 DOI: 10.1111/his.12461] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 05/14/2014] [Indexed: 01/21/2023]
Abstract
AIMS The assessment of thyroid nodules is a common clinical challenge. Fine-needle aspiration (FNA) is the standard pre-operative tool for thyroid nodule diagnosis. However, up to 30% of the samples are classified as indeterminate. This often leads to unnecessary surgery. In this study, we evaluated the added value of next-generation sequencing (NGS) for helping in the diagnosis of FNA samples. METHODS AND RESULTS We analysed retrospectively 34 indeterminate FNA samples for which surgical resection was performed. DNA was obtained from cell blocks or from stained smears and subjected to NGS to analyse mutations in 50 genes. Mutations in BRAF, NRAS, KRAS and PTEN, that are known to be involved in thyroid cancer biology, were detected in seven FNA samples. The presence of a mutation in these genes was a strong indicator of cancer because five (71%) of the mutation-positive FNA samples had a malignant diagnosis after surgery. Moreover, there was only an 8% cancer risk in nodules with an indeterminate cytological diagnosis but with a negative molecular test. CONCLUSION This study demonstrates that thyroid FNA can be analysed successfully by NGS. The detection of mutations known to be involved in thyroid cancer improves the sensitivity of thyroid FNA diagnosis.
Collapse
Affiliation(s)
- Marie Le Mercier
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | | | | | | | | | | |
Collapse
|