201
|
Udar N, Iyer A, Porter M, Haigis R, Smith S, Dhillon S, Meier K, Ward D, Lu J, Wenz P, Buchner L, Dunn T, Wise A, Mueller A, Gutekunst K. Development and Analytical Validation of a DNA Dual-Strand Approach for the US Food and Drug Administration-Approved Next-Generation Sequencing-Based Praxis Extended RAS Panel for Metastatic Colorectal Cancer Samples. J Mol Diagn 2019; 22:159-178. [PMID: 31837434 DOI: 10.1016/j.jmoldx.2019.09.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 09/03/2019] [Accepted: 09/27/2019] [Indexed: 12/28/2022] Open
Abstract
A next-generation sequencing method was developed that can distinguish single-stranded modifications from low-frequency somatic mutations present on both strands of DNA in formalin-fixed paraffin-embedded colorectal cancer samples. We applied this method for analytical validation of the Praxis Extended RAS Panel, a US Food and Drug Administration-approved companion diagnostic for panitumumab, on the Illumina MiSeqDx platform. With the use of the TruSeq amplicon workflow, both strands of DNA from the starting material were interrogated independently. Mutations were reported only if found on both strands; artifacts usually present on only one strand would not be reported. A total of 56 mutations were targeted within the KRAS and NRAS genes. A minimum read depth of 1800× per amplicon is required per sample but averaged >30,000× at maximum multiplexing levels. Analytical validation studies were performed to determine the simultaneous detection of mutations on both strands, reproducibility, assay detection level, precision of the assay across various factors, and the impact of interfering substances. In conclusion, this assay can clearly distinguish single-stranded artifacts from low-frequency mutations. Furthermore, the assay is accurate, precise, and reproducible, can achieve consistent detection of a mutation at 5% mutation frequency, exhibits minimal impact from tested interfering substances, and can simultaneously detect 56 mutations in a single run using 10 samples plus controls.
Collapse
Affiliation(s)
- Nitin Udar
- Department of Clinical Genomics Assay Development and Oncology, Illumina, Inc., San Diego, California
| | - Anita Iyer
- Department of Clinical Genomics Assay Development and Oncology, Illumina, Inc., San Diego, California.
| | - Margaret Porter
- Department of Clinical Genomics Assay Development and Oncology, Illumina, Inc., San Diego, California
| | - Robert Haigis
- Department of Clinical Genomics Assay Development and Oncology, Illumina, Inc., San Diego, California
| | - Shannon Smith
- Department of Clinical Genomics Assay Development and Oncology, Illumina, Inc., San Diego, California
| | - Shivani Dhillon
- Department of Clinical Genomics Assay Development and Oncology, Illumina, Inc., San Diego, California
| | - Kristen Meier
- Department of Biostatistics, Illumina, Inc., San Diego, California
| | - Diane Ward
- Department of Biostatistics, Illumina, Inc., San Diego, California
| | - Jing Lu
- Department of Biostatistics, Illumina, Inc., San Diego, California
| | - Paul Wenz
- Department of Biostatistics, Illumina, Inc., San Diego, California
| | - Leonard Buchner
- Department of Biostatistics, Illumina, Inc., San Diego, California
| | - Tamsen Dunn
- Department of Bioinformatics, Illumina, Inc., San Diego, California
| | - Aaron Wise
- Department of Bioinformatics, Illumina, Inc., San Diego, California
| | - Amy Mueller
- Department of Medical Affairs, Illumina, Inc., San Diego, California
| | - Karen Gutekunst
- Department of Clinical Genomics Assay Development and Oncology, Illumina, Inc., San Diego, California
| |
Collapse
|
202
|
Reliability of Whole-Exome Sequencing for Assessing Intratumor Genetic Heterogeneity. Cell Rep 2019; 25:1446-1457. [PMID: 30404001 PMCID: PMC6261536 DOI: 10.1016/j.celrep.2018.10.046] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/20/2018] [Accepted: 10/11/2018] [Indexed: 12/11/2022] Open
Abstract
Multi-region sequencing is used to detect intratumor genetic heterogeneity (ITGH) in tumors. To assess whether genuine ITGH can be distinguished from sequencing artifacts, we performed whole-exome sequencing (WES) on three anatomically distinct regions of the same tumor with technical replicates to estimate technical noise. Somatic variants were detected with three different WES pipelines and subsequently validated by high-depth amplicon sequencing. The cancer-only pipeline was unreliable, with about 69% of the identified somatic variants being false positive. Even with matched normal DNA for which 82% of the somatic variants were detected reliably, only 36%-78% were found consistently in technical replicate pairs. Overall, 34%-80% of the discordant somatic variants, which could be interpreted as ITGH, were found to constitute technical noise. Excluding mutations affecting low-mappability regions or occurring in certain mutational contexts was found to reduce artifacts, yet detection of subclonal mutations by WES in the absence of orthogonal validation remains unreliable.
Collapse
|
203
|
Megquier K, Turner-Maier J, Swofford R, Kim JH, Sarver AL, Wang C, Sakthikumar S, Johnson J, Koltookian M, Lewellen M, Scott MC, Schulte AJ, Borst L, Tonomura N, Alfoldi J, Painter C, Thomas R, Karlsson EK, Breen M, Modiano JF, Elvers I, Lindblad-Toh K. Comparative Genomics Reveals Shared Mutational Landscape in Canine Hemangiosarcoma and Human Angiosarcoma. Mol Cancer Res 2019; 17:2410-2421. [PMID: 31570656 PMCID: PMC7067513 DOI: 10.1158/1541-7786.mcr-19-0221] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 07/12/2019] [Accepted: 09/25/2019] [Indexed: 12/23/2022]
Abstract
Angiosarcoma is a highly aggressive cancer of blood vessel-forming cells with few effective treatment options and high patient mortality. It is both rare and heterogenous, making large, well-powered genomic studies nearly impossible. Dogs commonly suffer from a similar cancer, called hemangiosarcoma, with breeds like the golden retriever carrying heritable genetic factors that put them at high risk. If the clinical similarity of canine hemangiosarcoma and human angiosarcoma reflects shared genomic etiology, dogs could be a critically needed model for advancing angiosarcoma research. We assessed the genomic landscape of canine hemangiosarcoma via whole-exome sequencing (47 golden retriever hemangiosarcomas) and RNA sequencing (74 hemangiosarcomas from multiple breeds). Somatic coding mutations occurred most frequently in the tumor suppressor TP53 (59.6% of cases) as well as two genes in the PI3K pathway: the oncogene PIK3CA (29.8%) and its regulatory subunit PIK3R1 (8.5%). The predominant mutational signature was the age-associated deamination of cytosine to thymine. As reported in human angiosarcoma, CDKN2A/B was recurrently deleted and VEGFA, KDR, and KIT recurrently gained. We compared the canine data to human data recently released by The Angiosarcoma Project, and found many of the same genes and pathways significantly enriched for somatic mutations, particularly in breast and visceral angiosarcomas. Canine hemangiosarcoma closely models the genomic landscape of human angiosarcoma of the breast and viscera, and is a powerful tool for investigating the pathogenesis of this devastating disease. IMPLICATIONS: We characterize the genomic landscape of canine hemangiosarcoma and demonstrate its similarity to human angiosarcoma.
Collapse
Affiliation(s)
- Kate Megquier
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts.
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | | | - Ross Swofford
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Jong-Hyuk Kim
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Aaron L Sarver
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Institute for Health Informatics, University of Minnesota, Minneapolis, Minnesota
| | - Chao Wang
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Sharadha Sakthikumar
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Jeremy Johnson
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | | | - Mitzi Lewellen
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Milcah C Scott
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Ashley J Schulte
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Luke Borst
- Department of Clinical Sciences, North Carolina State College of Veterinary Medicine, Raleigh, North Carolina
| | - Noriko Tonomura
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts
| | - Jessica Alfoldi
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Corrie Painter
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Count Me In, Cambridge, Massachusetts
| | - Rachael Thomas
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, and Comparative Medicine Institute, Raleigh, North Carolina
| | - Elinor K Karlsson
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, Massachusetts
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Matthew Breen
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, and Comparative Medicine Institute, Raleigh, North Carolina
| | - Jaime F Modiano
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Center for Immunology, University of Minnesota, Minneapolis, Minneapolis
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
- Institute for Engineering in Medicine, University of Minnesota, Minneapolis, Minnesota
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Ingegerd Elvers
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Kerstin Lindblad-Toh
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts.
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
204
|
Southwood M, Krenz T, Cant N, Maurya M, Gazdova J, Maxwell P, McGready C, Moseley E, Hughes S, Stewart P, Salto-Tellez M, Groelz D, Rassl D. Systematic evaluation of PAXgene® tissue fixation for the histopathological and molecular study of lung cancer. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2019; 6:40-54. [PMID: 31571426 PMCID: PMC6966705 DOI: 10.1002/cjp2.145] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/04/2019] [Accepted: 09/13/2019] [Indexed: 12/25/2022]
Abstract
Whilst adequate for most existing pathological tests, formalin is generally considered a poor DNA preservative and use of alternative fixatives may prove advantageous for molecular testing of tumour material; an increasingly common approach to identify targetable driver mutations in lung cancer patients. We collected paired PAXgene® tissue-fixed and formalin-fixed samples of block-sized tumour and lung parenchyma, Temno-needle core tumour biopsies and fine needle tumour aspirates (FNAs) from non-small cell lung cancer resection specimens. Traditionally processed formalin fixed paraffin wax embedded (FFPE) samples were compared to paired PAXgene® tissue fixed paraffin-embedded (PFPE) samples. We evaluated suitability for common laboratory tests (H&E staining and immunohistochemistry) and performance for downstream molecular investigations relevant to lung cancer, including RT-PCR and next generation DNA sequencing (NGS). Adequate and comparable H&E staining was seen in all sample types and nuclear staining was preferable in PAXgene® fixed Temno tumour biopsies and tumour FNA samples. Immunohistochemical staining was broadly comparable. PFPE samples enabled greater yields of less-fragmented DNA than FFPE comparators. PFPE samples were also superior for PCR and NGS performance, both in terms of quality control metrics and for variant calling. Critically we identified a greater number of genetic variants in the epidermal growth factor receptor gene when using PFPE samples and the Ingenuity® Variant Analysis pipeline. In summary, PFPE samples are adequate for histopathological diagnosis and suitable for the majority of existing laboratory tests. PAXgene® fixation is superior for DNA and RNA integrity, particularly in low-yield samples and facilitates improved NGS performance, including the detection of actionable lung cancer mutations for precision medicine in lung cancer samples.
Collapse
Affiliation(s)
- Mark Southwood
- Pathology Research, Royal Papworth Hospital NHS Foundation Trust, University of Cambridge Clinical School of Medicine, Cambridge, UK
| | - Tomasz Krenz
- Sample Technologies Department, QIAGEN GmbH, Hilden, Germany
| | - Natasha Cant
- Sample Technologies Department, QIAGEN Ltd., Manchester, UK
| | - Manisha Maurya
- Northern Ireland Molecular Pathology Laboratory, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Jana Gazdova
- Northern Ireland Molecular Pathology Laboratory, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Perry Maxwell
- Northern Ireland Molecular Pathology Laboratory, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Claire McGready
- Northern Ireland Molecular Pathology Laboratory, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Ellen Moseley
- Pathology Research, Royal Papworth Hospital NHS Foundation Trust, University of Cambridge Clinical School of Medicine, Cambridge, UK
| | - Susan Hughes
- Northern Ireland Molecular Pathology Laboratory, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Peter Stewart
- Northern Ireland Molecular Pathology Laboratory, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Manuel Salto-Tellez
- Northern Ireland Molecular Pathology Laboratory, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Daniel Groelz
- Sample Technologies Department, QIAGEN GmbH, Hilden, Germany
| | - Doris Rassl
- Pathology Research, Royal Papworth Hospital NHS Foundation Trust, University of Cambridge Clinical School of Medicine, Cambridge, UK
| | | |
Collapse
|
205
|
Mutation of the PTCH1 gene predicts recurrence of breast cancer. Sci Rep 2019; 9:16359. [PMID: 31704974 PMCID: PMC6841698 DOI: 10.1038/s41598-019-52617-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 10/21/2019] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the most common cancer in women, and some patients develop recurrence after standard therapy. Effective predictors are urgently needed to detect recurrence earlier. The activation of Hedgehog signaling in breast cancer is correlated with poor prognosis. PTCH1 is an essential membrane receptor of Hedgehog. However, there are few reports about mutations in Hedgehog genes in breast cancer. We conducted a comprehensive study via an experimental and bioinformatics approach to detect mutated genes in breast cancer. Twenty-two breast cancer patients who developed recurrence within 24 months postoperatively were enrolled with 22 control cancer patients. Targeted deep sequencing was performed to assess the mutations among individuals with breast cancer using a panel of 143 cancer-associated genes. Bioinformatics and public databases were used to predict the protein functions of the mutated genes. Mutations were identified in 44 breast cancer specimens, and the most frequently mutated genes were BRCA2, APC, ATM, BRCA1, NF1, TET2, TSC1, TSC2, NOTCH1, MSH2, PTCH1, TP53, PIK3CA, FBXW7, and RB1. Mutation of these genes was correlated with protein phosphorylation and autophosphorylation, such as peptidyl-tyrosine and protein kinase C phosphorylation. Among these highly mutated genes, mutations of PTCH1 were associated with poor prognosis and increased recurrence of breast cancer, especially mutations in exons 22 and 23. The public sequencing data from the COSMIC database were exploited to predict the functions of the mutations. Our findings suggest that mutation of PTCH1 is correlated with early recurrence of breast cancer patients and will become a powerful predictor for recurrence of breast cancer.
Collapse
|
206
|
Abstract
Background The incorporation of novel biomarkers into therapy selection for patients with metastatic colorectal cancer (mcrc) has significantly improved outcomes. Optimal treatment planning now takes into account diverse characteristics of patients and their tumours to create personalized therapeutic plans. Discussion This review is split into two sections. In the first section, we review the prognostic and predictive significance of expanded RAS mutation testing, BRAF mutations, ERBB2 (her2) amplification, microsatellite instability (msi) and deficient mismatch repair (dmmr) protein, NTRK fusions, PIK3CA mutations, and met amplifications. The therapeutic implication of each of those biomarkers for personalizing therapies for each patient with mcrc is discussed. In the second section, we touch on testing methods and considerations of relevance to clinicians when they interpret companion diagnostics meant to guide therapy selection. The advantages and pitfalls of various methods are evaluated, and we also look at the potential of liquid biopsies and circulating tumour dna (ctdna) to change the landscape of therapeutic choice and biologic understanding of the disease. Summary Routine testing for extended RAS, BRAF, dmmr or high msi, and NTRK fusions is necessary to determine the best sequencing of chemotherapy and biologic agents for patients with mcrc. Although next-generation sequencing and ctdna are increasingly being adopted, other techniques such as immunohistochemistry retain their relevance in detection of her2 amplification, NTRK fusions, and dmmr.
Collapse
|
207
|
Alborelli I, Leonards K, Rothschild SI, Leuenberger LP, Savic Prince S, Mertz KD, Poechtrager S, Buess M, Zippelius A, Läubli H, Haegele J, Tolnay M, Bubendorf L, Quagliata L, Jermann P. Tumor mutational burden assessed by targeted NGS predicts clinical benefit from immune checkpoint inhibitors in non-small cell lung cancer. J Pathol 2019; 250:19-29. [PMID: 31471895 PMCID: PMC6972587 DOI: 10.1002/path.5344] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 07/22/2019] [Accepted: 08/22/2019] [Indexed: 02/06/2023]
Abstract
In non‐small cell lung cancer (NSCLC), immune checkpoint inhibitors (ICIs) significantly improve overall survival (OS). Tumor mutational burden (TMB) has emerged as a predictive biomarker for patients treated with ICIs. Here, we evaluated the predictive power of TMB measured by the Oncomine™ Tumor Mutational Load targeted sequencing assay in 76 NSCLC patients treated with ICIs. TMB was assessed retrospectively in 76 NSCLC patients receiving ICI therapy. Clinical data (RECIST 1.1) were collected and patients were classified as having either durable clinical benefit (DCB) or no durable benefit (NDB). Additionally, genetic alterations and PD‐L1 expression were assessed and compared with TMB and response rate. TMB was significantly higher in patients with DCB than in patients with NDB (median TMB = 8.5 versus 6.0 mutations/Mb, Mann–Whitney p = 0.0244). 64% of patients with high TMB (cut‐off = third tertile, TMB ≥ 9) were responders (DCB) compared to 33% and 29% of patients with intermediate and low TMB, respectively (cut‐off = second and first tertile, TMB = 5–9 and TMB ≤ 4, respectively). TMB‐high patients showed significantly longer progression‐free survival (PFS) and OS (log‐rank test p = 0.0014 for PFS and 0.0197 for OS). While identifying different subgroups of patients, combining PD‐L1 expression and TMB increased the predictive power (from AUC 0.63 to AUC 0.65). Our results show that the TML panel is an effective tool to stratify patients for ICI treatment. A combination of biomarkers might maximize the predictive precision for patient stratification. Our study supports TMB evaluation through targeted NGS in NSCLC patient samples as a tool to predict response to ICI therapy. We offer recommendations for a reliable and cost‐effective assessment of TMB in a routine diagnostic setting. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Ilaria Alborelli
- Department of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Katharina Leonards
- Department of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Sacha I Rothschild
- Laboratory of Cancer Immunology, Department of Biomedicine, University Hospital Basel, Basel, Switzerland.,Department of Medical Oncology, Department of Internal Medicine, University Hospital Basel, Basel, Switzerland
| | - Laura P Leuenberger
- Department of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Spasenija Savic Prince
- Department of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Kirsten D Mertz
- Department of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | | | - Martin Buess
- Department of Medical Oncology, St. Claraspital, Basel, Switzerland
| | - Alfred Zippelius
- Laboratory of Cancer Immunology, Department of Biomedicine, University Hospital Basel, Basel, Switzerland.,Department of Medical Oncology, Department of Internal Medicine, University Hospital Basel, Basel, Switzerland
| | - Heinz Läubli
- Laboratory of Cancer Immunology, Department of Biomedicine, University Hospital Basel, Basel, Switzerland.,Department of Medical Oncology, Department of Internal Medicine, University Hospital Basel, Basel, Switzerland
| | - Jasmin Haegele
- Department of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Markus Tolnay
- Department of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Lukas Bubendorf
- Department of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Luca Quagliata
- Department of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Philip Jermann
- Department of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
208
|
Sim Y, Ng GXP, Ng CCY, Rajasegaran V, Wong SF, Liu W, Guan P, Nagarajan S, Ng WY, Thike AA, Lim JCT, Nasir NDBM, Tan VKM, Madhukumar P, Yong WS, Wong CY, Tan BKT, Ong KW, Teh BT, Tan PH. A novel genomic panel as an adjunctive diagnostic tool for the characterization and profiling of breast Fibroepithelial lesions. BMC Med Genomics 2019; 12:142. [PMID: 31647027 PMCID: PMC6813086 DOI: 10.1186/s12920-019-0588-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 09/23/2019] [Indexed: 02/08/2023] Open
Abstract
Background Known collectively as breast fibroepithelial lesions (FELs), the common fibroadenomas (FAs) and the rarer phyllodes tumors (PTs) are a heterogenous group of biphasic neoplasms. Owing to limited tissue availability, inter-observer variability, overlapping histological features and heterogeneity of these lesions, diagnosing them accurately on core biopsies is challenging. As the choice management option depends on the histological diagnosis; a novel 16-gene panel assay was developed to improve the accuracy of preoperative diagnosis on core biopsy specimens. Methods Using this 16-gene panel, targeted amplicon-based sequencing was performed on 275 formalin-fixed, paraffin-embedded (FFPE) breast FEL specimens, archived at the Singapore General Hospital, from 2008 to 2012. Results In total, 167 FAs, 24 benign, 14 borderline and 6 malignant PTs, were profiled. Compared to FAs, PTs had significantly higher mutation rates in the TERT promoter (p < 0.001), RARA (p < 0.001), FLNA, RB1 and TP53 (p = 0.002, 0.020 and 0.018, respectively). In addition to a higher mutational count (p < 0.001), TERT promoter (p < 0.001), frameshift, nonsense and splice site (p = 0.001, < 0.001 and 0.043, respectively) mutations were also frequently observed in PTs. A multivariate logistic regression model was built using these as variables and a predictive scoring system was developed. It classifies a FEL at low or high risk (score < 1 and ≥ 1, respectively) of being a PT. This scoring system has good discrimination (ROC area = 0.773, 95% CI: 0.70 to 0.85), calibration (p = 0.945) and is significant in predicting PTs (p < 0.001). Conclusion This novel study demonstrates the ability to extract DNA of sufficient quality and quantity for targeted sequencing from FFPE breast core biopsy specimens, along with their successful characterization and profiling using our customized 16-gene panel. Prospective work includes validating the utility of this promising 16-gene panel assay as an adjunctive diagnostic tool in clinical practice.
Collapse
Affiliation(s)
- Yirong Sim
- Division of Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore. .,SingHealth Duke-NUS Breast Centre, Singapore General Hospital, Singapore, Singapore.
| | - Gwendolene Xin Pei Ng
- Division of Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore.,Integrated Genomics Platform, National Cancer Centre Singapore, Singapore, Singapore.,Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore
| | - Cedric Chuan Young Ng
- Integrated Genomics Platform, National Cancer Centre Singapore, Singapore, Singapore.,Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore
| | - Vikneswari Rajasegaran
- Integrated Genomics Platform, National Cancer Centre Singapore, Singapore, Singapore.,Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore
| | - Suet Far Wong
- Integrated Genomics Platform, National Cancer Centre Singapore, Singapore, Singapore.,Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore
| | - Wei Liu
- Integrated Genomics Platform, National Cancer Centre Singapore, Singapore, Singapore.,Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore
| | - Peiyong Guan
- Integrated Biostatistics and Bioinformatics Program, Duke-NUS Medical School, Singapore, Singapore
| | - Sanjanaa Nagarajan
- Integrated Genomics Platform, National Cancer Centre Singapore, Singapore, Singapore.,Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore
| | - Wai Yee Ng
- Division of Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Aye Aye Thike
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - Jeffrey Chun Tatt Lim
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | | | - Veronique Kiak Mien Tan
- Division of Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore.,SingHealth Duke-NUS Breast Centre, Singapore General Hospital, Singapore, Singapore
| | - Preetha Madhukumar
- Division of Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore.,SingHealth Duke-NUS Breast Centre, Singapore General Hospital, Singapore, Singapore
| | - Wei Sean Yong
- Division of Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore.,SingHealth Duke-NUS Breast Centre, Singapore General Hospital, Singapore, Singapore
| | - Chow Yin Wong
- SingHealth Duke-NUS Breast Centre, Singapore General Hospital, Singapore, Singapore
| | - Benita Kiat Tee Tan
- Division of Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore.,SingHealth Duke-NUS Breast Centre, Singapore General Hospital, Singapore, Singapore
| | - Kong Wee Ong
- Division of Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore.,SingHealth Duke-NUS Breast Centre, Singapore General Hospital, Singapore, Singapore
| | - Bin Tean Teh
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Puay Hoon Tan
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore.,Division of Pathology, Singapore General Hospital, Singapore, Singapore
| |
Collapse
|
209
|
Moelans CB, de Ligt J, van der Groep P, Prins P, Besselink NJM, Hoogstraat M, Ter Hoeve ND, Lacle MM, Kornegoor R, van der Pol CC, de Leng WWJ, Barbé E, van der Vegt B, Martens J, Bult P, Smit VTHBM, Koudijs MJ, Nijman IJ, Voest EE, Selenica P, Weigelt B, Reis-Filho JS, van der Wall E, Cuppen E, van Diest PJ. The molecular genetic make-up of male breast cancer. Endocr Relat Cancer 2019; 26:779-794. [PMID: 31340200 PMCID: PMC6938562 DOI: 10.1530/erc-19-0278] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 07/23/2019] [Indexed: 12/17/2022]
Abstract
Male breast cancer (MBC) is extremely rare and accounts for less than 1% of all breast malignancies. Therefore, clinical management of MBC is currently guided by research on the disease in females. In this study, DNA obtained from 45 formalin-fixed paraffin-embedded (FFPE) MBCs with and 90 MBCs (52 FFPE and 38 fresh-frozen) without matched normal tissues was subjected to massively parallel sequencing targeting all exons of 1943 cancer-related genes. The landscape of mutations and copy number alterations was compared to that of publicly available estrogen receptor (ER)-positive female breast cancers (smFBCs) and correlated to prognosis. From the 135 MBCs, 90% showed ductal histology, 96% were ER-positive, 66% were progesterone receptor (PR)-positive, and 2% HER2-positive, resulting in 50, 46 and 4% luminal A-like, luminal B-like and basal-like cases, respectively. Five patients had Klinefelter syndrome (4%) and 11% of patients harbored pathogenic BRCA2 germline mutations. The genomic landscape of MBC to some extent recapitulated that of smFBC, with recurrent PIK3CA (36%) and GATA3 (15%) somatic mutations, and with 40% of the most frequently amplified genes overlapping between both sexes. TP53 (3%) somatic mutations were significantly less frequent in MBC compared to smFBC, whereas somatic mutations in genes regulating chromatin function and homologous recombination deficiency-related signatures were more prevalent. MDM2 amplifications were frequent (13%), correlated with protein overexpression (P = 0.001) and predicted poor outcome (P = 0.007). In conclusion, despite similarities in the genomic landscape between MBC and smFBC, MBC is a molecularly unique and heterogeneous disease requiring its own clinical trials and treatment guidelines.
Collapse
Affiliation(s)
- Cathy B Moelans
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Joep de Ligt
- Department of Biomedical Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Petra van der Groep
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Internal Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Pjotr Prins
- Department of Biomedical Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Nicolle J M Besselink
- Department of Biomedical Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Center for Personalized Cancer Treatment, Rotterdam, The Netherlands
| | - Marlous Hoogstraat
- Department of Biomedical Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Natalie D Ter Hoeve
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Miangela M Lacle
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Robert Kornegoor
- Department of Pathology, Gelre Ziekenhuizen, Appeldoorn, The Netherlands
| | - Carmen C van der Pol
- Cancer Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Wendy W J de Leng
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Ellis Barbé
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Bert van der Vegt
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - John Martens
- Department of Medical Oncology, Daniel den Hoed Cancer Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Peter Bult
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Marco J Koudijs
- Department of Biomedical Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Center for Personalized Cancer Treatment, Rotterdam, The Netherlands
| | - Isaac J Nijman
- Department of Biomedical Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Center for Personalized Cancer Treatment, Rotterdam, The Netherlands
| | - Emile E Voest
- Center for Personalized Cancer Treatment, Rotterdam, The Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Pier Selenica
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Elsken van der Wall
- Cancer Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Edwin Cuppen
- Department of Biomedical Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Cancer Genomics.nl, Center for Molecular Medicine, UMC Utrecht, Utrecht, The Netherlands
| | - Paul J van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
210
|
Solomon IH, Lin C, Horback KL, Kanjilal S, Rojas-Rudilla V, Brigl M, Laga AC, Lindeman NI, Padera RF. Utility of Histologic and Histochemical Screening for 16S Ribosomal RNA Gene Sequencing of Formalin-Fixed, Paraffin-Embedded Tissue for Bacterial Endocarditis. Am J Clin Pathol 2019; 152:431-437. [PMID: 31172185 DOI: 10.1093/ajcp/aqz055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVES 16S ribosomal RNA (rRNA) sequencing is a powerful but expensive tool for the identification of bacteria in culture-negative endocarditis. Histologic criteria to screen formalin-fixed, paraffin-embedded (FFPE) specimens for testing are evaluated. METHODS Sixty-eight cases of infective endocarditis and controls were histologically reviewed and analyzed by 16S rRNA gene sequencing. RESULTS Sequencing identified a specific pathogenic organism in 33 (49%) of 68 cases with acute inflammation and in 0 of 10 controls (P = .004). Visualization of organisms by Gram or Grocott methenamine silver stains had the strongest association with positive sequencing, while antibiotic treatment effect and acid decalcification decreased sensitivity. Molecular identifications were concordant with blood culture results in 90% of the cases, and a positive sequencing result was obtained in approximately half of the cases with negative valve cultures. CONCLUSIONS Histologic screening criteria are extremely helpful for identifying cases likely to be positive by molecular testing and can provide significant cost savings in filtering out low-yield specimens.
Collapse
Affiliation(s)
- Isaac H Solomon
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Chieyu Lin
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Katharine L Horback
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Sanjat Kanjilal
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Vanesa Rojas-Rudilla
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Manfred Brigl
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Alvaro C Laga
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Neal I Lindeman
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Robert F Padera
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
211
|
Bhagwate AV, Liu Y, Winham SJ, McDonough SJ, Stallings-Mann ML, Heinzen EP, Davila JI, Vierkant RA, Hoskin TL, Frost M, Carter JM, Radisky DC, Cunningham JM, Degnim AC, Wang C. Bioinformatics and DNA-extraction strategies to reliably detect genetic variants from FFPE breast tissue samples. BMC Genomics 2019; 20:689. [PMID: 31477010 PMCID: PMC6720378 DOI: 10.1186/s12864-019-6056-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 08/22/2019] [Indexed: 01/20/2023] Open
Abstract
Background Archived formalin fixed paraffin embedded (FFPE) samples are valuable clinical resources to examine clinically relevant morphology features and also to study genetic changes. However, DNA quality and quantity of FFPE samples are often sub-optimal, and resulting NGS-based genetics variant detections are prone to false positives. Evaluations of wet-lab and bioinformatics approaches are needed to optimize variant detection from FFPE samples. Results As a pilot study, we designed within-subject triplicate samples of DNA derived from paired FFPE and fresh frozen breast tissues to highlight FFPE-specific artifacts. For FFPE samples, we tested two FFPE DNA extraction methods to determine impact of wet-lab procedures on variant calling: QIAGEN QIAamp DNA Mini Kit (“QA”), and QIAGEN GeneRead DNA FFPE Kit (“QGR”). We also used negative-control (NA12891) and positive control samples (Horizon Discovery Reference Standard FFPE). All DNA sample libraries were prepared for NGS according to the QIAseq Human Breast Cancer Targeted DNA Panel protocol and sequenced on the HiSeq 4000. Variant calling and filtering were performed using QIAGEN Gene Globe Data Portal. Detailed variant concordance comparisons and mutational signature analysis were performed to investigate effects of FFPE samples compared to paired fresh frozen samples, along with different DNA extraction methods. In this study, we found that five times or more variants were called with FFPE samples, compared to their paired fresh-frozen tissue samples even after applying molecular barcoding error-correction and default bioinformatics filtering recommended by the vendor. We also found that QGR as an optimized FFPE-DNA extraction approach leads to much fewer discordant variants between paired fresh frozen and FFPE samples. Approximately 92% of the uniquely called FFPE variants were of low allelic frequency range (< 5%), and collectively shared a “C > T|G > A” mutational signature known to be representative of FFPE artifacts resulting from cytosine deamination. Based on control samples and FFPE-frozen replicates, we derived an effective filtering strategy with associated empirical false-discovery estimates. Conclusions Through this study, we demonstrated feasibility of calling and filtering genetic variants from FFPE tissue samples using a combined strategy with molecular barcodes, optimized DNA extraction, and bioinformatics methods incorporating genomics context such as mutational signature and variant allelic frequency. Electronic supplementary material The online version of this article (10.1186/s12864-019-6056-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Aditya Vijay Bhagwate
- Departments of Health Science Research, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Yuanhang Liu
- Departments of Health Science Research, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Stacey J Winham
- Departments of Health Science Research, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Samantha J McDonough
- Departments of Laboratory Medicine and Pathology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | | | - Ethan P Heinzen
- Departments of Health Science Research, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Jaime I Davila
- Departments of Health Science Research, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Robert A Vierkant
- Departments of Health Science Research, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Tanya L Hoskin
- Departments of Health Science Research, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Marlene Frost
- Departments of Medical Oncology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Jodi M Carter
- Departments of Laboratory Medicine and Pathology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Derek C Radisky
- Departments of Cancer Biology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Julie M Cunningham
- Departments of Laboratory Medicine and Pathology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Amy C Degnim
- Departments of Surgery, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Chen Wang
- Departments of Health Science Research, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
212
|
Haile S, Corbett RD, Bilobram S, Bye MH, Kirk H, Pandoh P, Trinh E, MacLeod T, McDonald H, Bala M, Miller D, Novik K, Coope RJ, Moore RA, Zhao Y, Mungall AJ, Ma Y, Holt RA, Jones SJ, Marra MA. Sources of erroneous sequences and artifact chimeric reads in next generation sequencing of genomic DNA from formalin-fixed paraffin-embedded samples. Nucleic Acids Res 2019; 47:e12. [PMID: 30418619 PMCID: PMC6344851 DOI: 10.1093/nar/gky1142] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 11/06/2018] [Indexed: 01/01/2023] Open
Abstract
Tissues used in pathology laboratories are typically stored in the form of formalin-fixed, paraffin-embedded (FFPE) samples. One important consideration in repurposing FFPE material for next generation sequencing (NGS) analysis is the sequencing artifacts that can arise from the significant damage to nucleic acids due to treatment with formalin, storage at room temperature and extraction. One such class of artifacts consists of chimeric reads that appear to be derived from non-contiguous portions of the genome. Here, we show that a major proportion of such chimeric reads align to both the ‘Watson’ and ‘Crick’ strands of the reference genome. We refer to these as strand-split artifact reads (SSARs). This study provides a conceptual framework for the mechanistic basis of the genesis of SSARs and other chimeric artifacts along with supporting experimental evidence, which have led to approaches to reduce the levels of such artifacts. We demonstrate that one of these approaches, involving S1 nuclease-mediated removal of single-stranded fragments and overhangs, also reduces sequence bias, base error rates, and false positive detection of copy number and single nucleotide variants. Finally, we describe an analytical approach for quantifying SSARs from NGS data.
Collapse
Affiliation(s)
- Simon Haile
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Richard D Corbett
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Steve Bilobram
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Morgan H Bye
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Heather Kirk
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Pawan Pandoh
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Eva Trinh
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Tina MacLeod
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Helen McDonald
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Miruna Bala
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Diane Miller
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Karen Novik
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Robin J Coope
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Richard A Moore
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Yongjun Zhao
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Andrew J Mungall
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Yussanne Ma
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Rob A Holt
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Steven J Jones
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
213
|
Wu S, Shi X, Si X, Liu Y, Lu T, Zhang L, Liang Z, Zeng X. EGFR T790M detection in formalin-fixed paraffin-embedded tissues of patients with lung cancer using RNA-based in situ hybridization: A preliminary feasibility study. Thorac Cancer 2019; 10:1936-1944. [PMID: 31407509 PMCID: PMC6775006 DOI: 10.1111/1759-7714.13169] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/28/2019] [Accepted: 07/29/2019] [Indexed: 12/13/2022] Open
Abstract
Background Following drug resistance in patients with lung cancer treated by EGFR TKIs, a biopsy is required to obtain sufficient cancer tissue for T790M detection in order to select potential beneficiaries suitable for third‐generation EGFR TKIs, such as osimertinib. The purpose of this study was to explore the feasibility of using a new in situ analysis technique based on RNA target sequences to detect EGFR T790M in lung cancer. Methods A total of 28 formalin‐fixed paraffin‐embedded (FFPE) samples from 24 lung adenocarcinoma patients archived in Peking Union Medical College Hospital from 2015 to 2016 were collected. The BaseScope T790M detection technique by in situ hybridization on FFPE slides was used to analyze the mutation of EGFR T790M, and then the results were compared with the data acquired by Scorpions ARMS assay, which is the so‐called gold standard for EGFR gene mutation testing. The sensitivity and specificity of the BaseScope T790M detection technique were preliminarily evaluated. Results Of the 28 FFPE specimens, the average proportion of T790M‐positive cells was 35.78% ± 17.68% in 18 samples with EGFR T790M, confirmed by Scorpions ARMS assay, Compared with real‐time PCR assay, the sensitivity and specificity of BaseScope T790M were all 100% in our cohort. Conclusion BaseScope T790M assay could be completed on only one FFPE slide and the visualized molecular result overplayed with histomorphological information perfectly, so it may be the alternative method for EGFR T790M evaluation. BaseScope assay has potential clinical utility, and it will be necessary to carry out validation with a large number of cases.
Collapse
Affiliation(s)
- Shafei Wu
- Molecular Pathology Research Center, Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaohua Shi
- Molecular Pathology Research Center, Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoyan Si
- Department of Respiratory Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuanyuan Liu
- Molecular Pathology Research Center, Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Tao Lu
- Molecular Pathology Research Center, Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Li Zhang
- Department of Respiratory Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhiyong Liang
- Molecular Pathology Research Center, Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuan Zeng
- Molecular Pathology Research Center, Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
214
|
Pyrosequencing versus methylation-specific PCR for assessment of MGMT methylation in tumor and blood samples of glioblastoma patients. Sci Rep 2019; 9:11125. [PMID: 31366977 PMCID: PMC6668570 DOI: 10.1038/s41598-019-47642-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/16/2019] [Indexed: 12/20/2022] Open
Abstract
Circulating biomarkers in blood may provide an interesting alternative to risky tissue biopsies in the diagnosis and follow-up of glioblastoma patients. We have assessed MGMT methylation status in blood and tissue samples from unresected glioblastoma patients who had been included in the randomized GENOM-009 trial. Paired blood and tissue samples were assessed by methylation-specific PCR (MSP) and pyrosequencing (PYR). After establishing the minimum PYR cut-off that could yield a significant difference in overall survival, we assessed the sensitivity, specificity, positive predictive value and negative predictive value (NPV) of the analyses. Methylation could be detected in cfDNA by both MSP and PYR but with low concordance with results in tissue. Sensitivity was low for both methods (31% and 38%, respectively), while specificity was higher for MSP in blood than for PYR in plasma (96% vs 76%) and NPV was similar (56 vs 57%). Concordance of results in tissue by MSP and PYR was 84.3% (P < 0.001) and correlated with outcome. We conclude that detection of cfDNA in the blood of glioblastoma patients can be an alternative when tumor tissue is not available but methods for the detection of cfDNA in blood must improve before it can replace analysis in tumor tissue.
Collapse
|
215
|
Kinsler VA, Boccara O, Fraitag S, Torrelo A, Vabres P, Diociaiuti A. Mosaic abnormalities of the skin: review and guidelines from the European Reference Network for rare skin diseases. Br J Dermatol 2019; 182:552-563. [PMID: 30920652 DOI: 10.1111/bjd.17924] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Cutaneous mosaicism is an area of dermatology in which there has been an explosion of knowledge within the current decade. This has led to fundamental changes in the understanding of the conditions in this field, and to an ongoing paradigm shift in the approach to management of mosaic skin disorders. OBJECTIVES To lay out the general principles of mosaicism as they are currently understood, summarize the known cutaneous mosaic abnormalities of the skin with associated phenotypic and genotypic information, review the latest trials on targeted therapies and propose guidelines for the general approach to a patient with suspected mosaicism. METHODS This was a consensus expert review as part of the European Reference Network project (ERN-Skin). CONCLUSIONS This study provides clinicians with a practical approach to the patient with suspected mosaicism, redefines mosaicism for the modern genetic era, and proposes a new classification system based on genetic mechanism. What's already known about this topic? Cutaneous mosaicism is a complex field of dermatology that encompasses most birthmarks, and many rare syndromes. Some cutaneous patterns are known to be seen in mosaicism. Very few treatment options are available for most mosaic abnormalities of the skin. Recent high-sensitivity genetic techniques have led to an explosion of knowledge about genotype and phenotype in the literature. What does this study add? Expert consensus from the European Reference Network project. Review of knowledge of confirmed mosaic abnormalities of the skin, including cutaneous phenotype, extracutaneous associated features and genotype. Proposed new classification of mosaic abnormalities of the skin by genetic mechanism and therefore inheritance potential. Practical tips on correct sample collection and genetic investigation. Review of trials of targeted therapies. Guidelines for a practical clinical approach to the patient with suspected mosaicism.
Collapse
Affiliation(s)
- V A Kinsler
- Paediatric Dermatology, Great Ormond Street Hospital for Children, London, U.K.,Genetics and Genomic Medicine, UCL Institute of Child Health, London, U.K
| | - O Boccara
- Department of Dermatology and Reference Centre for Genodermatoses and Rare Skin Diseases (MAGEC), Université Paris Descartes - Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - S Fraitag
- Department of Pathology, Hôpital Universitaire Necker-Enfants Malades, APHP, Paris, France
| | - A Torrelo
- Department of Dermatology, Hospital Infantil del Niño Jesús, Madrid, Spain
| | - P Vabres
- Department of Dermatology and Reference Centre for Rare Skin Diseases, Dijon University Hospital, Dijon, France.,GAD, Genetics of Anomalies of Development, University of Bourgogne, Dijon, France
| | - A Diociaiuti
- Dermatology Unit, Bambino Gesù Children's Hospital, Rome, Italy
| |
Collapse
|
216
|
Berra CM, Torrezan GT, de Paula CA, Hsieh R, Lourenço SV, Carraro DM. Use of uracil-DNA glycosylase enzyme to reduce DNA-related artifacts from formalin-fixed and paraffin-embedded tissues in diagnostic routine. ACTA ACUST UNITED AC 2019. [DOI: 10.1186/s41241-019-0075-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
217
|
Dudez O, Dalstein V, Kanagaratnam L, Nasri S, Coquelet C, Fichel C, Bouland N, Lemaire E, Diebold MD, Marchal-Bressenot A, Boulagnon-Rombi C. Is the Mirror Image Method Really Useful in Tumor Tissue Bank Quality Control? Biopreserv Biobank 2019; 17:539-545. [PMID: 31233333 DOI: 10.1089/bio.2018.0123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Oncology research projects are highly dependent on the quality of tumor samples stored in the biobank. Microscopic control is important to ensure the quality of the frozen sample (Does the sample correspond to tumor tissue? Does the sample contain a sufficient number of tumor cells for molecular analysis?). The aim of this study was to evaluate the value of the mirror image method in quality control of colonic adenocarcinoma samples stored in a tumor bank. Microscopic concordance for the differentiation grade, malignant and normal cell percentages, necrosis, mucinous component, and ulceration was assessed on 82 colon adenocarcinoma banked samples and their paired, formalin-fixed, paraffin-embedded mirror controls. Molecular concordance for KRAS status was evaluated in 76 of these 82 cases. Morphological correspondence between frozen and mirror samples was good for the mucinous component (intraclass correlation coefficient [ICC] = 0.81), moderate for differentiation (Cohen's kappa coefficient [k] = 0.67), fair for malignant cells (ICC = 0.44), and poor for ulceration (k = 0.08), normal tissue (ICC = 0.36), and necrosis (ICC = 0.13) percentages. Molecular correspondence for KRAS status was almost perfect (95% correspondence, k = 0.88) between frozen and mirror samples. In conclusion, the mirror sample method is not a good alternative for microscopic and molecular control of frozen colonic adenocarcinoma samples.
Collapse
Affiliation(s)
- Oriane Dudez
- Pathology Department, Academic Hospital, Reims, France
| | - Véronique Dalstein
- Pathology Department, Academic Hospital, Reims, France.,Champagne Ardenne Cancers' Molecular Genetic Platform, Reims, France.,Inserm UMR-S 903, Université de Reims Champagne Ardenne, Reims, France
| | | | - Saviz Nasri
- Champagne Ardenne Biobank, Academic Hospital, Reims, France
| | | | | | - Nicole Bouland
- Pathology Department, Medicine University, Reims, France
| | - Emeric Lemaire
- Pathology Department, Academic Hospital, Reims, France.,Inserm UMR-S 903, Université de Reims Champagne Ardenne, Reims, France
| | - Marie-Danièle Diebold
- Pathology Department, Academic Hospital, Reims, France.,Champagne Ardenne Biobank, Academic Hospital, Reims, France.,Pathology Department, Medicine University, Reims, France
| | - Aude Marchal-Bressenot
- Pathology Department, Academic Hospital, Reims, France.,Pathology Department, Medicine University, Reims, France
| | - Camille Boulagnon-Rombi
- Pathology Department, Academic Hospital, Reims, France.,Pathology Department, Medicine University, Reims, France.,UMR CNRS 7369, Reims University, Reims, France
| |
Collapse
|
218
|
Berner AM, Morrissey GJ, Murugaesu N. Clinical Analysis of Whole Genome Sequencing in Cancer Patients. CURRENT GENETIC MEDICINE REPORTS 2019. [DOI: 10.1007/s40142-019-00169-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
219
|
Quy PN, Kanai M, Fukuyama K, Kou T, Kondo T, Yamamoto Y, Matsubara J, Hiroshima A, Mochizuki H, Sakuma T, Kamada M, Nakatsui M, Eso Y, Seno H, Masui T, Takaori K, Minamiguchi S, Matsumoto S, Muto M. Association Between Preanalytical Factors and Tumor Mutational Burden Estimated by Next-Generation Sequencing-Based Multiplex Gene Panel Assay. Oncologist 2019; 24:e1401-e1408. [PMID: 31186376 DOI: 10.1634/theoncologist.2018-0587] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 05/09/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Tumor mutational burden (TMB) measured via next-generation sequencing (NGS)-based gene panel is a promising biomarker for response to immune checkpoint inhibitors (ICIs) in solid tumors. However, little is known about the preanalytical factors that can affect the TMB score. MATERIALS AND METHODS Data of 199 patients with solid tumors who underwent multiplex NGS gene panel (OncoPrime), which was commercially provided by a Clinical Laboratory Improvement Amendments-licensed laboratory and covered 0.78 megabase (Mb) of capture size relevant to the TMB calculation, were reviewed. Associations between the TMB score and preanalytical factors, including sample DNA quality, sample type, sampling site, and storage period, were analyzed. Clinical outcomes of patients with a high TMB score (≥10 mutations per megabase) who received anti-programmed cell death protein 1 antibodies (n = 22) were also analyzed. RESULTS Low DNA library concentration (<5 nM), formalin-fixed paraffin-embedded tissue (FFPE), and the prolonged sample storage period (range, 0.9-58.1 months) correlated with a higher TMB score. After excluding low DNA library samples from the analysis, FFPE samples, but not the sample storage period, exhibited a marked correlation with a high TMB score. Of 22 patients with a high TMB score, we observed the partial response in 2 patients (9.1%). CONCLUSION Our results indicate that the TMB score estimated via NGS-based gene panel could be affected by the DNA library concentration and sample type. These factors could potentially increase the false-positive and/or artifactual variant calls. As each gene panel has its own pipeline for variant calling, it is unknown whether these factors have a significant effect in other platforms. IMPLICATIONS FOR PRACTICE A high tumor mutational burden score, as estimated via next-generation sequencing-based gene panel testing, should be carefully interpreted as it could be affected by the DNA library concentration and sample type.
Collapse
Affiliation(s)
- Pham Nguyen Quy
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masashi Kanai
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keita Fukuyama
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tadayuki Kou
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomohiro Kondo
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshihiro Yamamoto
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Junichi Matsubara
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akinori Hiroshima
- Biomedical Department, Mitsui Knowledge Industry Co., Ltd., Tokyo, Japan
| | - Hiroaki Mochizuki
- Biomedical Department, Mitsui Knowledge Industry Co., Ltd., Tokyo, Japan
| | - Tomohiro Sakuma
- Biomedical Department, Mitsui Knowledge Industry Co., Ltd., Tokyo, Japan
| | - Mayumi Kamada
- Department of Biomedical Data Intelligence, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masahiko Nakatsui
- Department of Biomedical Data Intelligence, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuji Eso
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toshihiko Masui
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kyoichi Takaori
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Sachiko Minamiguchi
- Department of Diagnostic Pathology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shigemi Matsumoto
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Manabu Muto
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
220
|
Zar G, Smith JG, Smith ML, Andersson B, Nilsson J. Whole-genome sequencing based on formalin-fixed paraffin-embedded endomyocardial biopsies for genetic studies on outcomes after heart transplantation. PLoS One 2019; 14:e0217747. [PMID: 31166960 PMCID: PMC6550412 DOI: 10.1371/journal.pone.0217747] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 05/14/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Whole-genome sequencing (WGS) of heart transplant recipient- and donor-derived cardiac biopsies may facilitate organ matching, graft failure prediction, and immunotolerance research. The objective of this study was to determine the feasibility of WGS based on formalin-fixed paraffin-embedded endomyocardial biopsies. METHODS AND RESULTS The study included serial donor- and recipient samples from patients who had undergone heart transplantation at Skane University Hospital, Lund, Sweden, between 1988 and 2009. DNA extraction and WGS were conducted. Additional WGS sequencing quality metrics and coverage were obtained with the Genome Analysis Toolkit (GATK). 455 endomyocardial samples from 37 heart transplant recipients were acquired from routine rejection monitoring and stored as formalin-fixed paraffin-embedded samples. They were analyzed after 3-26 years of storage. DNA was extracted from 114 samples and WGS was run on 85 samples. DNA extraction yielded 313 ng (IQR 96-601) for all samples. A coverage of 11.3x (IQR 9.0-15.9) was recorded for all WGS samples. Three samples stored for > 25 years yielded a coverage of > 25x. Data were generated for 1.7 billion reads per sample (IQR 1.4-2.7). A Transition/Transversion (TiTv) ratio of 2.09 ± 0.05 was calculated for all WGS samples. No associations were found among storage time, DNA yield, or sequencing quality metrics. CONCLUSIONS The present study demonstrated the feasibility of whole-genome sequencing based on endomyocardial biopsies. This process could enable large-scale retrospective genomic studies using stored histopathological samples.
Collapse
Affiliation(s)
- Gustav Zar
- Department of Clinical Sciences Lund, Cardiothoracic Surgery, Lund University and Skane University Hospital, Lund, Sweden
| | - J. Gustav Smith
- Department of Clinical Sciences, Cardiology, Lund University and Skane University Hospital, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Maya Landenhed Smith
- Department of Clinical Sciences Lund, Cardiothoracic Surgery, Lund University and Skane University Hospital, Lund, Sweden
| | - Bodil Andersson
- Department of Clinical Sciences Lund, Surgery, Lund University and Skane University Hospital, Lund, Sweden
| | - Johan Nilsson
- Department of Clinical Sciences Lund, Cardiothoracic Surgery, Lund University and Skane University Hospital, Lund, Sweden
- * E-mail:
| |
Collapse
|
221
|
Georgeson P, Walsh MD, Clendenning M, Daneshvar S, Pope BJ, Mahmood K, Joo JE, Jayasekara H, Jenkins MA, Winship IM, Buchanan DD. Tumor mutational signatures in sebaceous skin lesions from individuals with Lynch syndrome. Mol Genet Genomic Med 2019; 7:e00781. [PMID: 31162827 PMCID: PMC6625139 DOI: 10.1002/mgg3.781] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 05/16/2019] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Muir-Torre syndrome is defined by the development of sebaceous skin lesions in individuals who carry a germline mismatch repair (MMR) gene mutation. Loss of expression of MMR proteins is frequently observed in sebaceous skin lesions, but MMR-deficiency alone is not diagnostic for carrying a germline MMR gene mutation. METHODS Whole exome sequencing was performed on three MMR-deficient sebaceous lesions from individuals with MSH2 gene mutations (Lynch syndrome) and three MMR-proficient sebaceous lesions from individuals without Lynch syndrome with the aim of characterizing the tumor mutational signatures, somatic mutation burden, and microsatellite instability status. Thirty predefined somatic mutational signatures were calculated for each lesion. RESULTS Signature 1 was ubiquitous across the six lesions tested. Signatures 6 and 15, associated with defective DNA MMR, were significantly more prevalent in the MMR-deficient lesions from the MSH2 carriers compared with the MMR-proficient non-Lynch sebaceous lesions (mean ± SD=41.0 ± 8.2% vs. 2.3 ± 4.0%, p = 0.0018). Tumor mutation burden was, on average, significantly higher in the MMR-deficient lesions compared with the MMR-proficient lesions (23.3 ± 11.4 vs. 1.8 ± 0.8 mutations/Mb, p = 0.03). All four sebaceous lesions observed in sun exposed areas of the body demonstrated signature 7 related to ultraviolet light exposure. CONCLUSION Tumor mutational signatures 6 and 15 and somatic mutation burden were effective in differentiating Lynch-related from non-Lynch sebaceous lesions.
Collapse
Affiliation(s)
- Peter Georgeson
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, Vic., Australia.,Victorian Comprehensive Cancer Centre, University of Melbourne Centre for Cancer Research, Parkville, Vic., Australia
| | | | - Mark Clendenning
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, Vic., Australia.,Victorian Comprehensive Cancer Centre, University of Melbourne Centre for Cancer Research, Parkville, Vic., Australia
| | - Simin Daneshvar
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, Vic., Australia.,Dorevitch Pathology, Frankston Hospital, Frankston, Vic., Australia
| | - Bernard J Pope
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, Vic., Australia.,Melbourne Bioinformatics, The University of Melbourne, Carlton, Vic., Australia
| | - Khalid Mahmood
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, Vic., Australia.,Melbourne Bioinformatics, The University of Melbourne, Carlton, Vic., Australia
| | - Jihoon E Joo
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, Vic., Australia.,Victorian Comprehensive Cancer Centre, University of Melbourne Centre for Cancer Research, Parkville, Vic., Australia
| | - Harindra Jayasekara
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, Vic., Australia.,Victorian Comprehensive Cancer Centre, University of Melbourne Centre for Cancer Research, Parkville, Vic., Australia.,Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, Vic., Australia.,Centre for Alcohol Policy Research, La Trobe University, Melbourne, Vic., Australia
| | - Mark A Jenkins
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Carlton, Vic., Australia
| | - Ingrid M Winship
- Department of Medicine, The University of Melbourne, Parkville, Vic., Australia.,Genomic Medicine and Family Cancer Clinic, Royal Melbourne Hospital, Parkville, Vic., Australia
| | - Daniel D Buchanan
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, Vic., Australia.,Victorian Comprehensive Cancer Centre, University of Melbourne Centre for Cancer Research, Parkville, Vic., Australia.,Genomic Medicine and Family Cancer Clinic, Royal Melbourne Hospital, Parkville, Vic., Australia
| |
Collapse
|
222
|
Maura F, Agnelli L, Leongamornlert D, Bolli N, Chan WC, Dodero A, Carniti C, Heavican TB, Pellegrinelli A, Pruneri G, Butler A, Bhosle SG, Chiappella A, Di Rocco A, Zinzani PL, Zaja F, Piva R, Inghirami G, Wang W, Palomero T, Iqbal J, Neri A, Campbell PJ, Corradini P. Integration of transcriptional and mutational data simplifies the stratification of peripheral T-cell lymphoma. Am J Hematol 2019; 94:628-634. [PMID: 30829413 DOI: 10.1002/ajh.25450] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 02/25/2019] [Indexed: 12/21/2022]
Abstract
The histological diagnosis of peripheral T-cell lymphoma (PTCL) can represent a challenge, particularly in the case of closely related entities such as angioimmunoblastic T-lymphoma (AITL), PTCL-not otherwise specified (PTCL-NOS), and ALK-negative anaplastic large-cell lymphoma (ALCL). Although gene expression profiling and next generations sequencing have been proven to define specific features recurrently associated with distinct entities, genomic-based stratifications have not yet led to definitive diagnostic criteria and/or entered into the routine clinical practice. Herein, to improve the current molecular classification between AITL and PTCL-NOS, we analyzed the transcriptional profiles from 503 PTCLs stratified according to their molecular configuration and integrated them with genomic data of recurrently mutated genes (RHOA G17V , TET2, IDH2 R172 , and DNMT3A) in 53 cases (39 AITLs and 14 PTCL-NOSs) included in the series. Our analysis unraveled that the mutational status of RHOA G17V , TET2, and DNMT3A poorly correlated, individually, with peculiar transcriptional fingerprints. Conversely, in IDH2 R172 samples a strong transcriptional signature was identified that could act as a surrogate for mutational status. The integrated analysis of clinical, mutational, and molecular data led to a simplified 19-gene signature that retains high accuracy in differentiating the main nodal PTCL entities. The expression levels of those genes were confirmed in an independent cohort profiled by RNA-sequencing.
Collapse
Affiliation(s)
- Francesco Maura
- Myeloma Service, Department of MedicineMemorial Sloan Kettering Cancer Center New York New York
- Cancer, Ageing and Somatic Mutation ProgrammeWellcome Trust Sanger Institute Hinxton United Kingdom
- Department of Oncology and Hemato‐OncologyUniversity of Milan Milan Italy
| | - Luca Agnelli
- Department of Oncology and Hemato‐OncologyUniversity of Milan Milan Italy
- HematologyFoundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico Milan Italy
| | - Daniel Leongamornlert
- Cancer, Ageing and Somatic Mutation ProgrammeWellcome Trust Sanger Institute Hinxton United Kingdom
| | - Niccolò Bolli
- Cancer, Ageing and Somatic Mutation ProgrammeWellcome Trust Sanger Institute Hinxton United Kingdom
- Department of Oncology and Hemato‐OncologyUniversity of Milan Milan Italy
- Division of HematologyFondazione IRCCS Istituto Nazionale dei Tumori Milan Italy
| | - Wing C. Chan
- Department of PathologyCity of Hope National Medical Center Duarte California
| | - Anna Dodero
- Division of HematologyFondazione IRCCS Istituto Nazionale dei Tumori Milan Italy
| | - Cristiana Carniti
- Division of HematologyFondazione IRCCS Istituto Nazionale dei Tumori Milan Italy
| | - Tayla B. Heavican
- Department of Pathology and MicrobiologyUniversity of Nebraska Medical Center Omaha Nebraska
| | - Alessio Pellegrinelli
- Department of Pathology and Laboratory MedicineFondazione IRCCS Istituto Nazionale dei Tumori Milan Italy
| | - Giancarlo Pruneri
- Department of Pathology and Laboratory MedicineFondazione IRCCS Istituto Nazionale dei Tumori Milan Italy
| | - Adam Butler
- Cancer, Ageing and Somatic Mutation ProgrammeWellcome Trust Sanger Institute Hinxton United Kingdom
| | - Shriram G. Bhosle
- Cancer, Ageing and Somatic Mutation ProgrammeWellcome Trust Sanger Institute Hinxton United Kingdom
| | - Annalisa Chiappella
- Department of HematologyAzienda Ospedaliera Città della Salute e della Scienza Torino Italy
| | - Alice Di Rocco
- Department of Cellular Biotechnology and Hematology, Sapienza University of Rome Rome Italy
| | | | - Francesco Zaja
- Clinical Ematologica, DAMEUniversity of Udine Udine Italy
| | - Roberto Piva
- Department of Molecular Biotechnology and Health SciencesCenter for Experimental Research and Medical Studies, University of Torino Torino Italy
| | - Giorgio Inghirami
- Department of Molecular Biotechnology and Health SciencesCenter for Experimental Research and Medical Studies, University of Torino Torino Italy
- Pathology and Laboratory MedicinesWeill Cornell Medical College New York New York
| | - Wenyi Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center Houston Texas
| | - Teresa Palomero
- Institute for Cancer Genetics, Columbia University New York New York
| | - Javeed Iqbal
- Department of Pathology and MicrobiologyUniversity of Nebraska Medical Center Omaha Nebraska
| | - Antonino Neri
- Department of Oncology and Hemato‐OncologyUniversity of Milan Milan Italy
- HematologyFoundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico Milan Italy
| | - Peter J. Campbell
- Cancer, Ageing and Somatic Mutation ProgrammeWellcome Trust Sanger Institute Hinxton United Kingdom
| | - Paolo Corradini
- Department of Oncology and Hemato‐OncologyUniversity of Milan Milan Italy
- Division of HematologyFondazione IRCCS Istituto Nazionale dei Tumori Milan Italy
| |
Collapse
|
223
|
Baker AM, Cross W, Curtius K, Al Bakir I, Choi CHR, Davis HL, Temko D, Biswas S, Martinez P, Williams MJ, Lindsay JO, Feakins R, Vega R, Hayes SJ, Tomlinson IPM, McDonald SAC, Moorghen M, Silver A, East JE, Wright NA, Wang LM, Rodriguez-Justo M, Jansen M, Hart AL, Leedham SJ, Graham TA. Evolutionary history of human colitis-associated colorectal cancer. Gut 2019; 68:985-995. [PMID: 29991641 PMCID: PMC6580738 DOI: 10.1136/gutjnl-2018-316191] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 06/01/2018] [Accepted: 06/02/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE IBD confers an increased lifetime risk of developing colorectal cancer (CRC), and colitis-associated CRC (CA-CRC) is molecularly distinct from sporadic CRC (S-CRC). Here we have dissected the evolutionary history of CA-CRC using multiregion sequencing. DESIGN Exome sequencing was performed on fresh-frozen multiple regions of carcinoma, adjacent non-cancerous mucosa and blood from 12 patients with CA-CRC (n=55 exomes), and key variants were validated with orthogonal methods. Genome-wide copy number profiling was performed using single nucleotide polymorphism arrays and low-pass whole genome sequencing on archival non-dysplastic mucosa (n=9), low-grade dysplasia (LGD; n=30), high-grade dysplasia (HGD; n=13), mixed LGD/HGD (n=7) and CA-CRC (n=19). Phylogenetic trees were reconstructed, and evolutionary analysis used to reveal the temporal sequence of events leading to CA-CRC. RESULTS 10/12 tumours were microsatellite stable with a median mutation burden of 3.0 single nucleotide alterations (SNA) per Mb, ~20% higher than S-CRC (2.5 SNAs/Mb), and consistent with elevated ageing-associated mutational processes. Non-dysplastic mucosa had considerable mutation burden (median 47 SNAs), including mutations shared with the neighbouring CA-CRC, indicating a precancer mutational field. CA-CRCs were often near triploid (40%) or near tetraploid (20%) and phylogenetic analysis revealed that copy number alterations (CNAs) began to accrue in non-dysplastic bowel, but the LGD/HGD transition often involved a punctuated 'catastrophic' CNA increase. CONCLUSIONS Evolutionary genomic analysis revealed precancer clones bearing extensive SNAs and CNAs, with progression to cancer involving a dramatic accrual of CNAs at HGD. Detection of the cancerised field is an encouraging prospect for surveillance, but punctuated evolution may limit the window for early detection.
Collapse
Affiliation(s)
- Ann-Marie Baker
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - William Cross
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Kit Curtius
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Ibrahim Al Bakir
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Inflammatory Bowel Disease Unit, St Mark’s Hospital, London, UK
| | - Chang-Ho Ryan Choi
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Inflammatory Bowel Disease Unit, St Mark’s Hospital, London, UK
| | | | - Daniel Temko
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Department of Computer Science, University College London, London, UK
- Centre for Mathematics and Physics in the Life Sciences and Experimental Biology (CoMPLEX), University College London, London, UK
| | - Sujata Biswas
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Pierre Martinez
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Marc J Williams
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Centre for Mathematics and Physics in the Life Sciences and Experimental Biology (CoMPLEX), University College London, London, UK
- Department of Cell and Developmental Biology, University College London, London, UK
| | - James O Lindsay
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Roger Feakins
- Department of Histopathology, The Royal London Hospital, London, UK
| | - Roser Vega
- Department of Gastroenterology, University College London Hospital, London, UK
| | - Stephen J Hayes
- Department of Histopathology, Salford Royal NHS Foundation Trust, University of Manchester, Manchester, UK
| | - Ian P M Tomlinson
- Cancer Genetics and Evolution Laboratory, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Stuart A C McDonald
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Morgan Moorghen
- Inflammatory Bowel Disease Unit, St Mark’s Hospital, London, UK
| | - Andrew Silver
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - James E East
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Nicholas A Wright
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Lai Mun Wang
- Cellular Pathology, John Radcliffe Hospital, Oxford, UK
| | | | - Marnix Jansen
- Department of Histopathology, University College London Hospital, London, UK
| | - Ailsa L Hart
- Inflammatory Bowel Disease Unit, St Mark’s Hospital, London, UK
| | - Simon J Leedham
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Trevor A Graham
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
224
|
Parker JDK, Yap SQ, Starks E, Slind J, Swanson L, Docking TR, Fuller M, Zhou C, Walker B, Filipenko D, Xiong W, Karimuddin AA, Phang PT, Raval M, Brown CJ, Karsan A. Fixation Effects on Variant Calling in a Clinical Resequencing Panel. J Mol Diagn 2019; 21:705-717. [PMID: 31055024 DOI: 10.1016/j.jmoldx.2019.03.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 03/15/2019] [Accepted: 03/20/2019] [Indexed: 11/16/2022] Open
Abstract
Formalin fixation is the standard method for the preservation of tissue for diagnostic purposes, including pathologic review and molecular assays. However, this method is known to cause artifacts that can affect the accuracy of molecular genetic test results. We assessed the applicability of alternative fixatives to determine whether these perform significantly better on next-generation sequencing assays, and whether adequate morphology is retained for primary diagnosis, in a prospective study using a clinical-grade, laboratory-developed targeted resequencing assay. Several parameters relating to sequencing quality and variant calling were examined and quantified in tumor and normal colon epithelial tissues. We identified an alternative fixative that suppresses many formalin-related artifacts while retaining adequate morphology for pathologic review.
Collapse
Affiliation(s)
- Jeremy D K Parker
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Shyong Quin Yap
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Elizabeth Starks
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Jillian Slind
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Lucas Swanson
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - T Roderick Docking
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Megan Fuller
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Chen Zhou
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Blair Walker
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Douglas Filipenko
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Wei Xiong
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Ahmer A Karimuddin
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada; St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - P Terry Phang
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada; St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Manoj Raval
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada; St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Carl J Brown
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada; St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Aly Karsan
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
225
|
Zakrzewski F, Gieldon L, Rump A, Seifert M, Grützmann K, Krüger A, Loos S, Zeugner S, Hackmann K, Porrmann J, Wagner J, Kast K, Wimberger P, Baretton G, Schröck E, Aust D, Klink B. Targeted capture-based NGS is superior to multiplex PCR-based NGS for hereditary BRCA1 and BRCA2 gene analysis in FFPE tumor samples. BMC Cancer 2019; 19:396. [PMID: 31029168 PMCID: PMC6487025 DOI: 10.1186/s12885-019-5584-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 04/05/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND With the introduction of Olaparib treatment for BRCA-deficient recurrent ovarian cancer, testing for somatic and/or germline mutations in BRCA1/2 genes in tumor tissues became essential for treatment decisions. In most cases only formalin-fixed paraffin-embedded (FFPE) samples, containing fragmented and chemically modified DNA of minor quality, are available. Thus, multiplex PCR-based sequencing is most commonly applied in routine molecular testing, which is predominantly focused on the identification of known hot spot mutations in oncogenes. METHODS We compared the overall performance of an adjusted targeted capture-based enrichment protocol and a multiplex PCR-based approach for calling of pathogenic SNVs and InDels using DNA extracted from 13 FFPE tissue samples. We further applied both strategies to seven blood samples and five matched FFPE tumor tissues of patients with known germline exon-spanning deletions and gene-wide duplications in BRCA1/2 to evaluate CNV detection based solely on panel NGS data. Finally, we analyzed DNA from FFPE tissues of 11 index patients from families suspected of having hereditary breast and ovarian cancer, of whom no blood samples were available for testing, in order to identify underlying pathogenic germline BRCA1/2 mutations. RESULTS The multiplex PCR-based protocol produced inhomogeneous coverage among targets of each sample and between samples as well as sporadic amplicon drop out, leading to insufficiently or non-covered nucleotides, which subsequently hindered variant detection. This protocol further led to detection of PCR-artifacts that could easily have been misinterpreted as pathogenic mutations. No such limitations were observed by application of an adjusted targeted capture-based protocol, which allowed for CNV calling with 86% sensitivity and 100% specificity. All pathogenic CNVs were confirmed in the five matched FFPE tumor samples from patients carrying known pathogenic germline mutations and we additionally identified somatic loss of the second allele in BRCA1/2. Furthermore we detected pathogenic BRCA1/2 variants in four the eleven FFPE samples from patients of whom no blood was available for analysis. CONCLUSIONS We demonstrate that an adjusted targeted capture-based enrichment protocol is superior to commonly applied multiplex PCR-based protocols for reliable BRCA1/2 variant detection, including CNV-detection, using FFPE tumor samples.
Collapse
Affiliation(s)
- Falk Zakrzewski
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT), Schubertstraße 15, 01307 Dresden, Germany
| | - Laura Gieldon
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT), Schubertstraße 15, 01307 Dresden, Germany
- Institute for Clinical Genetics, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Andreas Rump
- Institute for Clinical Genetics, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Michael Seifert
- Institute for Medical Informatics and Biometry (IMB), Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
| | - Konrad Grützmann
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT), Schubertstraße 15, 01307 Dresden, Germany
| | - Alexander Krüger
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT), Schubertstraße 15, 01307 Dresden, Germany
| | - Sina Loos
- Institute of Pathology, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Silke Zeugner
- Institute of Pathology, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Karl Hackmann
- Institute for Clinical Genetics, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Joseph Porrmann
- Institute for Clinical Genetics, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Johannes Wagner
- Institute for Clinical Genetics, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Karin Kast
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Consortium (DKTK), Dresden, Germany
- Department of Gynecology and Obstetrics, University Hospital Carl Gustav Carus Dresden, TU Dresden, Dresden, Germany
| | - Pauline Wimberger
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Consortium (DKTK), Dresden, Germany
- Department of Gynecology and Obstetrics, University Hospital Carl Gustav Carus Dresden, TU Dresden, Dresden, Germany
| | - Gustavo Baretton
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT), Schubertstraße 15, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- Institute of Pathology, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Dresden, Germany
- Tumor- and Normal Tissue Bank of the University Cancer Center (UCC), University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, National Center for Tumor Diseases (NCT) Dresden, Dresden, Germany
| | - Evelin Schröck
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT), Schubertstraße 15, 01307 Dresden, Germany
- Institute for Clinical Genetics, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Consortium (DKTK), Dresden, Germany
| | - Daniela Aust
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT), Schubertstraße 15, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- Institute of Pathology, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Dresden, Germany
- Tumor- and Normal Tissue Bank of the University Cancer Center (UCC), University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, National Center for Tumor Diseases (NCT) Dresden, Dresden, Germany
| | - Barbara Klink
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT), Schubertstraße 15, 01307 Dresden, Germany
- Institute for Clinical Genetics, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Consortium (DKTK), Dresden, Germany
| |
Collapse
|
226
|
Lewis ER, Geisbüsch S, Chang YJ, Costa V, Husain S, Soteropoulos P, Griepp RB, Di Luozzo G. Paraspinous muscle gene expression profiling following simulated staged endovascular repair of thoracoabdominal aortic aneurysm: exploring potential therapeutic pathways. Eur J Cardiothorac Surg 2019; 57:30-38. [DOI: 10.1093/ejcts/ezz113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 03/11/2019] [Accepted: 03/13/2019] [Indexed: 02/01/2023] Open
Abstract
Abstract
OBJECTIVES
Thoracic endovascular techniques for aneurysm repair offer less invasive alternatives to open strategies. Both approaches, however, are associated with the risk for neurological complications. Despite adjuncts to maintain spinal cord perfusion, ischaemia and paraplegia continue to occur during thoracoabdominal aortic aneurysm (TAAA) repair. Staging of such extensive procedures has been proven to decrease the risk for spinal cord injury. Archived biopsy specimens may offer insight into the molecular signature of the reorganization and expansion of the spinal collateral network during staged endovascular interventions in the setting of TAAA.
METHODS
Biological replicates of total RNA were isolated from existing paraspinous muscle samples from 22 Yorkshire pigs randomized to 1 of 3 simulated TAAA repair strategies as part of a previous study employing coil embolization of spinal segmental arteries within the thoracic and lumbar spine. Gene expression profiling was performed using the Affymetrix GeneChip Porcine array.
RESULTS
Microarray analysis identified 649 differentially expressed porcine genes (≥1.3-fold change, P ≤ 0.05) when comparing paralysed and non-paralysed subjects. Of these, 355 were available for further analysis. When mapped to the human genome, 169 Homo sapiens orthologues were identified. Integrated interpretation of gene expression profiles indicated the significant regulation of transcriptional regulators (such as nuclear factor кB), cytokine (including CXCL12) elements contributing to hypoxia signalling in the cardiovascular system (vascular endothelial growth factor and UBE2) and cytoskeletal elements (like dystrophin (DMD) and matrix metallopeptidase (MMP)).
CONCLUSIONS
This study demonstrates the ability of microarray-based platforms to detect the differential expression of genes in paraspinous muscle during staged TAAA repair. Pathway enrichment analysis detected subcellular actors accompanying the neuroprotective effects of staged endovascular coiling. These observations provide new insight into the potential prognostic and therapeutic value of gene expression profiling in monitoring and modulating the arteriolar remodelling in the collateral network.
Collapse
Affiliation(s)
- Erin R Lewis
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Sarah Geisbüsch
- Department of Cardiovascular Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yun-Juan Chang
- Office of Advanced Research Computing, Rutgers University, Newark, NJ, USA
| | - Victor Costa
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Seema Husain
- Genomics Center, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | | | - Randall B Griepp
- Department of Cardiovascular Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gabriele Di Luozzo
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ, USA
- Department of Cardiovascular Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
227
|
McDonough SJ, Bhagwate A, Sun Z, Wang C, Zschunke M, Gorman JA, Kopp KJ, Cunningham JM. Use of FFPE-derived DNA in next generation sequencing: DNA extraction methods. PLoS One 2019; 14:e0211400. [PMID: 30973937 PMCID: PMC6459541 DOI: 10.1371/journal.pone.0211400] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/20/2019] [Indexed: 12/25/2022] Open
Abstract
Archival tissues represent a rich resource for clinical genomic studies, particularly when coupled with comprehensive medical records. Use of these in next generation sequencing (NGS) is a priority. Nine formalin-fixed paraffin-embedded (FFPE) DNA extraction methods were evaluated using twelve FFPE samples of varying tissue types. Quality assessment included total yield, percent dsDNA, fragment analysis and multiplex PCR. After assessment, three tissue types from four FFPE DNA methods were selected for NGS downstream evaluation, targeted and whole exome sequencing. In addition, two low input library protocols were evaluated for WES. Analysis revealed average coverage across the target regions for WES was ~20-30X for all four FFPE DNA extraction methods. For the targeted panels, the highest molecular tag coverage was obtained with the Kingfisher FFPE extraction method. The genotype concordance was 99% for the commonly called variant positions between all four extraction methods with the targeted PCR NGS panel and 96% with WES. Assessing quality of extracted DNA aids in selecting the optimal NGS approach, and the choice of both DNA extraction and library preparation approaches can impact the performance of archival tissue in NGS.
Collapse
Affiliation(s)
- Samantha J. McDonough
- Medical Genome Facility, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Aditya Bhagwate
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Zhifu Sun
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Chen Wang
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Michael Zschunke
- Pathology Research Core, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Joshua A. Gorman
- Pathology Research Core, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Karla J. Kopp
- Pathology Research Core, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Julie M. Cunningham
- Medical Genome Facility, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
228
|
DNA extraction from FFPE tissue samples - a comparison of three procedures. Contemp Oncol (Pozn) 2019; 23:52-58. [PMID: 31061638 PMCID: PMC6500389 DOI: 10.5114/wo.2019.83875] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/26/2019] [Indexed: 12/29/2022] Open
Abstract
Aim of the study One of the critical steps in molecular oncology diagnostics is obtaining high quality genomic DNA. Therefore, it is important to evaluate and compare the techniques used to extract DNA from tissue samples. Since formalin-fixed, paraffin-embedded (FFPE) tissues are routinely used for both retrospective and prospective studies, we compared three commercially available methods of nucleic acid extraction in terms of quantity and quality of isolated DNA. Material and methods Slides prepared from 42 FFPE blocks were macro-dissected. Resulting material was divided and processed simultaneously using three extraction kits: QIAamp DNA FFPE Tissue Kit (QIAGEN), Cobas DNA Sample Preparation Kit (Roche Molecular Systems) and Maxwell 16 FFPE Plus LEV DNA Purification Kit (Promega). Subsequently, quantity and quality of obtained DNA samples were analysed spectrophotometrically (NanoDrop 2000, Thermo Scientific). Results of quantitative analysis were confirmed by a fluorometric procedure (Qubit 3.0 Fluorometer, Life Technologies). Results The results demonstrated that the yields of total DNA extracted using either Maxwell or Cobas methods were significantly higher compared to the QIAamp method (p < 0.001). The Maxwell Extraction Kit delivered DNA samples of the highest quality (p < 0.01). However, the highest total yield of extracted DNA was achieved with the Cobas technique, which may be due to a higher volume of eluate compared to the Maxwell method. Conclusions To our knowledge, this is the first paper which directly compares three extraction methods: Cobas, Maxwell and QIAamp. The data herein provide information required for the selection of a protocol that best suits the needs of the overall study design in terms of the quantity and quality of the extracted DNA.
Collapse
|
229
|
Surrey LF, Oakley FD, Merker JD, Long TA, Vasalos P, Moncur JT, Kim AS. Next-Generation Sequencing (NGS) Methods Show Superior or Equivalent Performance to Non-NGS Methods on BRAF, EGFR, and KRAS Proficiency Testing Samples. Arch Pathol Lab Med 2019; 143:980-984. [DOI: 10.5858/arpa.2018-0394-cp] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Context.—
There has been a rapid expansion of next-generation sequencing (NGS)–based assays for the detection of somatic variants in solid tumors. However, limited data are available regarding the comparative performance of NGS and non-NGS assays using standardized samples across a large number of laboratories.
Objective.—
To compare the performance of NGS and non-NGS assays using well-characterized proficiency testing samples provided by the College of American Pathologists (CAP) Molecular Oncology Committee. A secondary goal was to compare the use of preanalytic and postanalytic practices.
Design.—
A total of 17 343 responses were obtained from participants in the BRAF, EGFR, KRAS, and the Multigene Tumor Panel surveys across 84 different proficiency testing samples interrogating 16 variants and 3 wild-type sequences. Performance and preanalytic/postanalytic practices were analyzed by method.
Results.—
While both NGS and non-NGS achieved an acceptable response rate of greater than 95%, the overall performance of NGS methods was significantly better than that of non-NGS methods for the identification of variants in BRAF (overall 97.8% versus 95.6% acceptable responses, P = .001) and EGFR (overall 98.5% versus 97.3%, P = .01) and was similar for KRAS (overall 98.8% and 97.6%, P = .10). There were specific variant differences, but in all discrepant cases, NGS methods outperformed non-NGS methods. NGS laboratories also more consistently used preanalytic and postanalytic practices suggested by the CAP checklist requirements than non-NGS laboratories.
Conclusions.—
The overall analytic performance of both methods was excellent. For specific BRAF and EGFR variants, NGS outperformed non-NGS methods and NGS laboratories report superior adherence to suggested laboratory practices.
Collapse
Affiliation(s)
- Lea F. Surrey
- From the Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Dr Surrey) Peoria Tazewell Pathology Group, Peoria, Illinois (Dr Oakley); the Department of Pathology, University of North Carolina, Chapel Hill (Dr Merker); Biostatistics (Mr Long) and Proficiency Testing (Ms Vasalos), College of Ame
| | - Fredrick D. Oakley
- From the Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Dr Surrey) Peoria Tazewell Pathology Group, Peoria, Illinois (Dr Oakley); the Department of Pathology, University of North Carolina, Chapel Hill (Dr Merker); Biostatistics (Mr Long) and Proficiency Testing (Ms Vasalos), College of Ame
| | - Jason D. Merker
- From the Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Dr Surrey) Peoria Tazewell Pathology Group, Peoria, Illinois (Dr Oakley); the Department of Pathology, University of North Carolina, Chapel Hill (Dr Merker); Biostatistics (Mr Long) and Proficiency Testing (Ms Vasalos), College of Ame
| | - Thomas A. Long
- From the Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Dr Surrey) Peoria Tazewell Pathology Group, Peoria, Illinois (Dr Oakley); the Department of Pathology, University of North Carolina, Chapel Hill (Dr Merker); Biostatistics (Mr Long) and Proficiency Testing (Ms Vasalos), College of Ame
| | - Patricia Vasalos
- From the Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Dr Surrey) Peoria Tazewell Pathology Group, Peoria, Illinois (Dr Oakley); the Department of Pathology, University of North Carolina, Chapel Hill (Dr Merker); Biostatistics (Mr Long) and Proficiency Testing (Ms Vasalos), College of Ame
| | - Joel T. Moncur
- From the Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Dr Surrey) Peoria Tazewell Pathology Group, Peoria, Illinois (Dr Oakley); the Department of Pathology, University of North Carolina, Chapel Hill (Dr Merker); Biostatistics (Mr Long) and Proficiency Testing (Ms Vasalos), College of Ame
| | - Annette S. Kim
- From the Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia (Dr Surrey) Peoria Tazewell Pathology Group, Peoria, Illinois (Dr Oakley); the Department of Pathology, University of North Carolina, Chapel Hill (Dr Merker); Biostatistics (Mr Long) and Proficiency Testing (Ms Vasalos), College of Ame
| |
Collapse
|
230
|
Kudryavtseva AV, Lukyanova EN, Kalinin DV, Zaretsky AR, Pokrovsky AV, Golovyuk AL, Fedorova MS, Pudova EA, Kharitonov SL, Pavlov VS, Kobelyatskaya AA, Melnikova NV, Dmitriev AA, Polyakov AP, Alekseev BY, Kiseleva MV, Kaprin AD, Krasnov GS, Snezhkina AV. Mutational load in carotid body tumor. BMC Med Genomics 2019; 12:39. [PMID: 30871634 PMCID: PMC6416835 DOI: 10.1186/s12920-019-0483-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Carotid body tumor (CBT) is a rare neoplasm arising from paraganglion located near the bifurcation of the carotid artery. There is great intra-tumor heterogeneity, and CBT development could be associated with both germline and somatic allelic variants. Studies on the molecular genetics of CBT are limited, and the molecular mechanisms of its pathogenesis are not fully understood. This work is focused on the estimation of mutational load (ML) in CBT. Methods Using the NextSeq 500 platform, we performed exome sequencing of tumors with matched lymph node tissues and peripheral blood obtained from six patients with CBT. To obtain reliable results in tumors with low ML, we developed and successfully applied a complex approach for the analysis of sequencing data. ML was evaluated as the number of somatic variants per megabase (Mb) of the target regions covered by the Illumina TruSeq Exome Library Prep Kit. Results The ML in CBT varied in the range of 0.09–0.28/Mb. Additionally, we identified several pathogenic/likely pathogenic somatic and germline allelic variants across six patients studied (including TP53 variants). Conclusions Using the developed approach, we estimated the ML in CBT, which is much lower than in common malignant tumors. Identified variants in known paraganglioma/pheochromocytoma-causative genes and novel genes could be associated with the pathogenesis of CBT. The obtained results expand our knowledge of the mutation process in CBT as well as the biology of tumor development.
Collapse
Affiliation(s)
- Anna V Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| | - Elena N Lukyanova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Dmitry V Kalinin
- Vishnevsky Institute of Surgery, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Andrew R Zaretsky
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Anatoly V Pokrovsky
- Vishnevsky Institute of Surgery, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Alexander L Golovyuk
- Vishnevsky Institute of Surgery, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Maria S Fedorova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Elena A Pudova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Sergey L Kharitonov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Vladislav S Pavlov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | | | - Nataliya V Melnikova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Alexey A Dmitriev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Andrey P Polyakov
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Boris Y Alekseev
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Marina V Kiseleva
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Andrey D Kaprin
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - George S Krasnov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | | |
Collapse
|
231
|
Kim J, Kim D, Lim JS, Maeng JH, Son H, Kang HC, Nam H, Lee JH, Kim S. The use of technical replication for detection of low-level somatic mutations in next-generation sequencing. Nat Commun 2019; 10:1047. [PMID: 30837471 PMCID: PMC6400950 DOI: 10.1038/s41467-019-09026-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 02/07/2019] [Indexed: 01/16/2023] Open
Abstract
Accurate genome-wide detection of somatic mutations with low variant allele frequency (VAF, <1%) has proven difficult, for which generalized, scalable methods are lacking. Herein, we describe a new computational method, called RePlow, that we developed to detect low-VAF somatic mutations based on simple, library-level replicates for next-generation sequencing on any platform. Through joint analysis of replicates, RePlow is able to remove prevailing background errors in next-generation sequencing analysis, facilitating remarkable improvement in the detection accuracy for low-VAF somatic mutations (up to ~99% reduction in false positives). The method is validated in independent cancer panel and brain tissue sequencing data. Our study suggests a new paradigm with which to exploit an overwhelming abundance of sequencing data for accurate variant detection. Somatic mutations of low allele frequencies are often difficult to detect. Here, the authors develop RePlow, a computational method that leverages technical replication for detecting low-level somatic mutations using next-generation sequencing.
Collapse
Affiliation(s)
- Junho Kim
- Department of Biomedical Systems Informatics and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Dachan Kim
- Department of Biomedical Systems Informatics and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Jae Seok Lim
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, South Korea
| | - Ju Heon Maeng
- Department of Biomedical Systems Informatics and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Hyeonju Son
- Department of Biomedical Systems Informatics and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Hoon-Chul Kang
- Department of Pediatrics, Division of Pediatric Neurology, Pediatric Epilepsy Clinics, Severance Children's Hospital, Epilepsy Research Institute, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Hojung Nam
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea
| | - Jeong Ho Lee
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, South Korea.
| | - Sangwoo Kim
- Department of Biomedical Systems Informatics and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| |
Collapse
|
232
|
Costa-Guda J. Commentary: Next-Generation Sequence Analysis of Parathyroid Carcinoma. J Endocr Soc 2019; 3:560-562. [PMID: 30793092 PMCID: PMC6378759 DOI: 10.1210/js.2018-00315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 10/16/2018] [Indexed: 02/04/2023] Open
Affiliation(s)
- Jessica Costa-Guda
- Center for Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, University of Connecticut School of Dental Medicine, Farmington, Connecticut
- Center for Molecular Oncology, Carole and Ray Neag Comprehensive Cancer Center, University of Connecticut School of Medicine, Farmington, Connecticut
| |
Collapse
|
233
|
Ingerslev K, Høgdall E, Skovrider-Ruminski W, Schnack TH, Lidang M, Høgdall C, Blaakaer J. The prevalence of EBV and CMV DNA in epithelial ovarian cancer. Infect Agent Cancer 2019; 14:7. [PMID: 30858874 PMCID: PMC6390605 DOI: 10.1186/s13027-019-0223-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 02/12/2019] [Indexed: 11/10/2022] Open
Abstract
Background The underlying cause of epithelial ovarian cancer (EOC) is unknown. It has been theorized that infectious agents could contribute to ovarian tumorigenesis. Objective To investigate the potential role of oncogenic viral infection in EOC, we examined the prevalence of Epstein-Barr Virus (EBV) DNA and cytomegalovirus (CMV) DNA in EOC tissue samples. Methods Formalin-fixed, paraffin-imbedded (FFPE) tumor tissue samples from 198 patients included in the Danish Pelvic Mass Study were studied: 163 with serous adenocarcinomas, 15 with endometrioid adenocarcinomas, 11 with mucinous adenocarcinomas, and nine with clear-cell carcinomas. For controls in the EBV analysis, we used 176 tissue samples from patients diagnosed with benign mucinous cystadenomas. EBV and CMV genotyping was performed by real-time polymerase chain reaction with CMV and EBV CE-IVD approved kits. In-situ hybridization (ISH) was performed on the EBV positive samples. Results Sufficient DNA material was obtained in 191 and 174 tissue samples from cases and controls, respectively. Ten of 191 case samples (5.2%) and one of 174 control samples (0.5%) were positive for EBV DNA (P value = 0.011). CMV DNA was detected in only one case sample (0.5%). ISH confirmed that three of the samples were of stromal origin, while the remaining seven tested negative for EBV. Conclusions This study is the first to demonstrate a higher prevalence of EBV DNA in tissue samples from patients with EOC than in a benign control group. However, the cellular origin of seven of the samples could not be determined by ISH analysis. Our study did not support an association between CMV and EOC.
Collapse
Affiliation(s)
- Kasper Ingerslev
- 1Department of Gynecology, Odense University Hospital, Sdr. Boulevard 29, 5000 Odense C, Denmark
| | - Estrid Høgdall
- 2Department of Pathology, Herlev Hospital, Herlev Ringvej 75, 2730 Herlev, Denmark
| | | | - Tine Henrichsen Schnack
- 3Department of Gynecology, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark
| | - Marianne Lidang
- 2Department of Pathology, Herlev Hospital, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - Claus Høgdall
- 3Department of Gynecology, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark
| | - Jan Blaakaer
- 1Department of Gynecology, Odense University Hospital, Sdr. Boulevard 29, 5000 Odense C, Denmark
| |
Collapse
|
234
|
da Cunha Santos G, Saieg MA, Troncone G, Zeppa P. Cytological preparations for molecular analysis: A review of technical procedures, advantages and limitations for referring samples for testing. Cytopathology 2019; 29:125-132. [PMID: 29575423 DOI: 10.1111/cyt.12534] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2018] [Indexed: 12/12/2022]
Abstract
Minimally invasive procedures such as endobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA) must yield not only good quality and quantity of material for morphological assessment, but also an adequate sample for analysis of molecular markers to guide patients to appropriate targeted therapies. In this context, cytopathologists worldwide should be familiar with minimum requirements for refereeing cytological samples for testing. The present manuscript is a review with comprehensive description of the content of the workshop entitled Cytological preparations for molecular analysis: pre-analytical issues for EBUS TBNA, presented at the 40th European Congress of Cytopathology in Liverpool, UK. The present review emphasises the advantages and limitations of different types of cytology substrates used for molecular analysis such as archival smears, liquid-based preparations, archival cytospin preparations and FTA (Flinders Technology Associates) cards, as well as their technical requirements/features. These various types of cytological specimens can be successfully used for an extensive array of molecular studies, but the quality and quantity of extracted nucleic acids rely directly on adequate pre-analytical assessment of those samples. In this setting, cytopathologists must not only be familiar with the different types of specimens and associated technical procedures, but also correctly handle the material provided by minimally invasive procedures, ensuring that there is sufficient amount of material for a precise diagnosis and correct management of the patient through personalised care.
Collapse
Affiliation(s)
- G da Cunha Santos
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Laboratory Medicine Program, University Health Network, Toronto, ON, Canada
| | - M A Saieg
- Department of Pathology, Santa Casa Medical School, São Paulo, Brazil.,Department of Pathology, AC Camargo Cancer Center, São Paulo, Brazil
| | - G Troncone
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - P Zeppa
- Department of Medicine and Surgery, University of Salerno, Salerno, Italy
| |
Collapse
|
235
|
Nagahashi M, Shimada Y, Ichikawa H, Kameyama H, Takabe K, Okuda S, Wakai T. Next generation sequencing-based gene panel tests for the management of solid tumors. Cancer Sci 2019; 110:6-15. [PMID: 30338623 PMCID: PMC6317963 DOI: 10.1111/cas.13837] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 10/13/2018] [Accepted: 10/16/2018] [Indexed: 12/16/2022] Open
Abstract
Next generation sequencing (NGS) has been an invaluable tool to put genomic sequencing into clinical practice. The incorporation of clinically relevant target sequences into NGS-based gene panel tests has generated practical diagnostic tools that enable individualized cancer-patient care. The clinical utility of gene panel testing includes investigation of the genetic basis for an individual's response to therapy, such as signaling pathways associated with a response to specific therapies, microsatellite instability and a hypermutated phenotype, and deficiency in the DNA double-strand break repair pathway. In this review, we describe the concept of precision cancer medicine using target sequences in gene panel tests as well as the importance of the control of sample quality in routine NGS-based genomic testing. We describe geographic and ethnic differences in cancer genomes, and discuss issues that need to be addressed in the future based on our experiences in Japan.
Collapse
Affiliation(s)
- Masayuki Nagahashi
- Division of Digestive and General SurgeryGraduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Yoshifumi Shimada
- Division of Digestive and General SurgeryGraduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Hiroshi Ichikawa
- Division of Digestive and General SurgeryGraduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Hitoshi Kameyama
- Division of Digestive and General SurgeryGraduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Kazuaki Takabe
- Division of Digestive and General SurgeryGraduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
- Breast SurgeryRoswell Park Cancer InstituteBuffaloNew York
- Department of SurgeryThe State University of New York Jacobs School of Medicine and Biomedical SciencesUniversity at BuffaloBuffaloNew York
| | - Shujiro Okuda
- Division of BioinformaticsGraduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Toshifumi Wakai
- Division of Digestive and General SurgeryGraduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| |
Collapse
|
236
|
Berland L, Heeke S, Humbert O, Macocco A, Long-Mira E, Lassalle S, Lespinet-Fabre V, Lalvée S, Bordone O, Cohen C, Leroy S, Hofman V, Hofman P, Ilié M. Current views on tumor mutational burden in patients with non-small cell lung cancer treated by immune checkpoint inhibitors. J Thorac Dis 2019; 11:S71-S80. [PMID: 30775030 DOI: 10.21037/jtd.2018.11.102] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In the last few years, the treatment of patients with non-small cell lung cancer (NSCLC) has impressively benefitted from immunotherapy, in particular from the inhibition of immune checkpoints such as programmed cell death-1 (PD-1) and programmed cell death ligand-1 (PD-L1). However, despite the significant survival benefit for some patients with advanced NSCLC, the objective response rates (ORRs) remain relatively low no more than 20-30% with a large proportion of patients demonstrating primary resistance. Although the selection of NSCLC patients for the first-line treatment is currently guided by the expression of PD-L1 in tumor cells as detected by immunohistochemistry, this is not the case for the second-line setting. Moreover, the sensitivity and specificity of PD-L1 expression is modest which has prompted the search for additional predictive biomarkers. In this context, the assessment of the tumor mutational burden (TMB), defined as the total number of nonsynonymous mutations in the coding regions of genes, has recently emerged as an additional powerful biomarker to select patients for immunotherapy. The purpose of our review is to highlight the recent advances as well as the challenges and perspectives in the field of TMB and immunotherapy for patients with NSCLC.
Collapse
Affiliation(s)
- Léa Berland
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Hospital Federation OncoAge, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France.,UFR Médicine, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France
| | - Simon Heeke
- Institute of Research on Cancer and Ageing of Nice (IRCAN), University Hospital Federation OncoAge, CNRS, INSERM, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France
| | - Olivier Humbert
- Department of Nuclear Medicine, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France
| | - Adam Macocco
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Hospital Federation OncoAge, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France.,UFR Médicine, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France
| | - Elodie Long-Mira
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Hospital Federation OncoAge, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France.,UFR Médicine, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France
| | - Sandra Lassalle
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Hospital Federation OncoAge, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France.,UFR Médicine, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France
| | - Virginie Lespinet-Fabre
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Hospital Federation OncoAge, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France
| | - Salomé Lalvée
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Hospital Federation OncoAge, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France
| | - Olivier Bordone
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Hospital Federation OncoAge, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France
| | - Charlotte Cohen
- Department of Thoracic Surgery, Pasteur Hospital, University Hospital Federation OncoAge, Université Côte d'Azur, Nice, France
| | - Sylvie Leroy
- Department of Pulmonary Medicine and Oncology, Pasteur Hospital, University Hospital Federation OncoAge, Université Côte d'Azur, Nice, France
| | - Véronique Hofman
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Hospital Federation OncoAge, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France.,UFR Médicine, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France.,Department of Thoracic Surgery, Pasteur Hospital, University Hospital Federation OncoAge, Université Côte d'Azur, Nice, France
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Hospital Federation OncoAge, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France.,UFR Médicine, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France.,Hospital-Related Biobank (BB-0033-00025), Pasteur Hospital, University Hospital Federation OncoAge, Université Côte d'Azur, Nice, France
| | - Marius Ilié
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Hospital Federation OncoAge, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France.,UFR Médicine, Antoine Lacassagne Comprehensive Cancer Center, Université Côte d'Azur, Nice, France.,Hospital-Related Biobank (BB-0033-00025), Pasteur Hospital, University Hospital Federation OncoAge, Université Côte d'Azur, Nice, France
| |
Collapse
|
237
|
Sohn J. Next generation sequencing and anti-cancer therapy. JOURNAL OF THE KOREAN MEDICAL ASSOCIATION 2019. [DOI: 10.5124/jkma.2019.62.2.119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Joohyuk Sohn
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
238
|
Kosovec JE, Zaidi AH, Pounardjian TS, Jobe BA. The Potential Clinical Utility of Circulating Tumor DNA in Esophageal Adenocarcinoma: From Early Detection to Therapy. Front Oncol 2018; 8:610. [PMID: 30619750 PMCID: PMC6297385 DOI: 10.3389/fonc.2018.00610] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/28/2018] [Indexed: 12/11/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) is a lethal cancer requiring improved screening strategies and treatment options due to poor detection methods, aggressive progression, and therapeutic resistance. Emerging circulating tumor DNA (ctDNA) technologies may offer a unique non-invasive strategy to better characterize the highly heterogeneous cancer and more clearly establish the genetic modulations leading to disease progression. The presented review describes the potential advantages of ctDNA methodologies as compared to current clinical strategies to improve clinical detection, enhance disease surveillance, evaluate prognosis, and personalize treatment. Specifically, we describe the ctDNA-targetable genetic markers of prognostic significance to stratify patients into risk of progression from benign to malignant disease and potentially offer cost-effective screening of established cancer. We also describe the application of ctDNA to more effectively characterize the heterogeneity and particular mutagenic resistance mechanisms in real-time to improve prognosis and therapeutic monitoring strategies. Lastly, we discuss the inconsistent clinical responses to currently approved therapies for EAC and the role of ctDNA to explore the dynamic regulation of novel targeted and immunotherapies to personalize therapy and improve patient outcomes. Although there are clear limitations of ctDNA technologies for immediate clinical deployment, this review presents the prospective role of such applications to potentially overcome many of the notable hurdles to treating EAC patients. A deeper understanding of complex EAC tumor biology may result in the progress toward improved clinical outcomes.
Collapse
Affiliation(s)
- Juliann E Kosovec
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, United States
| | - Ali H Zaidi
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, United States
| | - Tamar S Pounardjian
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, United States
| | - Blair A Jobe
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, United States
| |
Collapse
|
239
|
Fernando MR, Jiang C, Krzyzanowski GD, Somer-Shely T, Ryan WL. A novel approach to stabilize fetal cell-free DNA fraction in maternal blood samples for extended period of time. PLoS One 2018; 13:e0208508. [PMID: 30521613 PMCID: PMC6283530 DOI: 10.1371/journal.pone.0208508] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 11/19/2018] [Indexed: 02/04/2023] Open
Abstract
This study was undertaken to evaluate a novel method for stabilizing and preserving the original proportion of cell-free fetal DNA (cffDNA) in maternal blood for extended periods of time without using crosslinking agents, such as formaldehyde, which compromise DNA integrity and extraction efficiency. Blood was drawn from pregnant donors into K3EDTA and Blood Exo DNA ProTeck® (ProTeck) tubes. Blood drawn into both tubes were aliquoted and stored at three different temperatures. At indicated times sample aliquots were processed for cell-free DNA (cfDNA) extraction. Plasma cfDNA and cffDNA quantified by droplet digital PCR (ddPCR) assay which amplify RASSF1A gene promoter region. ProTeck reagent is formaldehyde free and inhibits blood cell metabolism in blood samples during storage. Cell-free DNA concentration increased over time in blood plasma stored in K3EDTA tubes at 4, 22 and 30°C. Blood stored in ProTeck tubes, cfDNA concentration was stable at 4, 22 and 30°C for 21, 28 and 7 days, respectively. In K3EDTA tubes cffDNA proportion decreases steadily over time whereas in ProTeck tubes cffDNA proportion remained stable. This novel technology stabilizes cffDNA proportion in maternal blood samples at 4, 22 and 30°C for 21, 28 and 7 days, respectively.
Collapse
Affiliation(s)
- M. Rohan Fernando
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha NE, United States of America
- Department of Research and Development, CFGenome Omaha NE, United States of America
- * E-mail:
| | - Chao Jiang
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha NE, United States of America
| | - Gary D. Krzyzanowski
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha NE, United States of America
- Department of Research and Development, CFGenome Omaha NE, United States of America
| | - Tifany Somer-Shely
- Methodist Physicians Clinic Women’s Center, Elkhorn, NE, United States of America
| | - Wayne L. Ryan
- Department of Research and Development, CFGenome Omaha NE, United States of America
| |
Collapse
|
240
|
Martínez-Pérez C, Turnbull AK, Dixon JM. The evolving role of receptors as predictive biomarkers for metastatic breast cancer. Expert Rev Anticancer Ther 2018; 19:121-138. [PMID: 30501540 DOI: 10.1080/14737140.2019.1552138] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION In breast cancer, estrogen receptor (ER) and human epidermal growth factor receptor 2 (HER2) are essential biomarkers to predict response to endocrine and anti-HER2 therapies, respectively. In metastatic breast cancer, the use of these receptors and targeted therapies present additional challenges: temporal heterogeneity, together with limited sampling methodologies, hinders receptor status assessment, and the constant evolution of the disease invariably leads to resistance to treatment. Areas covered: This review summarizes the genomic abnormalities in ER and HER2, such as mutations, amplifications, translocations, and alternative splicing, emerging as novel biomarkers that provide an insight into underlying mechanisms of resistance and hold potential predictive value to inform treatment selection. We also describe how liquid biopsies for sampling of circulating markers and ultrasensitive detection technologies have emerged which complement ongoing efforts for biomarker discovery and analysis. Expert commentary: While evidence suggests that genomic aberrations in ER and HER2 could contribute to meeting the pressing need for better predictive biomarkers, efforts need to be made to standardize assessment methods and better understand the resistance mechanisms these markers denote. Taking advantage of emerging technologies, research in upcoming years should include prospective trials incorporating these predictors into the study design to validate their potential clinical value.
Collapse
Affiliation(s)
- Carlos Martínez-Pérez
- a Breast Cancer Now Edinburgh Team, Institute of Genetics and Molecular Medicine , University of Edinburgh, Western General Hospital , Edinburgh , UK
| | - Arran K Turnbull
- a Breast Cancer Now Edinburgh Team, Institute of Genetics and Molecular Medicine , University of Edinburgh, Western General Hospital , Edinburgh , UK
| | - J Michael Dixon
- a Breast Cancer Now Edinburgh Team, Institute of Genetics and Molecular Medicine , University of Edinburgh, Western General Hospital , Edinburgh , UK.,b Edinburgh Breast Unit , Western General Hospital , Edinburgh , UK
| |
Collapse
|
241
|
Meléndez B, Van Campenhout C, Rorive S, Remmelink M, Salmon I, D'Haene N. Methods of measurement for tumor mutational burden in tumor tissue. Transl Lung Cancer Res 2018; 7:661-667. [PMID: 30505710 DOI: 10.21037/tlcr.2018.08.02] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Immunotherapies based on immune checkpoint inhibitors are emerging as an innovative treatment for different types of advanced cancers. While the utility of immune checkpoint inhibitors has been clearly demonstrated, the response rate is highly variable across individuals. Due to the cost and toxicity of these immunotherapies, a critical challenge in this field is the identification of predictive biomarkers to discriminate which patients may respond to immunotherapy. Recently, a high tumor mutational burden (TMB) has been identified as a genetic signature that is associated with a favorable outcome for immune checkpoint inhibitor therapy. The TMB is defined as the total number of nonsynonymous mutations per coding area of a tumor genome. Initially, it was determined using whole exome sequencing, but due to the high costs and long turnaround time of this method, targeted panel sequencing is currently being explored to measure TMB. In the near future, TMB evaluation may play an important role in immuno-oncology, but its implementation in a routine setting involves robust analytical and clinical validation. Standardization is also needed in order to make informed decisions about patients. This review presents the methodologies employed for determining TMB and discusses the factors that may have an impact on its measurement.
Collapse
Affiliation(s)
- Bárbara Meléndez
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Claude Van Campenhout
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Sandrine Rorive
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Myriam Remmelink
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Isabelle Salmon
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Nicky D'Haene
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
242
|
Devecchi A, De Cecco L, Dugo M, Penso D, Dagrada G, Brich S, Stacchiotti S, Sensi M, Canevari S, Pilotti S. The genomics of desmoplastic small round cell tumor reveals the deregulation of genes related to DNA damage response, epithelial-mesenchymal transition, and immune response. Cancer Commun (Lond) 2018; 38:70. [PMID: 30486883 PMCID: PMC6260689 DOI: 10.1186/s40880-018-0339-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 11/16/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Desmoplastic small round cell tumor (DSRCT) is a rare, aggressive, and poorly investigated simple sarcoma with a low frequency of genetic deregulation other than an Ewing sarcoma RNA binding protein 1 (EWSR1)-Wilm's tumor suppressor (WT1) translocation. We used whole-exome sequencing to interrogate six consecutive pre-treated DSRCTs whose gene expression was previously investigated. METHODS DNA libraries were prepared from formalin-fixed, paraffin-embedded archival tissue specimens following the Agilent SureSelectXT2 target enrichment protocol and sequenced on Illumina NextSeq 500. Raw sequence data were aligned to the reference genome with Burrows-Wheeler Aligner algorithm. Somatic mutations and copy number alterations (CNAs) were identified using MuTect2 and EXCAVATOR2, respectively. Biological functions associated with altered genes were investigated through Ingenuity Pathway Analysis (IPA) software. RESULTS A total of 137 unique somatic mutations were identified: 133 mutated genes were case-specific, and 2 were mutated in two cases but in different positions. Among the 135 mutated genes, 27% were related to two biological categories: DNA damage-response (DDR) network that was also identified through IPA and mesenchymal-epithelial reverse transition (MErT)/epithelial-mesenchymal transition (EMT) already demonstrated to be relevant in DSRCT. The mutated genes in the DDR network were involved in various steps of transcription and particularly affected pre-mRNA. Half of these genes encoded RNA-binding proteins or DNA/RNA-binding proteins, which were recently recognized as a new class of DDR players. CNAs in genes/gene families, involved in MErT/EMT and DDR, were recurrent across patients and mostly segregated in the MErT/EMT category. In addition, recurrent gains of regions in chromosome 1 involving many MErT/EMT gene families and loss of one arm or the entire chromosome 6 affecting relevant immune-regulatory genes were recorded. CONCLUSIONS The emerging picture is an extreme inter-tumor heterogeneity, characterized by the concurrent deregulation of the DDR and MErT/EMT dynamic and plastic programs that could favour genomic instability and explain the refractory DSRCT profile.
Collapse
Affiliation(s)
- Andrea Devecchi
- Platform of Integrated Biology, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 2133, Milan, Italy
| | - Loris De Cecco
- Platform of Integrated Biology, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 2133, Milan, Italy
| | - Matteo Dugo
- Platform of Integrated Biology, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 2133, Milan, Italy
| | - Donata Penso
- Platform of Integrated Biology, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 2133, Milan, Italy
| | - Gianpaolo Dagrada
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133, Milan, Italy
| | - Silvia Brich
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133, Milan, Italy
| | - Silvia Stacchiotti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133, Milan, Italy
| | - Marialuisa Sensi
- Platform of Integrated Biology, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 2133, Milan, Italy
| | - Silvana Canevari
- Platform of Integrated Biology, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 2133, Milan, Italy.
| | - Silvana Pilotti
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133, Milan, Italy.
| |
Collapse
|
243
|
Mutational Evolution in Relapsed Diffuse Large B-Cell Lymphoma. Cancers (Basel) 2018; 10:cancers10110459. [PMID: 30463380 PMCID: PMC6265691 DOI: 10.3390/cancers10110459] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/12/2018] [Accepted: 11/14/2018] [Indexed: 12/14/2022] Open
Abstract
Current genomic models in diffuse large B-cell lymphoma (DLBCL) are based on single tumor biopsies, which might underestimate heterogeneity. Data on mutational evolution largely remains unknown. An exploratory study using whole exome sequencing on paired (primary and relapse) formalin fixed paraffin embedded DLBCL biopsies (n = 14) of 6 patients was performed to globally assess the mutational evolution and to identify gene mutations specific for relapse samples from patients treated with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisolone. A minority of the mutations detected in the primary sample (median 7.6%, range 4.8–66.2%) could not be detected in the matching relapse sample. Relapsed DLBCL samples showed a mild increase of mutations (median 12.5%, range 9.4–87.6%) as compared to primary tumor biopsies. We identified 264 genes possibly related to therapy resistance, including tyrosine kinases (n = 18), (transmembrane) glycoproteins (n = 73), and genes involved in the JAK-STAT pathway (n = 7). Among the potentially resistance related genes were PIM1, SOCS1, and MYC, which have been reported to convey a risk for treatment failure. In conclusion, we show modest temporal heterogeneity between paired tumor samples with the acquisition of new mutations and identification of genes possibly related to therapy resistance. The mutational evolution could have implications for treatment decisions and development of novel targeted drugs.
Collapse
|
244
|
Lu XJD, Liu KYP, Zhu YS, Cui C, Poh CF. Using ddPCR to assess the DNA yield of FFPE samples. BIOMOLECULAR DETECTION AND QUANTIFICATION 2018; 16:5-11. [PMID: 30560062 PMCID: PMC6287546 DOI: 10.1016/j.bdq.2018.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 10/04/2018] [Accepted: 10/24/2018] [Indexed: 12/24/2022]
Abstract
Objectives Detection of genomic alterations in diseases can be achieved with current molecular technologies. However, the molecules extracted from formalin-fixed, paraffin-embedded (FFPE) bio-samples are often limited possibly due to DNA fragmentation and crosslinking caused by the sample fixation and processing. The study objective was to design a droplet digital PCR (ddPCR) assay to assess the quality and quantity of DNA derived from various DNA extraction conditions on FFPE samples. Methods We used 10 μm-thick sections from 5 FFPE oral tumoral blocks, each consisting of 10–15 sections. The protocol variables tested included: 1) tissue staining; 2) duration and 3) temperature of post-digestion heat treatment; and 4) DNA extraction method. DNA quantity was assessed using the NanoDrop 2000 (Thermo Fisher Scientific, USA), the Qubit fluorometer (Thermo Fisher Scientific, USA), and a ddPCR-based assay. DNA quality was assessed using a ddPCR assay for the degree of fragmentation and the effectiveness of removing crosslinks with varying guanine-cytosine (GC)-content. Results Deparaffinization with xylene helped to increase the DNA yield. Tissue staining (methyl green staining, pH 6) prior to microdissection, comparing to no staining, caused additional DNA fragmentation. Compared to column-based method, DNA extracted with phenol chloroform and ethanol precipitation increased the degree of fragmentation and lowered the yield of amplifiable DNA. The cross-linking derived from GC-contents may not be the only factor impacting on the DNA quality. Conclusions Samples undergoing different pre-treatment conditions prior to extraction can impact the yield of amplifiable DNA. Our ddPCR assay can be used to assess for both DNA quantity and quality.
Collapse
Affiliation(s)
- X J David Lu
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.,Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z 1L3, Canada
| | - Kelly Y P Liu
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.,Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z 1L3, Canada
| | - Yuqi Sarah Zhu
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z 1L3, Canada
| | - Cindy Cui
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z 1L3, Canada
| | - Catherine F Poh
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.,Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z 1L3, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada
| |
Collapse
|
245
|
Amemiya K, Hirotsu Y, Oyama T, Omata M. Relationship between formalin reagent and success rate of targeted sequencing analysis using formalin fixed paraffin embedded tissues. Clin Chim Acta 2018; 488:129-134. [PMID: 30395866 DOI: 10.1016/j.cca.2018.11.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/23/2018] [Accepted: 11/02/2018] [Indexed: 11/26/2022]
Abstract
BACKGROUND Tumor genetic alterations are determined to aid in selecting therapy and predicting prognosis. In routine clinical practice, targeted sequencing analysis is performed using formalin-fixed paraffin embedded (FFPE) tissues. However, successful genetic analysis remains challenging because FFPE DNA is fragmented during the sample preparation process. METHODS Real-time PCR was performed to assess DNA quality and quantities. Targeted sequencing was performed using FFPE tissues fixed with different types of formalin. RESULTS DNA was less fragmented from samples fixed in low formalin concentration (10% vs. 20%) and neutral buffered conditions (neutral buffered vs. non-neutral). DNA fragmentation increased over the fixation time. In a preliminary test study, we compared fixation using 10% neutral buffered formalin (n = 180) and 20% formalin (n = 26). The success rate of targeted analysis was higher using 10% neutral formalin (98.3%; 177/180) compared with 20% formalin (34.6%; 9/26). In a validation study with additional formalin-fixed paraffin embedded tissues fixed with 10% neutral buffered formalin (n = 860), we reproduced these results and achieved a high success rate for targeted sequencing analysis (98.4%; 846/860). CONCLUSION Our data show that 10% neutral buffered formalin is recommended for fixation of formalin-fixed paraffin embedded samples to achieve high success rate of targeted sequencing analysis.
Collapse
Affiliation(s)
- Kenji Amemiya
- Genome Analysis Center, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi 400-8506, Japan
| | - Yosuke Hirotsu
- Genome Analysis Center, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi 400-8506, Japan.
| | - Toshio Oyama
- Department of Pathology, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi 400-8506, Japan
| | - Masao Omata
- Genome Analysis Center, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi 400-8506, Japan; The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
| |
Collapse
|
246
|
Liu Y, He Q, Sun W. Association analysis using somatic mutations. PLoS Genet 2018; 14:e1007746. [PMID: 30388102 PMCID: PMC6235399 DOI: 10.1371/journal.pgen.1007746] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 11/14/2018] [Accepted: 10/07/2018] [Indexed: 11/18/2022] Open
Abstract
Somatic mutations drive the growth of tumor cells and are pivotal biomarkers for many cancer treatments. Genetic association analysis using somatic mutations is an effective approach to study the functional impact of somatic mutations. However, standard regression methods are not appropriate for somatic mutation association studies because somatic mutation calls often have non-ignorable false positive rate and/or false negative rate. While large scale association analysis using somatic mutations becomes feasible recently—thanks for the improvement of sequencing techniques and the reduction of sequencing cost—there is an urgent need for a new statistical method designed for somatic mutation association analysis. We propose such a method with computationally efficient software implementation: Somatic mutation Association test with Measurement Errors (SAME). SAME accounts for somatic mutation calling uncertainty using a likelihood based approach. It can be used to assess the associations between continuous/dichotomous outcomes and individual mutations or gene-level mutations. Through simulation studies across a wide range of realistic scenarios, we show that SAME can significantly improve statistical power than the naive generalized linear model that ignores mutation calling uncertainty. Finally, using the data collected from The Cancer Genome Atlas (TCGA) project, we apply SAME to study the associations between somatic mutations and gene expression in 12 cancer types, as well as the associations between somatic mutations and colon cancer subtype defined by DNA methylation data. SAME recovered some interesting findings that were missed by the generalized linear model. In addition, we demonstrated that mutation-level and gene-level analyses are often more appropriate for oncogene and tumor-suppressor gene, respectively. Cancer is a genetic disease that is driven by the accumulation of somatic mutations. Association studies using somatic mutations is a powerful approach to identify the potential impact of somatic mutations on molecular or clinical features. One challenge for such tasks is the non-ignorable somatic mutation calling errors. We have developed a statistical method to address this challenge and applied our method to study the gene expression traits associated with somatic mutations in 12 cancer types. Our results show that some somatic mutations affect gene expression in several cancer types. In particular, we show that the associations between gene expression traits and TP53 gene level mutation reveal some similarities across a few cancer types.
Collapse
Affiliation(s)
- Yang Liu
- Department of Mathematics and Statistics, Wright State University, Dayton, Ohio, United States of America
| | - Qianchan He
- Biostatistics Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Wei Sun
- Biostatistics Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
247
|
Zhang A, Li S, Apone L, Sun X, Chen L, Ettwiller LM, Langhorst BW, Noren CJ, Xu MQ. Solid-phase enzyme catalysis of DNA end repair and 3' A-tailing reduces GC-bias in next-generation sequencing of human genomic DNA. Sci Rep 2018; 8:15887. [PMID: 30367148 PMCID: PMC6203771 DOI: 10.1038/s41598-018-34079-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 10/06/2018] [Indexed: 01/28/2023] Open
Abstract
The use of next-generation sequencing (NGS) has been instrumental in advancing biological research and clinical diagnostics. To fully utilize the power of NGS, complete, uniform coverage of the entire genome is required. In this study, we identified the primary sources of bias observed in sequence coverage across AT-rich regions of the human genome with existing amplification-free DNA library preparation methods. We have found evidence that a major source of bias is the inefficient processing of AT-rich DNA in end repair and 3' A-tailing, causing under-representation of extremely AT-rich regions. We have employed immobilized DNA modifying enzymes to catalyze end repair and 3' A-tailing reactions, to notably reduce the GC bias observed with existing library construction methods.
Collapse
Affiliation(s)
- Aihua Zhang
- New England Biolabs, Inc., 240 County Road, Ipswich, MA, 01938, USA
| | - Shaohua Li
- New England Biolabs, Inc., 240 County Road, Ipswich, MA, 01938, USA
| | - Lynne Apone
- New England Biolabs, Inc., 240 County Road, Ipswich, MA, 01938, USA
| | - Xiaoli Sun
- New England Biolabs, Inc., 240 County Road, Ipswich, MA, 01938, USA
| | - Lixin Chen
- New England Biolabs, Inc., 240 County Road, Ipswich, MA, 01938, USA
| | | | | | | | - Ming-Qun Xu
- New England Biolabs, Inc., 240 County Road, Ipswich, MA, 01938, USA.
| |
Collapse
|
248
|
Matsuo Y, Yoshida T, Yamashita K, Satoh Y. Reducing DNA damage by formaldehyde in liquid-based cytology preservation solutions to enable the molecular testing of lung cancer specimens. Cancer Cytopathol 2018; 126:1011-1021. [PMID: 30343520 DOI: 10.1002/cncy.22069] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/18/2018] [Accepted: 09/11/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Liquid-based cytology (LBC) is a useful cytopathological method, and LBC lung adenocarcinoma specimens may be used for genetic analysis in the near future. In the current study, the authors determined whether LBC specimens can be used for epidermal growth factor receptor (EGFR) mutation analysis in human lung adenocarcinoma cell lines. METHODS Genomic DNA was extracted from 3 lung adenocarcinoma cell lines that were fixed in LBC preservation solution using 2 protocols (one for cultured cells and one for tissues) of a DNA extraction kit. Different fixation times were tested for each protocol: 30 minutes, 1 hour, and 1 to 9 days. As controls, cells also were fixed in 10% formalin or 95% ethanol. The authors investigated the effect of fixation time on DNA fragmentation, polymerase chain reaction (PCR) amplification, and EGFR mutation detection. RESULTS The DNA yield of LBC specimens tended to decrease depending on fixation time. When using the DNA extraction protocol for tissues, PCR amplification was successful after 9 days of fixation, although extracted genomic DNA that was fixed for >1 hour demonstrated fragmentation. Mutation analyses using the Cycleave PCR method were successful after 7 days of fixation. The DNA extraction protocol for tissues was appropriate for lung adenocarcinoma cell lines that were stored for >1 day in a preservative solution. The results of the current study demonstrated that EGFR mutations can be detected on day 7 using lung adenocarcinoma cell lines fixed in CytoRich Red preservative. CONCLUSIONS When LBC specimens are used for targeted molecular genetic testing, the appropriate preservative solution and extraction protocol first should be determined.
Collapse
Affiliation(s)
- Yukiko Matsuo
- Department of Thoracic Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Tsutomu Yoshida
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan.,Department of Pathology, Kitasato University Hospital, Kanagawa, Japan
| | - Kazuya Yamashita
- Department of Pathology, Kitasato University Hospital, Kanagawa, Japan
| | - Yukitoshi Satoh
- Department of Thoracic Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| |
Collapse
|
249
|
Rosa-Rosa JM, Caniego-Casas T, Leskela S, Muñoz G, Del Castillo F, Garrido P, Palacios J. Modified SureSelect QXT Target Enrichment Protocol for Illumina Multiplexed Sequencing of FFPE Samples. Biol Proced Online 2018; 20:19. [PMID: 30337841 PMCID: PMC6182866 DOI: 10.1186/s12575-018-0084-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/20/2018] [Indexed: 12/18/2022] Open
Abstract
Background Personalised medicine is nowadays a major objective in oncology. Molecular characterization of tumours through NGS offers the possibility to find possible therapeutic targets in a time- and cost-effective way. However, the low quality and complexity of FFPE DNA samples bring a series of disadvantages for massive parallel sequencing techniques compared to high-quality DNA samples (from blood cells, cell cultures, etc.). Results We performed several experiments to understand the behaviour of FFPE DNA samples during the construction of SureSelectQXT libraries. First, we designed a quality checkpoint for FFPE DNA samples based on the quantification of their amplification capability (qcPCR). We observed that FFPE DNA samples can be classified according to DIN value and qcPCR concentration into unusable, or low-quality (LQ) and good-quality (GQ) DNA. For GQ samples, we increased the amount of input DNA to 150 ng and the digestion time to 30 min, whereas for LQ samples, we used 50 ng of DNA as input but we decreased the digestion time to 1 min. In all cases, we increased the cycles of the pre-hyb PCR to 10 but decreased the cycles of the post-hyb PCR to 8. In addition, we confirmed that using half of the volume of reagents can be beneficial. Finally, in order to obtain better results, we designed a decision flow-chart to achieve a seeding concentration of 12–14 pM for MiSeq Reagent Kit v2. Conclusions Our experiments allowed us to unveil the behaviour of low-quality FFPE DNA samples during the construction of SureSelectQXT libraries. Sequencing results showed that, using our modified SureSelectQXT protocol, the final percentage of usable reads for low-quality samples was increased more than three times allowing to reach median depth/million reads values of 76.35. This value is equivalent to ~ 0.9 and ~ 0.7 of the values obtained for good-quality FFPE and high-quality DNA respectively. Electronic supplementary material The online version of this article (10.1186/s12575-018-0084-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- J M Rosa-Rosa
- 1CIBER-ONC, Instituto de Salud Carlos III, Madrid, Spain
| | - T Caniego-Casas
- 2Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - S Leskela
- 1CIBER-ONC, Instituto de Salud Carlos III, Madrid, Spain.,2Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - G Muñoz
- 2Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - F Del Castillo
- 2Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain.,3Servicio de Genética, Hospital Universitario Ramón y Cajal, Madrid, Spain.,4CIBER-ER, Instituto de Salud Carlos III, Madrid, Spain
| | - P Garrido
- 2Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain.,5Medical Oncology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain.,6Facultad de Medicina, Universidad de Alcalá de Henares, Madrid, Spain
| | - J Palacios
- 1CIBER-ONC, Instituto de Salud Carlos III, Madrid, Spain.,2Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain.,6Facultad de Medicina, Universidad de Alcalá de Henares, Madrid, Spain.,7Servicio de Anatomía Patológica, Hospital Ramón y Cajal, Ctra. Colmenar Viejo km 9,100, 28034 Madrid, Spain
| |
Collapse
|
250
|
Robbe P, Popitsch N, Knight SJL, Antoniou P, Becq J, He M, Kanapin A, Samsonova A, Vavoulis DV, Ross MT, Kingsbury Z, Cabes M, Ramos SDC, Page S, Dreau H, Ridout K, Jones LJ, Tuff-Lacey A, Henderson S, Mason J, Buffa FM, Verrill C, Maldonado-Perez D, Roxanis I, Collantes E, Browning L, Dhar S, Damato S, Davies S, Caulfield M, Bentley DR, Taylor JC, Turnbull C, Schuh A. Clinical whole-genome sequencing from routine formalin-fixed, paraffin-embedded specimens: pilot study for the 100,000 Genomes Project. Genet Med 2018; 20:1196-1205. [PMID: 29388947 PMCID: PMC6520241 DOI: 10.1038/gim.2017.241] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 11/06/2017] [Indexed: 12/16/2022] Open
Abstract
PURPOSE Fresh-frozen (FF) tissue is the optimal source of DNA for whole-genome sequencing (WGS) of cancer patients. However, it is not always available, limiting the widespread application of WGS in clinical practice. We explored the viability of using formalin-fixed, paraffin-embedded (FFPE) tissues, available routinely for cancer patients, as a source of DNA for clinical WGS. METHODS We conducted a prospective study using DNAs from matched FF, FFPE, and peripheral blood germ-line specimens collected from 52 cancer patients (156 samples) following routine diagnostic protocols. We compared somatic variants detected in FFPE and matching FF samples. RESULTS We found the single-nucleotide variant agreement reached 71% across the genome and somatic copy-number alterations (CNAs) detection from FFPE samples was suboptimal (0.44 median correlation with FF) due to nonuniform coverage. CNA detection was improved significantly with lower reverse crosslinking temperature in FFPE DNA extraction (80 °C or 65 °C depending on the methods). Our final data showed somatic variant detection from FFPE for clinical decision making is possible. We detected 98% of clinically actionable variants (including 30/31 CNAs). CONCLUSION We present the first prospective WGS study of cancer patients using FFPE specimens collected in a routine clinical environment proving WGS can be applied in the clinic.
Collapse
Affiliation(s)
- Pauline Robbe
- Oxford Molecular Diagnostics Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| | - Niko Popitsch
- Wellcome Trust Centre of Human Genetics, University of Oxford, Old Road Campus Research Building, Oxford, UK
| | - Samantha J L Knight
- Wellcome Trust Centre of Human Genetics, University of Oxford, Old Road Campus Research Building, Oxford, UK
| | - Pavlos Antoniou
- Oxford Molecular Diagnostics Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Jennifer Becq
- Illumina Cambridge Ltd., Chesterford Research Park, Saffron Walden, UK
| | - Miao He
- Illumina Cambridge Ltd., Chesterford Research Park, Saffron Walden, UK
| | | | | | - Dimitrios V Vavoulis
- Oxford Molecular Diagnostics Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Mark T Ross
- Illumina Cambridge Ltd., Chesterford Research Park, Saffron Walden, UK
| | - Zoya Kingsbury
- Illumina Cambridge Ltd., Chesterford Research Park, Saffron Walden, UK
| | - Maite Cabes
- Oxford Molecular Diagnostics Centre, John Radcliffe Hospital, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Sara D C Ramos
- Oxford Molecular Diagnostics Centre, John Radcliffe Hospital, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Suzanne Page
- Oxford Molecular Diagnostics Centre, John Radcliffe Hospital, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Helene Dreau
- Oxford Molecular Diagnostics Centre, John Radcliffe Hospital, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Kate Ridout
- Oxford Molecular Diagnostics Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Louise J Jones
- Genomics England, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Alice Tuff-Lacey
- Genomics England, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Shirley Henderson
- Oxford Molecular Diagnostics Centre, John Radcliffe Hospital, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Joanne Mason
- Genomics England, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Francesca M Buffa
- Computational Biology and Integrative Genomics, Department of Oncology, University of Oxford, Oxford, UK
| | - Clare Verrill
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - David Maldonado-Perez
- Department of Cellular Pathology, Oxford University Hospital Foundation Trust, Oxford, UK
| | - Ioannis Roxanis
- Department of Cellular Pathology, Oxford University Hospital Foundation Trust, Oxford, UK
| | - Elena Collantes
- Department of Cellular Pathology, Oxford University Hospital Foundation Trust, Oxford, UK
| | - Lisa Browning
- Department of Cellular Pathology, Oxford University Hospital Foundation Trust, Oxford, UK
| | - Sunanda Dhar
- Department of Cellular Pathology, Oxford University Hospital Foundation Trust, Oxford, UK
| | - Stephen Damato
- Department of Cellular Pathology, Oxford University Hospital Foundation Trust, Oxford, UK
| | - Susan Davies
- Department of Cellular Pathology, Oxford University Hospital Foundation Trust, Oxford, UK
| | - Mark Caulfield
- Genomics England, William Harvey Research Institute, Queen Mary University of London, London, UK
- NIHR Biomedical Research Centre at Barts Health NHS Trust, London, UK
| | - David R Bentley
- Illumina Cambridge Ltd., Chesterford Research Park, Saffron Walden, UK
| | - Jenny C Taylor
- Wellcome Trust Centre of Human Genetics, University of Oxford, Old Road Campus Research Building, Oxford, UK
- NIHR Comprehensive Biomedical Research Centre, Oxford, UK
| | - Clare Turnbull
- Genomics England, William Harvey Research Institute, Queen Mary University of London, London, UK
- Department of Cellular Pathology, Oxford University Hospital Foundation Trust, Oxford, UK
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Anna Schuh
- Oxford Molecular Diagnostics Centre, John Radcliffe Hospital, Oxford University Hospitals NHS Trust, Oxford, UK
- NIHR Comprehensive Biomedical Research Centre, Oxford, UK
- Oxford Molecular Diagnostics Centre, Department of Oncology, University of Oxford, Oxford, UK
| |
Collapse
|