1
|
Kadri S, Craven KE, Fussell AM, Gee EPS, Jordan D, Klee EW, Krumm N, Temple-Smolkin RL, Zehir A, Zhang W, Sboner A. Clinical Bioinformatician Body of Knowledge-Bioinformatics and Software Core: A Report of the Association for Molecular Pathology. J Mol Diagn 2025:S1525-1578(25)00118-7. [PMID: 40398560 DOI: 10.1016/j.jmoldx.2025.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 03/07/2025] [Accepted: 04/10/2025] [Indexed: 05/23/2025] Open
Abstract
With the evolution of next-generation sequencing-based testing in molecular diagnostics laboratories, the clinical role of bioinformaticians has also evolved. The Association for Molecular Pathology's Clinical Bioinformatician Body of Knowledge aims to define the various roles the clinical bioinformatician operates individually or within a clinical bioinformatics team, along with proficiencies and skill sets that may be required or desirable across these roles. One of the most common professional responsibilities of a clinical bioinformatician is to implement bioinformatics pipelines, either vendor supplied or custom built for the assays in the molecular diagnostics laboratory, along with analysis and quality control of clinical genomics data. This second article in the series describes the various stages in the life cycle of a clinical bioinformatics pipeline and the considerations, areas of expertise, and skill sets required in each stage. This information may help laboratory professionals to better work with clinical bioinformaticians and laboratory directors to hire the appropriate expertise based on the specific needs of the laboratory.
Collapse
Affiliation(s)
- Sabah Kadri
- AMP Clinical Bioinformatician Body of Knowledge Bioinformatics and Software Knowledge Core Working Group of the Informatics Subdivision, Association for Molecular Pathology, Rockville, Maryland; Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois; AbbVie, North Chicago, Illinois.
| | - Kelly E Craven
- AMP Clinical Bioinformatician Body of Knowledge Bioinformatics and Software Knowledge Core Working Group of the Informatics Subdivision, Association for Molecular Pathology, Rockville, Maryland; Johns Hopkins University School of Medicine, Baltimore, Maryland; Memorial Sloan Kettering Cancer Center, New York, New York
| | - Amber M Fussell
- The Association for Molecular Pathology, Rockville, Maryland
| | - Elaine P S Gee
- AMP Clinical Bioinformatician Body of Knowledge Bioinformatics and Software Knowledge Core Working Group of the Informatics Subdivision, Association for Molecular Pathology, Rockville, Maryland; BigHead Analytics Group, Windsor, California; Mirvie, Inc., South San Francisco, California
| | - Danielle Jordan
- The Association for Molecular Pathology, Rockville, Maryland
| | - Eric W Klee
- AMP Clinical Bioinformatician Body of Knowledge Bioinformatics and Software Knowledge Core Working Group of the Informatics Subdivision, Association for Molecular Pathology, Rockville, Maryland; Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Niklas Krumm
- AMP Clinical Bioinformatician Body of Knowledge Bioinformatics and Software Knowledge Core Working Group of the Informatics Subdivision, Association for Molecular Pathology, Rockville, Maryland; University of Washington, Seattle, Washington
| | | | - Ahmet Zehir
- AMP Clinical Bioinformatician Body of Knowledge Bioinformatics and Software Knowledge Core Working Group of the Informatics Subdivision, Association for Molecular Pathology, Rockville, Maryland; Memorial Sloan Kettering Cancer Center, New York, New York
| | - Weiwei Zhang
- AMP Clinical Bioinformatician Body of Knowledge Bioinformatics and Software Knowledge Core Working Group of the Informatics Subdivision, Association for Molecular Pathology, Rockville, Maryland; Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Andrea Sboner
- AMP Clinical Bioinformatician Body of Knowledge Bioinformatics and Software Knowledge Core Working Group of the Informatics Subdivision, Association for Molecular Pathology, Rockville, Maryland; Department of Pathology and Laboratory Medicine, Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Meyer Cancer Center, Weill Cornell Medicine, New York, New York
| |
Collapse
|
2
|
Loderer D, Hornáková A, Tobiášová K, Lešková K, Halašová E, Danková Z, Biringer K, Kúdela E, Rokos T, Dzian A, Miklušica J, Mikolajčík P, Smolár M, Plank L, Grendár M. Comparison of next‑generation sequencing quality metrics and concordance in the detection of cancer‑specific molecular alterations between formalin‑fixed paraffin‑embedded and fresh‑frozen samples in comprehensive genomic profiling with the Illumina® TruSight Oncology 500 assay. Exp Ther Med 2025; 29:64. [PMID: 39991725 PMCID: PMC11843196 DOI: 10.3892/etm.2025.12814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/19/2024] [Indexed: 02/25/2025] Open
Abstract
Next-generation sequencing (NGS) technology is routinely employed to detect clinically significant variants in the field of precision medicine. Formalin-fixed paraffin-embedded (FFPE) tissues remain a widely used source of genetic material for diagnostic purposes due to their long-term storage stability, which preserves the architecture of tumour tissues. However, the degradation of nucleic acids (NAs) that occurs during the fixation process can lead to unreliable results or hinder analysis. Considering the challenges posed by the quality and quantity of nucleic acids extracted from FFPE samples, the present study aimed to compare paired fresh-frozen (FF) and FFPE samples. The present study also aimed to assess the concordance rate of quality control metrics of sequenced libraries and identify variants and biomarkers between FFPE and FF tissues. The Illumina® TruSight Oncology 500 assay (TSO 500; Illumina, Inc.) was used to conduct comprehensive genomic profiling of samples from patients with various cancer types. All identified alterations were annotated using the Pierian Dx Clinical Genomics Workspace version 6.20 (PierianDx CGW). A total of 138 DNA and 138 RNA analyses on 69 paired samples were performed. These results demonstrated the significant potential of FF tissue as a primary source of higher-quality genetic material to detect small variants, microsatellite instabilities and the tumour mutational burden, using the TSO 500 assay compared with FFPE samples. This approach could effectively reduce the issues associated with poor-quality NAs extracted from FFPE samples. Based on our findings of lower concordance in the detection of splice variants, fusions and copy number variants in paired samples, we recommend that future studies using the TSO 500 assay should focus directly on this issue.
Collapse
Affiliation(s)
- Dušan Loderer
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, 036 01 Martin, Slovakia
| | - Andrea Hornáková
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, 036 01 Martin, Slovakia
| | - Katarína Tobiášová
- Department of Pathological Anatomy, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava and University Hospital in Martin, 036 01 Martin, Slovakia
| | - Katarína Lešková
- Department of Pathological Anatomy, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava and University Hospital in Martin, 036 01 Martin, Slovakia
| | - Erika Halašová
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, 036 01 Martin, Slovakia
| | - Zuzana Danková
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, 036 01 Martin, Slovakia
- Biobank for Cancer and Rare Diseases, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, 036 01 Martin, Slovakia
| | - Kamil Biringer
- Clinic of Obstetrics and Gynaecology, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, 036 01 Martin, Slovakia
| | - Erik Kúdela
- Clinic of Obstetrics and Gynaecology, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, 036 01 Martin, Slovakia
| | - Tomáš Rokos
- Clinic of Obstetrics and Gynaecology, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, 036 01 Martin, Slovakia
| | - Anton Dzian
- Department of Thoracic Surgery, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, 036 59 Martin, Slovakia
| | - Juraj Miklušica
- Clinic of General, Visceral and Transplant Surgery, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, 036 59 Martin, Slovakia
| | - Peter Mikolajčík
- Clinic of General, Visceral and Transplant Surgery, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, 036 59 Martin, Slovakia
| | - Marek Smolár
- Clinic of General, Visceral and Transplant Surgery, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, 036 59 Martin, Slovakia
| | - Lukáš Plank
- Department of Pathological Anatomy, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava and University Hospital in Martin, 036 01 Martin, Slovakia
| | - Marián Grendár
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, 036 01 Martin, Slovakia
| |
Collapse
|
3
|
GÖKER BAGCA B, GÖDE S, TURHAL G, ÖZATEŞ NP, VERAL A, GÜNDÜZ C, AVCI ÇB. Nadir paranazal sinüs kanserlerinde yeni tanımlanan reseptör tirozin kinaz mutasyonları ve potansiyel fonksiyonel etkileri. EGE TIP DERGISI 2023. [DOI: 10.19161/etd.1262612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023] Open
Abstract
Amaç: Paranazal sinüs kanserleri oldukça nadir görülen heterojen bir hastalık grubudur. Maksiler sinüs skuamoz hücreli karsinomu, paranazal sinüs kanserlerinin anatomik ve histolojik olarak en
yaygın alt tipidir. Bu kanserin genetik profiline dair bilginin sınırlı olması, hastaların hedefli tedavi seçeneklerinden yararlanamamasına neden olmaktadır. Çalışmamızda bu nadir kanserdeki reseptör tirozin kinaz mutasyonlarının tanımlanması ve mutasyonların olası fonksiyonel etkilerinin tahmin edilmesi amaçlanmıştır.
Gereç ve Yöntem: Bu amaçla 30 olgunun tümörüne ait FFPE dokulardan DNA izolasyonu gerçekleştirildi, olguların mutasyon profili yeni nesil sekanslama yöntemi ve biyoinformatik
değerlendirme ile belirlendi. Belirlenen patojenik/ olası patojenik varyantların fonksiyonel etkileri farklı in silico araçlar yardımıyla tahminlendi.
Bulgular: Olgularının tamamında en az bir adet patojenik/olası patojenik KIT, PDFGRA ve RETmutasyonu belirlendi. KIT geninin katalitik bölgesindeki mutasyonların kinaz aktivitesini arttıracağı
tahmin edildi. PDFGRA genindeki p.P567P ve p.D1074D mutasyonları, 30 olgunun tamamında ve SRA veritabanından elde edilen normal dokulara ait okumaların tümünde belirlendi.
Sonuç: Reseptör tirozin kinaz mutasyonlarının paranazal sinüs kanserlerinde de önemli rol oynayabileceğinin belirlenmiş olması özellikle artmış kinaz aktivitesini hedefleyen tedavi yaklaşımlarını
bu olguların erişimine sunma potansiyeli taşıması bakımından oldukça önemlidir.
Collapse
Affiliation(s)
- Bakiye GÖKER BAGCA
- Aydın Adnan Menderes Üniversitesi, Tıp Fakültesi, Tıbbi Biyoloji Anabilim Dalı, Aydın, Türkiye
| | - Sercan GÖDE
- Ege Üniversitesi, Tıp Fakültesi, Kulak Burun Boğaz Anabilim Dalı, İzmir, Türkiye
| | - Göksel TURHAL
- Ege Üniversitesi, Tıp Fakültesi, Kulak Burun Boğaz Anabilim Dalı, İzmir, Türkiye
| | - Neslihan Pınar ÖZATEŞ
- Harran Üniversitesi, Tıp Fakültesi, Tıbbi Biyoloji Anabilim Dalı, Şanlıurfa, Türkiye
| | - Ali VERAL
- Ege Üniversitesi, Tıp Fakültesi, Patoloji Anabilim Dalı, İzmir, Türkiye
| | - Cumhur GÜNDÜZ
- Ege Üniversitesi, Tıp Fakültesi, Tıbbi Biyoloji Anabilim Dalı, İzmir, Türkiye
| | - Çığır Biray AVCI
- Ege Üniversitesi, Tıp Fakültesi, Tıbbi Biyoloji Anabilim Dalı, İzmir, Türkiye
| |
Collapse
|
4
|
Fink JL, Jaradi B, Stone N, Anderson L, Leo PJ, Marshall M, Ellis J, Waring PM, O'Byrne K. Minimizing Sample Failure Rates for Challenging Clinical Tumor Samples. J Mol Diagn 2023; 25:263-273. [PMID: 36773702 DOI: 10.1016/j.jmoldx.2023.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 01/04/2023] [Accepted: 01/19/2023] [Indexed: 02/11/2023] Open
Abstract
Identification of somatic variants in cancer by high-throughput sequencing has become common clinical practice, largely because many of these variants may be predictive biomarkers for targeted therapies. However, there can be high sample quality control (QC) failure rates for some tests that prevent the return of results. Stem-loop inhibition mediated amplification (SLIMamp) is a patented technology that has been incorporated into commercially available cancer next-generation sequencing testing kits. The claimed advantage is that these kits can interrogate challenging formalin-fixed, paraffin-embedded tissue samples with low tumor purity, poor-quality DNA, and/or low-input DNA, resulting in a high sample QC pass rate. The study aimed to substantiate that claim using Pillar Biosciences oncoReveal Solid Tumor Panel. Forty-eight samples that had failed one or more preanalytical QC sample parameters for whole-exome sequencing from the Australian Translational Genomics Center's ISO15189-accredited diagnostic genomics laboratory were acquired. XING Genomic Services performed an exploratory data analysis to characterize the samples and then tested the samples in their ISO15189-accredited laboratory. Clinical reports could be generated for 37 (77%) samples, of which 29 (60%) contained clinically actionable or significant variants that would not otherwise have been identified. Eleven samples were deemed unreportable, and the sequencing data were likely dominated by artifacts. A novel postsequencing QC metric was developed that can discriminate between clinically reportable and unreportable samples.
Collapse
Affiliation(s)
- J Lynn Fink
- XING Genomic Services, Sinnamon Park, Queensland, Australia; Australian Translational Genomics Centre, Queensland University of Technology, Woolloongabba, Queensland, Australia; The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, Queensland, Australia.
| | - Binny Jaradi
- XING Genomic Services, Sinnamon Park, Queensland, Australia
| | - Nathan Stone
- XING Genomic Services, Sinnamon Park, Queensland, Australia
| | - Lisa Anderson
- Australian Translational Genomics Centre, Queensland University of Technology, Woolloongabba, Queensland, Australia; Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Paul J Leo
- Australian Translational Genomics Centre, Queensland University of Technology, Woolloongabba, Queensland, Australia; Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Mhairi Marshall
- Australian Translational Genomics Centre, Queensland University of Technology, Woolloongabba, Queensland, Australia; Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Jonathan Ellis
- Australian Translational Genomics Centre, Queensland University of Technology, Woolloongabba, Queensland, Australia; Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Paul M Waring
- XING Genomic Services, Sinnamon Park, Queensland, Australia
| | - Kenneth O'Byrne
- Australian Translational Genomics Centre, Queensland University of Technology, Woolloongabba, Queensland, Australia; The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, Queensland, Australia; Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| |
Collapse
|
5
|
Borgognone A, Serna G, Noguera-Julian M, Alonso L, Parera M, Català-Moll F, Sanchez L, Fasani R, Paredes R, Nuciforo P. Performance of 16S Metagenomic Profiling in Formalin-Fixed Paraffin-Embedded versus Fresh-Frozen Colorectal Cancer Tissues. Cancers (Basel) 2021; 13:cancers13215421. [PMID: 34771584 PMCID: PMC8582506 DOI: 10.3390/cancers13215421] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/19/2021] [Accepted: 10/26/2021] [Indexed: 01/04/2023] Open
Abstract
Formalin-fixed, paraffin-embedded (FFPE) tissues represent the most widely available clinical material to study colorectal cancer (CRC). However, the accuracy and clinical validity of FFPE microbiome profiling in CRC is uncertain. Here, we compared the microbial composition of 10 paired fresh-frozen (FF) and FFPE CRC tissues using 16S rRNA sequencing and RNA-ISH. Both sample types showed different microbial diversity and composition. FF samples were enriched in archaea and representative CRC-associated bacteria, such as Firmicutes, Bacteroidetes and Fusobacteria. Conversely, FFPE samples were mainly enriched in typical contaminants, such as Sphingomonadales and Rhodobacterales. RNA-ISH in FFPE tissues confirmed the presence of CRC-associated bacteria, such as Fusobacterium and Bacteroides, as well as Propionibacterium allowing discrimination between tumor-associated and contaminant taxa. An internal quality index showed that the degree of similarity within sample pairs inversely correlated with the dominance of contaminant taxa. Given the importance of FFPE specimens for larger studies in human cancer genomics, our findings may provide useful indications on potential confounding factors to consider for accurate and reproducible metagenomics analyses.
Collapse
Affiliation(s)
- Alessandra Borgognone
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Barcelona, Spain; (A.B.); (M.N.-J.); (M.P.); (F.C.-M.)
| | - Garazi Serna
- Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, 08035 Barcelona, Spain; (G.S.); (L.A.); (L.S.); (R.F.)
| | - Marc Noguera-Julian
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Barcelona, Spain; (A.B.); (M.N.-J.); (M.P.); (F.C.-M.)
- Faculty of Medicine, University of Vic–Central University of Catalonia (UVic–UCC), 08500 Vic, Barcelona, Spain
| | - Lidia Alonso
- Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, 08035 Barcelona, Spain; (G.S.); (L.A.); (L.S.); (R.F.)
| | - Mariona Parera
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Barcelona, Spain; (A.B.); (M.N.-J.); (M.P.); (F.C.-M.)
| | - Francesc Català-Moll
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Barcelona, Spain; (A.B.); (M.N.-J.); (M.P.); (F.C.-M.)
| | - Lidia Sanchez
- Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, 08035 Barcelona, Spain; (G.S.); (L.A.); (L.S.); (R.F.)
| | - Roberta Fasani
- Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, 08035 Barcelona, Spain; (G.S.); (L.A.); (L.S.); (R.F.)
| | - Roger Paredes
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Barcelona, Spain; (A.B.); (M.N.-J.); (M.P.); (F.C.-M.)
- Faculty of Medicine, University of Vic–Central University of Catalonia (UVic–UCC), 08500 Vic, Barcelona, Spain
- Facultat de Medicina, Universitat Autonoma de Barcelona (UAB), 08193 Barcelona, Spain
- Fight AIDS Foundation, Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Barcelona, Spain
- Infectious Diseases Service, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Barcelona, Spain
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
- Correspondence: (R.P.); (P.N.)
| | - Paolo Nuciforo
- Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, 08035 Barcelona, Spain; (G.S.); (L.A.); (L.S.); (R.F.)
- Correspondence: (R.P.); (P.N.)
| |
Collapse
|
6
|
Chong IY, Starling N, Rust A, Alexander J, Aronson L, Llorca-Cardenosa M, Chauhan R, Chaudry A, Kumar S, Fenwick K, Assiotis I, Matthews N, Begum R, Wotherspoon A, Terlizzo M, Watkins D, Chau I, Lord CJ, Haider S, Rao S, Cunningham D. The Mutational Concordance of Fixed Formalin Paraffin Embedded and Fresh Frozen Gastro-Oesophageal Tumours Using Whole Exome Sequencing. J Clin Med 2021; 10:E215. [PMID: 33435284 PMCID: PMC7826535 DOI: 10.3390/jcm10020215] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/01/2021] [Accepted: 01/04/2021] [Indexed: 12/31/2022] Open
Abstract
1. BACKGROUND The application of massively parallel sequencing has led to the identification of aberrant druggable pathways and somatic mutations within therapeutically relevant genes in gastro-oesophageal cancer. Given the widespread use of formalin-fixed paraffin-embedded (FFPE) samples in the study of this disease, it would be beneficial, especially for the purposes of biomarker evaluation, to assess the concordance between comprehensive exome-wide sequencing data from archival FFPE samples originating from a prospective clinical study and those derived from fresh-frozen material. 2. METHODS We analysed whole-exome sequencing data to define the mutational concordance of 16 matched fresh-frozen and FFPE gastro-oesophageal tumours (N = 32) from a prospective clinical study. We assessed DNA integrity prior to sequencing and then identified coding mutations in genes that have previously been implicated in other cancers. In addition, we calculated the mutant-allele heterogeneity (MATH) for these samples. 3. RESULTS Although there was increased degradation of DNA in FFPE samples compared with frozen samples, sequencing data from only two FFPE samples failed to reach an adequate mapping quality threshold. Using a filtering threshold of mutant read counts of at least ten and a minimum of 5% variant allele frequency (VAF) we found that there was a high median mutational concordance of 97% (range 80.1-98.68%) between fresh-frozen and FFPE gastro-oesophageal tumour-derived exomes. However, the majority of FFPE tumours had higher mutant-allele heterogeneity (MATH) scores when compared with corresponding frozen tumours (p < 0.001), suggesting that FFPE-based exome sequencing is likely to over-represent tumour heterogeneity in FFPE samples compared to fresh-frozen samples. Furthermore, we identified coding mutations in 120 cancer-related genes, including those associated with chromatin remodelling and Wnt/β-catenin and Receptor Tyrosine Kinase signalling. 4. CONCLUSIONS These data suggest that comprehensive genomic data can be generated from exome sequencing of selected DNA samples extracted from archival FFPE gastro-oesophageal tumour tissues within the context of prospective clinical trials.
Collapse
Affiliation(s)
- Irene Y. Chong
- The Division of Molecular Pathology, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK; (L.A.); (M.L.-C.)
- The Royal Marsden Hospital NHS Foundation Trust, 203 Fulham Road, London SW3 6JJ, UK; (N.S.); (A.C.); (S.K.); (R.B.); (A.W.); (M.T.); (D.W.); (I.C.); (S.R.); (D.C.)
| | - Naureen Starling
- The Royal Marsden Hospital NHS Foundation Trust, 203 Fulham Road, London SW3 6JJ, UK; (N.S.); (A.C.); (S.K.); (R.B.); (A.W.); (M.T.); (D.W.); (I.C.); (S.R.); (D.C.)
| | - Alistair Rust
- The Tissue Profiling Unit, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK; (A.R.); (R.C.); (K.F.); (I.A.); (N.M.)
| | - John Alexander
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; (J.A.); (C.J.L.); (S.H.)
| | - Lauren Aronson
- The Division of Molecular Pathology, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK; (L.A.); (M.L.-C.)
| | - Marta Llorca-Cardenosa
- The Division of Molecular Pathology, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK; (L.A.); (M.L.-C.)
| | - Ritika Chauhan
- The Tissue Profiling Unit, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK; (A.R.); (R.C.); (K.F.); (I.A.); (N.M.)
| | - Asif Chaudry
- The Royal Marsden Hospital NHS Foundation Trust, 203 Fulham Road, London SW3 6JJ, UK; (N.S.); (A.C.); (S.K.); (R.B.); (A.W.); (M.T.); (D.W.); (I.C.); (S.R.); (D.C.)
| | - Sacheen Kumar
- The Royal Marsden Hospital NHS Foundation Trust, 203 Fulham Road, London SW3 6JJ, UK; (N.S.); (A.C.); (S.K.); (R.B.); (A.W.); (M.T.); (D.W.); (I.C.); (S.R.); (D.C.)
| | - Kerry Fenwick
- The Tissue Profiling Unit, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK; (A.R.); (R.C.); (K.F.); (I.A.); (N.M.)
| | - Ioannis Assiotis
- The Tissue Profiling Unit, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK; (A.R.); (R.C.); (K.F.); (I.A.); (N.M.)
| | - Nik Matthews
- The Tissue Profiling Unit, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK; (A.R.); (R.C.); (K.F.); (I.A.); (N.M.)
| | - Ruwaida Begum
- The Royal Marsden Hospital NHS Foundation Trust, 203 Fulham Road, London SW3 6JJ, UK; (N.S.); (A.C.); (S.K.); (R.B.); (A.W.); (M.T.); (D.W.); (I.C.); (S.R.); (D.C.)
| | - Andrew Wotherspoon
- The Royal Marsden Hospital NHS Foundation Trust, 203 Fulham Road, London SW3 6JJ, UK; (N.S.); (A.C.); (S.K.); (R.B.); (A.W.); (M.T.); (D.W.); (I.C.); (S.R.); (D.C.)
| | - Monica Terlizzo
- The Royal Marsden Hospital NHS Foundation Trust, 203 Fulham Road, London SW3 6JJ, UK; (N.S.); (A.C.); (S.K.); (R.B.); (A.W.); (M.T.); (D.W.); (I.C.); (S.R.); (D.C.)
| | - David Watkins
- The Royal Marsden Hospital NHS Foundation Trust, 203 Fulham Road, London SW3 6JJ, UK; (N.S.); (A.C.); (S.K.); (R.B.); (A.W.); (M.T.); (D.W.); (I.C.); (S.R.); (D.C.)
| | - Ian Chau
- The Royal Marsden Hospital NHS Foundation Trust, 203 Fulham Road, London SW3 6JJ, UK; (N.S.); (A.C.); (S.K.); (R.B.); (A.W.); (M.T.); (D.W.); (I.C.); (S.R.); (D.C.)
| | - Christopher J. Lord
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; (J.A.); (C.J.L.); (S.H.)
| | - Syed Haider
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; (J.A.); (C.J.L.); (S.H.)
| | - Sheela Rao
- The Royal Marsden Hospital NHS Foundation Trust, 203 Fulham Road, London SW3 6JJ, UK; (N.S.); (A.C.); (S.K.); (R.B.); (A.W.); (M.T.); (D.W.); (I.C.); (S.R.); (D.C.)
| | - David Cunningham
- The Royal Marsden Hospital NHS Foundation Trust, 203 Fulham Road, London SW3 6JJ, UK; (N.S.); (A.C.); (S.K.); (R.B.); (A.W.); (M.T.); (D.W.); (I.C.); (S.R.); (D.C.)
| |
Collapse
|
7
|
Mathieson W, Thomas GA. Why Formalin-fixed, Paraffin-embedded Biospecimens Must Be Used in Genomic Medicine: An Evidence-based Review and Conclusion. J Histochem Cytochem 2020; 68:543-552. [PMID: 32697619 PMCID: PMC7400666 DOI: 10.1369/0022155420945050] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Fresh-frozen tissue is the “gold standard” biospecimen type for next-generation sequencing (NGS). However, collecting frozen tissue is usually not feasible because clinical workflows deliver formalin-fixed, paraffin-embedded (FFPE) tissue blocks. Some clinicians and researchers are reticent to embrace the use of FFPE tissue for NGS because FFPE tissue can yield low quantities of degraded DNA, containing formalin-induced mutations. We describe the process by which formalin-induced deamination can lead to artifactual cytosine (C) to thymine (T) and guanine (G) to adenine (A) (C:G > T:A) mutation calls and perform a literature review of 17 publications that compare NGS data from patient-matched fresh-frozen and FFPE tissue blocks. We conclude that although it is indeed true that sequencing data from FFPE tissue can be poorer than those from frozen tissue, any differences occur at an inconsequential magnitude, and FFPE biospecimens can be used in genomic medicine with confidence:
Collapse
|
8
|
Dunn T, Berry G, Emig-Agius D, Jiang Y, Lei S, Iyer A, Udar N, Chuang HY, Hegarty J, Dickover M, Klotzle B, Robbins J, Bibikova M, Peeters M, Strömberg M. Pisces: an accurate and versatile variant caller for somatic and germline next-generation sequencing data. Bioinformatics 2020; 35:1579-1581. [PMID: 30304370 PMCID: PMC6499249 DOI: 10.1093/bioinformatics/bty849] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 09/28/2018] [Accepted: 10/08/2018] [Indexed: 01/01/2023] Open
Abstract
Motivation Next-generation sequencing technology is transitioning quickly from research labs to clinical settings. The diagnosis and treatment selection for many acquired and autosomal conditions necessitate a method for accurately detecting somatic and germline variants. Results We have developed Pisces, a rapid, versatile and accurate small-variant calling suite designed for somatic and germline amplicon sequencing applications. Accuracy is achieved by four distinct modules, each incorporating a number of novel algorithmic strategies. Availability and implementation Pisces is distributed under an open source license and can be downloaded from https://github.com/Illumina/Pisces. Pisces is available on the BaseSpace™ SequenceHub. It is distributed on Illumina sequencing platforms such as the MiSeq™ and is included in the Praxis™ Extended RAS Panel test which was recently approved by the FDA. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Tamsen Dunn
- Departments of Bioinformatics and Clinical Genomics, Illumina Inc., San Diego, CA, USA
| | - Gwenn Berry
- Departments of Bioinformatics and Clinical Genomics, Illumina Inc., San Diego, CA, USA
| | - Dorothea Emig-Agius
- Departments of Bioinformatics and Clinical Genomics, Illumina Inc., San Diego, CA, USA
| | - Yu Jiang
- Departments of Bioinformatics and Clinical Genomics, Illumina Inc., San Diego, CA, USA
| | - Serena Lei
- Departments of Bioinformatics and Clinical Genomics, Illumina Inc., San Diego, CA, USA
| | - Anita Iyer
- Departments of Bioinformatics and Clinical Genomics, Illumina Inc., San Diego, CA, USA
| | - Nitin Udar
- Departments of Bioinformatics and Clinical Genomics, Illumina Inc., San Diego, CA, USA
| | - Han-Yu Chuang
- Departments of Bioinformatics and Clinical Genomics, Illumina Inc., San Diego, CA, USA
| | - Jeff Hegarty
- Departments of Bioinformatics and Clinical Genomics, Illumina Inc., San Diego, CA, USA
| | - Michael Dickover
- Departments of Bioinformatics and Clinical Genomics, Illumina Inc., San Diego, CA, USA
| | - Brandy Klotzle
- Departments of Bioinformatics and Clinical Genomics, Illumina Inc., San Diego, CA, USA
| | - Justin Robbins
- Departments of Bioinformatics and Clinical Genomics, Illumina Inc., San Diego, CA, USA
| | - Marina Bibikova
- Departments of Bioinformatics and Clinical Genomics, Illumina Inc., San Diego, CA, USA
| | - Marc Peeters
- Department of Oncology, Antwerp University Hospital, Edegem, Belgium
| | - Michael Strömberg
- Departments of Bioinformatics and Clinical Genomics, Illumina Inc., San Diego, CA, USA
| |
Collapse
|
9
|
Extended Molecular Profiling Improves Stratification and Prediction of Survival After Resection of Colorectal Liver Metastases. Ann Surg 2020; 270:799-805. [PMID: 31634180 DOI: 10.1097/sla.0000000000003527] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The aim of this study was to assess the effect of cancer-related genes and their mutations analyzed by next-generation sequencing (NGS) on the oncological outcome after resection of colorectal liver metastases (CRLM). BACKGROUND Traditional prognostic scores include clinical and pathological parameters of primary tumor and metastases. The modified clinical risk score (m-CS), based on size of metastases, primary tumor nodal status, and RAS mutation status outperformed traditional scores. We hypothesized to further improve the scoring system based on the results of NGS. METHODS Cancer tissues of 139 patients with CRLM were used for NGS. The work-up included the analysis of recurrent somatic mutations and copy number changes of 720 genes. Clinical data were extracted from a prospectively collected institutional liver database. RESULTS Depending on significance, the following cancer-related genes and their alterations (%) were further investigated: APC (86%), TP53 (78%), KRAS (29%), SMAD4 (15%), PIK3CA (14%), BRAF (8%), ERBB2 (6%), SMAD3 (5%), SMAD2 (4%), and NRAS (4%). The most predictive parameters for poor oncological outcome were alterations in the SMAD family (P = 0.0186) and RAS-RAF pathway (P = 0.032). Refining the m-CS by replacing RAS with RAS-RAF pathway and adding SMAD family resulted in an extended clinical risk score which is highly predictive for oncological outcome (P < 0.0001). CONCLUSION In conclusion, mutations of the SMAD family revealed a strong prognostic effect after surgery for CRLM. Integration of alterations of the SMAD family as well as the RAS/RAF pathway resulted in a new, still simple but highly prognostic score.
Collapse
|
10
|
Gao XH, Li J, Gong HF, Yu GY, Liu P, Hao LQ, Liu LJ, Bai CG, Zhang W. Comparison of Fresh Frozen Tissue With Formalin-Fixed Paraffin-Embedded Tissue for Mutation Analysis Using a Multi-Gene Panel in Patients With Colorectal Cancer. Front Oncol 2020; 10:310. [PMID: 32232001 PMCID: PMC7083147 DOI: 10.3389/fonc.2020.00310] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 02/21/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Next generation sequencing (NGS)-based multi-gene panel tests have been performed to predict the treatment response and prognosis in patients with colorectal cancer (CRC). Whether the multi-gene mutation results of formalin-fixed paraffin-embedded (FFPE) tissues are identical to those of fresh frozen tissues remains unknown. Methods: A 22-gene panel with 103 hotspots was used to detect mutations in paired fresh frozen tissue and FFPE tissue from 118 patients with CRC. Results: In our study, 117 patients (99.2%) had one or more variants, with 226 variants in FFPE tissue and 221 in fresh frozen tissue. Of the 129 variants identified in this study, 96 variants were present in both FFPE and fresh frozen tissues; 27 variants were found in FFPE tissues only; 6 variants were found only in fresh frozen tissues. The mutation results demonstrated >94.0% concordance in all variants, with Kappa coefficient >0.500 in 64.3% (83/129) of variants. At the gene level, concordance ranged from 73.8 to 100.0%, with Kappa coefficient >0.500 in 81.3% (13/16) of genes. Conclusions: The results of mutation analysis performed with a multi-gene panel and FFPE and fresh frozen tissue were highly concordant in patients with CRC, at both the variant and gene levels. There were, however, some important differences in mutation results between the two tissue types. Therefore, fresh frozen tissue should not routinely be replaced with FFPE tissue for mutation analysis with a multi-gene panel. Rather, FFPE tissue is a reasonable alternative for fresh frozen tissue when the latter is unavailable.
Collapse
Affiliation(s)
- Xian Hua Gao
- Department of Colorectal Surgery, Changhai Hospital, Shanghai, China
| | - Juan Li
- Department of Nephrology, Changhai Hospital, Shanghai, China
| | - Hai Feng Gong
- Department of Colorectal Surgery, Changhai Hospital, Shanghai, China
| | - Guan Yu Yu
- Department of Colorectal Surgery, Changhai Hospital, Shanghai, China
| | - Peng Liu
- Department of Colorectal Surgery, Changhai Hospital, Shanghai, China
| | - Li Qiang Hao
- Department of Colorectal Surgery, Changhai Hospital, Shanghai, China
| | - Lian Jie Liu
- Department of Colorectal Surgery, Changhai Hospital, Shanghai, China
| | - Chen Guang Bai
- Department of Pathology, Changhai Hospital, Shanghai, China
| | - Wei Zhang
- Department of Colorectal Surgery, Changhai Hospital, Shanghai, China
| |
Collapse
|
11
|
Systematic comparison of somatic variant calling performance among different sequencing depth and mutation frequency. Sci Rep 2020; 10:3501. [PMID: 32103116 PMCID: PMC7044309 DOI: 10.1038/s41598-020-60559-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 01/29/2020] [Indexed: 12/09/2022] Open
Abstract
In the past decade, treatments for tumors have made remarkable progress, such as the successful clinical application of targeted therapies. Nowadays, targeted therapies are based primarily on the detection of mutations, and next-generation sequencing (NGS) plays an important role in relevant clinical research. The mutation frequency is a major problem in tumor mutation detection and increasing sequencing depth is a widely used method to improve mutation calling performance. Therefore, it is necessary to evaluate the effect of different sequencing depth and mutation frequency as well as mutation calling tools. In this study, Strelka2 and Mutect2 tools were used in detecting the performance of 30 combinations of sequencing depth and mutation frequency. Results showed that the precision rate kept greater than 95% in most of the samples. Generally, for higher mutation frequency (≥20%), sequencing depth ≥200X is sufficient for calling 95% mutations; for lower mutation frequency (≤10%), we recommend improving experimental method rather than increasing sequencing depth. Besides, according to our results, although Strelka2 and Mutect2 performed similarly, the former performed slightly better than the latter one at higher mutation frequency (≥20%), while Mutect2 performed better when the mutation frequency was lower than 10%. Besides, Strelka2 was 17 to 22 times faster than Mutect2 on average. Our research will provide a useful and comprehensive guideline for clinical genomic researches on somatic mutation identification through systematic performance comparison among different sequencing depths and mutation frequency.
Collapse
|
12
|
Whole mitochondrial genome sequencing highlights mitochondrial impact in gastric cancer. Sci Rep 2019; 9:15716. [PMID: 31673122 PMCID: PMC6823544 DOI: 10.1038/s41598-019-51951-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 10/08/2019] [Indexed: 02/07/2023] Open
Abstract
Mitochondria are organelles that perform major roles in cellular operation. Thus, alterations in mitochondrial genome (mtGenome) may lead to mitochondrial dysfunction and cellular deregulation, influencing carcinogenesis. Gastric cancer (GC) is one of the most incident and mortal types of cancer in Brazil, particularly in the Amazon region. Here, we sequenced and compared the whole mtGenome extracted from FFPE tissue samples of GC patients (tumor and internal control – IC) and cancer-free individuals (external control – EC) from this region. We found 3-fold more variants and up to 9-fold more heteroplasmic regions in tumor when compared to paired IC samples. Moreover, tumor presented more heteroplasmic variants when compared to EC, while IC and EC showed no significant difference when compared to each other. Tumor also presented substantially more variants in the following regions: MT-RNR1, MT-ND5, MT-ND4, MT-ND2, MT-DLOOP1 and MT-CO1. In addition, our haplogroup results indicate an association of Native American ancestry (particularly haplogroup C) to gastric cancer development. To the best of our knowledge, this is the first study to sequence the whole mtGenome from FFPE samples and to apply mtGenome analysis in association to GC in Brazil.
Collapse
|
13
|
Zhang X, Shao Y, Tian J, Liao Y, Li P, Zhang Y, Chen J, Li Z. pTrimmer: An efficient tool to trim primers of multiplex deep sequencing data. BMC Bioinformatics 2019; 20:236. [PMID: 31077131 PMCID: PMC6511130 DOI: 10.1186/s12859-019-2854-x] [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: 08/09/2018] [Accepted: 04/26/2019] [Indexed: 11/23/2022] Open
Abstract
Background With the widespread use of multiple amplicon-sequencing (MAS) in genetic variation detection, an efficient tool is required to remove primer sequences from short reads to ensure the reliability of downstream analysis. Although some tools are currently available, their efficiency and accuracy require improvement in trimming large scale of primers in high throughput target genome sequencing. This issue is becoming more urgent considering the potential clinical implementation of MAS for processing patient samples. We here developed pTrimmer that could handle thousands of primers simultaneously with greatly improved accuracy and performance. Result pTrimmer combines the two algorithms of k-mers and Needleman-Wunsch algorithm, which ensures its accuracy even with the presence of sequencing errors. pTrimmer has an improvement of 28.59% sensitivity and 11.87% accuracy compared to the similar tools. The simulation showed pTrimmer has an ultra-high sensitivity rate of 99.96% and accuracy of 97.38% compared to cutPrimers (70.85% sensitivity rate and 58.73% accuracy). And the performance of pTrimmer is notably higher. It is about 370 times faster than cutPrimers and even 17,000 times faster than cutadapt per threads. Trimming 2158 pairs of primers from 11 million reads (Illumina PE 150 bp) takes only 37 s and no more than 100 MB of memory consumption. Conclusions pTrimmer is designed to trim primer sequence from multiplex amplicon sequencing and target sequencing. It is highly sensitive and specific compared to other three similar tools, which could help users to get more reliable mutational information for downstream analysis. Electronic supplementary material The online version of this article (10.1186/s12859-019-2854-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaolong Zhang
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Yanyan Shao
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Jichao Tian
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Yuwei Liao
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Peiying Li
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Yu Zhang
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Jun Chen
- The Second Hospital of Dalian Medical University, 467th Zhongshan Road, Shahekou District, Dalian, 116023, Liaoning, China.
| | - Zhiguang Li
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, Liaoning, China. .,The Second Affiliated Hospital, School of Medicine, Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
14
|
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: 0.3] [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
|
15
|
Tian J, Geng Y, Lv D, Li P, Cordova M, Liao Y, Tian X, Zhang X, Zhang Q, Zou K, Zhang Y, Zhang X, Li Y, Zhang J, Ma Z, Shao Y, Song L, Owen GI, Li T, Liu R, Liu Q, Zou L, Zhang Z, Li Z. Using plasma cell-free DNA to monitor the chemoradiotherapy course of cervical cancer. Int J Cancer 2019; 145:2547-2557. [PMID: 30919951 DOI: 10.1002/ijc.32295] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 02/10/2019] [Accepted: 03/12/2019] [Indexed: 12/16/2022]
Abstract
The liquid biopsy is being integrated into cancer diagnostics and surveillance. However, critical questions still remain, such as how to precisely evaluate cancer mutation burden and interpret the corresponding clinical implications. Herein, we evaluated the role of peripheral blood cell-free DNA (cfDNA) in characterizing the dynamic mutation alterations of 48 cancer driver genes from cervical cancer patients. We performed targeted deep sequencing on 93 plasma cfDNA from 57 cervical cancer patients and from this developed an algorithm, allele fraction deviation (AFD), to monitor in an unbiased manner the dynamic changes of genomic aberrations. Differing treatments, including chemotherapy (n = 22), radiotherapy (n = 14) and surgery (n = 15), led to a significant decrease in AFD values (Wilcoxon, p = 0.029). The decrease of cfDNA AFD values was accompanied by shrinkage in the size of the tumor in most patients. However, in a subgroup of patients where cfDNA AFD values did not reflect a reduction in tumor size, there was a detection of progressive disease (metastasis). Furthermore, a low AFD value at diagnosis followed a later increase of AFD value also successfully predicted relapse. These results show that plasma cfDNA, together with targeted deep sequencing, may help predict treatment response and disease development in cervical cancer.
Collapse
Affiliation(s)
- Jichao Tian
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Yan Geng
- Department of Radiotherapy, Ansteel Group Hospital, Anshan, Liaoning, China
| | - Dekang Lv
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Peiying Li
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Miguel Cordova
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Yuwei Liao
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Xiaoyuan Tian
- The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xiaolong Zhang
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Qingzheng Zhang
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Kun Zou
- The first affiliated hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yu Zhang
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Xia Zhang
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Yulong Li
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Jian Zhang
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Zhaokui Ma
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Yanyan Shao
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Luyao Song
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Gareth I Owen
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Tingting Li
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Ruimei Liu
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Quentin Liu
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Lijuan Zou
- The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Zhuo Zhang
- The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Zhiguang Li
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
16
|
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: 109] [Impact Index Per Article: 15.6] [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
|
17
|
Tack V, Spans L, Schuuring E, Keppens C, Zwaenepoel K, Pauwels P, Van Houdt J, Dequeker EMC. Describing the Reportable Range Is Important for Reliable Treatment Decisions: A Multiple Laboratory Study for Molecular Tumor Profiling Using Next-Generation Sequencing. J Mol Diagn 2018; 20:743-753. [PMID: 30055348 DOI: 10.1016/j.jmoldx.2018.06.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 05/14/2018] [Accepted: 06/05/2018] [Indexed: 01/04/2023] Open
Abstract
Because interpretation of next-generation sequencing (NGS) data remains challenging, optimization of the NGS process is needed to obtain correct sequencing results. Therefore, extensive validation and continuous monitoring of the quality is essential. NGS performance was compared with traditional detection methods and technical quality of nine NGS technologies was assessed. First, nine formalin-fixed, paraffin-embedded patient samples were analyzed by 114 laboratories by using different detection methods. No significant differences in performance were observed between analyses with NGS and traditional techniques. Second, two DNA control samples were analyzed for a selected number of variants by 26 participants with the use of nine different NGS technologies. Quality control metrics were analyzed from raw data files and a survey about routine procedures. Results showed large differences in coverages, but observed variant allele frequencies in raw data files were in line with predefined variant allele frequencies. Many false negative results were found because of low-quality regions, which were not reported as such. It is recommended to disclose the reportable range, the fraction of targeted genomic regions for which calls of acceptable quality can be generated, to avoid any errors in therapy decisions. NGS can be a reliable technique, only if essential quality control during analysis is applied and reported.
Collapse
Affiliation(s)
- Véronique Tack
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium
| | - Lien Spans
- Center for Human Genetics, University of Leuven, Leuven, Belgium
| | - Ed Schuuring
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Cleo Keppens
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium
| | - Karen Zwaenepoel
- Department of Pathology, University Hospital Antwerp, Edegem, Belgium
| | - Patrick Pauwels
- Center for Oncologic Research (CORE), University of Antwerp, Antwerp, Belgium
| | | | - Elisabeth M C Dequeker
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium; Department of Medical Diagnostics, University Hospital Leuven, Leuven, Belgium.
| |
Collapse
|
18
|
Bäumer C, Fisch E, Wedler H, Reinecke F, Korfhage C. Exploring DNA quality of single cells for genome analysis with simultaneous whole-genome amplification. Sci Rep 2018; 8:7476. [PMID: 29748573 PMCID: PMC5945709 DOI: 10.1038/s41598-018-25895-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 04/30/2018] [Indexed: 12/12/2022] Open
Abstract
Single cell genome analysis methods are powerful tools to define features of single cells and to identify differences between them. Since the DNA amount of a single cell is very limited, cellular DNA usually needs to be amplified by whole-genome amplification before being subjected to further analysis. A single nucleus only contains two haploid genomes. Thus, any DNA damage that prevents amplification results in loss of damaged DNA sites and induces an amplification bias. Therefore, the assessment of single cell DNA quality is urgently required. As of today, there is no simple method to determine the quality of a single cell DNA in a manner that will still retain the entire cellular DNA for amplification and downstream analysis. Here, we describe a method for whole-genome amplification with simultaneous quality control of single cell DNA by using a competitive spike-in DNA template.
Collapse
Affiliation(s)
- Christiane Bäumer
- QIAGEN GmbH, Department for Research & Foundation, QIAGEN-Strasse 1, 40724, Hilden, Germany
| | - Evelyn Fisch
- QIAGEN GmbH, Department for Research & Foundation, QIAGEN-Strasse 1, 40724, Hilden, Germany
| | - Holger Wedler
- QIAGEN GmbH, Department for NGS, PCR-Array and WGA-Service, QIAGEN-Strasse 1, 40724, Hilden, Germany
| | - Frank Reinecke
- QIAGEN GmbH, Department for Research & Foundation, QIAGEN-Strasse 1, 40724, Hilden, Germany
| | - Christian Korfhage
- QIAGEN GmbH, Department for Research & Foundation, QIAGEN-Strasse 1, 40724, Hilden, Germany.
| |
Collapse
|
19
|
Yamashita S, Chun YS, Kopetz SE, Vauthey JN. Biomarkers in colorectal liver metastases. Br J Surg 2018; 105:618-627. [PMID: 29579319 DOI: 10.1002/bjs.10834] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 01/05/2018] [Accepted: 01/11/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Despite a 5-year overall survival rate of 58 per cent after liver resection for colorectal liver metastases (CLMs), more than half of patients develop recurrence, highlighting the need for accurate risk stratification and prognostication. Traditional prognostic factors have been superseded by newer outcome predictors, including those defined by the molecular origin of the primary tumour. METHODS This review synthesized findings in the literature using the PubMed database of articles in the English language published between 1998 and 2017 on prognostic and predictive biomarkers in patients undergoing resection of CLMs. RESULTS Responses to preoperative chemotherapy define prognosis in patients undergoing CLM resection. There are differences by embryological origin too. Somatic mutations in the proto-oncogenes KRAS and NRAS are associated with positive surgical margins and tumour regrowth after ablation. Other mutations (such as BRAF) and co-occurring mutations in RAS/TP53 and APC/PIK3CA have emerged as important biomarkers that determine an individual patient's tumour biology and may be used to predict outcome after CLM resection. CONCLUSION Knowledge of somatic mutations can guide the use of preoperative therapy, extent of surgical margin and selection for ablation alone.
Collapse
Affiliation(s)
- S Yamashita
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Y S Chun
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - S E Kopetz
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - J-N Vauthey
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
20
|
Kechin A, Boyarskikh U, Kel A, Filipenko M. cutPrimers: A New Tool for Accurate Cutting of Primers from Reads of Targeted Next Generation Sequencing. J Comput Biol 2017; 24:1138-1143. [PMID: 28715235 DOI: 10.1089/cmb.2017.0096] [Citation(s) in RCA: 732] [Impact Index Per Article: 91.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cutting of primers from reads is an important step of processing targeted amplicon-based next generation sequencing data. Existing tools are adapted for cutting of one or several primer/adapter sequences from reads and removing all of their occurrences. Also most of the existing tools use kmers and may cut only part of primers or primers with studied sequence of gene. Because of this, use of such programs leads to incorrect trimming, reduction of coverage, and increase in the number of false-positive and/or false-negative results. We have developed a new tool named cutPrimers for accurate cutting of any number of primers from reads. Using sequencing reads that were obtained during study of BRCA1/2 genes, we compared it with cutadapt, AlienTrimmer, and BBDuk. All of them trimmed reads in such a way that coverage of at least two amplicons decreased to unacceptable level (<30 reads) comparing with reads trimmed with cutPrimers. At the same time, Trimmomatic and AlienTrimmer cut all occurrences of primer sequences, so the length of the remaining reads was less than prospective.
Collapse
Affiliation(s)
- Andrey Kechin
- 1 Institute of Chemical Biology and Fundamental Medicine SB RAS , Novosibirsk, Russia .,2 Novosivirsk State University , Novosibirsk, Russia
| | - Uljana Boyarskikh
- 1 Institute of Chemical Biology and Fundamental Medicine SB RAS , Novosibirsk, Russia
| | - Alexander Kel
- 1 Institute of Chemical Biology and Fundamental Medicine SB RAS , Novosibirsk, Russia .,3 geneXplain GmbH, Wolfenbuettel, Germany .,4 biosoft.ru , Novosibirsk, Russia
| | - Maxim Filipenko
- 1 Institute of Chemical Biology and Fundamental Medicine SB RAS , Novosibirsk, Russia
| |
Collapse
|
21
|
Ofner R, Ritter C, Ugurel S, Cerroni L, Stiller M, Bogenrieder T, Solca F, Schrama D, Becker JC. Non-reproducible sequence artifacts in FFPE tissue: an experience report. J Cancer Res Clin Oncol 2017; 143:1199-1207. [PMID: 28314930 DOI: 10.1007/s00432-017-2399-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 03/14/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND Recent advances in sequencing technologies supported the development of molecularly targeted therapy in cancer patients. Thus, genomic analyses are becoming a routine part in clinical practice and accurate detection of actionable mutations is essential to assist diagnosis and therapy choice. However, this is often challenging due to major problems associated with DNA from formalin-fixed paraffin-embedded tissue which is usually the primary source for genetic testing. OBJECTIVES Here we want to share our experience regarding major problems associated with FFPE DNA used for PCR-based sequencing as illustrated by the mutational analysis of ERBB4 in melanoma. We want to focus on two major problems including extensive DNA fragmentation and hydrolytic deamination as source of non-reproducible sequence artifacts. Further, we provide potential explanations and possible strategies to minimize these difficulties and improve the detection of targetable mutations. METHODS Genomic DNA from formalin-fixed paraffin-embedded tumor samples was isolated followed by PCR amplification, Sanger sequencing and statistical analysis. RESULTS Analysis of Sanger sequencing data revealed a total of 46 ERBB4 mutations in 27 of 96 samples including the identification of 11 mutations at three previously unknown mutational hotspots. Unfortunately, we were not able to confirm any assumed hotspot mutation within repeated sequencing of relevant amplicons suggesting the detection of sequence artifacts most likely caused by DNA lesions associated with FFPE tissues. CONCLUSION Since DNA from FFPE tissue is usually the primary source for mutational analyses, appropriate measures must be implemented in the workflow to assess DNA damage in formalin-fixed tissue to ensure accurate detection of actionable mutations and minimize the occurrence of sequence artifacts.
Collapse
Affiliation(s)
- Richard Ofner
- Department of General Dermatology, Medical University Graz, Graz, Austria
| | - Cathrin Ritter
- Department of General Dermatology, Medical University Graz, Graz, Austria
- Translational Skin Cancer Research-TSCR, German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, German Cancer Research Center (DKFZ), University Hospital Essen, Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital of Essen, Essen, Germany
| | - Lorenzo Cerroni
- Department of General Dermatology, Medical University Graz, Graz, Austria
| | - Mathias Stiller
- Translational Skin Cancer Research-TSCR, German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, German Cancer Research Center (DKFZ), University Hospital Essen, Essen, Germany
| | | | | | - David Schrama
- Department of Dermatology, University Hospital of Würzburg, Würzburg, Germany
| | - Jürgen C Becker
- Department of General Dermatology, Medical University Graz, Graz, Austria.
- Translational Skin Cancer Research-TSCR, German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, German Cancer Research Center (DKFZ), University Hospital Essen, Essen, Germany.
- Department of Dermatology, University Hospital of Essen, Essen, Germany.
| |
Collapse
|
22
|
Influence of mucinous and necrotic tissue in colorectal cancer samples on KRAS mutation analysis. Pathol Res Pract 2017; 213:606-611. [DOI: 10.1016/j.prp.2017.04.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/28/2017] [Accepted: 04/28/2017] [Indexed: 12/21/2022]
|
23
|
Darwanto A, Hein AM, Strauss S, Kong Y, Sheridan A, Richards D, Lader E, Ngowe M, Pelletier T, Adams D, Ricker A, Patel N, Kühne A, Hughes S, Shiffman D, Zimmermann D, Te Kaat K, Rothmann T. Use of the QIAGEN GeneReader NGS system for detection of KRAS mutations, validated by the QIAGEN Therascreen PCR kit and alternative NGS platform. BMC Cancer 2017; 17:358. [PMID: 28532404 PMCID: PMC5441096 DOI: 10.1186/s12885-017-3328-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 05/05/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The detection of somatic mutations in primary tumors is critical for the understanding of cancer evolution and targeting therapy. Multiple technologies have been developed to enable the detection of such mutations. Next generation sequencing (NGS) is a new platform that is gradually becoming the technology of choice for genotyping cancer samples, owing to its ability to simultaneously interrogate many genomic loci at massively high efficiency and increasingly lower cost. However, multiple barriers still exist for its broader adoption in clinical research practice, such as fragmented workflow and complex bioinformatics analysis and interpretation. METHODS We performed validation of the QIAGEN GeneReader NGS System using the QIAact Actionable Insights Tumor Panel, focusing on clinically meaningful mutations by using DNA extracted from formalin-fixed paraffin-embedded (FFPE) colorectal tissue with known KRAS mutations. The performance of the GeneReader was evaluated and compared to data generated from alternative technologies (PCR and pyrosequencing) as well as an alternative NGS platform. The results were further confirmed with Sanger sequencing. RESULTS The data generated from the GeneReader achieved 100% concordance with reference technologies. Furthermore, the GeneReader workflow provides a truly integrated workflow, eliminating artifacts resulting from routine sample preparation; and providing up-to-date interpretation of test results. CONCLUSION The GeneReader NGS system offers an effective and efficient method to identify somatic (KRAS) cancer mutations.
Collapse
Affiliation(s)
- Agus Darwanto
- QIAGEN Waltham, 35 Gatehouse Dr, Waltham, MA, 02451, USA.,Novartis Institutes for BioMedical Research, Cambridge, MA, 02139, USA
| | | | - Sascha Strauss
- QIAGEN GmbH, QIAGEN Strasse 1, 40724, Hilden, Nordrhein-Westfalen, Germany
| | - Yi Kong
- QIAGEN Redwood City, 1700 Seaport Blvd, Redwood, CA, 94063, USA
| | | | - Dan Richards
- QIAGEN Redwood City, 1700 Seaport Blvd, Redwood, CA, 94063, USA
| | - Eric Lader
- QIAGEN Frederick, 6951 Executive Way, Frederick, MD, 21703, USA
| | - Monika Ngowe
- QIAGEN Waltham, 35 Gatehouse Dr, Waltham, MA, 02451, USA.,T2 Biosystems, Lexington, MA, 02421, USA
| | | | - Danielle Adams
- QIAGEN Waltham, 35 Gatehouse Dr, Waltham, MA, 02451, USA.,Macherey-Nigel, Bethlehem, PA, 18020, USA
| | - Austin Ricker
- QIAGEN Waltham, 35 Gatehouse Dr, Waltham, MA, 02451, USA
| | - Nishit Patel
- QIAGEN Waltham, 35 Gatehouse Dr, Waltham, MA, 02451, USA
| | - Andreas Kühne
- QIAGEN GmbH, QIAGEN Strasse 1, 40724, Hilden, Nordrhein-Westfalen, Germany
| | - Simon Hughes
- QIAGEN Manchester, Skelton House Lloyd Street North, Manchester, M15 6SH, UK
| | - Dan Shiffman
- QIAGEN Redwood City, 1700 Seaport Blvd, Redwood, CA, 94063, USA
| | - Dirk Zimmermann
- QIAGEN GmbH, QIAGEN Strasse 1, 40724, Hilden, Nordrhein-Westfalen, Germany
| | - Kai Te Kaat
- QIAGEN GmbH, QIAGEN Strasse 1, 40724, Hilden, Nordrhein-Westfalen, Germany
| | - Thomas Rothmann
- QIAGEN GmbH, QIAGEN Strasse 1, 40724, Hilden, Nordrhein-Westfalen, Germany.
| |
Collapse
|
24
|
Einaga N, Yoshida A, Noda H, Suemitsu M, Nakayama Y, Sakurada A, Kawaji Y, Yamaguchi H, Sasaki Y, Tokino T, Esumi M. Assessment of the quality of DNA from various formalin-fixed paraffin-embedded (FFPE) tissues and the use of this DNA for next-generation sequencing (NGS) with no artifactual mutation. PLoS One 2017; 12:e0176280. [PMID: 28498833 PMCID: PMC5428915 DOI: 10.1371/journal.pone.0176280] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 04/07/2017] [Indexed: 01/09/2023] Open
Abstract
Formalin-fixed, paraffin-embedded (FFPE) tissues used for pathological diagnosis are valuable for studying cancer genomics. In particular, laser-capture microdissection of target cells determined by histopathology combined with FFPE tissue section immunohistochemistry (IHC) enables precise analysis by next-generation sequencing (NGS) of the genetic events occurring in cancer. The result is a new strategy for a pathological tool for cancer diagnosis: 'microgenomics'. To more conveniently and precisely perform microgenomics, we revealed by systematic analysis the following three details regarding FFPE DNA compared with paired frozen tissue DNA. 1) The best quality of FFPE DNA is obtained by tissue fixation with 10% neutral buffered formalin for 1 day and heat treatment of tissue lysates at 95°C for 30 minutes. 2) IHC staining of FFPE tissues decreases the quantity and quality of FFPE DNA to one-fourth, and antigen retrieval (at 120°C for 15 minutes, pH 6.0) is the major reason for this decrease. 3) FFPE DNA prepared as described herein is sufficient for NGS. For non-mutated tissue specimens, no artifactual mutation occurs during FFPE preparation, as shown by precise comparison of NGS of FFPE DNA and paired frozen tissue DNA followed by validation. These results demonstrate that even FFPE tissues used for routine clinical diagnosis can be utilized to obtain reliable NGS data if appropriate conditions of fixation and validation are applied.
Collapse
Affiliation(s)
- Naoki Einaga
- Department of Pathology, Nihon University School of Medicine, Itabashi-ku, Tokyo, Japan
| | - Akio Yoshida
- Department of Pathology, Nihon University School of Medicine, Itabashi-ku, Tokyo, Japan
- Department of Orthopaedic Surgery, Nihon University School of Medicine, Itabashi-ku, Tokyo, Japan
| | - Hiroko Noda
- Department of Pathology, Nihon University School of Medicine, Itabashi-ku, Tokyo, Japan
| | - Masaaki Suemitsu
- Department of Pathology, Nihon University School of Medicine, Itabashi-ku, Tokyo, Japan
- Department of Oral Pathology, Nihon University School of Dentistry at Matsudo, Matsudo, Chiba, Japan
| | - Yuki Nakayama
- Department of Pathology, Nihon University School of Medicine, Itabashi-ku, Tokyo, Japan
| | - Akihisa Sakurada
- Department of Pathology, Nihon University School of Medicine, Itabashi-ku, Tokyo, Japan
| | - Yoshiko Kawaji
- Department of Pathology, Nihon University School of Medicine, Itabashi-ku, Tokyo, Japan
| | - Hiromi Yamaguchi
- Department of Pathology, Nihon University School of Medicine, Itabashi-ku, Tokyo, Japan
| | - Yasushi Sasaki
- Department of Medical Genome Sciences, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Takashi Tokino
- Department of Medical Genome Sciences, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Mariko Esumi
- Department of Pathology, Nihon University School of Medicine, Itabashi-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
25
|
Ito C, Nishizuka SS, Ishida K, Uesugi N, Sugai T, Tamura G, Koeda K, Sasaki A. Analysis of PIK3CA mutations and PI3K pathway proteins in advanced gastric cancer. J Surg Res 2017; 212:195-204. [PMID: 28550907 DOI: 10.1016/j.jss.2017.01.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 01/09/2017] [Accepted: 01/19/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Although surgery and chemotherapy have extended advanced gastric cancer patient survival, some patients still experience relapse and metastasis. We postulated that PI3K pathway proteins could be prognostic biomarkers for the advanced gastric cancer patients. METHODS A retrospective cohort of 160 advanced gastric cancer patients receiving potentially curative surgery with/without chemotherapy was investigated for PIK3CA mutation and PI3K pathway protein level in the context of overall survival and relapse-free survival. RESULTS Thirteen patients (13 of 111, 11.7%) had PIK3CA mutations in codon 545, whereas one patient (1 of 94, 1.1%) had a mutation in PIK3CA codon 1047. PI3K pathway protein immunohistochemistry demonstrated that phosphorylated AKT positive [p-AKT (+)] patients in the surgery-only group had a good prognosis in terms of overall survival and relapse-free survival. No significant association between PIK3CA mutations and PI3K pathway protein level was seen. CONCLUSIONS This study revealed that (1) PIK3CA hotspot mutations occurred with low frequency in gastric cancer; (2) PIK3CA hotspot mutations were not directly associated with PI3K pathway activation; and (3) p-AKT (+) may be a biomarker for better outcomes for gastric cancer patients undergoing gastrectomy regardless of the PIK3CA mutation status.
Collapse
Affiliation(s)
- Chie Ito
- Molecular Therapeutics Laboratory, Iwate Medical University School of Medicine, Iwate, Japan; Department of Surgery, Iwate Medical University School of Medicine, Iwate, Japan
| | - Satoshi S Nishizuka
- Molecular Therapeutics Laboratory, Iwate Medical University School of Medicine, Iwate, Japan; Department of Surgery, Iwate Medical University School of Medicine, Iwate, Japan; Division of Biomedical Research and Development, Institute of Biomedical Science, Iwate Medical University, Iwate, Japan.
| | - Kazuyuki Ishida
- Department of Molecular Diagnostic Pathology, Iwate Medical University School of Medicine, Iwate, Japan
| | - Noriyuki Uesugi
- Department of Molecular Diagnostic Pathology, Iwate Medical University School of Medicine, Iwate, Japan
| | - Tamotsu Sugai
- Department of Molecular Diagnostic Pathology, Iwate Medical University School of Medicine, Iwate, Japan
| | - Gen Tamura
- Department of Pathology and Laboratory Medicine, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Keisuke Koeda
- Department of Surgery, Iwate Medical University School of Medicine, Iwate, Japan
| | - Akira Sasaki
- Department of Surgery, Iwate Medical University School of Medicine, Iwate, Japan
| |
Collapse
|
26
|
Kovaleva V, Geissler AL, Lutz L, Fritsch R, Makowiec F, Wiesemann S, Hopt UT, Passlick B, Werner M, Lassmann S. Spatio-temporal mutation profiles of case-matched colorectal carcinomas and their metastases reveal unique de novo mutations in metachronous lung metastases by targeted next generation sequencing. Mol Cancer 2016; 15:63. [PMID: 27756406 PMCID: PMC5069823 DOI: 10.1186/s12943-016-0549-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 10/06/2016] [Indexed: 12/13/2022] Open
Abstract
Background Targeted next generation sequencing (tNGS) has become part of molecular pathology diagnostics for determining RAS mutation status in colorectal cancer (CRC) patients as predictive tool for decision on EGFR-targeted therapy. Here, we investigated mutation profiles of case-matched tissue specimens throughout the disease course of CRC, to further specify RAS-status dynamics and to identify de novo mutations associated with distant metastases. Methods Case-matched formalin-fixed and paraffin-embedded (FFPE) resection specimens (n = 70; primary tumours, synchronous and/or metachronous liver and/or lung metastases) of 14 CRC cases were subjected to microdissection of normal colonic epithelial, primary and metastatic tumour cells, their DNA extraction and an adapted library protocol for limited DNA using the 48 gene TruSeq Amplicon Cancer PanelTM, MiSeq sequencing and data analyses (Illumina). Results By tNGS primary tumours were RAS wildtype in 5/14 and mutated in 9/14 (8/9 KRAS exon 2; 1/9 NRAS Exon 3) of cases. RAS mutation status was maintained in case-matched metastases throughout the disease course, albeit with altered allele frequencies. Case-matched analyses further identified a maximum of three sequence variants (mainly in APC, KRAS, NRAS, TP53) shared by all tumour specimens throughout the disease course per individual case. In addition, further case-matched de novo mutations were detected in synchronous and/or metachronous liver and/or lung metastases (e.g. in APC, ATM, FBXW7, FGFR3, GNAQ, KIT, PIK3CA, PTEN, SMAD4, SMO, STK11, TP53, VHL). Moreover, several de novo mutations were more frequent in synchronous (e.g. ATM, KIT, PIK3CA, SMAD4) or metachronous (e.g. FBXW7, SMO, STK11) lung metastases. Finally, some de novo mutations occurred only in metachronous lung metastases (CDKN2A, FGFR2, GNAS, JAK3, SRC). Conclusion Together, this study employs an adapted FFPE-based tNGS approach to confirm conservation of RAS mutation status in primary and metastatic tissue specimens of CRC patients. Moreover, it identifies genes preferentially mutated de novo in late disease stages of metachronous CRC lung metastases, several of which might be actionable by targeted therapies. Electronic supplementary material The online version of this article (doi:10.1186/s12943-016-0549-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Valentina Kovaleva
- Institute for Surgical Pathology, Medical Center-Faculty of Medicine, University of Freiburg, Breisacherstr. 115A, 79106, Freiburg, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anna-Lena Geissler
- Institute for Surgical Pathology, Medical Center-Faculty of Medicine, University of Freiburg, Breisacherstr. 115A, 79106, Freiburg, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Lisa Lutz
- Institute for Surgical Pathology, Medical Center-Faculty of Medicine, University of Freiburg, Breisacherstr. 115A, 79106, Freiburg, Germany
| | - Ralph Fritsch
- Department of Medicine I, Medical Center-Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Comprehensive Cancer Center Freiburg, Medical Center-Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Frank Makowiec
- Comprehensive Cancer Center Freiburg, Medical Center-Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of General and Visceral Surgery, Medical Center- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sebastian Wiesemann
- Comprehensive Cancer Center Freiburg, Medical Center-Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Thoracic Surgery, Medical Center-Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ulrich T Hopt
- Comprehensive Cancer Center Freiburg, Medical Center-Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of General and Visceral Surgery, Medical Center- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bernward Passlick
- Comprehensive Cancer Center Freiburg, Medical Center-Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Thoracic Surgery, Medical Center-Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martin Werner
- Institute for Surgical Pathology, Medical Center-Faculty of Medicine, University of Freiburg, Breisacherstr. 115A, 79106, Freiburg, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Comprehensive Cancer Center Freiburg, Medical Center-Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Silke Lassmann
- Institute for Surgical Pathology, Medical Center-Faculty of Medicine, University of Freiburg, Breisacherstr. 115A, 79106, Freiburg, Germany. .,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany. .,Comprehensive Cancer Center Freiburg, Medical Center-Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
27
|
Velizheva NP, Rechsteiner MP, Wong CE, Zhong Q, Rössle M, Bode B, Moch H, Soltermann A, Wild PJ, Tischler V. Cytology smears as excellent starting material for next-generation sequencing-based molecular testing of patients with adenocarcinoma of the lung. Cancer Cytopathol 2016; 125:30-40. [DOI: 10.1002/cncy.21771] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 07/08/2016] [Accepted: 07/27/2016] [Indexed: 11/10/2022]
Affiliation(s)
| | | | - Christine E. Wong
- Institute of Surgical Pathology; University Hospital Zurich; Zurich Switzerland
| | - Qing Zhong
- Institute of Surgical Pathology; University Hospital Zurich; Zurich Switzerland
| | - Matthias Rössle
- Cantonal Hospital Graubunden, Institute of Pathology; Chur Switzerland
| | - Beata Bode
- Institute of Surgical Pathology; University Hospital Zurich; Zurich Switzerland
| | - Holger Moch
- Institute of Surgical Pathology; University Hospital Zurich; Zurich Switzerland
| | - Alex Soltermann
- Institute of Surgical Pathology; University Hospital Zurich; Zurich Switzerland
| | - Peter J. Wild
- Institute of Surgical Pathology; University Hospital Zurich; Zurich Switzerland
| | - Verena Tischler
- Institute of Surgical Pathology; University Hospital Zurich; Zurich Switzerland
| |
Collapse
|
28
|
Mack E, Stabla K, Riera-Knorrenschild J, Moll R, Neubauer A, Brendel C. A rational two-step approach to KRAS mutation testing in colorectal cancer using high resolution melting analysis and pyrosequencing. BMC Cancer 2016; 16:585. [PMID: 27485514 PMCID: PMC4971616 DOI: 10.1186/s12885-016-2589-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 07/20/2016] [Indexed: 01/03/2023] Open
Abstract
Background KRAS mutation testing is mandatory in the management of metastatic colorectal cancer prior to treatment with anti-EGFR antibodies as patients whose tumors express mutant KRAS do not benefit from these agents. Although the U.S. Food and Drug Administration has recently approved two in-vitro diagnostics kits for determination of KRAS status, there is generally no consensus on the preferred method and new tests are continuously being developed. Most of these techniques focus on the hotspot mutations at codons 12 and 13 of the KRAS gene. Methods We describe a two-step approach to KRAS codon 12/13 mutation testing involving high resolution melting analysis (HRM) followed by pyrosequencing using the Therascreen KRAS Pyro kit (Qiagen) of only those samples that are not clearly identified as KRAS wildtype or mutant by HRM. First, we determined KRAS status in a panel of 61 colorectal cancer samples using both methods to compare technical performance and concordance of results. Subsequently, we evaluated practicability and costs of our concept in an independent set of 120 colorectal cancer samples in a routine diagnostic setting. Results HRM and pyrosequencing appeared to be equally sensitive, allowing for clear detection of mutant alleles at a mutant allele frequency ≥12.5 %. Pyrosequencing yielded more exploitable results due to lower input requirements and a lower rate of analysis failures. KRAS codon 12/13 status was called concordantly for 98.2 % (56/57) of all samples that could be successfully analysed by both methods and 100 % (19/19) of samples that were identified mutant by HRM. Reviewing the actual effort and expenses for KRAS mutation testing in our laboratory revealed, that the selective use of pyrosequencing for only those samples that could not be analysed by HRM increased the fraction of valid results from 87.5 % for HRM alone to 99.2 % (119/120) while allowing for a net reduction of operational costs of >75 % compared to pyrosequencing alone. Conclusions Combination of HRM and pyrosequencing in a two-step diagnostic procedure constitutes a reliable and economic analysis platform for KRAS mutation testing in colorectal cancer in a clinical setting. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2589-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elisabeth Mack
- Klinik für Hämatologie, Onkologie und Immunologie, Universitätsklinikum Gießen und Marburg, Standort Marburg, Philipps-Universität Marburg, Baldingerstraße, Marburg, Germany
| | - Kathleen Stabla
- Klinik für Hämatologie, Onkologie und Immunologie, Universitätsklinikum Gießen und Marburg, Standort Marburg, Philipps-Universität Marburg, Baldingerstraße, Marburg, Germany
| | - Jorge Riera-Knorrenschild
- Klinik für Hämatologie, Onkologie und Immunologie, Universitätsklinikum Gießen und Marburg, Standort Marburg, Philipps-Universität Marburg, Baldingerstraße, Marburg, Germany
| | - Roland Moll
- Institut für Pathologie, Universitätsklinikum Gießen und Marburg, Standort Marburg, Philipps-Universität Marburg, Baldingerstraße, Marburg, Germany
| | - Andreas Neubauer
- Klinik für Hämatologie, Onkologie und Immunologie, Universitätsklinikum Gießen und Marburg, Standort Marburg, Philipps-Universität Marburg, Baldingerstraße, Marburg, Germany
| | - Cornelia Brendel
- Klinik für Hämatologie, Onkologie und Immunologie, Universitätsklinikum Gießen und Marburg, Standort Marburg, Philipps-Universität Marburg, Baldingerstraße, Marburg, Germany.
| |
Collapse
|
29
|
Søreide K, Watson MM, Lea D, Nordgård O, Søreide JA, Hagland HR. Assessment of clinically related outcomes and biomarker analysis for translational integration in colorectal cancer (ACROBATICC): study protocol for a population-based, consecutive cohort of surgically treated colorectal cancers and resected colorectal liver metastasis. J Transl Med 2016; 14:192. [PMID: 27357108 PMCID: PMC4928276 DOI: 10.1186/s12967-016-0951-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 06/21/2016] [Indexed: 02/08/2023] Open
Abstract
Background More accurate predictive and prognostic biomarkers for patients with colorectal cancer (CRC) primaries or colorectal liver metastasis (CLM) are needed. Outside clinical trials, the translational integration of emerging pathways and novel techniques should facilitate exploration of biomarkers for improved staging and prognosis. Methods An observational study exploring predictive and prognostic biomarkers in a population-based, consecutive cohort of surgically treated colorectal cancers and resected colorectal liver metastases. Long-term outcomes will be cancer-specific survival, recurrence-free survival and overall survival at 5 years from diagnosis. Beyond routine clinicopathological and anthropometric characteristics and laboratory and biochemistry results, the project allows for additional blood samples and fresh-frozen tumour and normal tissue for investigation of circulating tumour cells (CTCs) and novel biomarkers (e.g. immune cells, microRNAs etc.). Tumour specimens will be investigated by immunohistochemistry in full slides. Extracted DNA/RNA will be analysed for genomic markers using specific PCR techniques and next-generation sequencing (NGS) panels. Flow cytometry will be used to characterise biomarkers in blood. Collaboration is open and welcomed, with particular interest in mutual opportunities for validation studies. Status and perspectives The project is ongoing and recruiting at an expected rate of 120–150 patients per year, since January 2013. A project on circulating tumour cells (CTCs) has commenced, with analysis being prepared. Investigating molecular classes beyond the TNM staging is under way, including characteristics of microsatellite instability (MSI) and elevated microsatellite alterations in selected tetranucleotides (EMAST). Hot spot panels for known mutations in CRC are being investigated using NGS. Immune-cell characteristics are being performed by IHC and flow cytometry in tumour and peripheral blood samples. The project has ethical approval (REK Helse Vest, #2012/742), is financially supported with a Ph.D.-Grant (EMAST project; Folke Hermansen Cancer Fund) and a CTC-project (Norwegian Research Council; O. Nordgård). The ACROBATICC clinical and molecular biobank repository will serve as a long-term source for novel exploratory analysis and invite collaborators for mutual validation of promising biomarker results. The project aims to generate results that can help better discern prognostic groups in stage II/III cancers; explore prognostic and predictive biomarkers, and help detail the biology of colorectal liver metastasis for better patient selection and tailored treatment. The project is registered at http://www.ClinicalTrials.gov NCT01762813.
Collapse
Affiliation(s)
- Kjetil Søreide
- Department of Gastrointestinal Surgery, Stavanger University Hospital, POB 8100, 4068, Stavanger, Norway. .,Gastrointestinal Translational Research Unit, Laboratory for Molecular Biology, Stavanger University Hospital, Stavanger, Norway. .,Department of Clinical Medicine, University of Bergen, Bergen, Norway.
| | - Martin M Watson
- Department of Gastrointestinal Surgery, Stavanger University Hospital, POB 8100, 4068, Stavanger, Norway.,Gastrointestinal Translational Research Unit, Laboratory for Molecular Biology, Stavanger University Hospital, Stavanger, Norway
| | - Dordi Lea
- Gastrointestinal Translational Research Unit, Laboratory for Molecular Biology, Stavanger University Hospital, Stavanger, Norway.,Department of Pathology, Stavanger University Hospital, Stavanger, Norway
| | - Oddmund Nordgård
- Department of Haematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - Jon Arne Søreide
- Department of Gastrointestinal Surgery, Stavanger University Hospital, POB 8100, 4068, Stavanger, Norway.,Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Hanne R Hagland
- Gastrointestinal Translational Research Unit, Laboratory for Molecular Biology, Stavanger University Hospital, Stavanger, Norway.,Centre of Organelle Research (CORE), University of Stavanger, Stavanger, Norway
| | | |
Collapse
|
30
|
Ballester LY, Luthra R, Kanagal-Shamanna R, Singh RR. Advances in clinical next-generation sequencing: target enrichment and sequencing technologies. Expert Rev Mol Diagn 2016; 16:357-72. [PMID: 26680590 DOI: 10.1586/14737159.2016.1133298] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The huge parallel sequencing capabilities of next generation sequencing technologies have made them the tools of choice to characterize genomic aberrations for research and diagnostic purposes. For clinical applications, screening the whole genome or exome is challenging owing to the large genomic area to be sequenced, associated costs, complexity of data, and lack of known clinical significance of all genes. Consequently, routine screening involves limited markers with established clinical relevance. This process, referred to as targeted genome sequencing, requires selective enrichment of the genomic areas comprising these markers via one of several primer or probe-based enrichment strategies, followed by sequencing of the enriched genomic areas. Here, the authors review current target enrichment approaches and next generation sequencing platforms, focusing on the underlying principles, capabilities, and limitations of each technology along with validation and implementation for clinical testing.
Collapse
Affiliation(s)
- Leomar Y Ballester
- a Department of Pathology and Immunology , Baylor College of Medicine , Houston , TX , USA
| | - Rajyalakshmi Luthra
- b Department of Hematopathology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Rashmi Kanagal-Shamanna
- b Department of Hematopathology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Rajesh R Singh
- b Department of Hematopathology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|