Analytical Validation of the Next-Generation Sequencing Assay for a Nationwide Signal- Finding Clinical Trial  Chih-Jian Lih, Robin D. Harrington, David.

Slides:



Advertisements
Similar presentations
Cancer Treatment from the DNA Perspective
Advertisements

Keyur P. Patel, Bedia A. Barkoh, Zhao Chen, Deqin Ma, Neelima Reddy, L
A Genome-Wide High-Resolution Array-CGH Analysis of Cutaneous Melanoma and Comparison of Array-CGH to FISH in Diagnostic Evaluation  Lu Wang, Mamta Rao,
Comparison of Clinical Targeted Next-Generation Sequence Data from Formalin-Fixed and Fresh-Frozen Tissue Specimens  David H. Spencer, Jennifer K. Sehn,
Development of a Next-Generation Sequencing Method for BRCA Mutation Screening  Maurice Chan, Shen Mo Ji, Zhen Xuan Yeo, Linda Gan, Eric Yap, Yoon Sim.
Molecular Diagnostic Profiling of Lung Cancer Specimens with a Semiconductor-Based Massive Parallel Sequencing Approach  Volker Endris, Roland Penzel,
Anna Sapino, Paul Roepman, Sabine C. Linn, Mireille H. J
DNA Extraction of Lung Cancer Samples for Advanced Diagnostic Testing
High-Resolution Genomic Profiling of Disseminated Tumor Cells in Prostate Cancer  Yu Wu, Jamie R. Schoenborn, Colm Morrissey, Jing Xia, Sandy Larson, Lisha.
Droplet Digital PCR for Absolute Quantification of EML4-ALK Gene Rearrangement in Lung Adenocarcinoma  Qiushi Wang, Xin Yang, Yong He, Qiang Ma, Li Lin,
Carol Beadling, Tanaya L. Neff, Michael C
A Targeted High-Throughput Next-Generation Sequencing Panel for Clinical Screening of Mutations, Gene Amplifications, and Fusions in Solid Tumors  Rajyalakshmi.
Assessing Copy Number Alterations in Targeted, Amplicon-Based Next-Generation Sequencing Data  Catherine Grasso, Timothy Butler, Katherine Rhodes, Michael.
Targeted Next-Generation Sequencing for Hereditary Cancer Syndromes
Application of Selected Reaction Monitoring for Multiplex Quantification of Clinically Validated Biomarkers in Formalin-Fixed, Paraffin-Embedded Tumor.
Emily M. Kudalkar, Naif A. M
In Silico Proficiency Testing for Clinical Next-Generation Sequencing
Comprehensive Screening of Gene Copy Number Aberrations in Formalin-Fixed, Paraffin-Embedded Solid Tumors Using Molecular Inversion Probe–Based Single-
A Method to Evaluate the Quality of Clinical Gene-Panel Sequencing Data for Single- Nucleotide Variant Detection  Chung Lee, Joon S. Bae, Gyu H. Ryu, Nayoung.
Validation of a Next-Generation Sequencing Pipeline for the Molecular Diagnosis of Multiple Inherited Cancer Predisposing Syndromes  Paula Paulo, Pedro.
Analytical Validation of a Next-Generation Sequencing Assay to Monitor Immune Responses in Solid Tumors  Jeffrey M. Conroy, Sarabjot Pabla, Sean T. Glenn,
Performance of Common Analysis Methods for Detecting Low-Frequency Single Nucleotide Variants in Targeted Next-Generation Sequence Data  David H. Spencer,
Somatic Mutation Screening Using Archival Formalin-Fixed, Paraffin-Embedded Tissues by Fluidigm Multiplex PCR and Illumina Sequencing  Ming Wang, Leire.
Targeted, High-Depth, Next-Generation Sequencing of Cancer Genes in Formalin- Fixed, Paraffin-Embedded and Fine-Needle Aspiration Tumor Specimens  Andrew.
Multiplex Preamplification of Serum DNA to Facilitate Reliable Detection of Extremely Rare Cancer Mutations in Circulating DNA by Digital PCR  Jennifer.
Analytical Validation of the Next-Generation Sequencing Assay for a Nationwide Signal- Finding Clinical Trial  Chih-Jian Lih, Robin D. Harrington, David.
Molecular Analysis of Circulating Cell-Free DNA from Lung Cancer Patients in Routine Laboratory Practice  Stephan Bartels, Sascha Persing, Britta Hasemeier,
Application of Single-Molecule Amplification and Resequencing Technology for Broad Surveillance of Plasma Mutations in Patients with Advanced Lung Adenocarcinoma 
Clinical Validation of a Next-Generation Sequencing Genomic Oncology Panel via Cross-Platform Benchmarking against Established Amplicon Sequencing Assays 
Clinical Genomic Profiling of a Diverse Array of Oncology Specimens at a Large Academic Cancer Center  Anthony N. Sireci, Vimla S. Aggarwal, Andrew T.
Plasmid-Based Materials as Multiplex Quality Controls and Calibrators for Clinical Next- Generation Sequencing Assays  David J. Sims, Robin D. Harrington,
Application of Single-Molecule Amplification and Resequencing Technology for Broad Surveillance of Plasma Mutations in Patients with Advanced Lung Adenocarcinoma 
Assessment of Capture and Amplicon-Based Approaches for the Development of a Targeted Next-Generation Sequencing Pipeline to Personalize Lymphoma Management 
A Method for Next-Generation Sequencing of Paired Diagnostic and Remission Samples to Detect Mitochondrial DNA Mutations Associated with Leukemia  Ilaria.
Maxim B. Freidin, Neesa Bhudia, Eric Lim, Andrew G
Utilization of Whole-Exome Next-Generation Sequencing Variant Read Frequency for Detection of Lesion-Specific, Somatic Loss of Heterozygosity in a Neurofibromatosis.
Christopher R. Cabanski, Vincent Magrini, Malachi Griffith, Obi L
CRISPR/Cas9 Technology–Based Xenograft Tumors as Candidate Reference Materials for Multiple EML4-ALK Rearrangements Testing  Rongxue Peng, Rui Zhang,
Quantitative Measurement of Cell-Free Plasma DNA and Applications for Detecting Tumor Genetic Variation and Promoter Methylation in a Clinical Setting 
Study of Preanalytic and Analytic Variables for Clinical Next-Generation Sequencing of Circulating Cell-Free Nucleic Acid  Meenakshi Mehrotra, Rajesh.
Anna Sapino, Paul Roepman, Sabine C. Linn, Mireille H. J
Certified DNA Reference Materials to Compare HER2 Gene Amplification Measurements Using Next-Generation Sequencing Methods  Chih-Jian Lih, Han Si, Biswajit.
A Genome-Wide High-Resolution Array-CGH Analysis of Cutaneous Melanoma and Comparison of Array-CGH to FISH in Diagnostic Evaluation  Lu Wang, Mamta Rao,
High-Throughput and Sensitive Quantification of Circulating Tumor DNA by Microfluidic- Based Multiplex PCR and Next-Generation Sequencing  Yinghui Guan,
Catherine E. Cottrell, Hussam Al-Kateb, Andrew J. Bredemeyer, Eric J
Detection of Dual IDH1 and IDH2 Mutations by Targeted Next-Generation Sequencing in Acute Myeloid Leukemia and Myelodysplastic Syndromes  Mia Y. Platt,
Validation and Implementation of a Custom Next-Generation Sequencing Clinical Assay for Hematologic Malignancies  Michael J. Kluk, R. Coleman Lindsley,
Comprehensive Diagnostic Testing for Stereocilin
Development of a Next-Generation Sequencing Method for BRCA Mutation Screening  Maurice Chan, Shen Mo Ji, Zhen Xuan Yeo, Linda Gan, Eric Yap, Yoon Sim.
Rapid Detection of TEM-Type Extended-Spectrum β-Lactamase (ESBL) Mutations Using Lights-On/Lights-Off Probes with Single-Stranded DNA Amplification  Kenneth.
Study of Preanalytic and Analytic Variables for Clinical Next-Generation Sequencing of Circulating Cell-Free Nucleic Acid  Meenakshi Mehrotra, Rajesh.
Analytical Validation of Clinical Whole-Genome and Transcriptome Sequencing of Patient-Derived Tumors for Reporting Targetable Variants in Cancer  Kazimierz.
Keyur P. Patel, Bedia A. Barkoh, Zhao Chen, Deqin Ma, Neelima Reddy, L
Utility of NIST Whole-Genome Reference Materials for the Technical Validation of a Multigene Next-Generation Sequencing Test  Bennett O.V. Shum, Ilya.
Development and Validation of a Template-Independent Next-Generation Sequencing Assay for Detecting Low-Level Resistance-Associated Variants of Hepatitis.
Ashleigh C. McEvoy, Benjamin A. Wood, Nima M. Ardakani, Michelle R
SeqReporter The Journal of Molecular Diagnostics
Analytical Validation and Application of a Targeted Next-Generation Sequencing Mutation-Detection Assay for Use in Treatment Assignment in the NCI-MPACT.
A Variant Detection Pipeline for Inherited Cardiomyopathy–Associated Genes Using Next-Generation Sequencing  Théo G.M. Oliveira, Miguel Mitne-Neto, Louise.
A Clinically Validated Diagnostic Second-Generation Sequencing Assay for Detection of Hereditary BRCA1 and BRCA2 Mutations  Ian E. Bosdet, T. Roderick.
Comprehensive Genomic Profiling of Malignant Effusions in Patients with Metastatic Lung Adenocarcinoma  Soo-Ryum Yang, Chieh-Yu Lin, Henning Stehr, Steven.
A Pyrosequencing-Based Assay for the Rapid Detection of the 22q11
A Platform for Rapid Detection of Multiple Oncogenic Mutations With Relevance to Targeted Therapy in Non–Small-Cell Lung Cancer  Zengliu Su, Dora Dias-Santagata,
Clinical Validation of a Next-Generation Sequencing Screen for Mutational Hotspots in 46 Cancer-Related Genes  Rajesh R. Singh, Keyur P. Patel, Mark J.
Validation and Reproducibility of a Microarray-Based Gene Expression Test for Tumor Identification in Formalin-Fixed, Paraffin-Embedded Specimens  Raji.
Nathan D. Montgomery, Sara R. Selitsky, Nirali M. Patel, D
Multilaboratory Assessment of a New Reference Material for Quality Assurance of Cell- Free Tumor DNA Measurements  Hua-Jun He, Erica V. Stein, Yves Konigshofer,
Development of a Novel Next-Generation Sequencing Assay for Carrier Screening in Old Order Amish and Mennonite Populations of Pennsylvania  Erin L. Crowgey,
MammaPrint and BluePrint Molecular Diagnostics Using Targeted RNA Next-Generation Sequencing Technology  Lorenza Mittempergher, Leonie J.M.J. Delahaye,
Preanalytic Variables and Tissue Stewardship for Reliable Next-Generation Sequencing (NGS) Clinical Analysis  Paolo A. Ascierto, Carlo Bifulco, Giuseppe.
Presentation transcript:

Analytical Validation of the Next-Generation Sequencing Assay for a Nationwide Signal- Finding Clinical Trial  Chih-Jian Lih, Robin D. Harrington, David J. Sims, Kneshay N. Harper, Courtney H. Bouk, Vivekananda Datta, Jonathan Yau, Rajesh R. Singh, Mark J. Routbort, Rajyalakshmi Luthra, Keyur P. Patel, Geeta S. Mantha, Savitri Krishnamurthy, Karyn Ronski, Zenta Walther, Karin E. Finberg, Sandra Canosa, Hayley Robinson, Amelia Raymond, Long P. Le, Lisa M. McShane, Eric C. Polley, Barbara A. Conley, James H. Doroshow, A. John Iafrate, Jeffrey L. Sklar, Stanley R. Hamilton, P. Mickey Williams  The Journal of Molecular Diagnostics  Volume 19, Issue 2, Pages 313-327 (March 2017) DOI: 10.1016/j.jmoldx.2016.10.007 Copyright © 2017 American Society for Investigative Pathology and the Association for Molecular Pathology Terms and Conditions

Figure 1 Workflows and components in the NCI-MATCH assay system. Four core needle specimens biopsied from a patient are shipped to central pathology laboratory at MD Anderson Cancer Center (MDACC). Tissue specimens are registered, and the preanalytic processes include formalin fixation and paraffin embedding (FFPE), hematoxylin and eosin staining, and tumor content assessment. The content of tumor cellularity of a specimen is required to be ≥70%, and enrichment by manual microdissections is performed on all specimens to attempt to achieve >70% tumor cellularity before nucleic acid (NA) extraction. A PTEN immunohistochemistry (IHC) stain is also performed as an inclusion biomarker for one treatment arm in the NCI-MATCH study. All residual tissue and nucleic acid samples are achieved in the ECOG-ACRIN Central Biorepository and Pathology Facility at the MDACC. One patient's genomic DNA (gDNA) and cDNA (reverse transcribed from total RNA) are shipped to one of four clinical laboratories to perform the NCI-MATCH next-generation sequencing (NGS) assay. The NGS data are analyzed by Ion Reporter (Torrent Suite version 4.4.2 Oncomine workflow) and the variant calling format (VCF) file and binary alignment map (BAM) file are automatically uploaded to MATCHBox, the centralized data processing system. MATCHBox identifies the actionable mutations of interest and assigns the treatment. The variant report and treatment assignment are reviewed by a group of experts composed of bioinformaticans, molecular biologists, statisticians, oncologists, and pathologists before sign off of the final report. The final results are sent to the enrolling physicians and patients and to a clinical medical database (DB) for long-term storage. The overall turnaround time from biopsy to final report is intended to be between 11 and 14 days. FNLCR, Frederick National Laboratory for Cancer Research; MGH, Massachusetts General Hospital; YSM, Yale University School of Medicine. The Journal of Molecular Diagnostics 2017 19, 313-327DOI: (10.1016/j.jmoldx.2016.10.007) Copyright © 2017 American Society for Investigative Pathology and the Association for Molecular Pathology Terms and Conditions

Figure 2 Tissue type has no effect on library yield. Library yields are plotted against the tissue types from which DNA samples (A) and RNA samples (B) were extracted. Each dot represents a sample, and each color code represents the laboratory site where the NCI-MATCH NGS assay was performed. The red dashed lines represent a minimal yield of 20 pmol/L required for template preparation. FNLCR, Frederick National Laboratory for Cancer Research; MDACC, MD Anderson Cancer Center; MGH, Massachusetts General Hospital; YSM, Yale University School of Medicine. The Journal of Molecular Diagnostics 2017 19, 313-327DOI: (10.1016/j.jmoldx.2016.10.007) Copyright © 2017 American Society for Investigative Pathology and the Association for Molecular Pathology Terms and Conditions

Figure 3 Reproducibility assessment across 16 technical replicates in four network laboratories. A and B: Reproducibility of 45 positive variants consisting of 38 variants detected in 9 DNA samples (A) and 7 fusion variants in 7 RNA samples (B) that were called at least once in 16 technical replicates across 4 network laboratories. Each row represents a variant, and the type of variant, sample, gene, Catalogue of Somatic Mutations in Cancer(COSMIC) identification or position (hg19) for variants without COSMIC identification are labeled. Each column represents a replicate; the laboratory and operator and replicate are indicated. The color code represents a variant call by the NCI-MATCH NGS assay (gray, no call; brown, a call). A: Eight DNA variants discordantly called (top) include 7 variants (6 indels and 1 SNV) detected once and 1 variant (Indel) detected only twice in 16 technical replicates. To simplify the presentation, replicates generated from OP2 and OP3 at the Yale University School of Medicine (YSM) are grouped and indicated as OP2. C and D: Allele frequencies of 27 variants (C) and copy numbers of 3 variants (D) were concordantly detected in 16 technical replicates and are plotted as box plot. Each row represents a variant, and the allele frequency copy number detected in 16 replicates are grouped by laboratory and color-coded as indicated in the legends. FNLCR, Frederick National Laboratory for Cancer Research; indel, insertion and deletion; MDACC, MD Anderson Cancer Center; MGH, Massachusetts General Hospital; OP, operator. The Journal of Molecular Diagnostics 2017 19, 313-327DOI: (10.1016/j.jmoldx.2016.10.007) Copyright © 2017 American Society for Investigative Pathology and the Association for Molecular Pathology Terms and Conditions