1
|
Yang Z, Chen K, Liu Y, Wang X, Wang S, Hao B. Regulation and analysis of Simiao Yong'an Decoction fermentation by Bacillus subtilis on the diversity of intestinal microbiota in Sprague-Dawley rats. Vet World 2024; 17:712-719. [PMID: 38680148 PMCID: PMC11045531 DOI: 10.14202/vetworld.2024.712-719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/05/2024] [Indexed: 05/01/2024] Open
Abstract
Background and Aim Simiao Yong'an decoction (SYD) is a classic traditional Chinese medicine (TCM) prescription that has the effects of clearing heat, detoxifying, promoting blood circulation, and relieving pain. In this study, we investigated the effect of SYD on the diversity of intestinal microbiota after fermentation by Bacillus subtilis. Materials and Methods SYD was fermented using B. subtilis. Female Sprague-Dawley rats were randomly divided into the following four groups with six rats in each group: Negative sample group (NS), water exaction non-fermentation group (WE), B. subtilis group (BS), and fermentation liquid group (FL). All rats were orally administered for 14 days. High-throughput Illumina sequencing was used to analyze 16S rRNA expression in rat fecal samples. Results A total of 2782 operational taxonomical units (OTUs) were identified in this study, and 634 OTUs were shared among all samples. Bacteroidetes (28.17%-53.20%) and Firmicutes (48.35%-67.83%) were the most abundant phyla identified among the four groups. The abundance of Escherichia and Alistipes was lower in the FL group than in the NS group, whereas the abundance of Bifidobacteria and Lactobacillus was increased in the FL group (p < 0.05). The abundance of Bifidobacterium was significantly upregulated in the FL group compared with the WE and BS groups (p < 0.05). Conclusion After fermentation, SYD had a significantly better effect than SYD or B. subtilis. SYD significantly promoted the growth of intestinal probiotics, inhibited the growth of pathogenic bacteria, and maintained the balance of intestinal microbiota in SD rats. This study provides new insights into the development and use of SYD.
Collapse
Affiliation(s)
- Zhen Yang
- Key Laboratory of New Animal Drug Project, Gansu Province, China; Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agriculture Sciences, Lanzhou, China
| | - Keyuan Chen
- Key Laboratory of New Animal Drug Project, Gansu Province, China; Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agriculture Sciences, Lanzhou, China
| | - Yu Liu
- Key Laboratory of New Animal Drug Project, Gansu Province, China; Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agriculture Sciences, Lanzhou, China
| | - Xuehong Wang
- Key Laboratory of New Animal Drug Project, Gansu Province, China; Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agriculture Sciences, Lanzhou, China
| | - Shengyi Wang
- Key Laboratory of New Animal Drug Project, Gansu Province, China; Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agriculture Sciences, Lanzhou, China
| | - Baocheng Hao
- Key Laboratory of New Animal Drug Project, Gansu Province, China; Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agriculture Sciences, Lanzhou, China
| |
Collapse
|
2
|
Maden SK, Kwon SH, Huuki-Myers LA, Collado-Torres L, Hicks SC, Maynard KR. Challenges and opportunities to computationally deconvolve heterogeneous tissue with varying cell sizes using single-cell RNA-sequencing datasets. Genome Biol 2023; 24:288. [PMID: 38098055 PMCID: PMC10722720 DOI: 10.1186/s13059-023-03123-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 11/24/2023] [Indexed: 12/17/2023] Open
Abstract
Deconvolution of cell mixtures in "bulk" transcriptomic samples from homogenate human tissue is important for understanding disease pathologies. However, several experimental and computational challenges impede transcriptomics-based deconvolution approaches using single-cell/nucleus RNA-seq reference atlases. Cells from the brain and blood have substantially different sizes, total mRNA, and transcriptional activities, and existing approaches may quantify total mRNA instead of cell type proportions. Further, standards are lacking for the use of cell reference atlases and integrative analyses of single-cell and spatial transcriptomics data. We discuss how to approach these key challenges with orthogonal "gold standard" datasets for evaluating deconvolution methods.
Collapse
Affiliation(s)
- Sean K Maden
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sang Ho Kwon
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Louise A Huuki-Myers
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Leonardo Collado-Torres
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Stephanie C Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA.
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, USA.
| | - Kristen R Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA.
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
3
|
Yang S, Zhang Z, Xian Q, Song Q, Liu Y, Gao Y, Wen W. An Aluminum-Based Microfluidic Chip for Polymerase Chain Reaction Diagnosis. Molecules 2023; 28:molecules28031085. [PMID: 36770751 PMCID: PMC9921548 DOI: 10.3390/molecules28031085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/13/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Real-time polymerase chain reaction (real-time PCR) tests were successfully conducted in an aluminum-based microfluidic chip developed in this work. The reaction chamber was coated with silicone-modified epoxy resin to isolate the reaction system from metal surfaces, preventing the metal ions from interfering with the reaction process. The patterned aluminum substrate was bonded with a hydroxylated glass mask using silicone sealant at room temperature. The effect of thermal expansion was counteracted by the elasticity of cured silicone. With the heating process closely monitored, real-time PCR testing in reaction chambers proceeded smoothly, and the results show similar quantification cycle values to those of traditional test sets. Scanning electron microscope (SEM) and atomic force microscopy (AFM) images showed that the surface of the reaction chamber was smoothly coated, illustrating the promising coating and isolating properties. Energy-dispersive X-ray spectroscopy (EDS), X-ray photoelectron spectroscopy (XPS), and inductively coupled plasma-optical emission spectrometer (ICP-OES) showed that no metal ions escaped from the metal to the chip surface. Fourier-transform infrared spectroscopy (FTIR) was used to check the surface chemical state before and after tests, and the unchanged infrared absorption peaks indicated the unreacted, antifouling surface. The limit of detection (LOD) of at least two copies can be obtained in this chip.
Collapse
Affiliation(s)
- Siyu Yang
- Division of Emerging Interdisciplinary Areas, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR 999077, China
| | - Ziyi Zhang
- Division of Emerging Interdisciplinary Areas, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR 999077, China
| | - Qingyue Xian
- Division of Emerging Interdisciplinary Areas, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR 999077, China
| | - Qi Song
- Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR 999077, China
| | - Yiteng Liu
- Division of Emerging Interdisciplinary Areas, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR 999077, China
| | - Yibo Gao
- Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR 999077, China
| | - Weijia Wen
- Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR 999077, China
- Thrust of Advanced Materials, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou 511400, China
- HKUST Shenzhen-Hong Kong Collaborative Innovation Research Institute, Futian, Shenzhen 518000, China
- Correspondence: ; Tel.: +852-2358-5781
| |
Collapse
|
4
|
Bias in RNA-seq Library Preparation: Current Challenges and Solutions. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6647597. [PMID: 33987443 PMCID: PMC8079181 DOI: 10.1155/2021/6647597] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 04/09/2021] [Indexed: 12/26/2022]
Abstract
Although RNA sequencing (RNA-seq) has become the most advanced technology for transcriptome analysis, it also confronts various challenges. As we all know, the workflow of RNA-seq is extremely complicated and it is easy to produce bias. This may damage the quality of RNA-seq dataset and lead to an incorrect interpretation for sequencing result. Thus, our detailed understanding of the source and nature of these biases is essential for the interpretation of RNA-seq data, finding methods to improve the quality of RNA-seq experimental, or development bioinformatics tools to compensate for these biases. Here, we discuss the sources of experimental bias in RNA-seq. And for each type of bias, we discussed the method for improvement, in order to provide some useful suggestions for researcher in RNA-seq experimental.
Collapse
|
5
|
Macken WL, Vandrovcova J, Hanna MG, Pitceathly RDS. Applying genomic and transcriptomic advances to mitochondrial medicine. Nat Rev Neurol 2021; 17:215-230. [PMID: 33623159 DOI: 10.1038/s41582-021-00455-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2021] [Indexed: 02/07/2023]
Abstract
Next-generation sequencing (NGS) has increased our understanding of the molecular basis of many primary mitochondrial diseases (PMDs). Despite this progress, many patients with suspected PMD remain without a genetic diagnosis, which restricts their access to in-depth genetic counselling, reproductive options and clinical trials, in addition to hampering efforts to understand the underlying disease mechanisms. Although they represent a considerable improvement over their predecessors, current methods for sequencing the mitochondrial and nuclear genomes have important limitations, and molecular diagnostic techniques are often manual and time consuming. However, recent advances in genomics and transcriptomics offer realistic solutions to these challenges. In this Review, we discuss the current genetic testing approach for PMDs and the opportunities that exist for increased use of whole-genome NGS of nuclear and mitochondrial DNA (mtDNA) in the clinical environment. We consider the possible role for long-read approaches in sequencing of mtDNA and in the identification of novel nuclear genomic causes of PMDs. We examine the expanding applications of RNA sequencing, including the detection of cryptic variants that affect splicing and gene expression and the interpretation of rare and novel mitochondrial transfer RNA variants.
Collapse
Affiliation(s)
- William L Macken
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Jana Vandrovcova
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Michael G Hanna
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Robert D S Pitceathly
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK.
| |
Collapse
|
6
|
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: 28] [Impact Index Per Article: 7.0] [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
|
7
|
Abstract
DNA contamination from external sources (reagents, environment, operator, etc.) has long been assumed to be the main cause of spurious signals that appear under low-bacterial-biomass conditions. Here, we demonstrate that contamination can be separated from another, random signal generated during low-biomass-sample sequencing. This stochastic noise is not reproduced between technical replicates; however, results for any one replicate taken alone could look like a microbial community different from the controls. Using this information, we investigated respiratory samples from healthy humans and determined the narrow range of bacterial biomass where samples transition from producing reproducible microbial sequences to ones dominated by noise. We present a rigorous approach to studies involving low-bacterial-biomass samples to detect this source of noise and provide a framework for deciding if a sample is likely to be dominated by noise. We anticipate that this work will facilitate increased reproducibility in the characterization of potentially important low-biomass communities. The bacterial microbiome of human body sites, previously considered sterile, remains highly controversial because it can be challenging to isolate signal from noise when low-biomass samples are being analyzed. We tested the hypothesis that stochastic sequencing noise, separable from reagent contamination, is generated during sequencing on the Illumina MiSeq platform when DNA input is below a critical threshold. We first purified DNA from serial dilutions of Pseudomonas aeruginosa and from negative controls using three DNA purification kits, quantified input using droplet digital PCR, and then sequenced the 16S rRNA gene in four technical replicates. This process identified reproducible contaminant signal that was separable from an irreproducible stochastic noise, which occurred as bacterial biomass of samples decreased. This approach was then applied to authentic respiratory samples from healthy individuals (n = 22) that ranged from high to ultralow bacterial biomass. Using oral rinse, bronchoalveolar lavage (BAL) fluid, and exhaled breath condensate (EBC) samples and matched controls, we were able to demonstrate (i) that stochastic noise dominates sequencing in real-world low-bacterial-biomass samples that contain fewer than 104 copies of the 16S rRNA gene per sample, (ii) that critical examination of the community composition of technical replicates can be used to separate signal from noise, and (iii) that EBC is an irreproducible sampling modality for sampling the microbiome of the lower airways. We anticipate that these results combined with suggested methods for identifying and dealing with noisy communities will facilitate increased reproducibility while simultaneously permitting characterization of potentially important low-biomass communities.
Collapse
|
8
|
Chen K, Gao Y, Wang X, Cheng F, Haq SU, Liang Y, Yang Z, Li B, Liu Y, Wang Y, Liang J, Hao B. Regulation and analysis of the diversity of intestinal microbiota in SD rats by Danggui Buxue Tang (DBT) fermented with Bacillus subtilis. ANN MICROBIOL 2020. [DOI: 10.1186/s13213-020-01563-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Purpose
To investigate the effect of Danggui Buxue Tang (DBT) on intestinal microbiota diversity after fermentation by Bacillus subtilis.
Methods
B. subtilis was used to ferment DBT. Sprague Dawley (SD) rats were randomly divided into the following four groups with six rats in each group: the control group, DBT nonfermentation group, B. subtilis group, and DBT fermentation group. Rats were fed continuously for 14 days. The 16S rRNA of faecal samples was analysed by high-throughput Illumina sequencing.
Results
In total, 3483 operational taxonomical units (OTUs) were identified in this study, and 1236 OTUs were shared among all samples. Moreover, the most abundant phyla identified in this study were Bacteroidetes (29.65–38.19%) and Firmicutes (48.30–67.04%). The F/B ratios of the DBT nonfermentation group (1.07%) and the DBT fermentation group (1.78%) were slightly lower than those of the control group (2.29%). Lactobacillus was most upregulated in the DBT fermentation group (38.4%), followed by the DBT nonfermentation group (18.97%), control group (14.61%), and probiotics group (8.39%). Moreover, the pathogenic bacteria Alistipes and Parabacteroides were found to be downregulated in the DBT fermentation group (the percentages of Alistipes and Parabacteroides were as follows: control group, 8.09% and 0.16%; DBT nonfermentation group, 4.31% and 0.37%; DBT fermentation group, 1.96 and 0.09%; and probiotics group, 6.25% and 0.12%, respectively).
Conclusion
This study is the first to research systematically the effects of DBT on the diversity of rat intestinal microbiota before and after fermentation. The structural characteristics of complex bacterial community in each group were clearly analysed, and DBT significantly increases probiotics and inhibits pathogenic bacterial growth in the intestinal tract of rats after fermentation, which plays a significant role in maintaining the balance of the intestinal microbiota of the rats. This research provides new insights into the development and utilization of traditional Chinese medicine.
Collapse
|
9
|
Roy-Chowdhuri S, Dacic S, Ghofrani M, Illei PB, Layfield LJ, Lee C, Michael CW, Miller RA, Mitchell JW, Nikolic B, Nowak JA, Pastis NJ, Rauch CA, Sharma A, Souter L, Billman BL, Thomas NE, VanderLaan PA, Voss JS, Wahidi MM, Yarmus LB, Gilbert CR. Collection and Handling of Thoracic Small Biopsy and Cytology Specimens for Ancillary Studies: Guideline From the College of American Pathologists in Collaboration With the American College of Chest Physicians, Association for Molecular Pathology, American Society of Cytopathology, American Thoracic Society, Pulmonary Pathology Society, Papanicolaou Society of Cytopathology, Society of Interventional Radiology, and Society of Thoracic Radiology. Arch Pathol Lab Med 2020; 144:933-958. [PMID: 32401054 DOI: 10.5858/arpa.2020-0119-cp] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT.— The need for appropriate specimen use for ancillary testing has become more commonplace in the practice of pathology. This, coupled with improvements in technology, often provides less invasive methods of testing, but presents new challenges to appropriate specimen collection and handling of these small specimens, including thoracic small biopsy and cytology samples. OBJECTIVE.— To develop a clinical practice guideline including recommendations on how to obtain, handle, and process thoracic small biopsy and cytology tissue specimens for diagnostic testing and ancillary studies. METHODS.— The College of American Pathologists convened an expert panel to perform a systematic review of the literature and develop recommendations. Core needle biopsy, touch preparation, fine-needle aspiration, and effusion specimens with thoracic diseases including malignancy, granulomatous process/sarcoidosis, and infection (eg, tuberculosis) were deemed within scope. Ancillary studies included immunohistochemistry and immunocytochemistry, fluorescence in situ hybridization, mutational analysis, flow cytometry, cytogenetics, and microbiologic studies routinely performed in the clinical pathology laboratory. The use of rapid on-site evaluation was also covered. RESULTS.— Sixteen guideline statements were developed to assist clinicians and pathologists in collecting and processing thoracic small biopsy and cytology tissue samples. CONCLUSIONS.— Based on the systematic review and expert panel consensus, thoracic small specimens can be handled and processed to perform downstream testing (eg, molecular markers, immunohistochemical biomarkers), core needle and fine-needle techniques can provide appropriate cytologic and histologic specimens for ancillary studies, and rapid on-site cytologic evaluation remains helpful in appropriate triage, handling, and processing of specimens.
Collapse
Affiliation(s)
- Sinchita Roy-Chowdhuri
- From the Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); Thoracic Surgery and Interventional Pulmonology, Swedish Cancer Institute, Seattle, Washington (Dr Gilbert); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Pathology, PeaceHealth Southwest Medical Center, Vancouver, Washington (Dr Ghofrani); the Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (Dr Illei); the Department of Pathology and Anatomic Sciences, University of Missouri, Columbia (Dr Layfield); the Department of Radiology, Keck Medical Center of the University of Southern California, Los Angeles (Dr Lee); the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Dr Michael); Memorial Pathology Consultants, PA, Houston, Texas (Dr Miller); the Department of Radiology, Capital Regional Medical Center, Tallahassee, Florida (Dr Mitchell); the Department of Interventional and Vascular Radiology, Cooley Dickinson Hospital, Northampton, Massachusetts (Dr Nikolic); the Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Division of Pulmonary and Critical Care, Medical University of South Carolina, Charleston (Dr Pastis); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Dr Rauch); the Department of Radiology, Massachusetts General Hospital, Boston (Dr Sharma); the Methodology Consultant, Ontario, Canada (Dr Souter); the Departments of Governance Services (Ms Billman) and Surveys (Ms Thomas), College of American Pathologists, Northfield, Illinois; the Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts (Dr VanderLaan); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Mr Voss); the Division of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina (Dr Wahidi); and the Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD (Dr Yarmus)
| | - Sanja Dacic
- From the Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); Thoracic Surgery and Interventional Pulmonology, Swedish Cancer Institute, Seattle, Washington (Dr Gilbert); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Pathology, PeaceHealth Southwest Medical Center, Vancouver, Washington (Dr Ghofrani); the Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (Dr Illei); the Department of Pathology and Anatomic Sciences, University of Missouri, Columbia (Dr Layfield); the Department of Radiology, Keck Medical Center of the University of Southern California, Los Angeles (Dr Lee); the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Dr Michael); Memorial Pathology Consultants, PA, Houston, Texas (Dr Miller); the Department of Radiology, Capital Regional Medical Center, Tallahassee, Florida (Dr Mitchell); the Department of Interventional and Vascular Radiology, Cooley Dickinson Hospital, Northampton, Massachusetts (Dr Nikolic); the Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Division of Pulmonary and Critical Care, Medical University of South Carolina, Charleston (Dr Pastis); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Dr Rauch); the Department of Radiology, Massachusetts General Hospital, Boston (Dr Sharma); the Methodology Consultant, Ontario, Canada (Dr Souter); the Departments of Governance Services (Ms Billman) and Surveys (Ms Thomas), College of American Pathologists, Northfield, Illinois; the Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts (Dr VanderLaan); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Mr Voss); the Division of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina (Dr Wahidi); and the Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD (Dr Yarmus)
| | - Mohiedean Ghofrani
- From the Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); Thoracic Surgery and Interventional Pulmonology, Swedish Cancer Institute, Seattle, Washington (Dr Gilbert); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Pathology, PeaceHealth Southwest Medical Center, Vancouver, Washington (Dr Ghofrani); the Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (Dr Illei); the Department of Pathology and Anatomic Sciences, University of Missouri, Columbia (Dr Layfield); the Department of Radiology, Keck Medical Center of the University of Southern California, Los Angeles (Dr Lee); the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Dr Michael); Memorial Pathology Consultants, PA, Houston, Texas (Dr Miller); the Department of Radiology, Capital Regional Medical Center, Tallahassee, Florida (Dr Mitchell); the Department of Interventional and Vascular Radiology, Cooley Dickinson Hospital, Northampton, Massachusetts (Dr Nikolic); the Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Division of Pulmonary and Critical Care, Medical University of South Carolina, Charleston (Dr Pastis); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Dr Rauch); the Department of Radiology, Massachusetts General Hospital, Boston (Dr Sharma); the Methodology Consultant, Ontario, Canada (Dr Souter); the Departments of Governance Services (Ms Billman) and Surveys (Ms Thomas), College of American Pathologists, Northfield, Illinois; the Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts (Dr VanderLaan); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Mr Voss); the Division of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina (Dr Wahidi); and the Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD (Dr Yarmus)
| | - Peter B Illei
- From the Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); Thoracic Surgery and Interventional Pulmonology, Swedish Cancer Institute, Seattle, Washington (Dr Gilbert); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Pathology, PeaceHealth Southwest Medical Center, Vancouver, Washington (Dr Ghofrani); the Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (Dr Illei); the Department of Pathology and Anatomic Sciences, University of Missouri, Columbia (Dr Layfield); the Department of Radiology, Keck Medical Center of the University of Southern California, Los Angeles (Dr Lee); the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Dr Michael); Memorial Pathology Consultants, PA, Houston, Texas (Dr Miller); the Department of Radiology, Capital Regional Medical Center, Tallahassee, Florida (Dr Mitchell); the Department of Interventional and Vascular Radiology, Cooley Dickinson Hospital, Northampton, Massachusetts (Dr Nikolic); the Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Division of Pulmonary and Critical Care, Medical University of South Carolina, Charleston (Dr Pastis); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Dr Rauch); the Department of Radiology, Massachusetts General Hospital, Boston (Dr Sharma); the Methodology Consultant, Ontario, Canada (Dr Souter); the Departments of Governance Services (Ms Billman) and Surveys (Ms Thomas), College of American Pathologists, Northfield, Illinois; the Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts (Dr VanderLaan); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Mr Voss); the Division of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina (Dr Wahidi); and the Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD (Dr Yarmus)
| | - Lester J Layfield
- From the Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); Thoracic Surgery and Interventional Pulmonology, Swedish Cancer Institute, Seattle, Washington (Dr Gilbert); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Pathology, PeaceHealth Southwest Medical Center, Vancouver, Washington (Dr Ghofrani); the Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (Dr Illei); the Department of Pathology and Anatomic Sciences, University of Missouri, Columbia (Dr Layfield); the Department of Radiology, Keck Medical Center of the University of Southern California, Los Angeles (Dr Lee); the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Dr Michael); Memorial Pathology Consultants, PA, Houston, Texas (Dr Miller); the Department of Radiology, Capital Regional Medical Center, Tallahassee, Florida (Dr Mitchell); the Department of Interventional and Vascular Radiology, Cooley Dickinson Hospital, Northampton, Massachusetts (Dr Nikolic); the Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Division of Pulmonary and Critical Care, Medical University of South Carolina, Charleston (Dr Pastis); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Dr Rauch); the Department of Radiology, Massachusetts General Hospital, Boston (Dr Sharma); the Methodology Consultant, Ontario, Canada (Dr Souter); the Departments of Governance Services (Ms Billman) and Surveys (Ms Thomas), College of American Pathologists, Northfield, Illinois; the Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts (Dr VanderLaan); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Mr Voss); the Division of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina (Dr Wahidi); and the Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD (Dr Yarmus)
| | - Christopher Lee
- From the Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); Thoracic Surgery and Interventional Pulmonology, Swedish Cancer Institute, Seattle, Washington (Dr Gilbert); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Pathology, PeaceHealth Southwest Medical Center, Vancouver, Washington (Dr Ghofrani); the Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (Dr Illei); the Department of Pathology and Anatomic Sciences, University of Missouri, Columbia (Dr Layfield); the Department of Radiology, Keck Medical Center of the University of Southern California, Los Angeles (Dr Lee); the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Dr Michael); Memorial Pathology Consultants, PA, Houston, Texas (Dr Miller); the Department of Radiology, Capital Regional Medical Center, Tallahassee, Florida (Dr Mitchell); the Department of Interventional and Vascular Radiology, Cooley Dickinson Hospital, Northampton, Massachusetts (Dr Nikolic); the Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Division of Pulmonary and Critical Care, Medical University of South Carolina, Charleston (Dr Pastis); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Dr Rauch); the Department of Radiology, Massachusetts General Hospital, Boston (Dr Sharma); the Methodology Consultant, Ontario, Canada (Dr Souter); the Departments of Governance Services (Ms Billman) and Surveys (Ms Thomas), College of American Pathologists, Northfield, Illinois; the Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts (Dr VanderLaan); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Mr Voss); the Division of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina (Dr Wahidi); and the Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD (Dr Yarmus)
| | - Claire W Michael
- From the Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); Thoracic Surgery and Interventional Pulmonology, Swedish Cancer Institute, Seattle, Washington (Dr Gilbert); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Pathology, PeaceHealth Southwest Medical Center, Vancouver, Washington (Dr Ghofrani); the Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (Dr Illei); the Department of Pathology and Anatomic Sciences, University of Missouri, Columbia (Dr Layfield); the Department of Radiology, Keck Medical Center of the University of Southern California, Los Angeles (Dr Lee); the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Dr Michael); Memorial Pathology Consultants, PA, Houston, Texas (Dr Miller); the Department of Radiology, Capital Regional Medical Center, Tallahassee, Florida (Dr Mitchell); the Department of Interventional and Vascular Radiology, Cooley Dickinson Hospital, Northampton, Massachusetts (Dr Nikolic); the Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Division of Pulmonary and Critical Care, Medical University of South Carolina, Charleston (Dr Pastis); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Dr Rauch); the Department of Radiology, Massachusetts General Hospital, Boston (Dr Sharma); the Methodology Consultant, Ontario, Canada (Dr Souter); the Departments of Governance Services (Ms Billman) and Surveys (Ms Thomas), College of American Pathologists, Northfield, Illinois; the Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts (Dr VanderLaan); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Mr Voss); the Division of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina (Dr Wahidi); and the Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD (Dr Yarmus)
| | - Ross A Miller
- From the Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); Thoracic Surgery and Interventional Pulmonology, Swedish Cancer Institute, Seattle, Washington (Dr Gilbert); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Pathology, PeaceHealth Southwest Medical Center, Vancouver, Washington (Dr Ghofrani); the Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (Dr Illei); the Department of Pathology and Anatomic Sciences, University of Missouri, Columbia (Dr Layfield); the Department of Radiology, Keck Medical Center of the University of Southern California, Los Angeles (Dr Lee); the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Dr Michael); Memorial Pathology Consultants, PA, Houston, Texas (Dr Miller); the Department of Radiology, Capital Regional Medical Center, Tallahassee, Florida (Dr Mitchell); the Department of Interventional and Vascular Radiology, Cooley Dickinson Hospital, Northampton, Massachusetts (Dr Nikolic); the Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Division of Pulmonary and Critical Care, Medical University of South Carolina, Charleston (Dr Pastis); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Dr Rauch); the Department of Radiology, Massachusetts General Hospital, Boston (Dr Sharma); the Methodology Consultant, Ontario, Canada (Dr Souter); the Departments of Governance Services (Ms Billman) and Surveys (Ms Thomas), College of American Pathologists, Northfield, Illinois; the Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts (Dr VanderLaan); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Mr Voss); the Division of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina (Dr Wahidi); and the Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD (Dr Yarmus)
| | - Jason W Mitchell
- From the Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); Thoracic Surgery and Interventional Pulmonology, Swedish Cancer Institute, Seattle, Washington (Dr Gilbert); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Pathology, PeaceHealth Southwest Medical Center, Vancouver, Washington (Dr Ghofrani); the Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (Dr Illei); the Department of Pathology and Anatomic Sciences, University of Missouri, Columbia (Dr Layfield); the Department of Radiology, Keck Medical Center of the University of Southern California, Los Angeles (Dr Lee); the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Dr Michael); Memorial Pathology Consultants, PA, Houston, Texas (Dr Miller); the Department of Radiology, Capital Regional Medical Center, Tallahassee, Florida (Dr Mitchell); the Department of Interventional and Vascular Radiology, Cooley Dickinson Hospital, Northampton, Massachusetts (Dr Nikolic); the Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Division of Pulmonary and Critical Care, Medical University of South Carolina, Charleston (Dr Pastis); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Dr Rauch); the Department of Radiology, Massachusetts General Hospital, Boston (Dr Sharma); the Methodology Consultant, Ontario, Canada (Dr Souter); the Departments of Governance Services (Ms Billman) and Surveys (Ms Thomas), College of American Pathologists, Northfield, Illinois; the Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts (Dr VanderLaan); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Mr Voss); the Division of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina (Dr Wahidi); and the Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD (Dr Yarmus)
| | - Boris Nikolic
- From the Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); Thoracic Surgery and Interventional Pulmonology, Swedish Cancer Institute, Seattle, Washington (Dr Gilbert); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Pathology, PeaceHealth Southwest Medical Center, Vancouver, Washington (Dr Ghofrani); the Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (Dr Illei); the Department of Pathology and Anatomic Sciences, University of Missouri, Columbia (Dr Layfield); the Department of Radiology, Keck Medical Center of the University of Southern California, Los Angeles (Dr Lee); the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Dr Michael); Memorial Pathology Consultants, PA, Houston, Texas (Dr Miller); the Department of Radiology, Capital Regional Medical Center, Tallahassee, Florida (Dr Mitchell); the Department of Interventional and Vascular Radiology, Cooley Dickinson Hospital, Northampton, Massachusetts (Dr Nikolic); the Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Division of Pulmonary and Critical Care, Medical University of South Carolina, Charleston (Dr Pastis); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Dr Rauch); the Department of Radiology, Massachusetts General Hospital, Boston (Dr Sharma); the Methodology Consultant, Ontario, Canada (Dr Souter); the Departments of Governance Services (Ms Billman) and Surveys (Ms Thomas), College of American Pathologists, Northfield, Illinois; the Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts (Dr VanderLaan); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Mr Voss); the Division of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina (Dr Wahidi); and the Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD (Dr Yarmus)
| | - Jan A Nowak
- From the Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); Thoracic Surgery and Interventional Pulmonology, Swedish Cancer Institute, Seattle, Washington (Dr Gilbert); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Pathology, PeaceHealth Southwest Medical Center, Vancouver, Washington (Dr Ghofrani); the Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (Dr Illei); the Department of Pathology and Anatomic Sciences, University of Missouri, Columbia (Dr Layfield); the Department of Radiology, Keck Medical Center of the University of Southern California, Los Angeles (Dr Lee); the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Dr Michael); Memorial Pathology Consultants, PA, Houston, Texas (Dr Miller); the Department of Radiology, Capital Regional Medical Center, Tallahassee, Florida (Dr Mitchell); the Department of Interventional and Vascular Radiology, Cooley Dickinson Hospital, Northampton, Massachusetts (Dr Nikolic); the Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Division of Pulmonary and Critical Care, Medical University of South Carolina, Charleston (Dr Pastis); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Dr Rauch); the Department of Radiology, Massachusetts General Hospital, Boston (Dr Sharma); the Methodology Consultant, Ontario, Canada (Dr Souter); the Departments of Governance Services (Ms Billman) and Surveys (Ms Thomas), College of American Pathologists, Northfield, Illinois; the Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts (Dr VanderLaan); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Mr Voss); the Division of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina (Dr Wahidi); and the Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD (Dr Yarmus)
| | - Nicholas J Pastis
- From the Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); Thoracic Surgery and Interventional Pulmonology, Swedish Cancer Institute, Seattle, Washington (Dr Gilbert); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Pathology, PeaceHealth Southwest Medical Center, Vancouver, Washington (Dr Ghofrani); the Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (Dr Illei); the Department of Pathology and Anatomic Sciences, University of Missouri, Columbia (Dr Layfield); the Department of Radiology, Keck Medical Center of the University of Southern California, Los Angeles (Dr Lee); the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Dr Michael); Memorial Pathology Consultants, PA, Houston, Texas (Dr Miller); the Department of Radiology, Capital Regional Medical Center, Tallahassee, Florida (Dr Mitchell); the Department of Interventional and Vascular Radiology, Cooley Dickinson Hospital, Northampton, Massachusetts (Dr Nikolic); the Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Division of Pulmonary and Critical Care, Medical University of South Carolina, Charleston (Dr Pastis); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Dr Rauch); the Department of Radiology, Massachusetts General Hospital, Boston (Dr Sharma); the Methodology Consultant, Ontario, Canada (Dr Souter); the Departments of Governance Services (Ms Billman) and Surveys (Ms Thomas), College of American Pathologists, Northfield, Illinois; the Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts (Dr VanderLaan); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Mr Voss); the Division of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina (Dr Wahidi); and the Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD (Dr Yarmus)
| | - Carol Ann Rauch
- From the Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); Thoracic Surgery and Interventional Pulmonology, Swedish Cancer Institute, Seattle, Washington (Dr Gilbert); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Pathology, PeaceHealth Southwest Medical Center, Vancouver, Washington (Dr Ghofrani); the Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (Dr Illei); the Department of Pathology and Anatomic Sciences, University of Missouri, Columbia (Dr Layfield); the Department of Radiology, Keck Medical Center of the University of Southern California, Los Angeles (Dr Lee); the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Dr Michael); Memorial Pathology Consultants, PA, Houston, Texas (Dr Miller); the Department of Radiology, Capital Regional Medical Center, Tallahassee, Florida (Dr Mitchell); the Department of Interventional and Vascular Radiology, Cooley Dickinson Hospital, Northampton, Massachusetts (Dr Nikolic); the Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Division of Pulmonary and Critical Care, Medical University of South Carolina, Charleston (Dr Pastis); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Dr Rauch); the Department of Radiology, Massachusetts General Hospital, Boston (Dr Sharma); the Methodology Consultant, Ontario, Canada (Dr Souter); the Departments of Governance Services (Ms Billman) and Surveys (Ms Thomas), College of American Pathologists, Northfield, Illinois; the Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts (Dr VanderLaan); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Mr Voss); the Division of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina (Dr Wahidi); and the Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD (Dr Yarmus)
| | - Amita Sharma
- From the Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); Thoracic Surgery and Interventional Pulmonology, Swedish Cancer Institute, Seattle, Washington (Dr Gilbert); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Pathology, PeaceHealth Southwest Medical Center, Vancouver, Washington (Dr Ghofrani); the Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (Dr Illei); the Department of Pathology and Anatomic Sciences, University of Missouri, Columbia (Dr Layfield); the Department of Radiology, Keck Medical Center of the University of Southern California, Los Angeles (Dr Lee); the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Dr Michael); Memorial Pathology Consultants, PA, Houston, Texas (Dr Miller); the Department of Radiology, Capital Regional Medical Center, Tallahassee, Florida (Dr Mitchell); the Department of Interventional and Vascular Radiology, Cooley Dickinson Hospital, Northampton, Massachusetts (Dr Nikolic); the Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Division of Pulmonary and Critical Care, Medical University of South Carolina, Charleston (Dr Pastis); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Dr Rauch); the Department of Radiology, Massachusetts General Hospital, Boston (Dr Sharma); the Methodology Consultant, Ontario, Canada (Dr Souter); the Departments of Governance Services (Ms Billman) and Surveys (Ms Thomas), College of American Pathologists, Northfield, Illinois; the Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts (Dr VanderLaan); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Mr Voss); the Division of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina (Dr Wahidi); and the Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD (Dr Yarmus)
| | - Lesley Souter
- From the Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); Thoracic Surgery and Interventional Pulmonology, Swedish Cancer Institute, Seattle, Washington (Dr Gilbert); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Pathology, PeaceHealth Southwest Medical Center, Vancouver, Washington (Dr Ghofrani); the Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (Dr Illei); the Department of Pathology and Anatomic Sciences, University of Missouri, Columbia (Dr Layfield); the Department of Radiology, Keck Medical Center of the University of Southern California, Los Angeles (Dr Lee); the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Dr Michael); Memorial Pathology Consultants, PA, Houston, Texas (Dr Miller); the Department of Radiology, Capital Regional Medical Center, Tallahassee, Florida (Dr Mitchell); the Department of Interventional and Vascular Radiology, Cooley Dickinson Hospital, Northampton, Massachusetts (Dr Nikolic); the Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Division of Pulmonary and Critical Care, Medical University of South Carolina, Charleston (Dr Pastis); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Dr Rauch); the Department of Radiology, Massachusetts General Hospital, Boston (Dr Sharma); the Methodology Consultant, Ontario, Canada (Dr Souter); the Departments of Governance Services (Ms Billman) and Surveys (Ms Thomas), College of American Pathologists, Northfield, Illinois; the Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts (Dr VanderLaan); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Mr Voss); the Division of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina (Dr Wahidi); and the Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD (Dr Yarmus)
| | - Brooke L Billman
- From the Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); Thoracic Surgery and Interventional Pulmonology, Swedish Cancer Institute, Seattle, Washington (Dr Gilbert); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Pathology, PeaceHealth Southwest Medical Center, Vancouver, Washington (Dr Ghofrani); the Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (Dr Illei); the Department of Pathology and Anatomic Sciences, University of Missouri, Columbia (Dr Layfield); the Department of Radiology, Keck Medical Center of the University of Southern California, Los Angeles (Dr Lee); the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Dr Michael); Memorial Pathology Consultants, PA, Houston, Texas (Dr Miller); the Department of Radiology, Capital Regional Medical Center, Tallahassee, Florida (Dr Mitchell); the Department of Interventional and Vascular Radiology, Cooley Dickinson Hospital, Northampton, Massachusetts (Dr Nikolic); the Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Division of Pulmonary and Critical Care, Medical University of South Carolina, Charleston (Dr Pastis); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Dr Rauch); the Department of Radiology, Massachusetts General Hospital, Boston (Dr Sharma); the Methodology Consultant, Ontario, Canada (Dr Souter); the Departments of Governance Services (Ms Billman) and Surveys (Ms Thomas), College of American Pathologists, Northfield, Illinois; the Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts (Dr VanderLaan); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Mr Voss); the Division of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina (Dr Wahidi); and the Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD (Dr Yarmus)
| | - Nicole E Thomas
- From the Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); Thoracic Surgery and Interventional Pulmonology, Swedish Cancer Institute, Seattle, Washington (Dr Gilbert); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Pathology, PeaceHealth Southwest Medical Center, Vancouver, Washington (Dr Ghofrani); the Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (Dr Illei); the Department of Pathology and Anatomic Sciences, University of Missouri, Columbia (Dr Layfield); the Department of Radiology, Keck Medical Center of the University of Southern California, Los Angeles (Dr Lee); the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Dr Michael); Memorial Pathology Consultants, PA, Houston, Texas (Dr Miller); the Department of Radiology, Capital Regional Medical Center, Tallahassee, Florida (Dr Mitchell); the Department of Interventional and Vascular Radiology, Cooley Dickinson Hospital, Northampton, Massachusetts (Dr Nikolic); the Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Division of Pulmonary and Critical Care, Medical University of South Carolina, Charleston (Dr Pastis); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Dr Rauch); the Department of Radiology, Massachusetts General Hospital, Boston (Dr Sharma); the Methodology Consultant, Ontario, Canada (Dr Souter); the Departments of Governance Services (Ms Billman) and Surveys (Ms Thomas), College of American Pathologists, Northfield, Illinois; the Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts (Dr VanderLaan); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Mr Voss); the Division of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina (Dr Wahidi); and the Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD (Dr Yarmus)
| | - Paul A VanderLaan
- From the Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); Thoracic Surgery and Interventional Pulmonology, Swedish Cancer Institute, Seattle, Washington (Dr Gilbert); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Pathology, PeaceHealth Southwest Medical Center, Vancouver, Washington (Dr Ghofrani); the Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (Dr Illei); the Department of Pathology and Anatomic Sciences, University of Missouri, Columbia (Dr Layfield); the Department of Radiology, Keck Medical Center of the University of Southern California, Los Angeles (Dr Lee); the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Dr Michael); Memorial Pathology Consultants, PA, Houston, Texas (Dr Miller); the Department of Radiology, Capital Regional Medical Center, Tallahassee, Florida (Dr Mitchell); the Department of Interventional and Vascular Radiology, Cooley Dickinson Hospital, Northampton, Massachusetts (Dr Nikolic); the Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Division of Pulmonary and Critical Care, Medical University of South Carolina, Charleston (Dr Pastis); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Dr Rauch); the Department of Radiology, Massachusetts General Hospital, Boston (Dr Sharma); the Methodology Consultant, Ontario, Canada (Dr Souter); the Departments of Governance Services (Ms Billman) and Surveys (Ms Thomas), College of American Pathologists, Northfield, Illinois; the Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts (Dr VanderLaan); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Mr Voss); the Division of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina (Dr Wahidi); and the Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD (Dr Yarmus)
| | - Jesse S Voss
- From the Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); Thoracic Surgery and Interventional Pulmonology, Swedish Cancer Institute, Seattle, Washington (Dr Gilbert); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Pathology, PeaceHealth Southwest Medical Center, Vancouver, Washington (Dr Ghofrani); the Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (Dr Illei); the Department of Pathology and Anatomic Sciences, University of Missouri, Columbia (Dr Layfield); the Department of Radiology, Keck Medical Center of the University of Southern California, Los Angeles (Dr Lee); the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Dr Michael); Memorial Pathology Consultants, PA, Houston, Texas (Dr Miller); the Department of Radiology, Capital Regional Medical Center, Tallahassee, Florida (Dr Mitchell); the Department of Interventional and Vascular Radiology, Cooley Dickinson Hospital, Northampton, Massachusetts (Dr Nikolic); the Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Division of Pulmonary and Critical Care, Medical University of South Carolina, Charleston (Dr Pastis); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Dr Rauch); the Department of Radiology, Massachusetts General Hospital, Boston (Dr Sharma); the Methodology Consultant, Ontario, Canada (Dr Souter); the Departments of Governance Services (Ms Billman) and Surveys (Ms Thomas), College of American Pathologists, Northfield, Illinois; the Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts (Dr VanderLaan); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Mr Voss); the Division of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina (Dr Wahidi); and the Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD (Dr Yarmus)
| | - Momen M Wahidi
- From the Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); Thoracic Surgery and Interventional Pulmonology, Swedish Cancer Institute, Seattle, Washington (Dr Gilbert); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Pathology, PeaceHealth Southwest Medical Center, Vancouver, Washington (Dr Ghofrani); the Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (Dr Illei); the Department of Pathology and Anatomic Sciences, University of Missouri, Columbia (Dr Layfield); the Department of Radiology, Keck Medical Center of the University of Southern California, Los Angeles (Dr Lee); the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Dr Michael); Memorial Pathology Consultants, PA, Houston, Texas (Dr Miller); the Department of Radiology, Capital Regional Medical Center, Tallahassee, Florida (Dr Mitchell); the Department of Interventional and Vascular Radiology, Cooley Dickinson Hospital, Northampton, Massachusetts (Dr Nikolic); the Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Division of Pulmonary and Critical Care, Medical University of South Carolina, Charleston (Dr Pastis); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Dr Rauch); the Department of Radiology, Massachusetts General Hospital, Boston (Dr Sharma); the Methodology Consultant, Ontario, Canada (Dr Souter); the Departments of Governance Services (Ms Billman) and Surveys (Ms Thomas), College of American Pathologists, Northfield, Illinois; the Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts (Dr VanderLaan); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Mr Voss); the Division of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina (Dr Wahidi); and the Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD (Dr Yarmus)
| | - Lonny B Yarmus
- From the Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); Thoracic Surgery and Interventional Pulmonology, Swedish Cancer Institute, Seattle, Washington (Dr Gilbert); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Pathology, PeaceHealth Southwest Medical Center, Vancouver, Washington (Dr Ghofrani); the Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (Dr Illei); the Department of Pathology and Anatomic Sciences, University of Missouri, Columbia (Dr Layfield); the Department of Radiology, Keck Medical Center of the University of Southern California, Los Angeles (Dr Lee); the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Dr Michael); Memorial Pathology Consultants, PA, Houston, Texas (Dr Miller); the Department of Radiology, Capital Regional Medical Center, Tallahassee, Florida (Dr Mitchell); the Department of Interventional and Vascular Radiology, Cooley Dickinson Hospital, Northampton, Massachusetts (Dr Nikolic); the Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Division of Pulmonary and Critical Care, Medical University of South Carolina, Charleston (Dr Pastis); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Dr Rauch); the Department of Radiology, Massachusetts General Hospital, Boston (Dr Sharma); the Methodology Consultant, Ontario, Canada (Dr Souter); the Departments of Governance Services (Ms Billman) and Surveys (Ms Thomas), College of American Pathologists, Northfield, Illinois; the Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts (Dr VanderLaan); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Mr Voss); the Division of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina (Dr Wahidi); and the Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD (Dr Yarmus)
| | - Christopher R Gilbert
- From the Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); Thoracic Surgery and Interventional Pulmonology, Swedish Cancer Institute, Seattle, Washington (Dr Gilbert); the Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Dacic); the Department of Pathology, PeaceHealth Southwest Medical Center, Vancouver, Washington (Dr Ghofrani); the Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (Dr Illei); the Department of Pathology and Anatomic Sciences, University of Missouri, Columbia (Dr Layfield); the Department of Radiology, Keck Medical Center of the University of Southern California, Los Angeles (Dr Lee); the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Dr Michael); Memorial Pathology Consultants, PA, Houston, Texas (Dr Miller); the Department of Radiology, Capital Regional Medical Center, Tallahassee, Florida (Dr Mitchell); the Department of Interventional and Vascular Radiology, Cooley Dickinson Hospital, Northampton, Massachusetts (Dr Nikolic); the Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York (Dr Nowak); the Division of Pulmonary and Critical Care, Medical University of South Carolina, Charleston (Dr Pastis); the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Dr Rauch); the Department of Radiology, Massachusetts General Hospital, Boston (Dr Sharma); the Methodology Consultant, Ontario, Canada (Dr Souter); the Departments of Governance Services (Ms Billman) and Surveys (Ms Thomas), College of American Pathologists, Northfield, Illinois; the Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts (Dr VanderLaan); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Mr Voss); the Division of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina (Dr Wahidi); and the Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD (Dr Yarmus)
| |
Collapse
|
10
|
Cryobiopsy increases the EGFR detection rate in non-small cell lung cancer. Lung Cancer 2020; 141:56-63. [PMID: 31955001 DOI: 10.1016/j.lungcan.2019.12.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 12/13/2019] [Accepted: 12/16/2019] [Indexed: 01/22/2023]
Abstract
OBJECTIVES Detection of activating epidermal growth factor receptor (EGFR) mutation is crucial for individualized treatment of advanced non-small-cell lung cancer (NSCLC). However little is known about how biopsy technique affects the detection rate of EGFR mutations. This retrospective, single center study evaluated the detection rate of EGFR mutations in tissue obtained by bronchoscopic cryobiopsy and compared this to other standard tissue sampling techniques. MATERIALS AND METHODS We retrospectively analyzed 414 patients with histologically confirmed NSCLC and known EGFR mutation status between 3/2008-7/2014. Tumor specimens obtained by tissue preserving bronchoscopic cryobiopsy were compared to those obtained by other techniques. RESULTS AND CONCLUSION Analysis of bronchoscopic cryobiopsy tissue detected 29 activating EGFR mutations in 27 (21.6 %) out of 125 patients, while analysis of tissue obtained by non-cryobiopsy techniques (bronchoscopic forceps biopsies, fine needle aspiration, imaging guided transthoracical and surgical procedures) detected 42 EGFR mutations in 40 (13.8 %) out of 298 patients (p < 0.05). Cryobiopsy increased detection rate of EGFR mutations in central tumors compared with forceps biopsy (19.6 % versus 6.5 %, p < 0.05), while an insignificant trend was detected also for peripheral tumors (33.3 % versus 26.9 %). Bronchosopic cryobiopsy increases the detection rate of activating EGFR mutations in NSCLC in comparison to other tissue sampling techniques. This will help to optimize individualized treatment of patients with advanced tumors. Because of the retrospective nature of this analysis, a prospective trial is mandatory for final assessment.
Collapse
|
11
|
Boggs LM, Scheible MKR, Machado G, Meiklejohn KA. Single Fragment or Bulk Soil DNA Metabarcoding: Which is Better for Characterizing Biological Taxa Found in Surface Soils for Sample Separation? Genes (Basel) 2019; 10:E431. [PMID: 31174412 PMCID: PMC6627524 DOI: 10.3390/genes10060431] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/02/2019] [Accepted: 06/05/2019] [Indexed: 12/18/2022] Open
Abstract
In forensic geology casework, sample size typically limits routine characterization of material using bulk approaches. To address this, DNA-based characterization of biological taxa has received attention, as the taxa present can be useful for sample-to-sample comparisons and source attribution. In our initial work, low biodiversity was captured when DNA barcodes were Sanger-sequenced from plant and insect fragments isolated from 10 forensic-type surface soils. Considering some forensic laboratories now have access to massively parallel sequencing platforms, we assessed whether biological taxa present in the same surface soils could be better characterized using DNA metabarcoding. To achieve this, plant and animal barcodes were amplified and sequenced on an Illumina MiniSeq for three different DNA sample types (n = 50): individual fragments used in our initial study, and 250 and 100 mg of bulk soil (from the 10 sites used in the initial study). A total of 572 unique target barcode sequences passed quality filtering and were used in downstream statistical analyses: 54, 321, and 285 for individual fragments, 100 mg, and 250 mg bulk soil samples, respectively. Plant barcodes permitted some spatial separation of sample sites in non-metric multidimensional scaling plots; better separation was obtained for samples prepared from bulk soil. This study confirmed that bulk soil DNA metabarcoding is a better approach for characterizing biological taxa present in surface soils, which could supplement traditional geologic examinations.
Collapse
Affiliation(s)
- Laura M Boggs
- Department of Population Health and Pathobiology, North Carolina State University, 1060 William Moore Drive, Raleigh, NC 27607, USA.
| | - Melissa K R Scheible
- Department of Population Health and Pathobiology, North Carolina State University, 1060 William Moore Drive, Raleigh, NC 27607, USA.
| | - Gustavo Machado
- Department of Population Health and Pathobiology, North Carolina State University, 1060 William Moore Drive, Raleigh, NC 27607, USA.
| | - Kelly A Meiklejohn
- Department of Population Health and Pathobiology, North Carolina State University, 1060 William Moore Drive, Raleigh, NC 27607, USA.
| |
Collapse
|
12
|
Santibanez-Koref M, Griffin H, Turnbull DM, Chinnery PF, Herbert M, Hudson G. Assessing mitochondrial heteroplasmy using next generation sequencing: A note of caution. Mitochondrion 2018; 46:302-306. [PMID: 30098421 PMCID: PMC6509278 DOI: 10.1016/j.mito.2018.08.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/09/2018] [Accepted: 08/02/2018] [Indexed: 12/31/2022]
Abstract
The mitochondrial genome has recently become the focus of several high-impact next-generation sequencing studies investigating the effect of mutations in disease and assessing the efficacy of mitochondrial replacement therapies. However, these studies have failed to take into consideration the capture of recurring translocations of mitochondrial DNA to the nuclear genome, known as nuclear mitochondrial sequences (NUMTs), continuing to align sequence data to the revised Cambridge reference sequence alone. Here, using different mtDNA enrichment techniques and a variety of tissues, we demonstrate that NUMTs are present in sequence data and that, dependent upon downstream analysis, are at a level which affects variant calling. Translocations of mtDNA to the nDNA genome are commonplace and present a challenge when performing next-generation-sequencing experiments aimed at identifying mtDNA heteroplasmy. Accurate next generation sequencing of mtDNA is affected by both target enrichment and downstream bioinformatic analysis strategy. NUMTs can affect heteroplasmy calling, but cannot wholly explain low-level sequencing artefacts.
Collapse
Affiliation(s)
- Mauro Santibanez-Koref
- Institute of Genetic Medicine, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Helen Griffin
- Institute of Genetic Medicine, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Douglass M Turnbull
- The Wellcome Centre for Mitochondrial Research, Newcastle University, Medical School, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Patrick F Chinnery
- MRC Mitochondrial Biology Unit, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Mary Herbert
- The Wellcome Centre for Mitochondrial Research, Newcastle University, Medical School, Framlington Place, Newcastle upon Tyne NE2 4HH, UK; Newcastle Fertility Centre, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Gavin Hudson
- Institute of Genetic Medicine, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK; The Wellcome Centre for Mitochondrial Research, Newcastle University, Medical School, Framlington Place, Newcastle upon Tyne NE2 4HH, UK.
| |
Collapse
|
13
|
ERIC recommendations for TP53 mutation analysis in chronic lymphocytic leukemia-update on methodological approaches and results interpretation. Leukemia 2018; 32:1070-1080. [PMID: 29467486 PMCID: PMC5940638 DOI: 10.1038/s41375-017-0007-7] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/05/2017] [Accepted: 12/08/2017] [Indexed: 02/06/2023]
Abstract
In chronic lymphocytic leukemia (CLL), TP53 gene defects, due to deletion of the 17p13 locus and/or mutation(s) within the TP53 gene, are associated with resistance to chemoimmunotherapy and a particularly dismal clinical outcome. On these grounds, analysis of TP53 aberrations has been incorporated into routine clinical diagnostics to improve patient stratification and optimize therapeutic decisions. The predictive implications of TP53 aberrations have increasing significance in the era of novel targeted therapies, i.e., inhibitors of B-cell receptor (BcR) signaling and anti-apoptotic BCL2 family members, owing to their efficacy in patients with TP53 defects. In this report, the TP53 Network of the European Research Initiative on Chronic Lymphocytic Leukemia (ERIC) presents updated recommendations on the methodological approaches for TP53 mutation analysis. Moreover, it provides guidance to ensure that the analysis is performed in a timely manner for all patients requiring treatment and that the data is interpreted and reported in a consistent, standardized, and accurate way. Since next-generation sequencing technologies are gaining prominence within diagnostic laboratories, this report also offers advice and recommendations for the interpretation of TP53 mutation data generated by this methodology.
Collapse
|
14
|
Memon AA, Zhang H, Gu Y, Luo Q, Shi J, Deng Z, Ma J, Ma W. EGFR with TKI-sensitive mutations in exon 19 is highly expressed and frequently detected in Chinese patients with lung squamous carcinoma. Onco Targets Ther 2017; 10:4607-4613. [PMID: 29075127 PMCID: PMC5609803 DOI: 10.2147/ott.s130051] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Recently, tyrosine kinase inhibitors (TKIs) have been recommended as a first-line treatment for advanced non-small cell lung cancer (NSCLC), significantly improving the treatment outcomes of lung adenocarcinoma patients with the EGFR mutation. However, the application of TKIs for lung squamous cell carcinoma (SCC), the second largest pathological subtype of NSCLC, remains controversial because available data for the EGFR mutation profile and frequency in SCC patients are limited. In this study, 89 bronchoscopic-biopsy specimens from Chinese SCC male patients were assayed for EGFR exon 19 mutation, using improved polymerase chain reaction-denature gel gradient electrophoresis. EGFR exon 19 mutations were detected in 77 of 89 (86.5%) patients, and included six kinds of point mutations (11.6%) and two deletions (Del_747-751 [64.9%] and Del_746-751 [23.3%]). We found that the proportion of mutated EGFR varied from 0.98% to 100% in positive specimens and increased with the development of the disease. The difference of proportion between Stage IV patients and Stage II patients or Stage III patients was significant (P<0.001). These results provided valuable clues to explain the reason why patients harboring the same mutation responded distinctly to TKI treatment. Del_747-751 and Del_746-751 were the dominant mutations in the assayed SCC patients (76.4%), and both belong to the EGFR-TKI-sensitive mutation. Recently research demonstrated that Del_746-751 patients have better response to EGFR-TKI than Del_L747-751 patients. However, our study indicated that majority of SCC patients (55.5%) carried Del_ L747-751. We suggest that the unique clinic features of SCC should be further studied to reveal the mechanism of poorer treatment outcome of EGFR-TKI therapy, and that a better treatment plan and more specific, potent targeted drugs for lung SCC need to be developed.
Collapse
Affiliation(s)
- Aadil Ahmed Memon
- State Key Laboratory of Microbial Metabolism, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University
| | | | - Ye Gu
- Endoscope Department, Shanghai Pulmonary Hospital, Tongji University School of Medicine
| | - Qian Luo
- Core Facility and Technical Service Center, School of Life Science and Biotechnology, Shanghai Jiao Tong University
| | - Jiajun Shi
- State Key Laboratory of Microbial Metabolism, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University
| | - Zixin Deng
- State Key Laboratory of Microbial Metabolism, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University
| | - Jian Ma
- Pneumology Department, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wei Ma
- State Key Laboratory of Microbial Metabolism, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University
| |
Collapse
|
15
|
Jain D, Iqbal S, Walia R, Malik P, Cyriac S, Mathur SR, Sharma MC, Madan K, Mohan A, Bhalla A, Pathy S, Kumar L, Guleria R. Evaluation of epidermal growth factor receptor mutations based on mutation specific immunohistochemistry in non-small cell lung cancer: A preliminary study. Indian J Med Res 2017; 143:308-14. [PMID: 27241644 PMCID: PMC4892077 DOI: 10.4103/0971-5916.182621] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND & OBJECTIVES Studies have shown that immunohistochemical (IHC) staining using epidermal growth factor receptor (EGFR) mutation specific antibodies, is an easy and cost-effective, screening method compared with molecular techniques. The purpose of present study was to assess the percentage positivity of IHC using EGFR mutation specific antibodies in lung biopsy samples from patients with primary lung adenocarcinoma (ADC). METHODS Two hundred and six biopsies of primary lung ADC were subjected to EGFR mutation specific antibodies against del E746-A750 and L858R. Detection of EGFR mutation done by high resolution melting analysis (HRM) was used as gold standard. A concordance was established between molecular and IHC results. Frequency of IHC positivity was assessed. RESULTS Of the 206 patients, 129 were male and 77 were female patients, with a mean age of 54.1 yr. Fifty five (26.6%) patients (36 men; 19 women) showed positivity for IHC of del E746-A750 (33) and L858R (22). HRM results were available in 14 patients which showed EGFR mutations in correspondence with del E746-750 or L858R in 64.2 per cent cases. Positive cases on HRM were further confirmed by DNA sequencing and fragment analysis. Three patients showed exon[20] variation. Two cases were negative for mutation. The genotype of del E746-750 mutation was more common than L858R. A concordance was established between molecular mutation and IHC in 85.7 per cent cases. INTERPRETATION & CONCLUSIONS In this preliminary study from India mutation specific IHC was used for assessment of mutation status of EGFR. Although the number tested was small, a good concordance was observed between molecular EGFR mutation and IHC expression. IHC methodology is a potentially useful tool to guide clinicians for personalized treatment in lung ADC, especially where facilities for molecular analysis are not readily available and for use in small biopsies where material is scant for molecular tests.
Collapse
Affiliation(s)
- Deepali Jain
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Sobuhi Iqbal
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Ritika Walia
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Prabhat Malik
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Sunu Cyriac
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Sandeep R Mathur
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Mehar C Sharma
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Karan Madan
- Department of Pulmonary Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Anant Mohan
- Department of Pulmonary Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Ashu Bhalla
- Department of Radiodiagnosis, All India Institute of Medical Sciences, New Delhi, India
| | - Sushmita Pathy
- Department of Radiation Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Lalit Kumar
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Randeep Guleria
- Department of Pulmonary Medicine, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
16
|
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: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
17
|
Performance Assessment of Epidermal Growth Factor Receptor Gene Sequencing According to Sample Size in Daily Practice Conditions. Appl Immunohistochem Mol Morphol 2017; 26:495-500. [PMID: 28248723 DOI: 10.1097/pai.0000000000000455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Lung carcinoma is the main cause of cancer death worldwide. Adenocarcinoma molecular biomarkers have been discovered, and targeted therapies have been developed with encouraging results. The epidermal growth factor receptor gene is one of these biomarkers. Exons 18 to 21 should be studied in patients with advanced adenocarcinoma, who are candidates for treatment with tyrosine kinase inhibitors. The objective was to compare the performance of the determination in large and small samples in daily practice conditions, trying to adjust to published consensus guidelines. A retrospective observational study of 141 cases was carried out, with exons 19 and 21 sequencing. Sample size (small vs. large), including number of satisfactory polymerase chain reaction (PCR), sequencing, deletions, and mutations, were evaluated. In small biopsies, sample type, fragment number, and percentage of tumor per sample were analyzed. The results shown 114/141 (80.8) cases that met selection criteria; 60/114 (53%) were large (surgical) and 54/114 (47%) were small samples (19/54 endoscopic, 17/54 fine needle aspiration clots, 4/54 lymph nodes, 14/54 core and other). All large samples were satisfactory PCR, 56/60 (93%) satisfactory sequencing, and 12/56 (21%) had deletions in exon 19. Small samples were satisfactory PCRs in 50/54 (93%) cases, and satisfactory sequencing in 35/50 (65%), 8/35 (23%) showed alterations in exon 19, and 1/35 (3%) in exon 21. In conclusion, the proportion of samples unfit for the study of the epidermal growth factor receptor gene mutational status increased from 7% in large samples to 35% in small ones. Nineteen small samples were inconclusive, with cell blocks predominating, 10/19 (53%).
Collapse
|
18
|
Han Y, Li J. Sample types applied for molecular diagnosis of therapeutic management of advanced non-small cell lung cancer in the precision medicine. ACTA ACUST UNITED AC 2017; 55:1817-1833. [DOI: 10.1515/cclm-2017-0112] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 04/07/2017] [Indexed: 12/17/2022]
Abstract
AbstractIn this era of precision medicine, molecular biology is becoming increasingly significant for the diagnosis and therapeutic management of non-small cell lung cancer. The specimen as the primary element of the whole testing flow is particularly important for maintaining the accuracy of gene alteration testing. Presently, the main sample types applied in routine diagnosis are tissue and cytology biopsies. Liquid biopsies are considered as the most promising alternatives when tissue and cytology samples are not available. Each sample type possesses its own strengths and weaknesses, pertaining to the disparity of sampling, preparation and preservation procedures, the heterogeneity of inter- or intratumors, the tumor cellularity (percentage and number of tumor cells) of specimens, etc., and none of them can individually be a “one size to fit all”. Therefore, in this review, we summarized the strengths and weaknesses of different sample types that are widely used in clinical practice, offered solutions to reduce the negative impact of the samples and proposed an optimized strategy for choice of samples during the entire diagnostic course. We hope to provide valuable information to laboratories for choosing optimal clinical specimens to achieve comprehensive functional genomic landscapes and formulate individually tailored treatment plans for NSCLC patients that are in advanced stages.
Collapse
|
19
|
Arreaza G, Qiu P, Pang L, Albright A, Hong LZ, Marton MJ, Levitan D. Pre-Analytical Considerations for Successful Next-Generation Sequencing (NGS): Challenges and Opportunities for Formalin-Fixed and Paraffin-Embedded Tumor Tissue (FFPE) Samples. Int J Mol Sci 2016; 17:ijms17091579. [PMID: 27657050 PMCID: PMC5037844 DOI: 10.3390/ijms17091579] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 09/08/2016] [Accepted: 09/13/2016] [Indexed: 12/11/2022] Open
Abstract
In cancer drug discovery, it is important to investigate the genetic determinants of response or resistance to cancer therapy as well as factors that contribute to adverse events in the course of clinical trials. Despite the emergence of new technologies and the ability to measure more diverse analytes (e.g., circulating tumor cell (CTC), circulating tumor DNA (ctDNA), etc.), tumor tissue is still the most common and reliable source for biomarker investigation. Because of its worldwide use and ability to preserve samples for many decades at ambient temperature, formalin-fixed, paraffin-embedded tumor tissue (FFPE) is likely to be the preferred choice for tissue preservation in clinical practice for the foreseeable future. Multiple analyses are routinely performed on the same FFPE samples (such as Immunohistochemistry (IHC), in situ hybridization, RNAseq, DNAseq, TILseq, Methyl-Seq, etc.). Thus, specimen prioritization and optimization of the isolation of analytes is critical to ensure successful completion of each assay. FFPE is notorious for producing suboptimal DNA quality and low DNA yield. However, commercial vendors tend to request higher DNA sample mass than what is actually required for downstream assays, which restricts the breadth of biomarker work that can be performed. We evaluated multiple genomics service laboratories to assess the current state of NGS pre-analytical processing of FFPE. Significant differences in pre-analytical capabilities were observed. Key aspects are highlighted and recommendations are made to improve the current practice in translational research.
Collapse
Affiliation(s)
- Gladys Arreaza
- Translational Medicine, Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ 07033, USA.
| | - Ping Qiu
- Translational Medicine, Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ 07033, USA.
| | - Ling Pang
- Translational Medicine, Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ 07033, USA.
| | - Andrew Albright
- Translational Medicine, Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ 07033, USA.
| | - Lewis Z Hong
- Translational Biomarkers, Merck Research Laboratories, Merck Sharp & Dohme, Singapore 609927, Singapore.
| | - Matthew J Marton
- Translational Medicine, Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ 07033, USA.
| | - Diane Levitan
- Translational Medicine, Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ 07033, USA.
| |
Collapse
|
20
|
|
21
|
Munch-Petersen HD, Asmar F, Dimopoulos K, Areškevičiūtė A, Brown P, Girkov MS, Pedersen A, Sjö LD, Heegaard S, Broholm H, Kristensen LS, Ralfkiaer E, Grønbæk K. TP53 hotspot mutations are predictive of survival in primary central nervous system lymphoma patients treated with combination chemotherapy. Acta Neuropathol Commun 2016; 4:40. [PMID: 27101868 PMCID: PMC4840983 DOI: 10.1186/s40478-016-0307-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Accepted: 03/27/2016] [Indexed: 12/18/2022] Open
Abstract
Primary central nervous system lymphoma (PCNSL) is an aggressive variant of diffuse large B-cell lymphoma (DLBCL) confined to the CNS. TP53 mutations (MUT-TP53) were investigated in the context of MIR34A/B/C- and DAPK promoter methylation status, and associated with clinical outcomes in PCNSL patients. In a total of 107 PCNSL patients clinical data were recorded, histopathology reassessed, and genetic and epigenetic aberrations of the p53-miR34-DAPK network studied. TP53 mutational status (exon 5-8), with structural classification of single nucleotide variations according to the IARC-TP53-Database, methylation status of MIR34A/B/C and DAPK, and p53-protein expression were assessed. The 57/107 (53.2 %) patients that were treated with combination chemotherapy +/- rituximab (CCT-treated) had a significantly better median overall survival (OS) (31.3 months) than patients treated with other regimens (high-dose methotrexate/whole brain radiation therapy, 6.0 months, or no therapy, 0.83 months), P < 0.0001. TP53 mutations were identified in 32/86 (37.2 %), among which 12 patients had hotspot/direct DNA contact mutations. CCT-treated patients with PCNSL harboring a hotspot/direct DNA contact MUT-TP53 (n = 9) had a significantly worse OS and progression free survival (PFS) compared to patients with non-hotspot/non-direct DNA contact MUT-TP53 or wild-type TP53 (median PFS 4.6 versus 18.2 or 45.7 months), P = 0.041 and P = 0.00076, respectively. Multivariate Cox regression analysis confirmed that hotspot/direct DNA contact MUT-TP53 was predictive of poor outcome in CCT-treated PCNSL patients, P = 0.012 and P = 0.008; HR: 1.86 and 1.95, for OS and PFS, respectively. MIR34A, MIR34B/C, and DAPK promoter methylation were detected in 53/93 (57.0 %), 80/84 (95.2 %), and 70/75 (93.3 %) of the PCNSL patients with no influence on survival. Combined MUT-TP53 and MIR34A methylation was associated with poor PFS (median 6.4 versus 38.0 months), P = 0.0070. This study suggests that disruption of the p53-pathway by MUT-TP53in hotspot/direct DNA contact codons is predictive of outcome in CCT-treated PCNSL patients, and concomitant MUT-TP53 and MIR34A methylation are associated with poor PFS.
Collapse
|
22
|
Comparison of the fecal microbiota of dholes high-throughput Illumina sequencing of the V3-V4 region of the 16S rRNA gene. Appl Microbiol Biotechnol 2016; 100:3577-86. [PMID: 26728019 DOI: 10.1007/s00253-015-7257-y] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 12/13/2015] [Accepted: 12/15/2015] [Indexed: 01/10/2023]
Abstract
Intestinal microbes are part of a complex ecosystem. They have a mutual relationship with the host and play an essential role in maintaining the host's health. To optimize the feeding strategies and improve the health status of the dhole, which is an endangered species, we analyzed the structure of fecal microbes in four captive dholes using high-throughput Illumina sequencing targeting the V3-V4 region of the 16S rRNA gene. The diversity indexes and rarefaction curves indicated high microbial diversity in the intestines of the four dholes. The average number of operational taxonomical units (OTUs) in the four samples was 1196, but the number of OTUs common to all libraries was 126, suggesting only a few dominant species. Phylogenetic analysis identified 19 prokaryotic phyla from the 16S rRNA gene sequences, of which only 5 phyla were core microbiota: Bacteroidetes (21.63-38.97 %), Firmicutes (20.97-44.01 %), Proteobacteria (9.33-17.60 %), Fusobacteria (9.11-17.90 %), and Actinobacteria (1.22-2.87 %). These five phyla accounted for 97 % of the bacteria in all the dholes apart from one, in which 78 % of the bacteria were from these phyla. The results of our study provide an effective theoretical basis from which to reach an understanding of the biological mechanisms relevant to the protection of this endangered species.
Collapse
|
23
|
|
24
|
Moens LNJ, Falk-Sörqvist E, Ljungström V, Mattsson J, Sundström M, La Fleur L, Mathot L, Micke P, Nilsson M, Botling J. HaloPlex Targeted Resequencing for Mutation Detection in Clinical Formalin-Fixed, Paraffin-Embedded Tumor Samples. J Mol Diagn 2015; 17:729-39. [PMID: 26354930 DOI: 10.1016/j.jmoldx.2015.06.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 05/29/2015] [Accepted: 06/17/2015] [Indexed: 12/13/2022] Open
Abstract
In recent years, the advent of massively parallel next-generation sequencing technologies has enabled substantial advances in the study of human diseases. Combined with targeted DNA enrichment methods, high sequence coverage can be obtained for different genes simultaneously at a reduced cost per sample, creating unique opportunities for clinical cancer diagnostics. However, the formalin-fixed, paraffin-embedded (FFPE) process of tissue samples, routinely used in pathology departments, results in DNA fragmentation and nucleotide modifications that introduce a number of technical challenges for downstream biomolecular analyses. We evaluated the HaloPlex target enrichment system for somatic mutation detection in 80 tissue fractions derived from 20 clinical cancer cases with paired tumor and normal tissue available in both FFPE and fresh-frozen format. Several modifications to the standard method were introduced, including a reduced target fragment length and two strand capturing. We found that FFPE material can be used for HaloPlex-based target enrichment and next-generation sequencing, even when starting from small amounts of DNA. By specifically capturing both strands for each target fragment, we were able to reduce the number of false-positive errors caused by FFPE-induced artifacts and lower the detection limit for somatic mutations. We believe that the HaloPlex method presented here will be broadly applicable as a tool for somatic mutation detection in clinical cancer settings.
Collapse
Affiliation(s)
- Lotte N J Moens
- Department of Immunology Genetics and Pathology, Uppsala University, Science for Life Laboratory, Uppsala, Sweden
| | - Elin Falk-Sörqvist
- Department of Immunology Genetics and Pathology, Uppsala University, Science for Life Laboratory, Uppsala, Sweden
| | - Viktor Ljungström
- Department of Immunology Genetics and Pathology, Uppsala University, Science for Life Laboratory, Uppsala, Sweden
| | - Johanna Mattsson
- Department of Immunology Genetics and Pathology, Uppsala University, Science for Life Laboratory, Uppsala, Sweden
| | - Magnus Sundström
- Department of Immunology Genetics and Pathology, Uppsala University, Science for Life Laboratory, Uppsala, Sweden
| | - Linnéa La Fleur
- Department of Immunology Genetics and Pathology, Uppsala University, Science for Life Laboratory, Uppsala, Sweden
| | - Lucy Mathot
- Department of Immunology Genetics and Pathology, Uppsala University, Science for Life Laboratory, Uppsala, Sweden
| | - Patrick Micke
- Department of Immunology Genetics and Pathology, Uppsala University, Science for Life Laboratory, Uppsala, Sweden
| | - Mats Nilsson
- Department of Immunology Genetics and Pathology, Uppsala University, Science for Life Laboratory, Uppsala, Sweden; Department of Biochemistry and Biophysics, Stockholm University, Science for Life Laboratory, Stockholm, Sweden.
| | - Johan Botling
- Department of Immunology Genetics and Pathology, Uppsala University, Science for Life Laboratory, Uppsala, Sweden.
| |
Collapse
|
25
|
da Cunha Santos G, Saieg MA. Preanalytic parameters in epidermal growth factor receptor mutation testing for non-small cell lung carcinoma: A review of cytologic series. Cancer Cytopathol 2015; 123:633-43. [PMID: 26288231 PMCID: PMC5042111 DOI: 10.1002/cncy.21595] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 07/03/2015] [Accepted: 07/07/2015] [Indexed: 12/31/2022]
Abstract
The results from molecular assays can be affected significantly by the preanalytic condition of cytologic samples. The authors review current knowledge on the use of cytologic samples for epidermal growth factor receptor (EGFR) mutation testing in non–small cell lung cancer with a focus on preanalytic parameters. A systematic electronic search of the MEDLINE database was performed to identify original articles that reported the use of cytologic samples for EGFR molecular analysis and included a minimum of 100 samples. The information collected included author(s), journal, and year of publication; number of patients and samples; sampling method; type of preparation; type of fixative; staining techniques; mutation analysis techniques; tumor cellularity; the percentage of tumor cells; data on DNA quantity, quality, and concentration; failed assays; and the mutation rate. EGFR mutation analysis was conducted on 4999 cytologic samples from 22 studies that fulfilled the inclusion criteria. Fine‐needle aspirates and pleural effusions were the most common types of specimens used. DNA was mainly extracted from cell blocks and smears, and the most commonly reported fixatives included formalin, ethanol, and CytoLyt. Cellularity assessments and DNA yields were available from 5 studies each. The average success rate for the assays that used cytologic specimens was 95.87% (range, 85.2%‐100%). The mutation rate ranged from 6% to 50.46%, and a higher mutation detection rate and lower numbers of insufficient cases were reported for pleural effusions and lymph node samples from endobronchial ultrasound‐guided transbronchial needle aspiration compared with histologic specimens. Low cellularity and a low percentage of tumor cells were associated with higher test failure rates. Future guidelines should consider the current data for specific recommendations regarding cytologic samples. Cancer (Cancer Cytopathol) 2015;123:633–643. © 2015 American Cancer Society. Preanalytic parameters for epidermal growth factor receptor mutation testing are reviewed in non–small cell lung cancer using 4999 cytologic samples from 22 studies. A higher mutation detection rate and lower numbers of insufficient cases are observed for pleural effusions and lymph node samples obtained using endobronchial ultrasound‐guided transbronchial needle aspiration compared with histologic specimens, and low cellularity and a lower percentage of tumor cells are associated with higher test failure rates. Future guidelines should consider the current data for specific recommendations regarding cytologic samples.
Collapse
Affiliation(s)
- Gilda da Cunha Santos
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Laboratory Medicine Program, University Health Network, Toronto, Ontario, Canada
| | - Mauro Ajaj Saieg
- Department of Pathology, Santa Casa Medical School, São Paulo, Brazil
| |
Collapse
|
26
|
|
27
|
Miles G, Rae J, Ramalingam SS, Pfeifer J. Genetic Testing and Tissue Banking for Personalized Oncology: Analytical and Institutional Factors. Semin Oncol 2015; 42:713-23. [PMID: 26433552 DOI: 10.1053/j.seminoncol.2015.07.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Personalized oncology, or more aptly precision oncogenomics, refers to the identification and implementation of clinically actionable targets tailored to an individual patient's cancer genomic information. Banking of human tissue and other biospecimens establishes a framework to extract and collect the data essential to our understanding of disease pathogenesis and treatment. Cancer cooperative groups in the United States have led the way in establishing robust biospecimen collection mechanisms to facilitate translational research, and combined with technological advances in molecular testing, tissue banking has expanded from its traditional base in academic research and is assuming an increasingly pivotal role in directing the clinical care of cancer patients. Comprehensive screening of tumors by DNA sequencing and the ability to mine and interpret these large data sets from well-organized tissue banks have defined molecular subtypes of cancer. Such stratification by genomic criteria has revolutionized our perspectives on cancer diagnosis and treatment, offering insight into prognosis, progression, and susceptibility or resistance to known therapeutic agents. In turn, this has enabled clinicians to offer treatments tailored to patients that can greatly improve their chances of survival. Unique challenges and opportunities accompany the rapidly evolving interplay between tissue banking and genomic sequencing, and are the driving forces underlying the revolution in precision medicine. Molecular testing and precision medicine clinical trials are now becoming the major thrust behind the cooperative groups' clinical research efforts.
Collapse
Affiliation(s)
- George Miles
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO.
| | - James Rae
- Department of Internal Medicine & Pharmacology, University of Michigan, Ann Arbor, MI
| | - Suresh S Ramalingam
- Department of Hematology and Medical Oncology, Emory School of Medicine, Winship Cancer Institute, Atlanta, GA
| | - John Pfeifer
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
28
|
Josefsen MH, Andersen SC, Christensen J, Hoorfar J. Microbial food safety: Potential of DNA extraction methods for use in diagnostic metagenomics. J Microbiol Methods 2015; 114:30-4. [PMID: 25937085 DOI: 10.1016/j.mimet.2015.04.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 04/30/2015] [Accepted: 04/30/2015] [Indexed: 12/26/2022]
Abstract
The efficiency of ten widely applied DNA extraction protocols was evaluated for suitability for diagnostic metagenomics. The protocols were selected based on a thorough literature study. Chicken fecal samples inoculated with about 1×10(3) and 1×10(6) CFU/g Campylobacter jejuni were used as a model. The evaluation was performed based on total DNA yield measured by fluorometry, and quality and quantity of C. jejuni DNA measured by real-time PCR. There was up to a 25-fold variance between the lowest (NucliSens miniMAG, BIOMÉRIEUX) and highest (PowerLyzer PowerSoil DNA Isolation Kit, MO BIO Laboratories) yielding protocols. The PowerLyzer PowerSoil DNA Isolation Kit performed significantly better than all other protocols tested. Selected protocols were modified, i.e., extended heating and homogenization, resulting in increased yields of total DNA. For QIAamp Fast DNA Stool Mini Kit (Qiagen) a 7-fold increase in total DNA was observed following the protocol for human DNA analysis and including a 5 min heating step at 70°C. For the PowerLyzer PowerSoil and the PowerFecal DNA Isolation Kit (MO BIO Laboratories) the total DNA fold increase was 1.6 to 1.8 when including an extra 10 min of bead-vortexing. There was no correlation between the yield of total DNA and the amount of PCR-amplifiable DNA from C. jejuni. The protocols resulting in the highest yield of total DNA did not show correspondingly increased levels of C. jejuni DNA as determined by PCR. In conclusion, substantial variation in the efficiency of the protocols to extract DNA was observed. The highest DNA yield was obtained with the PowerLyzer PowerSoil DNA Isolation Kit, whereas the FastDNA SPIN Kit for Feces (MP Biomedicals) resulted in the highest amount of PCR-amplifiable C. jejuni DNA.
Collapse
Affiliation(s)
- Mathilde H Josefsen
- Technical University of Denmark, National Food Institute, Mørkhøj Bygade 19, DK-2860 Søborg, Denmark
| | - Sandra C Andersen
- Technical University of Denmark, National Food Institute, Mørkhøj Bygade 19, DK-2860 Søborg, Denmark
| | - Julia Christensen
- Technical University of Denmark, National Food Institute, Mørkhøj Bygade 19, DK-2860 Søborg, Denmark
| | - Jeffrey Hoorfar
- Technical University of Denmark, National Food Institute, Mørkhøj Bygade 19, DK-2860 Søborg, Denmark.
| |
Collapse
|
29
|
Heydt C, Kumm N, Fassunke J, Künstlinger H, Ihle MA, Scheel A, Schildhaus HU, Haller F, Büttner R, Odenthal M, Wardelmann E, Merkelbach-Bruse S. Massively parallel sequencing fails to detect minor resistant subclones in tissue samples prior to tyrosine kinase inhibitor therapy. BMC Cancer 2015; 15:291. [PMID: 25886408 PMCID: PMC4404105 DOI: 10.1186/s12885-015-1311-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Accepted: 04/01/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Personalised medicine and targeted therapy have revolutionised cancer treatment. However, most patients develop drug resistance and relapse after showing an initial treatment response. Two theories have been postulated; either secondary resistance mutations develop de novo during therapy by mutagenesis or they are present in minor subclones prior to therapy. In this study, these two theories were evaluated in gastrointestinal stromal tumours (GISTs) where most patients develop secondary resistance mutations in the KIT gene during therapy with tyrosine kinase inhibitors. METHODS We used a cohort of 33 formalin-fixed, paraffin embedded (FFPE) primary GISTs and their corresponding recurrent tumours with known mutational status. The primary tumours were analysed for the secondary mutations of the recurrences, which had been identified previously. The primary tumours were resected prior to tyrosine kinase inhibitor therapy. Three ultrasensitive, massively parallel sequencing approaches on the GS Junior (Roche, Mannheim, Germany) and the MiSeq(TM) (Illumina, San Diego, CA, USA) were applied. Additionally, nine fresh-frozen samples resected prior to therapy were analysed for the most common secondary resistance mutations. RESULTS With a sensitivity level of down to 0.02%, no pre-existing resistant subclones with secondary KIT mutations were detected in primary GISTs. The sensitivity level varied for individual secondary mutations and was limited by sequencing artefacts on both systems. Artificial T > C substitutions at the position of the exon 13 p.V654A mutation, in particular, led to a lower sensitivity, independent from the source of the material. Fresh-frozen samples showed the same range of artificially mutated allele frequencies as the FFPE material. CONCLUSIONS Although we achieved a sufficiently high level of sensitivity, neither in the primary FFPE nor in the fresh-frozen GISTs we were able to detect pre-existing resistant subclones of the corresponding known secondary resistance mutations of the recurrent tumours. This supports the theory that secondary KIT resistance mutations develop under treatment by "de novo" mutagenesis. Alternatively, the detection limit of two mutated clones in 10,000 wild-type clones might not have been high enough or heterogeneous tissue samples, per se, might not be suitable for the detection of very small subpopulations of mutated cells.
Collapse
Affiliation(s)
- Carina Heydt
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany.
| | - Niklas Kumm
- Institute of Pathology, University Hospital Erlangen, Krankenhausstraße 8-10, 91054, Erlangen, Germany.
| | - Jana Fassunke
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany.
| | - Helen Künstlinger
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany.
| | - Michaela Angelika Ihle
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany.
| | - Andreas Scheel
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany.
| | - Hans-Ulrich Schildhaus
- Institute of Pathology, University Hospital Göttingen, Robert-Koch-Strasse 40, 37075, Göttingen, Germany.
| | - Florian Haller
- Institute of Pathology, University Hospital Erlangen, Krankenhausstraße 8-10, 91054, Erlangen, Germany.
| | - Reinhard Büttner
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany.
| | - Margarete Odenthal
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany.
| | - Eva Wardelmann
- Gerhard-Domagk-Institute of Pathology, University Hospital Münster, Albert-Schweitzer-Campus 1, Gebäude D17, 48149, Münster, Germany.
| | - Sabine Merkelbach-Bruse
- Institute of Pathology, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany.
| |
Collapse
|
30
|
Mojica WD, Oh KW, Lee H, Furlani EP, Sands AM. Maximizing derivable information from cytologic specimens for pathologic and molecular diagnostics. J Am Soc Cytopathol 2015; 4:141-147. [PMID: 31051695 DOI: 10.1016/j.jasc.2015.01.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 01/14/2015] [Accepted: 01/21/2015] [Indexed: 11/27/2022]
Abstract
INTRODUCTION The advent of precision medicine will increase the demand for molecular testing on patient tumor specimens. Cytology specimens have been shown to be ideal substrates for molecular testing, but their often paucicellular nature can lead to conflicts in prioritizing sample management. A microfluidic platform was investigated to determine whether cytologic and molecular data could be procured from the same cells, obviating the need for partitioning a sample by multiplexing it instead. MATERIALS AND METHODS Cytology samples were created from a tissue source, stained with a supravital dye, and enriched using immunomagnetic beads. These cells and the attached immunomagnetic beads were then run through a microfluidic channel, temporarily immobilized for cytologic examination, and then recovered. The cytologic characteristics of these cells was compared with cells from the same source prepared by conventional cytologic preparatory means. DNA was extracted from the cells recovered from the microfluidic channel and the nature of their integrity was assessed. RESULTS Cytologic features between cells run in a microfluidic channel and prepared by conventional means were similar. The DNA recovered from the cells run through the microfluidic channel was of high molecular weight. CONCLUSIONS Microfluidics enables multiplex testing of cytologic specimens, allowing for cytology-based diagnostic examination and recovery of high-quality DNA. This approach will be of particular benefit for cytology specimens that are paucicellular and will need molecular testing.
Collapse
Affiliation(s)
- Wilfrido D Mojica
- Department of Pathology and Anatomical Sciences, University at Buffalo, The State University of New York, 100 High Street, Buffalo, New York.
| | - Kwang W Oh
- Department of Electrical Engineering, University at Buffalo, The State University of New York, Buffalo, New York
| | - Hun Lee
- Department of Electrical Engineering, University at Buffalo, The State University of New York, Buffalo, New York
| | - Edward P Furlani
- Department of Electrical Engineering, University at Buffalo, The State University of New York, Buffalo, New York; Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, New York
| | - Amy M Sands
- Department of Pathology and Anatomical Sciences, University at Buffalo, The State University of New York, 100 High Street, Buffalo, New York
| |
Collapse
|
31
|
Do H, Dobrovic A. Sequence Artifacts in DNA from Formalin-Fixed Tissues: Causes and Strategies for Minimization. Clin Chem 2015; 61:64-71. [DOI: 10.1373/clinchem.2014.223040] [Citation(s) in RCA: 331] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Abstract
BACKGROUND
Precision medicine is dependent on identifying actionable mutations in tumors. Accurate detection of mutations is often problematic in formalin-fixed paraffin-embedded (FFPE) tissues. DNA extracted from formalin-fixed tissues is fragmented and also contains DNA lesions that are the sources of sequence artifacts. Sequence artifacts can be difficult to distinguish from true mutations, especially in the context of tumor heterogeneity, and are an increasing interpretive problem in this era of massively parallel sequencing. Understanding of the sources of sequence artifacts in FFPE tissues and implementation of preventative strategies are critical to improve the accurate detection of actionable mutations.
CONTENT
This mini-review focuses on DNA template lesions in FFPE tissues as the source of sequence artifacts in molecular analysis. In particular, fragmentation, base modification (including uracil and thymine deriving from cytosine deamination), and abasic sites are discussed as indirect or direct sources of sequence artifacts. We discuss strategies that can be implemented to minimize sequence artifacts and to distinguish true mutations from sequence artifacts. These strategies are applicable for the detection of actionable mutations in both single amplicon and massively parallel amplicon sequencing approaches.
SUMMARY
Because FFPE tissues are usually the only available material for DNA analysis, it is important to maximize the accurate informational content from FFPE DNA. Careful consideration of each step in the work flow is needed to minimize sequence artifacts. In addition, validation of actionable mutations either by appropriate experimental design or by orthogonal methods should be considered.
Collapse
Affiliation(s)
- Hongdo Do
- Translational Genomics and Epigenomics Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- Department of Pathology, University of Melbourne, Parkville, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Alexander Dobrovic
- Translational Genomics and Epigenomics Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- Department of Pathology, University of Melbourne, Parkville, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| |
Collapse
|
32
|
Roberts JD, Longoria J, Poon A, Gollob MH, Dewland TA, Kwok PY, Olgin JE, Deo RC, Marcus GM. Targeted deep sequencing reveals no definitive evidence for somatic mosaicism in atrial fibrillation. ACTA ACUST UNITED AC 2014; 8:50-7. [PMID: 25406240 DOI: 10.1161/circgenetics.114.000650] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Studies of ≤15 atrial fibrillation (AF) patients have identified atrial-specific mutations within connexin genes, suggesting that somatic mutations may account for sporadic cases of the arrhythmia. We sought to identify atrial somatic mutations among patients with and without AF using targeted deep next-generation sequencing of 560 genes, including genetic culprits implicated in AF, the Mendelian cardiomyopathies and channelopathies, and all ion channels within the genome. METHODS AND RESULTS Targeted gene capture and next-generation sequencing were performed on DNA from lymphocytes and left atrial appendages of 34 patients (25 with AF). Twenty AF patients had undergone cardiac surgery exclusively for pulmonary vein isolation and 17 had no structural heart disease. Sequence alignment and variant calling were performed for each atrial-lymphocyte pair using the Burrows-Wheeler Aligner, the Genome Analysis Toolkit, and MuTect packages. Next-generation sequencing yielded a median 265-fold coverage depth (interquartile range, 64-369). Comparison of the 3 million base pairs from each atrial-lymphocyte pair revealed a single potential somatic missense mutation in 3 AF patients and 2 in a single control (12 versus 11%; P=1). All potential discordant variants had low allelic fractions (range, 2.3%-7.3%) and none were detected with conventional sequencing. CONCLUSIONS Using high-depth next-generation sequencing and state-of-the art somatic mutation calling approaches, no pathogenic atrial somatic mutations could be confirmed among 25 AF patients in a comprehensive cardiac arrhythmia genetic panel. These findings indicate that atrial-specific mutations are rare and that somatic mosaicism is unlikely to exert a prominent role in AF pathogenesis.
Collapse
Affiliation(s)
- Jason D Roberts
- From the Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine (J.D.R., T.A.D., J.E.O., G.M.M.), Cardiovascular Research Institute (A.P., P.-Y.K., R.C.D.), and Department of Medicine, California Institute for Quantitative Biosciences, Institute for Human Genetics (R.C.D.), University of California San Francisco; Division of Cardiovascular Surgery, Sutter Health, Sacramento, CA (J.L.); and Arrhythmia Research Laboratory, University of Ottawa Heart Institute, Ottawa, Ontario, Canada (M.H.G.)
| | - James Longoria
- From the Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine (J.D.R., T.A.D., J.E.O., G.M.M.), Cardiovascular Research Institute (A.P., P.-Y.K., R.C.D.), and Department of Medicine, California Institute for Quantitative Biosciences, Institute for Human Genetics (R.C.D.), University of California San Francisco; Division of Cardiovascular Surgery, Sutter Health, Sacramento, CA (J.L.); and Arrhythmia Research Laboratory, University of Ottawa Heart Institute, Ottawa, Ontario, Canada (M.H.G.)
| | - Annie Poon
- From the Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine (J.D.R., T.A.D., J.E.O., G.M.M.), Cardiovascular Research Institute (A.P., P.-Y.K., R.C.D.), and Department of Medicine, California Institute for Quantitative Biosciences, Institute for Human Genetics (R.C.D.), University of California San Francisco; Division of Cardiovascular Surgery, Sutter Health, Sacramento, CA (J.L.); and Arrhythmia Research Laboratory, University of Ottawa Heart Institute, Ottawa, Ontario, Canada (M.H.G.)
| | - Michael H Gollob
- From the Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine (J.D.R., T.A.D., J.E.O., G.M.M.), Cardiovascular Research Institute (A.P., P.-Y.K., R.C.D.), and Department of Medicine, California Institute for Quantitative Biosciences, Institute for Human Genetics (R.C.D.), University of California San Francisco; Division of Cardiovascular Surgery, Sutter Health, Sacramento, CA (J.L.); and Arrhythmia Research Laboratory, University of Ottawa Heart Institute, Ottawa, Ontario, Canada (M.H.G.)
| | - Thomas A Dewland
- From the Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine (J.D.R., T.A.D., J.E.O., G.M.M.), Cardiovascular Research Institute (A.P., P.-Y.K., R.C.D.), and Department of Medicine, California Institute for Quantitative Biosciences, Institute for Human Genetics (R.C.D.), University of California San Francisco; Division of Cardiovascular Surgery, Sutter Health, Sacramento, CA (J.L.); and Arrhythmia Research Laboratory, University of Ottawa Heart Institute, Ottawa, Ontario, Canada (M.H.G.)
| | - Pui-Yan Kwok
- From the Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine (J.D.R., T.A.D., J.E.O., G.M.M.), Cardiovascular Research Institute (A.P., P.-Y.K., R.C.D.), and Department of Medicine, California Institute for Quantitative Biosciences, Institute for Human Genetics (R.C.D.), University of California San Francisco; Division of Cardiovascular Surgery, Sutter Health, Sacramento, CA (J.L.); and Arrhythmia Research Laboratory, University of Ottawa Heart Institute, Ottawa, Ontario, Canada (M.H.G.)
| | - Jeffrey E Olgin
- From the Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine (J.D.R., T.A.D., J.E.O., G.M.M.), Cardiovascular Research Institute (A.P., P.-Y.K., R.C.D.), and Department of Medicine, California Institute for Quantitative Biosciences, Institute for Human Genetics (R.C.D.), University of California San Francisco; Division of Cardiovascular Surgery, Sutter Health, Sacramento, CA (J.L.); and Arrhythmia Research Laboratory, University of Ottawa Heart Institute, Ottawa, Ontario, Canada (M.H.G.)
| | - Rahul C Deo
- From the Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine (J.D.R., T.A.D., J.E.O., G.M.M.), Cardiovascular Research Institute (A.P., P.-Y.K., R.C.D.), and Department of Medicine, California Institute for Quantitative Biosciences, Institute for Human Genetics (R.C.D.), University of California San Francisco; Division of Cardiovascular Surgery, Sutter Health, Sacramento, CA (J.L.); and Arrhythmia Research Laboratory, University of Ottawa Heart Institute, Ottawa, Ontario, Canada (M.H.G.).
| | - Gregory M Marcus
- From the Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine (J.D.R., T.A.D., J.E.O., G.M.M.), Cardiovascular Research Institute (A.P., P.-Y.K., R.C.D.), and Department of Medicine, California Institute for Quantitative Biosciences, Institute for Human Genetics (R.C.D.), University of California San Francisco; Division of Cardiovascular Surgery, Sutter Health, Sacramento, CA (J.L.); and Arrhythmia Research Laboratory, University of Ottawa Heart Institute, Ottawa, Ontario, Canada (M.H.G.).
| |
Collapse
|
33
|
Heydt C, Fassunke J, Künstlinger H, Ihle MA, König K, Heukamp LC, Schildhaus HU, Odenthal M, Büttner R, Merkelbach-Bruse S. Comparison of pre-analytical FFPE sample preparation methods and their impact on massively parallel sequencing in routine diagnostics. PLoS One 2014; 9:e104566. [PMID: 25105902 PMCID: PMC4126727 DOI: 10.1371/journal.pone.0104566] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 07/11/2014] [Indexed: 11/19/2022] Open
Abstract
Over the last years, massively parallel sequencing has rapidly evolved and has now transitioned into molecular pathology routine laboratories. It is an attractive platform for analysing multiple genes at the same time with very little input material. Therefore, the need for high quality DNA obtained from automated DNA extraction systems has increased, especially to those laboratories which are dealing with formalin-fixed paraffin-embedded (FFPE) material and high sample throughput. This study evaluated five automated FFPE DNA extraction systems as well as five DNA quantification systems using the three most common techniques, UV spectrophotometry, fluorescent dye-based quantification and quantitative PCR, on 26 FFPE tissue samples. Additionally, the effects on downstream applications were analysed to find the most suitable pre-analytical methods for massively parallel sequencing in routine diagnostics. The results revealed that the Maxwell 16 from Promega (Mannheim, Germany) seems to be the superior system for DNA extraction from FFPE material. The extracts had a 1.3-24.6-fold higher DNA concentration in comparison to the other extraction systems, a higher quality and were most suitable for downstream applications. The comparison of the five quantification methods showed intermethod variations but all methods could be used to estimate the right amount for PCR amplification and for massively parallel sequencing. Interestingly, the best results in massively parallel sequencing were obtained with a DNA input of 15 ng determined by the NanoDrop 2000c spectrophotometer (Thermo Fisher Scientific, Waltham, MA, USA). No difference could be detected in mutation analysis based on the results of the quantification methods. These findings emphasise, that it is particularly important to choose the most reliable and constant DNA extraction system, especially when using small biopsies and low elution volumes, and that all common DNA quantification techniques can be used for downstream applications like massively parallel sequencing.
Collapse
Affiliation(s)
- Carina Heydt
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
- * E-mail:
| | - Jana Fassunke
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Helen Künstlinger
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | | | - Katharina König
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | | | | | | | - Reinhard Büttner
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | | |
Collapse
|
34
|
Prendeville S, Brosnan T, Browne TJ, McCarthy J. Automated Cellient(™) cytoblocks: better, stronger, faster? Cytopathology 2014; 25:372-80. [PMID: 24943912 DOI: 10.1111/cyt.12159] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2014] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Cytoblocks (CBs), or cell blocks, provide additional morphological detail and a platform for immunocytochemistry (ICC) in cytopathology. The Cellient(™) system produces CBs in 45 minutes using methanol fixation, compared with traditional CBs, which require overnight formalin fixation. This study compares Cellient and traditional CB methods in terms of cellularity, morphology and immunoreactivity, evaluates the potential to add formalin fixation to the Cellient method for ICC studies and determines the optimal sectioning depth for maximal cellularity in Cellient CBs. METHODS One hundred and sixty CBs were prepared from 40 cytology samples (32 malignant, eight benign) using four processing methods: (A) traditional; (B) Cellient (methanol fixation); (C) Cellient using additional formalin fixation for 30 minutes; (D) Cellient using additional formalin fixation for 60 minutes. Haematoxylin and eosin-stained sections were assessed for cellularity and morphology. ICC was assessed on 14 cases with a panel of antibodies. Three additional Cellient samples were serially sectioned to determine the optimal sectioning depth. Scoring was performed by two independent, blinded reviewers. RESULTS For malignant cases, morphology was superior with Cellient relative to traditional CBs (P < 0.001). Cellularity was comparable across all methods. ICC was excellent in all groups and the addition of formalin at any stage during the Cellient process did not influence the staining quality. Serial sectioning through Cellient CBs showed optimum cellularity at 30-40 μm with at least 27 sections obtainable. CONCLUSIONS Cellient CBs provide superior morphology to traditional CBs and, if required, formalin fixation may be added to the Cellient process for ICC. Optimal Cellient CB cellularity is achieved at 30-40 μm, which will impact on the handling of cases in daily practice.
Collapse
Affiliation(s)
- S Prendeville
- Department of Cytopathology, Cork University Hospital, Wilton, Cork, Ireland
| | | | | | | |
Collapse
|
35
|
Mansuet-Lupo A, Zouiti F, Alifano M, Tallet A, Charpentier MC, Ducruit V, Devez F, Lemaitre F, Laurent-Puig P, Damotte D, Blons H. Intratumoral distribution of EGFR mutations and copy number in metastatic lung cancer, what impact on the initial molecular diagnosis? J Transl Med 2014; 12:131. [PMID: 24885034 PMCID: PMC4041917 DOI: 10.1186/1479-5876-12-131] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 04/07/2014] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Activating epidermal growth factor receptor (EGFR) mutations characterize a subgroup of non-small-cell lung cancer that benefit from first line EGFR tyrosine kinase inhibitors (EGFR-TKI). However, the existence of polyclonal cell populations may hinder personalized-medicine strategies as patients' screening often depends upon a single tumor-biopsy sample. The purpose of this study is to clarify and to validate in clinical testing conditions the accuracy of EGFR genotyping using different tumor sites and various types of samples (transthoracic, surgical or endoscopic biopsies and cytology specimens). METHODS We conducted a retrospective review of 357 consecutive patients addressed for EGFR mutation screening in accordance with the directive of the European Medicines Agency (stage IV NSCLC). Fifty-seven samples were EGFR mutated and 40 had adequate tumor specimens for analysis on multiple spatially separated sites. Ten wild type samples were also analyzed. A total of 153 and 39 tumor fragments, from mutated and non-mutated cases respectively, were generated to analyze tumor heterogeneity or primary-metastatic discordances. After histological review of all fragments, EGFR genotyping was assessed using the routine diagnostic tools: fragment analysis for insertions and deletions and allele specific TaqMan probes for point mutations. EGFR copy number (CN) was evaluated by qPCR using TaqMan probes. RESULTS The identification of EGFR mutations was independent of localization within primary tumor, of specimen type and consistent between primary and metastases. At the opposite, for half of the samples, tumor loci showed different EGFR copy number that may affect mutation detection cut-off. CONCLUSIONS This is the largest series reporting multiple EGFR testing in Caucasians. It validates the accuracy of EGFR mutation screening from single tumor-biopsy samples before first line EGFR-TKI. The unpredictable variability in EGFR CN and therefore in EGFR wild type/mutant allelic ratio justifies the implementation of sensitive methods to identify patients with EGFR mutated tumors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Hélène Blons
- Université Paris Descartes, Sorbonne, Paris cité, France.
| |
Collapse
|
36
|
Results of the First Italian External Quality Assurance Scheme for somatic EGFR mutation testing in non-small-cell lung cancer. J Thorac Oncol 2014; 8:773-8. [PMID: 23575414 DOI: 10.1097/jto.0b013e31828c2b08] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION The Italian Association of Medical Oncology (AIOM) and the Italian Society of Pathology and Cytology organized an external quality assessment (EQA) scheme for EGFR mutation testing in non-small-cell lung cancer. METHODS Ten specimens, including three small biopsies with known epidermal growth factor receptor (EGFR) mutation status, were validated in three referral laboratories and provided to 47 participating centers. The participants were requested to perform mutational analysis, using their usual method, and to submit results within a 4-week time frame. According to a predefined scoring system, two points were assigned to correct genotype and zero points to false-negative or false-positive results. The threshold to pass the EQA was set at higher than 18 of 20 points. Two rounds were preplanned. RESULTS All participating centers submitted the results within the time frame. Polymerase chain reaction (PCR)/sequencing was the main methodology used (n = 37 laboratories), although a few centers did use pyrosequencing (n = 8) or real-time PCR (n = 2). A significant number of analytical errors were observed (n = 20), with a high frequency of false-positive results (n = 16). The lower scores were obtained for the small biopsies. Fourteen of 47 centers (30%) that did not pass the first round, having a score less than or equal to 18 points, used PCR/sequencing, whereas 10 of 10 laboratories, using pyrosequencing or real-time PCR, passed the first round. Eight laboratories passed the second round. Overall, 41of 47 centers (87%) passed the EQA. CONCLUSION The results of the EQA for EGFR testing in non-small-cell lung cancer suggest that good quality EGFR mutational analysis is performed in Italian laboratories, although differences between testing methods were observed, especially for small biopsies.
Collapse
|
37
|
Robasky K, Lewis NE, Church GM. The role of replicates for error mitigation in next-generation sequencing. Nat Rev Genet 2013; 15:56-62. [PMID: 24322726 DOI: 10.1038/nrg3655] [Citation(s) in RCA: 199] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Advances in next-generation sequencing (NGS) technologies have rapidly improved sequencing fidelity and substantially decreased sequencing error rates. However, given that there are billions of nucleotides in a human genome, even low experimental error rates yield many errors in variant calls. Erroneous variants can mimic true somatic and rare variants, thus requiring costly confirmatory experiments to minimize the number of false positives. Here, we discuss sources of experimental errors in NGS and how replicates can be used to abate such errors.
Collapse
Affiliation(s)
- Kimberly Robasky
- 1] Program in Bioinformatics, Boston University, Massachusetts 02115, USA.Department of Genetics, Harvard Medical School, and the Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA. Present address: Expression Analysis, a Quintiles Company, Durham, North Carolina 27713, USA. [2]
| | - Nathan E Lewis
- 1] Department of Genetics, Harvard Medical School, and the Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA. Department of Biology, Brigham Young University, Provo, Utah 84602, USA. Present address: Division of Pediatric Pharmacology and Drug Discovery, University of California, San Diego School of Medicine, La Jolla, California 92093, USA. [2]
| | - George M Church
- Department of Genetics, Harvard Medical School, and the Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
| |
Collapse
|
38
|
Rossi G, Graziano P, Leone A, Migaldi M, Califano R. The role of molecular analyses in the diagnosis and treatment of non-small-cell lung carcinomas. Semin Diagn Pathol 2013; 30:298-312. [PMID: 24342286 DOI: 10.1053/j.semdp.2013.11.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Non-small-cell lung cancer (NSCLC) subtyping has recently been a key factor in determining patient management with novel drugs. In addition, the identification of distinct oncogenic driver mutations frequently associated with NSCLC histotype and coupled to the clinical responses to targeted therapies have revolutionized the impact of histologic type and molecular biomarkers in lung cancer. Several molecular alterations involving different genes (EGFR, KRAS, ALK, BRAF, and HER2) seem to have a remarkable predilection for adenocarcinoma and specific inhibitors of EGFR and ALK are now available for patients with adenocarcinoma harboring the relevant gene alterations. The efficacy of histology-based and molecular-targeted therapies had a deep impact in (1) re-defining classification of lung cancer (particularly adenocarcinomas) and (2) routine clinical practice of pathologists involved in optimization of handling of tissue samples in order to guarantee NSCLC subtyping with the help of immunohistochemistry and adequately preserve tumor cells for molecular analysis. In agreement with the modern multidisciplinary approach to lung cancer, we reviewed here the diagnostic and predictive value of molecular biomarkers according to the clinical, pathologic, and molecular biologist viewpoints.
Collapse
Affiliation(s)
- Giulio Rossi
- Department of Pathology, IRCCS Arcispedale Santa Maria Nuova, viale Risorgimento, 80 42100, Reggio Emilia, Italy; Section of Pathologic Anatomy, University of Modena and Reggio Emilia, Modena, Italy.
| | - Paolo Graziano
- Unit of Pathology San Camillo Hospital, Malpighi Pavillon, Rome, Italy
| | - Alvaro Leone
- Unit of Pathology San Camillo Hospital, Malpighi Pavillon, Rome, Italy
| | - Mario Migaldi
- Section of Pathologic Anatomy, University of Modena and Reggio Emilia, Modena, Italy
| | - Raffaele Califano
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, UK; Department of Medical Oncology, University Hospital of South Manchester, Southmoor Road, Manchester, UK
| |
Collapse
|
39
|
Sah S, Chen L, Houghton J, Kemppainen J, Marko AC, Zeigler R, Latham GJ. Functional DNA quantification guides accurate next-generation sequencing mutation detection in formalin-fixed, paraffin-embedded tumor biopsies. Genome Med 2013; 5:77. [PMID: 24001039 PMCID: PMC3978876 DOI: 10.1186/gm481] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 08/23/2013] [Indexed: 11/25/2022] Open
Abstract
The formalin-fixed, paraffin-embedded (FFPE) biopsy is a challenging sample for molecular assays such as targeted next-generation sequencing (NGS). We compared three methods for FFPE DNA quantification, including a novel PCR assay (‘QFI-PCR’) that measures the absolute copy number of amplifiable DNA, across 165 residual clinical specimens. The results reveal the limitations of commonly used approaches, and demonstrate the value of an integrated workflow using QFI-PCR to improve the accuracy of NGS mutation detection and guide changes in input that can rescue low quality FFPE DNA. These findings address a growing need for improved quality measures in NGS-based patient testing.
Collapse
|
40
|
Lindeman NI, Cagle PT, Beasley MB, Chitale DA, Dacic S, Giaccone G, Jenkins RB, Kwiatkowski DJ, Saldivar JS, Squire J, Thunnissen E, Ladanyi M, College of American Pathologists International Association for the Study of Lung Cancer and Association for Molecular Pathology. Molecular testing guideline for selection of lung cancer patients for EGFR and ALK tyrosine kinase inhibitors: guideline from the College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Pathology. J Mol Diagn 2013; 15:415-53. [PMID: 23562183 DOI: 10.1016/j.jmoldx.2013.03.001] [Citation(s) in RCA: 343] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 02/12/2013] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE To establish evidence-based recommendations for the molecular analysis of lung cancers that are required to guide EGFR- and ALK-directed therapies, addressing which patients and samples should be tested, and when and how testing should be performed. PARTICIPANTS Three cochairs without conflicts of interest were selected, one from each of the 3 sponsoring professional societies: College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Pathology. Writing and advisory panels were constituted from additional experts from these societies. EVIDENCE Three unbiased literature searches of electronic databases were performed to capture published articles from January 2004 through February 2012, yielding 1533 articles whose abstracts were screened to identify 521 pertinent articles that were then reviewed in detail for their relevance to the recommendations. EVIDENCE was formally graded for each recommendation. CONSENSUS PROCESS Initial recommendations were formulated by the cochairs and panel members at a public meeting. Each guideline section was assigned to at least 2 panelists. Drafts were circulated to the writing panel (version 1), advisory panel (version 2), and the public (version 3) before submission (version 4). CONCLUSIONS The 37 guideline items address 14 subjects, including 15 recommendations (evidence grade A/B). The major recommendations are to use testing for EGFR mutations and ALK fusions to guide patient selection for therapy with an epidermal growth factor receptor (EGFR) or anaplastic lymphoma kinase (ALK) inhibitor, respectively, in all patients with advanced-stage adenocarcinoma, regardless of sex, race, smoking history, or other clinical risk factors, and to prioritize EGFR and ALK testing over other molecular predictive tests. As scientific discoveries and clinical practice outpace the completion of randomized clinical trials, evidence-based guidelines developed by expert practitioners are vital for communicating emerging clinical standards. Already, new treatments targeting genetic alterations in other, less common driver oncogenes are being evaluated in lung cancer, and testing for these may be addressed in future versions of these guidelines.
Collapse
Affiliation(s)
- Neal I Lindeman
- Department of Pathology, Brigham & Women's Hospital, Boston, MA 02115-6110, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Lindeman NI, Cagle PT, Beasley MB, Chitale DA, Dacic S, Giaccone G, Jenkins RB, Kwiatkowski DJ, Saldivar JS, Squire J, Thunnissen E, Ladanyi M. Molecular testing guideline for selection of lung cancer patients for EGFR and ALK tyrosine kinase inhibitors: guideline from the College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Pathology. J Thorac Oncol 2013; 8:823-59. [PMID: 23552377 PMCID: PMC4159960 DOI: 10.1097/jto.0b013e318290868f] [Citation(s) in RCA: 616] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To establish evidence-based recommendations for the molecular analysis of lung cancers that are that are required to guide EGFR- and ALK-directed therapies, addressing which patients and samples should be tested, and when and how testing should be performed. PARTICIPANTS Three cochairs without conflicts of interest were selected, one from each of the 3 sponsoring professional societies: College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Pathology. Writing and advisory panels were constituted from additional experts from these societies. EVIDENCE Three unbiased literature searches of electronic databases were performed to capture articles published published from January 2004 through February 2012, yielding 1533 articles whose abstracts were screened to identify 521 pertinent articles that were then reviewed in detail for their relevance to the recommendations. Evidence was formally graded for each recommendation. CONSENSUS PROCESS Initial recommendations were formulated by the cochairs and panel members at a public meeting. Each guideline section was assigned to at least 2 panelists. Drafts were circulated to the writing panel (version 1), advisory panel (version 2), and the public (version 3) before submission (version 4). CONCLUSIONS The 37 guideline items address 14 subjects, including 15 recommendations (evidence grade A/B). The major recommendations are to use testing for EGFR mutations and ALK fusions to guide patient selection for therapy with an epidermal growth factor receptor (EGFR) or anaplastic lymphoma kinase (ALK) inhibitor, respectively, in all patients with advanced-stage adenocarcinoma, regardless of sex, race, smoking history, or other clinical risk factors, and to prioritize EGFR and ALK testing over other molecular predictive tests. As scientific discoveries and clinical practice outpace the completion of randomized clinical trials, evidence-based guidelines developed by expert practitioners are vital for communicating emerging clinical standards. Already, new treatments targeting genetic alterations in other, less common driver oncogenes are being evaluated in lung cancer, and testing for these may be addressed in future versions of these guidelines.
Collapse
Affiliation(s)
- Neal I Lindeman
- Department of Pathology, Brigham & Women's Hospital, Boston, Massachusetts 02115-6110, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Idowu MO. Epidermal growth factor receptor in lung cancer: The amazing interplay of molecular testing and cytopathology. Cancer Cytopathol 2013; 121:540-3. [DOI: 10.1002/cncy.21321] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 05/22/2013] [Accepted: 05/22/2013] [Indexed: 11/08/2022]
Affiliation(s)
- Michael O. Idowu
- Department of Pathology; Virginia Commonwealth University Health System; Richmond Virginia
| |
Collapse
|
43
|
Lindeman NI, Cagle PT, Beasley MB, Chitale DA, Dacic S, Giaccone G, Jenkins RB, Kwiatkowski DJ, Saldivar JS, Squire J, Thunnissen E, Ladanyi M. Molecular testing guideline for selection of lung cancer patients for EGFR and ALK tyrosine kinase inhibitors: guideline from the College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Pathology. Arch Pathol Lab Med 2013; 137:828-60. [PMID: 23551194 PMCID: PMC4162344 DOI: 10.5858/arpa.2012-0720-oa] [Citation(s) in RCA: 331] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE To establish evidence-based recommendations for the molecular analysis of lung cancers that are required to guide EGFR- and ALK-directed therapies, addressing which patients and samples should be tested, and when and how testing should be performed. PARTICIPANTS Three cochairs without conflicts of interest were selected, one from each of the 3 sponsoring professional societies: College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Pathology. Writing and advisory panels were constituted from additional experts from these societies. EVIDENCE Three unbiased literature searches of electronic databases were performed to capture articles published from January 2004 through February 2012, yielding 1533 articles whose abstracts were screened to identify 521 pertinent articles that were then reviewed in detail for their relevance to the recommendations. Evidence was formally graded for each recommendation. CONSENSUS PROCESS Initial recommendations were formulated by the cochairs and panel members at a public meeting. Each guideline section was assigned to at least 2 panelists. Drafts were circulated to the writing panel (version 1), advisory panel (version 2), and the public (version 3) before submission (version 4). CONCLUSIONS The 37 guideline items address 14 subjects, including 15 recommendations (evidence grade A/B). The major recommendations are to use testing for EGFR mutations and ALK fusions to guide patient selection for therapy with an epidermal growth factor receptor (EGFR) or anaplastic lymphoma kinase (ALK) inhibitor, respectively, in all patients with advanced-stage adenocarcinoma, regardless of sex, race, smoking history, or other clinical risk factors, and to prioritize EGFR and ALK testing over other molecular predictive tests. As scientific discoveries and clinical practice outpace the completion of randomized clinical trials, evidence-based guidelines developed by expert practitioners are vital for communicating emerging clinical standards. Already, new treatments targeting genetic alterations in other, less common driver oncogenes are being evaluated in lung cancer, and testing for these may be addressed in future versions of these guidelines.
Collapse
Affiliation(s)
- Neal I Lindeman
- Department of Pathology, Brigham & Women's Hospital, Boston, MA 02115-6110, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Righi L, Cuccurullo A, Vatrano S, Cappia S, Giachino D, De Giuli P, Ardine M, Novello S, Volante M, Scagliotti GV, Papotti M. Detection and characterization of classical and "uncommon" exon 19 Epidermal Growth Factor Receptor mutations in lung cancer by pyrosequencing. BMC Cancer 2013; 13:114. [PMID: 23497146 PMCID: PMC3608975 DOI: 10.1186/1471-2407-13-114] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 03/06/2013] [Indexed: 01/15/2023] Open
Abstract
Background The management of advanced stage non-small cell lung cancer is increasingly based on diagnostic and predictive analyses performed mostly on limited amounts of tumor tissue. The evaluation of Epidermal Growth Factor Receptor (EGFR) mutations have emerged as the strongest predictor of response to EGFR-tyrosine kinase inhibitors mainly in patients with adenocarcinoma. Several EGFR mutation detection techniques are available, having both sensitivity and specificity issues, being the Sanger sequencing technique the reference standard, with the limitation of a relatively high amount of mutated cells needed for the analysis. Methods A novel nucleotide dispensation order for pyrosequencing was established allowing the identification and characterization of EGFR mutation not definable with commercially and clinically approved kits, and validated in a consecutive series of 321 lung cancer patients (246 biopsies or cytology samples and 75 surgical specimens). Results 61/321 (19%) mutated cases were detected, 17 (27.9%) in exon 21 and 44 (72.1%) in exon 19, these latter corresponding to 32/44 (72.7%) classical and 12/44 (27.3%) uncommon mutations. Furthermore, a novel, never reported, point mutation, was found, which determined a premature stop codon in the aminoacidic sequence that resulted in a truncated protein in the tyrosine kinase domain, thus impairing the inhibitory effect of specific therapy. Conclusions The novel dispensation order allows to detect and characterize both classical and uncommon EGFR mutations. Although several phase III studies in genotypically defined groups of patients are already available, further prospective studies assessing the role of uncommon EGFR mutations are warranted.
Collapse
Affiliation(s)
- Luisella Righi
- Divisions of Pathology, University of Torino, Regione Gonzole 10, Torino, Orbassano 10043, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
de Biase D, Visani M, Malapelle U, Simonato F, Cesari V, Bellevicine C, Pession A, Troncone G, Fassina A, Tallini G. Next-generation sequencing of lung cancer EGFR exons 18-21 allows effective molecular diagnosis of small routine samples (cytology and biopsy). PLoS One 2013; 8:e83607. [PMID: 24376723 PMCID: PMC3871524 DOI: 10.1371/journal.pone.0083607] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Accepted: 11/05/2013] [Indexed: 02/05/2023] Open
Abstract
Selection of lung cancer patients for therapy with tyrosine kinase inhibitors directed at EGFR requires the identification of specific EGFR mutations. In most patients with advanced, inoperable lung carcinoma limited tumor samples often represent the only material available for both histologic typing and molecular analysis. We defined a next generation sequencing protocol targeted to EGFR exons 18-21 suitable for the routine diagnosis of such clinical samples. The protocol was validated in an unselected series of 80 small biopsies (n=14) and cytology (n=66) specimens representative of the material ordinarily submitted for diagnostic evaluation to three referral medical centers in Italy. Specimens were systematically evaluated for tumor cell number and proportion relative to non-neoplastic cells. They were analyzed in batches of 100-150 amplicons per run, reaching an analytical sensitivity of 1% and obtaining an adequate number of reads, to cover all exons on all samples analyzed. Next generation sequencing was compared with Sanger sequencing. The latter identified 15 EGFR mutations in 14/80 cases (17.5%) but did not detected mutations when the proportion of neoplastic cells was below 40%. Next generation sequencing identified 31 EGFR mutations in 24/80 cases (30.0%). Mutations were detected with a proportion of neoplastic cells as low as 5%. All mutations identified by the Sanger method were confirmed. In 6 cases next generation sequencing identified exon 19 deletions or the L858R mutation not seen after Sanger sequencing, allowing the patient to be treated with tyrosine kinase inhibitors. In one additional case the R831H mutation associated with treatment resistance was identified in an EGFR wild type tumor after Sanger sequencing. Next generation sequencing is robust, cost-effective and greatly improves the detection of EGFR mutations. Its use should be promoted for the clinical diagnosis of mutations in specimens with unfavorable tumor cell content.
Collapse
Affiliation(s)
- Dario de Biase
- Department of Medicine (DIMES) – Anatomic Pathology Unit, Bellaria Hospital, University of Bologna, Bologna, Italy
| | - Michela Visani
- Department of Pharmacology and Biotechnology (FaBiT), University of Bologna, Bologna, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Francesca Simonato
- Department of Medicine (DIMED) – Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy
| | - Valentina Cesari
- Department of Medicine (DIMES) – Anatomic Pathology Unit, Bellaria Hospital, University of Bologna, Bologna, Italy
- Department of Pharmacology and Biotechnology (FaBiT), University of Bologna, Bologna, Italy
| | - Claudio Bellevicine
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Annalisa Pession
- Department of Pharmacology and Biotechnology (FaBiT), University of Bologna, Bologna, Italy
| | - Giancarlo Troncone
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Ambrogio Fassina
- Department of Medicine (DIMED) – Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy
| | - Giovanni Tallini
- Department of Medicine (DIMES) – Anatomic Pathology Unit, Bellaria Hospital, University of Bologna, Bologna, Italy
- * E-mail:
| |
Collapse
|
46
|
Do H, Dobrovic A. Dramatic reduction of sequence artefacts from DNA isolated from formalin-fixed cancer biopsies by treatment with uracil- DNA glycosylase. Oncotarget 2012; 3:546-58. [PMID: 22643842 PMCID: PMC3388184 DOI: 10.18632/oncotarget.503] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Non-reproducible sequence artefacts are frequently detected in DNA from formalin-fixed and paraffin-embedded (FFPE) tissues. However, no rational strategy has been developed for reduction of sequence artefacts from FFPE DNA as the underlying causes of the artefacts are poorly understood. As cytosine deamination to uracil is a common form of DNA damage in ancient DNA, we set out to examine whether treatment of FFPE DNA with uracil-DNA glycosylase (UDG) would lead to the reduction of C>T (and G>A) sequence artefacts. Heteroduplex formation in high resolution melting (HRM)-based assays was used for the detection of sequence variants in FFPE DNA samples. A set of samples that gave false positive HRM results for screening of the E17K mutation in exon 4 of the AKT1 gene were chosen for analysis. Sequencing of these samples showed multiple non-reproducible C:G>T:A artefacts. Treatment of the FFPE DNA with UDG prior to PCR amplification led to a very marked reduction of the sequence artefacts as indicated by both HRM and sequencing analysis. Similar results were shown for the BRAFV600 region in the same sample set and EGFR exon 19 in another sample set. UDG treatment specifically suppressed the formation of artefacts in FFPE DNA as it did not affect the detection of true KRAS codon 12 and true EGFR exon 19 and 20 mutations. We conclude that uracil in FFPE DNA leads to a significant proportion of sequence artefacts. These can be minimised by a simple UDG pre-treatment, which can be readily carried out in the same tube as the PCR, immediately prior to commencing thermal cycling. HRM is a convenient way of monitoring both the degree of damage and the effectiveness of the UDG treatment. These findings have immediate and important implications for cancer diagnostics where FFPE DNA is used as the primary genetic material for mutational studies guiding personalised medicine strategies and where simple effective strategies to detect mutations are required.
Collapse
Affiliation(s)
- Hongdo Do
- Molecular Pathology Research and Development Laboratory, Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
| | | |
Collapse
|
47
|
Angulo B, Conde E, Suárez-Gauthier A, Plaza C, Martínez R, Redondo P, Izquierdo E, Rubio-Viqueira B, Paz-Ares L, Hidalgo M, López-Ríos F. A comparison of EGFR mutation testing methods in lung carcinoma: direct sequencing, real-time PCR and immunohistochemistry. PLoS One 2012; 7:e43842. [PMID: 22952784 PMCID: PMC3428292 DOI: 10.1371/journal.pone.0043842] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 07/30/2012] [Indexed: 01/06/2023] Open
Abstract
The objective of this study is to compare two EGFR testing methodologies (a commercial real-time PCR kit and a specific EGFR mutant immunohistochemistry), with direct sequencing and to investigate the limit of detection (LOD) of both PCR-based methods. We identified EGFR mutations in 21 (16%) of the 136 tumours analyzed by direct sequencing. Interestingly, the Therascreen EGFR Mutation Test kit was able to characterize as wild-type one tumour that could not be analyzed by direct sequencing of the PCR product. We then compared the LOD of the kit and that of direct sequencing using the available mutant tumours. The kit was able to detect the presence of a mutation in a 1% dilution of the total DNA in nine of the 18 tumours (50%), which tested positive with the real-time quantitative PCR method. In all cases, EGFR mutation was identified at a dilution of 5%. Where the mutant DNA represented 30% of the total DNA, sequencing was able to detect mutations in 12 out of 19 cases (63%). Additional experiments with genetically defined standards (EGFR ΔE746-A750/+ and EGFR L858R/+) yielded similar results. Immunohistochemistry (IHC) staining with exon 19-specific antibody was seen in eight out of nine cases with E746-A750del detected by direct sequencing. Neither of the two tumours with complex deletions were positive. Of the five L858R-mutated tumours detected by the PCR methods, only two were positive for the exon 21-specific antibody. The specificity was 100% for both antibodies. The LOD of the real-time PCR method was lower than that of direct sequencing. The mutation specific IHC produced excellent specificity.
Collapse
Affiliation(s)
- Bárbara Angulo
- Laboratorio de Dianas Terapéuticas, Faculty of Medicine, Centro Integral Oncológico “Clara Campal”, Hospital HM Universitario Sanchinarro, Universidad San Pablo-CEU, Madrid, Spain
| | - Esther Conde
- Laboratorio de Dianas Terapéuticas, Faculty of Medicine, Centro Integral Oncológico “Clara Campal”, Hospital HM Universitario Sanchinarro, Universidad San Pablo-CEU, Madrid, Spain
| | - Ana Suárez-Gauthier
- Laboratorio de Dianas Terapéuticas, Faculty of Medicine, Centro Integral Oncológico “Clara Campal”, Hospital HM Universitario Sanchinarro, Universidad San Pablo-CEU, Madrid, Spain
| | - Carlos Plaza
- Laboratorio de Dianas Terapéuticas, Faculty of Medicine, Centro Integral Oncológico “Clara Campal”, Hospital HM Universitario Sanchinarro, Universidad San Pablo-CEU, Madrid, Spain
| | - Rebeca Martínez
- Laboratorio de Dianas Terapéuticas, Faculty of Medicine, Centro Integral Oncológico “Clara Campal”, Hospital HM Universitario Sanchinarro, Universidad San Pablo-CEU, Madrid, Spain
| | - Pilar Redondo
- Laboratorio de Dianas Terapéuticas, Faculty of Medicine, Centro Integral Oncológico “Clara Campal”, Hospital HM Universitario Sanchinarro, Universidad San Pablo-CEU, Madrid, Spain
| | - Elisa Izquierdo
- Laboratorio de Dianas Terapéuticas, Faculty of Medicine, Centro Integral Oncológico “Clara Campal”, Hospital HM Universitario Sanchinarro, Universidad San Pablo-CEU, Madrid, Spain
| | - Belén Rubio-Viqueira
- Laboratorio de Dianas Terapéuticas, Faculty of Medicine, Centro Integral Oncológico “Clara Campal”, Hospital HM Universitario Sanchinarro, Universidad San Pablo-CEU, Madrid, Spain
- Gastrointestinal Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
| | - Luis Paz-Ares
- Department of Oncology, Instituto de Biomedicina de Sevilla (IBIS) & Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Manuel Hidalgo
- Laboratorio de Dianas Terapéuticas, Faculty of Medicine, Centro Integral Oncológico “Clara Campal”, Hospital HM Universitario Sanchinarro, Universidad San Pablo-CEU, Madrid, Spain
- Gastrointestinal Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
| | - Fernando López-Ríos
- Laboratorio de Dianas Terapéuticas, Faculty of Medicine, Centro Integral Oncológico “Clara Campal”, Hospital HM Universitario Sanchinarro, Universidad San Pablo-CEU, Madrid, Spain
- * E-mail:
| |
Collapse
|
48
|
Data-driven unbiased curation of the TP53 tumor suppressor gene mutation database and validation by ultradeep sequencing of human tumors. Proc Natl Acad Sci U S A 2012; 109:9551-6. [PMID: 22628563 DOI: 10.1073/pnas.1200019109] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cancer mutation databases are expected to play central roles in personalized medicine by providing targets for drug development and biomarkers to tailor treatments to each patient. The accuracy of reported mutations is a critical issue that is commonly overlooked, which leads to mutation databases that include a sizable number of spurious mutations, either sequencing errors or passenger mutations. Here we report an analysis of the latest version of the TP53 mutation database, including 34,453 mutations. By using several data-driven methods on multiple independent quality criteria, we obtained a quality score for each report contributing to the database. This score can now be used to filter for high-confidence mutations and reports within the database. Sequencing the entire TP53 gene from various types of cancer using next-generation sequencing with ultradeep coverage validated our approach for curation. In summary, 9.7% of all collected studies, mostly comprising numerous tumors with multiple infrequent TP53 mutations, should be excluded when analyzing TP53 mutations. Thus, by combining statistical and experimental analyses, we provide a curated mutation database for TP53 mutations and a framework for mutation database analysis.
Collapse
|
49
|
Aisner DL, Sams SB. The role of cytology specimens in molecular testing of solid tumors: Techniques, limitations, and opportunities. Diagn Cytopathol 2012; 40:511-24. [DOI: 10.1002/dc.22820] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
50
|
Boissière-Michot F, Lopez-Crapez E, Frugier H, Berthe ML, Ho-Pun-Cheung A, Assenat E, Maudelonde T, Lamy PJ, Bibeau F. KRAS genotyping in rectal adenocarcinoma specimens with low tumor cellularity after neoadjuvant treatment. Mod Pathol 2012; 25:731-9. [PMID: 22282307 DOI: 10.1038/modpathol.2011.210] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
KRAS status assessment is mandatory in patients with metastatic colorectal cancer before therapy with anti-epidermal growth factor receptor monoclonal antibodies, as KRAS mutations are associated with resistance to this treatment. However, KRAS genotyping may be very challenging in case of poor tumor cellularity, particularly when major tumor regression is achieved in locally advanced rectal adenocarcinomas after radiochemotherapy. We aimed at identifying the most reliable strategy to detect KRAS mutations in such samples. DNA was extracted from 31 surgical specimens with major tumor regression, following manual dissection, and from paired pre-treatment biopsies and analyzed by high-resolution melting. DNA samples displaying altered melting curve shapes were then sequenced. Samples with unmodified melting curves or wild-type sequence were further investigated by using an allele-specific PCR assay (TheraScreen) and laser microdissection (followed by high-resolution melting and sequencing analyses). In the 31 post-radiochemotherapy surgical specimens, seven KRAS mutations were identified by high-resolution melting analysis/sequencing. One additional mutation was detected by the TheraScreen assay and two mutations, including the one identified by the TheraScreen assay, were detected following laser microdissection. Altogether, 9/31 surgical specimens (29%) presented KRAS mutations. In the manually dissected pre-treatment biopsies, 12 mutations (39%) were identified by high-resolution melting analysis and sequencing. No additional mutations were found by using the TheraScreen assay or laser microdissection. These results indicate that, in the case of post-radiochemotherapy surgical specimens of colorectal cancer with low tumor cellularity, pre-treatment biopsies might represent the most cost-effective option for reliable KRAS genotyping. The use of more sensitive assays, such as allele-specific PCR or laser microdissection, can be envisaged but with higher costs and longer delays.
Collapse
|