No Evidence for Interference of H&E Staining in DNA Testing Usefulness of DNA Extraction From H&E-Stained Archival Tissue Sections

Similar documents
Clinical and Pathological Significance of Epigenomic Changes in Colorectal Cancer

Department of Pathology, Brigham and Women s Hospital, Harvard Medical School, Boston, MA, USA

Molecular biomarker profile of EGFR copy number, KRAS and BRAF mutations in colorectal carcinoma

Anatomic Molecular Pathology: An Emerging Field

Aspirin Use, Tumor PIK3CA Mutation, and Colorectal-Cancer Survival

T ranscriptional inactivation by cytosine methylation at

BRAF Testing In The Elderly: Same As in Younger Patients?

Supplementary Table 1. PIK3CA mutation in colorectal cancer

Molecular Diagnosis for Colorectal Cancer Patients

The Role of the CpG Island Methylator Phenotype on Survival Outcome in Colon Cancer

Development of Carcinoma Pathways

Colorectal cancer Chapelle, J Clin Oncol, 2010

CpG Island Methylator Phenotype in Primary Gastric Carcinoma

Integration of Cancer Genome into GECCO- Genetics and Epidemiology of Colorectal Cancer Consortium

Epigenetic profiling of synchronous colorectal neoplasias by quantitative DNA methylation analysis

Lynch Syndrome Screening for Endometrial Cancer: Basic Concepts 1/16/2017

CpG Island Methylator Phenotype-Low (CIMP-Low) in Colorectal Cancer: Possible Associations with Male Sex and KRAS Mutations

Evaluation of Markers for CpG Island Methylator Phenotype (CIMP) in Colorectal Cancer by a Large Population-Based Sample

Colorectal Cancer Expression of Peroxisome Proliferator-Activated Receptor (PPARG, PPARgamma) Is Associated With Good Prognosis

Supplementary Online Content

ASSESSMENT OF MLH1 PROMOTER METHYLATION IN ENDOMETRIAL CANCER USING PYROSEQUENCING TECHNOLOGY OBJECTIVES

AmoyDx TM BRAF V600E Mutation Detection Kit

MEDICAL POLICY. SUBJECT: GENOTYPING - RAS MUTATION ANALYSIS IN METASTATIC COLORECTAL CANCER (KRAS/NRAS) POLICY NUMBER: CATEGORY: Laboratory

Colon cancer: practical molecular diagnostics. Wade S. Samowitz, M.D. University of Utah and ARUP

A Meta-Regression Method for Studying Etiologic Heterogeneity across. Disease Subtypes Classified by Multiple Biomarkers

Prospective Study of Family History and Colorectal Cancer

Evolution of Pathology

Utilization of Cell-Transfer Technique for Molecular Testing on Hematoxylin-Eosin Stained Sections

Personalized Medicine: Lung Biopsy and Tumor

High risk stage II colon cancer

The estimation of tumor cell percentage for molecular testing by pathologists is not accurate

The molecular genetics of colorectal cancer

ORIGINAL ARTICLE. and treatment of patients

NGS in tissue and liquid biopsy

RNA preparation from extracted paraffin cores:

Colorectal Cancer - Working in Partnership. David Baty Genetics, Ninewells Hospital

Published OnlineFirst October 14, 2009; DOI: / CCR

Identification of Potential Therapeutic Targets by Molecular and Genomic Profiling of 628 Cases of Uterine Serous Carcinoma

Molecular Pathological Epidemiology (MPE): Novel Integrative Pathology Science (9/24/ min)

The diagnostic and prognostic value of genetic aberrations in resectable distal bile duct cancer Rijken, A.M.

METASTATIC COLORECTAL CANCER: TUMOR MUTATIONAL ANALYSIS AND ITS IMPACT ON CHEMOTHERAPY SUMA SATTI, MD

Gastric Carcinoma with Lymphoid Stroma: Association with Epstein Virus Genome demonstrated by PCR

Serrated Polyps and a Classification of Colorectal Cancer

Colorectal Cancer in 2017: From Biology to the Clinics. Rodrigo Dienstmann

Supplementary Appendix

CAP Laboratory Improvement Programs. Summary of Microsatellite Instability Test Results From Laboratories Participating in Proficiency Surveys

The College of American Pathologists (CAP) offers these

CpG islands are DNA segments of at least 0.5 kb in size,

Template for Reporting Results of Biomarker Testing of Specimens From Patients With Carcinoma of the Colon and Rectum

Accepted Manuscript. Pancreatic Cancer Subtypes: Beyond Lumping and Splitting. Andrew J. Aguirre

Prognostic significance of K-Ras mutation rate in metastatic colorectal cancer patients. Bruno Vincenzi Università Campus Bio-Medico di Roma

WHAT SHOULD WE DO WITH TUMOUR BUDDING IN EARLY COLORECTAL CANCER?

Microsatellite instability and other molecular markers: how useful are they?

BRAF mutation in colorectal carcinomas with signet ring cell component

Current Status of Biomarkers (including DNA Tumor Markers and Immunohistochemistry in the Laboratory Diagnosis of Tumors)

Research Article Analysis of KRAS Mutations of Exon 2 Codons 12 and 13 by SNaPshot Analysis in Comparison to Common DNA Sequencing

Molecular markers in colorectal cancer. Wolfram Jochum

Supplementary Information

Updated Molecular Testing Guideline for the Selection of Lung Cancer Patients for Treatment with Targeted Tyrosine Kinase Inhibitors

Collaboration between CEQAS and UK NEQAS for Molecular Genetics Solid Tumour EQAs and variant interpretation

Transform genomic data into real-life results

SSM signature genes are highly expressed in residual scar tissues after preoperative radiotherapy of rectal cancer.

TITLE: CYP1B1 Polymorphism as a Risk Factor for Race-Related Prostate Cancer

The Cancer Research UK Stratified Medicine Programme: Phases One and Two Dr Emily Shaw

Chapter 8. Summary. Chapter 8. Summary

Preanalytic Variables in Cytology: Lessons Learned from Next Generation Sequencing

QIAGEN's Growing Immuno-Oncology Testing Portfolio

Introduction. Why Do MSI/MMR Analysis?

Molecular Testing Updates. Karen Rasmussen, PhD, FACMG Clinical Molecular Genetics Spectrum Medical Group, Pathology Division Portland, Maine

Liquid biopsy: the experience of real life case studies

SALSA MLPA probemix P315-B1 EGFR

Fluxion Biosciences and Swift Biosciences Somatic variant detection from liquid biopsy samples using targeted NGS

EGFR methylation and outcome of patients with advanced colorectal cancer treated with cetuximab

IntelliGENSM. Integrated Oncology is making next generation sequencing faster and more accessible to the oncology community.

Supplementary Information Titles Journal: Nature Medicine

Liquid biopsy in lung cancer: The EGFR paradigm

Detection of Anaplastic Lymphoma Kinase (ALK) gene in Non-Small Cell lung Cancer (NSCLC) By CISH Technique

Biomarker development in the era of precision medicine. Bei Li, Interdisciplinary Technical Journal Club

DNA methylation in the ARG-NOS pathway is associated with exhaled nitric oxide in asthmatic children

Downregulation of serum mir-17 and mir-106b levels in gastric cancer and benign gastric diseases

Molecular Testing in Lung Cancer

M. Azzam Kayasseh,Dubai,UAE

FONS Nové sekvenační technologie vklinickédiagnostice?

Prognostic significance of nemo like kinase in nasopharyngeal carcinoma

The Incidence and Significance of Villous Change in Adenomatous Polyps

Contributions to Anatomic Pathology, over the years

Rapid BRAF mutation detection in melanoma patients by immunohistochemistry

EXAMPLE. ratio (%) Contraindication for treatment with panitumumab or cetuximab

Mutations in Both KRAS and BRAF May Contribute to the Methylator Phenotype in Colon Cancer

Caring for a Patient with Colorectal Cancer. Objectives. Poll question. UNC Cancer Network Presented on 10/15/18. For Educational Use Only 1

NGS ONCOPANELS: FDA S PERSPECTIVE

Frequency(%) KRAS G12 KRAS G13 KRAS A146 KRAS Q61 KRAS K117N PIK3CA H1047 PIK3CA E545 PIK3CA E542K PIK3CA Q546. EGFR exon19 NFS-indel EGFR L858R

Current and future applications of Molecular Pathology. Kathy Walsh Clinical Scientist NHS Lothian

Colon Cancer and Hereditary Cancer Syndromes

Original Article P53 mutations occur more commonly than KRAS mutations in colorectal adenoma

Applying Genomics to Cancer 21 st September The Frequency of EGFR mutations in Lung Adenocarcinoma: The Cardiff Experience

Precision Genetic Testing in Cancer Treatment and Prognosis

Supplementary Tables. Supplementary Figures

Related Policies None

Transcription:

Anatomic Pathology / H& Staining in DNA Testing No vidence for Interference of H& Staining in DNA Testing Usefulness of DNA xtraction From H&-Stained Archival Tissue Sections Teppei Morikawa, MD, PhD,,2* Kaori Shima, DDS, PhD,,2* Aya Kuchiba, PhD,,2* Mai Yamauchi, PhD,,2 Noriko Tanaka, PhD,,2 Yu Imamura, MD, PhD,,2 Xiaoyun Liao, MD, PhD,,2 Zhi Rong Qian, MD, PhD,,2 Mohan Brahmandam, MS, 2 Janina A. Longtine, MD, 3 Neal I. Lindeman, MD, 3 Charles S. Fuchs, MD, MPH, 2,4 and Shuji Ogino, MD, PhD, MS -3 Key Words: Histochemical staining; Polymerase chain reaction; Molecular diagnostics; Mutation detection; Microdissection; Solid tumor DOI: 0.309/AJCP28LAOOKSZSVW Abstract Although histochemical staining has been believed to inhibit the DNA amplification reaction, no previous study has systematically evaluated the influence of histochemical staining on downstream molecular assays. To evaluate an influence of H& staining on DNA testing, we isolated DNA from 0 unstained, 0 hematoxylin-stained, 0 eosin-stained, and 0 H&-stained tissue sections (ie, 4 groups), from each of 5 colon cancers. Among the 4 groups, we did not observe any significant or appreciable difference in DNA fragmentation by agarose gel electrophoresis, in DNA amplification by real-time polymerase chain reaction (PCR), in microsatellite PCR fragment analyses, or in a PCR-pyrosequencing assay. As a proof-of-principle study, we successfully performed microsatellite instability analysis and sequencing of KRAS and BRAF on more than,300 colorectal cancers using DNA extracted from H&-stained tissue sections. Our data provide no evidence for an interfering effect of H& staining on DNA testing, suggesting that DNA from H&-stained sections can be effectively used for routine DNA testing. Screening and identification of genetic alterations in formalin-fixed, paraffin-embedded (FFP) tumor materials have been important in molecular diagnosis and clinical medicine. -6 Solid tumors are inherently heterogeneous tissues that contain neoplastic cells admixed with nonneoplastic cells, including inflammatory cells, vascular and lymphatic endothelial cells, smooth muscle cells, and stromal fibroblasts. Therefore, an enrichment of neoplastic cells is commonly performed before DNA extraction in molecular testing to avoid false-negative results due to low neoplastic cellularity. 7 Macrodissection from tissue sections on glass slides has been a widely used method to exclude pure nonneoplastic areas and enrich neoplastic cellularity for molecular testing. For macrodissection of tumor areas, an accurate morphologic identification of tumor areas is essential. 8,9 Thus, macrodissection should be guided by an H&-stained tissue section with tumor areas marked by a pathologist under a microscope Image A. It would be ideal to dissect tumor tissue from an H&-stained section (without coverslip) because the H& stain enables us to easily identify tumor areas Image B, particularly next to the H& slide with the marked tumor areas (Image A). In contrast, it is difficult to accurately identify tumor areas of an unstained tissue section Image C, and tumor areas can be missed. With regard to the influence of histochemical staining on downstream molecular assays, some previous studies have shown interfering effects of histochemical staining, especially hematoxylin stain, 0-2 while another study failed to show a significant difference in polymerase chain reaction (PCR) amplification between DNA specimens from H&-stained and unstained tissue sections. 3 To avoid possible interfering 22 Am J Clin Pathol 202;38:22-29 22 DOI: 0.309/AJCP28LAOOKSZSVW

Anatomic Pathology / Original Article effects of histochemical staining on molecular assays, macrodissection of tumor from unstained tissue sections is commonly performed, despite the difficulty in identifying tumor areas (Image C) and the risk of missing tumor areas.,4-7 To our knowledge, no previous study has systematically evaluated the influence of histochemical staining on downstream molecular assays. We therefore conducted this study to evaluate various molecular assay results on DNA extracted from H&stained, hematoxylin-stained, and eosin-stained FFP tissue sections, in comparison with DNA extracted from unstained tissue sections. We compared results of agarose gel genomic DNA fragment analysis (without PCR), quantitative real-time PCR assay, PCR-pyrosequencing assay, and PCR capillary electrophoresis fragment analyses. We did not observe an appreciable difference in any of the results on DNA specimens from stained tissue. Furthermore, as a proof-of-principle study, we successfully used DNA from H&-stained tissue sections and obtained DNA analysis data from more than,300 colorectal cancers. Our data support the feasibility of DNA extraction from H&-stained tissue sections for routine clinical laboratory workflow. Materials and Methods FFP Tissue Specimens Specimen collection and analysis in this study were approved by the Harvard School of Public Health, Brigham and Women s Hospital, and Dana-Farber Cancer Institute institutional review boards (Boston, MA). FFP colorectal cancer tissue specimens (5 cases) were anonymized after collection from the archival file of the Department of Pathology, Brigham and Women s Hospital, to assess the effects of histochemical staining on downstream molecular testing Figure. As a proof-of-principle feasibility study, we tested DNA extraction from H&-stained tissue sections in a large cohort of colorectal cancer cases. We used 2 US nationwide prospective cohort studies, the Nurses Health Study (N = 2,70 women followed up since 976) and the Health Professionals Follow-up Study (N = 5,529 men followed up since 986). 8,9 We collected FFP tissue blocks from hospitals and pathology laboratories throughout the United States where cohort participants with colorectal cancer underwent cancer resections. 8,9 H&-stained tissue sections were reviewed by a pathologist (S.O.). Genomic DNA was extracted from H&-stained tissue sections, using another H&-stained section with marked tumor areas as a guide (as in Images A and B), for downstream KRAS, BRAF, and MSI testing, and results were available for,34,,33, and,293 cases, respectively. A B C Image xamples of H&-stained and unstained tissue sections. A, An H&-stained tissue section slide to guide tumor tissue dissection. A pathologist marked a tumor area under a microscope. B, An H&-stained tissue section without coverslip for subsequent DNA extraction. It is easy to identify the tumor area. C, An unstained tissue section for DNA extraction. It is not as easy to identify the tumor area as in the H&-stained tissue section (B). Overall Study Strategy, DNA xtraction, and Agarose Gel lectrophoresis Deparaffinized and hydrated 0-μm sections from the same tissue block were stained by hematoxylin (Harris hematoxylin, Surgipath, Richmond, IL) alone for 3 minutes (group 2), by eosin (Surgipath) alone for 5 seconds (group 3), or by H& (group 4; Figure ). Unstained slides were prepared as control slides (group ). Figure illustrates our overall strategy of experiments to assess the effects of hematoxylin and/or eosin staining on DNA integrity and downstream molecular assays. To avoid bias that could be introduced by tumor-directed macrodissection, a whole tissue section (including tumor and adjacent normal tissue) was entirely scraped off from a glass slide by using a sterile needle. Thus, DNA yields were very similar between the groups. The scraped tissue was collected into a microtube, and genomic DNA was extracted using QIAamp DNA Mini Kit (Qiagen, Valencia, CA). As a result, each of 0 tissue sections was aliquoted to tube, to yield 0 aliquoted DNA specimens for each group (Figure ). The 260/280 nm absorbance ratio of extracted DNA was approximately.8 (NanoDrop, Thermo Scientific, Waltham, MA). xtracted DNA from unstained tissue and H&-stained tissue from all 5 cases was analyzed by electrophoresis in 0.8% Am J Clin Pathol 202;38:22-29 23 23 DOI: 0.309/AJCP28LAOOKSZSVW 23

Morikawa et al / H& Staining in DNA Testing Unstained 0 sections 2 Hematoxylin-stained 0 sections 3 osin-stained 4 H&-stained 0 sections 0 sections agarose gel Image 2. Three cases were used for real-time PCR and PCR-pyrosequencing assay, and the other 2 cases were used for microsatellite PCR fragment analysis. Real-Time PCR Assay for GAPDH To assess PCR amplification efficiency, we performed real-time PCR using the primers for GAPDH and SYBR Green PCR Master Mix (Applied Biosystems, Foster City, CA) by the ABI 7300 Real-Time PCR system (Applied Biosystems). Primer sequences were DNA aliquots 0 0 0 0 Agarose gel (without PCR) Real-time PCR WGA PCR capillary electrophoresis Pyrosequencing Agarose gel (without PCR) Real-time PCR WGA PCR capillary electrophoresis Pyrosequencing Agarose gel (without PCR) Real-time PCR WGA PCR capillary electrophoresis Pyrosequencing Agarose gel (without PCR) Real-time PCR WGA PCR capillary electrophoresis Pyrosequencing Figure Our overall strategy to assess the influence of tissue staining on downstream DNA testing. DNA was extracted from unstained tissue (group ), hematoxylin-stained tissue (group 2), eosin-stained tissue (group 3), and H&-stained tissue (group 4). PCR, polymerase chain reaction; WGA, whole genome amplification. 5'-GTCATGGGTGTGAACCATGAGAA-3'and 5'-TGGT- CATGAGTCCTTCCACGAT-3'. The PCR reaction was repeated 4 times on each of the 0 DNA aliquots for groups through 4, and cycle threshold (Ct) values were compared. Microsatellite PCR Fragment Analysis To assess the effects of hematoxylin and/or eosin staining on PCR fragment analysis, we performed PCR for dinucleotide markers, D2S23 and D5S346, 20 after PCR-based whole genome amplification on genomic DNA as previously Image 2 Agarose gel (0.8%) electrophoresis to assess integrity of DNA from unstained and H&-stained tissue sections. There was no appreciable difference in DNA integrity between DNA specimens from unstained tissue and H&-stained tissue sections. The representative case (0 sections) was shown. Lane, size marker; lanes 2-, group (unstained tissue); lanes 2-2, group 4 (H&-stained tissue). bp, base pairs. 24 Am J Clin Pathol 202;38:22-29 24 DOI: 0.309/AJCP28LAOOKSZSVW

Anatomic Pathology / Original Article described. 2 Forward primers were labeled with fluorescence, and PCR product sizes were 80 base pairs (bp; D2S23) and 29 bp (D5S346). PCR products were electrophoresed and analyzed by the ABI 3730 DNA Analyzer (Applied Biosystems). PCR fragment analysis was repeated twice on each of the 0 DNA aliquots. For the large feasibility cohort, we performed microsatellite instability (MSI) analysis using a 0-marker panel (D2S23, D5S346, D7S250, BAT25, BAT26, BAT40, D8S55, D8S56, D8S67, and D8S487). 20 MSI-high was defined as the presence of instability in 30% or more of the markers and MSI-low/microsatellite stability as instability in fewer than 30% of the markers. 20 PCR-Pyrosequencing Assay for KRAS and BRAF To assess the effect of hematoxylin and/or eosin staining on pyrosequencing, we performed a PCR-pyrosequencing assay for KRAS (codons 2 and 3), which was previously developed and validated. 2 The size of PCR product was 82 bp, and 0 μl of each was sequenced by using the Pyrosequencing PSQ96 HS System (Qiagen). The PCRpyrosequencing reaction was repeated 3 times on each of the 0 aliquots. In addition, in the feasibility cohort, the PCRpyrosequencing assay for BRAF (codon 600) was performed as previously described. 22 Statistical Analysis For all statistical analyses, we used the SAS program (version 9., SAS Institute, Cary, NC). All P values were 2-sided. The Ct in real-time PCR, which reflected amplification efficiency of each PCR reaction given similar amounts A Mean Ct 40 35 30 25 2 3 4 of scraped tissue and extracted DNA, was compared by using the analysis of variance (ANOVA) test, adjusting for case, spot of each reaction, and plate. We performed the ANOVA test for comparing peak heights of the fluorescence signal on capillary electrophoretograms in microsatellite analysis. The distribution of the peak height values (median, 3,028.5; range, 0-0,68) was normalized by logarithmic transformation after adding 0.5 to each value (to log-transform the value of 0 ). A deviation from the null hypothesis in any of the comparisons might imply an interfering effect of histochemical staining on the DNA testing process. Results Integrity of DNA From Unstained and Stained Tissue Our overall strategy to assess the influence of H& staining on subsequent DNA testing for FFP tissue is shown in Figure. First, we assessed the influence of H& staining on DNA integrity. xtracted DNA specimens of group (unstained) and group 4 (H&-stained) were loaded onto 0.8% agarose gel (Image 2). There was no appreciable difference in DNA integrity between the DNA specimens from H&-stained and unstained tissue sections from all 5 cases. Comparison of Ct Values in Real-Time PCR We compared Ct values in real-time PCR for GAPDH in the 4 groups using 3 cases ( 0 sections 4 repeated runs) by the ANOVA test, which adjusted for case, spot of each reaction, and plate Figure 2A. There was no significant difference in Ct values between groups (P =.9). D2S23 ( 0 3 peak height) D5S346 ( 0 3 peak height) 5 4 3 2 0 0 8 6 4 2 0 Case 2 3 4 Case 2 3 4 5 4 3 2 0 0 8 6 4 2 0 Case 2 2 3 4 Case 2 2 3 4 Figure 2 Tissue staining and subsequent polymerase chain reaction (PCR) analysis. A, Mean cycle threshold (Ct) values in quantitative real-time PCR for GAPDH on DNA specimens (3 cases 0 sections 4 repeated runs) in each group. The error bars indicate the SD. There was no significant difference in the mean Ct values between groups (P =.9). B, Mean peak heights in electrophoretograms in PCR-fragment analysis of the microsatellite markers (D2S23 and D5S346) on DNA specimens (2 cases 0 sections 2 repeats) from each group. The error bars indicate the SD. There was no significant difference in the mean peak heights in the 4 groups (overall P =.35). B Am J Clin Pathol 202;38:22-29 25 25 DOI: 0.309/AJCP28LAOOKSZSVW 25

Morikawa et al / H& Staining in DNA Testing Microsatellite PCR Fragment Analysis We performed PCR fragment analysis for the 2 microsatellite markers, D2S23 and D5S346, and assessed the potential influence of tissue staining by using 2 cases ( 0 sections 2 repeats). We measured the peak height in electrophoretograms, and compared the mean peak height value for the 4 groups. There was no significant difference in the mean peak height in the 4 groups (overall P =.35) Figure 2B. PCR-Pyrosequencing Assay for KRAS We performed the PCR-pyrosequencing assay for KRAS codons 2 and 3 on 3 cases ( 0 sections 3 repeats) and compared results for the 4 groups. In 2 cases, there was a c.35g>a (p.g2d) mutation, and case showed only a wildtype sequence Figure 3. All pyrograms for KRAS codons 2 and 3 showed clear peaks and little background, and there was no appreciable difference in the 4 groups. MSI Testing and Pyrosequencing for KRAS and BRAF in the Large Feasibility Cohort Finally, we performed a proof-of-principle feasibility study on FFP tissue blocks that were fixed and processed in numerous pathology laboratories throughout the United States, using the 2 US nationwide prospective cohort studies. 8 We dissected tumor tissue from H&-stained sections (without coverslip) using a guide H& slide with marked tumor areas (as in Images A and B), and extracted DNA for subsequent MSI analysis and the PCR-pyrosequencing assay for KRAS and BRAF. KRAS and BRAF mutations were detected in 470 (35.8%) of,34 cases and 86 (4.2%) of,33 cases, respectively Table. MSI-high was present in A Wild-type G T GG C G T A GG C AA G Mutant A T GG C G T A GG C AA G A G 2 3 4 S A T C G A T C G A T C G A T C G A T C G A T C G A T C G S A T C G A T C G A T C G A T C G A T C G A T C G A T C G S A T C G A T C G A T C G A T C G A T C G A T C G A T C G S A T C G A T C G A T C G A T C G A T C G A T C G A T C G 97 (5.2%) of,293 cases. These frequencies are compatible with the published data on these molecular features in colorectal cancers, 23-27 particularly in population-based studies. 28-33 There was no evidence for an interfering effect of H& staining on these molecular assays. These data suggest that DNA from H&-stained sections can be used for routine tumor DNA testing on FFP tissue. Discussion We conducted this study to evaluate the potential influence of H& staining on subsequent DNA testing. Solid tumor tissue is inherently heterogeneous tissue with an admixture of neoplastic cells and nonneoplastic cells, Table Molecular Features of Colorectal Cancer by Analysis of DNA From H&-Stained Tissue in Two US Nationwide Prospective Cohort Studies, the Nurses Health Study and the Health Professionals Follow-up Study Molecular Feature No. (%) KRAS (n =,34) Codon 2 or 3 mutation 470 (35.8) Wild-type 844 (64.2) BRAF (n =,33) Codon 600 mutation 86 (4.2) Wild-type,27 (85.8) MSI (n =,293) MSI-high 97 (5.2) MSS/MSI-low,096 (84.8) MSI, microsatellite instability; MSS, microsatellite stable. B 2 3 4 G T GG C G T A GG C AA S A C T G A T C G A T C G A T C G A T C G A T C G A T C G S A C T G A T C G A T C G A T C G A T C G A T C G A T C G S A C T G A T C G A T C G A T C G A T C G A T C G A T C G S A C T G A T C G A T C G A T C G A T C G A T C G A T C G G Figure 3 Tissue staining and subsequent polymerase chain reaction (PCR)-pyrosequencing assay for KRAS. A, Case with mutant KRAS codon 2 (c.35g>a) admixed with wild-type sequence. The assay was repeated 3 times on 20 DNA aliquot specimens (3 cases 4 groups 0 sections), and representative results are shown. There was no appreciable difference between DNA specimens from the 4 groups. B, Case 3 with wild-type KRAS. The assay was repeated 3 times on 20 DNA aliquot specimens (3 cases 4 groups 0 sections), and representative results are shown. There was no appreciable difference between DNA specimens from the 4 groups. 26 Am J Clin Pathol 202;38:22-29 26 DOI: 0.309/AJCP28LAOOKSZSVW

Anatomic Pathology / Original Article including inflammatory, stromal, endothelial, and smooth muscle cells, even within tumor tissue areas. Macrodissection from H&-stained tissue sections enables accurate identification and dissection of tumor areas to obtain DNA specimens with a high content of neoplastic cell DNA. specially for small tumor nests or metastatic tumor foci that must be carefully and precisely dissected, it is very useful to stain and clearly visualize the tissue section. H& staining is a common and simple method to visualize the tumor area of interest. Although hematoxylin staining has been believed to inhibit DNA amplification reaction, only a small number of studies have been performed to assess the potential influence of tissue staining on amplification of DNA by PCR. 0,,3 Notably, no previous study has systematically evaluated the influence of H& staining on downstream molecular assays. Our current study provided no evidence for interfering effects of H& staining on subsequent DNA assays. Our data support routine clinical use of H& staining of tissue sections for subsequent tumor tissue dissection, DNA extraction, and molecular analyses. Previous studies evaluated potential influence of tissue staining on DNA assays. 0,,3 Burton et al 0 and Chen et al 34 showed that hematoxylin staining inhibited DNA amplification by PCR, and Diss et al reported that DNA from H&stained tissue was amplified less efficiently than DNA from unstained tissue. On the other hand, Murase et al 3 reported no significant difference in PCR amplification between DNA specimens from unstained and H&-stained tissue. However, none of the previous studies 0,,3 quantitatively evaluated the efficiencies of PCR amplification on DNA specimens from unstained and stained tissues. In the present study, there was no statistically significant difference in GAPDH amplification by quantitative real-time PCR between DNA specimens from unstained tissue, hematoxylin-stained tissue, eosin-stained tissue, and H&-stained tissue. Pyrosequencing is a nonelectrophoretic sequencing by nucleotide extension. Pyrosequencing is useful for sequencing of relatively short nucleotides (up to 40 to 50 bp). Therefore, pyrosequencing has been applied for single nucleotide polymorphism genotyping, 35 bacterial strain typing, 36 mutation detection in tumors, 2,37,38 quantitative promoter CpG island methylation analysis, 39,40 and measurement of LIN- methylation. 4 We evaluated the influence of H& staining for the PCR-pyrosequencing assay for KRAS and showed no appreciable difference in results between DNA specimens from unstained and H&-stained tissues. Capillary electrophoresis is commonly used for microsatellite analysis in FFP clinical tissue specimens (eg, MSI and loss of heterozygosity analyses). 42-45 We showed no significant difference in peak heights of the electrophoretograms of 2 microsatellite markers (D2S23 and D5S346) between DNA specimens from unstained and stained tissues. Murphy et al 46 reported an artifact (a peak at 7 bp) in capillary electrophoresis due to autofluorescence from eosin, which was probably contaminated by nonstringent DNA purification. A notable difference from our study was that Murphy et al 46 experienced the artifact after biopsy tissue (not a tissue section on a slide) was stained with eosin to enable identification of small tissue fragments during sectioning. We have not experienced this problem, despite the fact that we have performed microsatellite PCR fragment analysis on more than,300 cases using DNA from H&-stained tissue. Nevertheless, we agree with Murphy et al 46 that artifacts caused by eosin should be avoided by using stringent DNA purification steps, and that artifacts from histochemical staining should be considered when peaks of the same product size (eg, 7 bp) are present in multiple specimens or primary peaks contain additional underlying peaks of other colors. As a potential limitation of our study, we used a relatively small number of tissue blocks (N = 5) to systematically compare the effects of histochemical staining in downstream molecular assays. Nevertheless, we used as many as 0 sections and 0 DNA aliquots for each condition to achieve a precise measurement estimate. Moreover, as the proof-ofprinciple feasibility study, we used DNA extracted from H&-stained tissue of more than,300 colorectal cancers in 2 US nationwide prospective cohort studies (the Nurses Health Study and the Health Professionals Follow-up Study) for MSI analysis and pyrosequencing of KRAS and BRAF (Table ). The FFP tissue specimens were retrieved from numerous pathology laboratories throughout the United States. Despite the diversity of laboratories that processed and stored tissues, we were successful in performing those molecular assays in more than 95% of cases tested, and the frequencies of MSIhigh and mutations of KRAS and BRAF were compatible with published data on colorectal cancer, 23-27 particularly in population-based studies. 28-33 In addition, using DNA from H&-stained tissue sections, it has been shown that BRAF mutation and MSI-high in colon cancer are associated with the CpG island methylator phenotype (CIMP 20,28,47-52 ) and patient outcome, 20,53 and KRAS mutation is associated with CIMP-low in our cohort studies. 22 Our data indicate that H&-staining is useful for DNA analysis on FFP tissues from numerous pathology laboratories in multicenter clinical trials or population-based cohort studies, 42,54 which can contribute to improvement of patient care and public heath. Our present study represents the first systematic investigation on the effects of H& staining in downstream molecular assays. Our results provide no evidence to support the common conception that H& staining interferes with molecular assays on DNA from FFP tissue. Our data support routine clinical use of H& staining on tumor sections for subsequent tissue dissection and DNA extraction. H& staining enables easy and accurate identification of tumor areas to Am J Clin Pathol 202;38:22-29 27 27 DOI: 0.309/AJCP28LAOOKSZSVW 27

Morikawa et al / H& Staining in DNA Testing obtain DNA with a high content of neoplastic cellular DNA. H& staining can be easily incorporated into routine pathology workflow, so that laboratory personnel can easily identify and dissect tumor areas from H&-stained tissue sections, guided by an H&-stained slide with marked tumor areas. Thus, our data may have considerable influence on clinical molecular diagnostics and routine tumor molecular testing toward personalized medicine. From the Center for Molecular Oncologic Pathology, Dana- Farber Cancer Institute, Boston, MA; 2 Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School; 3 Department of Pathology, Brigham and Women s Hospital, and Harvard Medical School; and 4 Department of Medicine, Brigham and Women s Hospital, and Harvard Medical School. Supported by US National Institute of Health (NIH) grants P0 CA87969 (S.. Hankinson), P0 CA55075 (W.C. Willett), P50 CA27003 (Dr Fuchs), R0 CA8553 (Dr Fuchs), R0 CA24908 (Dr Fuchs), and R0 CA5993 (Dr Ogino). Address reprint requests to Dr Morikawa: Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Room JF-208, Boston, MA 0225; or Dr Ogino: Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Brigham and Women s Hospital, 450 Brookline Ave, Room JF-25C, Boston, MA 0225. * Drs Morikawa, Shima, and Kuchiba contributed equally to this work. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Cancer Institute or NIH. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. Acknowledgments: We deeply thank the Nurses Health Study (NHS) and Health Professionals Follow-up Study (HPFS) cohort participants who have agreed to provide us with information through questionnaires and biological specimens; hospitals and pathology departments throughout the United States for generously providing us with tissue specimens; the staff of the NHS and the HPFS for their valuable contributions; and all of the US state cancer registries for their help. References. Ausch C, Buxhofer-Ausch V, Oberkanins C, et al. Sensitive detection of KRAS mutations in archived formalin-fixed paraffin-embedded tissue using mutant-enriched PCR and reverse-hybridization. J Mol Diagn. 2009;:508-53. 2. Zheng S, Cummings OW, Saxena R, et al. Clonality and TP53 mutation analysis of focal nodular hyperplasia of the liver. Am J Clin Pathol. 200;34:65-70. 3. Battochio A, Mohammed S, Winthrop D, et al. Detection of c-kit and PDGFRA gene mutations in gastrointestinal stromal tumors: comparison of DHPLC and DNA sequencing methods using a single population-based cohort. Am J Clin Pathol. 200;33:49-55. 4. Hostein I, Faur N, Primois C, et al. BRAF mutation status in gastrointestinal stromal tumors. Am J Clin Pathol. 200;33:4-48. 5. Ross JS, Torres-Mora J, Wagle N, et al. Biomarker-based prediction of response to therapy for colorectal cancer: current perspective. Am J Clin Pathol. 200;34:478-490. 6. Tsiatis AC, Norris-Kirby A, Rich RG, et al. Comparison of Sanger sequencing, pyrosequencing, and melting curve analysis for the detection of KRAS mutations: diagnostic and clinical implications. J Mol Diagn. 200;2:425-432. 7. Monzon FA, Ogino S, Hammond M, et al. The role of KRAS mutation testing in the management of patients with metastatic colorectal cancer. Arch Pathol Lab Med. 2009;33:600-606. 8. Shibata D, Hawes D, Li ZH, et al. Specific genetic analysis of microscopic tissue after selective ultraviolet radiation fractionation and the polymerase chain reaction. Am J Pathol. 992;4:539-543. 9. Lukish JR, Muro K, DeNobile J, et al. Prognostic significance of DNA replication errors in young patients with colorectal cancer. Ann Surg. 998;227:5-56. 0. Burton MP, Schneider BG, Brown R, et al. Comparison of histologic stains for use in PCR analysis of microdissected, paraffin-embedded tissues. Biotechniques. 998;24:86-92.. Diss TC, Pan L, Peng H, et al. Sources of DNA for detecting B cell monoclonality using PCR. J Clin Pathol. 994;47:493-496. 2. de Lang A, Wilander. Sensitivity of HPV tests on stained vs unstained cervical smears. Acta Cytol. 2005;49:595-599. 3. Murase T, Inagaki H, imoto T. Influence of histochemical and immunohistochemical stains on polymerase chain reaction. Mod Pathol. 2000;3:47-5. 4. Rowe LR, Bentz BG, Bentz JS. Detection of BRAF V600 activating mutation in papillary thyroid carcinoma using PCR with allele-specific fluorescent probe melting curve analysis. J Clin Pathol. 2007;60:2-25. 5. van Puijenbroek M, Nielsen M, Tops CM, et al. Identification of patients with (atypical) MUTYH-associated polyposis by KRAS2 c.34g > T prescreening followed by MUTYH hotspot analysis in formalin-fixed paraffinembedded tissue. Clin Cancer Res. 2008;4:39-42. 6. Kristensen LS, Wojdacz TK, Thestrup BB, et al. Quality assessment of DNA derived from up to 30 years old formalin fixed paraffin embedded (FFP) tissue for PCR-based methylation analysis using SMART-MSP and MS-HRM. BMC Cancer. 2009;9:453. doi:0.86/47-2407-9-453. 7. Messick CA, Church JM, Liu X, et al. Stage III colorectal cancer: molecular disparity between primary cancers and lymph node metastases. Ann Surg Oncol. 200;7:425-43. 8. Morikawa T, Kuchiba A, Yamauchi M, et al. Association of CTNNB (β-catenin) alterations, body mass index, and physical activity with survival in patients with colorectal cancer. JAMA. 20;305:685-694. 9. Chan AT, Ogino S, Fuchs CS. Aspirin and the risk of colorectal cancer in relation to the expression of COX-2. N ngl J Med. 2007;356:23-242. 20. Nosho K, Irahara N, Shima K, et al. Comprehensive biostatistical analysis of CpG island methylator phenotype in colorectal cancer using a large population-based sample. PLoS One. 2008;3:e3698. doi:0.37/journal.pone.0003698. 2. Ogino S, Kawasaki T, Brahmandam M, et al. Sensitive sequencing method for KRAS mutation detection by pyrosequencing. J Mol Diagn. 2005;7:43-42. 22. Ogino S, Kawasaki T, Kirkner GJ, et al. CpG island methylator phenotype-low (CIMP-low) in colorectal cancer: possible associations with male sex and KRAS mutations. J Mol Diagn. 2006;8:582-588. 28 Am J Clin Pathol 202;38:22-29 28 DOI: 0.309/AJCP28LAOOKSZSVW

Anatomic Pathology / Original Article 23. De Roock W, Claes B, Bernasconi D, et al. ffects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis. Lancet Oncol. 200;:753-762. 24. Figueiredo JC, Grau MV, Wallace K, et al. Global DNA hypomethylation (LIN-) in the normal colon and lifestyle characteristics and dietary and genetic factors. Cancer pidemiol Biomarkers Prev. 2009;8:04-049. 25. Sartore-Bianchi A, Martini M, Molinari F, et al. PIK3CA mutations in colorectal cancer are associated with clinical resistance to GFR-targeted monoclonal antibodies. Cancer Res. 2009;69:85-857. 26. Barault L, Charon-Barra C, Jooste V, et al. Hypermethylator phenotype in sporadic colon cancer: study on a populationbased series of 582 cases. Cancer Res. 2008;68:854-8546. 27. Zlobec I, Bihl M, Foerster A, et al. Comprehensive analysis of CpG island methylator phenotype (CIMP)-high, -low, and -negative colorectal cancers based on protein marker expression and molecular features. J Pathol. 20;225:336-343. 28. Samowitz WS, Albertsen H, Herrick J, et al. valuation of a large, population-based sample supports a CpG island methylator phenotype in colon cancer. Gastroenterology. 2005;29:837-845. 29. Hughes LA, Simons CC, van den Brandt PA, et al. Body size, physical activity and risk of colorectal cancer with or without the CpG island methylator phenotype (CIMP). PLoS One. 20;6:e857. doi:0.37/journal.pone.00857. 30. Limsui D, Vierkant RA, Tillmans LS, et al. Cigarette smoking and colorectal cancer risk by molecularly defined subtypes. J Natl Cancer Inst. 200;02:02-022. 3. Rozek LS, Herron CM, Greenson JK, et al. Smoking, gender, and ethnicity predict somatic BRAF mutations in colorectal cancer. Cancer pidemiol Biomarkers Prev. 200;9:838-843. 32. Brink M, de Goeij AF, Weijenberg MP, et al. K-ras oncogene mutations in sporadic colorectal cancer in The Netherlands Cohort Study. Carcinogenesis. 2003;24:703-70. 33. nglish DR, Young JP, Simpson JA, et al. thnicity and risk for colorectal cancers showing somatic BRAF V600 mutation or CpG island methylator phenotype. Cancer pidemiol Biomarkers Prev. 2008;7:774-780. 34. Chen JT, Lane MA, Clark DP. Inhibitors of the polymerase chain reaction in Papanicolaou stain: removal with a simple destaining procedure. Acta Cytol. 996;40:873-877. 35. Uhlmann K, Brinckmann A, Toliat MR, et al. valuation of a potential epigenetic biomarker by quantitative methylsingle nucleotide polymorphism analysis. lectrophoresis. 2002;23:4072-4079. 36. Jordan JA, Butchko AR, Durso MB. Use of pyrosequencing of 6S rrna fragments to differentiate between bacteria responsible for neonatal sepsis. J Mol Diagn. 2005;7:05-0. 37. Garcia CA, Ahmadian A, Gharizadeh B, et al. Mutation detection by pyrosequencing: sequencing of exons 5-8 of the p53 tumor suppressor gene. Gene. 2000;253:249-257. 38. Nosho K, Kawasaki T, Ohnishi M, et al. PIK3CA mutation in colorectal cancer: relationship with genetic and epigenetic alterations. Neoplasia. 2008;0:534-54. 39. Colella S, Shen L, Baggerly KA, et al. Sensitive and quantitative universal pyrosequencing methylation analysis of CpG sites. Biotechniques. 2003;35:46-50. 40. Mikeska T, Bock C, l-maarri O, et al. Optimization of quantitative MGMT promoter methylation analysis using pyrosequencing and combined bisulfite restriction analysis. J Mol Diagn. 2007;9:368-38. 4. Irahara N, Nosho K, Baba Y, et al. Precision of pyrosequencing assay to measure LIN- methylation in colon cancer, normal colonic mucosa, and peripheral blood cells. J Mol Diagn. 200;2:77-83. 42. Ogino S, Chan AT, Fuchs CS, et al. Molecular pathological epidemiology of colorectal neoplasia: an emerging transdisciplinary and interdisciplinary field. Gut. 20;60:397-4. 43. arle JS, Luthra R, Romans A, et al. Association of microrna expression with microsatellite instability status in colorectal adenocarcinoma. J Mol Diagn. 200;2:433-440. 44. de la Chapelle A, Hampel H. Clinical relevance of microsatellite instability in colorectal cancer. J Clin Oncol. 200;28:3380-3387. 45. Pritchard CC, Grady WM. Colorectal cancer molecular biology moves into clinical practice. Gut. 20;60:6-29. 46. Murphy KM, Berg KD, Geiger T, et al. Capillary electrophoresis artifact due to eosin: implications for the interpretation of molecular diagnostic assays. J Mol Diagn. 2005;7:43-48. 47. Toyota M, Ahuja N, Ohe-Toyota M, et al. CpG island methylator phenotype in colorectal cancer. Proc Natl Acad Sci U S A. 999;96:868-8686. 48. Hinoue T, Weisenberger DJ, Lange CP, et al. Genome-scale analysis of aberrant DNA methylation in colorectal cancer. Genome Res. 202;22:27-282. doi:0.0/gr.7523.0. 49. Tanaka N, Huttenhower C, Nosho K, et al. Novel application of structural equation modeling to correlation structure analysis of CpG island methylation in colorectal cancer. Am J Pathol. 200;77:273-2740. 50. Lao VV, Grady WM. pigenetics and colorectal cancer. Nat Rev Gastroenterol Hepatol. 20;8:686-700. 5. Hughes LA, Khalid-de Bakker CA, Smits KM, et al. The CpG island methylator phenotype in colorectal cancer: progress and problems. Biochim Biophys Acta. 20;825:77-85. 52. Curtin K, Slattery ML, Samowitz WS. CpG island methylation in colorectal cancer: past, present and future. Patholog Res Int. 20;20:902674. doi:0.406/20/902674. 53. Ogino S, Nosho K, Kirkner GJ, et al. CpG island methylator phenotype, microsatellite instability, BRAF mutation and clinical outcome in colon cancer. Gut. 2009;58:90-96. 54. Ogino S, Galon J, Fuchs CS, et al. Cancer immunology: analysis of host and tumor factors for personalized medicine. Nat Rev Clin Oncol. 20;8:7-79. Am J Clin Pathol 202;38:22-29 29 29 DOI: 0.309/AJCP28LAOOKSZSVW 29