Elevated Microsatellite Alterations at Selected Tetranucleotides (EMAST) Is Not Attributed to MSH3 Loss in Stage I-III Colon cancer: An Automated, Digitalized Assessment by Immunohistochemistry of Whole Slides and Hot Spots

Martin M Watson, Dordi Lea, Hanne R Hagland, Kjetil Søreide, Martin M Watson, Dordi Lea, Hanne R Hagland, Kjetil Søreide

Abstract

Introduction: EMAST is a poorly understood form of microsatellite instability (MSI) in colorectal cancer (CRC) for which loss of MSH3 has been proposed as the underlying mechanism, based on experimental studies. We aimed to evaluate whether MSH3 loss is associated with EMAST in CRC.

Methods: A consecutive cohort of patients with stage I-III CRC. Digital image analysis using heatmap-derived hot spots investigated MSH3 expression by immunohistochemistry. Fragment analysis of multiplex PCR was used to assess MSI and EMAST, and results cross-examined with MSH3 protein expression.

Results: Of 152 patients, EMAST was found in 50 (33%) and exclusively in the colon. Most EMAST-positive cancers had instability at all 5 markers, and EMAST overlapped with MSI-H in 42/50 cases (84%). The most frequently altered tetranucleotide markers were D8S321 (38.2% of tumors) and D20S82 (34.4%). Subjective evaluation of MSH3 expression by IHC in tumor found ≤10% negative tumor cells in all samples, most being ≤5% negative. Digital analysis improved the detection but showed a similar spread of MSH3 loss (range 0.1-15.7%, mean 2.2%). Hotspot MSH3 negativity ranged between 0.1 to 95.0%, (mean 8.6%) with significant correlation with the whole slide analysis (Spearman's rho=0.677 P<.001). Loss of MSH3 expression did not correlate with EMAST.

Conclusions: In a well-defined cohort of patients with CRC, loss of MSH3 was not associated with EMAST. Further investigation into the mechanisms leading to EMAST in CRC is needed.

Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1
Figure 1
MSH3 immunohistochemistry virtual image analysis process. (A-C) An initial gross exclusion of stroma, tissue folds and normal tissue is carried out, selecting a “work area” where an app-based algorithm is then run to specifically mark tumor cells and exclude stroma. This results in a highlighted region of interest (ROI, marked in blue); (D) A second app-based algorithm classifies cells on the basis of their positivity (green) or negativity (blue) for the MSH3 protein; (E-F) A heatmap is created to highlight areas on the whole slide where the highest concentration of negative cells are located; (G) Based on the heatmap created in (E), a 0.8 mm2 round ROI (hotspot, light blue circle) is placed and becomes the focus of the analysis; (H) Fully classified, hotspot-derived ROI.
Figure 2
Figure 2
EMAST analysis. Electropherograms of multiplex PCR fragment analysis are shown for (A) an EMAST-negative and (B) an EMAST-positive patient. Arrows indicate extra PCR products at +/− 4n bp in unstable markers. (C) Stacked bar population graph showing frequency of instability (red) at each EMAST marker, in EMAST-positive and EMAST-negative populations. (D) bar chart showing proportions of patients grouped by total number of unstable EMAST markers. For patients bearing only 1 marker mutated (not EMAST, according to our thresholds) the bar is stacked to specify each marker's abundance.
Figure 3
Figure 3
Digitalized evaluation of MSH3 expression. (A) The entire cohort was scored by an experienced pathologist as >90% positive for MSH3 (majority >95%). (B) With the help of digitalized hotspot analysis, small areas where a higher proportion of negative cells were found in all the slides. (C) Within most MSH3-positive cells, some nuclei exhibited heterogeneous staining.

References

    1. Boland C.R., Goel A. Microsatellite instability in colorectal cancer. Gastroenterology. 2010;138(6):2073–2087. e2073.
    1. Soreide K., Janssen E.A., Soiland H., Korner H., Baak J.P. Microsatellite instability in colorectal cancer. Br J Surg. 2006;93(4):395–406.
    1. Dienstmann R., Mason M.J., Sinicrope F.A., Phipps A.I., Tejpar S., Nesbakken A., Danielsen S.A., Sveen A., Buchanan D.D., Clendenning M. Prediction of overall survival in stage II and III colon cancer beyond TNM system: a retrospective, pooled biomarker study. Ann Oncol. 2017;28(5):1023–1031.
    1. Domingo E., Camps C., Kaisaki P.J., Parsons M.J., Mouradov D., Pentony M.M., Makino S., Palmieri M., Ward R.L., Hawkins N.J. Mutation burden and other molecular markers of prognosis in colorectal cancer treated with curative intent: results from the QUASAR 2 clinical trial and an Australian community-based series. Lancet Gastroenterol Hepatol. 2018;3(9):635–643.
    1. Kang S., Na Y., Joung S.Y., Lee S.I., Oh S.C., Min B.W. The significance of microsatellite instability in colorectal cancer after controlling for clinicopathological factors. Medicine (Baltimore) 2018;97(9) e0019.
    1. Guinney J., Dienstmann R., Wang X., Reyniès Ad, Schlicker A., Soneson C., Marisa L., Roepman P., Nyamundanda G., Angelino P. The consensus molecular subtypes of colorectal cancer. Nat Med. 2015;21(11):1350–1356.
    1. Watson M.M., Berg M., Soreide K. Prevalence and implications of elevated microsatellite alterations at selected tetranucleotides in cancer. Br J Cancer. 2014;111(5):823–827.
    1. Boland C.R., Thibodeau S.N., Hamilton S.R., Sidransky D., Eshleman J.R., Burt R.W., Meltzer S.J., Rodriguez-Bigas M.A., Fodde R., Ranzani G.N. A National Cancer Institute Workshop on Microsatellite Instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res. 1998;58(22):5248–5257.
    1. Suraweera N., Duval A., Reperant M., Vaury C., Furlan D., Leroy K., Seruca R., Iacopetta B., Hamelin R. Evaluation of tumor microsatellite instability using five quasimonomorphic mononucleotide repeats and pentaplex PCR. Gastroenterology. 2002;123(6):1804–1811.
    1. Haugen A.C., Goel A., Yamada K., Marra G., Nguyen T.-P., Nagasaka T., Kanazawa S., Koike J., Kikuchi Y., Zhong X. Genetic instability caused by loss of MutS homologue 3 in human colorectal cancer. Cancer Res. 2008;68(20):8465–8472.
    1. Campregher C., Schmid G., Ferk F., Knasmüller S., Khare V., Kortüm B., Dammann K., Lang M., Scharl T., Spittler A. MSH3-deficiency initiates EMAST without oncogenic transformation of human colon epithelial cells. PLoS ONE. 2012;7(11) e50541.
    1. Tseng-Rogenski S.S., Chung H., Wilk M.B., Zhang S., Iwaizumi M., Carethers J.M. Oxidative stress induces nuclear-to-cytosol shift of hMSH3, a potential mechanism for EMAST in colorectal cancer cells. PLoS ONE. 2012;7(11) e50616.
    1. Tseng-Rogenski S., Hamaya Y., Choi D.Y., Carethers J.M. Interleukin 6 alters localization of hMSH3, leading to DNA mismatch repair defects in colorectal cancer cells. Gastroenterology. 2015;148(3):579–589.
    1. The Cancer Genome Atlas Network. Muzny D.M., Bainbridge M.N., Chang K., Dinh H.H., Drummond J.A., Fowler G., Kovar C.L., Lewis L.R., Morgan M.B. Comprehensive molecular characterization of human colon and rectal cancer. Nature. 2012;487:330.
    1. Kim T.-M., Laird Peter W., Park Peter J. The landscape of microsatellite instability in colorectal and endometrial cancer genomes. Cell. 2013;155(4):858–868.
    1. Lee S.Y., Chung H., Devaraj B., Iwaizumi M., Han H.S., Hwang D.Y., Seong M.K., Jung B.H., Carethers J.M. Microsatellite alterations at selected tetranucleotide repeats are associated with morphologies of colorectal neoplasias. Gastroenterology. 2010;139(5):1519–1525.
    1. Venderbosch S., van Lent-van Vliet S., de Haan A.F., Ligtenberg M.J., Goossens M., Punt C.J., Koopman M., Nagtegaal I.D. EMAST is associated with a poor prognosis in microsatellite instable metastatic colorectal cancer. PLoS One. 2015;10(4) e0124538.
    1. Soreide K., Watson M.M., Lea D., Nordgard O., Soreide J.A., Hagland H.R. Assessment of clinically related outcomes and biomarker analysis for translational integration in colorectal cancer (ACROBATICC): study protocol for a population-based, consecutive cohort of surgically treated colorectal cancers and resected colorectal liver metastasis. J Transl Med. 2016;14(1):192.
    1. Watson M.M., Lea D., Rewcastle E., Hagland H.R., Søreide K. Elevated microsatellite alterations at selected tetranucleotides in early-stage colorectal cancers with and without high-frequency microsatellite instability: same, same but different? Cancer Med. 2016;5(7):1580–1587.
    1. Buhard O., Suraweera N., Lectard A., Duval A., Hamelin R. Quasimonomorphic mononucleotide repeats for high-level microsatellite instability analysis. Disease Markers. 2004;20(4-5):251–257.
    1. Soreide K. High-fidelity of five quasimonomorphic mononucleotide repeats to high-frequency microsatellite instability distribution in early-stage adenocarcinoma of the colon. Anticancer Res. 2011;31(3):967–971.
    1. Koi M., Tseng-Rogenski S.S., Carethers J.M. Inflammation-associated microsatellite alterations: mechanisms and significance in the prognosis of patients with colorectal cancer. World Journal of Gastrointestinal Oncology. 2018;10(1):1–14.
    1. Stalhammar G., Robertson S., Wedlund L., Lippert M., Rantalainen M., Bergh J., Hartman J. Digital image analysis of Ki67 in hot spots is superior to both manual Ki67 and mitotic counts in breast cancer. Histopathology. 2018;72(6):974–989.
    1. Norwegian Health Council . 2018. Nasjonalt handlingsprogram med retningslinjer for diagnostikk, behandling og oppfølging av pasienter med brystkreft.
    1. Danish Breast Cancer Cooperative Group . 2017. Retningslinjer for brystkræft.
    1. Swedish Society of Pathology . 2018. Kvalitetsdokument för patologi.
    1. Pantanowitz L.R., David L. Imaging and Quantitative Immunohistochemistry. In: Dabbss David J., editor. Diagnostic Immunohistochemistry. 5th ed. Elsevier; Philadelphia, PA, USA: 2019.
    1. Bussolati G., Leonardo E. Technical pitfalls potentially affecting diagnoses in immunohistochemistry. J Clin Pathol. 2008;61(11):1184.
    1. Cartun R.W.T., Dabbs Clive R., David J. Techniques of immunohistochemistry: Principles, Pitfalls, and Standardization. In: Dabbss David J., editor. Diagnostic Immunohistochemistry. 5th ed. Elsevier; Philadelphia, PA, USA: 2019.
    1. Mori T., Hamaya Y., Uotani T., Yamade M., Iwaizumi M., Furuta T., Miyajima H., Osawa S., Sugimoto K. Prevalence of elevated microsatellite alterations at selected tetranucleotide repeats in pancreatic ductal adenocarcinoma. PLoS One. 2018;13(12) e0208557.
    1. Choi Y.D., Choi J., Kim J.H., Lee J.S., Lee J.H., Choi C., Choi H.S., Lee M.C., Park C.S., Juhng S.W. Microsatellite instability at a tetranucleotide repeat in type I endometrial carcinoma. J Exp Clin Cancer Res. 2008;27:88.
    1. Devaraj B., Lee A., Cabrera B.L., Miyai K., Luo L., Ramamoorthy S., Keku T., Sandler R.S., McGuire K.L., Carethers J.M. Relationship of EMAST and microsatellite instability among patients with rectal cancer. J Gastrointest Surg. 2010;14(10):1521–1528.
    1. Carethers J.M., Jung B.H. Genetics and genetic biomarkers in sporadic colorectal cancer. Gastroenterology. 2015;149(5):1177–1190. e1173.
    1. Lee H.S., Park K.U., Kim D.W., Lhn M.H., Kim W.H., Seo A.N., Chang H.E., Nam S.K., Lee S.Y., Oh H.K. Elevated microsatellite alterations at selected tetranucleotide repeats (EMAST) and microsatellite instability in patients with colorectal cancer and its clinical features. Curr Mol Med. 2016;16(9):829–839.

Source: PubMed

3
订阅