Article
Original Article
Aditya Agnihotri1, Anita P Javalgi*,2, Ajay Kumar Oli3, Vidisha Athanikar4,

1Department of Pathology, SDM College of Medical Sciences and Hospital, Shri Dharmasthala Manjunatheshwara University, Dharwad, Karnataka, India.

2Dr. Anita P Jawalgi, Department of Pathology, SDM College of Medical Sciences and Hospital, Shri Dharmasthala Manjunatheshwara University, Sattur, Dharwad, Karnataka, India.

3Department of Pathology, SDM College of Medical Sciences and Hospital, Shri Dharmasthala Manjunatheshwara University, Dharwad, Karnataka, India.

4Department of Pathology, SDM College of Medical Sciences and Hospital, Shri Dharmasthala Manjunatheshwara University, Dharwad, Karnataka, India.

*Corresponding Author:

Dr. Anita P Jawalgi, Department of Pathology, SDM College of Medical Sciences and Hospital, Shri Dharmasthala Manjunatheshwara University, Sattur, Dharwad, Karnataka, India., Email: anitajawalgi@gmail.com
Received Date: 2023-06-26,
Accepted Date: 2023-07-14,
Published Date: 2023-10-31
Year: 2023, Volume: 13, Issue: 4, Page no. 175-180, DOI: 10.26463/rjms.13_4_5
Views: 236, Downloads: 16
Licensing Information:
CC BY NC 4.0 ICON
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0.
Abstract

Background and aim: Colorectal adenocarcinoma is a major cause of morbidity and mortality worldwide. Mismatch repair (MMR) protein expression plays a crucial role in the development and progression of colorectal adenocarcinomas. Immunohistochemistry (IHC) is a widely used technique to assess MMR protein expression. However, there is limited data on the prospective evaluation of MMR protein expression by IHC on endoscopic biopsies of colorectal adenocarcinomas. In this study, we aimed to evaluate the MMR protein expression by IHC on endoscopic biopsies of colorectal adenocarcinomas at a tertiary care center.

Methods: Forty-Two colonoscopic endoscopic biopsies pathologically diagnosed as colorectal adenocarcinoma were analyzed for IHC expression of MMR proteins, MLH1, PMS2, MSH2 and MSH6.

Results: Out of the 42 cases, 22 (52.3%) were mismatch repair proficient (pMMR), while the remaining 20 (47.6%) were mismatch repair deficient (dMMR). MLH1+PMS2- was the most common pattern accounting for 20% of dMMR cases and MLH1-PMS2-MSH2-MSH6- or the “Null Pattern” was the least common with 10% of dMMR cases.

Conclusion: Evaluation of MMR proteins using immunohistochemistry is relatively easy in routine testing and is a useful tool in detecting MSI in CRC.  

<p><strong>Background and aim:</strong> Colorectal adenocarcinoma is a major cause of morbidity and mortality worldwide. Mismatch repair (MMR) protein expression plays a crucial role in the development and progression of colorectal adenocarcinomas. Immunohistochemistry (IHC) is a widely used technique to assess MMR protein expression. However, there is limited data on the prospective evaluation of MMR protein expression by IHC on endoscopic biopsies of colorectal adenocarcinomas. In this study, we aimed to evaluate the MMR protein expression by IHC on endoscopic biopsies of colorectal adenocarcinomas at a tertiary care center.</p> <p><strong>Methods: </strong>Forty-Two colonoscopic endoscopic biopsies pathologically diagnosed as colorectal adenocarcinoma were analyzed for IHC expression of MMR proteins, MLH1, PMS2, MSH2 and MSH6.</p> <p><strong>Results:</strong> Out of the 42 cases, 22 (52.3%) were mismatch repair proficient (pMMR), while the remaining 20 (47.6%) were mismatch repair deficient (dMMR). MLH1+PMS2- was the most common pattern accounting for 20% of dMMR cases and MLH1-PMS2-MSH2-MSH6- or the &ldquo;Null Pattern&rdquo; was the least common with 10% of dMMR cases.</p> <p><strong>Conclusion:</strong> Evaluation of MMR proteins using immunohistochemistry is relatively easy in routine testing and is a useful tool in detecting MSI in CRC.&nbsp;&nbsp;</p>
Keywords
Colorectal cancer, Mismatch repair (MMR), Microsatellite instability (MSI)
Downloads
  • 1
    FullTextPDF
Article
Introduction

Colorectal cancer (CRC) being the third most frequent cancer in men and the second most common in women worldwide, is a major contributor to cancer-related mortality.1 The highest age-adjusted incidence rate for CRCs was recorded as 4.1 for men and 5.2 for women in India, where it ranks ninth among the most frequent cancers in both sexes.2 The most frequent pathway for the development of colorectal neoplasia is chromosomal instability (75%), followed by the microsatellite instability (MSI) pathway (15%).3,4 The CpG island methylator phenotype, which results in epigenetic gene silencing in 10% of CRCs, is the third main route.4-6 Lynch syndrome (LS) or hereditary nonpolyposis colorectal cancer (HNPCC) accounts for about 15% of the MSI-related malignancies, while the remaining 85% are sporadic.3,4 

DNA mismatch repair (MMR) is a vital step in the cell cycle to correct mistakes in DNA replication. Loss of MMR pathway function results from mutations in MMR genes. Because the MMR system is ineffective at repairing replication-associated errors, mismatch mutations can persist throughout the genome, but especially in microsatellite regions, which are repetitive DNA structures. This instability in the microsatellite region causes the phenomenon of microsatellite instability (MSI), which leads to the rapid accumulation of new mutations and the emergence of carcinoma. MLH1, MSH2, MSH6, and PMS2 are the four DNA mismatch repair genes that are most frequently mutated and they function as a heterodimer. MSH2 and MSH6 are heterodimer where MSH2 is the dominant one i.e. loss of MSH2 leads to degradation of MSH6, but not the vice versa. Similarly MLH1 and PMS2 are heterodimer where MLH1 is the dominant one i.e. loss of MLH1 leads to degradation of PMS2, but not the vice versa. Defective MMR gene can be detected via immunohistochemistry and its absence is a surrogate marker for microsatellite instability. Testing for MSI (thus classifying microsatellite stable, MSI–H and MSI–L) is done by molecular testing of at least five loci recommended by National cancer institute using Polymerase Chain Reaction (PCR) or next-generation sequencing (NGS) technique.7

In 1996, Leach et al. developed monoclonal antibodies that identified the MSH2 protein in DNA mismatch-proficient cell lines 1, and this sparked a number of studies on the use of immunohistochemical detection of DNA mismatch repair (MMR) proteins in the recognition of colorectal tumors with microsatellite instability (MSI).8 The fundamental flaw in MSI has been discovered to be faulty DNA MMR function as a result of either germline mutation of one of multiple MMR genes, including MLH1, PMS2, MSH2, and MSH6 or abnormal methylation of the promoter of MLH1. Also, detection of MMR using Immunohistochemistry (IHC) is cost effective and easily available. Specific staining is performed on tumor tissue for each of the four mismatch repair proteins; mutations in MMR genes typically lead to nonfunctional proteins that do not stain by IHC suggestive of defective DNA mismatch repair within the tumor.

Detection of CRC with dMMR proteins has clinical importance in terms of prognostic value with better prognosis and reduced recurrence rates,9 as a predictive marker of response to 5FU chemotherapy specially in colorectal carcinomas and lynch syndrome screening tool.10

Immunohistochemical (IHC) analysis of MMR proteins is a promising approach to identify patients with CRC and MSI. This study aimed to evaluate MSI status using the IHC expression pattern of four MMR proteins in CRC endoscopic samples.

Materials and Methods

Forty-two colonoscopic endoscopic biopsies pathologically diagnosed as CRC were collected from the Department of Pathology, between June 2019 and June 2021. All samples satisfied the following criteria: (1) sporadic colon or rectal cancer confirmed by pathological diagnosis, (2) no preoperative therapy. Biopsies with inadequate tissue were excluded from the study. The patients’ demographic records, clinical details were retrieved from our digital archive. The study protocol was approved by the institutional review board, and written informed consent was obtained from all the participants.

IHC procedure and interpretation

Four µm thick sections were taken on positively charged slides. After overnight incubation at 37ºC and deparaffinization with repeated washes of xylene (10 min each), rehydration of tissues with graded alcohol was done. The slides were then washed with running water (10 min) and distilled water (5 min). Antigen retrieval with citrate or EDTA buffer at 95ºC was done and cooled at room temperature. Peroxide block was carried out for 10–15 min and washed in Phosphate buffer solution (PBS) for 5 min and incubated with Monoclonal Rabbit Anti-Human MutS Protein Homolog 6 Clone EP49 for MSH6, Monoclonal Mouse Anti-Human MutS Protein Homolog 2 Clone FE11 for MSH2, Monoclonal Mouse Anti-Human Mut L Protein Homolog 1 Clone ES05 for MLH1, Monoclonal Rabbit Anti-Human post meiotic segregation increased 2, clone EP51 for PMS2 for 45 min. Subsequently, tissues were incubated with Poly Excel Target Binder for 15 to 20 min and washed with PBS for five minutes. Again, incubation was done with Poly Excel Poly H.R.P (Horse Radish Peroxidase) for 15 to 20 min, washed with PBS for five minutes and developed with DAB (Diaminobenzidine) Chromogen for 5-8 minutes. Sections were counterstained with haematoxylin after washing with running water. Slides were mounted and observed under light microscope for interpretation. Normal colonic mucosa, lymphocytes and stromal cells were used as positive control.

The H&E slides for morphological features and the IHC results were interpreted by the consultant pathologists using a Nikon microscope. Reporting was done following the College of American Pathologists (CAP) protocol of biomarker testing for MMR protein. Any amount of intact nuclear staining by IHC was considered as positive (intact expression), cases with weak and focal nuclear positive were also considered as positive, but noted separately. The loss of nuclear staining by IHC with positive internal control (lymphocytes, normal colonic mucosa and stromal cells) was considered as loss of expression. All cases were categorized into one of the following six patterns: MLH1- PMS2-, MSH2+MSH6-, MSH2-MSH6-, MLH1+PMS2-, MLH1-PMS2-MSH2-MSH6, MLH1+ PMS2-MSH2+ MSH6-.

Furthermore, if all four markers showed intact nuclear expression, the biopsy was classified as Microsatellite Stable (MSS). If only one marker showed loss of expression, it was labelled as deficient MMR (dMMR) and categorized as Microsatellite instability -Low (MSI-L), and if two or more makers showed loss of expression, the tissue was dMMR and categorized as Microsatellite instability - High (MSI-H).

Descriptive statistics were used to summarize the demographic characteristics of the study population. All statistical studies were performed using IBM SPSS Version 29. The frequencies and percentages of positive and negative MMR protein expression were calculated.

Results

Forty-two colonoscopic biopsies with colorectal adenocarcinoma cases were studied. There were 27 males and 15 females (Figure 1) with a mean age of 56.65±15.4 years. Out of the 42 cases, 22 (52.3%) were mismatch repair proficient (pMMR), while the remaining 20 (47.6%) were mismatch repair deficient (dMMR). MSI-L accounted for six cases (14%) and MSI-H accounted for the remaining 14 cases (33%) (Figure 2). The mean age for dMMR cases was 53.5 years, while that of pMMR was 54 years.

Mismatch repair protein expression for MLH1, PMS2, MSH2 and MSH6 on Immunohistochemistry has been depicted in Table 1.

The pattern of expression was further classified into one of the following patterns: MLH1- PMS2-, MSH2+ MSH6-, MSH2-MSH6-, MLH1+PMS2-, MLH1 PMS2- MSH2-MSH6-, MLH1+PMS2-MSH2+ MSH6- as depicted in Figure 3. MLH1+PMS2- was the most common pattern accounting for 20% for dMMR cases and MLH1-PMS2-MSH2-MSH6- or the “Null IHC/ Pattern” was the least common with 10% of dMMR cases.

Figure 4 showcases a 62-year-old male with colorectal adenocarcinoma with pMMR immunohistochemistry status and was Microsatellite stable (MSS) with positive IHC expression for all four MMR proteins.

Figure 5 represents a 68-year-old male with colorectal adenocarcinoma with dMMR Immunohistochemistry status and was MSI-H with negative IHC expression of all four MMR proteins.

Discussion

The annual incidence of CRC worldwide raises a serious public health concern. Familial CRC represents 20% of all CRC and Lynch syndrome represents 3.5% of all CRC. These figures along with the recognition of the role of genomic instability in initiation and progression in CRC, raises the need for molecular screening of all CRC for Lynch syndrome.1 According to a study by Hashmi et al. (2017), the gold standard for identifying the MSI phenotype is still PCR amplification of microsatellite repeats, although this method is impractical for use in ordinary pathology labs.11 As a result, Hampel (2018) advised IHC be used to screen for Lynch syndrome because it is equally sensitive to PCR, is less expensive, is more widely available, and predicts the nonworking gene, which helps to limit the number of genes that need to be sequenced.12

The pathogenesis of colorectal cancer is still unclear, but it could be generally defined in two molecular pathways of genomic instability. One is chromosome instability, which is involved in certain oncogenes and tumour suppressor genes, such as APC, KRAS, and TP53. Another way is microsatellite instability (MSI), which is due to mis-match repair system defect, and it accounts for approximately 15% of all colorectal carcinomas. Microsatellites are simple repetitive sequences of DNA that are scattered throughout the genome. These sequences are stably inherited, vary from individual to individual, and have a low alteration rate. Instability within these sequences has been recognized as a marker for genome-wide mutations and DNA repair deficiencies. MSI has been detected in cancers associated with hereditary non-polyposis colorectal cancer syndrome (HNPCC), as well as in a variety of sporadic cancers, including colorectal cancers associated with ulcerative colitis (UC).13,14

Mismatch repair proteins are byproducts of MMR genes that are involved in the repair of mismatched bases that may have occurred during DNA replication, genetic recombination, chemical or physical damage.15 The effects of these mutations include a failure to recognize and repair mismatched nucleotides, as well as insertion/deletion loops induced by slippage of DNA polymerase.16 As a result, in tumors attributable to MMR protein deficiency, there are insertion and deletion mutations in stretches of short tandem DNA repeats (microsatellites) as well as nucleotide substitutions throughout the genome.17 Such CRC tends to display high levels of microsatellite instability (MSI-H), whereas in tumors with proficient MMR proteins, they may have either low levels of microsatellite instability (MSI-L) or microsatellite stable (MSS).17

In our study, out of the 42 colonic adenocarcinoma cases, 20 cases i.e. 47.6% of colonic adenocarcinoma were dMMR. In similar studies, Kalita Lachit et al. found dMMR in 27.1% of CRC.7 Pandey et al. using two markers MLH1 and MSH2 found 15.7% of colorectal carcinoma cases with dMMR.18 Singh et al. performed two marker MLH1 and MSH2 and observed MMR deficient in 63.3% cases.19 Nayak et al. performed IHC in total 236 cases of colorectal carcinomas using all four MMR proteins and found 22.9% cases with dMMR.20

A study conducted by Kumar A et al. in North India on pattern of mismatch repair protein loss and its clinicopathological correlation in colorectal cancer demonstrated high frequency of MMR protein loss (29%) in colorectal cancer in North Indian patients which was more common in right colon cancers and the sensitivity of IHC to detect MMR loss was 90-92% with use of all four antibodies (MLH1, MSH2, MSH6, PMS2).21

Regarding pattern of MMR protein loss, in our study, the loss of nuclear expression of PMS2 was the most common, followed by loss of MSH6, MSH 2 and MLH1 expression which is similar to most other studies.22,23

In two cases, there was absence of all four MMR proteins; such “null IHC/null pattern” phenotype is very rare. Wang et al. reported one CRC case which was associated with concurrent somatic inactivation of MLH1 and MSH2.24 Hagen et al. also reported one such “null pattern” IHC in a CRC which was associated with germline MSH2 mutation and somatic MLH1 hypermethylation.25

As a useful predictive and prognostic biomarker in CRC, immunohistochemical analysis of MMR proteins is reasonably simple to incorporate into routine testing in laboratories. Before assessing MMR proteins by IHC, tissue fixation and processing must follow established protocols to prevent antigen loss owing to procedural mistakes. In order to effectively manage patients, screen for Lynch syndrome, and find families with predisposed germ line mutations. There is a need for more extensive testing of MMR protein using IHC as a result of the introduction of immunological check point blocking therapy.

Conclusion

Our findings and the previous reports point out high incidence of MSI in our population and highlights the importance of molecular screening of patients with colorectal cancer for MSI using immunohistochemistry. The limitation of this study was the small sample size. A larger, multicenter study comparing the MMR protein expression using immunohistochemistry and comparing it with the gold standard MSI genetic analysis by polymerase chain reaction is recommended.

Conflict of interest

Nil

Source of funding

The study received funding from RGUHS research grant.

Supporting File
References
  1. Cancer Today - International agency for research on cancer. Available at: https://gco.iarc.fr/today/data/ factsheets/cancers/10_8_9-Colorectum-fact-sheet. pdf (Accessed: 25 June 2023). 
  2. Roy P, Nayak S, Arora N, Arun I, Roy M, Banerjee S, et al. Prevalence estimation of microsatellite instability in colorectal cancers using tissue microarray-based methods - a tertiary care center experience. Indian J Pathol Microbiol 2018;61 (4):520. 
  3. Samowitz WS, Curtin K, Ma KN, Schaffer D, Coleman LW, Leppert M, et al. Microsatellite instability in sporadic colon cancer is associated with an improved prognosis at the population level. Cancer Epidemiol Biomarkers Prev 2001;10: 917-23. 
  4. Sinicrope FA, Sargent DJ. Molecular pathways: Microsatellite instability in colorectal cancer: Prognostic, predictive, and therapeutic implications. Clin Cancer Res 2012;18:1506-12. 
  5. Al-Sohaily S, Biankin A, Leong R, Kohonen-Corish M, Warusavitarne J. Molecular pathways in colorectal cancer. J Gastroenterol Hepatol 2012;27:1423-31. 
  6. Hughes LA, Khalid-de Bakker CA, Smits KM, van den Brandt PA, Jonkers D, Ahuja N, et al. The cpG island methylator phenotype in colorectal cancer: Progress and problems. Biochim Biophys Acta 2012;1825:77-85. 
  7. Lachit K, Pant V. Study of mismatch repair protein expression by using immunohistochemistry in various carcinomas with special reference to colorectal adenocarcinomas - at a tertiary referral laboratory in India. APJCB 2022;7(4):3410-7. 
  8. Leach FS, Polyak K, Burrell M, Johnson KA, Hill D, Dunlop MG, et al. Expression of the human mismatch repair gene hMSH2 in normal and neoplastic tissues. Cancer Res 1996;56(2):235-40. 
  9. Legolvan MP, Taliano RJ, Resnick MB. Application of molecular techniques in the diagnosis, prognosis and management of patients with colorectal cancer: A practical approach. Hum Pathol 2012;43: 1157-68. 
  10. Sinicrope FA, Foster NR, Thibodeau SN, Marsoni S, Monges G, Labianca R, et al. DNA mismatch repair status and colon cancer recurrence and survival in clinical trials of 5-fluorouracil-based adjuvant therapy. J Natl Cancer Inst 2011;103: 863-75. 
  11. Hashmi AA, Ali R, Hussain ZF, Faridi N, Khan EY, Bakar SM, et al. Mismatch repair deficiency screening in colorectal carcinoma by a four-antibody immunohistochemical panel in Pakistani population and its correlation with histopathological parameters. World J Surg Oncol 2017;15(1):116. 
  12. Hampel H. Population screening for hereditary colorectal cancer. Surg Oncol Clin N Am 2018; 27(2):319-25. 
  13. Jefferys AJ, Wilson V, Neumann R, Keyte J. Amplification of human minisatellites by the polymerase chain reaction: towards DNA fingerprinting of single cells. Nucleic Acids Res 1998;16:10953-71.
  14. Weber JL, May PE. Abundant class of human DNA polymorphisms which can be typed using the polymerase chain reaction. Am J Hum Genet 1989;44:388-96. 
  15. Richman S. Deficient mismatch repair: Read all about it (Review). Int J Oncol 2015;47:1189-202.
  16. Pino MS, Mino-Kenudson M, Wildemore BM, Ganguly A, Batten J, Sperduti I, et al. Deficient DNA mismatch repair is common in Lynch Syndrome-associated colorectal adenomas. J Mol Diagn 2009;11:238-47. 
  17. Kheirelseid EAH, Miller N, Chang KH, Curran C, Hennessey E, Sheehan M, et al. Mismatch repair protein expression in colorectal cancer J Gastrointest Oncol 2013;4:397-408. 
  18. Pandey V, Prabhu JS, Payal K, Rajan V, Deepak C, Barde S, et al. Assessment of microsatellite instability in colorectal carcinoma at an Indian center. Int J Colorectal Dis 2007;22(7):777-782. 
  19. Singh P, Sharma K, Tewari R, Misra V, Dwivedi M, Misra SP. Clinico-histopathological features of colonic adenocarcinoma in young (< 50 years) patients with special reference to microsatellite instability status. Indian J Med Res Pharm Sci 2015;2:9-18. 
  20. Nayak SS, Roy P, Arora N, Arun I, Roy MK, Banerjee S, et al. Prevalence estimation of microsatellite instability in colorectal cancers using tissue microarray-based methods - A tertiary care center experience. Indian J Pathol Microbiol 2018;61(4):520-525. 
  21. Kumar A, Jain M, Saxena R, Yadav A, Kumari N, Krishnani N. Pattern of mismatch repair protein loss and its clinicopathological correlation in colorectal cancer in North India. S Afr J Surg 2018;56(1): 25-29. 
  22. Hall G, Clarkson A, Shi A, Langford E, Leung H, Eckstein RP, et al. Immunohistochemistry for PMS2 and MSH6 alone can replace a four antibody panel for mismatch repair deficiency screening in colorectal adenocarcinoma. Pathology 2010;42(5):409-413. 
  23. Watson N, Grieu F, Morris M, Harvey J, Stewart C, Schofield L, et al. Heterogeneous staining for mismatch repair proteins during population- based prescreening for hereditary nonpolyposis colorectal cancer. J Mol Diagn 2007;9(4):472-478. 
  24. Wang T, Stadler ZK, Zhang L, Weiser MR, Basturk O, Hechtman JF, et al. Immunohistochemical null phenotype for mismatch repair proteins in colonic carcinoma associated with concurrent MLH1 hypermethylation and somatic MSH2 mutations. Fam Cancer 2018 04;17(2):225-228. 
  25. Hagen CE, Lefferts J, Hornick JL, Srivastava A. “Null pattern” of immunoreactivity in a Lynch syndrome-associated colon cancer due to germline MSH2 mutation and somatic MLH1 hypermethylation. Am J Surg Pathol 2011;35 (12):1902-5.
We use and utilize cookies and other similar technologies necessary to understand, optimize, and improve visitor's experience in our site. By continuing to use our site you agree to our Cookies, Privacy and Terms of Use Policies.