Next Article in Journal
Deregulation of Transcriptional Enhancers in Cancer
Previous Article in Journal
Identification and Roles of miR-29b-1-3p and miR29a-3p-Regulated and Non-Regulated lncRNAs in Endocrine-Sensitive and Resistant Breast Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Tetranucleotide and Low Microsatellite Instability Are Inversely Associated with the CpG Island Methylator Phenotype in Colorectal Cancer

1
Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
2
The Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW 2037, Australia
3
Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
4
Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC 3000, Australia
5
Department of Clinical Pathology, University of Melbourne, Melbourne, VIC 3010, Australia
6
University of Melbourne Centre for Cancer Research, University of Melbourne, Melbourne, VIC 3010, Australia
7
Genomic Medicine and Family Cancer Clinic, Royal Melbourne Hospital, Melbourne, VIC 3010, Australia
8
Department of Oncology, Monash University and Monash Health, Melbourne, VIC 3168, Australia
9
ACT Pathology, The Canberra Hospital and Australian National University Medical School, Canberra, ACT 2605, Australia
10
Microbiome Research Centre, St George & Sutherland Clinical School, UNSW Sydney, Sydney, NSW 2217, Australia
11
School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
*
Author to whom correspondence should be addressed.
These authors contributed equally.
Submission received: 10 June 2021 / Revised: 8 July 2021 / Accepted: 8 July 2021 / Published: 14 July 2021
(This article belongs to the Section Cancer Biomarkers)

Abstract

:

Simple Summary

A type of DNA mismatch repair defect known as “elevated microsatellite alterations at selected tetranucleotide repeats” (EMAST) is found across many different cancers. Tetranucleotide microsatellite instability, which is caused by MSH3 mismatch repair gene/protein loss-of-function, shares a molecular basis with “low microsatellite instability” (MSI-L) in colorectal cancer. Tetranucleotide microsatellite instability is also a byproduct of “high microsatellite instability” (MSI-H) that arises from deficiency of mismatch repair due to MSH2, MSH6, MLH1 or PMS2 gene alterations. MSH3-related EMAST is emerging as a biomarker of poor prognosis in colorectal cancer and needs to be clearly differentiated from MSI-H. Here, we show that tumours with non-MSI-H-related EMAST or MSI-L rarely show concordant promoter methylation of multiple marker genes. Colorectal tumours that are positive for a single (1/5) tetranucleotide repeat marker are an important subset of the EMAST spectrum.

Abstract

MSH3 gene or protein deficiency or loss-of-function in colorectal cancer can cause a DNA mismatch repair defect known as “elevated microsatellite alterations at selected tetranucleotide repeats” (EMAST). A high percentage of MSI-H tumors exhibit EMAST, while MSI-L is also linked with EMAST. However, the distribution of CpG island methylator phenotype (CIMP) within the EMAST spectrum is not known. Five tetranucleotide repeat and five MSI markers were used to classify 100 sporadic colorectal tumours for EMAST, MSI-H and MSI-L according to the number of unstable markers detected. Promoter methylation was determined using methylation-specific PCR for MSH3, MCC, CDKN2A (p16) and five CIMP marker genes. EMAST was found in 55% of sporadic colorectal carcinomas. Carcinomas with only one positive marker (EMAST-1/5, 26%) were associated with advanced tumour stage, increased lymph node metastasis, MSI-L and lack of CIMP-H. EMAST-2/5 (16%) carcinomas displayed some methylation but MSI was rare. Carcinomas with 3 positive EMAST markers (13%) were more likely to have a proximal colon location and be MSI-H and CIMP-H. Our study suggests that EMAST/MSI-L is a valuable prognostic and predictive marker for colorectal carcinomas that do not display the high methylation phenotype CIMP-H.

1. Introduction

Microsatellite instability (MSI) in cancer is caused by deficient repair of insertion–deletion loop mismatches, which occur during DNA replication in tandem repeat sequences throughout the genome [1,2]. MSI-H is caused by DNA mismatch repair (MMR) deficiency as a result of alterations in the MLH1, MSH2, MSH6 or PMS2 genes. Another form of MSI is “elevated microsatellite alterations at selected tetranucleotide repeats” (EMAST) that has been identified in up to 60% of colorectal cancers [3,4,5,6], but no standard marker panel has been adopted yet for its detection. EMAST is caused by a deficiency or dysfunction of MMR involving the MSH3 gene or protein [5,7]. Loss of MSH3 expression or function can occur through multiple mechanisms, such as gene mutations, transcriptional or post-transcriptional repression, or cellular mislocalisation of the MSH3 protein during colon inflammation [4,5,8,9,10]. The MSH3 gene is silenced through promoter methylation in gastric cancer [11], but the relevance of promoter methylation for the generation of EMAST has not been reported yet in colorectal cancer.
The MSH3/MSH2 (MutSβ) heterodimer binds to DNA and initiates the repair of insertion–deletion loop mismatches of 2–4 nucleotides, while MSH6/MSH2 (MutSα) is more specific for mismatches of single base-pairs and insertion–deletion loop mismatches with 1–2 nucleotides [4]. The MLH1/PMS2 (MutLα) complex has a coordinator role in MMR. EMAST is caused by MutSβ dysfunction due to a defective MSH3 or MSH2. However, MSH2 deficiency disables both MutSβ and MutSα and can cause concurrent EMAST and MSI-H. Similarly, MutLα dysfunction due to a MLH1 or PMS2 defect can cause both EMAST and MSI-H. Therefore, there is an obligatory overlap of tetranucleotide repeat instability and MSI-H colorectal cancers, but MSH3-related EMAST is linked with the MSI-L phenotype [4,6,12,13].
The CpG island methylator phenotype (CIMP) is an important predictive factor in colorectal cancers involving concordant methylation silencing of multiple tumour suppressor genes [14]. Patients with CIMP-H cancers do not receive a benefit from 5-FU-based chemotherapy [15,16]. CIMP-high (H) is defined as hypermethylation of at least 3/5 markers while CIMP-low (L) is defined as hypermethylation of 1–2 out of 5 markers. CIMP-H is associated with MLH1 promoter methylation and the resulting MSI-H [17]. However, the extent of the association of CIMP with EMAST/non-MSI-H colorectal cancers has not been determined. Using the Weisenberger et al. panel of five markers [17], CIMP-H is found in ~11% and CIMP-L in ~20% of colorectal carcinomas. In addition to the five CIMP markers, genes that show concordant methylation in colorectal carcinoma include CDKN2A (p16) and MCC [18]. CDKN2A (p16) is an important cell cycle regulator that is silenced in ~30% of colorectal cancers [19]. The MCC gene is emerging as a regulator of the DNA damage response and epithelial cell–cell adhesion and is silenced by methylation in 44% and 24% of colon and rectal carcinomas, respectively [20,21,22,23]. We have shown that loss of Mcc down-regulates DNA repair genes, including Msh3, in a mouse model of inflammation-associated colon cancer [21].
Therefore, the aim of this study was to determine the association of EMAST with (i) clinicopathological features in colorectal carcinoma, (ii) MSI, (iii) CIMP and (iv) methylation of selected genes MSH3, MCC and CDKN2A (p16). We also wanted to investigate if these associations could help classify EMAST subtypes according to the number of markers present in each tumour.

2. Materials and Methods

2.1. Patient Cohorts

DNA was extracted from fresh frozen primary colorectal tumours and matched normal colon specimens (cohort 1) obtained from cancer resections (2000–2003). Thirty four percent of the patients received subsequent chemotherapy. This cohort has been previously analysed for clinicopathological and molecular variables from formalin-fixed paraffin embedded (FFPE) or fresh frozen tissue [20,24]. Clinical, pathological and follow-up data for all patients were obtained from the South Western Sydney Clinical Cancer Registry.
Further FFPE tumour DNA specimens and associated biomarker data were obtained from The Jeremy Jass Memorial Pathology Bank and the Australasian Colorectal Cancer Family Registry (ACCFR) housed at University of Melbourne (cohort 2) [25,26]. These included 22 Lynch syndrome associated carcinomas, with verified MMR gene deficiency (15 MLH1, 6 MSH2, 1 PMS2) and MSI-H status, and their matching control DNA extracted from peripheral blood [27].
The UNSW Biorepository at Mark Wainwright Analytical Centre, UNSW Sydney provided DNA specimens extracted from 13 colitis-associated colorectal carcinoma (fresh frozen) (cohort 3) [28]. Blood samples were not available and, therefore, EMAST-positivity could only be determined relative to the matching non-carcinoma inflamed tissue. The Australian Pancreatic Cancer Genome Initiative donated DNA extracted from 33 pancreatic cancers (fresh frozen) and their non-neoplastic tissue or blood control samples (cohort 4) [29,30]. Two of the tumours were selected for the study because of their known MSI-H status while 31 tumours were assigned randomly. No clinical follow-up data were available for cohorts 2–4.

2.2. Extraction of DNA and RNA from Tissue

DNA and RNA were simultaneously extracted from frozen colorectal tissue specimens using the Allprep DNA/RNA Mini Kit (Qiagen, Hilden, Germany), following homogenisation (Polytron, Kinematica, Littau, Switzerland).

2.3. Analysis of EMAST and MSI

There is no standardised reference panel available yet to define EMAST, but several suitable tetranucleotide markers have been identified. Tetranucleotide repeat markers D9S242, D20S82, L17686, UT5320, MYCL1 were selected from a previous publication [6]. PCR amplification of the markers was combined into multiplex reactions, using 1U of MyTaq polymerase and 1× MyTaq buffer (Bioline, London, UK) in each reaction. The first multiplex contained primer mixes for D9S242 and L17686 at 0.4 µM, and the second combined primers for D20S82, UT5320 and MYCL1 at 0.24 µM. Thermocycling was carried out on a Mastercycler Nexus (Eppendorf, Hamburg, Germany), with an initial denaturation of 1 min at 95 °C, followed by 35 cycles of 30 s at 95 °C, 15 s at 54–57 °C and 90 s at 72 °C, and a final extension of 10 min at 72 °C.
MSI analysis was performed using the reference panel BAT25, BAT26, D2S123, D5S346, D17S250 [3]. Cancers were classified as MSI-H if ≥2/5 of the marker genes were unstable and MSI-L when one marker was unstable. The PCRs were multiplexed, the first combining 0.2 µM BAT25 with 0.8 µM BAT26, and the second comprising 0.3 µM D2S123, 0.3 µM D5S346 and 0.5 µM D17S250. Amplification was carried out by 10 min denaturation at 95 °C, followed by 40 cycles of 45 s at 95 °C, 45 s at 55 °C and 60 s at 68 °C, and a final extension of 10 min at 68 °C.
Capillary electrophoresis of amplified products was carried out on the ABI3130XL (Thermo Fisher Scientific, Waltham, MA, USA), and data analysis performed using GeneMapper software (Thermo Fisher Scientific, Waltham, MA, USA).

2.4. Analysis of Gene Methylation and Gene Expression

Bisulphite conversion of tumour and matching control DNA was carried out using the Epitect Bisulfite Kit (Qiagen, Hilden, Germany). MSH3 methylation status was determined by methylation-specific PCR on bisulphite-treated DNA. PCR reactions specific for methylated and unmethylated MSH3 [11] were performed, with universally methylated and unmethylated DNA (Merck Millipore, Burlington, MA, USA) used as positive and negative controls. Amplified PCR products were run on agarose gels and visualised with the E-Box imager (Vilber Lourmat, Collégien, France). MCC and CDKN2A (p16) methylation and CIMP were analysed as previously described [18,19,31]. Cancers were classified as either CIMP-H if ≥3/5 of the marker genes showed methylation, CIMP-L when 1–2/5 markers were positive or CIMP-negative when 0/5 markers were positive.
cDNA was prepared from total RNA (or mRNA) using the Quantitect Reverse Transcription Kit (Qiagen, Hilden, Germany), following the manufacturer’s protocol. MSH3 expression was analysed by quantitative RT-PCR using Taqman assays (Thermo Fisher Scientific, Waltham, MA, USA). MSH3 (hs00989003_m1) was normalised to GAPDH (hs99999905_m1). Assays were carried out in triplicate on the ABI7900HT (Thermo Fisher Scientific) and the ΔΔCT method was used for data analysis.

2.5. Statistical Analysis

Survival curves were created using the Kaplan–Meier method and compared with the log-rank (Mantel–Cox) test (GraphPad Prism, San Diego, CA, USA). Overall survival was calculated from the date of diagnosis to the date of death due to any cause. Patients who were alive or lost to follow-up were censored. Fisher’s exact test and chi square was used to evaluate the significance of differences in contingency tables. The level for statistical significance was set at ≤0.05.

2.6. Analysis of The Cancer Genome Atlas (TCGA) Dataset

The TCGA PanCancer Atlas dataset containing comprehensive integrated molecular analyses was accessed through the cBioPortal for Cancer Genomics platform (https://www.cbioportal.org; accessed on 30 June 2021) [32,33,34]. The cohort of 594 Colorectal Adenocarcinoma patients was selected for data display [34]. The following genomic profiles were selected for the MSH3 gene: (i) mutations, (ii) putative copy-number alterations and (iii) mRNA expression z-scores relative to normal tissue samples. This returned data for 524 samples. The same analysis was performed for the TCGA PanCancer Atlas Pancreatic Adenocarcinoma cohort [34], except that mRNA expression z-scores were calculated relative to diploid samples (corresponding normal tissue data were not available). The graphs and statistical data were obtained using the function “Plots” and “Color samples by MSI MANTIS score” [35]. The Illumina Methylation450K data for colorectal and pancreatic cancers were obtained using the SMART App (http://www.bioinfo-zs.com/smartapp; accessed on 23 December 2020) [36].

3. Results

3.1. Patterns of EMAST Marker Positivity Are Different between Colorectal and Pancreatic Carcinomas

A panel of five tetranucleotide markers was selected from a previous publication to detect EMAST [6]. We first determined the prevalence of instability for each of these five EMAST markers across the four cohorts of carcinomas where MSI-H is found at a high frequency (100%, Lynch syndrome, cohort 2), intermediate (13–15%, sporadic colorectal and colitis-associated carcinomas, cohorts 1 and 3) or low frequency (sporadic pancreatic carcinomas, cohort 4). Instability for at least one of these EMAST markers was found in 100% of the 22 Lynch syndrome-associated colorectal carcinomas, 55% (55/100) of sporadic colorectal, 62% (8/13) of colitis-associated, and 74% (23/31) of sporadic non-MSI-H pancreatic carcinoma. Positivity for each of the five markers was evenly distributed in the 55 EMAST-positive sporadic colorectal carcinomas (19–24%). In contrast, the L17686 tetranucleotide marker was unstable in the majority of the colitis-associated (5/8) and sporadic pancreatic (21/23) carcinomas with any EMAST-positive markers. Representative images of the EMAST markers are shown in Figure 1.

3.2. EMAST≥3/5 Is Associated with MSI-H Across Different Cancer Types

Concurrent instability of two or more EMAST markers was found in 95% of Lynch syndrome (21/22), 29–31% of sporadic colorectal (29/100) or colitis-associated (4/13) carcinomas and 19% of the pancreatic carcinomas (6/31). All but two of the MSI-H sporadic colorectal carcinomas (cohort 1) had 3–5 unstable EMAST markers, which was rare in non-MSI-H tumours (Figure 2a, Table 1). Conversely, non-MSI-H colorectal carcinomas showed 1–2 EMAST-positive markers (47%) or were EMAST-negative (51%) (Figure 2a, Table 1). Most of the Lynch syndrome colorectal carcinomas (18/22), 2/2 MSI-H colitis-associated and 2/2 MSI-H pancreatic cancers showed instability with 3–5 EMAST markers.
We then examined the EMAST subsets separately in the sporadic colorectal cancer cohort, EMAST 3/5 (three or more EMAST-positive markers), EMAST-2/5 (two markers) and EMAST-1/5 (a single marker).

3.3. Presence of a Single EMAST Marker Is Associated with MSI-L and Lymph Node Metastases But Not Location in Sporadic Colorectal Carcinoma

We determined the relationship between the number of EMAST markers, MSI-L, tumour stage and location in the sporadic colorectal tumours (Table 1, Figure 2b–d). MSI-L was enriched in EMAST-1/5 tumours (9/26, 35%; Table 1, Table 2, Figure 2a,b). Dukes stage 3 and 4 cancers were also increased in EMAST-1/5 tumours (18/26, 69%), which was due to an association with increased lymph node metastasis (Table 1, Figure 2c,d). In general, these carcinomas were not poorly differentiated and there was no significant difference in location of EMAST-1/5 carcinomas along the length of the large bowel. Similar associations were seen for MSI-L sporadic colorectal carcinomas (Table 2). This suggests that carcinomas with MSI-L and/or a single EMAST positive marker are more likely to be of a higher stage with evidence of lymph node metastases at the time of detection. In survival analysis, there was no difference in overall survival between the EMAST subsets if the EMAST-2/5 group was combined with either EMAST-1/5 (Figure 2f) or EMAST-3/5 (Figure 2g). However, patients with EMAST-1/5 tumours showed significantly shorter overall survival when compared with all other patients as a single group (Figure 2h).
There was no significant association of the number of EMAST positive markers with MSI-L in the colitis-associated carcinomas, where MSI-L was detected at 38% frequency. Only three pancreatic specimens showed MSI-L.

3.4. EMAST-1/5 Colorectal Cancers Show Only Scattered Methylation

We next determined the relationship of EMAST with CIMP and methylation of CDKN2A (25%) and MCC (29%). EMAST 3/5 was strongly associated with CIMP-H and concurrent MCC/CDKN2A promoter methylation in sporadic colorectal cancer. In contrast, EMAST-1/5 carcinomas showed the lowest frequency of these markers (Table 1, Figure 2a,e). MSI-H was also strongly associated with CIMP-H in sporadic colorectal cancers, as expected, and with concurrent CDKN2A/MCC methylation. CIMP-H, MCC and CDKN2A methylation was absent or rare in MSI-L carcinomas (Table 2, Figure 2a). These data are consistent with our previous findings on CDKN2A [31] and suggest that carcinomas with MSI-L and/or a single EMAST positive marker are less likely to display high-level gene promoter methylation.
We reported previously that MCC and CDKN2A promoter methylation are frequently found in the same colon carcinoma specimens [18]. Here, the same strong association was observed in the colon but not in rectal carcinomas. MCC and CDKN2A methylation co-occurred in 57% (16/28) of the carcinomas positive for either marker in the colon but only 25% (2/8) in the rectum/rectosigmoid region.
The relationship of EMAST with the methylation markers was not studied in pancreatic or colitis-associated carcinomas.

3.5. Presence of 1–2 EMAST Markers Is Not Associated with Down-Regulation of MSH3 mRNA Expression

Defective MSH3 function or loss of gene expression causes EMAST and can occur through multiple mechanisms, but the role of MSH3 gene silencing has not yet been investigated in colorectal cancer. We analysed MSH3 promoter hypermethylation using methylation-specific primers [11] and RNA expression by qPCR. No promoter hypermethylation was evident in our specimens (data not shown). About 41% (39/96) of the tumours displayed at least a 2-fold reduction in MSH3 transcript levels compared to matching normal tissue; of these, 13% (12/96) showed >4-fold reduction. Down-regulation of MSH3 expression was associated with the presence of 3–5 EMAST markers but not with 1–2 positive markers (Table 3). Down-regulation of MSH3 was also more frequent in MSI-H or CIMP-H cancers but the strongest association was observed with MCC methylation (p = 0.0001, Table 3).

3.6. MSH3 Gene Mutations Are Rare But Copy Number Alterations Are More Common in Colorectal and Pancreatic Carcinoma

To evaluate MSH3 gene alteration and expression in a larger cohort, we analysed publicly available data from ~3000 colorectal cancer patients using cBioPortal for Cancer Genomics [32,33]. MSH3 mutations have been detected in 1–6% in various cohorts; about one quarter of these are truncating mutations, the rest are missense mutations. Two (2/524) truncating mutations are reported for colorectal carcinoma in the TCGA PanCancer Atlas cohort [34]. MSH3 copy number alterations occur in another ~32% of colorectal cancers; one copy is deleted in 24% (128/524), both copies in 0.2% (1/524) and copy gain is found in 8% (41/524) (Figure 3a) [34]. MSH3 copy number alterations show very little overlap with MSI-H that is mostly confined to MSH3 diploid cancers [35] (Figure 3b), but tetranucleotide repeat alterations have not yet been captured in the publicly available data.
MSH3 mRNA expression levels vary greatly in the TCGA colorectal cohort but can be considered low in 17% (87/524) of the tumours, when compared with the mean expression in normal tissue specimens (Figure 3a). There is a weak correlation between copy number alterations and mRNA expression (Pearson r = 0.35, p < 0.0001). The lowest level of MSH3 mRNA expression is seen in a subset of diploid tumours, including some MSI-H cancers (Figure 3b). Similar frequencies of MSH3 mutations and copy number alterations are found for pancreatic adenocarcinomas, but no MSI-H is evident (Figure 3c,d). The MSH3 gene promoter is not hypermethylated in colorectal or pancreatic carcinomas (Illumina Methylation450K CpG island probes cg11646734, cg16401290, cg14865507, cg07526021 and cg18200270) [34,36].
The TCGA data [34] indicate that MSH3 mutations, deep deletion or low mRNA expression are not frequent enough to explain the generation of all the observed EMAST in non-MSI-H colorectal or pancreatic carcinomas. Furthermore, our data suggest that down-regulation of MSH3 mRNA does not correlate with the presence of 1–2 EMAST-positive markers in colorectal cancer.

4. Discussion

This study has shown that the high methylation phenotype CIMP-H and concurrent MCC/CDKN2A methylation are rare in colorectal carcinomas that are positive for only one EMAST marker or MSI-L but more common in cancers with ≥2 markers. We previously reported in another cohort that CDKN2A (p16) methylation was significantly less frequent in MSI-L colon tumours compared to MSI-H or MSS tumours and that MSI-L tumours were associated with poor survival [31]. Since then, evidence has been strengthening that both MSI-L or EMAST/non-MSI-H are markers of poor prognosis in colorectal carcinoma and share a mechanistic basis [7,12,37,38]. Here, we show that the poor prognosis is likely due to increased lymph node metastasis, which is associated with the presence of one positive EMAST or MSI marker. Our data suggest poorer overall survival for the EMAST-1/5 group, which incorporates most of the MSI-L tumours in the cohort. This supports and extends the findings of Garcia et al., who found that EMAST/non-MSI-H cancers grouped with MSI-L were associated with shorter recurrence-free survival than cancers that were MSI-H or microsatellite stable [12].
Here, we also show that the MSH3 gene promoter is not methylated in colorectal cancer. Our data and the TCGA data indicate that tumour levels of MSH3 mRNA expression vary greatly. Down-regulation of MSH3 mRNA expression is associated with EMAST ≥ 3/5 and MSI-H but not with the EMAST/MSI-L phenotype. This is consistent with the finding that EMAST can be caused by MSH3 protein dysfunction, which occurs as a result of displacement of MSH3 from the nucleus to the cytoplasm. This can be induced by exposing colon cancer cells to IL6 or hydroxen peroxide in vitro and is thought to occur as a result of inflammation in sporadic and inflammatory bowel disease-associated cancers [5,39].
It remains to be determined whether MSH3 copy number alterations, found in 32% of the TCGA colorectal and 27% of the pancreatic cancer cohort (Figure 3a,c), are associated with the EMAST/non-MSI-H phenotype. MSI-H is rare in colorectal cancers with MSH3 copy number alterations but common in diploid cancers (Figure 3b). MSH3 copy number alterations reflect chromosomal instability (CIN), which is known to show inverse correlation with CIMP-H [40]. Here, we found that CIMP-H was rare in cancers with only one positive EMAST marker. Aberrant MSH3 protein expression and loss of heterozygosity of MSH3 markers have been reported in MSI-L cancers [38]. This may indicate a wider association of CIN with EMAST/MSI-L.
Genes that are silenced by methylation in colorectal carcinoma and associated with CIMP-H include many tumour suppressor genes [17]. Our data show that the EMAST-2/5 and high-level gene methylation show a small overlap in non-MSI-H cancers. The TCGA data show only low correlation between MSH3 copy number alterations and mRNA expression, indicating that multiple factors regulate mRNA expression. MSH3 mRNA down-regulation was strongly associated with MCC methylation in our cohort, which was not observed for CDKN2A methylation. This is consistent with our data from a mouse model of inflammation-associated colorectal cancer where deletion of Mcc caused down-regulation of multiple DNA repair genes in the inflamed tissue [21]. MCC methylation is also associated with more invasive colorectal cancers [20]. It is possible that methylation or alterations of non-MMR genes modify the EMAST phenotype spectrum, but this needs further investigation.
In this study, we used five tetranucleotide repeat markers, D9S242, D20S82, L17686, UT5320 and MYCL1, that were selected from a previous publication [6]. Two of these markers, L17686 and UT5320, have not been commonly used in other recent studies on EMAST. Here, we detected a higher frequency of EMAST in pancreatic cancer (74%) than previously reported (45%) [41], which was due to the L17686 marker. The L17686 marker was also common in the colitis-associated cancer specimens. Inflammation plays a critical role in the tumorigenesis of both pancreatic and colitis-associated cancer, and IL-6 is an important inflammatory cytokine that promotes pathogenesis in both conditions [10,42,43]. This may suggest that L17686 could be a marker that is particularly sensitive to inflammation-induced EMAST/MSI-L. The frequency of EMAST in colitis-associated cancers was 68%, which is comparable to a previously reported frequency of 71% [10]. However, since our control specimens were from matched inflamed tissue, which could be EMAST-positive, the frequency of EMAST may have been higher. Positivity of a single marker was common in pancreatic cancers (48%, 15/31) using the EMAST marker panel, but less frequent (10%, 3/31) using the MSI marker panel. This highlights the possible differences between cancer types and the need to standardise EMAST marker panels and the nomenclature, possibly incorporating MSI and EMAST markers, as was suggested by Garcia et al. [12] and Raeker et al. [13].
Our study highlights the need to include colorectal or pancreatic cancers that are positive for only one (1/5) tetranucleotide repeat marker as an important subgroup of the EMAST spectrum. In some recent studies, EMAST was defined as the presence of ≥2/5 tetranucleotide markers [44,45,46]. This definition results in a significant overlap with MSI-H and potentially excludes tumours that are associated with increased lymph node metastasis and lack of CIMP-H. The frequency of the EMAST-1/5 group could be inferred in at least two of the studies, as 23% [45] and 28% [46] of MSS tumours, which is comparable to our study.
MSH3-associated EMAST/MSI-L is a potentially valuable predictive biomarker for colorectal and other cancers, e.g., pancreatic and non-small cell lung carcinomas [41,47]. Apart from MMR, MSH3 has another function in the homologous recombination (HR) repair of DNA double-strand breaks that occur during carcinogenesis [48]. As a consequence, carcinomas with loss of MSH3 function may have a defect in HR and might respond better to targeted therapies with PARP and/or DNA-PKc inhibitors. These therapies have a synergistic effect based on the synthetic lethality principle [48]. Therefore, it is important to develop biomarkers that can identify the HR defective subset of carcinomas that may be responsive to these or other cancer-targeted drugs.

5. Conclusions

This study has identified new features of tetranucleotide instability that are clinically relevant in sporadic colorectal carcinomas. In particular, cancers with only one positive EMAST marker or MSI-L are associated with lack of CIMP-H and an increased likelihood of lymph node metastasis. EMAST-1/5 and MSI-L may be valuable surrogate markers for MSH3 dysfunction. Due to the dual function of MSH3 in both MMR and HR, EMAST/MSI-L cancers could be potentially responsive to therapies that exploit the concept of synthetic lethality.

Author Contributions

Conceptualization, M.R.J.K.-C.; Formal analysis, S.M., N.C., Z.J., H.W.P., J.E.D. and M.R.J.K.-C.; Funding acquisition, J.E.D. and M.R.J.K.-C.; Investigation, S.M. and N.C.; Resources, M.A.J., D.D.B., J.L.H., E.S., J.E.D. and M.R.J.K.-C.; Supervision, M.R.J.K.-C.; Writing—original draft, S.M., N.C. and M.R.J.K.-C.; Writing—review and editing, Z.J., H.W.P., M.A.J., D.D.B., J.L.H., E.S. and J.E.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Health and Medical Research Council of Australia (NHMRC) grant number 1020406, Cancer Council NSW grants RG17-05 and RG19-01, and the Gastroenterological Society of Australia GESA Project Grant. The Australasian Colorectal Cancer Family Registry (ACCFR) is supported in part by funding from the National Cancer Institute (NCI) of the U.S. National Institutes of Health (NIH), grant number U01 CA167551.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Research Governance Officer of Garvan Institute of Medical Research (24.12.2015) and the Human Research Ethics Committees of Sydney Local Health District (protocol code: X08-0224 and HREC/08/RPAH/376) and Melbourne University (ACCFR HREC #1954921).

Informed Consent Statement

Informed consent was obtained from all prospectively recruited participants. For the retrospectively obtained specimens, patient consent was waived due to multiple approved reasons, such as the high proportion of individuals who are likely to have died since the specimens were collected.

Data Availability Statement

Publicly available TCGA PanCancer Atlas datasets were analysed in this study. The colorectal data can be found at https://www.cbioportal.org/results/plots?cancer_study_list=coadread_tcga_pan_can_atlas (accessed on 30 June 2021). Other datasets generated in this study are available from the corresponding author on reasonable request.

Acknowledgments

We thank Andrew Biankin, Robyn Ward, Nicholas Hawkins, South Western Sydney Colorectal Tumour Group, South Western Sydney Clinical Cancer Registry, Australian Pancreatic Cancer Genome Initiative, the Australasian Colorectal Cancer Family Registry at the University of Melbourne and UNSW Biorepository at Mark Wainwright Analytical Centre UNSW Sydney for providing patient tissue, DNA specimens and data. We thank Owen Dent for advice with statistics. The ACCFR wishes to thank the generous contributions of their study participants, dedication of study staff, and the financial support from the U.S. National Cancer Institute, without which their registry would not exist.

Conflicts of Interest

The authors declare no conflict of interest. The contents of the published material are solely the responsibility of the administering institution, a participating institution or individual authors and do not reflect the views of the NHMRC or the NIH. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  1. Fishel, R. The selection for mismatch repair defects in hereditary nonpolyposis colorectal cancer: Revising the mutator hypothesis. Cancer Res. 2001, 61, 7369–7374. [Google Scholar]
  2. Boland, C.R.; Goel, A. Microsatellite instability in colorectal cancer. Gastroenterology 2010, 138, 2073–2087.e2073. [Google Scholar] [CrossRef]
  3. 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.; et al. 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, 5248–5257. [Google Scholar]
  4. Carethers, J.M.; Koi, M.; Tseng-Rogenski, S.S. Emast is a form of microsatellite instability that is initiated by inflammation and modulates colorectal cancer progression. Genes 2015, 6, 185–205. [Google Scholar] [CrossRef] [Green Version]
  5. Tseng-Rogenski, S.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, 579–589. [Google Scholar] [CrossRef] [Green Version]
  6. Yamada, K.; Kanazawa, S.; Koike, J.; Sugiyama, H.; Xu, C.; Funahashi, K.; Boland, C.R.; Koi, M.; Hemmi, H. Microsatellite instability at tetranucleotide repeats in sporadic colorectal cancer in japan. Oncol. Rep. 2010, 23, 551–561. [Google Scholar]
  7. Haugen, A.C.; Goel, A.; Yamada, K.; Marra, G.; Nguyen, T.P.; Nagasaka, T.; Kanazawa, S.; Koike, J.; Kikuchi, Y.; Zhong, X.; et al. Genetic instability caused by loss of muts homologue 3 in human colorectal cancer. Cancer Res. 2008, 68, 8465–8472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. 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, 1519–1525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. 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, 1521–1528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Munakata, K.; Koi, M.; Kitajima, T.; Tseng-Rogenski, S.; Uemura, M.; Matsuno, H.; Kawai, K.; Sekido, Y.; Mizushima, T.; Toiyama, Y.; et al. Inflammation-associated microsatellite alterations caused by msh3 dysfunction are prevalent in ulcerative colitis and increase with neoplastic advancement. Clin. Transl. Gastroenterol. 2019, 10, e00105. [Google Scholar] [CrossRef] [PubMed]
  11. Kim, H.G.; Lee, S.; Kim, D.Y.; Ryu, S.Y.; Joo, J.K.; Kim, J.C.; Lee, K.H.; Lee, J.H. Aberrant methylation of DNA mismatch repair genes in elderly patients with sporadic gastric carcinoma: A comparison with younger patients. J. Surg. Oncol. 2010, 101, 28–35. [Google Scholar] [CrossRef]
  12. Garcia, M.; Choi, C.; Kim, H.R.; Daoud, Y.; Toiyama, Y.; Takahashi, M.; Goel, A.; Boland, C.R.; Koi, M. Association between recurrent metastasis from stage ii and iii primary colorectal tumors and moderate microsatellite instability. Gastroenterology 2012, 143, 48–50.e41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Raeker, M.O.; Pierre-Charles, J.; Carethers, J.M. Tetranucleotide microsatellite mutational behavior assessed in real time: Implications for future microsatellite panels. Cell Mol. Gastroenterol. Hepatol. 2020, 9, 689–704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Toyota, M.; Ahuja, N.; Ohe-Toyota, M.; Herman, J.G.; Baylin, S.B.; Issa, J.P. Cpg island methylator phenotype in colorectal cancer. Proc. Natl. Acad. Sci. USA 1999, 96, 8681–8686. [Google Scholar] [CrossRef] [Green Version]
  15. Juo, Y.Y.; Johnston, F.M.; Zhang, D.Y.; Juo, H.H.; Wang, H.; Pappou, E.P.; Yu, T.; Easwaran, H.; Baylin, S.; van Engeland, M.; et al. Prognostic value of cpg island methylator phenotype among colorectal cancer patients: A systematic review and meta-analysis. Ann. Oncol. 2014, 25, 2314–2327. [Google Scholar] [CrossRef] [PubMed]
  16. Jover, R.; Nguyen, T.P.; Perez-Carbonell, L.; Zapater, P.; Paya, A.; Alenda, C.; Rojas, E.; Cubiella, J.; Balaguer, F.; Morillas, J.D.; et al. 5-fluorouracil adjuvant chemotherapy does not increase survival in patients with cpg island methylator phenotype colorectal cancer. Gastroenterology 2011, 140, 1174–1181. [Google Scholar] [CrossRef] [Green Version]
  17. Weisenberger, D.J.; Siegmund, K.D.; Campan, M.; Young, J.; Long, T.I.; Faasse, M.A.; Kang, G.H.; Widschwendter, M.; Weener, D.; Buchanan, D.; et al. Cpg island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with braf mutation in colorectal cancer. Nat. Genet. 2006, 38, 787–793. [Google Scholar] [CrossRef]
  18. Kohonen-Corish, M.R.; Sigglekow, N.D.; Susanto, J.; Chapuis, P.H.; Bokey, E.L.; Dent, O.F.; Chan, C.; Lin, B.P.; Seng, T.J.; Laird, P.W.; et al. Promoter methylation of the mutated in colorectal cancer gene is a frequent early event in colorectal cancer. Oncogene 2007, 26, 4435–4441. [Google Scholar] [CrossRef] [Green Version]
  19. Kohonen-Corish, M.R.; Tseung, J.; Chan, C.; Currey, N.; Dent, O.F.; Clarke, S.; Bokey, L.; Chapuis, P.H. Kras mutations and cdkn2a promoter methylation show an interactive adverse effect on survival and predict recurrence of rectal cancer. Int. J. Cancer. 2014, 134, 2820–2828. [Google Scholar] [CrossRef]
  20. Benthani, F.A.; Herrmann, D.; Tran, P.N.; Pangon, L.; Lucas, M.C.; Allam, A.H.; Currey, N.; Al-Sohaily, S.; Giry-Laterriere, M.; Warusavitarne, J.; et al. “Mcc” protein interacts with e-cadherin and beta-catenin strengthening cell-cell adhesion of hct116 colon cancer cells. Oncogene 2018, 37, 663–672. [Google Scholar] [CrossRef]
  21. Currey, N.; Jahan, Z.; Caldon, C.E.; Tran, P.N.; Benthani, F.; De Lacavalerie, P.; Roden, D.L.; Gloss, B.S.; Campos, C.; Bean, E.G.; et al. Mouse model of mutated in colorectal cancer gene deletion reveals novel pathways in inflammation and cancer. Cell Mol. Gastroenterol. Hepatol. 2019, 7, 819–839. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Pangon, L.; Sigglekow, N.D.; Larance, M.; Al-Sohaily, S.; Mladenova, D.N.; Selinger, C.I.; Musgrove, E.A.; Kohonen-Corish, M.R. The “mutated in colorectal cancer” protein is a novel target of the uv-induced DNA damage checkpoint. Genes Cancer 2010, 1, 917–926. [Google Scholar] [CrossRef] [Green Version]
  23. Pangon, L.; Van Kralingen, C.; Abas, M.; Daly, R.J.; Musgrove, E.A.; Kohonen-Corish, M.R. The pdz-binding motif of mcc is phosphorylated at position -1 and controls lamellipodia formation in colon epithelial cells. Biochim. Biophys. Acta 2012, 1823, 1058–1067. [Google Scholar] [CrossRef] [Green Version]
  24. Al-Sohaily, S.; Henderson, C.; Selinger, C.; Pangon, L.; Segelov, E.; Kohonen-Corish, M.R.; Warusavitarne, J. Loss of special at-rich sequence-binding protein 1 (satb1) predicts poor survival in patients with colorectal cancer. Histopathology 2014, 65, 155–163. [Google Scholar] [CrossRef] [PubMed]
  25. Jenkins, M.A.; Win, A.K.; Templeton, A.S.; Angelakos, M.S.; Buchanan, D.D.; Cotterchio, M.; Figueiredo, J.C.; Thibodeau, S.N.; Baron, J.A.; Potter, J.D.; et al. Cohort profile: The colon cancer family registry cohort (ccfrc). Int. J. Epidemiol. 2018, 47, 387–388i. [Google Scholar] [CrossRef] [PubMed]
  26. Newcomb, P.A.; Baron, J.; Cotterchio, M.; Gallinger, S.; Grove, J.; Haile, R.; Hall, D.; Hopper, J.L.; Jass, J.; Le Marchand, L.; et al. Colon cancer family registry: An international resource for studies of the genetic epidemiology of colon cancer. Cancer Epidemiol. Biomark. Prev. 2007, 16, 2331–2343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Buchanan, D.D.; Clendenning, M.; Rosty, C.; Eriksen, S.V.; Walsh, M.D.; Walters, R.J.; Thibodeau, S.N.; Stewart, J.; Preston, S.; Win, A.K.; et al. Tumor testing to identify lynch syndrome in two australian colorectal cancer cohorts. J. Gastroenterol. Hepatol. 2017, 32, 427–438. [Google Scholar] [CrossRef] [Green Version]
  28. Sigglekow, N.D.; Pangon, L.; Brummer, T.; Molloy, M.; Hawkins, N.J.; Ward, R.L.; Musgrove, E.A.; Kohonen-Corish, M.R. Mutated in colorectal cancer protein modulates the nfkappab pathway. Anticancer Res. 2012, 32, 73–79. [Google Scholar]
  29. Humphris, J.L.; Patch, A.M.; Nones, K.; Bailey, P.J.; Johns, A.L.; McKay, S.; Chang, D.K.; Miller, D.K.; Pajic, M.; Kassahn, K.S.; et al. Hypermutation in pancreatic cancer. Gastroenterology 2017, 152, 68–74.e62. [Google Scholar] [CrossRef] [Green Version]
  30. Biankin, A.V.; Waddell, N.; Kassahn, K.S.; Gingras, M.C.; Muthuswamy, L.B.; Johns, A.L.; Miller, D.K.; Wilson, P.J.; Patch, A.M.; Wu, J.; et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 2012, 491, 399–405. [Google Scholar] [CrossRef] [PubMed]
  31. Kohonen-Corish, M.R.; Daniel, J.J.; Chan, C.; Lin, B.P.; Kwun, S.Y.; Dent, O.F.; Dhillon, V.S.; Trent, R.J.; Chapuis, P.H.; Bokey, E.L. Low microsatellite instability is associated with poor prognosis in stage c colon cancer. J. Clin. Oncol. 2005, 23, 2318–2324. [Google Scholar] [CrossRef] [PubMed]
  32. Cerami, E.; Gao, J.; Dogrusoz, U.; Gross, B.E.; Sumer, S.O.; Aksoy, B.A.; Jacobsen, A.; Byrne, C.J.; Heuer, M.L.; Larsson, E.; et al. The cbio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012, 2, 401–404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Gao, J.; Aksoy, B.A.; Dogrusoz, U.; Dresdner, G.; Gross, B.; Sumer, S.O.; Sun, Y.; Jacobsen, A.; Sinha, R.; Larsson, E.; et al. Integrative analysis of complex cancer genomics and clinical profiles using the cbioportal. Sci. Signal. 2013, 6, pl1. [Google Scholar] [CrossRef] [Green Version]
  34. Hoadley, K.A.; Yau, C.; Hinoue, T.; Wolf, D.M.; Lazar, A.J.; Drill, E.; Shen, R.; Taylor, A.M.; Cherniack, A.D.; Thorsson, V.; et al. Cell-of-origin patterns dominate the molecular classification of 10,000 tumors from 33 types of cancer. Cell 2018, 173, 291–304.e296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Bonneville, R.; Krook, M.A.; Kautto, E.A.; Miya, J.; Wing, M.R.; Chen, H.Z.; Reeser, J.W.; Yu, L.; Roychowdhury, S. Landscape of microsatellite instability across 39 cancer types. JCO Precis. Oncol. 2017, 2017. [Google Scholar] [CrossRef]
  36. Li, Y.; Ge, D.; Lu, C. The smart app: An interactive web application for comprehensive DNA methylation analysis and visualization. Epigenetics. Chromatin 2019, 12, 71. [Google Scholar] [CrossRef] [Green Version]
  37. Torshizi Esfahani, A.; Seyedna, S.Y.; Nazemalhosseini Mojarad, E.; Majd, A.; Asadzadeh Aghdaei, H. Msi-l/emast is a predictive biomarker for metastasis in colorectal cancer patients. J. Cell Physiol. 2019, 234, 13128–13136. [Google Scholar] [CrossRef]
  38. Plaschke, J.; Preussler, M.; Ziegler, A.; Schackert, H.K. Aberrant protein expression and frequent allelic loss of msh3 in colorectal cancer with low-level microsatellite instability. Int. J. Colorectal Dis. 2012, 27, 911–919. [Google Scholar] [CrossRef]
  39. 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, e50616. [Google Scholar] [CrossRef] [Green Version]
  40. Goel, A.; Nagasaka, T.; Arnold, C.N.; Inoue, T.; Hamilton, C.; Niedzwiecki, D.; Compton, C.; Mayer, R.J.; Goldberg, R.; Bertagnolli, M.M.; et al. The cpg island methylator phenotype and chromosomal instability are inversely correlated in sporadic colorectal cancer. Gastroenterology 2007, 132, 127–138. [Google Scholar] [CrossRef]
  41. 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, e0208557. [Google Scholar] [CrossRef] [PubMed]
  42. Van Duijneveldt, G.; Griffin, M.D.W.; Putoczki, T.L. Emerging roles for the il-6 family of cytokines in pancreatic cancer. Clin. Sci. 2020, 134, 2091–2115. [Google Scholar] [CrossRef] [PubMed]
  43. 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 J. Gastrointest. Oncol. 2018, 10, 1–14. [Google Scholar] [CrossRef]
  44. Pirini, F.; Pasini, L.; Tedaldi, G.; Scarpi, E.; Marisi, G.; Molinari, C.; Calistri, D.; Passardi, A.; Ulivi, P. Instability of non-standard microsatellites in relation to prognosis in metastatic colorectal cancer patients. Int. J. Mol. Sci. 2020, 21, 3532. [Google Scholar] [CrossRef]
  45. Kondelin, J.; Martin, S.; Katainen, R.; Renkonen-Sinisalo, L.; Lepisto, A.; Koskensalo, S.; Bohm, J.; Mecklin, J.P.; Cajuso, T.; Hanninen, U.A.; et al. No evidence of emast in whole genome sequencing data from 248 colorectal cancers. Genes Chromosomes Cancer 2021, 60, 463–473. [Google Scholar] [CrossRef] [PubMed]
  46. 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, e0124538. [Google Scholar] [CrossRef]
  47. Ahrendt, S.A.; Decker, P.A.; Doffek, K.; Wang, B.; Xu, L.; Demeure, M.J.; Jen, J.; Sidransky, D. Microsatellite instability at selected tetranucleotide repeats is associated with p53 mutations in non-small cell lung cancer. Cancer Res. 2000, 60, 2488–2491. [Google Scholar]
  48. Dietlein, F.; Thelen, L.; Reinhardt, H.C. Cancer-specific defects in DNA repair pathways as targets for personalized therapeutic approaches. Trends Genet. 2014, 30, 326–339. [Google Scholar] [CrossRef]
Figure 1. Representative images of EMAST detection using the tetranucleotide repeat markers D9S242, D20S82, MYCL1, UT5320, and L17686 in colorectal cancer. New peaks or change of peak heights in cancer specimens indicate alteration of the repeat length compared to matching normal tissue.
Figure 1. Representative images of EMAST detection using the tetranucleotide repeat markers D9S242, D20S82, MYCL1, UT5320, and L17686 in colorectal cancer. New peaks or change of peak heights in cancer specimens indicate alteration of the repeat length compared to matching normal tissue.
Cancers 13 03529 g001
Figure 2. (a) Presence of CIMP-H, CDKN2A and MCC gene methylation (black), CIMP-L (grey), MSI-H (red) and MSI-L (blue) in 100 colorectal cancers grouped according to EMAST-≥3/5, 2/5, 1/5 or 0/5 status; distribution of MSI status (b), Dukes stages (c) N stages (d) and (e) CIMP status in colorectal cancers grouped according to EMAST-≥3/5, 2/5, 1/5 or 0/5 status (proportions taken from Table 1). Overall survival in patients grouped according to EMAST-positive (1–3 markers out of 5) or EMAST-negative tumours (f,g). Overall survival in patients with a single EMAST marker (EMAST-1/5) compared with all other patients (h).
Figure 2. (a) Presence of CIMP-H, CDKN2A and MCC gene methylation (black), CIMP-L (grey), MSI-H (red) and MSI-L (blue) in 100 colorectal cancers grouped according to EMAST-≥3/5, 2/5, 1/5 or 0/5 status; distribution of MSI status (b), Dukes stages (c) N stages (d) and (e) CIMP status in colorectal cancers grouped according to EMAST-≥3/5, 2/5, 1/5 or 0/5 status (proportions taken from Table 1). Overall survival in patients grouped according to EMAST-positive (1–3 markers out of 5) or EMAST-negative tumours (f,g). Overall survival in patients with a single EMAST marker (EMAST-1/5) compared with all other patients (h).
Cancers 13 03529 g002
Figure 3. Genomic data from 524 colorectal and 168 pancreatic adenocarcinomas that were profiled for MSH3 gene mutations, copy number alterations, mRNA expression and MSI MANTIS score in the TCGA PanCancer Atlas [32,33,34,35]. (a) Colorectal cancers have MSH3 truncating mutation in 0.4% (2/524), one copy deletion in 24% (128/524), deep deletion in 0.2% (1/524), copy gain in 8% (41/524) and mRNA expression z-score (relative to normal samples) less than 2 in 17% (87/524) of the patients. The z-score for each tumour specimen indicates the number of standard deviations away from the mean expression in the reference population. VUS = variants of uncertain significance; shallow deletion = shallow loss, possibly a heterozygous deletion; deep deletion = deep loss, possibly a homozygous deletion; gain = low-level gain, a few additional copies. GISTIC = Genomic Identification of Significant Targets in Cancer. (b) MSI-H (MSI MANTIS score > 0.6 [35]) is found in 12% (64/522) and is mostly confined to MSH3 diploid colorectal cancers. (c) Pancreatic carcinomas have MSH3 truncating mutation in 0.6% (1/168), one copy deletion in 19% (32/168) and copy gain in 7% (12/168) of the patients. (d) No MSI-H is found in pancreatic carcinoma, with MSI MANTIS score < 0.4 in all patients.
Figure 3. Genomic data from 524 colorectal and 168 pancreatic adenocarcinomas that were profiled for MSH3 gene mutations, copy number alterations, mRNA expression and MSI MANTIS score in the TCGA PanCancer Atlas [32,33,34,35]. (a) Colorectal cancers have MSH3 truncating mutation in 0.4% (2/524), one copy deletion in 24% (128/524), deep deletion in 0.2% (1/524), copy gain in 8% (41/524) and mRNA expression z-score (relative to normal samples) less than 2 in 17% (87/524) of the patients. The z-score for each tumour specimen indicates the number of standard deviations away from the mean expression in the reference population. VUS = variants of uncertain significance; shallow deletion = shallow loss, possibly a heterozygous deletion; deep deletion = deep loss, possibly a homozygous deletion; gain = low-level gain, a few additional copies. GISTIC = Genomic Identification of Significant Targets in Cancer. (b) MSI-H (MSI MANTIS score > 0.6 [35]) is found in 12% (64/522) and is mostly confined to MSH3 diploid colorectal cancers. (c) Pancreatic carcinomas have MSH3 truncating mutation in 0.6% (1/168), one copy deletion in 19% (32/168) and copy gain in 7% (12/168) of the patients. (d) No MSI-H is found in pancreatic carcinoma, with MSI MANTIS score < 0.4 in all patients.
Cancers 13 03529 g003
Table 1. Association of EMAST with clinicopathological features and molecular markers in colorectal cancer from cohort 1.
Table 1. Association of EMAST with clinicopathological features and molecular markers in colorectal cancer from cohort 1.
Tumour
Feature
VariableEMAST-
≥3/5
n = 13
(%)
EMAST-
2/5
n = 16
(%)
EMAST-
1/5
n = 26
(%)
No
EMAST
n = 45
(%)
p **
Microsatellite
instability
MSS2 (3)13 (17)17 (23)42 (57)<0.0001
MSI-L0 (0)2 (15)9 (70)2 (15)
MSI-H11 (84)1 (8)0 (0)1 (8)
CIMPAbsent3 (5)8 (14)21 (37)25 (44)<0.0001
CIMP-L2 (7)6 (20)4 (13)18 (60)
CIMP-H8 (62)2 (15)1 (8)2 (15)
CDKN2A (p16) methylationAbsent5 (7)11 (15)22 (29)37 (49)0.007
Present8 (32)5 (20)4 (16)8 (32)
MCC
methylation
Absent4 (6)9 (13)22 (31)35 (50)0.002
Present9 (31)6 (21)4 (14)10 (34)
CDKN2A & MCC methylationAbsent5(5)11 (13)25 (31)41 (51)<0.00001
Present8 (44)5* (28)1 * (6)4 (22)
Dukes stage1,210 (17)8 (14)8 (14)32 (55)0.028
32 (7)6 (21)11 (39)9 (32)
41 (7)2 (14)7 (50)4 (29)
T stage1,22 (7)5 (19)6 (22)14 (52)0.654
3,411 (15)11 (15)20 (27)31 (43)
N stage010 (16)8 (13)10 (16)33 (54)0.036
11 (5)4 (19)7 (33)9 (43)
22 (11)4 (22)9 (50)3 (17)
M stage012 (14)14 (16)20 (23)41 (47)0.343
11 (8)2 (15)6 (46)4 (31)
Tumour siteProximal11 (31)6 (17)7 (19)12 (33)0.004
Distal1 (3)8 (23)9 (26)17 (48)
Rectum1 (3)2 (7)10 (34)16 (55)
Tumour size
(median = 47)
<47 mm2 (4)9 (19)8 (17)29 (60)0.002
≥47 mm11 (22)7 (14)17 (35)14 (29)
SexFemale6 (12)7 (14)16 (33)20 (41)0.526
Male7 (14)9 (17)10 (20)25 (49)
DifferentiationWell/
Moderate
9 (10)14 (16)23 (27)40 (47)0.636
Poor3 (23)2 (15)3 (23)5 (38)
* EMAST-2/5 compared with EMAST-1/5, p = 0.023. ** Significant p values shown in bold.
Table 2. Association of MSI-H and MSI-L with clinicopathological features and molecular markers in sporadic colorectal cancer (cohort 1).
Table 2. Association of MSI-H and MSI-L with clinicopathological features and molecular markers in sporadic colorectal cancer (cohort 1).
Tumour
Feature
VariableMSI-H
n = 13
(%)
MSI-L
n = 13
(%)
MSS
n = 74
(%)
p *
Number of
EMAST
markers
01 (2)2 (4)42 (93)<0.0001
1–21 (2)11 (26)30 (71)-
3–511 (85)0 (0)2 (15)-
CIMPAbsent4 (7)10 (18)43 (75)0.003
CIMP-L3 (10)3 (10)24 (80)-
CIMP-H6 (46)0 (0)7 (54)-
CDKN2A (p16)
methylation
Absent7 (9)12 (16)56 (75)0.014
Present6 (24)1 (4)18 (72)-
MCC
methylation
Absent6 (8)11 (16)53 (76)0.004
Present7 (24)1 (3)21 (72)-
CDKN2A & MCC
methylation
Absent7 (8)13 (16)62 (76)0.007
Present6 (33)0 (0)12 (67)-
Dukes stage1.211 (19)2 (3)45 (78)0.007
32 (7)7 (25)19 (68)-
40 (0)4 (29)10 (71)-
T stage1,23 (11)4 (15)20 (74)0.907
3,410 (14)9 (12)54 (74)-
N stage011 (18)4 (7)46 (75)0.019
1,22 (5)9 (23)28 (72)-
M stage013 (15)10 (11)64 (74)0.190
10 (0)3 (23)10 (77)-
Tumour siteProximal10 (28)1 (3)25 (69)0.006
Distal2 (6)7 (20)26 (74)-
Rectum1 (3)5 (17)23 (79)-
Tumour size<47 mm4 (8)8 (17)36 (75)0.270
≥47 mm9 (18)5 (10)35 (71)-
SexFemale6 (12)6 (12)37 (76)0.944
Male7 (14)7 (14)37 (72)-
DifferentiationWell/
Moderate
10 (12)12 (14)64 (74)0.787
Poor2 (15)1 (8)10 (77)-
* Significant p values shown in bold.
Table 3. Association of MSH3 mRNA expression with MSI, EMAST and gene promoter methylation in sporadic colorectal cancers (cohort 1). Tumour MSH3 expression is shown relative to matched normal tissue and grouped according to median expression (0.63).
Table 3. Association of MSH3 mRNA expression with MSI, EMAST and gene promoter methylation in sporadic colorectal cancers (cohort 1). Tumour MSH3 expression is shown relative to matched normal tissue and grouped according to median expression (0.63).
Tumour FeatureVariableMSH3 mRNA
Expression <0.63
n = 48 (%)
MSH3 mRNA
Expression ≥0.63
n = 48 (%)
p *
Microsatellite
instability
H10 (77)3 (23)0.006
L2 (15)11 (85)-
S36 (51)34 (49)-
EMAST≥3/511 (85)2 (15)0.011
1–2/521 (51)20 (49)-
0/516 (38)26 (62)-
CIMPH11 (85)2 (15)0.006
L17 (57)13 (43)-
Absent20 (38)33 (62)-
CDKN2A (p16)
methylation
Yes16 (67)8 (33)0.098
No32 (44)40 (56)-
MCC
methylation
Yes23 (82)5 (18)0.0001
No25 (37)42 (63)-
* Significant p values shown in bold.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Meessen, S.; Currey, N.; Jahan, Z.; Parker, H.W.; Jenkins, M.A.; Buchanan, D.D.; Hopper, J.L.; Segelov, E.; Dahlstrom, J.E.; Kohonen-Corish, M.R.J. Tetranucleotide and Low Microsatellite Instability Are Inversely Associated with the CpG Island Methylator Phenotype in Colorectal Cancer. Cancers 2021, 13, 3529. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13143529

AMA Style

Meessen S, Currey N, Jahan Z, Parker HW, Jenkins MA, Buchanan DD, Hopper JL, Segelov E, Dahlstrom JE, Kohonen-Corish MRJ. Tetranucleotide and Low Microsatellite Instability Are Inversely Associated with the CpG Island Methylator Phenotype in Colorectal Cancer. Cancers. 2021; 13(14):3529. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13143529

Chicago/Turabian Style

Meessen, Sabine, Nicola Currey, Zeenat Jahan, Hannah W. Parker, Mark A. Jenkins, Daniel D. Buchanan, John L. Hopper, Eva Segelov, Jane E. Dahlstrom, and Maija R. J. Kohonen-Corish. 2021. "Tetranucleotide and Low Microsatellite Instability Are Inversely Associated with the CpG Island Methylator Phenotype in Colorectal Cancer" Cancers 13, no. 14: 3529. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13143529

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop