Next Article in Journal
Procoagulant Disorders in Patients with Newly Diagnosed Pancreatic Adenocarcinoma
Previous Article in Journal
Contrast-Enhanced Ultrasound for Assessing Abdominal Conditions in Pregnancy
Previous Article in Special Issue
Preliminary Results of an Exercise Program After Laparoscopic Resective Colorectal Cancer Surgery in Non-Metastatic Adenocarcinoma: A Pilot Study of a Randomized Control Trial
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Prognostic Value and Immunohistochemical Analysis of Mismatch Repair Deficiency in Patients with Stage II and III Colorectal Carcinoma—A Single-Center Study

by
Tijana Denčić
1,*,
Miljan Krstić
1,
Aleksandar Petrović
2,
Maja Jovičić-Milentijević
1,
Goran Radenković
2,
Marko Jović
2,
Nikola Živković
1,
Sonja Šalinger-Martinović
3,
Branko Branković
4 and
Simona Stojanović
5
1
Department of Pathology, Faculty of Medicine, University of Niš, Clinical Centre Niš, 18000 Niš, Serbia
2
Department of Histology and Embryology, Faculty of Medicine, University of Niš, 18000 Niš, Serbia
3
Department of Internal Medicine and Patient Care, Faculty of Medicine, University of Niš, Clinical Centre Niš, 18000 Niš, Serbia
4
Department of Surgery, Faculty of Medicine, University of Niš, Clinical Centre Niš, 18000 Niš, Serbia
5
Department of Oral Surgery, Faculty of Medicine, University of Niš, 18000 Niš, Serbia
*
Author to whom correspondence should be addressed.
Medicina 2020, 56(12), 676; https://doi.org/10.3390/medicina56120676
Submission received: 4 November 2020 / Revised: 1 December 2020 / Accepted: 4 December 2020 / Published: 8 December 2020
(This article belongs to the Special Issue Molecular and Therapeutic Landscapes in Colorectal Carcinoma)

Abstract

:
Background and objectives: Deficient mismatch repair (MMR) status is associated with good prognosis but poor therapeutic response to adjuvant chemotherapy in patients with colorectal cancer. However, there are some opposed arguments considering therapeutic outcomes in patients with evidenced MMR deficiency in colorectal cancer. The aim of the study was the investigation of prognostic value and immunohistochemical analysis of the MMR-deficiency tumors. Materials and Methods: The study enrolled 104 patients with resected stage II and III colorectal cancer samples from the period 2018–2019. Results: The tumors with deficient MMR status were significantly associated with age up to 50 years and right-sided localization (p < 0.001). During the follow-up period of 22.43 ± 6.66 months, 21 patients (20.2%) died, whereas 14 patients (13.5%) had relapses. The loss of mutL homologue 1/postmeiotic segregation increased 2 (MLH1/PMS2) expression, compared to proficient MMR tumors, was associated with shorter disease-free survival in patients with lymphovascular invasion (p < 0.05), perineural invasion (p < 0.01), stage III (p < 0.05) and high-grade tumor (p < 0.05). Conclusions: This retrospective pilot study of a single-center cohort of patients with stage II and III colorectal cancer highlights the clinical importance of using immunohistochemistry (IHC) analysis as a guide for diagnostic algorithm in a country with limited resources, but with a high prevalence of colorectal carcinoma in the young patients. MMR-deficiency tumors compared with proficient MMR colorectal cancer was not shown to be a significant predictor of disease-free and overall survival.

1. Introduction

Colorectal cancer (CRC) is a leading cause of cancer-related morbidity and mortality worldwide, especially in developing countries [1]. There are different pathways of colorectal carcinogenesis. Microsatellite instability (MSI) is a unique molecular change and a hypermutable phenotype that results from the loss of protein activities responsible for repairing errors during the DNA replication (MMR-mismatch repair), leading to the accumulation of mutations and alterations in microsatellite length [2]. Patients with hereditary non-polyposis colorectal cancer or Lynch syndrome demonstrate microsatellite instability in more than 90% of cases [3], while sporadic colorectal carcinoma is registered in 15% of cases [4].
MMR-deficiency (dMMR) tumor is associated with good prognosis but poor therapeutic response to adjuvant chemotherapy in patients with colorectal cancer stage II. Adjuvant chemotherapy may offer better survival benefit in stage III colorectal cancer [5]. On the other hand, in some published studies [6,7], dMMR was not associated with a favorable outcome and no survival differences were found between patients with dMMR colorectal cancers compared with those who had MMR-proficiency tumors (pMMR). However, there are some controversial arguments considering therapeutic outcomes in patients with evidenced dMMR tumors.
The College of American Pathologists recommended an initial evaluation for MMR deficiency by immunohistochemistry (IHC) testing with a four-antibody panel including MLH1, MSH2, MSH6, and PMS2. IHC testing for protein expression of the four MMR genes is a cheap, fast, and simple testing in everyday practice.
The aim of the study was the investigation of immunohistochemical analyses of dMMR tumors and the evaluation of the prognostic significance of MMR deficiency in paraffin-embedded tumor blocks of resected colorectal cancer stage II and III.

2. Materials and Methods

The study was conducted at the Clinical Center Niš, Republic of Serbia, and included 104 patients younger than 75 years of age, with resected colorectal carcinoma stage II and III. Patients with colorectal cancer who were treated with neoadjuvant chemotherapy and radiation therapy were excluded. The registry of pathological reports from the archive of the Pathology and Pathological Anatomy was used for the selection of formalin-fixed paraffin-embedded blocks with colorectal carcinoma samples collected in the period from 2018 to 2019. Follow-up data for these patients was collected until August the 10th 2020. This study obtained the approval of the local Ethical Committee No.:12-3340-2/1 (approval date: 26.05.2020). All experiments were performed in accordance with the relevant guidelines.
The histopathology reports comprised the following data: histopathological type of tumor, localization, low or high histological grade, pronounced or poor peritumoral lymphocytic infiltrate, positive or negative Crohn’s-like reaction, present or absent mucin secretion, and pathological stage.
Patients were distributed into two groups according to age—those younger than 50 and those over 50 years of age. Tumors localized in the cecum, ascending colon, and transverse colon are classified as right-sided, while tumors localized in descending colon, sigmoid colon, or the rectum are considered left-sided tumors. The tumor invasion depth, lymph nodes, and metastases status were used for pathological staging (TNM), as defined by the American Joint Committee on Cancer (AJCC). Crohn’s-like reaction, characterized by the presence of the lymphoid aggregates, of which some resemble the morphology of a lymph follicle, is present on the tumor periphery. Crohn’s-like reaction was reported as positive (the presence of at least two lymphoid follicles with germinal centers surrounding the tumor) or negative. Pronounced peritumoral lymphocytic infiltrate was a prominent lymphocytic reaction at the growing tumor margin without lymphoid aggregates. Other inflammatory cells were also present. Tumor budding was defined as single tumor cells or cluster of less than four cells at the invasive front of the tumor. The quantification of tumor budding was assessed on hematoxylin and eosin stained slides at magnification ×200. The specimen with the highest number of tumor budding was considered a hotspot (in a field measuring 0.785 mm 2). We used a two-tier system: low-grade tumor budding (0–9 tumor buds) and high-grade tumor budding (≥ 10 tumor buds). Adjuvant chemotherapy with 5-fluorouracil (5-FU) was given to patients with stage II and III tumors, following the standard schedules and doses.
All immunohistochemistry analyses were done manually and performed in the laboratories at the Department of Histology and Embryology, Faculty of Medicine, University of Niš, Serbia. The tissues slides were dewaxed by heating at 55 °C for 30 min and by three washes, 5 min each with xylene. Tissues were rehydrated by a series of five-minute washes in 100%, 95%, and 80% ethanol and distilled water. Endogenous peroxidase activity was blocked with 3% hydrogen peroxide for ten minutes. The antigen was unmasked in a water bath for 30 min and the samples were incubated with the primary antibody at room temperature for 40 min. The samples were then incubated with secondary antibody for 20 min, each at room temperature. The monoclonal antibodies (mutL homologue 1-MLH1, clone ES05; postmeiotic segregation increased 2-PMS2, clone EP51; mutS homologue 2-MSH2, clone FE11; and mutS homologue 6-MSH6, clone EP49) were manufactured by the same company (Dako, Glostrup, Denmark) in the form ready-to-use. The system DAKO En Vision with chromogen diaminobenzidine (DAB) was used for visualization. Counterstaining was performed with Mayer’s hematoxylin solution. After rewashing the slides in water for 5 minutes, we finally dehydrated, cleared, and mounted the sections. The slides were ready for microscopic observation. Microscopic images of pathohistological specimens were obtained at the Department of Histology and Embryology, Faculty of Medicine, Niš by recording tissue specimens on the optical microscope (Olympus BΧ50, Tokyo, Japan), equipped with digital camera DFC 295 (Leica Microsystems, Wetzlar, Germany). Immunohistochemical markers of the presence or absence of nuclear immunoreactivity of tumor cells were determined by using a positive internal control for each specimen: unchanged colorectal epithelium, lymphocytes and stromal cells. The presence of nuclear expression was regarded as a positive result (proficient MMR), while nuclear immunoreactivity loss of one or more MMR proteins was regarded as a negative result (deficient MMR).

Statistical Analysis

Statistical analysis was performed using the software SPSS ver. 17.0 (SPSS Inc, Chicago, IL, USA). Categorical variables are given as frequencies (n) and percentages (%). Comparison of the distribution of clinical and histopathological features between the examined groups was performed using the Chi-Square and Fisher’s exact test. Analysis of the relationship between MMR-deficient cancers and factors of interest was performed by univariate and multivariate logistic regression analysis (the odds ratio value (OR) and its 95% confidence interval—95% CI were calculated). Statistical significance was defined as p < 0.05.

3. Results

The loss of nuclear expression of at least one MMR protein was recorded in 12 (11.54%) cases. The loss of expression of heterodimer pair MLH1/PMS2 protein was found in 7 (6.73%) patients (Figure 1), while the loss of MSH2/MSH6 and MSH6 expression was evidenced in 1 (0.96%) patient. An isolated loss of PMS2 expression was present in 3 (2.88%) cases. The isolated loss of nuclear expression of MLH1 and MSH2 protein was not detected.
Demographic, clinical, and histopathological features of patients are given in Table 1.
Univariate logistic regression analysis showed that significant predictors of dMMR were: age ≤50 years, right-sided localization as well as the positive Crohn’s-like reaction and high grade tumor budding (Table 2). Expression of the following factors points to a higher probability of dMMR presence: age ≤50 years (p < 0.001), right-sided tumor localization (p < 0.001,) Crohn’s-like reaction (p < 0.001), and high-grade tumor budding (p < 0.001). There was no statistically significant association between the dMMR and the following parameters: gender, tumor type, mucin secretion, peritumoral lymphocytic infiltration, tumor grade, lymphovascular and perineural invasion, 5-FU based chemotherapy, and tumor stage.
Using a multivariate logistic regression analysis, age ≤50 years and the right-sided localization of tumor were shown as the most important predictors of dMMR tumors (p < 0.001 and p < 0.01, respectively) (Table 2).
Follow-up data for the whole group of patients was collected until August 10, 2020. During the follow-up period of 22.43 ± 6.66 months, 21 patients (20.2%) died, and 14 patients (13.5%) had relapses. Demographic, clinical, and histopathological features according to relapse occurrence are presented in Table 3. Relapses were more frequent in patients with high-grade tumor (p < 0.05).
Using a univariate Cox regression analysis, a possible predictor of disease-free survival was identified-the presence of a high-grade tumor. In the multivariate model (χ2 = 8.426, p < 0.05), none of the variables was shown to be an independent predictor of disease-free survival (Table 4).
MMR-deficiency tumors compared with proficient MMR colorectal cancer was not shown to be a significant predictor of disease-free survival. In colorectal carcinoma, MMR-proficiency tumors were predictors of longer disease-free survival compared to any loss of MMR protein expression (p < 0.05), especially MLH1/PMS2 loss (p < 0.01).
The loss of MLH1/PMS2 expression, compared to pMMR, was associated with shorter disease-free survival in patients with lymphovascular invasion (p < 0.05), perineural invasion (p < 0.01), stage III (p < 0.05), and high-grade tumor (p < 0.05)—Figure 2.
Demographic, clinical and histopathological features according to life status are given in Table 5. Death event was associated with mucinous secretion (p < 0.05), lymphovascular invasion or stage III (p < 0.05), high-grade tumor (p < 0.001), and previous relapse (p < 0.001).
Even though a univariate Cox regression analysis has identified a number of possible predictors of the overall survival in the studied group, the only independent predictors in the multivariate model (χ2 = 27.196, p < 0.001) were older age (p < 0.05) and high-grade tumor (p < 0.05) (Table 6).

4. Discussion

The molecular phenotype of colorectal carcinoma is closely related to demographic, clinical, and pathohistological features; biological behavior; prognosis; and therapy response [2]. Previous studies [8,9,10,11,12,13] showed different features of dMMR tumors, such as female gender, younger age of patients, poor differentiation, presence of extracellular and intracellular mucin, proximal localization in the colon, Crohn’s-like reaction, and pronounced lymphocytic infiltration.
Our results show that right-sided colon cancers are statistically significantly associated with dMMR, which is in keeping with literature data [4,8,9,10,11,12,13]. The association between younger age of a patient and the loss of MMR proteins expression by IHC was also markedly evident and consistent with literature data [4,8,9,10,11,12,13]. A statistically significant association between MMR deficiency and younger age of patients and right-sided tumor localization is another in a series of diagnostic parameters for the approach to a specific group of patients with CRC.
The use of IHC testing for deficient MMR to document protein expression in our center is not available despite international guidelines. The prevalence of dMMR in our study is relatively lower in comparison to literature data, but previous studies in Japan [13] and the USA [14] reported similar results a deficient MMR tumor prevalence of 8.4% and 10.7%, respectively. The most frequent expression pattern in our study was a combined loss of MLH1 and PMS2 proteins, with 6.73% of the overall tissue sample, which is also consistent with previous literature data [11]. In the Pakistan population, colorectal cancer with dMMR comprised 34% of the overall tissue sample [8], 33% in Egypt [11], 21% in Singapore [15], and 15% in western studies [16].
The most commonly used adjuvant chemotherapy for the treatment of CRC is the 5-FU. Since the initial discovery of MSI in CRC, a number of studies have confirmed a better outcome of patients with MMR-deficiency CRC in stage II and III compared with patients who have MMR-proficiency tumors [17,18]. Sinicrope et al. observed that, compared with patients who have proficient MMR tumors, patients with MMR-deficiency tumors experienced statistically significant improvements in disease-free and overall survival [19]. An association of dMMR tumors with improved outcomes was observed in patients with stage II and III but was statistically significant only in stage III when dMMR was compared with pMMR tumors [20,21]. A meta-analysis of patients with stage II and III CRC treated with 5-FU-based adjuvant chemotherapy demonstrated no survival benefit for patients with MMR-deficiency tumor [22]. Kontourakis et al. recommended adjuvant chemotherapy for all patients with high-risk stage II and stage III colorectal cancer [5]. Lamberti C et al. did not find an association of dMMR tumors with a favorable outcome compared with those who had MMR-proficiency tumors [6]. A relatively small subset of dMMR tumor cases may explain described data discrepancies and controversial arguments considering therapeutic outcomes in patients with evidenced MMR-deficiency tumors.
In our study, a median follow-up period was approximately two years (22.43 ± 6.66 months), and during this time 21 patients (20.2%) died (13.5%). In our patients, population relapses were more frequent in patients with high-grade tumor (p < 0.05). Using a univariate Cox regression analysis, we identified a possible predictor of disease-free survival, but in the multivariate model, none of the variables was shown to be a significant independent predictor. MMR-deficiency was not approved as a significant predictor of disease-free survival. MMR-proficiency colorectal adenocarcinoma was a predictor of longer disease-free survival compared to any loss of MMR expression, especially loss of MLH1/PMS2 protein expression. The loss of MLH1/PMS2 protein expression was associated with shorter disease-free survival in patients with lymphovascular invasion, perineural invasion, stage III, and high-grade tumors, compared to MMR-proficiency tumors. Lethal outcome was associated with mucinous adenocarcinoma, perineural and lymphovascular invasion, stage III, high-grade tumor, and previous relapse. Stage III, high-grade tumor, and lymphovascular and perineural invasion are independently associated with dMMR CRC recurrence, suggesting that screening for this factor in routine clinical practice may assist with adjuvant chemotherapy decision-making. There were no statistically significant differences in the overall survival between dMMR and pMMR tumors. The only independent predictors in the multivariate model were greater age and high tumor grade in survival times.
This retrospective study had a few limitations: small number of patients and unavailable IHC screening for MMR-deficiency in everyday, routine practice. There were no available data on comorbidities that may influence the outcome.

5. Conclusions

This retrospective pilot study of a single-center cohort of patients with stage II and III colorectal cancer highlights the clinical importance of using IHC analysis as a guide for diagnostic algorithm in an early stage of CRC. A statistically significant association of MMR deficiency with younger patients and right-sided tumor localization is another in a series of diagnostic parameters for the approach to a specific group of patients with colorectal cancer. In a country with limited resources, but with a high prevalence of colorectal carcinoma in the young patients, IHC analysis for deficient MMR proteins is recommended. MMR-deficiency was not shown as a significant predictor of disease-free survival and overall survival in stage II and III CRC. The objective of future research is to detect biomarkers that could provide the identification of the prognostic panel of biomarkers and the characterization of predictive biomarkers to help in the selection of the most appropriate therapy.

Author Contributions

T.D.—study design, wrote the manuscript, methodology, immunochistohemical analysis, corresponding author, M.K.—internal revision of the whole manuscript, A.P.—methodology, immunochistohemical analysis, M.J.-M.—Supervision of the research, G.R.—Methodology, M.J.—Coordinated the research, N.Ž.—revised the manuscript, S.Š.-M.—statistical analysis, B.B.—made the database, S.S.—checked the reference list. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

This work has been supported by the Internal Scientific Research Project of the Faculty of Medicine, University of Niš (INT-MFN, No.38/20).

Conflicts of Interest

The authors declare that they have no conflict of interests.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Battaglin, F.; Naseem, M.; Lenz, H.J.; Salem, M.E. Microsatellite instability in colorectal cancer: Overview of its clinical significance and novel perspectives. Clin. Adv. Hematol. Oncol. 2018, 16, 735–745. [Google Scholar] [PubMed]
  3. De Jong, A.E.; Morreau, H.; Van Puijenbroek, M.; Eilers, P.H.; Wijnen, J.; Nagengast, F.M.; Griffioen, G.; Cats, A.; Menko, F.H.; Kleibeuker, J.H.; et al. The role of mismatch repair gene defects in the development of adenomas in patients with HNPCC. Gastroenterology 2004, 126, 42–48. [Google Scholar] [CrossRef] [PubMed]
  4. Kim, H.; Jen, J.; Vogelstein, B.; Hamilton, S. Clinical and pathological characteristics of sporadic colorectal carcinomas with DNA replication errors in microsatellite sequences. Am. J. Pathol. 1994, 145, 148–156. [Google Scholar]
  5. Kountourakis, P.; Souglakos, J.; Gouvas, N.; Androulakis, N.; Athanasiadis, A.; Boukovinas, I.; Christodoulou, C.; Chrysou, E.; Dervenis, C.; Emmanouilidis, C.; et al. Adjuvant chemotherapy for colon cancer: A consensus statement of the Hellenic and Cypriot Colorectal Cancer Study Group by the HeSMO. Ann. Gastroenterol. 2016, 29, 18–23. [Google Scholar]
  6. Lamberti, C.; Lundin, S.; Bogdanow, M.; Pagenstecher, C.; Friedrichs, N.; Büttner, R.; Sauerbruch, T. Microsatellite instability did not predict individual survival of unselected patients with colorectal cancer. Int. J. Colorectal Dis. 2007, 22, 145–152. [Google Scholar] [CrossRef]
  7. Kim, G.P.; Colangelo, L.H.; Wieand, H.S.; Paik, S.; Kirsch, I.R.; Wolmark, N.; Allegra, C.J. Prognostic and predictive roles of high-degree microsatellite instability in colon cancer: A National Cancer Institute-National Surgical Adjuvant Breast and Bowel Project Collaborative Study. J. Clin. Oncol. 2007, 25, 767–772. [Google Scholar] [CrossRef]
  8. Hashmi, A.A.; Ali, R.; Hussain, Z.F.; Faridi, N.; Khan, E.Y.; Bakar, S.M.A.; Edhi, M.M.; Khan, M. 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, 116. [Google Scholar] [CrossRef] [Green Version]
  9. Raut, C.; Pawlik, T.; Rodriguez-Bigas, M. Clinicopathologic features in colorectal cancer patients with microsatellite instability. Mutat. Res./Fundam. Mol. Mech. Mutagenesis 2004, 568, 275–282. [Google Scholar] [CrossRef]
  10. Kawakami, H.; Zaanan, A.; Sinicrope, F. Microsatellite instability testing and its role in the management of colorectal cancer. Curr. Treat. Options Oncol. 2015, 16, 30. [Google Scholar] [CrossRef]
  11. Soliman, N.A.; Morsia, D.F.; Helmy, N.A.H. Immunohistochemical Expression of MMR Proteins with Clinicopathological Correlation in Colorectal Cancer in Egypt. Open Access Maced. J. Med. Sci. 2019, 7, 1608–1617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Bai, H.; Wang, R.; Cheng, W.; Shen, Y.; Li, H.; Xia, W.; Ding, Z.; Zhang, Y. Evaluation of Concordance Between Deficient Mismatch Repair and Microsatellite Instability Testing and Their Association with Clinicopathological Features in Colorectal Cancer. Cancer Manag. Res. 2020, 12, 2863–2873. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Suzuki, O.; Eguchi, H.; Chika, N.; Sakimoto, T.; Ishibashi, K.; Kumamoto, K.; Tamaru, J.I.; Tachikawa, T.; Akagi, K.; Arai, T.; et al. Prevalence and clinicopathologic/molecular characteristics of mismatch repair-deficient colorectal cancer in the under-50-year-old Japanese population. Surg. Today 2017, 47, 1135–1146. [Google Scholar] [CrossRef]
  14. Pearlman, R.; Frankel, W.L.; Swanson, B.; Zhao, W.; Yilmaz, A.; Miller, K.; Bacher, J.; Bigley, C.; Nelsen, L.; Goodfellow, P.J.; et al. Prevalence and Spectrum of Germline Cancer Susceptibility Gene Mutations Among Patients with Early-Onset Colorectal Cancer. JAMA Oncol. 2017, 3, 464–471. [Google Scholar] [CrossRef]
  15. Chew, M.H.; Koh, P.K.; Tan, M.; Lim, K.H.; Carol, L.; Tang, C.L. Mismatch repair deficiency screening via immunohistochemical staining in young Asians with colorectal cancers. World J. Surg. 2013, 37, 2468–2475. [Google Scholar] [CrossRef]
  16. Guastadisegni, C.; Colafranceschi, M.; Ottini, L.; Dogliotti, E. Microsatellite instability as a marker of prognosis and response to therapy: A meta-analysis of colorectal cancer survival data. Eur. J. Cancer 2010, 46, 2788–2798. [Google Scholar] [CrossRef]
  17. Lanza, G.; Gafa, R.; Santini, A.; Maestri, I.; Guerzoni, L.; Cavazzini, L. Immunohistochemical test for MLH1 and MSH2 expression predicts clinical outcome in stage II and III colorectal cancer patients. J. Clin. Oncol. 2006, 24, 2359–2367. [Google Scholar] [CrossRef] [PubMed]
  18. Gong, Q.; Zhang, H.H.; Sun, S.B.; Ge, W.M.; Li, Y.; Yhu, Y.C.; Le-Ping, L. Mismatch repair-deficient status associates with favorable prognosis of Eastern Chinese population with sporadic colorectal cancer. Oncol. Lett. 2018, 15, 7007–7013. [Google Scholar] [CrossRef] [PubMed]
  19. Sinicrope, F.A.; Foster, N.R.; Thibodeau, S.N.; Marsoni, S.; Monges, G.; Labianca, R.; Kim, G.P.; Yothers, G.; Allegra, C.; Moore, M.J.; 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, 863–875. [Google Scholar] [CrossRef] [Green Version]
  20. Roth, A.D.; Tejpar, S.; Delorenzi, M.; Yan, P.; Fiocca, R.; Klingbiel, D.; Dietrich, D.; Biesmans, B.; Bodoky, G.; Barone, C.; et al. Prognostic role of KRAS and BRAF in stage II and III resected colon cancer: Results of the translational study on the PETACC-3, EORTC 40993, SAKK 60-00 trial. J. Clin. Oncol. 2010, 28, 466–474. [Google Scholar] [CrossRef]
  21. Li, P.; Xiao, Z.; Braciak, T.A.; Ou, Q.; Chen, G.; Oduncu, F.S. Systematic immunohistochemical screening for mismatch repair and ERCC1 gene expression from colorectal cancers in China: Clinicopathological characteristics and effects on survival. PLoS ONE 2017, 12, doi. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Popat, S.; Hubner, R.; Houlston, R.S. Systematic review of microsatellite instability and colorectal cancer prognosis. J. Clin. Oncol. 2005, 23, 609–618. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Immunohistochemical staining pattern of a deficient mismatch repair (dMMR colorectal carcinoma ×100, with the loss of MLH1 (A), loss of PMS2 (B), and intact staining of MSH2 (C) and MSH6 (D).
Figure 1. Immunohistochemical staining pattern of a deficient mismatch repair (dMMR colorectal carcinoma ×100, with the loss of MLH1 (A), loss of PMS2 (B), and intact staining of MSH2 (C) and MSH6 (D).
Medicina 56 00676 g001
Figure 2. Kaplan–Meier curve of disease-free survival according to MMR status (deficient or proficient MMR) in patients with lymphovascular invasion (a), perineural invasion (b), stage III (c), high-grade tumor (d).
Figure 2. Kaplan–Meier curve of disease-free survival according to MMR status (deficient or proficient MMR) in patients with lymphovascular invasion (a), perineural invasion (b), stage III (c), high-grade tumor (d).
Medicina 56 00676 g002
Table 1. Expression of MMR proteins in colorectal carcinoma and its association with demographic, clinical, and histopathological features.
Table 1. Expression of MMR proteins in colorectal carcinoma and its association with demographic, clinical, and histopathological features.
FeaturesdMMR
(n = 12)
Expression of All MMR Proteins
(n = 92)
Total
(n = 104)
p Value
Age (years)
≤509(75.00%)6(6.52%)15(14.42%)0.0000 ***
>503(25.00%)86(93.48%)89(85.58%)
Gender
Female7(58.33%)39(42.39%)46(44.23%)0.2980
Male5(41.67%)53(57.61%)58(55.77%)
Tumor localization
Right10(83.33%)22(23.91%)32(30.77%)0.0001 ***
Left2(16.67%)70(76.09%)72(69.23%)
Chemotherapy given (yes)11(91.67%)62(67.39%)73(70.19%)0.1031
TNM staging
Stage III758.33%4852.17%5552.88%0.765
Stage II541.67%4447.83%4947.12%
Tumor histology
Conventional adenocarcinoma10(83.33%)66(71.74%)76(73.08%)0.5066
Mucinous adenocarcinoma2(16.67%)26(28.26%)28(26.92%)
Tumor grade
High-grade9(75.00%)53(57.61%)62(59.62%)0.3525
Low-grade3(25.00%)39(42.39%)42(40.38%)
Mucinous secretion
Present7(58.33%)28(30.43%)35(33.65%)0.0999
Absent5(41.67%)64(69.57%)69(66.35%)
Peritumoral lymphocytic infiltrate
Pronounced8(66.67%)39(42.39%)47(45.19%)0.1137
Poor4(33.33%)53(57.61%)57(54.81%)
Crohn’s-like reaction
Positive8(66.67%)13(14.13%)21(20.19%)0.0002 ***
Negative4(33.33%)79(85.87%)83(79.81%)
Tumor budding
High-grade9(75.00%)17(18.48%)26(25.00%)0.0000
Low-grade3(25.00%)7581.25%7875.00%
Lymphovascular invasion
Present866.67%4953.26%5754.81%0.540
Absent433.33%4346.74%4745.19%
Perineural invasion
Present650.00%4346.74%4947.12%10.000
Absent650.00%4953.26%5552.88%
Data were presented as frequencies (n) and in percentage (%)); ***—p < 0.001; tumor nodes metastases, TNM; deficient mismatch repair, dMMR; mismatch repair, MMR.
Table 2. Odds Ratio values for dMMR status prediction factors (univariate and multivariate logistic regression analysis).
Table 2. Odds Ratio values for dMMR status prediction factors (univariate and multivariate logistic regression analysis).
Univariate Logistic Regressionp ValueOR(95% CI of OR)
Factor
Age ≤50 years<0.00143.000(9.157–201.922)
Female gender0.30141.903(0.562–6.443)
Right-sided tumor localization<0.00115.909(3.238–78.168)
TNM stage (III)0.6881.283(0.379–4.340)
Conventional adenocarcinoma0.40181.970(0.404–9.606)
High-grade tumor0.25742.208(0.561–8.692)
Present mucin secretion0.06393.200(0.935–10.954)
Pronounced peritumoral lymphocytic infiltration0.12262.718(0.764–9.673)
Crohn’s-like reaction<0.00112.154(3.195–46.227)
Tumor budding (high-grade)<0.00113.235(3.235–54.142)
Lymphovascular invasion (yes)0.3851.755(0.494–6.238)
Perineural invasion (yes)0.8321.140(0.342–3.796)
Multivariate logistic regression (Step 2)p ValueOR(95% CI of OR)
Factor
Age ≤50 years<0.00177.924(8.029–756.225)
Right-sided tumor localization0.004830.491(2.843–327.060)
Crohn’s-like reaction0.2363.483(0.442–27.422)
Tumor budding (high-grade)0.2802.872(0.424–19.436)
Constant<0.0010.004(3.195–46.227)
OR—Odds ratio, CI—Confidence interval.
Table 3. Demographic, clinical, and histopathological features according to relapse occurrence.
Table 3. Demographic, clinical, and histopathological features according to relapse occurrence.
FeaturesRelapseNo Relapsep Value
Age (years)70.0 (62.8–73.2)66.0 (54.0–71.2)1.364 (0.172)
Gender (male)6 (42.9%)52 (57.8%)0.572 (0.388)
Localization (right-sided)7 (50.0%)25 (27.8%)1.862 (0.121)
Chemotherapy given (yes)11 (78.6%)62 (68.9%)0.179 (0.547)
TNM stage (III)10 (71.4%)45 (50.0%)1.456 (0.160)
Tumor histology (conventional adenocarcinoma)8 (57.1%)68 (75.6%)1.257 (0.195)
Tumor grade (high)12 (85.7%)50 (55.6%)3.410 (0.041)
Mucinous secretion (yes)7 (50.0%)28 (31.1%)1.182 (0.224)
Crohn-like reaction (positive)4 (28.6%)17 (18.9%)0.232 (0.474)
High-grade tumor budding (yes)5 (35.7%)21 (23.3%)0.440 (0.332)
Lymphovascular invasion (yes)8 (57.1%)49 (54.4%)0.000 (1.000)
Perineural invasion (yes)7 (50.0%)42 (46.7%)0.000 (1.000)
MMR status (proficient vs deficient)12 (85.7%)80 (88.9%)0.000 (0.663)
MMR status Proficient12 (85.7%)80 (88.9%)2.163 (0.706)
MLH1/PMS2 loss2 (14.3%)5 (5.6%)
PMS2 loss0 (0.0%)3 (3.3%)
MSH2/MSH6 loss0 (0.0%)1 (1.1%)
MSH6 loss0 (0.0%)1 (1.1%)
Table 4. Cox regression analysis of disease-free survival.
Table 4. Cox regression analysis of disease-free survival.
UnivariateMultivariate
HR (95% CI for HR)p ValueHR (95% CI for HR)p Value
Tumor Grade (High)4.914 (1.098–21.992)0.0372.935 (0.556–15.499)0.205
Hazard ratio, HR; Confidence interval, CI.
Table 5. Demographic, clinical, and histopathological features with reference to fatal outcome.
Table 5. Demographic, clinical, and histopathological features with reference to fatal outcome.
FeaturesDeathNo-Deathp Value
Age (years)70.0 (62.8–73.2)66.0 (54.0–71.2)1.364 (0.172)
Gender (male)11 (52.4%)47 (56.6%)0.011 (0.808)
Localization (right-sided)9 (42.9%)23 (27.7%)1.164 (0.195)
Chemotherapy given (yes)18 (85.7%)55 (66.3%)2.172 (0.110)
Relapse (yes)8 (38.1%)6 (7.2%)11.185 (0.001)
TNM stage (III)16 (76.2%)39 (47.0%)4.624 (0.026)
Tumor histology (adenocarcinoma)11 (52.4%)65 (78.3%)4.486 (0.026)
Tumor grade (high)19 (90.5%)43 (51.8%)8.865 (0.001)
Mucinous secretion (yes)10 (47.6%)25 (30.1%)1.581 (0.195)
Crohn-like reaction (positive)4 (19.0%)17 (20.5%)0.000 (1.000)
High-grade tumor budding (yes)3 (14.3%)23 (27.7%)0.975 (0.266)
Lymphovascular invasion (yes)15 (71.4%)42 (50.6%)2.154 (0.140)
Perineural invasion (yes)14 (66.7%)35 (42.2%)3.113 (0.053)
MMR status (proficient vs. deficient)20 (95.2%)72 (86.7%)0.498 (0.452)
MMR status Proficient20 (95.2%)72 (86.7%)1.553 (0.817)
MLH1/PMS2 loss1 (4.8%)6 (7.2%)
PMS2 loss0 (0.0%)3 (3.6%)
MSH2/MSH6 loss0 (0.0%)1 (1.2%)
MSH6 loss0 (0.0%)1 (1.2%)
Table 6. Cox regression analysis of the overall survival.
Table 6. Cox regression analysis of the overall survival.
FeaturesUnivariateMultivariate
HR (95% CI for HR)pHR (95% CI for HR)p
Age (years)1.095 (1.014–1.184)0.0211.104 (1.013–1.204)0.026
TNM stage (III)3.068 (1.123–8.381)0.0290.789 (0.224–2.776)0.711
Tumor grade (high)7.575 (1.764–32.532)0.0067.637 (1.163–50.162)0.034
Relapse (yes)5.395 (2.207–13.189)0.0002.678 (0.983–7.294)0.054
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Denčić, T.; Krstić, M.; Petrović, A.; Jovičić-Milentijević, M.; Radenković, G.; Jović, M.; Živković, N.; Šalinger-Martinović, S.; Branković, B.; Stojanović, S. Prognostic Value and Immunohistochemical Analysis of Mismatch Repair Deficiency in Patients with Stage II and III Colorectal Carcinoma—A Single-Center Study. Medicina 2020, 56, 676. https://doi.org/10.3390/medicina56120676

AMA Style

Denčić T, Krstić M, Petrović A, Jovičić-Milentijević M, Radenković G, Jović M, Živković N, Šalinger-Martinović S, Branković B, Stojanović S. Prognostic Value and Immunohistochemical Analysis of Mismatch Repair Deficiency in Patients with Stage II and III Colorectal Carcinoma—A Single-Center Study. Medicina. 2020; 56(12):676. https://doi.org/10.3390/medicina56120676

Chicago/Turabian Style

Denčić, Tijana, Miljan Krstić, Aleksandar Petrović, Maja Jovičić-Milentijević, Goran Radenković, Marko Jović, Nikola Živković, Sonja Šalinger-Martinović, Branko Branković, and Simona Stojanović. 2020. "Prognostic Value and Immunohistochemical Analysis of Mismatch Repair Deficiency in Patients with Stage II and III Colorectal Carcinoma—A Single-Center Study" Medicina 56, no. 12: 676. https://doi.org/10.3390/medicina56120676

Article Metrics

Back to TopTop