Skip to main content
International Journal of Environmental Research and Public Health logoLink to International Journal of Environmental Research and Public Health
. 2016 Sep 8;13(9):892. doi: 10.3390/ijerph13090892

Role of Deficient Mismatch Repair in the Personalized Management of Colorectal Cancer

Cong-Min Zhang 1,2, Jin-Feng Lv 3,4, Liang Gong 1,2, Lin-Yu Yu 1,2, Xiao-Ping Chen 1,2, Hong-Hao Zhou 1,2, Lan Fan 1,2,*
Editor: William Chi-shing Cho
PMCID: PMC5036725  PMID: 27618077

Abstract

Colorectal cancer (CRC) represents the third most common type of cancer in developed countries and one of the leading causes of cancer deaths worldwide. Personalized management of CRC has gained increasing attention since there are large inter-individual variations in the prognosis and response to drugs used to treat CRC owing to molecular heterogeneity. Approximately 15% of CRCs are caused by deficient mismatch repair (dMMR) characterized by microsatellite instability (MSI) phenotype. The present review is aimed at highlighting the role of MMR status in informing prognosis and personalized treatment of CRC including adjuvant chemotherapy, targeted therapy, and immune checkpoint inhibitor therapy to guide the individualized therapy of CRC.

Keywords: colorectal cancer, personalized treatment, mismatch repair system, microsatellite instability, adjuvant chemotherapy, targeted therapy, immune checkpoint inhibitor therapy

1. Introduction

Colorectal cancer (CRC) represents the third most common type of cancer in developed countries and one of the leading causes of cancer deaths worldwide [1]. For primary CRC, the tumor-node-metastasis (TNM) stage is considered the gold standard for informing prognosis and treatment after resection [2]. Nevertheless, it is fair to admit that there is considerable stage-independent variability in the clinical outcome of CRC, which might result from molecular heterogeneity of the tumors [3]. Hence, the identification of molecular markers for prognosis and treatment of CRC is urgently needed to achieve personalized management of CRC.

It has been shown that there are two recognized pathways contributing to CRC development [4]. The majority of CRCs develop via a chromosomal instability pathway (CIN), and approximately 15% are caused by deficient mismatch repair (dMMR) [4]. Molecular heterogeneity is commonly considered pivotal to guide the management of CRCs [5]. Coincident data demonstrated that compared with those with proficient MMR (pMMR) tumors, dMMR CRC patients had better stage-adjusted clinical outcome [6,7] and might benefit differently from a variety of therapies including adjuvant chemotherapy (fluoropyrimidine, platinum compounds, topoisomerase inhibitors, and alkylating agents), targeted therapy (anti-epithelial growth factor receptor (EGFR) or anti-vascular endothelial growth factor (VEGF) antibodies) and immune checkpoint inhibitor therapy (anti-programmed cell death 1 (PD-1) antibodies) [2,3,8,9]. Here we discuss the prognostic and predictive value of MMR status in the clinical personalized management of CRC.

2. dMMR in CRC

2.1. MMR System, dMMR, and Microsatellite Instability (MSI) Phenotype

DNA synthesis is an error-prone process which generates incorrect base-pairing (base-base mismatches) or unmatched DNA loops (insertion-deletion loops). As one of the DNA repair mechanisms, the MMR system can repair these errors to maintain genomic stability. The MMR system is composed of a series of MMR proteins including MutL homolog 1 (MLH1), MutL homolog 3 (MLH3), MutS homolog 2 (MSH2), MutS homolog 3 (MSH3), MutS homolog 6 (MSH6), postmeiotic segregation increased 1 (PMS1), and postmeiotic segregation increased 2 (PMS2) [10]. These MMR proteins function by forming heterodimer complexes MutS and MutL. Consisting of two major forms MutSα (a MSH2/MSH6 heterodimer) and MutSβ (a MSH2/MSH3 heterodimer), MutS performs the initial recognition of mismatches [11]. MutSα recognizes and binds to base-base mismatches and short (1–2) insertion deletion loops (IDLs), whereas MutSβ detects larger (≥2) IDLs [9]. Then, MutL together with other repair proteins including PCNA (proliferating-cell-nuclear-antigen) and exonucleases completes the DNA repair process [9,11]. MutL homologs include MutLα (a MLH1/PMS2 heterodimer), MutLβ (a MLH1/PMS1 heterodimer), and MutLγ (a MLH1/MLH3 heterodimer) [11]. MutLα plays a more major role in DNA mismatch repair compared with the other two [12]. MMR genes generally act as tumor suppressor genes. dMMR commonly results from a consequence of germline mutations in MMR genes, somatic MMR gene alterations, or epigenetic silencing of MMR gene expression [13].

Microsatellites are short (1–6 base pairs), tandem repeated sequences that are scattered throughout the genome and very susceptive to replication errors induced by the slippage of DNA polymerases [14]. Generally, these errors can be corrected by the MMR system to keep the stability of microsatellites. When the MMR system is deficient owing to genetic or epigenetic events, tumors exhibit MSI phenotype. Thus, it is a well-established concept that MSI serves as a phenotypic indicator of dMMR.

2.2. dMMR CRC

dMMR CRCs account for about 15% of all primary CRCs [13] including sporadic CRCs (12%) and Lynch syndrome (LS) (3%) [15]. CRC patients with dMMR tumors have unique clinicopathological features such as proximal colon preponderance, poor differentiation, early stage and abundant tumor-infiltrating lymphocytes compared with those displaying pMMR tumors (Box 1).

Box 1. Clinicopathological features of dMMR tumors [16,17,18,19,20].

  1. Proximal Colon Predominance (70% Proximal to the Splenic Flexure)

  2. Poor Differentiation

  3. Tumor Heterogeneity

  4. Large and Lymph-Node-negative

  5. Excess of Mucinous (15%), Signet Cell and Medullary Subtypes

  6. Prominent Anti-tumor Host Response (Increased Tumor-Infiltrating Lymphocytes as Well as “Crohn-like” Reaction)

  7. Accelerated Carcinogenesis from Tiny Adenoma to Carcinoma within 2–3 Years in Lynch Syndrome Cases

Abbreviation: dMMR, deficient mismatch repair.

2.2.1. dMMR in LS

LS is an inherited autosomal-dominant disorder, also known as hereditary nonpolyposis colorectal cancer (HNPCC). The development of LS requires ”two hit“ inactivation of both alleles of the MMR gene. LS is caused by a germline inactivating mutation in one of the MMR genes, commonly MLH1 or MSH2, infrequently MSH6 or PMS2 (the first hit). Subsequently, the remaining allele would lose function through somatic mutation, loss of heterozygosity (LOH) or promoter methylation (the second hit) leading to LS [13]. In addition, germline mutation in the epithelial cell adhesion molecule (EPCAM), a gene located upstream of MSH2 can cause epigenetic inactivation of MSH2 leading to LS [21]. LS patients are diagnosed at an earlier age, and are at high risk of various cancers such as stomach cancer, ovary cancer, urinary tract cancer, small intestine cancer, and prostate cancer. The revised Bethesda Guidelines (RBG) was developed to identify individuals at risk of LS by testing for dMMR/MSI of tumors (Box 2).

Box 2. Revised Bethesda Guidelines for MSI testing [22,23,24,25].
  1. CRC Diagnosed in a Patient Younger than 50 Years

  2. Presence of Synchronous or Metachronous CRC or Other Lynch Syndrome-Associated Tumor *, Regardless of Age

  3. CRC with MSI-H Pathological Features # Diagnosed in a Patient Younger than 60 Years

  4. Patient with CRC and CRC or Lynch Syndrome-associated Tumor * Diagnosed in at Least One First-Degree Relative Less than 50 Years of Age

  5. Patient with CRC and CRC or Lynch Syndrome-Associated Tumor * Diagnosed in Two or More First-Degree or Second-Degree Relatives, Regardless of Age

* Lynch syndrome-associated tumors include cancers of colorectum, endometrium, stomach, ovary, pancreas, biliary tract, small bowel, ureter, renal pelvis, and brain tumors, as well as sebaceous gland adenomas and keratoacanthomas. # MSI-H pathological features include tumor infiltrating lymphocytes, Crohn-like lymphocytic reaction, and mucinous or signet-ring cell differentiation, or medullary growth pattern. Abbreviations: CRC, colorectal cancer; MSI, microsatellite instability.

2.2.2. dMMR in Sporadic CRC

In sporadic CRCs, dMMR occurs more frequently in stage II (~20%) and stage III (~12%) tumors, and very rare in metastatic cases (~4%), which indicates that dMMR CRC is less metastatic and MMR status detection in earlier stage is of great importance [26,27]. The vast majority of sporadic CRCs are caused by suppression of MLH1 expression (~95%) due to hypermethylation of the MLH1 promoter known as the CpG island methylator phenotype (CIMP) [28,29], and inactivation of MSH2 and MSH6 account for the small percentage (~5% and ~1%) respectively. About half of sporadic dMMR cases carry BRAF V600E mutations which could distinguish sporadic tumors from LS cases [30,31]. Although sharing many similar features, dMMR sporadic CRCs have different clinical characteristics compared with LS, including older age and female predominance [19].

3. Identification of dMMR/MSI in CRC Tumors

There are two broadly accepted methods for dMMR/MSI detection including MSI testing and MMR protein expression analysis by immunohistochemistry (IHC). It has been shown that MSI testing and IHC are complimentary and the result of MMR proteins expression by IHC is concordant with DNA based MSI testing with a favorable sensitivity and a dramatic specificity [32,33]. IHC is commonly used as an alternative test when a molecular laboratory is not available and is able to pinpoint the affected gene by detecting its protein expression assisting in identifying patients with LS [33].

3.1. MSI Testing

MSI testing is performed by comparing allelic profiles of microsatellite markers in tumor tissue DNA with matching normal DNA from each patient through a PCR-based assay [33]. A panel of five microsatellite sequences, known as the Bethesda panel has been validated and recommended as a reference panel including 2 mononucleotide repeats (BAT26 and BAT25) and 3 dinucleotide repeats (D2S123, D5S346 and D17S250) [34]. According to these microsatellite markers, CRCs can be classified into three groups: MSI-H with two or more of the five microsatellite markers showing instability, MSI-L (low-frequency MSI) with only one of five markers showing instability and MSS (microsatellite stable) with none of the five markers showing instability. Notably, the 2002 National Cancer Institute Workshop made a revision recommending a secondary panel of microsatellite markers with mononucleotide repeats such as BAT-40 and/or MYCL to exclude MSI-L tumors in which only dinucleotide repeats were mutated [25].

3.2. MMR Protein Expression Detection by IHC

Using IHC, tumors exhibiting loss of a MMR protein are considered as dMMR/MSI and those with intact MMR proteins are classified as pMMR/MSS or MSI-L. Absent expression of a MMR protein (MLH1, MSH2, MSH6 or PMS2) can guide a follow-up germline test to find out the affected gene to screen for LS. LS cases can be diagnosed by isolated loss of MSH2 or MSH6 protein. MLH1 and PMS2 proteins are commonly lost concurrently, and so are MSH2 and MSH6 proteins [15]. Isolated loss of MLH1 protein has been described in sporadic CRCs [35]. In addition, CRCs with loss of MLH1 protein expression are always advised to detect BRAF mutations to confirm sporadic cases [36].

4. Predictive Value of MMR Status in CRC Treatment

4.1. Chemotherapeutic Agents

4.1.1. Fluoropyrimidine

A fluoropyrimidine (5-FU or capecitabine)-based adjuvant chemotherapy is considered as standard care for selected stage II and stage III CRC after surgery [37,38]. Clinicians commonly choose intravenous 5-FU or oral prodrug capecitabine or combine them with other chemotherapeutic agents such as irenotecan, leucovorin, and oxaliplatin to increase the response rate (RR) [39]. 5-FU functions by conversion to a series of active metabolites including fluorodeoxyuridine monophosphate (FdUMP), fluorodeoxyuridine triphosphate (FdUTP), and fluorodine triphosphate (FUTP), which kill tumor cells in different mechanisms. FdUMP inhibits thymidylate synthetase (TS), a pivotal element in generating nucleotides required for DNA replication, whilst FdUTP and FUTP incorporate into DNA and RNA respectively [40]. FUTP incorporation has been considered cytotoxic because it disrupts RNA processing while FdUTP lesion in DNA would be lethal by contributing to DNA strand breaks or apoptosis [40].

Although not all preclinical studies agreed [41,42], most supported that dMMR was associated with poor benefit from 5-FU treatment in CRC [43,44,45,46]. Carethers et al. [44] reported that HCT116 (a MLH1 deficient CRC cell line) was approximately 18-fold more resistant to 5-FU compared with pMMR cells. When introduced by chromosome 3 transfer containing the MLH1 gene, HCT116 was sensitive to 5-FU treatment [44]. Meyers et al. [45] obtained similar results in another study both in HCT116 and HEC59 (a MSH2 deficient human endometrial adenocarcinoma cell line). DNA mismatches produced by insertion of 5FdUTP into DNA could not be recognized in dMMR cells, which led to cell survival and drug resistance [47,48,49,50], and when MMR deficiency was corrected, resistance to 5-FU was reversed [49]. A MSH2 deficient xenograft experiment conducted by Pocard et al. also showed that dMMR CRC was resistant to 5-FU [51].

There are many clinical studies about the relationship between MMR and the response of CRC to 5-FU (Table 1). Abundant clinical data suggested that 5-FU-based adjuvant chemotherapy was ineffective in CRC patients with dMMR tumors. A retrospective analysis carried out by Ribic et al. [52] found that pMMR status was significantly correlated with increased overall survival (OS) (hazard ratio (HR), 0.72; 95% confidence interval (CI), 0.53–0.99) among stage II/III patients who received FU-based adjuvant chemotherapy, while dMMR CRC patients experienced no benefit. Another prospective analysis of data from randomized, clinical trials by Sargent et al. [53] demonstrated that no benefit from 5-FU treatment was observed for patients with either stage II (HR = 2.30; 95% CI = 0.85–6.24; p = 0.09) or stage III (HR = 1.01; 95% CI = 0.41–2.51; p = 0.98) dMMR CRCs. However, some studies considered that dMMR CRCs derived a similar or even a greater benefit from 5-FU-based adjuvant treatment compared with pMMR CRCs [54,55,56,57]. Conflicting results were likely due to the bimodal age distribution of CRC patients, limited sample size, inclusion of multiple tumor stages and different 5-FU-based adjuvant regimens etc. [58,59].

Table 1.

Studies evaluating the impact of MMR status on the efficacy of 5-FU-based treatment in CRC.

References Analyzed/Total MSI Frequency Disease Stage Treatment Result
Sargent et al. (2010) [53] 457 15% Stage II & III FU/LEV or FU/LV vs. No Treatment Reduced OS in dMMR Tumors Receiving FU-based Adjuvant Therapy (HR, 2.95; 95% CI, 1.02–8.54; p = 0.04)
Jover et al. (2009) [60] 505/754 10.1% Stage II & III 5-FU-based vs. No Treatment Reduced Survival in dMMR Tumors Receiving FU-based Adjuvant Therapy (pMMR Log Rank p = 0.00001; dMMR Log Rank p = 0.7)
Tejpar et al. (2009) [61] 1254/3278 22% stage II
12% stage III
Stage II & III 5-FU/FO vs. 5-FU/FO/CPT-11 Prognostic Effect of dMMR in Patients Treated with 5-FU
Sargent et al. (2008) [62] 341 13.8% Stage II & III 5-FU/LEV, 5-FU/FO vs. No Treatment Reduced OS (pMMR HR, 0.69; p = 0.047; dMMR HR, 1.26; p = 0.68) and DFS (pMMR HR, 0.59; p = 0.004; dMMR HR, 1.41; p = 0.53) in dMMR Tumors Receiving 5-FU-based Adjuvant Therapy
Kim et al. (2007) [56] 542 18.1% Stage II & III FU/LV vs. No Treatment No Difference was Found by dMMR Status.
Westra et al. (2005) [54] 273/391 16% Stage III FU-based Chemotherapy In a Multivariate Model, dMMR Status was not Associated with DFS.
Ribic et al. (2003) [52] 570 16.7% Stage II & III 5-FU-based Chemotherapy vs. No Treatment Reduced OS (pMMR HR, 0.72; p = 0.04; dMMR HR, 1.07; p = 0.80) in dMMR Tumors Receiving 5-FU-based Adjuvant Therapy
Hemminki et al. (2000) [55] 1044 12% Stage III 5-FU-based Chemotherapy Improved RFS in dMMR Tumors (p = 0.020)
Elsaleh et al. (2000) [57] 656 8.5% Stage III 5-FU-based Chemotherapy Better Survival in dMMR Tumors (p = 0.0007)

Abbreviations: dMMR, deficient mismatch repair; CRC, colorectal cancer; 5-FU, 5-fluorouracil; FO, folinic acid; CPT-11, irinotecan; LV, leucovorin; LEV, levamisole; PFS, progression-free survival; HR, hazard ratio; MSI, microsatellite instability; OS, overall survival; DFS, disease-free survival; RFS, recurrence-free survival; pMMR, proficient mismatch repair.

4.1.2. Platinum Compounds

Platinum compounds such as carboplatin, cisplatin, and oxaliplatin play an important role in chemotherapeutic treatment of many malignant tumors [63]. These platinum-containing drugs kill cancer cells by forming DNA adducts contributing to intrastrand or interstrand cross-links, which change the DNA molecule leading to cell cycle arrest and apoptosis [63].

Unfortunately, intrinsic or acquired drug resistance has limited the usage of platinum compounds in most cancers including CRC. Aebi et al. [42] showed that two dMMR tumor cells, HCT116 and HEC59 were both resistant to cisplatin and carboplatin compared with pMMR subline HCT116 + ch3 cell (complemented with chromosome 3 bearing the wild-type gene for MLH1) and pMMR subline HEC59 + ch2 cell (complemented with chromosome 2 bearing the wild-type gene for MSH2). The mechanism involved was that platinum complexes could interfere with the functional MMR system to stop a complete repair of DNA damage leading to cell apoptosis [64]. When MMR was deficient, cells could proliferate with DNA damage induced by platinating agents and drug resistance would occur. Thus, dMMR is a key determinant in resistance of CRC cells to cisplatin and carboplatin management.

However, it was shown that MMR deficiency seemed not to confer drug resistance to a new platinum compound oxaliplatin. A pooled analysis of the NSABP-C07 and NSABP-C08 trials demonstrated that the benefit of adding oxaliplatin to 5-FU was not associated with MMR status [65]. Meanwhile, several retrospective studies reported that adding oxaliplatin to 5-FU could restore the efficacy of adjuvant chemotherapy in stage III dMMR CRCs [38,66,67]. A retrospective study carried out by Zaanan et al. showed that addition of oxaliplatin to 5-fluorouracil and leucovorin (FL) significantly improved disease-free survival (DFS) of stage III CRC patients with dMMR tumors (HR 0.17; 95% CI = 0.04–0.68; p = 0.01) [66]. Flejou et al. [68] were also in favor of FOLFOX4 (a fluorouracil, leucovorin, and oxaliplatin regimen) vs. LV5FU2 (a fluorouracil and leucovorin regimen) in dMMR CRC patients. The reason why dMMR CRC cells were sensitive to oxaliplatin was that the special 1, 2-diaminocyclohexane (DACH) ligand of oxaliplatin prevented MMR complex from binding to its DNA adducts, which led to failure of repair and subsequent apoptosis of tumor cells [69]. Thus oxaliplatin had efficacy in CRC cells resistant to cisplatin and carboplatin [42,69,70].

4.1.3. Topoisomerase I Inhibitors

Camptothecin (CPT) and its derivative irinotecan (CPT-11) are topoisomerase I inhibitors that induce transient DNA single-strand breaks which are subsequently converted into permanent DNA double strand breaks (DSB) and ultimately cause cell apoptosis. CPT-11 has been commonly applied in the treatment of metastatic CRC (mCRC) as an effective complement for 5-FU.

When it comes to the relationship between MMR status and the response of CRC to CPT-11, there has been a long-term controversy. There appeared to be strong preclinical evidence indicating that dMMR CRC tumors were more sensitive to CPT-11 treatment than pMMR tumors [71,72]. Magrini et al. [73] suggested the percentage of apoptotic cells after treatment with CPT-11 was higher in dMMR CRC cells than that in pMMR cells, suggesting that an intact MMR system might prevent CPT-11-induced apoptosis. A recent research showed that dMMR CRC cells were more sensitive to CPT-11 and MMR status might be a predictive biomarker of response to CPT-11-based chemotherapy in mCRC [74]. The reasons below might explain the fact that dMMR CRC tumors were more sensitive to CPT-11 treatment: Firstly, when the MMR system was defected, the DSB induced by CPT-11 could not be repaired and the apoptosis of tumor cells occurred [74]. Secondly, dMMR CRC cells commonly generated secondary mutations in DSB repair genes such as MRE11A and hRAD50, which might have improved the efficacy of CPT-11 [75]. A few studies showed that dMMR CRC cells were resistant to camptothecin derivatives [76,77], which might have resulted from different detection methods such as the clonogenic assay and MTT test.

As shown in Table 2, the clinical results about the relationship between MMR status and CPT-11 were quite inconclusive. Some studies found that dMMR was associated with better clinical outcome to CPT-11 management in mCRC patients. A retrospective study of 72 mCRC patients conducted by Fallik et al. [78] found that dMMR tumors experienced improved RR to CPT-11 compared with pMMR tumors (57.1% vs. 10.8%). Charara et al. [79] demonstrated that dMMR was predictive of an improved response to neoadjuvant chemotherapy containing CPT-11 and radiation therapy in early stage rectal cancer. Similarly, Bertagnolli et al. [80] showed that CRC patients with dMMR tumors had improved 5-year DFS as compared with pMMR tumors when treated with irinotecan, FU, and leucovorin (IFL). However, there were clinical data indicating that MMR status was not associated with the response to CPT-11 in CRC treatment [6,81,82]. These varied conclusions might be due to the fact that most studies were small-scale, retrospective, or nonrandomized, with a significant bias as well as the different CPT-11-based regimens involved in the trials [6]. For instance, CPT-11 was often used in combination with 5-FU, to which dMMR cells were resistant. Hence, further studies are definitely needed to confirm the efficacy of CPT-11 in dMMR tumors to guide its individual administration in CRC subsets.

Table 2.

Studies assessing the impact of MMR status on the efficacy of CPT-11 in CRC therapy.

References Analyzed/Total MSI Frequency Disease Stage Treatment Result
Tejpar et al. (2015) [6] 1254/3278 21.8% in Stage II
12.1% in Stage III
Stage II/III 5-FU/LV vs. 5-FU/LV/CPT-11 No Difference was Found by dMMR Status
Kim et al. (2011) [81] 197/297 11.7% mCRC CPT-11-based Chemotherapy No Difference in RR and PFS
Bertagnolli et al. (2009) [80] 723/1264 13.3% Stage III FU/LV vs. Weekly IFL Improved Survival in dMMR Patients (p = 0.03)
Braun et al. (2008) [82] 931/2135 4.4% mCRC Palliative 1st-line 5-FU/CPT-11 or 5-FU/oxaliplatin No Difference in PFS (HR, 0.93; p = 0.7) and OS (HR, 0.66; p = 0.2)
Charara et al. (2004) [79] 57 23% Early Stage Rectal Cancer 5-FU, CPT-11, Radiotherapy and Surgery Improved Complete RR in dMMR patients
Fallik et al. (2003) [78] 44/72 15.9% mCRC CPT-11 Improved RR in dMMR Patients (57% vs. 10.8%; p = 0.009)

Abbreviations: mCRC, metastatic colorectal cancer; RR, response rate; IFL, irinotecan, FU and LV.

4.1.4. Alkylating Agents

Alkylating agents including N-Methyl-N-nitrosurea (MNU), N-methyl-N’-nitro-N-nitrosoguanidine (MNNG), procarbazine, and temozolomide (an activated form of procarbazine) induce various adducts on DNA, notably O6-methylguanine [83]. O6-methylguanine DNA methyltransferase (MGMT) possesses the ability to remove the carcinogenic O6-methylguanine (O6-MeG) DNA adducts and has been demonstrated to serve as a biomarker to define the likely benefit from alkylating agents [84]. Some in vitro experiments highlighted that the MMR system was a pivotal determinant in the response of CRC cells to alkylating agents. The dMMR CRC cell line HCT116 was found to be tolerant to MNNG and the resistance phenomenon could be reversed when the MMR defect was corrected [85]. A similar result was shown in another CRC cell LOVO, which contained a mutated MSH2 gene from exon 3 to exon 8 [86]. The underlying mechanism was probably that a competent MMR system was necessary for G2 arrest after alkylating agent treatment, and dMMR tumor cells failed to undergo this arrest [87]. Also, MMR proteins could recognize lethal DNA adducts which were neglected by MGMT, and then initiated an apoptotic progress contributing to cell death [88]. In conclusion, sensitivity to alkylating agents likely requires taking both the MGMT expression and the functional MMR system into consideration.

To date, little clinical trial data describing the role of dMMR in response to alkylating agents in CRC has been reported. A phase II study of temozolomide found that all 5 pMMR CRC patients were with partial responses (PR) and 83% dMMR CRC patients had progressive disease, suggesting that a proficient MMR system seemed to be required for the response to temozolomide, but the overall RR (6%) was too low to permit this conclusion [89].

4.2. Targeted Therapy

4.2.1. Anti-EGFR Targeted Therapy

EGFR is a receptor tyrosine kinase involved in the development and metastasis of CRC. Monoclonal antibodies (mAbs) targeting EGFR including cetuximab and panitumumab are used alone or in combination with chemotherapy in mCRC treatment. KRAS or BRAF mutations in mCRCs have been identified as well validated markers of a poor response to EGFR-targeted antibodies [90,91,92]. Previous works demonstrated that BRAF mutations were significantly associated with dMMR CRCs especially sporadic dMMR cases. Wang et al. [93] found that 34% dMMR CRCs had BRAF mutations and most occurred in those with promoter hypermethylation of MLH1, whereas only 12% pMMR CRC displayed BRAF mutations. Tran et al. [94] also showed that BRAF mutations were significantly more common in dMMR CRC tumors. Therefore, whether dMMR mCRC could benefit more from anti-EGFR targeted therapy deserves further exploration.

4.2.2. Anti-VEGF Targeted Therapy

VEGF, a potent regulator of physiologic and pathologic angiogenesis plays an essential role in tumor progression and metastasis. VEGF-A has been proven not only to suppress the maturation of dendritic cells (DCs) [95] and T cells [96] but also to alter the function of dendritic cells potently generating a tumor-associated immune suppression [97]. Moreover, VEGF-A could directly induce regulatory T cells (Tregs) proliferation in tumor-bearing mice through VEGF-A/VEGFR pathway [98], and specific blockade of this pathway could prevent the accumulation of Tregs in tumor-bearing mice and mCRC patients [98].

Bevacizumab in combination with chemotherapy exhibited attractive clinical activity in mCRC patients [99]. A retrospective analysis by Pogue-Geile et al. [100] revealed that stage II/III dMMR CRC patients significantly derived potential survival benefit from the addition of bevacizumab to standard FOLFOX (HR = 0.52; 95% CI = 0.29–0.94) in contrast to pMMR patients (HR = 1.03; 95% CI = 0.84–1.27). Hansen et al. [101] suggested that CRC patients with dMMR tumors had higher serum VEGF-A levels than those with pMMR tumors. Additionally, adding bevacizumab to adjuvant chemotherapy induced a decrease in Treg percentages but not conventional T cells in the peripheral blood of mCRCs [101]. Thus dMMR CRC patients might get significant clinical benefit from bevacizumab owing to their immunosuppressive microenvironment and high serum levels of VEGF-A. These findings seem to pave the road to individualized VEGF targeted therapies in mCRC, but more investigations are needed to confirm them.

4.3. Immune Checkpoint Inhibitor Therapy: Monoclonal Antibodies Inhibiting PD-1

dMMR CRC is hypermutated and expresses numerous neoantigens (frameshift peptides) which induce an active immune microenvironment characterized by abundant tumor infiltrating lymphocytes (TILs) [102]. dMMR tumors are able to evade immune destruction of the vigorous immune system owing to elevated expression of multiple checkpoint proteins including the immune cell co-receptor PD-1, programmed cell death-ligand 1 (PD-L1) and programmed cell death-ligand 2 (PD-L2), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), lymphocyte-activation gene 3 (LAG-3) and indoleamine (2,3)-dioxygenase (IDO) [103]. Recently, anti-PD-1 (nivolumab and pembrolizumab) antibodies and anti-PD-L1 (MPDL3280A, Medi4736, and BMS-936559) antibodies, namely immune checkpoint inhibitors (ICIs) have been developed to restore T cell activity and are implemented in the treatment of various human malignancies (e.g., melanoma cancer, renal cancer, and non-small cell lung cancer) [104,105,106].

There were clinical data demonstrating that dMMR status could predict the clinical benefit of anti-PD-1 therapy in mCRC patients and pMMR mCRC patients might not receive anti-PD-1 therapy in clinic due to the complete absence of objective responses. The anti-PD-1 antibody nivolumab did not demonstrate clinically significant activity in a phase I study of mCRC patients [106]. Only one patient with a dMMR tumor from this cohort, who had a PD-L1 positive tumor displayed complete response after treatment with five doses of nivolumab for six months [107]. A phase II study conducted by Le et al. [108] demonstrated that dMMR CRC patients displayed the immune-related objective response and immune-related progression-free survival (PFS) rates 40% and 78% respectively, compared with pMMR CRC patients 0% and 11%. Thus, the Food and Drug Administration (FDA) approved rapidly pembrolizumab for metastatic/refractory dMMR CRC treatment. dMMR CRC could benefit from immune checkpoint blockade therapy due to high expression levels of checkpoint proteins in the local immune microenvironment [108] and the strong T cell response induced by large amounts of neoantigens [109]. However, only 3%–6% of mCRCs are dMMR phenotype, which indicates that the targeted population in mCRC is very small [110]. Thus the anti-PD-1 therapy might be applied to earlier stage CRC patients to expand its application range [110].

5. Prognostic Value of MMR Status in CRC

5.1. Prognostic Value of MMR Status in Early Stage CRC

Rich evidence indicated that stage II/III CRC patients with dMMR tumors had a better clinical outcome than those with pMMR tumors [52,111,112]. Bertagnolli et al. [112] reported in their study that CRC patients with dMMR tumors had better 5-year DFS (0.76 vs. 0.67; p < 0.001) and OS (0.81 vs. 0.78; p = 0.029) than those with pMMR tumors. It was shown that the prognostic effect of dMMR was stronger in stage II than stage III CRC patients [6,7]. In a study by Klingbiel et al. [6] they found that in stage II, relapse-free survival (RFS) and OS were better for CRC patients with pMMR than with dMMR tumors (HR = 0.26; 95% CI = 0.10–0.65; p = 0.004 and 0.16; 95% CI = 0.04–0.64; p = 0.01). In stage III, RFS was slightly better for dMMR CRC patients (HR = 0.67; 95% CI = 0.46–0.99; p = 0.04) [6]. And the better prognosis of dMMR CRC patients might result from a stronger immunologic response driven by abundant TILs in the tumor microenvironment [113]. In addition, several studies confirmed that dMMR CRC had reduced levels of VEGF compared to pMMR CRC [114,115], which might partly explain that patients with dMMR tumors had more favorable prognosis.

5.2. Prognostic Value of MMR Status in mCRC

The very small fraction of dMMR tumors in mCRC patients brought an obstacle to evaluate the prognostic value of dMMR status in mCRC [116]. It was well reported that dMMR was a good prognostic marker in early stage CRC, and several researches showed that dMMR displayed little prognostic value in mCRC. Recently, a nationwide cohort study of 6692 patients suggested that MMR status was not related to survival in mCRC patients [117]. Similarly, Nöpel-Dünnebacke et al. [118] found that dMMR status was not correlated with overall response rate (ORR), PFS and OS in mCRC. Overman et al. [110] found that mCRC patients with dMMR tumors had no improved outcomes and BRAF V600E mutation was associated with a poor prognosis in dMMR mCRC. A pooled analysis of four phase III studies demonstrated that compared with those with pMMR tumors, CRC patients with dMMR tumors displayed reduced PFS and OS (HR = 1.33; 95% CI = 1.12–1.57 and HR = 1.35; 95% CI = 1.13–1.61, respectively) which might be driven by BRAF V600E mutations [119]. Another study showed that dMMR CRC tumors had significantly poorer survival compared with pMMR CRC tumors (11.1 months vs. 22.1 months, p = 0 .017) [94].

6. Conclusions and Perspectives

Though the TNM stage remains the key determinant of CRC prognosis and treatment in clinic, there are considerable stage independent inter-individual differences in clinical outcome and therapy response of CRC patients. Hence, prognostic and predictive biomarkers are urgently demanded to accurately inform clinical outcome and guide treatment selection in CRC management. Improved knowledge of the molecular characterization of CRC has allowed the personalized management of this malignancy to advance rapidly. As one of the important molecular characterizations of CRC, dMMR status has been demonstrated as a crucial biomarker for prognosis and response to many drugs used in CRC, which helps the clinicians and patients use medications rationally to avoid dispensable treatment and reduce the burden of patients in CRC therapy.

The paired MMR deficient/proficient models or the panel of dMMR versus pMMR cell lines were always chosen in preclinical studies. The former models might not take the effects of secondary mutations into account whereas the latter would. In addition, different detection methods for cells apoptosis induced by drugs, such as the clonogenic assay and MTT test were applied in preclinical experiments. All the above factors would lead to inconsistent preclinical results concerning the relationship between the MMR system and the sensitivity to drugs used in CRC treatment. Meanwhile, clinical studies determining the role of MMR status in CRC management were different in scale, whether retrospective or prospective, randomized or not, and different chemotherapy regimens were involved, which might have led to the varied conclusions. Thus, studies in pooled data from similar clinical trials may help to further explore tumor heterogeneity and to highlight the impact of MMR status and other molecular features on prognosis and the efficacy of drugs used to treat CRC. Although dMMR has been shown to provide valuable predictive and prognostic information, it is more important to combine it with other molecular markers such as KRAS, BRAF V600E mutations [120] or gene expression profiling by next-generation sequencing (NGS) platforms to precisely guide the individualized treatment of CRC [3].

In conclusion, dMMR is a crucial molecular biomarker and plays an important role in CRC management to guide decision-making in clinic. A better understanding of molecular pathways involved in dMMR CRC is pivotal to develop novel therapies and inform suitable therapies of dMMR CRC subsets. With the development of some key scientific discoveries in the molecular biology of CRC, personalized drug therapy of CRC is an undoubted tendency. Although we are a long way from personalized treatment of CRC, we are very optimistic regarding the direction of this field and expect revolutionary progress in the future.

Acknowledgments

This work was supported by China Scholarship Council, the National Natural Science Foundation of China (No. 30901834), NCET-11-0509, Hunan Provincial Natural Science Foundation of China (No. 12K005) and Research Funds of teachers for the Central South University.

Author Contributions

Cong-Min Zhang conceived the review, generated the first draft, and overall, led the writing of the manuscript and the manuscript was supervised and finalized by Lan Fan. Jin-Feng Lv, Liang Gong and Lin-Yu Yu read and edited the whole paper. Xiao-Ping Chen and Hong-Hao Zhou provided a critical revision of the manuscript. All the authors read, and approved the final version to be published.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  • 1.Siegel R.L., Miller K.D., Jemal A. Cancer statistics, 2015. CA Cancer J. Clin. 2015;65:5–29. doi: 10.3322/caac.21254. [DOI] [PubMed] [Google Scholar]
  • 2.Kawakami H., Zaanan A., Sinicrope F.A. Microsatellite instability testing and its role in the management of colorectal cancer. Curr. Treat. Opt. Oncol. 2015;16:30. doi: 10.1007/s11864-015-0348-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Sinicrope F.A., Okamoto K., Kasi P.M., Kawakami H. Molecular biomarkers in the personalized treatment of colorectal cancer. Clin. Gastroenterol. Hepatol. 2016;14:651–658. doi: 10.1016/j.cgh.2016.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Sinicrope F.A., Sargent D.J. Molecular pathways: Microsatellite instability in colorectal cancer: Prognostic, predictive, and therapeutic implications. Am. J. Clin. Cancer Res. 2012;18:1506–1512. doi: 10.1158/1078-0432.CCR-11-1469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Wong A., Ma B.B.Y. Personalizing therapy for colorectal cancer. Clin. Gastroenterol. Hepatol. 2014;12:139–144. doi: 10.1016/j.cgh.2013.08.040. [DOI] [PubMed] [Google Scholar]
  • 6.Klingbiel D., Saridaki Z., Roth A.D., Bosman F.T., Delorenzi M., Tejpar S. Prognosis of stage II and III colon cancer treated with adjuvant 5-fluorouracil or FOLFIRI in relation to microsatellite status: Results of the PETACC-3 trial. Ann. Oncol. 2015;26:126–132. doi: 10.1093/annonc/mdu499. [DOI] [PubMed] [Google Scholar]
  • 7.Roth A.D., Delorenzi M., Tejpar S., Yan P., Klingbiel D., Fiocca R., d’Ario G., Cisar L., Labianca R., Cunningham D., et al. Integrated analysis of molecular and clinical prognostic factors in stage II/III colon cancer. J. Natl. Cancer Inst. 2012;104:1635–1646. doi: 10.1093/jnci/djs427. [DOI] [PubMed] [Google Scholar]
  • 8.Gatalica Z., Vranic S., Xiu J., Swensen J., Reddy S. High microsatellite instability (MSI-H) colorectal carcinoma: A brief review of predictive biomarkers in the era of personalized medicine. Fam. Cancer. 2016;15:405–412. doi: 10.1007/s10689-016-9884-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hewish M., Lord C.J., Martin S.A., Cunningham D., Ashworth A. Mismatch repair deficient colorectal cancer in the era of personalized treatment. Nat. Rev. Clin. Oncol. 2010;7:197–208. doi: 10.1038/nrclinonc.2010.18. [DOI] [PubMed] [Google Scholar]
  • 10.Silva F.C., Valentin M.D., Ferreira Fde O., Carraro D.M., Rossi B.M. Mismatch repair genes in Lynch syndrome: A review. Sao Paulo Med. J. 2009;127:46–51. doi: 10.1590/S1516-31802009000100010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Hsieh P., Yamane K. DNA mismatch repair: Molecular mechanism, cancer, and ageing. Mech. Ageing Dev. 2008;129:391–407. doi: 10.1016/j.mad.2008.02.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Li G.M. Mechanisms and functions of DNA mismatch repair. Cell Res. 2008;18:85–98. doi: 10.1038/cr.2007.115. [DOI] [PubMed] [Google Scholar]
  • 13.Imai K., Yamamoto H. Carcinogenesis and microsatellite instability: The interrelationship between genetics and epigenetics. Carcinogenesis. 2008;29:673–680. doi: 10.1093/carcin/bgm228. [DOI] [PubMed] [Google Scholar]
  • 14.Zhang L. Immunohistochemistry versus microsatellite instability testing for screening colorectal cancer patients at risk for hereditary nonpolyposis colorectal cancer syndrome. Part II. The utility of microsatellite instability testing. J. Mol. Diagn. 2008;10:301–307. doi: 10.2353/jmoldx.2008.080062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Yuan L., Chi Y., Chen W., Chen X., Wei P., Sheng W., Zhou X., Shi D. Immunohistochemistry and microsatellite instability analysis in molecular subtyping of colorectal carcinoma based on mismatch repair competency. Int. J. Clin. Exp. Med. 2015;8:20988–21000. [PMC free article] [PubMed] [Google Scholar]
  • 16.Zlobec I., Bihl M.P., Foerster A., Rufle A., Lugli A. The impact of CpG island methylator phenotype and microsatellite instability on tumour budding in colorectal cancer. Histopathology. 2012;61:777–787. doi: 10.1111/j.1365-2559.2012.04273.x. [DOI] [PubMed] [Google Scholar]
  • 17.Greenson J.K., Huang S.C., Herron C., Moreno V., Bonner J.D., Tomsho L.P., Ben-Izhak O., Cohen H.I., Trougouboff P., Bejhar J., et al. Pathologic predictors of microsatellite instability in colorectal cancer. Am. J. Surg. Pathol. 2009;33:126–133. doi: 10.1097/PAS.0b013e31817ec2b1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Lynch H.T., Lynch J.F., Lynch P.M., Attard T. Hereditary colorectal cancer syndromes: Molecular genetics, genetic counseling, diagnosis and management. Fam. Cancer. 2008;7:27–39. doi: 10.1007/s10689-007-9165-5. [DOI] [PubMed] [Google Scholar]
  • 19.Gatalica Z., Torlakovic E. Pathology of the hereditary colorectal carcinoma. Fam. Cancer. 2008;7:15–26. doi: 10.1007/s10689-007-9146-8. [DOI] [PubMed] [Google Scholar]
  • 20.Boland C.R., Goel A. Microsatellite instability in colorectal cancer. Gastroenterology. 2010;138:e2073–e2087. doi: 10.1053/j.gastro.2009.12.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Ligtenberg M.J., Kuiper R.P., Chan T.L., Goossens M., Hebeda K.M., Voorendt M., Lee T.Y., Bodmer D., Hoenselaar E., Hendriks-Cornelissen S.J., et al. Heritable somatic methylation and inactivation of MSH2 in families with Lynch syndrome due to deletion of the 3’ exons of TACSTD1. Nat. Genet. 2009;41:112–117. doi: 10.1038/ng.283. [DOI] [PubMed] [Google Scholar]
  • 22.Setaffy L., Langner C. Microsatellite instability in colorectal cancer: Clinicopathological significance. Pol. J. Pathol. 2015;66:203–218. doi: 10.5114/pjp.2015.54953. [DOI] [PubMed] [Google Scholar]
  • 23.Tiwari A.K., Roy H.K., Lynch H.T. Lynch syndrome in the 21st century: Clinical perspectives. Qjm. 2016;109:151–158. doi: 10.1093/qjmed/hcv137. [DOI] [PubMed] [Google Scholar]
  • 24.Giardiello F.M., Allen J.I., Axilbund J.E., Boland C.R., Burke C.A., Burt R.W., Church J.M., Dominitz J.A., Johnson D.A., Kaltenbach T., et al. Guidelines on genetic evaluation and management of Lynch syndrome: A consensus statement by the US multi-society task force on colorectal cancer. Gastroenterology. 2014;147:502–526. doi: 10.1053/j.gastro.2014.04.001. [DOI] [PubMed] [Google Scholar]
  • 25.Umar A., Boland C.R., Terdiman J.P., Syngal S., de la Chapelle A., Ruschoff J., Fishel R., Lindor N.M., Burgart L.J., Hamelin R., et al. Revised bethesda guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability. J. Natl. Cancer Inst. 2004;96:261–268. doi: 10.1093/jnci/djh034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.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. doi: 10.1200/JCO.2009.23.3452. [DOI] [PubMed] [Google Scholar]
  • 27.Koopman M., Kortman G.A., Mekenkamp L., Ligtenberg M.J., Hoogerbrugge N., Antonini N.F., Punt C.J., van Krieken J.H. Deficient mismatch repair system in patients with sporadic advanced colorectal cancer. Br. J. Cancer. 2009;100:266–273. doi: 10.1038/sj.bjc.6604867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ogino S., Goel A. Molecular classification and correlates in colorectal cancer. J. Mol. Diagn. 2008;10:13–27. doi: 10.2353/jmoldx.2008.070082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Devaud N., Gallinger S. Chemotherapy of MMR-deficient colorectal cancer. Fam. Cancer. 2013;12:301–306. doi: 10.1007/s10689-013-9633-z. [DOI] [PubMed] [Google Scholar]
  • 30.Tol J., Nagtegaal I.D., Punt C.J. Braf mutation in metastatic colorectal cancer. N. Engl. J. Med. 2009;361:98–99. doi: 10.1056/NEJMc0904160. [DOI] [PubMed] [Google Scholar]
  • 31.Domingo E., Niessen R.C., Oliveira C., Alhopuro P., Moutinho C., Espin E., Armengol M., Sijmons R.H., Kleibeuker J.H., Seruca R., et al. BRAF-V600E is not involved in the colorectal tumorigenesis of HNPCC in patients with functional MLH1 and MSH2 genes. Oncogene. 2005;24:3995–3998. doi: 10.1038/sj.onc.1208569. [DOI] [PubMed] [Google Scholar]
  • 32.Shia J. Immunohistochemistry versus microsatellite instability testing for screening colorectal cancer patients at risk for hereditary nonpolyposis colorectal cancer syndrome. Part I. The utility of immunohistochemistry. J. Mol. Diagn. 2008;10:293–300. doi: 10.2353/jmoldx.2008.080031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Lindor N.M., Burgart L.J., Leontovich O., Goldberg R.M., Cunningham J.M., Sargent D.J., Walsh-Vockley C., Petersen G.M., Walsh M.D., Leggett B.A., et al. Immunohistochemistry versus microsatellite instability testing in phenotyping colorectal tumors. J. Clin. Oncol. 2002;20:1043–1048. doi: 10.1200/JCO.20.4.1043. [DOI] [PubMed] [Google Scholar]
  • 34.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. [PubMed] [Google Scholar]
  • 35.Herman J.G., Umar A., Polyak K., Graff J.R., Ahuja N., Issa J.P., Markowitz S., Willson J.K., Hamilton S.R., Kinzler K.W., et al. Incidence and functional consequences of hMLH1 promoter hypermethylation in colorectal carcinoma. Proc. Natl. Acad. Sci. USA. 1998;95:6870–6875. doi: 10.1073/pnas.95.12.6870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ladabaum U., Wang G., Terdiman J., Blanco A., Kuppermann M., Boland C.R., Ford J., Elkin E., Phillips K.A. Strategies to identify the Lynch syndrome among patients with colorectal cancer: A cost-effectiveness analysis. Ann. Intern. Med. 2011;155:69–79. doi: 10.7326/0003-4819-155-2-201107190-00002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Andre T., Boni C., Navarro M., Tabernero J., Hickish T., Topham C., Bonetti A., Clingan P., Bridgewater J., Rivera F., et al. Improved overall survival with oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment in stage II or III colon cancer in the mosaic trial. J. Clin. Oncol. 2009;27:3109–3116. doi: 10.1200/JCO.2008.20.6771. [DOI] [PubMed] [Google Scholar]
  • 38.Andre T., Boni C., Mounedji-Boudiaf L., Navarro M., Tabernero J., Hickish T., Topham C., Zaninelli M., Clingan P., Bridgewater J., et al. Oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment for colon cancer. N. Engl. J. Med. 2004;350:2343–2351. doi: 10.1056/NEJMoa032709. [DOI] [PubMed] [Google Scholar]
  • 39.Cascinu S., Georgoulias V., Kerr D., Maughan T., Labianca R., Ychou M. Colorectal cancer in the adjuvant setting: Perspectives on treatment and the role of prognostic factors. Ann. Oncol. 2003;14(Suppl. S2):ii25–ii29. doi: 10.1093/annonc/mdg725. [DOI] [PubMed] [Google Scholar]
  • 40.Parker W.B., Cheng Y.C. Metabolism and mechanism of action of 5-fluorouracil. Pharmacol. Ther. 1990;48:381–395. doi: 10.1016/0163-7258(90)90056-8. [DOI] [PubMed] [Google Scholar]
  • 41.Chen X.X., Lai M.D., Zhang Y.L., Huang Q. Less cytotoxicity to combination therapy of 5-fluorouracil and cisplatin than 5-fluorouracil alone in human colon cancer cell lines. World J. Gastroenterol. 2002;8:841–846. doi: 10.3748/wjg.v8.i5.841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Aebi S., Fink D., Gordon R., Kim H.K., Zheng H., Fink J.L., Howell S.B. Resistance to cytotoxic drugs in DNA mismatch repair-deficient cells. Clin. Cancer Res. 1997;3:1763–1767. [PubMed] [Google Scholar]
  • 43.Bras-Goncalves R.A., Pocard M., Formento J.L., Poirson-Bichat F., De Pinieux G., Pandrea I., Arvelo F., Ronco G., Villa P., Coquelle A., et al. Synergistic efficacy of 3n-butyrate and 5-fluorouracil in human colorectal cancer xenografts via modulation of DNA synthesis. Gastroenterology. 2001;120:874–888. doi: 10.1053/gast.2001.22440. [DOI] [PubMed] [Google Scholar]
  • 44.Carethers J.M., Chauhan D.P., Fink D., Nebel S., Bresalier R.S., Howell S.B., Boland C.R. Mismatch repair proficiency and in vitro response to 5-fluorouracil. Gastroenterology. 1999;117:123–131. doi: 10.1016/S0016-5085(99)70558-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Meyers M., Wagner M.W., Hwang H.S., Kinsella T.J., Boothman D.A. Role of the hMLH1 DNA mismatch repair protein in fluoropyrimidine-mediated cell death and cell cycle responses. Cancer Res. 2001;61:5193–5201. [PubMed] [Google Scholar]
  • 46.Tokunaga E., Oda S., Fukushima M., Maehara Y., Sugimachi K. Differential growth inhibition by 5-fluorouracil in human colorectal carcinoma cell lines. Eur. J. Cancer. 2000;36:1998–2006. doi: 10.1016/S0959-8049(00)00200-8. [DOI] [PubMed] [Google Scholar]
  • 47.Karran P. Mechanisms of tolerance to DNA damaging therapeutic drugs. Carcinogenesis. 2001;22:1931–1937. doi: 10.1093/carcin/22.12.1931. [DOI] [PubMed] [Google Scholar]
  • 48.Jiricny J. The multifaceted mismatch-repair system. Nat. Rev. Mol. Cell Biol. 2006;7:335–346. doi: 10.1038/nrm1907. [DOI] [PubMed] [Google Scholar]
  • 49.Arnold C.N., Goel A., Boland C.R. Role of hMLH1 promoter hypermethylation in drug resistance to 5-fluorouracil in colorectal cancer cell lines. Int. J. Cancer. 2003;106:66–73. doi: 10.1002/ijc.11176. [DOI] [PubMed] [Google Scholar]
  • 50.Tajima A., Hess M.T., Cabrera B.L., Kolodner R.D., Carethers J.M. The mismatch repair complex hMutS alpha recognizes 5-fluorouracil-modified DNA: Implications for chemosensitivity and resistance. Gastroenterology. 2004;127:1678–1684. doi: 10.1053/j.gastro.2004.10.001. [DOI] [PubMed] [Google Scholar]
  • 51.Pocard M., Bras-Goncalves R., Hamelin R., Northover J., Poupon M.F. Response to 5-fluorouracil of orthotopically xenografted human colon cancers with a microsatellite instability: Influence of P53 status. Anticancer Res. 2000;20:85–90. [PubMed] [Google Scholar]
  • 52.Ribic C.M., Sargent D.J., Moore M.J., Thibodeau S.N., French A.J., Goldberg R.M., Hamilton S.R., Laurent-Puig P., Gryfe R., Shepherd L.E., et al. Tumor microsatellite-instability status as a predictor of benefit from fluorouracil-based adjuvant chemotherapy for colon cancer. N. Engl. J. Med. 2003;349:247–257. doi: 10.1056/NEJMoa022289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Sargent D.J., Marsoni S., Monges G., Thibodeau S.N., Labianca R., Hamilton S.R., French A.J., Kabat B., Foster N.R., Torri V., et al. Defective mismatch repair as a predictive marker for lack of efficacy of fluorouracil-based adjuvant therapy in colon cancer. J. Clin. Oncol. 2010;28:3219–3226. doi: 10.1200/JCO.2009.27.1825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Westra J.L., Schaapveld M., Hollema H., de Boer J.P., Kraak M.M.J., de Jong D., ter Elst A., Mulder N.H., Buys C.H.C.M., Hofstra R.M.W., et al. Determination of TP53 mutation is more relevant than microsatellite instability status for the prediction of disease-free survival in adjuvant-treated stage III colon cancer patients. J. Clin. Oncol. 2005;23:5635–5643. doi: 10.1200/JCO.2005.04.096. [DOI] [PubMed] [Google Scholar]
  • 55.Hemminki A., Mecklin J.P., Jarvinen H., Aaltonen L.A., Joensuu H. Microsatellite instability is a favorable prognostic indicator in patients with colorectal cancer receiving chemotherapy. Gastroenterology. 2000;119:921–928. doi: 10.1053/gast.2000.18161. [DOI] [PubMed] [Google Scholar]
  • 56.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. doi: 10.1200/JCO.2006.05.8172. [DOI] [PubMed] [Google Scholar]
  • 57.Elsaleh H., Joseph D., Grieu F., Zeps N., Spry N., Iacopetta B. Association of tumour site and sex with survival benefit from adjuvant chemotherapy in colorectal cancer. Lancet. 2000;355:1745–1750. doi: 10.1016/S0140-6736(00)02261-3. [DOI] [PubMed] [Google Scholar]
  • 58.Kawakami H., Zaanan A., Sinicrope F.A. Implications of mismatch repair-deficient status on management of early stage colorectal cancer. J. Gastrointest Oncol. 2015;6:676–684. doi: 10.3978/j.issn.2078-6891.2015.065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Zaanan A., Meunier K., Sangar F., Flejou J.F., Praz F. Microsatellite instability in colorectal cancer: From molecular oncogenic mechanisms to clinical implications. Cell Oncol. 2011;34:155–176. doi: 10.1007/s13402-011-0024-x. [DOI] [PubMed] [Google Scholar]
  • 60.Jover R., Zapater P., Castells A., Llor X., Andreu M., Cubiella J., Balaguer F., Sempere L., Xicola R.M., Bujanda L., et al. The efficacy of adjuvant chemotherapy with 5-fluorouracil in colorectal cancer depends on the mismatch repair status. Eur. J. Cancer. 2009;45:365–373. doi: 10.1016/j.ejca.2008.07.016. [DOI] [PubMed] [Google Scholar]
  • 61.Tejpar S., Bosman F., Delorenzi M., Fiocca R., Yan P., Klingbiel D., Dietrich D., Van Cutsem E., Labianca R., Roth A. Microsatellite instability (MSI) in stage II and III colon cancer treated with 5FU-LV or 5FU-LV and irinotecan (PETACC 3-EORTC 40993-SAKK 60/00 trial) J. Clin. Oncol. 2009;27 [Google Scholar]
  • 62.Sargent D.J., Marsoni S., Thibodeau S.N., Labianca R., Hamilton S.R., Torri V., Monges G., Ribic C., Grothey A., Gallinger S. Confirmation of deficient mismatch repair (dMMR) as a predictive marker for lack of benefit from 5-FU based chemotherapy in stage II and III colon cancer (CC): A pooled molecular reanalysis of randomized chemotherapy trials. J. Clin. Oncol. 2008;26 [Google Scholar]
  • 63.Rabik C.A., Dolan M.E. Molecular mechanisms of resistance and toxicity associated with platinating agents. Cancer Treat. Rev. 2007;33:9–23. doi: 10.1016/j.ctrv.2006.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Martin L.P., Hamilton T.C., Schilder R.J. Platinum resistance: The role of DNA repair pathways. Clin. Cancer Res. 2008;14:1291–1295. doi: 10.1158/1078-0432.CCR-07-2238. [DOI] [PubMed] [Google Scholar]
  • 65.Gavin P.G., Colangelo L.H., Fumagalli D., Tanaka N., Remillard M.Y., Yothers G., Kim C., Taniyama Y., Kim S.I., Choi H.J., et al. Mutation profiling and microsatellite instability in stage II and III colon cancer: An assessment of their prognostic and oxaliplatin predictive value. Clin. Cancer Res. 2012;18:6531–6541. doi: 10.1158/1078-0432.CCR-12-0605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Zaanan A., Cuilliere-Dartigues P., Guilloux A., Parc Y., Louvet C., de Gramont A., Tiret E., Dumont S., Gayet B., Validire P., et al. Impact of P53 expression and microsatellite instability on stage iii colon cancer disease-free survival in patients treated by 5-fluorouracil and leucovorin with or without oxaliplatin. Ann. Oncol. 2010;21:772–780. doi: 10.1093/annonc/mdp383. [DOI] [PubMed] [Google Scholar]
  • 67.Des Guetz G., Schischmanoff O., Nicolas P., Perret G.Y., Morere J.F., Uzzan B. Does microsatellite instability predict the efficacy of adjuvant chemotherapy in colorectal cancer? A systematic review with meta-analysis. Eur. J. Cancer. 2009;45:1890–1896. doi: 10.1016/j.ejca.2009.04.018. [DOI] [PubMed] [Google Scholar]
  • 68.Flejou J.F., André T., Chibaudel B., Scriva A., Hickish T., Tabernero J. Effect of adding oxaliplatin to adjuvant 5-fluorouracil/leucovorin (5FU/LV) in patients with defective mismatch repair (dMMR) colon cancer stage II and III included in the mosaic study. J. Clin. Oncol. 2013;31:3524. [Google Scholar]
  • 69.Raymond E., Chaney S.G., Taamma A., Cvitkovic E. Oxaliplatin: A review of preclinical and clinical studies. Annals. Oncol. 1998;9:1053–1071. doi: 10.1023/A:1008213732429. [DOI] [PubMed] [Google Scholar]
  • 70.Fink D., Zheng H., Nebel S., Norris P.S., Aebi S., Lin T.P., Nehme A., Christen R.D., Haas M., MacLeod C.L., et al. In vitro and in vivo resistance to cisplatin in cells that have lost DNA mismatch repair. Cancer Res. 1997;57:1841–1845. [PubMed] [Google Scholar]
  • 71.Park J.M., Huang S., Tougeron D., Sinicrope F.A. MSH3 mismatch repair protein regulates sensitivity to cytotoxic drugs and a histone deacetylase inhibitor in human colon carcinoma cells. PLoS ONE. 2013;8:892. doi: 10.1371/journal.pone.0065369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Bras-Goncalves R.A., Rosty C., Laurent-Puig P., Soulie P., Dutrillaux B., Poupon M.F. Sensitivity to CPT-11 of xenografted human colorectal cancers as a function of microsatellite instability and p53 status. Br. J. Cancer. 2000;82:913–923. doi: 10.1054/bjoc.1999.1019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Magrini R., Bhonde M.R., Hanski M.L., Notter M., Scherubl H., Boland C.R., Zeitz M., Hanski C. Cellular effects of CPT-11 on colon carcinoma cells: Dependence on p53 and hMLH1 status. Int. J. Cancer. 2002;101:23–31. doi: 10.1002/ijc.10565. [DOI] [PubMed] [Google Scholar]
  • 74.Ma J., Zhang Y., Shen H., Kapesa L., Liu W., Zeng M., Zeng S. Association between mismatch repair gene and irinotecan-based chemotherapy in metastatic colon cancer. Tumour Biol. 2015;36:9599–9609. doi: 10.1007/s13277-015-3723-5. [DOI] [PubMed] [Google Scholar]
  • 75.Bhonde M.R., Hanski M.L., Stehr J., Jebautzke B., Peiro-Jordan R., Fechner H., Yokoyama K.K., Lin W.C., Zeitz M., Hanski C. Mismatch repair system decreases cell survival by stabilizing the tetraploid G1 arrest in response to SN-38. Int. J. Cancer. 2010;126:2813–2825. doi: 10.1002/ijc.24893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Hausner P., Venzon D.J., Grogan L., Kirsch I.R. The “comparative growth assay”: Examining the interplay of anti-cancer agents with cells carrying single gene alterations. Neoplasia. 1999;1:356–367. doi: 10.1038/sj.neo.7900047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Fedier A., Schwarz V.A., Walt H., Carpini R.D., Haller U., Fink D. Resistance to topoisomerase poisons due to loss of DNA mismatch repair. Int. J. Cancer. 2001;93:571–576. doi: 10.1002/ijc.1356. [DOI] [PubMed] [Google Scholar]
  • 78.Fallik D., Borrini F., Boige V., Viguier J., Jacob S., Miquel C., Sabourin J.C., Ducreux M., Praz F. Microsatellite instability is a predictive factor of the tumor response to irinotecan in patients with advanced colorectal cancer. Cancer Res. 2003;63:5738–5744. [PubMed] [Google Scholar]
  • 79.Charara M., Edmonston T.B., Burkholder S., Walters R., Anne P., Mitchell E., Fry R., Boman B., Rose D., Fishel R., et al. Microsatellite status and cell cycle associated markers in rectal cancer patients undergoing a combined regimen of 5-FU and CPT-11 chemotherapy and radiotherapy. Anticancer Res. 2004;24:3161–3167. [PubMed] [Google Scholar]
  • 80.Bertagnolli M.M., Niedzwiecki D., Compton C.C., Hahn H.P., Hall M., Damas B., Jewell S.D., Mayer R.J., Goldberg R.M., Saltz L.B., et al. Microsatellite instability predicts improved response to adjuvant therapy with irinotecan, fluorouracil, and leucovorin in stage III colon cancer: Cancer and leukemia group B protocol 89803. J. Clin. Oncol. 2009;27:1814–1821. doi: 10.1200/JCO.2008.18.2071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Kim J.E., Hong Y.S., Ryu M.H., Lee J.L., Chang H.M., Lim S.B., Kim J.H., Jang S.J., Kim M.J., Yu C.S., et al. Association between deficient mismatch repair system and efficacy to irinotecan-containing chemotherapy in metastatic colon cancer. Cancer Sci. 2011;102:1706–1711. doi: 10.1111/j.1349-7006.2011.02009.x. [DOI] [PubMed] [Google Scholar]
  • 82.Braun M.S., Richman S.D., Quirke P., Daly C., Adlard J.W., Elliott F., Barrett J.H., Selby P., Meade A.M., Stephens R.J., et al. Predictive biomarkers of chemotherapy efficacy in colorectal cancer: Results from the UK MRC FOCUS trial. J. Clin. Oncol. 2008;26:2690–2698. doi: 10.1200/JCO.2007.15.5580. [DOI] [PubMed] [Google Scholar]
  • 83.Hunter C., Smith R., Cahill D.P., Stephens P., Stevens C., Teague J., Greenman C., Edkins S., Bignell G., Davies H., et al. A hypermutation phenotype and somatic MSH6 mutations in recurrent human malignant gliomas after alkylator chemotherapy. Cancer Res. 2006;66:3987–3991. doi: 10.1158/0008-5472.CAN-06-0127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Coppede F., Lopomo A., Spisni R., Migliore L. Genetic and epigenetic biomarkers for diagnosis, prognosis and treatment of colorectal cancer. World J. Gastroenterol. 2014;20:943–956. doi: 10.3748/wjg.v20.i4.943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Boyer J.C., Umar A., Risinger J.I., Lipford J.R., Kane M., Yin S., Barrett J.C., Kolodner R.D., Kunkel T.A. Microsatellite instability, mismatch repair deficiency, and genetic defects in human cancer cell lines. Cancer Res. 1995;55:6063–6070. [PubMed] [Google Scholar]
  • 86.Watanabe Y., Haugen-Strano A., Umar A., Yamada K., Hemmi H., Kikuchi Y., Takano S., Shibata Y., Barrett J.C., Kunkel T.A., et al. Complementation of an hMSH2 defect in human colorectal carcinoma cells by human chromosome 2 transfer. Mol. Carcinog. 2000;29:37–49. doi: 10.1002/1098-2744(200009)29:1&#x0003c;37::AID-MC5&#x0003e;3.0.CO;2-2. [DOI] [PubMed] [Google Scholar]
  • 87.Carethers J.M., Hawn M.T., Chauhan D.P., Luce M.C., Marra G., Koi M., Boland C.R. Competency in mismatch repair prohibits clonal expansion of cancer cells treated with N-methyl-N’-nitro-N-nitrosoguanidine. J. Clin. Investig. 1996;98:199–206. doi: 10.1172/JCI118767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.York S.J., Modrich P. Mismatch repair-dependent iterative excision at irreparable O6-methylguanine lesions in human nuclear extracts. J. Biol. Chem. 2006;281:22674–22683. doi: 10.1074/jbc.M603667200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Hochhauser D., Glynne-Jones R., Potter V., Gravalos C., Doyle T.J., Pathiraja K., Zhang Q., Zhang L., Sausville E.A. A phase II study of temozolomide in patients with advanced aerodigestive tract and colorectal cancers and methylation of the O6-methylguanine-DNA methyltransferase promoter. Mol. Cancer Ther. 2013;12:809–818. doi: 10.1158/1535-7163.MCT-12-0710. [DOI] [PubMed] [Google Scholar]
  • 90.Douillard J.Y., Oliner K.S., Siena S., Tabernero J., Burkes R., Barugel M., Humblet Y., Bodoky G., Cunningham D., Jassem J., et al. Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N. Engl. J. Med. 2013;369:1023–1034. doi: 10.1056/NEJMoa1305275. [DOI] [PubMed] [Google Scholar]
  • 91.Schwartzberg L.S., Rivera F., Karthaus M., Fasola G., Canon J.L., Hecht J.R., Yu H., Oliner K.S., Go W.Y. Peak: A randomized, multicenter phase II study of panitumumab plus modified fluorouracil, leucovorin, and oxaliplatin (mFOLFOX6) or bevacizumab plus mFOLFOX6 in patients with previously untreated, unresectable, wild-type KRAS exon 2 metastatic colorectal cancer. J. Clin. Oncol. 2014;32:2240–2247. doi: 10.1200/JCO.2013.53.2473. [DOI] [PubMed] [Google Scholar]
  • 92.Karapetis C.S., Khambata-Ford S., Jonker D.J., O'Callaghan C.J., Tu D., Tebbutt N.C., Simes R.J., Chalchal H., Shapiro J.D., Robitaille S., et al. K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N. Engl. J. Med. 2008;359:1757–1765. doi: 10.1056/NEJMoa0804385. [DOI] [PubMed] [Google Scholar]
  • 93.Wang L., Cunningham J.M., Winters J.L., Guenther J.C., French A.J., Boardman L.A., Burgart L.J., McDonnell S.K., Schaid D.J., Thibodeau S.N. Braf mutations in colon cancer are not likely attributable to defective DNA mismatch repair. Cancer Res. 2003;63:5209–5212. [PubMed] [Google Scholar]
  • 94.Tran B., Kopetz S., Tie J., Gibbs P., Jiang Z.Q., Lieu C.H., Agarwal A., Maru D.M., Sieber O., Desai J. Impact of BRAF mutation and microsatellite instability on the pattern of metastatic spread and prognosis in metastatic colorectal cancer. Cancer. 2011;117:4623–4632. doi: 10.1002/cncr.26086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Oyama T., Ran S., Ishida T., Nadaf S., Kerr L., Carbone D.P., Gabrilovich D.I. Vascular endothelial growth factor affects dendritic cell maturation through the inhibition of nuclear factor-kappa B activation in hemopoietic progenitor cells. J. Immunol. 1998;160:1224–1232. [PubMed] [Google Scholar]
  • 96.Ohm J.E., Gabrilovich D.I., Sempowski G.D., Kisseleva E., Parman K.S., Nadaf S., Carbone D.P. VEGF inhibits T-cell development and may contribute to tumor-induced immune suppression. Blood. 2003;101:4878–4886. doi: 10.1182/blood-2002-07-1956. [DOI] [PubMed] [Google Scholar]
  • 97.Laxmanan S., Robertson S.W., Wang E., Lau J.S., Briscoe D.M., Mukhopadhyay D. Vascular endothelial growth factor impairs the functional ability of dendritic cells through id pathways. Biochem. Biophys. Res. Commun. 2005;334:193–198. doi: 10.1016/j.bbrc.2005.06.065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Terme M., Pernot S., Marcheteau E., Sandoval F., Benhamouda N., Colussi O., Dubreuil O., Carpentier A.F., Tartour E., Taieb J. Vegfa-vegfr pathway blockade inhibits tumor-induced regulatory T-cell proliferation in colorectal cancer. Cancer Res. 2013;73:539–549. doi: 10.1158/0008-5472.CAN-12-2325. [DOI] [PubMed] [Google Scholar]
  • 99.Kabbinavar F.F., Hambleton J., Mass R.D., Hurwitz H.I., Bergsland E., Sarkar S. Combined analysis of efficacy: The addition of bevacizumab to fluorouracil/leucovorin improves survival for patients with metastatic colorectal cancer. J. Clin. Oncol. 2005;23:3706–3712. doi: 10.1200/JCO.2005.00.232. [DOI] [PubMed] [Google Scholar]
  • 100.Pogue-Geile K., Yothers G., Taniyama Y., Tanaka N., Gavin P., Colangelo L., Blackmon N., Lipchik C., Kim S.R., Sharif S., et al. Defective mismatch repair and benefit from bevacizumab for colon cancer: Findings from NSABP C-08. J. Natl. Cancer Inst. 2013;105:989–992. doi: 10.1093/jnci/djt140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Hansen T.F., Jensen L.H., Spindler K.L., Lindebjerg J., Brandslund I., Jakobsen A. The relationship between serum vascular endothelial growth factor A and microsatellite instability in colorectal cancer. Colorectal Dis. 2011;13:984–988. doi: 10.1111/j.1463-1318.2010.02357.x. [DOI] [PubMed] [Google Scholar]
  • 102.Yamamoto H., Imai K. Microsatellite instability: An update. Arch. Toxicol. 2015;89:899–921. doi: 10.1007/s00204-015-1474-0. [DOI] [PubMed] [Google Scholar]
  • 103.Llosa N.J., Cruise M., Tam A., Wicks E.C., Hechenbleikner E.M., Taube J.M., Blosser R.L., Fan H., Wang H., Luber B.S., et al. The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints. Cancer Discov. 2015;5:43–51. doi: 10.1158/2159-8290.CD-14-0863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Hamid O., Robert C., Daud A., Hodi F.S., Hwu W.J., Kefford R., Wolchok J.D., Hersey P., Joseph R.W., Weber J.S., et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N. Engl. J. Med. 2013;369:134–144. doi: 10.1056/NEJMoa1305133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Brahmer J.R., Tykodi S.S., Chow L.Q., Hwu W.J., Topalian S.L., Hwu P., Drake C.G., Camacho L.H., Kauh J., Odunsi K., et al. Safety and activity of anti-PD-l1 antibody in patients with advanced cancer. N. Engl. J. Med. 2012;366:2455–2465. doi: 10.1056/NEJMoa1200694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Topalian S.L., Hodi F.S., Brahmer J.R., Gettinger S.N., Smith D.C., McDermott D.F., Powderly J.D., Carvajal R.D., Sosman J.A., Atkins M.B., et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 2012;366:2443–2454. doi: 10.1056/NEJMoa1200690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Lipson E.J., Sharfman W.H., Drake C.G., Wollner I., Taube J.M., Anders R.A., Xu H., Yao S., Pons A., Chen L., et al. Durable cancer regression off-treatment and effective reinduction therapy with an anti-PD-1 antibody. Clin. Cancer Res. 2013;19:462–468. doi: 10.1158/1078-0432.CCR-12-2625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Le D.T., Uram J.N., Wang H., Bartlett B.R., Kemberling H., Eyring A.D., Skora A.D., Luber B.S., Azad N.S., Laheru D., et al. PD-1 blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med. 2015;372:2509–2520. doi: 10.1056/NEJMoa1500596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Kelderman S., Schumacher T.N., Kvistborg P. Mismatch repair-deficient cancers are targets for anti-PD-1 therapy. Cancer Cell. 2015;28:11–13. doi: 10.1016/j.ccell.2015.06.012. [DOI] [PubMed] [Google Scholar]
  • 110.Goldstein J., Tran B., Ensor J., Gibbs P., Wong H.L., Wong S.F., Vilar E., Tie J., Broaddus R., Kopetz S., et al. Multicenter retrospective analysis of metastatic colorectal cancer (CRC) with high-level microsatellite instability (MSI-H) Ann. Oncol. 2014;25:1032–1038. doi: 10.1093/annonc/mdu100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Korphaisarn K., Pongpaibul A., Limwongse C., Roothumnong E., Klaisuban W., Nimmannit A., Jinawath A., Akewanlop C. Deficient DNA mismatch repair is associated with favorable prognosis in Thai patients with sporadic colorectal cancer. World J. Gastroenterol. 2015;21:926–934. doi: 10.3748/wjg.v21.i3.926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Bertagnolli M.M., Redston M., Compton C.C., Niedzwiecki D., Mayer R.J., Goldberg R.M., Colacchio T.A., Saltz L.B., Warren R.S. Microsatellite instability and loss of heterozygosity at chromosomal location 18q: Prospective evaluation of biomarkers for stages II and III colon cancer—A study of CALGB 9581 and 89803. J. Clin. Oncol. 2011;29:3153–3162. doi: 10.1200/JCO.2010.33.0092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Drescher K.M., Sharma P., Lynch H.T. Current hypotheses on how microsatellite instability leads to enhanced survival of lynch syndrome patients. Clin. Dev. Immunol. 2010;2010:170432. doi: 10.1155/2010/170432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Ricciardiello L., Ceccarelli C., Angiolini G., Pariali M., Chieco P., Paterini P., Biasco G., Martinelli G.N., Roda E., Bazzoli F. High thymidylate synthase expression in colorectal cancer with microsatellite instability: Implications for chemotherapeutic strategies. Clin. Cancer Res. 2005;11:4234–4240. doi: 10.1158/1078-0432.CCR-05-0141. [DOI] [PubMed] [Google Scholar]
  • 115.Wendum D., Comperat E., Boelle P.Y., Parc R., Masliah J., Trugnan G., Flejou J.F. Cytoplasmic phospholipase A2 alpha overexpression in stromal cells is correlated with angiogenesis in human colorectal cancer. Mod. Pathol. 2005;18:212–220. doi: 10.1038/modpathol.3800284. [DOI] [PubMed] [Google Scholar]
  • 116.Cohen R., Svrcek M., Dreyer C., Cervera P., Duval A., Pocard M., Flejou J.F., de Gramont A., Andre T. New therapeutic opportunities based on DNA mismatch repair and BRAF status in metastatic colorectal cancer. Curr. Oncol. Rep. 2016;18:18. doi: 10.1007/s11912-016-0504-2. [DOI] [PubMed] [Google Scholar]
  • 117.Nordholm-Carstensen A., Krarup P.M., Morton D., Harling H., Danish Colorectal Cancer G. Mismatch repair status and synchronous metastases in colorectal cancer: A nationwide cohort study. Int. J. Cancer. 2015;137:2139–2148. doi: 10.1002/ijc.29585. [DOI] [PubMed] [Google Scholar]
  • 118.Nopel-Dunnebacke S., Schulmann K., Reinacher-Schick A., Porschen R., Schmiegel W., Tannapfel A., Graeven U. Prognostic value of microsatellite instability and p53 expression in metastatic colorectal cancer treated with oxaliplatin and fluoropyrimidine-based chemotherapy. Z. Gastroenterol. 2014;52:1394–1401. doi: 10.1055/s-0034-1366781. [DOI] [PubMed] [Google Scholar]
  • 119.Venderbosch S., Nagtegaal I.D., Maughan T.S., Smith C.G., Cheadle J.P., Fisher D., Kaplan R., Quirke P., Seymour M.T., Richman S.D., et al. Mismatch repair status and BRAF mutation status in metastatic colorectal cancer patients: A pooled analysis of the CAIRO, CAIRO2, COIN, and FOCUS studies. Clin. Cancer Res. 2014;20:5322–5330. doi: 10.1158/1078-0432.CCR-14-0332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Ooki A., Akagi K., Yatsuoka T., Asayama M., Hara H., Takahashi A., Kakuta M., Nishimura Y., Yamaguchi K. Combined microsatellite instability and BRAF gene status as biomarkers for adjuvant chemotherapy in stage III colorectal cancer. J. Surg. Oncol. 2014;110:982–988. doi: 10.1002/jso.23755. [DOI] [PubMed] [Google Scholar]

Articles from International Journal of Environmental Research and Public Health are provided here courtesy of Multidisciplinary Digital Publishing Institute (MDPI)

RESOURCES