Diagnosis and Management of DNA Mismatch Repair-Deficient Colorectal Cancer




Colorectal tumors exhibiting defective DNA mismatch repair (MMR-D)/microsatellite instability (MSI-H) form a distinct subgroup of CRCs associated with important clinical and pathologic features. The identification of MMR-D/MSI-H may impact CRC prognosis, prediction of response to chemotherapeutic agents, and may necessitate the need for genetic assessment for Lynch syndrome. Oncologists remain at the forefront of diagnosing, treating, and managing patients with MMR-D/MSI-H CRC and ensuring that the clinical care of these patients reflect our evolving understanding of this unique CRC subtype.


Key points








  • Defective DNA mismatch repair (MMR-D), also referred to as microsatellite instability (MSI-H), is present in approximately 15% of colorectal cancers (CRCs).



  • In most cases, MMR-D/MSI-H in colorectal tumors is caused by a noninherited epigenetic event; however, in about one-third of cases, it is caused by Lynch syndrome.



  • Identification of MMR-D/MSI-H in patients with CRC may lead to a diagnosis of Lynch syndrome, with important clinical implications for future cancer surveillance and risk-reduction options for the patient as well as for at-risk family members.



  • MMR-D/MSI-H is associated with distinct clinical and pathologic features, including right-sided colon predominance, early stage at diagnosis, prominent lymphocytic infiltrate, and a poorly differentiated mucinous adenocarcinoma, often showing a medullary component.



  • Although MMR-D/MSI-H in CRC is associated with a favorable prognosis, it also seems to predict for a lack of benefit from 5-fluorouracil chemotherapy.




Introduction


Colorectal cancers (CRCs) may be divided via molecular phenotyping into tumors with normal DNA mismatch repair (MMR) function and those with DNA MMR deficiency (MMR-D), representing ∼15% of CRCs. The hallmark of MMR-D CRC is a distinct type of genomic instability, referred to as high-frequency microsatellite instability (MSI-H). MMR-D/MSI-H is associated with specific clinical and pathologic features, and in some cases, may provide the initial clinical indication suggesting the presence of Lynch syndrome, an inherited form of CRC, characterized by germline genetic defects in one of the DNA MMR genes ( MLH1, MSH2, MSH6, PMS2, EPCAM ). Lynch syndrome remains significantly underdiagnosed, and recent measures to expand screening of patients with Lynch-associated cancers, including those with CRC and endometrial cancer, provides one of the most effective ways to identify at-risk families. The MMR-D/MSI-H phenotype also serves as an important prognostic and predictive marker in CRC. Although MMR-D/MSI-H CRCs tend to be early stage at diagnosis and are associated with a good prognosis, the value of 5-fluorouracil (5-FU) as an effective chemotherapeutic treatment of this type of CRC has been debated. As our understanding of MMR-D/MSI-H CRCs continues to evolve, oncologists remain at the forefront of diagnosing, treating, and managing patients with this unique subtype of CRC and ensuring that, for appropriate patients, referral to clinical cancer genetics for Lynch syndrome evaluation is implemented.


The adenoma-carcinoma model of colorectal cancer


The development of CRCs via the adenoma-carcinoma sequence, initially described by Fearon and Vogelstein in 1990, proposed a multistep genetic model of CRC carcinogenesis. Most CRCs arise along the chromosomal instability pathway, characterized by widespread imbalances in chromosome number (aneuploidy) and loss of heterozygosity, as well as the accumulation of characteristic mutations in tumor suppressor genes and oncogenes critical for CRC initiation and progression. The second pathway, accounting for 15% to 20% of CRCs, results from defective DNA MMR, which leads to the molecular phenomenon of MSI-H shown within the tumor. Microsatellites are short, tandemly repeated DNA sequences that are distributed throughout the human genome and consist of mononucleotide, dinucleotide, or higher-order DNA base repeats. Such repetitive sequences are prone to the accumulation of mutations mostly because of base-base mismatches and insertion-deletion loops. The correction of errors in microsatellites is performed by the MMR proteins, the most notable of which are MLH1, MSH2, MSH6, and PMS2. In tumors with MMR-D, either because of germline, somatic, or epigenetic inactivation, the correction of such mutations is impaired, leading to the accumulation of DNA errors, resulting in the MSI-H phenotype.


In most cases, MMR-D is a result of an epigenetic phenomenon in the tumor, most commonly hypermethylation of the MLH1 promoter, which leads to the development of a sporadic (noninherited) CRC. However, in about 30% of cases, MMR-D is secondary to a germline mutation in one of the MMR genes, leading to a diagnosis of Lynch syndrome, an inherited form of CRC. Deciphering the cause of the MMR-D is of upmost importance, because it helps to differentiate sporadic from inherited forms of colon cancer.


Screening for defective DNA mismatch repair/high-frequency microsatellite instability in patients with colorectal cancer: should a universal approach be applied?


The identification of the MMR-D/MSI-H phenotype in a colorectal tumor may be the first indication that a patient may have a diagnosis of Lynch syndrome. Patients identified to have Lynch syndrome benefit from life-saving cancer surveillance measures. In addition, their at-risk family members would benefit from predictive genetic testing and, if needed, implementation of high-risk cancer surveillance. Although measures aimed at increasing awareness about hereditary CRC, both among individuals as well as among physicians, may be undertaken, one of the most effective ways to identify potential at-risk Lynch families is by evaluating patients who are diagnosed with either CRC or endometrial cancer, the 2 most common Lynch-associated malignancies.


Originally, the Amsterdam criteria and subsequently, the Revised Bethesda guidelines, have served as criteria to select patients with CRC or endometrial cancer in whom further evaluation for MMR-D/MSI-H was indicated. However, in the clinical setting these criteria have proved to be cumbersome to implement and lack specificity and sensitivity. Some groups have recommended expansion of the criteria for MMR/MSI testing to either universal testing, with testing of all patients with CRC, or to include all individuals with CRC diagnosed younger than 70 years. Using universal testing, Lynch syndrome was diagnosed in 2.4% to 3.7% of all unselected patients with CRC. Implementing universal screening for endometrial cancer yielded similar results, with about 1.8% to 3.9% of patients being diagnosed with Lynch syndrome. In the CRC cases, a Lynch syndrome diagnosis would have been missed in ∼12% to 28% of patients if the Revised Bethesda guidelines were used for selection criteria. More expansive testing of CRC and endometrial cases has also proved to be cost effective.


Based on these data, the Mallorca group, consisting of European experts in Lynch syndrome, the Evaluation of Genomic Applications in Practice and Prevention Working Group, and the National Comprehensive Cancer Network have recently updated their recommendations to suggest that either all patients with CRC or all patients with CRC younger than 70 years at diagnosis, and those 70 years or older who meet Bethesda guidelines, should undergo testing by either immunohistochemistry (IHC) or MSI for Lynch syndrome. Implementation of such reflex testing is already occurring across large medical centers, with a recent study suggesting that the specific implementation procedures influenced patient follow-through with genetic testing after an initial positive screening test. For example, institutions with a high level of involvement of genetic counselors in the tracking and communicating of screening results improved patient follow-through and reduced barriers to patient contact.


Identification of defective DNA mismatch repair/high-frequency microsatellite instability tumors


Two accepted methods for the detection of MMR-D are available. MSI testing relies on polymerase chain reaction for amplification of specific microsatellites repeats. The original accepted panel of 5 microsatellite markers, referred to as the Bethesda panel, includes 2 mononucleotides (BAT-25, BAT-26) and 3 dinucleotides (D5S346, D2S123, D17S250). The presence of instability is determined based on comparison of the length of a specific microsatellite marker in the tumor versus the normal DNA, such as adjacent colonic mucosa or a blood sample. Instability in 2 or more markers is defined as an MSI-high tumor, whereas those with 1 unstable marker are designated MSI low. For a sample to be microsatellite stable (MSS), no instability in any of the markers should be present. Given concerns over limited sensitivity of dinucleotide repeats, the 2002 National Cancer Institute workshop made further revisions, with recommendations to include a secondary panel of mononucleotide markers, such as BAT-40, to exclude MSI-low cases in which only a dinucleotide repeat is mutated.


In 1996, monoclonal antibodies against MMR proteins became available, rendering IHC detection of MMR proteins possible. A lack of expression of 1 or more of these proteins is diagnostic of MMR-D, with the specific pattern of expression also helping to pinpoint which gene is most likely to harbor a mutation or may be inactivated by another mechanism ( Table 1 ). Initial concerns over lower sensitivity of IHC as opposed to MSI screening have largely been overcome with the introduction of the 4-antibody panel (MLH1, MSH2, MSH6, PMS2). In the clinical setting, both IHC and MSI testing are used broadly and are essentially interchangeable techniques with greater than 90% concordance and similar sensitivity of around 85% to 92%. At our institution, the preference to use IHC as opposed to MSI testing for initial screening was largely based on the ability of IHC to pinpoint which genes should be targeted for subsequent germline analysis in MMR-D cases, thereby simplifying the genetic testing process.



Table 1

Commonly observed patterns of DNA MMR protein expression by IHC staining
















































Protein Expression by IHC Staining MMR Status Most Likely Inactivated Gene
MLH1 MSH2 MSH6 PMS2
Present Present Present Present Proficient None
Absent Present Present Absent Deficient MLH1 (epigenetic or germline)
Present Present Present Absent Deficient PMS2
Present Absent Absent Present Deficient MSH2 , possibly EPCAM
Present Present Absent Present Deficient MSH6


Defective DNA mismatch repair/high-frequency microsatellite instability colorectal cancer: making the diagnosis of Lynch syndrome


An MSI-H or an abnormal IHC result does not distinguish between a somatic (sporadic) versus a germline (inherited) defect in the MMR system. Without further analysis, the cause of the defect remains elusive. In approximately two-thirds of cases, MSI-H indicates a sporadic CRC, with the molecular phenomenon being caused by an epigenetic event, most commonly MLH1 promoter hypermethylation, leading to MLH1 gene inactivation. In about 70% of such promoter hypermethylated MSI-H cases, the presence of a BRAF V600E mutation can be identified. The presence of a BRAF V600E mutation in a colorectal tumor would make the diagnosis of Lynch syndrome less likely, and together with an absence of suggestive family history, may reassure clinicians of the sporadic cause of the MSI-H CRC. Cases of BRAF mutated tumors in Lynch syndrome–associated CRCs have been reported, and therefore a careful review of the clinical and family history should be undertaken to determine whether further genetic counseling/testing is indicated. In many centers, an MSI-H CRC or a CRC showing loss of MLH1/PMS2 protein expression is immediately tested via direct MLH1 hypermethylation analysis or BRAF V600E mutation analysis. These are generally not considered genetic tests but are used for risk stratification to determine which patients may benefit from subsequent genetic testing.


In one-third of MSI-H tumors, the molecular defect is secondary to a germline mutation in one of the MMR genes, diagnostic of Lynch syndrome. In patients with CRC in whom Lynch syndrome is suspected based on early age at diagnosis or family history of Lynch-associated malignancies, an abnormal IHC or MSI-H result should prompt referral for genetic counseling and genetic testing. If IHC was performed, then the specific protein expression loss helps guide genetic testing (see Table 1 ). For example, in a tumor with absence of MSH2 and MSH6 expression, genetic testing for mutations in the MSH2 gene should be undertaken initially. Identification of a genetic mutation confirms the diagnosis of Lynch syndrome. In such patients, appropriate cancer surveillance and options for cancer risk reduction need to be reviewed and implemented. Moreover, expansion of genetic counseling and testing for family members should be undertaken.


A difficult clinical situation that arises not infrequently is that workup of a patient with an MSI-H or MMR-D CRC results in ambiguous or uninformative results with respect to the origin of the MMR defect. For example, in a young patient with CRC with absence of MLH1/PMS2 protein expression, genetic testing of the MLH1 and PMS2 genes may be unrevealing, with no mutation identified. Coupled with the absence of MLH1 promoter hypermethylation and absence of a BRAF mutation, in such a clinical circumstance, Lynch syndrome cannot be ruled out. In such situations, a careful evaluation of the family history may help to identify other cancer-affected family members whose tumors could be evaluated for the MMR-D/MSI-H molecular signature and, if positive, would confirm the presence of an occult mutation. These cases require the careful input of genetic counselors and cancer geneticists for recommendations for appropriate cancer surveillance for both the patient and at-risk family members.


Clinical and pathologic features of defective DNA mismatch repair/high-frequency microsatellite instability colorectal cancers


Regardless of the cause of the MMR deficiency, MMR-D/MSI-H CRCs seem to be associated with distinct clinical and pathologic features, which often serve as the initial clue that a particular tumor may harbor an MMR defect ( Table 2 ). As opposed to the 25% of MSS colorectal tumors, 85% of MMR-D/MSI-H tumors are proximal to the splenic flexure. Moreover, the age distribution of MMR-D/MSI-H tumors seems to be bimodal, with about 24% of patients with CRC younger than 40 years and 19% older than 70 years having MSI-H tumors, as opposed to only 8% of patients with CRC aged between 50 and 59 years. This U-shaped age distribution reflects the higher prevalence of Lynch-associated MMR-D/MSI-H CRCs in young patients and the similarly higher prevalence of sporadic MMR-D/MSI-H phenotype observed in older patients.



Table 2

Clinical and pathologic features of MMR-D/MSI-H CRCs





















Right-colon predominance
Mucinous, signet-ring cells
Tumor-infiltrating lymphocytes, Crohn-like nodular infiltrate
Absence of necrotic cellular debris
Poorly differentiated adenocarcinoma (medullary subtype)
Low pathologic stage
Sporadic, Noninherited MMR-D Inherited MMR-D (Lynch Syndrome)
Presence of epigenetic MLH1 promoter hypermethylation Presence of a germline mutation in one of the MMR genes ( MLH1 , MSH2 , MSH6 , PMS2 )
Often positive for V600E BRAF somatic mutation Generally, no somatic BRAF mutation identified
More common in older and female patients Increased risk of synchronous and metachronous CRC
Risk of extracolonic cancers (eg, endometrial, ovarian, gastric, ureter, pancreas)
Requires genetic counseling and testing of at-risk family members


Despite the improved prognosis (see later discussion) associated with the MMR-D/MSI-H phenotype, such tumors tend to show a higher histologic grade and are often mucinous tumors with presence of signet-ring cells. A recently recognized feature is the presence of the distinct medullary subtype of colorectal adenocarcinoma, associated with a unique histologic appearance and a more favorable prognosis compared with standard poorly differentiated colonic carcinomas. Medullary carcinomas are generally associated with a right-sided predominance, older female patients, and a lower incidence of lymph node metastases. Morphologic features of such tumors are characterized by a syncytial growth pattern and large vesicular nuclei, with conspicuous nucleoli and prominent peritumoral lymphocytic infiltrates. Because there is significant morphologic overlap between medullary carcinomas and poorly differentiated carcinomas, these may be difficult to differentiate. However, the presence of medullary carcinoma subtype, even if categorized under a poorly differentiated carcinoma, is associated with a favorable prognosis. In 1 study, MLH1 antibody staining was absent in nearly 80% of medullary carcinomas, indicating the close association of MMR-D/MSI-H with this subtype of CRC. Other common features include prominent tumor-infiltrating lymphocytes, lack of necrotic debris, and a Crohn-like lymphocytic host reaction. These distinct histologic features evoke the possible presence of MMR-D/MSI-H, prompting the pathologist to perform subsequent MSI or IHC testing.


Defective DNA mismatch repair/high-frequency microsatellite instability as a prognostic marker in colorectal cancer


In addition to the distinct clinicopathologic features, numerous studies have also suggested a favorable prognosis of MMR-D/MSI-H colon tumors. Patients with MMR-D/MSI-H tumors tend to have a lower tumor stage at diagnosis and rarely develop metastatic disease. When assessed stage by stage, the presence of MMR-D/MSI-H is noted in ∼20% of stage I/II, ∼12% of stage III, but only ∼4% of stage IV patients, again suggesting an association with earlier stage of disease. Substantial evidence has accumulated from retrospective studies, population-based studies, as well as a meta-analysis to suggest a more favorable prognosis in MMR-D/MSI-H colorectal tumors.


In 2000, Gryfe and colleagues initially reported an association of MSI-H status, with a survival advantage that was independent of all standard prognostic features. This analysis was limited to CRCs diagnosed at age 50 years or younger, limiting the generalizability of this result to all patients with CRC. In the seminal 2003 publication by Ribic and colleagues, of patients with stage II and III colon cancer who did not receive adjuvant chemotherapy, those with MMR-D/MSI-H had significantly improved overall survival compared with MMR-P/MSS tumors with a hazard ratio (HR) for death of 0.31 (95% confidence interval [CI], 0.14–0.72, P = .004).The improved prognosis associated with MMR-D/MSI-H was despite the presence of a higher percentage of poorly differentiated tumors in the MMR-D/MSI-H group (26% vs 9%). A subsequent meta-analyses of 32 studies, including a total of 7642 patients with CRC, including all stages of disease, also found a favorable overall survival in the 1277 MSI-H tumors with an HR of 0.65 (95% CI, 0.59–0.71). When the analysis was restricted to clinical trials only, the benefit was maintained with an HR of 0.69. In this meta-analysis, both treated (5-FU–based adjuvant treatment) and untreated patients were included from phase 3 randomized trials. In the Quick And Simple And Reliable (QUASAR) trial assessing stage II CRCs, MMR-D was an independent prognostic factor, with an improvement in survival with an HR of 0.31. Nonetheless, several studies do not corroborate the survival advantage associated with MMR-D/MSI-H tumors. Such discrepant results may be caused by limited power to detect prognostic differences because of small sample size, different methodologies used for the assessment of defective MMR (ie, 4 vs 2 antibody IHC testing; MSI analysis with different panel and numbers of markers), different treatment and stage of disease included in the analysis, and selection bias. Because patients with MMR-D/MSI-H caused by Lynch syndrome comprise an even smaller group (∼2–3% of all CRCs), power to detect prognostic differences specifically for patients with Lynch syndrome is even more limited. Nonetheless, based on the preponderance of evidence, it is generally well accepted that MMR-D/MSI-H tumors are associated with an improved prognosis, with the more contentious issue being the importance of MMR-D/MSI-H status as a predictive marker in the treatment of CRC.


Defective DNA mismatch repair/high-frequency microsatellite instability status as a predictive marker in colorectal cancer


5-Fluorouracil–Based Chemotherapy


In addition to being a prognostic marker for CRC outcome, the MMR-D/MSI-H phenotype has also been implicated as a predictive marker, with studies suggesting that such tumors do not derive a benefit from adjuvant 5-FU–based chemotherapy. However, this conclusion has been fraught with controversy, because the lack of benefit from 5-FU–based chemotherapy is not consistent across all studies evaluating MMR-D/MSI-H CRCs. 5-FU is a mainstay of chemotherapy for CRC in both the early stage and metastatic disease. In vitro studies have attempted to assess the effect of 5-FU on MSI-H CRC cell lines. Preclinical data using cell lines show that MMR-D cells seem to have a growth advantage and are more resistant to the cytotoxic effects of 5-FU compared with MMR-P cells. Moreover, if MMR function was restored via chromosome 3 transfer, then, the growth advantage in the presence of 5-FU was no longer present. Conflicting data showed that MMR-D cell lines, specifically, LOVO, showed similar sensitivity to 5-FU as MMR-D cell lines, SW480. Subsequent evaluations of the MSI-H LOVO cell line showed increased thymidylate synthase activity and reduced sensitivity to 5-FU. Data are also starting to emerge that the underlying cause of the MSI-H phenotype may be relevant in predicting response to 5-FU. For example, it has been suggested that MSI-H tumors with a CpG island methylator phenotype may show specific sensitivity to 5-FU.


Initial clinical studies evaluating response to 5-FU in MMR-D/MSI-H CRCs indicated a beneficial impact of 5-FU on stage III colon cancers with improved overall or recurrence-free survivals. However, the sample size was limited in both of these studies. A pivotal study published by Ribic and colleagues analyzed patients with stage II and III colon cancer from pooled randomized clinical trials in which patients received either 5-FU chemotherapy or no treatment after surgical resection. Of 570 patients with tissue available, 95 had an MSI-H tumor, 53 of whom received 5-FU chemotherapy. In the MSI-H patients who did not receive chemotherapy, an improved 5-year survival rate was reported; however, this survival advantage was not present in the MSI-H group who received 5-FU chemotherapy. Moreover, the study seemed to suggest that MSI-H patients with colon cancer who received 5-FU had a reduced survival compared with those who did not receive treatment. In an effort to provide validation of these findings, an international collaboration was formed to pool data from patients with stage II/III colon cancer receiving adjuvant 5-FU versus surgery alone as part of 5 different adjuvant colon cancer clinical trials. Of the 457 patients, 70 (15%) had MMR-D/MSI-H colon cancer. Although adjuvant 5-FU significantly improved disease-free survival (DFS) in the MSS/MMR-P tumors, in the MMR-D/MSI-H tumors receiving 5-FU, no improvement in DFS was seen. When this data set (n = 457) and the Ribic data set (n = 570) were pooled, in the 515 untreated patients with stage II and III CRC, those with MMR-D/MSI-H had a clear improved 5-year DFS over MSS tumors. On the other hand, in the 512 treated patients with CRC, the improvement in DFS within the MMR-D/MSI-H subgroup was no longer apparent, suggesting that 5-FU may have abrogated the benefit associated with the MMR-D/MSI-H phenotype. Moreover, in patients with stage II colon cancer with MMR-D/MSI-H tumors, a reduced overall survival was seen (HR, 2.95; 95% CI, 1.02–8.54; P = .04). The findings of a potential detrimental effect of 5-FU in MMR-D/MSI-H cases were specific to patients with stage II disease only, and applicability to other stages may not be appropriate.


Despite the data from these trials, there have been additional studies, including an analysis of patients treated in adjuvant colon cancer trials conducted by the National Surgery Adjuvant Breast and Bowel Project (NSABP), which reported no interaction between MMR status and 5-FU response. Although an advantage of this trial was that MSI analysis was performed in a uniform way with the NCI-based reference of 5 markers, only 20% of all patients enrolled in the NSABP C-01 to C-04 studies had paraffin blocks that were suitable for MSI analysis. In a more recent analysis including more then 600 patients with stage II/III colon cancer treated with 5-FU in the control arm of the Pan European Trial Adjuvant Colon Cancer (PETACC-3) study, a significant improvement in 5-year DFS was seen with MMR-D/MSI-H compared with MSS tumors, indicating that perhaps the improved prognosis is maintained even under 5-FU treatment. Further analysis of this data set suggested that the prognostic impact of MSI-H phenotype is substantially stronger in stage II as opposed to stage III patients. This finding may suggest stage-specific biological effects of the MMR-D/MSI-H phenotype. In contrast to the previous reports, in this study, the prognostic effect of MMR-D/MSI-H remained significant, despite treatment with 5-FU even in the stage II setting.


Adding to the evidence is a recent study by Sinicrope and colleagues, who assessed stage II/III colon cancers and MMR status using samples from patients enrolled in adjuvant therapy clinical trials evaluating 5-FU with levamisole or leucovorin versus surgery alone. MMR-D/MSI-H was associated with reduced recurrence rates, delayed time to recurrence, and fewer distant recurrences. Distant recurrences were reduced with 5-FU–based adjuvant treatment in stage III but not in stage II MMR-D/MSI-H patients. Again, this finding seems to suggest that stage III colon cancers with MMR-D/MSI-H seem to benefit from 5-FU–based adjuvant treatment and should be treated with adjuvant chemotherapy per the current standard of care. In a subgroup analysis, this study also suggested that any treatment benefit was restricted to patients with suspected germline as opposed to sporadic MMR-D/MSI-H. Studies assessing the response of MMR-D/MSI-H tumors to FOLFOX (5-FU, leucovorin, oxaliplatin) are awaited and may provide further insight into the outcome and the optimal treatment of MMR-D/MSI-H tumors.


Evidence overwhelmingly suggests that MMR-D/MSI-H CRC carries a better prognosis, yet, the efficacy of 5-FU chemotherapy in this subtype of CRC has been called into question. Bearing in mind the conflicting data, our general approach to the treatment of MMR-D/MSI-H CRC in the adjuvant setting has been that of avoiding the administration of 5-FU alone. Specifically, given the improved prognosis, as well as the lack of clear benefit (and suggestion of a potential detrimental effect), in stage II MMR-D/MSI-H CRCs, adjuvant treatment can generally be spared. In stage III MMR-D/MSI-H CRCs, there is no evidence to suggest that standard oxaliplatin-based treatment is ineffective, although the prognostic impact of MMR-D may be more modest in stage III as opposed to stage II CRCs. Our approach within the stage III setting has been to administer standard adjuvant treatment with an oxaliplatin-containing regimen. Studies assessing the response of MMR-D/MSI-H tumors to FOLFOX (5-FU, leucovorin, oxaliplatin) are awaited and may provide further insight into the outcome and the optimal treatment of MMR-D/MSI-H tumors.


Irinotecan


Preclinical data also suggest that MMR-D/MSI-H CRC cell lines may be more sensitive to irinotecan, a topoisomerase I inhibitor, with known clinical efficacy in advanced CRC. Irinotecan introduces double-strand DNA breaks usually repaired by either homologous recombination or nonhomologous end joining. In tumors with MMR-D, mutations arising in microsatellite repeats in more than 30 genes have been described. Colorectal tumors and cell lines with MMR-D frequently accumulate mutations within microsatellite repeats of genes implicated in the double-strand DNA break repair pathway, including the MRE11A and h RAD50 genes. A mutation in either one of these genes results in significantly decreased expression of the Mre11/Nbs1/Rad50 (or MRN) protein complex. Mutations in MRE11 lead to a weak interaction with RAD50 and low affinity to Nbs1, compared with wild-type MRE11 . It has been postulated that decreased expression of the MRN protein complex leading to impaired double-stranded DNA break repair pathway in MMR-D tumors/cell lines may make these cells especially sensitive to agents that lead to disruption of DNA repair, such as irinotecan and potentially poly(ADP-ribose) polymerase inhibitors. MMR-D colon cancer cell lines that show an intronic frameshift mutation of MRE11 show the greatest sensitivity to irinotecan.


In the clinical setting, the evidence for the impact of MMR-D/MSI-H on irinotecan sensitivity remains limited. In a retrospective analysis of 73 patients with metastatic CRC receiving second-line irinotecan-based therapy, a higher response rate was observed in MSI-H than in MSI-low or MSS tumors. DFS according to MSI status was assessed through a retrospective evaluation of the Cancer and Leukemia Group B (CALGB) 89803 adjuvant trial, which assigned patients with resected stage III colon cancer to 5-FU/leucovorin with or without irinotecan. Although in the overall group, no advantage for the addition of irinotecan to 5-FU/leucovorin was shown, in patients with MMR-D tumors, irinotecan resulted in a modest improvement in DFS. The third study, presented at ASCO 2009, reported conflicting results. In a retrospective analysis of the PETACC-3 trial, randomizing patients with stage II and III colon cancer to folinic acid, 5-FU, and irinotecan versus biweekly infusional 5-FU and folinic acid, of the 188 MSI-H cases, those treated with irinotecan did not have an improved survival. In a retrospective analysis of the Capecitabine, Irinotecan, and Oxaliplatin in advanced colorectal cancer (CAIRO) trial, which included sequential irinotecan-containing treatments for metastatic CRC, the MSI-H cases were limited to only 14 and because both treatment arms received irinotecan at some point, interpretation of the MSI-H data was not possible. More studies to clarify the role of irinotecan in MSI-H cancers are clearly necessary. At this point, the use of irinotecan should be limited to advanced/metastatic CRC, irrespective of MMR status, because there is insufficient evidence for its use in the adjuvant setting, even in MMR-D/MSI-H tumors.


Summary


The identification of the MMR-D/MSI-H subtype of CRC has important clinical implications for the prognosis of CRC, prediction of response to chemotherapeutic agents, specifically efficacy of 5-FU, and for the need for genetic assessment for Lynch syndrome. Recent expert consensus recommendations suggest that either all patients with CRC or those with CRC at younger than 70 years, should undergo testing by either IHC or MSI, thereby leading to more patients with CRC being recognized as having this subtype of tumor. There is a pressing need for unambiguous answers with respect to the prognostic and predictive impact of MMR-D/MSI-H. Provocative, but preliminary, data suggesting a stage-specific impact and possible inherent biological differences between sporadic versus germline MMR-D/MSI-H tumors remain important questions awaiting validation. With advances in gene expression profiling, next-generation sequencing technologies, and microRNA assessments, further biologically relevant classifications of CRC are anticipated, with MMR-D/MSI-H being just one of many relevant subgroups. Drug development and clinical trial design centered on specific molecular subtypes of tumors is a rapidly emerging approach to novel oncological treatments, and the MMR-D/MSI-H subtype may prove to be a useful marker for such targeted approaches.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Mar 1, 2017 | Posted by in HEMATOLOGY | Comments Off on Diagnosis and Management of DNA Mismatch Repair-Deficient Colorectal Cancer

Full access? Get Clinical Tree

Get Clinical Tree app for offline access