Skip to main content
The Journal of Molecular Diagnostics : JMD logoLink to The Journal of Molecular Diagnostics : JMD
. 2008 Jul;10(4):293–300. doi: 10.2353/jmoldx.2008.080031

Immunohistochemistry versus Microsatellite Instability Testing For Screening Colorectal Cancer Patients at Risk For Hereditary Nonpolyposis Colorectal Cancer Syndrome

Part I. The Utility of Immunohistochemistry

Jinru Shia 1,*
PMCID: PMC2438196  PMID: 18556767

Abstract

The utility of immunohistochemical detection of DNA mismatch repair (MMR) protein in screening colorectal tumors for hereditary nonpolyposis colorectal cancer (HNPCC) syndrome has been the focus of much intensive research over the last 10 years. Particular attention has been given to the relative usefulness of immunohistochemistry (IHC) versus testing of tumor microsatellite instability (MSI). Earlier work that focused on mutL homolog 1 (MLH1) and mutS homolog 2 (MSH2) has created a false impression that IHC has a lower sensitivity than MSI testing in predicting germline mutation. More recent studies that included postmeiotic segregation increased 2 (PMS2) and MSH6, on the other hand, have demonstrated an IHC predictive value that is virtually equivalent to that of MSI testing. Such added value of PMS2 and MSH6 can be explained by the biological and biochemical properties of the MMR proteins. On the premise that IHC with PMS2 and MSH6 is as sensitive as MSI testing, given that IHC is easily available and generally inexpensive and, importantly, identifies the affected gene, it is reasonable to regard IHC as a more optimal first-line screening tool than MSI testing for identifying HNPCC. MSI testing can provide a fallback position in equivocal situations, while remaining an important research tool. However, for IHC to be used as a first-line screening test requires that both pathologists and clinicians be aware that IHC results may be construed as “genetic information,” and that appropriate procedures should be established to ensure patient understanding and consent.


Hereditary nonpolyposis colorectal cancer (HNPCC), also known as Lynch syndrome—a condition originally characterized as familial clustering of colorectal and other types of cancer,1,2 is now molecularly defined as a cancer-predisposing syndrome secondary to a deleterious germline mutation in one of a set of DNA mismatch repair (MMR) genes, namely, mutL homolog 1 (MLH1), mutS homologs 2 and 6 (MSH2, MSH6), and postmeiotic segregation increased 2 (PMS2).3 In this condition, affected persons carry one mutated copy of the gene in all their tissues, and a somatic mutation or loss of the second normal allele in colorectal or other epithelium inactivates the gene and impairs mismatch repair function, resulting in neoplasia. As such, detection of the deleterious germline mutation has evolved into the ultimate diagnostic criterion for HNPCC.

Screening for mutation, however, is both time-consuming and expensive. This is largely because of the heterogeneity of the mutation spectrum of the MMR genes. Such difficulties make preselection of high-risk patients necessary. Consequently, there have been tremendous research efforts over the last 10 years that aim at designing the most efficient workup algorithm.3,4,5 Among the commonly studied screening tools, ie, family history, tumor pathological characteristics, tumor DNA microsatellite instability (MSI), and tumor MMR protein detection by immunohistochemistry (IHC), the relative usefulness of MSI testing versus IHC has been the topic of much intensive investigation.

Historically, DNA MSI, a molecular manifestation of tumors bearing DNA MMR deficiency, was the phenotype that led to the discovery of human MMR genes and their causative role in HNPCC.6,7,8,9 Such a breakthrough occurred in 1993; testing of MSI has since remained a prominent methodology in HNPCC-related research and clinical work.

Around 1996, monoclonal antibodies against MMR proteins started to become available; first came antibodies to MSH210,11 and then to others. Such antibodies rendered IHC detection of MMR protein possible, providing an alterative methodology for detecting MMR deficiency.

Paralleling the recognition of the greater impact of germline mutations in MLH1 and MSH2 in HNPCC, much research work on the utility of IHC focused on the products of these two genes. Such work, unfortunately, has accentuated the limitations of MLH1 IHC in predicting MLH1 mutation,12 therefore creating a false impression13 that IHC was inferior to MSI testing. Other, mostly more recent work realized a significant compensatory effect of PMS2 antibody on the detection of MLH1 mutation,14 and as such, demonstrated an apparent improvement of the performance of IHC by adding PMS2 to the antibody panel.4,15,16 In this article, we summarize both the early and more recent literature data on the use of IHC, discuss the biological and biochemical basis of how IHC works in MMR-deficient tumors, and outline the advantages and limitations of this methodology. Our analysis indicates that IHC is in fact a more optimal first-line screening tool than MSI testing for identifying HNPCC.

Literature Review

In this review, pertinent literature reports that analyzed the effectiveness of IHC versus MSI in predicting germline mutation in colorectal tumors are divided into two groups according to the IHC antibodies used. The first group includes those that primarily assessed MLH1 and MSH2 (with or without MSH6),12,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31 and the second group, those that assessed all four proteins (MLH1, MSH2, MSH6, and PMS2).4,14,15

Results from the most pertinent studies belonging to the first group are summarized in Table 1. After weighing for sample size, IHC with MLH1/MSH2 antibodies had a sensitivity of 85% in predicting germline mutation in MLH1/MSH2. This was apparently lower than the 93% sensitivity of MSI testing. It became clear on further analysis that this low sensitivity of IHC was largely caused by a low rate of MLH1 mutation detection by MLH1 IHC (74%).

Table 1.

Literature Data on Sensitivity of MLH1- and MSH2-IHC versus MSI Testing in Predicting Germline Mutation in MLH1 and MSH2 in Colorectal Carcinoma

Abnormal IHC or MSI-H/pathogenic mutation
IHC
MSI
References Overall MLH1 MSH2 Overall MLH1 MSH2
Marcus et al 199917 16/16 5/5 11/11 16/16 5/5 11/11
Debniak et al 200018 4/6 2/4 2/2 5/6 3/4 2/2
Dieumegard et al 200019 5/7 3/4 2/3 8/8 5/5 3/3
Cunningham et al 200120 5/5 2/2 3/3 5/5 2/2 3/3
Salahshor et al 200112 22/28 14/20 8/8 27/30 19/22 8/8
Stone et al 200121 4/4 4/4 4/4 4/4
Terdiman et al 200122 11/13 6/7 5/6 13/13 7/7 6/6
Furukawa et al 200223 8/8 3/3 5/5
Wahlberg et al 200224 4/9 1/5 3/4 14/14 8/8 6/6
Hendriks et al 200325 29/33 18/21 11/12 19/21 13/15 6/6
Hoedema et al 200326 4/4 4/4 3/4 3/4
Mangold et al 200527 67/82 29/44 38/38
Pinol et al 200528 11/11* 3/4 6/7 10/11 4/4 6/7
Shia et al 200529 25/32 4/9 21/23 27/28 6/7 21/21
Barnetson et al 200630 21/23 12/12 9/11 19/24 10/12 9/12
Niessen et al 200631 15/16 7/8 8/8 17/17 8/8 9/9
Total 251/297 (85%) 113/152 (74%) 136/145 (94%) 187/201 (93%) 94/103 (91%) 93/98 (95%)
*

One case: MLH1 present/MSH2 lost but with mutation on MLH1; one case: MLH1 lost/MSH2 present but with mutation on MSH2.

Results from the most pertinent studies belonging to the second group are summarized in Table 2. As shown here, with the use of all four antibodies, the sensitivity of IHC in predicting MLH1 mutation increased to 92%, which was equivalent to that of MSI testing. Such an improvement was largely the result of the ability of PMS2 IHC to predict MLH1 mutation (mechanisms to be explained below). This is further supported by a study by de Jong et al14 (not included in Table 2 because of lack of correlative data on other genes) in which PMS2 IHC detected 23% (8/35) of MLH1 mutated tumors that were missed by MLH1 IHC. Table 2 also shows that MSH6 IHC predicted MSH6 mutation in 8 of 8 tumors; in contrast, high frequency MSI was seen in only 2 of the 8 cases.

Table 2.

Literature Data on Sensitivity of MLH1-, MSH2-, MSH6-, and PMS2-IHC versus MSI Testing in Predicting Germline Mutation in MLH1, MSH2, MSH6, and PMS2 in Colorectal Carcinoma

Abnormal IHC or MSI-H/pathogenic mutation
IHC
MSI
Reference Overall MLH1 MSH2 MSH6 PMS2 Overall MLH1 MSH2 MSH6 PMS2
Hampel et al 20054 21/23 4/5 12/13 3/3 2/2 21/23 5/5 12/13 2/3 2/2
Southey et al 200515 18/18 9/9 4/4 4/4 1/1 13/18 9/9 4/4 0/4* 0/1
Lagerstedt Robinson et al 200716 23/25 10/11 12/13 1/1 0 21/25 9/11 12/13 0/1 0
Total 62/66 (94%) 23/25 (92%) 28/30 (93%) 8/8 (100%) 3/3 (100%) 55/66 (83%) 23/25 (92%) 28/30 (93%) 2/8 (25%) 2/3 (67%)
*

All 4 are MSI-L (Ten microsatellite markers assessed: 3 dinucleotide repeats - D5S346, D17S250, and 2S123, and 7 mononucleotide repeats - BAT-25, BAT-26, BAT-40, MYB, TGFßRII, IGFIIR, and BAX. In this study, the degree of instability was scored as stable, low, and high when 0 to 1, 2 to 5, and 6 to 10 markers were identified as unstable, respectively).

In summary, literature review suggests that IHC with MLH1/MSH2 has a lower sensitivity than MSI testing in predicting gene mutation; however, inclusion of PMS2 and MSH6 significantly increases the sensitivity of IHC, resulting in a predictive value that is virtually equivalent to that of MSI testing.

With regard to the specificity of IHC in predicting germline mutation, data are more limited as the analysis requires that large numbers of cases (including clinically unsuspected cases) be tested for mutation so as to achieve a meaningful group of mutation-negative cases. Many studies reported IHC data in mutation-positive cases only. In general, however, the specificity is believed to be high; one study29 reported 95% specificity for both MLH1 and MSH2 IHC.

It is to be noted that whereas both MSI and the four-antibody IHC have similar value in predicting germline mutation, the concordance rate between these two tests is less than perfect; the sensitivity of IHC in predicting MSI is about 92%.32 This is because both tests may miss cases that are detectable by the other. Specifically, MSI may detect cases that have abnormalities in MMR genes that are not covered by the IHC antibody panel and therefore not detectable by IHC; on the other hand, as shown in Table 2, IHC can detect MSH6 mutation cases that may not show high frequency MSI and therefore can be missed by MSI testing.

The Biological and Biochemical Basis of How IHC Works in MMR-Deficient Tumors

Not All Pathogenic Mutations Result in Loss of Protein by IHC

In general, IHC is reliable in screening for mutations that result in truncation or degradation of the protein. IHC, however, cannot distinguish between mutant proteins commonly resulting from missense mutations and wild-type polypeptides.

Most mutations in MSH2 are protein truncating; consequently, most MSH2-mutant colorectal tumors are expected to show absent MSH2 expression by IHC.27 However, more than one-third of the mutations in MLH1 are missense mutations that may result in mutant proteins that are catalytically inactive but antigenically intact.12,33 Thus, on IHC, these mutant proteins may result in a false-normal staining pattern. Moreover, false-normal staining for MLH1 can occur even with protein-truncating mutations and large in-frame deletions in MLH1,12,24,27,34 the mechanism for which is unclear.

Yet another element that could potentially result in a false-normal staining relates to the second hit that inactivates the second normal allele. This second hit might result in a nonfunctional but antibody-binding MLH1 protein detectable by IHC. Such a possibility exists as various individuals with an identical germline mutation in MLH1 have been shown to exhibit different MLH1 IHC staining patterns.27

Mutation of MLH1 or MSH2 Will Result in Concurrent Loss of MLH1/PMS2 or MSH2/MSH6, Respectively, by IHC, Whereas Mutation of PSM2 or MSH6 Will Result in Isolated Loss of PMS2 or MSH6 Only

In their functional state, the MMR proteins form heterodimers.35,36,37,38 MSH2 dimerizes with MSH6, forming the functional complex, MutSα38; and MLH1 dimerizes with PMS2, forming MutLα.37,39 It has been shown that the MSH2 and MLH1 proteins are the obligatory partner of their respective heterodimer.14,40,41,42 Their abnormalities can result in proteolytic degradation of their dimer and consequent loss of both the obligatory and secondary partner proteins (exceptions include some MLH1 mutations: when the mutation results in an antigenically active mutant MLH1 protein as discussed above, there may be loss of PMS2 only). The reverse, however, is not true. When mutation occurs in genes of the secondary proteins, ie, MSH6 and PMS2, there may not be concurrent loss of the obligatory proteins, MSH2 and MLH1. This is because the function of the secondary proteins may be compensated by other proteins, such as MSH3, MLH3, and PMS1. As a result, mutations of MLH1 or MSH2 often cause concurrent loss of MLH1/PMS2 or MSH2/MSH6, respectively, by IHC, whereas mutations of PSM2 or MSH6 often cause isolated loss of PMS2 or MSH6 only.

A curious phenomenon worth noting here is that mononucleotide repeats of 7 or more elements (ie, A7, C8, etc) exist in the coding sequence of some MMR genes, namely MSH2, MSH6, and PMS2.43,44 Theoretically, such encoded microsatellites can undergo secondary mutation in HNPCC and result in a pattern of protein loss that deviates from the common situations described above. For example, concurrent loss of MLH1 and MSH6 may occur because of secondary loss of MSH6 in an MLH1 mutation case. Such aberrant patterns (with complete loss of a second, “unexpected” protein), however, are extremely rare.45 This is so, probably because secondary mutation does not occur during carcinogenesis, but rather, only in subclones of an established MSI cancer. It follows that such a mechanism may explain partial loss of IHC staining in some tumors (see section on IHC staining pattern below).

Summary

In summary, IHC with antibodies to only MLH1/MSH2 is not able to detect all MLH1 or MSH2 abnormalities, as certain pathogenic mutations may be associated with retained protein expression. However, PMS2 and MSH6 antibodies have the capability of detecting most abnormalities in MLH1 and MSH2, in addition to detecting mutations in the genes that encode themselves, ie, PSM2 and MSH6 (notably, mutations in MSH6 have been reported to account for up to 13% of families with MMR gene mutations46). Thus, it becomes easy to understand why the studies that used MLH1/MSH2 IHC yielded a lower predictive value than those that included PMS2 and MSH6 (as shown above, under Literature Review). Parenthetically, the secondary nature of MSH6 protein in MutS complexes, ie, the functional redundancy between MSH6 and MSH3, may explain, at least in part, why some MSH6 mutations do not result in high frequency MSI in the tumor.

Advantages of IHC

IHC Is Easily Available; Testing at the Time of Colectomy Is Efficient

As IHC is available as part of the routine service in the general pathology laboratories, it constitutes a convenient technique to general pathologists. This is important because pathologists are at the forefront in recognizing HNPCC. The lack of a premorbid clinical phenotype (hence the difficulty to identify it clinically), coupled with the presence of specific histological patterns in its cancers, renders HNPCC a disease that is often first recognized by the pathologists. Indeed, the revised Bethesda Guidelines have defined an important role for pathologists.3 For colorectal cancer patients age 50 to 60 years, the Guidelines recommend that further testing be done when the tumor morphology is suggestive as determined by the pathologist. Currently, the Guidelines are defined to select tumors for “MSI testing.” With today's knowledge about IHC, it is foreseeable that the next version will state that the Guidelines are to select tumors for “either IHC or MSI testing.” In that scenario (IHC as the standard first-line screening tool), an at-risk colorectal cancer patient will be able to obtain the information about the MMR proteins from the routine pathology report at the time of colectomy. When the patient reaches the Clinical Genetics Service, the next test would be germline mutation on the gene indicated by IHC. There is no longer any need to retrospectively retrieve the paraffin tumor material. Such an approach should be applicable to the majority of at-risk patients, restricting the need for MSI testing to only the rare cases in which IHC is not informative and yet there is a clinical suspicion.

IHC Is in General Regarded As an Inexpensive Technique

The cost of IHC and MSI testing may vary among different countries and health care systems. Earlier analysis indicated that IHC was about threefold less expensive than MSI testing.18 Such estimation, however, may no longer be accurate today as newer techniques are being used. Indeed, the cost of MSI can be reduced by new methods such as fluorescent multiplex PCR-capillary electrophoresis. The cost of IHC may potentially be further reduced as well. We have recently proposed a new two-antibody panel (composed of PMS2 and MSH6) to replace the current four-antibody panel for IHC use as a first-line screening tool.45 Such a proposal is based on the biochemical properties of MMR proteins as described above, and data from the literature and our own work indicating that PMS2 and MSH6 have the ability to predict mutations on all four MMR genes. According to this proposal, in cases in which abnormality is detected by the two-antibody panel, a second-step IHC using additional antibodies (MLH1 in the case of PMS2 abnormality and MSH2 in the case of MSH6 abnormality) may be performed to pinpoint further the gene for mutation testing. (It is expected that the majority of cases picked up by PMS2 will more likely have a mutation in MLH1 than in PMS2, and the majority of cases picked up by MSH6 will more likely have a mutation in MSH2.) In cases where no abnormality is detected by PMS2 or MSH6, no further IHC testing would be necessary. This implies that the majority of colorectal cancer patients who fulfill the revised Bethesda Guidelines but do not harbor MMR deficiency (estimated to be about 70%)28 could have been spared half the IHC work and yet still have the ability to achieve the same predictive value from this test.

IHC Helps Identify the Mutated Gene

IHC reveals which particular MMR gene may be defective, and as such it enables efficient mutation analysis on the target gene. Such ability is not possessed by MSI testing. The ability of IHC to identify the mutated gene also encourages the use of alternative procedures in cases in which standard methods fail. For example, by applying the multiplex ligation-dependent probe amplification method to tumors that showed abnormal PMS2 IHC, Halvarsson et al47 identified a deletion of exons 14 to 19 of MLH1 that was missed by conventional sequencing.

IHC May Detect MMR-Deficient Cases That Can Potentially Be Missed by MSI Testing

As noted by our literature review (Table 2), mutations in MSH6 tend to result in weaker or no MSI in the tumors, a phenomenon also well demonstrated both by cell line studies48,49 and by studies with MSH6-mutant mice.50 Such MSH6 cases may be missed by MSI testing but can be detectable by MSH6 IHC.

Limitations of IHC

IHC with MLH1 Antibody Alone Has a Low Sensitivity in Detecting Mutation of MLH1

Early IHC work noted a low sensitivity of MLH1 IHC, and this was regarded as a major drawback of IHC. However, as demonstrated in the more recent studies and discussed above, such a drawback is largely overcome by the addition of PMS2 to the IHC antibody panel.

IHC Staining Pattern May Vary, Resulting in Uncertainty in Interpretation

A limitation that has drawn much attention is the IHC staining quality.32 Three major patterns have caused much confusion: i) focal staining (with or without weakened intensity); ii) lack of positive internal control, mostly in negatively stained tumors; and iii) cytoplasmic staining. These patterns are also referred to as “weak staining,” “heterogeneous staining,” or “clonal pattern” in the literature.

Focal staining, often with weak intensity, with or without positive internal control in the negatively stained regions, is a pattern most commonly seen with MLH1. This pattern has been suggested to reflect certain types of gene mutation.32 Careful analyses, however, fail to support such an association, as its occurrence spans the mutation spectrum27 and can be seen in mutation-negative cases as well.29 Thus, alternative mechanisms have been suggested and include tumor microenvironment and tissue preservation. It is interesting to note here that tissue hypoxia and oxidative stress have been demonstrated to impair MMR function in genetically MMR-proficient tissues.51,52 As such, regional hypoxia in a tumor could potentially be a reason for regional staining to be lost or weak. Better tissue preservation could potentially reduce such effects of tissue microenvironment and improve the performance of IHC.

Focal staining with unimpaired intensity is a pattern that seems to be more frequently seen with MSH6. In such cases, the areas of tumor cell staining can be very focal, sometimes, constituting only <10% of the tumor. Positive internal control is often present throughout the tumor. The mechanism for this phenomenon is unclear. Anecdotally, we have encountered such extremely focal staining for MSH6 in MLH1-deficient tumors (cases initially misinterpreted as concurrent loss of MLH1/MSH6). Such experience prompted us to speculate that secondary mutation in MSH6 in subclonal populations of an MSI tumor might be a potential explanation (as discussed above, under The Biological and Biochemical Basis of How IHC Works in MMR-Deficient Tumors).

The second staining pattern of concern, lack of convincing positive internal control in an otherwise IHC-negative (protein lost) case, is again not well understood from a mechanistic point of view. Such cases had been regarded as “staining un-interpretable” and therefore excluded from analysis in some early IHC studies.29,32 It has been our experience that, on careful examination or repeated staining in such cases, at least some positive staining can be revealed in the background benign cells, whereas the tumor cells remain negative. Therefore, these cases are probably best regarded as IHC-abnormal before more definitive data become available. Recently, we have encountered a case of an endometrial carcinoma in which the stain for MSH6 demonstrated this very staining variation and the patient was found to have a pathogenic germline mutation on MSH6. The MSI testing in this case did not show instability in any of the five standardized markers.

The third staining variation, cytoplasmic staining, bears no known significance. There are no data as yet to indicate that its presence is reflective of protein deficiency.

Although these various staining patterns exist, with experience, an accurate interpretation of IHC staining is still easily achievable, especially in colorectal tumor specimens. In general, the presence of nuclear staining in the tumor cells, even when it is focal and weak, is good evidence of retained MMR protein. In the rare situation where there is lack of positive internal control in an otherwise IHC-negatively stained tumor, we recommend repeating the stain in search for positive stromal cells. If such repeated attempts still fail to reveal positive stromal cells (a scenario rare in colorectal cancer cases in our experience), as long as the tumor cells remain negative, this tumor is best regarded as IHC abnormal or inconclusive.

IHC May Not Be Reliable in Small Biopsy Samples

Given that IHC staining for MMR protein can be focal, its performance may be affected by tissue sampling. Specifically, lack of staining in a small biopsy sample may not be reliably interpreted as loss of protein in the entire tumor, thus limiting the utility of IHC in biopsy specimens. Specific data to demonstrate such an effect of sampling in biopsy specimens, however, are scare in the literature. Work in this area is ongoing at our institution.

IHC Results May Be Interpreted As “Genetic Information,” Which Can Complicate the Use of IHC as a First-Line Screening Tool

Although loss of MLH1 is a frequent occurrence in sporadic MSI colorectal cancers, loss of the other MMR proteins most often indicates an underlying germline defect. Therefore, although no germline testing has been performed, genetic information can be gleaned from IHC testing (that is not available from MSI testing). As it is the general perception that reporting of genetic information requires patient consent, IHC testing can therefore be construed as medicolegally relevant if it is to be initiated by pathologists who do not have the ability to directly obtain patient consent. Such an issue deserves our attention, and may be resolved by good communication between the pathologists and surgeons or clinicians. An agreement needs to be reached whereby the surgeons and clinicians will discuss IHC tests with their patients who fall in a defined high-risk category for which IHC will be routinely performed and obtain patient consent before the pathology material reaches the pathology department. Under such an agreement, the pathologists can then automatically perform the IHC in predefined patient populations.

The Sensitivity of IHC Is Dependent on Its Antibody Panel

Although the genes covered by the four IHC antibodies are believed to account for the majority of hereditary MMR-deficient cases, other putative MMR genes exist that could potentially be pathogenic as well. Unless new antibodies become available, IHC will not be able to detect such other genes. In contrast, MSI testing has the potential to predict pathogenic abnormalities in any MMR gene (although exceptions are already known to exist, such as the MSH6 mutation cases as discussed above). Vasen et al53 noted that, as long as the role of other putative MMR genes has not been elucidated, IHC analysis cannot completely replace MSI. This is true particularly from a research and investigational point of view.

Summary

In summary, limitations to IHC do exist. Most limitations, however, can be remedied by an appropriate IHC antibody panel, optimized laboratory procedures, and knowledge and experience in MMR IHC interpretation. The inability of IHC to reliably detect MMR abnormality in biopsy specimens, should it be proven by further data, however, would represent a true drawback, as diagnosis at the time of biopsy could significantly enhance the management decision-making (eg, segmental resection versus subtotal colectomy). (In this scenario, however, MSI testing is not superior to IHC; the presence of MSI in a small biopsy would be equally, or even more, unreliable.) In addition, it is to be recognized that on the one hand, IHC has the ability to pinpoint the specific gene and therefore it may be construed as a “genetic test” that carries medicolegal implications; on the other hand, IHC's ability is dependent on its antibody panel, and it may miss cases in which the fundamental defect resides in other untested genes.

Conclusion And Perspectives

Nearly 100 years after a pathologist, Warthin, first recognized the clinical condition of what is now known as HNPCC,2 we are finally able to pinpoint the specific germline mutation in at least a significant proportion of these patients and families. Of the two techniques—MSI testing and IHC (with antibodies including PMS2 and MSH6)—that are commonly evaluated as screening tools for identification of HNPCC, our literature review demonstrates a virtually equivalent informative value in predicting germline mutation (IHC may even be superior in the case of MSH6 mutation). Under such a premise, given that IHC is more easily available and in general inexpensive, that it also detects the mutated gene, and that its limitations can be largely overcome by the pathologists' experience, it is our view that IHC should be placed as the first choice for first-line screening to identify patients for genetic testing. However, such a strategy requires that both the pathologists and clinicians are cognizant of the fact that genetic information can be gleaned from IHC testing, and that appropriate procedures need to be established to ensure patients' understanding and consent. Meanwhile, MSI testing is not to be abandoned by any means, as it can provide a fallback position in situations where IHC is normal and yet clinical suspicion exists, or IHC is inconclusive. MSI testing will also remain a significant research tool for discovering new MMR gene abnormalities.

Notably, amid all constructive debate on the utility of IHC versus MSI testing, there is a constant improvement of laboratory techniques and emergence of new knowledge and new methods. For example, tests on MLH1 promoter methylation and B-raf mutation have been shown to be useful in distinguishing sporadic MSI cases from HNPCC. It is hopeful that in the future, tests such as IHC and MSI (or even mutation testing) will become equally easily accessible and affordable; and detection of HNPCC cancers and mutation carriers will be as simple as diagnosing any other routine pathological entity.

References

  • 1.Lynch HT, Krush AJ. Cancer family “G” revisited: 1895–1970. Cancer. 1971;27:1505–1511. doi: 10.1002/1097-0142(197106)27:6<1505::aid-cncr2820270635>3.0.co;2-l. [DOI] [PubMed] [Google Scholar]
  • 2.Classics in oncology. Heredity with reference to carcinoma as shown by the study of the cases examined in the pathological laboratory of the University of Michigan, 1895–1913. By Aldred Scott Warthin. 1913. CA Cancer J Clin. 1985;35:348–359. doi: 10.3322/canjclin.35.6.348. [DOI] [PubMed] [Google Scholar]
  • 3.Umar A, Boland CR, Terdiman JP, Syngal S, de la Chapelle A, Ruschoff J, Fishel R, Lindor NM, Burgart LJ, Hamelin R, Hamilton SR, Hiatt RA, Jass J, Lindblom A, Lynch HT, Peltomaki P, Ramsey SD, Rodriguez-Bigas MA, Vasen HF, Hawk ET, Barrett JC, Freedman AN, Srivastava S. 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]
  • 4.Hampel H, Frankel WL, Martin E, Arnold M, Khanduja K, Kuebler P, Nakagawa H, Sotamaa K, Prior TW, Westman J, Panescu J, Fix D, Lockman J, Comeras I, de la Chapelle A. Screening for the Lynch syndrome (hereditary nonpolyposis colorectal cancer) N Engl J Med. 2005;352:1851–1860. doi: 10.1056/NEJMoa043146. [DOI] [PubMed] [Google Scholar]
  • 5.Vasen HF, Moslein G, Alonso A, Bernstein I, Bertario L, Blanco I, Burn J, Capella G, Engel C, Frayling I, Friedl W, Hes FJ, Hodgson S, Mecklin JP, Moller P, Nagengast F, Parc Y, Renkonen-Sinisalo L, Sampson JR, Stormorken A, Wijnen J. Guidelines for the clinical management of Lynch syndrome (hereditary non-polyposis cancer) J Med Genet. 2007;44:353–362. doi: 10.1136/jmg.2007.048991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Fishel R, Lescoe MK, Rao MR, Copeland NG, Jenkins NA, Garber J, Kane M, Kolodner R. The human mutator gene homolog MSH2 and its association with hereditary nonpolyposis colon cancer. Cell. 1993;75:1027–1038. doi: 10.1016/0092-8674(93)90546-3. [DOI] [PubMed] [Google Scholar]
  • 7.Leach FS, Nicolaides NC, Papadopoulos N, Liu B, Jen J, Parsons R, Peltomaki P, Sistonen P, Aaltonen LA, Nystrom-Lahti M, Guan XY, Zhang J, Meltzer PS, Yu JW, Kao FT, Chen DJ, Cerosaletti KM, Fournier REK, Todd S, Lewis T, Leach RJ, Naylor SL, Weissenbach J, Mecklin JP, Järvinen H, Petersen GM, Hamilton SR, Green J, Jass J, Watson P, Lynch HT, Trent JM, de la Chapelle A, Kinzler KW, Vogelstein B. Mutations of a mutS homolog in hereditary nonpolyposis colorectal cancer. Cell. 1993;75:1215–1225. doi: 10.1016/0092-8674(93)90330-s. [DOI] [PubMed] [Google Scholar]
  • 8.Lindblom A, Tannergard P, Werelius B, Nordenskjold M. Genetic mapping of a second locus predisposing to hereditary non-polyposis colon cancer. Nat Genet. 1993;5:279–282. doi: 10.1038/ng1193-279. [DOI] [PubMed] [Google Scholar]
  • 9.Ionov Y, Peinado MA, Malkhosyan S, Shibata D, Perucho M. Ubiquitous somatic mutations in simple repeated sequences reveal a new mechanism for colonic carcinogenesis. Nature. 1993;363:558–561. doi: 10.1038/363558a0. [DOI] [PubMed] [Google Scholar]
  • 10.Wilson TM, Ewel A, Duguid JR, Eble JN, Lescoe MK, Fishel R, Kelley MR. Differential cellular expression of the human MSH2 repair enzyme in small and large intestine. Cancer Res. 1995;55:5146–5150. [PubMed] [Google Scholar]
  • 11.Leach FS, Polyak K, Burrell M, Johnson KA, Hill D, Dunlop MG, Wyllie AH, Peltomaki P, de la Chapelle A, Hamilton SR, Kinzler KW, Vogelstein B. Expression of the human mismatch repair gene hMSH2 in normal and neoplastic tissues. Cancer Res. 1996;56:235–240. [PubMed] [Google Scholar]
  • 12.Salahshor S, Koelble K, Rubio C, Lindblom A. Microsatellite instability and hMLH1 and hMSH2 expression analysis in familial and sporadic colorectal cancer. Lab Invest. 2001;81:535–541. doi: 10.1038/labinvest.3780262. [DOI] [PubMed] [Google Scholar]
  • 13.Soreide K. Molecular testing for microsatellite instability and DNA mismatch repair defects in hereditary and sporadic colorectal cancers—ready for prime time? Tumour Biol. 2007;28:290–300. doi: 10.1159/000110427. [DOI] [PubMed] [Google Scholar]
  • 14.de Jong AE, van Puijenbroek M, Hendriks Y, Tops C, Wijnen J, Ausems MG, Meijers-Heijboer H, Wagner A, van Os TA, Brocker-Vriends AH, Vasen HF, Morreau H. Microsatellite instability, immunohistochemistry, and additional PMS2 staining in suspected hereditary nonpolyposis colorectal cancer. Clin Cancer Res. 2004;10:972–980. doi: 10.1158/1078-0432.ccr-0956-3. [DOI] [PubMed] [Google Scholar]
  • 15.Southey MC, Jenkins MA, Mead L, Whitty J, Trivett M, Tesoriero AA, Smith LD, Jennings K, Grubb G, Royce SG, Walsh MD, Barker MA, Young JP, Jass JR, St John DJ, Macrae FA, Giles GG, Hopper JL. Use of molecular tumor characteristics to prioritize mismatch repair gene testing in early-onset colorectal cancer. J Clin Oncol. 2005;23:6524–6532. doi: 10.1200/JCO.2005.04.671. [DOI] [PubMed] [Google Scholar]
  • 16.Lagerstedt Robinson K, Liu T, Vandrovcova J, Halvarsson B, Clendenning M, Frebourg T, Papadopoulos N, Kinzler KW, Vogelstein B, Peltomaki P, Kolodner RD, Nilbert M, Lindblom A. Lynch syndrome (hereditary nonpolyposis colorectal cancer) diagnostics. J Natl Cancer Inst. 2007;99:291–299. doi: 10.1093/jnci/djk051. [DOI] [PubMed] [Google Scholar]
  • 17.Marcus VA, Madlensky L, Gryfe R, Kim H, So K, Millar A, Temple LK, Hsieh E, Hiruki T, Narod S, Bapat BV, Gallinger S, Redston M. Immunohistochemistry for hMLH1 and hMSH2: a practical test for DNA mismatch repair-deficient tumors. Am J Surg Pathol. 1999;23:1248–1255. doi: 10.1097/00000478-199910000-00010. [DOI] [PubMed] [Google Scholar]
  • 18.Debniak T, Kurzawski G, Gorski B, Kladny J, Domagala W, Lubinski J. Value of pedigree/clinical data, immunohistochemistry and microsatellite instability analyses in reducing the cost of determining hMLH1 and hMSH2 gene mutations in patients with colorectal cancer. Eur J Cancer. 2000;36:49–54. doi: 10.1016/s0959-8049(99)00208-7. [DOI] [PubMed] [Google Scholar]
  • 19.Dieumegard B, Grandjouan S, Sabourin JC, Le Bihan ML, Lefrere I, Bellefqih, Pignon JP, Rougier P, Lasser P, Benard J, Couturier D, Bressac-de Paillerets B. Extensive molecular screening for hereditary non-polyposis colorectal cancer. Br J Cancer. 2000;82:871–880. doi: 10.1054/bjoc.1999.1014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Cunningham JM, Kim CY, Christensen ER, Tester DJ, Parc Y, Burgart LJ, Halling KC, McDonnell SK, Schaid DJ, Walsh Vockley C, Kubly V, Nelson H, Michels VV, Thibodeau SN. The frequency of hereditary defective mismatch repair in a prospective series of unselected colorectal carcinomas. Am J Hum Genet. 2001;69:780–790. doi: 10.1086/323658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Stone JG, Robertson D, Houlston RS. Immunohistochemistry for MSH2 and MHL1: a method for identifying mismatch repair deficient colorectal cancer. J Clin Pathol. 2001;54:484–487. doi: 10.1136/jcp.54.6.484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Terdiman JP, Gum JR, Jr, Conrad PG, Miller GA, Weinberg V, Crawley SC, Levin TR, Reeves C, Schmitt A, Hepburn M, Sleisenger MH, Kim YS. Efficient detection of hereditary nonpolyposis colorectal cancer gene carriers by screening for tumor microsatellite instability before germline genetic testing. Gastroenterology. 2001;120:21–30. doi: 10.1053/gast.2001.20874. [DOI] [PubMed] [Google Scholar]
  • 23.Furukawa T, Konishi F, Shitoh K, Kojima M, Nagai H, Tsukamoto T. Evaluation of screening strategy for detecting hereditary nonpolyposis colorectal carcinoma. Cancer. 2002;94:911–920. [PubMed] [Google Scholar]
  • 24.Wahlberg SS, Schmeits J, Thomas G, Loda M, Garber J, Syngal S, Kolodner RD, Fox E. Evaluation of microsatellite instability and immunohistochemistry for the prediction of germ-line MSH2 and MLH1 mutations in hereditary nonpolyposis colon cancer families. Cancer Res. 2002;62:3485–3492. [PubMed] [Google Scholar]
  • 25.Hendriks Y, Franken P, Dierssen JW, De Leeuw W, Wijnen J, Dreef E, Tops C, Breuning M, Brocker-Vriends A, Vasen H, Fodde R, Morreau H. Conventional and tissue microarray immunohistochemical expression analysis of mismatch repair in hereditary colorectal tumors. Am J Pathol. 2003;162:469–477. doi: 10.1016/S0002-9440(10)63841-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Hoedema R, Monroe T, Bos C, Palmer S, Kim D, Marvin M, Luchtefeld M. Genetic testing for hereditary nonpolyposis colorectal cancer. Am Surg. 2003;69:387–391. discussion 391–382. [PubMed] [Google Scholar]
  • 27.Mangold E, Pagenstecher C, Friedl W, Fischer HP, Merkelbach-Bruse S, Ohlendorf M, Friedrichs N, Aretz S, Buettner R, Propping P, Mathiak M. Tumours from MSH2 mutation carriers show loss of MSH2 expression but many tumours from MLH1 mutation carriers exhibit weak positive MLH1 staining. J Pathol. 2005;207:385–395. doi: 10.1002/path.1858. [DOI] [PubMed] [Google Scholar]
  • 28.Pinol V, Castells A, Andreu M, Castellvi-Bel S, Alenda C, Llor X, Xicola RM, Rodriguez-Moranta F, Paya A, Jover R, Bessa X. Accuracy of revised Bethesda guidelines, microsatellite instability, and immunohistochemistry for the identification of patients with hereditary nonpolyposis colorectal cancer. JAMA. 2005;293:1986–1994. doi: 10.1001/jama.293.16.1986. [DOI] [PubMed] [Google Scholar]
  • 29.Shia J, Klimstra DS, Nafa K, Offit K, Guillem JG, Markowitz AJ, Gerald WL, Ellis NA. Value of immunohistochemical detection of DNA mismatch repair proteins in predicting germline mutation in hereditary colorectal neoplasms. Am J Surg Pathol. 2005;29:96–104. doi: 10.1097/01.pas.0000146009.85309.3b. [DOI] [PubMed] [Google Scholar]
  • 30.Barnetson RA, Tenesa A, Farrington SM, Nicholl ID, Cetnarskyj R, Porteous ME, Campbell H, Dunlop MG. Identification and survival of carriers of mutations in DNA mismatch-repair genes in colon cancer. N Engl J Med. 2006;354:2751–2763. doi: 10.1056/NEJMoa053493. [DOI] [PubMed] [Google Scholar]
  • 31.Niessen RC, Berends MJ, Wu Y, Sijmons RH, Hollema H, Ligtenberg MJ, de Walle HE, de Vries EG, Karrenbeld A, Buys CH, van der Zee AG, Hofstra RM, Kleibeuker JH. Identification of mismatch repair gene mutations in young patients with colorectal cancer and in patients with multiple tumours associated with hereditary non-polyposis colorectal cancer. Gut. 2006;55:1781–1788. doi: 10.1136/gut.2005.090159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Shia J, Ellis NA, Klimstra DS. The utility of immunohistochemical detection of DNA mismatch repair gene proteins. Virchows Arch. 2004;445:431–441. doi: 10.1007/s00428-004-1090-5. [DOI] [PubMed] [Google Scholar]
  • 33.Peltomaki P, Vasen H. Mutations associated with HNPCC predisposition—update of ICG-HNPCC/INSiGHT mutation database. Dis Markers. 2004;20:269–276. doi: 10.1155/2004/305058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Christensen M, Katballe N, Wikman F, Primdahl H, Sorensen FB, Laurberg S, Orntoft TF. Antibody-based screening for hereditary nonpolyposis colorectal carcinoma compared with microsatellite analysis and sequencing. Cancer. 2002;95:2422–2430. doi: 10.1002/cncr.10979. [DOI] [PubMed] [Google Scholar]
  • 35.Jascur T, Boland CR. Structure and function of the components of the human DNA mismatch repair system. Int J Cancer. 2006;119:2030–2035. doi: 10.1002/ijc.22023. [DOI] [PubMed] [Google Scholar]
  • 36.Fishel R. The selection for mismatch repair defects in hereditary nonpolyposis colorectal cancer: revising the mutator hypothesis. Cancer Res. 2001;61:7369–7374. [PubMed] [Google Scholar]
  • 37.Kadyrov FA, Dzantiev L, Constantin N, Modrich P. Endonucleolytic function of MutLalpha in human mismatch repair. Cell. 2006;126:297–308. doi: 10.1016/j.cell.2006.05.039. [DOI] [PubMed] [Google Scholar]
  • 38.Acharya S, Wilson T, Gradia S, Kane MF, Guerrette S, Marsischky GT, Kolodner R, Fishel R. hMSH2 forms specific mispair-binding complexes with hMSH3 and hMSH6. Proc Natl Acad Sci USA. 1996;93:13629–13634. doi: 10.1073/pnas.93.24.13629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Harfe BD, Minesinger BK, Jinks-Robertson S. Discrete in vivo roles for the MutL homologs Mlh2p and Mlh3p in the removal of frameshift intermediates in budding yeast. Curr Biol. 2000;10:145–148. doi: 10.1016/s0960-9822(00)00314-6. [DOI] [PubMed] [Google Scholar]
  • 40.Koi M, Umar A, Chauhan DP, Cherian SP, Carethers JM, Kunkel TA, Boland CR. Human chromosome 3 corrects mismatch repair deficiency and microsatellite instability and reduces N-methyl-N′-nitro-N-nitrosoguanidine tolerance in colon tumor cells with homozygous hMLH1 mutation. Cancer Res. 1994;54:4308–4312. [PubMed] [Google Scholar]
  • 41.Umar A, Koi M, Risinger JI, Glaab WE, Tindall KR, Kolodner RD, Boland CR, Barrett JC, Kunkel TA. Correction of hypermutability, N-methyl-N′-nitro-N-nitrosoguanidine resistance, and defective DNA mismatch repair by introducing chromosome 2 into human tumor cells with mutations in MSH2 and MSH6. Cancer Res. 1997;57:3949–3955. [PubMed] [Google Scholar]
  • 42.Watanabe Y, Haugen-Strano A, Umar A, Yamada K, Hemmi H, Kikuchi Y, Takano S, Shibata Y, Barrett JC, Kunkel TA, Koi M. Complementation of an hMSH2 defect in human colorectal carcinoma cells by human chromosome 2 transfer. Mol Carcinog. 2000;29:37–49. [PubMed] [Google Scholar]
  • 43.Boland CR, Koi M, Chang DK, Carethers JM. The biochemical basis of microsatellite instability and abnormal immunohistochemistry and clinical behavior in Lynch Syndrome: from bench to bedside. Fam Cancer. 2008;7:41–52. doi: 10.1007/s10689-007-9145-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Chang DK, Metzgar D, Wills C, Boland CR. Microsatellites in the eukaryotic DNA mismatch repair genes as modulators of evolutionary mutation rate. Genome Res. 2001;11:1145–1146. doi: 10.1101/gr.186301. [DOI] [PubMed] [Google Scholar]
  • 45.Shia J, Vakiani E, Guillem J, Tang L, Paty P, Offit K, Klimstra D. Immunohistochemistry (IHC) as first-line screening tool for detecting colorectal cancer (CRC) patients at risk for Lynch syndrome: a 2-antibody panel is as predictive as a 4-antibody panel (Abstr) Mod Pathol. 2008;21(Suppl 1):138A. doi: 10.1097/PAS.0b013e3181b15aa2. [DOI] [PubMed] [Google Scholar]
  • 46.Roncari B, Pedroni M, Maffei S, Di Gregorio C, Ponti G, Scarselli A, Losi L, Benatti P, Roncucci L, De Gaetani C, Camellini L, Lucci-Cordisco E, Tricarico R, Genuardi M, Ponz de Leon M. Frequency of constitutional MSH6 mutations in a consecutive series of families with clinical suspicion of HNPCC. Clin Genet. 2007;72:230–237. doi: 10.1111/j.1399-0004.2007.00856.x. [DOI] [PubMed] [Google Scholar]
  • 47.Halvarsson B, Lindblom A, Rambech E, Lagerstedt K, Nilbert M. The added value of PMS2 immunostaining in the diagnosis of hereditary nonpolyposis colorectal cancer. Fam Cancer. 2006;5:353–358. doi: 10.1007/s10689-006-0005-9. [DOI] [PubMed] [Google Scholar]
  • 48.Umar A, Risinger JI, Glaab WE, Tindall KR, Barrett JC, Kunkel TA. Functional overlap in mismatch repair by human MSH3 and MSH6. Genetics. 1998;148:1637–1646. doi: 10.1093/genetics/148.4.1637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Yamada NA, Castro A, Farber RA. Variation in the extent of microsatellite instability in human cell lines with defects in different mismatch repair genes. Mutagenesis. 2003;18:277–282. doi: 10.1093/mutage/18.3.277. [DOI] [PubMed] [Google Scholar]
  • 50.Edelmann W, Yang K, Umar A, Heyer J, Lau K, Fan K, Liedtke W, Cohen PE, Kane MF, Lipford JR, Yu N, Crouse GF, Pollard JW, Kunkel T, Lipkin M, Kolodner R, Kucherlapati R. Mutation in the mismatch repair gene Msh6 causes cancer susceptibility. Cell. 1997;91:467–477. doi: 10.1016/s0092-8674(00)80433-x. [DOI] [PubMed] [Google Scholar]
  • 51.Chang CL, Marra G, Chauhan DP, Ha HT, Chang DK, Ricciardiello L, Randolph A, Carethers JM, Boland CR. Oxidative stress inactivates the human DNA mismatch repair system. Am J Physiol Cell Physiol. 2002;283:C148–C154. doi: 10.1152/ajpcell.00422.2001. [DOI] [PubMed] [Google Scholar]
  • 52.Bindra RS, Crosby ME, Glazer PM. Regulation of DNA repair in hypoxic cancer cells. Cancer Metastasis Rev. 2007;26:249–260. doi: 10.1007/s10555-007-9061-3. [DOI] [PubMed] [Google Scholar]
  • 53.Vasen HF, Hendriks Y, de Jong AE, van Puijenbroek M, Tops C, Brocker-Vriends AH, Wijnen JT, Morreau H. Identification of HNPCC by molecular analysis of colorectal and endometrial tumors. Dis Markers. 2004;20:207–213. doi: 10.1155/2004/391039. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from The Journal of Molecular Diagnostics : JMD are provided here courtesy of American Society for Investigative Pathology

RESOURCES