Lynch syndrome: clinical, pathological, and genetic insights

13
REVIEW ARTICLE Lynch syndrome: clinical, pathological, and genetic insights Ralph Schneider & Claudia Schneider & Matthias Kloor & Alois Fürst & Gabriela Möslein Received: 25 January 2012 / Accepted: 27 January 2012 / Published online: 24 February 2012 # Springer-Verlag 2012 Abstract Introduction Lynch syndrome as the most common heredi- tary colorectal cancer syndrome and the most common cause of hereditary endometrial cancer is characterized by an autosomal dominant inheritance with a penetrance of 8590%. The molecular genetic underlying mechanism is a mutation in one of the mismatch repair genes. Methods In order to identify patients with Lynch syndrome, a nuclear family history should be ascertained and matched with the Amsterdam criteria. A different approach for identification is the adherence to Bethesda criteria and subsequent testing for microsatellite instability. In patients with unstable tumors as an indicator for mismatch repair deficiency, genetic counseling and mutation analysis are warranted. For families fulfilling the Amsterdam criteria, intensified screening is rec- ommended, even if a pathogenic mutation is not identified. Results Individuals from families with a proven pathogenic mutation that are tested negative are at normal population risk for cancers and may be dismissed from intensified surveillance. Prophylactic surgery in high-risk individuals without neoplasia is not generally recommended. At the time of a colon primary, however, extended surgery should be discussed in the light of a high rate of metachronous cancers. The worries of impairing functional results have now been evaluated in the light of quality of life in a large international cohort. Interestingly, extended (prophylactic) surgery does not lead to inferior quality of life with equal perioperative risks. Conclusions Therefore, taking the risk reduction into account, extended surgery at the time of the first colon primary should at least be discussed, if not recommended. Also, prophylactic hysterectomy and bilateral oophorecto- my at the time of a colorectal primary should be recommen- ded if family planning has been completed. Keywords Lynch syndrome . Prophylactic surgery . Mismatch repair genes . Genetic testing . MSI analysis . Immunohistochemistry Epidemiology Colorectal (CRC), the most common malignancy in Europe, results as an interaction of genetic and environmental factors and can be attributed to three risk categories: sporadic, famil- ial, and hereditary [1]. Sporadic CRC, which accounts for approximately 70% of all CRC, originates from acquired somatic mutations over time and therefore is associated with older age. Approximately 2030% of all CRCs are classified as familial. Polymorphisms and low penetrance susceptibility loci associated with an increased risk of CRC have been identified by genetic association and population studies in these patients [24]. These genetic risk factors combined with R. Schneider Department of Visceral, Thoracic and Vascular Surgery, Philipps University, Marburg, Germany C. Schneider : G. Möslein Department of General and Visceral Surgery, HELIOS St. Josefs-Hospital, Bochum-Linden, Germany M. Kloor Department of Pathology, University of Heidelberg, Heidelberg, Germany A. Fürst Department of Surgery, Caritas Hospital St. Josef, Regensburg, Germany G. Möslein (*) Department of General and Visceral Surgery, Coloproctology, HELIOS St. Josefs-Hospital, Axstrasse 35, 44879 Bochum, Germany e-mail: [email protected] Langenbecks Arch Surg (2012) 397:513525 DOI 10.1007/s00423-012-0918-8

Transcript of Lynch syndrome: clinical, pathological, and genetic insights

REVIEWARTICLE

Lynch syndrome: clinical, pathological, and genetic insights

Ralph Schneider & Claudia Schneider & Matthias Kloor &

Alois Fürst & Gabriela Möslein

Received: 25 January 2012 /Accepted: 27 January 2012 /Published online: 24 February 2012# Springer-Verlag 2012

AbstractIntroduction Lynch syndrome as the most common heredi-tary colorectal cancer syndrome and the most commoncause of hereditary endometrial cancer is characterized byan autosomal dominant inheritance with a penetrance of85–90%. The molecular genetic underlying mechanism isa mutation in one of the mismatch repair genes.Methods In order to identify patients with Lynch syndrome, anuclear family history should be ascertained and matched withthe Amsterdam criteria. A different approach for identificationis the adherence to Bethesda criteria and subsequent testingfor microsatellite instability. In patients with unstable tumorsas an indicator for mismatch repair deficiency, geneticcounseling and mutation analysis are warranted. For familiesfulfilling the Amsterdam criteria, intensified screening is rec-ommended, even if a pathogenic mutation is not identified.

Results Individuals from families with a proven pathogenicmutation that are tested negative are at normal populationrisk for cancers and may be dismissed from intensifiedsurveillance. Prophylactic surgery in high-risk individualswithout neoplasia is not generally recommended. At thetime of a colon primary, however, extended surgery shouldbe discussed in the light of a high rate of metachronouscancers. The worries of impairing functional results havenow been evaluated in the light of quality of life in a largeinternational cohort. Interestingly, extended (prophylactic)surgery does not lead to inferior quality of life with equalperioperative risks.Conclusions Therefore, taking the risk reduction intoaccount, extended surgery at the time of the first colonprimary should at least be discussed, if not recommended.Also, prophylactic hysterectomy and bilateral oophorecto-my at the time of a colorectal primary should be recommen-ded if family planning has been completed.

Keywords Lynch syndrome . Prophylactic surgery .

Mismatch repair genes . Genetic testing .MSI analysis .

Immunohistochemistry

Epidemiology

Colorectal (CRC), the most common malignancy in Europe,results as an interaction of genetic and environmental factorsand can be attributed to three risk categories: sporadic, famil-ial, and hereditary [1]. Sporadic CRC, which accounts forapproximately 70% of all CRC, originates from acquiredsomatic mutations over time and therefore is associated witholder age. Approximately 20–30% of all CRCs are classifiedas familial. Polymorphisms and low penetrance susceptibilityloci associated with an increased risk of CRC have beenidentified by genetic association and population studies inthese patients [2–4]. These genetic risk factors combined with

R. SchneiderDepartment of Visceral, Thoracic and Vascular Surgery,Philipps University,Marburg, Germany

C. Schneider :G. MösleinDepartment of General and Visceral Surgery,HELIOS St. Josefs-Hospital,Bochum-Linden, Germany

M. KloorDepartment of Pathology, University of Heidelberg,Heidelberg, Germany

A. FürstDepartment of Surgery, Caritas Hospital St. Josef,Regensburg, Germany

G. Möslein (*)Department of General and Visceral Surgery, Coloproctology,HELIOS St. Josefs-Hospital,Axstrasse 35,44879 Bochum, Germanye-mail: [email protected]

Langenbecks Arch Surg (2012) 397:513–525DOI 10.1007/s00423-012-0918-8

environmental factors lead to an increased risk of CRCobserved in particular families [5]. Thus, familial CRC canalso be characterized as a susceptibility with a classical mul-tifactorial inheritance. The third category of CRC—hereditaryCRC—accounts for approximately 5% of all CRC and ischaracterized by inherited, highly penetrant mutations [3, 5,6]. Lynch syndrome (LS), which accounts for approximately1–3% of all CRC [7–9] and also for 2% of all endometrialcancers [8], is the most common form of hereditary CRC. Thepopulation risk can only be estimated: approximately 6% ofall persons in Germany (80 million inhabitants) will develop aCRC which corresponds to 4.8 million inhabitants. If 2% ofthese are attributed to hereditary predisposition (a conserva-tive estimate), this would translate to 96,000 affected, insinu-ating that 1.2% of the general population would have apathogenic mutation. Taking into account a penetrance of65% for pathogenic mismatch repair (MMR) mutations(MSH2, MLH1, and MSH6), the frequency of MMR muta-tions would be 1.8% or 1:550.

Terminology

The first description of a family with a hereditary cancerpattern dates back to Aldred Scott Warthin in 1913 whodescribed the family of his seamstress. Henry T. Lynch laterfollowed up on this first family “G” and added observations offurther families, triggered by the consultation of a youngpatient with CRC in the absence of overt polyposis denomi-nating this observation “cancer family syndromes” [10–14].In 1985, the term “hereditary non-polyposis colorectal cancer(HNPCC)” was introduced emphasizing the heritable nature,the predisposition to CRC, and the absence of a widespreadpolyposis [15, 16]. In the following decades, HNPCC and“Lynch syndrome” have been used synonymously [17, 18].Today, there is an agreement on the terminology to classifyfamilies with an identified mutation in one of the mismatchrepair genes as LS, whereas other families fulfilling theBethesda or Amsterdam criteria (Tables 1 and 2) withoutmutation detection are denominated HNPCC [19, 20].

This overlap is a problem since as many as 40% of thepatients meeting the Amsterdam I criteria lack evidence of

hereditary deficiency in mismatch repair genes [21]. Onehundred sixty-one pedigrees fulfilling the Amsterdam I cri-teria were grouped according to the microsatellite instability(MSI) status. Patients with an MSI-high status which wasinterpreted as a surrogate for a germline mismatch repairgene mutation had a standardized incidence ratio for CRC inthe first-degree and second-degree relatives of the patientsof 6.1 and a statistically significant increased risk for thepresentation of LS-associated extracolonic tumors [21]. Incontrast, patients with an MSI-low status or a microsatellitestable tumor had a lower standardized incidence ratio of 2.3and had no increased risk for extracolonic tumors [21].

We strongly encourage the use of Lynch syndrome sincethe term HNPCC is misleading for several reasons [21, 22].HNPCC implies the absence of polyps, although studieshave shown that LS patients harbor similar numbers ofpolyps as the general population [23]. Moreover, the termfails to acknowledge the wide spectrum of frequent extrac-olonic cancers such as cancers of the endometrium, stom-ach, small bowel, ovary, pelviureter, and skin [24].

Clinical insights

Clinical features

LS is characterized by an increased cancer risk at a youngerage than in the sporadic counterpart (Table 3). In theGerman HNPCC Consortium, a total of 1,309 families withan identified mutation have been reported. MLH1 mutationcarriers have a lifetime risk (until age 75) for a CRC of80.1%, followed by a risk of 65.1% for MSH2 carriers. Therisk of CRC in MSH6 carriers in accordance to literaturewas significantly lower being 37.3%. Males have a higherlifelong CRC risk of 73% compared to females with a risk of59%. After 30 years, 54.3% of all CRC patients had devel-oped a metachronous CRC.

Younger age at diagnosis is typical for LS-associatedCRC with a reported mean age of CRC diagnosis rangingfrom 42 to 61 years [25, 26] as compared to 71 years in thegeneral population [27]. The mean age of endometrial

Table 1 Amsterdam I criteria [121]

1. CRC has been diagnosed in at least three relatives.

2. One of them should be a first-degree relative of the other two.

3. At least two successive generations are affected.

4. At least one CRC was diagnosed before the age of 50 years.

5. Familial adenomatous polyposis (FAP) has been excluded.

6. CRC are verified by histopathological examination.

Families must fulfill all criteria

Table 2 Amsterdam II criteria [17]

1. Lynch syndrome-associated cancer has been diagnosed in at leastthree relatives.a

2. One of them should be a first-degree relative of the other two.

3. At least two successive generations are affected.

4. At least one cancer was diagnosed before the age of 50 years.

5. Familial adenomatous polyposis (FAP) has been excluded.

6. Tumors are verified by histopathological examination.

Families must fulfill all criteriaa CRC, cancer of the endometrium, small bowel, ureter, or renal pelvis

514 Langenbecks Arch Surg (2012) 397:513–525

cancer diagnosis is reported to be 47–55 years in somestudies, which is younger than in general population [25,28, 29]. In contrast, others studies found a mean age ofendometrial cancer diagnosis of 62 years, which is the sameas in the general population [27, 30].

Although patients with LS have an earlier age of CRCdiagnosis, they have improved survival compared to spo-radic CRC. This could be demonstrated in a study fromFinland with a one third lower mortality of LS-associatedCRC [31]. In concordance, the 5-year survival rate of LS-associated CRC is reported to be 53% as compared to 35%for sporadic CRC [32]. This effect may be attributed to theobservation that LS-associated cancers have a much lowerdisposition to metastasize.

Further typical features of LS-associated CRC are the slightpredominance of cancers proximal to the left colonic flexureof 56–62% [33, 34] as well as the frequent presentation ofsynchronous and/or metachronous CRC. The frequency ofsynchronous CRC for LS is 18% and of metachronous CRCis 30% at 10 years and 50% at 15 years [35, 36]. In compar-ison, the risk for synchronous and metachronous CRC inpatients with sporadic CRC is 2–4% and 2–3%, respectively[32, 37]. An analysis of 1,381 tumors in LS families revealedCRC as the most frequent malignancy with 78% in MLH1mutation carriers and 65% in MSH2 mutation carriers. Asalready described above, there was a high number of right-sided CRC of 60%. Interestingly, the number of rectal cancerswas 21% in MLH1 mutation carriers and 20% in MSH2mutation carriers [38].

As the presentation of keratoacanthomas or sebaceousskin tumors such as sebaceous adenomas, sebaceous carci-nomas, or sebaceous epitheliomas is fairly common inpatients with LS, all patients with LS have to be consideredat risk for these skin tumors [39–41]. For patients with LS

and the presence of skin tumors, the term “Muir–Torresyndrome” is widely used [40–42].

Identification of patients at risk for Lynch syndrome

Unfortunately, an increased familial risk for cancers per se isnot frequently questioned or acknowledged in clinical (sur-gical) practice. In the event of a colorectal cancer or a cancerin one of the associated target organs, alertness for thecondition is pivotal. Since not all index LS patients areyoung, nuclear family history in all cancer patients mustbe obtained and documented, as a basis for selecting thosepatients. Genetic counseling and perhaps molecular testingshould be recommended. In a clinical setting, the nuclearpedigree suffices. Due to the structure of small families andsocial separation, reliable family information is restricted toclose relatives. If the nuclear pedigree detects a pattern ofhereditary susceptibility, an extended pedigree is essential(Fig. 1). To date, it is recommended to test for mismatchrepair deficiency [MSI or immunohistochemistry (IHC)] inneoplastic lesions from affected persons whenever possible.The Bethesda criteria have clearly been set up as a guidelineto decide when to perform this testing in order to thenrecommend mutation analysis for microsatellite instable ormismatch repair-deficient tumors in index persons. For sin-gle patients, that appear to have an underlying geneticpredisposition. The Bethesda Guidelines including patho-logical and morphological characteristics were published in1997 to detect patients whose tumors should be analyzed formicrosatellite instability [43]. In 2004, the revised BethesdaGuidelines were published (Table 4) [18] which have asensitivity of approximately 90% [7–9, 26, 44–48].

MSI testing can either be performed via PCR or muchmore economical in the immunohistochemical assessmentof loss of protein expression in one of the mismatch repairgenes (IHC) as a standardized procedure in pathologicalinstitutes. The additional benefit of IHC lies in the fact thatloss of expression in one of the genes pinpoints towards theunderlying gene disabled, enabling more directed sequenceanalysis. Due to economic constraints, it does not seenreasonable to test all patients with CRC as a non-directedgermline mutation detection exercise with costs involvingapproximately US $1,000 per gene with four genes requir-ing testing [24, 49]. However, since sequencing costs arerapidly decreasing and logistics rapidly evolving, it isenvisaged that broader testing will be available in the nearfuture at an affordable cost. Nevertheless, until today, theAmsterdam II criteria especially, taking associated extraco-lonic cancers into regards, have proven to be clinically mostvaluable in identifying LS families.

The Amsterdam II criteria (Table 2) were published in1999 [17]. However, many families with LS do not meetthese criteria due to small family size, late onset of the

Table 3 Cumulative lifetime risks for patients with Lynch syndrome[26–30, 39, 40, 122–127]

Cancer Lynchsyndrome (%)

Generalpopulation (%)

CRC—male 54–74 5CRC—female 30–52

Endometrium 28–60 2

Ovarian 6–7 1

Stomach 6–9 <1

Small bowel 3–4 <1

Pancreatic <1–4 1

Hepatobiliary 1 Rare

Urinary tract 3–8 Rare

Brain 2–3 <1

Sebaceous skin tumor/keratoacanthoma

1–9 Rare

Langenbecks Arch Surg (2012) 397:513–525 515

disease, young age of death due to other causes, or adoption[50]. Others reasons for reduced sensitivity may be non-

paternity, new mutations as well as reduced penetrance [18].Moreover, recording an accurate family history is often verydifficult in clinical practice since patients often have limitedknowledge of their family history [51–54].

Pathological insights

Histology

The first systematic histological evaluation of LS-associatedCRC revealed an increased incidence of mucinous cancersin 39% of tumors compared to 20% in sporadic controlswith more poorly differentiated tumors (24% versus 12%)[23]. Based on this observation, several tumor types andhistological features have been described to be common inLS-associated CRC.

Characteristic tumor types include mucinous cancer cellscomposed of more than 50%mucin and signet ring cell cancercontaining more than 50% signet ring cells [24]. The thirdmost common tumor types were medullary carcinomas [24].

Fig. 1 Pedigree of a Lynch syndrome family

Table 4 Revised Bethesda Guidelines [18]

1. CRC has been diagnosed before the age of 50 years.

2. Presence of synchronous, metachronous CRC or other Lynchsyndrome associated tumorsa, regardless of age.

3. CRC with MSI-H histologyb diagnosed in a patients who is less than60 years of age.

4. CRC diagnosed in a patient with one or more first-degree relativeswith Lynch syndrome associated tumor, with one of the cancersbeing diagnosed before the age of 50 years.

5. CRC diagnosed in a patient with two or more first- or second-degreerelatives with Lynch syndrome associated tumors, regardless of age.

Fulfillment of only one criterion necessary to warrant MSI testinga Endometrial, stomach, ovarian, pancreas, biliary tract, small intestine,brain tumors (usually glioblastoma in Turcot syndrome), sebaceous glandadenomas and keratoacanthomas in Muir–Torre syndrome, hepatobiliary,transitional cell carcinoma of renal pelvis or ureterb Presence of tumor-infiltrating lymphocytes, Crohn’s-like lymphocyticreaction, mucinous/signet ring differentiation, or medullary growth pattern

516 Langenbecks Arch Surg (2012) 397:513–525

In all of these, neoplasia tumor-infiltrating lymphocytes,which largely consist of CD3/CD8 coexpressing cytotoxic Tcells and peritumoral lymphocytes have been a very commonfeature. These “Crohn’s-like” lesions are prominent nodularlymphoid aggregates at the infiltrating edge of the tumor [24,55–58]. A poor differentiation and tumor heterogeneity areadditional characteristic features of LS-associated CRC [23,33, 55–57, 59–64].

Immunohistochemistry

If a patient fulfills the Amsterdam or Bethesda criteria,testing for microsatellite instability via PCR or IHC stainingof the tumor is recommended [18]. A negative IHC in one ormore of the mismatch repair genes is caused by a geneticevent in the particular gene and is indicative for the defec-tive gene. As further described below, a large number of lossof expression is due to promoter methylation and not mis-match repair deficiency. Particularly for the other mismatchrepair genes, molecular genetic testing can be focused on thegene with the loss of protein expression [27].

However, 10% of patients with LS do not demonstrateloss of protein expression of the mismatch repair genes[65–67]. The reasons may be a lack of standardization infixatives of material, a varying ability in performing andinterpreting IHC staining results, and also differences in thestaining protocol (lack of expertise or standardization,insufficient protocol, historical blocks, and type of fixation)[68].

Genetic insights

Mismatch repair genes

The MMR system corrects errors in DNA replication. It is acombination of proteins, specifically MLH1, MSH2, MSH6,and PMS2, whereby MLH1 and PMS2 as well as MSH2and MSH6 work as partners at the binding site. A deficiencyin the MMR system leads to increased mutability in onco-genes and tumor suppressor genes, leading to the clinicalexpression (phenotype) of LS. The estimated 2–5% of allCRCs [26] are attributed to LS, although this may still be anunderestimate. MLH1 and MSH2 are the predominantlyinvolved genes notably leading to nonsense or frameshiftmutations.

The loss of function of the MMR system is not complete,since the second (wild type) allele further encodes intactMMR proteins. In malignant tumors in LS patients, thesecond gene is somatically mutated [69]. Adenomas in LSpatients demonstrate a complete loss of one of the MMRproteins in 66% of cases, with a higher percentage in prox-imal and larger adenomas [70].

Microsatellite instability

Microsatellites are repetitive DNA sequences throughout thewhole genome. Due to these frequent repetitions, micro-satellites are liable for errors during DNA replication—errors that are corrected by an intact mismatch repair sys-tem. Alterations in the length of the microsatellite sequenceslead to MSI. As microsatellites are also present in genesinvolved in the regulation of cell growth, their dysfunctionmay cause tumorigenesis. MSI-unstable tumors account forapproximately 15–20% of unselected CRC cases [59] andare the underlying pathogenic mechanism for the vastmajority of CRC in LS patients [71].

Testing for MSI (via PCR) requires a comparisonbetween healthy and tumor tissue due to individual variabil-ity of microsatellite size and can be performed in fresh aswell as in a formalin-fixed specimens. Criteria and panels todefine microsatellite instability-high (MSI-H), microsatelliteinstability-low (MSI-L), and microsatellite stable (MSS)were defined on two NCI workshops [18, 72]. However,to date, the clinical significance of MSI-L remains unclear.While MSI colon tumors generally have an overall betterprognosis [73], it is still unclear, whether they have a worseresponse to 5-fluorouracil [74, 75].

The domain of MSI testing is to prescreen for LSpatients. However, in the past years, IHC loss of proteinexpression in one of the known mismatch repair genesidentifies reliably the vast majority of unstable tumors at amuch lower cost. Additionally, this approach reduces theeffort of sequencing, since the gene that showed loss ofexpression can be prioritized for sequence analysis. Never-theless, it is necessary to distinguish clearly between LSpatients and patients with BRAF-V600 mutation.

Promoter hypermethylation/BRAF analysis

CRC evolves via three different pathways. Approximately85% of unselected CRC show chromosomal instability andconsecutive loss of genes like APC or p53 and are predomi-nantly microsatellite stable MSS. Only a minority of the MSItumors are due to inactive MMR genes caused by germlinemutation in LS [76, 77], called MSI pathway. Most sporadicMSI tumors follow the serrated pathway (approximately 12%of all CRC and 75% ofMSI tumors), characterized by biallelicloss of the MMR system, due to hypermethylation of thepromoter region of the MLH1 gene [78, 79]. Sessile serratedadenomas are regarded as precursor lesions for these sporadicMSI cancers [80, 81]. The serrated pathway accounts for up to30% of all CRC [82].

A characteristic feature of serrated pathway is methylationof cytosine–guanosine dinucleotides, so-called CpG islands.If located in the promoter region of genes, methylation causesdownregulation of the affected genes, leading to the so-called

Langenbecks Arch Surg (2012) 397:513–525 517

CpG island methylator phenotype (CIMP) [83, 84]. Thehypermethylation affects tumor suppressor genes, characteris-tically MLH1, leading to inactivation and consecutively loss ofexpression in IHC. BRAF mutations are significantly associ-ated with CIMP or the serrated pathway [84]. The BRAF gene,encoding serine/threonine-protein kinase B-Raf, is involved incell growth signaling. BRAF mutations are known to be asso-ciated with different cancers, such as malignant melanoma(40–60%) [85], papillary thyroid cancer (45%) [86], recentlyalso in hairy cell leukemia (100%) [87] and in (MSI) CRC;almost in all cases caused by BRAF-V600E mutation [valine(V) in codon 600 was substituted by glutamate (E)].

The frequency of BRAFmutations in CRC ranges from 5%inMSS to 45% inMSI tumors [88]. BRAF mutations seem tobe associated with a deteriorated outcome in CRC, especiallyin combination with MSS tumors [74, 89, 90]. WhetherBRAF mutation status has an influence on the response tocetuximab remains unclear [91–93]. BRAF-V600E mutationsare common in sporadic MSI colorectal cancer (40–50%), butextremely rare in LS. Therefore, prior testing for BRAF-V600E mutations before sequence analysis is recommendedfor patients with a loss of MLH1 in IHC [94].

Limitations of molecular analysis

MSI testing has its merits as a prescreening for identificationof LS patients, with a reported sensitivity of close to 100%[76], but there are limitations. MSI is still not broadlyavailable and remains costly. In contrast, IHC is availablein all pathologic institutes and is a routine procedure alreadyfor several indicators of prognosis such as response toadjuvant treatment. Also, MSI does not indicate the specificMMR gene involved and further does not distinguish be-tween LS and sporadic MSI cancer arising from BRAF-V600E mutation. The main limitation of IHC testing is thehigh frequency of MLH1 loss in sporadic cancers due topromoter methylation, mostly in old patients.

The gold standard for demonstrating MMR deficiencyremains germline analysis with the identification of a clearlypathogenic mutation in one of the MMR genes. Homozy-gous mutations have been demonstrated and lead to delete-rious outcome at young age. As a standard, full genesequencing has been established for mutation detection innewly identified index patients, that either fulfill Bethesdacriteria and have shown MSI or loss of expression in one ofthe MMR proteins in neoplasia (Fig. 2) or were identifieddue to fulfillment of clinical criteria (Amsterdam criteria). Ifa pathogenic mutation was not identified after sequenceanalysis of four MMR genes (MSH2, MLH1, MSH6, andPMS2), LS cannot be ruled out. However, the opportunity ofpredictive testing for at-risk relatives cannot be offered, imply-ing that 50% of relatives are subjected to intensified screeningprocedures without having an elevated cancer risk.

Genetic counseling considerations for Germany

A new Genetic Diagnostics Act was introduced in Germanyin February 2010. As from February 2012, a new regulationentered into force, determining that special training forgenetic counseling is required—a regulation that affectssurgeons in their practice with hereditary conditions. Aformalized special training will become necessary in orderto perform genetic counseling in case of diagnostic or pre-dictive genetic examinations, for all physicians that are not aspecialist in human genetics (Facharzt für Humangenetik)(§10 Abs. 1 and 2 as well as §7 Abs. 1 and 3 GenDG).

In the absence of such special training courses, a transi-tional period of 5 years has been determined. Within thisperiod, any physician who counsels patients with geneticconditions is required to accomplish an online formalizedinterrogation. A voluntary preparatory class is being estab-lished, but to date, it is not in place.

We recommend formalizing the expertise for surgeonsthat are interested in hereditary conditions, especially sinceour knowledge regarding the recommendations towardsprophylactic surgery on one hand and identification ofunderlying genetic causes on the other is rapidly increasing.In the interest of good clinical practice and patients’ right toreceive adequate counseling prior to surgeries, this aspect isenvisaged to become important for daily surgical routine.After 2016, physicians will be required to absolve a72-h obligatory class prior to a knowledge test in order tolegally counsel patients with genetic conditions, accordingto §10 GenDG.

Screening recommendations

As illustrated in Table 5, LS patients require an intensifiedsurveillance not only for the substantial risk of CRC, butalso for a variety of extracolonic cancers, especially endo-metrial, gastric, and small bowel cancer among others.Recently, unpublished observations report a higher thanestimated rate of cancers of the bladder and breast. Severalguidelines regarding surveillance for LS mutation carriersrecommend slightly different surveillance programs. TheGerman S3 Guideline [95] in comparison with the recom-mendations of the Mallorca Group [96] (European branch ofInSiGHT; www.mallorca-group.eu), the EGAPP [97], andthe NCCN [98] recommendations are shown in Table 5.

Yearly versus two- or three-yearly colonoscopies has notshown any additional benefit to date. Interval cancers –referring to those metachronous cancers arising betweentwo scheduled screening colonoscopies – are the expressionof the biology of LS with a vastly accelerated adenomacarcinoma sequence. The benefit of yearly gastroscopies thathas been established due to a high frequency of gastric

518 Langenbecks Arch Surg (2012) 397:513–525

cancer regardless of familial clustering for this condition hasnot yet been proven to be beneficial but continues to berecommended. Screening for endometrial cancer has beenupgraded towards the truly invasive recommendation ofyearly endometrial biopsies. Since then, more women haverequested prophylactic hysterectomy, which has been rec-ommended as the procedure of choice for women aftercompletion of family planning elsewhere.

Surgical recommendations

Despite regular surveillance, the relative risk ratio of devel-oping cancer (CRC, endometrial and ovarian cancer) is 5.8compared with a mutation negative cohort in a setting withcolonoscopy, transvaginal ultrasound, and endometrial bi-opsy every 2 or 3 years [99]. The rate of interval CRC in thisstudy was 12.4% in 11.5 years, whereas in a Dutch cohort ofLS families with colonoscopy every 1–2 years, the rate ofinterval CRC was 6% in a 10-year follow-up. In a Germanstudy, 19 of 43 interval cancers occurred after a normalsurveillance colonoscopy within the recommended intervalof 12 months. Therefore, the option of extended resection asa subtotal colectomy with an ileosigmoidal anastomosismust be addressed [100].

Despite the high rate of metachronous colon cancers, inguidelines to date, at the most, there is the view of equipoise

regarding the recommended extent of colonic resection (onco-logic segmental versus prophylactically extended subtotal re-section) at the time of first colon cancer. The high risk ofmetachronous cancer after segmental resection must beweighed against a facilitated surveillance (sigmoidoscopy ver-sus colonoscopy) and a supposed worse functional outcomeafter subtotal colectomy [100–102].

Results from a large international study demonstrate anequal postoperative quality of life after subtotal colectomycompared to segmental resection (Schneider et al. “Qualityof life (QOL) in LS patients with colon cancer”, in prepara-tion) showed no impairment after extended (prophylactic)colonic resection. The authors carefully recommend subtotalcolectomy in LS patients at the time of their first coloncancer.

In biopsies of colorectal cancer, MSI can be identified with100% sensitivity and specificity [103]. In colorectal adeno-mas, a combination of MSI and immunohistochemical assaydetects pathological findings in 73% of LS patients [104]. Itmay be concluded that preoperative assessment of colon can-cer patients with suspected LS is easy to perform and hasmajor implications for patients’ right for knowledge andextensive information prior to surgery (Fig. 2). Not taking anunderlying genetic predisposition into regard that may influ-ence therapeutic decisions could be regarded as an offenseagainst patients right “Patientenrechtegesetz” (BürgerlichesGesetzbuch, §630a, Absatz 2). Also, the guidelines of the

Fig. 2 Strategy for CRC patients with a suspected MMR gene defect

Langenbecks Arch Surg (2012) 397:513–525 519

Mallorca Group [96] and others have clearly claimed the needof discussing the option of prophylactic hysterectomy andsalpingo-oophorectomy at the time of abdominal surgery,especially after completion of family planning or olderage.

Effects of aspirin on colorectal neoplasia

Aspirin is known to act as a preventive agent in the devel-opment of sporadic CRC, merely the weighing of this ben-efit versus side effects of the medication impedes the generalrecommendation towards aspirin intake. In a recent meta-analysis of long-term data from five randomized trials ofcardiovascular prevention, aspirin reduced the 20-year riskof colorectal cancer-associated mortality by 35% [105].Overall, only two aspirin studies published failed to dem-onstrate a cancer preventive effect, one of these being theWomen’s Health Study [106]. The interval of aspirin admin-istration has been argued to be more influential on this effectthan the actual doses (daily versus every second day). Insummary, aspirin studies appear to correlate the overallintake of aspirin with a long-term benefit in cancer preven-tion. However, to date, prospective randomized trials withcancer prevention as an endpoint have not been pursued.

The Colorectal Adenoma/Carcinoma Prevention Programme2 (CAPP2) study of aspirin in LS was designed as such a trial.In CAPP2, 937 carriers of LS were randomly assigned to fourgroups in a two-by-two factorial design: high-dose aspirin(600 mg daily) plus resistant starch placebo, resistant starch(30 g) plus aspirin placebo, aspirin plus resistant starch, oraspirin placebo plus starch placebo. The initially reportedanalysis (after a mean 29 months on treatment) suggested thataspirin or resistant starch, or both, did not reduce the risk ofcolorectal neoplasia (adenomas plus colorectal cancer). Burnet al. now most recently reported the long-term (mean56 months) follow-up analysis in 861 individuals. Contrastingstrikingly with the short-term findings, colorectal cancer (notcombined with adenoma) developed in fewer patients withaspirin (4%) than in those on aspirin (7%, hazard ratio 0.63,95% CI 0.35–1.13, p00.12) in an intention to treat analysis;for participants completing 2 years of intervention, the hazardratio in a per protocol analysis was 0.41 (95% CI 0.19–0.86,p00.02). This effect is as preventive for CRC as the yearlycolonoscopies recommended as a screening measure! Also,aspirin reduces approximately 50% of all extracolonic cancersassociated with LS [107]. This study arguably supports moregeneral recommendations to consider aspirin for prevention ofcolorectal cancer in the context of individualized risk–benefitassessments.

The authors recommend intake of low-dose aspirin forLS patients, taking into account the risk–benefit of aspirinversus expected side effects. It is to be expected that theseT

able5

Recom

mendatio

nsforsurveillancein

Lyn

chsynd

romemutationcarriersby

theGerman

S3Guidelin

e[95]

incomparisonwith

therecommendatio

nsof

theMallorcaGroup

[96]

(Europ

ean

branch

ofInSiGHT,

www.m

allorca-grou

p.eu),theEGAPP[97],andtheNCCN

[98]

Colon

oscopy

interval

(years)

Low

erage

limit

Gastroscopy

interval

(years)

Low

erage

limit

Abd

ominal

ultrasou

ndinterval

(years)

Low

erage

limit

Gyn

ecolog

yinterval

Low

eragelim

itOther

S3Guidelin

e1

25a

1b25

125

1,TVU

25Genetic

coun

selin

gat

18yearsold

MallorcaGroup

1–2

20–25

1–2b

30–35

130–35

1–2,

TVU,aspiratio

nbiop

sy30–35

Urine

analysisandcytology,

ifurinarytractcancer

runs

inthefamily

EGAPP

1–2

20–25

1–2,

TVU,endo

metrial

biop

sy30–35

Genetic

coun

selin

g

NCCN

1–2

20–25

c1,

TVU

orendo

metrial

aspirate

TVU

transvaginal

ultrasou

ndaEqu

alsor

atleast5yearsyo

ungerthan

theyo

ungestageat

diagno

sisin

thefamily

bIfcancer

runs

inthefamily

cEqu

alsor

10yearsyo

ungerthan

theyo

ungestageat

diagno

sisin

thefamily

520 Langenbecks Arch Surg (2012) 397:513–525

compelling results will eventually find integration in guide-lines for LS patients.

The use of vaccines in the prevention of colorectal cancer

A typical feature of LS-associated CRC is pronouncedinfiltration with immune cells [108, 109]. This suggestedearly that the host’s antitumoral immune response plays animportant role for the comparatively favorable prognosis ofLS-associated CRC patients [110, 111]. Studies on the anti-gen specificity of the observed immune responses identifiedMSI-associated antigens [112, 113]. These frameshift pep-tide (FSP) antigens directly result from mismatch repairdeficiency-induced frameshift mutations affecting codingmicrosatellite-bearing genes, like TGFBR2, TAF1B, AIM2,and others, and readily elicit antigen-specific cellular andhumoral immune responses of the host [114, 115]. Recentstudies suggested that FSP antigens are promising targets fornovel therapeutic approaches in MSI-H colorectal cancerpatients. Moreover, prophylactic vaccination with FSP anti-gens may be applicable for cancer prevention in LS mutationcarriers in the future. A clinical trial evaluating vaccinationwith FSP antigens has recently been initiated.

Initiation of a surgical Lynch syndrome database

LS is one of the most frequent monogenetic heritable con-ditions, leading to a high rate of colorectal cancer. Unfortu-nately, even to date, most index persons that lead topredisposed families are identified at the time of a colorectalcancer. However, patients are largely underidentified due toa lack of knowledge and understanding of the significanceof the predisposition.

In order to improve awareness and identify and counselmore patients with the hereditary conditions, a surgicalnetwork has been initiated. Patients at the time of theircancer should be counseled clinically and be embedded ingenetic counseling and testing. More information may befound at www.indexfam.de.

Clinical overlap with other predisposing syndromes(FAP + MYH-associated polyposis)

Biallelic germline mutations in the mutY human homologue(MYH) gene lead to the autosomal recessive MYH poly-posis. MYH plays a key role in base excision repair ofdamages caused by reactive oxygen species [116]. MYHdeficiency leads to somatic C:G –>T:A transversions result-ing in deficient APC and K-ras genes [117]. The clinicalfeature shows a polyp burden of less than 20 to more than100 polyps predominantly located in the proximal colon.The age at diagnosis varies widely with a mean of about

45–55 years. The colorectal cancer risk is estimated of about80% by the age of 70 [118]. Clinically polyposis syndromeswith a low polyp burden and LS are difficult to differentiate.Increased awareness of an underlying hereditary condition iswarranted and genetic counseling is recommended forpatients with multiple polyps. For identification, it is impor-tant to acknowledge that two to three or five synchronouspolyps should be added to the number of polyps at subse-quent colonoscopies.

Due to the recessive nature of MYH polyposis, about1–2% of the general population harbors a deleterious muta-tion of the MYH gene [119] with obviously only a slightlyincreased risk of developing colorectal cancer [120]. Therisk to develop MAP for children of a patient affected withMAP is, assumed a healthy patient’s partner, negligible, butall children are obligate carriers.

Conclusions

The molecular clinical, pathological, and genetic back-ground of LS is well known. Due to this fact, predictivegenetic testing is possible and prophylactic surgery is arecommendable option at the time of colon cancer. Toidentify patients with LS, a high clinical awareness is war-ranted. Therefore, the knowledge of a detailed family histo-ry and the correlation with the Amsterdam I and II criteria aswell as with the Bethesda guidelines is essential.

Conflicts of interest GM has received a fee as speaker at a Bayerworkshop in 2010.

References

1. Boyle P, Ferlay J (2005) Cancer incidence and mortality inEurope, 2004. Ann Oncol 16:481–488

2. Lichtenstein P, Holm NV, Verkasalo PK et al (2000) Environmentaland heritable factors in the causation of cancer—analyses of cohortsof twins from Sweden, Denmark, and Finland. N Engl J Med343:78–85

3. Jasperson KW, Tuohy TM, Neklason DW, Burt RW (2010)Hereditary and familial colon cancer. Gastroenterology 138:2044–2058

4. Johns LE, Houlston RS (2001) A systematic review and meta-analysis of familial colorectal cancer risk. Am J Gastroenterol96:2992–3003

5. Goodenberger M, Lindor NM (2011) Lynch syndrome andMYH-associated polyposis: review and testing strategy. J ClinGastroenterol 45:488–500

6. Vasen HF, van der Meulen-de Jong AE, de Vos Tot NederveenCappel WH, Oliveira J (2009) Familial colorectal cancer risk:ESMO clinical recommendations. Ann Oncol 20(Suppl 4):51–53

7. Aaltonen LA, Salovaara R, Kristo P et al (1998) Incidence ofhereditary nonpolyposis colorectal cancer and the feasibility ofmolecular screening for the disease. N Engl J Med 338:1481–1487

Langenbecks Arch Surg (2012) 397:513–525 521

8. Hampel H, Frankel WL, Martin E et al (2008) Feasibility ofscreening for Lynch syndrome among patients with colorectalcancer. J Clin Oncol 26:5783–5788

9. Cunningham JM, Kim CY, Christensen ER et al (2001) Thefrequency of hereditary defective mismatch repair in a prospec-tive series of unselected colorectal carcinomas. Am J Hum Genet69:780–790

10. Cantor D (2006) The frustrations of families: Henry Lynch,heredity, and cancer control, 1962-1975. Med Hist 50:279–302

11. Lynch HT, Smyrk T, Lynch JF (1998) Molecular genetics andclinical-pathology features of hereditary nonpolyposis colorectalcarcinoma (Lynch syndrome): historical journey from pedigreeanecdote to molecular genetic confirmation. Oncology 55:103–108

12. Lynch HT, Shaw MW, Magnuson CW, Larsen AL, Krush AJ(1966) Hereditary factors in cancer. Study of two large midwest-ern kindreds. Arch Intern Med 117:206–212

13. Lynch HT, Krush AJ (1971) Cancer family “G” revisited: 1895–1970. Cancer 27:1505–1511

14. Warthin AS (1913) Hereditary with reference to carcinoma asshown by the study of the cases examined in the pathologicallaboratory of the University of Michigan, 1895–1913. Arch In-tern Med 117:206–212, Ref Type: Journal (Full)

15. Lynch HT, Drouhard TJ, Schuelke GS, Biscone KA, LynchJF, Danes BS (1985) Hereditary nonpolyposis colorectal can-cer in a Navajo Indian family. Cancer Genet Cytogenet15:209–213

16. Boland CR, Troncale FJ (1984) Familial colonic cancer withoutantecedent polyposis. Ann Intern Med 100:700–701

17. Vasen HF, Watson P, Mecklin JP, Lynch HT (1999) New clinicalcriteria for hereditary nonpolyposis colorectal cancer (HNPCC,Lynch syndrome) proposed by the International Collaborativegroup on HNPCC. Gastroenterology 116:1453–1456

18. Umar A, Boland CR, Terdiman JP et al (2004) Revised BethesdaGuidelines for hereditary nonpolyposis colorectal cancer (Lynchsyndrome) and microsatellite instability. J Natl Cancer Inst 96:261–268

19. Lynch HT, Lynch PM, Lanspa SJ, Snyder CL, Lynch JF, BolandCR (2009) Review of the Lynch syndrome: history, moleculargenetics, screening, differential diagnosis, and medicolegal ram-ifications. Clin Genet 76:1–18

20. Moslein G (2008) Hereditary colorectal cancer. Chirurg 79:1038–1046

21. Lindor NM, Rabe K, Petersen GM et al (2005) Lower cancerincidence in Amsterdam-I criteria families without mismatch repairdeficiency: familial colorectal cancer type X. JAMA 293:1979–1985

22. Jass JR (2006) Hereditary non-polyposis colorectal cancer: therise and fall of a confusing term. World J Gastroenterol 12:4943–4950

23. Mecklin JP, Sipponen P, Jarvinen HJ (1986) Histopathology ofcolorectal carcinomas and adenomas in cancer family syndrome.Dis Colon Rectum 29:849–853

24. Bellizzi AM, Frankel WL (2009) Colorectal cancer due to defi-ciency in DNA mismatch repair function: a review. Adv AnatPathol 16:405–417

25. Stoffel E, Mukherjee B, Raymond VM et al (2009) Calculation ofrisk of colorectal and endometrial cancer among patients withLynch syndrome. Gastroenterology 137:1621–1627

26. Hampel H, Frankel WL, Martin E et al (2005) Screening for theLynch syndrome (hereditary nonpolyposis colorectal cancer). NEngl J Med 352:1851–1860

27. Weissman SM, Bellcross C, Bittner CC et al (2011) Geneticcounseling considerations in the evaluation of families for Lynchsyndrome—a review. J Genet Couns 20:5–19

28. Hampel H, Frankel W, Panescu J et al (2006) Screening forLynch syndrome (hereditary nonpolyposis colorectal cancer)among endometrial cancer patients. Cancer Res 66:7810–7817

29. Vasen HF, Watson P, Mecklin JP et al (1994) The epidemiologyof endometrial cancer in hereditary nonpolyposis colorectal cancer.Anticancer Res 14:1675–1678

30. Hampel H, Stephens JA, Pukkala E et al (2005) Cancer risk inhereditary nonpolyposis colorectal cancer syndrome: later age ofonset. Gastroenterology 129:415–421

31. Sankila R, Aaltonen LA, Jarvinen HJ, Mecklin JP (1996) Bettersurvival rates in patients withMLH1-associated hereditary colorectalcancer. Gastroenterology 110:682–687

32. Albano WA, Recabaren JA, Lynch HT et al (1982) Naturalhistory of hereditary cancer of the breast and colon. Cancer50:360–363

33. Jass JR, Smyrk TC, Stewart SM, Lane MR, Lanspa SJ, Lynch HT(1994) Pathology of hereditary non-polyposis colorectal cancer.Anticancer Res 14:1631–1634

34. Vasen HF, Mecklin JP, Watson P et al (1993) Surveillance inhereditary nonpolyposis colorectal cancer: an international cooper-ative study of 165 families. The International Collaborative Groupon HNPCC. Dis Colon Rectum 36:1–4

35. Lynch HT, Harris RE, Lynch PM, Guirgis HA, Lynch JF, BardawilWA (1977) Role of heredity in multiple primary cancer. Cancer40:1849–1854

36. Mecklin JP, Jarvinen HJ (1986) Clinical features of colorectalcarcinoma in cancer family syndrome. Dis Colon Rectum 29:160–164

37. Lynch HT, de la Chapelle A (1999) Genetic susceptibility to non-polyposis colorectal cancer. J Med Genet 36:801–818

38. Goecke T, Schulmann K, Engel C et al (2006) Genotype-phenotype comparison of German MLH1 and MSH2 mutationcarriers clinically affected with Lynch syndrome: a report by theGerman HNPCC Consortium. J Clin Oncol 24:4285–4292

39. Ponti G, Losi L, Pedroni M et al (2006) Value of MLH1 andMSH2 mutations in the appearance of Muir-Torre syndromephenotype in HNPCC patients presenting sebaceous gland tumorsor keratoacanthomas. J Invest Dermatol 126:2302–2307

40. South CD, Hampel H, Comeras I, Westman JA, Frankel WL, dela Chapelle A (2008) The frequency of Muir-Torre syndromeamong Lynch syndrome families. J Natl Cancer Inst 100:277–281

41. Ollila S, Fitzpatrick R, Sarantaus L et al (2006) The importanceof functional testing in the genetic assessment of Muir-Torresyndrome, a clinical subphenotype of HNPCC. Int J Oncol28:149–153

42. Lynch HT, Fusaro RM, Roberts L, Voorhees GJ, Lynch JF (1985)Muir-Torre syndrome in several members of a family with avariant of the cancer family syndrome. Br J Dermatol 113:295–301

43. Rodriguez-Bigas MA, Boland CR, Hamilton SR et al (1997) ANational Cancer Institute Workshop on Hereditary NonpolyposisColorectal Cancer Syndrome: meeting highlights and Bethesdaguidelines. J Natl Cancer Inst 89:1758–1762

44. Kievit W, de Bruin JH, Adang EM et al (2004) Current clinicalselection strategies for identification of hereditary non-polyposiscolorectal cancer families are inadequate: a meta-analysis. ClinGenet 65:308–316

45. Debniak T, Kurzawski G, Gorski B, Kladny J, Domagala W,Lubinski J (2000) Value of pedigree/clinical data, immunohisto-chemistry and microsatellite instability analyses in reducing thecost of determining hMLH1 and hMSH2 gene mutations inpatients with colorectal cancer. Eur J Cancer 36:49–54

46. Pinol V, Castells A, Andreu M et al (2005) Accuracy ofrevised Bethesda guidelines, microsatellite instability, andimmunohistochemistry for the identification of patients with

522 Langenbecks Arch Surg (2012) 397:513–525

hereditary nonpolyposis colorectal cancer. JAMA 293:1986–1994

47. Salovaara R, Loukola A, Kristo P et al (2000) Population-basedmolecular detection of hereditary nonpolyposis colorectal cancer.J Clin Oncol 18:2193–2200

48. Julie C, Tresallet C, Brouquet A et al (2008) Identification indaily practice of patients with Lynch syndrome (hereditarynonpolyposis colorectal cancer): revised Bethesda guidelines-based approach versus molecular screening. Am J Gastroenterol103:2825–2835

49. Palomaki GE, McClain MR, Melillo S, Hampel HL, ThibodeauSN (2009) EGAPP supplementary evidence review: DNA testingstrategies aimed at reducing morbidity and mortality from Lynchsyndrome. Genet Med 11:42–65

50. Lindor NM, Petersen GM, Hadley DW et al (2006) Recommen-dations for the care of individuals with an inherited predispositionto Lynch syndrome: a systematic review. JAMA 296:1507–1517

51. Church J, McGannon E (2000) Family history of colorectalcancer: how often and how accurately is it recorded? Dis ColonRectum 43:1540–1544

52. Katballe N, Juul S, Christensen M, Orntoft TF, Wikman FP,Laurberg S (2001) Patient accuracy of reporting on hereditarynon-polyposis colorectal cancer-related malignancy in familymembers. Br J Surg 88:1228–1233

53. Sijmons RH, Boonstra AE, Reefhuis J et al (2000) Accuracy offamily history of cancer: clinical genetic implications. Eur J HumGenet 8:181–186

54. van Lier MG, Wagner A, van Leerdam ME et al (2010) A reviewon the molecular diagnostics of Lynch syndrome: a central rolefor the pathology laboratory. J Cell Mol Med 14:181–197

55. Jenkins MA, Hayashi S, O’Shea AM et al (2007) Pathologyfeatures in Bethesda guidelines predict colorectal cancer micro-satellite instability: a population-based study. Gastroenterology133:48–56

56. Young J, Simms LA, Biden KG et al (2001) Features of colorectalcancers with high-level microsatellite instability occurring infamilial and sporadic settings: parallel pathways of tumorigenesis.Am J Pathol 159:2107–2116

57. Greenson JK, Bonner JD, Ben-Yzhak O et al (2003) Phenotype ofmicrosatellite unstable colorectal carcinomas: well-differentiated andfocally mucinous tumors and the absence of dirty necrosis correlatewith microsatellite instability. Am J Surg Pathol 27:563–570

58. Alexander J, Watanabe T, Wu TT, Rashid A, Li S, Hamilton SR(2001) Histopathological identification of colon cancer withmicrosatellite instability. Am J Pathol 158:527–535

59. Kim H, Jen J, Vogelstein B, Hamilton SR (1994) Clinical andpathological characteristics of sporadic colorectal carcinomaswith DNA replication errors in microsatellite sequences. Am JPathol 145:148–156

60. Greenson JK, Huang SC, Herron C et al (2009) Pathologicpredictors of microsatellite instability in colorectal cancer. Am JSurg Pathol 33:126–133

61. Jass JR, Do KA, Simms LA et al (1998) Morphology of sporadiccolorectal cancer with DNA replication errors. Gut 42:673–679

62. Smyrk TC, Watson P, Kaul K, Lynch HT (2001) Tumor-infiltrating lymphocytes are a marker for microsatellite instabilityin colorectal carcinoma. Cancer 91:2417–2422

63. Yearsley M, Hampel H, Lehman A, Nakagawa H, de la Chapelle A,Frankel WL (2006) Histologic features distinguish microsatellite-high from microsatellite-low and microsatellite-stable colorectalcarcinomas, but do not differentiate germline mutations from meth-ylation of the MLH1 promoter. Hum Pathol 37:831–838

64. Michael-Robinson JM, Biemer-Huttmann A, Purdie DM et al(2001) Tumour infiltrating lymphocytes and apoptosis are inde-pendent features in colorectal cancer stratified according tomicrosatellite instability status. Gut 48:360–366

65. Lindor NM, Burgart LJ, Leontovich O et al (2002) Immunohis-tochemistry versus microsatellite instability testing in phenotyp-ing colorectal tumors. J Clin Oncol 20:1043–1048

66. Muller A, Giuffre G, Edmonston TB et al (2004) Challenges andpitfalls in HNPCC screening by microsatellite analysis andimmunohistochemistry. J Mol Diagn 6:308–315

67. Ruszkiewicz A, Bennett G, Moore J et al (2002) Correlation ofmismatch repair genes immunohistochemistry and microsatelliteinstability status in HNPCC-associated tumours. Pathology34:541–547

68. Muller W, Burgart LJ, Krause-Paulus R et al (2001) The reliabilityof immunohistochemistry as a prescreening method for the diagno-sis of hereditary nonpolyposis colorectal cancer (HNPCC)—resultsof an international collaborative study. Fam Cancer 1:87–92

69. Lazar V, Grandjouan S, Bognel C et al (1994) Accumulation ofmultiple mutations in tumour suppressor genes during colorectaltumorigenesis in HNPCC patients. Hum Mol Genet 3:2257–2260

70. Halvarsson B, Lindblom A, Johansson L, Lagerstedt K, NilbertM (2005) Loss of mismatch repair protein immunostaining incolorectal adenomas from patients with hereditary nonpolyposiscolorectal cancer. Mod Pathol 18:1095–1101

71. Boland CR (2000) Molecular genetics of hereditary nonpolyposiscolorectal cancer. Ann N YAcad Sci 910:50–59

72. Boland CR, Thibodeau SN, Hamilton SR et al (1998) A NationalCancer Institute Workshop on Microsatellite Instability for cancerdetection and familial predisposition: development of internation-al criteria for the determination of microsatellite instability incolorectal cancer. Cancer Res 58:5248–5257

73. Guastadisegni C, Colafranceschi M, Ottini L, Dogliotti E (2010)Microsatellite instability as a marker of prognosis and response totherapy: a meta-analysis of colorectal cancer survival data. Eur JCancer 46:2788–2798

74. Ogino S, Shima K, Meyerhardt JA et al (2012) Predictive andprognostic roles of BRAF mutation in stage III colon cancer:results from intergroup trial CALGB 89803. Clin Cancer Res18:890–900

75. Stein A, Hiemer S, Schmoll HJ (2011) Adjuvant therapy for earlycolon cancer: current status. Drugs 71:2257–2275

76. Bedeir A, Krasinskas AM (2011) Molecular diagnostics of colo-rectal cancer. Arch Pathol Lab Med 135:578–587

77. Kang GH (2011) Four molecular subtypes of colorectal cancerand their precursor lesions. Arch Pathol Lab Med 135:698–703

78. Kane MF, Loda M, Gaida GM et al (1997) Methylation of thehMLH1 promoter correlates with lack of expression of hMLH1 insporadic colon tumors and mismatch repair-defective humantumor cell lines. Cancer Res 57:808–811

79. Herman JG, Umar A, Polyak K et al (1998) Incidence andfunctional consequences of hMLH1 promoter hypermethylationin colorectal carcinoma. Proc Natl Acad Sci USA 95:6870–6875

80. Snover DC, Jass JR, Fenoglio-Preiser C, Batts KP (2005) Serrat-ed polyps of the large intestine: a morphologic and molecularreview of an evolving concept. Am J Clin Pathol 124:380–391

81. Carr NJ,Mahajan H, Tan KL, Hawkins NJ,Ward RL (2009) Serratedand non-serrated polyps of the colorectum: their prevalence in anunselected case series and correlation of BRAF mutation analysiswith the diagnosis of sessile serrated adenoma. J Clin Pathol 62:516–518

82. Jover R, Nguyen TP, Perez-Carbonell L et al (2011) 5-Fluorouracil adjuvant chemotherapy does not increase survivalin patients with CpG island methylator phenotype colorectalcancer. Gastroenterology 140:1174–1181

83. Ahuja N, Mohan AL, Li Q et al (1997) Association between CpGisland methylation andmicrosatellite instability in colorectal cancer.Cancer Res 57:3370–3374

84. Ogino S, Cantor M, Kawasaki T et al (2006) CpG island methylatorphenotype (CIMP) of colorectal cancer is best characterised by

Langenbecks Arch Surg (2012) 397:513–525 523

quantitative DNA methylation analysis and prospective cohortstudies. Gut 55:1000–1006

85. Sullivan RJ, Flaherty KT (2011) BRAF inmelanoma: pathogenesis,diagnosis, inhibition, and resistance. J Skin Cancer 2011:423239

86. Kim TH, Park YJ, Lim JA et al (2011) The association of theBRAF(V600E) mutation with prognostic factors and poor clinicaloutcome in papillary thyroid cancer: A meta-analysis. Cancer.doi:10.1002/cncr.26500

87. Tiacci E, Schiavoni G, Forconi F et al (2012) Simple geneticdiagnosis of hairy cell leukemia by sensitive detection of theBRAF-V600E mutation. Blood 119:192–195

88. Rajagopalan H, Bardelli A, Lengauer C, Kinzler KW, VogelsteinB, Velculescu VE (2002) Tumorigenesis: RAF/RAS oncogenesand mismatch-repair status. Nature 418:934

89. Samowitz WS, Sweeney C, Herrick J et al (2005) Poor survivalassociated with the BRAF V600E mutation in microsatellite-stable colon cancers. Cancer Res 65:6063–6069

90. Bae JM, Kim MJ, Kim JH et al (2011) Differential clinicopath-ological features in microsatellite instability-positive colorectalcancers depending on CIMP status. Virchows Arch 459:55–63

91. Van CE, Kohne CH, Lang I et al (2011) Cetuximab plus irinote-can, fluorouracil, and leucovorin as first-line treatment for meta-static colorectal cancer: updated analysis of overall survivalaccording to tumor KRAS and BRAF mutation status. J ClinOncol 29:2011–2019

92. Di NF, Martini M, Molinari F et al (2008) Wild-type BRAF isrequired for response to panitumumab or cetuximab in metastaticcolorectal cancer. J Clin Oncol 26:5705–5712

93. Lin JS, Webber EM, Senger CA, Holmes RS, Whitlock EP (2011)Systematic review of pharmacogenetic testing for predicting clin-ical benefit to anti-EGFR therapy in metastatic colorectal cancer.Am J Cancer Res 1:650–662

94. Domingo E, Laiho P, Ollikainen M et al (2004) BRAF screeningas a low-cost effective strategy for simplifying HNPCC genetictesting. J Med Genet 41:664–668

95. Schmiegel W, Pox C, Reinacher-Schick A et al (2010) S3 guide-lines for colorectal carcinoma: results of an evidence-based con-sensus conference on February 6/7, 2004 and June 8/9, 2007 (forthe topics IV, VI and VII). Z Gastroenterol 48:65–136

96. Vasen HF, Moslein G, Alonso A et al (2007) Guidelines for theclinical management of Lynch syndrome (hereditary non-polyposis cancer). J Med Genet 44:353–362

97. Teutsch SM, Bradley LA, Palomaki GE et al (2009) The Evalu-ation of Genomic Applications in Practice and Prevention(EGAPP) Initiative: methods of the EGAPP Working Group.Genet Med 11:3–14

98. Burt RW, Barthel JS, Dunn KB et al (2010) NCCN clinicalpractice guidelines in oncology. Colorectal cancer screening. JNatl Compr Canc Netw 8:8–61

99. Jarvinen HJ, Renkonen-Sinisalo L, Ktan-Collan K, Peltomaki P,Aaltonen LA, Mecklin JP (2009) Ten years after mutation testingfor Lynch syndrome: cancer incidence and outcome in mutation-positive and mutation-negative family members. J Clin Oncol27:4793–4797

100. Engel C, Rahner N, Schulmann K et al (2010) Efficacy of annualcolonoscopic surveillance in individuals with hereditary nonpo-lyposis colorectal cancer. Clin Gastroenterol Hepatol 8:174–182

101. de Vos Tot Nederveen Cappel WH, Nagengast FM, Griffioen G etal (2002) Surveillance for hereditary nonpolyposis colorectalcancer: a long-term study on 114 families. Dis Colon Rectum45:1588–1594

102. Parry S, Win AK, Parry B et al (2011) Metachronous colorectalcancer risk for mismatch repair gene mutation carriers: theadvantage of more extensive colon surgery. Gut 60:950–957

103. Warrier SK, Trainer AH, Lynch AC et al (2011) Preoperativediagnosis of Lynch syndrome with DNA mismatch repair

immunohistochemistry on a diagnostic biopsy. Dis Colon Rectum54:1480–1487

104. Pino MS, Mino-Kenudson M, Wildemore BM et al (2009) Defi-cient DNA mismatch repair is common in Lynch syndrome-associated colorectal adenomas. J Mol Diagn 11:238–247

105. Rothwell PM, Wilson M, Elwin CE et al (2010) Long-term effectof aspirin on colorectal cancer incidence and mortality: 20-yearfollow-up of five randomised trials. Lancet 376:1741–1750

106. Cook NR, Lee IM, Gaziano JM et al (2005) Low-dose aspirin inthe primary prevention of cancer: the Women’s Health Study: arandomized controlled trial. JAMA 294:47–55

107. Burn J, Gerdes AM, Macrae F et al (2011) Long-term effect ofaspirin on cancer risk in carriers of hereditary colorectal cancer:an analysis from the CAPP2 randomised controlled trial. Lancet378:2081–2087

108. Dolcetti R, Viel A, Doglioni C et al (1999) High prevalence ofactivated intraepithelial cytotoxic T lymphocytes and increasedneoplastic cell apoptosis in colorectal carcinomas with micro-satellite instability. Am J Pathol 154:1805–1813

109. Shia J, Ellis NA, Paty PB et al (2003) Value of histopathology inpredicting microsatellite instability in hereditary nonpolyposiscolorectal cancer and sporadic colorectal cancer. Am J SurgPathol 27:1407–1417

110. Watson P, Lin KM, Rodriguez-Bigas MA et al (1998) Colorectalcarcinoma survival among hereditary nonpolyposis colorectalcarcinoma family members. Cancer 83:259–266

111. Buckowitz A, Knaebel HP, Benner A et al (2005) Microsatelliteinstability in colorectal cancer is associated with local lymphocyteinfiltration and low frequency of distant metastases. Br J Cancer92:1746–1753

112. Linnebacher M, Gebert J, Rudy W et al (2001) Frameshiftpeptide-derived T-cell epitopes: a source of novel tumor-specific antigens. Int J Cancer 93:6–11

113. Saeterdal I, Bjorheim J, Lislerud K et al (2001) Frameshift-mutation-derived peptides as tumor-specific antigens in inheritedand spontaneous colorectal cancer. Proc Natl Acad Sci USA98:13255–13260

114. Schwitalle Y, Kloor M, Eiermann S et al (2008) Immune responseagainst frameshift-induced neopeptides in HNPCC patients andhealthy HNPCC mutation carriers. Gastroenterology 134:988–997

115. Reuschenbach M, Kloor M, Morak M et al (2010) Serum anti-bodies against frameshift peptides in microsatellite unstablecolorectal cancer patients with Lynch syndrome. Fam Cancer9:173–179

116. Slupska MM, Baikalov C, Luther WM, Chiang JH, Wei YF,Miller JH (1996) Cloning and sequencing a human homolog(hMYH) of the Escherichia coli mutY gene whose function isrequired for the repair of oxidative DNA damage. J Bacteriol178:3885–3892

117. Jones S, Emmerson P, Maynard J et al (2002) Biallelicgermline mutations in MYH predispose to multiple colorectaladenoma and somatic G:C–>T:A mutations. Hum Mol Genet11:2961–2967

118. Jenkins MA, Croitoru ME, Monga N et al (2006) Risk of colo-rectal cancer in monoallelic and biallelic carriers of MYH muta-tions: a population-based case-family study. Cancer EpidemiolBiomarkers Prev 15:312–314

119. Sieber OM, Lipton L, Crabtree M et al (2003) Multiple colorectaladenomas, classic adenomatous polyposis, and germ-line muta-tions in MYH. N Engl J Med 348:791–799

120. Croitoru ME, Cleary SP, Di NN et al (2004) Association betweenbiallelic and monoallelic germline MYH gene mutations andcolorectal cancer risk. J Natl Cancer Inst 96:1631–1634

121. Vasen HF, Mecklin JP, Khan PM, Lynch HT (1991) The Interna-tional Collaborative Group on Hereditary Non-Polyposis Colo-rectal Cancer (ICG-HNPCC). Dis Colon Rectum 34:424–425

524 Langenbecks Arch Surg (2012) 397:513–525

122. AarnioM, Sankila R, Pukkala E et al (1999) Cancer risk inmutationcarriers of DNA-mismatch-repair genes. Int J Cancer 81:214–218

123. Barrow E, Alduaij W, Robinson L et al (2008) Colorectal cancerin HNPCC: cumulative lifetime incidence, survival and tumourdistribution. A report of 121 families with proven mutations. ClinGenet 74:233–242

124. Barrow E, Robinson L, Alduaij W et al (2009) Cumulative lifetimeincidence of extracolonic cancers in Lynch syndrome: a report of121 families with proven mutations. Clin Genet 75:141–149

125. Dunlop MG, Farrington SM, Carothers AD et al (1997) Cancerrisk associated with germline DNA mismatch repair gene muta-tions. Hum Mol Genet 6:105–110

126. Kastrinos F, Mukherjee B, Tayob N et al (2009) Risk of pancre-atic cancer in families with Lynch syndrome. JAMA 302:1790–1795

127. Watson P, Vasen HF, Mecklin JP et al (2008) The risk of extra-colonic, extra-endometrial cancer in the Lynch syndrome. Int JCancer 123:444–449

Langenbecks Arch Surg (2012) 397:513–525 525