ArticlePDF Available

Microsatellite Marker Analysis in Screening for Hereditary Nonpolyposis Colorectal Cancer (HNPCC)

Authors:

Abstract

Hereditary nonpolyposis colorectal cancer (HNPCC) is an autosomal dominant cancer predisposition syndrome caused by germ-line mutations in DNA mismatch repair genes. It is relevant to identify HNPCC patients because colonoscopic screening of individuals with HNPCC mutations reduces cancer morbidity and mortality. Microsatellite instability (MSI) is characteristic of HNPCC tumors. A panel of five markers (BAT25, BAT26, D2S123, D5S346, and D17S250, the so-called Bethesda markers) has been proposed for screening for MSI. To test a hypothesis that the use of BAT26 alone is feasible in screening for MLH1/MSH2 mutation-positive HNPCC patients, we compared the MSI results of 494 colorectal cancer patients obtained using BAT26 with results obtained using the Bethesda markers. BAT26 was able to identify all 27 mutation-positive individuals in this series. The marker failed to identify 2 high MSI tumors and 20 low MSI tumors, all of which expressed MLH1, MSH2, and MSH6 when scrutinized by immunohistochemistry.
2001;61:4545-4549. Cancer Res
Anu Loukola, Katja Eklin, Päivi Laiho, et al.
Nonpolyposis Colorectal Cancer (HNPCC)
Microsatellite Marker Analysis in Screening for Hereditary
Updated version
http://cancerres.aacrjournals.org/content/61/11/4545
Access the most recent version of this article at:
Cited Articles
http://cancerres.aacrjournals.org/content/61/11/4545.full.html#ref-list-1
This article cites by 35 articles, 16 of which you can access for free at:
Citing articles
http://cancerres.aacrjournals.org/content/61/11/4545.full.html#related-urls
This article has been cited by 27 HighWire-hosted articles. Access the articles at:
E-mail alerts
related to this article or journal.Sign up to receive free email-alerts
Subscriptions
Reprints and
.pubs@aacr.orgDepartment at
To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Permissions
.permissions@aacr.orgDepartment at
To request permission to re-use all or part of this article, contact the AACR Publications
on June 4, 2013. © 2001 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from
[CANCER RESEARCH 61, 4545–4549, June 1, 2001]
Microsatellite Marker Analysis in Screening for Hereditary Nonpolyposis
Colorectal Cancer (HNPCC)
1
Anu Loukola,
2
Katja Eklin,
2
Pa¨ivi Laiho,
2
Reijo Salovaara, Paula Kristo, Heikki Ja¨rvinen, Jukka-Pekka Mecklin,
Virpi Launonen, and Lauri A. Aaltonen
3
Departments of Medical Genetics [A. L., K. E., P. L., R. S., P. K., V. L., L. A. A.] and Pathology [R. S.], Biomedicum Helsinki, FIN-00014 University of Helsinki, Finland; Second
Department of Surgery, Helsinki University Central Hospital, FIN-00029 Helsinki, Finland [H. J.]; Department of Surgery, Jyva¨skyla¨ Central Hospital, FIN-40620 Jyva¨skyla¨,
Finland [J-P. M.]; and Department of Oncology, Helsinki University Central Hospital, FIN-00029 Helsinki, Finland [L. A. A.]
ABSTRACT
Hereditary nonpolyposis colorectal cancer (HNPCC) is an autosomal
dominant cancer predisposition syndrome caused by germ-line mutations
in DNA mismatch repair genes. It is relevant to identify HNPCC patients
because colonoscopic screening of individuals with HNPCC mutations
reduces cancer morbidity and mortality. Microsatellite instability (MSI) is
characteristic of HNPCC tumors. A panel of five markers (BAT25,
BAT26, D2S123, D5S346, and D17S250, the so-called Bethesda markers)
has been proposed for screening for MSI. To test a hypothesis that the use
of BAT26 alone is feasible in screening for MLH1/MSH2 mutation-positive
HNPCC patients, we compared the MSI results of 494 colorectal cancer
patients obtained using BAT26 with results obtained using the Bethesda
markers. BAT26 was able to identify all 27 mutation-positive individuals
in this series. The marker failed to identify 2 high MSI tumors and 20 low
MSI tumors, all of which expressed MLH1, MSH2, and MSH6 when
scrutinized by immunohistochemistry.
INTRODUCTION
HNPCC
4
is an autosomal dominant inherited cancer susceptibility
syndrome. The condition is characterized by the development of CRC
at an early age and by frequently occurring extracolonic tumors, e.g.,
cancers of the endometrium, stomach, ovaries, small bowel, ureter,
biliary tract, and renal pelvis (1, 2). Early removal of benign and
malignant tumors reduces cancer morbidity and mortality (3, 4).
HNPCC is caused by an inherited mutation in one of five mismatch
repair genes: (a) MLH1; (b) MSH2; (c) PMS1; (d) PMS2; and (e)
MSH6 (5, 6). Defective DNA mismatch repair results in RERs and
genetic instability, which can easily be observed in short repetitive
sequences such as microsatellites (7–9) and is thus referred to as MSI.
In the majority of families with HNPCC, the mutations affect MLH1
or MSH2 (10), and few mutations have been reported in PMS1, PMS2,
and MSH6.
5
A total of 85–90% of HNPCC patients show MSI, and this propor-
tion is even higher in mutation-positive families (7, 11, 12), whereas
only 10–15% of sporadic colorectal tumors do so (7–9). Thus, MSI is
a relatively sensitive but unspecific marker for HNPCC. In 1997, the
International Workshop on MSI and RER Phenotypes in Cancer
Detection and Familial Predisposition proposed a panel of five mic-
rosatellite markers to be used in MSI analysis (13). For the purpose of
providing some uniformity, two mononucleotide repeats (BAT25 and
BAT26) and three dinucleotide repeats (D2S123, D5S346, and
D17S250) were recommended. Using this reference panel (known as
the Bethesda markers), tumors with instability in two or more markers
are defined as MSI-H, tumors with instability in one marker are
defined as MSI-L, and tumors in which none of the markers exhibit
MSI are defined as MSS. Arguments in favor of combining the MSI-L
and MSS groups include the facts that the baseline mutation rate for
microsatellites in apparently stable CRCs is not precisely known and
that the clinical features in the two groups are similar (14, 15). The
distinction between these two groups is dependent on both the type
and the number of microsatellites analyzed. If a large number of
markers are used, all CRCs may exhibit some level of MSI (13).
Nevertheless, MSI-L CRCs seem to be distinct from both MSI-H
and MSS CRC. They appear to have more in common with MSS
cancers (16), although they can be distinguished on the basis of a
higher frequency of K-ras mutations (16, 17) and reduced expression
of BCL-2 (16, 18). MSI-L may represent a subtype of CRC combining
features of the common chromosomal instability and mild mutator
pathways (16, 19). However, MLH1 and MSH2 do not appear to be
implicated in the MSI-L subset (20–22); thus, for the purpose of
identifying patients with defects in these two genes, MSI-L and MSS
cases may not need to be distinguished.
BAT26 has some advantages over many other markers in MSI
analysis. It is extremely sensitive in detecting tumors with instability
(20, 23–26) and shows negligible size variation either between both
alleles of one individual or among individuals (23). Several studies
support the use of BAT26 on tumor DNA alone (23, 24, 27). How-
ever, a germ-line polymorphism in the BAT26 locus has been detected
in 7.7–12.6% of African Americans (26, 28) and in 0.8% of Cauca-
sians (26). The presence of allelic variations, although rare in some
populations, emphasizes the need for matching normal DNA in MSI-
positive cases to avoid misclassifications.
This work was performed to test MSI analysis using the Bethesda
panel of five markers in a series of 494 CRCs including 27 patients
with identified MLH1 or MSH2 mutations. This series is part of a
larger population-based study (24, 29). The MSI status of these
samples had already been determined using BAT26 alone. The aim of
this study was to evaluate the possible benefit of using the Bethesda
set, as compared with using BAT26 alone, to identify MLH1 or MSH2
mutation-positive HNPCC patients.
MATERIALS AND METHODS
Patients. The study was approved by the appropriate ethics review com-
mittees. Colorectal tumor and normal tissue specimens were derived from 494
CRC patients treated at nine large regional hospitals in southeastern Finland.
Altogether, 484 patients were derived from a consecutive series, and 10
patients with a germ-line MLH1 or MSH2 mutation were added to enrich the
proportion of mutation-positive patients (24, 29). The individuals ranged in age
from 34–92 years, with a mean age of 67 years. The freshly frozen samples
Received 11/20/00; accepted 3/26/01.
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance with
18 U.S.C. Section 1734 solely to indicate this fact.
1
This work was supported by grants from the Finnish Cancer Society, the Academy of
Finland, Sigrid Juselius Foundation, Duodecim, Ida Montin Foundation, Jalmari and
Rauha Ahokas Foundation, Emil Aaltonen Foundation, Finnish-Norwegian Medical
Foundation, Nordisk Cancer Union, Paulo Foundation, and Helsinki University Central
Hospital and carried out at the Center of Excellence in Disease Genetics of the Academy
of Finland (Project 44870).
2
A. L., K. E., and P. L. contributed equally to this work.
3
To whom requests for reprints should be addressed, at Department of Medical
Genetics, Haartman Institute, P. O. Box 63, FIN-00014 University of Helsinki, Finland.
Phone: 358-9-19125595; Fax: 358-9-19125105; E-mail: lauri.aaltonen@helsinki.fi.
4
The abbreviations used are: HNPCC, hereditary nonpolyposis colorectal cancer;
CRC, colorectal cancer; MSI, microsatellite instability; MSI-H, high MSI; MSI-L, low
MSI; MSS, microsatellite stable; RER, replication error; TGF-
RII, transforming growth
factor
receptor II.
5
http.//www.nfdht.nl/.
4545
on June 4, 2013. © 2001 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from
were evaluated histologically by a pathologist before DNA extraction to
document the proportion of tumor tissue. A total of 480 of 494 (97%) samples
displayed 50% or more carcinoma tissue. The specimens representing normal
mucosa were always derived from a separate site rather than from the tumor
margins.
MSI Analysis. All 494 samples had already been analyzed for MSI using
BAT26 and tumor DNA (24, 29). In this study, these tumor DNAs and
respective normal tissue DNAs were used to independently study MSI status
using five fluorescence-labeled microsatellite markers (BAT25, BAT26,
D2S123, D5S346, and D17S250, the Bethesda panel). Primer sequences are
presented in Table 1. PCR reactions were carried out in a 10-
l reaction
volume containing 50–100 ng of genomic DNA, 1 PCR buffer (Perkin-
Elmer Applied Biosystems Division, Foster City, CA), 250
M each de-
oxynucleotide triphosphate (Finnzymes, Espoo, Finland), 0.5
M each primer,
and 1 unit of AmpliTaq Gold polymerase (Perkin-Elmer). The MgCl
2
concen-
tration was 2.5 mM for BAT25 and 2.75 mM for BAT26, D2S123, D5S346, and
D17S250. The PCR cycles for each marker are indicated in Table 1. Prede-
naturation was performed at 95°C for 10 min, and final extension was per-
formed at 72°C for 10 min in all reactions. PCR products were loaded on a 5%
Long Ranger 6 M urea gel (FMC BioProducts, Rockland, ME) and run in an
ABI PRISM 377 DNA Sequencer (Perkin-Elmer) according to the manufac-
turer’s instructions. The data were collected automatically and analyzed by
GeneScan 3.1 software (Perkin-Elmer).
Patients whose tumor DNA showed alleles that were not present in the
corresponding normal DNA were classified as MSI positive. With BAT25 and
BAT26, shifts of 3 or more bp were considered MSI positive. For the dinu-
cleotide markers (D2S123, D5S346, and D17S250), all deviations starting
from shifts of one CA repeat were acknowledged. If only one of the five
markers showed MSI, the tumor was classified as MSI-L, and if two or more
markers showed MSI, the tumor was classified as MSI-H. The results were
evaluated visually by three independent reviewers (A. L., V. L., L. A. A.). All
samples showing MSI-L were analyzed using five additional markers (TGF-
RII, D18S363, D18S1156, D5S318, and TP53). Primer sequences and PCR
conditions are available on request.
In 16 cases, the distinction between MSI-positive and MSS in individual
markers was difficult to perform visually, and in these cases, a previously
described (30) mathematical model for the calculation of a RER score was
used. The RER or MSI score was calculated individually for each dinucleotide
marker and used to determine the MSI status of each marker separately. A MSI
score of 25% or higher was used as a cutoff level for positivity (30).
Immunohistochemistry. Immunohistochemistry for MLH1, MSH2, and
MSH6 was performed for MSI-L cases and novel MSI-H cases. Paraffin-
embedded surgical resection specimens were collected from the files of pa-
thology departments in different hospitals.
The tissue sections were mounted on ChemMate Capillary Gap microscopy
slides (DAKO A/S, Glostrup, Denmark; BioTek Solutions) and dried at 37°C.
The sections were deparaffinized in xylene and rehydrated through a graded
alcohol series to distilled water. The samples were then microwaved at high
power four times (5 min each) in citrate buffer and then cooled and washed
in PBS.
The following monoclonal antibodies were used: (a) MLH1 (clone G168-
15; catalogue number 13271; PharMingen); (b) MSH2 (clone FE 11; catalogue
number NA27; Oncogene Sciences); and (c) MSH6 (clone 44; G70220;
Transduction Laboratories). For immunohistochemical analysis, avidin-biotin
complex immunoperoxidase technique was performed by using a commercial
ChemMate detection kit (DAKO A/S) in a Techmate automate machine.
Endogenous peroxidase was blocked by incubation in hydrogen peroxide with
methanol. Incubation with nonimmune horse serum was followed by incuba-
tion with primary antibody. The sections were then incubated in biotinylated
second antibody and peroxidase-labeled avidin-biotin complex. All dilutions
were made in PBS (pH 7.2). The stainings were visualized with diaminoben-
zidine tetrahydrochloride solution. The sections were counterstained in May-
er’s hematoxylin, rinsed with water, and mounted in an aqueous mounting
media (Aquamount, BDH, Poole, United Kingdom). The percentage of posi-
tive nuclei was evaluated and scored as follows: (a) , 0%; (b) , 1–10%; (c)
⫹⫹, 11–50%; (d) ⫹⫹⫹, 51–80%; and (e) ⫹⫹⫹⫹, 81% or more. The slides
were analyzed by one pathologist (R. S.).
Detection of Germ-line Mutations. All 494 patients had been scrutinized
previously for the two most common mismatch repair gene mutations in
Finland (24, 29). Founder mutation 1 is a 3.5-kb genomic deletion of MLH1
comprising exon 16, and founder mutation 2 is MLH1 exon 6 splice site
mutation G3 A at 454-1 (last intronic base before exon 6). If neither founder
mutation was detected, but the patient’s tumor displayed MSI (analyzed using
BAT26 alone), mutation analysis of MLH1 and MSH2 was performed by direct
genomic sequencing of the coding exons including the flanking intronic
regions and promoter region, as described previously (24, 29).
The new MSI-H cases appearing after MSI analysis using the Bethesda
markers were similarly analyzed for mutations in MLH1 and MSH2.To
exclude the possibility of large deletions of MLH1 and MSH2, the new MSI-H
cases were analyzed by Southern blotting according to standard procedures.
Genomic DNA was digested with EcoRI and analyzed with two different
cDNA probes encompassing MLH1 exons 11–19 and MSH2 exons 1–8. The
new MSI-H cases were also screened for MSH6 mutations by direct sequenc-
ing covering 98% of the coding region. Primer sequences and PCR conditions
are available on request. Direct sequencing of the PCR products was performed
using cycle sequencing with Big Dye Terminator kit (Perkin-Elmer), and
reactions were run on an ABI 3100 capillary sequencer (Perkin-Elmer) ac-
cording to the manufacturer’s instructions.
A total of 182 cancer-free control individuals and 83 CRC patients were
analyzed for a MSH6 variant in exon 2 by single-strand conformational
polymorphism analysis using mutation detection enhancement gel solution
(BioWhittaker Molecular Applications, Rockland, ME). PCR products were
run on 0.6mutation detection enhancement gels at 4 W for 22 h. The running
buffer was 0.6 Tris-borate EDTA. Single-strand conformational polymor-
phism gels were silver-stained according to standard procedures.
RESULTS AND DISCUSSION
HNPCC is the most common hereditary CRC syndrome. The pa-
tients benefit greatly from early diagnosis because CRC deaths can be
efficiently reduced by removing adenomas and early carcinomas (3,
4), emphasizing the need for development of molecular identification
procedures for HNPCC. MSI analysis is a practical tool for prescreen-
ing HNPCC (24, 29, 31–33). A panel containing two mononucleotide
markers (BAT25 and BAT26) and three dinucleotide markers
(D2S123, D5S346, and D17S250), the so-called Bethesda markers,
has been proposed for MSI analysis (13). BAT26 has some advan-
tages over dinucleotide markers. It is quasimonomorphic (23), and
germ-line polymorphisms are rare in the Caucasian population (26). If
analysis of tumor tissue DNA reveals no putative shifts, use of normal
Table 1 Primer sequences (http://www.gdb.org) and PCR cycles for the five Bethesda markers
Marker Primer sequences PCR cycles
BAT25 TCG-CCT-CCA-AGA-ATG-TAA-GT 28 cycles of 95°C for 1 min, 56°C for 45 s, 72°C for 45 s
TCT-GGA-TTT-TAA-CTA-TGG-CTC
BAT26 TGA-CTA-CTT-TTG-ACT-TCA-GCC 32 cycles of 95°C for 45 s, 55°C for 1 min, 72°C for 30 s
AAC-CAT-TCA-ACA-TTT-TTA-ACC
D2S123 AAA-CAG-GAT-GCC-TGC-CTT-TA 35 cycles of 95°C for 45 s, 55°C for 45 s, 72°C for 45 s
GGA-CTT-TCC-ACC-TAT-GGG-AC
D5S346 ACT-CAC-TCT-AGT-GAT-AAA-TCG-GG 30 cycles of 95°C for 1 min, 57°C for 45 s, 72°C for 45 s
AGC-AGA-TAA-GAC-AAG-TAT-TAC-TAG
D17S250 GGA-AGA-ATC-AAA-TAG-ACA-AT 35 cycles of 95°C for 45 s, 55°C for 45 s, 72°C for 45 s
GCT-GGC-CAT-ATA-TAT-ATT-TAA-ACC
4546
MICROSATELLITE MARKERS IN SCREENING FOR HNPCC
on June 4, 2013. © 2001 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from
control DNA is not necessary. The use of Bethesda markers, on the
other hand, harbors some technical difficulties. Reamplifications are
frequently needed to obtain successful amplifications and unambigu-
ous results for all markers and for both normal and tumor DNA; in this
study, there were 31 reamplifications for BAT25, 68 reamplifications
for BAT26, 119 reamplifications for D2S123, 57 reamplifications for
D5S346, and 182 reamplifications for D17S250. In addition, the
interpretation of D2S123, D5S346, and D17S250 is not trivial and
cannot be accomplished without the matching normal DNA, even if
the sample is MSS (see Fig. 1). The cutoff level for MSI positivity for
the mononucleotide markers was set at shifts of 3 bp or greater. The
cutoff level of 3 bp was chosen because all 10 cases displaying such
deviation also had instability at other loci. Eight and seven patients
showed 1–2-bp deviations at BAT25 and BAT26, respectively, and
none of these cases displayed additional evidence of MSI at other loci.
In 31 individual cases, the three reviewers had discrepancies when
scoring dinucleotide markers, whereas not a single scoring discrep-
ancy occurred with BAT25 and BAT26.
A total of 494 CRC patients were successfully analyzed for MSI
using the Bethesda panel of five microsatellite markers (13). A total
of 73 of 494 patients (14.8%) had previously shown MSI when
analyzed using BAT26 alone (24, 29). When the five Bethesda mark-
ers were used, all 73 appeared as MSI-H (Table 2). A total of 95 of
494 patients (19.2%) were classified as MSI with the Bethesda panel;
75 patients (15.2%) were classified as MSI-H, and 20 patients (4.0%)
were classified as MSI-L (Table 2).
Twenty-two new MSI-positive cases appeared when the panel of
five markers was used (2 MSI-H cases and 20 MSI-L cases; Table 2).
Germ-line mutation analysis of MLH1 and MSH2 was performed for
the two new MSI-H cases. No mutations were found in the coding
regions, exon-intron boundaries, or the promoter region of these
genes. Neither case had a family history of cancer, and the ages at
Fig. 1. MSI analysis using BAT25, BAT26, D2S123, D5S346, and D17S250. For each marker, the top graph represents tumor DNA, and the bottom graph represents matching
normal DNA. A, case c145 (Finnish founder mutation 1, 70% tumor tissue) shows instability at all markers except D5S346. B, case c1034 (no MLH1/MSH2 germ-line mutation
identified, 70% tumor tissue) shows instability only at BAT25 and BAT26. The novel allele in BAT25 represents the cutoff level for positivity (shift of 3 bp in tumor DNA). Marker
D5S346 shows a weak extra peak on this patient’s tumor (indicated by an arrow). The RER score (30) was calculated to confirm the MSI status and resulted in a score of 3.2%, consistent
with visual MSI scoring, excluding MSI positivity. However, considering the patterns in other loci, the peak indicated by the arrow may reflect true MSI. Scoring is much more trivial
with the mononucleotide markers because the cutoff level can be determined as the number of bp deleted. The BAT25 and BAT26 patterns in this study were highly reproducible. C,
examples of shifts in BAT26 that are below the cutoff level: sample c859, top graph pair; and sample c1079, bottom graph pair.
Table 2 Comparison of MSI analysis based on BAT26 alone and MSI analysis based on the Bethesda markers (BAT25, BAT26, D2S123, D5S346, and D17S250)
The total number of samples studied was 494.
MSI with BAT26 alone
MSI with the Bethesda markers
0 positive
markers
1 positive
marker
2 positive
markers
3 positive
markers
4 positive
markers
5 positive
markers
BAT26 positive (n 73) n 4 n 8 n 25 n 36
BAT26 negative (n 421) n 399 n 20 n 2
4547
MICROSATELLITE MARKERS IN SCREENING FOR HNPCC
on June 4, 2013. © 2001 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from
diagnosis were 64 and 65 years. To examine the molecular back-
ground of the observed MSI-H phenotype in more detail, additional
experiments were performed on the two cases. Because direct
genomic sequencing cannot detect defects such as genomic deletions
affecting whole exons, we examined both MLH1 and MSH2 by
Southern hybridization. No aberrations were detected. Direct genomic
sequencing of MSH6 revealed a missense change in one case; MSH6
exon 2 S144I (AGC3 ATC). The tumor DNA showed no loss of
heterozygosity at this change. This variant has been reported previ-
ously as a pathogenic mutation (34). We analyzed 182 cancer-free
controls and 83 individuals with CRC for the change and found it in
1 control sample. It thus appears to be a rare polymorphism, although
additional studies are needed to confirm its nature.
Five additional microsatellite markers (TGF-
RII, D18S363,
D18S1156, D5S318, and TP53) were used to confirm the status of the
20 MSI-L cases. Three samples showed further instability with one of
the markers. This degree of instability (2 of 10 or 20%) should be
considered as MSI-L (13).
In addition, immunohistochemistry of MLH1, MSH2, and MSH6
was performed for the 20 MSI-L and the 2 new MSI-H cases. Because
no lack of expression was revealed in the MSI-L cases (all scored
⫹⫹, ⫹⫹⫹,or⫹⫹⫹⫹), no further procedures were performed. Also,
previous studies have confirmed that the expression of MLH1 and
MSH2 is not altered in MSI-L cases, whereas loss of expression in one
of them, typically MLH1, can be seen in most MSI-H cases (20, 21).
However, because MLH1 antibody has a tendency to show a weak
positive staining even in cases harboring deleterious mutations (35),
we must acknowledge the possible involvement of a gene defect even
in cases showing MLH1 immunostaining. Importantly, the two new
MSI-H cases expressed MLH1, MSH2, and MSH6 (all scored ⫹⫹⫹
or ⫹⫹⫹⫹). This result, together with their BAT26 negativity, im-
plies that they are possibly MSI-L despite the two unstable dinucle-
otide markers.
All 27 mutation-positive CRCs in the series of 494 tumors showed
instability at both the BAT26 and BAT25 loci. However, in other
studies, we have detected one sample derived from a mutation-
positive patient with no instability at BAT26. The patient was diag-
nosed at 37 years of age with a proximal tumor, has Finnish founder
mutation 1, and has a strong family history of CRC. Thus, the lesion
is most likely a HNPCC tumor. The degree of MSI in tumors is related
to past patterns of tumorigenesis, primarily to mutation rate and the
number of divisions since loss of mismatch repair. Relatively young
lesions are likely to show less MSI, and rare cases of HNPCC may
display MSI at relatively few loci (36). In this study, D2S123,
D5S346, and D17S250 identified 24 (89%), 16 (59%), and 22 (81%)
of 27 mutation-positive individuals, respectively. Despite negative
results in genomic sequencing, some of the 46 cases that showed
BAT26 instability may have had a MSH2 or MLH1 mutation that was
not detected by the method. Of these 46 individuals, 35 had no family
history of CRC or endometrial cancer, but undetected mutations may
underlie a subset of the 11 cases with some HNPCC features (1–3
relatives with CRC or endometrial cancer). None of the 11 pedigrees
fulfilled the Amsterdam criteria for HNPCC (2).
It is possible that the two common founder mutations in Finland
cause a bias toward MSI-H phenotypes in our HNPCC series. To-
gether, these two mutations accounted for 18 of 27 (67%) mutations
in this series. However, all of the nine other patients with five
different mutations exhibited MSI-H phenotypes, supporting the no-
tion that deleterious MLH1 and MSH2 mutations tend to cause MSI-H
phenotypes. A minority of HNPCC tumors are MSS. The genes
responsible for HNPCC are not yet fully known, and some of these
appear to be associated with more attenuated phenotypes (13). It is
possible that MSH6, TGF-
RII, and perhaps other currently uniden-
tified predisposing genes cause a MSI-L or MSS phenotype (34).
Relying on BAT26 alone in MSI analysis has prompted the ques-
tion of possible loss of sensitivity and specificity. The proposed panel
of five microsatellite markers (13) aims at dividing tumors into
categories of MSI-H, MSI-L, and MSS, of which typically only
MSI-H tumors are considered as candidates for MLH1/MSH2 muta-
tion analysis (20–22). When relying on one marker, distinguishing
between MSI-H and MSI-L is impossible. According to this study,
BAT26 detects MSI-H cases with high sensitivity because 73 of 75
(97%) MSI-H cases were BAT26 positive. The sensitivity in detecting
MLH1/MSH2 mutation-positive individuals was 100% (27 of 27).
Only 2 of 421 (0.5%) BAT26-negative tumors were found to be
MSI-H, and no MLH1/MSH2 mutations could be found in these
patients. Twenty of 421 (4.8%) BAT26-negative tumors were found
to be MSI-L. These 22 BAT26-negative tumors expressed MLH1,
MSH2, and MSH6 according to immunohistochemistry and are un-
likely to represent HNPCC. The specificity of BAT26 in detecting
mutation-positive individuals (27 of 73 cases, 37%) was very similar
to the specificity of the Bethesda panel (27 of 75 cases, 36%). The
data suggest that there is little loss of specificity when relying on
BAT26 alone.
On the basis of analyzing 494 CRC patients for MSI using the
Bethesda markers, we conclude that BAT26 alone was sufficient to
identify 97% of MSI-H cases but seemingly fails to detect MSI-L
cases. Thus, additional markers are needed when aiming at distin-
guishing MSI-L and MSS subgroups. However, this may not be
necessary when prescreening patients for MLH1/MSH2 germ-line
mutations. It seems that BAT26 identifies MLH1 or MSH2 mutation-
positive CRC patients with high sensitivity. Whereas utilization of
more markers may be useful if extensive resources are available, the
benefit appears marginal and may not be cost effective. Utilization of
numerous markers resulted in increased workload and expense, as
well as difficulties in scoring even in experienced hands. Our data
indicate that BAT26 alone can be used as a tool in detection of
MLH1/MSH2-associated HNPCC in circumstances where resources
and experience in interpretation are limited.
ACKNOWLEDGMENTS
We thank Susanna Anjala, Annika Lahti, Kirsi Laukkanen, Siv Lindroos,
Sinikka Lindh, and Anitta Hottinen for technical assistance.
REFERENCES
1. Watson, P., and Lynch, H. T. Extracolonic cancer in hereditary nonpolyposis colo-
rectal cancer. Cancer (Phila.), 71: 677–685, 1993.
2. Vasen, H. F. A., Watson, P., Mecklin, J-P., Lynch, H. T., and the ICG-HNPCC. New
clinical criteria for hereditary nonpolyposis colorectal cancer (HNPCC, Lynch syn-
drome) proposed by the International Collaborative Group on HNPCC. Gastroenter-
ology, 116: 1453–1456, 1999.
3. Ja¨rvinen, H., Mecklin, J-P., and Sistonen, P. Screening reduces colorectal cancer rate
in families with hereditary nonpolyposis colorectal cancer. Gastroenterology, 108:
1405–1411, 1995.
4. Ja¨rvinen, H. J., Aarnio, M., Mustonen, H., Aktan-Collan, K., Aaltonen, L. A.,
Peltoma¨ki, P., de la Chapelle, A., and Mecklin, J-P. Controlled 15-year trial on
screening for colorectal cancer in families with hereditary nonpolyposis colorectal
cancer. Gastroenterology, 118: 829834, 2000.
5. Kinzler, K. W., and Vogelstein, B. Lessons from hereditary colorectal cancer. Cell,
87: 159–170, 1996.
6. Miyaki, M., Konishi, M., Tanaka, K., Kikuchi-Yanoshita, R., Muraoka, M., Yasuno,
M., Igari, T., Koike, M., Chiba, M., and Mori, T. Germline mutation of MSH6 as the
cause of hereditary nonpolyposis colorectal cancer. Nat. Genet., 17: 271–272, 1997.
7. Aaltonen, L. A., Peltoma¨ki, P., Leach, F., Sistonen, P., Pylkka¨nen, L., Mecklin, J-P.,
Ja¨rvinen, H., Powell, S., Jen, J., Hamilton, S. R., Petersen, G. M., Kinzler, K. W.,
Vogelstein, B., and de la Chapelle, A. Clues to the pathogenesis of familial colorectal
cancer. Science (Wash. DC), 260: 812–816, 1993.
8. Ionov, Y., Peinado, M. A., Malkhosyan, S., Shibata, D., and Perucho, M. Ubiquitous
somatic mutations in simple repeated sequences reveal a new mechanism for colonic
carcinogenesis. Nature (Lond.), 363: 558–561, 1993.
4548
MICROSATELLITE MARKERS IN SCREENING FOR HNPCC
on June 4, 2013. © 2001 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from
9. Thibodeau, S. N., Bren, G., and Schaid, D. Microsatellite instability in cancer of the
proximal colon. Science (Wash. DC), 260: 816819, 1993.
10. Peltoma¨ki, P., and Vasen, H. F. Mutations predisposing to hereditary nonpolyposis
colorectal cancer: database and results of a collaborative study. The International
Collaborative Group on Hereditary Nonpolyposis Colorectal Cancer. Gastroenterol-
ogy, 113: 1146–1158, 1997.
11. Aaltonen, L. A., Peltoma¨ki, P., Mecklin, J-P., Ja¨rvinen, H., Jass, J. R., Green, J. S.,
Lynch, H. T., Watson, P., Tallqvist, G., Juhola, M., Sistonen, P., Hamilton, S. R.,
Kinzler, K. W., Vogelstein, B., and de la Chapelle, A. Replication errors in benign
and malignant tumors from hereditary nonpolyposis colorectal cancer patients. Can-
cer Res., 54: 1645–1648, 1994.
12. Moslein, G., Tester, D. J., Lindor, N. M., Honchel, R., Cunningham, J. M., French,
A. J., Halling, K. C., Schwab, M., Goretzki, P., and Thibodeau, S. N. Microsatellite
instability and mutation analysis of hMSH2 and hMLH1 in patients with sporadic,
familial and hereditary colorectal cancer. Hum. Mol. Genet., 5: 1245–1252, 1996.
13. Boland, C. R., Thibodeau, S. N., Hamilton, S. R., Sudransky, D., Eshleman, J. R.,
Burt, R. W., Meltzer, S. J., Rodriguez-Bigas, M. A., Fodde, R., Ranzani, G. N., and
Srivastava, S. A National Cancer Institute Workshop on microsatellite instability for
cancer detection and familial predisposition: development of international criteria for
the determination of microsatellite instability in colorectal cancer. Cancer Res., 58:
5248–5257, 1998.
14. Lothe, R. A., Peltoma¨ki, P., Meling, G. I., Aaltonen, L. A., Nystro¨m-Lahri, M.,
Pylkka¨nen, L., Heimdal, K., Andersen, T. I., Møller, P., Rognum, T. O., Fossa, S. D.,
Haldorsen, T., Langmark, F., Brøgger, A., de la Chapelle, A., and Børresen, A-L.
Genomic instability in colorectal cancer: relationship to clinicopathological variables
and family history. Cancer Res., 53: 5849–5852, 1993.
15. Jass, J. R., Do, K. A., Simms, L. A., Iino, H., Wynter, C., Pillay, S. P., Searle, J.,
Radford-Smith, G., Young, J., and Leggett, B. Morphology of sporadic colorectal
cancer with DNA replication errors. Gut, 42: 673–679, 1998.
16. Jass, J. R., Biden, K. G., Cummings, M. C., Simms, L. A., Walsh, M., Schoch, E.,
Meltzer, S. J., Wright, C., Searle, J., Young, J., and Leggett, B. A. Characterization
of a subtype of colorectal cancer combining features of the suppressor and mild
mutator pathways. J. Clin. Pathol., 52: 455–460, 1999.
17. Konishi, M., Kikuchi-Yanoshita, R., Tanaka, K., Muraoka, M., Onda, A., Okumura,
Y., Kishi, N., Iwama, T., Mori, T., Koike, M., Ushio, K., Chiba, M., Nomizu, S.,
Konishi, F., Utsunomiya, J., and Miyaki, M. Molecular nature of colon tumors in
hereditary nonpolyposis colorectal cancer, familial polyposis, and sporadic colon
cancer. Gastroenterology, 111: 307–317, 1996.
18. Biden, K. G., Simms, L. A., Cummings, M., Buttenshaw, R., Schoch, E., Searle, J.,
Gobe, G., Jass, J. R., Meltzer, S. J., Leggett, B. A., and Young, J. Expression of Bcl-2
protein is decreased in colorectal adenocarcinomas with microsatellite instability.
Oncogene, 18: 1245–1249, 1999.
19. Iino, H., Jass, J. R., Simms, L. A., Young, J., Leggett, B., Ajioka, Y., and Watanabe,
H. DNA microsatellite instability in hyperplastic polyps, serrated adenomas, and
mixed polyposis: a mild mutator pathway for colorectal cancer? J. Clin. Pathol., 52:
5–9, 1999.
20. Dietmaier, W., Wallinger, S., Bocker, T., Kullmann, F., Fishel, R., and Ruschoff, J.
Diagnostic microsatellite instability: definition and correlation with mismatch repair
protein expression. Cancer Res., 57: 47494756, 1997.
21. Bocker, T., Diermann, J., Friedl, W., Gebert, J., Holinski-Feder, E., Karner-Hanusch,
J., von Knebel-Doeberitz, M., Koelble, K., Moeslein, G., Schackert, H. K., Wirtz,
H. C., Fishel, R., and Ruschoff, J. Microsatellite instability analysis: a multicenter
study for reliability and quality control. Cancer Res., 57: 47394743, 1997.
22. Thibodeau, S. N., French, A. J., Cunningham, J. M., Tester, D., Burgart, L. J., Roche,
P. C., McDonnell, S. K., Schaid, D. J., Vockley, C. W., Michels, V. V., Farr, G. H.,
Jr., and O’Connell, M. J. Microsatellite instability in colorectal cancer: different
mutator phenotypes and the principle involvement of. hMLH1. Cancer Res., 58:
1713–1718, 1998.
23. Hoang, J-M., Cottu, P. H., Thuille, B., Salmon, R. J., Thomas, G., and Hamelin, R.
BAT-26, an indicator of the replication error phenotype in colorectal cancers and cell
lines. Cancer Res., 57: 300–303, 1997.
24. Aaltonen, L. A., Salovaara, R., Kristo, P., Canzian, F., Hemminki, A., Peltoma¨ki, P.,
Chadwick, R. B., Percesepe, A., Ka¨a¨ria¨inen, H., Ahtola, H., Eskelinen, M., Ha¨rko¨nen,
N., Julkunen, R., Kangas, E., Ojala, S., Tulikoura, J., Valkamo, E., Ja¨rvinen, H.,
Mecklin, J-P., and de la Chapelle, A. Incidence of hereditary nonpolyposis colorectal
cancer, and molecular screening for the disease. N. Engl. J. Med., 338: 1481–1487,
1998.
25. Sutter, C., Gebert, J., Bischoff, P., Herfarth, C., and von Knebel Doeberitz, M.
Molecular screening of potential HNPCC patients using a multiplex microsatellite
PCR system. Mol. Cell Probes, 13: 157–165, 1999.
26. Samowitz, W. S., Slattery, M. L., Potter, J. D., and Leppert, M. F. BAT-26 and
BAT-40 instability in colorectal adenomas and carcinomas and germline polymor-
phisms. Am. J. Pathol., 154: 1637–1641, 1999.
27. Zhou, X. P., Hoang, J. M., Li, Y. J., Seruca, R., Carneiro, F., Sobrinho-Simoes, M.,
Lothe, R. A., Gleeson, C. M., Russell, S. E., Muzeau, F., Flejou, J. F., Hoang-Xuan,
K., Liderau, R., Thomas, G., and Hamelin, R. Determination of the replication error
phenotype in human tumors without the requirement for matching normal DNA by
analysis of mononucleotide repeat microsatellites. Genes Chromosomes Cancer, 21:
101–107, 1998.
28. Pyatt, R., Chadwick, R. B., Johnson, C. K., Adebamowo, C., de la Chapelle, A., and
Prior, T. W. Polymorphic variation at the BAT-25 and BAT-26 loci in individuals of
African origin. Am. J. Pathol., 155: 349–353, 1999.
29. Salovaara, R., Loukola, A., Kristo, P., Ka¨a¨ria¨inen, H., Ahtola, H., Eskelinen, M.,
Ha¨rko¨nen, N., Julkunen, R., Kangas, E., Ojala, S., Tulikoura, J., Valkamo, E.,
Ja¨rvinen, H., Mecklin, J-P., Aaltonen, L. A., and de la Chapelle, A. Population-based
molecular detection of hereditary nonpolyposis colorectal cancer. J. Clin. Oncol., 18:
2193–2200, 2000.
30. Canzian, F., Salovaara, R., Hemminki, A., Kristo, P., Chadwick, R. B., Aaltonen,
L. A., and de la Chapelle, A. Semiautomated assessment of loss of heterozygosity and
replication error in tumors. Cancer Res., 56: 3331–3337, 1996.
31. Jass, J. R., Cottier, D. S., Jeevaratnam, P., Pokos, V., Holdaway, K. M., Bowden,
M. L., Van de Water, N. S., and Browett, P. J. Diagnostic use of microsatellite
instability in hereditary non-polyposis colorectal cancer. Lancet, 346: 1200–1201,
1995.
32. Liu, T., Wahlberg, S., Burek, E., Lindblom, P., Rubio, C., and Lindblom, A.
Microsatellite instability as a predictor of a mutation in a DNA mismatch repair gene
in familial colorectal cancer. Genes Chromosomes Cancer, 27: 17–25, 2000.
33. Lamberti, C., Kruse, R., Ruelfs, C., Caspari, R., Wang, Y., Jungck, M., Mathiak, M.,
Malayeri, H. R., Friedl, W., Sauerbruch, T., and Propping, P. Microsatellite instabil-
ity: a useful diagnostic tool to select patients at high risk for hereditary non-polyposis
colorectal cancer: a study in different groups of patients with colorectal cancer. Gut,
44: 839843, 1999.
34. Wu, Y., Berends, M. J., Mensink, R. G., Kempinga, C., Sijmons, R. H., van Der Zee,
A. G., Hollema, H., Kleibeuker, J. H., Buys, C. H., and Hofstra, R. M. Association of
hereditary nonpolyposis colorectal cancer-related tumors displaying low microsatel-
lite instability with MSH6 germline mutations. Am. J. Hum. Genet., 65: 1291–1298,
1999.
35. Mangold, E., Mathiak, M., Friedl, W., Lamberti, C., Jungck, M., Steinhoff, M.,
Sengteller, H-P., and Propping, P. Correlation between immunohistochemistry for
MLH1/MSH2 and microsatellite instability in tumour tissue of HNPCC patients with
known germline mutations. In: Abstract Book of the 12
th
ICG-HNPCC Meeting in
Tiberias, Galilee, Israel, p. 31. , 2000.
36. Shibata, D., and Aaltonen, L. A. Genetic predisposition and somatic diversification in
tumor development and progression. Adv. Cancer Res., 80: 83–114, 2001.
4549
MICROSATELLITE MARKERS IN SCREENING FOR HNPCC
on June 4, 2013. © 2001 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from
... Converted DNA was then subjected to desulphonation and clean-up using washing buffer. DNA (approximately 10 μL) was eluted and collected for methylation specific PCR (MS-PCR) [15]. Reaction was carried out using Qiagen Taq Core Kit in a reaction volume of 25 μL with 10x PCR Buffer, 2.5 mmol/L of MgCl 2 , 50 pmol of primer, 1.25 mmol/L of dNTPs and 1.25 units of Taq DNA polymerase added to 1.5-2 μL of converted DNA. ...
... Reaction was carried out using Qiagen Taq Core Kit in a reaction volume of 25 μL with 10x PCR Buffer, 2.5 mmol/L of MgCl 2 , 50 pmol of primer, 1.25 mmol/L of dNTPs and 1.25 units of Taq DNA polymerase added to 1.5-2 μL of converted DNA. Primer sets used for hMLH1 MS-PCR are taken from published work of Fox et al [15]. December 15, 2021 Volume 13 Issue 12 ...
Article
Full-text available
Background: Colorectal cancer (CRC) ranks third in terms of incidence and second in mortality worldwide. In CRC, the silencing of mismatch repair genes, including the mutL homolog 1 (hMLH1) has been linked to microsatellite instability (MSI), the lengthening or shortening of microsatellite repeats. Very limited data have been presented so far on the link of hMLH1 methylation and MSI in Southeast Asia populations with sporadic CRC, and on its clinical significance. Aim: To investigate the significance of the MSI status and hMLH1 methylation in CRC Filipino patients. Methods: Fifty-four sporadic CRC patients with complete clinical data were included in this study. Genomic DNA from CRC tumor biopsies and their normal tissue counterparts were profiled for MSI by high resolution melting (HRM) analysis using the Bethesda Panel of Markers (BAT25, BAT26, D2S123, D5S346, and D17S250). hMLH1 methylation screening was performed using bisulfite conversion and methylation specific polymerase chain reaction. Statistical analysis was conducted to calculate their associations to clinicopathological characteristics and survival relevance (Kaplan-Meier curves and the log-rank test). Results: hMLH1 methylation was observed in 9% and 35% of CRC and normal samples, respectively. Higher incidence of consistently methylated hMLH1 found in both normal and CRC was noticed for relation to location of tumor (P < 0.05). As for MSI status, D2S123 the most common unstable microsatellite and MSI-high (MSI-H) was the most common MSI profile, counted for 46% and 50% of normal and CRC tissues, respectively. The presence of MSI-low (MSI-L) and microsatellite stable (MSS) was 43% and 11% for normal, and 31% and 19% for CRC samples. The mean month of patients' survival was shorter in patients whose normal and tumor tissues had methylated compared to those with unmethylated hMLH1 and with MSI-H compared to those with MSI-L/MSS (P < 0.05). This was supported by significant difference in Kaplan-Meier with log-rank analysis. This data indicated that hMLH1 methylation and high MSI status have prognostic value. Conclusion: This study showed the clinical significance of hMLH1 methylation and MSI status in sporadic CRC Filipino patients, especially in the normal part of the tumor.
... Discrepant samples between IHC and Idylla test were further analyzed with RER test using BAT25 and BAT26 mononucleotide repeats, in which 10-μm FFPE tissue flakes were cut from whenever 50% or more tumor cells [12] could be retrieved and DNA was extracted from the deparaffinized flakes by using a Maxwell® CSC Blood DNA Kit (Promega Corporation, Madison, WI) according to the manufacturer's instructions. MSI status was then assessed using the two mononucleotide-repeat markers using fluorescently labeled PCR. ...
... As a third method to evaluate the MSI phenotype, the discrepant cases (from EC Set I to Set II) were further tested for RER status using BAT25 and BAT26 microsatellite markers (Table 6). For this analysis, two samples (B14 and C26) did not meet the required 50% tumor cell content [12], and two (B107 and C9) did not have enough tissue available. Of the 14 samples that were subjected to RER analysis, four were shown to be positive for both BAT25 and BAT26, indicating MSI, and rest of them were negative (Table 6). ...
Article
Full-text available
Universal testing of microsatellite instability (MSI) is recommended for colorectal cancer (CRC) and endometrial cancer (EC) to screen for Lynch syndrome and to aid in assessing prognosis and optimal treatment. We compared the performance of Idylla MSI test to immunohistochemistry (IHC) of mismatch repair (MMR) proteins in consecutive series of 100 CRC and 108 EC samples, as well as in retrospective series of 28 CRC and 33 EC specimens with known deficient MMR protein expression. The concordance between the Idylla test and IHC was 100% in all CRC samples ( n =128) but lower in EC samples (87.2%; n =141). In the EC samples, sensitivity of Idylla test was 72.7% and specificity 100%. EC MSI/dMMR agreement was 85.4% for MLH1, 87.5% for MSH2, and only 35.3% for MSH6. When we analyzed 14 EC samples that were discrepant, i.e., dMMR using IHC and microsatellite stable using Idylla, with microsatellite markers BAT25 and BAT26, we found four cases to be replication error (RER) positive. All RER positive cases were deficient for MSH6 protein expression. We also re-analyzed EC samples with variable tumor cellularity to determine the limit of detection of the Idylla test and found that a 30% or higher tumor cellularity is required. We conclude that Idylla MSI test offers a sensitive and specific method for CRC diagnostics but is less sensitive in EC samples especially in the case of MSH6 deficiency.
... In contrast, all LS CRCs were determined to be MSI. Markers were selected based on previous studies where they had been shown to function as sensitive and specific markers of high-degree MSI [78][79][80] and shown high concordance between MSI status and loss of MMR protein expression in Lynch syndrome-associated colorectal tumours. 11,81 Also in our analyses, all tumours identified as unstable according to MSI analysis showed a deficiency of corresponding MMR protein in IHC analysis. ...
Article
Full-text available
Background Lynch syndrome (LS) is one of the most common hereditary cancer syndromes worldwide. Dominantly inherited mutation in one of four DNA mismatch repair genes combined with somatic events leads to mismatch repair deficiency and microsatellite instability (MSI) in tumours. Due to a high lifetime risk of cancer, regular surveillance plays a key role in cancer prevention; yet the observation of frequent interval cancers points to insufficient cancer prevention by colonoscopy-based methods alone. This study aimed to identify precancerous functional changes in colonic mucosa that could facilitate the monitoring and prevention of cancer development in LS. Methods The study material comprised colon biopsy specimens (n = 71) collected during colonoscopy examinations from LS carriers (tumour-free, or diagnosed with adenoma, or diagnosed with carcinoma) and a control group, which included sporadic cases without LS or neoplasia. The majority (80%) of LS carriers had an inherited genetic MLH1 mutation. The remaining 20% included MSH2 mutation carriers (13%) and MSH6 mutation carriers (7%). The transcriptomes were first analysed with RNA-sequencing and followed up with Gorilla Ontology analysis and Reactome Knowledgebase and Ingenuity Pathway Analyses to detect functional changes that might be associated with the initiation of the neoplastic process in LS individuals. Findings With pathway and gene ontology analyses combined with measurement of mitotic perimeters from colonic mucosa and tumours, we found an increased tendency to chromosomal instability (CIN), already present in macroscopically normal LS mucosa. Our results suggest that CIN is an earlier aberration than MSI and may be the initial cancer driving aberration, whereas MSI accelerates tumour formation. Furthermore, our results suggest that MLH1 deficiency plays a significant role in the development of CIN. Interpretation The results validate our previous findings from mice and highlight early mitotic abnormalities as an important contributor and precancerous marker of colorectal tumourigenesis in LS. Funding This work was supported by grants from the 10.13039/501100004012Jane and Aatos Erkko Foundation, the 10.13039/501100002341Academy of Finland (330606 and 331284), 10.13039/501100010711Cancer Foundation Finland sr, and the 10.13039/501100006306Sigrid Jusélius Foundation. Open access is funded by Helsinki University Library.
... Recent studies indicate that individuals with CRC who display MSI have a favourable prognosis, but is usually absent in 80-85% of CRCs. 16 MSI-H CRC is associated with immune checkpoint upregulation such as programmed cell death receptor (PD-1) and programmed death ligand (PDL-1), the expression of which is usually used as an indication to use immunotherapeutic PD-1 blocking agents e.g. pembrolizumab (Keytruda) and dostarlimab (Jemperli), the former is the first-line therapeutic agent in advanced metastatic or inoperable CRCs. ...
Article
Full-text available
Objectives: To identify the frequency and types of microsatellite instability among a group of sporadic CRC patients and to correlate the findings with clinicopathological characteristics. Methods: During an 8-month period, all patients with sporadic CRC who attended to two teaching hospitals in Baghdad, Iraq were recruited to this cross-sectional study regardless of age, sex, ethnicity, or tumor characteristics. Demographic, clinical, and histopathological features were recorded. DNA was extracted from FFPE-blocks of the resected tumors and normal tissues. PCR amplification of five microsatellite mononucleotide repeat loci (BAT25, BAT26, NR-21, NR-24, and MONO-27) and 2 pentanucleotide repeat control markers (Penta C and Penta D) was performed to determine the MSI status. Capillary electrophoresis and Genetic Analyzer 3500 (Applied Biosystem, Japan) were used to separate and examine the products. Data were analyzed by Genescan software (Promega, USA). Instability of two or more loci is considered MSI-H. Result: In this study, ages of the 45 recruited patients ranged between 20-80 years, with a mean±SD of 55±12.3 years; of them, 31(68.9%) were ≥50 years; 25 (55.6%) were males. Rectal bleeding was the most frequent presenting feature [22 (48.9%)] patients; 23 (51.1%) of CRCs were located at recto-sigmoid region, 29 (64.4%) were T3 tumors, 34(75.5%) were non-mucinous adenocarcinoma, 39(86.7%) were moderately differentiated, 17 (37.8%) patients had stage III tumors; and 25 (55.5%) had lymphovascular invasion. MSI-H was seen in 5/45 (11.1%) patients; 3(60%) of them were ≥50 years, 4(80%) were males, 3(60%) were smokers, 2 (40%) presented with intestinal obstruction and altered bowel habits each; 4(80%) had T3 tumors, 3(60%) had mucinous adenocarcinomas [p=0.004], 2(40%) had stage II tumor and stage III each. Conclusion: The frequency of MSI-H among the recruited patients with CRC was 5/45 (11.1%) and it was significantly associated with mucinous adenocarcinoma subtype. NR-24 and NR-21 were the most prevalent instable markers.
... Lynch syndrome (LS) is the most common cause of hereditary colon cancer, with a prevalence noted to be as high as 1 in 279 individuals in the general population and as common as 1 in 35 patients with colon cancer [5,6]. The overwhelming majority of colon cancers arising in this setting exhibit MSI and develop proximal to the splenic flexure [7][8][9][10]. Synchronous and metachronous tumors are characteristic of this syndrome, with individuals typically in their mid-40s ...
Article
Full-text available
Purpose of Review Patients with Lynch syndrome have a high probability of developing colorectal and other carcinomas. This review provides a comprehensive assessment of the immunologic aspects of Lynch syndrome pathogenesis and provides an overview of potential immune interventions for patients with Lynch syndrome polyps and Lynch syndrome–associated carcinomas. Recent Findings Immunogenic properties of the majority of Lynch syndrome polyps and associated cancers include microsatellite instability leading to a high mutational burden and the development of novel frameshift peptides, i.e., neoantigens. In addition, patients with Lynch syndrome develop T cell responses in the periphery and in the tumor microenvironment (TME) to tumor-associated antigens, and a proinflammatory cytokine TME has also been identified. However, Lynch syndrome lesions also possess immunosuppressive entities such as alterations in MHC class I antigen presentation, TGFβ receptor mutations, regulatory T cells, and upregulation of PD-L1 on tumor-associated lymphocytes. Summary The rich immune microenvironment of Lynch syndrome polyps and associated carcinomas provides an opportunity to employ the spectrum of immune-mediating agents now available to induce and enhance host immune responses and/or to also reduce immunosuppressive entities. These agents can be employed in the so-called prevention trials for the treatment of patients with Lynch syndrome polyps and for trials in patients with Lynch syndrome–associated cancers.
... Analysis for MSI was performed using a modified procedure by Loukola et al. 15 Two sets of primers from the Bethesda panel of markers were used, BAT25 and BAT26. They were chosen because they are the best-known mononucleotide markers for MSI testing. ...
Article
Background: Microsatellite instability (MSI) represents as a molecular hallmark of deficient MMR system at the genomic level. Increasing clinical significance of MSI status highlights the necessity of simple, accurate markers for detection. Although 2B3D NCI panel is the most widely applied, it has been questioned whether the performance of NCI panel is second to none in MSI detection. Methods: We evaluated the efficacy of the NCI panel versus a 6-mononucleotide site panel (BAT25, BAT26, NR21, NR24, NR27, and MONO-27) in assessing MSI status of 468 Chinese patients with CRC, and compared MSI test results with the results by immunohistochemistry of four MMR proteins (MLH1, PMS2, MSH2, MSH6) in the present study. Clinicopathological variables were also collected, and their associations with MSI or MMR proteins status were analyzed using either the chi-square test or the Fisher's exact test. Results: MSI-H/dMMR was significantly associated with right colon involvement, poor differentiation, early stage, mucinous adenocarcinoma, negative lymph node, less neural invasion, and KRAS/NRAS/BRAF wild-type. As to the efficiency of detecting deficient MMR system, both panels had good concordance with MMR proteins expression by IHC, and 6-mononucleotide site panel outperformed NCI panel in sensitivity, specificity, positive predictive value, and negative predictive value numerically despite the lack of statistical significance. The advantage was more obvious in the sensitivity and specificity analyses of each single microsatellite markers from 6-mononucleotide site panel in comparison with NCI panel. Additionally, the rate of MSI-L detected by 6-mononucleotide site panel was much lower than that detected by the NCI panel (0.64% vs. 2.86%, P = 0.0326). Conclusion: 6-mononucleotide site panel had a greater ability to help resolve cases of MSI-L into either MSI-H or MSS. We propose that 6-mononucleotide site panel may be potentially more suitable than NCI panel for Chinese CRC population. Large-scale studies are warranted to validate our findings.
Article
Background Lynch syndrome (LS) is the first cause of inherited colorectal cancer (CRC), being responsible for 2–4% of all diagnoses. Identification of affected individuals is important as they have an increased lifetime risk of multiple CRC and other neoplasms, however, LS is consistently underdiagnosed at the population level. We aimed to evaluate the yield of LS screening in CRC in a single-referral centre and to identify the barriers to its effective implementation. Methods LS screening programme included individuals with CRC < 70 years, multiple CRC, or endometrial cancer at any age. Mismatch repair (MMR) protein immunohistochemistry (IHC) analysis was performed in routine practice on the surgical specimen and, if MLH1 IHC was altered, MLH1 gene promoter methylation was analysed. Results were collected in the CRC multidisciplinary board database. LS suspected individuals (altered MMR IHC without MLH1 promoter methylation) were referred to the Cancer Genetic Counselling Unit (CGCU). If accepted, a genetic study was performed. Two checkpoints were included: review of the pathology data and verification of patient referral by a genetic counsellor. Results Between 2016 and 2019, 381 individuals were included. MMR IHC analysis was performed in 374/381 (98.2 %) CRC cases and MLH1 promoter methylation in 18/21 (85.7 %). Seventeen of the 20 LS suspected individuals were invited for referral at the CGCU. Two cases were not invited and the remaining patient died of cancer before completion of tumour screening. Fifteen individuals attended and a genetic analysis was performed in 15/20 (75 %) LS suspected individuals. Ten individuals were diagnosed with LS, in concordance with the IHC profile (2.7 % of the total cohort). This led to cascade testing in 58/75 (77.3 %) of the available adult relatives at risk, identifying 26 individuals with LS. Conclusions Establishing a standardized institutional LS screening programme with checkpoints in the workflow is key to increasing the yield of LS identification.
Article
Full-text available
Objective: Hepatocellular carcinoma (HCC) accounts for more than 80% of primary liver cancers. Moreover, in the next 10 years, more than one million patients are expected to die from liver cancer as estimated by the World Health Organization. The aim of the present study is to define the microsatellite phenotype in the blood, tumor and nontumor tissue samples from hepatocellular carcinoma cases to develop a simple non-invasive method for diagnosis and detection of the disease. Methods: A total of 100 patients with histologically-proven HCC were enrolled in this study, blood samples and tissue specimens from tumor and nontumor tissue were obtained from each patient. DNA was extracted and microsatellite instability MSI status was determined by polymerase chain reaction (PCR) using 5 mononucleotide and 5 dinucleotide repeats. Results: Among the 100 HCC tumors analyzed, (8%) considered as displaying a typical MSI-H phenotype as defined by instability in at least 3 of the 10 repeats analyzed, (61%) tumors displayed MSI-L and (31%) displayed MSS while in plasma the instability was (40%) for MSI-H, (44%) for MSI-L and (16%) for MSS. Conclusion: our findings could point to the achievement that HCC patients could be diagnosed by MSI analysis using blood sample as non-invasive way and this conclusion achieved our aim as the study shows impressive and promising results.
Article
Full-text available
Ulcerative colitis increases colorectal cancer risk by mechanisms that remain incompletely understood. We approached this question by determining the genetic and epigenetic profiles of colitis‐associated colorectal carcinomas (CA‐CRC). The findings were compared to Lynch syndrome (LS), a different form of cancer predisposition that shares the importance of immunological factors in tumorigenesis. CA‐CRCs (n = 27) were investigated for microsatellite instability, CpG island methylator phenotype and somatic mutations of 999 cancer‐relevant genes (“Pan‐cancer” panel). A subpanel of “Pan‐cancer” design (578 genes) was used for LS colorectal tumors (n = 28). Mutational loads and signatures stratified CA‐CRCs into three subgroups: hypermutated microsatellite‐unstable (Group 1, n = 1), hypermutated microsatellite‐stable (Group 2, n = 9) and nonhypermutated microsatellite‐stable (Group 3, n = 17). The Group 1 tumor was the only one with MLH1 promoter hypermethylation and exhibited the mismatch repair deficiency‐associated Signatures 21 and 15. Signatures 30 and 32 characterized Group 2, whereas no prominent single signature existed in Group 3. TP53, the most common mutational target in CA‐CRC (16/27, 59%), was similarly affected in Groups 2 and 3, but DNA repair genes and Wnt signaling genes were mutated significantly more often in Group 2. In LS tumors, the degree of hypermutability exceeded that of the hypermutated CA‐CRC Groups 1 and 2, and somatic mutational profiles and signatures were different. In conclusion, Groups 1 (4%) and 3 (63%) comply with published studies, whereas Group 2 (33%) is novel. The existence of molecularly distinct subgroups within CA‐CRC may guide clinical management, such as therapy options.
Article
Full-text available
A replication error (RER) phenotype has been documented both in sporadic colorectal tumors and in tumors from patients with hereditary nonpolyposis colorectal cancer (HNPCC). In the current study 8 of 49 (16%) sporadic colorectal cancers (CRCs) and 25 of 29 (86%) CRCs from HNPCC patients were found to be RER+. All 9 (100%) CRCs from HNPCC patients with germline mutations of the mismatch repair gene MSH2 were found to be RER+, while 16 of 20 CRCs from HNPCC kindreds unlinked or not studied for linkage to MSH2 were RER+. Corresponding analysis in colorectal adenomas revealed that only 1 of 33 (3%) sporadic tumors but 8 of 14 (57%) HNPCC tumors were RER+. Moreover, RER was found in all 6 extracolonic cancers (endometrium, 2; kidney, 1; stomach, 1; duodenum, 1; and ovary, 1) derived from members of HNPCC families. These data suggest the involvement of mismatch repair deficiency in the premalignant stage of tumorigenesis in HNPCC cases, and suggest that mismatch repair genes (MSH2 or others) are defective in the germline of nearly all these patients.
Article
Full-text available
Recent reports have suggested that one or more genes may cause replication errors (RER) during colorectal tumorigenesis. Additional alleles are seen in the tumors when analyzing random microsatellite loci. We have studied seven dinucleotide repeat loci, located on seven different chromosomes, by use of polymerase chain reaction amplification and denaturing polyacrylamide gel electrophoresis. We found that 16.5% (40 of 243) colorectal cancers showed RER at one or several loci (RER+). This includes 31% (4 of 13) among cases with a strong positive family history according to previously published criteria and 17% (35 of 207) among cases with no history of familial cancer. Interestingly, no significant association was found between RER+ tumors and a general familial clustering of cancer. Microsatellite instability was significantly associated with DNA diploid status of the tumor (P < 0.001), with the location of the tumor in the proximal colon (P < 0.001), and with poorly differentiated tumor phenotype (P < 0.001). Patients with RER+ at > or = 2 loci tumors had an increased survival (P = 0.05). We further analyzed 84 breast cancers and 86 male germ cell cancers using the same seven markers. None of the tumors were RER+, indicating that this phenomenon may be specific to certain types of tumors.
Article
SummaryHereditary nonpolyposis colorectal cancer (HNPCC) (Amsterdam criteria) is often caused by mutations in mismatch repair (MMR) genes, and tumors of patients with HNPCC show microsatellite instability (MSI-high phenotype). Germline mutations of MMR genes have rarely been found in families that have HNPCC or suspected HNPCC and that do not show microsatellite instability (MSI-low phenotype). Therefore, an MSI-high phenotype is often used as an inclusion criterion for mutation testing of MMR genes. Correction of base-base mismatches is the major function of MSH6. Since mismatches present with an MSI-low phenotype, we assumed that the phenotype in patients with HNPCC-related tumors might be associated with MSH6 germline mutations. We divided 36 patients with suspected HNPCC into an MSI-low group (n=18) and an MSI-high group (n=18), on the basis of the results of MSI testing. Additionally, three unrelated patients from Amsterdam families with MSI-low tumors were investigated. All patients were screened for MSH2, MLH1, and MSH6 mutations. Four presumably causative MSH6 mutations were detected in the patients (22%) who had suspected HNPCC and MSI-low tumors. Furthermore, we detected one frameshift mutation in one of the three patients with HNPCC and MSI-low tumors. In the MSI-high group, one MSH6 missense mutation was found, but the same patient also had an MLH1 mutation, which may explain the MSI-high phenotype. These results suggest that MSH6 may be involved in a substantial proportion of patients with HNPCC or suspected HNPCC and MSI-low tumors. Our data emphasize that an MSI-low phenotype cannot be considered an exclusion criterion for mutation testing of MMR genes in general.
Article
50 families with a history of colorectal cancer were divided according to whether criteria for hereditary non-polyposis colorectal cancer (HNPCC) were fulfilled totally (A, n = 19) or partly (B, n = 31) and stratified by the demonstration that at least half the cancers tested per family were positive for DNA replication errors (RER+). Accepted clinical and pathological characteristics of HNPCC were found to cluster within 12 A/RER+ families in which the mean number of affected individuals per family was 10.1. Reliance upon clinical data alone may result in over-diagnosis of HNPCC, in small families who just meet the minimum criteria, whereas underdiagnosis is rare. The criteria could be refined by inclusion of RER status.
Article
The inherited susceptibility to hereditary nonpolyposis colorectal cancer (HNPCC) provides an opportunity for secondary prevention of colorectal cancer (CRC) in family members who are at 50% lifetime risk. The aim of this study was to evaluate the effectiveness of long-term screening during a 10-year period. The CRC and death rates were compared between two groups of asymptomatic at-risk members of 22 families with HNPCC: 133 subjects screened at 3-year intervals by colonoscopy or barium enema and sigmoidoscopy and 118 control subjects without screening. The screening was complete in 118 subjects (89%), whereas 18 control subjects (15%) had screening examinations outside of the study. CRC occurred in 6 study subjects (4.5%) and in 14 controls (11.9%; P = 0.03), a difference of 7.4% in favor of the study group, which corresponds to a reduction by 62% that is presumably because of polypectomies. The tumor stage was more favorable in the screening group with no deaths caused by CRC compared with 5 of 14 cases in controls. Overall, there were 6 and 12 deaths within the 10-year period in the study and control groups, respectively (P = 0.08). The 3-year interval screening more than halves the CRC rate in at-risk members of families with HNPCC and seems to prevent CRC deaths.
Article
It has been hypothesized that in some but not all families with hereditary nonpolyposis colorectal cancer (HNPCC) there is a high risk of certain cancers other than colon cancer. The authors compared the observed frequency of cancer at specific sites in more than 1300 high-risk members of 23 kindreds with HNPCC with expectations based on general population incidence and evaluated the hypothesis that there was heterogeneity in cancer frequency among families. The authors observed significantly increased numbers of cancers of the stomach, small intestine, upper urologic tract (renal pelvis and ureter), and ovary. No excess was seen in other cancer types that have been associated previously with HNPCC, including cancer of the breast, pancreas, and urinary bladder. Significant heterogeneity among families was observed in the frequencies of endometrial, ovarian, and upper urologic system cancer. In addition to early onset cancers of the colorectum, HNPCC family members are at increased risk for cancers of other gastrointestinal tract organs, and, especially in some families, cancers of the upper urologic and female genital tract.
Article
Colorectal tumor DNA was examined for somatic instability at (CA)n repeats on human chromosomes 5q, 15q, 17p, and 18q. Differences between tumor and normal DNA were detected in 25 of the 90 (28 percent) tumors examined. This instability appeared as either a substantial change in repeat length (often heterogeneous in nature) or a minor change (typically two base pairs). Microsatellite instability was significantly correlated with the tumor's location in the proximal colon (P = 0.003), with increased patient survival (P = 0.02), and, inversely, with loss of heterozygosity for chromosomes 5q, 17p, and 18q. These data suggest that some colorectal cancers may arise through a mechanism that does not necessarily involve loss of heterozygosity.
Article
A predisposition to colorectal cancer is shown to be linked to markers on chromosome 2 in some families. Molecular features of "familial" cancers were compared with those of sporadic colon cancers. Neither the familial nor sporadic cancers showed loss of heterozygosity for chromosome 2 markers, and the incidence of mutations in KRAS, P53, and APC was similar in the two groups of tumors. Most of the familial cancers, however, had widespread alterations in short repeated DNA sequences, suggesting that numerous replication errors had occurred during tumor development. Thirteen percent of sporadic cancers had identical abnormalities and these cancers shared biologic properties with the familial cases. These data suggest a mechanism for familial tumorigenesis different from that mediated by classic tumor suppressor genes.
Article
Microsatellite instability (replication error [RER]) is a characteristic of tumors in hereditary nonpolyposis colon cancer (HNPCC), but the mechanism of HNPCC carcinogenesis is not yet understood. To clarify the nature of HNPCC tumors, RER and genetic changes were compared between HNPCC and non-HNPCC tumors. RER and genetic changes were analyzed in 21 HNPCC, 389 familial adenomatous polyposis, and 206 sporadic tumors using polymerase chain reaction, single-strand conformation polymorphism, sequencing, and Southern hybridization. RESULTS. in HNPCC, 95% tumors at all stages showed RER positivity (altered loci, 4.3 of 5). In familial adenomatous polyposis and sporadic tumors, RER positivity (1.7 of 5) was 3% in adenoma and intramucosal carcinoma, 13%-24% in invasive carcinoma, and 35% in carcinoma metastasized to liver. Fifty percent of RER-positive HNPCC tumors had both germline and somatic mutations of hMSH2 or hMLH1 gene, whereas 6% of RER-positive non-HNPCC had somatic mutation. APC, p53, and K-ras-2 mutations and loss of heterozygosity of tumor-suppressor genes were significantly less frequent (P = 0.03 to 0.0006) but transforming growth factor beta type II receptor mutation was significantly more frequent (P = 0.000001) in HNPCC than in non-HNPCC. RER positivity occurs from an early stage of carcinogenesis in HNPCC but in later stages in non-HNPCC. Most HNPCC tumors may develop through different genetic changes from those in the adenoma-carcinoma sequence, although a certain percentage develops through APC mutation.