ArticlePDF Available

Microsatellite Instability and Mutation of DNA Mismatch Repair Genes in Gliomas

Authors:

Abstract and Figures

Microsatellite instability (MSI) has been identified in various human cancers, particularly those associated with the hereditary nonpolyposis colorectal cancer syndrome. Although gliomas have been reported in a few hereditary nonpolyposis colorectal cancer syndrome kindred, data on the incidence of MSI in gliomas are conflicting, and the nature of the mismatch repair (MMR) defect is not known. We established the incidence of MSI and the underlying MMR gene mutation in 22 patients ages 45 years or less with sporadic high-grade gliomas (17 glioblastomas, 3 anaplastic astrocytomas, and 2 mixed gliomas, grade III). Using five microsatellite loci, four patients (18%) had high level MSI, with at least 40% unstable loci. Germline MMR gene mutation was detected in all four patients, with inactivation of the second allele of the corresponding MMR gene or loss of protein expression in the tumor tissue. Frameshift mutation in the mononucleotide tract of insulin-like growth factor type II receptor was found in one high-level MSI glioma, but none was found in the transforming growth factor beta type II receptor and the Bax genes. There was no family history of cancer in three of the patients, and although one patient did have a family history of colorectal carcinoma, the case did not satisfy the Amsterdam criteria for hereditary nonpolyposis colorectal cancer syndrome. Three patients developed metachronous colorectal adenocarcinomas, fitting the criteria of Turcot's syndrome. Thus, MSI and germline MMR gene mutation is present in a subset of young glioma patients, and these patients and their family members are at risk of developing other hereditary nonpolyposis colorectal cancer syndrome-related tumors, in particular colorectal carcinomas. These results have important implications in the genetic testing and management of young patients with glioma and their families.
Content may be subject to copyright.
Microsatellite Instability and Mutation of DNA
Mismatch Repair Genes in Gliomas
Suet Yi Leung,* Tsun Leung Chan,*
Lap Ping Chung,*
Annie S. Y. Chan,*
Yiu Wah Fan,
Kwan Ngai Hung,
Wai Kay Kwong,
§
Judy W. C. Ho,
†‡
and Siu Tsan Yuen*
From the Departments of Pathology,*Surgery,
and Radiation
Oncology,
§
and the Hereditary Gastrointestinal Cancer Registry,
Queen Mary Hospital, The University of Hong Kong, Hong Kong
Microsatellite instability (MSI) has been identified in
various human cancers, particularly those associated
with the hereditary nonpolyposis colorectal cancer
syndrome. Although gliomas have been reported in a
few hereditary nonpolyposis colorectal cancer syn-
drome kindred, data on the incidence of MSI in glio-
mas are conflicting, and the nature of the mismatch
repair (MMR) defect is not known. We established the
incidence of MSI and the underlying MMR gene mu-
tation in 22 patients ages 45 years or less with spo-
radic high-grade gliomas (17 glioblastomas, 3 ana-
plastic astrocytomas, and 2 mixed gliomas, grade III).
Using five microsatellite loci, four patients (18%) had
high level MSI, with at least 40% unstable loci. Germ-
line MMR gene mutation was detected in all four pa-
tients, with inactivation of the second allele of the
corresponding MMR gene or loss of protein expres-
sion in the tumor tissue. Frameshift mutation in the
mononucleotide tract of insulin-like growth factor
type II receptor was found in one high-level MSI gli-
oma, but none was found in the transforming growth
factor
b
type II receptor and the Bax genes. There was
no family history of cancer in three of the patients,
and although one patient did have a family history of
colorectal carcinoma, the case did not satisfy the Am-
sterdam criteria for hereditary nonpolyposis colorec-
tal cancer syndrome. Three patients developed meta-
chronous colorectal adenocarcinomas, fitting the
criteria of Turcot’s syndrome. Thus, MSI and germline
MMR gene mutation is present in a subset of young
glioma patients, and these patients and their family
members are at risk of developing other hereditary
nonpolyposis colorectal cancer syndrome-related tu-
mors, in particular colorectal carcinomas. These
results have important implications in the genetic
testing and management of young patients with
glioma and their families. (Am J Pathol 1998,
153:1181–1188)
Microsatellite instability (MSI) is characterized by the ex-
pansion and contraction of small repeat sequences dur-
ing DNA replication and is present in the majority of
tumors in the hereditary nonpolyposis colon cancer
(HNPCC) syndrome.
1
HNPCC is characterized by familial
occurrence of cancer in various sites, including the co-
lon, endometrium, and urinary tract, at an early age.
2
The
mechanism leading to MSI is related to a defect in the
DNA mismatch repair (MMR) system, of which more than
5 DNA mismatch repair (MMR) genes are now known.
3–12
Mutation of the hMSH2 and hMLH1 genes accounts for
more than 60% of individuals with the syndrome.
13
Many sporadic cancers have also been found to show
MSI.
14
For gliomas, relatively few studies have been per-
formed, and the results are conflicting. Izumoto et al
15
and Dams et al
16
demonstrated the presence of MSI in 20
to 45% of glioblastomas and anaplastic astrocytomas
and in no low-grade astrocytomas. Zhu et al
17
found MSI
in 17% of oligodendrogliomas and 3% of astrocytic tu-
mors. Wooster et al
18
and Amariglio et al
19
found MSI in
1.9% of brain tumors and in no brain tumors, respectively.
In most of these series, MSI was considered positive if
there was an allelic shift in a single locus only. The status
of the MMR gene, be it germline or somatic, is largely
unknown for these MSI-positive gliomas. On the other
hand, there have been reports of increased risk for brain
tumors in HNPCC kindred,
20–22
and a few patients with
Turcot’s syndrome, characterized by the development of
both colorectal and brain cancers, have been shown to
have MSI and to harbor germline mutation in the MMR
genes.
23,24
We have previously reported an unusually high inci-
dence of colorectal carcinoma in the young Hong Kong
population.
25
Coincidentally, there have been several
studies reporting an unexpected tendency for the occur-
rence of glioblastomas and anaplastic astrocytomas in
young Chinese in Taiwan, the People’s Republic of
China, and Hong Kong,
26–28
when compared with the
West.
29,30
The incidence of MSI is high in cases of spo-
radic colorectal carcinoma in the young of Hong Kong
31
and elsewhere,
32
and germline mutation of the MMR
Supported by Committee on Research and Conference Grant 337/046/
0024 and University Research Committee Grant 344/046/0003 from the
University of Hong Kong and by Croucher Foundation Research Grant
394/046/1238. TLC is a Ph.D. student of the University of Hong Kong.
Accepted for publication July 18, 1998.
Address reprint requests to Dr. Siu Tsan Yuen, Department of Pathol-
ogy, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong
Kong. E-mail: styuen@hkucc.hku.hk.
American Journal of Pathology, Vol. 153, No. 4, October 1998
Copyright © American Society for Investigative Pathology
1181
genes has been found in a high proportion of these
young patients with MSI,
31,32
but little is known of the MSI
status or mutation of the MMR genes in the young pa-
tients with gliomas. Using stringent criteria for MSI,
33,34
we examined a series of local young patients with high-
grade gliomas (grades III to IV by the World Health Or-
ganization system
35
), to look for the presence of MSI and
examined for mutation, both somatic and germline, in the
hMSH2 and hMLH1 genes.
Materials and Methods
Materials
Twenty-two patients, ages 45 years or less, with gliomas
of grades III to IV, were included in this study. The mean
age of the patients was 33 years (range, 13 to 44). The
tumors included 17 glioblastomas, 3 anaplastic astrocy-
tomas, 2 mixed gliomas (grade III), and the histological
classification used was based on the World Health Orga-
nization system.
35
Either frozen or paraffin-embedded
tumor tissue with more than 80% tumor cell content was
used. For normal tissue, either blood leukocytes obtained
by venipuncture with the patients’ consent or normal
brain tissue adjacent to the tumor was used. For all tissue
used for DNA extraction, frozen or paraffin sections were
used to confirm the absence of tumor cell contamination
in the normal tissues and to confirm the percentage of
tumor in tumor blocks. DNA and RNA were extracted
from blood leukocytes and frozen tumor blocks for germ-
line and somatic MMR gene mutational analysis using
standard methods.
MSI Analysis
Paired tumor and normal tissues were amplified by poly-
merase chain reaction (PCR) using 5 microsatellite loci.
These included dinucleotide repeats (Tp53, D18S58, and
D2S123) and polyadenine tracts (Bat40 and Bat26/A26).
Tp53 was purchased from Research Genetics (Hunts-
ville, AL). D18S58, D2S123, and Bat40 were synthesized
according to the sequence published previously.
13
For
the polyadenine tract in intron 5 of the hMSH2 gene, two
pairs of primers were used, including Bat26 as previously
published,
13
and another pair named A26, correspond-
ing to nucleotides 123 to 143 (forward) and nucleotides
222 to 241 (reverse) of the hMSH2 exon 5 genomic
sequence (GenBank accession no. U41210). In all cases,
all five loci were analyzed.
The PCR was performed in a 10-
m
l reaction solution
containing 50 ng of DNA, 10 mmol/L of Tris (ph 8.3), 50
mmol/L of KCl, 2 to 3 mmol/L of Mg
21
, 200
m
mol/L de-
oxynucleotide triphosphate, 1
m
Ci [
a
-
32
P]dCTP, 0.2 to 1
m
mol/L of each primer, and 0.1 U Taq polymerase. A
hot-start reaction was performed by preheating the mix-
ture in the thermocycler at 95°C for 5 minutes, then cool-
ing to 80°C before adding the Taq polymerase. An initial
denaturation step of 95°C for 5 minutes and 25 to 40
cycles, including 95°C for 45 seconds (1 minute), 1
minute (1.5 minutes) in 52 to 64°C annealing temperature
according to the specific primers, and 72°C for 1 minute
(2 minutes) in frozen DNA (paraffin DNA), was performed,
followed by a final extension of 5 minutes at 72°C.
The PCR products were diluted by loading buffer,
heated at 95°C for 5 minutes, and loaded onto 6% vertical
polyacrylamide gel. After electrophoresis, the gels were
fixed, dried, and exposed to X-ray film for 12 hours to 7
days.
The results were interpreted independently by two ob-
servers. Results with discrepancy in interpretation were
discussed and PCR was repeated if necessary. MSI was
defined as the presence of allelic shift or additional
bands in the tumor compared with normal tissue. All
cases with MSI were repeated once. A case was defined
as high-level MSI if there were more than 40% unstable
loci, low-level MSI if less than 40%, and microsatellite
stable if there were no unstable loci.
33,34
MSI in the (A)
10
tract of type II transforming growth
factor
b
receptor (T
b
RII), (G)
8
tracts of Bax, and insulin-
like growth factor type II receptor (IGFIIR) genes was also
analyzed in the microsatellite-unstable cases. The primer
sequences were as described previously.
36–38
hMSH2 and hMLH1 Mutational Analysis
Mutational analysis for hMSH2 and hMLH1 was per-
formed for the high-level MSI cases using the following
methods.
In Vitro Synthesized Protein Assay
In vitro-synthesized protein assays to screen for trunca-
tion mutations in the MMR genes hMSH2 and hMLH1
were performed with primer sequences as described.
5,13
In brief, 3
m
g of total RNA was reverse transcribed using
20 to 200 ng of random hexamers or oligo(dT), 20 units of
RNAsin, 20 pmol of deoxynucleotide triphosphates, and
200 units of Superscript II reverse transcriptase (Life
Technologies, Inc., Grand Island, NY) in a 20-
m
l reaction
volume, using the manufacturers’ suggested reaction
conditions. Forty cycles of PCR were performed in 50
m
l
and included 2 to 4
m
l of first-strand cDNA mix, 10
mmol/L of Tris-HCl (pH 8.3), 50 mmol/L of KCl, 3 to 5
mmol/L of MgCl
2
, 5 pmol of each primer, 200
m
mol/L of
each nucleotide, and 2.5 units Taq polymerase (Life
Technologies). Both hMSH2 and hMLH1 were amplified
in two overlapping segments ranging between 1.2 and
2.0 kb. The left-hand primers of each segment were
tagged with a T7 promoter sequence and a translation
initiation site. The products were then subjected to in vitro
transcription/translation using the linked T7 transcription-
translation system (Amersham Corp., Little Chalfont, UK).
DNA Sequencing Analysis
Individual exons of hMSH2 and hMLH1 genes, including
intron-exon boundaries, were PCR amplified. The prim-
ers’ sequences are available on request. The PCR prod-
ucts were then purified by High Pure PCR Product Puri-
fication Kit (Boehringer Mannheim, Mannheim, Germany)
1182 Leung et al
AJP October 1998, Vol. 153, No. 4
and directly sequenced by Sequenase V2.0 (Amersham)
using both forward and reverse primers following the
manufacturer’s protocols. The sequencing products were
denatured at 80°C for 5 minutes and electrophoresed
through a 6% polyacrylamide/urea gel at 70 W for 2 to 3
hours. The gels were then fixed, dried, and exposed to
autoradiographic films.
Immunohistochemistry
Immunostaining for hMSH2 and hMLH1 was performed in
the cases showing allelic shift in one or more loci, using
the standard streptavidin-biotin-peroxidase complex
method with 3,39-diaminobenzidine as chromogen. Sec-
tions 6
m
m thick of 10% neutral buffered formalin-fixed,
paraffin-embedded tumor tissue were incubated for 1
hour at 37°C with monoclonal antibodies against the ami-
no-terminal fragment (clone GB12; dilution 1:20; Onco-
gene Research Products, Cambridge, MA) and carboxy-
terminal fragment of hMSH2 (clone FE11; dilution 1:200;
Oncogene Research Products, Cambridge, MA). For
hMLH1, sections were incubated at 37°C for 1 hour using
a monoclonal antibody (clone G168-15; dilution 1:10;
PharMingen, San Diego, CA). Microwave pretreatment at
95°C for 30 minutes in citrate buffer, pH 6.0, was per-
formed after deparaffinization. For negative control, the
primary antibodies were replaced by mouse immuno-
globulin G (Dakopatts, Glostrup, Denmark).
Results
MSI
Four of the 22 high-grade gliomas from patients ages 45
years or less (18%) showed high-level MSI (Table 1).
These included three glioblastomas and one malignant
mixed glioma. In all four cases, there was gross MSI with
75 to 100% loci involved. One additional tumor showed
MSI in one locus only, and this case was thus considered
low-level MSI. None of the tumors showed mutation in the
mononucleotide tracts in the T
b
RII and Bax genes. One
tumor showed frameshift mutation in the (G)
8
tract of the
IGFIIR gene. Representative results of the microsatellite
analysis are shown in Figure 1 and 2.
Clinicopathological Data and Family History
The clinical data of the four patients with high-level MSI
gliomas are shown in Table 2. The patients were all
relatively young; two of them were below 30 when they
developed the glioblastoma. Histologically, the three gli-
oblastomas showed primitive anaplastic cells in the
background and the presence of many multinucleated
Table 1. Results of Microsatellite Analysis and Immunohistochemistry for Cases Showing MSI
Patients
ABCDE
Sex/age F/27 M/23 M/35 M/37 F/38
Diagnosis GBM Mixed glioma (grade III)* GBM GBM Astro III
Tp53 11 111
D18S58 21 112
D2S123 11 112
Bat40 11 112
Bat26/A26
11 112
% loci with MSI 75% 100% 100% 100% 20%
T
b
RII (A)
10
22 222
Bax (G)
8
22 222
IGFIIR (G)
8
21 222
hMSH2 protein (IHC) 12 221
hMLH1 protein (IHC) 21 111
GBM, glioblastoma multiforme; Astro III, anaplastic astrocytoma; IHC, immunohistochemistry.
Both Bat26 and A26 amplify a polyadenine tract in intron 5 of the hMSH2 gene.
*According to World Health Organization system.
Figure 1. MSI at various loci in gliomas: A, Tp53; B, D2S123; C, D18S58; D,
Bat26; E, A26; and F, Bat40. N, normal; T, tumor.
MSI and DNA Mismatch Repair Gene Mutation in Gliomas 1183
AJP October 1998, Vol. 153, No. 4
tumor giant cells. Patient B had a malignant mixed gli-
oma, with a prominent oligodendroglial element, although
ependymal and astrocytic elements were noted in some
areas. None except patient D had a family history of
cancer. Patient D had a positive family history of colorec-
tal carcinoma, but that did not satisfy the Amsterdam
criteria for HNPCC syndrome. Interestingly, three of the
patients had metachronous colorectal adenocarcinomas,
thus satisfying the criteria of Turcot’s syndrome.
39
Patient
A was only 27 years old when she developed a glioblas-
toma, and there was no previous tumor nor any family
history of cancer.
Germline and Somatic Mutation of the
MMR Genes
All four patients with high-level MSI gliomas showed
germline mutation of the MMR genes, three of them in the
hMSH2 and one in hMLH1 (Table 2). Three of the muta-
tions resulted in truncated protein products. One case
(patient D) showed a missense mutation resulting in
amino acid substitution in an evolutionary conserved res-
idue. The wild-type allele was lost in the tumor in this
patient.
In three cases, a second hit could be identified in the
gliomas. Patient A showed two truncated protein prod-
ucts in the in vitro-synthesized protein assay of the tumor
RNA (Figure 3). The germline mutation was found in exon
8 of the hMLH1 gene, which resulted in skipping of the
exon (Figure 4A and 5). A second mutation, found only in
the tumor DNA, resulted in a stop codon (Figure 4B). For
patients C and D, the normal allele was absent when
tumor tissue was sequenced (Figure 6). For patient B, the
wild-type allele was retained in sequencing of exon 11 in
the brain tumor. We did not screen for other somatic
mutation in the hMSH2, because only paraffin blocks of
the tumor were available.
Expression of the hMSH2 and hMLH1 Protein
Immunohistochemical staining revealed complete loss of
hMSH2 protein when both antibodies on the tumor cells in
patients B, C, and D were used, whereas the normal
neurons and glial cells at the tumor borders were posi-
Figure 2. Frameshift mutation in the mononucleotide repeats of IGFIIR gene
(C) but not in the T
b
RII (A) and Bax (B) genes in microsatellite-unstable
gliomas. Lanes N, normal; lanes T, tumor.
Table 2. Clinical Data and DNA MMR Gene Mutations in Four Patients with Microsatellite-Unstable Gliomas
Patient
Sex/
age Histology Family history
Other cancers
(age, years)
Survival after
craniotomy
MMR germline
mutation
MMR somatic mutation
in glioma
A F/27 GBM None None Died of disease at 8
months
hMLH1 last nucleotide
of exon 8
(CGgt3CAgt),
resulting in splicing
defect, skipping of
exon 8, deletion of
codon 197–226, and
frameshift
hMLH1 exon 13, codon
487 CGA3TGA(stop)
B M/23 Malignant
mixed
glioma
None Colon (25) Died of disease
(brain) at 4 years
hMSH2 exon 11 codon
580
GAA3TAA(stop)
Not determined;
(absence of hMSH2
protein by
immunostaining)
C M/35 GBM Not available
(adopted son)
Colon (29) Alive with disease
(brain) at 6
months
hMSH2 first nucleotide
of exon 12 codon
587, deletion of G
(agGCT3agCT),
generating a stop
codon 6 bp
downstream
Wild-type allele loss in
tumor by sequencing
D M/37 GBM Two sisters
had colorectal
adenocar-
cinomas
Rectum (28) Died of disease
(brain) at 0.5
months
hMSH2 exon 3 codon
199 (TGT3CGT)
resulting in change
of an evolutionary
conserved cysteine
to arginine
Wild-type allele loss in
tumor by sequencing
GBM, glioblastoma multiforme.
1184 Leung et al
AJP October 1998, Vol. 153, No. 4
tive. Staining for hMLH1 was retained in the tumors in
these three patients. The tumor cells in patient A were
negative for hMLH1 but positive for hMSH2 proteins,
whereas the normal cells were positive for both.
Discussion
Three important pieces of information resulted from this
study. 1) A proportion of young patients (18%) with high-
grade gliomas showed gross replication error, character-
istic of defects in the DNA MMR system. 2) In all of these
patients with gross replication errors, germline mutation
in one of the MMR genes could be detected. 3) In three
of the four cases, inactivation of the second allele was
found in the tumor tissue. Although the second hit could
not be detected in patient B, the tumor cells were immu-
nohistochemically negative for hMSH2, supporting the
presence of a second inactivating event. This double-hit
phenomenon for MMR genes, identical to Knudson’s the-
ory for a tumor suppressor gene, was also previously
suggested in HNPCC-related and sporadic colorectal
carcinomas.
4,6,7,40
To our knowledge, this is the first study documenting
the presence of a germline MMR gene mutation in young
patients with sporadic gliomas. A similar study in young
patients with sporadic colorectal carcinoma revealed MSI
in 58%, with germline MMR mutation detected in 42% of
the MSI-positive cases.
32
Although germline MMR gene
mutation has previously been found in four patients with
Turcot’s syndrome,
21,23,24
patient A developed and died
Figure 4. A: Sequencing result of hMLH1 exon 8 from the blood leukocytes
(lanes N) and glioblastoma (lanes T) of patient A. Both tumor and blood
leukocytes demonstrate a mutation in the last nucleotide of exon 8, CGgt to
CAgt (arrow), indicating that it is a germline mutation. B: Sequencing result
of hMLH1 exon 13 from the blood leukocytes (lanes N) and glioblastoma
(lanes T) of patient A. Somatic mutation in codon 487, CGA to TGA (arrow),
generating a stop codon, is found in the tumor but not in blood leukocytes.
Figure 5. Reverse transcription-PCR analysis using a pair of primers ampli-
fying nucleotides 471 to 813 of hMLH1 cDNA. A wild-type band of 342 bp
and an abnormal band of 253 bp (arrow), resulting from skipping of exon 8,
are noted in the RNA extracted from the leukocytes of patient A (lane A)
compared with a normal individual (lane C). Lane M: Bluescript-MSP1
marker.
Figure 3. In vitro synthesized protein assay of 59(A) and 39(B) segments of
the hMLH1 gene from the glioblastoma of patient A. The bands with highest
molecular weight and strongest intensity correspond to the normal products.
Truncated products of 21 and 18 kd (arrowheads) are noted in the 59and 39
segments. Lanes C, blood leukocytes from a normal individual as control;
lanes T, glioblastoma.
MSI and DNA Mismatch Repair Gene Mutation in Gliomas 1185
AJP October 1998, Vol. 153, No. 4
of the glioma without antecedent cancer. Patient B pre-
sented with the glioma first and only subsequently devel-
oped the colorectal carcinoma. Neither A nor B had a
family history of cancer. Two other patients have ante-
cedent colorectal carcinoma. In patient D, a family history
of colorectal carcinomas was also obtained. This raises
an important point concerning the management of young
patients with microsatellite-unstable gliomas and their
family members. From the information in our study, we
conclude that screening for replication error is useful in
young patients with high-grade gliomas. For high-level
MSI patients, germline mutation of the MMR gene should
be sought. Regular colonoscopic screening for colorectal
carcinoma should be offered to the patient and the family
members with demonstrable MMR gene mutation. Also,
we should be alert to and check for the possibility of brain
tumor by regular neurological examination. It is of note
that, whereas the colorectal carcinoma could be suc-
cessfully treated, three of the patients succumbed to the
gliomas 0.5 months to 4 years after the craniotomy. This
was in contrast to the prolonged survival noted in three
patients with MSI-positive glioblastoma in a previous
series.
23
Concerning the histological type of high-level MSI gli-
omas, three were glioblastomas. Interestingly, one case
was a malignant mixed glioma with a prominent oligoden-
droglial component and also focal ependymal differenti-
ation. In HNPCC kindred, the possible histological type of
brain tumor includes not only astrocytomas but also oli-
godendrogliomas and rarely ependymomas.
21
Thus, mu-
tation of the MMR gene may lead not only to glioblas-
tomas, but to high-grade gliomas of oligodendroglial or
even ependymal differentiation.
Mononucleotide tracts of various growth-regulatory
genes are frequently the target of mutational inactivation
in microsatellite-unstable tumors. The (A)
10
tract in the
T
b
RII gene is mutated in 70 to 90% of microsatellite-
unstable colorectal and gastric cancers.
36,41
Frameshift
mutation of the (G)
8
tract in Bax is also reported in more
than 50% of these cancers.
37,42,43
Apart from frameshift
mutation in the (G)
8
tract, somatic mutation of Bax genes
is frequent in MSI-positive gastric and colorectal carci-
nomas,
43
but not in gliomas.
44
Interestingly, none of the
MSI-positive gliomas in this study showed mutation in the
T
b
RII and Bax genes. This may be the result of selection
pressure, in which mutation of genes caused by MMR
defects are selected for if they confer growth advantage
in that organ.
We identified a frameshift mutation in the IGFIIR gene
in the malignant mixed glioma from patient B, the first
reported mutation in this gene in a glioma, although IG-
FIIR mutation has been reported in MSI colorectal, gas-
tric, and endometrial carcinomas.
38,45
IGFIIR plays a role
in activation of transforming growth factor
b
,
46
which is a
potent growth inhibitor. Also, it antagonizes the growth-
stimulatory effect of IGFII by internalizing and degrading
the protein.
47
Given that enhanced expression of IGFII
mRNA has been reported in gliomas,
48
inactivating mu-
tation of IGFIIR may remove the growth-inhibitory signal
and confer growth advantage.
The molecular genetic pathways of different subsets of
glioblastoma have been increasingly clarified in recent
years.
29,49
Those arising de novo are referred to as pri-
mary glioblastomas, and those developed from a pre-
existing astrocytoma are referred to as secondary glio-
blastomas. Most primary glioblastomas develop in older
patients (mean, 55 years) with epidermal growth factor
receptor amplification or overexpression, loss of het-
erozygosity in chromosome 10, and p16 deletion. Sec-
ondary glioblastomas tend to occur in younger patients
(mean, 39 years), and most of them harbor p53 muta-
tions.
50–54
We have demonstrated that a proportion of
primary glioblastomas in young patients can be caused
by germline MMR gene mutations, and these patients
and their family members are at risk of developing other
HNPCC-related tumors, in particular colorectal carcino-
mas. Screening for MSI and MMR gene mutation is thus
of importance in the management of these patients.
Acknowledgments
We thank Mr. Samson W. C. Shum for the technical
assistance, Miss Kedo Kwan for collecting the family
history, and Dr. R. J. Collins for his assistance with the
manuscript.
References
1. Aaltonen LA, Peltomaki P, Leach FS, Sistonen P, Pylkkanen L, Mecklin
JP, Jarvinen H, Powell SM, Jen J, Hamilton SR, Petersen GM, Kinzler
KW, Vogelstein B, de la Chapell A: Clues to the pathogenesis of
familial colorectal cancer. Science 1993, 260:812– 816
2. Lynch HT, Smyrk TC, Watson P, Lanspa SJ, Lynch JF, Lynch PM,
Cavalieri RJ, Boland CR: Genetics, natural history, tumor spectrum,
and pathology of hereditary nonpolyposis colorectal cancer: an up-
dated review. Gastroenterology 1993, 104:1535–1549
3. Fishel R, Lescoe MK, Rao MR, Copeland NG, Jenkins NA, Garber J,
Kane M, Kolodner R: The human mutator gene homolog MSH2 and its
Figure 6. Sequencing result of hMSH2 exon 12 from the blood leukocytes
(lanes L) and glioblastoma (lanes G) of patient C. Deletion of a guanosine
in the first nucleotide of exon 12 (agGCT to agCT) is noted in both blood
leukocytes and tumor tissue. The normal allele is lost in the tumor tissue.
1186 Leung et al
AJP October 1998, Vol. 153, No. 4
association with hereditary nonpolyposis colon cancer. Cell 1993,
75:1027–1038
4. Leach FS, Nicolaides NC, Papadopoulos N, Liu B, Jen J, Parsons R,
Peltomaki P, Sistonen P, Aaltonen LA, Nystrom Lahti M, Guan XY,
Zhang J, Meltzer PS, Yu JW, Kao FT, Chen DJ, Cerosaletti KM,
Fournier RE, Todd S, Lewis T, Leach RJ, Naylor SL, Weissenbach J,
Mecklin JP, Jarvinen H, Petersen GM, Hamilton SR, Green J, Jass J,
Watson P, Lynch HT, Trent JM, de la Chapelle A, Kinzler KW, Vo-
gelstein B: Mutations of a mutS homolog in hereditary nonpolyposis
colorectal cancer. Cell 1993, 75:1215–1225
5. Liu B, Parsons RE, Hamilton SR, Petersen GM, Lynch HT, Watson P,
Markowitz S, Willson JK, Green J, de la Chapelle A, Kinzler KW,
Vogelstein B: hMSH2 mutations in hereditary nonpolyposis colorectal
cancer kindreds. Cancer Res 1994;54:4590 4594
6. Papadopoulos N, Nicolaides NC, Wei YF, Ruben SM, Carter KC,
Rosen C, Haseltine WA, Fleischmann RD, Fraser CM, Adams MD,
Venter JC, Hamilton SR, Petersen GM, Watson P, Lynch HT, Pelto-
maki P, Mecklin JP, de la Chapelle A, Kinzler KW, Vogelstein B:
Mutation of a mutL homolog in hereditary colon cancer. Science
1994, 263:1625–1629
7. Nicolaides NC, Papadopoulos N, Liu B, Wei YF, Carter KC, Ruben
SM, Rosen CA, Haseltine WA, Fleischmann RD, Fraser CM, Adams
MD, Venter JC, Dunlop MG, Hamilton SR, Petersen GM, de la
Chapelle A, Vogelstein B, Kinzler KW: Mutations of two PMS homo-
logues in hereditary nonpolyposis colon cancer. Nature 1994, 371:
75– 80
8. Bronner CE, Baker SM, Morrison PT, Warren G, Smith LG, Lescoe MK,
Kane M, Earabino C, Lipford J, Lindblom A, Tannergard P, Bollag RJ,
Godwin AR, Ward DC, Nordenskjold M, Fishel R, Kolodner R, Liskay
RM: Mutation in the DNA mismatch repair gene homologue hMLH1 is
associated with hereditary non-polyposis colon cancer. Nature 1994,
368:258 –261
9. Palombo F, Hughes M, Jiricny J, Truong O, Hsuan J: Mismatch repair
and cancer. Nature 1994, 367:417
10. Palombo F, Gallinari P, Iaccarino I, Lettieri T, Hughes M, D’Arrigo A,
Truong O, Hsuan JJ, Jiricny J: GTBP, a 160-kilodalton protein essen-
tial for mismatch-binding activity in human cells. Science 1995, 268:
1912–1914
11. Drummond JT, Li GM, Longley MJ, Modrich P: Isolation of an hMSH2–
p160 heterodimer that restores DNA mismatch repair to tumor cells.
Science 1995, 268:1909 –1912
12. Papadopoulos N, Nicolaides NC, Liu B, Parsons R, Lengauer C,
Palombo F, D’Arrigo A, Markowitz S, Willson JK, Kinzler KW, Jiricny J,
Vogelstein B: Mutations of GTBP in genetically unstable cells. Sci-
ence 1995, 268:1915–1917
13. Liu B, Parsons R, Papadopoulos N, Nicolaides NC, Lynch HT, Watson
P, Jass JR, Dunlop M, Wyllie A, Peltomaki P, de la Chapelle A,
Hamilton SR, Vogelstein B, Kinzler KW: Analysis of mismatch repair
genes in hereditary non-polyposis colorectal cancer patients. Nat
Med 1996, 2:169 –174
14. Eshleman JR, Markowitz SD: Microsatellite instability in inherited and
sporadic neoplasms. Curr Opin Oncol 1995, 7:83– 89
15. Izumoto S, Arita N, Ohnishi T, Hiraga S, Taki T, Tomita N, Ohue M,
Hayakawa T: Microsatellite instability and mutated type II transform-
ing growth factor-
b
receptor gene in gliomas. Cancer Lett 1997,
112:251–256
16. Dams E, Van de Kelft EJ, Martin JJ, Verlooy J, Willems PJ: Instability
of microsatellites in human gliomas. Cancer Res 1995, 55:1547–1549
17. Zhu J, Guo SZ, Beggs AH, Maruyama T, Santarius T, Dashner K,
Olsen N, Wu JK, Black P: Microsatellite instability analysis of primary
human brain tumors. Oncogene 1996, 12:1417–1423
18. Wooster R, Cleton Jansen AM, Collins N, Mangion J, Cornelis RS,
Cooper CS, Gusterson BA, Ponder BA, von Deimling A, Wiestler OD,
Cornelisse CJ, Devilee P, Stratton MR: Instability of short tandem
repeats (microsatellites) in human cancers. Nat Genet 1994, 6:152–
156
19. Amariglio N, Friedman E, Mor O, Stiebel H, Phelan C, Collins P,
Nordenskjold M, Brok Simoni F, Rechavi G: Analysis of microsatellite
repeats in pediatric brain tumors. Cancer Genet Cytogenet 1995,
84:56 –59
20. Ponz de Leon M, Benatti P, Pedroni M, Sassatelli R, Roncucci L: Risk
of cancer revealed by follow-up of families with hereditary non-pol-
yposis colorectal cancer: a population-based study. Int J Cancer
1993, 55:202–207
21. Vasen HF, Sanders EA, Taal BG, Nagengast FM, Griffioen G, Menko
FH, Kleibeuker JH, Houwing Duistermaat JJ, Meera Khan P: The risk
of brain tumours in hereditary non-polyposis colorectal cancer
(HNPCC). Int J Cancer 1996, 65:422– 425
22. Heinimann K, Muller H, Weber W, Scott RJ: Disease expression in
Swiss hereditary non-polyposis colorectal cancer (HNPCC) kindreds.
Int J Cancer 1997, 74:281–285
23. Hamilton SR, Liu B, Parsons RE, Papadopoulos N, Jen J, Powell SM,
Krush AJ, Berk T, Cohen Z, Tetu B, Burger PC, Wood PA, Taqi F,
Booker SV, Petersden GM, Offerhaus GJA, Tersmette AC, Giardiello
FM, Vogelstein B, Kinzler KW: The molecular basis of Turcot’s syn-
drome. N Engl J Med 1995, 332:839 847
24. Miyaki M, Nishio J, Konishi M, Kikuchi-Yanoshita R, Tanaka K,
Muraoka M, Nagato M, Chong JM, Koika M, Terada T, Yutaka K,
Fukutome A, Tomiyama J, Chuganji Y, Momoi M, Utsunomiya J:
Drastic genetic instability of tumours and normal tissues in Turcot
syndrome. Oncogene 1997, 15:2877–2881
25. Yuen ST, Chung LP, Leung SY, Luk ISC, Chan SY, Ho JWC, Wyllie AH:
Colorectal carcinoma in Hong Kong: epidemiology and genetic mu-
tations. Br J Cancer 1997, 76:1610 –1616
26. Huang WQ, Zheng SJ, Tian QS, Huang JQ, Li Yx, Xu QZ, Liu ZJ,
Zhang WC: Statistical analysis of central nervous system tumors in
China. J Neurosurg 1982, 56:555–564
27. Kepes JJ, Chen WY, Pang LC, Kepes M: Tumors of the central
nervous system in Taiwan, Republic of China. Surg Neurol 1984,
22:149 –156
28. Ng HK, Poon WS, South JR, Lee JC: Tumours of the central nervous
system in Chinese in Hong Kong: a histological review. Aust N Z
J Surg 1988, 58:573–578
29. Kleihues P, Burger PC, Plate KH, Ohgaki H, Cavenee WK:
Glioblastoma: pathology and genetics of tumours of the nervous
system. Edited by P Kleihues, WK Cavenee. Lyon: International
Agency for Research on Cancer, 1997, pp 16 –28
30. Zulch KJ: Brain Tumors: Their Biology and Pathology, ed 3. Berlin,
Springer-Verlag, 1986
31. Chan TL, Yuen ST, Chung LP, Ho J, Leung SY, Chan AS, Chan LC:
Replication errors (RER) and mismatch repair (MMR) gene mutations
in young colorectal carcinoma patients in Hong Kong Chinese. Ge-
netics of human cancer: pathogenesis and diagnosis, Proceedings of
Keystone Symposium on Molecular and Cellular Biology, 1997, p 9
32. Liu B, Farrington SM, Petersen GM, Hamilton SR, Parsons R, Pap-
adopoulos N, Fujiwara T, Jen J, Kinzler KW, Wyllie AH, Vogelstein B,
Dunlop MG: Genetic instability occurs in the majority of young pa-
tients with colorectal cancer. Nat Med 1995, 1:348 –352
33. Dietmaier W, Wallinger S, Bocker T, Kullmann F, Fishel R, Ruschoff J:
Diagnostic microsatellite instability: definition and correlation with
mismatch repair protein expression. Cancer Res 1997, 57:4749
4756
34. Bocker T, Diermann J, Friedl W, Gebert J, Holinski Feder E, Karner H,
von Knebel Doeberitz M, Koelble K, Moeslein G, Schackert HK, Wirtz
HC, Fishel R, Ruschoff J: Microsatellite instability analysis: a multi-
center study for reliability and quality control. Cancer Res 1997,
57:4739 4743
35. Kleihues P, Burger PC, Scheithauer BW: Histological Typing of Tu-
mours of the Central Nervous System. Berlin, Springer-Verlag, 1993
36. Myeroff LL, Parsons R, Kim SJ, Hedrick L, Cho KR, Orth K, Mathis M,
Kinzler KW, Lutterbaugh J, Park K, Bang YJ, Lee HY, Park JG, Lynch
HT, Roberts AB, Vogelstein B, Markowitz SD: A transforming growth
factor
b
receptor type II gene mutation common in colon and gastric
but rare in endometrial cancers with microsatellite instability. Cancer
Res 1995, 55:5545–5547
37. Rampino N, Yamamoto H, Ionov Y, Li Y, Sawai H, Reed JC, Perucho
M: Somatic frameshift mutations in the BAX gene in colon cancers of
the microsatellite mutator phenotype. Science 1997, 275:967–969
38. Souza RF, Appel R, Yin J, Wang S, Smolinski KN, Abraham JM, Zou
TT, Shi YQ, Lei J, Cottrell J, Cymes K, Biden K, Simms L, Leggett B,
Lynch PM, Frazier M, Powell SM, Harpaz N, Sugimura H, Young J,
Meltzer SJ: Microsatellite instability in the insulin-like growth factor II
receptor gene in gastrointestinal tumours. Nat Genet 1996, 14:255–
257
39. Turcot J, Despres JP, St. Pierre F: Malignant tumors of the central
nervous system associated with familial polyposis of the colon: report
of two cases. Dis Colon Rectum 1959, 2:465– 468
40. Liu B, Nicolaides NC, Markowitz S, Willson JK, Parsons RE, Jen J,
MSI and DNA Mismatch Repair Gene Mutation in Gliomas 1187
AJP October 1998, Vol. 153, No. 4
Papadopoulos N, Peltomaki P, de la Chapelle A, Hamilton SR, Kinzler
KW, Vogelstein B: Mismatch repair gene defects in sporadic colorec-
tal cancers with microsatellite instability. Nat Genet 1995, 9:48 –55
41. Parsons R, Myeroff LL, Liu B, Willson JK, Markowitz SD, Kinzler KW,
Vogelstein B: Microsatellite instability and mutations of the transform-
ing growth factor
b
type II receptor gene in colorectal cancer. Cancer
Res 1995, 55:5548 –5550
42. Chung YJ, Park SW, Song JM, Lee KY, Seo EJ, Choi SW, Rhyu MG:
Evidence of genetic progression in human gastric carcinomas with
microsatellite instability. Oncogene 1997, 15:1719 –1726
43. Yamamoto H, Sawai H, Perucho M: Frameshift somatic mutations in
gastrointestinal cancer of the microsatellite mutator phenotype. Can-
cer Res 1997, 57:4420 4426
44. Chou D, Miyashita T, Mohrenweiser HW, Ueki K, Kastury K, Druck T,
von Deimling A, Huebner K, Reed JC, Louis DN: The BAX gene maps
to the glioma candidate region at 19q13.3, but is not altered in human
gliomas. Cancer Genet Cytogenet 1996, 88:136 –140
45. Ouyang H, Shiwaku HO, Hagiwara H, Miura K, Abe T, Kato Y, Ohtani
H, Shiiba K, Souza RF, Meltzer SJ, Horii A: The insulin-like growth
factor II receptor gene is mutated in genetically unstable cancers of
the endometrium, stomach, and colorectum. Cancer Res 1997, 57:
1851–1854
46. Dennis PA, Rifkin DB: Cellular activation of latent transforming growth
factor
b
requires binding to the cation-independent mannose 6-phos-
phate/insulin-like growth factor type II receptor. Proc Natl Acad Sci
USA 1991, 88:580 –584
47. Kornfeld S: Structure and function of the mannose 6-phosphate/
insulin-like growth factor II receptors. Annu Rev Biochem 1992, 61:
307–330
48. Sandberg AC, Engberg C, Lake M, von Holst H, Sara VR: The ex-
pression of insulin-like growth factor I and insulin-like growth factor II
genes in the human fetal and adult brain and in glioma. Neurosci Lett
1988, 93:114–119
49. Kleihues P, Ohgaki H: Genetics of glioma progression and the defi-
nition of primary and secondary glioblastoma. Brain Pathol 1997,
7:1131–1136
50. Lang FF, Miller DC, Koslow M, Newcomb EW: Pathways leading to
glioblastoma multiforme: a molecular analysis of genetic alterations in
65 astrocytic tumors. J Neurosurg 1994, 81:427– 436
51. von Deimling A, von Ammon K, Schoenfeld D, Wiestler OD, Seizinger
B, Louis DN: Subsets of glioblastoma multiforme defined by molec-
ular genetic analysis. Brain Pathol 1993, 3:19 –26
52. Watanabe K, Tachibana O, Sata K, Yonekawa Y, Kleihues P, Ohgaki
H: Overexpression of the EGF receptor and p53 mutations are mu-
tually exclusive in the evolution of primary and secondary glioblas-
tomas. Brain Pathol 1996, 6:217–223
53. Hayashi Y, Ueki K, Waha A, Wiestler OD, Louis DN, von Deimling A:
Association of EGFR gene amplification and CDKN2 (p16/MTS1)
gene deletion in glioblastoma multiforme. Brain Pathol 1997, 7:871–
875
54. Biernat W, Tohma Y, Yonekawa Y, Kleihues P, Ohgaki H: Alterations
of cell cycle regulatory genes in primary (de novo) and secondary
glioblastomas. Acta Neuropathol Berl 1997, 94:303–309
1188 Leung et al
AJP October 1998, Vol. 153, No. 4
... MSI is associated with all types of cancers, including brain cancer (Eckert et al., 2007;Latham et al., 2019), even if MSI phenotyping appears to be closely linked with specific clinicopathological features, primarily in colorectal cancer (Boland and Goel, 2010). Screening for gene mutations in MSI and MMR has been seen as important in the treatment of patients with glioma (Leung et al., 1998;Xu et al., 2021b). Thus, we analyzed the correlation between MSI and the expression of URB2 in glioma. ...
Article
Full-text available
Introduction: Glioma is the most common primary brain tumor and primary malignant tumor of the brain in clinical practice. Conventional treatment has not significantly altered the prognosis of patients with glioma. As research into immunotherapy continues, glioma immunotherapy has shown great potential. Methods: The clinical data were acquired from the Chinese Glioma Genome Atlas (CGGA) database and validated by the Gene Expression Omnibus (GEO) database, The Cancer Genome Atlas (TCGA) dataset, Clinical Proteomic Tumor Analysis Consortium (CPTAP) database, and Western blot (WB) analysis. By Cox regression analyses, we examined the association between different variables and overall survival (OS) and its potential as an independent prognostic factor. By constructing a nomogram that incorporates both clinicopathological variables and the expression of URB2, we provide a model for the prediction of prognosis. Moreover, we explored the relationship between immunity and URB2 and elucidated its underlying mechanism of action. Results: Our study shows that URB2 likely plays an oncogenic role in glioma and confirms that URB2 is a prognostic independent risk factor for glioma. Furthermore, we revealed a close relationship between immunity and URB2, which suggests a new approach for the immunotherapy of glioma. Conclusion: URB2 can be used for prognosis prediction and immunotherapy of glioma.
... MSI-H is a sufficient condition for administering pembrolizumab, and the existence of such targeted molecular aberration is mandatory for applying those molecularly targeting agents. However, no patient was diagnosed with MSI-H (among 65 patients) in this study, which is compatible with previous literature [27][28][29]. Although a daily clinical practice, the TMB score is not a definite prognostic indicator to assess the immune checkpoint inhibitors (e.g., pembrolizumab) applicable in glioma patients; the temozolomide-induced hypermutation increases the TMB scores in glioma [30,31]. ...
Article
Full-text available
Simple Summary Cancer patients suffer from recurrence after the completion of standard treatments and exhaustion of treatment options. The comprehensive genomic profiling test (CGPT) is a platform that enables those patients to access the eligible promising therapeutic agents based on their genomic aberrations, using next-generation sequencing. Though CGPTs have been utilized since 2019 in Japan, only limited findings have been available about their use for glioma patients. The aim of this study was to reveal the comprehensive results of CGPT in glioma patients, especially the clinical actionability, which means the probability of being able to receive appropriate molecular targeting therapeutic agents. In our cohort, the clinical actionability was 18.5%, which was compatible with the results of previous reports for tumors other than glioma. We confirmed that CGPT is also useful for glioma patients, and our result will encourage a future increase of CGPT use in our clinical practices. Abstract Next-generation sequencing-based comprehensive genomic profiling test (CGPT) enables clinicians and patients to access promising molecularly targeted therapeutic agents. Approximately 10% of patients who undergo CGPT receive an appropriate agent. However, its coverage of glioma patients is seldom reported. The aim of this study was to reveal the comprehensive results of CGPT in glioma patients in our institution, especially the clinical actionability. We analyzed the genomic aberrations, tumor mutation burden scores, and microsatellite instability status. The Molecular Tumor Board (MTB) individually recommended a therapeutic agent and suggested further confirmation of germline mutations after considering the results. The results of 65/104 patients with glioma who underwent CGPTs were reviewed by MTB. Among them, 12 (18.5%) could access at least one therapeutic agent, and 5 (7.7%) were suspected of germline mutations. A total of 49 patients with IDH-wildtype glioblastoma showed frequent genomic aberrations in the following genes: TERT promoter (67%), CDKN2A (57%), CDKN2B (51%), MTAP (41%), TP53 (35%), EGFR (31%), PTEN (31%), NF1 (18%), BRAF (12%), PDGFRA (12%), CDK4 (10%), and PIK3CA (10%). Since glioma patients currently have very limited standard treatment options and a high recurrence rate, CGPT might be a facilitative tool for glioma patients in terms of clinical actionability and diagnostic value.
... Along with TMB, we also predict MSI status and possible POLE deficiency. As previously reported, the incidence of MSI in diffuse gliomas was low [59][60][61]. ...
Article
Full-text available
Background In the clinical setting, workflows for analyzing individual genomics data should be both comprehensive and convenient for clinical interpretation. In an effort for comprehensiveness and practicality, we attempted to create a clinical individual whole exome sequencing (WES) analysis workflow, allowing identification of genomic alterations and presentation of neurooncologically-relevant findings. Methods The analysis workflow detects germline and somatic variants and presents: (1) germline variants, (2) somatic short variants, (3) tumor mutational burden (TMB), (4) microsatellite instability (MSI), (5) somatic copy number alterations (SCNA), (6) SCNA burden, (7) loss of heterozygosity, (8) genes with double-hit, (9) mutational signatures, and (10) pathway enrichment analyses. Using the workflow, 58 WES analyses from matched blood and tumor samples of 52 patients were analyzed: 47 primary and 11 recurrent diffuse gliomas. Results The median mean read depths were 199.88 for tumor and 110.955 for normal samples. For germline variants, a median of 22 (14–33) variants per patient was reported. There was a median of 6 (0–590) reported somatic short variants per tumor. A median of 19 (0–94) broad SCNAs and a median of 6 (0–12) gene-level SCNAs were reported per tumor. The gene with the most frequent somatic short variants was TP53 (41.38%). The most frequent chromosome-/arm-level SCNA events were chr7 amplification, chr22q loss, and chr10 loss. TMB in primary gliomas were significantly lower than in recurrent tumors ( p = 0.002). MSI incidence was low (6.9%). Conclusions We demonstrate that WES can be practically and efficiently utilized for clinical analysis of individual brain tumors. The results display that NOTATES produces clinically relevant results in a concise but exhaustive manner.
... Malfunctioning of MMR proteins, due either to mutation, or reduced expression, suggests the correlation of cancer development to the aberrations of all or the majority of MMR proteins. Besides colon tumors, many studies report on MSI in diverse malignancies including endometrial, ovarian, gastric, melanoma, prostate, lung, stomach, and glioblastoma (Leung et al., 1998;Alvino et al., 2000;Arai et al., 2016;Hause et al., 2016;Kubeček and Kopecký, 2016). ...
Article
Full-text available
The acquisition of genomic instability is one of the key characteristics of the cancer cell, and microsatellite instability (MSI) is an important segment of this phenomenon. This review aims to describe the mismatch DNA repair (MMR) system whose deficiency is responsible for MSI and discuss the cellular roles of MMR genes. Malfunctioning of the MMR repair pathway increases the mutational burden of specific cancers and is often involved in its etiology, sometimes as an influential bystander and sometimes as the main driving force. Detecting the presence of MSI has for a long time been an important part of clinical diagnostics, but has still not achieved its full potential. The MSI blueprints of specific tumors are useful for precize grading, evaluation of cancer chance and prognosis and to help us understand how and why therapy-resistant cancers arise. Furthermore, evidence indicates that MSI is an important predictive biomarker for the application of immunotherapy.
... This includes 5 colorectal cancers, 5 ovarian cancers, 1 endometrial cancer, 3 brain tumours, and 1 testicular tumour. The 3 patients with brain tumours were included in our earlier studies of early-onset glioma and Turcot's syndrome (Leung et al., 1998;Chan et al., 1999a). Fifty-five patients received a median of 1 surveillance colonoscopy (range 1-5, SD 1.36) at a median duration of 2 years (range 1-15 years, SD 3.75) after the index colorectal cancer resection. ...
Article
The Hong Kong Chinese population has an unusually high incidence of colorectal cancer in the young, suggestive of hereditary susceptibility. To search for a genetic basis for this predisposition, we studied the incidence of microsatellite instability (MSI) in paraffin‐embedded colectomy specimens of 124 young (<50 years old) Chinese colorectal cancer patients referred to the Hong Kong Hereditary Gastrointestinal Cancer Registry from 1995 to 1998. By medical record review and personal interview, we searched for distinct clinical features associated with the manifestation of MSI in this group of patients. For patients with MSI tumours, blood was taken for detection of germline mutation in 2 mismatch repair (MMR) genes. MSI was present in 33 tumours from 23 males and 10 females (26.6%). Ongoing mutation analysis has so far identified MMR gene mutations in 8 patients with MSI tumours. The incidence of MSI increased significantly with decreasing age at cancer diagnosis. For patients aged 30 to 49, MSI tumours were located mainly at the proximal colon. However, for exceptionally young patients (<30 years), MSI tumours tended to be at the distal large bowel. This observation suggested a differential activity of the MMR pathway in colorectal carcinogenesis in different age groups. On multivariate analysis, young age at cancer diagnosis, proximal tumour location, a strong family history of colorectal cancer, and a personal history of metachronous cancer were independent predictors for MSI status. This knowledge may have an impact on the management of young colorectal cancer patients and their families. Int. J. Cancer 89:356–360, 2000. © 2000 Wiley‐Liss, Inc.
Article
Full-text available
Background/aim: Microsatellite instability tests and programmed cell death-1 (PD-1) / programmed cell death ligand-1 (PD-L1) in the immune checkpoint pathway are the tests that determine who will benefit from immune checkpoint inhibitor therapy. We aimed to show the expression of DNA mismatch repair proteins and PD-1 / PD-L1 molecules that inhibit immune checkpoints, to explain the relationship between them, and to demonstrate their predictive role in recurrent and non-recurrent glioblastoma. Materials and methods: We analyzed 27 recurrent and 47 non-recurrent cases at our archive. We performed immunohistochemical analysis to determine expressions of PD-1, PD-L1 and mismatch repair proteins in glioblastoma. We evaluated the relationship between these two group and compared the results with the clinicopathological features. Results: The mean age of diagnosis was significantly lower in recurrent glioblastoma patients. Median survival was longer in this group. We found that PD-L1 expression was reduced in recurrent cases. Additionally, recurrent cases had a significantly higher rate of microsatellite instability. Loss of PMS2 was high in both group but was substantially higher in recurrent cases. Conclusion: The presence of microsatellite instability and low PD-L1 levels, which are among the causes of treatment resistance in glioblastoma, were found to be compatible with the literature in our study, with higher rates in recurrent cases. In recurrent cases with higher mutations and where immunotherapy resistance is expected less, low PD-L1 levels thought that different combinations with other immune checkpoint inhibitors can be tried as predictive and prognostic marker in GBM patients.
Article
Central nervous system tumors in adolescents and young adults (AYA) are rarely reported in the literature. The association with cancer predisposition syndrome is not established. Programmed death ligand 1 (PD-L1) can predict the potential response of patients to immunotherapy. A link between mismatch repair protein deficiency (MMRP-D) and response to immunotherapy is established. P53 is reported to be positive in MMRD-D cases. We aim to investigate the frequency of MMRP-D in AYA with high-grade glioma and any potential association with PD-L1. A total of 96 cases were tested including 49 (51.0%) cases of glioblastoma. Six cases (6.25%) were MMRP-D, 17 (17.7%) were PD-L1 positive, mostly in grade IV tumors (8.7% in grade III compared to 26% in grade IV, p value = 0.027), and 69 (71.9%) were P3 positive. None of the MMRP-D cases expressed PD-L1. P53-positive cases were mostly MMRP proficient (n = 67; 74.4%, p value 0.051). Fourteen cases (28.7%) were positive for both PD-L1 and P53, while p53-positive grade IV tumors were mostly associated with negative PD-L1 (n = 29, 58%, p value = 0.043). MMRP deficiency does not appear to be prevalent in high-grade glioma in AYA. Expression of PD-L1 in a quarter of cases might suggest a role for immunotherapy in high-grade glioma.
Article
Glioblastoma (WHO grade IV glioma) is the most common malignant primary brain tumor in adults. Survival has remained largely static for decades, despite significant efforts to develop new effective therapies. Immunotherapy, and especially immune checkpoint inhibitors and programmed cell death (PD)-1/PD-L1 inhibitors have transformed the landscape of cancer treatment and improved patient survival in a number of different cancer types. With the exception of few select cases (e.g., patients with Lynch syndrome) the neuro-oncology community is still awaiting evidence that PD-1 blockade can lead to meaningful clinical benefit in glioblastoma. This lack of progress in the field is likely to be due to multiple reasons, including inherent challenges in brain tumor drug development, the blood-brain barrier, the unique immune environment in the brain, the impact of corticosteroids, as well as inter- and intra-tumoral heterogeneity. Here we critically review the clinical literature, address the unique aspects of glioma immunobiology and potential immunobiological barriers to progress, and contextualize new approaches to increase the efficacy of PD-1/PD-L1 inhibitors in glioblastoma that may identify gaps and testable relevant hypotheses for future basic and clinical research and to provide a novel perspective to further stimulate pre-clinical and clinical research to ultimately help patients with glioma, including glioblastoma, which is arguably one of the greatest areas of unmet need in cancer. Moving forward, we need to build on our existing knowledge by conducting further fundamental glioma immunobiology research in parallel with innovative and methodologically sound clinical trials.
Article
Full-text available
Purpose: DNA mismatch repair (MMR) genes play important roles in maintaining genome stability. Mutations in MMR genes disrupt their mismatch repair function, cause genome instability and lead to increased risk of cancer in the mutation carriers as represented by Lynch Syndrome. Studies have identified a large number of MMR variants, mostly in the Caucasian population, whereas data from non-Caucasian populations remain poorly illustrated. With the population size of 1.4 billion, knowledge of MMR variants in the Chinese population can be valuable in understanding the roles of ethnic MMR variation and cancer and to further guide clinical applications in MMR-related cancer prevention and treatment in the Chinese population. In this study, we systematically analysed the MMR variants from the Chinese population. Experimental design: We performed a comprehensive MMR data mining and collected all the MMR variation data reported from 33,998 Chinese individuals consisting of 23,938 cancer and 10,060 non-cancer cases between January 1997 to May 2019. For the collected data, we performed standardisation following Human Genome Variation Society nomenclature and reannotated the MMR variant data following American College of Medical Genetics and Genomics guidelines and comparing with non-Chinese MMR data on various aspects. Results: We identified a total of 540 MMR variants in the Chinese population, including 194 in MLH1, 181 in MSH2, 59 in MSH6, 53 in PMS2 single-base/indel changes and 53 large deletions/duplications in MLH1, MSH2, MSH6 and PMS2, respectively. We determined that the pathogenic/likely pathogenic carrier rate in the Chinese population was 1.6%. Comparative analysis in variant spectrum, variant types, clinical classification and founder mutations showed substantial differences of MMR variation between Chinese and non-Chinese populations and the fact that over 90% of the variants were only present in the Chinese ethnicity reveals the highly ethnic-specific nature of the Chinese MMR variation . We also developed an open-access database, dbMMR-Chinese, to host all data (https://dbMMR-chinese.fhs.um.edu.mo). The rich MMR data from a large non-Caucasian population should be valuable to study MMR variation and its relationship with cancer and provide a valuable reference resource for MMR-related cancer prevention and treatment. Conclusion: Our study provides the largest MMR data set from a single non-Caucasian population and reveals that MMR variation in the humans can be highly ethnic-specific.
Article
Hereditary non‐polyposis colorectal cancer (HNPCC) is known to be associated with several extracolonic cancers, e.g., cancers of the endometrium, stomach, urinary tract, small bowel and ovary. An association between HNPCC and brain tumours has also been reported, although previous risk analysis did not reveal an excess of this type of tumour. To determine whether HNPCC predisposes patients to brain tumours, we used risk analysis to compare families with HNPCC to those in the general population. Of the 1,321 subjects from 50 HNPCC families (with 60,237 person‐years of follow‐up) in the Dutch HNPCC Registry which satisfy the Amsterdam Criteria, 312 had colorectal cancer. The registry revealed 14 brain tumours in the HNPCC‐patients and their first‐degree relatives: 5 astrocytomas, 3 oligodendrogliomas, 1 ependymoma and 5 tumours for which a pathological report was not available. The relative risk of brain tumour in patients with HNPCC and their first‐degree relatives was 6 times greater than in the general population (95% confidence interval, 3.5 to 10.1). After exclusion of the cases based only on family history, the relative risk was 4.3 (95% confidence interval, 2.3 to 8.0). Although the relative risk of brain tumour was increased, the lifetime risk was low (3.35%). Because it is not certain whether an improvement of the overall prognosis can be achieved by early diagnosis and intervention, and in view of the low lifetime risk, we do not recommend screening for brain tumours in HNPCC families. © 1996 Wiley‐Liss, Inc.
Article
The genetics of Hereditary Non‐Polyposis Colorectal Cancer (HNPCC) has recently been established and found to be associated with DNA mismatch repair deficiency. As the molecular basis of this syndrome does not appear to predict any particular disease, we compared families selected according to the “Amsterdam” criteria (AC) against families that were selected because of an aggregation of colonic and extracolonic malignancies (EC), all of which have been observed in HNPCC families. A comparison of the 2 groups revealed that there were significant differences between them. Age at disease onset for both groups was 20‐30 years younger than in the general population; however, a normal age distribution was observed for the AC group whereas for the EC group a bimodal distribution was apparent. The prognosis for both groups together did not differ from that of the general population; however, if split, the AC group had a significantly better outcome than the EC group. Furthermore, dividing the AC group into hMSH2‐ and hMLH1‐linked families revealed that there was no difference in severity of disease between these 2 groups with respect to survival and mean age of disease onset. Both the AC and EC groups displayed a similar tumour spectrum with a virtually identical tumour distribution. A significant finding was the over‐representation, of brain tumours in this family set, which comprised the third most common malignancy after endometrial and stomach cancer. Int. J. Cancer 74:281‐285, 1997. © 1997 Wiley‐Liss, Inc.
Article
Hereditary nonpolyposis colorectal cancer (HNPCC) dates to Warthin's description of family G, which he began studying in 1895. Warthin's observations were not fully appreciated until 1966 when two families with an autosomal dominant inheritance pattern of nonpolyposis colorectal cancer (CRC) and endometrial cancer were described. This condition was first termed the “cancer family syndrome” and was later renamed HNPCC. Some have proposed that HNPCC consists of at least two syndromes: Lynch syndrome I, with hereditary predisposition for CRC having early (~44 years) age of onset, a proclivity (70%) for the proximal colon, and an excess of synchronous and metachronous colonic cancers and Lynch syndrome II, featuring a similar colonic phenotype accompanied by a high risk for carcinoma of the endometrium. Transitional cell carcinoma of the ureter and renal pelvis and carcinomas of the stomach, small bowel, ovary, and pancreas also afflict some families. Current estimates indicate that HNPCC may account for as much as 6% of the total CRC burden. There are no known premonitory phenotypic signs or biomarkers of cancer susceptibility in the Lynch syndromes. This report will summarize current knowledge, with emphasis on the manner in which this knowledge can be employed effectively for diagnosis and management of HNPCC.
Article
HEREDITARY nonpolyposis colorectal cancer (HNPCC) is one of man's commonest hereditary diseases1. Several studies have implicated a defect in DNA mismatch repair in the pathogenesis of this disease2-8. In particular, hMSH2 and hMLHl homologues of the bacterial DNA mismatch repair genes mutS and mutL, respectively, were shown to be mutated in a subset of HNPCC cases9-16. Here we report the nucleotide sequence, chromosome localization and mutational analysis of hPMSl and hPMS2, two additional homologues of the prokaryotic mutL gene. Both hPMSl and hPMS2 were found to be mutated in the germline of HNPCC patients. This doubles the number of genes implicated in HNPCC and may help explain the relatively high incidence of this disease.
Article
The TGF@type H receptor(RI!) was found to be mutatedwithin a polyadeninetract in 100of 111(90%) colorectal cancers with microsatellite instability. Other polyadenine tracts of similar length were mutated in these samplesbut not as frequentlyas RI!. In most cases, the polyadeninetract mutationsaffectedboth allelesofRI!, and in four tumors heterozygousfor the polyadenine mutations, three had additional mutations that were expected to inactivate the other RH allele. These genetic data support the idea that RU behaves like a tumor suppressor during CR cancer development and is a critical target of inactivation in mismatch repair-deficient tumors.