ArticlePDF Available

Differential colorectal carcinogenesis: Molecular basis and clinical relevance

Authors:

Abstract and Figures

Colorectal cancer (CCR) is one of the most frequent cancers in developed countries. It poses a major public health problem and there is renewed interest in understanding the basic principles of the molecular biology of colorectal cancer. It has been established that sporadic CCRs can arise from at least two different carcinogenic pathways. The traditional pathway, also called the suppressor or chromosomal instability pathway, follows the Fearon and Vogelstein model and shows mutation in classical oncogenes and tumour suppressor genes, such as K-ras, adenomatous polyposis coli, deleted in colorectal cancer, or p53. Alterations in the Wnt pathway are also very common in this type of tumour. The second main colorectal carcinogenesis pathway is the mutator pathway. This pathway is present in nearly 15% of all cases of sporadic colorectal cancer. It is characterized by the presence of mutations in the microsatellite sequences caused by a defect in the DNA mismatch repair genes, mostly in hMLH1 or hMSH2. These two pathways have clear molecular differences, which will be reviewed in this article, but they also present distinct histopathological features. More strikingly, their clinical behaviours are completely different, having the "mutator" tumours a better outcome than the "suppressor" tumours.
Content may be subject to copyright.
Alberto Morán, Paloma Ortega, Carmen de Juan, Tamara Fernández-Marcelo, Cristina Frías, Andrés Sánchez-
Pernaute, Antonio José Torres, Eduardo Díaz-Rubio, Pilar Iniesta, Manuel Benito
REVIEW
Differential colorectal carcinogenesis: Molecular basis and
clinical relevance
Alberto Morán, Paloma Ortega, Carmen de Juan, Tamara
Fernández-Marcelo, Cristina Frías, Pilar Iniesta, Manuel
Benito, the second Department of Biochemistry and Molecular
Biology, School of Pharmacy, Complutense University, 28040-
Madrid, Spain
Andrés Sánchez-Pernaute, Antonio Jo Torres, Surgery
Service II, Clínico San Carlos Hospital, 28040-Madrid, Spain
Eduardo Díaz-Rubio, Oncology Service, Clínico San Carlos
Hospital, 28040-Madrid, Spain
Author contributions: Morán A, de Juan C, Frías C, Ortega P
and Fernández-Marcelo T performed the molecular analyses;
Sánchez-Pernaute A and Torres AJ assessed the clinical correlations;
Díaz-Rubio E was assessor of this work; Benito M directed and
coordinated this work; Benito M, Iniesta P and Morán A were
involved in writing the manuscript.
Supported by Grants from Ministerio de Sanidad y Consumo,
FIS PI080033; Fundación de Investigación Médica Mutua
Madrileña and RTICC RD06/0020/0021
Co rre spo nde nce to : Man uel Be nit o, PhD, the second
Department of Biochemistry and Molecular Biology, School of
Pharmacy, Complutense University, 28040-Madrid,
Spain. benito@farm.ucm.es
Telephone: +34-91-3941777 Fax: +34-91-3941779
Received: March 19, 2009 Revised: August 24, 2009
Accepted: August 31, 2009
Published online: March 15, 2010
Abstract
Colorectal cancer (CCR) is one of the most frequent
cancers in developed c ountr ies. It poses a ma jor
public health problem and there is renewed interest in
understanding the basic principles of the molecular biology
of colorectal cancer. It has been established that sporadic
CCRs can arise from at least two different carcinogenic
pathways. The traditional pathway, also called the
suppressor or chromosomal instability pathway, follows
the Fearon and Vogelstein model and shows mutation
in classical oncogenes and tumour suppressor genes,
such as
K-ras
, adenomatous polyposis coli, deleted in
colorectal cancer, or
p53
. Alterations in the Wnt pathway
are also very common in this type of tumour. The second
main colorectal carcinogenesis pathway is the mutator
pathway. This pathway is present in nearly 15% of all
cases of sporadic colorectal cancer. It is characterized by
the presence of mutations in the microsatellite sequences
caused by a defect in the DNA mismatch repair genes,
mostly in hMLH1 or hMSH2. These two pathways have
clear molecular differences, which will be reviewed in this
article, but they also present distinct histopathological
features. More strikingly, their clinical behaviours are
completely different, having the “mutator” tumours a
better outcome than the “suppressor” tumours.
© 2010 Baishideng. All rights reserved.
Key words: Colorectal cancer; Microsatellite instability;
Clinical outcome
Peer reviewer: Tzu-Chen Yen, PhD, Professor, Department of
Nuclear Medicine, Chang Gung Memorial Hospital, No.5, Fu-
Hsin St., Taoyuan 333, Taiwan, China
Morán A, Ortega P, de Juan C, Fernández-Marcelo T, Frías C,
Sánchez-Pernaute A, Torres AJ, Díaz-Rubio E, Iniesta P, Benito M.
Differential colorectal carcinogenesis: Molecular basis and clinical
relevance. World J Gastrointest Oncol
2010; 2(3): 151-158
Available from: URL: http://www.wjgnet.com/1948-5204/full/v2/
i3/151.htm DOI: http://dx.doi.org/10.4251/wjgo.v2.i3.151
INTRODUCTION
Colorectal cancer (CRC) is one of the most common
cancers in developed countries. The American Cancer
Society estimated that up to 153 760 new colorectal cancer
cases were diagnosed in USA during 2007 (the fourth
most common cancer for that period of time), with
52 180 associated deaths[1].
Online Submissions: http://www.wjgnet.com/1948-5204ofce
wjgo@wjgnet.com
doi:10.4251/wjgo.v2.i3.151
World J Gastrointest Oncol 2010 March 15; 2(3): 151-158
ISSN 1948-5204 (online)
© 2010 Baishideng. All rights reserved.
151 March 15, 2010
|
Volume 2
|
Issue 3
|
WJGO
|
www.wjgnet.com
Morán A
et al
. Differential colorectal carcinogenesis
Ferlay et al[2] estimated that colorectal cancer was the
second most common form of cancer in Europe during
2006 with 412,900 cancer diagnoses (12.9% of total
cancers) and 207,400 deaths (ranking second position).
These figures illustrate the clinical impact of colorectal
cancer. Due to the worldwide scale of the problem,
colorectal carcinogenesis is one of the most extensively
studied types of cancers.
CRC is traditionally divided into sporadic and familial
(hereditary) cases. Approximately, 75%-80% of colorectal
tumours have a sporadic origin. Of all patients, a high
proportion have one first to third-degree relative with
CRC. It is quite clear that even in sporadic CRC cases, the
descendants have a higher risk of suffering colorectal can-
cer. In this review, we will focus on sporadic CRC.
SPORADIC COLORECTAL CANCER:
DIFFERENTIAL CARCINOGENESIS,
DIFFERENT CLINICAL BEHAVIOUR
Currently, it is considered that there are two major path-
ways in colorectal carcinogenesis. One of them is called
the “canonical” (adenoma-carcinoma sequence) or “sup-
pressor” pathway and involves chromosomal instability
(CIN)[3]. It is characterized by allelic losses on chromo-
some 5q (APC), 17p (p53), and 18q (DCC/SMAD4).
The second pathway of colorectal carcinogenesis involves
microsatellite instability (MSI), and is called the “mutator”
pathway. The MSI pathway is present in approximately
15%-20% of sporadic CRCs[4].
Apart from their molecular differences, these two
pathways present different clinical behaviours and distinct
histopathological features, as will be discussed below.
SUPPRESSOR OR CANONICAL
PATHWAY
The “canonical” pathway is present in 80%-85% of
colorectal carcinomas and it is assumed to follow the
Fearon and Vogelstein approach. It is accepted that in
the majority of cases, carcinomas arise from pre-existing
adenomas. Fearon and Vogelstein[5] proposed a model of
colorectal carcinogenesis that correlates specic genetic
events with evolving tissue morphology. As is shown in
Figure 1, every step from the normal mucosa towards
the carcinoma involves specic and well-dened genetic
alterations. This linear model has evolved to a more
complex, comprehensive, and mechanistic approach[6].
However, in spite of the impact of new knowledge on
the Fearon and Vogelstein scheme, the model as such still
stands[7]. Alterations in tumour suppressor genes, such
as APC, p53, and DCC, and in oncogenes, such as K-ras,
are characteristic of this model and of the suppressor
pathway. CIN tumours are also characterized by a high
frequency of allelic imbalance (most commonly involving
chromosomal arms 5q, 8p, 17p, and 18q), chromosomal
amplications, and translocations[8].
APC
The adenomatous polyposis coli (APC, 5q21) gene contains
15 exons and it is mutated in 60% and 82% of colon and
rectal cancers, respectively[9]. Its best-known role is in the
Wnt pathway, where it is part of a multiprotein complex
that joins β-catenin and causes its phosphorylation, subse-
quent ubiquitination, and destruction in the proteosome.
This complex is mainly constituted by APC, axin, and
GSK3β. If this complex is disrupted, by multiple causes,
β-catenin is not directed towards degradation and is avail-
able to translocate to the nucleus and co-transactivate seve-
ral genes[10]. The list of Wnt target genes is quite long, but it
is important to note some cell cycle regulating genes (cyclin
D, c-Myc), and some genes related to tumour progression
(MMP-7, MMP-26). One of the main causes of disruption
of the multiprotein complex is mutations in APC. These
mutations interfere with binding to β-catenin and result in
the Wnt pathways becoming constitutively active.
However, APC also plays Wnt-independent roles,
whose alteration can also be related to carcinogenesis[11]. It
participates in cytoskeletal regulation, as it has been shown
to associate with microtubules and actin cytoskeleton,
suggesting that one role for APC may be in regulating
directed cell migration[12]. APC also has a role in mitosis.
APC has been reported at kinetochores, where it might
promote correct chromosomal alignment[13] and at centro-
somes, where it could inuence centrosome duplication[11].
It has been described that APC deficient cells cannot
properly detect chromosomal abnormalities during anap-
hase[14]. Therefore, loss of APC might interfere with the
correct regulation of mitosis and contribute to CIN[3].
Inactivation of APC has also been related to the promotion
of tumorigenesis, through loss of cell adhesion[15]. It has
been shown that a mutation in Apc in mice can decrease the
level of E-cadherin at the cell membrane[16]
Thus, we can consider that mutations in APC are a
frequent early event in the carcinogenesis of CCR and APC
is related to carcinogenesis at different levels: its activity in
the Wnt pathway, its relation with the cytoskeleton, its role
in chromosome segregation and, nally, its role in adhesion.
K-ras
K-ras is a proto-oncogene located at 12p12.1 that encodes
a 21-kDa GTP-binding protein. K-ras is frequently mu-
tated during the very early stages of colorectal cancer de-
velopment (35%-42% of colorectal cancers and advanced
adenomas present mutations on this proto-oncogene)[17].
When it is bound to GTP, the ras protein is active. This
protein is involved in many different processes. It activates
a large number of transduction signal pathways, among
them the mitogen-activated protein kinases (MAPK) path-
way. Recently, it has been demonstrated that mutant K-ras
promotes hyperplastic growth in the colonic epithelium
(signalling through MEK) and suppresses differentiation
in APC-mutant colon cancers[18]. It also regulates epithelial
cell polarity. During the development of CRC, epithelial
cells can loose their polarity and it has been described that
152
WJGO
|
www.wjgnet.com March 15, 2010
|
Volume 2
|
Issue 3
|
an acquired mutation in K-ras reduces adherens junction-
mediated cell–cell contacts[19].
DCC
DCC (deleted in colorectal cancer) is located at 18q21.1 and
has been proposed as a tumour suppressor gene. About
70% of colorectal cancers show allelic losses in DCC[20];
some cancers had somatic mutations of this gene, and its
expression is often reduced in colorectal cancer tissues
and cell lines[21]. The DCC protein is a transmembrane
receptor of the Ig superfamily for netrins, factors involved
in axon guidance in the developing nervous system.
However, DCC has a role not only in axon guidance, but
in intracellular signalling. Chen et al[22] demonstrated that
the wild-type DCC, but not the mutant, induced apoptosis
and activated caspase-3, and that DCC expression induces
a rapid G2/M cell cycle arrest in some cell lines. DCC was
also shown to activate Rac-1 when netrin-1 is present[23];
thus it is implicated in actin organization and cell motility.
As reviewed by Mehlen and Fearon[21], transgenic mice
expressing a constitutive form of Rac-1 in the intestine
showed differentiation of the epithelium with accompanying
alterations in their apical actin. Hence, DCC-mediated Rac-1
activation might be important for epithelium differentiation.
p53
p53 is encoded by the TP53 gene located on 17p13.1. Its
expression is abnormal in more than 50% of human tu-
mours[24]. Mutation or loss of p53 usually occurs at the time
of the transition from adenoma to cancer in the Fearon
and Vogelstein sequence. As reviewed by Worthley[3], the
frequency of p53 abnormalities increases with the progres-
sion of the lesion. Thus alterations are found in 4%-26%
of adenomas, 50% of adenomas with invasive foci, and in
50%-75% of CRCs[3,17]. P53 protein induces G1 cell-cycle
arrest to facilitate DNA repair during replication of cells
exposed to environmental or oncogenic stress[20]. When
DNA damage is too great to be repaired, it can induce
apoptosis and this is considered a major pathway whereby
p53 exerts its tumour suppressor function[25].
MSI PATHWAY
Molecular alterations
The MSI, or mutator pathway, is present in approximately
15%-20% of sporadic CRCs. MSI tumours (also called
Replication ERor, RER+) are characterized by a huge
accumulation of mutations (mutation rates in these tumour
cells are 100- to 1000-fold more common compared to
normal cells[8]) in microsatellite sequences (High Micro-
satellite Instability, MSI-H). Microsatellites are short sequ-
ences repeated in tandem throughout the genome[26,27]. This
accumulation of frameshift mutations is caused by a primary
defect in the mismatch repair (MMR) genes (Figure 2).
There are at least seven genes in the MMR system: hMLH1,
hMLH3, hMSH2, hMSH3, hMSH6, hPMS1 and hPMS2[28].
When MMR proteins are functional, errors made by DNA
polymerase in microsatellite sequences during replication,
are repaired. The acquisition of thousands of mutations
characteristic of the MSI-H phenotype, requires the
inactivation of the MMR genes[4]. Germline mutations, or
epigenetic changes, in hMLH1 (mainly silencing caused by
methylation) and hMSH2 are the most common cause of
MSI-H in sporadic CRC (and in HNPCC, Hereditary Non
Polyposis Colorectal Cancer). hMSH6 mutations are less
frequent and alterations of the other MMR genes are very
rare[4]. These data enforce the idea that loss of hMLH1 and
hMSH2 is associated with complete inactivation of MMR,
whereas defects in other proteins cause only a partial MMR
deciency[28].
MSI-H sporadic colorectal cancers do not show big
cytogenetic abnormalities and are usually not aneuploid[29].
This type of tumour presents reduced frequency, or
absence, of mutation or allelic losses at the genes usually
altered in the “suppressor” pathway, APC, K-ras and p53,
and loss of heterozygosity at 5q, 17p, and 18q[30]. Instead,
mutations are described in microsatellite sequences present
in genes implicated in colorectal carcinogenesis, such
as TGF
β
RII[31] , IGF2R[ 32], BAX[33], MSH3[34] , MSH6[34],
caspase 5[35], APC[36],
β
-catenin[37], Tcf-4[38], axin[39], MMP-3[40],
E2F-4[41], BCL-10[42], cdx-2[43], and hRAD50[44] (See Table 1
for further information). Additionally, a number of normally
153WJGO
|
www.wjgnet.com March 15, 2010
|
Volume 2
|
Issue 3
|
Morán A
et al
. Differential colorectal carcinogenesis
Normal
epithelium
Micro
adenoma
Early
adenoma
Intermediate
adenoma
Late
adenoma Carcinoma Metastasis
Suppressor pathway
APC
mutation
Chromosome
5q LOH
K-ras
mutation
18q LOH,
DCC mutation
p53 mutation,
17p LOH
Other
alterations
Figure 1 Molecular alterations in the suppressor pathway.
functioning genes are silenced by methylation. Most sporadic
MSI-H cancers show the CpG island methylator phenotype,
characterised by widespread DNA hypermethylation[30].
Dening MSI-H tumours
International consensus criteria for classifying a tumour
as MSI-H were established in 1998[45]. A panel of five
microsatellite sequences was proposed for defining
the MSI-H tumour groups. The recommended panel
is composed of two mononucleotide repeats (BAT26
and A4725) and three dinucleotide repeats (D5S346,
D2S123, and D17S250). MSI-H tumours are dened as
having instability in two or more markers, whereas MSI-L
tumours are dened as having instability in one marker.
Microsatellite stability (MSS) is dened by no instability at
those ve loci. It is also important to stress that instability
is dened as a change of any length due to either insertion
or deletion of repeating units, in a microsatellite within a
tumour, when compared to normal tissue.
Clinical and histopathological characteristics
One of the most important and intriguing characteristics
of individuals with MSI-H tumours is that they have
distinct clinical and histopathological features. This is why
it is so important to determine a patients carcinogenic
pathway. Samowitz et al[46] reported that MSI-H was more
frequent in individuals with colorectal cancer diagnosed
before the age of 55 or over the age of 70, than in those
between 55 years and 70 years of age. However, these
data have not been confir med by other authors[47,48].
MSI-H tumours are located predominantly in the right-
sided colon[48-52] and have generally been reported more
frequently in women[46,53]. It has been proposed that the
analysis of MSI in CRCs might be helpful in predicting
the development of metachronous multiple colorectal
carcinomas[54].
MSI-H colorectal tumours typically present with a grea-
ter depth of invasion but with a lower overall stage[49,50,52].
MSI has also been associated with the presence of local
lymphocyte inltration and low frequency of distant metas-
tases[48,50,55]. In spite of its longer survival after surgical
resection (see below, in MSI-H and prognosis), MSI-H
carcinomas tend to be poorly differentiated[52]. Some studies
have demonstrated that MSI-H occurs more frequently
in mucinous-cell type tumours[53,56,57], but others have not
found any difference in histological cell type[49]. The absence
of dirty necrosis is also associated with MSI-H[50]. However,
as reviewed in Raut[52], it is not yet possible to use a single
pathological feature to diagnosis MSI-H. Greenson et al[50,58]
have recently developed a model that permits pathologists to
predict the likelihood of MSI using a combination of simple
histological and clinical data (mucinous differentiation,
lymphocyte inltration, and dirty necrosis).
MSI-H and prognosis
Many authors have reported a better outcome for MSI-H
tumours (whether sporadic or inherited) than those with
MSI-L or MSS tumours[46,47,59-62], though others have
not[63,64]. The prognostic advantage conferred by the
presence of high instability has been shown to be most
evident in stage and disease[62]. Individuals with
distant metastases present (stage ) showing MSI-H
in the TGF-
β
R
gene (transforming growth factor-β
receptor ) had improved prognosis as compared with
those with native TGF-βR[46]. Moreover, MSI status
is considered to be predictive of a favourable outcome,
independent of tumour stage and of patient treatment[49].
Therefore, the MSI-H phenotype is associated with a good
prognosis, independently of the molecular biology (germ
line mutations or transcriptional silencing via hyperme-
thylation) provoking it[52]. In the short term, therapeutic
decisions might be taken in MSI-H patients considering
this differential prognosis. For example, Benatti et al[62]
demonstrated that 5-FU-based chemotherapy does not
seem to provide survival benefits among patients with
MSI-H tumours, so the use of 5-FU in patients with
MSI-H tumours should be limited to avoid harmful side
effects of unnecessary chemotherapeutic regimens
Classication of colorectal cancer by MSI status might
also have prognostic value in patients undergoing curative
surgery, as suggested by Banerjea[65] . MSI-H cancers
display enhanced immunogenic properties and this might
contribute to their better prognosis.
154WJGO
|
www.wjgnet.com March 15, 2010
|
Volume 2
|
Issue 3
|
Morán A
et al
. Differential colorectal carcinogenesis
hMLH1 hMSH2
hPMS1
hPMS2 hMLH3
hMSH6 hMSH3
hMLH1 hMSH2
hPMS1
hPMS2 hMLH3
hMSH6 hMSH3
DNA mismatch error
(base-base mismatch,
insertion, deletion
mismatch)
loop
DNA repaired Loss of function
Thousands of frameshift
mutations in coding and
non-coding
microsatellite sequences
Figure 2 Origin of the high microsatellite instability phenotype.
MSI-H VS MSI-L
As mentioned above, MSI-L tumours (also called mild
mutator phenotype) are defined as having instability
in one marker out of the five consensus microsatellite
sequences (as defined by Boland in 1998[45]). However,
not everybody denes MSI-L with the same criteria. The
distinction between MSI-H and MSI-L depends on both
the type and the number of microsatellites analyzed. For
example, mononucleotide markers, such as BAT26 and
BAT40, are relatively specic for MSI-H cancers[8]. This is
the reason why some groups use specic markers, such as
MYCL, for dening MSI-L tumours.
MSI-L tumours have been considered by some authors
to be halfway between MSI-H and MSS. However, MSI-L
colorectal tumours do not show clear differences in their
clinicopathological features when compared with the
classical “suppressor” tumours[66]. Yearsley et al[57] found
no difference between MSI-L vs MSS using clinical and
histological parameters such as percentage of mucin,
histological type, grade, and lymphoid host response.
Moreover, its molecular characteristics are more similar to
those from MSS than MSI-H tumours (reviewed in[8]). For
example, it has been described that LOH at 1p32, 2p16,
7q31, 8p12-22, and 17q11 is more frequent in MSI-L
than in MSI-H[66-68] and that K-ras mutations occur more
frequently in MSI-L carcinomas than in MSI-H colorectal
tumours, with no difference in frequency between MSS
and MSS-L cancers, by some authors[66,68]. The rate of
K-ras mutation is higher in the MSI-L group than in the
stable cancers[69]. Analysis of mutations in MSI-H target
genes revealed that they are absent in MSI-L tumours[4].
Some authors have even wondered about the real exist-
ence of the MSI-L group of tumours[70]. However, Jass
and others defended the notion that MSI-L is a separate
group of tumours, arguing that when a panel of sensitive
markers is used, approximately 8% of sporadic colorectal
cancers can be classied as MSI-L[69,71]. Others authors have
demonstrated the existence of specic markers for MSI-L,
such as MYCL and D2S123, which are mutated at a higher
rate outside the MSI-H subset[66].
In conclusion, we can consider that MSI-L CRCs are
indistinguishable from MSS using most clinicopathological
parameters. However, these tumours can be validated as
a distinct molecular phenotypic category, as they present
molecular alterations different from MSI-H and MSS
(reviewed in reference [4]).
SPORADIC MSI-H TUMOURS VS HNPCC
HNPCC (Lynch syndrome) constitutes approximately
2%-4% of all CRC cases[72]. The presence of MSI is also
a hallmark of this type of hereditary cancer. However,
the molecular mechanism causing MSI-H is different
in sporadic CRC than in HNPCC. In sporadic CRCs,
MSI-H is p rovoked mainly by epig enetic sile ncing
(hypermethylation) on hMLH1, whereas in HNPCC is
more frequent a germ line mutation in an MMR gene,
followed by a “second hit”.
Most of the molecular characteristics of sporadic
MSI-H tumours and HNPCCs are similar. However, some
small differences have been described recently. Oliveira
et al[73] demonstrated the presence of distinct patterns of
K-ras mutations in cancers according to hMLH1 methy-
lation status and germ line DNA MMR defects. BRAF
mutations (a serine/threonine kinase involved in the
RAS/RAF/MAPK pathway) in a specific hotspot site
have been more frequently detected in sporadic MSI-H
tumours than in HNPCCs[74].
There is no difference in overall survival amongst
MSI-H patients with HNPCC and those with sporadic
CRC[49].
CONCLUSION
Sporadic colorectal cancers can be classied in two clear-
ly different subtypes, according to the molecular events
that give rise to the tumour. The rst one is the so-called
canonical, CIN or suppressor pathway. It is the most fre-
quent pathway and it is characterized by mutation or de-
letion of K-ras, APC, DCC, and p53, among others genes.
The specic genetic events that occur during this pathway
have clear correlations with evolving tissue morphology.
The second pathway is the mutator or MSI-H pathway,
155WJGO
|
www.wjgnet.com March 15, 2010
|
Volume 2
|
Issue 3
|
Morán A
et al
. Differential colorectal carcinogenesis
Table 1 Genes with microsatellite instability in their coding
sequences. Adapted from reference [75].
Gene Function Coding repeat
ACTR
Growth/differentiation factor
receptor
(A) 8
AIM2 IFN inducible (A) 10
AXIN-2 Wnt signaling (A) 6*2, (G) 7, (C) 6
BAX Proapoptotic factor (G) 8
BCL-10 Proapoptotic factor (A) 8
BLM Response to DNA damage (A) 9
Caspase-5 Proapoptotic factor (A) 10
CDX2 Homeobox transcription factor (G) 7
CHEK1 Response to DNA damage (A) 9
FAS Proapoptotic factor (T) 7
GRB-14 Growth factor bound protein (A) 9
hG4-1 Cell cycle (A) 8
IGFIIR Growth factor receptor (G) 8
KIAAO977 Homologue to mouse cordon bleu (T) 9
MBD-4 DNA glycosylase and methyl CpG
binding protein
(A) 10
MLH3 MMR (A) 9
MSH3 MMR (A) 8
MSH6 MMR (C) 8
NADH-UOB NADH ubiquinone oxidoreductase (T) 9
OGT O-linked GlcNAc transferase (T) 10
PTEN Cell cycle (A) 6*2
RAD-50 Response to DNA damage (A) 9
RHAMM Cell motility (A) 9
RIZ Cell cycle and apoptotic protein (A) 8, (A) 9
SEC63 ER membrane protein (A) 10, (A) 9
SLC23AI Nucleobase transporter (C) 9
TCF-4 Transcription factor (Wnt pathway) (A) 9
TGF
β
R
Growth factor receptor (A) 10
WISP-3 Growth factor (Wnt pathway) (A) 9
which is less frequent. Its main molecular characteristic
is a huge accumulation of mutations in microsatellite
sequences throughout the genome, caused by primary
alteration in the MMR genes. As well as their important
molecular differences, the existence of these two path-
ways is relevant for their different phenotypes. MSI-H
tumours and CIN tumours have distinct clinical and his-
topathological features. Known molecular differences be-
tween the two groups of tumours are still not sufcient to
fully explain why MSI-H tumours have a better outcome;
its most intriguing characteristic. Recent studies are begin-
ning to shed light on this differential clinical behaviour,
but further work is required.
REFERENCES
1 Data obtained from the American Cancer Society. Available
from: URL: http://www.cancer.org/downloads/stt/
CFF2007EstCsDths07.pdf
2 Ferlay J, Autier P, Boniol M, Heanue M, Colombet M, Boyle P.
Estimates of the cancer incidence and mortality in Europe in
2006. Ann Oncol 2007; 18: 581-592
3 Worthley DL, Whitehall VL, Spring KJ, Leggett BA. Colorectal
carcinogenesis: road maps to cancer. World J Gastroenterol 2007;
13: 3784-3791
4 Imai K, Yamamoto H. Carcinogenesis and microsatellite insta-
bility: the interrelationship between genetics and epigenetics.
Carcinogenesis 2008; 29: 673-680
5 Fearon ER, Vogelstein B. A genetic model for colorectal
tumorigenesis. Cell 1990; 61: 759-767
6 Gatenby RA, Vincent TL. An evolutionary model of carcino-
genesis. Cancer Res 2003; 63: 6212-6220
7 Arends JW. Molecular interactions in the Vogelstein model of
colorectal carcinoma. J Pathol 2000; 190: 412-416
8 Pawlik TM, Raut CP, Rodriguez-Bigas MA. Colorectal
carcinogenesis: MSI-H versus MSI-L. Dis Markers 2004; 20:
199-206
9 Jass JR. Pathogenesis of colorectal cancer. Surg Clin North Am
2002; 82: 891-904
10 Barker N. The canonical Wnt/beta-catenin signalling pathway.
Methods Mol Biol 2008; 468: 5-15
11 Rusan NM, Peifer M. Original CIN: reviewing roles for APC in
chromosome instability. J Cell Biol 2008; 181: 719-726
12 Näthke IS, Adams CL, Polakis P, Sellin JH, Nelson WJ. The
adenomatous polyposis coli tumor suppressor protein localizes
to plasma membrane sites involved in active cell migration. J
Cell Biol 1996; 134: 165-179
13 Green RA, Wollman R, Kaplan KB. APC and EB1 function
together in mitosis to regulate spindle dynamics and chrom-
osome alignment. Mol Biol Cell 2005; 16: 4609-4622
14 Draviam VM, Shapiro I, Aldridge B, Sorger PK. Misorientation
and reduced stretching of aligned sister kinetochores promote
chromosome missegregation in EB1- or APC-depleted cells.
EMBO J 2006; 25: 2814-2827
15 Senda T, Iizuka-Kogo A, Onouchi T, Shimomura A.
Adenomatous polyposis coli (APC) plays multiple roles in the
intestinal and colorectal epithelia. Med Mol Morphol 2007; 40:
68-81
16 Carothers AM, Melstrom KA Jr, Mueller JD, Weyant MJ,
Bertagnolli MM. Progressive changes in adherens junction
structure during intestinal adenoma formation in Apc mutant
mice. J Biol Chem 2001; 276: 39094-39102
17 Leslie A, Carey FA, Pratt NR, Steele RJ. The colorectal
adenoma-carcinoma sequence. Br J Surg 2002; 89: 845-860
18 Haigis KM, Kendall KR, Wang Y, Cheung A, Haigis MC,
Glickman JN, Niwa-Kawakita M, Sweet-Cordero A, Sebolt-
Leopold J, Shannon KM, Settleman J, Giovannini M, Jacks
T. Dif ferential e ffe cts of oncogen ic K-Ras and N-Ra s on
proliferation, differentiation and tumor progression in the
colon. Nat Genet 2008; 40: 600-608
19 Smakman N, Borel Rinkes IH, Voest EE, Kranenburg O.
Control of colorectal metastasis formation by K-Ras. Biochim
Biophys Acta 2005; 1756: 103-114
20 Takayama T, Miyanishi K, Hayashi T, Sato Y, Niitsu Y.
Colorectal cancer: genetics of development and metastasis. J
Gastroenterol 2006; 41: 185-192
21 Mehlen P, Fearon ER. Role of the dependence receptor DCC in
colorectal cancer pathogenesis. J Clin Oncol 2004; 22: 3420-3428
22 Chen YQ, Hsieh JT, Yao F, Fang B, Pong RC, Cipriano SC,
Krepulat F. Induction of apoptosis and G2/M cell cycle arrest
by DCC. Oncogene 1999; 18: 2747-2754
23 Shekarabi M, Kennedy TE. The netrin-1 receptor DCC
promotes lopodia formation and cell spreading by activating
Cdc42 and Rac1. Mol Cell Neurosci 2002; 19: 1-17
24 Munro AJ, Lain S, Lane DP. P53 abnormalities and outcomes
in colorectal cancer: a systematic review. Br J Cancer 2005; 92:
434-444
25 Pietsch EC, Sykes SM, McMahon SB, Murphy ME. The
p53 family and programmed cell death. Oncogene 2008; 27:
6507-6521
26 Thibodeau SN, Bren G, Schaid D. Microsatellite instability in
cancer of the proximal colon. Science 1993; 260: 816-819
27 Ionov Y, Peinado MA, Malkhosyan S, Shibata D, Perucho M.
Ubiquitous somatic mutations in simple repeated sequences
reveal a new mechanism for colonic carcinogenesis. Nature
1993; 363: 558-561
28 Hoeijmakers JH. Genome maintenance mechanisms for
preventing cancer. Nature 2001; 411: 366-734
29 Eshleman JR, Casey G, Kochera ME, Sedwick WD, Swinler
SE, Veigl ML, Willson JK, Schwartz S, Markowitz SD.
Chromosome number and structure both are markedly stable
in RER colorectal cancers and are not destabilized by mutation
of p53. Oncogene 1998; 17: 719-725
30 Jass JR, Walsh MD, Barker M, Simms LA, Young J, Leggett BA.
Distinction between familial and sporadic forms of colorectal
cancer showing DNA microsatellite instability. Eur J Cancer
2002; 38: 858-866
31 Markowitz S, Wang J, Myeroff L, Parsons R, Sun L, Lutter-
baugh J, Fan RS, Zborowska E, Kinzler KW, Vogelstein B.
Inactivation of the type II TGF-beta receptor in colon cancer
cells with microsatellite instability. Science 1995; 268: 1336-1338
32 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
33 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
34 Yamamoto H, Sawai H, Perucho M. Frameshift somatic
mutations in gastrointestinal cancer of the microsatellite
mutator phenotype. Cancer Res 1997; 57: 4420-4426
35 Schwartz S Jr, Yamamoto H, Navarro M, Maestro M, Reventós
J, Perucho M. Frameshift mutations at mononucleotide
repeats in caspase-5 and other target genes in endometrial and
gastrointestinal cancer of the microsatellite mutator phenotype.
Cancer Res 1999; 59: 2995-3002
36 Fang DC, Luo YH, Yang SM, Li XA, Ling XL, Fang L. Mutation
analysis of APC gene in gastric cancer with microsatellite
instability. World J Gastroenterol 2002; 8: 787-791
37 Kitaeva MN, Grogan L, Williams JP, Dimond E, Nakahara K,
Hausner P, DeNobile JW, Soballe PW, Kirsch IR. Mutations in
beta-catenin are uncommon in colorectal cancer occurring in
occasional replication error-positive tumors. Cancer Res 1997;
57: 4478-4481
156WJGO
|
www.wjgnet.com March 15, 2010
|
Volume 2
|
Issue 3
|
Morán A
et al
. Differential colorectal carcinogenesis
157WJGO
|
www.wjgnet.com March 15, 2010
|
Volume 2
|
Issue 3
|
Morán A
et al
. Differential colorectal carcinogenesis
38 Duval A, Iacopetta B, Ranzani GN, Lothe RA, Thomas G,
Hamelin R. Variable mutation frequencies in coding repeats of
TCF-4 and other target genes in colon, gastric and endometrial
carcinoma showing microsatellite instability. Oncogene 1999; 18:
6806-6809
39 Shimizu Y, Ikeda S, Fujimori M, Kodama S, Nakahara M,
Okajima M, Asahara T. Frequent alterations in the Wnt
signaling pathway in colorectal cancer with microsatellite
instability. Genes Chromosomes Cancer 2002; 33: 73-81
40 Morán A, Iniesta P, de Juan C, González-Quevedo R, Sánchez-
Pernaute A, Díaz-Rubio E, Ramón y Cajal S, Torres A, Balibrea
JL, Benito M . S tromelysin-1 promote r m utations impair
gelatinase B activation in high microsatellite instability sporadic
colorectal tumors. Cancer Res 2002; 62: 3855-3860
41 Fujiwara T, Stolker JM, Watanabe T, Rashid A, Longo P,
Eshleman JR, Booker S, Lynch HT, Jass JR, Green JS, Kim H,
Jen J, Vogelstein B, Hamilton SR. Accumulated clonal genetic
alterations in familial and sporadic colorectal carcinomas with
widespread instability in microsatellite sequences. Am J Pathol
1998; 153: 1063-1078
42 Simms LA, Young J, Wicking C, Meltzer SJ, Jass JR, Leggett BA.
The apoptotic regulatory gene, BCL10, is mutated in sporadic
mismatch repair deficient colorectal cancers. Cell Death Differ
2000; 7: 236-237
43 Wicking C, Simms LA, Evans T, Walsh M, Wicking C, Simms
LA, Evans T, Walsh M, Chawengsaksophak K, Beck F,
Chenevix-Trench G, Young J, Jass J, Leggett B, Wainwright B.
CDX2, a human homologue of Drosophila caudal, is mutated
in both alleles in a replication error positive colorectal cancer.
Oncogene 1998; 17: 657-659
44 Kim NG, Choi YR, Baek MJ, Kim YH, Kang H, Kim NK, Min
JS, Kim H. Frameshift mutations at coding mononucleotide
repeats of the hRAD50 gene in gastrointestinal carcinomas with
microsatellite instability. Cancer Res 2001; 61: 36-38
45 Boland CR, Thibodeau SN, Hamilton SR, Sidransky D,
Eshleman JR, Burt RW, Meltzer SJ, Rodriguez-Bigas MA, Fodde
R, Ranzani GN, 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 1998; 58: 5248-5257
46 Samowitz WS, Curtin K, Ma KN, Schaffer D, Coleman LW,
Leppert M, Slattery ML. Microsatellite instability in sporadic
colon cancer is associated with an improved prognosis at the
population level. Cancer Epidemiol Biomarkers Prev 2001; 10:
917-923
47 Ribic CM, Sargent DJ, Moore MJ, Thibodeau SN, French AJ,
Goldberg RM, Hamilton SR, Laurent-Puig P, Gryfe R, Shepherd
LE, Tu D, Redston M, Gallinger S. Tumor microsatellite-
instability status as a predictor of benefit from fluorouracil-
based adjuvant chemotherapy for colon cancer. N Engl J Med
2003; 349: 247-257
48 Jeong SY, Shin KH, Shin JH, Ku JL, Shin YK, Park SY, Kim
WH, Park JG. Microsatellite instability and mutations in DNA
mismatch repair genes in sporadic colorectal cancers. Dis Colon
Rectum 2003; 46: 1069-1077
49 Gryfe R, Kim H, Hsieh ET, Aronson MD, Holowaty EJ, Bull SB,
Redston M, Gallinger S. Tumor microsatellite instability and
clinical outcome in young patients with colorectal cancer. N
Engl J Med 2000; 342: 69-77
50 Greenson JK, Bonner JD, Ben-Yzhak O, Cohen HI, Miselevich
I, Resnick MB, Trougouboff P, Tomsho LD, Kim E, Low M,
Almog R, Rennert G, Gruber SB. Phenotype of microsatellite
unstable colorectal carcinomas: Well-differentiated and focally
mucinous tumors and the absence of dirty necrosis correlate
with microsatellite instability. Am J Surg Pathol 2003; 27: 563-570
51 Gerva z P, Bucher P, Morel P. Two colons-two cancers:
paradigm shift and clinical implications. J Surg Oncol 2004; 88:
261-266
52 Raut CP, Pawlik TM, Rodriguez-Bigas MA. Clinicopathologic
features in colorectal cancer patients with microsatellite
instability. Mutat Res 2004; 568: 275-282
53 Ward R, Meagher A, Tomlinson I, O'Connor T, Norrie
M, Wu R, Hawkins N. Microsatellite instability and the
clinicopathological features of sporadic colorectal cancer. Gut
2001; 48: 821-829
54 Masubuchi S, Konishi F, Togashi K, Okamoto T, Senba S,
Shitoh K, Kashiwagi H, Kanazawa K, Tsukamoto T. The
significance of microsatellite instability in predicting the
development of metachronous multiple colorectal carcinomas
in patients with nonfamilial colorectal carcinoma. Cancer 1999;
85: 1917-1924
55 Buckowitz A, Knaebel HP, Benner A, Bläker H, Gebert J, Kienle
P, von Knebel Doeberitz M, Kloor M. Microsatellite instability
in colorectal cancer is associated with local lymphocyte
inltration and low frequency of distant metastases. Br J Cancer
2005; 92: 1746-1753
56 Leopo ldo S, Loren a B, Cinzia A, Gabr iella DC, Angela
Luciana B, Renato C, Antonio M, Carlo S, Cristina P, Stefano
C, Maurizio T, Luigi R, Cesare B. Two subtypes of mucinous
adenocarcinoma of the colorectum: clinicopathological and
genetic features. Ann Surg Oncol 2008; 15: 1429-1439
57 Yearsley M, Hampel H, Lehman A, Nakagawa H, de la Chapelle
A, Frankel WL. Histologic features distinguish microsatellite-
high from microsatellite-low and microsatellite-stable colorectal
carcinomas, but do not differentiate germline mutations from
methylation of the MLH1 promoter. Hum Pathol 2006; 37:
831-838
58 Greenson JK, Huang SC, Herron C, Moreno V, Bonner JD,
Tomsho LP, Ben-Izhak O, Cohen HI, Trougouboff P, Bejhar
J, Sova Y, Pinchev M, Rennert G, Gruber SB. Pathologic
predictors of microsatellite instability in colorectal cancer. Am J
Surg Pathol 2009; 33: 126-133
59 Watanabe T, Wu TT, Catalano PJ, Ueki T, Satriano R, Haller
DG, Benson AB 3rd, Hamilton SR. Molecular predictors of
survival after adjuvant chemotherapy for colon cancer. N Engl J
Med 2001; 344: 1196-1206
60 Elsaleh H, Iacopetta B. Microsatellite instability is a predictive
marker for survival benet from adjuvant chemotherapy in a
population-based series of stage III colorectal carcinoma. Clin
Colorectal Cancer 2001; 1: 104-109
61 Guidoboni M, Gafà R, Viel A, Doglioni C, Russo A, Santini
A, Del Tin L, Macrì E, Lanza G, Boiocchi M, Dolcetti R.
Microsatellite instability and high content of activated cytotoxic
lymphocytes identify colon cancer patients with a favorable
prognosis. Am J Pathol 2001; 159: 297-304
62 Benatti P, Gafà R, Barana D, Marino M, Scarselli A, Pedroni
M, Maestri I, Guerzoni L, Roncucci L, Menigatti M, Roncari
B, Maffei S, Rossi G, Ponti G, Santini A, Losi L, Di Gregorio C,
Oliani C, Ponz de Leon M, Lanza G. Microsatellite instability
and colorectal cancer prognosis. Clin Cancer Res 2005; 11:
8332-8340
63 Ko JM, Cheung MH, Kwan MW, Wong CM, Lau KW, Tang
CM, Lung ML. Genomic instability and alterations in Apc, Mcc
and Dcc in Hong Kong patients with colorectal carcinoma. Int J
Cancer 1999; 84: 404-409
64 Salahshor S, Kressner U, Fischer H, Lindmark G, Glimelius B,
Påhlman L, Lindblom A. Microsatellite instability in sporadic
colorectal cancer is not an independent prognostic factor. Br J
Cancer 1999; 81: 190-193
65 Banerjea A, Hands RE, Powar MP, Bustin SA, Dorudi S. Microsa-
tellite and chromosomal stable colorectal cancers demonstrate
poor immunogenicity and early disease recurrence. Colorectal
Dis 2009; 11: 601-608
66 Laiho P, Launonen V, Lahermo P, Esteller M, Guo M, Herman
JG, Mecklin JP, Järvinen H, Sistonen P, Kim KM, Shibata D,
Houlston RS, Aaltonen LA. Low-level microsatellite instability
in most colorectal carcinomas. Cancer Res 2002; 62: 1166-1170
67 Kambara T, Matsubara N, Nakagawa H, Notohara K, Nagasaka
T, Yoshino T, Isozaki H, Sharp GB, Shimizu K, Jass J, Tanaka N.
158WJGO
|
www.wjgnet.com March 15, 2010
|
Volume 2
|
Issue 3
|
Morán A
et al
. Differential colorectal carcinogenesis
High frequency of low-level microsatellite instability in early
colorectal cancer. Cancer Res 2001; 61: 7743-7746
68 Gebert J, Sun M, Ridder R, Hinz U, Lehnert T, Möller P,
Schackert HK, Herfarth C, von Knebel Doeberitz M. Molecular
profiling of sporadic colorectal tumors by microsatellite
analysis. Int J Oncol 2000; 16: 169-179
69 Jass JR, Biden KG, Cummings MC, Simms LA, Walsh M,
Schoch E, Meltzer SJ, Wright C, Searle J, Young J, Leggett BA.
Characterisation of a subtype of colorectal cancer combining
features of the suppressor and mild mutator pathways. J Clin
Pathol 1999; 52: 455-460
70 Tomlinson I, Halford S, Aaltonen L, Hawkins N, Ward R. Does
MSI-low exist? J Pathol 2002; 197: 6-13
71 Jass JR, Whitehall VL, Young J, Leggett B, Meltzer SJ,
Matsubara N, Fishel R. Correspondence re: P. Laiho et al., Low-
level microsatellite instability in most colorectal carcinomas.
Cancer Res., 62: 1166-1170, 2002. Cancer Res 2002; 62: 5988-5989;
author reply 5989-5990
72 Umar A. Lynch syndrome (HNPCC) and mic rosat ellit e
instability. Dis Markers 2004; 20: 179-180
73 Oliveira C, Westra JL, Arango D, Ollikainen M, Domingo E,
Ferreira A, Velho S, Niessen R, Lagerstedt K, Alhopuro P, Laiho
P, Veiga I, Teixeira MR, Ligtenberg M, Kleibeuker JH, Sijmons
RH, Plukker JT, Imai K, Lage P, Hamelin R, Albuquerque
C, Schwartz S Jr, Lindblom A, Peltomaki P, Yamamoto H,
Aaltonen LA, Seruca R, Hofstra RM. Distinct patterns of KRAS
mutations in colorectal carcinomas according to germline
mismatch repair defects and hMLH1 methylation status. Hum
Mol Genet 2004; 13: 2303-2311
74 Deng G, Bell I, Crawley S, Gum J, Terdiman JP, Allen BA, Truta
B, Sleisenger MH, Kim YS. BRAF mutation is frequently present
in sporadic colorectal cancer with methylated hMLH1, but not
in hereditary nonpolyposis colorectal cancer. Clin Cancer Res
2004; 10: 191-195
75 Duval A, Hamelin R. Mutations at coding repeat sequences
in mismatch repair-deficient human cancers: toward a new
concept of target genes for instability. Cancer Res 2002; 62:
2447-2454
S- Editor Li LF L- Editor Stewart G E- Editor Yang C
... With this tool, an AI model was developed to recognize persistent histological inflammation with a specificity of 97% and sensitivity of 74% [20]. Mossotto et al [22] presented a model that used histological and endoscopic data to differentiate pediatric IBD between ulcerative colitis and Crohn's disease. The accuracy of this method was 82.7%, and the presence of ileal disease was the most important factor in the classification of the disease [22]. ...
... Mossotto et al [22] presented a model that used histological and endoscopic data to differentiate pediatric IBD between ulcerative colitis and Crohn's disease. The accuracy of this method was 82.7%, and the presence of ileal disease was the most important factor in the classification of the disease [22]. ...
... e etiologies of CRC are either genetic or environmental or both [11]. CRC is divided into hereditary and sporadic forms, with approximately 75-80% of sporadic forms [12]. ...
... With respect to sporadic CRC, Fearon et al. have suggested a colorectal carcinogenesis model that correlated specific genetic landscapes with changing tissue morphology, from adenomas to carcinomas [12,20]. Genomic instability is considered an essential component of this transformation process [14]. ...
Article
Full-text available
Colorectal cancer (CRC) is the third most common cancer type and the second cause of death worldwide. %e advancement in understanding molecular pathways involved in CRC has led to new classifications based on the molecular characteristics of each tumor and also improved CRC management through the integration of targeted therapy into clinical practice. In this review, we will present the main molecular pathways involved in CRC carcinogenesis, the molecular classifications. %e anti-VEGF and anti-EGFR therapies currently used in CRC treatment and those under clinical investigation will also be outlined, as well as the mechanisms of primary and acquired resistance to anti-EGFR monoclonal antibodies (cetuximab and panitumumab). Targeted therapy has led to great improvement in the treatment of metastatic CRC. However, there has been variability in CRC treatment outcomes due to molecular heterogeneity in colorectal tumors, which underscores the need for identifying prognostic and predictive biomarkers for CRC-targeted drugs.
... Two primary mechanisms of genomic instability have been identified in the progression of sporadic CRC. The first, called chromosomal instability, arises from a cascade of genetic changes that involve the activation of oncogenes, such as K-ras, and the inactivation of TSGs, including p53, DCC/Smad4, and APC [58][59][60].The second mechanism, referred to as microsatellite instability (MSI), originates from the inactivation of DNA mismatch repair (MMR) genes, such as MLH1 and/or MSH2, mostly via promoter hypermethylation (MLH1ph). This leads to mutations in genes containing coding microsatellites, such as transforming growth factor receptor II (TGF-RII) and BAX genes [38]. ...
Article
Full-text available
Prime editing (PE), a recent progression in CRISPR-based technologies, holds promise for precise genome editing without the risks associated with double-strand breaks. It can introduce a wide range of changes, including single-nucleotide variants, insertions, and small deletions. Despite these advancements, there is a need for further optimization to overcome certain limitations to increase efficiency. One such approach to enhance PE efficiency involves the inhibition of the DNA mismatch repair (MMR) system, specifically MLH1. The rationale behind this approach lies in the MMR system’s role in correcting mismatched nucleotides during DNA replication. Inhibiting this repair pathway creates a window of opportunity for the PE machinery to incorporate the desired edits before permanent DNA repair actions. However, as the MMR system plays a crucial role in various cellular processes, it is important to consider the potential risks associated with manipulating this system. The new versions of PE with enhanced efficiency while blocking MLH1 are called PE4 and PE5. Here, we explore the potential risks associated with manipulating the MMR system. We pay special attention to the possible implications for human health, particularly the development of cancer.
... Patients with cancer suffer a long intrusive procedure to eradicate CRC [10]. Therefore, a novel diagnosis procedure needs to enhance its specificity and effectiveness [11]. For several decades now, nanotechnology has emerged as a promising technology in various fields such as biomedical sciences, energy production, nanoelectronics, and consumer products [12][13][14][15][16][17]. ...
Article
Full-text available
Colorectal cancer (CRC) is the third cancer among the known causes of cancer that impact people. Although CRC drug options are imperfect, primary detection of CRC can play a key role in treating the disease and reducing mortality. Cancer tissues show many molecular markers that can be used as a new way to advance therapeutic methods. Nanotechnology includes a wide range of nanomaterials with high diagnostic and therapeutic power. Several nanomaterials and nanoformulations can be used to treat cancer, especially CRC. In this review, we discuss recent insights into nanotechnology in colorectal cancer.
... Colorectal cancer (CRC) is one of the most prevalent cancers in men followed by lung and prostate cancer and in women after breast cancer (Jemal et al. 2011). A multistage genetic model of cancer development encompassing genetic variations of the APC gene, Kras, PI3k, and Wnt/-catenin, as well as bridge between these mechanisms, had a significant role in cell cycle advancement deregulation, cell death prevention, initiation of genomic instability, and improved intrusiveness and metastasis (Moran et al. 2010;Wu et al. 2013). Reduced apoptosis in the colon epithelium is linked to a higher probability of colorectal cancer. ...
Article
Full-text available
The rhizomes of ginger have been in use in many forms of traditional and alternative medicines. Besides being employed as condiment and flavoring agent, it is used in the treatment of nausea, osteoarthritis, muscle pain, menstrual pain, chronic indigestion, Alzheimer’s disease, and cancer. Ginger rhizome contains volatile oils, phenolic compounds and resins, and characterization studies showed that [6]-gingerol, [6]-shogaol, and [6]-paradol are reported to be the pharmacologically active components. Gingerol is a major chemical constituent found as volatile oil in the rhizomes of ginger. It has several medicinal benefits and used for the treatment of rheumatoid arthritis, nausea, cancer, and diabetes. Many studies have been carried out in various parts of the world to isolate and standardize gingerol for their use as a complementary medicine. The present review summarizes wide range of research studies on gingerol and its pharmacological roles in various metabolic diseases. Graphical Abstract
... Les causes et les facteurs conduisant à un CCR sont soit génétiques, soit environnementaux ou les deux à la fois [2]. Le CCR est divisé en deux formes : la forme sporadique et la forme héréditaire, cette dernière représente entre 15 et 20 % de formes détectées [3]. ...
Thesis
Le cancer colorectal (CCR) est le troisième cancer en terme d’incidence et la deuxième cause de décès par cancer dans le monde. Au sein des tumeurs solides, les cellules souches cancéreuses (CSCs) représentent une sous-population rare de cellules possédant des propriétés d’auto-renouvellement, de différenciation et de tumorigenèse. Elles sont caractérisées par leurs résistances à la radiothérapie et à la chimiothérapie. Par conséquent, il est crucial de développer de nouvelles approches thérapeutiques ciblant spécifiquement les CSC afin de prévenir la résistance aux traitements chez les patients atteints de cancer du côlon. En outre, il a été mis en évidence qu’une grande majorité des protéines impliquées dans la mise en place du phénotype malin des CSCs sont produites sous forme de précurseurs qui nécessitent un clivage protéolytique pour être actives. Ce processus de maturation est contrôlé par des enzymes appelées proprotéines convertases (PCs).Précédemment, il a été montré que le niveau d’expression des PCs est fortement altéré dans le CCR et que ces PCs sont responsables de la maturation de plusieurs protéines impliquées dans la prolifération, l’angiogenèse et la dissémination métastatique. Cependant, le rôle des proprotéines convertases dans le phénotype malin de la population spécifique des CSCs du côlon n’avait pas encore été étudié jusqu’à présent.Durant mon projet de thèse, nous avons mis en évidence que le niveau d’expression des PCs (Furine, PACE4, PC5 et PC7) est fortement augmenté dans différentes lignées de CSCs de côlon et que l’inhibition de leurs activités réduit la prolifération, l’invasion et la survie de ces CSCs. Une analyse transcriptomique de CSCs traitées par un inhibiteur général des PCs (le décanoyle-RVKR-Chlorométhylekétone) montre une augmentation de l'expression de neuf métallothionéines, ces dernières étant connues pour être des suppresseurs de tumeur. De plus, l’inhibition spécifique de la Furine est associée à une diminution de l’expression des marqueurs de cellules souches LGR5 et NANOG et à l’altération de l’expression de plusieurs régulateurs du calcium intracellulaire. Nous avons également montré que ces modifications d’expression sont dépendantes de la mutation KRAS ou BRAF dans des lignées cancéreuses du côlon.Ces résultats mettent en évidence le rôle important des convertases dans le phénotype malin des CSCs et suggèrent que le ciblage de l’activité des PCs pourrait représenter une nouvelle stratégie thérapeutique dans le traitement du CCR, en particulier ceux possédant une mutation KRAS ou BRAF.
... q25.3 and aids in the occurrence and development of tumors by encouraging cell prolif¬eration, stopping cell apoptosis, inducing angiogenesis and suppressing immune reactions (Yu et al., 2016;Chen et al., 2015). The best-known tumor suppressor gene P53, it is located on chromosome 17p13.1 (Morán et al., 2010). In reaction to DNA damage, this gene codes for P53 protein which induces cell-cycle arrest in the G1 phase and promotes DNA repair genes. ...
Article
Full-text available
Background: Cancer is a significant health problem around the world and one of the leading causes of human death. The need for novel, selective and non-toxic anti-cancer agents is still urging. Aim of the work: to investigate the anti-proliferative and pro-apoptotic effects of the synthesized ciprofloxacin 3,4,5 tri-methoxy chalcone hybrid (CCH) on the HepG2 hepatocellular carcinoma and MCF7 breast carcinoma cell lines. Materials and methods: HepG2 and MCF7cell lines were treated with CCH. Cell viability and cell cycle analysis were performed. Protein and mRNA expression levels of P53, COX-2 and TNF-α were analyzed by western blotting and RT-PCR respectively. Results: CCH caused concentration and time-dependent reduction in the viability of human HepG2 and MCF7 cells, pre-G1 apoptosis and cell cycle arrest at G2/M stage, significantly higher P53 and TNF-α mRNA and protein expression levels but significantly lower COX2 mRNA and protein expression levels. Conclusion: CCH showed obvious anti-proliferative and apoptosis-inducing activities in both cell lines.
... Faulty repair of replication errors is mainly caused by a methylation anomaly of the MLH1 replication error repair gene, being a sporadic process without the involvement of heredity. Another mechanism may be a mutation of a hereditary nature, if the error-repair genes (MLH1, MSH2, MSH6) produce a genetic predisposition to CRC [118]. ...
Article
Full-text available
In CRC, screening compliance is decreased due to the experienced discomfort associated with colonoscopy, although this method is the gold standard in terms of sensitivity and specificity. Promoter DNA methylation (hypomethylation or hypermethylation) has been linked to all CRC stages. Study objectives: to systematically review the current knowledge on approved biomarkers, reveal new potential ones, and inspect tactics that can improve performance. This research was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines; the risk of bias was evaluated using the revised Quality Assessment of Diagnostic Accuracy Studies criteria (QUADAS-2). The Web of Science® Core Collection, MEDLINE® and Scopus® databases were searched for original articles published in peer-reviewed journals with the specific keywords “colorectal cancer”, “early detection”, “early-stage colorectal cancer”, “epigenetics”, “biomarkers”, “DNA methylation biomarkers”, “stool or blood or tissue or biopsy”, “NDRG4”, “BMP3”, “SEPT9”, and “SDC2”. Based on eligibility criteria, 74 articles were accepted for analysis. mSDC2 and mSEPT9 were frequently assessed in studies, alone or together as part of the ColoDefense panel test—the latter with the greatest performance. mBMP3 may not be an appropriate marker for detecting CRC. A panel of five methylated binding sites of the CTCF gene holds the promise for early-stage specific detection of CRC. CRC screening compliance and accuracy can be enhanced by employing a stool mt-DNA methylation test.
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.
Article
Full-text available
Our aim was to reveal the significance of tumor‐suppressor genes and genomic instability in 99 Hong Kong Chinese colorectal carcinoma (CRC) patients by PCR‐LOH analysis and PCR‐PTT assay. The frequencies of allelic loss of Apc, Mcc and Dcc and of APC truncation were 31.3% (15/48), 11.6% (5/43), 44.4% (20/45) and 46/93 (49.5%), respectively. The frequency of Apc LOH was similar to, the Mcc LOH was lower than, and the Dcc LOH was higher than that reported for Caucasians and Japanese. In Hong Kong CRC patients, the replication error‐positive (RER⁺) phenotype occurred with a frequency of 10% (10/99), which was similar to other results using microsatellite markers where RER⁺ frequencies ranged from 11% to 28%. The rates of genetic alteration in RER⁺ tumors were lower in tumors harboring p53, Mcc and Dcc alterations; similar in Apc; and higher in Ki‐ras tumors compared with RER– tumors, though these differences did not achieve statistical significance. None of the biomarkers examined were predictive of survival independently, but strong trends confirming earlier observations of associations between RER⁺ phenotypes with proximal tumor location and poorly differentiated tumor status were noted. The RER⁺ phenotype was correlated significantly to the less aggressive Duke's stage B and improved prognosis. Additionally, tumors with RER⁺ phenotypes were positively correlated with young age and sex. Our results support the observation that a subset of younger male CRC patients in Hong Kong may develop CRC via the RER pathway and show differences in RER status and sex. A significantly higher percentage of older Hong Kong Chinese CRC patients had APC truncations. Int. J. Cancer (Pred. Oncol.) 84:404–409, 1999. © 1999 Wiley‐Liss, Inc.
Article
Full-text available
You may have seen the bumper sticker "Eve was framed." Thousands of years of being blamed for original sin and still many wonder, where's the evidence? Today, the tumor suppressor adenomatous polyposis coli (APC) may have the same complaint about accusations of a different type of CIN, chromosome instability. A series of recent papers, including three in this journal, propose that loss of APC function plays an important role in the CIN seen in many colon cancer cells. However, a closer look reveals a complex story that raises more questions than answers.
Article
Full-text available
A subset of hereditary and sporadic colorectal carcinomas is defined by microsatellite instability (MSI), but the spectra of gene mutations have not been characterized extensively. Thirty-nine hereditary nonpolyposis colorectal cancer syndrome carcinomas (HNPCCa) and 57 sporadic right-sided colonic carcinomas (SRSCCa) were evaluated. Of HNPCCa, 95% (37/39) were MSI-positive as contrasted with 31% (18/57) of SRSCCa (P < 0.000001), but instability tended to be more widespread in SRSCCa (P = 0.08). Absence of nuclear hMSH2 mismatch repair gene product by immunohistochemistry was associated with germline hMSH2 mutation (P = 0.0007). The prevalence of K-ras proto-oncogene mutations was similar in HNPCCa and SRSCCa (30% (11/37) and 30% (16/54)), but no HNPCCa from patients with germline hMSH2 mutation had codon 13 mutation (P = 0.02), and two other HNPCCa had multiple K-ras mutations attributable to subclones. 18q allelic deletion and p53 gene product overexpression were inversely related to MSI (P = 0.0004 and P = 0.0001, respectively). Frameshift mutation of the transforming growth factor β type II receptor gene was frequent in all MSI-positive cancers (85%, 46/54), but mutation of the E2F-4 transcription factor gene was more common in HNPCCa of patients with germline hMSH2 mutation than in those with germline hMLH1 mutation (100% (8/8) versus 40% (2/5), P = 0.04), and mutation of the Bax proapoptotic gene was more frequent in HNPCCa than in MSI-positive SRSCCa (55% (17/31) versus 13% (2/15), P = 0.01). The most common combination of mutations occurred in only 23% (8/35) of evaluable MSI-positive cancers. Our findings suggest that the accumulation of specific genetic alterations in MSI-positive colorectal cancers is markedly heterogeneous, because the occurrence of some mutations (eg, ras, E2F-4, and Bax genes), but not others (eg, transforming growth factor β type II receptor gene), depends on the underlying basis of the mismatch repair deficiency. This genetic heterogeneity may contribute to the heterogeneous clinical and pathological features of MSI-positive cancers.
Article
Full-text available
The p53 tumor suppressor continues to hold distinction as the most frequently mutated gene in human cancer. The ability of p53 to induce programmed cell death, or apoptosis, of cells exposed to environmental or oncogenic stress constitutes a major pathway whereby p53 exerts its tumor suppressor function. In the past decade, we have discovered that p53 is not alone in its mission to destroy damaged or aberrantly proliferating cells: it has two homologs, p63 and p73, that in various cellular contexts and stresses contribute to this process. In this review, the mechanisms whereby p53, and in some cases p63 and p73, induce apoptosis are discussed. Other reviews have focused more extensively on the contribution of individual p53-regulated genes to apoptosis induction by this protein, whereas in this review, we focus more on those factors that mediate the decision between growth arrest and apoptosis by p53, p63 and p73, and on the post-translational modifications and protein-protein interactions that influence this decision.
Article
BACKGROUND AND AIMS In this study, we prospectively examined the clinical significance of the microsatellite instability (MSI) phenotype in sporadic colorectal cancer, and investigated methods for effective identification of these tumours in routine pathology practice. METHODS DNA was extracted from 310 tumours collected from 302 consecutive individuals undergoing curative surgery for sporadic colorectal cancer. Microsatellite status was determined by polymerase chain reaction amplification using standard markers, while immunostaining was used to examine expression of MLH1, MSH2, and p53. RESULTS Eleven per cent of tumours showed high level instability (MSI-H), 6.8% had low level instability (MSI-L), and the remainder were stable. MSI-H tumours were significantly more likely to be of high histopathological grade, have a mucinous phenotype, and to harbour increased numbers of intraepithelial lymphocytes. They were also more likely to be right sided, occur in women, and be associated with improved overall survival. In total, 25 (8%) tumours showed loss of staining for MLH1 and a further three tumours showed absence of staining for MSH2. The positive and negative predictive value of immunohistochemistry in the detection of MSI-H tumours was greater than 95%. CONCLUSIONS We conclude that the MSI-H phenotype constitutes a pathologically and clinically distinct subtype of sporadic colorectal cancer. Immunohistochemical staining for MLH1 and MSH2 represents an inexpensive and accurate means of identifying such tumours.
Article
BACKGROUND Patients with metachronous multiple colorectal carcinomas have been reported to have a higher frequency of a family history of colorectal carcinoma, associated colorectal adenomas, and extracolonic malignancies. These clinicopathologic factors also are considered to be related to the development of metachronous multiple colorectal carcinomas after surgery for colorectal carcinoma. In this article, the authors investigated whether genetic markers such as microsatellite instability (MSI) were helpful in predicting the development of metachronous multiple colorectal carcinomas.METHODS Between 1990–1997, 312 colorectal carcinoma patients underwent yearly surveillance colonoscopy after surgery. Among these patients, there were 19 with nonfamilial colorectal carcinoma in whom metachronous multiple colorectal carcinomas were diagnosed during the yearly surveillance colonoscopy. A control group was comprised of 28 patients who did not demonstrate either synchronous or metachronous carcinomas over a period of ≥5 years. Six microsatellite markers (D2S123, D3S1029, D3S1611, TP53, Mfd26, and Mfd36) were used to determine MSI by polymerase chain reaction.RESULTSThe frequency of MSI positive cases was significantly higher in patients with sporadic metachronous multiple colorectal carcinomas than in those with a single carcinoma (17/19 [89%] vs. 4/28 [14%]; P < 0.0001). In tumors occurring in the distal colon and rectum, the percentage of MSI positive carcinomas was significantly higher in the patients with metachronous multiple carcinomas than in those with a single carcinoma (13/15 [87%] vs. 0/19 [0%]; P < 0.0001). No such difference was observed in the proximal colon.CONCLUSIONS Based on the findings of the current study, the analysis of MSI in sporadic carcinomas of the distal colon and rectum may be helpful in predicting the development of metachronous multiple colorectal carcinomas. Cancer 1999;85:1917–24. © 1999 American Cancer Society.
Article
Colorectal cancers may demonstrate chromosomal instability (CSI) or microsatellite instability (MSI-H). A third group of microsatellite and chromosome stable (MACS) colorectal cancer has been described more recently. Patients with MSI-H colorectal cancers demonstrate improved outcome and a pronounced inflammatory infiltrate. Enhanced host immune response and increased immunogenicity might explain these observations. This study aims to further characterize colorectal cancer immunogenicity. Microsatellite stability status was determined in resected tumour samples. Microsatellite stable (MSS) tumour samples were stratified by DNA ploidy status, as determined by flow cytometry into aneuploid MSS (CSI) and diploid MSS (MACS) cancers. Lymphocyte proliferation, quantified by bromodeoxyuridine incorporation assays assessed tumour protein immunogenicity and ELISA assays quantified inflammatory cytokine release. Kaplan-Meier survival curves and multivariate analyses were used to determine prognostic value. Patients with MSI-H colorectal cancer had improved outcome but those with MACS cancers undergoing curative surgery had significantly poorer disease-free survival (P = 0.002). The MACS phenotype was an independent predictor of poor outcome (HR = 2.44, 1.33-4.47, P = 0.004). Lymphocyte proliferation assays confirmed enhanced immunogenicity of MSI-H proteins and reduced immunogenicity of MACS proteins (P < 0.0001). In vitro levels of IFN-gamma (P = 0.004) and IL-18 (P < 0.0001) mirrored these differences in lymphocyte activity. Stratification of colorectal cancer by MSI and ploidy status may have prognostic value in patients undergoing curative surgery. MSI-H cancers display enhanced immunogenic properties but the immune response to MACS cancers appears to be absent and this may contribute to their poor prognosis.
Article
Embryonic development of multicellular organisms is an incredibly complex process that relies heavily on evolutionarily conserved signalling pathways to provide crucial cell-cell communication. Typically, secreted signalling proteins such as Wnts, BMPs, and Hedgehogs released by one cell population will trigger concentration-dependent responses in other cells located some distance away. In adults, the same signalling pathways orchestrate tissue renewal in organs such as the intestine and skin, and direct tissue regeneration in many organs following injury. Strict regulation of these signalling pathways is critical, with insufficient or excess activity having catastrophic consequences including severe developmental defects or, later in life, cancer. This chapter deals with the beta-catenin-dependent branch of Wnt signalling (also referred to the canonical pathway).