SlideShare uma empresa Scribd logo
1 de 28
Skip Navigation
Oxford Journals
      Contact Us
      My Basket
      My Account

   HUMAN MOLECULAR GENETICS
        About This Journal
        Contact This Journal
           Subscriptions
            View Current Issue (Volume 21 Issue 2 January 15, 2012)
             Archive
               Search

                                      Institution: Benghazi University
                                        Sign In as Personal Subscriber

               Oxford Journals

               Life Sciences & Medicine

               Human Molecular Genetics

               Volume13, Issuesuppl 2

               Pp. R177-R185.

                  o    Zoe Kemp,
                  o    Cristina Thirlwell,
                  o    Oliver Sieber,
                  o    Andrew Silver,
                  o    and Ian Tomlinson

               An update on the genetics of colorectal cancer Hum. Mol. Genet.
               (2004) 13(suppl 2): R177-R185 doi:10.1093/hmg/ddh247




                      1. Hum. Mol. Genet. (2004) 13 (suppl 2): R177-R185. doi:
                10.1093/hmg/ddh247       An update on the genetics
of colorectal cancer
Next Section
Abstract
Colorectal carcinoma (CRC) remains a frequent cause of cancer-associated mortality in
the UK and still has a relatively poor outcome. Single gene defects account for up to 2–6%
of cases, but twin studies suggest a hereditary component in 35%. CRC represents a
paradigm for cancer genetics. Almost all the major-gene influences on CRC have been
identified, and the identification of the remaining susceptibility alleles is proving
troublesome. Only a few low-penetrance alleles, such as methylene tetrahydrofolate
reductase C677T, appear convincingly to be associated with CRC risk. To identify the
remaining CRC genes, parallel approaches, including strategies based on linkage and
association and complementary analyses such as searches for modifier genes, must be
employed. To gain sufficient evidence to prove that a gene is involved in CRC
predisposition, it is probably necessary for multiple, adequately-powered studies to
demonstrate an association with the disease, especially if the allelic variants have only a
small differential effect on risk. It may also be possible to show how genes interact with
each other and the environment, although this will be even more difficult. Accurate
quantitation of the allele-specific risks in different populations will be necessary, but
problematic, especially if those risks combine in a fashion which is not of a
straightforward additive or multiplicative type. Without any good prior evidence of the
nature of the remaining genetic influence on CRC, the possibility remains that this is a
truly polygenic trait or that multiple, rare variants contribute to the increased risk; in
these cases, identification of the genes involved will be very difficult. Despite these
potential problems, the effectiveness of preventive measures for CRC, especially in high-
risk individuals, means that the search for new predisposition genes is justified.

Previous SectionNext Section
Colorectal cancer (CRC) is the third most common cause of cancer-related death in the
western world. The annual incidence in the United Kingdom is 35 000, and worldwide it
has been estimated to be at least half a million. There exists a number of syndromes with
Mendelian dominant inheritance in which there is a primary predisposition to benign or
malignant tumours of the large bowel (Table 1). Familial adenomatous polyposis (FAP)
patients inherit a mutated copy of the adenomatous polyposis gene ( APC), whereas
hereditary non-polyposis colon cancer (HNPCC) is caused by inheritance of defective DNA
mismatch repair genes (MLH1, MSH2, PMS2 and MSH6). Germline mutations of the
LKB1/STK11 gene have been shown to cause the Peutz–Jeghers syndrome (PJS), and
mutation of SMAD4 or ALK3 underly juvenile polyposis (JPS). In contrast, the MYH-
associated polyposis (MAP) syndrome has autosomal recessive inheritance and results
from bi-allelic mutations in the MYH gene.

                                        View this table:


                                             In this window
                                            In a new window


Table 1.
Genes responsible for known polyposis and CRC syndromes

Taken together, these syndromes account only for ∼2–6% of CRC cases. The great
majority of CRCs do not have a recognizable inherited cause, but a number of studies
have suggested a role for genetic factors in predisposition to a substantial minority of
colorectal tumours. The relatives of patients with ‘sporadic’ CRC are themselves at
increased risk of the disease and segregation analysis has suggested dominant
inheritance of the uncharacterized susceptibility genes (1–3). The relative risk in siblings
of affected individuals is 2–3-fold increased for both colorectal adenomas and CRC, the
risk being higher in relatives of patients diagnosed young, and in those with two or more
affected family members. In addition, an extensive analysis of twins has suggested that
35% of CRCs arise through inherited factors (4). Although some high and moderate
penetrance genes are probably undiscovered, inherited predisposition to CRC in humans
is more likely to be the result of low-penetrance alleles at several or many genetic loci. In
support of this contention, experimental data from rodent models have demonstrated
that numerous genes with relatively small effects control susceptibility to cancer
including CRC (reviewed in 5). Furthermore, hereditary factors may influence the severity
of the disease in the Mendelian CRC syndromes, as exemplified by the number of
colorectal polyps presenting within FAP families or in individuals with identical germline
APC mutations (6).

There are two factors that, in combination, make the search for additional CRC
predisposition genes so important. First, identification and removal of colorectal
adenomas (and possibly other types of bowel polyp) almost certainly greatly reduce
cancer incidence; identification of cancers before they present clinically may have similar
benefits. Second, regular, frequent and effective screening for colorectal tumours is not
currently available at the population level. The identification of controlling genetic factors
therefore allows individual cancer risk assessment and the targeting of screening to
those at highest risk; there may also be spin-offs in terms of effective chemoprevention
and the development of new therapies. The challenge, therefore, is to identify all the
relevant genes, and to specify their contribution in controlling CRC predisposition and
severity in human populations. A major part of this challenge is to choose the most
appropriate methods to locate and identify novel susceptibility genes.

In this short review, we shall detail some of the recent developments in the study of
inherited susceptibility to CRC. Progress has been made in the understanding of the
Mendelian syndromes and towards the identification of the small number of remaining
high-penetrance CRC genes. We shall also present recent evidence for low-penetrance
CRC alleles and for modifier genes of the Mendelian syndromes' phenotypes. We
complete our review by outlining future strategies for use in the continued search for
genes predisposing to CRC.

Previous SectionNext Section
PROGRESS IN UNDERSTANDING THE HEREDITARY
COLORECTAL TUMOUR SYNDROMES
MYH-associated polyposis
MYH is a DNA glycosylase involved in the response to oxidative damage of DNA by base
excision repair (BER). MYH was identified as a recessive CRC and adenoma predisposition
gene by Al-Tassan et al. (7), who found an unusual somatic APC mutation spectrum
(mostly G→T changes) in the polyps of a family that they were investigating for the
pathogenic effects of the APC missense variant E1317Q. The MAP phenotype generally
resembles attenuated (<100 adenomas) or mild, classical (100–1000 adenomas) FAP (8),
although MAP tumours seem to be confined to the gastrointestinal tract. MAP individuals
with early-onset CRC but no detected colorectal adenomas have been reported (9),
although small adenomas can be missed using colonoscopy without dye-spray. MAP
accounts for about 1% of all CRCs and one-third of patients with 15–100 colorectal
adenomas (8,10). MYH screening should be considered in patients with multiple
colorectal adenomas, although APC remains as a much stronger candidate in families
with vertical transmission of disease. Testing MYH in patients with early-onset CRC and
no adenomas would be justified if further data support those of Wang et al. (9). Mutation
testing should be tailored to the ethnic group, with mutations Y165C and G382D more
common in Caucasians, Y90X in Pakistanis, E466X in Indians and nt1395delGGA in those
of Mediterranean origin (10–13). The site-specific nature of MAP tumours remains
puzzling. BER deficiency does not specifically target APC and many MAP tumours harbour
specific G→T changes in K-ras (14). The few MAP cancers studied so far are
microsatellite-stable and near-diploid (14).

AXIN2
Apart from APC, studies have generally failed to find germline mutations in other
components of the Wnt pathway (such as beta-catenin, APC2, axin, conductin (axin2) and
glycogen synthase kinase 3-beta) in patients with multiple or early-onset colorectal
tumours (15). Recently, however, a Finnish family with dominantly-transmitted
oligodontia and colorectal polyps was shown to carry a germline R656X mutation in
AXIN2 (16). The colorectal phenotype in the family was highly variable, ranging from 68
adenomas through 10 hyperplastic polyps to no disease. Loss of the wild-type AXIN2
allele was not tested in the polyps.

Peutz–Jeghers syndrome
Germline mutations of the serine/threonine kinase gene LKB1(STK11) cause PJS. PJS
patients develop harmatomatous polyps of the gastrointestinal tract, and characteristic
freckling, and are at increased risk of colorectal and other cancers. LKB1 has been
reported to function in a variety of pathways, including those involving mTOR/AMP-
activated protein kinase, Wnt, COX2, NF-kappaB, VEGF, cell polarity and p53 (17–25). It
remains to be seen whether mutations in such genes play a role in PJS. From a strictly
genetic perspective, there is ongoing controversy as to the existence of a second PJS
locus, primarily because LKB1 mutations cannot be found in 30–80% of PJS patients. PJS
kindreds unlinked to LKB1 (26) and some linked to 19q13 (27) have been reported, but
no second locus has been identified. In this regard, it should be noted that, although PJS
polyps are probably pathognomic, several conditions have pigmented lesions that may
mimic PJS (28).

Hereditary non-polyposis CRC
Hereditary non-polyposis colon cancer (HNPCC) is an autosomal dominant disorder
characterized by early-onset CRC (often right-sided), few adenomas and specific extra-
colonic cancers, including those of the ovary, stomach, small bowel and uroepithelial
tract. HNPCC accounts for 1–5% of CRCs and results from germline mutations in MLH1
and MSH2 in about 90% of cases and more rarely in PMS2 and MSH6 (remaining 10% of
cases). HNPCC cancers are characterized by microsatellite instability (MSI). The MMR
genes are not mutated in sporadic CRCs, although MLH1 is silenced by promoter
methylation in 10–15% of these tumours, most of which are right-sided and tend to occur
in older females. In the light of these data, Suter et al. (29) hypothesized that methylation
of the MLH1 promoter could occur in the germline and act as an alternative to mutation
in predisposing to HNPCC. They found evidence of such a MLH1 ‘epimutation’ in two
patients with multiple, MSI+ primary tumours which showed mismatch repair deficiency.
The epimutation occurred as a mosaic and was also present in spermatozoa of one of the
individuals, indicating a germline defect and the potential for transmission to offspring. It
was not clear whether an uncharacterized, primary genetic defect had led to the
epimutation. Other workers have focussed on the role of additional MMR genes in the
HNPCC families, with evidence that MLH3 (30–32) and EXO1 (33–36) might be involved in
a small number of cases, although data are inconsistent. There is also accumulating
evidence to show that the genetic pathways of sporadic, MSI+ CRCs and HNPCC CRCs are
non-identical. Johnson et al. (37), for example, showed that beta-catenin mutations were
specific to MSI+ carcinomas from HNPCC, but were rare in the HNPCC adenomas,
suggesting that Wnt activation was not always an early event in HNPCC tumorigenesis.
Two further issues the functions of MMR proteins outside simple DNA repair (38) and
whether or not MMR loss initiates tumorigenesis in HNPCC, remain under close scrutiny.

Familial adenomatous polyposis and APC modifier genes
FAP is caused by germline APC mutations. Affected individuals develop hundreds to
thousands of adenomatous colorectal polyps during the second and third decades,
progressing to cancer in nearly 100% of cases without treatment. Specific extra-colonic
features exist (39). We shall focus on three aspects of recent FAP research. First, it is
clear, in both FAP and sporadic CRCs, that APC is not a simple tumour suppressor gene:
the ‘two hits’ are strongly associated in terms of their location within the gene (39–42).
This association probably provides an optimal level of Wnt signalling for tumour growth,
mediated through beta-catenin levels. The mutation spectrum, and presumably optimal
level of Wnt signalling, varies among tissues (40,43). The ‘first hit–second hit’ association
at APC has also provided a new mechanism for determining disease severity (40), given
that certain ‘second hits’ such as allelic loss probably occur spontaneously at a relatively
high frequency and are only selected in certain FAP patients (with mutations close to
codon 1300), who consequently have severe disease. Second, clues have emerged as to
the cause of attenuated disease (AFAP, AAPC) in individuals with germline APC mutations
in exons 1–4, 9 and the latter half of exon 15, owing to the discovery of ‘third hits’ in the
tumours of some of these patients (44). In order to explain the paradox that a
requirement for ‘third hits’ should in theory cause AFAP patients to have no more
tumours than the general population, Su et al. (45) suggested that a broader spectrum of
somatic mutations was selected in AFAP individuals than in those with no germline APC
mutation (although their model was not placed in the context of the ‘first hit–second hit’
associations described earlier). Third, the site of the germline APC mutations cannot
explain all the phenotypic variation which is seen in FAP and AFAP. It is likely that
modifier genes distinct from APC influence features such as the number of tumours in
each region of the gastrointestinal tract (46), the development of desmoids (47) and
progression to cancer. In ApcMin mice, a modifier gene, Mom1/Pla2g2a, influences polyp
number (48–50). FAP modifier genes in humans are likely to be common polymorphisms,
and candidate gene approaches have been used to analyse these. One study has
suggested effects of polymorphisms in the N-acetyltransferases (NAT1 and NAT2) on
polyp numbers in FAP (51).

APC I1307K
The APC I1307K variant is present in about 6% of Ashkenazi Jews, but is much rarer in
those of other ethnic groups. I1307K creates an A8 tract that appears to be somatically
unstable, leading to frameshift mutations (52). Originally, I1307K was proposed to lead
to an AFAP-like phenotype, although extra-colonic disease does not appear to feature in
this group. Recent data have shown that APC I1307K only confers a small excess risk of
CRC (53), inconsistent prima facie with an AFAP-like phenotype. Moreover, only a
minority of colorectal tumours from I1307K carriers shows slippage of the A 8 tract. It may
be the case, therefore, that although the 1307K allele is somewhat unstable, this effect is
subtle in most carriers; only some individuals, perhaps with an unidentified, additional
germline factor, go on to develop an AFAP-like phenotype (54).

Previous SectionNext Section
RECENT DEVELOPMENTS THROUGH LINKAGE
ANALYSIS
Hereditary mixed polyposis syndrome
Hereditary mixed polyposis syndrome (HMPS) was described as a distinct syndrome of
colorectal adenomas, hyperplastic polyps, atypical ‘mixed’ polyps and carcinoma by
Whitelaw et al. (55) in a large Ashkenazi family. The extra-colonic features of FAP and the
typical features of PJS were absent. The phenotype had limited similarity to JPS. Linkage
of HMPS to chromosome 6q16–q21 was demonstrated by Thomas et al. (56). Tomlinson
et al. (57) showed linkage to chromosome 15q14–q15 (CRAC1 locus) in another
Ashkenazi family with multiple colorectal adenomas (including serrated lesions) and CRC.
The development of multiple adenomas in a HMPS individual without the disease-
associated haplotype led to the reassignment and stricter implementation of affection
status in that family in order to minimize the phenocopy rate. Subsequent linkage
analysis showed HMPS and CRAC1 to be one-and-the-same (58). Affected individuals in
these families and in several other Ashkenazi kindreds with a similar phenotype were
shown to share an ancestral haplotype between D15S1030 and D15S118. The gene itself
is, as yet, unidentified and its influence in other population groups, undetermined.

A CRC locus on chromosome 9q?
Wiesner et al. (59) undertook linkage analysis using the affected sibling pair method in
53 families with individuals affected at <65 years of age by CRC or adenomas (>1 cm or
displaying high-grade dysplasia). Six potential loci were identified with excess allele
sharing among concordantly affected sib-pairs (P<0.016). Of these, a region on
chromosome 9 gave a significant result when additionally examined for deficient allele
sharing among discordant sib pairs (P=0.014). The Haseman–Elston regression method
provided an overall probability of linkage of P=0.00045.

Previous SectionNext Section
EVIDENCE FOR LOW-PENETRANCE ALLELES
ASSOCIATED WITH AN INCREASED RISK OF CRC
Some high- or moderate-penetrance CRC predisposition genes remain to be detected
and their pursuit is of great importance. It is generally believed, nevertheless, that low-
penetrance variants, such as common alleles at SNPs, are responsible for much of the
uncharacterized inherited influence on CRC. Many different SNPs and other common
polymorphisms have been evaluated for their effects on CRC risk, and space precludes
detailed descriptions or analysis here. Case–control (association) analysis has been used
most frequently. Such studies have often been criticized, some of the reasons being the
following:

               sample sizes produce insufficient statistical power;

               cases and controls are inappropriately chosen;

               statistical thresholds are chosen with insufficient rigour, so that most
               reported associations are inevitably false (60);

               the ‘multiple testing’ problem and tendency to inappropriate sub-group
               analyses are endemic;
publication bias remains problematic, even though methods have been
              developed to assess this (61,62);

              studies rarely attempt to replicate their findings in the same population;

              findings which are not replicated in different populations may be
              inappropriately dismissed;

              the functional effects of variants have often not been clearly shown;

              some methods, such as PCR–RFLP analysis, are prone to systematic error
              unless control measures are carefully applied.

The recognition of these problems has arguably led to too pessimistic an attitude to
studies of low-penetrance variants as CRC susceptibility alleles. Most recent studies have
used larger, better defined populations of cases and controls, together with genotyping
methods with greater accuracy and throughput. Formal assessments of the roles of
several polymorphisms in CRC predisposition were made by Houlston and Tomlinson (63)
and by de Jong et al. (64). In summary, there is already good evidence to show that a
small number of polymorphisms is associated with a differential risk of CRC (Table 2).
These include methylene tetrahydrofolate reductase MTHFRC677T, H-ras VNTR rare
alleles and NAT2 alleles which cause a rapid acetylator phenotype. Other polymorphisms
have been tested (Table 2) and many are unproven, although in a small number of cases,
loci continue to be cited as being associated with CRC risk despite the evidence in their
favour being minimal.

                                        View this table:


                                             In this window
                                            In a new window


Table 2.
Summary of polymorphisms tested for association with differential CRC risk

Recent data have suggested that loss of imprinting (LOI) of the insulin-like growth factor
II (IGF2) in normal colonic mucosa and/or lymphocytes is indicative of increased CRC risk
(89–92). It has been suggested that IGF2 LOI is a genetically determined trait. It is not
clear whether the association between IGF2 LOI and CRC might be a direct effect of
increased IGF2 levels, or a marker of a general tendency to, for example,
hypomethylation or aberrant gene silencing.

Studies are increasingly testing risks associated with combinations of putative CRC
genes, or of genes and specific environments, although inherent problems of statistical
power mean that most of the reported associations of this type must currently be taken
as no more than suggestive. Even for the MTHFR polymorphisms, advice on dietary
modification so that folate intake is increased cannot yet be given reliably or with
confidence. A further question is whether or not a true CRC risk factor should affect the
risk of both rectal and colonic cancers, conditions that are usually treated as essentially
the same disease (albeit with some different molecular and clinical features) by
geneticists, but as different conditions by epidemiologists. Further studies are beginning
to test the influence of germline variation on factors other than disease susceptibility,
including the presenting features of CRCs, response to therapy and toxicity (including
pharmacogenetic analyses), and recurrence/survival.

In general, acceptance of a polymorphism as a CRC risk factor can only come through
repeated analysis by different studies and by consequent slow progress to a consensus
judgement. Accurate quantitation of the associated risk will be just as important as its
identification in the first place, and this will require multiple studies in individual
populations which may differ in genetic background, environment and availability of
screening and medical care. Quantitation of risks associated with variants at multiple
predisposition loci will be even more difficult, especially if risks do not combine in a
simple additive or multiplicative manner, yet if testing for low-penetrance alleles is to
come into routine clinical practice, such risks must be determined.

Without any good prior evidence of the nature of the remaining genetic influence on CRC,
the possibility remains that it is a truly polygenic trait in which risks associated with any
single polymorphism are so low that identification of the genes involved (and their
testing in clinical practice) will be very difficult. This is, perhaps, the worst case scenario.
An alternative is that multiple, rare variants may contribute to the increased familial risk
of CRC: in this case, identification of the variants will be possible through mutation
screening methods, although evaluation of their functional importance will be time-
consuming. The case of the sub-polymorphic variant APCE1317Q provides a case in point
(93–97); it currently seems likely that this variant, which was discovered by mutation
screening, has little functional effect, although ‘closing the door’ on such a variant is not
easy. Nevertheless, the modestly increased risk of breast cancer associated with the
CHEK2del1100C sub-polymorphic variant (98) shows that important alleles of this type
do exist. (Any association between CHEK2del1100C and CRC remains controversial.)
Should many of the remaining CRC alleles prove to be of the low-frequency/low-risk
type, genetic testing in the clinical setting may not be worthwhile. Despite these potential
problems, however, the effectiveness of preventive measures for CRC, especially in high-
risk individuals, means that the search for new predisposition genes is justified.

Previous SectionNext Section
FUTURE STRATEGIES FOR IDENTIFYING CRC
PREDISPOSITION GENES
The successful detection of predisposition genes for cancer has largely been limited to
high-penetrance genes such as APC, BRCA1/2 and CDNK2A. Although the search for
low-penetrance genes has been informative in some other complex diseases, for
example, APOE-4 in Alzheimer's disease, those underlying cancer remain largely elusive.
The characteristics of cancer create obstacles. The usual late onset of CRC means parents
may be deceased and the disease is often fatal, making sample collection difficult. In
addition, the requirement of random somatic mutations for the progression to carcinoma
may cloud the picture. However, some of these problems can be circumvented by the
selection of early-onset cases to increase the genetic component and to reduce the
environmental component, and by the study of benign, precursor lesions.

The question therefore arises of which methods to use to detect novel cancer genes. In
our view, given that there are undoubtedly still high and moderate penetrance CRC genes
yet to be discovered, linkage analysis should certainly not be abandoned. The latter
would require the use of non-parametric methodology in view of genetic heterogeneity
and the existence of phenocopies. However, even for moderate penetrance genes, the
numbers of multi-case families required to yield results are large. For example, under a
dominant model with a gene frequency of 0.1 and a risk ratio of 6, about 500 affected sib
pairs or 20 affected sib trios would be needed to give a LOD score of 3.3 (99).

The increasing attention being given to association studies to identify CRC genes is
therefore justified. For CRC, these have previously been limited to examining the
frequency of alleles at candidate loci in cases and controls. However, the advent of SNP
maps has created the potential for genome-wide association (100). The power of these
studies, especially for rare alleles can be increased by using cases with a family history of
CRC (101) and by selecting in other ways for ‘genetic’ disease (for example, multiple
tumours, cancers of more than one site or early age of presentation). The detection of a
dominant gene with a frequency of 0.1 and a relative risk of 4 would require 200 cases
and 200 controls in an association study, but over 1000 affected sib pairs in a traditional
linkage analysis (99).

A more controversial area in the optimal design of association studies revolves around
the question of how many SNPs to use and whether the approach should be haplotype-
map or sequence based. Public and private SNP discovery projects report the
identification of 1.4 and 2.1 million SNPs, respectively, although the total is estimated
(102) to be about 15 million in reality. Obviously only a small proportion of these could
currently be genotyped in any association study. A map-based approach depends on
linkage disequilibrium occurring between blocks of haplotypes within which only a few
common haplotypes are observed and there is little evidence of historical recombination.
This allows the genome to be represented by a carefully chosen group of haplotype tag
SNPs (htSNPs) (103). The number of SNPs required will increase with the genetic diversity
of a population because of shorter haplotype blocks. For example, the International
HapMap Project has recently decided to add 2.25 million SNPs to the original 600 000 for
each of three population maps, owing to the longer evolutionary history of the Yuroban
(Nigerian) population (88). This approach will mainly detect alleles with a frequency of
≥5%. Advocates of a sequence-based approach argue that SNPs should be chosen on the
basis of their probable contribution to the phenotype. The focus of this approach is
therefore on coding regions and SNPs resulting in amino acid changes and/or SNPs found
in evolutionarily conserved areas. This would require far fewer SNPs (50 000–100 000) for
a genome-wide screen and would potentially detect lower frequency alleles (1–20%).
Botstein and Risch (102) have suggested that, at least initially, a ‘sequence-based’
approach on a selected 5% of the genome might provide the power to detect some of the
CRC-associated variants.

For identifying sub-polymorphic variants associated with disease, a mutation screening
strategy based on candidate genes (and followed if possible by a suitable association
study) is probably the only practical method for the short term. It remains to be seen
whether or not new technologies such as re-sequencing microarrays prove cost-effective
for research and diagnosis here. The requirement for functional studies will be
particularly important for sub-polymorphic variants, especially those which lead to
conserved amino acid changes, or affect splice sites, promoters or control regions.

Although it is hoped that linkage, association and mutation screening studies will yield
answers, the concurrent use of more indirect methods is prudent and may contribute to
the fine mapping of an identified region of interest. As with SMAD4 and more recently,
MYH, the search for somatic mutations may also identify the site of germline mutations,
as their effects in the cell are functionally similar. Other approaches include the study of
cancer-associated phenotypes including normal variation (for example in hormone
levels). Inflammatory bowel disease, which we have not discussed in detail earlier, is
associated with an increase risk of CRC yet has a higher sibling relative risk (104), and
some of the predisposition genes might be the same for the two conditions. The search
for modifier genes is also potentially important, as a gene which influences the severity
of a Mendelian syndrome should also, in principle, act as a low-penetrance
predisposition gene.

Intensive, effective population-based screening for CRC and its precursor lesions is likely
in the foreseeable future to be expensive and/or to have a relatively low take-up rate.
Even if this screening for CRC becomes more acceptable, it will optimally be targeted to
those at highest risk. The identification of CRC risk genes will permit targeted screening,
and will open up possibilities for other preventive measures and, perhaps, therapies. It is
highly likely that at least some CRC genes can be identified and will provide useful
targets for routine genetic testing and preventive intervention.

Previous SectionNext Section
ACKNOWLEDGEMENTS
We are grateful to colleagues, particularly Richard Houlston and Walter Bodmer, for
discussion and help.

  1. Zoe Kemp1,
  2. Cristina Thirlwell1,
  3. Oliver Sieber1,
  4. Andrew Silver2 and
  5. Ian Tomlinson1,2,*
+ Author Affiliations

           1.  Molecular and Population Genetics Laboratory, London Research Institute,
                1


              Cancer Research UK, 44 Lincoln's Inn Fields, London WC2A 3PX, UK and
           2. 2Colorectal Cancer Unit, Cancer Research UK, St Mark's Hospital, Watford
              Road, Harrow HA1 3UJ, UK

  1.   To whom correspondence should be addressed. Tel: +44 1712692884; Fax: +44
       *


       1712692885; Email: i.tomlinson@cancer.org.uk


                                                                     Received July 23, 2004.
                                                                    Accepted July 27, 2004.


                Human Molecular Genetics, Vol. 13, Review Issue 2 © Oxford University
                Press 2004; all rights reserved


Previous Section


References
1. ↵
   Cannon-Albright, L.A., Skolnick, M.H., Bishop, D.T., Lee, R.G. and
   Burt, R.W. (1988) Common inheritance of susceptibility to colonic
   adenomatous polyps and associated colorectal cancers. N. Engl. J.
   Med., 319, 533–537.
   MedlineWeb of Science
2. Houlston, R.S., Collins, A., Slack, J. and Morton, N.E. (1992)
   Dominant genes for colorectal cancer are not rare. Ann. Hum. Genet.,
   56 (Pt 2), 99–103.
   MedlineWeb of Science
3. ↵
   Johns, L.E. and Houlston, R.S. (2001) A systematic review and meta-
   analysis of familial colorectal cancer risk. Am. J. Gasrtoenterol., 96,
   2992–3003.
4. ↵
   Lichtenstein, P., Holm, N.V., Verkasalo, P.K., Iliadou, A., Kaprio, J.,
   Koskenvuo, M., Pukkala, E., Skytthe, A. and Hemminki, K. (2000)
   Environmental and heritable factors in the causation of cancer—
   analyses of cohorts of twins from Sweden, Denmark, and Finland. N.
   Engl. J. Med., 343, 78–85.
   CrossRefMedlineWeb of Science
5. ↵
   Demant, P. (2003) Cancer susceptibility in the mouse: genetics,
   biology and implications for human cancer. Nat. Rev. Genet., 4, 721–
   734.
   MedlineWeb of Science
6. ↵
   Crabtree, M.D., Tomlinson, I.P., Hodgson, S.V., Neale, K., Phillips,
   R.K. and Houlston, R.S. (2002) Explaining variation in familial
   adenomatous polyposis: relationship between genotype and
   phenotype and evidence for modifier genes. Gut, 51, 420–423.
   Abstract/FREE Full Text
7. ↵
   Al-Tassan, N., Chmiel, N.H., Maynard, J., Fleming, N., Livingston,
   A.L., Williams, G.T., Hodges, A.K., Davies, D.R., David, S.S., Sampson,
   J.R. et al. (2002) Inherited variants of MYH associated with somatic
   G:C→T:A mutations in colorectal tumors. Nat. Genet., 30, 227–232.
   CrossRefMedlineWeb of Science
8. ↵
   Sieber, O.M., Lipton, L., Crabtree, M., Heinimann, K., Fidalgo, P.,
   Phillips, R.K., Bisgaard, M.L., Orntoft, T.F., Aaltonen, L.A., Hodgson,
   S.V. et al. (2003) Multiple colorectal adenomas, classic adenomatous
polyposis, and germ-line mutations in MYH. N. Engl. J. Med., 348,
   791–799.
   CrossRefMedlineWeb of Science
9. ↵
   Wang, L., Baudhuin, L.M., Boardman, L.A., Steenblock, K.J., Petersen,
   G.M., Halling, K.C., French, A.J., Johnson, R.A., Burgart, L.J., Rabe, K.
   et al. (2004) MYH mutations in patients with attenuated and classic
   polyposis and with young-onset colorectal cancer without polyps.
   Gastroenterology, 127, 9–16.
   CrossRefMedlineWeb of Science
10. ↵
   Halford, S.E., Rowan, A.J., Lipton, L., Sieber, O.M., Pack, K., Thomas,
   H.J., Hodgson, S.V., Bodmer, W.F. and Tomlinson, I.P. (2003)
   Germline mutations but not somatic changes at the MYH locus
   contribute to the pathogenesis of unselected colorectal cancers. Am.
   J. Pathol., 162, 1545–1548.
   MedlineWeb of Science
11. Jones, S., Emmerson, P., Maynard, J., Best, J.M., Jordan, S., Williams,
   G.T., Sampson, J.R. and Cheadle, J.P. (2002) Biallelic germline
   mutations in MYH predispose to multiple colorectal adenoma and
   somatic G:C→T:A mutations. Hum. Mol. Genet., 11, 2961–2967.
   Abstract/FREE Full Text
12. Enholm, S., Hienonen, T., Suomalainen, A., Lipton, L., Tomlinson, I.,
   Karja, V., Eskelinen, M., Mecklin, J.P., Karhu, A., Jarvinen, H.J. et al.
   (2003) Proportion and phenotype of MYH-associated colorectal
   neoplasia in a population-based series of Finnish colorectal cancer
   patients. Am. J. Pathol., 163, 827–832.
   MedlineWeb of Science
13. ↵
   Gismondi, V., Meta, M., Bonelli, L., Radice, P., Sala, P., Bertario, L.,
   Viel, A., Fornasarig, M., Arrigoni, A., Gentile, M. et al. (2004)
   Prevalence of the Y165C, G382D and 1395delGGA germline
   mutations of the MYH gene in Italian patients with adenomatous
   polyposis coli and colorectal adenomas. Int. J. Cancer, 109, 680–684.
   CrossRefMedlineWeb of Science
14. ↵
   Lipton, L., Halford, S.E., Johnson, V., Novelli, M.R., Jones, A.,
   Cummings, C., Barclay, E., Sieber, O., Sadat, A., Bisgaard, M.L. et al.
   (2003) Carcinogenesis in MYH-associated polyposis follows a distinct
   genetic pathway. Cancer Res., 63, 7595–7599.
   Abstract/FREE Full Text
15. ↵
Lipton, L., Sieber, O.M., Thomas, H.J., Hodgson, S.V., Tomlinson, I.P.
   and Woodford-Richens, K. (2003) Germline mutations in the TGF-
   beta and Wnt signalling pathways are a rare cause of the ‘multiple’
   adenoma phenotype. J. Med. Genet., 40, e35.
   FREE Full Text
16. ↵
   Lammi, L., Arte, S., Somer, M., Jarvinen, H., Lahermo, P., Thesleff, I.,
   Pirinen, S. and Nieminen, P. (2004) Mutations in AXIN2 cause familial
   tooth agenesis and predispose to colorectal cancer. Am. J. Hum.
   Genet., 74, 1043–1050.
   CrossRefMedlineWeb of Science
17. ↵
   De Leng, W.W., Westerman, A.M., Weterman, M.A., De Rooij, F.W.,
   Dekken Hv, H., De Goeij, A.F., Gruber, S.B., Wilson, J.H., Offerhaus,
   G.J., Giardiello, F.M. et al. (2003) Cyclooxygenase 2 expression and
   molecular alterations in Peutz–Jeghers hamartomas and carcinomas.
   Clin. Cancer Res., 9, 3065–3072.
   Abstract/FREE Full Text
18. Liu, W.K., Chien, C.Y., Chou, C.K. and Su, J.Y. (2003) An LKB1-
   interacting protein negatively regulates TNFalpha-induced NF-
   kappaB activation. J. Biomed. Sci., 10, 242–252.
   MedlineWeb of Science
19. Martin, S.G. and St Johnston, D. (2003) A role for Drosophila LKB1 in
   anterior–posterior axis formation and epithelial polarity. Nature, 421,
   379–384.
   CrossRefMedline
20. Ossipova, O., Bardeesy, N., DePinho, R.A. and Green, J.B. (2003) LKB1
   (XEEK1) regulates Wnt signalling in vertebrate development. Nat. Cell
   Biol., 5, 889–894.
   CrossRefMedlineWeb of Science
21. Baas, A.F., Smit, L. and Clevers, H. (2004) LKB1 tumor suppressor
   protein: PARtaker in cell polarity. Trends Cell. Biol., 14, 312–319.
   CrossRefMedlineWeb of Science
22. Baas, A.F., Kuipers, J., van der Wel, N.N., Batlle, E., Koerten, H.K.,
   Peters, P.J. and Clevers, H.C. (2004) Complete polarization of single
   intestinal epithelial cells upon activation of LKB1 by STRAD. Cell,
   116, 457–466.
   CrossRefMedlineWeb of Science
23. Lizcano, J.M., Goransson, O., Toth, R., Deak, M., Morrice, N.A.,
   Boudeau, J., Hawley, S.A., Udd, L., Makela, T.P., Hardie, D.G. et al.
   (2004) LKB1 is a master kinase that activates 13 kinases of the AMPK
   subfamily, including MARK/PAR-1. EMBO J., 23, 833–843.
   CrossRefMedlineWeb of Science
24. Shaw, R.J., Bardeesy, N., Manning, B.D., Lopez, L., Kosmatka, M.,
   DePinho, R.A. and Cantley, L.C. (2004) The LKB1 tumor suppressor
   negatively regulates mTOR signaling. Cancer Cell, 6, 91–99.
   CrossRefMedlineWeb of Science
25. ↵
   Spicer, J. and Ashworth, A. (2004) LKB1 kinase: master and
   commander of metabolism and polarity. Curr. Biol., 14, R383–R385.
   CrossRefMedlineWeb of Science
26. ↵
   Olschwang, S., Markie, D., Seal, S., Neale, K., Phillips, R., Cottrell, S.,
   Ellis, I., Hodgson, S., Zauber, P., Spigelman, A. et al. (1998) Peutz–
   Jeghers disease: most, but not all, families are compatible with
   linkage to 19p13.3. J. Med. Genet., 35, 42–44.
   Abstract/FREE Full Text
27. ↵
   Buchet-Poyau, K., Mehenni, H., Radhakrishna, U. and Antonarakis,
   S.E. (2002) Search for the second Peutz–Jeghers syndrome locus:
   exclusion of the STK13, PRKCG, KLK10, and PSCD2 genes on
   chromosome 19 and the STK11IP gene on chromosome 2.
   Cytogenet. Genome Res., 97, 171–178.
   CrossRefMedlineWeb of Science
28. ↵
   Lampe, A.K., Hampton, P.J., Woodford-Richens, K., Tomlinson, I.,
   Lawrence, C.M. and Douglas, F.S. (2003) Laugier–Hunziker syndrome:
   an important differential diagnosis for Peutz–Jeghers syndrome. J.
   Med. Genet., 40, e77.
   FREE Full Text
29. ↵
   Suter, C.M., Martin, D.I. and Ward, R.L. (2004) Germline epimutation
   of MLH1 in individuals with multiple cancers. Nat. Genet., 36, 497–
   501.
   CrossRefMedlineWeb of Science
30. ↵
   Wu, Y., Berends, M.J., Sijmons, R.H., Mensink, R.G., Verlind, E., Kooi,
   K.A., van der Sluis, T., Kempinga, C., van der Zee, A.G., Hollema, H.
   et al. (2001) A role for MLH3 in hereditary nonpolyposis colorectal
   cancer. Nat. Genet., 29, 137–138.
   CrossRefMedlineWeb of Science
31. Liu, H.X., Zhou, X.L., Liu, T., Werelius, B., Lindmark, G., Dahl, N. and
   Lindblom, A. (2003) The role of hMLH3 in familial colorectal cancer.
   Cancer Res., 63, 1894–1899.
   Abstract/FREE Full Text
32. ↵
Hienonen, T., Laiho, P., Salovaara, R., Mecklin, J.P., Jarvinen, H.,
   Sistonen, P., Peltomaki, P., Lehtonen, R., Nupponen, N.N., Launonen,
   V. et al. (2003) Little evidence for involvement of MLH3 in colorectal
   cancer predisposition. Int. J. Cancer, 106, 292–296.
   CrossRefMedlineWeb of Science
33. ↵
   Wu, Y., Berends, M.J., Post, J.G., Mensink, R.G., Verlind, E., van der
   Sluis, T., Kempinga, C., Sijmons, R.H., van der Zee, A.G., Hollema, H.
   et al. (2001) Germline mutations of EXO1 gene in patients with
   hereditary nonpolyposis colorectal cancer (HNPCC) and atypical
   HNPCC forms. Gastroenterology, 120, 1580–1587.
   CrossRefMedlineWeb of Science
34. Tomlinson, I. and Houlston, R. (2001) Is EXO1 a colon cancer
   predisposition gene? Gastroenterology, 120, 1860–1862.
   MedlineWeb of Science
35. Alam, N.A., Gorman, P., Jaeger, E.E., Kelsell, D., Leigh, I.M., Ratnavel,
   R., Murdoch, M.E., Houlston, R.S., Aaltonen, L.A., Roylance, R.R. et al.
   (2003) Germline deletions of EXO1 do not cause colorectal tumors
   and lesions which are null for EXO1 do not have microsatellite
   instability. Cancer Genet. Cytogenet., 147, 121–127.
   CrossRefMedlineWeb of Science
36. ↵
   Jagmohan-Changur, S., Poikonen, T., Vilkki, S., Launonen, V.,
   Wikman, F., Orntoft, T.F., Moller, P., Vasen, H., Tops, C., Kolodner,
   R.D. et al. (2003) EXO1 variants occur commonly in normal
   population: evidence against a role in hereditary nonpolyposis
   colorectal cancer. Cancer Res., 63, 154–158.
   Abstract/FREE Full Text
37. ↵
   Johnson, V., Volikos, E., Talbot, I., Tomlinson, I. and Silver, A. (2004)
   Beta-catenin mutations are specific to carcinomas in the HNPCC
   syndrome. Gut, in press.
38. ↵
   Jiricny, J. and Marra, G. (2003) DNA repair defects in colon cancer.
   Curr. Opin. Genet. Dev., 13, 61–69.
   CrossRefMedlineWeb of Science
39. ↵
   Crabtree, M., Sieber, O.M., Lipton, L., Hodgson, S.V., Lamlum, H.,
   Thomas, H.J., Neale, K., Phillips, R.K., Heinimann, K. and Tomlinson,
   I.P. (2003) Refining the relation between ‘first hits’ and ‘second hits’
   at the APC locus: the ‘loose fit’ model and evidence for differences in
   somatic mutation spectra among patients. Oncogene, 22, 4257–
   4265.
CrossRefMedlineWeb of Science
40. ↵
   Lamlum, H., Ilyas, M., Rowan, A., Clark, S., Johnson, V., Bell, J.,
   Frayling, I., Efstathiou, J., Pack, K., Payne, S. et al. (1999) The type of
   somatic mutation at APC in familial adenomatous polyposis is
   determined by the site of the germline mutation: a new facet to
   Knudson's ‘two-hit’ hypothesis. Nat. Med., 5, 1071–1075.
   CrossRefMedlineWeb of Science
41. Rowan, A.J., Lamlum, H., Ilyas, M., Wheeler, J., Straub, J.,
   Papadopoulou, A., Bicknell, D., Bodmer, W.F. and Tomlinson, I.P.
   (2000) APC mutations in sporadic colorectal tumors: a mutational
   ‘hotspot’ and interdependence of the ‘two hits’. Proc. Natl Acad. Sci.
   USA, 97, 3352–3357.
   CrossRefMedlineWeb of Science
42. ↵
   Albuquerque, C., Breukel, C., van der Luijt, R., Fidalgo, P., Lage, P.,
   Slors, F.J., Leitao, C.N., Fodde, R. and Smits, R. (2002) The ‘just-
   right’ signaling model: APC somatic mutations are selected based on
   a specific level of activation of the beta-catenin signaling cascade.
   Hum. Mol. Genet., 11, 1549–1560.
   Abstract/FREE Full Text
43. ↵
   Groves, C., Lamlum, H., Crabtree, M., Williamson, J., Taylor, C., Bass,
   S., Cuthbert-Heavens, D., Hodgson, S., Phillips, R. and Tomlinson, I.
   (2002) Mutation cluster region, association between germline and
   somatic mutations and genotype–phenotype correlation in upper
   gastrointestinal familial adenomatous polyposis. Am. J. Pathol., 160,
   2055–2061.
   MedlineWeb of Science
44. ↵
   Spirio, L.N., Samowitz, W., Robertson, J., Robertson, M., Burt, R.W.,
   Leppert, M. and White, R. (1998) Alleles of APC modulate the
   frequency and classes of mutations that lead to colon polyps. Nat.
   Genet., 20, 385–388.
   CrossRefMedlineWeb of Science
45. ↵
   Su, L.K., Barnes, C.J., Yao, W., Qi, Y., Lynch, P.M. and Steinbach, G.
   (2000) Inactivation of germline mutant APC alleles by attenuated
   somatic mutations: a molecular genetic mechanism for attenuated
   familial adenomatous polyposis. Am. J. Hum. Genet., 67, 582–590.
   CrossRefMedlineWeb of Science
46. ↵
Crabtree, M.D., Tomlinson, I.P., Talbot, I.C. and Phillips, R.K. (2001)
   Variability in the severity of colonic disease in familial adenomatous
   polyposis results from differences in tumour initiation rather than
   progression and depends relatively little on patient age. Gut, 49,
   540–543.
   Abstract/FREE Full Text
47. ↵
   Bertario, L., Russo, A., Sala, P., Eboli, M., Giarola, M., D'Amico, F.,
   Gismondi, V., Varesco, L., Pierotti, M.A. and Radice, P. (2001)
   Genotype and phenotype factors as determinants of desmoid tumors
   in patients with familial adenomatous polyposis. Int. J. Cancer, 95,
   102–107.
   CrossRefMedlineWeb of Science
48. ↵
   Dietrich, W.F., Lander, E.S., Smith, J.S., Moser, A.R., Gould, K.A.,
   Luongo, C., Borenstein, N. and Dove, W. (1993) Genetic identification
   of Mom-1, a major modifier locus affecting Min-induced intestinal
   neoplasia in the mouse. Cell, 75, 631–639.
   CrossRefMedlineWeb of Science
49. MacPhee, M., Chepenik, K.P., Liddell, R.A., Nelson, K.K., Siracusa,
   L.D. and Buchberg, A.M. (1995) The secretory phospholipase A2
   gene is a candidate for the Mom1 locus, a major modifier of ApcMin-
   induced intestinal neoplasia. Cell, 81, 957–966.
   CrossRefMedlineWeb of Science
50. ↵
   Cormier, R.T., Hong, K.H., Halberg, R.B., Hawkins, T.L., Richardson,
   P., Mulherkar, R., Dove, W.F. and Lander, E.S. (1997) Secretory
   phospholipase Pla2g2a confers resistance to intestinal
   tumorigenesis. Nat. Genet., 17, 88–91.
   CrossRefMedlineWeb of Science
51. ↵
   Crabtree, M.D., Fletcher, C., Churchman, M., Hodgson, S.V., Neale,
   K., Phillips, R.K. and Tomlinson, I.P. (2004) Analysis of candidate
   modifier loci for the severity of colonic familial adenomatous
   polyposis, with evidence for the importance of the N-acetyl
   transferases. Gut, 53, 271–276.
   Abstract/FREE Full Text
52. ↵
   Laken, S.J., Petersen, G.M., Gruber, S.B., Oddoux, C., Ostrer, H.,
   Giardiello, F.M., Hamilton, S.R., Hampel, H., Markowitz, A., Klimstra,
   D. et al. (1997) Familial colorectal cancer in Ashkenazim due to a
   hypermutable tract in APC. Nat. Genet., 17, 79–83.
   CrossRefMedlineWeb of Science
53. ↵
   Strul, H., Barenboim, E., Leshno, M., Gartner, M., Kariv, R., Aljadeff,
   E., Aljadeff, Y., Kazanov, D., Strier, L., Keidar, A. et al. (2003) The
   I1307K adenomatous polyposis coli gene variant does not contribute
   in the assessment of the risk for colorectal cancer in Ashkenazi Jews.
   Cancer Epidemiol. Biomark. Prev., 12, 1012–1015.
   Abstract/FREE Full Text
54. ↵
   Sieber, O., Lipton, L., Heinimann, K. and Tomlinson, I. (2003)
   Colorectal tumourigenesis in carriers of the APC I1307K variant: lone
   gunman or conspiracy? J. Pathol., 199, 137–1379.
   CrossRefMedlineWeb of Science
55. ↵
   Whitelaw, S.C., Murday, V.A., Tomlinson, I.P., Thomas, H.J., Cottrell,
   S., Ginsberg, A., Bukofzer, S., Hodgson, S.V., Skudowitz, R.B., Jass,
   J.R. et al. (1997) Clinical and molecular features of the hereditary
   mixed polyposis syndrome. Gastroenterology, 112, 327–334.
   CrossRefMedlineWeb of Science
56. ↵
   Thomas, H.J., Whitelaw, S.C., Cottrell, S.E., Murday, V.A., Tomlinson,
   I.P., Markie, D., Jones, T., Bishop, D.T., Hodgson, S.V., Sheer, D. et al.
   (1996) Genetic mapping of hereditary mixed polyposis syndrome to
   chromosome 6q. Am. J. Hum. Genet., 58, 770–776.
   MedlineWeb of Science
57. ↵
   Tomlinson, I., Rahman, N., Frayling, I., Mangion, J., Barfoot, R.,
   Hamoudi, R., Seal, S., Northover, J., Thomas, H.J., Neale, K. et al.
   (1999) Inherited susceptibility to colorectal adenomas and
   carcinomas: evidence for a new predisposition gene on 15q14–q22.
   Gastroenterology, 116, 789–795.
   CrossRefMedlineWeb of Science
58. ↵
   Jaeger, E.E., Woodford-Richens, K.L., Lockett, M., Rowan, A.J.,
   Sawyer, E.J., Heinimann, K., Rozen, P., Murday, V.A., Whitelaw, S.C.,
   Ginsberg, A. et al. (2003) An ancestral Ashkenazi haplotype at the
   HMPS/CRAC1 locus on 15q13–q14 is associated with hereditary
   mixed polyposis syndrome. Am. J. Hum. Genet., 72, 1261–1267.
   CrossRefMedlineWeb of Science
59. ↵
   Wiesner, G.L., Daley, D., Lewis, S., Ticknor, C., Platzer, P.,
   Lutterbaugh, J., MacMillen, M., Baliner, B., Willis, J., Elston, R.C. et al.
   (2003) A subset of familial colorectal neoplasia kindreds linked to
chromosome 9q22.2–31.2. Proc. Natl Acad. Sci. USA, 100, 12961–
   12965.
   Abstract/FREE Full Text
60. ↵
   Wacholder, S., Chanock, S., Garcia-Closas, M., El Ghormli, L. and
   Rothman, N. (2004) Assessing the probability that a positive report is
   false: an approach for molecular epidemiology studies. J. Natl Cancer
   Inst., 96, 434–442.
   Abstract/FREE Full Text
61. ↵
   Egger, M., Smith, G.D. and Sterne, J.A. (2001) Uses and abuses of
   meta-analysis. Clin. Med., 1, 478–484.
   MedlineWeb of Science
62. ↵
   Sterne, J.A. and Egger, M. (2001) Funnel plots for detecting bias in
   meta-analysis: guidelines on choice of axis. J. Clin. Epidemiol., 54,
   1046–1055.
   CrossRefMedlineWeb of Science
63. ↵
   Houlston, R.S. and Tomlinson, I.P. (2001) Polymorphisms and
   colorectal tumor risk. Gastroenterology, 121, 282–301.
   CrossRefMedlineWeb of Science
64. ↵
   de Jong, M.M., Nolte, I.M., te Meerman, G.J., van der Graaf, W.T., de
   Vries, E.G., Sijmons, R.H., Hofstra, R.M. and Kleibeuker, J.H. (2002)
   Low-penetrance genes and their involvement in colorectal cancer
   susceptibility. Cancer Epidemiol. Biomark. Prev., 11, 1332–1352.
   Abstract/FREE Full Text
65. ↵
   Gruber, S.B., Ellis, N.A., Scott, K.K., Almog, R., Kolachana, P., Bonner,
   J.D., Kirchhoff, T., Tomsho, L.P., Nafa, K., Pierce, H. et al. (2002) BLM
   heterozygosity and the risk of colorectal cancer. Science, 297, 2013.
   FREE Full Text
66. ↵
   Cleary, S.P., Zhang, W., Di Nicola, N., Aronson, M., Aube, J.,
   Steinman, A., Haddad, R., Redston, M., Gallinger, S., Narod, S.A. et al.
   (2003) Heterozygosity for the BLM(Ash) mutation and cancer risk.
   Cancer Res., 63, 1769–1771.
   Abstract/FREE Full Text
67. ↵
   Sharp, L. and Little, J. (2004) Polymorphisms in genes involved in
   folate metabolism and colorectal neoplasia: a HuGE review. Am. J.
   Epidemiol., 159, 423–443.
Abstract/FREE Full Text
68. ↵
   Grieu, F., Malaney, S., Ward, R., Joseph, D. and Iacopetta, B. (2003)
   Lack of association between CCND1 G870A polymorphism and the
   risk of breast and colorectal cancers. Anticancer Res., 23, 4257–
   4259.
   MedlineWeb of Science
69. ↵
   Le Marchand, L., Seifried, A., Lum-Jones, A., Donlon, T. and Wilkens,
   L.R. (2003) Association of the cyclin D1 A870G polymorphism with
   advanced colorectal cancer. J. Am. Med. Assoc., 290, 2843–2848.
   Abstract/FREE Full Text
70. ↵
   Shin, Y., Kim, I.J., Kang, H.C., Park, J.H., Park, H.W., Jang, S.G., Lee,
   M.R., Jeong, S.Y., Chang, H.J., Ku, J.L. et al. (2004) A functional
   polymorphism (−347 G→GA) in the E-cadherin gene is associated
   with colorectal cancer. Carcinogenesis, in press.
71. ↵
   Ulrich, C.M., Bigler, J., Sparks, R., Whitton, J., Sibert, J.G., Goode, E.L.,
   Yasui, Y. and Potter, J.D. (2004) Polymorphisms in PTGS1 (=COX-1)
   and risk of colorectal polyps. Cancer Epidemiol. Biomark. Prev., 13,
   889–893.
   Abstract/FREE Full Text
72. ↵
   Lin, H.J., Lakkides, K.M., Keku, T.O., Reddy, S.T., Louie, A.D., Kau,
   I.H., Zhou, H., Gim, J.S., Ma, H.L., Matthies, C.F. et al. (2002)
   Prostaglandin H synthase 2 variant (Val511Ala) in African Americans
   may reduce the risk for colorectal neoplasia. Cancer Epidemiol.
   Biomark. Prev., 11, 1305–1315.
   Abstract/FREE Full Text
73. ↵
   Shaheen, N.J., Silverman, L.M., Keku, T., Lawrence, L.B., Rohlfs, E.M.,
   Martin, C.F., Galanko, J. and Sandler, R.S. (2003) Association between
   hemochromatosis (HFE) gene mutation carrier status and the risk of
   colon cancer. J. Natl Cancer Inst., 95, 154–159.
   Abstract/FREE Full Text
74. ↵
   Slattery, M.L., Samowitz, W., Curtin, K., Ma, K.N., Hoffman, M., Caan,
   B. and Neuhausen, S. (2004) Associations among IRS1, IRS2, IGF1,
   and IGFBP3 genetic polymorphisms and colorectal cancer. Cancer
   Epidemiol. Biomark. Prev., 13, 1206–1214.
   Abstract/FREE Full Text
75. ↵
Slattery, M.L., Samowitz, W., Hoffman, M., Ma, K.N., Levin, T.R. and
   Neuhausen, S. (2004) Aspirin, NSAIDs, and colorectal cancer:
   possible involvement in an insulin-related pathway. Cancer
   Epidemiol. Biomark. Prev., 13, 538–545.
   Abstract/FREE Full Text
76. ↵
   Landi, S., Moreno, V., Gioia-Patricola, L., Guino, E., Navarro, M., de
   Oca, J., Capella, G. and Canzian, F. (2003) Association of common
   polymorphisms in inflammatory genes interleukin (IL)6, IL8, tumor
   necrosis factor alpha, NFKB1, and peroxisome proliferator-activated
   receptor gamma with colorectal cancer. Cancer Res., 63, 3560–3566.
   Abstract/FREE Full Text
77. ↵
   Lipkin, S.M., Rozek, L.S., Rennert, G., Yang, W., Chen, P.C., Hacia, J.,
   Hunt, N., Shin, B., Fodor, S., Kokoris, M. et al. (2004) The MLH1
   D132H variant is associated with susceptibility to sporadic colorectal
   cancer. Nat. Genet., 36, 694–699.
   CrossRefMedlineWeb of Science
78. ↵
   de Jong, M.M., Hofstra, R.M., Kooi, K.A., Westra, J.L., Berends, M.J.,
   Wu, Y., Hollema, H., van der Sluis, T., van der Graaf, W.T., de Vries,
   E.G. et al. (2004) No association between two MLH3 variants (S845G
   and P844L) and colorectal cancer risk. Cancer Genet. Cytogenet.,
   152, 70–71.
   CrossRefMedlineWeb of Science
79. ↵
   Hinoda, Y., Okayama, N., Takano, N., Fujimura, K., Suehiro, Y.,
   Hamanaka, Y., Hazama, S., Kitamura, Y., Kamatani, N. and Oka, M.
   (2002) Association of functional polymorphisms of matrix
   metalloproteinase (MMP)-1 and MMP-3 genes with colorectal cancer.
   Int. J. Cancer, 102, 526–529.
   CrossRefMedlineWeb of Science
80. ↵
   Matsuo, K., Hamajima, N., Hirai, T., Kato, T., Inoue, M., Takezaki, T.
   and Tajima, K. (2002) Methionine synthase reductase gene A66G
   polymorphism is associated with risk of colorectal cancer. Asian Pac.
   J. Cancer Prev., 3, 353–359.
   Medline
81. ↵
   Goode, E.L., Potter, J.D., Bigler, J. and Ulrich, C.M. (2004) Methionine
   synthase D919G polymorphism, folate metabolism, and colorectal
   adenoma risk. Cancer Epidemiol. Biomark. Prev., 13, 157–162.
   Abstract/FREE Full Text
82. ↵
   Loktionov, A., Watson, M.A., Stebbings, W.S., Speakman, C.T. and
   Bingham, S.A. (2003) Plasminogen activator inhibitor-1 gene
   polymorphism and colorectal cancer risk and prognosis. Cancer Lett.,
   189, 189–196.
   CrossRefMedlineWeb of Science
83. ↵
   Sparks, R., Bigler, J., Sibert, J.G., Potter, J.D., Yasui, Y. and Ulrich,
   C.M. (2004) TGF{beta}1 polymorphism (L10P) and risk of colorectal
   adenomatous and hyperplastic polyps. Int. J. Epidemiol., in press.
84. ↵
   Pasche, B., Kaklamani, V., Hou, N., Young, T., Rademaker, A.,
   Peterlongo, P., Ellis, N., Offit, K., Caldes, T., Reiss, M. et al. (2004)
   TGFBR1*6A and cancer: a meta-analysis of 12 case-control studies.
   J. Clin. Oncol., 22, 756–758.
   FREE Full Text
85. ↵
   Chen, J., Hunter, D.J., Stampfer, M.J., Kyte, C., Chan, W., Wetmur, J.G.,
   Mosig, R., Selhub, J. and Ma, J. (2003) Polymorphism in the
   thymidylate synthase promoter enhancer region modifies the risk
   and survival of colorectal cancer. Cancer Epidemiol Biomark. Prev.,
   12, 958–962.
   Abstract/FREE Full Text
86. ↵
   Ulrich, C.M., Bigler, J., Bostick, R., Fosdick, L. and Potter, J.D. (2002)
   Thymidylate synthase promoter polymorphism, interaction with
   folate intake, and risk of colorectal adenomas. Cancer Res., 62,
   3361–3364.
   Abstract/FREE Full Text
87. ↵
   Wong, H.L., Seow, A., Arakawa, K., Lee, H.P., Yu, M.C. and Ingles, S.A.
   (2003) Vitamin D receptor start codon polymorphism and colorectal
   cancer risk: effect modification by dietary calcium and fat in
   Singapore Chinese. Carcinogenesis, 24, 1091–1095.
   Abstract/FREE Full Text
88. ↵
   Couzin, J. (2004) Genomics. Consensus emerges on HapMap
   strategy. Science, 304, 671–673.
   Abstract/FREE Full Text
89. ↵
   Cui, H., Cruz-Correa, M., Giardiello, F.M., Hutcheon, D.F., Kafonek,
   D.R., Brandenburg, S., Wu, Y., He, X., Powe, N.R. and Feinberg, A.P.
(2003) Loss of IGF2 imprinting: a potential marker of colorectal
   cancer risk. Science, 299, 1753–1755.
   Abstract/FREE Full Text
90. Cruz-Correa, M., Cui, H., Giardiello, F.M., Powe, N.R., Hylind, L.,
   Robinson, A., Hutcheon, D.F., Kafonek, D.R., Brandenburg, S., Wu, Y.
   et al. (2004) Loss of imprinting of insulin growth factor II gene: a
   potential heritable biomarker for colon neoplasia predisposition.
   Gastroenterology, 126, 964–970.
   CrossRefMedlineWeb of Science
91. Woodson, K., Flood, A., Green, L., Tangrea, J.A., Hanson, J., Cash, B.,
   Schatzkin, A. and Schoenfeld, P. (2004) Loss of insulin-like growth
   factor-II imprinting and the presence of screen-detected colorectal
   adenomas in women. J. Natl Cancer Inst., 96, 407–410.
   Abstract/FREE Full Text
92. ↵
   Jirtle, R.L. (2004) IGF2 loss of imprinting: a potential heritable risk
   factor for colorectal cancer. Gastroenterology, 126, 1190–1193.
   CrossRefMedlineWeb of Science
93. ↵
   White, S., Bubb, V.J. and Wyllie, A.H. (1996) Germline APC mutation
   (Gln1317) in a cancer-prone family that does not result in familial
   adenomatous polyposis. Genes Chromosomes Cancer, 15, 122–128.
   CrossRefMedlineWeb of Science
94. Frayling, I.M., Beck, N.E., Ilyas, M., Dove-Edwin, I., Goodman, P.,
   Pack, K., Bell, J.A., Williams, C.B., Hodgson, S.V., Thomas, H.J. et al.
   (1998) The APC variants I1307K and E1317Q are associated with
   colorectal tumors, but not always with a family history. Proc. Natl
   Acad. Sci. USA, 95, 10722–10727.
   Abstract/FREE Full Text
95. Popat, S., Stone, J., Coleman, G., Marshall, G., Peto, J., Frayling, I. and
   Houlston, R. (2000) Prevalence of the APC E1317Q variant in
   colorectal cancer patients. Cancer Lett., 149, 203–206.
   CrossRefMedlineWeb of Science
96. Gismondi, V., Bonelli, L., Sciallero, S., Margiocco, P., Viel, A., Radice,
   P., Mondini, P., Sala, P., Montera, M.P., Mareni, C. et al. (2002)
   Prevalence of the E1317Q variant of the APC gene in Italian patients
   with colorectal adenomas. Genet. Test., 6, 313–317.
   CrossRefMedlineWeb of Science
97. ↵
   Hahnloser, D., Petersen, G.M., Rabe, K., Snow, K., Lindor, N.M.,
   Boardman, L., Koch, B., Doescher, D., Wang, L., Steenblock, K. et al.
   (2003) The APC E1317Q variant in adenomatous polyps and
   colorectal cancers. Cancer Epidemiol. Biomark. Prev., 12, 1023–1028.
Abstract/FREE Full Text
      98. ↵
           Meijers-Heijboer, H., van den Ouweland, A., Klijn, J., Wasielewski, M.,
           de Snoo, A., Oldenburg, R., Hollestelle, A., Houben, M., Crepin, E.,
           van Veghel-Plandsoen, M. et al. (2002) Low-penetrance susceptibility
           to breast cancer due to CHEK2(*)1100delC in noncarriers of BRCA1
           or BRCA2 mutations. Nat. Genet., 31, 55–59.
           CrossRefMedlineWeb of Science
      99. ↵
           Houlston, R.S. and Tomlinson, I.P. (2000) Detecting low penetrance
           genes in cancer: the way ahead. J. Med. Genet., 37, 161–167.
           Abstract/FREE Full Text
      100. ↵
           Risch, N. (2001) Implications of multilocus inheritance for gene–
           disease association studies. Theor. Popul. Biol., 60, 215–220.
           CrossRefMedlineWeb of Science
      101. ↵
           Houlston, R.S. and Peto, J. (2003) The future of association studies of
           common cancers. Hum. Genet., 112, 434–435.
           MedlineWeb of Science
      102. ↵
           Botstein, D. and Risch, N. (2003) Discovering genotypes underlying
           human phenotypes: past successes for mendelian disease, future
           approaches for complex disease. Nat. Genet., 33 (suppl.), 228–237.
      103. ↵
           Gabriel, S.B., Schaffner, S.F., Nguyen, H., Moore, J.M., Roy, J.,
           Blumenstiel, B., Higgins, J., DeFelice, M., Lochner, A., Faggart, M.
           et al. (2002) The structure of haplotype blocks in the human
           genome. Science, 296, 2225–2229.
           Abstract/FREE Full Text
      104. ↵
           Satsangi, J., Parkes, M., Jewell, D.P. and Bell, J.I. (1998) Genetics of
           inflammatory bowel disease (Review). Clin. Sci. (London), 94, 473–
           478.


Articles citing this article
    Characterization of 9p24 Risk Locus and Colorectal Adenoma and Cancer:
    Gene-Environment Interaction and Meta-Analysis Cancer Epidemiol. Biomarkers Prev.
    (2010) 19(12): 3131-3139
       o Abstract
       o Full Text (HTML)
       o Full Text (PDF)
    Allele-specific expression of TGFBR1 in colon cancer patients   Carcinogenesis (2010)
    31(10): 1800-1804
      o    Abstract
o Full Text (HTML)
             o Full Text (PDF)
          Association of the ARLTS1 Cys148Arg variant with sporadic and familial
          colorectal cancer Carcinogenesis (2007) 28(8): 1687-1691
             o Abstract
             o Full Text (HTML)
             o Full Text (PDF)
          TGF-{beta} signaling alterations and susceptibility to colorectal cancer Hum Mol
          Genet (2007) 16(R1): R14-R20
             o       Abstract
             o       Full Text (HTML)
             o       Full Text (PDF)
                                   « Previous | Next Article »Table of Contents
This Article

                     1.   Hum. Mol. Genet. (2004) 13 (suppl 2): R177-R185. doi: 10.1093/hmg/ddh247


    1.    Abstract
    2.    » Full Text (HTML)
    3.    Full Text (PDF)
- Classifications
     1.
                 o    Review
                 o    Review
- Services


    1.    Alert me when cited
    2.    Alert me if corrected
    3.    Find similar articles
    5.    Similar articles in PubMed
    6.    Add to my archive
    7.    Download citation
    8.    Request Permissions
+   Citing Articles
+   Google Scholar
+   PubMed
+   Related Content
+   Share

             o
             o
             o
             o
             o
       o
       o
Navigate This Article

    1.    Top
    2.    Abstract
    3.    PROGRESS IN UNDERSTANDING THE HEREDITARY COLORECTAL TUMOUR SYNDROMES
    4.    RECENT DEVELOPMENTS THROUGH LINKAGE ANALYSIS
    5.    EVIDENCE FOR LOW-PENETRANCE ALLELES ASSOCIATED WITH AN INCREASED RISK OF CRC
    6.    FUTURE STRATEGIES FOR IDENTIFYING CRC PREDISPOSITION GENES
    7.    ACKNOWLEDGEMENTS
    8.    References
- People also read [Beta]
1.    Specific codon 13 K-ras mutations are predictive of clinical outcome in
                         colorectal cancer patients, whereas codon 12 K-ras mutations are associated
                         with mucinous histotype




                What's this?



Search this journal:



Advanced »
Current Issue
                                      1. January 15, 2012 21 (2)




                                      1.

                                       1. Alert me to new issues


The Journal
              About this journal
              Rights & Permissions
              Dispatch date of the next issue
              This journal is a member of the Committee on Publication Ethics (COPE)
              We are mobile – find out more

Impact factor: 8.058
5-Yr impact factor: 8.145
Executive Editors
 Professor Kay Davies
 Professor Anthony Wynshaw-Boris
 Professor Joel Hirschhorn

                        View full editorial board

       For Authors

                                Instructions to authors
                                Online submission
                                Submit Now!
                                Self-archiving policy
Open access options for authors - visit Oxford Open




                  This journal enables compliance with the
                  NIH Public Access Policy

      Alerting Services

                          Email table of contents
                          Email Advance Access
                          CiteTrack
                          XML RSS feed

             Corporate Services

                                 Advertising sales
                                 Reprints
                                 Supplements

                      Most

                                    Most Read
                                    Most Cited


                                   Most Read
                                              1. Epistasis: what it
                                                 means, what it
                                                 doesn't mean, and
                                                 statistical methods
                                                 to detect it in
                                                 humans
                                              2. Genetics of cleft lip
                                                 and palate:
                                                 syndromic genes
                                                 contribute to the
                                                 incidence of non-
                                                 syndromic clefts
                                              3. The genetics of
                                                 psoriasis, psoriatic
                                                 arthritis and atopic
                                                 dermatitis
                                              4. Non-coding RNA
                                              5. Synergy Between
                                                 the Genes for
                                                 Butyrylcholinestera
                                                 se K Variant and
                                                 Apolipoprotein E4
                                                 in Late-Onset
Confirmed
                                              Alzheimer's
                                              Disease

                                » View all Most Read articles

                                Most Cited
                                          1. The DNA
                                             methyltransferases
                                             of mammals
                                          2. Nonsense-Mediated
                                             mRNA Decay in
                                             Health and Disease
                                          3. Isolation of a
                                             Candidate Human
                                             Telomerase
                                             Catalytic Subunit
                                             Gene, Which
                                             Reveals Complex
                                             Splicing Patterns in
                                             Different Cell Types
                                          4. Human Y
                                             Chromosome
                                             Azoospermia
                                             Factors (AZF)
                                             Mapped to
                                             Different
                                             Subregions in Yq11
                                          5. Mutation of human
                                             short tandem
                                             repeats

                                » View all Most Cited articles

                     Online ISSN 1460-2083 - Print ISSN 0964-6906
                          Copyright © 2011 Oxford University Press
Oxford Journals Oxford University Press
                                                        Site Map
                                                   Privacy Policy
                                     Frequently Asked Questions
Other Oxford University Press sites:

   Oxford University Press

Mais conteúdo relacionado

Mais procurados

Key achievements in cancer research at Cold Spring Harbor Laboratory: May 201...
Key achievements in cancer research at Cold Spring Harbor Laboratory: May 201...Key achievements in cancer research at Cold Spring Harbor Laboratory: May 201...
Key achievements in cancer research at Cold Spring Harbor Laboratory: May 201...cshlnews
 
Dott.ssa Serena Merante, “RISCHIO DI SECONDI TUMORI E LEUCEMIA MIELOIDE CRONICA”
Dott.ssa Serena Merante, “RISCHIO DI SECONDI TUMORI E LEUCEMIA MIELOIDE CRONICA”Dott.ssa Serena Merante, “RISCHIO DI SECONDI TUMORI E LEUCEMIA MIELOIDE CRONICA”
Dott.ssa Serena Merante, “RISCHIO DI SECONDI TUMORI E LEUCEMIA MIELOIDE CRONICA”AIP LEUCEMIA MIELOIDE CRONICA
 
Microsatellite instability pathway and molecular genetics of colorectal cancer
Microsatellite instability pathway and molecular genetics of colorectal cancerMicrosatellite instability pathway and molecular genetics of colorectal cancer
Microsatellite instability pathway and molecular genetics of colorectal cancerDr.Juel Chowdhury MD
 
Alfred knudson and the two hit hypothesis
Alfred knudson and the two hit hypothesisAlfred knudson and the two hit hypothesis
Alfred knudson and the two hit hypothesisWEI LIN
 
Kurrey_et_al-2009-STEM_CELLS
Kurrey_et_al-2009-STEM_CELLSKurrey_et_al-2009-STEM_CELLS
Kurrey_et_al-2009-STEM_CELLSSwati Jalgaonkar
 
Zeine et al. Poster 2007 Cancer Associated Fibroblasts in Neuroblastoma
Zeine et al. Poster 2007 Cancer Associated Fibroblasts in NeuroblastomaZeine et al. Poster 2007 Cancer Associated Fibroblasts in Neuroblastoma
Zeine et al. Poster 2007 Cancer Associated Fibroblasts in NeuroblastomaRana ZEINE, MD, PhD, MBA
 
Peddinti, Zeine et al. Clinical Cancer Research 2007
Peddinti, Zeine et al. Clinical Cancer Research 2007Peddinti, Zeine et al. Clinical Cancer Research 2007
Peddinti, Zeine et al. Clinical Cancer Research 2007Rana ZEINE, MD, PhD, MBA
 
Different Faces of Fas Signaling in Cancer Cells-Crimson Publishers
Different Faces of Fas Signaling in Cancer Cells-Crimson PublishersDifferent Faces of Fas Signaling in Cancer Cells-Crimson Publishers
Different Faces of Fas Signaling in Cancer Cells-Crimson PublishersCrimsonpublishersCancer
 
Molecular profiling of breast cancer
Molecular profiling of breast cancerMolecular profiling of breast cancer
Molecular profiling of breast cancerHriman Sharma Sarkar
 
Lombardi et al: XMRV/CFS Inflammatory Signature
Lombardi et al: XMRV/CFS Inflammatory SignatureLombardi et al: XMRV/CFS Inflammatory Signature
Lombardi et al: XMRV/CFS Inflammatory Signaturedegarden
 
CCO Rational Options in Breast Cancer : How Molecular Understanding Informs T...
CCO Rational Options in Breast Cancer : How Molecular Understanding Informs T...CCO Rational Options in Breast Cancer : How Molecular Understanding Informs T...
CCO Rational Options in Breast Cancer : How Molecular Understanding Informs T...Adonis Guancia
 
Genetic predipositio to cancer
Genetic predipositio to cancerGenetic predipositio to cancer
Genetic predipositio to cancerMahran Alnahmi
 
Pró reitoria de pós-graduação e pesquisa programa de pós-graduação stricto se...
Pró reitoria de pós-graduação e pesquisa programa de pós-graduação stricto se...Pró reitoria de pós-graduação e pesquisa programa de pós-graduação stricto se...
Pró reitoria de pós-graduação e pesquisa programa de pós-graduação stricto se...José Antonio Paniagua
 
Case 1 & 2
Case 1 & 2Case 1 & 2
Case 1 & 2kciapm
 
Biología molecular
Biología molecularBiología molecular
Biología molecularIMSS
 
Molecular profiling in breast cancer
Molecular profiling in breast cancerMolecular profiling in breast cancer
Molecular profiling in breast cancerShashidhara TS
 

Mais procurados (20)

Key achievements in cancer research at Cold Spring Harbor Laboratory: May 201...
Key achievements in cancer research at Cold Spring Harbor Laboratory: May 201...Key achievements in cancer research at Cold Spring Harbor Laboratory: May 201...
Key achievements in cancer research at Cold Spring Harbor Laboratory: May 201...
 
Dott.ssa Serena Merante, “RISCHIO DI SECONDI TUMORI E LEUCEMIA MIELOIDE CRONICA”
Dott.ssa Serena Merante, “RISCHIO DI SECONDI TUMORI E LEUCEMIA MIELOIDE CRONICA”Dott.ssa Serena Merante, “RISCHIO DI SECONDI TUMORI E LEUCEMIA MIELOIDE CRONICA”
Dott.ssa Serena Merante, “RISCHIO DI SECONDI TUMORI E LEUCEMIA MIELOIDE CRONICA”
 
Microsatellite instability pathway and molecular genetics of colorectal cancer
Microsatellite instability pathway and molecular genetics of colorectal cancerMicrosatellite instability pathway and molecular genetics of colorectal cancer
Microsatellite instability pathway and molecular genetics of colorectal cancer
 
Genome wide study
Genome wide studyGenome wide study
Genome wide study
 
Alfred knudson and the two hit hypothesis
Alfred knudson and the two hit hypothesisAlfred knudson and the two hit hypothesis
Alfred knudson and the two hit hypothesis
 
Kurrey_et_al-2009-STEM_CELLS
Kurrey_et_al-2009-STEM_CELLSKurrey_et_al-2009-STEM_CELLS
Kurrey_et_al-2009-STEM_CELLS
 
Zeine et al. Poster 2007 Cancer Associated Fibroblasts in Neuroblastoma
Zeine et al. Poster 2007 Cancer Associated Fibroblasts in NeuroblastomaZeine et al. Poster 2007 Cancer Associated Fibroblasts in Neuroblastoma
Zeine et al. Poster 2007 Cancer Associated Fibroblasts in Neuroblastoma
 
ASCO heterogeneity presentation El-Deiry 6-4-17
ASCO heterogeneity presentation El-Deiry 6-4-17ASCO heterogeneity presentation El-Deiry 6-4-17
ASCO heterogeneity presentation El-Deiry 6-4-17
 
Peddinti, Zeine et al. Clinical Cancer Research 2007
Peddinti, Zeine et al. Clinical Cancer Research 2007Peddinti, Zeine et al. Clinical Cancer Research 2007
Peddinti, Zeine et al. Clinical Cancer Research 2007
 
Different Faces of Fas Signaling in Cancer Cells-Crimson Publishers
Different Faces of Fas Signaling in Cancer Cells-Crimson PublishersDifferent Faces of Fas Signaling in Cancer Cells-Crimson Publishers
Different Faces of Fas Signaling in Cancer Cells-Crimson Publishers
 
Molecular profiling of breast cancer
Molecular profiling of breast cancerMolecular profiling of breast cancer
Molecular profiling of breast cancer
 
CRC CHP final PDF
CRC CHP final PDFCRC CHP final PDF
CRC CHP final PDF
 
Lombardi et al: XMRV/CFS Inflammatory Signature
Lombardi et al: XMRV/CFS Inflammatory SignatureLombardi et al: XMRV/CFS Inflammatory Signature
Lombardi et al: XMRV/CFS Inflammatory Signature
 
CCO Rational Options in Breast Cancer : How Molecular Understanding Informs T...
CCO Rational Options in Breast Cancer : How Molecular Understanding Informs T...CCO Rational Options in Breast Cancer : How Molecular Understanding Informs T...
CCO Rational Options in Breast Cancer : How Molecular Understanding Informs T...
 
2016_11_2_066-072
2016_11_2_066-0722016_11_2_066-072
2016_11_2_066-072
 
Genetic predipositio to cancer
Genetic predipositio to cancerGenetic predipositio to cancer
Genetic predipositio to cancer
 
Pró reitoria de pós-graduação e pesquisa programa de pós-graduação stricto se...
Pró reitoria de pós-graduação e pesquisa programa de pós-graduação stricto se...Pró reitoria de pós-graduação e pesquisa programa de pós-graduação stricto se...
Pró reitoria de pós-graduação e pesquisa programa de pós-graduação stricto se...
 
Case 1 & 2
Case 1 & 2Case 1 & 2
Case 1 & 2
 
Biología molecular
Biología molecularBiología molecular
Biología molecular
 
Molecular profiling in breast cancer
Molecular profiling in breast cancerMolecular profiling in breast cancer
Molecular profiling in breast cancer
 

Destaque

Sabay magz30
Sabay magz30Sabay magz30
Sabay magz30Barang CK
 
Phnom Penh Post Khmer
Phnom Penh Post KhmerPhnom Penh Post Khmer
Phnom Penh Post KhmerBarang CK
 
Sabay magz29
Sabay magz29Sabay magz29
Sabay magz29Barang CK
 
Sabay magz27
Sabay magz27Sabay magz27
Sabay magz27Barang CK
 
Keamanan internet dengan dns
Keamanan internet dengan dnsKeamanan internet dengan dns
Keamanan internet dengan dnsAinul Hakim
 
Sabay magz22
Sabay magz22Sabay magz22
Sabay magz22Barang CK
 
03 top 15-spectacular-bridges-and-viaducts-in-the-world-day
03 top 15-spectacular-bridges-and-viaducts-in-the-world-day03 top 15-spectacular-bridges-and-viaducts-in-the-world-day
03 top 15-spectacular-bridges-and-viaducts-in-the-world-day8888888anna
 

Destaque (9)

Figure 1
Figure 1Figure 1
Figure 1
 
Sabay magz30
Sabay magz30Sabay magz30
Sabay magz30
 
Phnom Penh Post Khmer
Phnom Penh Post KhmerPhnom Penh Post Khmer
Phnom Penh Post Khmer
 
Sabay magz29
Sabay magz29Sabay magz29
Sabay magz29
 
Sabay magz27
Sabay magz27Sabay magz27
Sabay magz27
 
Keamanan internet dengan dns
Keamanan internet dengan dnsKeamanan internet dengan dns
Keamanan internet dengan dns
 
Sabay magz22
Sabay magz22Sabay magz22
Sabay magz22
 
03 top 15-spectacular-bridges-and-viaducts-in-the-world-day
03 top 15-spectacular-bridges-and-viaducts-in-the-world-day03 top 15-spectacular-bridges-and-viaducts-in-the-world-day
03 top 15-spectacular-bridges-and-viaducts-in-the-world-day
 
Lecture 12
Lecture 12Lecture 12
Lecture 12
 

Semelhante a Cadher

Cancro del colon-retto: biotecnologie e prevenzione nell’eredità di Darwin e ...
Cancro del colon-retto: biotecnologie e prevenzione nell’eredità di Darwin e ...Cancro del colon-retto: biotecnologie e prevenzione nell’eredità di Darwin e ...
Cancro del colon-retto: biotecnologie e prevenzione nell’eredità di Darwin e ...Gentile Warschauer Emilio
 
Cancro del colon-retto: biotecnologie e prevenzione nell’eredità di Darwin e ...
Cancro del colon-retto: biotecnologie e prevenzione nell’eredità di Darwin e ...Cancro del colon-retto: biotecnologie e prevenzione nell’eredità di Darwin e ...
Cancro del colon-retto: biotecnologie e prevenzione nell’eredità di Darwin e ...Gentile Warschauer Emilio
 
Book left and rigth colon cancer gradient and local factors involved in c...
Book left and rigth colon cancer    gradient  and local factors involved in c...Book left and rigth colon cancer    gradient  and local factors involved in c...
Book left and rigth colon cancer gradient and local factors involved in c...M. Luisetto Pharm.D.Spec. Pharmacology
 
An Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch SyndromeAn Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch Syndromedaranisaha
 
An Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch SyndromeAn Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch SyndromeJohnJulie1
 
An Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch SyndromeAn Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch SyndromeAnonIshanvi
 
An Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch SyndromeAn Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch SyndromeEditorSara
 
An_Adrenal_Mass_in_a_Patient_with_Lynch.pdf
An_Adrenal_Mass_in_a_Patient_with_Lynch.pdfAn_Adrenal_Mass_in_a_Patient_with_Lynch.pdf
An_Adrenal_Mass_in_a_Patient_with_Lynch.pdfsemualkaira
 
An Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch SyndromeAn Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch SyndromeClinicsofOncology
 
An Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch SyndromeAn Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch Syndromesemualkaira
 
An Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch SyndromeAn Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch Syndromesemualkaira
 
An Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch SyndromeAn Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch SyndromeNainaAnon
 
13. Hyperplastic Polyposis Syndrome
13. Hyperplastic Polyposis Syndrome13. Hyperplastic Polyposis Syndrome
13. Hyperplastic Polyposis Syndromeensteve
 
CANCER DE COLON UNA DESCRIPCION Y REVISION DE LOS FACTORES DE RIESGO
CANCER DE COLON UNA DESCRIPCION Y REVISION DE LOS FACTORES DE RIESGOCANCER DE COLON UNA DESCRIPCION Y REVISION DE LOS FACTORES DE RIESGO
CANCER DE COLON UNA DESCRIPCION Y REVISION DE LOS FACTORES DE RIESGOCarlosRodrguezSantil
 
7.Cancer Genetics.Oct.09
7.Cancer Genetics.Oct.097.Cancer Genetics.Oct.09
7.Cancer Genetics.Oct.09ghalan
 
A 3′-untranslated region KRAS variant and triple-negative breast cancer: a ca...
A 3′-untranslated region KRAS variant and triple-negative breast cancer: a ca...A 3′-untranslated region KRAS variant and triple-negative breast cancer: a ca...
A 3′-untranslated region KRAS variant and triple-negative breast cancer: a ca...UCLA
 
Acs0515 Adenocarcinoma Of The Colon And Rectum 2005
Acs0515 Adenocarcinoma Of The Colon And Rectum 2005Acs0515 Adenocarcinoma Of The Colon And Rectum 2005
Acs0515 Adenocarcinoma Of The Colon And Rectum 2005medbookonline
 

Semelhante a Cadher (20)

Cancro del colon-retto: biotecnologie e prevenzione nell’eredità di Darwin e ...
Cancro del colon-retto: biotecnologie e prevenzione nell’eredità di Darwin e ...Cancro del colon-retto: biotecnologie e prevenzione nell’eredità di Darwin e ...
Cancro del colon-retto: biotecnologie e prevenzione nell’eredità di Darwin e ...
 
Cancro del colon-retto: biotecnologie e prevenzione nell’eredità di Darwin e ...
Cancro del colon-retto: biotecnologie e prevenzione nell’eredità di Darwin e ...Cancro del colon-retto: biotecnologie e prevenzione nell’eredità di Darwin e ...
Cancro del colon-retto: biotecnologie e prevenzione nell’eredità di Darwin e ...
 
Book left and rigth colon cancer gradient and local factors involved in c...
Book left and rigth colon cancer    gradient  and local factors involved in c...Book left and rigth colon cancer    gradient  and local factors involved in c...
Book left and rigth colon cancer gradient and local factors involved in c...
 
Brca acog 2017
Brca acog 2017Brca acog 2017
Brca acog 2017
 
An Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch SyndromeAn Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch Syndrome
 
An Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch SyndromeAn Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch Syndrome
 
An Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch SyndromeAn Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch Syndrome
 
An Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch SyndromeAn Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch Syndrome
 
An_Adrenal_Mass_in_a_Patient_with_Lynch.pdf
An_Adrenal_Mass_in_a_Patient_with_Lynch.pdfAn_Adrenal_Mass_in_a_Patient_with_Lynch.pdf
An_Adrenal_Mass_in_a_Patient_with_Lynch.pdf
 
An Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch SyndromeAn Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch Syndrome
 
An Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch SyndromeAn Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch Syndrome
 
An Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch SyndromeAn Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch Syndrome
 
An Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch SyndromeAn Adrenal Mass in a Patient with Lynch Syndrome
An Adrenal Mass in a Patient with Lynch Syndrome
 
13. Hyperplastic Polyposis Syndrome
13. Hyperplastic Polyposis Syndrome13. Hyperplastic Polyposis Syndrome
13. Hyperplastic Polyposis Syndrome
 
CANCER DE COLON UNA DESCRIPCION Y REVISION DE LOS FACTORES DE RIESGO
CANCER DE COLON UNA DESCRIPCION Y REVISION DE LOS FACTORES DE RIESGOCANCER DE COLON UNA DESCRIPCION Y REVISION DE LOS FACTORES DE RIESGO
CANCER DE COLON UNA DESCRIPCION Y REVISION DE LOS FACTORES DE RIESGO
 
CRC for 5th.
CRC for 5th.CRC for 5th.
CRC for 5th.
 
Cancer genetics
Cancer geneticsCancer genetics
Cancer genetics
 
7.Cancer Genetics.Oct.09
7.Cancer Genetics.Oct.097.Cancer Genetics.Oct.09
7.Cancer Genetics.Oct.09
 
A 3′-untranslated region KRAS variant and triple-negative breast cancer: a ca...
A 3′-untranslated region KRAS variant and triple-negative breast cancer: a ca...A 3′-untranslated region KRAS variant and triple-negative breast cancer: a ca...
A 3′-untranslated region KRAS variant and triple-negative breast cancer: a ca...
 
Acs0515 Adenocarcinoma Of The Colon And Rectum 2005
Acs0515 Adenocarcinoma Of The Colon And Rectum 2005Acs0515 Adenocarcinoma Of The Colon And Rectum 2005
Acs0515 Adenocarcinoma Of The Colon And Rectum 2005
 

Último

From Event to Action: Accelerate Your Decision Making with Real-Time Automation
From Event to Action: Accelerate Your Decision Making with Real-Time AutomationFrom Event to Action: Accelerate Your Decision Making with Real-Time Automation
From Event to Action: Accelerate Your Decision Making with Real-Time AutomationSafe Software
 
Breaking the Kubernetes Kill Chain: Host Path Mount
Breaking the Kubernetes Kill Chain: Host Path MountBreaking the Kubernetes Kill Chain: Host Path Mount
Breaking the Kubernetes Kill Chain: Host Path MountPuma Security, LLC
 
Mastering MySQL Database Architecture: Deep Dive into MySQL Shell and MySQL R...
Mastering MySQL Database Architecture: Deep Dive into MySQL Shell and MySQL R...Mastering MySQL Database Architecture: Deep Dive into MySQL Shell and MySQL R...
Mastering MySQL Database Architecture: Deep Dive into MySQL Shell and MySQL R...Miguel Araújo
 
Unblocking The Main Thread Solving ANRs and Frozen Frames
Unblocking The Main Thread Solving ANRs and Frozen FramesUnblocking The Main Thread Solving ANRs and Frozen Frames
Unblocking The Main Thread Solving ANRs and Frozen FramesSinan KOZAK
 
Presentation on how to chat with PDF using ChatGPT code interpreter
Presentation on how to chat with PDF using ChatGPT code interpreterPresentation on how to chat with PDF using ChatGPT code interpreter
Presentation on how to chat with PDF using ChatGPT code interpreternaman860154
 
Handwritten Text Recognition for manuscripts and early printed texts
Handwritten Text Recognition for manuscripts and early printed textsHandwritten Text Recognition for manuscripts and early printed texts
Handwritten Text Recognition for manuscripts and early printed textsMaria Levchenko
 
How to convert PDF to text with Nanonets
How to convert PDF to text with NanonetsHow to convert PDF to text with Nanonets
How to convert PDF to text with Nanonetsnaman860154
 
A Domino Admins Adventures (Engage 2024)
A Domino Admins Adventures (Engage 2024)A Domino Admins Adventures (Engage 2024)
A Domino Admins Adventures (Engage 2024)Gabriella Davis
 
Driving Behavioral Change for Information Management through Data-Driven Gree...
Driving Behavioral Change for Information Management through Data-Driven Gree...Driving Behavioral Change for Information Management through Data-Driven Gree...
Driving Behavioral Change for Information Management through Data-Driven Gree...Enterprise Knowledge
 
Workshop - Best of Both Worlds_ Combine KG and Vector search for enhanced R...
Workshop - Best of Both Worlds_ Combine  KG and Vector search for  enhanced R...Workshop - Best of Both Worlds_ Combine  KG and Vector search for  enhanced R...
Workshop - Best of Both Worlds_ Combine KG and Vector search for enhanced R...Neo4j
 
A Call to Action for Generative AI in 2024
A Call to Action for Generative AI in 2024A Call to Action for Generative AI in 2024
A Call to Action for Generative AI in 2024Results
 
08448380779 Call Girls In Greater Kailash - I Women Seeking Men
08448380779 Call Girls In Greater Kailash - I Women Seeking Men08448380779 Call Girls In Greater Kailash - I Women Seeking Men
08448380779 Call Girls In Greater Kailash - I Women Seeking MenDelhi Call girls
 
Partners Life - Insurer Innovation Award 2024
Partners Life - Insurer Innovation Award 2024Partners Life - Insurer Innovation Award 2024
Partners Life - Insurer Innovation Award 2024The Digital Insurer
 
Neo4j - How KGs are shaping the future of Generative AI at AWS Summit London ...
Neo4j - How KGs are shaping the future of Generative AI at AWS Summit London ...Neo4j - How KGs are shaping the future of Generative AI at AWS Summit London ...
Neo4j - How KGs are shaping the future of Generative AI at AWS Summit London ...Neo4j
 
Top 5 Benefits OF Using Muvi Live Paywall For Live Streams
Top 5 Benefits OF Using Muvi Live Paywall For Live StreamsTop 5 Benefits OF Using Muvi Live Paywall For Live Streams
Top 5 Benefits OF Using Muvi Live Paywall For Live StreamsRoshan Dwivedi
 
[2024]Digital Global Overview Report 2024 Meltwater.pdf
[2024]Digital Global Overview Report 2024 Meltwater.pdf[2024]Digital Global Overview Report 2024 Meltwater.pdf
[2024]Digital Global Overview Report 2024 Meltwater.pdfhans926745
 
Injustice - Developers Among Us (SciFiDevCon 2024)
Injustice - Developers Among Us (SciFiDevCon 2024)Injustice - Developers Among Us (SciFiDevCon 2024)
Injustice - Developers Among Us (SciFiDevCon 2024)Allon Mureinik
 
Data Cloud, More than a CDP by Matt Robison
Data Cloud, More than a CDP by Matt RobisonData Cloud, More than a CDP by Matt Robison
Data Cloud, More than a CDP by Matt RobisonAnna Loughnan Colquhoun
 
Strategies for Unlocking Knowledge Management in Microsoft 365 in the Copilot...
Strategies for Unlocking Knowledge Management in Microsoft 365 in the Copilot...Strategies for Unlocking Knowledge Management in Microsoft 365 in the Copilot...
Strategies for Unlocking Knowledge Management in Microsoft 365 in the Copilot...Drew Madelung
 
2024: Domino Containers - The Next Step. News from the Domino Container commu...
2024: Domino Containers - The Next Step. News from the Domino Container commu...2024: Domino Containers - The Next Step. News from the Domino Container commu...
2024: Domino Containers - The Next Step. News from the Domino Container commu...Martijn de Jong
 

Último (20)

From Event to Action: Accelerate Your Decision Making with Real-Time Automation
From Event to Action: Accelerate Your Decision Making with Real-Time AutomationFrom Event to Action: Accelerate Your Decision Making with Real-Time Automation
From Event to Action: Accelerate Your Decision Making with Real-Time Automation
 
Breaking the Kubernetes Kill Chain: Host Path Mount
Breaking the Kubernetes Kill Chain: Host Path MountBreaking the Kubernetes Kill Chain: Host Path Mount
Breaking the Kubernetes Kill Chain: Host Path Mount
 
Mastering MySQL Database Architecture: Deep Dive into MySQL Shell and MySQL R...
Mastering MySQL Database Architecture: Deep Dive into MySQL Shell and MySQL R...Mastering MySQL Database Architecture: Deep Dive into MySQL Shell and MySQL R...
Mastering MySQL Database Architecture: Deep Dive into MySQL Shell and MySQL R...
 
Unblocking The Main Thread Solving ANRs and Frozen Frames
Unblocking The Main Thread Solving ANRs and Frozen FramesUnblocking The Main Thread Solving ANRs and Frozen Frames
Unblocking The Main Thread Solving ANRs and Frozen Frames
 
Presentation on how to chat with PDF using ChatGPT code interpreter
Presentation on how to chat with PDF using ChatGPT code interpreterPresentation on how to chat with PDF using ChatGPT code interpreter
Presentation on how to chat with PDF using ChatGPT code interpreter
 
Handwritten Text Recognition for manuscripts and early printed texts
Handwritten Text Recognition for manuscripts and early printed textsHandwritten Text Recognition for manuscripts and early printed texts
Handwritten Text Recognition for manuscripts and early printed texts
 
How to convert PDF to text with Nanonets
How to convert PDF to text with NanonetsHow to convert PDF to text with Nanonets
How to convert PDF to text with Nanonets
 
A Domino Admins Adventures (Engage 2024)
A Domino Admins Adventures (Engage 2024)A Domino Admins Adventures (Engage 2024)
A Domino Admins Adventures (Engage 2024)
 
Driving Behavioral Change for Information Management through Data-Driven Gree...
Driving Behavioral Change for Information Management through Data-Driven Gree...Driving Behavioral Change for Information Management through Data-Driven Gree...
Driving Behavioral Change for Information Management through Data-Driven Gree...
 
Workshop - Best of Both Worlds_ Combine KG and Vector search for enhanced R...
Workshop - Best of Both Worlds_ Combine  KG and Vector search for  enhanced R...Workshop - Best of Both Worlds_ Combine  KG and Vector search for  enhanced R...
Workshop - Best of Both Worlds_ Combine KG and Vector search for enhanced R...
 
A Call to Action for Generative AI in 2024
A Call to Action for Generative AI in 2024A Call to Action for Generative AI in 2024
A Call to Action for Generative AI in 2024
 
08448380779 Call Girls In Greater Kailash - I Women Seeking Men
08448380779 Call Girls In Greater Kailash - I Women Seeking Men08448380779 Call Girls In Greater Kailash - I Women Seeking Men
08448380779 Call Girls In Greater Kailash - I Women Seeking Men
 
Partners Life - Insurer Innovation Award 2024
Partners Life - Insurer Innovation Award 2024Partners Life - Insurer Innovation Award 2024
Partners Life - Insurer Innovation Award 2024
 
Neo4j - How KGs are shaping the future of Generative AI at AWS Summit London ...
Neo4j - How KGs are shaping the future of Generative AI at AWS Summit London ...Neo4j - How KGs are shaping the future of Generative AI at AWS Summit London ...
Neo4j - How KGs are shaping the future of Generative AI at AWS Summit London ...
 
Top 5 Benefits OF Using Muvi Live Paywall For Live Streams
Top 5 Benefits OF Using Muvi Live Paywall For Live StreamsTop 5 Benefits OF Using Muvi Live Paywall For Live Streams
Top 5 Benefits OF Using Muvi Live Paywall For Live Streams
 
[2024]Digital Global Overview Report 2024 Meltwater.pdf
[2024]Digital Global Overview Report 2024 Meltwater.pdf[2024]Digital Global Overview Report 2024 Meltwater.pdf
[2024]Digital Global Overview Report 2024 Meltwater.pdf
 
Injustice - Developers Among Us (SciFiDevCon 2024)
Injustice - Developers Among Us (SciFiDevCon 2024)Injustice - Developers Among Us (SciFiDevCon 2024)
Injustice - Developers Among Us (SciFiDevCon 2024)
 
Data Cloud, More than a CDP by Matt Robison
Data Cloud, More than a CDP by Matt RobisonData Cloud, More than a CDP by Matt Robison
Data Cloud, More than a CDP by Matt Robison
 
Strategies for Unlocking Knowledge Management in Microsoft 365 in the Copilot...
Strategies for Unlocking Knowledge Management in Microsoft 365 in the Copilot...Strategies for Unlocking Knowledge Management in Microsoft 365 in the Copilot...
Strategies for Unlocking Knowledge Management in Microsoft 365 in the Copilot...
 
2024: Domino Containers - The Next Step. News from the Domino Container commu...
2024: Domino Containers - The Next Step. News from the Domino Container commu...2024: Domino Containers - The Next Step. News from the Domino Container commu...
2024: Domino Containers - The Next Step. News from the Domino Container commu...
 

Cadher

  • 1. Skip Navigation Oxford Journals Contact Us My Basket My Account HUMAN MOLECULAR GENETICS About This Journal Contact This Journal Subscriptions View Current Issue (Volume 21 Issue 2 January 15, 2012) Archive Search Institution: Benghazi University Sign In as Personal Subscriber Oxford Journals Life Sciences & Medicine Human Molecular Genetics Volume13, Issuesuppl 2 Pp. R177-R185. o Zoe Kemp, o Cristina Thirlwell, o Oliver Sieber, o Andrew Silver, o and Ian Tomlinson An update on the genetics of colorectal cancer Hum. Mol. Genet. (2004) 13(suppl 2): R177-R185 doi:10.1093/hmg/ddh247 1. Hum. Mol. Genet. (2004) 13 (suppl 2): R177-R185. doi: 10.1093/hmg/ddh247 An update on the genetics of colorectal cancer Next Section Abstract Colorectal carcinoma (CRC) remains a frequent cause of cancer-associated mortality in the UK and still has a relatively poor outcome. Single gene defects account for up to 2–6% of cases, but twin studies suggest a hereditary component in 35%. CRC represents a paradigm for cancer genetics. Almost all the major-gene influences on CRC have been identified, and the identification of the remaining susceptibility alleles is proving troublesome. Only a few low-penetrance alleles, such as methylene tetrahydrofolate reductase C677T, appear convincingly to be associated with CRC risk. To identify the remaining CRC genes, parallel approaches, including strategies based on linkage and association and complementary analyses such as searches for modifier genes, must be
  • 2. employed. To gain sufficient evidence to prove that a gene is involved in CRC predisposition, it is probably necessary for multiple, adequately-powered studies to demonstrate an association with the disease, especially if the allelic variants have only a small differential effect on risk. It may also be possible to show how genes interact with each other and the environment, although this will be even more difficult. Accurate quantitation of the allele-specific risks in different populations will be necessary, but problematic, especially if those risks combine in a fashion which is not of a straightforward additive or multiplicative type. Without any good prior evidence of the nature of the remaining genetic influence on CRC, the possibility remains that this is a truly polygenic trait or that multiple, rare variants contribute to the increased risk; in these cases, identification of the genes involved will be very difficult. Despite these potential problems, the effectiveness of preventive measures for CRC, especially in high- risk individuals, means that the search for new predisposition genes is justified. Previous SectionNext Section Colorectal cancer (CRC) is the third most common cause of cancer-related death in the western world. The annual incidence in the United Kingdom is 35 000, and worldwide it has been estimated to be at least half a million. There exists a number of syndromes with Mendelian dominant inheritance in which there is a primary predisposition to benign or malignant tumours of the large bowel (Table 1). Familial adenomatous polyposis (FAP) patients inherit a mutated copy of the adenomatous polyposis gene ( APC), whereas hereditary non-polyposis colon cancer (HNPCC) is caused by inheritance of defective DNA mismatch repair genes (MLH1, MSH2, PMS2 and MSH6). Germline mutations of the LKB1/STK11 gene have been shown to cause the Peutz–Jeghers syndrome (PJS), and mutation of SMAD4 or ALK3 underly juvenile polyposis (JPS). In contrast, the MYH- associated polyposis (MAP) syndrome has autosomal recessive inheritance and results from bi-allelic mutations in the MYH gene. View this table: In this window In a new window Table 1. Genes responsible for known polyposis and CRC syndromes Taken together, these syndromes account only for ∼2–6% of CRC cases. The great majority of CRCs do not have a recognizable inherited cause, but a number of studies have suggested a role for genetic factors in predisposition to a substantial minority of colorectal tumours. The relatives of patients with ‘sporadic’ CRC are themselves at increased risk of the disease and segregation analysis has suggested dominant inheritance of the uncharacterized susceptibility genes (1–3). The relative risk in siblings of affected individuals is 2–3-fold increased for both colorectal adenomas and CRC, the risk being higher in relatives of patients diagnosed young, and in those with two or more affected family members. In addition, an extensive analysis of twins has suggested that 35% of CRCs arise through inherited factors (4). Although some high and moderate penetrance genes are probably undiscovered, inherited predisposition to CRC in humans is more likely to be the result of low-penetrance alleles at several or many genetic loci. In support of this contention, experimental data from rodent models have demonstrated
  • 3. that numerous genes with relatively small effects control susceptibility to cancer including CRC (reviewed in 5). Furthermore, hereditary factors may influence the severity of the disease in the Mendelian CRC syndromes, as exemplified by the number of colorectal polyps presenting within FAP families or in individuals with identical germline APC mutations (6). There are two factors that, in combination, make the search for additional CRC predisposition genes so important. First, identification and removal of colorectal adenomas (and possibly other types of bowel polyp) almost certainly greatly reduce cancer incidence; identification of cancers before they present clinically may have similar benefits. Second, regular, frequent and effective screening for colorectal tumours is not currently available at the population level. The identification of controlling genetic factors therefore allows individual cancer risk assessment and the targeting of screening to those at highest risk; there may also be spin-offs in terms of effective chemoprevention and the development of new therapies. The challenge, therefore, is to identify all the relevant genes, and to specify their contribution in controlling CRC predisposition and severity in human populations. A major part of this challenge is to choose the most appropriate methods to locate and identify novel susceptibility genes. In this short review, we shall detail some of the recent developments in the study of inherited susceptibility to CRC. Progress has been made in the understanding of the Mendelian syndromes and towards the identification of the small number of remaining high-penetrance CRC genes. We shall also present recent evidence for low-penetrance CRC alleles and for modifier genes of the Mendelian syndromes' phenotypes. We complete our review by outlining future strategies for use in the continued search for genes predisposing to CRC. Previous SectionNext Section PROGRESS IN UNDERSTANDING THE HEREDITARY COLORECTAL TUMOUR SYNDROMES MYH-associated polyposis MYH is a DNA glycosylase involved in the response to oxidative damage of DNA by base excision repair (BER). MYH was identified as a recessive CRC and adenoma predisposition gene by Al-Tassan et al. (7), who found an unusual somatic APC mutation spectrum (mostly G→T changes) in the polyps of a family that they were investigating for the pathogenic effects of the APC missense variant E1317Q. The MAP phenotype generally resembles attenuated (<100 adenomas) or mild, classical (100–1000 adenomas) FAP (8), although MAP tumours seem to be confined to the gastrointestinal tract. MAP individuals with early-onset CRC but no detected colorectal adenomas have been reported (9), although small adenomas can be missed using colonoscopy without dye-spray. MAP accounts for about 1% of all CRCs and one-third of patients with 15–100 colorectal adenomas (8,10). MYH screening should be considered in patients with multiple colorectal adenomas, although APC remains as a much stronger candidate in families with vertical transmission of disease. Testing MYH in patients with early-onset CRC and no adenomas would be justified if further data support those of Wang et al. (9). Mutation testing should be tailored to the ethnic group, with mutations Y165C and G382D more common in Caucasians, Y90X in Pakistanis, E466X in Indians and nt1395delGGA in those of Mediterranean origin (10–13). The site-specific nature of MAP tumours remains
  • 4. puzzling. BER deficiency does not specifically target APC and many MAP tumours harbour specific G→T changes in K-ras (14). The few MAP cancers studied so far are microsatellite-stable and near-diploid (14). AXIN2 Apart from APC, studies have generally failed to find germline mutations in other components of the Wnt pathway (such as beta-catenin, APC2, axin, conductin (axin2) and glycogen synthase kinase 3-beta) in patients with multiple or early-onset colorectal tumours (15). Recently, however, a Finnish family with dominantly-transmitted oligodontia and colorectal polyps was shown to carry a germline R656X mutation in AXIN2 (16). The colorectal phenotype in the family was highly variable, ranging from 68 adenomas through 10 hyperplastic polyps to no disease. Loss of the wild-type AXIN2 allele was not tested in the polyps. Peutz–Jeghers syndrome Germline mutations of the serine/threonine kinase gene LKB1(STK11) cause PJS. PJS patients develop harmatomatous polyps of the gastrointestinal tract, and characteristic freckling, and are at increased risk of colorectal and other cancers. LKB1 has been reported to function in a variety of pathways, including those involving mTOR/AMP- activated protein kinase, Wnt, COX2, NF-kappaB, VEGF, cell polarity and p53 (17–25). It remains to be seen whether mutations in such genes play a role in PJS. From a strictly genetic perspective, there is ongoing controversy as to the existence of a second PJS locus, primarily because LKB1 mutations cannot be found in 30–80% of PJS patients. PJS kindreds unlinked to LKB1 (26) and some linked to 19q13 (27) have been reported, but no second locus has been identified. In this regard, it should be noted that, although PJS polyps are probably pathognomic, several conditions have pigmented lesions that may mimic PJS (28). Hereditary non-polyposis CRC Hereditary non-polyposis colon cancer (HNPCC) is an autosomal dominant disorder characterized by early-onset CRC (often right-sided), few adenomas and specific extra- colonic cancers, including those of the ovary, stomach, small bowel and uroepithelial tract. HNPCC accounts for 1–5% of CRCs and results from germline mutations in MLH1 and MSH2 in about 90% of cases and more rarely in PMS2 and MSH6 (remaining 10% of cases). HNPCC cancers are characterized by microsatellite instability (MSI). The MMR genes are not mutated in sporadic CRCs, although MLH1 is silenced by promoter methylation in 10–15% of these tumours, most of which are right-sided and tend to occur in older females. In the light of these data, Suter et al. (29) hypothesized that methylation of the MLH1 promoter could occur in the germline and act as an alternative to mutation in predisposing to HNPCC. They found evidence of such a MLH1 ‘epimutation’ in two patients with multiple, MSI+ primary tumours which showed mismatch repair deficiency. The epimutation occurred as a mosaic and was also present in spermatozoa of one of the individuals, indicating a germline defect and the potential for transmission to offspring. It was not clear whether an uncharacterized, primary genetic defect had led to the epimutation. Other workers have focussed on the role of additional MMR genes in the HNPCC families, with evidence that MLH3 (30–32) and EXO1 (33–36) might be involved in a small number of cases, although data are inconsistent. There is also accumulating evidence to show that the genetic pathways of sporadic, MSI+ CRCs and HNPCC CRCs are non-identical. Johnson et al. (37), for example, showed that beta-catenin mutations were
  • 5. specific to MSI+ carcinomas from HNPCC, but were rare in the HNPCC adenomas, suggesting that Wnt activation was not always an early event in HNPCC tumorigenesis. Two further issues the functions of MMR proteins outside simple DNA repair (38) and whether or not MMR loss initiates tumorigenesis in HNPCC, remain under close scrutiny. Familial adenomatous polyposis and APC modifier genes FAP is caused by germline APC mutations. Affected individuals develop hundreds to thousands of adenomatous colorectal polyps during the second and third decades, progressing to cancer in nearly 100% of cases without treatment. Specific extra-colonic features exist (39). We shall focus on three aspects of recent FAP research. First, it is clear, in both FAP and sporadic CRCs, that APC is not a simple tumour suppressor gene: the ‘two hits’ are strongly associated in terms of their location within the gene (39–42). This association probably provides an optimal level of Wnt signalling for tumour growth, mediated through beta-catenin levels. The mutation spectrum, and presumably optimal level of Wnt signalling, varies among tissues (40,43). The ‘first hit–second hit’ association at APC has also provided a new mechanism for determining disease severity (40), given that certain ‘second hits’ such as allelic loss probably occur spontaneously at a relatively high frequency and are only selected in certain FAP patients (with mutations close to codon 1300), who consequently have severe disease. Second, clues have emerged as to the cause of attenuated disease (AFAP, AAPC) in individuals with germline APC mutations in exons 1–4, 9 and the latter half of exon 15, owing to the discovery of ‘third hits’ in the tumours of some of these patients (44). In order to explain the paradox that a requirement for ‘third hits’ should in theory cause AFAP patients to have no more tumours than the general population, Su et al. (45) suggested that a broader spectrum of somatic mutations was selected in AFAP individuals than in those with no germline APC mutation (although their model was not placed in the context of the ‘first hit–second hit’ associations described earlier). Third, the site of the germline APC mutations cannot explain all the phenotypic variation which is seen in FAP and AFAP. It is likely that modifier genes distinct from APC influence features such as the number of tumours in each region of the gastrointestinal tract (46), the development of desmoids (47) and progression to cancer. In ApcMin mice, a modifier gene, Mom1/Pla2g2a, influences polyp number (48–50). FAP modifier genes in humans are likely to be common polymorphisms, and candidate gene approaches have been used to analyse these. One study has suggested effects of polymorphisms in the N-acetyltransferases (NAT1 and NAT2) on polyp numbers in FAP (51). APC I1307K The APC I1307K variant is present in about 6% of Ashkenazi Jews, but is much rarer in those of other ethnic groups. I1307K creates an A8 tract that appears to be somatically unstable, leading to frameshift mutations (52). Originally, I1307K was proposed to lead to an AFAP-like phenotype, although extra-colonic disease does not appear to feature in this group. Recent data have shown that APC I1307K only confers a small excess risk of CRC (53), inconsistent prima facie with an AFAP-like phenotype. Moreover, only a minority of colorectal tumours from I1307K carriers shows slippage of the A 8 tract. It may be the case, therefore, that although the 1307K allele is somewhat unstable, this effect is subtle in most carriers; only some individuals, perhaps with an unidentified, additional germline factor, go on to develop an AFAP-like phenotype (54). Previous SectionNext Section
  • 6. RECENT DEVELOPMENTS THROUGH LINKAGE ANALYSIS Hereditary mixed polyposis syndrome Hereditary mixed polyposis syndrome (HMPS) was described as a distinct syndrome of colorectal adenomas, hyperplastic polyps, atypical ‘mixed’ polyps and carcinoma by Whitelaw et al. (55) in a large Ashkenazi family. The extra-colonic features of FAP and the typical features of PJS were absent. The phenotype had limited similarity to JPS. Linkage of HMPS to chromosome 6q16–q21 was demonstrated by Thomas et al. (56). Tomlinson et al. (57) showed linkage to chromosome 15q14–q15 (CRAC1 locus) in another Ashkenazi family with multiple colorectal adenomas (including serrated lesions) and CRC. The development of multiple adenomas in a HMPS individual without the disease- associated haplotype led to the reassignment and stricter implementation of affection status in that family in order to minimize the phenocopy rate. Subsequent linkage analysis showed HMPS and CRAC1 to be one-and-the-same (58). Affected individuals in these families and in several other Ashkenazi kindreds with a similar phenotype were shown to share an ancestral haplotype between D15S1030 and D15S118. The gene itself is, as yet, unidentified and its influence in other population groups, undetermined. A CRC locus on chromosome 9q? Wiesner et al. (59) undertook linkage analysis using the affected sibling pair method in 53 families with individuals affected at <65 years of age by CRC or adenomas (>1 cm or displaying high-grade dysplasia). Six potential loci were identified with excess allele sharing among concordantly affected sib-pairs (P<0.016). Of these, a region on chromosome 9 gave a significant result when additionally examined for deficient allele sharing among discordant sib pairs (P=0.014). The Haseman–Elston regression method provided an overall probability of linkage of P=0.00045. Previous SectionNext Section EVIDENCE FOR LOW-PENETRANCE ALLELES ASSOCIATED WITH AN INCREASED RISK OF CRC Some high- or moderate-penetrance CRC predisposition genes remain to be detected and their pursuit is of great importance. It is generally believed, nevertheless, that low- penetrance variants, such as common alleles at SNPs, are responsible for much of the uncharacterized inherited influence on CRC. Many different SNPs and other common polymorphisms have been evaluated for their effects on CRC risk, and space precludes detailed descriptions or analysis here. Case–control (association) analysis has been used most frequently. Such studies have often been criticized, some of the reasons being the following: sample sizes produce insufficient statistical power; cases and controls are inappropriately chosen; statistical thresholds are chosen with insufficient rigour, so that most reported associations are inevitably false (60); the ‘multiple testing’ problem and tendency to inappropriate sub-group analyses are endemic;
  • 7. publication bias remains problematic, even though methods have been developed to assess this (61,62); studies rarely attempt to replicate their findings in the same population; findings which are not replicated in different populations may be inappropriately dismissed; the functional effects of variants have often not been clearly shown; some methods, such as PCR–RFLP analysis, are prone to systematic error unless control measures are carefully applied. The recognition of these problems has arguably led to too pessimistic an attitude to studies of low-penetrance variants as CRC susceptibility alleles. Most recent studies have used larger, better defined populations of cases and controls, together with genotyping methods with greater accuracy and throughput. Formal assessments of the roles of several polymorphisms in CRC predisposition were made by Houlston and Tomlinson (63) and by de Jong et al. (64). In summary, there is already good evidence to show that a small number of polymorphisms is associated with a differential risk of CRC (Table 2). These include methylene tetrahydrofolate reductase MTHFRC677T, H-ras VNTR rare alleles and NAT2 alleles which cause a rapid acetylator phenotype. Other polymorphisms have been tested (Table 2) and many are unproven, although in a small number of cases, loci continue to be cited as being associated with CRC risk despite the evidence in their favour being minimal. View this table: In this window In a new window Table 2. Summary of polymorphisms tested for association with differential CRC risk Recent data have suggested that loss of imprinting (LOI) of the insulin-like growth factor II (IGF2) in normal colonic mucosa and/or lymphocytes is indicative of increased CRC risk (89–92). It has been suggested that IGF2 LOI is a genetically determined trait. It is not clear whether the association between IGF2 LOI and CRC might be a direct effect of increased IGF2 levels, or a marker of a general tendency to, for example, hypomethylation or aberrant gene silencing. Studies are increasingly testing risks associated with combinations of putative CRC genes, or of genes and specific environments, although inherent problems of statistical power mean that most of the reported associations of this type must currently be taken as no more than suggestive. Even for the MTHFR polymorphisms, advice on dietary modification so that folate intake is increased cannot yet be given reliably or with confidence. A further question is whether or not a true CRC risk factor should affect the risk of both rectal and colonic cancers, conditions that are usually treated as essentially the same disease (albeit with some different molecular and clinical features) by geneticists, but as different conditions by epidemiologists. Further studies are beginning to test the influence of germline variation on factors other than disease susceptibility,
  • 8. including the presenting features of CRCs, response to therapy and toxicity (including pharmacogenetic analyses), and recurrence/survival. In general, acceptance of a polymorphism as a CRC risk factor can only come through repeated analysis by different studies and by consequent slow progress to a consensus judgement. Accurate quantitation of the associated risk will be just as important as its identification in the first place, and this will require multiple studies in individual populations which may differ in genetic background, environment and availability of screening and medical care. Quantitation of risks associated with variants at multiple predisposition loci will be even more difficult, especially if risks do not combine in a simple additive or multiplicative manner, yet if testing for low-penetrance alleles is to come into routine clinical practice, such risks must be determined. Without any good prior evidence of the nature of the remaining genetic influence on CRC, the possibility remains that it is a truly polygenic trait in which risks associated with any single polymorphism are so low that identification of the genes involved (and their testing in clinical practice) will be very difficult. This is, perhaps, the worst case scenario. An alternative is that multiple, rare variants may contribute to the increased familial risk of CRC: in this case, identification of the variants will be possible through mutation screening methods, although evaluation of their functional importance will be time- consuming. The case of the sub-polymorphic variant APCE1317Q provides a case in point (93–97); it currently seems likely that this variant, which was discovered by mutation screening, has little functional effect, although ‘closing the door’ on such a variant is not easy. Nevertheless, the modestly increased risk of breast cancer associated with the CHEK2del1100C sub-polymorphic variant (98) shows that important alleles of this type do exist. (Any association between CHEK2del1100C and CRC remains controversial.) Should many of the remaining CRC alleles prove to be of the low-frequency/low-risk type, genetic testing in the clinical setting may not be worthwhile. Despite these potential problems, however, the effectiveness of preventive measures for CRC, especially in high- risk individuals, means that the search for new predisposition genes is justified. Previous SectionNext Section FUTURE STRATEGIES FOR IDENTIFYING CRC PREDISPOSITION GENES The successful detection of predisposition genes for cancer has largely been limited to high-penetrance genes such as APC, BRCA1/2 and CDNK2A. Although the search for low-penetrance genes has been informative in some other complex diseases, for example, APOE-4 in Alzheimer's disease, those underlying cancer remain largely elusive. The characteristics of cancer create obstacles. The usual late onset of CRC means parents may be deceased and the disease is often fatal, making sample collection difficult. In addition, the requirement of random somatic mutations for the progression to carcinoma may cloud the picture. However, some of these problems can be circumvented by the selection of early-onset cases to increase the genetic component and to reduce the environmental component, and by the study of benign, precursor lesions. The question therefore arises of which methods to use to detect novel cancer genes. In our view, given that there are undoubtedly still high and moderate penetrance CRC genes yet to be discovered, linkage analysis should certainly not be abandoned. The latter would require the use of non-parametric methodology in view of genetic heterogeneity
  • 9. and the existence of phenocopies. However, even for moderate penetrance genes, the numbers of multi-case families required to yield results are large. For example, under a dominant model with a gene frequency of 0.1 and a risk ratio of 6, about 500 affected sib pairs or 20 affected sib trios would be needed to give a LOD score of 3.3 (99). The increasing attention being given to association studies to identify CRC genes is therefore justified. For CRC, these have previously been limited to examining the frequency of alleles at candidate loci in cases and controls. However, the advent of SNP maps has created the potential for genome-wide association (100). The power of these studies, especially for rare alleles can be increased by using cases with a family history of CRC (101) and by selecting in other ways for ‘genetic’ disease (for example, multiple tumours, cancers of more than one site or early age of presentation). The detection of a dominant gene with a frequency of 0.1 and a relative risk of 4 would require 200 cases and 200 controls in an association study, but over 1000 affected sib pairs in a traditional linkage analysis (99). A more controversial area in the optimal design of association studies revolves around the question of how many SNPs to use and whether the approach should be haplotype- map or sequence based. Public and private SNP discovery projects report the identification of 1.4 and 2.1 million SNPs, respectively, although the total is estimated (102) to be about 15 million in reality. Obviously only a small proportion of these could currently be genotyped in any association study. A map-based approach depends on linkage disequilibrium occurring between blocks of haplotypes within which only a few common haplotypes are observed and there is little evidence of historical recombination. This allows the genome to be represented by a carefully chosen group of haplotype tag SNPs (htSNPs) (103). The number of SNPs required will increase with the genetic diversity of a population because of shorter haplotype blocks. For example, the International HapMap Project has recently decided to add 2.25 million SNPs to the original 600 000 for each of three population maps, owing to the longer evolutionary history of the Yuroban (Nigerian) population (88). This approach will mainly detect alleles with a frequency of ≥5%. Advocates of a sequence-based approach argue that SNPs should be chosen on the basis of their probable contribution to the phenotype. The focus of this approach is therefore on coding regions and SNPs resulting in amino acid changes and/or SNPs found in evolutionarily conserved areas. This would require far fewer SNPs (50 000–100 000) for a genome-wide screen and would potentially detect lower frequency alleles (1–20%). Botstein and Risch (102) have suggested that, at least initially, a ‘sequence-based’ approach on a selected 5% of the genome might provide the power to detect some of the CRC-associated variants. For identifying sub-polymorphic variants associated with disease, a mutation screening strategy based on candidate genes (and followed if possible by a suitable association study) is probably the only practical method for the short term. It remains to be seen whether or not new technologies such as re-sequencing microarrays prove cost-effective for research and diagnosis here. The requirement for functional studies will be particularly important for sub-polymorphic variants, especially those which lead to conserved amino acid changes, or affect splice sites, promoters or control regions. Although it is hoped that linkage, association and mutation screening studies will yield answers, the concurrent use of more indirect methods is prudent and may contribute to
  • 10. the fine mapping of an identified region of interest. As with SMAD4 and more recently, MYH, the search for somatic mutations may also identify the site of germline mutations, as their effects in the cell are functionally similar. Other approaches include the study of cancer-associated phenotypes including normal variation (for example in hormone levels). Inflammatory bowel disease, which we have not discussed in detail earlier, is associated with an increase risk of CRC yet has a higher sibling relative risk (104), and some of the predisposition genes might be the same for the two conditions. The search for modifier genes is also potentially important, as a gene which influences the severity of a Mendelian syndrome should also, in principle, act as a low-penetrance predisposition gene. Intensive, effective population-based screening for CRC and its precursor lesions is likely in the foreseeable future to be expensive and/or to have a relatively low take-up rate. Even if this screening for CRC becomes more acceptable, it will optimally be targeted to those at highest risk. The identification of CRC risk genes will permit targeted screening, and will open up possibilities for other preventive measures and, perhaps, therapies. It is highly likely that at least some CRC genes can be identified and will provide useful targets for routine genetic testing and preventive intervention. Previous SectionNext Section ACKNOWLEDGEMENTS We are grateful to colleagues, particularly Richard Houlston and Walter Bodmer, for discussion and help. 1. Zoe Kemp1, 2. Cristina Thirlwell1, 3. Oliver Sieber1, 4. Andrew Silver2 and 5. Ian Tomlinson1,2,* + Author Affiliations 1. Molecular and Population Genetics Laboratory, London Research Institute, 1 Cancer Research UK, 44 Lincoln's Inn Fields, London WC2A 3PX, UK and 2. 2Colorectal Cancer Unit, Cancer Research UK, St Mark's Hospital, Watford Road, Harrow HA1 3UJ, UK 1. To whom correspondence should be addressed. Tel: +44 1712692884; Fax: +44 * 1712692885; Email: i.tomlinson@cancer.org.uk Received July 23, 2004. Accepted July 27, 2004. Human Molecular Genetics, Vol. 13, Review Issue 2 © Oxford University Press 2004; all rights reserved Previous Section References
  • 11. 1. ↵ Cannon-Albright, L.A., Skolnick, M.H., Bishop, D.T., Lee, R.G. and Burt, R.W. (1988) Common inheritance of susceptibility to colonic adenomatous polyps and associated colorectal cancers. N. Engl. J. Med., 319, 533–537. MedlineWeb of Science 2. Houlston, R.S., Collins, A., Slack, J. and Morton, N.E. (1992) Dominant genes for colorectal cancer are not rare. Ann. Hum. Genet., 56 (Pt 2), 99–103. MedlineWeb of Science 3. ↵ Johns, L.E. and Houlston, R.S. (2001) A systematic review and meta- analysis of familial colorectal cancer risk. Am. J. Gasrtoenterol., 96, 2992–3003. 4. ↵ Lichtenstein, P., Holm, N.V., Verkasalo, P.K., Iliadou, A., Kaprio, J., Koskenvuo, M., Pukkala, E., Skytthe, A. and Hemminki, K. (2000) Environmental and heritable factors in the causation of cancer— analyses of cohorts of twins from Sweden, Denmark, and Finland. N. Engl. J. Med., 343, 78–85. CrossRefMedlineWeb of Science 5. ↵ Demant, P. (2003) Cancer susceptibility in the mouse: genetics, biology and implications for human cancer. Nat. Rev. Genet., 4, 721– 734. MedlineWeb of Science 6. ↵ Crabtree, M.D., Tomlinson, I.P., Hodgson, S.V., Neale, K., Phillips, R.K. and Houlston, R.S. (2002) Explaining variation in familial adenomatous polyposis: relationship between genotype and phenotype and evidence for modifier genes. Gut, 51, 420–423. Abstract/FREE Full Text 7. ↵ Al-Tassan, N., Chmiel, N.H., Maynard, J., Fleming, N., Livingston, A.L., Williams, G.T., Hodges, A.K., Davies, D.R., David, S.S., Sampson, J.R. et al. (2002) Inherited variants of MYH associated with somatic G:C→T:A mutations in colorectal tumors. Nat. Genet., 30, 227–232. CrossRefMedlineWeb of Science 8. ↵ Sieber, O.M., Lipton, L., Crabtree, M., Heinimann, K., Fidalgo, P., Phillips, R.K., Bisgaard, M.L., Orntoft, T.F., Aaltonen, L.A., Hodgson, S.V. et al. (2003) Multiple colorectal adenomas, classic adenomatous
  • 12. polyposis, and germ-line mutations in MYH. N. Engl. J. Med., 348, 791–799. CrossRefMedlineWeb of Science 9. ↵ Wang, L., Baudhuin, L.M., Boardman, L.A., Steenblock, K.J., Petersen, G.M., Halling, K.C., French, A.J., Johnson, R.A., Burgart, L.J., Rabe, K. et al. (2004) MYH mutations in patients with attenuated and classic polyposis and with young-onset colorectal cancer without polyps. Gastroenterology, 127, 9–16. CrossRefMedlineWeb of Science 10. ↵ Halford, S.E., Rowan, A.J., Lipton, L., Sieber, O.M., Pack, K., Thomas, H.J., Hodgson, S.V., Bodmer, W.F. and Tomlinson, I.P. (2003) Germline mutations but not somatic changes at the MYH locus contribute to the pathogenesis of unselected colorectal cancers. Am. J. Pathol., 162, 1545–1548. MedlineWeb of Science 11. Jones, S., Emmerson, P., Maynard, J., Best, J.M., Jordan, S., Williams, G.T., Sampson, J.R. and Cheadle, J.P. (2002) Biallelic germline mutations in MYH predispose to multiple colorectal adenoma and somatic G:C→T:A mutations. Hum. Mol. Genet., 11, 2961–2967. Abstract/FREE Full Text 12. Enholm, S., Hienonen, T., Suomalainen, A., Lipton, L., Tomlinson, I., Karja, V., Eskelinen, M., Mecklin, J.P., Karhu, A., Jarvinen, H.J. et al. (2003) Proportion and phenotype of MYH-associated colorectal neoplasia in a population-based series of Finnish colorectal cancer patients. Am. J. Pathol., 163, 827–832. MedlineWeb of Science 13. ↵ Gismondi, V., Meta, M., Bonelli, L., Radice, P., Sala, P., Bertario, L., Viel, A., Fornasarig, M., Arrigoni, A., Gentile, M. et al. (2004) Prevalence of the Y165C, G382D and 1395delGGA germline mutations of the MYH gene in Italian patients with adenomatous polyposis coli and colorectal adenomas. Int. J. Cancer, 109, 680–684. CrossRefMedlineWeb of Science 14. ↵ Lipton, L., Halford, S.E., Johnson, V., Novelli, M.R., Jones, A., Cummings, C., Barclay, E., Sieber, O., Sadat, A., Bisgaard, M.L. et al. (2003) Carcinogenesis in MYH-associated polyposis follows a distinct genetic pathway. Cancer Res., 63, 7595–7599. Abstract/FREE Full Text 15. ↵
  • 13. Lipton, L., Sieber, O.M., Thomas, H.J., Hodgson, S.V., Tomlinson, I.P. and Woodford-Richens, K. (2003) Germline mutations in the TGF- beta and Wnt signalling pathways are a rare cause of the ‘multiple’ adenoma phenotype. J. Med. Genet., 40, e35. FREE Full Text 16. ↵ Lammi, L., Arte, S., Somer, M., Jarvinen, H., Lahermo, P., Thesleff, I., Pirinen, S. and Nieminen, P. (2004) Mutations in AXIN2 cause familial tooth agenesis and predispose to colorectal cancer. Am. J. Hum. Genet., 74, 1043–1050. CrossRefMedlineWeb of Science 17. ↵ De Leng, W.W., Westerman, A.M., Weterman, M.A., De Rooij, F.W., Dekken Hv, H., De Goeij, A.F., Gruber, S.B., Wilson, J.H., Offerhaus, G.J., Giardiello, F.M. et al. (2003) Cyclooxygenase 2 expression and molecular alterations in Peutz–Jeghers hamartomas and carcinomas. Clin. Cancer Res., 9, 3065–3072. Abstract/FREE Full Text 18. Liu, W.K., Chien, C.Y., Chou, C.K. and Su, J.Y. (2003) An LKB1- interacting protein negatively regulates TNFalpha-induced NF- kappaB activation. J. Biomed. Sci., 10, 242–252. MedlineWeb of Science 19. Martin, S.G. and St Johnston, D. (2003) A role for Drosophila LKB1 in anterior–posterior axis formation and epithelial polarity. Nature, 421, 379–384. CrossRefMedline 20. Ossipova, O., Bardeesy, N., DePinho, R.A. and Green, J.B. (2003) LKB1 (XEEK1) regulates Wnt signalling in vertebrate development. Nat. Cell Biol., 5, 889–894. CrossRefMedlineWeb of Science 21. Baas, A.F., Smit, L. and Clevers, H. (2004) LKB1 tumor suppressor protein: PARtaker in cell polarity. Trends Cell. Biol., 14, 312–319. CrossRefMedlineWeb of Science 22. Baas, A.F., Kuipers, J., van der Wel, N.N., Batlle, E., Koerten, H.K., Peters, P.J. and Clevers, H.C. (2004) Complete polarization of single intestinal epithelial cells upon activation of LKB1 by STRAD. Cell, 116, 457–466. CrossRefMedlineWeb of Science 23. Lizcano, J.M., Goransson, O., Toth, R., Deak, M., Morrice, N.A., Boudeau, J., Hawley, S.A., Udd, L., Makela, T.P., Hardie, D.G. et al. (2004) LKB1 is a master kinase that activates 13 kinases of the AMPK subfamily, including MARK/PAR-1. EMBO J., 23, 833–843. CrossRefMedlineWeb of Science
  • 14. 24. Shaw, R.J., Bardeesy, N., Manning, B.D., Lopez, L., Kosmatka, M., DePinho, R.A. and Cantley, L.C. (2004) The LKB1 tumor suppressor negatively regulates mTOR signaling. Cancer Cell, 6, 91–99. CrossRefMedlineWeb of Science 25. ↵ Spicer, J. and Ashworth, A. (2004) LKB1 kinase: master and commander of metabolism and polarity. Curr. Biol., 14, R383–R385. CrossRefMedlineWeb of Science 26. ↵ Olschwang, S., Markie, D., Seal, S., Neale, K., Phillips, R., Cottrell, S., Ellis, I., Hodgson, S., Zauber, P., Spigelman, A. et al. (1998) Peutz– Jeghers disease: most, but not all, families are compatible with linkage to 19p13.3. J. Med. Genet., 35, 42–44. Abstract/FREE Full Text 27. ↵ Buchet-Poyau, K., Mehenni, H., Radhakrishna, U. and Antonarakis, S.E. (2002) Search for the second Peutz–Jeghers syndrome locus: exclusion of the STK13, PRKCG, KLK10, and PSCD2 genes on chromosome 19 and the STK11IP gene on chromosome 2. Cytogenet. Genome Res., 97, 171–178. CrossRefMedlineWeb of Science 28. ↵ Lampe, A.K., Hampton, P.J., Woodford-Richens, K., Tomlinson, I., Lawrence, C.M. and Douglas, F.S. (2003) Laugier–Hunziker syndrome: an important differential diagnosis for Peutz–Jeghers syndrome. J. Med. Genet., 40, e77. FREE Full Text 29. ↵ Suter, C.M., Martin, D.I. and Ward, R.L. (2004) Germline epimutation of MLH1 in individuals with multiple cancers. Nat. Genet., 36, 497– 501. CrossRefMedlineWeb of Science 30. ↵ Wu, Y., Berends, M.J., Sijmons, R.H., Mensink, R.G., Verlind, E., Kooi, K.A., van der Sluis, T., Kempinga, C., van der Zee, A.G., Hollema, H. et al. (2001) A role for MLH3 in hereditary nonpolyposis colorectal cancer. Nat. Genet., 29, 137–138. CrossRefMedlineWeb of Science 31. Liu, H.X., Zhou, X.L., Liu, T., Werelius, B., Lindmark, G., Dahl, N. and Lindblom, A. (2003) The role of hMLH3 in familial colorectal cancer. Cancer Res., 63, 1894–1899. Abstract/FREE Full Text 32. ↵
  • 15. Hienonen, T., Laiho, P., Salovaara, R., Mecklin, J.P., Jarvinen, H., Sistonen, P., Peltomaki, P., Lehtonen, R., Nupponen, N.N., Launonen, V. et al. (2003) Little evidence for involvement of MLH3 in colorectal cancer predisposition. Int. J. Cancer, 106, 292–296. CrossRefMedlineWeb of Science 33. ↵ Wu, Y., Berends, M.J., Post, J.G., Mensink, R.G., Verlind, E., van der Sluis, T., Kempinga, C., Sijmons, R.H., van der Zee, A.G., Hollema, H. et al. (2001) Germline mutations of EXO1 gene in patients with hereditary nonpolyposis colorectal cancer (HNPCC) and atypical HNPCC forms. Gastroenterology, 120, 1580–1587. CrossRefMedlineWeb of Science 34. Tomlinson, I. and Houlston, R. (2001) Is EXO1 a colon cancer predisposition gene? Gastroenterology, 120, 1860–1862. MedlineWeb of Science 35. Alam, N.A., Gorman, P., Jaeger, E.E., Kelsell, D., Leigh, I.M., Ratnavel, R., Murdoch, M.E., Houlston, R.S., Aaltonen, L.A., Roylance, R.R. et al. (2003) Germline deletions of EXO1 do not cause colorectal tumors and lesions which are null for EXO1 do not have microsatellite instability. Cancer Genet. Cytogenet., 147, 121–127. CrossRefMedlineWeb of Science 36. ↵ Jagmohan-Changur, S., Poikonen, T., Vilkki, S., Launonen, V., Wikman, F., Orntoft, T.F., Moller, P., Vasen, H., Tops, C., Kolodner, R.D. et al. (2003) EXO1 variants occur commonly in normal population: evidence against a role in hereditary nonpolyposis colorectal cancer. Cancer Res., 63, 154–158. Abstract/FREE Full Text 37. ↵ Johnson, V., Volikos, E., Talbot, I., Tomlinson, I. and Silver, A. (2004) Beta-catenin mutations are specific to carcinomas in the HNPCC syndrome. Gut, in press. 38. ↵ Jiricny, J. and Marra, G. (2003) DNA repair defects in colon cancer. Curr. Opin. Genet. Dev., 13, 61–69. CrossRefMedlineWeb of Science 39. ↵ Crabtree, M., Sieber, O.M., Lipton, L., Hodgson, S.V., Lamlum, H., Thomas, H.J., Neale, K., Phillips, R.K., Heinimann, K. and Tomlinson, I.P. (2003) Refining the relation between ‘first hits’ and ‘second hits’ at the APC locus: the ‘loose fit’ model and evidence for differences in somatic mutation spectra among patients. Oncogene, 22, 4257– 4265.
  • 16. CrossRefMedlineWeb of Science 40. ↵ Lamlum, H., Ilyas, M., Rowan, A., Clark, S., Johnson, V., Bell, J., Frayling, I., Efstathiou, J., Pack, K., Payne, S. et al. (1999) The type of somatic mutation at APC in familial adenomatous polyposis is determined by the site of the germline mutation: a new facet to Knudson's ‘two-hit’ hypothesis. Nat. Med., 5, 1071–1075. CrossRefMedlineWeb of Science 41. Rowan, A.J., Lamlum, H., Ilyas, M., Wheeler, J., Straub, J., Papadopoulou, A., Bicknell, D., Bodmer, W.F. and Tomlinson, I.P. (2000) APC mutations in sporadic colorectal tumors: a mutational ‘hotspot’ and interdependence of the ‘two hits’. Proc. Natl Acad. Sci. USA, 97, 3352–3357. CrossRefMedlineWeb of Science 42. ↵ Albuquerque, C., Breukel, C., van der Luijt, R., Fidalgo, P., Lage, P., Slors, F.J., Leitao, C.N., Fodde, R. and Smits, R. (2002) The ‘just- right’ signaling model: APC somatic mutations are selected based on a specific level of activation of the beta-catenin signaling cascade. Hum. Mol. Genet., 11, 1549–1560. Abstract/FREE Full Text 43. ↵ Groves, C., Lamlum, H., Crabtree, M., Williamson, J., Taylor, C., Bass, S., Cuthbert-Heavens, D., Hodgson, S., Phillips, R. and Tomlinson, I. (2002) Mutation cluster region, association between germline and somatic mutations and genotype–phenotype correlation in upper gastrointestinal familial adenomatous polyposis. Am. J. Pathol., 160, 2055–2061. MedlineWeb of Science 44. ↵ Spirio, L.N., Samowitz, W., Robertson, J., Robertson, M., Burt, R.W., Leppert, M. and White, R. (1998) Alleles of APC modulate the frequency and classes of mutations that lead to colon polyps. Nat. Genet., 20, 385–388. CrossRefMedlineWeb of Science 45. ↵ Su, L.K., Barnes, C.J., Yao, W., Qi, Y., Lynch, P.M. and Steinbach, G. (2000) Inactivation of germline mutant APC alleles by attenuated somatic mutations: a molecular genetic mechanism for attenuated familial adenomatous polyposis. Am. J. Hum. Genet., 67, 582–590. CrossRefMedlineWeb of Science 46. ↵
  • 17. Crabtree, M.D., Tomlinson, I.P., Talbot, I.C. and Phillips, R.K. (2001) Variability in the severity of colonic disease in familial adenomatous polyposis results from differences in tumour initiation rather than progression and depends relatively little on patient age. Gut, 49, 540–543. Abstract/FREE Full Text 47. ↵ Bertario, L., Russo, A., Sala, P., Eboli, M., Giarola, M., D'Amico, F., Gismondi, V., Varesco, L., Pierotti, M.A. and Radice, P. (2001) Genotype and phenotype factors as determinants of desmoid tumors in patients with familial adenomatous polyposis. Int. J. Cancer, 95, 102–107. CrossRefMedlineWeb of Science 48. ↵ Dietrich, W.F., Lander, E.S., Smith, J.S., Moser, A.R., Gould, K.A., Luongo, C., Borenstein, N. and Dove, W. (1993) Genetic identification of Mom-1, a major modifier locus affecting Min-induced intestinal neoplasia in the mouse. Cell, 75, 631–639. CrossRefMedlineWeb of Science 49. MacPhee, M., Chepenik, K.P., Liddell, R.A., Nelson, K.K., Siracusa, L.D. and Buchberg, A.M. (1995) The secretory phospholipase A2 gene is a candidate for the Mom1 locus, a major modifier of ApcMin- induced intestinal neoplasia. Cell, 81, 957–966. CrossRefMedlineWeb of Science 50. ↵ Cormier, R.T., Hong, K.H., Halberg, R.B., Hawkins, T.L., Richardson, P., Mulherkar, R., Dove, W.F. and Lander, E.S. (1997) Secretory phospholipase Pla2g2a confers resistance to intestinal tumorigenesis. Nat. Genet., 17, 88–91. CrossRefMedlineWeb of Science 51. ↵ Crabtree, M.D., Fletcher, C., Churchman, M., Hodgson, S.V., Neale, K., Phillips, R.K. and Tomlinson, I.P. (2004) Analysis of candidate modifier loci for the severity of colonic familial adenomatous polyposis, with evidence for the importance of the N-acetyl transferases. Gut, 53, 271–276. Abstract/FREE Full Text 52. ↵ Laken, S.J., Petersen, G.M., Gruber, S.B., Oddoux, C., Ostrer, H., Giardiello, F.M., Hamilton, S.R., Hampel, H., Markowitz, A., Klimstra, D. et al. (1997) Familial colorectal cancer in Ashkenazim due to a hypermutable tract in APC. Nat. Genet., 17, 79–83. CrossRefMedlineWeb of Science
  • 18. 53. ↵ Strul, H., Barenboim, E., Leshno, M., Gartner, M., Kariv, R., Aljadeff, E., Aljadeff, Y., Kazanov, D., Strier, L., Keidar, A. et al. (2003) The I1307K adenomatous polyposis coli gene variant does not contribute in the assessment of the risk for colorectal cancer in Ashkenazi Jews. Cancer Epidemiol. Biomark. Prev., 12, 1012–1015. Abstract/FREE Full Text 54. ↵ Sieber, O., Lipton, L., Heinimann, K. and Tomlinson, I. (2003) Colorectal tumourigenesis in carriers of the APC I1307K variant: lone gunman or conspiracy? J. Pathol., 199, 137–1379. CrossRefMedlineWeb of Science 55. ↵ Whitelaw, S.C., Murday, V.A., Tomlinson, I.P., Thomas, H.J., Cottrell, S., Ginsberg, A., Bukofzer, S., Hodgson, S.V., Skudowitz, R.B., Jass, J.R. et al. (1997) Clinical and molecular features of the hereditary mixed polyposis syndrome. Gastroenterology, 112, 327–334. CrossRefMedlineWeb of Science 56. ↵ Thomas, H.J., Whitelaw, S.C., Cottrell, S.E., Murday, V.A., Tomlinson, I.P., Markie, D., Jones, T., Bishop, D.T., Hodgson, S.V., Sheer, D. et al. (1996) Genetic mapping of hereditary mixed polyposis syndrome to chromosome 6q. Am. J. Hum. Genet., 58, 770–776. MedlineWeb of Science 57. ↵ Tomlinson, I., Rahman, N., Frayling, I., Mangion, J., Barfoot, R., Hamoudi, R., Seal, S., Northover, J., Thomas, H.J., Neale, K. et al. (1999) Inherited susceptibility to colorectal adenomas and carcinomas: evidence for a new predisposition gene on 15q14–q22. Gastroenterology, 116, 789–795. CrossRefMedlineWeb of Science 58. ↵ Jaeger, E.E., Woodford-Richens, K.L., Lockett, M., Rowan, A.J., Sawyer, E.J., Heinimann, K., Rozen, P., Murday, V.A., Whitelaw, S.C., Ginsberg, A. et al. (2003) An ancestral Ashkenazi haplotype at the HMPS/CRAC1 locus on 15q13–q14 is associated with hereditary mixed polyposis syndrome. Am. J. Hum. Genet., 72, 1261–1267. CrossRefMedlineWeb of Science 59. ↵ Wiesner, G.L., Daley, D., Lewis, S., Ticknor, C., Platzer, P., Lutterbaugh, J., MacMillen, M., Baliner, B., Willis, J., Elston, R.C. et al. (2003) A subset of familial colorectal neoplasia kindreds linked to
  • 19. chromosome 9q22.2–31.2. Proc. Natl Acad. Sci. USA, 100, 12961– 12965. Abstract/FREE Full Text 60. ↵ Wacholder, S., Chanock, S., Garcia-Closas, M., El Ghormli, L. and Rothman, N. (2004) Assessing the probability that a positive report is false: an approach for molecular epidemiology studies. J. Natl Cancer Inst., 96, 434–442. Abstract/FREE Full Text 61. ↵ Egger, M., Smith, G.D. and Sterne, J.A. (2001) Uses and abuses of meta-analysis. Clin. Med., 1, 478–484. MedlineWeb of Science 62. ↵ Sterne, J.A. and Egger, M. (2001) Funnel plots for detecting bias in meta-analysis: guidelines on choice of axis. J. Clin. Epidemiol., 54, 1046–1055. CrossRefMedlineWeb of Science 63. ↵ Houlston, R.S. and Tomlinson, I.P. (2001) Polymorphisms and colorectal tumor risk. Gastroenterology, 121, 282–301. CrossRefMedlineWeb of Science 64. ↵ de Jong, M.M., Nolte, I.M., te Meerman, G.J., van der Graaf, W.T., de Vries, E.G., Sijmons, R.H., Hofstra, R.M. and Kleibeuker, J.H. (2002) Low-penetrance genes and their involvement in colorectal cancer susceptibility. Cancer Epidemiol. Biomark. Prev., 11, 1332–1352. Abstract/FREE Full Text 65. ↵ Gruber, S.B., Ellis, N.A., Scott, K.K., Almog, R., Kolachana, P., Bonner, J.D., Kirchhoff, T., Tomsho, L.P., Nafa, K., Pierce, H. et al. (2002) BLM heterozygosity and the risk of colorectal cancer. Science, 297, 2013. FREE Full Text 66. ↵ Cleary, S.P., Zhang, W., Di Nicola, N., Aronson, M., Aube, J., Steinman, A., Haddad, R., Redston, M., Gallinger, S., Narod, S.A. et al. (2003) Heterozygosity for the BLM(Ash) mutation and cancer risk. Cancer Res., 63, 1769–1771. Abstract/FREE Full Text 67. ↵ Sharp, L. and Little, J. (2004) Polymorphisms in genes involved in folate metabolism and colorectal neoplasia: a HuGE review. Am. J. Epidemiol., 159, 423–443.
  • 20. Abstract/FREE Full Text 68. ↵ Grieu, F., Malaney, S., Ward, R., Joseph, D. and Iacopetta, B. (2003) Lack of association between CCND1 G870A polymorphism and the risk of breast and colorectal cancers. Anticancer Res., 23, 4257– 4259. MedlineWeb of Science 69. ↵ Le Marchand, L., Seifried, A., Lum-Jones, A., Donlon, T. and Wilkens, L.R. (2003) Association of the cyclin D1 A870G polymorphism with advanced colorectal cancer. J. Am. Med. Assoc., 290, 2843–2848. Abstract/FREE Full Text 70. ↵ Shin, Y., Kim, I.J., Kang, H.C., Park, J.H., Park, H.W., Jang, S.G., Lee, M.R., Jeong, S.Y., Chang, H.J., Ku, J.L. et al. (2004) A functional polymorphism (−347 G→GA) in the E-cadherin gene is associated with colorectal cancer. Carcinogenesis, in press. 71. ↵ Ulrich, C.M., Bigler, J., Sparks, R., Whitton, J., Sibert, J.G., Goode, E.L., Yasui, Y. and Potter, J.D. (2004) Polymorphisms in PTGS1 (=COX-1) and risk of colorectal polyps. Cancer Epidemiol. Biomark. Prev., 13, 889–893. Abstract/FREE Full Text 72. ↵ Lin, H.J., Lakkides, K.M., Keku, T.O., Reddy, S.T., Louie, A.D., Kau, I.H., Zhou, H., Gim, J.S., Ma, H.L., Matthies, C.F. et al. (2002) Prostaglandin H synthase 2 variant (Val511Ala) in African Americans may reduce the risk for colorectal neoplasia. Cancer Epidemiol. Biomark. Prev., 11, 1305–1315. Abstract/FREE Full Text 73. ↵ Shaheen, N.J., Silverman, L.M., Keku, T., Lawrence, L.B., Rohlfs, E.M., Martin, C.F., Galanko, J. and Sandler, R.S. (2003) Association between hemochromatosis (HFE) gene mutation carrier status and the risk of colon cancer. J. Natl Cancer Inst., 95, 154–159. Abstract/FREE Full Text 74. ↵ Slattery, M.L., Samowitz, W., Curtin, K., Ma, K.N., Hoffman, M., Caan, B. and Neuhausen, S. (2004) Associations among IRS1, IRS2, IGF1, and IGFBP3 genetic polymorphisms and colorectal cancer. Cancer Epidemiol. Biomark. Prev., 13, 1206–1214. Abstract/FREE Full Text 75. ↵
  • 21. Slattery, M.L., Samowitz, W., Hoffman, M., Ma, K.N., Levin, T.R. and Neuhausen, S. (2004) Aspirin, NSAIDs, and colorectal cancer: possible involvement in an insulin-related pathway. Cancer Epidemiol. Biomark. Prev., 13, 538–545. Abstract/FREE Full Text 76. ↵ Landi, S., Moreno, V., Gioia-Patricola, L., Guino, E., Navarro, M., de Oca, J., Capella, G. and Canzian, F. (2003) Association of common polymorphisms in inflammatory genes interleukin (IL)6, IL8, tumor necrosis factor alpha, NFKB1, and peroxisome proliferator-activated receptor gamma with colorectal cancer. Cancer Res., 63, 3560–3566. Abstract/FREE Full Text 77. ↵ Lipkin, S.M., Rozek, L.S., Rennert, G., Yang, W., Chen, P.C., Hacia, J., Hunt, N., Shin, B., Fodor, S., Kokoris, M. et al. (2004) The MLH1 D132H variant is associated with susceptibility to sporadic colorectal cancer. Nat. Genet., 36, 694–699. CrossRefMedlineWeb of Science 78. ↵ de Jong, M.M., Hofstra, R.M., Kooi, K.A., Westra, J.L., Berends, M.J., Wu, Y., Hollema, H., van der Sluis, T., van der Graaf, W.T., de Vries, E.G. et al. (2004) No association between two MLH3 variants (S845G and P844L) and colorectal cancer risk. Cancer Genet. Cytogenet., 152, 70–71. CrossRefMedlineWeb of Science 79. ↵ Hinoda, Y., Okayama, N., Takano, N., Fujimura, K., Suehiro, Y., Hamanaka, Y., Hazama, S., Kitamura, Y., Kamatani, N. and Oka, M. (2002) Association of functional polymorphisms of matrix metalloproteinase (MMP)-1 and MMP-3 genes with colorectal cancer. Int. J. Cancer, 102, 526–529. CrossRefMedlineWeb of Science 80. ↵ Matsuo, K., Hamajima, N., Hirai, T., Kato, T., Inoue, M., Takezaki, T. and Tajima, K. (2002) Methionine synthase reductase gene A66G polymorphism is associated with risk of colorectal cancer. Asian Pac. J. Cancer Prev., 3, 353–359. Medline 81. ↵ Goode, E.L., Potter, J.D., Bigler, J. and Ulrich, C.M. (2004) Methionine synthase D919G polymorphism, folate metabolism, and colorectal adenoma risk. Cancer Epidemiol. Biomark. Prev., 13, 157–162. Abstract/FREE Full Text
  • 22. 82. ↵ Loktionov, A., Watson, M.A., Stebbings, W.S., Speakman, C.T. and Bingham, S.A. (2003) Plasminogen activator inhibitor-1 gene polymorphism and colorectal cancer risk and prognosis. Cancer Lett., 189, 189–196. CrossRefMedlineWeb of Science 83. ↵ Sparks, R., Bigler, J., Sibert, J.G., Potter, J.D., Yasui, Y. and Ulrich, C.M. (2004) TGF{beta}1 polymorphism (L10P) and risk of colorectal adenomatous and hyperplastic polyps. Int. J. Epidemiol., in press. 84. ↵ Pasche, B., Kaklamani, V., Hou, N., Young, T., Rademaker, A., Peterlongo, P., Ellis, N., Offit, K., Caldes, T., Reiss, M. et al. (2004) TGFBR1*6A and cancer: a meta-analysis of 12 case-control studies. J. Clin. Oncol., 22, 756–758. FREE Full Text 85. ↵ Chen, J., Hunter, D.J., Stampfer, M.J., Kyte, C., Chan, W., Wetmur, J.G., Mosig, R., Selhub, J. and Ma, J. (2003) Polymorphism in the thymidylate synthase promoter enhancer region modifies the risk and survival of colorectal cancer. Cancer Epidemiol Biomark. Prev., 12, 958–962. Abstract/FREE Full Text 86. ↵ Ulrich, C.M., Bigler, J., Bostick, R., Fosdick, L. and Potter, J.D. (2002) Thymidylate synthase promoter polymorphism, interaction with folate intake, and risk of colorectal adenomas. Cancer Res., 62, 3361–3364. Abstract/FREE Full Text 87. ↵ Wong, H.L., Seow, A., Arakawa, K., Lee, H.P., Yu, M.C. and Ingles, S.A. (2003) Vitamin D receptor start codon polymorphism and colorectal cancer risk: effect modification by dietary calcium and fat in Singapore Chinese. Carcinogenesis, 24, 1091–1095. Abstract/FREE Full Text 88. ↵ Couzin, J. (2004) Genomics. Consensus emerges on HapMap strategy. Science, 304, 671–673. Abstract/FREE Full Text 89. ↵ Cui, H., Cruz-Correa, M., Giardiello, F.M., Hutcheon, D.F., Kafonek, D.R., Brandenburg, S., Wu, Y., He, X., Powe, N.R. and Feinberg, A.P.
  • 23. (2003) Loss of IGF2 imprinting: a potential marker of colorectal cancer risk. Science, 299, 1753–1755. Abstract/FREE Full Text 90. Cruz-Correa, M., Cui, H., Giardiello, F.M., Powe, N.R., Hylind, L., Robinson, A., Hutcheon, D.F., Kafonek, D.R., Brandenburg, S., Wu, Y. et al. (2004) Loss of imprinting of insulin growth factor II gene: a potential heritable biomarker for colon neoplasia predisposition. Gastroenterology, 126, 964–970. CrossRefMedlineWeb of Science 91. Woodson, K., Flood, A., Green, L., Tangrea, J.A., Hanson, J., Cash, B., Schatzkin, A. and Schoenfeld, P. (2004) Loss of insulin-like growth factor-II imprinting and the presence of screen-detected colorectal adenomas in women. J. Natl Cancer Inst., 96, 407–410. Abstract/FREE Full Text 92. ↵ Jirtle, R.L. (2004) IGF2 loss of imprinting: a potential heritable risk factor for colorectal cancer. Gastroenterology, 126, 1190–1193. CrossRefMedlineWeb of Science 93. ↵ White, S., Bubb, V.J. and Wyllie, A.H. (1996) Germline APC mutation (Gln1317) in a cancer-prone family that does not result in familial adenomatous polyposis. Genes Chromosomes Cancer, 15, 122–128. CrossRefMedlineWeb of Science 94. Frayling, I.M., Beck, N.E., Ilyas, M., Dove-Edwin, I., Goodman, P., Pack, K., Bell, J.A., Williams, C.B., Hodgson, S.V., Thomas, H.J. et al. (1998) The APC variants I1307K and E1317Q are associated with colorectal tumors, but not always with a family history. Proc. Natl Acad. Sci. USA, 95, 10722–10727. Abstract/FREE Full Text 95. Popat, S., Stone, J., Coleman, G., Marshall, G., Peto, J., Frayling, I. and Houlston, R. (2000) Prevalence of the APC E1317Q variant in colorectal cancer patients. Cancer Lett., 149, 203–206. CrossRefMedlineWeb of Science 96. Gismondi, V., Bonelli, L., Sciallero, S., Margiocco, P., Viel, A., Radice, P., Mondini, P., Sala, P., Montera, M.P., Mareni, C. et al. (2002) Prevalence of the E1317Q variant of the APC gene in Italian patients with colorectal adenomas. Genet. Test., 6, 313–317. CrossRefMedlineWeb of Science 97. ↵ Hahnloser, D., Petersen, G.M., Rabe, K., Snow, K., Lindor, N.M., Boardman, L., Koch, B., Doescher, D., Wang, L., Steenblock, K. et al. (2003) The APC E1317Q variant in adenomatous polyps and colorectal cancers. Cancer Epidemiol. Biomark. Prev., 12, 1023–1028.
  • 24. Abstract/FREE Full Text 98. ↵ Meijers-Heijboer, H., van den Ouweland, A., Klijn, J., Wasielewski, M., de Snoo, A., Oldenburg, R., Hollestelle, A., Houben, M., Crepin, E., van Veghel-Plandsoen, M. et al. (2002) Low-penetrance susceptibility to breast cancer due to CHEK2(*)1100delC in noncarriers of BRCA1 or BRCA2 mutations. Nat. Genet., 31, 55–59. CrossRefMedlineWeb of Science 99. ↵ Houlston, R.S. and Tomlinson, I.P. (2000) Detecting low penetrance genes in cancer: the way ahead. J. Med. Genet., 37, 161–167. Abstract/FREE Full Text 100. ↵ Risch, N. (2001) Implications of multilocus inheritance for gene– disease association studies. Theor. Popul. Biol., 60, 215–220. CrossRefMedlineWeb of Science 101. ↵ Houlston, R.S. and Peto, J. (2003) The future of association studies of common cancers. Hum. Genet., 112, 434–435. MedlineWeb of Science 102. ↵ Botstein, D. and Risch, N. (2003) Discovering genotypes underlying human phenotypes: past successes for mendelian disease, future approaches for complex disease. Nat. Genet., 33 (suppl.), 228–237. 103. ↵ Gabriel, S.B., Schaffner, S.F., Nguyen, H., Moore, J.M., Roy, J., Blumenstiel, B., Higgins, J., DeFelice, M., Lochner, A., Faggart, M. et al. (2002) The structure of haplotype blocks in the human genome. Science, 296, 2225–2229. Abstract/FREE Full Text 104. ↵ Satsangi, J., Parkes, M., Jewell, D.P. and Bell, J.I. (1998) Genetics of inflammatory bowel disease (Review). Clin. Sci. (London), 94, 473– 478. Articles citing this article Characterization of 9p24 Risk Locus and Colorectal Adenoma and Cancer: Gene-Environment Interaction and Meta-Analysis Cancer Epidemiol. Biomarkers Prev. (2010) 19(12): 3131-3139 o Abstract o Full Text (HTML) o Full Text (PDF) Allele-specific expression of TGFBR1 in colon cancer patients Carcinogenesis (2010) 31(10): 1800-1804 o Abstract
  • 25. o Full Text (HTML) o Full Text (PDF) Association of the ARLTS1 Cys148Arg variant with sporadic and familial colorectal cancer Carcinogenesis (2007) 28(8): 1687-1691 o Abstract o Full Text (HTML) o Full Text (PDF) TGF-{beta} signaling alterations and susceptibility to colorectal cancer Hum Mol Genet (2007) 16(R1): R14-R20 o Abstract o Full Text (HTML) o Full Text (PDF) « Previous | Next Article »Table of Contents This Article 1. Hum. Mol. Genet. (2004) 13 (suppl 2): R177-R185. doi: 10.1093/hmg/ddh247 1. Abstract 2. » Full Text (HTML) 3. Full Text (PDF) - Classifications 1. o Review o Review - Services 1. Alert me when cited 2. Alert me if corrected 3. Find similar articles 5. Similar articles in PubMed 6. Add to my archive 7. Download citation 8. Request Permissions + Citing Articles + Google Scholar + PubMed + Related Content + Share o o o o o o o Navigate This Article 1. Top 2. Abstract 3. PROGRESS IN UNDERSTANDING THE HEREDITARY COLORECTAL TUMOUR SYNDROMES 4. RECENT DEVELOPMENTS THROUGH LINKAGE ANALYSIS 5. EVIDENCE FOR LOW-PENETRANCE ALLELES ASSOCIATED WITH AN INCREASED RISK OF CRC 6. FUTURE STRATEGIES FOR IDENTIFYING CRC PREDISPOSITION GENES 7. ACKNOWLEDGEMENTS 8. References - People also read [Beta]
  • 26. 1. Specific codon 13 K-ras mutations are predictive of clinical outcome in colorectal cancer patients, whereas codon 12 K-ras mutations are associated with mucinous histotype What's this? Search this journal: Advanced » Current Issue 1. January 15, 2012 21 (2) 1. 1. Alert me to new issues The Journal About this journal Rights & Permissions Dispatch date of the next issue This journal is a member of the Committee on Publication Ethics (COPE) We are mobile – find out more Impact factor: 8.058 5-Yr impact factor: 8.145 Executive Editors Professor Kay Davies Professor Anthony Wynshaw-Boris Professor Joel Hirschhorn View full editorial board For Authors Instructions to authors Online submission Submit Now! Self-archiving policy
  • 27. Open access options for authors - visit Oxford Open This journal enables compliance with the NIH Public Access Policy Alerting Services Email table of contents Email Advance Access CiteTrack XML RSS feed Corporate Services Advertising sales Reprints Supplements Most Most Read Most Cited Most Read 1. Epistasis: what it means, what it doesn't mean, and statistical methods to detect it in humans 2. Genetics of cleft lip and palate: syndromic genes contribute to the incidence of non- syndromic clefts 3. The genetics of psoriasis, psoriatic arthritis and atopic dermatitis 4. Non-coding RNA 5. Synergy Between the Genes for Butyrylcholinestera se K Variant and Apolipoprotein E4 in Late-Onset
  • 28. Confirmed Alzheimer's Disease » View all Most Read articles Most Cited 1. The DNA methyltransferases of mammals 2. Nonsense-Mediated mRNA Decay in Health and Disease 3. Isolation of a Candidate Human Telomerase Catalytic Subunit Gene, Which Reveals Complex Splicing Patterns in Different Cell Types 4. Human Y Chromosome Azoospermia Factors (AZF) Mapped to Different Subregions in Yq11 5. Mutation of human short tandem repeats » View all Most Cited articles Online ISSN 1460-2083 - Print ISSN 0964-6906 Copyright © 2011 Oxford University Press Oxford Journals Oxford University Press Site Map Privacy Policy Frequently Asked Questions Other Oxford University Press sites: Oxford University Press