Download Hereditary Colorectal Cancer

Document related concepts

Epigenetics of neurodegenerative diseases wikipedia , lookup

Nutriepigenomics wikipedia , lookup

Epistasis wikipedia , lookup

Designer baby wikipedia , lookup

Neuronal ceroid lipofuscinosis wikipedia , lookup

Microevolution wikipedia , lookup

Cancer epigenetics wikipedia , lookup

RNA-Seq wikipedia , lookup

Mutagen wikipedia , lookup

BRCA mutation wikipedia , lookup

NEDD9 wikipedia , lookup

Genome (book) wikipedia , lookup

Mutation wikipedia , lookup

Frameshift mutation wikipedia , lookup

Point mutation wikipedia , lookup

Oncogenomics wikipedia , lookup

Transcript
Hereditary Colorectal Cancer-Part II
Lynch Syndrome
There are 2 broad classes of hereditary forms of colorectal cancer
(CRC), based on the predominant anatomic location of the CRC: distal
versus proximal. CRCs involving the distal colon are more likely to show
aneuploid DNA content, harbor mutations in APC, p53, and K-ras genes,
and behave more aggressively303,304; proximal CRCs are more likely to
show diploid DNA, possess microsatellite instability, harbor mutations in
the mismatch repair (MMR) genes, and behave less aggressively, as in
hereditary nonpolyposis colorectal cancer (HNPCC) or Lynch syndrome.303,304 Familial adenomatous polyposis (FAP) and most sporadic
cases may be considered a paradigm for the first, or distal, CRC class,
whereas Lynch syndrome more clearly represents the second, or proximal, CRC class.303,304
The term Lynch syndrome is used in preference to HNPCC, given the
fact that the syndrome’s phenotype is not completely without polyps, and
in addition to CRC it involves a litany of extracolonic cancer types that
are integral to the syndrome. It is the most commonly occurring
hereditary syndrome that predisposes to CRC304 (Fig 2; Part I). Its
diagnosis requires a sufficiently detailed family history of colorectal
carcinoma as well as cancer of all anatomic sites, particularly those that
are integral to this disorder (e.g., carcinoma of the endometrium [the
second most common cancer], ovary, stomach [particularly in families
from Japan and Korea],305 hepatobiliary tract, small bowel, pancreas,
upper uroepithelial tract, and brain).303,304,306,307
History of Lynch Syndrome
The history of what is known as Lynch syndrome dates to an
observation of Aldred Warthin, pathologist at the University of Michigan
School of Medicine.308 He became deeply moved when his seamstress, in
1895, told him that she would likely die of cancer of the colon, stomach,
or her female organs, because of the enormous proclivity to these cancers
in her family (unfortunately, just as she had told Warthin, she died at a
Curr Probl Surg 2005;42:267-334.
0011-3840/2005/$30.00 ⫹ 0
doi:10.1067/j.cpsurg.2005.02.003
Curr Probl Surg, May 2005
267
young age of metastatic endometrial carcinoma). Warthin listened intently, developed her pedigree, and along with other similar cancer prone
families published this work in 1913.309 Warthin updated the family’s
history in 1925.310 The seamstress’s family has since been known as
Family G.
Lynch and colleagues311 described the natural history and genetics of 2
large Midwestern kindreds (Families N and M) in 1966. Dr. A. James
French, Warthin’s successor as chairman of pathology at the University of
Michigan, heard about Lynch’s research on Families N and M, and
recalled that Warthin, his predecessor, had discovered a similar family
(Family G) in 1895. Lynch was then invited by French to take custody of
all the detailed documents and pathology specimens that the meticulous
Warthin had investigated, catalogued, and published over a span of more
than 30 years.309,310 The history of Family G was then updated and
published in 1971.312 This material is discussed in a more detailed review
of the history of HNPCC.313 Through the use of conversion technology,
an MSH2 mutation was identified in Family G in the year 2000.314 Table
7 summarizes the important points in the history of HNPCC.
Clinical Features and Diagnostic Criteria
The cardinal clinical features of the Lynch syndrome are listed in Table
8 and are discussed in more detail in other review articles.303,304,315
Several international diagnostic criteria for the Lynch syndrome have
been developed, the foremost being the Amsterdam I,316 Amsterdam
II,317 and the Bethesda Guidelines.318 The variety of criteria that have
been developed for the syndrome testifies to its complexity. The original
Amsterdam I criteria were developed for the purpose of consistent
classification of subjects in research programs and focused on CRC
(Table 9). The subsequent Amsterdam II criteria added the syndrome’s
integral extracolonic cancers to the focus to be more useful for clinical
diagnosis purposes. The Bethesda Guidelines introduced molecular genetic considerations, linking the diagnostic criteria of the Lynch syndrome to the high percentage of Lynch syndrome tumors that are positive
for microsatellite instability (MSI), to provide guidelines for MSI testing.
Other criteria, such as those developed in Korea and Japan, make use of
emerging knowledge regarding the syndrome’s phenotypic variability in
different geographic regions. These criteria are based heavily on the
family history, which we consider to be potentially the most costbeneficial component of a patient’s medical evaluation; however, we are
also cognizant of the fact that it remains 1 of the most neglected portions
268
Curr Probl Surg, May 2005
TABLE 7. Landmarks of Lynch syndrome history*
Feature
First
report
References
Family G of Warthin (study began 1895)
Genetic counseling
First report of Lynch et al. on Families N and M
Early age of cancer onset
Autosomal dominant inheritance pattern
Family information session (FIS)
Screening recommendations
Update of Family G
Proximal colon involvement
Beginning of study of Lynch syndrome in Uruguay
Recommendation of prophylactic TAH-BSO
Muir-Torre syndrome (as variant of Lynch syndrome)
Increased incidence of synchronous and metachronous CRC
Lynch syndrome studies begin with the Navajo
Tritiated thymidine distribution studies of rectal mucosa
HNPCC named “Lynch syndrome”
Selenium levels in Lynch syndrome studied
Formation of ICG-HNPCC
Lectin binding studies in FAP and HNPCC
Amsterdam I criteria
Accelerated carcinogenesis and interval CRC
1913
1965
1966
1966
1966
1966
1967
1971
1977
1977
1978
1980
1982
1983
1983
1984
1984
1989
1990
1991
1992
First cancer susceptibility locus found on 2p through linkage
analysis
Second cancer susceptibility locus found on 3p through linkage
analysis
DNA mismatch repair genes reported
1993
309
582, 583
311
311
311
311, 584, 585
586
312
376
587
588
382, 589
377, 378
590–592
593
594
595
317, 596
597–599
316
365, 395, 396,
600, 601
326
1993
327
1993
RER⫹ (MSI) phenotype described
Germline mutations in the syndrome
MSH2 mutation identified
Extracolonic adenocarcinomas
Distinctive pathology features
MSH2; MLH1 mutations
Creighton group’s involvement in Uruguayan study
Historical perspective through 1995
Role of DNA MMR genes in CRC tumorigenesis
Recommendations of prophylactic subtotal colectomy
Survival advantage
NIH NCI workshop on HNPCC (Bethesda Guidelines)
MSH6 mutation
NIH NCI update on MSI
Small bowel involvement
Founder mutation in Finland
Amsterdam II criteria
Tumor infiltrating lymphocytes and their association with MSI
Conversion technology
1993
1993
1993
1994
1994
1994
1995
1995
1995
1996
1996
1996
1997
1997
1998
1998
1999
1999
2000
328, 329, 334,
335
322
328
329
307
365
329, 335
602
603
604, 605
391, 392
374, 379
318
347, 606
607
608
356
317
380
314
Curr Probl Surg, May 2005
269
TABLE 7. Continued
Feature
A complex mutation of MLH1 at codon 222 is associated with
adolescent onset of CRC (more early-onset CRC families
needed for study)
Fluorouracil-based adjuvant chemotherapy benefits patients
with stage II or stage III CRC with MSS or MSI-L tumors but
not those with MSI-H tumors
H2O2 effect improves survival in DNA MMR-deficient cell line
MSH2 del1-6 founder mutation in the United States
First
report
References
2001
609
2003
610
2003
2004
611
315
*Reproduced from Lynch HT, Shaw TG, Lynch JF. Inherited predisposition to cancer a historical
overview. Am J Med Genet C (Semin Med Genet) 2004;129C:5–22.
Abbreviations: CRC, colorectal cancer; HNPCC, hereditary nonpolyposis colorectal cancer;
TAH-BSO, total abdominal hysterectomy and bilateral salpingo-oophorectomy; ICGHNPCC, International Collaborative Group on HNPCC; FAP, familial adenomatous polyposis; MMR, mismatch repair; NIH, National Institutes of Health; NCI, National Cancer
Institute; MSI, Microsatellite instability; MSS, Microsatellite stable.
TABLE 8. Cardinal features of Lynch syndrome
● Earlier average age of CRC onset than in the general population; the average age of CRC
onset in HNPCC is ⬃45 years, whereas the average age of onset in sporadic CRC is
⬃63 years
● Proximal colon involvement (70% of CRCs arise proximal to the splenic flexure)
● A significant excess of synchronous and metachronous CRCs (⬃25% to 30% among
patients having a second primary CRC within 10 years of surgical resection for initial
CRC, if the operation was anything less than a subtotal colectomy)
● Autosomal dominant inheritance pattern
● Increased risk for malignancy at certain extracolonic sites, foremost of which is
endometrial carcinoma, followed by carcinoma of the ovary, stomach, small bowel,
hepatobiliary tract, pancreas, upper uroepithelial tract, and brain
● CRC tumors in HNPCC are more often poorly differentiated, with an excess of mucoid and
signet-cell features, show a Crohn’s-like reaction, and contain a significant excess of
infiltrating lymphocytes within the tumor. Microsatellite instability (MSI) is found in most
CRC tumors in the Lynch syndrome
● Increased survival from CRC
● Accelerated carcinogenesis and interval CRC; a tiny adenoma may emerge into a
carcinoma within 2–3 years, as opposed to 8–10 years in the general population
● Sebaceous adenomas, sebaceous carcinomas, and multiple keratoacanthomas in the
Muir-Torre syndrome (MTS) variant of Lynch syndrome
● The sine qua non, the identification of a germline MMR mutation segregating with
syndrome-affected individuals in the family
Abbreviations: HNPCC, hereditary nonpolyposis colorectal cancer; CRC, colorectal cancer;
MMR, mismatch repair.
of a cancer patient’s medical evaluation in the average clinical practice
setting.319,320
Differing stages in the development of a Lynch syndrome pedigree
270
Curr Probl Surg, May 2005
TABLE 9. Amsterdam I and Amsterdam II criteria and Bethesda Guidelines
Amsterdam I criteria316
At least 3 relatives with histologically verified colorectal cancer:
1. One is a first degree relative of the other 2
2. At least 2 successive generations affected
3. At least 1 of the relatives with colorectal cancer diagnosed at less than 50 years of
age
4. Familial adenomatous polyposis has been excluded
Amsterdam II criteria317
At least 3 relatives with an hereditary nonpolyposis colorectal cancer-associated cancer
(colorectal cancer, endometrial, stomach, ovary, ureter/renal pelvis, brain, small
bowel, hepatobiliary tract, and skin [sebaceous tumors]):
1. One is a first degree relative of the other 2
2. At least 2 successive generations affected
3. At least 1 of the hereditary nonpolyposis colorectal cancer-associated cancers
should be diagnosed at less than 50 years of age
4. Familial adenomatous polyposis should be excluded in any colorectal cancer cases
Tumors should be verified whenever possible.
Bethesda Guidelines for testing of colorectal tumors for microsatellite instability318
1. Individuals with cancer in families that meet the Amsterdam criteria
2. Individuals with 2 HNPCC-related cancers, including synchronous and metachronous
colorectal cancers or associated extracolonic cancers*
3. Individuals with colorectal cancer and a first degree relative with colorectal cancer
and/or HNPCC-related extracolonic cancer and/or a colorectal adenoma; 1 of the
cancers diagnosed at age less than 45 years and the adenoma diagnosed at age
less than 40 years
4. Individuals with colorectal cancer or endometrial cancer diagnosed at age less than
45 years
5. Individuals with right-sided colorectal cancer with an undifferentiated pattern (solid/
cribiform) on histopathology diagnosed at age less than 45 years†
6. Individuals with signet-ring-cell-type colorectal cancer diagnosed at age less than 45
years‡
7. Individuals with adenomas diagnosed at age less than 40 years
*Endometrial, ovarian, gastric, hepatobiliary, or small-bowel cancer or transitional cell
carcinoma of the renal pelvis or ureter.
†Solid/cribiform defined as poorly differentiated or undifferentiated carcinoma composed of
irregular, solid sheets of large eosinophilic cells and containing small gland-like spaces.
‡Composed of more than 50% signet ring cells.
wherein each component provides a pictorial review of its genesis are
shown in Figure 16. For example, note that the proband had multiple
primary cancers at an early age (Panel A), which should provide the
clinician with vital clues of the possibility of the Lynch syndrome. This
much information does not fulfill any of the criteria in Table 9, yet we
believe the information is sufficient to test for a mismatch repair gene
(MMR) germline mutation. However, when the pedigree is further
extended, it essentially epitomizes an HNPCC diagnosis (Panels B and
C), and when extended even further, as shown in Panel D, it essentially
Curr Probl Surg, May 2005
271
FIG 16. Four stages of a pedigree. (Reproduced with permission from Lynch HT, de la Chapelle A. Hereditary
colorectal cancer. N Engl J Med 2003;348:919-32.) A) The initial ascertainment of what turned out to be a
classical hereditary nonpolyposis colorectal cancer pedigree. The proband (IV-1) showed early-onset colorectal
carcinoma and carcinoma of the ureter. These findings by themselves are highly significant. However, his
mother (III-1) had uterine cervical carcinoma at age 37, a tumor not associated with the syndrome, but at age
55 had colorectal carcinoma. B) Further inquiry indicated that the proband’s mother (III-1) had 2 sisters with
endometrial carcinoma at ages 57 (III-2) and 61 (III-3). This pattern, notwithstanding Amsterdam criteria, would
be sufficient for a hereditary nonpolyposis colorectal cancer diagnosis. C) Extending the pedigree further, 1 of
these maternal aunts (III-2) had 3 cancer-affected sons, 1 with early-onset metachronous colon cancer (IV-3), a
second with colon cancer and carcinoma of the ureter (IV-4), and the other with colon cancer only (IV-5). The
other aunt (III-3) had a daughter (IV-6) with cancer of the bile duct. These findings provide strong evidence for
the diagnosis of hereditary nonpolyposis colorectal cancer. D) The fully extended pedigree, with findings
continuing to support a diagnosis of hereditary nonpolyposis colorectal cancer.
evolves into highly detailed information that is used for research
purposes. The extended pedigree is also valuable for identifying other
family members at risk for developing HNPCC and its related extracolonic cancers.
272
Curr Probl Surg, May 2005
FIG 17. The necessary information on first and second degree relatives, which will aid significantly
in hereditary cancer syndrome diagnosis.
From a practical standpoint, the clinician does not need to go into this
much depth to diagnose the Lynch syndrome. We believe that the
modified nuclear pedigree (Fig 17), wherein attention is given to a
detailed description of the proband, with pathologic documentation of
cancer, and is extended to that individual’s primary relatives (parents,
siblings, and progeny), and then to secondary relatives (maternal and
paternal aunts and uncles, and both sets of grandparents), who will be
highly genetically informative because they have passed through the
cancer risk age, will, in most cases, provide enough information to
diagnose the Lynch syndrome, should it be present in the family. The sine
qua non for this diagnosis will be the presence of a germline mutation in
1 of the mismatch repair genes.
Genetics of Lynch Syndrome
Microsatellite Instability. Microsatellite instability (MSI) is a phenomenon observed in some colorectal tumors, especially in patients with
HNPCC. It is present in approximately 90% to 95% of cancers in Lynch
syndrome, and in approximately 15% of sporadic CRCs. When DNA is
amplified using primers derived from highly repetitive DNA sequences,
new alleles frequently appear in tumor DNA relative to normal DNA (Fig
18). These new alleles represent small insertions or deletions as a
consequence of uncorrected errors in DNA replication. In 1 of the first
studies, Thibodeau and colleagues321 found MSI in 25 of 90 (28%)
colorectal cancers, which was more common in proximal versus distal
cancers (P ⫽ 0.003), and these tumors were associated with a better
Curr Probl Surg, May 2005
273
FIG 18. Microsatellite instability, as demonstrated in 3 normal (N) and colon tumor (T) tissue pairs
using the dinucleotide repeat marker D3S1029. Note an extra group of bands in each tumor,
representing a new allele (reproduced with permission from Lindblom A, Tannergard P, Werelius B,
Nordenskjold M. Genetic mapping of a second locus predisposing to hereditary non-polyposis colon
cancer. Nat Genet 1993;5:279-82).
prognosis than those without microsatellite instability (P ⫽ 0.02).
Aaltonen and colleagues322,323 found 86% (25 of 29) of HNPCC
colorectal tumors to be replication error positive (RER⫹, synonymous
with MSI) but only 16% (8 of 49) of sporadic colorectal cancers were
RER⫹. This group found that 3% (1 of 33) of sporadic adenomas and
16% (8 of 49) of sporadic carcinomas were RER⫹, whereas 57% (8 of
14) of adenomas from HNPCC patients were RER⫹. This suggested that
adenomatous polyps were precursor lesions to colorectal cancer in
HNPCC.323
MSI in colorectal cancers reminded investigators of abnormalities seen
in DNA mismatch repair genes from yeast cells, where increases or
decreases in the number of repeat units within repetitive DNA sequences
274
Curr Probl Surg, May 2005
FIG 19. An overview of DNA mismatch repair enzymes (reproduced with permission from Chung DC,
Rustgi AK. The hereditary nonpolyposis colorectal cancer syndrome: genetics and clinical implications. Ann Intern Med 2003;138:560-70)576.
result.324 DNA mismatch repair genes in bacteria encode for the enzymes
mutS (which binds to base mismatches), mutH (which binds to methylated CpG nucleotides and cuts the opposite strand), mutL (a facilitator of
mutS and mutH), UvrD (a helicase that excises mismatched strands),
DNA polymerase III holoenzyme (which adds new bases), and DNA
ligase.325 Since yeast lack methylated DNA, they do not have mutH. The
homologous 2 mutL proteins in yeast are PMS1 and MLH1, and the
homologous protein to mutS is MSH2 (Fig 19). When these yeast genes
are mutated, there is a corresponding 100- to 700-fold increase in MSI. It
is believed that these genetic changes lead to errors in the number of
repetitive sequences replicated, due to lack of repair and DNA polymerCurr Probl Surg, May 2005
275
ase slippage.324 The observation that tumors in patients with Lynch
syndrome had similar changes to those seen in bacterial and yeast cells
with DNA mismatch repair gene mutations led investigators to examine
the human homologs of these genes in HNPCC.
Identification of Lynch Syndrome Genes. In 1993, Peltomäki and
colleagues326 from Finland and Lindblom and colleagues327 from Sweden discovered sites on chromosomes 2p and 3p by linkage analysis,
respectively, as being etiologic for cancer susceptibility in the Lynch
syndrome. Aaltonen and colleagues322 also demonstrated that tumors in
Lynch syndrome were RER⫹.
hMSH2 Gene Mutations. Fischel and colleagues328 identified the mutS
homolog MSH2 in humans (hMSH2) and mapped it to chromosome 2.
They found that 2 of 7 MSI⫹ tumors had point mutations affecting a
splice acceptor site in hMSH2 and that this same mutation was present in
affected members of 2 Lynch syndrome families. The same gene was
identified by Leach and colleagues329 by using a positional cloning
strategy, and a gene homologous to mutS was found to map within an
interval defined by critical recombinants in Lynch syndrome families
linked to a 2p locus. The gene was 2802 bp in length, and a missense
mutation was found in 11 affected members of 1 kindred, a splice site
mutation in another, and a nonsense mutation in another. Both germline
and somatic mutations were found in 1 of 4 RER⫹ sporadic CRC,
suggesting that this gene was a tumor suppressor gene. The important role
of hMSH2 in Lynch syndrome was subsequently confirmed by several
investigators. Nystrom-Lahti and colleagues330 found that 4 of 6 Lynch
syndrome families linked to chromosome 2p had hMSH2 mutations. Liu
and colleagues331 reported that 10 of 25 Lynch syndrome kindreds had
germline hMSH2 mutations, with 9 mutations leading to a truncated
protein. Wijnen and colleagues332 described 7 hMSH2 mutations in 34
Lynch syndrome kindreds leading to protein truncation, and found no
evidence of correlation of genotype with phenotype in Lynch syndrome.
Maliaka and colleagues333 found that 4 of 11 Russian and Moldavian
Lynch syndrome kindreds had hMSH2 mutations.
hMLH1 Gene Mutations. Papadopoulos and colleagues334 screened
human gene databases for sequences with similarities to the yeast mutL
genes and found 3 genes, called hMLH1, hPMS1, and hPMS2. These
genes were localized to chromosomes 3, 2, and 7, respectively, by
somatic cell hybrids. hMLH1 held immediate interest due to the previous
finding of genetic linkage in some Lynch syndrome families to chromosome 3p21.327 This gene was 2268 bp in length and expressed in a wide
variety of tissues. They found that 9 of the 10 Lynch syndrome families
276
Curr Probl Surg, May 2005
demonstrating linkage to markers on 3p had germline hMLH1 mutations
whereas only 1 of 18 Lynch syndrome kindreds that were too small to be
studied by linkage had mutations, thereby establishing hMLH1 as another
important predisposition gene for Lynch syndrome.334
Bronner and colleagues335 independently identified hMLH1 by searching a human cDNA library for genes homologous to mutL. They found a
gene encoding for a 756-amino acid protein that localized to chromosome
3p21.3-23. They also found that 4 affected members of a Lynch syndrome
family had a germline substitution in a highly conserved exon of
hMLH1.335 Han and colleagues336 reported that 8 of 34 (24%) unrelated
Lynch syndrome patients had hMLH1 mutations, and Nystrom-Lahti and
colleagues330 demonstrated that 3 of 4 Lynch syndrome kindreds linked
to 3p had hMLH1 mutations. Kolodner and colleagues337 defined the
genomic structure of hMLH1 and found a frameshift mutation in 9 of 19
members of a Lynch syndrome kindred linked to markers on 3p. Wijnen
and colleagues332 found 12 of 34 (35%) kindreds with hMLH1 mutations,
and exons 15 and 16 were hot spots for mutation, accounting for 50% of
reported mutations. No genotype-phenotype correlations were observed
for hMLH1 either, as suggested by 3 Lynch syndrome families with a
common exon 16 deletion, where 1 had Turcot’s syndrome and the other
2 did not have family members with CNS tumors.338
In cell lines, Parsons and colleagues339 demonstrated that mutation of 1
allele in a DNA mismatch repair gene alone did not result in the RER⫹
phenotype. Hemminki and colleagues340 found that one third of patients
with germline mutation in hMLH1 also had somatic loss of wild-type
hMLH1 in RER⫹ tumors, consistent with a tumor-suppressor gene
function.
Herman and colleagues341 described methylation of the hMLH1 promoter in sporadic CRC demonstrating MSI, with subsequent loss of
hMLH1 expression. No methylation of hMSH2 was found. Methylation
of the hMLH1 promoter is also common in MSI-positive sporadic
endometrial cancers, and Simpkins and colleagues342 reported this in 41
of 53 such cases.
hPMS1 and hPMS2 Mutations. Papadopoulos and colleagues334 discovered the human mutL homologs hPMS1 and hPMS2 in 1994 and
mapped them to chromosomes 2 and 7. Nicolaides and colleagues343
mapped hPMS1 to chromosome to 2q31-33 by FISH and demonstrated
the gene encoded for a 932-amino acid protein; hPMS2 mapped to 7p22
and encoded for a 862-amino acid protein. They examined 22 hMSH2 and
hMLH1 mutation-negative Lynch syndrome patients and found 1 with a
hPMS1 missense mutation and another with a hPMS2 deletion. A tumor
Curr Probl Surg, May 2005
277
from the latter patient demonstrated deletion of the other hPMS2 allele,
supporting a tumor suppressor role.343 A family with Turcot’s syndrome
has also been reported with a missense mutation of hPMS2.29
hMSH6 Gene Mutations. Another mutS gene homolog was identified
in 1995, by virtue of it forming heterodimers with hMSH2, called GTBP
(G/T binding protein) or hMSH6.344,345 It was found to map in close
proximity to hMSH2 on chromosome 2p16, and mutations were rare in
Lynch syndrome patients. Cells lacking hMSH6 generally have changes
in mononucleotide tracts rather than other simple tandem repeat sequences more commonly present with other MMR genes.346 Miyaki and
colleagues347 found that 1 of 5 HNPCC families lacking hMSH2 or
hMLH1 mutations had an hMSH6 mutation. Wijnen and colleagues348
reported 10 hMSH6 mutations in 214 hMLH1 and hMSH2 mutation
negative families, 71 meeting the Amsterdam criteria (3 mutations) and
143 suspected of having Lynch syndrome but not meeting the Amsterdam
criteria (7 mutations). The age at which colon cancer developed was later
in these patients, and there was a higher incidence of atypical endometrial
hyperplasia and cancer (73%, versus 29% in hMSH2 and 31% in hMLH1
mutation carriers).348 Wu and colleagues349 demonstrated that 4 of 18
individuals with Lynch syndrome-associated tumors that were MSI-low
had hMSH6 mutations. Berends and colleagues350 found hMSH6 mutations in 25 of 316 suspected Lynch syndrome cases, and Huang and
colleagues351 detected only 1 of 90 cases with hMSH6 mutations in which
hMSH2 or hMLH1 mutations were not found.
Endometrial Cancer and MMR Germline Mutations. Berends and
colleagues352 examined the frequency of MMR germline mutations in 58
patients with endometrial carcinoma who were diagnosed at less than 50
years of age. The objective was to relate the occurrence of MMR
mutations to family history, as well as to histopathologic data in the
presence of MSI positivity and immunostaining to develop criteria for
genetic testing. The results showed that 5 of 22 patients who had a
positive first degree family history for Lynch syndrome-related cancers
had pathogenic germline mutations (1 MLH1, 3 MSH2, and 1 MSH6).
Four of the mutation carriers were members of families that fulfilled the
revised Amsterdam criteria, whereas no mutations were identified in the
35 patients who lacked such family history (P ⫽ 0.006). In turn, MSI was
identified in 20 of 57 cancers in which 1 or more MMR proteins were
absent. Among all 5 MMR mutation carriers, immunostaining identified
the involved MMR gene.352 These findings support the conclusion that an
HNPCC-related cancer in a first degree relative is an important selection
278
Curr Probl Surg, May 2005
criterion for mutation analysis. Immunostaining of MMR proteins will
then aid in targeting the gene(s) that should be analyzed.
Overview of Genetic Changes. Lynch syndrome is an autosomal
dominant condition, caused by germline changes in DNA mismatch repair
genes, which behave as tumor suppressor genes. Individuals born with
these germline alterations have 1 normal copy (wild-type) of the gene in
each cell and 1 mutant copy. When the wild-type (functional) copy is lost
as a somatic event (in a nongerm cell), that cell develops the characteristic
of microsatellite instability. The inevitable mistakes that occur during
DNA replication are not efficiently repaired, and these changes may lead
to unrestrained growth that leads to adenoma, then carcinoma. The
numbers of the mutations of various MMR genes in 49 Lynch syndrome
families fulfilling the Amsterdam criteria were 21 hMLH1 (43%; 19 point
mutations, 2 deletions), 22 hMSH2 (45%; 10 point mutations, 12
deletions), 2 hMSH6 (4%), and 4 (8%) were negative for point mutations
or genomic rearrangements.353 The frequency of hMSH2 mutations may
have been overestimated in this study, however, because 7 families had
the same 20 kb deletion encompassing exons 1-6 and appeared potentially
to share a common ancestor. A compilation of published and unpublished
mutations have been established by the International Collaborative Group
on HNPCC (http://www.nfdht.nl), in which the numbers of hMLH1
mutations reported were 323 (63% of total), hMSH2 156 (25%), hMSH6
30 (6%), hPMS2 2 (0.4%), and hPMS1 1 (0.2%). The mutations found in
the hMLH1 and hMSH2 genes are distributed throughout the exons, and
no clear correlations between genotype and phenotype are known, with
the exception of the higher incidence of MSI-low and endometrial cancers
in those patients with hMSH6 mutations.
One might appropriately probe more deeply into the question about why
only approximately one half of Lynch syndrome families will have an
MMR mutation identified, or why some classical FAP families may not
have an APC germline mutation identified. This vexing problem has been
addressed by Renkonen and colleagues354 in both HNPCC and FAP
families that lack any sequence changes for their respective known
susceptibility genes. These authors suggest that these genes either have
alterations that have escaped detection by conventional techniques, or
alternatively, other, as yet unknown, susceptibility genes are involved.
Furthermore, they suggest that significant proportions of families that are
presumed to be mutation negative (11 of 26 families in their study, or
42% for Lynch syndrome; 4 of 16 families, or 25% for FAP) harbor
“hidden” alterations in known predisposition genes, leading to their
Curr Probl Surg, May 2005
279
conclusion that such changes may be detected by expression-based
methods.354,355
Founder Mutations in Lynch Syndrome. De la Chapelle and Wright356
evaluated 2 founder mutations in the hMLH1 gene in Finland, which
accounted for approximately one half of all Lynch syndrome germline
mutations in that country.357,358 They identified haplotype sharing “. . .
over a genomic region as large as 18 cM indicated a relatively recent
founding of the more prevalent mutation. . . wherein the ‘age’ of this
mutation in most of the 19 kindreds studied could be estimated at 16-43
generations in keeping with historical records and compatible with a
founding in a regional subisolate in new Finland in the early
1500s.”357,359 This research, once the mutations became more fully
understood and characterized, indicated that their presence in a patient or
family could identify individuals who could benefit significantly from
highly targeted cancer control programs.360
A founder mutation consisting of a 4-bp deletion beginning at the first
nucleotide of codon 727 in hMLH1 has been identified in Navajo families
from Arizona. A deletion encompassing exons 1-6 of the MSH2 gene has
been identified in an extended American kindred of German origin.315
Foulkes and colleagues361 described a founder mutation of
hMSH2(1906G¡C) in the Ashkenazi Jewish population. This mutation
results in a substitution of proline for alanine at codon 636 in the MSH2
protein and was identified in 15 unrelated Ashkenazi Jewish families with
Lynch syndrome who met the Amsterdam criteria. These families shared
the same alleles with 18 polymorphic loci within and flanking the MSH2
region, which was consistent with a single origin for this mutation.
Through immunohistochemical analysis, all CRCs that were tested were
found to harbor MSI and absence of the MSH2 protein. These authors
then provided an analysis of a population-based incidence series of 686
Ashkenazi Jews from Israel who manifested CRC. Their results showed
that 3 (0.44%) were mutation carriers. Furthermore, those patients with a
family history of CRC or endometrial cancer were more likely to harbor
the mutations than those without such a family history (P ⫽ 0.042).
Individuals with CRC who were mutation carriers were, on average,
younger than CRC-affected individuals who were noncarriers (P ⫽
0.033). Five hundred sixty-six unaffected Ashkenazi Jews from Israel and
1022 controls from New York were all found to be noncarriers. In
hospital-based series, the 1906 allele was identified in 5 of 463 Ashkenazi
Jews with CRC, 2 of 197 with endometrial cancer, and 0 of 83 with
ovarian cancer.
Guillem and colleagues362 identified this rare founder mutation, hMSH2
280
Curr Probl Surg, May 2005
1906C¡G, in Ashkenazi Jews in 8 of 1345 individuals (0.6%) with CRC.
They then investigated the proportion of individuals of Ashkenazi
heritage who manifested early-onset CRC (age ⱕ 40 years) that could be
explained by hMSH2 1906C¡G. They detected this mutation in 3 of the
41 samples (7.14%) among Ashkenazi patients with CRC diagnosed
before age 40, an incidence found to be significantly greater than the 8 in
1345 (0.6%) that was observed in CRC among Ashkenazi Jews who were
not selected for age (P ⫽ 0.004). They concluded that their results merit
testing for the hMSH2 1906C¡G mutation among Ashkenazi Jews
manifesting early-onset CRC.362
Strategy for Genetic Testing. As in other autosomal dominant
conditions, the affected proband should be studied thoroughly for
mutations to allow for directed testing later in other members at risk.
This should be performed in patients meeting the Amsterdam criteria,
or in those who are suspected of having Lynch syndrome but do not
meet the criteria (as in the Bethesda guidelines). The Lynch syndrome
patient’s tumor should first be tested for MSI, and testing for MSH2 or
MLH1 expression by immunohistochemistry can also be quite helpful
in determining which gene is most likely affected in the family. If MSI
is not found, then sequencing of MMR genes is not warranted. If the
tumor is MSI-positive or loss of protein expression is found, then
sequencing of hMLH1 and/or hMSH2 is indicated. The frequency of
PMS gene mutations is so low that their sequencing is not generally
performed; hMSH6 sequencing may be indicated in cases with a
family history suggestive of Lynch syndrome where tumors are
MSI-low or there is a high incidence of endometrial cancer. In rare
cases, there may be mutations in PMS2, MLH3, and EXO1.363
Lynch Syndrome Versus Early Onset Sporadic Colon Cancer. Even in
those cases in which the Amsterdam criteria have not been met, Jass has
suggested that the profile of early-onset MSI-high (MSI-H) CRCs
resembles that of Lynch syndrome cancer, with emphasis on the histologic features and site of tumors. Furthermore, Jass provides 5 cogent and
interrelated reasons for questioning the presumption that early-onset
“sporadic” MSI-H CRCs are truly sporadic. Specifically, he notes that
“. . . first is the fact that the incidence of HNPCC peaks at around 45
years. Second is the finding of germline mutations in DNA mismatch
repair genes in subjects presenting with early-onset “sporadic” MSI-H
colorectal cancer. Third is the evidence that methylation of hMLH1 in
sporadic MSI-H cancer is strongly age-related. Fourth is the fact that
methylation of hMLH1 may occur selectively in HNPCC cancers in
subjects who carry a germline mutation in hMLH1. Fifth is the finding of
Curr Probl Surg, May 2005
281
HNPCC-type molecular features among early-onset “sporadic” MSI-H
colorectal cancers.”364
Pathologic Features in Lynch Syndrome. The pathologic features of
Lynch syndrome CRCs include a solid growth pattern, which appears to
be responsible for the high frequency of poorly differentiated carcinomas.365,366 Surprisingly, however, is the finding that CRCs in the Lynch
syndrome are not as aggressive as their failure to form tubules might
suggest. Lynch and colleagues have called attention to the “undifferentiated carcinoma” described by Gibbs367 and the “medullary carcinoma”
described by Jessurun and Manivel,368 which were published in small
case series and shown to have a more favorable prognosis when compared
with typical CRC.369 It is also noteworthy that similar histologic features
characterize the 15% of sporadic CRCs that harbor microsatellite instability (MSI⫹), which have been found to constitute a molecular aberration in CRCs that lack MMR activity.370 This special histologic feature
has been referred to as “solid cribiform growth” wherein there is a
positive predictive value of 53% for MSI⫹ status.371
Smyrk and colleagues372 described a postlymphoid response referred to
as “Crohn’s-like reaction” to be more common in Lynch syndromes as
opposed to sporadic cancers, a finding that has been shown to be
consistent in all series,365 but which is similar to the tendency to form
lymphoid aggregates around the tumor, and which is a frequent feature of
sporadic MSI⫹ CRCs.370 Importantly, the Crohn’s-like reaction is often
associated with improved prognosis373 in the general population, suggesting that this phenomenon may account for the more favorable prognosis
of CRC that occurs in HNPCC.374 De Jong and colleagues375 studied the
role of MMR defects and the manner in which they promote development
of adenomas in Lynch syndrome families compared with controls from
the Dutch HNPCC Registry. They identified 249 MMR mutation carriers
and 247 controls. They found that the proportion of patients lacking an
adenoma at the age of 60 years was 29.7% for carriers and 70.8% for
controls (P ⬍ 0.05). They verified previous studies showing that the
adenomas in carriers were larger and showed a higher proportion of
villous components and/or high-grade dysplasia (P ⬍ 0.05, all analyses).
Furthermore, they found the adenomas and carcinomas of the carriers to
be located predominantly in the proximal colon, and most of the
adenomas showed absent staining of the MMR proteins. They concluded
that their study “. . . indicates that the MMR defect is involved in the early
stages of development of adenomas.” They recommended immunohistochemical staining of large adenomas showing high-grade dysplasia in
282
Curr Probl Surg, May 2005
patients younger than 50 years old, in the interest of identifying patients
with suspected Lynch syndrome.375
Pattern Recognition. An alternative to the Amsterdam I, Amsterdam II,
Bethesda, and other criteria is what we have described as “pattern
recognition.” This involves careful scrutiny of the phenotypic cancer
features expressed in the family in accord with their mode of distribution
and transmission throughout the family. This is particularly important
when evaluating small families, or for problems of reduced penetrance of
the deleterious MMR mutation, should this be present in the family. It is
valuable then to follow what have been considered to be the cardinal
features of hereditary forms of cancer, including the Lynch syndrome,
when present even in a single patient. These phenotypic features have
often been sufficient to raise the confidence level of a possible Lynch
syndrome diagnosis and include the following: 1) autosomal dominant
inheritance pattern311; 2) gene penetrance for CRC of ⬃85% to 90%304;
3) gene carriers develop CRC at an early age (⬃45 years)311; 4) most
(⬃70%) of the CRCs are proximal to the splenic flexure376; 5) multiple
CRCs, both synchronous and metachronous, are common377,378; 6) the
prognosis is better than that for sporadic CRC374,379; 7) the pathologic
features365 of CRC are often distinguishable (but not pathognomic) and
include poor differentiation, mucoid excess with increased signet cells,
medullary features, peritumoral lymphocytic infiltration, Crohn’s-like
reaction, and tumor infiltrating lymphocytes (TILs) admixed with tumor
cells380; and 8) there is an increased risk for malignancy at several
extracolonic sites, particularly the endometrium, ovary, stomach, small
bowel, hepatobiliary tract, pancreas, ureter, renal pelvis, and brain.306 In
addition, breast cancer excess may be present in some HNPCC families.381 Although gastric cancer has declined in Lynch syndrome families
since 1900, paralleling its decline in the general population,312 it is still an
important feature of the syndrome in geographic areas in which gastric
cancer is endemic, most notably Japan and Korea.305
Muir-Torre Syndrome (MTS). In 1981, Lynch and colleagues382
reported the first observation of MTS in the Lynch syndrome. MTS is
characterized by multiple cutaneous sebaceous adenomas, sebaceous
carcinomas, multiple keratoacanthomas, and multiple visceral cancers, in
Lynch syndrome families. Several publications have elucidated the
clinical and molecular genetic features of MTS in the Lynch syndrome.383-386 Kruse and colleagues387 examined 13 patients with CRC
and sebaceous lesions of the skin and found that 8 had hMSH2 mutations
and 1 had an hMLH1 mutation. Data suggest that the identification of
these MTS cutaneous features in a patient merit a detailed family history
Curr Probl Surg, May 2005
283
in the search for evidence of the Lynch syndrome. Indeed, patients with
these stigmata merit MMR germline testing, particularly for evidence of
hMSH2 germline mutation. The pedigree of an extended Lynch syndrome
family with MTS that is known to carry an hMSH2 mutation is shown in
Fig 20.
Surgical Management. In Lynch syndrome patients diagnosed with
colorectal cancer, the preferred treatment is subtotal colectomy. Fitzgibbons and colleagues388 studied 116 patients with Lynch syndrome and
demonstrated several significant differences in this patient population
relative to patients with sporadic CRC. They found that 69% of Lynch
syndrome CRC were proximal to the splenic flexure, whereas 23% were
in the rectosigmoid. Eighteen percent had synchronous cancers, and 40%
developed metachronous lesions over 10 years of follow-up.388 Because
of the predominance of proximal lesions and the high risk of developing
new cancers in the future, subtotal colectomy with IRA and lifetime
surveillance of the rectosigmoid is recommended. For women beyond
childbearing age, serious consideration should also be given to performing prophylactic hysterectomy and bilateral salpingo-oophorectomy.388
In reviewing this subject, Church389 suggests that interval CRCs
develop from normal epithelium within 3 years and/or from adenomas
that were missed. It is also important to realize that colonoscopy “miss”
rates are as high as 29% for polyps less than 5 mm in diameter.390
Therefore, patients should be advised that colonoscopy, although not a
perfect screening procedure, is nevertheless highly effective.360 The
option of prophylactic colectomy should be discussed, weighing it against
lifetime colonoscopy.391-394 The problem in effective cancer control
through the use of prophylactic colectomy is a decision based heavily on
the attitudes, feelings, and concerns of the patient and the manner in
which these could impact on his/her compliance with annual colonoscopy. For example, the presence of fear and anxiety about what might be
found at colonoscopy, in our experience, has deterred patients in showing
compliance with this screening procedure for CRC. Criteria for prophylactic surgery are the following: 1) lack of compliance with surveillance;
2) morbid fear and desire to have these high risk organs surgically
excised; 3) presence of colonic adenomas, particularly at a young age
and/or with features of moderate to severe dysplasia and carcinoma in situ
in polyps.
Women at risk for the Lynch syndrome should have annual screening
for endometrial and ovarian cancer beginning at age 30 to 35 years.
Endometrial aspiration coupled with transvaginal ultrasound is advised.
CA-125 testing, in addition to transvaginal ultrasound and Doppler color
284
Curr Probl Surg, May 2005
Curr Probl Surg, May 2005
FIG 20. Pedigree shows the Muir-Torre syndrome.
285
blood flow imagery, should be performed semiannually for ovarian
cancer. Prophylactic hysterectomy and oophorectomy can be considered
when childbearing is completed.
Screening in Lynch Syndrome. Jass395 has elucidated the pathologic
nature of precursor lesions in HNPCC. He has postulated the “aggressive adenoma” theory (i.e., adenomas form earlier but about as often
in HNPCC patients as in the general population).395 However, once
formed, these colonic adenomas progress to carcinoma more quickly
and/or more often than their sporadic counterparts. A tiny colonic
adenoma in the Lynch syndrome may evolve into a carcinoma in as
short a time frame as 2 to 3 years, compared with approximately 8 to
10 years in the general population.303,365,396 Strong clinical evidence
in support of a more rapid evolution of adenoma to carcinoma comes
from a Finnish study showing a marked decrease in colon cancer
incidence for HNPCC patients given regular colonoscopic surveillance
with removal of adenomas.397 Because of accelerated carcinogenesis,
proximal colonic predilection, and early age of CRC onset in the
Lynch syndrome, we strongly recommend that annual full colonoscopy be initiated at age 25 (Fig 21). A good clean-out is necessary and
excellent visualization of the cecum is mandatory, because approximately one third of CRCs occur at that particular site.
Järvinen and colleagues360 demonstrated the benefit of colonoscopic
screening in HNPCC through a controlled clinical trial extending over
15 years. The incidence of CRC was compared in 2 cohorts of at-risk
members of 22 HNPCC families. CRC developed in 8 screened
subjects (6%), compared with 19 controls (16%; P ⫽ 0.014). The CRC
rate was reduced by 62% in those who were screened. All CRCs in the
screened group were local, causing no deaths, compared with 9 deaths
caused by CRC in the controls. It was concluded that CRC screening
at 3-year intervals more than cuts in half the risk of CRC, prevents
CRC deaths, and decreases the overall mortality rate by approximately
65% in HNPCC families.360 The relatively high incidence of CRC
even in the screened subjects (albeit without deaths) in our opinion
argues for shorter screening intervals (i.e., 1 year). For example,
Vasen and colleagues398 discovered 5 interval cancers in HNPCC
patients within 3.5 years following a normal colonoscopy. Prophylactic colectomy, as discussed, is an alternative to intensive colonoscopic
surveillance in highly selected members of Lynch syndrome families.
An algorithm showing the appropriate sequence for diagnosis and
management of Lynch syndrome is shown in Figure 21.
286
Curr Probl Surg, May 2005
FIG 21. Algorithm depicting the appropriate sequence for A) diagnosis and management and B)
screening and management of the Lynch syndrome. CRC, colorectal cancer; MSI, microsatellite
instability; MMR, mismatch repair.
Juvenile Polyposis
The colorectal polyps most commonly found in children are different
from those found in adults and are called juvenile polyps. Polyps in adults
are generally adenomatous, whereas polyps in children tend to be
hamartomatous (juvenile polyps being a type of hamartomatous polyp). In
most children, the juvenile polyps are few in number or solitary, will
slough off and not require treatment, and are not associated with a
heritable predisposition. Juvenile polyposis (JP; OMIM 174900) is a
heritable syndrome in which there are multiple juvenile polyps (Fig 22).
Patients with juvenile polyposis, Cowden syndrome (CS; OMIM
158350), and Bannayan-Riley-Ruvalcaba syndrome (BRRS; OMIM
153480) may all develop juvenile polyps of the GI tract, which are
indistinguishable microscopically. Patients with Peutz-Jeghers syndrome
(PJS; OMIM 175200) also have hamartomatous polyps, but these can be
differentiated histologically from juvenile polyps.
Curr Probl Surg, May 2005
287
FIG 21. Continued
288
Curr Probl Surg, May 2005
FIG 22. Juvenile polyposis of the colon.
History. The first description of a juvenile polyp was by Diamond399 in
1939, who originally called this a rectal adenoma. Diamond399 described
it grossly as a 2.5 by 2.0 cm pedunculated tumor that was injected and
friable. One small area on the polyp surface was covered with a fibrinous
exudate. Histologically, the polyp was lined with high columnar epithelial
cells with an occasional bare area. The glands were distended and filled
with a fibrinous cellular material. The stroma was diffusely infiltrated
with lymphocytes, plasma cells, eosinophils, and neutrophils.399 In 1957,
Mauro and Prior400 clearly distinguished between juvenile polyps and
adenomatous polyps. Contrasting the 2, they found juvenile polyps to be
globular in shape, gray to slightly red, with a granular external surface.
Villous processes were not present. It was common to have areas of
denudation of the columnar epithelium replaced by granulation tissue.
There was cystic gland dilation, containing mucous and nuclear debris.
The abundant stroma contained an inflammatory cell infiltrate with a
predominance of eosinophils.400 Later in the same year, Horrilleno and
colleagues401 coined the term juvenile polyp for these histologically
similar polyps described previously by Diamond and by Mauro and Prior.
Curr Probl Surg, May 2005
289
FIG 23. Photomicrograph of a juvenile polyp. Note the normal epithelium, with a markedly expanded
lamina propria, infiltrated by inflammatory cells and dense stroma. Cystically dilated glands are also
prominent in this layer.
In 1962, Morson402 supported the view that juvenile polyps were different
from adenomatous polyps and suggested they were hamartomatous
polyps. Hamartomas are an overgrowth of mature cells and tissues that
normally occur in the affected part, but often with 1 element predominating. Juvenile polyps satisfy this definition with their excessive proliferation of the lamina propia and attenuation of the muscularis mucosa
(Fig 23).
Definition and Manifestations. The term juvenile polyposis (JP) was
defined by McColl and colleagues403 in 1964. This distinguished juvenile
polyps from JP, a heritable syndrome in which there are multiple juvenile
polyps involving the GI tract. In 1974, following the definition of JP by
McColl, Sachatello and colleagues404 proposed a working definition, later
modified by Jass and colleagues,405 with the following diagnostic criteria:
1) more than 5 juvenile polyps involving the colon and rectum, 2) juvenile
polyps throughout the GI tract, and 3) any number of juvenile polyps
involving the GI tract with a family history of JP. JP has been further
divided based on the clinical presentation and disease course into the
following three classifications404,405: 1) juvenile polyposis of infancy, 2)
290
Curr Probl Surg, May 2005
FIG 24. Juvenile polyposis affecting the stomach, resulting in a dense carpet of polyps.
juvenile polyposis coli, and 3) generalized juvenile polyposis. Sachatello
and colleagues defined juvenile polyposis of infancy in a case report and
reviewed 6 similar cases in the literature.404,406-410 In all 7 cases, the
infants died before 1 year of age. Characteristic symptoms included
diarrhea (usually bloody), protein losing enteropathy, hypoproteinemia,
anemia, anasarca, and eventually failure to thrive. It was common to find
rectal polyp prolapse and intussusception. Congenital anomalies ranged in
these series from none to clubbing, motor retardation, macrocephaly,
alopecia, double renal pelvis, and bifid uterus/vagina.404
Juvenile polyposis coli and generalized juvenile polyposis are defined
by the sites of GI involvement with juvenile polyps. As the name implies,
juvenile polyposis coli occurs when juvenile polyps are limited to the
colon and rectum, whereas in generalized juvenile polyposis, the polyps
are present in both the upper and lower GI tract (Fig 24). Both subtypes
will usually manifest in the first and second decades with rectal bleeding,
a prolapsed rectal polyp, abdominal cramps or pain, diarrhea, or aneCurr Probl Surg, May 2005
291
mia.411,412 In addition, generalized GI juvenile polyposis may include
protein-losing enteropathy, intussusception, and severe malnutrition.
However, these children, unlike those with juvenile polyposis of infancy,
eventually recover. Coburn and colleagues413 reviewed the English
literature and identified 218 patients meeting the criteria for JP. These
patients with JP were stratified into 2 groups: juvenile polyposis coli and
generalized juvenile polyposis. Patients with juvenile polyposis coli
usually presented during childhood between the ages of 5 and 15 years,
whereas patients diagnosed with generalized juvenile polyposis present at
an earlier age. Otherwise, there did not appear to be any difference in the
family history of juvenile polyposis in either subtype.413 It should be
mentioned that the distinction between juvenile polyposis of infancy,
juvenile polyposis coli, and generalized juvenile polyposis may be
somewhat arbitrary. Stemper and colleagues414 reported a kindred with
some members having generalized GI juvenile polyposis whereas others
had juvenile polyposis coli, suggesting the phenotype of juvenile polyposis is of variable penetrance. Similarly, Grotsky and colleagues415
described an extended family with both juvenile polyposis coli and
juvenile polyposis of infancy.
Associated Anomalies. In their 1964 series, McColl and colleagues403
described 4 of 11 patients with associated congenital anomalies. Coburn
and colleagues413 reported nearly a 15% prevalence of congenital
malformations with JP. Interestingly, Desai and colleagues,416 who
routinely obtained radiographs and an echocardiogram if clinically
indicated, reported 18 of 23 (78%) patients with JP having extracolonic
manifestations. This number is clearly skewed since after the extensive
re-evaluation 5 patients had alternative syndromes diagnosed, and an
unspecified number were described as having similarities to known
syndromes. Removing these cases leaves 8 of 13 (62%) patients with
presumed JP associated with extracolonic manifestations. Throughout
the JP literature, some of the recurrent anomalies include the following: macrocephaly, mental retardation, atrial and ventricular septal
defects, pulmonary arteriovenous malformations, pulmonary stenosis,
Meckel’s diverticulum, malrotation, cryptorchidism, hypertelorism,
and telangectasias.403,413,416
Malignant Potential. An extensive portion of the literature on JP has
been dedicated to the discussion of its malignant potential. It was initially
thought that JP did not predispose to GI malignancy, since juvenile polyps
are hamartomatous. As it became apparent that adenomatous polyps had
malignant potential, an increased prevalence of colorectal cancer was
recognized with JP patients. During this time adenomatous changes in
292
Curr Probl Surg, May 2005
juvenile polyps were recognized414,415,417-434 and hypothesized occasionally to undergo dysplastic change and eventually progress to adenocarcinoma. In 1979, Goodman and colleagues422 reported a 23-year-old
woman undergoing a colectomy with numerous juvenile polyps mainly
localized to the cecum, ascending colon, and rectum. Several of the
juvenile polyps showed adenomatous changes, and an adenocarcinoma
had also developed in the rectum.
After adenomatous changes, the next step in the progression to
malignancy would be dysplasia/carcinoma in situ changes in the juvenile
polyps of patients with JP. Jass and colleagues405 reviewed the pathologic
features of 1038 polyps collected from 80 patients in the St. Mark’s
Polyposis Registry with JP. They divided the polyps into 2 groups: 1)
multilobulated or having a villous configuration, and 2) typical juvenile
polyps. Seventy-nine of 169 (47%) atypical juvenile polyps had epithelial
dysplasia, whereas 76 of the 840 (9%) typical juvenile polyps had similar
dysplastic changes.405 Several other investigators have reported the
finding of dysplastic changes in juvenile polyps in JP patients.428,435,436
There have been many cases of documented colorectal adenocarcinoma
reported in patients with JP.414,422,427,428,430-432,437,438 These cancers are
often closely associated with juvenile polyps and juvenile polyps with
adenomatous changes, supporting a juvenile polyp-adenomatous changesdysplasia-carcinoma sequence of events.
The establishment of JP registries has allowed estimates of the cumulative risk for developing GI malignancies to be made. Howe and
colleagues439 examined a 117-member Iowa JP family and found that 11
of the 59 (38%) affected kindred members developed colon cancer,
whereas 6 of 59 (21%) developed upper GI cancers. The cumulative risk
for upper and lower GI cancers was 55%. The cumulative risk for
colorectal cancer in JP patients has ranged from 17% to 68% in other
reports.413,440
The distinction between JP and patients with juvenile polyps is
important when estimating the risk of GI malignancy. Many authorities
have suggested that there is no inherent increased risk of GI malignancy
for patients with solitary juvenile polyps not meeting the definition of JP.
Giardello and colleagues432 found that only 1 of 26 patients with only 1
or 2 juvenile polyps and a negative family history had adenomatous
changes in a polyp, whereas 9 of 31 (29%) patients with 3 or more
juvenile polyps or a positive family history of JP had colonic adenomas
or adenocarcinoma. There is only 1 reported case of an adenocarcinoma
arising from a presumed solitary juvenile polyp in a 16-year-old boy.441
Nugent and colleagues442 reported that the relative risk of dying of
Curr Probl Surg, May 2005
293
colorectal malignancy in a patient with a solitary juvenile polyp was 0.66,
and therefore these patients are not at an increased risk of death from
colorectal carcinoma.
Genetics. The autosomal dominant nature of JP was first suggested by
Smilow and colleagues437 and Veale and colleagues443 in 1966. Further
descriptions of kindreds with juvenile polyposis confirmed an autosomal
dominant inheritance.414,444 More recent investigation has shifted from
the clinical to the molecular basis of JP. Jacoby and colleagues described
a patient in 1997 with JP having a deletion at chromosome 10q22.445
Soon thereafter, Olschwang and colleagues446 reported 3 cases of JP with
germline PTEN mutations, which maps to 10q22. However, on closer
evaluation by Eng and Ji,447 these patients either had a phenotype
suggestive of CS or, due to their young age, CS could not be ruled out.
Howe and colleagues448 examined a family of 43 individuals of which 13
had the clinical diagnosis of JP and found no linkage to chromosome 10q
markers. Marsh and colleagues449 were unable to identify germline
mutations in PTEN in 14 familial and 11 sporadic JP cases. Riggins and
colleagues450 also found no PTEN mutations in 11 JP patients. In 1998,
Howe and colleagues448 demonstrated genetic linkage to markers on
chromosome 18q21, and subsequently reported germline mutations of
MADH4 in 5 of 9 JP cases.451 This was independently confirmed in
multiple small series.452-455 Woodford-Richens and colleagues456 found
7 of 44 (16%) cases of JP with MADH4 mutations, and Howe and
colleagues457 reported mutations in 14 of 77 JP cases (18.2%). In the
latter report, 8 mutations were deletions and 6 were substitutions (1
nonsense, 5 missense). Reviewing all published mutations, Howe and
colleagues457 described a total prevalence of 23% (32 of 141 cases with
MADH4 mutations). One mutation had been described in 6 cases, a 4-bp
deletion in exon 9.458 Of 32 total mutations, 1 mapped to the MH1
domain, 5 to the linker region and the remainder to the MH2 domain (Fig
25).457
The MADH4 protein is the common mediator involved in the transforming growth factor-␤ (TGF-␤) superfamily, which includes TGF-␤,
activin, and bone morphogenetic protein (BMP) signal transduction
pathways. Functionally, MADH4 acts as a tumor suppressor gene, as
demonstrated by Hahn and colleagues,459 who found loss of heteorozygosity (LOH) of MADH4 in 25 of 84 (30%) pancreatic xenografts.
Members of the TGF-␤ superfamily initiate a wide spectrum of effects on
a variety of cell types, including control of differentiation, proliferation,
and apoptosis. TGF-␤ binds to plasma membrane serine/threonine kinases, specifically the type II TGF-␤ receptor. TGF-␤ and the type II
294
Curr Probl Surg, May 2005
FIG 25. MADH4 mutations reported in juvenile polyposis (JP). The upper rectangle represents the
MADH4 gene, with the exons shown within the rectangle and nucleotides above and below. Lines
designate mutations identified in JP cases, with those shown above each exon corresponding to those
identified by Howe and colleagues,457 whereas those below are derived from the reports of Roth and
colleagues,454 Kim and colleagues,455 Woodford-Richens and colleagues,456 and Friedl and
colleagues.464 (Reproduced with permission from Howe JR, Sayed MG, Ahmed AF, Ringold J,
Larsen-Haidle J, Merg A, et al. The prevalence of MADH4 and BMPR1A mutations in juvenile
polyposis and absence of BMPR2, BMPR1B, and ACVR1 mutations. J Med Genet 2004;41:484-91.)
TGF-␤ receptor complex then binds with the type I receptor, causing
phosphorylation in a glycine-serine-rich domain of the receptor (Fig 26).
In the TGF-␤ pathway, these activated type I receptors then phosphorylate cytoplasmic MADH2 or MADH3. Phosphorylation allows these
proteins to oligomerize and then to associate with MADH4. The complex
then migrates to the nucleus, where it recruits a DNA binding protein, and
the complex binds to specific DNA sequences, regulating the transcription
of various genes, many of which remain to be identified. The MH2
domain of MADH4, which is frequently altered in JP patients and
sporadic pancreatic cancers, is important in nuclear localization, MADH4
binding, and activation of transcription. There are 8 known MADHs in
Curr Probl Surg, May 2005
295
FIG 26. Overview of the TGF-␤ signaling pathway (reproduced with permission from Merg A, Howe
JR. Genetic conditions associated with intestinal juvenile polyps. Am J Med Genet 2004;129C:4455).577
vertebrates, with MADH2 and MADH3 functioning as the cytoplasmic
effectors in the TGF-␤ and activin pathways; MADH1, MADH5, and
MADH8 are those involved in the BMP pathway. MADH4 is the
common mediator for these pathways, whereas MADH6 and MADH7
function as inhibitors of all these pathways by binding to the type I
receptors and by interfering with phosphorylation.460 To date, no other
MADH genes have been implicated in the development of JP. Roth and
colleagues454 found no mutations in MADH2, MADH3, or MADH7 in 4
unrelated patients without MADH4 mutations, and Bevan and colleagues461 found no mutations in MADH1, MADH2, MADH3, or MADH5
in 30 JP patients without MADH4 mutations.
Since MADH4 mutations only accounted for one fifth of JP cases, Howe
and colleagues462 searched for another JP gene through a linkage-based
296
Curr Probl Surg, May 2005
genome screen in 4 additional JP families. They found linkage to markers
on chromosome 10q22-23, and subsequently each family was found to
have a truncating mutation in the BMPR1A gene.462 Since the initial
report of BMPR1A mutations in JP patients, there have been 2 confirmatory studies published.463,464 In a total of 77 JP cases studied by Howe
and colleagues, there have been 16 (21%) with BMPR1A mutations,
consisting of 6 deletions and 10 substitutions (4 nonsense, 6 missense).457
Pooling all 3 reports, 31 unique mutations have been described, distributed in 8 of 11 exons with nearly one half affecting the intracellular
protein kinase region in exons 7 and 8. One quarter of mutations affect the
extracellular cysteine-rich domain (Fig 27).457 BMPR1A is another
member of the TGF-␤ superfamily involved in a pathway that also
depends on MADH4 as the intracellular mediator of signal transduction
(Fig 26). Like TGF-␤, the various BMPs bind to specific type II receptors
or to the type II and type I receptors together, then the type II receptor
phosphorylates a type I receptor, such as BMPR1A or BMPR1B.465 The
type I/type II receptor complex then phosphorylates MADH1, MADH5,
and/or MADH8, which then form oligomers with MADH4, and these
proteins migrate to the nucleus to regulate transcription of specific
genes.460 BMPs exert many effects on a variety of tissues, including
control of osteogenesis, chondrogenesis, mesoderm development, synthesis of extracellular matrix, and epithelial/mesenchymal cell relationships
contributing to morphogenesis.466
A recent study examining MADH4 and BMPR1A mutation-positive
(MUT⫹) and negative (MUT⫺) cases found that that 17 of 19 (89%)
MUT⫹ patients and 13 of 25 (52%) MUT⫺ patients cases had a family
history of GI cancer (P ⫽ 0.01), and that 89% of MUT⫹ and 63% of
MUT– cases were familial (P ⫽ 0.09).467 The familial prevalence of
upper GI juvenile polyps was 86% in MADH4 mutation-positive JP
families, 10% in BMPR1A families, and 23% in MUT– cases. Friedl and
colleagues464 reported gastric polyposis in 4 of 7 MADH4 mutationpositive cases, in 1 of 5 cases with BMPR1A mutations, and 2 of 17
mutation-negative cases. It therefore appears that MADH4 mutations
predispose to generalized juvenile polyposis, whereas MADH4 mutationnegative cases more commonly cause juvenile polyposis coli. Since
MADH4 and BMPR1A mutations only account for 40% of JP cases, there
are likely other genes responsible for JP that await discovery.
Treatment. Because of the malignant potential of juvenile polyps in JP,
endoscopic screening and polypectomy, and prophylactic surgery in some
cases, are very important means of cancer control. Screening should begin at
age 15, and include esophagogastroduodenoscopy, colonoscopy, and a
Curr Probl Surg, May 2005
297
FIG 27. BMPR1A mutations reported in juvenile polyposis (JP). The upper rectangle represents the
BMPR1A gene, with the exons shown within the rectangle and nucleotides above and below. Lines
designate mutations identified in JP cases, with those shown above each exon corresponding to those
identified by,457 while those below are derived from the reports of.463,464 The lower rectangle
represents the different known domains of the BMPR1A gene (reproduced with permission from Howe
JR, Sayed MG, Ahmed AF, Ringold J, Larsen-Haidle J, Merg A, et al. The prevalence of MADH4 and
BMPR1A mutations in juvenile polyposis and absence of BMPR2, BMPR1B, and ACVR1 mutations.
J Med Genet 2004;41:484-91).
hemogram in those at risk for JP.468 If a mutation of MADH4 or BMPR1A is
found in a family, then the process is simplified by testing individuals at risk
for the same mutation. If no mutation is found, then that individual can be
followed by using the guidelines for screening the general population for
sporadic colorectal cancer. If either a mutation is found in an at-risk patient,
or a mutation has not been found in the family, then colonoscopy and upper
endoscopy should be performed every 3 years. When juvenile polyps are
found in the colon, they should be removed endoscopically, which may
require multiple sessions if the number is greater than 20 to 50 polyps.
298
Curr Probl Surg, May 2005
TABLE 10. International Cowden Consortium diagnostic criteria*
Pathognomonic criteria
Facial trichilemmomas
Acral keratoses
Papillomatous papules
Mucosal lesions
Major criteria
Breast carcinoma
Thyroid carcinoma, nonmedullary
Macrocephaly
Lhermitte-Duclos disease (LDD)
Endometrial carcinoma
Minor criteria
Thyroid lesions, adenoma or multinodular goiter
Mental retardation
Gastrointestinal hamartomas
Fibrocystic disease of the breast
Lipomas
Fibromas
Genito-urinary tumors (renal cell carcinoma or uterine fibroids) or malformations
Operational diagnosis in an individual
1) Mucocutaneous lesions alone if there are
i) Six or more facial papules, of which 3 or more must be trichilemmomas
ii) Cutaneous facial papules and oral mucosal papillomatosis
iii) Oral mucosal papillomatosis and acral keratoses
iv) Six or more palmar or plantar keratoses
2) Two major criteria, 1 of which must be macrocephaly or LDD
3) One major and 3 minor criteria
4) Four minor criteria
Operational diagnosis in a family in which 1 individual has had a diagnosis of Cowden
syndrome
1) One or more of pathognomonic criteria
2) Any 1 major criterion with or without minor criteria
3) Two minor criteria
*Reproduced with permission from Waite KA, Eng C. Protean PTEN: form and function. Am J
Hum Genet 2002;70:829 – 44.612
Subsequently, annual endoscopy should be performed until the all polyps are
removed, and then every 3 years after this if no dysplastic polyps are found.
If the colon cannot be rendered polyp free using the endoscope, then surgical
management may be needed. The range of surgical options include segmental
colectomy, subtotal colectomy, and total proctocolectomy with ileoanal
pull-through.468 Since the malignant potential of juvenile polyps in JP is low
relative to more aggressive polyposis syndromes, one must consider carefully
the impact of the chosen procedure on quality of life versus the need for
future endoscopic surveillance. Subtotal colectomy is preferred over segmental colectomy since it limits the amount of colon at risk, and patients can be
screened easily by flexible sigmoidoscopy. Total proctocolectomy should be
Curr Probl Surg, May 2005
299
FIG 28. A and B) Trichilemmomas of the face in patients with Cowden syndrome (reproduced with
permission from Brownstein MH, Wolf M, Bikowski JB. Cowden’s disease: a cutaneous marker of
breast cancer. Cancer 1978;41:2393-8).578
reserved for those with significant involvement of both the colon and the
rectum, or those with dysplastic rectal polyps. Polyps of the stomach are
usually diffuse and cannot be effectively removed endoscopically. Significant
anemia is often present, and these patients may require subtotal or total
gastrectomy.468 Patients with JP or at risk for JP should receive genetic
counseling. Here, recommendations for endoscopic screening, the risk of
upper and lower GI symptoms and cancer, and the ramifications of genetic
testing should be discussed.
Cowden Syndrome
Cowden Syndrome (CS) was first described by Lloyd and Dennis469 in
the propositus Rachel Cowden, and Weary and colleagues470 later
300
Curr Probl Surg, May 2005
FIG 28. Continued
designated CS as a multiple hamartoma syndrome after observing its
autosomal dominant inheritance in her family. CS involves hamartomatous changes in all 3 germ layers, including predominantly the skin,
mucous membranes, breast, thyroid, GI tract, and CNS. The International
Cowden Consortium has established a group of features that establish the
diagnosis of CS (Table 10). Lhermitte and Duclos disease (LDD) is
included in these criteria, which causes a variety of CNS symptoms.471,472 In the 1990s, it was realized that LDD and CS were variants
of the same genetic condition, which manifest as neurologic and cutaneous hamartomas, respectively.473-477
Phenotype. Lesions of the skin and mucous membranes are pathognomonic for CS, and are present with nearly 100% prevalence.478,479
Trichilemmoma, 1 of the characteristic lesions in CS, is a hamartoma of
the hair follicle’s root sheath. Typically trichilemmomas are found around
the eyes, nose, and mouth (Fig 28). Oral fibromas with a papillomatosis
morphology can cause a cobblestoning appearance of the gingiva and/or
tongue, also referred to as a scrotal tongue (Fig 29). Acral keratosis is also
prominent in CS (Fig 30).478,480,481 Three quarters of patients with CS
have thyroid abnormalities, most commonly goiters or adenomas. NonCurr Probl Surg, May 2005
301
FIG 29. A) Papillomatous lesions of the tongue seen in a patient with Cowden syndrome (CS)
(reproduced with permission from Botma M, Russell DI, Kell RA. Cowden’s disease: a rare cause of
oral papillomatosis. J Laryngol Otol 2002;116:221-3).579 B) Gingival papules in a CS patient
(reproduced with permission from Sogol PB, Sugawara M, Gordon HE, Shellow WV, Hernandez F,
Hershman JM. Cowden’s disease: familial goiter and skin hamartomas. A report of three cases. West
J Med 1983;139:324-8).580
medullary thyroid malignancies are more common than in the general
population and range in prevalence from 3% to 7%.478,479,482-486
Two large series of CS patients have demonstrated breast involvement
in approximately 70%. Fifty percent to 60% have benign lesions, and
breast cancer is found in approximately 20% to 30% of cases.478,479
Owing to the lack of prospective, long-term follow-up studies, the
lifetime risk of breast cancer in CS patients may be underestimated.
302
Curr Probl Surg, May 2005
FIG 30. Keratodermas of the palm in a patient with Cowden syndrome (reproduced with permission
from Hover AR, Cawthern T, McDanial W. Cowden disease. A hereditary polyposis syndrome
diagnosable by mucocutaneous inspection. J Clin Gastroenterol 1986;8:576-9).
Approximately 30% of patients with breast cancer have bilateral involvement, and the mean age at presentation is younger than for the general
population, ranging from 38 to 46 years.478,487 Breast cancer is not
limited to women with CS; 2 cases of breast cancer have been reported in
men with CS.488
The GI tract manifestations in CS are adenomas, hamartomas, ganglioneuromas, leiomyomas, lipomas, lymphoid, neuromas, as well as hyperplastic, inflammatory, and occasionally juvenile polyps.470,485,489-495 The
incidence of GI involvement may range from 35% to 85%, depending
on whether patients have contrast studies and endoscopy performed.478,495,496 In contrast to JP, there are only 4 reported cases of
colon carcinoma.478,482,497,498
Menstrual irregularities and ovarian cysts are found in approximately
20% of patients with CS, and leiomyomas of the uterus may also be
present.478,491,496 Other cancers associated with CS are uterine adenocarcinoma (6%), transitional cell carcinoma (5%), and cervical carcinoma
(3%).478 Eng499 recently suggested a possible association between CS
and both renal cell carcinoma as well as melanoma, based on anecdotal
evidence.
Curr Probl Surg, May 2005
303
Lhermitte-Duclos Disease. Lhermitte and Duclos500 first described this
syndrome in the French literature in 1920. This was later referred to as
dysplastic gangliocytoma of the cerebellum, due to the histologic features
of this peculiar hamartoma. This cerebellar hamartoma usually manifests
with symptoms of increased pressure or cerebellar ataxia, which include
headaches (75%), papilledema (70%), cerebellar syndrome (50%), megalencephaly (40%), cranial nerve deficit (30%), ataxia (20%), and pyramidal syndrome (20%). Associated anomalies found in patients with
Lhermitte-Duclos disease (LDD) have also included macrocephaly,
megalencephaly, hydrocephalus, heterotopia, and hydromyelia.471,472
Ambler and colleagues501 found LDD to be autosomal dominant in
nature. By the 1980s, the diagnosis could be established by MRI findings,
manifesting as a nonenhancing gyriform pattern with enlargement of the
cerebellar folia.502 In 1921, Padberg and colleagues473 reported 2 patients
with biopsy-proven LDD and CS. This was followed by similar reports,
leading to the realization that these were the same hamartomatous
syndrome involving cutaneous and neurologic ectoderm.474-477,503-506
This was supported in 1996, when Nelen and colleagues507 reported the
localization of the predisposing gene for CS to chromosome 10q22-23.
Among the12 families studied for linkage, 4 families had clear evidence
of LDD that showed the same linkage to 10q22-33.507 Hence, LDD was
incorporated into the diagnostic criteria for CS (Table 10).
Genetics. CS was demonstrated to have an autosomal dominant
inheritance by Gentry and colleagues494 in 1974 and Nuss and colleagues508 in 1978. Although within families there were generally equal
numbers of affected men and women, between families, interfamilial
series may show a female predisposition ranging from 1:1 to 1.6:
1.478,496,509 This could also reflect a bias for presentation in females due
to the breast disease predisposition in CS.
Excellent progress has been made in understanding the genetic basis of
CS in the last decade. In 1996, Nelen and colleagues507 found linkage for
CS to markers on chromosome 10q22-23, with a lod score of 8.92 (theta
⫽ 0.02) in 12 families. This was closely followed by independent
identification of the PTEN gene on chromosome 10 by 3 groups: Li and
Sun,510 Li and colleagues,511 and Steck and colleagues.512 Li and
colleagues511 found PTEN to have a sequence consistent with a protein
tyrosine phosphatase domain, and homology to the chicken protein tensin
and bovine protein auxilin. In the name PTEN, the phosphatase function
is represented by P; TEN refers to the homology to tensin and its location
on chromosome 10. They also found mutations of the PTEN gene in
breast cancer, glioblastoma, and prostate cancer cell lines.511 Liaw and
304
Curr Probl Surg, May 2005
colleagues513 found that 4 of 5 CS families had PTEN germline mutations
in 1997 and Nelen and colleagues514 found that 9 of 19 CS cases had
PTEN mutations.
PTEN is a dual specificity phosphatase that causes dephosphorylation of
serine/threonine and tyrosine residues and of protein and lipid substrates.515 PTEN is involved in embryonic development, the cell cycle,
apoptosis, and tumor formation. It plays an important role in the
phosphatidylinositol 3-kinase (PI3K) protein kinase B (PKB)/antiapoptotic serine threonine kinase (AKT) pathway. Upregulation of PI3K-PKB/
AKT promotes cell growth and survival, which is regulated by PTENmediated dephosphorylation of the phosphatidylinositol3-5 triphosphate
(PIP3) substrate. Reduced PIP3 levels leads to lower PKB/AKT in the cell
membrane, causing cell cycle arrest in G1 and apoptosis.516 Other
pathways regulated by PTEN may include the mitogen-activated kinase
(MAPK) pathway, which is involved in cell proliferation and differentiation and the insulin signaling pathway.517 Li and Sun,510 Li and
colleagues,511 and Steck and colleagues512 all recognized that the
mutation of PTEN leads to inactivation of its phosphatase activity and
inactivation of its role in growth inhibition, implying a tumor suppressor
role.
Marsh and colleagues518 examined a series of 37 families with CS and
identified 30 families with germline PTEN mutations, for an incidence of
81%. Zhou and colleagues519 re-examined 97 patients with CS that were
negative for PTEN mutations. They found mutations in the promoter
region of PTEN in 9 (9.3%) patients. Combining both mutations in the
PTEN promoter and coding sequence therefore accounts for approximately 90% of germline mutations seen in patients with CS.
Treatment. The first step in appropriate screening for CS is recognition
of its predisposition to malignancy. Once CS is diagnosed, patients should
be considered for genetic counseling. This provides an opportunity for
support and education, including its social, familial, and personal ramifications. In a family in which a PTEN mutation has been detected, if the
individuals at risk are negative for the same mutation, they may then
undergo routine health and cancer screening. At-risk CS patients or
patients in families without identified PTEN mutations should undergo a
general comprehensive physical examination starting at 18 years of age or
5 years before the earliest cancer was diagnosed in another family
member.
The most common cancers encountered in CS patients are of the breast
and thyroid gland. Starink and colleagues478 reported 3 patients younger
than the age of 30 years in whom breast cancer developed. Therefore,
Curr Probl Surg, May 2005
305
at-risk patients should perform monthly breast self-examinations starting
at age 18, with annual physical examinations beginning at age 25, or 5
to 10 years earlier than the youngest case of breast cancer in the
family.499,520 Mammography should be performed beginning at age 30
to 35 years, or at least 5 years before the youngest age of onset of
breast cancer in the family.499,521
Thyroid cancer in CS patients may develop by 10 to 30 years of
age.470,485,486,522 Screening should begin with a thyroid ultrasound at age
18, with annual physical examinations thereafter and repeat ultrasounds
as determined by the level of clinical suspicion.
To screen for endometrial cancer, annual endometrial suction biopsies
should be performed beginning at the age of 35 to 40 years, or 5 years
younger than the earliest premenopausal case of endometrial cancer in the
family. Postmenopausal women should have an annual endometrial
ultrasound. Families with a history of renal cancer should have annual
urine cytology, and renal ultrasound as indicated. Dermatologic examination is also suggested annually to screen for melanoma.499
Bannayan-Riley-Ruvalcaba Syndrome
The name Bannayan-Riley-Ruvalcaba syndrome (BRRS) was proposed
in 1990 to encompass 3 phenotypically similar syndromes (RuvalcabaMyhre-Smith syndrome, Bannayan-Zonana syndrome, and the RileySmith syndrome). The syndromes are defined by macrocephaly, and
variable penetrance of developmental delay, hemangiomas, lipomas,
genital pigmentation, intestinal polyps, and lipid myopathy.523-536 The
predominant features of BRRS as reported by Gorlin and colleagues are
listed in Table 11.
The GI polyps associated with BRRS were initially described as
hamartomatous.523,527,529,537 Further review by Haggitt and Reid538 of
the histologic features of polyps reported by Ruvalcaba found that they
were juvenile polyps, except for 1 ganglioneuroma. Recent reports have
confirmed that most polyps are juvenile polyps, which occasionally will
display adenomatous changes.539-541
Genetics. It may be difficult to distinguish between BRRS and CS.
Fargnoli and colleagues542 described a patient with findings consistent
with both BRRS and meeting the diagnostic criteria for CS. This patient
had macrocephaly, 2 lipoma resections, a thyroid follicular adenoma (that
had been removed), scoliosis, colonic polyps, and hyperpigmented
macules on the glans penis (Fig 31).542 Both BRRS and CS have an
autosomal dominant inheritance.527,531,532,536,543 Marsh and colleagues518 found a PTEN germline mutation in 4 of 7 unrelated BRRS
306
Curr Probl Surg, May 2005
TABLE 11. Bannayan-Riley-Ruvalcaba syndrome, common signs*
Craniofacial
Macrocephaly
Prominent corneal nerves/Schwalbe lines
Neurologic
Developmental delays (motor, mental, speech)
Muscular
Lipid myopathy
Cutaneous
Lipomas
Genitalia pigmentation
Angiolipomas
Gastrointestinal
Intestinal polyps (hamartomatous, juvenile)
Skeletal
Accelerated growth of 1st metacarpal, and 1st proximal and 2nd middle phalanges
Joint hyperflexibility, pectus excavatum, and scoliosis
*Reproduced with permission from Gorlin RJ, Cohen MM Jr, Codon LM, Burke BA. BannayanRiley-Ruvalcaba syndrome. Am J Med Genet 1992;44:307–14.
families in 1998. One of the these PTEN mutations had previously been
found in 2 CS families,513 suggesting that the 2 syndromes were allelic.
The largest series to date has been that of Marsh and colleagues,544 who
evaluated 43 individuals with BRRS. Of these 43 patients with BRRS, 16
were sporadic and 27 were familial.544 Eleven of the 27 familial cases
were mixed, having individuals of both CS and BRRS phenotypes. PTEN
mutations were found in 26 of 43 individuals, for a prevalence of 60%.544
Additional reports of PTEN mutations associated with patients having
BRRS have been published.545,546
In families with BRRS, the recommendations for follow-up are less
clear, because the risk for cancers has not been established. The findings
from genetic studies that BRRS and CS are allelic syndromes suggests
that the clinician maintain an elevated level of suspicion and examine the
patient annually for the same tumors seen in patients with CS.
Peutz-Jeghers Syndrome
Peutz-Jeghers syndrome (PJS) is characterized by hamartomatous
polyps of the upper and lower GI tract, and lentigines (pigmented lesions)
of the buccal mucosa, perioral region, palms, feet, or anus (Fig 32). The
association of melanosis and polyposis was first recognized by Peutz547 in
1921, and Jeghers and colleagues548 described an autosomal dominant
inheritance in 1949. The prevalence of PJS has been estimated to be
between 1 in 8300 and 1 in 29,000.549 The mean age of diagnosis of PJS
ranges between 22 and 26 years.174,550
Curr Probl Surg, May 2005
307
FIG 31. Macules of the glans penis seen in a patient with Bannayan-Riley-Ruvalcaba syndrome
(reproduced with permission from Gorlin RJ, Cohen MM Jr, Condon LM, Burke BA. Bannayan-RileyRuvalcaba syndrome. Am J Med Genet 1992;44:307-14).
The intestinal polyps are most commonly found in the jejunum, and
large series have found the incidence of polyps in the small intestine to be
78%, colon 42%, stomach 38%, and rectum 28%.174,550,551 These polyps
resemble the hamartomatous polyps in patients with juvenile polyposis
and Cowden syndrome, but differ in the fact that there is prominent
smooth muscle tissue found within the lamina propria (Fig 33). Adenomatous and hyperplastic polyps may also be encountered. Gastric polyps
tend to be more sessile, whereas small and large intestinal polyps are
generally pedunculated. The most common problems arising from these
GI polyps are intussusception and GI bleeding, as highlighted by the
report of 1 family with 12 affected members that had 32 abdominal
operations and more than 70 endoscopic procedures performed for
removal of polyps.552
The skin lesions in PJS patients are pigmented macules, caused by
increased numbers of melanocytes. These lesions are not premalignant.
They are most commonly seen around the lips, mouth, hands, feet, buccal
mucosa, eyes, nose, and perianal areas.174,548 The degree of pigmentation
in cutaneous regions may decrease with age, but it may persist in the
buccal mucosa.
308
Curr Probl Surg, May 2005
FIG 32. Pigmented lesions of the oral mucosa in a patient with Peutz-Jeghers syndrome (reproduced
with permission from Aaltonen LA, Järvinen H, Gruber SB. Peutz-Jeghers syndrome. In: Hamilton SR,
Aaltonen LA, editors. WHO Classification: Tumours of the Digestive System. Lyon: International
Agency for Research on Cancer; 2000:74-6).581
In a study of 72 PJS patients, Spigelman and colleagues553 found that
16 (22%) had developed cancer, with 10 GI cancers (4 small bowel, 3
stomach, 2 colon, 1 pancreas) and 7 non-GI cancers (2 unknown primary,
1 ovary, 1 lung, 1 thyroid, 1 fallopian tube, 1 basal cell carcinoma). The
relative risk of GI cancer death was 13, and 48% of those with cancer had
died by age 57.553 Giardiello and colleagues554 found that 15 of 31 (48%)
PJS patients developed cancers over a 12-year follow-up interval. Four
had GI cancers (2 stomach, 2 colon), 4 had pancreatic cancers, and 7 had
other cancers (2 breast, 2 lung, 1 ovary, 1 uterus, 1 myeloma). The
relative risk of cancer was calculated to be 18 times higher than the
normal population, and the average age at diagnosis was 25 years
(ranging from 1 to 64 years) after the diagnosis of PJS (at mean age of 17
years).554 Lim and colleagues555 found that 8 of 70 affected PJS patients
developed cancers (2 stomach, 2 breast, 2 colorectal, 1 pancreas, 1
cervical). They calculated the risk of developing cancer by age 65 in
mutation carriers to be 47%, with a standardized mortality ratio of 13.2
for all cancers, 32.0 for GI cancer, and 13.9 for breast cancer. Boardman
and colleagues556 found that 18 of 34 PJS patients developed cancer, in
Curr Probl Surg, May 2005
309
which there were 10 GI (7 colon, 1 duodenum, 1 esophagus, 1 stomach)
and 16 extraintestinal cancers (6 breast, 3 lung, 2 cervix, 1 ovary, 1 uterus,
1 thyroid, 1 prostate, 1 unknown primary). Six patients had multiple
cancers. Women were at an 18.5-fold increased risk for cancer, 20.3 times
higher for breast and gynecologic malignancies. Men were at a 6.2-fold
increased risk for cancer, and the mean age of cancer diagnosis was 39.4
years in all patients.556 In a review of PJS, Hemminki557 found 39 cases
of small bowel, 31 colorectal, 17 gastric, 8 pancreatic, and 1 esophageal
cancer. There were 22 cases of breast cancer, 13 cervical, 9 lung, 8
ovarian, 3 hepatobiliary, 2 thyroid, 2 myelomas, 2 leiomyosarcomas, 2
endometrial, 1 testicular, 1 fallopian tube cancer, and 1 osteosarcoma.
Benign tumors of the breast, ovary, thyroid, and of sex cord origin have
also been reported.557 Giardiello and colleagues558 performed a metaanalysis and showed that the relative risk of small bowel cancer in PJS
was 520, stomach 213, pancreas 132, colon 84, esophagus 57, ovary 27,
lung 17, breast 15, and uterus 16. The risk of cervical and testicular cancer
was not increased.558 The mechanism of GI cancer development is likely
through hamartoma to adenoma to carcinoma, although this has not been
completely elucidated. Spigelman and colleagues553 did find that 4 to 5 of
their 10 GI cancers appeared to have developed within a hamartomatous
polyp.
Genetics of PJS. In 1997, evidence for a PJS locus on chromosome 19p
was discovered by Hemminki and colleagues,559 who performed comparative genomic hybridization and detected a region that appeared to be
lost in PJS tumors relative to normal tissues. Loss of heterozygosity
studies found consistent losses of markers from 19p in tumors. Then by
genetic linkage, they found a multipoint lod score of 7.00 at theta ⫽ 0
with the marker D19S886 in 12 PJS families.559 Amos and colleagues560
performed genetic linkage studies in 5 PJS families and confirmed linkage
to the same marker (multipoint lod score of 7.52 at theta ⫽ 0), placing the
PJS susceptibility gene within the telomeric 3 megabases of chromosome
19p. Mehenni and colleagues561 also found linkage to D19S886 with a
2-point lod score of 4.74 at theta ⫽ 0.045. Interestingly, they found
FIG 33. A) Gross picture of a small intestinal polyp in a patient with Peutz-Jeghers syndrome (PJS). B)
Photomicrograph of a colonic PJS polyp, similar to the hamartomatous juvenile polyposis polyp shown
in Fig 23, but the lamina propria has large amounts of smooth muscle present (reproduced with
permission from Aaltonen LA, Järvinen H, Gruber SB. Peutz-Jeghers syndrome. In: Hamilton SR,
Aaltonen LA, editors. WHO Classification: Tumours of the Digestive System. Lyon: International
Agency for Research on Cancer; 2000:74-6).581
310
Curr Probl Surg, May 2005
Curr Probl Surg, May 2005
311
linkage to the marker D19S891 from the long arm of chromosome 19
(19q) in 1 Indian PJS family (lod of 3.52 at theta ⫽ 0), suggesting a
possible second PJS locus.
Hemminki and colleagues562 mapped cDNA clones to a cosmid contig
created from the human chromosome 19 mapping project, spanning an
800-kb region near D19S886 known to contain the PJS gene by critical
recombinants in PJS families. Twenty-seven genes were mapped to this
region, and mutations were found in 1 called LKB1, a previously
unmapped serine threonine protein kinase gene. Of 12 unrelated PJS
patients sequenced, 11 were found to have LKB1 mutations (5 deletions,
1 insertion, 5 substitutions; Fig 34). This gene was expressed in all tissues
examined, and the mutations found were predicted to diminish the kinase
activity of the protein, in contrast to the activation of kinases seen in other
heritable cancer syndromes (such as RET in MEN2, and CDKN2 in
familial melanoma).562 A similar effort from Jenne and colleagues563
mapped 21 genes from overlapping cosmid clones spanning 400 kb in the
region of D18S886, and 1 of these genes was found to have a rearrangement in a PJS patient. This was a serine threonine protein kinase gene
consisting of 9 exons over 23 kb of genomic DNA, which they called
STK11 (although it had previously been given the name LKB1). The PJS
proband had a deletion of exons 4 and 5, and inversion of exons 6 and 7,
and all 3 affected members of her family also had this rearrangement.
Analysis of 4 other unrelated PJS patients revealed 3 nonsense and 1
splice site mutation of the gene.563
Gruber and colleagues564 found an LKB1 mutation in the original
family reported by Jeghers,548 as well as 5 others (2 deletions, 2
insertions, 2 nonsense mutations). They confirmed that LKB1 was a tumor
suppressor gene by demonstrating that 11 of 12 hamartomas or adenocarcinomas from 4 PJS patients had loss of the wild-type allele with
retention of the mutated allele. The losses seen in hamartomas implied
that this gene is involved at an early stage of tumor progression.564 Jiang
and colleagues565 found only 1 mutation of LKB1 in 10 unrelated PJS
patients, and suggested that there may be other genes responsible for JPS.
Nakagawa and colleagues566 analyzed 15 unrelated PJS patients for
mutations, and found them in 10. Four were deletions causing frameshifts,
3 were insertions, 2 were splice site mutations, and 1 was a nonsense
mutation.
Miyaki and colleagues567 found LKB1 mutations in 6 of 9 PJS families
studied, which consisted of 3 deletions, 2 insertions, and 1 splice site
mutation, all predicted to cause protein truncation. They also found
somatic LKB1 mutations in 5 of 27 (19%) polyps, 4 of which shared a
312
Curr Probl Surg, May 2005
4-bp deletion in exon 6. Fourteen other polyps had LOH of 19p markers,
for a total of 70% of polyps showing potential somatic inactivation of
LKB1. Six larger polyps also had ␤-catenin mutations, suggesting these
changes may be important in polyp progression. One carcinomatous
polyp examined had 19p LOH, ␤-catenin mutation, and p53 mutation,
suggesting a hamartoma-adenoma-carcinoma sequence in PJS polyps.567
Gruber and colleagues564 found that 11 of 12 hamartomas and adenocarcinomas had 19p LOH, 1 of 18 had 18q LOH, and 0 of 18 had K-ras
mutations.
Wang and colleagues568 found LKB1 germline mutations in 7 of 12
(58%) PJS patients, which included 4 deletions, 2 nonsense mutations,
and 1 missense mutation. Four families without mutations had findings
compatible with LKB1 linkage, suggesting that alterations might be
present but were not detected by their sequencing or single strand
conformational polymorphism (SSCP) analysis.568 Westerman and colleagues569 found LKB1 mutations in 12 of 19 PJS families (63%), which
consisted of 3 deletions, 3 insertions, 3 missense mutations, 2 splice site
mutations, and 1 nonsense mutation.
Boardman and colleagues570 screened 10 familial and 23 sporadic cases
of PJS for LKB1 changes by conformation-sensitive gel electrophoresis
(CSGE), and only found mutations in 2 and 4 cases, respectively. The low
mutation rate found in this study may have been due to the lower
sensitivity rate of CSGE for identifying base-pair changes (which is
approximately 90%), other mechanisms of LKB1 alteration, or the
possibility that other genetic loci for JPS may exist (genetic heterogeneity).570 Lim and colleagues555 also used CSGE to screen 33 JPS families
for LKB1 mutations, and found mutations in 17 (52%; 5 deletions, 3
splice site mutations, 2 insertions, 3 nonsense, and 4 missense mutations).
Boudeau and colleagues571 studied the functional consequences of 6
germline LKB1 mutations (4 deletions and 2 missense mutations) in PJS
patients by cloning the cDNA into a CMV expression vector. They found
that each of these LKB1 isoforms was unable to autophosphorylate at
Thr336, and to suppress the growth of a melanoma cell line, in contrast
to wild-type LKB1. They concluded that these were loss-of-function
mutations, and that loss of kinase activity was responsible for PJS.571
Esteller and colleagues572 studied 51 cancer cell lines (15 colon, 11
lung, 7 ovary, 5 breast, 3 thyroid, 3 brain, 3 prostate, 3 leukemia, 1 cervix)
for methylation of a CpG island just upstream from LKB1, and found this
in 3 colorectal and 1 cervical cancer cell lines with coincident loss of
LKB1 expression. They also found methylation in 4 of 22 (18%)
Curr Probl Surg, May 2005
313
314
Curr Probl Surg, May 2005
hamartomatous polyps in PJS patients, suggesting that this mechanism of
gene inactivation may be involved in a subset of PJS cases.572
Entius and colleagues573 examined 39 polyps and 5 carcinomas from 17
PJS patients for LOH for markers at the LKB1 locus on 19p13, 5q21
(APC), 17p13 (p53), and 18q21 to 22 (MADH2, MADH4). Fifteen of 39
(38%) polyps had 19p13 LOH, as did 5 of 5 carcinomas. Six of 7 polyps
from the 5 patients with carcinoma demonstrated 19p LOH. No LOH was
seen at 5q21, but 1 cancer had an APC mutation. LOH was not found at
17p13 in any of these samples, but was found in 1 cancer at 18q; however,
all polyps and cancers had p53 and MADH4 expression by immunohistochemistry. The authors concluded that LKB1 loss was important in
tumorigenesis in PJS patients, and the genetic mechanisms responsible
differed from those in sporadic colorectal carcinogenesis.573
Surveillance and Follow-Up. Recommendations for surveillance of
patients with PJS or at risk for PJS are not clear, but must address the
significantly elevated risk of a variety of cancers, such as the aforementioned increased relative risk of small bowel cancer (520-fold increased
risk), stomach,213 pancreas,132 colon,84 esophagus,57 ovary,27 lung,17
uterus,16 and breast.15,558 Dunlop574 recommended that colonoscopy or
barium enema be performed every 3 years beginning at age 18, and that
upper endoscopy be performed every 3 years beginning at age 25.
Hemminki557 recommends biannual upper and lower endoscopy beginning in the mid teen years, and others suggest these be performed every
2 to 5 years.575 How to follow the small bowel in PJS patients remains a
challenge. These polyps can lead to intussusception or anemia and
develop into cancers. Small bowel follow-through and enteroclysis will
show larger polyps, but the relatively new technique of capsule endoscopy may be the best available method for visualization of small bowel
polyps. It would seem prudent to perform capsule endoscopy at least
every 3 years in asymptomatic PJS gene carriers, which might preclude
the need for screening colonoscopy and upper endoscopy, which could be
reserved for therapeutic interventions. If multiple polyps are found, then
screening should be performed at least annually until the polyps are
removed.
Gastric and colonic polyps may be removed endoscopically, with
FIG 34. A) Location of LKB1 on chromosome 19p. B) Genomic and protein structure and mutations
described in LKB1 (reproduced with permission from Hemminki A, Markie D, Tomlinson I, Avizienyte
E, Roth S, Loukola A, et al. A serine/threonine kinase gene defective in Peutz-Jeghers syndrome.
Nature 1998;391:184-7).
Curr Probl Surg, May 2005
315
surgery being reserved for lesions with dysplasia or cancer. Less invasive
therapeutic interventions for small bowel polyps are more limited, since
most enteroscopes will only reach the proximal jejunum. Surgery is
therefore the required treatment for symptomatic polyps (causing intussusception, anemia, or pain) or those suspicious for cancer. At operation,
intraoperative endoscopy through an enterotomy can be performed to
examine the entire small bowel, and polyps may be removed through the
endoscope by using a snare cautery. Larger or suspicious polyps will
require enterotomy or small bowel resection for their removal.
Surveillance recommendations for extraintestinal cancers in PJS patients are not well established. Screening for breast cancers should include
annual breast examination and mammography (or ultrasound in patients
younger than age 35), with the latter beginning between the ages of 25
and 35. Gynecologic malignancy should be ruled out by annual transvaginal ultrasound, pap smear, and pelvic examination. Annual testicular
examination and ultrasound should be initiated in males by age 10.
Screening for pancreatic cancer by abdominal ultrasound, endoscopic
ultrasound, or computed tomography should be performed every 1 to 2
years starting at age 30.552,557,575
The diagnosis of PJS gene carriers may be evident from within the first
year of life based on the characteristic mucocutaneous pigmentation.
These patients will clearly need surveillance. Gene testing for PJS
involves direct sequencing of the entire gene for probands and directed
mutation screening of other at-risk individuals once a mutation is found
in the family. Genetic testing of at-risk individuals from families with
known LKB1 mutations will benefit those found not to harbor the
disease-causing mutation because they may be excused from the screening regimens. Those found to be gene carriers will need to be educated
regarding their risk for developing cancer and be given these surveillance
recommendations.
References
303. Lynch HT, de la Chapelle A. Genetic susceptibility to non-polyposis colorectal
cancer. J Med Genet 1999;36:801-18.
304. Lynch HT, de la Chapelle A. Hereditary colorectal cancer. N Engl J Med
2003;348:919-32.
305. Park YJ, Shin K-H, Park J-G. Risk of gastric cancer in hereditary nonpolyposis
colorectal cancer in Korea. Clin Cancer Res 2000;6:2994-8.
306. Watson P, Lynch HT. Extracolonic cancer in hereditary nonpolyposis colorectal
cancer. Cancer 1993;71:677-85.
307. Watson P, Lynch HT. The tumor spectrum in HNPCC. Anticancer Res
1994;14:1635-40.
316
Curr Probl Surg, May 2005
308. Thorson A, Knezetic JA, Lynch HT. A century of progress in hereditary
nonpolyposis colorectal cancer (Lynch syndrome). Dis Colon Rectum 1999;42:1-9.
309. Warthin AS. Heredity with reference to carcinoma as shown by the study of the
cases examined in the pathological laboratory of the University of Michigan,
1895-1913. Arch Intern Med 1913;12:546-55.
310. Warthin AS. The further study of a cancer family. J Cancer Res 1925;9:279-86.
311. Lynch HT, Shaw MW, Magnuson CW, Larsen AL, Krush AJ. Hereditary factors in
cancer: study of two large Midwestern kindreds. Arch Intern Med
1966;117:206-12.
312. Lynch HT, Krush AJ. Cancer family “G” revisited: 1895-1970. Cancer
1971;27:1505-11.
313. Lynch HT, Smyrk T, Lynch JF. Molecular genetics and clinical-pathology features
of hereditary nonpolyposis colorectal carcinoma (Lynch syndrome): historical
journey from pedigree anecdote to molecular genetic confirmation. Oncology
1998;55:103-8.
314. Yan H, Papadopoulos N, Marra G, Perrera C, Jiricny J, Boland CR, et al.
Conversion of diploidy to haploidy. Nature 2000;403:723-4.
315. Lynch HT, Coronel SM, Okimoto R, Hampel H, Sweet K, Lynch JF, et al. An
American founder mutation of the MSH2 gene may account for a significant
proportion of all Lynch syndrome cases in the United States. JAMA
2004;291:718-24.
316. Vasen HF, Mecklin JP, Khan P, Lynch HT. The International Collaborative Group
on Hereditary Nonpolyposis Colorectal Cancer (ICG-HNPCC). Dis Colon Rectum
1991;34:424-5.
317. Vasen HFA, Watson P, Mecklin J-P, Lynch HT. ICG-HNPCC. New clinical criteria
for hereditary nonpolyposis colorectal cancer (HNPCC, Lynch syndrome) proposed
by the International Collaborative Group on HNPCC. Gastroenterology
1999;116:1453-6.
318. Rodriguez-Bigas MA, Boland CR, Hamilton SR, Henson DE, Jass JR, Khan PM,
et al. A National Cancer Institute Workshop on Hereditary Nonpolyposis Colorectal
Cancer Syndrome: meeting highlights and Bethesda guidelines. J Natl Cancer Inst
1997;89:1758-62.
319. David KL, Steiner-Grossman P. The potential use of tumor registry data in the
recognition and prevention of hereditary and familial cancer. NY State J Med
1991;91:150-2.
320. Lynch HT, Follett KL, Lynch PM, Albano WA, Mailliard JL, Pierson RL. Family
history in an oncology clinic: implications for cancer genetics. JAMA
1979;242:1268-72.
321. Thibodeau SN, Bren G, Schaid D. Microsatellite instability in cancer of the
proximal colon. Science 1993;260:816-9.
322. Aaltonen LA, Peltomaki P, Leach FS, Sistonen P, Pylkkanen L, Mecklin JP, et al.
Clues to the pathogenesis of familial colorectal cancer. Science 1993;260:812-6.
323. Aaltonen LA, Peltomaki P, Mecklin JP, Järvinen H, Jass JR, Green JS, et al.
Replication errors in benign and malignant tumors from hereditary nonpolyposis
colorectal cancer patients. Cancer Res 1994;54:1645-8.
324. Strand M, Prolia TA, Liskay RM, Petes TD. Destabilization of tracts of simple
repetitive DNA in yeast affecting DNA mismatch repair. Nature 1993;365:274-6.
Curr Probl Surg, May 2005
317
325. Modrich P. Mechanisms and biological effects of mismatch repair. Annu Rev
Genet 1991;25:229-53.
326. Peltomäki P, Aaltonen LA, Sistonen P, Pylkkanen L, Mecklin JP, Järvinen H, et al.
Genetic mapping of a locus predisposing to human colorectal cancer. Science
1993;260:810-2.
327. Lindblom A, Tannergard P, Werelius B, Nordenskjold M. Genetic mapping of a
second locus predisposing to hereditary non-polyposis colon cancer. Nat Genet
1993;5:279-82.
328. Fischel R, Lescoe MK, Rao MRS, Copeland NG, Jenkins NA, Garber J, et al. The
human mutator gene homolog MSH2 and its association with hereditary nonpolposis colon cancer. Cell 1993;75:1027-38.
329. Leach FS, Nicolaides NC, Papadopoulus N, Liu B, Jen J, Parsons R, et al.
Mutations of a mutS homolog in hereditary nonpolyposis colorectal cancer. Cell
1993;75:1215-25.
330. Nystrom-Lahti M, Parsons R, Sistonen P, Pylkkanen L, Aaltonen LA, Leach FS, et
al. Mismatch repair genes on chromosome 2p and 3p account for a major share of
hereditary nonpolyposis colorectal cancer families evaluable by linkage. Am J Hum
Genet 1994;55:659-65.
331. Liu B, Parsons RE, Hamilton SR, Petersen GM, Lynch HT, Watson P, et al.
hMSH2 mutations in hereditary nonpolyposis colorectal cancer kindreds. Cancer
Res 1994;54:4590-4.
332. Wijnen J, Vasen H, Khan PM, Menko FH, van der Klift H, van Leeuwen C, et al.
Seven new mutations in hMSH2, an HNPCC gene, identified by denaturing
gradient-gel electrophoresis. Am J Hum Genet 1995;56:1060-6.
333. Maliaka YK, Chudina AP, Belev NF, Alday P, Bochkov NP, Buerstedde J-M. CpG
dinucleotides in the hMSH2 and hMLH1 genes are hotspots for HNPCC mutations.
Hum Genet 1996;97:251-5.
334. Papadopoulos N, Nicolaides NC, Wei YF, Ruben SM, Carter KC, Rosen CA, et al.
Mutation of a mutL homolog in hereditary colon cancer. Science 1994;263:1625-9.
335. Bronner CE, Baker SM, Morrison PT, Warren G, Smith LG, Lescoe MK, et al.
Mutation in the DNA mismatch repair gene homologue hMLH1 is associated with
hereditary non-polyposis colon cancer. Nature 1994;368:258-61.
336. Han H-J, Maruyama M, Baba S, Park J-G, Nakamura Y. Genomic structure of
human mismatch repair gene, hMLH1, and its mutation analysis in patients with
hereditary nonpolyposis colorectal cancer (HNPCC). Hum Mol Genet
1995;4:237-42.
337. Kolodner RD, Hall NR, Lipford J, Kane MF, Morrison PT, Finan PJ, et al. Structure
of the human MLH1 locus and analysis of a large hereditary nonpolyposis
colorectal carcinoma kindred for mlh1 mutations. Cancer Res 1995;55:242-8.
338. Wijnen J, Khan PM, Vasen H, Menko FH, van der Klift H, van der Broek M, et al.
Majority of hMLH1 mutations responsible for hereditary nonpolyposis colorectal
cancer cluster at the exonic region 15-16. Am J Hum Genet 1996;58:300-7.
339. Parsons R, Li GM, Longley MJ, Fang WH, Papadopoulos N, Jen J, et al.
Hypermutability and mismatch repair deficiency in RER⫹ tumor cells. Cell
1993;75:1227-36.
340. Hemminki A, Peltomaki P, Mecklin JP, Järvinen H, Salovaara R, Nystrom-Lahti
M, et al. Loss of the wild type MLH1 gene is a feature of hereditary nonpolyposis
colorectal cancer. Nat Genet 1994;8:405-10.
318
Curr Probl Surg, May 2005
341. Herman JG, Umar A, Polyak K, Graff JR, Ahuja N, Issa JP, et al. Incidence and
functional consequences of hMLH1 promoter hypermethylation in colorectal
carcinoma. Proc Natl Acad Sci USA 1998;95:6870-5.
342. Simpkins SB, Bocker T, Swisher EM, Mutch DG, Gersell DJ, Kovatich AJ, et al.
MLH1 promoter methylation and gene silencing is the primary cause of microsatellite instability in sporadic endometrial cancers. Hum Mol Genet 1999;8:661-6.
343. Nicolaides NC, Papadopoulos N, Liu B, Wei YF, Carter KC, Ruben SM, et al.
Mutations of two PMS homologues in hereditary nonpolyposis colon cancer.
Nature 1994;371:75-80.
344. Drummond JT, Li GM, Longley MJ, Modrich P. Isolation of an hMSH2-p160
heterodimer that restores DNA mismatch repair to tumor cells. Science
1995;268:1909-12.
345. Palombo F, Gallinari P, Iaccarino I, Lettieri T, Hughes M, D’Arrigo A, et al. GTBP,
a 160-kilodalton protein essential for mismatch-binding activity in human cells.
Science 1995;268:1912-4.
346. Papadopoulos N, Nicolaides NC, Liu B, Parsons R, Lengauer C, Palombo F, et al.
Mutations of GTBP in genetically unstable cells. Science 1995;268:1915-7.
347. Miyaki M, Konishi M, Tanaka K, Kikuchi-Yanoshita R, Muraoka M, Yasuno M,
et al. Germline mutation of MSH6 as the cause of hereditary nonpolyposis
colorectal cancer. Nat Genet 1997;17:271-2.
348. Wijnen J, de Leeuw W, Vasen H, van der Klift H, Moller P, Stormorken A, et al.
Familial endometrial cancer in female carriers of MSH6 germline mutations. Nat
Genet 1999;23:142-4.
349. Wu Y, Berends MJ, Mensink RG, Kempinga C, Sijmons RH, van Der Zee AG, et
al. Association of hereditary nonpolyposis colorectal cancer-related tumors displaying low microsatellite instability with MSH6 germline mutations. Am J Hum Genet
1999;65:1291-8.
350. Berends MJ, Wu Y, Sijmons RH, Mensink RG, van der Sluis T, Hordijk-Hos JM,
et al. Molecular and clinical characteristics of MSH6 variants: an analysis of 25
index carriers of a germline variant. Am J Hum Genet 2002;70:26-37.
351. Huang J, Kuismanen SA, Liu T, Chadwick RB, Johnson CK, Stevens MW, et al.
MSH6 and MSH3 are rarely involved in genetic predisposition to nonpolypotic
colon cancer. Cancer Res 2001;61:1619-23.
352. Berends MJW, Wu Y, Sijmons RH, van der Sluis T, Boersmavan Ek W, Ligtenberg
MJL, et al. Toward new strategies to select young endometrial cancer patients for
mismatch repair gene mutation analysis. J Clin Oncol 2003;21:4364-70.
353. Wagner A, Barrows A, Wijnen JT, van der Klift H, Franken PF, Verkuijlen P, et
al. Molecular analysis of hereditary nonpolyposis colorectal cancer in the United
States: high mutation detection rate among clinically selected families and
characterization of an American founder genomic deletion of the MSH2 gene. Am J
Hum Genet 2003;72:1088-100.
354. Renkonen E, Lohi H, Zhang Y, Salovaara R, Abdel-Rahman W, Moisio A-L.
Expression changes in predisposition to HNPCC and FAP: a population based
study. Familial Cancer 2003;2(Suppl 1):37 (abstract).
355. Renkonen E, Zhang Y, Lohi H, Salovaara R, Abdel-Rahman WM, Nilbert M, et al.
Altered expression of MLH1, MSH2, and MSH6 in predisposition to hereditary
nonpolyposis colorectal cancer. J Clin Oncol 2003;21:3629-37.
356. De la Chapelle A, Wright FA. Linkage disequilibrium mapping in isolated
Curr Probl Surg, May 2005
319
357.
358.
359.
360.
361.
362.
363.
364.
365.
366.
367.
368.
369.
370.
371.
372.
320
populations: the example of Finland revisited. Proc Natl Acad Sci USA
1998;95:12416-23.
Nystrom-Lahti M, Sistonen P, Mecklin JP, Pylkkanen L, Aaltonen LA, Järvinen H,
et al. Close linkage to chromosome 3p and conservation of ancestral founding
haplotype in hereditary nonpolyposis colorectal cancer families. Proc Natl Acad Sci
USA 1994;91:6054-8.
Nystrom-Lahti M, Wu Y, Moisio AL, Hofstra RM, Osinga J, Mecklin JP, et al.
DNA mismatch repair gene mutations in 55 kindreds with verified or putative
hereditary non-polyposis colorectal cancer. Hum Mol Genet 1996;5:763-9.
Moisio AL, Sistonen P, Weissenbach J, de la Chapelle APP. Age and origin of two
common MLH1 mutations predisposing to hereditary colon cancer. Am J Hum
Genet 1996;59:1243-51.
Järvinen HJ, Aarnio M, Mustonen H, Aktan-Collan K, Aaltonen LA, Peltomaki P,
et al. Controlled 15-year trial on screening for colorectal cancer in families with
hereditary nonpolyposis colorectal cancer. Gastroenterology 2000;118:829-34.
Foulkes WD, Thiffault I, Gruber SB, Horwitz M, Hamel N, Lee C, et al. The
founder mutation MSH2*1906G–⬎C is an important cause of hereditary nonpolyposis colorectal cancer in the Ashkenazi Jewish population. Am J Hum Genet
2002;71:1395-412.
Guillem JG, Rapaport BS, Kirchhoff T, Kolachana P, Nafa K, Glogowski E, et al.
A636P is associated with early-onset colon cancer in Ashkenazi Jews. J Am Coll
Surg 2003;196:222-5.
Peltomäki P. Role of DNA mismatch repair defects in the pathogenesis of human
cancer. J Clin Oncol 2003;21:1174-9.
Jass JR. Clinical significance of early-onset “sporadic” colorectal cancer with
microsatellite instability. Dis Colon Rectum 2003;46:1305-9.
Jass JR, Smyrk TC, Stewart SM, Lane MR, Lanspa SJ, Lynch HT. Pathology of
hereditary non-polyposis colorectal cancer. Anticancer Res 1994;14:1631-4.
Mecklin JP, Järvinen HJ. Clinical features of colorectal carcinoma in cancer family
syndrome. Dis Colon Rectum 1986;29:160-4.
Gibbs NM. Undifferentiated carcinoma of the large intestine. Histopathology
1977;1:77-84.
Jessurun MR, Manivel JC. Cecal, poorly differentiated adenocarcinomas, medullary-type. Mod Pathol 1992;5:43A (abstract).
Lynch HT, Smyrk T, Lanspa SJ, Marcus J, Bowman BM, Watson P. Pathology and
genetic markers of colorectal cancer in Lynch syndromes I and II. J Tumor Markers
Oncol 1988;3:341-50.
Kim H, Jen J, Vogelstein B, Hamilton SR. Clinical and pathological characteristics
of sporadic colorectal carcinomas with DNA replication errors in microsatellie
sequences. Am J Pathol 1994;145:148-56.
Thibodeau SN, French AJ, Roche PC, Cunningham JM, Tester DJ, Lindor NM, et
al. Altered expression of hMSH2 and hMLH1 in tumors with microsatellite
instability and genetic alterations in mismatch repair genes. Cancer Res
1996;56:4836-40.
Smyrk TC, Lynchn HT, Watson PA, Appelman HD. Histologic features of
hereditary nonpolyposis colorectal carcinoma. In: Utsunomiya J, Lynch HT,
editors. Hereditary Colorectal Cancer. Tokyo: Springer-Verlag; 1990:357– 62.
Curr Probl Surg, May 2005
373. Graham DM, Appelman HD. Crohn’s-like lymphoid reaction and colorectal
carcinoma: a potential histologic prognosticator. Mod Pathol 1990;3:332-5.
374. Watson P, Lin KM, Rodriguez-Bigas MA, Smyrk T, Lemon S, Shashidharan M, et
al. Colorectal carcinoma survival among hereditary nonpolyposis colorectal carcinoma family members. Cancer 1998;83:259-66.
375. De Jong AE, Morreau H, Van Puijenbroek M, Eilers PH, Wijnen J, Nagengast FM,
et al. The role of mismatch repair gene defects in the development of adenomas in
patients with HNPCC. Gastroenterology 2004;126:42-8.
376. Lynch PM, Lynch HT, Harris RE. Hereditary proximal colonic cancer. Dis Colon
Rectum 1977;20:661-8.
377. Albano WA, Recabaren JA, Lynch HT, Campbell AS, Mailliard JA, Organ CH, et
al. Natural history of hereditary cancer of the breast and colon. Cancer
1982;50:360-3.
378. Lynch HT, Watson P, Lanspa SJ, Marcus J, Smyrk T, Fitzgibbons RJ Jr, et al.
Natural history of colorectal cancer in hereditary nonpolyposis colorectal cancer
(Lynch syndromes I and II). Dis Colon Rectum 1988;31:439-44.
379. Lynch HT, Lin K, Smyrk T, Watson P, Thorson A, Shashidharan M. Colorectal
cancer, pathology staging and survival in HNPCC. ASCO Program/Proceedings
1997;16:530A (abstract).
380. Smyrk TC, Watson P, Kaul K, Lynch HT. Tumor-infiltrating lymphocytes are a
marker for microsatellite instability in colorectal carcinoma. Cancer
2001;91:2417-22.
381. Risinger JI, Barrett JC, Watson P, Lynch HT, Boyd J. Molecular genetic evidence
of the occurrence of breast cancer as an integral tumor in patients with the
hereditary nonpolyposis colorectal carcinoma syndrome. Cancer 1996;77:1836-43.
382. Lynch HT, Lynch PM, Pester J, Fusaro RM. The cancer family syndrome. Rare
cutaneous phenotypic linkage of Torre’s syndrome. Arch Intern Med
1981;141:607-11.
383. Lynch HT, Lynch PM, Pester JA, Fusaro RM. Sebaceous neoplasia and visceral
cancer (Torre’s syndrome) and its relationship to the cancer family syndrome. In:
Lynch HT, Fusaro RM, editors. Cancer-Associated Genodermatoses. New York:
Van Nostrand Reinhold; 1982.
384. Lynch HT, Fusaro RM, Roberts L, Voorhees GJ, Lynch JF. Muir-Torre syndrome
in several members of a family with a variant of the cancer family syndrome. Br J
Dermatol 1985;113:295-301.
385. Lynch HT, Fusaro RM. Muir-Torre syndrome: heterogeneity, natural history,
diagnosis, and management. Prob Gen Surg 1993;10:1-14.
386. Lynch HT, Leibowitz R, Smyrk T, Fusaro RM, Lynch JF, Smith A, et al. Colorectal
cancer and the Muir-Torre syndrome in a Gypsy family: a review. Am J
Gastroenterol 1999;94:575-80.
387. Kruse R, Rutten A, Lamberti C, Hosseiny-Malayeri HR, Wang Y, Ruelfs C, et al.
Muir-Torre phenotype has a frequency of DNA mismatch-repair-gene mutations
similar to that in hereditary nonpolyposis colorectal cancer families defined by the
Amsterdam criteria. Am J Hum Genet 1998;63:63-70.
388. Fitzgibbons RJ Jr, Lynch HT, Stanislav GV, Watson PA, Lanspa SJ, Marcus JN, et
al. Recognition and treatment of patients with hereditary nonpolyposis colon cancer
(Lynch syndromes I and II). Ann Surg 1987;206:289-95.
Curr Probl Surg, May 2005
321
389. Church J. Hereditary colon cancers can be tiny: a cautionary case report of the
results of colonoscopic surveillance. Am J Gastroenterol 1998;93:2289-90.
390. Rex DK, Cutler CS, Lemmel GT, Rahmani EY, Clark DW, Helper DJ, et al.
Colonoscopic miss rates of adenomas determined by back-to-back colonoscopies.
Gastroenterology 1997;112:24-8.
391. Lynch HT. Is there a role for prophylactic subtotal colectomy among hereditary
nonpolyposis colorectal cancer germline mutation carriers? Dis Colon Rectum
1996;39:109-10.
392. Church JM. Prophylactic colectomy in patients with hereditary nonpolyposis
colorectal cancer. Ann Med 1996;28:479-82.
393. Scaife CL, Rodriguez-Bigas MA. Lynch syndrome: implications for the surgeon.
Clin Colorectal Cancer 2003;3:92-8.
394. Lynch HT, Lynch JF, Fitzgibbons R Jr. Role of prophylactic colectomy in Lynch
syndrome. Clin Colorectal Cancer 2003;3:99-101.
395. Jass JR. Colorectal adenoma progression and genetic change: is there a link? Ann
Med 1995;27:301-6.
396. Jass JR, Stewart SM. Evolution of hereditary non-polyposis colorectal cancer. Gut
1992;33:783-6.
397. Sankila R, Aaltonen LA, Järvinen HJ, Mecklin J-P. Better survival rates in patients
with MLH1-associated hereditary colorectal cancer. Gastroenterology
1996;110:682-7.
398. Vasen HF, Nagengast FM, Khan PM. Interval cancers in hereditary non-polyposis
colorectal cancer (Lynch syndrome). Lancet 1995;345:1183-4.
399. Diamond M. Adenoma of the rectum in children: report of a case in a thirty month
old girl. Am J Dis Child 1939;57:360-7.
400. Mauro J, Prior JT. Gastrointestinal polypoid lesions in childhood. Cancer
1957;10:131-7.
401. Horrilleno EG, Eckert C, Ackerman LV. Polyps of the rectum and colon in
children. Cancer 1957;10:1210-2.
402. Morson BC. Some peculiarities in the histology of intestinal polyps. Dis Colon
Rectum 1962;5:337-44.
403. McColl I, Bussey HJR, Veale AMO, Morson BC. Juvenile polyposis coli. Proc R
Soc Med 1964;57:896-7.
404. Sachatello CR, Hahn IL, Carrington CB. Juvenile gastrointestinal polyposis in a
female infant: report of a case and review of the literature of a recently recognized
syndrome. Surgery 1974;75:107-14.
405. Jass JR, Williams CB, Bussey HJR, Morson BC. Juvenile polyposis—a precancerous condition. Histopathology 1988;13:619-30.
406. Ravitch MM. Polypoid adenomatosis of the entire gastrointestinal tract. Ann Surg
1948;128:283-98.
407. LeFevre HW Jr, Jacques TF. Multiple polyposis in an infant of 4 months. Am J
Surg 1951;81:90-1.
408. Raymann FB. Juvenile polyps with cachexia. Gastroenterology 1969;57:431.
409. Arbeter AM, Courtney RA, Gaynor MF Jr. Diffuse gastrointestinal polyposis
associated with chronic blood loss, hypoproteinemia, and anasarca in an infant.
J Pediatr 1970;76:609-11.
410. Soper RT, Kent TH. Fatal juvenile polyposis of infancy. Surgery 1971;69:692-8.
322
Curr Probl Surg, May 2005
411. Roth SI, Helwig EB. Juvenile polyps of the colon and rectum. Cancer
1963;16:468-79.
412. Reed K, Vose PC. Diffuse juvenile polyposis of the colon: a premalignant
condition? Dis Colon Rectum 1981;24:205-10.
413. Coburn MC, Pricolo VE, DeLuca FG, Bland KI. Malignant potential in intestinal
juvenile polyposis syndromes. Ann Surg Oncol 1995;2:386-91.
414. Stemper TJ, Kent TH, Summers RW. Juvenile polyposis and gastrointestinal
carcinoma. Ann Intern Med 1975;83:639-46.
415. Grotsky HW, Rickert RR, Smith WD, Newsome JF. Familial juvenile polyposis
coli: a clinical and pathologic study of a large kindred. Gastroenterology
1982;82:494-501.
416. Desai DC, Murday V, Phillips RKS, Neale KF, Milla P, Hodgson SV. A survey of
phenotypic features in juvenile polyposis. J Med Genet 1998;35:476-81.
417. Gelb AM, Minkowitz S, Tresser M. Rectal and colonic polyps occurring in young
people. NY State J Med 1962;62:513-8.
418. Haggitt RC, Pitcock JA. Familial juvenile polyposis of the colon. Cancer
1970;26:1232-8.
419. Kaschula RO. Mixed juvenile, adenomatous and intermediate polyposis coli: report
of a case. Dis Colon Rectum 1971;14:368-74.
420. Velcek FT, Coopersmith IS, Chen CK, Kassner EG, Klotz DH Jr, Kottmeier PK.
Familial juvenile adenomatous polyposis. J Pediatr Surg 1976;11:781-7.
421. Beacham CH, Shields HM, Raffensperger EC, Enterline HT. Juvenile and adenomatous gastrointestinal polyposis. Dig Dis 1978;23:1137-43.
422. Goodman ZD, Yardley JH, Milligan FD. Pathogenesis of colonic polyps in multiple
juvenile polyposis. Cancer 1979;43:1906-13.
423. Monga G, Mazzucco G, Rossini FP, Presti F. Colorectal polyposis with mixed
juvenile and adenomatous patterns. Virchows Arch A Pathol Anat Histol
1979;382:355-60.
424. Sandler RS, Lipper S. Multiple adenomas in juvenile polyposis. Am J Gastroenterol
1981;75:361-6.
425. Friedman CJ, Fechner RE. A solitary juvenile polyp with hyperplastic and
adenomatous glands. Dig Dis Sci 1982;27:946-8.
426. Mills SE, Fechner RE. Unusual adenomatous polyps in juvenile polyposis coli.
Am J Surg Pathol 1982;6:177-83.
427. Rozen P, Baratz M. Familial juvenile colonic polyposis with associated colon
cancer. Cancer 1982;49:1500-3.
428. Järvinen HJ, Franssila KO. Familial juvenile polyposis coli: increased risk of
colorectal cancer. Gut 1984;25:792-800.
429. Yoshida T, Haraguchi Y, Tanaka A, Higa A, Daimon Y, Mizuta Y, et al. A case of
generalized juvenile gastrointestinal polyposis associated with gastric carcinoma.
Endoscopy 1988;20:33-5.
430. Bentley E, Chandrasoma P, Radin R, Cohen H. Generalized juvenile polyposis with
carcinoma. Am J Gastroenterol 1989;84:1456-9.
431. Longo WE, Touloukian RJ, West B, Ballantyne GH. Malignant potential of
juvenile polyposis coli. Dis Colon Rectum 1990;33:980-4.
432. Giardiello FM, Hamilton SR, Kern SE, Offerhaus GJA, Green PA, Celano P, et al.
Colorectal neoplasia in juvenile polyposis or juvenile polyps. Arch Dis Child
1991;66:971-5.
Curr Probl Surg, May 2005
323
433. O’Riordain DS, O’Dwyer PJ, Cullen BF, McDermott EW, Murphy JJ. Familial
juvenile polyposis coli and colorectal cancer. Cancer 1991;68:889-92.
434. Heiss KF, Schaffner D, Ricketts RR, Winn K. Malignant risk in juvenile polyposis
coli: increasing documentation in the pediatric age group. J Pediatr Surg
1993;28:1188-93.
435. Ramaswamy G, Elhosseiny AA, Tchertkoff V. Juvenile polyposis of the colon with
atypical adenomatous changes and carcinoma in situ. Dis Colon Rectum
1984;27:393-8.
436. Jones MA, Hebert JC, Trainer TD. Juvenile polyp with intramucosal carcinoma.
Arch Pathol Lab Med 1987;111:200-1.
437. Smilow PC, Pryor CA, Swinton NW. Juvenile polyposis coli: a report of three
patients in three generations of one family. Dis Colon Rectum 1966;9:248-54.
438. Restrepo C, Moreno J, Duque E, Cuello C, Amsel J, Correa P. Juvenile colonic
polyposis in Colombia. Dis Colon Rectum 1978;21:600-12.
439. Howe JR, Mitros FA, Summers RW. The risk of gastrointestinal carcinoma in
familial juvenile polyposis. Ann Surg Oncol 1998;5:751-6.
440. Murday V, Slack J. Inherited disorders associated with colorectal cancer. Cancer
Surv 1989;8:139-57.
441. Liu T-H, Chen M-C, Tseng H-C. Malignant change of juvenile polyp of colon.
Chin Med J (Engl) 1978;4:434-9.
442. Nugent KP, Talbot IC, Hodgson SV, Phillips RK. Solitary juvenile polyps: not a
marker for subsequent malignancy. Gastroenterology 1993;105:698-700.
443. Veale AMO, McColl I, Bussey HJR, Morson BC. Juvenile polyposis coli. J Med
Genet 1966;3:5-16.
444. Sachatello CR, Pickren JW, Grace JT. Generalized juvenile gastrointestinal
polyposis. Gastroenterology 1970;58:699-708.
445. Jacoby RF, Schlack S, Sekhon G, Laxova R. Del(10)(q22.3q24.1) associated with
juvenile polyposis. Am J Med Genet 1997;70:361-4.
446. Olschwang S, Serova-Sinilnikova OM, Lenoir GM, Thomas G. PTEN germ-line
mutations in juvenile polyposis coli. Nat Genet 1998;18:12-4.
447. Eng C, Ji H. Molecular classification of the inherited hamartoma polyposis
syndromes: clearing the muddied waters. Am J Hum Genet 1998;62:1020-2.
448. Howe JR, Ringold JC, Summers RW, Mitros FA, Nishimura DY, Stone EM. A
gene for familial juvenile polyposis maps to chromosome 18q21.1. Am J Hum
Genet 1998;62:1129-36.
449. Marsh DJ, Roth S, Lunetta KL, Hemminki A, Dahia PLM, Sistonen P, et al.
Exclusion of PTEN and 10q22-24 as the susceptibility locus for juvenile polyposis
syndrome. Cancer Res 1997;57:5017-21.
450. Riggins GJ, Hamilton SR, Kinzler KW, Vogelstein B. Normal PTEN gene in
juvenile polyposis. J Neg Observ Genet Oncol 1997;1:1.
451. Howe JR, Roth S, Ringold JC, Summers RW, Järvinen HJ, Sistonen P, et al.
Mutations in the SMAD4/DPC4 gene in juvenile polyposis. Science
1998;280:1086-8.
452. Houlston R, Bevan S, Williams A, Young J, Dunlop M, Rozen P, et al. Mutations
in DPC4 (SMAD4) cause juvenile polyposis syndrome, but only account for a
minority of cases. Hum Mol Genet 1998;7:1907-12.
453. Friedl W, Kruse R, Uhlhaas S, Stolte M, Schartmann B, Keller KM, et al. Frequent
324
Curr Probl Surg, May 2005
454.
455.
456.
457.
458.
459.
460.
461.
462.
463.
464.
465.
466.
467.
468.
469.
470.
4-bp deletion in exon 9 of the SMAD4/MADH4 gene in familial juvenile polyposis
patients. Genes Chromosomes Cancer 1999;25:403-6.
Roth S, Sistonen P, Salovaara R, Hemminki A, Loukola A, Johansson M, et al.
SMAD genes in juvenile polyposis. Genes Chromosomes Cancer 1999;26:54-61.
Kim IJ, Ku JL, Yoon KA, Heo SC, Jeong SY, Choi HS, et al. Germline mutations
of the dpc4 gene in Korean juvenile polyposis patients. Int J Cancer
2000;86:529-32.
Woodford-Richens K, Bevan S, Churchman M, Dowling B, Jones D, Norbury CG,
et al. Analysis of genetic and phenotypic heterogeneity in juvenile polyposis. Gut
2000;46:656-60.
Howe JR, Sayed MG, Ahmed AF, Ringold J, Larsen-Haidle J, Merg A, et al. The
prevalence of MADH4 and BMPR1A mutations in juvenile polyposis and absence
of BMPR2, BMPR1B, and ACVR1 mutations. J Med Genet 2004;41:484-91.
Howe JR, Shellnut J, Wagner B, Ringold JC, Sayed MG, Ahmed AF, et al.
Common deletion of SMAD4 in juvenile polyposis is a mutational hotspot. Am J
Hum Genet 2002;70:1357-62.
Hahn SA, Shutte M, Shamsul Hoque ATM, Moskaluk CA, da Costa LT,
Rozenblum E, et al. DPC4, a candidate tumor suppressor gene at human
chromosome 18q21.1. Science 1996;271:350-3.
Heldin C-H, Miyazono K, Ten Dijke P. TGF-␤ signaling from cell membrane to
nucleus through SMAD proteins. Nature 1997;390:465-71.
Bevan S, Woodford-Richens K, Rozen P, Eng C, Young J, Dunlop M, et al.
Screening SMAD1, SMAD2, SMAD3, and SMAD5 for germline mutations in
juvenile polyposis syndrome. Gut 1999;45:406-8.
Howe JR, Bair JL, Sayed MG, Anderson ME, Mitros FA, Petersen GM, et al.
Germline mutations of the gene encoding bone morphogenetic protein receptor 1A
in juvenile polyposis. Nat Genet 2001;28:184-7.
Zhou XP, Woodford-Richens K, Lehtonen R, Kurose K, Aldred M, Hampel H, et
al. Germline mutations in BMPR1A/ALK3 cause a subset of cases of juvenile
polyposis syndrome and of Cowden and Bannayan-Riley-Ruvalcaba syndromes.
Am J Hum Genet 2001;69:704-11.
Friedl W, Uhlhaas S, Schulman K, Stolte M, Loff S, Back W, et al. Juvenile
polyposis: massive gastric polyposis is more common in MADH4 mutation carriers
than in BMPR1A mutation carriers. Hum Genet 2002;111:108-11.
Mehra A, Wrana JL. TGF-b and the Smad signal transduction pathway. Biochem
Cell Biol 2002;80:605-22.
Reddi AH. Bone morphogenetic proteins: an unconventional approach to isolation
of first mammalian morphogens. Cytokine Growth Factor Rev 1997;8:11-20.
Sayed MG, Ahmed AF, Ringold JC, ME. A, Bair JL, Mitros FA, et al. Germline
SMAD4 or BMPR1A mutations and phenotype of juvenile polyposis. Ann Surg
Oncol 2002;9:901-6.
Howe JR, Ringold JC, Hughes J, Summers RW. Direct genetic testing for Smad4
mutations in patients at risk for juvenile polyposis. Surgery 1999;126:162-70.
Lloyd KM 2nd, Dennis M. Cowden’s disease: a possible new symptom complex
with multiple system involvement. Ann Intern Med 1963;58:136-42.
Weary PE, Gorlin RJ, Gentry WC Jr, Comer JE, Greer KE. Multiple hamartoma
syndrome (Cowden’s disease). Arch Dermatol 1972;106:682-90.
Curr Probl Surg, May 2005
325
471. Marano SR, Johnson PC, Spetzler RF. Recurrent Lhermitte-Duclos disease in a
child: case report. J Neurosurg 1988;69:599-603.
472. Milbouw G, Born JD, Martin D, Collignon J, Hans P, Reznik M, et al. Clinical and
radiological aspects of dysplastic gangliocytoma (Lhermitte-Duclos disease): a
report of two cases with review of the literature. Neurosurgery 1988;22:124-8.
473. Padberg GW, Schot JD, Vielvoye GJ, Bots GT, de Beer FC. Lhermitte-Duclos
disease and Cowden disease: a single phakomatosis. Ann Neurol 1991;29:517-23.
474. Rimbau J, Isamat F. Dysplastic gangliocytoma of the cerebellum (LhermitteDuclos disease) and its relation to the multiple hamartoma syndrome (Cowden
disease). J Neurooncol 1994;18:191-7.
475. Vinchon M, Blond S, Lejeune JP, Krivosik I, Fossati P, Assaker R, et al.
Association of Lhermitte-Duclos and Cowden disease: report of a new case and
review of the literature. J Neurol Neurosurg Psychiatry 1994;57:699-704.
476. Wells GB, Lasner TM, Yousem DM, Zager EL. Lhermitte-Duclos disease and
Cowden’s syndrome in an adolescent patient: case report. J Neurosurg
1994;81:133-6.
477. Thomas DW, Lewis MA. Lhermitte-Duclos disease associated with Cowden’s
disease. Int J Oral Maxillofac Surg 1995;24:369-71.
478. Starink TM, van der Veen JP, Arwert F, de Waal LP, de Lange GG, Gille JJ, et al.
The Cowden syndrome: a clinical and genetic study in 21 patients. Clin Genet
1986;29:222-33.
479. Hanssen AM, Fryns JP. Cowden syndrome. J Med Genet 1995;32:117-9.
480. Brownstein MH, Mehregan AH, Bilowski JB. Trichilemmomas in Cowden’s
disease. JAMA 1977;238:26 (Letter).
481. Brownstein MH, Mehregan AH, Bikowski JB, Lupulescu A, Patterson JC. The
dermatopathology of Cowden’s syndrome. Br J Dermatol 1979;100:667-73.
482. Burnett JW, Goldner R, Calton GJ. Cowden disease: report of two additional cases.
Br J Dermatol 1975;93:329-36.
483. Starink TM. Cowden’s disease: analysis of fourteen new cases. J Am Acad
Dermatol 1984;11:1127-41.
484. Harach HR, Soubeyran I, Brown A, Bonneau D, Longy M. Thyroid pathologic
findings in patients with Cowden disease. Ann Diagn Pathol 1999;3:331-40.
485. Thyresson HN, Doyle JA. Cowden’s disease (multiple hamartoma syndrome).
Mayo Clin Proc 1981;56:179-84.
486. Kameyama K, Takami H, Miyajima K, Mimura T, Hosoda Y, Ito K. Papillary
carcinoma occurring within an adenomatous goiter of the thyroid gland in
Cowden’s disease. Endocr Pathol 2001;12:73-6.
487. Tsubosa Y, Fukutomi T, Tsuda H, Kanai Y, Akashi-Tanaka S, Nanasawa T, et al.
Breast cancer in Cowden’s disease: a case report with review of the literature. Jpn
J Clin Oncol 1998;28:42-6.
488. Fackenthal JD, Marsh DJ, Richardson AL, Cummings SA, Eng C, Robinson BG,
et al. Male breast cancer in Cowden syndrome patients with germline PTEN
mutations. J Med Genet 2001;38:159-64.
489. Lashner BA, Riddell RH, Winans CS. Ganglioneuromatosis of the colon and
extensive glycogenic acanthosis in Cowden’s disease. Dig Dis Sci 1986;31:213-6.
490. Taylor AJ, Dodds WJ, Stewart ET. Alimentary tract lesions in Cowden’s disease.
Br J Radiol 1989;62:890-2.
326
Curr Probl Surg, May 2005
491. Carlson GJ, Nivatvongs S, Snover DC. Colorectal polyps in Cowden’s disease
(multiple hamartoma syndrome). Am J Surg Pathol 1984;8:763-70.
492. Hauser H, Ody B, Plojoux O, Wettstein P. Radiological findings in multiple
hamartoma syndrome (Cowden disease): a report of three cases. Radiology
1980;137:317-23.
493. Hizawa K, Iida M, Matsumoto T, Kohrogi N, Suekane H, Yao T, et al.
Gastrointestinal manifestations of Cowden’s disease: report of four cases. J Clin
Gastroenterol 1994;18:13-8.
494. Gentry WC Jr, Eskritt NR, Gorlin RJ. Multiple hamartoma syndrome (Cowden
disease). Arch Dermatol 1974;109:521-5.
495. Marra G, Armelao F, Vecchio FM, Percesepe A, Anti M. Cowden’s disease with
extensive gastrointestinal polyposis. J Clin Gastroenterol 1994;18:42-7.
496. Allen BS, Fitch MH, Smith JG Jr. Multiple hamartoma syndrome: a report of a new
case with associated carcinoma of the uterine cervix and angioid streaks of the eyes.
J Am Acad Dermatol 1980;2:303-8.
497. Gorensek M, Matko I, Skralovnik A, Rode M, Satler J, Jutersek A. Disseminated
hereditary gastrointestinal polyposis with orocutaneous hamartomatosis (Cowden’s
disease). Endoscopy 1984;16:59-63.
498. Hover AR, Cawthern T, McDanial W. Cowden disease. A hereditary polyposis
syndrome diagnosable by mucocutaneous inspection. J Clin Gastroenterol
1986;8:576-9.
499. Eng C. Will the real Cowden syndrome please stand up: revised diagnostic criteria.
J Med Genet 2000;37:828-30.
500. Lhermitte J, Duclos P. Sur un Ganglioneurome diffus du cortex du cervelet. Bull
Assoc Fr Etude Cancer 1920;9:99-107.
501. Ambler M, Pogacar S, Sidman R. Lhermitte-Duclos disease (granule cell hypertrophy of the cerebellum) pathological analysis of the first familial cases. J Neuropathol Exp Neurol 1969;28:622-47.
502. Smith RR, Grossman RI, Goldberg HI, Hackney DB, Bilaniuk LT, Zimmerman
RA. MR imaging of Lhermitte-Duclos disease: a case report. AJNR Am J
Neuroradiol 1989;10:187-9.
503. Williams DW 3rd, Elster AD, Ginsberg LE, Stanton C. Recurrent Lhermitte-Duclos
disease: report of two cases and association with Cowden’s disease. AJNR Am J
Neuroradiol 1992;13:287-90.
504. Eng C, Murday V, Seal S, Mohammed S, Hodgson SV, Chaudary MA, et al.
Cowden syndrome and Lhermitte-Duclos disease in a family: a single genetic
syndrome with pleiotropy? J Med Genet 1994;31:458-61.
505. Robinson S, Cohen AR. Cowden disease and Lhermitte-Duclos disease: characterization of a new phakomatosis. Neurosurgery 2000;46:371-83.
506. Vantomme N, Van Calenbergh F, Goffin J, Sciot R, Demaerel P, Plets C.
Lhermitte-Duclos disease is a clinical manifestation of Cowden’s syndrome. Surg
Neurol 2001;56:201-4; discussion 204-5.
507. Nelen MR, Padberg GW, Peeters EA, Lin AY, van den Helm B, Frants RR, et al.
Localization of the gene for Cowden disease to chromosome 10q22-23. Nat Genet
1996;13:114-6.
508. Nuss DD, Aeling JL, Clemons DE, Weber WN. Multiple hamartoma syndrome
(Cowden’s disease). Arch Dermatol 1978;114:743-6.
509. Salem OS, Steck WD. Cowden’s disease (multiple hamartoma and neoplasia
Curr Probl Surg, May 2005
327
510.
511.
512.
513.
514.
515.
516.
517.
518.
519.
520.
521.
522.
523.
524.
525.
526.
328
syndrome): a case report and review of the English literature. J Am Acad Dermatol
1983;8:686-96.
Li DM, Sun H. TEP1, encoded by a candidate tumor suppressor locus, is a novel
protein tyrosine phosphatase regulated by transforming growth factor beta. Cancer
Res 1997;57:2124-9.
Li J, Yen C, Liaw D, Podsypanina K, Bose S, Wang SI, et al. PTEN, a putative
protein tyrosine phosphatase gene mutated in human brain, breast, and prostate
cancer. Science 1997;275:1943-7.
Steck PA, Pershouse MA, Jasser SA, Yung WK, Lin H, Ligon AH, et al.
Identification of a candidate tumour suppressor gene, MMAC1, at chromosome
10q23.3 that is mutated in multiple advanced cancers. Nat Genet 1997;15:356-62.
Liaw D, Marsh DJ, Li J, Dahia PL, Wang SI, Zheng Z, et al. Germline mutations
of the PTEN gene in Cowden disease, an inherited breast and thyroid cancer
syndrome. Nat Genet 1997;16:64-7.
Nelen MR, van Staveren WC, Peeters EA, Hassel MB, Gorlin RJ, Hamm H, et al.
Germline mutations in the PTEN/MMAC1 gene in patients with Cowden disease.
Hum Mol Genet 1997;6:1383-7.
Myers MP, Stolarov JP, Eng C, Li J, Wang SI, Wigler MH, et al. P-TEN, the tumor
suppressor from human chromosome 10q23, is a dual-specificity phosphatase. Proc
Natl Acad Sci USA 1997;94:9052-7.
Myers MP, Pass I, Batty IH, Van der Kaay J, Stolarov JP, Hemmings BA, et al. The
lipid phosphatase activity of PTEN is critical for its tumor suppressor function. Proc
Natl Acad Sci USA 1998;95:13513-8.
Weng LP, Smith WM, Brown JL, Eng C. PTEN inhibits insulin-stimulated
MEK/MAPK activation and cell growth by blocking IRS-1 phosphorylation and
IRS-1/Grb-2/Sos complex formation in a breast cancer model. Hum Mol Genet
2001;10:605-16.
Marsh DJ, Coulon V, Lunetta KL, Rocca-Serra P, Dahia PL, Zheng Z, et al.
Mutation spectrum and genotype-phenotype analyses in Cowden disease and
Bannayan-Zonana syndrome, two hamartoma syndromes with germline PTEN
mutation. Hum Mol Genet 1998;7:507-15.
Zhou XP, Waite KA, Pilarski R, Hampel H, Fernandez MJ, Bos C, et al. Germline
PTEN promoter mutations and deletions in Cowden/Bannayan-Riley-Ruvalcaba
syndrome result in aberrant PTEN protein and dysregulation of the phosphoinositol-3-kinase/Akt pathway. Am J Hum Genet 2003;73:404-11.
Eng C. Cowden syndrome. J Genet Counsel 1997;6:181-92.
Eng C. Genetics of Cowden syndrome: through the looking glass of oncology. Int
J Oncol 1998;12:701-10.
Yen BC, Kahn H, Schiller AL, Klein MJ, Phelps RG, Lebwohl MG. Multiple
hamartoma syndrome with osteosarcoma. Arch Pathol Lab Med 1993;117:1252-4.
Ruvalcaba RH, Myhre S, Smith DW. Sotos syndrome with intestinal polyposis and
pigmentary changes of the genitalia. Clin Genet 1980;18:413-6.
Halal F. Male to male transmission of cerebral gigantism. Am J Med Genet
1982;12:411-9.
Halal F. Cerebral gigantism, intestinal polyposis, and pigmentary spotting of the
genitalia. Am J Med Genet 1983;15:161.
Cohen MM, Jr. Bannayan-Riley-Ruvalcaba syndrome: renaming three formerly
recognized syndromes as one etiologic entity. Am J Med Genet 1990;35:291-2.
Curr Probl Surg, May 2005
527. DiLiberti JH, Weleber RG, Budden S. Ruvalcaba-Myhre-Smith syndrome: a case
with probable autosomal-dominant inheritance and additional manifestations. Am J
Med Genet 1983;15:491-5.
528. DiLiberti JH, D’Agostino AN, Ruvalcaba RH, Schimschock JR. A new lipid
storage myopathy observed in individuals with the Ruvalcaba-Myhre-Smith syndrome. Am J Med Genet 1984;18:163-7.
529. Gretzula JC, Hevia O, Schachner LS, DiLiberti JH, Ruvalcaba RH, Schimschock
JR, et al. Ruvalcaba-Myhre-Smith syndrome. Pediatr Dermatol 1988;5:28-32.
530. Bannayan GA. Lipomatosis, angiomatosis, and macrencephalia: a previously
undescribed congenital syndrome. Arch Pathol 1971;92:1-5.
531. Zonana J, Rimoin DL, Davis DC. Macrocephaly with multiple lipomas and
hemangiomas. J Pediatr 1976;89:600-3.
532. Higginbottom MC, Schultz P. The Bannayan syndrome: an autosomal dominant
disorder consisting of macrocephaly, lipomas, hemangiomas, and risk for intracranial tumors. Pediatrics 1982;69:632-4.
533. Saul RA, Stevenson RE, Bley R. Mental retardation in the Bannayan syndrome.
Pediatrics 1982;69:642-4.
534. Miles JH, Zonana J, McFarlane J, Aleck KA, Bawle E. Macrocephaly with
hamartomas: Bannayan-Zonana syndrome. Am J Med Genet 1984;19:225-34.
535. Klein JA, Barr RJ. Bannayan-Zonana syndrome associated with lymphangiomyomatous lesions. Pediatr Dermatol 1990;7:48-53.
536. Riley HD, Smith WR. Macrocephaly, pseudopapilledema and multiple hemangiomata. Pediatrics 1960:293-300.
537. Foster MA, Kilcoyne RF. Ruvalcaba-Myhre-Smith syndrome: a new consideration
in the differential diagnosis of intestinal polyposis. Gastrointest Radiol
1986;11:349-50.
538. Haggitt RC, Reid BJ. Hereditary gastrointestinal polyposis syndrome. Am J Surg
Pathol 1986;10:871-87.
539. Zigman AF, Lavine JE, Jones MC, Boland CR, Carethers JM. Localization of the
Bannayan-Riley-Ruvalcaba syndrome gene to chromosome 10q23. Gastroenterology 1997;113:1433-7.
540. Coyle WJ, Ryan MT, Nowicki MJ. Adenomatous changes in the hamartomatous
polyps of Ruvalcaba-Myhre-Smith syndrome. J Clin Gastroenterol 1998;26:85-7.
541. Lowichik A, White FV, Timmons CF, Weinberg AG, Gunasekaran TS, Nathan K,
et al. Bannayan-Riley-Ruvalcaba syndrome: spectrum of intestinal pathology
including juvenile polyps. Pediatr Dev Pathol 2000;3:155-61.
542. Fargnoli MC, Orlow SJ, Semel-Concepcion J, Bolognia JL. Clinicopathologic findings
in the Bannayan-Riley-Ruvalcaba syndrome. Arch Dermatol 1996;132:1214-8.
543. Gorlin RJ, Cohen MM Jr, Condon LM, Burke BA. Bannayan-Riley-Ruvalcaba
syndrome. Am J Med Genet 1992;44:307-14.
544. Marsh DJ, Kum JB, Lunetta KL, Bennett MJ, Gorlin RJ, Ahmed SF, et al. PTEN mutation
spectrum and genotype-phenotype correlations in Bannayan-Riley-Ruvalcaba syndrome
suggest a single entity with Cowden syndrome. Hum Mol Genet 1999;8:1461-72.
545. Parisi MA, Dinulos MB, Leppig KA, Sybert VP, Eng C, Hudgins L. The spectrum
and evolution of phenotypic findings in PTEN mutation positive cases of Bannayan-Riley-Ruvalcaba syndrome. J Med Genet 2001;38:52-8.
546. Figer A, Kaplan A, Frydman M, Lev D, Paswell J, Papa MZ, et al. Germline
Curr Probl Surg, May 2005
329
563.
mutations in the PTEN gene in Israeli patients with Bannayan-Riley-Ruvalcaba
syndrome and women with familial breast cancer. Clin Genet 2002;62:298-302.
Peutz JL. A very remarkable case of familial polyposis of mucous membrane of
intestinal tract and accompanied by peculiar pigmentations of skin and mucous
membrane. Ned Tijdschr Geneeskd 1921;10:134-46.
Jeghers H, McKusick VA, Katz KH. Generalized intestinal polyposis and melanin
spots of oral mucosa, lips, and digits. N Engl J Med 1949;241:993-1005, 1031-6.
Mallory SB, Stough DBt.Genodermatoses with malignant potential. Dermatol Clin
1987;5:221-30.
Bartholomew LG, Dahlin DC, Waugh JM. Intestinal polyposis associated with
mucocutaneous melanin pigmentation (Peutz-Jeghers syndrome). Gastroenterology
1957;32:434-51.
Bartholomew LG, More CE, Dahlin DC, Waugh JM. Intestinal polyposis associated with mucocutaneous pigmentation. Surg Gynecol Obstet 1962;115:1-11.
McGarrity TJ, Kulin HE, Zaino RJ. Peutz-Jeghers syndrome. Am J Gastroenterol
2000;95:596-604.
Spigelman AD, Murday V, Phillips RK. Cancer and the Peutz-Jeghers syndrome.
Gut 1989;30:1588-90.
Giardiello FM, Welsh SB, Hamilton SR, Offerhaus GJ, Gittelsohn AM, Booker SV,
et al. Increased risk of cancer in the Peutz-Jeghers syndrome. N Engl J Med
1987;316:1511-4.
Lim W, Hearle N, Shah B, Murday V, Hodgson SV, Lucassen A, et al. Further
observations on LKB1/STK11 status and cancer risk in Peutz-Jeghers syndrome.
Br J Cancer 2003;89:308-13.
Boardman LA, Thibodeau SN, Schaid DJ, Lindor NM, McDonnel SK, Burgart LJ,
et al. Increased risk for cancer in patients with the Peutz-Jeghers syndrome. Ann
Intern Med 1998128:896 –9.
Hemminki A. The molecular basis and clinical aspects of Peutz-Jeghers syndrome.
Cell Mol Life Sci 1999;55:735-50.
Giardiello FM, Brensinger JD, Tersmette AC, Goodman SN, Petersen GM, Booker
SV, et al. Very high risk of cancer in familial Peutz-Jeghers syndrome. Gastroenterology 2000;119:1447-53.
Hemminki A, Tomlinson I, Markie D, Järvinen H, Sistonen P, Bjorkqvist AM, et
al. Localization of a susceptibility locus for Peutz-Jeghers syndrome to 19p using
comparative genomic hybridization and targeted linkage analysis. Nat Genet
1997;15:87-90.
Amos CI, Bali D, Thiel TJ, Anderson JP, Gourley I, Frazier ML, et al. Fine
mapping of a genetic locus for Peutz-Jeghers syndrome on chromosome 19p.
Cancer Res 1997;57:3653-6.
Mehenni H, Blouin J-L, Radhakrishna U, Bhardwaj SS, Bhardwaj K, Dixit VB, et
al. Peutz-Jeghers syndrome: confirmation of linkage to chromosome 19p13.3 and
identification of a potential second locus, on 19q13.4. Am J Hum Genet
1997;61:1327-34.
Hemminki A, Markie D, Tomlinson I, Avizienyte E, Roth S, Loukola A, et al. A
serine/threonine kinase gene defective in Peutz-Jeghers syndrome. Nature
1998;391:184-7.
Jenne DE, Reimann H, Nezu J-I, Friedel W, Loff S, Jeschke J, et al. Peutz-Jeghers
330
Curr Probl Surg, May 2005
547.
548.
549.
550.
551.
552.
553.
554.
555.
556.
557.
558.
559.
560.
561.
562.
564.
565.
566.
567.
568.
569.
570.
571.
572.
573.
574.
575.
576.
577.
578.
579.
580.
581.
syndrome is caused by mutations in a novel serine threonine kinase. Nat Genet
1998;18:38-43.
Gruber SB, Entius MM, Petersen GM, Laken SJ, Longo PA, Boyer R, et al.
Pathogenesis of adenocarcinoma in Peutz-Jeghers syndrome. Cancer Res
1998;58:5267-70.
Jiang CY, Esufali S, Berk T, Gallinger S, Cohen Z, Tobi M, et al. STK11/LKB1
germline mutations are not identified in most Peutz-Jeghers syndrome patients. Clin
Genet 1999;56:136-41.
Nakagawa H, Koyama K, Miyoshi Y, Ando H, Baba S, Watatani M, et al. Nine
novel germline mutations of STK11 in ten families with Peutz-Jeghers syndrome.
Hum Genet 1998;103:168-72.
Miyaki M, Iijima T, Hosono K, Ishii R, Yasuno M, Mori T, et al. Somatic mutations
of LKB1 and beta-catenin genes in gastrointestinal polyps from patients with
Peutz-Jeghers syndrome. Cancer Res 2000;60:6311-3.
Wang ZJ, Churchman M, Avizienyte E, McKeown C, Davies S, Evans DG, et al.
Germline mutations of the LKB1 (STK11) gene in Peutz-Jeghers patients. J Med
Genet 1999;36:365-8.
Westerman AM, Entius MM, Boor PP, Koole R, de Baar E, Offerhaus GJ, et al.
Novel mutations in the LKB1/STK11 gene in Dutch Peutz-Jeghers families. Hum
Mutat 1999;13:476-81.
Boardman LA, Couch FJ, Burgart LJ, Schwartz D, Berry R, McDonnell SK, et al.
Genetic heterogeneity in Peutz-Jeghers syndrome. Hum Mutat 2000;16:23-30.
Boudeau J, Kieloch A, Alessi DR, Stella A, Guanti G, Resta N. Functional analysis
of LKB1/STK11 mutants and two aberrant isoforms found in Peutz-Jeghers
Syndrome patients. Hum Mutat 2003;21:172.
Esteller M, Avizienyte E, Corn PG, Lothe RA, Baylin SB, Aaltonen LA, et al.
Epigenetic inactivation of LKB1 in primary tumors associated with the PeutzJeghers syndrome. Oncogene 2000;19:164-8.
Entius MM, Keller JJ, Westerman AM, van Rees BP, van Velthuysen ML, de Goeij
AF, et al. Molecular genetic alterations in hamartomatous polyps and carcinomas of
patients with Peutz-Jeghers syndrome. J Clin Pathol 2001;54:126-31.
Dunlop MG. Guidance on large bowel surveillance for hereditary non-polyposis
colorectal cancer, familial adenomatous polyposis, juvenile polyposis, and PeutzJeghers syndrome. Gut 2002;51(Suppl 5):V21-V27.
Ballhausen WG, Gunther K. Genetic screening for Peutz-Jeghers syndrome. Expert
Rev Mol Diagn 2003;3:471-9.
Chung DC, Rustgi AK. The hereditary nonpolyposis colorectal cancer syndrome:
genetics and clinical implications. Ann Intern Med 2003;138:560-70.
Merg A, Howe JR. Genetic conditions associated with intestinal juvenile polyps.
Am J Med Genet 2004;129C:44-55.
Brownstein MH, Wolf M, Bikowski JB. Cowden’s disease: a cutaneous marker of
breast cancer. Cancer 1978;41:2393-8.
Botma M, Russell DI, Kell RA. Cowden’s disease: a rare cause of oral papillomatosis. J Laryngol Otol 2002;116:221-3.
Sogol PB, Sugawara M, Gordon HE, Shellow WV, Hernandez F, Hershman JM.
Cowden’s disease: familial goiter and skin hamartomas. A report of three cases.
West J Med 1983;139:324-8.
Aaltonen LA, Järvinen H, Gruber SB. Peutz-Jeghers syndrome. In: Hamilton SR,
Curr Probl Surg, May 2005
331
582.
583.
584.
585.
586.
587.
588.
589.
590.
591.
592.
593.
594.
595.
596.
597.
598.
599.
332
Aaltonen LA, editors.WHO Classification: Tumours of the Digestive System.
Lyon: International Agency for Research on Cancer; 2000:74-6.
Krush AJ, Lynch HT, Magnuson C. Attitudes toward cancer in a “cancer family”:
implications for cancer detection. Am J Med Sci 1965;249:432-8.
Lynch HT, Tips RL, Krush A, Magnuson C. Family centered genetic counseling:
role of the physician and the medical genetics clinic. Nebr State Med J
1965;50:155-9.
Cristofaro G, Lynch HT, Caruso ML, Attolini A, DiMatteo G, Giorgio P, et al. New
phenotypic aspects in a family with Lynch syndrome II. Cancer 1987;60:51-8.
Lynch HT. Family information service and hereditary cancer. Cancer
2001;91:625-8.
Lynch HT, Krush AJ, Larsen AL. Heredity and multiple primary malignant
neoplasms: six cancer families. Am J Med Sci 1967;254:322-9.
Sarroca C, Ferreira WA, Quadrelli R. Cáncer colónico familiar sin poliposis:
enfoque clínico y anátomo-patológico. Perspectivas de estudio genético. Cir del
Uruguay 1977;47:515-20.
Lynch HT, Lynch PM, Harris RE. Minimal genetic findings and their cancer control
implications: a family with the cancer family syndrome. JAMA 1978;240:535-8.
Fusaro RM, Lynch HT, Pester J, Lynch PM. Torre’s syndrome as phenotypic
expression of cancer family syndrome. Arch Dermatol 1980;116:986-7.
Lynch HT, Drouhard T, Lanspa S, Smyrk T, Lynch P, Lynch J, et al. Mutation of
an mutL momologue in a Navajo family with hereditary nonpolyposis colorectal
cancer. J Natl Cancer Inst 1994;86:1417-9.
Lynch HT, Drouhard TJ, Schuelke GS, Biscone KA, Lynch JF, Danes BS.
Hereditary nonpolyposis colorectal cancer in a Navajo Indian family. Cancer Genet
Cytogenet 1985;15:209-13.
Lynch HT, Drouhard T, Vasen HF, Cavalieri J, Lynch J, Nord S, et al. Genetic
counseling in a Navajo hereditary nonpolyposis colorectal cancer kindred. Cancer
1996;77:30-5.
Lipkin M, Blattner WE, Fraumeni JF Jr, Lynch HT, Deschner E, Winawer S.
Tritiated thymidine (phi p, phi h) labeling distribution as a marker for hereditary
predisposition to colon cancer. Cancer Res 1983;43:1899-904.
Boland CR, Troncale FJ. Familial colonic cancer without antecedent polyposis.
Ann Intern Med 1984;100:700-1.
Tempero MA, Jacobs MM, Lynch HT, Graham CL, Blotcky AJ. Serum and hair
selenium levels in hereditary nonpolyposis colorectal cancer. Biol Trace Elem Res
1984;6:51-5.
Vasen HF, Mecklin JP, Khan PM, Lynch HT. The International Collaborative
Group on HNPCC. Anticancer Res 1994;14:1661-4.
Lynch HT, Marcus J, Watson P, Lynch J. Familial breast cancer, cancer family
syndromes, and predisposition to breast neoplasms. In: Bland K, Copeland EM,
editors. The Breast. Philadelphia: W.B. Saunders; 1991:262-91.
Boland CR, Chen YF, Rinderle SJ, Resau JH, Luk GD, Lynch HT, et al. Use of the
lectin from Amaranthus caudatus as a histochemical probe of proliferating colonic
epithelial cells. Cancer Res 1991;51:657-65.
Boland CR, Martin MA, Goldstein IJ. Lectin reactivities as intermediate biomarkers in premalignant colorectal epithelium. J Cell Biochem Suppl 1992;16G:103-9.
Curr Probl Surg, May 2005
600. Jass JR, Stewart SM, Stewart J, Lane MR. Hereditary non-polyposis colorectal
cancer—morphologies, genes and mutations. Mutat Res 1994;310:125-33.
601. Jass JR. Natural history of hereditary non-polyposis colorectal cancer. J Tumor
Marker Oncol 1995;10:65-71.
602. Sarroca C, Alfano N, Bendin GT, Della Valle A, Dominguez A, Quadrelli R, et al.
Hereditary nonpolyposis colorectal cancer (Lynch syndrome II) in Uruguay. Dis
Colon Rectum 2000;43:353-60; discussion 360-2.
603. Marra G, Boland CR. Hereditary nonpolyposis colorectal cancer: the syndrome, the
genes, and historical perspectives. J Natl Cancer Inst 1995;87:1114-25.
604. Hawn MT, Umar A, Carethers JM, Marra G, Kunkel TA, Boland CR, et al.
Evidence for a connection between the mismatch repair system and the G2 cell
cycle checkpoint. Cancer Res 1995;55:3721-5.
605. Boland CR. Roles of the DNA mismatch repair genes in colorectal tumorigenesis.
Int J Cancer 1996;69:47-9.
606. Akiyama Y, Sato H, Yamada T, Nagasaki H, Tsuchiya A, Abe R, et al. Germ-line
mutation of the hMSH6/GTBP gene in an atypical hereditary nonpolyposis
colorectal cancer kindred. Cancer Res 1997;57:3920-3.
607. Boland CR, Thibodeau SN, Hamilton SR, Sidransky D, Eshleman JR, Burt RW, et
al. A National Cancer Institute Workshop on Microsatellite Instability for cancer
detection and familial predisposition: development of international criteria for the
determination of microsatellite instability in colorectal cancer. Cancer Res
1998;58:5248-57.
608. Rodriguez-Bigas MA, Vasen HF, Lynch HT, Watson P, Myrhoj T, Järvinen HJ, et
al. Characteristics of small bowel carcinoma in hereditary nonpolyposis colorectal
carcinoma. International Collaborative Group on HNPCC. Cancer 1998;83:240-4.
609. Huang SC, Lavine JE, Boland PS, Newbury RO, Kolodner R, Pham TT, et al.
Germline characterization of early-aged onset of hereditary non-polyposis colorectal cancer. J Pediatr 2001;138:629-35.
610. Ribic CM, Sargent DJ, Moore MJ, Thibodeau SN, French AJ, Goldberg RM, et al.
Tumor microsatellite-instability status as a predictor of benefit from fluorouracilbased adjuvant chemotherapy for colon cancer. N Engl J Med 2003;349:247-57.
611. Chang DK, Goel A, Ricciardiello L, Lee DH, Chang CL, Carethers JM, et al. Effect
of H2O2 on cell cycle and survival in DNA mismatch repair-deficient and
-proficient cell lines. Cancer Lett 2003;195:243-51.
612. Waite KA, Eng C. Protean PTEN: form and function. Am J Hum Genet
2002;70:829-44.
Curr Probl Surg, May 2005
333