Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Cancer Res ; 61(16): 6131-6, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11507063

ABSTRACT

Epidemiological studies suggest that nonsteroidal anti-inflammatory agents decrease the risk of colorectal cancer. This is believed to be mediated, at least in part, by inhibition of cyclooxygenase (COX) activity. There are two COX isoenzymes, namely the constitutively expressed COX-1 and the inducible COX-2. COX-2 is overexpressed in adenomas and colorectal cancers, and COX-2-specific inhibitors have been shown to inhibit intestinal polyps in Apc(Delta716) mice more effectively than dual COX-1/COX-2 inhibitors such as sulindac. Various Apc knockout mice, including the multiple intestinal neoplasia (Min) mouse and the Apc(Delta716) mouse, are limited by their lack of large numbers of colonic adenomas and aberrant crypt foci, the putative precursors of large-bowel polyps and cancers. Our DNA mismatch-repair-deficient Min mouse model (Apc+/-Msh2-/-) has genetic features of both familial adenomatous polyposis and hereditary nonpolyposis colorectal cancer, and most importantly, rapidly develops numerous small- and large-bowel adenomas, as well as colonic aberrant crypt foci. The purpose of this study was to determine the effects of COX inhibitors on intestinal adenomas and colonic aberrant crypt foci in this accelerated polyposis, mismatch-repair-deficient Min mouse model, in addition to a standard Min mouse model. Weanling Apc+/-Msh2-/- and Min mice were fed diets containing no drug, sulindac, or a specific COX-2 inhibitor (MF-tricyclic). Apc+/-Msh2-/- and Min mice were sacrificed after 4 weeks and 5 months on diet, respectively. Apc+/-Msh2-/- mice treated with MF-tricyclic had significantly fewer small-bowel polyps (mean +/- SD, 178 +/- 29) compared with mice on sulindac (278 +/- 80), or control diet (341 +/- 43; P < 0.001). There was no difference in numbers of large-bowel polyps or aberrant crypt foci in mice in the three groups. MF-tricyclic was also effective in reducing both small- and large-bowel polyps in Min mice. Western analysis demonstrated COX-2 expression in both large- and small-bowel polyps from mice of both genotypes. This study demonstrates that a specific COX-2 inhibitor is effective in preventing small-bowel polyps in mismatch-repair-deficient Min mice and both small- and large-bowel polyps in standard Min mice. Therefore, specific COX-2 inhibitors may be useful as chemopreventive and therapeutic agents in humans at risk for colorectal neoplasia.


Subject(s)
Colorectal Neoplasms/drug therapy , Cyclooxygenase Inhibitors/pharmacology , DNA-Binding Proteins , Intestinal Polyps/drug therapy , Isoenzymes/antagonists & inhibitors , Precancerous Conditions/drug therapy , Proto-Oncogene Proteins/physiology , Adenoma/drug therapy , Adenoma/enzymology , Adenoma/genetics , Animals , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Crosses, Genetic , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/blood , DNA Repair/genetics , Female , Furans/pharmacology , Genes, APC/genetics , Intestinal Polyps/enzymology , Intestinal Polyps/genetics , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , MutS Homolog 2 Protein , Precancerous Conditions/enzymology , Precancerous Conditions/genetics , Prostaglandin-Endoperoxide Synthases , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Substrate Specificity , Sulindac/blood , Sulindac/pharmacology
2.
Gastroenterology ; 121(2): 275-81, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11487537

ABSTRACT

BACKGROUND & AIMS: Recent reports have suggested the mucosa of an ileal reservoir could be at risk of neoplasia. Risk factors may include the age of the pouch, chronic pouchitis, and previous colonic neoplasia. This study examined a group of such patients to determine the risk of dysplasia. METHODS: From a cohort of 1221 patients with ileal pouches, 171 patients with possible risk factors were selected. Successful contact was made with 138 patients who were invited for endoscopy and multiple biopsies. Biopsy specimens were stained with H&E and p53, scored for inflammatory changes including villous atrophy, and analyzed by flow cytometry. RESULTS: One hundred six patients took part and fell into 1 or more of the following clinical categories: chronic pouchitis (n = 34), pelvic pouch for > or =12 years (n = 42); Kock pouch for > or =14 years (n = 29), and neoplasia in colectomy specimen (n = 11). Thirty-three patients had severe villous atrophy. One patient of 106 (95% confidence interval, 0.9% +/- 1.6%) with a long-standing pouch had low-grade dysplasia that was multifocal. DNA analysis by flow cytometry showed aneuploidy in this patient and 2 others. CONCLUSIONS: These data suggest that the development of dysplasia in ileal pouches performed for ulcerative colitis is probably a rare event within 15-20 years of pouch surgery.


Subject(s)
Pouchitis/epidemiology , Pouchitis/pathology , Proctocolectomy, Restorative , Adult , Aged , Aneuploidy , Biopsy , Chronic Disease , Colitis, Ulcerative/epidemiology , Colitis, Ulcerative/pathology , Colonic Polyps/epidemiology , Colonic Polyps/pathology , Endoscopy, Gastrointestinal , Flow Cytometry , Humans , Intestinal Mucosa/chemistry , Intestinal Mucosa/pathology , Middle Aged , Morbidity , Risk Factors , Tumor Suppressor Protein p53/analysis
3.
J Natl Cancer Inst ; 93(9): 700-4, 2001 May 02.
Article in English | MEDLINE | ID: mdl-11333292

ABSTRACT

BACKGROUND: To understand the role of sporadic mutations in the tumor suppressor gene p53 (also known as TP53) in the pathogenesis of breast cancer, it is important to identify at which histologic stage such mutations first occur. We previously showed that a p53 mutation present in invasive breast cancer was found in all surrounding areas of ductal carcinoma in situ (DCIS) but not in areas of hyperplasia or normal breast epithelium. In the present investigation, we studied patients with DCIS, but without invasive breast cancer, to determine the spectrum of DCIS types that can harbor a p53 mutation. METHODS: Formalin-fixed, paraffin-embedded tissues from 94 patients with DCIS were evaluated histologically for the predominant cellular architectural pattern, degree of necrosis, and nuclear grade. Each specimen was also assigned an overall histologic grade (with the use of the Van Nuys Prognostic Index pathologic classification). Tissue specimens were stained immunohistochemically with an anti-p53 antibody. Positively stained tissue areas were analyzed for the presence of p53 mutations by single-strand conformation polymorphism and direct sequencing. All statistical tests were two-sided. RESULTS: DCIS from 10 of 94 patients were found to contain p53 missense mutations. All 10 were of a solid or a comedo histologic pattern and contained cells of nuclear grade 2 or 3 (i.e., more abnormal nuclei). The frequency of p53 missense mutations was statistically significantly different among the three overall histologic grade categories (zero [0%] of 49 with low-grade DCIS, one [4.35%] of 23 with intermediate-grade DCIS, and nine [40.9%] of 22 with high-grade DCIS; df = 2 and P<.0001). CONCLUSION: The DCIS types in patients in this series are representative of clinically detected DCIS. Our finding that p53 mutations can occur before the development of invasive breast cancer, particularly in DCIS of high histologic grade, has potentially important implications for prevention and treatment.


Subject(s)
Breast Neoplasms/genetics , Carcinoma in Situ/genetics , Carcinoma, Ductal, Breast/genetics , Genes, p53 , Mutation, Missense , Female , Humans , Immunohistochemistry , Tumor Suppressor Protein p53/analysis
4.
Dis Colon Rectum ; 44(3): 347-53, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11289279

ABSTRACT

PURPOSE: The aim of this study was to establish the prevalence of adenomatous polyps in the ileal pouch of patients with familial adenomatous polyposis. METHOD: Forty-three patients who had an ileal pouch for familial adenomatous polyposis were invited to have a careful endoscopic examination of their pouch, including dye spraying. The number of polyps was recorded, and up to ten were biopsied. In addition, four random biopsy specimens were taken from the proximal and four from the distal pouch. RESULTS: Thirty-three patients with a median age of 36 (range, 14-63) years who had a pouch (5 Kock and 28 pelvic) for a median of 7 (range, 1-19) years accepted the invitation. Twenty-one patients (64 percent) had endoscopically identified polyps, the number of polyps ranging from 1 to 100 (median, 10) and varying in size from 1 to 3 mm. Fourteen patients (42 percent) had adenomatous polyps and 4 of these also had microadenomas on random biopsies. Nine of the 14 patients with adenomas also had lymphoid polyps. Seven patients had lymphoid polyps only and two of these patients had a microadenoma on random biopsy. Four of 12 patients with no visible polyps had microadenomas in their random biopsies. The presence of adenomatous polyps (Pearson's correlation; P < 0.01) increased with the age of the pouch. In total, 20 of 33 (60 percent) patients had adenomas and or microadenomas. CONCLUSION: Adenomatous polyps occur frequently in ileal pouches. These findings are of concern, and therefore, regular surveillance seems warranted until the natural history of these adenomatous polyps is determined.


Subject(s)
Adenomatous Polyposis Coli/surgery , Adenomatous Polyps/diagnosis , Endoscopy, Gastrointestinal , Neoplasm Recurrence, Local/diagnosis , Proctocolectomy, Restorative , Adenomatous Polyposis Coli/diagnosis , Adenomatous Polyposis Coli/pathology , Adenomatous Polyps/pathology , Adolescent , Adult , Biopsy , Female , Follow-Up Studies , Humans , Intestinal Mucosa/pathology , Male , Middle Aged , Neoplasm Recurrence, Local/pathology
5.
Mod Pathol ; 14(3): 236-45, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11266532

ABSTRACT

The genetic alterations in colorectal cancer progression are determined by one of two separate and distinct underlying pathways of genomic instability. The first pathway, chromosomal instability, is characterized by allelic losses and aneuploidy. The second pathway, microsatellite instability, is characterized by an abundance of subtle DNA mutations and diploidy. Although the genes causing chromosomal instability remain unknown, microsatellite instability is caused by inactivation of a DNA mismatch repair gene (predominantly MLH1 or MSH2). Microsatellite instability is present in 15% of colorectal cancers, and is diagnosed by analysis of tumor DNA from paraffin blocks and by demonstration of loss of mismatch repair protein expression in cancers. In addition to the unique profile of genetic alterations, colorectal cancers with microsatellite instability have distinct pathologic features and improved survival. Finally, cancers from most patients with hereditary non-polyposis colorectal cancer (or Lynch syndrome) have microsatellite instability due to germline mutations in the DNA mismatch repair genes. Identification of the microsatellite instability pathway has enormous implications for the clinical investigation and management of colorectal cancer patients.


Subject(s)
Aneuploidy , Colorectal Neoplasms , DNA-Binding Proteins , Loss of Heterozygosity , Microsatellite Repeats , Adaptor Proteins, Signal Transducing , Base Pair Mismatch , Carrier Proteins , Colorectal Neoplasms/chemistry , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , DNA, Neoplasm/analysis , Humans , Intestinal Mucosa/pathology , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Neoplasm Proteins/analysis , Nuclear Proteins , Proto-Oncogene Proteins/analysis
6.
Breast Cancer Res Treat ; 65(2): 111-8, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11261826

ABSTRACT

p53 mutation is a common event in sporadic breast cancer being found in 15-50% of invasive carcinomas. The purpose of this study was to determine the earliest histologic stage at which p53 mutation could be detected with a widely used anti-p53 antibody (DO7, Novocastra) which recognizes both wild type and mutant forms. p53 expression was assessed immunohistochemically in 12 primary breast carcinomas with known p53 mutations and in all pre-malignant epithelial lesions surrounding these invasive cancers. Strong p53 nuclear staining was found in all of the tumors known to have missense mutations and none of the tumors with truncation mutations. In cases with intense staining in the invasive carcinoma, a similar quality of staining was also seen in all areas of DCIS (ductal carcinoma in situ) and was representative of missense p53 mutations. Lighter nuclear staining intensity was observed in up to 40% of cells in areas of hyperplasia and in up to 30% of normal breast lobules irrespective of the type of mutation found in the invasive carcinoma. This weak staining was not specific to mutated p53 and may indicate increased amounts of normal p53 protein. We conclude that p53 inactivation occurs prior to invasion in breast carcinogenesis, with mutations being uniformly identified in DCIS associated with p53-mutated invasive carcinomas. In contrast, there is no evidence that epithelial hyperplasia or epithelial cells of the terminal duct lobular unit harbor the same mutations as their associated invasive carcinoma.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Intraductal, Noninfiltrating/genetics , Cell Transformation, Neoplastic/genetics , Tumor Suppressor Protein p53/genetics , Adult , Aged , Female , Humans , Immunohistochemistry , Middle Aged , Mutation , Tumor Suppressor Protein p53/metabolism
7.
Nature ; 406(6798): 897-902, 2000 Aug 24.
Article in English | MEDLINE | ID: mdl-10972292

ABSTRACT

Phosphoinositide-3-OH kinases (PI(3)Ks) constitute a family of evolutionarily conserved lipid kinases that regulate a vast array of fundamental cellular responses, including proliferation, transformation, differentiation and protection from apoptosis. PI(3)K-mediated activation of the cell survival kinase PKB/Akt, and negative regulation of PI(3)K signalling by the tumour suppressor PTEN (refs 3, 4) are key regulatory events in tumorigenesis. Thus, a model has arisen that PI(3)Ks promote development of cancers. Here we report that genetic inactivation of the p110gamma catalytic subunit of PI(3)Kgamma (ref. 8) leads to development of invasive colorectal adenocarcinomas in mice. In humans, p110gamma protein expression is lost in primary colorectal adenocarcinomas from patients and in colon cancer cell lines. Overexpression of wild-type or kinase-dead p110gamma in human colon cancer cells with mutations of the tumour suppressors APC and p53, or the oncogenes beta-catenin and Ki-ras, suppressed tumorigenesis. Thus, loss of p110gamma in mice leads to spontaneous, malignant epithelial tumours in the colorectum and p110gamma can block the growth of human colon cancer cells.


Subject(s)
Colorectal Neoplasms/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Animals , Carcinoma/enzymology , Carcinoma/genetics , Catalytic Domain/genetics , Cell Cycle Proteins/biosynthesis , Chromosome Mapping , Chromosomes, Human, Pair 7 , Colorectal Neoplasms/genetics , Humans , Longevity , Mice , Mice, Nude , Molecular Sequence Data , Phosphatidylinositol 3-Kinases/genetics , Protein Biosynthesis , Tumor Cells, Cultured
8.
Am J Pathol ; 156(5): 1641-51, 2000 May.
Article in English | MEDLINE | ID: mdl-10793075

ABSTRACT

Medullary carcinomas of the pancreas are a recently described, histologically distinct subset of poorly differentiated adenocarcinomas that may have a unique pathogenesis and clinical course. To further evaluate these neoplasms, we studied genetic, pathological, and clinical features of 13 newly identified medullary carcinomas of the pancreas. Nine (69%) of these had wild-type K-ras genes, and one had microsatellite instability (MSI). This MSI medullary carcinoma, along with three previously reported MSI medullary carcinomas, were examined immunohistochemically for Mlh1 and Msh2 expression, and all four expressed Msh2 but did not express Mlh1. In contrast, all of the medullary carcinomas without MSI expressed both Msh2 and Mlh1. Remarkably, the MSI medullary carcinoma of the pancreas in the present series arose in a patient with a synchronous but histologically distinct cecal carcinoma that also had MSI and did not express Mlh1. The synchronous occurrence of two MSI carcinomas suggests an inherited basis for the development of these carcinomas. Indeed, the medullary phenotype, irrespective of MSI, was highly associated with a family history of cancer in first-degree relatives (P < 0.001). Finally, one medullary carcinoma with lymphoepithelioma-like features contained Epstein-Barr virus-encoded RNA-1 by in situ hybridization. Therefore, because of medullary carcinoma's special genetic, immunohistochemical, and clinical features, recognition of the medullary variant of pancreatic adenocarcinoma is important. Only by classifying medullary carcinoma as special subset of adenocarcinoma can we hope to further elucidate its unique pathogenesis.


Subject(s)
Carcinoma, Medullary/pathology , DNA-Binding Proteins , Pancreatic Neoplasms/pathology , Adaptor Proteins, Signal Transducing , Adult , Aged , Aged, 80 and over , Carcinoma, Medullary/genetics , Carcinoma, Medullary/metabolism , Carrier Proteins , Epstein-Barr Virus Infections/virology , Family Health , Female , Genes, ras/genetics , Herpesvirus 4, Human/genetics , Humans , Immunohistochemistry , In Situ Hybridization , Male , Microsatellite Repeats/genetics , Middle Aged , Multivariate Analysis , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Mutation , Neoplasm Proteins/analysis , Nuclear Proteins , Pancreas/chemistry , Pancreas/pathology , Pancreas/virology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Phenotype , Proto-Oncogene Proteins/analysis , RNA, Viral/genetics , Survival Analysis
9.
Genes Chromosomes Cancer ; 27(4): 358-61, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10719365

ABSTRACT

Germline mutations of the CDKN2A tumor suppressor gene have been identified in melanoma kindreds linked to 9p21, and pancreatic adenocarcinoma is the second most common malignancy in some of these families. We hypothesized that unselected patients with both primary cancers, i.e., pancreatic cancer and malignant melanoma, have a genetic predisposition to tumor development, and that this susceptibility may be due to germline CDKN2A mutations. Fourteen patients, with both pathologically verified pancreatic adenocarcinoma and melanoma, were assessed for germline CDKN2A mutations by polymerase chain reaction amplification and sequencing of six overlapping fragments encompassing exons 1alpha and 2. A yeast two-hybrid assay was used to assess the functional consequences of CDKN2A variants. Germline CDKN2A mutations were identified in 2/14 patients: I49S, a novel substitution in exon 1alpha, and M53I, a previously reported missense mutation in exon 2. Both variants lead to compromised CDKN2A function. We conclude that the occurrence of both pancreatic cancer and melanoma, in the same patient, signals an inherited susceptibility to cancer, and that this predisposition is, in some cases, due to germline CDKN2A mutations. This finding has important implications not only for the proband, but also for other family members.


Subject(s)
Adenocarcinoma/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Germ-Line Mutation/genetics , Melanoma/genetics , Neoplasms, Multiple Primary/genetics , Pancreatic Neoplasms/genetics , Adult , Aged , DNA Mutational Analysis , DNA, Neoplasm/analysis , Female , Genetic Carrier Screening , Genetic Predisposition to Disease , Humans , Male , Middle Aged
10.
Cancer Res ; 60(2): 409-16, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10667595

ABSTRACT

Susceptibility to pancreatic adenocarcinoma appears to be linked to germ-line mutations in genes causing various familial cancer syndromes. The objectives of this study were to estimate the proportion of unselected pancreatic cancer patients belonging to hereditary cancer syndrome families and to determine the frequency ofp16, BRCA1, BRCA2, hMSH2, and hMLH1 germ-line mutations in patients with a personal or family history of cancer. The study population consisted of 102 patients with histologically verified pancreatic adenocarcinoma, unselected for age, sex, family history, or ethnic origin. Patients completed a family history questionnaire and provided blood for mutation analysis. Three-generation pedigrees were constructed and classified as high risk/familial, intermediate risk/ familial, intermediate risk/nonfamilial, or low risk according to defined criteria. High- and intermediate-risk cases were analyzed for germ-line mutations using a combination of methods. Thirty-eight of 102 (37%) patients were characterized as high or intermediate risk, and the remainder were classified as low risk. Germ-line mutations were identified in five (13%) of these cases [one in p16 (I49S); one in BRCA1 (5382 insC); and three in BRCA2 (6174delT)]. The BRCA1 and BRCA2 mutations were identified in Ashkenazi Jewish patients. Four of the mutation carriers had strong family histories of the syndromes associated with the mutated genes. No mutations were identified in patients in whom the sole risk factor was a family history of pancreatic cancer, and only one mutation was found among patients with early-onset disease. We conclude that known causes of genetic predisposition are an important risk factor in a small proportion of pancreatic cancer patients, especially if associated with a strong family history of syndromes associated with the disease. The lack of detectable germ-line mutations in most high- and intermediate-risk cases suggests that there are probably additional, as yet unidentified genes predisposing to this disease.


Subject(s)
Adenocarcinoma/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA-Binding Proteins , Genes, BRCA1 , Genetic Predisposition to Disease , Germ-Line Mutation , Mutation , Neoplasm Proteins/genetics , Pancreatic Neoplasms/genetics , Transcription Factors/genetics , Adaptor Proteins, Signal Transducing , BRCA1 Protein/genetics , BRCA2 Protein , Carrier Proteins , Europe/ethnology , Exons , Family , Female , Genetic Linkage , Genetic Markers , Humans , Jews/genetics , Male , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Nuclear Proteins , Ontario , Pedigree , Proto-Oncogene Proteins/genetics , Risk Assessment , Sequence Deletion
11.
N Engl J Med ; 342(2): 69-77, 2000 Jan 13.
Article in English | MEDLINE | ID: mdl-10631274

ABSTRACT

BACKGROUND: Colorectal cancer can arise through two distinct mutational pathways: microsatellite instability or chromosomal instability. We tested the hypothesis that colorectal cancers arising from the microsatellite-instability pathway have distinctive clinical attributes that affect clinical outcome. METHODS: We tested specimens of colorectal cancer from a population-based series of 607 patients (50 years of age or younger at diagnosis) for microsatellite instability. We compared the clinical features and survival of patients who had colorectal cancer characterized by high-frequency microsatellite instability with these characteristics in patients who had colorectal cancers with microsatellite stability. RESULT: We found high-frequency microsatellite instability in 17 percent of the colorectal cancers in 607 patients, and in a multivariate analysis, microsatellite instability was associated with a significant survival advantage independently of all standard prognostic factors, including tumor stage (hazard ratio, 0.42; 95 percent confidence interval, 0.27 to 0.67; P< 0.001). Furthermore, regardless of the depth of tumor invasion, colorectal cancers with high-frequency microsatellite instability had a decreased likelihood of metastasizing to regional lymph nodes (odds ratio, 0.33; 95 percent confidence interval, 0.21 to 0.53; P< 0.001) or distant organs (odds ratio, 0.49; 95 percent confidence interval, 0.27 to 0.89; P=0.02). CONCLUSION: High-frequency microsatellite instability in colorectal cancer is independently predictive of a relatively favorable outcome and, in addition, reduces the likelihood of metastases.


Subject(s)
Colorectal Neoplasms/genetics , Microsatellite Repeats/genetics , Adult , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Female , Humans , Male , Middle Aged , Multivariate Analysis , Mutation , Prognosis , Survival Analysis
12.
Clin Genet ; 56(2): 136-41, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10517250

ABSTRACT

Germline mutations of the STK11 gene mapped to chromosome 19p13.3 are responsible for Peutz Jeghers syndrome (PJS), a dominant disorder associated with characteristic gastrointestinal hamartomatous polyps and a predisposition to various cancers. We conducted a detailed investigation of germline STK11 alterations by protein truncation test and genomic DNA sequence analysis in ten unrelated PJS families. We identified a novel truncating deletion spanning STK11 exons 2-7 in a single patient and several known polymorphisms. Loss of heterozygosity studies in PJS polyps of four of these patients identified an allelic deletion of D19S886 in another patient. Our results suggest that STK11 mutations account for only a proportion of PJS cases.


Subject(s)
Germ-Line Mutation , Peutz-Jeghers Syndrome/genetics , Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinase Kinases , Adolescent , Adult , Child , Chromosomes, Human, Pair 19/genetics , Female , Gene Deletion , Genetic Markers , Humans , Introns , Loss of Heterozygosity , Male , Middle Aged , Pedigree , Phenotype , Polymorphism, Genetic , Sequence Analysis, DNA
13.
Am J Surg Pathol ; 23(10): 1248-55, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10524526

ABSTRACT

Inactivation of deoxyribonucleic acid (DNA) mismatch repair genes, most commonly human mutL homologue 1 (hMLH1) or human mutS homologue 2 (hMSH2), is a recently described alternate pathway in cancer development and progression. The resulting genetic instability is characterized by widespread somatic mutations in tumor DNA, and is termed high-frequency microsatellite instability (MSI-H). Although described in a variety of tumors, mismatch repair deficiency has been studied predominantly in colorectal carcinoma. Most MSI-H colorectal carcinomas are sporadic, but some occur in patients with hereditary nonpolyposis colorectal cancer (HNPCC), and are associated with germline mutations in mismatch repair genes. Until now, the identification of MSI-H cancers has required molecular testing. To evaluate the role of immunohistochemistry as a new screening tool for mismatch repair-deficient neoplasms, the authors studied the expression of hMLH1 and hMSH2, using commercially available monoclonal antibodies, in 72 formalin-fixed, paraffin-embedded tumors that had been tested previously for microsatellite instability. They compared immunohistochemical patterns of 38 MSI-H neoplasms, including 16 cases from HNPCC patients with known germline mutations in hMLH1 or hMSH2, with 34 neoplasms that did not show microsatellite instability. Thirty-seven of 38 MSI-H neoplasms were predicted to have a mismatch repair gene defect, as demonstrated by the absence of hMLH1 and/or hMSH2 expression. This included correspondence with all 16 cases with germline mutations. All 34 microsatellite-stable cancers had intact staining with both antibodies. These findings clearly demonstrate that immunohistochemistry can discriminate accurately between MSI-H and microsatellite-stable tumors, providing a practical new technique with important clinical and research applications.


Subject(s)
Adenocarcinoma/genetics , Base Pair Mismatch/genetics , Colorectal Neoplasms/genetics , DNA Repair , DNA-Binding Proteins , Neoplasm Proteins/analysis , Proto-Oncogene Proteins/analysis , Adaptor Proteins, Signal Transducing , Adenocarcinoma/chemistry , Adenocarcinoma/pathology , Adult , Carrier Proteins , Colorectal Neoplasms/chemistry , Colorectal Neoplasms/pathology , DNA, Neoplasm/analysis , Genes, DCC/genetics , Genetic Predisposition to Disease/genetics , Humans , Immunoenzyme Techniques , Microsatellite Repeats/genetics , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Nuclear Proteins , Polymerase Chain Reaction
15.
Cancer Res ; 59(14): 3346-51, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10416591

ABSTRACT

Some colorectal tumors with wild-type adenomatous polyposis coli gene have activating mutations in beta-catenin (encoded by CTNNB1) that result in decreased phosphorylation by GSK-3beta and increased signaling through the Tcf/Lef transcription factors. To investigate the relationship between CTNNB1 mutations and underlying pathways of genomic instability, we examined 80 colorectal cancers stratified by the presence or absence of microsatellite instability (MSI). CTNNB1 mutations were identified in 13 (25%) of 53 cancers with high frequency MSI (MSI-H), including 12 point mutations at exon 3 phosphorylation sites (codons 41 and 45) and one deletion of the entire exon 3 degradation box. No CTNNB1 mutations were identified in 27 microsatellite stable or low frequency MSI (MSI-L) colorectal cancers (P < 0.01). In contrast, CTNNB1 mutations were identified in 3 of 9 (33%) MSI-H and 10 of 20 (50%) MSS/MSI-L endometrial carcinomas, suggesting a more generalized involvement in these tumors. Only six (46%) of the endometrial carcinoma CTNNB1 mutations occurred at residues directly phosphorylated by GSK-3beta, and only one of these was at either codon 41 or 45. All point mutations in MSI-H cancers were transitions, whereas 64% of those in MSS/MSI-L cancers were transversions (P < 0.01). The differences in the mutation profiles suggest that there may be molecular fingerprints of CTNNB1 mutations, determined by biological factors related to both tumor type and underlying pathways of genomic instability.


Subject(s)
Adenocarcinoma/genetics , Amino Acid Substitution , Cell Transformation, Neoplastic/genetics , Colonic Neoplasms/genetics , Cytoskeletal Proteins/genetics , Endometrial Neoplasms/genetics , Microsatellite Repeats , Point Mutation , Trans-Activators , Adenoma/genetics , Adenomatous Polyposis Coli/genetics , Cell Differentiation , Codon/genetics , DNA Mutational Analysis , DNA, Neoplasm/genetics , Exons/genetics , Female , Genes, APC , Humans , Male , Organ Specificity , Phosphorylation , Protein Processing, Post-Translational , Signal Transduction/genetics , beta Catenin
16.
Hum Mol Genet ; 8(5): 823-9, 1999 May.
Article in English | MEDLINE | ID: mdl-10196371

ABSTRACT

Hereditary non-polyposis colorectal cancer (HNPCC) is a dominantly inherited cancer syndrome caused by germline defects of mismatch repair (MMR) genes. Endometrial cancer is the most common extracolonic neoplasm in HNPCC and is the primary clinical manifestation of the syndrome in some families. The cumulative incidence of endometrial cancer among HNPCC mutation carriers is high, estimated to be from 22 to 43%. We hypothesized that women with double primary cancers of the colorectum and endometrium are likely to be members of HNPCC families. In order to determine how frequently HNPCC manifests in the context of double primary cancers, we examined alterations of two MMR genes, hMSH2 and hMLH1, in 40 unrelated women affected with double primary cancers. These cases were identified using hospital-based and population-based cancer registries in Ontario, Canada. MMR gene mutations were screened by single-strand conformation polymorphism analysis and confirmed by direct sequencing. Eighteen percent (seven of 40) were found to harbor mutations of one of the two MMR genes. Analysis of colorectal and/or endometrial tumors of mutation-negative probands found microsatellite instability in seven of 20 cases. Six of seven mutation-positive probands had strong family histories suggestive of HNPCC. First degree relatives of mutation-positive probands had a very high relative risk (RR) of colorectal cancer (RR = 8.1, CI 3. 5-15.9) and endometrial cancer (RR = 23.8, CI 6.4-61.0). The relative risk of mutation-negative cases was 2.8 (CI 1.7-4.5) for colorectal cancer and 5.4 (CI 2.0-11.7) for endometrial cancer. We recommend that all double primary patients with cancers at these sites should have a genetic evaluation, including molecular analysis for HNPCC where appropriate.


Subject(s)
Colorectal Neoplasms/genetics , DNA-Binding Proteins , Endometrial Neoplasms/genetics , Neoplasm Proteins/genetics , Neoplasms, Multiple Primary/genetics , Proto-Oncogene Proteins/genetics , Adaptor Proteins, Signal Transducing , Adult , Base Pair Mismatch/genetics , Carrier Proteins , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Repair/genetics , Female , Genetic Predisposition to Disease , Humans , Male , Microsatellite Repeats , Middle Aged , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Mutation , Nuclear Proteins , Pedigree
17.
Hum Genet ; 104(2): 167-76, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10190329

ABSTRACT

Recent characterization of the molecular genetic basis of hereditary nonpolyposis colorectal cancer provides an important opportunity for identification of individuals and their families with germline mutations in mismatch repair genes. Cancer family history criteria that accurately define hereditary colorectal cancer are necessary for cost-effective testing for germline mutations in mismatch repair genes. The present report describes the results of analysis of 33 colorectal cancer cases/families that satisfy our modified family history criteria (Mount Sinai criteria) for colorectal cancer. Fourteen of these families met the more stringent Amsterdam criteria. Germline MSH2 and MLH1 mutations were identified by the reverse transcription-polymerase chain reaction and the protein truncation test, and confirmed by sequencing. Microsatellite instability analysis was performed on available tumors from affected patients. MSH2 or MLH1 mutations were detected in 8 of 14 Amsterdam criteria families and in 5 of the remaining 19 cases/families that only satisfied the Mount Sinai criteria. Three of the latter families had features of the Muir-Torre syndrome. A high level of microsatellite instability (MSI-H) was detected in almost all (16/18) colorectal cancers from individuals with MSH2 and MLH1 mutations, and infrequently (1/21) in colorectal cancer specimens from cases without detectable mutations. Families with germline MSH2 and MLH1 mutations tended to have individuals affected at younger ages and with multiple tumors. The Amsterdam criteria are useful, but not sufficient, for detecting hereditary colorectal cancer families with germline MSH2 and MLH1 mutations, since a proportion of cases and families with mutations in mismatch repair genes will be missed. Further development of cancer family history criteria are needed, using unbiased prospectively collected cases, to define more accurately those who will benefit from MSH2 and MLH1 mutation analysis.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA, Neoplasm , DNA-Binding Proteins , Germ-Line Mutation , Neoplasm Proteins/genetics , Proto-Oncogene Proteins/genetics , Adaptor Proteins, Signal Transducing , Base Pair Mismatch , Carrier Proteins , DNA Repair , Female , Humans , Male , Microsatellite Repeats , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Nuclear Proteins , Pedigree
18.
Am J Hum Genet ; 64(2): 378-84, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9973276

ABSTRACT

Germ-line and somatic truncating mutations of the APC gene are thought to initiate colorectal tumor formation in familial adenomatous polyposis syndrome and sporadic colorectal carcinogenesis, respectively. Recently, an isoleucine-->lysine polymorphism at codon 1307 (I1307K) of the APC gene has been identified in 6%-7% of the Ashkenazi Jewish population. To assess the risk of this common APC allelic variant in colorectal carcinogenesis, we have analyzed a large cohort of unselected Ashkenazi Jewish subjects with adenomatous polyps and.or colorectal cancer, for the APC I1307K polymorphism. The APC I1307K allele was identified in 48 (10.1%) of 476 patients. Compared with the frequency in two separate population control groups, the APC I1307K allele is associated with an estimated relative risk of 1.5-1.7 for colorectal neoplasia (both P=.01). Furthermore, compared with noncarriers, APC I1307K carriers had increased numbers of adenomas and colorectal cancers per patient (P=.03), as well as a younger age at diagnosis. We conclude that the APC I1307K variant leads to increased adenoma formation and directly contributes to 3%-4% of all Ashkenazi Jewish colorectal cancer. The estimated relative risk for carriers may justify specific clinical screening for the 360,000 Americans expected to harbor this allele, and genetic testing in the setting of long-term-outcome studies may impact significantly on colorectal cancer prevention in this population.


Subject(s)
Adenomatous Polyposis Coli/genetics , Colonic Neoplasms/genetics , Cytoskeletal Proteins/genetics , Polymorphism, Genetic , Adenomatous Polyposis Coli Protein , Aged , Female , Genetic Diseases, Inborn , Humans , Male , Pancreatic Neoplasms/genetics , Risk Factors
19.
Int J Cancer ; 78(5): 600-5, 1998 Nov 23.
Article in English | MEDLINE | ID: mdl-9808529

ABSTRACT

Loss of heterozygosity (LOH) studies reported thus far suggest that tumor suppressor loci on chromosome 5q are important in esophageal cancer (EC) while little is known about the involvement of chromosome 5p. To investigate the potential existence of tumor suppressor gene(s) on chromosome 5 contributing to the development of EC, we performed LOH studies using a total of 24 polymorphic markers spanning the entire chromosome 5. Seventy primary esophageal cancers were microdissected and allelic deletions were detected by polymerase chain reaction (PCR)-single strand conformation polymorphism or by microsatellite analysis. LOH was observed in at least 1 of the loci in 47 of 70 (67%) esophageal tumors. Initially, 40 tumors [24 squamous cell carcinomas (SCC) and 16 adenocarcinomas (ADC)], each with matched histologically normal esophageal mucosa, were analyzed at 15 marker loci on 5p and 5q. A novel locus, D5S667 on 5p15.2, exhibited the highest frequency of LOH (44%) in these tumors along with another previously reported region of frequent deletion, irf-1 (5q31.1). In a series of 30 additional EC tumors (11 SCC and 19 ADC), a detailed LOH analysis of chromosome 5p15.2 region was conducted using 10 additional polymorphic markers, which mapped the frequently deleted region within 1 cM. Overall, LOH at the D5S667 locus was observed more frequently in SCC than in ADC (62% vs. 23%, p = 0.01). This significant rate of LOH of a distinct region of chromosome 5p implicates the existence of a putative tumor suppressor gene locus involved in EC.


Subject(s)
Chromosomes, Human, Pair 5 , Esophageal Neoplasms/genetics , Loss of Heterozygosity , Adenocarcinoma/genetics , Carcinoma, Squamous Cell/genetics , Chromosome Mapping , Humans
SELECTION OF CITATIONS
SEARCH DETAIL