27 results on '"Redston M"'
Search Results
2. Giant Filiform Polyposis of the Colon in a 6-Year Old Male With Ulcerative Colitis.
- Author
-
Slack JC, Redston M, Goldsmith JD, Bousvaros B, and Levy Zitomersky N
- Subjects
- Child, Humans, Male, Colitis, Ulcerative complications, Colonic Polyps complications, Colonic Polyps diagnosis, Colonic Polyps surgery, Colorectal Neoplasms
- Abstract
Competing Interests: The authors report no conflicts of interest.
- Published
- 2022
- Full Text
- View/download PDF
3. Five-year efficacy and safety analysis of the Adenoma Prevention with Celecoxib Trial.
- Author
-
Bertagnolli MM, Eagle CJ, Zauber AG, Redston M, Breazna A, Kim K, Tang J, Rosenstein RB, Umar A, Bagheri D, Collins NT, Burn J, Chung DC, Dewar T, Foley TR, Hoffman N, Macrae F, Pruitt RE, Saltzman JR, Salzberg B, Sylwestrowicz T, and Hawk ET
- Subjects
- Adult, Aged, Aged, 80 and over, Celecoxib, Colonoscopy, Cyclooxygenase 2 Inhibitors administration & dosage, Dose-Response Relationship, Drug, Female, Follow-Up Studies, Humans, Male, Middle Aged, Neoplasm Recurrence, Local prevention & control, Pyrazoles administration & dosage, Sulfonamides administration & dosage, Adenoma prevention & control, Cardiovascular Diseases chemically induced, Colorectal Neoplasms prevention & control, Cyclooxygenase 2 Inhibitors adverse effects, Pyrazoles adverse effects, Sulfonamides adverse effects
- Abstract
The Adenoma Prevention with Celecoxib Trial examined the efficacy and safety of the cyclooxygenase (Cox)-2 inhibitor, celecoxib, for sporadic colorectal adenoma prevention in patients at high risk for colorectal cancer. The trial randomized 2,035 subjects to receive either placebo, celecoxib 200 mg twice daily, or celecoxib 400 mg twice daily. The primary study safety and efficacy analyses involved 3 years of treatment. The results showed significant antitumor effect but also indicated increased cardiovascular adverse events in patients treated with celecoxib compared with placebo. A total of 933 patients participated in an extension of the Adenoma Prevention with Celecoxib Trial, with a planned total treatment and surveillance duration of 5 years. Study medication was stopped early, resulting in a median treatment duration of 3.1 years for those with a year 5 colonoscopy. Patients treated on the placebo arm had a cumulative adenoma incidence of 68.4% over 5 years of observation. This figure was 59.0% (P < 0.0001) for those receiving low-dose celecoxib, and 60.1% (P < 0.0001) for those receiving high-dose celecoxib. The cumulative incidence of advanced adenomas over 5 years was 21.3% of those taking placebo, 12.5% (P < 0.0001) of those taking low dose celecoxib and 15.8% (P < 0.0001) of those taking high-dose celecoxib. Investigator reported treatment emergent adverse events were similar across all treatment groups for categories including renal and hypertensive events and gastrointestinal ulceration and hemorrhage events. For a category composed of cardiovascular and thrombotic events, the risk relative to placebo was 1.6 (95% confidence interval, 1.0, 2.5) for those using 200 mg twice daily celecoxib and 1.9 (95% confidence interval, 1.2, 3.1) for those using 400 mg twice daily celecoxib. Secondary analysis showed an interaction between a baseline history of atherosclerotic heart disease and study drug use with respect to cardiovascular and thrombotic adverse events (P = 0.004). These results confirm the inhibitory effect of celecoxib on colorectal adenoma formation, and provide additional safety data indicating an elevated risk for cardiovascular and thrombotic adverse events, particularly for patients with preexisting atherosclerotic heart disease.
- Published
- 2009
- Full Text
- View/download PDF
4. Missense polymorphisms in the adenomatous polyposis coli gene and colorectal cancer risk.
- Author
-
Cleary SP, Kim H, Croitoru ME, Redston M, Knight JA, Gallinger S, and Gryfe R
- Subjects
- Adenomatous Polyposis Coli epidemiology, Adult, Aged, Alleles, Analysis of Variance, Case-Control Studies, Colorectal Neoplasms epidemiology, DNA Mutational Analysis, Female, Humans, Incidence, Male, Middle Aged, Ontario epidemiology, Open Reading Frames, Polymerase Chain Reaction, Risk Factors, Statistics, Nonparametric, Surveys and Questionnaires, Adenomatous Polyposis Coli genetics, Colorectal Neoplasms genetics, Genes, APC, Mutation, Missense, Polymorphism, Genetic
- Abstract
Purpose: Whereas truncating germline mutations of the adenomatous polyposis coli (APC) gene give rise to familial adenomatous polyposis, missense polymorphisms of APC may confer a weaker risk for colorectal cancer., Methods: We sequenced the entire open reading frame of the APC gene and tested for two common MYH mutations in a population-based series of patients with colorectal cancer and 5 to 99 adenomas. Missense adenomatous polyposis coli alterations identified in this colorectal cancer multiple-polyp population were analyzed in a population-based series of patients with colorectal cancer and healthy control subjects., Results: Germline APC or mutY human homologue (MYH) alterations were identified in 16 of 39 colorectal cancer-multiple polyp patients. Four missense APC gene alterations (S130G, E1317Q, D1822V, G2502S) were observed in 13 individuals and 3 additional patients carried presumed pathogenic (APC Y94X, biallelic MYH Y165C and heterozygous MYH G382D) mutations. When independently assessed in 971 patients with colorectal cancer and 954 healthy control subjects, none of the identified missense APC alterations conferred a significantly increased risk for colorectal cancer, odds ratio (95 percent confidence intervals): S130G = 3.1 (0.29-32.25), E1317Q = 1.08 (0.59-2.74), G2502S = 1 (0.65-1.63), D1822V (heterozygous) = 0.79 (0.64-0.98), D1822V (homozygous) = 0.82 (0.63-1.27)., Conclusions: Germline missense APC alterations observed in 33 percent of patients with multiple colorectal neoplasms seemed to play a limited role in colorectal cancer risk when independently assessed by a population-based, case-control analysis.
- Published
- 2008
- Full Text
- View/download PDF
5. Common and distinct genomic events in sporadic colorectal cancer and diverse cancer types.
- Author
-
Martin ES, Tonon G, Sinha R, Xiao Y, Feng B, Kimmelman AC, Protopopov A, Ivanova E, Brennan C, Montgomery K, Kucherlapati R, Bailey G, Redston M, Chin L, and DePinho RA
- Subjects
- Cell Line, Tumor, Chromosome Aberrations, Gene Expression Profiling, Genes, Neoplasm, Genome, Humans, Immunohistochemistry methods, Models, Genetic, Mutation, Nucleic Acid Hybridization, Sequence Analysis, DNA, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Gene Expression Regulation, Neoplastic
- Abstract
Colorectal cancer (CRC) is a major cause of cancer morbidity and mortality, and elucidation of its underlying genetics has advanced diagnostic screening, early detection, and treatment. Because CRC genomes are characterized by numerous non-random chromosomal structural alterations, we sought to delimit regions of recurrent amplifications and deletions in a collection of 42 primary specimens and 37 tumor cell lines derived from chromosomal instability neoplasia and microsatellite instability neoplasia CRC subtypes and to compare the pattern of genomic aberrations in CRC with those in other cancers. Application of oligomer-based array-comparative genome hybridization and custom analytic tools identified 50 minimal common regions (MCRs) of copy number alterations, 28 amplifications, and 22 deletions. Fifteen were highly recurrent and focal (<12 genes) MCRs, five of them harboring known CRC genes including EGFR and MYC with the remaining 10 containing a total of 65 resident genes with established links to cancer. Furthermore, comparisons of these delimited genomic profiles revealed that 22 of the 50 CRC MCRs are also present in lung cancer, glioblastoma, and/or multiple myeloma. Among 22 shared MCRs, nine do not contain genes previously shown genetically altered in cancer, whereas the remaining 13 harbor 35 known cancer genes, of which only 14 have been linked to CRC pathogenesis. Together, these observations point to the existence of many yet-to-be discovered cancer genes driving CRC development, as well as other human cancers, and show the utility of high-resolution copy number analysis in the identification of genetic events common and specific to the development of various tumor types.
- Published
- 2007
- Full Text
- View/download PDF
6. Filiform serrated adenomas: a clinicopathologic and immunophenotypic study of 18 cases.
- Author
-
Yantiss RK, Oh KY, Chen YT, Redston M, and Odze RD
- Subjects
- Adenomatous Polyps chemistry, Adenomatous Polyps genetics, Adenomatous Polyps surgery, Adult, Aged, Aged, 80 and over, Biomarkers, Tumor analysis, Colorectal Neoplasms chemistry, Colorectal Neoplasms genetics, Colorectal Neoplasms surgery, DNA Mutational Analysis, DNA, Neoplasm analysis, Epithelial Cells chemistry, Epithelial Cells pathology, Female, Genes, p53 genetics, Genes, ras genetics, Genetic Markers, Humans, Immunoenzyme Techniques, Male, Microsatellite Instability, Middle Aged, Proto-Oncogene Proteins B-raf genetics, Proto-Oncogene Proteins B-raf metabolism, Adenomatous Polyps pathology, Colorectal Neoplasms pathology
- Abstract
In this study, we describe a previously uncharacterized type of adenomatous polyp of the colorectum that shows prominent, thin, elongated projections of neoplastic epithelium with a serrated contour, which we have termed "filiform serrated adenoma" (SA). Routinely processed polypectomy specimens from 18 patients with filiform SA and 23 controls with traditional (nonfiliform) SA were evaluated for their clinical and pathologic features, and immunohistochemically stained for a variety of markers (O-methylguanine methyltransferase, MLH1, MSH2, CDX2, nuclear beta-catenin, p53, and Ki-67) designed to evaluate their molecular and proliferative characteristics. DNA was extracted from the paraffin-embedded materials, amplified by polymerase chain reaction, and analyzed for microsatellite instability, BRAF, K-ras, and p53 mutational status. Five cases contained sufficient non-neoplastic tissue for dissection and DNA extraction, allowing analysis of loss of heterozygosity. The study group consisted of 7 males and 11 females of mean age 64 years (range: 42 to 89 y). All 18 filiform SAs were located in the left colon, including 15 (83%) that occurred in the rectum, compared with 43% of the control group (P=0.03). Filiform SAs were also larger (1.6 cm) than SAs (mean: 1.2 cm, P=0.02), but no other clinical differences were noted. Most (56%) filiform SAs contained marked stromal edema and tall nonmucinous cells with abundant eosinophilic cytoplasm (61%). High-grade dysplasia was present in 4/18 (22%) cases. Four (22%) filiform SAs also contained nonserrated adenomatous elements with a villous (3 cases) or tubular (1 case) growth pattern. Two (11%) cases contained adjacent areas of sessile SAs and 4 (22%) had hyperplastic areas. None of the polyps in the control group showed stromal edema, high-grade dysplasia, or mixed elements. Polyps in both groups demonstrated comparable staining patterns for O-methylguanine methyltransferase, MLH-1, MSH-2, CDX2, beta-catenin, and Ki-67, and none showed increased nuclear p53 expression. Low-frequency microsatellite instability was present in 5/12 (42%) filiform SAs, 7/12 (58%) were microsatellite stable. Mitogen-activated protein kinase pathway abnormalities were present in 71% of the cases [7/14 (50%) with BRAF and 3/14 (21%) with K-ras mutations]. Four cases showed silent p53 mutations upon direct sequencing and 4 revealed loss of heterozygosity at the loci evaluated, including 1 at D5S346 [adenomatous polyposis coli (APC) gene], 1 at D17S250 (p53 gene), and 2 at MYCL (chromosome 1p34). We conclude that filiform SA potentially represents an unusual variant of SA with a predilection for the left colon, particularly the rectum.
- Published
- 2007
- Full Text
- View/download PDF
7. Celecoxib for the prevention of sporadic colorectal adenomas.
- Author
-
Bertagnolli MM, Eagle CJ, Zauber AG, Redston M, Solomon SD, Kim K, Tang J, Rosenstein RB, Wittes J, Corle D, Hess TM, Woloj GM, Boisserie F, Anderson WF, Viner JL, Bagheri D, Burn J, Chung DC, Dewar T, Foley TR, Hoffman N, Macrae F, Pruitt RE, Saltzman JR, Salzberg B, Sylwestrowicz T, Gordon GB, and Hawk ET
- Subjects
- Adenoma drug therapy, Adult, Aged, Aged, 80 and over, Anti-Inflammatory Agents, Non-Steroidal administration & dosage, Aspirin administration & dosage, Cardiovascular Diseases chemically induced, Cardiovascular Diseases mortality, Celecoxib, Colorectal Neoplasms drug therapy, Cyclooxygenase 2 Inhibitors administration & dosage, Cyclooxygenase 2 Inhibitors adverse effects, Drug Therapy, Combination, Female, Gastrointestinal Diseases chemically induced, Humans, Kidney Diseases chemically induced, Male, Middle Aged, Pyrazoles administration & dosage, Pyrazoles adverse effects, Secondary Prevention, Sulfonamides administration & dosage, Sulfonamides adverse effects, Adenoma prevention & control, Colorectal Neoplasms prevention & control, Cyclooxygenase 2 Inhibitors therapeutic use, Pyrazoles therapeutic use, Sulfonamides therapeutic use
- Abstract
Background: Studies showing that drugs that inhibit cyclooxygenase-2 (COX-2) reduce the number of colorectal adenomas in animals and patients with familial adenomatous polyposis suggest that COX-2 inhibitors may also prevent sporadic colorectal neoplasia., Methods: We randomly assigned patients who had adenomas removed before study entry to receive placebo (679 patients) or 200 mg (685 patients) or 400 mg (671 patients) of celecoxib twice daily. Randomization was stratified for the use of low-dose aspirin. Follow-up colonoscopies were performed at one and three years after randomization. The occurrence of newly detected colorectal adenomas was compared among the groups with the life-table extension of the Mantel-Haenszel test., Results: Follow-up colonoscopies were completed at year 1 in 89.5 percent of randomized patients, and at year 3 in 75.7 percent. The estimated cumulative incidence of the detection of one or more adenomas by year 3 was 60.7 percent for patients receiving placebo, as compared with 43.2 percent for those receiving 200 mg of celecoxib twice a day (risk ratio, 0.67; 95 percent confidence interval, 0.59 to 0.77; P<0.001) and 37.5 percent for those receiving 400 mg of celecoxib twice a day (risk ratio, 0.55; 95 percent confidence interval, 0.48 to 0.64; P<0.001). Serious adverse events occurred in 18.8 percent of patients in the placebo group, as compared with 20.4 percent of those in the low-dose celecoxib group (risk ratio, 1.1; 95 percent confidence interval, 0.9 to 1.3; P=0.5) and 23.0 percent of those in the high-dose group (risk ratio, 1.2; 95 percent confidence interval, 1.0 to 1.5; P=0.06). As compared with placebo, celecoxib was associated with an increased risk of cardiovascular events (risk ratio for the low dose, 2.6; 95 percent confidence interval, 1.1 to 6.1; and risk ratio for the high dose, 3.4; 95 percent confidence interval, 1.5 to 7.9)., Conclusions: These findings indicate that celecoxib is an effective agent for the prevention of colorectal adenomas but, because of potential cardiovascular events, cannot be routinely recommended for this indication. (ClinicalTrials.gov number, NCT00005094 [ClinicalTrials.gov].)., (Copyright 2006 Massachusetts Medical Society.)
- Published
- 2006
- Full Text
- View/download PDF
8. Modulation of tumor formation and intestinal cell migration by estrogens in the Apc(Min/+) mouse model of colorectal cancer.
- Author
-
Javid SH, Moran AE, Carothers AM, Redston M, and Bertagnolli MM
- Subjects
- Animals, Colorectal Neoplasms genetics, Colorectal Neoplasms metabolism, Colorectal Neoplasms pathology, Female, Immunohistochemistry, Immunoprecipitation, Intestinal Mucosa metabolism, Intestines pathology, Mice, Mice, Inbred C57BL, Ovariectomy, Receptors, Estrogen metabolism, Cell Movement drug effects, Colorectal Neoplasms prevention & control, Coumestrol pharmacology, Disease Models, Animal, Genes, APC, Genistein pharmacology, Intestines drug effects, Phytoestrogens pharmacology
- Abstract
Epidemiological studies suggest that post-menopausal hormone replacement therapy (HRT) reduces colorectal cancer (CRC) incidence. Phytoestrogens, including the soy isoflavone genistein and coumestrol, are used by many women as alternatives to HRT. Previous studies showed that ovariectomy induced a 77% increase in intestinal adenoma number in the C57BL/6J-Min/+ (Min/+) mouse, an animal model of adenomatous polyposis coli (APC)-associated CRC. Replacement of estradiol (E(2)) in ovariectomized Min/+ mice reduced tumor number to baseline and up-regulated the expression of estrogen receptor beta (ERbeta). We hypothesized that the phytoestrogens genistein and coumestrol would inhibit intestinal tumorigenesis in ovariectomized Min/+ mice. Min/+ and Apc(+/+) (WT) mice were ovariectomized and assigned to either a control diet or treatment with E(2), genistein or coumestrol. Treatment of ovariectomized Min/+ (Min/+ OX) mice with genistein resulted in a non-significant reduction in tumor number. Min/+ OX mice treated with coumestrol had significantly fewer tumors than untreated Min/+ OX controls and the same number of tumors as non-ovariectomized Min/+ mice. Bromodeoxyuridine migration assays also demonstrated that treatment with E(2) or coumestrol improved enterocyte migration rate. Immunoprecipitation and immunohistochemistry analyses showed that impaired association of the adherens junction proteins E-cadherin and beta-catenin in Min/+ mice was improved by treatment with either E(2) or coumestrol. Immunoblot analyses also showed that expression of ERbeta was elevated in enterocytes of Min/+ OX mice treated with E(2) or coumestrol as compared with those of untreated Min/+ OX mice. In conclusion, both coumestrol and E(2) prevent intestinal tumorigenesis and ameliorate enterocyte migration and intercellular adhesion in the Apc(Min/+) mouse model of CRC.
- Published
- 2005
- Full Text
- View/download PDF
9. Association between biallelic and monoallelic germline MYH gene mutations and colorectal cancer risk.
- Author
-
Croitoru ME, Cleary SP, Di Nicola N, Manno M, Selander T, Aronson M, Redston M, Cotterchio M, Knight J, Gryfe R, and Gallinger S
- Subjects
- Adenomatous Polyposis Coli genetics, Aspartic Acid, Base Pair Mismatch, Case-Control Studies, Colorectal Neoplasms epidemiology, Colorectal Neoplasms, Hereditary Nonpolyposis genetics, Cysteine, DNA Mutational Analysis, DNA, Neoplasm analysis, Gene Frequency, Genetic Predisposition to Disease, Glycine, Humans, Ontario epidemiology, Phenotype, Risk Factors, Tyrosine, Biomarkers, Tumor genetics, Colorectal Neoplasms genetics, DNA Glycosylases genetics, Germ-Line Mutation, Loss of Heterozygosity
- Abstract
The MutY human homologue (MYH) gene encodes a member of the base excision repair pathway that is involved in repairing oxidative damage to DNA. Two germline MYH gene mutations that result in Myh proteins containing amino acid substitutions Y165C and G382D (hereafter called the Y165C and G382D mutations) are associated with adenomatous poly-posis and colorectal cancer among patients from several European poly-posis registries. We used a population-based series of 1238 colorectal cancer patients and 1255 healthy control subjects from Ontario, Canada, to examine the risk of colorectal cancer among biallelic and monoallelic germline MYH Y165C and G382D mutation carriers. The entire MYH gene coding region was screened in all MYH Y165C and G382D mutation carriers. Compared with noncarriers, biallelic and monoallelic germline MYH gene mutation carriers had an increased risk of colorectal cancer and were more likely to have first-or second-degree relatives with colorectal cancer (relative risk = 1.54, 95% confidence interval = 1.10 to 2.16). The increased risk of colorectal cancer in biallelic and monoallelic MYH gene mutation carriers was not consistently associated with the development of multiple adenomatous polyps. Loss of heterozygosity in at least one of four loci in MYH was detected in eight (47%) of 17 colorectal tumors from monoallelic MYH gene mutation carriers but in only two (20%) of 10 colorectal tumors from biallelic MYH gene mutation carriers. These two MYH gene mutations may account for a substantial fraction of hereditary colorectal cancer.
- Published
- 2004
- Full Text
- View/download PDF
10. Microsatellite instability as a prognostic factor in resected colorectal cancer liver metastases.
- Author
-
Haddad R, Ogilvie RT, Croitoru M, Muniz V, Gryfe R, Pollet A, Shanmugathasan P, Fitzgerald T, Law CH, Hanna SS, Jothy S, Redston M, Gallinger S, and Smith AJ
- Subjects
- Aged, Cohort Studies, Female, Humans, Male, Middle Aged, Multivariate Analysis, Polymerase Chain Reaction, Prognosis, Survival Analysis, Chromosomal Instability genetics, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Liver Neoplasms genetics, Liver Neoplasms secondary, Microsatellite Repeats genetics
- Abstract
Background: Two distinct genetic mutational pathways characterized by either chromosomal instability or high-frequency microsatellite instability (MSI-H) are currently recognized in the pathogenesis of colorectal cancer (CRC). Recently, it has been shown that patients with primary CRC that displays MSI-H have a significant, stage-independent, multivariate survival advantage. Untreated CRC hepatic metastases are incurable and are associated with a median survival of 4 to 12 months. Conversely, surgical resection in selected patients results in a 20% to 50% cure rate. The aim of this study was to investigate the prognostic importance of MSI-H in patients undergoing resection of hepatic CRC metastases., Methods: DNA was extracted from paraffin-embedded, resected metastatic CRC liver lesions and corresponding normal liver parenchyma from 190 patients. MSI-H status was determined by polymerase chain reaction-based evaluation of the noncoding mononucleotide repeats BAT-25 and BAT-26., Results: MSI was detected in tumors from 5 (2.7%) of the 190 CRC patients. All MSI-H tumors were in patients with node-positive CRC primary tumors. The median survival after hepatic resection of MSI-H and non-MSI-H tumors was 67 and 61 months, respectively (P = .9)., Conclusions: These data suggest that MSI-H is not a common feature in resected CRC liver metastases and do not suggest a role for MSI in stratifying good versus poor prognosis in these patients.
- Published
- 2004
- Full Text
- View/download PDF
11. alpha2HS-glycoprotein, an antagonist of transforming growth factor beta in vivo, inhibits intestinal tumor progression.
- Author
-
Swallow CJ, Partridge EA, Macmillan JC, Tajirian T, DiGuglielmo GM, Hay K, Szweras M, Jahnen-Dechent W, Wrana JL, Redston M, Gallinger S, and Dennis JW
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Binding, Competitive, Blood Proteins deficiency, Blood Proteins genetics, Blood Proteins pharmacology, Cattle, Colorectal Neoplasms genetics, Colorectal Neoplasms immunology, Colorectal Neoplasms metabolism, Female, Humans, Macrophage Activation physiology, Macrophages, Peritoneal immunology, Male, Mice, Mice, Inbred C57BL, Middle Aged, Protein Serine-Threonine Kinases, Receptor, Transforming Growth Factor-beta Type II, Receptors, Transforming Growth Factor beta metabolism, Signal Transduction physiology, Transforming Growth Factor beta immunology, Transforming Growth Factor beta metabolism, Transforming Growth Factor beta physiology, Transforming Growth Factor beta1, alpha-2-HS-Glycoprotein, Blood Proteins physiology, Colorectal Neoplasms pathology, Transforming Growth Factor beta antagonists & inhibitors
- Abstract
Transforming growth factor (TGF)-beta1 is associated with tumor progression and resistance to chemotherapy in established cancers, as well as host immune suppression. Here, we show that the serum glycoprotein alpha2-HS-glycoprotein (AHSG) blocks TGF-beta1 binding to cell surface receptors, suppresses TGF-beta signal transduction, and inhibits TGF-beta-induced epithelial-mesenchymal transition, suggesting that AHSG may play a role in tumor progression. In 66 consecutive sporadic human colorectal cancer specimens, we observed a 3-fold depletion of ASHG in tumor compared with normal tissue, whereas levels of other abundant plasma proteins, albumin and transferrin, were equivalent. Using the Multiple intestinal neoplasia/+ (Min/+) mouse model of intestinal tumorigenesis, we found twice as many intestinal polyps overall, twice as many large polyps (>3 mm diameter), and more progression to invasive adenocarcinoma in Min/+ Ahsg-/- mice than in littermates expressing Ahsg. Phosphorylated Smad2 was more abundant in the intestinal mucosa and tumors of Min/+ mice lacking Ahsg, demonstrating increased TGF-beta signaling in vivo. Furthermore, TGF-beta-mediated suppression of immune cell function was exaggerated in Ahsg-/- animals, as shown by inhibition of macrophage activation and reduction in 12-O-tetradecanoylphorbol 13-acetate-induced cutaneous inflammation. Reconstitution of Ahsg-/- mice with bovine Ahsg suppressed endogenous TGF-beta-dependent signaling to wild-type levels, suggesting that therapeutic enhancement of AHSG levels may benefit patients whose tumors are driven by TGF-beta.
- Published
- 2004
- Full Text
- View/download PDF
12. Chromosome 7q31 allelic imbalance and somatic mutations of RAY1/ST7 gene in colorectal cancer.
- Author
-
Haddad R, Vincent JB, Gryfe R, Kim H, Wen J, Redston M, Scherer SW, and Gallinger S
- Subjects
- Chromosomes, Human, Pair 7, Humans, Mutation, Adenocarcinoma genetics, Allelic Imbalance, Colorectal Neoplasms genetics, Proteins genetics, Tumor Suppressor Proteins genetics
- Abstract
ST7 is a putative tumor suppressor gene on chromosome 7q31. However, the role of ST7 as a tumor suppressor is uncertain as somatic mutations have been difficult to demonstrate. In order to investigate the genetic role of RAY1/ST7 in tumorigenesis, we have screened 135 colorectal cancers for loss of heterozygosity (LOH) at chromosome 7q31. The entire RAY1/ST7 gene, including intron/exon boundaries and alternate 5' and 3' sequences of 15/124 (12%) informative cancers with LOH were characterized. No somatic mutations of the RAY1/ST7 gene were observed. Our results do not support a role for RAY1/ST7 as a colorectal cancer tumor suppressor gene.
- Published
- 2004
- Full Text
- View/download PDF
13. Tumor microsatellite-instability status as a predictor of benefit from fluorouracil-based adjuvant chemotherapy for colon cancer.
- Author
-
Ribic CM, Sargent DJ, Moore MJ, Thibodeau SN, French AJ, Goldberg RM, Hamilton SR, Laurent-Puig P, Gryfe R, Shepherd LE, Tu D, Redston M, and Gallinger S
- Subjects
- Analysis of Variance, Base Pair Mismatch, Chemotherapy, Adjuvant, Colorectal Neoplasms mortality, Colorectal Neoplasms surgery, DNA Repair, Follow-Up Studies, Humans, Neoplasm Staging, Prognosis, Survival Analysis, Antimetabolites, Antineoplastic therapeutic use, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics, DNA, Neoplasm analysis, Fluorouracil therapeutic use, Microsatellite Repeats genetics
- Abstract
Background: Colon cancers with high-frequency microsatellite instability have clinical and pathological features that distinguish them from microsatellite-stable tumors. We investigated the usefulness of microsatellite-instability status as a predictor of the benefit of adjuvant chemotherapy with fluorouracil in stage II and stage III colon cancer., Methods: Tumor specimens were collected from patients with colon cancer who were enrolled in randomized trials of fluorouracil-based adjuvant chemotherapy. Microsatellite instability was assessed with the use of mononucleotide and dinucleotide markers., Results: Of 570 tissue specimens, 95 (16.7 percent) exhibited high-frequency microsatellite instability. Among 287 patients who did not receive adjuvant therapy, those with tumors displaying high-frequency microsatellite instability had a better five-year rate of overall survival than patients with tumors exhibiting microsatellite stability or low-frequency instability (hazard ratio for death, 0.31 [95 percent confidence interval, 0.14 to 0.72]; P=0.004). Among patients who received adjuvant chemotherapy, high-frequency microsatellite instability was not correlated with increased overall survival (hazard ratio for death, 1.07 [95 percent confidence interval, 0.62 to 1.86]; P=0.80). The benefit of treatment differed significantly according to the microsatellite-instability status (P=0.01). Adjuvant chemotherapy improved overall survival among patients with microsatellite-stable tumors or tumors exhibiting low-frequency microsatellite instability, according to a multivariate analysis adjusted for stage and grade (hazard ratio for death, 0.72 [95 percent confidence interval, 0.53 to 0.99]; P=0.04). By contrast, there was no benefit of adjuvant chemotherapy in the group with high-frequency microsatellite instability., Conclusions: Fluorouracil-based adjuvant chemotherapy benefited patients with stage II or stage III colon cancer with microsatellite-stable tumors or tumors exhibiting low-frequency microsatellite instability but not those with tumors exhibiting high-frequency microsatellite instability., (Copyright 2003 Massachusetts Medical Society)
- Published
- 2003
- Full Text
- View/download PDF
14. Heterozygosity for the BLM(Ash) mutation and cancer risk.
- Author
-
Cleary SP, Zhang W, Di Nicola N, Aronson M, Aube J, Steinman A, Haddad R, Redston M, Gallinger S, Narod SA, and Gryfe R
- Subjects
- Aged, Aged, 80 and over, Alleles, Case-Control Studies, Female, Genetic Predisposition to Disease, Heterozygote, Humans, Male, Middle Aged, Bloom Syndrome genetics, Colorectal Neoplasms genetics, Jews genetics, Mutation
- Abstract
Bloom syndrome is an autosomal recessive disorder whose characteristics include an increased risk for many types of cancers. In contrast to the homozygous mutations of Bloom syndrome, heterozygous carriers of BLM mutations may be at increased risk for developing colorectal cancer. We have screened 2,333 Jewish individuals, including 497 individuals with colorectal cancer, 125 with adenomatous polyps, 767 with noncolorectal cancers and 944 controls for the truncating BLM(Ash) founder mutation. The BLM(Ash) mutation was carried by 0.80% of individuals with colorectal neoplasia, 0.87% of those with any type of cancer and 0.85% of controls. In addition to case-control data, we found no evidence to support a significant relationship between increased cancer risk and heterozygous BLM(Ash) mutations with respect to age of cancer diagnosis, tumor multiplicity or family cancer history.
- Published
- 2003
15. Mutation profiling of mismatch repair-deficient colorectal cncers using an in silico genome scan to identify coding microsatellites.
- Author
-
Park J, Betel D, Gryfe R, Michalickova K, Di Nicola N, Gallinger S, Hogue CW, and Redston M
- Subjects
- Algorithms, Computational Biology, DNA Repair, Humans, Base Pair Mismatch, Colorectal Neoplasms genetics, Microsatellite Repeats, Mutation
- Abstract
Human colorectal, endometrial, and gastric cancers with defective DNA mismatch repair (MMR) have microsatellite instability, a unique molecular alteration characterized by widespread frameshift mutations of repetitive DNA sequences. We developed "Kangaroo," a bioinformatics program for searches in nucleotide and protein sequence databases, and performed an in silico genome scan for DNA coding microsatellites that may have novel mutations in MMR-deficient cancers. Examination of 29 previously untested coding polyadenines revealed widespread mutations in MMR-deficient colorectal cancers, with the highest frequencies in ERCC5, CASP8AP2, p72, RAD50, CDC25, RECQL1, CBF2, RACK7, GRK4, and DNAPK (range, 10-33%). This algorithm allows comprehensive mutation profiling of MMR-deficient cancers, an important step in understanding the pathogenesis of these neoplasms.
- Published
- 2002
16. Immunohistochemistry versus microsatellite instability testing in phenotyping colorectal tumors.
- Author
-
Lindor NM, Burgart LJ, Leontovich O, Goldberg RM, Cunningham JM, Sargent DJ, Walsh-Vockley C, Petersen GM, Walsh MD, Leggett BA, Young JP, Barker MA, Jass JR, Hopper J, Gallinger S, Bapat B, Redston M, and Thibodeau SN
- Subjects
- Aged, Colorectal Neoplasms pathology, Cost-Benefit Analysis, DNA Mutational Analysis, DNA Repair, Female, Humans, Immunohistochemistry, Male, Middle Aged, Phenotype, Polymerase Chain Reaction, Predictive Value of Tests, Sensitivity and Specificity, Base Pair Mismatch genetics, Colorectal Neoplasms genetics, DNA, Neoplasm genetics, Microsatellite Repeats genetics
- Abstract
Purpose: To compare microsatellite instability (MSI) testing with immunohistochemical (IHC) detection of hMLH1 and hMSH2 in colorectal cancer., Patients and Methods: Colorectal cancers from 1,144 patients were assessed for DNA mismatch repair deficiency by two methods: MSI testing and IHC detection of hMLH1 and hMSH2 gene products. High-frequency MSI (MSI-H) was defined as more than 30% instability of at least five markers; low-level MSI (MSI-L) was defined as 1% to 29% of loci unstable., Results: Of 1,144 tumors tested, 818 showed intact expression of hMLH1 and hMSH2. Of these, 680 were microsatellite stable (MSS), 27 were MSI-H, and 111 were MSI-L. In all, 228 tumors showed absence of hMLH1 expression and 98 showed absence of hMSH2 expression: all were MSI-H., Conclusion: IHC in colorectal tumors for protein products hMLH1 and hMSH2 provides a rapid, cost-effective, sensitive (92.3%), and extremely specific (100%) method for screening for DNA mismatch repair defects. The predictive value of normal IHC for an MSS/MSI-L phenotype was 96.7%, and the predictive value of abnormal IHC was 100% for an MSI-H phenotype. Testing strategies must take into account acceptability of missing some cases of MSI-H tumors if only IHC is performed.
- Published
- 2002
- Full Text
- View/download PDF
17. Suppression of intestinal polyps in Msh2-deficient and non-Msh2-deficient multiple intestinal neoplasia mice by a specific cyclooxygenase-2 inhibitor and by a dual cyclooxygenase-1/2 inhibitor.
- Author
-
Lal G, Ash C, Hay K, Redston M, Kwong E, Hancock B, Mak T, Kargman S, Evans JF, and Gallinger S
- Subjects
- 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, 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
- 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.
- Published
- 2001
18. Carcinogenesis in the GI tract: from morphology to genetics and back again.
- Author
-
Redston M
- Subjects
- Adaptor Proteins, Signal Transducing, Base Pair Mismatch, Carrier Proteins, 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, Aneuploidy, Colorectal Neoplasms chemistry, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, DNA-Binding Proteins, Loss of Heterozygosity, Microsatellite Repeats
- 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.
- Published
- 2001
- Full Text
- View/download PDF
19. Colorectal carcinomas in mice lacking the catalytic subunit of PI(3)Kgamma.
- Author
-
Sasaki T, Irie-Sasaki J, Horie Y, Bachmaier K, Fata JE, Li M, Suzuki A, Bouchard D, Ho A, Redston M, Gallinger S, Khokha R, Mak TW, Hawkins PT, Stephens L, Scherer SW, Tsao M, and Penninger JM
- Subjects
- 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, Colorectal Neoplasms enzymology, Phosphatidylinositol 3-Kinases metabolism
- 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.
- Published
- 2000
- Full Text
- View/download PDF
20. Tumor microsatellite instability and clinical outcome in young patients with colorectal cancer.
- Author
-
Gryfe R, Kim H, Hsieh ET, Aronson MD, Holowaty EJ, Bull SB, Redston M, and Gallinger S
- Subjects
- Adult, Colorectal Neoplasms mortality, Colorectal Neoplasms pathology, Colorectal Neoplasms therapy, Female, Humans, Male, Middle Aged, Multivariate Analysis, Mutation, Prognosis, Survival Analysis, Colorectal Neoplasms genetics, Microsatellite Repeats genetics
- 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.
- Published
- 2000
- Full Text
- View/download PDF
21. Immunohistochemistry for hMLH1 and hMSH2: a practical test for DNA mismatch repair-deficient tumors.
- Author
-
Marcus VA, Madlensky L, Gryfe R, Kim H, So K, Millar A, Temple LK, Hsieh E, Hiruki T, Narod S, Bapat BV, Gallinger S, and Redston M
- Subjects
- 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, Adenocarcinoma genetics, Base Pair Mismatch genetics, Colorectal Neoplasms genetics, DNA Repair, DNA-Binding Proteins, Neoplasm Proteins analysis, Proto-Oncogene Proteins analysis
- 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.
- Published
- 1999
- Full Text
- View/download PDF
22. Mismatch repair gene defects contribute to the genetic basis of double primary cancers of the colorectum and endometrium.
- Author
-
Millar AL, Pal T, Madlensky L, Sherman C, Temple L, Mitri A, Cheng H, Marcus V, Gallinger S, Redston M, Bapat B, and Narod S
- Subjects
- 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, Colorectal Neoplasms genetics, DNA-Binding Proteins, Endometrial Neoplasms genetics, Neoplasm Proteins genetics, Neoplasms, Multiple Primary genetics, Proto-Oncogene Proteins genetics
- 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.
- Published
- 1999
- Full Text
- View/download PDF
23. Somatic instability of the APC I1307K allele in colorectal neoplasia.
- Author
-
Gryfe R, Di Nicola N, Gallinger S, and Redston M
- Subjects
- Colorectal Neoplasms ethnology, DNA Mutational Analysis, Gene Deletion, Germ-Line Mutation genetics, Humans, Adenomatous Polyposis Coli genetics, Alleles, Colorectal Neoplasms genetics, Genes, APC genetics, Jews genetics, Microsatellite Repeats genetics, Point Mutation genetics
- Abstract
The adenomatous polyposis coli (APC) gene is proposed to function as a gatekeeper of colorectal neoplasia. A germ-line variant of this gene, the APC I1307K allele, is present in approximately 6% of the Ashkenazi Jewish population. To assess the role in tumorigenesis of the variant (A)8 tract produced by this allele, we undertook a somatic mutation analysis of the region surrounding codon 1307 in colorectal tumors from APC I1307K carriers. Somatic mutations involving the variant (A)8 tract were identified in 53 of 127 (42%) tumors from APC I1307K carriers compared with 5 of 127 (4%) mutations involving the wild-type allele of these tumors (P < 0.0001). Loss of heterozygosity of the wild-type allele was significantly more common in tumors with APC I1307K allele mutations (25 of 41, 61%) compared with APC I1307K carrier tumors without mutation of the variant (A)8 tract (12 of 53, 23%; P < 0.0005). This somatic biallelic APC inactivation further confirms the biological importance of the I1307K germ-line variant. The vast majority of APC I1307K somatic mutations consisted of a single adenine insertion (insA) involving the variant (A)8 tract. This insA mutation was mutually exclusive of the presence of microsatellite instability with 0 of 49 tumors with insA displaying BAT-26 instability compared with 9 of 78 tumors without insA (P=0.01). These findings support a model where somatic instability of the (A)8 tract produced by the APC I1307K allele leads to increased APC gene inactivation and directly accounts for 42% of the colorectal neoplasms occurring in APC I1307K carriers.
- Published
- 1998
24. Molecular biology of colorectal cancer.
- Author
-
Gryfe R, Swallow C, Bapat B, Redston M, Gallinger S, and Couture J
- Subjects
- Colorectal Neoplasms diagnosis, Colorectal Neoplasms therapy, DNA, Neoplasm analysis, Disease Progression, Genes, Tumor Suppressor genetics, Genetic Markers, Genetic Testing methods, Humans, Models, Genetic, Molecular Biology, Mutation, Oncogenes genetics, Pedigree, Risk Assessment, Colorectal Neoplasms genetics
- Abstract
Colorectal cancer is a significant cause of morbidity and mortality in Western populations. This cancer develops as a result of the pathologic transformation of normal colonic epithelium to an adenomatous polyp and ultimately an invasive cancer. The multistep progression requires years and possibly decades and is accompanied by a number of recently characterized genetic alterations. Mutations in two classes of genes, tumor-suppressor genes and proto-oncogenes, are thought to impart a proliferative advantage to cells and contribute to development of the malignant phenotype. Inactivating mutations of both copies (alleles) of the adenomatous polyposis coli (APC) gene--a tumor-suppressor gene on chromosome 5q--mark one of the earliest events in colorectal carcinogenesis. Germline mutation of the APC gene and subsequent somatic mutation of the second APC allele cause the inherited familial adenomatous polyposis syndrome. This syndrome is characterized by the presence of hundreds to thousands of colonic adenomatous polyps. If these polyps are left untreated, colorectal cancer develops. Mutation leading to dysregulation of the K-ras protooncogene is also thought to be an early event in colon cancer formation. Conversely, loss of heterozygosity on the long arm of chromosome 18 (18q) occurs later in the sequence of development from adenoma to carcinoma, and this mutation may predict poor prognosis. Loss of the 18q region is thought to contribute to inactivation of the DCC tumor-suppressor gene. More recent evidence suggests that other tumor-suppressor genes--DPC4 and MADR2 of the transforming growth factor beta (TGF-beta) pathway--also may be inactivated by allelic loss on chromosome 18q. In addition, mutation of the tumor-suppressor gene p53 on chromosome 17p appears to be a late phenomenon in colorectal carcinogenesis. This mutation may allow the growing tumor with multiple genetic alterations to evade cell cycle arrest and apoptosis. Neoplastic progression is probably accompanied by additional, undiscovered genetic events, which are indicated by allelic loss on chromosomes 1q, 4p, 6p, 8p, 9q, and 22q in 25% to 50% of colorectal cancers. Recently, a third class of genes, DNA repair genes, has been implicated in tumorigenesis of colorectal cancer. Study findings suggest that DNA mismatch repair deficiency, due to germline mutation of the hMSH2, hMLH1, hPMS1, or hPMS2 genes, contributes to development of hereditary nonpolyposis colorectal cancer. The majority of tumors in patients with this disease and 10% to 15% of sporadic colon cancers display microsatellite instability, also know as the replication error positive (RER+) phenotype. This molecular marker of DNA mismatch repair deficiency may predict improved patient survival. Mismatch repair deficiency is thought to lead to mutation and inactivation of the genes for type II TGF-beta receptor and insulin-like growth-factor II receptor. Individuals from families at high risk for colorectal cancer (hereditary nonpolyposis colorectal cancer or familial adenomatous polyposis) should be offered genetic counseling, predictive molecular testing, and when indicated, endoscopic surveillance at appropriate intervals. Recent studies have examined colorectal carcinogenesis in the light of other genetic processes. Telomerase activity is present in almost all cancers, including colorectal cancer, but rarely in benign lesions such as adenomatous polyps or normal tissues. Furthermore, genetic alterations that allow transformed colorectal epithelial cells to escape cell cycle arrest or apoptosis also have been recognized. In addition, hypomethylation or hypermethylation of DNA sequences may alter gene expression without nucleic acid mutation.
- Published
- 1997
- Full Text
- View/download PDF
25. A preventive registry for hereditary nonpolyposis colorectal cancer.
- Author
-
Madlensky L, Berk TC, Bapat BV, McLeod RS, Couture J, Baron D, Hiruki T, Redston M, Cohen Z, and Gallinger S
- Subjects
- Colorectal Neoplasms genetics, Colorectal Neoplasms, Hereditary Nonpolyposis genetics, Databases, Factual, Family, Health Education, Humans, Pedigree, Referral and Consultation, Colorectal Neoplasms prevention & control, Colorectal Neoplasms, Hereditary Nonpolyposis prevention & control, Registries standards
- Abstract
Hereditary nonpolyposis colorectal cancer (HNPCC) is a genetic disorder characterized by a strong family history of colorectal and extracolonic cancers, usually at a young age. This article presents a new provincial service for families with HNPCC. The Steve Atanas Stavro Familial Gastrointestinal Cancer Registry at Mount Sinai Hospital is accruing patients that meet a set of criteria establishing a putative diagnosis of HNPCC. The objectives of the Registry are to develop and assess patient pedigrees, to coordinate screening procedures for at-risk persons, to maintain a prospective database of patient information, to provide education and support for families and to contribute to research. To date, surgeons and patients are the most common referral sources, while oncologists and geneticists are the least common. The ultimate goal of the HNPCC service is the secondary prevention of cancer and a corresponding decrease in mortality for HNPCC family members.
- Published
- 1995
26. Molecular genetic profiles of colitis-associated neoplasms.
- Author
-
Kern SE, Redston M, Seymour AB, Caldas C, Powell SM, Kornacki S, and Kinzler KW
- Subjects
- Adenocarcinoma etiology, Adenocarcinoma genetics, Adenocarcinoma metabolism, Adult, Aged, Alleles, Colon metabolism, Colon pathology, Colorectal Neoplasms etiology, Colorectal Neoplasms metabolism, Genes, APC genetics, Genes, p53 genetics, Genes, ras genetics, Humans, Immunohistochemistry, Middle Aged, Mutation, Retinoblastoma Protein metabolism, Colitis complications, Colorectal Neoplasms genetics
- Abstract
Background/aims: A major long-term risk for patients with chronic idiopathic colitis is the development of colorectal dysplasia and adenocarcinoma. The presence or absence of specific genetic changes in these lesions will provide important insights into the relationship of colitis-associated dysplasia and the development of carcinoma., Methods: A case-study approach was used to develop detailed molecular genetic profiles of advanced dysplasias and carcinomas from six patients with ulcerative colitis or Crohn's colitis., Results: Numerous genetic alterations were identified in each of the dysplasias and carcinomas profiled. These genetic alterations involved many of the same targets found in sporadic colorectal tumors and included multiple sites of allelic deletion, microsatellite instabilities, and mutations of the K-ras, p53, and APC genes. The progression of dysplasia to carcinoma was often accompanied by an accumulation of these mutations., Conclusions: Genetic alterations are common in colitis-associated neoplasia, just as in sporadic colorectal neoplasia. This could have important implications for the evaluation and treatment of patients with colitis.
- Published
- 1994
- Full Text
- View/download PDF
27. Common and distinct genomic events in sporadic colorectal cancer and diverse cancer types
- Author
-
Eric S. Martin, Raju Kucherlapati, Alexei Protopopov, Elena Ivanova, Bin Feng, Cameron Brennan, Yonghong Xiao, Giovanni Tonon, Lynda Chin, Gerald Bailey, Ronald A. DePinho, Kate Montgomery, Raktim Sinha, Alec C. Kimmelman, Mark Redston, Martin, E, Tonon, G, Sinha, R, Xiao, Y, Feng, B, Kimmelman, Ac, Protopopov, A, Ivanova, E, Brennan, C, Montgomery, K, Kucherlapati, R, Bailey, G, Redston, M, Chin, L, and Depinho, Ra
- Subjects
Cancer Research ,Colorectal cancer ,Copy number analysis ,Genomics ,Biology ,Genome ,Chromosome instability ,Cell Line, Tumor ,medicine ,Humans ,Genetics ,Chromosome Aberrations ,Models, Genetic ,Gene Expression Profiling ,Microsatellite instability ,Cancer ,Nucleic Acid Hybridization ,Sequence Analysis, DNA ,medicine.disease ,Immunohistochemistry ,Gene expression profiling ,Gene Expression Regulation, Neoplastic ,Oncology ,Mutation ,Colorectal Neoplasms ,Genes, Neoplasm - Abstract
Colorectal cancer (CRC) is a major cause of cancer morbidity and mortality, and elucidation of its underlying genetics has advanced diagnostic screening, early detection, and treatment. Because CRC genomes are characterized by numerous non-random chromosomal structural alterations, we sought to delimit regions of recurrent amplifications and deletions in a collection of 42 primary specimens and 37 tumor cell lines derived from chromosomal instability neoplasia and microsatellite instability neoplasia CRC subtypes and to compare the pattern of genomic aberrations in CRC with those in other cancers. Application of oligomer-based array-comparative genome hybridization and custom analytic tools identified 50 minimal common regions (MCRs) of copy number alterations, 28 amplifications, and 22 deletions. Fifteen were highly recurrent and focal (
- Published
- 2007
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.