Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 123
Filtrar
1.
J Clin Pathol ; 62(11): 1034-6, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19861563

RESUMO

Mucosal prolapse syndrome comprises a variety of clinical and histopathological entities, with mucosal prolapse as the underlying pathogenic mechanism. Due to variable clinical, endoscopic and histopathological presentation and rareness of symptomatic mucosal prolapse, misdiagnosis resulting in delayed or inappropriate treatment is frequent. This report describes a patient initially diagnosed with a colorectal polyposis syndrome consisting of multiple rectal hyperplastic and adenomatous polyps. But after careful review of medical history and histopathology, the patient was found to have a rare variant of solitary rectal ulcer syndrome presenting as rectal polyposis. The recognition of rectal polyposis as a manifestation of solitary rectal ulcer syndrome/mucosal prolapse syndrome will improve diagnosis and treatment and prevent inappropriate management of this condition.


Assuntos
Polipose Intestinal/etiologia , Prolapso Retal/complicações , Colonoscopia , Feminino , Humanos , Mucosa Intestinal/patologia , Polipose Intestinal/patologia , Pessoa de Meia-Idade , Prolapso Retal/diagnóstico , Prolapso Retal/patologia , Síndrome
3.
Gut ; 57(5): 623-7, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18178612

RESUMO

BACKGROUND/AIMS: Juvenile polyposis syndrome (JPS) is a rare autosomal dominant disorder characterised by multiple gastrointestinal juvenile polyps and an increased risk of colorectal cancer. This syndrome is caused by germline mutation of either SMAD4 or BMPR1A, and possibly ENG. PTEN, originally linked to Cowden syndrome and Bannayan-Riley-Ruvalcaba syndrome, has also been associated with JPS. By direct sequencing, germline mutations are found in only 30-40% of patients with a JPS phenotype. Therefore, alternative ways of inactivation of the known JPS genes, or additional genes predisposing to JPS may be involved. In this study, a comprehensive genetic analysis of SMAD4, BMPR1A, PTEN and ENG is performed through direct sequencing and multiplex ligation-dependent probe amplification (MLPA) in JPS patients. METHODS: Archival material of 29 patients with JPS from 27 families was collected. Direct sequencing and MLPA analysis were performed to search for germline defects in SMAD4, BMPR1A, PTEN and ENG. RESULTS: A germline defect in SMAD4, BMPR1A or PTEN was found in 13 of 27 (48.1%) unrelated JPS patients. Nine mutations (33.3%) were detected by direct sequencing, including six (22.2%) SMAD4 mutations and three (11.1%) BMPR1A mutations. MLPA identified four additional patients (14.8%) with germline hemizygous large genomic deletions, including one deletion of SMAD4, one deletion of exons 10 and 11 of BMPR1A, and two unrelated patients with deletion of both BMPR1A and PTEN. No ENG gene mutations were found. CONCLUSION: Large genomic deletions of SMAD4, BMPR1A and PTEN are a common cause of JPS. Using direct sequencing and MLPA, a germline defect was detected in 48.1% of JPS patients. MLPA identified 14.8% (4/27) of these mutations. Since a substantial percentage of JPS patients carry a germline deletion and MLPA is a reliable and user-friendly technique, it is concluded that MLPA is a valuable adjunct in JPS diagnosis.


Assuntos
Polipose Adenomatosa do Colo/genética , Sequência de Bases/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , PTEN Fosfo-Hidrolase/genética , Deleção de Sequência/genética , Proteína Smad4/genética , Sequência de Bases/fisiologia , Análise Mutacional de DNA/métodos , Feminino , Genoma , Genótipo , Mutação em Linhagem Germinativa/genética , Humanos , Masculino , Linhagem , Fenótipo , Deleção de Sequência/fisiologia
4.
Clin Genet ; 72(6): 568-73, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17924967

RESUMO

LKB1/STK11 germline inactivations are identified in the majority (66-94%) of Peutz-Jeghers syndrome (PJS) patients. Therefore, defects in other genes or so far unidentified ways of LKB1 inactivation may cause PJS. The genes encoding the MARK proteins, homologues of the Par1 polarity protein that associates with Par4/Lkb1, were analyzed in this study because of their link to LKB1 and cell polarity. The genetic defect underlying PJS was determined through analysis of both LKB1 and all four MARK genes. LKB1 point mutations and small deletions were identified in 18 of 23 PJS families using direct sequencing and multiplex ligation-dependent probe amplification analysis identified exon deletions in 3 of 23 families. In total, 91% of the studied families showed LKB1 inactivation. Furthermore, a MARK1, MARK2, MARK3 and MARK4 mutation analysis and an MARK4 quantitative multiplex polymerase chain reaction analysis to identify exon deletions on another eight PJS families without identified LKB1 germline mutation did not identify mutations in the MARK genes. LKB1 defects are the major cause of PJS and genes of the MARK family do not represent alternative PJS genes. Other mechanisms of inactivation of LKB1 may cause PJS in the remaining families.


Assuntos
Família Multigênica , Síndrome de Peutz-Jeghers/genética , Proteínas Serina-Treonina Quinases/genética , Quinases Proteína-Quinases Ativadas por AMP , Éxons , Feminino , Mutação em Linhagem Germinativa , Humanos , Íntrons , Masculino , Síndrome de Peutz-Jeghers/enzimologia , Deleção de Sequência
6.
Oral Dis ; 13(4): 360-5, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17577321

RESUMO

Patients with familial adenomatous polyposis (FAP) develop multiple premalignant colorectal adenomas. Untreated, one or more of these polyps will progress to colorectal carcinoma in middle-aged adults. Extra-intestinal manifestations of FAP are frequently observed and this combination has been called Gardner's syndrome. Oral and maxillofacial symptoms of FAP include an increased risk of jaw osteomas, odontomas and supernumerary or unerupted teeth. Early diagnosis of FAP is crucial and may be life saving. As oral signs usually precede gastrointestinal symptoms, the dentist may play an important role in the diagnosis of FAP.


Assuntos
Polipose Adenomatosa do Colo/complicações , Neoplasias Faciais/etiologia , Neoplasias Maxilomandibulares/etiologia , Doenças Dentárias/etiologia , Polipose Adenomatosa do Colo/genética , Síndrome de Gardner/diagnóstico , Humanos , Odontoma/etiologia , Osteoma/etiologia , Dente Supranumerário/etiologia , Dente não Erupcionado/etiologia
7.
Dig Dis Sci ; 52(6): 1462-70, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17372834

RESUMO

Dietary folate status appears to influence risk for colorectal cancer possibly by alterations in DNA methylation and nucleotide precursor pools. Polymorphisms (677C-->T and 1298A-->C) in methylenetetrahydrofolate reductase (MTHFR), a key enzyme in folate metabolism, determines enzyme activity. The frequency of polymorphisms in the gene varies extensively in different populations. We sought to determine the association between folate status, folate metabolism, DNA methylation, tobacco, alcohol consumption, and the risk of colorectal adenomas in African Americans. Among 58 patients who underwent a clinically indicated colonoscopy, 23 patients with histology confirmed colorectal polyps and 35 patients without were recruited for a case-control study. Blood samples were collected from fasting patients for determination of serum and red blood cell (RBC) folate, homocysteine, vitamin B(12), and methylation status. Polymerase chain reaction (PCR) restriction fragment length polymorphism (RFLP) technique was performed to identify the MTHFR 677 C-->T polymorphism and specific PCR was used to analyze adenomatous polyposis coli (APC) gene-promoter sequence methylation. Among 23 cases, 49 polyps (adenomatous, n = 41 and hyperplastic, n= 8) were identified. Twenty-eight (57%) of the polyps were on the left side and 21 (42%) were on the right side of the colon. There was no association between the presence of colon polyps and levels of folate (serum, RBC), vitamin B(12), or homocysteine. Forty-eight individuals (84%) were homozygous for 677 CC. Of these individuals, 18 (37.5%) had >/=1 colorectal polyps, whereas 30 (62.5%) had no polyps. Nine individuals were heterozygous for 677 CT, and 4 (44%) of these individuals had colon polyps. Eighty-eight percent of the APC gene-promoter sequences tested using peripheral blood DNA from patients were unmethylated. Among the individuals who showed APC methylation, 66% had polyps; 33% were polyp free using their blood DNA. There was highly significant association between smoking and alcohol consumption with the presence of a colon polyp (P= .0006 and P= .05, respectively). In conclusion, the lack of the 677 TT may be a significant risk factor for colon neoplasm in the African-American population. Smoking and alcohol consumption were found to be risk factors for colon polyps. APC gene-promoter sequence methylation found in peripheral blood may be an indicator of risk for polyp formation and an important screening tool.


Assuntos
Adenoma/metabolismo , Negro ou Afro-Americano , Pólipos do Colo/metabolismo , Neoplasias Colorretais/metabolismo , Ácido Fólico/metabolismo , Genes APC , Tetra-Hidrofolatos/genética , Adenoma/etnologia , Adenoma/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Consumo de Bebidas Alcoólicas/efeitos adversos , Estudos de Casos e Controles , Pólipos do Colo/etnologia , Pólipos do Colo/genética , Neoplasias Colorretais/etnologia , Neoplasias Colorretais/genética , Metilação de DNA , Feminino , Homocisteína/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Regiões Promotoras Genéticas , Fumar/efeitos adversos , Vitamina B 12/metabolismo , Vitaminas/administração & dosagem
8.
J Med Genet ; 43(8): e41, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16882735

RESUMO

BACKGROUND: Peutz-Jeghers syndrome (PJS) is caused by germline STK11 mutations and characterised by gastrointestinal polyposis. Although small bowel intussusception is a recognised complication of PJS, risk varies between patients. OBJECTIVE: To analyse the time to onset of intussusception in a large series of PJS probands. METHODS: STK11 mutation status was evaluated in 225 PJS probands and medical histories of the patients reviewed. RESULTS: 135 (60%) of the probands possessed a germline STK11 mutation; 109 (48%) probands had a history of intussusception at a median age of 15.0 years but with wide variability (range 3.7 to 45.4 years). Median time to onset of intussusception was not significantly different between those with identified mutations and those with no mutation detected, at 14.7 years and 16.4 years, respectively (log-rank test of difference, chi(2) = 0.58, with 1df; p = 0.45). Similarly no differences were observed between patient groups on the basis of the type or site of STK11 mutation. CONCLUSIONS: The risk of intussusception in PJS is not influenced by STK11 mutation status.


Assuntos
Intussuscepção/genética , Síndrome de Peutz-Jeghers/genética , Proteínas Serina-Treonina Quinases/genética , Quinases Proteína-Quinases Ativadas por AMP , Adolescente , Adulto , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fatores de Risco
9.
Gut ; 55(1): 1-5, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16344569

RESUMO

Germline mutations in LKB1 cause the rare cancer prone disorder Peutz-Jeghers syndrome (PJS). Gastrointestinal hamartomatous polyps constitute the major phenotypic trait in PJS. Hamartomatous polyps arising in PJS patients are generally considered to lack premalignant potential although rare neoplastic changes in these polyps and an increased gastrointestinal cancer risk in PJS are well documented. These conflicting observations are resolved in the current hypothesis by providing a unifying explanation for these contrasting features of PJS polyposis. We postulate that a genetic predisposition to epithelial prolapse underlies the formation of the polyps associated with PJS. Conventional sporadic adenomas arising in PJS patients will similarly show mucosal prolapse and carry the associated histological features.


Assuntos
Mucosa Intestinal/patologia , Síndrome de Peutz-Jeghers/genética , Quinases Proteína-Quinases Ativadas por AMP , Predisposição Genética para Doença , Mutação em Linhagem Germinativa , Humanos , Prolapso , Proteínas Serina-Treonina Quinases/genética
11.
J Med Genet ; 41(7): 484-91, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15235019

RESUMO

BACKGROUND: Juvenile polyposis (JP) is an autosomal dominant syndrome predisposing to colorectal and gastric cancer. We have identified mutations in two genes causing JP, MADH4 and bone morphogenetic protein receptor 1A (BMPR1A): both are involved in bone morphogenetic protein (BMP) mediated signalling and are members of the TGF-beta superfamily. This study determined the prevalence of mutations in MADH4 and BMPR1A, as well as three other BMP/activin pathway candidate genes in a large number of JP patients. METHODS: DNA was extracted from the blood of JP patients and used for PCR amplification of each exon of these five genes, using primers flanking each intron-exon boundary. Mutations were determined by comparison to wild type sequences using sequence analysis software. A total of 77 JP cases were sequenced for mutations in the MADH4, BMPR1A, BMPR1B, BMPR2, and/or ACVR1 (activin A receptor) genes. The latter three genes were analysed when MADH4 and BMPR1A sequencing found no mutations. RESULTS: Germline MADH4 mutations were found in 14 cases (18.2%) and BMPR1A mutations in 16 cases (20.8%). No mutations were found in BMPR1B, BMPR2, or ACVR1 in 32 MADH4 and BMPR1A mutation negative cases. DISCUSSION: In the largest series of JP patients reported to date, the prevalence of germline MADH4 and BMPR1A mutations is approximately 20% for each gene. Since mutations were not found in more than half the JP patients, either additional genes predisposing to JP remain to be discovered, or alternate means of inactivation of the two known genes are responsible for these JP cases.


Assuntos
Polipose Adenomatosa do Colo/epidemiologia , Polipose Adenomatosa do Colo/genética , Neoplasias do Colo/epidemiologia , Neoplasias do Colo/genética , Proteínas de Ligação a DNA/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas/genética , Receptores de Fatores de Crescimento/genética , Transativadores/genética , Receptores de Ativinas Tipo I , Receptores de Proteínas Morfogenéticas Ósseas Tipo I , Receptores de Proteínas Morfogenéticas Ósseas Tipo II , Éxons/genética , Humanos , Epidemiologia Molecular/métodos , Prevalência , Proteína Smad4
12.
Br J Cancer ; 90(1): 224-9, 2004 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-14710233

RESUMO

Nonsteroidal anti-inflammatory drugs (NSAIDs) have chemopreventive potential against colorectal carcinomas (CRCs). Inhibition of cyclooxygenase (COX)-2 underlies part of this effect, although COX-2-independent mechanisms may also exist. Nonsteroidal anti-inflammatory drugs appear to inhibit the initial stages of the adenoma-carcinoma sequence, suggesting a link to the APC/beta-catenin/TCF pathway (Wnt-signalling pathway). Therefore, the effect of the NSAID sulindac on nuclear (nonphosphorylated) beta-catenin and beta-catenin/TCF-mediated transcription was investigated. Nuclear beta-catenin expression was assessed in pretreatment colorectal adenomas and in adenomas after treatment with sulindac from five patients with familial adenomatous polyposis (FAP). Also, the effect of sulindac sulphide on beta-catenin/TCF-mediated transcription was studied. Adenomas of FAP patients collected after treatment with sulindac for up to 6 months showed less nuclear beta-catenin expression compared to pretreatment adenomas of the same patients. Sulindac sulphide abrogated beta-catenin/TCF-mediated transcription in the CRC cell lines DLD1 and SW480, and decreased the levels of nonphosphorylated beta-catenin. As a result, the protein levels of the positively regulated TCF targets Met and cyclin D1 were downregulated after sulindac treatment. This study provides in vivo and in vitro evidence that nuclear beta-catenin localisation and beta-catenin/TCF-regulated transcription of target genes can be inhibited by sulindac. The inhibition of Wnt-signalling provides an explanation for the COX-2-independent mechanism of chemoprevention by NSAIDs.


Assuntos
Adenoma/genética , Polipose Adenomatosa do Colo/genética , Anti-Inflamatórios não Esteroides/farmacologia , Transformação Celular Neoplásica , Neoplasias Colorretais/genética , Proteínas Proto-Oncogênicas/fisiologia , Sulindaco/farmacologia , Proteínas de Peixe-Zebra , Adenoma/fisiopatologia , Polipose Adenomatosa do Colo/fisiopatologia , Adulto , Núcleo Celular , Quimioprevenção , Neoplasias Colorretais/fisiopatologia , Proteínas do Citoesqueleto , Feminino , Humanos , Mitógenos , Transdução de Sinais , Transativadores , Transcrição Gênica , Células Tumorais Cultivadas , Proteínas Wnt , beta Catenina
13.
Gut ; 52(6): 903-4, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12740351

RESUMO

The risk of periampullary cancer in patients with classic familial adenomatous polyposis (FAP) is significantly increased compared with the general population. However, the incidence of this extracolonic manifestation in attenuated FAP (AFAP) is unknown. We report the case of a 38 year old woman with no known family history of polyposis or colorectal cancer, who presented with ampullary adenocarcinoma. Diagnosis of AFAP was made only after evaluation of the patient's extended family history and genetic testing. This case report suggests that AFAP should be included in the differential diagnosis of patients with ampullary/duodenal tumours.


Assuntos
Adenocarcinoma/genética , Polipose Adenomatosa do Colo/genética , Ampola Hepatopancreática , Neoplasias do Ducto Colédoco/genética , Polipose Adenomatosa do Colo/diagnóstico , Adulto , Feminino , Humanos , Linhagem
14.
Ann Surg Oncol ; 9(9): 901-6, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12417513

RESUMO

BACKGROUND: Juvenile polyposis (JP) is an inherited condition predisposing to upper gastrointestinal (UGI) polyps and colorectal cancer. Two genes are known to predispose to JP, SMAD4 and bone morphogenetic protein receptor type 1A (BMPR1A). The object of this study was to determine the differences in phenotype of patients with SMAD4 or BMPR1A mutations (MUT+) compared with those without (MUT-). METHODS: DNA was extracted from 54 JP probands and used for polymerase chain reaction of all exons of SMAD4 and BMPR1A. Products were then sequenced and analyzed for mutations. Medical record data were used to create a JP database, and statistical analysis was performed using Fisher's exact and unpaired t-tests. RESULTS: Nine of 54 patients had germline SMAD4 mutations, 13 had BMPR1A mutations, and 32 had neither. There were no significant differences between SMAD4+ and BMPR1A+ cases in terms of clinical factors examined, except for a family history of UGI involvement (P <.01). There was a higher prevalence of familial cases in MUT+ patients (P =.09), >10 lower gastrointestinal polyps (P =.06), and frequency of family history of gastrointestinal cancer compared with MUT- patients (P =.01). CONCLUSIONS: Patients with germline SMAD4 or BMPR1A mutations have a more prominent JP phenotype than those without, and SMAD4 mutations predispose to UGI polyposis.


Assuntos
Proteínas de Ligação a DNA/genética , Genes Supressores de Tumor , Mutação em Linhagem Germinativa , Pólipos Intestinais/genética , Proteínas Serina-Treonina Quinases/genética , Receptores de Fatores de Crescimento/genética , Transativadores/genética , Adolescente , Receptores de Proteínas Morfogenéticas Ósseas Tipo I , Criança , Análise Mutacional de DNA , Predisposição Genética para Doença , Humanos , Fenótipo , Proteína Smad4
15.
Aliment Pharmacol Ther ; 16(11): 1843-57, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12390093

RESUMO

Colorectal cancer is the second leading cause of cancer death, after lung cancer, in the USA. The great majority (80%) of patients with colorectal cancer have sporadic disease with no evidence of having inherited the disorder. In the remaining 20%, a potentially definable genetic component exists. With the discovery of gene mutations related to hereditary colorectal cancer, risk assessment based on genetic test results is now feasible. The following review focuses on the two well-described colorectal cancer genetic syndromes-familial adenomatous polyposis and hereditary nonpolyposis colorectal cancer, the process of genetic counselling, currently available genetic tests, and indications for their use.


Assuntos
Polipose Adenomatosa do Colo/diagnóstico , Neoplasias Colorretais Hereditárias sem Polipose/diagnóstico , Aconselhamento Genético/métodos , Testes Genéticos/métodos , Polipose Adenomatosa do Colo/genética , Neoplasias Colorretais Hereditárias sem Polipose/genética , Predisposição Genética para Doença , Humanos , Mutação
17.
J Clin Pathol ; 55(5): 340-5, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11986336

RESUMO

BACKGROUND: Peptic ulcer surgery may carry an increased risk for pancreatic cancer development. Molecular analysis of K-ras codon 12, frequently mutated in conventional pancreatic cancers, might provide insight into the aetiological mechanisms. METHODS: The relative risk of pancreatic cancer was computed by multivariate and person-year analysis in a cohort of 2633 patients who had undergone gastrectomy. Lung cancer risk was analysed as an indirect means of assessing smoking behaviour. K-ras codon 12 mutational analysis was performed on 15 postgastrectomy pancreatic cancers. RESULTS: There was an overall increased risk of pancreatic carcinoma of 1.8 (95% confidence interval, 1.3 to 2.6) five to 59 years postoperatively, which gradually increased to 3.6 at 35 years or more after surgery (chi(2) test for trend, p < 0.05). Multivariate analysis indicated that parameters other than postoperative interval did not influence the risk. Lung cancer risk was significantly increased after surgery, but no time trend was observed. The spectrum and prevalence of K-ras codon 12 mutations were comparable to conventional pancreatic cancer. CONCLUSIONS: Remote partial gastrectomy is associated with an increased risk of pancreatic cancer. Postgastrectomy and non-postgastrectomy pancreatic cancers may share similar aetiological factors, such as smoking. However, the neoplastic process in patients who have undergone gastrectomy appears to be accelerated by factors related to the surgery itself.


Assuntos
Neoplasias Pancreáticas/etiologia , Úlcera Péptica/cirurgia , Complicações Pós-Operatórias/etiologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Estudos de Coortes , Úlcera Duodenal/cirurgia , Feminino , Genes ras , Humanos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Mutação/genética , Países Baixos/epidemiologia , Neoplasias Pancreáticas/mortalidade , Complicações Pós-Operatórias/mortalidade , Período Pós-Operatório , Medição de Risco , Fatores de Risco , Úlcera Gástrica/cirurgia
18.
Clin Cancer Res ; 7(12): 4000-7, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11751493

RESUMO

PURPOSE: Sulindac causes the reduction of adenomas in familial adenomatous polyposis (FAP) patients, but complete regression is unusual, and breakthrough of colorectal carcinoma during sulindac treatment has been described. The molecular features related to sulindac resistance are unknown. Therefore, we investigated molecular alterations in adenomas from FAP patients with complete adenoma regression on sulindac (responsive patients) and from FAP patients with sulindac-resistant adenomas (resistant patients). DESIGN: Fourteen baseline adenomas (removed before sulindac treatment) from six responsive patients were studied. Also, 9 baseline adenomas and 34 resistant adenomas (removed during sulindac treatment) from three resistant patients were analyzed. Using immunohistochemistry, we evaluated the expression of beta-catenin, cyclooxygenase-2 (Cox-2), p53, Bcl-2, and Bax. K-ras codon 12 mutations, loss of heterozygosity at 5q (APC locus), and microsatellite instability were studied with PCR-based techniques. RESULTS: There were no significant differences between baseline adenomas from sulindac-responsive and -resistant patients (P > 0.05). There was less loss of membranous beta-catenin staining and less nuclear beta-catenin accumulation in resistant adenomas compared with baseline adenomas from the same (sulindac-resistant) patients (P < 0.01) or baseline adenomas from responsive patients (P < 0.01). Epithelial Cox-2 expression was less, though not significant, in resistant adenomas compared with baseline adenomas from resistant patients, but was significantly less in baseline adenomas from responsive patients (P < 0.01). K-ras mutations were found in 8 of 34 resistant adenomas (24%) and in none of the baseline adenomas (P < 0.05). Stromal Cox-2 expression, staining of p53 and Bcl-2, and loss of heterozygosity at 5q were comparable in both groups. Loss of Bax staining and microsatellite instability were not found in any adenoma. CONCLUSIONS: Sulindac-resistant adenomas display less alteration in beta-catenin staining and less epithelial Cox-2 expression when compared with adenomas removed before sulindac treatment. K-ras mutations may contribute to sulindac-resistance. Continued research is needed to investigate molecular alterations related to sulindac resistance.


Assuntos
Adenoma/tratamento farmacológico , Polipose Adenomatosa do Colo/tratamento farmacológico , Antineoplásicos/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Sulindaco/uso terapêutico , Transativadores , Adenoma/genética , Adenoma/patologia , Adenoma/cirurgia , Polipose Adenomatosa do Colo/genética , Polipose Adenomatosa do Colo/patologia , Polipose Adenomatosa do Colo/cirurgia , Apoptose , Biomarcadores Tumorais/análise , Códon , Ciclo-Oxigenase 2 , Proteínas do Citoesqueleto/análise , Genes ras , Humanos , Imuno-Histoquímica , Isoenzimas/análise , Perda de Heterozigosidade , Proteínas de Membrana , Mutação , Prostaglandina-Endoperóxido Sintases/análise , Proteínas Proto-Oncogênicas/análise , Proteínas Proto-Oncogênicas c-bcl-2/análise , Neoplasias Retais/tratamento farmacológico , Proteína Supressora de Tumor p53/análise , Proteína X Associada a bcl-2 , beta Catenina
19.
Gastroenterology ; 121(1): 198-213, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11438509

RESUMO

This literature review and the recommendations therein were prepared for the American Gastroenterological Association (AGA) Clinical Practice and Practice Economics Committee. The paper was approved by the Committee on March 20, 2001, and by the AGA Governing Board on April 18, 2001.


Assuntos
Polipose Adenomatosa do Colo/genética , Neoplasias Colorretais Hereditárias sem Polipose/genética , Testes Genéticos/métodos , Polipose Adenomatosa do Colo/diagnóstico , Neoplasias Colorretais Hereditárias sem Polipose/diagnóstico , Família , Feminino , Testes Genéticos/economia , Genótipo , Humanos , Masculino , Mutação , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA