Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
Add more filters











Publication year range
1.
Ann Oncol ; 28(8): 1869-1875, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28460000

ABSTRACT

BACKGROUND: We assessed the novel MACC1 gene to further stratify stage II colon cancer patients with proficient mismatch repair (pMMR). PATIENTS AND METHODS: Four cohorts with 596 patients were analyzed: Charité 1 discovery cohort was assayed for MACC1 mRNA expression and MMR in cryo-preserved tumors. Charité 2 comparison cohort was used to translate MACC1 qRT-PCR analyses to FFPE samples. In the BIOGRID 1 training cohort MACC1 mRNA levels were related to MACC1 protein levels from immunohistochemistry in FFPE sections; also analyzed for MMR. Chemotherapy-naïve pMMR patients were stratified by MACC1 mRNA and protein expression to establish risk groups based on recurrence-free survival (RFS). Risk stratification from BIOGRID 1 was confirmed in the BIOGRID 2 validation cohort. Pooled BIOGRID datasets produced a best effect-size estimate. RESULTS: In BIOGRID 1, using qRT-PCR and immunohistochemistry for MACC1 detection, pMMR/MACC1-low patients had a lower recurrence probability versus pMMR/MACC1-high patients (5-year RFS of 92% and 67% versus 100% and 68%, respectively). In BIOGRID 2, longer RFS was confirmed for pMMR/MACC1-low versus pMMR/MACC1-high patients (5-year RFS of 100% versus 90%, respectively). In the pooled dataset, 6.5% of patients were pMMR/MACC1-low with no disease recurrence, resulting in a 17% higher 5-year RFS [95% confidence interval (CI) (12.6%-21.3%)] versus pMMR/MACC1-high patients (P = 0.037). Outcomes were similar for pMMR/MACC1-low and deficient MMR (dMMR) patients (5-year RFS of 100% and 96%, respectively). CONCLUSIONS: MACC1 expression stratifies colon cancer patients with unfavorable pMMR status. Stage II colon cancer patients with pMMR/MACC1-low tumors have a similar favorable prognosis to those with dMMR with potential implications for the role of adjuvant therapy.


Subject(s)
Colonic Neoplasms/pathology , DNA Mismatch Repair , Neoplasm Recurrence, Local/genetics , Transcription Factors/genetics , Cohort Studies , Colonic Neoplasms/genetics , Disease-Free Survival , Humans , Neoplasm Staging , Prognosis , Risk Factors , Trans-Activators
2.
Oncogene ; 35(19): 2475-84, 2016 05 12.
Article in English | MEDLINE | ID: mdl-26300002

ABSTRACT

Transcription factor Myb is overexpressed in most colorectal cancers (CRC). Patients with CRC expressing the highest Myb are more likely to relapse. We previously showed that mono-allelic loss of Myb in an Adenomatous polyposis coli (APC)-driven CRC mouse model (Apc(Min/+)) significantly improves survival. Here we directly investigated the association of Myb with poor prognosis and how Myb co-operates with tumor suppressor genes (TSGs) (Apc) and cell cycle regulator, p27. Here we generated the first intestinal-specific, inducible transgenic model; a MybER transgene encoding a tamoxifen-inducible fusion protein between Myb and the estrogen receptor-α ligand-binding domain driven by the intestinal-specific promoter, Gpa33. This was to mimic human CRC with constitutive Myb activity in a highly tractable mouse model. We confirmed that the transgene was faithfully expressed and inducible in intestinal stem cells (ISCs) before embarking on carcinogenesis studies. Activation of the MybER did not change colon homeostasis unless one p27 allele was lost. We then established that MybER activation during CRC initiation using a pro-carcinogen treatment, azoxymethane (AOM), augmented most measured aspects of ISC gene expression and function and accelerated tumorigenesis in mice. CRC-associated symptoms of patients including intestinal bleeding and anaemia were faithfully mimicked in AOM-treated MybER transgenic mice and implicated hypoxia and vessel leakage identifying an additional pathogenic role for Myb. Collectively, the results suggest that Myb expands the ISC pool within which CRC is initiated while co-operating with TSG loss. Myb further exacerbates CRC pathology partly explaining why high MYB is a predictor of worse patient outcome.


Subject(s)
Carcinogenesis , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Intestinal Mucosa/metabolism , Intestines/pathology , Proto-Oncogene Proteins c-myb/metabolism , Animals , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Mice , Mice, Transgenic , Organ Specificity , Stem Cells/pathology , Tumor Hypoxia , Vascular Endothelial Growth Factor A/genetics
3.
Cell Death Differ ; 22(5): 767-78, 2015 May.
Article in English | MEDLINE | ID: mdl-25361085

ABSTRACT

FASL/FAS signaling imposes a critical barrier against autoimmune disease and lymphadenopathy. Mutant mice unable to produce membrane-bound FASL (FasL(Δm/Δm)), a prerequisite for FAS-induced apoptosis, develop lymphadenopathy and systemic autoimmune disease with immune complex-mediated glomerulonephritis. Prior to disease onset, FasL(Δm/Δm) mice contain abnormally high numbers of leukocytes displaying activated and elevated NF-κB-regulated cytokine levels, indicating that NF-κB-dependent inflammation may be a key pathological driver in this multifaceted autoimmune disease. We tested this hypothesis by genetically impairing canonical or non-canonical NF-κB signaling in FasL(Δm/Δm) mice by deleting the c-Rel or NF-κB2 genes, respectively. Although the loss of NF-κB2 reduced the levels of inflammatory cytokines and autoantibodies, the impact on animal survival was minor due to substantially accelerated and exacerbated lymphoproliferative disease. In contrast, a marked increase in lifespan resulting from the loss of c-REL coincided with a striking reduction in classical parameters of autoimmune pathology, including the levels of cytokines and antinuclear autoantibodies. Notably, the decrease in regulatory T-cell numbers associated with loss of c-REL did not exacerbate autoimmunity in FasL(Δm/Δm)c-rel(-/-) mice. These findings indicate that selective inhibition of c-REL may be an attractive strategy for the treatment of autoimmune pathologies driven by defects in FASL/FAS signaling that would be expected to circumvent many of the complications caused by pan-NF-κB inhibition.


Subject(s)
Autoimmune Diseases/metabolism , Fas Ligand Protein/metabolism , Mutation , NF-kappa B p52 Subunit/metabolism , Proto-Oncogene Proteins c-rel/metabolism , fas Receptor/metabolism , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Autoimmune Diseases/prevention & control , Fas Ligand Protein/genetics , Mice , Mice, Knockout , NF-kappa B p52 Subunit/genetics , Proto-Oncogene Proteins c-rel/genetics , Signal Transduction/genetics , fas Receptor/genetics
4.
Pathology ; 44(2): 89-98, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22198256

ABSTRACT

Genes have been identified for which germline mutations are associated with high lifetime risks of breast, colorectal and other cancers. Identification of mutation carriers through genetic testing is important as it could help lower cancer incidence and mortality. The translation of genetic information into better health outcomes is expensive because of the costs of genetic counselling as well as laboratory testing. Approaches to triage for mutation screening of known genes which rely on cancer family history are not necessarily sensitive and specific or the most cost-effective. Recent population-based research has shown that the cancers and precancerous lesions arising in mutation carriers have specific molecular and morphological characteristics. People with colorectal cancer, especially those diagnosed at a young age, whose tumours exhibit microsatellite instability and some specific pathology and immunohistochemically-defined features are more likely to carry a germline mutation in one of four mismatch repair genes. Some morphological and immunohistochemically-defined features are associated with breast cancers arising in women who carry BRCA1 or BRCA2 germline mutations, especially if at a young age. Screening paradigms based on molecular and morphological features that predict mutation status, especially if focused on early-onset disease, have the potential to identify mutation carriers with greater sensitivity and specificity, and in a more cost-effective way, than those based on family history alone. Genetic testing results could help inform treatment if those affected are tested soon after diagnosis using pathology-led selection strategies to identify cases most likely to carry germline mutations. We propose how this new approach could be undertaken by having genetic testing and counselling prioritised to those with the greatest probability of carrying a germline mutation in these known cancer predisposition genes.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Genetic Predisposition to Disease , Female , Germ-Line Mutation , Humans , Male
5.
Cell Death Differ ; 19(4): 623-32, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21997189

ABSTRACT

The tumour suppressor p53 transcriptionally regulates a range of target genes that control cell growth and survival. Mutations of p53 have been implicated in the development of approximately 50% of human cancers, including those instigated by exposure to mutagens. Although numerically rare, cancers can arise as a consequence of inherited mutations, such as in the Li-Fraumeni syndrome, which is caused by mutation of one p53 allele. Gene-targeted mice deficient for p53 have been generated to study this familial cancer syndrome. On a C57BL/6 background, p53-deficient mice develop primarily thymic lymphoma and more rarely sarcoma. Evasion of apoptosis is considered to be essential for neoplastic transformation. As proteins of the Bcl-2 family are the critical regulators of apoptosis, we investigated the role of the pro-survival members Bcl-2, Bcl-x(L) and Bcl-w in cancer development in p53(+/-) and p53(-/-) mice by testing whether ABT-737, a pharmacological inhibitor of these proteins, could prevent or delay tumourigenesis. Our studies showed that ABT-737 prophylaxis only caused a minor delay and reduction in γ-radiation-induced thymic lymphoma development in p53(-/-) mice, but this was accompanied by a concomitant increase in sarcoma. These data show that, collectively, Bcl-2, Bcl-x(L) and Bcl-w have only minor roles in thymic lymphoma development elicited by defects in p53, and this may indicate that Mcl-1 and/or A1 may feature more prominently in this process.


Subject(s)
Biomimetic Materials/pharmacology , Biphenyl Compounds/pharmacology , Lymphoma/metabolism , Neoplasms, Radiation-Induced/metabolism , Neoplastic Syndromes, Hereditary/metabolism , Nitrophenols/pharmacology , Proteins/antagonists & inhibitors , Sulfonamides/pharmacology , Thymus Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , bcl-X Protein/antagonists & inhibitors , Alleles , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins , Humans , Lymphoma/genetics , Lymphoma/prevention & control , Mice , Mice, Knockout , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasms, Radiation-Induced/genetics , Neoplasms, Radiation-Induced/prevention & control , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/prevention & control , Piperazines/pharmacology , Proteins/genetics , Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2 , Thymus Neoplasms/genetics , Thymus Neoplasms/prevention & control , Tumor Suppressor Protein p53/genetics , bcl-X Protein/genetics , bcl-X Protein/metabolism
6.
Fam Cancer ; 6(3): 301-10, 2007.
Article in English | MEDLINE | ID: mdl-17453358

ABSTRACT

Patients suspected on clinical grounds to have hereditary non-polyposis colorectal cancer (HNPCC) may be offered laboratory testing in order to confirm the diagnosis and to facilitate screening of pre-symptomatic family members. Tumours from an affected family member are usually pre-screened for microsatellite instability (MSI) and/or loss of immunohistochemical expression of mismatch repair (MMR) genes prior to germline MMR gene mutation testing. The efficiency of this triage process is compromised by the more frequent occurrence of sporadic colorectal cancer (CRC) showing high levels of MSI (MSI-H) due to epigenetic loss of MLH1 expression. Somatic BRAF mutations, most frequently V600E, have been described in a significant proportion of sporadic MSI-H CRC but not in HNPCC-associated cancers. BRAF mutation testing has therefore been proposed as a means to more definitively identify and exclude sporadic MSI-H CRC cases from germline MMR gene testing. However, the clinical validity and utility of this approach have not been previously evaluated in a familial cancer clinic setting. Testing for the V600E mutation was performed on MSI-H CRC samples from 68 individuals referred for laboratory investigation of suspected HNPCC. The V600E mutation was identified in 17 of 40 (42%) tumours showing loss of MLH1 protein expression by immunohistochemistry but in none of the 28 tumours that exhibited loss of MSH2 expression (P < 0.001). The assay was negative in all patients with an identified germline MMR gene mutation. Although biased by the fact that germline testing was not pursued beyond direct sequencing in many cases lacking a high clinical index of suspicion of HNPCC, BRAF V600E detection was therefore considered to be 100% specific and 48% sensitive in detecting sporadic MSI-H CRC amongst those cases showing loss of MLH1 protein expression, in a population of patients with MSI-H CRC and clinical features suggestive of HNPCC. Accordingly, we recommend the incorporation of BRAF V600E mutation testing into the laboratory algorithm for pre-screening patients with suspected HNPCC, whose CRCs show loss of expression of MLH1. In such tumours, the presence of a BRAF V600E mutation indicates the tumour is not related to HNPCC and that germline testing of MLH1 in that individual is not warranted. We also recommend that in families where the clinical suspicion of HNPCC is high, germline testing should not be performed on an individual whose CRC harbours a somatic BRAF mutation, as this may compromise identification of the familial mutation.


Subject(s)
Algorithms , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Genetic Testing , Proto-Oncogene Proteins B-raf/genetics , Adaptor Proteins, Signal Transducing/genetics , Adolescent , Adult , Alleles , Base Sequence , DNA Mutational Analysis , Female , Humans , Male , Microsatellite Instability , Middle Aged , Molecular Sequence Data , MutL Protein Homolog 1 , MutS Homolog 2 Protein/genetics , Mutation , Nuclear Proteins/genetics
7.
Oncogene ; 26(27): 4009-17, 2007 Jun 07.
Article in English | MEDLINE | ID: mdl-17173065

ABSTRACT

To identify possible genetic interactions between the mechanisms of tumor suppression of menin and pRb, we intercrossed mice with targeted deletions of Men1 and Rb1, and compared tumor development in cohorts of animals carrying single or dual mutations of these tumor-suppressor genes. In mice lacking one copy of Men1, pancreatic islet and anterior pituitary adenomas are common. In animals lacking one copy of Rb1, intermediate pituitary and thyroid tumors occur at high frequency, with less frequent development of pancreatic islet hyperplasia and parathyroid lesions. In mice heterozygous for both Men1 and Rb1, pancreatic hyperplasia and tumors of the intermediate pituitary and thyroid occurred at high frequency. Serum measurements of calcium and glucose did not vary significantly between genotypic groups. Loss of heterozygosity at the Rb1 locus was common in pituitary and thyroid tumors, whereas loss of menin was observed in pancreatic and parathyroid lesions. The tumor spectrum in the double heterozygotes was a combination of pathologies seen in each of the individual heterozygotes, without decrease in age of onset, indicating independent, non-additive effects of the two mutations. Together with the lack of increased tumor spectrum, this suggests that menin and pRb function in a common pathway of tumor suppression.


Subject(s)
Neoplasms/pathology , Proto-Oncogene Proteins/physiology , Retinoblastoma Protein/physiology , Animals , Genotype , Heterozygote , Immunohistochemistry , Loss of Heterozygosity , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/genetics , Pancreas/metabolism , Pancreas/pathology , Pituitary Gland/metabolism , Pituitary Gland/pathology , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/genetics , Retinoblastoma Protein/genetics , Severity of Illness Index , Thyroid Gland/metabolism , Thyroid Gland/pathology
8.
Histopathology ; 49(5): 523-32, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17064299

ABSTRACT

AIMS: Germline variants in the ataxia telangiectasia mutated (ATM) gene have been implicated in increased breast cancer risk. The aim of this study was to determine whether the histopathology of breast cancers occurring in ATM variant carriers is distinctive or resembles the described BRCA1 mutation-associated phenotype. METHODS: The histopathological features of breast cancers occurring in ATM variant carriers from multiple-case breast cancer families were compared with matched controls. The test group included 21 cases of in situ and/or invasive cancer from carriers of either the IVS10-6T-->G, 2424V-->G or 1420L-->F ATM variants in the absence of BRCA1 or BRCA2 mutations. An additional four invasive cancers from carriers of a pathogenic BRCA1 mutation in the context of a familial ATM variant were also examined. RESULTS: The histopathology of breast cancers in ATM variant-only carriers was not significantly different from controls and known features of BRCA1 mutation-associated cancer were rarely seen. In contrast, these features were prominent in the small group of cases with a pathogenic BRCA1 mutation. CONCLUSIONS: Breast cancer occurring in carriers of ATM variants is not associated with distinctive histopathological features and does not resemble the tumour phenotype commonly observed in BRCA1 mutation carriers.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Genetic Predisposition to Disease , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics , Adult , Aged , Ataxia Telangiectasia Mutated Proteins , Biomarkers, Tumor/metabolism , Carcinoma, Ductal, Breast/genetics , Carcinoma, Intraductal, Noninfiltrating/genetics , Cohort Studies , Female , Genes, BRCA1 , Genes, BRCA2 , Genetic Carrier Screening , Germ-Line Mutation/genetics , Humans , Middle Aged , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism
9.
Histopathology ; 49(1): 52-65, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16842246

ABSTRACT

AIMS: With the availability of effective but expensive treatment in the form of imatinib, accurate diagnosis of gastrointestinal stromal tumour (GIST) is extremely important. The aims of this study were: to describe the clinicopathological, immunohistochemical and molecular features of cases referred to a cancer centre with a possible diagnosis of GIST; to identify pitfalls in the performance and interpretation of KIT immunohistochemistry; to define the role of KIT mutation testing in making a diagnosis of GIST. METHODS AND RESULTS: Morphological review, KIT immunohistochemistry and mutation testing were performed on all cases referred with a diagnosis of GIST or where the diagnosis was under serious consideration on the basis of KIT immunopositivity with a view to treating with imatinib. Thirty-seven cases met the inclusion criteria. Of these, 26 were classified as GIST and 11 as non-GIST. Most GISTs showed strong diffuse membranous, cytoplasmic or paranuclear KIT immunopositivity. Some non-GISTs demonstrated patchy cytoplasmic KIT immunopositivity related to the immunohistochemical protocol used in the external laboratory, which led to erroneous diagnoses of GIST in nine (24%) cases. KIT mutations involving exons 11 or 9 were identified in 22 (88%) GISTs tested and none of the non-GISTs. CONCLUSIONS: An accurate diagnosis of GIST can be made on clinicopathological and immunohistochemical criteria without the need for mutational analysis in most cases, provided proper attention is paid to the immunohistochemical protocol used and, most importantly, control material. False-positive diagnoses of GIST potentially leading to inappropriate treatment with imatinib are more common than missed diagnoses.


Subject(s)
Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/metabolism , Mutation , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Antineoplastic Agents/therapeutic use , Base Sequence , Benzamides , DNA Primers/genetics , DNA, Neoplasm/genetics , Female , Gastrointestinal Stromal Tumors/classification , Gastrointestinal Stromal Tumors/diagnosis , Gastrointestinal Stromal Tumors/drug therapy , Humans , Imatinib Mesylate , Immunohistochemistry , Male , Middle Aged , Molecular Sequence Data , Piperazines/therapeutic use , Polymerase Chain Reaction , Pyrimidines/therapeutic use , Receptor, Platelet-Derived Growth Factor alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL