Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
2.
Pharmacotherapy ; 34(11): 1159-66, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25266970

ABSTRACT

STUDY OBJECTIVE: To assess the reproducibility of Naranjo Adverse Drug Reaction Probability Scale (APS) scores in published case reports. DESIGN: Reliability analysis. MEASUREMENTS AND MAIN RESULTS: Randomly selected case reports using the APS were identified from the Web of Science database. The APS scores were blinded from the case reports, and scores were then independently calculated by four raters, using the APS. The percentage of exact agreement between raters' and the published APS scores was calculated for all case reports. Categorical scores were compared by using a weighted κ statistic. For numerical scores, descriptive statistics were computed by using raw and absolute difference scores. Twenty-four case reports were independently scored by four raters. Exact agreement between all raters' scores and the published APS scores was found in five (21%) of the 24 reports. Agreement between individual rater's scores and the published categorical score ranged from 42% to 79%. Weighted κ ranged from 0.12 to 0.61, corresponding to strengths of agreement between poor and good. Difference in scoring by raters resulted in 18% and 27% of case reports being reclassified into higher and lower than reported APS categories, respectively. CONCLUSION: Exact agreement between raters' scores and the published APS score was infrequent. We recommend that authors of case reports include all pertinent details of the case and that journals ensure the robustness of the causality assessment during peer review.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/epidemiology , Probability Theory , Causality , Databases, Factual , Drug-Related Side Effects and Adverse Reactions/physiopathology , Humans , Internet , Medical Records , Periodicals as Topic , Reproducibility of Results , Risk Assessment/methods
3.
Can J Hosp Pharm ; 65(3): 202-8, 2012 May.
Article in English | MEDLINE | ID: mdl-22783031

ABSTRACT

BACKGROUND: Canadian pharmacy residency programs rely on preceptors to support the growing demand of graduates wishing to pursue hospital residencies. Understanding the educational needs of these preceptors is important to ensure that they are well prepared to deliver successful programs. OBJECTIVE: To determine what new and experienced residency preceptors self-identify as learning needs in order to become more effective preceptors for pharmacy residents. METHODS: A needs assessment of preceptors from the 31 accredited Canadian general hospital pharmacy residency programs was conducted. The study had 4 key components: interviews and focus group discussions with key informants, a pilot study, an online survey, and member checking (seeking clarification and further explanation from study participants). The residency coordinators and a convenience sample of 5 preceptors from each program were invited to participate in the survey component. RESULTS: Of a possible 186 participants, 132 (71%) responded to the survey. Of these, 128 (97%) were confident that they met the 2010 standards of the Canadian Hospital Pharmacy Residency Board (CHPRB). Preceptors ranked communication skills, giving effective feedback, and clinical knowledge as the most important elements of being an effective preceptor. Managing workload, performing evaluations, and dealing with difficult residents were commonly reported challenges. Preceptors expressed a preference for interactive workshops and mentorship programs with experienced colleagues when first becoming preceptors, followed by 1-day training sessions or online learning modules every other year for ongoing educational support. The most beneficial support topics selected were providing constructive feedback, practical assessment strategies, small-group teaching strategies, effective communication skills, and setting goals and objectives. CONCLUSIONS: This study identified several learning needs of hospital residency preceptors and showed that preceptors would appreciate educational support. Utilization of these results by residency program administrators, the CHPRB, and faculties of pharmacy could be beneficial for residency programs across Canada.

4.
Gut ; 60(4): 499-508, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21068132

ABSTRACT

BACKGROUND AND AIMS: Although aberrant methylation of key genes in the progression of colorectal neoplasia has been reported, no model-based analysis of the incremental changes through the intermediate adenoma stage has been described. In addition, the biological drivers for these methylation changes have yet to be defined. Linear mixed-effects modelling was used in this study to understand the onset and patterns of the methylation changes of SFRP2, IGF2 DMR0, H19, LINE-1 and a CpG island methylator phenotype (CIMP) marker panel, and they were correlated with DNA methyltransferase 3B (DNMT3B) levels of expression in a sample set representative of colorectal neoplastic progression. METHODS: Methylation of the above CpG islands was measured using quantitative pyrosequencing assays in 261 tissue samples. This included a prospective collection of 44 colectomy specimens with concurrent normal mucosa, adenoma and invasive cancer tissues. Tissue microarrays from a subset of 64 cases were used for immunohistochemical analysis of DNMT3B expression. RESULTS: It is shown that the onset and pattern of methylation changes during colorectal neoplastic progression are locus dependent. The CIMP marker RUNX3 was the earliest CpG island showing significant change, followed by the CIMP markers NEUROG1 and CACNA1G at the hyperplastic polyp stage. SFRP2 and IGF2 DMR0 showed significant methylation changes at the adenomatous polyp stage, followed by the CIMP markers CDKN2A and hMLH1 at the adenocarcinoma stage. DNMT3B levels of immunohistochemical expression increased significantly (p < 0.001) from normal to hyperplastic and from adenomatous polyps to carcinoma samples. DNMT3B expression correlated positively with SFRP2 methylation (r = 0.42, p < 0.001, 95% CI 0.25 to 0.56), but correlated negatively with IGF2 DMR0 methylation (r = 0.26, p = 0.01, 95% CI -0.45 to -0.05). A subset of the CIMP panel (NEUROG1, CACNA1G and CDKN2A) positively correlated with DNMT3B levels of expression (p < 0.05). CONCLUSION: Hierarchical epigenetic alterations occur at transition points during colorectal neoplastic progression. These cumulative changes are closely correlated with a gain of DNMT3B expression, suggesting a causal relationship.


Subject(s)
Adenocarcinoma/genetics , Colorectal Neoplasms/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , DNA, Neoplasm/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/secondary , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Colon/pathology , Colonic Polyps/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , CpG Islands/genetics , Disease Progression , Female , Humans , Hyperplasia/pathology , Insulin-Like Growth Factor II/genetics , Liver Neoplasms/secondary , Male , Membrane Proteins/genetics , Microsatellite Repeats , Middle Aged , Precancerous Conditions/genetics , DNA Methyltransferase 3B
5.
Proc Natl Acad Sci U S A ; 107(34): 15145-50, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20696900

ABSTRACT

In 100 primary colorectal carcinomas, we demonstrate by array comparative genomic hybridization (aCGH) that 33% show DNA copy number (DCN) loss involving PARK2, the gene encoding PARKIN, the E3 ubiquitin ligase whose deficiency is responsible for a form of autosomal recessive juvenile parkinsonism. PARK2 is located on chromosome 6 (at 6q25-27), a chromosome with one of the lowest overall frequencies of DNA copy number alterations recorded in colorectal cancers. The PARK2 deletions are mostly focal (31% approximately 0.5 Mb on average), heterozygous, and show maximum incidence in exons 3 and 4. As PARK2 lies within FRA6E, a large common fragile site, it has been argued that the observed DCN losses in PARK2 in cancer may represent merely the result of enforced replication of locally vulnerable DNA. However, we show that deficiency in expression of PARK2 is significantly associated with adenomatous polyposis coli (APC) deficiency in human colorectal cancer. Evidence of some PARK2 mutations and promoter hypermethylation is described. PARK2 overexpression inhibits cell proliferation in vitro. Moreover, interbreeding of Park2 heterozygous knockout mice with Apc(Min) mice resulted in a dramatic acceleration of intestinal adenoma development and increased polyp multiplicity. We conclude that PARK2 is a tumor suppressor gene whose haploinsufficiency cooperates with mutant APC in colorectal carcinogenesis.


Subject(s)
Adenomatous Polyposis Coli/genetics , Colorectal Neoplasms/genetics , Gene Deletion , Gene Dosage , Genes, APC , Ubiquitin-Protein Ligases/genetics , Adenomatous Polyposis Coli/etiology , Adenomatous Polyposis Coli/metabolism , Adenomatous Polyposis Coli/pathology , Animals , Base Sequence , Cell Line, Tumor , Cell Proliferation , Chromosomes, Human, Pair 6/genetics , Cocarcinogenesis , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA Methylation , DNA Primers/genetics , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , Genes, Tumor Suppressor , Heterozygote , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Models, Molecular , Mutation , Spectral Karyotyping , Ubiquitin-Protein Ligases/chemistry
6.
Mol Neurobiol ; 42(1): 4-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20552413

ABSTRACT

Apoptosis was a term introduced in 1972 to distinguish a mode of cell death with characteristic morphology and apparently regulated, endogenously driven mechanisms. The effector processes responsible for apoptosis are now mostly well known, involving activation of caspases and Bcl2 family members in response to a wide variety of physiological and injury-induced signals. The factors that lead of the decision to activate apoptosis as opposed to adaptive responses to such signals (e.g. autophagy, cycle arrest, protein synthesis shutoff) are less well understood, but the intranuclear Promyelocytic Leukaemia Body (PML body) may create a local microenvironment in which the audit of DNA damage may occur, informed by the extent of the damage, the adequacy of its repair and other aspects of cell status.


Subject(s)
Apoptosis/physiology , Animals , Caspases/metabolism , Cell Survival , DNA Damage , Enzyme Activation , Humans , Proto-Oncogene Proteins c-bcl-2/metabolism
7.
J Pathol ; 220(3): 338-47, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19911421

ABSTRACT

In a study of 109 colorectal cancers, DNA copy number aberrations were identified by comparative genomic hybridization using a DNA microarray covering the entire genome at an average interval of less than 1 Mbase. Four patterns were revealed by unsupervised clustering analysis, one of them associated with significantly better prognosis than the others. This group contained tumours with short, dispersed, and relatively few regions of copy number gain or loss. The good prognosis of this group was not attributable to the presence of tumours showing microsatellite instability (MSI-H). Supervised methods were employed to determine those genomic regions where copy number alterations correlate significantly with multiple indices of aggressive growth (lymphatic spread, recurrence, and early death). Multivariate analysis identified DNA copy number loss at 18q12.2, harbouring a single gene, BRUNOL4 that encodes the Bruno-like 4 splicing factor, as an independent prognostic indicator. The data show that the different patterns of DNA copy number alterations in primary tumours reveal prognostic information and can aid identification of novel prognosis-associated genes.


Subject(s)
Colorectal Neoplasms/genetics , DNA Copy Number Variations , DNA, Neoplasm/genetics , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/pathology , Comparative Genomic Hybridization/methods , Epidemiologic Methods , Female , Humans , Lymphatic Metastasis , Male , Microsatellite Instability , Middle Aged , Neoplasm Staging , Prognosis , Recurrence
10.
J Proteome Res ; 6(5): 2027-32, 2007 May.
Article in English | MEDLINE | ID: mdl-17419608

ABSTRACT

The marriage between transducers of cell stress stimuli and their nuclear targets is likely to be achieved in part by some spatial-temporal compartmentalization of the relevant effectors. A candidate compartment for these events is the promyelocytic leukemia nuclear domain (PML-ND), within which are found numerous effectors of damage recognition, repair, and cell death. We predicted that the identification of PML-ND cargo proteins would clarify those biochemical pathways that straddle the recognition of cellular damage and cell fate. We now use mass spectrometry of peptides eluted from PML coprecipitates to demonstrate that the gamma 1 (gamma1) isoform of PLC associates with nuclear PML. Though thought to act primarily in the cytoplasm, we use biochemical fractionation combined with immunocytochemistry to verify the nuclear expression of PLC-gamma1 and its interaction with PML. These are the first data to show an interaction between endogenous levels of a phosphoinositide metabolizing protein and the biophysically labile PML-ND by mass spectrometry and add weight to the view that PML-NDs may act as tumor suppressors by sequestering mitogenic effectors.


Subject(s)
Cell Nucleus/metabolism , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Phospholipase C gamma/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Cell Line , Chromatography, Gel , Co-Repressor Proteins , Humans , Mass Spectrometry , Molecular Chaperones , Neoplasm Proteins/chemistry , Nuclear Proteins/chemistry , Peptides/chemistry , Peptides/metabolism , Phospholipase C gamma/chemistry , Promyelocytic Leukemia Protein , Transcription Factors/chemistry , Tumor Suppressor Proteins/chemistry
11.
Nucleic Acids Res ; 34(20): e136, 2006.
Article in English | MEDLINE | ID: mdl-17041235

ABSTRACT

We describe an optimized microarray method for identifying genome-wide CpG island methylation called microarray-based methylation assessment of single samples (MMASS) which directly compares methylated to unmethylated sequences within a single sample. To improve previous methods we used bioinformatic analysis to predict an optimized combination of methylation-sensitive enzymes that had the highest utility for CpG-island probes and different methods to produce unmethylated representations of test DNA for more sensitive detection of differential methylation by hybridization. Subtraction or methylation-dependent digestion with McrBC was used with optimized (MMASS-v2) or previously described (MMASS-v1, MMASS-sub) methylation-sensitive enzyme combinations and compared with a published McrBC method. Comparison was performed using DNA from the cell line HCT116. We show that the distribution of methylation microarray data is inherently skewed and requires exogenous spiked controls for normalization and that analysis of digestion of methylated and unmethylated control sequences together with linear fit models of replicate data showed superior statistical power for the MMASS-v2 method. Comparison with previous methylation data for HCT116 and validation of CpG islands from PXMP4, SFRP2, DCC, RARB and TSEN2 confirmed the accuracy of MMASS-v2 results. The MMASS-v2 method offers improved sensitivity and statistical power for high-throughput microarray identification of differential methylation.


Subject(s)
CpG Islands , DNA Methylation , Oligonucleotide Array Sequence Analysis/methods , Cell Line, Tumor , Computational Biology , DNA Probes/chemistry , DNA Restriction Enzymes , Genes, Neoplasm , Genomics/methods , Humans , Sequence Analysis, DNA
12.
J Pathol ; 203(4): 877-83, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15258989

ABSTRACT

The polyglutamine diseases are characterized by expansion of triplet CAG repeats that encode polyglutamine tracts in otherwise unrelated proteins. One plausible explanation for the neurodegeneration of these disorders proposes that inclusions of such proteins sequester other significant nuclear proteins in inactive form. The present study shows that PML protein is sequestered by inclusions of the pathogenic mutant form of the polyglutamine protein ataxin-1 and that this sequestration removes from the nucleus the free 0.2-1 microm diameter PML nuclear domains (PML-NDs), together with at least one of their many cargo proteins (Sp100). The present study demonstrates that this sequestration can be effected equally by another nuclear protein, RED, which lacks a polyglutamine tract, but expresses a polar zipper repeat. The sequestered PML-NDs no longer respond to stress signals (heat shock or ionizing radiation) to which they are normally sensitive. In both cases, there is independent evidence that the cells initiate other responses to their injury (nuclear translocation of heat shock protein or generation of gamma-H2AX-rich nuclear foci, respectively). The data thus provide strong evidence that multiple species of nuclear inclusion functionally sequester PML-NDs. This mechanism is likely to distort cellular responses to injury of many different types.


Subject(s)
Cell Nucleus/metabolism , Intranuclear Inclusion Bodies/physiology , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/physiology , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , Nucleoproteins/physiology , Transcription Factors/metabolism , Ataxin-1 , Ataxins , Cell Line , Cell Nucleus/radiation effects , DNA Damage , Gamma Rays , Heat-Shock Proteins/metabolism , Hot Temperature , Humans , Intranuclear Inclusion Bodies/radiation effects , Oxidative Stress/physiology , Promyelocytic Leukemia Protein , Tumor Suppressor Proteins
13.
Oncogene ; 21(49): 7585-92, 2002 Oct 24.
Article in English | MEDLINE | ID: mdl-12386821

ABSTRACT

High-frequency microsatellite instability (MSI-H) results from deficiency in nucleotide mismatch repair. It contributes significantly to carcinogenesis in the human colorectal mucosa. Here we study 41 colorectal and three other HNPCC-related cancers with MSI-H to provide comprehensive information on the mechanisms of inactivation of the two major proteins involved, hMLH1 and hMSH2. Seventeen of the patients had family histories meeting the criteria for Bethesda grades 1, 2 or 3. Of these familial cases, 14 (83%) had early-onset disease, defined on the basis of diagnosis prior to the age of 50, but in three the disease was of late onset (>50 years). A second subset of 20 patients had early onset disease without family history. The remaining seven patients were selected to allow comparisons with sporadic, late-onset disease, the molecular basis of which has been extensively reported elsewhere. We stratified the tumours initially on the basis of hMLH1 or hMSH2 protein deficiency, detected by immunohistochemistry, and then by analysis of germline and somatic mutation, mRNA transcription, loss of heterozygosity (LOH) at the hMLH1 and hMSH2 loci, and methylation status in two regions of the hMLH1 promoter. The functional significance of several of these changes in the MSI-H tumours was confirmed by comparisons with 16 tumours with low-frequency microsatellite instability and 56 tumours with stable microsatellites. As anticipated, patients with family histories usually showed germline mutation of hMSH2 or hMLH1. In many cases the residual normal allele was silenced in their tumours by loss of heterozygosity (LOH). The small subset of late-onset, sporadic cases confirmed the preponderance in this group of biallelic hMLH1 promoter methylation. In the early-onset, apparently sporadic subset there were 11 tumours with hMLH1 deficiency, five with hMSH2 deficiency and four with no detectable abnormality in expression of either protein. These showed a complex mixture of lesions, including germline and somatic mutations, promoter methylation, LOH, suppression of wild-type RNA by as yet undiscovered mechanisms, or no detectable abnormality in any of these parameters. Evidence is presented to indicate that methylation in proximal region of the hMLH1 promoter is a more reliable correlate of transcriptional silencing in colorectal cancers than methylation in upstream region. These observations have significant implications for management of patients with MSI-H tumours.


Subject(s)
Base Pair Mismatch , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms/genetics , DNA-Binding Proteins , Germ-Line Mutation , Adaptor Proteins, Signal Transducing , Base Sequence , Carrier Proteins , DNA Methylation , DNA Primers , Humans , Immunohistochemistry , Loss of Heterozygosity , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Neoplasm Proteins/genetics , Nuclear Proteins , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Transcription, Genetic
14.
Oncogene ; 21(41): 6299-306, 2002 Sep 12.
Article in English | MEDLINE | ID: mdl-12214270

ABSTRACT

In colorectal tumorigenesis, loss of function of the mismatch repair genes is closely associated with genomic instability at the nucleotide level whereas p53 deficiency has been linked with gross chromosomal instability. We have addressed the contribution of these two forms of genetic instability to tumorigenesis using mice mutant for Msh2 and p53. As previously reported, deficiency of both genes leads to rapid lymphomagenesis Here we show that heterozygosity for p53 also markedly reduces survival on an Msh2 null background. We characterized the patterns of genomic instability in a small set of tumours and showed that, as predicted p53 deficiency predisposes to aneuploidy and Msh2 deficiency leads to microsatellite instability (MSI). However, heterozygosity for p53 in the absence of Msh2 resulted in increased MSI and not aneuploidy. This implied role for p53 in modulating MSI was confirmed using a large cohort of primary fibroblast clones. The differences observed were highly significant (P<0.01) in both the fibroblast clones (which all retained p53 functionality) and the tumours, a proportion of which retained p53 functionality. Our results therefore demonstrate a dose sensitive role for p53 in the maintenance of genomic integrity at the nucleotide level.


Subject(s)
DNA Repair , DNA-Binding Proteins , Genes, p53 , Microsatellite Repeats/genetics , Proto-Oncogene Proteins/genetics , Animals , Base Pair Mismatch , Gene Expression Regulation, Neoplastic , Heterozygote , Mice , MutS Homolog 2 Protein , Neoplasms, Experimental/genetics , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/deficiency
15.
J Pathol ; 198(2): 139-41, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12237872

ABSTRACT

E2F1 is a transcription factor involved in both cell cycle progression and apoptosis. Perhaps surprisingly, these two processes are closely related, and the choice between them appears to be made on the basis of the aggregate of signals flowing into the cell at the time. This may be the means whereby normal cells tune their threshold for apoptosis with respect to the availability of external growth factors, so that cells that are supernumerary to the tissue's needs at the time can be immediately deleted. In many tumours, however, the pathways that link E2F1 activity to apoptosis have been interrupted, sometimes at multiple points. Non-small-cell lung carcinoma provides a striking example of this, with the result that expression of E2F1 in these tumours does not correlate with apoptosis but is a good surrogate marker for replicative status. This relationship does not necessarily pertain in other tumour types. Molecules such as E2F1 lie at the core of very significant cell fate decisions, but they are part of a complex matrix of interactions, all of which must be surveyed before interpretation in terms of tumour behaviour is possible. Microarray analysis may provide a way to do this. In the future, however, such interpretations, including predictions of therapeutic response, may be possible through interrogation of the status of a relatively limited number of molecules. Those that preside over critical cellular decision forks (such as the choice between proliferation or death) are good candidates for this role. E2F1 clearly qualifies as one member of this group.


Subject(s)
Apoptosis/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Cell Cycle Proteins/physiology , DNA-Binding Proteins , Lung Neoplasms/genetics , Transcription Factors/physiology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle/genetics , E2F Transcription Factors , E2F1 Transcription Factor , Humans , Lung Neoplasms/pathology , Neoplasm Proteins/physiology
16.
Int J Cancer ; 98(6): 844-50, 2002 Apr 20.
Article in English | MEDLINE | ID: mdl-11948461

ABSTRACT

It is well established that microsatellite instability (MSI), the hallmark of defective DNA mismatch repair (MMR), is associated with prolonged survival in colorectal cancer compared with tumours that are microsatellite stable (MSS). MSI in sporadic colorectal tumours is primarily due to epigenetic silencing of MLH1. However, there are no prospective population-based studies of survival in patients with germline MMR gene mutations who develop cancer. Although MSI is almost universal in tumours from HNPCC family members, there is a potential confounding effect of ascertainment and other biases that could explain the apparent survival benefit in HNPCC families. Resolving whether germline MMR gene mutations impact on survival is important because it potentially undermines the rationale for surveillance of mutation carriers. Here, we report an investigation of the influence of MSI on survival in cohorts of cancer patients (aged < 30 years at diagnosis, n = 118; non-age-selected, n = 181) in the context of clinicopathologic variables. There was a substantial age-related influence of tumour MSI status on survival. In young patients with tumour MSI, 65% of patients with MSI tumours had germline MSH2 or MLH1 mutations. Clinicopathologic variables and tumour MSI of the cohort were studied with respect to survival and compared with control groups. Young patients had excess MSI tumours (p < 0.000001), mucinous tumours (p < 0.01), advanced disease (p approximately 0.001) and poorer 5-year survival compared with older cases. Cox proportional hazard analysis identified Dukes' stage, age at diagnosis and calendar year of treatment as independent predictors of survival. There was no detectable association between tumour MSI and survival in young patients, although we confirmed previous observations that MSI is associated with better prognosis in later onset cohorts. These findings underscore the rationale for surveillance and early identification of tumours in MMR gene carriers as well as refining understanding of the influence of MSI on cancer progression.


Subject(s)
Adenocarcinoma, Mucinous/genetics , Colorectal Neoplasms/genetics , DNA-Binding Proteins , Microsatellite Repeats , Neoplasm Proteins/genetics , Proto-Oncogene Proteins/genetics , Adaptor Proteins, Signal Transducing , Adenocarcinoma, Mucinous/mortality , Adenocarcinoma, Mucinous/pathology , Adult , Age Factors , Base Pair Mismatch , Carrier Proteins , Cohort Studies , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , DNA Repair , Female , Germ-Line Mutation , Humans , Male , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Neoplasm Staging , Nuclear Proteins , Survival Rate , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...