Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
J Med Genet ; 52(11): 770-8, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26318770

ABSTRACT

BACKGROUND: Constitutional mismatch repair deficiency (CMMRD) syndrome is a childhood cancer predisposition syndrome involving biallelic germline mutations of MMR genes, poorly recognised by clinicians so far. METHODS: Retrospective review of all 31 patients with CMMRD diagnosed in French genetics laboratories in order to describe the characteristics, treatment and outcome of the malignancies and biological diagnostic data. RESULTS: 67 tumours were diagnosed in 31 patients, 25 (37%) Lynch syndrome-associated malignancies, 22 (33%) brain tumours, 17 (25%) haematological malignancies and 3 (5%) sarcomas. The median age of onset of the first tumour was 6.9 years (1.2-33.5). Overall, 22 patients died, 9 (41%) due to the primary tumour. Median survival after the diagnosis of the primary tumour was 27 months (0.26-213.2). Failure rate seemed to be higher than expected especially for T-cell non-Hodgkin's lymphoma (progression/relapse in 6/12 patients). A familial history of Lynch syndrome was identified in 6/23 families, and consanguinity in 9/23 families. PMS2 mutations (n=18) were more frequent than other mutations (MSH6 (n=6), MLH1 (n=4) and MSH2 (n=3)). CONCLUSIONS: In conclusion, this unselected series of patients confirms the extreme severity of this syndrome with a high mortality rate mostly related to multiple childhood cancers, and highlights the need for its early detection in order to adapt treatment and surveillance.


Subject(s)
Brain Neoplasms/diagnosis , Colorectal Neoplasms/diagnosis , Neoplastic Syndromes, Hereditary/diagnosis , Adaptor Proteins, Signal Transducing/genetics , Adenosine Triphosphatases/genetics , Adolescent , Adult , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Child , Child, Preschool , Colorectal Neoplasms/genetics , Colorectal Neoplasms/therapy , DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , Female , Humans , Infant , Male , Mismatch Repair Endonuclease PMS2 , MutL Protein Homolog 1 , MutS Homolog 2 Protein/genetics , Mutation , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/therapy , Nuclear Proteins/genetics , Treatment Outcome , Young Adult
2.
Mutat Res Genet Toxicol Environ Mutagen ; 789-790: 28-35, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26232255

ABSTRACT

The p53 tumor suppressor protein has emerged as a universal sensor of genotoxic stress that regulates the transcription of numerous genes required for appropriate cellular response to DNA damage. Therefore, transcriptional induction of p53 target genes can be considered as a global and early indicator of genotoxic stress. By performing expression microarrays and RNA-Seq analysis on wild-type and mutant TP53 human lymphocytes respectively derived from controls and Li-Fraumeni patients and exposed to different classes of genotoxic agents, we first determined a common p53-dependent transcriptional signature of DNA damage. We then derived a simple and fast assay based on the exposure of wild-type TP53 lymphocytes to physical or chemical agents and on the quantitative measurement of selected p53 target gene transcriptional induction. The specificity of the p53 genotoxicity assay can easily be demonstrated by performing the same experiment in control lymphocytes with heterozygous TP53 mutations, which compromise responses to DNA damage. This assay allowed us to show that most of the drugs commonly used in cancer treatment, except the microtubule poisons, are highly genotoxic. The p53 genotoxicity assay should facilitate the measurement of the genotoxic effects of chemical and physical agents and the identification of drugs that are not genotoxic and do not expose patients to the risk of secondary malignancies, especially those with a constitutional defect in response to DNA damage, such as patients with Li-Fraumeni syndrome.


Subject(s)
Lymphocytes/metabolism , Mutagenicity Tests/methods , Transcriptome/genetics , Tumor Suppressor Protein p53/genetics , Antineoplastic Agents/pharmacology , Cells, Cultured , Cisplatin/pharmacology , DNA Damage , Doxorubicin/pharmacology , Fluorouracil/pharmacology , Humans , Li-Fraumeni Syndrome/blood , Li-Fraumeni Syndrome/genetics , Lymphocytes/drug effects , Mutation , Oligonucleotide Array Sequence Analysis , Reproducibility of Results , Transcriptional Activation/drug effects , Transcriptome/drug effects
3.
Br J Cancer ; 100(8): 1330-5, 2009 Apr 21.
Article in English | MEDLINE | ID: mdl-19367287

ABSTRACT

Recent studies have suggested that activation of the EGFR pathway leads to malignant transformation only if the p53 protein is inactivated. Therefore, we evaluated the impact of TP53 mutations on cetuximab-based chemotherapy (CT) sensitivity in combination with KRAS mutations that have been associated with cetuximab resistance. KRAS and TP53 status were assessed in tumours from 64 metastatic colorectal cancer patients treated with cetuximab-based CT and correlated to clinical response using the Fisher's exact test. Times to progression (TTPs) according to gene status were calculated using the Kaplan-Meier method and compared with log-rank test. TP53 mutations were found in 41 patients and were significantly associated with controlled disease (CD), as defined as complete response, partial response or stable disease (P=0.037) and higher TTP (20 vs 12 weeks, P=0.004). Remarkably, in the subgroup of 46 patients without KRAS mutation, but not in patients with KRAS mutation, TP53 mutations were also associated with CD (P=0.008) and higher TTP (24 vs 12 weeks, P=0.0007). This study suggests that TP53 mutations are predictive of cetuximab sensitivity, particularly in patients without KRAS mutation, and that TP53 genotyping could have a clinical interest to select patients who should benefit from cetuximab-based CT.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Mutation , Tumor Suppressor Protein p53/genetics , Aged , Amino Acid Substitution , Antibodies, Monoclonal, Humanized , Cetuximab , Colorectal Neoplasms/pathology , DNA, Neoplasm/genetics , DNA, Neoplasm/isolation & purification , Disease Progression , Dose-Response Relationship, Drug , Exons , Female , Genotype , Humans , Male , Middle Aged , Neoplasm Metastasis , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , ras Proteins/genetics
4.
J Med Genet ; 45(8): 535-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18511570

ABSTRACT

We have performed an extensive analysis of TP53 in 474 French families suggestive of Li-Fraumeni syndrome (LFS), including 232 families fulfilling the Chompret criteria. We identified a germline alteration of TP53 in 82 families (17%), in 67/232 of the families fulfilling the Chompret criteria (29%) and in 15/242 which did not fulfil these criteria (6%). Most of the alterations corresponded to missense mutations (67%), and we identified in four families genomic deletions removing the entire TP53 locus, the promoter and the non-coding exon 1, or exons 2-10. These results represent a definitive argument demonstrating that LFS results from TP53 haplodeficiency. The mean ages of tumour onset were significantly different between patients harbouring TP53 missense mutations and other types of alterations, missense mutations being associated with a 9 year earlier tumour onset. These results confirm that missense mutations not only inactivate p53 but also have an additional oncogenic effect. Germline alterations of TP53 that lead exclusively to loss of function are therefore associated with a later age of tumour onset and the presence of such mutations should be considered in atypical LFS families with tumours diagnosed after 40 years.


Subject(s)
Genes, p53 , Genetic Predisposition to Disease , Li-Fraumeni Syndrome/genetics , Female , France , Gene Deletion , Humans , Male , Mutation, Missense , Neoplasms/genetics , Pedigree
5.
J Med Genet ; 43(6): 531-3, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16258005

ABSTRACT

Li-Fraumeni syndrome, resulting from p53 (TP53) germline mutations, represents one of the most devastating genetic predispositions to cancer. Recently, the MDM2 SNP309 (T-->G variation) was shown to be associated with accelerated tumour formation in p53 mutation carriers. The impact of the common p53 codon 72 polymorphism on cancer risk remains controversial. We therefore investigated the effect of these two polymorphisms in 61 French carriers of the p53 germline mutation. The mean age of tumour onset in MDMD2 SNP309 G allele carriers (19.6 years) was significantly different from that observed in patients homozygous for the T allele (29.9 years, p<0.05). For the p53 codon 72 polymorphism, the mean age of tumour onset in Arg allele carriers (21.8 years) was also different from that of Pro/Pro patients (34.4 years, p<0.05). We observed a cumulative effect of both polymorphisms because the mean ages of tumour onset in carriers of the MDM2G and p53Arg alleles (16.9 years) and those with the MDM2T/T and p53Pro/Pro genotypes (43 years) were clearly different (p<0.02). Therefore, our results confirm the impact of the MDM2 SNP309 G allele on the age of tumour onset in germline p53 mutation carriers, and suggest that this effect may be amplified by the p53 72Arg allele. Polymorphisms affecting p53 degradation therefore represent one of the rare examples of modifier genetic factors identified to date in mendelian predispositions to cancer.


Subject(s)
Genes, p53 , Genetic Predisposition to Disease , Li-Fraumeni Syndrome/genetics , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-mdm2/genetics , Adolescent , Adult , Age of Onset , DNA Mutational Analysis , Disease Progression , Female , Gene Frequency , Germ-Line Mutation , Heterozygote , Humans , Li-Fraumeni Syndrome/diagnosis , Li-Fraumeni Syndrome/epidemiology , Male , Middle Aged , Risk Factors
6.
Eur J Hum Genet ; 9(8): 642-5, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11528512

ABSTRACT

TP63 gene mutations have recently been shown to be disease causing in EEC and SHFM. Two other overlapping syndromes with ectrodactyly as a major feature, have been mapped to chromosome 3q27 close by the TP63 locus, namely the LMS and ADULT syndromes. Here, we report on a missense TP63 gene mutation in an isolated ADULT syndrome case. This finding widens the spectrum of abnormalities to be ascribed to TP63 gene in human and emphasise on the variable roles of the different Tp63 isotypes.


Subject(s)
Abnormalities, Multiple/genetics , Genes, Dominant/genetics , Mutation/genetics , Abnormalities, Multiple/diagnosis , Alleles , Child , Female , Genetic Linkage , Genotype , Humans , Limb Deformities, Congenital/diagnosis , Limb Deformities, Congenital/genetics , Nipples/abnormalities , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL