Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 13.556
Filtrer
1.
JCO Precis Oncol ; 8: e2400094, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39088769

RÉSUMÉ

PURPOSE: Genetic susceptibility to nonsyndromic renal cell carcinoma (RCC) remains poorly understood, especially for different histological subtypes, as does variations in genetic predisposition in different populations. The objectives of this study were to identify risk genes for RCC in the Canadian population, investigate their clinical significance, and evaluate variations in germline pathogenic variants (PVs) among patients with RCC across the globe. MATERIALS AND METHODS: We conducted targeted sequencing of 19 RCC-related and 27 cancer predisposition genes for 960 patients with RCC from Canada and identified genes enriched in rare germline PVs in RCC compared with cancer-free controls. We combined our results with those reported for patients from Japan, the United Kingdom, and the United States to investigate PV variations in different populations. Furthermore, we evaluated the performance of referral criteria for genetic screening for including patients with rare PVs. RESULTS: We identified 39 germline PVs in 56 patients (5.8%) from the Canadian cohort. Compared with cancer-free controls, PVs in CHEK2 (odds ratio [OR], 4.8 [95% CI, 2.7 to 7.9], P = 3.94 × 10-5) and ATM (OR, 4.5 [95% CI, 2.0 to 8.7], P = .016) were significantly enriched in patients with clear cell, whereas PVs in FH (OR, 215.1 [95% CI, 64.4 to 597.8], P = 6.14 × 10-9) were enriched in patients with non-clear cell RCCs. PVs in BRCA1, BRCA2, and ATM were associated with metastasis (P = .003). Comparative analyses showed an enrichment of TP53 PVs in patients from Japan, of CHEK2 and ATM in patients from Canada, the United States and the United Kingdom, and of FH and BAP1 in the United States. CONCLUSION: CHEK2, ATM, and FH are risk genes for RCC in the Canadian population, whereas PVs in BRCA1/2 and ATM are associated with risk of metastasis. Globally, clinical guidelines for genetic screening in RCC fail to include more than 70% of patients with rare germline PVs.


Sujet(s)
Néphrocarcinome , Prédisposition génétique à une maladie , Dépistage génétique , Mutation germinale , Tumeurs du rein , Humains , Néphrocarcinome/génétique , Dépistage génétique/méthodes , Tumeurs du rein/génétique , Tumeurs du rein/anatomopathologie , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Adulte , Canada
2.
Cancer Rep (Hoboken) ; 7(8): e2141, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39118233

RÉSUMÉ

BACKGROUND: Myeloid neoplasms, including acute myeloid leukemia, have been traditionally among the less investigated cancer types concerning germline predisposition. Indeed, myeloid neoplasms with germline predisposition are challenging to identify because often display similar clinical and morphological characteristics of sporadic cases and have similar age at diagnosis. However, a misidentifications of familiarity in myeloid neoplasms have a critical impact on clinical management both for the carriers and their relatives. AIMS: We conducted a family segregation study, in order to identify novel cancer predisposing genes in myeloid neoplasms and classify novel identified variants. METHODS AND RESULTS: We performed a thorough genomic analysis using a large custom gene panel (256 genes), the Myelo-Panel, targeted on cancer predisposing genes. In particular, we assessed both germline and somatic variants in four families, each with two siblings, who developed hematological neoplasms: seven acute myeloid leukemia and one Philadelphia-positive chronic myeloid leukemia. In each family, we identified at least one novel potentially predisposing variant, affecting also genes not included in the current European LeukemiaNet guidelines for AML management. Moreover, we suggest reclassification of two germline variants as pathogenic: likely pathogenic p.S21Tfs*139 in CEPBA and VUS p.K392Afs*66 in DDX41. CONCLUSION: We believe that predisposition to hematological neoplasms is still underestimated and particularly difficult to diagnosed. Considering that misidentification of familiarity in myeloid neoplasms have a critical impact on the clinical management both for the carriers and their relatives, our study highlights the importance of revision, in this clinical context, of clinical practices that should include thorough reconstruction of family history and in-depth genetic testing.


Sujet(s)
Prédisposition génétique à une maladie , Mutation germinale , Leucémie aigüe myéloïde , Pedigree , Humains , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/anatomopathologie , Leucémie aigüe myéloïde/diagnostic , Femelle , Mâle , Adulte d'âge moyen , Adulte , DEAD-box RNA helicases/génétique , Sujet âgé
3.
Int J Mol Sci ; 25(15)2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-39125758

RÉSUMÉ

APC is a tumor suppressor gene that exerts its effect through the regulation of the Wnt signaling pathway. Loss of function mutations of the gene are associated with familial adenomatous polyposis (FAP). Early diagnosis in FAP patients is essential to prevent the development of colorectal cancer. Extraintestinal manifestations often precede the formation of the polyposis; therefore, these manifestations may serve as a clinical indicator for the condition. The aim of this study was to assess genotype-phenotype associations between the location of APC mutations and various extraintestinal features, mainly focusing on osseous and dental anomalies. Analyses of our cases and the mutations available in the literature with these manifestations revealed that mutations in the N-terminal region (amino acids 1-~1000) of the protein are more frequently associated with only osseous anomalies, whereas dental manifestations are more prevalent in mutations in the middle region (amino acids 1000-~2100). In addition, supernumerary teeth were found to be the most common dental feature. Since dental abnormalities often precede intestinal polyposis, dentists have a crucial role in the early identification of patients at risk.


Sujet(s)
Protéine de la polypose adénomateuse colique , Polypose adénomateuse colique , Mutation germinale , Humains , Protéine de la polypose adénomateuse colique/génétique , Polypose adénomateuse colique/génétique , Malformations dentaires/génétique , Études d'associations génétiques , Dent surnuméraire/génétique , Prédisposition génétique à une maladie , Mâle , Femelle
4.
BMC Med Genomics ; 17(1): 215, 2024 Aug 19.
Article de Anglais | MEDLINE | ID: mdl-39160519

RÉSUMÉ

Hereditary leiomyomatosis and renal cell cancer (HLRCC) is a rare autosomal dominant syndrome caused by a germline mutation in the fumarate hydratase (FH) gene that manifests with cutaneous leiomyomas, uterine fibroids, and renal cell cancer (RCC). Patients with HLRCC-associated RCC (HLRCC-RCC) have aggressive clinical courses, but there is no standardized therapy for advanced HLRCC-RCC. In this study, we described a case of aggressive HLRCC in a 33-year-old female who exhibited a novel heterozygous germline insertion mutation in exon 8 of the FH gene (c.1126 C > T; p.Q376*). The patient underwent laparoscopic resection of the right kidney, but metastases appeared within 3 months after surgery. Histological staining of the resected tumor revealed high expression levels of programmed cell death-ligand 1 (PD-L1). Therefore, the patient was treated with immunotherapy. The patient achieved a partial response to immunotherapy, and the treatment of metastatic lesions has continued to improve. A thorough literature review pinpointed 76 historical cases of HLRCC-RCC that had undergone immunotherapy. From this pool, 46 patients were selected for this study to scrutinize the association between mutations in the FH gene and the effectiveness of immunotherapy. Our results indicate that immunotherapy could significantly improve the overall survival (OS) of patients with HLRCC-RCC. However, no influence of different mutations in the FH germline gene on the therapeutic efficacy of immunotherapy was observed. Therefore, our study suggested that immunotherapy was an effective therapeutic option for patients with HLRCC regardless of the type of FH germline mutation.


Sujet(s)
Fumarate hydratase , Immunothérapie , Léiomyomatose , Syndromes néoplasiques héréditaires , Tumeurs cutanées , Tumeurs de l'utérus , Humains , Femelle , Léiomyomatose/génétique , Léiomyomatose/anatomopathologie , Léiomyomatose/thérapie , Fumarate hydratase/génétique , Adulte , Syndromes néoplasiques héréditaires/génétique , Syndromes néoplasiques héréditaires/thérapie , Syndromes néoplasiques héréditaires/complications , Syndromes néoplasiques héréditaires/anatomopathologie , Tumeurs de l'utérus/génétique , Tumeurs de l'utérus/anatomopathologie , Tumeurs de l'utérus/thérapie , Tumeurs cutanées/génétique , Tumeurs cutanées/anatomopathologie , Tumeurs cutanées/thérapie , Mutation germinale , Tumeurs du rein/génétique , Tumeurs du rein/anatomopathologie , Tumeurs du rein/thérapie , Néphrocarcinome/génétique , Néphrocarcinome/anatomopathologie , Néphrocarcinome/thérapie
5.
Cancer Med ; 13(16): e70103, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39149814

RÉSUMÉ

BACKGROUND: Monoallelic germline pathogenic variants (GPVs) in five Fanconi anemia (FA) genes (BRCA1/FANCS, BRCA2/FANCD1, PALB2/FANCN, BRIP1/FANCJ, and RAD51C/FANCO) confer an increased risk of breast (BC) and/or ovarian (OC) cancer, but the role of GPVs in 17 other FA genes remains unclear. METHODS: Here, we investigated the association of germline variants in FANCG/XRCC9 with BC and OC risk. RESULTS: The frequency of truncating GPVs in FANCG did not differ between BC (20/10,204; 0.20%) and OC (8/2966; 0.27%) patients compared to controls (6/3250; 0.18%). In addition, only one out of five tumor samples showed loss-of-heterozygosity of the wild-type FANCG allele. Finally, none of the nine functionally tested rare recurrent missense FANCG variants impaired DNA repair activities (FANCD2 monoubiquitination and FANCD2 foci formation) upon DNA damage, in contrast to all tested FANCG truncations. CONCLUSION: Our study suggests that heterozygous germline FANCG variants are unlikely to contribute to the development of BC or OC.


Sujet(s)
Tumeurs du sein , Protéine du groupe de complémentation G de l'anémie de Fanconi , Prédisposition génétique à une maladie , Mutation germinale , Tumeurs de l'ovaire , Humains , Femelle , Tumeurs du sein/génétique , Tumeurs de l'ovaire/génétique , Protéine du groupe de complémentation G de l'anémie de Fanconi/génétique , Adulte d'âge moyen , Adulte , Réparation de l'ADN/génétique , Études cas-témoins , Sujet âgé
6.
Genome Med ; 16(1): 101, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39148102

RÉSUMÉ

BACKGROUND: The Alpe-DPD study (NCT02324452) demonstrated that prospective genotyping and dose-individualization using four alleles in DPYD (DPYD*2A/rs3918290, c.1236G > A/rs75017182, c.2846A > T/rs67376798 and c.1679 T > G/rs56038477) can mitigate the risk of severe fluoropyrimidine toxicity. However, this could not prevent all toxicities. The goal of this study was to identify additional genetic variants, both inside and outside DPYD, that may contribute to fluoropyrimidine toxicity. METHODS: Biospecimens and data from the Alpe-DPD study were used. Exon sequencing was performed to identify risk variants inside DPYD. In silico and in vitro analyses were used to classify DPYD variants. A genome-wide association study (GWAS) with severe fluoropyrimidine-related toxicity was performed to identify variants outside DPYD. Association with severe toxicity was assessed using matched-pair analyses for the exon sequencing and logistic, Cox, and ordinal regression analyses for GWAS. RESULTS: Twenty-four non-synonymous, frameshift, and splice site DPYD variants were detected in ten of 986 patients. Seven of these variants (c.1670C > T, c.1913 T > C, c.1925 T > C, c.506delC, c.731A > C, c.1740 + 1G > T, c.763 - 2A > G) were predicted to be deleterious. The carriers of either of these variants showed a trend towards a 2.14-fold (95% CI, 0.41-11.3, P = 0.388) increased risk of severe toxicity compared to matched controls (N = 30). After GWAS of 942 patients, no individual single nucleotide polymorphisms achieved genome-wide significance (P ≤ 5 × 10-8), however, five variants were suggestive of association (P < 5 × 10-6) with severe toxicity. CONCLUSIONS: Results from DPYD exon sequencing and GWAS analysis did not identify additional genetic variants associated with severe toxicity, which suggests that testing for single markers at a population level currently has limited clinical value. Identifying additional variants on an individual level is still promising to explain fluoropyrimidine-related severe toxicity. In addition, studies with larger samples sizes, in more diverse cohorts are needed to identify potential clinically relevant genetic variants related to severe fluoropyrimidine toxicity.


Sujet(s)
Dihydrouracil dehydrogenase (NADP) , Humains , Dihydrouracil dehydrogenase (NADP)/génétique , Femelle , Mâle , Adulte d'âge moyen , Étude d'association pangénomique , Mutation germinale , Sujet âgé , Polymorphisme de nucléotide simple , Adulte , Fluorouracil/effets indésirables , Pyrimidines/effets indésirables , Antimétabolites antinéoplasiques/effets indésirables , Exons
7.
Am J Case Rep ; 25: e943882, 2024 Aug 11.
Article de Anglais | MEDLINE | ID: mdl-39127886

RÉSUMÉ

BACKGROUND Hereditary breast cancer arising in BRCA1-deficient patients is commonly diagnosed as invasive carcinoma of no special type (NST) with medullary features, while invasive lobular carcinoma (ILC) appears to be significantly under-represented in BRCA1 mutation carriers. We report a case of pleomorphic ILC arising in a 28-year-old woman harboring a germline BRCA1 c.3756_3759delGTCT p.(Ser1253Argfs*10) pathogenic variant. CASE REPORT A nulliparous 28-year-old woman with a family history of BRCA1 mutation presented to the symptomatic breast clinic with a several-week history of a left 80-mm breast lump. Core biopsy established a diagnosis of a poorly differentiated triple-negative breast cancer (TNBC) of pleomorphic lobular phenotype. Her clinical diagnosis was cT3, N0, M0, cStageIIB. The MDT recommended CT staging, MRI breast imaging and neoadjuvant chemotherapy (NACT). PET CT imaging showed no evidence of distant metastatic disease. The patient had a good radiological response to NACT with a FEC-T carboplatin regimen. Post-NACT imaging showed a residual cystic mass and the patient underwent a mastectomy and sentinel lymph node biopsy with plans for a delayed latissimus dorsi reconstruction following her adjuvant radiotherapy treatment. A complete pathological response was subsequently demonstrated without any evidence of metastatic disease. CONCLUSIONS This case is the first report of pleomorphic ILC with a triple-negative receptor status and a complete pathological response in a BRCA1 mutation carrier. Our study expands the heterogeneous spectrum of TNBC and contributes to a better understanding of the molecular genetic landscape that characterizes invasive pleomorphic lobular neoplasia.


Sujet(s)
Carcinome lobulaire , Tumeurs du sein triple-négatives , Humains , Femelle , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/thérapie , Adulte , Carcinome lobulaire/génétique , Carcinome lobulaire/anatomopathologie , Carcinome lobulaire/thérapie , Protéine BRCA1/génétique , Mutation germinale
8.
BMC Cancer ; 24(1): 1000, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-39134950

RÉSUMÉ

BACKGROUND: Germline breast cancer susceptibility gene (gBRCA) mutation in patients with pancreatic cancer (PC) is not common in clinical practice. Therefore, factors that efficiently show gBRCA mutations and the real-world outcomes of olaparib maintenance therapy have not been fully established. In the present study, we clarified the indicators for the effective detection of gBRCA mutation and the efficacy and safety of olaparib as maintenance therapy. METHODS: We retrospectively analyzed 84 patients with PC who underwent gBRCA testing (BRACAnalysis, Myriad Genetics, Salt Lake City, UT, USA) at our institute between January 2021 and March 2022. For each patient, clinical data were extracted from medical records. RESULTS: The median patient age was 64 y (29-85 y), and 41 patients (48.8%) were male. The gBRCA mutations were identified in 10 (11.9%) patients; two patients had BRCA1 mutation and eight had BRCA2 mutation. All patients with gBRCA mutation had a family history of any cancer, and eight of them had a family history of Hereditary Breast and Ovarian Cancer syndrome (HBOC)-related cancer. The gBRCA mutation rate was higher for patients with PC with a family history of HBOC-related cancer compared to that in patients with PC having a family history of other cancers and no family history of cancer (22.9% vs. 4.1%; P = 0.014). In our study, eight out of 10 patients with gBRCA-positive PC received olaparib after platinum-based chemotherapy. The best responses to platinum-based chemotherapy included a complete response in one patient (12.5%) and a partial response in seven patients (87.5%). The median duration of treatment with platinum-based chemotherapy plus olaparib was 17.5 months (8-87 months), and the duration of treatment with olaparib maintenance therapy was 11 months (1-30 months). During olaparib maintenance therapy, three patients showed no disease progression. One of these three patients underwent conversion surgery after receiving olaparib for 12 months. CONCLUSIONS: The gBRCA testing should be considered proactively, especially in patients with PC with a family history of HBOC-related cancer.


Sujet(s)
Protéine BRCA1 , Protéine BRCA2 , Prédisposition génétique à une maladie , Mutation germinale , Tumeurs du pancréas , Phtalazines , Pipérazines , Humains , Phtalazines/usage thérapeutique , Adulte d'âge moyen , Femelle , Sujet âgé , Mâle , Adulte , Études rétrospectives , Tumeurs du pancréas/génétique , Tumeurs du pancréas/traitement médicamenteux , Sujet âgé de 80 ans ou plus , Pipérazines/usage thérapeutique , Pipérazines/administration et posologie , Protéine BRCA2/génétique , Protéine BRCA1/génétique , Chimiothérapie de maintenance , Dépistage génétique/méthodes , Pertinence clinique
10.
Sci Rep ; 14(1): 16183, 2024 07 13.
Article de Anglais | MEDLINE | ID: mdl-39003285

RÉSUMÉ

The subset of ovarian cancer (OC) diagnosed ≤ 30yo represents a distinct subgroup exhibiting disparities from late-onset OC in many aspects, including indefinite germline cancer predisposition. We performed DNA/RNA-WES with HLA-typing, PRS assessment and survival analysis in 123 early-onset OC-patients compared to histology/stage-matched late-onset and unselected OC-patients, and population-matched controls. Only 6/123(4.9%) early-onset OC-patients carried a germline pathogenic variant (GPV) in high-penetrance OC-predisposition genes. Nevertheless, our comprehensive germline analysis of early-onset OC-patients revealed two divergent trajectories of potential germline susceptibility. Firstly, overrepresentation analysis highlighted a connection to breast cancer (BC) that was supported by the CHEK2 GPV enrichment in early-onset OC(p = 1.2 × 10-4), and the presumably BC-specific PRS313, which successfully stratified early-onset OC-patients from controls(p = 0.03). The second avenue pointed towards the impaired immune response, indicated by LY75-CD302 GPV(p = 8.3 × 10-4) and diminished HLA diversity compared with controls(p = 3 × 10-7). Furthermore, we found a significantly higher overall GPV burden in early-onset OC-patients compared to controls(p = 3.8 × 10-4). The genetic predisposition to early-onset OC appears to be a heterogeneous and complex process that goes beyond the traditional Mendelian monogenic understanding of hereditary cancer predisposition, with a significant role of the immune system. We speculate that rather a cumulative overall GPV burden than specific GPV may potentially increase OC risk, concomitantly with reduced HLA diversity.


Sujet(s)
Âge de début , Prédisposition génétique à une maladie , Mutation germinale , Tumeurs de l'ovaire , Humains , Femelle , Tumeurs de l'ovaire/génétique , Adulte , Adulte d'âge moyen , Études cas-témoins , Jeune adulte , Checkpoint kinase 2/génétique
11.
PLoS One ; 19(7): e0306612, 2024.
Article de Anglais | MEDLINE | ID: mdl-39012888

RÉSUMÉ

PURPOSE: We present a methodically devised protocol for conducting a systematic review and meta-analysis aimed at ascertaining the prevalence of BReast CAncer gene (BRCA) mutations in breast and ovarian cancer (BOC) among women in India. The review will include cross-sectional, cohort, case-series, and registry-based studies focusing on females clinically diagnosed with any stage of BOC, tested for BRCA germline mutation and undergone any form of treatment. METHODS: A Cochrane literature search will be carried out to identify all the published and unpublished articles available in English from 2010 till date across various electronic databases including PubMed, Psych Info, SCI, Cochrane Central, Embase, Scopus, IND Med and Google Scholar. A step-by-step process will be followed to select all the relevant studies for final inclusion using Rayyan software. The selection process of the review will be reported based on Preferred Reporting Items for Systematic Review and Meta-analysis Protocols (PRISMA) checklist. The protocol has been registered in PROSPERO (ID: CRD42023463452). Joanna Briggs Institute Critical Appraisal Checklist will be used to evaluate the methodological quality of the included studies. The outcome measure will be the prevalence of BRCA1/2 gene mutation in this population. Meta-analysis will be performed to report the pooled prevalence along with 95% confidence interval. DISCUSSION: The results of this review study will provide valuable insights for clinicians, and policy makers, enabling them to formulate guidelines that underscore the importance of screening for BRCA mutations in cases of BOC.


Sujet(s)
Tumeurs du sein , Tumeurs de l'ovaire , Revues systématiques comme sujet , Femelle , Humains , Protéine BRCA1/génétique , Protéine BRCA2/génétique , Tumeurs du sein/génétique , Tumeurs du sein/épidémiologie , Mutation germinale , Inde/épidémiologie , Méta-analyse comme sujet , Mutation , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/épidémiologie , Prévalence
12.
Zhonghua Yi Xue Za Zhi ; 104(28): 2613-2618, 2024 Jul 23.
Article de Chinois | MEDLINE | ID: mdl-39019817

RÉSUMÉ

Objective: To explore the correlation between clinical characteristics and pathological features in patients with pheochromocytoma/paraganglioma (PPGLs). Methods: A case series study. A retrospective analysis was conducted on patients with single and primary PPGLs after postoperative pathological diagnosis who were admitted to Peking Union Medical College Hospital between January 2019 and December 2022. The patients were divided into the Ki-67<3% group and the Ki-67≥3% group with Ki-67 proliferation index of 3% as the threshold. The relationship between clinical and pathological characteristics of PPGLs was analyzed. Results: A total of 399 PPGLs patients were included, with 177 males and 222 females, aged [M(Q1, Q3)] 45.0(35.5, 53.0) years. Among them, 226 (56.6%) cases originated from the adrenal gland, while 104 cases (26.1%) from the retroperitoneum. 20.9% (27/129) of the patients were found to harbor germline mutations of susceptibility genes, with SDHB mutations being the most common (10.1%, 13/129). The Ki-67 staining was performed on 302 cases, with a Ki-67 proliferation index [M(Q1, Q3)] of 2.0% (1.0%, 3.0%). There were 194 cases in Ki-67<3% group and 108 cases in Ki-67≥3% group. Compared with the patients in Ki-67<3% group, the age of onset in Ki-67≥3% group was younger (P=0.029). Compared with the patients with paragangliomas without SDHB or Cluster 1A-related gene mutations, positive 131I-meta-iodobenzylguanidine (131I-MIBG) imaging or negative O-6-methylguanine-DNA methyltransferase (MGMT) immunohistochemistry staining, those with SDHB or Cluster 1A-related gene mutations, negative 131I-MIBG imaging or positive MGMT immunohistochemistry staining had a higher Ki-67 index (all P<0.05). Compared with adrenal pheochromocytoma, retroperitoneal paragangliomas had a higher proportion of SDHB mutations and a higher proportion of normetanephrine (NMN) secretory types (all P<0.05). Compared with adrenal pheochromocytoma, the maximum diameter of head and neck paraganglioma tumors was smaller [3.0 (1.9, 3.8) cm vs 4.7 (3.4, 6.4) cm, P<0.001] and the proportion of Ki-67≥3% was higher (61.3% vs 33.8%, P=0.007). Conclusions: PPGLs patients with earlier onset age, SDHB or Cluster 1A-related gene mutations, negative 131I-MIBG imaging, or positive MGMT immunohistochemistry staining tend to have a higher Ki-67 index. Head and neck tumors, though smaller, exhibit a higher proliferation potential.


Sujet(s)
Tumeurs de la surrénale , Antigène KI-67 , Paragangliome , Phéochromocytome , Humains , Phéochromocytome/anatomopathologie , Phéochromocytome/génétique , Mâle , Femelle , Adulte , Études rétrospectives , Adulte d'âge moyen , Paragangliome/anatomopathologie , Paragangliome/génétique , Tumeurs de la surrénale/anatomopathologie , Tumeurs de la surrénale/génétique , Antigène KI-67/métabolisme , Mutation germinale , Succinate Dehydrogenase/génétique
13.
Hematology ; 29(1): 2377860, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39007733

RÉSUMÉ

BACKGROUD: Li-Fraumeni syndrome is a hereditary tumor syndrome characterized by an elevated risk of malignancy, particularly acute lymphoblastic leukemia (ALL), which can be caused by the heterozygous germline mutation. TP53 gene germline mutation is considered a potential risk factor and crucial prognostic parameter for acute leukemia development and diagnosis, but rarely occurs in adults, and its specific pathogenic significance in acute leukemia is unclear. CASE PRESENTATION: We describes a case of a 45-year-old woman diagnosed with ALL. Whole-exome sequencing approach identified one of the TP53 germline mutations from her bone marrow sample with possible pathogenic significance, c.848G>A (p.Arg283His) heterozygous missense mutation located on exon 8, which was further verified in her hair, oral mucous and nail samples. Family pedigree screening revealed that the same TP53 genetic variant was present in the patient's father and non-donor son, whereas not in the donor. Digital PCR observed that this point mutation frequency dropped post-transplantation but remained low during maintenance therapy when the patient was leukemia-free. CONCLUSION: This suspected Li-Fraumeni syndrome case report with a likely pathogenic heterozygous TP53 variant expands the cancer genetic spectrum. Screening her family members for mutations facilitates identifying the optimal relative donor and avoids unnecessary treatment by monitoring TP53 germline mutations for minimal residual disease following hematopoietic stem cell transplantation. Its potential roles in hematological malignant tumor development and clinical pathogenic implications necessitate further probing.


Sujet(s)
Mutation germinale , Syndrome de Li-Fraumeni , Leucémie-lymphome lymphoblastique à précurseurs B et T , Protéine p53 suppresseur de tumeur , Humains , Femelle , Leucémie-lymphome lymphoblastique à précurseurs B et T/génétique , Leucémie-lymphome lymphoblastique à précurseurs B et T/diagnostic , Leucémie-lymphome lymphoblastique à précurseurs B et T/thérapie , Adulte d'âge moyen , Protéine p53 suppresseur de tumeur/génétique , Syndrome de Li-Fraumeni/génétique , Syndrome de Li-Fraumeni/diagnostic , Pedigree
14.
Nat Commun ; 15(1): 6025, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-39019934

RÉSUMÉ

Therapy-related myeloid neoplasms (t-MN) arise as a complication of chemo- and/or radiotherapy. Although t-MN can occur both in adult and childhood cancer survivors, the mechanisms driving therapy-related leukemogenesis likely vary across different ages. Chemotherapy is thought to induce driver mutations in children, whereas in adults pre-existing mutant clones are selected by the exposure. However, selective pressures induced by chemotherapy early in life are less well studied. Here, we use single-cell whole genome sequencing and phylogenetic inference to show that the founding cell of t-MN in children starts expanding after cessation of platinum exposure. In patients with Li-Fraumeni syndrome, characterized by a germline TP53 mutation, we find that the t-MN already expands during treatment, suggesting that platinum-induced growth inhibition is TP53-dependent. Our results demonstrate that germline aberrations can interact with treatment exposures in inducing t-MN, which is important for the development of more targeted, patient-specific treatment regimens and follow-up.


Sujet(s)
Mutation germinale , Syndrome de Li-Fraumeni , Seconde tumeur primitive , Protéine p53 suppresseur de tumeur , Humains , Protéine p53 suppresseur de tumeur/génétique , Syndrome de Li-Fraumeni/génétique , Enfant , Seconde tumeur primitive/génétique , Mâle , Femelle , Composés du platine/usage thérapeutique , Adulte , Adolescent , Séquençage du génome entier , Phylogenèse , Enfant d'âge préscolaire , Antinéoplasiques/usage thérapeutique , Analyse sur cellule unique
15.
Brief Bioinform ; 25(4)2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-39038933

RÉSUMÉ

Breast cancer (BC) is the most common malignancy affecting Western women today. It is estimated that as many as 10% of BC cases can be attributed to germline variants. However, the genetic basis of the majority of familial BC cases has yet to be identified. Discovering predisposing genes contributing to familial BC is challenging due to their presumed rarity, low penetrance, and complex biological mechanisms. Here, we focused on an analysis of rare missense variants in a cohort of 12 families of Middle Eastern origins characterized by a high incidence of BC cases. We devised a novel, high-throughput, variant analysis pipeline adapted for family studies, which aims to analyze variants at the protein level by employing state-of-the-art machine learning models and three-dimensional protein structural analysis. Using our pipeline, we analyzed 1218 rare missense variants that are shared between affected family members and classified 80 genes as candidate pathogenic. Among these genes, we found significant functional enrichment in peroxisomal and mitochondrial biological pathways which segregated across seven families in the study and covered diverse ethnic groups. We present multiple evidence that peroxisomal and mitochondrial pathways play an important, yet underappreciated, role in both germline BC predisposition and BC survival.


Sujet(s)
Tumeurs du sein , Apprentissage profond , Prédisposition génétique à une maladie , Humains , Tumeurs du sein/génétique , Femelle , Mutation faux-sens , Pedigree , Mutation germinale
16.
Pediatr Blood Cancer ; 71(9): e31175, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38961591

RÉSUMÉ

Atypical hemolytic uremic syndrome (aHUS) is a complement-mediated thrombotic microangiopathy sometimes associated with germline variants in genes of the complement system. Clinical findings of microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injury arise due to aberrant complement protein activation in the circulation. A 13-month-old boy with metastatic neuroblastoma (NB) developed aHUS during his first cycle of induction chemotherapy with germline testing revealing a complement factor H (CFH) gene mutation, currently classified as a variant of uncertain significance (VUS). Now he is in disease remission after successful complement blockade therapy, thus highlighting a unique presentation of aHUS in a patient with newly diagnosed NB.


Sujet(s)
Syndrome hémolytique et urémique atypique , Chimiothérapie d'induction , Neuroblastome , Humains , Neuroblastome/traitement médicamenteux , Neuroblastome/anatomopathologie , Neuroblastome/génétique , Mâle , Syndrome hémolytique et urémique atypique/traitement médicamenteux , Syndrome hémolytique et urémique atypique/génétique , Syndrome hémolytique et urémique atypique/anatomopathologie , Chimiothérapie d'induction/effets indésirables , Nourrisson , Facteur H du complément/génétique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Mutation germinale
17.
Mol Genet Genomic Med ; 12(7): e2496, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39056403

RÉSUMÉ

BACKGROUND: Hereditary diffuse gastric cancer (HDGC) (OMIM# 137215) is an autosomal dominant cancer syndrome associated with CDH1 (OMIM# 192090) mutations. Prophylactic total gastrectomy (PTG) is the most recommended preventive treatment when a pathogenic mutation is found. However, the increasing use of genetic testing has led to the identification of incidental CDH1 mutations in individuals without a family history of gastric cancer. It remains unclear whether these patients should undergo prophylactic total gastrectomy. METHODS: Germline DNA, obtained from peripheral blood, was analysed by NGS. RESULTS: A 47-year-old woman was diagnosed with high-grade serous ovarian carcinoma, FIGO stage IIIC, with a Homologous Recombination Deficiency (HRD) GIS status of 78 (positive, cut-off: 43). She received chemotherapy and niraparib treatment. A multigene panel test revealed no pathogenic mutations in BRCA1 (OMIM# 113705)/BRCA2 (OMIM# 600185) genes, but a de novo deletion of exon 16 in CDH1 was found incidentally. She had no previous family history of gastric or breast cancer. The patient was enrolled in a surveillance program involving periodic endoscopy and was diagnosed with diffuse gastric cancer through biopsies of a pale area in the antrum after 1 year of close endoscopic follow-up. CONCLUSION: This case presents supportive evidence for the pathogenic classification of the loss of the last exon of CDH1.


Sujet(s)
Antigènes CD , Cadhérines , Tumeurs de l'ovaire , Tumeurs de l'estomac , Humains , Femelle , Adulte d'âge moyen , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Antigènes CD/génétique , Cadhérines/génétique , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , Gastrectomie , Mutation germinale
18.
Genes (Basel) ; 15(7)2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-39062660

RÉSUMÉ

Breast cancer (BC) risks imparted by CHEK2 c.1100delC ("1100delC") germline pathogenic/likely pathogenic variant (GPV) are 20-30%, compared to CHEK2 c.470T>C ("I157T") GPV with <20%, leading to different breast screening recommendations through MRI. We compared cancer risk management (CRM) across these two GPVs. Study participants were adult females with an 1100delC or I157T GPV drawn from the Inherited Cancer Registry (ICARE) across the United States. Cancer history, clinical characteristics, and CRM were compared using chi-squared tests, t-tests, and logistic regression. Of 150 CHEK2 carriers, 40.7% had BC, with a mean age of 50. Comparing 1100delC and I157T GPVs, there were no differences in rates of (1) breast MRI among those with (65.2% versus 55.6% of 23 and 9; p = 0.612) and without (44.0% versus 44.8% of 50 and 29; p = 0.943) BC; (2) risk-reducing mastectomy among those with (50% versus 38.9% of 46 and 15; p = 0.501) and without (13.8% versus 6.5% of 58 and 31; p = 0.296) BC; and (3) risk-reducing salpingo-oophorectomy among those with (24.2% versus 22.2% of 45 and 18; p = 0.852) and without (17.5% versus 16.7% of 57 and 30; p = 0.918) BC. The results suggest over-screening with breast MRI among CHEK2 I157T GPV carriers and possible overuse of risk-reducing surgeries among CHEK2 carriers.


Sujet(s)
Tumeurs du sein , Checkpoint kinase 2 , Prédisposition génétique à une maladie , Humains , Checkpoint kinase 2/génétique , Femelle , Tumeurs du sein/génétique , Adulte d'âge moyen , Adulte , Sujet âgé , Mutation faux-sens , Gestion du risque , Mutation germinale , Imagerie par résonance magnétique
19.
Genes (Basel) ; 15(7)2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39062721

RÉSUMÉ

Germline BRCA1/2 alteration has been linked to an increased risk of hereditary breast and ovarian cancer syndromes. As a result, genetic testing, based on NGS, allows us to identify a high number of variants of uncertain significance (VUS) or conflicting interpretation of pathogenicity (CIP) variants. The identification of CIP/VUS is often considered inconclusive and clinically not actionable for the patients' and unaffected carriers' management. In this context, their assessment and classification remain a significant challenge. The aim of the study was to investigate whether the in silico prediction tools (PolyPhen-2, SIFT, Mutation Taster and PROVEAN) could predict the potential clinical impact and significance of BRCA1/2 CIP/VUS alterations, eventually impacting the clinical management of Breast Cancer subjects. In a cohort of 860 BC patients, 10.6% harbored BRCA1 or BRCA2 CIP/VUS alterations, mostly observed in BRCA2 sequences (85%). Among them, forty-two out of fifty-five alterations were predicted as damaging, with at least one in silico that used tools. Prediction agreement of the four tools was achieved in 45.5% of patients. Moreover, the highest consensus was obtained in twelve out of forty-two (28.6%) mutations by considering three out of four in silico algorithms. The use of prediction tools may help to identify variants with a potentially damaging effect. The lack of substantial agreement between the different algorithms suggests that the bioinformatic approaches should be combined with the personal and family history of the cancer patients.


Sujet(s)
Protéine BRCA1 , Protéine BRCA2 , Tumeurs du sein , Simulation numérique , Humains , Femelle , Tumeurs du sein/génétique , Protéine BRCA2/génétique , Protéine BRCA1/génétique , Adulte d'âge moyen , Adulte , Prédisposition génétique à une maladie , Dépistage génétique/méthodes , Mutation germinale , Sujet âgé , Études de cohortes
20.
Cancer Med ; 13(13): e7394, 2024 07.
Article de Anglais | MEDLINE | ID: mdl-38970307

RÉSUMÉ

BACKGROUND: Germline mutations have been identified in a small number of hereditary cancers, but the genetic predisposition for many familial cancers remains to be elucidated. METHODS: This study identified a Chinese pedigree that presented different cancers (breast cancer, BRCA; adenocarcinoma of the esophagogastric junction, AEG; and B-cell acute lymphoblastic leukemia, B-ALL) in each of the three generations. Whole-genome sequencing and whole-exome sequencing were performed on peripheral blood or bone marrow and cancer biopsy samples. Whole-genome bisulfite sequencing was conducted on the monozygotic twin brothers, one of whom developed B-ALL. RESULTS: According to the ACMG guidelines, bioinformatic analysis of the genome sequencing revealed 20 germline mutations, particularly mutations in the DNAH11 (c.9463G > A) and CFH (c.2314G > A) genes that were documented in the COSMIC database and validated by Sanger sequencing. Forty-one common somatic mutated genes were identified in the cancer samples, displaying the same type of single nucleotide substitution Signature 5. Meanwhile, hypomethylation of PLEK2, MRAS, and RXRA as well as hypermethylation of CpG island associated with WT1 was shown in the twin with B-ALL. CONCLUSIONS: These findings reveal genomic alterations in a pedigree with multiple cancers. Mutations found in the DNAH11, CFH genes, and other genes predispose to malignancies in this family. Dysregulated methylation of WT1, PLEK2, MRAS, and RXRA in the twin with B-ALL increases cancer susceptibility. The similarity of the somatic genetic changes among the three cancers indicates a hereditary impact on the pedigree. These familial cancers with germline and somatic mutations, as well as epigenomic alterations, represent a common molecular basis for many multiple cancer pedigrees.


Sujet(s)
Méthylation de l'ADN , , Prédisposition génétique à une maladie , Mutation germinale , Pedigree , Humains , Mâle , Femelle , Séquençage du génome entier , Adulte d'âge moyen , Génomique/méthodes , Adulte , Épigenèse génétique , Ilots CpG , Épigénomique/méthodes , Dynéines de l'axonème/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE