Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.208
Filtrer
1.
Front Endocrinol (Lausanne) ; 15: 1385079, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948517

RÉSUMÉ

Background: 177Lu-oxodotreotide peptide receptor therapy (LuPRRT) is an efficient treatment for midgut neuroendocrine tumors (NETs) of variable radiological response. Several clinical, biological, and imaging parameters may be used to establish a relative disease prognosis but none is able to predict early efficacy or toxicities. We investigated expression levels for mRNA and miRNA involved in radiosensitivity and tumor progression searching for correlations related to patient outcome during LuPRRT therapy. Methods: Thirty-five patients received LuPRRT for G1/G2 midgut NETs between May 2019 and September 2021. Peripheral blood samples were collected prior to irradiation, before and 48 h after the second and the fourth LuPRRT, and at 6-month follow-up. Multiple regression analyses and Pearson correlations were performed to identify the miRNA/mRNA signature that will best predict response to LuPRRT. Results: Focusing on four mRNAs and three miRNAs, we identified a miRNA/mRNA signature enabling the early identification of responders to LuPRRT with significant reduced miRNA/mRNA expression after the first LuPRRT administration for patients with progressive disease at 1 year (p < 0.001). The relevance of this signature was reinforced by studying its evolution up to 6 months post-LuPRRT. Moreover, nadir absolute lymphocyte count within the first 2 months after the first LuPRRT administration was significantly related to low miRNA/mRNA expression level (p < 0.05) for patients with progressive disease. Conclusion: We present a pilot study exploring a miRNA/mRNA signature that correlates with early hematologic toxicity and therapeutic response 12 months following LuPRRT. This signature will be tested prospectively in a larger series of patients.


Sujet(s)
Tumeurs de l'intestin , microARN , Tumeurs neuroendocrines , ARN messager , Humains , Tumeurs neuroendocrines/génétique , Tumeurs neuroendocrines/sang , Tumeurs neuroendocrines/thérapie , Tumeurs neuroendocrines/radiothérapie , Tumeurs neuroendocrines/anatomopathologie , Mâle , Femelle , microARN/sang , microARN/génétique , Adulte d'âge moyen , Tumeurs de l'intestin/sang , Tumeurs de l'intestin/anatomopathologie , Tumeurs de l'intestin/génétique , Tumeurs de l'intestin/traitement médicamenteux , ARN messager/génétique , ARN messager/sang , Sujet âgé , Études de suivi , Adulte , Pronostic , Marqueurs biologiques tumoraux/sang , Marqueurs biologiques tumoraux/génétique , Somatostatine/analogues et dérivés , Somatostatine/usage thérapeutique , Récepteurs peptidiques/génétique , Radiopharmaceutiques/usage thérapeutique , Radiopharmaceutiques/administration et posologie , Lutétium , Radio-isotopes
2.
Mol Biol Rep ; 51(1): 704, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38824233

RÉSUMÉ

BACKGROUND: Tumor modeling using organoids holds potential in studies of cancer development, enlightening both the intracellular and extracellular molecular mechanisms behind different cancer types, biobanking, and drug screening. Intestinal organoids can be generated in vitro using a unique type of adult stem cells which are found at the base of crypts and are characterized by their high Lgr5 expression levels. METHODS AND RESULTS: In this study, we successfully established intestinal cancer organoid models by using both the BALB/c derived and mouse embryonic stem cells (mESCs)-derived intestinal organoids. In both cases, carcinogenesis-like model was developed by using azoxymethane (AOM) treatment. Carcinogenesis-like model was verified by H&E staining, immunostaining, relative mRNA expression analysis, and LC/MS analysis. The morphologic analysis demonstrated that the number of generated organoids, the number of crypts, and the intensity of the organoids were significantly augmented in AOM-treated intestinal organoids compared to non-AOM-treated ones. Relative mRNA expression data revealed that there was a significant increase in both Wnt signaling pathway-related genes and pluripotency transcription factors in the AOM-induced intestinal organoids. CONCLUSION: We successfully developed simple carcinogenesis-like models using mESC-based and Lgr5 + stem cell-based intestinal organoids. Intestinal organoid based carcinogenesi models might be used for personalized cancer therapy in the future.


Sujet(s)
Oxyde de diméthyl-diazène , Carcinogenèse , Cellules souches embryonnaires de souris , Organoïdes , Voie de signalisation Wnt , Animaux , Organoïdes/métabolisme , Organoïdes/anatomopathologie , Souris , Oxyde de diméthyl-diazène/toxicité , Carcinogenèse/anatomopathologie , Carcinogenèse/induit chimiquement , Carcinogenèse/génétique , Cellules souches embryonnaires de souris/métabolisme , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Souris de lignée BALB C , Intestins/anatomopathologie , Tumeurs de l'intestin/anatomopathologie , Tumeurs de l'intestin/induit chimiquement , Tumeurs de l'intestin/génétique , Tumeurs de l'intestin/métabolisme , Modèles animaux de maladie humaine , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie
3.
Nutrients ; 16(9)2024 Apr 27.
Article de Anglais | MEDLINE | ID: mdl-38732562

RÉSUMÉ

Iron supplements are widely consumed. However, excess iron may accelerate intestinal tumorigenesis. To determine the effect of excess iron on intestinal tumor burden and protein expression changes between tumor and normal tissues, ApcMin/+ mice were fed control (adequate) and excess iron (45 and 450 mg iron/kg diet, respectively; n = 9/group) for 10 wk. Tumor burden was measured, and two-dimensional fluorescence difference gel electrophoresis was used to identify differentially expressed proteins in tumor and normal intestinal tissues. There was a significant increase (78.3%; p ≤ 0.05) in intestinal tumor burden (mm2/cm) with excess iron at wk 10. Of 980 analyzed protein spots, 69 differentially expressed (p ≤ 0.05) protein isoforms were identified, representing 55 genes. Of the isoforms, 56 differed (p ≤ 0.05) between tumor vs. normal tissues from the adequate iron group and 23 differed (p ≤ 0.05) between tumors from the adequate vs. excess iron. Differentially expressed proteins include those involved in cell integrity and adaptive response to reactive oxygen species (including, by gene ID: ANPEP, DPP7, ITGB1, PSMA1 HSPA5). Biochemical pathway analysis found that iron supplementation modulated four highly significant (p ≤ 0.05) functional networks. These findings enhance our understanding of interplay between dietary iron and intestinal tumorigenesis and may help develop more specific dietary guidelines regarding trace element intake.


Sujet(s)
Compléments alimentaires , Modèles animaux de maladie humaine , Tumeurs de l'intestin , Charge tumorale , Animaux , Souris , Humains , Tumeurs de l'intestin/métabolisme , Tumeurs de l'intestin/anatomopathologie , Tumeurs de l'intestin/génétique , Fer/métabolisme , Fer alimentaire/administration et posologie , Souris de lignée C57BL , Mâle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Protéomique/méthodes
4.
J Pathol ; 263(4-5): 418-428, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38795318

RÉSUMÉ

Neuroendocrine neoplasms (NENs) encompass tumors arising from neuroendocrine cells in various organs, including the gastrointestinal tract, pancreas, adrenal gland, and paraganglia. Despite advancements, accurately predicting the aggressiveness of gastroenteropancreatic (GEP) NENs based solely on pathological data remains challenging, thereby limiting optimal clinical management. Our previous research unveiled a crucial link between hypermethylation of the protocadherin PCDHGC3 gene and neuroendocrine tumors originating from the paraganglia and adrenal medulla. This epigenetic alteration was associated with increased metastatic potential and succinate dehydrogenase complex (SDH) dysfunction. Expanding upon this discovery, the current study explored PCDHGC3 gene methylation within the context of GEP-NENs in a cohort comprising 34 cases. We uncovered promoter hypermethylation of PCDHGC3 in 29% of GEP-NENs, with a significantly higher prevalence in gastrointestinal (GI) neuroendocrine carcinomas (NECs) compared with both pancreatic (Pan) NECs and neuroendocrine tumors (NETs) of GI and Pan origin. Importantly, these findings were validated in one of the largest multi-center GEP-NEN cohorts. Mechanistic analysis revealed that PCDHGC3 hypermethylation was not associated with SDH mutations or protein loss, indicating an SDH-independent epigenetic mechanism. Clinically, PCDHGC3 hypermethylation emerged as a significant prognostic factor, correlating with reduced overall survival rates in both patient cohorts. Significantly, whereas PCDHGC3 hypermethylation exhibited a strong correlation with TP53 somatic mutations, a hallmark of NEC, its predictive value surpassed that of TP53 mutations, with an area under the curve (AUC) of 0.95 (95% CI 0.83-1.0) for discriminating GI-NECs from GI-NETs, highlighting its superior predictive performance. In conclusion, our findings position PCDHGC3 methylation status as a promising molecular biomarker for effectively stratifying patients with GI-NENs. This discovery has the potential to advance patient care by enabling more precise risk assessments and tailored treatment strategies. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Sujet(s)
Marqueurs biologiques tumoraux , Carcinome neuroendocrine , Méthylation de l'ADN , Tumeurs de l'intestin , Humains , Marqueurs biologiques tumoraux/génétique , Carcinome neuroendocrine/génétique , Carcinome neuroendocrine/anatomopathologie , Mâle , Femelle , Tumeurs de l'intestin/génétique , Tumeurs de l'intestin/anatomopathologie , Adulte d'âge moyen , Cadhérines/génétique , Sujet âgé , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Épigenèse génétique , Régions promotrices (génétique) , Tumeurs neuroendocrines/génétique , Tumeurs neuroendocrines/anatomopathologie , Adulte
5.
J Vet Med Sci ; 86(7): 748-755, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38811188

RÉSUMÉ

Nuclear expression of ß-catenin has been reported in canine intestinal epithelial tumors (IETs) and colorectal inflammatory polyps (CIPs) with dysplastic epithelia. However, the role of the Wnt/ß-catenin signaling pathway in these lesions remains unclear. To investigate the association between the nuclear ß-catenin expression and the activation of the Wnt/ß-catenin signaling pathway, immunohistochemistry and mutation analyses were conducted on 64 IETs and 20 CIPs. IETs and CIPs with ß-catenin nuclear and/or cytoplasm immunolabeling were classified as ß-catenin (+). The immunostaining scores of c-Myc and Cyclin D1 and Ki-67 index were significantly higher in ß-catenin (+) cases than in ß-catenin (-) cases. Identical APC mutations (p.E154D and p.K155X) were detected in 6/41 ß-catenin (+) IETs; all 6 of IETs with APC mutations were Jack Russell Terriers. CTNNB1 mutations were detected in 29/42 ß-catenin (+) IETs, 3/11 ß-catenin (+) CIPs, and 2/22 ß-catenin (-) IETs, most of which were hotspots associated with human colorectal carcinoma. In one Miniature Dachshund diagnosed with a CIP that subsequently developed into an IET, the same CTNNB1 mutation was detected in both lesions. The immunohistochemical results suggest that cell proliferative activity in ß-catenin (+) cases may be associated with activation of the Wnt/ß-catenin signaling pathway. The mutation analysis results suggest that CTNNB1 mutations may be associated with cytoplasmic ß-catenin accumulation in IET and CIP. Furthermore, the dysplastic epithelium in CIP may progress to IET through the activation of the Wnt/ß-catenin signaling pathway by the CTNNB1 mutation.


Sujet(s)
Maladies des chiens , Mutation , Voie de signalisation Wnt , bêta-Caténine , Animaux , Chiens , bêta-Caténine/génétique , bêta-Caténine/métabolisme , Maladies des chiens/génétique , Maladies des chiens/anatomopathologie , Maladies des chiens/métabolisme , Voie de signalisation Wnt/génétique , Femelle , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Mâle , Immunohistochimie/médecine vétérinaire , Tumeurs de l'intestin/médecine vétérinaire , Tumeurs de l'intestin/génétique , Tumeurs de l'intestin/anatomopathologie , Tumeurs de l'intestin/métabolisme
6.
Br J Cancer ; 131(1): 159-170, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38729995

RÉSUMÉ

BACKGROUND: High-grade gastro-entero-pancreatic neoplasms (HG GEP-NENs) can be stratified according to their morphology and Ki-67 values into three prognostic classes: neuroendocrine tumors grade 3 (NETs G3), neuroendocrine carcinomas with Ki-67 < 55% (NECs <55) and NECs with Ki-67 ≥ 55% (NECs ≥55). METHODS: We analyzed a cohort of 49 HG GEP-NENs by targeted Next-Generation Sequencing (TrueSight Oncology 500), RNA-seq, and immunohistochemistry for p53, Rb1, SSTR-2A, and PD-L1. RESULTS: Frequent genomic alterations affected TP53 (26%), APC (20%), KRAS and MEN1 (both 11%) genes. NET G3 were enriched in MEN1 (p = 0.02) mutations, while both NECs groups were enriched in TP53 (p = 0.001), APC (p = 0.002) and KRAS (p = 0.02) mutations and tumors with TMB ≥ 10 muts/Mb (p = 0.01). No differentially expressed (DE) gene was found between NECs <55% and NECs ≥55%, while 1129 DE genes were identified between NET G3 and NECs. A slight enrichment of CD4+ and CD8+ T cells in NECs and of cancer-associated fibroblasts and macrophages (M2-like) in NET G3. Multivariate analysis identified histologic type and Rb1 loss as independent prognostic factors for overall survival. CONCLUSIONS: This study showed that GEP-NET G3 and GEP-NECs exhibit clear genomic and transcriptomic differences, differently from GEP-NECs <55% and GEP-NECs ≥55%, and provided molecular findings with prognostic and potentially predictive value.


Sujet(s)
Tumeurs neuroendocrines , Tumeurs du pancréas , Transcriptome , Humains , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Tumeurs neuroendocrines/génétique , Tumeurs neuroendocrines/anatomopathologie , Mutation , Adulte , Pronostic , Génomique/méthodes , Grading des tumeurs , Sujet âgé de 80 ans ou plus , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Marqueurs biologiques tumoraux/génétique , Tumeurs de l'intestin/génétique , Tumeurs de l'intestin/anatomopathologie
7.
Br J Cancer ; 131(1): 49-62, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38745088

RÉSUMÉ

BACKGROUND: Small bowel adenocarcinoma is a rare disease. The genomic profiling tumours according to clinical characteristics and its impact on the prognosis remains unclear. METHODS: A pooled analysis of clinical data, genomic profiling and MisMatch Repair (MMR) status from three databases was performed. RESULTS: A total of 188 tumour samples were analysed. A predisposing disease was reported in 22.3%, mainly Lynch syndrome and Crohn's disease. The tumours were localized in 80.2% and metastatic in 18.8%. The most frequent mutations were KRAS (42.0%) among them 7/79 are G12C, TP53 (40.4%), APC (19.1%), PIK3CA (18.6%), SMAD4 (12.8%) and ERBB2 (9.6%). Mutation distribution differed according to predisposing disease for TP53, ERBB2, IDH1, FGFR3, FGFR1 and KDR. KRAS and SMAD4 mutations were more frequent in metastatic tumour, whereas ERBB2 mutations were absent in metastatic tumour. For localized tumour, APC mutation was independently associated with a poor overall survival (OS) (p = 0.0254). 31.8% of localized tumours and 11.3% of metastatic tumours were dMMR (29.8% of the entire cohort). A dMMR status was associated with a better OS (HR = 0.61 [0.39-0.96], p = 0.0316). CONCLUSIONS: There is a different genomic profile according to the stage and predisposing disease. dMMR and APC mutation in localized tumour predict a better prognosis.


Sujet(s)
Adénocarcinome , Tumeurs de l'intestin , Mutation , Humains , Adénocarcinome/génétique , Adénocarcinome/anatomopathologie , Tumeurs de l'intestin/génétique , Tumeurs de l'intestin/anatomopathologie , Tumeurs de l'intestin/mortalité , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Intestin grêle/anatomopathologie , Adulte , Pronostic , Sujet âgé de 80 ans ou plus , Analyse de profil d'expression de gènes , Réparation de mésappariement de l'ADN/génétique
8.
Cell Rep ; 43(4): 113975, 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38507411

RÉSUMÉ

The intestine is a highly metabolic tissue, but the metabolic programs that influence intestinal crypt proliferation, differentiation, and regeneration are still emerging. Here, we investigate how mitochondrial sirtuin 4 (SIRT4) affects intestinal homeostasis. Intestinal SIRT4 loss promotes cell proliferation in the intestine following ionizing radiation (IR). SIRT4 functions as a tumor suppressor in a mouse model of intestinal cancer, and SIRT4 loss drives dysregulated glutamine and nucleotide metabolism in intestinal adenomas. Intestinal organoids lacking SIRT4 display increased proliferation after IR stress, along with increased glutamine uptake and a shift toward de novo nucleotide biosynthesis over salvage pathways. Inhibition of de novo nucleotide biosynthesis diminishes the growth advantage of SIRT4-deficient organoids after IR stress. This work establishes SIRT4 as a modulator of intestinal metabolism and homeostasis in the setting of DNA-damaging stress.


Sujet(s)
Prolifération cellulaire , Tumeurs de l'intestin , Intestins , Sirtuines , Animaux , Humains , Souris , Glutamine/métabolisme , Homéostasie , Muqueuse intestinale/métabolisme , Tumeurs de l'intestin/métabolisme , Tumeurs de l'intestin/anatomopathologie , Tumeurs de l'intestin/génétique , Intestins/métabolisme , Intestins/anatomopathologie , Souris de lignée C57BL , Protéines mitochondriales , Nucléotides/métabolisme , Organoïdes/métabolisme , Sirtuines/métabolisme
9.
Cancer Res Commun ; 4(1): 164-169, 2024 01 19.
Article de Anglais | MEDLINE | ID: mdl-38259096

RÉSUMÉ

The extent to which non-genetic environmental factors, such as diet, contribute to carcinogenesis has been long debated. One potential mechanism for the effects of environmental factors is through epigenetic modifications that affect gene expression without changing the underlying DNA sequence. However, the functional cooperation between dietary factors and cancer-causing epigenetic regulation is largely unknown. Here, we use a mouse model of age-dependent p16 epimutation, in which the p16 gene activity is directly controlled by promoter DNA methylation. We show p16 epimutation is modulated by folate and cofactors in dietary supplementation, which leads to increased colon cancer risk. Importantly, our findings provide functional evidence concerning the safety of folate fortification in the general population. SIGNIFICANCE: Our study demonstrates that dietary folate and cofactors modulate tumor-suppressor gene methylation to increase intestinal tumorigenesis. Our findings highlight the need for monitoring the long-term safety of folate fortification in high-risk individuals.


Sujet(s)
Carcinogenèse , Inhibiteur p16 de kinase cycline-dépendante , Épigenèse génétique , Tumeurs de l'intestin , Animaux , Humains , Souris , Carcinogenèse/génétique , Transformation cellulaire néoplasique , Régime alimentaire , Acide folique , Tumeurs de l'intestin/génétique , Inhibiteur p16 de kinase cycline-dépendante/génétique
10.
Histopathology ; 84(2): 255-265, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-37565289

RÉSUMÉ

Multiple recurrent genetic and epigenetic aberrations have been associated with worse prognosis in multiple studies of neuroendocrine tumours (NETs), but these have been mainly small cohorts and univariate analysis. This review and meta-analysis will focus upon the literature available on NETs of the gastrointestinal (GI) tract, liver, biliary tract and pancreas. PubMed and Embase were searched for publications that investigated the prognostic value of (epi)genetic changes of neuroendocrine tumours. A meta-analysis was performed assessing the association of the (epi)genetic alterations with overall survival (OS), disease-free survival (DFS) or locoregional control (LRC). In the pancreas DAXX/ATRX [hazard ratio (HR) = 3.29; 95% confidence interval (CI) = 2.28-4.74] and alternative lengthening telomeres (ALT) activation (HR = 8.20; 95% CI = 1.40-48.07) showed a pooled worse survival. In the small bowel NETs gains on chromosome 14 were associated with worse survival (HR 2.85; 95% CI = 1.40-5.81). NETs from different anatomical locations must be regarded as different biological entities with diverging molecular prognosticators, and epigenetic changes being important to the pathogenesis of these tumours. This review underpins the prognostic drivers of pancreatic NET which lie in mutations of DAXX/ATRX and ALT pathways. However, there is reaffirmation that prognostic molecular biomarkers of small bowel NETs should be sought in copy number variations (CNVs) rather than in single nucleotide variations (SNVs). This review also reveals how little is known about the prognostic significance of epigenetics in NETs.


Sujet(s)
Voies biliaires , Tumeurs de l'intestin , Tumeurs neuroendocrines , Tumeurs du pancréas , Humains , Tumeurs neuroendocrines/génétique , Tumeurs neuroendocrines/anatomopathologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Variations de nombre de copies de segment d'ADN , Pronostic , Tumeurs de l'intestin/génétique , Épigenèse génétique , Pancréas/anatomopathologie , Foie/anatomopathologie , Voies biliaires/anatomopathologie
11.
Aging Dis ; 15(1): 226-244, 2024 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-37962464

RÉSUMÉ

Diets that restrict caloric or protein intake offer a variety of benefits, including decreasing the incidence of cancer. However, whether such diets pose a substantial therapeutic benefit as auxiliary cancer treatments remains unclear. We determined the effects of severe protein depletion on tumorigenesis in a Drosophila melanogaster intestinal tumor model, using a human RAF gain-of-function allele. Severe and continuous protein restriction significantly reduced tumor growth but resulted in premature death. Therefore, we developed a diet in which short periods of severe protein restriction alternated cyclically with periods of complete feeding. This nutritional regime reduced tumor mass, restored gut functionality, and rescued the lifespan of oncogene-expressing flies to the levels observed in healthy flies on a continuous, fully nutritious diet. Furthermore, this diet reduced the chemotherapy-induced stem cell activity associated with tumor recurrence. Transcriptome analysis revealed long-lasting changes in the expression of key genes involved in multiple major developmental signaling pathways. Overall, the data suggest that recurrent severe protein depletion effectively mimics the health benefits of continuous protein restriction, without undesired nutritional shortcomings. This provides seminal insights into the mechanisms of the memory effect required to maintain the positive effects of protein restriction throughout the phases of a full diet. Finally, the repetitive form of strict protein restriction is an ideal strategy for adjuvant cancer therapy that is useful in many tumor contexts.


Sujet(s)
Drosophila , Tumeurs de l'intestin , Animaux , Humains , Longévité/génétique , Drosophila melanogaster/génétique , Restriction calorique , Récidive tumorale locale , Tumeurs de l'intestin/génétique
12.
Endocr Rev ; 45(3): 343-350, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38123518

RÉSUMÉ

Midgut neuroendocrine neoplasms (NENs) are one of the most common subtypes of NEN, and their incidence is rising globally. Despite being the most frequently diagnosed malignancy of the small intestine, little is known about their underlying molecular biology. Their unusually low mutational burden compared to other solid tumors and the unexplained occurrence of multifocal tumors makes the molecular biology of midgut NENs a particularly fascinating field of research. This review provides an overview of recent advances in the understanding of the interplay of the genetic, epigenetic, and transcriptomic landscape in the development of midgut NENs, a topic that is critical to understanding their biology and improving treatment options and outcomes for patients.


Sujet(s)
Tumeurs de l'intestin , Tumeurs neuroendocrines , Humains , Tumeurs neuroendocrines/génétique , Tumeurs neuroendocrines/métabolisme , Tumeurs de l'intestin/génétique , Tumeurs de l'intestin/métabolisme , Tumeurs de l'intestin/anatomopathologie , Animaux , Épigenèse génétique
13.
F1000Res ; 12: 417, 2023.
Article de Anglais | MEDLINE | ID: mdl-37954063

RÉSUMÉ

Background: Small bowel carcinoids are insidious tumors that are often metastatic when diagnosed. Limited mutation landscape studies of carcinoids indicate that these tumors have a relatively low mutational burden. The development of targeted therapies will depend upon the identification of mutations that drive the pathogenesis and metastasis of carcinoid tumors. Methods: Whole exome and RNA sequencing of 5 matched sets of normal tissue, primary small intestine carcinoid tumors, and liver metastases were investigated. Germline and somatic variants included: single nucleotide variants (SNVs), insertions/deletions (indels), structural variants, and copy number alterations (CNAs). The functional impact of mutations was predicted using Ensembl Variant Effect Predictor. Results: Large-scale CNAs were observed including the loss of chromosome 18 in all 5 metastases and 3/5 primary tumors. Certain somatic SNVs were metastasis-specific; including mutations in ATRX, CDKN1B, MXRA5 (leading to the activation of a cryptic splice site and loss of mRNA), SMARCA2, and the loss of UBE4B. Additional mutations in ATRX, and splice site loss of PYGL, leading to intron retention observed in primary and metastatic tumors. Conclusions: We observed novel mutations in primary/metastatic carcinoid tumor pairs, and some have been observed in other types of neuroendocrine tumors. We confirmed a previously observed loss of chromosome 18 and CDKN1B. Transcriptome sequencing added relevant information that would not have been appreciated with DNA sequencing alone. The detection of several splicing mutations on the DNA level and their consequences at the RNA level suggests that RNA splicing aberrations may be an important mechanism underlying carcinoid tumors.


Sujet(s)
Tumeur carcinoïde , Tumeurs de l'intestin , Tumeurs neuroendocrines , Humains , Multi-omique , Tumeur carcinoïde/génétique , Tumeur carcinoïde/anatomopathologie , Tumeur carcinoïde/secondaire , Tumeurs neuroendocrines/génétique , Tumeurs neuroendocrines/anatomopathologie , Tumeurs de l'intestin/génétique , Tumeurs de l'intestin/anatomopathologie , Ubiquitin-protein ligases
14.
Curr Oncol ; 30(10): 9244-9261, 2023 10 19.
Article de Anglais | MEDLINE | ID: mdl-37887568

RÉSUMÉ

Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) account for 80% of gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs). GEP-NETs are well-differentiated tumors, highly heterogeneous in biology and origin, and are often diagnosed at the metastatic stage. Diagnosis is commonly through clinical symptoms, histopathology, and PET-CT imaging, while molecular markers for metastasis and the primary site are unknown. Here, we report the identification of multi-gene signatures for hepatic metastasis and primary sites through analyses on RNA-SEQ datasets of pancreatic and small intestinal NETs tissue samples. Relevant gene features, identified from the normalized RNA-SEQ data using the mRMRe algorithm, were used to develop seven Machine Learning models (LDA, RF, CART, k-NN, SVM, XGBOOST, GBM). Two multi-gene random forest (RF) models classified primary and metastatic samples with 100% accuracy in training and test cohorts and >90% accuracy in an independent validation cohort. Similarly, three multi-gene RF models identified the pancreas or small intestine as the primary site with 100% accuracy in training and test cohorts, and >95% accuracy in an independent cohort. Multi-label models for concurrent prediction of hepatic metastasis and primary site returned >98.42% and >87.42% accuracies on training and test cohorts, respectively. A robust molecular signature to predict liver metastasis or the primary site for GEP-NETs is reported for the first time and could complement the clinical management of GEP-NETs.


Sujet(s)
Tumeurs de l'intestin , Tumeurs du foie , Tumeurs neuroendocrines , Humains , Tumeurs neuroendocrines/génétique , Tumeurs neuroendocrines/anatomopathologie , Tomographie par émission de positons couplée à la tomodensitométrie , Tumeurs de l'intestin/génétique , Tumeurs de l'intestin/anatomopathologie , Tumeurs du foie/génétique
15.
PLoS One ; 18(10): e0292643, 2023.
Article de Anglais | MEDLINE | ID: mdl-37824459

RÉSUMÉ

Among the small intestinal tumors that occur in irradiated mice of the established mouse model B6/B6-Chr18MSM-F1 ApcMin/+, loss of heterozygosity analysis can be utilized to estimate whether a deletion in the wild-type allele containing the Adenomatous polyposis coli (Apc) region (hereafter referred to as Deletion), a duplication in the mutant allele with a nonsense mutation at codon 850 of Apc (Duplication), or no aberration (Unidentified) has occurred. Previous research has revealed that the number of Unidentified tumors tends to increase with the radiation dose. In the present study, we investigated the molecular mechanisms underlying the development of an Unidentified tumor type in response to radiation exposure. The mRNA expression levels of Apc were significantly lower in Unidentified tumors than in normal tissues. We focused on epigenetic suppression as the mechanism underlying this decreased expression; however, hypermethylation of the Apc promoter region was not observed. To investigate whether deletions occur that cannot be captured by loss of heterozygosity analysis, we analyzed chromosome 18 using a customized array comparative genomic hybridization approach designed to detect copy-number changes in chromosome 18. However, the copy number of the Apc region was not altered in Unidentified tumors. Finally, gene mutation analysis of the Apc region using next-generation sequencing suggested the existence of a small deletion (approximately 3.5 kbp) in an Unidentified tumor from a mouse in the irradiated group. Furthermore, nonsense and frameshift mutations in Apc were found in approximately 30% of the Unidentified tumors analyzed. These results suggest that radiation-induced Unidentified tumors arise mainly due to decreased Apc expression of an unknown regulatory mechanism that does not depend on promoter hypermethylation, and that some tumors may result from nonsense mutations which are as-yet undefined point mutations.


Sujet(s)
Polypose adénomateuse colique , Tumeurs de l'intestin , Tumeurs radio-induites , Souris , Animaux , Gènes APC , Hybridation génomique comparative , Mutation , Polypose adénomateuse colique/génétique , Tumeurs de l'intestin/génétique , Tumeurs de l'intestin/anatomopathologie , Tumeurs radio-induites/génétique , Génomique
16.
Cell Rep ; 42(10): 113245, 2023 10 31.
Article de Anglais | MEDLINE | ID: mdl-37837622

RÉSUMÉ

Many tumors recapitulate the developmental and differentiation program of their tissue of origin, a basis for tumor cell heterogeneity. Although stem-cell-like tumor cells are well studied, the roles of tumor cells undergoing differentiation remain to be elucidated. We employ Drosophila genetics to demonstrate that the differentiation program of intestinal stem cells is crucial for enabling intestinal tumors to invade and induce non-tumor-autonomous phenotypes. The differentiation program that generates absorptive cells is aberrantly recapitulated in the intestinal tumors generated by activation of the Yap1 ortholog Yorkie. Inhibiting it allows stem-cell-like tumor cells to grow but suppresses invasiveness and reshapes various phenotypes associated with cachexia-like wasting by altering the expression of tumor-derived factors. Our study provides insight into how a native differentiation program determines a tumor's capacity to induce advanced cancer phenotypes and suggests that manipulating the differentiation programs co-opted in tumors might alleviate complications of cancer, including cachexia.


Sujet(s)
Drosophila , Tumeurs de l'intestin , Animaux , Cachexie/génétique , Différenciation cellulaire/génétique , Intestins/anatomopathologie , Tumeurs de l'intestin/génétique
17.
Sci Adv ; 9(39): eadd9668, 2023 09 29.
Article de Anglais | MEDLINE | ID: mdl-37756410

RÉSUMÉ

Neuroendocrine tumors (NETs) are rare cancers that most often arise in the gastrointestinal tract and pancreas. The fundamental mechanisms driving gastroenteropancreatic (GEP)-NET growth remain incompletely elucidated; however, the heterogeneous clinical behavior of GEP-NETs suggests that both cellular lineage dynamics and tumor microenvironment influence tumor pathophysiology. Here, we investigated the single-cell transcriptomes of tumor and immune cells from patients with gastroenteropancreatic NETs. Malignant GEP-NET cells expressed genes and regulons associated with normal, gastrointestinal endocrine cell differentiation, and fate determination stages. Tumor and lymphoid compartments sparsely expressed immunosuppressive targets commonly investigated in clinical trials, such as the programmed cell death protein-1/programmed death ligand-1 axis. However, infiltrating myeloid cell types within both primary and metastatic GEP-NETs were enriched for genes encoding other immune checkpoints, including VSIR (VISTA), HAVCR2 (TIM3), LGALS9 (Gal-9), and SIGLEC10. Our findings highlight the transcriptomic heterogeneity that distinguishes the cellular landscapes of GEP-NET anatomic subtypes and reveal potential avenues for future precision medicine therapeutics.


Sujet(s)
Tumeurs de l'intestin , Tumeurs neuroendocrines , Tumeurs du pancréas , Tumeurs de l'estomac , Humains , Tumeurs neuroendocrines/génétique , Tumeurs de l'intestin/génétique , Tumeurs de l'estomac/génétique , Tumeurs du pancréas/génétique , Microenvironnement tumoral/génétique
18.
Gastroenterology ; 165(4): 861-873, 2023 10.
Article de Anglais | MEDLINE | ID: mdl-37453564

RÉSUMÉ

BACKGROUND & AIMS: Small intestinal neuroendocrine tumor (SI-NET) is a rare disease, but its incidence has increased over the past 4 decades. Understanding the genetic risk factors underlying SI-NETs can help in disease prevention and may provide clinically beneficial markers for diagnosis. Here the results of the largest genome-wide association study of SI-NETs performed to date with 405 cases and 614,666 controls are reported. METHODS: Samples from 307 patients with SI-NETs and 287,137 controls in the FinnGen study were used for the identification of SI-NET risk-associated genetic variants. The results were also meta-analyzed with summary statistics from the UK Biobank (n = 98 patients with SI-NET and n = 327,529 controls). RESULTS: We identified 6 genome-wide significant (P < 5 × 10-8) loci associated with SI-NET risk, of which 4 (near SEMA6A, LGR5, CDKAL1, and FERMT2) are novel and 2 (near LTA4H-ELK and in KIF16B) have been reported previously. Interestingly, the top hit (rs200138614; P = 1.80 × 10-19) was a missense variant (p.Cys712Phe) in the LGR5 gene, a bona-fide marker of adult intestinal stem cells and a potentiator of canonical WNT signaling. The association was validated in an independent Finnish collection of 70 patients with SI-NETs, as well as in the UK Biobank exome sequence data (n = 92 cases and n = 392,814 controls). Overexpression of LGR5 p.Cys712Phe in intestinal organoids abolished the ability of R-Spondin1 to support organoid growth, indicating that the mutation perturbed R-Spondin-LGR5 signaling. CONCLUSIONS: Our study is the largest genome-wide association study to date on SI-NETs and reported 4 new associated genome-wide association study loci, including a novel missense mutation (rs200138614, p.Cys712Phe) in LGR5, a canonical marker of adult intestinal stem cells.


Sujet(s)
Tumeurs de l'intestin , Tumeurs neuroendocrines , Adulte , Humains , Tumeurs neuroendocrines/génétique , Tumeurs neuroendocrines/anatomopathologie , Mutation faux-sens , Étude d'association pangénomique , Tumeurs de l'intestin/génétique , Tumeurs de l'intestin/anatomopathologie , Récepteurs couplés aux protéines G/génétique , Kinésine/génétique
19.
Endocr Relat Cancer ; 30(10)2023 10 01.
Article de Anglais | MEDLINE | ID: mdl-37410378

RÉSUMÉ

High-grade gastroenteropancreatic (HG-GEP) neuroendocrine neoplasms (NENs) are highly aggressive cancers. The molecular etiology of these tumors remains unclear, and the prevalence of pathogenic germline variants in patients with HG-GEP NENs is unknown. We assessed sequencing data of 360 cancer genes in normal tissue from 240 patients with HG-GEP NENs; 198 patients with neuroendocrine carcinomas (NECs) and 42 with grade 3 neuroendocrine tumors (NET G3). Applying strict criteria, we identified pathogenic germline variants and compared the frequency with previously reported data from 33 different cancer types. We found a recurrent MYOC variant in three patients and a recurrent MUTYH variant in two patients, indicating that these genes may be important underlying risk factors for HG-GEP NENs when mutated. Further, germline variants were found in canonical tumor-suppressor genes, such as TP53, RB1, BRIP1 and BAP1. Overall, we found that 4.5% of patients with NEC and 9.5% of patients with NET G3 carry germline pathogenic or highly likely pathogenic variants. Applying identical criteria for variant classification in silico to mined data from 33 other cancer types, the median percentage of patients carrying pathogenic or highly likely pathogenic variants was 3.4% (range: 0-17%). The patients with NEC and pathogenic germline variants had a median overall survival of 9 months, similar to what is generally expected for metastatic GEP NECs. A patient with NET G3 and pathogenic MUTYH variant had much shorter overall survival than expected. The fraction of HG-GEP NENs with germline pathogenic variants is relatively high, but still <10%, meaning that that germline mutations cannot be the major underlying cause of HG-GEP NENs.


Sujet(s)
Carcinome neuroendocrine , Tumeurs gastro-intestinales , Tumeurs de l'intestin , Tumeurs neuroendocrines , Tumeurs du pancréas , Tumeurs de l'estomac , Humains , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Mutation germinale , Tumeurs gastro-intestinales/génétique , Tumeurs neuroendocrines/anatomopathologie , Carcinome neuroendocrine/anatomopathologie , Tumeurs de l'intestin/génétique , Tumeurs de l'intestin/anatomopathologie , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie
20.
Best Pract Res Clin Endocrinol Metab ; 37(5): 101794, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-37414651

RÉSUMÉ

Precision medicine describes a target-related approach to tailoring diagnosis and treatment of the individual patient. While this personalized approach is revoluzionizing many areas of oncology, it is quite late in the field of gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs), in which there are few molecular alterations to be therapeutically targeted. We critically reviewed the current evidence about precision medicine in GEP NENs, focusing on potential clinically relevant actionable targets for GEP NENs, such as the mTOR pathway, MGMT, hypoxia markers, RET, DLL-3, and some general agnostic targets. We analysed the main investigational approaches with solid and liquid biopsies. Furthermore, we reviewed a model of precision medicine more specific for NENs that is the theragnostic use of radionuclides. Overall, currently no true predictive factors for therapy have been validated so far in GEP NENs, and the personalized approach is based more on clinical thinking within a NEN-dedicated multidisciplinary team. However, there is a robust background to suppose that precision medicine, with the theragnostic model will yield new insights in this context soon.


Sujet(s)
Tumeurs gastro-intestinales , Tumeurs de l'intestin , Tumeurs neuroendocrines , Tumeurs du pancréas , Tumeurs de l'estomac , Humains , Médecine de précision , Tumeurs du pancréas/génétique , Tumeurs du pancréas/thérapie , Tumeurs gastro-intestinales/génétique , Tumeurs gastro-intestinales/thérapie , Tumeurs neuroendocrines/génétique , Tumeurs neuroendocrines/thérapie , Tumeurs neuroendocrines/diagnostic , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/thérapie , Tumeurs de l'estomac/métabolisme , Tumeurs de l'intestin/génétique , Tumeurs de l'intestin/thérapie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...