Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 11 de 11
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Nature ; 629(8013): 927-936, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38588697

RÉSUMÉ

Broad-spectrum RAS inhibition has the potential to benefit roughly a quarter of human patients with cancer whose tumours are driven by RAS mutations1,2. RMC-7977 is a highly selective inhibitor of the active GTP-bound forms of KRAS, HRAS and NRAS, with affinity for both mutant and wild-type variants3. More than 90% of cases of human pancreatic ductal adenocarcinoma (PDAC) are driven by activating mutations in KRAS4. Here we assessed the therapeutic potential of RMC-7977 in a comprehensive range of PDAC models. We observed broad and pronounced anti-tumour activity across models following direct RAS inhibition at exposures that were well-tolerated in vivo. Pharmacological analyses revealed divergent responses to RMC-7977 in tumour versus normal tissues. Treated tumours exhibited waves of apoptosis along with sustained proliferative arrest, whereas normal tissues underwent only transient decreases in proliferation, with no evidence of apoptosis. In the autochthonous KPC mouse model, RMC-7977 treatment resulted in a profound extension of survival followed by on-treatment relapse. Analysis of relapsed tumours identified Myc copy number gain as a prevalent candidate resistance mechanism, which could be overcome by combinatorial TEAD inhibition in vitro. Together, these data establish a strong preclinical rationale for the use of broad-spectrum RAS-GTP inhibition in the setting of PDAC and identify a promising candidate combination therapeutic regimen to overcome monotherapy resistance.


Sujet(s)
Antinéoplasiques , Carcinome du canal pancréatique , Guanosine triphosphate , Tumeurs du pancréas , Protéines proto-oncogènes p21(ras) , Animaux , Femelle , Humains , Souris , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Apoptose/effets des médicaments et des substances chimiques , Carcinome du canal pancréatique/traitement médicamenteux , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Variations de nombre de copies de segment d'ADN , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Gènes myc , Guanosine triphosphate/métabolisme , Souris de lignée BALB C , Souris de lignée C57BL , Récidive tumorale locale/traitement médicamenteux , Récidive tumorale locale/génétique , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/métabolisme , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes p21(ras)/métabolisme , Protéines proto-oncogènes p21(ras)/antagonistes et inhibiteurs , Résultat thérapeutique , Tests d'activité antitumorale sur modèle de xénogreffe , Mutation
2.
Cancer Discov ; 14(2): 348-361, 2024 Feb 08.
Article de Anglais | MEDLINE | ID: mdl-37966260

RÉSUMÉ

The sparse vascularity of pancreatic ductal adenocarcinoma (PDAC) presents a mystery: What prevents this aggressive malignancy from undergoing neoangiogenesis to counteract hypoxia and better support growth? An incidental finding from prior work on paracrine communication between malignant PDAC cells and fibroblasts revealed that inhibition of the Hedgehog (HH) pathway partially relieved angiosuppression, increasing tumor vascularity through unknown mechanisms. Initial efforts to study this phenotype were hindered by difficulties replicating the complex interactions of multiple cell types in vitro. Here we identify a cascade of paracrine signals between multiple cell types that act sequentially to suppress angiogenesis in PDAC. Malignant epithelial cells promote HH signaling in fibroblasts, leading to inhibition of noncanonical WNT signaling in fibroblasts and epithelial cells, thereby limiting VEGFR2-dependent activation of endothelial hypersprouting. This cascade was elucidated using human and murine PDAC explant models, which effectively retain the complex cellular interactions of native tumor tissues. SIGNIFICANCE: We present a key mechanism of tumor angiosuppression, a process that sculpts the physiologic, cellular, and metabolic environment of PDAC. We further present a computational and experimental framework for the dissection of complex signaling cascades that propagate among multiple cell types in the tissue environment. This article is featured in Selected Articles from This Issue, p. 201.


Sujet(s)
Carcinome du canal pancréatique , Tumeurs du pancréas , Animaux , Humains , Souris , Carcinome du canal pancréatique/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire , Protéines Hedgehog/génétique , Tumeurs du pancréas/anatomopathologie , Facteur de croissance endothéliale vasculaire de type A
3.
bioRxiv ; 2023 Dec 04.
Article de Anglais | MEDLINE | ID: mdl-38105998

RÉSUMÉ

Broad-spectrum RAS inhibition holds the potential to benefit roughly a quarter of human cancer patients whose tumors are driven by RAS mutations. However, the impact of inhibiting RAS functions in normal tissues is not known. RMC-7977 is a highly selective inhibitor of the active (GTP-bound) forms of KRAS, HRAS, and NRAS, with affinity for both mutant and wild type (WT) variants. As >90% of human pancreatic ductal adenocarcinoma (PDAC) cases are driven by activating mutations in KRAS, we assessed the therapeutic potential of RMC-7977 in a comprehensive range of PDAC models, including human and murine cell lines, human patient-derived organoids, human PDAC explants, subcutaneous and orthotopic cell-line or patient derived xenografts, syngeneic allografts, and genetically engineered mouse models. We observed broad and pronounced anti-tumor activity across these models following direct RAS inhibition at doses and concentrations that were well-tolerated in vivo. Pharmacological analyses revealed divergent responses to RMC-7977 in tumor versus normal tissues. Treated tumors exhibited waves of apoptosis along with sustained proliferative arrest whereas normal tissues underwent only transient decreases in proliferation, with no evidence of apoptosis. Together, these data establish a strong preclinical rationale for the use of broad-spectrum RAS inhibition in the setting of PDAC.

4.
bioRxiv ; 2023 Mar 21.
Article de Anglais | MEDLINE | ID: mdl-36993718

RÉSUMÉ

To identify novel drivers of malignancy in pancreatic ductal adenocarcinoma (PDAC), we employed regulatory network analysis, which calculates the activity of transcription factors and other regulatory proteins based on the integrated expression of their positive and negative target genes. We generated a regulatory network for the malignant epithelial cells of human PDAC using gene expression data from a set of 197 laser capture microdissected human PDAC samples and 45 low-grade precursors, for which we had matched histopathological, clinical, and epidemiological annotation. We then identified the most highly activated and repressed regulatory proteins (e.g. master regulators or MRs) associated with four malignancy phenotypes: precursors vs. PDAC (initiation), low-grade vs. high grade histopathology (progression), survival post resection, and association with KRAS activity. Integrating across these phenotypes, the top MR of PDAC malignancy was found to be BMAL2, a member of the PAS family of bHLH transcription factors. Although the canonical function of BMAL2 is linked to the circadian rhythm protein CLOCK, annotation of BMAL2 target genes highlighted a potential role in hypoxia response. We previously demonstrated that PDAC is hypovascularized and hypoperfused, and here show that PDAC from the genetically engineered KPC model exists in a state of extreme hypoxia, with a partial oxygen pressure of <1mmHg. Given the close homology of BMAL2 to HIF1ß (ARNT) and its potential to heterodimerize with HIF1A and HIF2A, we investigated whether BMAL2 plays a role in the hypoxic response of PDAC. Indeed, BMAL2 controlled numerous hypoxia response genes and could be inhibited following treatment with multiple RAF, MEK, and ERK inhibitors, validating its association with RAS activity. Knockout of BMAL2 in four human PDAC cell lines led to defects in growth and invasion in the setting of hypoxia. Strikingly, BMAL2 null cells failed to induce glycolysis upon exposure to severe hypoxia and this was associated with a loss of expression of the glycolytic enzyme LDHA. Moreover, HIF1A was no longer stabilized under hypoxia in BMAL2 knockout cells. By contrast, HIF2A was hyper-stabilized under hypoxia, indicating a dysregulation of hypoxia metabolism in response to BMAL2 loss. We conclude that BMAL2 is a master regulator of hypoxic metabolism in PDAC, serving as a molecular switch between the disparate metabolic roles of HIF1A- and HIF2A-dependent hypoxia responses.

5.
Biomed Pharmacother ; 158: 114162, 2023 Feb.
Article de Anglais | MEDLINE | ID: mdl-36571997

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive disease characterized by its metastatic potential and chemoresistance. These traits are partially attributable to the highly tumorigenic pancreatic cancer stem cells (PaCSCs). Interestingly, these cells show unique features in order to sustain their identity and functionality, some of them amenable for therapeutic intervention. Screening of phospho-receptor tyrosine kinases revealed that PaCSCs harbored increased activation of anaplastic lymphoma kinase (ALK). We subsequently demonstrated that oncogenic ALK signaling contributes to tumorigenicity in PDAC patient-derived xenografts (PDXs) by promoting stemness through ligand-dependent activation. Indeed, the ALK ligands midkine (MDK) or pleiotrophin (PTN) increased self-renewal, clonogenicity and CSC frequency in several in vitro local and metastatic PDX models. Conversely, treatment with the clinically-approved ALK inhibitors Crizotinib and Ensartinib decreased PaCSC content and functionality in vitro and in vivo, by inducing cell death. Strikingly, ALK inhibitors sensitized chemoresistant PaCSCs to Gemcitabine, as the most used chemotherapeutic agent for PDAC treatment. Consequently, ALK inhibition delayed tumor relapse after chemotherapy in vivo by effectively decreasing the content of PaCSCs. In summary, our results demonstrate that targeting the MDK/PTN-ALK axis with clinically-approved inhibitors impairs in vivo tumorigenicity and chemoresistance in PDAC suggesting a new treatment approach to improve the long-term survival of PDAC patients.


Sujet(s)
Carcinome du canal pancréatique , Tumeurs du pancréas , Humains , Kinase du lymphome anaplasique , Résistance aux médicaments antinéoplasiques , Récidive tumorale locale , Récepteurs à activité tyrosine kinase , Carcinome du canal pancréatique/traitement médicamenteux , Carcinome du canal pancréatique/anatomopathologie , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/anatomopathologie , Lignée cellulaire tumorale , Tumeurs du pancréas
6.
STAR Protoc ; 3(4): 101899, 2022 12 16.
Article de Anglais | MEDLINE | ID: mdl-36595938

RÉSUMÉ

Orthotopic patient-derived xenograft models recapitulate the genomic complexity of the original tumor and some aspects of local microenvironment, useful for rapid development and validation of personalized treatment strategies. Here, we precisely describe a protocol for generating tumor slices from human or murine-derived pancreatic cancer. They are then implanted directly into the murine pancreas, monitored using ultrasound, with a 90% success rate. This assay creates a clinically relevant in vivo model facilitating personalized treatment development.


Sujet(s)
Tumeurs du pancréas , Humains , Animaux , Souris , Hétérogreffes , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Pancréas/anatomopathologie , Microenvironnement tumoral , Tumeurs du pancréas
7.
Mol Cancer Ther ; 20(6): 1029-1038, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-33846235

RÉSUMÉ

Glioblastoma (GBM) is the most frequent and aggressive primary tumor type in the central nervous system in adults. Resistance to chemotherapy remains one of the major obstacles in GBM treatment. Identifying and overcoming the mechanisms of therapy resistance is instrumental to develop novel therapeutic approaches for patients with GBM. To determine the major drivers of temozolomide (TMZ) sensitivity, we performed shRNA screenings in GBM lines with different O6-methylguanine-DNA methyl-transferase (MGMT) status. We then evaluated dianhydrogalactitol (Val-083), a small alkylating molecule that induces interstrand DNA crosslinking, as a potential treatment to bypass TMZ-resistance mechanisms. We found that loss of mismatch repair (MMR) components and MGMT expression are mutually exclusive mechanisms driving TMZ resistance in vitro Treatment of established GBM cells and tumorsphere lines with Val-083 induces DNA damage and cell-cycle arrest in G2-M phase, independently of MGMT or MMR status, thus circumventing conventional resistance mechanisms to TMZ. Combination of TMZ and Val-083 shows a synergic cytotoxic effect in tumor cells in vitro, ex vivo, and in vivo We propose this combinatorial treatment as a potential approach for patients with GBM.


Sujet(s)
Dianhydro-galactitol/usage thérapeutique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Glioblastome/traitement médicamenteux , Témozolomide/pharmacologie , Animaux , Lignée cellulaire tumorale , Dianhydro-galactitol/pharmacologie , Humains , Souris , Transfection , Tests d'activité antitumorale sur modèle de xénogreffe
8.
Trends Mol Med ; 25(5): 361-362, 2019 05.
Article de Anglais | MEDLINE | ID: mdl-30878400

RÉSUMÉ

Genetically engineered mouse models have revolutionized the study of pancreatic cancer, but have several technical and practical limitations. A new adeno-associated virus (AAV)-driven somatic genome-editing model of pancreatic ductal adenocarcinoma reported by Ideno et al. (Lab. Invest. published online February 6, 2019; https://doi.org/10.1038/s41374-018-0171-z) addresses several of these limitations, achieving rapid and penetrant induction of multiple targeted alterations in the adult murine pancreas.


Sujet(s)
Dependovirus/génétique , Édition de gène , Études d'associations génétiques , Prédisposition génétique à une maladie , Vecteurs génétiques/génétique , Tumeurs du pancréas/étiologie , Animaux , Marqueurs biologiques tumoraux , Carcinome du canal pancréatique/étiologie , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/anatomopathologie , Édition de gène/méthodes , Humains , Souris , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie
9.
Nat Commun ; 9(1): 1466, 2018 04 13.
Article de Anglais | MEDLINE | ID: mdl-29654229

RÉSUMÉ

To accurately recapitulate the heterogeneity of human diseases, animal models require to recreate multiple complex genetic alterations. Here, we combine the RCAS-TVA system with the CRISPR-Cas9 genome editing tools for precise modeling of human tumors. We show that somatic deletion in neural stem cells of a variety of known tumor suppressor genes (Trp53, Cdkn2a, and Pten) leads to high-grade glioma formation. Moreover, by simultaneous delivery of pairs of guide RNAs we generate different gene fusions with oncogenic potential, either by chromosomal deletion (Bcan-Ntrk1) or by chromosomal translocation (Myb-Qk). Lastly, using homology-directed-repair, we also produce tumors carrying the homologous mutation to human BRAF V600E, frequently identified in a variety of tumors, including different types of gliomas. In summary, we have developed an extremely versatile mouse model for in vivo somatic genome editing, that will elicit the generation of more accurate cancer models particularly appropriate for pre-clinical testing.


Sujet(s)
Tumeurs du cerveau/génétique , Systèmes CRISPR-Cas , Édition de gène , /génétique , Animaux , Antigènes néoplasiques/génétique , Benzamides/pharmacologie , Tumeurs du cerveau/traitement médicamenteux , Brévicane/génétique , Réparation de l'ADN , Faux positifs , Fréquence d'allèle , Techniques de transfert de gènes , Gliome/métabolisme , Humains , Hybridation fluorescente in situ , Indazoles/pharmacologie , Souris , Souris SCID , Souris transgéniques , Mutation , Cellules NIH 3T3 , Récepteur trkA/génétique
10.
Nat Commun ; 8(1): 2035, 2017 12 11.
Article de Anglais | MEDLINE | ID: mdl-29229958

RÉSUMÉ

Transport of macromolecules through the nuclear pore by importins and exportins plays a critical role in the spatial regulation of protein activity. How cancer cells co-opt this process to promote tumorigenesis remains unclear. The epidermal growth factor receptor (EGFR) plays a critical role in normal development and in human cancer. Here we describe a mechanism of EGFR regulation through the importin ß family member RAN-binding protein 6 (RanBP6), a protein of hitherto unknown functions. We show that RanBP6 silencing impairs nuclear translocation of signal transducer and activator of transcription 3 (STAT3), reduces STAT3 binding to the EGFR promoter, results in transcriptional derepression of EGFR, and increased EGFR pathway output. Focal deletions of the RanBP6 locus on chromosome 9p were found in a subset of glioblastoma (GBM) and silencing of RanBP6 promoted glioma growth in vivo. Our results provide an example of EGFR deregulation in cancer through silencing of components of the nuclear import pathway.


Sujet(s)
Récepteurs ErbB/génétique , Régulation de l'expression des gènes tumoraux , Gliome/génétique , Caryophérines bêta/génétique , Protéine G ran/génétique , Transport nucléaire actif/génétique , Animaux , Antibiotiques antinéoplasiques/pharmacologie , Lignée cellulaire tumorale , Cellules cultivées , Doxorubicine/pharmacologie , Récepteurs ErbB/métabolisme , Rétrocontrôle physiologique , Femelle , Techniques de knock-down de gènes , Gliome/traitement médicamenteux , Gliome/métabolisme , Cellules HEK293 , Humains , Souris knockout , Souris SCID , Facteur de transcription STAT-3/génétique , Facteur de transcription STAT-3/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe , Caryophérines bêta/métabolisme , Protéine G ran/métabolisme
11.
Hum Mutat ; 36(2): 210-21, 2015 Feb.
Article de Anglais | MEDLINE | ID: mdl-25382762

RÉSUMÉ

Numerous pathogenic DNA variants impair the splicing mechanism in human genetic diseases. Minigenes are optimal approaches to test variants under the splicing viewpoint without the need of patient samples. We aimed to design a robust minigene construct of the breast cancer gene BRCA2 in order to investigate the impact of variants on splicing. BRCA2 exons 19-27 (MGBR2_ex19-27) were cloned in the new vector pSAD. It produced a large transcript of the expected size (2,174 nucleotides) and exon structure (V1-ex19-27-V2). Splicing assays showed that 18 (17 splice-site and 1 silencer variants) out of 40 candidate DNA variants induced aberrant patterns. Twenty-four anomalous transcripts were accurately detected by fluorescent-RT-PCR that were generated by exon-skipping, alternative site usage, and intron-retention events. Fourteen variants induced major anomalies and were predicted to disrupt protein function so they could be classified as pathogenic. Furthermore, minigene mimicked previously reported patient RNA outcomes of seven variants supporting the reproducibility of minigene assays. Therefore, a relevant fraction of variants are involved in breast cancer through splicing alterations. MGBR2_ex19-27 is the largest reported BRCA2 minigene and constitutes a valuable tool for the functional and clinical classification of sequence variations.


Sujet(s)
Épissage alternatif , Protéine BRCA2/génétique , Protéine BRCA2/métabolisme , Exons , Études d'associations génétiques , Cellules HeLa , Syndrome héréditaire de cancer du sein et de l'ovaire/génétique , Humains , Cellules MCF-7 , Isoformes de protéines/génétique , Isoformes de protéines/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...