Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 27
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Cell Death Dis ; 15(4): 255, 2024 Apr 10.
Article de Anglais | MEDLINE | ID: mdl-38600086

RÉSUMÉ

Excessive STAT3 signalling via gp130, the shared receptor subunit for IL-6 and IL-11, contributes to disease progression and poor survival outcomes in patients with colorectal cancer. Here, we provide evidence that bazedoxifene inhibits tumour growth via direct interaction with the gp130 receptor to suppress IL-6 and IL-11-mediated STAT3 signalling. Additionally, bazedoxifene combined with chemotherapy synergistically reduced cell proliferation and induced apoptosis in patient-derived colon cancer organoids. We elucidated that the primary mechanism of anti-tumour activity conferred by bazedoxifene treatment occurs via pro-apoptotic responses in tumour cells. Co-treatment with bazedoxifene and the SMAC-mimetics, LCL161 or Birinapant, that target the IAP family of proteins, demonstrated increased apoptosis and reduced proliferation in colorectal cancer cells. Our findings provide evidence that bazedoxifene treatment could be combined with SMAC-mimetics and chemotherapy to enhance tumour cell apoptosis in colorectal cancer, where gp130 receptor signalling promotes tumour growth and progression.


Sujet(s)
Tumeurs du côlon , Indoles , Interleukine-11 , Humains , Interleukine-11/usage thérapeutique , Lignée cellulaire tumorale , Interleukine-6/métabolisme , Récepteur gp130 de cytokines/métabolisme , Tumeurs du côlon/traitement médicamenteux , Apoptose
2.
Life Sci Alliance ; 7(2)2024 02.
Article de Anglais | MEDLINE | ID: mdl-37957015

RÉSUMÉ

Deregulation of the Hippo pathway is a driver for cancer progression and treatment resistance. In the context of gastric cancer, YAP1 is a biomarker for poor patient prognosis. Although genomic tumor profiling provides information of Hippo pathway activation, the present study demonstrates that inhibition of Yap1 activity has anti-tumor effects in gastric tumors driven by oncogenic mutations and inflammatory cytokines. We show that Yap1 is a key regulator of cell metabolism, proliferation, and immune responses in normal and neoplastic gastric epithelium. We propose that the Hippo pathway is targetable across gastric cancer subtypes and its therapeutic benefits are likely to be mediated by both cancer cell-intrinsic and -extrinsic mechanisms.


Sujet(s)
Tumeurs de l'estomac , Humains , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/anatomopathologie , Microenvironnement tumoral , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Voie de signalisation Hippo , Facteur de transcription STAT-3/métabolisme
3.
Trends Immunol ; 44(12): 971-985, 2023 12.
Article de Anglais | MEDLINE | ID: mdl-37995659

RÉSUMÉ

Macrophages represent a key component of the tumor microenvironment (TME) and are largely associated with poor prognosis. Therapeutic targeting of macrophages has historically focused on inhibiting their recruitment or reprogramming their phenotype from a protumor (M2-like) to an antitumor (M1-like) one. Unfortunately, this approach has not provided clinical breakthroughs that have changed practice. Emerging studies utilizing single-cell RNA-sequencing (scRNA-seq) and spatial transcriptomics have improved our understanding of the ontogeny, phenotype, and functional plasticity of macrophages. Overlaying the wealth of current information regarding macrophage molecular subtypes and functions has also identified novel therapeutic vulnerabilities that might drive better control of tumor-associated macrophages (TAMs). Here, we discuss the functional profiling of macrophages and provide an update of novel macrophage-targeted therapies in development.


Sujet(s)
Tumeurs , Humains , Tumeurs/thérapie , Tumeurs/anatomopathologie , Macrophages/anatomopathologie , Phénotype , Microenvironnement tumoral
4.
Nat Commun ; 14(1): 6872, 2023 10 28.
Article de Anglais | MEDLINE | ID: mdl-37898600

RÉSUMÉ

Although gastric cancer is a leading cause of cancer-related deaths, systemic treatment strategies remain scarce. Here, we report the pro-tumorigenic properties of the crosstalk between intestinal tuft cells and type 2 innate lymphoid cells (ILC2) that is evolutionarily optimized for epithelial remodeling in response to helminth infection. We demonstrate that tuft cell-derived interleukin 25 (IL25) drives ILC2 activation, inducing the release of IL13 and promoting epithelial tuft cell hyperplasia. While the resulting tuft cell - ILC2 feed-forward circuit promotes gastric metaplasia and tumor formation, genetic depletion of tuft cells or ILC2s, or therapeutic targeting of IL13 or IL25 alleviates these pathologies in mice. In gastric cancer patients, tuft cell and ILC2 gene signatures predict worsening survival in intestinal-type gastric cancer where ~40% of the corresponding cancers show enriched co-existence of tuft cells and ILC2s. Our findings suggest a role for ILC2 and tuft cells, along with their associated cytokine IL13 and IL25 as gatekeepers and enablers of metaplastic transformation and gastric tumorigenesis, thereby providing an opportunity to therapeutically inhibit early-stage gastric cancer through repurposing antibody-mediated therapies.


Sujet(s)
Immunité innée , Tumeurs de l'estomac , Humains , Souris , Animaux , Interleukine-13/métabolisme , Tumeurs de l'estomac/anatomopathologie , Lymphocytes/métabolisme , Hyperplasie/métabolisme , Métaplasie/métabolisme
6.
Oncogene ; 42(22): 1786-1801, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-37120696

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignant disease with a 5-year survival rate of <10%. Aberrant activation or elevated expression of the tyrosine kinase c-SRC (SRC) is frequently observed in PDAC and is associated with a poor prognosis. Preclinical studies have revealed a multifaceted role for SRC activation in PDAC, including promoting chronic inflammation, tumor cell proliferation and survival, cancer cell stemness, desmoplasia, hypoxia, angiogenesis, invasion, metastasis, and drug resistance. Strategies to inhibit SRC signaling include suppressing its catalytic activity, inhibiting protein stability, or by interfering with signaling components of the SRC signaling pathway including suppressing protein interactions of SRC. In this review, we discuss the molecular and immunological mechanisms by which aberrant SRC activity promotes PDAC tumorigenesis. We also provide a comprehensive update of SRC inhibitors in the clinic, and discuss the clinical challenges associated with targeting SRC in pancreatic cancer.


Sujet(s)
Carcinome du canal pancréatique , Tumeurs du pancréas , Humains , Lignée cellulaire tumorale , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/génétique , Tumeurs du pancréas/métabolisme , Transduction du signal , Carcinome du canal pancréatique/génétique , Prolifération cellulaire , Mouvement cellulaire , Régulation de l'expression des gènes tumoraux , Tumeurs du pancréas
7.
STAR Protoc ; 4(1): 102110, 2023 03 17.
Article de Anglais | MEDLINE | ID: mdl-36853665

RÉSUMÉ

Tumor-derived organoids are valuable for testing anti-cancer drugs in vitro, but existing lysis-based protocols for viability measurement are laborious and restricted at a single time point. Here, we provide a lysis-free protocol for longitudinal and rapid assessment of mouse gastric tumor organoid viability and growth. We describe organoid plating, viability assessment via luminescence measurement, quantification of organoid growth by microscopy imaging, and treatment of organoids with test compounds to evaluate the effects on viability and growth at various time points.


Sujet(s)
Antinéoplasiques , Tumeurs de l'estomac , Animaux , Souris , Microscopie , Luminescence , Antinéoplasiques/pharmacologie , Organoïdes/anatomopathologie
8.
STAR Protoc ; 4(1): 102021, 2023 03 17.
Article de Anglais | MEDLINE | ID: mdl-36638017

RÉSUMÉ

Here, we provide a protocol for an intrasplenic injection model to establish pancreatic tumors in the mouse liver. We describe the steps to inject tumor cells into mouse spleen and to perform a splenectomy, followed by animal recovery and end point analysis of tumors in the liver. This model allows rapid and reproducible tumor growth in a clinically relevant metastatic site, providing a platform to evaluate the efficacy of anti-cancer drugs. This technique can be expanded to other cancer cell lines. For complete details on the use and execution of this protocol, please refer to Poh et al. (2022).1.


Sujet(s)
Tumeurs du foie , Tumeurs du pancréas , Souris , Animaux , Transplantation tumorale , Tumeurs du pancréas/anatomopathologie , Tumeurs du foie/anatomopathologie , Tumeurs du pancréas
9.
Cells ; 11(24)2022 12 17.
Article de Anglais | MEDLINE | ID: mdl-36552868

RÉSUMÉ

Aberrant expression of the oncoprotein c-Myc (Myc) is frequently observed in solid tumors and is associated with reduced overall survival. In addition to well-recognized cancer cell-intrinsic roles of Myc, studies have also suggested tumor-promoting roles for Myc in cells of the tumor microenvironment, including macrophages and other myeloid cells. Here, we benchmark Myc inactivation in tumor cells against the contribution of its expression in myeloid cells of murine hosts that harbor endogenous or allograft tumors. Surprisingly, we observe that LysMCre-mediated Myc ablation in host macrophages does not attenuate tumor growth regardless of immunogenicity, the cellular origin of the tumor, the site it develops, or the stage along the tumor progression cascade. Likewise, we find no evidence for Myc ablation to revert or antagonize the polarization of alternatively activated immunosuppressive macrophages. Thus, we surmise that systemic targeting of Myc activity may confer therapeutic benefits primarily through limiting Myc activity in tumor cells rather than reinvigorating the anti-tumor activity of macrophages.


Sujet(s)
Macrophages , Tumeurs , Souris , Animaux , Macrophages/métabolisme , Tumeurs/métabolisme , Cellules myéloïdes/métabolisme , Microenvironnement tumoral
10.
Cell Rep ; 41(2): 111479, 2022 10 11.
Article de Anglais | MEDLINE | ID: mdl-36223746

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with a low 5-year survival rate and is associated with poor response to therapy. Elevated expression of the myeloid-specific hematopoietic cell kinase (HCK) is observed in PDAC and correlates with reduced patient survival. To determine whether aberrant HCK signaling in myeloid cells is involved in PDAC growth and metastasis, we established orthotopic and intrasplenic PDAC tumors in wild-type and HCK knockout mice. Genetic ablation of HCK impaired PDAC growth and metastasis by inducing an immune-stimulatory endotype in myeloid cells, which in turn reduced the desmoplastic microenvironment and enhanced cytotoxic effector cell infiltration. Consequently, genetic ablation or therapeutic inhibition of HCK minimized metastatic spread, enhanced the efficacy of chemotherapy, and overcame resistance to anti-PD1, anti-CTLA4, or stimulatory anti-CD40 immunotherapy. Our results provide strong rationale for HCK to be developed as a therapeutic target to improve the response of PDAC to chemo- and immunotherapy.


Sujet(s)
Carcinome du canal pancréatique , Tumeurs du pancréas , Protéines proto-oncogènes c-hck , Animaux , Carcinome du canal pancréatique/génétique , Souris , Cellules myéloïdes/métabolisme , Tumeurs du pancréas/anatomopathologie , Protéines proto-oncogènes c-hck/génétique , Microenvironnement tumoral , Tumeurs du pancréas
11.
Sci Adv ; 8(25): eabl7882, 2022 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-35731867

RÉSUMÉ

Although immunotherapy has revolutionized cancer treatment, many immunogenic tumors remain refractory to treatment. This can be largely attributed to an immunologically "cold" tumor microenvironment characterized by an accumulation of immunosuppressive myeloid cells and exclusion of activated T cells. Here, we demonstrate that genetic ablation or therapeutic inhibition of the myeloid-specific hematopoietic cell kinase (HCK) enables activity of antagonistic anti-programmed cell death protein 1 (anti-PD1), anti-CTLA4, or agonistic anti-CD40 immunotherapies in otherwise refractory tumors and augments response in treatment-susceptible tumors. Mechanistically, HCK ablation reprograms tumor-associated macrophages and dendritic cells toward an inflammatory endotype and enhances CD8+ T cell recruitment and activation when combined with immunotherapy in mice. Meanwhile, therapeutic inhibition of HCK in humanized mice engrafted with patient-derived xenografts counteracts tumor immunosuppression, improves T cell recruitment, and impairs tumor growth. Collectively, our results suggest that therapeutic targeting of HCK activity enhances response to immunotherapy by simultaneously stimulating immune cell activation and inhibiting the immunosuppressive tumor microenvironment.

12.
Cancers (Basel) ; 13(24)2021 Dec 11.
Article de Anglais | MEDLINE | ID: mdl-34944848

RÉSUMÉ

Non-small cell lung cancer (NSCLC) is the most common type of lung cancer and accounts for 85% of lung cancer cases. Aberrant activation of the Signal Transducer and Activator of Transcription 3 (STAT3) is frequently observed in NSCLC and is associated with a poor prognosis. Pre-clinical studies have revealed an unequivocal role for tumor cell-intrinsic and extrinsic STAT3 signaling in NSCLC by promoting angiogenesis, cell survival, cancer cell stemness, drug resistance, and evasion of anti-tumor immunity. Several STAT3-targeting strategies have also been investigated in pre-clinical models, and include preventing upstream receptor/ligand interactions, promoting the degradation of STAT3 mRNA, and interfering with STAT3 DNA binding. In this review, we discuss the molecular and immunological mechanisms by which persistent STAT3 activation promotes NSCLC development, and the utility of STAT3 as a prognostic and predictive biomarker in NSCLC. We also provide a comprehensive update of STAT3-targeting therapies that are currently undergoing clinical evaluation, and discuss the challenges associated with these treatment modalities in human patients.

13.
Cancers (Basel) ; 13(12)2021 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-34201127

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignant disease with a 5-year survival rate of less than 10%. Macrophages are one of the earliest infiltrating cells in the pancreatic tumor microenvironment, and are associated with an increased risk of disease progression, recurrence, metastasis, and shorter overall survival. Pre-clinical studies have demonstrated an unequivocal role of macrophages in PDAC by contributing to chronic inflammation, cancer cell stemness, desmoplasia, immune suppression, angiogenesis, invasion, metastasis, and drug resistance. Several macrophage-targeting therapies have also been investigated in pre-clinical models, and include macrophage depletion, inhibiting macrophage recruitment, and macrophage reprogramming. However, the effectiveness of these drugs in pre-clinical models has not always translated into clinical trials. In this review, we discuss the molecular mechanisms that underpin macrophage heterogeneity within the pancreatic tumor microenvironment, and examine the contribution of macrophages at various stages of PDAC progression. We also provide a comprehensive update of macrophage-targeting therapies that are currently undergoing clinical evaluation, and discuss clinical challenges associated with these treatment modalities in human PDAC patients.

15.
Cell Death Discov ; 7(1): 122, 2021 May 28.
Article de Anglais | MEDLINE | ID: mdl-34050131

RÉSUMÉ

Malignant pleural mesothelioma (MPM) is an aggressive cancer with treatment limited to Cisplatin and Pemetrexed chemotherapy. Recently, we showed that drugs targeting the BCL-2-regulated apoptosis pathway could kill MPM cell lines in vitro, and control tumor growth in vivo. These studies showed BCL-XL was the dominant pro-survival BCL-2 family member correlating with its high-level expression in cells and patient tumor samples. In this study we show another inhibitor, AZD4320 that targets BCL-XL (and BCL-2), can also potently kill MPM tumor cells in vitro (EC50 values in the 200 nM range) and this effect is enhanced by co-inhibition of MCL-1 using AZD5991. Moreover, we show that a novel nanoparticle, AZD0466, where AZD4320 is chemically conjugated to a PEGylated poly-lysine dendrimer, was as effective as standard-of-care chemotherapy, Cisplatin, at inhibiting tumor growth in mouse xenograft studies, and this effect was enhanced when both drugs were combined. Critically, the degree of thrombocytopenia, an on-target toxicity associated with BCL-XL inhibition, was significantly reduced throughout the treatment period compared to other BCL-XL-targeting BH3-mimetics. These pre-clinical findings provide a rationale for the future clinical evaluation for novel BH3-mimetic formulations in MPM, and indeed, other solid tumor types dependent on BCL-XL.

16.
Cell Death Discov ; 6(1): 114, 2020 Oct 31.
Article de Anglais | MEDLINE | ID: mdl-33298868

RÉSUMÉ

Despite having one of the lowest survival rates of all cancers, there have been no new approved treatments for malignant pleural mesothelioma (MPM) in over a decade. Standard-of-care treatment relies on Cisplatin plus Pemetrexed chemotherapy. Here, we tested a suite of BH3-mimetic drugs targeting BCL-2 pro-survival proteins of the intrinsic apoptotic pathway. We found BCL-XL is the dominant pro-survival protein in a panel of cell lines in vitro, though potent, synergistic cell killing occurred with MCL-1 co-targeting. This correlates with high-level expression of BCL-XL and MCL-1 in cell lines and a large cohort of patient tumour samples. BCL-XL inhibition combined with Cisplatin also enhanced cell killing. In vivo BCL-XL inhibition was as effective as Cisplatin, and the combination enhanced tumour growth control and survival. Genetic ablation of MCL-1 also enhanced the effects of BCL-XL inhibitors, in vivo. Combined, these data provide a compelling rationale for the clinical investigation of BH3-mimetics targeting BCL-XL in MPM.

17.
Cancer Immunol Res ; 8(4): 428-435, 2020 04.
Article de Anglais | MEDLINE | ID: mdl-31992566

RÉSUMÉ

Persistent activation of the latent transcription factor STAT3 is observed in gastric tumor epithelial and immune cells and is associated with a poor patient prognosis. Although targeting STAT3-activating upstream kinases offers therapeutically viable targets with limited specificity, direct inhibition of STAT3 remains challenging. Here we provide functional evidence that myeloid-specific hematopoietic cell kinase (HCK) activity can drive STAT3-dependent epithelial tumor growth in mice and is associated with alternative macrophage activation alongside matrix remodeling and tumor cell invasion. Accordingly, genetic reduction of HCK expression in bone marrow-derived cells or systemic pharmacologic inhibition of HCK activity suppresses alternative macrophage polarization and epithelial STAT3 activation, and impairs tumor growth. These data validate HCK as a molecular target for the treatment of human solid tumors harboring excessive STAT3 activity.


Sujet(s)
Protéines proto-oncogènes c-hck/antagonistes et inhibiteurs , Pyrimidines/pharmacologie , Pyrroles/pharmacologie , Facteur de transcription STAT-3/métabolisme , Tumeurs de l'estomac/traitement médicamenteux , Animaux , Femelle , Humains , Activation des macrophages/effets des médicaments et des substances chimiques , Mâle , Souris , Souris transgéniques , Phosphorylation/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-hck/métabolisme , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/anatomopathologie , Taux de survie
18.
Cell Death Differ ; 27(2): 742-757, 2020 02.
Article de Anglais | MEDLINE | ID: mdl-31296963

RÉSUMÉ

Gastrointestinal epithelial cells provide a selective barrier that segregates the host immune system from luminal microorganisms, thereby contributing directly to the regulation of homeostasis. We have shown that from early embryonic development Bcl-G, a Bcl-2 protein family member with unknown function, was highly expressed in gastrointestinal epithelial cells. While Bcl-G was dispensable for normal growth and development in mice, the loss of Bcl-G resulted in accelerated progression of colitis-associated cancer. A label-free quantitative proteomics approach revealed that Bcl-G may contribute to the stability of a mucin network, which when disrupted, is linked to colon tumorigenesis. Consistent with this, we observed a significant reduction in Bcl-G expression in human colorectal tumors. Our study identifies an unappreciated role for Bcl-G in colon cancer.


Sujet(s)
Tumeurs colorectales/métabolisme , Inflammation/métabolisme , Protéines proto-oncogènes c-bcl-2/métabolisme , Animaux , Colite/métabolisme , Colite/anatomopathologie , Tumeurs colorectales/anatomopathologie , Humains , Inflammation/anatomopathologie , Souris , Souris knockout , Protéines proto-oncogènes c-bcl-2/déficit , Protéines proto-oncogènes c-bcl-2/génétique
19.
Nat Commun ; 10(1): 2735, 2019 06 21.
Article de Anglais | MEDLINE | ID: mdl-31227713

RÉSUMÉ

The contribution of mast cells in the microenvironment of solid malignancies remains controversial. Here we functionally assess the impact of tumor-adjacent, submucosal mast cell accumulation in murine and human intestinal-type gastric cancer. We find that genetic ablation or therapeutic inactivation of mast cells suppresses accumulation of tumor-associated macrophages, reduces tumor cell proliferation and angiogenesis, and diminishes tumor burden. Mast cells are activated by interleukin (IL)-33, an alarmin produced by the tumor epithelium in response to the inflammatory cytokine IL-11, which is required for the growth of gastric cancers in mice. Accordingly, ablation of the cognate IL-33 receptor St2 limits tumor growth, and reduces mast cell-dependent production and release of the macrophage-attracting factors Csf2, Ccl3, and Il6. Conversely, genetic or therapeutic macrophage depletion reduces tumor burden without affecting mast cell abundance. Therefore, tumor-derived IL-33 sustains a mast cell and macrophage-dependent signaling cascade that is amenable for the treatment of gastric cancer.


Sujet(s)
Interleukine-33/immunologie , Macrophages/immunologie , Mastocytes/immunologie , Tumeurs de l'estomac/immunologie , Aminopyridines/administration et posologie , Animaux , Dégranulation cellulaire/effets des médicaments et des substances chimiques , Dégranulation cellulaire/immunologie , Cromoglicate de sodium/administration et posologie , Modèles animaux de maladie humaine , Épithélium/immunologie , Épithélium/anatomopathologie , Femelle , Muqueuse gastrique/cytologie , Muqueuse gastrique/immunologie , Muqueuse gastrique/anatomopathologie , Humains , Protéine-1 analogue au récepteur de l'interleukin-1/immunologie , Protéine-1 analogue au récepteur de l'interleukin-1/métabolisme , Interleukine-33/métabolisme , Mâle , Souris , Souris transgéniques , Pyrroles/administration et posologie , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/immunologie , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/mortalité , Tumeurs de l'estomac/anatomopathologie , Analyse sur puce à tissus , Microenvironnement tumoral/immunologie
20.
EMBO Mol Med ; 11(4)2019 04.
Article de Anglais | MEDLINE | ID: mdl-30885958

RÉSUMÉ

Excessive signaling through gp130, the shared receptor for the interleukin (IL)6 family of cytokines, is a common hallmark in solid malignancies and promotes their progression. Here, we established the in vivo utility of bazedoxifene, a steroid analog clinically approved for the treatment of osteoporosis, to suppress gp130-dependent tumor growth of the gastrointestinal epithelium. Bazedoxifene administration reduced gastric tumor burden in gp130Y757F mice, where tumors arise exclusively through excessive gp130/STAT3 signaling in response to the IL6 family cytokine IL11. Likewise, in mouse models of sporadic colon and intestinal cancers, which arise from oncogenic mutations in the tumor suppressor gene Apc and the associated ß-catenin/canonical WNT pathway, bazedoxifene treatment reduces tumor burden. Consistent with the proposed orthogonal tumor-promoting activity of IL11-dependent gp130/STAT3 signaling, tumors of bazedoxifene-treated Apc-mutant mice retain excessive nuclear accumulation of ß-catenin and aberrant WNT pathway activation. Likewise, bazedoxifene treatment of human colon cancer cells harboring mutant APC did not reduce aberrant canonical WNT signaling, but suppressed IL11-dependent STAT3 signaling. Our findings provide compelling proof of concept to support the repurposing of bazedoxifene for the treatment of gastrointestinal cancers in which IL11 plays a tumor-promoting role.


Sujet(s)
Repositionnement des médicaments , Tumeurs gastro-intestinales/traitement médicamenteux , Indoles/usage thérapeutique , Modulateurs sélectifs des récepteurs des oestrogènes/usage thérapeutique , Protéine de la polypose adénomateuse colique/génétique , Animaux , Prolifération cellulaire/effets des médicaments et des substances chimiques , Récepteur gp130 de cytokines/composition chimique , Récepteur gp130 de cytokines/métabolisme , Modèles animaux de maladie humaine , Femelle , Tumeurs gastro-intestinales/anatomopathologie , Humains , Indoles/métabolisme , Indoles/pharmacologie , Interleukine-11/composition chimique , Interleukine-11/métabolisme , Interleukine-11/pharmacologie , Mâle , Souris , Souris de lignée C57BL , Souris transgéniques , Facteur de transcription STAT-3/métabolisme , Modulateurs sélectifs des récepteurs des oestrogènes/métabolisme , Modulateurs sélectifs des récepteurs des oestrogènes/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , bêta-Caténine/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...