Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 19 de 19
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
J Neurooncol ; 168(2): 317-332, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38630384

RÉSUMÉ

INTRODUCTION: Patients with pediatric low-grade gliomas (pLGGs), the most common primary brain tumors in children, can often benefit from MAPK inhibitor (MAPKi) treatment. However, rapid tumor regrowth, also referred to as rebound growth, may occur once treatment is stopped, constituting a significant clinical challenge. METHODS: Four patient-derived pediatric glioma models were investigated to model rebound growth in vitro based on viable cell counts in response to MAPKi treatment and withdrawal. A multi-omics dataset (RNA sequencing and LC-MS/MS based phospho-/proteomics) was generated to investigate possible rebound-driving mechanisms. Following in vitro validation, putative rebound-driving mechanisms were validated in vivo using the BT-40 orthotopic xenograft model. RESULTS: Of the tested models, only a BRAFV600E-driven model (BT-40, with additional CDKN2A/Bdel) showed rebound growth upon MAPKi withdrawal. Using this model, we identified a rapid reactivation of the MAPK pathway upon MAPKi withdrawal in vitro, also confirmed in vivo. Furthermore, transient overactivation of key MAPK molecules at transcriptional (e.g. FOS) and phosphorylation (e.g. pMEK) levels, was observed in vitro. Additionally, we detected increased expression and secretion of cytokines (CCL2, CX3CL1, CXCL10 and CCL7) upon MAPKi treatment, maintained during early withdrawal. While increased cytokine expression did not have tumor cell intrinsic effects, presence of these cytokines in conditioned media led to increased attraction of microglia cells in vitro. CONCLUSION: Taken together, these data indicate rapid MAPK reactivation upon MAPKi withdrawal as a tumor cell intrinsic rebound-driving mechanism. Furthermore, increased secretion of microglia-recruiting cytokines may play a role in treatment response and rebound growth upon withdrawal, warranting further evaluation.


Sujet(s)
Tumeurs du cerveau , Cytokines , Gliome , Microglie , Mutation , Inhibiteurs de protéines kinases , Protéines proto-oncogènes B-raf , Humains , Protéines proto-oncogènes B-raf/génétique , Protéines proto-oncogènes B-raf/métabolisme , Microglie/métabolisme , Microglie/effets des médicaments et des substances chimiques , Gliome/métabolisme , Gliome/traitement médicamenteux , Gliome/anatomopathologie , Gliome/génétique , Cytokines/métabolisme , Animaux , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/génétique , Inhibiteurs de protéines kinases/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe , Enfant , Souris , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques
2.
Oral Dis ; 2024 Feb 20.
Article de Anglais | MEDLINE | ID: mdl-38376129

RÉSUMÉ

OBJECTIVES: Investigation of the therapeutic effect of zoledronic acid (ZA) in a preclinical model of jaw osteosarcoma (JO). MATERIALS AND METHODS: The effect of 100 µg/kg ZA administered twice a week was assessed in a xenogenic mouse model of JO. The clinical (tumor growth, development of lung metastasis), radiological (bone microarchitecture by micro-CT analysis), and molecular and immunohistochemical (TRAP, RANK/RANKL, VEGF, and CD146) parameters were investigated. RESULTS: Animals receiving ZA exhibited an increased tumor volume compared with nontreated animals (71.3 ± 14.3 mm3 vs. 51.9 ± 19.9 mm3 at D14, respectively; p = 0.06) as well as increased numbers of lung metastases (mean 4.88 ± 4.45 vs. 0.50 ± 1.07 metastases, respectively; p = 0.02). ZA protected mandibular bone against tumor osteolysis (mean bone volume of 12.81 ± 0.53 mm3 in the ZA group vs. 11.55 ± 1.18 mm3 in the control group; p = 0.01). ZA induced a nonsignificant decrease in mRNA expression of the osteoclastic marker TRAP and an increase in RANK/RANKL bone remodeling markers. CONCLUSION: The use of bisphosphonates in the therapeutic strategy for JO should be further explored, as should the role of bone resorption in the pathophysiology of the disease.

3.
Nat Cell Biol ; 25(12): 1804-1820, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-38012402

RÉSUMÉ

Drugs that selectively kill senescent cells (senolytics) improve the outcomes of cancer, fibrosis and age-related diseases. Despite their potential, our knowledge of the molecular pathways that affect the survival of senescent cells is limited. To discover senolytic targets, we performed RNAi screens and identified coatomer complex I (COPI) vesicle formation as a liability of senescent cells. Genetic or pharmacological inhibition of COPI results in Golgi dispersal, dysfunctional autophagy, and unfolded protein response-dependent apoptosis of senescent cells, and knockdown of COPI subunits improves the outcomes of cancer and fibrosis in mouse models. Drugs targeting COPI have poor pharmacological properties, but we find that N-myristoyltransferase inhibitors (NMTi) phenocopy COPI inhibition and are potent senolytics. NMTi selectively eliminated senescent cells and improved outcomes in models of cancer and non-alcoholic steatohepatitis. Our results suggest that senescent cells rely on a hyperactive secretory apparatus and that inhibiting trafficking kills senescent cells with the potential to treat various senescence-associated diseases.


Sujet(s)
Tumeurs , Sénothérapie , Souris , Animaux , Appareil de Golgi/métabolisme , Vieillissement de la cellule , Tumeurs/métabolisme , Fibrose
4.
Aging (Albany NY) ; 15(17): 8576-8593, 2023 09 01.
Article de Anglais | MEDLINE | ID: mdl-37659108

RÉSUMÉ

Senescent cells (SnCs) have been described to accumulate in osteoarthritis (OA) joint tissues in response to injury, thereby participating in OA development and progression. However, clinical therapeutic approaches targeting SnCs using senolysis, although promising in preclinical OA models, have not yet proven their efficacy in patients with knee OA. This pitfall may be due to the lack of understanding of the mechanisms underlying chondrocyte senescence. Therefore, our study aimed to generate models of chondrocyte senescence. This study used etoposide, to induce DNA damage-related senescence or chronic exposure to IL-1ß to entail inflammation-related senescence in human OA chondrocytes. Several hallmarks of cellular senescence, such as cell cycle arrest, expression of cyclin-dependent kinase inhibitors, DNA damages, and senescence-associated secretory profile were evaluated. Chronic exposure to IL-1ß induces only partial expression of senescence markers and does not allow us to conclude on its ability to induce senescence in chondrocytes. On the other hand, etoposide treatment reliably induces DNA damage-related senescence in human articular chondrocytes evidenced by loss of proliferative capacity, DNA damage accumulation, and expression of some SASP components. Etoposide-induced senescence model may help investigate the initiation of cellular senescence in chondrocytes, and provide a useful model to develop therapeutic approaches to target senescence in OA.


Sujet(s)
Chondrocytes , Gonarthrose , Humains , Étoposide/pharmacologie , Gonarthrose/génétique , Transport biologique , Altération de l'ADN
5.
Cancer Cell ; 41(7): 1242-1260.e6, 2023 07 10.
Article de Anglais | MEDLINE | ID: mdl-37267953

RÉSUMÉ

The accumulation of senescent cells in the tumor microenvironment can drive tumorigenesis in a paracrine manner through the senescence-associated secretory phenotype (SASP). Using a new p16-FDR mouse line, we show that macrophages and endothelial cells are the predominant senescent cell types in murine KRAS-driven lung tumors. Through single cell transcriptomics, we identify a population of tumor-associated macrophages that express a unique array of pro-tumorigenic SASP factors and surface proteins and are also present in normal aged lungs. Genetic or senolytic ablation of senescent cells, or macrophage depletion, result in a significant decrease in tumor burden and increased survival in KRAS-driven lung cancer models. Moreover, we reveal the presence of macrophages with senescent features in human lung pre-malignant lesions, but not in adenocarcinomas. Taken together, our results have uncovered the important role of senescent macrophages in the initiation and progression of lung cancer, highlighting potential therapeutic avenues and cancer preventative strategies.


Sujet(s)
Vieillissement de la cellule , Tumeurs du poumon , Sujet âgé , Animaux , Humains , Souris , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Vieillissement de la cellule/génétique , Cellules endothéliales , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Macrophages/métabolisme , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes p21(ras)/métabolisme , Microenvironnement tumoral
6.
Neuro Oncol ; 25(4): 735-747, 2023 04 06.
Article de Anglais | MEDLINE | ID: mdl-35977048

RÉSUMÉ

BACKGROUND: Pilocytic astrocytoma (PA) is the most common pediatric brain tumor and a mitogen-activated protein kinase (MAPK)-driven disease. Oncogenic MAPK-signaling drives the majority of cells into oncogene-induced senescence (OIS). While OIS induces resistance to antiproliferative therapies, it represents a potential vulnerability exploitable by senolytic agents. METHODS: We established new patient-derived PA cell lines that preserve molecular features of the primary tumors and can be studied in OIS and proliferation depending on expression or repression of the SV40 large T antigen. We determined expression of anti-apoptotic BCL-2 members in these models and primary PA. Dependence of senescent PA cells on anti-apoptotic BCL-2 members was investigated using a comprehensive set of BH3 mimetics. RESULTS: Senescent PA cells upregulate BCL-XL upon senescence induction and show dependency on BCL-XL for survival. BH3 mimetics with high affinity for BCL-XL (BCL-XLi) reduce metabolic activity and induce mitochondrial apoptosis in senescent PA cells at nano-molar concentrations. In contrast, BH3 mimetics without BCL-XLi activity, conventional chemotherapy, and MEK inhibitors show no effect. CONCLUSIONS: Our data demonstrate that BCL-XL is critical for survival of senescent PA tumor cells and provides proof-of-principle for the use of clinically available BCL-XL-dependent senolytics.


Sujet(s)
Astrocytome , Tumeurs du cerveau , Enfant , Humains , Protéines proto-oncogènes c-bcl-2 , Apoptose , Astrocytome/anatomopathologie , Tumeurs du cerveau/anatomopathologie , Mitogen-Activated Protein Kinases , Lignée cellulaire tumorale
7.
Cancers (Basel) ; 14(22)2022 Nov 16.
Article de Anglais | MEDLINE | ID: mdl-36428719

RÉSUMÉ

Despite advances in clinical management, osteosarcoma and Ewing sarcoma, the two most frequent malignant primary bone tumors at pediatric age, still have a poor prognosis for high-risk patients (i.e., relapsed or metastatic disease). Triggering a TRAIL pro-apoptotic pathway represents a promising therapeutic approach, but previous studies have described resistance mechanisms that could explain the declining interest of such an approach in clinical trials. In this study, eight relevant human cell lines were used to represent the heterogeneity of the response to the TRAIL pro-apoptotic effect in pediatric bone tumors and two cell-derived xenograft models were developed, originating from a sensitive and a resistant cell line. The DR5 agonist antibody AMG655 (Conatumumab) was selected as an example of TRAIL-based therapy. In both TRAIL-sensitive and TRAIL-resistant cell lines, two signaling pathways were activated following AMG655 treatment, the canonical extrinsic apoptotic pathway and a non-apoptotic pathway, involving the recruitment of RIPK1 on the DR5 protein complex, activating both pro-survival and pro-proliferative effectors. However, the resulting balance of these two pathways was different, leading to apoptosis only in sensitive cells. In vivo, AMG655 treatment reduced tumor development of the sensitive model but accelerated tumor growth of the resistant one. We proposed two independent strategies to overcome this issue: (1) a proof-of-concept targeting of RIPK1 by shRNA approach and (2) the use of a novel highly-potent TRAIL-receptor agonist; both shifting the balance in favor of apoptosis. These observations are paving the way to resurrect TRAIL-based therapies in pediatric bone tumors to help predict the response to treatment, and propose a relevant adjuvant strategy for future therapeutic development.

8.
Neuropathol Appl Neurobiol ; 47(3): 359-378, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-33378554

RÉSUMÉ

The study of cell senescence is a burgeoning field. Senescent cells can modify the cellular microenvironment through the secretion of a plethora of biologically active products referred to as the senescence-associated secretory phenotype (SASP). The consequences of these paracrine signals can be either beneficial for tissue homeostasis, if senescent cells are properly cleared and SASP activation is transient, or result in organ dysfunction, when senescent cells accumulate within the tissues and SASP activation is persistent. Several studies have provided evidence for the role of senescence and SASP in promoting age-related diseases or driving organismal ageing. The hype about senescence has been further amplified by the fact that a group of drugs, named senolytics, have been used to successfully ameliorate the burden of age-related diseases and increase health and life span in mice. Ablation of senescent cells in the brain prevents disease progression and improves cognition in murine models of neurodegenerative conditions. The role of senescence in cancer has been more thoroughly investigated, and it is now accepted that senescence is a double-edged sword that can paradoxically prevent or promote tumourigenesis in a context-dependent manner. In addition, senescence induction followed by senolytic treatment is starting to emerge as a novel therapeutic avenue that could improve current anti-cancer therapies and reduce tumour recurrence. In this review, we discuss recent findings supporting the role of cell senescence in the pathogenesis of neurodegenerative diseases and in brain tumours. A better understanding of senescence is likely to result in the development of novel and efficacious anti-senescence therapies against these brain pathologies.


Sujet(s)
Tumeurs du cerveau/anatomopathologie , Vieillissement de la cellule/physiologie , Dégénérescence nerveuse/anatomopathologie , Animaux , Carcinogenèse/anatomopathologie , Humains
9.
Aging Cell ; 19(4): e13133, 2020 04.
Article de Anglais | MEDLINE | ID: mdl-32175667

RÉSUMÉ

Senescence is a stable growth arrest that impairs the replication of damaged, old or preneoplastic cells, therefore contributing to tissue homeostasis. Senescent cells accumulate during ageing and are associated with cancer, fibrosis and many age-related pathologies. Recent evidence suggests that the selective elimination of senescent cells can be effective on the treatment of many of these senescence-associated diseases. A universal characteristic of senescent cells is that they display elevated activity of the lysosomal ß-galactosidase, and this has been exploited as a marker for senescence (senescence-associated ß-galactosidase activity). Consequently, we hypothesized that galactose-modified cytotoxic prodrugs will be preferentially processed by senescent cells, resulting in their selective killing. Here, we show that different galactose-modified duocarmycin (GMD) derivatives preferentially kill senescent cells. GMD prodrugs induce selective apoptosis of senescent cells in a lysosomal ß-galactosidase (GLB1)-dependent manner. GMD prodrugs can eliminate a broad range of senescent cells in culture, and treatment with a GMD prodrug enhances the elimination of bystander senescent cells that accumulate upon whole-body irradiation treatment of mice. Moreover, taking advantage of a mouse model of adamantinomatous craniopharyngioma (ACP), we show that treatment with a GMD prodrug selectively reduced the number of ß-catenin-positive preneoplastic senescent cells. In summary, the above results make a case for testing the potential of galactose-modified duocarmycin prodrugs to treat senescence-related pathologies.


Sujet(s)
Antibiotiques antinéoplasiques/pharmacologie , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Craniopharyngiome/traitement médicamenteux , Duocarmycines/pharmacologie , Galactose/pharmacologie , Promédicaments/pharmacologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire , Techniques de coculture , Craniopharyngiome/métabolisme , Craniopharyngiome/anatomopathologie , Humains , Souris , Souris de lignée C57BL , Tumeurs expérimentales/traitement médicamenteux , Tumeurs expérimentales/métabolisme , Tumeurs expérimentales/anatomopathologie , beta-Galactosidase/métabolisme
10.
Nat Metab ; 1(11): 1074-1088, 2019 11.
Article de Anglais | MEDLINE | ID: mdl-31799499

RÉSUMÉ

Senescence is a cellular stress response that results in the stable arrest of old, damaged or preneoplastic cells. Oncogene-induced senescence is tumor suppressive but can also exacerbate tumorigenesis through the secretion of pro-inflammatory factors from senescent cells. Drugs that selectively kill senescent cells, termed senolytics, have proved beneficial in animal models of many age-associated diseases. Here, we show that the cardiac glycoside, ouabain, is a senolytic agent with broad activity. Senescent cells are sensitized to ouabain-induced apoptosis, a process mediated in part by induction of the pro-apoptotic Bcl2-family protein NOXA. We show that cardiac glycosides synergize with anti-cancer drugs to kill tumor cells and eliminate senescent cells that accumulate after irradiation or in old mice. Ouabain also eliminates senescent preneoplastic cells. Our findings suggest that cardiac glycosides may be effective anti-cancer drugs by acting through multiple mechanism. Given the broad range of senescent cells targeted by cardiac glycosides their use against age-related diseases warrants further exploration.


Sujet(s)
Glucosides cardiotoniques/pharmacologie , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Animaux , Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Humains , Souris , Ouabaïne/pharmacologie , Quercétine/pharmacologie , Rats
11.
Clin Cancer Res ; 25(6): 1851-1866, 2019 03 15.
Article de Anglais | MEDLINE | ID: mdl-30530705

RÉSUMÉ

PURPOSE: Pilocytic astrocytoma is the most common childhood brain tumor, characterized by constitutive MAPK activation. MAPK signaling induces oncogene-induced senescence (OIS), which may cause unpredictable growth behavior of pilocytic astrocytomas. The senescence-associated secretory phenotype (SASP) has been shown to regulate OIS, but its role in pilocytic astrocytoma remains unknown.Experimental Design: The patient-derived pilocytic astrocytoma cell culture model, DKFZ-BT66, was used to demonstrate presence of the SASP and analyze its impact on OIS in pilocytic astrocytoma. The model allows for doxycycline-inducible switching between proliferation and OIS. Both states were studied using gene expression profiling (GEP), Western blot, ELISA, and cell viability testing. Primary pilocytic astrocytoma tumors were analyzed by GEP and multiplex assay. RESULTS: SASP factors were upregulated in primary human and murine pilocytic astrocytoma and during OIS in DKFZ-BT66 cells. Conditioned medium induced growth arrest of proliferating pilocytic astrocytoma cells. The SASP factors IL1B and IL6 were upregulated in primary pilocytic astrocytoma, and both pathways were regulated during OIS in DKFZ-BT66. Stimulation with rIL1B but not rIL6 reduced growth of DKFZ-BT66 cells and induced the SASP. Anti-inflammatory treatment with dexamethasone induced regrowth of senescent cells and inhibited the SASP. Senescent DKFZ-BT66 cells responded to senolytic BCL2 inhibitors. High IL1B and SASP expression in pilocytic astrocytoma tumors was associated with favorable progression-free survival. CONCLUSIONS: We provide evidence for the SASP regulating OIS in pediatric pilocytic astrocytoma, with IL1B as a relevant mediator. SASP expression could enable prediction of progression in patients with pilocytic astrocytoma. Further investigation of the SASP driving the unpredictable growth of pilocytic astrocytomas, and its possible therapeutic application, is warranted.


Sujet(s)
Astrocytome/anatomopathologie , Tumeurs du cerveau/anatomopathologie , Vieillissement de la cellule , Interleukine-1 bêta/métabolisme , Animaux , Astrocytome/mortalité , Astrocytome/chirurgie , Tumeurs du cerveau/mortalité , Tumeurs du cerveau/chirurgie , Prolifération cellulaire , Enfant , Milieux de culture conditionnés/métabolisme , Jeux de données comme sujet , Modèles animaux de maladie humaine , Femelle , Analyse de profil d'expression de gènes , Humains , Mâle , Souris , Culture de cellules primaires , Pronostic , Survie sans progression , Cellules cancéreuses en culture
12.
Cytotherapy ; 20(8): 1037-1045, 2018 08.
Article de Anglais | MEDLINE | ID: mdl-30093324

RÉSUMÉ

BACKGROUND: Osteosarcoma (OS) is the most frequent pediatric malignant bone tumor. OS patients have not seen any major therapeutic progress in the last 30 years, in particular in the case of metastatic disease, which requires new therapeutic strategies. The pro-apoptotic cytokine Tumor necrosis factor (TNF)-Related Apoptosis Inducing Ligand (TRAIL) can selectively kill tumor cells while sparing normal cells, making it a promising therapeutic tool in several types of cancer. However, many OS cell lines appear resistant to recombinant human (rh)TRAIL-induced apoptosis. We, therefore, hypothesized that TRAIL presentation at the membrane level of carrier cells might overcome this resistance and trigger apoptosis. METHODS: To address this, human adipose mesenchymal stromal cells (MSCs) transfected in a stable manner to express membrane-bound full-length human TRAIL (mbTRAIL) were co-cultured with several human OS cell lines. RESULTS: This induced apoptosis by cell-to-cell contact even in cell lines initially resistant to rhTRAIL. In contrast, mbTRAIL delivered by MSCs was not able to counteract tumor progression in this OS orthotopic model. DISCUSSION: This was partly due to the fact that MSCs showed a potential to support tumor development. Moreover, the expression of mbTRAIL did not show caspase activation in adjacent tumor cells.


Sujet(s)
Apoptose , Tumeurs osseuses/thérapie , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses/métabolisme , Ostéosarcome/thérapie , Ligand TRAIL/métabolisme , Ligand TRAIL/pharmacologie , Transfert adoptif/méthodes , Animaux , Apoptose/effets des médicaments et des substances chimiques , Apoptose/génétique , Tumeurs osseuses/génétique , Tumeurs osseuses/anatomopathologie , Cellules cultivées , Thérapie génétique/méthodes , Humains , Techniques in vitro , Transplantation de cellules souches mésenchymateuses/méthodes , Cellules souches mésenchymateuses/cytologie , Souris , Souris nude , Ostéosarcome/génétique , Ostéosarcome/anatomopathologie , Ligand TRAIL/génétique , Transfection , Tests d'activité antitumorale sur modèle de xénogreffe
13.
Acta Neuropathol ; 135(5): 757-777, 2018 05.
Article de Anglais | MEDLINE | ID: mdl-29541918

RÉSUMÉ

Adamantinomatous craniopharyngiomas (ACPs) are clinically challenging tumours, the majority of which have activating mutations in CTNNB1. They are histologically complex, showing cystic and solid components, the latter comprised of different morphological cell types (e.g. ß-catenin-accumulating cluster cells and palisading epithelium), surrounded by a florid glial reaction with immune cells. Here, we have carried out RNA sequencing on 18 ACP samples and integrated these data with an existing ACP transcriptomic dataset. No studies so far have examined the patterns of gene expression within the different cellular compartments of the tumour. To achieve this goal, we have combined laser capture microdissection with computational analyses to reveal groups of genes that are associated with either epithelial tumour cells (clusters and palisading epithelium), glial tissue or immune infiltrate. We use these human ACP molecular signatures and RNA-Seq data from two ACP mouse models to reveal that cell clusters are molecularly analogous to the enamel knot, a critical signalling centre controlling normal tooth morphogenesis. Supporting this finding, we show that human cluster cells express high levels of several members of the FGF, TGFB and BMP families of secreted factors, which signal to neighbouring cells as evidenced by immunostaining against the phosphorylated proteins pERK1/2, pSMAD3 and pSMAD1/5/9 in both human and mouse ACP. We reveal that inhibiting the MAPK/ERK pathway with trametinib, a clinically approved MEK inhibitor, results in reduced proliferation and increased apoptosis in explant cultures of human and mouse ACP. Finally, we analyse a prominent molecular signature in the glial reactive tissue to characterise the inflammatory microenvironment and uncover the activation of inflammasomes in human ACP. We validate these results by immunostaining against immune cell markers, cytokine ELISA and proteome analysis in both solid tumour and cystic fluid from ACP patients. Our data support a new molecular paradigm for understanding ACP tumorigenesis as an aberrant mimic of natural tooth development and opens new therapeutic opportunities by revealing the activation of the MAPK/ERK and inflammasome pathways in human ACP.


Sujet(s)
Craniopharyngiome/métabolisme , Système de signalisation des MAP kinases , Tumeurs de l'hypophyse/métabolisme , Transcriptome , Microenvironnement tumoral/physiologie , Animaux , Biologie informatique , Craniopharyngiome/anatomopathologie , Craniopharyngiome/thérapie , Cytokines/métabolisme , Modèles animaux de maladie humaine , Humains , Inflammation/métabolisme , Inflammation/thérapie , Microdissection au laser , Souris , Névroglie/métabolisme , Odontogenèse/physiologie , Hypophyse/embryologie , Hypophyse/anatomopathologie , Tumeurs de l'hypophyse/anatomopathologie , Tumeurs de l'hypophyse/thérapie , Analyse de séquence d'ARN , Techniques de culture de tissus
14.
Am J Cancer Res ; 7(11): 2333-2349, 2017.
Article de Anglais | MEDLINE | ID: mdl-29218254

RÉSUMÉ

Osteosarcoma, the most common malignant primary bone tumor, is currently treated with chemotherapy and surgery. The effectiveness of chemotherapy is evaluated by means of histological analysis of tumor necrosis, known as "the Huvos score". However, 25% of the patients initially considered good responders will relapse. In our practice, strong tissue heterogeneity around the residual viable cells of the osteosarcoma is observed, but this is not taken into account by the Huvos score, as it is only an average. The objective is to determine whether heterogeneity in the osteosarcoma's microenvironment can play a role in the histological response to chemotherapy. Two complementary approaches have been developed: (i) the therapeutic response to several monotherapies (ifosfamide, cisplatin, doxorubicin) has been compared to tumor growth and the necrosis levels in different preclinical syngeneic osteosarcoma models, mimicking various microenvironments by injecting the tumor cells into subcutaneous, intra-muscular paratibial, or intra-osseous sites; (ii) a retrospective analysis was performed on patients' osteoblastic osteosarcoma biopsies. Tissue localization mapping of residual live tumor cell colonies was evaluated for potential correlation with overall survival. The results of the preclinical studies showed a difference in tumor growth depending on the osteosarcoma model, with a higher rate in bone sites compared to subcutaneous tumors. For the therapeutic response, a higher response to doxorubicin was observed in the intra-osseous model compared to the intra-muscular model for tumor growth (P = 0.013) and necrosis (P = 0.007). These data strongly suggest that the microenvironment plays a role in how osteosarcoma responds to chemotherapy. The retrospective analysis showed no significant survival difference between residual cell sites, although the soft tissues may be seen as a potential negative factor.

15.
Int J Cancer ; 139(12): 2802-2811, 2016 Dec 15.
Article de Anglais | MEDLINE | ID: mdl-27558972

RÉSUMÉ

Ewing sarcoma (EWS) is the second most frequent pediatric malignant bone tumor. EWS patients have not seen any major therapeutic progress in the last 30 years, in particular in the case of metastatic disease, which requires new therapeutic strategies. The pro-apoptotic cytokine TNF-Related Apoptosis Inducing Ligand (TRAIL) can selectively kill tumor cells while sparing normal cells, making it a promising therapeutic tool in several types of cancer. However, certain EWS cell lines appear resistant to recombinant human (rh) TRAIL-induced apoptosis. We therefore hypothesized that a TRAIL presentation at the surface of the carrier cells might overcome this resistance and trigger apoptosis. For this purpose, human adipose mesenchymal stromal/stem cells (MSC) transfected in a stable manner to express full-length human TRAIL were co-cultured with several human EWS cell lines, inducing apoptosis by cell-to-cell contact even in cell lines initially resistant to rhTRAIL or AMG655, an antibody agonist to the death receptor, DR5. In vivo, TRAIL delivered by MSCs was able to counteract tumor progression in two orthotopic models of Ewing sarcoma, associated with caspase activation, indicating that a cell-based delivery of a potent apoptosis-inducing factor could be relevant in EWS.


Sujet(s)
Transformation cellulaire néoplasique/génétique , Cellules souches mésenchymateuses/métabolisme , Sarcome d'Ewing/étiologie , Sarcome d'Ewing/métabolisme , Ligand TRAIL/génétique , Animaux , Apoptose/génétique , Caspases/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire , Transformation cellulaire néoplasique/métabolisme , Modèles animaux de maladie humaine , Expression des gènes , Gènes rapporteurs , Hétérogreffes , Humains , Souris , Sarcome d'Ewing/mortalité , Sarcome d'Ewing/anatomopathologie , Ligand TRAIL/métabolisme , Transduction génétique
16.
Am J Cancer Res ; 6(3): 677-89, 2016.
Article de Anglais | MEDLINE | ID: mdl-27152244

RÉSUMÉ

Osteosarcoma, the most frequent malignant primary bone tumor in pediatric patients is characterized by osteolysis promoting tumor growth. Lung metastasis is the major bad prognosis factor of this disease. Zoledronic Acid (ZA), a potent inhibitor of bone resorption is currently evaluated in phase III randomized studies in Europe for the treatment of osteosarcoma and Ewing sarcoma. The beneficial effect of the liposomal form of Muramyl-TriPeptide-Phosphatidyl Ethanolamine (L-mifamurtide, MEPACT®), an activator of macrophage populations has been demonstrated to eradicate lung metastatic foci in osteosarcoma. The objective of this study was to evaluate the potential therapeutic benefit and the safety of the ZA and L-mifamurtide combination in preclinical models of osteosarcoma, as a prerequisite before translation to patients. The effects of ZA (100 µg/kg) and L-mifamurtide (1 mg/kg) were investigated in vivo in xenogeneic and syngeneic mice models of osteosarcoma, at clinical (tumor proliferation, spontaneous lung metastases development), radiological (bone microarchitecture by microCT analysis), biological and histological levels. No interference between the two drugs could be observed on ZA-induced bone protection and on L-mifamurtide-induced inhibition of lung metastasis development. Unexpectedly, ZA and L-mifamurtide association induced an additional and in some cases synergistic inhibition of primary tumor progression. L-mifamurtide has no effect on tumor proliferation in vitro or in vivo, and macrophage population was not affected at the tumor site whatever the treatment. This study evidenced for the first time a significant inhibition of primary osteosarcoma progression when both drugs are combined. This result constitutes a first proof-of-principle for clinical application in osteosarcoma patients.

17.
Future Oncol ; 11(3): 535-42, 2015.
Article de Anglais | MEDLINE | ID: mdl-25675131

RÉSUMÉ

Osteosarcoma and Ewing's sarcoma, the two most frequent malignant primary tumors preferentially arise in children and young adults, and have a poor prognosis. TRAIL represents a promising therapeutic approach for most cancers but in the case of primary bone tumors, osteosarcoma cell lines are highly resistant to this pro-apoptotic cytokine. In addition, another signaling pathway mediating cell proliferation and migration may be even activated in this subset of resistant cells leading to protumoral effect. Therapeutic perspectives are linked to possibility to overcome TRAIL resistance by combining other drugs with TRAIL or death receptors agonistic antibodies. We hypothesized that the bone microenvironment may provide a favorable niche for TRAIL resistance that might be targeted by new resensitizing agents.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Tumeurs osseuses/traitement médicamenteux , Tumeurs osseuses/métabolisme , Thérapie moléculaire ciblée , Transduction du signal/effets des médicaments et des substances chimiques , Ligand TRAIL/métabolisme , Facteurs âges , Antinéoplasiques/pharmacologie , Enfant , Enfant d'âge préscolaire , Résistance aux médicaments antinéoplasiques , Humains , Ostéosarcome/traitement médicamenteux , Ostéosarcome/métabolisme , Microenvironnement tumoral/effets des médicaments et des substances chimiques
18.
Oncotarget ; 5(17): 7805-19, 2014 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-25138053

RÉSUMÉ

PURPOSE: Despite recent improvements in therapeutic management of osteosarcoma, ongoing challenges in improving the response to chemotherapy warrants new strategies still needed to improve overall patient survival. Among new therapeutic approaches, zoledronic acid (ZOL) represents a promising adjuvant molecule to chemotherapy to limit the osteolytic component of bone tumors. However, ZOL triggers the elevation of heat shock proteins (Hsp), including Hsp27 and clusterin (CLU), which could enhance tumor cell survival and treatment resistance. We hypothesized that targeting CLU using siRNA or the antisense drug, OGX-011, will suppress treatment-induced CLU induction and enhance ZOL-induced cell death in osteosarcoma (OS) cells. METHODS: The combined effects of OGX-011 and ZOL were investigated in vitro on cell growth, viability, apoptosis and cell cycle repartition of ZOL-sensitive or -resistant human OS cell lines (SaOS2, U2OS, MG63 and MNNG/HOS). RESULTS: In OS cell lines, ZOL increased levels of HSPs, especially CLU, in a dose- and time-dependent manner by mechanism including increased HSF1 transcription activity. The OS resistant cells to ZOL exhibited higher CLU expression level than the sensitive cells. Moreover, CLU overexpression protects OS sensitive cells to ZOL-induced cell death by modulating the MDR1 and farnesyl diphosphate synthase expression. OGX-011 suppressed treatment-induced increases in CLU and synergistically enhanced the activity of ZOL on cell growth and apoptosis. These biologic events were accompanied by decreased expression of HSPs, MDR1 and HSF1 transcriptional activity. In vivo, OGX-011, administered 3 times a week (IP, 20mg/kg), potentiated the effect of ZOL (s.c; 50µg/kg), significantly inhibiting tumor growth by 50% and prolonging survival in MNNG/HOS xenograft model compared to ZOL alone. CONCLUSION: These results indicate that ZOL-mediated induction of CLU can be attenuated by OGX-011, with synergistic effects on delaying progression of osteosarcoma.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Tumeurs osseuses/anatomopathologie , Clusterine/biosynthèse , Diphosphonates/administration et posologie , Imidazoles/administration et posologie , Ostéosarcome/anatomopathologie , Thionucléotides/administration et posologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Technique de Western , Tumeurs osseuses/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Synergie des médicaments , Femelle , Humains , Immunohistochimie , Souris , Souris nude , Ostéosarcome/métabolisme , RT-PCR , Tests d'activité antitumorale sur modèle de xénogreffe , Acide zolédronique
19.
Mol Cancer Res ; 10(3): 336-46, 2012 Mar.
Article de Anglais | MEDLINE | ID: mdl-22258765

RÉSUMÉ

Ewing's sarcoma (ES) is a high-grade neoplasm arising in bones of children and adolescents. Survival rate decreases from greater than 50% to only 20% after 5 years for patients not responding to treatment or presenting metastases at diagnosis. TRAIL, which has strong antitumoral activity, is a promising therapeutic candidate. To address TRAIL sensitivity, 7 human ES cell lines were used. Cell viability experiments [3'[1-(phenylaminocarbonyl)-3,4-tetrazolium]-bis(4-methoxy-6-nitro-)benzene sulfonic acid hydrate (XTT) assay] showed that 4 of the 7 ES cell lines were resistant to TRAIL. Western blotting and flow cytometry analyses revealed that DR5 was uniformly expressed by all ES cell lines, whereas DR4 levels were higher in sensitive cell lines. In TRAIL-sensitive TC-71 cells, knockdown of TNFRSF10A/DR4 by short hairpin RNA (shRNA) was associated with a loss of sensitivity to TRAIL, in spite of DR5 presence. Interestingly, we identified a new transcript variant that results from an alternative splicing and encodes a 310-amino acid protein which corresponds to the 468 aa of DR4 original isoform but truncated of aa 11 to 168 within the extracellular TRAIL-binding domain. According to modeling studies, the contact of this new DR4 isoform (bDR4) with TRAIL seemed largely preserved. The overexpression of bDR4 in a TRAIL-resistant cell line restored TRAIL sensitivity. TRAIL resensitization was also observed after c-FLIP knockdown by shRNA in two TRAIL-resistant cell lines, as shown by XTT assay and caspase-3 assay. The results presented in this study showed that DR4, both as the complete form or as its new short isoform, is involved in TRAIL sensitivity in ES.


Sujet(s)
Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Récepteurs de TRAIL/métabolisme , Sarcome d'Ewing/métabolisme , Sarcome d'Ewing/anatomopathologie , Ligand TRAIL/pharmacologie , Épissage alternatif/effets des médicaments et des substances chimiques , Épissage alternatif/génétique , Séquence d'acides aminés , Protéine de régulation de l'apoptose CASP8 et FADD-like/métabolisme , Lignée cellulaire tumorale , Tests de criblage d'agents antitumoraux , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Techniques de knock-down de gènes , Humains , Modèles moléculaires , Données de séquences moléculaires , Protéines mutantes/composition chimique , Protéines mutantes/métabolisme , Isoformes de protéines/composition chimique , Isoformes de protéines/métabolisme , Structure tertiaire des protéines , ARN messager/génétique , ARN messager/métabolisme , Récepteurs de TRAIL/composition chimique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...