Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 8 de 8
Filtrer
1.
Sci Rep ; 11(1): 14062, 2021 07 07.
Article de Anglais | MEDLINE | ID: mdl-34234256

RÉSUMÉ

Medulloblastoma is the most common high-grade brain tumor in childhood. Medulloblastomas with c-myc amplification, classified as group 3, are the most aggressive among the four disease subtypes resulting in a 5-year overall survival of just above 50%. Despite current intensive therapy regimens, patients suffering from group 3 medulloblastoma urgently require new therapeutic options. Using a recently established c-myc amplified human medulloblastoma cell line, we performed an in-vitro-drug screen with single and combinatorial drugs that are either already clinically approved or agents in the advanced stage of clinical development. Candidate drugs were identified in vitro and then evaluated in vivo. Tumor growth was closely monitored by BLI. Vessel development was assessed by 3D light-sheet-fluorescence-microscopy. We identified the combination of gemcitabine and axitinib to be highly cytotoxic, requiring only low picomolar concentrations when used in combination. In the orthotopic model, gemcitabine and axitinib showed efficacy in terms of tumor control and survival. In both models, gemcitabine and axitinib were better tolerated than the standard regimen comprising of cisplatin and etoposide phosphate. 3D light-sheet-fluorescence-microscopy of intact tumors revealed thinning and rarefication of tumor vessels, providing one explanation for reduced tumor growth. Thus, the combination of the two drugs gemcitabine and axitinib has favorable effects on preventing tumor progression in an orthotopic group 3 medulloblastoma xenograft model while exhibiting a favorable toxicity profile. The combination merits further exploration as a new approach to treat high-risk group 3 medulloblastoma.


Sujet(s)
Antinéoplasiques/pharmacologie , Axitinib/pharmacologie , Désoxycytidine/analogues et dérivés , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Amplification de gène , Médulloblastome/génétique , Protéines proto-oncogènes c-myc/génétique , Animaux , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/génétique , Désoxycytidine/pharmacologie , Modèles animaux de maladie humaine , Expression des gènes , Humains , Médulloblastome/traitement médicamenteux , Médulloblastome/anatomopathologie , Souris , Néovascularisation pathologique/traitement médicamenteux , Néovascularisation pathologique/génétique , Néovascularisation pathologique/métabolisme , Récepteurs aux facteurs de croissance endothéliale vasculaire/génétique , Récepteurs aux facteurs de croissance endothéliale vasculaire/métabolisme , Résultat thérapeutique , Facteur de croissance endothéliale vasculaire de type A/génétique , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe ,
2.
J Exp Med ; 213(9): 1881-900, 2016 08 22.
Article de Anglais | MEDLINE | ID: mdl-27526711

RÉSUMÉ

Donor CD4(+)Foxp3(+) regulatory T cells (T reg cells) suppress graft-versus-host disease (GvHD) after allogeneic hematopoietic stem cell transplantation (HCT [allo-HCT]). Current clinical study protocols rely on the ex vivo expansion of donor T reg cells and their infusion in high numbers. In this study, we present a novel strategy for inhibiting GvHD that is based on the in vivo expansion of recipient T reg cells before allo-HCT, exploiting the crucial role of tumor necrosis factor receptor 2 (TNFR2) in T reg cell biology. Expanding radiation-resistant host T reg cells in recipient mice using a mouse TNFR2-selective agonist before allo-HCT significantly prolonged survival and reduced GvHD severity in a TNFR2- and T reg cell-dependent manner. The beneficial effects of transplanted T cells against leukemia cells and infectious pathogens remained unaffected. A corresponding human TNFR2-specific agonist expanded human T reg cells in vitro. These observations indicate the potential of our strategy to protect allo-HCT patients from acute GvHD by expanding T reg cells via selective TNFR2 activation in vivo.


Sujet(s)
Maladie du greffon contre l'hôte/prévention et contrôle , Récepteur au facteur de nécrose tumorale de type II/physiologie , Lymphocytes T régulateurs/immunologie , Maladie aigüe , Animaux , Femelle , Maladie du greffon contre l'hôte/immunologie , Transplantation de cellules souches hématopoïétiques , Interleukine-2/pharmacologie , Souris , Lignées consanguines de souris , Cellules myéloïdes suppressives/physiologie
3.
BMC Cancer ; 16: 115, 2016 Feb 17.
Article de Anglais | MEDLINE | ID: mdl-26883117

RÉSUMÉ

BACKGROUND: Medulloblastoma is the most common malignant brain tumor in children and can be divided in different molecular subgroups. Patients whose tumor is classified as a Group 3 tumor have a dismal prognosis. However only very few tumor models are available for this subgroup. METHODS: We established a robust orthotopic xenograft model with a cell line derived from the malignant pleural effusions of a child suffering from a Group 3 medulloblastoma. RESULTS: Besides classical characteristics of this tumor subgroup, the cells display cancer stem cell characteristics including neurosphere formation, multilineage differentiation, CD133/CD15 expression, high ALDH-activity and high tumorigenicity in immunocompromised mice with xenografts exactly recapitulating the original tumor architecture. CONCLUSIONS: This model using unmanipulated, human medulloblastoma cells will enable translational research, specifically focused on Group 3 medulloblastoma.


Sujet(s)
Médulloblastome/anatomopathologie , Tumeurs expérimentales/anatomopathologie , Animaux , Marqueurs biologiques tumoraux , Lignée cellulaire tumorale , Femelle , Humains , Nourrisson , Mâle , Souris , Souris SCID , Cellules souches tumorales , Tests d'activité antitumorale sur modèle de xénogreffe
4.
Blood ; 126(4): 437-44, 2015 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-26012567

RÉSUMÉ

Inhibition of the tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK)/fibroblast growth factor-inducible 14 (Fn14) system reduces intestinal cell death and disease development in several models of colitis. In view of the crucial role of TNF and intestinal cell death in graft-versus-host disease (GVHD) and the ability of TWEAK to enhance TNF-induced cell death, we tested here the therapeutic potential of Fn14 blockade on allogeneic hematopoietic cell transplantation (allo-HCT)-induced intestinal GVHD. An Fn14-specific blocking human immunoglobulin G1 antibody variant with compromised antibody-dependent cellular cytotoxicity (ADCC) activity strongly inhibited the severity of murine allo-HCT-induced GVHD. Treatment of the allo-HCT recipients with this monoclonal antibody reduced cell death of gastrointestinal cells but neither affected organ infiltration by donor T cells nor cytokine production. Fn14 blockade also inhibited intestinal cell death in mice challenged with TNF. This suggests that the protective effect of Fn14 blockade in allo-HCT is based on the protection of intestinal cells from TNF-induced apoptosis and not due to immune suppression. Importantly, Fn14 blockade showed no negative effect on graft-versus-leukemia/lymphoma (GVL) activity. Thus, ADCC-defective Fn14-blocking antibodies are not only possible novel GVL effect-sparing therapeutics for the treatment of GVHD but might also be useful for the treatment of other inflammatory bowel diseases where TNF-induced cell death is of relevance.


Sujet(s)
Anticorps monoclonaux d'origine murine/usage thérapeutique , Apoptose , Maladie du greffon contre l'hôte/prévention et contrôle , Transplantation de cellules souches hématopoïétiques/effets indésirables , Intestins/anatomopathologie , Récepteurs aux facteurs de nécrose tumorale/antagonistes et inhibiteurs , Inhibiteurs du facteur de nécrose tumorale , Animaux , Cytotoxicité à médiation cellulaire dépendante des anticorps , Technique de Western , Cellules cultivées , Cytokine TWEAK , Modèles animaux de maladie humaine , Femelle , Technique d'immunofluorescence , Maladie du greffon contre l'hôte/étiologie , Maladie du greffon contre l'hôte/métabolisme , Maladie du greffon contre l'hôte/anatomopathologie , Humains , Immunoglobuline G/administration et posologie , Immunoglobuline G/immunologie , Muqueuse intestinale/métabolisme , Intestins/immunologie , Mesures de luminescence , Mâle , Souris , Souris de lignée BALB C , Souris de lignée C57BL , ARN messager/génétique , Réaction de polymérisation en chaine en temps réel , Récepteurs du fragment Fc des IgG/immunologie , Récepteurs du fragment Fc des IgG/métabolisme , Récepteurs aux facteurs de nécrose tumorale/génétique , Récepteurs aux facteurs de nécrose tumorale/immunologie , Récepteurs aux facteurs de nécrose tumorale/métabolisme , RT-PCR , Rituximab , Récepteur TWEAK , Facteur de nécrose tumorale alpha/pharmacologie , Facteurs de nécrose tumorale/immunologie , Facteurs de nécrose tumorale/métabolisme
5.
J Leukoc Biol ; 96(4): 633-45, 2014 Oct.
Article de Anglais | MEDLINE | ID: mdl-25030422

RÉSUMÉ

Cytokine secretion and degranulation represent key components of CD8(+) T-cell cytotoxicity. While transcriptional blockade of IFN-γ and inhibition of degranulation by TGF-ß are well established, we wondered whether TGF-ß could also induce immune-regulatory miRNAs in human CD8(+) T cells. We used miRNA microarrays and high-throughput sequencing in combination with qRT-PCR and found that TGF-ß promotes expression of the miR-23a cluster in human CD8(+) T cells. Likewise, TGF-ß up-regulated expression of the cluster in CD8(+) T cells from wild-type mice, but not in cells from mice with tissue-specific expression of a dominant-negative TGF-ß type II receptor. Reporter gene assays including site mutations confirmed that miR-23a specifically targets the 3'UTR of CD107a/LAMP1 mRNA, whereas the further miRNAs expressed in this cluster-namely, miR-27a and -24-target the 3'UTR of IFN-γ mRNA. Upon modulation of the miR-23a cluster by the respective miRNA antagomirs and mimics, we observed significant changes in IFN-γ expression, but only slight effects on CD107a/LAMP1 expression. Still, overexpression of the cluster attenuated the cytotoxic activity of antigen-specific CD8(+) T cells. These functional data thus reveal that the miR-23a cluster not only is induced by TGF-ß, but also exerts a suppressive effect on CD8(+) T-cell effector functions, even in the absence of TGF-ß signaling.


Sujet(s)
Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Cytotoxicité immunologique , Déterminants antigéniques des lymphocytes T/immunologie , Interféron gamma/métabolisme , microARN/génétique , Facteur de croissance transformant bêta/métabolisme , Régions 3' non traduites , Séquence nucléotidique , Sites de fixation , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lignée cellulaire , Cellules cultivées , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Humains , Interféron gamma/composition chimique , Interféron gamma/génétique , Activation des lymphocytes/génétique , Activation des lymphocytes/immunologie , Protéine de membrane-1 associée au lysosome/composition chimique , Protéine de membrane-1 associée au lysosome/génétique , Antigène MART-1/immunologie , Famille multigénique , Interférence par ARN , Facteur de croissance transformant bêta/pharmacologie
6.
PLoS One ; 8(12): e81320, 2013.
Article de Anglais | MEDLINE | ID: mdl-24349055

RÉSUMÉ

To promote cancer research and to develop innovative therapies, refined pre-clinical mouse tumor models that mimic the actual disease in humans are of dire need. A number of neoplasms along the B cell lineage are commonly initiated by a translocation recombining c-myc with the immunoglobulin heavy-chain gene locus. The translocation is modeled in the C.129S1-Igha(tm1(Myc)Janz)/J mouse which has been previously engineered to express c-myc under the control of the endogenous IgH promoter. This transgenic mouse exhibits B cell hyperplasia and develops diverse B cell tumors. We have isolated tumor cells from the spleen of a C.129S1-Igha(tm1(Myc)Janz)/J mouse that spontaneously developed a plasmablastic lymphoma-like disease. These cells were cultured, transduced to express eGFP and firefly luciferase, and gave rise to a highly aggressive, transplantable B cell lymphoma cell line, termed IM380. This model bears several advantages over other models as it is genetically induced and mimics the translocation that is detectable in a number of human B cell lymphomas. The growth of the tumor cells, their dissemination, and response to treatment within immunocompetent hosts can be imaged non-invasively in vivo due to their expression of firefly luciferase. IM380 cells are radioresistant in vivo and mice with established tumors can be allogeneically transplanted to analyze graft-versus-tumor effects of transplanted T cells. Allogeneic hematopoietic stem cell transplantation of tumor-bearing mice results in prolonged survival. These traits make the IM380 model very valuable for the study of B cell lymphoma pathophysiology and for the development of innovative cancer therapies.


Sujet(s)
Imagerie diagnostique/méthodes , Mesures de luminescence/méthodes , Lymphome B/anatomopathologie , Animaux , Lymphocytes B , Lignée cellulaire tumorale , Femelle , Cytométrie en flux , Transplantation de cellules souches hématopoïétiques , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Transplantation homologue
7.
PLoS One ; 8(9): e75737, 2013.
Article de Anglais | MEDLINE | ID: mdl-24098720

RÉSUMÉ

Multiple activities are ascribed to the cytokine tumor necrosis factor (TNF) in health and disease. In particular, TNF was shown to affect carcinogenesis in multiple ways. This cytokine acts via the activation of two cell surface receptors, TNFR1, which is associated with inflammation, and TNFR2, which was shown to cause anti-inflammatory signaling. We assessed the effects of TNF and its two receptors on the progression of pancreatic cancer by in vivo bioluminescence imaging in a syngeneic orthotopic tumor mouse model with Panc02 cells. Mice deficient for TNFR1 were unable to spontaneously reject Panc02 tumors and furthermore displayed enhanced tumor progression. In contrast, a fraction of wild type (37.5%), TNF deficient (12.5%), and TNFR2 deficient mice (22.2%) were able to fully reject the tumor within two weeks. Pancreatic tumors in TNFR1 deficient mice displayed increased vascular density, enhanced infiltration of CD4(+) T cells and CD4(+) forkhead box P3 (FoxP3)(+) regulatory T cells (Treg) but reduced numbers of CD8(+) T cells. These alterations were further accompanied by transcriptional upregulation of IL4. Thus, TNF and TNFR1 are required in pancreatic ductal carcinoma to ensure optimal CD8(+) T cell-mediated immunosurveillance and tumor rejection. Exogenous systemic administration of human TNF, however, which only interacts with murine TNFR1, accelerated tumor progression. This suggests that TNFR1 has basically the capability in the Panc02 model to trigger pro-and anti-tumoral effects but the spatiotemporal availability of TNF seems to determine finally the overall outcome.


Sujet(s)
Carcinome canalaire/physiopathologie , Régulation de l'expression des gènes/immunologie , Tumeurs du pancréas/physiopathologie , Récepteur au facteur de nécrose tumorale de type II/métabolisme , Récepteur au facteur de nécrose tumorale de type I/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Animaux , Lymphocytes T CD4+/immunologie , Carcinome canalaire/immunologie , Carcinome canalaire/métabolisme , Lignée cellulaire tumorale , Amorces ADN/génétique , Cytométrie en flux , Interleukine-4/métabolisme , Mesures de luminescence , Souris , Souris de lignée C57BL , Microscopie de fluorescence , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/métabolisme , Réaction de polymérisation en chaine en temps réel , Récepteur au facteur de nécrose tumorale de type I/déficit , Récepteur au facteur de nécrose tumorale de type II/déficit , RT-PCR
8.
Blood ; 122(7): 1203-13, 2013 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-23836556

RÉSUMÉ

Src-kinase inhibitors hold great potential as targeted therapy against malignant cells. However, such inhibitors may also affect nonmalignant cells and cause pronounced off-target effects. We investigated the role of the dual kinase inhibitor dasatinib on human myeloid cells. Dasatinib is clinically used for the treatment of bcr/abl⁺ leukemias because it blocks the mutated tyrosine kinase abl. To understand its effect on the development of antigen-specific T-cell responses, we assessed antigen-specific priming of human, naïve T cells. In surprising contrast to the direct inhibition of T-cell activation by dasatinib, pretreatment of maturing dendritic cells (DCs) with dasatinib strongly enhanced their stimulatory activity. This effect strictly depended on the activating DC stimulus and led to enhanced interleukin 12 (IL-12) production and T-cell responses of higher functional avidity. Src-kinase inhibitors, and not conventional tyrosine kinase inhibitors, increased IL-12 production in several cell types of myeloid origin, such as monocytes and classical or nonclassical DCs. Interestingly, only human cells, but not mouse or macaques DCs, were affected. These data highlight the potential immunostimulatory capacity of a group of novel drugs, src-kinase inhibitors, thereby opening new opportunities for chemoimmunotherapy. These data also provide evidence for a regulatory role of src kinases in the activation of myeloid cells.


Sujet(s)
Cellules dendritiques/effets des médicaments et des substances chimiques , Interleukine-12/métabolisme , Cellules myéloïdes/effets des médicaments et des substances chimiques , Pyrimidines/pharmacologie , Lymphocytes T/effets des médicaments et des substances chimiques , Thiazoles/pharmacologie , Récepteurs de type Toll/métabolisme , src-Family kinases/antagonistes et inhibiteurs , Animaux , Cellules cultivées , Dasatinib , Cellules dendritiques/immunologie , Cellules dendritiques/anatomopathologie , Cytométrie en flux , Humains , Activation des lymphocytes/effets des médicaments et des substances chimiques , Macaca mulatta , Souris , Cellules myéloïdes/immunologie , Cellules myéloïdes/anatomopathologie , Facteur de transcription NF-kappa B/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Inhibiteurs de protéines kinases/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Lymphocytes T/immunologie , Lymphocytes T/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...