Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 5 de 5
1.
Bone ; 73: 51-9, 2015 Apr.
Article En | MEDLINE | ID: mdl-25532478

High doses of bone resorption inhibitors are currently under evaluation in pediatric oncology. Previous works have evidenced transient arrest in long bone and skull bone growth and tooth eruption blockage when mice were treated with zoledronic acid (ZOL). The question of potential similar effects with a RANKL-blocking antibody (IK22.5) was raised. Sensitivity disparities in these inhibitors between mouse strains and synergic effects of zoledronic acid and a RANKL-blocking antibody were subsidiary questions. In order to answer these questions, newborn C57BL/6J and CD1 mice were injected every two or three days (4 injections in total so 7 or 10 days of treatment length) with high doses of a RANKL-blocking antibody. The consequences on the tibia, craniofacial bones and teeth were analyzed by µCT and histology at the end of the treatment and one, two and three months later. The results obtained showed that RANKL-blocking antibody injections induced a transient arrest of tibia and skull bone growth and an irreversible blockage of tooth eruption in C57BL/6J mice. In CD1 mice, tooth eruption defects were also present but only at much higher doses. Similar mouse strain differences were obtained with zoledronic acid. Finally, a synergic effect of the two inhibitors was evidenced. In conclusion as previously observed for bisphosphonates (ZOL), a RANKL-blocking antibody induced a transient arrest in long bone and skull bone growth and a blockage of tooth eruption with however disparities between mouse strains with regard to this last effect. A synergic effect of both bone resorption inhibitors was also demonstrated.


Antibodies/administration & dosage , Bone Density Conservation Agents/pharmacology , Bone Development/immunology , Diphosphonates/pharmacology , Imidazoles/pharmacology , RANK Ligand/immunology , Animals , Animals, Newborn , Antibodies/immunology , Bone Development/drug effects , Female , Mice , Mice, Inbred C57BL , Pregnancy , Tooth Eruption/immunology , Zoledronic Acid
2.
Int J Cancer ; 136(4): 784-96, 2015 Feb 15.
Article En | MEDLINE | ID: mdl-24961790

It has been established that disturbances in intracellular signaling pathways play a considerable part in the oncologic process. Phosphatidylinositol-3-kinase (PI3K) has become of key interest in cancer therapy because of its high mutation frequency and/or gain in function of its catalytic subunits in cancer cells. We investigated the therapeutic value of BYL719, a new specific PI3Kα inhibitor that blocks the ATP site, on osteosarcoma and bone cells. The in vitro effects of BYL719 on proliferation, apoptosis, and cell cycle were assessed in human and murine osteosarcoma cell. Its impact on bone cells was determined using human mesenchymal stem cells (hMSC) and human CD14+ osteoclast precursors. Two different murine preclinical models of osteosarcoma were used to analyze the in vivo biological activities of BYL719. BYL719 decreased cell proliferation by blocking cell cycle in G0/G1 phase with no outstanding effects on apoptosis cell death in HOS and MOS-J tumor cells. BYL719 inhibited cell migration and can thus be considered as a cytostatic drug for osteosarcoma. In murine preclinical models of osteosarcoma, BYL719 significantly decreased tumor progression and tumor ectopic bone formation as shown by a decrease of Ki67+ cells and tumor vascularization. To explore the maximum therapeutic potential of BYL719, the drug was studied in combination with conventional chemotherapeutic drugs, revealing promising efficacy with ifosfamide. BYL719 also exhibited dual activities on osteoblast and osteoclast differentiation. Overall, the present work shows that BYL719 is a promising drug in either a single or multidrug approach to curing bone sarcoma.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bone Neoplasms/drug therapy , Osteosarcoma/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Neoplasms/pathology , Cell Line, Tumor , Drug Synergism , Female , Humans , Inhibitory Concentration 50 , Male , Mice, Inbred C57BL , Mice, Nude , Osteoblasts/drug effects , Osteoclasts/drug effects , Osteosarcoma/pathology , Thiazoles/administration & dosage , Tumor Burden , Xenograft Model Antitumor Assays
3.
Clin Sci (Lond) ; 127(5): 277-93, 2014 Sep.
Article En | MEDLINE | ID: mdl-24827940

Aseptic loosening as a result of wear debris is considered to be the main cause of long-term implant failure in orthopaedic surgery and improved biomaterials for bearing surfaces decreases significantly the release of micrometric wear particles. Increasingly, in-depth knowledge of osteoimmunology highlights the role of nanoparticles and ions released from some of these new bearing couples, opening up a new era in the comprehension of aseptic loosening. Mouse models have been essential in the progress made in the early comprehension of pathophysiology and in testing new therapeutic agents for particle-induced osteolysis. However, despite this encouraging progress, there is still no valid clinical alternative to revision surgery. The present review provides an update of the most commonly used bearing couples, the current concepts regarding particle-cell interactions and the approaches used to study the biology of periprosthetic osteolysis. It also discusses the contribution and future challenges of mouse models for successful translation of the preclinical progress into clinical applications.


Arthroplasty, Replacement/adverse effects , Macrophages/physiology , Osteolysis/etiology , Animals , Biomechanical Phenomena , Ceramics/adverse effects , Clinical Trials as Topic , Disease Models, Animal , Equipment Failure , Humans , Inflammation/physiopathology , Mice , Nanoparticles/adverse effects , Osteolysis/physiopathology , Particle Size , Polyethylenes/adverse effects , Polyethylenes/chemistry , Polymethyl Methacrylate/adverse effects , Reoperation , Translational Research, Biomedical
4.
PLoS One ; 9(3): e90795, 2014.
Article En | MEDLINE | ID: mdl-24599309

Osteosarcoma is the most common primary malignant bone tumour characterized by osteoid production and/or osteolytic lesions of bone. A lack of response to chemotherapeutic treatments shows the importance of exploring new therapeutic methods. Imatinib mesylate (Gleevec, Novartis Pharma), a tyrosine kinase inhibitor, was originally developed for the treatment of chronic myeloid leukemia. Several studies revealed that imatinib mesylate inhibits osteoclast differentiation through the M-CSFR pathway and activates osteoblast differentiation through PDGFR pathway, two key cells involved in the vicious cycle controlling the tumour development. The present study investigated the in vitro effects of imatinib mesylate on the proliferation, apoptosis, cell cycle, and migration ability of five osteosarcoma cell lines (human: MG-63, HOS; rat: OSRGA; mice: MOS-J, POS-1). Imatinib mesylate was also assessed as a curative and preventive treatment in two syngenic osteosarcoma models: MOS-J (mixed osteoblastic/osteolytic osteosarcoma) and POS-1 (undifferentiated osteosarcoma). Imatinib mesylate exhibited a dose-dependent anti-proliferative effect in all cell lines studied. The drug induced a G0/G1 cell cycle arrest in most cell lines, except for POS-1 and HOS cells that were blocked in the S phase. In addition, imatinib mesylate induced cell death and strongly inhibited osteosarcoma cell migration. In the MOS-J osteosarcoma model, oral administration of imatinib mesylate significantly inhibited the tumour development in both preventive and curative approaches. A phospho-receptor tyrosine kinase array kit revealed that PDGFRα, among 7 other receptors (PDFGFRß, Axl, RYK, EGFR, EphA2 and 10, IGF1R), appears as one of the main molecular targets for imatinib mesylate. In the light of the present study and the literature, it would be particularly interesting to revisit therapeutic evaluation of imatinib mesylate in osteosarcoma according to the tyrosine-kinase receptor status of patients.


Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Benzamides/pharmacology , Benzamides/therapeutic use , Immunocompetence/drug effects , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Piperazines/pharmacology , Piperazines/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Animals , Caspases/metabolism , Cell Cycle Checkpoints/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Disease Progression , Humans , Imatinib Mesylate , Male , Mice , Mice, Inbred C57BL , Mitosis/drug effects , Osteosarcoma/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
5.
Cancer Lett ; 344(2): 291-8, 2014 Mar 28.
Article En | MEDLINE | ID: mdl-24333720

Despite recent improvements in chemotherapy and surgery, the problem of non-response osteosarcoma to chemotherapy remains, and is a parameter that is critical for prognosis. The present work investigated the therapeutic value of NVP-BEZ235, a dual class I PI3K/mTOR inhibitor. NVP-BEZ235 inhibited osteosarcoma cell proliferation by inducing G0/G1 cell cycle arrest with no caspase activation. In murine pre-clinical models, NVP-BEZ235 significantly slowed down tumor progression and ectopic tumor bone formation with decreased numbers of Ki67(+) cells and reduced tumor vasculature. Finally, NVP-BEZ235 considerably improved the survival rate of mice with osteosarcoma. Taken together, the results of the present work show that NVP-BEZ235 exhibits therapeutic interest in osteosarcoma and may be a promising adjuvant drug for bone sarcomas.


Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Imidazoles/pharmacology , Osteosarcoma/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Quinolines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Bone Neoplasms/enzymology , Bone Neoplasms/pathology , Cell Growth Processes/drug effects , Cell Line, Tumor , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Osteosarcoma/enzymology , Osteosarcoma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Random Allocation , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
...