Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Crit Rev Oncol Hematol ; 156: 103126, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33113487

ABSTRACT

Osteosarcoma (OS) is the most common cancer of bone. Jaw osteosarcoma (JOS) is rare and it differs from long-bone OS (LBOS) in terms of the time of onset (two decades later), lower metastatic spread, and better survival. OS is characterized by the proliferation of osteoblastic precursor cells and the production of osteoid or immature bone. OS arises from a combination of genetic aberrations and a favourable microenvironment. This local microenvironment includes bone cells, blood vessels, stromal cells, and immune infiltrates, all of which may constitute potential targets for anti-cancer drugs. Differences in the clinical and biological behaviour of JOS versus LBOS are likely to at least in part be due to differences in the microenvironment between the two sites. The present review provides a brief overview of the known pathophysiological parameters involved in JOS.


Subject(s)
Bone Neoplasms , Osteosarcoma , Bone and Bones , Humans , Nigeria , Stromal Cells , Tumor Microenvironment
2.
Sarcoma ; 2011: 932451, 2011.
Article in English | MEDLINE | ID: mdl-21647363

ABSTRACT

Chondrosarcomas are malignant cartilage-forming tumours representing around 20% of malignant primary tumours of bone and affect mainly adults in the third to sixth decade of life. Unfortunately, the molecular pathways controlling the genesis and the growth of chondrosarcoma cells are still not fully defined. It is well admitted that the invasion of bone by tumour cells affects the balance between early bone resorption and formation and induces an "inflammatory-like" environment which establishes a dialogue between tumour cells and their environment. The bone tumour microenvironment is then described as a sanctuary that contributes to the drug resistance patterns and may control at least in part the tumour growth. The concept of "niche" defined as a specialized microenvironment that can promote the emergence of tumour stem cells and provide all the required factors for their development recently emerges in the literature. The present paper aims to summarize the main evidence sustaining the existence of a specific bone niche in the pathogenesis of chondrosarcomas.

3.
Curr Pharm Des ; 16(27): 2981-7, 2010.
Article in English | MEDLINE | ID: mdl-20722622

ABSTRACT

Bone tumours can be dissociated in two main categories: i) primary bone tumours (benign or malignant) including mainly osteosarcoma and other sarcomas.ii)and giant cell tumour and bone metastases originate from others cancer (Breast, prostate, kidney cancer, etc). These tumours are able to destroy or/and induce a new calcified matrix. However, the first step of bone tumour development is associated with an induction of bone resorption and the establishment of a vicious cycle between the osteoclasts and the tumour growth. Indeed, bone resorption contributes to the pathogenesis of bone tumour by the release of cytokines (IL6, TNFα) which govern the bone tumour's development and which are trapped into the bone matrix. Bisphosphonates (BPs) are chemical compounds of P-C-P structure with a high affinity for bone hydroxyapatite crystals. Thus, they have been used as a carrier for radio nucleotides to develop novel approaches of bone imaging. BPs exert also indirect anti-tumour activities in vivo. Indeed, BPs directly interfere with the bone microenvironment and target osteoclasts, endothelial cells and immune cells (tumour-associated macrophages, γ9δ2 T cells). BPs induce tumour cell death in vitro and same activity is suspected in vivo. The present review summarizes the mechanisms of actions of BPs as well as their clinical interests in bone primary tumours.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Bone Neoplasms/drug therapy , Diphosphonates/pharmacology , Diphosphonates/therapeutic use , Drug Design , Animals , Bone Density Conservation Agents/pharmacology , Bone Density Conservation Agents/therapeutic use , Bone Neoplasms/diagnostic imaging , Chondrosarcoma/drug therapy , Giant Cell Tumor of Bone/drug therapy , Humans , Osteosarcoma/drug therapy , Radiography
4.
Cancer Gene Ther ; 17(6): 387-97, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20075983

ABSTRACT

Osteosarcoma is the most common malignant primary bone tumor for which pertinent preclinical models are still needed to develop new therapeutic strategies. As osteosarcoma growth is strongly supported by bone resorption, previous studies have inhibited the cytokine receptor activator of nuclear factor-kappaB ligand using antibodies or recombinant proteins. However, its expression has not yet been inhibited using genetic approaches using small interfering RNA. To optimize the delivery of small interfering RNA to its cellular target and demonstrate their efficiency in vivo, two new osteosarcoma models expressing the firefly luciferase enzyme were developed. These luciferase-expressing osteosarcomas showed conserved osteolytic and osteogenic activities in mice and were detectable by in vivo bioluminescence imaging. In comparison with measurement of tumor volume, bioluminescence analysis enabled earlier tumor detection and revealed extensive cell death in response to ifosfamide treatment. Finally, by targeting the luciferase expression into osteosarcoma, we established a protocol for in vivo administration of small interfering RNA combined with cationic liposome.


Subject(s)
Luminescent Proteins/metabolism , Osteosarcoma/metabolism , RNA Interference , RNA, Small Interfering/genetics , Animals , Cell Line , Cell Line, Tumor , Flow Cytometry , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Lentivirus/genetics , Luciferases, Firefly/genetics , Luciferases, Firefly/metabolism , Luminescent Proteins/genetics , Male , Mice , Mice, Inbred C3H , Mice, Inbred Strains , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Rats , Transfection , Transplantation, Heterologous , Tumor Cells, Cultured
5.
Biochim Biophys Acta ; 1805(1): 17-24, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19733222

ABSTRACT

Tumor development in bone is often associated with fractures, bone loss and bone pain, and improvement is still needed in therapeutic approaches. Bone tumors are related to the existence of a vicious cycle between bone resorption and tumor proliferation in which the molecular triad osteoprotegerin (OPG)/receptor activator of NF-kappaB (RANK)/RANK ligand (RANKL) plays a pivotal role. RANKL, a member of the TNF superfamily, is one of the main inducers of bone resorption. Its soluble receptor OPG represents a promising therapeutic candidate as it prevents bone lesions and inhibits associated tumor growth. However, its therapeutic use in bone tumors remains controversial due to its ability to bind and inhibit another member of the TNF superfamily, TNF related apoptosis inducing ligand (TRAIL), which is a potent inducer of tumor cell apoptosis. Through its heparin binding domain, OPG is also able to bind proteoglycans present in the bone matrix. This paper is an overview of the involvement of the micro-environment, as represented by the balance of RANKL/TRAIL and the presence of proteoglycans in the regulation of OPG biological activity in bone tumors.


Subject(s)
Bone Neoplasms/metabolism , Osteoprotegerin/metabolism , RANK Ligand/physiology , TNF-Related Apoptosis-Inducing Ligand/physiology , Bone Resorption , Humans
6.
Histol Histopathol ; 24(2): 235-42, 2009 02.
Article in English | MEDLINE | ID: mdl-19085839

ABSTRACT

Primary and secondary bone tumors clearly deteriorate quality of life and the activity of daily living of patients. These undesirable diseases become a major social and economic burden. As both primary and secondary bone tumors develop in the unique bone tissue, it is therefore necessary to understand bone cell biology in tumor bone environment. Recent findings of the Receptor Activator of Nuclear Factor-kappaB ligand (RANKL)/RANK/osteoprotegerin (OPG) molecular triad, the key regulators of bone remodeling, opened new era of bone research. Although RANK is an essential receptor for osteoclast formation, activation and survival, functional RANK expression has been recently identified on several bone-associated tumor cells. When RANK is expressed on secondary bone tumor cells, it is implicated in tumor cell migration, whereas this is not the case for primary bone tumors. In any case, RANK is not involved in RANK-positive cell proliferation or death. In two models of bone metastases secondary to melanoma or prostate carcinoma, in vivo neutralization of RANKL by OPG resulted in complete protection from paralysis, due to metastases of vertebral body, and a marked reduction in tumor burden in bones, but not in other organs. OPG also decreased tumor formation and tumor burden in a mouse model of primary bone tumor, osteosarcoma. In all these models, tumor cells express RANK. These data revealed that local differentiation factors, such as RANKL, play an important role in cell migration in a metastatic tissue-specific manner. These findings substantiate the novel direct role of RANKL/RANK in bone-associated tumors, and its capability of representing new therapeutic targets.


Subject(s)
Bone Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Neoplasms/metabolism , RANK Ligand/physiology , Receptor Activator of Nuclear Factor-kappa B/physiology , Animals , Apoptosis , Bone and Bones/metabolism , Cell Movement , Female , Humans , Male , Mice , Models, Biological , Neoplasm Metastasis , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism
7.
J Cell Mol Med ; 12(3): 928-41, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18494934

ABSTRACT

We recently demonstrated original anti-tumor effects of zoledronic acid (Zol) on osteosarcoma cell lines independently of their p53 and Rb status. The present study investigated the potential Zol-resistance acquired by osteosarcoma cells after prolonged treatment. After 12 weeks of culture in the presence of 1 microm Zol, the effects of high doses of Zol (10-100 microm) were compared between the untreated rat (OSRGA, ROS) and human (MG63, SAOS2) osteosarcoma cells and Zol-pretreated cells in terms of cell proliferation, cell cycle analysis, migration assay and cytoskeleton organization. Long-term treatment with 1 microm Zol reduced the sensitivity of osteosarcoma cells to high concentrations of Zol. Furthermore, the Zol-resistant cells were sensitive to conventional anti-cancer agents demonstrating that this resistance process is independent of the multidrug resistance phenotype. However, as similar experiments performed in the presence of clodronate and pamidronate evidenced that this drug resistance was restricted to the nitrogen-containing bisphosphonates, we then hypothesized that this resistance could be associated with a differential expression of farnesyl diphos-phate synthase (FPPS) also observed in human osteosarcoma samples. The transfection of Zol-resistant cells with FPPS siRNA strongly increased their sensitivity to Zol. This study demonstrates for the first time the induction of metabolic resistance after prolonged Zol treatment of osteosarcoma cells confirming the therapeutic potential of Zol for the treatment of bone malignant pathologies, but points out the importance of the treatment regimen may be important in terms of duration and dose to avoid the development of drug metabolic resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Diphosphonates/pharmacology , Drug Resistance, Neoplasm/drug effects , Geranyltranstransferase/metabolism , Imidazoles/pharmacology , Osteosarcoma/drug therapy , Adolescent , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Osteosarcoma/diagnosis , Osteosarcoma/metabolism , Osteosarcoma/pathology , RNA, Small Interfering/pharmacology , Time Factors , Transfection , Zoledronic Acid
8.
Cell Mol Life Sci ; 64(18): 2334-50, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17530461

ABSTRACT

1997 saw the identification of a novel set of proteins within the tumor necrosis factor (TNF)/TNF receptor families that are required for the control of bone remodeling. Therefore, these receptors, receptor activator of nuclear factor kappa B (RANK), osteoprotegerin (OPG) and their ligand RANK ligand (RANKL) became the critical molecular triad controlling osteoclastogenesis and pathophysiologic bone remodeling. However, the establishment of the corresponding knock-out and transgenic mice revealed unexpected results, most particularly, the involvement of these factors in the vascular system and immunity. Thus, the OPG/RANK/RANKL molecular triad appears to be associated with vascular calcifications and plays a pivotal function in the development of the immune system through dendritic cells. OPG/RANK/RANKL thus constitute a molecular bridge spanning bone metabolism, vascular biology and immunity. This review summarizes recent knowledge of OPG/RANK/RANKL interactions and activities as well as the current evidence for their participation in osteoimmunology and vascular diseases. In fine, the targeting of the OPG/RANK/RANKL axis as novel therapeutic approaches will be discussed.


Subject(s)
Bone and Bones/immunology , Osteoprotegerin/metabolism , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Vascular Diseases/metabolism , Animals , Bone and Bones/cytology , Humans , Mice , Osteoprotegerin/chemistry , RANK Ligand/chemistry , Receptor Activator of Nuclear Factor-kappa B/chemistry
9.
Oncogene ; 26(46): 6653-64, 2007 Oct 11.
Article in English | MEDLINE | ID: mdl-17471233

ABSTRACT

Oncostatin M (OSM), a cytokine of the interleukin-6 family, induces growth arrest and differentiation of osteoblastic cells into glial-like/osteocytic cells. Here, we asked whether OSM regulates apoptosis of normal or transformed (osteosarcoma) osteoblasts. We show that OSM sensitizes cells to apoptosis induced by various death inducers such as staurosporine, ultraviolet or tumor necrosis factor-alpha. Apoptosis is mediated by the mitochondrial pathway, with release of cytochrome c from the mitochondria to the cytosol and activation of caspases-9 and -3. DNA micro-arrays revealed that OSM modulates the expression of Bax, Bad, Bnip3, Bcl-2 and Mcl-1. Pharmacological inhibitors, dominant-negative signal transducer and activator of transcriptions (STATs), stable RNA interference and knockout cells indicated that the transcription factors p53 and STAT5, which are activated by OSM, are implicated in the sensitization to apoptosis, being responsible for Bax induction and Bcl-2 reduction, respectively. These results indicate that, in addition to growth arrest and induced differentiation, OSM also sensitizes normal and transformed osteoblasts to apoptosis by a mechanism implicating (i) activation and nuclear translocation of STAT5 and p53 and (ii) an increased Bax/Bcl-2 ratio. Therefore, association of OSM with kinase inhibitors such as Sts represents new therapeutic opportunities for wild-type p53 osteosarcoma.


Subject(s)
Apoptosis , Oncostatin M/pharmacology , Apoptosis/drug effects , Carbazoles/pharmacology , Caspases/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Indole Alkaloids/pharmacology , Osteosarcoma , Proto-Oncogene Proteins c-bcl-2/metabolism , STAT5 Transcription Factor , Tumor Suppressor Protein p53 , bcl-2-Associated X Protein/metabolism
10.
J Pathol ; 211(5): 555-562, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17323424

ABSTRACT

RANK, RANK ligand (RANKL) and osteoprotegerin (OPG) are the key regulators of bone metabolism, both in normal and pathological conditions. Previous data have demonstrated that human osteosarcoma biopsies express RANKL as well as OPG, and functional RANK is expressed in a murine osteosarcoma cell line. As RANK expression in human osteosarcoma remains controversial, the aim of the present study was to analyse its expression in vitro in human osteosarcoma cell lines, ex vivo using pathological tissues, and then to determine its functionality in terms of signal transduction pathways modulated by RANKL. RT-PCR analysis and immunohistochemistry experiments revealed that RANK is expressed at both transcriptional and protein levels in MNNG/HOS, Saos-2 and MG-63 human osteosarcoma cell lines, in contrast to the U-2 OS osteosarcoma cell line and human osteoblasts, which were negative. RANK was also expressed in 57% of osteosarcoma biopsies. Furthermore, western blot experiments clearly demonstrated the functionality of RANK. Thus, RANKL significantly induced the phosphorylation of ERK1/2, p38 and IkappaB in RANK-positive osteosarcoma cells. This study is the first report of functional RANK expression in human osteosarcoma cells: this strengthens the involvement of the RANK-RANKL-OPG axis in primary bone tumour biology and identifies novel therapeutic approaches targeting RANK-positive osteosarcoma.


Subject(s)
Bone Neoplasms/chemistry , Neoplasm Proteins/analysis , Osteosarcoma/chemistry , Receptor Activator of Nuclear Factor-kappa B/analysis , Adolescent , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Child , Cross-Sectional Studies , Female , Humans , Immunohistochemistry/methods , Male , Middle Aged , Osteoblasts/chemistry , Osteoprotegerin/analysis , Phosphorylation , RANK Ligand/metabolism , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Reverse Transcriptase Polymerase Chain Reaction/methods , Signal Transduction/physiology
11.
Bone ; 40(4): 981-90, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17196895

ABSTRACT

Prostate cancer metastases to bone are observed in around 80% of prostate cancer patients and represent the most critical complication of advanced prostate cancer, frequently resulting in significant morbidity and mortality. As the underlying mechanisms are not fully characterized, understanding the biological mechanisms that govern prostate cancer metastases to bone at the molecular level should lead to the determination of new potential therapeutic targets. Receptor activator of NFkappaB ligand (RANKL)/RANK/Osteoprotegerin (OPG) are the key regulators of bone metabolism both in normal and pathological condition, including prostate cancer bone metastases. In the present study, we demonstrated that human prostate cancer cell lines, DU145 and PC3 express biologically functional RANK. Indeed, soluble human RANKL (shRANKL, 100 ng/ml) treatment induced ERK 1/2, p38 and IkappaB phosphorylations in these cells. shRANKL administration also promoted DU145 and PC3 prostate cancer cell invasion in vitro. Whereas human OPG (hOPG) administration alone (100 ng/ml) had no marked effect, combined association of both agents abolished the RANKL-induced DU145 cell invasion. As RANKL had no direct effect on DU145 cell proliferation, the observed effects were indeed related to RANKL-induced cell migration. DU145 human prostate cancer cells promoted osteoclastogenesis of osteoclast precursors generated from mouse bone marrow. Moreover, DU145 cells produced soluble factor(s) that up-regulate the proliferation of MC3T3-E1 pre-osteoblasts through the activation of the ERK 1/2 and STAT3 signal transduction pathways. This stimulation of pre-osteoblast proliferation resulted in an increased local RANKL expression that can activate both osteoclasts/osteoclast precursors and prostate cancer cells, thus facilitating prostate cancer metastasis development in bone. We confirm that RANKL is a factor that facilitates metastasis to bone by acting as an activator of both osteoclasts and RANK-positive prostate cancer cells in our model. Furthermore, the present study provides the evidence that blocking RANKL-RANK interaction offer new therapeutic approach not only at the level of bone resorbing cells, but also by interfering with RANK-positive prostate cancer cells in the prostate cancer bone metastasis development.


Subject(s)
Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Prostatic Neoplasms/metabolism , RANK Ligand/metabolism , 3T3 Cells , Animals , Bone Neoplasms/etiology , Cell Line, Tumor , Culture Media, Conditioned , Humans , I-kappa B Proteins/metabolism , MAP Kinase Signaling System/drug effects , Male , Mice , Models, Biological , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoprotegerin/metabolism , Osteoprotegerin/pharmacology , RANK Ligand/pharmacology , Receptor Activator of Nuclear Factor-kappa B/metabolism , Recombinant Proteins/pharmacology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects
12.
Mol Pharmacol ; 71(1): 333-43, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17050806

ABSTRACT

The molecular mechanisms responsible for the cellular effects of the nitrogen-containing bisphosphonate zoledronic acid (Zol) were assessed on several osteosarcoma cell lines differing in their p53 and retinoblastoma (Rb) status. Zol inhibited cell proliferation and increased atypical apoptosis. The Zol effects on proliferation were due to cell cycle arrest in S and G2/M phases subsequent to the activation of the intra-S DNA damage checkpoint with an increase in P-ATR, P-chk1, Wee1, and P-cdc2 levels and a decrease in cdc25c, regardless of the p53 and Rb status. In addition, the atypic apoptosis induced by Zol was independent of caspase activation, and it was characterized by nuclear alterations, increased Bax expression, and reduced Bcl-2 level. Furthermore, mitochondrial permeability was up-regulated by Zol independently of p53 in association with the translocation of apoptosis-inducing factor (AIF) and endonuclease-G (EndoG). Zol also disturbed cytoskeletal organization and cell junctions and inhibited cell migration and phosphorylation of focal adhesion kinases. The main difficulty encountered in treating cancer relates to mutations in key genes such as p53, Rb, or proteins affecting caspase signaling carried by many tumor cells. We have demonstrated for the first time that zoledronic acid activated the DNA damage S-phase checkpoint and the mitochondrial pathway via AIF and EndoG translocation, and it inhibited cell proliferation and induced cell death, bypassing these potentials mutations. Therefore, zoledronic acid may be considered as an effective therapeutic agent in clinical trials of osteosarcoma in which mutation for p53 and Rb very often occur, and where current treatment with traditional chemotherapeutic agents is ineffective.


Subject(s)
Apoptosis Inducing Factor/metabolism , Cell Death/drug effects , DNA, Neoplasm/genetics , Diphosphonates/pharmacology , Endodeoxyribonucleases/metabolism , Imidazoles/pharmacology , Animals , Bone Density Conservation Agents/pharmacology , Bone Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Humans , Microscopy, Confocal , Osteosarcoma/pathology , Protein Transport , Rats , S Phase/drug effects , Tumor Suppressor Protein p53/metabolism , Zoledronic Acid
13.
Biochem Biophys Res Commun ; 347(2): 460-7, 2006 Aug 25.
Article in English | MEDLINE | ID: mdl-16828054

ABSTRACT

Osteoprotegerin (OPG) is a decoy receptor for receptor activator of nuclear factor kappaB ligand (RANKL), a key inducer of osteoclastogenesis via its receptor RANK. We previously showed that RANK, RANKL, and OPG are able to form a tertiary complex and that OPG must be also considered as a direct effector of osteoclast functions. As OPG contains a heparin-binding domain, the present study investigated the interactions between OPG and glycosaminoglycans (GAGs) by surface plasmon resonance and their involvement in the OPG functions. Kinetic data demonstrated that OPG binds to heparin with a high-affinity (KD: 0.28 nM) and that the pre-incubation of OPG with heparin inhibits in a dose-dependent manner the OPG binding to the complex RANK-RANKL. GAGs from different structure/origin (heparan sulfate, dermatan sulfate, and chondroitin sulfate) exert similar activity on OPG binding. The contribution of the sulfation pattern and the size of the oligosaccharide were determined in this inhibitory mechanism. The results demonstrated that sulfation is essential in the OPG-blocking function of GAGs since a totally desulfated heparin loses its capacity to bind and to block OPG binding to RANKL. Moreover, a decasaccharide is the minimal structure that totally inhibits the OPG binding to the complex RANK-RANKL. Western blot analysis performed in 293 cells surexpressing RANKL revealed that the pre-incubation of OPG with these GAGs strongly inhibits the OPG-induced decrease of membrane RANKL half-life. These data support an essential function of the related glycosaminoglycans heparin and heparan sulfate in the activity of the triad RANK-RANKL-OPG.


Subject(s)
Carrier Proteins/metabolism , Glycoproteins/metabolism , Glycosaminoglycans/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Surface Plasmon Resonance/methods , Binding, Competitive/drug effects , Blotting, Western , Carrier Proteins/genetics , Cell Line , Glycoproteins/chemistry , Glycoproteins/pharmacology , Glycosaminoglycans/chemistry , Glycosaminoglycans/pharmacology , Heparin/chemistry , Heparin/metabolism , Humans , Kinetics , Membrane Glycoproteins/genetics , Models, Biological , Oligosaccharides/pharmacology , Osteoprotegerin , Protein Binding/drug effects , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Tumor Necrosis Factor/chemistry , Sulfates/chemistry , Sulfates/metabolism , Time Factors
14.
BMC Cancer ; 5: 123, 2005 Sep 27.
Article in English | MEDLINE | ID: mdl-16188028

ABSTRACT

BACKGROUND: Osteosarcoma is the most common type of primary bone tumor. The use of aggressive chemotherapy has drastically improved the prognosis of the patients with non-metastatic osteosarcomas, however the prognosis of the patients with metastasis is still very poor. Then, new and more effective treatments for curing osteosarcoma, such as immunotherapy are needed. Tumor-infiltrating lymphocytes (TIL) have been involved in the control of tumor development and already assessed with success for the treatment of several cancers including melanoma. While TIL represent a fascinating therapeutic approach in numerous malignant pathologies, there is few report concerning adult bone-associated tumors including osteosarcoma. METHODS: Human TIL were isolated and characterized (phenotype, lytic activity) from twenty-seven patients with bone-associated tumors (osteosarcoma, Ewing's sarcoma, giant cell tumor, chondrosarcoma, plasmocytoma and bone metastases). Similar experiments were performed using rat osteosarcoma model. RESULTS: While TIL with a main CD4+ profile were easily isolated from most of the tumor samples, only TIL extracted from osteosarcoma were cytotoxic against allogeneic tumor cells. In all cases, TIL lytic activity was significantly higher compared to autologous peripheral blood leukocytes. Similar data were observed in rat osteosarcoma model where TIL were characterized by a main CD4+ profile and high lytic activity against allogeneic and autologous tumor cells. Moreover, rat TIL expansion was not accompanied by refractoriness to further activation stimulus mainly by tumor antigens. CONCLUSION: These results demonstrated that TIL therapy could be a very efficient strategy for the treatment of adult osteosarcoma.


Subject(s)
Bone Neoplasms/pathology , Lymphocytes/pathology , Osteolysis , Osteosarcoma/pathology , Adolescent , Adult , Aged , Animals , Bone Neoplasms/metabolism , CD4-Positive T-Lymphocytes/cytology , Cell Line, Tumor , Disease Models, Animal , Female , Flow Cytometry , Humans , Immunotherapy/methods , Leukocytes/pathology , Lymphocytes/metabolism , Lymphocytes, Tumor-Infiltrating/cytology , Male , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Osteosarcoma/metabolism , Osteosarcoma/therapy , Phenotype , Prognosis , Rats
15.
Tumour Biol ; 26(3): 121-30, 2005.
Article in English | MEDLINE | ID: mdl-15970646

ABSTRACT

Satisfactory experimental models for preclinical cancer studies must follow several criteria: (1) reproducibility of the method used to induce the tumor and (2) clinical, pathological and kinetic similarity with the corresponding human tumors. We developed a model of osteosarcoma locally induced by the intrafemoral injection of osteosarcoma (OSR) cells in Sprague-Dawley rats. This method yields nearly 80% of bone tumors at the injection site. These tumors double their volume fairly slowly (in approximately 20 days) and lung metastases occur in 96% of the animals. The OSR cell-induced tumor is characterized by a direct production of mineralized matrix by the tumor cells themselves, as revealed by histochemical analysis. The microarchitectural parameters which were quantified by a microscanner show an increased trabecular bone volume (+238%) when OSR cells were injected in the femur, as compared to controls injected with vehicle. Osteoblastic markers such as alkaline phosphatase, osteopontin, osteocalcin and bone sialoprotein were expressed by the tumor in vivo, whereas the initially injected OSR cells did not express some of these markers, suggesting that OSR cells reacquired an osteoblastic phenotype in a favorable environment. The clinical, radiological and histological data show that this model shares high similarities with the osteocondensing forms of osteosarcoma in humans.


Subject(s)
Biomarkers, Tumor/analysis , Bone Neoplasms/pathology , Bone Neoplasms/veterinary , Disease Models, Animal , Osteosarcoma/pathology , Osteosarcoma/veterinary , Animals , Bone Neoplasms/genetics , Cell Proliferation , Lung Neoplasms/secondary , Lung Neoplasms/veterinary , Osteosarcoma/genetics , Phenotype , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
16.
Bone ; 37(1): 74-86, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15894525

ABSTRACT

The efficacy of zoledronic acid (ZOL), with or without the anticancer drug ifosfamide (IFO), was tested on primary bone tumor growth using a rat-transplantable model of osteosarcoma. The effects on bone remodeling and tumor growth were analyzed by radiography, micro-computed tomography (micro-CT), and histological staining. The in vitro effects of ZOL were studied by proliferation, apoptosis, and cell cycle analyses on the osteosarcoma cells OSRGA compared to rat primary osteoblasts. Treatment with ZOL was effective in preventing the formation of osteolytic lesions that developed in bone sites and in reducing the local tumor growth, as compared to the untreated rats. The combination of ZOL and IFO was more effective than each agent alone in preventing tumor recurrence, improving tissue repair, and increasing bone formation as revealed by the analysis of trabecular architecture. In vitro studies demonstrated that ZOL was more potent against the OSRGA cell line than osteoblasts (with a half-maximal inhibitory effect on proliferation seen at 0.2 and 20 microM, respectively), the ZOL-induced inhibition of OSRGA proliferation being due to cell cycle arrest in S-phase. No effect on OSRGA apoptosis could be observed in vitro, as assessed by Hoechst staining and caspase-1 and -3 activation. In situ cell death was determined by TUNEL staining on tumor tissue sections. No significant difference in TUNEL-positive cells could be observed between ZOL-treated and -untreated rats. This is the first report of the anti-bone resorption and antitumoral activities of zoledronic acid in a rat model of osteosarcoma, and its beneficial association with an antitumoral chemotherapeutic drug in preventing tumor recurrence.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Osteosarcoma/drug therapy , Animals , Bone Remodeling/drug effects , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Diphosphonates/administration & dosage , Fibrosis , Ifosfamide/administration & dosage , Imidazoles/administration & dosage , Male , Necrosis , Neoplasm Metastasis/drug therapy , Neoplasm Recurrence, Local/drug therapy , Osteoblasts/metabolism , Osteogenesis/drug effects , Osteosarcoma/mortality , Osteosarcoma/pathology , Rats , Rats, Sprague-Dawley , S Phase/drug effects , Survival Rate , Tibia/diagnostic imaging , Tibia/pathology , Time Factors , Tomography, X-Ray Computed , Treatment Outcome , Zoledronic Acid
17.
Biochim Biophys Acta ; 1704(2): 49-57, 2004 Sep 20.
Article in English | MEDLINE | ID: mdl-15363860

ABSTRACT

The emergence of the molecular triad osteoprotegerin (OPG)/Receptor Activator of NF-kB (RANK)/RANK Ligand (RANKL) has helped elucidate a key signalling pathway between stromal cells and osteoclasts. The interaction between RANK and RANKL plays a critical role in promoting osteoclast differentiation and activation leading to bone resorption. OPG is a soluble decoy receptor for RANKL that blocks osteoclast formation by inhibiting RANKL binding to RANK. The OPG/RANK/RANKL system has been shown to be abnormally regulated in several malignant osteolytic pathologies such as multiple myeloma [MM, where enhanced RANKL expression (directly by tumour cells or indirectly by stromal bone cells or T-lymphocytes)] plays an important role in associated bone destruction. By contrast, production of its endogenous counteracting decoy receptor OPG is either inhibited or too low to compensate for the increase in RANKL production. Therefore, targeting the OPG/RANK/RANKL axis may offer a novel therapeutic approach to malignant osteolytic pathologies. In animal models, OPG or soluble RANK was shown both to control hypercalcaemia of malignancy and the establishment and progression of osteolytic metastases caused by various malignant tumours. To this day, only one phase I study has been performed using a recombinant OPG construct that suppressed bone resorption in patients with multiple myeloma or breast carcinoma with radiologically confirmed bone lesions. RANK-Fc also exhibits promising therapeutic effects, as revealed in animal models of prostate cancer and multiple myeloma. If the animal results translate to similar clinical benefits in humans, using RANK-Fc or OPG may yield novel and potent strategies for treating patients with established or imminent malignant bone diseases and where standard therapeutic regimens have failed.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Bone Neoplasms/physiopathology , Carrier Proteins/pharmacology , Glycoproteins/pharmacology , Membrane Glycoproteins/pharmacology , Multiple Myeloma/drug therapy , Multiple Myeloma/physiopathology , Bone Neoplasms/secondary , Cell Transformation, Neoplastic , Humans , Osteoprotegerin , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Receptors, Cytoplasmic and Nuclear , Receptors, Tumor Necrosis Factor , Signal Transduction
18.
Exp Cell Res ; 293(2): 292-301, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14729467

ABSTRACT

Receptor activator of NF-kB Ligand (RANKL) is an essential requirement for osteoclastogenesis and its activity is neutralized by binding to the soluble decoy receptor osteoprotegerin (OPG). The purpose of this work was to study the effects of RANKL and OPG during osteoclastogenesis using the murine monocytic cell line RAW 264.7 that can differentiate into osteoclasts in vitro. RAW 264.7 cells plated at 10(4) cells/cm(2) and cultured for 4 days in the presence of RANKL represent the optimal culture conditions for osteoclast differentiation, with an up-regulation of all parameters related to bone resorption: tartrate resistant acid phosphatase (TRAP), calcitonin receptor (CTR), RANK, cathepsin K, matrix metalloproteinase (MMP)-9 mRNA expressions. RANKL and OPG biological effects vary according to the differentiation state of the cells: in undifferentiated RAW 264.7 cells, TRAP expression was decreased by OPG and RANKL, RANK expression was inhibited by OPG, while MMP-9 and cathepsin K mRNA expressions were not modulated. In differentiated RAW 264.7 cells, RANKL and OPG both exert an overall inhibitory effect on the expression of all the parameters studied. In these experimental conditions, OPG-induced MMP-9 inhibition was abrogated in the presence of a blocking anti-RANKL antibody, suggesting that part of OPG effects are RANKL-dependent.


Subject(s)
Bone Remodeling/physiology , Carrier Proteins/metabolism , Cell Differentiation/physiology , Glycoproteins/metabolism , Membrane Glycoproteins/metabolism , Osteoclasts/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Stem Cells/metabolism , Acid Phosphatase/genetics , Animals , Antibodies/pharmacology , Biomarkers , Bone Remodeling/drug effects , Carrier Proteins/antagonists & inhibitors , Cathepsin K , Cathepsins/genetics , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Cell Line , Glycoproteins/genetics , Glycoproteins/pharmacology , Isoenzymes/genetics , Matrix Metalloproteinase 9/genetics , Membrane Glycoproteins/antagonists & inhibitors , Mice , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Osteoclasts/drug effects , Osteoprotegerin , RANK Ligand , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptor Activator of Nuclear Factor-kappa B , Receptors, Calcitonin/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Tumor Necrosis Factor , Reproducibility of Results , Stem Cells/drug effects , Tartrate-Resistant Acid Phosphatase
19.
Biochim Biophys Acta ; 1644(1): 1-7, 2004 Feb 02.
Article in English | MEDLINE | ID: mdl-14741739

ABSTRACT

Osteoprotegerin (OPG) is a decoy receptor for receptor activator of nuclear factor kappaB ligand (RANKL), an inducer of osteoclastogenesis via its receptor RANK. We recently demonstrated that OPG also exerts a direct effect in osteoclasts by regulating protease expression. Herein, we showed that OPG-induced pro-matrix metalloproteinase-9 activity was abolished by ras/MAPK inhibitors in purified osteoclasts. OPG induced the phosphorylation of p38 and ERK1/2 in RAW264.7 cells. Only p38 activation was totally abolished by a blocking anti-RANKL antibody or an excess of RANKL. Surface plasmon resonance experiments revealed that RANK, RANKL and OPG are able to form a tertiary complex. These results suggested a potential formation of a tertiary complex RANK-RANKL-OPG on osteoclasts. Thus, OPG is not only a soluble decoy receptor for RANKL but must be also considered as a direct effector of osteoclast functions.


Subject(s)
Glycoproteins/physiology , Osteoclasts/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/pharmacology , Cell Line , Cells, Cultured , Enzyme Activation , Glycoproteins/antagonists & inhibitors , Glycoproteins/pharmacology , Ligands , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase Inhibitors , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/pharmacology , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Models, Molecular , Osteoclasts/drug effects , Osteoclasts/enzymology , Osteoprotegerin , Phosphorylation , RANK Ligand , Rabbits , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Tumor Necrosis Factor , Signal Transduction/physiology , Surface Plasmon Resonance , Transfection , p38 Mitogen-Activated Protein Kinases
20.
Clin Orthop Relat Res ; (416): 278-84, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14646771

ABSTRACT

Light and electron microscopic investigations and studies of the resorption ability in vitro of giant cells were done in two patients with giant cell osteolytic lesions of metatarsal bone. Giant cells harvested from both patients were similar in morphologic features and ability to resorb dentin. After other diagnoses of osteolytic lesions of metatarsal bone were ruled out, one lesion was considered a giant cell reparative granuloma and the other a true giant cell tumor of bone. Clinical, radiologic, ultrastructural, functional studies, and data in the literature, indicated that giant cell reparative granuloma only can be differentiated from giant cell tumor by younger age at diagnosis and the occurrence of giant cell clusters. All other features (cortical erosion, high rate of recurrence, hemorrhage areas, predominant intercellular collagenous substance) are characteristic of both lesions. If these two giant cell lesions are different entities, more accurate means are needed to distinguish them.


Subject(s)
Bone Neoplasms/pathology , Giant Cell Tumors/pathology , Granuloma, Giant Cell/pathology , Metatarsal Bones , Adolescent , Adult , Diagnosis, Differential , Female , Humans , Microscopy, Electron
SELECTION OF CITATIONS
SEARCH DETAIL
...