Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
J Bone Oncol ; 29: 100364, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34150488

ABSTRACT

More than 35% of lung adenocarcinoma patients have bone metastases at diagnosis and have a poor survival. Periostin, a carboxylated matrix protein, mediates lung cancer cell dissemination by promoting epithelial-mesenchymal transition, and is involved in bone response to mechanical stress and bone formation regulation. This suggests that periostin may be used as a biomarker to predict survival in lung cancer patients. Serum periostin was assessed at diagnosis in a prospective cohort of 133 patients with lung adenocarcinoma of all stages. Patients were divided into localized and bone metastatic groups. Both groups were matched to healthy controls. Survival analysis and Cox proportional hazards models were conducted in the total population and in bone metastatic group. The median serum periostin level was higher in bone metastatic (n = 67; median: 1752 pmol/L) than in the localized group (n = 66; 861 pmol/L; p < 0.0001). Patients with high periostin (>median) had a poorer overall survival in the whole population (33.3 weeks vs. NR; p < 0.0001) and the bone metastatic group (24.4 vs. 66.1 weeks; p < 0.001). In multivariate analysis, patients with high periostin had increased risk of death (HR = 2.09, 95%CI [1.06-4.13]; p = 0.03). This was also found in the bone metastatic group (HR = 3.62, 95%CI [1.74-7.52]; p = 0.0005). Immunohistochemistry on bone metastasis biopsies showed periostin expression in the bone matrix and nuclear and cytoplasmic staining in cancer cells. Serum periostin was an independent survival biomarker in all-stage and in bone metastatic lung adenocarcinoma patients. IHC data suggest that periostin might be induced in cancer cells in bone metastatic niche in addition to bone microenvironment expression.

2.
Clin Transl Oncol ; 21(8): 977-991, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30656607

ABSTRACT

Drug repurposing offers advantages over traditional drug development in terms of cost, speed and improved patient outcomes. The receptor activator of nuclear factor kappa B (RANK) ligand (RANKL) inhibitor denosumab is approved for the prevention of skeletal-related events in patients with advanced malignancies involving bone, including solid tumours and multiple myeloma. Following improved understanding of the role of RANK/RANKL in cancer biology, denosumab has already been repurposed as a treatment for giant cell tumour of bone. Here, we review the role of RANK/RANKL in tumourigenesis, including effects on tumour initiation, progression and metastasis and consider the impact of RANK/RANKL on tumour immunology and immune evasion. Finally, we look briefly at ongoing trials and future opportunities for therapeutic synergy when combining denosumab with anti-cancer agents such as immune checkpoint inhibitors.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Repositioning , Neoplasms/drug therapy , Neoplasms/metabolism , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Humans , Prognosis
3.
Oncogene ; 38(7): 950-964, 2019 02.
Article in English | MEDLINE | ID: mdl-30478447

ABSTRACT

Bone is the most common metastatic site for breast cancer. Estrogen-related-receptor alpha (ERRα) has been implicated in cancer cell invasiveness. Here, we established that ERRα promotes spontaneous metastatic dissemination of breast cancer cells from primary mammary tumors to the skeleton. We carried out cohort studies, pharmacological inhibition, gain-of-function analyses in vivo and cellular and molecular studies in vitro to identify new biomarkers in breast cancer metastases. Meta-analysis of human primary breast tumors revealed that high ERRα expression levels were associated with bone but not lung metastases. ERRα expression was also detected in circulating tumor cells from metastatic breast cancer patients. ERRα overexpression in murine 4T1 breast cancer cells promoted spontaneous bone micro-metastases formation when tumor cells were inoculated orthotopically, whereas lung metastases occurred irrespective of ERRα expression level. In vivo, Rank was identified as a target for ERRα. That was confirmed in vitro in Rankl stimulated tumor cell invasion, in mTOR/pS6K phosphorylation, by transactivation assay, ChIP and bioinformatics analyses. Moreover, pharmacological inhibition of ERRα reduced primary tumor growth, bone micro-metastases formation and Rank expression in vitro and in vivo. Transcriptomic studies and meta-analysis confirmed a positive association between metastases and ERRα/RANK in breast cancer patients and also revealed a positive correlation between ERRα and BRCA1mut carriers. Taken together, our results reveal a novel ERRα/RANK axis by which ERRα in primary breast cancer promotes early dissemination of cancer cells to bone. These findings suggest that ERRα may be a useful therapeutic target to prevent bone metastases.


Subject(s)
Bone Neoplasms/metabolism , Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Animal/metabolism , Neoplasm Proteins/metabolism , Receptor Activator of Nuclear Factor-kappa B/biosynthesis , Receptors, Estrogen/metabolism , Animals , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Humans , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice , Mice, Inbred BALB C , Neoplasm Proteins/genetics , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptors, Estrogen/genetics , ERRalpha Estrogen-Related Receptor
5.
Ann Oncol ; 27(3): 379-90, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26681681

ABSTRACT

Bisphosphonates have been studied in randomised trials in early breast cancer to investigate their ability to prevent cancer treatment-induced bone loss (CTIBL) and reduce the risk of disease recurrence and metastasis. Treatment benefits have been reported but bisphosphonates do not currently have regulatory approval for either of these potential indications. This consensus paper provides a review of the evidence and offers guidance to breast cancer clinicians on the use of bisphosphonates in early breast cancer. Using the nominal group methodology for consensus, a systematic review of the literature was augmented by a workshop held in October 2014 for breast cancer and bone specialists to present and debate the available pre-clinical and clinical evidence for the use of adjuvant bisphosphonates. This was followed by a questionnaire to all members of the writing committee to identify areas of consensus. The panel recommended that bisphosphonates should be considered as part of routine clinical practice for the prevention of CTIBL in all patients with a T score of <-2.0 or ≥2 clinical risk factors for fracture. Compelling evidence from a meta-analysis of trial data of >18,000 patients supports clinically significant benefits of bisphosphonates on the development of bone metastases and breast cancer mortality in post-menopausal women or those receiving ovarian suppression therapy. Therefore, the panel recommends that bisphosphonates (either intravenous zoledronic acid or oral clodronate) are considered as part of the adjuvant breast cancer treatment in this population and the potential benefits and risks discussed with relevant patients.


Subject(s)
Antineoplastic Agents/adverse effects , Bone Density Conservation Agents/therapeutic use , Bone Neoplasms/prevention & control , Breast Neoplasms/drug therapy , Diphosphonates/therapeutic use , Neoplasm Recurrence, Local/prevention & control , Osteoporosis/prevention & control , Antineoplastic Agents/therapeutic use , Bone Neoplasms/secondary , Chemotherapy, Adjuvant , Clodronic Acid/adverse effects , Clodronic Acid/therapeutic use , Consensus , Diphosphonates/adverse effects , Europe , Female , Humans , Imidazoles/adverse effects , Imidazoles/therapeutic use , Neoplasm Recurrence, Local/drug therapy , Osteoporosis/chemically induced , Osteoporosis/drug therapy , Surveys and Questionnaires , Zoledronic Acid
6.
Cancer Lett ; 357(1): 279-285, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25444923

ABSTRACT

Zoledronate (ZOL) inhibits farnesyl pyrophosphate synthase leading to intracellular accumulation of isopentenyl pyrophosphate/triphosphoric acid 1-adenosin-5'-yl ester 3-(3-methylbut-3-enyl) ester (IPP/ApppI). Cytotoxic Vγ9Vδ2 T cells have been shown to recognize IPP/ApppI in breast cancer cells. Further, human breast cancer cells have been shown to differ remarkably in their ZOL treatment induced IPP/ApppI production and responses to that. In this communication we analysed the responsiveness of prostate cancer cells PC-3 and DU-145, Caki-2 renal carcinoma cells and U87MG glioblastoma cells to ZOL treatment, and the subsequent activation of Vγ9Vδ2 T-cell cytotoxicity. Of the cell lines tested, PC-3 cells were not susceptible to Vγ9Vδ2 T-cell cytotoxicity due to low activity of the mevalonate pathway and low amount of IPP formed. However, the resistance of PC-3 cells to Vγ9Vδ2 T-cell cytotoxicity could be abrogated by upregulation of the mevalonate pathway through cholesterol depletion.


Subject(s)
Cholesterol/deficiency , Diphosphonates/pharmacology , Imidazoles/pharmacology , Mevalonic Acid/metabolism , Prostatic Neoplasms/drug therapy , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Cell Line, Tumor , Cholesterol/metabolism , Cytotoxicity, Immunologic/drug effects , Humans , Male , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects , Zoledronic Acid
7.
Oncogene ; 32(6): 724-35, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-22430214

ABSTRACT

Little is known about metastatic pathways that are specific to the lung rather than other organs. We previously showed that antioxidant proteins such as peroxiredoxins were specifically upregulated in lung metastatic breast cancer cells. We hypothesize that cancer cells that live under aerobic conditions, as might be the case in lungs, protect themselves against the damage caused by reactive oxygen species (ROS). To examine this hypothesis, we studied the role of peroxiredoxin-2 (PRDX2) in lung vs bone metastasis formation. A metastatic variant of MDA-MB-435 breast cancer cells that specifically metastasize to lungs (435-L3) was transduced with short hairpin RNAs to specifically silence PRDX2. Conversely, a bone metastatic variant of MDA-MB-231 cells (BO2) was stably transfected to overexpress PRDX2. The 435-L3 cells silenced for PRDX2 were significantly more sensitive to H(2)O(2)-induced oxidative stress than the parental and scrambled transfected cells. BO2/PRDX2 cells produced less ROS than BO2/green fluorescent protein control cells under oxidative stress. Moreover, PRDX2 knockdown inhibited the growth of 435-L3 cells in the lungs, whereas lymph node metastasis remained unaffected. In contrast, PRDX2 overexpression in bone metastatic BO2 breast cancer cells led to drastic inhibition of the skeletal tumor burden and reduction of bone destruction. Furthermore, PRDX2 expression in breast cancer cells was associated with a glucose-dependent phenotype, different from bone metastatic cells. Overall, our results strongly suggest that PRDX2 is a targetable 'metabolic adaptor' driver protein implicated in the selective growth of metastatic cells in the lungs by protecting them against oxidative stress.


Subject(s)
Breast Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Oxidative Stress/genetics , Peroxiredoxins/physiology , Stress, Physiological/genetics , Bone Neoplasms/secondary , Breast Neoplasms/genetics , Cell Line, Tumor , Female , Humans , Hydrogen Peroxide/metabolism , Lung Neoplasms/genetics , Lymphatic Metastasis , Transfection
8.
Cancer Radiother ; 16(5-6): 330-8, 2012 Sep.
Article in French | MEDLINE | ID: mdl-22921977

ABSTRACT

Solitary metastases have been reported in up to 30% of cases in imaging series. Local treatment aims at consolidating the injured bone and to prevent neurologic complications. Since the prognosis of bony metastatic disease is about 30 months and includes some long survivors, the multisdisciplinary committee in charge of the patient should ask the question and decide on the type of radical/ablative intervention in case of oligometastases. A literature search was performed using MESH terms (bone, metastases, radiotherapy, radiology, cement, radiofrequency ablation, chemoembolisation). Local ablative treatments can yield symptomatic relief and local control rates of about 90%. Stereotactic hypofractionated irradiation and cementoplasty are increasingly used. In conclusion, local ablative treatment of bony oligometastases is an efficient treatment. Its potential impact on survival remains to be demonstrated prospectively in clinical trials.


Subject(s)
Bone Neoplasms/secondary , Bone Neoplasms/therapy , Ablation Techniques , Bone Neoplasms/mortality , Cementoplasty , Chemoembolization, Therapeutic , Dose Fractionation, Radiation , Humans , Radiosurgery
9.
Br J Cancer ; 107(1): 63-70, 2012 Jun 26.
Article in English | MEDLINE | ID: mdl-22627320

ABSTRACT

BACKGROUND: The small stress heat shock protein 27 (Hsp27) has recently turned as a promising target for cancer treatment. Hsp27 upregulation is associated with tumour growth and resistance to chemo- and radio-therapeutic treatments, and several ongoing drugs inhibiting Hsp27 expression are under clinical trial. Hsp27 is now well described to counteract apoptosis and its elevated expression is associated with increased aggressiveness of several primary tumours. However, its role in the later stage of tumour progression and, more specifically, in the later and most deadly stage of tumour metastasis is still unclear. METHODS/RESULTS: In the present study, we showed by qRT-PCR that Hsp27 gene is overexpressed in a large fraction of the metastatic breast cancer area in 53 patients. We further analysed the role of this protein in mice during bone metastasis invasion and establishment by using Hsp27 genetically depleted MDA-MB231/B02 human breast cancer cell line as a model. We demonstrate that Hsp27 silencing led to reduced cell migration and invasion in vitro and that in vivo it correlated with a decreased ability of breast cancer cells to metastasise and grow in the skeleton. CONCLUSION: Altogether, these data characterised Hsp27 as a potent therapeutic target in breast cancer bone metastasis and skeletal tumour growth.


Subject(s)
Bone Neoplasms/prevention & control , Breast Neoplasms/genetics , HSP27 Heat-Shock Proteins/genetics , Animals , Apoptosis/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Female , Gene Targeting , Humans , Mice , Mice, Nude , Neoplasm Transplantation , RNA, Small Interfering/pharmacology , Transfection , Transplantation, Heterologous
10.
Curr Pharm Des ; 16(27): 3007-2014, 2010.
Article in English | MEDLINE | ID: mdl-20722623

ABSTRACT

Bisphosphonates, especially nitrogen-containing bisphosphonates (N-BPs), are widely used to block bone destruction in cancer patients with bone metastasis because they are effective inhibitors of osteoclast-mediated bone resorption. In addition to their antiresorptive effects, preclinical evidence strongly suggests that N-BPs have anticancer activity. Some of the activities associated with N-BPs are observed in human γδT cells that straddle the interface of innate and adaptive immunity and have potent anti-tumour activity. This review examines the molecular and cellular mechanisms through which N-BPs stimulate the expansion and cytotoxic activity of human γδ T cells. In addition, we discuss the emerging clinical evidence that N-BPs have a role in cancer immunotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Diphosphonates/therapeutic use , Immunotherapy, Adoptive/methods , Lymphocyte Activation/drug effects , Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Adaptive Immunity/drug effects , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Bone Density Conservation Agents/chemistry , Bone Density Conservation Agents/pharmacology , Bone Density Conservation Agents/therapeutic use , Bone Neoplasms/drug therapy , Bone Neoplasms/immunology , Bone Neoplasms/secondary , Bone Resorption/drug therapy , Bone Resorption/prevention & control , Diphosphonates/chemistry , Diphosphonates/pharmacology , Humans , Neoplasms/immunology , Nitrogen/chemistry , T-Lymphocytes, Cytotoxic/drug effects
11.
Bull Cancer ; 97(8): 937-49, 2010 Aug.
Article in French | MEDLINE | ID: mdl-20595092

ABSTRACT

Zoledronate, just as other bisphosphonates, inhibit osteoclast mediated bone resorption. This is the reason why they are used in the treatment of bone metastasis, in order to block osteolysis. Zoledronate and some other bisphosphonates (clodronate, pamidronate, ibandronate, alendronate, risédronate, minodronate) also exhibit antitumor properties in vitro. They act directly on tumor cells by blocking tumor cell adhesion, invasion and proliferation, and by inducing tumor cell apoptosis. However, their high bone mineral affinity decreases their bioavailability to a significant extent and, thus, should weaken their in vivo antitumor potential. Despite of this, several studies (most of them being performed with zoledronate) show that bisphosphonates have an in vivo antitumor activity. This review focuses on zoledronate and on results obtained in several experimental models showing that this bisphosphonate interferes with the growth of tumors and metastases which are thriving in tissues others than the skeletal tissue. The significance of these findings is discussed in the light of several ongoing clinical trials which examine the benefits of using zoledronate and other bisphosphonates in the adjuvant treatment of cancers at an early stage of the disease.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Density Conservation Agents/therapeutic use , Diphosphonates/therapeutic use , Imidazoles/therapeutic use , Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Bone Density Conservation Agents/pharmacokinetics , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Diphosphonates/pharmacokinetics , Humans , Imidazoles/pharmacokinetics , Neoplasm Metastasis/drug therapy , Neoplasms/metabolism , Zoledronic Acid
12.
Curr Cancer Drug Targets ; 9(7): 801-6, 2009 Nov.
Article in English | MEDLINE | ID: mdl-20025568

ABSTRACT

Integrins constitute a family of cell surface receptors that are heterodimers composed of noncovalently associated alpha and beta subunits. Integrins bind to extracellular matrix proteins and immunogobulin superfamily molecules. They exert a stringent control on cell migration, survival and proliferation. However, their expression and functions are often deregulated in cancer, and many lines of evidence implicate them as key regulators during progression from primary tumor growth to metastasis. Here, we review the role of integrins in bone metastasis formation and present evidence that the use of integrin-targeted therapeutic agents may be an efficient strategy to block tumor metastasis.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Integrins/physiology , Bone Neoplasms/physiopathology , Drug Delivery Systems , Humans , Integrins/drug effects , Models, Biological , Neoplasm Invasiveness/physiopathology , Protein Structure, Quaternary
13.
Br J Cancer ; 97(7): 964-70, 2007 Oct 08.
Article in English | MEDLINE | ID: mdl-17876334

ABSTRACT

The aim of this study was to determine whether Dickkopf-1 (Dkk-1) expression in breast cancer was associated with bone metastases. We first analysed Dkk-1 expression by human breast cancer cell lines that induce osteolytic or osteoblastic lesions in animals. Dickkopf-1 levels were then measured in the bone marrow aspirates of hind limbs from eight NMRI mice inoculated with breast cancer cells that induced bone metastases and 11 age-matched non-inoculated control animals. Finally, Dkk-1 was measured in the serum of 17 women with breast cancer in complete remission, 19 women with breast cancer and bone metastases, 16 women with breast cancer and metastases at non-bone sites and 16 healthy women. Only breast cancer cells that induce osteolytic lesions in animals produced Dkk-1. There was a six-fold increase in Dkk-1 levels in the bone marrow from animals inoculated with MDA-B02 cells when compared with that of control non-inoculated animals (P=0.003). Median Dkk-1 levels in the serum of patients with breast cancer and bone metastases were significantly higher than levels of patients in complete remission (P=0.016), patients with breast cancer having metastases at non-bone sites (P<0.0001) and healthy women (P=0.047), although there was a large overlap in individual levels between the different groups. In conclusion, Dkk-1 is secreted by osteolytic human breast cancer cells lines and increased circulating levels are associated with the presence of bone metastases in patients with breast cancer. Measurements of circulating Dkk-1 levels may be useful for the clinical investigation of patients with breast cancer and bone metastases.


Subject(s)
Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Breast Neoplasms/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Animals , Blotting, Western , Bone Resorption , Breast Neoplasms/pathology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Nude , Osteolysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
14.
Breast Cancer Res Treat ; 101(2): 135-48, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17028989

ABSTRACT

Metastatic breast cancer cells exhibit the selective ability to seed and grow in the skeleton. We and others have previously reported that human breast tumors which metastasize to the skeleton overexpress bone matrix extracellular proteins. In an attempt to reveal the osteoblast-like phenotype of osteotropic breast cancer cells, we performed a microarray study on a model of breast cancer bone metastasis consisting of the MDA-MB-231 human cell line and its variant B02 selected for its high capacity to form bone metastases in vivo. Analysis of B02 cells transcriptional profile revealed that 11 and 9 out of the 50 most up- and down-regulated mRNAs, respectively, corresponded to genes which expression has been previously associated with osteoblastic differentiation process. Thus, osteoblast specific cadherin 11 which mediates the differentiation of mesenchymal cells into osteoblastic cells is up-regulated in B02. While S100A4, recently described as a key negative regulator of osteoblast differentiation, is the most down-regulated gene in B02 cells. RT-PCR and western blotting experiments allowed the validation of the modulation of several genes of interest. Using immunohistochemistry, performed on human breast primary tumors and their matched liver and bone metastases, we were able to confirm that the osteoblast-like pattern of gene expression observed in our model holds true in vivo. This is the first report demonstrating a gene-expression pattern corresponding to the acquisition of an osteomimetic phenotype by bone metastatic breast cancer cells.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Neoplasm Metastasis/genetics , Blotting, Western , Cell Line, Tumor , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Oligonucleotide Array Sequence Analysis , Osteoblasts/physiology , Phenotype , Reverse Transcriptase Polymerase Chain Reaction
15.
Cell Tissue Res ; 323(3): 457-63, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16341551

ABSTRACT

Integrin alphabeta heterodimers mediate adhesion to the extracellular matrix and at cell-cell contacts and initiate intracellular signalling cascades in response to a variety of inductive factors. Apart from the expression of alphavbeta3 that we have previously reported, little is known about the expression of integrins in odontoblasts. Here, we investigated the expression of alphav-binding beta integrin subunits in healthy human dental pulp in vivo and in odontoblasts differentiated in vitro. Reverse transcription/polymerase chain reaction analysis revealed the expression of alphav, beta1, beta5 and beta8 integrin mRNA, but not beta6, in whole pulp cells. Flow cytometry showed that the alphav and beta1 subunits were the most intensely expressed. Immunohistochemistry demonstrated that the beta1 subunit was localised in newly differentiated odontoblasts in the root and in mature odontoblasts in the crown, including their intradentinal cell processes. The alphav chain was predominantly expressed by mature odontoblasts and alphavbeta5 was only observed in mature odontoblasts. In vitro differentiated odontoblasts expressed genes for alphav, beta1 and beta5, but not for beta6 and beta8. A comparison of integrin profiles between cultured pulp cells and in vitro differentiated odontoblasts revealed that odontoblast maturation was characterised by a significant increase in the expression of alphav and beta1 subunits and alphavbeta5 integrin. The beta8 subunit was detected in nerve cells only. Histological analysis of teeth from alphav knockout mice showed no obvious structural modification in the odontoblast layer. Thus, human mature odontoblasts express alphavbeta3, alphavbeta5 and perhaps alphavbeta1 integrins, with the possible presence of alpha-beta1 pairs. The roles that these molecules play in the exchange of information throughout the odontoblast layer remain to be determined.


Subject(s)
Dental Pulp/cytology , Integrin alphaV/metabolism , Odontoblasts/cytology , Adolescent , Animals , Cell Differentiation , Cells, Cultured , Dental Pulp/metabolism , Humans , Immunohistochemistry , Mice , Mice, Knockout , Odontoblasts/metabolism , Reverse Transcriptase Polymerase Chain Reaction
16.
J Bone Miner Res ; 16(11): 2027-34, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11697798

ABSTRACT

A very common metastatic site for human breast cancer is bone. The traditional bone metastasis model requires human MDA-MB-231 breast carcinoma cell inoculation into the left heart ventricle of nude mice. MDA-MB-231 cells usually develop osteolytic lesions 3-4 weeks after intracardiac inoculation in these animals. Here, we report a new approach to study the formation of bone metastasis in animals using breast carcinoma cells expressing the bioluminescent jellyfish protein (green fluorescent protein [GFP]). We first established a subclone of MDA-MB-231 cells by repeated in vivo passages in bone using the heart injection model. On stable transfection of this subclone with an expression vector for GFP and subsequent inoculation of GFP-expressing tumor cells (B02/GFP.2) in the mouse tail vein, B02/GFP.2 cells displayed a unique predilection for dissemination to bone. Externally fluorescence imaging of live animals allowed the detection of fluorescent bone metastases approximately 1 week before the occurrence of radiologically distinctive osteolytic lesions. The number, size, and intensity of fluorescent bone metastases increased progressively with time and was indicative of breast cancer cell progression within bone. Histological examination of fluorescent long bones from B02/GFP.2-bearing mice revealed the occurrence of profound bone destruction. Treatment of B02/GFP.2-bearing mice with the bisphosphonate zoledronic acid markedly inhibited the progression of established osteolytic lesions and the expansion of breast cancer cells within bone. Overall, this new bone metastasis model of breast cancer combining both fluorescence imaging and radiography should provide an invaluable tool to study the effectiveness of pharmaceutical agents that could suppress cancer colonization in bone.


Subject(s)
Bone Neoplasms/diagnosis , Bone Neoplasms/secondary , Breast Neoplasms , Animals , Bone Neoplasms/drug therapy , Breast Neoplasms/genetics , Diphosphonates/therapeutic use , Female , Green Fluorescent Proteins , Humans , Imidazoles/therapeutic use , Luminescent Proteins/genetics , Mice , Mice, Nude , Microscopy, Fluorescence , Osteolysis/drug therapy , Osteolysis/etiology , Recombinant Proteins/genetics , Tumor Cells, Cultured , Zoledronic Acid
17.
Genes Dev ; 15(11): 1373-82, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11390357

ABSTRACT

Thrombospondin 1 (TSP1) is a multifunctional protein able to activate TGFbeta and to inhibit angiogenesis in vivo. Although usually thought of as an inhibitor of tumor growth, TSP1 may sometimes be present at high levels during tumor progression, suggesting that tumors can eventually overcome their anti-tumor effects. Using a tet-repressible expression system, we demonstrate that murine TSP1 delayed the onset of tumor growth when produced in the tumor bed by rat fibrosarcoma tumor cells or by stromal fibroblasts coinjected with unmodified C6 glioma tumor cells. Yet upon prolonged exposure to TSP1, tumors came to grow at the same rate in the presence as in the absence of TSP1 and transplantation experiments showed that they had become insensitive to inhibition by TSP1 in both syngeneic and immune compromised hosts. Tumor resistance to TSP1 developed as a result of the in vivo outgrowth of pre-existing tumor cell variants that (1) secreted increased amounts of angiogenic factors that counterbalanced the inhibitory effect of TSP1 on neovascularization and (2) grew more efficiently in the presence of TSP1-activated TGFbeta. These results indicate that prolonged and continuous local delivery of a single multifunctional angiogenesis inhibitor like TSP1 to fast-growing tumors can lead to tumor resistance in vivo by fostering the outgrowth of subpopulations that are a by-product of the genetic instability of the tumor cells themselves.


Subject(s)
Angiogenesis Inhibitors/metabolism , Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Thrombospondin 1/metabolism , Transforming Growth Factor beta/metabolism , Animals , Blotting, Northern , Fibrosarcoma/blood supply , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Glioblastoma/blood supply , Glioblastoma/metabolism , Glioblastoma/pathology , Immunoblotting , Immunohistochemistry , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms/blood supply , Neoplasms/pathology , Rats , Rats, Inbred F344 , Tumor Cells, Cultured
18.
Cancer Res ; 60(11): 2949-54, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10850442

ABSTRACT

The molecular mechanisms by which tumor cells metastasize to bone are likely to involve invasion, cell adhesion to bone, and the release of soluble mediators from tumor cells that stimulate osteoclast-mediated bone resorption. Bisphosphonates (BPs) are powerful inhibitors of the osteoclast activity and are, therefore, used in the treatment of patients with osteolytic metastases. However, an added beneficial effect of BPs may be direct antitumor activity. We previously reported that BPs inhibit breast and prostate carcinoma cell adhesion to bone (Boissier et al., Cancer Res., 57: 3890-3894, 1997). Here, we provided evidence that BP pretreatment of breast and prostate carcinoma cells inhibited tumor cell invasion in a dose-dependent manner. The order of potency for four BPs in inhibiting tumor cell invasion was: zoledronate > ibandronate > NE-10244 (active pyridinium analogue of risedronate) > clodronate. In addition, NE-58051 (the inactive pyridylpropylidene analogue of risedronate) had no inhibitory effect, whereas NE-10790 (a phosphonocarboxylate analogue of risedronate in which one of the phosphonate groups is substituted by a carboxyl group) inhibited tumor cell invasion to an extent similar to that observed with NE-10244, indicating that the inhibitory activity of BPs on tumor cells involved the R2 chain of the molecule. BPs did not induce apoptosis in tumor cells, nor did they inhibit tumor cell migration at concentrations that did inhibit tumor cell invasion. However, although BPs did not interfere with the production of matrix metalloproteinases (MMPs) by tumor cells, they inhibited their proteolytic activity. The inhibitory effect of BPs on MMP activity was completely reversed in the presence of an excess of zinc. In addition, NE-10790 did not inhibit MMP activity, suggesting that phosphonate groups of BPs are responsible for the chelation of zinc and the subsequent inhibition of MMP activity. In conclusion, our results provide evidence for a direct cellular effect of BPs in preventing tumor cell invasion and an inhibitory effect of BPs on the proteolytic activity of MMPs through zinc chelation. These results suggest, therefore, that BPs may be useful agents for the prophylactic treatment of patients with cancers that are known to preferentially metastasize to bone.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/drug therapy , Diphosphonates/pharmacology , Prostatic Neoplasms/drug therapy , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Cell Adhesion , Cell Cycle/drug effects , Cell Movement , Clodronic Acid/pharmacology , Collagen/metabolism , Dose-Response Relationship, Drug , Drug Combinations , Flow Cytometry , Fluorometry , Humans , Ibandronic Acid , Imidazoles/pharmacology , Laminin/metabolism , Male , Matrix Metalloproteinases/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Paclitaxel/pharmacology , Proteoglycans/metabolism , Pyridines/pharmacology , Pyridinium Compounds/pharmacology , Tumor Cells, Cultured , Zinc/metabolism , Zoledronic Acid
19.
Joint Bone Spine ; 67(1): 22-9, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10773965

ABSTRACT

Malignant osteolysis is a common complication of many cancers, most notably breast cancer, prostate cancer, and multiple myeloma. Hypercalcemia, pain, and fractures are the main manifestations. Malignant osteolysis can be fatal or cause a rapid deterioration in quality of life. The underlying mechanism in tumor cem-mediated activation of osteoclasts, whose function is normally to resorb bone. It follows that pharmacological agents capable of inhibiting osteoclast activity, including bisphosphonates, are likely to be useful in the treatment of malignant osteolysis. Also, experimental evidence suggest that bisphosphonates act on the tumor cells themselves, either by inhibiting mechanisms involved in the development of bone metastasis (tumor invasion, adhesion of tumor cells to the bone matrix) or by inducing apoptosis of tumor cells. Many clinical trials have found bisphophonates to be effective in the treatment of complications due to malignant osteolysis. Based on these studies, bisphosphonates are now indicated to treat hypercalcemia and to prevent skeletal complications of metastatic breast cancer and myeloma, in a dosage of 1600 mg.d orally for clodronate or 90 mg every four weeks intravenously for pamidronate. Osteoclast inhibition is clearly the mechanism underlying the efficacy of bisphosphonates in these clinical trials. Recent clinical trials found that prophylactic bisphosphonates therapy in patients with nonmetastasic breast cancer decreased the incidence of bone metastases, thus supporting a direct effect of biphosphonates on tumor cells. However, conflicting experimental and clinical data have been reported, so that it remains uncertain whether bisphosphonates have anti-tumor effects in vivo in humans. Nevertheless, biphosphonates now have an undisputed place in the therapeutic armamentarium for cancer.


Subject(s)
Bone Neoplasms/drug therapy , Diphosphonates/pharmacology , Diphosphonates/therapeutic use , Multiple Myeloma/drug therapy , Osteoclasts/drug effects , Osteolysis/drug therapy , Animals , Bone Neoplasms/complications , Bone Neoplasms/secondary , Humans , Multiple Myeloma/complications , Osteolysis/etiology , Tumor Cells, Cultured/drug effects
20.
Presse Med ; 29(9): 487-91, 2000 Mar 11.
Article in French | MEDLINE | ID: mdl-10745942

ABSTRACT

MECHANISMS OF BONE LOSS: In patients with bone metastases, bone loss is the consequence of a dissociated process combining excessive bone resorption and inhibited bone formation. This destructive process occurs in response to soluble factors secreted by metastatic cells (PTH-rP, cytokines) which activate osteoclasts. These cells also secrete proteases (cathepsin K, metalloprotease MMP-9) which degrade the bone's collagen network. Excessive bone resorption is also favored by direct interaction between the metastatic cells and stromal cells in the bone marrow in response to the activation of membrane receptors (integrins a4 beta 1 and a4 beta 7). Finally, metastatic cells secrete non-identified soluble factors capable of inhibiting osteoblast proliferation in vitro. EFFECT OF BONE LOSS ON THE METASTASIS: Bone is a major reservoir of growth factors (mainly TGF beta and IGF-1). Positive feedback mechanism operates at the site of osteolysis where TGF beta released by bone tissue induces a paracrine stimulation of PTH-rP production by metastatic cells. IGF-1 released by the bone favors grow of metastatic cells present in the marrow. In addition, IGF-1 as well as collagen proteolytic fragments may stimulate recruitment of new metastatic cells at the site of the bone metastasis. This creates a vicious circle of mutual stimulation between bone destruction and tumor proliferation.


Subject(s)
Bone Neoplasms/physiopathology , Bone Neoplasms/secondary , Bone Resorption , Animals , Bone Marrow/pathology , Bone Matrix/pathology , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Bone Resorption/pathology , Bone Resorption/physiopathology , Bone Resorption/prevention & control , Breast Neoplasms/pathology , Cytokines/physiology , Diphosphonates/therapeutic use , Female , Humans , Mammary Neoplasms, Animal/pathology , Melanoma/pathology , Mice , Multiple Myeloma/pathology , Neoplasm Proteins/physiology , Osteoblasts/pathology , Osteoclasts/pathology , Osteolysis/pathology , Osteolysis/physiopathology , Parathyroid Hormone/physiology , Parathyroid Hormone-Related Protein , Proteins/physiology , Stromal Cells/pathology , Tumor Cells, Cultured/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...