Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 13(6): e0199517, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29924867

RESUMO

The receptor tyrosine kinase c-Met, its ligand HGF, and components of the downstream signalling pathway, have all been implicated in the pathogenesis of myeloma, both as modulators of plasma cell proliferation and as agents driving osteoclast differentiation and osteoblast inhibition thus, all these contribute substantially to the bone destruction typically caused by myeloma. Patients with elevated levels of HGF have a poor prognosis, therefore, targeting these entities in such patients may be of substantial benefit. We hypothesized that ARQ-197 (Tivantinib), a small molecule c-Met inhibitor, would reduce myeloma cell growth and prevent myeloma-associated bone disease in a murine model. In vitro we assessed the effects of ARQ-197 on myeloma cell proliferation, cytotoxicity and c-Met protein expression in human myeloma cell lines. In vivo we injected NOD/SCID-γ mice with PBS (non-tumour bearing) or JJN3 cells and treated them with either ARQ-197 or vehicle. In vitro exposure of JJN3, U266 or NCI-H929 cells to ARQ-197 resulted in a significant inhibition of cell proliferation and an induction of cell death by necrosis, probably caused by significantly reduced levels of phosphorylated c-Met. In vivo ARQ-197 treatment of JJN3 tumour-bearing mice resulted in a significant reduction in tumour burden, tumour cell proliferation, bone lesion number, trabecular bone loss and prevented significant decreases in the bone formation rate on the cortico-endosteal bone surface compared to the vehicle group. However, no significant differences on bone parameters were observed in non-tumour mice treated with ARQ-197 compared to vehicle, implying that in tumour-bearing mice the effects of ARQ-197 on bone cells was indirect. In summary, these res ults suggest that ARQ-197 could be a promising therapeutic in myeloma patients, leading to both a reduction in tumour burden and an inhibition of myeloma-induced bone disease.


Assuntos
Antineoplásicos/farmacologia , Doenças Ósseas/prevenção & controle , Mieloma Múltiplo/tratamento farmacológico , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Pirrolidinonas/farmacologia , Quinolinas/farmacologia , Animais , Doenças Ósseas/diagnóstico por imagem , Doenças Ósseas/metabolismo , Doenças Ósseas/patologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Feminino , Humanos , Camundongos Endogâmicos NOD , Camundongos SCID , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Transplante de Neoplasias , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteoclastos/patologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Distribuição Aleatória , Carga Tumoral/efeitos dos fármacos
2.
Biotechnol Appl Biochem ; 65(3): 407-418, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-28981962

RESUMO

Bone metastasis is a key event responsible for morbidity in prostate cancer patients. Interactions between prostate cancer cells and the bone microenvironment facilitate survival of tumor cells and alter bone turnover, a process that is thought to enhance the growth of metastases in this site. This study aimed to test the hypothesis that the presence of tumors cells increases transforming growth factor beta (TGF-ß) signaling in bone and that this regulates the proliferation and differentiation of osteoblastic lineage cells in metastatic sites. Initial studies showed that factors produced by prostate cancer cells increased the proliferation of osteoblastic cells and suppressed the early phase of their differentiation. We subsequently showed that interactions between prostate cancer and osteoblastic cells affected the expression of TGF-ß superfamily genes in the latter. Noggin was expressed and secreted by prostate cancer cells but expressed at very low levels by osteoblastic cells when these cells were grown alone. This pattern changed when osteoblasic cells were treated with conditioned medium derived from prostate cancer cells or were cocultured with the latter, with strong induction of Noggin being demonstrated. Immunohistochemical examination of prostate cancer xenografts showed strong Noggin protein staining on endosteal bone surfaces and in bone lining cells in close proximity to tumor foci. These studies support previous work that suggest Noggin is an important suppressor of the differentiation of osteoblast lineage cells in bone metastases. Importantly, we have now also shown that this protein can be induced in bone cells themselves by factors derived from prostate cancer cells.


Assuntos
Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Proteínas de Transporte/biossíntese , Osteoblastos/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Diferenciação Celular , Proliferação de Células , Humanos , Masculino , Fator de Crescimento Transformador beta1/metabolismo , Células Tumorais Cultivadas
3.
Br Med Bull ; 113(1): 15-30, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25662536

RESUMO

BACKGROUND: Despite advances in the treatment of multiple myeloma (MM), it remains an incurable malignant disease. Myeloma genetics is intrinsically complex, but it offers an opportunity to categorize the disease and apply a personalized medicine approach. AREAS OF AGREEMENT: Research into the genetics of myeloma is moving at a fast pace and is highlighting areas and patient cohorts likely to benefit from specific treatment. Targeting residual disease is likely to be crucial to improved clinical outcome. AREAS OF CONTROVERSY: Patients in clinical trials are more likely to receive genetic diagnosis than non-trial patients, for whom access is ad hoc and dependent upon regional commissioning arrangements. AREAS TIMELY FOR DEVELOPING RESEARCH: Relating genetics to potential treatment pathways will become crucial for improved myeloma outcomes. Universal access to standardized genetic testing will facilitate modern personalized treatments.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Testes Genéticos , Imunomodulação/genética , Mieloma Múltiplo/diagnóstico , Medicina de Precisão/métodos , Transplante de Células-Tronco/métodos , Testes Genéticos/métodos , Testes Genéticos/tendências , Humanos , Mieloma Múltiplo/genética , Mieloma Múltiplo/terapia , Medicina de Precisão/tendências , Prognóstico
4.
Br Med Bull ; 111(1): 117-38, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25190762

RESUMO

INTRODUCTION: Patients with myeloma develop localized and generalized bone loss leading to hypercalcaemia, accelerated osteoporosis, vertebral wedge fractures, other pathological fractures, spinal cord compression and bone pain. Bone loss is mediated by a variety of biological modifiers including osteoclast-activating factors (OAF) and osteoblast (OB) inhibitory factors produced either directly by malignant plasma cells (MPCs) or as a consequence of their interaction with the bone marrow microenvironment (BMM). Raised levels of OAFs such as receptor activator of nuclear factor-kappa B ligand (RANKL), macrophage inflammatory protein 1 alpha, tumour necrosis factor-alpha and interleukin 6 stimulate bone resorption by recruiting additional osteoclasts. Via opposing mechanisms, increases in OB inhibitory factors, such as dickkopf-1 (Dkk-1), soluble frizzled-related protein-3 and hepatocyte growth factor (HGF), suppress bone formation by inhibiting the differentiation and recruitment of OBs. These changes result in an uncoupling of physiological bone remodelling, leading to myeloma bone disease (MBD). Moreover, the altered BMM provides a fertile ground for the growth and survival of MPCs. Current clinical management of MBD is both reactive (to pain and fractures) and preventive, with bisphosphonates (BPs) being the mainstay of pharmacological treatment. However, side effects and uncertainties associated with BPs warrant the search for more targeted treatments for MBD. This review will summarize recent developments in understanding the intimate relationship between MBD and the BMM and the novel ways in which they are being therapeutically targeted. SOURCES OF DATA: All data included were sourced and referenced from PubMed. AREAS OF AGREEMENT: The clinical utility of BP therapy is well established. However, there is general acknowledgement that BPs are only partially successful in the treatment of MBD. The number of skeletal events attributable to myeloma are reduced by BPs but not totally eliminated. Furthermore, existing damage is not repaired. It is widely recognized that more effective treatments are needed. AREAS OF CONTROVERSY: There remains controversy concerning the duration of BP therapy. Whether denosumab is a viable alternative to BP therapy is also contested. Many of the new therapeutic strategies discussed are yet to translate to clinical practice and demonstrate equal efficacy or superiority to BP therapy. It also remains controversial whether reported anti-tumour effects of bone-modulating therapies are clinically significant. GROWING POINTS: The potential clinical utility of bone anabolic therapies including agents such as anti-Dkk-1, anti-sclerostin and anti-HGF is becoming increasingly recognized. AREAS TIMELY FOR DEVELOPING RESEARCH: Further research effectively targeting the mediators of MBD, targeting both bone resorption and bone formation, is urgently needed. This should translate promptly to clinical trials of combination therapy comprising anti-resorptives and bone anabolic therapies to demonstrate efficacy and improved outcomes over BPs.


Assuntos
Doenças Ósseas/etiologia , Mieloma Múltiplo/complicações , Conservadores da Densidade Óssea/uso terapêutico , Doenças Ósseas/terapia , Moléculas de Adesão Celular/fisiologia , Humanos , Fatores Imunológicos/uso terapêutico , Mieloma Múltiplo/terapia , Osteoblastos/fisiologia , Osteoclastos/fisiologia , Osteoporose/etiologia , Osteoporose/terapia
5.
PLoS One ; 7(8): e41127, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22952578

RESUMO

Patients with multiple myeloma commonly develop focal osteolytic bone disease, as well as generalised osteoporosis. The mechanisms underlying the development of osteoporosis in patients with myeloma are poorly understood. Although disruption of the RANKL/OPG pathway has been shown to underlie formation of focal osteolytic lesions, its role in the development of osteoporosis in myeloma remains unclear. Increased soluble RANKL in serum from patients with myeloma raises the possibility that this molecule plays a key role. The aim of the present study was to establish whether sRANKL produced by myeloma cells contributes directly to osteoporosis. C57BL/KaLwRij mice were injected with either 5T2MM or 5T33MM murine myeloma cells. 5T2MM-bearing mice developed osteolytic bone lesions (p<0.05) with increased osteoclast surface (p<0.01) and reduced trabecular bone volume (p<0.05). Bone volume was also reduced at sites where 5T2MM cells were not present (p<0.05). In 5T2MM-bearing mice soluble mRANKL was increased (p<0.05), whereas OPG was not altered. In contrast, 5T33MM-bearing mice had no changes in osteoclast surface or trabecular bone volume and did not develop osteolytic lesions. Soluble mRANKL was undetectable in serum from 5T33MM-bearing mice. In separate experiments, RPMI-8226 human myeloma cells were transduced with an human RANKL/eGFP construct, or eGFP alone. RPMI-8226/hRANKL/eGFP cells, but not RPMI-8226/eGFP cells, stimulated osteoclastic bone resorption (p<0.05) in vitro. Sub-cutaneous injection of NOD/SCID mice with RPMI-8226/hRANKL/eGFP or RPMI-8226/eGFP cells resulted in tumour development in all mice. RPMI-8226/hRANKL/eGFP-bearing mice exhibited increased serum soluble hRANKL (p<0.05) and a three-fold increase in osteoclast number (p<0.05) compared to RPMI-8226/eGFP-bearing mice. This was associated with reduced trabecular bone volume (27%, p<0.05), decreased trabecular number (29%, p<0.05) and increased trabecular thickness (8%, p<0.05). Our findings demonstrate that soluble RANKL produced by myeloma cells causes generalised bone loss, suggesting that targeting RANKL may prevent osteoporosis in patients with myeloma.


Assuntos
Osso e Ossos/metabolismo , Mieloma Múltiplo/metabolismo , Osteoclastos/citologia , Animais , Linhagem Celular Tumoral , DNA Complementar/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Vértebras Lombares/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Transplante de Neoplasias , Osteoporose/genética , Osteoporose/fisiopatologia , Osteoprotegerina/metabolismo , Ligante RANK/metabolismo
6.
J Bone Miner Res ; 24(3): 425-36, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19016584

RESUMO

Multiple myeloma (MM) is associated with the development of osteolytic bone disease, mediated by increased osteoclastic bone resorption and impaired osteoblastic bone formation. Dickkopf-1 (Dkk1), a soluble inhibitor of wingless/int (Wnt) signaling and osteoblastogenesis, is elevated in patients with MM and correlates with osteolytic bone disease. In this study, we investigated the effect of inhibiting Dkk1 on the development of osteolytic lesions in the 5T2MM murine model of myeloma. We showed that Dkk1 is expressed by murine 5T2MM myeloma cells. Injection of 5T2MM cells into C57BL/KaLwRij mice resulted in the development of osteolytic bone lesions (p < 0.05), mediated by increased osteoclast numbers (p < 0.001) and a decrease in osteoblast numbers (p < 0.001) and mineralizing surface (p < 0.05). Mice bearing 5T2MM cells were treated with an anti-Dkk1 antibody (BHQ880, 10 mg/kg, IV, twice weekly for 4 wk) from time of paraprotein detection. Anti-Dkk1 treatment prevented 5T2MM-induced suppression of osteoblast numbers (p < 0.001) and surface (p < 0.001). Treatment increased mineralizing surface by 28% and bone formation rate by 25%; however, there was no change in mineral apposition rate. Inhibiting Dkk1 had no effect on osteoclast numbers. muCT analysis showed that anti-Dkk1 treatment significantly protected against 5T2MM-induced trabecular bone loss (p < 0.05) and reduced the development of osteolytic bone lesions (p < 0.05). Treatment had no significant effect on tumor burden. These data suggest that inhibiting Dkk1 prevents the suppression of bone formation and in doing so is effective in preventing the development of osteolytic bone disease in myeloma, offering an effective therapeutic approach to treating this clinically important aspect of myeloma.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Mieloma Múltiplo/complicações , Mieloma Múltiplo/fisiopatologia , Osteogênese , Osteólise/complicações , Osteólise/prevenção & controle , Animais , Anticorpos/farmacologia , Contagem de Células , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Mieloma Múltiplo/patologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/patologia , Osteogênese/efeitos dos fármacos , Osteólise/patologia , Osteólise/fisiopatologia , Solubilidade/efeitos dos fármacos , Carga Tumoral/efeitos dos fármacos , Proteínas Wnt/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...