Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
J Bone Oncol ; 29: 100375, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34131559

RESUMO

Optimum management of patients with cancer during the COVID-19 pandemic has proved extremely challenging. Patients, clinicians and hospital authorities have had to balance the risks to patients of attending hospital, many of whom are especially vulnerable, with the risks of delaying or modifying cancer treatment. Those whose care has been significantly impacted include patients suffering from the effects of cancer on bone, where delivering the usual standard of care for bone support has often not been possible and clinicians have been forced to seek alternative options for adequate management. At a virtual meeting of the Cancer and Bone Society in July 2020, an expert group shared experiences and solutions to this challenge, following which a questionnaire was sent internationally to the symposium's participants, to explore the issues faced and solutions offered. 70 respondents, from 9 countries (majority USA, 39%, followed by UK, 19%) included 50 clinicians, spread across a diverse range of specialties (but with a high proportion, 64%, of medical oncologists) and 20 who classified themselves as non-clinical (solely lab-based). Spread of clinician specialty across tumour types was breast (65%), prostate (27%), followed by renal, myeloma and melanoma. Analysis showed that management of metastatic bone disease in all solid tumour types and myeloma, adjuvant bisphosphonate breast cancer therapy and cancer treatment induced bone loss, was substantially impacted. Respondents reported delays to routine CT scans (58%), standard bone scans (48%) and MRI scans (46%), though emergency scans were less affected. Delays in palliative radiotherapy for bone pain were reported by 31% of respondents with treatments often involving only a single dose without fractionation. Delays to, or cancellation of, prophylactic surgery for bone pain were reported by 35% of respondents. Access to treatments with intravenous bisphosphonates and subcutaneous denosumab was a major problem, mitigated by provision of drug administration at home or in a local clinic, reduced frequency of administration or switching to oral bisphosphonates taken at home. The questionnaire also revealed damaging delays or complete stopping of both clinical and laboratory research. In addition to an analysis of the questionnaire, this paper presents a rationale and recommendations for adaptation of the normal guidelines for protection of bone health during the pandemic.

3.
Bone ; 122: 82-92, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30776499

RESUMO

Multiple myeloma (MM) is characterised by destructive lytic bone disease, caused by induction of bone resorption and impaired bone formation. Our understanding of the molecular mechanisms responsible for osteoblast suppression, are limited. Using the 5T2MM murine model of MM we have previously shown that suppression of the activity of a known inhibitor of bone formation Dikkopf-1 (Dkk1) prevents the development of lytic bone disease. Here we have demonstrated that another potential inhibitor of bone formation, sclerostin domain containing 1 (Sostdc1) is expressed at low levels in MM and osteoblast lineage cells when these cells are grown separately in cell culture but its expression is significantly induced in both cell types when these cells are in contact. The distribution of Sostdc1 staining in bones infiltrated with 5TGM1 myeloma cells in vivo suggested its presence in both myeloma and osteoblast lineage populations when in close proximity. We have also shown that recombinant Sostdc1 inhibits both bone morphogenic proteins (BMP2 and 7) and Wnt signalling in primary osteoblasts and suppresses differentiation of these cells. Together, these findings suggest that Sostdc1 expression in 5TGM1-infiltrated bones as a result of the interaction between myeloma and osteoblast lineage populations, could result in suppression of osteoblast differentiation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Comunicação Celular , Linhagem da Célula , Mieloma Múltiplo/patologia , Osteoblastos/patologia , Proteínas Wnt/antagonistas & inibidores , Animais , Calcificação Fisiológica/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Solubilidade , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Tíbia/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
4.
Curr Pharm Des ; 16(27): 3028-36, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20722619

RESUMO

Multiple myeloma is an incurable B cell neoplasm caused by the monoclonal expansion of malignant plasma cells in the bone marrow, often resulting in devastating bone disease. For over 2 decades bisphosphonates have been successfully used to treat the tumour-induced bone disease associated with multiple myeloma. This review will focus on preclinical studies and investigations in patients with multiple myeloma that have led to our current understanding of the mechanisms of action of bisphosphonates in myeloma bone disease. Major advances in the use of bisphosphonates, including findings that they may have additional benefits such as anti-tumour effects and promoting patient survival will be discussed.


Assuntos
Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Doenças Ósseas/tratamento farmacológico , Doenças Ósseas/etiologia , Difosfonatos/farmacologia , Difosfonatos/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Animais , Antineoplásicos/efeitos adversos , Conservadores da Densidade Óssea/efeitos adversos , Conservadores da Densidade Óssea/farmacologia , Conservadores da Densidade Óssea/uso terapêutico , Difosfonatos/efeitos adversos , Desenho de Fármacos , Humanos , Mieloma Múltiplo/complicações , Mieloma Múltiplo/metabolismo , Osteoclastos/efeitos dos fármacos
5.
Curr Pharm Des ; 16(11): 1272-83, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20166980

RESUMO

Cancers which damage the human skeleton include multiple myeloma, where the primary tumour colonises bone directly, or breast and prostate cancer, where malignant cells travel from the primary tumour to form clonal outgrowths within the bone. Owing to the interaction of tumour cells with those normally found in the bone microenvironment, such as osteoclasts and osteoblasts, these cancers affect the closely linked processes of bone formation and resorption. As a result, these twin processes contribute to the clinical manifestations of cancer metastasis, including bone pain and pathological fractures. A critical component of physiologically normal bone remodelling, the RANK/RANKL/OPG pathway, has been implicated in the formation of osteolytic, and possibly osteoblastic, lesions, which characterise the bone disease associated with these malignancies. In these cancers that affect the skeleton in this way the abnormally regulated RANK/RANKL system appears to be the final effector pathway. As a result, there has been much research focused upon targeting these molecules using OPG constructs, peptidomimetics, soluble receptor constructs and antibodies to RANKL, in pre-clinical studies. The success of these studies has paved the way for a clinical programme, the success of which is likely to lead to a new therapeutic approach to treating cancers that develop in the skeleton.


Assuntos
Mieloma Múltiplo/tratamento farmacológico , Neoplasias/tratamento farmacológico , Ligante RANK/antagonistas & inibidores , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Animais , Reabsorção Óssea/fisiopatologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Feminino , Humanos , Masculino , Modelos Biológicos , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/fisiopatologia , Neoplasias/metabolismo , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
6.
Ann Oncol ; 20(8): 1303-17, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19465418

RESUMO

BACKGROUND: Bisphosphonates (BPs) prevent, reduce, and delay multiple myeloma (MM)-related skeletal complications. Intravenous pamidronate and zoledronic acid, and oral clodronate are used for the management of MM bone disease. The purpose of this paper is to review the current evidence for the use of BPs in MM and provide European Union-specific recommendations to support the clinical practice of treating myeloma bone disease. DESIGN AND METHODS: An interdisciplinary, expert panel of specialists on MM and myeloma-related bone disease convened for a face-to-face meeting to review and assess the evidence and develop the recommendations. The panel reviewed and graded the evidence available from randomized clinical trials, clinical practice guidelines, and the body of published literature. Where published data were weak or unavailable, the panel used their own clinical experience to put forward recommendations based solely on their expert opinions. RESULTS: The panel recommends the use of BPs in MM patients suffering from lytic bone disease or severe osteoporosis. Intravenous administration may be preferable; however, oral administration can be considered for patients unable to make hospital visits. Dosing should follow approved indications with adjustments if necessary. In general, BPs are well tolerated, but preventive steps should be taken to avoid renal impairment and osteonecrosis of the jaw (ONJ). The panel agrees that BPs should be given for 2 years, but this may be extended if there is evidence of active myeloma bone disease. Initial therapy of ONJ should include discontinuation of BPs until healing occurs. BPs should be restarted if there is disease progression. CONCLUSIONS: BPs are an essential component of MM therapy for minimizing skeletal morbidity. Recent retrospective data indicate that a modified dosing regimen and preventive measures can greatly reduce the incidence of ONJ.


Assuntos
Neoplasias Ósseas/prevenção & controle , Difosfonatos/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Neoplasias Ósseas/patologia , Humanos , Mieloma Múltiplo/patologia
7.
Leukemia ; 21(10): 2181-91, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17657224

RESUMO

We describe a new model of myeloma bone disease in which beta2m NOD/SCID mice injected with KMS-12-BM cells develop medullary disease after tail vein administration. Micro-computed tomography analysis demonstrated significant bone loss in the tibiae and vertebrae of diseased animals compared to controls, with loss of cortical bone (P<0.01), as well as trabecular bone volume, thickness and number (P<0.05 for all). Bone marrow of diseased animals demonstrated an increase in osteoclasts (P<0.01) and reduction in osteoblasts (P<0.01) compared to control animals. Both bone loss and osteoclast increase correlated with the degree of disease involvement. Mesenchymal stem cells (MSCs) were lentivirally transduced to express human osteoprotegerin (hOPG). Systemic administration of OPG expressing MSC reduced osteoclast activation (P<0.01) and trabecular bone loss in the vertebrae (P<0.05) and tibiae of diseased animals, to levels comparable to non-diseased controls. Because of its predominantly medullary involvement and quantifiable parameters of bone disease, the KMS-12-BM xenogeneic model provides unique opportunities to test therapies targeted at the bone marrow microenvironment.


Assuntos
Modelos Animais de Doenças , Lentivirus/genética , Células-Tronco Mesenquimais/citologia , Mieloma Múltiplo/patologia , Mieloma Múltiplo/terapia , Osteoprotegerina/biossíntese , Animais , Osso e Ossos/metabolismo , Linhagem Celular , Terapia Genética/métodos , Humanos , Cinética , Lentivirus/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante de Neoplasias , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Tíbia/patologia
8.
Leukemia ; 21(4): 805-12, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17315027

RESUMO

The growth and survival of myeloma cells is critically regulated by cells of the bone marrow microenvironment, including osteoblasts. Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) is a potent inducer of myeloma cell apoptosis, however, this antimyeloma activity is inhibited by osteoprotegerin (OPG) released from osteoblasts. Therefore, we hypothesized that specific agonists of TRAIL death receptors would not be inhibited by OPG released from osteoblasts and thus may represent a novel therapeutic approach in multiple myeloma. In the present study, TRAIL-induced apoptosis was demonstrated to be mediated through both DR4 and DR5. Specific agonist antibodies to DR4 or DR5 dose-dependently induced myeloma cell apoptosis, which was not prevented by OPG or by medium conditioned by osteoblasts. Co-culture of myeloma cells with osteoblasts protected against TRAIL-induced apoptosis of myeloma cells, and this protective effect was due to OPG. In contrast, the co-culture of myeloma cells with osteoblasts had no protective effect on apoptosis induced by specific agonists of DR4 or DR5. TRAIL has been proposed as a potential antitumour therapy, but within the bone marrow microenvironment OPG may interfere with the action of TRAIL. Specific agonists of TRAIL death receptors would not be subject to this inhibition and thus may provide an alternative specific antimyeloma therapy.


Assuntos
Apoptose/fisiologia , Células da Medula Óssea/fisiologia , Mieloma Múltiplo/patologia , Osteoprotegerina/farmacologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/efeitos dos fármacos , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Clorometilcetonas de Aminoácidos/farmacologia , Linhagem Celular Tumoral , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Humanos , Osteoblastos/efeitos dos fármacos , Osteoblastos/fisiologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/agonistas , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Immunity ; 19(6): 849-61, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14670302

RESUMO

Autoimmune associated bone disease and intestinal inflammation are closely linked with deregulation and hyperactivation of autoreactive CD4 T cells. How these T cells are activated and mediate disease is not clear. Here we show that in the Interleukin 2-deficient mouse model of autoimmunity spontaneous osteopenia and colitis are caused by increased production of the ligand for receptor activator of NFkappaB (RANKL). RANKL acting via its receptor, receptor activator of NFkappaB (RANK), increases bone turnover and promotes intestinal dendritic cell (DC) survival in vivo. Modulation of RANKL-RANK interactions with exogenous recombinant osteoprotegerin (Fc-OPG) reverses skeletal abnormalities and reduces colitis by decreasing colonic DC numbers. This study identifies a common causal link between bone disease and intestinal inflammation and establishes the importance of DC in mediating colonic inflammation in vivo.


Assuntos
Adjuvantes Imunológicos/farmacologia , Osso e Ossos/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Glicoproteínas/farmacologia , Inflamação/tratamento farmacológico , Animais , Doenças Ósseas Metabólicas/tratamento farmacológico , Doenças Ósseas Metabólicas/genética , Doenças Ósseas Metabólicas/imunologia , Osso e Ossos/imunologia , Proteínas de Transporte/metabolismo , Colo/efeitos dos fármacos , Colo/imunologia , Células Dendríticas/imunologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Inflamação/imunologia , Interleucina-2/genética , Interleucina-2/imunologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Osteoprotegerina , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Receptores Citoplasmáticos e Nucleares , Receptores do Fator de Necrose Tumoral , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
11.
Blood ; 98(13): 3534-40, 2001 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11739154

RESUMO

Multiple myeloma is a B-cell malignancy characterized by the accumulation of plasma cells in the bone marrow and the development of osteolytic bone disease. The present study demonstrates that myeloma cells express the critical osteoclastogenic factor RANKL (the ligand for receptor activator of NF-kappa B). Injection of 5T2MM myeloma cells into C57BL/KaLwRij mice resulted in the development of bone disease characterized by a significant decrease in cancellous bone volume in the tibial and femoral metaphyses, an increase in osteoclast formation, and radiologic evidence of osteolytic bone lesions. Dual-energy x-ray absorptiometry demonstrated a decrease in bone mineral density (BMD) at each of these sites. Treatment of mice with established myeloma with recombinant osteoprotegerin (OPG) protein, the soluble decoy receptor for RANKL, prevented the development of lytic bone lesions. OPG treatment was associated with preservation of cancellous bone volume and inhibition of osteoclast formation. OPG also promoted an increase in femoral, tibial, and vertebral BMD. These data suggest that the RANKL/RANK/OPG system may play a critical role in the development of osteolytic bone disease in multiple myeloma and that targeting this system may have therapeutic potential.


Assuntos
Glicoproteínas/uso terapêutico , Mieloma Múltiplo/complicações , Osteólise/prevenção & controle , Receptores Citoplasmáticos e Nucleares/uso terapêutico , Animais , Densidade Óssea , Osso e Ossos/patologia , Proteínas de Transporte/análise , Proteínas de Transporte/genética , Citometria de Fluxo , Expressão Gênica , Glicoproteínas/administração & dosagem , Masculino , Glicoproteínas de Membrana/análise , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Mieloma Múltiplo/química , Mieloma Múltiplo/patologia , Transplante de Neoplasias , Osteólise/etiologia , Osteólise/patologia , Osteoprotegerina , Ligante RANK , RNA Mensageiro/análise , Receptor Ativador de Fator Nuclear kappa-B , Receptores Citoplasmáticos e Nucleares/administração & dosagem , Receptores do Fator de Necrose Tumoral , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Br J Haematol ; 114(2): 414-21, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11529866

RESUMO

Syndecan-1 is a cell surface proteoglycan that is expressed on human myeloma cells and is thought to act as a co-receptor for certain extracellular matrix proteins and growth factors. The ectodomain of syndecan-1 is thought to be shed from the surface of myeloma cells, although the exact mechanism of release remains unclear. In this study, we used a panel of inhibitors to identify the class of proteinase responsible for shedding the soluble syndecan-1 ectodomain from human myeloma cells. Using enzyme-linked immunosorbent assay, flow cytometry and immunocytochemistry, we demonstrated that myeloma cell lines expressed syndecan-1 on their surface and that this was shed constitutively, but to a varying extent. In addition, phorbol 12-myristate 13-acetate (PMA), an activator of protein kinase C, stimulated a marked loss of cell surface syndecan-1 from each of the cell lines and this was associated with a corresponding increase in soluble syndecan-1. Inhibitors of serine and cysteine proteinases, and matrix-type metalloproteinases, did not inhibit constitutive or PMA-stimulated syndecan-1 shedding from JJN3 and RPMI 8226 cells. However, BB-94, a hydroxamate-based, broad-spectrum, metalloproteinase inhibitor, substantially suppressed constitutive and PMA-stimulated syndecan-1 loss from myeloma cells. These data indicate that a non-matrix-type metalloproteinase is responsible for syndecan-1 shedding from the surface of myeloma cells.


Assuntos
Glicoproteínas de Membrana/metabolismo , Metaloendopeptidases/metabolismo , Mieloma Múltiplo/metabolismo , Proteoglicanas/metabolismo , Anticorpos Monoclonais , Membrana Celular/química , Citometria de Fluxo , Fluoresceína , Humanos , Imuno-Histoquímica/métodos , Isotiocianatos/imunologia , Glicoproteínas de Membrana/análise , Proteína Quinase C/metabolismo , Proteoglicanas/análise , Estatísticas não Paramétricas , Estimulação Química , Sindecana-1 , Sindecanas , Acetato de Tetradecanoilforbol/farmacologia , Células Tumorais Cultivadas/metabolismo
13.
Br J Cancer ; 84(8): 1126-34, 2001 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-11308265

RESUMO

Bisphosphonates are well established in the management of breast-cancer-induced bone disease. Recent studies have suggested that these compounds are effective in preventing the development of bone metastases. However, it is unclear whether this reflects an indirect effect via an inhibition of bone resorption or a direct anti-tumour effect. The breast cancer cell lines, MCF-7 and MDA-MB-231 cells were treated with increasing concentrations of the bisphosphonate, zoledronic acid, for varying time periods, in the presence or absence of paclitaxel. The effects of zoledronic acid were determined by assessing cell number and rate of apoptosis by evaluating changes in nuclear morphology and using a fluorescence nick translation assay. Zoledronic acid caused a dose- and time-dependent decrease in cell number (P< 0.001) and a concomitant increase in tumour cell apoptosis (P< 0.005). Short-term exposure to zoledronic acid was sufficient to cause a significant reduction in cell number and increase in apoptosis (P< 0.05). These effects could be prevented by incubation with geranyl geraniol, suggesting that zoledronic acid-induced apoptosis is mediated by inhibiting the mevalonate pathway. Treatment with zoledronic acid and clinically achievable concentrations of paclitaxel resulted in a 4-5-fold increase in tumour cell apoptosis (P< 0.02). Isobologram analysis revealed synergistic effects on tumour cell number and apoptosis when zoledronic acid and paclitaxel were combined. Short-term treatment with zoledronic acid, which closely resembles the clinical setting, has a clear anti-tumour effect on breast cancer cells. Importantly, the commonly used anti-neoplastic agent, paclitaxel, potentiates the anti-tumour effects of zoledronic acid. These data suggest that, in addition to inhibiting bone resorption, zoledronic acid has a direct anti-tumour activity on breast cancer cells in vitro.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias da Mama/prevenção & controle , Difosfonatos/farmacologia , Imidazóis/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Neoplasias da Mama/patologia , Contagem de Células , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Humanos , Ácido Mevalônico/metabolismo , Paclitaxel/farmacologia , Fatores de Tempo , Células Tumorais Cultivadas , Ácido Zoledrônico
14.
Biochem Biophys Res Commun ; 280(2): 574-80, 2001 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-11162558

RESUMO

MDC-9 is a widely expressed member of the metalloproteinase/disintegrin/cysteine-rich protein family. The disintegrin domain of MDC-9 lacks an RGD motif but has recently been reported to bind the alpha(6)beta(1) integrin; however, it is unclear whether MDC-9 can bind other integrins. In the present study myeloma cells, but not lymphoblastoid cells, were shown to bind to immobilised, recombinantly expressed MDC-9 disintegrin domain (A9dis). Binding was divalent cation-dependent, being supported by Mn(2+) and Ca(2+). Adhesion of myeloma cells to A9dis was completely inhibited by an antibody to the alpha(v)beta(5) integrin but not by antibodies to other subunits. RGD-containing peptides had no effect on binding, suggesting that MDC-9 interacts with alpha(v)beta(5) in an RGD-independent manner. Flow cytometric analyses demonstrated that myeloma cells, but not lymphoblastoid cells, expressed alpha(v)beta(5) on the cell membrane. These data indicated that the disintegrin domain of MDC-9 can function as an adhesion molecule by interacting with an alpha(v)beta(5) integrin.


Assuntos
Moléculas de Adesão Celular/metabolismo , Integrinas/metabolismo , Proteínas de Membrana/metabolismo , Metaloendopeptidases , Proteínas Musculares/metabolismo , Receptores de Vitronectina , Proteínas ADAM , Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/genética , Desintegrinas/química , Citometria de Fluxo , Humanos , Integrinas/química , Linfoma/química , Proteínas de Membrana/química , Proteínas de Membrana/genética , Mieloma Múltiplo/química , Mieloma Múltiplo/metabolismo , Proteínas Musculares/química , Proteínas Musculares/genética , Oligopeptídeos/farmacologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Células Tumorais Cultivadas
15.
Br J Haematol ; 111(1): 283-6, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11091214

RESUMO

Bisphosphonates are effective in the management of bone disease in patients with multiple myeloma and recent reports have suggested that they may also have an anti-tumour activity. In support of this, we have previously demonstrated that bisphosphonates can induce myeloma cell apoptosis in vitro; however, it remains unclear whether this occurs in vivo. We have therefore investigated the effect of the potent bisphosphonate ibandronate in the 5T2MM murine model of established multiple myeloma. Short-term treatment with a high dose of ibandronate had no effect on either myeloma cell number or the proportion of myeloma cells undergoing apoptosis. These observations suggest that although bisphosphonates induce apoptosis in myeloma cells in vitro, they may not have the same anti-tumour effects in vivo.


Assuntos
Difosfonatos/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Neoplasias Experimentais/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Células da Medula Óssea/patologia , Ácido Ibandrônico , Camundongos , Camundongos Endogâmicos C57BL , Mieloma Múltiplo/patologia , Falha de Tratamento , Células Tumorais Cultivadas
16.
J Bone Miner Res ; 15(10): 1935-43, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11028445

RESUMO

Multiple myeloma is associated with the development of osteolytic bone disease characterized by a disruption to normal bone resorption and bone formation. Although studies have shown that myeloma cells produce factors that promote bone resorption little data are available examining the mechanism of decreased bone formation or the factors that mediate this effect. In the present study we describe a novel in vitro coculture system in which to investigate the effect of myeloma cells on osteoblast recruitment and differentiation. Under appropriate conditions mesenchymal stem cells were shown to differentiate into colonies of cells, a proportion of which show characteristics of osteoblasts, in that they express alkaline phosphatase activity and stain positively for collagen and calcium. The addition of the human myeloma cells JJN-3, RPMI-8226, or NCI-H929 to these cultures stimulated a significant increase in the total number of colonies (p < 0.005) and the proportion of osteoblastic colonies (p < 0.005). Media conditioned by these cells also were able to promote the formation of both total and osteoblastic colonies (p < 0.005). The addition of an antibody against the interleukin-6 receptor (IL-6R) blocked myeloma cell and myeloma cell-conditioned media induced osteoblast recruitment (p < 0.01). Furthermore, media conditioned by myeloma cells incubated with phorbol ester, which promotes IL-6R shedding, or a metalloproteinase inhibitor, which inhibits IL-6R shedding, were able to stimulate (p < 0.005) and inhibit osteoblast recruitment (p < 0.005), respectively. In addition, soluble IL-6R (sIL-6R) and IL-6 together, but not alone, were able to promote osteoblastic colony formation (p < 0.01). Taken together these data show that myeloma cells promote osteoblast recruitment by release of sIL-6R from myeloma cells.


Assuntos
Interleucina-6/metabolismo , Interleucina-6/farmacologia , Mieloma Múltiplo/metabolismo , Osteoblastos/efeitos dos fármacos , Receptores de Interleucina-6/metabolismo , Células-Tronco/efeitos dos fármacos , Fosfatase Alcalina/metabolismo , Anticorpos/farmacologia , Remodelação Óssea/efeitos dos fármacos , Cálcio/metabolismo , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Técnicas de Cocultura , Colágeno/metabolismo , Meios de Cultivo Condicionados/farmacologia , Humanos , Mesoderma/citologia , Mesoderma/efeitos dos fármacos , Mieloma Múltiplo/patologia , Osteoblastos/citologia , Osteoblastos/enzimologia , Osteoblastos/metabolismo , Receptores de Interleucina-6/administração & dosagem , Receptores de Interleucina-6/antagonistas & inibidores , Solubilidade , Células-Tronco/citologia , Células-Tronco/enzimologia , Células-Tronco/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Células Tumorais Cultivadas
17.
Br J Haematol ; 108(2): 383-90, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10691869

RESUMO

Interleukin-6 (IL-6) is an important growth and survival factor for myeloma cells. However, the identity of the cells producing IL-6 in vivo remains unclear. Myeloma cells are found closely associated with sites of active bone turnover, and cells of the osteogenic lineage, including bone marrow osteoprogenitors, osteoblasts and bone lining cells, may therefore be ideally placed to synthesize IL-6. We have examined the possibility that human osteogenic cells may produce IL-6 in response to stimulation by myeloma cells. Primary human osteoblasts (hOBs) were isolated from normal donors, co-cultured with the human myeloma cell lines, JJN-3, RPMI-8226 and NCI-H929, and the amount of IL-6 released was determined by enzyme-linked immunosorbent assay (ELISA). All myeloma cells stimulated a significant increase in the production of IL-6 when cultured with hOBs (P < 0.05). Prior fixation of hOBs completely abrogated release of IL-6 in the co-cultures. In contrast, fixed myeloma cells retained the ability to induce IL-6 production, suggesting that hOBs were the principal source of IL-6. Physical separation of myeloma cells from hOBs using transwell inserts caused a partial inhibition of IL-6 release (P < 0.05), whereas the addition of media conditioned by myeloma cells to cultures of hOBs stimulated a significant increase in IL-6 production (P < 0.05). hOBs secreted greater amounts of IL-6 than human bone marrow stromal cells (hBMSCs) (2.2- to 3.5-fold, P < 0.05), but incubating hBMSCs with dexamethasone to stimulate osteoblastic differentiation resulted in an increase in their ability to produce IL-6 (1.7- to 4. 8-fold, P < 0.05) and to respond to myeloma cells (P < 0.05). These data clearly indicate that cells of the osteoblast lineage release significant amounts of IL-6 in response to stimulation by myeloma cells and may contribute to the IL-6 that promotes the proliferation and survival of myeloma cells in vivo.


Assuntos
Interleucina-6/biossíntese , Mieloma Múltiplo/metabolismo , Osteoblastos/metabolismo , Comunicação Celular , Ensaio de Imunoadsorção Enzimática , Humanos , Células Estromais/metabolismo , Células Tumorais Cultivadas
18.
Acta Oncol ; 39(7): 829-35, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11145441

RESUMO

Bisphosphonates are a class of anti-resorptive drugs, which are effective in the treatment of osteoclast-mediated bone disease, including the osteolytic bone disease. which is a major clinical feature of patients with multiple myeloma. Recently, increases in survival following treatment with pamidronate have been observed in some patients with multiple myeloma, raising the possibility that bisphosphonates may also have an anti-tumour effect. We have demonstrated that bisphosphonates can have an anti-tumour effect in human myeloma cell in vitro, and that these anti-tumour effects induced by potent nitrogen-containing bisphosphonates are a result of inhibition of enzymes of the mevalonate pathway. However, we and others have been unable to demonstrate an anti-tumour effect of the potent bisphosphonate ibandronate in vivo, using murine models of multiple myeloma. It is therefore likely that only by studying patients receiving bisphosphonates will we be able to determine whether these compounds have a clinically important anti-tumour effect.


Assuntos
Difosfonatos/farmacologia , Mieloma Múltiplo/tratamento farmacológico , Animais , Difosfonatos/uso terapêutico , Modelos Animais de Doenças , Humanos , Ácido Mevalônico/metabolismo , Camundongos , Mieloma Múltiplo/complicações , Mieloma Múltiplo/fisiopatologia , Osteólise/tratamento farmacológico , Osteólise/etiologia
20.
J Bone Miner Res ; 14 Suppl 2: 53-65, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10510215

RESUMO

Bisphosphonates are chemically stable analogs of inorganic pyrophosphate, which are resistant to breakdown by enzymatic hydrolysis. The biological effects of bisphosphonates on calcium metabolism were originally ascribed to their physico-chemical effects on hydroxyapatite crystals. Although such effects may contribute to their overall action, their effects on cells are probably of greater importance, particularly for the more potent compounds. Remarkable progress has been made in increasing the potency of bisphosphonates as inhibitors of bone resorption, and the most potent compounds in current use are characterized by the presence of a nitrogen atom at critical positions in the side chain which, together with the bisphosphonate moiety itself, seems to be essential for maximal activity. As a class the bisphosphonates offer a very effective means of treating Paget's disease.


Assuntos
Doenças Ósseas/tratamento farmacológico , Difosfonatos/uso terapêutico , Animais , Reabsorção Óssea , Células Cultivadas , Humanos , Modelos Químicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA