Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Br J Haematol ; 190(6): 877-890, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32232850

RESUMO

Future progress in the treatment of multiple myeloma (MM) requires both the characterisation of key drivers of the disease and novel, innovative approaches to tackle these vulnerabilities. The present study focussed on the pre-clinical evaluation of a novel drug class, BMI-1 modulators, in MM. We demonstrate potent activity of PTC-028 and PTC596 in a comprehensive set of in vitro and in vivo models, including models of drug resistance and stromal support. Treatment of MM cells with PTC-028 and PTC596 downregulated BMI-1 protein levels, which was found to correlate with drug activity. Surprisingly, BMI-1 was dispensable for the activity of BMI-1 modulators and MM cell growth. Our data rather point to mitotic arrest accompanied by myeloid cell leukaemia-1 (MCL-1) loss as key anti-MM mechanisms and reveal impaired MYC and AKT signalling activity due to BMI-1 modulator treatment. Moreover, we observed a complete eradication of MM after PTC596 treatment in the 5TGM.1 in vivo model and define epigenetic compounds and B cell leukaemia/lymphoma 2 homology domain 3 (BH3) mimetics as promising combination partners. These results bring into question the postulated role of BMI-1 as an essential MM gene and confirm BMI-1 modulators as potent anti-mitotic agents with encouraging pre-clinical activity that supports their rapid translation into clinical trials.


Assuntos
Antineoplásicos/farmacologia , Benzimidazóis/farmacologia , Mitose/efeitos dos fármacos , Mieloma Múltiplo , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias Experimentais , Complexo Repressor Polycomb 1/antagonistas & inibidores , Pirazinas/farmacologia , Animais , Feminino , Humanos , Masculino , Camundongos , Mieloma Múltiplo/dietoterapia , Mieloma Múltiplo/enzimologia , Mieloma Múltiplo/patologia , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/enzimologia , Neoplasias Experimentais/patologia , Complexo Repressor Polycomb 1/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Adv Biol Regul ; 76: 100651, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31519471

RESUMO

Opsismodysplasia (OPS) is a rare but severe autosomal recessive skeletal chondrodysplasia caused by inactivating mutations in the Inppl1/Ship2 gene. The molecular mechanism leading from Ship2 gene inactivation to OPS is currently unknown. Here, we used our Ship2Δ/Δ mouse expressing reduced amount of a catalytically-inactive SHIP2 protein and a previously reported SHIP2 inhibitor to investigate growth plate development and mineralization in vivo, ex vivo and in vitro. First, as observed in OPS patients, catalytic inactivation of SHIP2 in mouse leads to reduced body length, shortening of long bones, craniofacial dysmorphism, reduced height of the hyperthrophic chondrocyte zone and to defects in growth plate mineralization. Second, intrinsic Ship2Δ/Δ bone defects were sufficient to induce the characteristic OPS alterations in bone growth, histology and mineralization ex vivo. Third, expression of osteocalcin was significantly increased in SHIP2-inactivated chondrocyte cultures whereas production of mineralized nodules was markedly decreased. Targeting osteocalcin mRNA with a specific shRNA increased the production of mineralized nodules. Fourth, levels of p-MEK and p-Erk1/2 were significantly increased in SHIP2-inactivated chondrocytes in response to serum and IGF-1, but not to FGF2, as compared to control chondrocytes. Treatment of chondrocytes and bones in culture with a MEK inhibitor partially rescued the production of mineralized nodules, the size of the hypertrophic chondrocyte zone and bone growth, raising the possibility of a treatment that could partially reduce the phenotype of this severe condition. Altogether, our results indicate that Ship2Δ/Δ mice represent a relevant model for human OPS. They also highlight the important role of SHIP2 in chondrocytes during endochondral ossification and its different differentiation steps. Finally, we identified a role of osteocalcin in mineralized nodules production and for the MEK-Erk1/2 signaling pathway in the OPS phenotype.


Assuntos
Condrócitos/metabolismo , MAP Quinase Quinase Quinases/genética , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/genética , Osteocalcina/genética , Osteocondrodisplasias/genética , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases/genética , Aminoacetonitrila/análogos & derivados , Aminoacetonitrila/farmacologia , Animais , Calcificação Fisiológica/genética , Diferenciação Celular , Condrócitos/patologia , Modelos Animais de Doenças , Fator 2 de Crescimento de Fibroblastos/farmacologia , Regulação da Expressão Gênica , Lâmina de Crescimento/metabolismo , Lâmina de Crescimento/patologia , Humanos , Fator de Crescimento Insulin-Like I/farmacologia , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Osteocalcina/antagonistas & inibidores , Osteocalcina/metabolismo , Osteocondrodisplasias/metabolismo , Osteocondrodisplasias/patologia , Osteogênese/genética , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases/antagonistas & inibidores , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases/deficiência , Fosforilação/efeitos dos fármacos , Cultura Primária de Células , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Tiofenos/farmacologia
3.
Cancers (Basel) ; 11(2)2019 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-30813402

RESUMO

Multiple myeloma osteolytic disease is caused by an uncoupled bone-remodelling process with an increased osteoclast activity. Disease development relies on interactions between myeloma cells and bone marrow stromal cells. Recent findings suggest a role for glycan-binding proteins in myeloma microenvironment. Here, we investigated lectins involved in osteoclastogenesis and their role in myeloma bone disease. Microarray data analysis showed a lower expression of galectin-1 (gal-1) in mature osteoclasts compared to monocytic progenitor cells, confirmed at the RNA and protein levels in osteoclast cultures. Confocal microscopy showed that gal-1 localised predominantly in the sealing zone of mature osteoclasts. Although equal differentiated-osteoclast numbers, gal-1-/- osteoclasts showed a higher resorption activity compared to wild-type controls. Micro-computed tomography showed an aberrant bone phenotype with decreased bone densities in gal-1-/- mice. In vivo, tumour progression was faster in gal-1-/- mice and associated with a marked bone loss. Additionally, myeloma cells were found to decrease gal-1 expression in osteoclasts. Our results demonstrate that galectin-1 regulates osteoclast activity with an increased resorption by gal-1-/- osteoclasts and decreased bone densities in gal-1-/- mice. We observed an enhanced tumour development in gal-1-/- mice compared to wild-type mice, suggesting that galectin-1 has a functional role in stromal cells in myeloma microenvironment.

4.
Blood Cancer J ; 8(11): 105, 2018 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-30409995

RESUMO

Progression of multiple myeloma (MM) is largely dependent on the bone marrow (BM) microenvironment wherein communication through different factors including extracellular vesicles takes place. This cross-talk not only leads to drug resistance but also to the development of osteolysis. Targeting vesicle secretion could therefore simultaneously ameliorate drug response and bone disease. In this paper, we examined the effects of MM exosomes on different aspects of osteolysis using the 5TGM1 murine model. We found that 5TGM1 sEVs, or 'exosomes', not only enhanced osteoclast activity, they also blocked osteoblast differentiation and functionality in vitro. Mechanistically, we could demonstrate that transfer of DKK-1 led to a reduction in Runx2, Osterix, and Collagen 1A1 in osteoblasts. In vivo, we uncovered that 5TGM1 exosomes could induce osteolysis in a similar pattern as the MM cells themselves. Blocking exosome secretion using the sphingomyelinase inhibitor GW4869 not only increased cortical bone volume, but also it sensitized the myeloma cells to bortezomib, leading to a strong anti-tumor response when GW4869 and bortezomib were combined. Altogether, our results indicate an important role for exosomes in the BM microenvironment and suggest a novel therapeutic target for anti-myeloma therapy.


Assuntos
Doenças Ósseas/etiologia , Doenças Ósseas/metabolismo , Exossomos/metabolismo , Mieloma Múltiplo/complicações , Mieloma Múltiplo/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Compostos de Anilina/farmacologia , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Compostos de Benzilideno/farmacologia , Biomarcadores , Reabsorção Óssea/metabolismo , Bortezomib/farmacologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/metabolismo , Modelos Animais de Doenças , Exossomos/ultraestrutura , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/ultraestrutura , Feminino , Humanos , Camundongos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/etiologia , Osteoblastos/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Osteólise , Padrão de Cuidado , Carga Tumoral , Via de Sinalização Wnt
5.
Front Immunol ; 9: 1943, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30214443

RESUMO

Despite the increasing use of humanized mouse models to study new approaches of graft-versus-host disease (GVHD) prevention, the pathogenesis of xenogeneic GVHD (xGVHD) in these models remains misunderstood. The aim of this study is to describe this pathogenesis in NOD/LtSz-PrkdcscidIL2rγtm1Wjl (NSG) mice infused with human PBMCs and to assess the impact of the expression of HLA-A0201 by NSG mice cells (NSG-HLA-A2/HHD mice) on xGVHD and graft-versus-leukemia (GvL) effects, by taking advantage of next-generation technologies. We found that T cells recovered from NSG mice after transplantation had upregulated expression of genes involved in cell proliferation, as well as in TCR, co-stimulatory, IL-2/STAT5, mTOR and Aurora kinase A pathways. T cells had mainly an effector memory or an effector phenotype and exhibited a Th1/Tc1-skewed differentiation. TCRß repertoire diversity was markedly lower both in the spleen and lungs (a xGVHD target organ) than at infusion. There was no correlation between the frequencies of specific clonotypes at baseline and in transplanted mice. Finally, expression of HLA-A0201 by NSG mice led to more severe xGVHD and enhanced GvL effects toward HLA-A2+ leukemic cells. Altogether our data demonstrate that the pathogenesis of xGVHD shares important features with human GVHD and that NSG-HLA-A2/HHD mice could serve as better model to study GVHD and GvL effects.


Assuntos
Regulação da Expressão Gênica/imunologia , Doença Enxerto-Hospedeiro/imunologia , Antígeno HLA-A2/imunologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/transplante , Animais , Doença Enxerto-Hospedeiro/genética , Doença Enxerto-Hospedeiro/patologia , Efeito Enxerto vs Leucemia/genética , Efeito Enxerto vs Leucemia/imunologia , Antígeno HLA-A2/genética , Xenoenxertos , Humanos , Leucócitos Mononucleares/patologia , Camundongos
6.
Rev Med Suisse ; 14(615): 1438-1442, 2018 Aug 22.
Artigo em Francês | MEDLINE | ID: mdl-30136458

RESUMO

Multiple myeloma is the second most frequent hematological malignancy. Unfortunately, it is still incurable. A better understanding of the myeloma pathophysiology favored the development of new therapeutic molecules that improved both survival and quality of life of patients. Diagnostic and prognostic criteria for myeloma have been reviewed and help to detect multiple myeloma more early and further help to define the best therapeutic strategy. These new regimens are associated with side effects that differ from those of classic molecules and that we have to be able to recognize and to treat appropriately.


Le myélome multiple est le second cancer hématologique le plus fréquent. Il reste malheureusement incurable à l'heure actuelle. Pourtant, ces deux dernières décennies ont été marquées par des progrès dans la compréhension de la physiopathologie du myélome, ce qui a permis le développement de nouvelles molécules thérapeutiques, améliorant la survie et la qualité de vie des patients. Les critères diagnostiques et pronostiques du myélome ont été revus et sont maintenant plus performants pour détecter des stades précoces et déterminer ainsi la stratégie thérapeutique à entreprendre. Les nouveaux régimes de traitement sont grevés d'un profil d'effets secondaires différent qu'il faut pouvoir reconnaître et traiter.


Assuntos
Mieloma Múltiplo , Humanos , Mieloma Múltiplo/terapia , Prognóstico , Qualidade de Vida
7.
Haematologica ; 103(8): 1359-1368, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29748441

RESUMO

Multiple myeloma bone disease is characterized by an uncoupling of bone remodeling in the multiple myeloma microenvironment, resulting in the development of lytic bone lesions. Most myeloma patients suffer from these bone lesions, which not only cause morbidity but also negatively impact survival. The development of novel therapies, ideally with a combined anti-resorptive and bone-anabolic effect, is of great interest because lesions persist with the current standard of care, even in patients in complete remission. We have previously shown that MELK plays a central role in proliferation-associated high-risk multiple myeloma and its inhibition with OTSSP167 resulted in decreased tumor load. MELK inhibition in bone cells has not yet been explored, although some reports suggest that factors downstream of MELK stimulate osteoclast activity and inhibit osteoblast activity, which makes MELK inhibition a promising therapeutic approach. Therefore, we assessed the effect of OTSSP167 on bone cell activity and the development of myeloma-induced bone disease. OTSSP167 inhibited osteoclast activity in vitro by decreasing progenitor viability as well as via a direct anti-resorptive effect on mature osteoclasts. In addition, OTSSP167 stimulated matrix deposition and mineralization by osteoblasts in vitro This combined anti-resorptive and osteoblast-stimulating effect of OTSSP167 resulted in the complete prevention of lytic lesions and bone loss in myeloma-bearing mice. Immunohistomorphometric analyses corroborated our in vitro findings. In conclusion, we show that OTSSP167 has a direct effect on myeloma-induced bone disease in addition to its anti-multiple myeloma effect, which warrants further clinical development of MELK inhibition in multiple myeloma.


Assuntos
Doenças Ósseas/tratamento farmacológico , Mieloma Múltiplo/tratamento farmacológico , Naftiridinas/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Animais , Doenças Ósseas/etiologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Feminino , Xenoenxertos , Humanos , Camundongos , Mães , Mieloma Múltiplo/complicações , Mieloma Múltiplo/patologia , Naftiridinas/uso terapêutico , Osteoblastos/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Osteólise/tratamento farmacológico , Osteólise/prevenção & controle , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico
8.
Leuk Lymphoma ; 59(1): 14-28, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28573897

RESUMO

Multiple myeloma (MM) bone disease is a major cause of morbidity and mortality in MM patients and persists even in patients in remission. This bone disease is caused by an uncoupling of bone remodeling, with increased osteoclast and decreased osteoblast activity and formation, culminating in lytic bone destruction. Bisphosphonates are the current standard of care but new therapies are needed. As the molecular mechanisms controlling MM bone disease are increasingly well understood, new therapeutic targets are extensively explored in the preclinical setting and initial clinical trials with novel compounds now show promising results. In this review, we will provide a comprehensive overview of the biology of MM bone disease, summarize its current clinical management and discuss preclinical and clinical data on next generation therapies.


Assuntos
Doenças Ósseas/etiologia , Doenças Ósseas/terapia , Mieloma Múltiplo/complicações , Animais , Conservadores da Densidade Óssea/farmacologia , Conservadores da Densidade Óssea/uso terapêutico , Doenças Ósseas/diagnóstico , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , Medula Óssea/patologia , Remodelação Óssea , Reabsorção Óssea/tratamento farmacológico , Ensaios Clínicos como Assunto , Difosfonatos/farmacologia , Difosfonatos/uso terapêutico , Gerenciamento Clínico , Avaliação Pré-Clínica de Medicamentos , Humanos , Mieloma Múltiplo/patologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteogênese/efeitos dos fármacos , Transdução de Sinais , Resultado do Tratamento , Microambiente Tumoral
9.
Haematologica ; 103(2): 325-335, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29122991

RESUMO

Treatment of high-risk patients is a major challenge in multiple myeloma. This is especially true for patients assigned to the gene expression profiling-defined proliferation subgroup. Although recent efforts have identified some key players of proliferative myeloma, genetic interactions and players that can be targeted with clinically effective drugs have to be identified in order to overcome the poor prognosis of these patients. We therefore examined maternal embryonic leucine zipper kinase (MELK) for its implications in hyper-proliferative myeloma and analyzed the activity of the MELK inhibitor OTSSP167 both in vitro and in vivoMELK was found to be significantly overexpressed in the proliferative subgroup of myeloma. This finding translated into poor overall survival in patients with high vs low MELK expression. Enrichment analysis of upregulated genes in myeloma cells of MELKhigh patients confirmed the strong implications in myeloma cell proliferation. Targeting MELK with OTSSP167 impaired the growth and survival of myeloma cells, thereby affecting central survival factors such as MCL-1 and IRF4 This activity was also observed in the 5TGM.1 murine model of myeloma. OTSSP167 reduced bone marrow infiltration and serum paraprotein levels in a dose-dependent manner. In addition, we revealed a strong link between MELK and other proliferation-associated high-risk genes (PLK-1, EZH2, FOXM1, DEPDC1) and MELK inhibition also impaired the expression of those genes. We therefore conclude that MELK is an essential component of a proliferative gene signature and that pharmacological inhibition of MELK represents an attractive novel approach to overcome the poor prognosis of high-risk patients with a proliferative expression pattern.


Assuntos
Proliferação de Células/efeitos dos fármacos , Mieloma Múltiplo/tratamento farmacológico , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Camundongos , Mieloma Múltiplo/patologia , Naftiridinas/farmacologia , Prognóstico , Proteínas Serina-Treonina Quinases/metabolismo , Medição de Risco
10.
J Hematol Oncol ; 9(1): 53, 2016 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-27377819

RESUMO

BACKGROUND: Previous studies have demonstrated that regulatory T cells (Tregs) play a protective role in the pathogenesis of chronic graft-versus-host disease (cGVHD). Tregs constitutively express the gene of the transcription factor Foxp3 whose CNS2 region is heavily methylated in conventional CD4(+) T cells (CD4(+)Tconvs) but demethylated in Tregs. METHODS: Here, we assessed the impact of azacytidine (AZA) on cGVHD in a well-established murine model of sclerodermic cGVHD (B10.D2 (H-2d) → BALB/cJ (H-2d)). RESULTS: The administration of AZA every 48 h from day +10 to day +30 at the dose of 0.5 mg/kg or 2 mg/kg mitigated chronic GVHD. Further, AZA-treated mice exhibited higher blood and thymic Treg frequencies on day +35, as well as higher demethylation levels of the Foxp3 enhancer and the IL-2 promoter in splenocytes at day +52. Interestingly, Tregs from AZA-treated mice expressed more frequently the activation marker CD103 on day +52. AZA-treated mice had also lower counts of CD4(+)Tconvs and CD8(+) T cells from day +21 to day +35 after transplantation, as well as a lower proportion of CD4(+)Tconvs expressing the Ki67 antigen on day +21 demonstrating an anti-proliferating effect of the drug on T cells. CONCLUSIONS: Our results indicate that AZA prevented sclerodermic cGVHD in a well-established murine model of cGVHD. These data might serve as the basis for a pilot study of AZA administration for cGVHD prevention in patients at high risk for cGVHD.


Assuntos
Azacitidina/administração & dosagem , Doença Enxerto-Hospedeiro/prevenção & controle , Escleroderma Sistêmico/prevenção & controle , Animais , Antimetabólitos Antineoplásicos/administração & dosagem , Transplante de Medula Óssea/efeitos adversos , Proliferação de Células/efeitos dos fármacos , Metilação de DNA , Esquema de Medicação , Fatores de Transcrição Forkhead/genética , Contagem de Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Baço/citologia , Linfócitos T Reguladores/imunologia
11.
Oncotarget ; 7(25): 37931-37943, 2016 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-27177328

RESUMO

Multiple myeloma (MM) is a plasma cell malignancy characterized by the accumulation of tumor cells in the bone marrow (BM) and is associated with immunosuppression, angiogenesis and osteolysis. Myeloid-derived suppressor cells (MDSCs) represent a heterogeneous population of immature, immunosuppressive myeloid cells that promote tumor progression through different mechanisms.In this work, we studied the contribution of MDSC subsets to different disease-promoting aspects in MM. We observed an expansion of polymorphonuclear/granulocytic (PMN-)MDSCs in two immunocompetent murine MM models, while this was not observed for monocytic (MO-)MDSCs. Both MDSC subpopulations from MM-bearing mice were immunosuppressive, but PMN-MDSCs displayed a higher suppressive potential. Soluble factors secreted by MM cells increased the viability of MDSCs, whereas the presence of MDSCs did not affect the proliferation of MM cells in vitro or in vivo. Interestingly, we observed a pro-angiogenic effect of PMN-MDSCs in the context of MM using the chick chorioallantoic membrane assay. Consistently, MM-derived PMN-MDSCs showed an up-regulation of angiogenesis-related factors and reduced PMN-MDSC levels were associated with less angiogenesis in vivo. Finally, we identified MO-MDSCs as osteoclast precursors.These results suggest that MDSC subpopulations play diverging roles in MM. We show for the first time that PMN-MDSCs exert a pro-angiogenic role in MM.


Assuntos
Granulócitos/metabolismo , Mieloma Múltiplo/genética , Células Supressoras Mieloides/metabolismo , Neovascularização Patológica/metabolismo , Animais , Humanos , Camundongos , Mieloma Múltiplo/patologia , Neovascularização Patológica/patologia
12.
Oncotarget ; 7(21): 30712-29, 2016 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-27095574

RESUMO

Multiple myeloma (MM)-associated osteolytic bone disease is a major cause of morbidity and mortality in MM patients and the development of new therapeutic strategies is of great interest. The proto-oncogene SRC is an attractive target for such a strategy. In the current study, we investigated the effect of treatment with the SRC inhibitor saracatinib (AZD0530) on osteoclast and osteoblast differentiation and function, and on the development of MM and its associated bone disease in the 5TGM.1 and 5T2MM murine MM models. In vitro data showed an inhibitory effect of saracatinib on osteoclast differentiation, polarization and resorptive function. In osteoblasts, collagen deposition and matrix mineralization were affected by saracatinib. MM cell proliferation and tumor burden remained unaltered following saracatinib treatment and we could not detect any synergistic effects with drugs that are part of standard care in MM. We observed a marked reduction of bone loss after treatment of MM-bearing mice with saracatinib as reflected by a restoration of trabecular bone parameters to levels observed in naive control mice. Histomorphometric analyses support that this occurs through an inhibition of bone resorption. In conclusion, these data further establish SRC inhibition as a promising therapeutic approach for the treatment of MM-associated osteolytic bone disease.


Assuntos
Benzodioxóis/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Osteólise/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Proto-Oncogenes/efeitos dos fármacos , Quinazolinas/uso terapêutico , Quinases da Família src/antagonistas & inibidores , Administração Oral , Animais , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/patologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mieloma Múltiplo/complicações , Mieloma Múltiplo/patologia , Osteoblastos/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Osteólise/etiologia , Osteólise/patologia , Proto-Oncogene Mas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...