Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Br J Haematol ; 156(5): 633-42, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22211565

RESUMO

In view of the fact that histone deacetylases (HDACs) are promising targets for myeloma therapy, we investigated the effects of the HDAC inhibitor CR2408 on multiple myeloma (MM) cells in vitro. CR2408 is a direct pan-HDAC inhibitor and inhibits all known 11 HDACs with a 50% inhibitory concentration (IC(50) ) of 12 nmol/l (HDAC 6) to 520 nmol/l (HDAC 8). Correspondingly, CR2408 induces hyperacetylation of histone H4, inhibits cell growth and strongly induces apoptosis (IC(50) =0.1-0.5 µmol/l) in MM cell lines and primary MM cells. CR2408 leads to fragmentation of cells and induces an accumulation in the subG1 phase accompanied with moderately decreased levels of cyclin D1 and cdk4 and strongly decreased levels of cdc25a, pRb and p53. Interruption of the cell cycle is reflected by inhibition of cell proliferation and is accompanied by decreased phosphorylation of 4E-BP1 and p70S6k. Treatment with CR2408 results in increased protein levels of Bim and pJNK and downregulation of Bad and Bcl-xL and activation of Caspases 3, 8 and 9. Furthermore, as HDAC inhibitors have shown synergism with other drugs, these effects were investigated and synergism was observed for combinations of CR2408 with doxorubicin and bortezomib. In conclusion, we have identified potent anti-myeloma activity for this novel HDAC inhibitor that gives further insights into the biological sequelae of HDAC inhibition in MM.


Assuntos
Antineoplásicos/farmacologia , Inibidores de Histona Desacetilases/farmacologia , Mieloma Múltiplo/tratamento farmacológico , Pirróis/farmacologia , Sulfonamidas/farmacologia , Acetilação/efeitos dos fármacos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Ácidos Borônicos/administração & dosagem , Bortezomib , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Doxorrubicina/administração & dosagem , Avaliação Pré-Clínica de Medicamentos/métodos , Sinergismo Farmacológico , Histonas/metabolismo , Humanos , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Pirazinas/administração & dosagem , Pirróis/administração & dosagem , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/administração & dosagem , Células Tumorais Cultivadas
2.
Anticancer Drugs ; 23(1): 131-8, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21959532

RESUMO

Multiple myeloma is still uncurable. Myeloma cells become resistant to common drugs and patients eventually die of tumour progression. Therefore, new targets and drugs are urgently needed. NVP-BGT226 is a novel, orally bioavailable small-molecule inhibitor of phosphoinositol-3-kinase and mammalian target of rapamycin. Here, we show that NVP-BGT226 inhibits growth in common myeloma cell lines and primary myeloma cells at nanomolar concentrations in a time-dependent and dose-dependent manner. Western blots for the detection of caspase 3 cleavage and annexin-V-fluorescein isothiocyanate/propidium iodide assays revealed induction of apoptosis in common myeloma cells lines. Induction of apoptosis was accompanied by upregulation of proapoptotic Bim and a moderate upregulation of Mcl-1 and Bad and a downregulation of Bcl-2, Bax and Bcl-Xl. Inhibition of cell growth was mainly due to inhibition of myeloma cell proliferation, as shown by the 5-bromo-2'-deoxyuridine assay. Cell cycle analysis revealed induction of cell cycle arrest in the G1 phase, which was due to downregulation of cyclin D1, cyclin D2, pRb and cdc25a. NVP-BGT226 inhibited phosphorylation of protein kinase B (Akt), P70S6k and 4E-BP-1 in a time-dependent and dose-dependent manner. Furthermore, we show that the stimulatory effect of insulin-like growth factor 1, interleukin-6 and conditioned medium of HS-5 stromal cells on myeloma cell growth is completely abrogated by NVP-BGT226. Overall, inhibition of phosphoinositol-3-kinase/mammalian target of rapamycin by NVP-BGT226 is highly effective, and NVP-BGT226 represents a potential new candidate for targeted therapy in multiple myeloma.


Assuntos
Antineoplásicos/farmacologia , Imidazóis/farmacologia , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Quinolinas/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Meios de Cultura/farmacologia , Relação Dose-Resposta a Droga , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Humanos , Fator de Crescimento Insulin-Like I/farmacologia , Interleucina-6/farmacologia , Transdução de Sinais/efeitos dos fármacos , Células Estromais/química
3.
Am J Surg Pathol ; 34(9): 1382-7, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20739840

RESUMO

We report a case of primary Epstein Barr virus (EBV) negative peripheral T-cell lymphoma (PTCL) NOS in a 56-year-old female who-after an initially indolent course - simultaneously developed an aggressive, EBV+ cytotoxic large T-cell lymphoma, clonally related to the primary PTCL, and an EBV+, clonal large B-cell lymphoproliferation. The initial, EBV-negative PTCL had shown some features of angioimmunoblastic T-cell lymphoma and had responded well to steroid therapy. Two years later, rapidly fatal, progressive disease with multivisceral involvement developed. Histologically, extensive infiltrates of EBV+, CD8+ large cells were present, in addition to areas of the initial PTCL. Extensive comparative phenotypic and molecular analyses confirmed the presence of an identical CD8+ T-cell clone in the initial EBV-negative PTCL and the EBV+, CD8+ large cell lymphoma at the time of aggressive transformation. These results also justified the retrospective classification of PTCL, NOS for the initial lymphoma. This case shows that secondary EBV infection of an established malignant T-cell clone can occur and may contribute to aggressive transformation of PTCL.


Assuntos
Infecções por Vírus Epstein-Barr/patologia , Herpesvirus Humano 4/isolamento & purificação , Linfoma de Células T Periférico/patologia , Segunda Neoplasia Primária/patologia , Linfócitos T Citotóxicos/patologia , Transformação Celular Neoplásica , Células Clonais , DNA de Neoplasias/análise , DNA Viral/análise , Progressão da Doença , Infecções por Vírus Epstein-Barr/complicações , Evolução Fatal , Feminino , Herpesvirus Humano 4/fisiologia , Humanos , Imuno-Histoquímica , Imunofenotipagem , Hibridização In Situ , Linfonodos/patologia , Linfoma de Células T Periférico/virologia , Pessoa de Meia-Idade , Segunda Neoplasia Primária/virologia , Linfócitos T Citotóxicos/virologia
4.
Br J Haematol ; 149(4): 518-28, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20201941

RESUMO

Inhibition of histone deacetylase (HDAC) is a promising mechanism for novel, anti-myeloma agents. We investigated the effects of the novel HDAC inhibitor resminostat on multiple myeloma (MM) cells in vitro. Resminostat is a potent inhibitor of HDACs 1, 3 and 6 [50% inhibitory concentration (IC50)=43-72 nmol/l] representing HDAC classes I and II and induces hyperacetylation of histone H4 in MM cells. Low micromolar concentrations of resminostat abrogated cell growth and strongly induced apoptosis (IC50=2.5-3 micromol/l in 3 out of 4 MM cell lines) in MM cell lines as well as primary MM cells. At 1 micromol/l, resminostat inhibited proliferation and induced G0/G1 cell cycle arrest in 3 out of 4 MM cell lines accompanied with decreased levels of cyclin D1, cdc25a, Cdk4 and pRb as well as upregulation of p21. Resminostat decreased phosphorylation of 4E-BP1 and p70S6k indicating an interference with Akt pathway signalling. Treatment with resminostat resulted in increased protein levels of Bim and Bax and decreased levels of Bcl-xL. Caspases 3, 8 and 9 were activated by resminostat. Furthermore, synergistic effects were observed for combinations of resminostat with melphalan and the proteasome inhibitors bortezomib and S-2209. In conclusion, we have identified potent anti-myeloma activity for this novel HDAC inhibitor.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Mieloma Múltiplo/patologia , Sulfonamidas/farmacologia , Acetilação/efeitos dos fármacos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Histonas/metabolismo , Humanos , Mieloma Múltiplo/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas
5.
Mol Cancer Ther ; 9(2): 300-10, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20124446

RESUMO

Evidence is increasing that aberrant NF-kappaB activation is crucial for multiple myeloma pathophysiology and a promising target for new antimyeloma therapies. In this study, we assessed the in vitro antimyeloma activity of the novel NF-kappaB inhibitor V1810. Pharmacokinetics and toxicity were studied in vivo. In mice, V1810 plasma concentrations of 10 micromol/L can be reached without relevant toxicity. At this concentration, V1810 potently induces apoptosis in all four multiple myeloma cell lines assessed (IC(50) = 5-12 micromol/L) as well as in primary multiple myeloma cells (IC(50) = 5-40 micromol/L). Apoptosis induced by V1810 is associated with proteasome-independent inhibition of NF-kappaB signaling (41% relative reduction), downregulation of Mcl-1, and caspase 3 cleavage. In OPM2, U266, and RPMI-8226 cells, induction of apoptosis is accompanied by cell cycle arrest. Western blots revealed downregulation of Cdk4 as well as cyclin D1 (U266) or cyclin D2 (OPM2, NCI-H929, RPMI-8226), but not cyclin D3. Consistently, retinoblastoma protein was found to be hypophosphorylated. Furthermore, V1810 reverses NF-kappaB activation induced by the genotoxic drugs melphalan and doxorubicin. V1810 and melphalan synergistically decrease multiple myeloma cell viability. Taken together, the novel, proteasome-independent NF-kappaB inhibitor V1810 induces apoptosis and cell cycle arrest in multiple myeloma cells at a concentration range that can be achieved in vivo. Moreover, V1810 reverses NF-kappaB activation by alkylating drugs and overcomes NF-kappaB-mediated resistance to melphalan.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Benzimidazóis/farmacologia , Resistencia a Medicamentos Antineoplásicos , Mieloma Múltiplo/metabolismo , NF-kappa B/antagonistas & inibidores , Inibidores de Proteassoma , Tiazóis/farmacologia , Animais , Ciclo Celular , Sobrevivência Celular , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Concentração Inibidora 50 , Melfalan/farmacologia , Camundongos , Fosforilação , Complexo de Endopeptidases do Proteassoma/metabolismo
6.
Anticancer Drugs ; 20(4): 259-66, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19240643

RESUMO

Multiple myeloma is still incurable. Myeloma cells become resistant to common drugs and patients eventually die of tumour progression. Therefore, new targets and drugs are needed immediately. NVP-AEW541 is a new, orally bioavailable small molecule inhibitor of the insulin-like growth factor-1 receptor (IGF-1R). Here, we show that NVP-AEW541 inhibits cell growth in myeloma cells at low concentrations in a time-dependent and a dose-dependent manner. Further experiments using the annexin-V-fluorescein isothiocyanate/propidium iodide assay revealed induction of apoptosis in common myeloma cell lines, but not in peripheral blood mononuclear cell from healthy donors. Stimulation of myeloma cells with IGF-1 led to a vast increase of cell growth and this was blocked by low doses of NVP-AEW541. Stimulation of myeloma cells with conditioned medium obtained from a 48-h-old HS-5 stromal cell culture was only partly blocked by NVP-AEW541. Western blotting experiments revealed that NVP-AEW541 decreased the phosphorylation status of P70S6 kinase and 4E-BP-1 but not of mammalian target of rapamycin (mTOR). Combined inhibition of IGF-1R and mTOR using the novel mTOR inhibitor Rad001 led to additive/synergistic increase of cell growth inhibition in multiple myeloma cells, which was accompanied by a stronger dephosphorylation of P70S6 kinase and 4E-BP-1. Taken together, we show that the combined inhibition of IGF-1R and mTOR by combining NVP-AEW541 and Rad001 is highly effective in multiple myeloma and might represent a potential new treatment strategy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Mieloma Múltiplo/tratamento farmacológico , Proteínas Quinases/efeitos dos fármacos , Receptor IGF Tipo 1/antagonistas & inibidores , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Apoptose/efeitos dos fármacos , Western Blotting , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Sistemas de Liberação de Medicamentos , Sinergismo Farmacológico , Everolimo , Humanos , Mieloma Múltiplo/patologia , Fosforilação/efeitos dos fármacos , Proteínas Quinases/metabolismo , Pirimidinas/administração & dosagem , Pirróis/administração & dosagem , Sirolimo/administração & dosagem , Sirolimo/análogos & derivados , Serina-Treonina Quinases TOR , Fatores de Tempo
7.
Br J Haematol ; 144(6): 875-86, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19170678

RESUMO

Bortezomib is the first approved member of a new class of anti-myeloma agents, the proteasome inhibitors. Further proteasome inhibitors are needed to optimise this promising treatment option. S-2209 [1-[1-{1-[(2,4-Dioxo-imidazolidin-1-ylimino)-methyl]-2-phenyl-ethylcarbamoyl}-2-(1H-indol-3-yl)-ethylcarbamoyl]-2-(1H-indol)] inhibits the chymotryptic activity of the human 20S proteasome (half maximal effective concentration, IC(50) approximately 220 nmol/l) which was determined by a proteasome inhibition assay. A nuclear factor kappaB inhibition assay revealed a half maximal effective concentration (EC(50)) of 0.9 micromol/l. The WST-1 growth assay showed inhibition of cell growth of all tested multiple myeloma (MM) cell lines with an IC(50) between 100 nmol/l and 600 nmol/l. Strong induction of apoptosis was seen in MM cells at nanomolar concentrations (IC(50) approximately 300 nm) as well as in primary myeloma cells. No induction of apoptosis was detected in peripheral blood mononuclear cells from healthy humans. Upregulation of p53, activation of JNK protein, and downregulation of Mcl-1 was revealed. Despite the administration of 15 mg S-2209/kg/d in wistar rats, no toxicity with respect to body weight, hepatic enzymes, creatinine or haemoglobin was seen. Proteasome inhibition in white blood cells isolated from the treated rats was higher in the S-2209 treated animals in comparison with the control animals treated with 0.1 mg/kg/d bortezomib. S-2209 is active in myeloma cells and shows a favourable toxicity profile in first in-vivo studies. S-2209 is a promising agent for further clinical development.


Assuntos
Mieloma Múltiplo/tratamento farmacológico , Peptídeos/uso terapêutico , Inibidores de Proteassoma , Semicarbazonas/uso terapêutico , Animais , Antimetabólitos/uso terapêutico , Apoptose/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Doxorrubicina/uso terapêutico , Sinergismo Farmacológico , Humanos , MAP Quinase Quinase 4/metabolismo , Masculino , Dose Máxima Tolerável , Melfalan/uso terapêutico , Mieloma Múltiplo/metabolismo , Proteína de Sequência 1 de Leucemia de Células Mieloides , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Peptídeos/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Wistar , Semicarbazonas/farmacologia , Testes de Toxicidade , Proteína Supressora de Tumor p53/metabolismo
8.
Mol Cancer Ther ; 8(2): 366-75, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19174558

RESUMO

Multiple myeloma is still an incurable disease; therefore, new therapeutics are urgently needed. A771726 is the active metabolite of the immunosuppressive drug leflunomide, which is currently applied in the treatment of rheumatoid arthritis, BK virus nephropathy, and cytomegaly viremia. Here, we show that dihydroorotate dehydrogenase (DHODH) is commonly expressed in multiple myeloma cell lines and primary multiple myeloma cells. The DHODH inhibitor A771726 inhibits cell growth in common myeloma cell lines at clinically achievable concentrations in a time- and dose-dependent manner. Annexin V-FITC/propidium iodide staining revealed induction of apoptosis of multiple myeloma cell lines and primary multiple myeloma cells. The 5-bromo-2'-deoxyuridine cell proliferation assay showed that inhibition of cell growth was partly due to inhibition of multiple myeloma cell proliferation. A771726 induced G(1) cell cycle arrest via modulation of cyclin D2 and pRb expression. A771726 decreased phosphorylation of protein kinase B (Akt), p70S6K, and eukaryotic translation initiation factor 4E-binding protein-1 as shown by Western blotting experiments. Furthermore, we show that the stimulatory effect of conditioned medium of HS-5 bone marrow stromal cells on multiple myeloma cell growth is completely abrogated by A771726. In addition, synergism studies revealed synergistic and additive activity of A771726 together with the genotoxic agents melphalan, treosulfan, and doxorubicin as well as with dexamethasone and bortezomib. Taken together, we show that inhibition of DHODH by A771726/leflunomide is effective in multiple myeloma. Considering the favorable toxicity profile and the great clinical experience with leflunomide in rheumatoid arthritis, this drug represents a potential new candidate for targeted therapy in multiple myeloma.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Isoxazóis/farmacologia , Mieloma Múltiplo/enzimologia , Mieloma Múltiplo/patologia , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Antineoplásicos/farmacologia , Moléculas de Adesão Celular/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Meios de Cultura , Di-Hidro-Orotato Desidrogenase , Regulação para Baixo/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Fase G1/efeitos dos fármacos , Humanos , Leflunomida , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores de Citocinas/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Células Estromais/efeitos dos fármacos , Uridina/deficiência
9.
Exp Cell Res ; 315(3): 485-97, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19071109

RESUMO

NVP-BEZ235 is a new inhibitor of phosphoinositol-3-kinase (PI3 kinase) and mammalian target of rapamycin (mTOR) whose efficacy in advanced solid tumours is currently being evaluated in a phase I/II clinical trial. Here we show that NVP-BEZ235 inhibits growth in common myeloma cell lines as well as primary myeloma cells at nanomolar concentrations in a time and dose dependent fashion. Further experiments revealed induction of apoptosis in three of four cell lines. Inhibition of cell growth was mainly due to inhibition of myeloma cell proliferation, as shown by the BrdU assay. Cell cycle analysis revealed induction of cell cycle arrest in the G1 phase, which was due to downregulation of cyclin D1, pRb and cdc25a. NVP-BEZ235 inhibited phosphorylation of protein kinase B (Akt), P70S6k and 4E-BP-1. Furthermore we show that the stimulatory effect of CD40-ligand (CD40L), insulin-like growth factor 1 (IGF-1), interleukin-6 (IL-6) and conditioned medium of HS-5 stromal cells on myeloma cell growth is completely abrogated by NVP-BEZ235. In addition, synergism studies revealed synergistic and additive activity of NVP-BEZ235 together with melphalan, doxorubicin and bortezomib. Taken together, inhibition of PI3 kinase/mTOR by NVP-BEZ235 is highly effective and NVP-BEZ235 represents a potential new candidate for targeted therapy in multiple myeloma.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Imidazóis/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Quinases/metabolismo , Quinolinas/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Antineoplásicos/administração & dosagem , Ácidos Borônicos/farmacologia , Bortezomib , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Sinergismo Farmacológico , Fase G1/efeitos dos fármacos , Humanos , Imidazóis/administração & dosagem , Melfalan/farmacologia , Mieloma Múltiplo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pirazinas/farmacologia , Quinolinas/administração & dosagem , Proteínas Recombinantes/farmacologia , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Serina-Treonina Quinases TOR
10.
Leuk Res ; 32(7): 1144-7, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18083229

RESUMO

Multiple myeloma is still not curable and drug combination strategies are currently being evaluated in order to achieve high remission rates with tolerable toxicity. Bortezomib has been shown to exert inhibitory effects on NFkappaB activity. NFkappaB in turn is known to be activated by cytokines, growth factors and by cellular adhesion to bone marrow stromal cells and represents an important mediator of primary and secondary drug resistance in multiple myeloma that confers to proliferation and survival. In this study we confirm that bortezomib sensitized MM cells to the DNA-damaging drugs melphalan and doxorubicin. Further, we demonstrate that the sole incubation of MM cells with melphalan or doxorubicin leads to a vast activation of NFkappaB activity. Additionally, we show that the co-incubation of bortezomib with melphalan or doxorubicin reduces activation of NFkappaB. These data suggest that the drug-sensitizing effect of bortezomib on MM cells is due to inhibition of melphalan- and doxorubicin-induced activation of NFkappaB activity. This study, therefore, supports the idea of combining a NFkappaB inhibitor with alkylating drugs in the therapy of multiple myeloma.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Mieloma Múltiplo/metabolismo , NF-kappa B/metabolismo , Ácidos Borônicos/farmacologia , Bortezomib , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Humanos , Melfalan/farmacologia , Mieloma Múltiplo/patologia , Pirazinas/farmacologia
11.
Eur J Haematol ; 80(1): 37-45, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18028419

RESUMO

OBJECTIVES: Despite high dose treatment regimes multiple myeloma (MM) disease is still not curable. Patients become resistant to cytotoxic drugs and die of disease progression. Therefore, besides new cytotoxic compounds drug sensitisers are urgently needed. METHODS: The MM cell lines U266, OPM-2, RPMI-8226 and NCI-H929 were incubated with the common anti-myeloma drugs like melphalan together with protein kinase C (PKC) inhibitors. Growth inhibition was measured using the water-soluble tetrazolium salt 4-[3-(4-iodophenyl)-2-(4-nitrophenyl)-2H-5-tetrazolio]-1,3-benzene disulfonate (WST-1 assay), and apoptosis was determined by flow cytometry after staining with fluorescein isothiocyanate-labeled annexin V (annexin-V-FITC) and propidium iodide. Intracellular signalling was shown by western blotting. RESULTS: In this study we show that the combination of melphalan or doxorubicin with a PKC inhibitor, Gö6976 or enzastaurin, strongly increases cell toxicity. Increase of cytotoxicity is shown to be due to increased induction of apoptosis. Furthermore, we show that the protective effect of human bone marrow stromal cells (hBMSC) is abrogated by the PKC inhibitors. Finally, western blotting experiments revealed that incubation of myeloma cells with cytotoxic drugs like melphalan or doxorubicin leads to increased phosphorylation and therefore degradation of inhibitor of nuclear factor kappa B (IkappaB) and release of nuclear factor kappa B (NFkappaB). In contrast, enzastaurin inhibits phosphorylation of IkappaB. CONCLUSIONS: We conclude that the combination of conventional drugs and PKC inhibitors might be very effective and represents a new strategy in the treatment of MM.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Mieloma Múltiplo/tratamento farmacológico , Mutagênicos/farmacologia , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Medula Óssea/patologia , Carbazóis/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Doxorrubicina/farmacologia , Sinergismo Farmacológico , Humanos , Indóis/farmacologia , Melfalan/farmacologia , Mieloma Múltiplo/patologia
12.
J Cancer Res Clin Oncol ; 134(2): 245-53, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17653574

RESUMO

PURPOSE: Multiple myeloma is a fatal hematological disease caused by malignant transformation of plasma cells. Bendamustine has been proven to be a potent alternative to melphalan in phase 3 studies, yet its molecular mode of action is still poorly understood. METHODS: The four-myeloma cell lines NCI-H929, OPM-2, RPMI-8226, and U266 were cultured in vitro. Apoptosis was measured by flow cytometry after annexin V FITC and propidium iodide staining. Cell cycle distribution of cells was determined by DNA staining with propidium iodide. Intracellular levels of (phosphorylated) proteins were determined by western blot. RESULTS: We show that bendamustine induces apoptosis with an IC50 of 35-65 mug/ml and with cleavage of caspase 3. Incubation with 10-30 mug/ml results in G2 cell cycle arrest in all four-cell lines. The primary DNA-damage signaling kinases ATM and Chk2, but not ATR and Chk1, are activated. The Chk2 substrate Cdc25A phosphatase is degraded and Cdc2 is inhibited by inhibitory phosphorylation of Tyr15 accompanied by increased cyclin B levels. Additionally, p53 activation occurs as phosphorylation of Ser15, the phosphorylation site for ATM. p53 promotes Cdc2 inhibition by upregulation of p21. Targeting of p38 MAPK by the selective inhibitor SB202190 significantly increases bendamustine induced apoptosis. Additionally, SB202190 completely abrogates G2 cell cycle arrest. CONCLUSION: Bendamustine induces ATM-Chk2-Cdc2-mediated G2 arrest and p53 mediated apoptosis. Inhibition of p38 MAPK augments apoptosis and abrogates G2 arrest and can be considered as a new therapeutic strategy in combination with bendamustine.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Proteínas de Ligação a DNA/metabolismo , Fase G2/efeitos dos fármacos , Mieloma Múltiplo/patologia , Compostos de Mostarda Nitrogenada/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Fosfatases cdc25/metabolismo , Apoptose/fisiologia , Proteínas Mutadas de Ataxia Telangiectasia , Cloridrato de Bendamustina , Western Blotting , Proliferação de Células/efeitos dos fármacos , Quinase do Ponto de Checagem 2 , Citometria de Fluxo , Fase G2/fisiologia , Humanos , Mieloma Múltiplo/metabolismo , Fosforilação , Tirosina/metabolismo
13.
Int J Oncol ; 31(4): 969-76, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17786331

RESUMO

Multiple myeloma (MM) is still an incurable disease and adhesion of MM cells to bone marrow stromal cells is one of the hallmarks of the disease. Lymphocyte function associated antigen 1 (LFA-1) is an adhesion molecule that mediates lymphocyte adhesion, but its role in MM is only poorly understood. The aim of the presented study was to improve knowledge on LFA-1 and associated pathways in MM for the development of molecular targeted therapies. We demonstrate that LFA-1 is expressed in U266, RPMI-8226, OPM-2, and NCI-H929 MM cell lines and in primary cells of eight tested patients. The LFA-1 inhibitor LFA878 induces apoptosis in all four cell lines as revealed by annexin V staining and caspase 3 cleavage. Apoptosis is not hampered by adhesion to stromal cells. Additionally, the soluble ligand, intracellular adhesion molecule 1 (ICAM-1), which is increased in the serum of MM patients, does not protect from melphalan-induced apoptosis. Western blots demonstrate downregulation of FAK, PI3-K, and Akt upon LFA878 treatment. Additionally, sequential inhibition of the pathway by simultaneous application of Src family kinase or PI3-K inhibitors significantly increases LFA878 induced apoptosis. We conclude that LFA-1/FAK/PI3-K/Akt is a survival pathway in MM and that targeted inhibition may provide new therapeutic options.


Assuntos
Apoptose/efeitos dos fármacos , Quinase 1 de Adesão Focal/metabolismo , Antígeno-1 Associado à Função Linfocitária/metabolismo , Mieloma Múltiplo/tratamento farmacológico , Naftalenos/farmacologia , Oxazinas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Caspases/metabolismo , Ciclo Celular/efeitos dos fármacos , Ciclinas/metabolismo , Regulação para Baixo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Immunoblotting , Antígeno-1 Associado à Função Linfocitária/química , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Células Tumorais Cultivadas/efeitos dos fármacos
14.
Exp Cell Res ; 313(16): 3592-603, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17669398

RESUMO

The role of adenosine monophosphate activated protein kinase (AMPK) in regulating multiple myeloma (MM) cell growth is not yet clear. In this study, we show that the AMPK activators 5-aminoimidazole-4-carboxamide riboside (AICAr) and D942 inhibit cell growth in MM cell lines. AICAr also induced an S-phase cell cycle arrest in all four tested cell lines and led to phosphorylation and thus activation of AMPK. Furthermore, the inhibition of a nucleoside transporter by nitrobenzyl-thio-9-beta-d-ribofuranosylpurine (NBTI), inhibition of the adenosine kinase by iodotubericidine and inhibition of AMPK by AMPKI Compound C reversed AICAr effects, indicating that the cellular effects of AICAr were mediated by AMPK. Activation of AMPK inhibited basal extracellular signal-regulated kinase (ERK), mammalian target of rapamycin (mTOR) and P70S6 kinase (P70S6K) as well as AKT phosphorylation, and blocked IL-6, IGF-1, and HS-5 stromal cell conditioned medium-induced increase of cell growth. Troglitazone, which has previously been shown to activate AMPK, similarly inhibited MM cell growth, activated AMPK, and decreased ERK and P70S6K phosphorylation. Our results suggest that activation of AMPK inhibits MM cell growth despite stimulation with IL-6, IGF-1, or HS-5 stromal cell conditioned medium and represents a potential new target in the therapy of MM.


Assuntos
Complexos Multienzimáticos/metabolismo , Mieloma Múltiplo/enzimologia , Mieloma Múltiplo/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases Ativadas por AMP , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cromanos/farmacologia , Meios de Cultivo Condicionados , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/farmacologia , Interleucina-6/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Quinases/metabolismo , Ribonucleotídeos/farmacologia , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Fase S/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Células Estromais/efeitos dos fármacos , Serina-Treonina Quinases TOR , Tiazolidinedionas/farmacologia , Troglitazona
15.
Anticancer Drugs ; 18(4): 405-10, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17351392

RESUMO

In this study, we show that adenosine monophosphate-activated protein kinase (AMPK) is expressed and activated in multiple myeloma cells. The inhibition of AMPK induced growth arrest and reduction of cell viability in the cell viability assay using the water-soluble tetrazolium salt 4-[3-(4-iodophenyl)-2-(4-nitrophenyl)-2H-5-tetrazolio]-1,3-benzene disulfonate (WST-1 assay). Induction of apoptosis was determined by annexin-V and propidium iodide staining. The prevention of apoptosis using the pancaspase inhibitor ZVAD-fmk and caspase-3 cleavage upon incubation with the AMPK inhibitor (AMPKI) is shown. Furthermore, incubation of myeloma cells with AMPKI resulted in the downregulation of pAMPK, Mcl-1 and Bcl-xL. Coincubation of AMPKI and melphalan led to a strong additional increase of apoptosis in myeloma cells. We conclude that AMPKI has a strong antimyeloma activity in vitro and represents a new targeted strategy in the treatment of multiple myeloma.


Assuntos
Antineoplásicos , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Mieloma Múltiplo/tratamento farmacológico , Anexinas/biossíntese , Antineoplásicos Alquilantes/farmacologia , Western Blotting , Neoplasias Encefálicas/patologia , Sobrevivência Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Humanos , Indicadores e Reagentes , Melfalan/farmacologia , Mieloma Múltiplo/patologia , Proteína de Sequência 1 de Leucemia de Células Mieloides , Proteínas de Neoplasias/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteína bcl-X/biossíntese
16.
Blood ; 104(4): 1120-6, 2004 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15090460

RESUMO

The t(11;14)(q13;q32) is the most common translocation in multiple myeloma (MM), resulting in up-regulation of cyclin D1. We used a segregation fluorescence in situ hybridization (FISH) assay to detect t(11;14) breakpoints in primary MM cases and real-time reverse transcriptase-polymerase chain reaction (RT-PCR) to quantify cyclin D1 and MYEOV (myeloma overexpressed) expression, another putative oncogene located on chromosome 11q13. High levels of cyclin D1 mRNA (cyclin D1/TBP [TATA box binding protein] ratio > 95) were found exclusively in the presence of a t(11;14) translocation (11/48 cases; P <.00001). In addition, a subgroup of MM cases (15/48) with intermediate to low cyclin D1 mRNA (cyclin D1/TBP ratio between 2.3 and 20) was identified. FISH analysis ruled out a t(11; 14) translocation and 11q13 amplification in these cases; however, in 13 of 15 patients a chromosome 11 polysomy was demonstrated (P <.0001). These results indicate an effect of gene dosage as an alternative mechanism of cyclin D1 deregulation in MM. The absence of chromosome 11 abnormalities in 2 of 15 patients with intermediate cyclin D1 expression supports that there are presumably other mechanism(s) of cyclin D1 deregulation in MM patients. Our data indicate that deregulation of MYEOV is not favored in MM and further strengthens the role of cyclin D1 overexpression in lymphoid malignancies with a t(11;14)(q13;q32) translocation.


Assuntos
Ciclina D1/genética , Mieloma Múltiplo/genética , RNA Mensageiro/análise , Cromossomos Humanos Par 11 , Cromossomos Humanos Par 14 , Ciclina D1/análise , Dosagem de Genes , Humanos , Hibridização in Situ Fluorescente , Mieloma Múltiplo/patologia , Proteínas Oncogênicas/análise , Proteínas Oncogênicas/genética , Osteólise/patologia , Proteínas Proto-Oncogênicas , Translocação Genética
17.
Lab Invest ; 83(1): 107-14, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12533691

RESUMO

Discordant bone marrow (BM) involvement in patients with a diagnosis of large-cell non-Hodgkin's lymphoma (NHL) is characterized by marrow infiltrates predominantly composed of small lymphoid cell, cytologically compatible with low-grade NHL. Although this phenomenon is well described morphologically, molecular data concerning the relationship of the two lesions are lacking. The aim of the study was to investigate the clonal relationship of discordant lymphoma manifestations by using immunoglobulin heavy chain gene (IgH), as well as bcl-2 rearrangements, as molecular markers. IgH rearrangements were amplified by PCR with consensus primers directed against framework regions 3 or 2 (FR3 and FR2), followed by automated fragment length analysis and sequencing in selected cases. Rearrangements of the bcl-2 gene were identified with primers against the major breakpoint region. Small BM infiltrates were isolated by laser capture microdissection. In addition, immunohistochemistry was performed on paraffin sections using antibodies against CD3, CD10, CD20, bcl-2, bcl-6, p53, and the Ki67 antigen. Paraffin-embedded tissues of 21 cases diagnosed as diffuse large B-cell lymphoma (DLBCL) with discordant BM involvement and no previous history of low-grade B-cell NHL were analyzed. After review of immunohistochemical stains, 5 cases were excluded either as concordant BM infiltrates by large-cell lymphoma with abundant reactive T-cells (2 cases) or as benign, reactive lymphoid infiltrates (3 cases), as confirmed by a polyclonal pattern in the IgH analysis. Of the remaining 16 cases, a common clonal origin was confirmed in 8 cases by the presence of an identical clonal IgH rearrangement or bcl-2 rearrangement. In 4 cases, identification of distinct IgH or bcl-2 rearrangements gave evidence for the presence of two clonally unrelated neoplasms. The remaining 4 cases were not evaluable for technical reasons. Morphological, phenotypical, and molecular findings were compatible with a lymphoma of germinal center origin in the majority of cases. However, in 4 cases, flow cytometric analysis of the BM infiltrates revealed a B-cell chronic lymphocytic leukemia phenotype. Two of these cases were clonally related to the DLBCL and thus represented Richter's transformation. In summary, discordant BM infiltrates in DLBCL represent a heterogeneous group of disorders, encompassing cases with a clonally related, clinically occult small-cell component, as well as cases with two clonally distinct, unrelated B-cell neoplasms presenting synchronously at different locations.


Assuntos
Medula Óssea/patologia , Linfoma de Células B/patologia , Linfoma Difuso de Grandes Células B/patologia , Adulto , Idoso , Feminino , Humanos , Imuno-Histoquímica , Imunofenotipagem , Linfoma de Células B/classificação , Linfoma de Células B/imunologia , Linfoma Difuso de Grandes Células B/classificação , Linfoma Difuso de Grandes Células B/imunologia , Masculino , Pessoa de Meia-Idade
18.
Kidney Int ; 61(2): 570-8, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11849398

RESUMO

BACKGROUND: Vascular endothelial growth factor (VEGF) was recently found in peritoneal effluents of peritoneal dialysis (PD) patients. It was suggested that human peritoneal mesothelial cells (HMC) contribute to the intraperitoneal production of VEGF, which may augment vascular permeability, vasodilation and neoangiogenesis in the peritoneal membrane. The present study was designed to assess the influence of proinflammatory cytokines, thrombin, d-glucose and glycated albumin in the regulation of VEGF synthesis in primary HMC cultures. METHODS: VEGF antigen concentrations were measured in the cell supernatant by ELISA and VEGF mRNA expression was evaluated by real time RT-PCR. RESULTS: Incubation of HMC with interleukin-1alpha (IL-1alpha; 10 to 100 U/mL), tumor necrosis factor-alpha (TNF-alpha; 500 to 1000 U/mL) or thrombin (1 to 10 U/mL) resulted in a time (24 to 72 hours) and concentration dependent increase in VEGF synthesis. In contrast, d-glucose (30 to 90 mmol/L), which is commonly used as an osmotic agent in peritoneal dialysis, was not able to up-regulate VEGF expression. High glucose levels even decreased VEGF production. However, exposure of HMC to Amadori-modified glycated albumin, which is generated in the peritoneal cavity in the presence of glucose-based dialysis solutions, resulted in a dose and time dependent increase in VEGF mRNA expression and antigen secretion. CONCLUSIONS: These results demonstrate, to our knowledge for the first time, that glycated serum albumin, not glucose, increases VEGF production in HMC. HMC play an important role as a source of intraperitoneal VEGF synthesis, and VEGF expression also is up-regulated in the presence of proinflammatory cytokines and thrombin. Additionally, these results confirm clinical data that the continuous exposure of the peritoneal membrane to glucose-based dialysis solutions is an important stimulus for VEGF expression. However, it is not glucose per se, but nonenzymatic glycation products like glycated albumin that up-regulate VEGF expression.


Assuntos
Fatores de Crescimento Endotelial/genética , Linfocinas/genética , Peritônio/citologia , Peritônio/metabolismo , Antineoplásicos/farmacologia , Células Cultivadas , Fatores de Crescimento Endotelial/metabolismo , Epitélio/metabolismo , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Glucose/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Hemostáticos/farmacologia , Humanos , Interleucina-1/farmacologia , Falência Renal Crônica/metabolismo , Falência Renal Crônica/terapia , Linfocinas/metabolismo , Diálise Peritoneal , RNA Mensageiro/análise , Albumina Sérica/farmacologia , Trombina/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular , Albumina Sérica Glicada
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...