Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Ann Anat ; 246: 152023, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36400339

RESUMO

Porphyromonas gingivalis lipopolysaccharide (PG-LPS) is an important virulence factor potentially contributing to periodontal tissue destruction. Toll-like receptor 4 (Tlr4) is a key mediator of NF-kB activation during pathogen recognition. Previous work using Tlr4-specific antibodies demonstrated a partial neutralization of PG-LPS effects on murine cementoblasts, which can affect cell function and regulate gene expression of osteoclastic markers. PG-LPS also potentially influence the inflammation process and the resorption of mineralized tissues. Yet, such inflammatory responses and cell signaling events remain to be characterized at the protein level. We thus investigated the effect of 1 and 10 µg/ml of PG-LPS, respectively, on cell morphology, cell viability, and selected key downstream molecules of the Tlr4 signaling cascade in cementoblasts. High concentrations of PG-LPS (10 µg/ml) significantly reduced cell viability after 48 h. Upon PG-LPS-stimulation, Tlr4 was significantly downregulated. Equally, IκBα, a downstream molecule, was downregulated in terms of phosphorylation and protein production. Furthermore, downstream signaling kinases, like serine/threonine kinase phospho-AKT and the mitogen-activated protein kinase (MAPK)-family, specifically phospho-ERK1/2, were significantly upregulated under high PG-LPS-concentrations. We provide new insights into PG-LPS-triggered intracellular signaling pathways in cementoblasts and thus deliver a basis for further research in PG-mediated periodontal inflammation.


Assuntos
Lipopolissacarídeos , Porphyromonas gingivalis , Proteínas Proto-Oncogênicas c-akt , Receptor 4 Toll-Like , Animais , Camundongos , Cemento Dentário/metabolismo , Inflamação , Lipopolissacarídeos/toxicidade , Fosforilação , Porphyromonas gingivalis/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor 4 Toll-Like/metabolismo
2.
J Cell Mol Med ; 26(23): 5832-5845, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36377725

RESUMO

Metastatic disease is the leading cause of death in children suffering from medulloblastoma and a major treatment challenge. The evidence of leptomeningeal dissemination defines the most aggressive tumours and is associated with increased mortality; thus, inhibition of migration as a factor involved in the process of metastatic disease is fundamental for the treatment and prevention of metastatic dissemination. Targeting the small Rho GTPases Rac1 has been shown to effectively impair medulloblastoma cell migration in vitro. Yet clinically applicable selective Rac1 inhibitors are still lacking. In view of the pertinent oncogenic role of the PI3K signalling cascade and tyrosine kinase-mediated signalling pathways in medulloblastoma, we explored clinically available targeted therapeutics to this effect. Here, we show that Rac1 is expressed in both the cytoplasm and nucleus in the medulloblastoma cell lines Daoy and MEB-Med-8A representative of two high risk medulloblastoma entities. We demonstrate that activated Rac1 is subject to substantial downmodulation following administration of the clinically available inhibitor of the PI3K pathway Pictilisib (GDC-0941) and the multityrosine kinase inhibitors Pazopanib and Sorafenib. The application of those drugs was associated with reduced mobility of the medulloblastoma cells and alterations of the actin skeleton. Of note, PI3K inhibition reveals the strongest anti-migratory effect in Daoy cells. Thus, our in vitro observations provide new insights into different strategies of blocking Rac1 and inhibiting migration in medulloblastoma employing clinically available agents paving the way for confirmatory studies in in vivo models.


Assuntos
Neoplasias Cerebelares , Meduloblastoma , Proteínas rac1 de Ligação ao GTP , Humanos , Linhagem Celular Tumoral , Movimento Celular , Neoplasias Cerebelares/tratamento farmacológico , Meduloblastoma/tratamento farmacológico , Meduloblastoma/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas rac1 de Ligação ao GTP/metabolismo , Sorafenibe/farmacologia , Sorafenibe/uso terapêutico
3.
Sci Rep ; 12(1): 14970, 2022 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-36056072

RESUMO

Xanthohumol (XN) is a prenylated plant polyphenol that naturally occurs in hops and its products, e.g. beer. It has shown to have anti-inflammatory and angiogenesis inhibiting effects and it prevents the proliferation of cancer cells. These effects could be in particular interesting for processes within the periodontal ligament, as previous studies have shown that orthodontic tooth movement is associated with a sterile inflammatory reaction. Based on this, the study evaluates the anti-inflammatory effect of XN in cementoblasts in an in vitro model of the early phase of orthodontic tooth movement by compressive stimulation. XN shows a concentration-dependent influence on cell viability. Low concentrations between 0.2 and 0.8 µM increase viability, while high concentrations between 4 and 8 µM cause a significant decrease in viability. Compressive force induces an upregulation of pro-inflammatory gene (Il-6, Cox2, Vegfa) and protein (IL-6) expression. XN significantly reduces compression related IL-6 protein and gene expression. Furthermore, the expression of phosphorylated ERK and AKT under compression was upregulated while XN re-established the expression to a level similar to control. Accordingly, we demonstrated a selective anti-inflammatory effect of XN in cementoblasts. Our findings provide the base for further examination of XN in modulation of inflammation during orthodontic therapy and treatment of periodontitis.


Assuntos
Cemento Dentário , Propiofenonas , Anti-Inflamatórios/farmacologia , Flavonoides/farmacologia , Humanos , Inflamação/tratamento farmacológico , Interleucina-6 , Propiofenonas/farmacologia
4.
Int J Mol Sci ; 23(15)2022 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-35897640

RESUMO

Mechanical compression simulating orthodontic tooth movement in in vitro models induces pro-inflammatory cytokine expression in periodontal ligament (PDL) cells. Our previous work shows that TLR4 is involved in this process. Here, primary PDL cells are isolated and characterized to better understand the cell signaling downstream of key molecules involved in the process of sterile inflammation via TLR4. The TLR4 monoclonal blocking antibody significantly reverses the upregulation of phospho-AKT, caused by compressive force, to levels comparable to controls by inhibition of TLR4. Phospho-ERK and phospho-p38 are also modulated in the short term via TLR4. Additionally, moderate compressive forces of 2 g/cm2, a gold standard for static compressive mechanical stimulation, are not able to induce translocation of Nf-kB and phospho-ERK into the nucleus. Accordingly, we demonstrated for the first time that TLR4 is also one of the triggers for signal transduction under compressive force. The TLR4, one of the pattern recognition receptors, is involved through its specific molecular structures on damaged cells during mechanical stress. Our findings provide the basis for further research on TLR4 in the modulation of sterile inflammation during orthodontic therapy and periodontal remodeling.


Assuntos
Ligamento Periodontal , Receptor 4 Toll-Like , Técnicas de Movimentação Dentária , Células Cultivadas , Humanos , Inflamação/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Ligamento Periodontal/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Estresse Mecânico , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
5.
Basic Clin Pharmacol Toxicol ; 130(1): 132-140, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34740282

RESUMO

In genome-wide association studies, the CYP2C8 gene locus has been reported to be associated with bisphosphonate-related osteonecrosis of the jaw, a severe devastating side effect of antiresorptive bone treatment. The aim of this study was to elucidate the putative pathomechanism explaining the association between the genetic polymorphism with the alleles CYP2C8*2 and *3 causing low CYP2C8 activity, and disturbed periodontal remodelling in periodontal fibroblasts cultured from patients undergoing orthodontic treatment. CYP2C8 activity, enzyme expression and substrate metabolism were detected in human periodontal fibroblast cultures. Zoledronic acid caused enhanced reactive oxygen species (ROS) production in periodontal fibroblasts, which was enhanced by arachidonic acid as inflammatory signal. Enhanced bisphosphonate-induced uncoupling of the CYP2C8 enzyme was detected in the variant allele (CYP2C8*3) with the result of increased H2 O2 production and lowered substrate oxidation. Conversely, substrate (amodiaquine) addition led to decreased H2 O2 production in isolated CYP2C8 enzymes, but in CYP2C8*3 enzyme, increased H2 O2 was still detected, especially in presence of arachidonic acid. CYP2C8 variants leading to decreased enzyme activity in substrate oxidation may enhance ROS production by reaction uncoupling, and thus, contribute to difficulties in orthodontic treatment and the risk of side effects of antiresorptive drugs.


Assuntos
Citocromo P-450 CYP2C8/genética , Fibroblastos/efeitos dos fármacos , Ligamento Periodontal/efeitos dos fármacos , Ácido Zoledrônico/toxicidade , Alelos , Amodiaquina/farmacologia , Ácido Araquidônico/metabolismo , Conservadores da Densidade Óssea/toxicidade , Células Cultivadas , Fibroblastos/citologia , Estudo de Associação Genômica Ampla , Humanos , Peróxido de Hidrogênio/metabolismo , Ortodontia , Oxirredução , Ligamento Periodontal/citologia , Polimorfismo Genético , Espécies Reativas de Oxigênio/metabolismo
6.
Ann Anat ; 234: 151668, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33400981

RESUMO

Cementoblasts, located on the tooth root surface covered with cementum, are considered to have tooth protecting abilities. They prevent tissue damage and secure teeth anchorage inside the periodontal ligament during mechanical stress. However, the involvement of cementoblasts in mechanical compression induced periodontal remodeling needs to be identified and better understood. Here, we investigated the effect of static compressive stimulation, simulating the compression side of orthodontic force and cell confluence on a murine cementoblast cell line (OC/CM). The influence of cell confluence in cementoblast cells was analyzed by MTS assay and immunostaining. Furthermore, mRNA and protein expression were investigated by real-time RT-PCR and western blotting at different confluence grades and after mechanical stimulation. We observed that cementoblast cell proliferation increases with increasing confluence grades, while cell viability decreases in parallel. Gene expression of remodeling markers is regulated by compressive force. In addition, cementoblast confluence plays a crucial role in this regulation. Confluent cementoblasts show a significantly higher basal expression of Bsp, Osterix, Alpl, Vegfa, Mmp9, Tlr2 and Tlr4 compared to sub-confluent cells. After compressive force of 48 h at 60% confluence, an upregulation of Bsp, Osterix, Alpl, Vegf and Mmp9 is observed. In contrast, at high confluence, all analyzed genes were downregulated through mechanical stress. We also proved a regulation of ERK, phospho-ERK and phospho-AKT dependent on compressive force. In summary, our findings provide evidence that cementoblast physiology and metabolism is highly regulated in a cell confluence-dependent manner and by mechanical stimulation.


Assuntos
Cemento Dentário , Proteínas Proto-Oncogênicas c-akt , Animais , Expressão Gênica , Camundongos , Ligamento Periodontal , Fosforilação
7.
Cancers (Basel) ; 12(5)2020 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-32438548

RESUMO

Outcome in high-risk patients with refractory or relapsed germ cell tumours (GCT) remains poor. Novel strategies enhancing therapeutic efficacy whilst limiting therapeutic burden are warranted, yet immunotherapy approaches geared towards activating endogenous antitumor responses have not been successful thus far. Redirection of cytotoxic effector cells by bispecific antibodies represents a promising approach in this setting. We demonstrate that the Epithelial Cell Adhesion Molecule (EpCAM) is broadly expressed in GCT cell lines of different histologic origin including seminoma, choriocarcinoma (CHC), and embryonal carcinoma (EC). In these GCT lines of variable EpCAM surface expression, targeting T cells by the prototypic bispecific EpCAM/CD3-antibody (bAb) Catumaxomab together with natural killer (NK) cell engagement via the Fc domain promotes profound cytotoxicity across a broad range of antibody dilutions. In contrast, tumor cell lysis mediated by either immune cell subset alone is influenced by surface density of the target antigen. In the CHC line JAR, NK cell-dependent cytotoxicity dominates, which may be attributed to differential surface expression of immunomodulatory proteins such as MHC-I, CD24, and Fas receptors on CHC and EC. In view of redirecting T cell therapy mediated by bispecific antibodies, such differences in GCT immunophenotype potentially favoring immune escape are worth further investigation.

8.
Clin Oral Investig ; 24(4): 1411-1419, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31691860

RESUMO

OBJECTIVE: Biomechanical orthodontics loading of the periodontium initiates a cascade of inflammatory signaling events that induce periodontal remodeling and finally facilitate orthodontic tooth movement. Pattern recognition receptors such as toll-like receptors (TLRs) have been well characterized for their ability to induce the activation of inflammatory, immunomodulatory cytokines. Here, we examined whether the cellular response of human periodontal ligament (hPDL) cells to mechanical stress involves TLR-4 signaling in vitro. MATERIALS AND METHODS: Confluent hPDL cells were cultured in the presence of 5 µg/ml TLR-4 antibody (TLR-4ab) for 1 h prior to the induction of compressive forces by the use of round glass plates for 24 h. At harvest, interleukin-6 and interleukin-8 (IL-6, IL-8) mRNA and protein expression were analyzed by real-time PCR and ELISA. The immunomodulatory role of mechanical cell stress and TLR-4 signaling was addressed in co-culture experiments of hPDL and THP-1 cells targeting monocyte adhesion and by culturing osteoclastic precursors (RAW 264.7) in the presence of the conditioned medium of hPDL cells that had been mechanically loaded before. RESULTS: Basal expression of IL-6 and IL-8 was not affected by TLR-4ab, but increased significantly upon mechanical loading of hPDL cells. When cells were mechanically stressed in the presence of TLR-4ab, the effect seen for loading alone was markedly reduced. Likewise, monocyte adhesion and osteoclastic differentiation were enhanced significantly by mechanical stress of hPDL cells and this effect was partially inhibited by TLR-4ab. CONCLUSIONS: The results of the present study indicate a proinflammatory and immunomodulatory influence of mechanical loading on hPDL cells. Intracellular signaling involves a TLR-4-dependent pathway. CLINICAL RELEVANCE: These findings hold out the prospect of interfering with the cellular response to mechanical cell stress in order to minimize undesired side effects of orthodontic tooth movement.


Assuntos
Interleucina-6/imunologia , Interleucina-8/imunologia , Ligamento Periodontal/citologia , Estresse Mecânico , Receptor 4 Toll-Like/metabolismo , Animais , Diferenciação Celular , Humanos , Camundongos , Osteoclastos/citologia , Células RAW 264.7 , Transdução de Sinais , Células THP-1
9.
J Cell Mol Med ; 22(4): 2153-2161, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29377550

RESUMO

Aberrant receptor kinase signalling and tumour neovascularization are hallmarks of medulloblastoma development and are both considered valuable therapeutic targets. In addition to VEGFR1/2, expression of PDGFR α/ß in particular has been documented as characteristic of metastatic disease correlating with poor prognosis. Therefore, we have been suggested that the clinically approved multi-kinase angiogenesis inhibitor Axitinib, which specifically targets these kinases, might constitute a promising option for medulloblastoma treatment. Indeed, our results delineate anti-neoplastic activity of Axitinib in medulloblastoma cell lines modelling the most aggressive c-myc-amplified Non-WNT/Non-SHH and SHH-TP53-mutated tumours. Exposure of medulloblastoma cell lines to Axitinib results in marked inhibition of proliferation and profound induction of cell death. The differential efficacy of Axitinib is in line with target expression of medulloblastoma cells identifying VEGFR 1/2, PDGFR α/ß and c-kit as potential markers for drug application. The high specificity of Axitinib and the consequential low impact on the haematopoietic and immune system render this drug ideal multi-modal treatment approaches. In this context, we demonstrate that the clinically available PI3K inhibitor GDC-0941 enhances the anti-neoplastic efficacy of Axitinib against c-myc-amplified medulloblastoma. Our findings provide a rational to further evaluate Axitinib alone and in combination with other therapeutic agents for the treatment of most aggressive medulloblastoma subtypes.


Assuntos
Axitinibe/farmacologia , Indazóis/farmacologia , Meduloblastoma/patologia , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Sulfonamidas/farmacologia , Antineoplásicos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Células Clonais , Sinergismo Farmacológico , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fatores de Risco , Fator de Transcrição STAT3/metabolismo , Estados Unidos , United States Food and Drug Administration
10.
J Cell Mol Med ; 22(1): 568-575, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28941150

RESUMO

Prognosis in patients suffering from high-risk, refractory and relapsed germ cell tumours (GCT) often comprising of CD30-positive embryonal carcinoma (EC) components remains poor. Thus, novel treatment strategies are warranted. The antibody-drug conjugate (ADC) brentuximab vedotin delivers the potent antimitotic drug monomethyl auristatin E (MMAE) to CD30-expressing tumour cells. After CD30 binding, internalization and intracellular linker cleavage cytotoxic MMAE can efflux and eradicate neighbouring CD30-negative cells. To analyse cytotoxicity and a potential bystander effect of brentuximab vedotin in GCT, we established an in vitro coculture model mimicking GCT of heterogeneous CD30 positivity and measured cell viability, proliferation and apoptosis after exposure to brentuximab vedotin and unbound MMAE by MTS- and flow cytometry-based CFSE/Hoechst assay. CD30 expression being assessed by quantitative RT-PCR and immunohistochemistry was apparent in all EC cell lines with different intensity. Brentuximab vedotin abrogates cell viability of CD30-positive GCT27 EC line exerting marked time-dependent antiproliferative and pro-apoptotic activity. CD30-negative JAR cultured alone barely responds to brentuximab vedotin, while in coculture with GCT27 brentuximab vedotin induces clear dose-dependent cytotoxicity. Cellular proliferation and cell death are significantly enhanced in CD30-negative JAR cocultured with CD30-positive GCT27 compared to JAR cultured alone in proof of substantial bystander activity of brentuximab vedotin in CD30-negative GCT. We present first evidence that in an in vitro model mimicking GCT of heterogeneous histology, brentuximab vedotin exerts potent antiproliferative and pro-apoptotic activity against both CD30-positive as well as CD30-negative GCT subsets. Our results strongly support translational efforts to evaluate clinical efficacy of brentuximab vedotin in high-risk GCT of heterogeneous CD30 positivity.


Assuntos
Apoptose/efeitos dos fármacos , Imunoconjugados/farmacologia , Antígeno Ki-1/metabolismo , Neoplasias Embrionárias de Células Germinativas/patologia , Brentuximab Vedotin , Efeito Espectador/efeitos dos fármacos , Contagem de Células , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular , Técnicas de Cocultura , Humanos , Antígeno Ki-1/genética , Oligopeptídeos/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Tempo
11.
Oncotarget ; 8(29): 46915-46927, 2017 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-28159923

RESUMO

Medulloblastoma is comprised of at least four molecular subgroups with distinct clinical outcome (WHO classification 2016). SHH-TP53-mutated as well as MYC-amplified Non-WNT/Non-SHH medulloblastoma show the worst prognosis.Here we present evidence that single application of the multi-kinase inhibitor Vandetanib displays anti-neoplastic efficacy against cell lines derived from high-risk SHH-TP53-mutated and MYC-amplified Non-WNT/Non-SHH medulloblastoma. The narrow target spectrum of Vandetanib along with a favourable toxicity profile renders this drug ideal for multimodal treatment approaches. In this context our investigation documents that Vandetanib in combination with the clinically available PI3K inhibitor GDC-0941 leads to enhanced cytotoxicity against MYC-amplified and SHH-TP53-mutated medulloblastoma. In line with these findings we show for MYC-amplified medulloblastoma a profound reduction in activity of the oncogenes STAT3 and AKT. Furthermore, we document that Vandetanib and the standard chemotherapeutic Etoposide display additive anti-neoplastic efficacy in the investigated medulloblastoma cell lines that could be further enhanced by PI3K inhibition. Of note, the combination of Vandetanib, GDC-0941 and Etoposide results in MYC-amplified and SHH-TP53-mutated cell lines in complete loss of cell viability. Our findings therefore provide a rational to further evaluate Vandetanib in combination with PI3K inhibitors as well as standard chemotherapeutics in vivo for the treatment of most aggressive medulloblastoma variants.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Cerebelares/metabolismo , Meduloblastoma/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Piperidinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Neoplasias Cerebelares/tratamento farmacológico , Neoplasias Cerebelares/patologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Etoposídeo/farmacologia , Humanos , Indazóis/farmacologia , Meduloblastoma/tratamento farmacológico , Meduloblastoma/patologia , Terapia de Alvo Molecular , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/farmacologia
12.
Oncotarget ; 6(2): 802-13, 2015 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-25596739

RESUMO

Deregulation of the Phosphoinositide 3-kinase (PI3K)/AKT signalling network is a hallmark of oncogenesis. Also medulloblastoma, the most common malignant brain tumor in children, is characterized by high levels of AKT phosphorylation and activated PI3K signalling in medulloblastoma is associated with enhanced cellular motility, survival and chemoresistency underscoring its role of as a potential therapeutic target. Here we demonstrate that GDC-0941, a highly specific PI3K inhibitor with good clinical tolerability and promising anti-neoplastic activity in adult cancer, also displays anti-proliferative and pro-apoptotic effects in pediatric human medulloblastoma cell lines. Loss in cell viability is accompanied by reduced phosphorylation of AKT, a downstream target of PI3K. Furthermore, we show that GDC-0941 attenuates the migratory capacity of medulloblastoma cells and targets subpopulations expressing the stem cell marker CD133. GDC-0941 also synergizes with the standard medulloblastoma chemotherapeutic etoposide. In an orthotopic xenograft model of the most aggressive human medulloblastoma variant we document that oral adminstration of GDC-0941 impairs tumor growth and significantly prolongs survival. These findings provide a rational to further investigate GDC-0941 alone and in combination with standard chemotherapeutics for medulloblastoma treatment.


Assuntos
Neoplasias Cerebelares/tratamento farmacológico , Indazóis/farmacologia , Meduloblastoma/tratamento farmacológico , Sulfonamidas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias Cerebelares/enzimologia , Neoplasias Cerebelares/patologia , Células HEK293 , Humanos , Técnicas In Vitro , Meduloblastoma/enzimologia , Meduloblastoma/patologia , Camundongos , Inibidores de Fosfoinositídeo-3 Quinase , Distribuição Aleatória , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Oncotarget ; 5(16): 7149-61, 2014 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-25216529

RESUMO

Regardless of the recent advances in cytotoxic therapies, 30% of children diagnosed with medulloblastoma. succumb to the disease. Therefore, novel therapeutic approaches are warranted. Here we demonstrate that Pazopanib a clinically approved multi-kinase angiogenesis inhibitor (MKI) inhibits proliferation and apoptosis in medulloblastoma cell lines. Moreover, Pazopanib profoundly attenuates medulloblastoma cell migration, a prerequisite for tumor invasion and metastasis. In keeping with the observed anti-neoplastic activity of Pazopanib, we also delineate reduced phosphorylation of the STAT3 protein, a key regulator of medulloblastoma proliferation and cell survival. Finally, we document profound in vivo activity of Pazopanib in an orthotopic mouse model of the most aggressive c-myc amplified human medulloblastoma variant. Pazopanib reduced the growth rate of intracranial growing medulloblastoma and significantly prolonged the survival. Furthermore, to put these results into a broader perspective we analysed Pazopanib side by side with the MKI Sorafenib. Both compounds share a similar target profile but display different pharmacodynamics and pharmacokinetics with distinct cytotoxic activity in different tumor entities. Thus, we identified Pazopanib as a new promising candidate for a rational clinical assessment for targeted paediatric medulloblastoma therapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias Cerebelares/tratamento farmacológico , Meduloblastoma/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias Cerebelares/enzimologia , Neoplasias Cerebelares/patologia , Relação Dose-Resposta a Droga , Feminino , Humanos , Indazóis , Masculino , Meduloblastoma/enzimologia , Meduloblastoma/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Niacinamida/administração & dosagem , Niacinamida/análogos & derivados , Niacinamida/farmacologia , Compostos de Fenilureia/administração & dosagem , Compostos de Fenilureia/farmacologia , Inibidores de Proteínas Quinases/administração & dosagem , Pirimidinas/administração & dosagem , Pirimidinas/farmacologia , Sorafenibe , Sulfonamidas/administração & dosagem , Sulfonamidas/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA