Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Genes Dev ; 24(16): 1731-45, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20713517

RESUMO

Human solid tumors frequently have pronounced heterogeneity of both neoplastic and normal cells on the histological, genetic, and gene expression levels. While current efforts are focused on understanding heterotypic interactions between tumor cells and surrounding normal cells, much less is known about the interactions between and among heterogeneous tumor cells within a neoplasm. In glioblastoma multiforme (GBM), epidermal growth factor receptor gene (EGFR) amplification and mutation (EGFRvIII/DeltaEGFR) are signature pathogenetic events that are invariably expressed in a heterogeneous manner. Strikingly, despite its greater biological activity than wild-type EGFR (wtEGFR), individual GBM tumors expressing both amplified receptors typically express wtEGFR in far greater abundance than the DeltaEGFR lesion. We hypothesized that the minor DeltaEGFR-expressing subpopulation enhances tumorigenicity of the entire tumor cell population, and thereby maintains heterogeneity of expression of the two receptor forms in different cells. Using mixtures of glioma cells as well as immortalized murine astrocytes, we demonstrate that a paracrine mechanism driven by DeltaEGFR is the primary means for recruiting wtEGFR-expressing cells into accelerated proliferation in vivo. We determined that human glioma tissues, glioma cell lines, glioma stem cells, and immortalized mouse Ink4a/Arf(-/-) astrocytes that express DeltaEGFR each also express IL-6 and/or leukemia inhibitory factor (LIF) cytokines. These cytokines activate gp130, which in turn activates wtEGFR in neighboring cells, leading to enhanced rates of tumor growth. Ablating IL-6, LIF, or gp130 uncouples this cellular cross-talk, and potently attenuates tumor growth enhancement. These findings support the view that a minor tumor cell population can potently drive accelerated growth of the entire tumor mass, and thereby actively maintain tumor cell heterogeneity within a tumor mass. Such interactions between genetically dissimilar cancer cells could provide novel points of therapeutic intervention.


Assuntos
Receptores ErbB/genética , Receptores ErbB/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioblastoma/fisiopatologia , Mutação/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular/fisiologia , Receptor gp130 de Citocina/metabolismo , Citocinas/metabolismo , Glioblastoma/genética , Glioma/fisiopatologia , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Fator Inibidor de Leucemia/genética , Fator Inibidor de Leucemia/metabolismo , Ligantes , Camundongos , Camundongos Nus , Células-Tronco Neoplásicas/patologia , Regulação para Cima
2.
Life Sci Alliance ; 6(8)2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37169593

RESUMO

The epidermal growth factor receptor (EGFR) has been studied extensively because of its critical role in cellular signaling and association with disease. Previous models have elucidated interactions between EGFR and downstream adaptor proteins or showed phenotypes affected by EGFR. However, the link between specific EGFR phosphorylation sites and phenotypic outcomes is still poorly understood. Here, we employed a suite of isogenic cell lines expressing site-specific mutations at each of the EGFR C-terminal phosphorylation sites to interrogate their role in the signaling network and cell biological response to stimulation. Our results demonstrate the resilience of the EGFR network, which was largely similar even in the context of multiple Y-to-F mutations in the EGFR C-terminal tail, while also revealing nodes in the network that have not previously been linked to EGFR signaling. Our data-driven model highlights the signaling network nodes associated with distinct EGF-driven cell responses, including migration, proliferation, and receptor trafficking. Application of this same approach to less-studied RTKs should provide a plethora of novel associations that should lead to an improved understanding of these signaling networks.


Assuntos
Fator de Crescimento Epidérmico , Tirosina , Tirosina/genética , Tirosina/metabolismo , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Transdução de Sinais/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Fosforilação
3.
Cancer Res ; 63(8): 1969-74, 2003 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-12702590

RESUMO

In this paper, we describe the role of chemokine receptor CXCR4 activation by its natural ligand, the chemokine stromal cell-derived factor (SDF-1) (CXCL12), in glioblastoma cell growth in vitro. We show that both CXC chemokine receptor 4 (CXCR4) and SDF-1 mRNA are expressed in several human glioblastoma multiforme tumor tissues and in two human glioblastoma cell lines, U87-MG and DBTRG-05MG. These cells are able to secrete SDF-1 under basal conditions, and the rate of secretion is highly increased after lipopolysaccharide or 1% fetal bovine serum treatment. Exogenous SDF-1alpha induces proliferation in a dose-dependent manner in both cell lines. Moreover, we observed that SDF-1alpha-dependent proliferation is correlated with phosphorylation and activation of both extracellular signal-regulated kinases 1/2 and Akt and that these kinases are independently involved in glioblastoma cell proliferation. The role of CXCR4 stimulation in glioblastoma cell growth is further demonstrated by the ability of human monoclonal CXCR4 antibody (clone 12G5) to inhibit the SDF-1alpha-induced proliferation as well as the proliferation induced by SDF-1-releasing treatments (lipopolysaccharide and 1% fetal bovine serum). These data support a role for SDF-1alpha in the regulation of glioblastoma growth in vitro, likely through an autocrine/paracrine mechanism.


Assuntos
Quimiocinas CXC/fisiologia , Glioblastoma/patologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Receptores CXCR4/fisiologia , Animais , Divisão Celular/fisiologia , Quimiocina CXCL12 , Quimiocinas CXC/biossíntese , Quimiocinas CXC/metabolismo , DNA de Neoplasias/biossíntese , Ativação Enzimática , Glioblastoma/enzimologia , Glioblastoma/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , Proteína Quinase 3 Ativada por Mitógeno , Fosforilação , Proteínas Proto-Oncogênicas c-akt , Ratos , Receptores CXCR4/biossíntese , Receptores CXCR4/metabolismo , Células Tumorais Cultivadas
4.
Neurochem Int ; 43(1): 31-8, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12605880

RESUMO

Pyrrolidinedithiocarbamate (PDTC) is a compound displaying antioxidant, pro-oxidant and metal chelator properties in different cell types. It has been described that PDTC may exert either anti-apoptotic or apoptotic activity. Moreover it is known that this agent regulates the activity of redox-sensitive transcription factors, such as AP-1 and NF-kappaB. Using cerebellar granule cells (CGCs), a well-described model of neuronal primary cultures, we investigated the effects of different concentrations of this compound on cell viability and the intracellular mechanisms involved. PDTC used at concentrations, as low as 1 microM, exerts cytotoxic effects on CGC through the activation of the apoptotic machinery with a maximal efficacy for concentration of 10 microM. The PDTC-dependent apoptosis is correlated to a biphasic and long-lasting increase of AP-1 binding to the DNA, apparently without affecting the NF-kappaB whose activity was reduced only at much higher concentrations (100 microM). PDTC treatment enhanced ERK phosphorylation (maximal effect 1h) and p38 phosphorylation (maximal effect 7h) that was accompanied by an increase of both mRNA and protein of c-Jun. In conclusion the results presented show that PDTC exerts apoptotic effects on CGC, that are correlated to the activation of stress-pathways, involving mainly AP-1 and MAPKs.


Assuntos
Apoptose/efeitos dos fármacos , Cerebelo/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Pirrolidinas/farmacologia , Tiocarbamatos/farmacologia , Fator de Transcrição AP-1/efeitos dos fármacos , Animais , Bisbenzimidazol , Western Blotting , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/ultraestrutura , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Cerebelo/citologia , Grânulos Citoplasmáticos/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Ensaio de Desvio de Mobilidade Eletroforética , Corantes Fluorescentes , Técnicas In Vitro , Proteínas Quinases Ativadas por Mitógeno/genética , NF-kappa B/antagonistas & inibidores , NF-kappa B/fisiologia , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/química , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sais de Tetrazólio , Tiazóis , Proteínas Quinases p38 Ativadas por Mitógeno
5.
Ann N Y Acad Sci ; 973: 402-13, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12485901

RESUMO

Many activities of neuronal cells, such as synaptic transmission, inflammation, neuroprotection, and neurotoxicity, are regulated by the activity of the transcription factor nuclear factor-kappaB (NF-kappaB). In resting cells, NF-kappaB activity is present both in the cytoplasm, as an inducible-inactive complex, and in the nucleus as a constitutive form. The activation of its inducible form is related to processing of IkappaB(s), which occurs through the proteasome. To understand whether NF-kappaB is involved in the survival of cerebellar granule cells (CGCs) maintained under conditions of mild depolarization (25 mM KCl), these cells were treated with different proteasome inhibitors. The results presented show that these pharmacological tools reduce CGC survival with changes in nuclear morphology and induction of apoptosis. Furthermore, we demonstrate that PSI-induced apoptosis is reverted by inhibitors of transcription and translation, as well as by specific caspase inhibitors. These issues are also associated with a redistribution of NF-kappaB, in that a reduced amount of nuclear NF-kappaB and an increased p65 cytoplasmic level have been observed. Finally, we propose that, at least in part, p65 metabolism could also be regulated by the ubiquitin-proteasome complex. Altogether, the results presented define an important role for NF-kappaB in maintainig CGC survival.


Assuntos
Cerebelo/citologia , Cisteína Endopeptidases/metabolismo , Citarabina/farmacologia , Dactinomicina/farmacologia , Complexos Multienzimáticos/metabolismo , NF-kappa B/antagonistas & inibidores , Inibidores de Proteases/farmacologia , Animais , Apoptose/efeitos dos fármacos , Inibidores de Caspase , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cerebelo/efeitos dos fármacos , Cinética , Neurônios/citologia , Neurônios/efeitos dos fármacos , Oligopeptídeos/farmacologia , Complexo de Endopeptidases do Proteassoma , Ratos , Ratos Sprague-Dawley
6.
Ann N Y Acad Sci ; 973: 60-9, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12485835

RESUMO

Chemokines are a family of proteins that chemoattract and activate cells by interacting with specific receptors on the surface of their targets. They are grouped into four classes based on the position of key cysteine residues: C, CC, CXC, and CX3C. Stromal cell-derived factor 1 (SDF1), the ligand of the CXCR4 receptor, is a CXC chemokine involved in chemotaxis and brain development that also acts as coreceptor for HIV-1 infection. It has been proposed that CXCR4 is overexpressed and required for proliferation in human brain tumor cells. We previously demonstrated that CXCR4 and SDF1 are expressed in culture of cortical type I rat astrocytes, cortical neurons, and cerebellar granule cells. In this study, we analyzed the expression of CXCR4 and SDF1 in four human brain tumor tissues, showing that CXCR4 is expressed in all tumors analyzed, whereas SDF1 is expressed only in two tumor tissues. We also investigated the possible functions of CXCR4 expressed in rat type I cortical astrocytes, demonstrating that SDF1alpha stimulates the proliferation of these cells in vitro. Moreover, we studied by western blot the intracellular pathway involved in cell proliferation, demonstrating that SDF1alpha induces the ERK1/2 phosphorylation that is reduced by the PD98059 compound, an MEK inhibitor.


Assuntos
Neoplasias Encefálicas/genética , Quimiocinas CXC/genética , Neuroglia/patologia , Receptores CXCR4/genética , Animais , Animais Recém-Nascidos , Astrócitos/citologia , Astrócitos/fisiologia , Neoplasias Encefálicas/imunologia , Divisão Celular , Quimiocina CXCL12 , Regulação Neoplásica da Expressão Gênica , Glioblastoma , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Timidina/metabolismo , Células Tumorais Cultivadas
7.
Toxicol Lett ; 139(2-3): 181-9, 2003 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-12628753

RESUMO

The study of chemokine role in the CNS indubitably represents an important step to understanding many aspects of brain pathology, physiology and development. Here we discuss our recent research on the expression of chemokines and chemokine receptors in brain tissues and in cultured CNS cells, with particular regard to the CXCL12/SDF-1-CXCR4 system. We showed their expression in both glial and neuronal cells in basal conditions and their modulation upon stimulation. We demonstrated that CXCL12/SDF-1 in vitro act as a growth factor for astrocytes by stimulating their proliferation, a phenomenon that could represent the basis of pathological conditions such as gliosis and malignant transformation. We investigated the signal transduction pathways, identifying in the sequential activation of G-protein-PI-3Kinase-ERK1/2 the main signaling cascade linked to the CXCL12/SDF-1-induced proliferation in astrocytes.


Assuntos
Astrócitos/citologia , Astrócitos/metabolismo , Sistema Nervoso Central/citologia , Sistema Nervoso Central/metabolismo , Quimiocinas CXC/metabolismo , Quimiocinas/metabolismo , Regulação da Expressão Gênica , Receptores CXCR4/metabolismo , Animais , Divisão Celular , Quimiocina CXCL12 , Quimiocinas CXC/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Receptores CXCR4/genética
8.
Cancers (Basel) ; 6(1): 226-39, 2014 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-24473088

RESUMO

Heterogeneity is a hallmark of tumors and has a crucial role in the outcome of the malignancy, because it not only confounds diagnosis, but also challenges the design of effective therapies. There are two types of heterogeneity: inter-tumor and intra-tumor heterogeneity. While inter-tumor heterogeneity has been studied widely, intra-tumor heterogeneity has been neglected even though numerous studies support this aspect of tumor pathobiology. The main reason has been the technical difficulties, but with new advances in single-cell technology, intra-tumor heterogeneity is becoming a key area in the study of cancer. Several models try to explain the origin and maintenance of intra-tumor heterogeneity, however, one prominent model compares cancer with a tree where the ubiquitous mutations compose the trunk and mutations present in subpopulations of cells are represented by the branches. In this review we will focus on the intra-tumor heterogeneity of glioblastoma multiforme (GBM), the most common brain tumor in adults that is characterized by a marked heterogeneity at the cellular and molecular levels. Better understanding of this heterogeneity will be essential to design effective therapies against this devastating disease to avoid tumor escape.

9.
Cancer Res ; 71(12): 4055-60, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21628493

RESUMO

Glioblastoma multiforme (GBM), the most common intracranial tumor in adults, is characterized by extensive heterogeneity at the cellular and molecular levels. This insidious feature arises inevitably in almost all cancers and has great significance for the general outcome of the malignancy, because it confounds our understanding of the disease and also intrinsically contributes to the tumor's aggressiveness and poses an obstacle to the design of effective therapies. The classic view that heterogeneity arises as the result of a tumor's "genetic chaos" and the more contemporary cancer stem cell (CSC) hypothesis tend to identify a single cell population as the therapeutic target: the prevailing clone over time in the first case and the CSC in the latter. However, there is growing evidence that the different tumor cell populations may not be simple bystanders. Rather, they can establish a complex network of interactions between each other and with the tumor microenvironment that eventually strengthens tumor growth and increases chances to escape therapy. These differing but complementary ideas about the origin and maintenance of tumor heterogeneity and its importance in GBM are reviewed here.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Comunicação Celular , Modelos Animais de Doenças , Receptores ErbB/genética , Glioblastoma/genética , Glioblastoma/terapia , Humanos , Camundongos , Mutação , Células-Tronco Neoplásicas/patologia
10.
Proc Natl Acad Sci U S A ; 104(31): 12867-72, 2007 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-17646646

RESUMO

Glioblastoma multiforme (GBM) is the most aggressive brain tumor in adults and remains incurable despite multimodal intensive treatment regimens. EGFRvIII is a truncated extracellular mutant of the EGF receptor (EGFR) commonly found in GBMs that confers enhanced tumorigenic behavior. To gain a molecular understanding of the mechanisms by which EGFRvIII acts, we have performed a large-scale analysis of EGFRvIII-activated phosphotyrosine-mediated signaling pathways and thereby have identified and quantified 99 phosphorylation sites on 69 proteins. Distinct signaling responses were observed as a function of titrated EGFRvIII receptor levels with the phosphatidylinositol 3-kinase pathway being dominant over the MAPK and STAT3 pathways at a high level of EGFRvIII expression. Within this data set, the activating phosphorylation site on the c-Met receptor was found to be highly responsive to EGFRvIII levels, indicating cross-activation of the c-Met receptor tyrosine kinase by EGFRvIII. To determine the significance of this finding, we devised a combined treatment regimen that used a c-Met kinase inhibitor and either an EGFR kinase inhibitor or cisplatin. This regimen resulted in enhanced cytotoxicity of EGFRvIII-expressing cells compared with treatment with either compound alone. These results suggest that the clinical use of c-Met kinase inhibitors in combination with either EGFR inhibitors or standard chemotherapeutics might represent a previously undescribed therapeutic approach to overcome the observed chemoresistance in patients with GBMs expressing EGFRvIII.


Assuntos
Receptores ErbB/metabolismo , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Transdução de Sinais , Animais , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Receptores ErbB/antagonistas & inibidores , Glioblastoma/patologia , Humanos , Camundongos , Camundongos Nus , Fosforilação , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Quinases/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Exp Cell Res ; 308(2): 241-53, 2005 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15921680

RESUMO

Ovarian cancer (OC) is the leading cause of death in gynecologic diseases in which there is evidence for a complex chemokine network. Chemokines are a family of proteins that play an important role in tumor progression influencing cell proliferation, angiogenic/angiostatic processes, cell migration and metastasis, and, finally, regulating the immune cells recruitment into the tumor mass. We previously demonstrated that astrocytes and glioblastoma cells express both the chemokine receptor CXCR4 and its ligand stromal cell-derived factor-1 (SDF-1), and that SDF-1alpha treatment induced cell proliferation, supporting the hypothesis that chemokines may play an important role in tumor cells' growth in vitro. In the present study, we report that CXCR4 and SDF-1 are expressed in OC cell lines. We demonstrate that SDF-1alpha induces a dose-dependent proliferation in OC cells, by the specific interaction with CXCR4 and a biphasic activation of ERK1/2 and Akt kinases. Our results further indicate that CXCR4 activation induces EGF receptor (EGFR) phosphorylation that in turn was linked to the downstream intracellular kinases activation, ERK1/2 and Akt. In addition, we provide evidence for cytoplasmic tyrosine kinase (c-Src) involvement in the SDF-1/CXCR4-EGFR transactivation. These results suggest a possible important "cross-talk" between SDF-1/CXCR4 and EGFR intracellular pathways that may link signals of cell proliferation in ovarian cancer.


Assuntos
Carcinoma/metabolismo , Quimiocinas CXC/metabolismo , Receptores ErbB/metabolismo , Neoplasias Ovarianas/metabolismo , Receptores CXCR4/metabolismo , Ativação Transcricional , Carcinoma/fisiopatologia , Linhagem Celular Tumoral , Proliferação de Células , Quimiocina CXCL12 , Quimiocinas CXC/farmacologia , Relação Dose-Resposta a Droga , Feminino , Genes src/genética , Humanos , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neoplasias Ovarianas/fisiopatologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Receptor Cross-Talk/fisiologia
12.
J Neurochem ; 82(6): 1311-29, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12354279

RESUMO

Chemokines represent key factors in the outburst of the immune response, by activating and directing the leukocyte traffic, both in lymphopoiesis and in immune surveillance. Neurobiologists took little interest in chemokines for many years, until their link to acquired immune deficiency syndrome-associated dementia became established, and thus their importance in this field has been neglected. Nevertheless, the body of data on their expression and role in the CNS has grown in the past few years, along with a new vision of brain as an immunologically competent and active organ. A large number of chemokines and chemokine receptors are expressed in neurons, astrocytes, microglia and oligodendrocytes, either constitutively or induced by inflammatory mediators. They are involved in many neuropathological processes in which an inflammatory state persists, as well as in brain tumor progression and metastasis. Moreover, there is evidence for a crucial role of CNS chemokines under physiological conditions, similar to well known functions in the immune system, such as proliferation and developmental patterning, but also peculiar to the CNS, such as regulation of neural transmission, plasticity and survival.


Assuntos
Doenças do Sistema Nervoso Central/fisiopatologia , Sistema Nervoso Central/fisiopatologia , Quimiocinas/metabolismo , Receptores de Quimiocinas/metabolismo , Animais , Quimiocinas/classificação , Humanos , Neuroglia/metabolismo , Neurônios/metabolismo , Receptores de Quimiocinas/classificação , Transdução de Sinais/imunologia
13.
J Neurooncol ; 60(2): 109-16, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12635657

RESUMO

Malignant gliomas are the most common primary brain tumors in humans. However, poor response to conventional therapeutic approaches, including chemotherapy, leads invariably to disease recurrence and progression. The organo-tin derivative triethyltin(IV)lupinylsulfide hydrochloride (IST-FS 29) was identified and developed as potential antiproliferative agent in human cancer cell lines. However, for its peculiar chemical structure and good lipophilicity, this compound also appeared an eligible candidate for the treatment of gliobastoma cells. The present experiments were designed to explore the in vitro effects of IST-FS 29 on four human glioblastoma cell lines: A-172, DBTRG.05MG, U-87MG and CAS-1. The average IC50 values were obtained by MTT assay and ranged between 3 and 10 microM. Time-course assays with cell recovery after drug withdrawal, demonstrated marked cytotoxicity following exposure to IST-FS 29 for 8, 24 and 72 h. Cultures treated for 8 h were able to partially re-grow by 144 h; on the contrary, longer times of exposure did not allow surviving cells to recover from the damage and actively proliferate. Cell morphology of cultures exposed to IST-FS 29 was assessed by inverted light microscopy after 24 and 72 h and was more consistent with cell death by necrosis which included cell size reduction, vacuolation of cytoplasm, round dying cells. The present results and our previous data, in vitro and in vivo, indicate the relevant cytotoxic activity of this organo-tin compound and suggest that IST-FS 29 might be a promising novel agent to be developed for the treatment of malignant brain neoplasms.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Encefálicas , Glioblastoma , Compostos de Trietilestanho/farmacologia , Cálcio/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa