Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 8(10): e78632, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24205284

RESUMO

Lysophosphatidic acid acyltransferase (LPAAT-ß) is a phosphatidic acid (PA) generating enzyme that plays an essential role in triglyceride synthesis. However, LPAAT-ß is now being studied as an important regulator of cell growth and differentiation and as a potential therapeutic target in cancer since PA is necessary for the activity of key proteins such as Raf, PKC-ζ and mTOR. In this report we determine the effect of LPAAT-ß silencing with siRNA in pancreatic adenocarcinoma cell lines. We show for the first time that LPAAT-ß knockdown inhibits proliferation and anchorage-independent growth of pancreatic cancer cells. This is associated with inhibition of signaling by mTOR as determined by levels of mTORC1- and mTORC2-specific phosphorylation sites on 4E-BP1, S6K and Akt. Since PA regulates the activity of mTOR by modulating its binding to FKBP38, we explored the possibility that LPAAT-ß might regulate mTOR by affecting its association with FKBP38. Coimmunoprecipitation studies of FKBP38 with mTOR show increased levels of FKBP38 associated with mTOR when LPAAT-ß protein levels are knocked down. Furthermore, depletion of LPAAT-ß results in increased Lipin 1 nuclear localization which is associated with increased nuclear eccentricity, a nuclear shape change that is dependent on mTOR, further confirming the ability of LPAAT-ß to regulate mTOR function. Our results provide support for the hypothesis that PA generated by LPAAT-ß regulates mTOR signaling. We discuss the implications of these findings for using LPAAT-ß as a therapeutic target.


Assuntos
Aciltransferases/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Transporte Ativo do Núcleo Celular , Aciltransferases/deficiência , Aciltransferases/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adenocarcinoma/patologia , Sequência de Bases , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proliferação de Células , Regulação Enzimológica da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Pancreáticas/patologia , Fosfatidato Fosfatase/metabolismo , Ácidos Fosfatídicos/biossíntese , Fosfoproteínas/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética
2.
J Transl Med ; 10: 246, 2012 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-23228035

RESUMO

BACKGROUND: Multiple farnesylated proteins are involved in signal transduction in cancer. Farnesyltransferase inhibitors (FTIs) have been developed as a strategy to inhibit the function of these proteins. As FTIs inhibit proliferation of melanoma cell lines, we undertook a study to assess the impact of a FTI in advanced melanoma. As farnesylated proteins are also important for T cell activation, measurement of effects on T cell function was also pursued. METHODS: A 3-stage trial design was developed with a maximum of 40 patients and early stopping if there were no responders in the first 14, or fewer than 2 responders in the first 28 patients. Eligibility included performance status of 0-1, no prior chemotherapy, at most 1 prior immunotherapy, no brain metastases, and presence of at least 2 cutaneous lesions amenable to biopsy. R115777 was administered twice per day for 21 days of a 28-day cycle. Patients were evaluated every 2 cycles by RECIST. Blood and tumor were analyzed pre-treatment and during week 7. RESULTS: Fourteen patients were enrolled. Two patients had grade 3 toxicities, which included myelosuppression, nausea/vomiting, elevated BUN, and anorexia. There were no clinical responses. All patients analyzed showed potent inhibition of FT activity (85-98%) in tumor tissue; inhibition of phosphorylated ERK and Akt was also observed. T cells showed evidence of FT inhibition and diminished IFN-γ production. CONCLUSIONS: Despite potent target inhibition, R115777 showed no evidence of clinical activity in this cohort of melanoma patients. Inhibition of T cell function by FTIs has potential clinical implications. Clinicaltrials.gov number NCT00060125.


Assuntos
Inibidores Enzimáticos/uso terapêutico , Farnesiltranstransferase/antagonistas & inibidores , Melanoma/tratamento farmacológico , Melanoma/patologia , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Biópsia , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Farnesiltranstransferase/metabolismo , Feminino , Proteínas de Choque Térmico HSP40/metabolismo , Humanos , Interferon gama/biossíntese , Masculino , Melanoma/sangue , Melanoma/enzimologia , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinolonas/administração & dosagem , Quinolonas/efeitos adversos , Quinolonas/farmacologia , Quinolonas/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Neoplasias Cutâneas/sangue , Neoplasias Cutâneas/enzimologia , Linfócitos T/efeitos dos fármacos , Resultado do Tratamento
3.
Cancer Immunol Immunother ; 61(4): 523-33, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21983879

RESUMO

Large granular lymphocyte (LGL) leukemia is a chronic lymphoproliferative disease in which T-bet [T-box transcription factor 21 gene (tbx21)] overexpression may play a pathogenic role. T-bet orchestrates the differentiation of mature peripheral T-cells into interferon-γ (IFN-γ) and tumor necrosis factor-α producing CD4+ T-helper type I (Th1) and CD8+ T cytotoxic cells that are necessary for antiviral responses. When IL-12 is produced by antigen-presenting cells, T-bet expression is induced, causing direct stimulation of ifng gene transcription while simultaneously acting as a transcriptional repressor of the IL4 gene, which then leads to Th1 dominance and T-helper type 2 differentiation blockade. Additionally, T-bet has been shown to regulate histone acetylation of the ifng promoter and enhancer to loosen condensed DNA, creating greater accessibility for other transcription factor binding, which further amplifies IFNγ production. We found that treatment with a farnesyltransferase inhibitor tipifarnib reduced Th1 cytokines in LGL leukemia patient T-cells and blocked T-bet protein expression and IL-12 responsiveness in T-cells from healthy donors. The mechanism of suppression was based on modulation of histone acetylation of the ifng gene, which culminated in Th1 blockade.


Assuntos
Antineoplásicos/farmacologia , Farnesiltranstransferase/antagonistas & inibidores , Leucemia Linfocítica Granular Grande/imunologia , Quinolonas/farmacologia , Proteínas com Domínio T/metabolismo , Células Th1/efeitos dos fármacos , Células Th2/efeitos dos fármacos , Acetilação/efeitos dos fármacos , Adulto , Idoso , Citocinas/genética , Citocinas/imunologia , Citocinas/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Terapia de Imunossupressão , Leucemia Linfocítica Granular Grande/patologia , Masculino , Pessoa de Meia-Idade , Transdução de Sinais/efeitos dos fármacos , Proteínas com Domínio T/genética , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/patologia , Equilíbrio Th1-Th2/efeitos dos fármacos , Células Th2/imunologia , Células Th2/metabolismo , Células Th2/patologia
4.
Clin Cancer Res ; 17(5): 1140-6, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21233404

RESUMO

PURPOSE: To determine the safety, target inhibition, and signals of clinical activity of tipifarnib in combination with bortezomib in patients with advanced acute leukemias. EXPERIMENTAL DESIGN: In a "3 + 3" design, patients received escalating doses of tipifarnib (days 1-14) and bortezomib (days 1, 4, 8, 11) every 3 weeks until maximum tolerated dose was reached. Peripheral blood mononuclear cells (PBMC) were collected at days 1, 8, and 22 for measurement of chymotrypsin-like and farnesyltransferase activity. Purified bone marrow leukemic blasts were collected at baseline and at day 8 for measurement of NF-κB activity. RESULTS: The combination was well-tolerated, and maximum tolerated dose was not reached. Dose-limiting toxicities included diarrhea, fatigue, and sensorimotor neuropathy. Chymotrypsin-like and farnesyltransferase activity within PBMCs were decreased in a majority of patients at day 8. NF-κB activity within leukemic blasts was decreased in a majority of patients at day 8. Complete response with incomplete count recovery was observed in 2 patients, and additional 5 patients had stable disease. CONCLUSIONS: Tipifarnib and bortezomib combination in patients with advanced leukemias was well-tolerated, demonstrated relevant target inhibition, and was associated with signals of clinical activity in patients with advanced and refractory acute leukemias. Future studies of this combination may be warranted in more selected groups of patients in whom these molecular targets are of particular importance.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Ácidos Borônicos/administração & dosagem , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Pirazinas/administração & dosagem , Quinolonas/administração & dosagem , Idoso , Idoso de 80 Anos ou mais , Ácidos Borônicos/efeitos adversos , Ácidos Borônicos/farmacocinética , Bortezomib , Quimotripsina/sangue , Quimotripsina/metabolismo , Farnesiltranstransferase/antagonistas & inibidores , Farnesiltranstransferase/sangue , Feminino , Humanos , Leucócitos Mononucleares/metabolismo , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Inibidores de Proteassoma , Pirazinas/efeitos adversos , Pirazinas/farmacocinética , Quinolonas/efeitos adversos , Quinolonas/farmacocinética , Quinolonas/uso terapêutico
5.
J Med Chem ; 53(19): 6867-88, 2010 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-20822181

RESUMO

A potent class of anticancer, human farnesyltransferase (hFTase) inhibitors has been identified by "piggy-backing" on potent, antimalarial inhibitors of Plasmodium falciparum farnesyltransferase (PfFTase). On the basis of a 4-fold substituted ethylenediamine scaffold, the inhibitors are structurally simple and readily derivatized, facilitating the extensive structure-activity relationship (SAR) study reported herein. Our most potent inhibitor is compound 1f, which exhibited an in vitro hFTase IC(50) value of 25 nM and a whole cell H-Ras processing IC(50) value of 90 nM. Moreover, it is noteworthy that several of our inhibitors proved highly selective for hFTase (up to 333-fold) over the related prenyltransferase enzyme geranylgeranyltransferase-I (GGTase-I). A crystal structure of inhibitor 1a co-crystallized with farnesyl pyrophosphate (FPP) in the active site of rat FTase illustrates that the para-benzonitrile moiety of 1a is stabilized by a π-π stacking interaction with the Y361ß residue, suggesting a structural explanation for the observed importance of this component of our inhibitors.


Assuntos
Antineoplásicos/síntese química , Etilenodiaminas/síntese química , Farnesiltranstransferase/antagonistas & inibidores , Modelos Moleculares , Compostos de Anilina/síntese química , Compostos de Anilina/química , Compostos de Anilina/farmacologia , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Domínio Catalítico , Linhagem Celular , Cristalografia por Raios X , Desenho de Fármacos , Etilenodiaminas/química , Etilenodiaminas/farmacologia , Humanos , Estrutura Molecular , Nitrilas/síntese química , Nitrilas/química , Nitrilas/farmacologia , Plasmodium falciparum/enzimologia , Ligação Proteica , Ratos , Relação Estrutura-Atividade , Sulfonamidas/síntese química , Sulfonamidas/química , Sulfonamidas/farmacologia
6.
Clin Cancer Res ; 15(22): 7061-8, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19903778

RESUMO

PURPOSE: We evaluated the safety, maximum tolerated dose, pharmacokinetics, and biological effects of the combination of the Raf-1, RET, KIT, platelet-derived growth factor receptor, and vascular endothelial growth factor receptor 2 kinase inhibitor sorafenib and the farnesyltransferase inhibitor tipifarnib. EXPERIMENTAL DESIGN: A standard 3 + 3 phase I dose-escalation design was used with a 28-day cycle (sorafenib daily and tipifarnib for 21 days, by mouth). RESULTS: Fifty patients were treated; 43 reached restaging evaluation after cycle 2. The most common side effects were grade 1 to 2 rash, hyperglycemia, and diarrhea. Dose-limiting toxicity was rash, and the recommended phase II dose is sorafenib 400 mg p.o. qam/200 mg p.o. qpm and tipifarnib p.o. 100 mg bd. Despite the low doses of tipifarnib, one quarter of patients had > or =50% reduction in farnesyltransferase levels. Interestingly, six of eight patients with medullary thyroid cancer had durable stable disease (n = 3) or partial remissions (n = 3), lasting 12 to 26+ months. Five of the six responders had available tissue, and RET gene mutations were identified in them. Prolonged (> or =6 months) stable disease was also seen in nine patients as follows: papillary thyroid cancer (n = 4; 18+ to 27+ months), adrenocortical cancer (n = 2; 7 and 11 months), and one each of melanoma (platelet-derived growth factor receptor mutation positive; 14 months), renal (6 months), and pancreatic cancer (6 months). CONCLUSIONS: Our study shows that the combination of tipifarnib and sorafenib is well tolerated. Activity was seen, especially in patients with medullary thyroid cancer, a tumor characterized by RET mutations.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Benzenossulfonatos/administração & dosagem , Farnesiltranstransferase/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Piridinas/administração & dosagem , Quinolonas/administração & dosagem , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Mutação , Niacinamida/análogos & derivados , Compostos de Fenilureia , Proteínas Proto-Oncogênicas c-ret/genética , Sorafenibe , Resultado do Tratamento
7.
Mol Cell Biol ; 29(8): 2254-63, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19204084

RESUMO

We describe the design of a potent and selective peptidomimetic inhibitor of geranylgeranyltransferase I (GGTI), GGTI-2418, and its methyl ester GGTI-2417, which increases the levels of the cyclin-dependent kinase (Cdk) inhibitor p27(Kip1) and induces breast tumor regression in vivo. Experiments with p27(Kip1) small interfering RNA in breast cancer cells and p27(Kip1) null murine embryonic fibroblasts demonstrate that the ability of GGTI-2417 to induce cell death requires p27(Kip1). GGTI-2417 inhibits the Cdk2-mediated phosphorylation of p27(Kip1) at Thr187 and accumulates p27(Kip1) in the nucleus. In nude mouse xenografts, GGTI-2418 suppresses the growth of human breast tumors. Furthermore, in ErbB2 transgenic mice, GGTI-2418 increases p27(Kip1) and induces significant regression of breast tumors. We conclude that GGTIs' antitumor activity is, at least in part, due to inhibiting Cdk2-dependent p27(Kip1) phosphorylation at Thr187 and accumulating nuclear p27(Kip1). Thus, GGTI treatment might improve the poor prognosis of breast cancer patients with low nuclear p27(Kip1) levels.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Prenilação de Proteína/efeitos dos fármacos , Animais , Antineoplásicos/farmacologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Feminino , Humanos , Camundongos , Células NIH 3T3 , Fosforilação/efeitos dos fármacos , Treonina/metabolismo
8.
Chem Biol ; 16(2): 181-92, 2009 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-19246009

RESUMO

Protein farnesyltransferase (FTase) catalyzes an essential posttranslational lipid modification of more than 60 proteins involved in intracellular signal transduction networks. FTase inhibitors have emerged as a significant target for development of anticancer therapeutics and, more recently, for the treatment of parasitic diseases caused by protozoan pathogens, including malaria (Plasmodium falciparum). We present the X-ray crystallographic structures of complexes of mammalian FTase with five inhibitors based on an ethylenediamine scaffold, two of which exhibit over 1000-fold selective inhibition of P. falciparum FTase. These structures reveal the dominant determinants in both the inhibitor and enzyme that control binding and selectivity. Comparison to a homology model constructed for the P. falciparum FTase suggests opportunities for further improving selectivity of a new generation of antimalarial inhibitors.


Assuntos
Antimaláricos/química , Antineoplásicos/química , Inibidores Enzimáticos/química , Farnesiltranstransferase/antagonistas & inibidores , Farnesiltranstransferase/química , Animais , Antimaláricos/metabolismo , Antineoplásicos/metabolismo , Linhagem Celular Tumoral , Cristalografia por Raios X , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/metabolismo , Etilenodiaminas/química , Farnesiltranstransferase/metabolismo , Humanos , Plasmodium falciparum/enzimologia , Ligação Proteica , Conformação Proteica , Proteínas de Protozoários/antagonistas & inibidores , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo , Ratos , Homologia Estrutural de Proteína , Relação Estrutura-Atividade , Especificidade por Substrato
9.
Blood ; 112(12): 4694-8, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18791165

RESUMO

Large granular lymphocyte (LGL) leukemia is commonly associated with poor hematopoiesis. The first case of pulmonary artery hypertension (PAH) was observed in a 57-year-old woman with natural killer (NK)-LGL leukemia and transfusion-dependent anemia. Using a genetic approach, we demonstrated that killing of pulmonary endothelial cells by patient NK cells was mediated by dysregulated balance in activating and inhibitory NK-receptor signaling. Elevated pulmonary artery pressure and erythroid differentiation improved after disrupting the NK-receptor signaling pathway with 4 courses of a farnesyltransferase inhibitor, tipifarnib. Coincidental association between PAH and LGL leukemia suggest a causal relationship between the expanded lymphocyte population and these clinical manifestations. This trial is registered at www.ClinicalTrials.gov as NCI 6823.


Assuntos
Antineoplásicos/uso terapêutico , Farnesiltranstransferase/antagonistas & inibidores , Leucemia Linfocítica Granular Grande/tratamento farmacológico , Receptores de Células Matadoras Naturais/genética , Células Cultivadas , Inibidores Enzimáticos/uso terapêutico , Feminino , Humanos , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/etiologia , Células K562 , Leucemia Linfocítica Granular Grande/complicações , Leucemia Linfocítica Granular Grande/genética , Pessoa de Meia-Idade , Quinolonas/uso terapêutico , Transdução de Sinais/genética , Resultado do Tratamento
10.
Chemistry ; 14(5): 1392-401, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18200641

RESUMO

Synthetic chemical probes designed to simultaneously targeting multiple sites of protein surfaces are of interest owing to their potential application as site specific modulators of protein-protein interactions. A new approach toward bivalent inhibitors of mammalian type I geranylgeranyltransferase (GGTase I) based on module assembly for simultaneous recognition of both interior and exterior protein surfaces is reported. The inhibitors synthesized in this study consist of two modules linked by an alkyl spacer; one is the tetrapeptide CVIL module for binding to the interior protein surface (active pocket) and the other is a 3,4,5-alkoxy substituted benzoyl motif that contains three aminoalkyl groups designed to bind to the negatively charged protein exterior surface near the active site. The compounds were screened by two distinct enzyme inhibition assays based on fluorescence spectroscopy and incorporation of a [(3)H]-labeled prenyl group onto a protein substrate. The bivalent inhibitors block GGTase I enzymatic activity with K(i) values in the submicromolar range and are approximately one order of magnitude and more than 150 times more effective than the tetrapeptide CVIL and the methyl benzoate derivatives, respectively. The bivalent compounds 6 and 8 were shown to be competitive inhibitors, suggesting that the CVIL module anchors the whole molecule to the GGTase I active site and delivers the other module to the targeting protein surface. Thus, our module-assembly approach resulted in simultaneous multiple-site recognition, and as a consequence, synergetic inhibition of GGTase I activity, thereby providing a new approach in designing protein-surface-directed inhibitors for targeting protein-protein interactions.


Assuntos
Alquil e Aril Transferases/antagonistas & inibidores , Alquil e Aril Transferases/metabolismo , Inibidores Enzimáticos/farmacologia , Álcoois/química , Álcoois/metabolismo , Alquil e Aril Transferases/química , Alquilação , Aminas/química , Aminas/metabolismo , Sequência de Aminoácidos , Benzoatos/química , Benzoatos/metabolismo , Sítios de Ligação , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/química , Marcação por Isótopo , Cinética , Dados de Sequência Molecular , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Prenilação , Ligação Proteica , Espectrometria de Fluorescência , Relação Estrutura-Atividade , Especificidade por Substrato
11.
Proc Natl Acad Sci U S A ; 104(18): 7391-6, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17463090

RESUMO

S3I-201 (NSC 74859) is a chemical probe inhibitor of Stat3 activity, which was identified from the National Cancer Institute chemical libraries by using structure-based virtual screening with a computer model of the Stat3 SH2 domain bound to its Stat3 phosphotyrosine peptide derived from the x-ray crystal structure of the Stat3beta homodimer. S3I-201 inhibits Stat3.Stat3 complex formation and Stat3 DNA-binding and transcriptional activities. Furthermore, S3I-201 inhibits growth and induces apoptosis preferentially in tumor cells that contain persistently activated Stat3. Constitutively dimerized and active Stat3C and Stat3 SH2 domain rescue tumor cells from S3I-201-induced apoptosis. Finally, S3I-201 inhibits the expression of the Stat3-regulated genes encoding cyclin D1, Bcl-xL, and survivin and inhibits the growth of human breast tumors in vivo. These findings strongly suggest that the antitumor activity of S3I-201 is mediated in part through inhibition of aberrant Stat3 activation and provide the proof-of-concept for the potential clinical use of Stat3 inhibitors such as S3I-201 in tumors harboring aberrant Stat3.


Assuntos
Ácidos Aminossalicílicos , Antineoplásicos/química , Antineoplásicos/metabolismo , Benzenossulfonatos/química , Benzenossulfonatos/metabolismo , Avaliação Pré-Clínica de Medicamentos , Fator de Transcrição STAT3/química , Fator de Transcrição STAT3/metabolismo , Ácido Aminossalicílico/química , Ácido Aminossalicílico/metabolismo , Ácido Aminossalicílico/uso terapêutico , Animais , Antineoplásicos/uso terapêutico , Apoptose , Benzenossulfonatos/uso terapêutico , Linhagem Celular , Biologia Computacional , DNA/química , DNA/metabolismo , Regulação da Expressão Gênica , Humanos , Camundongos , Modelos Moleculares , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Fosfotirosina/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/genética , Transcrição Gênica/genética , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Org Biomol Chem ; 4(3): 482-92, 2006 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-16446806

RESUMO

A series of imidazole-containing peptidomimetic PFTase inhibitors and their co-crystal structures bound to PFTase and FPP are reported. The structures reveal that the peptidomimetics adopt a similar conformation to that of the extended CVIM tetrapeptide, with the imidazole group coordinating to the catalytic zinc ion. Both mono- and bis-imidazole-containing derivatives, 13 and 16, showed remarkably high enzyme inhibition activity against PFTase in vitro with IC50 values of 0.86 and 1.7 nM, respectively. The peptidomimetics were also highly selective for PFTase over PGGTase-I both in vitro and in intact cells. In addition, peptidomimetics and were found to suppress tumor growth in nude mouse xenograft models with no gross toxicity at a daily dose of 25 mg kg(-1).


Assuntos
Desenho de Fármacos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Farnesiltranstransferase/antagonistas & inibidores , Imidazóis/química , Peptídeos/química , Peptídeos/farmacologia , Animais , Linhagem Celular , Cristalografia por Raios X , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/uso terapêutico , Farnesiltranstransferase/química , Farnesiltranstransferase/metabolismo , Humanos , Camundongos , Camundongos Nus , Modelos Moleculares , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Peptídeos/síntese química , Peptídeos/uso terapêutico , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Oncogene ; 24(29): 4701-9, 2005 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-15897913

RESUMO

Angiogenesis depends on vascular endothelial growth factor (VEGF) for initiation and platelet-derived growth factor (PDGF) for maintenance of blood vessels. We have designed a targeted library of compounds from which we identified a novel molecule, GFB-204, that binds PDGF and VEGF, blocks binding of PDGF and VEGF to their receptors (200-500 nM) and subsequently inhibits PDGFR and Flk-1 tyrosine phosphorylation and stimulation of the protein kinases Erk1, Erk2 and Akt and the signal transducer and activator of transcription STAT3. GFB-204 is selective for PDGF and VEGF and does not inhibit EGF, IGF-1 and FGF stimulation of Erk1/2, Akt and STAT3. GFB-204 inhibits endothelial cell migration and capillary network formation in vitro. Finally, treatment of mice with GFB-204 suppresses human tumor growth and angiogenesis. Thus, inhibition of VEGF and PDGF receptor binding with a synthetic molecule results in potent inhibition of angiogenesis and tumorigenesis.


Assuntos
Calixarenos/farmacologia , Transformação Celular Neoplásica , Neovascularização Patológica , Fator de Crescimento Derivado de Plaquetas/fisiologia , Receptores do Fator de Crescimento Derivado de Plaquetas/fisiologia , Receptores de Fatores de Crescimento do Endotélio Vascular/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Animais , Movimento Celular , Humanos , Camundongos , Camundongos Nus , Neoplasias Experimentais , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Transplante Heterólogo
14.
Oncogene ; 24(20): 3236-45, 2005 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-15735720

RESUMO

Constitutive activation of the JAK/STAT3 pathway is a major contributor to oncogenesis. In the present study, structure-activity relationship (SAR) studies with five cucurbitacin (Cuc) analogs, A, B, E, I, and Q, led to the discovery of Cuc Q, which inhibits the activation of STAT3 but not JAK2; Cuc A which inhibits JAK2 but not STAT3 activation; and Cuc B, E, and I, which inhibit the activation of both. Furthermore, these SAR studies demonstrated that conversion of the C3 carbonyl of the cucurbitacins to a hydroxyl results in loss of anti-JAK2 activity, whereas addition of a hydroxyl group to C11 of the cucurbitacins results in loss of anti-STAT3 activity. Cuc Q inhibits selectively the activation of STAT3 and induces apoptosis without inhibition of JAK2, Src, Akt, Erk, or JNK activation. Furthermore, Cuc Q induces apoptosis more potently in human and murine tumors that contain constitutively activated STAT3 (i.e., A549, MDA-MB-435, and v-Src/NIH 3T3) as compared to those that do not (i.e., H-Ras/NIH 3T3, MDA-MB-453, and NIH 3T3 cells). Finally, in a nude mouse tumor xenograft model, Cuc Q, but not Cuc A, suppresses tumor growth indicating that JAK2 inhibition is not sufficient to inhibit tumor growth and suggesting that the ability of Cuc Q to inhibit tumor growth is related to its anti-STAT3 activity. These studies further validate STAT3 as a drug discovery target and provide evidence that pharmacological agents that can selectively reduce the P-STAT3 levels in human cancer cells result in tumor apoptosis and growth inhibition.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Transativadores/antagonistas & inibidores , Transativadores/metabolismo , Triterpenos/farmacologia , Células 3T3 , Animais , Apoptose , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células , Cucurbitacinas , Humanos , Imuno-Histoquímica , Imunoprecipitação , Marcação In Situ das Extremidades Cortadas , Concentração Inibidora 50 , Janus Quinase 2 , Camundongos , Camundongos Nus , Modelos Químicos , Células NIH 3T3 , Transplante de Neoplasias , Fosfotirosina/química , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Fator de Transcrição STAT3 , Transdução de Sinais , Relação Estrutura-Atividade , Fatores de Tempo , Triterpenos/química , Quinases da Família src/metabolismo
15.
Bioorg Med Chem ; 13(3): 677-88, 2005 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15653335

RESUMO

A series of novel protein geranylgeranyltransferase-I (PGGTase-I) inhibitors based on a benzoyleneurea scaffold has been synthesized. Using a benzoyleneurea scaffold as a mimetic for the central dipeptide (AA), we have developed CAAX peptidomimetic inhibitors that selectively block the activity of PGGTase-I over the closely related enzyme protein farnesyltransferase. In this new class of PGGTase-I inhibitors, compound (6c) with X=L-phenylalanine, displayed the highest inhibition activity against PGGTase-I with an IC50 value of 170 nM. The inhibitors described in this study represent novel and promising leads for the development of potent and selective inhibitors of mammalian PGGTase-I for potential application as antitumor agents.


Assuntos
Alquil e Aril Transferases/antagonistas & inibidores , Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Ureia/química , Ureia/farmacologia , Animais , Espectroscopia de Ressonância Magnética , Espectrometria de Massas/métodos , Modelos Moleculares , Estrutura Molecular
16.
Bioorg Med Chem ; 12(24): 6517-26, 2004 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-15556768

RESUMO

A series of protein farnesyltransferase inhibitor ester prodrugs of FTI-2148 (17) were synthesized in order to evaluate the effects of ester structure modification on antimalarial activity and for further development of a farnesyltransferase inhibitor with in vivo activity. Evaluation against P. falciparum in red blood cells showed that all the investigated esters exhibited significant antimalarial activity, with the benzyl ester 16 showing the best inhibition (ED50=150 nM). Additionally, compound 16 displayed in vivo activity and was found to suppress parasitemia by 46.1% at a dose of 50 mg kg(-1) day(-1) against Plasmodium berghei in mice. The enhanced inhibition potency of the esters is consistent with improved cell membrane permeability compared to that of the free acid. The results of this study suggest that protein farnesyltransferase is a valid antimalarial drug target and that the antimalarial activity of these compounds derives from a balance between the hydrophobic character and the size and conformation of the ester moiety.


Assuntos
Alquil e Aril Transferases/antagonistas & inibidores , Antimaláricos/síntese química , Antimaláricos/farmacocinética , Permeabilidade da Membrana Celular , Peptídeos/farmacocinética , Animais , Eritrócitos/parasitologia , Ésteres/química , Farnesiltranstransferase , Interações Hidrofóbicas e Hidrofílicas , Malária/tratamento farmacológico , Camundongos , Mimetismo Molecular , Células NIH 3T3 , Peptídeos/síntese química , Plasmodium berghei/efeitos dos fármacos , Plasmodium falciparum/efeitos dos fármacos , Relação Estrutura-Atividade
17.
J Immunol Methods ; 292(1-2): 59-71, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15350512

RESUMO

In this report, we describe a new flow cytometry technique termed flow cytometric high-content screening (FC-HCS) which involves semi-automated processing and analysis of multiparameter flow cytometry samples. As a first test of the FC-HCS technique, we used it to screen a 2000-compound library, called the National Cancer Institute (NCI) Diversity Set, to identify agents that would enhance the anti-lymphoma activity of the therapeutic monoclonal antibody rituximab. FC-HCS identified 15 compounds from the Diversity Set that significantly enhanced the ability of rituximab to inhibit cell cycle progression and induce apoptosis in lymphoma cells. The validity of the screening results was confirmed for several compounds using additional assays of cell proliferation, apoptosis and cell growth. The FC-HCS technique was relatively simple and reliable and could process up to 1000 samples/day on a single flow cytometer. The FC-HCS technique may be useful for a variety of applications including drug discovery, immunologic monitoring of patients, functional genomics studies and tissue engineering efforts.


Assuntos
Anticorpos Monoclonais/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Citometria de Fluxo/métodos , Linfoma/tratamento farmacológico , Anticorpos Monoclonais Murinos , Afidicolina/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Linfoma/patologia , Fenantrolinas/farmacologia , Rituximab , Topotecan/farmacologia
18.
Cancer Res ; 64(10): 3586-92, 2004 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15150116

RESUMO

A small synthetic library of cyclohexapeptidomimetic calixarenes was prepared to identify disrupters of vascular endothelial growth factor (VEGF) binding to its receptor that inhibits angiogenesis. From this library, we discovered GFA-116, which potently inhibits (125)I-VEGF binding to Flk-1 in Flk-1-overexpressing NIH 3T3 cells and human prostate tumor cells with an IC(50) of 750 nM. This inhibition is highly selective for VEGF in that (125)I- platelet-derived growth factor binding to its receptor is not affected. GFA-116 inhibits VEGF-stimulated Flk-1 tyrosine phosphorylation and subsequent activation of Erk1/2 mitogen-activated protein kinases. Furthermore, epidermal growth factor, platelet-derived growth factor, and fibroblast growth factor-dependent stimulation of Erk1/2 phosphorylation are not affected at concentrations as high as 10 microM. In vitro, GFA-116 inhibits angiogenesis as measured by inhibition of migration and formation of capillary-like structures by human endothelial cells as well as suppression of microvessel outgrowth in rat aortic rings and rat cornea angiogenesis. In vivo, GFA-116 (50 mpk/day) inhibits tumor growth and angiogenesis as measured by CD31 staining of A-549 human lung tumors in nude mice. Furthermore, GFA-116 is also effective at inhibiting tumor growth and metastasis to the lung of B16-F10 melanoma cells injected into immunocompetent mice. Taken together, these results demonstrate that a synthetic molecule capable of disrupting the binding of VEGF to its receptor selectively inhibits VEGF-dependent signaling and suppresses angiogenesis and tumorigenesis.


Assuntos
Inibidores da Angiogênese/farmacologia , Benzoatos/farmacologia , Neovascularização Patológica/tratamento farmacológico , Peptídeos Cíclicos/farmacologia , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Encéfalo/irrigação sanguínea , Linhagem Celular Tumoral , Córnea/irrigação sanguínea , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/crescimento & desenvolvimento , Endotélio Vascular/metabolismo , Humanos , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Masculino , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/patologia , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Células NIH 3T3 , Neovascularização Patológica/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Fosforilação , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Ratos , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Prog Cell Cycle Res ; 5: 211-7, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14593715

RESUMO

The presence of mutated Ras in more that 30% of human cancers has spurred interest in the identification of molecules that can block its uncontrolled signaling function. A particular focus in recent years has been a key posttranslational modification of Ras that places a farnesyl group on a cysteine residue near the C-terminus of the protein. In this chapter we describe recent progress in the design of inhibitors for the enzyme that catalyzes this step, protein farnesyltransferase, and show their potential for blocking oncogenic cell growth.


Assuntos
Alquil e Aril Transferases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Neoplasias/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Proteínas ras/metabolismo , Alquil e Aril Transferases/metabolismo , Animais , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Inibidores Enzimáticos/uso terapêutico , Humanos , Estrutura Molecular , Neoplasias/enzimologia , Prenilação de Proteína/efeitos dos fármacos , Prenilação de Proteína/fisiologia , Transdução de Sinais/fisiologia
20.
Cancer Res ; 63(6): 1270-9, 2003 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-12649187

RESUMO

Constitutively activated, tyrosine-phosphorylated signal transducer and activator of transcription (STAT) 3 plays a pivotal role in human tumor malignancy. To discover disrupters of aberrant STAT3 signaling pathways as novel anticancer drugs, we developed a phosphotyrosine STAT3 cytoblot. Using this high throughput 96-well plate assay, we identified JSI-124 (cucurbitacin I) from the National Cancer Institute Diversity Set. JSI-124 suppressed the levels of phosphotyrosine STAT3 in v-Src-transformed NIH 3T3 cells and human cancer cells potently (IC(50) value of 500 nM in the human lung adenocarcinoma A549) and rapidly (complete inhibition within 1-2 h). The suppression of phosphotyrosine STAT3 levels resulted in the inhibition of STAT3 DNA binding and STAT3-mediated but not serum response element-mediated gene transcription. JSI-124 also decreased the levels of tyrosine-phosphorylated Janus kinase (JAK) but not those of Src. JSI-124 was highly selective for JAK/STAT3 and did not inhibit other oncogenic and tumor survival pathways such as those mediated by Akt, extracellular signal-regulated kinase 1/2, or c-Jun NH(2)-terminal kinase. Finally, JSI-124 (1 mg/kg/day) potently inhibited the growth in nude mice of A549 tumors, v-Src-transformed NIH 3T3 tumors, and the human breast carcinoma MDA-MB-468, all of which express high levels of constitutively activated STAT3, but it did not affect the growth of oncogenic Ras-transformed NIH 3T3 tumors that are STAT3 independent or of the human lung adenocarcinoma Calu-1, which has barely detectable levels of phosphotyrosine STAT3. JSI-124 also inhibited tumor growth and significantly increased survival of immunologically competent mice bearing murine melanoma with constitutively activated STAT3. These results give strong support for pharmacologically targeting the JAK/STAT3 signaling pathway for anticancer drug discovery.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas Serina-Treonina Quinases , Proteínas Tirosina Quinases/antagonistas & inibidores , Transativadores/antagonistas & inibidores , Triterpenos/farmacologia , Células 3T3 , Animais , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Humanos , Janus Quinase 2 , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Melanoma Experimental/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Fosforilação , Fosfotirosina/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Fator de Transcrição STAT3 , Transdução de Sinais/efeitos dos fármacos , Transativadores/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...