Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 109(41): 16666-71, 2012 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-23012407

RESUMO

Hutchinson-Gilford progeria syndrome (HGPS) is an extremely rare, fatal, segmental premature aging syndrome caused by a mutation in LMNA that produces the farnesylated aberrant lamin A protein, progerin. This multisystem disorder causes failure to thrive and accelerated atherosclerosis leading to early death. Farnesyltransferase inhibitors have ameliorated disease phenotypes in preclinical studies. Twenty-five patients with HGPS received the farnesyltransferase inhibitor lonafarnib for a minimum of 2 y. Primary outcome success was predefined as a 50% increase over pretherapy in estimated annual rate of weight gain, or change from pretherapy weight loss to statistically significant on-study weight gain. Nine patients experienced a ≥50% increase, six experienced a ≥50% decrease, and 10 remained stable with respect to rate of weight gain. Secondary outcomes included decreases in arterial pulse wave velocity and carotid artery echodensity and increases in skeletal rigidity and sensorineural hearing within patient subgroups. All patients improved in one or more of these outcomes. Results from this clinical treatment trial for children with HGPS provide preliminary evidence that lonafarnib may improve vascular stiffness, bone structure, and audiological status.


Assuntos
Inibidores Enzimáticos/uso terapêutico , Farnesiltranstransferase/antagonistas & inibidores , Piperidinas/uso terapêutico , Progéria/tratamento farmacológico , Piridinas/uso terapêutico , Adolescente , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/patologia , Criança , Pré-Escolar , Diarreia/induzido quimicamente , Relação Dose-Resposta a Droga , Esquema de Medicação , Inibidores Enzimáticos/efeitos adversos , Inibidores Enzimáticos/farmacocinética , Farnesiltranstransferase/metabolismo , Fadiga/induzido quimicamente , Feminino , Humanos , Masculino , Piperidinas/efeitos adversos , Piperidinas/farmacocinética , Progéria/patologia , Progéria/fisiopatologia , Análise de Onda de Pulso , Piridinas/efeitos adversos , Piridinas/farmacocinética , Resultado do Tratamento , Vômito/induzido quimicamente , Aumento de Peso/efeitos dos fármacos
2.
J Biol Chem ; 285(14): 10198-212, 2010 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-20124408

RESUMO

Abrogation of p53 function occurs in almost all human cancers, with more than 50% of cancers harboring inactivating mutations in p53 itself. Mutation of p53 is indicative of highly aggressive cancers and poor prognosis. The vast majority of mutations in p53 occur in its core DNA binding domain (DBD) and result in inactivation of p53 by reducing its thermodynamic stability at physiological temperature. Here, we report a small molecule, SCH529074, that binds specifically to the p53 DBD in a saturable manner with an affinity of 1-2 microm. Binding restores wild type function to many oncogenic mutant forms of p53. This small molecule reactivates mutant p53 by acting as a chaperone, in a manner similar to that previously reported for the peptide CDB3. Binding of SCH529074 to the p53 DBD is specifically displaced by an oligonucleotide with a sequence derived from the p53-response element. In addition to reactivating mutant p53, SCH529074 binding inhibits ubiquitination of p53 by HDM2. We have also developed a novel variant of p53 by changing a single amino acid in the core domain of p53 (N268R), which abolishes binding of SCH529074. This amino acid change also inhibits HDM2-mediated ubiquitination of p53. Our novel findings indicate that through its interaction with p53 DBD, SCH529074 restores DNA binding activity to mutant p53 and inhibits HDM2-mediated ubiquitination.


Assuntos
Proliferação de Células/efeitos dos fármacos , DNA/metabolismo , Mutação/genética , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Quinazolinas/farmacologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Ubiquitinação , Animais , Apoptose/efeitos dos fármacos , Sítios de Ligação , Western Blotting , Imunoprecipitação da Cromatina , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/prevenção & controle , DNA/química , DNA/genética , Feminino , Humanos , Imunoprecipitação , Camundongos , Camundongos Nus , Chaperonas Moleculares , Piperazinas/isolamento & purificação , Ligação Proteica , Proteínas Proto-Oncogênicas c-mdm2/genética , Quinazolinas/isolamento & purificação , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Am J Med Genet C Semin Med Genet ; 157C(2): 136-46, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-21495172

RESUMO

The RASopathies, one of the largest groups of multiple congenital anomaly syndromes known, are caused by germline mutations in various genes encoding components of the Ras/mitogen-activated protein kinase (MAPK) pathway. The RASopathies have many overlapping characteristics, including craniofacial manifestations, cardiac malformations, cutaneous, musculoskeletal, gastrointestinal, and ocular abnormalities, neurocognitive impairment, hypotonia, and an increased risk of developing cancer. Costello syndrome (CS) and cardio-facio-cutaneous (CFC) syndrome are two of the more rare RASopathies. CS is caused by activating mutations in HRAS, and CFC is caused by dysregulation of signaling in the Ras/MAPK pathway due to mutations in BRAF, MEK1, or MEK2. The Ras/MAPK pathway, which has been well-studied in cancer, is an attractive target for inhibition in the treatment of various malignancies utilizing small molecule therapeutics that specifically inhibit the pathway. With many inhibitors of the Ras/MAPK pathway in clinical trials, the notion of using these molecules to ameliorate developmental defects in CS and CFC is under consideration. CS and CFC, like other syndromes in their class, have a progressive phenotype and may be amenable to inhibition or normalization of signaling.


Assuntos
Ensaios Clínicos como Assunto/métodos , Síndrome de Costello/genética , Neurofibromatose 1/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Projetos de Pesquisa , Transdução de Sinais/genética , Síndrome de Costello/tratamento farmacológico , Displasia Ectodérmica/tratamento farmacológico , Displasia Ectodérmica/genética , Fácies , Insuficiência de Crescimento/tratamento farmacológico , Insuficiência de Crescimento/genética , Farnesiltranstransferase/antagonistas & inibidores , Cardiopatias Congênitas/tratamento farmacológico , Cardiopatias Congênitas/genética , Humanos , MAP Quinase Quinase 1/antagonistas & inibidores , Parcerias Público-Privadas , Quinases raf/antagonistas & inibidores
4.
Bioorg Med Chem Lett ; 20(3): 1134-6, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20056542

RESUMO

The discovery of C-linked imidazole azaheptapyridine bridgehead FPT inhibitors is described. This novel class of compounds are sub nM FPT enzyme inhibitors with potent cellular inhibitory activities. This series also has reduced hERG activity versus previous N-linked imidazole series. X-ray of compound 10a bound to FTase revealed strong interaction between bridgehead imidazole 3N with catalytic zinc atom.


Assuntos
Descoberta de Drogas/métodos , Farnesiltranstransferase/antagonistas & inibidores , Imidazóis/química , Piridinas/química , Linhagem Celular Tumoral , Cristalografia por Raios X , Farnesiltranstransferase/metabolismo , Humanos , Imidazóis/metabolismo , Imidazóis/farmacologia , Piridinas/metabolismo , Piridinas/farmacologia
5.
Int J Cancer ; 125(11): 2711-20, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19530253

RESUMO

Lonafarnib is a potent, selective farnesyltransferase inhibitor (FTI) undergoing clinical studies for the treatment of solid tumors and hematological malignancies. Preclinically, a number of FTIs, including lonafarnib, interact with taxanes to inhibit cancer cell growth in an additive/synergistic manner. These observations provided rationale for investigating the effects of combining lonafarnib and docetaxel on preclinical prostate cancer models. To date, docetaxel is the only chemotherapeutic agent in clinical use for hormone-refractory prostate cancer. In vitro experiments with 22Rv1, LNCaP, DU-145, PC3 and PC3-M prostate cancer cell lines showed significantly enhanced inhibition of cell proliferation and apoptosis when lonafarnib was added to docetaxel. In human tumor xenograft models, continuous coadministration of lonafarnib with docetaxel caused marked tumor regressions (24-47%) in tumors from all of the cell types as well as parental CWR22 xenografts. Intermittent dosing of lonafarnib (5 days on then 5 days off) coadministered with docetaxel produced similar regressions in hormone-refractory 22Rv1 tumors. 22Rv1 tumors progressing on docetaxel treatment also responded to treatment with intermittent lonafarnib (5 days on then 5 days off). Moreover, animals did not exhibit any signs of toxicity during coadministration of lonafarnib and docetaxel. In conclusion, coadministration of continuous and intermittent lonafarnib enhanced the antitumor activity of docetaxel in a panel of prostate cancer models. An intermittent dosing schedule of lonafarnib coadministered with docetaxel may allow enhanced efficacy to that of continuous dosing by improving the tolerability of higher doses of lonafarnib.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Piperidinas/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Piridinas/uso terapêutico , Taxoides/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Western Blotting , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Docetaxel , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , Masculino , Camundongos , Camundongos Nus , Camundongos SCID , Neoplasias Hormônio-Dependentes/sangue , Neoplasias Hormônio-Dependentes/patologia , Piperidinas/sangue , Piperidinas/farmacocinética , Neoplasias da Próstata/sangue , Neoplasias da Próstata/patologia , Piridinas/sangue , Piridinas/farmacocinética
6.
Gynecol Oncol ; 109(1): 97-106, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18237771

RESUMO

OBJECTIVES: To determine the effects of combining lonafarnib with paclitaxel on the growth of human ovarian cancer cells and tumor xenografts as well as to monitor a pharmacodynamic marker of farnesyltransferase inhibition (HDJ-2) in peripheral blood mononuclear cells (PBMCs) isolated from tumor-bearing animals after treatment with this combination. METHODS: Proliferation of A2780, PA-1, IGROV-1, and TOV-112D cells was assessed after treatment with lonafarnib and paclitaxel. Cell cycle progression was determined by flow cytometry, and apoptosis was evaluated by assaying for caspase-3 and cleaved PARP. The effects of lonafarnib and paclitaxel on the tumor growth of each model were determined in immunocompromised mice. Proteins extracted from cells, tumors, and PBMCs were assayed for HDJ-2 mobility shifts by Western blotting as well as for farnesyl protein transferase (FTase) enzyme activity by biochemical analyses. RESULTS: In A2780, PA-1, IGROV-1, and TOV-112D cells lonafarnib potentiated the growth inhibitory effects of paclitaxel. In each of the models lonafarnib enhanced paclitaxel-induced mitotic arrest and apoptosis. The combination of lonafarnib plus paclitaxel resulted in marked tumor regressions in A2780, TOV-112D, PA-1, and IGROV-1 tumor xenografts. Western blotting demonstrated that in PBMCs isolated from the animals, paclitaxel treatment suppressed lonafarnib-induced HDJ-2 mobility shifts. Paclitaxel did not affect lonafarnib inhibition of FTase enzyme activity levels in these PBMCs. CONCLUSIONS: Lonafarnib enhances the antiproliferative effects of paclitaxel on ovarian cancer cells in vitro and ovarian tumor xenografts in vivo. Measuring FTase enzyme activity levels rather than HDJ-2 shifts in PBMCs may be a more accurate biomarker to predict levels of farnesyltransferase inhibition in patients who are also receiving paclitaxel chemotherapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Farnesiltranstransferase/antagonistas & inibidores , Neoplasias Ovarianas/tratamento farmacológico , Piperidinas/farmacologia , Piridinas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/sangue , Ciclo Celular/efeitos dos fármacos , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Farnesiltranstransferase/sangue , Farnesiltranstransferase/metabolismo , Feminino , Proteínas de Choque Térmico HSP40/metabolismo , Humanos , Leucócitos Mononucleares/enzimologia , Camundongos , Camundongos Nus , Camundongos SCID , Neoplasias Ovarianas/sangue , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/patologia , Paclitaxel/administração & dosagem , Piperidinas/administração & dosagem , Piridinas/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
7.
JCI Insight ; 3(4)2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29467321

RESUMO

BACKGROUND: Constitutive activation of ERK1/2 occurs in various cancers, and its reactivation is a well-described resistance mechanism to MAPK inhibitors. ERK inhibitors may overcome the limitations of MAPK inhibitor blockade. The dual mechanism inhibitor SCH772984 has shown promising preclinical activity across various BRAFV600/RAS-mutant cancer cell lines and human cancer xenografts. METHODS: We have developed an orally bioavailable ERK inhibitor, MK-8353; conducted preclinical studies to demonstrate activity, pharmacodynamic endpoints, dosing, and schedule; completed a study in healthy volunteers (P07652); and subsequently performed a phase I clinical trial in patients with advanced solid tumors (MK-8353-001). In the P07652 study, MK-8353 was administered as a single dose in 10- to 400-mg dose cohorts, whereas in the MK-8353-001 study, MK-8353 was administered in 100- to 800-mg dose cohorts orally twice daily. Safety, tolerability, pharmacokinetics, pharmacodynamics, and antitumor activity were analyzed. RESULTS: MK-8353 exhibited comparable potency with SCH772984 across various preclinical cancer models. Forty-eight patients were enrolled in the P07652 study, and twenty-six patients were enrolled in the MK-8353-001 study. Adverse events included diarrhea (44%), fatigue (40%), nausea (32%), and rash (28%). Dose-limiting toxicity was observed in the 400-mg and 800-mg dose cohorts. Sufficient exposure to MK-8353 was noted that correlated with biological activity in preclinical data. Three of fifteen patients evaluable for treatment response in the MK-8353-001 study had partial response, all with BRAFV600-mutant melanomas. CONCLUSION: MK-8353 was well tolerated up to 400 mg twice daily and exhibited antitumor activity in patients with BRAFV600-mutant melanoma. However, antitumor activity was not particularly correlated with pharmacodynamic parameters. TRIAL REGISTRATION: ClinicalTrials.gov NCT01358331. FUNDING: Merck Sharp & Dohme Corp., a subsidiary of Merck & Co. Inc., and NIH (P01 CA168585 and R35 CA197633).


Assuntos
Indazóis/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Pirrolidinas/farmacologia , Triazóis/farmacologia , Administração Oral , Adulto , Animais , Disponibilidade Biológica , Linhagem Celular Tumoral , Diarreia/induzido quimicamente , Diarreia/epidemiologia , Cães , Relação Dose-Resposta a Droga , Toxidermias/epidemiologia , Toxidermias/etiologia , Avaliação Pré-Clínica de Medicamentos , Fadiga/induzido quimicamente , Fadiga/epidemiologia , Feminino , Humanos , Indazóis/uso terapêutico , Masculino , Dose Máxima Tolerável , Camundongos , Pessoa de Meia-Idade , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Náusea/induzido quimicamente , Náusea/epidemiologia , Estadiamento de Neoplasias , Neoplasias/genética , Neoplasias/patologia , Inibidores de Proteínas Quinases/uso terapêutico , Piridinas/uso terapêutico , Pirrolidinas/uso terapêutico , Ratos , Triazóis/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto , Adulto Jovem
8.
Mol Cancer Ther ; 4(8): 1214-21, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16093437

RESUMO

Insulin-like growth factor-I receptor (IGF-IR) plays an important role in tumor cell growth and survival. On ligand stimulation, IGF-IR, a receptor tyrosine kinase, phosphorylates tyrosine residues on two major substrates, IRS-1 and Shc, which subsequently signal through the Ras/mitogen-activated protein kinase and phosphatidylinositol 3-kinase/AKT pathways. Here, we describe the characterization of a fully human anti-IGF-IR monoclonal antibody 19D12 that inhibits IGF binding and autophosphorylation of both IGF-IR/IGF-IR homodimers and IGF-IR/insulin receptor heterodimers. 19D12 does not recognize insulin receptor homodimers. In addition to inhibiting IGF-IR autophosphorylation, 19D12 also inhibits IRS-1 phosphorylation and activation of the major downstream signaling molecules AKT and extracellular signal-regulated kinase 1/2. Furthermore, the antibody down-regulates the total IGF-IR protein level and can exhibit antibody-dependent cellular cytotoxicity activity against a non-small cell adenocarcinoma cell line in vitro in the presence of isolated human natural killer cells. 19D12 binds tightly to the receptor, with an affinity of 3.8 pmol/L as measured by KinExA. In cell culture, 19D12 inhibits proliferation and soft agar growth of various tumor cell lines. In vivo, 19D12 inhibits the tumor growth of a very aggressive human ovarian tumor xenograft model A2780. These data support the development of this anti-IGF-IR monoclonal antibody as a promising anticancer agent.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Receptor IGF Tipo 1/antagonistas & inibidores , Animais , Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Dimerização , Regulação para Baixo , Humanos , Proteínas Substratos do Receptor de Insulina , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Neoplasias/metabolismo , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Receptor IGF Tipo 1/imunologia , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
9.
ACS Med Chem Lett ; 7(3): 324-9, 2016 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-26985323

RESUMO

A new subseries of substituted piperidines as p53-HDM2 inhibitors exemplified by 21 has been developed from the initial lead 1. Research focused on optimization of a crucial HDM2 Trp23-ligand interaction led to the identification of 2-(trifluoromethyl)thiophene as the preferred moiety. Further investigation of the Leu26 pocket resulted in potent, novel substituted piperidine inhibitors of the HDM2-p53 interaction that demonstrated tumor regression in several human cancer xenograft models in mice. The structure of HDM2 in complex with inhibitors 3, 10, and 21 is described.

10.
Oncogene ; 22(23): 3645-54, 2003 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-12789273

RESUMO

The temporal gene expression profile during the entire process of apoptosis and cell cycle progression in response to p53 in human ovarian cancer cells was explored with cDNA microarrays representing 33 615 individual human genes. A total of 1501 genes (4.4%) were found to respond to p53 (approximately 80% of these were repressed by p53) using 2.5-fold change as a cutoff. It was anticipated that most of p53 responsive genes resulted from the secondary effect of p53 expression at late stage of apoptosis. To delineate potential p53 direct and indirect target genes during the process of apoptosis and cell cycle progression, microarray data were combined with global p53 DNA-binding site analysis. Here we showed that 361 out of 1501 p53 responsive genes contained p53 consensus DNA-binding sequence(s) in their regulatory region, approximately 80% of which were repressed by p53. This is the first time that a large number of p53-repressed genes have been identified to contain p53 consensus DNA-binding sequence(s) in their regulatory region. Hierarchical cluster analysis of these genes revealed distinct temporal expression patterns of transcriptional activation and repression by p53. More genes were activated at early time points, while more repressed genes were found after the onset of apoptosis. A small-scale quantitative chromatin immunoprecipitation analysis indicated that in vivo p53-DNA interaction was detected in eight out of 10 genes, most of which were repressed by p53 at the early onset of apoptosis, suggesting that a portion of p53 target genes in the human genome could be negatively regulated by p53 via sequence-specific DNA binding. The approaches and genes described here should aid the understanding of global gene regulatory network of p53.


Assuntos
Apoptose/genética , Ciclo Celular/genética , Neoplasias Ovarianas/genética , Transcrição Gênica , Proteína Supressora de Tumor p53/genética , Adenoviridae/genética , Sequência de Aminoácidos , Sítios de Ligação , Cromatina/metabolismo , Análise por Conglomerados , Sequência Conservada , Feminino , Perfilação da Expressão Gênica , Genoma Humano , Humanos , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Ovarianas/patologia , Testes de Precipitina , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transfecção/métodos , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo
11.
Oncogene ; 21(17): 2613-22, 2002 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-11965534

RESUMO

Survivin is an inhibitor of apoptosis protein, which is over-expressed in most tumors. Aberrant expression of survivin and loss of wild-type p53 in many tumors prompted us to investigate a possible link between these two events. Here we show that wild-type p53 represses survivin expression at both mRNA and protein levels. Transient transfection analyses revealed that the expression of wild-type p53, but not mutant p53, was associated with strong repression of the survivin promoter in various cell types. The over-expression of exogenous survivin protein rescues cells from p53-induced apoptosis in a dose-dependent manner, suggesting that loss of survivin mediates, at least, in part the p53-dependent apoptotic pathway. In spite of the presence of two putative p53-binding sites in the survivin promoter, deletion and mutation analyses suggested that neither site is required for transcriptional repression of survivin expression. This was confirmed by chromatin immunoprecipitation assays. Further analyses suggested that the modification of chromatin within the survivin promoter could be a molecular explanation for silencing of survivin gene transcription by p53.


Assuntos
Apoptose , Proteínas Cromossômicas não Histona/genética , Inibidores de Cisteína Proteinase/genética , Proteínas Associadas aos Microtúbulos , Proteína Supressora de Tumor p53/genética , Adenoviridae , Antibióticos Antineoplásicos/farmacologia , Sequência de Bases , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Inibidores de Cisteína Proteinase/metabolismo , Primers do DNA/química , Doxorrubicina/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas Inibidoras de Apoptose , Luciferases/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Dados de Sequência Molecular , Proteínas de Neoplasias , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Testes de Precipitina , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , RNA Neoplásico/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Survivina , Transcrição Gênica , Transfecção , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo
12.
Clin Cancer Res ; 10(9): 2968-76, 2004 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15131032

RESUMO

PURPOSE: To establish the maximum tolerated dose of lonafarnib, a novel farnesyltransferase inhibitor, in combination with paclitaxel in patients with solid tumors and to characterize the safety, tolerability, dose-limiting toxicity, and pharmacokinetics of this combination regimen. EXPERIMENTAL DESIGN: In a Phase I trial, lonafarnib was administered p.o., twice daily (b.i.d.) on continuously scheduled doses of 100 mg, 125 mg, and 150 mg in combination with i.v. paclitaxel at doses of 135 mg/m(2) or 175 mg/m(2) administered over 3 h on day 8 of every 21-day cycle. Plasma paclitaxel and lonafarnib concentrations were collected at selected time points from each patient. RESULTS: Twenty-four patients were enrolled; 21 patients were evaluable. The principal grade 3/4 toxicity was diarrhea (5 of 21 patients), which was most likely due to lonafarnib. dose-limiting toxicities included grade 3 hyperbilirubinemia at dose level 3 (100 mg b.i.d. lonafarnib and 175 mg/m(2) paclitaxel); grade 4 diarrhea and grade 3 peripheral neuropathy at dose level 3A (125 mg b.i.d. lonafarnib and 175 mg/m(2) paclitaxel); and grade 4 neutropenia with fever and grade 4 diarrhea at level 4 (150 mg b.i.d. lonafarnib and 175 mg/m(2) paclitaxel). The maximum tolerated dose established by the continual reassessment method was lonafarnib 100 mg b.i.d. and paclitaxel 175 mg/m(2). Paclitaxel appeared to have no effect on the pharmacokinetics of lonafarnib. The median duration of therapy was eight cycles, including seven cycles with paclitaxel. Six of 15 previously treated patients had a durable partial response, including 3 patients who had previous taxane therapy. Notably, two of five patients with taxane-resistant metastatic non-small cell lung cancer had partial responses. CONCLUSIONS: When combined with paclitaxel, the recommended dose of lonafarnib for Phase II trials is 100 mg p.o. twice daily with 175 mg/m(2) of paclitaxel i.v. every 3 weeks. Additional studies of lonafarnib in combination regimens appear warranted, particularly in patients with non-small cell lung cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias/tratamento farmacológico , Adulto , Idoso , Alquil e Aril Transferases/antagonistas & inibidores , Anemia/induzido quimicamente , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Área Sob a Curva , Relação Dose-Resposta a Droga , Farnesiltranstransferase , Fadiga/induzido quimicamente , Feminino , Parada Cardíaca/induzido quimicamente , Humanos , Leucopenia/induzido quimicamente , Masculino , Pessoa de Meia-Idade , Neoplasias/metabolismo , Neoplasias/patologia , Neutropenia/induzido quimicamente , Paclitaxel/administração & dosagem , Paclitaxel/efeitos adversos , Paclitaxel/farmacocinética , Piperidinas/administração & dosagem , Piperidinas/efeitos adversos , Piperidinas/farmacocinética , Piridinas/administração & dosagem , Piridinas/efeitos adversos , Piridinas/farmacocinética , Resultado do Tratamento
13.
Mol Cancer Ther ; 2(3): 219-24, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12657715

RESUMO

BCR-ABL fusion proteins exhibit elevated tyrosine kinase activity and transforming properties. Genetic and biochemical data suggest that Ras activation plays a central role in leukemogenic transformation by BCR-ABL. Imatinib (Novartis, Basel, Switzerland) is a potent and selective inhibitor of the tyrosine kinase activity of BCR-ABL. Although imatinib has shown promise against Ph-positive leukemia in human clinical trials, the emergence of imatinib resistance in patients with acute forms of Ph-positive leukemia has highlighted the need for combination chemotherapy to eradicate this disease. In the present study, combined use of a farnesyl transferase inhibitor, SCH66336 (lonafarnib), with the antileukemic agents imatinib, daunorubicin, cytosine arabinoside, or etoposide was investigated by cell proliferation assays. The effects of the combination of SCH66336 and imatinib were also investigated by apoptosis assay and colony-forming assay. In proliferation assays with BCR-ABL-expressing cells, combination of SCH66336 with imatinib or cytosine arabinoside showed enhanced antiproliferative activity, whereas combination of SCH66336 with daunorubicin or etoposide demonstrated an antagonistic effect. The combination of imatinib plus SCH66336 more effectively inhibited hematopoietic colony formation by primary human chronic myelogenous leukemia cells. SCH66336 combined with imatinib was shown to induce apoptosis in imatinib-resistant BCR-ABL cells by flow cytometric analysis with an APO2.7 monoclonal antibody. These results indicate that SCH66336 is a promising candidate for use in the treatment of patients with imatinib-resistant, Ph-positive leukemia and that the combination of SCH66336 plus imatinib may be useful to circumvent resistance.


Assuntos
Alquil e Aril Transferases/antagonistas & inibidores , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Proteínas de Fusão bcr-abl/metabolismo , Leucemia Mieloide Aguda/patologia , Piperazinas/administração & dosagem , Piperidinas/administração & dosagem , Piridinas/administração & dosagem , Pirimidinas/administração & dosagem , Apoptose/efeitos dos fármacos , Benzamidas , Divisão Celular/efeitos dos fármacos , Ensaio de Unidades Formadoras de Colônias , Citarabina/farmacologia , Daunorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Inibidores Enzimáticos/farmacologia , Etoposídeo/farmacologia , Farnesiltranstransferase , Proteínas de Fusão bcr-abl/genética , Humanos , Mesilato de Imatinib , Interleucina-3/farmacologia , Leucemia Mieloide Aguda/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Transfecção , Resultado do Tratamento , Células Tumorais Cultivadas
14.
Mol Cancer Ther ; 1(14): 1349-53, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12516969

RESUMO

Insulin-like growth factor receptor 1 (IGFR1) plays a crucial role in oncogenic transformation [C. Sell et al., Mol. Cell. Biol., 14: 3604-3612,1994]. Compared with the normal human mammary epithelial cell line MCF12A, MCF7 human mammary carcinoma cells overexpress IGFR1 on the cell surface. To measure the effects of IGFR1 inhibition on tumor cells, we tested two mouse neutralizing antibodies against human IGFR1 in cell-based assays. Both MAB391 and anti-IR3 antibodies inhibit IGFR1 autophosphorylation upon IGF-I ligand stimulation with IC50s of 0.58 and 0.80 nM, respectively. When cells were treated with neutralizing anti-IGFR1 antibodies for > or = 4 h, the total receptor level was dramatically decreased. IGF-I-stimulated activation of AKT was also inhibited by anti-IGFR1 antibodies. Furthermore, MAB391 and anti-IR3 inhibited the growth of MCF7 cells in soft agar. In addition to MCF7 cells, MAB391 also inhibited IGFR1 autophosphorylation and induced IGFR1 down-modulation in HT29 colorectal and Du145 prostate cancer cells. Therefore, neutralizing antibodies against IGFR1 represent a valid approach to inhibit growth of tumor cells.


Assuntos
Receptor IGF Tipo 1/química , Receptor IGF Tipo 1/imunologia , Ágar/farmacologia , Anticorpos Monoclonais/farmacologia , Western Blotting , Transformação Celular Neoplásica , Relação Dose-Resposta a Droga , Regulação para Baixo , Eletroforese em Gel de Poliacrilamida , Humanos , Concentração Inibidora 50 , Fator de Crescimento Insulin-Like I/metabolismo , Lisossomos/metabolismo , Fosforilação , Testes de Precipitina , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais , Fatores de Tempo , Células Tumorais Cultivadas
15.
Curr Opin Drug Discov Devel ; 7(4): 478-86, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15338957

RESUMO

Farnesyltransferase (FT) inhibitors were originally designed as anticancer agents, and were thought to act by inhibiting the farnesylation of mutant Ras proteins. However, these compounds were subsequently demonstrated to have antitumor effects even in the absence of Ras mutations and it has now become clear that other protein targets are involved. This article discusses the preclinical and clinical development of FT inhibitors. To date, tipifarnib (Zarnestra; Janssen Pharmaceutica NV) and lonafarnib (Sarasar; Schering-Plough Research Institute) are the only two FT inhibitors to have been evaluated in phase III clinical trials. The clinical results of these two compounds are presented below, with emphasis on ways of enhancing the possibility of a successful FT inhibitor anticancer drug. Details of new FT inhibitors disclosed since the beginning of 2003 are also included.


Assuntos
Alquil e Aril Transferases/antagonistas & inibidores , Alquil e Aril Transferases/uso terapêutico , Antineoplásicos/uso terapêutico , Alquil e Aril Transferases/farmacologia , Animais , Antineoplásicos/farmacologia , Ensaios Clínicos Fase II como Assunto , Ensaios Clínicos Fase III como Assunto , Sistemas de Liberação de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Farnesiltranstransferase , Humanos , Estrutura Molecular , Proteínas Oncogênicas/biossíntese , Proteínas Oncogênicas/efeitos dos fármacos
16.
Cancer Discov ; 3(7): 742-50, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23614898

RESUMO

The high frequency of activating RAS or BRAF mutations in cancer provides strong rationale for targeting the mitogen-activated protein kinase (MAPK) pathway. Selective BRAF and MAP-ERK kinase (MEK) inhibitors have shown clinical efficacy in patients with melanoma. However, the majority of responses are transient, and resistance is often associated with pathway reactivation of the extracellular signal-regulated kinase (ERK) signaling pathway. Here, we describe the identification and characterization of SCH772984, a novel and selective inhibitor of ERK1/2 that displays behaviors of both type I and type II kinase inhibitors. SCH772984 has nanomolar cellular potency in tumor cells with mutations in BRAF, NRAS, or KRAS and induces tumor regressions in xenograft models at tolerated doses. Importantly, SCH772984 effectively inhibited MAPK signaling and cell proliferation in BRAF or MEK inhibitor-resistant models as well as in tumor cells resistant to concurrent treatment with BRAF and MEK inhibitors. These data support the clinical development of ERK inhibitors for tumors refractory to MAPK inhibitors.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinase Quinase Quinases/genética , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , MAP Quinase Quinase Quinases/antagonistas & inibidores , Mutação , Neoplasias/tratamento farmacológico , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos
17.
Mol Cancer Ther ; 9(2): 410-8, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20124453

RESUMO

The insulin-like growth factor-I receptor (IGF-IR) and its ligands (IGF-I and IGF-II) have been implicated in the growth, survival, and metastasis of a broad range of malignancies including pediatric tumors. Blocking the IGF-IR action is a potential cancer treatment. A fully human neutralizing monoclonal antibody, SCH 717454 (19D12, robatumumab), specific to IGF-IR, has shown potent antitumor effects in ovarian cancer in vitro and in vivo. In this study, SCH 717454 was evaluated in several pediatric solid tumors including neuroblastoma, osteosarcoma, and rhabdomyosarcoma. SCH 717454 is shown here to downregulate IGF-IR as well as inhibit IGF-IR and insulin receptor substrate-1 phosphorylation in pediatric tumor cells. IGF-IR and insulin receptor substrate-1 phosphorylation in the tumor cells. In vivo, SCH 717454 exhibits activity as a single agent and significantly inhibited growth of neuroblastoma, osteosarcoma, and rhabdomyosarcoma tumor xenografts. Combination of SCH 717454 with cisplatin or cyclophosphamide enhanced both the degree and the duration of the in vivo antitumor activity compared with single-agent treatments. Furthermore, SCH 717454 treatment markedly reduced Ki-67 expression and blood vessel formation in tumor xenografts, showing that the in vivo activity is derived from its inhibition of tumor cell proliferation and angiogenesis activity.


Assuntos
Anticorpos Monoclonais/química , Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Receptor IGF Tipo 1/imunologia , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Fator de Crescimento Insulin-Like I/imunologia , Antígeno Ki-67/biossíntese , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neovascularização Patológica , Fosforilação
19.
J Lipid Res ; 47(1): 15-31, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16278491

RESUMO

Some proteins undergo posttranslational modification by the addition of an isoprenyl lipid (farnesyl- or geranylgeranyl-isoprenoid) to a cysteine residue proximal to the C terminus. Protein isoprenylation promotes membrane association and contributes to protein-protein interactions. Farnesylated proteins include small GTPases, tyrosine phosphatases, nuclear lamina, cochaperones, and centromere-associated proteins. Prenylation is required for the transforming activity of Ras. Because of the high frequency of Ras mutations in cancer, farnesyl transferase inhibitors (FTIs) were investigated as a means to antagonize Ras function. Evaluation of FTIs led to the finding that both K- and N-Ras are alternatively modified by geranylgeranyl prenyltransferase-1 in FTI-treated cells. Geranylgeranylated forms of Ras retain the ability to associate with the plasma membrane and activate substrates. Despite this, FTIs are effective at inhibiting the growth of human tumor cells in vitro, suggesting that activity is dependent on blocking the farnesylation of other proteins. FTIs also inhibit the in vivo growth of human tumor xenografts and sensitize these models to chemotherapeutics, most notably taxanes. Several FTIs have entered clinical trials for various cancer indications. In some clinical settings, primarily hematologic malignancies, FTIs have displayed evidence of single-agent activity. Clinical studies in progress are exploring the antitumor activity of FTIs as single agents and in combination. This review will summarize the basic biology of FTIs, their antitumor activity in preclinical models, and the current status of clinical studies with these agents.


Assuntos
Farnesiltranstransferase/antagonistas & inibidores , Metabolismo dos Lipídeos , Processamento de Proteína Pós-Traducional , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Prenilação de Proteína/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas ras/metabolismo
20.
Bioorg Med Chem Lett ; 16(4): 984-8, 2006 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-16298128

RESUMO

Benzocycloheptapyridine tricyclic compounds with piperazine or substituted piperidine moieties extending either from the 5- or 6-position of the tricyclic bridgehead exhibited enhanced FTase activity: this resulted from favorable binding of the ligand nitrogen with the catalytic zinc found in the FTase. A single isomer at C-11 with piperazine adduct extending from the 6-position, compound 24, exhibited excellent FTase activity with IC50 = 0.007 microM, soft agar IC50 = 72 nM, and Rat AUC(PO, 10 mpk) = 4.0 microM x h. X-ray of (-)-[8-chloro-6-(1-piperazinyl)-1H-benzo[5,6]]cyclohepta[1,2-b]pyridine-11-yl]-1-(methylsulfonyl)piperidine 24 bound to Ftase revealed favorable interaction between piperazine nitrogen and catalytic zinc atom.


Assuntos
Alquil e Aril Transferases/química , Compostos Organometálicos/química , Piperazinas/química , Zinco/química , Alquil e Aril Transferases/antagonistas & inibidores , Sítios de Ligação/efeitos dos fármacos , Catálise/efeitos dos fármacos , Cristalografia por Raios X , Ligantes , Modelos Moleculares , Estrutura Molecular , Compostos Organometálicos/farmacologia , Piperazina , Estereoisomerismo , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA