Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(19): 9533-9542, 2019 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-31019077

RESUMO

T cell-invigorating cancer immunotherapies have near-curative potential. However, their clinical benefit is currently limited, as only a fraction of patients respond, suggesting that these regimens may benefit from combination with tumor-targeting treatments. As oncogenic progression is accompanied by alterations in metabolic pathways, tumors often become heavily reliant on antioxidant machinery and may be susceptible to increases in oxidative stress. The cystine-glutamate antiporter xCT is frequently overexpressed in cancer and fuels the production of the antioxidant glutathione; thus, tumors prone to redox stress may be selectively vulnerable to xCT disruption. However, systemic inhibition of xCT may compromise antitumor immunity, as xCT is implicated in supporting antigen-induced T cell proliferation. Therefore, we utilized immune-competent murine tumor models to investigate whether cancer cell expression of xCT was required for tumor growth in vivo and if deletion of host xCT impacted antitumor immune responses. Deletion of xCT in tumor cells led to defective cystine uptake, accumulation of reactive oxygen species, and impaired tumor growth, supporting a cancer cell-autonomous role for xCT. In contrast, we observed that, although T cell proliferation in culture was exquisitely dependent on xCT expression, xCT was dispensable for T cell proliferation in vivo and for the generation of primary and memory immune responses to tumors. These findings prompted the combination of tumor cell xCT deletion with the immunotherapeutic agent anti-CTLA-4, which dramatically increased the frequency and durability of antitumor responses. Together, these results identify a metabolic vulnerability specific to tumors and demonstrate that xCT disruption can expand the efficacy of anticancer immunotherapies.


Assuntos
Sistema y+ de Transporte de Aminoácidos/deficiência , Células Apresentadoras de Antígenos/imunologia , Proliferação de Células , Memória Imunológica , Neoplasias Experimentais/imunologia , Linfócitos T/imunologia , Sistema y+ de Transporte de Aminoácidos/imunologia , Animais , Células Apresentadoras de Antígenos/patologia , Linhagem Celular , Deleção de Genes , Glutationa/genética , Glutationa/imunologia , Imunoterapia , Camundongos , Camundongos Knockout , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Neoplasias Experimentais/terapia , Linfócitos T/patologia
2.
Proc Natl Acad Sci U S A ; 113(1): 182-7, 2016 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-26677873

RESUMO

Macroautophagy is a key stress-response pathway that can suppress or promote tumorigenesis depending on the cellular context. Notably, Kirsten rat sarcoma (KRAS)-driven tumors have been reported to rely on macroautophagy for growth and survival, suggesting a potential therapeutic approach of using autophagy inhibitors based on genetic stratification. In this study, we evaluated whether KRAS mutation status can predict the efficacy to macroautophagy inhibition. By profiling 47 cell lines with pharmacological and genetic loss-of-function tools, we were unable to confirm that KRAS-driven tumor lines require macroautophagy for growth. Deletion of autophagy-related 7 (ATG7) by genome editing completely blocked macroautophagy in several tumor lines with oncogenic mutations in KRAS but did not inhibit cell proliferation in vitro or tumorigenesis in vivo. Furthermore, ATG7 knockout did not sensitize cells to irradiation or to several anticancer agents tested. Interestingly, ATG7-deficient and -proficient cells were equally sensitive to the antiproliferative effect of chloroquine, a lysosomotropic agent often used as a pharmacological tool to evaluate the response to macroautophagy inhibition. Moreover, both cell types manifested synergistic growth inhibition when treated with chloroquine plus the tyrosine kinase inhibitors erlotinib or sunitinib, suggesting that the antiproliferative effects of chloroquine are independent of its suppressive actions on autophagy.


Assuntos
Antineoplásicos/farmacologia , Autofagia/efeitos dos fármacos , Transformação Celular Neoplásica/efeitos dos fármacos , Cloroquina/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Autofagia/genética , Proteína 7 Relacionada à Autofagia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Cloridrato de Erlotinib/farmacologia , Técnicas de Inativação de Genes , Humanos , Indóis/farmacologia , Mutação , Inibidores de Proteínas Quinases/farmacologia , Pirróis/farmacologia , Tolerância a Radiação/genética , Sunitinibe , Enzimas Ativadoras de Ubiquitina/genética
3.
J Biol Chem ; 285(20): 15380-15392, 2010 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-20233713

RESUMO

The mammalian target of rapamycin (mTOR) regulates growth via promoting translation and transcription. Here, employing an mTOR active-site inhibitor WYE-125132 (WYE-132), we have performed quantitative phospho-proteomics and identified a Ser-75-containing phosphopeptide from Maf1, a known repressor of RNA polymerase III (Pol III) transcription. Treatment of cancer cells with WYE-132 or the rapamycin analog CCI-779 led to a rapid loss of the phosphorylation at Ser-75, whereas this effect was not seen in cells treated with cytotoxic agents or unrelated inhibitors. WYE-132-induced Maf1 dephosphorylation correlated with its accumulation in the nucleus and a marked decline in the cellular levels of pre-tRNAs. Depletion of cellular Maf1 via small interfering RNA increased basal pre-tRNA and rendered tRNA synthesis refractory to mTOR inhibitors. Maf1 mutant proteins carrying S75A alone or with S60A, T64A, and S68A (Maf1-S75A, Maf1-4A) progressively enhanced basal repression of tRNA in actively proliferating cells and attenuated amino acid-induced tRNA transcription. Gene alignment revealed conservation of all four Ser/Thr sites in high eukaryotes, further supporting a critical role of these residues in Maf1 function. Interestingly, mTOR inhibition led to an increase in the occupancy of Maf1 on a set of Pol III-dependent genes, with concomitant reduction in the binding of Pol III and Brf1. Unexpectedly, mTORC1 itself was also enriched at the same set of Pol III templates, but this association was not influenced by mTOR inhibitor treatment. Our results highlight a new and unique mode of regulation of Pol III transcription by mTOR and suggest that normalization of Pol III activity may contribute to the therapeutic efficacy of mTOR inhibitors.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , RNA Polimerase III/metabolismo , Proteínas Repressoras/metabolismo , Transcrição Gênica , Sequência de Bases , Western Blotting , Linhagem Celular Tumoral , Cromatografia Líquida , Humanos , Fosforilação , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serina-Treonina Quinases TOR , Espectrometria de Massas em Tandem
4.
Bioorg Med Chem Lett ; 20(1): 375-9, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19897362

RESUMO

2-Aryl-4-morpholinothieno[3,2-d]pyrimidines are known PI3K inhibitors. This class of compounds also potently inhibited the homologous enzyme mTOR. Replacement of the morpholine group in these compounds with an 8-oxa-3-azabicyclo[3.2.1]octane group led to mTOR inhibitors with selectivity over PI3K. Optimization of the 2-aryl substituent led to the discovery of 2-(4-ureidophenyl)-thienopyrimidines as highly potent (IC(50) <1nM) mTOR inhibitors with excellent selectivity (up to >1000-fold) over PI3K and good potency in a cellular proliferation assay (IC(50) <50nM).


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Pirimidinas/química , Tropanos/química , Linhagem Celular Tumoral , Descoberta de Drogas , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Morfolinas/química , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Pirimidinas/síntese química , Pirimidinas/farmacologia , Relação Estrutura-Atividade , Serina-Treonina Quinases TOR
5.
Bioorg Med Chem Lett ; 20(2): 640-3, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19963384
6.
Bioorg Med Chem Lett ; 20(7): 2259-63, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20188551
7.
Bioorg Med Chem Lett ; 20(8): 2644-7, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20227881

RESUMO

Incorporation of bridged morpholines in monocyclic triazine PI3K/mTOR inhibitors gave compounds with increased mTOR selectivity relative to the corresponding morpholine analogs. Compounds with ureidophenyl groups gave highly potent and selective mTOR inhibitors. Potency and selectivity was demonstrated both in vitro and in vivo through biomarker suppression studies. Select compounds exhibited potent inhibition of tumor growth in nude mouse xenograft assays upon PO and IV administration.


Assuntos
Trifosfato de Adenosina/química , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Morfolinas/química , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Triazinas/química , Triazinas/farmacologia , Animais , Descoberta de Drogas , Concentração Inibidora 50 , Camundongos , Camundongos Nus , Serina-Treonina Quinases TOR
8.
Bioorg Med Chem Lett ; 20(4): 1440-4, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20089401

RESUMO

A series of pyrazolopyrimidine mammalian Target Of Rapamycin (mTOR) inhibitors with various substituents at the 1-position have been prepared, resulting in compounds with excellent potency, selectivity and microsomal stability. Combination of a 1-cyclohexyl ketal group with a 2,6-ethylene bridged morpholine in the 4-position and a ureidophenyl group in the 6-positon resulted in compound 8a, that selectively suppressed key mTOR biomarkers in vivo for at least 8h following iv administration and showed excellent oral activity in a xenograft tumor model.


Assuntos
Trifosfato de Adenosina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/química , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/química , Pirazóis/química , Piridinas/química , Trifosfato de Adenosina/química , Administração Oral , Animais , Ligação Competitiva , Estabilidade de Medicamentos , Humanos , Concentração Inibidora 50 , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Microssomos/enzimologia , Estrutura Molecular , Proteínas Serina-Treonina Quinases/metabolismo , Pirazóis/síntese química , Piridinas/síntese química , Relação Estrutura-Atividade , Serina-Treonina Quinases TOR , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Bioorg Med Chem Lett ; 20(7): 2321-5, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20188552

RESUMO

We discovered 2-(4-substituted-pyrrolo[2,3-b]pyridin-3-yl)methylene-4-hydroxybenzofuran-3(2H)-ones as potent and selective ATP-competitive inhibitors of the mammalian target of rapamycin (mTOR). Since phenolic OH groups pose metabolic liability, one of the two hydroxyl groups was selectively removed. The SAR data showed the structural features necessary for subnanomolar inhibitory activity against mTOR kinase as well as selectivity over PI3Kalpha. An X-ray co-crystal structure of one inhibitor with the mTOR-related PI3Kgamma revealed the key hydrogen bonding interactions.


Assuntos
Benzofuranos/química , Benzofuranos/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Cristalografia por Raios X , Humanos , Concentração Inibidora 50 , Camundongos , Camundongos Nus , Microssomos/metabolismo , Modelos Moleculares , Neoplasias/tratamento farmacológico , Fosfatidilinositol 3-Quinases/química , Fosfatidilinositol 3-Quinases/metabolismo , Relação Estrutura-Atividade , Serina-Treonina Quinases TOR
10.
Bioorg Med Chem Lett ; 19(24): 6830-5, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19896845

RESUMO

A series of highly potent and selective pyrazolopyrimidine mTOR inhibitors which contain water-solubilizing groups attached to the 6-arylureidophenyl moiety have been prepared. Such derivatives displayed superior potency to those in which these appendages were attached to alternative sites. In comparison to unfunctionalized arylureido compounds, these analogs demonstrated enhanced cellular potency and significantly improved stability towards human microsomes, resulting in an mTOR inhibitor with impressive efficacy in a xenograft model with an intermittent dosing regimen.


Assuntos
Antineoplásicos/química , Peptídeos e Proteínas de Sinalização Intracelular/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/química , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Pirazóis/química , Pirimidinas/química , Animais , Antineoplásicos/síntese química , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Descoberta de Drogas , Estabilidade de Medicamentos , Humanos , Camundongos , Camundongos Nus , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/farmacocinética , Pirazóis/síntese química , Pirazóis/farmacocinética , Pirimidinas/síntese química , Pirimidinas/farmacocinética , Solubilidade , Serina-Treonina Quinases TOR , Água/química , Ensaios Antitumorais Modelo de Xenoenxerto
11.
J Med Chem ; 51(5): 1319-23, 2008 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-18269228

RESUMO

The phosphoinositide 3-kinase (PI3K) signaling pathway is frequently up-regulated in human cancer and is a promising target for the treatment of cancer. Wortmannin and its analogues are potent inhibitors of PI3K but suffer from inherent defects such as instability, insolubility, and toxicity. Opening of the reactive furan ring of 17-hydroxywortmannin with amines gives compounds with improved properties such as greater stability and aqueous solubility and a larger therapeutic index. Ring-opened analogues such as compound 13 containing basic amine groups have significantly increased PI3K inhibitory potency and greater efficacy in nude mouse xenograft assays.


Assuntos
Androstadienos/síntese química , Antineoplásicos/síntese química , Benzopiranos/síntese química , Indenos/síntese química , Inibidores de Fosfoinositídeo-3 Quinase , Androstadienos/química , Androstadienos/farmacologia , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Benzopiranos/química , Benzopiranos/farmacologia , Linhagem Celular Tumoral , Cristalografia por Raios X , Indenos/química , Indenos/farmacologia , Camundongos , Camundongos Nus , Fosfatidilinositol 3-Quinases/química , Solubilidade , Estereoisomerismo , Relação Estrutura-Atividade , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Mol Cancer Ther ; 6(11): 3028-38, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17989320

RESUMO

The serine/threonine kinase AKT/PKB plays a critical role in cancer and represents a rational target for therapy. Although efforts in targeting AKT pathway have accelerated in recent years, relatively few small molecule inhibitors of AKT have been reported. The development of selective AKT inhibitors is further challenged by the extensive conservation of the ATP-binding sites of the AGC kinase family. In this report, we have conducted a high-throughput screen for inhibitors of activated AKT1. We have identified lactoquinomycin as a potent inhibitor of AKT kinases (AKT1 IC(50), 0.149 +/- 0.045 micromol/L). Biochemical studies implicated a novel irreversible interaction of the inhibitor and AKT involving a critical cysteine residue(s). To examine the role of conserved cysteines in the activation loop (T-loop), we studied mutant AKT1 harboring C296A, C310A, and C296A/C310A. Whereas the ATP-pocket inhibitor, staurosporine, indiscriminately targeted the wild-type and all three mutant-enzymes, the inhibition by lactoquinomycin was drastically diminished in the single mutants C296A and C310A, and completely abolished in the double mutant C296A/C310A. These data strongly implicate the binding of lactoquinomycin to the T-loop cysteines as critical for abrogation of catalysis, and define an unprecedented mechanism of AKT inhibition by a small molecule. Lactoquinomycin inhibited cellular AKT substrate phosphorylation induced by growth factor, loss of PTEN, and myristoylated AKT. The inhibition was substantially attenuated by coexpression of C296A/C310A. Moreover, lactoquinomycin reduced cellular mammalian target of rapamycin signaling and cap-dependent mRNA translation initiation. Our results highlight T-loop targeting as a new strategy for the generation of selective AKT inhibitors.


Assuntos
Cisteína/metabolismo , Inibidores Enzimáticos/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Trifosfato de Adenosina/farmacologia , Regulação Alostérica/efeitos dos fármacos , Animais , Catálise/efeitos dos fármacos , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/química , Humanos , Cinética , Naftoquinonas/química , Naftoquinonas/farmacologia , Fosforilação/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , Proteínas Quinases/metabolismo , Capuzes de RNA/metabolismo , Ratos , Relação Estrutura-Atividade , Especificidade por Substrato/efeitos dos fármacos , Serina-Treonina Quinases TOR , Fatores de Tempo
13.
Oncotarget ; 7(18): 25224-40, 2016 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-27015560

RESUMO

The mechanistic target of rapamycin (mTOR) is a major regulator of cell growth and is frequently dysregulated in cancer. While mTOR complex-1 (mTORC1) is a validated cancer target, the role of mTOR complex-2 (mTORC2) remains less defined. Here, we reveal mTORC2 as a critical regulator of breast cancer metabolism. We showed that hyperphosphorylation in ATP citrate lyase (ACL) occurs frequently in human breast tumors and correlates well with HER2+ and/or PIK3CA-mutant (HER2+/PIK3CAmut) status in breast tumor cell lines. In HER2+/PIK3CAmut cells, mTORC2 controls Ser-455 phosphorylation of ACL thereby promoting acetyl-CoA production, de novo lipogenesis and mitochondrial physiology, all of which were inhibited by an mTORC1/mTORC2 kinase inhibitor (mTOR-KI) or cellular depletion of mTORC2 or ACL. mTOR-KI but not rapamycin blocked the IGF-1-induced ACL phosphorylation and glucose to lipid conversion. Depletion of mTORC2 but not mTORC1 specifically inhibited the ACL-dependent acetyl-CoA production. In the HER2+/PIK3CAmut MDA361, MDA453, BT-474 and T47D cells, depletion of mTORC2 or ACL led to growth inhibition and mitochondrial hyperpolarization, which were partially rescued by an alternate source of acetyl-CoA. These same changes were not apparent in mTORC2- or ACL-depleted HER2-/PIK3CAwt MDA231 and HCC1806 cells, highlighting a differential dependence of mTORC2-ACL for survival in these two cell types. Moreover, ACL Ser-455 mutants S455E (phosphomimetic) and S455A (non-phosphorylatable) each increased or decreased, respectively, the acetyl-CoA production, mitochondrial homeostasis and survival in ACL-depleted MDA453 cells. These studies define a new and rapamycin-resistant mechanism of mTORC2-ACL in lipogenesis and acetyl-CoA biology and provide a rationale for targeting of mTORC1 and mTORC2 in HER2+/PIK3CAmut breast cancer.


Assuntos
ATP Citrato (pro-S)-Liase/metabolismo , Acetilcoenzima A/metabolismo , Neoplasias da Mama/metabolismo , Lipogênese/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Classe I de Fosfatidilinositol 3-Quinases/genética , Feminino , Humanos , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Receptor ErbB-2/genética
14.
Cancer Biol Ther ; 4(5): 538-45, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15846106

RESUMO

Deregulated phosphatidylinositol 3-kinase (PI3K) signaling pathway is widely implicated in tumor growth and resistance to chemotherapy. While a strong rationale exists for pharmacological targeting of PI3K, only a few proof-of-principle in vivo efficacy studies are currently available. PWT-458, pegylated-17-hydroxywortmannin, is a novel and highly potent inhibitor of PI3K in animal models. Upon in vivo cleavage of its poly(ethyleneglycol) (PEG), PWT-458 releases its active moiety 17-hydroxywortmannin (17-HWT), the most potent inhibitor in its class. Here we show that a single intravenous injection of PWT-458 rapidly inhibited PI3K signaling, as measured by a complete loss of AKT (Ser-473) phosphorylation in xenograft tumors grown in nude mice. Following a daily X5 dosing regimen, PWT-458 demonstrated single-agent antitumor activity in nude mouse xenograft models of U87MG glioma, nonsmall cell lung cancer (NSCLC) A549, and renal cell carcinoma (RCC) A498. Efficacious doses ranged from 0.5 mg/kg to 10 mg/kg, achieving a superior therapeutic index over 17-HWT. PWT-458 augmented anticancer efficacy of a suboptimal dose of paclitaxel against A549 and U87MG tumors. Combination treatment of PWT-458 and an mTOR inhibitor, Pegylated-Rapamycin (Peg-Rapa), resulted in an enhanced antitumor efficacy in U87MG. Finally, PWT-458 in combination with interferon-alpha (Intron-A) caused a dramatic regression of RCC A498, which was not achieved by either agent alone. These studies identify PWT-458 as an effective anticancer agent and provide strong proof-of-principle for targeting the PI3K pathway as novel anticancer therapy.


Assuntos
Androstadienos/farmacologia , Neoplasias/tratamento farmacológico , Inibidores de Fosfoinositídeo-3 Quinase , Transdução de Sinais/efeitos dos fármacos , Androstadienos/química , Animais , Antibióticos Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antineoplásicos Fitogênicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Quimioterapia Combinada , Feminino , Glioma/tratamento farmacológico , Glioma/metabolismo , Glioma/patologia , Humanos , Interferon-alfa/uso terapêutico , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Estrutura Molecular , Peso Molecular , Transplante de Neoplasias , Neoplasias/metabolismo , Neoplasias/patologia , Paclitaxel/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sirolimo/farmacologia , Transplante Heterólogo , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Methods Mol Biol ; 821: 15-28, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22125057

RESUMO

The mammalian target of rapamycin (mTOR) is the catalytic subunit of two multiprotein complexes, mTOR complex-1 (mTORC1) and mTOR complex-2 (mTORC2). Clinically used rapamycin and rapalogs are FKBP12-dependent allosteric inhibitors of mTORC1. The recently discovered WYE-125132 and related drugs represent a new generation of ATP competitive and highly specific inhibitors targeting mTOR globally. As mTORC1 and mTORC2 mediate diverse sets of both redundant and distinctive cellular pathways of growth, nutrient and energy homeostasis, rapamycin and WYE-125132 elicit both overlapping and distinctive pharmacological properties with important implications in treating cancer, metabolic, and age-related degenerative diseases. Detailed methods are described for the determination of mTOR inhibition by rapamycin and WYE-125132 in assays of recombinant mTOR enzyme, immunprecipitated native mTOR complexes, growth factor- and amino acid-induced cellular phosphorylation cascades as well as the PI3K/AKT/mTOR hyperactive breast tumor model in vitro and in vivo. The methods have been particularly useful in discovery and biochemical characterization of mTOR inhibitors, cellular and in vivo mTOR substrate phosphorylation analysis, and in deciphering novel biomarkers of mTORC1 and mTORC2 signaling pathways.


Assuntos
Descoberta de Drogas/métodos , Inibidores de Proteínas Quinases/química , Proteínas/antagonistas & inibidores , Sirolimo/farmacologia , Fatores de Transcrição/antagonistas & inibidores , Trifosfato de Adenosina/metabolismo , Biomarcadores Tumorais/antagonistas & inibidores , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos , Compostos de Fenilureia/farmacologia , Inibidores de Proteínas Quinases/isolamento & purificação , Inibidores de Proteínas Quinases/farmacologia , Proteínas/metabolismo , Pirazóis/farmacologia , Transdução de Sinais , Serina-Treonina Quinases TOR , Proteína 1A de Ligação a Tacrolimo/farmacologia , Fatores de Transcrição/metabolismo
16.
Cancer Res ; 70(2): 621-31, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20068177

RESUMO

The mammalian target of rapamycin (mTOR) is a major component of the phosphoinositide 3-kinase (PI3K)/AKT signaling pathway that is dysregulated in 50% of all human malignancies. Rapamycin and its analogues (rapalogs) partially inhibit mTOR through allosteric binding to mTOR complex 1 (mTORC1) but not mTOR complex 2 (mTORC2), an emerging player in cancer. Here, we report WYE-125132 (WYE-132), a highly potent, ATP-competitive, and specific mTOR kinase inhibitor (IC(50): 0.19 +/- 0.07 nmol/L; >5,000-fold selective versus PI3Ks). WYE-132 inhibited mTORC1 and mTORC2 in diverse cancer models in vitro and in vivo. Importantly, consistent with genetic ablation of mTORC2, WYE-132 targeted P-AKT(S473) and AKT function without significantly reducing the steady-state level of the PI3K/PDK1 activity biomarker P-AKT(T308), highlighting a prominent and direct regulation of AKT by mTORC2 in cancer cells. Compared with the rapalog temsirolimus/CCI-779, WYE-132 elicited a substantially stronger inhibition of cancer cell growth and survival, protein synthesis, cell size, bioenergetic metabolism, and adaptation to hypoxia. Oral administration of WYE-132 to tumor-bearing mice showed potent single-agent antitumor activity against MDA361 breast, U87MG glioma, A549 and H1975 lung, as well as A498 and 786-O renal tumors. An optimal dose of WYE-132 achieved a substantial regression of MDA361 and A549 large tumors and caused complete regression of A498 large tumors when coadministered with bevacizumab. Our results further validate mTOR as a critical driver for tumor growth, establish WYE-132 as a potent and profound anticancer agent, and provide a strong rationale for clinical development of specific mTOR kinase inhibitors as new cancer therapy.


Assuntos
Neoplasias/tratamento farmacológico , Compostos de Fenilureia/farmacologia , Pirazóis/farmacologia , Sirolimo/análogos & derivados , Fatores de Transcrição/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Processos de Crescimento Celular/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Complexos Multiproteicos , Neoplasias/metabolismo , Neoplasias/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR
17.
J Med Chem ; 52(24): 7942-5, 2009 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-19916508

RESUMO

Dramatic improvements in mTOR-targeting selectivity were achieved by replacing morpholine in pyrazolopyrimidine inhibitors with bridged morpholines. Analogues with subnanomolar mTOR IC(50) values and up to 26000-fold selectivity versus PI3Kalpha were prepared. Chiral morpholines gave inhibitors whose enantiomers had different selectivity and potency profiles. Molecular modeling suggests that a single amino acid difference between PI3K and mTOR (Phe961Leu) accounts for the profound selectivity seen by creating a deeper pocket in mTOR that can accommodate bridged morpholines.


Assuntos
Morfolinas/farmacologia , Proteínas Quinases/química , Modelos Moleculares , Morfolinas/síntese química , Morfolinas/química , Fosfatidilinositol 3-Quinases/química , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Quinases/metabolismo , Pirazóis/síntese química , Pirazóis/química , Pirazóis/farmacologia , Pirimidinas/síntese química , Pirimidinas/química , Pirimidinas/farmacologia , Relação Estrutura-Atividade , Serina-Treonina Quinases TOR
18.
J Med Chem ; 52(22): 7081-9, 2009 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-19848404

RESUMO

The mammalian target of rapamycin (mTOR) is a central regulator of cell growth, metabolism, and angiogenesis and an emerging target in cancer research. High throughput screening (HTS) of our compound collection led to the identification of 3-(4-morpholin-4-yl-1-piperidin-4-yl-1H-pyrazolo[3,4-d]pyrimidin-6-yl)phenol (5a), a modestly potent and nonselective inhibitor of mTOR and phosphoinositide 3-kinase (PI3K). Optimization of compound 5a, employing an mTOR homology model based on an X-ray crystal structure of closely related PI3Kgamma led to the discovery of 6-(1H-indol-5-yl)-4-morpholin-4-yl-1-[1-(pyridin-3-ylmethyl)piperidin-4-yl]-1H-pyrazolo[3,4-d]pyrimidine (5u), a potent and selective mTOR inhibitor (mTOR IC(50) = 9 nM; PI3Kalpha IC(50) = 1962 nM). Compound 5u selectively inhibited cellular biomarker of mTORC1 (P-S6K, P-4EBP1) and mTORC2 (P-AKT S473) over the biomarker of PI3K/PDK1 (P-AKT T308) and did not inhibit PI3K-related kinases (PIKKs) in cellular assays. These pyrazolopyrimidines represent an exciting new series of mTOR-selective inhibitors with potential for development for cancer therapy.


Assuntos
Descoberta de Drogas , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/metabolismo , Pirimidinas/farmacologia , Ligação Competitiva , Linhagem Celular Tumoral , Humanos , Concentração Inibidora 50 , Modelos Moleculares , Conformação Molecular , Peso Molecular , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/metabolismo , Proteínas Quinases/química , Pirimidinas/síntese química , Pirimidinas/química , Pirimidinas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Especificidade por Substrato , Serina-Treonina Quinases TOR
19.
J Med Chem ; 52(24): 8010-24, 2009 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-19894727

RESUMO

Design and synthesis of a series of 4-morpholino-6-aryl-1H-pyrazolo[3,4-d]pyrimidines as potent and selective inhibitors of the mammalian target of rapamycin (mTOR) are described. Optimization of the 6-aryl substituent led to the discovery of inhibitors carrying 6-ureidophenyl groups, the first reported active site inhibitors of mTOR with subnanomolar inhibitory concentrations. The data presented in this paper show that 6-arylureidophenyl substituents led to potent mixed inhibitors of mTOR and phosphatidylinositol 3-kinase alpha (PI3K-alpha), whereas 6-alkylureidophenyl appendages gave highly selective mTOR inhibitors. Combination of 6-alkylureidophenyl groups with 1-carbamoylpiperidine substitution resulted in compounds with subnanomolar IC(50) against mTOR and greater than 1000-fold selectivity over PI3K-alpha. In addition, structure based drug design resulted in the preparation of several 6-arylureidophenyl-1H-pyrazolo[3,4-d]pyrimidines, substituted in the 4-position of the arylureido moiety with water solubilizing groups. These compounds combined potent mTOR inhibition (IC(50) < 1 nM) with unprecedented activity in cellular proliferation assays (IC(50) < 1 nM).


Assuntos
Trifosfato de Adenosina/química , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/química , Pirimidinas/farmacologia , Trifosfato de Adenosina/metabolismo , Ligação Competitiva , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Masculino , Modelos Moleculares , Morfolinas/síntese química , Morfolinas/química , Morfolinas/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Proteínas Quinases/metabolismo , Pirazóis/síntese química , Pirazóis/química , Pirazóis/farmacologia , Pirimidinas/síntese química , Pirimidinas/química , Relação Estrutura-Atividade , Serina-Treonina Quinases TOR
20.
J Med Chem ; 52(8): 2181-4, 2009 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-19309081

RESUMO

The naturally occurring pyranonaphthoquinone (PNQ) antibiotic lactoquinomycin and related aglycones were found to be selective inhibitors of the serine-threonine kinase AKT. A set of synthetic PNQs were prepared and a minimum active feature set and preliminary SAR were determined. PNQ lactones inhibit the proliferation of human tumor cell lines containing constitutively activated AKT and show expected effects on cellular biomarkers. Biochemical data are presented supporting a proposed bioreductive alkylation mechanism of action.


Assuntos
Antineoplásicos/síntese química , Cisteína/metabolismo , Lactonas/síntese química , Proteína Oncogênica v-akt/antagonistas & inibidores , Piranos/síntese química , Alquilação , Antineoplásicos/química , Antineoplásicos/farmacologia , Biomarcadores/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Lactonas/química , Lactonas/farmacologia , Naftoquinonas/síntese química , Naftoquinonas/química , Naftoquinonas/farmacologia , Piranos/química , Piranos/farmacologia , Estereoisomerismo , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA