Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Bioorg Med Chem Lett ; 30(12): 127202, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32331933

RESUMO

The therapeutic potential of interfering with dysregulated proteins by inducing its selective degradation has been pursued using different mechanisms. In the present article, we review representative examples of monovalent protein-degraders that, contrary to the proteolysis targeting chimeras, achieve target degradation without displaying recognition motifs for the recruitment of E3 ubiquitin ligases. We also highlight new technologies and assays that may brought to bear on the discovery of common elements that could predict and enable the selective degradation of pathogenic targets by monovalent protein-degraders. The successful application of these methods would pave the way to the advancement of new drugs with unique efficacy and tolerability properties.


Assuntos
Proteólise/efeitos dos fármacos , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo , Técnicas Biossensoriais , Cinamatos/farmacologia , Descoberta de Drogas , Fulvestranto/farmacologia , Humanos , Indóis/farmacologia , Ligantes , Ligação Proteica , Receptores de Estrogênio/metabolismo , Estilbenos/farmacologia , Tamoxifeno/farmacologia
2.
Nucleic Acids Res ; 43(2): 1204-15, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25550434

RESUMO

Mechanisms of unassisted delivery of RNA therapeutics, including inhibitors of microRNAs, remain poorly understood. We observed that the hepatocellular carcinoma cell line SKHEP1 retains productive free uptake of a miR-21 inhibitor (anti-miR-21). Uptake of anti-miR-21, but not a mismatch (MM) control, induces expression of known miR-21 targets (DDAH1, ANKRD46) and leads to dose-dependent inhibition of cell growth. To elucidate mechanisms of SKHEP1 sensitivity to anti-miR-21, we conducted an unbiased shRNA screen that revealed tumor susceptibility gene 101 (TSG101), a component of the endosomal sorting complex required for transport (ESCRT-I), as an important determinant of anti-proliferative effects of anti-miR-21. RNA interference-mediated knockdown of TSG101 and another ESCRT-I protein, VPS28, improved uptake of anti-miR-21 in parental SKHEP1 cells and restored productive uptake to SKHEP1 clones with acquired resistance to anti-miR-21. Depletion of ESCRT-I in several additional cancer cell lines with inherently poor uptake resulted in improved activity of anti-miR-21. Finally, knockdown of TSG101 increased uptake of anti-miR-21 by cancer cells in vivo following systemic delivery. Collectively, these data support an important role for the ESCRT-I complex in the regulation of productive free uptake of anti-miRs and reveal potential avenues for improving oligonucleotide free uptake by cancer cells.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , MicroRNAs/antagonistas & inibidores , Neoplasias/metabolismo , Oligonucleotídeos/metabolismo , Animais , Transporte Biológico , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/fisiologia , Complexos Endossomais de Distribuição Requeridos para Transporte/antagonistas & inibidores , Complexos Endossomais de Distribuição Requeridos para Transporte/fisiologia , Feminino , Humanos , Camundongos SCID , MicroRNAs/metabolismo , Neoplasias/genética , Fatores de Transcrição/fisiologia
3.
Blood ; 124(11): 1777-89, 2014 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-25006129

RESUMO

Postchemotherapy relapse presents a major unmet medical need in acute myeloid leukemia (AML), where treatment options are limited. CD25 is a leukemic stem cell marker and a conspicuous prognostic marker for overall/relapse-free survival in AML. Rare occurrence of genetic alterations among PIM family members imposes a substantial hurdle in formulating a compelling patient stratification strategy for the clinical development of selective PIM inhibitors in cancer. Here we show that CD25, a bona fide STAT5 regulated gene, is a mechanistically relevant predictive biomarker for sensitivity to PIM kinase inhibitors. Alone or in combination with tyrosine kinase inhibitors, PIM inhibitors can suppress STAT5 activation and significantly shorten the half-life of MYC to achieve substantial growth inhibition of high CD25-expressing AML cells. Our results highlight the importance of STAT5 and MYC in rendering cancer cells sensitive to PIM inhibitors. Because the presence of a CD25-positive subpopulation in leukemic blasts correlates with poor overall or relapse-free survival, our data suggest that a combination of PIM inhibitors with chemotherapy and tyrosine kinase inhibitors could improve long-term therapeutic outcomes in CD25-positive AML.


Assuntos
Antineoplásicos/farmacologia , Crise Blástica , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Proteólise/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Proto-Oncogênicas c-pim-1/antagonistas & inibidores , Fator de Transcrição STAT5/metabolismo , Antineoplásicos/química , Crise Blástica/tratamento farmacológico , Crise Blástica/genética , Crise Blástica/metabolismo , Crise Blástica/patologia , Feminino , Células HL-60 , Humanos , Subunidade alfa de Receptor de Interleucina-2/genética , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Masculino , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-pim-1/genética , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , Fator de Transcrição STAT5/genética
4.
Nat Chem Biol ; 10(12): 1013-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25326666

RESUMO

Vps34 is a phosphoinositide 3-kinase (PI3K) class III isoform that has attracted major attention over the recent years because of its role in autophagy. Herein we describe the biological characterization of SAR405, which is a low-molecular-mass kinase inhibitor of Vps34 (KD 1.5 nM). This compound has an exquisite protein and lipid kinase selectivity profile that is explained by its unique binding mode and molecular interactions within the ATP binding cleft of human Vps34. To the best of our knowledge, this is the first potent and specific Vps34 inhibitor described so far. Our results demonstrate that inhibition of Vps34 kinase activity by SAR405 affects both late endosome-lysosome compartments and prevents autophagy. Moreover, we show that the concomitant inhibition of Vps34 and mTOR, with SAR405 and the US Food and Drug Administration-approved mTOR inhibitor everolimus, results in synergistic antiproliferative activity in renal tumor cell lines, indicating a potential clinical application in cancer.


Assuntos
Antineoplásicos/farmacologia , Autofagia/efeitos dos fármacos , Classe III de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Pirimidinonas/farmacologia , Sirolimo/análogos & derivados , Serina-Treonina Quinases TOR/antagonistas & inibidores , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Antineoplásicos/síntese química , Autofagia/genética , Domínio Catalítico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Classe III de Fosfatidilinositol 3-Quinases/química , Classe III de Fosfatidilinositol 3-Quinases/genética , Sinergismo Farmacológico , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Everolimo , Expressão Gênica , Humanos , Rim/enzimologia , Rim/patologia , Cinética , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Simulação de Acoplamento Molecular , Inibidores de Proteínas Quinases/síntese química , Piridinas/síntese química , Pirimidinonas/síntese química , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Transdução de Sinais , Sirolimo/síntese química , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/química , Serina-Treonina Quinases TOR/genética
5.
J Biol Chem ; 288(42): 30125-30138, 2013 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-24003220

RESUMO

PRP4 kinase is known for its roles in regulating pre-mRNA splicing and beyond. Therefore, a wider spectrum of PRP4 kinase substrates could be expected. The role of PRP4 kinase in cancer is also yet to be fully elucidated. Attaining specific and potent PRP4 inhibitors would greatly facilitate the study of PRP4 biological function and its validation as a credible cancer target. In this report, we verified the requirement of enzymatic activity of PRP4 in regulating cancer cell growth and identified an array of potential novel substrates through orthogonal proteomics approaches. The ensuing effort in structural biology unveiled for the first time unique features of PRP4 kinase domain and its potential mode of interaction with a low molecular weight inhibitor. These results provide new and important information for further exploration of PRP4 kinase function in cancer.


Assuntos
Proteínas de Neoplasias , Neoplasias , Inibidores de Proteínas Quinases , Ribonucleoproteína Nuclear Pequena U4-U6 , Linhagem Celular Tumoral , Humanos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Neoplasias/genética , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Proteômica/métodos , Ribonucleoproteína Nuclear Pequena U4-U6/antagonistas & inibidores , Ribonucleoproteína Nuclear Pequena U4-U6/química , Ribonucleoproteína Nuclear Pequena U4-U6/genética , Ribonucleoproteína Nuclear Pequena U4-U6/metabolismo
6.
Bioorg Med Chem Lett ; 24(23): 5357-63, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25455482

RESUMO

Antibody-drug conjugates (ADCs) consist of cytotoxic drugs covalently linked to monoclonal antibodies directed to antigens differentially overexpressed in tumor cells. These loaded antibodies are expected to selectively deliver lethal cargoes to tumor cells and provide sustained clinical benefit to pre-selected cancer patients while, at the same time, minimizing systemic toxicity. Although on-target adverse events are not completely avoided and the true efficacy of these innovative agents still requires further clarification, proof-of-concept has already been achieved in clinical settings with immunoconjugates containing calicheamicin, auristatin or maytansine-based cytotoxic payloads. In this present article we review the characteristics of the preceding cytotoxic platforms and their chemical conjugation approaches.


Assuntos
Anticorpos Monoclonais/metabolismo , Neoplasias/tratamento farmacológico , Produtos Biológicos , Humanos , Estrutura Molecular
7.
Bioorg Med Chem Lett ; 24(6): 1506-10, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24560540
8.
Bioorg Med Chem Lett ; 23(9): 2480-5, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23541651

RESUMO

A growing number of the elements identified in intracellular signaling events that affect cell growth and transformation are proteins that physically interact with each other via domains or specifically recognized amino acid sequences. Some of these intracellular protein-protein interactions are attractive targets for anticancer targeted therapy, but progress in this field has been compromised by the paucity of compounds with suitable biological profiles and pharmacological properties. This Letter covers salient achievements in the identification and development of inhibitors of the p53-hdm2 protein-protein interaction, and highlights different screening techniques and structure-based design approaches that may be brought to bear on the discovery and development of inhibitors of other therapeutically relevant intracellular protein-protein interactions.


Assuntos
Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Proteína Supressora de Tumor p53/antagonistas & inibidores , Sítios de Ligação , Desenho de Fármacos , Células HCT116 , Humanos , Imidazolinas/síntese química , Imidazolinas/química , Imidazolinas/farmacologia , Simulação de Acoplamento Molecular , Piperidinas/química , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Relação Estrutura-Atividade , Proteína Supressora de Tumor p53/metabolismo
9.
Cancer Cell ; 5(3): 221-30, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15050914

RESUMO

Insulin-like growth factors and their receptor (IGF-1R) have been implicated in cancer pathophysiology. We demonstrate that IGF-1R is universally expressed in various hematologic (multiple myeloma, lymphoma, leukemia) and solid tumor (breast, prostate, lung, colon, thyroid, renal, adrenal cancer, retinoblastoma, and sarcoma) cells. Specific IGF-1R inhibition with neutralizing antibody, antagonistic peptide, or the selective kinase inhibitor NVP-ADW742 has in vitro activity against diverse tumor cell types (particularly multiple myeloma), even those resistant to conventional therapies, and triggers pleiotropic antiproliferative/proapoptotic molecular sequelae, delineated by global transcriptional and proteomic profiling. NVP-ADW742 monotherapy or its combination with cytotoxic chemotherapy had significant antitumor activity in an orthotopic xenograft MM model, providing in vivo proof of principle for therapeutic use of selective IGF-1R inhibitors in cancer.


Assuntos
Neoplasias Hematológicas/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Receptor IGF Tipo 1/antagonistas & inibidores , Receptor IGF Tipo 1/metabolismo , Antineoplásicos , Medula Óssea/metabolismo , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo , Perfilação da Expressão Gênica , Neoplasias Hematológicas/tratamento farmacológico , Humanos , Mieloma Múltiplo , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Pirimidinas/farmacologia , Pirróis/farmacologia , Transplante Heterólogo/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
Cancer Cell ; 5(3): 231-9, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15050915

RESUMO

IGF-IR-mediated signaling promotes survival, anchorage-independent growth, and oncogenic transformation, as well as tumor growth and metastasis formation in vivo. NVP-AEW541 is a pyrrolo[2,3-d]pyrimidine derivative small molecular weight kinase inhibitor of the IGF-IR, capable of distinguishing between the IGF-IR (IC50 = 0.086 microM) and the closely related InsR (IC50 = 2.3 microM) in cells. As expected for a specific IGF-IR kinase inhibitor, NVP-AEW541 abrogates IGF-I-mediated survival and colony formation in soft agar at concentrations that are consistent with inhibition of IGF-IR autophosphorylation. In vivo, this orally bioavailable compound inhibits IGF-IR signaling in tumor xenografts and significantly reduces the growth of IGF-IR-driven fibrosarcomas. Thus, NVP-AEW541 represents a class of selective, small molecule IGF-IR kinase inhibitors with proven in vivo antitumor activity and potential therapeutic application.


Assuntos
Antineoplásicos/farmacologia , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais/fisiologia , Células 3T3 , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Divisão Celular , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Fosforilação/efeitos dos fármacos , Receptor IGF Tipo 1/efeitos dos fármacos , Receptor de Insulina/efeitos dos fármacos , Receptor de Insulina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Quinases da Família src/efeitos dos fármacos , Quinases da Família src/metabolismo
11.
Proc Natl Acad Sci U S A ; 106(52): 22299-304, 2009 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-20007781

RESUMO

NVP-BEZ235 is a dual PI3K/mTOR inhibitor currently in phase I clinical trials. We profiled this compound against a panel of breast tumor cell lines to identify the patient populations that would benefit from such treatment. In this setting, NVP-BEZ235 selectively induced cell death in cell lines presenting either HER2 amplification and/or PIK3CA mutation, but not in cell lines with PTEN loss of function or KRAS mutations, for which resistance could be attributed, in part to ERK pathway activity. An in depth analysis of death markers revealed that the cell death observed upon NVP-BEZ235 treatment could be recapitulated with other PI3K inhibitors and that this event is linked to active PARP cleavage indicative of an apoptotic process. Moreover, the effect seemed to be partly independent of the caspase-9 executioner and mitochondrial activated caspases, suggesting an alternate route for apoptosis induction by PI3K inhibitors. Overall, this study will provide guidance for patient stratification for forthcoming breast cancer phase II trials for NVP-BEZ235.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Genes erbB-2 , Imidazóis/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/efeitos dos fármacos , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Quinolinas/farmacologia , Apoptose/genética , Apoptose/fisiologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Caspase 9/metabolismo , Linhagem Celular Tumoral , Classe I de Fosfatidilinositol 3-Quinases , Resistencia a Medicamentos Antineoplásicos/genética , Resistencia a Medicamentos Antineoplásicos/fisiologia , Inibidores Enzimáticos/farmacologia , Feminino , Amplificação de Genes , Genes erbB-2/efeitos dos fármacos , Humanos , Mutação , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR
12.
Proc Natl Acad Sci U S A ; 106(1): 268-73, 2009 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-19116269

RESUMO

Characterization of the molecular pathways that are required for the viability and maintenance of self-renewing tumor-initiating cells may ultimately lead to improved therapies for cancer. In this study, we show that a CD133(+)/CD44(+) population of cells enriched in prostate cancer progenitors (PCaPs) has tumor-initiating potential and that these progenitors can be expanded under nonadherent, serum-free, sphere-forming conditions. Cells grown under these conditions have increased in vitro clonogenic and in vivo tumorigenic potential. mRNA expression analysis of cells grown under sphere-forming conditions, compared with long-term monolayer cultures, revealed preferential activation of the PI3K/AKT signaling pathway. PI3K p110alpha and beta-protein levels were higher in cells grown under sphere-forming conditions, and phosphatase and tensin homolog (PTEN) knockdown by shRNA led to an increase in sphere formation as well as increased clonogenic and tumorigenic potential. Similarly, shRNA knockdown of FoxO3a led to an increase in tumorigenic potential. Consistent with these results, inhibition of PI3K activity by the dual PI3K/mTOR inhibitor NVP-BEZ235 led to growth inhibition of PCaPs. Taken together, our data strongly suggest that the PTEN/PI3K/Akt pathways are critical for prostate cancer stem-like cell maintenance and that targeting PI3K signaling may be beneficial in prostate cancer treatment by eliminating prostate cancer stem-like cells.


Assuntos
Células-Tronco Neoplásicas/patologia , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Antígeno AC133 , Antígenos CD , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Glicoproteínas , Humanos , Receptores de Hialuronatos , Masculino , Peptídeos , RNA Mensageiro/análise , RNA Neoplásico/análise , Transdução de Sinais/fisiologia
13.
Proc Natl Acad Sci U S A ; 106(46): 19503-8, 2009 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-19850869

RESUMO

Non-small cell lung cancers with activating mutations in the epidermal growth factor receptor (EGFR) are highly responsive to EGFR tyrosine kinase inhibitors (TKIs), such as gefitinib and erlotinib. Such cancers are "addicted" to EGFR, and treatment with a TKI invariably leads to down-regulation of the PI3K-AKT-mTOR and MEK-ERK signaling pathways, resulting in apoptosis. Using a dual PI3K-mTOR inhibitor, NVP-BEZ235, we evaluated whether PI3K-mTOR inhibition alone induced apoptosis in these cancers. In contrast to HER2-amplified breast cancers, we found that PI3K-mTOR inhibition did not promote substantial apoptosis in the EGFR mutant lung cancers. However, blocking both PI3K-mTOR and MEK simultaneously led to apoptosis to similar levels as the EGFR TKIs, suggesting that down-regulation of these pathways may account for much of the apoptosis promoted by EGFR inhibition. In EGFR mutant lung cancers, down-regulation of both intracellular pathways converged on the BH3 family of proteins regulating apoptosis. PI3K inhibition led to down-regulation of Mcl-1, and MEK inhibition led to up-regulation of BIM. In fact, down-regulation of Mcl-1 by siRNA was sufficient to sensitize these cancers to single-agent MEK inhibitors. Surprisingly, an AKT inhibitor did not decrease Mcl-1 levels, and when combined with MEK inhibitors, failed to induce apoptosis. Importantly, we observed that the combination of PI3K-mTOR and MEK inhibitors effectively shrunk tumors in a transgenic and xenograft model of EGFR T790M-L858R cancers. These data indicate simultaneous inhibition of PI3K-mTOR and MEK signaling is an effective strategy for treating EGFR mutant lung cancers, including those with acquired resistance to EGFR TKIs.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica , Apoptose , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/uso terapêutico , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Linhagem Celular Tumoral , Receptores ErbB/genética , Cloridrato de Erlotinib , Feminino , Gefitinibe , Humanos , Imidazóis/farmacologia , Neoplasias Pulmonares/genética , MAP Quinase Quinase Quinases/antagonistas & inibidores , Camundongos , Proteínas Quinases/metabolismo , Quinazolinas/farmacologia , Quinolinas/farmacologia , Receptor ErbB-2/genética , Serina-Treonina Quinases TOR , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Biochem Soc Trans ; 39(2): 451-5, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21428918

RESUMO

Substantial drug discovery efforts have been devoted, over the last few years, to identifying and developing mTOR (mammalian target of rapamycin) kinase modulators. This has resulted in a number of mTOR inhibitors with different mechanisms of action and/or distinct protein and lipid kinase selectivity profiles. As briefly reviewed in the present paper, these compounds have provided us with a better understanding of the roles of mTOR and other phosphoinositide 3-kinase/mTOR pathway components in human cancer biology, and a few of them have already demonstrated clinical benefit in cancer patients.


Assuntos
Descoberta de Drogas/métodos , Terapia de Alvo Molecular/métodos , Inibidores de Proteínas Quinases/isolamento & purificação , Serina-Treonina Quinases TOR/antagonistas & inibidores , Regulação Alostérica/fisiologia , Animais , Ligação Competitiva , Humanos , Inibidores de Proteínas Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Sirolimo/metabolismo , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo
15.
Curr Top Microbiol Immunol ; 347: 209-39, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20582534

RESUMO

A number of intracellular kinase components of the PI3K/Akt/mTOR pathway have been targeted over the past few years, leading to a new generation of anticancer agents that effectively and specifically disrupt this pathway in tumor cells. Here, progress in the identification and clinical evaluation of compounds designed to modulate the enzymatic activity of PI3K, Akt, mTOR, and Hsp90 is reviewed.


Assuntos
Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Animais , Artrite/tratamento farmacológico , Descoberta de Drogas , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Humanos , Isoenzimas/antagonistas & inibidores , Pneumopatias/tratamento farmacológico , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Neoplasias/tratamento farmacológico , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Serina-Treonina Quinases TOR/antagonistas & inibidores
16.
Mol Cancer Ther ; 20(2): 250-262, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33310762

RESUMO

Primary treatment for estrogen receptor-positive (ER+) breast cancer is endocrine therapy. However, substantial evidence indicates a continued role for ER signaling in tumor progression. Selective estrogen receptor degraders (SERD), such as fulvestrant, induce effective ER signaling inhibition, although clinical studies with fulvestrant report insufficient blockade of ER signaling, possibly due to suboptimal pharmaceutical properties. Furthermore, activating mutations in the ER have emerged as a resistance mechanism to current endocrine therapies. New oral SERDs with improved drug properties are under clinical investigation, but the biological profile that could translate to improved therapeutic benefit remains unclear. Here, we describe the discovery of SAR439859, a novel, orally bioavailable SERD with potent antagonist and degradation activities against both wild-type and mutant Y537S ER. Driven by its fluoropropyl pyrrolidinyl side chain, SAR439859 has demonstrated broader and superior ER antagonist and degrader activities across a large panel of ER+ cells, compared with other SERDs characterized by a cinnamic acid side chain, including improved inhibition of ER signaling and tumor cell growth. Similarly, in vivo treatment with SAR439859 demonstrated significant tumor regression in ER+ breast cancer models, including MCF7-ESR1 wild-type and mutant-Y537S mouse tumors, and HCI013, a patient-derived tamoxifen-resistant xenograft tumor. These findings indicate that SAR439859 may provide therapeutic benefit to patients with ER+ breast cancer, including those who have resistance to endocrine therapy with both wild-type and mutant ER.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Receptores de Estrogênio/uso terapêutico , Animais , Modelos Animais de Doenças , Feminino , Humanos , Camundongos
17.
Mol Cancer Res ; 7(4): 601-13, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19372588

RESUMO

Phosphoinositide 3-kinase (PI3K)/protein kinase B/Akt and Ras/mitogen-activated protein kinase pathways are often constitutively activated in melanoma and have thus been considered as promising drug targets. Exposure of melanoma cells to NVP-BAG956, NVP-BBD130, and NVP-BEZ235, a series of novel, potent, and stable dual PI3K/mammalian target of rapamycin (mTOR) inhibitors, resulted in complete G1 growth arrest, reduction of cyclin D1, and increased levels of p27(KIP1), but negligible apoptosis. In contrast, treatment of melanoma with the pan-class I PI3K inhibitor ZSTK474 or the mTORC1 inhibitor rapamycin resulted only in minor reduction of cell proliferation. In a syngeneic B16 mouse melanoma tumor model, orally administered NVP-BBD130 and NVP-BEZ235 efficiently attenuated tumor growth at primary and lymph node metastatic sites with no obvious toxicity. Metastatic melanoma in inhibitor-treated mice displayed reduced numbers of proliferating and significantly smaller tumor cells. In addition, neovascularization was blocked and tumoral necrosis increased when compared with vehicle-treated mice. In conclusion, compounds targeting PI3K and mTOR simultaneously were advantageous to attenuate melanoma growth and they develop their potential by targeting tumor growth directly, and indirectly via their interference with angiogenesis. Based on the above results, NVP-BEZ235, which has entered phase I/II clinical trials in patients with advanced solid tumors, has a potential in metastatic melanoma therapy.


Assuntos
Imidazóis/farmacologia , Melanoma Experimental/tratamento farmacológico , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Quinases/metabolismo , Quinolinas/farmacologia , Sirolimo/farmacologia , Triazinas/farmacologia , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Administração Oral , Animais , Antibióticos Antineoplásicos/farmacologia , Apoptose/fisiologia , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Feminino , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/metabolismo , Humanos , Immunoblotting , Técnicas Imunoenzimáticas , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Proteico , Serina-Treonina Quinases TOR , Células Tumorais Cultivadas
18.
Eur J Haematol ; 84(4): 337-44, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20028416

RESUMO

Heat shock protein 90 (HSP90) is a promising target for tumor therapy. The novel HSP90 inhibitor NVP-AUY922 has preclinical activity in multiple myeloma, however, little is known about effective combination partners to design clinical studies. Multiple myeloma cell lines, OPM-2, RPMI-8226, U-266, LP-1, MM1.S, and primary myeloma cells were exposed to NVP-AUY922 and one of the combination partners histone deacetylase inhibitor NVP-LBH589, suberoylanilide hydroxamic acid (SAHA), melphalan, or doxorubicin, either simultaneously or in sequential patterns. Effects on cell proliferation and apoptosis were determined. Synergistic effects were evaluated using the method of Chou and Talalay. Combined sequential incubation with NVP-AUY922 and SAHA showed that best synergistic effects were achieved with 24 h preincubation with SAHA followed by another 48 h of combination treatment. Combination of NVP-AUY922 with SAHA, NVP-LBH589, melphalan, or doxorubicin resulted in synergistic inhibition of viability, with strong synergy (combination index < 0.3) in the case of melphalan. Importantly, resistance of the RPMI-8226 cell line and relative resistance of some primary myeloma cells against NVP-AUY922 could be overcome by combination treatment. These data show impressive synergistic action of the novel HSP90 inhibitor NVP-AUY922 with melphalan, doxorubicin, NVP-LBH589, and SAHA in multiple myeloma and build the frame work for clinical trials.


Assuntos
Doxorrubicina/farmacologia , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Inibidores de Histona Desacetilases/farmacologia , Isoxazóis/farmacologia , Melfalan/farmacologia , Mieloma Múltiplo/tratamento farmacológico , Resorcinóis/farmacologia , Antibióticos Antineoplásicos , Antineoplásicos Alquilantes , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/agonistas , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Proteínas de Choque Térmico HSP90/metabolismo , Inibidores de Histona Desacetilases/agonistas , Histona Desacetilases/metabolismo , Humanos , Isoxazóis/agonistas , Isoxazóis/uso terapêutico , Melfalan/agonistas , Mieloma Múltiplo/metabolismo , Resorcinóis/agonistas , Resorcinóis/uso terapêutico
19.
Bioorg Med Chem Lett ; 20(15): 4308-12, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20561789

RESUMO

Over the past few years a number of components of the PI3K/mTOR pathway have been the subject of intense drug discovery activities both in pharmaceutical companies and in academia. This review article summarizes progress made in the identification and development of allosteric and ATP-competitive kinase inhibitors of mTOR and their potential therapeutic use in oncology.


Assuntos
Antineoplásicos/química , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/química , Serina-Treonina Quinases TOR/antagonistas & inibidores , Trifosfato de Adenosina/metabolismo , Regulação Alostérica , Antineoplásicos/uso terapêutico , Humanos , Fosfatidilinositol 3-Quinase/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/uso terapêutico , Pirimidinas/química , Pirimidinas/uso terapêutico , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
20.
Clin Cancer Res ; 26(24): 6589-6599, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33046521

RESUMO

PURPOSE: Carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) is a glycoprotein that has limited expression in normal adult tissues, but is overexpressed in carcinomas of the gastrointestinal tract, the genitourinary and respiratory systems, and breast cancer. As such, CEACAM5 is an attractive target for antibody-based therapies designed to selectively deliver cytotoxic drugs to certain epithelial tumors. Here, we describe preclinical data for a novel antibody-drug conjugate (ADC), SAR408701, which consists of an anti-CEACAM5 antibody (SAR408377) coupled to a maytansinoid agent DM4 via a cleavable linker. EXPERIMENTAL DESIGN: The specificity and binding affinity of SAR408701 to human and cynomolgus monkey CEACAM5 were tested in vitro. The cytotoxic activity of SAR408701 was assessed in CEACAM5-expressing tumor cell lines and using patient-derived xenograft mouse models of CEACAM5-positive tumors. Pharmacokinetic-pharmacodynamic and pharmacokinetic-efficacy relationships were established. SAR408701 toxicity was evaluated in cynomolgus monkey. RESULTS: SAR408701 bound selectively to human and cynomolgus monkey CEACAM5 with similar apparent Kd values (0.017 nmol/L and 0.024 nmol/L, respectively). Both in vitro and in vivo evaluations showed that SAR408701 has cytotoxic activity, leading to in vivo efficacy in single and repeated dosing. Single doses of SAR408701 induced significant increases in the tumor expression of phosphorylated histone H3, confirming the tubulin-targeting mechanism of action. The overall toxicity profile of SAR408701 in cynomolgus monkey was similar to that observed after intravenous administration of DM4 alone. CONCLUSIONS: On the basis of these preclinical data, the ADC SAR408701 is a promising candidate for development as a potential treatment for patients with CEACAM5-positive tumors.


Assuntos
Anticorpos Monoclonais/química , Anticorpos/farmacologia , Antineoplásicos/farmacologia , Imunoconjugados/farmacologia , Maitansina/química , Neoplasias Epiteliais e Glandulares/tratamento farmacológico , Animais , Anticorpos/química , Anticorpos/uso terapêutico , Anticorpos Monoclonais/imunologia , Antineoplásicos/química , Apoptose , Antígeno Carcinoembrionário/imunologia , Proliferação de Células , Feminino , Proteínas Ligadas por GPI/antagonistas & inibidores , Proteínas Ligadas por GPI/imunologia , Humanos , Macaca fascicularis , Camundongos , Camundongos SCID , Neoplasias Epiteliais e Glandulares/imunologia , Neoplasias Epiteliais e Glandulares/patologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA