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








Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 131(22)2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34779417

RESUMO

Alpelisib selectively inhibits the p110α catalytic subunit of PI3Kα and is approved for treatment of breast cancers harboring canonical PIK3CA mutations. In head and neck squamous cell carcinoma (HNSCC), 63% of PIK3CA mutations occur at canonical hotspots. The oncogenic role of the remaining 37% of PIK3CA noncanonical mutations is incompletely understood. We report a patient with HNSCC with a noncanonical PIK3CA mutation (Q75E) who exhibited a durable (12 months) response to alpelisib in a phase II clinical trial. Characterization of all 32 noncanonical PIK3CA mutations found in HNSCC using several functional and phenotypic assays revealed that the majority (69%) were activating, including Q75E. The oncogenic impact of these mutations was validated in 4 cellular models, demonstrating that their activity was lineage independent. Further, alpelisib exhibited antitumor effects in a xenograft derived from a patient with HNSCC containing an activating noncanonical PIK3CA mutation. Structural analyses revealed plausible mechanisms for the functional phenotypes of the majority of the noncanonical PIK3CA mutations. Collectively, these findings highlight the importance of characterizing the function of noncanonical PIK3CA mutations and suggest that patients with HNSCC whose tumors harbor activating noncanonical PIK3CA mutations may benefit from treatment with PI3Kα inhibitors.


Assuntos
Classe I de Fosfatidilinositol 3-Quinases/genética , Neoplasias de Cabeça e Pescoço/genética , Mutação , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Tiazóis/uso terapêutico , Animais , Classe I de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Classe I de Fosfatidilinositol 3-Quinases/química , Classe I de Fosfatidilinositol 3-Quinases/fisiologia , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Domínios Proteicos , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico
2.
Front Immunol ; 12: 708908, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34421914

RESUMO

PI3K signalling is required for activation, differentiation, and trafficking of T cells. PI3Kδ, the dominant PI3K isoform in T cells, has been extensively characterised using PI3Kδ mutant mouse models and PI3K inhibitors. Furthermore, characterisation of patients with Activated PI3K Delta Syndrome (APDS) and mouse models with hyperactive PI3Kδ have shed light on how increased PI3Kδ activity affects T cell functions. An important function of PI3Kδ is that it acts downstream of TCR stimulation to activate the major T cell integrin, LFA-1, which controls transendothelial migration of T cells as well as their interaction with antigen-presenting cells. PI3Kδ also suppresses the cell surface expression of CD62L and CCR7 which controls the migration of T cells across high endothelial venules in the lymph nodes and S1PR1 which controls lymph node egress. Therefore, PI3Kδ can control both entry and exit of T cells from lymph nodes as well as the recruitment to and retention of T cells within inflamed tissues. This review will focus on the regulation of adhesion receptors by PI3Kδ and how this contributes to T cell trafficking and localisation. These findings are relevant for our understanding of how PI3Kδ inhibitors may affect T cell redistribution and function.


Assuntos
Classe I de Fosfatidilinositol 3-Quinases/fisiologia , Linfócitos T/fisiologia , Animais , Adesão Celular , Movimento Celular , Classe I de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Humanos , Sinapses Imunológicas/fisiologia , Integrinas/fisiologia , Antígeno-1 Associado à Função Linfocitária/fisiologia , Camundongos , Doenças da Imunodeficiência Primária/etiologia , Transdução de Sinais/fisiologia , Quinases Associadas a rho/fisiologia
3.
J Pathol ; 250(2): 231-242, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31674011

RESUMO

We genetically engineered expression of an activated form of P110 alpha, the catalytic subunit of PI3K, in mouse prostate epithelium to create a mouse model of direct PI3K activation (Pbsn-cre4Prb;PI3KGOF/+ ). We hypothesized that direct activation would cause rapid neoplasia and cancer progression. Pbsn-cre4Prb;PI3KGOF/+ mice developed widespread prostate intraepithelial hyperplasia, but stromal invasion was limited and overall progression was slower than anticipated. However, the model produced profound and progressive stromal remodeling prior to explicit epithelial neoplasia. Increased stromal cellularity and inflammatory infiltrate were evident as early as 4 months of age and progressively increased through 12 months of age, the terminal endpoint of this study. Prostatic collagen density and phosphorylated SMAD2-positive prostatic stromal cells were expansive and accumulated with age, consistent with pro-fibrotic TGF-ß pathway activation. Few reported mouse models accumulate prostate-specific collagen to the degree observed in Pbsn-cre4Prb;PI3KGOF/+ . Our results indicate a signaling process beginning with prostatic epithelial PI3K and TGF-ß signaling that drives prostatic stromal hypertrophy and collagen accumulation. These mice afford a unique opportunity to explore molecular mechanisms of prostatic collagen accumulation that is relevant to cancer progression, metastasis, inflammation and urinary dysfunction. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Classe I de Fosfatidilinositol 3-Quinases/fisiologia , Colágeno/metabolismo , Próstata/enzimologia , Neoplasia Prostática Intraepitelial/enzimologia , Neoplasias da Próstata/enzimologia , Envelhecimento/patologia , Animais , Modelos Animais de Doenças , Progressão da Doença , Epitélio/enzimologia , Masculino , Camundongos Mutantes , Fosforilação , Próstata/metabolismo , Próstata/patologia , Hiperplasia Prostática/enzimologia , Hiperplasia Prostática/metabolismo , Hiperplasia Prostática/patologia , Neoplasia Prostática Intraepitelial/metabolismo , Neoplasia Prostática Intraepitelial/patologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Transdução de Sinais , Proteína Smad2/metabolismo , Células Estromais/metabolismo , Células Estromais/patologia , Fator de Crescimento Transformador beta/fisiologia
4.
Cells ; 8(10)2019 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-31546615

RESUMO

The phosphoinositide 3-kinase catalytic subunit p110δ (PI3Kδ) gene maps to a human inflammatory bowel diseases (IBD) susceptibility locus, and genetic deletion of PI3Kδ signaling causes spontaneous colitis in mice. However, little is known regarding the role of PI3Kδ on IL-10-producing B cells that help regulate mucosal inflammation in IBD. We investigated the role of PI3Kδ signaling in B cell production of IL-10, following stimulation by resident bacteria and B cell regulatory function against colitis. In vitro, B cells from PI3KδD910A/D910A mice or wild-type B cells treated with PI3K specific inhibitors secreted significantly less IL-10 with greater IL-12p40 following bacterial stimulation. These B cells failed to suppress inflammatory cytokines by co-cultured microbiota-activated macrophages or CD4+ T cells. In vivo, co-transferred wild-type B cells ameliorated T cell-mediated colitis, while PI3KδD910A/D910A B cells did not confer protection from mucosal inflammation. These results indicate that PI3Kδ-signaling mediates regulatory B cell immune differentiation when stimulated with resident microbiota or their components, and is critical for induction and regulatory function of IL-10-producing B cells in intestinal homeostasis and inflammation.


Assuntos
Linfócitos B/fisiologia , Classe I de Fosfatidilinositol 3-Quinases/fisiologia , Enterite/genética , Interleucina-10/metabolismo , Microbiota/fisiologia , Animais , Linfócitos B/metabolismo , Células Cultivadas , Classe I de Fosfatidilinositol 3-Quinases/genética , Enterite/metabolismo , Enterite/microbiologia , Enterite/patologia , Inflamação/genética , Inflamação/metabolismo , Inflamação/microbiologia , Inflamação/patologia , Interleucina-10/farmacologia , Intestinos/efeitos dos fármacos , Intestinos/microbiologia , Intestinos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
5.
Proc Natl Acad Sci U S A ; 116(17): 8380-8389, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-30948643

RESUMO

The PIK3CA gene, which encodes the p110α catalytic subunit of PI3 kinase (PI3K), is mutationally activated in cancer and in overgrowth disorders known as PIK3CA-related overgrowth spectrum (PROS). To determine the consequences of genetic PIK3CA activation in a developmental context of relevance to both PROS and cancer, we engineered isogenic human induced pluripotent stem cells (iPSCs) with heterozygous or homozygous knockin of PIK3CAH1047R While heterozygous iPSCs remained largely similar to wild-type cells, homozygosity for PIK3CAH1047R caused widespread, cancer-like transcriptional remodeling, partial loss of epithelial morphology, up-regulation of stemness markers, and impaired differentiation to all three germ layers in vitro and in vivo. Genetic analysis of PIK3CA-associated cancers revealed that 64% had multiple oncogenic PIK3CA copies (39%) or additional PI3K signaling pathway-activating "hits" (25%). This contrasts with the prevailing view that PIK3CA mutations occur heterozygously in cancer. Our findings suggest that a PI3K activity threshold determines pathological consequences of oncogenic PIK3CA activation and provide insight into the specific role of this pathway in human pluripotent stem cells.


Assuntos
Classe I de Fosfatidilinositol 3-Quinases , Células-Tronco Pluripotentes Induzidas , Neoplasias , Adolescente , Linhagem Celular Tumoral , Classe I de Fosfatidilinositol 3-Quinases/genética , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Classe I de Fosfatidilinositol 3-Quinases/fisiologia , Feminino , Edição de Genes , Regulação Neoplásica da Expressão Gênica/genética , Técnicas de Introdução de Genes , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/fisiologia , Masculino , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Transdução de Sinais/genética
6.
Nat Commun ; 10(1): 1546, 2019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30948720

RESUMO

The insulin/IGF-1 signalling pathway is a key regulator of metabolism and the rate of ageing. We previously documented that systemic inactivation of phosphoinositide 3-kinase (PI3K) p110α, the principal PI3K isoform that positively regulates insulin signalling, results in a beneficial metabolic effect in aged mice. Here we demonstrate that deletion of p110α specifically in the adipose tissue leads to less fat accumulation over a significant part of adult life and allows the maintenance of normal glucose tolerance despite insulin resistance. This effect of p110α inactivation is due to a potentiating effect on ß-adrenergic signalling, which leads to increased catecholamine-induced energy expenditure in the adipose tissue. Our findings provide a paradigm of how partial inactivation of an essential component of the insulin signalling pathway can have an overall beneficial metabolic effect and suggest that PI3K inhibition could potentiate the effect of ß-adrenergic agonists in the treatment of obesity and its associated comorbidities.


Assuntos
Tecido Adiposo/metabolismo , Classe I de Fosfatidilinositol 3-Quinases/fisiologia , Fatores Etários , Animais , Classe I de Fosfatidilinositol 3-Quinases/genética , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Resistência à Insulina/genética , Camundongos Transgênicos , Obesidade/metabolismo , Transdução de Sinais
7.
Mol Cell ; 71(2): 343-351.e4, 2018 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-30029007

RESUMO

Class II phosphoinositide 3-kinases (PI3K-C2) are large multidomain enzymes that control cellular functions ranging from membrane dynamics to cell signaling via synthesis of 3'-phosphorylated phosphoinositides. Activity of the alpha isoform (PI3K-C2α) is associated with endocytosis, angiogenesis, and glucose metabolism. How PI3K-C2α activity is controlled at sites of endocytosis remains largely enigmatic. Here we show that the lipid-binding PX-C2 module unique to class II PI3Ks autoinhibits kinase activity in solution but is essential for full enzymatic activity at PtdIns(4,5)P2-rich membranes. Using HDX-MS, we show that the PX-C2 module folds back onto the kinase domain, inhibiting its basal activity. Destabilization of this intramolecular contact increases PI3K-C2α activity in vitro and in cells, leading to accumulation of its lipid product, increased recruitment of the endocytic effector SNX9, and facilitated endocytosis. Our studies uncover a regulatory mechanism in which coincident binding of phosphoinositide substrate and cofactor selectively activate PI3K-C2α at sites of endocytosis.


Assuntos
Classe II de Fosfatidilinositol 3-Quinases/metabolismo , Classe II de Fosfatidilinositol 3-Quinases/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Animais , Domínios C2/fisiologia , Células COS , Chlorocebus aethiops , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Classe I de Fosfatidilinositol 3-Quinases/fisiologia , Clatrina/fisiologia , Endocitose/fisiologia , Células HEK293 , Homeostase , Humanos , Lipídeos/fisiologia , Espectrometria de Massas , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Ligação Proteica , Domínios Proteicos , Transdução de Sinais
8.
Physiol Rep ; 6(12): e13733, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29932504

RESUMO

Nitric oxide, produced by eNOS, plays critical roles in the regulation of vascular function and maintenance. Chronic PI3K signaling has recently been associated with vascular malformations. A well described substrate downstream of PI3K signaling is eNOS. Another critical downstream target of PI3K is the metabolic regulator, mTORc1. The relationship between mTORc1 and eNOS regulation, has not been determined. We generated cells with manipulated PI3K signaling by expressing the activating mutation, PIK3CAH1047R , or knocking down PTEN expression. We investigated eNOSS1177 phosphorylation, a major activating regulatory site, following mTORC1 inhibition. We also tested the sufficiency of mTORc1 activation to stimulate eNOSS1177 phosphorylation. Our data indicate mTORc1 activity is required for the phosphorylation of eNOSS1177 , even in the presence of robust AKT activation. Moreover, we found that expression of RHEB, which functions in the absence of AKT activation to activate mTORc1, is sufficient to phosphorylate this site. Our data indicate that mTORc1, rather than AKT, may be the critical determinant of eNOSS1177 phosphorylation. As mTORc1 is a central regulator of cellular metabolism, the finding that this regulatory complex can directly participate in the regulation of eNOS provides new insights into metabolic uncoupling and vascular disease that often accompanies diabetes, high fat diets, and aging.


Assuntos
Células Endoteliais/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/fisiologia , Óxido Nítrico Sintase Tipo III/metabolismo , Células Cultivadas , Classe I de Fosfatidilinositol 3-Quinases/fisiologia , Ativação Enzimática/fisiologia , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/fisiologia , Proteína Enriquecida em Homólogo de Ras do Encéfalo/fisiologia
9.
EMBO J ; 37(8)2018 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-29581096

RESUMO

Adult mouse muscle satellite cells (MuSCs) are quiescent in uninjured muscles. Upon injury, MuSCs exit quiescence in vivo to become activated, re-enter the cell cycle to proliferate, and differentiate to repair the damaged muscles. It remains unclear which extrinsic cues and intrinsic signaling pathways regulate quiescence exit during MuSC activation. Here, we demonstrated that inducible MuSC-specific deletion of p110α, a catalytic subunit of phosphatidylinositol 3-kinase (PI3K), rendered MuSCs unable to exit quiescence, resulting in severely impaired MuSC proliferation and muscle regeneration. Genetic reactivation of mTORC1, or knockdown of FoxOs, in p110α-null MuSCs partially rescued the above defects, making them key effectors downstream of PI3K in regulating quiescence exit. c-Jun was found to be a key transcriptional target of the PI3K/mTORC1 signaling axis essential for MuSC quiescence exit. Moreover, induction of a constitutively active PI3K in quiescent MuSCs resulted in spontaneous MuSC activation in uninjured muscles and subsequent depletion of the MuSC pool. Thus, PI3K-p110α is both necessary and sufficient for MuSCs to exit quiescence in response to activating signals.


Assuntos
Classe I de Fosfatidilinositol 3-Quinases/fisiologia , Células Satélites de Músculo Esquelético/fisiologia , Animais , Proliferação de Células , Células Cultivadas , Camundongos Mutantes , Músculo Esquelético/lesões , Músculo Esquelético/fisiologia , Regeneração
10.
Genet Med ; 20(8): 882-889, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29446767

RESUMO

PURPOSE: CLAPO syndrome is a rare vascular disorder characterized by capillary malformation of the lower lip, lymphatic malformation predominant on the face and neck, asymmetry, and partial/generalized overgrowth. Here we tested the hypothesis that, although the genetic cause is not known, the tissue distribution of the clinical manifestations in CLAPO seems to follow a pattern of somatic mosaicism. METHODS: We clinically evaluated a cohort of 13 patients with CLAPO and screened 20 DNA blood/tissue samples from 9 patients using high-throughput, deep sequencing. RESULTS: We identified five activating mutations in the PIK3CA gene in affected tissues from 6 of the 9 patients studied; one of the variants (NM_006218.2:c.248T>C; p.Phe83Ser) has not been previously described in developmental disorders. CONCLUSION: We describe for the first time the presence of somatic activating PIK3CA mutations in patients with CLAPO. We also report an update of the phenotype and natural history of the syndrome.


Assuntos
Malformações Arteriovenosas/genética , Malformações Arteriovenosas/fisiopatologia , Classe I de Fosfatidilinositol 3-Quinases/genética , Doenças Linfáticas/genética , Doenças Linfáticas/fisiopatologia , Adolescente , Adulto , Criança , Classe I de Fosfatidilinositol 3-Quinases/fisiologia , Feminino , Estudos de Associação Genética/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Masculino , Mutação , Fosfatidilinositol 3-Quinases/genética , Estudos Retrospectivos
11.
J Leukoc Biol ; 100(4): 801-810, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27154356

RESUMO

Upon in vitro stimulation, neutrophils undergo a cell death named netosis. This process is characterized by extracellular release of chromatin scaffold associated with granular and cytoplasmic proteins, which together, ensnare and kill microbes. We have previously described that interaction of Leishmania amazonensis with human neutrophils leads to the release of neutrophil extracellular traps, which trap and kill the parasite. However, the signaling leading to Leishmania induced netosis is still unknown. Thus, we sought to evaluate signaling events that drive L. amazonensis induced neutrophil extracellular trap release from human neutrophils. Here, we found that PI3K, independently of protein kinase B, has a role in parasite-induced netosis. We also described that the main isoforms involved are PI3Kγ and PI3Kδ, which work in reactive oxygen species-dependent and -independent ways, respectively. We demonstrated that activation of ERK downstream of PI3Kγ is important to trigger reactive oxygen species-dependent, parasite-induced netosis. Pharmacological inhibition of protein kinase C also significantly decreased parasite-induced neutrophil extracellular trap release. Intracellular calcium, regulated by PI3Kδ, represents an alternative reactive oxygen species-independent pathway of netosis stimulated by L. amazonensis Finally, intracellular calcium mobilization and reactive oxygen species generation are the major regulators of parasite-induced netosis. Our results contribute to a better understanding of the signaling behind netosis induced by interactions between Leishmania and neutrophils.


Assuntos
Sinalização do Cálcio/fisiologia , Classe I de Fosfatidilinositol 3-Quinases/fisiologia , Classe Ib de Fosfatidilinositol 3-Quinase/fisiologia , Armadilhas Extracelulares/parasitologia , Leishmania mexicana/imunologia , Sistema de Sinalização das MAP Quinases , Neutrófilos/imunologia , Proteína Quinase C/fisiologia , Cromatina/ultraestrutura , Classe I de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Humanos , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/fisiologia , Espécies Reativas de Oxigênio/metabolismo
12.
Mol Autism ; 7: 3, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26770665

RESUMO

BACKGROUND: Dysfunctions in the PI3K/mTOR pathway have gained a lot of attention in autism research. This was initially based on the discovery of several monogenic autism spectrum disorders with mutations or defects in PI3K/mTOR signaling components. Recent genetic studies corroborate that defective PI3K/mTOR signaling might be a shared pathomechanism in autism disorders of so far unknown etiology, but functional molecular analyses in human cells are rare. The goals of this study were to perform a functional screen of cell lines from patients with idiopathic autism for defects in PI3K/mTOR signaling, to test if further functional analyses are suitable to detect underlying molecular mechanisms, and to evaluate this approach as a biomarker tool to identify therapeutic targets. METHODS: We performed phospho-S6- and S6-specific ELISA experiments on 21 lymphoblastoid cell lines from the AGRE collection and on 37 lymphoblastoid cell lines from the Simons Simplex Collection and their healthy siblings. Cell lines from one individual with increased S6 phosphorylation and his multiplex family were analyzed in further detail to identify upstream defects in PI3K signaling associated with autism diagnosis. RESULTS: We detected significantly increased S6 phosphorylation in 3 of the 21 lymphoblastoid cell lines from AGRE compared to a healthy control and in 1 of the 37 lymphoblastoid cell lines from the Simons Simplex Collection compared to the healthy sibling. Further analysis of cells from one individual with elevated S6 phosphorylation showed increased expression of the PI3K catalytic subunit p110δ, which was also observed in lymphoblastoid cells from other autistic siblings but not unaffected members in his multiplex family. The p110δ-selective inhibitor IC87114 reduced elevated S6 phosphorylation and protein synthesis in this cell line. CONCLUSIONS: Our results suggest that functional analysis of PI3K/mTOR signaling is a biomarker tool to identify disease-associated molecular defects that could serve as therapeutic targets in autism. Using this approach, we discovered impaired signaling and protein synthesis through the PI3K catalytic subunit p110δ as an underlying molecular defect and potential treatment target in select autism spectrum disorders. Increased p110δ activity was recently associated with schizophrenia, and our results suggest that p110δ may also be implicated in autism.


Assuntos
Transtorno Autístico/enzimologia , Classe I de Fosfatidilinositol 3-Quinases/biossíntese , Proteínas do Tecido Nervoso/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Adenina/análogos & derivados , Adenina/farmacologia , Transtorno Autístico/genética , Transtorno Autístico/patologia , Biomarcadores , Linhagem Celular , Classe I de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Classe I de Fosfatidilinositol 3-Quinases/genética , Classe I de Fosfatidilinositol 3-Quinases/fisiologia , Doenças em Gêmeos , Ensaio de Imunoadsorção Enzimática , Saúde da Família , Feminino , Humanos , Linfócitos/enzimologia , Masculino , Terapia de Alvo Molecular , Proteínas do Tecido Nervoso/genética , Fosforilação , Processamento de Proteína Pós-Traducional , Quinazolinas/farmacologia , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/fisiologia
13.
J Med Chem ; 55(11): 5188-219, 2012 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-22548365

RESUMO

A highly selective series of inhibitors of the class I phosphatidylinositol 3-kinases (PI3Ks) has been designed and synthesized. Starting from the dual PI3K/mTOR inhibitor 5, a structure-based approach was used to improve potency and selectivity, resulting in the identification of 54 as a potent inhibitor of the class I PI3Ks with excellent selectivity over mTOR, related phosphatidylinositol kinases, and a broad panel of protein kinases. Compound 54 demonstrated a robust PD-PK relationship inhibiting the PI3K/Akt pathway in vivo in a mouse model, and it potently inhibited tumor growth in a U-87 MG xenograft model with an activated PI3K/Akt pathway.


Assuntos
Classe I de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Piperazinas/síntese química , Piridinas/síntese química , Sulfonamidas/síntese química , Triazinas/síntese química , Animais , Disponibilidade Biológica , Classe I de Fosfatidilinositol 3-Quinases/fisiologia , Cristalografia por Raios X , Desenho de Fármacos , Feminino , Humanos , Indazóis/síntese química , Indazóis/farmacocinética , Indazóis/farmacologia , Camundongos , Camundongos Nus , Microssomos Hepáticos/metabolismo , Modelos Moleculares , Piperazinas/farmacocinética , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Purinas/síntese química , Purinas/farmacocinética , Purinas/farmacologia , Pirazóis/síntese química , Pirazóis/farmacocinética , Pirazóis/farmacologia , Piridinas/farmacocinética , Piridinas/farmacologia , Pirimidinas/síntese química , Pirimidinas/farmacocinética , Pirimidinas/farmacologia , Ratos , Transdução de Sinais , Relação Estrutura-Atividade , Sulfonamidas/farmacocinética , Sulfonamidas/farmacologia , Sulfonas/síntese química , Sulfonas/farmacocinética , Sulfonas/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Triazinas/farmacocinética , Triazinas/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA