Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Lancet Oncol ; 20(6): 806-815, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31036468

RESUMO

BACKGROUND: Tucidinostat (formerly known as chidamide) is an oral subtype-selective histone deacetylase inhibitor. In an exploratory study, the combination of tucidinostat with exemestane showed preliminary signs of encouraging anti-tumour activity in patients with advanced hormone receptor-positive breast cancer. To build on these findings, we aimed to assess the efficacy and safety of this combination in a randomised trial in a larger population of postmenopausal patients with advanced, hormone receptor-positive breast cancer. METHODS: We did the randomised, double-blind, placebo-controlled, phase 3 ACE trial at 22 specialist cancer centres in China. Eligible patients were postmenopausal women (aged ≥60 years or aged <60 years if their serum follicle-stimulating hormone and oestradiol concentrations were within postmenopausal ranges) with hormone receptor-positive, HER2-negative breast cancer, whose disease had relapsed or progressed after at least one endocrine therapy (either in advanced or metastatic or adjuvant setting), and who had at least one measurable lesion, adequate organ function, Eastern Cooperative Oncology Group (ECOG) performance status of 0-1, and adequate haematological and biochemical parameters. Endocrine therapy did not have to be the most recent therapy before randomisation, but recurrence or progression after the most recent therapy was a prerequisite. Patients were randomly assigned (2:1) by a dynamic randomisation scheme via an interactive web-response system to receive 30 mg oral tucidinostat or placebo twice weekly. All patients in both groups also received 25 mg oral exemestane daily. Randomisation was stratified according to the presence of visceral metastases (yes vs no). Patients, investigators, study site staff, and the sponsor were masked to treatment assignment. The primary endpoint was investigator-assessed progression-free survival. Efficacy analyses were done in the full analysis set population, comprising all patients who received at least one dose of any study treatment, and safety analyses were done in all patients who received at least one dose of any study treatment and for whom at least one safety case report form was available. This study is registered with ClinicalTrials.gov, number NCT02482753. The study has reached the required number of events for final analysis of the primary endpoint. The trial is no longer enrolling patients, but follow-up for investigation of overall survival is ongoing. FINDINGS: Between July 20, 2015, and June 26, 2017, 365 patients were enrolled and randomly assigned, 244 to the tucidinostat group and 121 to the placebo group. The median duration of follow-up was 13·9 months (IQR 9·8-17·5). Investigator-assessed median progression-free survival was 7·4 months (95% CI 5·5-9·2) in the tucidinostat group and 3·8 months (3·7-5·5) in the placebo group (HR 0·75 [95% CI 0·58-0·98]; p=0·033). The most common grade 3 or 4 adverse events in either group were neutropenia (124 [51%] of 244 patients in the tucidinostat group vs three [2%] of 121 patients in the placebo group), thrombocytopenia (67 [27%] vs three [2%]), and leucopenia (46 [19%] vs three [2%]). Serious adverse events of any cause occurred in 51 (21%) of 244 patients in the tucidinostat group and seven (6%) of 121 patients in the placebo group. No treatment-related deaths were reported. INTERPRETATION: Tucidinostat plus exemestane improved progression-free survival compared with placebo plus exemestane in patients with advanced, hormone receptor-positive, HER2-negative breast cancer that progressed after previous endocrine therapy. Grade 3-4 haematological adverse events were more common in the tucidinostat plus exemestane group than in the placebo plus exemestane group. Tucidinostat plus exemestane could represent a new treatment option for these patients. FUNDING: Chipscreen Biosciences.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Ósseas/tratamento farmacológico , Neoplasias da Mama/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Terapia de Salvação , Aminopiridinas/administração & dosagem , Androstadienos/administração & dosagem , Benzamidas/administração & dosagem , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/secundário , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Método Duplo-Cego , Feminino , Seguimentos , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/patologia , Pós-Menopausa , Receptor ErbB-2/metabolismo , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Taxa de Sobrevida , Resultado do Tratamento
2.
Anticancer Drugs ; 30(9): 909-916, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30998512

RESUMO

As a novel orally active multitarget small molecule inhibitor, CS2164 has shown broad antitumor activities against several human tumor xenograft models in immune-compromised mice. However, the ability of CS2164 to modulate antitumor immunity in an immune-competent mouse tumor model remains undefined, although antiangiogenic treatment has been reported to affect immune cell infiltration and remodel the tumor immune microenvironment. In the present study, the subcutaneous and ascites hepatocellular carcinoma (HCC) models in syngeneic Balb/c mice established by inoculation of an H22 hepatoma cell line were utilized to investigate the antitumor and immunomodulatory effects of CS2164. Although the antitumor effects of CS2164 were validated in both subcutaneous and ascites HCC models in syngeneic mice, CS2164 treatment consistently modulated immune cell populations, both in the periphery and in tumor microenvironments, with upregulation of CD4 and CD8 T cells in the spleen, but downregulation of immunosuppressive populations including regulatory T cells, myeloid-derived suppressor cells, and tumor-associated macrophages in the spleen and tumor tissues. Furthermore, CS2164 increased the relative gene expression and protein production of several proinflammatory cytokines in tumor-related ascites. These results indicate that CS2164 exerts an antitumor effect associated with its immunomodulatory activities in mouse HCC models, and may also provide evidence for the immunotherapy potentiation of CS2164 in future cancer treatment.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Fatores Imunológicos/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Naftalenos/farmacologia , Quinolinas/farmacologia , Inibidores da Angiogênese/farmacologia , Animais , Apoptose/efeitos dos fármacos , Ascite/tratamento farmacológico , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Imunoterapia/métodos , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Microambiente Tumoral/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
3.
Cancer Sci ; 108(3): 469-477, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28004478

RESUMO

Although inhibitors targeting tumor angiogenic pathway have provided improvement for clinical treatment in patients with various solid tumors, the still very limited anti-cancer efficacy and acquired drug resistance demand new agents that may offer better clinical benefits. In the effort to find a small molecule potentially targeting several key pathways for tumor development, we designed, discovered and evaluated a novel multi-kinase inhibitor, CS2164. CS2164 inhibited the angiogenesis-related kinases (VEGFR2, VEGFR1, VEGFR3, PDGFRα and c-Kit), mitosis-related kinase Aurora B and chronic inflammation-related kinase CSF-1R in a high potency manner with the IC50 at a single-digit nanomolar range. Consequently, CS2164 displayed anti-angiogenic activities through suppression of VEGFR/PDGFR phosphorylation, inhibition of ligand-dependent cell proliferation and capillary tube formation, and prevention of vasculature formation in tumor tissues. CS2164 also showed induction of G2/M cell cycle arrest and suppression of cell proliferation in tumor tissues through the inhibition of Aurora B-mediated H3 phosphorylation. Furthermore, CS2164 demonstrated the inhibitory effect on CSF-1R phosphorylation that led to the suppression of ligand-stimulated monocyte-to-macrophage differentiation and reduced CSF-1R+ cells in tumor tissues. The in vivo animal efficacy studies revealed that CS2164 induced remarkable regression or complete inhibition of tumor growth at well-tolerated oral doses in several human tumor xenograft models. Collectively, these results indicate that CS2164 is a highly selective multi-kinase inhibitor with potent anti-tumor activities against tumor angiogenesis, mitosis and chronic inflammation, which may provide the rationale for further clinical assessment of CS2164 as a therapeutic agent in the treatment of cancer.


Assuntos
Adenocarcinoma/tratamento farmacológico , Inibidores da Angiogênese/uso terapêutico , Neoplasias do Colo/tratamento farmacológico , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Mitose/efeitos dos fármacos , Neovascularização Patológica/tratamento farmacológico , Fenilenodiaminas/uso terapêutico , Quinolinas/uso terapêutico , Células 3T3 , Animais , Aurora Quinase B/antagonistas & inibidores , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Histonas/metabolismo , Humanos , Inflamação/tratamento farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Simulação de Acoplamento Molecular , Naftalenos , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-kit/antagonistas & inibidores , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto
4.
J Diabetes ; 16(2): e13484, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37853916

RESUMO

BACKGROUND: To evaluate the glycemic control effects of vhiglitazar (carfloglitazar), a novel peroxisome proliferator-activated receptor pan-agonist, in patients with type 2 diabetes mellitus (T2DM) with metabolic syndrome (MetS) or insulin resistance (IR) using pooled data analysis of two phase III clinical trials. METHODS: Data were collected from two randomized phase III clinical trials in China, comparing chiglitazar to placebo or sitagliptin in T2DM patients. The MetS was defined by the Adult Treatment Panel III MetS criteria, and IR was defined by homeostatic model assessment for insulin resistance (HOMA-IR) ≥4.31 (male) or 4.51 (female). The main end point of this analysis was glycemic control in the different arms within each subgroup. RESULTS: In the MetS subgroup, changes in glycated hemoglobin (HbA1c) from baseline at week 24 in the chiglitazar 32 mg, chiglitazar 48 mg, and sitagliptin 100 mg arms were -1.44%, -1.68%, and -1.37%, respectively; p < .05 was obtained when chiglitazar 48 mg was compared with sitagliptin. In the IR subgroup, the changes in HbA1c were -1.58%, -1.56%, and -1.26% in chiglitazar 32 mg, chiglitazar 48 mg, and sitagliptin 100 mg arms, respectively; p < .05 was obtained when chiglitazar 32 mg was compared with sitaligptin. The two doses of chiglitazar demonstrated a greater reduction in fasting plasma glucose and 2 h postprandial plasma glucose than sitagliptin in the pooled population and in the MetS and IR subgroups. CONCLUSIONS: Chiglitazar shows promising efficacy for glycemic control in patients with T2DM associated with MetS or IR. Further prospective trials are required to validate these findings.


Assuntos
Carbazóis , Diabetes Mellitus Tipo 2 , Resistência à Insulina , Síndrome Metabólica , Propionatos , Adulto , Humanos , Masculino , Feminino , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/tratamento farmacológico , Síndrome Metabólica/complicações , Síndrome Metabólica/tratamento farmacológico , Hemoglobinas Glicadas , Glicemia/metabolismo , Controle Glicêmico , Hipoglicemiantes/uso terapêutico , Fosfato de Sitagliptina/uso terapêutico
5.
J Hematol Oncol ; 17(1): 37, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38822399

RESUMO

Histone deacetylase (HDAC) serves as a critical molecular regulator in the pathobiology of various malignancies and have garnered attention as a viable target for therapeutic intervention. A variety of HDAC inhibitors (HDACis) have been developed to target HDACs. Many preclinical studies have conclusively demonstrated the antitumor effects of HDACis, whether used as monotherapy or in combination treatments. On this basis, researchers have conducted various clinical studies to evaluate the potential of selective and pan-HDACis in clinical settings. In our work, we extensively summarized and organized current clinical trials, providing a comprehensive overview of the current clinical advancements in targeting HDAC therapy. Furthermore, we engaged in discussions about several clinical trials that did not yield positive outcomes, analyzing the factors that led to their lack of anticipated therapeutic effectiveness. Apart from the experimental design factors, issues such as toxicological side effects, tumor heterogeneity, and unexpected off-target effects also contributed to these less-than-expected results. These challenges have naturally become significant barriers to the application of HDACis. Despite these challenges, we believe that advancements in HDACi research and improvements in combination therapies will pave the way or lead to a broad and hopeful future in the treatment of solid tumors.


Assuntos
Inibidores de Histona Desacetilases , Histona Desacetilases , Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Inibidores de Histona Desacetilases/uso terapêutico , Histona Desacetilases/metabolismo , Animais , Ensaios Clínicos como Assunto , Antineoplásicos/uso terapêutico , Antineoplásicos/farmacologia , Terapia de Alvo Molecular/métodos
6.
Nat Genet ; 56(5): 846-860, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38641644

RESUMO

Methylation quantitative trait loci (mQTLs) are essential for understanding the role of DNA methylation changes in genetic predisposition, yet they have not been fully characterized in East Asians (EAs). Here we identified mQTLs in whole blood from 3,523 Chinese individuals and replicated them in additional 1,858 Chinese individuals from two cohorts. Over 9% of mQTLs displayed specificity to EAs, facilitating the fine-mapping of EA-specific genetic associations, as shown for variants associated with height. Trans-mQTL hotspots revealed biological pathways contributing to EA-specific genetic associations, including an ERG-mediated 233 trans-mCpG network, implicated in hematopoietic cell differentiation, which likely reflects binding efficiency modulation of the ERG protein complex. More than 90% of mQTLs were shared between different blood cell lineages, with a smaller fraction of lineage-specific mQTLs displaying preferential hypomethylation in the respective lineages. Our study provides new insights into the mQTL landscape across genetic ancestries and their downstream effects on cellular processes and diseases/traits.


Assuntos
Metilação de DNA , População do Leste Asiático , Locos de Características Quantitativas , Feminino , Humanos , Masculino , População do Leste Asiático/genética , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla/métodos , Herança Multifatorial , Polimorfismo de Nucleotídeo Único
7.
Front Pharmacol ; 13: 932914, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36120308

RESUMO

Histone deacetylase (HDAC) is one of the most characterized epigenetic modifiers, modulating chromatin structure and gene expression, which plays an important role in cell cycle, differentiation and apoptosis. Dysregulation of HDAC promotes cancer progression, thus inhibitors targeting HDACs have evidently shown therapeutic efficacy in multiple cancers. Tucidinostat (formerly known as chidamide), a novel subtype-selective HDAC inhibitor, inhibits Class I HDAC1, HDAC2, HDAC3, as well as Class IIb HDAC10. Tucidinostat is approved in relapsed or refractory (R/R) peripheral T-cell lymphoma (PTCL), advanced breast cancer and R/R adult T-cell leukemia-lymphoma (ATLL). Compared with other HDAC inhibitors, tucidinostat shows notable antitumor activity, remarkable synergistic effect with immunotherapy, and manageable toxicity. Here, we comprehensively summarize recent advances in tucidinostat as both monotherapy and a regimen of combination therapy in both hematological and solid malignancies in clinic. Further studies will endeavor to identify more combination strategies with tucidinostat and to identify specific clinical biomarkers to predict the therapeutic effect.

8.
Bioorg Med Chem Lett ; 21(1): 110-6, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21145737

RESUMO

Novel 2-aminoanilide histone deacetylase (HDAC) inhibitors were designed to increase their contact with surface residues surrounding the HDAC active site compared to the contacts made by existing clinical 2-aminoanilides such as SNDX-275, MGCD0103, and Chidamide. Their HDAC selectivity was assessed using p21 and klf2 reporter gene assays in HeLa and A204 cells, respectively, which provide a cell-based readout for the inhibition of HDACs associated either with the p21 or klf2 promoter. A subset of the designed compounds selectively induced p21 over klf2 relative to the clinical reference compound SNDX-275. A representative lead compound from this subset had antiproliferative effects in cancer cells associated with induction of acetylated histone H4, endogenous p21, cell cycle arrest, and apoptosis. The p21- versus klf2-selective compounds described herein may provide a chemical starting point for developing clinically-differentiated HDAC inhibitors for cancer therapy.


Assuntos
Antineoplásicos/química , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidores de Histona Desacetilases/química , Histona Desacetilases/química , Fatores de Transcrição Kruppel-Like/genética , Aminopiridinas/uso terapêutico , Anilidas/síntese química , Anilidas/química , Anilidas/uso terapêutico , Antineoplásicos/síntese química , Antineoplásicos/uso terapêutico , Benzamidas/uso terapêutico , Domínio Catalítico , Linhagem Celular Tumoral , Genes Reporter , Inibidores de Histona Desacetilases/síntese química , Inibidores de Histona Desacetilases/uso terapêutico , Histona Desacetilases/metabolismo , Humanos , Neoplasias/tratamento farmacológico , Regiões Promotoras Genéticas , Pirimidinas/uso terapêutico , Relação Estrutura-Atividade
9.
J Oncol ; 2021: 4936207, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34194495

RESUMO

As a selective histone deacetylase (HDAC) inhibitor developed in China, chidamide has been applied for the treatment of refractory peripheral T-cell lymphoma (PTCL) and multiple solid tumors, including lung cancer. However, the underlying mechanisms are not well elucidated. In our present study, we found that chidamide and radiation acted synergistically to suppress cell and xenograft growth of lung squamous cell carcinoma cells by inducing cell apoptosis. Moreover, chidamide alone or a combination of chidamide and radiation treatment inhibited cancer cell stemness. miRNA microarray analysis demonstrated that miR-375 was the highest upregulated microRNA (miRNA) in NCI-2170 and NCI-H226 cells treated with chidamide alone or treated with chidamide plus radiation, compared with normal control. Inhibition of miR-375 attenuated the promoting effect of chidamide alone and chidamide plus radiation-induced NCI-2170 and NCI-H226 cell apoptosis and reverted the suppression of cancer stemness caused by chidamide alone or chidamide plus radiation treatment. Moreover, EIF4G3, a scaffold protein in the translation initiation complex, was found to be a direct target of miR-375 based on the luciferase reporter assay and western blot analysis. Interestingly, both chidamide alone and chidamide plus radiation treatments suppressed the mRNA and protein expression of EIF4G3. Silence of EIF4G3 also induced cell apoptosis and suppressed tumor growth in NCI-2170 and NCI-H226 cells. These data suggest that chidamide shows a synergistic effect with radiation therapy on lung squamous cell carcinomas by modulating the miR-375-EIF4G3 axis, which may afford an effective strategy to overcome the drug resistance of chidamide in clinical cancer therapy.

10.
Sci Bull (Beijing) ; 66(15): 1571-1580, 2021 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-36654286

RESUMO

Chiglitazar (Carfloglitazar) is a novel non-thiazolidinedione (TZD) structured peroxisome proliferator-activated receptor (PPAR) pan-agonist that has shown promising effects on glycemic control and lipid regulation in patients with type 2 diabetes in previous clinical studies. This randomized phase 3 trial aimed to compare the efficacy and safety of chiglitazar with placebo in patients with type 2 diabetes with insufficient glycemic control by strict diet and exercise alone. Eligible patients were randomly assigned to receive chiglitazar 32 mg (n = 167), chiglitazar 48 mg (n = 166), or placebo (n = 202) once daily. The primary endpoint was the change in glycosylated hemoglobin A1c (HbA1c) at week 24 with superiority of chiglitazar over placebo. The results showed that both chiglitazar 32 and 48 mg resulted in significant and clinically meaningful reductions in HbA1c, and placebo-adjusted estimated treatment differences at week 24 for chiglitazar 32 and 48 mg were -0.87% (95% confidential interval (CI): -1.10 to -0.65; P < 0.0001) and -1.05% (95% CI: -1.29 to -0.81; P < 0.0001), respectively. Secondary efficacy parameters including glycemic control, insulin sensitivity and triglyceride reduction were also significantly improved in the chiglitazar groups. The overall frequency of adverse events and study discontinuation attributable to adverse events were similar among the groups. Low incidences of mild edema and body weight gain were reported in the chiglitazar dose groups. The results from this phase 3 trial demonstrated that the PPAR pan-agonist chiglitazar possesses an overall good efficacy and safety profile in patients with type 2 diabetes inadequately controlled with lifestyle interventions, thereby providing adequate supporting evidence for using this PPAR pan-agonist as a treatment option for type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2 , Humanos , Diabetes Mellitus Tipo 2/tratamento farmacológico , Receptores Ativados por Proliferador de Peroxissomo/uso terapêutico , Hipoglicemiantes/efeitos adversos , Carbazóis
11.
Sci Bull (Beijing) ; 66(15): 1581-1590, 2021 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-36654287

RESUMO

Chiglitazar (Carfloglitazar) is a novel peroxisome proliferator-activated receptor (PPAR) pan-agonist that has shown promising effects on glycemic control and lipid regulation in patients with type 2 diabetes. In this randomized phase 3 trial, we compared the efficacy and safety of chiglitazar with sitagliptin in patients with type 2 diabetes who had insufficient glycemic control despite a strict diet and exercise regimen. Eligible patients were randomized (1:1:1) to receive chiglitazar 32 mg (n = 245), chiglitazar 48 mg (n = 246), or sitagliptin 100 mg (n = 248) once daily for 24 weeks. The primary endpoint was the change in glycosylated hemoglobin A1C (HbA1c) from baseline at week 24 with the non-inferiority of chiglitazar over sitagliptin. Both chiglitazar and sitagliptin significantly reduced HbA1c at week 24 with values of -1.40%, -1.47%, and -1.39% for chiglitazar 32 mg, chiglitazar 48 mg, and sitagliptin 100 mg, respectively. Chiglitazar 32 and 48 mg were both non-inferior to sitagliptin 100 mg, with mean differences of -0.04% (95% confidential interval (CI) -0.22 to 0.15) and -0.08% (95% CI -0.27 to 0.10), respectively. Compared with sitagliptin, greater reduction in fasting and 2-h postprandial plasma glucose and fasting insulin was observed with chiglitazar. Overall adverse event rates were similar between the groups. A small increase in mild edema in the chiglitazar 48 mg group and slight weight gain in both chiglitazar groups were reported. The overall results demonstrated that chiglitazar possesses good efficacy and safety profile in patients with type 2 diabetes inadequately controlled with lifestyle interventions, thereby providing adequate supporting evidence for using this PPAR pan-agonist as a treatment option for type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2 , Fosfato de Sitagliptina , Humanos , Fosfato de Sitagliptina/efeitos adversos , Diabetes Mellitus Tipo 2/tratamento farmacológico , Receptores Ativados por Proliferador de Peroxissomo , Hipoglicemiantes/efeitos adversos
12.
Eur Neurol ; 63(1): 52-9, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20029217

RESUMO

BACKGROUND/AIMS: Between 50 and 70% of stroke survivors suffer from severe disabilities such as paralysis and aphasia. Poor stroke outcome is a reflection of our incomplete understanding of the underlying mechanisms, and hence the capacity to implement appropriate treatment(s). We evaluated hypoxic tissue after stroke and patient condition severity and prognosis. METHODS: Hypoxic tissue volume was quantified within 14 days after stroke. Patients were classified as hypoxic positive or negative. Patients were evaluated at imaging and 21 days later. Prognosis was assessed at 30 and 90 days. RESULTS: Significant improvement was shown in hypoxia-positive (vs. hypoxia-negative) patients (p < 0.05). There were significant positive relationships between the volume of hypoxic tissue and the improvement in specialized test scores at 90 days (p < 0.05 for both). Presence of hypoxic tissue within 14 days after cerebral stroke was related to recovery at 3 weeks and prognosis at 90 days. CONCLUSIONS: The assessment of hypoxic tissue volume after stroke may be useful in predicting patient recovery.


Assuntos
Infarto Encefálico/diagnóstico , Infarto Encefálico/patologia , Cérebro/patologia , Hipóxia Encefálica/patologia , Acidente Vascular Cerebral/diagnóstico , Acidente Vascular Cerebral/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Infarto Encefálico/diagnóstico por imagem , Circulação Cerebrovascular , Cérebro/diagnóstico por imagem , Feminino , Humanos , Hipóxia Encefálica/diagnóstico por imagem , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Prognóstico , Análise de Regressão , Índice de Gravidade de Doença , Acidente Vascular Cerebral/diagnóstico por imagem , Fatores de Tempo , Tomografia Computadorizada de Emissão de Fóton Único , Tomografia Computadorizada por Raios X
13.
Int Immunopharmacol ; 77: 105914, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31634789

RESUMO

Rheumatoid arthritis (RA) is a representative autoimmune disease characterized by chronic inflammation and joint destruction. Although biological inhibitors such as TNF-α and IL-6 antibodies have achieved success in clinical therapy, small molecule inhibitors against the Janus kinases (JAKs) involved in the signaling pathways of various cytokine receptors have gained more attraction as safe and efficacious options. In this study, we identified CS12192 as a novel selective JAK3/JAK1/TBK1 inhibitor and investigated its pharmacological effects on the experimental arthritis models in rat and mouse. We found that CS12192 showed a more selective inhibitory activity on JAK3, and to a less extent on JAK1 and TBK1, that were verified by decreased activation of p-STATs and p-IRF3 as well as down-regulation of IFN gene expression in the cultured cells with relevant stimuli. Furthermore, oral treatment with CS12192 dose-dependently ameliorated the disease severity, hind paw swelling, body weight loss, and bone destruction in rat models of adjuvant-induced arthritis (AIA) and collagen-induced arthritis (CIA). In a mouse CIA model, CS12192 also attenuated the disease severity, which was correlated with the suppressed CD4+ T cell activation and Th17 function, as well as the reduced cytokine levels in sera and pro-inflammatory cytokine and chemokine gene expression in joint tissue. Corroboratively, RANKL-induced osteoclast formation was inhibited by CS12192. Thus, these results suggest that CS12192 as a novel selective JAK inhibitor has therapeutic potential for the treatment of RA and may provide a new strategy for the control of autoimmune diseases.


Assuntos
Artrite Experimental/tratamento farmacológico , Artrite Reumatoide/tratamento farmacológico , Janus Quinase 1/antagonistas & inibidores , Janus Quinase 3/antagonistas & inibidores , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Animais , Artrite Experimental/sangue , Artrite Experimental/imunologia , Artrite Reumatoide/sangue , Artrite Reumatoide/imunologia , Citocinas/sangue , Citocinas/genética , Feminino , Humanos , Camundongos , Camundongos Endogâmicos DBA , Osteoclastos/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Células RAW 264.7 , Ratos Endogâmicos Lew , Células THP-1
14.
Phytother Res ; 22(10): 1400-3, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18690615

RESUMO

The dried, immature fruit of Citrus aurantium L., 'Zhiqiao' in Chinese, has been used to treat cardiovascular diseases in traditional Chinese medicine for centuries. Naringenin and hesperetin and their glycosides present in considerable amounts (about 10 approximately 15%) in the herb. The aim of this study is to test whether naringenin and hesperetin influence adiponectin expression, which plays an important role in glucose and lipid metabolism with antiatherogenic and anti-inflammatory properties. Treatment with naringenin and hesperetin enhanced adiponectin transcription in differentiated 3T3-L1 cells. Both naringenin and hesperetin induced peroxisome proliferator-activated receptor (PPAR)gamma-controlled luciferase expression in a dose-dependent manner (20-160 microM), whereas only naringenin possessed significant activity to activate PPARalpha. These results suggested the two flavonoids might exert antiatherogenic effects partly through activating PPAR and up-regulating adiponectin expression in adipocytes. Our findings give new insight for the molecular explanations for the therapeutic effects of Zhiqiao.


Assuntos
Adiponectina/genética , Citrus/química , Flavanonas/farmacologia , Hesperidina/farmacologia , Transcrição Gênica/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Células 3T3-L1 , Animais , Sequência de Bases , Primers do DNA , Relação Dose-Resposta a Droga , Flavanonas/isolamento & purificação , Hesperidina/isolamento & purificação , Camundongos , PPAR alfa/genética , PPAR gama/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Zhonghua Zhong Liu Za Zhi ; 29(5): 369-72, 2007 May.
Artigo em Zh | MEDLINE | ID: mdl-17892134

RESUMO

OBJECTIVE: To investigate the feasibility of 9Tc(m)-HL91 imaging in prediction of 34 radiotherapy sensitivity of naqsopharyngeal cancer( NPC) and its relationship with prognosis. METHODS: patients with NPC confirmed by pathology, staging from II-IVa, underwent 99Tc(m)-HL91 SPECT imaging one week before radiotherapy. 18 of them received adjuvant chemotherapy. The hypoxia in primary nasopharyngeal lesions and cervical lymph node metastases were calculated semi-quantitatively, and compared with clinical findings in medium-term therapy at 4 months and 1 year post therapy. RESULTS: (1) There was no significant relationship between the total preliminary curative effect of adjuvant chemotherapy and the degree of nasopharyngeal lesion hypoxia (T/Mu, gamma = -0.394, P = 0.145; T/ Ce gamma = -0.510, P = 0.052). But there was a significant difference between the partial curative effect group and significant curative effect group. (2) The degree of NPC regression in the medium-term radiotherapy group was negatively correlated with the degree of hypoxia (T/Mu, gamma = -0.602; T/Ce, gamma = -0.643, P < 0.01). (3) 23 patients had good local control except one case with lung and bone metastasis 4 months post-therapy. The lesions disappeared or not developed in 6 patients (T/Mu 1.30 +/- 0.23, T/Ce 3.61 +/- 0.84). Two patients at stage III and IVa relapsed (T/Mu were 1.40 and 1.27, respectively; T/Ce were 4.10 and 3.85, respectively), there was no significant difference. (4) The degree of lymph node hypoxia had no correlation with the curative effect on medium-term radiotherapy. CONCLUSION: 99 Tc(m)-HL91 hypoxia imaging may predict sensitivity to radiotherapy in patients with NPC, with a potential help to carry out individual therapy. However, further investigation is needed to ascertain whether it could predict the long-time curative effect on NPC radiotherapy.


Assuntos
Neoplasias Nasofaríngeas/diagnóstico por imagem , Compostos de Organotecnécio , Oximas , Adulto , Idoso , Quimioterapia Adjuvante , Feminino , Seguimentos , Humanos , Hipóxia , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Neoplasias Nasofaríngeas/tratamento farmacológico , Neoplasias Nasofaríngeas/radioterapia , Estadiamento de Neoplasias , Aceleradores de Partículas , Cuidados Pré-Operatórios/métodos , Prognóstico , Radioterapia de Alta Energia , Indução de Remissão , Reprodutibilidade dos Testes , Tomografia Computadorizada de Emissão de Fóton Único
16.
PPAR Res ; 2017: 4313561, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29056962

RESUMO

Type 2 diabetes mellitus is often treated with insulin-sensitizing drugs called thiazolidinediones (TZD), which improve insulin resistance and glycemic control. Despite their effectiveness in treating diabetes, these drugs provide little protection from eminent cardiovascular disease associated with diabetes. Here we demonstrate how chiglitazar, a configuration-restricted non-TZD peroxisome proliferator-activated receptor (PPAR) pan agonist with moderate transcription activity, preferentially regulates ANGPTL4 and PDK4, which are involved in glucose and lipid metabolism. CDK5-mediated phosphorylation at serine 273 (S273) is a unique regulatory mechanism reserved for PPARγ, and this event is linked to insulin resistance in type 2 diabetes mellitus. Our data demonstrates that chiglitazar modulates gene expression differently from two TZDs, rosiglitazone and pioglitazone, via its configuration-restricted binding and phosphorylation inhibition of PPARγ. Chiglitazar induced significantly greater expression of ANGPTL4 and PDK4 than rosiglitazone and pioglitazone in different cell models. These increased expressions were dependent on the phosphorylation status of PPARγ at S273. Furthermore, ChIP and AlphaScreen assays showed that phosphorylation at S273 inhibited promoter binding and cofactor recruitment by PPARγ. Based on these results, activities from pan agonist chiglitazar can be an effective part of a long-term therapeutic strategy for treating type 2 diabetes in a more balanced action among its targeted organs.

17.
Endocrinology ; 147(2): 875-84, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16269451

RESUMO

Both peroxisome proliferator-activated receptor (PPAR)-gamma and hormone-sensitive lipase (HSL) play important roles in lipid metabolism and insulin sensitivity. We demonstrate that expression of the HSL gene is up-regulated by PPARgamma and PPARgamma agonists (rosiglitazone and pioglitazone) in the cultured hepatic cells and differentiating preadipocytes. Rosiglitazone treatment also results in up-regulation of the HSL gene in liver and skeleton muscle from an experimental obese rat model, accompanied by the decreased triglyceride content in these tissues. The proximal promoter (-87 bp of the human HSL gene) was found to be essential for PPARgamma-mediated transactivating activity. This important promoter region contains two GC-boxes and binds the transcription factor specificity protein-1 (Sp1) but not PPARgamma. The Sp1-promoter binding activity can be endogenously enhanced by PPARgamma and rosiglitazone, as demonstrated by analysis of EMSA and chromatin immunoprecipitation assay. Mutations in the GC-box sequences reduce the promoter binding activity of Sp1 and the transactivating activity of PPARgamma. In addition, mithramycin A, the specific inhibitor for Sp1-DNA binding activity, abolishes the PPARgamma-mediated up-regulation of HSL. These results indicate that PPARgamma positively regulates the HSL gene expression, and up-regulation of HSL by PPARgamma requires the involvement of Sp1. Taken together, this study suggests that HSL may be a newly identified PPARgamma target gene, and up-regulation of HSL may be an important mechanism involved in action of PPARgamma agonists in type 2 diabetes.


Assuntos
PPAR gama/metabolismo , Regiões Promotoras Genéticas/fisiologia , Fator de Transcrição Sp1/fisiologia , Esterol Esterase/metabolismo , Ativação Transcricional/fisiologia , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Hipoglicemiantes/farmacologia , Fígado/citologia , Masculino , PPAR gama/agonistas , Pioglitazona , Regiões Promotoras Genéticas/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Rosiglitazona , Estatísticas não Paramétricas , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Esterol Esterase/genética , Tiazolidinedionas/farmacologia , Regulação para Cima
18.
Br J Pharmacol ; 148(5): 610-8, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16751799

RESUMO

1. The aim of this study was to investigate the capacity of chiglitazar to improve insulin resistance and dyslipidemia in monosodium L-glutamate (MSG) obese rats and to determine whether its lipid-lowering effect is mediated through its activation of PPARalpha. 2. Chiglitazar is a PPARalpha/gamma dual agonist. 3. The compound improved impaired insulin and glucose tolerance; decreased plasma insulin level and increased the insulin sensitivity index and decreased HOMA index. Euglycemic hyperinsulinemic clamp studies showed chiglitazar increased the glucose infusion rate in MSG obese rats. 4. Chiglitazar inhibited alanine gluconeogenesis, lowered the hepatic glycogen level in MSG obese rats. Like rosiglitazone, chiglitazar promoted the differentiation of adipocytes and decreased the maximal diameter of adipocytes. In addition, chiglitazar decreased the fibrosis and lipid accumulation in the islets and increased the size of islets. 5. Chiglitazar reduced plasma triglyceride, total cholesterol (TCHO), nonesterified fatty acids (NEFA) and low density lipoprotein-cholesterol levels; lowered hepatic triglyceride and TCHO contents; decreased muscular NEFA level. Unlike rosiglitazone, chiglitazar showed significant increase of mRNA expression of PPARalpha, CPT1, BIFEZ, ACO and CYP4A10 in the liver of MSG obese rats. 6. These data suggest that PPARalpha/gamma coagonist, such as chiglitazar, affect lipid homeostasis with different mechanisms from rosiglitazone, chiglitazar may have better effects on lipid homeostasis in diabetic patients than selective PPARgamma agonists.


Assuntos
Carbazóis/farmacologia , Dislipidemias/induzido quimicamente , Resistência à Insulina , Obesidade/induzido quimicamente , PPAR alfa/agonistas , PPAR gama/agonistas , Propionatos/farmacologia , Glutamato de Sódio , Tecido Adiposo/citologia , Tecido Adiposo/efeitos dos fármacos , Animais , Glicemia/efeitos dos fármacos , Modelos Animais de Doenças , Dislipidemias/tratamento farmacológico , Expressão Gênica/efeitos dos fármacos , Gluconeogênese/efeitos dos fármacos , Hipoglicemiantes/uso terapêutico , Lipídeos/análise , Lipídeos/sangue , Fígado/química , Fígado/efeitos dos fármacos , Glicogênio Hepático/análise , Músculos/química , Músculos/efeitos dos fármacos , Pâncreas/citologia , Pâncreas/efeitos dos fármacos , Pioglitazona , Pirimidinas/farmacologia , Ratos , Ratos Wistar , Rosiglitazona , Tiazolidinedionas/farmacologia
19.
Intractable Rare Dis Res ; 5(3): 185-91, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27672541

RESUMO

Peripheral T-cell lymphoma (PTCL) is a set of rare and highly heterogeneous group of mature T- and NK-cell neoplasms associated with poor outcomes and lack of standard and effective therapies. The total number of newly diagnosed cases of PTCL yearly in China is estimated about 50,000. Chidamide (CS055) is a novel and orally active benzamide class of histone deacetylase (HDAC) inhibitor that selectively inhibits activity of HDAC1, 2, 3 and 10, the enzymes that are involved and play an important role in tumor initiation and development in both tumor cells and their surrounding micro-environment. Functioning as a genuine epigenetic modulator, chidamide induces growth arrest and apoptosis in tumor cells and enhances cellular antitumor immunity. Based on the overall results from preclinical and phase I clinical studies, exploratory and pivotal phase II trials of chidamide for relapsed or refractory PTCL were conducted from March 2009 to May 2012, and the results led to CFDA approval of chidamide for the indication in December 2014, being the first approved orphan drug according to the research & development approach of orphan drugs in China, as well as the first orally active drug for PTCL in China and worldwide.

20.
J Radiat Res ; 57(6): 702-708, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27618833

RESUMO

The aims of the present study were to analyze the outcomes of pregnancy, after 131I treatment, in patients of reproductive age with Graves' hyperthyroidism and to investigate the effects, if any, of the 131I treatment on the mothers and newborns. From 2009 to 2014, 257 pregnant female patients with Graves' hyperthyroidism in the outpatients at the Department of Nuclear Medicine and 166 healthy pregnant women from the Department of Obstetrics at Sun Yat-Sen Memorial Hospital were included in our study. They were divided into a 131I therapy group (n = 130) and an anti-thyroid drug (ATD) group (n = 127) according to their therapy before conception. The neonatal gender, rate of preterm birth, body weight ratio and occurrence of low birth weight [except for higher rates of abortion (odds ratio; OR = 2.023) and cesarean delivery (OR = 1.552) in patients with Graves' hyperthyroidism] showed no statistically significant differences from those of the healthy group (P > 0.05). The level of intrauterine growth restriction did not differ between the Graves' hyperthyroidism group and the healthy group (8 vs 2, 3.0% vs 1.2%). The outcomes of pregnancy among the 131I therapy group, ATD group and healthy group also showed no significant differences. Of the patients treated with 131I, no significant differences were observed in the outcomes of their pregnancies, whether they received propylthiouracil (PTU), levothyroxine or no additional drug treatment during pregnancy. Women with hyperthyroidism who were treated with 131I therapy could have normal delivery if they ceased 131I treatment for at least six months prior to conception and if their thyroid function was reasonably controlled and maintained using the medication: anti-thyroid drug and levothyroxine before and during pregnancy.


Assuntos
Doença de Graves/terapia , Radioisótopos do Iodo/uso terapêutico , Complicações na Gravidez/terapia , Adulto , Peso ao Nascer , Peso Corporal , Cesárea , Feminino , Seguimentos , Humanos , Recém-Nascido , Masculino , Razão de Chances , Gravidez , Resultado da Gravidez , Nascimento Prematuro
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA