Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Clin Cancer Res ; 30(16): 3364-3370, 2024 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-38875108

RESUMO

On November 15, 2023, the U.S. Food and Drug Administration (FDA) granted traditional approval to repotrectinib (Augtyro, Bristol Myers Squibb Corporation) for the treatment of adult patients with locally advanced or metastatic receptor tyrosine kinase encoded by the ROS1 gene (ROS1)-positive non-small cell lung cancer (NSCLC). The approval was based on TRIDENT-1, a single-arm trial with multiple cohorts of patients with ROS1 fusion-positive (hereafter "ROS1-positive") NSCLC (NCT03093116), who were either treatment naïve or had received prior ROS1 tyrosine kinase inhibitor (TKI) and/or platinum-based chemotherapy. The primary efficacy outcome measure is objective response rate (ORR) assessed by blinded independent central review (BICR) using response evaluation criteria in solid tumors version 1.1. ORR was assessed in 71 patients who were ROS1 TKI naïve and 56 patients who had received a prior ROS1 TKI. Among the 71 patients who were ROS1 TKI naïve, the ORR was 79% (95% CI, 68-88), median duration of response was 34.1 months (95% CI, 26-NE). In patients who had received a prior ROS1 TKI and no prior chemotherapy, the ORR was 38% (95% CI, 25-52). The median duration of response was 14.8 months (95% CI, 7.6-NE); BICR-assessed responses were observed in CNS metastases in patients in both cohorts and in patients who developed resistance mutations following prior TKI therapy. The most common (>20%) adverse reactions were dizziness, dysgeusia, peripheral neuropathy, constipation, dyspnea, ataxia, fatigue, cognitive disorders, and muscular weakness. A unique feature of this ROS1 TKI approval is the inclusion of robust evidence of efficacy in patients with ROS1-positive NSCLC who had progressed on prior ROS1 TKIs.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Aprovação de Drogas , Neoplasias Pulmonares , Proteínas Tirosina Quinases , Proteínas Proto-Oncogênicas , United States Food and Drug Administration , Humanos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Tirosina Quinases/antagonistas & inibidores , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/genética , Estados Unidos , Masculino , Feminino , Pessoa de Meia-Idade , Inibidores de Proteínas Quinases/uso terapêutico , Inibidores de Proteínas Quinases/efeitos adversos , Idoso , Pirimidinas/uso terapêutico , Adulto , Pirazóis/uso terapêutico , Idoso de 80 Anos ou mais
2.
J Clin Oncol ; 42(10): 1193-1201, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38381994

RESUMO

PURPOSE: The US Food and Drug Administration (FDA) approved elacestrant for the treatment of postmenopausal women or adult men with estrogen receptor-positive (ER+), human epidermal growth factor receptor 2-negative (HER2-), estrogen receptor 1 (ESR1)-mutated advanced or metastatic breast cancer with disease progression after at least one line of endocrine therapy (ET). PATIENTS AND METHODS: Approval was based on EMERALD (Study RAD1901-308), a randomized, open-label, active-controlled, multicenter trial in 478 patients with ER+, HER2- advanced or metastatic breast cancer, including 228 patients with ESR1 mutations. Patients were randomly assigned (1:1) to receive either elacestrant 345 mg orally once daily (n = 239) or investigator's choice of ET (n = 239). RESULTS: In the ESR1-mut subgroup, EMERALD demonstrated a statistically significant improvement in progression-free survival (PFS) by blinded independent central review assessment (n = 228; hazard ratio [HR], 0.55 [95% CI, 0.39 to 0.77]; P value = .0005). Although the overall survival (OS) end point was not met, there was no trend toward a potential OS detriment (HR, 0.90 [95% CI, 0.63 to 1.30]) in the ESR1-mut subgroup. PFS also reached statistical significance in the intention-to-treat population (ITT, N = 478; HR, 0.70 [95% CI, 0.55 to 0.88]; P value = .0018). However, improvement in PFS in the ITT population was primarily attributed to results from patients in the ESR1-mut subgroup. More patients who received elacestrant experienced nausea, vomiting, and dyslipidemia. CONCLUSION: The approval of elacestrant in ER+, HER2- advanced or metastatic breast cancer was restricted to patients with ESR1 mutations. Benefit-risk assessment in the ESR1-mut subgroup was favorable on the basis of a statistically significant improvement in PFS in the context of an acceptable safety profile including no evidence of a potential detriment in OS. By contrast, the benefit-risk assessment in patients without ESR1 mutations was not favorable. Elacestrant is the first oral estrogen receptor antagonist to receive FDA approval for patients with ESR1 mutations.


Assuntos
Neoplasias da Mama , Tetra-Hidronaftalenos , Adulto , Estados Unidos , Humanos , Feminino , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Receptor alfa de Estrogênio/genética , United States Food and Drug Administration , Receptor ErbB-2/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico
3.
Clin Cancer Res ; 29(20): 4027-4031, 2023 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-37289037

RESUMO

On September 30, 2022, the FDA granted accelerated approval to futibatinib for the treatment of adult patients with previously treated, unresectable, locally advanced or metastatic intrahepatic cholangiocarcinoma (iCCA) with FGFR2 fusions or other rearrangements. Approval was based on Study TAS-120-101, a multicenter open-label, single-arm trial. Patients received futibatinib 20-mg orally once daily. The major efficacy outcome measures were overall response rate (ORR) and duration of response (DoR) as determined by an independent review committee (IRC) according to RECIST v1.1. ORR was 42% (95% confidence interval, 32%-52%). Median DoR was 9.7 months. Adverse reactions occurring in ≥30% patients were nail toxicity, musculoskeletal pain, constipation, diarrhea, fatigue, dry mouth, alopecia, stomatitis, and abdominal pain. The most common laboratory abnormalities (≥50%) were increased phosphate, increased creatinine, decreased hemoglobin, and increased glucose. Ocular toxicity (including dry eye, keratitis, and retinal epithelial detachment) and hyperphosphatemia are important risks of futibatinib, which are listed under Warnings and Precautions. This article summarizes the FDA's thought process and data supporting the approval of futibatinib.


Assuntos
Neoplasias dos Ductos Biliares , Colangiocarcinoma , Pirazóis , Pirróis , Adulto , Humanos , Pirimidinas/efeitos adversos , Colangiocarcinoma/tratamento farmacológico , Neoplasias dos Ductos Biliares/tratamento farmacológico , Neoplasias dos Ductos Biliares/genética , Ductos Biliares Intra-Hepáticos , Aprovação de Drogas , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética
4.
Clin Pharmacol Ther ; 111(3): 624-634, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34656075

RESUMO

Remdesivir (RDV) is the first drug approved by the US Food and Drug Administration (FDA) for the treatment of coronavirus disease 2019 (COVID-19) in certain patients requiring hospitalization. As a nucleoside analogue prodrug, RDV undergoes intracellular multistep activation to form its pharmacologically active species, GS-443902, which is not detectable in the plasma. A question arises that whether the observed plasma exposure of RDV and its metabolites would correlate with or be informative about the exposure of GS-443902 in tissues. A whole body physiologically-based pharmacokinetic (PBPK) modeling and simulation approach was utilized to elucidate the disposition mechanism of RDV and its metabolites in the lungs and liver and explore the relationship between plasma and tissue pharmacokinetics (PK) of RDV and its metabolites in healthy subjects. In addition, the potential alteration of plasma and tissue PK of RDV and its metabolites in patients with organ dysfunction was explored. Our simulation results indicated that intracellular exposure of GS-443902 was decreased in the liver and increased in the lungs in subjects with hepatic impairment relative to the subjects with normal liver function. In subjects with severe renal impairment, the exposure of GS-443902 in the liver was slightly increased, whereas the lung exposure of GS-443902 was not impacted. These predictions along with the organ impairment study results may be used to support decision making regarding the RDV dosage adjustment in these patient subgroups. The modeling exercise illustrated the potential of whole body PBPK modeling to aid in decision making for nucleotide analogue prodrugs, particularly when the active metabolite exposure in the target tissues is not available.


Assuntos
Monofosfato de Adenosina/análogos & derivados , Alanina/análogos & derivados , Fígado/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Modelos Biológicos , Insuficiência de Múltiplos Órgãos/metabolismo , Monofosfato de Adenosina/sangue , Monofosfato de Adenosina/metabolismo , Monofosfato de Adenosina/farmacocinética , Monofosfato de Adenosina/urina , Adulto , Alanina/sangue , Alanina/metabolismo , Alanina/farmacocinética , Alanina/urina , Humanos , Fígado/metabolismo , Pulmão/metabolismo , Masculino , Insuficiência de Múltiplos Órgãos/tratamento farmacológico , Distribuição Tecidual
5.
Cell Death Dis ; 12(6): 508, 2021 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-34006831

RESUMO

There is limited and discordant evidence on the role of nicotine in diabetic vascular disease. Exacerbated endothelial cell dysregulation in smokers with diabetes is associated with the disrupted adipose function. Adipokines possess vascular protective, anti-inflammatory, and anti-diabetic properties. However, whether and how nicotine primes and aggravates diabetic vascular disorders remain uncertain. In this study, we evaluated the alteration of adiponectin (APN) level in high-fat diet (HFD) mice with nicotine (NIC) administration. The vascular pathophysiological response was evaluated with vascular ring assay. Confocal and co-immunoprecipitation analysis were applied to identify the signal interaction and transduction. These results indicated that the circulating APN level in nicotine-administrated diabetic Apolipoprotein E-deficient (ApoE-/-) mice was elevated in advance of 2 weeks of diabetic ApoE-/- mice. NIC and NIC addition in HFD groups (NIC + HFD) reduced the vascular relaxation and signaling response to APN at 6 weeks. Mechanistically, APN receptor 1 (AdipoR1) level was decreased in NIC and further significantly reduced in NIC + HFD group at 6 weeks, while elevated suppressor of cytokine signaling 3 (SOCS3) expression was induced by NIC and further augmented in NIC + HFD group. Additionally, nicotine provoked SOCS3, degraded AdipoR1, and attenuated APN-activated ERK1/2 in the presence of high glucose and high lipid (HG/HL) in human umbilical vein endothelial cells (HUVECs). MG132 (proteasome inhibitor) administration manifested that AdipoR1 was ubiquitinated, while inhibited SOCS3 rescued the reduced AdipoR1. In summary, this study demonstrated for the first time that nicotine primed vascular APN resistance via SOCS3-mediated degradation of ubiquitinated AdipoR1, accelerating diabetic endothelial dysfunction. This discovery provides a potential therapeutic target for preventing nicotine-accelerated diabetic vascular dysfunction.


Assuntos
Adiponectina/metabolismo , Apolipoproteínas E/metabolismo , Nicotina/efeitos adversos , Animais , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Knockout , Transfecção , Ubiquitina
6.
Clin Cancer Res ; 27(15): 4142-4146, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-33712511

RESUMO

On April 10, 2020, the FDA approved selumetinib (KOSELUGO, AstraZeneca) for the treatment of pediatric patients 2 years of age and older with neurofibromatosis type 1 who have symptomatic, inoperable plexiform neurofibromas. Approval was based on demonstration of a durable overall response rate per Response Evaluation in Neurofibromatosis and Schwannomatosis criteria and supported by observed clinical improvements in plexiform neurofibroma-related symptoms and functional impairments in 50 pediatric patients with inoperable plexiform neurofibromas in a single-arm, multicenter trial. The overall reponse rate per NCI investigator assessment was 66% (95% confidence interval, 51-79) with at least 12 months of follow-up. The median duration of response was not reached, and 82% of responding patients experienced duration of response ≥12 months. Clinical outcome assessment endpoints provided supportive efficacy data. Risks of selumetinib are consistent with MAPK (MEK) inhibitor class effects, including ocular, cardiac, musculoskeletal, gastrointestinal, and dermatologic toxicities. Safety was assessed across a pooled database of 74 pediatric patients with plexiform neurofibromas and supported by adult and pediatric selumetinib clinical trial data in cancer indications. The benefit-risk assessment for selumetinib in patients with inoperable plexiform neurofibromas was considered favorable.


Assuntos
Benzimidazóis/uso terapêutico , Aprovação de Drogas , Neurofibroma Plexiforme/tratamento farmacológico , Adolescente , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Estados Unidos
7.
Clin Cancer Res ; 27(5): 1220-1226, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33055172

RESUMO

On April 17, 2020, the FDA approved tucatinib in combination with trastuzumab and capecitabine for the treatment of patients with advanced unresectable or metastatic HER2-positive breast cancer, including patients with brain metastases, who have received one or more prior anti-HER2-based regimens in the metastatic setting. This was the first new molecular entity evaluated under Project Orbis, an FDA Oncology Center of Excellence initiative, which supports concurrent review of oncology drugs by multiple global health authorities. Approval was based on the HER2CLIMB trial, which randomized patients to receive tucatinib or placebo with trastuzumab and capecitabine. Tucatinib demonstrated efficacy compared with placebo in progression-free survival [PFS; HR: 0.54; 95% confidence interval (CI): 0.42-0.71; P < 0.00001] and overall survival (OS; HR: 0.66; 95% CI, 0.50-0.87; P = 0.00480). Patients with either treated and stable or active brain metastases made up 48% of the study population. PFS in patients with brain metastases confirmed benefit (HR: 0.48; 95% CI, 0.34-0.69; P < 0.00001). The benefit in patients with brain metastases allowed for inclusion of this specific population in the indication. Important safety signals included diarrhea and hepatotoxicity which are listed under Warnings and Precautions. This article summarizes the FDA thought process and data supporting the favorable benefit-risk profile and approval of tucatinib.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Aprovação de Drogas , Oxazóis/uso terapêutico , Piridinas/uso terapêutico , Quinazolinas/uso terapêutico , Receptor ErbB-2/metabolismo , Antineoplásicos/uso terapêutico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/secundário , Feminino , Humanos , Estados Unidos , United States Food and Drug Administration
8.
Clin Cancer Res ; 27(7): 1842-1849, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33168657

RESUMO

On May 24, 2019, the FDA granted regular approval to alpelisib in combination with fulvestrant for postmenopausal women, and men, with hormone receptor (HR)-positive, HER2-negative, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA)-mutated, advanced or metastatic breast cancer as detected by an FDA-approved test following progression on or after an endocrine-based regimen. Approval was based on the SOLAR-1 study, a randomized, double-blind, placebo-controlled trial of alpelisib plus fulvestrant versus placebo plus fulvestrant. The primary endpoint was investigator-assessed progression-free survival (PFS) per RECIST v1.1 in the cohort of trial participants whose tumors had a PIK3CA mutation. The estimated median PFS by investigator assessment in the alpelisib plus fulvestrant arm was 11 months [95% confidence interval (CI), 7.5-14.5] compared with 5.7 months (95% CI, 3.7-7.4) in the placebo plus fulvestrant arm (HR, 0.65; 95% CI, 0.50-0.85; two-sided P = 0.001). The median overall survival was not yet reached for the alpelisib plus fulvestrant arm (95% CI, 28.1-NE) and was 26.9 months (95% CI, 21.9-NE) for the fulvestrant control arm. No PFS benefit was observed in trial participants whose tumors did not have a PIK3CA mutation (HR, 0.85; 95% CI, 0.58-1.25). The most common adverse reactions, including laboratory abnormalities, on the alpelisib plus fulvestrant arm were increased glucose, increased creatinine, diarrhea, rash, decreased lymphocyte count, increased gamma glutamyl transferase, nausea, increased alanine aminotransferase, fatigue, decreased hemoglobin, increased lipase, decreased appetite, stomatitis, vomiting, decreased weight, decreased calcium, decreased glucose, prolonged activated partial thromboplastin time, and alopecia.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Classe I de Fosfatidilinositol 3-Quinases/genética , Fulvestranto/administração & dosagem , Mutação , Tiazóis/administração & dosagem , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias da Mama/genética , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Método Duplo-Cego , Aprovação de Drogas , Feminino , Fulvestranto/efeitos adversos , Fulvestranto/farmacologia , Humanos , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Medidas de Resultados Relatados pelo Paciente , Receptor ErbB-2/análise , Receptores de Estrogênio/análise , Tiazóis/efeitos adversos , Tiazóis/farmacologia
9.
Eur J Pharm Biopharm ; 142: 153-164, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31226367

RESUMO

The distribution of nanomedicines inside solid tumors is often restricted to perivascular areas, leaving most distal tumor cells out of reach. This partly explains modest patient benefit of many nanomedicines compared to their free-form counterparts. The objective for this study is to develop a mathematical model to quantitatively analyze this phenomenon and the influencing factors to such perivascular distribution and seek for effective strategies to alleviate this. A spatial tumor distribution model was firstly constructed to mimic the geometrical structure of tumor vessels and the surrounding tumor cells. This tumor model was further integrated with a systemic pharmacokinetics model for nanoparticles. A variety of factors on the tumor spatial distributions of nanomedicines were considered in the model. With the model, we quantified the effect of these influencing factors on tumor delivery efficacy (ID %), the magnitude of heterogeneous distribution (H index), and the effect of enhanced permeability and retention (EPR). In particularly, we compared the spatial distributions of the nanoparticles and the free payloads insides tumors. The model predicted high degrees of distributional heterogeneity for both nanoparticles and free payloads. The degree of heterogeneity and the influencing factors for free payloads were markedly different from those for nanoparticles. We found that nanoparticle diffusion coefficient was the most effective factor in reducing the nanoparticle H index but exerted moderate influence on the free payloads H index. The most effective factor in reducing the H index of free payload was payload diffusion coefficient. The factors that improved free payload distribution were closely associated with higher drug efficacy. In contrast, the factors that improved nanoparticle spatial distributions did not always confer improved anti-tumor efficacy of the delivered drug. These findings highlight the importance of assessing the heterogeneous free payload distribution in tumors for the development of effective nanomedicines.


Assuntos
Antineoplásicos/metabolismo , Antineoplásicos/uso terapêutico , Nanopartículas/metabolismo , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Humanos , Modelos Teóricos , Nanomedicina/métodos , Permeabilidade/efeitos dos fármacos , Distribuição Tecidual/efeitos dos fármacos
10.
Pharm Res ; 35(9): 174, 2018 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-29987398

RESUMO

PURPOSE: The mechanisms underlying doxorubicin cytotoxicity and cardiotoxicity were broadly explored but remain incompletely understood. A multiscale physiologically-based pharmacokinetic (PBPK) model was developed to assess doxorubicin dispositions at levels of system, tissue interstitial, cell, and cellular organelles. This model was adopted to explore the mechanisms-of-action/toxicity of doxorubicin in humans. METHODS: The PBPK model was developed by analyzing data from mice and the model was verified by scaling up to predict doxorubicin multiscale dispositions in rats and humans. The multiscale dispositions of doxorubicin in human heart and tumors were explicitly simulated to elucidate the potential mechanisms of its cytotoxicity and cardiotoxicity. RESULTS: The developed PBPK model was able to adequately describe doxorubicin dispositions in mice, rats and humans. In humans, prolonged infusion, a dosing regimen with less cardiotoxicity, was predicted with substantially reduced free doxorubicin concentrations at human heart interstitium, which were lower than the concentrations associated with oxidative stress. However, prolonged infusion did not reduce doxorubicin-DNA adduct at tumor nucleus, consistent with clinical observations that prolonged infusion did not compromise anti-tumor effect, indicating that one primary anti-tumor mechanism was DNA torsion. CONCLUSIONS: A multiscale PBPK model for doxorubicin was developed and further applied to explore its cytotoxic and cardiotoxic mechanisms.


Assuntos
Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/toxicidade , Cardiotoxicidade/etiologia , Doxorrubicina/farmacocinética , Doxorrubicina/toxicidade , Animais , Simulação por Computador , Citotoxinas/farmacocinética , Citotoxinas/toxicidade , Humanos , Modelos Biológicos , Neoplasias/tratamento farmacológico , Ratos , Distribuição Tecidual
11.
J Cell Physiol ; 220(1): 72-81, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19229878

RESUMO

Hepatic progenitor cells are local stem cells in the liver and they can be differentiated into either hepatocytes or cholangiocytes depending on different stimulations. These stimulations include extracellular growth factors and intracellular transcription factors. Bone morphogenetic protein 4 (BMP4) is a member of transforming growth factor beta (TGF-beta) superfamily and was first identified as growth factor to induce ectopic bone formation from skeletal muscle. Role of BMP4 in the liver is still unclear especially its role in hepatic progenitor cells (HPCs) differentiation. BMP4 was used to stimulate rat HPCs (WB-F344 cells) and differentiation of WB-F344 cells was investigated by reverse transcriptase polymerase chain reaction (RT-PCR) and Western blot analysis. Both adenovirus delivered BMP4 and recombinant BMP4 were able to induce expression of hepatocyte markers such as albumin, TAT-1, and G6Pase but not cholangiocyte markers such as beta4-integrin and CK19. BMP4 induced differentiation of WB-F344 cells toward hepatocytes was mediated by increase in phosphorylation of Smad1 and ERK1/2. Moreover, BMP4 also stimulated expression of transcription factor--C/EBP-alpha, which involved in differentiation of WB-F344 cells toward hepatocytes. BMP4 is able to stimulate WB-F344 cells differentiation toward hepatocyte lineage.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Diferenciação Celular , Linhagem da Célula , Hepatócitos/metabolismo , Células-Tronco/metabolismo , Albuminas/metabolismo , Animais , Biomarcadores/metabolismo , Proteína Morfogenética Óssea 4/genética , Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Linhagem Celular , Glucose-6-Fosfatase/metabolismo , Hepatócitos/enzimologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Ratos , Proteínas Recombinantes/metabolismo , Proteína Smad1/metabolismo , Células-Tronco/enzimologia , Fatores de Tempo , Transfecção , Tirosina Transaminase/metabolismo
12.
J Proteome Res ; 6(7): 2495-501, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17542627

RESUMO

BACKGROUND: Understanding the proteins associated with lymph node metastasis (LNM) in colorectal cancer (CRC) will benefit us in the prediction of CRC prognosis and provide us new potential targets in the intervention of CRC. The aim of this study is to investigate the LNM-associated proteins and to evaluate the clinicopathological characteristics of these target proteins' expression in CRC. METHODS: Fresh tumor and paired normal mucosa from five cases for each group of non-LNM CRC and LNM CRC were analyzed by two-dimensional electrophoresis coupled with MALDI-TOF-MS, followed by Western blotting confirmation. In 40 paraffin-embedded CRC samples, each for non-LNM CRC and LNM CRC, four differentially expressed proteins identified by proteomics analysis were detected by tissue microarray with immunohistochemistry staining to access the clinicopathological characteristics of these proteins in LNM of CRC. RESULTS: Twenty-five proteins were found to be differentially expressed between normal mucosa and CRC tissue. Increased expression levels of heat shock protein-27 (HSP-27), glutathione S-transferase (GST), and Annexin II, but a decreased expression level of liver-fatty acid binding protein (L-FABP), existed in LNM CRC as compared with non-LNM CRC (p<0.01 or p<0.05, respectively). CONCLUSION: The techniques of proteomic analysis combined with tissue microarray provide us a dramatic tool for screening of LNM-associated proteins in cancer research. The increased expression of HSP-27, GST, and Annexin II, but decreased expression of L-FABP, suggests a significantly elevated incidence of LNM in CRC.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias Colorretais/patologia , Proteínas de Neoplasias/análise , Análise Serial de Proteínas/métodos , Proteoma/análise , Proteômica/métodos , Adulto , Idoso , Western Blotting , Eletroforese em Gel Bidimensional , Feminino , Humanos , Imuno-Histoquímica , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Análise Serial de Tecidos/métodos
13.
Biochem Cell Biol ; 85(2): 189-95, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17534399

RESUMO

Treatment of liver fibrosis and cirrhosis remains a challenging field. Hepatocyte injury and the activation of hepatic stellate cells are the 2 major events in the development of liver fibrosis and cirrhosis. It is known that several Chinese herbs have significant beneficial effects on the liver; therefore, the purpose of the present study was to investigate the therapeutic effect of saikosaponin-d (SSd) on liver fibrosis and cirrhosis. A rat model of liver fibrosis was established using the dimethylnitrosamine method. Liver tissue and serum were used to examine the effect of SSd on liver fibrosis. A hepatocyte culture was also used to investigate how SSd can protect hepatocytes from oxidative injury induced by carbon tetrachloride. The results showed that SSd significantly reduced collagen I deposition in the liver and alanine aminotransferase level in the serum. Moreover, SSd decreased the content of TGF-beta1 in the liver, which was significantly elevated after dimethylnitrosamine induced liver fibrosis. Furthermore, SSd was able to alleviate hepatocyte injury from oxidative stress. In conclusion, SSd could postpone the development of liver fibrosis by attenuating hepatocyte injury.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Hepatócitos/metabolismo , Cirrose Hepática Experimental/prevenção & controle , Ácido Oleanólico/análogos & derivados , Saponinas/farmacologia , Alanina Transaminase/sangue , Animais , Linhagem Celular Tumoral , Colágeno Tipo I/biossíntese , Dimetilnitrosamina/toxicidade , Hepatócitos/patologia , Cirrose Hepática Experimental/sangue , Cirrose Hepática Experimental/induzido quimicamente , Cirrose Hepática Experimental/patologia , Ácido Oleanólico/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Fator de Crescimento Transformador beta1/metabolismo
14.
J Cell Physiol ; 212(3): 764-70, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17525996

RESUMO

Smad1 is a receptor-activated intracellular signaling protein, which mediates signal transduction of bone morphogenetic proteins. Current study investigated the expression and transcriptional activity of Smad1 during hepatic stellate cell (HSC) activation. Rat HSCs were isolated from rats at 1, 2, 3 and 4 days after gavaged with carbon tetrachloride (CCl(4)) or corn oil. RT-PCR, Western blot, gel-shift assay and luciferase assay were employed to examine Smad1 expression and transcriptional activity, respectively. CCl(4)-cirrhotic liver fat-storing cells-8B (CFSC-8B) cells were infected with recombinant adenoviruses of Smad1 and/or Smad1 shRNA. Both mRNA and protein levels of Smad1 were significantly increased at 48 h after gavage of CCl(4). Gel shift assays demonstrated a significant increase in nuclear Smad1 in day 9 HSCs. Transfection of HSCs with Smad1 responsible luciferase indicated an increase in Smad1 transcriptional activity in day 6 HSCs (1.563 +/- 0.229 in day 6 versus 0.785 +/- 0.192 in day 3). When CFSC-8B cells were infected with adenoviruses with Smad1 or Smad1 short hairpin RNA (shRNA), there was an increase or decrease in Smad1 mRNA and protein, respectively. Smooth muscle alpha-actin expression was increased or decreased according to induction or reduction of Smad1. In conclusion, there were significantly increases in Smad1 expression and transcriptional activity during in vivo activation of hepatic stellate cells.


Assuntos
Diferenciação Celular , Proliferação de Células , Cirrose Hepática/metabolismo , Fígado/metabolismo , Proteína Smad1/metabolismo , Transcrição Gênica , Actinas/metabolismo , Adenoviridae/genética , Animais , Tetracloreto de Carbono , Diferenciação Celular/genética , Linhagem Celular , Modelos Animais de Doenças , Vetores Genéticos , Fígado/patologia , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/genética , Cirrose Hepática/patologia , Masculino , Fenótipo , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Proteína Smad1/genética , Fatores de Tempo , Transfecção , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA