Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Colloids Surf B Biointerfaces ; 236: 113823, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38442502

RESUMO

Hydrophobic antimicrobial peptide L30, a potential antibiotic candidate, has poor water solubility and hemolytic activity. Herein, a biocompatible nano-formulation composed of liposomes and dendritic mesoporous silica encapsulation (LDMSNs@L30) was constructed for L30 to solve the limits for its clinical development. The characterization, antimicrobial activity and therapeutic effect of LDMSNs@L30 on Staphylococcus aureus 9 (cfr+) infected mice models were investigated. LDMSNs@L30 displayed a smooth, spherical, and monodisperse nanoparticle with a hydrodynamic diameter of 177.40 nm, an encapsulation rate of 56.13%, a loading efficiency of 32.26%, a release rate of 66.5%, and effective slow-release of L30. Compared with free L30, the formulation could significantly increase the solubility of L30 in PBS with the maximum concentration from 8 µg/mL to 2.25 mg/mL and decrease the hemolytic activity of hydrophobic peptide L30 with the HC5 from 65.36 µg/mL to more than 500 µg/mL. The nano delivery system LDMSNs@L30 also exhibited higher therapeutic effects on mice models infected with S. aureus 9 (cfr+) than those of free L30 after 7 days of treatment by reducing the lung inflammation and the inflammatory cytokines levels in plasma, showing better health score and pulmonary pathological improvement. Our research suggests that nano-formulation can be expected to be a promising strategy for peptide drugs in therapeutic applications.


Assuntos
Infecções Estafilocócicas , Staphylococcus aureus , Animais , Camundongos , Peptídeos Antimicrobianos , Antibacterianos/química , Infecções Estafilocócicas/tratamento farmacológico , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Nanotecnologia
2.
Nat Med ; 28(6): 1189-1198, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35534566

RESUMO

Despite success in hematologic malignancies, the treatment landscape of chimeric antigen receptor (CAR) T cell therapy for solid tumors remains limited. Claudin18.2 (CLDN18.2)-redirected CAR T cells showed promising efficacy against gastric cancer (GC) in a preclinical study. Here we report the interim analysis results of an ongoing, open-label, single-arm, phase 1 clinical trial of CLDN18.2-targeted CAR T cells (CT041) in patients with previously treated, CLDN18.2-positive digestive system cancers ( NCT03874897 ). The primary objective was safety after CT041 infusion; secondary objectives included CT041 efficacy, pharmacokinetics and immunogenicity. We treated 37 patients with one of three CT041 doses: 2.5 × 108, 3.75 × 108 or 5.0 × 108 cells. All patients experienced a grade 3 or higher hematologic toxicity. Grade 1 or 2 cytokine release syndrome (CRS) occurred in 94.6% of patients. No grade 3 or higher CRS or neurotoxicities, treatment-related deaths or dose-limiting toxicities were reported. The overall response rate (ORR) and disease control rate (DCR) reached 48.6% and 73.0%, respectively. The 6-month duration of response rate was 44.8%. In patients with GC, the ORR and DCR reached 57.1% and 75.0%, respectively, and the 6-month overall survival rate was 81.2%. These initial results suggest that CT041 has promising efficacy with an acceptable safety profile in patients with heavily pretreated, CLDN18.2-positive digestive system cancers, particularly in those with GC.


Assuntos
Imunoterapia Adotiva , Neoplasias Gástricas , Claudinas , Síndrome da Liberação de Citocina , Humanos , Imunoterapia Adotiva/efeitos adversos , Imunoterapia Adotiva/métodos , Neoplasias Gástricas/terapia , Linfócitos T
3.
PLoS One ; 16(5): e0250573, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33961634

RESUMO

OBJECTIVE: Dyslipidemia is a leading risk factor for cardiovascular and cerebrovascular diseases. By collecting the blood lipid profiles among adult residents of Shenmu City in Shaanxi Province, China, we aim to assess and elucidate the prevalence and risk factors of dyslipidemia in this city. METHOD: Stratified multistage sampling was used to survey 4,598 permanent adult residents in five areas of Shenmu (2 communities in the county seat, 2 in the southern area and 2 in the northern area) from September 2019 to December 2019. Questionnaire surveys and physical examinations were conducted. Data were analyzed using SPSS software version 26.0. RESULTS: The average level of total cholesterol (TC) is 4.47mmol/L, that of triglyceride (TG) 1.32mmol/L, high-density lipoprotein cholesterol (HDL-C) 1.27mmol/L, apolipoprotein A1 (ApoA1) 1.44g/L, low-density lipoprotein cholesterol (LDL-C) 2.7mmol/L and apolipoprotein B (ApoB) 0.97g/L. The prevalence of hypercholesterolemia (HTC), hypertriglyceridemia (HTG), low high-density lipoprotein (HDL-C) and high low-density lipoprotein (LDL-C) is 22.4%, 33.3%, 14.5%, and 5.81%, respectively, and the overall prevalence of dyslipidemia is 48.27%. Furthermore, blood lipid levels and prevalence of dyslipidemia vary by region, age, gender, occupation and educational level. Nine risk factors of dyslipidemia were identified, which are living in county seat or northern industrial area, increasing age, male, overweight or obesity, abdominal obesity, smoking, hypertension, abnormal glucose metabolism (pre-diabetes or diabetes) and hyperuricemia. CONCLUSION: The blood lipid levels and dyslipidemia prevalence of adults in Shenmu City are higher comparing to national averages of China. Combining risk factors of dyslipidemia, early detection and public health interventions are necessary in high-risk population for associated cardiovascular and cerebrovascular diseases prevention.


Assuntos
Diabetes Mellitus/fisiopatologia , Dislipidemias/epidemiologia , Hipertensão/fisiopatologia , Obesidade/fisiopatologia , Sobrepeso/fisiopatologia , Adolescente , Adulto , China/epidemiologia , Dislipidemias/sangue , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Prevalência , Fatores de Risco , Inquéritos e Questionários , Adulto Jovem
4.
Clin Cancer Res ; 26(15): 3979-3989, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32371538

RESUMO

PURPOSE: Our preclinical studies demonstrated the potential of chimeric antigen receptor (CAR)-glypican-3 (GPC3) T-cell therapy for hepatocellular carcinoma (HCC). We report herein the first published results of CAR-GPC3 T-cell therapy for HCC. PATIENTS AND METHODS: In two prospective phase I studies, adult patients with advanced GPC3+ HCC (Child-Pugh A) received autologous CAR-GPC3 T-cell therapy following cyclophosphamide- and fludarabine-induced lymphodepletion. The primary objective was to assess the treatment's safety. Adverse events were graded using the Common Terminology Criteria for Adverse Events (version 4.03). Tumor responses were evaluated using the RECIST (version 1.1). RESULTS: A total of 13 patients received a median of 19.9 × 108 CAR-GPC3 T cells by a data cutoff date of July 24, 2019. We observed pyrexia, decreased lymphocyte count, and cytokine release syndrome (CRS) in 13, 12, and nine patients, respectively. CRS (grade 1/2) was reversible in eight patients. One patient experienced grade 5 CRS. No patients had grade 3/4 neurotoxicity. The overall survival rates at 3 years, 1 year, and 6 months were 10.5%, 42.0%, and 50.3%, respectively, according to the Kaplan-Meier method. We confirmed two partial responses. One patient with sustained stable disease was alive after 44.2 months. CAR T-cell expansion tended to be positively associated with tumor response. CONCLUSIONS: This report demonstrated the initial safety profile of CAR-GPC3 T-cell therapy. We observed early signs of antitumor activity of CAR-GPC3 T cells in patients with advanced HCC.


Assuntos
Carcinoma Hepatocelular/terapia , Síndrome da Liberação de Citocina/epidemiologia , Febre/epidemiologia , Imunoterapia Adotiva/efeitos adversos , Neoplasias Hepáticas/terapia , Recidiva Local de Neoplasia/terapia , Adulto , Idoso , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Ensaios Clínicos Fase I como Assunto , Síndrome da Liberação de Citocina/imunologia , Feminino , Febre/imunologia , Glipicanas/genética , Glipicanas/imunologia , Humanos , Imunoterapia Adotiva/métodos , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , Contagem de Linfócitos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/diagnóstico , Recidiva Local de Neoplasia/imunologia , Recidiva Local de Neoplasia/patologia , Estadiamento de Neoplasias , Estudos Prospectivos , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Linfócitos T/transplante , Transplante Autólogo/efeitos adversos , Transplante Autólogo/métodos , Resultado do Tratamento
5.
Am J Physiol Renal Physiol ; 318(4): F994-F1005, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32068461

RESUMO

Renal ischemia-reperfusion (IR) injury is one of the most common acute kidney injuries, but there is still a lack of effective treatment in the clinical setting. Trehalose (Tre), a natural disaccharide, has been demonstrated to protect against oxidative stress, inflammation, and apoptosis. However, whether it could protect against IR-induced renal injury needs to be investigated. In an in vivo experiment, C57BL/6J mice were pretreated with or without Tre (2 g/kg) through a daily single intraperitoneal injection from 3 days before renal IR surgery. Renal function, apoptosis, oxidative stress, and inflammation were analyzed to evaluate kidney injury. In an in vitro experiment, mouse proximal tubular cells were treated with or without Tre under a hypoxia/reoxygenation condition. Western blot analysis, autophagy flux detection, and apoptosis assay were performed to evaluate the level of autophagy and antiapoptotic effect of Tre. The in vivo results showed that the renal damage induced by IR was ameliorated by Tre treatment, as renal histology and renal function were improved and the enhanced protein levels of kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin were blocked. Moreover, autophagy was activated by Tre pretreatment along with inhibition of the IR injury-induced apoptosis, oxidative stress, and inflammation. The in vitro results showed that Tre treatment activated autophagy and protected against hypoxia/reoxygenation-induced tubular cell apoptosis and oxidative stress. Our results demonstrated that Tre protects against IR-induced renal injury, possibly by enhancing autophagy and blocking oxidative stress, inflammation, and apoptosis, suggesting its potential use for the clinical treatment of renal IR injury.


Assuntos
Injúria Renal Aguda/prevenção & controle , Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Autofagia/efeitos dos fármacos , Mediadores da Inflamação/metabolismo , Rim/efeitos dos fármacos , Nefrite/prevenção & controle , Estresse Oxidativo/efeitos dos fármacos , Traumatismo por Reperfusão/prevenção & controle , Trealose/farmacologia , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Animais , Apoptose/efeitos dos fármacos , Biomarcadores/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Rim/metabolismo , Rim/patologia , Masculino , Camundongos Endogâmicos C57BL , Nefrite/metabolismo , Nefrite/patologia , Infiltração de Neutrófilos/efeitos dos fármacos , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Transdução de Sinais
6.
Free Radic Biol Med ; 152: 821-837, 2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32004633

RESUMO

Mitochondrial dysfunction plays an important role in acute kidney injury (AKI). Thus, the agents improving the mitochondrial function could be beneficial for treating AKI. Ursodeoxycholic acid (UDCA) has been demonstrated to prevent mitochondrial dysfunction under pathology, however, its role in AKI and the underlying mechanism remain unknown. This study aimed to evaluate the effect of UDCA on cisplatin-induced AKI. In vivo, C57BL/6 J mice were treated with cisplatin (25 mg/kg) for 72 h to induce AKI through a single intraperitoneal (i.p.) injection with or without UDCA (60 mg/kg/day) administration by gavage. Renal function, mitochondrial function and oxidative stress were analyzed to evaluate kidney injury. In vitro, mouse proximal tubular cells (mPTCs) and human proximal tubule epithelial cells (HK2) were treated with cisplatin with or without UDCA treatment for 24 h. Transcriptomic RNA-seq was preformed to analyze possible targets of UDCA. Our results showed that cisplatin-induced increments of serum creatinine (Scr), blood urea nitrogen (BUN), and cystatin C were significantly reduced by UDCA along with ameliorated renal tubular injury evidenced by improved renal histology and blocked upregulation of neutrophil gelatinase associated lipocalin (NGAL) and kidney injury molecule 1 (KIM-1). Meanwhile, the apoptosis induced by cisplatin was also markedly attenuated by UDCA administration. In vitro, UDCA treatment protected against tubular cell apoptosis possibly through antagonizing mitochondrial dysfunction and oxidative stress by targeting ALDH1L2 which was screened out by an RNA-seq analysis. Knockout of ALDH1L2 by CRISPR/Cas9 greatly blunted the protective effects of UDCA in renal tubular cells. Moreover, UDCA did not diminish cisplatin's antineoplastic effect in human cancer cells. In all, our results demonstrated that UDCA protects against cisplatin-induced AKI through improving mitochondrial function through acting on the expression of ALDH1L2, suggesting a clinical potential of UDCA for the treatment of AKI.


Assuntos
Injúria Renal Aguda , Cisplatino , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/genética , Animais , Apoptose , Cisplatino/toxicidade , Rim/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias , Ácido Ursodesoxicólico/metabolismo , Ácido Ursodesoxicólico/farmacologia
7.
Front Med ; 13(1): 83-93, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30671888

RESUMO

Colorectal cancer (CRC) is a common malignant tumor in the digestive tract, and 30%-85% of CRCs express epidermal growth factor receptors (EGFRs). Recently, treatments using cetuximab, also named C225, an anti-EGFR monoclonal antibody, for CRC have been demonstrated to cause an S492R mutation in EGFR. However, little is known about the biological function of S492R EGFR. Therefore, we attempted to elucidate its biological function in CRC cells and explore new treatment strategies for this mutant form. Our study indicated that EGFR and S492R EGFR accelerate the growth of CRC cells in vitro and in vivo and monoclonal antibody CH12, which specifically recognizes an EGFR tumor-specific epitope, can bind efficiently to S492R EGFR. Furthermore, mAb CH12 showed significantly stronger growth suppression activities and induced a more potent antibody-dependent cellular cytotoxicity effect on CRC cells bearing S492R EGFR than mAb C225. mAb CH12 obviously suppressed the growth of CRC xenografts with S492R EGFR mutations in vivo. Thus, mAb CH12 may be a promising therapeutic agent in treating patients with CRC bearing an S492R EGFR mutation.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos Imunológicos/farmacologia , Neoplasias Colorretais/terapia , Receptores ErbB/genética , Animais , Células CACO-2 , Proliferação de Células/efeitos dos fármacos , Receptores ErbB/imunologia , Feminino , Células HT29 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Cancer Immunol Res ; 6(11): 1314-1326, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30201736

RESUMO

The heterogeneous expression of EGFRvIII [variant III mutant of epidermal growth factor receptor (EGFR)] in glioblastoma has significant impact on the clinical response to the treatment of EGFRvIII-specific chimeric antigen receptor-engineered T (CAR T) cells. We hypothesized that CAR T cells that could target both EGFRvIII and the form of EGFR expressed on tumor cells, but not EGFR on normal cells, would greatly improve efficacy without inducing on-target, off-tumor toxicity. Therefore, we developed a humanized single-chain antibody, M27, with a single specificity that binds to an epitope found both on wild-type EGFR- and EGFRvIII-overexpressing tumor cells, but not EGFR-expressing normal cells, including primary keratinocytes, on which wild-type EGFR is highly expressed. M27-derived CAR T cells effectively lysed EGFRvIII- or EGFR-overexpressing tumor cells, but showed no observable toxicity on normal cells. Inclusion of the CD137 (4-1BB) costimulatory intracellular domain in the M27-28BBZ CAR provided CAR T cells with higher tumor lysis activity than when not included (as in the M27-28Z CAR). The growth of established EGFR- or EGFRvIII-overexpressing glioma xenografts was suppressed by M27-28BBZ CAR T cells as well. The growth of heterogeneic xenograft tumors, created by mixing EGFR- and EGFR-overexpressing glioblastoma cells, was also effectively inhibited by M27-28BBZ CAR T cells. The survival of mice in the orthotopic models was significantly prolonged after M27-28BBZ CAR T-cell infusion. These results suggested that tumor-selective, bitargeted anti-EGFR/EGFRvIII CAR T cells may be a promising modality for the treatment of patients with EGFR/EGFRvIII-overexpressing glioblastoma. Cancer Immunol Res; 6(11); 1314-26. ©2018 AACR.


Assuntos
Anticorpos Monoclonais Humanizados/imunologia , Glioblastoma/terapia , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/transplante , Animais , Anticorpos Monoclonais Humanizados/genética , Anticorpos Monoclonais Humanizados/metabolismo , Linhagem Celular Tumoral , Citocinas/metabolismo , Epitopos/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Feminino , Glioblastoma/imunologia , Glioblastoma/patologia , Humanos , Camundongos SCID , Terapia de Alvo Molecular , Receptores de Antígenos Quiméricos/genética , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Mol Ther ; 25(10): 2270-2279, 2017 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-28757080

RESUMO

The incorporation of an endogenous safety switch represents a rational strategy for the control of toxicities following the administration of adoptive T cell therapies. An ideal safety switch should be capable of depleting the transferred T cells with minimal injury to normal tissues. We generated a fusion receptor by engineering a cryptic 806 epitope of human epidermal growth factor receptor (EGFR) into the N terminus of the full-length human folate receptor 1 (FOLR1), designated as FR806. The expression of FR806 allows transduced T cells to be targeted with CH12, a monoclonal antibody recognizing the 806 epitope, but not wild-type EGFR in healthy tissues. FR806, therefore, constitutes a specific cell-surface marker for the elimination of transduced T cells. We demonstrate that the antibody-drug conjugate (ADC) CH12-MMAF is efficiently internalized by FR806-expressing T cells and has the potential to eliminate them. Transfected T cells could, furthermore, be efficiently detected and purified using CH12 antibodies. In immuno-compromised mice, CH12-MMAF eliminated the majority of transferred T cells expressing FR806 and anti-CD19 chimeric antigen receptor (CAR). The selectivity for the 806 epitope and internalization capacity of FOLR1 makes FR806 an efficient safety switch, which may additionally be used as a detection and purification biomarker for human T cell immunotherapies.


Assuntos
Transferência Adotiva/métodos , Biomarcadores/sangue , Linfócitos T/imunologia , Animais , Linhagem Celular , Humanos , Interferon gama/metabolismo , Interleucina-2/metabolismo , Camundongos , Camundongos SCID , Linfócitos T/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
10.
Cancer Immunol Immunother ; 66(4): 475-489, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28035433

RESUMO

Adoptive immunotherapy leveraging chimeric antigen receptor-modified T (CAR-T) cells holds great promise for the treatment of cancer. However, tumor-associated antigens often have low expression levels in normal tissues, which can cause on-target, off-tumor toxicity. Recently, we reported that GPC3-targeted CAR-T cells could eradicate hepatocellular carcinoma (HCC) xenografts in mice. However, it remains unknown whether on-target, off-tumor toxicity can occur. Therefore, we proposed that dual-targeted CAR-T cells co-expressing glypican-3 (GPC3) and asialoglycoprotein receptor 1 (ASGR1) (a liver tissue-specific protein)-targeted CARs featuring CD3ζ and 28BB (containing both CD28 and 4-1BB signaling domains), respectively, may have reduced on-target, off-tumor toxicity. Our results demonstrated that dual-targeted CAR-T cells caused no cytotoxicity to ASGR1+GPC3- tumor cells, but they exhibited a similar cytotoxicity against GPC3+ASGR1- and GPC3+ASGR1+ HCC cells in vitro. We found that dual-targeted CAR-T cells showed significantly higher cytokine secretion, proliferation and antiapoptosis ability against tumor cells bearing both antigens than single-targeted CAR-T cells in vitro. Furthermore, the dual-targeted CAR-T cells displayed potent growth suppression activity on GPC3+ASGR1+ HCC tumor xenografts, while no obvious growth suppression was seen with single or double antigen-negative tumor xenografts. Additionally, the dual-targeted T cells exerted superior anticancer activity and persistence against single-targeted T cells in two GPC3+ASGR1+ HCC xenograft models. Together, T cells carrying two complementary CARs against GPC3 and ASGR1 may reduce the risk of on-target, off-tumor toxicity while maintaining relatively potent antitumor activities on GPC3+ASGR1+ HCC.


Assuntos
Vacinas Anticâncer/imunologia , Carcinoma Hepatocelular/terapia , Imunoterapia Adotiva/métodos , Neoplasias Hepáticas/terapia , Receptores de Antígenos de Linfócitos T/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Linfócitos T/fisiologia , Animais , Receptor de Asialoglicoproteína/imunologia , Carcinoma Hepatocelular/imunologia , Linhagem Celular Tumoral , Proliferação de Células , Citocinas/metabolismo , Glipicanas/imunologia , Humanos , Neoplasias Hepáticas/imunologia , Ativação Linfocitária , Camundongos , Camundongos SCID , Especificidade de Órgãos , Receptores de Antígenos de Linfócitos T/genética , Proteínas Recombinantes de Fusão/genética , Especificidade do Receptor de Antígeno de Linfócitos T , Linfócitos T/transplante , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Oncotarget ; 7(3): 2496-507, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26684028

RESUMO

There are unmet medical needs for patients with lung squamous cell carcinoma (LSCC). Therefore, in this study, we explored the antitumor potential of third-generation glypican 3 (GPC3)-redirected chimeric antigen receptor (CAR)-engineered T lymphocytes (CARgpc3 T cells) in tumor models of LSCC. First, we demonstrated by immunohistochemistry (IHC) that GPC3 was expressed in 66.3% of LSCC samples and in 3.3% of lung adenocarcinoma (LAD) samples but not in normal lung tissues. In the presence of GPC3-positive LSCC cells, CARgpc3 T cells were highly activated and increased in number. CARgpc3 T cells could specifically lyse GPC3-positive LSCC cells in vitro. In two established LSCC xenograft models, CARgpc3 T cells could almost completely eliminate the growth of GPC3-positive cells. Additionally, the CARgpc3 T cells were able to persist in vivo and efficiently infiltrate the cancerous tissues. Taken together, these findings indicate that CARgpc3 T cells might be a novel potential therapeutic agent for the treatment of patients with LSCC.


Assuntos
Carcinoma de Células Escamosas/terapia , Glipicanas/imunologia , Imunoterapia Adotiva , Neoplasias Pulmonares/terapia , Linfócitos T Citotóxicos/imunologia , Animais , Apoptose , Western Blotting , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Proliferação de Células , Citometria de Fluxo , Glipicanas/metabolismo , Humanos , Técnicas Imunoenzimáticas , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Receptores de Antígenos de Linfócitos T/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Clin Cancer Res ; 20(24): 6418-28, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25320357

RESUMO

PURPOSE: The aim of our study is to elucidate whether T cells expressing GPC3-targeted chimeric antigen receptor (CAR) can efficiently eliminate GPC3-positive HCC cells and their potential in the treatment of HCC. EXPERIMENTAL DESIGN: T cells expressing a first-generation and third-generation GPC3-targeted CAR were prepared using lentiviral vector transduction. The in vitro and in vivo cytotoxic activities of the genetically engineered CAR T cells were evaluated against various HCC cell lines. RESULTS: GPC3-targeted CAR T cells could efficiently kill GPC3-positive HCC cells but not GPC3-negative cells in vitro. These cytotoxic activities seemed to be positively correlated with GPC3 expression levels in the target cells. In addition, T cells expressing the third-generation GPC3-targeted CAR could eradicate HCC xenografts with high level of GPC3 expression and efficiently suppress the growth of HCC xenografts with low GPC3 expression level in vivo. The survival of the mice bearing established orthotopic Huh-7 xenografts was significantly prolonged by the treatment with the third-generation GPC3-targeted CAR T cells. CONCLUSIONS: GPC3-targeted CAR T cells could potently eliminate GPC3-positive HCC cells, thereby providing a promising therapeutic intervention for GPC3-positive HCC.


Assuntos
Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Glipicanas/imunologia , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , Subpopulações de Linfócitos T/imunologia , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/mortalidade , Carcinoma Hepatocelular/terapia , Linhagem Celular Tumoral , Citocinas/biossíntese , Citotoxicidade Imunológica , Modelos Animais de Doenças , Ordem dos Genes , Vetores Genéticos/genética , Humanos , Imunoterapia , Lentivirus/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/mortalidade , Neoplasias Hepáticas/terapia , Camundongos , Receptores de Antígenos de Linfócitos T/genética , Proteínas Recombinantes de Fusão/genética , Anticorpos de Domínio Único/genética , Anticorpos de Domínio Único/imunologia , Especificidade do Receptor de Antígeno de Linfócitos T/genética , Especificidade do Receptor de Antígeno de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Transdução Genética , Carga Tumoral/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Cancer Lett ; 342(1): 113-20, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24007863

RESUMO

5-Fluorouracil (5-FU) is one of the most common chemotherapeutic agents used for the treatment of hepatocellular carcinoma (HCC). However, chemoresistance has precluded the use of 5-FU alone in clinical regimens. Combination therapies with 5-FU and other anticancer agents are considered to be a therapeutic option for patients with HCC. We previously reported that the expression of epidermal growth factor receptor variant III (EGFRvIII) can decrease the sensitivity of HCC cells to 5-FU. To overcome this problem, in this study, we elucidated the mechanism underlying EGFRvIII-mediated 5-FU resistance. We observed that EGFRvIII expression can induce miR-520d-3p downregulation and the ensuing upregulation of the transcription factor E2F-1 and the enzyme thymidylate synthase (TS), which may lead to drug resistance. Intriguingly, we found that CH12, a monoclonal antibody directed against EGFRvIII, and 5-FU together had an additive antitumor effect on EGFRvIII-positive HCC xenografts and significantly improved survival in all mice with established tumors when compared with either 5-FU or CH12 alone. Mechanistically, compared with 5-FU alone, the combination more noticeably downregulated EGFR phosphorylation and Akt phosphorylation as well as the expression of the apoptotic protector Bcl-xL and the cell cycle regulator cyclin D1. Additionally, the combination upregulated the expression of the cell cycle inhibitor p27 in in vivo treatment. More interestingly, CH12 treatment upregulated miR-520-3p and downregulated E2F-1 and TS at the mRNA and protein levels. Collectively, these observations suggest that the combination of 5-FU with mAb CH12 is a potential means of circumventing EGFRvIII-mediated 5-FU resistance in HCC.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Hepatocelular/tratamento farmacológico , Receptores ErbB/antagonistas & inibidores , Neoplasias Hepáticas/tratamento farmacológico , Animais , Anticorpos Monoclonais/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Di-Hidrouracila Desidrogenase (NAD+)/metabolismo , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F1/metabolismo , Receptores ErbB/imunologia , Fluoruracila/administração & dosagem , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Nus , MicroRNAs/genética , Orotato Fosforribosiltransferase/metabolismo , Interferência de RNA , Timidilato Sintase/genética , Timidilato Sintase/metabolismo , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Cancer Immunol Immunother ; 63(2): 121-32, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24177984

RESUMO

There have been several studies suggesting that cancer stem cells (CSCs) contribute to the high rates of recurrence and resistance to therapies observed in hepatocellular carcinoma (HCC). Epithelial cell adhesion molecule (EpCAM) has been demonstrated to be a biomarker of CSCs and a potential therapeutic target in HCC. Here, we prepared two anti-EpCAM monoclonal antibodies (1H8 and 2F2) and an anti-EpCAM bispecific T cell engager (BiTE) 1H8/CD3, which was derived from 1H8, and used them to treat HCC in vitro and in vivo. The results demonstrated that all of the developed anti-EpCAM antibodies specifically bound to EpCAM. Neither anti-EpCAM monoclonal antibody had obvious anti-HCC activities in vitro or in vivo. However, anti-EpCAM BiTE 1H8/CD3 induced strong peripheral blood mononuclear cell-dependent cellular cytotoxicity in Huh-7 and Hep3B cells but not EpCAM-negative SK-Hep-1 cells. Notably, 1H8/CD3 completely inhibited the growth of Huh-7 and Hep3B xenografts in vivo. Treatment of the Huh-7 HCC xenografts with 1H8/CD3 significantly suppressed tumor proliferation and reduced the expression of most CSC biomarkers. Intriguingly, galectin-1 (Gal-1) overexpression inhibited 1H8/CD3-induced lymphocytotoxicity in HCCs while knockdown of Gal-1 increased the lymphocytotoxicity. Collectively, these results indicate that anti-EpCAM BiTE 1H8/CD3 is a promising therapeutic agent for HCC treatment. Gal-1 may contribute to the resistance of HCC cells to 1H8/CD3-induced lysis.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Antígenos de Neoplasias/imunologia , Complexo CD3/imunologia , Carcinoma Hepatocelular/terapia , Moléculas de Adesão Celular/imunologia , Galectina 1/análise , Neoplasias Hepáticas/terapia , Antígeno AC133 , Animais , Antígenos CD/análise , Antígenos de Neoplasias/análise , Moléculas de Adesão Celular/análise , Linhagem Celular Tumoral , Molécula de Adesão da Célula Epitelial , Galectina 1/fisiologia , Glicoproteínas/análise , Humanos , Camundongos , Peptídeos/análise
15.
Cancer Res ; 73(23): 7056-67, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-24240702

RESUMO

As a validated therapeutic target in several human cancers, the EGF receptor (EGFR) provides a focus to gain deeper insights into cancer pathophysiology. In this study, we report the identification of a naturally occurring and widely expressed EGFR isoform termed EGFRvA, which substitutes a Ser/Thr-rich peptide for part of the carboxyl-terminal regulatory domain of the receptor. Intriguingly, EGFRvA expression relates more closely to histopathologic grade and poor prognosis in patients with glioma. Ectopic expression of EGFRvA in cancer cells conferred a higher invasive capacity than EGFR in vitro and in vivo. Mechanistically, EGFRvA stimulated expression of STAT3, which upregulated heparin-binding EGF (HB-EGF). Reciprocally, HB-EGF stimulated phosphorylation of EGFRvA at Y845 along with STAT3, generating a positive feedback loop that may reinforce invasive function. The significance of EGFRvA expression was reinforced by findings that it is attenuated by miR-542-5p, a microRNA that is a known tumor suppressor. Taken together, our findings define this newfound EGFR isoform as a key theranostic molecule.


Assuntos
Receptores ErbB/fisiologia , Invasividade Neoplásica/genética , Neoplasias/patologia , Animais , Movimento Celular/genética , Células HEK293 , Humanos , Camundongos , Células NIH 3T3 , Neoplasias/genética , Prognóstico , Isoformas de Proteínas/fisiologia , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Análise de Sobrevida , Células Tumorais Cultivadas
16.
Carcinogenesis ; 34(11): 2639-46, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23764753

RESUMO

Recently, de4 EGFR, a variant of epidermal growth factor receptor (EGFR) with exon 4 deletion, was identified in glioblastoma and ovarian cancer. However, its biological function on ovarian cancer is still not clear. In this study, the expression profile of de4 EGFR and its contribution to epithelial ovarian cancer cells proliferation, invasiveness and drug resistance were studied. Our results showed that 48.6% (35/72) of epithelial ovarian cancer tissues had de4 EGFR expression and the expression ratio positively correlated with clinical stages. Compared with EGFR transfectants, de4 EGFR transfectants exhibited significantly higher level of invasiveness in vitro. Mechanistically, de4 EGFR significantly upregulated the extracellular regulated protein kinase, AKT, focal adhesion kinase (FAK) and Src phosphorylation and matrix metalloproteinase-9 expression while downregulated the expression of E-cadherin. Additionally, knockdown of FAK obviously suppressed de4 EGFR-induced invasiveness. Interestingly, de4 EGFR transfectants displayed significantly lower sensitivity to cisplatin than EGFR transfectants, which could be ascribed to the upregulation of Bcl-2 and downregulation of BAD in the de4 EGFR transfectants. Collectively, these results demonstrate that de4 EGFR plays an important role in the invasiveness and cisplatin resistance in epithelial ovarian cancer cells and may provide a new potential therapeutic target for epithelial ovarian cancer.


Assuntos
Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Receptores ErbB/genética , Éxons/genética , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/patologia , Deleção de Sequência , Animais , Apoptose , Western Blotting , Carcinoma Epitelial do Ovário , Adesão Celular , Movimento Celular , Proliferação de Células , Feminino , Proteína-Tirosina Quinases de Adesão Focal/antagonistas & inibidores , Proteína-Tirosina Quinases de Adesão Focal/genética , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Técnicas Imunoenzimáticas , Camundongos , Camundongos Nus , Neoplasias Epiteliais e Glandulares/tratamento farmacológico , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Fosforilação , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Células Tumorais Cultivadas
17.
Oncol Rep ; 28(4): 1453-60, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22895605

RESUMO

Hepatocellular carcinoma (HCC) overexpresses insulin-like growth factor-I receptor (IGF-IR), as compared with normal hepatocytes. Since IGF-1R-mediated signaling promotes survival, oncogenic transformation and tumor growth and spread, it represents a potential target for treating HCC. Here, we have generated a murine anti-IGF-1R antibody, 4F2, that recognizes the IGF-IRα subunit and blocks in vitro IGF-I and IGF-II-induced cell proliferation of SMMC-7721 and Bel-7402 HCC cell lines. 4F2 can inhibit IGF-IR autophosphorylation, IRS-1 phosphorylation and the activation of the major downstream signaling molecules AKT and mitogen-activated protein kinase. Additionally, we observed a moderate increase in apoptosis as demonstrated by detection of changes in the expression of the pro-apoptotic and anti-apoptotic proteins Bax and Bcl-2 after 4F2 treatment. Combined treatment with 4F2 and doxorubicin was more effective in reducing cell proliferation and promoting apoptosis than either agent alone. These data support that therapeutic anti-IGF-IR antibodies are potential new agents for treating HCC.


Assuntos
Anticorpos Monoclonais/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Receptor IGF Tipo 1/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Regulação Neoplásica da Expressão Gênica , Humanos , Fator de Crescimento Insulin-Like I/farmacologia , Fator de Crescimento Insulin-Like II/farmacologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptor IGF Tipo 1/genética , Transdução de Sinais/efeitos dos fármacos , Proteína X Associada a bcl-2/metabolismo
18.
Neoplasia ; 14(6): 509-18, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22787432

RESUMO

The multikinase inhibitor sorafenib is the first oral agent to show activity against human hepatocellular carcinoma (HCC). Although the clinical application of sorafenib has shown good tolerability in the studied populations, it also causes multiple human dose-limiting toxicities. Thus, there is a strong need to reduce the overall dose of sorafenib. We have reported that the epidermal growth factor receptor variant III (EGFRvIII) expression can decrease the sensitivity of HCC cells to chemotherapeutic drugs. Therefore, we sought to explore whether EGFRvIII can affect the sensitivity of HCC cells to sorafenib. In this study, we observed that EGFRvIII expression significantly decreased the sensitivity of HCC cells to sorafenib. To enhance the antitumor effect and reduce the overall dose of sorafenib, we evaluated the combined effects of CH12, a monoclonal antibody against EGFRvIII, and sorafenib on the growth of HCC cells expressing EGFRvIII in vitro and in vivo. The results showed that, when CH12 was combined with sorafenib, the tumor growth suppression effect was significantly increased, and the concentration of sorafenib required for growth inhibition was substantially reduced. Mechanistically, the combination could more noticeably downregulate the phosphorylation of constitutively active extracellular signal-regulated kinase (ERK), Akt (Thr308), and signal transducer and activator of transcription 3 (STAT3) than sorafenib alone. Collectively, these findings demonstrate that CH12 interacts additively with sorafenib to strongly inhibit the tumor growth of HCC xenografts expressing EGFRvIII by enhancing the sorafenib-mediated inhibition of the MEK/ERK, phosphoinositide 3-kinase/AKT, and STAT3 pathways.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Benzenossulfonatos/farmacologia , Carcinoma Hepatocelular/metabolismo , Receptores ErbB/metabolismo , Neoplasias Hepáticas/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Animais , Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Benzenossulfonatos/administração & dosagem , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Sinergismo Farmacológico , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Nus , Neovascularização Patológica/tratamento farmacológico , Niacinamida/análogos & derivados , Compostos de Fenilureia , Inibidores de Proteínas Quinases/administração & dosagem , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piridinas/administração & dosagem , Fator de Transcrição STAT3/metabolismo , Sorafenibe , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Biomarkers ; 15(2): 128-34, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19839718

RESUMO

OBJECTIVE: We investigated whether or not there are autoantibodies for DKK1 (Dickkopf-1) in patients with non-small cell lung cancer (NSCLC) and whether this autoantibody can be used for cancer detection. METHODS: The levels of DKK1 autoantibodies were determined in 93 NSCLC patients and 87 healthy controls. RESULTS: We found that, in the sera, the presence of autoantibody against DKK1 was highly correlated with NSCLC. High anti-DKK1 autoantibody titres were found in the sera of NSCLC patients, whereas low or negative titres were found in the control group. The ROC curve results showed that autoantibody immunoassay exhibited 62% sensitivity and 84% specificity. The sensitivity for the detection of NSCLC in stage I also reach 64.3%. Furthermore, a combined ELISA assays for both DKK1 and autoantibody DKK1 increased sensitivity and classified 81.7% (76/93) of the NSCLC patients as positive, whereas only 13.8 % (12/87) of healthy volunteers were falsely diagnosed as positive. CONCLUSIONS: Our results suggest that the detection of circulating DKK1 autoantibody could potentially serve as a useful non-invasive marker for determining lung cancer status.


Assuntos
Autoanticorpos/sangue , Biomarcadores Tumorais/sangue , Carcinoma Pulmonar de Células não Pequenas/genética , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Neoplasias Pulmonares/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma Pulmonar de Células não Pequenas/imunologia , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Pulmonares/imunologia , Masculino , Pessoa de Meia-Idade , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA