Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Immunother Cancer ; 12(3)2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38538093

RESUMO

BACKGROUND: Choosing treatments for epidermal growth factor receptor (EGFR)-mutated non-small cell lung cancer (NSCLC) patients with osimertinib resistance is challenging. We evaluated the safety and efficacy of SNK01 (autologous natural killer (NK) cells) in combination with cytotoxic chemotherapy and/or cetuximab (an anti-EGFR monoclonal antibody) in treating EGFR-mutated NSCLC in this non-clinical and phase I/IIa clinical trial. METHODS: We developed a cell line-derived xenograft-humanized mouse model with an osimertinib-resistant lung cancer cell line. The mice were divided into four groups based on treatment (no treatment, cetuximab, SNK01, and combination groups) and treated weekly for 5 weeks. In the clinical study, 12 patients with EGFR-mutated NSCLC who failed prior tyrosine kinase inhibitor (TKI) received SNK01 weekly in combination with gemcitabine/carboplatin (n=6) or cetuximab/gemcitabine/carboplatin (n=6) and dose escalation of SNK01 following the "3+3" design. RESULTS: In the non-clinical study, an increase in NK cells in the blood and enhanced NK cell tumor infiltration were observed in the SNK01 group. The volume of tumor extracted after treatment was the smallest in the combination group. In the clinical study, 12 patients (median age, 60.9 years; all adenocarcinoma cases) received SNK01 weekly for 7-8 weeks (4×109 cells/dose (n=6); 6×109 cells/dose (n=6)). The maximum feasible dose of SNK01 was 6×109 cells/dose without dose-limiting toxicity. Efficacy outcomes showed an objective response rate of 25%, disease control rate of 100%, and median progression-free survival of 143 days. CONCLUSION: SNK01 in combination with cytotoxic chemotherapy, including cetuximab, for EGFR-mutated NSCLC with TKI resistance was safe and exerted a potential antitumor effect. TRIAL REGISTRATION NUMBER: NCT04872634.


Assuntos
Acrilamidas , Compostos de Anilina , Carcinoma Pulmonar de Células não Pequenas , Indóis , Neoplasias Pulmonares , Pirimidinas , Humanos , Camundongos , Animais , Pessoa de Meia-Idade , Carcinoma Pulmonar de Células não Pequenas/patologia , Cetuximab/farmacologia , Cetuximab/uso terapêutico , Neoplasias Pulmonares/patologia , Carboplatina/uso terapêutico , Gencitabina , Receptores ErbB/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Células Matadoras Naturais/metabolismo
2.
Ann Plast Surg ; 69(3): 331-7, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22907186

RESUMO

BACKGROUND: Parry-Romberg disease is a rare condition that results in progressive hemifacial atrophy, involving the skin, dermis, subcutaneous fat, muscle, and, finally, cartilage and bone. Patients have been treated with dermofat or fat grafts or by microvascular free flap transfer. We hypothesized that adipose-derived stem cells (ASCs) may improve the results of microfat grafting through enhancing angiogenesis. We evaluated the utility of ASC in microfat grafting of patients with Parry-Romberg disease by measuring the change in the hemifacial volumes after injection of ASCs with microfat grafts or microfat grafts alone. METHODS: In April 2008, this investigation was approved by the Korean Food and Drug Administration and the institutional review board of the Asan Medical Center (Seoul, Korea) that monitor investigator-initiated trials. Between May 2008 and January 2009, 10 volunteers with Parry-Romberg disease (5 men and 5 women; mean age, 28 y) were recruited; 5 received ASC and microfat grafts and 5 received microfat grafts only. The mean follow-up period was 15 months. Adipose-derived stem cells were obtained from abdominal fat by liposuction and were cultured for 2 weeks. On day 14, patients were injected with fat grafts alone or plus (in the test group) 1 × 10 ASCs. Patients were evaluated postoperatively using a 3-dimensional camera and 3-dimensional CT scans, and grafted fat volumes were objectively calculated. RESULTS: Successful outcomes were evident in all 5 patients receiving microfat grafts and ASCs, and the survival of grafted fat was better than in patients receiving microfat grafts alone. Before surgery, the mean difference between ipsilateral and contralateral hemiface volume in patients receiving microfat grafts and ASCs was 21.71 mL decreasing to 4.47 mL after surgery. Overall resorption in this ASC group was 20.59%. The mean preoperative difference in hemiface volume in those receiving microfat grafts alone was 8.32 mL decreasing to 3.89 mL after surgery. Overall resorption in this group was 46.81%. The preoperative and postoperative volume differences between the groups was statistically significant (P = 0.002; random-effects model [SAS 9.1]). CONCLUSIONS: Adipose-derived stem cells enhance the survival of fat grafted into the face. A microfat graft with simultaneous ASC injection may be used to treat Parry-Romberg disease without the need for microvascular free flap transfer.


Assuntos
Tecido Adiposo/transplante , Hemiatrofia Facial/cirurgia , Imageamento Tridimensional , Transplante de Células-Tronco Mesenquimais , Procedimentos de Cirurgia Plástica/métodos , Cirurgia Assistida por Computador , Tomografia Computadorizada por Raios X/métodos , Tecido Adiposo/citologia , Adulto , Feminino , Humanos , Imageamento Tridimensional/instrumentação , Masculino
3.
Thorac Cancer ; 13(14): 2050-2056, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35670036

RESUMO

BACKGROUND: A previous trial showed that autologous ex-vivo expanded NK cell (SNK01) treatment combined with pembrolizumab showed better efficacy than pembrolizumab monotherapy in advanced non-small cell lung cancer (NSCLC). This study was a 2-year follow-up of that previous study to determine the long-term efficacy of the combination treatment. METHODS: This trial included 20 patients with advanced NSCLC with a PD-L1 tumor proportion score of 1% or greater who failed prior to front-line platinum-based therapy. The patients received pembrolizumab with low-dose SNK01 (2 × 109 cells/dose) or high-dose SNK01 (4 × 109 cells/dose), or pembrolizumab monotherapy. The primary study endpoint was overall survival (OS), and the secondary endpoint was progression-free survival (PFS). RESULTS: Two patients were excluded following serious adverse events. Among the 11 patients who died, five were from the NK groups (41.6%, n = 5/12), and six received pembrolizumab monotherapy (100%, n = 6/6). The estimated 2-year survival rate was 58.3% versus 16.7% (pembrolizumab plus SNK01 vs. pembrolizumab monotherapy). The hazard ratio of pembrolizumab plus SNK01 compared with pembrolizumab monotherapy was 0.32 (95% CI: 0.1, 1.08, p-value: 0.066). Although the median PFS was significantly higher in the pembrolizumab plus SNK01 group than in the pembrolizumab alone group, OS and PFS did not differ statistically between patients who received low doses of NK cells and those who received high doses of NK cells. CONCLUSIONS: Autologous NK cells can enhance the long-term OS and PFS for NSCLC. A larger study is needed to confirm this result. Clinical Research Information Service number: KCT0003463.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Antígeno B7-H1/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/patologia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Humanos , Células Matadoras Naturais/transplante , Neoplasias Pulmonares/patologia , Intervalo Livre de Progressão
4.
Cancer Res Treat ; 54(4): 1005-1016, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34856706

RESUMO

PURPOSE: The aim of this study is to evaluate the safety and efficacy of ex vivo activated and expanded natural killer (NK) cell therapy (SNK01) plus pembrolizumab in a randomized phase I/IIa clinical trial. MATERIALS AND METHODS: Overall, 18 patients with advanced non-small cell lung cancer (NSCLC) and a programmed death ligand 1 tumor proportion score of 1% or greater who had a history of failed frontline platinum-based therapy were randomized (2:1) to receive pembrolizumab every 3 weeks +/- 6 weekly infusions of SNK01 at either 2×109 or 4×109 cells per infusion (pembrolizumab monotherapy vs. SNK01 combination). The primary endpoint was safety, whereas the secondary endpoints were the objective response rate (ORR), progression-free survival (PFS), overall survival, and quality of life. RESULTS: Since no dose-limiting toxicity was observed, the maximum tolerated dose was determined as SNK01 4×109 cells/dose. The safety data did not show any new safety signals when SNK01 was combined with pembrolizumab. The ORR and the 1-year survival rate in the NK combination group were higher than those in patients who underwent pembrolizumab monotherapy (ORR, 41.7% vs. 0%; 1-year survival rate, 66.7% vs. 50.0%). Furthermore, the median PFS was higher in the SNK01 combination group (6.2 months vs. 1.6 months, p=0.001). CONCLUSION: Based on the findings of this study, the NK cell combination therapy may consider as a safe treatment method for stage IV NSCLC patients who had a history of failed platinum-based therapy without an increase in adverse events.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Anticorpos Monoclonais Humanizados , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Antígeno B7-H1 , Carcinoma Pulmonar de Células não Pequenas/patologia , Humanos , Neoplasias Pulmonares/patologia , Qualidade de Vida
5.
Exp Mol Med ; 35(6): 486-93, 2003 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-14749525

RESUMO

The 2',3'-cyclic nucleotide 3'-phosphodiesterase (CNP), a protein of unknown function in vivo, is abundantly expressed in myelinating glia in two isoforms, CNP1 and CNP2. In this study, immunoblot analysis showed that CNP1 is the major isoform in adult forebrain, and that both isoforms are included in the postsynaptic density (PSD) fraction and tyrosine-phosphorylated at the basal level. However, subcellular distribution and detergent extraction data showed that CNP is nonspecifically associated with the PSD fraction. Immunocytochemistry revealed that CNP is detected, in a weak but punctate pattern, in dissociated rat hippocampal neurons of 3 days to 2 weeks in vitro. The CNP-positive punctae were distributed throughout soma and dendrites, and distinct from PSD95-positive ones. Immunoblot analysis indicated that CNP is also expressed in neuronal stem cell lines, HiB5 and F11. Interestingly, in addition to the known two isoforms, a new CNP isoform of MW 45 kDa was expressed in these cell lines and was the major type of isoform in F11 cells. Taken together, our data suggest that CNP is expressed in the early stage of in vitro development and nonspecifically included in the adult rat PSD fraction.


Assuntos
2',3'-Nucleotídeo Cíclico Fosfodiesterases/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Prosencéfalo/metabolismo , Envelhecimento/fisiologia , Animais , Células Cultivadas , Hipocampo/citologia , Hipocampo/metabolismo , Imuno-Histoquímica , Neurônios/metabolismo , Fosfotirosina/metabolismo , Prosencéfalo/citologia , Ratos , Ratos Sprague-Dawley , Especificidade por Substrato
6.
Mol Cells ; 16(1): 128-35, 2003 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-14503857

RESUMO

We have shown by protein sequencing that the phosphotyrosine-containing 48 kDa protein band of the rat cerebellar postsynaptic density fraction (CBL-PSD) is 2', 3'-cyclic nucleotide 3'-phosphodiesterase 2 (CNP2). Immunoblot analysis indicated that both CNP1 and CNP2 isoforms are present in the CBL-PSD fraction, whereas there is little CNP2 in the forebrain (FB)-PSD fraction. Both isoforms in the CBL-PSD fraction were tyrosine-phosphorylated to a basal extent. They were efficiently dissociated from the complexes in the PSD fraction by salt, but not by non-ionic detergents such as n-octyl glucoside (OG) and Triton X-100. Immunocytochemistry of dissociated cerebellar cultures revealed patchy CNP staining in oligodendrocytes (OLs), Purkinje cells (PCs), and unidentified PSD95-positive cells, but no staining in granule cells (GCs). Our results indicate that both CNP1 and CNP2 are expressed in cerian populations of cerebellar cells in addition to OL, and that they are associated with complexes that are co-isolated with the PSD.


Assuntos
2',3'-Nucleotídeo Cíclico Fosfodiesterases/metabolismo , Cerebelo/citologia , Isoenzimas/metabolismo , Neurônios/metabolismo , Sinapses/metabolismo , Animais , Cerebelo/metabolismo , Detergentes , Camundongos , Neuroglia/citologia , Neuroglia/metabolismo , Neurônios/citologia , Peptídeos/metabolismo , Fosfotirosina/metabolismo , Ratos , Ratos Sprague-Dawley , Frações Subcelulares/química , Frações Subcelulares/metabolismo
7.
Mol Cells ; 14(2): 238-44, 2002 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-12442896

RESUMO

We investigated the effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on the expression of synaptic proteins in dissociated E18 rat cortical cells. TCDD (0-50 nM) was added to plating media, and cell viability and expression of synaptic proteins were assayed on 4 and 7 days in vitro (DIV), respectively. TCDD had no apparent effect on early neurite outgrowth 12 h after plating. However, on 4 DIV, cell viability was reduced significantly, and neurons often revealed vacuoles in 20 or 50 nM culture and had limited secondary or higher order dendritic processes in 20 or 50 nM culture. Immunoblot analyses of cell homogenates indicated upregulation of NMDA receptor subunits (NR1, NR2A, and NR2B), but downregulation of synaptic organizing proteins (PSD-95, densin-180, and septin6 homologue) and a synapse-enriched enzyme (alphaCaMKII). Changes in the expression of synaptic proteins may be a underlying mechanism for altered synaptic transmission and neuropathy by TCDD.


Assuntos
Córtex Cerebral/metabolismo , Poluentes Ambientais/farmacologia , Dibenzodioxinas Policloradas/farmacologia , Sinapses/metabolismo , Animais , Linhagem Celular , Poluentes Ambientais/toxicidade , Técnicas In Vitro , Dibenzodioxinas Policloradas/toxicidade , Ratos , Receptores de N-Metil-D-Aspartato/biossíntese , Receptores de N-Metil-D-Aspartato/genética , Sinapses/efeitos dos fármacos
8.
Neurosci Lett ; 366(1): 29-33, 2004 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-15265584

RESUMO

The postsynaptic density (PSD) is a proteinaceous cellular structure that is specialized for postsynaptic signal transduction. Here, we show that eukaryotic translation factor-1A (eEF1A; formerly known as eEF-1alpha) is associated with the excitatory PSD in rat forebrain. Immunoblot analysis showed that eEF1A in the PSD fraction is enriched over homogenate. Salt (1.0M NaCl), but not non-ionic detergents such as Triton X-100 (1.0%) and n-octyl glucoside (1.0%), could dissociate eEF1A from the PSD core. In cultured cortical neurons, eEF1A was colocalized with postsynaptic markers (PSD95 and SynGAPalpha), but not with a presynaptic marker (synaptophysin). These results indicate that eEF1A is present in the PSD of the excitatory synapses.


Assuntos
Córtex Cerebral/metabolismo , Fator 1 de Elongação de Peptídeos/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/ultraestrutura , Immunoblotting , Prosencéfalo/citologia , Prosencéfalo/metabolismo , Prosencéfalo/ultraestrutura , Ratos , Transdução de Sinais , Sinapses/metabolismo
9.
Neurosci Lett ; 362(1): 53-6, 2004 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-15147779

RESUMO

This study examined a 55 kDa protein in the rat cerebellar postsynaptic density (PSD) fraction. The amino acid sequence of an HPLC-purified peptide derived from tryptic digestion of the protein was contained in eukaryotic translation elongation factor-1A (eEF1A, formerly known as eEF-1alpha). Immunoblot analysis showed that eEF1A is enriched in the PSD fraction and is tightly associated with the PSD 'core'. The association of eEF1A with the PSD was further evidenced by colocalization of the protein with PSD95, a PSD marker, in dissociated cerebellar cultures and immunoelectron microscopy of the adult rat cerebellum. Combined, our results indicate that eEF1A is associated with the PSD.


Assuntos
Cerebelo/química , Fator 1 de Elongação de Peptídeos/análise , Sinapses/química , Animais , Contagem de Células/métodos , Cerebelo/ultraestrutura , Ratos , Sinapses/ultraestrutura
10.
Neurobiol Dis ; 21(3): 457-67, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16202616

RESUMO

The aim of this study was to investigate the role of microglia in radiation-induced astrocyte gliosis. We found that a single dose of 15 Gy radiation to a whole rat brain increased immunostaining of glial fibrillary acidic protein in astrocytes 6 h later, and even more so 24 h later, indicating the initiation of gliosis. While irradiation of cultured rat astrocytes had little effect, irradiation of microglia-astrocyte mixed-cultures displayed altered astrocyte phenotype into more processed, which is another characteristic of gliosis. Experiments using microglia-conditioned media indicated this astrocyte change was due to factors released from irradiated microglia. Irradiation of cultured mouse microglial cells induced a dose-dependent increase in mRNA levels for cyclooxygenase-2 (COX-2), interleukin (IL)-1beta, IL-6, IL-18, tumor necrosis factor-alpha and interferon-gamma-inducible protein-10, which are usually associated with microglia activation. Consistent with these findings, irradiation of microglia activated NF-kappaB, a transcription factor that regulates microglial activation. Addition of prostaglandin E2 (PGE2: a metabolic product of the COX-2 enzyme) to primary cultured rat astrocytes resulted in phenotypic changes similar to those observed in mixed-culture experiments. Therefore, it appears that PGE(2) released from irradiated microglia is a key mediator of irradiation-induced gliosis or astrocyte phenotype change. These data suggest that radiation-induced microglial activation and resultant production of PGE2 seems to be associated with an underlying cause of inflammatory complications associated with radiation therapy for malignant gliomas.


Assuntos
Astrócitos/efeitos da radiação , Gliose/etiologia , Microglia/efeitos da radiação , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Células Cultivadas , Técnicas de Cocultura , Meios de Cultivo Condicionados , Ciclo-Oxigenase 2/efeitos dos fármacos , Ciclo-Oxigenase 2/metabolismo , Inibidores de Ciclo-Oxigenase/farmacologia , Dinoprostona/metabolismo , Dinoprostona/efeitos da radiação , Ensaio de Desvio de Mobilidade Eletroforética , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/patologia , Immunoblotting , Imuno-Histoquímica , Camundongos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Nitrobenzenos/farmacologia , RNA Mensageiro/análise , Radiação Ionizante , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sulfonamidas/farmacologia
11.
Biochem Biophys Res Commun ; 318(3): 691-7, 2004 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-15144893

RESUMO

The current study examined the potential involvement of phosphatidylinositol 3 phosphate kinase (PI3K) in interferon-gamma (IFN-gamma)-stimulated nitric oxide (NO) generation in BV2 murine microglial cells. We found that LY294002, a PI3K inhibitor, markedly reduced IFN-gamma-induced morphological changes, NO production, and cell death. The inhibitory effect of LY294002 on NO generation may be mediated through specific inhibition of signal transducer and activator-1 (STAT1) and NF-kappaB, which are activated by IFN-gamma. Induction of the mRNA for IFN-gamma-mediated interferon response factor (IRF-1) and inducible protein-10 (IP-10) was not significantly affected by LY294002, indicating that suppression of PI3K may not be sufficient for downregulation of these genes. Although it remains unclear how PI3K signaling is involved in IFN-gamma-mediated inflammatory reactions in the brain, our findings provide some insight into the inflammatory mechanisms of IFN-gamma in the brain and suggest that regulators of the PI3K pathway may act as anti-inflammatory agents in microglia.


Assuntos
Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Interferon gama/antagonistas & inibidores , Microglia/efeitos dos fármacos , Morfolinas/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Interações Medicamentosas , Ativação Enzimática/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Interferon gama/farmacologia , Camundongos , Microglia/enzimologia , Microglia/metabolismo , NF-kappa B/metabolismo , Óxido Nítrico/antagonistas & inibidores , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico Sintase Tipo II , Proteína Oncogênica v-akt , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Proteínas Oncogênicas de Retroviridae/metabolismo , Fator de Transcrição STAT1 , Transdução de Sinais/efeitos dos fármacos , Transativadores/metabolismo , Tirosina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA