Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Cancer Ther ; 18(8): 1430-1438, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31171582

RESUMEN

In clinical N0 (cN0) cases with head and neck squamous cell carcinoma (HNSCC), a treatment selection is still controversial: elective neck dissection or watchful waiting. We focused on sentinel lymph node (SLN)-targeted therapy using the urokinase-type plasminogen activator (uPA)-dependent oncolytic Sendai virus "BioKnife." The objectives of this study were to investigate BioKnife migration into SLNs and elucidate its antitumor effect on lymph node metastases (LNM). We established an orthotopic nude mouse model of HNSCC, with LNM being frequently induced. We inoculated HSC-3-M3, human highly metastatic tongue squamous cell carcinoma cells, in the tongue of the nude mice, and after 2 weeks, we injected BioKnife into the primary tumor. We tracked BioKnife migration into the SLNs by immunostaining, RT-PCR, and an in vivo imaging system. We also examined its antitumor effects and mechanisms through serial section analysis of lymph nodes. GFP reporter expression was clearly visible in the lymph nodes of virus groups, which corresponded to SLNs. Relative GFP mRNA was significantly increased in both the tongues and lymph nodes in the virus groups compared with that in the control group (P < 0.05). Serial section analysis showed that BioKnife infected cancer cells and exhibited significant antitumor effect against LNM compared with the control groups (P < 0.05). We detected apoptosis in LNM infected by BioKnife. BioKnife migrated into SLNs after its injection into the primary tumor and effectively suppressed LNM, suggesting that SLN-targeted therapy using BioKnife has great potential to provide a novel and promising alternative to elective neck dissection in cN0 patients with HNSCC.


Asunto(s)
Viroterapia Oncolítica , Virus Oncolíticos , Virus Sendai , Ganglio Linfático Centinela/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/terapia , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Metástasis Linfática , Ratones , Micrometástasis de Neoplasia , Virus Oncolíticos/genética , Virus Sendai/genética , Ganglio Linfático Centinela/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/diagnóstico por imagen , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Head Neck ; 41(9): 2873-2882, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30969451

RESUMEN

BACKGROUND: This study was designed to assess the therapeutic effect of urokinase-targeted recombinant oncolytic Sendai virus, termed "BioKnife," on anaplastic thyroid carcinoma (ATC). METHODS: Urokinase activity was investigated in human ATC cell lines, and BioKnife cytotoxicity against the cell lines was evaluated in vitro. Orthotopic mouse models of ATC were treated with three intratumoral injections of BioKnife, control virus, or phosphate-buffered saline (PBS) and were observed daily until >20% weight loss occurred. RESULTS: All three ATC cell lines showed a high level of urokinase activity. BioKnife induced urokinase-dependent cell fusion and cytotoxicity in all cell lines. Orthotopic models treated with BioKnife showed significantly prolonged survival compared with models treated with control virus or PBS (BioKnife 41.6 ± 15.0, control virus 17.0 ± 2.9, PBS 17.7 ± 6.3 days). CONCLUSIONS: BioKnife exerted therapeutic effects in orthotopic ATC mouse models. Thus, BioKnife represents a possible treatment option for ATC.


Asunto(s)
Viroterapia Oncolítica , Virus Sendai/fisiología , Carcinoma Anaplásico de Tiroides/terapia , Neoplasias de la Tiroides/terapia , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Animales , Apoptosis , Fusión Celular , Línea Celular Tumoral , Modelos Animales de Enfermedad , Células Gigantes/metabolismo , Ratones Endogámicos BALB C , Carcinoma Anaplásico de Tiroides/metabolismo , Neoplasias de la Tiroides/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Head Neck ; 41(6): 1676-1686, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30620422

RESUMEN

BACKGROUND: The objectives of this study were to demonstrate anti-metastatic effect of BioKnife, uPA activity-dependent oncolytic Sendai virus, after BioKnife treatment for primary tumor, and analyze its mechanisms in a simulated metastasis mouse model of head and neck squamous cell carcinoma (HNSCC). METHODS: We established a simulated metastasis mouse model using a murine HNSCC cell line "SCCVII." We assessed a tumor size and an induction of tumor-specific immunoresponses using cytotoxic T-lymphocyte (CTL) assay, flow cytometry (FCM) in spleen and immunohistochemistry (IHC) in secondary tumor. RESULTS: Secondary tumors were significantly smaller in BioKnife-treated group. CTL activities were significantly improved in BioKnife group. FCM revealed that induction of dendritic cells and CD4+ /CD8+ lymphocytes was significantly higher in BioKnife group. IHC showed that CD8+ lymphocytes invaded secondary tumor. CONCLUSION: Tumor-specific immunoresponses induced by BioKnife has great potential to be a novel, safe, and less invasive option for control and prevention of metastasis.


Asunto(s)
Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/terapia , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/terapia , Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Virus Sendai/fisiología , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Inmunocompetencia , Ratones Endogámicos C3H , Activador de Plasminógeno de Tipo Uroquinasa , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Pharmacol Rep ; 69(3): 485-491, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28324846

RESUMEN

BACKGROUND: Androgenic alopecia (AGA) occurs as a result of the contraction of the anagen phase because of the action of androgens on hair follicles. TGF-ß production from dermal papillae is enhanced by androgens, and growth inhibition of hair-follicle cells is induced by TGF-ß, and the hair cycle progresses from the anagen phase to the catagen phase. We investigated both the in vitro and in vivo potency of the newly identified ALK5 inhibitor TP0427736 {6-[4-(4-methyl-1,3-thiazol-2-yl)-1H-imidazol-5-yl]-1,3-benzothiazole}. METHODS: For in vitro study, kinase inhibitory activity was evaluated with ELISA, and inhibitory activity against TGF-ß-induced Smad2/3 phosphorylation in A549 cells and TGF-ß-induced growth inhibition of human outer root sheath cells were assayed using ELISA. For in vivo study, we used a mouse model that had been synchronized through dorsal hair depilation. RESULTS: TP0427736 inhibited ALK5 kinase activity with an IC50 of 2.72nM; this effect was 300-fold higher than the inhibitory effect on ALK3. In cell-based assays, TP0427736 inhibited Smad2/3 phosphorylation in A549 cells and decreased the growth inhibition of human outer root sheath cells. The topical application of TP0427736 significantly decreased Smad2 phosphorylation in mouse skin, and its repeated application suppressed the shortening of average hair follicle length during the transition from the late anagen phase to the catagen phase. CONCLUSIONS: TP0427736, a potent ALK5 inhibitor with appropriate in vitro and in vivo profiles, may serve as a potential new therapy for AGA. .


Asunto(s)
Alopecia/tratamiento farmacológico , Benzotiazoles/farmacología , Folículo Piloso/efectos de los fármacos , Imidazoles/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Administración Tópica , Alopecia/patología , Animales , Benzotiazoles/administración & dosificación , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/antagonistas & inhibidores , Ensayo de Inmunoadsorción Enzimática , Humanos , Imidazoles/administración & dosificación , Concentración 50 Inhibidora , Ratones , Fosforilación , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , Receptor Tipo I de Factor de Crecimiento Transformador beta , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
5.
Biol Open ; 5(6): 709-19, 2016 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-27170256

RESUMEN

Direct reprogramming is a promising, simple and low-cost approach to generate target cells from somatic cells without using induced pluripotent stem cells. Recently, peripheral blood mononuclear cells (PBMCs) have attracted considerable attention as a somatic cell source for reprogramming. As a cell source, PBMCs have an advantage over dermal fibroblasts with respect to the ease of collecting tissues. Based on our studies involving generation of photosensitive photoreceptor cells from human iris cells and human dermal fibroblasts by transduction of photoreceptor-related transcription factors via retrovirus vectors, we transduced these transcription factors into PBMCs via Sendai virus vectors. We found that retinal disease-related genes were efficiently detected in CRX-transduced cells, most of which are crucial to photoreceptor functions. In functional studies, a light-induced inward current was detected in some CRX-transduced cells. Moreover, by modification of the culture conditions including additional transduction of RAX1 and NEUROD1, we found a greater variety of retinal disease-related genes than that observed in CRX-transduced PBMCs. These data suggest that CRX acts as a master control gene for reprogramming PBMCs into photoreceptor-like cells and that our induced photoreceptor-like cells might contribute to individualized drug screening and disease modeling of inherited retinal degeneration.

6.
Cancer Sci ; 106(10): 1351-61, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26190440

RESUMEN

Neuroblastoma (NB) is the most common extracranial solid tumor that originates from multipotent neural crest cells. NB cell populations that express embryonic stem cell-associated genes have been identified and shown to retain a multipotent phenotype. However, whether somatic reprogramming of NB cells can produce similar stem-cell like populations is unknown. Here, we sought to reprogram NB cell lines using an integration-free Sendai virus vector system. Of four NB cell lines examined, only SH-IN cells formed induced pluripotent stem cell-like colonies (SH-IN 4F colonies) at approximately 6 weeks following transduction. These SH-IN 4F colonies were alkaline phosphatase-positive. Array comparative genomic hybridization analysis indicated identical genomic aberrations in the SH-IN 4F cells as in the parental cells. SH-IN 4F cells had the ability to differentiate into the three embryonic germ layers in vitro, but rather formed NBs in vivo. Furthermore, SH-IN 4F cells exhibited resistance to cisplatin treatment and differentiated into endothelial-like cells expressing CD31 in the presence of vascular endothelial growth factor. These results suggest that SH-IN 4F cells are partially reprogrammed NB cells, and could be a suitable model for investigating the plasticity of aggressive tumors.


Asunto(s)
Plasticidad de la Célula/genética , Reprogramación Celular/genética , Células Madre Pluripotentes Inducidas/citología , Neuroblastoma/genética , Neuroblastoma/patología , Diferenciación Celular , Línea Celular Tumoral , Cisplatino/farmacología , Hibridación Genómica Comparativa , Resistencia a Antineoplásicos , Células Endoteliales/citología , Vectores Genéticos/genética , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Humanos , Células Madre Pluripotentes Inducidas/virología , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/biosíntesis , Factores de Transcripción de Tipo Kruppel/genética , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Factor 3 de Transcripción de Unión a Octámeros/genética , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Proteínas Proto-Oncogénicas c-myc/genética , Factores de Transcripción SOXB1/biosíntesis , Factores de Transcripción SOXB1/genética , Virus Sendai
7.
PLoS One ; 10(1): e0116279, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25569246

RESUMEN

Acquired tracheal stenosis remains a challenging problem for otolaryngologists. The objective of this study was to determine whether the Sendai virus (SeV)-mediated c-myc suppressor, a far upstream element (FUSE)-binding protein (FBP)-interacting repressor (FIR), modulates wound healing of the airway mucosa, and whether it prevents tracheal stenosis in an animal model of induced mucosal injury. A fusion gene-deleted, non-transmissible SeV vector encoding FIR (FIR-SeV/ΔF) was prepared. Rats with scraped airway mucosae were administered FIR-SeV/ΔF through the tracheostoma. The pathological changes in the airway mucosa and in the tracheal lumen were assessed five days after scraping. Untreated animals showed hyperplasia of the airway epithelium and a thickened submucosal layer with extensive fibrosis, angiogenesis, and collagen deposition causing lumen stenosis. By contrast, the administration of FIR-SeV/ΔF decreased the degree of tracheal stenosis (P < 0.05) and improved the survival rate (P < 0.05). Immunohistochemical staining showed that c-Myc expression was downregulated in the tracheal basal cells of the FIR-SeV/ΔF-treated animals, suggesting that c-myc was suppressed by FIR-SeV/ΔF in the regenerating airway epithelium of the injured tracheal mucosa. The airway-targeted gene therapy of the c-myc suppressor FIR, using a recombinant SeV vector, prevented tracheal stenosis in a rat model of airway mucosal injury.


Asunto(s)
Proteínas Proto-Oncogénicas c-myc/genética , Proteínas de Unión al ARN/genética , Proteínas Represoras/genética , Virus Sendai/genética , Estenosis Traqueal/terapia , Animales , Modelos Animales de Enfermedad , Femenino , Terapia Genética , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Inmunohistoquímica , Estimación de Kaplan-Meier , Factores de Empalme de ARN , Ratas , Ratas Sprague-Dawley , Tráquea/metabolismo , Estenosis Traqueal/mortalidad , Estenosis Traqueal/patología
8.
ACS Synth Biol ; 3(2): 83-90, 2014 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-25101815

RESUMEN

Genetic manipulations with mammalian cells often require introduction of two or more genes that have to be in trans-configuration. However, conventional gene delivery vectors have several limitations, including a limited cloning capacity and a risk of insertional mutagenesis. In this paper, we describe a novel gene expression system that consists of two differently marked HAC vectors containing unique gene loading sites. One HAC, 21HAC, is stably propagated during cell divisions; therefore, it is suitable for complementation of a gene deficiency. The other HAC, tet-O HAC, can be eliminated, providing a unique opportunity for transient gene expression (e.g., for cell reprogramming). Efficiency and accuracy of a novel bi-HAC vector system have been evaluated after loading of two different transgenes into these HACs. Based on analysis of transgenes expression and HACs stability in the proof of principle experiments, the combination of two HAC vectors may provide a powerful tool toward gene and cell therapy.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Cromosomas Artificiales Humanos/genética , Terapia Genética , Vectores Genéticos/metabolismo , Animales , Células CHO , Línea Celular Tumoral , Reprogramación Celular , Cricetinae , Cricetulus , Vectores Genéticos/genética , Recombinación Homóloga , Humanos , Neoplasias/terapia
9.
World J Gastroenterol ; 20(15): 4316-28, 2014 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-24764668

RESUMEN

AIM: To investigate a novel therapeutic strategy to target and suppress c-myc in human cancers using far up stream element (FUSE)-binding protein-interacting repressor (FIR). METHODS: Endogenous c-Myc suppression and apoptosis induction by a transient FIR-expressing vector was examined in vivo via a HA-tagged FIR (HA-FIR) expression vector. A fusion gene-deficient, non-transmissible, Sendai virus (SeV) vector encoding FIR cDNA, SeV/dF/FIR, was prepared. SeV/dF/FIR was examined for its gene transduction efficiency, viral dose dependency of antitumor effect and apoptosis induction in HeLa (cervical squamous cell carcinoma) cells and SW480 (colon adenocarcinoma) cells. Antitumor efficacy in a mouse xenograft model was also examined. The molecular mechanism of the anti-tumor effect and c-Myc suppression by SeV/dF/FIR was examined using Spliceostatin A (SSA), a SAP155 inhibitor, or SAP155 siRNA which induce c-Myc by increasing FIR∆exon2 in HeLa cells. RESULTS: FIR was found to repress c-myc transcription and in turn the overexpression of FIR drove apoptosis through c-myc suppression. Thus, FIR expressing vectors are potentially applicable for cancer therapy. FIR is alternatively spliced by SAP155 in cancer cells lacking the transcriptional repression domain within exon 2 (FIR∆exon2), counteracting FIR for c-Myc protein expression. Furthermore, FIR forms a complex with SAP155 and inhibits mutual well-established functions. Thus, both the valuable effects and side effects of exogenous FIR stimuli should be tested for future clinical application. SeV/dF/FIR, a cytoplasmic RNA virus, was successfully prepared and showed highly efficient gene transduction in in vivo experiments. Furthermore, in nude mouse tumor xenograft models, SeV/dF/FIR displayed high antitumor efficiency against human cancer cells. SeV/dF/FIR suppressed SSA-activated c-Myc. SAP155 siRNA, potentially produces FIR∆exon2, and led to c-Myc overexpression with phosphorylation at Ser62. HA-FIR suppressed endogenous c-Myc expression and induced apoptosis in HeLa and SW480 cells. A c-myc transcriptional suppressor FIR expressing SeV/dF/FIR showed high gene transduction efficiency with significant antitumor effects and apoptosis induction in HeLa and SW480 cells. CONCLUSION: SeV/dF/FIR showed strong tumor growth suppression with no significant side effects in an animal xenograft model, thus SeV/dF/FIR is potentially applicable for future clinical cancer treatment.


Asunto(s)
Neoplasias del Colon/terapia , Genes myc , Vectores Genéticos , Factores de Intercambio de Guanina Nucleótido/genética , Proteínas de Unión al ARN/genética , Proteínas Represoras/genética , Virus Sendai/genética , Animales , Apoptosis , Línea Celular Tumoral , Supervivencia Celular , Neoplasias del Colon/metabolismo , ADN Complementario/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Fosforilación , Plásmidos/metabolismo , Piranos/química , Factores de Empalme de ARN , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/metabolismo , Compuestos de Espiro/química
10.
Chem Pharm Bull (Tokyo) ; 61(3): 286-91, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23449197

RESUMEN

7-Methoxy-6-[4-(4-methyl-1,3-thiazol-2-yl)-1H-imidazol-5-yl]-1,3-benzothiazole 11 (TASP0382088) was synthesized and evaluated as transforming growth factor-ß (TGF-ß) type I receptor (also known as activin receptor-like kinase 5 or ALK5) inhibitor. Compound 11, a potent and selective ALK5 inhibitor, exhibited good enzyme inhibitory activity (IC50=4.8 nM) as well as inhibitory activity against TGF-ß-induced Smad2/3 phosphorylation at a cellular level (IC50=17 nM). The introduction of a methoxy group to the benzothiazole ring in 1 and the break up of the planarity between the imidazole ring and the thiazole ring improved the solubility in the lotion base of 11. Furthermore, the topical application of 3% 11 lotion significantly inhibited Smad2 phosphorylation in mouse skin at 8 h after application (71% inhibition, compared with vehicle-treated animals).


Asunto(s)
Alopecia/tratamiento farmacológico , Benzotiazoles/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Animales , Benzotiazoles/química , Femenino , Imidazoles/química , Imidazoles/farmacología , Ratones , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteína Smad2/antagonistas & inhibidores , Proteína Smad2/metabolismo , Proteína smad3/antagonistas & inhibidores , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/metabolismo
11.
Biochem Biophys Res Commun ; 419(4): 754-60, 2012 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-22387544

RESUMEN

Sphingosine 1-phosphate receptor type 1 (S1P(1)) was shown to be essential for vascular maturation during embryonic development and it has been demonstrated that substantial crosstalk exists between S1P(1) and other pro-angiogenic growth factors, such as vascular endothelial growth factor (VEGF) and basic fibroblast growth factor. We developed a novel S1P(1)-selective antagonist, TASP0277308, which is structurally unrelated to S1P as well as previously described S1P(1) antagonists. TASP0277308 inhibited S1P- as well as VEGF-induced cellular responses, including migration and proliferation of human umbilical vein endothelial cells. Furthermore, TASP0277308 effectively blocked a VEGF-induced tube formation in vitro and significantly suppressed tumor cell-induced angiogenesis in vivo. These findings revealed that S1P(1) is a critical component of VEGF-related angiogenic responses and also provide evidence for the efficacy of TASP0277308 for anti-cancer therapies.


Asunto(s)
Neovascularización Fisiológica/efectos de los fármacos , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Sulfonas/farmacología , Triazoles/farmacología , Línea Celular Tumoral , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Lisofosfolípidos/metabolismo , Lisofosfolípidos/farmacología , Neoplasias/irrigación sanguínea , Neovascularización Patológica/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Esfingosina/farmacología , Receptores de Esfingosina-1-Fosfato , Factor A de Crecimiento Endotelial Vascular/farmacología
12.
Exp Hematol ; 39(1): 47-54, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20875838

RESUMEN

OBJECTIVE: The homeobox B4 (HoxB4) gene promotes expansion of hematopoietic stem cells (HSCs). However, frequent development of leukemia in large animals due to retrovirally transduced HoxB4 gene has been reported. To prevent tumorigenesis, we developed a nonintegrating and nonreplicating Sendai virus vector that did not contain the phosphoprotein gene (SeV/ΔP), which enabled clearance of the vector and transgene shortly after transduction. We tested the SeV/ΔP vector expressing the HoxB4 gene (SeV/ΔP/HoxB4) for the ex vivo expansion of human cord blood CD34(+) cells (HSCs) using a sheep in utero transplantation assay. MATERIALS AND METHODS: Human HSCs were ex vivo-expanded by transduction with SeV/ΔP/HoxB4 vector and transplanted into the abdominal cavity of fetal sheep. The engraftment of human HSCs in the lambs was quantitatively evaluated by hematopoietic colony-forming unit assays. RESULTS: After transplantation, the HoxB4-transduced HSCs contributed to longer-period (up to 20 months) repopulation in sheep, and human hematopoietic progenitors were detected more frequently in the bone marrow of the HoxB4 group as compared with the control untreated group (p < 0.05). The expansion of human HSCs with the SeV/ΔP/HoxB4 vector was comparable with previously reported retroviral vectors expressing HoxB4. The SeV/ΔP/HoxB4 vector and the transgene were cleared from the recipient sheep and leukemia was not detected at 20 months post-transplantation. CONCLUSIONS: The SeV/ΔP vector would be suitable for transient expression of HoxB4 in human CD34(+) cells. In addition, the SeV/ΔP vector is free of concern about transgene-related and insertional leukemogenesis and should be safer than retroviral vectors.


Asunto(s)
Vectores Genéticos , Células Madre Hematopoyéticas/citología , Proteínas de Homeodominio/genética , Virus Sendai/genética , Factores de Transcripción/genética , Animales , Secuencia de Bases , Cartilla de ADN , Femenino , Humanos , Reacción en Cadena de la Polimerasa , Embarazo , Ovinos , Trasplante Heterólogo
13.
Sci Rep ; 1: 174, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22355689

RESUMEN

Dendritic cells (DCs) play a crucial role in maintaining the immune system. Though DC-based cancer immunotherapy has been suggested as a potential treatment for various kinds of malignancies, its clinical efficacies are still insufficient in many human trials. Issues that limit the clinical efficacy of DC-based immunotherapy, as well as the difficulty of the industrial production of DCs, are largely due to the limited number of autologous DCs available from each patient. We here established a possible breakthrough, a simple cytokine-based culture method to expand the log-scale order of functional human DCs. Floating cultivation of cord-blood CD34(+) cells under an optimized cytokine cocktail led these progenitor cells to stable log-scale proliferation and to DC differentiation. The expanded DCs had typical features of conventional myeloid DCs in vitro. Therefore, the concept of DC expansion should contribute significantly to the progress of DC immunotherapy.


Asunto(s)
Antígenos CD34/metabolismo , Técnicas de Cultivo de Célula , Citocinas/metabolismo , Células Dendríticas/citología , Regulación de la Expresión Génica , Antígeno CD11c/metabolismo , Cadherinas/metabolismo , Proliferación Celular , Células Cultivadas , Sangre Fetal/citología , Citometría de Flujo , Humanos , Inmunoterapia , Linfocitos T/metabolismo
14.
Neoplasia ; 12(11): 906-14, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21076616

RESUMEN

We recently demonstrated highly efficient antitumor immunity against dermal tumors of B16F10 murine melanoma with the use of dendritic cells (DCs) activated by replication-competent, as well as nontransmissible-type, recombinant Sendai viruses (rSeV), and proposed a new concept, "immunostimulatory virotherapy," for cancer immunotherapy. However, there has been little information on the efficacies of this method: 1) in more clinically relevant situations including metastatic diseases, 2) on other tumor types and other animal species, and 3) on the related molecular/cellular mechanisms. In this study, therefore, we investigated the efficacy of vaccinating DCs activated by fusion gene-deleted nontransmissible rSeV on a rat model of lung metastasis using a highly malignant subline of Dunning R-3327 prostate cancer, AT6.3. rSeV/dF-green fluorescent protein (GFP)-activated bone marrow-derived DCs (rSeV/dF-GFP-DC), consistent with results previously observed in murine DCs. Vaccination of rSeV/dF-GFP-DC was highly effective at preventing lung metastasis after intravenous loading of R-3327 tumor cells, compared with the effects observed with immature DCs or lipopolysaccharide-activated DCs. Interestingly, neither CTL activity nor DC trafficking showed any apparent difference among groups. Notably, rSeV/dF-DCs expressing a dominant-negative mutant of retinoic acid-inducible gene I (RIG-I) (rSeV/dF-RIGIC-DC), an RNA helicase that recognizes the rSeV genome for inducing type I interferons, largely lost the expression of proinflammatory cytokines without any impairment of antitumor activity. These results indicate the essential role of RIG-I-independent signaling on antimetastatic effect induced by rSeV-activated DCs and may provide important insights to DC-based immunotherapy for advanced malignancies.


Asunto(s)
Células Dendríticas/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias de la Próstata/inmunología , Virus Sendai/inmunología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Citocinas/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/trasplante , Citometría de Flujo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inmunoterapia Adoptiva/métodos , Mediadores de Inflamación/metabolismo , Interferón gamma/metabolismo , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Masculino , Mutación , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/terapia , ARN Helicasas/genética , ARN Helicasas/metabolismo , Ratas , Virus Sendai/genética , Transducción de Señal/inmunología
15.
Mol Ther ; 18(10): 1778-86, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20606645

RESUMEN

Glioblastoma multiforme (GM), the most frequent primary malignant brain tumor, is highly invasive due to the expression of proteases, including urokinase-type plasminogen activator (uPA). Here, we show the potential of our new and powerful recombinant Sendai virus (rSeV) showing uPA-specific cell-to-cell fusion activity [rSeV/dMFct14 (uPA2), named "BioKnife"] for GM treatment, an effect that was synergistically enhanced by arming BioKnife with the interferon-ß (IFN-ß) gene. BioKnife killed human GM cell lines efficiently in a uPA-dependent fashion, and this killing was prevented by PA inhibitor-1. Rat gliosarcoma 9L cells expressing both uPA and its functional receptor uPAR (9L-L/R) exhibited high uPA activity on the cellular surface and were highly susceptible to BioKnife. Although parent 9L cells (9L-P) were resistant to BioKnife and to BioKnife expressing IFN-ß (BioKnife-IFNß), cell-cell fusion of 9L-L/R strongly facilitated the expression of IFN-ß, and in turn, IFN-ß significantly accelerated the fusion activity of BioKnife. A similar synergy was seen in a rat orthotopic brain GM model with 9L-L/R in vivo; therefore, these results suggest that BioKnife-IFNß may have significant potential to improve the survival of GM patients in a clinical setting.


Asunto(s)
Glioblastoma/terapia , Interferón beta/metabolismo , Virus Oncolíticos/fisiología , Virus Sendai/fisiología , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Animales , Western Blotting , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Ensayo de Inmunoadsorción Enzimática , Femenino , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Interferón beta/genética , Imagen por Resonancia Magnética , Ratones , Ratones Desnudos , Virus Oncolíticos/genética , Inhibidor 1 de Activador Plasminogénico/farmacología , Ratas , Ratas Endogámicas F344 , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Virus Sendai/genética , Activador de Plasminógeno de Tipo Uroquinasa/genética
16.
Cancer ; 116(15): 3740-8, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20564091

RESUMEN

BACKGROUND: Carbon-ion beam (CIB) treatment is a powerful tool for controlling primary tumors in the clinical setting. However, to date, few clinical or experimental studies have investigated the effects of CIB treatment on tumor recurrence and antitumor immunity. METHODS: A multiple challenge test was performed using syngenic and nude mouse models of a poorly immunogenic squamous cell carcinoma cell line (SCCVII) after CIB treatment at a clinically available dose (77 kiloelectron volts [keV]/microm) to primary tumors. To further examine changes in antitumor immunity in this model, the authors used dendritic cell (DC)-based immunotherapy. RESULTS: In a syngenic model, CIB treatment itself resulted not only in efficient elimination of the primary tumor but also in a dramatic reduction of tumor formation after secondary tumor challenge at a contralateral site (P<.0001). Conversely, CIB treatment eliminated neither the primary nor the secondary tumor in nude mice. This antitumor effect produced by CIB treatment was enhanced significantly by combining it with DC immunotherapy (P=.0007). Combined CIB and DC treatment induced more intense cytolytic activity than CIB in a chromium-release assay. The third challenge tests, which included challenge with a third-party tumor cell line (FM3A) and effector depletion, revealed that the antitumor effects were the results of tumor-specific, long-lasting antitumor immunity through CD8-positive T lymphocytes. CONCLUSIONS: To the authors' knowledge, this is the first demonstration of strong antitumor immunity induced by CIB treatment in a dermal tumor, and this effect was enhanced by combining it with DC-based immunotherapy. The authors concluded that this combination warrants further investigation as a promising modality for the prevention of tumor recurrence.


Asunto(s)
Carbono , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/radioterapia , Inmunoterapia Adoptiva/métodos , Animales , Línea Celular Tumoral , Células Dendríticas , Femenino , Rechazo de Injerto , Iones , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Desnudos , Trasplante de Neoplasias , Linfocitos T Citotóxicos/inmunología
17.
Mol Ther ; 18(6): 1173-82, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20332767

RESUMEN

Gene therapy for cystic fibrosis (CF) is making encouraging progress into clinical trials. However, further improvements in transduction efficiency are desired. To develop a novel gene transfer vector that is improved and truly effective for CF gene therapy, a simian immunodeficiency virus (SIV) was pseudotyped with envelope proteins from Sendai virus (SeV), which is known to efficiently transduce unconditioned airway epithelial cells from the apical side. This novel vector was evaluated in mice in vivo and in vitro directed toward CF gene therapy. Here, we show that (i) we can produce relevant titers of an SIV vector pseudotyped with SeV envelope proteins for in vivo use, (ii) this vector can transduce the respiratory epithelium of the murine nose in vivo at levels that may be relevant for clinical benefit in CF, (iii) this can be achieved in a single formulation, and without the need for preconditioning, (iv) expression can last for 15 months, (v) readministration is feasible, (vi) the vector can transduce human air-liquid interface (ALI) cultures, and (vii) functional CF transmembrane conductance regulator (CFTR) chloride channels can be generated in vitro. Our data suggest that this lentiviral vector may provide a step change in airway transduction efficiency relevant to a clinical programme of gene therapy for CF.


Asunto(s)
Fibrosis Quística/terapia , Terapia Genética , Vectores Genéticos , Lentivirus/genética , Virus Sendai/genética , Proteínas del Envoltorio Viral/genética , Animales , Diferenciación Celular , Línea Celular , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Transducción Genética
18.
J Immunol ; 183(7): 4211-9, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19734206

RESUMEN

We recently demonstrated efficient antitumor immunity against murine tumors using dendritic cells (DCs) activated by recombinant Sendai viruses (rSeVs), and proposed a new concept, "immunostimulatory virotherapy," for cancer immunotherapy. However, there has been little information on the efficacy of this method in preventing metastatic diseases. In this study, we investigated the efficacy of vaccinating DCs activated by fusion gene-deleted nontransmissible rSeV (rSeV/dF) using a murine model of lung metastasis. Bolus and i.v. administration of DCs harboring rSeV/dF-expressing GFP without pulsation of tumor Ag (DC-rSeV/dF-GFP) 2 days before tumor inoculation showed efficient prevention against lung metastasis of c1300 neuroblastoma, but not of RM-9 prostatic cancer. We found that the timing of DC therapy was critical for the inhibition of pulmonary metastasis of RM-9, and that the optimal effect of DCs was seen 28 days before tumor inoculation. Interestingly, the antimetastatic effect was sustained for over 3 mo, even when administered DCs were already cleared from the lung and organs related to the immune system. Although NK cell activity had already declined to baseline at the time of tumor inoculation, Ab-mediated depletion studies revealed that CD4+ cells as well as the presence of, but not the activation of, NK cells were crucial to the prevention of lung metastasis. These results are the first demonstration of efficient inhibition of lung metastasis via bolus administration of virally activated DCs that was sustained and NK/CD4+ cell-dependent, and may suggest a potentially new mechanism of DC-based immunotherapy for advanced malignancies.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Células Dendríticas/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Virus Sendai/inmunología , Animales , Proliferación Celular , Citotoxicidad Inmunológica/genética , Células Dendríticas/virología , Neoplasias Pulmonares/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Masculino , Ratones , Ratones Endogámicos A , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Neuroblastoma/inmunología , Viroterapia Oncolítica , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/virología , Virus Sendai/genética , Factores de Tiempo , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
19.
PLoS One ; 4(8): e6674, 2009 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-19688095

RESUMEN

BACKGROUND: Limitations of the clinical efficacy of dendritic cell (DC)-based immunotherapy, as well as difficulties in their industrial production, are largely related to the limited number of autologous DCs from each patient. We here established a possible breakthrough, a simple and cytokine-based culture method to realize a log-scale order of functional murine DCs (>1,000-fold), which cells were used as a model before moving to human studies. METHODOLOGY/PRINCIPAL FINDINGS: Floating cultivation of lineage-negative hematopoietic progenitors from bone marrow in an optimized cytokine cocktail (FLT3-L, IL-3, IL-6, and SCF) led to a stable log-scale proliferation of these cells, and a subsequent differentiation study using IL-4/GM-CSF revealed that 3-weeks of expansion was optimal to produce CD11b+/CD11c+ DC-like cells. The expanded DCs had typical features of conventional myeloid DCs in vitro and in vivo, including identical efficacy as tumor vaccines. CONCLUSIONS/SIGNIFICANCE: The concept of DC expansion should make a significant contribution to the progress of DC-based immunotherapy.


Asunto(s)
Citocinas/fisiología , Células Dendríticas/citología , Animales , Células Dendríticas/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Prueba de Cultivo Mixto de Linfocitos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL
20.
Exp Hematol ; 37(10): 1250-1257.e1, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19638293

RESUMEN

OBJECTIVE: Hematopoietic stem cells (HSCs) reside in the osteoblastic niche, which consists of osteoblasts. Mesenchymal stromal cells (MSCs) have an ability to differentiate into osteoblasts. Here, using nonhuman primates, we investigated the effects of cotransplantation with MSCs on the engraftment of HSCs after autologous intra-bone marrow transplantation. MATERIALS AND METHODS: From three cynomolgus monkeys, CD34-positive cells (as HSCs) and MSCs were obtained. The former were divided into two equal aliquots and each aliquot was genetically marked with a distinctive retroviral vector to track the in vivo fate. Each HSC aliquot with or without MSCs was autologously injected into the bone marrow (BM) cavity of right or left side, enabling the comparison of in vivo fates of the two HSC grafts in the same body. RESULTS: In the three monkeys, CD34(+) cells transplanted with MSCs engrafted 4.4, 6.0, and 1.6 times more efficiently than CD34(+) cells alone, as assessed by BM colony polymerase chain reaction. In addition, virtually all marked cells detected in the peripheral blood were derived from the cotransplantation aliquots. Notably, colony-forming units derived from the cotransplantation aliquots were frequently detected in BM distant sites from the injection site, implying that cotransplantation with MSCs also restored the ability of gene-marked HSCs to migrate and achieve homing in the distant BM. CONCLUSION: Cotransplantation with MSCs would improve the efficacy of transplantation of gene-modified HSCs in primates, with enhanced engraftment in BM as well as increased chimerism in peripheral blood through migration and homing.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Animales , Linaje de la Célula , Células Cultivadas/trasplante , Genes Reporteros , Vectores Genéticos/análisis , Supervivencia de Injerto , Macaca fascicularis , Osteoblastos/citología , Células del Estroma/trasplante , Acondicionamiento Pretrasplante , Trasplante Autólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...