Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 13(1): 13033, 2023 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-37563266

RESUMO

Emerging SARS-CoV-2 Omicron variants are highly contagious with enhanced immune escape mechanisms against the initially approved COVID-19 vaccines. Therefore, we require stable alternative-platform vaccines that confer protection against newer variants of SARS-CoV-2. We designed an Omicron B.1.1.529 specific DNA vaccine using our DNA vaccine platform and evaluated the humoral and cellular immune responses. SD rats intradermally administered with Omicron-specific DNA vaccine via pyro-drive jet injector (PJI) thrice at 2-week intervals elicited high antibody titers against the Omicron subvariants as well as the ancestral strain. Indeed, the Omicron B.1.1.529-specific antibody titer and neutralizing antibody were higher than that of other strains. Longitudinal monitoring indicated that anti-spike (ancestral and Omicron) antibody titers decreased toward 30 weeks after the first vaccination dose. However, neutralization activity remained unaltered. Germinal center formation was histologically detected in lymph nodes in rats immunized with Omicron DNA vaccine. Ancestral spike-specific immune cell response was slightly weaker than Omicron spike-specific response in splenocytes with Omicron-adapted DNA vaccine, evaluated by ELISpot assay. Collectively, our findings suggest that Omicron targeting DNA vaccines via PJI can elicit robust durable antibody production mediated by germinal center reaction against this new variant as well as partially against the spike protein of other SARS-CoV-2 variants.


Assuntos
COVID-19 , Vacinas de DNA , Animais , Humanos , Ratos , Ratos Sprague-Dawley , Anticorpos Neutralizantes , Vacinas contra COVID-19 , SARS-CoV-2 , COVID-19/prevenção & controle , Centro Germinativo , Anticorpos Antivirais
2.
FEBS Open Bio ; 13(3): 478-489, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36651034

RESUMO

Cell-cell fusion involves the fusion of somatic cells into a single hybrid cell. It is not only a physiological process but also an important cell engineering technology which can be applied to various fields, such as regenerative medicine, antibody engineering, genetic engineering, and cancer therapy. There are three major methods of cell fusion: electrical cell fusion, polyethylene glycol (PEG) cell fusion, and virus-mediated cell fusion. Although PEG cell fusion is the most economical approach and does not require expensive instrumentation, it has a poor fusion rate and induces a high rate of cell cytotoxicity. To improve the fusion rate of the PEG method, we combined it with the pyro-drive jet injector (PJI). PJI provides instant pressure instead of cell agitation to increase the probability of cell-to-cell contact and shorten the distance between cells in the process of cell fusion. Here, we report that this improved fusion method not only decreased cell cytotoxicity during the fusion process, but also increased fusion rate compared with the conventional PEG method. Furthermore, we tested the functionality of cells fused using the PJI-PEG method and found them to be comparable to those fused using the conventional PEG method in terms of their application for dendritic cell (DC)-tumor cell fusion vaccine production; in addition, the PJI-PEG method demonstrated excellent performance in hybridoma cell preparation. Taken together, our data indicate that this method improves cell fusion efficiency as compared to the PEG method and thus has the potential for use in various applications that require cell fusion technology.


Assuntos
Engenharia Genética , Polietilenoglicóis , Polietilenoglicóis/farmacologia , Fusão Celular
3.
Sci Rep ; 12(1): 20923, 2022 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-36463322

RESUMO

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has led to a global pandemic. New technologies have been utilized to develop several types of vaccines to prevent the spread of SARS-CoV-2 infection, including mRNA vaccines. Our group previously developed an effective DNA-based vaccine. However, emerging SARS-CoV-2 variants of concern (VOCs), such as the delta variant, have escaped mutations against vaccine-induced neutralizing antibodies. This suggests that modified vaccines accommodating VOCs need to be developed promptly. Here, we first modified the current DNA vaccine to enhance antigenicity. Compared with the parental DNA vaccine, the modified version (GP∆-DNA vaccine) induced rapid antibody production. Next, we updated the GP∆-DNA vaccine to spike glycoprotein of the delta variant (GP∆-delta DNA vaccine) and compared the efficacy of different injection routes, namely intramuscular injection using a needle and syringe and intradermal injection using a pyro-drive jet injector (PJI). We found that the levels of neutralizing antibodies induced by the intradermal PJI injection were higher than intramuscular injection. Furthermore, the PJI-injected GP∆-delta DNA vaccine effectively protected human angiotensin-converting enzyme 2 (hACE2) knock-in mice from delta-variant infection. These results indicate that the improved DNA vaccine was effective against emerging VOCs and was a potential DNA vaccine platform for future VOCs or global pandemics.


Assuntos
COVID-19 , Vacinas de DNA , Humanos , Animais , Camundongos , SARS-CoV-2/genética , Imunidade Humoral , Vacinas de DNA/genética , COVID-19/prevenção & controle , Anticorpos Neutralizantes
4.
Immunol Med ; 45(4): 251-264, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36001011

RESUMO

There is an urgent need to stop the coronavirus disease 2019 (COVID-19) pandemic through the development of efficient and safe vaccination methods. Over the short term, plasmid DNA vaccines can be developed as they are molecularly stable, thus facilitating easy transport and storage. pVAX1-SARS-CoV2-co was designed for the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) S protein. The antibodies produced led to immunoreactions against the S protein, an anti-receptor-binding-domain, and a neutralizing action of the pVAX1-SARS-CoV2-co, as previously confirmed. To promote the efficacy of the pVAX1-SARS-CoV2-co vaccine a pyro-drive jet injector (PJI) was used. An intradermally adjusted PJI demonstrated that the pVAX1-SARS-CoV2-co vaccine injection caused a high production of anti-S protein antibodies, triggered immunoreactions, and neutralized the actions against SARS-CoV-2. A high-dose pVAX1-SARS-CoV2-co intradermal injection using PJI did not cause any serious disorders in the rat model. A viral challenge confirmed that intradermally immunized mice were potently protected from COVID-19. A pVAX1-SARS-CoV2-co intradermal injection using PJI is a safe and promising vaccination method for overcoming the COVID-19 pandemic.


Assuntos
COVID-19 , Vacinas de DNA , Vacinas Virais , Camundongos , Humanos , Ratos , Animais , COVID-19/prevenção & controle , Pandemias/prevenção & controle , SARS-CoV-2 , RNA Viral , Roedores , Anticorpos Antivirais , Vacinação/métodos , Formação de Anticorpos , Plasmídeos
5.
Mol Ther Oncolytics ; 20: 175-186, 2021 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-33575480

RESUMO

Breast cancer has a high risk of metastasis; however, no effective treatment has been established. We developed a novel immunotherapy for breast cancer to enhance cytotoxic T lymphocytes against cancer cells using N1-type neutrophils with anti-tumor properties. For this purpose, we combined CXCL2 (CXC chemokine ligand 2) plasmid DNA with inactivated Sendai virus (hemagglutinating virus of Japan)-envelope (HVJ-E). The combination of CXCL2 DNA and HVJ-E (C/H) suppressed the growth of murine breast cancers in orthotopic syngeneic models by enhancing cytotoxic T lymphocytes and inhibited lung metastasis of breast cancer from primary lesions. N1-type neutrophils (CD11b+ Ly6G+ FAS+) increased in the tumor microenvironment with C/H treatment, and tumor suppression and cytotoxic T lymphocyte activation from C/H was blocked after administrating anti-neutrophil antibodies, which indicates the role of N1-type neutrophils in cancer immunotherapy. We also demonstrated that the anti-tumor activities of C/H treatment were enhanced by the administration of anti-PD-1 antibodies through neutrophil-mediated cytotoxic T lymphocyte activation. Thus, the triple combination of C/H and anti-PD-1 antibody C/H treatment may provide an improvement in cancer immunotherapy.

6.
RNA ; 27(2): 163-173, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33177188

RESUMO

Small interfering RNAs (siRNAs) can be utilized not only as functional biological research tools but also as therapeutic agents. For the clinical use of siRNA as drugs, various chemical modifications have been used to improve the activity of siRNA drugs, and further chemical modifications are expected to improve the utility of siRNA therapeutics. As the 5' nucleobase of the guide strand affects the interaction between an siRNA and AGO2 and target cleavage activity, structural optimization of this specific position may be a useful strategy for improving siRNA activity. Here, using the in silico model of the complex between human AGO2 MID domain and nucleoside monophosphates, we screened and synthesized an original adenine-derived analog, 6-(3-(2-carboxyethyl)phenyl)purine (6-mCEPh-purine), that fits better than the natural nucleotide bases into the MID domain of AGO2. Introduction of the 6-mCEPh-purine analog at the 5'-end of the siRNA guide strand significantly enhanced target knockdown activity in both cultured cell lines and in vivo animal models. Our findings can help expand strategies for rationally optimizing siRNA activity via chemical modifications of nucleotide bases.


Assuntos
Adenina/farmacologia , Proteínas Argonautas/genética , Interferência de RNA/efeitos dos fármacos , RNA de Cadeia Dupla/genética , RNA Interferente Pequeno/agonistas , Complexo de Inativação Induzido por RNA/agonistas , Adenina/análogos & derivados , Adenina/síntese química , Monofosfato de Adenosina/química , Monofosfato de Adenosina/metabolismo , Animais , Apolipoproteína B-100/antagonistas & inibidores , Apolipoproteína B-100/sangue , Apolipoproteína B-100/química , Apolipoproteína B-100/genética , Proteínas Argonautas/metabolismo , Pareamento de Bases , Sequência de Bases , Sítios de Ligação , Colesterol/sangue , Células HeLa , Humanos , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Masculino , Metilação , Camundongos , Camundongos Knockout , Modelos Moleculares , Ligação Proteica , RNA de Cadeia Dupla/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Complexo de Inativação Induzido por RNA/genética , Complexo de Inativação Induzido por RNA/metabolismo , Uridina Monofosfato/química , Uridina Monofosfato/metabolismo
7.
Lasers Med Sci ; 36(4): 743-750, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32592133

RESUMO

To enhance the potency of photosensitizer, we developed a novel photosensitizer, Laserphyrin®-HVJ-E (L-HVJ-E), by incorporating talaporfin sodium (Laserphyrin®, Meiji Seika Pharma) into hemagglutinating virus of Japan envelope (HVJ-E). In this study, we examined the optimal Laserphyrin® concentration for preparation of Laserphyrin®-HVJ-E which had photocytotoxicity and maintained direct cytotoxicity derived from HVJ-E. Then, potency of Laserphyrin®-HVJ-E and Laserphyrin® were compared in vitro using castration-resistant prostate cancer cell line (PC-3). A laser diode (L660P120, Thorlabs, USA) with a wavelength of 664 nm was used for light activation of Laserphyrin®, which corresponds to an absorption peak of Laserphyrin® and provides a high therapeutic efficiency. The photocytotoxicity and direct cytotoxicity of Laserphyrin®-HVJ-E prepared using various Laserphyrin® concentrations were evaluated using PC-3 cell in vitro. We categorized the treatment groups as Group 1: 50 µL of D-MEM treatment group, Group 2: HVJ-E treatment group, Group 3: Laserphyrin®-HVJ-E treatment group, and Group 4: Laserphyrin® treatment group. Group 3 was subjected to different concentrations of Laserphyrin®-HVJ-E suspension, and all groups were subjected to different incubation periods (24, 48 h), (30 min, 1 h, or 3 h,) respectively, without and after PDT. Laserphyrin®-HVJ-E prepared using 15 mM Laserphyrin® had high photocytotoxicity and maintained HVJ-E's ability to induce direct cytotoxicity. Therapeutic effect of Laserphyrin®-HVJ-E was substantially equivalent to that of Laserphyrin® alone even at half Laserphyrin® concentration. By utilizing Laserphyrin®-HVJ-E, PDT could be performed with lower Laserphyrin® concentration. In addition, Laserphyrin®-HVJ-E showed higher potency than Laserphyrin® by combining cytotoxicities of HVJ-E and PDT.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Fotoquimioterapia , Porfirinas/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Vírion/fisiologia , Animais , Antineoplásicos/uso terapêutico , Humanos , Lasers Semicondutores , Masculino , Células PC-3 , Fármacos Fotossensibilizantes/farmacologia , Porfirinas/farmacologia , Vírus Sendai/efeitos dos fármacos
9.
Cancer Gene Ther ; 27(6): 498-508, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31383952

RESUMO

Inactivated Sendai virus particle (or hemagglutinating virus of Japan envelope; HVJ-E) has been previously reported to possess antitumour properties that activate antitumour immunity. Two glycoproteins, fusion (F) and hemagglutinin-neuraminidase (HN), are present on the surface of HVJ-E. HN is necessary for binding to receptors such as acidic gangliosides, and F induces membrane fusion by associating with membrane lipids. We previously reported that liposomes reconstituted with F but not HN showed antitumour activity by inducing IL-6 secretion in dendritic cells (DCs), suggesting that F protein is capable of eliciting antitumour activity. Here, we attempted to deliver F gene into tumour tissue in mice by electroporation and demonstrated that F gene therapy retarded tumour growth, increased CD4+ and CD8+ T-cell infiltration into tumours and induced tumour-specific IFN-γ T-cell response. However, neutralisation of IL-6R signalling did not impact F plasmid-mediated antitumour effect. Instead, we found that F plasmid treatment resulted in a significant increase in the secretion of the chemokine RANTES (regulated upon activation, normal T cell expressed and secreted) by tumour-infiltrating T cells. Neutralising antibody against RANTES abolished the antitumour effect of F plasmid treatment in a dose-dependent manner. Thus, F gene therapy may show promise as a novel therapeutic for single or combined cancer immunotherapy.


Assuntos
Imunoterapia/métodos , Melanoma Experimental/terapia , Vírus Sendai/imunologia , Animais , Feminino , Fusão Gênica , Linfócitos do Interstício Tumoral/imunologia , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmídeos/administração & dosagem , Plasmídeos/genética , Plasmídeos/imunologia , Vírus Sendai/genética , Linfócitos T/imunologia , Transfecção
10.
AAPS PharmSciTech ; 21(1): 19, 2019 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-31820256

RESUMO

DNA vaccination can be applied to the treatment of various infectious diseases and cancers; however, technical difficulties have hindered the development of an effective delivery method. The efficacy of a DNA vaccine depends on optimal antigen expression by the injected plasmid DNA. The pyro-drive jet injector (PJI) is a novel system that allows for adjustment of injection depth and may, thus, provide a targeted delivery approach for various therapeutic or preventative compounds. Herein, we investigated its potential for use in delivering DNA vaccines. This study evaluated the optimal ignition powder dosage, as well as its delivery effectiveness in both rat and mouse models, while comparing the results of the PJI with that of a needle syringe delivery system. We found that the PJI effectively delivered plasmid DNA to intradermal regions in both rats and mice. Further, it efficiently transfected plasmid DNA directly into the nuclei, resulting in higher protein expression than that achieved via needle syringe injection. Moreover, results from animal ovalbumin (OVA) antigen induction models revealed that animals receiving OVA expression plasmids (pOVA) via PJI exhibited dose-dependent (10 µg, 60 µg, and 120 µg) production of anti-OVA antibodies; while only low titers (< 1/100) of OVA antibodies were detected when 120 µg of pOVA was injected via needle syringe. Thus, PJI is an effective, novel method for delivery of plasmid DNA into epidermal and dermal cells suggesting its promise as a tool for DNA vaccination.


Assuntos
Injeções/instrumentação , Vacinas de DNA/imunologia , Administração Cutânea , Animais , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Agulhas , Ratos , Ratos Sprague-Dawley , Vacinação/métodos , Vacinas de DNA/administração & dosagem
11.
Laser Ther ; 28(4): 245-256, 2019 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-32255916

RESUMO

BACKGROUND: Photodynamic therapy (PDT), a minimally invasive cancer treatment involving the activation of photosensitizer by a specific wavelength of light, is considered to be a promising treatment option for drug-resistant prostate cancer. Hemagglutinating virus of Japan envelope (HVJ-E) has the potential to serve as a highly effective cancer therapy through selective drug delivery and enhancement of the anti-tumor immune response. OBJECTIVES: To improve therapeutic efficacy and selective accumulation of photosensitizer into tumor cells, we developed a novel photosensitizer, Laserphyrin®-HVJ-E (L-HVJ-E), by incorporating talaporfin sodium (Laserphyrin®, Meiji Seika Pharma) into HVJ-E. MATERIALS AND METHODS: The therapeutic effect of PDT with Laserphyrin® or L-HVJ-E was evaluated in the human prostate cancer cell line PC-3 in vitro. The subcellular localizations of Laserphyrin® and L-HVJ-E were observed by confocal microscopy. Apoptosis or necrosis following PDT was detected by annexin V-fluorescein/propidium iodide double staining. RESULTS: The cytotoxic effect of Laserphyrin®- and L-HVJ-E-mediated PDT were determined by evaluating cell survival rate and production of reactive oxygen species. The cytotoxicity of L-HVJ-E-mediated PDT was dependent on drug concentration and light dose. Laserphyrin® and L-HVJ-E gradually entered cells as incubation time increased, and both agents tended to be distributed in lysosomes rather than mitochondria. Time and dose dependent increase in ROS production was observed, and induction of both apoptotic and necrotic cell death was confirmed. CONCLUSIONS: Laserphyrin® and L-HVJ-E were distributed mainly in lysosomes and induced cell death by both apoptosis and necrosis. Furthermore, L-HVJ-E-mediated PDT effectively killed cultured PC-3 cells and exerted higher photocytotoxicity than Laserphyrin®-mediated PDT.

12.
Photodiagnosis Photodyn Ther ; 20: 238-245, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29079353

RESUMO

BACKGROUND: Subcellular localization of a photosensitizer is known to determine the therapeutic efficacy of photodynamic therapy (PDT). Cell membrane is an optimal target that promises an effective treatment outcome. OBJECTIVES: We previously developed a novel photosensitizer named porphyrus envelope (PE) by combining hemagglutinating virus of Japan envelope (HVJ-E) with lipidated protoporphyrin IX (PpIX lipid). In the current study, the cellular localization of PE and its ability to induce multiple anti-tumor effect were characterized. MATERIALS AND METHODS: The localization and uptake of PpIX lipid in cells were evaluated with confocal laser scanning microscopy and a cell-based fluorescent assay, respectively. The ability of PE to suppress the migration and proliferation of cancer cells was assessed using a scratch-wound assay. The synergistic effect of PDT and HVJ-E treatment was evaluated using an in vitro experiment with PC-3 cells. RESULTS: PE localized along the cell membrane and PpIX lipid accumulated selectively in the prostate cancer cells within 10min. Also, PE maintained the ability to undergo fusion and induce cancer cell death even after light irradiation at the dose for PDT. Incubation with PE resulted in delayed migratory and proliferative activity of PC-3 cells. PE-mediated PDT was twice as effective when cells were further incubated with PE following PDT. CONCLUSIONS: PE allows rapid drug delivery targeting the cell membrane. Because the cytotoxicity of HVJ-E was maintained, synergistic effect of HVJ-E and the photochemical reactions resulted in highly effective killing of prostate cancer cells in vitro and thus represents a promising treatment for prostate cancer.


Assuntos
Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia , Protoporfirinas/farmacologia , Vírus Sendai , Proteínas do Envelope Viral/química , Ácido Aminolevulínico/farmacologia , Morte Celular , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos/métodos , Humanos , Microscopia Confocal , Fármacos Fotossensibilizantes/administração & dosagem , Protoporfirinas/administração & dosagem
13.
Cancer Sci ; 108(12): 2333-2341, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28945328

RESUMO

We have already reported that the inactivated Sendai virus (hemagglutinating virus of Japan; HVJ) envelope (HVJ-E) has multiple anticancer effects, including induction of cancer-selective cell death and activation of anticancer immunity. The HVJ-E stimulates dendritic cells to produce cytokines and chemokines such as ß-interferon, interleukin-6, chemokine (C-C motif) ligand 5, and chemokine (C-X-C motif) ligand 10, which activate both CD8+ T cells and natural killer (NK) cells and recruit them to the tumor microenvironment. However, the effect of HVJ-E on modulating the sensitivity of cancer cells to immune cell attack has yet to be investigated. In this study, we found that HVJ-E induced the production of intercellular adhesion molecule-1 (ICAM-1, CD54), a ligand of lymphocyte function-associated antigen 1, in several cancer cell lines through the activation of nuclear factor-κB downstream of retinoic acid-inducible gene I and the mitochondrial antiviral signaling pathway. The upregulation of ICAM-1 on the surface of cancer cells increased the sensitivity of cancer cells to NK cells. Knocking out expression of ICAM-1 in MDA-MB-231 cells using the CRISPR/Cas9 method significantly reduced the killing effect of NK cells on ICAM-1-depleted MDA-MB-231 cells. In addition, HVJ-E suppressed tumor growth in MDA-MB-231 tumor-bearing SCID mice, and the HVJ-E antitumor effect was impaired when NK cells were depleted by treatment with the anti-asialo GM1 antibody. Our findings suggest that HVJ-E enhances NK cell sensitivity against cancer cells by increasing ICAM-1 expression on the cancer cell surface.


Assuntos
Imunoterapia/métodos , Molécula 1 de Adesão Intercelular/biossíntese , Células Matadoras Naturais/imunologia , Neoplasias/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Linhagem Celular Tumoral , Xenoenxertos , Humanos , Camundongos , Camundongos SCID , Vírus Sendai , Regulação para Cima , Proteínas do Envelope Viral/farmacologia
14.
Oncotarget ; 8(21): 35048-35060, 2017 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-28456782

RESUMO

Salmonella can target to tumor microenvironments after systemic treatment. The hemagglutinating virus of Japan-envelope (HVJ-E) induced apoptosis in tumor cells without toxicity in normal cells. Current HVJ-E therapeutic strategies, aimed at using HVJ-E for intratumor treatment, have shown great promise in animal models but have achieved only limited systemic treatment. The purpose of this study was to investigate the modulation of the anti-tumor efficiency of HVJ-E by coating the particles with poly (allylamine hydrochloride) (PAH), designated as P-HVJ-E. Treatment with P-HVJ-E resulted in decreased hemagglutinating activity and maintained tumor cell-selective apoptosis and anti-tumor immunity. The use of Salmonella as a coating for P-HVJ-E (PHS) enhanced the antitumor activity and maintained the tumor-targeting activity. Treatment with PHS resulted in delayed tumor growth in tumor-bearing mice. Furthermore, a Western blot assay of the tumors revealed that HVJ-E targeted to the tumor after systemic treatment with PHS. These results indicate that Salmonella coating viral particles may provide a new approach for tumor therapy.


Assuntos
Neoplasias Experimentais/tratamento farmacológico , Poliaminas/química , Salmonella/fisiologia , Vírus Sendai/metabolismo , Proteínas do Envelope Viral/administração & dosagem , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , Camundongos , Microambiente Tumoral , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Oncotarget ; 7(27): 42195-42207, 2016 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-27259252

RESUMO

The tumor microenvironment (TME) fosters tumors by attenuating anti-tumor immunity, reinforcing tumor cell survival and increasing angiogenesis. Among the constituents of the TME, here, we focused on tumor-associated neutrophils (TANs). First, we found that the combination of poly I:C and inactivated Sendai virus particles (hemagglutinating virus of Japan envelope; HVJ-E) synergistically suppressed tumor growth in the B16-F10 melanoma mouse model. In this model, poly I:C contributed to the recruitment of CD11b+Ly6G+ neutrophils to the TME, and co-injection of poly I:C and HVJ-E increased CD11b+Ly6G+FAS+ TAN in the TME. Depletion of neutrophils abolished the synergistic anti-tumor effect of HVJ-E and poly I:C in B16-F10 tumors. We revealed that C-X-C motif chemokine ligand 2 (CXCL2) is produced in the TME by poly I:C, but HVJ-E enhanced neutrophil infiltration of the TME does not occur. An anti-CXCL2 antibody inhibited the tumor suppression by HVJ-E+poly I:C. HVJ-E in combination with recombinant CXCL2 protein or CXCL2 pDNA suppressed mouse melanoma by increasing cytotoxic T lymphocyte activity against B16-F10 melanoma, which was abolished by an anti-Ly6G antibody. HVJ-E directly and indirectly increased FAS and ICAM-1 expression in cultured bone marrow-derived naïve neutrophils. Thus, HVJ-E activates anti-tumor immunity via anti-tumorigenic neutrophils in the TME. An HVJ-E vector containing the CXCL2 gene may be applicable as a novel cancer gene therapy strategy.


Assuntos
Melanoma Experimental/virologia , Neutrófilos/imunologia , Neutrófilos/virologia , Linfócitos T/imunologia , Microambiente Tumoral , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Quimiocina CXCL2/metabolismo , Ligantes , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Terapia Viral Oncolítica , Poli I-C/imunologia , Vírus Sendai , Transdução de Sinais , Baço/citologia
16.
Sci Rep ; 6: 25077, 2016 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-27117228

RESUMO

Tumours constitute unique microenvironments where various blood cells and factors are exposed as a result of leaky vasculature. In the present study, we report that thrombin enrichment in B16F10 melanoma led to platelet aggregation, and this property was exploited to administer an anticancer cytokine, interferon-gamma induced protein 10 (IP10), through the formation of a platelet-IP10 complex. When intravenously infused, the complex reached platelet microaggregates in the tumour. The responses induced by the complex were solely immune-mediated, and tumour cytotoxicity was not observed. The complex suppressed the growth of mouse melanoma in vivo, while both platelets and the complex suppressed the accumulation of FoxP3(+) regulatory T cells in the tumour. These results demonstrated that thrombin-dependent platelet aggregation in B16F10 tumours defines platelets as a vector to deliver anticancer cytokines and provide specific treatment benefits.


Assuntos
Coagulação Sanguínea , Plaquetas/metabolismo , Quimiocina CXCL10/administração & dosagem , Melanoma/terapia , Agregação Plaquetária , Trombina/metabolismo , Animais , Modelos Animais de Doenças , Infusões Intravenosas , Melanoma/patologia , Camundongos Endogâmicos C57BL
17.
Mol Ther ; 24(1): 135-45, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26548591

RESUMO

Inactivated Sendai virus (hemagglutinating virus of Japan; HVJ) envelope (HVJ-E) induces anticancer immunity and cancer cell-selective apoptosis through the recognition of viral RNA genome fragments by retinoic acid-inducible gene-I (RIG-I). Here, we discovered that the "copy-back" type of defective-interfering (DI) particles that exist in the Cantell strain of HVJ induced the human PC3 prostate cancer cell death more effectively than the Sendai/52 strain or Cantell strain, which contain fewer DI particles. DI particle genomic RNA (~550 bases) activated proapoptotic genes such as Noxa and/or TNF-related apoptosis-inducing ligand (TRAIL) in human prostate cancer cells to induce cancer cell-selective apoptosis. DI particle-derived RNA was synthesized by in vitro transcription (in vitro transcribed (IVT)-B2). IVT-B2 RNA, which has a double-stranded region in its secondary structure, promoted a stronger anticancer effect than IVT-HN RNA, which does not have a double-stranded region in its secondary structure. The intratumoral transfection of IVT-B2 significantly reduced the volume of a human prostate tumor and induced tumor cell apoptosis in the xenograft mouse model. Moreover, the involvement of natural killer (NK) cells in IVT-B2-RNA-induced anticancer effects was also suggested. These findings provide a novel nucleic acid medicine for the treatment of cancer.


Assuntos
Vírus Defeituosos/genética , Neoplasias da Próstata/terapia , RNA de Cadeia Dupla/administração & dosagem , RNA Viral/administração & dosagem , Vírus Sendai/genética , Animais , Apoptose , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Proteínas Quinases Ativadas por Mitógeno , Terapia Viral Oncolítica , Neoplasias da Próstata/genética , Proteínas Serina-Treonina Quinases , RNA de Cadeia Dupla/química , RNA de Cadeia Dupla/farmacologia , RNA Viral/química , RNA Viral/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Mol Ther ; 22(12): 2046-2055, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25023327

RESUMO

Tumor microenvironments include a number of fibrin clots due to the microbleeding caused by cancer cell invasion into blood vessels, which suggests the potential utility of a platelet vector for systemic cancer treatment. We previously reported that inactivated Sendai virus (hemagglutinating virus of Japan; HVJ) envelope (HVJ-E) activates anti-tumor immunity and induces cancer cell-selective apoptosis. The hemagglutination activity that blocks the systemic administration of HVJ-E was dramatically attenuated by incorporation into platelets. Platelets incorporating HVJ-E (PH complex) were then injected into the tail veins of B16F10 melanoma-bearing mice. The PH complex primarily accumulated in tumor tissues and caused the significant accumulation of various immune cells in the tumor bed. Injections of the PH complex to the melanoma-bearing mouse significantly reduced the tumor size, and the tumor growth was ultimately arrested. Secretion of the chemokine regulated upon activation normal T-expressed and presumably secreted (RANTES) was upregulated following PH stimulation. The RANTES-depletion in melanoma-bearing mice significantly attenuated the cytotoxic T lymphocyte activity and led to a dramatic abrogation of the mouse melanoma suppression induced by the PH complex. Thus, a platelet vector incorporating viral particles, a Trojan horse for cancer treatment, will provide a new approach for cancer therapy using oncolytic viruses.


Assuntos
Plaquetas/virologia , Hemaglutininas Virais/imunologia , Melanoma/imunologia , Células-Tronco Mesenquimais/citologia , Vírus Oncolíticos/fisiologia , Vírus Sendai/fisiologia , Animais , Plaquetas/citologia , Plaquetas/imunologia , Linhagem Celular , Feminino , Imunoterapia , Melanoma/terapia , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Terapia Viral Oncolítica , Inativação de Vírus
19.
Clin Cancer Res ; 18(22): 6271-83, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23014529

RESUMO

PURPOSE: The treatment of cancer with oncolytic viruses primarily depends on the selective viral replication in cancer cells. However, a replication-incompetent hemagglutinating virus of Japan (HVJ; Sendai virus) envelope (HVJ-E) suppresses the growth of human cancer cells as effectively as replication-competent live HVJ without producing toxic effects in nonmalignant cells. Here, we analyze the molecular mechanism of the oncolytic activity of HVJ-E. EXPERIMENTAL DESIGN: The molecules responsible for HVJ-E-induced cancer cell death were elucidated in prostate cancer cell lines, and the effect of HVJ-E on orthotopic prostate cancers was evaluated in nonobese diabetic-severe combined immunodeficient (NOD-SCID) mice. RESULTS: The liposome-mediated transfer of viral RNA genome fragments from HVJ-E suppressed the viability of prostate cancer cells but not the viability of the noncancerous prostate epithelium. Knockdown experiments using siRNAs showed that the cancer cell-selective killing induced by HVJ-E was mediated by retinoic acid-inducible gene I (RIG-I) and mitochondrial antiviral signaling protein (MAVS). Downstream of the RIG-I/MAVS pathway, both TNF-related apoptosis-inducing ligand (TRAIL) and Noxa were upregulated by HVJ-E in the castration-resistant prostate cancer cell line PC3 but not in the noncancerous prostate epithelial cell line PNT2. TRAIL siRNA but not Noxa siRNA significantly inhibited HVJ-E-induced cell death in PC3 cells. However, Noxa siRNA effectively suppressed HVJ-E-induced cell death in DU145 cells, another castration-resistant prostate cancer cell line, in which Noxa but not TRAIL was upregulated by HVJ-E. Furthermore, the orthotopic prostate cancers were dramatically eradicated in immunodeficient mice injected with HVJ-E. CONCLUSION: The RIG-I/MAVS signaling pathway represents an attractive target for cancer therapy.


Assuntos
Neoplasias da Próstata/terapia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Vírus Sendai/fisiologia , Transdução de Sinais , Ligante Indutor de Apoptose Relacionado a TNF/genética , Regulação para Cima , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Genoma Viral , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Vírus Oncolíticos/fisiologia , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Viral/imunologia , Receptores Imunológicos , Vírus Sendai/genética , Vírus Sendai/imunologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Carga Tumoral , Vírion/fisiologia , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
20.
J Cell Mol Med ; 16(7): 1629-39, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21812915

RESUMO

We previously identified a novel angiogenic peptide, AG30, with antibacterial effects that could serve as a foundation molecule for the design of wound-healing drugs. Toward clinical application, in this study we have developed a modified version of the AG30 peptide characterized by improved antibacterial and angiogenic action, thus establishing a lead compound for a feasibility study. Because AG30 has an α-helix structure with a number of hydrophobic and cationic amino acids, we designed a modified AG30 peptide by replacing several of the amino acids. The replacement of cationic amino acids (yielding a new molecule, AG30/5C), but not hydrophobic amino acids, increased both the angiogenic and the antimicrobial properties of the peptide. AG30/5C was also effective against methicillin-resistant Staphylococcus aureus (MRSA) and antibiotic-resistant Pseudomonas aeruginosa. In a diabetic mouse wound-healing model, the topical application of AG30/5C accelerated wound healing with increased angiogenesis and attenuated MRSA infection. To facilitate the eventual clinical investigation/application of these compounds, we developed a large-scale procedure for the synthesis of AG30/5C that employed the conventional solution method and met Good Manufacturing Practice guidelines. In the evaluation of stability of this peptide in saline solution, RP-HPLC analysis revealed that AG30/5C was fairly stable under 5°C for 12 months. Therefore, we propose the use of AG30/5C as a wound-healing drug with antibacterial and angiogenic actions.


Assuntos
Proteínas Angiogênicas/farmacologia , Peptídeos Catiônicos Antimicrobianos/farmacologia , Desenho de Fármacos , Peptídeos/farmacologia , Proteínas Angiogênicas/síntese química , Animais , Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/síntese química , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Dicroísmo Circular , Estudos de Viabilidade , Humanos , Masculino , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Testes de Sensibilidade Microbiana , Peptídeos/síntese química , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/crescimento & desenvolvimento , Cicatrização/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...