Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
BMC Immunol ; 15: 48, 2014 Oct 18.
Article in English | MEDLINE | ID: mdl-25323934

ABSTRACT

BACKGROUND: The application of vaccine adjuvants has been vigorously studied for a diverse range of diseases in order to improve immune responses and reduce toxicity. However, most adjuvants have limited uses in clinical practice due to their toxicity. METHODS: Therefore, to reduce health risks associated with the use of such adjuvants, we developed an advanced non-toxic adjuvant utilizing biodegradable chitosan hydrogel (CH-HG) containing ovalbumin (OVA) and granulocyte-macrophage colony-stimulating factor (GM-CSF) as a local antigen delivery system. RESULTS: After subcutaneous injection into mice, OVA/GM-CSF-loaded CH-HG demonstrated improved safety and enhanced OVA-specific antibody production compared to oil-based adjuvants such as Complete Freund's adjuvant (CFA) or Incomplete Freund's adjuvant (IFA). Moreover, CH-HG system-mediated immune responses was characterized by increased number of OVA-specific CD4(+) and CD8(+) INF-γ(+) T cells, leading to enhanced humoral and cellular immunity. CONCLUSIONS: In this study, the improved safety and enhanced immune response characteristics of our novel adjuvant system suggest the possibility of the extended use of adjuvants in clinical practice with reduced apprehension about toxic side effects.


Subject(s)
Adjuvants, Immunologic/toxicity , Chitosan/toxicity , Epitopes/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hydrogel, Polyethylene Glycol Dimethacrylate/toxicity , Immunity/drug effects , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Female , Freund's Adjuvant , Immunization , Immunoglobulin G/immunology , Injections, Subcutaneous , Lipids , Mice, Inbred C57BL , Ovalbumin/immunology
2.
Sci Rep ; 13(1): 6864, 2023 04 26.
Article in English | MEDLINE | ID: mdl-37100873

ABSTRACT

Mycoplasma fermentans is a proposed risk factor of several neurological diseases that has been detected in necrotic brain lesions of acquired immunodeficiency syndrome patients, implying brain invasiveness. However, the pathogenic roles of M. fermentans in neuronal cells have not been investigated. In this study, we found that M. fermentans can infect and replicate in human neuronal cells, inducing necrotic cell death. Necrotic neuronal cell death was accompanied by intracellular amyloid-ß (1-42) deposition, and targeted depletion of amyloid precursor protein by a short hairpin RNA (shRNA) abolished necrotic neuronal cell death. Differential gene expression analysis by RNA sequencing (RNA-seq) showed that interferon-induced transmembrane protein 3 (IFITM3) was dramatically upregulated by M. fermentans infection, and knockdown of IFITM3 abolished both amyloid-ß (1-42) deposition and necrotic cell death. A toll-like receptor 4 antagonist inhibited M. fermentans infection-mediated IFITM3 upregulation. M. fermentans infection also induced necrotic neuronal cell death in the brain organoid. Thus, neuronal cell infection by M. fermentans directly induces necrotic cell death through IFITM3-mediated amyloid-ß deposition. Our results suggest that M. fermentans is involved in neurological disease development and progression through necrotic neuronal cell death.


Subject(s)
Mycoplasma Infections , Mycoplasma fermentans , Humans , Cell Death , Membrane Proteins/metabolism , Mycoplasma fermentans/metabolism , Mycoplasma Infections/complications , Necrosis/complications , RNA-Binding Proteins , Tumor Necrosis Factor-alpha/metabolism
3.
Eur J Cancer ; 153: 179-189, 2021 08.
Article in English | MEDLINE | ID: mdl-34182269

ABSTRACT

OBJECTIVE: Anti-programmed death (PD)-1 therapy confers sustainable clinical benefits for patients with non-small-cell lung cancer (NSCLC), but only some patients respond to the treatment. Various clinical characteristics, including the PD-ligand 1 (PD-L1) level, are related to the anti-PD-1 response; however, none of these can independently serve as predictive biomarkers. Herein, we established a machine learning (ML)-based clinical decision support algorithm to predict the anti-PD-1 response by comprehensively combining the clinical information. MATERIALS AND METHODS: We collected clinical data, including patient characteristics, mutations and laboratory findings, from the electronic medical records of 142 patients with NSCLC treated with anti-PD-1 therapy; these were analysed for the clinical outcome as the discovery set. Nineteen clinically meaningful features were used in supervised ML algorithms, including LightGBM, XGBoost, multilayer neural network, ridge regression and linear discriminant analysis, to predict anti-PD-1 responses. Based on each ML algorithm's prediction performance, the optimal ML was selected and validated in an independent validation set of PD-1 inhibitor-treated patients. RESULTS: Several factors, including PD-L1 expression, tumour burden and neutrophil-to-lymphocyte ratio, could independently predict the anti-PD-1 response in the discovery set. ML platforms based on the LightGBM algorithm using 19 clinical features showed more significant prediction performance (area under the curve [AUC] 0.788) than on individual clinical features and traditional multivariate logistic regression (AUC 0.759). CONCLUSION: Collectively, our LightGBM algorithm offers a clinical decision support model to predict the anti-PD-1 response in patients with NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Machine Learning/standards , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Immune Checkpoint Inhibitors/pharmacology , Lung Neoplasms/pathology , Male , Middle Aged
4.
Exp Mol Med ; 52(5): 750-761, 2020 05.
Article in English | MEDLINE | ID: mdl-32439954

ABSTRACT

T cell activation requires extracellular stimulatory signals that are mainly mediated by T cell receptor (TCR) complexes. The TCR recognizes antigens on major histocompatibility complex molecules with the cooperation of CD4 or CD8 coreceptors. After recognition, TCR-induced signaling cascades that propagate signals via various molecules and second messengers are induced. Consequently, many features of T cell-mediated immune responses are determined by these intracellular signaling cascades. Furthermore, differences in the magnitude of TCR signaling direct T cells toward distinct effector linages. Therefore, stringent regulation of T cell activation is crucial for T cell homeostasis and proper immune responses. Dysregulation of TCR signaling can result in anergy or autoimmunity. In this review, we summarize current knowledge on the pathways that govern how the TCR complex transmits signals into cells and the roles of effector molecules that are involved in these pathways.


Subject(s)
Cell Differentiation , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Humans , Immunity, Cellular , Immunomodulation , Lymphocyte Activation/genetics , T-Lymphocytes/cytology
5.
J Biomed Nanotechnol ; 16(2): 201-211, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-32252881

ABSTRACT

A key issue in dendritic cell (DC)-based cancer immunotherapy is the effective delivery of tumor-specific antigens to DCs. To deliver antigens, non-viral vaccine system has been used in ex vivo manipulation. However, ex vivo manipulation is time-consuming, lacks quality control of DCs, and demonstrates low antigen delivery efficiency, which implicates that there are serious problems in therapeutic DC preparations. Therefore, we developed mannose (MN)-labeled poly(d, l-lactide-co-glycolide) (PLGA) nanoparticles (MN-PLGA-NPs) encapsulating tumor-specific antigens for targeted delivery to mannose receptors (MN-R) on DC surfaces without ex vivo manipulation. The MN-PLGA-NPs showed DC-selective delivery in tumor-bearing mice, leading to highly mature and activated DCs, which migrated to lymphoid organs, resulting in activation of cytotoxic CD8+ T cells. Additionally, MN-PLGA-NPs showed significant therapeutic efficacy in EG7 lymphoma tumorbearing mice. Our nano-platform technology can be used as a vaccine system to bypass ex vivo manipulation and enhance targeted delivery of tumor-specific antigens to DCs, which is well-suited for cancer immunotherapy.


Subject(s)
Dendritic Cells , Nanoparticles , Neoplasms , Animals , Dioxanes , Immunotherapy , Lactic Acid , Mannose , Mice , Mice, Inbred C57BL , Neoplasms/therapy , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer
6.
ACS Appl Bio Mater ; 2(6): 2481-2489, 2019 Jun 17.
Article in English | MEDLINE | ID: mdl-35030704

ABSTRACT

Dendritic cell (DC)-based cancer immunotherapies have been studied extensively. In cancer immunotherapy, the initial key step is the delivery of tumor-specific antigens, leading to the maturation and activation of DCs. To promote effective antigen delivery, liposome-based delivery systems for tumor-specific antigens have been investigated, and although promising, a triggered release of the antigen from the liposome is required to attain an optimum immune response. In this study, we developed CO2-bubble-generating thermosensitive liposomes (BG-TSLs) that encapsulate whole tumor cell lysates (TCLs). The release of the lysate from BG-TSLs can be triggered using near-infrared (NIR) irradiation. We also developed BG-TSLs able to encapsulate doxorubicin (DOX) for combination therapy. The DOX-BG-TSLs and TCL-BG-TSLs have a mean particle size of 114.17 ± 8.28 nm and 123.8 ± 10.2 nm and a surface charge of -22.56 ± 1.3 mV and -28.9 ± 0.8 mV, respectively. CO2 bubble generation within TCL-BG-TSLs and DOX-BG-TSLs by NIR irradiation led to the burst release of TCL or DOX. TCL release from TCL-BG-TSLs promoted dendritic cell maturation and activation, leading to the emergence of antigen-specific cytotoxic CD8+ T cells. The combination of TCL-BG-TSLs with DOX-BG-TSLs showed a significantly greater antitumor efficacy in B16F10 tumor-bearing mice compared to that seen in the control mice (P < 0.001). Taken together, our liposomal delivery system, combined with NIR irradiation, could enhance the therapeutic efficacy of cancer immunotherapies.

7.
Cancer Res ; 78(21): 6247-6256, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30115698

ABSTRACT

Chemotherapy is commonly used in the treatment of ovarian cancer, yet most ovarian cancers harbor inherent resistance or develop acquired resistance. Therefore, novel therapeutic approaches to overcome chemoresistance are required. In this study, we developed a hyaluronic acid-labeled poly(d,l-lactide-co-glycolide) nanoparticle (HA-PLGA-NP) encapsulating both paclitaxel (PTX) and focal adhesion kinase (FAK) siRNA as a selective delivery system against chemoresistant ovarian cancer. The mean size and zeta potential of the HA-PLGA-NP were 220 nm and -7.3 mV, respectively. Incorporation efficiencies for PTX and FAK siRNA in the HA-PLGA-NPs were 77% and 85%, respectively. HA-PLGA-NP showed higher binding efficiency for CD44-positive tumor cells as compared with CD44-negative cells. HA-PLGA (PTX+FAK siRNA)-NP caused increased cytotoxicity and apoptosis in drug-resistant tumor cells. Treatment of human epithelial ovarian cancer tumor models HeyA8-MDR (P < 0.001) and SKOV3-TR (P < 0.001) with HA-PLGA (PTX+FAK siRNA)-NP resulted in significant inhibition of tumor growth. Moreover, in a drug-resistant, patient-derived xenograft (PDX) model, HA-PLGA (PTX+FAK siRNA)-NP significantly inhibited tumor growth compared with PTX alone (P < 0.002). Taken together, HA-PLGA-NP acts as an effective and selective delivery system for both the chemotherapeutic and the siRNA in order to overcome chemoresistance in ovarian carcinoma.Significance: These findings demonstrate the efficacy of a novel, selective, two-in-one delivery system to overcome chemoresistance in epithelial ovarian cancer. Cancer Res; 78(21); 6247-56. ©2018 AACR.


Subject(s)
Carcinoma, Ovarian Epithelial/metabolism , Focal Adhesion Kinase 1/metabolism , Hyaluronan Receptors/chemistry , Nanoparticles/chemistry , Paclitaxel/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , RNA, Small Interfering/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Ovarian Epithelial/drug therapy , Cell Line, Tumor , Cell Survival , Drug Carriers/chemistry , Drug Resistance, Neoplasm , Female , Gene Silencing , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism
8.
Drug Deliv ; 25(1): 1394-1402, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29890852

ABSTRACT

Angiogenesis plays an essential role in the growth and metastasis of tumor cells, and the modulation of angiogenesis can be an effective approach for cancer therapy. We focused on silencing the angiogenic gene PLXDC1 as an important factor for anti-angiogenesis tumor therapy. Herein, we developed PLXDC1 small interfering siRNA (siRNA)-incorporated chitosan nanoparticle (CH-NP/siRNA) coated with hyaluronic acid (HA) to target the CD44 receptor on tumor endothelial cells. This study aimed to improve targeted delivery and enhance therapeutic efficacy for tumor anti-angiogenesis. The HA-CH-NP/siRNA was 200 ± 10 nm in size with a zeta potential of 26.4 mV. The loading efficiency of siRNA to the HA-CH-NP/siRNA was up to 60%. The selective binding of HA-CH-NP/siRNA to CD44-positive tumor endothelial cells increased by 2.1-fold compared with that of the CD44 nontargeted CH-NP/siRNA. PLXDC1 silencing by the HA-CH-NP/siRNA significantly inhibited tumor growth in A2780 tumor-bearing mice compared with that in the control group (p < .01), and mRNA expression of PLXDC1 was significantly reduced in the HA-CH-NP/siRNA-treated group. Furthermore, treatment with HA-CH-NP/siRNA resulted in significant inhibition of cell proliferation (p < .001), reduced microvessel density (p < .001), and increased cell apoptosis (p < .001). This study demonstrates that HA-CH-NP/siRNA is a highly selective delivery platform for siRNA, and has broad potential to be used in anti-angiogenesis tumor therapy.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Chitosan/chemistry , Endothelial Cells/drug effects , Hyaluronan Receptors/genetics , Nanoparticles/chemistry , Neoplasm Proteins/genetics , Neoplasms, Glandular and Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , RNA, Small Interfering/administration & dosage , Receptors, Cell Surface/genetics , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Carcinoma, Ovarian Epithelial , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Delivery Systems/methods , Female , Gene Silencing/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Hyaluronic Acid/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/administration & dosage , Particle Size , RNA, Messenger/genetics , Xenograft Model Antitumor Assays/methods
9.
Int J Nanomedicine ; 11: 5729-5742, 2016.
Article in English | MEDLINE | ID: mdl-27843314

ABSTRACT

Dendritic cells (DCs) are potent professional antigen-presenting cells that are capable of initiating a primary immune response and activating T cells, and they play a pivotal role in the immune responses of the host to cancer. Prior to antigen presentation, efficient antigen and adjuvant uptake by DCs is necessary to induce their maturation and cytokine generation. Nanoparticles (NPs) are capable of intracellular delivery of both antigen and adjuvant to DCs. Here, we developed an advanced poly(d,l-lactide-co-glycolide) (PLGA)-NP encapsulating both ovalbumin (OVA) as a model antigen and polyinosinic-polycytidylic acid sodium salt (Toll-like receptor 3 ligand) as an adjuvant to increase intracellular delivery and promote DC maturation. The PLGA-NPs were taken up by DCs, and their uptake greatly facilitated major histocompatibility class I antigen presentation in vitro. Moreover, vaccination with PLGA-NP-treated DCs led to the generation of ovalbumin-specific CD8+ T cells, and the resulting antitumor efficacy was significantly increased in EG.7 and TC-1 tumor-bearing mice compared to control mice (P<0.01). Taken together, these findings demonstrated that the PLGA-NP platform may be an effective method for delivering tumor-specific antigens or adjuvants to DCs.


Subject(s)
Dendritic Cells/immunology , Immunotherapy , Lactic Acid/chemistry , Nanoparticles/chemistry , Neoplasms, Experimental/therapy , Polyglycolic Acid/chemistry , Thymoma/therapy , Toll-Like Receptor 3/immunology , Adjuvants, Immunologic , Animals , Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Female , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Ovalbumin/immunology , Polylactic Acid-Polyglycolic Acid Copolymer , T-Lymphocytes/immunology , T-Lymphocytes, Cytotoxic/immunology , Thymoma/immunology , Thymoma/pathology , Thymus Neoplasms/immunology , Thymus Neoplasms/pathology , Thymus Neoplasms/therapy , Toll-Like Receptor 3/metabolism
10.
Mol Cancer Ther ; 15(4): 618-27, 2016 04.
Article in English | MEDLINE | ID: mdl-26861249

ABSTRACT

Although cytotoxic chemotherapy is widely used against epithelial ovarian cancer (EOC), adverse side effects and emergence of resistance can limit its utility. Therefore, new drugs with systemic delivery platforms are urgently needed for this disease. In this study, we developed linalool-incorporated nanoparticles (LIN-NP) as a novel anticancer agent. We prepared LIN-NPs by the self-assembly water-in-oil-in-water (w/o/w) emulsion method. LIN-NP-mediated cytotoxicity and apoptosis was assessed in EOC cells, and the role of reactive oxygen species (ROS) generation as the mechanism of action was evaluated. In addition, therapeutic efficacy of LIN-NP was assessed in cell lines and patient-derived xenograft (PDX) models for EOC. LIN-NPs had significant cytotoxicity and apoptotic activity against EOC cells, including A2780, HeyA8, and SKOV3ip1. LIN-NP treatment increased apoptosis in EOC cells through ROS generation and a subsequent decrease in mitochondrial membrane potential and increase in caspase-3 levels. In addition, 100 mg/kg LIN-NPs significantly decreased tumor weight in the HeyA8 (P < 0.001) and SKOV3ip1 (P = 0.006) in vivo models. Although treatment with 50 mg/kg LIN-NP did not decrease tumor weight compared with the control group, combination treatment with paclitaxel significantly decreased tumor weight compared with paclitaxel alone in SKOV3ip1 xenografts (P = 0.004) and the patient-derived xenograft model (P = 0.020). We have developed LIN-NPs that induce ROS generation as a novel anticancer agent for EOC. These findings have broad applications for cancer therapy. Mol Cancer Ther; 15(4); 618-27. ©2016 AACR.


Subject(s)
Antineoplastic Agents/administration & dosage , Monoterpenes/administration & dosage , Nanoparticles , Acyclic Monoterpenes , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Drug Therapy, Combination , Female , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Monoterpenes/chemistry , Nanoparticles/chemistry , Neoplasms, Glandular and Epithelial/drug therapy , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Paclitaxel/pharmacology , Reactive Oxygen Species/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
11.
Sci Rep ; 6: 38348, 2016 12 02.
Article in English | MEDLINE | ID: mdl-27910914

ABSTRACT

Dentritic cell (DC)-based cancer immunotherapy faces challenges in both efficacy and practicality. However, DC-based vaccination requires multiple injections and elaborates ex vivo manipulation, which substantially limits their use. Therefore, we sought to develop a chitosan nanoparticle (CH-NP)-based platform for the next generation of vaccines to bypass the ex vivo manipulation and induce immune responses via active delivery of polyinosinic-polycytidylic acid sodium salt (poly I:C) to target Toll-like receptor 3 (TLR3) in endosomes. We developed CH-NPs encapsulating ovalbumin (OVA) as a model antigen and poly I:C as the adjuvant in an ionic complex. These CH-NPs showed increased in vivo intracellular delivery to the DCs in comparison with controls after injection into tumor-bearing mice, and promoted DC maturation, leading to emergence of antigen-specific cytotoxic CD8+ T cells. Finally, the CH-NPs showed significantly greater antitumor efficacy in EG.7 and TC-1 tumor-bearing mice compared to the control (p < 0.01). Taken together, these data show that the CH-NP platform can be used as an immune response modulatory vaccine for active cancer immunotherapy without ex vivo manipulation, thus resulting in increased anticancer efficacy.


Subject(s)
Antigens/immunology , Immunotherapy/methods , Lung Neoplasms/therapy , Lymphoma/therapy , Ovalbumin/immunology , Poly I-C/administration & dosage , Adjuvants, Immunologic/administration & dosage , Animals , Antigen Presentation/drug effects , Antigens/administration & dosage , Antigens/chemistry , Cancer Vaccines/administration & dosage , Cancer Vaccines/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Chitosan/chemistry , Chitosan/metabolism , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Female , Gene Expression , Immunomodulation/drug effects , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lymphoma/immunology , Lymphoma/pathology , Mice , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Nanotechnology/methods , Ovalbumin/administration & dosage , Ovalbumin/chemistry , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/immunology , Transfection
12.
J Control Release ; 216: 132-9, 2015 Oct 28.
Article in English | MEDLINE | ID: mdl-26247553

ABSTRACT

Stimulus-triggered drug release based on the liposomal drug delivery platform has been studied vigorously to increase drug release at the target site. Although the delivery system has been developed, an effective carrier system is needed to achieve effective therapeutic efficacy. Therefore, we focused on the development of gold cluster bound thermosensitive liposomes (G-TSL), which are capable of triggered drug release when stimulated by external near-infrared (NIR) irradiation in the tumor microenvironment. The size of doxorubicin (DOX)-loaded G-TSL (DOX/G-TSL) was 171.5 ± 8.3 nm, and the efficiency of DOX encapsulation was up to 90%. The release of DOX from DOX/G-TSL was increased 70% by NIR irradiation (1.50 W/cm(2) for 0.5 min) compared to non-gold-coated TSL. Consequentially, the gold cluster on the TSL enabled the light-controlled DOX release through the photothermal conversion of the energy of NIR-absorbed light, leading to membrane destabilization. Cell cytotoxicity of DOX/G-TSL was also increased by their NIR irradiation-triggered DOX release compared to non-NIR-irradiated DOX/G-TSL. In addition, we demonstrated the therapeutic efficacy of DOX/G-TSL against the MDA-MB-231 tumor model. The NIR-irradiated DOX/G-TSL treatment showed greater therapeutic efficacy than that of the non-NIR-irradiated DOX/G-TSL and control (p<0.05). Taken together, DOX/G-TSL has the potential for remote-triggered drug release upon stimulation with NIR irradiation in the tumor microenvironment, and may be applied to a broad range of photothermal-based disease therapies.


Subject(s)
Gold/pharmacology , Liposomes/chemistry , Neoplasms/metabolism , Tumor Microenvironment/drug effects , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Compounding , Drug Delivery Systems , Female , Gold/chemistry , Hot Temperature , Humans , Infrared Rays , Light , Mice , Mice, Nude , Particle Size
13.
Acta Biomater ; 24: 279-285, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26102337

ABSTRACT

Drug delivery using thermosensitive liposomes (TSL) has significant potential for tumor drug targeting and can be combined with local hyperthermia to trigger drug release. Although TSL-mediated drug delivery can be effective by itself, we developed doxorubicin (DOX)-containing CO2 bubble-generating TSL (TSL-C) that were found to enhance the antitumor effects of DOX owing to the synergism between burst release of drug and hyperthermia-induced CO2 generation. An ultrasound imaging system was used to monitor hyperthermia-induced CO2 generation in TSL-C and the results revealed that hyperthermia-induced CO2 generation in TSL-C led to increased DOX release compared to that observed for non-CO2-generating TSL. Moreover, TSL-C significantly inhibited the tumor growth in MDA-MB-231 tumor-bearing mice compared to TSL (p<0.004). Taken together, we demonstrated that the TSL-C platform increased the therapeutic efficacy of cancer chemotherapy and showed the applicability of this approach to increase drug release within the tumor microenvironment. As a novel and highly effective drug delivery platform, TSL-C has great potential for use in a broad range of applications for the treatment of various human diseases. STATEMENT OF SIGNIFICANCE: We have developed a novel method for drug release from liposomes by gas (CO2) generation in tumor microenvironment. In addition, we demonstrate therapeutic efficacy in breast carcinoma. CO2-generated liposomal doxorubicin is a novel and highly attractive delivery system for anticancer drug with the potential for broad applications in human disease.


Subject(s)
Breast Neoplasms/drug therapy , Carbon Dioxide/chemistry , Doxorubicin/chemistry , Doxorubicin/pharmacology , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Humans , Liposomes , Mice , Mice, Nude , Xenograft Model Antitumor Assays
14.
Nanoscale Res Lett ; 9(1): 209, 2014.
Article in English | MEDLINE | ID: mdl-24855464

ABSTRACT

Liposome-based drug delivery systems hold great potential for cancer therapy. However, to enhance the localization of payloads, an efficient method of systemic delivery of liposomes to tumor tissues is required. In this study, we developed cationic liposomes composed of polyethylenimine (PEI)-conjugated distearoylglycerophosphoethanolamine (DSPE) as an enhanced local drug delivery system. The particle size of DSPE-PEI liposomes was 130 ± 10 nm and the zeta potential of liposomes was increased from -25 to 30 mV by the incorporation of cationic PEI onto the liposomal membrane. Intracellular uptake of DSPE-PEI liposomes by tumor cells was 14-fold higher than that of DSPE liposomes. After intratumoral injection of liposomes into tumor-bearing mice, DSPE-PEI liposomes showed higher and sustained localization in tumor tissue compared to DSPE liposomes. Taken together, our findings suggest that DSPE-PEI liposomes have the potential to be used as effective drug carriers for enhanced intracellular uptake and localization of anticancer drugs in tumor tissue through intratumoral injection.

SELECTION OF CITATIONS
SEARCH DETAIL