Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Control Release ; 282: 131-139, 2018 07 28.
Article in English | MEDLINE | ID: mdl-29702142

ABSTRACT

Recent studies have shown that certain combinations of Toll-like receptor (TLR) agonists can induce synergistic immune activation. However, it remains challenging to achieve such robust responses in vivo in a manner that is effective, facile, and amenable for clinical translation. Here, we show that MPLA, a TLR4 agonist, and CpG, a TLR9 agonist, can be efficiently co-loaded into synthetic high-density lipoprotein nanodiscs, forming a potent adjuvant system (ND-MPLA/CpG) that can be readily combined with a variety of subunit antigens, including proteins and peptides. ND-MPLA/CpG significantly enhanced activation of dendritic cells, compared with free dual adjuvants or nanodiscs delivering a single TLR agonist. Importantly, mice immunized with physical mixtures of protein antigens ND-MPLA/CpG generated strong humoral responses, including induction of IgG responses against protein convertase subtilisin/kexin 9 (PCSK9), leading to 17-30% reduction of the total plasma cholesterol levels. Moreover, ND-MPLA/CpG exerted strong anti-tumor efficacy in multiple murine tumor models. Compared with free adjuvants, ND-MPLA/CpG admixed with ovalbumin markedly improved antigen-specific CD8+ T cell responses by 8-fold and promoted regression of B16F10-OVA melanoma (P < 0.0001). Furthermore, ND-MPLA/CpG admixed with E7 peptide antigen elicited ~20% E7-specific CD8+ T cell responses and achieved complete regression of established TC-1 tumors in all treated animals. Taken together, our work highlights the simplicity, versatility, and potency of dual TLR agonist nanodiscs for applications in vaccines and cancer immunotherapy.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Lipid A/analogs & derivatives , Oligodeoxyribonucleotides/administration & dosage , Toll-Like Receptor 4/agonists , Toll-Like Receptor 9/agonists , Vaccines/administration & dosage , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/therapeutic use , Animals , Cells, Cultured , Drug Carriers/chemistry , Female , Humans , Immunity, Humoral , Immunization , Immunotherapy , Lipid A/administration & dosage , Lipid A/immunology , Lipid A/therapeutic use , Melanoma/immunology , Melanoma/therapy , Mice , Mice, Inbred C57BL , Nanostructures/chemistry , Oligodeoxyribonucleotides/immunology , Oligodeoxyribonucleotides/therapeutic use , Toll-Like Receptor 4/immunology , Toll-Like Receptor 9/immunology , Vaccines/immunology , Vaccines/therapeutic use
2.
Int J Pharm ; 510(1): 394-405, 2016 Aug 20.
Article in English | MEDLINE | ID: mdl-27374198

ABSTRACT

Treatment of brain tumor remains a great challenge worldwide. Development of a stable, safe, and effective siRNA delivery system which is able to cross the impermeable blood-brain barrier (BBB) and target glioma cells is necessary. This study aims to investigate the therapeutic effects of intravenous administration of T7 peptide modified core-shell nanoparticles (named T7-LPC/siRNA NPs) on brain tumors. Layer-by-layer assembling of protamine/chondroitin sulfate/siRNA/cationic liposomes followed by T7 peptide modification has been carried out in order to obtain a targeted siRNA delivery system. In vitro cellular uptake experiments demonstrated a higher intracellular fluorescence intensity of siRNA in brain microvascular endothelial cells (BMVECs) and U87 glioma cells when treated with T7-LPC/siRNA NPs compared with PEG-LPC/siRNA NPs. In the co-culture model of BMVECs and U87 cells, a significant down-regulation of EGFR protein expression occurred in the U87 glioma cells after treatment with the T7-LPC/siEGFR NPs. Moreover, the T7-LPC/siRNA NPs had an advantage in penetrating into a deep region of the tumor spheroid compared with PEG-LPC/siRNA NPs. In vivo imaging revealed that T7-LPC/siRNA NPs accumulated more specifically in brain tumor tissues than the non-targeted NPs. Also, in vivo tumor therapy experiments demonstrated that the longest survival period along with the greatest downregulation of EGFR expression in tumor tissues was observed in mice with an intracranial U87 glioma treated with T7-LPC/siEGFR NPs compared with mice receiving other formulations. Therefore, we believe that these transferrin receptor-mediated core-shell nanoparticles are an important potential siRNA delivery system for brain tumor-targeted therapy.


Subject(s)
Brain Neoplasms/therapy , Gene Transfer Techniques , Nanoparticles/administration & dosage , RNA, Small Interfering/administration & dosage , Receptors, Transferrin/administration & dosage , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Line, Tumor , Coculture Techniques , Humans , Male , Mice , Mice, Nude , Nanoparticles/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism
3.
Eur J Pharm Sci ; 92: 39-48, 2016 Sep 20.
Article in English | MEDLINE | ID: mdl-27355138

ABSTRACT

Systemic delivery of siRNA is the most challenging step to transfer RNAi to clinical application for breast cancer therapy. In this study, the tumor targeted, T7 peptide modified core-shell nanoparticles (named as T7-LPC/siRNA NPs) were constructed to achieve effective systemic delivery of siRNA. The core-shell structure of T7-LPC/siRNA NPs enables them to encapsulate siRNA in the core and protect it from RNase degradation during circulation. In vitro cellular uptake and gene silencing experiments demonstrated that T7-LPC/siEGFR NPs could deliver EGFR siRNA into breast cancer cells through receptor mediated endocytosis and effectively down-regulate the EGFR expression. In vivo distribution study proved the T7-LPC/siRNA NPs could deliver fluorescence labeled siRNA to the tumor site more efficiently than the non-targeted PEG-LPC/siRNA NPs after intravenous administration. Furthermore, the experiments of in vivo tumor therapy confirmed that intravenous administration of T7-LPC/siEGFR NPs led to an effective EGFR down-regulation and an obvious inhibition of breast tumor growth, with little activation of immune responses and negligible body weight loss. These results suggested that T7-LPC/siRNA NPs could be an effective and safe systemic siRNA delivery system for RNAi-based breast cancer therapy.


Subject(s)
Breast Neoplasms/drug therapy , Collagen Type IV/administration & dosage , Nanoparticles/administration & dosage , Peptide Fragments/administration & dosage , RNA, Small Interfering/administration & dosage , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Collagen Type IV/chemistry , Collagen Type IV/pharmacokinetics , Collagen Type IV/therapeutic use , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Humans , Interferon-gamma/blood , Interleukin-6/blood , MCF-7 Cells , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Peptide Fragments/chemistry , Peptide Fragments/pharmacokinetics , Peptide Fragments/therapeutic use , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemistry , RNA Interference , RNA, Small Interfering/chemistry , RNA, Small Interfering/pharmacokinetics , RNA, Small Interfering/therapeutic use , Tissue Distribution , Tumor Burden/drug effects
4.
J Control Release ; 235: 99-111, 2016 08 10.
Article in English | MEDLINE | ID: mdl-27242198

ABSTRACT

In our previous study, a H-shape gemini-like cationic lipid (ssGLCL, formerly named as CLD), composed of two hydrophilic lysine heads and two hydrophobic oleyl alcohol tails with a bridge of the redox-active disulfide-bond, had been synthesized and used as a nanocarrier for delivering small interfering RNAs (siRNAs) into cells. In order to further elucidate the role of disulfide (-S-S-) bridge on the activity of ssGLCL based siRNA delivery, a comparable ccGLCL bridged with a non-reducible carbon-carbon bond was synthesized and used as control in this study. Both two H-shape GLCL molecules could individually self-assemble into cationic nanoparticles in water phase and complex with negatively-charged siRNA into nanoplexes with particle size of ~200nm and zeta potential of ~ +30mV, and exhibit effective siRNA delivery both in vitro and in vivo. Investigation of internalization pathway displayed that both ssGLCL/siRNA and ccGLCL/siRNA nanoplexes were predominantly internalized into MCF-7 cells by the clathrin-mediated endocytosis pattern. Although a lower cellular uptake of siRNA was found in the human breast cancer MCF-7 cells, the ssGLCL/siRNA nanoplexes could exhibit similar or even stronger down-regulation effects on the targeted EGFR mRNA and protein in MCF-7 cells when compared to the ccGLCL/siRNA nanoplexes. Furthermore, mechanistic study showed that the enhanced down-regulation effects of ssGLCL/siRNA nanoplexes on targeted mRNA and protein were probably attributed to the increased release of siRNA from lysosomes to cytoplasm following the cleavage of redox-active disulfide-bridge in ssGLCL. Therefore, we believed that the redox-active H-shape ssGLCL could be a potential nanocarrier towards improving siRNA delivery.


Subject(s)
Disulfides/chemistry , Gene Transfer Techniques , Lipids/administration & dosage , Nanoparticles/administration & dosage , RNA, Small Interfering/administration & dosage , Animals , Cell Survival/drug effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , Erythrocytes/drug effects , Female , Gene Silencing , HeLa Cells , Hemolysis/drug effects , Humans , Lipids/chemistry , MCF-7 Cells , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/therapy , RNA, Messenger/metabolism , RNA, Small Interfering/chemistry , Rats , Xenograft Model Antitumor Assays
5.
Biomaterials ; 35(18): 5028-38, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24680191

ABSTRACT

A somatostatin analog, vapreotide (VAP), can be used as a ligand for targeting drug delivery based on its high affinity to somatostatin receptors (SSTRs), which is overexpressed in many tumor cells. RNA interference plays an important role on downregulation of vascular endothelial growth factor (VEGF), which is important for tumor growth, progression and metastasis. To improve tumor therapy efficacy, the vapreotide-modified core-shell type nanoparticles co-encapsulating VEGF targeted siRNA (siVEGF) and paclitaxel (PTX), termed as VAP-PLPC/siRNA NPs, were developed in this study. When targeted via somatostatin receptors to tumor cells, the VAP-PLPC/siRNA NPs could simultaneously delivery siVEGF and PTX into cells and achieve a synergistic inhibition of tumor growth. Interestingly, in vitro cell uptake and gene silencing experiments demonstrated that the targeted VAP-PLPC/siRNA NPs exhibited significant higher intracellular siRNA accumulation and VEGF downregulation in human breast cancer MCF-7 cells, compared to those of the non-targeted PEG-PLPC/siRNA NPs. More importantly, in vivo results further demonstrated that the targeted VAP-PLPC/siRNA NPs had significant stronger drug distribution in tumor tissues and tumor growth inhibition efficacy via receptor-mediated targeting delivery, accompany with an obvious inhibition of neovascularization induced by siVEGF silencing. These results suggested that the co-delivery of siRNA and paclitaxel via vapreotide-modified core-shell nanoparticles would be a promising approach for tumor targeted therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Molecular Targeted Therapy , Nanoparticles/chemistry , Paclitaxel/pharmacology , RNA, Small Interfering/genetics , Somatostatin/analogs & derivatives , Vascular Endothelial Growth Factor A/genetics , Animals , Breast Neoplasms/therapy , Cell Cycle/drug effects , Down-Regulation , Drug Delivery Systems , Female , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Somatostatin/metabolism , Somatostatin/pharmacology , Vascular Endothelial Growth Factor A/metabolism
6.
Biomaterials ; 35(6): 2066-78, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24315577

ABSTRACT

Our previous study had reported that cholesterol-grafted poly(amidoamine) (rPAA-Chol polymer) was able to self-assemble into cationic nanoparticles and act as a potential carrier for siRNA transfection. In this study, the core-shell type lipid/rPAA-Chol hybrid nanoparticles (PEG-LP/siRNA NPs and T7-LP/siRNA NPs) were developed for improving in vivo siRNA delivery by modifying the surface of rPAA-Chol/siRNA nanoplex core with a lipid shell, followed by post-insertion of polyethylene glycol phospholipid (DSPE-PEG) and/or peptide (HAIYPRH, named as T7) modified DSPE-PEG-T7. The integrative hybrid nanostructures of LP/siRNA NPs were evidenced by dynamic light scattering (DLS), confocal laser scanning microscope (CLSM), cryo-transmission electron microscope (Cryo-TEM) and surface plasmon resonance (SPR) assay. It was demonstrated that the T7 peptide modified LP/siRNA NPs (T7-LP/siRNA NPs) exhibited uniform and spherical structures with particle size of 99.39 ± 0.65 nm and surface potential of 42.53 ± 1.03 mV, and showed high cellular uptake efficiency and rapid endosomal/lysosomal escape ability in MCF-7 cells. Importantly, in vitro gene silencing experiment demonstrated that both of pegylated and targeted LP/siEGFR NPs exhibited significantly stronger downregulation of EGFR protein expression level in MCF-7 cells, compared to that of the physical mixture of siRNA lipoplexes and rPAA-Chol/siRNA nanoplexes. In vivo tumor therapy on nude mice bearing MCF-7 tumors further confirmed that the targeted T7-LP/siEGFR NPs exhibited the greatest inhibition on tumor growth via transferrin receptor-mediated targeting delivery, without any activation of immune responses and significant body weight loss following systemic administration. These findings indicated that the core-shell type T7-LP/siRNA nanoparticles would be promising siRNA delivery systems for in vivo tumor-targeted therapy.


Subject(s)
Nanoparticles/chemistry , Polymers/chemistry , RNA, Small Interfering/chemistry , Animals , Cell Survival/genetics , Cell Survival/physiology , Flow Cytometry , Humans , MCF-7 Cells , Mice , Microscopy, Confocal , Polyethylene Glycols/chemistry , RNA, Small Interfering/physiology , Surface Plasmon Resonance
SELECTION OF CITATIONS
SEARCH DETAIL
...