25 research outputs found

    Nanoparticle Delivery of siRNA for Cancer Therapy

    Get PDF
    We have developed lipid-polycation-DNA (LPD) nanoparticles containing DOTAP and targeted with polyethylene glycol (PEG) tethered with a targeting ligand such as anisamide (AA) to specifically deliver siRNA to cancer cells. Two novel non-glycerol based cationic lipids which contain both a guanidinium and a lysine or an arginine residue as the cationic headgroup are synthesized to replace DOTAP and form nanoparticles. DSGLA, which contains a lysine residue, down-regulated pERK more efficiently in H460 cells than DOTAP. DSAA, which contains an arginine residue, induced reactive oxygen species (ROS), triggered apoptosis and down-regulated anti-apoptotic protein Bcl-2 in B16F10 melanoma cells. A significant improvement in tumor growth inhibition was observed after dosing with targeted nanoparticles containing DSGLA or DSAA. We further designed a LPD nanoparticle modification with NGR (aspargine-glycine-arginine) peptide, targeting aminopeptidase N (CD13) expressed in the tumor cells or tumor vascular endothelium. The targeted nanoparticles efficiently delivered c-myc siRNA into the cytoplasm of HT-1080 xenograft tumor and effectively suppressed c-myc expression and triggered cellular apoptosis in the tumor, resulting in a partial tumor growth inhibition. When doxorubicin (Dox) and siRNA were co-formulated in the multi-functional nanoparticles, an enhanced therapeutic effect was observed. Furthermore, we explored the application of the multi-functional nanoparticles in multi-drug resistant cells which are new targets for cancer therapy. In this study, we have used a multi-functional anionic LPD (LPD-II) nanoparticle for efficient systemic co-delivery of siRNA against c-myc and Dox, into P-gp-positive NCI/ADR-RES tumors in a xenograft model. c-Myc siRNA delivered by the targeted nanoparticles significantly down-regulated both c-myc and P-gp expressions in the tumor, caused enhanced Dox uptake and sensitized tumor cells to the co-delivered Dox. Three daily intravenous injections of c-myc siRNA and Dox co-formulated in the targeted nanoparticles showed a significant improvement in tumor growth inhibition. We have further developed a LPH (liposome-polycation-hyaluronic acid) nanoparticle formulation modified with tumor specific scFv (single chain variable fragment) for systemic delivery of small interfering RNA (siRNA) and microRNA (miRNA) into lung metastasis of murine B16F10 melanoma. When miR-34a and therapeutic siRNAs were co-formulated in C4-targeted nanoparticles, an enhanced anti-cancer effect was observed

    Tumor-targeted delivery of siRNA by non-viral vector: safe and effective cancer therapy

    Get PDF
    RNA interference technology has been developed as a potential therapeutic agent for many indications, including cancer. Silencing a specific oncogene in tumor cells brings about cell death both in vitro and in vivo. However, there is a great need for powerful delivery strategies to enhance the therapeutic effect of small interfering RNA (siRNA). This review summarizes different signaling pathways inhibited by siRNA and the advantages of targeted siRNA as a delivery system

    In vivo delivery of miRNAs for cancer therapy: Challenges and strategies

    Get PDF
    MicroRNAs (miRNAs), small non-coding RNAs, can regulate post-transcriptional gene expressions and silence a broad set of target genes. miRNAs, aberrantly expressed in cancer cells, play an important role in modulating gene expressions, thereby regulating downstream signaling pathways and affecting cancer formation and progression. Oncogenes or tumor suppressor genes regulated by miRNAs mediate cell cycle progression, metabolism, cell death, angiogenesis, metastasis and immunosuppression in cancer. Recently, miRNAs have emerged as therapeutic targets or tools and biomarkers for diagnosis and therapy monitoring in cancer. Since miRNAs can regulate multiple cancer-related genes simultaneously, using miRNAs as a therapeutic approach plays an important role in cancer therapy. However, one of the major challenges of miRNA-based cancer therapy is to achieve specific, efficient and safe systemic delivery of therapeutic miRNAs In vivo. This review discusses the key challenges to the development of the carriers for miRNA-based therapy and explores current strategies to systemically deliver miRNAs to cancer without induction of toxicity

    Targeted Nanoparticles Deliver siRNA to Melanoma

    Get PDF
    Melanoma is a severe skin cancer that often leads to death. To examine the potential of small interfering RNA (siRNA) therapy for melanoma, we have developed anisamide-targeted nanoparticles that can systemically deliver siRNA into the cytoplasm of B16F10 murine melanoma cells, which express the sigma receptor. A c-Myc siRNA delivered by the targeted nanoparticles effectively suppressed c-Myc expression in the tumor and partially inhibited tumor growth. More significant tumor growth inhibition was observed with nanoparticles composed of N,N-distearyl-N-methyl-N-2-(N'-arginyl) aminoethyl ammonium chloride (DSAA), a guanidinium-containing cationic lipid, than with a commonly used cationic lipid, 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP). Three daily injections of c-Myc siRNA formulated in the targeted nanoparticles containing DSAA could impair tumor growth, and the ED(50) of c-Myc siRNA was about 0.55  mg  kg(-1). The targeted DSAA nanoparticles containing c-Myc siRNA sensitized B16F10 cells to paclitaxel (Taxol), resulting in a complete inhibition of tumor growth for 1 week. Treatments of c-Myc siRNA in the targeted nanoparticles containing DSAA also showed significant inhibition on the growth of MDA-MB-435 tumor. The enhanced anti-melanoma activity is probably related to the fact that DSAA, but not DOTAP, induced reactive oxygen species, triggered apoptosis, and downregulated antiapoptotic protein Bcl-2 in B16F10 melanoma cells. Thus, the targeted nanoparticles containing c-Myc siRNA may serve as an effective therapeutic agent for melanoma

    Novel Cationic Lipid That Delivers siRNA and Enhances Therapeutic Effect in Lung Cancer Cells

    Get PDF
    We have developed lipid–polycation–DNA (LPD) nanoparticles containing DOTAP and targeted with polyethylene glycol (PEG) tethered with anisamide (AA) to specifically deliver siRNA to H460 human lung carcinoma cells which express the sigma receptor. A novel non-glycerol based cationic lipid which contains both a guanidinium and a lysine residue as the cationic headgroup, i.e. DSGLA, downregulated pERK more efficiently in H460 cells than DOTAP. As demonstrated by using fluorescently labeled siRNA, LPD-PEG-AA prepared with DSGLA efficiently delivered siRNA to the cytoplasm of the H460 cells. Although the siRNA delivered by LPD-PEG-AA containing either DOTAP or DSGLA could effectively silence EGFR expression, a synergistic cell killing effect in promoting cellular apoptosis was only observed with DSGLA. The fluorescently labeled siRNA was efficiently delivered into the cytoplasm of H460 xenograft tumor by the LPD-PEG-AA containing either DOTAP or DSGLA 4 h after intravenous injection. Three daily injections (0.6 mg/kg) of siRNA formulated in the LPD-PEG-AA containing either DOTAP or DSGLA could effectively silence the epidermal growth factor receptor (EGFR) in the tumor, but the formulation containing DSGLA could induce more cellular apoptosis. A significant improvement in tumor growth inhibition was observed after dosing with LPD-PEG-AA containing DSGLA. Thus, DSGLA served as both a formulation component as well as a therapeutic agent which synergistically enhanced the activity of siRNA

    Quantum dot/antibody conjugates for in vivo cytometric imaging in mice

    Get PDF
    Multiplexed, phenotypic, intravital cytometric imaging requires novel fluorophore conjugates that have an appropriate size for long circulation and diffusion and show virtually no nonspecific binding to cells/serum while binding to cells of interest with high specificity. In addition, these conjugates must be stable and maintain a high quantum yield in the in vivo environments. Here, we show that this can be achieved using compact (~15 nm in hydrodynamic diameter) and biocompatible quantum dot (QD) -Ab conjugates. We developed these conjugates by coupling whole mAbs to QDs coated with norbornene-displaying polyimidazole ligands using tetrazine–norbornene cycloaddition. Our QD immunoconstructs were used for in vivo single-cell labeling in bone marrow. The intravital imaging studies using a chronic calvarial bone window showed that our QD-Ab conjugates diffuse into the entire bone marrow and efficiently label single cells belonging to rare populations of hematopoietic stem and progenitor cells (Sca1[superscript +]c-Kit[superscript +] cells). This in vivo cytometric technique may be useful in a wide range of structural and functional imaging to study the interactions between cells and between a cell and its environment in intact and diseased tissues.National Institutes of Health (U.S.) (Grant U54-CA151884)National Institutes of Health (U.S.) (Grant P41-EB015871-26A1)Samsung Scholarship Foundation (Graduate Student Fellowship)Massachusetts Institute of Technology. Institute for Soldier Nanotechnologies (Grant W911NF-07-D-0004

    Nanoparticles Targeted With NGR Motif Deliver c-myc siRNA and Doxorubicin for Anticancer Therapy

    Get PDF
    We have designed a PEGylated LPD (liposome-polycation-DNA) nanoparticle for systemic, specific, and efficient delivery of small interfering RNA (siRNA) into solid tumors in mice by modification with NGR (aspargine–glycine–arginine) peptide, targeting aminopeptidase N (CD13) expressed in the tumor cells or tumor vascular endothelium. LPD-PEG-NGR efficiently delivered siRNA to the cytoplasm and downregulated the target gene in the HT-1080 cells but not CD13− HT-29 cells, whereas nanoparticles containing a control peptide, LPD-PEG-ARA, showed only little siRNA uptake and gene silencing activity. LPD-PEG-NGR efficiently delivered siRNA into the cytoplasm of HT-1080 xenograft tumor 4 hours after intravenous injection. Three daily injections (1.2 mg/kg) of c-myc siRNA formulated in the LPD-PEG-NGR effectively suppressed c-myc expression and triggered cellular apoptosis in the tumor, resulting in a partial tumor growth inhibition. When doxorubicin (DOX) and siRNA were co-formulated in LPD-PEG-NGR, an enhanced therapeutic effect was observed

    Nanoparticles Modified With Tumor-targeting scFv Deliver siRNA and miRNA for Cancer Therapy

    Get PDF
    Targeted delivery of RNA-based therapeutics for cancer therapy remains a challenge. We have developed a LPH (liposome-polycation-hyaluronic acid) nanoparticle formulation modified with tumor-targeting single-chain antibody fragment (scFv) for systemic delivery of small interfering RNA (siRNA) and microRNA (miRNA) into experimental lung metastasis of murine B16F10 melanoma. The siRNAs delivered by the scFv targeted nanoparticles efficiently downregulated the target genes (c-Myc/MDM2/VEGF) in the lung metastasis. Two daily intravenous injections of the combined siRNAs in the GC4-targeted nanoparticles significantly reduced the tumor load in the lung. miRNA-34a (miR-34a) induced apoptosis, inhibited survivin expression, and downregulated MAPK pathway in B16F10 cells. miR-34a delivered by the GC4-targeted nanoparticles significantly downregulated the survivin expression in the metastatic tumor and reduced tumor load in the lung. When miR-34a and siRNAs were co-formulated in GC4-targeted nanoparticles, an enhanced anticancer effect was observed
    corecore