Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Mol Pharm ; 18(5): 2032-2038, 2021 05 03.
Article in English | MEDLINE | ID: mdl-33877834

ABSTRACT

Triple negative breast cancer (TNBC) remains one of the most challenging subtypes of breast cancer to treat and is responsible for approximately 12% of breast cancer cases in the US per year. In 2019, the protein Tinagl1 was identified as a key factor for improved prognoses in certain TNBC patients. While the intracellular mechanism of action has been thoroughly studied, little is known about the role of Tinagl1 in the tumor microenvironment. In this study, we developed a lipid nanoparticle-based gene therapy to directly target the expression of Tinagl1 in tumor cells for localized expression. Additionally, we sought to characterize the changes to the tumor microenvironment induced by Tinagl1 treatment, with the goal of informing future choices for combination therapies including Tinagl1. We found that Tinagl1 gene therapy was able to slow tumor growth from the first dose and that the effects held steady for nearly a week following the final dose. No toxicity was found with this treatment. Additionally, the use of Tinagl1 increases the tumor vasculature by 3-fold but does not increase the tumor permeability or risk of metastasis. However, the increase in vasculature arising from Tinagl1 therapy reduced the expression of Hif1a significantly (p < 0.01), which may decrease the risk of drug resistance.


Subject(s)
Extracellular Matrix Proteins/genetics , Genetic Therapy/methods , Lipocalins/genetics , Nanoparticles/chemistry , Plasmids/administration & dosage , Triple Negative Breast Neoplasms/therapy , Animals , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Liposomes , Mammary Glands, Animal/pathology , Mice , Plasmids/genetics , Recombinant Proteins/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment/genetics
2.
Nanoscale ; 12(5): 3400-3410, 2020 Feb 07.
Article in English | MEDLINE | ID: mdl-31989142

ABSTRACT

Highly fibrotic and collagen-rich properties in desmoplastic melanoma (DM) result in an immune-suppressive fibrotic tumor microenvironment (TME) that resists clinical therapies. The different clinical and pathological properties, as compared to conventional melanoma, lead to delayed diagnosis and it is difficult to deliver drugs effectively due to fibrosis. Herein, we designed a chemo-immuno strategy focused on combining vaccination immunotherapy with multi-targeting sunitinib (SUN) nano-therapy to remodel TME and generate a robust immune response and a stronger synergistic anti-cancer effect. This strategy was evaluated side-by-side with non-desmoplastic melanoma and achieved significant improvement in therapeutic efficacy. The combination treatment was also synergistically assessed with the desmoplastic melanoma model. This strategy can remodel the fibrotic immunosuppressive TME and result in a robust cytotoxic T-cell response by reducing the collagen content, normalizing blood vessels, inhibiting tumor-associated fibroblasts and reducing high levels of suppressor immune cells. The modification of fibrotic immunosuppressive TME may serve as a good approach to further enhance immunotherapy for desmoplastic tumors.


Subject(s)
Cancer Vaccines , Immunotherapy , Melanoma, Experimental , Melanoma , Skin Neoplasms , Sunitinib/pharmacology , Tumor Microenvironment , Animals , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Cell Line, Tumor , Melanoma/immunology , Melanoma/pathology , Melanoma/therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Melanoma, Cutaneous Malignant
3.
Chem Soc Rev ; 48(10): 2698-2737, 2019 May 20.
Article in English | MEDLINE | ID: mdl-31080987

ABSTRACT

Calcium phosphates (CaPs) are ubiquitous in nature and vertebrate bones and teeth, and have high biocompatibility and promising applications in various biomedical fields. Nanostructured calcium phosphates (NCaPs) are recognized as promising nanocarriers for drug/gene/protein delivery owing to their high specific surface area, pH-responsive degradability, high drug/gene/protein loading capacity and sustained release performance. In order to control the structure and surface properties of NCaPs, various biomolecules with high biocompatibility such as nucleic acids, proteins, peptides, liposomes and phosphorus-containing biomolecules are used in the synthesis of NCaPs. Moreover, biomolecules play important roles in the synthesis processes, resulting in the formation of various NCaPs with different sizes and morphologies. At room temperature, biomolecules can play the following roles: (1) acting as a biocompatible organic phase to form biomolecule/CaP hybrid nanostructured materials; (2) serving as a biotemplate for the biomimetic mineralization of NCaPs; (3) acting as a biocompatible modifier to coat the surface of NCaPs, preventing their aggregation and increasing their colloidal stability. Under heating conditions, biomolecules can (1) control the crystallization process of NCaPs by forming biomolecule/CaP nanocomposites before heating; (2) prevent the rapid and disordered growth of NCaPs by chelating with Ca2+ ions to form precursors; (3) provide the phosphorus source for the controlled synthesis of NCaPs by using phosphorus-containing biomolecules. This review focuses on the important roles of biomolecules in the synthesis of NCaPs, which are expected to guide the design and controlled synthesis of NCaPs. Moreover, we will also summarize the biomedical applications of NCaPs in nanomedicine and tissue engineering, and discuss their current research trends and future prospects.


Subject(s)
Calcium Phosphates/chemistry , Nanocomposites/chemistry , Biocompatible Materials/chemistry , DNA, Single-Stranded/chemistry , Drug Carriers/chemistry , Green Chemistry Technology , Humans , Nanomedicine , Serum Albumin/chemistry , Tissue Engineering
4.
Nucleic Acid Ther ; 29(2): 61-66, 2019 04.
Article in English | MEDLINE | ID: mdl-30562145

ABSTRACT

Nucleic acid-based therapeutics like synthetic small interfering RNAs have been exploited to modulate gene function, taking advantage of RNA interference (RNAi), an evolutionally conserved biological process. Recently, the world's first RNAi drug was approved for a rare genetic disorder in the liver. However, there are significant challenges that need to be resolved before RNAi can be translated in other genetic diseases like cancer. Current drug delivery platforms for therapeutic silencing RNAs are tailored to hepatic targets. RNAi therapies for nonhepatic conditions are still at early clinical phases. In this study, we discuss the critical design considerations in anticancer RNAi drug development, insights gained from initial clinical trials, and new strategies that are entering clinical development, shaping the future of RNAi in cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , RNA Interference , RNA, Small Interfering/therapeutic use , Clinical Trials as Topic , Drug Delivery Systems , Gene Silencing/drug effects , Humans , Neoplasms/genetics , RNA, Small Interfering/genetics
5.
ACS Nano ; 12(12): 11740-11755, 2018 12 26.
Article in English | MEDLINE | ID: mdl-30508378

ABSTRACT

Nanoscience has long been lauded as a method through which tumor-associated barriers could be overcome. As successful as cancer immunotherapy has been, limitations associated with the tumor microenvironment or side effects of systemic treatment have become more apparent. In this Review, we seek to lay out the therapeutic challenges associated with the tumor microenvironment and the ways in which nanoscience is being applied to remodel the tumor microenvironment and increase the susceptibility of many cancer types to immunotherapy. We detail the nanomedicines on the cutting edge of cancer immunotherapy and how their interactions with the tumor microenvironment make them more effective than systemically administered immunotherapies.


Subject(s)
Immunotherapy/methods , Nanoparticles/chemistry , Neoplasms/therapy , Tumor Microenvironment , Animals , Apoptosis , Cancer Vaccines/therapeutic use , Drug Delivery Systems/methods , Humans , Neoplasms/immunology
7.
Mol Ther ; 26(1): 45-55, 2018 01 03.
Article in English | MEDLINE | ID: mdl-29258739

ABSTRACT

Triple negative breast cancer (TNBC), which constitutes 10%-20% of all breast cancers, is associated with aggressive progression, a high rate of metastasis, and poor prognosis. The treatment of patients with TNBC remains a great clinical challenge. Preclinical reports support the combination immunotherapy of cancer vaccines and immune checkpoint blockades in non-immunogenic tumors. In this study, we constructed nanoparticles (NPs) to deliver an mRNA vaccine encoding tumor antigen MUC1 to dendritic cells (DCs) in lymph nodes to activate and expand tumor-specific T cells. An anti-CTLA-4 (cytotoxic T-lymphocyte-associated protein 4) monoclonal antibody was combined with the mRNA vaccine to enhance the anti-tumor benefits. In vivo studies demonstrated that the NP-based mRNA vaccine, targeted to mannose receptors on DCs, could successfully express tumor antigen in the DCs of the lymph node; that the NP vaccine could induce a strong, antigen-specific, in vivo cytotoxic T lymphocyte response against TNBC 4T1 cells; and that combination immunotherapy of the vaccine and anti-CTLA-4 monoclonal antibody could significantly enhance anti-tumor immune response compared to the vaccine or monoclonal antibody alone. These data support both the NP as a carrier for delivery of mRNA vaccine and a potential combination immunotherapy of the NP-based mRNA vaccine and the CTLA-4 inhibitor for TNBC.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , CTLA-4 Antigen/antagonists & inhibitors , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Mucin-1/genetics , Mucin-1/immunology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology , Animals , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Cell Line, Tumor , Combined Modality Therapy , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Female , Humans , Immunotherapy , Interferon-gamma/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Recombinant Fusion Proteins , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
8.
Cancer Immunol Immunother ; 67(2): 299-310, 2018 02.
Article in English | MEDLINE | ID: mdl-29094184

ABSTRACT

Approximately, 50% of human melanomas are driven by BRAF mutations, which produce tumors that are highly immunosuppressive and often resistant to vaccine therapy. We introduced lipid-coated calcium phosphate nanoparticles (LCP NPs) as a carrier to efficiently deliver a tumor-specific antigen, the BRAFV600E peptide, to drive dendritic cell (DC) maturation and antigen presentation in C57BL6 mice. The BRAF peptide vaccine elicited a robust, antigen-specific cytotoxic T cell response and potent tumor growth inhibition in a murine BRAF-mutant melanoma model. Advanced BRAF-specific immune response was illustrated by IFN-γ production assay and cytotoxic T lymphocyte (CTL) assay. Remodeling of immunosuppressive modules within the tumor microenvironment further facilitated CTL infiltration. Thus, using LCP NPs to deliver the BRAF peptide vaccine is a promising strategy for the BRAF-mutant melanoma therapy.


Subject(s)
Cancer Vaccines/pharmacology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Proto-Oncogene Proteins B-raf/immunology , Animals , Cancer Vaccines/immunology , Cell Line, Tumor , Female , Melanoma, Experimental/genetics , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins B-raf/genetics , Tumor Microenvironment/immunology
9.
Acta Biomater ; 65: 405-416, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29037897

ABSTRACT

Oral administration of nanocarriers remains a significant challenge in the pharmaceutical sciences. The nanocarriers must efficiently overcome multiple gastrointestinal barriers including the harsh gastrointestinal environment, the mucosal layer, and the epithelium. Neutral hydrophilic surfaces are reportedly necessary for mucus permeation, but hydrophobic and cationic surfaces are important for efficient epithelial absorption. To accommodate these conflicting surface property requirements, we developed a strategy to modify nanocarrier surfaces with cationic cell-penetrating peptides (CPP) concealed by a hydrophilic succinylated casein (SCN) layer. SCN is a mucus-inert natural material specifically degraded in the intestine, thus protecting nanocarriers from the harsh gastric environment, facilitating their mucus permeation, and inducing exposure of CPPs after degradation for further effective transepithelial transport. Quantum dots doped hollow silica nanoparticles (HSQN) with a diameter around 180 nm was used as the nanocarrier and demonstrated as high as 50% loading efficacy of paclitaxel, a model drug with poor solubility and permeability. The dual layer modification strategy prevented premature drug leakage in stomach and maintained high mucus permeation (the trajectory spanned 9-fold larger area than single CPP modification). After intestinal degradation of SCN by trypsin, these nanocarriers exhibited strong interaction with epithelial membranes and a 5-fold increase in cellular uptake. Significant transepithelial transport and intestinal distribution were also observed for this dual-modified formulation. A pharmacokinetics study on the paclitaxel-loaded nanocarrier found 40% absolute bioavailability and 7.8-fold higher AUC compared to oral Taxol®. Compared with single CPP modified nanocarriers, our formulation showed increased in vivo efficacy and tumor accumulation of the model drug with negligible intestinal toxicity. In summary, sequential modification with CPP and SCN layers on HSQN offers a potential strategy to overcome the multiple barriers of the gastrointestinal tract. STATEMENT OF SIGNIFICANCE: Oral administration of nanocarriers remains a big challenge due to the multiple gastrointestinal barriers. In order to achieve both strong mucus permeation and efficient epithelial absorption, we modified the surface of silica nanoparticles with two layers: cell penetrating peptide (CPP) layer and succinylated casein (SCN) layer. The newly developed nanoformulations are demonstrated to have the following advantages: 1) versatile carrier with easy preparation, 2) high drug loading especially for poor soluble molecules, 3) reduced drug leakage in the stomach, 4) effective mucus penetration and transepithelial transport and 5) good biocompatibility, which in all indicate a great potential of this bilayer-modification strategy to facilitate the oral delivery of therapeutic agents.


Subject(s)
Drug Carriers/chemistry , Gastrointestinal Tract/metabolism , Nanoparticles/chemistry , Silicon Dioxide/chemistry , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Biocompatible Materials , Biological Availability , Caseins/chemistry , Cell Line, Tumor , Cell-Penetrating Peptides/chemistry , Drug Carriers/pharmacokinetics , Female , Humans , Hydrophobic and Hydrophilic Interactions , Intestinal Mucosa/metabolism , Mice, Inbred BALB C , Mucus/metabolism , Paclitaxel/administration & dosage , Paclitaxel/pharmacokinetics , Porosity , Quantum Dots
10.
Biomaterials ; 148: 16-30, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28961532

ABSTRACT

Cancer immunotherapy is quickly growing to be the fourth most important cancer therapy, after surgery, radiation therapy, and chemotherapy. Immunotherapy is the most promising cancer management strategy because it orchestrates the body's own immune system to target and eradicate cancer cells, which may result in durable antitumor responses and reduce metastasis and recurrence more than traditional treatments. Nanomaterials hold great promise in further improving the efficiency of cancer immunotherapy - in many cases, they are even necessary for effective delivery. In this review, we briefly summarize the basic principles of cancer immunotherapy and explain why and where to apply nanomaterials in cancer immunotherapy, with special emphasis on cancer vaccines and tumor microenvironment modulation.


Subject(s)
Immunotherapy/methods , Nanostructures/chemistry , Neoplasms/diagnosis , Neoplasms/therapy , Animals , Antineoplastic Agents/therapeutic use , Cancer Vaccines/immunology , Cell Line, Tumor , Humans , Neoplasms/immunology , Tumor Microenvironment/immunology
11.
Mol Ther ; 25(7): 1567-1579, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28274796

ABSTRACT

The success of small interfering RNA (siRNA)-mediated gene silencing for cancer therapy is still limited because of its instability and poor intracellular internalization. Traditional cationic carriers cannot adequately meet the need for clinical application of siRNA. We herein report a dual-functional liposome containing a cholesterol derivative of metformin, i.e., LipoMET, which takes advantage of the fusogenic activity as well as intrinsic tumor apoptosis inducing ability of biguanide moiety to achieve a combinational anti-oncogenic effect. In this study, the vascular endothelial growth factor (VEGF)-specific siRNAs were first electrostatically condensed into a ternary nanocomplex composed of polycation and hyaluronate, which was subsequently enveloped by LipoMET through membrane fusion. In comparison with common cationic control group, the resulting envelope-type nanoparticles (PH@LipoMET nanoparticles [NPs]) showed the ability of rapid cellular internalization and effective endosomal escape of siRNA during intracellular trafficking studies. Systemic administration of the targeted LipoMETs was capable of inducing apoptosis and tumor growth inhibition in the NCI-H460 xenograft model. When carrying VEGF-specific siRNAs, PH@LipoMET NPs remarkably downregulated the expression of VEGF and led to even more tumor suppression in vivo. Thus, LipoMET originated envelope-type nanoparticles may serve as a potential dual-functional siRNA delivery system to improve therapeutic effect of oncogene silencing.


Subject(s)
Carcinoma, Non-Small-Cell Lung/therapy , Gene Expression Regulation, Neoplastic , Liposomes/administration & dosage , Lung Neoplasms/therapy , Metformin/pharmacology , RNA, Small Interfering/genetics , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cholesterol/chemistry , Cholesterol/metabolism , Endosomes/metabolism , Female , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/metabolism , Liposomes/chemistry , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Metformin/analogs & derivatives , Metformin/metabolism , Mice , Mice, Nude , RNA, Small Interfering/chemistry , RNA, Small Interfering/metabolism , Receptors, sigma/genetics , Receptors, sigma/metabolism , Survival Analysis , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
12.
Sci Transl Med ; 8(364): 364ra153, 2016 11 09.
Article in English | MEDLINE | ID: mdl-27831902

ABSTRACT

The liver is the primary site of metastasis for gastrointestinal cancers and is a location highly susceptible to the establishment of metastasis in numerous other primary cancers, including breast, lung, and pancreatic cancers. The current standard of care typically consists of primary tumor resection and systemic administration of potent but toxic chemotherapeutics, yielding a minimal improvement in the median survival rate. CXCL12, a chemokine, is a key factor for activating the migration/survival pathways of CXCR4+ cancer cells and for recruiting immunosuppressive cells to areas of inflammation. Therefore, reducing CXCL12 concentrations within the liver has the potential to decrease tumor and immunosuppressive cell activation/migration within the liver. However, because of off-target toxicities associated with systemic administration of anti-CXCL12 therapies, transient and liver-specific expression of a CXCL12 trap is necessary. To address this challenge, we developed a lipid calcium phosphate nanoparticle optimized for delivering plasmid DNA, encoding an engineered CXCL12 protein trap, to the nucleus of liver hepatocytes. This pCXCL12-trap formulation yielded transient (4 days) liver-specific expression, which greatly decreased the occurrence of liver metastasis in two aggressive liver metastasis models, including colorectal [CT-26(FL3)] and breast (4T1) cancers. Subsequent studies in an aggressive human colorectal liver metastasis model (HT-29) decreased the establishment of liver metastasis more effectively than did systemic administration of the CXCL12 protein trap and to a level comparable to a high-dose regimen of a potent CXCR4 antagonist (AMD3100).


Subject(s)
Chemokine CXCL12/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/secondary , Molecular Targeted Therapy , Neoplasm Metastasis/prevention & control , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , DNA/chemistry , Disease Models, Animal , Female , Gene Expression Profiling , HEK293 Cells , Humans , Immunosuppressive Agents/therapeutic use , Inflammation , Interferometry , Male , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Neoplasm Invasiveness , Recombinant Proteins/chemistry , Signal Transduction
13.
Nano Lett ; 16(9): 5401-8, 2016 09 14.
Article in English | MEDLINE | ID: mdl-27490088

ABSTRACT

Chemotherapeutic efficacy can be greatly improved by developing nanoparticulate drug delivery systems (nano-DDS) with high drug loading capacity and smart stimulus-triggered drug release in tumor cells. Herein, we report a novel redox dual-responsive prodrug-nanosystem self-assembled by hydrophobic small-molecule conjugates of paclitaxel (PTX) and oleic acid (OA). Thioether linked conjugates (PTX-S-OA) and dithioether inserted conjugates (PTX-2S-OA) are designed to respond to the redox-heterogeneity in tumor. Dithioether has been reported to show redox dual-responsiveness, but we find that PTX-S-OA exhibits superior redox sensitivity over PTX-2S-OA, achieving more rapid and selective release of free PTX from the prodrug nanoassemblies triggered by redox stimuli. PEGylated PTX-S-OA nanoassemblies, with impressively high drug loading (57.4%), exhibit potent antitumor activity in a human epidermoid carcinoma xenograft. This novel prodrug-nanosystem addresses concerns related to the low drug loading and inefficient drug release from hydrophobic prodrugs of PTX, and provides possibilities for the development of redox dual-sensitive conjugates or polymers for efficient anticancer drug delivery.


Subject(s)
Drug Carriers , Fatty Acids/chemistry , Nanoconjugates , Paclitaxel/administration & dosage , Prodrugs/administration & dosage , Animals , Cell Line, Tumor , Drug Liberation , Humans , Oxidation-Reduction , Rats, Sprague-Dawley , Sulfides
14.
Biomaterials ; 102: 239-48, 2016 09.
Article in English | MEDLINE | ID: mdl-27344367

ABSTRACT

Metformin (dimethylbiguanide) has been found to be effective for the treatment of a wide range of cancer. Herein, a novel lipid (1,2-di-(9Z-octadecenoyl)-3-biguanide-propane (DOBP)) was elaborately designed by utilizing biguanide as the cationic head group. This novel cationic lipid was intended to act as a gene carrier with intrinsic antitumor activity. When compared with 1,2-di-(9Z-octadecenoyl)-3-trimethylammonium-propane (DOTAP), a commercially available cationic lipid with a similar structure, the blank liposomes consisting of DOBP showed much more potent antitumor effects than DOTAP in human lung tumor xenografts, following an antitumor mechanism similar to metformin. Given its cationic head group, biguanide, DOBP could encapsulate TNF-related apoptosis-inducing ligand (TRAIL) plasmids into Lipid-Protamine-DNA (LPD) nanoparticles (NPs) for systemic gene delivery. DOBP-LPD-TRAIL NPs demonstrated distinct superiority in delaying tumor progression over DOTAP-LPD-TRAIL NPs, due to the intrinsic antitumor activity combined with TRAIL-induced apoptosis in the tumor. These results indicate that DOBP could be used as a versatile and promising cationic lipid for improving the therapeutic index of gene therapy in cancer treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Biguanides/therapeutic use , Carcinoma, Non-Small-Cell Lung/therapy , DNA/therapeutic use , Liposomes/therapeutic use , Lung Neoplasms/therapy , TNF-Related Apoptosis-Inducing Ligand/genetics , Animals , Antineoplastic Agents/chemistry , Biguanides/chemistry , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line , Cell Line, Tumor , DNA/administration & dosage , DNA/genetics , Female , Gene Transfer Techniques , Genetic Therapy , Lipids/chemistry , Lipids/therapeutic use , Liposomes/chemistry , Lung Neoplasms/genetics , Mice, Nude
SELECTION OF CITATIONS
SEARCH DETAIL
...