Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Control Release ; 192: 114-21, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-25016158

ABSTRACT

There is no effective clinical therapy yet for triple-negative breast cancer (TNBC) without particular human epidermal growth factor receptor-2, estrogen and progesterone receptor expression. In this study, we report a molecularly targeted and synthetic lethality-based siRNA therapy for TNBC treatment, using cationic lipid assisted poly(ethylene glycol)-b-poly(d,l-lactide) (PEG-PLA) nanoparticles as the siRNA carrier. It is demonstrated that only in c-Myc overexpressed TNBC cells, while not in normal mammary epithelial cells, delivery of siRNA targeting cyclin-dependent kinase 1 (CDK1) with the nanoparticle carrier (NPsiCDK1) induces cell viability decreasing and cell apoptosis through RNAi-mediated CDK1 expression inhibition, indicating the synthetic lethality between c-Myc with CDK1 in TNBC cells. Moreover, systemic delivery of NPsiCDK1 is able to suppress tumor growth in mice bearing SUM149 and BT549 xenograft and cause no systemic toxicity or activate the innate immune response, suggesting the therapeutic promise with such nanoparticles carrying siCDK1 for c-Myc overexpressed triple negative breast cancer.


Subject(s)
CDC2 Protein Kinase/genetics , Drug Carriers/chemistry , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , RNA Interference , RNA, Small Interfering/therapeutic use , Triple Negative Breast Neoplasms/therapy , Animals , Breast/metabolism , Breast/pathology , Cell Line, Tumor , Female , Genetic Therapy , Humans , Mice , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
2.
Biomaterials ; 35(26): 7622-34, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24929619

ABSTRACT

Systemic delivery of small interfering RNA (siRNA) into cancer cells remains the major obstacle to siRNA drug development. An ideal siRNA delivery vehicle for systemic administration should have long circulation time in blood, accumulate at tumor site, and sufficiently internalize into cancer cells for high-efficiency of gene silence. Herein, we report a core-shell Micelleplex delivery system that made from block copolymer bearing poly(ethylene glycol) (PEG), matrix metalloproteinase 2 (MMP-2)-degradable peptide PLG*LAG, cationic cell penetrating peptide polyarginine r9 and poly(ε-caprolactone) (PCL) for siRNA delivery. We show clear evidences in vitro and in vivo to prove that the micelle carrying siRNA can circulate enough time in blood, enrich accumulation at tumor sites, shed the PEG layer when triggered by tumor overexpressing MMP-2, and then the exposing cell penetrating peptide r9 enhanced cellular uptake of siRNA. Accordingly, this design strategy enhances the inhibition of breast tumor growth following systemic injection of this system carrying siRNA against Polo-like kinase 1, which demonstrating this Micelleplex can be a potential delivery system for systemic siRNA delivery in cancer therapy.


Subject(s)
Drug Carriers/metabolism , Matrix Metalloproteinase 2/metabolism , Peptides/metabolism , RNA, Small Interfering/administration & dosage , Transfection , Amino Acid Sequence , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Cell Cycle Proteins/genetics , Cell Line, Tumor , Drug Carriers/chemistry , Female , HEK293 Cells , Humans , Mice, Inbred BALB C , Mice, Nude , Micelles , Molecular Sequence Data , Peptides/chemistry , Polyethylene Glycols/chemistry , Polyethylene Glycols/metabolism , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Polo-Like Kinase 1
3.
Mol Ther ; 22(5): 964-73, 2014 May.
Article in English | MEDLINE | ID: mdl-24496383

ABSTRACT

The KRAS mutation is present in ~20% of lung cancers and has not yet been effectively targeted for therapy. This mutation is associated with a poor prognosis in non-small-cell lung carcinomas (NSCLCs) and confers resistance to standard anticancer treatment drugs, including epidermal growth factor receptor tyrosine kinase inhibitors. In this study, we exploited a new therapeutic strategy based on the synthetic lethal interaction between cyclin-dependent kinase 4 (CDK4) downregulation and the KRAS mutation to deliver micellar nanoparticles (MNPs) containing small interfering RNA targeting CDK4 (MNPsiCDK4) for treatment in NSCLCs harboring the oncogenic KRAS mutation. Following MNPsiCDK4 administration, CDK4 expression was decreased, accompanied by inhibited cell proliferation, specifically in KRAS mutant NSCLCs. However, this intervention was harmless to normal KRAS wild-type cells, confirming the proposed mechanism of synthetic lethality. Moreover, systemic delivery of MNPsiCDK4 significantly inhibited tumor growth in an A549 NSCLC xenograft murine model, with depressed expression of CDK4 and mutational KRAS status, suggesting the therapeutic promise of MNPsiCDK4 delivery in KRAS mutant NSCLCs via a synthetic lethal interaction between KRAS and CDK4.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Cyclin-Dependent Kinase 4/genetics , Genetic Therapy , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/genetics , ras Proteins/genetics , Animals , Carcinoma, Non-Small-Cell Lung/therapy , Cell Line, Tumor , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Gene Transfer Techniques , Humans , Mice , Nanoparticles/therapeutic use , Proto-Oncogene Proteins p21(ras) , RNA, Small Interfering/therapeutic use , Xenograft Model Antitumor Assays
4.
Biotechnol Adv ; 32(4): 789-803, 2014.
Article in English | MEDLINE | ID: mdl-23933109

ABSTRACT

pH-responsive nanoparticles (NPs) are currently under intense development as drug delivery systems for cancer therapy. Among various pH-responsiveness, NPs that are designed to target slightly acidic extracellular pH environment (pHe) of solid tumors provide a new paradigm of tumor targeted drug delivery. Compared to conventional specific surface targeting approaches, the pHe-targeting strategy is considered to be more general due to the common occurrence of acidic microenvironment in solid tumors. This review mainly focuses on the design and applications of pHe-activated NPs, with special emphasis on pHe-activated surface charge reversal NPs, for drug and siRNA delivery to tumors. The novel development of NPs described here offers great potential for achieving better therapeutic effects in cancer treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Carriers/chemistry , Drug Delivery Systems , Nanomedicine/methods , Nanoparticles/administration & dosage , Nanotechnology/methods , Neoplasms/drug therapy , Animals , Cell Line, Tumor , Endocytosis , Female , Humans , Hydrogen-Ion Concentration , MCF-7 Cells , Mice , Nanoparticles/chemistry , Neoplasm Transplantation , RNA, Small Interfering/metabolism , Surface Properties
5.
Biomaterials ; 35(2): 836-45, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24144908

ABSTRACT

Nanoparticle-mediated delivery of chemotherapies has demonstrated enhanced anti-cancer efficacy, mainly through the mechanisms of both passive and active targeting. Herein, we report other than these well-elucidated mechanisms, rationally designed nanoparticles can efficiently deliver drugs to cancer stem cells (CSCs), which in turn contributes significantly to the improved anti-cancer efficacy. We demonstrate that doxorubicin-tethered gold nanoparticles via a poly(ethylene glycol) spacer and an acid-labile hydrazone bond mediate potent doxorubicin delivery to breast CSCs, which reduces their mammosphere formation capacity and their cancer initiation activity, eliciting marked enhancement in tumor growth inhibition in murine models. The drug delivery mediated by the nanoparticles also markedly attenuates tumor growth during off-therapy stage by reducing breast CSCs in tumors, while the therapy with doxorubicin alone conversely evokes an enrichment of breast CSCs. Our findings suggest that with well-designed drug delivery system, the conventional chemotherapeutic agents are promising for cancer stem cell therapy.


Subject(s)
Doxorubicin/pharmacology , Drug Delivery Systems , Gold/chemistry , Nanoparticles/chemistry , Neoplastic Stem Cells/drug effects , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Female , Humans , MCF-7 Cells , Mice , Mice, Inbred NOD , Polyethylene Glycols/chemistry
6.
Adv Mater ; 24(40): 5476-80, 2012 Oct 23.
Article in English | MEDLINE | ID: mdl-22886872

ABSTRACT

Two faced nanoparticles: A zwitterionic polymer-based nanoparticle with response to tumor acidity is developed for enhanced drug delivery to tumors. The nanoparticles are neutrally charged at physiological conditions and show prolonged circulation time; after leaking into tumor sites, in the acidic extracellular tumor environment (pH(e) ), nanoparticles are activated and become positively charged and are therefore efficiently taken up by tumor cells, leading to enhanced therapeutic effects in cancer treatment.


Subject(s)
Drug Carriers/chemistry , Nanoparticles/chemistry , Polymers/chemistry , Animals , Cell Line, Tumor , Doxorubicin/administration & dosage , Humans , Hydrogen-Ion Concentration , Mice , Mice, Nude , Neoplasms/drug therapy , Polyesters/chemistry , Transplantation, Heterologous
7.
J Control Release ; 161(3): 875-83, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22762887

ABSTRACT

The targeted delivery of small interfering RNA (siRNA) to specific tumor tissues and tumor cells remains as one of the key challenges in the development of RNA interference as a therapeutic application. To target breast cancer, we developed a therapeutic delivery system using a fusion protein of an anti-Her2 single-chain antibody fragment with a positively charged protamine, namely F5-P, as the carrier to specifically deliver siRNA-targeting DNA methyltransferases 1 and/or 3b genes (siDNMTs) into Her2-expressing breast tumor cells. The carrier F5-P, expressed by the Escherichia coli system, was able to bind siRNA molecules and specifically deliver the siRNA to Her2-expressing BT474 breast cancer cells but not Her2-nonexpressing MDA-MB-231 breast cancer cells, while delivery of siDNMTs to BT474 cells successfully silenced the expression of targeted DNA methyltransferases (DNMTs) and facilitated the de-methylation of the RASSF1A tumor suppressor gene promoter, leading to the suppression of tumor cell proliferation. Moreover, as demonstrated in the BT474 xenograft murine model, F5-P successfully delivered siRNA into a Her2-expressing breast tumor, and tumor growth inhibition was mediated by an intravenous injection of F5-P/siDNMTs complex by down-regulating the expression of DNMTs and restoring tumor suppressor gene expression. These data suggest that the delivery of siDNMTs by F5-P could be used to treat Her2-expressing breast cancer.


Subject(s)
Breast Neoplasms/therapy , DNA Modification Methylases/genetics , Immunoglobulin Fragments/administration & dosage , Protamines/administration & dosage , RNA, Small Interfering/administration & dosage , Receptor, ErbB-2/immunology , Animals , Cell Line, Tumor , DNA Modification Methylases/metabolism , Female , Gene Silencing , Humans , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/administration & dosage , Xenograft Model Antitumor Assays
8.
ACS Nano ; 6(6): 4955-65, 2012 Jun 26.
Article in English | MEDLINE | ID: mdl-22646867

ABSTRACT

The clinical success of therapeutics of small interfering RNA (siRNA) is still hindered by its delivery systems. Cationic polymer or lipid-based vehicles as the major delivery systems of siRNA cannot sufficiently satisfy siRNA therapeutic applications. It is hypothesized that cationic lipid-polymer hybrid nanoparticles may take advantage of both polymeric and lipid-based nanoparticles for siRNA delivery, while diminishing the shortcomings of both. In this study, cationic lipid-polymer hybrid nanoparticles were prepared by a single-step nanoprecipitation of a cationic lipid (N,N-bis(2-hydroxyethyl)-N-methyl-N-(2-cholesteryloxycarbonyl aminoethyl) ammonium bromide, BHEM-Chol) and amphiphilic polymers for systemic delivery of siRNA. The formed hybrid nanoparticles comprised a hydrophobic polylactide core, a hydrophilic poly(ethylene glycol) shell, and a cationic lipid monolayer at the interface of the core and the shell. Such hybrid nanoparticles exhibited excellent stability in serum and showed significantly improved biocompatibility compared to that of pure BHEM-Chol particles. The hybrid nanoparticles were capable of delivering siRNA into BT474 cells and facilitated the escape of loaded siRNA from the endosome into the cytoplasm. The hybrid nanoparticles carrying polo-like kinase 1 (Plk1)-specific siRNA (siPlk1) remarkably and specifically downregulated expression of the oncogene Plk1 and induced cancer cell apoptosis both in vitro and in vivo and significantly suppressed tumor growth following systemic administration. We demonstrate that this system is stable, nontoxic, highly efficient, and easy to scale up, bringing the clinical application of siRNA therapy one important step closer to reality.


Subject(s)
Lipids/chemistry , Nanocapsules/chemistry , Neoplasms, Experimental/genetics , Neoplasms, Experimental/therapy , Polymers/chemistry , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/chemistry , Animals , Cations , Cell Line, Tumor , Genetic Therapy/methods , Mice , Nanocapsules/administration & dosage , Treatment Outcome
10.
Sci Transl Med ; 4(130): 130ra48, 2012 Apr 18.
Article in English | MEDLINE | ID: mdl-22517885

ABSTRACT

A major obstacle to developing small interfering RNAs (siRNAs) as cancer drugs is their intracellular delivery to disseminated cancer cells. Fusion proteins of single-chain fragmented antibodies (ScFvs) and positively charged peptides deliver siRNAs into specific target cells. However, the therapeutic potential of ScFv-mediated siRNA delivery has not been evaluated in cancer. Here, we tested whether Polo-like kinase 1 (PLK1) siRNAs complexed with a Her2-ScFv-protamine peptide fusion protein (F5-P) could suppress Her2(+) breast cancer cell lines and primary human cancers in orthotopic breast cancer models. PLK1-siRNAs transferred by F5-P inhibited target gene expression, reduced proliferation, and induced apoptosis of Her2(+) breast cancer cell lines and primary human cancer cells in vitro without triggering an interferon response. Intravenously injected F5-P/PLK1-siRNA complexes concentrated in orthotopic Her2(+) breast cancer xenografts and persisted for at least 72 hours, leading to suppressed PLK1 gene expression and tumor cell apoptosis. The intravenously injected siRNA complexes retarded Her2(+) breast tumor growth, reduced metastasis, and prolonged survival without evident toxicity. F5-P-mediated delivery of a cocktail of PLK1, CCND1, and AKT siRNAs was more effective than an equivalent dose of PLK1-siRNAs alone. These data suggest that F5-P could be used to deliver siRNAs to treat Her2(+) breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Cell Cycle Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Receptor, ErbB-2/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Line, Tumor , Cell Proliferation , Female , Humans , Mice , RNA, Small Interfering/genetics , RNA, Small Interfering/physiology , Single-Chain Antibodies/administration & dosage , Single-Chain Antibodies/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
11.
ACS Nano ; 6(1): 771-81, 2012 Jan 24.
Article in English | MEDLINE | ID: mdl-22136582

ABSTRACT

Drug delivery systems for cancer therapy usually need to be sterically stabilized by a poly(ethylene glycol) (PEG) layer during blood circulation to minimize nonspecific interactions with serum components. However, PEGylation significantly reduces cellular uptake of the delivery systems after they accumulate at the tumor site, which markedly impairs the in vivo antitumor efficiency. Here, we develop a ternary small interfering RNA (siRNA) delivery system with tumor acidity-activated sheddable PEG layer to overcome the challenge. The sheddable nanoparticle is fabricated by introducing a tumor acidity-responsive PEGylated anionic polymer to the surface of positively charged polycation/siRNA complexes via electrostatic interaction. We show clear evidence that introducing the PEGylated anionic polymer to the surface of a nanoparticle markedly reduces its nonspecific interactions with protein. We further demonstrate that the nanoparticle is capable of deshielding the PEG layer at the slightly acidic tumor extracellular microenvironment to facilitate the delivery of siRNA to the tumor cells after accumulation at the tumor site. Accordingly, this promotes the RNA-interfering efficiencies and enhances the inhibition of tumor growth. Such delivery system with the ability to deshield the PEG layer at the target tissues has remarkable potential in cancer therapy.


Subject(s)
Nanocapsules/chemistry , Neoplasms, Experimental/genetics , Neoplasms, Experimental/therapy , Polyethylene Glycols/chemistry , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Cell Line, Tumor , Humans , Hydrogen-Ion Concentration
12.
J Am Chem Soc ; 133(44): 17560-3, 2011 Nov 09.
Article in English | MEDLINE | ID: mdl-21985458

ABSTRACT

Efficient delivery of therapeutics into tumor cells to increase the intracellular drug concentration is a major challenge for cancer therapy due to drug resistance and inefficient cellular uptake. Herein, we have designed a tailor-made dual pH-sensitive polymer-drug conjugate nanoparticulate system to overcome the challenges. The nanoparticle is capable of reversing its surface charge from negative to positive at tumor extracellular pH (∼6.8) to facilitate cell internalization. Subsequently, the significantly increased acidity in subcellular compartments such as the endosome (∼5.0) further promotes doxorubicin release from the endocytosed drug carriers. This dual pH-sensitive nanoparticle has showed enhanced cytotoxicity in drug-resistant cancer stem cells, indicating its great potential for cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Doxorubicin/pharmacology , Drug Delivery Systems , Nanoparticles/chemistry , Polymers/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Doxorubicin/chemistry , Drug Design , Drug Screening Assays, Antitumor , Humans , Hydrogen-Ion Concentration , Polymers/chemical synthesis , Polymers/chemistry , Structure-Activity Relationship
13.
J Control Release ; 156(2): 203-11, 2011 Dec 10.
Article in English | MEDLINE | ID: mdl-21839126

ABSTRACT

Delivery of small interfering RNA (siRNA) has been one of the major hurdles for the application of RNA interference in therapeutics. Here, we describe a cationic lipid assisted polymeric nanoparticle system with stealthy property for efficient siRNA encapsulation and delivery, which was fabricated with poly(ethylene glycol)-b-poly(d,l-lactide), siRNA and a cationic lipid, using a double emulsion-solvent evaporation technique. By incorporation of the cationic lipid, the encapsulation efficiency of siRNA into the nanoparticles could be above 90% and the siRNA loading weight ratio was up to 4.47%, while the diameter of the nanoparticles was around 170 to 200nm. The siRNA retained its integrity within the nanoparticles, which were effectively internalized by cancer cells and escaped from the endosome, resulting in significant gene knockdown. This effect was demonstrated by significant down-regulation of luciferase expression in HepG2-luciferase cells which stably express luciferase, and suppression of polo-like kinase 1 (Plk1) expression in HepG2 cells, following delivery of specific siRNAs by the nanoparticles. Furthermore, the nanoparticles carrying siRNA targeting the Plk1 gene were found to induce remarkable apoptosis in both HepG2 and MDA-MB-435s cancer cells. Systemic delivery of specific siRNA by nanoparticles significantly inhibited luciferase expression in an orthotopic murine liver cancer model and suppressed tumor growth in a MDA-MB-435s murine xenograft model, suggesting its therapeutic promise in disease treatment.


Subject(s)
Nanoparticles/chemistry , Neoplasms/therapy , Polyethylene Glycols/chemistry , RNA Interference , RNA, Small Interfering/administration & dosage , Animals , Cell Cycle Proteins/genetics , Cell Line, Tumor , Genetic Therapy , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/ultrastructure , Neoplasms/genetics , Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/genetics , Polo-Like Kinase 1
14.
Biomaterials ; 32(11): 3124-33, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21277018

ABSTRACT

One of the key challenges in the development of RNA interference-based cancer therapy is the lack of an efficient delivery system for synthetic small interfering RNAs (siRNAs) that would enable efficient uptake by tumor cells and allow for significant knockdown of a target transcript in vivo. Here, we describe a micelleplex system based on an amphiphilic and cationic triblock copolymer, which can systemically deliver siRNA targeting the acid ceramidase (AC) gene for cancer therapy. This triblock copolymer, consisting of monomethoxy poly(ethylene glycol), poly(ε-caprolactone) and poly(2-aminoethyl ethylene phosphate), self-assembles into micellar nanoparticles (MNPs) in aqueous solution with an average diameter of 60 nm and a zeta potential of approximately 48 mV. The resulting micelleplex, formed by the interaction of MNPs and siRNA, was effectively internalized by BT474 breast cancer cells and siRNA was subsequently released, resulting in significant gene knockdown. This effect was demonstrated by significant down-regulation of luciferase expression in BT474-luciferase cells which stably express luciferase, and suppression of AC expression in BT474 cells at both the transcriptional and protein level, following delivery of specific siRNAs by the micelleplex. Furthermore, a micelleplex carrying siRNA targeting the AC (micelleplex(siAC)) gene was found to induce remarkable apoptosis and reduce the proliferation of cancer cells. Systemic delivery of micelleplex(siAC) significantly inhibited tumor growth in a BT474 xenograft murine model, with depressed expression of AC and no positive activation of the innate immune response, suggesting therapeutic promise for micelleplex siRNA delivery in cancer therapy.


Subject(s)
Biocompatible Materials/chemistry , Breast Neoplasms/therapy , Nanoparticles/chemistry , Polymers/chemistry , RNA, Small Interfering/genetics , Acid Ceramidase/genetics , Animals , Apoptosis/genetics , Blotting, Western , Breast Neoplasms/enzymology , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Mice , Mice, Nude , Micelles , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/chemistry , Reverse Transcriptase Polymerase Chain Reaction
15.
ACS Nano ; 5(2): 1483-94, 2011 Feb 22.
Article in English | MEDLINE | ID: mdl-21204585

ABSTRACT

Combination of two or more therapeutic strategies with different mechanisms can cooperatively prohibit cancer development. Combination of chemotherapy and small interfering RNA (siRNA)-based therapy represents an example of this approach. Hypothesizing that the chemotherapeutic drug and the siRNA should be simultaneously delivered to the same tumoral cell to exert their synergistic effect, the development of delivery systems that can efficiently encapsulate two drugs and successfully deliver payloads to targeted sites via systemic administration has proven to be challenging. Here, we demonstrate an innovative "two-in-one" micelleplex approach based on micellar nanoparticles of a biodegradable triblock copolymer poly(ethylene glycol)-b-poly(ε-caprolactone)-b-poly(2-aminoethyl ethylene phosphate) to systemically deliver the siRNA and chemotherapeutic drug. We show clear evidence that the micelleplex is capable of delivering siRNA and paclitaxel simultaneously to the same tumoral cells both in vitro and in vivo. We further demonstrate that systemic administration of the micelleplex carrying polo-like kinase 1 (Plk1) specific siRNA and paclitaxel can induce a synergistic tumor suppression effect in the MDA-MB-435s xenograft murine model, requiring a thousand-fold less paclitaxel than needed for paclitaxel monotherapy delivered by the micelleplex and without activation of the innate immune response or generation of carrier-associated toxicity.


Subject(s)
Micelles , Neoplasms/genetics , Neoplasms/metabolism , Paclitaxel/metabolism , Paclitaxel/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Animals , Base Sequence , Cell Cycle Proteins/genetics , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Carriers/chemistry , Drug Synergism , Female , Humans , Hydrophobic and Hydrophilic Interactions , Mice , Nanoparticles/chemistry , Neoplasms/pathology , Polymers/chemistry , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Time Factors , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
SELECTION OF CITATIONS
SEARCH DETAIL
...