Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Int J Nanomedicine ; 16: 7697-7709, 2021.
Article in English | MEDLINE | ID: mdl-34819728

ABSTRACT

INTRODUCTION: ß-lactamase (LACTB) is a tumor suppressor gene in various tumors including melanoma. However, it remains challenging to efficiently deliver the LACTB gene into melanoma. Recently, we designed a nonviral nanocarrier iRGD/DOTAP/MPEG-PDLLA (iDPP) that could deliver gene targetedly to melanoma efficiently without obvious adverse effects. METHODS: In this study, the tumor-targeted nanoparticle iDPP was prepared to deliver LACTB gene to treat melanoma in vitro and in vivo. First, the expression level of LACTB in 6 clinical specimens of melanoma patients was evaluated. Subsequently, the characteristics of iDPP/LACTB nanocomplexes were studied. Afterwards, the in vitro and in vivo anti-tumor efficacy of the iDPP/LACTB nanocomplexes were explored utilizing the B16-F10 mouse melanoma cell line and the B16-F10 subcutaneous melanoma model. RESULTS: Compared with the normal epithelium, the expression level of LACTB in melanoma tissues was significantly downregulated. In vitro B16-F10 cell tests showed iDPP/LACTB nanocomplexes could increase the mRNA levels of P21, Bid, Bax, Pidd1, and Sival genes and up-regulate the p53 signaling pathway of melanoma cells, thus promoting cell apoptosis and blocking the cell cycle. Injected intravenously, iDPP nanoparticles could deliver DNA to the subcutaneous melanoma targetedly. Based on in vivo mouse xenograft model, iDPP/LACTB nanocomplexes could effectively inhibit tumor proliferation and induce tumor apoptosis, thus significantly inhibiting melanoma growth (tumor inhibition rate is about 68%) in the subcutaneous B16-F10 melanoma model. CONCLUSION: The downregulated LACTB might be a potential target for melanoma therapy. The iDPP/LACTB nanocomplexes could inhibit the growth of the mouse melanoma without obvious side effects, which provide a new option for melanoma gene therapy research.


Subject(s)
Antineoplastic Agents , Melanoma, Experimental , Nanoparticles , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Genetic Therapy , Humans , Melanoma, Experimental/drug therapy , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/genetics , beta-Lactamases/pharmacology , beta-Lactamases/therapeutic use
2.
Int J Nanomedicine ; 16: 2833-2847, 2021.
Article in English | MEDLINE | ID: mdl-33883894

ABSTRACT

INTRODUCTION: Peptides can be rationally designed as non-covalent inhibitors for molecularly targeted therapy. However, it remains challenging to efficiently deliver the peptides into the targeted cells, which often severely affects their therapeutic efficiency. METHODS: Herein, we created a novel non-covalent peptide inhibitor against nuclear export factor CRM1 by a structure-guided drug design method and targetedly delivered the peptide into cancer cells by a nanoparticle-mediated gene expression system for use as a cancer therapy. RESULTS: The nuclear export signal (NES)-optimized CRM1 peptide inhibitor colocalized with CRM1 to the nuclear envelope and inhibited nuclear export in cancer cell lines in vitro. The crystal structures of the inhibitors complexed with CRM1 were solved. In contrast to the covalent inhibitors, the peptides were similarly effective against cells harboring the CRM1 C528S mutation. Moreover, a plasmid encoding the peptides was delivered by a iRGD-modified nanoparticle to efficiently target and transfect the cancer cells in vivo after intravenous administration. The peptides could be selectively expressed in the tumor, resulting in the efficient inhibition of subcutaneous melanoma xenografts without obvious systemic toxicity. DISCUSSION: This work provides an effective strategy to design peptide-based molecularly targeted therapeutics, which could lead to the development of future targeted therapy.


Subject(s)
Intracellular Space/metabolism , Karyopherins/antagonists & inhibitors , Melanoma, Experimental/drug therapy , Nanoparticles/chemistry , Peptides/pharmacology , Peptides/therapeutic use , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , A549 Cells , Active Transport, Cell Nucleus/drug effects , Amino Acid Sequence , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Gene Transfer Techniques , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Karyopherins/chemistry , Karyopherins/metabolism , Melanoma, Experimental/pathology , Mutant Proteins/metabolism , Mutation/genetics , Nanoparticles/ultrastructure , Nuclear Export Signals , Peptides/chemistry , Protein Binding/drug effects , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Viral Nonstructural Proteins/chemistry , Exportin 1 Protein
4.
ACS Appl Mater Interfaces ; 11(40): 36371-36382, 2019 Oct 09.
Article in English | MEDLINE | ID: mdl-31490057

ABSTRACT

Combination of chemotherapeutics and immunomodulators can generate synergistic anticancer efficacy, exerting efficient chemoimmunotherapy for cancer treatment. Nanoparticulate delivery systems hold great promise to promote synergistic anticancer efficacy for the codelivery of drugs. However, there remain challenges to precisely coencapsulate and deliver combinational drugs at designed ratios due to the difference of compatibility between drugs and nanocarriers. In this study, coassembled nanoparticles of lipophilic prodrugs (LPs) were designed to codeliver chemotherapeutics and immunomodulators for cancer treatment. Such nanoassemblies (NAs) could act as platforms to ratiometrically coencapsulate chemotherapeutics and immunomodulators. Based on this method, NAs formed by the self-assembly of iRGD peptide derivatives, paclitaxel (PTX) LPs, and imiquimod (R837) LPs were demonstrated to target the tumor at unified pharmacokinetics, further inducing the effective tumor inhibition and tumor recurrence prevention. This work provided an alternative to prepare chemoimmunotherapeutic NAs with advantages of ratiometric drug coencapsulation and unified pharmacokinetics, which may advance the future cancer chemoimmunotherapy.


Subject(s)
Drug Delivery Systems/methods , Drug Therapy , Immunotherapy , Nanoparticles/chemistry , Neoplasms/therapy , Animals , Antigen Presentation , Apoptosis/drug effects , Cell Line, Tumor , Drug Liberation , Female , Imiquimod/administration & dosage , Imiquimod/pharmacokinetics , Imiquimod/therapeutic use , Mice, Inbred BALB C , Nanoparticles/ultrastructure , Neoplasm Recurrence, Local/prevention & control , Neoplasms/drug therapy , Neoplasms/pathology , Paclitaxel/administration & dosage , Paclitaxel/pharmacokinetics , Paclitaxel/therapeutic use , Rats, Sprague-Dawley , Tissue Distribution
5.
Int J Nanomedicine ; 14: 3455-3468, 2019.
Article in English | MEDLINE | ID: mdl-31190803

ABSTRACT

Background: Phospholysine phosphohistidine inorganic pyrophosphate phosphatase (LHPP) is a novel tumor suppressor. However, whether LHPP is effective to melanoma has not been investigated. Gene therapy provides a new strategy for the treatment of melanoma. Currently, it suffers from the lack of safe and effective gene delivery systems. Methods: A CRGDKGPDC peptide (iRGD) modified hybrid monomethoxy poly(ethylene glycol)-poly(D,L-lactide) nanoparticle (iDPP) was prepared and complexed with a LHPP plasmid, forming an iDPP/LHPP nanocomplex. The iDPP/LHPP nanocomplex was characterized by particle size distribution, zeta potential, morphology, cytotoxicity, and transfection efficiency. The antitumor efficacy of the nanocomplex against melanoma was studied both in vitro and in vivo. Further, the potential epigenetic changes in melanoma induced by iDPP/LHPP nanocomplex were evaluated. Results: The iDPP/LHPP nanocomplex showed high transfection efficiency and low toxicity. Moreover, the nanocomplex displayed a neutral charge that can meet the requirement of intravenous injection for targeted gene therapy. In vitro and in vivo experiments indicated that the iDPP/LHPP nanocomplex significantly inhibited the melanoma growth without causing notable adverse effects. We also found that LHPP played an important role in epigenetics. It regulated the expression of genes related to the proliferation and apoptosis chiefly at the level of transcription. Conclusion: This work demonstrates that the iDPP nanoparticle-delivered LHPP gene has a potential application in melanoma therapy through regulation of the genes associated with epigenetics.


Subject(s)
Inorganic Pyrophosphatase/therapeutic use , Melanoma, Experimental/drug therapy , Nanoparticles/chemistry , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Epigenesis, Genetic/drug effects , Humans , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Nanoparticles/ultrastructure , Oligopeptides/chemistry , Organ Specificity/drug effects , Polyesters/chemistry , Polyethylene Glycols/chemistry
6.
Acta Biomater ; 90: 49-59, 2019 05.
Article in English | MEDLINE | ID: mdl-30930306

ABSTRACT

Nerve conduits provide an advanced tool for repairing the injured peripheral nerve that often causes disability and mortality. Currently, the efficiency of conduits in repairing peripheral nerve is unsatisfying. Here, we show a functional nanoparticle-enhanced nerve conduit for promoting the regeneration of peripheral nerves. This conduit, which consists of gelatin-methacryloyl (GelMA) hydrogels with drug loaded poly(ethylene glycol)- poly(3-caprolactone) (MPEG-PCL) nanoparticles dispersed in the hydrogel matrix, is rapidly fabricated by a continuous three-dimensional (3D) printing process. While the 3D-printed hydrogel conduit with customized size, shape and structure provides a physical microenvironment for axonal elongation, the nanoparticles sustained release the drug to facilitate the nerve regeneration. The drug, 4-((5,10-dimethyl-6-oxo-6,10-dihydro-5H-pyrimido[5,4-b]thieno[3,2-e][1,4]diazepin-2-yl)amino) benzenesulfonamide, is a Hippo pathway inhibitor with multiple functions including improving the proliferation and migration of Schwann cells and up-regulating neurotrophic factors genes. The descried functional nerve conduit efficiently induced the recovery of sciatic injuries in morphology, histopathology and functions in vivo, showing the potential clinical application in peripheral nerve repair. STATEMENTS OF SIGNIFICANCE: Functional nerve conduit provides a promising strategy alternative to autografts. In this work, we rapidly customized a nanoparticle-enhanced conduit by the continuous bioprinting process. This nanoparticle in the conduit can release a Hippo pathway inhibitor to facilitate the nerve regeneration and function restoration. The efficacy of the conduits is comparable to that of autograft, suggesting the potential clinical applications.


Subject(s)
Bioprinting , Nanoparticles/chemistry , Nerve Regeneration , Schwann Cells/metabolism , Sciatic Nerve/physiology , Tissue Scaffolds/chemistry , Animals , Cell Line , Gelatin/chemistry , Gelatin/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Male , Polyesters/chemistry , Polyesters/pharmacology , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Printing, Three-Dimensional , Rats , Rats, Sprague-Dawley
7.
J Biomed Nanotechnol ; 14(12): 2092-2101, 2018 Dec 01.
Article in English | MEDLINE | ID: mdl-30305216

ABSTRACT

Gene therapy is emerging as a promising tool for cancer treatment. Down-regulation of survivin gene can lead to the cancer inhibition. However, the lack of efficient and safe gene delivery system is still a critical obstacle to clinical gene therapy. In this study, we use a biodegradable nanoparticle to deliver human survivin-T34A (T34A) to dominant-negatively regulate survivin gene for ovarian cancer therapy. This nanoparticle, self-assembled from monomethoxy poly(ethylene glycol)-poly(D,L-lactide) (MPEG-PLA) copolymer and N-[1-(2,3-dioleoyloxy) propyl]-N,N,N-trimethylammonium chloride (DOTAP), has high transfection capability and negligible cytotoxicity. The nanoparticle-delivered T34A gene can efficiently inhibit the growth of SKOV3 ovarian cancer cells through induction of apoptosis in vitro. After intraperitoneal injection, the nanoparticle-delivered T34A gene significantly inhibited the growth of intraperitoneal metastasis of SKOV3 ovarian cancer, with no obvious adverse effects. Our data suggest that the nanoparticle-delivered T34A gene has promising clinical applications in ovarian cancer treatment.


Subject(s)
Nanoparticles , Ovarian Neoplasms , Apoptosis , Cell Line, Tumor , Female , Genetic Therapy , Humans , Inhibitor of Apoptosis Proteins , Survivin , Transfection
8.
Adv Sci (Weinh) ; 5(3): 1700263, 2018 03.
Article in English | MEDLINE | ID: mdl-29593949

ABSTRACT

Gene therapy provides a novel method for cancer therapy. This study shows a DNA nanocomplex that is inspired from vesicular stomatitis virus (VSV) for ovarian cancer therapy. This DNA nanocomplex consists of a cationized monomethoxy poly (ethylene glycol)-poly (d,l-lactide) (MPEG-PLA) nanoparticle and a plasmid encoding the matrix protein of vesicular stomatitis virus (VSVMP) that plays a critical role in the VSV-induced apoptosis of cancer cells. The cationized MPEG-PLA nanoparticle that is self-assembled by MPEG-PLA copolymer and N -[1-(2,3-dioleoyloxy) propyl]-N,N,N-trimethylammonium chloride (DOTAP) has low cytotoxicity and high transfection efficiency (>80%). Intraperitoneal administration of the p VSVMP nanocomplex remarkably inhibits the intraperitoneal metastasis of ovarian cancer and does not cause significant systemic toxicity. The apoptosis induction and anti-angiogenesis are involved in the anticancer mechanism. This work demonstrates a VSV-inspired DNA nanocomplex that has potential application for the treatment of intraperitoneal metastasis of ovarian cancer.

9.
Oncotarget ; 8(12): 20441-20451, 2017 Mar 21.
Article in English | MEDLINE | ID: mdl-28099901

ABSTRACT

The most efficient sequence of targeted agents for metastatic renal cell carcinoma patients has yet to be identified. Whether the sequence of sorafenib and sunitinib really matters is controversial and not answered clearly until now. This meta-analysis aims to estimate the efficacy of receptor tyrosine kinase inhibitors sorafenib-sunitinib and sunitinib-sorafenib for metastatic renal cell carcinoma, on the outcome of first-line progression-free survival, second-line progression-free survival, total progression-free survival and overall survival.We searched PubMed, Embase, Cochrane Library and ClinicalTrails.gov for eligible studies. Data were analyzed using random or fixed effects model depending on the heterogeneity of the eligible studies. Heterogeneity across studies were analyzed using Q and I2 statistics.Of 902 identified studies, ten were qualified in our analysis (N = 1732 patients). Sorafenib-sunitinib yielded no statistically significant benefit in first-line progression-free survival (fixed effects; HR = 0.95; 95%CI 0.75-1.21; p = 0.702), total progression-free survival (random effects; HR = 0.92; 95%CI 0.71-1.19; p = 0.531) and overall survival (fixed effects; HR = 0.89; 95%CI 0.72-1.09; p = 0.257), compared with sunitinib-sorafenib. Second-line progression-free survival was longer for sorafenib-sunitinib than sunitinib-sorafenib (fixed effects; HR = 0.55; 95%CI 0.44-0.68; p = 0.000).Sequential therapies with sorafenib and sunitinib is well tolerated and efficient in mRCC. However, there are no evidence supported that sorafenib-sunitinib has the superiority to sunitinib-sorafenib in sequence. The ideal sequence of targeted agents requires further elucidation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Renal Cell/drug therapy , Indoles/administration & dosage , Kidney Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/administration & dosage , Pyrroles/administration & dosage , Carcinoma, Renal Cell/mortality , Disease-Free Survival , Humans , Kidney Neoplasms/mortality , Niacinamide/administration & dosage , Sorafenib , Sunitinib , Treatment Outcome
10.
Acta Pharm Sin B ; 6(4): 319-28, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27471672

ABSTRACT

Considering that some antibacterial agents can identify the outer structure of pathogens like cell wall and/or cell membrane, we explored a self-enhanced targeted delivery strategy by which a small amount of the antibiotic molecules were modified on the surface of carriers as targeting ligands of certain bacteria while more antibiotic molecules were loaded inside the carriers, and thus has the potential to improve the drug concentration at the infection site, enhance efficacy and reduce potential toxicity. In this study, a novel targeted delivery system against methicillin-resistant Staphylococcus aureus (MRSA) pneumonia was constructed with daptomycin, a lipopeptide antibiotic, which can bind to the cell wall of S. aureus via its hydrophobic tail. Daptomycin was conjugated with N-hydroxysuccinimidyl-polyethylene glycol-1,2-distearoyl-sn-glycero-3-phosphoethanolamine to synthesize a targeting compound (Dapt-PEG-DSPE) which could be anchored on the surface of liposomes, while additional daptomycin molecules were encapsulated inside the liposomes. These daptomycin-modified, daptomycin-loaded liposomes (DPD-L[D]) showed specific binding to MRSA as detected by flow cytometry and good targeting capabilities in vivo to MRSA-infected lungs in a pneumonia model. DPD-L[D] exhibited more favorable antibacterial efficacy against MRSA than conventional PEGylated liposomal daptomycin both in vitro and in vivo. Our study demonstrates that daptomycin-modified liposomes can enhance MRSA-targeted delivery of encapsulated antibiotic, suggesting a novel drug delivery approach for existing antimicrobial agents.

SELECTION OF CITATIONS
SEARCH DETAIL
...