Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Am J Emerg Med ; 38(10): 2153-2159, 2020 10.
Article in English | MEDLINE | ID: mdl-33071103

ABSTRACT

INTRODUCTION: Chinese medicine (CM) has been used to treat Novel Coronavirus 2019 (COVID-19) pneumonia in China. This meta-analysis was conducted to evaluate the clinical efficacy and safety of CM in the treatment of COVID-19 pneumonia. METHODS: Randomized controlled trials (RCTs) involving CM in the treatment of COVID-19 pneumonia were identified from Cochrane Central Register of Controlled Trials, PubMed, EMBASE, Chinese National Knowledge Infrastructure, Chinese Biomedical Database, Wanfang Database and VIP Information Database. The methodological quality of trials was evaluated with Cochrane Hanadbook criteria, and the Cochrane Collaboration's Review Manager 5.3 software was used for meta-analysis. RESULTS: A total of 7 valid studies involving 681 patients were included. The meta-analysis exhibited in comparison to conventional treatment, CM combined with conventional treatment significantly improved clinical efficacy (RR = 1.21, 95% CI [1.08,1.36]), and significantly increased viral nucleic acid negative conversion rate (RR = 1.49, 95% CI [1.13,1.97]). CM also prominently reduced pulmonary inflammation (RR = 1.27, 95% CI [1.12,1.44]), and improved host immune function (WBC, MD = 0.92, 95% CI [0.07,1.76]; LYM, MD = 0.33, 95% CI [0.08,0.57]; LYM%, MD = 2.90, 95% CI [2.09,3.71]; CRP, MD = -12.66, 95% CI [-24.40, -0.92]). Meanwhile, CM did not increase the incidence of adverse reactions (RR = 1.17, 95% CI [0.39,3.52]). CONCLUSION: According to the allocated data, CM has demonstrated clinical efficacy and safety on COVID-19 pneumonia, which need to be confirmed by high quality, multiple-center, large sample randomized controlled trials.


Subject(s)
COVID-19 Drug Treatment , Drugs, Chinese Herbal/therapeutic use , Humans , Randomized Controlled Trials as Topic , SARS-CoV-2/drug effects , Treatment Outcome
2.
J Mater Chem B ; 8(40): 9351-9361, 2020 10 21.
Article in English | MEDLINE | ID: mdl-32996552

ABSTRACT

Combined X-ray-induced photodynamic therapy (X-PDT) and chemotherapy are of great interest for tumor treatment, but their outcome is still hindered by insufficient drug delivery without tumor specificity and the difficulty of switching to chemotherapy during the X-PDT process. Herein, we report an efficient strategy for preparing a nanocarrier, DANPVP&DOX, with slight-acidity-induced charge conversion and hypoxia-motivated doxorubicin (DOX) release properties to achieve a more precise and synchronous therapeutic effect. Upon a change in the extracellular pH (pHe) in the tumor matrix, the surface charge of DANPVP&DOX converted from negative to positive via dimethyl maleate degradation. Following the increased internalization by tumoral cells, exposure of verteporfin (VP) in DANPVP&DOX to low-dose X-ray radiation resulted in O2 consumption in the cytoplasm to produce cytotoxic reactive oxygen species (ROS), which caused cell killing. Moreover, the hypoxic conditions formed in the tumor area specifically promoted DANPVP&DOX dissociation and on-demand DOX release. Consequently, DANPVP&DOX significantly increased the therapeutic efficacy through X-PDT and cascade chemotherapy. More importantly, this strategy could potentially be extended to various therapeutic agents other than anticancer drugs for precise drug delivery and cancer treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Doxorubicin/therapeutic use , Drug Carriers/chemistry , Nanoparticles/chemistry , Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Drug Carriers/chemical synthesis , Drug Carriers/pharmacokinetics , Drug Liberation , Female , Humans , Hydrogen-Ion Concentration , Mice, Inbred BALB C , Organophosphates/chemical synthesis , Organophosphates/chemistry , Organophosphates/pharmacokinetics , Photochemotherapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/radiation effects , Photosensitizing Agents/therapeutic use , Polyethylene Glycols/chemical synthesis , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics , Singlet Oxygen/metabolism , Verteporfin/chemistry , Verteporfin/pharmacokinetics , Verteporfin/radiation effects , Verteporfin/therapeutic use , X-Rays , Xenograft Model Antitumor Assays
4.
J Mater Chem B ; 7(31): 4843-4853, 2019 08 07.
Article in English | MEDLINE | ID: mdl-31389968

ABSTRACT

Hyaluronic acid (HA)-based nanocarriers are of great interest in the drug delivery field due to the tumor targetability via CD44-mediated recognition and endocytosis. However, sufficient tumor-specific release of encapsulated cargoes with steady controllability is necessary to optimize their outcome for cancer therapy. In this study, we constructed a light-activated nanocarrier TKHCENPDOX to enable on-demand drug release at the desired site (tumor). Particularly, TKHCENPDOX encapsulating doxorubicin (DOX) was self-assembled from a HA-photosensitizer conjugate (HA-TK-Ce6) containing reactive oxygen species (ROS)-sensitive thioketal (TK) linkers. Following i.v. injection, TKHCENPDOX was accumulated in the MDA-MB-231 breast tumor xenograft more efficiently through preventing drug leakage in the bloodstream and the HA-mediated targeting effect. Upon internalization into tumoral cells, 660 nm laser irradiation generated ROS during a photodynamic (PDT) process to cleave the TK linker next to Ce6, resulting in light-induced TKHCENPDOX dissociation and selective DOX release in the tumor area. Consequently, TKHCENPDOX showed a remarkable therapeutic effect and minimized toxicity in vivo. This strategy might provide new insight for designing cancer-selective nanoplatforms with active targeting and locoregional drug release simultaneously.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Doxorubicin/therapeutic use , Drug Carriers/chemistry , Hyaluronic Acid/chemistry , Nanoconjugates/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Chlorophyllides , Doxorubicin/pharmacokinetics , Drug Carriers/radiation effects , Drug Carriers/toxicity , Drug Liberation/radiation effects , Female , Humans , Hyaluronic Acid/radiation effects , Hyaluronic Acid/toxicity , Light , Mice, Inbred BALB C , Mice, Inbred ICR , Nanoconjugates/radiation effects , Nanoconjugates/toxicity , Nanoparticles/chemistry , Nanoparticles/radiation effects , Nanoparticles/toxicity , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Photosensitizing Agents/radiation effects , Photosensitizing Agents/toxicity , Porphyrins/pharmacology , Porphyrins/radiation effects , Porphyrins/toxicity , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
5.
Theranostics ; 8(11): 2939-2953, 2018.
Article in English | MEDLINE | ID: mdl-29896295

ABSTRACT

The simple integration of chemotherapeutic drugs and photosensitizers (PSs) into the same nanocarriers only achieves a combination of chemo-photodynamic therapy but may not confer synergistic effects. The boosted intracellular release of chemotherapeutic drugs during the photodynamic therapy (PDT) process is necessary to achieve a cascade of amplified synergistic therapeutic effects of chemo-photodynamic therapy. Methods: In this study, we explored an innovative hyperbranched polyphosphate (RHPPE) containing a singlet oxygen (SO)-labile crosslinker to boost drug release during the PDT process. The photosensitizer chlorin e6 (Ce6) and doxorubicin (DOX) were simultaneously loaded into RHPPE nanoparticles (denoted as SOHNPCe6/DOX). The therapeutic efficacy of SOHNPCe6/DOX against drug-resistant cancer was evaluated in vitro and in vivo. Results: Under 660-nm light irradiation, SOHNPCe6/DOX can produce SO, which not only induces PDT against cancer but also cleaves the thioketal linkers to destroy the nanoparticles. Subsequently, boosted DOX release can be achieved, activating a chemotherapy cascade to synergistically destroy the remaining tumor cells after the initial round of PDT. Furthermore, SOHNPCe6/DOX also efficiently detected the tumor area by photoacoustic/magnetic resonance bimodal imaging. Under the guidance of bimodal imaging, the laser beam was precisely focused on the tumor areas, and subsequently, SOHNPCe6/DOX realized a cascade of amplified synergistic chemo-photodynamic therapeutic effects. High antitumor efficacy was achieved even in a drug-resistant tumor model. Conclusion: The designed SOHNPCe6/DOX with great biocompatibility is promising for use as a co-delivery carrier for combined chemo-photodynamic therapy, providing an alternative avenue to achieve a cascade of amplified synergistic effects of chemo-photodynamic therapy for cancer treatment.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Neoplasms/drug therapy , Photochemotherapy , Photosensitizing Agents/pharmacology , Porphyrins/pharmacology , Radiation-Sensitizing Agents/pharmacology , Antibiotics, Antineoplastic/chemistry , Cell Line, Tumor , Chlorophyllides , Doxorubicin/chemistry , Drug Carriers , Drug Delivery Systems , Drug Liberation , Drug Synergism , Humans , Nanoparticles , Photosensitizing Agents/chemistry , Polymers/chemistry , Porphyrins/chemistry , Radiation-Sensitizing Agents/chemistry , Reactive Oxygen Species/chemistry , Theranostic Nanomedicine
6.
Biomater Sci ; 5(8): 1612-1621, 2017 Jul 25.
Article in English | MEDLINE | ID: mdl-28580971

ABSTRACT

Platinum-based chemotherapy as first-line treatment for lung cancers encounters insufficient selectivity, severe side effects and drug resistance in clinics. In this study, we developed an amphiphilic prodrug of cisplatin-poly(ethylene glycol)-block-polycaprolactone and demonstrated that the prodrug formed micellar nanoparticles, NPPt(IV), with an average diameter of ∼100 nm. NPPt(IV) released platinum in response to the intracellular acidic and reductive environment, and in turn induced significant anti-proliferative activity in lung cancer cells. More importantly, NPPt(IV) exhibited a prominent inhibitory effect on CD133+ lung cancer stem cells (CSCs) and suppressed tumor growth in vivo. Unlike cisplatin treatment which eventually enriches CSCs, NPPt(IV) treatment prevents the accumulation of CD133+ lung CSCs in tumors. Therefore, NPPt(IV) simutaneously targeting CSCs and non-CSCs might represent a superior strategy to improve conventional anticancer therapy directed predominantly to tumor bulk populations.


Subject(s)
Cisplatin/metabolism , Cisplatin/pharmacology , Lung Neoplasms/pathology , Micelles , Neoplastic Stem Cells/drug effects , Prodrugs/metabolism , AC133 Antigen/metabolism , Biological Transport , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Liberation , Drug Resistance, Neoplasm/drug effects , Humans , Hydrogen-Ion Concentration , Nanoparticles/chemistry , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Polyesters/chemistry , Polyethylene Glycols/chemistry , Prodrugs/chemistry , Time Factors
7.
Proc Natl Acad Sci U S A ; 113(15): 4164-9, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27035960

ABSTRACT

A principal goal of cancer nanomedicine is to deliver therapeutics effectively to cancer cells within solid tumors. However, there are a series of biological barriers that impede nanomedicine from reaching target cells. Here, we report a stimuli-responsive clustered nanoparticle to systematically overcome these multiple barriers by sequentially responding to the endogenous attributes of the tumor microenvironment. The smart polymeric clustered nanoparticle (iCluster) has an initial size of ∼100 nm, which is favorable for long blood circulation and high propensity of extravasation through tumor vascular fenestrations. Once iCluster accumulates at tumor sites, the intrinsic tumor extracellular acidity would trigger the discharge of platinum prodrug-conjugated poly(amidoamine) dendrimers (diameter ∼5 nm). Such a structural alteration greatly facilitates tumor penetration and cell internalization of the therapeutics. The internalized dendrimer prodrugs are further reduced intracellularly to release cisplatin to kill cancer cells. The superior in vivo antitumor activities of iCluster are validated in varying intractable tumor models including poorly permeable pancreatic cancer, drug-resistant cancer, and metastatic cancer, demonstrating its versatility and broad applicability.


Subject(s)
Antineoplastic Agents/therapeutic use , Nanoparticles , Neoplasms/drug therapy , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Apoptosis , Cell Line, Tumor , Humans , Neoplasm Metastasis , Neoplasms/pathology , Spheroids, Cellular
8.
Biomaterials ; 94: 9-19, 2016 07.
Article in English | MEDLINE | ID: mdl-27088406

ABSTRACT

Chemotherapy resistance has become a major challenge in the clinical treatment of lung cancer which is the leading cancer type for the estimated deaths. Recent studies have shown that nanoparticles as drug carriers can raise intracellular drug concentration by achieving effectively cellular uptake and rapid drug release, and therefore reverse the acquired chemoresistance of tumors. In this context, nanoparticles-based chemotherapy represents a promising strategy for treating malignancies with chemoresistance. In the present study, we developed cationic lipid assisted nanoparticles (CLAN) to deliver polylactide-cisplatin prodrugs to drug resistant lung cancer cells. The nanoparticles were formulated through self-assembly of a biodegradable poly(ethylene glycol)-block-poly(lactide) (PEG-PLA), a hydrophobic polylactide-cisplatin prodrug, and a cationic lipid. The cationic nanoparticles were proven to significantly improve cell uptake of cisplatin, leading to an increased DNA-Pt adduct and significantly promoted DNA damage in vitro. Moreover, our study reveals that cationic nanoparticles, although are slightly inferior in blood circulation and tumor accumulation, are more effective in blood vessel extravasation. The CLANs ultimately enhances the cellular drug availability and leads to the reversal of cisplatin resistance.


Subject(s)
Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Lipids/chemistry , Nanoparticles/chemistry , Prodrugs/pharmacology , A549 Cells , Animals , Cations , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/chemistry , Cisplatin/pharmacokinetics , Fatty Acids, Monounsaturated/chemistry , Female , Humans , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/ultrastructure , Polyesters/chemistry , Polyethylene Glycols/chemistry , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Quaternary Ammonium Compounds/chemistry , Tissue Distribution/drug effects
9.
Biomaterials ; 88: 48-59, 2016 May.
Article in English | MEDLINE | ID: mdl-26945455

ABSTRACT

The design of ideal nanoparticle delivery systems should be capable of meeting the requirements of several stages of drug delivery, including prolonged circulation, enhanced accumulation and penetration in the tumor, facilitated cellular internalization and rapid release of the active drug in the tumor cells. However, among the current design strategies, meeting the requirements of one stage often conflicts with the other. Herein, a tumor pH-labile linkage-bridged block copolymer of poly(ethylene glycol) with poly(lacide-co-glycolide) (PEG-Dlinkm-PLGA) was used for siRNA delivery to fulfill all aforementioned requirements of these delivery stages. The obtained siRNA-encapsulating PEG-Dlinkm-PLGA nanoparticle gained efficiently prolonged circulation in the blood and preferential accumulation in tumor sites via the PEGylation. Furthermore, the PEG surface layer was detached in response to the tumor acidic microenvironment to facilitate cellular uptake, and the siRNA was rapidly released within tumor cells due to the hydrophobic PLGA layer. Hence, PEG-Dlinkm-PLGA nanoparticles met the requirements of several stages of drug delivery, and resulted in the enhanced therapeutic effect of the nanoparticular delivery systems.


Subject(s)
Breast Neoplasms/therapy , Delayed-Action Preparations/chemistry , Lactic Acid/chemistry , Polyethylene Glycols/chemistry , Polyglycolic Acid/chemistry , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/therapeutic use , RNAi Therapeutics , Animals , Breast/metabolism , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle Proteins/genetics , Cell Line, Tumor , Female , Humans , Hydrogen-Ion Concentration , Mice , Nanoparticles/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacokinetics , Polo-Like Kinase 1
10.
Angew Chem Int Ed Engl ; 55(3): 1010-4, 2016 Jan 18.
Article in English | MEDLINE | ID: mdl-26756443

ABSTRACT

Successful bench-to-bedside translation of nanomedicine relies heavily on the development of nanocarriers with superior therapeutic efficacy and high biocompatibility. However, the optimal strategy for improving one aspect often conflicts with the other. Herein, we report a tactic of designing tumor-pH-labile linkage-bridged copolymers of clinically validated poly(D,L-lactide) and poly(ethylene glycol) (PEG-Dlink(m)-PDLLA) for safe and effective drug delivery. Upon arriving at the tumor site, PEG-Dlink(m)-PDLLA nanoparticles will lose the PEG layer and increase zeta potential by responding to tumor acidity, which significantly enhances cellular uptake and improves the in vivo tumor inhibition rate to 78.1% in comparison to 47.8% of the non-responsive control. Furthermore, PEG-Dlink(m)-PDLLA nanoparticles show comparable biocompatibility with the clinically used PEG-b-PDLLA micelle. The improved therapeutic efficacy and safety demonstrate great promise for our strategy in future translational studies.


Subject(s)
Antineoplastic Agents/therapeutic use , Hydrogen-Ion Concentration , Neoplasms/drug therapy , Polymers/chemistry , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Humans , Nanoparticles , Neoplasms/chemistry
11.
ACS Biomater Sci Eng ; 2(8): 1319-1329, 2016 Aug 08.
Article in English | MEDLINE | ID: mdl-33434985

ABSTRACT

Spinal cord injuries (SCIs) are followed by a complex series of events that contribute to the failure of regeneration. To date, there is no robust treatment that can restore the injury-induced loss of function. Since damaged spinal axons do not spontaneously regenerate in their native inhibitory microenvironment, a combined application of biomaterials and neurotrophic factors that induce nerve regeneration emerges as an attractive treatment for SCIs. In this study, we report the novel use of a three-dimensional (3D) hybrid scaffold to provide contact guidance for regrowth of axons in vivo. The scaffold comprises 3D aligned sparsely distributed poly(ε-caprolactone-co-ethyl ethylene phosphate) nanofibers that are supported and dispersed within a collagen hydrogel. Neurotrophin-3 was incorporated into the scaffold as an additional biochemical signal. To evaluate the efficacy of the scaffold in supporting nerve regeneration after SCIs, the construct was implanted into an incision injury, which was created at level C5 in the rat spinal cord. After 3 months of implantation, scaffolds with NT-3 incorporation showed the highest average neurite length (391.9 ± 12.9 µm, p ≤ 0.001) as compared to all the other experimental groups. In addition, these regenerated axons formed along the direction of the aligned nanofibers, regardless of their orientation. Moreover, the presence of the hybrid scaffolds did not affect tissue scarring and inflammatory reaction. Taken together, these findings demonstrate that our scaffold design can serve as a potential platform to support axonal regeneration following SCIs.

12.
J Am Chem Soc ; 137(48): 15217-24, 2015 Dec 09.
Article in English | MEDLINE | ID: mdl-26571079

ABSTRACT

Although surface PEGylation of siRNA vectors is effective for preventing protein adsorption and thereby helps these vectors to evade the reticuloendothelial system (RES) in vivo, it also suppresses the cellular uptake of these vectors by target cells. This dilemma could be overcome by employing stimuli-responsive shell-detachable nanovectors to achieve enhanced cellular internalization while maintaining prolonged blood circulation. Among the possible stimuli, dysregulated pH in tumor (pHe) is the most universal and practical. However, the design of pHe-sensitive system is problematic because of the subtle differences between the pHe and pH in other tissues. Here, a simple acid-sensitive bridged copolymer is developed and used for tumor-targeted systemic delivery of siRNA. After forming the micelleplex delivery system, the corresponding nanoparticles (Dm-NP) might undergo several modifications as follows: (i) a poly(ethylene glycol) (PEG) corona, which is stable in the circulatory system and protects nanovectors from RES clearance; (ii) a pHe responsive linkage breakage, which induces PEG detachment at tumor sites and thereby facilitates cell targeting; and (iii) a cell-penetration peptide, which is exposed upon the removal of PEG and further enhances cellular uptake. Thus, Dm-NP achieved both prolonged circulation and effective accumulation in tumor cells and resulted in the safe and enhanced inhibition of non-small cell lung cancer growth.


Subject(s)
Neoplasms/metabolism , Polymers/chemistry , RNA, Small Interfering/administration & dosage , Micelles , RNA, Small Interfering/pharmacokinetics , Tissue Distribution
13.
ACS Appl Mater Interfaces ; 7(47): 26315-25, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26552849

ABSTRACT

Multidrug resistance (MDR) has been recognized as a key factor contributing to the failure of chemotherapy for cancer in the clinic, often due to insufficient delivery of anticancer drugs to target cells. For addressing this issue, a redox-responsive polyphosphoester-based micellar nanomedicine, which can be triggered to release transported drugs in tumor cells, has been developed. The micelles are composed of diblock copolymers with a hydrophilic PEG block and a hydrophobic polyphosphoester (PPE) block bearing a disulfide bond in a side group. After incubating the redox-responsive micelles with drug-resistant tumor cells, the intracellular accumulation and retention of DOX were significantly enhanced. Moreover, after internalization by MDR cancer cells, the disulfide bond in the side group was cleaved by the high intracellular glutathione levels, resulting in a hydrophobic to hydrophilic transition of the PPE block and subsequent disassembly of the micelles. Thus, the encapsulated DOX was rapidly released, and abrogation of drug resistance in the cancer cells was observed in vitro. Moreover, the DOX-loaded redox-responsive micelles exhibited significantly enhanced inhibition of tumor growth in nude mice bearing MCF-7/ADR xenograft tumors via tail vein injection, indicating that such micelles have great potential in overcoming MDR for cancer therapy.


Subject(s)
Breast Neoplasms/pathology , Drug Resistance, Neoplasm , Esters/chemistry , Micelles , Nanomedicine/methods , Polyphosphates/chemistry , Animals , Antineoplastic Agents/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Doxorubicin/pharmacology , Drug Resistance, Multiple/drug effects , Dynamic Light Scattering , Female , Flow Cytometry , Humans , Mice, Inbred BALB C , Mice, Nude , Oxidation-Reduction , Proton Magnetic Resonance Spectroscopy , Pyrenes/chemistry , Xenograft Model Antitumor Assays
14.
Biomaterials ; 69: 1-11, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26275857

ABSTRACT

Poly(ethylene glycol) (PEG) is usually used to protect nanoparticles from rapid clearance in blood. The effects are highly dependent on the surface PEG density of nanoparticles. However, there lacks a detailed and informative study in PEG density and in vivo drug delivery due to the critical techniques to precisely control the surface PEG density when maintaining other nano-properties. Here, we regulated the polymeric nanoparticles' size and surface PEG density by incorporating poly(ε-caprolactone) (PCL) homopolymer into poly(ethylene glycol)-block-poly(ε-caprolactone) (PEG-PCL) and adjusting the mass ratio of PCL to PEG-PCL during the nanoparticles preparation. We further developed a library of polymeric nanoparticles with different but controllable sizes and surface PEG densities by changing the molecular weight of the PCL block in PEG-PCL and tuning the molar ratio of repeating units of PCL (CL) to that of PEG (EG). We thus obtained a group of nanoparticles with variable surface PEG densities but with other nano-properties identical, and investigated the effects of surface PEG densities on the biological behaviors of nanoparticles in mice. We found that, high surface PEG density made the nanoparticles resistant to absorption of serum protein and uptake by macrophages, leading to a greater accumulation of nanoparticles in tumor tissue, which recuperated the defects of decreased internalization by tumor cells, resulting in superior antitumor efficacy when carrying docetaxel.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Drug Carriers/chemistry , Lactones/chemistry , Nanoparticles/chemistry , Polyesters/chemistry , Polyethylene Glycols/chemistry , Taxoids/administration & dosage , Animals , Antineoplastic Agents/therapeutic use , Breast/drug effects , Breast/pathology , Breast Neoplasms/pathology , Cell Line, Tumor , Docetaxel , Drug Carriers/metabolism , Female , Humans , Lactones/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/metabolism , Polyesters/metabolism , Polyethylene Glycols/metabolism , RAW 264.7 Cells , Surface Properties , Taxoids/pharmacokinetics , Taxoids/therapeutic use
15.
Biomaterials ; 70: 71-83, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26302232

ABSTRACT

As part of HCC tumor cellularity, cancer stem cells (CSCs) are considered a major obstacle to eradicate hepatocellular carcinoma (HCC), which is the third most common cause of cancer-related death worldwide, and the accumulation of chemotherapeutic drug-resistant CSCs invariably accounts for poor prognosis and HCC relapse. In the present study, we explored the efficacy of co-delivery of platinum drug and siRNA targeting Notch1 to treat CSCs-harboring HCC. To overcome the challenging obstacles of platinum drug and siRNA in the systemic administration, we developed a micellar nanoparticle (MNP) to deliver platinum(IV) prodrug and siNotch1, hereafter referred to as (Pt(IV))MNP/siNotch1. We demonstrated that (Pt(IV))MNP/siNotch1 was able to efficiently deliver two drugs into both non-CSCs and CSCs of SMMC7721, a HCC cell line. We further found that siRNA-mediated inhibition of Notch1 suppression can increase the sensitivity of HCC cells to platinum drugs and decrease the percentage of HCC CSCs, and consequently resulting in enhanced proliferation inhibition and apoptosis induction in HCC cells in vitro. Moreover, our results indicated that the combined drug delivery system can remarkably augment drug enrichment in tumor tissues, substantially suppressing the tumor growth while avoiding the accumulation of CSCs in a synergistic manner in the SMMC7721 xenograft model.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Drug Delivery Systems , Liver Neoplasms/drug therapy , Micelles , Platinum/therapeutic use , RNA, Small Interfering/metabolism , Receptor, Notch1/metabolism , AC133 Antigen , Animals , Antigens, CD/metabolism , Carcinoma, Hepatocellular/pathology , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Combined Modality Therapy , Glycoproteins/metabolism , Humans , Liver Neoplasms/pathology , Mice, Inbred NOD , Mice, SCID , Nanoparticles/chemistry , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Peptides/metabolism , Xenograft Model Antitumor Assays
16.
Biomater Sci ; 3(7): 1105-13, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26221944

ABSTRACT

Polymeric nanoparticles have been widely used as nano-drug delivery systems in preclinical and clinical trials for cancer therapy, and these systems usually need to be sterically stabilized by poly(ethylene glycol) (PEG) to maintain stability and avoid rapid clearance by the immune system. Recently, zwitterionic materials have been demonstrated to be potential alternatives to the classic PEG. Herein, we developed two drug delivery systems stabilized by zwitterionic polyphosphoesters. These nanoparticles showed favourable stability and anti-protein absorption ability in vitro. Meanwhile, as drug carriers, these zwitterionic polyphosphoester-stabilized nanoparticles significantly prolonged drug circulation half-lives and increased drug accumulation in tumors, which was comparable to PEG-stabilized nanoparticles. Systemic delivery of doxorubicin (DOX) by zwitterionic polyphosphoester-stabilized nanoparticles significantly inhibited tumor growth in a MDA-MB-231 tumor model, suggesting the potential of zwitterionic polyphosphoester-based nanoparticles in anticancer drug delivery.


Subject(s)
Amino Acid Transport Systems, Neutral/chemistry , Doxorubicin/therapeutic use , Drug Delivery Systems/methods , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Polymers/chemistry , Amino Acid Transport Systems, Neutral/pharmacokinetics , Cell Line, Tumor , Doxorubicin/chemistry , Drug Carriers , Humans , Hydrogen-Ion Concentration
17.
ACS Appl Mater Interfaces ; 7(1): 1012-20, 2015 Jan 14.
Article in English | MEDLINE | ID: mdl-25522342

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common malignant human tumors worldwide, but no effective therapeutic options are currently available. The cancer stem cell (CSC) has proven to play a central role in the development, metastasis, and recurrence of HCC. In this study, we report a dual functional mitogen-activated protein kinase inhibitor (U0126)-based therapy for treating both bulk HCC and HCC CSCs, using poly(ethylene glycol)-b-poly(d,l-lactide) (PEG-PLA) nanoparticles as the drug carrier. It is demonstrated that nanoparticle encapsulation enhanced the cell uptake of U0126 in HCC CSCs and that enhanced endocytosis lead to augmented cytotoxicity of U0126 in HCC CSCs. Moreover, the nanoparticle encapsulation increased the inhibition of self-renewal capability, prolonged the circulation time, and increased the tumor accumulation of U0126 when compared with the use of the free inhibitor. The systemic delivery of U0126 remarkably enhanced the suppression of tumor development with decreased CSCs in the HepG2 xenograft simultaneously with reduced systemic toxicity.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Stem Cell Transplantation/methods , Animals , Butadienes/chemistry , Cell Line, Tumor , Drug Carriers , Endocytosis , Enzyme Inhibitors/chemistry , Female , Hep G2 Cells , Humans , Immunohistochemistry , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis , Neoplasm Recurrence, Local , Neoplasm Transplantation , Nitriles/chemistry , Polyethylene Glycols/chemistry
18.
ACS Appl Mater Interfaces ; 6(24): 22709-18, 2014 Dec 24.
Article in English | MEDLINE | ID: mdl-25426800

ABSTRACT

Recently, micelles, which are self-assembled by amphiphilic copolymers, have attracted tremendous attention as promising drug delivery systems for cancer treatment. Thus, the hydrophobic core of the micelles, which could efficiently encapsulate small molecular drug, will play a significant role for the anticancer efficiency. Unfortunately, the effect of hydrophobicity of micellar core on its anticancer efficiency was rarely reported. Herein, the amphiphilic diblock polymers of poly(ethylene glycol) and polyphosphoester with different side groups (butyl, hexyl, octyl) were synthesized to tune the hydrophobicity of the micellar core. We found that the in vitro cytotoxicity of the DOX-loaded micelles decreased with the increasing hydrophobicity of micellar core due to the drug release rate. However, following systemic delivery, the DOX-loaded micelles with the most hydrophobic core exhibited the most significant inhibition of tumor growth in a MDA-MB-231 tumor model, indicating the importance of hydrophobicity of core on the antitumor efficacy of drug delivery systems.


Subject(s)
Doxorubicin/administration & dosage , Nanocapsules/chemistry , Nanocapsules/ultrastructure , Neoplasms, Experimental/drug therapy , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/chemistry , Cell Line, Tumor , Diffusion , Doxorubicin/chemistry , Drug Synergism , Hydrophobic and Hydrophilic Interactions , Mice , Mice, Inbred BALB C , Mice, Nude , Micelles , Nanocapsules/administration & dosage , Neoplasms, Experimental/pathology , Particle Size , Surface Properties , Treatment Outcome
19.
Adv Healthc Mater ; 3(11): 1792-803, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24947820

ABSTRACT

Patients with Her2-overexpressing (Her2(+)) breast cancers generally have a poorer prognosis due to the high aggressiveness and chemoresistance of the disease. Small interfering RNA (siRNA) targeting the gene encoding polo-like kinase 1 (Plk1; siPlk1) has emerged as an efficient therapeutic agent for Her2(+) breast cancers. Poly(ethylene glycol)-block-poly(D,L-lactide) (PEG-PLA)-based nanoparticles for siRNA delivery were previously developed and optimized. In this study, for targeted delivery of siPlk1 to Her2(+) breast cancer, anti-Her2 single-chain variable fragment antibody (ScFv(Her2))-decorated PEG-PLA-based nanoparticles with si Plk1 encapsulation (ScFv(Her2)-NP(si) Plk1) are developed. With the rationally designed conjugation site, ScFv(Her2)-NP(siRNA) can specifically bind to the Her2 antigen overexpressed on the surface of Her2(+) breast cancer cells. Therefore, ScFv(Her2)-NP(si) Plk1 exhibits improved cellular uptake, promoted Plk1 silencing efficiency, and induced enhanced tumor cell apoptosis in Her2(+) breast cancer cells, when compared with nontargeted NP(si) Plk1. More importantly, ScFv(Her2)-NP(siRNA) markedly enhances the accumulation of siRNA in Her2(+) breast tumor tissue, and remarkably improves the efficacy of tumor suppression. Dose-dependent anti-tumor efficacy further demonstrates that ScFvHer2 -decorated PEG-PLA-based nanoparticles with siPlk1 encapsulation can significantly enhance the inhibition of Her2(+) breast tumor growth and reduce the dose of injected siRNA. These results suggest that ScFvHer2 -decorated PEG-PLA-based nanoparticles show great potential for targeted RNA interference therapy of Her2(+) breast tumor.


Subject(s)
Breast Neoplasms/therapy , Nanoparticles/administration & dosage , Polyethylene Glycols/administration & dosage , RNA, Small Interfering/administration & dosage , Single-Chain Antibodies/administration & dosage , Apoptosis/drug effects , Breast Neoplasms/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Female , Genetic Therapy/methods , Humans , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, ErbB-2/metabolism , Polo-Like Kinase 1
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...