Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38032100

ABSTRACT

S-scheme heterojunction photocatalyst MAPbI3@PCN-222 with light absorption extending to the NIR region is constructed by embedding organic-inorganic hybrid perovskite (MAPbI3) into porphyrinic Zr-MOF (PCN-222). Both in situ X-ray photoelectron spectroscopy, ultraviolet photoelectron spectral characterization, and photocatalytic polymerization experiment prove the formation of S-scheme heterojunction. MAPbI3@PCN-222 with a low dosage (90 ppm) displays an impressive photocatalytic ability for 980 nm light-mediated photoinduced electron/energy-transfer-reversible addition-fragmentation chain-transfer (PET-RAFT) polymerization in air. The well-defined controllable-molecular weight polymers including block copolymers and ultrahigh-molecular weight polymers can be achieved with narrow distributions (Mw/Mn < 1.20) via rapid photopolymerization. The industrial application potential of the photocatalyst also has been proved by scale-up synthesis of polymers with low polydispersity under NIR light-induced photopolymerization in a large-volume reaction system (200 mL) with high monomer conversion up to 99%. The penetration photopolymerization through the 5 mm polytetrafluoroethylene plate and excellent photocontrollable behavior illustrate the existence of long-term photogenerated electron transfer of heterojunction and abundant free radicals in photopolymerization. The photocatalyst still retains high catalytic activity after 10 cycles of photopolymerization in air. It is revealed for the first time that the special PET-RAFT polymerization pathway is initiated by the aldehyde-bearing α-aminoalkyl radical derived from the oxidization of triethanolamine (TEOA) by the heterojunction photocatalyst. This research offers a new insight into understanding the NIR-light-activated PET-RAFT polymerization mechanism in the presence of TEOA.

2.
ACS Appl Mater Interfaces ; 15(21): 25615-25623, 2023 May 31.
Article in English | MEDLINE | ID: mdl-37194188

ABSTRACT

Synthesis of alloy-type materials (X) is one of the most effective approaches to limit lithium dendrites in Li metal anode (LMA) because of their satisfactory lithiophilicity and easy electrochemical reaction with lithium. However, current investigations have only focused on the influence of the resulting alloyed products (LiX) on the properties of LMA, but the alloying reaction process between Li+ and X has been mostly ignored. Herein, by masterly taking advantage of the alloying reaction process, a novel approach is developed to more effectively inhibit lithium dendrites than the conventional strategy that just considers the utilization of alloyed products LiX. A three-dimensional substrate material loaded with metallic Zn on the surface of Cu foam is synthesized by a simple electrodeposition process. During Li plating/stripping, both alloy reaction processes between Li+ and Zn and LiZn product are involved, which makes the disordered Li+ flux near the substrate first react with Zn metal and then results in an even Li+ concentration for more uniform Li nucleation and growth. The full cell (Li-Cu@Zn-15//LFP) exhibits the reversible capacity of 122.5 mAh g-1, and a high capacity retention of 95% is achieved after 180 cycles. This work proposes a valuable concept for the development of alloy-type materials in energy storage devices.

3.
Asian J Pharm Sci ; 17(4): 571-582, 2022 Jul.
Article in English | MEDLINE | ID: mdl-36105315

ABSTRACT

Poly(2-oxazoline) (POx) has been regarded as a potential candidate for drug delivery carrier to meet the challenges of nanomedicine clinical translation, due to its excellent biocompatibility and self-assembly properties. The drug loading capacity and stability of amphiphilic POxs as drug nanocarriers, however, tend to be insufficient. Herein, we report a strategy to prepare nucleobase-crosslinked POx nanoparticles (NPs) with enhanced stability and ultra-high paclitaxel (PTX) loading capacity for breast cancer therapy. An amphiphilic amine-functionalized POx (PMBEOx-NH2) was firstly prepared through a click reaction between cysteamines and vinyl groups in poly(2-methyl-2-oxazoline)-block-poly (2­butyl­2-oxazoline-co-2-butenyl-2-oxazoline) (PMBEOx). Complementary nucleobase-pairs adenine (A) and uracil (U) were subsequently conjugated to PMBEOx-NH2 to give functional POxs (POxA and POxU), respectively. Due to the nucleobase interactions formed between A and U, NPs formed by POxA and POxU at a molar ratio of 1:1 displayed ultrahigh PTX loading capacity (38.2%, PTX/POxA@U), excellent stability, and reduced particle size compared to the uncross-linked PTX-loaded NPs (PTX/PMBEOx). Besides the prolonged blood circulation and enhanced tumor accumulation, the smaller PTX/POxA@U NPs also have better tumor penetration ability compared with PTX/PMBEOx, thus leading to a higher tumor suppression rate in two murine breast cancer models (E0711 and 4T1). These results proved that the therapeutic effect of chemotherapeutic drugs could be improved remarkably through a reasonable optimization of nanocarriers.

4.
Adv Healthc Mater ; 11(16): e2200268, 2022 08.
Article in English | MEDLINE | ID: mdl-35758640

ABSTRACT

Persistent fungal infections caused by biofilms seriously endanger human health. In this study, a photosensitizer-polypeptide conjugate (PPa-cP) comprising a photosensitizer, pyropheophorbide a (PPa), and a cationic polypeptide (cP) is readily synthesized for effective antifungal and antibiofilm treatment. Compared with free PPa, the cationic PPa-cP shows enhanced binding ability to the negatively charged surface of Candida albicans (C. albicans) through electrostatic interactions. As a result, PPa-cP exhibits effective antifungal efficiency against both C. albicans and fluconazole-resistant C. albicans in vitro under light irradiation. The minimum inhibitory concentration (MIC) of PPa-cP for both C. albicans and fluconazole-resistant C. albicans is 1 µm. In addition, PPa-cP also shows improved penetration in a C. albicans biofilm, thus effectively eliminating the C. albicans biofilm by photodynamic effects. More importantly, PPa-cP demonstrats significantly enhanced therapeutic effects in a fluconazole-resistant C. albicans-infected rat model with minimal side effects. In conclusion, the current work presents an effective strategy to combat biofilm infections associated with biomedical equipment.


Subject(s)
Candida albicans , Fluconazole , Animals , Antifungal Agents/pharmacology , Biofilms , Fluconazole/pharmacology , Humans , Microbial Sensitivity Tests , Peptides/pharmacology , Photosensitizing Agents/pharmacology , Rats
5.
Macromol Biosci ; 22(7): e2200105, 2022 07.
Article in English | MEDLINE | ID: mdl-35526119

ABSTRACT

Recently, continuous emergence of resistant bacteria has appeared as one of the most serious threats to human health. Therefore, systematic exploration of new antibacterial materials is of guiding significance. In this study, a series of photosensitizer-polypeptide conjugate (PPa-cP) is readily synthesized through simple ring-opening reactions to realize the synergistic antibacterial effects on Staphylococcus aureus (S. aureus) and methicillin-resistant S. aureus (MRSA) under light irradiation. Compared with free PPa, the cationic PPa-cP shows enhanced binding ability with the negative surface of S. aureus through electrostatic interaction, exhibiting effective antibacterial activity against both S. aureus and MRSA in vitro under light irradiation. Among the synthesized PPa-cP, PPa-cP5 with the degree of polymerization of 37 and modified with a 1-methylimidazole side group exhibits the best antibacterial activity with a minimum inhibitory concentration value of 2 µm without light irradiation and 0.25 µm with light irradiation. Moreover, PPa-cP5 shows good hemocompatibility. The above-mentioned results elucidate that the positively charged PPa-cP5 can significantly increase the efficiency of photodynamic therapy and effectively eradicate S. aureus biofilm due to its potent penetration ability into S. aureus biofilms. Overall, the present study establishes an efficient strategy for the treatment of S. aureus and S. aureus biofilm infections.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Anti-Bacterial Agents/pharmacology , Biofilms , Humans , Microbial Sensitivity Tests , Peptides/pharmacology , Photosensitizing Agents/pharmacology , Staphylococcal Infections/drug therapy , Staphylococcus aureus
6.
ACS Biomater Sci Eng ; 8(2): 903-911, 2022 02 14.
Article in English | MEDLINE | ID: mdl-35050580

ABSTRACT

The steady development of bacterial resistance has become a global public health issue, and new antibacterial agents that are active against drug-resistant bacteria and less susceptible to bacterial resistance are urgently needed. Here, a series of low-molecular-weight cationic polylysines (Cx-PLLn) with different hydrophobic end groups (Cx) and degrees of polymerization (PLLn) was synthesized and used in antibacterial applications. All the obtained Cx-PLLn have antibacterial activity. Among them, C6-PLL13 displays the best antibacterial effect for Gram-positive bacteria, that is, Staphylococcus aureus (S. aureus) and methicillin-resistant Staphylococcus aureus (MRSA), and highest selectivity against Gram-positive bacteria. A mechanistic study revealed that the C6-PLL13 destroys the integrity of the bacterial cell membrane and causes effective bacterial death. Owing to this membrane-disrupting property, C6-PLL13 showed rapid bacterial killing kinetics and was not likely to develop resistance after repeat treatment (up to 13 generations). Moreover, C6-PLL13 demonstrated a significant therapeutic effect on an MRSA infection mouse model, which further proved that this synthetic polymer could be used as an effective weapon against bacterial infections.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Hemolysis , Mice , Microbial Sensitivity Tests , Polylysine/pharmacology , Staphylococcus aureus
7.
Int J Pharm ; 550(1-2): 79-88, 2018 Oct 25.
Article in English | MEDLINE | ID: mdl-30138704

ABSTRACT

Gemcitabine is widely used for anticancer therapy. However, its short blood circulation time and poor stability greatly impair its application. To solve this problem, we prepared a poly (l-glutamic acid)-g-methoxy poly (ethylene glycol)-gemcitabine conjugate (l-Gem) with a 14.3 wt% drug-loading content. l-Gem showed concentration- and time-dependent cytotoxicity towards 4T1, LLC, MIA PaCa-2 and A2780 in vitro. Pharmacokinetic and biodistribution studies indicated that l-Gem had remarkably enhanced blood stability, prolonged blood circulation time and greatly improved selective tumor distribution compared with free gemcitabine. The area under the concentration-time curve from zero to infinity [AUC(0-∞)] of l-Gem in plasma was 43-fold higher than that of free gemcitabine. The AUC(0-∞) of the inactive metabolite, 2'-deoxy-2',2'-difluorouridine in the l-Gem group was ∼20% of that observed in the free gemcitabine group. The drug tumor accumulation ratio in the l-Gem group relative to the free gemcitabine group was 9.9 at 36 h, while the tumor AUC ratio was 15.8. Testing on Balb/C mice bearing the 4T1 tumor further demonstrated that l-Gem had significantly higher anticancer efficacy than free gemcitabine in vivo. These findings indicated that l-Gem has great potential for cancer treatment.


Subject(s)
Antineoplastic Agents/chemistry , Deoxycytidine/analogs & derivatives , Polyethylene Glycols , Polyglutamic Acid , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Cell Survival/drug effects , Deoxycytidine/chemistry , Deoxycytidine/pharmacokinetics , Female , Humans , Male , Mice, Inbred BALB C , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Tissue Distribution , Gemcitabine
8.
Acta Biomater ; 73: 388-399, 2018 06.
Article in English | MEDLINE | ID: mdl-29694920

ABSTRACT

Podophyllotoxin (PPT) is a chemotherapeutic agent which has shown significant activity against P-glycoprotein (P-gp) mediated multi drug resistant cancer cells. However, because of the poor aqueous solubility and high toxicity, PPT cannot be used in clinical cancer therapy. In order to enhance the efficiency and reduce side effect of PPT, a polypeptide based PPT conjugate PLG-g-mPEG-PPT was developed and used for the treatment of multi drug resistant breast cancer. The PLG-g-mPEG-PPT was prepared by conjugating PPT to poly(l-glutamic acid)-g-methoxy poly(ethylene glycol) (PLG-g-mPEG) via ester bonds. The PPT conjugates self-assembled into nanoparticles with average sizes about 100 nm in aqueous solution. Western blotting assay showed that the PLG-g-mPEG-PPT could effectively inhibit the expression of P-gp in the multiple drug resistant MCF-7/ADR cells. In vitro cytotoxicity assay indicated that the resistance index (RI) values of PLG-g-mPEG-PPT on different drug-resistant cancer cell lines exhibited 57-270 folds reduction than of traditional microtubule inhibitor chemotherapeutic drug PTX or DTX. Hemolysis assay demonstrated that the conjugation greatly decreased the hemolytic activity of free PPT. Maximum tolerated dose (MTD) of PLG-g-mPEG-PPT increased greatly (13.3 folds) as compared to that of free PPT. In vivo study showed that the PLG-g-mPEG-PPT conjugate remarkably enhanced the antitumor efficacy against MCF-7/ADR xenograft tumors with a tumor suppression rate (TSR) of 82.5%, displayed significantly improved anticancer efficacy as compared to free PPT (TSR = 37.1%) with minimal toxicity when both of the two formulations were used in MTD. STATEMENT OF SIGNIFICANCE: The development of multiple drug resistance (MDR) of cancer cells is the main cause of chemotherapy failure. The over-expression of P-glycoprotein (P-gp) has been recognized to be the most important cause of MDR in cancer. Podophyllotoxin (PPT) is a chemotherapeutic agent which has shown strong activity against P-gp mediated multidrug resistant cancer cells by simultaneously inhibiting the over-expression of P-gp and the growth of cancer cells. However, PPT can not be used in clinical cancer treatment due to its poor aqueous solubility and high toxicity. Herein, we developed a polypeptide based PPT conjugate PLG-g-mPEG-PPT by conjugating PPT to poly(l-glutamic acid)-g-methoxy poly(ethylene glycol). The PLG-g-mPEG-PPT shows significantly decreased hemolytic activity, greatly improved maximum tolerated dose and remarkably enhanced antitumor efficacy against MCF-7/ADR xenograft tumors as compared to free PPT.


Subject(s)
Breast Neoplasms/drug therapy , Drug Delivery Systems , Drug Resistance, Neoplasm , Peptides/chemistry , Podophyllotoxin/chemistry , A549 Cells , ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , Animals , Antineoplastic Agents/chemistry , Apoptosis , Cell Line, Tumor , Drug Evaluation, Preclinical , Drug Resistance, Multiple/drug effects , Female , Hemolysis , Humans , MCF-7 Cells , Maximum Tolerated Dose , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Neoplasm Transplantation , Polyethylene Glycols/chemistry , Polyglutamic Acid/chemistry
9.
Biomacromolecules ; 18(12): 4341-4348, 2017 Dec 11.
Article in English | MEDLINE | ID: mdl-29141405

ABSTRACT

In this study, a type of novel thermosensitive polypeptide-based hydrogel with tunable gelation behavior through changing the content of carboxyl groups was developed for the purpose of improving the cisplatin (CDDP) release behavior and enhancing the localized antitumor efficiency. The introduction of carboxyl groups in methoxy-poly(ethylene glycol)-b-(poly(γ-ethyl-l-glutamate-co-l-glutamic acid) (mPEG-b-P(ELG-co-LG)) not only led to adjustable mechanical properties of the hydrogel but also significantly reduced the burst release of the drug through the complexation between the carboxyl groups of polypeptide and CDDP. Furthermore, both the good biocompatibility and the biodegradable properties of mPEG-b-P(ELG-co-LG) hydrogel were observed in vivo. Interestingly, the CDDP-complexed mPEG-b-P(ELG-co-LG) hydrogel exhibited significantly enhanced antitumor efficacy in vivo compared to the mPEG-b-PELG hydrogel loaded with CDDP without complexation, although a lower cytotoxicity and IC50 of the CDDP-complexed hydrogel was observed in vitro. Overall, the new type of injectable CDDP-complexed hydrogel may serve as an efficient platform for sustained CDDP delivery in localized tumor therapy.


Subject(s)
Antineoplastic Agents/chemistry , Cisplatin/chemistry , Glutamic Acid/chemistry , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Peptides/chemistry , Polyethylene Glycols/chemistry , Animals , Antineoplastic Agents/pharmacology , Biocompatible Materials/chemistry , Biodegradable Plastics/chemistry , Cell Line, Tumor , Cisplatin/pharmacology , Drug Carriers/chemistry , Female , HeLa Cells , Humans , Injections/methods , MCF-7 Cells , Mice , Mice, Inbred BALB C , Rats , Rats, Sprague-Dawley
10.
Biomater Sci ; 5(10): 2169-2178, 2017 Sep 26.
Article in English | MEDLINE | ID: mdl-28914292

ABSTRACT

A pH and redox dual-sensitive biodegradable polysaccharide, succinic acid-decorated dextran-g-phenylalanine ethyl ester-g-cysteine ethyl ester (Dex-SA-l-Phe-l-Cys), was synthesized to load doxorubicin hydrochloride (DOX·HCl). The DOX-loaded nanoparticles, which were prepared in aqueous solution and free of organic solvent, could spontaneously self-assemble into uniform sizes. When loading DOX·HCl, mercapto Dex-SA-l-Phe-l-Cys was oxidized into a crosslinked disulfide linkage to form pH and redox dual-sensitive nanoparticles (DOX-S-NPs). The amphiphilic polymer loaded DOX·HCl into the core through electrostatic and hydrophobic interactions, meanwhile the crosslinked disulfide bond could stabilize the drug loaded nanoparticles. As a control with similar polymer structure, succinic acid decorated dextran-g-phenylalanine ethyl ester (Dex-SA-l-Phe) was prepared to obtain pH-sensitive DOX-loaded micelles (DOX-N-NPs). The controlled pH and redox-dependent release profiles of the DOX-S-NPs in vitro were certified in different releasing mediums. Furthermore, the cellular uptake of the DOX-S-NPs was comparable with that of free DOX·HCl, determined by confocal laser scanning microscopy (CLSM) and flow cytometry. Cytotoxicity assay in vitro showed that the DOX-S-NPs and free DOX·HCl were similar in inhibiting the proliferation of non-small cell lung carcinoma A549 and breast cancer MCF-7 cell lines. DOX-S-NPs displayed similar antitumor efficiency compared with free DOX·HCl, but lower toxicity by body weight. These dual-sensitive DOX-S-NPs provide a useful strategy for anti-tumor therapy.


Subject(s)
Antineoplastic Agents/chemistry , Doxorubicin/chemistry , Drug Carriers/chemistry , Nanoparticles/chemistry , Polysaccharides/chemistry , Animals , Antineoplastic Agents/pharmacology , Biological Transport , Doxorubicin/pharmacology , Drug Carriers/metabolism , Drug Carriers/pharmacology , Drug Liberation , Female , Humans , Hydrogen-Ion Concentration , MCF-7 Cells , Materials Testing , Mice , Micelles , Oxidation-Reduction , Polysaccharides/metabolism , Polysaccharides/pharmacology
11.
J Biomater Sci Polym Ed ; 28(4): 394-414, 2017 03.
Article in English | MEDLINE | ID: mdl-28027693

ABSTRACT

Patupilone, an original natural anti-cancer agent, also known as epothilone B or Epo906, has shown promise for the treatment of a variety of cancers, however, the systematic side effects of patupilone significantly impaired its clinical translation. Herein, patupilone-loaded PLG-g-mPEG micelles were prepared. Patupilone was grafted to a poly(L-glutamic acid)-graft-methoxy-poly(ethylene glycol) (PLG-g-mPEG) by Steglich esterification reaction to give PLG-g-mPEG/Epo906 that could self-assemble to form patupilone-loaded micelles (Epo906-M). The Epo906-M was able to inhibit the proliferation of A549, MCF-7 cancer cells and BEAs-2B cells in vitro. For in vivo treatment of orthotopic xenograft tumor models (MCF-7), the Epo906-M exhibited higher tumor inhibition efficiency with lower side effects as compared with free Epo906. Seventeen percent of the body weight loss appeared in the group treated with free Epo906 of 0.25 mg kg-1, while the group treated with Epo906-M of 10 mg kg-1 showed less than ten percent of body weight loss and displayed stronger tumor inhibiting effect. Therefore, the polypeptide-patupilone conjugate has improved potential for oncotherapy.


Subject(s)
Antineoplastic Agents/chemistry , Drug Carriers/chemistry , Epothilones/chemistry , Micelles , Polyethylene Glycols/chemistry , Polyglutamic Acid/analogs & derivatives , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Biological Transport , Cell Line, Tumor , Epothilones/metabolism , Epothilones/pharmacology , Humans , Polyglutamic Acid/chemistry
12.
ACS Appl Mater Interfaces ; 8(46): 31558-31566, 2016 Nov 23.
Article in English | MEDLINE | ID: mdl-27775317

ABSTRACT

Multifunctional nanoparticles with high gene transfection activity, low cytotoxicity, photoacoustic imaging ability, and photothermal therapeutic properties were prepared by conjugating low-molecular-weight polyethylenimine onto the surfaces of gold nanorods through the formation of stable S-Au bonded conjugates. Results revealed that the gene transfection efficiency of the prepared polyethylenimine-modified gold nanorods (GNRs-PEI1.8k) was higher and their cytotoxicity was less than those of the commercial reagent PEI25k. GNRs-PEI1.8k could also be potentially used as a photoacoustic and photothermal reagent to evaluate the pharmacokinetics, biodistribution, and antitumor effects of gene/drug nanoparticles. Therefore, GNRs-PEI1.8k can be considered a promising candidate for the clinical diagnosis and treatment of tumors.


Subject(s)
Nanotubes , Gold , Photoacoustic Techniques , Phototherapy , Tissue Distribution
13.
Acta Biomater ; 43: 262-268, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27431878

ABSTRACT

UNLABELLED: Photoacoustic imaging (PAI) is an emerging modality in biomedical imaging. Photoacoustic effect is the basis for PAI, where a photoacoustic contrast agent absorbs optical pulses to initiate localized heating and rapid thermal expansion, thus generating thermoelastic stress waves. Therefore, ideal PAI dyes should have strong NIR light absorbance and high light-heat conversion efficiency. However, most current low molecular weight organic PAI contrast agents are fluorescent dyes, where the light-heat conversion efficiency is dramatically impaired due to the energy loss by fluorescence emission. Herein, we report a series of highly efficient photoacoustic dyes with COOH, NH2 and NHS ester functionalities, from an inexpensive industrial computer-to-plate NIR absorber (IR830 p-toluenesulfonate) that has a strong NIR absorbance but an extremely low fluorescence emission. In vitro and in vivo studies show that the functional IR830 dyes have low cytotoxicity, and are 2.1 folds brighter in photoacoustic imaging than traditional photoacoustic dye indocyanine green (ICG). The Lowest Limit of Quantification of the IR830 series dyes is as low as the 1/7 of that of ICG. These indicate that the functional IR830 dyes have great potential as highly efficient photoacoustic dyes. STATEMENT OF SIGNIFICANCE: Photoacoustic imaging (PAI) is an emerging modality in biomedical imaging. Ideal PAI dyes should have strong NIR absorbance and high light-heat conversion efficiency. However, most current low molecular weight organic PAI contrast agents are fluorescent dyes, where the light-heat conversion efficiency is dramatically impaired due to the energy loss by fluorescence emission. Herein we report a series of highly efficient functional photoacoustic dyes from an inexpensive industrial computer-to-plate NIR absorber (IR830) that has a strong NIR absorbance but an extremely low fluorescence emission. The functional IR830 dyes show low cytotoxicity, much brighter in photoacoustic imaging than traditional photoacoustic dye indocyanine green. These indicate that the functional IR830 dyes have great potential as highly efficient photoacoustic dyes.


Subject(s)
Fluorescent Dyes/chemistry , Photoacoustic Techniques/methods , Spectroscopy, Near-Infrared/methods , Animals , Cell Death , Cell Line , Fluorescent Dyes/chemical synthesis , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet
14.
J Biomed Nanotechnol ; 12(1): 69-78, 2016 Jan.
Article in English | MEDLINE | ID: mdl-27301173

ABSTRACT

A series of novel polypeptide-based graft copolymer poly(L-glutamic acid)-graft-methoxy poly(ethylene glycol) (PLG-g-mPEG) was synthesized through a Steglich esterification reaction of PLG with mPEG. The structure of the copolymers was confirmed by nuclear magnetic resonance spectra (NMR) and gel permeation chromatography (GPC). MTT assay demonstrated that the PLG-g-mPEGs had good cell compatibility. The unreacted carboxyl groups of the PLG-g-mPEGs were used to complex cisplatin to form polymer-metal complex nanoparticles (CDDP/PLG-g-mPEG) for cancer therapy. The average hydrodynamic radius of the CDDP/PLG-g-mPEG nanoparticles was inr the range of 14-25 nm, which was beneficial for solid tumor targeting delivery. A sustained release without initial burst was achieved for the CDDP/PLG-g-mPEG nanoparticles, indicating that the CDDP-loaded nanoparticles had great potential to suppress the drug release in blood circulation before the nanoparticles had arrived at targeting tumors. The CDDP/PLG-g-mPEG nanoparticles showed a much longer blood retention profile as compared with the free CDDP. This indicated that the CDDP-loaded nanoparticles had much more opportunity to accumulate in tumor tissue by exerting the EPR effect. In vitro tests demonstrated that the CDDP/PLG-g-mPEG nanoparticles could inhibit the proliferation of HeLa, MCF-7 and A549 cancer cells. At equal dose (4 mg kg(-1)), the CDDP/PLG-g-mPEG nanoparticles showed comparable in vivo antitumor efficacy and significantly lower systemic toxicity as compared with free cis-Diaminedichloroplatinum (cisplatin, CDDP) in MCF-7 tumor bearing mice. These suggested that the CDDP/PLG-g-mPEG nanoparticle drug delivery system had a great potential to be used for cancer therapy.


Subject(s)
Cisplatin/administration & dosage , Delayed-Action Preparations/chemical synthesis , Nanocapsules/chemistry , Neoplasms, Experimental/drug therapy , Polyethylene Glycols/chemistry , Polyglutamic Acid/chemistry , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/chemistry , Cisplatin/pharmacokinetics , Delayed-Action Preparations/administration & dosage , Diffusion , Feasibility Studies , Female , Humans , Metabolic Clearance Rate , Mice , Mice, Inbred BALB C , Nanocapsules/administration & dosage , Nanocapsules/ultrastructure , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Organ Specificity , Particle Size , Tissue Distribution , Treatment Outcome
15.
Theranostics ; 6(7): 1023-30, 2016.
Article in English | MEDLINE | ID: mdl-27217835

ABSTRACT

Nanocarrier-based anti-tumor drugs hold great promise for reducing side effects and improving tumor-site drug retention in the treatment of solid tumors. However, therapeutic outcomes are still limited, primarily due to a lack of drug penetration within most tumor tissues. Herein, we propose a strategy using a nanocarrier-based combination of vascular disrupting agents (VDAs) and cytotoxic drugs for solid tumor therapy. Specifically, combretastatin A-4 (CA4) serves as a "cannon" by eradicating tumor cells at a distance from blood vessels; concomitantly, doxorubicin (DOX) serves as a "pawn" by killing tumor cells in close proximity to blood vessels. This "cannon and pawn" combination strategy acts without a need to penetrate every tumor cell and is expected to eliminate all tumor cells in a solid tumor. In a murine C26 colon tumor model, this strategy proved effective in eradicating greater than 94% of tumor cells and efficiently inhibited tumor growth with a weekly injection. In large solid tumor models (C26 and 4T1 tumors with volumes of approximately 250 mm(3)), this strategy also proved effective for inhibiting tumor growth. These results showing remarkable inhibition of tumor growth provide a valuable therapeutic choice for solid tumor therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Colonic Neoplasms/drug therapy , Doxorubicin/pharmacology , Drug Carriers/pharmacokinetics , Nanoparticles/metabolism , Stilbenes/pharmacology , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Disease Models, Animal , Doxorubicin/pharmacokinetics , Drug Therapy, Combination/methods , Mice , Stilbenes/pharmacokinetics , Treatment Outcome
16.
Nanomedicine ; 12(2): 377-86, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26711966

ABSTRACT

Disulfiram (DSF) showed great potential in an in vitro tumor therapy study; however, those results could not be applied to an in vivo study due to the extreme instability of DSF in blood. Here, we describe a system of methoxy poly(ethylene glycol)-b-poly(lactide-co-glycolide)/poly(ε-caprolactone) (mPEG-PLGA/PCL) mixed nanoparticles (NPs) for DSF loading and delivery. By adjusting the mPEG-PLGA/PCL content ratios, the DSF loading capacity increased to 7.8%, while the hydrodynamic radii of the NPs were around 50-100nm. The DSF-loaded NPs showed high stability in distilled water and 10% serum-containing phosphate buffered saline. The NPs efficiently protected DSF from degradation while maintaining its anti-tumor properties. Furthermore, a pharmacokinetics study demonstrated that NP delivery system enhanced the DSF concentration in the blood after tail vein injection. Finally, DSF delivery using this model effectively slowed the growth of a 4T1 murine xenograft tumor. FROM THE CLINICAL EDITOR: The anti-tumor efficacy of the anti-alcoholic drug disulfiram has been known for some time. However, its use in the clinical setting is limited due to the underlying instability of the drug. In this study, the authors utilized a nanocarrier system of mPEG-PLGA/PCL for the delivery of this drug. The promising results may allow encapsulation of other drugs.


Subject(s)
Alcohol Deterrents/therapeutic use , Breast Neoplasms/drug therapy , Disulfiram/therapeutic use , Drug Carriers/chemistry , Nanoparticles/chemistry , Polyesters/chemistry , Polyethylene Glycols/chemistry , Acetaldehyde Dehydrogenase Inhibitors/administration & dosage , Acetaldehyde Dehydrogenase Inhibitors/blood , Acetaldehyde Dehydrogenase Inhibitors/therapeutic use , Alcohol Deterrents/administration & dosage , Alcohol Deterrents/blood , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Antineoplastic Agents/therapeutic use , Breast/drug effects , Breast/pathology , Breast Neoplasms/pathology , Disulfiram/administration & dosage , Disulfiram/blood , Female , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Nanoparticles/ultrastructure , Rats, Sprague-Dawley
17.
Biomaterials ; 35(9): 3005-14, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24388813

ABSTRACT

There are two important obstacles for the currently applied anti-cancer drug delivery systems. One is the conflict between long-circulation and cellular uptake while the other one is the achievement of ideal anti-cancer efficacy. To solve these problems, we designed a polypeptide-based micelle system that combined the advantages of receptor mediated endocytosis and multi-drug delivery. Firstly, an amphiphilic PLG-g-Ve/PEG graft copolymer was prepared by grafting α-tocopherol (Ve) and polyethylene glycol (PEG) to poly(l-glutamic acid) (PLG). Then docetaxel (DTX) and cisplatin (CDDP) were co-loaded into the PLG-g-Ve/PEG micelles via hydrophobic and chelation effect. After that, the surface of the dual-drug-loaded micelles was decorated with an αvß3 integrin targeting peptide c(RGDfK). The targeted dual-drug-loaded micelles showed synergistic cytotoxicity and enhanced internalization rate in mouse melanoma (B16F1) cells. In vivo tests demonstrated that remarkable long circulation, anti-tumor and anti-metastasis efficacy could be achieved using this drug delivery system. This work revealed a strategy for the design and preparation of anti-cancer drug delivery systems with reduced side effect, enhanced anti-tumor and anti-metastasis efficacy.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Micelles , Peptides/chemistry , Taxoids/pharmacology , Amino Acid Sequence , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Cell Survival/drug effects , Cisplatin/pharmacokinetics , Cisplatin/therapeutic use , Docetaxel , Endocytosis/drug effects , HeLa Cells , Humans , Hydrodynamics , Magnetic Resonance Spectroscopy , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neoplasm Metastasis , Polyethylene Glycols/chemical synthesis , Polyethylene Glycols/chemistry , Polyglutamic Acid/chemical synthesis , Polyglutamic Acid/chemistry , Taxoids/pharmacokinetics , Taxoids/therapeutic use , Tissue Distribution/drug effects , Tumor Burden/drug effects
18.
Acta Biomater ; 10(3): 1392-402, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24316362

ABSTRACT

A novel methoxy poly(ethylene glycol)-b-poly(l-glutamic acid)-b-poly(l-phenylalanine) (mPEG-b-P(Glu)-b-P(Phe)) triblock copolymer was prepared and explored as a micelle carrier for the co-delivery of paclitaxel (PTX) and cisplatin (cis-diamminedichlo-platinum, CDDP). PTX and CDDP were loaded inside the hydrophobic P(Phe) inner core and chelated to the middle P(Glu) shell, respectively, while mPEG provided the outer corona for prolonged circulation. An in vitro release profile of the PTX+CDDP-loaded micelles showed that the CDDP chelation cross-link prevented an initial burst release of PTX. The PTX+CDDP-loaded micelles exhibited a high synergism effect in the inhibition of A549 human lung cancer cell line proliferation over 72 h incubation. For the in vivo treatment of xenograft human lung tumor, the PTX+CDDP-loaded micelles displayed an obvious tumor inhibiting effect with a 83.1% tumor suppression rate (TSR%), which was significantly higher than that of a free drug combination or micelles with a single drug. In addition, more importantly, the enhanced anti-tumor efficacy of the PTX+CDDP-loaded micelles came with reduced side-effects. No obvious body weight loss occurred during the treatment of A549 tumor-bearing mice with the PTX+CDDP-loaded micelles. Thus, the polypeptide-based combination of PTX and CDDP may provide useful guidance for effective and safe cancer chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Paclitaxel/pharmacology , Peptides/chemistry , Animals , Body Weight/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Endocytosis/drug effects , Glutamic Acid/analogs & derivatives , Glutamic Acid/chemical synthesis , Glutamic Acid/chemistry , Humans , Immunohistochemistry , Inhibitory Concentration 50 , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Micelles , Polyethylene Glycols/chemical synthesis , Polyethylene Glycols/chemistry , Proton Magnetic Resonance Spectroscopy , Tumor Burden/drug effects
19.
Int J Pharm ; 455(1-2): 259-66, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23876502

ABSTRACT

This study developed a novel oral insulin formulation centered on microspheres consisting of a blend of biodegradable poly(ester amide) (PEA). In the formulation, L-lysine-/L-leucine-based PEA with pendant COOH groups (PEA-COOH) was used as a pH-responsive material for the protection of insulin from the harsh environmental conditions of the stomach. Arginine-based PEA (Arg-PEA) was introduced to improve the intestinal absorption of the drug. The influence of both the hydrophobicity of PEA-COOH and the content of Arg-PEA was investigated in detail on microsphere surface morphology, drug loading, and the in vitro release profile of insulin. The PEA-COOH/Arg-PEA blend microspheres protected the loaded insulin in simulated gastric fluid and released insulin in a fast and sustained manner in simulated intestinal fluid. The in vivo test demonstrated that the oral administration of insulin-loaded PEA blend microspheres could effectively suppress the blood glucose level in diabetic rats for 10h, and the oral bioavailability was improved to 5.89+1.84% in healthy rats. These results indicate that the PEA blend microspheres are promising vehicles for the oral delivery of insulin.


Subject(s)
Hypoglycemic Agents/chemistry , Insulin/chemistry , Nylons/chemistry , Polyesters/chemistry , Administration, Oral , Animals , Arginine/chemistry , Biological Availability , Diabetes Mellitus, Experimental , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacokinetics , Insulin/administration & dosage , Insulin/pharmacokinetics , Leucine/chemistry , Lysine/chemistry , Male , Microspheres , Rats , Rats, Wistar
20.
Biomacromolecules ; 13(9): 2881-9, 2012 Sep 10.
Article in English | MEDLINE | ID: mdl-22909313

ABSTRACT

Biodegradable poly(ester amide)s have recently been used as biomaterials due to their desirable chemical and biological characteristics as well as their mechanical properties, which are amendable for material processing. In this study, electroactive tetraaniline (TA) grafted poly(ester amide)s were successfully synthesized and characterized. The poly(ester amide)s-graft-tetraaniline copolymers (PEA-g-TA) exhibited good electroactivity, mechanical properties, and biodegradability. The biocompatibility of the PEA-g-TA copolymers in vitro was systematically studied, which demonstrated that they were nontoxic and led to favorable adhesion and proliferation of mouse preosteoblastic MC3T3-E1 cells. Moreover, the PEA-g-TA copolymers stimulated by pulsed electrical signal could serve to promote the differentiation of MC3T3-E1 cells compared with TCPs. Hence, the biodegradable and electroactive PEA-g-TA copolymers possessed the properties in favor of the long-time potential application in vivo (electrical stimulation directly to the desired area) as bone repair scaffold materials in tissue engineering.


Subject(s)
Biocompatible Materials/chemical synthesis , Oligopeptides/chemistry , Osteoblasts/drug effects , Polyesters/chemical synthesis , Amides , Animals , Biocompatible Materials/pharmacology , Bone and Bones/cytology , Bone and Bones/drug effects , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Survival/drug effects , Electric Stimulation , Electricity , Magnetic Resonance Spectroscopy , Mice , Osteoblasts/cytology , Polyesters/pharmacology , Tissue Engineering , Tissue Scaffolds
SELECTION OF CITATIONS
SEARCH DETAIL
...