1
|
Meng X, Deng J, Liu F, Guo T, Liu M, Dai P, Fan A, Wang Z, Zhao Y. Triggered All-Active Metal Organic Framework: Ferroptosis Machinery Contributes to the Apoptotic Photodynamic Antitumor Therapy. NANO LETTERS 2019; 19:7866-7876. [PMID: 31594301 DOI: 10.1021/acs.nanolett.9b02904] [Citation(s) in RCA: 219] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Nanoscale photodynamic therapy (PDT) is an appealing antitumor modality for which apoptosis is the major mechanism of toxicity induction. It was postulated that the highly reactive singlet oxygen in PDT could deplete glutathione (GSH) and activate ferroptosis, the extent to which could be further manipulated by a redox-responsive nanocarrier. To validate this, a disulfide-bearing imidazole ligand coordinated with zinc to form an all-active metal organic framework (MOF) nanocarrier where a photosensitizer (chlorin e6/Ce6) was encapsulated. Regardless of light irradiation, the Ce6-loaded nanocarrier caused the depletion of intracellular GSH via the disulfide-thiol exchange reaction in a murine mammary carcinoma cell line (4T1). The GSH depletion further caused the inactivation of glutathione peroxide 4 (GPX4) and the enhancement of cytotoxicity that was alleviated by ferroptosis inhibitors. The superior in vivo antitumor efficacy of the all-active nanocarrier was corroborated in a 4T1 tumor-bearing mice model regarding tumor growth suppression and animal survival rate. The coadministration of an iron chelator weakened the antitumor potency of the nanocarrier due to ferroptosis inhibition, which was supported by the fact of tumor growth upsurge and the recovered GPX4 activity. The current work highlights the contribution of ferroptotic machinery to antitumor PDT via an activatable, adaptable, all-active MOF nanocarrier.
Collapse
|
|
6 |
219 |
2
|
Xiao D, Jia HZ, Zhang J, Liu CW, Zhuo RX, Zhang XZ. A dual-responsive mesoporous silica nanoparticle for tumor-triggered targeting drug delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2014; 10:591-598. [PMID: 24106109 DOI: 10.1002/smll.201301926] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Revised: 08/16/2013] [Indexed: 06/02/2023]
Abstract
A novel pH- and redox- dual-responsive tumor-triggered targeting mesoporous silica nanoparticle (TTTMSN) is designed as a drug carrier. The peptide RGDFFFFC is anchored on the surface of mesoporous silica nanoparticles via disulfide bonds, which are redox-responsive, as a gatekeeper as well as a tumor-targeting ligand. PEGylated technology is employed to protect the anchored peptide ligands. The peptide and monomethoxypolyethylene glycol (MPEG) with benzoic-imine bond, which is pH-sensitive, are then connected via "click" chemistry to obtain TTTMSN. In vitro cell research demonstrates that the targeting property of TTTMSN is switched off in normal tissues with neutral pH condition, and switched on in tumor tissues with acidic pH condition after removing the MPEG segment by hydrolysis of benzoic-imine bond under acidic conditions. After deshielding of the MPEG segment, the drug-loaded nanoparticles are easily taken up by tumor cells due to the exposed peptide targeting ligand, and subsequently the redox signal glutathione in tumor cells induces rapid drug release intracellularly after the cleavage of disulfide bond. This novel intelligent TTTMSN drug delivery system has great potential for cancer therapy.
Collapse
|
|
11 |
157 |
3
|
Lei B, Wang M, Jiang Z, Qi W, Su R, He Z. Constructing Redox-Responsive Metal-Organic Framework Nanocarriers for Anticancer Drug Delivery. ACS APPLIED MATERIALS & INTERFACES 2018; 10:16698-16706. [PMID: 29692177 DOI: 10.1021/acsami.7b19693] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Metal-organic frameworks (MOFs), which are a unique class of hybrid porous materials built from metal ions and organic ligands, have attracted significant interest in recent years as a promising platform for controlled drug delivery. Current approaches for creating MOFs-based responsive drug carriers involve encapsulation of stimuli-responsive compositions into MOFs or postsynthetic surface modification with sensitive molecules. In this study, we developed a novel intrinsic redox-responsive MOFs carrier, MOF-M(DTBA) (M = Fe, Al or Zr) by using iron, aluminum, or zirconium as metal nodes and 4,4'-dithiobisbenzoic acid (4,4'-DTBA) as the organic ligand. The disulfide bond in 4,4'-DTBA is cleavable by glutathione (GSH), which is often overexpressed in tumor cells. It was found that MOF-Zr(DTBA) synthesized at 40 °C displayed the appropriate size and properties as a drug carrier. By incorporating curcumin (CCM) into MOF-Zr(DTBA), CCM@MOF-Zr(DTBA) nanoparticles were obtained that displayed a faster releasing behavior in vitro and enhanced the cell death compared with free CCM. The in vivo anticancer experiments indicate that CCM@ MOF-Zr(DTBA) exhibits much higher antitumor efficacy than free CCM. This strategy for constructing responsive MOFs-based nanocarriers might open new possibilities for the application of MOFs in drug delivery, molecular imaging, or theranostics.
Collapse
|
|
7 |
118 |
4
|
Zhao Q, Geng H, Wang Y, Gao Y, Huang J, Wang Y, Zhang J, Wang S. Hyaluronic acid oligosaccharide modified redox-responsive mesoporous silica nanoparticles for targeted drug delivery. ACS APPLIED MATERIALS & INTERFACES 2014; 6:20290-20299. [PMID: 25311422 DOI: 10.1021/am505824d] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
A redox-responsive delivery system based on colloidal mesoporous silica (CMS) has been developed, in which 6-mercaptopurine (6-MP) was conjugated to vehicles by cleavable disulfide bonds. The oligosaccharide of hyaluronic acid (oHA) was modified on the surface of CMS by disulfide bonds as a targeting ligand and was able to increase the stability and biocompatibility of CMS under physiological conditions. In vitro release studies indicated that the cumulative release of 6-MP was less than 3% in the absence of glutathione (GSH), and reached nearly 80% within 2 h in the presence of 3 mM GSH. Confocal microscopy and fluorescence-activated cell sorter (FACS) methods were used to evaluate the cellular uptake performance of fluorescein isothiocyanate (FITC) labeled CMS, with and without oHA modification. The CMS-SS-oHA exhibited a higher cellular uptake performance via CD44 receptor-mediated endocytosis in HCT-116 (CD44 receptor-positive) cells than in NIH-3T3 (CD44 receptor-negative) cells. 6-MP loaded CMS-SS-oHA exhibited greater cytotoxicity against HCT-116 cells than NIH-3T3 cells due to the enhanced cell uptake behavior of CMS-SS-oHA. This study provides a novel strategy to covalently link bioactive drug and targeting ligand to the interiors and exteriors of mesoporous silica to construct a stimulus-responsive targeted drug delivery system.
Collapse
|
|
11 |
112 |
5
|
Municoy S, Álvarez Echazú MI, Antezana PE, Galdopórpora JM, Olivetti C, Mebert AM, Foglia ML, Tuttolomondo MV, Alvarez GS, Hardy JG, Desimone MF. Stimuli-Responsive Materials for Tissue Engineering and Drug Delivery. Int J Mol Sci 2020; 21:E4724. [PMID: 32630690 PMCID: PMC7369929 DOI: 10.3390/ijms21134724] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/28/2020] [Accepted: 06/30/2020] [Indexed: 02/07/2023] Open
Abstract
Smart or stimuli-responsive materials are an emerging class of materials used for tissue engineering and drug delivery. A variety of stimuli (including temperature, pH, redox-state, light, and magnet fields) are being investigated for their potential to change a material's properties, interactions, structure, and/or dimensions. The specificity of stimuli response, and ability to respond to endogenous cues inherently present in living systems provide possibilities to develop novel tissue engineering and drug delivery strategies (for example materials composed of stimuli responsive polymers that self-assemble or undergo phase transitions or morphology transformations). Herein, smart materials as controlled drug release vehicles for tissue engineering are described, highlighting their potential for the delivery of precise quantities of drugs at specific locations and times promoting the controlled repair or remodeling of tissues.
Collapse
|
Review |
5 |
97 |
6
|
Chen L, Zhou X, Nie W, Zhang Q, Wang W, Zhang Y, He C. Multifunctional Redox-Responsive Mesoporous Silica Nanoparticles for Efficient Targeting Drug Delivery and Magnetic Resonance Imaging. ACS APPLIED MATERIALS & INTERFACES 2016; 8:33829-33841. [PMID: 27960384 DOI: 10.1021/acsami.6b11802] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The convenient modification of mesoporous silica nanoparticles (MSN) can provide great opportunities for constructing a new generation of nanocarriers with multiple functions. In the current study, we fabricated a new multifunctional drug delivery system based on MSN capped by gadolinium-based bovine serum albumin complex (BSA-Gd) and hyaluronic acid (HA) via reductive-cleavable disulfide bond. In this multifunctional nanoparticle (MSN-ss-GHA), BSA-Gd component was prepared by biomineralization and acted as both smart gatekeeper and contrast agent for magnetic resonance (MR) imaging, while HA served as the targeted molecule to improve the specific affinity of MSN-ss-GHA toward cancer cells. The successful fabrication of MSN-ss-GHA was demonstrated by a series of physicochemical characterization. The redox-sensitive drug release behavior of doxorubicin hydrochloride (DOX) loaded MSN-ss-GHA (DOX@MSN-ss-GHA) was also verified. Comparatively, the MSN-ss-GHA exhibited excellent biocompatibility and distinctly enhanced cell uptake by 4T1 cells. More importantly, the improved in vitro MR imaging ability of MSN-ss-GHA over that of Gd-DTPA was also confirmed. The results also suggested that the DOX@MSN-ss-GHA could efficiently deliver DOX into 4T1 cells and showed enhanced cytotoxicity as compared to that of nontargeted nanocarrier. The in vivo experiment also demonstrated the negligible toxicity of MSN-ss-GHA and improved antitumor suppression of DOX@MSN-ss-GHA. Thus, this multifunctional MSN-based theranostic agent holds potential for efficient redox-responsive targeting drug delivery and MR imaging.
Collapse
|
|
9 |
90 |
7
|
Su Y, Hu Y, Du Y, Huang X, He J, You J, Yuan H, Hu F. Redox-responsive polymer-drug conjugates based on doxorubicin and chitosan oligosaccharide-g-stearic acid for cancer therapy. Mol Pharm 2015; 12:1193-202. [PMID: 25751168 DOI: 10.1021/mp500710x] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Here, a biodegradable polymer-drug conjugate of doxorubicin (DOX) conjugated with a stearic acid-grafted chitosan oligosaccharide (CSO-SA) was synthesized via disulfide linkers. The obtained polymer-drug conjugate DOX-SS-CSO-SA could self-assemble into nanosized micelles in aqueous medium with a low critical micelle concentration. The size of the micelles was 62.8 nm with a narrow size distribution. In reducing environments, the DOX-SS-CSO-SA could rapidly disassemble result from the cleavage of the disulfide linkers and release the DOX. DOX-SS-CSO-SA had high efficiency for cellular uptake and rapidly released DOX in reductive intracellular environments. In vitro antitumor activity tests showed that the DOX-SS-CSO-SA had higher cytotoxicity against DOX-resistant cells than free DOX, with reversal ability up to 34.8-fold. DOX-SS-CSO-SA altered the drug distribution in vivo, which showed selectively accumulation in tumor and reduced nonspecific accumulation in hearts. In vivo antitumor studies demonstrated that DOX-SS-CSO-SA showed efficient suppression on tumor growth and relieved the DOX-induced cardiac injury. Therefore, DOX-SS-CSO-SA is a potential drug delivery system for safe and effective cancer therapy.
Collapse
|
Research Support, Non-U.S. Gov't |
10 |
78 |
8
|
Xu X, Wu J, Liu S, Saw PE, Tao W, Li Y, Krygsman L, Yegnasubramanian S, De Marzo AM, Shi J, Bieberich CJ, Farokhzad OC. Redox-Responsive Nanoparticle-Mediated Systemic RNAi for Effective Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1802565. [PMID: 30230235 PMCID: PMC6286670 DOI: 10.1002/smll.201802565] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/21/2018] [Indexed: 05/16/2023]
Abstract
Biodegradable polymeric nanoparticles (NPs) have demonstrated significant potential to improve the systemic delivery of RNA interference (RNAi) therapeutics, such as small interfering RNA (siRNA), for cancer therapy. However, the slow and inefficient siRNA release inside tumor cells generally observed for most biodegradable polymeric NPs may result in compromised gene silencing efficacy. Herein, a biodegradable and redox-responsive NP platform, composed of a solid poly(disulfide amide) (PDSA)/cationic lipid core and a lipid-poly(ethylene glycol) (lipid-PEG) shell for systemic siRNA delivery to tumor cells, is developed. This newly generated NP platform can efficiently encapsulate siRNA under extracellular environments and can respond to the highly concentrated glutathione (GSH) in the cytoplasm to induce fast intracellular siRNA release. By screening a library of PDSA polymers with different structures and chain lengths, the optimized NP platform shows the unique features of i) long blood circulation, ii) high tumor accumulation, iii) fast GSH-triggered intracellular siRNA release, and iv) exceptionally effective gene silencing. Together with the facile polymer synthesis technique and robust NP formulation enabling scale-up, this new redox-responsive NP platform may become an effective tool for RNAi-based cancer therapy.
Collapse
|
Research Support, N.I.H., Extramural |
7 |
78 |
9
|
Xiao Y, Lu C, Liu Y, Kong L, Bai H, Mu H, Li Z, Geng H, Duan J. Encapsulation of Lactobacillus rhamnosus in Hyaluronic Acid-Based Hydrogel for Pathogen-Targeted Delivery to Ameliorate Enteritis. ACS APPLIED MATERIALS & INTERFACES 2020; 12:36967-36977. [PMID: 32702229 DOI: 10.1021/acsami.0c11959] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Probiotics were found to be effective in ameliorating the microbial dysbiosis and inflammation caused by intestinal pathogens. However, biological challenges encountered during oral delivery have greatly limited their potential health benefits. Here, a model probiotic (Lactobacillus rhamnosus) was encapsulated in an intestinal-targeted hydrogel to alleviate bacterial enteritis in a novel mode. The hydrogel was prepared simply by the self-cross-linking of thiolated hyaluronic acid. Upon exposure to H2S which were excreted by surrounding intestinal pathogens, the hydrogel can locally degrade and rapidly release cargos to compete with source pathogens in turn for binding to the host. The mechanical properties of hydrogel were studied by rheological analysis, and the ideal stability was achieved at a polymer concentration of 4% (w/v). The morphology of the optimal encapsulation system was further measured by a scanning electron microscope, exhibiting uniform payload of probiotics. Endurance experiments indicated that the encapsulation of L. rhamnosus significantly enhanced their viability under gastrointestinal tract insults. Compared with free cells, encapsulated L. rhamnosus exerted better therapeutic effect against Salmonella-induced enteritis with negligible toxicity in vivo. These results demonstrate that this redox-responsive hydrogel may be a promising encapsulation and delivery system for improving the efficacy of orally administered probiotics.
Collapse
|
|
5 |
75 |
10
|
Zhang L, Wang Y, Zhang X, Wei X, Xiong X, Zhou S. Enzyme and Redox Dual-Triggered Intracellular Release from Actively Targeted Polymeric Micelles. ACS APPLIED MATERIALS & INTERFACES 2017; 9:3388-3399. [PMID: 28071889 DOI: 10.1021/acsami.6b14078] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Highly effective delivery of therapeutic agents into target cells using nanocarriers and subsequently rapid intracellular release are of great importance in cancer treatment. Here, we developed an enzyme and redox dual-responsive polymeric micelle with active targeting abilities to achieve rapid intracellular drug release. To overcome both its poor solubility in water and instability in the blood circulation, camptothecin (CPT) was chemically conjugated to monomethyl poly(ethylene glycol) (mPEG) via a redox-responsive linker to form polymeric prodrugs. The enzyme-responsive function is achieved by connecting hydrophobic polycaprolactone segments and hydrophilic PEG segments with azo bonds. Additionally, the end of the PEG segment was decorated with phenylboronic acid (PBA), endowing the nanocarriers with active targeting abilities. The dual-responsive targeting polymeric micelles can be generated by self-assembly of a mixture of the polymeric prodrug and enzyme-responsive copolymer. The in vitro drug release profile revealed that CPT was rapidly released from the micelles under a simulated condition similar to the tumor cell microenvironment. In vivo and ex vivo fluorescence imaging indicated that these micelles possess excellent specificity to target hepatoma carcinoma cells. The antitumor effect in mice liver cancer cells (H22) in tumor-bearing Kunming (KM) mice demonstrated that this nanocarrier exhibits high therapeutic efficiency in artificial solid tumors and low toxicity to normal tissues, with a survival rate of approximately 100% after 160 days of treatment.
Collapse
|
|
8 |
69 |
11
|
Kang Y, Ju X, Ding LS, Zhang S, Li BJ. Reactive Oxygen Species and Glutathione Dual Redox-Responsive Supramolecular Assemblies with Controllable Release Capability. ACS APPLIED MATERIALS & INTERFACES 2017; 9:4475-4484. [PMID: 28103014 DOI: 10.1021/acsami.6b14640] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
A dual redox and biorelevant triggered supramolecular system is developed through noncovalent supramolecular inclusion interactions between the ferrocene (Fc) modified on camptothecin (CPT) and β-cyclodextrin (β-CD) at the end of methoxy polyethylene glycol (mPEG). With these two segments, a stable noncovalent supramolecular structure, i.e., mPEG-β-CD/Fc-CPT, can be formed, and then self-assembled into micellar structures in water. Interestingly, these supramolecular micelles showed uniform sphere structure, high and constant drug loading content, hyper-fast redox-responsive drug release, and exhibited equal cellular proliferation inhibition toward A549 cancer cells. The cytotoxicity evaluation of mPEG-β-CD also indicated good biocompatibility. In vivo results revealed the mPEG-β-CD/Fc-CPT nanoparticles had higher in vivo efficacy without side effects. It is anticipated this supramolecular complex may serve as a new kind of promising alternative for drug delivery systems.
Collapse
|
|
8 |
67 |
12
|
Du D, Wang K, Wen Y, Li Y, Li YY. Photodynamic Graphene Quantum Dot: Reduction Condition Regulated Photoactivity and Size Dependent Efficacy. ACS APPLIED MATERIALS & INTERFACES 2016; 8:3287-94. [PMID: 26761130 DOI: 10.1021/acsami.5b11154] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Prequenching and selective activation of photosensitizer (PS) are highly desired in photodynamic therapy (PDT) to avoid off-target effect due to nonspecific activation and poor targeting selectivity of PS. In this study, nanographene materials as a unique π-conjugated planar system for electronic transfer were employed as the robust platform for temporarily quenching of PS. Photosensitizer chlorin e6 (Ce6) was integrated onto planar structure of graphene quantum dot (GQD) or graphene oxide (GO) via a reduction cleavable disulfide linker. The formed hybrid nanosystem displayed considerable fluorescence quenching and slight phototoxicity, even under the condition of light irradiation, while the photoactivity of PS could be selectively recovered in the presence of the reducing agent. Compared with graphene oxide system with larger size (around 200 nm), GQD nanosystem exhibited significantly improved tumor accumulation via enhanced permeation and retention effect (EPR effect). The in vivo study demonstrated extremely effective suppression of tumor growth for the group treated with the GQD nanosystem with cleavable linker, revealing the promising application of the presented novel strategy.
Collapse
|
|
9 |
62 |
13
|
Li Y, Hu H, Zhou Q, Ao Y, Xiao C, Wan J, Wan Y, Xu H, Li Z, Yang X. α-Amylase- and Redox-Responsive Nanoparticles for Tumor-Targeted Drug Delivery. ACS APPLIED MATERIALS & INTERFACES 2017; 9:19215-19230. [PMID: 28513132 DOI: 10.1021/acsami.7b04066] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Paclitaxel (PTX) is an effective antineoplastic agent and shows potent antitumor activity against a wide spectrum of cancers. Yet, the wide clinical use of PTX is limited by its poor aqueous solubility and the side effects associated with its current therapeutic formulation. To tackle these obstacles, we report, for the first time, α-amylase- and redox-responsive nanoparticles based on hydroxyethyl starch (HES) for the tumor-targeted delivery of PTX. PTX is conjugated onto HES by a redox-sensitive disulfide bond to form HES-SS-PTX, which was confirmed by results from NMR, high-performance liquid chromatography-mass spectrometry, and Fourier transform infrared spectrometry. The HES-SS-PTX conjugates assemble into stable and monodispersed nanoparticles (NPs), as characterized with Dynamic light scattering, transmission electron microscopy, and atomic force microscopy. In blood, α-amylase will degrade the HES shell and thus decrease the size of the HES-SS-PTX NPs, facilitating NP extravasation and penetration into the tumor. A pharmacokinetic study demonstrated that the HES-SS-PTX NPs have a longer half-life than that of the commercial PTX formulation (Taxol). As a consequence, HES-SS-PTX NPs accumulate more in the tumor compared with the extent of Taxol, as shown in an in vivo imaging study. Under reductive conditions, the HES-SS-PTX NPs could disassemble quickly as evidenced by their triggered collapse, burst drug release, and enhanced cytotoxicity against 4T1 tumor cells in the presence of a reducing agent. Collectively, the HES-SS-PTX NPs show improved in vivo antitumor efficacy (63.6 vs 52.4%) and reduced toxicity in 4T1 tumor-bearing mice compared with those of Taxol. These results highlight the advantages of HES-based α-amylase- and redox-responsive NPs, showing their great clinical translation potential for cancer chemotherapy.
Collapse
|
|
8 |
58 |
14
|
Guo Y, Zhang Y, Li J, Zhang Y, Lu Y, Jiang X, He X, Ma H, An S, Jiang C. Cell microenvironment-controlled antitumor drug releasing-nanomicelles for GLUT1-targeting hepatocellular carcinoma therapy. ACS APPLIED MATERIALS & INTERFACES 2015; 7:5444-5453. [PMID: 25686400 DOI: 10.1021/am5091462] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
In clinical therapy, the poor prognosis of hepatocellular carcinoma (HCC) is mainly attributed to the failure of chemotherapeutical agents to accumulate in tumor as well as their serious systemic toxicity. In this work, we developed actively tumor-targeting trilayer micelles with microenvironment-sensitive cross-links as a novel nanocarrier for HCC therapy. These micelles comprised biodegradable PEG-pLys-pPhe polymers, in which pLys could react with a disulfide-containing agent to form redox-responsive cross-links. In vitro drug release and pharmacokinetics studies showed that these cross-links were stable in physiological condition whereas cleaved once internalized into cells due to the high level of glutathione, resulting in facilitated intracellular doxorubicin release. In addition, dehydroascorbic acid (DHAA) was decorated on the surface of micelles for specific recognition of tumor cells via GLUT1, a member of glucose transporter family overexpressed on hepatocarcinoma cells. Moreover, DHAA exhibited a "one-way" continuous accumulation within tumor cells. Cellular uptake and in vivo imaging studies proved that these micelles had remarkable targeting property toward hepatocarcinoma cells and tumor. Enhanced anti-HCC efficacy of the micelles was also confirmed both in vitro and in vivo. Therefore, this micellar system may be a potential platform of chemotherapeutics delivery for HCC therapy.
Collapse
|
|
10 |
55 |
15
|
Moghaddam SPH, Saikia J, Yazdimamaghani M, Ghandehari H. Redox-Responsive Polysulfide-Based Biodegradable Organosilica Nanoparticles for Delivery of Bioactive Agents. ACS APPLIED MATERIALS & INTERFACES 2017; 9:21133-21146. [PMID: 28609092 PMCID: PMC5665166 DOI: 10.1021/acsami.7b04351] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Design and development of silica nanoparticles (SiO2 NPs) with a controlled degradation profile promises effective drug delivery with a predetermined carrier elimination profile. In this research, we fabricated a series of redox-responsive polysulfide-based biodegradable SiO2 NPs with low polydispersity and with variations in size (average diameters of 58 ± 7, 108 ± 11, 110 ± 9, 124 ± 9, and 332 ± 6 nm), porosity, and composition (disulfide vs tetrasulfide bonds). The degradation kinetics of the nanoparticles was analyzed in the presence of 8 mM glutathione (GSH), mimicking the intracellular reducing condition. Results indicate that porosity and core composition play the predominant roles in the degradation rate of these nanoparticles. The 108 nm mesoporous disulfide-based nanoparticles showed the highest degradation rate among all the synthesized nanoparticles. Transmission electron microscopy (TEM) reveals that nonporous nanoparticles undergo surface erosion, while porous nanoparticles undergo both surface and bulk erosion under reducing environment. The cytotoxicity of these nanoparticles in RAW 264.7 macrophages was evaluated. Results show that all these nanoparticles with the IC50 values ranging from 233 ± 42 to 705 ± 17 μg mL-1 do not have cytotoxic effect in macrophages at concentrations less than 125 μg mL-1. The degradation products of these nanoparticles collected within 15 days did not show cytotoxicity in the same macrophage cell line after 24 h of incubation. In vitro doxorubicin (DOX) release was examined in 108 nm mesoporous disulfide-based nanoparticles in the absence and presence of 8 mM GSH. It was shown that drug release depends on intracellular reducing conditions. Due to their ease of synthesis and scale up, robust structure, and the ability to control size, composition, release, and elimination, biodegradable SiO2 NPs provide an alternative platform for delivery of bioactive and imaging agents.
Collapse
|
research-article |
8 |
51 |
16
|
Yang Y, Zhu H, Wang J, Fang Q, Peng Z. Enzymatically Disulfide-Crosslinked Chitosan/Hyaluronic Acid Layer-by-Layer Self-Assembled Microcapsules for Redox-Responsive Controlled Release of Protein. ACS APPLIED MATERIALS & INTERFACES 2018; 10:33493-33506. [PMID: 30203959 DOI: 10.1021/acsami.8b07120] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Disulfide-crosslinked hollow polyelectrolyte microcapsules composed of thiolated chitosan (CS-SH) and hyaluronic acid (HA-SH) were prepared by combining the layer-by-layer (LBL) technique and horseradish peroxidase (HRP)-mediated oxidative cross-linking reaction in mild conditions. FITC-dextran-doped CaCO3 microspheres were used as template core and removed after LBL depositing CS-SH and HA-SH on the surface. The disulfide-crosslinked (CS/HA) microcapsules were readily fabricated by HRP-mediated oxidative coupling of the thiol groups in CS/HA shell layer in the presence of HRP (10 units/mL) and Tyramine hydrochloride (Tyr, 35 mmol/L). The kinetics of enzymatic disulfide-crosslinking reaction was investigated through the real-time monitoring of the consumption of thiol groups by UV absorption spectra. It found that the formation of disulfide linkages by the enzymatic thiol oxidation reaction showed a gradual acceleration. The disulfide-crosslinked CS/HA hydrogel were rapidly formed in gelation time between approximately 17 and 30 min, which were dependent on the concentrations of HRP and Tyr. The disulfide linkages endowed the microcapsule-enhanced physical stability and low permeability under physiological conditions and redox-responsive degradability in reducing environments. The structural stability of disulfide-crosslinked (CS/HA) microcapsules was visualized by confocal laser scanning microscopy in phosphate-buffered saline containing 5.0 mmol/L dithiothreitol (DTT) to evaluate the redox-responsive disassembly process. Redox-responsive controlled release of encapsulated FITC-dextran from the disulfide-crosslinked (CS/HA) microcapsules were obtained. The release profiles of FITC-dextran could be manipulated by controlling the shell thickness and the concentration of DTT. The conformational stability analyses and more than 94% esterase activity of released bovine serum albumin (BSA) from (CS/HA) microcapsules conformed that the structural integrity and bioactivity were well preserved during the encapsulation and release process. The microcapsules exhibited excellent cytocompatibility for HEK 293 cells up to a concentration of 1.0 mg/mL. The microcapsules efficiently delivered loaded FITC-BSA into HeLa cells and released the protein in the reducing cytosol. This study proposed a novel approach for producing disulfide-crosslinked microcarriers for intracellular delivery and redox-responsive controlled release of protein.
Collapse
|
|
7 |
47 |
17
|
Liu N, Tan Y, Hu Y, Meng T, Wen L, Liu J, Cheng B, Yuan H, Huang X, Hu F. A54 Peptide Modified and Redox-Responsive Glucolipid Conjugate Micelles for Intracellular Delivery of Doxorubicin in Hepatocarcinoma Therapy. ACS APPLIED MATERIALS & INTERFACES 2016; 8:33148-33156. [PMID: 27934140 DOI: 10.1021/acsami.6b09333] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Redox-responsive nanomaterials applied in drug delivery systems (DDS) have attracted an increasing attention in pharmaceutical research as a carrier for antitumor therapy. However, there would be unwanted drug release from a redox-responsive DDS with no selection at nontarget sites, leading to undesirable toxicities in normal tissues and cells. Here, an A54 peptide modified and PEGylated reduction cleavable glucolipid conjugate (A54-PEG-CSO-ss-SA, abbreviated to APCssA) was designed for intracellular delivery of doxorubicin (DOX). The synthesized APCssA could be assembled via micellization self-assembly in aqueous water above the critical micelle concentration (54.9 μg/mL) and exhibited a high drug encapsulation efficiency (77.92%). The APCssA micelles showed an enhanced redox sensitivity in that the disulfide bond could be degraded quickly and the drug would be released from micelles in 10 mM levels of glutathione (GSH). The cellular uptake studies highlighted the affinity of APCssA micelles toward the hepatoma cells (BEL-7402) compared to that toward HepG2 cells. In contrast with the nonresponsive conjugate, the drug was released from APCssA micelles more quickly in 10 mM level of GSH concentration (tumor cells). Moreover, the DOX-loaded APCssA micelles displayed an increased cytotoxicity which was 1.6- to 2.0-fold that of unmodified and nonresponsive micelles. In vivo, the APCssA micelles had stronger distribution to liver and hepatoma tissue and prolonged the circulation and retention time, while the drug release only occurred in the tumor tissue. The APCssA/DOX showed the tumor inhibition rate equal to that of commercial doxorubicin hydrochloric without negative consequence. This study suggested that the APCssA/DOX showed promising potential to treat the tumor for its special tumor targeting, selective intracellular drug release, enhanced antitumor activity, and reduced toxicity on normal tissues.
Collapse
|
|
9 |
46 |
18
|
Xu C, Song RJ, Lu P, Chen JC, Zhou YQ, Shen G, Jiang MJ, Zhang W. pH-triggered charge-reversal and redox-sensitive drug-release polymer micelles codeliver doxorubicin and triptolide for prostate tumor therapy. Int J Nanomedicine 2018; 13:7229-7249. [PMID: 30510415 PMCID: PMC6231516 DOI: 10.2147/ijn.s182197] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
AIM To significantly promote cancer cell uptake and to achieve combination therapy and on-demand drug release, a pH-triggered charge-switchable and redox-responsive drug-release nanovehicle was developed in this study. MATERIALS AND METHODS The nanocarrier was constructed by conjugating 3,3'-dithiodipropionic acid-modified doxorubicin (DTPA-DOX) and 2,3-dimethylmaleic anhydride (DMA) to the side amino groups of poly(ethylene glycol)-b-poly(L-lysine) (PEG-b-PLL) and by encapsulating triptolide (TRI) into the hydrophobic core. The surface charge of the obtained nanocarriers (DA-ss-DT) can change from negative to positive in response to tumor extracellular acidity pH, and the nanocarriers capably release two drugs in response to intracellular high glutathione (GSH) environment. RESULTS Compared to the control group, the in vitro cellular uptake of DA-ss-DT by human prostate cancer PC-3 cells was significantly promoted in slightly acidic conditions, and the drug could be rapidly released in the high concentration of GSH conditions. The in vitro and in vivo antitumor experiments exhibited that the DA-ss-DT nanoparticles have a great antitumor effect in comparison to the control group. CONCLUSION These findings demonstrated that the DA-ss-DT nanoparticles supply a useful strategy for promoting cellular uptake and synergetic anticancer therapy.
Collapse
|
research-article |
7 |
43 |
19
|
Zhao N, Lin X, Zhang Q, Ji Z, Xu FJ. Redox-Triggered Gatekeeper-Enveloped Starlike Hollow Silica Nanoparticles for Intelligent Delivery Systems. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2015; 11:6467-6479. [PMID: 26528765 DOI: 10.1002/smll.201502760] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Indexed: 06/05/2023]
Abstract
The design and development of multifunctional carriers for drug delivery based on hollow nanoparticles (HNPs) have attracted intense interests. Ordinary spherical HNPs are demonstrated to be promising candidates. However, the application of HNPs with special morphologies has rarely been reported. HNPs with sharp horns are expected to own higher endocytosis efficiencies than spherical counterparts. In this work, novel starlike hollow silica nanoparticles (SHNPs) with different sizes are proposed as platforms for the fabrication of redox-triggered multifunctional systems for synergy of gene therapy and chemotherapy. The CD-PGEA gene vectors (consisting of β-CD cores and ethanolamine-functionalized poly(glycidyl methacrylate) (denoted BUCT-PGEA) arms) are introduced ingeniously onto the surfaces of SHNPs with plentiful disulfide bond-linked adamantine guests. The resulting supramolecular assemblies (SHNP-PGEAs) possess redox-responsive gatekeepers for loaded drugs in the cavities of SHNPs. Meanwhile, they also demonstrate excellent performances to deliver genes. The gene transfection efficiencies, controlled drug release behaviors, and synergistic antitumor effect of hollow silica-based carriers with different morphologies are investigated in detail. Compared with ordinary spherical HNP-based counterparts, SHNP-PGEA carriers with six sharp horns are proven to be superior gene vectors and possess better efficacy for cellular uptake and antitumor effects. The present multifunctional carriers based on SHNPs will have promising applications in drug/gene codelivery and cancer treatment.
Collapse
|
|
10 |
43 |
20
|
Park HK, Lee SJ, Oh JS, Lee SG, Jeong YIL, Lee HC. Smart Nanoparticles Based on Hyaluronic Acid for Redox-Responsive and CD44 Receptor-Mediated Targeting of Tumor. NANOSCALE RESEARCH LETTERS 2015; 10:981. [PMID: 26163139 PMCID: PMC4499038 DOI: 10.1186/s11671-015-0981-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 06/14/2015] [Indexed: 05/27/2023]
Abstract
BACKGROUND Since aggressive cancer cells highly express the CD44 receptor compared to normal cells, hyaluronic acid (HA) can be used for CD44 targeting molecule. Since glutathione (GSH) level is normally elevated in the intracellular compartment and in the tumor cell, the fact that disulfide bond can be cleaved by GSH is widely used for intracellular drug delivery. METHODS HA was connected with poly(DL-lactide-co-glycolide) (PLGA) using disulfide linkage, and then a diblock copolymer (HAssLG) was prepared. Doxorubicin (DOX)-loaded HAssLG nanoparticles were prepared by dialysis procedures. RESULTS AND DISCUSSION DOX-loaded HAssLG nanoparticles have spherical shapes with small particle size of less than 300 nm. In fluorescence measurement, DOX was dose-dependently liberated from nanoparticles by the addition of GSH. DOX release rate from HAssLG nanoparticles was increased by the addition of GSH. To confirm CD44 receptor-mediated endocytosis of nanoparticles, CD44-positive MDA-MB231 cells were employed and fluorescence intensity was strong when nanoparticles were treated to tumor cells. However, fluorescence intensity was significantly decreased through blocking of the CD44 receptor by pretreatment of cells with free HA. Fluorescence intensity of cells was increased again when GSH was added, indicating that HAssLG nanoparticles have CD44 receptor targetability and potential of redox-responsive drug delivery. For animal imaging study, CD44-positive MDA-MB231 cells and CD44-negative NIH3T3 cells were simultaneously implanted into the right flank and left flank of mice, respectively. Fluorescence intensity was significantly stronger at tumor mass of MDA-MB231 cells than solid mass of NIH3T3 cells, indicating that HAssLG nanoparticles were specifically delivered to tumor cells. CONCLUSIONS The results indicated that HAssLG nanoparticles have specificity against the CD44 receptor and can be used for anticancer drug targeting. We recommend HAssLG nanoparticles as a promising vehicle for cancer drug targeting.
Collapse
|
research-article |
10 |
41 |
21
|
Devnarain N, Osman N, Fasiku VO, Makhathini S, Salih M, Ibrahim UH, Govender T. Intrinsic stimuli-responsive nanocarriers for smart drug delivery of antibacterial agents-An in-depth review of the last two decades. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 13:e1664. [PMID: 32808486 DOI: 10.1002/wnan.1664] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/30/2020] [Accepted: 07/20/2020] [Indexed: 12/18/2022]
Abstract
Antibiotic resistance due to suboptimal targeting and inconsistent antibiotic release at bacterial infection sites has driven the formulation of stimuli-responsive nanocarriers for antibacterial therapy. Unlike conventional nanocarriers, stimuli-responsive nanocarriers have the ability to specifically enhance targeting and drug release profiles. There has been a significant escalation in the design and development of novel nanomaterials worldwide; in particular, intrinsic stimuli-responsive antibiotic nanocarriers, due to their enhanced activity, improved targeted delivery, and superior potential for bacterial penetration and eradication. Herein, we provide an extensive and critical review of pH-, enzyme-, redox-, and ionic microenvironment-responsive nanocarriers that have been reported in literature to date, with an emphasis on the mechanisms of drug release, the nanomaterials used, the nanosystems constructed and the antibacterial efficacy of the nanocarriers. The review also highlights further avenues of research for optimizing their potential and commercialization. This review confirms the potential of intrinsic stimuli-responsive nanocarriers for enhanced drug delivery and antibacterial killing. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
|
Research Support, Non-U.S. Gov't |
5 |
40 |
22
|
Ma YC, Wang JX, Tao W, Sun CY, Wang YC, Li DD, Fan F, Qian HS, Yang XZ. Redox-Responsive Polyphosphoester-Based Micellar Nanomedicines for Overriding Chemoresistance in Breast Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2015; 7:26315-26325. [PMID: 26552849 DOI: 10.1021/acsami.5b09195] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
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.
Collapse
|
|
10 |
37 |
23
|
Yang Q, Li L, Sun W, Zhou Z, Huang Y. Dual Stimuli-Responsive Hybrid Polymeric Nanoparticles Self-Assembled from POSS-Based Starlike Copolymer-Drug Conjugates for Efficient Intracellular Delivery of Hydrophobic Drugs. ACS APPLIED MATERIALS & INTERFACES 2016; 8:13251-61. [PMID: 27167898 DOI: 10.1021/acsami.6b02403] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
To further fine tune drug release and enhance therapeutic effects of polyhedral oligomericsilsesquioxane (POSS)-based nanomedicine, a starlike organic-inorganic conjugate was synthesized by grafting semitelechelic N-(2-hydroxypropyl) methacrylamide (HPMA) copolymers to a POSS rigid core through reductively degradable disulfide bonds. The hydrophobic docetaxel (DTX) was attached to the grafts by pH-sensitive hydrazone bonds and also encapsulated into the POSS core (SP-DTX). Thus, the final amphiphilic star-shaped conjugates could self-assemble into nanoparticles and exhibited conspicuous drug-loading capacity (20.1 wt %) based on the covalently conjugated accompanied by physically encapsulated DTX. The stimuli-responsive DTX release under acidic lysosomal and reducing cytoplasmic environments was verified, leading to enhanced cytotoxicity against PC-3 human prostate carcinoma cells. To evaluate the in vivo therapeutic effects of the DTX-loaded nanovehicles objectively, a stroma-rich, prostate xenograft tumor model was generated. SP-DTX displayed uniform tumor distribution and suppressed tumor growth to a more pronounced level (tumor inhibition of 78.9%) than nonredox-sensitive SP-DTX-A (67.4%), SP-DTX-C contained DTX only in the core (65.5%) or linear P-DTX (60.7%) through enhanced depletion of cancer-associated fibroblasts and induction of apoptosis. The hybrid POSS-based polymeric nanoparticles offer an efficient approach to transport hydrophobic drugs for cancer therapy.
Collapse
|
|
9 |
36 |
24
|
Zhou Y, Wang S, Ying X, Wang Y, Geng P, Deng A, Yu Z. Doxorubicin-loaded redox-responsive micelles based on dextran and indomethacin for resistant breast cancer. Int J Nanomedicine 2017; 12:6153-6168. [PMID: 28883726 PMCID: PMC5574666 DOI: 10.2147/ijn.s141229] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Multidrug resistance (MDR) against chemotherapeutic agents has become one of the major obstacles to successful cancer therapy and MDR-associated proteins (MRPs)-mediated drug efflux is the key factor for MDR. In this study, a redox-responsive polymer based on dextran (DEX) and indomethacin (IND), which could reduce MRPs-mediated efflux of chemotherapeutics, was synthesized, and the obtained polymer could spontaneously form stable micelles with well-defined core-shell structure and a uniform size distribution with an average diameter of 50 nm and effectively encapsulate doxorubicin (DOX); the micelles contain a disulfide bridge (cystamine, SS) between IND and DEX (DEX-SS-IND). In vitro drug release results indicated that DEX-SS-IND/DOX micelles could maintain good stability in a stimulated normal physiological environment and promptly depolymerized and released DOX in a reducing environment. After incubating DEX-SS-IND/DOX micelles with drug-resistant tumor (MCF-7/ADR) cells, the intracellular accumulation and retention of DOX were significantly increased under the synergistic effects of redox-responsive delivery and the inhibitory effect of IND on MRPs. In vitro cytotoxicity showed that DEX-SS-IND/DOX micelles exhibited higher cytotoxicity against MCF-7/ADR cells. Moreover, DEX-SS-IND/DOX micelles showed significantly enhanced inhibition of tumor in BALB/c nude mice bearing MCF-7/ADR tumors and reduced systemic toxicity. Overall, the cumulative evidence indicates that DEX-SS-IND/DOX micelles hold significant promise for overcoming MDR for cancer therapy.
Collapse
|
research-article |
8 |
36 |
25
|
Tan S, Wang G. Redox-responsive and pH-sensitive nanoparticles enhanced stability and anticancer ability of erlotinib to treat lung cancer in vivo. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:3519-3529. [PMID: 29263650 PMCID: PMC5726363 DOI: 10.2147/dddt.s151422] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Purpose Erlotinib (ETB) is a well-established therapeutic for non-small-cell lung cancer (NSCLC). To overcome drug resistance and severe toxicities in the clinical application, redox-responsive and pH-sensitive nanoparticle drug delivery systems were designed for the encapsulation of ETB. Methods Poly(acrylic acid)-cystamine-oleic acid (PAA-ss-OA) was synthesized. PAA-ss-OA-modified ETB-loaded lipid nanoparticles (PAA-ETB-NPs) were prepared using the emulsification and solvent evaporation method. The tumor inhibition efficacy of PAA-ETB-NPs was compared with that of ETB-loaded lipid nanoparticles (ETB-NPs) and free ETB anticancer drugs in tumor-bearing mice. Results PAA-ETB-NPs had a size of 170 nm, with a zeta potential of −32 mV. The encapsulation efficiency and drug loading capacity of PAA-ETB-NPs were over 85% and 2.6%, respectively. In vitro cytotoxicity of ETB-NPs were higher than that of ETB solution. The cytotoxicity of PAA-ETB-NPs was the highest. The in vivo tumor growth inhibition by PAA-ETB-NP treatment was significantly higher than that by ETB-NPs and ETB solution. No obvious weight loss was observed in any of the treatment groups, indicating that all the treatments were well tolerated. Conclusion PAA-ETB-NPs could enhance the stability and anti-cancer ability of ETB to treat lung cancer and are a promising drug delivery system for lung cancer treatment.
Collapse
|
Journal Article |
8 |
36 |