Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Eur J Pharm Biopharm ; : 114411, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39009192

ABSTRACT

Combination therapy using chemo-photothermal therapy (chemo-PTT) shows great efficacy toward tumor ablation in preclinical studies. Besides, lipopolymersomes as a hybrid nanocarriers, integrate advantages of liposomes and polymersomes in a single platform in order to provide tremendous biocompatibility, biodegradability, noteworthy loading efficacy for both hydrophobic and hydrophilic drugs with adjustable drug release and high stability. In this study, a multipurpose lipopolymersome was fabricated for guided chemotherapy-PTT and CT-scan imaging of melanoma. A lipopolymerosomal hybrid nanovesicle consisting of equal molar ratio of 1,2-dioleoyl-3-trimethylammonium propane (DOTAP) and poly (ethylene glycol)-poly (lactic acid) (PEG-PLA) diblock copolymer (molar ratio 1:1) was fabricated. The nanoparticulate system was prepared through film rehydration technique for encapsulation of doxorubicin (DOX) and indocyanine green (ICG) to form DOX-ICG-LP platform. At the next stage, AS1411 DNA aptamer was conjugated to the surface of lipopolymersome (Apt-DOX-ICG-LP) for selective delivery. The sizes of DOX-ICG-LP and Apt-DOX-ICG-LP were obtained through DLS analysis (61.0 ±â€¯6 and 74 ±â€¯5, respectively). Near Infrared-responsive release pattern of the prepared lipopolymersome was verified in vitro. The formulated platform showed efficient photothermal conversion, and superior stability with acceptable encapsulation efficiency. Consistent with the in vitro studies, NIR-responsive lipopolymersome exhibited significantly higher cellular toxicity for Chemo-PTT versus single anti-cancer treatment. Moreover, superlative tumor shrinkage with favorable survival profile were attained in B16F10 tumor-bearing mice received Apt-DOX-ICG-LP and irradiated with 808 nm laser compared to those treated with either DOX-ICG-LP or Apt-DOX-ICG-LP without laser irradiation. The diagnostic capability of Apt-DOX-ICG-LP was addressed using in vivo NIR imaging, 6 and 24 h post-intravenous administration. The results indicated desirable feature of an established targeted theranostic capability of Apt-DOX-ICG-LP for both diagnostics and dual chemo-PTT of melanoma.

2.
Eur J Pharm Biopharm ; 198: 114259, 2024 May.
Article in English | MEDLINE | ID: mdl-38479563

ABSTRACT

Liquid crystalline nanoparticles (LCNPs) have gained much attention in cancer nanomedicines due to their unique features such as high surface area, storage stability, and sustained-release profile. In the current study, a novel LCNP for co-encapsulation of Bi2O3 and hydrophilic doxorubicin (DOX) was fabricated and functionalized with folic acid (FA) to achieve efficient tumor targeting toward CT-scan imaging and chemotherapy of melanoma in vitro and in vivo. LCNPs Bi2O3 NPs were prepared using glycerol monooleate-pluronic F-127 (GMO/PF127/water). Firstly, GMO/water were homogenized to prepare LC gel. Then, the stabilizer aqueous solution (PF127/Bi2O3/DOX) was added to the prepared LC gel and homogenized using homogenization and ultrasonication. The formulated NPs exhibited superior stability with encapsulation efficiency. High cytotoxicity and cellular internalization of the FA-Bi2O3-DOX-NPs were observed in comparison with Bi2O3-DOX-NPs and the free DOX in folate-receptor (FR) overexpressing cells (B16F10) in vitro. Moreover, ideal tumor suppression with increased survival rate were observed in tumorized mice treated with FA-Bi2O3-DOX-NPs compared to those treated with non-targeted one. On the other hand, the CT-imaging ability of the Bi2O3-DOX-NPs was tested inB16F10 tumor-bearing mice. The obtained data indicated a high potential of the developed targeted theranostic FA-Bi2O3-DOX-NPs for diagnostics and treatment of melanoma.


Subject(s)
Bismuth , Melanoma , Nanoparticles , Animals , Mice , Drug Delivery Systems/methods , Precision Medicine , Folic Acid/chemistry , Doxorubicin , Nanoparticles/chemistry , Water , Cell Line, Tumor
3.
Int J Biol Macromol ; 248: 125882, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37473882

ABSTRACT

In this study, a targeted pH-sensitive polymersome incorporating doxorubicin (DOX) was manufactured implementing diblock copolymer of hyaluronic acid-b-pPoly (ß-amino ester) (HA-PBAE). The hydrophilic DOX was loaded into the aqueous compartment of HA-PBAE polymersomal structure during nanoprecipitation process with 60 % ± 3.0 entrapment efficiency (EE%) and 5.3 % ± 0.2 loading content (LC%) while demonstrating spherical morphology with size of 196 ± 3.8 nm and PDI of 0.3. The prepared platform (DOX-HA-PBAE) illustrated accelerated DOX release in acidic pH 5.4, and showed significantly higher cytotoxicity and cellular internalization in comparison with free DOX against 4T1 cell line (CD44 positive cell). In contrast, no significant growth inhibition was observed in CHO cell line (CD44 negative cell). Furthermore, DOX-HA-PBAE platform displayed higher therapeutic efficacy, favorable tumor accumulation and lower systemic toxicity in comparison with free DOX based on obtained experimental data in ectopic 4T1 tumor model in BALB/c Female mice in terms of tumor growth rate, survival rate, body weight loss, ex vivo biodistribution and pathological evaluations. The obtained results demonstrated that DOX-HA-PBAE polymersomes have potential to be used in metastatic breast cancer therapy with promising characteristics in terms of tumor growth suppression and safety profile.


Subject(s)
Nanoparticles , Neoplasms , Animals , Mice , Cricetinae , Female , Hyaluronic Acid/chemistry , Tissue Distribution , Doxorubicin , Neoplasms/drug therapy , CHO Cells , Hydrogen-Ion Concentration , Drug Delivery Systems/methods , Cell Line, Tumor , Nanoparticles/chemistry , Mice, Inbred BALB C
4.
Eur J Pharm Biopharm ; 187: 76-86, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37100090

ABSTRACT

Mesenchymal stem cell membrane (MSCM)-coated biomimetic doxorubicin-loaded hollow gold nanoparticles were fabricated and decorated with MUC1 aptamer in order to provide smart theranostic platform. The prepared targeted nanoscale biomimetic platform was extensively characterized and evaluated in terms of selective delivery of DOX and CT-scan imaging. The fabricated system illustrated spherical morphology with 118 nm in diameter. Doxorubicin was loaded into the hollow gold nanoparticles through physical absorption technique with encapsulation efficiency and loading content of 77%±10 and 31%±4, respectively. The in vitro release profile demonstrated that the designed platform could respond to acidic environment, pH 5.5 and release 50% of the encapsulated doxorubicin during 48 h, while 14% of the encapsulated doxorubicin was released in physiological condition, pH 7.4 up to 48 h. The in vitro cytotoxicity experiments on 4T1 as MUC1 positive cell line illustrated that the targeted formulation could significantly increase mortality at 0.468 and 0.23 µg/ml of equivalent DOX concentration compared to non-targeted formulation while this cytotoxicity was not observed in CHO as MUC1 negative cell line. Furthermore, in vivo experiments showed high tumor accumulation of the targeted formulation even 24 h after intravenous injection which induced effective tumor growth suppression against 4T1 tumor bearing mice. On the other hand, existence of hollow gold in this platform provided CT scan imaging capability of the tumor tissue in 4T1 tumor bearing mice up to 24 h post-administration. The obtained results indicated that the designed paradigm are promising and safe theranostic system for fighting against metastatic breast cancer.


Subject(s)
Mesenchymal Stem Cells , Metal Nanoparticles , Nanoparticles , Neoplasms , Animals , Mice , Gold/chemistry , Precision Medicine , Cell Line, Tumor , Nanoparticles/chemistry , Doxorubicin , Oligonucleotides , Theranostic Nanomedicine/methods , Drug Delivery Systems
5.
J Drug Target ; 30(10): 1106-1112, 2022 12.
Article in English | MEDLINE | ID: mdl-35736221

ABSTRACT

Herein, we presented a novel DOX-loaded multi-storey DNA nanostructure, including AS1411 aptamer as a targeting agent for treatment of target cells (MCF-7 and 4T1). Gel retardation test and fluorometric analysis were used to examine the construction of DNA nanostructure and loading of DOX in the complex. At pH 5.5 and 7.4, the release patterns of DOX from the prepared formulation were studied. Cell viability test was conducted to analyse the cell cytotoxicity ability of the DOX loaded multi-storey DNA nanostructure compared to free DOX in 4T1, MCF-7 (target) and CHO cells (non-target). Flow cytometry analysis was used to examine the DOX-loaded DNA nanostructure internalisation. Finally, the developed DOX-loaded multi-storey DNA nanostructure was tested in vivo to see if it could prevent tumour growth. The drug was released from the nanocomplex in a pH-related process (higher release in acidic pH compared to neutral pH). According to MTT assay, DOX-loaded DNA nanostructure damaged nucleolin positive cells while not significantly affecting nucleolin negative cells. The formulation was efficaciously internalised into target cells (4T1 and MCF-7), but not into non-target ones. Moreover, DOX-loaded DNA nanostructure can restrict tumour growth, increase survival rate, and accumulate significantly more in the tumour site than free DOX.


Subject(s)
Aptamers, Nucleotide , Breast Neoplasms , Nanostructures , Cricetinae , Animals , Humans , Female , Cricetulus , Breast Neoplasms/drug therapy , Doxorubicin/chemistry , Aptamers, Nucleotide/chemistry , Nanostructures/chemistry , DNA/chemistry , Cell Line, Tumor , Drug Delivery Systems , MCF-7 Cells
6.
J Control Release ; 339: 164-194, 2021 11 10.
Article in English | MEDLINE | ID: mdl-34592384

ABSTRACT

Targeted therapy and early accurate detection of malignant lesions are essential for the effectiveness of treatment and prognosis in cancer patients. The development of gaseous system as a versatile platform for the fabricated nanobubbles, has attracted much interest in improving the efficacy of ultrasound therapeutic, diagnostic, and theranostic platforms. Nano-sized bubble, as an ultrasound contrast agent, with spherical gas-filled structures exhibited contrast enhancement capability due to their inherent EPR effect. Additionally, nanobubbles exhibited good stability with extended retention time in the blood stream. The current review summarized various nanobubbles and discussed about the crucial parameters affecting the stability of ultrafine bubbles. Furthermore, therapeutic and theranostic gaseous systems for fighting against cancer were described.


Subject(s)
Nanomedicine , Neoplasms , Contrast Media , Humans , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Precision Medicine , Ultrasonography
7.
Expert Opin Drug Deliv ; 18(9): 1309-1322, 2021 09.
Article in English | MEDLINE | ID: mdl-33970721

ABSTRACT

BACKGROUND: Hybrid vesicular systems (lipopolymersomes) are promising platforms for minimizing the liposomes and polymersomes disadvantages in terms of chemotherapeutic transportation. In this regard, lipopolymersome has been designed to integrate the advantage of both polymersomes and liposomes to enable better structural integrity of the bilayer after encapsulation of hydrophobic drugs while maintaining the soft nature of liposomes, superior serum stability, and high encapsulation efficiency of cargos in the bilayer segment. RESEARCH DESIGN AND METHODS: In the present study, we reported preparation and characterization of five camptothecin (CPT)-loaded lipopolymersomal formulations composed of poly (ethylene glycol)-poly (lactic acid) (PEG-PLA) and dipalmitoylphosphatidylcholine (DPPC) at different molar ratios using film rehydration method. Afterward, the preferred formulation was tagged with AS1411 DNA aptamer in order to evaluate the therapeutic index using nucleolin-positive colon cancer cell lines (HT29 and C26). RESULTS: The obtained data indicated that the prepared CPT-loaded lipopolymersome at a PEG-PLA: DPPC ratio of 75:25 exhibited superior stability and high loading capacity compared to other systems. Moreover, high cytotoxicity of the aptamer-targeted lipopolymersome and increased tumor accumulation were observed in comparison with non-targeted one. CONCLUSIONS: The designed polymer-rich lipopolymersomal platform offers bright future for the development of potent nanomedicine against cancer.


Subject(s)
Adenocarcinoma , Colonic Neoplasms , Adenocarcinoma/drug therapy , Camptothecin/pharmacology , Colonic Neoplasms/drug therapy , Drug Delivery Systems , Humans
8.
Int J Pharm ; 591: 120030, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-33137452

ABSTRACT

Despite the noticeable advantages of the liposomes and polymersomes, they also revealed some drawbacks that could be minimized by preparing hybrid vesicular systems and integrating the advantage of both vehicles into one system named lipopolymersome. Lipopolymesome incorporates the biodegradability, stability, adjustability and chemical flexibility of polymersomes with the elasticity, soft nature and biocompatibility of liposomes. In the current study, wereported the development of five nanoscale lipopolymersomal hybrid vesicular systems consisting different molar ratios of dipalmitoylphosphatidylcholine (DPPC) and poly (ethylene glycol)-poly (lactic acid) (PEG-PLA) (PEG-PLA: DPPC ratio of 100:0, 50:50: 25:75, 75:25 and 0:100). Rhod-6G-loaded hybrid vesicles were prepared via film rehydration. Then, the efficacy of five formulations were evaluated in terms of loading capacity, release pattern, cellular uptake, andin vivobiodistribution in ectopic tumor model in mice. The obtained results demonstrated that the self-assembly, loading capacity, cargo release and stability of hybrid nanoscale lipopolymersomes can be tuned by incorporation of amphiphilic lipid-polymers at various ratios. In this regard, the prepared hybrid nanovesicles consisting of DPPC-PEG-PLA (25:75) exhibited great potential through superior loading capacity, stability and tumor accumulation compared with other systems. It could be concluded that the prepared lipopolymersome offers important opportunities for the development of novel hybrid carriers for efficient transportation of therapeutics into tumor site.


Subject(s)
Neoplasms , Phospholipids , Animals , Drug Carriers , Drug Delivery Systems , Liposomes , Mice , Polyethylene Glycols , Polymers
9.
Colloids Surf B Biointerfaces ; 193: 111135, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32447200

ABSTRACT

In the current study, polyethylene glycol (PEG) was linked to polylactide (PLA) through the synthetic peptide PVGLIG which can be selectively cleaved by the tumor-associated matrix metalloproteinase 2 (MMP-2) enzyme. The synthesized chimeric triblock polymer of PEG-b-PVGLIG-PLA was implemented to form nanoscale self-assemble chimeric polymersomes. The hydrophobic SN38 was loaded into polymersomes with 70.3% ± 3.0% encapsulation efficiency demonstrating monodispersed spherical morphologies with 172 ±â€¯30 nm dimension. The prepared chimeric polymersomal formulation provided controlled release of SN38 at physiological condition while illustrating seven-folds higher release rate when exposed to MMP-2 enzyme. At the next stage, AS1411 aptamer was conjugated onto the surface of MMP-2 responsive polymersomal formulation in order to provide guided drug delivery against nucleolin positive cells. In vitro cellular toxicity assay against C26 cell line (nucleolin positive) demonstrated significantly higher toxicity of targeted formulation in comparison with non-targeted one in low SN38 concentrations (0.15-1.25 µg/mL). In vivo study in mice bearing subcutaneous C26 tumor showed higher therapeutic index for MMP-2 responsive chimeric polymersomal formulation of SN38 in comparison with non-responsive one. On the other hand, AS1411 aptamer-targeted MMP-2 responsive chimeric polymersomal formulation exhibited highest therapeutic index compared to other groups. It could be concluded that the targeted chimeric polymersomes bearing both cleavable peptide sequence between their blocks and targeting ligand on their surface, provide favorable characteristics as an ideal delivery system against cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Peptides/pharmacology , Polyesters/pharmacology , Polyethylene Glycols/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , CHO Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cricetulus , Drug Screening Assays, Antitumor , Female , Matrix Metalloproteinase Inhibitors/chemical synthesis , Matrix Metalloproteinase Inhibitors/chemistry , Mice , Mice, Inbred BALB C , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Particle Size , Peptides/chemical synthesis , Peptides/chemistry , Polyesters/chemistry , Polyethylene Glycols/chemistry , Surface Properties
10.
J Cell Physiol ; 235(2): 1036-1050, 2020 02.
Article in English | MEDLINE | ID: mdl-31276199

ABSTRACT

In this study, the chemical features of dendritic mesoporous silica nanoparticles (DMSNs) provided the opportunity to design a nanostructure with the capability to intelligently transport the payload to the tumor cells. In this regard, doxorubicin (DOX)-encapsulated DMSNs was electrostatically surface-coated with polycarboxylic acid dextran (PCAD) to provide biocompatible dextran-capped DMSNs (PCAD-DMSN@DOX) with controlled pH-dependent drug release. Moreover, a RNA aptamer against a cancer stem cell (CSC) marker, CD133 was covalently attached to the carboxyl groups of DEX to produce a CD133-PCAD-DMSN@DOX. Then, the fabricated nanosystem was utilized to efficiently deliver DOX to CD133+ colorectal cancer cells (HT29). The in vitro evaluation in terms of cellular uptake and cytotoxicity demonstrated that the CD133-PCAD-DMSN@DOX specifically targets HT29 as a CD133 overexpressed cancer cells confirmed by flow cytometry and 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide assay. The potentially promising intelligent-targeted platform suggests that targeted dextran-capped DMSNs may find impressive application in cancer therapy.


Subject(s)
Dextrans/chemistry , Drug Delivery Systems , Nanoparticles/chemistry , Silicon Dioxide/chemistry , AC133 Antigen , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacology , CHO Cells , Cell Survival/drug effects , Cricetinae , Cricetulus , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Doxorubicin/pharmacology , HT29 Cells , Humans
11.
J Control Release ; 308: 172-189, 2019 08 28.
Article in English | MEDLINE | ID: mdl-31295542

ABSTRACT

Recent technological approaches in drug delivery have attracted scientist interest for improving therapeutic index of medicines and drug compliance. One of the powerful strategies to control the transportation of drugs is implementation of intelligent stimuli-responsive drug delivery system (DDS). In this regard, tumor tissues with unique characteristics including leaky vasculature and diverse enzyme expression profiles facilitate the development of efficient enzyme-responsive nanoscale delivery systems. Based on the stimuli nature (physical, chemical and biological), these systems can be categorized into three groups according to the nature of trigger initiating the drug release. Enzymes are substantial constituents of the biotechnology toolbox offering promising capabilities and ideal characteristics to accelerate chemical reactions. Nanoparticles which have the ability to trigger their cargo release in the presence of specific enzymes are fabricated implementing fascinating physico-chemical properties of different materials in a nanoscale dimension. In order to reduce the adverse effects of the therapeutic agents, nanocarriers can be utilized and modified with enzyme-labile linkages to provide on-demand enzyme-responsive drug release. In the current review, we give an overview of drug delivery systems which can deliver drugs to the tumor microenvironment and initiate the drug release in response to specific enzymes highly expressed in particular tumor tissues. This strategy offers a versatile platform for intelligent drug release at the site of action.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems , Enzymes/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Drug Liberation , Humans , Nanoparticles , Neoplasms/drug therapy , Neoplasms/enzymology , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL