Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.115
Filter
1.
ACS Nano ; 18(20): 12716-12736, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38718220

ABSTRACT

Mesoporous silica nanoparticles (MSNs) represent a promising avenue for targeted brain tumor therapy. However, the blood-brain barrier (BBB) often presents a formidable obstacle to efficient drug delivery. This study introduces a ligand-free PEGylated MSN variant (RMSN25-PEG-TA) with a 25 nm size and a slight positive charge, which exhibits superior BBB penetration. Utilizing two-photon imaging, RMSN25-PEG-TA particles remained in circulation for over 24 h, indicating significant traversal beyond the cerebrovascular realm. Importantly, DOX@RMSN25-PEG-TA, our MSN loaded with doxorubicin (DOX), harnessed the enhanced permeability and retention (EPR) effect to achieve a 6-fold increase in brain accumulation compared to free DOX. In vivo evaluations confirmed the potent inhibition of orthotopic glioma growth by DOX@RMSN25-PEG-TA, extending survival rates in spontaneous brain tumor models by over 28% and offering an improved biosafety profile. Advanced LC-MS/MS investigations unveiled a distinctive protein corona surrounding RMSN25-PEG-TA, suggesting proteins such as apolipoprotein E and albumin could play pivotal roles in enabling its BBB penetration. Our results underscore the potential of ligand-free MSNs in treating brain tumors, which supports the development of future drug-nanoparticle design paradigms.


Subject(s)
Blood-Brain Barrier , Doxorubicin , Drug Delivery Systems , Nanoparticles , Silicon Dioxide , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Silicon Dioxide/chemistry , Doxorubicin/pharmacology , Doxorubicin/chemistry , Nanoparticles/chemistry , Animals , Porosity , Mice , Humans , Polyethylene Glycols/chemistry , Drug Carriers/chemistry , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Particle Size , Cell Line, Tumor , Glioma/drug therapy , Glioma/metabolism , Glioma/pathology , Ligands , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/administration & dosage
2.
Int J Pharm ; 658: 124231, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38759741

ABSTRACT

Two frequent problems hindering clinical translation of nanomedicine are low drug loading and low colloidal stability. Previous efforts to achieve ultrahigh drug loading (>30 %) introduce new hurdles, including lower colloidal stability and others, for clinical translation. Herein, we report a new class of drug nano-carriers based on our recent finding in protein-nanoparticle co-assembly supraparticle (PNCAS), with both ultrahigh drug loading (58 % for doxorubicin, i.e., DOX) and ultrahigh colloidal stability (no significant change in hydrodynamic size after one year). We further show that our PNCAS-based drug nano-carrier possesses a built-in environment-responsive drug release feature: once in lysosomes, the loaded drug molecules are released instantly (<1 min) and completely (∼100 %). Our PNCAS-based drug delivery system is spontaneously formed by simple mixing of hydrophobic nanoparticles, albumin and drugs. Several issues related to industrial production are studied. The ultrahigh drug loading and stability of DOX-loaded PNCAS enabled the delivery of an exceptionally high dose of DOX into a mouse model of breast cancer, yielding high efficacy and no observed toxicity. With further developments, our PNCAS-based delivery systems could serve as a platform technology to meet the multiple requirements of clinical translation of nanomedicines.


Subject(s)
Doxorubicin , Drug Liberation , Lysosomes , Nanoparticles , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Animals , Nanoparticles/chemistry , Female , Drug Carriers/chemistry , Mice , Colloids/chemistry , Humans , Drug Delivery Systems , Mice, Inbred BALB C , Drug Stability , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacokinetics , Cell Line, Tumor , Breast Neoplasms/drug therapy
3.
Mol Pharm ; 21(6): 2970-2980, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38742943

ABSTRACT

One of the most significant reasons hindering the clinical translation of nanomedicines is the rapid clearance of intravenously injected nanoparticles by the mononuclear phagocyte system, particularly by Kupffer cells in the liver, leading to an inefficient delivery of nanomedicines for tumor treatment. The threshold theory suggests that the liver's capacity to clear nanoparticles is limited, and a single high dose of nanoparticles can reduce the hepatic clearance efficiency, allowing more nanomedicines to reach tumor tissues and enhance therapeutic efficacy. Building upon this theory, researchers have conducted numerous validation studies based on the same nanoparticle carrier systems. These studies involve the use of albumin nanoparticles to improve the therapeutic efficacy of albumin nanomedicines as well as polyethylene glycol (PEG)-modified liposomal nanoparticles to enhance the efficacy of PEGylated liposomal nanomedicines. However, there is no research indicating the feasibility of the threshold theory when blank nanoparticles and nanomedicine belong to different nanoparticle carrier systems currently. In this study, we prepared two different sizes of albumin nanoparticles by using bovine serum albumin. We used the marketed nanomedicine liposomal doxorubicin hydrochloride injection (trade name: LIBOD, manufacturer: Shanghai Fudan-zhangjiang Biopharmaceutical Co., Ltd.), as the representative nanomedicine. Through in vivo experiments, we found that using threshold doses of albumin nanoparticles still can reduce the clearance rate of LIBOD, prolong its time in vivo, increase the area under the plasma concentration-time curve (AUC), and also lead to an increased accumulation of the drug at the tumor site. Furthermore, evaluation of in vivo efficacy and safety further indicates that threshold doses of 100 nm albumin nanoparticles can enhance the antitumor effect of LIBOD without causing harm to the animals. During the study, we found that the particle size of albumin nanoparticles influenced the in vivo distribution of the nanomedicine at the same threshold dose. Compared with 200 nm albumin nanoparticles, 100 nm albumin nanoparticles more effectively reduce the clearance efficiency of LIBOD and enhance nanomedicine accumulation at the tumor site, warranting further investigation. This study utilized albumin nanoparticles to reduce hepatic clearance efficiency and enhance the delivery efficiency of nonalbumin nanocarrier liposomal nanomedicine, providing a new avenue to improve the efficacy and clinical translation of nanomedicines with different carrier systems.


Subject(s)
Doxorubicin , Nanoparticles , Polyethylene Glycols , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Doxorubicin/chemistry , Doxorubicin/pharmacology , Doxorubicin/analogs & derivatives , Animals , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Mice , Liposomes/chemistry , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/administration & dosage , Tissue Distribution , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacology , Mice, Inbred BALB C , Liver/drug effects , Liver/metabolism , Particle Size , Nanomedicine/methods , Humans , Male , Female
4.
Int J Pharm ; 657: 124183, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38692500

ABSTRACT

We developed cyclic RGD-tagged polymeric micellar nanoassemblies for sustained delivery of Doxorubicin (Dox) endowed with significant cytotoxic effect against MG63, SAOS-2, and U2-OS osteosarcoma cells without compromising the viability of healthy osteoblasts (hFOBs). Targeted polymeric micellar nanoassemblies (RGD-NanoStar@Dox) enabled Dox to reach the nucleus of MG63, SAOS-2, and U2-OS cells causing the same cytotoxic effect as free Dox, unlike untargeted micellar nanoassemblies (NanoStar@Dox) which failed to reach the nucleus and resulted ineffective, demonstrating the crucial role of cyclic RGD peptide in driving cellular uptake and accumulation mechanisms in osteosarcoma cells. Micellar nanoassemblies were obtained by nanoformulation of three-armed star PLA-PEG copolymers properly synthetized with and without decoration with the cyclic-RGDyK peptide (Arg-Gly-Asp-D-Tyr-Lys). The optimal RGD-NanoStar@Dox nanoformulation obtained by nanoprecipitation method (8 % drug loading; 35 % encapsulation efficiency) provided a prolonged and sustained drug release with a rate significantly lower than the free drug under the same experimental conditions. Moreover, the nanosystem preserved Dox from the natural degradation occurring under physiological conditions (i.e., dimerization and consequent precipitation) serving as a slow-release "drug reservoir" ensuring an extended biological activity over the time.


Subject(s)
Bone Neoplasms , Cell Survival , Doxorubicin , Micelles , Oligopeptides , Osteosarcoma , Polyethylene Glycols , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Doxorubicin/chemistry , Osteosarcoma/drug therapy , Humans , Polyethylene Glycols/chemistry , Cell Line, Tumor , Oligopeptides/chemistry , Oligopeptides/administration & dosage , Bone Neoplasms/drug therapy , Cell Survival/drug effects , Nanoparticles/chemistry , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/chemistry , Drug Liberation , Drug Carriers/chemistry
5.
Sci Rep ; 14(1): 11988, 2024 05 25.
Article in English | MEDLINE | ID: mdl-38796555

ABSTRACT

This study employs a combination of mathematical derivation and optimization technique to investigate the adsorption of drug molecules on nanocarriers. Specifically, the chemotherapy drugs, fluorouracil, proflavine, and methylene blue, are non-covalently bonded with either a flat graphene sheet or a spherical C 60 fullerene. Mathematical expressions for the interaction energy between an atom and graphene, as well as between an atom and C 60 fullerene, are derived. Subsequently, a discrete summation is evaluated for all atoms on the drug molecule utilizing the U-NSGA-III algorithm. The stable configurations' three-dimensional architectures are presented, accompanied by numerical values for crucial parameters. The results indicate that the nanocarrier's structure effectively accommodates the atoms on the drug's carbon planes. The three drug types' molecules disperse across the graphene surface, whereas only fluorouracil spreads on the C 60 surface; proflavine and methylene blue stack vertically to form a layer. Furthermore, all atomic positions of equilibrium configurations for all systems are obtained. This hybrid method, integrating analytical expressions and an optimization process, significantly reduces computational time, representing an initial step in studying the binding of drug molecules on nanocarriers.


Subject(s)
Drug Carriers , Fluorouracil , Graphite , Methylene Blue , Adsorption , Graphite/chemistry , Methylene Blue/chemistry , Fluorouracil/chemistry , Drug Carriers/chemistry , Models, Theoretical , Algorithms , Fullerenes/chemistry , Carbon/chemistry , Proflavine/chemistry , Nanoparticles/chemistry , Antibiotics, Antineoplastic/chemistry , Antineoplastic Agents/chemistry
6.
Anal Chem ; 96(22): 8905-8913, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38771097

ABSTRACT

Chemotherapeutic anthracyclines, like doxorubicin (DOX), are drugs endowed with cytostatic activity and are widely used in antitumor therapy. Their molecular mechanism of action involves the formation of a stable anthracycline-DNA complex, which prevents cell division and results in cell death. It is known that elevated DOX concentrations induce DNA chain loops and overlaps. Here, for the first time, tip-enhanced Raman scattering was used to identify and localize intercalated DOX in isolated double-stranded calf thymus DNA, and the correlated near-field spectroscopic and morphologic experiments locate the DOX molecules in the DNA and provide further information regarding specific DOX-nucleobase interactions. Thus, the study provides a tool specifically for identifying intercalation markers and generally analyzing drug-DNA interactions. The structure of such complexes down to the molecular level provides mechanistic information about cytotoxicity and the development of potential anticancer drugs.


Subject(s)
DNA , Doxorubicin , Spectrum Analysis, Raman , Doxorubicin/pharmacology , Doxorubicin/chemistry , DNA/chemistry , Animals , Cattle , Intercalating Agents/chemistry , Intercalating Agents/pharmacology , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/chemistry
7.
Colloids Surf B Biointerfaces ; 239: 113937, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38749166

ABSTRACT

Osteosarcoma conventional chemotherapeutics are known for their side effects, limited options, and induction of drug resistance. This creates the need to develop new therapeutics capable of effectively destroying cancer cells with low toxicity, improving patient survival rate and their life quality. This work reports a novel drug delivery nanoplataform made of Natural Melanin Nanoparticles (MNPs), obtained from Sepia officinalis ink, with 99% incorporation efficiency of doxorubicin (Dox) without the use of non-toxic solvents. A significant photothermal effect was shown by a 36ºC increment after 10 min of laser irradiation, surpassing reported values for synthetic melanin. A sustained drug release of ca. 23% with photothermal stimuli was observed, compared to 15% without stimuli, after 48 h. This nanoplatform is obtained as a food industry side product, which makes it a natural cost-effective biomedical material. Natural MPs were applied in an osteosarcoma cell line (SaOs-2), and internalized by the cells in less than 2 h, showing cytocompatibility up to 1000 µg/mL after 72 h of contact with cells. On the contrary, when natural MNPs loaded with Dox (Dox-MNPs) were placed in contact with the SaOs-2 cells and were simultaneously receiving NIR light it was observed a 93% reduction in cancer cells in 48 h, revealing a synergistic effect between chemotherapy and phototherapy. To our knowledge this is the first time that natural MNPs extracted from Sepia officinalis were tested on an osteosarcoma cell line as chemo-photothermal agent, showing these NPs are an effective, cost-effective, reproducible, non-toxic nanoplatform for osteosarcoma treatment using combined effects.


Subject(s)
Cell Survival , Doxorubicin , Melanins , Nanoparticles , Osteosarcoma , Sepia , Humans , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Doxorubicin/pharmacology , Doxorubicin/chemistry , Melanins/metabolism , Nanoparticles/chemistry , Sepia/chemistry , Cell Survival/drug effects , Cell Line, Tumor , Drug Liberation , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/chemistry , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Particle Size , Cost-Benefit Analysis , Drug Screening Assays, Antitumor
8.
ACS Nano ; 18(22): 14441-14456, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38758604

ABSTRACT

The active targeting drug delivery system based on special types of endogenous cells such as macrophages has emerged as a promising strategy for tumor therapy, owing to its tumor homing property and biocompatibility. In this work, the active tumor-targeting drug delivery system carrying doxorubicin-loaded nanoparticles (DOX@MPF127-MCP-1, DMPM) on macrophage (RAW264.7) surfaces via the mediation of interaction with the CCR2/MCP-1 axis was exploited. Initially, the amphiphilic block copolymer Pluronic F127 (PF127) was carboxylated to MPF127 at the hydroxyl terminus. Subsequently, MPF127 was modified with MCP-1 peptide to prepare MPF127-MCP-1 (MPM). The DOX was wrapped in MPM to form DMPM nanomicelles (approximately 100 nm) during the self-assembly process of MPM. The DMPM spontaneously bound to macrophages (RAW264.7), which resulted in the construction of an actively targeting delivery system (macrophage-DMPM, MA-DMPM) in vitro and in vivo. The DOX in MA-DMPM was released in the acidic tumor microenvironment (TME) in a pH-responsive manner to increase DOX accumulation and enhance the tumor treatment effect. The ratio of MA-DMPM homing reached 220% in vitro compared with the control group, indicating that the MA-DMPM was excellently capable of tumor-targeting delivery. In in vivo experiments, nonsmall cell lung cancer cell (NCI-H1299) tumor models were established. The results of the fluorescence imaging system (IVIS) showed that MA-DMPM demonstrated tremendous tumor-targeting ability in vivo. The antitumor effects of MA-DMPM in vivo indicated that the proportion of tumor cell apoptosis in the DMPM-treated group was 63.33%. The findings of the tumor-bearing mouse experiment proved that MA-DMPM significantly suppressed tumor cell growth, which confirmed its immense potential and promising applications in tumor therapy.


Subject(s)
Doxorubicin , Macrophages , Nanoparticles , Poloxamer , Tumor Microenvironment , Doxorubicin/pharmacology , Doxorubicin/chemistry , Doxorubicin/administration & dosage , Animals , Tumor Microenvironment/drug effects , Mice , Poloxamer/chemistry , Nanoparticles/chemistry , Macrophages/metabolism , Macrophages/drug effects , RAW 264.7 Cells , Drug Delivery Systems , Humans , Drug Carriers/chemistry , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/administration & dosage , Mice, Inbred BALB C , Chemokine CCL2/metabolism
9.
Anal Chem ; 96(22): 9254-9261, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38778440

ABSTRACT

Targeted therapy to the tumor would greatly advance precision medicine. Many drug delivery vehicles have emerged, but liposomes are cited as the most successful to date. Recent efforts to develop liposomal drug delivery systems focus on drug distribution in tissues and ignore liposomal fate. In this study, we developed a novel method to elucidate both drug and liposomal bilayer distribution in a three-dimensional cell culture model using quantitative matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI qMSI) alongside fluorescence microscopy. Imaging liposomal distribution in a cell culture model is challenging, as lipids forming the bilayer are endogenous to the model system. To resolve this issue, we functionalized the bilayer by chemically cross-linking a fluorescent tag to the alkyne-containing lipid hexynoyl phosphoethanolamine (HPE). We synthesized liposomes incorporating the tagged HPE lipid and encapsulated within them doxorubicin, yielding a theranostic liposome capable of both drug delivery and monitoring liposomal uptake. We employed an "in-tissue" MALDI qMSI approach to generate a calibration curve with R2 = 0.9687, allowing for quantification of doxorubicin within spheroid sections at multiple time points. After 72 h of treatment with the theranostic liposomes, full doxorubicin penetration was observed. The metabolites doxorubicinone and 7-deoxydoxorubicinone were also detected after 48 h. Modification of the bilayer allowed for fluorescence microscopy tracking of liposomes, while MALDI MSI simultaneously permitted the imaging of drugs and metabolites. While we demonstrated the utility of our method with doxorubicin, this system could be applied to examine the uptake, release, and metabolism of many other liposome-encapsulated drugs.


Subject(s)
Doxorubicin , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Doxorubicin/chemistry , Doxorubicin/administration & dosage , Doxorubicin/analogs & derivatives , Humans , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Drug Delivery Systems , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/administration & dosage , Liposomes/chemistry , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Polyethylene Glycols/chemistry , Microscopy, Fluorescence , Cell Line, Tumor
10.
Sci Rep ; 14(1): 11350, 2024 05 18.
Article in English | MEDLINE | ID: mdl-38762628

ABSTRACT

A new 3D metal-organic frameworks [Cd6(L)4(bipy)3(H2O)2·H2O] (1) was gained by employing Cd(II) and organic ligand [H3L = 4,4',4''-(benzene-1,3,5-triyltris(oxy))tribenzoic acid)benzene acid; bipy = 4,4'-bipyridine] in the solvothermal condition, which has been fully examined via single-X ray diffraction, FTIR and elemental analysis and so on. Using natural polysaccharides hyaluronic acid (HA) and carboxymethyl chitosan (CMCS) as raw materials, we successfully prepared HA/CMCS hydrogels and observed their internal micromorphology by scanning electron microscopy. Using doxorubicin (Dox) as a drug model, we synthesized a novel metal gel particle loaded with doxorubicin, and their encapsulation and release effects were studied using fluorescence spectroscopy, followed by further investigation of their components through thermogravimetric analysis. Based on this, the therapeutic effect on leukemia was evaluated. Finally, an enhanced learning method for automatically designing new ligand structures from host ligands was proposed. Through generative modeling and molecular docking simulations, the biological behavior of the host and predicted cadmium complexes was extensively studied.


Subject(s)
Chitosan , Doxorubicin , Hydrogels , Leukemia , Doxorubicin/chemistry , Doxorubicin/pharmacology , Hydrogels/chemistry , Chitosan/chemistry , Chitosan/analogs & derivatives , Humans , Leukemia/drug therapy , Molecular Docking Simulation , Cadmium/chemistry , Hyaluronic Acid/chemistry , Metal-Organic Frameworks/chemistry , Drug Carriers/chemistry , Cell Line, Tumor , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacology
11.
Biomacromolecules ; 25(5): 2980-2989, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38587905

ABSTRACT

We developed a supramolecular system for codelivery of doxorubicin (Dox) and p53 gene based on a ß-CD-containing star-shaped cationic polymer. First, a star-shaped cationic polymer consisting of a ß-CD core and 3 arms of oligoethylenimine (OEI), named CD-OEI, was used to form a supramolecular inclusion complex with hydrophobic Dox. The CD-OEI/Dox complex was subsequently used to condense plasmid DNA via electrostatic interactions to form CD-OEI/Dox/DNA polyplex nanoparticles with positive surface charges that enhanced the cellular uptake of both Dox and DNA. This supramolecular drug and gene codelivery system showed high gene transfection efficiency and effective protein expression in cancer cells. The codelivery of Dox and DNA encoding the p53 gene resulted in reduced cell viability and enhanced antitumor effects at low Dox concentrations. With its enhanced cellular uptake and anticancer efficacy, the system holds promise as a delivery carrier for potential combination cancer therapies.


Subject(s)
Doxorubicin , Nanoparticles , Static Electricity , beta-Cyclodextrins , Doxorubicin/pharmacology , Doxorubicin/chemistry , Doxorubicin/administration & dosage , Humans , Nanoparticles/chemistry , beta-Cyclodextrins/chemistry , Tumor Suppressor Protein p53/genetics , DNA/chemistry , Cell Survival/drug effects , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/administration & dosage , Gene Transfer Techniques , Drug Carriers/chemistry
12.
Colloids Surf B Biointerfaces ; 238: 113892, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38581834

ABSTRACT

Receptor and ligand binding mediated targeted drug delivery systems (DDS) sometimes fail to target to tumor sites, and cancer cell membrane (CCM) coating can overcome the dilemma of immune clearance and nonspecific binding of DDS in vivo. In order to enhance the targeting ability and improve the anti-tumor effect, a dual targeting DDS was established based on U87MG CCM mediated homologous targeting and cyclic peptide RGD mediated active targeting. The DDS was prepared by coating RGD doped CCM onto doxorubicin (DOX) loaded liposomes. The homologous and active dual targeting ability endowed the DDS (RGD-CCM-LP-DOX) exhibited superior cancer cell affinity, improved tissue distribution and enhanced anti-tumor effects. In vivo pharmacodynamic studies revealed that RGD-CCM-LP-DOX exhibited superior therapeutic effect compared with homologous targeting CCM-LP-DOX and non-targetable LP-DOX injection. H&E staining, Ki 67 staining and TUNEL staining confirmed that RGD-CCM-LP-DOX not only increased anti-tumor efficacy, but also reduced tissue toxicity by changing the distribution in vivo. The experimental results showed that the RGD doped CCM camouflaged liposome DDS is a better choice for chemotherapeutics delivery.


Subject(s)
Cell Membrane , Doxorubicin , Drug Delivery Systems , Liposomes , Doxorubicin/pharmacology , Doxorubicin/chemistry , Doxorubicin/administration & dosage , Liposomes/chemistry , Animals , Humans , Mice , Cell Membrane/metabolism , Cell Membrane/drug effects , Cell Membrane/chemistry , Oligopeptides/chemistry , Mice, Inbred BALB C , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/administration & dosage , Cell Line, Tumor , Mice, Nude , Cell Proliferation/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Tissue Distribution , Drug Screening Assays, Antitumor
13.
Colloids Surf B Biointerfaces ; 238: 113909, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38599076

ABSTRACT

Nasopharyngeal carcinoma (NPC) is a common head and neck malignancy, which is characterized by high incidence and aggression with poor diagnosis and limited therapeutic opportunity. The innovative strategy for achieving precise NPC active-targeting drug delivery has emerged as a prominent focus in clinical research. Here, a minimalist cancer cell membrane (CCM) shielded biomimetic nanoparticle (NP) was designed for NPC active-targeting therapy. Chemotherapeutant model drug doxorubicin (DOX) was loaded in polyamidoamine (PAMAM) dendrimer. The PAMAM/DOX (PD) NP was further shielded by human CNE-2 NPC CCM. Characterization results verified that the biomimetic PAMAM/DOX@CCM (abbreviated as PDC) NPs had satisfactory physical properties with high DOX-loading and excellent stability. Cell experiments demonstrated that the CNE-2 membrane-cloaked PDC NPs presented powerful cellular uptake in the sourcing cells by homologous targeting and adhesive interaction. Further in vivo results confirmed that this biomimetic nanoplatform had extended circulation and remarkable tumor-targeting capability, and the PDC NPs effectively suppressed the progression of CNE-2 tumors by systemic administration. This CCM-shielded biomimetic NP displayed a minimalist paradigm nanoplatform for precise NPC therapy, and the strategy of CCM-shielded biomimetic drug delivery system (DDS) has great potential for extensive cancer active-targeting therapy.


Subject(s)
Biomimetic Materials , Cell Membrane , Doxorubicin , Nanoparticles , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Humans , Doxorubicin/pharmacology , Doxorubicin/chemistry , Nanoparticles/chemistry , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Carcinoma/pathology , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Membrane/drug effects , Animals , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/pathology , Dendrimers/chemistry , Mice , Cell Line, Tumor , Drug Delivery Systems , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/administration & dosage , Cell Proliferation/drug effects , Mice, Nude , Mice, Inbred BALB C , Biomimetics , Particle Size
14.
J Colloid Interface Sci ; 667: 1-11, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38615618

ABSTRACT

A major challenge in combining cancer immunotherapy is the efficient delivery of multiple types of immunological stimulators to elicit a robust anti-tumor immune response and reprogram the immunosuppressive tumor microenvironment (TME). Here, we developed a DNA nanodevice that was generated by precisely assembling three types of immunological stimulators. The doxorubicin (Dox) component induced immunogenic cell death (ICD) in tumor cells and enhanced phagocytosis of antigen-presenting cells (APCs). Exogenous double-stranded DNA (dsDNA) could act as a molecular adjuvant to activate the stimulator of interferon genes (STING) signaling in APCs by engulfing dying tumor cells. Interleukin (IL)-12 and small hairpin programmed cell death-ligand 1 (shPD-L1) transcription templates were designed to regulate TME. Additionally, for targeted drug delivery, multiple cyclo[Arg-Gly-Asp-(d-Phe)-Cys] (cRGD) peptide units on DNA origami were employed. The incorporation of disulfide bonds allowed the release of multiple modules in response to intracellular glutathione (GSH) in tumors. The nanodevice promoted the infiltration of CD8+ and CD4+ cells into the tumor and generated a highly inflamed TME, thereby enhancing the effectiveness of cancer immunotherapy. Our research results indicate that the nanodevice we constructed can effectively inhibit tumor growth and prevent lung metastasis without obvious systemic toxicity, providing a promising strategy for cancer combination treatment.


Subject(s)
DNA , Doxorubicin , Immunotherapy , DNA/chemistry , Doxorubicin/pharmacology , Doxorubicin/chemistry , Doxorubicin/administration & dosage , Mice , Animals , Tumor Microenvironment/drug effects , Humans , Drug Delivery Systems , Mice, Inbred C57BL , Mice, Inbred BALB C , Cell Line, Tumor , Antigen-Presenting Cells/immunology , Nanoparticles/chemistry , Neoplasms/therapy , Neoplasms/drug therapy , Neoplasms/immunology , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/administration & dosage , Particle Size
15.
Mol Pharm ; 21(5): 2394-2405, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38647653

ABSTRACT

Doxorubicin (DOX) is one of the most commonly used anticancer drugs; however, its clinical application is greatly limited due to its toxicity and chemotherapy resistance. The delivery of DOX by liposomes (Lipos) can improve the blood circulation time in vivo and reduce toxic side effects, but the drug's accumulation in the tumor is often insufficient for effective treatment. In this study, we present a calcium cross-linked liposome gel for the encapsulation of DOX, demonstrating its superior long-term release capabilities compared to conventional Lipos. By leveraging this enhanced long-term release, we can enhance drug accumulation within tumors, ultimately leading to improved antitumor efficacy. Lipos were prepared using the thin-film dispersion method in this study. We utilized the ion-responsiveness of glutathione-gelatin (GSH-GG) to form the gel outside the Lipos and named the nanoparticles coated with GSH-GG on the outside of Lipos as Lipos@GSH-GG. The average size of Lipos@GSH-GG was around 342.9 nm, with a negative charge of -25.6 mV. The in vitro experiments revealed that Lipos@GSH-GG exhibited excellent biocompatibility and slower drug release compared to conventional Lipos. Further analysis of cellular uptake and cytotoxicity demonstrated that Lipos@GSH-GG loading DOX (DOX&Lipos@GSH-GG) exhibited superior long-term release effects and lower toxic side effects compared to Lipos loading DOX (DOX&Lipos). Additionally, the findings regarding the long-term release effect in vivo and the tumor accumulation within tumor-bearing mice of Lipos@GSH-GG suggested that, compared to Lipos, it demonstrated superior long-term release capabilities and achieved greater drug accumulation within tumors. In vivo antitumor efficacy experiments showed that DOX&Lipos@GSH-GG demonstrated superior antitumor efficacy to DOX&Lipos. Our study highlights Lipos@GSH-GG as a promising nanocarrier with the potential to enhance efficacy and safety by means of long-term release effects and may offer an alternative approach for effective antitumor therapy in the future.


Subject(s)
Calcium , Doxorubicin , Drug Liberation , Glutathione , Liposomes , Doxorubicin/pharmacology , Doxorubicin/chemistry , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Animals , Mice , Liposomes/chemistry , Humans , Calcium/chemistry , Calcium/metabolism , Glutathione/chemistry , Female , Gels/chemistry , Gelatin/chemistry , Mice, Nude , Nanoparticles/chemistry , Mice, Inbred BALB C , Cell Line, Tumor , Xenograft Model Antitumor Assays , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacokinetics , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Cross-Linking Reagents/chemistry , Drug Delivery Systems/methods
16.
Nanoscale ; 16(19): 9412-9425, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38650478

ABSTRACT

Nanotechnology has the potential to provide formulations of antitumor agents with increased selectivity towards cancer tissue thereby decreasing systemic toxicity. This in vivo study evaluated the potential of novel nanoformulation based on poly(lactic-co-glycolic acid) (PLGA) to reduce the cardiotoxic potential of doxorubicin (DOX). In vivo toxicity of PLGADOX was compared with clinically approved non-PEGylated, liposomal nanoformulation of DOX (LipoDOX) and conventional DOX form (ConvDOX). The study was performed using Wistar Han rats of both sexes that were treated intravenously for 28 days with 5 doses of tested substances at intervals of 5 days. Histopathological analyses of heart tissues showed the presence of myofiber necrosis, degeneration processes, myocytolysis, and hemorrhage after treatment with ConvDOX, whereas only myofiber degeneration and hemorrhage were present after the treatment with nanoformulations. All DOX formulations caused an increase in the troponin T with the greatest increase caused by convDOX. qPCR analyses revealed an increase in the expression of inflammatory markers IL-6 and IL-8 after ConvDOX and an increase in IL-8 expression after lipoDOX treatments. The mass spectra imaging (MSI) of heart tissue indicates numerous metabolic and lipidomic changes caused by ConvDOX, while less severe cardiac damages were found after treatment with nanoformulations. In the case of LipoDOX, autophagy and apoptosis were still detectable, whereas PLGADOX induced only detectable mitochondrial toxicity. Cardiotoxic effects were frequently sex-related with the greater risk of cardiotoxicity observed mostly in male rats.


Subject(s)
Cardiotoxicity , Doxorubicin , Polylactic Acid-Polyglycolic Acid Copolymer , Rats, Wistar , Doxorubicin/chemistry , Doxorubicin/pharmacology , Doxorubicin/analogs & derivatives , Animals , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Rats , Male , Cardiotoxicity/prevention & control , Female , Apoptosis/drug effects , Nanoparticles/chemistry , Myocardium/pathology , Myocardium/metabolism , Polyethylene Glycols/chemistry , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacology , Heart/drug effects , Liposomes/chemistry
17.
ACS Biomater Sci Eng ; 10(5): 3425-3437, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38622760

ABSTRACT

Triple-negative breast cancer (TNBC) lacks expressed protein targets, making therapy development challenging. Hydrogels offer a promising new route in this regard by improving the chemotherapeutic efficacy through increased solubility and sustained release. Moreover, subcutaneous hydrogel administration reduces patient burden by requiring less therapy and shorter treatment times. We recently established the design principles for the supramolecular assembly of single-domain coiled-coils into hydrogels. Using a modified computational design algorithm, we designed Q8, a hydrogel with rapid assembly for faster therapeutic hydrogel preparation. Q8 encapsulates and releases doxorubicin (Dox), enabling localized sustained release via subcutaneous injection. Remarkably, a single subcutaneous injection of Dox-laden Q8 (Q8•Dox) significantly suppresses tumors within just 1 week. This work showcases the bottom-up engineering of a fully protein-based drug delivery vehicle for improved TBNC treatment via noninvasive localized therapy.


Subject(s)
Delayed-Action Preparations , Doxorubicin , Hydrogels , Triple Negative Breast Neoplasms , Doxorubicin/pharmacology , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Doxorubicin/therapeutic use , Hydrogels/chemistry , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Female , Humans , Animals , Delayed-Action Preparations/chemistry , Cell Line, Tumor , Protein Engineering , Mice , Drug Liberation , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/therapeutic use , Antibiotics, Antineoplastic/chemistry
18.
J Colloid Interface Sci ; 668: 88-97, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38669999

ABSTRACT

Nanotheranostic platforms, which can respond to tumor microenvironments (TME, such as low pH and hypoxia), are immensely appealing for photodynamic therapy (PDT). However, hypoxia in solid tumors harms the treatment outcome of PDT which depends on oxygen molecules to generate cytotoxic singlet oxygen (1O2). Herein, we report the design of TME-responsive smart nanotheranostic platform (DOX/ZnO2@Zr-Ce6/Pt/PEG) which can generate endogenously hydrogen peroxide (H2O2) and oxygen (O2) to alleviate hypoxia for improving photodynamic-chemo combination therapy of tumors. DOX/ZnO2@Zr-Ce6/Pt/PEG nanocomposite was prepared by the synthesis of ZnO2 nanoparticles, in-situ assembly of Zr-Ce6 as typical metal-organic framework (MOF) on ZnO2 surface, in-situ reduction of Pt nanozymes, amphiphilic lipids surface coating and then doxorubicin (DOX) loading. DOX/ZnO2@Zr-Ce6/Pt/PEG nanocomposite exhibits average sizes of ∼78 nm and possesses a good loading capacity (48.8 %) for DOX. When DOX/ZnO2@Zr-Ce6/Pt/PEG dispersions are intratumorally injected into mice, the weak acidic TEM induces the decomposition of ZnO2 core to generate endogenously H2O2, then Pt nanozymes catalyze H2O2 to produce O2 for alleviating tumor hypoxia. Upon laser (630 nm) irradiation, the Zr-Ce6 component in DOX/ZnO2@Zr-Ce6/Pt/PEG can produce cytotoxic 1O2, and 1O2 generation rate can be enhanced by 2.94 times due to the cascaded generation of endogenous H2O2/O2. Furthermore, the generated O2 can suppress the expression of hypoxia-inducible factor α, and further enable tumor cells to become more sensitive to chemotherapy, thereby leading to an increased effectiveness of chemotherapy treatment. The photodynamic-chemo combination therapy from DOX/ZnO2@Zr-Ce6/Pt/PEG nanoplatform exhibits remarkable tumor growth inhibition compared to chemotherapy or PDT. Thus, the present study is a good demonstration of a TME-responsive nanoplatform in a multimodal approach for cancer therapy.


Subject(s)
Doxorubicin , Hydrogen Peroxide , Oxygen , Photochemotherapy , Theranostic Nanomedicine , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Animals , Mice , Doxorubicin/pharmacology , Doxorubicin/chemistry , Doxorubicin/administration & dosage , Oxygen/chemistry , Oxygen/metabolism , Humans , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Particle Size , Surface Properties , Drug Screening Assays, Antitumor , Cell Survival/drug effects , Cell Proliferation/drug effects , Cell Line, Tumor , Peroxides/chemistry , Peroxides/pharmacology , Nanoparticles/chemistry , Mice, Inbred BALB C , Zinc/chemistry , Zinc/pharmacology , Tumor Microenvironment/drug effects , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/administration & dosage
19.
Chem Phys Lipids ; 261: 105396, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38621603

ABSTRACT

In this study, we have developed a redox-sensitive (RS) liposomal doxorubicin formulation by incorporating 10,10'-diselanediylbis decanoic acid (DDA) organoselenium compound as the RS moiety. Hence, several RS liposomal formulations were prepared by using DOPE, HSPC, DDA, mPEG2000-DSPE, and cholesterol. In situ drug loading using a pH gradient and citrate complex yielded high drug to lipid ratio and encapsulation efficiency (100%) for RS liposomes. Liposomal formulations were characterized in terms of size, surface charge and morphology, drug loading, release properties, cell uptake and cytotoxicity, as well as therapeutic efficacy in BALB/c mice bearing C26 tumor cells. The formulations showed an average particle size of 200 nm with narrow size distributions (PDI < 0.3), and negative surface charges varying from -6 mV to -18.6 mV. Our study confirms that the presence of the DDA compound in liposomes is highly sensitive to hydrogen peroxide at 0.1% w/v, resulting in a significant burst release of up to 40%. The in vivo therapeutic efficacy study in BALB/c mice bearing C26 colon carcinoma confirmed the promising function of RS liposomes in the tumor microenvironment which led to a prolonged median survival time (MST). The addition of hydrogenated soy phosphatidylcholine (HSPC) with a high transition temperature (Tm: 52-53.5°C) extended the MST of our 3-component formulation of F14 (DOPE/HSPC/DDA) to 60 days in comparison to Caelyx (PEGylated liposomal Dox), which is not RS-sensitive (39 days). Overall, HSPC liposomes bearing RS-sensitive moiety enhanced therapeutic efficacy against colon cancer in vitro and in vivo. This achievement unequivocally underscores the criticality of high-TM phospholipids, particularly HSPC, in significantly enhancing liposome stability within the bloodstream. In addition, RS liposomes enable the on-demand release of drugs, leveraging the redox environment of tumor cells, thereby augmenting the efficacy of the formulation.


Subject(s)
Colonic Neoplasms , Doxorubicin , Mice, Inbred BALB C , Oxidation-Reduction , Phospholipids , Doxorubicin/chemistry , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Doxorubicin/analogs & derivatives , Animals , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Mice , Phospholipids/chemistry , Temperature , Polyethylene Glycols/chemistry , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/administration & dosage , Cell Line, Tumor , Liposomes/chemistry , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Humans , Particle Size
20.
Expert Opin Drug Deliv ; 21(3): 399-422, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38623735

ABSTRACT

INTRODUCTION: Doxorubicin (DOX) emerges as a cornerstone in the arsenal of potent chemotherapeutic agents. Yet, the clinical deployment of DOX is tarnished by its proclivity to induce severe cardiotoxic effects, culminating in heart failure and other consequential morbidities. In response, a panoply of strategies has undergone rigorous exploration over recent decades, all aimed at attenuating DOX's cardiotoxic impact. The advent of encapsulating DOX within lipidic or polymeric nanocarriers has yielded a dual triumph, augmenting DOX's therapeutic efficacy while mitigating its deleterious side effects. AREAS COVERED: Recent strides have spotlighted the emergence of DOX conjugates as particularly auspicious avenues for ameliorating DOX-induced cardiotoxicity. These conjugates entail the fusion of DOX through physical or chemical bonds with diminutive natural or synthetic moieties, polymers, biomolecules, and nanoparticles. This spectrum encompasses interventions that impinge upon DOX's cardiotoxic mechanism, modulate cellular uptake and localization, confer antioxidative properties, or refine cellular targeting. EXPERT OPINION: The endorsement of DOX conjugates as a compelling stratagem to mitigate DOX-induced cardiotoxicity resounds from this exegesis, amplifying safety margins and the therapeutic profile of this venerated chemotherapeutic agent. Within this ambit, DOX conjugates stand as a beacon of promise in the perpetual pursuit of refining chemotherapy-induced cardiac compromise.


Subject(s)
Antibiotics, Antineoplastic , Cardiotoxicity , Doxorubicin , Drug Carriers , Nanoparticles , Doxorubicin/adverse effects , Doxorubicin/administration & dosage , Cardiotoxicity/prevention & control , Cardiotoxicity/etiology , Humans , Animals , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/chemistry , Drug Carriers/chemistry , Nanoparticles/chemistry , Drug Delivery Systems , Polymers/chemistry , Heart Failure/drug therapy , Heart Failure/chemically induced , Lipids/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...