Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 509
Filter
1.
Bull Exp Biol Med ; 171(4): 517-522, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34542769

ABSTRACT

Visualization of transplanted stem cells in the brain is an important issue in the study of the mechanisms of their therapeutic action. MRI allowing visualization of single transplanted cells previously labeled with superparamagnetic iron oxide particles is among the most informative methods of non-invasive intravital imaging. Verification of MRI data using pathomorphological examination at the microscopic level helps to avoid errors in data interpretation. However, making serial sections of the whole brain and searching for transplanted cells under the microscope is laborious and time-consuming. We have developed a method for 3D modeling of the distribution of transplanted cells in the brain allowing navigating through various brain structures and identifying the areas of accumulation of transplanted cells, which significantly increases the efficiency and reduces the time of histological examination.


Subject(s)
Brain/pathology , Cell Tracking/methods , Ischemic Stroke/therapy , Mesenchymal Stem Cell Transplantation/methods , Animals , Brain/blood supply , Brain/metabolism , Cells, Cultured , Disease Models, Animal , Female , Ferric Compounds/chemistry , Ferric Compounds/pharmacokinetics , Humans , Imaging, Three-Dimensional , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/therapy , Infusions, Intra-Arterial , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Magnetic Resonance Imaging , Magnetite Nanoparticles/chemistry , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Pregnancy , Rats , Rats, Wistar
2.
Inorg Chem ; 60(12): 8651-8664, 2021 Jun 21.
Article in English | MEDLINE | ID: mdl-34110140

ABSTRACT

Four high-spin Fe(III) macrocyclic complexes, including three dinuclear and one mononuclear complex, were prepared toward the development of more effective iron-based magnetic resonance imaging (MRI) contrast agents. All four complexes contain a 1,4,7-triazacyclononane macrocyclic backbone with two hydroxypropyl pendant groups, an ancillary aryl or biphenyl group, and a coordination site for a water ligand. The pH potentiometric titrations support one or two deprotonations of the complexes, most likely deprotonation of hydroxypropyl groups at near-neutral pH. Variable-temperature 17O NMR studies suggest that the inner-sphere water ligand is slow to exchange with bulk water on the NMR time scale. Water proton T1 relaxation times measured for solutions of the Fe(III) complexes at pH 7.2 showed that the dinuclear complexes have a 2- to 3-fold increase in r1 relaxivity in comparison to the mononuclear complex per molecule at field strengths ranging from 1.4 T to 9.4 T. The most effective agent, a dinuclear complex with macrocycles linked through para-substitution of an aryl group (Fe2(PARA)), has an r1 of 6.7 mM-1 s-1 at 37 °C and 4.7 T or 3.3 mM-1 s-1 per iron center in the presence of serum albumin and shows enhanced blood pool and kidney contrast in mice MRI studies.


Subject(s)
Contrast Media/chemistry , Coordination Complexes/chemistry , Ferric Compounds/chemistry , Macrocyclic Compounds/chemistry , Magnetic Resonance Imaging , Animals , Contrast Media/chemical synthesis , Contrast Media/pharmacokinetics , Coordination Complexes/chemical synthesis , Coordination Complexes/pharmacokinetics , Ferric Compounds/pharmacokinetics , Humans , Macrocyclic Compounds/chemical synthesis , Macrocyclic Compounds/pharmacokinetics , Mice , Mice, Inbred BALB C , Molecular Structure , Serum Albumin, Human/chemistry
3.
Nanomedicine ; 35: 102406, 2021 07.
Article in English | MEDLINE | ID: mdl-33932592

ABSTRACT

The harnessing of the cancer X-ray radiation therapy by gold-decorated Fe3O4 theranostic nanoparticles (Au-Fe3O4 NPs) under electromagnetic field was articulated. The applied electromagnetic field could assemble the NPs inside cell in oriented field direction and enhance the local irradiation dose inside cell. By materializing NPs, the absorption of the energy exposed by X-ray radiation under electromagnetic field was restricted. The cytotoxic properties of the Au-Fe3O4 NPs were assessed using MTT assay in L929, HeLa and PC3 cell lines under radiation and dark conditions. The efficiency of the Au-Fe3O4 NPs under 2 Gy dose radiations was higher than 6 Gy radiations in untreated cells. The in vitro measurements showed that under electromagnetic field and X-ray radiation therapy with Au-Fe3O4 NPs, around 90% of the cancer cells population was annihilated. The in vivo measurements indicated that the tumor shape and size under X-ray with Au-Fe3O4 NPs after 3 weeks were efficiently deteriorated.


Subject(s)
Antineoplastic Agents , Electromagnetic Fields , Ferric Compounds , Gold , Nanoparticles , Neoplasms , Theranostic Nanomedicine , X-Ray Therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Ferric Compounds/chemistry , Ferric Compounds/pharmacokinetics , Ferric Compounds/pharmacology , Gold/chemistry , Gold/pharmacokinetics , Gold/pharmacology , HeLa Cells , Humans , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasms/metabolism , Neoplasms/radiotherapy , PC-3 Cells
4.
Nanomedicine ; 35: 102392, 2021 07.
Article in English | MEDLINE | ID: mdl-33872772

ABSTRACT

This paper reports a smart intracellular nanocarrier for sustainable and controlled drug release in non-invasive neuroregeneration. The nanocarrier is composed by superparamagnetic iron oxide-gold (SPIO-Au) core-shell nanoparticles (NPs) conjugated with porous coordination cages (PCCs) through the thiol-containing molecules as bridges. The negatively charged PCC-2 and positively charged PCC-3 are compared for intracellular targeting. Both types result in intracellular targeting via direct penetration across cellular membranes. However, the pyrene (Py)-PEG-SH bridge enabled functionalization of SPIO-Au NPs with PCC-3 exhibits higher interaction with PC-12 neuron-like cells, compared with the rhodamine B (RhB)-PEG-SH bridge enabled case and the stand-alone SPIO-Au NPs. With neglectable toxicities to PC-12 cells, the proposed SPIO-Au-RhB(Py)-PCC-2(3) nanocarriers exhibit effective drug loading capacity of retinoic acid (RA) at 13.505 µg/mg of RA/NPs within 24 h. A controlled release of RA is achieved by using a low-intensity 525 nm LED light (100% compared to 40% for control group within 96 h).


Subject(s)
Drug Carriers , Ferric Compounds , Gold , Nanoparticles , Nerve Regeneration/drug effects , Tretinoin , Animals , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Ferric Compounds/chemistry , Ferric Compounds/pharmacokinetics , Ferric Compounds/pharmacology , Gold/chemistry , Gold/pharmacokinetics , Gold/pharmacology , Nanoparticles/chemistry , Nanoparticles/therapeutic use , PC12 Cells , Porosity , Rats , Tretinoin/chemistry , Tretinoin/pharmacokinetics , Tretinoin/pharmacology
5.
Mol Pharm ; 18(4): 1544-1557, 2021 04 05.
Article in English | MEDLINE | ID: mdl-33621099

ABSTRACT

Complex iron nanoparticle-based drugs are one of the oldest and most frequently administered classes of nanomedicines. In the US, there are seven FDA-approved iron nanoparticle reference drug products, of which one also has an approved generic drug product (i.e., sodium ferric gluconate (SFG)). These products are indicated for the treatment of iron deficiency anemia and are administered intravenously. On the molecular level, iron nanomedicines are colloids composed of an iron oxide core with a carbohydrate coating. This formulation makes nanomedicines more complex than conventional small molecule drugs. As such, these products are often referred to as nonbiological complex drugs (e.g., by the nonbiological complex drugs (NBCD) working group) or complex drug products (e.g., by the FDA). Herein, we report a comprehensive study of the physiochemical properties of the iron nanoparticle product SFG. SFG is the single drug for which both an innovator (Ferrlecit) and generic product are available in the US, allowing for comparative studies to be performed. Measurements focused on the iron core of SFG included optical spectroscopy, inductively coupled plasma mass spectrometry (ICP-MS), X-ray powder diffraction (XRPD), 57Fe Mössbauer spectroscopy, and X-ray absorbance spectroscopy (XAS). The analysis revealed similar ferric-iron-oxide structures. Measurements focused on the carbohydrate shell comprised of the gluconate ligands included forced acid degradation, dynamic light scattering (DLS), analytical ultracentrifugation (AUC), and gel permeation chromatography (GPC). Such analysis revealed differences in composition for the innovator versus the generic SFG. These studies have the potential to contribute to future quality assessment of iron complex products and will inform on a pharmacokinetic study of two therapeutically equivalent iron gluconate products.


Subject(s)
Drugs, Generic/chemistry , Ferric Compounds/chemistry , Nanoparticles/chemistry , Anemia, Iron-Deficiency/drug therapy , Chemistry, Pharmaceutical , Chromatography, Gel , Drugs, Generic/administration & dosage , Drugs, Generic/pharmacokinetics , Drugs, Generic/standards , Dynamic Light Scattering , Equivalence Trials as Topic , Ferric Compounds/administration & dosage , Ferric Compounds/pharmacokinetics , Ferric Compounds/standards , Humans , Nanoparticles/administration & dosage , Nanoparticles/standards , Quality Control , Ultracentrifugation
6.
Sci Rep ; 11(1): 3286, 2021 02 08.
Article in English | MEDLINE | ID: mdl-33558583

ABSTRACT

Iron oxide particles (IOP) are commonly used for Cellular Magnetic Resonance Imaging (MRI) and in combination with several treatments, like Magnetic Fluid Hyperthermia (MFH), due to the rise in temperature they provoke under an Alternating Magnetic Field (AMF). Micrometric IOP have a high sensitivity of detection. Nevertheless, little is known about their internalization processes or their potential heat power. Two micrometric commercial IOP (from Bangs Laboratories and Chemicell) were characterized by Transmission Electron Microscopy (TEM) and their endocytic pathways into glioma cells were analyzed. Their Specific Absorption Rate (SAR) and cytotoxicity were evaluated using a commercial AMF inductor. T2-weighted imaging was used to monitor tumor growth in vivo after MFH treatment in mice. The two micron-sized IOP had similar structures and r2 relaxivities (100 mM-1 s-1) but involved different endocytic pathways. Only ScreenMAG particles generated a significant rise in temperature following AMF (SAR = 113 W g-1 Fe). After 1 h of AMF exposure, 60% of ScreenMAG-labeled cells died. Translated to a glioma model, 89% of mice responded to the treatment with smaller tumor volume 42 days post-implantation. Micrometric particles were investigated from their characterization to their intracellular internalization pathways and applied in one in vivo cancer treatment, i.e. MFH.


Subject(s)
Cell Tracking , Ferric Compounds , Glioma , Hyperthermia, Induced , Magnetic Resonance Imaging , Animals , Cell Line, Tumor , Ferric Compounds/pharmacokinetics , Ferric Compounds/pharmacology , Glioma/diagnostic imaging , Glioma/therapy , Humans , Mice, Nude , Xenograft Model Antitumor Assays
7.
Expert Rev Hematol ; 14(1): 7-29, 2021 01.
Article in English | MEDLINE | ID: mdl-33317356

ABSTRACT

Introduction: Originally approved in Europe in 2009, ferric derisomaltose is the most recently authorized intravenous iron compound in the United States of America (2020). Ferric derisomaltose given as a rapid high-dose infusion can allow complete iron repletion in a single dose and it is now widely used in the treatment of iron deficiency. Areas covered: The chemistry, pharmacodynamics and pharmacokinetics of ferric derisomaltose are reviewed. Results from phase II, III and IV trials regarding efficacy and safety are presented. Mechanisms behind minor infusion reactions, hypersensitivity and hypophosphatemia are discussed. The economic impact of ferric derisomaltose use is presented. Data pertaining to the use of ferric derisomaltose in iron deficiency anemia, chronic kidney disease, inflammatory bowel disease, chronic heart failure, perioperative care and other patient groups are comprehensively covered. Expert opinion: Ferric derisomaltose is an effective intravenous iron formulation with a good safety profile, providing rapid, cost-effective iron repletion. Ferric derisomaltose releases low quantities of labile iron relative to older compounds. Anaphylaxis is extremely rare, and 'Fishbane' reactions are uncommon. Hypophosphatemia following ferric derisomaltose administration is infrequent in comparison to other intravenous irons such as ferric carboxymaltose. The scope of ferric derisomaltose use is growing with increasing research in these areas.


Subject(s)
Anemia/drug therapy , Disaccharides/therapeutic use , Animals , Clinical Trials as Topic , Disaccharides/adverse effects , Disaccharides/pharmacokinetics , Disaccharides/pharmacology , Drug and Narcotic Control , Ferric Compounds/adverse effects , Ferric Compounds/pharmacokinetics , Ferric Compounds/pharmacology , Ferric Compounds/therapeutic use , Humans , Treatment Outcome
8.
Ann Pharmacother ; 55(2): 222-229, 2021 02.
Article in English | MEDLINE | ID: mdl-32633548

ABSTRACT

OBJECTIVE: To review the pharmacology, efficacy, and safety of ferric maltol (FM), an oral iron formulation, for iron deficiency anemia (IDA). DATA SOURCES: A MEDLINE/PubMed and EMBASE (January 1, 1985, to June 19, 2020) literature search was performed using the terms ferric maltol, accrufer, feraccru, iron maltol, ferric trimaltol, iron deficiency, iron deficiency anemia, inflammatory bowel disease, and chronic kidney disease. Additional data sources included prescribing information, abstracts, and the National Institutes of Health Clinical Trials Registry. STUDY SELECTION/DATA EXTRACTION: English language literature evaluating FM pharmacology, pharmacokinetics, efficacy, or safety in the treatment of IDA were reviewed. DATA SYNTHESIS: FM is a ferric, non-salt-based oral iron formulation demonstrating improved tolerance in patients with previous intolerance to other iron formulations. Phase 3 trials demonstrated significant improvements in anemia and serum iron parameters in patients with inflammatory bowel disease (IBD) and chronic kidney disease (CKD). Common adverse effects were gastrointestinal intolerance. RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE: FM is an effective and well-tolerated alternative to oral iron salts for patients with IBD or CKD and IDA. Emerging data suggest that FM is noninferior to intravenous (IV) ferric carboxymaltose in patients with IBD and IDA. Prior to selecting FM over IV iron products, consideration should be given to time to normalization of Hb, ease of administration, cost, and tolerability. CONCLUSION: FM is a relatively safe, effective oral iron therapy that may be better tolerated than other oral iron formulations. FM may be an effective alternative to IV iron in patients with IBD.


Subject(s)
Anemia, Iron-Deficiency/drug therapy , Ferric Compounds/therapeutic use , Hematinics/therapeutic use , Pyrones/therapeutic use , Administration, Intravenous , Administration, Oral , Adult , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/complications , Clinical Trials as Topic , Female , Ferric Compounds/administration & dosage , Ferric Compounds/adverse effects , Ferric Compounds/pharmacokinetics , Hematinics/administration & dosage , Hematinics/adverse effects , Hematinics/pharmacokinetics , Humans , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/drug therapy , Male , Maltose/administration & dosage , Maltose/adverse effects , Maltose/analogs & derivatives , Maltose/therapeutic use , Pyrones/administration & dosage , Pyrones/adverse effects , Pyrones/pharmacokinetics , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/drug therapy , Treatment Outcome
9.
Int J Nanomedicine ; 15: 4677-4689, 2020.
Article in English | MEDLINE | ID: mdl-32669844

ABSTRACT

BACKGROUND: Superparamagnetic iron oxide nanoparticles (SPIONs) have displayed multifunctional applications in cancer theranostics following systemic delivery. In an effort to increase the therapeutic potential of local therapies (including focal hyperthermia), nanoparticles can also be administered intratumorally. Therefore, the development of a reliable pharmacokinetic model for the prediction of nanoparticle distribution for both clinically relevant routes of delivery is of high importance. MATERIALS AND METHODS: The biodistribution of SPIONs (of two different sizes - 130 nm and 60 nm) radiolabeled with zirconium-89 or technetium-99m following intratumoral or intravenous injection was investigated in C57/Bl6 mice bearing subcutaneous GL261 glioblastomas. Based on PET/CT biodistribution data, a novel pharmacokinetic model was established for a better understanding of the pharmacokinetics of the SPIONs after both administration routes. RESULTS: The PET image analysis of the nanoparticles (confirmed by histology) demonstrated the presence of radiolabeled nanoparticles within the glioma site (with low amounts in the liver and spleen) at all investigated time points following intratumoral injection. The mathematical model confirmed the dynamic nanoparticle redistribution in the organism over a period of 72 h with an equilibrium reached after 100 h. Intravenous injection of nanoparticles demonstrated a different distribution pattern with a rapid particle retention in all organs (particularly in liver and spleen) and a subsequent slow release rate. CONCLUSION: The mathematical model demonstrated good agreement with experimental data derived from tumor mouse models suggesting the value of this tool to predict the real-time pharmacokinetic features of SPIONs in vivo. In the future, it is planned to adapt our model to other nanoparticle formulations to more precisely describe their biodistribution in in vivo model systems.


Subject(s)
Ferric Compounds/administration & dosage , Ferric Compounds/pharmacokinetics , Glioblastoma/diagnostic imaging , Magnetite Nanoparticles/administration & dosage , Animals , Female , Glioblastoma/pathology , Injections , Injections, Intravenous , Magnetite Nanoparticles/chemistry , Mice, Inbred C57BL , Models, Biological , Positron Emission Tomography Computed Tomography , Radioisotopes/pharmacokinetics , Technetium/pharmacokinetics , Theranostic Nanomedicine/methods , Tissue Distribution , Xenograft Model Antitumor Assays , Zirconium/pharmacokinetics
10.
Nephrol Dial Transplant ; 35(7): 1136-1144, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32514572

ABSTRACT

BACKGROUND: Anemia of chronic kidney disease (CKD) is, in part, caused by hepcidin-mediated impaired iron absorption. However, phosphate binder, ferric citrate (FC) overcomes the CKD-induced impairment of iron absorption and increases serum iron, transferrin saturation, and iron stores and reduces erythropoietin requirements in CKD/ESRD patients. The mechanism and sites of intestinal absorption of iron contained in FC were explored here. METHODS: Eight-week old rats were randomized to sham-operated or 5/6 nephrectomized (CKD) groups and fed either regular rat chow or rat chow containing 4% FC for 6 weeks. They were then euthanized, and tissues were processed for histological and biochemical analysis using Prussian blue staining, Western blot analysis to quantify intestinal epithelial tight junction proteins and real-time PCR to measure Fatty Acid receptors 2 (FFA2) and 3 (FFA3) expressions. RESULTS: CKD rats exhibited hypertension, anemia, azotemia, and hyperphosphatemia. FC-treated CKD rats showed significant reductions in blood pressure, serum urea, phosphate and creatinine levels and higher serum iron and blood hemoglobin levels. This was associated with marked increase in iron content of the epithelial and subepithelial wall of the descending colon and modest iron deposits in the proximal tubular epithelial cells of their remnant kidneys. No significant difference was found in hepatic tissue iron content between untreated and FC-treated CKD or control groups. Distal colon's epithelial tight Junction proteins, Occludin, JAM-1 and ZO-1 were markedly reduced in the CKD groups. The FFA2 expression in the jejunum and FFA3 expression in the distal colon were significantly reduced in the CKD rats and markedly increased with FC administration. CONCLUSION: Iron contained in the phosphate binder, FC, is absorbed by the distal colon of the CKD animals via disrupted colonic epithelial barrier and upregulation of short chain fatty acid transporters.


Subject(s)
Ferric Compounds/metabolism , Ferric Compounds/pharmacokinetics , Hyperphosphatemia/prevention & control , Intestinal Absorption , Iron/metabolism , Phosphates/metabolism , Renal Insufficiency, Chronic/complications , Animals , Colon/metabolism , Erythropoietin/metabolism , Hyperphosphatemia/etiology , Hyperphosphatemia/metabolism , Male , Rats , Rats, Sprague-Dawley , Tissue Distribution
11.
Food Funct ; 11(7): 6240-6250, 2020 Jul 01.
Article in English | MEDLINE | ID: mdl-32596698

ABSTRACT

Nanosized iron is a promising candidate as an iron fortificant due to its good solubility and bioavailability. Here, ferric hydrolysis in the presence of salmon/herring sperm DNA yielded irregularly shaped, highly negatively charged DNA-stabilized ferric oxyhydroxide nanoparticles (DNA-FeONPs) aggregated from 2-4 nm primary spherical monomers, in which phosphodioxy groups of the DNA backbone served as the iron-nucleation sites with high molecular weight (>500 bp), double-stranded winding, and acidic environmental pH disfavoring DNA's iron-loading capacity. The calcein fluorescence-quenching kinetics of polarized Caco-2 cells revealed the involvement of divalent transporter 1, macropinocytosis and nucleolin-mediated endocytosis in intestinal iron absorption from DNA-FeONPs with low molecular weight (<500 bp) favoring the performance of DNA in aiding iron absorption. In anemic rats, dietary DNA-FeONPs showed >80% relative iron bioavailability compared to FeSO4 as per hemoglobin regeneration efficiencies and delivered intestinally available nanosized iron, as determined by luminal iron speciation analysis. Overall, fish sperm DNA is promising in stabilizing and delivering bioavailable nanosized iron.


Subject(s)
DNA/chemistry , Ferric Compounds/administration & dosage , Ferric Compounds/pharmacokinetics , Fishes , Nanoparticles/administration & dosage , Spermatozoa/chemistry , Animals , Biological Availability , Caco-2 Cells , Drug Stability , Endocytosis , Humans , Intestinal Absorption , Iron/administration & dosage , Male , Rats , Rats, Sprague-Dawley , Salmon
12.
Molecules ; 25(10)2020 May 13.
Article in English | MEDLINE | ID: mdl-32414058

ABSTRACT

Complexes of Fe(III) that contain a triazacyclononane (TACN) macrocycle, two pendant hydroxyl groups, and a third ancillary pendant show promise as MRI contrast agents. The ancillary group plays an important role in tuning the solution relaxivity of the Fe(III) complex and leads to large changes in MRI contrast enhancement in mice. Two new Fe(III) complexes, one with a third coordinating hydroxypropyl pendant, Fe(L2), and one with an anionic non-coordinating sulfonate group, Fe(L1)(OH2), are compared. Both complexes have a deprotonated hydroxyl group at neutral pH and electrode potentials representative of a stabilized trivalent iron center. The r1 relaxivity of the Fe(L1)(OH2) complex is double that of the saturated complex, Fe(L2), at 4.7 T, 37 °C in buffered solutions. However, variable-temperature 17O-NMR experiments show that the inner-sphere water of Fe(L1)(OH2) does not exchange rapidly with bulk water under these conditions. The pendant sulfonate group in Fe(L1)(OH2) confers high solubility to the complex in comparison to Fe(L2) or previously studied analogues with benzyl groups. Dynamic MRI studies of the two complexes showed major differences in their pharmacokinetics clearance rates compared to an analogue containing a benzyl ancillary group. Rapid blood clearance and poor binding to serum albumin identify Fe(L1)(OH2) for development as an extracellular fluid contrast agent.


Subject(s)
Contrast Media , Ferric Compounds , Macrocyclic Compounds , Magnetic Resonance Imaging , Animals , Contrast Media/chemistry , Contrast Media/pharmacokinetics , Contrast Media/pharmacology , Ferric Compounds/chemistry , Ferric Compounds/pharmacokinetics , Ferric Compounds/pharmacology , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/pharmacokinetics , Macrocyclic Compounds/pharmacology , Mice , Mice, Inbred BALB C
13.
Bull Exp Biol Med ; 168(6): 789-792, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32328946

ABSTRACT

We studied the distribution of ferrihydrite nanoparticles isolated from bacteria Klebsiella oxytoca in the whole body in vivo and in a cultured isolated organ (liver). The possibility of controlling these nanoparticles in the body using a magnetic field was assessed. One hour after intravenous injection of ferrihydrite nanoparticles to mice, their accumulation was observed in the liver, lungs, and kidneys. Experiment with cultured isolated rat liver showed that these nanoparticles can be controlled by a magnetic field and the influence of magnetic nanoparticles on the liver over 1 h does not lead to destruction of liver cells associated with the release of the marker enzyme AST. These results show the possibility of using magnetic nanoparticles as a system for controlled drug delivery in the body.


Subject(s)
Ferric Compounds/chemistry , Liver/diagnostic imaging , Lung/diagnostic imaging , Magnetite Nanoparticles/administration & dosage , Animals , Animals, Outbred Strains , Ferric Compounds/pharmacokinetics , Injections, Intravenous , Kidney/diagnostic imaging , Kidney/metabolism , Klebsiella oxytoca/chemistry , Liver/metabolism , Lung/metabolism , Magnetic Fields , Magnetic Resonance Imaging/methods , Magnetite Nanoparticles/chemistry , Male , Mice , Organ Culture Techniques , Rats , Spleen/diagnostic imaging , Spleen/metabolism
14.
Mater Sci Eng C Mater Biol Appl ; 109: 110579, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32228948

ABSTRACT

3D multifunctional bone scaffolds have recently attracted more attention in bone tissue engineering because of addressing critical issues like bone cancer and inflammation beside bone regeneration. In this study, a 3D bone scaffold is fabricated from Mg2SiO4-CoFe2O4 nanocomposite which is synthesized via a two-step synthesis strategy and then the scaffold's surface is modified with poly-3-hydroxybutyrate (P3HB)-ordered mesoporous magnesium silicate (OMMS) composite to improve its physicochemical and biological properties. The Mg2SiO4-CoFe2O4 scaffold is fabricated through polymer sponge technique and the scaffold exhibits an interconnected porous structure in the range of 100-600 µm. The scaffold is then coated with OMMS/P3HB composite via dip coating and the physical, chemical, and biological-related properties of OMMS/P3HB composite-coated scaffold are assessed and compared to the non-coated and P3HB-coated scaffolds in vitro. It is found that, on the one hand, P3HB increases the cell attachment, proliferation, and compressive strength of the scaffold, but on the other hand, it weakens the bioactivity kinetic. Addition of OMMS to the coating composition is accompanied with significant increase in bioactivity kinetic. Besides, OMMS/P3HB composite-coated scaffold exhibits higher drug loading capacity and more controlled release manner up to 240 h than the other samples because of OMMS which has a high surface area and ordered mesoporous structure suitable for controlled release applications. The overall results indicate that OMMS/P3HB coating on Mg2SiO4-CoFe2O4 scaffold leads to a great improvement in bioactivity, drug delivery potential, compressive strength, cell viability, and proliferation. Moreover, OMMS/P3HB composite-coated scaffold has heat generation capability for hyperthermia-based bone cancer therapy and so it is suggested as a multifunctional scaffold with great potentials for bone cancer therapy and regeneration.


Subject(s)
Bone Neoplasms/therapy , Bone Regeneration , Coated Materials, Biocompatible , Hyperthermia, Induced , Nanocomposites , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacokinetics , Coated Materials, Biocompatible/pharmacology , Cobalt/chemistry , Cobalt/pharmacokinetics , Cobalt/pharmacology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Ferric Compounds/chemistry , Ferric Compounds/pharmacokinetics , Ferric Compounds/pharmacology , Humans , Hydroxybutyrates , Magnesium Silicates/chemistry , Magnesium Silicates/pharmacokinetics , Magnesium Silicates/pharmacology , Nanocomposites/chemistry , Nanocomposites/therapeutic use , Polyesters , Porosity
15.
Biomater Sci ; 8(9): 2590-2599, 2020 May 06.
Article in English | MEDLINE | ID: mdl-32238997

ABSTRACT

In this work we describe the formulation and characterisation of red-emitting polymeric nanocapsules (NCs) incorporating superparamagnetic iron oxide nanoparticles (SPIONs) for magnetic tumour targeting. The self-fluorescent oligomers were synthesised and chemically conjugated to PLGA which was confirmed by NMR spectroscopy, FT-IR spectroscopy and mass spectrometry. Hydrophobic SPIONs were synthesised through thermal decomposition and their magnetic and heating properties were assessed by SQUID magnetometry and calorimetric measurements, respectively. Magnetic nanocapsules (m-NCs) were prepared by a single emulsification/solvent evaporation method. Their in vitro cytotoxicity was examined in CT26 colon cancer cells. The formulated fluorescent m-NCs showed good stability and biocompatibility both in vitro and in vivo in CT 26 colon cancer models. Following intravenous injection, accumulation of m-NCs in tumours was observed by optical imaging. A higher iron content in the tumours exposed to a magnetic field, compared to the contralateral tumours without magnetic exposure in the same animal, further confirmed the magnetic tumour targeting in vivo. The overall results show that the engineered red-emitting m-NCs have great potential as multifunctional nanocarriers for multi-model bioimaging and magnetic-targeted drug delivery.


Subject(s)
Ferric Compounds/administration & dosage , Fluorescent Dyes/administration & dosage , Nanocapsules/administration & dosage , Neoplasms/diagnostic imaging , Neoplasms/therapy , Animals , Cell Line, Tumor , Drug Delivery Systems , Female , Ferric Compounds/pharmacokinetics , Fluorescent Dyes/pharmacokinetics , Hyperthermia, Induced , Iron/metabolism , Magnetic Phenomena , Mice, Inbred BALB C , Neoplasms/metabolism , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/pharmacokinetics , Polyglactin 910/administration & dosage , Polyglactin 910/pharmacokinetics , Tissue Distribution
16.
Sci Rep ; 10(1): 5339, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32210349

ABSTRACT

Bouillon cubes are widely consumed and when fortified with iron could contribute in preventing iron deficiency. We report the development (part I) and evaluation (current part II) of a novel ferric phytate compound to be used as iron fortificant in condiments such as bouillon. Ferric pyrophosphate (FePP), is the compound of choice due to its high stability in foods, but has a modest absorption in humans. Our objective was to assess iron bioavailability from a novel iron fortificant consisting of ferric iron complexed with phytic acid and hydrolyzed corn protein (Fe-PA-HCP), used in bouillon with and without an inhibitory food matrix. In a randomised single blind, cross-over study, we measured iron absorption in healthy adult women (n = 22). In vitro iron bioaccessibility was assessed using a Caco-2 cell model. Iron absorption from Fe-PA-HCP was 1.5% and 4.1% in bouillon with and without inhibitory matrix, respectively. Relative iron bioavailability to FeSO4 was 2.4 times higher than from FePP in bouillon (17% vs 7%) and 5.2 times higher when consumed with the inhibitory meal (41% vs 8%). Similar results were found in vitro. Fe-PA-HCP has a higher relative bioavailability versus FePP, especially when bouillon is served with an inhibitory food matrix.


Subject(s)
Ferric Compounds/pharmacokinetics , Food, Fortified , Iron/pharmacokinetics , Phytic Acid/chemistry , Adult , Caco-2 Cells , Cross-Over Studies , Female , Ferric Compounds/chemistry , Ferritins/blood , Humans , Hydrolysis , Iron Radioisotopes/pharmacokinetics , Plant Proteins, Dietary/chemistry , Single-Blind Method , Young Adult , Zea mays/chemistry
17.
Artif Cells Nanomed Biotechnol ; 48(1): 443-451, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32024389

ABSTRACT

Superparamagnetic iron oxide nanoparticles (SPIONs) have been employed in several biomedical applications where they facilitate both diagnostic and therapeutic aims. Although the potential benefits of SPIONs with different surface chemistry and conjugated targeting ligands/proteins are considerable, complicated interactions between these nanoparticles (NPs) and cells leading to toxic impacts could limit their clinical applications. Hence, elevation of our knowledge regarding the SPION-related toxicity is necessary. Here, the present review article will consider current studies and compare the potential toxic effect of SPIONs with or without identical surface chemistries on different cell lines. It centers on cellular and molecular mechanisms underlying toxicity of SPIONs. Likewise, emphasis is being dedicated for toxicity of SPIONs in various cell lines, in vitro and animal models, in vivo.


Subject(s)
Ferric Compounds/pharmacokinetics , Ferric Compounds/toxicity , Magnetite Nanoparticles/toxicity , Animals , Cell Line , Cell Survival , Coated Materials, Biocompatible/toxicity , Ferric Compounds/chemistry , Humans , Magnetite Nanoparticles/chemistry , Nanomedicine , Reactive Oxygen Species/chemistry , Reactive Oxygen Species/toxicity , Tissue Distribution
18.
Sci Rep ; 10(1): 1171, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31980695

ABSTRACT

The use of magnetic fluid hyperthermia (MFH) for cancer therapy has shown promise but lacks suitable methods for quantifying exogenous irons such as superparamagnetic iron oxide (SPIO) nanoparticles as a source of heat generation under an alternating magnetic field (AMF). Application of quantitative susceptibility mapping (QSM) technique to prediction of SPIO in preclinical models has been challenging due to a large variation of susceptibility values, chemical shift from tissue fat, and noisier data arising from the higher resolution required to visualize the anatomy of small animals. In this study, we developed a robust QSM for the SPIO ferumoxytol in live mice to examine its potential application in MFH for cancer therapy. We demonstrated that QSM was able to simultaneously detect high level ferumoxytol accumulation in the liver and low level localization near the periphery of tumors. Detection of ferumoxytol distribution in the body by QSM, however, required imaging prior to and post ferumoxytol injection to discriminate exogenous iron susceptibility from other endogenous sources. Intratumoral injection of ferumoxytol combined with AMF produced a ferumoxytol-dose dependent tumor killing. Histology of tumor sections corroborated QSM visualization of ferumoxytol distribution near the tumor periphery, and confirmed the spatial correlation of cell death with ferumoxytol distribution. Due to the dissipation of SPIOs from the injection site, quantitative mapping of SPIO distribution will aid in estimating a change in temperature in tissues, thereby maximizing MFH effects on tumors and minimizing side-effects by avoiding unwanted tissue heating.


Subject(s)
Ferric Compounds/analysis , Ferrosoferric Oxide/analysis , Hyperthermia, Induced , Nanoparticles/analysis , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Animals , Cell Line, Tumor , Contrast Media , Ferric Compounds/pharmacokinetics , Ferric Compounds/therapeutic use , Ferrosoferric Oxide/pharmacokinetics , Ferrosoferric Oxide/therapeutic use , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred NOD , Nanoparticles/therapeutic use , Positron Emission Tomography Computed Tomography , Prostatic Neoplasms/pathology , Radioisotopes , Radiopharmaceuticals , Subcutaneous Tissue , Tissue Distribution , Tumor Burden , Xenograft Model Antitumor Assays , Zirconium
19.
Mol Pharm ; 17(3): 837-851, 2020 03 02.
Article in English | MEDLINE | ID: mdl-31977228

ABSTRACT

Delivery efficiencies of theranostic nanoparticles (NPs) based on passive tumor targeting strongly depend either on their blood circulation time or on appropriate modulations of the tumor microenvironment. Therefore, predicting the NP delivery efficiency before and after a tumor microenvironment modulation is highly desirable. Here, we present a new erythrocyte membrane-camouflaged magnetofluorescent nanocarrier (MMFn) with long blood circulation time (92 h) and high delivery efficiency (10% ID for Ehrlich murine tumor model). MMFns owe their magnetic and fluorescent properties to the incorporation of manganese ferrite nanoparticles (MnFe2O4 NPs) and IR-780 (a lipophilic indocyanine fluorescent dye), respectively, to their erythrocyte membrane-derived camouflage. MMFn composition, morphology, and size, as well as optical absorption, zeta potential, and fluorescent, magnetic, and magnetothermal properties, are thoroughly examined in vitro. We then present an analytical pharmacokinetic (PK) model capable of predicting the delivery efficiency (DE) and the time of peak tumor uptake (tmax), as well as changes in DE and tmax due to modulations of the tumor microenvironment, for potentially any nanocarrier. Experimental PK data sets (blood and tumor amounts of MMFns) are simultaneously fit to the model equations using the PK modeling software Monolix. We then validate our model analytical solutions with the numerical solutions provided by Monolix. We also demonstrate how our a priori nonmechanistic model for passive targeting relates to a previously reported mechanistic model for active targeting. All in vivo PK studies, as well as in vivo and ex vivo biodistribution studies, were conducted using two noninvasive techniques, namely, fluorescence molecular tomography (FMT) and alternating current biosusceptometry (ACB). Finally, histopathology corroborates our PK and biodistribution results.


Subject(s)
Drug Carriers/chemistry , Erythrocyte Membrane/chemistry , Ferric Compounds/chemistry , Fluorescent Dyes/chemistry , Magnetic Iron Oxide Nanoparticles/chemistry , Magnets/chemistry , Manganese Compounds/chemistry , Photothermal Therapy/methods , Animals , Carcinoma, Ehrlich Tumor/drug therapy , Disease Models, Animal , Drug Carriers/pharmacokinetics , Female , Ferric Compounds/pharmacokinetics , Fluorescent Dyes/pharmacokinetics , Hyperthermia, Induced/methods , Manganese Compounds/pharmacokinetics , Mice , Particle Size , Theranostic Nanomedicine/methods , Tissue Distribution , Tumor Burden/drug effects , Tumor Microenvironment/drug effects
20.
Clin Ther ; 42(2): 276-285, 2020 02.
Article in English | MEDLINE | ID: mdl-31937462

ABSTRACT

PURPOSE: Iron deficiency (ID) is one of the most commonly known nutritional deficiencies and is considered the primary cause of anemia (iron-deficiency anemia). Ferric carboxymaltose (FCM), an intravenous iron preparation, has been widely used for >10 years for iron-deficiency anemia treatment worldwide because of its many advantages. METHODS: This single-center, open-label, single dose escalation study in Chinese subjects was designed to assess the pharmacokinetic/pharmacodynamic parameters and safety of FCM in this population. The first 12 subjects received a 500-mg dose; after assessing safety data from the first 6 subjects in this cohort, another 12 subjects were assigned to the 1000-mg dose cohort. FINDINGS: After an infusion of FCM over 15 min, a rapid dose-dependent increase in total serum iron levels was observed with a median Tmax of 30 min following the start of the infusion for both cohorts. The Cmax and AUC for the 1000-mg dose were ~1.8-fold (p = 0.2929) and 2.3-fold (p = 0.0318) those associated with the 500-mg dose, respectively. Mean terminal t1/2 values were 12.3 and 10.5 h for the 2 cohorts. The renal elimination of FCM was negligible (<0.1%). Increase in mean serum iron levels and ferritin concentrations showed dose dependency. Iron-binding capacity was transiently well utilized after dosing, as indicated by transferrin saturation >88% with 500-mg FCM and >90% with 1000-mg FCM. Hemoglobin levels did not show significant changes during the 7-day observation period, whereas mean reticulocyte counts significantly increased in both cohorts, suggesting activation of the hematopoietic system. FCM was well tolerated in these Chinese subjects. No new or unexpected treatment-emergent adverse events were attributable to FCM. IMPLICATIONS: The pharmacokinetic/pharmacodynamic and safety profiles in Chinese subjects seemed comparable to those in white and Japanese populations. ChinaDrugTrials.org.cn identifier: CTR20160863.


Subject(s)
Anemia, Iron-Deficiency/blood , Ferric Compounds/pharmacology , Ferric Compounds/pharmacokinetics , Ferritins/blood , Iron/blood , Maltose/analogs & derivatives , Administration, Intravenous , Adult , Asian People , Female , Ferric Compounds/adverse effects , Humans , Male , Maltose/adverse effects , Maltose/pharmacokinetics , Maltose/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...