Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Mol Pharm ; 20(9): 4546-4558, 2023 09 04.
Article in English | MEDLINE | ID: mdl-37578286

ABSTRACT

Delamanid (DLM) is a hydrophobic small molecule therapeutic used to treat drug-resistant tuberculosis (DR-TB). Due to its hydrophobicity and resulting poor aqueous solubility, formulation strategies such as amorphous solid dispersions (ASDs) have been investigated to enhance its aqueous dissolution kinetics and thereby improve oral bioavailability. However, ASD formulations are susceptible to temperature- and humidity-induced phase separation and recrystallization under harsh storage conditions typically encountered in areas with high tuberculosis incidence. Nanoencapsulation represents an alternative formulation strategy to increase aqueous dissolution kinetics while remaining stable at elevated temperature and humidity. The stabilizer layer coating the nanoparticle drug core limits the formation of large drug domains by diffusion during storage, representing an advantage over ASDs. Initial attempts to form DLM-loaded nanoparticles via precipitation-driven self-assembly were unsuccessful, as the trifluoromethyl and nitro functional groups present on DLM were thought to interfere with surface stabilizer attachment. Therefore, in this work, we investigated the nanoencapsulation of DLM via emulsification, avoiding the formation of a solid drug core and instead keeping DLM dissolved in a dichloromethane dispersed phase during nanoparticle formation. Initial emulsion formulation screening by probe-tip ultrasonication revealed that a 1:1 mass ratio of lecithin and HPMC stabilizers formed 250 nm size-stable emulsion droplets with 40% DLM loading. Scale-up studies were performed to produce nearly identical droplet size distribution at larger scale using high-pressure homogenization, a continuous and industrially scalable technique. The resulting emulsions were spray-dried to form a dried powder, and in vitro dissolution studies showed dramatically enhanced dissolution kinetics compared to both as-received crystalline DLM and micronized crystalline DLM, owing to the increased specific surface area and partially amorphous character of the DLM-loaded nanoparticles. Solid-state NMR and dissolution studies showed good physical stability of the emulsion powders during accelerated stability testing (50 °C/75% RH, open vial).


Subject(s)
Nanoparticles , Tuberculosis, Oral , Humans , Emulsions , Nanoparticles/chemistry , Solubility , Excipients/chemistry , Water/chemistry , Particle Size
2.
Int J Pharm ; 640: 122985, 2023 Jun 10.
Article in English | MEDLINE | ID: mdl-37121493

ABSTRACT

Flash NanoPrecipitation (FNP) is a scalable, single-step process that uses rapid mixing to prepare nanoparticles with a hydrophobic core and amphiphilic stabilizing shell. Because the two steps of particle self-assembly - (1) core nucleation and growth and (2) adsorption of a stabilizing polymer onto the growing core surface - occur simultaneously during FNP, nanoparticles formulated at core loadings above approximately 70% typically exhibit poor stability or do not form at all. Additionally, a fundamental limit on the concentration of total solids that can be introduced into the FNP process has been reported previously. These limits are believed to share a common mechanism: entrainment of the stabilizing polymer into the growing particle core, leading to destabilization and aggregation. Here, we demonstrate a variation of FNP which separates the nucleation and stabilization steps of particle formation into separate sequential mixers. This scheme allows the hydrophobic core to nucleate and grow in the first mixing chamber unimpeded by adsorption of the stabilizing polymer, which is later introduced to the growing nuclei in the second mixer. Using this Sequential Flash NanoPrecipitation (SNaP) technique, we formulate stable nanoparticles with up to 90% core loading by mass and at 6-fold higher total input solids concentrations than typically reported.


Subject(s)
Nanoparticles , Polymers , Particle Size , Polymers/chemistry , Nanoparticles/chemistry , Hydrophobic and Hydrophilic Interactions
3.
Mol Pharm ; 19(5): 1515-1525, 2022 05 02.
Article in English | MEDLINE | ID: mdl-35412842

ABSTRACT

Nanoparticle encapsulation is an attractive approach to improve the oral bioavailability of hydrophobic therapeutics. The high specific surface area of nanoparticle formulations, combined with the thermodynamically driven increased solubility of an amorphous drug core, promotes rapid drug dissolution. However, the physicochemical properties of the hydrophobic therapeutic can present obstacles to in vitro characterization of nanoparticle formulations. Namely, drugs with low density and high membrane binding affinity frustrate traditional analytical methods to monitor release kinetics from nanoparticles. In this work, cannabidiol (CBD) was encapsulated into nanoparticles with low polydispersity and high drug loading via Flash NanoPrecipitation (FNP), a scalable self-assembly process. Hydroxypropyl methylcellulose acetate succinate (HPMCAS) and lecithin were employed as amphiphilic particle stabilizers during the FNP process. However, the low density and high membrane binding affinity of the amorphous CBD nanoparticle core prevented the characterization of in vitro release kinetics by conventional methods. Released CBD could not be separated from intact nanoparticles by filtration or centrifugation. To address this challenge, an alternative approach is described to coencapsulate 6 nm hydrophobic Fe3O4 colloids with CBD during FNP. The Fe3O4 colloids were added at 33% by mass (approximately 20% by volume) to increase the density of the nanoparticles, resulting in particles with an average diameter of 160 nm (CBD-lecithin-Fe3O4) or 280 nm (CBD-HPMCAS-Fe3O4). This densification enabled the centrifugal separation of dissolved (released) CBD from unreleased CBD during the in vitro assay while avoiding the losses associated with a filtration step. The resulting nanoparticle formulations provided more rapid and complete in vitro dissolution kinetics than bulk CBD, representing a 6-fold improvement in dissolution compared to crystalline CBD. The coencapsulation of high-density Fe3O4 colloids to enable the separation of nanoparticles from release media is a novel approach to measuring in vitro release kinetics of nanoencapsulated low-density, hydrophobic drug molecules.


Subject(s)
Cannabidiol , Nanoparticles , Colloids/chemistry , Lecithins , Nanoparticles/chemistry , Particle Size , Solubility
5.
J Colloid Interface Sci ; 604: 208-220, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34265681

ABSTRACT

HYPOTHESIS: Dynamic Light Scattering (DLS) generated particle size distributions (PSD) of polymer-stabilized nanoparticles are dependent on the optimization parameters used to generate an inversion solution fit to the measured autocorrelation function. The accuracy of the DLS PSD average and polydispersity can be determined by comparing analyzed Transmission Electron Microscopy (TEM) images with the DLS results if the TEM measured sizes can be corrected for the thickness of the hydrated polymer corona that impacts particle hydrodynamics but is a collapsed, desiccated shell in the TEM images. EXPERIMENTS: Nanoparticles were prepared by Flash NanoPrecipitation with either poly(ethylene glycol) (PEG) or hydroxypropyl methylcellulose acetate succinate (HPMCAS) stabilizing polymers. Solvated nanoparticle size distributions were measured by DLS in aqueous media. The same nanoparticle dispersions were lyophilized onto TEM grids and stained by ruthenium tetroxide (RuO4) vapor to improve electron contrast. Desiccated particle size distributions were generated by measuring a minimum of 300 particle diameters in the stained TEM images. FINDINGS: Using our protocol for staining soft matter nanoparticles in TEM measurements, we have quantitatively analyzed the correlation between DLS and TEM generated PSDs. Average diameters disagree by the hydrated polymer corona thickness for each stabilizer due to the high-vacuum TEM environment, with 21.4 nm for PEG and 51.2 nm for HPMCAS. While corrected average diameter agrees within 10% for each technique, DLS consistently over-estimates the standard deviation of the PSD by 100% compared to the TEM measurement.


Subject(s)
Nanoparticles , Polymers , Microscopy, Electron, Transmission , Particle Size , Ruthenium Compounds
6.
Langmuir ; 37(28): 8517-8524, 2021 07 20.
Article in English | MEDLINE | ID: mdl-34236205

ABSTRACT

Chitosan-coated nanoparticles are a promising class of drug delivery vehicles that have been studied as tools for improving the gastrointestinal delivery of therapeutics. Here we present an analysis of chitosan-coated nanoparticles with an emphasis on characterizing the chitosan polymer properties. Cationic nanoparticles are produced by adsorbing a layer of chitosan HCl on an anionic (-40 mV ζ-potential) polyacrylic acid (PAA) coated primary nanoparticle. Commercially available chitosan (90% deacetylated) must be processed into a nearly completely deacetylated HCl salt form (99% deacetylation); otherwise, primary nanoparticle aggregation occurs. Deacetylated chitosan HCl produces stable, cationic (+35 mV ζ-potential) nanoparticles within 10% of the original anionic particle hydrodynamic diameter at a 1:2 molar ratio of chitosan glucosamine HCl monomers to PAA acrylic acid monomers.


Subject(s)
Chitosan , Nanoparticles , Adsorption , Drug Carriers , Drug Delivery Systems , Particle Size , Polyelectrolytes
7.
Mol Pharm ; 18(3): 1093-1101, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33440941

ABSTRACT

Nanocarriers (NCs) are an attractive class of vehicles for drug delivery with the potential to improve drug efficacy and safety, particularly for intravenous parenteral delivery. Many therapeutics remain challenging to formulate in NCs due to their intrinsic solubilities that frustrate NC loading or result in too rapid release in vivo. Therapeutic conjugate approaches that alter the solubility of a conjugate "prodrug" have been used to enable NC formation and controlled release from NCs using labile linker chemistry. A limitation of this approach has been that a different linker chemistry must be used to produce an adjustable release rate for a single therapeutic. We report on a new approach where the therapeutic conjugate hydrolysis rates are varied by adjusting the excipient formulation of the NC core, not the conjugate linker chemistry. A hydrophobic therapeutic conjugate of camptothecin (PROCPT) is synthesized by conjugating camptothecin (CPT) with an acid derivative of α-tocopherol (vitamin E). The PROCPT compound can be loaded to 50% wt in poly(lactic acid)-block-poly(ethylene glycol) (PLA-b-PEG)-stabilized NCs produced by Flash NanoPrecipitation with particle diameters between 60 and 80 nm. Co-loading a zwitterionic lipid, 1-palmitoyl-2-oleoyl-glycero-3-phosphocholine, from 0 to 67% core loading tunes the PROCPT hydrolysis from no observable therapeutic release over 200 h to therapeutic conjugate half-life times of 31 h. For a single therapeutic conjugate molecule, the hydrolysis rate can be tuned by modifying the NC formulation with different excipient concentrations. NCs containing a 50% core loading of PROCPT were lyophilized and encapsulated in a PEG hydrogel matrix to make microparticles for depot delivery with an average diameter of 65 ± 10 µm that provide a sustained, first-order release of CPT with a therapeutic conjugate half-life of 240 h. These results demonstrate a new approach to the formulation of therapeutic NCs with variable release profiles using a single molecular entity therapeutic conjugate.


Subject(s)
Camptothecin/chemistry , Delayed-Action Preparations/chemistry , Drug Carriers/chemistry , Excipients/chemistry , Microgels/chemistry , Nanoparticles/chemistry , Prodrugs/chemistry , Drug Delivery Systems/methods , Drug Liberation/drug effects , Drug Stability , Hydrolysis/drug effects , Hydrophobic and Hydrophilic Interactions , Lactates/chemistry , Polyesters/chemistry , Polyethylene Glycols/chemistry , Solubility/drug effects , alpha-Tocopherol/chemistry
8.
J Colloid Interface Sci ; 582(Pt B): 815-824, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-32911422

ABSTRACT

HYPOTHESIS: Hydrophobic ion pairing (HIP), a solubility engineering technique in which ionic hydrophilic molecules are paired with a hydrophobic counterion, is an attractive strategy for encapsulating ionic water-soluble species into nanocarriers (NCs). Drug release from NCs containing HIP complexes is sensitive to ionic strength, pH, and drug:counterion charge ratio, but the exact mechanism for this was unknown, as was the underlying microstructure inside the NCs. We hypothesize that HIP complexes arrange into liquid crystalline structures in NC cores and that these structures are responsible for salt- and pH-dependent release. EXPERIMENT: A model hydrophobic ion pair from the cationic antimicrobial peptide polymyxin B sulfate and the anionic counterion sodium oleate is encapsulated into ~100 nm NCs formed using Flash NanoPrecipitation (FNP) and stabilized with an amphiphilic diblock copolymer, poly(caprolactone)-b-poly(ethylene glycol). Internal structures are observed using synchrotron small angle X-ray scattering (SAXS) and transmission electron microscopy (TEM) following NC formulation and are found to vary with polymyxin:oleate charge ratio. In vitro drug release is also measured with changes in pH and two charge ratio. FINDINGS: For a formulation containing a four-fold charge excess of oleate relative to polymyxin, internal structures rearranged from a lamellar phase into an inverse hexagonal phase. The hexagonal phase formation corresponds to a greatly reduced rate of polymyxin release, suggesting that the polymyxin was incorporated into the center of hexagonally-packed rods. When release tests were repeated using phosphate-buffered saline (PBS) at pH 2.0 to ensure protonation of the oleic acid, all internal structures were eliminated and release occurs much faster than at neutral pH, regardless of charge ratio. These findings shed light on the mechanism behind stimulus-responsive drug release from systems containing hydrophobic ion pairs and enable the rational design of controlled-release formulations by manipulating the formation and dynamics of liquid crystalline phases inside NCs.


Subject(s)
Liquid Crystals , Delayed-Action Preparations , Drug Liberation , Hydrophobic and Hydrophilic Interactions , Scattering, Small Angle , X-Ray Diffraction
9.
J Vis Exp ; (143)2019 01 07.
Article in English | MEDLINE | ID: mdl-30663705

ABSTRACT

The formulation of a therapeutic compound into nanoparticles (NPs) can impart unique properties. For poorly water-soluble drugs, NP formulations can improve bioavailability and modify drug distribution within the body. For hydrophilic drugs like peptides or proteins, encapsulation within NPs can also provide protection from natural clearance mechanisms. There are few techniques for the production of polymeric NPs that are scalable. Flash NanoPrecipitation (FNP) is a process that uses engineered mixing geometries to produce NPs with narrow size distributions and tunable sizes between 30 and 400 nm. This protocol provides instructions on the laboratory-scale production of core-shell polymeric nanoparticles of a target size using FNP. The protocol can be implemented to encapsulate either hydrophilic or hydrophobic compounds with only minor modifications. The technique can be readily employed in the laboratory at milligram scale to screen formulations. Lead hits can directly be scaled up to gram- and kilogram-scale. As a continuous process, scale-up involves longer mixing process run time rather than translation to new process vessels. NPs produced by FNP are highly loaded with therapeutic, feature a dense stabilizing polymer brush, and have a size reproducibility of ± 6%.


Subject(s)
Chemical Precipitation , Hydrophobic and Hydrophilic Interactions , Nanoparticles/chemistry , Polymers/chemistry , Particle Size , Polyethylene Glycols/chemistry , Reproducibility of Results , Solvents , Vitamin E/chemistry , Water
10.
ACS Appl Mater Interfaces ; 10(4): 3191-3199, 2018 Jan 31.
Article in English | MEDLINE | ID: mdl-29272577

ABSTRACT

Nanoparticles (NP) are promising contrast agents for positron emission tomography (PET) radionuclide imaging that can increase signal intensity by localizing clusters of PET radionuclides together. However, methods to load NPs with PET radionuclides suffer from harsh loading conditions or poor loading efficacies or result in NP surface modifications that alter targeting in vivo. We present the formation of water-dispersible, polyethylene glycol coated NPs that encapsulate phthalocyanines into NP cores at greater than 50 wt % loading, using the self-assembly technique Flash NanoPrecipitation. Particles from 70 to 160 nm are produced. Phthalocyanine NPs rapidly and spontaneously chelate metals under mild conditions and can act as sinks for PET radionuclides such as 64-Cu to produce PET-active NPs. NPs chelate copper(II) with characteristic rates of 1845 M-1 h-1 at pH 6 and 37 °C, which produced >90% radionuclide chelation within 1 h. NP physical properties, such as core composition, core fluidity, and size, can be tuned to modulate chelation kinetics. These NPs retain 64Cu even in the presence of the strong chelator ethylene diamine tetraacetic acid. The development of these constructs for rapid and facile radionuclide labeling expands the applications of NP-based PET imaging.


Subject(s)
Nanoparticles , Copper , Copper Radioisotopes , Positron-Emission Tomography
11.
ACS Biomater Sci Eng ; 3(3): 443-451, 2017 Mar 13.
Article in English | MEDLINE | ID: mdl-33465939

ABSTRACT

Photoacoustic (PA) imaging is a developing diagnostic technique where multiple species can be simultaneously imaged with high spatial resolution in 3D if the absorbance spectrum of each species is distinct and separable. However, multiplexed PA imaging has been greatly limited by the availability of spectrally separable contrast agents that can be used in vivo. Toward this end, we present the formation and application of a series of poly ethylene glycol (PEG)-coated nanoparticles (NPs) with unique separable absorbance profiles suitable for simultaneous multiplexed imaging. As a proof-of-concept, we demonstrate this form of mixed-sample multiplexed imaging, using cRGD peptide surface-modified NPs with nonmodified NPs in a murine subcutaneous Lewis lung carcinoma tumor model. The simultaneous imaging of nonmodified NPs provides an "internal standard", to deconvolute the contributions of active-ligand and passive-NP targeting effects. Particles with 25% surface cRGD modification display 52 ± 22 fold higher liver to tumor ratio accumulation levels, while the same set of particles display only 9.8 ± 4 fold accumulation levels when internally normalized. The pharmacokinetic profiles of targeted and nontargeted NPs can be simultaneously tracked in real-time to study how biodistribtions of particles are affected by ligand modification. The internal normalization of control particles greatly enhances the precision and decreases the number of animals needed in studies of nanoparticle targeting. These new dyes are an enabling technology for PA imaging of NP fate and targeting. This is the first demonstration of real-time multiplexed PA imaging of mixed-targeted samples in vivo.

12.
ACS Appl Mater Interfaces ; 8(23): 14379-88, 2016 Jun 15.
Article in English | MEDLINE | ID: mdl-27153806

ABSTRACT

Photoacoustic (PA) imaging is an emerging hybrid optical-ultrasound based imaging technique that can be used to visualize optical absorbers in deep tissue. Free organic dyes can be used as PA contrast agents to concurrently provide additional physiological and molecular information during imaging, but their use in vivo is generally limited by rapid renal clearance for soluble dyes and by the difficulty of delivery for hydrophobic dyes. We here report the use of the block copolymer directed self-assembly process, Flash NanoPrecipitation (FNP), to form series of highly hydrophobic optical dyes into stable, biocompatible, and water-dispersible nanoparticles (NPs) with sizes from 38 to 88 nm and with polyethylene glycol (PEG) surface coatings suitable for in vivo use. The incorporation of dyes with absorption profiles within the infrared range, that is optimal for PA imaging, produces the PA activity of the particles. The hydrophobicity of the dyes allows their sequestration in the NP cores, so that they do not interfere with targeting, and high loadings of >75 wt % dye are achieved. The optical extinction coefficients (ε (mL mg(-1) cm(-1))) were essentially invariant to the loading of the dye in NP core. Co-encapsulation of dye with vitamin E or polystyrene demonstrates the ability to simultaneously image and deliver a second agent. The PEG chains on the NP surface were functionalized with folate to demonstrate folate-dependent targeting. The spectral separation of different dyes among different sets of particles enables multiplexed imaging, such as the simultaneous imaging of two sets of particles within the same animal. We provide the first demonstration of this capability with PA imaging, by simultaneously imaging nontargeted and folate-targeted nanoparticles within the same animal. These results highlight Flash NanoPrecipitation as a platform to develop photoacoustic tools with new diagnostic capabilities.


Subject(s)
Diagnostic Imaging/methods , Nanoparticles/chemistry , Photoacoustic Techniques , Animals , Infrared Rays , Polyethylene Glycols/chemistry , Polymers/chemistry
13.
J Pharm Sci ; 101(10): 3763-78, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22777686

ABSTRACT

Monoclonal antibodies continue to command a large market for treatment of a variety of diseases. In many cases, the doses required for therapeutic efficacy are large, limiting options for antibody delivery and administration. We report a novel formulation strategy based on dispersions of antibody nanoclusters that allows for subcutaneous injection of highly concentrated antibody (≈ 190 mg/mL). A solution of monoclonal antibody 1B7 was rapidly frozen and lyophilized using a novel spiral-wound in-situ freezing technology to generate amorphous particles. Upon gentle stirring, a translucent dispersion of approximately 430 nm protein clusters with low apparent viscosity (≈ 24 cp) formed rapidly in buffer containing the pharmaceutically acceptable crowding agents such as trehalose, polyethylene glycol, and n-methyl-2-pyrrolidone. Upon in vitro dilution of the dispersion, the nanoclusters rapidly reverted to monomeric protein with full activity, as monitored by dynamic light scattering and antigen binding. When administered to mice as an intravenous solution, subcutaneous solution, or subcutaneous dispersion at similar (4.6-7.3 mg/kg) or ultra-high dosages (51.6 mg/kg), the distribution and elimination kinetics were within error and the protein retained full activity. Overall, this method of generating high-concentration, low-viscosity dispersions of antibody nanoclusters could lead to improved administration and patient compliance, providing new opportunities for the biotechnology industry.


Subject(s)
Antibodies, Monoclonal/chemistry , Nanoparticles/chemistry , Pharmaceutical Solutions/chemistry , Proteins/chemistry , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Biological Availability , CHO Cells , Cell Line , Chemistry, Pharmaceutical/methods , Cricetinae , Drug Stability , Female , Injections, Subcutaneous , Mice , Mice, Inbred BALB C , Nanoparticles/administration & dosage , Pharmaceutical Solutions/administration & dosage , Pharmaceutical Solutions/pharmacokinetics , Proteins/administration & dosage , Proteins/pharmacokinetics , Solutions/administration & dosage , Solutions/chemistry , Solutions/pharmacokinetics , Viscosity
14.
ACS Nano ; 6(2): 1357-69, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22260218

ABSTRACT

Stabilizing proteins at high concentration is of broad interest in drug delivery, for treatment of cancer and many other diseases. Herein, we create highly concentrated antibody dispersions (up to 260 mg/mL) comprising dense equilibrium nanoclusters of protein (monoclonal antibody 1B7, polyclonal sheep immunoglobulin G, and bovine serum albumin) molecules which, upon dilution in vitro or administration in vivo, remain conformationally stable and biologically active. The extremely concentrated environment within the nanoclusters (∼700 mg/mL) provides conformational stability to the protein through a novel self-crowding mechanism, as shown by computer simulation, while the primarily repulsive nanocluster interactions result in colloidally stable, transparent dispersions. The nanoclusters are formed by adding trehalose as a cosolute which strengthens the short-ranged attraction between protein molecules. The protein cluster diameter was reversibly tuned from 50 to 300 nm by balancing short-ranged attraction against long-ranged electrostatic repulsion of weakly charged protein at a pH near the isoelectric point. This behavior is described semiquantitatively with a free energy model which includes the fractal dimension of the clusters. Upon dilution of the dispersion in vitro, the clusters rapidly dissociated into fully active protein monomers as shown with biophysical analysis (SEC, DLS, CD, and SDS-PAGE) and sensitive biological assays. Since the concept of forming nanoclusters by tuning colloid interactions is shown to be general, it is likely applicable to a variety of biological therapeutics, mitigating the need to engineer protein stability through amino acid modification. In vivo subcutaneous injection into mice results in indistinguishable pharmacokinetics versus a standard antibody solution. Stable protein dispersions with low viscosities may potentially enable patient self-administration by subcutaneous injection of antibody therapeutics being discovered and developed.


Subject(s)
Drug Carriers/chemistry , Nanostructures/chemistry , Proteins/chemistry , Animals , Buffers , Cattle , Drug Carriers/pharmacokinetics , Mice , Models, Molecular , Protein Conformation , Protein Stability , Trehalose/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...