Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Adv Healthc Mater ; 13(11): e2303910, 2024 04.
Article in English | MEDLINE | ID: mdl-38180445

ABSTRACT

Self-assembling protein nanoparticles are a promising class of materials for targeted drug delivery. Here, the use of a computationally designed, two-component, icosahedral protein nanoparticle is reported to encapsulate multiple macromolecular cargoes via simple and controlled self-assembly in vitro. Single-stranded RNA molecules between 200 and 2500 nucleotides in length are encapsulated and protected from enzymatic degradation for up to a month with length-dependent decay rates. Immunogenicity studies of nanoparticles packaging synthetic polymers carrying a small-molecule TLR7/8 agonist show that co-delivery of antigen and adjuvant results in a more than 20-fold increase in humoral immune responses while minimizing systemic cytokine secretion associated with free adjuvant. Coupled with the precise control over nanoparticle structure offered by computational design, robust and versatile encapsulation via in vitro assembly opens the door to a new generation of cargo-loaded protein nanoparticles that can combine the therapeutic effects of multiple drug classes.


Subject(s)
Nanoparticles , Nanoparticles/chemistry , Animals , Mice , Proteins/chemistry , Toll-Like Receptor 8/metabolism , Toll-Like Receptor 8/agonists , Toll-Like Receptor 8/chemistry , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 7/agonists
2.
Adv Ther (Weinh) ; 6(12)2023 Dec.
Article in English | MEDLINE | ID: mdl-38464558

ABSTRACT

Following a traumatic brain injury (TBI), excess reactive oxygen species (ROS) and lipid peroxidation products (LPOx) are generated and lead to secondary injury beyond the primary insult. A major limitation of current treatments is poor target engagement, which has prevented success in clinical trials. Thus, nanoparticle-based treatments have received recent attention because of their ability to increase accumulation and retention in damaged brain. Theranostic neuroprotective copolymers (NPC3) containing thiol functional groups can neutralize ROS and LPOx. Immediate administration of NPC3 following injury in a controlled cortical impact (CCI) mouse model provides a therapeutic window in reducing ROS levels at 2.08-20.83 mg/kg in males and 5.52-27.62 mg/kg in females. This NPC3-mediated reduction in oxidative stress improves spatial learning and memory in males, while females show minimal improvement. Notably, NPC3-mediated reduction in oxidative stress prevents the bilateral spread of necrosis in male mice, which was not observed in female mice and likely accounts for the sex-based spatial learning and memory differences. Overall, these findings suggest sex-based differences to oxidative stress scavenger nanoparticle treatments, and a possible upper threshold of antioxidant activity that provides therapeutic benefit in injured brain since female mice benefit from NPC3 treatment to a lesser extent than male mice.

3.
Biomacromolecules ; 23(4): 1703-1712, 2022 04 11.
Article in English | MEDLINE | ID: mdl-35316025

ABSTRACT

Traumatic brain injury (TBI) results in the generation of reactive oxygen species (ROS) and lipid peroxidation product (LPOx), including acrolein and 4-hydroxynonenal (4HNE). The presence of these biochemical derangements results in neurodegeneration during the secondary phase of the injury. The ability to rapidly neutralize multiple species could significantly improve outcomes for TBI patients. However, the difficulty in creating therapies that target multiple biochemical derangements simultaneously has greatly limited therapeutic efficacy. Therefore, our goal was to design a material that could rapidly bind and neutralize both ROS and LPOx following TBI. To do this, a series of thiol-functionalized biocompatible copolymers based on lipoic acid methacrylate and polyethylene glycol monomethyl ether methacrylate (FW ∼ 950 Da) (O950) were prepared. A polymerizable gadolinium-DOTA methacrylate monomer (Gd-MA) was also synthesized starting from cyclen to facilitate direct magnetic resonance imaging and in vivo tracking of accumulation. These neuroprotective copolymers (NPCs) were shown to rapidly and effectively neutralize both ROS and LPOx. Horseradish peroxidase absorbance assays showed that the NPCs efficiently neutralized H2O2, while R-phycoerythrin protection assays demonstrated their ability to protect the fluorescent protein from oxidative damage. 1H NMR studies indicated that the thiol-functional NPCs rapidly form covalent bonds with acrolein, efficiently removing it from solution. In vitro cell studies with SH-SY5Y-differentiated neurons showed that NPCs provide unique protection against toxic concentrations of both H2O2 and acrolein. NPCs rapidly accumulate and are retained in the injured brain in controlled cortical impact mice and reduce post-traumatic oxidative stress. Therefore, these materials show promise for improved target engagement of multiple biochemical derangements in hopes of improving TBI therapeutic outcomes.


Subject(s)
Acrolein , Brain Injuries, Traumatic , Acrolein/pharmacology , Animals , Brain Injuries, Traumatic/drug therapy , Humans , Hydrogen Peroxide/pharmacology , Lipid Peroxidation/physiology , Methacrylates/pharmacology , Mice , Oxidative Stress , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/pharmacology , Theranostic Nanomedicine
4.
Biomaterials ; 272: 120766, 2021 05.
Article in English | MEDLINE | ID: mdl-33819812

ABSTRACT

The secondary phase of traumatic brain injury (TBI) is partly caused by the release of excess reactive oxygen species (ROS) from the primary injury. However, there are currently no therapies that have been shown to reduce the secondary spread of injury beyond the primary insult. Nanoparticles offer the ability to rapidly accumulate and be retained in injured brain for improved target engagement. Here, we utilized systemically administered antioxidant thioether core-cross-linked nanoparticles (NP1) that scavenge and inactivate ROS to reduce this secondary spread of injury in a mild controlled cortical impact (CCI) mouse model of TBI. We found that NP1 treatment protected CCI mice from injury induced learning and memory deficits observed in the Morris water maze (MWM) test at 1-month post-CCI. This protection was likely a result of NP1-mediated reduction in oxidative stress in the ipsilateral hemisphere as determined by immunofluorescence imaging of markers of oxidative stress and the spread of neuroinflammation into the contralateral hippocampus as determined by immunofluorescence imaging of activated microglia and neuron-astrocyte-microglia triad formation. These data suggest NP1-mediated reduction in post-traumatic oxidative stress correlates with the reduction in the spread of injury to the contralateral hippocampus to protect spatial memory and learning in CCI mice. Therefore, these materials may offer an improved treatment strategy to reduce the secondary spread of TBI.


Subject(s)
Brain Injuries, Traumatic , Nanoparticles , Animals , Antioxidants , Brain Injuries, Traumatic/drug therapy , Disease Models, Animal , Maze Learning , Mice , Spatial Learning , Sulfides
5.
ACS Infect Dis ; 6(11): 2866-2871, 2020 11 13.
Article in English | MEDLINE | ID: mdl-33074651

ABSTRACT

Biofilms are one of the most challenging obstacles in bacterial infections. By providing protection against immune responses and antibiotic therapies, biofilms enable chronic colonization and the development of antibiotic resistance. As previous clinical observations and studies have shown, traditional antibiotic therapy alone cannot effectively treat and eliminate biofilm forming infections due to the protection conferred by the biofilm. A new strategy specifically targeting biofilms must be developed. Here, we specifically target and bind to the PAO1 biofilm and elucidate the molecular mechanism behind the interaction between a glycan targeted polymer and biofilm using a continuous flow biofilm model. The incubation of biofilms with fluorescent glycan targeted polymers demonstrated strong and persistent interactions with the mannose-containing polymer even after 24 h of continuous flow. To evaluate the role of major biofilm proteins LecB and CdrA, loss of function experiments with knockout variants established the dual involvement of both proteins in mannose targeted polymer retention. These results identify a persistent and specific targeting strategy to the biofilm, emphasizing its potential value as a delivery strategy and encouraging further exploration of biofilm targeted delivery.


Subject(s)
Mannose , Pseudomonas aeruginosa , Bacterial Proteins , Biofilms , Polymers
6.
Biomaterials ; 195: 38-50, 2019 03.
Article in English | MEDLINE | ID: mdl-30610992

ABSTRACT

Alveolar macrophages resident in the lung are prominent phagocytic effector cells of the pulmonary innate immune response, and paradoxically, are attractive harbors for pathogens. Consequently, facultative intracellular bacteria, such as Francisella tularensis, can cause severe systemic disease and sepsis, with high morbidity and mortality associated with pulmonary infection. Current clinical treatment, which involves exhaustive oral or intravenous antibiotic therapy, has limitations such as systemic toxicity and off-target effects. Pulmonary administration represents a promising alternative to systemic dosing for delivering antibiotics directly to the lung. Here, we present synthesized mannosylated ciprofloxacin polymeric prodrugs for efficient pulmonary delivery, targeting, and subsequent internalization by alveolar macrophages. We demonstrate significant improvement in efficacy against intracellular infections in an otherwise uniformly lethal airborne Francisella murine model (F. novicida). When administered to the lungs of mice in a prophylactic regimen, the mannosylated ciprofloxacin polymeric prodrugs led to 50% survival. In a treatment regimen that was concurrent with infection, the survival of mice increased to 87.5%. Free ciprofloxacin antibiotic was ineffective in both cases. This significant difference in antibacterial efficacy demonstrates the impact of this delivery platform based on improved physiochemical, pharmacokinetic, and pharmacodynamic properties of ciprofloxacin administered via our glycan polymeric prodrug. This modular platform provides a route for overcoming the limitations of free drug and increasing efficacy in treatment of intracellular infection.


Subject(s)
Macrophages, Alveolar/metabolism , Polysaccharides/chemistry , Prodrugs/chemistry , Francisella tularensis/metabolism , Magnetic Resonance Spectroscopy , Mannose/metabolism , Microbial Sensitivity Tests
7.
J Control Release ; 287: 1-11, 2018 10 10.
Article in English | MEDLINE | ID: mdl-30099019

ABSTRACT

Intracellular bacterial infections localized to the lung alveolar macrophage (AM) remain one of the most challenging settings for antimicrobial therapy. Current systemic antibiotic treatment fails to deliver sustained doses to intracellular bacterial reservoirs, which necessitates prolonged treatment regimens. Herein, we demonstrate a new intracellular enzyme-cleavable polymeric prodrug with tailored ciprofloxacin release profiles in the lungs and AM. The targeted polymeric prodrug, termed "drugamers", incorporates (1) hydrophilic mannose residues to solubilize the antibiotic cargo and to target and enhance AM uptake and intracellular delivery, and (2) enzyme-cleavable linkage chemistry to provide high and sustained intracellular AM drug dosing. Prodrug monomers, derived from the antibiotic ciprofloxacin, were synthesized with either an intracellular protease cleavable dipeptide linker or a hydrolytic phenyl ester linker. RAFT polymerization was used to copolymerize the prodrug monomers and mannose monomer to synthesize well-defined drugamers without requiring a post-polymerization conjugation step. In addition to favorable in vivo safety profiles following intratracheal administration, a single dose of the drugamers sustained ciprofloxacin dosing in lungs and AMs above the minimum inhibitory concentration (MIC) over at least a 48 h period. The enzyme-cleavable therapeutic achieved a >10-fold increase in sustained ciprofloxacin in AM, and maintained a significantly higher whole lung PK as well. Ciprofloxacin dosed in identical fashion displayed rapid clearance with a half-life of approximately 30 min. Notably, inhalation of the mannose-targeted ciprofloxacin drugamers achieved full survival (100%) in a highly lethal mouse model of pneumonic tularemia, contrasted with 0% survival using free ciprofloxacin. These findings demonstrate the versatility of the drugamer platform for engineering the intracellular pharmacokinetic profiles and its strong therapeutic activity in treating pulmonary intracellular infections.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Ciprofloxacin/administration & dosage , Delayed-Action Preparations/chemistry , Francisella/drug effects , Gram-Negative Bacterial Infections/drug therapy , Lung Diseases/drug therapy , Administration, Inhalation , Animals , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Ciprofloxacin/pharmacokinetics , Ciprofloxacin/pharmacology , Ciprofloxacin/therapeutic use , Drug Delivery Systems , Female , Lung/drug effects , Lung/metabolism , Lung Diseases/metabolism , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Mannose/analogs & derivatives , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Polymers/chemistry , RAW 264.7 Cells , Tularemia/drug therapy
8.
Biomater Sci ; 6(7): 1976-1985, 2018 Jun 25.
Article in English | MEDLINE | ID: mdl-29850694

ABSTRACT

Pulmonary intracellular infections, such as tuberculosis, anthrax, and tularemia, have remained a significant challenge to conventional antibiotic therapy. Ineffective antibiotic treatment of these infections can lead not only to undesired side effects, but also to the emergence of antibiotic resistance. Aminoglycosides (e.g., streptomycin) have long been part of the therapeutic regiment for many pulmonary intracellular infections. Their bioavailability for intracellular bacterial pools, however, is limited by poor membrane permeability and rapid elimination. To address this challenge, polymer-augmented liposomes (PALs) were developed to provide improved cytosolic delivery of streptomycin to alveolar macrophages, an important host cell for intracellular pathogens. A multifunctional diblock copolymer was engineered to functionalize PALs with carbohydrate-mediated targeting, pH-responsive drug release, and endosomal release activity with a single functional polymer that replaces the pegylated lipid component to simplify the liposome formulation. The pH-sensing functionality enabled PALs to provide enhanced release of streptomycin under endosomal pH conditions (70% release in 6 hours) with limited release at physiological pH 7.4 (16%). The membrane-destabilizing activity connected to endosomal release was characterized in a hemolysis assay and PALs displayed a sharp pH profile across the endosomal pH development target range. The direct connection of this membrane-destabilizing pH profile to model drug release was demonstrated in an established pyranine/p-xylene bispyridinium dibromide (DPX) fluorescence dequenching assay. PALs displayed similar sharp pH-responsive release, whereas PEGylated control liposomes did not, and similar profiles were then shown for streptomycin release. The mannose-targeting capability of the PALs was also demonstrated with 2.5 times higher internalization compared to non-targeted PEGylated liposomes. Finally, the streptomycin-loaded PALs were shown to have a significantly improved intracellular antibacterial activity in a Francisella-macrophage co-culture model, compared with free streptomycin or streptomycin delivered by control PEGylated liposomes (13× and 16×, respectively). This study suggests the potential of PALs as a useful platform to deliver antibiotics for the treatment of intracellular macrophage infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Delivery Systems/methods , Francisella tularensis/drug effects , Liposomes/pharmacology , Streptomycin/pharmacology , Animals , Anti-Bacterial Agents/metabolism , Arylsulfonates/chemistry , Drug Compounding/methods , Drug Liberation , Endosomes/drug effects , Endosomes/metabolism , Endosomes/microbiology , Fluorescent Dyes/chemistry , Francisella tularensis/growth & development , Francisella tularensis/metabolism , Hydrogen-Ion Concentration , Kinetics , Liposomes/chemical synthesis , Liposomes/metabolism , Mannose/metabolism , Methacrylates/chemistry , Mice , Phosphatidylcholines/chemistry , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemistry , Pyridinium Compounds/chemistry , RAW 264.7 Cells , Streptomycin/metabolism
9.
Polym Chem ; 9(42): 5224-5233, 2018 Nov 14.
Article in English | MEDLINE | ID: mdl-36660314

ABSTRACT

Herein, we developed a fully polymerizable, peptide-targeted, camptothecin polymeric prodrug system. Two prodrug monomers were synthesized via esterification of campothecin (20Cam) and 10-hydroxycamptothecin (10Cam) with mono-2-(methacryloyloxy)ethyl succinate (SMA) resulting in polymerizable forms of the aliphatic ester- and aromatic ester-linked drugs respectively. These monomers were then incorporated into zwitterionic polymers via RAFT copolymerization of the prodrug monomers with a tert-butyl ester protected carboxy betaine monomer. Subsequent deprotection of the tert-butyl residues with TFA yielded carboxy betaine methacrylate (CBM) scaffolds with controlled prodrug incorporation. Reverse phase HPLC was then employed to establish drug release kinetics in human serum at 37 oC for the resultant polymeric prodrugs. Copolymers containing 10Cam residues linked via aromatic esters showed faster hydrolysis rates with 59 % drug released at 7 days, while copolymers with Cam residues linked via aliphatic esters showed only 28 % drug release over the same time period. These differences in drug release kinetics were then shown to correlate with large differences in cytotoxic activity in SKOV3 ovarian cancer cell cultures. At 72 hours, the IC50s of aromatic- and aliphatic- ester linked prodrugs were 56 nM and 4776 nM, respectively. An EGFR-targeting peptide sequence, GE11, was then directly incorporated into the polymeric prodrugs via RAFT copolymerization of the polymeric prodrugs with a peptide macronomer. The GE11-targeted polymeric prodrugs showed enhanced targeting and cytotoxic activity in SKOV3 cell cultures relative to untargeted polymers containing the negative control sequence HW12. Following pulse-chase treatment (15 min, 37 °C), the 72 hour IC50 of GE11 targeted prodrug was determined to be 1597 nM, in contrast to 3399 nM for the non-targeted control.

10.
ACS Nano ; 11(9): 8600-8611, 2017 09 26.
Article in English | MEDLINE | ID: mdl-28783305

ABSTRACT

Traumatic brain injury (TBI) is the leading cause of death and disability in children and young adults, yet there are currently no treatments available that prevent the secondary spread of damage beyond the initial insult. The chronic progression of this secondary injury is in part caused by the release of reactive oxygen species (ROS) into surrounding normal brain. Thus, treatments that can enter the brain and reduce the spread of ROS should improve outcome from TBI. Here a highly versatile, reproducible, and scalable method to synthesize core-cross-linked nanoparticles (NPs) from polysorbate 80 (PS80) using a combination of thiol-ene and thiol-Michael chemistry is described. The resultant NPs consist of a ROS-reactive thioether cross-linked core stabilized in aqueous solution by hydroxy-functional oligoethylene oxide segments. These NPs show narrow molecular weight distributions and have a high proportion of thioether units that reduce local levels of ROS. In a controlled cortical impact mouse model of TBI, the NPs are able to rapidly accumulate and be retained in damaged brain as visualized through fluorescence imaging, reduce neuroinflammation and the secondary spread of injury as determined through magnetic resonance imaging and histopathology, and improve functional outcome as determined through behavioral analyses. Our findings provide strong evidence that these NPs may, upon further development and testing, provide a useful strategy to help improve the outcome of patients following a TBI.


Subject(s)
Antioxidants/therapeutic use , Brain Injuries, Traumatic/therapy , Nanoparticles/therapeutic use , Reactive Oxygen Species/metabolism , Sulfides/therapeutic use , Animals , Antioxidants/chemistry , Brain/metabolism , Brain/pathology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Inflammation/metabolism , Inflammation/pathology , Inflammation/therapy , Mice , Mice, Inbred C57BL , Nanomedicine/methods , Nanoparticles/chemistry , Oxidative Stress/drug effects , Polysorbates/chemistry , Polysorbates/therapeutic use , RAW 264.7 Cells , Sulfides/chemistry
11.
Mol Pharm ; 14(6): 1988-1997, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28394614

ABSTRACT

Lung-based intracellular bacterial infections remain one of the most challenging infectious disease settings. For example, the current standard for treating Franciscella tularensis pneumonia (tularemia) relies on administration of oral or intravenous antibiotics that poorly achieve and sustain pulmonary drug bioavailability. Inhalable antibiotic formulations are approved and in clinical development for upper respiratory infections, but sustained drug dosing from inhaled antibiotics against alveolar intracellular infections remains a current unmet need. To provide an extended therapy against alveolar intracellular infections, we have developed a macromolecular therapeutic platform that provides sustained local delivery of ciprofloxacin with controlled dosing profiles. Synthesized using RAFT polymerization, these macromolecular prodrugs characteristically have high drug loading (16-17 wt % drug), tunable hydrolysis kinetics mediated by drug linkage chemistry (slow-releasing alkyllic vs fast-releasing phenolic esters), and, in general, represent new fully synthetic nanotherapeutics with streamlined manufacturing profiles. In aerosolized and completely lethal F.t. novicida mouse challenge models, the fast-releasing ciprofloxacin macromolecular prodrug provided high cure efficiencies (75% survival rate under therapeutic treatment), and the importance of release kinetics was demonstrated by the inactivity of the similar but slow-releasing prodrug system. Pharmacokinetics and biodistribution studies further demonstrated that the efficacious fast-releasing prodrug retained drug dosing in the lung above the MIC over a 48 h period with corresponding Cmax/MIC and AUC0-24h/MIC ratios being greater than 10 and 125, respectively; the thresholds for optimal bactericidal efficacy. These findings identify the macromolecular prodrug platform as a potential therapeutic system to better treat alveolar intracellular infections such as F. tularensis, where positive patient outcomes require tailored antibiotic pharmacokinetic and treatment profiles.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Ciprofloxacin/therapeutic use , Administration, Intranasal , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacokinetics , Ciprofloxacin/administration & dosage , Ciprofloxacin/pharmacokinetics , Disease Models, Animal , Female , Francisella tularensis/drug effects , Francisella tularensis/pathogenicity , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Tissue Distribution
12.
Mol Pharm ; 14(5): 1450-1459, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28277671

ABSTRACT

Peptides derived from the third Bcl-2 homology domain (BH3) renormalize apoptotic signaling by antagonizing prosurvival Bcl-2 family members. These potential peptide drugs exhibit therapeutic activities but are limited by barriers including short circulation half-lives and poor penetration into cells. A diblock polymeric micelle carrier for the BIM BH3 peptide was recently described that demonstrated antitumor activity in a B-cell lymphoma xenograft model [Berguig et al., Mol. Ther. 2015, 23, 907-917]. However, the disulfide linkage used to conjugate the BIM peptide was shown to have nonoptimal blood stability. Here we describe a peptide macromonomer composed of BIM capped with a four amino acid cathepsin B substrate (FKFL) that possesses high blood stability and is cleaved to release the drug inside of target cells. Employing RAFT polymerization, the peptide macromonomer was directly integrated into a multifunctional diblock copolymer tailored for peptide delivery. The first polymer block was made as a macro-chain transfer agent (CTA) and composed of a pH-responsive endosomolytic formulation of N,N-diethylaminoethyl methacrylate (DEAEMA) and butyl methacrylate (BMA). The second polymer block was a copolymer of the peptide and polyethylene glycol methacrylate (PEGMA). PEGMA monomers of two sizes were investigated (300 Da and 950 Da). Protein gel analysis, high performance liquid chromatography, and coupled mass spectrometry (MS) showed that incubation with cathepsin B specifically cleaved the FKFL linker and released active BIM peptide with PEGMA300 but not with PEGMA950. MALDI-TOF MS showed that incubation of the peptide monomers alone in human serum resulted in partial cleavage at the FKFL linker after 12 h. However, formulation of the peptides into polymers protected against serum-mediated peptide degradation. Dynamic light scattering (DLS) demonstrated pH-dependent micelle disassembly (25 nm polymer micelles at pH 7.4 versus 6 nm unimers at pH 6.6), and a red blood cell lysis assay showed a corresponding increase in membrane destabilizing activity (<1% lysis at pH 7.4 versus 95% lysis at pH 6.6). The full carrier-drug system successfully induced apoptosis in SKOV3 ovarian cancer cells in a dose-dependent manner, in comparison to a control polymer containing a scrambled BIM peptide sequence. Mechanistic analysis verified target-dependent activation of caspase 3/7 activity (8.1-fold increase), and positive annexin V staining (72% increase). The increased blood stability of this enzyme-cleavable peptide polymer design, together with the direct polymerization approach that eliminated postsynthetic conjugation steps, suggests that this new carrier design could provide important benefits for intracellular peptide drug delivery.


Subject(s)
Micelles , Peptides/chemistry , Polymers/chemistry , Cathepsin B/chemistry , Cell Line, Tumor , Drug Carriers/chemistry , Drug Delivery Systems , Humans , Hydrogen-Ion Concentration , Methacrylates/chemistry , Polyethylene Glycols/chemistry
13.
Langmuir ; 32(36): 9286-92, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27556595

ABSTRACT

Copolymers with well-defined architectures, controlled molecular weights, and narrow molar mass dispersities (D) were synthesized by reversible addition-fragmentation chain transfer (RAFT) polymerization. The resultant polymers contain different combinations of the pH-responsive monomer 2-(diethylaminoethyl) methacrylate (DEAEMA), the hydrophobic comonomer butyl methacrylate (BMA), and a neutral hydrophilic stabilizing monomer polyethylene glycol monomethyl ether methacrylate (designated O950). Surface tension and cryo-TEM measurements of native and heavy-atom stained samples were used to characterize the pH and salt responsiveness of the different polymers as a function of their composition. These studies indicate that while the polymers predominately self-assemble to form spherical micelles, a narrow size distribution is observed in aqueous solutions of poly(O950)-b-(BMA) and poly(O950)-b-(DEAEMA-co-BMA), whereas a broad size distribution characterizes the assemblies of poly(O950)-b-(DEAEMA) and poly(DEAEMA-co-BMA). In the latter case, micelles having diameters around 15-25 nm are found along with smaller aggregates (about 10 nm) mostly arranged in elongated necklace-like structures. The pH and salt-responsiveness of the DEAEMA residue, as indicated by the surface activity of the copolymers, was found to depend on the nature of the additional components: covalently linked hydrophobic groups (BMA) moderated the pH response of the copolymer as compared to nonionic and hydrophilic groups as in poly(O950)-b-(DEAEMA). These results suggest that mutual interactions among the building blocks of self-assembling copolymers should be taken into account when designing responsive copolymers.

14.
Nanomedicine ; 12(7): 2031-2041, 2016 10.
Article in English | MEDLINE | ID: mdl-27184097

ABSTRACT

Carbohydrate receptors on alveolar macrophages are attractive targets for receptor-mediated delivery of nanostructured therapeutics. In this study, we employed reversible addition fragmentation chain transfer polymerization to synthesize neoglycopolymers, consisting of mannose- and galactose methacrylate-based monomers copolymerized with cholesterol methacrylate for use in functional liposome studies. Glycopolymer-functional liposomes were employed to elucidate macrophage mannose receptor (CD206) and macrophage galactose-type lectin (CD301) targeting in both primary macrophage and immortal macrophage cell lines. Expression of CD206 and CD301 was observed to vary significantly between cell lines (murine alveolar macrophage, murine bone marrow-derived macrophage, RAW264.7, and MH-S), which has significant implications in in vitro targeting and uptake studies. Synthetic glycopolymers and glycopolymer augmented liposomes demonstrated specific receptor-mediated uptake in a manner dependent on carbohydrate receptor expression. These results establish a platform capable of probing endogenous carbohydrate receptor-mediated targeting via glycofunctional nanomaterials.


Subject(s)
Carbohydrate Metabolism , Liposomes , Macrophages, Alveolar , Animals , Cell Line , Drug Carriers , Humans , Lectins , Macrophages , Mannose , Mice
15.
Oncotarget ; 7(8): 9561-75, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26840082

ABSTRACT

The therapeutic potential of RNA interference (RNAi) has been limited by inefficient delivery of short interfering RNA (siRNA). Tumor-specific recognition can be effectively achieved by antibodies directed against highly expressed cancer cell surface receptors. We investigated the utility of linking an internalizing streptavidin-conjugated HER2 antibody to an endosome-disruptive biotinylated polymeric nanocarrier to improve the functional cytoplasmic delivery of siRNA in breast and ovarian cancer cells in vitro and in an intraperitoneal ovarian cancer xenograft model in vivo, yielding an 80% reduction of target mRNA and protein levels with sustained repression for at least 96 hours. RNAi-mediated site specific cleavage of target mRNA was demonstrated using the 5' RLM-RACE (RNA ligase mediated-rapid amplification of cDNA ends) assay. Mice bearing intraperitoneal human ovarian tumor xenografts demonstrated increased tumor accumulation of Cy5.5 fluorescently labeled siRNA and 70% target gene suppression after treatment with HER2 antibody-directed siRNA nanocarriers. Detection of the expected mRNA cleavage product by 5' RLM-RACE assay confirmed that suppression occurs via the expected RNAi pathway. Delivery of siRNA via antibody-directed endosomolytic nanoparticles may be a promising strategy for cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/therapy , Genetic Therapy/methods , Ovarian Neoplasms/therapy , RNA, Small Interfering/therapeutic use , Receptor, ErbB-2/immunology , Trastuzumab/therapeutic use , Animals , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , RNA Interference , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Trastuzumab/immunology , Xenograft Model Antitumor Assays
16.
J Control Release ; 219: 345-354, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26342661

ABSTRACT

The development of drug delivery systems based on well-defined polymer nanostructures could lead to significant improvements in the treatment of cancer. The design of these therapeutic nanosystems must account for numerous systemic and circulation obstacles as well as the specific pathophysiology of the tumor. Nanoparticle size and surface charge must also be carefully selected in order to maintain long circulation times, allow tumor penetration, and avoid clearance by the reticuloendothelial system (RES). Targeting ligands such as vitamins, peptides, and antibodies can improve the accumulation of nanoparticle-based therapies in tumor tissue but must be optimized to allow for intratumoral penetration. In this review, we will highlight factors influencing the design of nanoparticle therapies as well as the development of modern controlled "living" polymerization techniques (e.g. ATRP, RAFT, ROMP) that are leading to the creation of sophisticated new polymer architectures with discrete spatially-defined functional modules. These innovative materials (e.g. star polymers, polymer brushes, macrocyclic polymers, and hyperbranched polymers) combine many of the desirable properties of traditional nanoparticle therapies while substantially reducing or eliminating the need for complex formulations.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems , Nanostructures/administration & dosage , Neoplasms/drug therapy , Polymers/administration & dosage , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Humans , Nanostructures/therapeutic use , Polymerization , Polymers/pharmacokinetics , Polymers/therapeutic use
17.
Mol Ther ; 23(5): 907-917, 2015 May.
Article in English | MEDLINE | ID: mdl-25669432

ABSTRACT

Antibodies armed with biologic drugs could greatly expand the therapeutic potential of antibody-drug conjugates for cancer therapy, broadening their application to disease targets currently limited by intracellular delivery barriers. Additional selectivity and new therapeutic approaches could be realized with intracellular protein drugs that more specifically target dysregulated pathways in hematologic cancers and other malignancies. A multifunctional polymeric delivery system for enhanced cytosolic delivery of protein drugs has been developed that incorporates endosomal-releasing activity, antibody targeting, and a biocompatible long-chain ethylene glycol component for optimized safety, pharmacokinetics, and tumor biodistribution. The pH-responsive polymeric micelle carrier, with an internalizing anti-CD22 monoclonal targeting antibody, effectively delivered a proapoptotic Bcl-2 interacting mediator (BIM) peptide drug that suppressed tumor growth for the duration of treatment and prolonged survival in a xenograft mouse model of human B-cell lymphoma. Antitumor drug activity was correlated with a mechanistic induction of the Bcl-2 pathway biomarker cleaved caspase-3 and a marked decrease in the Ki-67 proliferation biomarker. Broadening the intracellular target space by more effective delivery of protein/peptide drugs could expand the repertoire of antibody-drug conjugates to currently undruggable disease-specific targets and permit tailored drug strategies to stratified subpopulations and personalized medicines.


Subject(s)
Antibodies, Monoclonal , Drug Delivery Systems , Immunoconjugates/pharmacology , Peptides , Animals , Apoptosis/drug effects , Biological Availability , Biomarkers , Cell Line, Tumor , Cytochromes c/biosynthesis , Disease Models, Animal , Drug Stability , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Immunoconjugates/toxicity , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/mortality , Lymphoma, B-Cell/pathology , Mice , Micelles , Polymers/chemistry , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/metabolism , Tissue Distribution , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
18.
AAPS J ; 17(2): 358-69, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25501498

ABSTRACT

Protein-based vaccines offer a number of important advantages over organism-based vaccines but generally elicit poor CD8(+) T cell responses. We have previously demonstrated that pH-responsive, endosomolytic polymers can enhance protein antigen delivery to major histocompatibility complex class I (MHC-I) antigen presentation pathways thereby augmenting CD8(+) T cell responses following immunization. Here, we describe a new family of nanocarriers for protein antigen delivery assembled using architecturally distinct pH-responsive polymers. Reversible addition-fragmentation chain transfer (RAFT) polymerization was used to synthesize linear, hyperbranched, and core-crosslinked copolymers of 2-(N,N-diethylamino)ethyl methacrylate (DEAEMA) and butyl methacrylate (BMA) that were subsequently chain extended with a hydrophilic N,N-dimethylacrylamide (DMA) segment copolymerized with thiol-reactive pyridyl disulfide (PDS) groups. In aqueous solution, polymer chains assembled into 25 nm micellar nanoparticles and enabled efficient and reducible conjugation of a thiolated protein antigen, ovalbumin. Polymers demonstrated pH-dependent membrane-destabilizing activity in an erythrocyte lysis assay, with the hyperbranched and cross-linked polymer architectures exhibiting significantly higher hemolysis at pH ≤ 7.0 than the linear diblock. Antigen delivery with the hyperbranched and cross-linked polymer architecture enhanced in vitro MHC-I antigen presentation relative to free antigen, whereas the linear construct did not have a discernible effect. The hyperbranched system elicited a four- to fivefold increase in MHC-I presentation relative to the cross-linked architecture, demonstrating the superior capacity of the hyperbranched architecture in enhancing MHC-I presentation. This work demonstrates that the architecture of pH-responsive, endosomolytic polymers can have dramatic effects on intracellular antigen delivery, and offers a promising strategy for enhancing CD8(+) T cell responses to protein-based vaccines.


Subject(s)
Histocompatibility Antigens Class I/immunology , Nanoparticles , Polymers/chemistry , Vaccines/immunology , Acrylamides/chemistry , Animals , Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Cross-Linking Reagents/chemistry , Endosomes/metabolism , Hemolysis/drug effects , Humans , Hydrogen-Ion Concentration , Mice , Micelles , Ovalbumin/immunology
19.
Polym Chem ; 5(5): 1791-1799, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-25221630

ABSTRACT

Reversible addition-fragmentation chain transfer (RAFT) polymerization was employed to prepare a nanoparticulate drug delivery system for chemotherapeutics. The nanoparticles contain a PEG "stealth" corona as well as reactive anhydride functionality designed for conjugating targeting proteins. The multifunctional carrier functionality was achieved by controlling the copolymerization of the hydrophobic monomer lauryl methacrylate (LMA), with a reactive anhydride functional methacrylate (TMA), and a large polyethyleneglycol methacrylate monomer (Mn~950 Da) (O950). RAFT polymerization kinetics of O950 were evaluated as a function of target degrees of polymerization (DP), initial chain transfer agent to initiator ratio ([CTA]o/[I]o), and solvent concentration. Excellent control over the polymerization was observed for target DPs of 25 and 50 at [CTA]o/[I]o ratio of 10 as evidenced by narrow and symmetric molecular weight distributions and the ability to prepare block copolymers. The TMA-functional copolymers were conjugated to the tumor targeting protein transferrin (Tf). The targeted copolymer was shown to encapsulate docetaxel at concentrations comparable to the commercial single vial formulation of docetaxel (Taxotere). In vitro cytotoxicity studies conducted in HeLa cells show that the Tf targeting enhances the cancer killing properties relative to the polymer encapsulated docetaxel formulation.

20.
Cell ; 157(7): 1644-1656, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24949974

ABSTRACT

Because apoptosis of infected cells can limit virus production and spread, some viruses have co-opted prosurvival genes from the host. This includes the Epstein-Barr virus (EBV) gene BHRF1, a homolog of human Bcl-2 proteins that block apoptosis and are associated with cancer. Computational design and experimental optimization were used to generate a novel protein called BINDI that binds BHRF1 with picomolar affinity. BINDI recognizes the hydrophobic cleft of BHRF1 in a manner similar to other Bcl-2 protein interactions but makes many additional contacts to achieve exceptional affinity and specificity. BINDI induces apoptosis in EBV-infected cancer lines, and when delivered with an antibody-targeted intracellular delivery carrier, BINDI suppressed tumor growth and extended survival in a xenograft disease model of EBV-positive human lymphoma. High-specificity-designed proteins that selectively kill target cells may provide an advantage over the toxic compounds used in current generation antibody-drug conjugates.


Subject(s)
Herpesvirus 4, Human/chemistry , Protein Engineering , Proteins/pharmacology , Viral Proteins/antagonists & inhibitors , Amino Acid Sequence , Animals , Apoptosis/drug effects , Computational Biology , Crystallography, X-Ray , Epstein-Barr Virus Infections/drug therapy , Herpesvirus 4, Human/physiology , Heterografts , Humans , Lymphoma, B-Cell/drug therapy , Mice , Models, Molecular , Molecular Sequence Data , Neoplasm Transplantation , Proteins/chemistry , Proteins/metabolism , Sequence Alignment , Viral Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...