Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
Add more filters











Publication year range
1.
Cell Chem Biol ; 31(8): 1490-1502.e42, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39116881

ABSTRACT

We describe a protein proximity inducing therapeutic modality called Regulated Induced Proximity Targeting Chimeras or RIPTACs: heterobifunctional small molecules that elicit a stable ternary complex between a target protein (TP) selectively expressed in tumor cells and a pan-expressed protein essential for cell survival. The resulting co-operative protein-protein interaction (PPI) abrogates the function of the essential protein, thus leading to death selectively in cells expressing the TP. This approach leverages differentially expressed intracellular proteins as novel cancer targets, with the advantage of not requiring the target to be a disease driver. In this chemical biology study, we design RIPTACs that incorporate a ligand against a model TP connected via a linker to effector ligands such as JQ1 (BRD4) or BI2536 (PLK1) or CDK inhibitors such as TMX3013 or dinaciclib. RIPTACs accumulate selectively in cells expressing the HaloTag-FKBP target, form co-operative intracellular ternary complexes, and induce an anti-proliferative response in target-expressing cells.


Subject(s)
Antineoplastic Agents , Cell Cycle Proteins , Small Molecule Libraries , Humans , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Cell Proliferation/drug effects , Triazoles/chemistry , Triazoles/pharmacology , Polo-Like Kinase 1 , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Azepines/pharmacology , Azepines/chemistry , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Transcription Factors/metabolism , Transcription Factors/antagonists & inhibitors , Indolizines/chemistry , Indolizines/pharmacology , Cell Line, Tumor , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Ligands , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Heterocyclic Compounds, 2-Ring/pharmacology , Heterocyclic Compounds, 2-Ring/chemistry , Heterocyclic Compounds, 2-Ring/chemical synthesis , Nuclear Proteins/metabolism , Nuclear Proteins/antagonists & inhibitors , Bromodomain Containing Proteins , Cyclic N-Oxides , Pyridinium Compounds
2.
Clin Exp Dent Res ; 9(2): 398-408, 2023 04.
Article in English | MEDLINE | ID: mdl-36779270

ABSTRACT

OBJECTIVE: The primary objective of this research was to develop a poly(l-lactic acid) (PLLA) scaffold and evaluate critical characteristics essential for its biologic use as a craniofacial implant. MATERIALS AND METHODS: PLLA scaffolds were designed and fabricated using fused deposition modeling technology. The surface morphology and microarchitecture were analyzed using scanning electron microscopy (SEM) and microCT, respectively. Crystallography, compressive modulus, and the piezoelectric potential generated upon mechanical distortion were characterized. Hydrolytic degradation was studied. MG63 osteoblast-like cell proliferation and morphology were assessed. RESULTS: The porosity of the scaffolds was 73%, with an average pore size of 450 µm and an average scaffold fiber thickness of 130 µm. The average compressive modulus was 244 MPa, and the scaffolds generated an electric potential of 25 mV upon cyclic/repeated loading. The crystallinity reduced from 27.5% to 13.9% during the 3D printing process. The hydrolytic degradation was minimal during a 12-week period. Osteoblast-like cells did not attach to the uncoated scaffold but attached well after coating the scaffold with fibrinogen. They then proliferated to cover the complete scaffold by Day 14. CONCLUSION: The PLLA scaffolds were designed and printed, proving the feasibility of 3D printing as a method of fabricating PLLA scaffolds. The elastic modulus was comparable to that of trabecular bone, and the piezoelectric properties of the PLLA were retained after 3D printing. The scaffolds were cytocompatible. These 3D-printed PLLA scaffolds showed promising properties akin to the natural bone, and they warrant further investigation for bone regeneration.


Subject(s)
Bone Regeneration , Tissue Scaffolds , Tissue Scaffolds/chemistry , Printing, Three-Dimensional , Porosity
3.
J Biomed Mater Res A ; 111(6): 790-800, 2023 06.
Article in English | MEDLINE | ID: mdl-36606344

ABSTRACT

There is an urgent unmet medical need to develop therapeutic options for the ~50% of depression patients suffering from treatment-resistant depression, which is difficult to treat with existing psycho- and pharmaco-therapeutic options. Classical psychedelics, such as the 5HT2A agonists, have re-emerged as a treatment paradigm for depression. Recent clinical trials highlight the potential effectiveness of 5HT2A agonists to improve mood and psychotherapeutic growth in treatment-resistant depression patients, even in those who have failed a median of four previous medications in their lifetime. Moreover, microdosing could be a promising way to achieve long-term alleviation of depression symptoms without a hallucinogenic experience. However, there are a gamut of practical barriers that stymie further investigation of microdosing 5HT2A agonists, including: low compliance with the complicated dosing regimen, high risk of diversion of controlled substances, and difficulty and cost administering the long-term treatment regimens in controlled settings. Here, we developed a drug delivery system composed of multilayered cellulose acetate phthalate (CAP)/Pluronic F-127 (P) films for the encapsulation and interval delivery of 5HT2A agonists from a fully biodegradable and biocompatible implant. CAPP film composition, thickness, and layering strategies were optimized, and we demonstrated three distinct pulses from the multilayered CAPP films in vitro. Additionally, the pharmacokinetics and biodistribution of the 5HT2A agonist 2,5-Dimethoxy-4-iodoamphetamine (DOI) were quantified following the subcutaneous implantation of DOI-loaded single and multilayered CAPP films. Our results demonstrate, for the first time, the interval delivery of psychedelics from an implantable drug delivery system and open the door to future studies into the therapeutic potential of psychedelic delivery.


Subject(s)
Hallucinogens , Humans , Polymers , Tissue Distribution , Pharmaceutical Preparations
4.
bioRxiv ; 2023 Jan 02.
Article in English | MEDLINE | ID: mdl-36711980

ABSTRACT

While specific cell signaling pathway inhibitors have yielded great success in oncology, directly triggering cancer cell death is one of the great drug discovery challenges facing biomedical research in the era of precision oncology. Attempts to eradicate cancer cells expressing unique target proteins, such as antibody-drug conjugates (ADCs), T-cell engaging therapies, and radiopharmaceuticals have been successful in the clinic, but they are limited by the number of targets given the inability to target intracellular proteins. More recently, heterobifunctional small molecules such as Proteolysis Targeting Chimera (PROTACs) have paved the way for protein proximity inducing therapeutic modalities. Here, we describe a proof-of-concept study using novel heterobifunctional small molecules called Regulated Induced Proximity Targeting Chimeras or RIPTACs, which elicit a stable ternary complex between a target protein selectively expressed in cancer tissue and a pan-expressed protein essential for cell survival. The resulting cooperative protein:protein interaction (PPI) abrogates the function of the essential protein, thus leading to cell death selectively in cells expressing the target protein. This approach not only opens new target space by leveraging differentially expressed intracellular proteins but also has the advantage of not requiring the target to be a driver of disease. Thus, RIPTACs can address non-target mechanisms of resistance given that cell killing is driven by inactivation of the essential protein. Using the HaloTag7-FKBP model system as a target protein, we describe RIPTACs that incorporate a covalent or non-covalent target ligand connected via a linker to effector ligands such as JQ1 (BRD4), BI2536 (PLK1), or multi-CDK inhibitors such as TMX3013 or dinaciclib. We show that these RIPTACs exhibit positive co-operativity, accumulate selectively in cells expressing HaloTag7-FKBP, form stable target:RIPTAC:effector trimers in cells, and induce an anti-proliferative response in target-expressing cells. We propose that RIPTACs are a novel heterobifunctional therapeutic modality to treat cancers that are known to selectively express a specific intracellular protein.

5.
Tetrahedron Lett ; 772021 Aug 03.
Article in English | MEDLINE | ID: mdl-34393283

ABSTRACT

Small molecules that selectively bind to the pseudokinase JH2 domain over the JH1 kinase domain of JAK2 kinase are sought. Virtual screening led to the purchase of 17 compounds among which 9 were found to bind to V617F JAK2 JH2 with affinities of 40 - 300 µM in a fluorogenic assay. Ten analogues were then purchased yielding 9 additional active compounds. Aminoanilinyltriazine 22 was particularly notable as it shows no detectable binding to JAK2 JH1, and it has a 65-µM dissociation constant K d with V617F JAK2 JH2. A crystal structure for 22 in complex with wild-type JAK2 JH2 was obtained to elucidate the binding mode. Additional de novo design led to the synthesis of 19 analogues of 22 with the most potent being 33n with K d values of 2-3 µM for WT and V617F JAK2 JH2, and with 16-fold selectivity relative to binding with WT JAK2 JH1.

6.
Mater Sci Eng C Mater Biol Appl ; 118: 111395, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33255000

ABSTRACT

Bone pain is the leading cause of morbidity in patients with metastatic cancer. Systemic administration of zoledronic acid (ZA) decreases skeletally-related events in bone cancer patients but is associated with major side effects. This project investigated two biomaterials, poly(methyl methacrylate) (PMMA) bone cement and poly(lactic-co-glycolic acid) (PLGA), for local ZA delivery. Compressive properties of PMMA samples were tested with increased drug loading, and in vitro ZA release profiles from PMMA cylinders and PLGA films were measured over 8 weeks. The activity of ZA eluted from both materials was evaluated on the RAW 264.7 macrophage cell line. PMMA samples released up to only 17% of incorporated drug, whereas PLGA films released over 95%. A burst profile was observed for PMMA, while ZA release from PLGA exhibited a typical triphasic profile. Drug bioactivity remained above 50% for both materials. Local ZA delivery from these materials may be useful in the treatment of metastatic bone cancer.


Subject(s)
Bone Diseases , Neoplasms , Bone and Bones , Delayed-Action Preparations , Humans , Zoledronic Acid/pharmacology
7.
J Med Chem ; 63(10): 5324-5340, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32329617

ABSTRACT

Janus kinases (JAKs) are non-receptor tyrosine kinases that are essential components of the JAK-STAT signaling pathway. Associated aberrant signaling is responsible for many forms of cancer and disorders of the immune system. The present focus is on the discovery of molecules that may regulate the activity of JAK2 by selective binding to the JAK2 pseudokinase domain, JH2. Specifically, the Val617Phe mutation in JH2 stimulates the activity of the adjacent kinase domain (JH1) resulting in myeloproliferative disorders. Starting from a non-selective screening hit, we have achieved the goal of discovering molecules that preferentially bind to the ATP binding site in JH2 instead of JH1. We report the design and synthesis of the compounds and binding results for the JH1, JH2, and JH2 V617F domains, as well as five crystal structures for JH2 complexes. Testing with a selective and non-selective JH2 binder on the autophosphorylation of wild-type and V617F JAK2 is also contrasted.


Subject(s)
Amitrole/chemistry , Amitrole/metabolism , Enzyme Activators/chemistry , Enzyme Activators/metabolism , Janus Kinase 2/chemistry , Janus Kinase 2/metabolism , Animals , HEK293 Cells , Humans , Ligands , Protein Binding/physiology , Sf9 Cells , X-Ray Diffraction/methods
8.
Spine Deform ; 8(4): 553-559, 2020 08.
Article in English | MEDLINE | ID: mdl-32078142

ABSTRACT

STUDY DESIGN: Basic science. OBJECTIVE: Investigate the ability of local applicaiton of vancomycin, either in powder form or suspended within poly(lactic-co-glycolic acid) microspheres (MS), to treat infection using a rat spinal model. Surgical site infections (SSIs) are a serious complication after spine surgery and are associated with high morbidity and mortality and often caused my coagulase negative staphylococci. A comprehensive approach to reduce SSIs has been recommended including the use of topical vancomycin. Animal and human studies have shown improved control of infection with local compared to systemic antibiotics. METHODS: K-wires seeded with methicillin-resistant Staphylococcus epidermidis RP62A (MRSE) were treated with vancomycin powder, carboxymethylcellulose sodium salt (CMC) (microsphere carrier), vancomycin powder, blank MS or vancomycin-loaded MS for 24 or 48 h in vitro after which bacteria were enumerated. In addition, a spinal instrumentation model was developed in rats with a bacterial seeded K-wire implanted into the right side of L4 and L5. Rats underwent no treatment or were treated locally with either vancomycin powder, blank MS or vancomycin-loaded MS. After 8 weeks, the K-wire, bone, soft tissue and wire fastener were cultured and results analyzed. RESULTS: Vancomycin powder and vancomycin-loaded MS resulted in significantly fewer bacteria remaining in vitro than did CMC. Vancomycin powder- treated animals' cultures were significantly lower than all other groups (P < 0.0001) with negative culture results, except for one animal. The vancomycin-loaded MS-treated animals had lower bone bacterial counts than the controls (P < 0.0279); blank MS-treated animals had no differences in bacterial densities when compared to non-treated animals. CONCLUSION: Vancomycin powder and vancomycin-loaded MS were active against MRSE in vitro, in a rat MRSE implant model; however, vancomycin MS were inferior to the topical vancomycin powder. Vancomycin powder prevented MRSE infection in a rat spinal implant infection model.


Subject(s)
Bone Wires/microbiology , Spine/surgery , Staphylococcus epidermidis , Surgical Wound Infection/drug therapy , Surgical Wound Infection/microbiology , Vancomycin/administration & dosage , Animals , Bacterial Load , Disease Models, Animal , Drug Resistance, Bacterial , Female , Methicillin Resistance , Microspheres , Polylactic Acid-Polyglycolic Acid Copolymer , Powders , Rats, Sprague-Dawley , Staphylococcus epidermidis/drug effects , Surgical Wound Infection/prevention & control , Vancomycin/pharmacology
9.
Acta Biomater ; 93: 192-199, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31055123

ABSTRACT

Previous research from our labs demonstrated the synthesis of polymerized simvastatin by ring-opening polymerization and slow degradation with controlled release of simvastatin in vitro. The objective of the present study was to evaluate the degradation and intramembranous bone-forming potential of simvastatin-containing polyprodrugs in vivo using a rat calvarial onlay model. Poly(ethylene glycol)-block-poly(simvastatin) and poly(ethylene glycol)-block-poly(simvastatin)-ran-poly(glycolide) were compared with simvastatin conventionally encapsulated in poly(lactic-co-glycolic acid) (PLGA) and pure PLGA. The rate of degradation was higher for PLGA with and without simvastatin relative to the simvastatin polyprodrugs. Significant new bone growth at the circumference of poly(ethylene glycol)-block-poly(simvastatin) disks was observed beginning at 4 weeks, whereas severe bone resorption (4 weeks) and bone loss (8 weeks) were observed for PLGA loaded with simvastatin. No significant systemic effects were observed for serum total cholesterol and body weight. Increased expression of osteogenic (BMP-2, Runx2, and ALP), angiogenic (VEGF), and inflammatory cytokines (IL-6 and NF-ĸB) genes was seen with all polymers at the end of 8 weeks. Poly(ethylene glycol)-block-poly(simvastatin), with slow degradation and drug release, controlled inflammation, and significant osteogenic effect, is a candidate for use in bone regeneration applications. STATEMENT OF SIGNIFICANCE: Traditional drug delivery systems, e.g., drug encapsulated in poly(lactic-co-glycolic acid) (PLGA), are typically passive and have limited drug payload. As an alternative, we polymerized the drug simvastatin, which has multiple physiological effects, into macromolecules ("polysimvastatin") via ring-opening polymerization. We previously demonstrated that the rate of degradation and drug (simvastatin) release can be adjusted by copolymerizing it with other monomers. The present results demonstrate significant new bone growth around polysimvastatin, whereas severe bone loss occurred for PLGA loaded with simvastatin. This degradable biomaterial with biofunctionality integrated into the polymeric backbone is a useful candidate for bone regeneration applications.


Subject(s)
Absorbable Implants , Bone Regeneration/drug effects , Osteogenesis/drug effects , Polymers/chemistry , Simvastatin/chemistry , Tissue Scaffolds/chemistry , Angiogenesis Inducing Agents/metabolism , Animals , Body Weight/drug effects , Bone Morphogenetic Protein 2/metabolism , Cholesterol/blood , Core Binding Factor Alpha 1 Subunit/metabolism , Cytokines/metabolism , Cytoskeletal Proteins/metabolism , Drug Delivery Systems , Drug Liberation , Male , Models, Animal , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polymers/pharmacology , Rats , Rats, Sprague-Dawley , Simvastatin/pharmacology , Skull/drug effects , Skull/surgery
10.
Am J Orthod Dentofacial Orthop ; 155(2): 254-263, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30712697

ABSTRACT

INTRODUCTION: Nonhuman animal models have been used extensively to study orthodontic tooth movement (OTM). However, rodent models have disadvantages, including a reported reduction in bone volume during OTM. The purpose of this study was to determine the viability of a skeletal anchorage and the effect of low force (∼3 cN) on interradicular bone volume during OTM. METHODS: Ninety Sprague-Dawley rats were divided into 5 time points. A miniscrew and a nickel titanium coil spring placed a load of 3 cN (experimental) or 0 cN (sham) on the maxillary first molar in a split-mouth design. Displacement of the first molar and bone volume/total volume (BV/TV) in the interradicular region were quantified. RESULTS: The success rate of the miniscrew was 98.9% (89 out of 90). Linear and angular tooth movement increased steadily (mean 0.1 mm/wk, 0.48 mm at 40 days). BV/TV was significantly reduced between the tooth movement and non-tooth movement sides in the 3 cN group: by 13%, 23%, 15%, 23%, and 16% at 3, 7, 14, 28, and 40 days, respectively. CONCLUSIONS: Our model resulted in efficient OTM without skeletal anchorage failure. BV/TV reduction was lower than in previous reports. This novel validated model is likely to be the basis for future studies.


Subject(s)
Maxilla/anatomy & histology , Orthodontic Anchorage Procedures , Tooth Movement Techniques/methods , Animals , Male , Models, Animal , Organ Size , Rats , Rats, Sprague-Dawley
11.
ACS Appl Bio Mater ; 2(6): 2511-2519, 2019 Jun 17.
Article in English | MEDLINE | ID: mdl-33912813

ABSTRACT

Biodegradable scaffolds are widely use in drug delivery and tissue engineering applications. The scaffolds can be modified to provide the necessary mechanical support for tissue formation and to deliver one or more drugs to stimulate tissue formation or for the treatment of a specific condition. In the current study, we developed biodegradable scaffolds that have the potential for dual drug delivery. The scaffolds consisted of simvastatin-containing prodrug, poly(simvastatin) entrapped in poly(ß-amino ester) (PBAE) porogen particles and vancomycin encapsulated in poly(lactic-co-glycolic acid) (PLGA) microspheres, which were fused together around the PBAE porogens to create a slow-degrading matrix. Upon hydrolysis, poly(simvastatin) releases simvastatin acid, which has angiogenic and osteogenic properties, while the PLGA microspheres release vancomycin as an antibacterial agent. Degradation of PBAE porogens through hydrolysis of ester linkages led to the development of porosity in a controlled manner and led to water penetration that facilitated hydrolysis of PLGA. Higher porogen loading (~60% by weight) gave rise to ~70% interconnected porosity with pore spacing of ~180 µm. This open volume facilitated simvastatin acid release upon hydrolysis and entrapped vancomycin release via diffusion through and degradation of PLGA. During the study, ~162 µg of simvastatin acid and ~18 mg vancomycin were released from the highest porosity scaffolds. Bioactivity studies showed that released simvastatin acid stimulated preosteoblastic activity, indicating that scaffold fabrication did not damage the polymeric prodrug. Regarding mechanical properties, compressive modulus, failure strain, and failure stress decreased with increasing PBAE porogen content. These dual drug releasing scaffolds with controlled development of microarchitecture can be useful in bone tissue engineering applications.

12.
Proc Natl Acad Sci U S A ; 115(33): 8340-8345, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30061385

ABSTRACT

Elucidating the physiological roles and modes of action of the recently discovered ligands (designated ALKAL1,2 or AUG-α,ß) of the receptor tyrosine kinases Anaplastic Lymphoma Kinase (ALK) and Leukocyte Tyrosine Kinase (LTK) has been limited by difficulties in producing sufficient amounts of the two ligands and their poor stability. Here we describe procedures for expression and purification of AUG-α and a deletion mutant lacking the N-terminal variable region. Detailed biochemical characterization of AUG-α by mass spectrometry shows that the four conserved cysteines located in the augmentor domain (AD) form two intramolecular disulfide bridges while a fifth, primate-specific cysteine located in the N-terminal variable region mediates dimerization through formation of a disulfide bridge between two AUG-α molecules. In contrast to AUG-α, the capacity of AUG-α AD to undergo dimerization is strongly compromised. However, full-length AUG-α and the AUG-α AD deletion mutant stimulate similar tyrosine phosphorylation of cells expressing either ALK or LTK. Both AUG-α and AUG-α AD also stimulate a similar profile of MAP kinase response in L6 cells and colony formation in soft agar by autocrine stimulation of NIH 3T3 cells expressing ALK. Moreover, both AUG-α and AUG-α AD stimulate neuronal differentiation of human neuroblastoma NB1 and PC12 cells in a similar dose-dependent manner. Taken together, these experiments show that deletion of the N-terminal variable region minimally affects the activity of AUG-α toward LTK or ALK stimulation in cultured cells. Reduced dimerization might be compensated by high local concentration of AUG-α AD bound to ALK at the cell membrane and by potential ligand-induced receptor-receptor interactions.


Subject(s)
Cytokines/isolation & purification , Receptor Protein-Tyrosine Kinases/isolation & purification , Amino Acid Motifs , Anaplastic Lymphoma Kinase , Animals , Cytokines/chemistry , Cytokines/physiology , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , PC12 Cells , Protein Multimerization , Rats , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/metabolism
13.
Acta Biomater ; 68: 178-189, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29289681

ABSTRACT

Recently, biomaterials have been designed to contain redox-sensitive moieties, such as thiols and disulfides, to impart responsive degradation and/or controlled release. However, due to the high sensitivity of cellular redox-based systems which maintain free-radical homeostasis (e.g. glutathione/glutathione disulfide), if these biomaterials modify the cellular redox environment, they may inadvertently affect cellular compatibility and/or oxidative stress defenses. In this work, we hypothesize that the degradation products of a poly(ß-amino ester) (PBAE) hydrogel formed with redox sensitive disulfide (cystamine) crosslinking could serve as a supplement to the environmental cellular antioxidant defenses. Upon introduction into a reducing environment, these disulfide-containing hydrogels cleave to present bound-thiol groups, yet remain in the bulk form at up to 66 mol% cystamine of the total amines. By controlling the molar fraction of cystamine, it was apparent that the thiol content varied human umbilical vein endothelial cell (HUVEC) viability IC50 values across an order of magnitude. Further, upon introduction of an enzymatic oxidative stress generator to the cell culture (HX/XO), pre-incubated thiolated hydrogel degradation products conferred cellular and mitochondrial protection from acute oxidative stress, whereas non-reduced disulfide-containing degradation products offered no protection. This polymer may be an advantageous implantable drug delivery system for use in acute oxidative stress prophylaxis and/or chronic oxidative stress cell therapies due to its solid/liquid reversibility in a redox environment, controlled thiolation, high loading capacity through covalent drug-addition, and simple post-synthesis modification which bound-thiols introduce. STATEMENT OF SIGNIFICANCE: In this work, we demonstrate a unique property of disulfide containing degradable biomaterials. By changing the redox state of the degradation products (from oxidized to reduced), it is possible to increase the IC50 of the material by an order of magnitude. This dramatic shift is linked directly to the oxidative stress response of the cells and suggests a possible mechanism by which one can tune the cellular response to degradable biomaterials.


Subject(s)
Antioxidants/pharmacology , Disulfides/chemistry , Drug Delivery Systems , Hydrogels/chemistry , Polymers/chemistry , Cell Death/drug effects , Cell Survival/drug effects , Drug Liberation , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Inhibitory Concentration 50 , Mitochondria/drug effects , Mitochondria/metabolism , Nanoparticles/chemistry , Oxidation-Reduction , Oxidative Stress/drug effects , Oxygen Consumption/drug effects , Polymers/chemical synthesis , Sulfhydryl Compounds/chemistry
14.
Gels ; 4(4)2018 Oct 08.
Article in English | MEDLINE | ID: mdl-30674856

ABSTRACT

Disulfides are used extensively in reversible cross-linking because of the ease of reduction into click-reactive thiols. However, the free-radical scavenging properties upon reduction are often under-considered. The free thiols produced upon reduction of this disulfide material mimic the cellular reducing chemistry (glutathione) that serves as a buffer against acute oxidative stress. A nanoparticle formulation producing biologically relevant concentrations of thiols may not only provide ample chemical conjugation sites, but potentially be useful against severe acute oxidative stress exposure, such as in targeted radioprotection. In this work, we describe the synthesis and characterization of highly thiolated poly (ß-amino ester) (PBAE) nanoparticles formed from the reduction of bulk disulfide cross-linked PBAE hydrogels. Degradation-tunable PBAE hydrogels were initially synthesized containing up to 26 wt % cystamine, which were reduced into soluble thiolated oligomers and formulated into nanoparticles upon single emulsion. These thiolated nanoparticles were size-stable in phosphate buffered saline consisting of up to 11.0 ± 1.1 mM (3.7 ± 0.3 mmol thiol/g, n = 3 M ± SD), which is an antioxidant concentration within the order of magnitude of cellular glutathione (1⁻10 mM).

15.
ACS Biomater Sci Eng ; 4(12): 4193-4199, 2018 Dec 10.
Article in English | MEDLINE | ID: mdl-30631799

ABSTRACT

Simvastatin was previously converted to a polymeric prodrug with higher drug loading, but the hydrophobic nature of the poly(simvastatin) component of the block copolymer led to slow release of the drug in vitro. In this study, we hypothesized that degradation could be accelerated by chemically modifying the polymer backbone by introducing glycolide and lactide comonomers. Copolymers were formed by ring-opening polymerization using 5 kDa monomethyl ether poly(ethylene glycol) as the microinitiator in presence of triazabicyclodecene catalyst. In addition to simvastatin, modified reaction mixtures contained lactide or glycolide. Incorporation of the less hydrophobic glycolide comonomer led to in vitro degradation of up to two times greater mass loss, release of up to ~7 times more simvastatin, and a 2-3 times increase in compressive modulus compared to the lactide-containing and parent polymers.

16.
J Biomed Mater Res B Appl Biomater ; 106(5): 1878-1886, 2018 07.
Article in English | MEDLINE | ID: mdl-28926192

ABSTRACT

Large infected bone defects, often resulting from high energy traumas, are difficult to treat due to their variability in complexity and location. Standard treatment for infected bone defects begins with a protocol that includes a series of debridements in conjunction with an extended course of systemic antibiotics. Only after the infection has been eliminated will repair of the defect commence, typically with implantation of autologous bone. To address some of the shortcomings of the standard treatment methods, such as serial procedures, limited grafting material, and the need for a second surgical site for autologous bone, a sequential, dual drug-releasing, moldable, calcium sulfate-based bone graft substitute was developed previously. In the present studies, the effectiveness of the material for treating both the infection with vancomycin and bone defect with simvastatin was evaluated in vivo using a critically sized, infected segmental defect model in rat femurs. Although the infection was not fully eliminated, the local release of vancomycin increased survivorship of infected animals by 464% compared to nontreated controls. Infected animals receiving antimicrobial treatment showed comparable amounts of new bone formation within the defect site when compared to noninfected controls. Incorporating agents capable of disrupting established biofilms into bone graft substitutes may enhance effectiveness in treating a biofilm infection within a bone defect. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 1878-1886, 2018.


Subject(s)
Bone Substitutes , Calcium Sulfate , Femur , Vancomycin , Animals , Bone Substitutes/chemistry , Bone Substitutes/pharmacology , Calcium Sulfate/chemistry , Calcium Sulfate/pharmacology , Femur/injuries , Femur/metabolism , Femur/microbiology , Femur/pathology , Infection Control , Infections/microbiology , Male , Rats , Rats, Sprague-Dawley , Vancomycin/chemistry , Vancomycin/pharmacology
17.
React Funct Polym ; 119: 37-46, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29217967

ABSTRACT

Simvastatin was polymerized into copolymers to better control drug loading and release for therapeutic delivery. When using the conventional stannous octoate catalyst in ring-opening polymerization (ROP), reaction temperatures ≥200 °C were required, which promoted uncontrollable and undesirable side reactions. Triazabicyclodecene (TBD), a highly reactive guanidine base organocatalyst, was used as an alternative to polymerize simvastatin. Polymerization was achieved at 150 °C using 5 kDa methyl-terminated poly(ethylene glycol) (mPEG) as the initiator. ROP reactions with 2 kDa or 550 Da mPEG initiators were also successful using TBD at 150 °C instead of stannous octoate, which required a higher reaction temperature. Biodegradability of the poly(simvastatin) copolymer in phosphate-buffered saline was also improved, losing twice as much mass than the copolymer synthesized via stannous octoate. The three copolymers exhibited modified rates of simvastatin release, demonstrating tunablity for drug delivery applications.

19.
Int J Polym Mater ; 66(11): 569-576, 2017.
Article in English | MEDLINE | ID: mdl-28736462

ABSTRACT

Sequential release of drugs aligned with the phases of tissue healing could reduce scarring. To achieve this aim, layered film devices comprising cellulose acetate phthalate (CAP) and Pluronic F-127 (Pluronic) were loaded with ketoprofen, quercetin, and pirfenidone. Citrate plasticizers were added to impart flexibility. Release of two or three drugs in sequence over several days was obtained for all multilayered devices tested. Mechanical analysis showed that elongation increased and modulus decreased with increasing plasticizer content. Release profiles can be tailored by order of layers, plasticizer concentration, and drug loaded, making CAP-Pluronic an appealing system for inhibiting scar tissue formation.

20.
ACS Med Chem Lett ; 8(6): 614-617, 2017 Jun 08.
Article in English | MEDLINE | ID: mdl-28626520

ABSTRACT

A competitive fluorescence polarization (FP) assay is reported for determining binding affinities of probe molecules with the pseudokinase JAK2 JH2 allosteric site. The syntheses of the fluorescent 5 and 6 used in the assay are reported as well as Kd results for 10 compounds, including JNJ7706621, NVP-BSK805, and filgotinib (GLPG0634). X-ray crystal structures of JAK2 JH2 in complex with NVP-BSK805, filgotinib, and diaminopyrimidine 8 elucidate the binding poses.

SELECTION OF CITATIONS
SEARCH DETAIL