Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Biomater Sci Eng ; 7(7): 3209-3220, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34160196

ABSTRACT

Successful treatment of glioblastoma (GBM) is hampered by primary tumor recurrence after surgical resection and poor prognosis, despite adjuvant radiotherapy and chemotherapy. In search of improved outcomes for this disease, quisinostat appeared as a lead compound in drug screening. A delivery system was devised for this drug and to exploit current clinical methodology: an injectable hydrogel, loaded with both the quisinostat drug and radiopaque gold nanoparticles (AuNP) as contrast agent, that can release these payloads as a response to radiation. This hydrogel grants high local drug concentrations, overcoming issues with current standards of care. Significant hydrogel degradation and quisinostat release were observed due to the radiation trigger, providing high in vitro anticancer activity. In vivo, the combination of radiotherapy and the radiation-induced delivery of quisinostat from the hydrogel, successfully inhibited tumor growth in a mice model bearing xenografted human GBM tumors with a total response rate of 67%. Long-term tolerability was observed after intratumoral injection of the quisinostat loaded hydrogel. The AuNP payload enabled precise image-guided radiation delivery and the monitoring of hydrogel degradation using computed tomography (CT). These exciting results highlight this hydrogel as a versatile imageable drug delivery platform that can be activated simultaneously to radiation therapy and potentially offers improved treatment for GBM.


Subject(s)
Glioblastoma , Metal Nanoparticles , Glioblastoma/diagnostic imaging , Gold , Humans , Hydrogels , Neoplasm Recurrence, Local
2.
Macromol Biosci ; 19(1): e1800248, 2019 01.
Article in English | MEDLINE | ID: mdl-30259658

ABSTRACT

Shear-thinning hydrogels are useful for biomedical applications, from 3D bioprinting to injectable biomaterials. Although they have the appropriate properties for injection, it may be advantageous to decouple injectability from the controlled release of encapsulated therapeutics. Toward this, composites of hydrogels and encapsulated microgels are introduced with microgels that are fabricated via microfluidics. The microgel cross-linker controls degradation and entrapped molecule release, and the concentration of microgels alters composite hydrogel rheological properties. For the treatment of myocardial infarction (MI), interleukin-10 (IL-10) is encapsulated in microgels and released from composites. In a rat model of MI, composites with IL-10 reduce macrophage density after 1 week and improve scar thickness, ejection fraction, cardiac output, and the size of vascular structures after 4 weeks when compared to saline injection. Improvements are also observed with the composite without IL-10 over saline, emphasizing the role of injectable hydrogels alone on tissue repair.


Subject(s)
Biocompatible Materials , Hydrogels , Interleukin-10 , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacokinetics , Biocompatible Materials/pharmacology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Disease Models, Animal , Hydrogels/chemistry , Hydrogels/pharmacokinetics , Hydrogels/pharmacology , Interleukin-10/chemistry , Interleukin-10/pharmacokinetics , Interleukin-10/pharmacology , Myocardial Infarction/metabolism , Rats
3.
J Thorac Cardiovasc Surg ; 157(4): 1479-1490, 2019 04.
Article in English | MEDLINE | ID: mdl-30579534

ABSTRACT

OBJECTIVES: The ventricle undergoes adverse remodeling after myocardial infarction, resulting in abnormal biomechanics and decreased function. We hypothesize that tissue-engineered therapy could minimize postischemic remodeling through mechanical stress reduction and retention of tensile myocardial properties due to improved endothelial progenitor cell retention and intrinsic biomechanical properties of the hyaluronic acid shear-thinning gel. METHODS: Endothelial progenitor cells were harvested from adult Wistar rats and resuspended in shear-thinning gel. The constructs were injected at the border zone of ischemic rat myocardium in an acute model of myocardial infarction. Myocardial remodeling, tensile properties, and hemodynamic function were analyzed: control (phosphate-buffered saline), endothelial progenitor cells, shear-thinning gel, and shear-thinning gel + endothelial progenitor cells. Novel high-resolution, high-sensitivity ultrasound with speckle tracking allowed for global strain analysis. Uniaxial testing assessed tensile biomechanical properties. RESULTS: Shear-thinning gel + endothelial progenitor cell injection significantly increased engraftment and retention of the endothelial progenitor cells within the myocardium compared with endothelial progenitor cells alone. With the use of strain echocardiography, a significant improvement in left ventricular ejection fraction was noted in the shear-thinning gel + endothelial progenitor cell cohort compared with control (69.5% ± 10.8% vs 40.1% ± 4.6%, P = .04). A significant normalization of myocardial longitudinal displacement with subsequent stabilization of myocardial velocity with shear-thinning gel + endothelial progenitor cell therapy compared with control was also evident (0.84 + 0.3 cm/s vs 0.11 ± 0.01 cm/s, P = .03). A significantly positive and higher myocardial strain was observed in shear-thinning gel + endothelial progenitor cell (4.5% ± 0.45%) compared with shear-thinning gel (3.7% ± 0.24%), endothelial progenitor cell (3.5% ± 0.97%), and control (8.6% ± 0.3%, P = .05). A resultant reduction in dynamic stiffness was noted in the shear-thinning gel + endothelial progenitor cell cohort. CONCLUSIONS: This novel injectable shear-thinning hyaluronic acid hydrogel demonstrates stabilization of border zone myocardium with reduction in adverse myocardial remodeling and preservation of myocardial biomechanics. The cellular construct provides a normalization of strain measurements and reduces left ventricular dilatation, thus resulting in improvement of left ventricular function.


Subject(s)
Endothelial Progenitor Cells/transplantation , Hemodynamics , Hyaluronic Acid/administration & dosage , Myocardial Infarction/surgery , Myocardium/pathology , Stem Cell Transplantation/methods , Ventricular Function, Left , Ventricular Remodeling , Animals , Biomechanical Phenomena , Cell Survival , Cells, Cultured , Disease Models, Animal , Graft Survival , Hydrogels , Injections , Male , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Neovascularization, Physiologic , Rats, Wistar , Recovery of Function , Stress, Mechanical , Tensile Strength
4.
Bioconjug Chem ; 29(4): 905-913, 2018 04 18.
Article in English | MEDLINE | ID: mdl-29406696

ABSTRACT

Biomimetic hydrogels fabricated from biologically derived polymers, such as hyaluronic acid (HA), are useful for numerous biomedical applications. Due to the dynamic nature of biological processes, it is of great interest to synthesize hydrogels with dynamically tunable network properties where various functions (e.g., cargo delivery, mechanical signaling) can be changed over time. Among the various stimuli developed to control hydrogel properties, light stands out for its exquisite spatiotemporal control; however, most light-based chemistries are unidirectional in their ability to manipulate network changes. Here, we report a strategy to reversibly modulate HA hydrogel properties with light, using supramolecular cross-links formed via azobenzene bound to ß-cyclodextrin. Upon isomerization with 365 nm or 400-500 nm light, the binding affinity between azobenzene and ß-cyclodextrin changed and altered the network connectivity. The hydrogel mechanical properties depended on both the azobenzene modification and isomeric state (lower for cis state), with up to a 60% change in storage modulus with light exposure. Furthermore, the release of a fluorescently labeled protein was accelerated with light exposure under conditions that were cytocompatible to encapsulated cells. These results indicate that the developed hydrogels may be suitable for applications in which temporal regulation of material properties is important, such as drug delivery or mechanobiology studies.


Subject(s)
Azo Compounds/chemistry , Biomimetic Materials/chemistry , Delayed-Action Preparations/chemistry , Hyaluronic Acid/chemistry , Hydrogels/chemistry , beta-Cyclodextrins/chemistry , Animals , Cattle , Drug Liberation , Fluorescein-5-isothiocyanate/administration & dosage , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/pharmacokinetics , Isomerism , Light , Materials Testing , Mice , NIH 3T3 Cells , Serum Albumin, Bovine/administration & dosage , Serum Albumin, Bovine/pharmacokinetics
5.
ACS Biomater Sci Eng ; 3(12): 3146-3160, 2017 Dec 11.
Article in English | MEDLINE | ID: mdl-29250593

ABSTRACT

Injectable hydrogels have gained popularity as a vehicle for the delivery of cells, growth factors, and other molecules to localize and improve their retention at the injection site, as well as for the mechanical bulking of tissues. However, there are many factors, such as viscosity, storage and loss moduli, and injection force, to consider when evaluating hydrogels for such applications. There are now numerous tools that can be used to quantitatively assess these factors, including for shear-thinning hydrogels because their properties change under mechanical load. Here, we describe relevant rheological tests and ways to measure injection force using a force sensor or a mechanical testing machine toward the evaluation of injectable hydrogels. Injectable, shear-thinning hydrogels can be used in a variety of clinical applications, and as an example we focus on methods for injection into the heart, where an understanding of injection properties and mechanical forces is imperative for consistent hydrogel delivery and retention. We discuss methods for delivery of hydrogels to mouse, rat, and pig hearts in models of myocardial infarction, and compare methods of tissue postprocessing for hydrogel preservation. Our intent is that the methods described herein can be helpful in the design and assessment of shear-thinning hydrogels for widespread biomedical applications.

6.
Nat Protoc ; 12(8): 1521-1541, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28683063

ABSTRACT

The design of injectable hydrogel systems addresses the growing demand for minimally invasive approaches for local and sustained delivery of therapeutics. We developed a class of hyaluronic acid (HA) hydrogels that form through noncovalent guest-host interactions, undergo disassembly (shear-thinning) when injected through a syringe and then reassemble within seconds (self-healing) when shear forces are removed. Its unique properties enable the use of this hydrogel system for numerous applications, such as injection in vivo (including with cells and therapeutic molecules) or as a 'bioink' in 3D-printing applications. Here, we describe the functionalization of HA either with adamantanes (guest moieties) via controlled esterification or with ß-cyclodextrins (host moieties) through amidation. We also describe how to modify the HA derivatives with methacrylates for secondary covalent cross-linking and for reaction with fluorophores for in vitro and in vivo imaging. HA polymers are rationally designed from relatively low-molecular-weight starting materials, with the degree of modification controlled, and have matched guest-to-host stoichiometry, allowing the preparation of hydrogels with tailored properties. This procedure takes 3-4 weeks to complete. We detail the preparation and characterization of the guest-host hydrogels, including assessment of their rheological properties, erosion and biomolecule release in vitro. We furthermore demonstrate how to encapsulate cells in vitro and provide procedures for quantitative assessment of in vivo hydrogel degradation by imaging of fluorescently derivatized materials.


Subject(s)
Drug Carriers/administration & dosage , Hyaluronic Acid/administration & dosage , Hydrogels/administration & dosage , Hydrogels/chemistry , Printing, Three-Dimensional , Adamantane/metabolism , Animals , Injections , Mice , beta-Cyclodextrins/metabolism
7.
Soft Matter ; 12(37): 7839-7847, 2016 Sep 20.
Article in English | MEDLINE | ID: mdl-27714343

ABSTRACT

Macromolecular interactions are used to form supramolecular assemblies, including through the interaction of guest-host chemical pairs. Microstructural heterogeneity has been observed within such physical hydrogels; yet, systematic investigation of the microstructure and its determining inputs are lacking. Herein, we investigated the hierarchical self-assembly of hyaluronic acid (HA) modified by the guest-host pair adamantane (Ad-HA, guest) and ß-cyclodextrin (CD-HA, host), as well as with methacrylate groups to both tether fluorescent agents and to covalently stabilize the material structure. We observed microporous materials in the hydrated state, which temporally arose from initially homogenous hydrogels composed of the two polymers. Independent fluorescent labeling of Ad-HA and CD-HA demonstrated spatiotemporal co-localization, indicative of guest-host polymer condensation on the microscale. The hydrogel void fractions and pore diameters were independently tuned through incubation time (0-7 days), polymer concentration (1.25-10 wt%), and polymer modification (25-50% Ad-HA modification). Void fractions as great as 93.3 ± 2.4% were achieved and pore diameters ranged from 2.1 ± 0.5 to 1025.4 ± 209.4 µm. The segregation of discrete solid and solute phases was measured with both atomic force microscopy and diffusive microparticle tracking analysis, where the solute phase contained only dilute polymer. The study represents a systematic investigation of hierarchical self-assembly in binary associating hydrogels, and provides insights on mechanisms that control microstructure within supramolecular hydrogels.

8.
J Thorac Cardiovasc Surg ; 150(5): 1268-76, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26293548

ABSTRACT

OBJECTIVES: The clinical translation of cell-based therapies for ischemic heart disease has been limited because of low cell retention (<1%) within, and poor targeting to, ischemic myocardium. To address these issues, we developed an injectable hyaluronic acid (HA) shear-thinning hydrogel (STG) and endothelial progenitor cell (EPC) construct (STG-EPC). The STG assembles as a result of interactions of adamantine- and ß-cyclodextrin-modified HA. It is shear-thinning to permit delivery via a syringe, and self-heals upon injection within the ischemic myocardium. This directed therapy to the ischemic myocardial border zone enables direct cell delivery to address adverse remodeling after myocardial infarction. We hypothesize that this system will enhance vasculogenesis to improve myocardial stabilization in the context of a clinically translatable therapy. METHODS: Endothelial progenitor cells (DiLDL(+) VEGFR2(+) CD34(+)) were harvested from adult male rats, cultured, and suspended in the STG. In vitro viability was quantified using a live-dead stain of EPCs. The STG-EPC constructs were injected at the border zone of ischemic rat myocardium after acute myocardial infarction (left anterior descending coronary artery ligation). The migration of the enhanced green fluorescent proteins from the construct to ischemic myocardium was analyzed using fluorescent microscopy. Vasculogenesis, myocardial remodeling, and hemodynamic function were analyzed in 4 groups: control (phosphate buffered saline injection); intramyocardial injection of EPCs alone; injection of the STG alone; and treatment with the STG-EPC construct. Hemodynamics and ventricular geometry were quantified using echocardiography and Doppler flow analysis. RESULTS: Endothelial progenitor cells demonstrated viability within the STG. A marked increase in EPC engraftment was observed 1-week postinjection within the treated myocardium with gel delivery, compared with EPC injection alone (17.2 ± 0.8 cells per high power field (HPF) vs 3.5 cells ± 1.3 cells per HPF, P = .0002). A statistically significant increase in vasculogenesis was noted with the STG-EPC construct (15.3 ± 5.8 vessels per HPF), compared with the control (P < .0001), EPC (P < .0001), and STG (P < .0001) groups. Statistically significant improvements in ventricular function, scar fraction, and geometry were noted after STG-EPC treatment compared with the control. CONCLUSIONS: A novel injectable shear-thinning HA hydrogel seeded with EPCs enhanced cell retention and vasculogenesis after delivery to ischemic myocardium. This therapy limited adverse myocardial remodeling while preserving contractility.


Subject(s)
Endothelial Progenitor Cells/transplantation , Hyaluronic Acid/chemistry , Myocardial Ischemia/surgery , Myocardium/pathology , Regeneration , Tissue Scaffolds , Animals , Cell Movement , Cell Survival , Cells, Cultured , Disease Models, Animal , Echocardiography, Doppler , Endothelial Progenitor Cells/metabolism , Fibrosis , Genes, Reporter , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Hydrogels , Male , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myocardium/metabolism , Neovascularization, Physiologic , Rats, Wistar , Recovery of Function , Time Factors , Transfection , Ventricular Function, Left , Ventricular Pressure , Ventricular Remodeling , beta-Cyclodextrins/chemistry
9.
J Cardiovasc Transl Res ; 7(2): 242-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24429838

ABSTRACT

Hemocompatibility of tissue-engineered vascular grafts remains a major hurdle to clinical utility for small-diameter grafts. Here we assessed the feasibility of using autologous blood outgrowth endothelial cells to create an endothelium via lumenal seeding on completely biological, decellularized engineered allografts prior to implantation in the sheep femoral artery. The 4-mm-diameter, 2- to 3-cm-long grafts were fabricated from fibrin gel remodeled into an aligned tissue tube in vitro by ovine dermal fibroblasts prior to decellularization. Decellularized grafts pre-seeded with blood outgrowth endothelial cells (n = 3) retained unprecedented (>95 %) monolayer coverage 1 h post-implantation and had greater endothelial coverage, smaller wall thickness, and more basement membrane after 9-week implantation, including a final week without anti-coagulation therapy, compared with contralateral non-seeded controls. These results support the use of autologous blood outgrowth endothelial cells as a viable source of endothelial cells for creating an endothelium with biological function on decellularized engineered allografts made from fibroblast-remodeled fibrin.


Subject(s)
Blood Vessel Prosthesis Implantation/instrumentation , Blood Vessel Prosthesis , Cell Proliferation , Endothelial Cells/transplantation , Femoral Artery/surgery , Tissue Engineering/methods , Allografts , Animals , Basement Membrane/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Feasibility Studies , Femoral Artery/metabolism , Fibrin/metabolism , Models, Animal , Prosthesis Design , Pulsatile Flow , Sheep , Stress, Mechanical , Time Factors , Tissue Scaffolds
SELECTION OF CITATIONS
SEARCH DETAIL
...