WO2023102150A1 - Microfibres polymères imprégnées de nanoparticules à base de silice pour améliorer le dépôt de minéraux - Google Patents

Microfibres polymères imprégnées de nanoparticules à base de silice pour améliorer le dépôt de minéraux Download PDF

Info

Publication number
WO2023102150A1
WO2023102150A1 PCT/US2022/051589 US2022051589W WO2023102150A1 WO 2023102150 A1 WO2023102150 A1 WO 2023102150A1 US 2022051589 W US2022051589 W US 2022051589W WO 2023102150 A1 WO2023102150 A1 WO 2023102150A1
Authority
WO
WIPO (PCT)
Prior art keywords
composition
nbg
mole fraction
cao
nano
Prior art date
Application number
PCT/US2022/051589
Other languages
English (en)
Inventor
Helen H. Lu
Ming C. CHAN
Original Assignee
The Trustees Of Columbia University In The City Of New York
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Trustees Of Columbia University In The City Of New York filed Critical The Trustees Of Columbia University In The City Of New York
Publication of WO2023102150A1 publication Critical patent/WO2023102150A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C03GLASS; MINERAL OR SLAG WOOL
    • C03CCHEMICAL COMPOSITION OF GLASSES, GLAZES OR VITREOUS ENAMELS; SURFACE TREATMENT OF GLASS; SURFACE TREATMENT OF FIBRES OR FILAMENTS MADE FROM GLASS, MINERALS OR SLAGS; JOINING GLASS TO GLASS OR OTHER MATERIALS
    • C03C4/00Compositions for glass with special properties
    • C03C4/0007Compositions for glass with special properties for biologically-compatible glass
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/40Composite materials, i.e. containing one material dispersed in a matrix of the same or different material
    • A61L27/44Composite materials, i.e. containing one material dispersed in a matrix of the same or different material having a macromolecular matrix
    • A61L27/46Composite materials, i.e. containing one material dispersed in a matrix of the same or different material having a macromolecular matrix with phosphorus-containing inorganic fillers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/50Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L27/54Biologically active materials, e.g. therapeutic substances
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/50Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L27/56Porous materials, e.g. foams or sponges
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/50Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L27/58Materials at least partially resorbable by the body
    • CCHEMISTRY; METALLURGY
    • C03GLASS; MINERAL OR SLAG WOOL
    • C03CCHEMICAL COMPOSITION OF GLASSES, GLAZES OR VITREOUS ENAMELS; SURFACE TREATMENT OF GLASS; SURFACE TREATMENT OF FIBRES OR FILAMENTS MADE FROM GLASS, MINERALS OR SLAGS; JOINING GLASS TO GLASS OR OTHER MATERIALS
    • C03C1/00Ingredients generally applicable to manufacture of glasses, glazes, or vitreous enamels
    • C03C1/006Ingredients generally applicable to manufacture of glasses, glazes, or vitreous enamels to produce glass through wet route
    • CCHEMISTRY; METALLURGY
    • C03GLASS; MINERAL OR SLAG WOOL
    • C03CCHEMICAL COMPOSITION OF GLASSES, GLAZES OR VITREOUS ENAMELS; SURFACE TREATMENT OF GLASS; SURFACE TREATMENT OF FIBRES OR FILAMENTS MADE FROM GLASS, MINERALS OR SLAGS; JOINING GLASS TO GLASS OR OTHER MATERIALS
    • C03C13/00Fibre or filament compositions
    • CCHEMISTRY; METALLURGY
    • C03GLASS; MINERAL OR SLAG WOOL
    • C03CCHEMICAL COMPOSITION OF GLASSES, GLAZES OR VITREOUS ENAMELS; SURFACE TREATMENT OF GLASS; SURFACE TREATMENT OF FIBRES OR FILAMENTS MADE FROM GLASS, MINERALS OR SLAGS; JOINING GLASS TO GLASS OR OTHER MATERIALS
    • C03C3/00Glass compositions
    • C03C3/04Glass compositions containing silica
    • C03C3/062Glass compositions containing silica with less than 40% silica by weight
    • CCHEMISTRY; METALLURGY
    • C03GLASS; MINERAL OR SLAG WOOL
    • C03CCHEMICAL COMPOSITION OF GLASSES, GLAZES OR VITREOUS ENAMELS; SURFACE TREATMENT OF GLASS; SURFACE TREATMENT OF FIBRES OR FILAMENTS MADE FROM GLASS, MINERALS OR SLAGS; JOINING GLASS TO GLASS OR OTHER MATERIALS
    • C03C3/00Glass compositions
    • C03C3/04Glass compositions containing silica
    • C03C3/076Glass compositions containing silica with 40% to 90% silica, by weight
    • C03C3/097Glass compositions containing silica with 40% to 90% silica, by weight containing phosphorus, niobium or tantalum
    • CCHEMISTRY; METALLURGY
    • C03GLASS; MINERAL OR SLAG WOOL
    • C03CCHEMICAL COMPOSITION OF GLASSES, GLAZES OR VITREOUS ENAMELS; SURFACE TREATMENT OF GLASS; SURFACE TREATMENT OF FIBRES OR FILAMENTS MADE FROM GLASS, MINERALS OR SLAGS; JOINING GLASS TO GLASS OR OTHER MATERIALS
    • C03C4/00Compositions for glass with special properties
    • C03C4/0007Compositions for glass with special properties for biologically-compatible glass
    • C03C4/0021Compositions for glass with special properties for biologically-compatible glass for dental use
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/40Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a specific therapeutic activity or mode of action
    • A61L2300/404Biocides, antimicrobial agents, antiseptic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2400/00Materials characterised by their function or physical properties
    • A61L2400/12Nanosized materials, e.g. nanofibres, nanoparticles, nanowires, nanotubes; Nanostructured surfaces
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2430/00Materials or treatment for tissue regeneration
    • A61L2430/02Materials or treatment for tissue regeneration for reconstruction of bones; weight-bearing implants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2430/00Materials or treatment for tissue regeneration
    • A61L2430/12Materials or treatment for tissue regeneration for dental implants or prostheses
    • CCHEMISTRY; METALLURGY
    • C03GLASS; MINERAL OR SLAG WOOL
    • C03CCHEMICAL COMPOSITION OF GLASSES, GLAZES OR VITREOUS ENAMELS; SURFACE TREATMENT OF GLASS; SURFACE TREATMENT OF FIBRES OR FILAMENTS MADE FROM GLASS, MINERALS OR SLAGS; JOINING GLASS TO GLASS OR OTHER MATERIALS
    • C03C2205/00Compositions applicable for the manufacture of vitreous enamels or glazes
    • C03C2205/06Compositions applicable for the manufacture of vitreous enamels or glazes for dental use

Definitions

  • Bone fractures remain a global burden on public health. Over 2 million bone graft procedures are performed annually (2nd most frequent tissue transplantation) in the world (J.W. Lee et. al. 2016) ( Figure 1). Bone fractures usually result loss of mobility, impaired quality of life and increased healthcare costs. The most common treatments for patients suffering from slow or incomplete bone healing are autologous or allogenic bone grafts. Autologous bone grafts require harvesting large bone volumes from the patient, while allogenic bone grafts carry a high risk of disease transmission (Suchomel et al. 2004). These treatments have inherent limitations including a lack of graft availability, donor site morbidity and disease transmission. Therefore, there is a need for a treatment approach that is more biomimetic and improves material tissue integration.
  • Tissue-engineered bone grafts using bioactive glasses are a promising alternative because the glasses strongly bind to bone (Gao et al. 2014).
  • Composite materials such as Bioactive Glass nanofibers (Figure 3A), Particle filled/coated Polymer, Bioactive Glass -Hydrogel, Particle filled/coated scaffold, BG nanoparticle-Polymer scaffold, are manufactured to improve mechanical properties and modulate ion release.
  • bioactive glass is fabricated using melt-quenching technique at extreme temperatures and it dissolves in physiological fluid and bonds to native bone.
  • a ternary system SiCf-CaO- P2O5 fabricated via Sol-gel processing is developed ( Figure 3B) .
  • BG nanocomposite platform may provide 3D structure template to guide bone healing.
  • bio-glasses are brittle and unable to share load with the bone.
  • nBG nano-bioactive glass
  • nBG nano-bioactive glass
  • the nBG comprises S i O 2- CaO and P2O5, and wherein CaO has at least 40% mole fraction.
  • nBG nano-bioactive glass
  • the nBG comprises SiO2, CaO and P2O5, and wherein P2O5 has at least 5% mole fraction.
  • composition in the manufacture of a medicament for treating bone fractures on a subject may be provided.
  • a method of producing a nano-bioactive glass may comprise:
  • a method of stimulating bone regeneration may be performed by treating a subject with the composition disclosed in the present invention.
  • a method of repairing bone fractures may be performed by treating a subject with the composition disclosed in the present invention.
  • Figure 1 An X-ray showing Bone fracture.
  • SiO2 enhances osteoblast differentiation and mineralization in vitro.
  • Figure 3A 45S5 Bioactive Glass (Biogalss®), containing 46.1 mol.% SiCL, 24.4 mol.%Na2O, 26.9 mol. % CaO and 2.6 mol.% P2O5.
  • FIG. 3B Ternary system (SiO2-CaO-P2O5) via Sol-gel processing.
  • Figure 9A Morphology of nBG.
  • FIG. 11 A SEM of PLGA (5: 1) PCL Fibers.
  • FIG 11 B FTIR and XDR of PLGA (5: 1) PCL Fibers.
  • Figure 12 A SEM of nBG Fibers.
  • Figure 12 B FTIR and XDR of nBG Fibers.
  • Figure 14 A FTIR of nBG after 14 days.
  • Figure 14 B Crystallinity change of nBG after 14 days.
  • FIG. 15 A Bioactivity Test of PLGA (5: 1) PCL.
  • FIG. 17 A Cell Viability at Day 1 and Day 7.
  • nBG nano-bioactive glass
  • the nBG comprises SiCh, CaO and P2O5, and wherein SiO2has less than 60% mole fraction.
  • the mole fraction of CaO is at least 40%.
  • the mole fraction of P2O5 is at least 5%.
  • nBG nano-bioactive glass
  • the nBG comprises SiO2, CaO and P2O5, wherein CaO has at least 40% mole fraction.
  • the mole fraction of SiO2 is less than 60%.
  • the mole fraction of P2O5 is at least 5%.
  • nBG nano -bioactive glass
  • the nBG comprises SiO2, CaO and P2O5, wherein P2O5has at least 5% mole fraction.
  • the mole fraction of SiO2 is less than 60%.
  • the mole fraction of CaO is at least 40%.
  • the mole fraction of SiO2 is 30%-60%.
  • the mole fraction of SiO2 is 40%-55%.
  • the mole fraction of CaO is 37%-57%.
  • the mole fraction of P2O5 is 5%-10%.
  • the mole fraction of SiO2 is 55%.
  • the mole fraction of CaO is 40%.
  • a polymer-bioactive glass fiber composite can be provided.
  • the porous structure of the composite allows for controlled release of silica and calcium phosphate ions.
  • the polymer component allows for enhanced flexibility.
  • the composite is biocompatible and enhances bioactivity via mineralization. Such composite may promote cell attachment, and therefore promote bone regeneration, and have dental and orthopedic applications.
  • the microarchitecture of bone indicates that bone is a biological composite material. It is composed mainly type I collagen fibers and calcium phosphate/hydroxyapatite crystals. Mineralized collagen is aggregated into small fibrils, which further combine to form fibers a few microns in diameter and several mm long. For trabecular bone, the fibers are randomly laid out. For the cortical bone, fibers are wrapped around a hollow core. Since the advent of tissue engineering, many strategies have an aim to mimic the bone microarchitecture in order to enhance and facilitate tissue regeneration. The Hydroxyapatite (HA) nanocrystals are principally arranged with their c-axes parallel to the collagen fibrils and organized in a periodic, staggered arrangement along the fibrils.
  • HA Hydroxyapatite
  • This biomineralization process of bone proceeds via a matrix vesicle-mediated mechanism, in which the matrix vesicles are secreted by the outer membranes of bone-forming osteoblasts.
  • the enzyme alkaline phosphatase present in the matrix vesicles cleaves the phosphate esters and acts as the foci for calcium and phosphate deposition (Wei et al. 2007).
  • the self-assembly process of collagen molecules defines the framework and spatial constraints for HA nucleation and propagation
  • Bioactive glass is a synthetic material which is able to bond to bone via the formation of an apatite layer on their surface (Rezwan et al. 2006). This material was first developed in the 1970s. The composition of the first bioglass contained a main silica network which contained CaO and P2O5, the natural constituents of bone and silica enhancing osteoblast differentiation and mineralization (Yunos et al. 2013, Kumar et al. 2019). Traditionally it is fabricated using a high temperature melt-quenching technique. However, with the improvement of technology, now bioactive glass can be fabricated under ambient conditions, typically using a ternary system (Magazzini et al. 2002).
  • the mole fraction of P2O5 is 5%.
  • composition of nano-bioactive glass further comprising a polymer blend of at least two polymers.
  • the polymer blend consists of two polymers
  • the polymer blend consists of three polymers.
  • the polymer amongst the polymers comprises aliphatic polyesters, poly( amino acids), copoly (ether-esters), polyalkylenes oxalates, polyamides, tyrosine derived polycarbonates, poly(iminocarbonates), polyorthoe-sters, polyoxaesters, polyamidoesters, polyoxaesters containing amine groups, poly(anhydrides), polyphosphazenes, polydiglycolates, polylactic-co-glycolic acid (PLGA) and poly(8-caprolactone) (PCL), or any combination thereof.
  • the polymer comprises polylactic-co-glycolic acid (PLGA) and poly(s-caprolactonc) (PCL).
  • PLGA polylactic-co-glycolic acid
  • PCL poly(s-caprolactonc)
  • the polymer blend consists of polylactic-co-glycolic acid (PLGA) and poly(s- caprolactone) (PCL).
  • PLGA polylactic-co-glycolic acid
  • PCL poly(s- caprolactone)
  • the mass ratio between PLGA and PCL is 1: 1 to 10: 1.
  • the mass ratio between PLGA and PCL is 3: 1 to 7: 1.
  • the mass ratio between PLGA and PCL is 5 : 1.
  • the polymer amongst the polymers is a biocompatible polymer.
  • the biocompatible polymer has a diameter between 1.50-3.00 pm.
  • the composition of nano-bioactive glass (nBG) further comprising a therapeutic agent.
  • the therapeutic agent is selected from the group consisting of antibiotics, antivirals, adhesion preventative s, contraceptives, and analgesics.
  • composition of nano-bioactive glass further comprising an antimicrobial agent.
  • the antimicrobial agent is poly chloro phenoxy phenol.
  • a method of producing a nano-bioactive glass comprises: a) mixing calcium nitrate solution with Tetraethoxysilane (TEOS) solution; b) adding dropwise of a mixture from step a) to an ammonium dibasic phosphate solution; c) stirring the mixture from step b) for 30-60 hours and permitting the reaction mixture to rest for 20-30 hours for further precipitation; d) separating a precipitate from step c); e) freezing the precipitate from step d) at -60 to -40 °C for 20-30 hours; and f) heating a sample of the precipitate from step e) at 650 to 800 °C for 2-5 hours and cool down the heated precipitate for 8-14 hours.
  • TEOS Tetraethoxysilane
  • step a) the TEOS was dissolved in anhydrous ethanol.
  • step a) is conducted at a pH between 1-2 and a reaction mixture is stirred for 20-40 minutes.
  • step a) is conducted at a pH between 1-2 and a reaction mixture is stirred for 30 minutes.
  • step b) further comprises adding ammonium hydroxide solution to adjust the pH to be between 9-12. In some embodiments, step b) further comprises adding ammonium hydroxide solution to adjust the pH to 11.
  • step d) further comprises freezing the precipitate at -20-30 °C for 8-14 hours.
  • step d) further comprises freezing the precipitate at-25 °C for 12 hours.
  • a method of stimulating bone regeneration by treating a subject with the composition disclosed in the present application is disclosed.
  • composition in the manufacture of a medicament for treating bone fractures on a subject is provided.
  • the treatment comprises oral administration or surgical implantation of the composition.
  • the subject of treatment is a mammal.
  • the subject of treatment is human.
  • the subject was treated for dental treatment, or orthopedic surgery.
  • a numeric value may have a value that is ⁇ 1 % of the stated value (or range of values), ⁇ 2% of the stated value (or range of values), ⁇ 5% of the stated value (or range of values), ⁇ 10% of the stated value (or range of values), ⁇ 15% of the stated value (or range of values), or ⁇ 20% of the stated value (or range of values).
  • the nano-bioactive glass comprises at least one alkali metal such as, for example, lithium, sodium, potassium, rubidium, cesium, francium, or combinations of these metals. In other embodiments, however, the nano-bioactive glass has little to no alkali metal. For example, in certain embodiments, the nano-bioactive glass has 30% or less of alkali metal. In other embodiments, the nanobioactive glass has 25% or less of alkali metal. In yet other embodiments, the nano-bioactive glass has 20% or less of alkali metal. In yet other embodiments, the nano-bioactive glass has 15% or less of alkali metal.
  • alkali metal such as, for example, lithium, sodium, potassium, rubidium, cesium, francium, or combinations of these metals. In other embodiments, however, the nano-bioactive glass has little to no alkali metal. For example, in certain embodiments, the nano-bioactive glass has 30% or less of alkali metal. In other embodiments, the nanobioactive glass has 25% or less of al
  • the nano-bioactive glass has 10% or less of alkali metal. In still other embodiments, the nano-bioactive glass has 5% or less of alkali metal. In yet other embodiments, the nano-bioactive glass has substantially no alkali metal. Without intending to be bound by any particular theory, it is believed that the presence of certain metals may catalyze further polymerization of the biocompatible polymer such as, for example, PLGA or PCL, thereby (1) increasing its molecular weight and/or (2) increasing its degree of cross-linking/cross-link density. Either event increases the viscosity of the polymer and may seize up the equipment used to process the com-posite material. As such, a nano-bioactive glass with a low percentage of alkali metal may be utilized to prevent equip-ment failure and/or to allow a high percentage of nanobioactive glass to be utilized.
  • the biocompatible polymer such as, for example, PLGA or PCL
  • the nano-bioactive glass has osteoproductive properties.
  • osteoproductive refers to an ability to allow osteoblasts to proliferate, allowing bone to regenerate.
  • Oxsteoproductive may also be defined as conducive to a process in which a bioactive surface is colonized by osteogenic stem cells and which results in more rapid filling of defects than that produced by merely osteoconductive materials.
  • nBG fiber as disclosed in the present invention is an example of an osteoproductive, bioactive material.
  • Osteoconductivity of a biomaterial is largely dependent on the biocompatibility and bioactivity of the biomaterial's surface [Smith D C, Pilliar R M and Chemecky R 1991 Dental implant materials. I. Some effects of preparative procedures on surface topography J of Biomedical Materials Research 25 1045-68], Factors affecting properties of hydroxyapatite in cell adhesion and cell proliferation include its structure, phase purity, porosity, surface properties and sintering temperature [Khan A, Wong F, McKay I, Whiley R, Rehman I. Structural, mechanical, and biocompatibility analyses of a novel dental restorative nanocomposite.
  • the water used herein may be tap water, distilled water, bidistilled water, deionized water, deionized distilled water, reverse osmosis water, and/or some other water.
  • the water is bidistilled to eliminate trace met-als.
  • the water is bidistilled, deionized, deinonized distilled, or reverse osmosis water and at 25° C.
  • a conductivity at less than 10 pS cm 1 preferably less than 1 pS cm 1 , a resistivity greater than 0.1 MQ cm, preferably greater than 1 MQ cm, more preferably greater than 10 MQ cm, a total solid concentration less than 5 mg/kg, preferably less than 1 mg/kg, and a total organic carbon concentration less than 1000 pg/L, preferably less than 200 pg/L, more preferably less than 50 pg/L.
  • the nBG fiber may comprise up to about 50% of the nano-bioactive glass.
  • the nano-bioactive glass is present in an amount of about 5 to 50% by weight of the compounded composite material.
  • the nano-bioactive glass is present in an amount of about 15 to 30% by weight of the nBG fiber.
  • the nano bioactive glass is present in an amount of about 20 to 30% by weight of the nBG fiber.
  • bioactive glass may be present in higher weight percentages, such as 60% by weight of the compounded composite material.
  • a coupling agent is added to the mixture of the biocompatible polymer and the nano-bioactive glass.
  • the coupling agent acts as a bonding agent between the biocompatible polymer and the nano-bioactive glass which translates into increased tensile/flexural strength of the nano-bioactive composite.
  • Non-limiting examples of coupling agents suitable for use in the present invention include, for example, silane, titanium-based and zirconium-based coupling agents, specifically, organotitanate, multifunctional amine compounds such as 4-aminophenyl sulfone, azo compounds such as 4-cyanovaleric acid, and combinations thereof.
  • the preferred coupling agent is one that includes multifunctional groups that are capable of chemically bonding with a functional group of the biocompatible polymer and binding the bioactive glass.
  • the nano-bioactive glass may be coated with the coupling agent prior to being combined/ mixed with the biocompatible polymer.
  • both the nano-bioactive glass and biocompatible polymer may be individually coated with the coupling agent before being combined.
  • At least one other agent may be added to the mixture of the biocompatible polymer and nano-bioactive glass.
  • Such agents can comprise, at least partially, reinforcing fibers.
  • Non-limiting examples of other agents include carbon, glass, radiopaque material, barium glass, resorbable material, strontium, strontium nitrate, strontium -calcium-zinc-silicate glasses, silver, calcium apatite, calcium silicate or mixtures of these materials.
  • the other agent is barium sulfate, barium- boroaluminosilicate (BBAS) glass, silica or e-glass fibers.
  • BBAS barium- boroaluminosilicate
  • the other agents include radiopaque markers situated in predetermined locations within the shaped implant to aid in visualizing the implant once in the body.
  • FIGS. Sa and Sb show titanium alloy (Ti-6A1-4V ELI) markers incorporated into the composite shaped body.
  • the other agent may comprise calcium phosphate having macro-, mesa-, and microporosity. More preferably, the porosity of the calcium phosphate is interconnected. The preparation of preferred forms of calcium phosphate for use in the present invention is described in U.S. Pat. No. 6,383,519 and U.S. Pat. No. 6,521,246, incorporated into this application by reference in their entireties.
  • the at least one other agent may be incorporated within the bioactive composite, or in the case of a shaped implant be used to fill cavities of the implant. For instance, when used with a shaped spinal implant, the other agent may be present within the center cavity of the implant to facilitate fusion of the adjacent vertebral bodies.
  • bone augmentation materials or bone cements may be used in conjunction with the bioactive composite (nBG fiber) in applications where additional reinforcement is required.
  • nBG fiber bioactive composite
  • An exemplary bone augmen-tation product is Cortoss® Bone Augmentation Material (available from Orthovita, Inc. of Malvern, Pa.).
  • a nBG fiber (bioactive composite) is formed upon combining a biocompatible polymer with a nano-bioactive glass as described in the present invention.
  • the biocompatible polymer preferably has a diameter of 1.5 - 3 pm.
  • the biocompatible polymer and the nano-bioactive glass may be dry mixed for a period of time and under conditions sufficient to achieve substantial homogeneity of the mixture.
  • dry mixed refers to mixing the components in a dry state, i.e., in the absence of added liquid water or organic solvent.
  • the dry mixing of the bioactive glass with the biocompatible polymer granules or pellets may be accomplished using any methods known in the art per se, including milling, spinning, tumbling, sonication, vibrating, or shaking.
  • the mixture is tumbled on rollers for about one to about two hours.
  • homogeneity and “homogeneous” describe a composition that is substantially uniform in structure and/or composition throughout.
  • substantially homogeneous is to be understood within the context of the invention and is not to be taken as an absolute.
  • suitable polymers that may be included in the polymer blends of the present invention include: suitable biocompat-ible, biodegradable polymers which may be synthetic or natu-ral polymers.
  • suitable synthetic biocompatible, biodegrad-able polymers include polymers selected from the group consisting of aliphatic polyesters, poly( amino acids), copoly (ether-esters), polyalkylenes oxalates, polyamides, tyrosine derived polycarbonates, poly(iminocarbonates), polyorthoe-sters, polyoxaesters, polyamidoesters, polyoxaesters containing amine groups, poly(anhydrides), polyphosphazenes, polydiglycolates, polylactic-co-glycolic acid (PLGA) and poly(s-caprolactonc) (PCL), and combinations thereof. It is to be understood that inclusion of additional suitable polymers is dependent upon obtaining dimensional stability in the fabricated device.
  • the above optional aliphatic polyesters include, but are not limited to, homopolymers and copolymers of lactide (which include lactic acid, D-, L- and mesa lactide), glycolide (including glycolic acid), epsilon-caprolactone, p-dioxanone (1 ,4-dioxan-2-one ), trimethylene carbonate (l,3-dioxan-2-one), alkyl derivatives of trimethylene carbonate, and blends thereof.
  • lactide which include lactic acid, D-, L- and mesa lactide
  • glycolide including glycolic acid
  • epsilon-caprolactone p-dioxanone (1 ,4-dioxan-2-one
  • trimethylene carbonate l,3-dioxan-2-one
  • alkyl derivatives of trimethylene carbonate and blends thereof.
  • the term “activity” refers to the activation, production, expression, synthesis, intercellular effect, and/or pathological or aberrant effect of the referenced molecule, either inside and/or outside of a cell.
  • Such molecules include, but are not limited to, cytokines, enzymes, growth factors, pro-growth factors, active growth factors, and pro-enzymes. Molecules such as cytokines, enzymes, growth factors, pro-growth factors, active growth factors, and pro-enzymes may be produced, expressed, or synthesized within a cell where they may exert an effect. Such molecules may also be transported outside of the cell to the extracellular matrix where they may induce an effect on the extracellular matrix or on a neighboring cell.
  • inactive cytokines activation of inactive cytokines, enzymes and pro-enzymes may occur inside and/or outside of a cell and that both inactive and active forms may be present at any point inside and/or outside of a cell. It is also understood that cells may possess basal levels of such molecules for normal function and that abnormally high or low levels of such active molecules may lead to pathological or aberrant effects that may be corrected by pharmacological intervention.
  • the present invention may also provide isotopic variants of the compounds disclosed herein, including wherein the isotopic atom is 2 H and/or wherein the isotopic atom 13 C. Accordingly, in the compounds provided herein hydrogen can be enriched in the deuterium isotope. It is to be understood that the invention encompasses all such isotopic forms. It is understood that the structures described in the embodiments of the methods hereinabove can be the same as the structures of the compounds described hereinabove.
  • each stereogenic carbon may be of the R or S configuration.
  • isomers arising from such asymmetry e.g., all enantiomers and diastereomers
  • Such isomers can be obtained in substantially pure form by classical separation techniques and by stereochemically controlled synthesis, such as those described in "Enantiomers, Racemates and Resolutions" by J. Jacques, A. Collet and S. Wilen, Pub. John Wiley & Sons, NY, 1981.
  • the resolution may be carried out by preparative chromatography on a chiral column.
  • the subject invention is also intended to include all isotopes of atoms occurring on the compounds disclosed herein.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • isotopes of hydrogen include tritium and deuterium.
  • isotopes of carbon include C-13 and C-14.
  • any notation of a carbon in structures throughout this application when used without further notation, are intended to represent all isotopes of carbon, such as 12 C, 13 C, or 14 C.
  • any compounds containing 13 C or 14 C may specifically have the structure of any of the compounds disclosed herein.
  • any notation of a hydrogen in structures throughout this application when used without further notation, are intended to represent all isotopes of hydrogen, such as 1 H, 2 H, or 3 H.
  • any compounds containing 2 H or 3 H may specifically have the structure of any of the compounds disclosed herein.
  • Isotopically-labeled compounds can generally be prepared by conventional techniques known to those skilled in the art using appropriate isotopically-labeled reagents in place of the non-labeled reagents employed.
  • the substituents may be substituted or unsubstituted, unless specifically defined otherwise.
  • alkyl, heteroalkyl, monocycle, bicycle, aryl, heteroaryl and heterocycle groups can be further substituted by replacing one or more hydrogen atoms with alternative non-hydrogen groups.
  • non-hydrogen groups include, but are not limited to, halo, hydroxy, mercapto, amino, carboxy, cyano and carbamoyl.
  • substituents and substitution patterns on the compounds used in the method of the present invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art from readily available starting materials. If a substituent is itself substituted with more than one group, it is understood that these multiple groups may be on the same carbon or on different carbons, so long as a stable structure result.
  • the compounds used in the method of the present invention may be prepared by techniques well known in organic synthesis and familiar to a practitioner ordinarily skilled in the art. However, these may not be the only means by which to synthesize or obtain the desired compounds.
  • the compounds used in the method of the present invention may be prepared by techniques described in Vogel’s Textbook of Practical Organic Chemistry, A. I. Vogel, A.R. Tatchell, B.S. Fumis, A. J. Hannaford, P.W.G. Smith, (Prentice Hall) 5 th Edition (1996), March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, Michael B. Smith, Jerry March, (Wiley-Interscience) 5 th Edition (2007), and references therein, which are incorporated by reference herein. However, these may not be the only means by which to synthesize or obtain the desired compounds.
  • Another aspect of the invention comprises a compound used in the method of the present invention as a pharmaceutical composition.
  • a pharmaceutical composition comprising the nBG Fiber disclosed in the present invention and a pharmaceutically acceptable carrier.
  • the term “pharmaceutically active agent” means any substance or compound suitable for administration to a subject and furnishes biological activity or other direct effect in the treatment, cure, mitigation, diagnosis, or prevention of disease, or affects the structure or any function of the subject.
  • Pharmaceutically active agents include, but are not limited to, substances and compounds described in the Physicians’ Desk Reference (PDR Network, LLC; 64th edition; November 15, 2009) and “Approved Drug Products with Therapeutic Equivalence Evaluations” (U.S. Department Of Health And Human Services, 30 th edition, 2010), which are hereby incorporated by reference.
  • compositions which have pendant carboxylic acid groups may be modified in accordance with the present invention using standard esterification reactions and methods readily available and known to those having ordinary skill in the art of chemical synthesis. Where a pharmaceutically active agent does not possess a carboxylic acid group, the ordinarily skilled artisan will be able to design and incorporate a carboxylic acid group into the pharmaceutically active agent where esterification may subsequently be carried out so long as the modification does not interfere with the pharmaceutically active agent’s biological activity or effect.
  • the compounds used in the method of the present invention may be in a salt form.
  • a “salt” is a salt of the instant compounds which has been modified by making acid or base salts of the compounds.
  • the salt is pharmaceutically acceptable.
  • pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as phenols.
  • the salts can be made using an organic or inorganic acid.
  • Such acid salts are chlorides, bromides, sulfates, nitrates, phosphates, sulfonates, formates, tartrates, maleates, malates, citrates, benzoates, salicylates, ascorbates, and the like.
  • Phenolate salts are the alkaline earth metal salts, sodium, potassium or lithium.
  • pharmaceutically acceptable salt in this respect, refers to the relatively non-toxic, inorganic and organic acid or base addition salts of compounds of the present invention.
  • salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or by separately reacting a purified compound of the invention in its free base or free acid form with a suitable organic or inorganic acid or base, and isolating the salt thus formed.
  • Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, napthylate, mesylate, glucoheptonate, lactobionate, and lauryl sulphonate salts and the like. (See, e.g., Berge et al. (1977) "Pharmaceutical Salts", Pharm. Set. 66: 1-19).
  • treating means preventing, slowing, halting, or reversing the progression of a disease or infection. Treating may also mean improving one or more symptoms of a disease or infection.
  • the compounds used in the method of the present invention may be administered in various forms, including those detailed herein.
  • the treatment with the compound may be a component of a combination therapy or an adjunct therapy, i.e. the subject or patient in need of the drug is treated or given another drug for the disease in conjunction with one or more of the instant compounds.
  • This combination therapy can be sequential therapy where the patient is treated first with one drug and then the other or the two drugs are given simultaneously.
  • These can be administered independently by the same route or by two or more different routes of administration depending on the dosage forms employed.
  • a "pharmaceutically acceptable carrier” is a pharmaceutically acceptable solvent, suspending agent or vehicle, for delivering the instant compounds to the animal or human.
  • the carrier may be liquid or solid and is selected with the planned manner of administration in mind.
  • Liposomes are also a pharmaceutically acceptable carrier.
  • the dosage of the compounds administered in treatment will vary depending upon factors such as the pharmacodynamic characteristics of a specific chemotherapeutic agent and its mode and route of administration; the age, sex, metabolic rate, absorptive efficiency, health and weight of the recipient; the nature and extent of the symptoms; the kind of concurrent treatment being administered; the frequency of treatment with; and the desired therapeutic effect.
  • a dosage unit of the compounds used in the method of the present invention may comprise a single compound or mixtures thereof with additional antibacterial agents.
  • the compounds can be administered in oral dosage forms as tablets, capsules, pills, powders, granules, elixirs, tinctures, suspensions, syrups, and emulsions.
  • the compounds may also be administered in intravenous (bolus or infusion), intraperitoneal, subcutaneous, or intramuscular form, or introduced directly, e.g. by injection, topical application, or other methods, into or onto a site of infection, all using dosage forms well known to those of ordinary skill in the pharmaceutical arts.
  • the compounds used in the method of the present invention can be administered in admixture with suitable pharmaceutical diluents, extenders, excipients, or carriers (collectively referred to herein as a pharmaceutically acceptable carrier) suitably selected with respect to the intended form of administration and as consistent with conventional pharmaceutical practices.
  • a pharmaceutically acceptable carrier suitably selected with respect to the intended form of administration and as consistent with conventional pharmaceutical practices.
  • the unit will be in a form suitable for oral, rectal, topical, intravenous or direct injection or parenteral administration.
  • the compounds can be administered alone or mixed with a pharmaceutically acceptable carrier.
  • This carrier can be a solid or liquid, and the type of carrier is generally chosen based on the type of administration being used.
  • the active agent can be co-administered in the form of a tablet or capsule, liposome, as an agglomerated powder or in a liquid form.
  • suitable solid carriers include lactose, sucrose, gelatin and agar.
  • Capsule or tablets can be easily formulated and can be made easy to swallow or chew; other solid forms include granules, and bulk powders. Tablets may contain suitable binders, lubricants, diluents, disintegrating agents, coloring agents, flavoring agents, flow-inducing agents, and melting agents.
  • suitable liquid dosage forms include solutions or suspensions in water, pharmaceutically acceptable fats and oils, alcohols or other organic solvents, including esters, emulsions, syrups or elixirs, suspensions, solutions and/or suspensions reconstituted from non-efferve scent granules and effervescent preparations reconstituted from effervescent granules.
  • Such liquid dosage forms may contain, for example, suitable solvents, preservatives, emulsifying agents, suspending agents, diluents, sweeteners, thickeners, and melting agents.
  • Oral dosage forms optionally contain flavorants and coloring agents.
  • Parenteral and intravenous forms may also include minerals and other materials to make them compatible with the type of injection or delivery system chosen.
  • Tablets may contain suitable binders, lubricants, disintegrating agents, coloring agents, flavoring agents, flow -inducing agents, and melting agents.
  • the active drug component can be combined with an oral, non-toxic, pharmaceutically acceptable, inert carrier such as lactose, gelatin, agar, starch, sucrose, glucose, methyl cellulose, magnesium stearate, dicalcium phosphate, calcium sulfate, mannitol, sorbitol and the like.
  • Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, com sweeteners, natural and synthetic gums such as acacia, tragacanth, or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes, and the like.
  • Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, and the like.
  • Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum, and the like.
  • the compound/composition used in the method of the present invention may also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamallar vesicles, and multilamellar vesicles.
  • Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine, or phosphatidylcholines.
  • the compounds may be administered as components of tissue- targeted emulsions.
  • the compound/composition used in the method of the present invention may also be coupled to soluble polymers as targetable drug carriers or as a prodrug.
  • soluble polymers include polyvinylpyrrolidone, pyran copolymer, polyhydroxylpropylmethacrylamide-phenol, polyhydroxyethylasparta-midephenol, or polyethyleneoxide-polylysine substituted with palmitoyl residues.
  • the compounds may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyglycolic acid, copolymers of polylactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacylates, and crosslinked or amphipathic block copolymers of hydrogels.
  • a class of biodegradable polymers useful in achieving controlled release of a drug
  • a drug for example, polylactic acid, polyglycolic acid, copolymers of polylactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacylates, and crosslinked or amphipathic block copolymers of hydrogels.
  • Gelatin capsules may contain the active ingredient compounds and powdered carriers, such as lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, and the like. Similar diluents can be used to make compressed tablets. Both tablets and capsules can be manufactured as immediate release products or as sustained release products to provide for continuous release of medication over a period of hours. Compressed tablets can be sugar coated or film coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric coated for selective disintegration in the gastrointestinal tract.
  • the oral drug components are combined with any oral, nontoxic, pharmaceutically acceptable inert carrier such as ethanol, glycerol, water, and the like.
  • suitable liquid dosage forms include solutions or suspensions in water, pharmaceutically acceptable fats and oils, alcohols or other organic solvents, including esters, emulsions, syrups or elixirs, suspensions, solutions and/or suspensions reconstituted from non-effervescent granules and effervescent preparations reconstituted from effervescent granules.
  • Such liquid dosage forms may contain, for example, suitable solvents, preservatives, emulsifying agents, suspending agents, diluents, sweeteners, thickeners, and melting agents.
  • Liquid dosage forms for oral administration can contain coloring and flavoring to increase patient acceptance.
  • water a suitable oil, saline, aqueous dextrose (glucose), and related sugar solutions and glycols such as propylene glycol or polyethylene glycols are suitable carriers for parenteral solutions.
  • Solutions for parenteral administration preferably contain a water soluble salt of the active ingredient, suitable stabilizing agents, and if necessary, buffer substances.
  • Antioxidizing agents such as sodium bisulfite, sodium sulfite, or ascorbic acid, either alone or combined, are suitable stabilizing agents.
  • citric acid and its salts and sodium EDTA are also used.
  • parenteral solutions can contain preservatives, such as benzalkonium chloride, methyl- or propyl-paraben, and chlorobutanol.
  • preservatives such as benzalkonium chloride, methyl- or propyl-paraben, and chlorobutanol.
  • Suitable pharmaceutical carriers are described in Remington's Pharmaceutical Sciences, Mack Publishing Company, a standard reference text in this field.
  • the compound/composition used in the method of the present invention may also be administered in intranasal form via use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in that art.
  • the dosage administration will generally be continuous rather than intermittent throughout the dosage regimen.
  • Parenteral and intravenous forms may also include minerals and other materials to make them compatible with the type of injection or delivery system chosen.
  • the compounds and compositions of the present invention can be administered in oral dosage forms as tablets, capsules, pills, powders, granules, elixirs, tinctures, suspensions, syrups, and emulsions.
  • the compounds may also be administered in intravenous (bolus or infusion), intraperitoneal, subcutaneous, or intramuscular form, or introduced directly, e.g. by topical administration, injection or other methods, to the afflicted area, such as a wound, including ulcers of the skin, all using dosage forms well known to those of ordinary skill in the pharmaceutical arts.
  • prodrug refers to any compound that when administered to a biological system generates the compound of the invention, as a result of spontaneous chemical reaction(s), enzyme catalyzed chemical reaction(s), photolysis, and/or metabolic chemical reaction(s).
  • a prodrug is thus a covalently modified analog or latent form of a compound of the invention.
  • the active ingredient can be administered orally in solid dosage forms, such as capsules, tablets, powders, and chewing gum; or in liquid dosage forms, such as elixirs, syrups, and suspensions, including, but not limited to, mouthwash and toothpaste. It can also be administered parentally, in sterile liquid dosage forms.
  • Solid dosage forms such as capsules and tablets, may be enteric coated to prevent release of the active ingredient compounds before they reach the small intestine.
  • Materials that may be used as enteric coatings include, but are not limited to, sugars, fatty acids, waxes, shellac, cellulose acetate phthalate (CAP), methyl acrylate-methacrylic acid copolymers, cellulose acetate succinate, hydroxy propyl methyl cellulose phthalate, hydroxy propyl methyl cellulose acetate succinate (hypromellose acetate succinate), polyvinyl acetate phthalate (PVAP), and methyl methacrylate -methacrylic acid copolymers.
  • CAP cellulose acetate phthalate
  • PVAP polyvinyl acetate phthalate
  • the compounds and compositions of the invention can be coated onto stents for temporary or permanent implantation into the cardiovascular system of a subject.
  • nBG nano-Bioactive Glass
  • BGNPs Bioactive Glass Nanoparticles
  • TEOS, Calcium Nitrate and Ammonium Dibasic Phosphate are first dissolved in ethanol and water to facilitate reaction with each other.
  • Citric acid is used as a catalyst to initiate the hydrolysis reaction with TEOS to form the glass network.
  • Addition of calcium nitrate during the reaction is to act as a network modifier to improve the dissolution of bioactive glass allowing HCA layer deposition.
  • Ammonium dibasic phosphate also provides a glass network to improve dissolution of the bioactive glass.
  • Reaction mixture is aged in the presence of ammonium hydroxide to catalyze the gelation process to form Bioactive Glass Nanoparticles (BGNPs). Drying is completed to prevent aggregation of the discrete particles and lastly sintering stage is done to remove any organic residuals within the BGNPs.
  • BGNPs Bioactive Glass Nanoparticles
  • TEOS Tetraethyl orthosilicate
  • calcium nitrate after dissolution in ethanol and water.
  • Ammonium dibasic phosphate was dissolved in deionized water.
  • TEOS Tetraethoxysilane
  • Citric Acid Monohydrate (Sigma Aldrich - 120M0186V).
  • Citric Acid Monohydrate 42.028 g was weighed and dissolved in 200 ml of diH 2 O in a beaker and stir at 150 rpm for 5 mins until a uniform solution is formed.
  • BGNPs Fabrication Sol-Gel Processing (Acid/Base Co-catalyzed method):
  • nBG (5wt%) was mixed with polymer solution and pumped through a syringe needle under high voltage.
  • Polymeric fibers were collected on a ground collector and sectioned into flat meshes.
  • bioactivity of the nBG and nBG fibers were evaluated.
  • the materials were immersed in DMEM media and incubated at 37 °C and 5% CO2 to mimic the physiological environment without changing the media throughout the study.
  • the changes in crystallinity were analyzed using XRD. Composition changes were monitored using FTIR. pH levels of the media were recorded.
  • the supernatant of the DMEM were collected at each timepoint and the ion profiles of Si Ca and P were analyzed using inductive coupled plasma spectroscopy.
  • Bioactivity Test 37°C, 5% CO2) ( Figure 8):
  • EDS showed presence of Si, Ca and P ( Figure 10). Quantification of these ion was also conducted using EDS. The theoretical mass % three elements were calculated based on the molar ratio and were compared to the actual mass% calculated from EDS, and showed larger proportion of Si and lower Ca and P.
  • nBG and nBG fibers also maintained pH throughout the 14 days. From stat analysis, PLGA+PCL at day 14 showed significant difference compared to the rest of the groups. Therefore, it is suggested that nBG incorporation into the polymeric fibers may cause a pH buffering effect.
  • Bioactive glass nanoparticles have been used for a wide range of biomedical applications including bone regeneration due to its high specific surface area and reactivity.
  • bioactive glass nanoparticles derived from a SiCL-CaOJ ⁇ CL ternary system may be incorporated into polymer blend (PLGA: PCL) fibers, and this application explores nBG synthesis and evaluates the bioactivity and osteogenic potential of this composite fiber platform.
  • PLGA polymer blend
  • nBG 55%SiO2-40%CaO-5%P2C>5, wt%).
  • nBG 55%SiO2-40%CaO-5%P2C>5, wt%).
  • polymer-nBG fibers 5 wt% of nBG were added to a polymeric blend of PLGA: PCL (5: 1) and electrospun at 10 kV.
  • DMEM 1 mg/ml
  • nBG appeared globular and are 10-20 nm in diameter under SEM.
  • the fibers with or without nBG were 2.21 ⁇ 0.64 pm and 2.24 ⁇ 0.58 pm in diameter, respectively.
  • FTIR analysis reveals a peak at 1100 cm' 1 representing the Si-0 bond of the silica network.
  • pH level remained similar across all experimental groups except for day 14 where particle-free control measured significantly lower pH compared to all other groups, indicative of polymer degradation. Coupling the polymer fiber with nBG served to neutralize the negative impact on pH and allow maintenance of physiological pH.
  • XRD patterns showed crystallization peaks at 31.8° and 45° suggesting crystallization of phosphate and the early formation of carbonated apatite. This was confirmed by FTIR spectra showing growing peaks at 603 and 568 cm' 1 and 960 cm' 1 suggesting dissociation of Si-0 bonds and mineral crystal formation by day 14. These bulk and surface characterization analysis confirm the bioactivity and mineralized in this novel composite system. It also confirms both osteoconductive and osteoinductive potential of these composites for bone regeneration.
  • nBG and nBG fibers supported mineral deposition of bioactivity.
  • Silica release indicates formation of silica gel layer, which is important in mineralization (Li. et. al. 2003). It also indicates CaP release and re -precipitation onto the silica gel layer.
  • nBG releases alkaline products which balances acidic degradation products of PLGA which indicates biocompatibility.
  • nBG showed potential to serve as an apatite nucleation site and nBG fibers showed potential to serve as a structural template for mineral deposition.
  • Antrobus RM Childs HR, Chan MC, Liu J, Brudnicki PA, & Lu HH; Tissue Engineering — Bone Mimics, in Encyclopedia of Bone Biology, Zaidi M ed, Oxford Academic Press, 2020.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Materials Engineering (AREA)
  • Organic Chemistry (AREA)
  • Geochemistry & Mineralogy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Medicinal Chemistry (AREA)
  • Oral & Maxillofacial Surgery (AREA)
  • General Health & Medical Sciences (AREA)
  • Epidemiology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Dermatology (AREA)
  • Transplantation (AREA)
  • Molecular Biology (AREA)
  • Composite Materials (AREA)
  • Inorganic Chemistry (AREA)
  • Dispersion Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Ceramic Engineering (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne une composition d'une plateforme à fibres de verre nano-bioactif (nBG) qui comprend SiO2, CaO, P2O5, PLGA et PLC.
PCT/US2022/051589 2021-12-01 2022-12-01 Microfibres polymères imprégnées de nanoparticules à base de silice pour améliorer le dépôt de minéraux WO2023102150A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163285008P 2021-12-01 2021-12-01
US63/285,008 2021-12-01

Publications (1)

Publication Number Publication Date
WO2023102150A1 true WO2023102150A1 (fr) 2023-06-08

Family

ID=86612981

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/051589 WO2023102150A1 (fr) 2021-12-01 2022-12-01 Microfibres polymères imprégnées de nanoparticules à base de silice pour améliorer le dépôt de minéraux

Country Status (1)

Country Link
WO (1) WO2023102150A1 (fr)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102923957A (zh) * 2012-11-30 2013-02-13 浙江理工大学 一种有序介孔生物活性微晶玻璃的制备方法
CN103100110A (zh) * 2012-11-13 2013-05-15 苏州谷力生物科技有限公司 一种纳米生物活性玻璃及制备工艺
US20140019349A1 (en) * 2008-10-10 2014-01-16 Rent A Toll, Ltd. Method and system for processing vehicular violations
CN107129585A (zh) * 2017-05-02 2017-09-05 佛山今兰生物科技有限公司 一种以微纳米生物活性玻璃作为Pickering乳液稳定粒子制备聚合物微球的方法
CN109437574A (zh) * 2018-10-31 2019-03-08 华南理工大学 一种高钙磷含量的微纳米生物活性玻璃微球及其制备方法
CN111744049A (zh) * 2020-06-12 2020-10-09 广东工贸职业技术学院 一种具有细胞生长调控功能的创面修复材料的制备方法

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140019349A1 (en) * 2008-10-10 2014-01-16 Rent A Toll, Ltd. Method and system for processing vehicular violations
CN103100110A (zh) * 2012-11-13 2013-05-15 苏州谷力生物科技有限公司 一种纳米生物活性玻璃及制备工艺
CN102923957A (zh) * 2012-11-30 2013-02-13 浙江理工大学 一种有序介孔生物活性微晶玻璃的制备方法
CN107129585A (zh) * 2017-05-02 2017-09-05 佛山今兰生物科技有限公司 一种以微纳米生物活性玻璃作为Pickering乳液稳定粒子制备聚合物微球的方法
CN109437574A (zh) * 2018-10-31 2019-03-08 华南理工大学 一种高钙磷含量的微纳米生物活性玻璃微球及其制备方法
CN111744049A (zh) * 2020-06-12 2020-10-09 广东工贸职业技术学院 一种具有细胞生长调控功能的创面修复材料的制备方法

Similar Documents

Publication Publication Date Title
Gómez-Cerezo et al. Mesoporous bioactive glass/ɛ-polycaprolactone scaffolds promote bone regeneration in osteoporotic sheep
Jiménez et al. Bibliographic review on the state of the art of strontium and zinc based regenerative therapies. Recent developments and clinical applications
US8282911B2 (en) Implant material
Amudha et al. Enhanced mechanical and biocompatible properties of strontium ions doped mesoporous bioactive glass
Cui et al. A novel injectable borate bioactive glass cement for local delivery of vancomycin to cure osteomyelitis and regenerate bone
Balasubramanian et al. Biomedical applications of ceramic nanomaterials: a review
CN107149698B (zh) 生物相容的材料及其用途
Jalise et al. The effects of strontium incorporation on a novel gelatin/bioactive glass bone graft: In vitro and in vivo characterization
Vafa et al. A better roadmap for designing novel bioactive glasses: effective approaches for the development of innovative revolutionary bioglasses for future biomedical applications
Xu et al. Noninvasive monitoring of bone regeneration using NaYF4: Yb3+, Er3+ upconversion hollow microtubes supporting PLGA-PEG-PLGA hydrogel
Ma et al. Recent progress on fabrication of calcium-based inorganic biodegradable nanomaterials
Durgalakshmi et al. Nano-bioglass: a versatile antidote for bone tissue engineering problems
Zhao et al. The effect of poly (lactic-co-glycolic acid)(PLGA) coating on the mechanical, biodegradable, bioactive properties and drug release of porous calcium silicate scaffolds
CN111840652B (zh) 骨修复材料及其制备方法
Xu et al. Calcium phosphate ceramics and synergistic bioactive agents for osteogenesis in implant dentistry
Zhu et al. Advances in osseointegration of biomimetic mineralized collagen and inorganic metal elements of natural bone for bone repair
Zhu et al. Cemented injectable multi-phased porous bone grafts for the treatment of femoral head necrosis
WO2023102150A1 (fr) Microfibres polymères imprégnées de nanoparticules à base de silice pour améliorer le dépôt de minéraux
ES2378341T3 (es) Cemento óseo compuesto
Sha et al. Physico-chemical and biological properties of novel Eu-doped carbonization modified tricalcium silicate composite bone cement
Oltean-Dan et al. BIOCOMPATIBILITY OF TITANIUM IMPLANTS COATED WITH BIOCOMPOSITE IN A RAT MODEL OF FEMORAL FRACTURE.
Sezer et al. In vivo performance of poly (ε-caprolactone) constructs loaded with gentamicin releasing composite microspheres for use in bone regeneration
Garcés-Villalá et al. Evaluation of two highly porous microcrystalline biphasic calcium phosphate-based bone grafts for bone regeneration: an experimental study in rabbits
Qi et al. Advances in magnesium-containing bioceramics for bone repair
Ito et al. Magnesium-and zinc-substituted beta-tricalcium phosphates as potential bone substitute biomaterials

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22902204

Country of ref document: EP

Kind code of ref document: A1