GB2532283A - Morphogenetically active calcium polyphosphate nanoparticles - Google Patents

Morphogenetically active calcium polyphosphate nanoparticles Download PDF

Info

Publication number
GB2532283A
GB2532283A GB1420363.2A GB201420363A GB2532283A GB 2532283 A GB2532283 A GB 2532283A GB 201420363 A GB201420363 A GB 201420363A GB 2532283 A GB2532283 A GB 2532283A
Authority
GB
United Kingdom
Prior art keywords
polyphosphate
calcium
polyp
nanoparticles
solution
Prior art date
Legal status (The legal status 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 status listed.)
Withdrawn
Application number
GB1420363.2A
Other versions
GB201420363D0 (en
Inventor
Ernst Ludwig Georg Muller Werner
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
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 Individual filed Critical Individual
Priority to GB1420363.2A priority Critical patent/GB2532283A/en
Publication of GB201420363D0 publication Critical patent/GB201420363D0/en
Priority to PCT/EP2015/076222 priority patent/WO2016078971A1/en
Priority to CN201580067850.6A priority patent/CN106999624B/en
Priority to EP15794143.6A priority patent/EP3220967B8/en
Priority to CN201580069333.2A priority patent/CN106999630A/en
Priority to PCT/EP2015/076172 priority patent/WO2016078963A2/en
Priority to US15/527,479 priority patent/US10307350B2/en
Priority to US15/527,520 priority patent/US20190083679A1/en
Priority to EP15794139.4A priority patent/EP3220966A2/en
Priority to PCT/EP2015/076468 priority patent/WO2016079006A1/en
Priority to CN201580072176.0A priority patent/CN107106729A/en
Priority to EP15794552.8A priority patent/EP3220968A1/en
Priority to US15/527,553 priority patent/US20170319740A1/en
Publication of GB2532283A publication Critical patent/GB2532283A/en
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/02Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K6/00Preparations for dentistry
    • A61K6/15Compositions characterised by their physical properties
    • A61K6/17Particle size
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K6/00Preparations for dentistry
    • A61K6/80Preparations for artificial teeth, for filling teeth or for capping teeth
    • A61K6/831Preparations for artificial teeth, for filling teeth or for capping teeth comprising non-metallic elements or compounds thereof, e.g. carbon
    • A61K6/838Phosphorus compounds, e.g. apatite
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K6/00Preparations for dentistry
    • A61K6/80Preparations for artificial teeth, for filling teeth or for capping teeth
    • A61K6/849Preparations for artificial teeth, for filling teeth or for capping teeth comprising inorganic cements
    • A61K6/864Phosphate cements
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/5115Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5192Processes
    • 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
    • A61L24/00Surgical adhesives or cements; Adhesives for colostomy devices
    • A61L24/02Surgical adhesives or cements; Adhesives for colostomy devices containing inorganic materials
    • 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
    • A61L24/00Surgical adhesives or cements; Adhesives for colostomy devices
    • A61L24/04Surgical adhesives or cements; Adhesives for colostomy devices containing macromolecular materials
    • 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
    • A61L27/02Inorganic materials
    • A61L27/12Phosphorus-containing materials, e.g. apatite
    • 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
    • C01INORGANIC CHEMISTRY
    • C01BNON-METALLIC ELEMENTS; COMPOUNDS THEREOF; METALLOIDS OR COMPOUNDS THEREOF NOT COVERED BY SUBCLASS C01C
    • C01B25/00Phosphorus; Compounds thereof
    • C01B25/16Oxyacids of phosphorus; Salts thereof
    • C01B25/26Phosphates
    • C01B25/38Condensed phosphates
    • C01B25/40Polyphosphates
    • 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

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Oral & Maxillofacial Surgery (AREA)
  • Inorganic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Transplantation (AREA)
  • Dermatology (AREA)
  • Organic Chemistry (AREA)
  • Optics & Photonics (AREA)
  • Physics & Mathematics (AREA)
  • Biomedical Technology (AREA)
  • Nanotechnology (AREA)
  • Plastic & Reconstructive Surgery (AREA)
  • Surgery (AREA)
  • Dispersion Chemistry (AREA)
  • Materials For Medical Uses (AREA)

Abstract

A method for the production of a solid, degradable polyphosphate material having calcium counterions comprises: (i) dissolving a polyphosphate salt in an aqueous solvent, such as water, and adjusting the pH value of the solution to alkaline values; (ii) adding a solution of a calcium salt solution to the polyphosphate salt solution, and adjusting the pH value to alkaline values, (iii) optionally, washing with a solvent, for example ethanol, and (iv) collecting the particles formed. The method in steps (i) to (iii) is performed at ambient temperature, and the material has a hardness similar to that of bone tissue, and is morphogenetically active. Preferably, the polyphosphate is sodium phosphate, and has a chain length in the range of 3 to 1000 phosphate units. The calcium salt solution may be calcium chloride, and the pH may be adjusted to 10. The amorphous nanoparticles made by the method may be used in bone regeneration and repair and in dentistry, or for drug delivery.

Description

MORPHOGENETICALLY ACTIVE CALCIUM POLYPHOSPHATE
NANOPARTICLES
This invention concerns a calcium polyphosphate material consisting of amorphous nanoparticles with a diameter of approximately 0.2 pm that displays a considerable hardness (elastic modulus) of about 1.3 GPa. The inventive non-crystalline and biodegradable material that is fabricated under mild conditions, at room temperature, is morphogenetically active and induces bone formation and the expression of the marker gene for osteoblast activity, alkaline phosphatase.
Background of Invention
Inorganic polyphosphate (polyP) is a nontoxic polymer existing in a wide range of organisms (Schroder HC, Muller WEG, eds (1999) Inorganic Polyphosphates -Biochemistry, Biology, Biotechnology. Prog Mol Subcell Biol 23:45-81; Kulaev IS, Vagabov V, Kulakovskaya T (2004) The Biochemistry of Inorganic Polyphosphates. New York: John Wiley & Sons Inc). It consists of usually linear molecules of tens to hundreds of phosphate units which are linked together via high energy phosphoanhydride bonds. Living organisms can produce this polymer metabolically at ambient temperatures, while the chemical synthesis of polyP requires high temperatures of several hundred degrees.
PolyP in bone Previous studies revealed that polyP molecules of different chain lengths accumulate especially in bone cells (Leyhausen G, Lorenz B, Zhu H, Geurtsen W, Bohnensack R, Muller WEG, Schroder HC. Inorganic polyphosphate in human osteoblast-like cells. J Bone Mineral Res 1998;13:803-812; Schroder HC, Kurz L, Muller WEG, Lorenz B. Polyphosphate in bone. Biochemistry (Moscow) 2000;65:296-303). In addition, human osteoblast-like cells contain enzymes that hydrolyze polyP, e.g. the alkaline phosphatase (ALP) (Lorenz B, Schroder HC. Mammalian intestinal alkaline phosphatase acts as highly active exopolyphosphatase. Biochim Biophys Acta 2001;1547:254-261). PolyP is also found in platelets (Smith SA, Mutch NJ, Baskar D, Rohloff P, Docampo R, Morrissey TH. Polyphosphate modulates blood coagulation and fibrinolysis. Proc Natl Acad Sci USA 2006;103:903-908), possibly playing a role in initiation of healing of bone fractures.
PolyP has an inductive effect on osteoblasts mainly as an anabolic polymer that stimulates differentiation of bone cells and mineralization (reviewed in: Wang XH, Schroder HC, Wiens M, Ushijima H, Muller WEG. Bio-silica and bio-polyphosphate: applications in biomedicine (bone formation). Current Opinion Biotechnol 2012;23:570-578; Wang XH, Schroder HC and Muller WEG. Enzymatically synthesized inorganic polymers as morphogenetically active bone scaffolds: application in regenerative medicine. Int Rev Cell Mol Biol 2014;313:27-77). In addition, polyP induces the ALP (Muller WEG, Wang XH, Diehl-Seifert B, Kropf K, Schloamacher U, Lieberwirth I, Glasser G, Wiens M and SchrOder HC. Inorganic polymeric phosphate/polyphosphate as an inducer of alkaline phosphatase and a modulator of intracellular Can level in osteoblasts (SaOS-2 cells) in vitro. Acta Biomater 2011;7:266h 2671).
Bone graft materials Bone graft materials must be biocompatible and comprise similar biomechanical properties like the physiological bone tissue. Favored are substitutes that are degradable and allow the surrounding tissue to migrate into at least the surface region of the non-self implant to strengthen the bridging of the biofabricated material to the surrounding tissue and to avoid inflammatory reactions.
Besides of cell-based bone graft substitutes ceramic-based materials have proven to be useful bone scaffold for implants. Among the calcium phosphate-based ceramics, the following materials are of particular relevance: Hydroxvapatite (HA). HA is produced at a high-temperature reaction and comprises a crystalline form of calcium phosphate (Noshi T, Yoshikawa T, Ikeuchi NI, Dohi Y, Ohgushi H, Horiuchi K, Sugimura M, Ichijima K, Yonemasu K. Enhancement of the in vivo osteogenic potential of marrow/hydroxyapatite composites by bovine bone morphogenetic protein. J Biomed Mater Res 2000;52:621-630). Since the composition of HA is Calo(PO4)6(OH)2 with a calcium-to-phosphate ratio of 1.67 this material is chemically very similar to the mineralized phase of physiological bone. Based on this property HA shows suitable biocompatibility (Nandi SK, Kundu B, Ghosh SK, De DK, Basu D. Efficacy of nanohydroxyapatite prepared by an aqueous solution combustion technique in healing bone defects of goat. J Vet Sci 2008;9:183-191).
13-Tricalcium phosphate ((3-TCP). Similar as HA, the chemical composition Ca3(PO4)2 and crystallinity of 13-TCP match the ones of the mineral phase of bone; in addition, this material is bioabsorbable and biocompatible (Daculsi G, LeGeros RZ, Heughebaert M, Barbieux I. Formation of carbonate apatite crystals after implantation of calcium phosphate ceramics. Calcif Tissue Int 1990;46:20-27). TCP implants have been successfully used as synthetic bone void fillers both in orthopedics and in dentistry (Shigaku S, Katsuyuki F. Beta-tricalcium phosphate as a bone graft substitute. Jikeikai Med J 2005;52:47-54).
Bioactive glass. Finally, bioactive glass ceramics ("bioglass") initially developed by Hench et al (Hench LL, Splinter RI, Allen WC, Greenlee TK. Bonding mechanisms at the interface of ceramic prosthetic materials. J Biomed Mater Res Symp 1971;2:117-141; reviewed in: Chen Q, Roether JA, Boccaccini AR. Tissue engineering scaffolds from bioactive glass and composite materials. In: Ashammakhi N, Reis R, Chiellini F Topics in Tissue Engineering, Vol 4, pp. 1-27, 2008) is not only biocompatible but also osteoconductive and allows bone to bind without any intervening fibrous connective tissue interface (Zhang H, Ye XJ, Li IS. Preparation and biocompatibility evaluation of apatite/ wollastonite-derived porous bioactive glass ceramic scaffolds. Biomed Mater 2009;4: 45007). This mineralic glass is frequently used as filling material for bone defects either alone or in combination with autogeneic or allogenic cancellous bone graft (Dorea HC, McLaughlin RM, Cantwell HD, Read R, Armbrust L, Pool R, Roush JK, Boyle C. Evaluation of healing in feline femoral defects tilled with cancellous autograft, cancellous allograft or Bioglass. Vet Comp Orthop Traumatol 2005;18:157-168) or with morphogenetically active inorganic polymers, e.g. polyphosphate [polyP] (Wang XH, Tolba E, Schroder HC, Neufurth NI, Feng Q, Diehl-Seifert B, Muller WEG. Effect of bioglass on growth and biomineralization of SaOS-2 cells in hydrogel after 3D cell bioprinting. PLoS ONE 2014;9:e112497).
Since all of these mineralic implant materials are fabricated at temperatures higher than 700°C and, in turn, are not, or only at a limited degree, osteoinductive like the bioglass, the inventors developed a new potential biomaterial likewise suitable to be used as bone implant.
The new material according to this invention is based on polyP. Previously polyP has been used, after calcinations, as potential scaffold for bone implants (Pilliar RM, Filiaggi MJ, Wells JD, Grynpas MD, Kandel RA. Porous calcium polyphosphate scaffolds for bone substitute applications -in vitro characterization. Biomaterials 2001;22:963-972; Qiu K, Wan CX, Zhao CS, Chen X, Tang CW, Chen YW. Fabrication and characterization of porous calcium polyphosphate scaffolds. J Materials Sci 2006;41:2429-2434; Ding YL, Chen YW, Qin YJ, Shi GQ, Yu XX, Wan CX. Effect of polymerization degree of calcium polyphosphate on its microstructure and in vitro degradation performance. J Mater Sci Mater Med 2008;19:1291-1295; Wang D, Wallace AF, De Yoreo JJ, Dove PM. Carboxylated molecules regulate magnesium content of amorphous calcium carbonates during calcification. Proc Natl Acad Sci USA 2009;106:21511-21516). In all those preparations, polyP had to be subjected to calcination with the result that the polyP chain might be degraded and the exact chain length is impossible to be determined. Furthermore those polyP scaffolds have not been described to act in an osteoinductive manner.
Summary of the invention
The material according to this invention is a hard amorphous polyP-based biomaterial that is produced at ambient conditions (i.e. at around 20°C ± 10°C) in the presence of a distinctly adjusted concentration of CaC12. The material obtained comprises a porous scaffold built of spherical, amorphous nanoparticles that are biodegradable and retain the morphogenetic activity of the inorganic polymer.
Thus, in a first aspect thereof, this invention concerns a new morphogenetically active material consisting of calcium polyphosphate (polyP) nanoparticles that are (7) amorphous and (ii) display an unusual hardness not found for calcium polyphosphate materials prepared by state-of-the-art methods. The inventors developed a controlled and slow fabrication process that is performed at room temperature and unexpectedly resulted in the formation of a material showing these properties. The polyP material formed in the presence of CaCl2 at a stoichiometric ratio of around 1 or 2 (phosphate to calcium) is an amorphous powder that is composed of nanospheres with a diameter of approximately 0.2 p.m. The inventive material is degradable, in contrast to the Ca-polyP salt prepared by conventional methods which resists hydrolytic cleavage by phosphatases present in medium over longer time periods in cell culture experiments.
The following patent applications on polyP are deemed relevant; GB1406840.7. Morphogenetically active hydrogel for bioprinting of bioartificial tissue. Inventors: Milner WEG, Schroder HC, Wang XH; GB1403899.6. Synergistic composition comprising quercetin and polyphosphate for treatment of bone disorders. Inventors: Muller WEG, Schroder HC, Wang XH.
Detailed description of the invention
The inventors describe a novel polyphosphate (polyP) material that is characterized by the following properties: a) The material is amorphous (non-crystalline); b) The material has an unusual hardness (e.g., the elastic modulus of the Ca-polyP2 biopolymer according to this invention amounts to approximately 1.3 GPa (in the context of the present invention, "hard" or "hardness" means a value of approximately 1.3 GPa, such as between 0.8 and 1.8 GPa, preferably between 1.0 and 1.6 GPa), close to value measured for trabecular tissue surrounding human bone [6.9 GPa]); and preferably, c) The material consist of nanoparticles with a diameter of about 0.2 um (Ca-polyP2 particles according to this invention). -4 -
Advantageously, the inventive material can be prepared under mild conditions, such as ambient conditions, and particularly at room temperature.
Hardness can be measured using the Brinell and/or Vickers hardness test method, as known to the person of skill.
It was surprisingly found, that the inventive material is morphogenetically active, it induces bone alkaline phosphatase activity and new bone formation (hydroxyapatite synthesis); and that the material is biodegradable (e.g. by polyphosphatases, such as bone alkaline phosphatase).
This inventive material, whose properties make it superior compared to conventional polyphosphate preparations for a use, for example, in bone regeneration and replacement materials (e.g. GB1406840.7. Morphogenetically active hydrogel for bioprinting of bioartificial tissue [Inventors: Muller WEG, Schroder MC, Wang XH]; GB1403899.6. Synergistic composition comprising quercetin and polyphosphate for treatment of bone disorders [Inventors: Muller WEG, Schroder HC, Wang XH]), can be prepared according to the following method, according to this invention: a) Dissolution of a polyP salt in water and adjustment of the pH to alkaline values, b) (slow) Addition of a solution of a calcium salt to the polyP salt (with Na+) solution, and adjustment of the pH to alkaline value, and c) Collection and drying of the particles thus formed, optionally after washing with ethanol.
This procedure is performed at room temperature.
As a preferred example, the method can be carried out using sodium polyphosphate (NapolyP) as a polyP salt (chain length: 50 phosphate units) in solution and calcium chloride as a calcium salt in solution as follows.
a) Dissolution of 10 g of Na-polyP in 500 ml of distilled water and adjustment of the pH to 10 with 1 M NaOH (room temperature), b) Slow, dropwise (1 ml/min), addition of a solution of 14 g of calcium chloride (Ca-polyP1) or 28 g of calcium chloride (Ca-polyP2) in 250 ml salt to the Na-polyP solution and adjustment of the pH to 10 (room temperature); c) Stirring of the thus formed suspension for about 4 h, and d) Collection of the particles formed, and optionally washing them with ethanol while filtering (0.45 pm filter; e.g., Nalgene Rapid-Flow), and drying at 60°C.
The Ca-polyP material obtained with 14 g of CaCl2 was named Ca-polyPl (stoichiometric ratio phosphate: calcium of 1); and the Ca-polyP material obtained with 28 g of CaCI1 was named "Ca-polyP2" (stoichiometric ratio phosphate: calcium of 1:2).
The nanoparticles consisting of the inventive Ca-polyP material are characterized by a high hardness (about 1.3 GPa; Ca-polyP2) compared to larger particles that are produced at a different phosphate to calcium ratio in solution or at harsh reaction conditions, e.g. in acid-flux of phosphoric acid with Ca(OH)) at high (250°C) temperature (Jackson LE, Kariuki BM, Smith ME, Barralet JE, Wright AJ. Synthesis and structure of a calcium polyphosphate with a unique criss-cross arrangement of helical phosphate chains. Chem Mater 2005;17:4642-4646), or the described calcium-polyphosphate complex (Muller WEG, Wang XH, Diehl-Seifert B, Kropf K, Schlo3macher U, Lieberwirth I, Glasser G, Wiens NI, Schroder HC. Inorganic -5 -polymeric phosphate/polyphosphate is an inducer of alkaline phosphatase and a modulator of intracellular Ca2+ level in osteoblasts (SaOS-2 cells) in vitro. Acta Biomater 2011;7:26612671).
The new, hard polyP material according to this invention is biodegradable and displays superior morphogenetic activity, compared to the Ca-polyP salts prepared by conventional techniques.
In addition, the inventive hard Ca-polyP nanoparticles are prone to cellular uptake (even observed for larger, 600 nm particles; Zhao Y, Sun X, Zhang G, Trewyn BG, Slowing II, Lin VS. Interaction of mesoporous silica nanoparticles with human red blood cell membranes: size and surface effects. ACS Nano 2011;5:1366-1375) and subsequent metabolization, a property that is not possible for the free polyanionic polyP polymer.
The chain lengths of the polyP molecules can be in the range 3 to up to 1000 phosphate units. Optimal results are achieved with polyP molecules with an average chain length of approximately 200 to 20, and optimally about 50 phosphate units.
A further aspect of the invention concerns the material as obtained by one of the methods described above.
Further, the inventors demonstrate that the inventive method can be used for the preparation of hard amorphous and morphogenetically active polyP nanoparticles.
A further aspect of the invention concerns a material comprising the nanoparticles as obtained by one of the methods described above.
The technology according to this invention can be used for the fabrication of nanoparticles or for a material containing such nanoparticles to be used, preferably, in bone regeneration and repair and in dentistry.
A further aspect of the invention concerns the application of the inventive nanoparticles in drug delivery, again, preferably, in bone regeneration and repair and in dentistry, in analogy to, for example, systems described in Kwon et al. and Yang et al. (Kwon S, Singh RK, Perez RA, Abou Neel EA, Kim EIW, Chrzanowski W. Silica-based mesoporous nanoparticles for controlled drug delivery. J Tissue Eng. 2013 Sep 3; 4:2041731413503357. eCollection 2013. Yang P, Gai S, Lin J. Functionalized mesoporous silica materials for controlled drug delivery. Chem Soc Rev. 2012 May 7;41(9):3679-98).
The invention will now be described further in the following preferred examples, nevertheless, without being limited thereto. For the purposes of the present invention, all references as cited herein are incorporated by reference in their entireties. In the Figures listing, Figure 1 shows the FTIR spectra of Na-polyP, Ca-polyP1 and Ca-polyP2; (A) wavenumbers 4000 to 600 cm-1 and (B) wavenumbers 1400 and 600 cm-1 Figure 2 shows the SEM micrographs taken from the following polyP powders: (A and B) Na-polyP, (C and D) Ca-polyP1 and (E and F) Ca-polyP2. -6 -
Figure 3 shows the biological properties of the Ca-polyP material, developed here. (A) SaOS2 cells remained without phosphate material (none), or were incubated with 10 ttg/mL of NapolyP, Ca-polyP1, Ca-polyP2, HA, or 0-TCP for 7 d in the presence of the activation cocktail OC. Then the coverslips with the cells were stained with Alizarin Red S. (B) Expression level of ALP in SaOS-2 cells, in response to exposure to phosphate material. After an 1 or 7 d incubation period the cells were harvested and RNA was extracted and subjected to qRT-PCR analysis. The steady-state-levels of the ALI' transcripts were measured and normalized to the expression of GA1'011. Data are expressed as mean values ± SD for four independent experiments; each experiment was carried out in duplicate. Differences between the groups were evaluated using unpaired f-test. *p < 0.05. (C) Degradation of polyP in vitro, using the 16.5% polycrylamide/7 NI urea gel electrophoresis technique. The cells were incubated with 50 ttg/m1 of solid Ca-polyP2 either in the presence of SaOS-2 cells and medium/serum or PBS [phosphate buffered saline]; after an incubation period of 1 or 7 d, samples were taken for chain length determination. Synthetic polyP markers with an average chain length of 80, 40 and 3 units were run in parallel.
Examples
In the following examples, the inventive method described only for polyP molecules with a chain length of 50 phosphate units. Similar results can be obtained by using polyP molecules with lower and higher chain lengths, such as between 100 to 20 units.
FTIR analyses The two phosphate materials, Ca-polyP1 and Ca-polyP2, were characterized by FTIR and compared with the spectrum obtained with Na-polyP (Fig. 1). The complete spectra between the wavenumbers 4000 and 600 cm1 are shown in Fig. 1A, while segments between 1400 and 600 cm-I are given in Fig. 1B. The band near 1250 cm-I is assigned to the asymmetric stretching mode of the two non-bridging oxygen atoms bonded to phosphorus atoms in the P02 metaphosphate units, vas(P02)-. The weak band at 1190 cm-1 is the P02 symmetric stretching mode vs(P02)-.The absorption bands close to 1083 and 999 cm-1 are assigned to the asymmetric and symmetric stretching modes of chain-terminating P03 groups [vas(P03)2 and vs(P03)21. The absorption band near 864 cm-I is attributed to the asymmetric stretching modes of the P-O-P linkages, vas (P-O-P) and the partially split band centered around 763 cm-1 is due to the symmetric stretching modes of these linkages, vs (P-O-P).
The comparison of the spectra of Na-polyP and Ca-polyP shows that the polyP features are seen in the 1300-1000 cm-1 region. There, most of the chemical characteristics of polyP chains are found. In all three samples a similar pattern is seen, reflecting that the polyP chain backbones are not broken down during the reaction with the Ca2+ ions. However, the peaks are shifted in the Ca-polyP1 and Ca-polyP2 samples if compared the Na-polyP spectrum. This shifting is also seen for other bands in the Ca-polyP spectra; they even increase by increasing the Cat-content in the polyP sample (from Ca-polyP1 to Ca-polyP2). Moreover, it has been reported that the adsorptions bands near 1100-1000 cm-1 are attributed to the ionic stretching mode of the P-0-group, the shifting as well as broadening of this peak of the Ca-polyP samples is attributed to the formation of P-O-N1-(±) (where NI is Ca2-). In conclusion, the 1R spectra confirm the interaction between Ca2+ and polyP and the formation of Ca-PolyP.
XRD analyses were performed with both Ca-polyP1 and Ca-polyP2; the patterns showed no sign of crystallinity, like Na-polyP (Fig. 1C). -7 -
Morphology of polyP samples The three samples, Na-polyP, Ca-polyP1 and Ca-polyP2, were analyzed by SEM (Fig. 2). The Na-polyP particles, of a non-regular shapes, often show a tapered morphology (Fig. 2A and B). The sizes of the particles vary between 1 and 300 um with an average size of =---1 00 pm. Likewise non-regular shapes show the Ca-polyPl particles (Fig. 2C and D). They are smaller than the Na-polyP particles with an average diameter of =4 um. Even smaller are the CapolyP2 particles with an average diameter of =02 pm (Fig. 2 E and F).
PolyP-induced mineralization of SaOS-2 cells The cells were incubated with 10 ug/mL of Na-polyP, Ca-polyP1, Ca-polyP2, HA, or f3-TCP in the presence of the activation cocktail OC for 7 d. Then the coverslips onto which the cells had been cultivated were removed and stained with Alizarin Red S, as described under Methods. Eye-inspection revealed that the intensity of the color reaction, which reflects the extent of minerals being present in the samples, is highest for Ca-polyP1 and for Ca-polyP2. The degree of color reaction is lower for Na-polyP, 13-TCP and HA; the intensities of those samples are only slightly higher, compared to the control (Fig. 3A).
Expression level of ALP Since the Alizarin Red S color reaction might not be sensitive enough due to cross-reactivity with exogenously added grains, the steady-state-expression level of ALP in SaOS-2 cells was quantified by qRT-PCR. The inventors determined previously that the expression level of this enzyme is a reliable marker for the polyP-induced activation of bone cells (Muller WEG, Wang XH, Diehl-Seifert B, Kropf K, Schlol3macher U, Lieberwirth I, Glasser G, Wiens M and Schroder HC. Inorganic polymeric phosphate/polyphosphate as an inducer of alkaline phosphatase and a modulator of intracellular Ca--level in osteoblasts (SaOS-2 cells) in vitro. Acta Biomaterialia 2011j;7:2661-2671). Therefore, the inventors subjected the cells, after incubation with the different phosphate samples and in the presence of the OC activation cocktail to qRT-PCR analysis. The data revealed that at day 1 the expression level of ALP is statistically not different between the different polyphosphate samples used. However, after an incubation period of 7 d the steady-state-expression levels of ALP in the cells exposed to Ca-polyP1 and Ca-polyP2 are significantly higher 0.10 expression units compared to the one of the reference GAPDH) than those measured for Na-polyP, fl-TCP or HA (cc. 0.055 expression units). The expression levels of the latter three assays are not significantly higher than the one seen in control assays (no phosphate sample added); Fig. 3B.
Hardness of the Ca-polyP2 material Determination of the mechanical properties (elastic modulus) of the Ca-polyP2 biopolymer was performed with a ferrule-top cantilever and found to be of 1.3 GPa, close to values measured for trabecular tissue that is surrounding human bone with 6.9 GPa (Zysset PK, Guo XE, Hoffler CE, Moore KE, Goldstein SA. Elastic modulus and hardness of cortical and trabecular bone lamellae measured by nanoindentation in the human femur. J Biomech 1999; 321005-1012).
Degradation of polyP in vitro In one series of experiments, the SaOS-2 cells were incubated with 50 pg/m1 of solid CapolyP2 and incubated in the standard assay for 1 d or 7 d either in the presence of SaOS-2 cells and medium/serum or in PBS. Then samples were taken and assayed for the chain length of polyP (Lorenz B, Schroder I-1C. Mammalian intestinal alkaline phosphatase acts as highly active exopolyphosphatase. Biochim Biophys Acta 2001;1547:254-261). The gels were stained with o-toluidine blue. The results (Fig. 3C) show that Ca-polyP2, added to the assays, does not undergo hydrolytic degradation during the 7 d long incubation period if dissolved in -8 -PBS. In contrast the average chain length of Ca-polyP2 drops from an average chain length of 40 to around 3 Pi units if the polymer is incubated in the assays which contained SaOS-2 cells and medium/serum. This result indicates that Ca-polyP2 is prone to hydrolysis by ALP (exopolyphosphatase), but also to other polyP hydrolyzing enzymes, endophosphatases, that exist in serum.
Methods Polyphosphate The sodium polyphosphate (Na-polyP of an average chain of 40 phosphate units) used in the Examples has been obtained from Chemische Fabrik Budenheim (Budenheim; Germany).
Preparation of calcium polyphosphate nanospheres Ten g of Na-polyP are dissolved in 500 ml of distilled water and the pH is adjusted to 10 with 1 M NaOH. A solution of 14 g of calcium chloride dihydrate or 28 g of CaC12 in 250 ml is added slowly, dropwise (1 ml/min) to the Na-polyP solution, adjusting steadily the pH to 10 at room temperature. The suspension formed is stirred for 4 hr. Then the particles formed are collected by washing twice with ethanol while filtering through a 0.45 p.m filter (e.g., Nalgene Rapid-Flow). Then the particles are dried at 60°C. The Ca-polyP material obtained by addition of 14 g of CaC12 is named "Ca-polyP1", and the one with 28 g of CaC12 is termed "Ca-polyP2".
Chemical characterization by FTIR Fourier transformed infrared (FTIR) spectroscopy in the attenuated total reflectance (ATR) mode is applied, using, for example, the Varian 660-IR spectrometer with Golden Gate ATR auxiliary (Agilent). Spectra between the wavenumbers 4000 and 600 cm-1 are recorded.
Scanning electron microscopy For the scanning electron microscopic (SEM) analyses, for example, a HITACHI SU 8000 can be employed at low voltage (<1 kV; analysis of near-surface organic surfaces).
XRD analyses X-ray diffraction (XRD) is performed using established procedures (Fischer V, Lieberwirth I, Jakob G, Landfester K, Mulioz-Espi R. Metal oxide/polymer hybrid nanoparticles with versatile functionality prepared by controlled surface crystallization. Adv Funct Mat 2013;23:451-466).
Cells and cell culture conditions Human osteogenic sarcoma cells, SaOS-2 cells, are used for the experiments and cultivated in McCoy's medium with fetal calf serum [FCS] (Wiens M, Wang XH, Schlof3macher U, Lieberwirth I, Glasser G, Ushijima H, Schroder HC, Muller WEG. Osteogenic potential of bio-silica on human osteoblast-like (SaOS-2) cells. Calcif Tissue Intern 2010;87:513-524). Cultivation of the cells is performed in 24-well plates; 3X104 cells are seeded per well. After an initial incubation period of 3 d, the cultures are supplemented either with 10 mg/mL of solid Na-polyP, supplemented with CaC12 in a 2:1 stoichiometric ratio, in order to compensate for the chelating activity of polyP as described (Muller WEG, Wang XH, Diehl-Seifert B, Kropf K, SchloBmacher U, Lieberwirth I, Glasser G, Wiens M, Schroder HC. Inorganic polymeric phosphate/polyphosphate as an inducer of alkaline phosphatase and a modulator of intracellular Cat-1 level in osteoblasts (SaOS-2 cells) in vitro. Acta Biomaterialia 2011; 7;2661-26719), or with the two polyP samples, prepared here, with Ca-polyP1 or with CapolyP2. In comparison also nano-hydroxyapatite (HA; for example: 677418 Sigma-Aldrich) or p-TCP (for example: 13204 Sigma-Aldrich) had been included in the test series. As controls none of those polymers is added. Then the cells are continued to be incubated in the presence of the osteogenic cocktail [OC], containing 10 nM dexamethasone, 5 mM 13-glycerophosphate and 50 mM ascorbic acid.
After a 7 d incubation period the cells are either stained with Alizarin Red S to assess the extent of mineralization (Schroder HC, Borejko A, Krasko A, Reiber A, Schwertner H, Muller WEG. Mineralization of SaOS-2 cells on enzymatically (silicatein) modified bioactive osteoblast-stimulating surfaces. J Biomed Mater Res B Appl Biomater 2005; 75:387-392) or subjected to qRT-PCR [quantitative real-time RT-PCR] analysis (Wiens M, Wang XH, Schloemacher U, Lieberwirth I, Glasser G. Ushijima H, Schroder HC, Muller WEG. Osteogenic potential of bio-silica on human osteoblast-like (SaOS-2) cells. Calcif Tissue Intern 2010;87:513-524).
Transcripts expression For qRT-PCR reactions the primer pair Fwd: 5'-TGCAGTACGAGCTGAACAGGAACA-3' (SEQ ID NO: 1) [ntii4i to nti ii,4] and Rev: 5'-TCCACCAAATGTGAAGACGTGGGA-3' (SEQ ID NO: 2) [nti418 to nti395; PCR product length 278 bp] can be used for the quantification of the alkaline phosphatase [ALP] transcripts (accession number NM 000478.4). The expression level of the ALI' can be normalized to the one of the reference gene GAPIEI (glyceralclehyde 3-phosphate dehydr*ogenase; NM 002046.3) using the primer pair Fwd: 5'-CCGTCTAGAAAAACCTGCC-3' (SEQ ID NO: 3) [ntios to ntii63] and Rev: 5'-GCCAAATTCGTTGTCATACC-3' (SEQ ID NO: 4) [ntio5u to ntinm; 215 bp].
Determination of the hardness The hardness of the polyphosphate material Ca-polyP2 can be determined, for example, by a ferruled optical fiber-based nanoindenter as described (Chavan D, Andres D, Iannuzzi D. Ferrule-top atomic force microscope. II. Imaging in tapping mode and at low temperature. Rev Sci Instrum 2011; 82:046107. doi: 10.1063/1.3579496).
PolyP degradation in vitro In one series of experiments the SaOS-2 cells are incubated with 50 pg/ml of solid Ca-polyP or Ca-polyP2 and incubated in the standard assay for 4 d. Then samples (50 pi) are taken and assayed for the chain length of polyP (for example, see: Lorenz B, Schroder HC. Mammalian intestinal alkaline phosphatase acts as highly active exopolyphosphatase. Biochim Biophys Acta 2001;1547:254-261). The gels are stained with o-toluidine blue.
Statistical analysis The results can be statistically evaluated using the paired Student's t-test.

Claims (10)

  1. -10 -CLAIMS1. Method for the production of a solid, degradable amorphous polyphosphate material having calcium counterions, comprising the steps of i) dissolving of a polyphosphate salt in an aqueous solvent, such as water, and adjusting the pH value of the solution to alkaline values, ii) adding a solution of a calcium salt solution to said polyphosphate salt solution, and adjusting the pH value to alkaline values, iii) optionally, washing with a solvent, for example ethanol, and iv) collecting of the particles formed, wherein said method in steps i) to iii) is performed at ambient temperature, and wherein said material hasa hardness similar to that of bone tissue, and is morphogenetically active.
  2. 2. The method according to claim 1, wherein the said polyphosphate salt is sodium polyphosphate.
  3. 3. The method according to claim 1 or 2, wherein the chain length of the polyphosphate is in the range of about 3 to about 1000 phosphate units, preferably in the range of about 10 to about 100 phosphate units, and most preferred about 50 phosphate units.
  4. 4. The method according to any of claims 1 to 3, wherein the calcium salt is calcium chloride.
  5. 5. The method according to any of claims 1 to 4, wherein the pH is adjusted to 10.
  6. 6. The method according to any of claims 1 to 5, wherein the calcium polyphosphate material is formed from sodium polyphosphate in the presence of calcium chloride at a stoichiometric ratio of 0.1 to 10 (phosphate to calcium), preferably of 1 to 2.
  7. 7. The method according to any of claims 1 to 6, wherein the calcium polyphosphate material is obtained by addition of a solution containing 14 g/L of calcium chloride or 28 g/L of calcium chloride to a solution containing 10 g/L of sodium polyphosphate.
  8. 8. The method according to any of claims 1 to 7, further comprising the step of producing hard amorphous and morphogenetically active polyphosphate nanoparticles, and/or a material containing such nanoparticles.
  9. 9. The nanoparticles or the material containing such nanoparticles produced according to claim 8 for use in bone regeneration and repair and in dentistry.
  10. 10. The nanoparticles or the material containing such nanoparticles produced according to claim 8 for use in in drug delivery.
GB1420363.2A 2014-11-17 2014-11-17 Morphogenetically active calcium polyphosphate nanoparticles Withdrawn GB2532283A (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
GB1420363.2A GB2532283A (en) 2014-11-17 2014-11-17 Morphogenetically active calcium polyphosphate nanoparticles
CN201580067850.6A CN106999624B (en) 2014-11-17 2015-11-10 Amorphous calcium polyphosphate nanoparticles having morphology-generating activity for use in therapy
US15/527,479 US10307350B2 (en) 2014-11-17 2015-11-10 Morphogenetically active amorphous calcium polyphosphate nanoparticles for therapeutic applications
EP15794139.4A EP3220966A2 (en) 2014-11-17 2015-11-10 Amorphous inorganic polyphosphate-calcium-phosphate and carbonate particles as morphogenetically active coatings and scaffolds
EP15794143.6A EP3220967B8 (en) 2014-11-17 2015-11-10 Morphogenetically active amorphous calcium polyphosphate nanoparticles for therapeutic applications
CN201580069333.2A CN106999630A (en) 2014-11-17 2015-11-10 It is used as the coated layer with morphogenic activity and the amorphous inorganic polyphosphate calcium phosphate and calcium carbonate granule of support
PCT/EP2015/076172 WO2016078963A2 (en) 2014-11-17 2015-11-10 Amorphous inorganic polyphosphate-calcium-phosphate and carbonate particles as morphogenetically active coatings and scaffolds
PCT/EP2015/076222 WO2016078971A1 (en) 2014-11-17 2015-11-10 Morphogenetically active amorphous calcium polyphosphate nanoparticles containing retinol for therapeutic applications
US15/527,520 US20190083679A1 (en) 2014-11-17 2015-11-10 Amorphous Inorganic Polyphosphate-Calcium-Phosphate And Carbonate Particles As Morphogenetically Active Coatings and Scaffolds
PCT/EP2015/076468 WO2016079006A1 (en) 2014-11-17 2015-11-12 Bioactive wound dressing and teeth coating based on morphogenetically active amorphous calcium polyphosphate
US15/527,553 US20170319740A1 (en) 2014-11-17 2015-11-12 Bioactive Wound Dressing and Teeth Coating Based on Morphogenetically Active Amorphous Calcium Polyphosphate
CN201580072176.0A CN107106729A (en) 2014-11-17 2015-11-12 Bioactivity wound dressing and tooth coating based on the amorphous calcium polyphosphate with morphogenic activity
EP15794552.8A EP3220968A1 (en) 2014-11-17 2015-11-12 Bioactive wound dressing and teeth coating based on morphogenetically active amorphous calcium polyphosphate

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
GB1420363.2A GB2532283A (en) 2014-11-17 2014-11-17 Morphogenetically active calcium polyphosphate nanoparticles

Publications (2)

Publication Number Publication Date
GB201420363D0 GB201420363D0 (en) 2014-12-31
GB2532283A true GB2532283A (en) 2016-05-18

Family

ID=52248440

Family Applications (1)

Application Number Title Priority Date Filing Date
GB1420363.2A Withdrawn GB2532283A (en) 2014-11-17 2014-11-17 Morphogenetically active calcium polyphosphate nanoparticles

Country Status (1)

Country Link
GB (1) GB2532283A (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016178748A1 (en) * 2015-05-06 2016-11-10 Sunny Delight Beverages Co. Calcium polyphosphate salts, methods of making and use in beverage compositions
GB2541897A (en) * 2015-09-02 2017-03-08 Ernst Ludwig Georg Muller Werner Amorphous inorganic polyphosphate-calcium-phosphate particles inducing bone formation
EP3269400A1 (en) * 2016-07-15 2018-01-17 NanotecMARIN GmbH Amoprhous hyaluronic acid-magnesium/calcium polyphosphate microparticles for cartilage regeneration and repair
GB2552649A (en) * 2016-07-25 2018-02-07 Ernst Ludwig Georg Muller Werner Amorphous strontium polyphosphate microparticles for treatment of osteoporosis and inducing bone cell mineralization
GB2563899A (en) * 2017-06-29 2019-01-02 Ernst Ludwig Georg Muller Werner Inorganic polyphosphate formulations for use in the treatment of Alzheimer disease
WO2019108815A1 (en) * 2017-11-30 2019-06-06 Colgate-Palmolive Company Oral care compositions

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH07219217A (en) * 1994-01-31 1995-08-18 Japan Synthetic Rubber Co Ltd Chemically amplified resist composition
WO2004007399A2 (en) * 2002-07-12 2004-01-22 Pilliar Robert M Method of manufacture of porous inorganic structures and infiltration with organic polymers
WO2008006204A2 (en) * 2006-07-12 2008-01-17 Mcgill University Fibrous calcium pyrophosphate particles and methods of making and using same

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH07219217A (en) * 1994-01-31 1995-08-18 Japan Synthetic Rubber Co Ltd Chemically amplified resist composition
WO2004007399A2 (en) * 2002-07-12 2004-01-22 Pilliar Robert M Method of manufacture of porous inorganic structures and infiltration with organic polymers
WO2008006204A2 (en) * 2006-07-12 2008-01-17 Mcgill University Fibrous calcium pyrophosphate particles and methods of making and using same

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10005670B2 (en) 2015-05-06 2018-06-26 Sunny Delight Beverages Co. Calcium polyphosphate salt particles and method of making
WO2016178747A1 (en) * 2015-05-06 2016-11-10 Sunny Delight Beverages Co. Calcium polyphosphate salt particles and method of making
WO2016178748A1 (en) * 2015-05-06 2016-11-10 Sunny Delight Beverages Co. Calcium polyphosphate salts, methods of making and use in beverage compositions
GB2541897A (en) * 2015-09-02 2017-03-08 Ernst Ludwig Georg Muller Werner Amorphous inorganic polyphosphate-calcium-phosphate particles inducing bone formation
EP3269400A1 (en) * 2016-07-15 2018-01-17 NanotecMARIN GmbH Amoprhous hyaluronic acid-magnesium/calcium polyphosphate microparticles for cartilage regeneration and repair
GB2552462A (en) * 2016-07-15 2018-01-31 Nanotecmarin Gmbh Amorphous hyaluronic acid magnesium/calcium polyphosphate microparticles for cartilage regeneration and repair
GB2552649A (en) * 2016-07-25 2018-02-07 Ernst Ludwig Georg Muller Werner Amorphous strontium polyphosphate microparticles for treatment of osteoporosis and inducing bone cell mineralization
GB2563899A (en) * 2017-06-29 2019-01-02 Ernst Ludwig Georg Muller Werner Inorganic polyphosphate formulations for use in the treatment of Alzheimer disease
WO2019108815A1 (en) * 2017-11-30 2019-06-06 Colgate-Palmolive Company Oral care compositions
CN111405928A (en) * 2017-11-30 2020-07-10 高露洁-棕榄公司 Oral care compositions
AU2018375426B2 (en) * 2017-11-30 2021-07-29 Colgate-Palmolive Company Oral care compositions
US11083674B2 (en) 2017-11-30 2021-08-10 Colgate-Palmolive Company Oral care compositions
AU2018375426C1 (en) * 2017-11-30 2022-02-03 Colgate-Palmolive Company Oral care compositions

Also Published As

Publication number Publication date
GB201420363D0 (en) 2014-12-31

Similar Documents

Publication Publication Date Title
Bellucci et al. Bioactive glass/hydroxyapatite composites: Mechanical properties and biological evaluation
Habraken et al. Calcium phosphates in biomedical applications: materials for the future?
GB2532283A (en) Morphogenetically active calcium polyphosphate nanoparticles
US8715744B2 (en) Inorganic resorbable bone substitute material
EP2794471B1 (en) Calcium phosphate material
Bellucci et al. Mg-and/or Sr-doped tricalcium phosphate/bioactive glass composites: Synthesis, microstructure and biological responsiveness
CN107185034B (en) Bone-cartilage defect integrated repair biological ceramic scaffold and preparation method and application thereof
Venkatraman et al. Biomineralization, mechanical, antibacterial and biological investigation of larnite and rankinite bioceramics
Niu et al. Development of a bioactive composite of nano fluorapatite and poly (butylene succinate) for bone tissue regeneration
US10307350B2 (en) Morphogenetically active amorphous calcium polyphosphate nanoparticles for therapeutic applications
Guo et al. Preparation, characterization, bioactivity and degradation behavior in vitro of copper-doped calcium polyphosphate as a candidate material for bone tissue engineering
Bulut et al. Evaluation of mechanical behavior, bioactivity, and cytotoxicity of chitosan/akermanite-TiO2 3D-printed scaffolds for bone tissue applications
Müller et al. Fabrication of a new physiological macroporous hybrid biomaterial/bioscaffold material based on polyphosphate and collagen by freeze-extraction
Chen et al. Biomimetic preparation of trace element-codoped calcium phosphate for promoting osteoporotic bone defect repair
WO2016078963A2 (en) Amorphous inorganic polyphosphate-calcium-phosphate and carbonate particles as morphogenetically active coatings and scaffolds
JP5354562B2 (en) Cement material, method for producing cement material, method for producing cement, and cement
Wu et al. Developing a novel calcium magnesium silicate/graphene oxide incorporated silk fibroin porous scaffold with enhanced osteogenesis, angiogenesis and inhibited osteoclastogenesis
Navarro et al. Biomimetic mineralization of ceramics and glasses
De Paula et al. Hydroxyapatite-alginate biocomposite promotes bone mineralization in different length scales in vivo
Korkusuz et al. Nanocrystalline apatite-based biomaterials and stem cells in orthopaedics
Medvecky et al. Osteogenic potential and properties of injectable silk fibroin/tetracalcium phosphate/monetite composite powder biocement systems
El-Maghraby et al. Preparation, structural characterization, and biomedical applications of gypsum-based nanocomposite bone cements
CN110624129B (en) Corrosion-resistant osteoinductive silk fibroin/hydroxyapatite/magnesium oxide gel sponge and preparation method thereof
Zanetti Characterization of Novel Akermanite: Poly-E-Caprolactone Scaffolds for Bone Tissue Engineering Aapplications Combined with Human Adipose-Derived Stem Cells
Chen et al. Mesoporous Bioactive Glass Promoted Osteoblast Proliferation and Differentiation In Vitro

Legal Events

Date Code Title Description
WAP Application withdrawn, taken to be withdrawn or refused ** after publication under section 16(1)