US20190365869A1 - Bioresponsive Particles - Google Patents

Bioresponsive Particles Download PDF

Info

Publication number
US20190365869A1
US20190365869A1 US16/428,986 US201916428986A US2019365869A1 US 20190365869 A1 US20190365869 A1 US 20190365869A1 US 201916428986 A US201916428986 A US 201916428986A US 2019365869 A1 US2019365869 A1 US 2019365869A1
Authority
US
United States
Prior art keywords
enzyme
silica
nanoparticles
groups
silyl
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.)
Abandoned
Application number
US16/428,986
Inventor
Jacques Lux
Robert F. Mattrey
Annie Yang
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.)
University of Texas System
Original Assignee
University of Texas System
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 University of Texas System filed Critical University of Texas System
Priority to US16/428,986 priority Critical patent/US20190365869A1/en
Publication of US20190365869A1 publication Critical patent/US20190365869A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/44Oxidoreductases (1)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/44Oxidoreductases (1)
    • A61K38/446Superoxide dismutase (1.15)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/4813Exopeptidases (3.4.11. to 3.4.19)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/50Hydrolases (3) acting on carbon-nitrogen bonds, other than peptide bonds (3.5), e.g. asparaginase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/51Lyases (4)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/22Echographic preparations; Ultrasound imaging preparations ; Optoacoustic imaging preparations
    • A61K49/221Echographic preparations; Ultrasound imaging preparations ; Optoacoustic imaging preparations characterised by the targeting agent or modifying agent linked to the acoustically-active agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/22Echographic preparations; Ultrasound imaging preparations ; Optoacoustic imaging preparations
    • A61K49/222Echographic preparations; Ultrasound imaging preparations ; Optoacoustic imaging preparations characterised by a special physical form, e.g. emulsions, liposomes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/22Echographic preparations; Ultrasound imaging preparations ; Optoacoustic imaging preparations
    • A61K49/222Echographic preparations; Ultrasound imaging preparations ; Optoacoustic imaging preparations characterised by a special physical form, e.g. emulsions, liposomes
    • A61K49/223Microbubbles, hollow microspheres, free gas bubbles, gas microspheres
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/22Echographic preparations; Ultrasound imaging preparations ; Optoacoustic imaging preparations
    • A61K49/222Echographic preparations; Ultrasound imaging preparations ; Optoacoustic imaging preparations characterised by a special physical form, e.g. emulsions, liposomes
    • A61K49/225Microparticles, microcapsules
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/088Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins conjugates with carriers being peptides, polyamino acids or proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/12Preparations containing radioactive substances for use in therapy or testing in vivo characterised by a special physical form, e.g. emulsion, microcapsules, liposomes, characterized by a special physical form, e.g. emulsions, dispersions, microcapsules
    • A61K51/1241Preparations containing radioactive substances for use in therapy or testing in vivo characterised by a special physical form, e.g. emulsion, microcapsules, liposomes, characterized by a special physical form, e.g. emulsions, dispersions, microcapsules particles, powders, lyophilizates, adsorbates, e.g. polymers or resins for adsorption or ion-exchange resins
    • A61K51/1244Preparations containing radioactive substances for use in therapy or testing in vivo characterised by a special physical form, e.g. emulsion, microcapsules, liposomes, characterized by a special physical form, e.g. emulsions, dispersions, microcapsules particles, powders, lyophilizates, adsorbates, e.g. polymers or resins for adsorption or ion-exchange resins microparticles or nanoparticles, e.g. polymeric nanoparticles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/107General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides
    • C07K1/1072General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides by covalent attachment of residues or functional groups
    • C07K1/1077General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides by covalent attachment of residues or functional groups by covalent attachment of residues other than amino acids or peptide residues, e.g. sugars, polyols, fatty acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N11/00Carrier-bound or immobilised enzymes; Carrier-bound or immobilised microbial cells; Preparation thereof
    • C12N11/14Enzymes or microbial cells immobilised on or in an inorganic carrier
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/0004Oxidoreductases (1.)
    • C12N9/0065Oxidoreductases (1.) acting on hydrogen peroxide as acceptor (1.11)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/96Stabilising an enzyme by forming an adduct or a composition; Forming enzyme conjugates
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y111/00Oxidoreductases acting on a peroxide as acceptor (1.11)
    • C12Y111/01Peroxidases (1.11.1)
    • C12Y111/01006Catalase (1.11.1.6)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y113/00Oxidoreductases acting on single donors with incorporation of molecular oxygen (oxygenases) (1.13)
    • C12Y113/12Oxidoreductases acting on single donors with incorporation of molecular oxygen (oxygenases) (1.13) with incorporation of one atom of oxygen (internal monooxygenases or internal mixed function oxidases)(1.13.12)
    • C12Y113/12005Renilla-luciferin 2-monooxygenase (1.13.12.5), i.e. renilla-luciferase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y115/00Oxidoreductases acting on superoxide as acceptor (1.15)
    • C12Y115/01Oxidoreductases acting on superoxide as acceptor (1.15) with NAD or NADP as acceptor (1.15.1)
    • C12Y115/01001Superoxide dismutase (1.15.1.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/17Metallocarboxypeptidases (3.4.17)
    • C12Y304/17011Glutamate carboxypeptidase (3.4.17.11)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y305/00Hydrolases acting on carbon-nitrogen bonds, other than peptide bonds (3.5)
    • C12Y305/01Hydrolases acting on carbon-nitrogen bonds, other than peptide bonds (3.5) in linear amides (3.5.1)
    • C12Y305/01001Asparaginase (3.5.1.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y404/00Carbon-sulfur lyases (4.4)
    • C12Y404/01Carbon-sulfur lyases (4.4.1)
    • C12Y404/01011Methionine gamma-lyase (4.4.1.11)

Definitions

  • ALL Acute lymphoblastic leukemia
  • our invention provides for incorporating this treatment enzyme in ultra-small particles that are porous to asparagine, but at the same time, prevent the entry of large components of the immune system, will protect a child from immune reactions, but will effectively deplete asparagine. This will not only increase the enzyme's functional life in the child's body, but will also eliminate the harmful immune responses. By creating this nanoscale “force field” around the cancer treatment enzyme, it can do its job to cure the children of cancer much more safely.
  • Prior enzyme shielding strategies include the silica coating of single enzymes for industrial use (US2014/0127778) and the passive accumulation of enzymes inside hollow silica shells (US2016/0243262). Neither approach is optimal. The first produces ultra-small nanoparticles; however, shielding is suboptimal as enzyme activity decreases by 90 percent at room temperature in one week, and even faster in vivo and at body temperature, since plasma contains protein-cleaving enzymes to weaken the shield and deactivate the enzyme. The second approach has particle size and enzyme loading limitations. Since it is produced using two templates of markedly different sizes to create hollow silica shells with relatively large pores to allow entry of enzymes prior to closing the pores, particles cannot be made smaller than 100-200 nanometers.
  • Our invention allows continuous fine-tuning of enzyme activity per particle and particle size. Because we begin by attaching anchors on each enzyme molecule without affecting its function that serve as seeds upon which silica can deposit, we can control how many enzyme molecules we can put together in each particle to provide full control of number of enzyme molecules per particle and ultimately particle size.
  • the invention provides methods and compositions for shielding enzymes with silica. We use enone groups to decorate enzymes, which then allow the facile reaction of the silyl amine derivative to obtain silyl groups on the enzyme which acts as the seed for the growth of the siloxane scaffold around the enzyme (nanoporous silica network/shell that protects the enzymes).
  • the invention provides a silica modified enzyme comprising an enzyme covalently decorated with enone groups, around which is grown a siloxane scaffold, to form a hybrid enzyme-silica nanoparticle (HES-NP). Loading the silica modified enzyme into a silica nanoshell protects the enzyme, such as from inactivation by proteolysis
  • the invention provides a method of making a silica-modified enzyme comprising the steps of: a) reacting an acrylic compound (acryloyl derivative) with amine groups of an enzyme to covalently decorate the enzyme with enone groups; and b) coupling a silyl amine to the enone groups to covalently decorate the enzyme with silyl groups, forming a silica-modified enzyme.
  • the invention provides a method of making hybrid enzyme-silica nanoparticles (HES-NPs) comprising the steps of: (i) growing a siloxane scaffold around a silica-modified enzyme wherein the silyl groups seed the growth of the siloxane scaffold (e.g., in an emulsion or aqueous medium) to form hybrid enzyme-silica nanoparticles (HES-NPs); and (ii) isolating (e.g. from the emulsion or medium) the hybrid enzyme-silica nanoparticles.
  • HES-NPs hybrid enzyme-silica nanoparticles
  • the invention includes all combinations of the recited particular embodiments as if each combination had been laboriously separately recited.
  • FIG. 1 Schematic representation of the preparation of hybrid enzyme-silica nanoparticles using catalase as a model enzyme (CAT-HES-NP).
  • FIG. 2 Representative TEM images of HES-NP produced under aqueous (left) or reverse emulsion (right) conditions.
  • FIG. 3A Characterization of HES-NP obtained by reverse emulsion: TRPS and intensity-weighted DLS size distribution.
  • FIG. 3B Number-weighted size distribution.
  • FIG. 4 Activity of free catalase and CAT-HES-NP after incubation at 37° C. for 16 h with or without proteinase K.
  • our method to encapsulate catalase and also to encapsulate asparaginase.
  • Our novel approach is not enzyme-specific and can be applied to any enzymes.
  • Other exemplary enzymes include but are not limited to superoxide dismutase, methioninase, carboxypeptidase G2 and luciferase.
  • the invention provides a method for coating enzymes in nanoporous silica that allows free access to small molecules substrates, but not larger molecules such as antibodies or immune cells to be used as a treatment or imaging tool without interacting with the immune system. This approach extends the enzyme's activity in vivo and limits or prevents immune reactions.
  • Enzyme i.e., catalase, superoxide dismutase, asparaginase etc.
  • 36 mg was dissolved in sodium carbonate buffer (7.2 mL, 20 mM, pH 9.15) and a solution of N-acryloxysuccinimide (36 mg, in DMSO (72 ⁇ L) was added.
  • the silica-modified enzyme was filtered through a syringe filter (0.2 ⁇ m) to remove large aggregates.
  • the silica-modified enzyme was then formulated into particles using two different formulations.
  • the first method aqueous conditions
  • the second method reverse emulsion
  • Tetraethoxysilane (240 ⁇ L) was added to the silica-modified enzyme solution in water (1.5 mg/mL, 2 mL). The resulting mixture was stirred vigorously for 10 minutes and ammonium hydroxide (7.2 ⁇ L of 28% NH 4 OH solution) was added to hydrolyze silane groups and start the silica particle growth. The resulting emulsion was stirred vigorously for 2 hours at room temperature particles were collected by high speed centrifugation at 20,000 g for 15 minutes. After this time, supernatant was discarded and pellets were redispersed in water (4 mL) for a second wash and centrifugation. The supernatant was discarded a second time and pelleted particles were dispersed in water for storage and characterization.
  • Tetraethoxysilane (142 ⁇ L) was added to the silica-modified enzyme solution (1.5 mg/mL, 500 ⁇ L) under reverse emulsion conditions with decane (oil phase, 28.409 mL), IGEPAL® CO-520 (surfactant, 2.318 mL) n-hexanol (co-surfactant, 784 ⁇ L). The resulting mixture was stirred vigorously for 10 minutes and ammonium hydroxide (71 ⁇ L of 28% NH 4 OH solution) was added to hydrolyze silane groups and start the silica particle growth. The resulting emulsion was stirred vigorously overnight at room temperature and ethanol (16 mL) was added to remove surfactants and precipitate the particles.
  • decane oil phase, 28.409 mL
  • IGEPAL® CO-520 surfactant, 2.318 mL
  • n-hexanol co-surfactant, 784 ⁇ L
  • the resulting bottom layer was extracted and submitted to high speed centrifugation at 20,000 g for 15 minutes. After this time, supernatant was discarded and pellets were redispersed in water (4 mL) for a second wash and centrifugation. The supernatant was discarded a second time and pelleted particles were dispersed in water for storage and characterization.
  • Nanoparticles were sonicated at 10° C. for three minutes in a bath sonicator before size measurements to prevent aggregation.
  • Transmission electron microscopy (TEM, FEI Tecnai G2 Spirit transmission electron microscope equipped with a Gatan camera operating at 120 kV with Digital Micrograph software) was performed with negative staining (2% uranyl acetate in water) and TEM pictures were taken and showed monodisperse particles with sizes between 30 and 60 nm ( FIG. 2 )
  • DLS Dynamic Light Scattering
  • TRPS Resistive Pulse Sensing

Abstract

Shielding enzymes are made by modifying the enzyme surface with silica precursors and then depositing silica to a desired thickness while retaining biological activity of the enzyme.

Description

  • Priority: This application claims priority to Ser No. 62/679,762, filed: Jun. 01, 2018.
  • This invention was made with government support under Grant Number UL1TR001105 awarded by the National Institutes of Health (NIH). The government has certain rights in the invention.
  • INTRODUCTION
  • Acute lymphoblastic leukemia (ALL) is the most common childhood cancer accounting for more than 25 percent of all pediatric cancers in the U.S. Unfortunately, 30 percent of children have immune responses to one of the most effective treatments for ALL that is highly allergenic. These immune responses either render the treatment completely ineffective, particularly when children relapse, or worse, immediately threaten the life of the child, or both. Because this treatment is essential for permanently curing children of ALL, it is critical that novel strategies be devised to completely eliminate these immune reactions.
  • In an aspect, our invention provides for incorporating this treatment enzyme in ultra-small particles that are porous to asparagine, but at the same time, prevent the entry of large components of the immune system, will protect a child from immune reactions, but will effectively deplete asparagine. This will not only increase the enzyme's functional life in the child's body, but will also eliminate the harmful immune responses. By creating this nanoscale “force field” around the cancer treatment enzyme, it can do its job to cure the children of cancer much more safely.
  • Prior enzyme shielding strategies include the silica coating of single enzymes for industrial use (US2014/0127778) and the passive accumulation of enzymes inside hollow silica shells (US2016/0243262). Neither approach is optimal. The first produces ultra-small nanoparticles; however, shielding is suboptimal as enzyme activity decreases by 90 percent at room temperature in one week, and even faster in vivo and at body temperature, since plasma contains protein-cleaving enzymes to weaken the shield and deactivate the enzyme. The second approach has particle size and enzyme loading limitations. Since it is produced using two templates of markedly different sizes to create hollow silica shells with relatively large pores to allow entry of enzymes prior to closing the pores, particles cannot be made smaller than 100-200 nanometers. This relatively large size forces them to stay in blood speeding their removal by the liver and spleen, and limiting their ability to reach the cellular microenvironment in sufficient quantities to adequately fight cancer or provide enzymes to cells that so desperately need them. Further, since they are filled by suspending them in aqueous solutions of the enzyme, they can only trap the amount that can be dissolved without precipitation limiting enzyme loading. Low enzyme activity requires higher dosages to achieve the enzyme activity needed for the desired application, increasing toxicity.
  • Our invention allows continuous fine-tuning of enzyme activity per particle and particle size. Because we begin by attaching anchors on each enzyme molecule without affecting its function that serve as seeds upon which silica can deposit, we can control how many enzyme molecules we can put together in each particle to provide full control of number of enzyme molecules per particle and ultimately particle size.
  • Relevant Literature
  • Trogler et al., US20150273061
  • Yang et al., In situ synthesis of porous silica nanoparticles for covalent immobilization of Enzymes, Nanoscale 2012, 4, 414
  • Ortac I, Simberg O, Yeh Y S, Yang J, Messmer B, Trogler W C, Tsien R Y, Esener S. Dualporosity hollow nanoparticles for the immunoprotection and delivery of nonhuman enzymes. Nano Lett. 2014;14(6):3023-32. doi: 10.1021/nl404360k. PubMed PMID: 24471767; PMCID: PMC4059531.
  • Olson E S, Ortac I, Malone C, Esener S, Mattrey R. Ultrasound Detection of Regional Oxidative Stress in Deep Tissues Using Novel Enzyme Loaded Nanoparticles. Adv Healthc Mater. 2017;6(5). doi: 10.1002/adhm.201601163. PubMed PMID: 28081299; PMCID: PMC5516546.
  • Aspects of this disclosure were presented at Bioengineering Seminar at UT Arlington. Bioresponsive Particles for the Detection of Disease by Ultrasound. Nov. 1, 2017.
  • SUMMARY OF THE INVENTION
  • The invention provides methods and compositions for shielding enzymes with silica. We use enone groups to decorate enzymes, which then allow the facile reaction of the silyl amine derivative to obtain silyl groups on the enzyme which acts as the seed for the growth of the siloxane scaffold around the enzyme (nanoporous silica network/shell that protects the enzymes).
  • The invention provides a silica modified enzyme comprising an enzyme covalently decorated with enone groups, around which is grown a siloxane scaffold, to form a hybrid enzyme-silica nanoparticle (HES-NP). Loading the silica modified enzyme into a silica nanoshell protects the enzyme, such as from inactivation by proteolysis
  • In an aspect the invention provides a method of making a silica-modified enzyme comprising the steps of: a) reacting an acrylic compound (acryloyl derivative) with amine groups of an enzyme to covalently decorate the enzyme with enone groups; and b) coupling a silyl amine to the enone groups to covalently decorate the enzyme with silyl groups, forming a silica-modified enzyme.
  • In embodiments:
      • the acrylic compound comprises an acryloyl group and an N-hydroxysuccinimide group, such as N-acryloxysuccinimide or acrylate-polyethyleneglycol N-hydroxysuccinimide; and/or
      • the silyl amine is comprises a silyl ether group and an amine group, such as 3-aminopropyl trimethoxysilane (APTMS) or 3-aminopropyl triethoxysilane (APTES).
  • In another aspect the invention provides a method of making hybrid enzyme-silica nanoparticles (HES-NPs) comprising the steps of: (i) growing a siloxane scaffold around a silica-modified enzyme wherein the silyl groups seed the growth of the siloxane scaffold (e.g., in an emulsion or aqueous medium) to form hybrid enzyme-silica nanoparticles (HES-NPs); and (ii) isolating (e.g. from the emulsion or medium) the hybrid enzyme-silica nanoparticles.
  • In embodiments:
      • step (i) comprises contacting the silica-modified enzyme with tetraethoxysilane under aqueous conditions and hydrolyzing (e.g., with ammonium hydroxide) silane groups to start the growth of the siloxane scaffold;
      • step (i) comprises contacting the silica-modified enzyme with tetraethoxysilane under reverse emulsion conditions and hydrolyzing silane groups to start the growth of the siloxane scaffold;
      • the method further comprises the antecedent steps of: a) reacting an acrylic compound (acryloyl derivative) with amine groups of an enzyme to covalently decorate the enzyme with enone groups; and b) coupling a silyl amine to the enone groups to covalently decorate the enzyme with silyl groups, forming a silica-modified enzyme, wherein embodiment: the acrylic compound comprises an acryloyl group and a N-hydroxysuccinimide group, such as N-acryloxysuccinimide or acrylate-polyethyleneglycol N-hydroxysuccinimide; and/or the silyl amine comprises a silyl ether group and an amine group, such as 3-aminopropyl trimethoxysilane (APTMS) or 3-aminopropyl triethoxysilane (APTES);
      • the enzyme is selected from catalase, superoxide dismutase, asparaginase, methioninase, carboxypeptidase G2 and luciferase;
      • the nanoparticles are of average size 20-100 nm or 20-50 nm diameter;
      • the method further comprises the step of administering the nanoparticles to a person in need thereof, and particularly wherein:
      • the enzyme is catalase;
      • the nanoparticles provide a bioresponsive ultrasound contrast agent, and imaging the patient by ultrasound, such as wherein the enzyme is catalase, effective to generate O2 bubbles;
      • the person has or is at (imminent, demonstrable) risk of reperfusion injury and the enzyme is catalase, effective to scavenge reactive oxygen species (ROS);
      • the person has leukemia (e.g. acute lymphoblastic leukemia, ALL) and the enzyme is asparaginase, effective to deplete asparagine; and/or
      • the person is, has been or will be administered a prodrug, and the enzyme is prodrug converting enzyme, effective to convert the prodrug to a therapeutic drug.
  • The invention includes all combinations of the recited particular embodiments as if each combination had been laboriously separately recited.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1. Schematic representation of the preparation of hybrid enzyme-silica nanoparticles using catalase as a model enzyme (CAT-HES-NP).
  • FIG. 2. Representative TEM images of HES-NP produced under aqueous (left) or reverse emulsion (right) conditions.
  • FIG. 3A. Characterization of HES-NP obtained by reverse emulsion: TRPS and intensity-weighted DLS size distribution. FIG. 3B. Number-weighted size distribution.
  • FIG. 4. Activity of free catalase and CAT-HES-NP after incubation at 37° C. for 16 h with or without proteinase K.
  • DESCRIPTION OF PARTICULAR EMBODIMENTS OF THE INVENTION
  • We disclose a novel hybrid approach to shielding enzymes. We first modify the enzyme surface with silica precursors and then proceed to deposit silica to a desired thickness while retaining its biological activity. An advantage of this approach is that we can control final nanoparticle size and desired enzyme activity per particle by incorporating one or more or different enzyme molecules to optimize delivery and efficacy. Unlike passive trapping of enzymes in hollow silica spheres that utilize templates ≥100 nanometers, our nanoparticles can be made as small as 20-50 nanometer to achieve optimal delivery and enzyme activity. In an embodiment we exemplify the method with catalase as a model enzyme because it can be used to detect tissues in oxidative stress using ultrasound imaging, can be used as an anti-oxidant, and its activity is easily measured using commercial assay kits. In another embodiment example, we used our method to encapsulate catalase and also to encapsulate asparaginase. Our novel approach is not enzyme-specific and can be applied to any enzymes. Other exemplary enzymes include but are not limited to superoxide dismutase, methioninase, carboxypeptidase G2 and luciferase.
  • The invention provides a method for coating enzymes in nanoporous silica that allows free access to small molecules substrates, but not larger molecules such as antibodies or immune cells to be used as a treatment or imaging tool without interacting with the immune system. This approach extends the enzyme's activity in vivo and limits or prevents immune reactions.
  • General procedure for the preparation and characterization of hybrid enzyme-silica nanoparticles (HES-NP):
  • Preparation of HES-NP
  • Enzyme (i.e., catalase, superoxide dismutase, asparaginase etc.) (36 mg) was dissolved in sodium carbonate buffer (7.2 mL, 20 mM, pH 9.15) and a solution of N-acryloxysuccinimide (36 mg, in DMSO (72 μL) was added. The resulting mixture was stirred for 1 hour at room temperature and was purified by spin filtration in Amicon spin filters (Molecular weight Cutoff=10 kDa) at 4,000 g for 10 min. The filtrate was discarded, and the retentate was washed with water and spin filtered again at 4,000 g for 10 more minutes to yield the enone-modified enzyme (FIG. 1, step a). The purified enone-modified enzyme (3 mL, 12 mg/mL) was diluted down to a concentration of 1.5 mg/mL in 1M phosphate buffer pH 6.0 and water. (3-aminopropyl) trimethoxysilane (96 μL) was then added the resulting mixture was stirred for 1 hour at room temperature and purified by spin filtration in Amicon spin filters (Molecular weight Cutoff=10 kDa) at 4,000 g for 10 min. The filtrate was discarded, and the retentate was washed with water and spin filtered again at 4,000 g for 10 more minutes to yield the silica-modified enzyme (FIG. 1. Step b).
  • Before particle formulations, the silica-modified enzyme was filtered through a syringe filter (0.2 μm) to remove large aggregates. The silica-modified enzyme was then formulated into particles using two different formulations. The first method (aqueous conditions) yields nanoparticles around 100 nm and the second method (reverse emulsion) yield ultrasmall nanoparticles around 50 nm.
  • A] Aqueous conditions
  • Tetraethoxysilane (240 μL) was added to the silica-modified enzyme solution in water (1.5 mg/mL, 2 mL). The resulting mixture was stirred vigorously for 10 minutes and ammonium hydroxide (7.2 μL of 28% NH4OH solution) was added to hydrolyze silane groups and start the silica particle growth. The resulting emulsion was stirred vigorously for 2 hours at room temperature particles were collected by high speed centrifugation at 20,000 g for 15 minutes. After this time, supernatant was discarded and pellets were redispersed in water (4 mL) for a second wash and centrifugation. The supernatant was discarded a second time and pelleted particles were dispersed in water for storage and characterization.
  • B] Reverse emulsion conditions
  • Tetraethoxysilane (142 μL) was added to the silica-modified enzyme solution (1.5 mg/mL, 500 μL) under reverse emulsion conditions with decane (oil phase, 28.409 mL), IGEPAL® CO-520 (surfactant, 2.318 mL) n-hexanol (co-surfactant, 784 μL). The resulting mixture was stirred vigorously for 10 minutes and ammonium hydroxide (71 μL of 28% NH4OH solution) was added to hydrolyze silane groups and start the silica particle growth. The resulting emulsion was stirred vigorously overnight at room temperature and ethanol (16 mL) was added to remove surfactants and precipitate the particles. The resulting bottom layer was extracted and submitted to high speed centrifugation at 20,000 g for 15 minutes. After this time, supernatant was discarded and pellets were redispersed in water (4 mL) for a second wash and centrifugation. The supernatant was discarded a second time and pelleted particles were dispersed in water for storage and characterization.
  • Characterization of HES-NP:
  • Nanoparticles were sonicated at 10° C. for three minutes in a bath sonicator before size measurements to prevent aggregation. Transmission electron microscopy (TEM, FEI Tecnai G2 Spirit transmission electron microscope equipped with a Gatan camera operating at 120 kV with Digital Micrograph software) was performed with negative staining (2% uranyl acetate in water) and TEM pictures were taken and showed monodisperse particles with sizes between 30 and 60 nm (FIG. 2)
  • The hydrodynamic diameter of HES-NP was measured at 122.6 nm with a PdI of 0.168 by Dynamic Light Scattering (DLS, FIG. 3A) (Zetasizer ZS, Malvern Instruments). HES-NP were measured with a mean diameter of 117 nm (StdDev=40.5) with a concentration of 1.7×1012 NPs/mL by Tunable Resistive Pulse Sensing (TRPS, IZON, qNano Gold). The difference in size between the real size (TEM) and the size measured by DLS is explained by the higher scattering from larger molecules increasing the overall hydrodiameter of the particle population. This is demonstrated by displaying the size distribution of the particle population weighted by number, which provides a mean diameter around 50 nm (FIG. 3B). In the case of TRPS the overestimation of the size is due to the physical limitations of the instrument which uses a nanopore that cannot detect particles under the size of 50 nm, which means that we overestimate the size and underestimate the count of our nanoparticles using this method.
  • Stability Measurements of HES-NP:
  • To confirm that enzyme-loaded silica nanoshells protect enzymes from inactivation by proteolysis, I evaluated the activity of free enzyme and encapsulated enzyme in the presence of proteinase K, a serine protease that cleaves a wide range of proteins. In this experiment, we used catalase as a model protein, as it is not expensive, allows facile observation of activity by naked eye (bubbles generated upon addition of H2O2) and quantitative measurement of the enzymatic activity using a fluorometric assay. Specifically, we incubated free catalase and CAT-HES-NP overnight at 37° C. in pure water in the presence of CaCl2 (10 mM, 50 μL) and proteinase K (50 μL at 1 mg/mL). After 16 h, free catalase kept only 6% of its activity, while CAT-HES-NP kept 87% of its activity (FIG. 4). This slight activity loss is most likely due to the degradation of catalase attached at the surface of the particle.
  • It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims. All publications, patents, and patent applications cited herein, including citations therein, are hereby incorporated by reference in their entirety for all purposes.

Claims (20)

What is claimed is:
1. A method of making a silica-modified enzyme, comprising the steps of:
a) reacting an acrylic compound (acryloyl derivative) with amine groups of an enzyme to covalently decorate the enzyme with enone groups; and
b) coupling a silyl amine to the enone groups to covalently decorate the enzyme with silyl groups, forming a silica-modified enzyme.
2. The method of claim 1 wherein the acrylic compound comprises an acryloyl group and a N-hydroxysuccinimide group, such as N-acryloxysuccinimide or acrylate-polyethyleneglycol N-hydroxysuccinimide.
3. The method of claim 1 wherein the silyl amine is comprises a silyl ether group and an amine group, such as 3-aminopropyl trimethoxysilane (APTMS) or 3-aminopropyl triethoxysilane (APTES).
4. The method of claim 2 wherein the silyl amine is comprises a silyl ether group and an amine group, such as 3-aminopropyl trimethoxysilane (APTMS) or 3-aminopropyl triethoxysilane (APTES).
5. The method of claim 1 wherein the enzyme is selected from catalase, superoxide dismutase, asparaginase, methioninase, carboxypeptidase G2 and luciferase.
6. The method of claim 4 wherein the enzyme is selected from catalase, superoxide dismutase, asparaginase, methioninase, carboxypeptidase G2 and luciferase.
7. A method of making hybrid enzyme-silica nanoparticles (HES-NPs) using a silica-modified enzyme synthesizable by the method of claim 1, comprising the steps of:
i) growing a siloxane scaffold around the silica-modified enzyme, wherein the silyl groups seed the growth of the siloxane scaffold (e.g., in an emulsion or aqueous medium) to form hybrid enzyme-silica nanoparticles (HES-NPs); and
ii) isolating (e.g. from the emulsion or medium) the hybrid enzyme-silica nanoparticles.
8. The method of claim 7 wherein step (i) comprises contacting the silica-modified enzyme with tetraethoxysilane under surfactant-free aqueous conditions and hydrolyzing (e.g. with ammonium hydroxide) silane groups to start the growth of the siloxane scaffold.
9. The method of claim 7 wherein step (i) comprises contacting the silica-modified enzyme with tetraethoxysilane under reverse emulsion conditions and hydrolyzing silane groups to start the growth of the siloxane scaffold.
10. The method of claim 7 further comprising the antecedent steps of:
a) reacting an acrylic compound (acryloyl derivative) with amine groups of an enzyme to covalently decorate the enzyme with enone groups; and
b) coupling a silyl amine to the enone groups to covalently decorate the enzyme with silyl groups, forming a silica-modified enzyme.
11. The method of claim 10 wherein:
the acrylic compound comprises an acryloyl group and a N-hydroxysuccinimide group, such as N-acryloxysuccinimide or acrylate-polyethyleneglycol N-hydroxysuccinimide; and
the silyl amine is comprises a silyl ether group and an amine group, such as 3-aminopropyl trimethoxysilane (APTMS) or 3-aminopropyl triethoxysilane (APTES).
12. The method of claim 7 wherein the enzyme is selected from catalase, superoxide dismutase, asparaginase, methioninase, carboxypeptidase G2 and luciferase.
13. The method of claim 11 wherein the enzyme is selected from catalase, superoxide dismutase, asparaginase, methioninase, carboxypeptidase G2 and luciferase.
14. The method of claim 7 wherein the nanoparticles are of average size 20-100 nm or 20-50 nm diameter.
15. The method of claim 7 further comprising the step of administering the nanoparticles to a person in need thereof.
16. The method of claim 7 further comprising the steps of administering the nanoparticles to a person in need thereof, the enzyme is catalase.
17. The method of claim 7 further comprising the step of administering the nanoparticles to a person in need thereof, wherein the nanoparticles provide a bioresponsive ultrasound contrast agent, and imaging the patient by ultrasound, such as wherein the enzyme is catalase, effective to generate O2 bubbles.
18. The method of claim 7 further comprising the step of administering the nanoparticles to a person in need thereof, wherein the person has or is at (imminent, demonstrable) risk of reperfusion injury and the enzyme is catalase, effective to scavenge reactive oxygen species (ROS).
19. The method of claim 7 further comprising the step of administering the nanoparticles to a person in need thereof, wherein the person has leukemia (e.g. acute lymphoblastic leukemia, ALL) and the enzyme is asparaginase, effective to deplete asparagine.
20. The method of claim 7 further comprising the step of administering the nanoparticles to a person in need thereof, wherein the person is, has been or will be administered a prodrug, and the enzyme is prodrug converting enzyme, effective to convert the prodrug to a therapeutic drug.
US16/428,986 2018-06-01 2019-06-01 Bioresponsive Particles Abandoned US20190365869A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/428,986 US20190365869A1 (en) 2018-06-01 2019-06-01 Bioresponsive Particles

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862679762P 2018-06-01 2018-06-01
US16/428,986 US20190365869A1 (en) 2018-06-01 2019-06-01 Bioresponsive Particles

Publications (1)

Publication Number Publication Date
US20190365869A1 true US20190365869A1 (en) 2019-12-05

Family

ID=68694411

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/428,986 Abandoned US20190365869A1 (en) 2018-06-01 2019-06-01 Bioresponsive Particles
US16/429,026 Active 2040-05-08 US11648297B2 (en) 2018-06-01 2019-06-02 Bioresponsive particles

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/429,026 Active 2040-05-08 US11648297B2 (en) 2018-06-01 2019-06-02 Bioresponsive particles

Country Status (1)

Country Link
US (2) US20190365869A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021168996A1 (en) * 2020-02-27 2021-09-02 Vivibaba, Inc. Catalase nanocapsules and methods for use

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8507606B2 (en) * 2002-12-20 2013-08-13 Battelle Memorial Institute Biocomposite materials and methods for making the same

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8507606B2 (en) * 2002-12-20 2013-08-13 Battelle Memorial Institute Biocomposite materials and methods for making the same

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021168996A1 (en) * 2020-02-27 2021-09-02 Vivibaba, Inc. Catalase nanocapsules and methods for use

Also Published As

Publication number Publication date
US20190367902A1 (en) 2019-12-05
US11648297B2 (en) 2023-05-16

Similar Documents

Publication Publication Date Title
CN106806343B (en) Folic acid and polydopamine modified tumor targeting mesoporous silica nanoparticle and preparation method and application thereof
Rancan et al. Skin penetration and cellular uptake of amorphous silica nanoparticles with variable size, surface functionalization, and colloidal stability
EP3284483B1 (en) Hollow silica nanoparticles with encapsulated bioactive ingredients, preparation process and applications thereof
US20190111000A1 (en) Multistage delivery of active agents
CN106806344B (en) Polydopamine and polyethylene glycol vitamin E succinate modified mesoporous silica nanoparticle and preparation method and application thereof
EP2109445B1 (en) Compositions for topical application comprising a peroxide and retinoid
CN100408026C (en) Ceramic based nanoparticles for entrapping therapeutic agents for photodynamic therapy and method of using same
Ali et al. Fabrication and evaluation of ketorolac loaded cubosome for ocular drug delivery
CN101658533A (en) Delivery of stem cells of antitumor medicament
EP3154521B1 (en) Disintegratable core/shell silica particles for encapsulating and releasing bioactive macromolecules
JP2008501509A (en) Non-aggregating core / shell nanocomposite particles
CN111265533A (en) Preparation method of core-shell nanoparticles based on lipid membrane and metal organic framework
CN108126189B (en) Nano composite carrier drug delivery system assembled in grading manner and application thereof
Gunay et al. Development of nanosized, pramipexole-encapsulated liposomes and niosomes for the treatment of Parkinson’s disease
US20190365869A1 (en) Bioresponsive Particles
KR102391328B1 (en) Method for Manufacturing and Use of Urease-powered Polydopamine Nanomotors
CN114259477A (en) Nano delivery system capable of promoting penetration, relieving tumor hypoxia and targeting tumor cells, and preparation method and application thereof
CN108721321B (en) Tumor-targeted nano diagnosis and treatment agent, preparation method and application thereof
Sailor Biodegradable luminescent porous silicon nanoparticles for in vivo applications
Kenechukwu et al. Novel Bos indicus fat-based nanoparticulate lipospheres of miconazole nitrate as enhanced mucoadhesive therapy for oral candidiasis
US20230122688A1 (en) Method for preparing polydopamine nanomotor using urease, and use of same
Mahjoub et al. Preparation and optimization of controlled release nanoparticles containing cefixime using central composite design: an attempt to enrich its antimicrobial activity
CN113058031B (en) Galgi-body and genetic-engineering-exosome hybrid-membrane-coated retinoic acid in-situ spray hydrogel vaccine, and preparation method and application thereof
CN115350280B (en) Bionic nano-drug loaded with indocyanine green and inhibitor together and preparation method thereof
CN114796523B (en) Acid-resistant glycolipid modified liposome and application thereof

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION