US20160038608A1 - Silica-based mesoporous carrier and delivery method of using the same - Google Patents

Silica-based mesoporous carrier and delivery method of using the same Download PDF

Info

Publication number
US20160038608A1
US20160038608A1 US14/819,438 US201514819438A US2016038608A1 US 20160038608 A1 US20160038608 A1 US 20160038608A1 US 201514819438 A US201514819438 A US 201514819438A US 2016038608 A1 US2016038608 A1 US 2016038608A1
Authority
US
United States
Prior art keywords
targets
carriers
silica nanoparticles
mesoporous silica
cell
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
US14/819,438
Inventor
Chung-Yuan Mou
Yi-Ping Chen
Si-Han Wu
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.)
National Taiwan University NTU
Original Assignee
National Taiwan University NTU
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 National Taiwan University NTU filed Critical National Taiwan University NTU
Priority to US14/819,438 priority Critical patent/US20160038608A1/en
Assigned to NATIONAL TAIWAN UNIVERSITY reassignment NATIONAL TAIWAN UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHEN, YI-PING, MOU, CHUNG-YUAN, WU, Si-han
Priority to CA2994809A priority patent/CA2994809C/en
Priority to AU2016303039A priority patent/AU2016303039B2/en
Priority to EP16833417.5A priority patent/EP3331947A4/en
Priority to US15/750,759 priority patent/US11666662B2/en
Priority to PCT/US2016/014194 priority patent/WO2017023358A1/en
Priority to CN201680046902.6A priority patent/CN108368303A/en
Priority to JP2018506167A priority patent/JP2018533543A/en
Priority to TW105103067A priority patent/TWI611812B/en
Publication of US20160038608A1 publication Critical patent/US20160038608A1/en
Priority to IL257337A priority patent/IL257337A/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • A61K47/48869
    • 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
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6923Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being an inorganic particle, e.g. ceramic particles, silica particles, ferrite or synsorb
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/385Haptens or antigens, bound to carriers
    • 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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/24Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing atoms other than carbon, hydrogen, oxygen, halogen, nitrogen or sulfur, e.g. cyclomethicone or phospholipids
    • 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
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/42Proteins; Polypeptides; Degradation products thereof; Derivatives thereof, e.g. albumin, gelatin or zein
    • A61K47/48015
    • 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
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • 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
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6925Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a microcapsule, nanocapsule, microbubble or nanobubble
    • 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
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • G01N33/551Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals the carrier being inorganic
    • G01N33/552Glass or silica
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/005Assays involving biological materials from specific organisms or of a specific nature from viruses
    • G01N2333/08RNA viruses
    • G01N2333/15Retroviridae, e.g. bovine leukaemia virus, feline leukaemia virus, feline leukaemia virus, human T-cell leukaemia-lymphoma virus
    • G01N2333/155Lentiviridae, e.g. visna-maedi virus, equine infectious virus, FIV, SIV
    • G01N2333/16HIV-1, HIV-2
    • G01N2333/163Regulatory proteins, e.g. tat, nef, rev, vif, vpu, vpr, vpt, vpx

Definitions

  • the present invention generally relates to a mesoporous carrier, in particular, to silica-based mesoporous carriers and the delivery methods by using the silica-based mesoporous carriers.
  • porous materials can be classified as microporous materials having pore sizes of less than 2 nm, mesoporous materials having pore sizes of 2-50 nm and macroporous materials having pore sizes of greater than 50 nm.
  • the size of the pores and the large surface area of the pores allow the mesoporous materials to be ideal vehicles for carrying chemicals or drugs.
  • mesoporous carriers for delivering targets into a cell.
  • the mesoporous carriers comprise hollow silica nanospheres (HSN) or mesoporous silica nanoparticles (MSN) and the targets bound to or encapsulated by the hollow silica nanospheres or the mesoporous silica nanoparticles.
  • the targets includes first targets and second targets, and the first and second targets are different.
  • the targets may include peptides, proteins, enzymes and/or enzymatic mimetics.
  • a method of delivering targets into a cell is proposed.
  • mesoporous carriers are prepared and provided.
  • the mesoporous carriers comprises hollow silica nanospheres (HSN) or mesoporous silica nanoparticles (MSN) and the targets bound to or encapsulated by the hollow silica nanospheres or the mesoporous silica nanoparticles.
  • the targets include first targets and second targets, and the first and second targets are different.
  • the mesoporous carriers contact with the cell by incubating the cell with the mesoporous carriers.
  • the first targets and the second targets are co-delivered into the cell at the same time, as the mesoporous carriers and the targets carried by the hollow silica nanospheres or the mesoporous silica nanoparticles enter into the cell.
  • FIGS. 1A-1B show the reaction schemes for the PEI-modification of the enzymes SOD and CAT and the encapsulation of PEI-SOD and PEI-CAT within HSN.
  • FIG. 2 shows the adsorption and desorption isotherms of HSN according to one embodiment of the present invention.
  • FIGS. 3A-3D show transmission electron microscopy (TEM) images of various enzyme encapsulated HSN.
  • FIG. 4A shows the relative enzyme activity for different enzyme-encapsulated HSN.
  • FIG. 4B shows the fluorescence intensity over the wavelength for different enzyme-encapsulated HSN.
  • FIG. 5 shows the cell viability results using WST-1 assay when exposed to HSN or PEI-SOD/CAT@HSN.
  • FIG. 6 shows the quantification of fluorescence intensity of oxidized DHE from HeLa cells were treated with various enzyme encapsulated HSN.
  • FIG. 7 describes the reaction scheme for the conjugation of NF- ⁇ B p65 antibody and Cys-TAT peptide to the surface functionalized MSN.
  • FIGS. 8A-8D show transmission electron microscopy (TEM) images of various functionalized MSN nanoparticles.
  • FIG. 9A-9C show the results of in vitro pull-down assay of various functionalized MSN nanoparticles.
  • FIG. 10A shows the conjugation of FMSN-PEG/PEI nanoparticles.
  • FIG. 10B shows the TEM images of FMSN-PEG/PEI nanoparticles.
  • FIGS. 11A-11C show the protection effects of co-delivery of TAT-SOD and TAT-GPx into Hela cells.
  • silica-based mesoporous carrier materials are described.
  • the silica-based mesoporous materials may be classified as hollow silica nanospheres (HSN) and mesoporous silica nanoparticles (MSN) and the targets may be carried on the surface of the silica-based mesoporous materials or be encapsulated within the silica-based mesoporous materials.
  • HSN hollow silica nanospheres
  • MSN mesoporous silica nanoparticles
  • the targets may be carried on the surface of the silica-based mesoporous materials or be encapsulated within the silica-based mesoporous materials.
  • the ingredients, the reaction conditions or parameters illustrated in the examples are merely for illustration purposes, but it is not intended to limit the material or the preparation method by the exemplary embodiments described herein.
  • the target suitable for being carried by the silica-based mesoporous materials may include an enzymes containing cysteine (thiol group), lysine (amino group), aspartate or glutamate (carboxyl group), a peptide containing cysteine (thiol group), lysine (amino group), aspartate or glutamate (carboxyl group) or an antibody containing cysteine (thiol group), lysine (amino group), aspartate or glutamate (carboxyl group).
  • the target suitable for being carried by the silica-based mesoporous materials may include an enzymes containing polyhistidine-tag (His-tag), a peptide containing polyhistidine-tag or an antibody containing polyhistidine-tag.
  • the polyhistidine-tag consists of at least six histidine (His) residues.
  • hollow silica nanospheres with porous silica shells and large interior spaces (cavities) have been synthesized, which is suitable for loading enzymes within the cavities of HSN for intracellular catalysis.
  • a microemulsion method has been developed for synthesizing hollow silica nanospheres (HSN), of which one or more enzymes may be loaded within the cavities.
  • the enzymes may be antioxidant enzymes, including horseradish peroxidase (HRP), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase, glutathione reductase and their enzymatic mimetics.
  • the enzymes may be enzymes involved in biochemical enzymatic cascades, which refers to a series of biochemical reactions involving enzymes, such as blood coagulation, metabolism pathways, and signal transduction pathways.
  • HSN may be synthesized by silica sol-gel process of water-in-oil microemulsion, and polyethyleneimine (PEI) modified enzymes in aqueous phase are then encapsulated inside HSN. It has been demonstrated that encapsulation of HRP in the cavities of HSN (HRP@HSN) is feasible and the intracellular delivery of HRP@HSN showing its function as a catalytic nanoreactor inside Hela cells for converting prodrug into toxic agent to kill the cancer cells.
  • PEI polyethyleneimine
  • PEI grafted superoxide dismutase (PEI-SOD) and catalase (PEI-CAT) are prepared and then co-encapsulated within HSN as the loaded enzymes.
  • FIGS. 1A-1B describe the reaction schemes for the PEI-modification of the enzymes (SOD and CAT) and the encapsulation of the positively charged PEI-SOD and PEI-CAT within HSN.
  • SOD and CAT the enzymes
  • the silica shell of HSN shields off PEI from contacting with cellular machineries, but the pores of the silica shells allow protons to diffuse inside the hollow spheres of HSN and keep the proton sponge effects of PEI.
  • FIG. 1A the PEI-enzyme conjugation is achieved through the amidation of the carboxyl group with PEI.
  • FIG. 1B illustrates the synthesis of PEI-SOD and PEI-CAT co-encapsulated HSN (denoted as PEI-SOD/CAT@HSN).
  • PEI is covalently linked to the enzymes by the conventional EDC/NHS coupling reaction.
  • PEI-grafted enzymes (PEI-SOD, PEI-CAT), either individually or together, are encapsulated inside HSN by an one-pot water-in-oil (w/o) microemulsion approach as shown in FIG. 1B .
  • an aqueous solution containing PEI-grafted enzyme is emulsified in an oil system containing surfactant (isooctylphenyl ether, CA-520), co-surfactant (n-hexanol) and organic solvent (decane).
  • surfactant isooctylphenyl ether, CA-520
  • co-surfactant n-hexanol
  • organic solvent decane
  • TEOS tetraethoxysilane
  • APIMS aminopropyltrimethoxy silane
  • aqueous ammonia aqueous ammonia
  • the water-in-oil (w/o) mixture was stirred for 10 h at 20° C. to form enzyme-encapsulated silica nanoparticles.
  • Enzymes-loaded particles were foiiiied after the nanoparticles were further suspended in waiin deionized water (40° C.) for 40 min and washed with water, which is critical for the transformation to hollow nanospheres.
  • SOD and CAT Modification of SOD and CAT with PEI: PEI-conjugation of enzymes was carried using NHS/EDC crosslinking reaction. Briefly, 3 mg of SOD and CAT were dissolved in 50 mM sodium phosphate buffer (pH 7.8) at a concentration of 3 mg/mL. Amine-reactive NHS esters and EDC ⁇ HCl were prepared in 200 ⁇ L sodium phosphate buffer (50 mM, pH 7.8) and then added to the enzyme solution. The SOD solution contained 17 mg NHS/14 mg EDC ⁇ HCl and CAT solution contained 15 mg NHS/13 mg EDC ⁇ HCl.
  • PEI-SOD PEI-SOD
  • CAT PEI-CAT
  • Synthesis of PEI-SOD@HSN, PEI-CAT@HSN, and PEI-SOD/CAT@HSN Hollow silica nanospheres (HSN) were synthesized by a reverse microemulsion method. Typically, 20 mL of decane, 1.63 mL of CA-520, 550 ⁇ L of n-hexanol and 350 ⁇ L of H 2 O with concentrated PEI-enzymes (PEI-SOD, PEI-CAT, or a mixture of PEI-SOD and PEI-CAT) were mixed at room temperature to generate the water-in-oil microemulsion. Then, silica sources (100 ⁇ L of TEOS and 25 ⁇ L of ethanolic APTMS solution) were added with stirring.
  • silica sources 100 ⁇ L of TEOS and 25 ⁇ L of ethanolic APTMS solution
  • the ethanolic APTMS solution was prepared by adding 200 mL of APTMS to 1.4 mL of absolute ethanol. After 10 min, 250 ⁇ L of aqueous ammonia (35 wt %) was introduced to the mixture which was stirred for 10 h at 20° C. Then 95% ethanol was added to destabilize the microemulsion system and solid products were centrifuged at 11000 rpm for 30 min.
  • the samples were suspended in 40 mL warm water (40° C.), stirred for 40 minutes, and isolated by centrifugation to obtain PEI-SOD encapsulated HSN (denoted as PEI-SOD@HSN), PEI-CAT encapsulated HSN (denoted as PEI-CAT@HSN), PEI-SOD/CAT@HSN or HSN only (without adding enzymes). Finally, all nanoparticles were washed several times with ethanol and deionized water to extract the surfactants in the pores.
  • the amount of PEI-enzymes used to prepare enzyme-loaded HSN 36 nmole of PEI-SOD for PEI-SOD@HSN; 4 nmole of PEI-CAT for PEI-CAT@HSN; 36 nmole of PEI-SOD and 4 nmole of PEI-CAT for PEI-SOD/CAT@HSN.
  • FIGS. 3A-3D show transmission electron microscopy (TEM) images of enzyme encapsulated silica nanospheres; FIG. 3A : PEI-SOD/CAT@HSN, FIG. 3B : PEI-SOD/CAT@HSN stained with uranyl acetate (UA); FIG. 3C : PEI-SOD@HSN and FIG. 3D : PEI-CAT@HSN.
  • TEM transmission electron microscopy
  • PEI-SOD The loading efficiency of PEI-SOD (12.2%) and PEI-CAT (8.7%) were much higher than that of SOD (2.8%) and CAT (2.8%). This is due to the positively charged PEI-SOD and PEI-CAT are effective in attracting the negatively charged hydrolyzed silica precursors for encapsulation. The higher loading will improve the enzyme activity for biomedical use and thus PEI-coated enzymes are chosen for further study thereafter.
  • the loading yields of PEI-SOD@HSN and PEI-CAT@HSN were 44.7 and 29.5 ⁇ g enzymes/mg HSN. Compared with single enzyme loading, about the same level of efficiency and loading yields were observed when two enzymes, PEI-SOD and PEI-CAT, were co-encapsulated in HSN.
  • the loading efficiency of PEI-SOD and PEI-CAT were 12.3% and 9.4%.
  • PEI-SOD/CAT@HSN contains about 44.5 and 26.1 ⁇ g enzymes/mg HSN (Table
  • PEI-SOD/CAT@HSN and PEI-SOD@HSN maintain 46.6% and 18.6% of the native SOD activity (Table 2). Also, as seen in Table 2, when compared with the reaction rate of H 2 O 2 decomposition between native CAT and CAT-loaded particles, PEI-SOD/CAT@HSN and CAT@HSN keep 57.9% and 62.7% of the native CAT activity.
  • FIG. 4A shows the stability of CAT-loaded particles and native CAT after treated with KO 2 solution.
  • KO 2 solution which generates 0 2 • ⁇
  • native CAT lost almost all its activity
  • CAT activity of PEI-SOD/CAT@HSN and PEI-CAT@HSN still remained at about 80% and 70%.
  • SOD and CAT are antioxidant enzymes and work together as primary defence system against free radical damage in living cells.
  • PEI-SOD and PEI-CAT could be simultaneously encapsulated in HSN and the final nanoparticle system showed activities in both enzymes.
  • fluorescence assay was performed in which the resorufin formation was recorded by monitoring the fluorescence at 570 nm.
  • the O 2 • ⁇ generated by KO 2 was unstable and converted to H 2 O 2 in the presence or absence of SOD in aqueous solution.
  • the dismutation rate of O 2 • ⁇ in the presence of SOD is faster.
  • H 2 O 2 could be either reduced by CAT or further involved in peroxidase/Amplex red oxidation reaction.
  • the fluorescence intensity of PEI-SOD@HSN was significantly higher than that of blank (control), HSN (negative control), and PEI-SOD/CAT@HSN.
  • the cascade reaction occurs. Hydrogen peroxide produced by SOD could be further decomposed by CAT and the system showed weaker fluorescence.
  • FIG. 4B shows the cascade reactions within PEI-SOD/CAT@HSN.
  • the fluorescence intensity reflects the amount of H 2 O 2 in the assay buffer.
  • the same SOD units of PEI-SOD/CAT@HSN and PEI-SOD@HSN were used for comparison.
  • Cell proliferation assay 2 ⁇ 10 4 cells were seeded in 24-well plates and allowed to attach for 24 h. To determine the particle toxicity, cells were incubated in fresh serum-free medium containing different amounts of BSA-stabilized particles (0-400 ⁇ g/mL) for 2.5 h. After washing twice with phosphate-buffered saline (PBS), particle-treated cells were cultured in regular growth medium. After 24 h, cells were washed twice with PBS and incubated with 200 ⁇ L WST-1 (10%) in Dulbecco's modified eagles medium (DMEM). Cell viability was determined by the fornazan dye generated by the live cells and the absorbance at 450 nm was measured using a microplate reader (Bio-Rad, model 680).
  • PBS phosphate-buffered saline
  • DMEM Dulbecco's modified eagles medium
  • FIG. 5 shows the cell viability results for Hela cells exposed to HSN or PEI-SOD/CAT@HSN using WST-1 assay.
  • FIG. 6 shows the quantification of fluorescence intensity of oxidized DHE from HeLa cells were treated with various nanoparticles.
  • the PQ-induced ROS productions are the same for the un-protected cells (positive control and HSN only). All enzyme-nanoparticle treated cells obviously inhibit PQ-induced ROS production.
  • PEI-SOD@HSN and PEI-CAT@HSN show similar levels of reduction of DHE-positive cells by about 25% (compared to HSN only).
  • PEI-SOD/CAT@HSN shows a much higher level of reduction of DHE-positive cells by about 60%.
  • the PEI-SOD/CAT@HSN nanosystem clearly shows a synergetic effect in ROS reduction.
  • DHE-positive cells and fluorescence intensity quantitated indicates that PEI-SOD/CAT@HSN having the synergetic effect of both antioxidant enzymes displays the weakest fluorescence intensity under microscopic observation.
  • the enzyme-loaded particles, especially PEI-SOD/CAT@HSN show better cell attaching morphology, which implies the treatment by PEI-SOD/CAT@HSN detoxify the imposed ROS.
  • the nanoparticle-treated cells after incubation with PQ were further assayed with Western blotting to check the level of p-p38 and COX2 expression.
  • Down-regulation of PQ-induced activation of p-p38 and COX2 is observed for cells pre-treated with PEI-SOD/CAT@HSN, PEI-SOD@HSN and PEI-CAT@HSN.
  • the present invention provides a mesoporous hollow silica nanosphere (HSN) materials (about 40 nm size) based on templating water-in-oil with sol-gel condensation.
  • the enzyme SOD and/or catalase (CAT) can be individually or jointly encapsulated within HSN, with the help of the polyamine PEI.
  • the mesopores on the silica shell allow easy access of small molecules while keeping the enzymes inside the nanosphere from undesirable protein-protein interaction.
  • PEI-SOD and PEI-CAT@HSN can complete cascade transformation of superoxide through hydrogen peroxide to water with synergism.
  • HSN Downstream reactive oxygen species (ROS) production and COX-2/p-p38 expressions show that co-encapsulated SOD/CAT inside HSN give the highest cell protection against the toxicants.
  • ROS reactive oxygen species
  • PEI-SOD/CAT@HSN may be applied as antioxidants for medical treatments, such as inflammation, ischemia, stroke and other strong oxidative stress situation.
  • the sizes of many proteins or enzymes are significantly bigger than the inner space of HSN or MSN or certain proteins are not suitable to be encapsulated within the nanomaterials and the loading efficiency is limited. In this case, it is feasible to link or conjugate the enzymes or proteins to the exterior or outer surface of the nanoparticles or nanomaterials for better delivery efficiency.
  • mesoporous silica nanoparticle (MSN) materials are synthesized and functionalized to carry peptides and/or antibodies.
  • the peptide may be any peptide containing cysteine or a polyhistidine tag, including nucleus localization sequence (NLS)-peptide, cancer-targeting peptide and lysosomal targeting peptide.
  • the antibody may be any antibody containing cysteine or a polyhistidine tag, including signal transduction antibody and cancer-targeting antibody.
  • the present invention provides nanoparticles consisting of mesoporous silica nanoparticle (MSN) with surface functionalization of NF- ⁇ B (nuclear factor-kappa B) p65 antibody and TAT transducing peptide (i.e., HIV trans-activator of transcription (TAT) protein transduction domain).
  • TAT transducing peptide i.e., HIV trans-activator of transcription (TAT) protein transduction domain.
  • TAT transducing peptide i.e., HIV trans-activator of transcription (TAT) protein transduction domain.
  • TAT transducing peptide i.e., HIV trans-activator of transcription (TAT) protein transduction domain.
  • TAT transducing peptide i.e., HIV trans-activator of transcription (TAT) protein transduction domain
  • FIG. 7 describes the reaction scheme for the conjugation of NF- ⁇ B p65 antibody and Cys-TAT peptide to the surface functionalized MSN.
  • amine groups are formed on the surface of MSN by reacting with 3-aminopropyltrimethoxysilane (APTMS) to foim MSN-APTMS with an average loading of nitrogen content of APTMS at 2.6 wt % by elemental analysis.
  • APTMS 3-aminopropyltrimethoxysilane
  • PEG polyethylene glycol
  • MAL maleimide
  • PEG 2k or PEG 3.4k polyethylene glycol (PEG) having an average molecular weight of 2000 or 3400
  • SCM succinimidyl carboxymethyl.
  • the MAL-PEG-SCM crosslinkers containing a succinimidyl moiety react with the amine groups of MSN-APTMS through an active succinimidyl link to obtain the MSN-PEGs (MSN-PEGs: MSN-PEG 2k , MSN-PEG 34k ).
  • the MAL-end of MSN-PEG reacted with the thiol groups of the antibody and Cys-TAT peptide.
  • FITC-APTMS N-1-(3-trimethoxysilyl propyl)-N′-fluoreceylthiourea
  • FITC fluorescein isothiocyanate
  • APIMS 3-aminopropyltrimethoxysilane
  • 200 mg of as-synthesized samples were redispersed in 25 mL of 95% ethanol with 0.5 g of 37 wt % HCl.
  • Surfactant was extracted by heating the ethanol suspension at 60° C. for 24 h.
  • the product, called FITC-MSN (MSN) was collected by centrifugation and washed with ethanol several times and stored in ethanol.
  • MSN The surface of MSN was functionalized with amine groups by treatment with APTMS.
  • MSNs 200 mg were first dispersed in 50 mL of ethanol, and then the solution was refluxed for 18 h after the addition of 500 ⁇ L of APTMS. After centrifugation and washing with ethanol, amine-functionalized MSNs were redispersed in ethanol. To remove the surfactants, the amine-functionalized MSNs were suspended in acidic ethanol (1 g of HCl in 50 mL of EtOH) and refluxed for 24 h. After centrifugation and washing with ethanol, amine-functionalized MSN (MSN-APTMS) were redispersed in ethanol.
  • TEM images were taken using a Hitachi H-7100 instrument with an operating voltage of 75 KV. Samples were sonicated to disperse in ethanol and 10 ⁇ L of the suspension was dropped to fix on a microgrid.
  • the p65 antibody was covalently immobilized with the MAL-end of MSN-PEG 3.4k in different ratios (1:6, 1:12, 1:24) via C-S binding.
  • the Cys-TAT peptide was conjugated to fill up the free MAL-end of MSN-PEG 3.4k .
  • MSN-PEG 3.4k without antibody coupling was also directly conjugated with Cys-TAT as a control.
  • the physical properties of the nanoparticles were characterized by nitrogen adsorption-desorption isotherms, powder X-ray diffraction (XRD), FT-IR, TEM, dynamic light scattering (DLS) and zeta potential.
  • TEM 8A-8D show transmission electron microscopy (TEM) images of various functionalized MSN. From the TEM images, it is shown that the MSN particles possess well-ordered mesoporous structures and the average particle size obtained from TEM images is about 40 nm.
  • the WST-1 assay was applied to measure the cell viability and growth inhibition assay: 2 ⁇ 10 4 HeLa cells per well were seeded in 24-well plates for 16 h for HeLa cell viability assay. HeLa cells were incubated in serum-free medium containing different amounts of MSN-PEG 3.4k -Ab-TAT (100 ⁇ g/L) for 4 h.
  • HNSCC growth inhibition assay HNSCC cells were seeded in 24-well plates with a density of 4 ⁇ 10 4 cells/well for 16 h and incubated with 200 ⁇ g/mL of MSN-PEG 3.4k -Ab(1:24)-TAT, MSN-PEG 3.4k -TAT or anti-TNF antibody (100 ng/mL) in serum-free medium for 4 h. Following medium replacement with culture medium, HNSCC cells were incubated for further 72 h.
  • WST-1 assay HeLa or HNSCC cells were allowed to grow in culture medium containing WST-1 (Clontech) for 4 h at 37° C. The dark red formazan dye generated by the live cells was proportional to the number of live cells and the absorbance at 450 nm was measured using a microplate reader (Bio-Rad, model 680).
  • the cell viability of the MSN-PEG 34k -Ab-TAT was examined by using WST-1 assay and MSN-PEG 3.4k -Ab-TAT shows no significant cytotoxicity.
  • PVDF polyvinylidene difluoride
  • MSN-APTMS 100 ⁇ g/mL of MSN-APTMS, MSN-PEG 2k and MSN-PEG 3.4k were treated in HeLa cells for 4 h, and then incubation without or with TNF- ⁇ (50 ng/mL), a NF- ⁇ B activator, for another 0.5 h.
  • TNF- ⁇ 50 ng/mL
  • a NF- ⁇ B activator 50 ng/mL
  • the p65 expression level in either cytosol or nucleus was determined by western blotting experiments.
  • the MSN-PEG 3.4k -Ab-TAT 100 ⁇ g/mL was mixed and incubated with total lysate of HeLa cell at 4° C. for 18 h in vitro. Then, the mixture was centrifuged at 12,000 rpm for 20 mins and the supernatant (10 ⁇ L) was assayed for the free p65 expression level by Western blotting.
  • FIGS. 9A-9C show the results of in vitro pull-down assay of various functionalized MSN nanoparticles.
  • MSN-PEG 3.4k -Ab-TAT blocks NF- ⁇ B p65 nuclear translocation and thus inhibits the NF- ⁇ B p65 downstream protein expression.
  • HeLa cells were treated with MSN-PEG 3.4k -TAT or MSN-PEG 3.4k -Ab-TAT for 4 h at different doses (100 ⁇ ug/mL for FIG. 9B , 50-200 ⁇ g/mL for FIG. 9C ). After the delivery, the cells were stimulated with or without 5Ong/mL TNF- ⁇ for another 0.5 h.
  • Dose-dependence study of the blockage as shown in FIG. 9C indicates that nuclear p65 level decreases with the increasing concentration of MSN-PEG 3.4k -Ab(1:24)-TAT.
  • MSN-PEG 3.4k -Ab(1:12)-TAT and MSN-PEG 3.4k -Ab(1:24)-TAT show obvious suppression of the p65 translocation to nucleus, whereas MSN-PEG 3.4k -TAT did not prevent the TNF- ⁇ inducing nuclear p65 translocation.
  • MSN-PEG 3.4k -Ab-TAT shows the specificity and effectiveness to block NF- ⁇ B p65 nuclear translocation through immunogenic binding.
  • a nanoparticle/antibody complex targeting NF- ⁇ B is employed to catch the Rel protein p65 in perinuclear region and thus blocking the translocation near the nuclear pore gate.
  • TAT peptide conjugated on mesoporous silica nanoparticles (MSN) help non-endocytosis cell-membrane transducing and converge toward perinuclear region, where the p65 specific antibody performed the targeting and catching against active NF- ⁇ B p65 effectively.
  • a protein delivery system combining MSN nanoparticle carriers and one or more denatured fusion proteins has been developed.
  • Such combination of the nanomaterial and one or more fusion proteins not only solves the problems of protein delivery, including chemical solvents, stability, and permeability, but also provide a new approach for protein therapy.
  • TAT-SOD and TAT-GPX protein conjugation we constructed and overexpressed the His-tag human Cu, Zn-superoxide dismutase (SOD) and human glutathione peroxidase (GPx) which contain a human immunodeficiency virus (HIV) transducing domain (TAT, residues 49-57).
  • the sequence of TAT transducing peptide RKKRRQRRR.
  • the genes of TAT-SOD and TAT-GPx were cloned and inserted into prokaryotic protein expression vector of pQE-30 to form pQE-TAT-SOD and pQE-TAT-GPx. The vectors were transformed into JM109 E.
  • TAT-SOD and TAT-GPx with high protein overexpression were displayed in accordance with increasing induction time in 10% SDS-PAGE electrophoresis. Finally, the supernatants of pellets of E. coli crude lysates expressed TAT-SOD or TAT-GPx were tried to further directly conjugate in 8M urea.
  • FITC solution was prepared by dissolving 1 mg of FITC in 5 ml of anhydrous ethanol. 100 L of APTMS was added with rapid stirring at room temperature in darkness for 24 hours. 0.58 g of C 16 TAB was dissolved in 300 g of 0.17 M NH 3 solution, and 5 mL of dilute TEOS solution (5% v/v TEOS/ethanol) was added with stirring for 5 h. FITC-APTMS solution added before 5 ml of concentrate TEOS solution (25% v/v TEOS/ethanol) was added dropwise with vigorous stirring for 1 h. The solution was then aged at 40° C. for 24 hours to complete the silica condensation. As-synthesized products was collected by centrifugation and washed with 95% ethanol three times. The products called FITC-MSN (FMSN) were stored in absolute ethanol.
  • FMSN-NTA-Ni were obtained and stored in absolute ethanol. FMSN-NTA-Ni with an average loading of Ni content is 0.6 wt % by ICP-MS analysis.
  • FITC solution was prepared by dissolving 1 mg of FITC in 5 mL of anhydrous ethanol. 100 of APTMS was added with rapid stirring at room temperature in darkness for 24 hours. 0.58 g C 16 TAB was dissolved in 300 g of 0.17 M NH 3 solution, and 5 mL of dilute TEOS solution (5% v/v TEOS/ethanol) was added with stirring for 5 h. FITC-APTMS solution added before 5 mL of concentrate TEOS solution (25% v/v TEOS/ethanol) was added dropwise with vigorous stirring for 1 h.
  • TEM images were taken on a JEOL JSM-1200 EX II operating at 120 kV.
  • the nickel amount of sample was determined by inductively coupled plasma mass spectrometry (ICP-MS) using Agilent 7700e instrument. Size measurements were performed using dynamic light scattering (DLS) on a Malvern Zetasizer Nano ZS (Malvern, UK). Zeta potential was determined by the electrophoretic mobility and then applying the Henry equation on Malven Zetasizer Nano ZS (Malvern, UK).
  • Table 3 shows dynamic light scattering (DLS) data for average particle size of FMSN-PEG/PEI nanoparticles in different solutions.
  • FIG. 10A shows the conjugation of FMSN-PEG/PEI nanoparticles
  • FIG. 10B shows the TEM images of FMSN-PEG/PEI nanoparticles.
  • the TEM images show that these FMSN-PEG/PEI particles possess well-ordered mesoporous structure with an average particle size of about 60-70 nm. DLS-determined size indicates very little aggregation in biological solutions (Table 3).
  • FMSN-PEG/PEI 20 mg was dispersed in 2.5 mL of PBS buffer, and then 6.8 mg of NHS-PEG-MAL(3.4k) was dissolved in 2.5 mL of PBS and then added to FMSN-PEG/PEI solution. The solution was stirred for 2 hours at room temperature.
  • Thiolated Na,Na-Bis(carboxymethyl)-L-lysine hydrate (BCLH) solution was prepared by added 400 ⁇ L of Traut's reagent (100 ⁇ M) and 5.24 mg of Na,Na-Bis(carboxymethyl)-L-lysine hydrate in 5 mL of PBS buffer and stirred for 30 mins.
  • the lysate of E.coli containing His-TAT-SOD or His-TAT-GPx was mixed with FMSN-NTA-Ni at 4° C. overnight. Based on the metal affinity between the Ni (II) and His-tag protein offered a tight linkage with a very low dissociation constant, the FMSN-NTA-Ni was directly mixed with TAT-SOD or TAT-GPx proteins from the supernatants of pellets of E. coli crude lysates under 8M urea without purifying. The protein-conjugated particles were isolated by centrifugation and washed by ethanol. The protein-functionalized particles were denoted as FMSN-TAT-SOD or FMSN-TAT-GPx.
  • samples were prepared in 300 ⁇ L and monitored using a microplate reader (Bio Tek, SynergyTM H1). Firstly, a stock of cocktail reagents contained EDTA (10 ⁇ 4 M), cytochrome c (10 ⁇ 5 M), and xanthine (5 ⁇ 10 ⁇ 5 M) in 1 mL of 50 mM K 3 PO 4 was prepared. Then, 280 ⁇ L of cocktail reagent was added with various samples, xanthine oxidase (10 ⁇ L of 58 mU/mL) and completed with D.I. water up to 300 ⁇ L total volume. Finally, 200 ⁇ L of each sample was transferred to microplate reader and the absorbance at 550 nm was detection.
  • cocktail reagents contained EDTA (10 ⁇ 4 M), cytochrome c (10 ⁇ 5 M), and xanthine (5 ⁇ 10 ⁇ 5 M) in 1 mL of 50 mM K 3 PO 4 was prepared. Then, 280 ⁇ L of cocktail reagent
  • SOD specific activity was expressed as unit per milligram (U/mg) of total lysate proteins (The Journal of Biological Chemistry, 1969, 244, 6049-6055.).
  • GPx activity in HeLa cell was measured using the Glutathione Peroxidase Assay Kit (Cayman Chemical), based on the method of Paglia and Valentine, with hydrogen peroxide as substrate.
  • the method was based on an NADPH-coupled reaction, whereby GPx reduces hydrogen peroxide while oxidizing GSH to GSSG.
  • the generated GSSG is reduced to GSH with consumption of NADPH by GR.
  • Enzyme activity was measured at 340 nm and expressed in units representing oxidation of 1 ⁇ mole NADPH per minute per mL sample.
  • GPX specific activity was expressed as unit per milligram (U/mg) of protein.
  • Cell Viability Assay 3 ⁇ 10 4 cells per well were seeded in 24-well plates for proliferation assays. After incubation with different amounts of nanoparticles suspended in serum-free medium for 4 h, respectively, then the 500 ⁇ M N, N′-dimethyl-4, 4′-bipyridinium dichloride (paraquat) was added to the culture medium for 24 h. Particle-treated cells were then washed twice with PBS and incubated with 200 ⁇ L WST-1 (10%) in DMEM. Cells viability was estimated by a formazan dye generated by the live cells and the absorbance at 450 nm was measured using a microplate reader (Bio-Rad, model 680).
  • FIGS. 11A-C show the protection effects of co-delivery of TAT-SOD and TAT-GPx into Hela cells.
  • FIG. 11A shows the enhanced cell viability results for various nanoparticles by using WST-1 assay.
  • FIG. 3B shows the results of ROS detection for various nanoparticles. The levels of ROS were stained by DHE assays and quantified by flow cytometry.
  • FIG. 11C shows the results of Western blotting assays to show the levels of COX II and p-p38.
  • the concentration of PQ and co-delivery of FMSN-TAT-SOD and FMSN-TAT-GPx (1:1 ratio) are 500 ⁇ M and 25 ⁇ g/mL, respectively.
  • the denatured TAT-SOD or TAT-GPx fusion protein can be co-delivered into Hela cells and the denatured fusion proteins can be refolded and exhibit the specific enzymatic activities after delivering into the cells.
  • the TAT-SOD or TAT-GPx fusion protein functionalized FMSN named as FMSN-TAT-SOD or FMSN-TAT-GPx, still has the enzymatic activity by the refolding mechanism of the cells.
  • the mesoporous carriers of the present disclosure embodiments can deliver peptides, proteins, enzymes or enzymatic mimetics into the cells as needed and the native activities of the peptides, proteins, enzymes or enzymatic mimetics being delivered into the cell are maintained.
  • the mesoporous carriers can function as nanoreactors located within the cells and the delivered peptides, proteins, enzymes or enzymatic mimetics can work together to provide multiple functions.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Hematology (AREA)
  • Inorganic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Nanotechnology (AREA)
  • Urology & Nephrology (AREA)
  • Microbiology (AREA)
  • Biomedical Technology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Ceramic Engineering (AREA)
  • Mycology (AREA)
  • Cell Biology (AREA)
  • Pathology (AREA)
  • Biochemistry (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biotechnology (AREA)
  • General Physics & Mathematics (AREA)
  • Diabetes (AREA)
  • Obesity (AREA)
  • Medicinal Preparation (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Immobilizing And Processing Of Enzymes And Microorganisms (AREA)

Abstract

Mesoporous carriers for delivering targets into a cell are provided. The mesoporous carriers comprise hollow silica nanospheres (HSN) or mesoporous silica nanoparticles (MSN) and the targets bound to or encapsulated by the hollow silica nanospheres or the mesoporous silica nanoparticles. The targets includes at least two different targets, and the targets may include peptides, proteins, enzymes and/or enzymatic mimetics.

Description

    CROSS-REFERENCE TO RELATED APPLICATION
  • This application claims the priority benefits of U.S. provisional application Ser. No. 62/034,181, filed on Aug. 7, 2014, U.S. provisional application Ser. No. 62/034,192, filed on Aug. 7, 2014, and U.S. provisional application Ser. No. 62/034,282, filed on Aug. 7, 2014. The entirety of each of the above-mentioned patent applications is hereby incorporated by reference herein and made a part of this specification.
  • BACKGROUND OF THE INVENTION
  • 1. Field of the Invention
  • The present invention generally relates to a mesoporous carrier, in particular, to silica-based mesoporous carriers and the delivery methods by using the silica-based mesoporous carriers.
  • 2. Description of Related Art
  • Classified by the pore sizes, porous materials can be classified as microporous materials having pore sizes of less than 2 nm, mesoporous materials having pore sizes of 2-50 nm and macroporous materials having pore sizes of greater than 50 nm. The size of the pores and the large surface area of the pores allow the mesoporous materials to be ideal vehicles for carrying chemicals or drugs.
  • SUMMARY OF THE INVENTION
  • According to embodiments of the present invention, mesoporous carriers for delivering targets into a cell are provided. The mesoporous carriers comprise hollow silica nanospheres (HSN) or mesoporous silica nanoparticles (MSN) and the targets bound to or encapsulated by the hollow silica nanospheres or the mesoporous silica nanoparticles. The targets includes first targets and second targets, and the first and second targets are different. The targets may include peptides, proteins, enzymes and/or enzymatic mimetics.
  • According to embodiments of the present invention, a method of delivering targets into a cell is proposed. At first, mesoporous carriers are prepared and provided. The mesoporous carriers comprises hollow silica nanospheres (HSN) or mesoporous silica nanoparticles (MSN) and the targets bound to or encapsulated by the hollow silica nanospheres or the mesoporous silica nanoparticles. The targets include first targets and second targets, and the first and second targets are different. Then the mesoporous carriers contact with the cell by incubating the cell with the mesoporous carriers. The first targets and the second targets are co-delivered into the cell at the same time, as the mesoporous carriers and the targets carried by the hollow silica nanospheres or the mesoporous silica nanoparticles enter into the cell.
  • In order to make the aforementioned and other features and advantages of the invention more comprehensible, several embodiments accompanied with figures are described in detail below.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The accompanying drawings are included to provide a further understanding of the invention, and are incorporated in and constitute a part of this specification. The drawings illustrate embodiments of the invention and, together with the description, serve to explain the principles of the invention.
  • FIGS. 1A-1B show the reaction schemes for the PEI-modification of the enzymes SOD and CAT and the encapsulation of PEI-SOD and PEI-CAT within HSN.
  • FIG. 2 shows the adsorption and desorption isotherms of HSN according to one embodiment of the present invention.
  • FIGS. 3A-3D show transmission electron microscopy (TEM) images of various enzyme encapsulated HSN.
  • FIG. 4A shows the relative enzyme activity for different enzyme-encapsulated HSN.
  • FIG. 4B shows the fluorescence intensity over the wavelength for different enzyme-encapsulated HSN.
  • FIG. 5 shows the cell viability results using WST-1 assay when exposed to HSN or PEI-SOD/CAT@HSN.
  • FIG. 6 shows the quantification of fluorescence intensity of oxidized DHE from HeLa cells were treated with various enzyme encapsulated HSN.
  • FIG. 7 describes the reaction scheme for the conjugation of NF-κB p65 antibody and Cys-TAT peptide to the surface functionalized MSN.
  • FIGS. 8A-8D show transmission electron microscopy (TEM) images of various functionalized MSN nanoparticles.
  • FIG. 9A-9C show the results of in vitro pull-down assay of various functionalized MSN nanoparticles.
  • FIG. 10A shows the conjugation of FMSN-PEG/PEI nanoparticles.
  • FIG. 10B shows the TEM images of FMSN-PEG/PEI nanoparticles.
  • FIGS. 11A-11C show the protection effects of co-delivery of TAT-SOD and TAT-GPx into Hela cells.
  • DESCRIPTION OF THE EMBODIMENTS
  • Reference will now be made in detail to the present preferred embodiments of the invention, examples of which are illustrated in the accompanying drawings. Wherever possible, the same reference numbers are used in the drawings and the description to refer to the same or like parts. In the following embodiments, one or more silica-based mesoporous carrier materials are described. In the embodiments, the silica-based mesoporous materials may be classified as hollow silica nanospheres (HSN) and mesoporous silica nanoparticles (MSN) and the targets may be carried on the surface of the silica-based mesoporous materials or be encapsulated within the silica-based mesoporous materials. The cited examples, the ingredients, the reaction conditions or parameters illustrated in the examples are merely for illustration purposes, but it is not intended to limit the material or the preparation method by the exemplary embodiments described herein.
  • The target suitable for being carried by the silica-based mesoporous materials may include an enzymes containing cysteine (thiol group), lysine (amino group), aspartate or glutamate (carboxyl group), a peptide containing cysteine (thiol group), lysine (amino group), aspartate or glutamate (carboxyl group) or an antibody containing cysteine (thiol group), lysine (amino group), aspartate or glutamate (carboxyl group). Alternatively, the target suitable for being carried by the silica-based mesoporous materials may include an enzymes containing polyhistidine-tag (His-tag), a peptide containing polyhistidine-tag or an antibody containing polyhistidine-tag. Herein, the polyhistidine-tag consists of at least six histidine (His) residues.
  • In one embodiment, hollow silica nanospheres (HSN) with porous silica shells and large interior spaces (cavities) have been synthesized, which is suitable for loading enzymes within the cavities of HSN for intracellular catalysis. A microemulsion method has been developed for synthesizing hollow silica nanospheres (HSN), of which one or more enzymes may be loaded within the cavities. The enzymes may be antioxidant enzymes, including horseradish peroxidase (HRP), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase, glutathione reductase and their enzymatic mimetics. Alternatively, the enzymes may be enzymes involved in biochemical enzymatic cascades, which refers to a series of biochemical reactions involving enzymes, such as blood coagulation, metabolism pathways, and signal transduction pathways. HSN may be synthesized by silica sol-gel process of water-in-oil microemulsion, and polyethyleneimine (PEI) modified enzymes in aqueous phase are then encapsulated inside HSN. It has been demonstrated that encapsulation of HRP in the cavities of HSN (HRP@HSN) is feasible and the intracellular delivery of HRP@HSN showing its function as a catalytic nanoreactor inside Hela cells for converting prodrug into toxic agent to kill the cancer cells.
  • In one embodiment, PEI grafted superoxide dismutase (PEI-SOD) and catalase (PEI-CAT) are prepared and then co-encapsulated within HSN as the loaded enzymes.
  • Synthesis of Enzyme-Encapsulated HSN
  • Herein, polyethyleneimine (PEI) is introduced to improve the enzyme loading. PEI, with rich amine groups, has been demonstrated to be good proton sponge for endosome escape effect. FIGS. 1A-1B describe the reaction schemes for the PEI-modification of the enzymes (SOD and CAT) and the encapsulation of the positively charged PEI-SOD and PEI-CAT within HSN. The silica shell of HSN shields off PEI from contacting with cellular machineries, but the pores of the silica shells allow protons to diffuse inside the hollow spheres of HSN and keep the proton sponge effects of PEI.
  • In FIG. 1A, the PEI-enzyme conjugation is achieved through the amidation of the carboxyl group with PEI. FIG. 1B illustrates the synthesis of PEI-SOD and PEI-CAT co-encapsulated HSN (denoted as PEI-SOD/CAT@HSN).
  • Both SOD and CAT are important antioxidant enzymes in living systems. PEI is covalently linked to the enzymes by the conventional EDC/NHS coupling reaction. Subsequently, the PEI-grafted enzymes (PEI-SOD, PEI-CAT), either individually or together, are encapsulated inside HSN by an one-pot water-in-oil (w/o) microemulsion approach as shown in FIG. 1B. In brief, an aqueous solution containing PEI-grafted enzyme is emulsified in an oil system containing surfactant (isooctylphenyl ether, CA-520), co-surfactant (n-hexanol) and organic solvent (decane). After introducing the silica sources, tetraethoxysilane (TEOS) and aminopropyltrimethoxy silane (APTMS), and aqueous ammonia, the water-in-oil (w/o) mixture was stirred for 10 h at 20° C. to form enzyme-encapsulated silica nanoparticles. Enzymes-loaded particles were foiiiied after the nanoparticles were further suspended in waiin deionized water (40° C.) for 40 min and washed with water, which is critical for the transformation to hollow nanospheres.
  • Modification of SOD and CAT with PEI: PEI-conjugation of enzymes was carried using NHS/EDC crosslinking reaction. Briefly, 3 mg of SOD and CAT were dissolved in 50 mM sodium phosphate buffer (pH 7.8) at a concentration of 3 mg/mL. Amine-reactive NHS esters and EDC·HCl were prepared in 200 μL sodium phosphate buffer (50 mM, pH 7.8) and then added to the enzyme solution. The SOD solution contained 17 mg NHS/14 mg EDC·HCl and CAT solution contained 15 mg NHS/13 mg EDC·HCl. After stirring for 1 h at 4° C., 43 and 38 μL of PEI reagent were added to the SOD and CAT solutions separately and the mixtures were further stirred at 4° C. for 24 h to complete the reactions. Finally, the PEI-conjugated SOD (PEI-SOD) and CAT (PEI-CAT) were purified and concentrated with Amicon Ultra filters (MA, USA).
  • Synthesis of PEI-SOD@HSN, PEI-CAT@HSN, and PEI-SOD/CAT@HSN: Hollow silica nanospheres (HSN) were synthesized by a reverse microemulsion method. Typically, 20 mL of decane, 1.63 mL of CA-520, 550 μL of n-hexanol and 350 μL of H2O with concentrated PEI-enzymes (PEI-SOD, PEI-CAT, or a mixture of PEI-SOD and PEI-CAT) were mixed at room temperature to generate the water-in-oil microemulsion. Then, silica sources (100 μL of TEOS and 25 μL of ethanolic APTMS solution) were added with stirring. The ethanolic APTMS solution was prepared by adding 200 mL of APTMS to 1.4 mL of absolute ethanol. After 10 min, 250 μL of aqueous ammonia (35 wt %) was introduced to the mixture which was stirred for 10 h at 20° C. Then 95% ethanol was added to destabilize the microemulsion system and solid products were centrifuged at 11000 rpm for 30 min. To obtain the hollow structure, the samples were suspended in 40 mL warm water (40° C.), stirred for 40 minutes, and isolated by centrifugation to obtain PEI-SOD encapsulated HSN (denoted as PEI-SOD@HSN), PEI-CAT encapsulated HSN (denoted as PEI-CAT@HSN), PEI-SOD/CAT@HSN or HSN only (without adding enzymes). Finally, all nanoparticles were washed several times with ethanol and deionized water to extract the surfactants in the pores. The amount of PEI-enzymes used to prepare enzyme-loaded HSN: 36 nmole of PEI-SOD for PEI-SOD@HSN; 4 nmole of PEI-CAT for PEI-CAT@HSN; 36 nmole of PEI-SOD and 4 nmole of PEI-CAT for PEI-SOD/CAT@HSN.
  • Physicochemical Characterization.
  • The surface area of HSN (231.2 m2/g) as determined by N2 adsorption—desorption isotherms exhibited characteristic type IV BET isotherms (FIG. 1). The high surface area is attributed to the porous structure on the shell, indicating most of the surfactants have been removed. FIGS. 3A-3D show transmission electron microscopy (TEM) images of enzyme encapsulated silica nanospheres; FIG. 3A: PEI-SOD/CAT@HSN, FIG. 3B: PEI-SOD/CAT@HSN stained with uranyl acetate (UA); FIG. 3C: PEI-SOD@HSN and FIG. 3D: PEI-CAT@HSN. All particles show a narrow size distribution, with an average diameter of about 40 nm. Uranyl acetate (UA) staining displayed an enhanced electron density inside PEI-SOD/CAT@HSN but no staining outside the particles (FIG. 3B). This indicates the enzymes were indeed entrapped within the hollow spheres.
  • The enzyme-encapsulation efficiency and loading yields have been examined and the results are listed in Table 1. Significantly higher amount of enzymes are encapsulated in HSN with the cationic polymer PEI as shown in Table 1.
  • TABLE 1
    SOD-Loading CAT-Loading
    yield yield
    SOD-encapsulation CAT-encapsulation (μg (μg
    efficiency efficiency enzyme/mg enzyme/mg
    Sample (%) (%) particles) particles)
    SOD@HSN  2.8% (±0.2%)
    CAT@HSN 2.8% (±0.4%)
    PEI-SOD@HSN 12.2% (±3.1%) 44.7 (±8.1)
    PEI-CAT@HSN 8.7% (±2.3%) 29.5 (±4.6)
    PEI-SOD/CAT@HSN 12.3% (±3.4%) 9.4% (±0.8%) 44.5 (±5.7) 26.1 (±3.3)
  • The loading efficiency of PEI-SOD (12.2%) and PEI-CAT (8.7%) were much higher than that of SOD (2.8%) and CAT (2.8%). This is due to the positively charged PEI-SOD and PEI-CAT are effective in attracting the negatively charged hydrolyzed silica precursors for encapsulation. The higher loading will improve the enzyme activity for biomedical use and thus PEI-coated enzymes are chosen for further study thereafter. The loading yields of PEI-SOD@HSN and PEI-CAT@HSN were 44.7 and 29.5 μg enzymes/mg HSN. Compared with single enzyme loading, about the same level of efficiency and loading yields were observed when two enzymes, PEI-SOD and PEI-CAT, were co-encapsulated in HSN. The loading efficiency of PEI-SOD and PEI-CAT were 12.3% and 9.4%. PEI-SOD/CAT@HSN contains about 44.5 and 26.1 μg enzymes/mg HSN (Table 1).
  • Enzymatic Activity Assays
  • In the case of SOD, samples were prepared in 300 μL and monitored using a microplate reader (Bio Tek, Synergy™ H1). First, a stock of cocktail reagent contained EDTA (10-4 M), cytochrome c (10-5 M), and xanthine (5×10-5 M) in 1 mL of 50 mM K3PO4 was prepared. Then, 280 μL of cocktail reagent was added with various samples (native SOD, PEI-SOD/CAT@HSN, PEI-SOD@HSN, PEI-CAT@HSN or HSN). Then, xanthine oxidase (10 μL of 58 mU/mL) was added to the above solution completing with D.I. water up to 300 μL total volume. Finally, 200 μL of each sample was transferred to microplate reader and the absorbance at 550 nm was determined. To measure the SOD activity, the inhibition rate of cytochrome c reduction between native SOD (3000 U/mg) and SOD loaded particles (PEI-SOD/CAT@HSN and PEI-SOD@HSN) were compared using the slopes of absorbance between t=0 sec and t=180 sec.
  • In the case of CAT assay, 90 μL of H2O2 solution (5 μM in 50 mM sodium phosphate buffer; pH 7.8) and various nanoparticles solutions (native CAT, PEI-SOD/CAT@HSN, PEI-CAT@HSN, PEI-SOD@HSN or HSN) were mixed and completed with D.I. water up to 100 μL total volume. After incubation for 30 min, HRP (100 μL of 2 U/mL) and Amplex red (10 μL of 0.1 mM) were added. Each sample (200 μL) was transferred to a microplate reader and the absorbance was monitored at 570 nm. To determine the CAT activity, we calculated the ΔOD570 of each sample between t=0 min and t=30 min and compared with native CAT. Table 2 shows the results of relative activity of SOD and CAT after encapsulated inside HSN.
  • TABLE 2
    Relative activity Relative activity
    Sample (SOD) (CAT)
    Native SOD 100
    PEI-SOD@HSN 18.6 (±13.4)
    Native CAT 100
    PEI-CAT@HSN 62.7 (±29.2)
    PEI-SOD/CAT@HSN 46.6 (±5.8)  57.9 (±30.5)
  • Compared with the inhibition percentage of cytochrome c reduction between native SOD and SOD-loaded particles, the encapsulated PEI-SOD/CAT@HSN and PEI-SOD@HSN maintain 46.6% and 18.6% of the native SOD activity (Table 2). Also, as seen in Table 2, when compared with the reaction rate of H2O2 decomposition between native CAT and CAT-loaded particles, PEI-SOD/CAT@HSN and CAT@HSN keep 57.9% and 62.7% of the native CAT activity.
  • The synergism between SOD and CAT in the presence of O2 •− was investigated. FIG. 4A shows the stability of CAT-loaded particles and native CAT after treated with KO2 solution. As shown in FIG. 4A, after exposure to KO2 solution, which generates 02 •−, native CAT lost almost all its activity, whereas CAT activity of PEI-SOD/CAT@HSN and PEI-CAT@HSN still remained at about 80% and 70%.
  • Cascade reactions in PEI-SOD/CAT@HSN
  • SOD and CAT are antioxidant enzymes and work together as primary defence system against free radical damage in living cells. In aforementioned study, we had demonstrated that PEI-SOD and PEI-CAT could be simultaneously encapsulated in HSN and the final nanoparticle system showed activities in both enzymes. In order to investigate whether both the encapsulated enzymes are involved in cascade reactions, fluorescence assay was performed in which the resorufin formation was recorded by monitoring the fluorescence at 570 nm. In the reaction, the O2 •− generated by KO2 was unstable and converted to H2O2 in the presence or absence of SOD in aqueous solution. However, the dismutation rate of O2 •− in the presence of SOD is faster. Subsequently H2O2 could be either reduced by CAT or further involved in peroxidase/Amplex red oxidation reaction. As shown in FIG. 4(B), the fluorescence intensity of PEI-SOD@HSN was significantly higher than that of blank (control), HSN (negative control), and PEI-SOD/CAT@HSN. The results indicated PEI-SOD@HSN catalyzed the reduction of O2 •− to hydrogen peroxide and increase the rate of resorufin formation. However, in the presence of PEI-SOD/CAT@HSN, the cascade reaction occurs. Hydrogen peroxide produced by SOD could be further decomposed by CAT and the system showed weaker fluorescence. Relative enzyme activity was expressed as a ratio of change in absorption at 570 nm before and after KO2 treatment. FIG. 4B shows the cascade reactions within PEI-SOD/CAT@HSN. The fluorescence intensity reflects the amount of H2O2 in the assay buffer. Herein, the same SOD units of PEI-SOD/CAT@HSN and PEI-SOD@HSN were used for comparison.
  • Cell proliferation assay: 2×104 cells were seeded in 24-well plates and allowed to attach for 24 h. To determine the particle toxicity, cells were incubated in fresh serum-free medium containing different amounts of BSA-stabilized particles (0-400 μg/mL) for 2.5 h. After washing twice with phosphate-buffered saline (PBS), particle-treated cells were cultured in regular growth medium. After 24 h, cells were washed twice with PBS and incubated with 200 μL WST-1 (10%) in Dulbecco's modified eagles medium (DMEM). Cell viability was determined by the fornazan dye generated by the live cells and the absorbance at 450 nm was measured using a microplate reader (Bio-Rad, model 680).
  • Cell Uptake, Distribution Pattern, and Cytotoxicity of PEI-Enzyme Loaded HSN
  • From the experimental observations, nanoparticles, HSN and PEI-SOD/CAT@HSN are well-internalized into cells, and most of the nanoparticles have escaped from endosome and resided in the cytoplasm. The cell uptake of the nanoparticles, at the treated doses of 50, 100 and 200 μg/mL for 2.5 h, leads to remarkably high cellular uptake of PEI-SOD/CAT@HSN and a dose-dependent uptake behaviour of nanoparticles by HeLa cells. FIG. 5 shows the cell viability results for Hela cells exposed to HSN or PEI-SOD/CAT@HSN using WST-1 assay. More importantly, no significant cytotoxicity of HSN and PEI-SOD@HSN are found in cell proliferation assay while the doses of particles were up to 400 μg/mL (FIG. 5). Cell viability was examined using WST-1 assay at doses of 25, 50, 100, 200, and 400 μg/mL, after 2.5 h of particle treatment and followed by additional 24 h growth.
  • Protective Effect of Enzyme-Loaded Particles in Cells after Paraquat (PQ) Treatment
  • It has been demonstrated that tandem reaction occurs within PEI-SOD/CAT@HSN in test tube experiments. The nanoparticles exhibited excellent synergism effect between SOD and CAT. They also displayed high bio-compatibility. These advantages provided opportunities for their uses in protein therapy. The capability of enzyme-loaded particles in living cells for removing paraquat (PQ)-induced ROS is evaluated. ROS production is measured by flow cytometry with dihydroethidium (DHE), a superoxide indicator with fluorescence upon oxidation, to evaluate the antioxidant effect of each kind of nanoparticle. HeLa cells were treated with various nanoparticles at a dose of 100 μg/mL for 2.5 h. After washing twice with PBS, cells were exposed to 500 μM of paraquat (PQ) for 18 h and then stained with 2 μM of dihydroethidine (DHE) and the oxidized DHE with fluorescence was determined by flow cytometry analysis. FIG. 6 shows the quantification of fluorescence intensity of oxidized DHE from HeLa cells were treated with various nanoparticles. The PQ-induced ROS productions are the same for the un-protected cells (positive control and HSN only). All enzyme-nanoparticle treated cells obviously inhibit PQ-induced ROS production. PEI-SOD@HSN and PEI-CAT@HSN show similar levels of reduction of DHE-positive cells by about 25% (compared to HSN only). However, co-loaded PEI-SOD/CAT@HSN shows a much higher level of reduction of DHE-positive cells by about 60%. The PEI-SOD/CAT@HSN nanosystem clearly shows a synergetic effect in ROS reduction. DHE-positive cells and fluorescence intensity quantitated indicates that PEI-SOD/CAT@HSN having the synergetic effect of both antioxidant enzymes displays the weakest fluorescence intensity under microscopic observation. The enzyme-loaded particles, especially PEI-SOD/CAT@HSN, show better cell attaching morphology, which implies the treatment by PEI-SOD/CAT@HSN detoxify the imposed ROS. The nanoparticle-treated cells after incubation with PQ were further assayed with Western blotting to check the level of p-p38 and COX2 expression. Down-regulation of PQ-induced activation of p-p38 and COX2 is observed for cells pre-treated with PEI-SOD/CAT@HSN, PEI-SOD@HSN and PEI-CAT@HSN.
  • In one aspect, the present invention provides a mesoporous hollow silica nanosphere (HSN) materials (about 40 nm size) based on templating water-in-oil with sol-gel condensation. The enzyme SOD and/or catalase (CAT) can be individually or jointly encapsulated within HSN, with the help of the polyamine PEI. The mesopores on the silica shell allow easy access of small molecules while keeping the enzymes inside the nanosphere from undesirable protein-protein interaction. We found excellent catalytic activities of superoxide dismutation by SOD and then transformation of H2O2 to water by catalase encapsulated in HSN. When PEI-SOD and PEI-CAT are co-encapsulated, PEI-SOD/CAT@HSN can complete cascade transformation of superoxide through hydrogen peroxide to water with synergism.
  • After delivery to cells, substantial fractions of HSN exhibited endosome escape to cytosol. Downstream reactive oxygen species (ROS) production and COX-2/p-p38 expressions show that co-encapsulated SOD/CAT inside HSN give the highest cell protection against the toxicants. The rapid cell-uptake and strong detoxification effect on superoxide radicals by the SOD/CAT-encapsulated hollow mesoporous silica nanoparticles demonstrate that the enzyme-encapsulated hollow mesoporous silica nanoparticles can function as a catalytic nanoreactor after cell uptake. PEI-SOD/CAT@HSN may be applied as antioxidants for medical treatments, such as inflammation, ischemia, stroke and other strong oxidative stress situation.
  • However, the sizes of many proteins or enzymes are significantly bigger than the inner space of HSN or MSN or certain proteins are not suitable to be encapsulated within the nanomaterials and the loading efficiency is limited. In this case, it is feasible to link or conjugate the enzymes or proteins to the exterior or outer surface of the nanoparticles or nanomaterials for better delivery efficiency.
  • Alternatively, in another embodiment, mesoporous silica nanoparticle (MSN) materials are synthesized and functionalized to carry peptides and/or antibodies. The peptide may be any peptide containing cysteine or a polyhistidine tag, including nucleus localization sequence (NLS)-peptide, cancer-targeting peptide and lysosomal targeting peptide. The antibody may be any antibody containing cysteine or a polyhistidine tag, including signal transduction antibody and cancer-targeting antibody.
  • The present invention provides nanoparticles consisting of mesoporous silica nanoparticle (MSN) with surface functionalization of NF-κB (nuclear factor-kappa B) p65 antibody and TAT transducing peptide (i.e., HIV trans-activator of transcription (TAT) protein transduction domain). The sequence of TAT transducing peptide: CGRKKRRQRRR. These nanoparticles can move near nuclear membrane and block nuclear translocation of the activated p65.
  • FIG. 7 describes the reaction scheme for the conjugation of NF-κB p65 antibody and Cys-TAT peptide to the surface functionalized MSN. To synthesize the functionalized MSN, amine groups are formed on the surface of MSN by reacting with 3-aminopropyltrimethoxysilane (APTMS) to foim MSN-APTMS with an average loading of nitrogen content of APTMS at 2.6 wt % by elemental analysis. In order to immobilize p65 antibody on MSN, we chose two polyethylene glycol (PEG) crosslinkers with different lengths, MAL-PEG2k-SCM and MAL-PEG3.4k-SCM, to MSN-APTMS. Herein, the abbreviations are explained, MAL: maleimide, PEG2k or PEG3.4k: polyethylene glycol (PEG) having an average molecular weight of 2000 or 3400, SCM: succinimidyl carboxymethyl. The MAL-PEG-SCM crosslinkers containing a succinimidyl moiety react with the amine groups of MSN-APTMS through an active succinimidyl link to obtain the MSN-PEGs (MSN-PEGs: MSN-PEG2k, MSN-PEG34k). The MAL-end of MSN-PEG reacted with the thiol groups of the antibody and Cys-TAT peptide.
  • Synthesis of Green Fluorescent Mesoporous Silica Nanoparticles (MSN) C16TABr (0.58 g, 1.64×10−3 mole) and 5 mL of 0.226 M ethanol solution of tetraethoxysilane (TEOS, 1 mL TEOS in 20 mL 99.5% ethanol) were dissolved in 300 g of 0.17 M aqueous ammonia solution. The stock solution was stirred at 40° C. for 5 h. 5 mL of 1.13 M ethanol solution of TEOS (5 mL of TEOS in 20 mL 99.5% ethanol) and FITC-APTMS were added with vigorous stirring for 1 h and then aged statically at 40° C. for 24 h. FITC-APTMS, N-1-(3-trimethoxysilyl propyl)-N′-fluoreceylthiourea), was prepared in advance by stirring fluorescein isothiocyanate (FITC, 1 mg) and 3-aminopropyltrimethoxysilane (APTMS, 100 μL) in 5 mL ethanol (99%) at room temperature for 24 h. As synthesized samples were then collected by centrifugation with 12000 rpm for 20 min and washed five times with 99% ethanol. 200 mg of as-synthesized samples were redispersed in 25 mL of 95% ethanol with 0.5 g of 37 wt % HCl. Surfactant was extracted by heating the ethanol suspension at 60° C. for 24 h. The product, called FITC-MSN (MSN), was collected by centrifugation and washed with ethanol several times and stored in ethanol.
  • Preparation of Amine-Functionalized MSN (MSN-APTMS)
  • The surface of MSN was functionalized with amine groups by treatment with APTMS. MSNs (200 mg) were first dispersed in 50 mL of ethanol, and then the solution was refluxed for 18 h after the addition of 500 μL of APTMS. After centrifugation and washing with ethanol, amine-functionalized MSNs were redispersed in ethanol. To remove the surfactants, the amine-functionalized MSNs were suspended in acidic ethanol (1 g of HCl in 50 mL of EtOH) and refluxed for 24 h. After centrifugation and washing with ethanol, amine-functionalized MSN (MSN-APTMS) were redispersed in ethanol.
  • Transmission Electron Microscopy (TEM)
  • TEM images were taken using a Hitachi H-7100 instrument with an operating voltage of 75 KV. Samples were sonicated to disperse in ethanol and 10 μL of the suspension was dropped to fix on a microgrid.
  • For evaluation of immunological efficiency, the p65 antibody was covalently immobilized with the MAL-end of MSN-PEG3.4k in different ratios (1:6, 1:12, 1:24) via C-S binding. After the p65 antibody conjugation, the Cys-TAT peptide was conjugated to fill up the free MAL-end of MSN-PEG3.4k. MSN-PEG3.4k without antibody coupling was also directly conjugated with Cys-TAT as a control. The physical properties of the nanoparticles were characterized by nitrogen adsorption-desorption isotherms, powder X-ray diffraction (XRD), FT-IR, TEM, dynamic light scattering (DLS) and zeta potential. FIGS. 8A-8D show transmission electron microscopy (TEM) images of various functionalized MSN. From the TEM images, it is shown that the MSN particles possess well-ordered mesoporous structures and the average particle size obtained from TEM images is about 40 nm.
  • Cell Viability and Growth Inhibition Assay.
  • The WST-1 assay was applied to measure the cell viability and growth inhibition assay: 2×104 HeLa cells per well were seeded in 24-well plates for 16 h for HeLa cell viability assay. HeLa cells were incubated in serum-free medium containing different amounts of MSN-PEG3.4k-Ab-TAT (100 μg/L) for 4 h. For HNSCC growth inhibition assay, HNSCC cells were seeded in 24-well plates with a density of 4×104 cells/well for 16 h and incubated with 200 μg/mL of MSN-PEG3.4k-Ab(1:24)-TAT, MSN-PEG3.4k-TAT or anti-TNF antibody (100 ng/mL) in serum-free medium for 4 h. Following medium replacement with culture medium, HNSCC cells were incubated for further 72 h. For WST-1 assay, HeLa or HNSCC cells were allowed to grow in culture medium containing WST-1 (Clontech) for 4 h at 37° C. The dark red formazan dye generated by the live cells was proportional to the number of live cells and the absorbance at 450 nm was measured using a microplate reader (Bio-Rad, model 680).
  • The cell viability of the MSN-PEG34k-Ab-TAT was examined by using WST-1 assay and MSN-PEG3.4k-Ab-TAT shows no significant cytotoxicity.
  • Western Blotting Analysis
  • Cell lysates were separated on a 10% SDS-PAGE, and the proteins were then electrophoretically transferred to a polyvinylidene difluoride (PVDF) membrane and blocked 1 h at room temperature in blocking buffer [1×Tris-buffered saline (TBS)-0.1 % Tween 20, 5% w/v nonfat dry milk]. Membranes were incubated overnight at 4° C. with primary antibodies: NF-κB p65, TNF-α, Lamin B and GAPDH from Santa Cruz Biotechnology (Santa Cruz, Calif.), along with COX-2 from Cayman (Cayman, Ann Arbor, Mich., USA). All primary antibodies were diluted in blocking buffer (NF-κB p65: 1:500, TNF-α: 1:500, Lamin B: 1:3500, GAPDH: 1:5000 and COX-2: 1:500 dilution). The PVDF membranes were extensively washed and incubated with a horseradish peroxidase—conjugated secondary immunoglobulin G antibody (1: 2000 dilution, Santa Cruz Biotechnology) for 1 h at room temperature. Immunoreactive bands were visualized with an enhanced chemiluminescence substrate kit (Amersham Pharmacia Biotech, GE Healthcare UK Ltd, Bucks, UK) according to the manufacturer's protocol.
  • Cellular Response of NF-κB on MSN-PEGs and in vitro Pull-Down Assay of MSN-PEG3.4k-Ab-TAT
  • 100 μg/mL of MSN-APTMS, MSN-PEG2k and MSN-PEG3.4k were treated in HeLa cells for 4 h, and then incubation without or with TNF-α (50 ng/mL), a NF-κB activator, for another 0.5 h. After the cells were harvested, cytosolic and nuclear protein were isolated, the p65 expression level in either cytosol or nucleus was determined by western blotting experiments. For in vitro pull-down assay, the MSN-PEG3.4k-Ab-TAT (100 μg/mL) was mixed and incubated with total lysate of HeLa cell at 4° C. for 18 h in vitro. Then, the mixture was centrifuged at 12,000 rpm for 20 mins and the supernatant (10 μL) was assayed for the free p65 expression level by Western blotting.
  • FIGS. 9A-9C show the results of in vitro pull-down assay of various functionalized MSN nanoparticles. MSN-PEG3.4k-Ab-TAT blocks NF-κB p65 nuclear translocation and thus inhibits the NF-κB p65 downstream protein expression. HeLa cells were treated with MSN-PEG3.4k-TAT or MSN-PEG3.4k-Ab-TAT for 4 h at different doses (100 μug/mL for FIG. 9B, 50-200 μg/mL for FIG. 9C). After the delivery, the cells were stimulated with or without 5Ong/mL TNF-α for another 0.5 h. Dose-dependence study of the blockage as shown in FIG. 9C indicates that nuclear p65 level decreases with the increasing concentration of MSN-PEG3.4k-Ab(1:24)-TAT.
  • As shown in FIG. 9B, western blotting was carried out to quantify the p65 level in HeLa cells. After the treatment, the cell lysates were harvested for the p65 level in nucleus and cytoplasm. The western blotting results indicated that MSN-PEG3.4k-Ab-TATs did not induce any nuclear translocation of p65 without TNF-α. However, under the TNF-α treatment, a significant increase of p65 level appeared in the nucleus in absence of MSN-PEG3.4k-Ab-TATs. Once the MSN-PEG34k-Ab-TATs with different amount of conjugated antibody were added, the level of nuclear p65 gradually reduced with increasing amount of antibody. Both MSN-PEG3.4k-Ab(1:12)-TAT and MSN-PEG3.4k-Ab(1:24)-TAT show obvious suppression of the p65 translocation to nucleus, whereas MSN-PEG3.4k-TAT did not prevent the TNF-α inducing nuclear p65 translocation. Hence, MSN-PEG3.4k-Ab-TAT shows the specificity and effectiveness to block NF-κB p65 nuclear translocation through immunogenic binding.
  • Herein, a nanoparticle/antibody complex targeting NF-κB is employed to catch the Rel protein p65 in perinuclear region and thus blocking the translocation near the nuclear pore gate. TAT peptide conjugated on mesoporous silica nanoparticles (MSN) help non-endocytosis cell-membrane transducing and converge toward perinuclear region, where the p65 specific antibody performed the targeting and catching against active NF-κB p65 effectively.
  • In another embodiment, a protein delivery system combining MSN nanoparticle carriers and one or more denatured fusion proteins has been developed. Such combination of the nanomaterial and one or more fusion proteins not only solves the problems of protein delivery, including chemical solvents, stability, and permeability, but also provide a new approach for protein therapy.
  • Herein, two antioxidant enzyme proteins with similar function for free radicals scavenging, superoxide dismutase (SOD) and glutathione peroxidase (GPx), are demonstrated as the co-delivered enzymes carried by the nanoparticles.
  • For TAT-SOD and TAT-GPX protein conjugation, we constructed and overexpressed the His-tag human Cu, Zn-superoxide dismutase (SOD) and human glutathione peroxidase (GPx) which contain a human immunodeficiency virus (HIV) transducing domain (TAT, residues 49-57). The sequence of TAT transducing peptide: RKKRRQRRR. The genes of TAT-SOD and TAT-GPx were cloned and inserted into prokaryotic protein expression vector of pQE-30 to form pQE-TAT-SOD and pQE-TAT-GPx. The vectors were transformed into JM109 E. coli and cultured in LB broth with IPTG protein induction for 1 and 3 h. The TAT-SOD and TAT-GPx with high protein overexpression were displayed in accordance with increasing induction time in 10% SDS-PAGE electrophoresis. Finally, the supernatants of pellets of E. coli crude lysates expressed TAT-SOD or TAT-GPx were tried to further directly conjugate in 8M urea.
  • Synthesis of Fluorescent Mesoporous Silica Nanoparticles (FMSN)
  • Dye-functionalized MSNs were synthesized by co-condensation process. FITC solution was prepared by dissolving 1 mg of FITC in 5 ml of anhydrous ethanol. 100 L of APTMS was added with rapid stirring at room temperature in darkness for 24 hours. 0.58 g of C16TAB was dissolved in 300 g of 0.17 M NH3 solution, and 5 mL of dilute TEOS solution (5% v/v TEOS/ethanol) was added with stirring for 5 h. FITC-APTMS solution added before 5 ml of concentrate TEOS solution (25% v/v TEOS/ethanol) was added dropwise with vigorous stirring for 1 h. The solution was then aged at 40° C. for 24 hours to complete the silica condensation. As-synthesized products was collected by centrifugation and washed with 95% ethanol three times. The products called FITC-MSN (FMSN) were stored in absolute ethanol.
  • Conjugation of NTA-Silane and Ni (II) with FMSN
  • 100 mg of FMSN were suspended in 50 mL of toluene containing 50 mg of NTA-silane and reflux for 18 h. The products were cleaned by ethanol to eliminate excess silane. In order to remove the C16TABr templates, the particles were dispersed in acidic solution (1 g of HCl in 50 mL ethanol) and stirred at 60° C. for 24 h. Subsequently, hydrolysis of methoxycarbonyl on NTA linker was achieved in the presence of aqueous p-TsOH (0.266 g, pH=2.0) under stirring at 65° C. for 6 h. After cleaned by ethanol, the particles were reacted with 50 mM NiCl2 aqueous solution for 6 h at room temperature. Followed the same cleaned procedure described above, the
  • FMSN-NTA-Ni were obtained and stored in absolute ethanol. FMSN-NTA-Ni with an average loading of Ni content is 0.6 wt % by ICP-MS analysis.
  • Synthesis of FMSN-PEG/PEI Nanoparticles
  • Dye-functionalized MSNs were synthesized by co-condensation process. FITC solution was prepared by dissolving 1 mg of FITC in 5 mL of anhydrous ethanol. 100 of APTMS was added with rapid stirring at room temperature in darkness for 24 hours. 0.58 g C16TAB was dissolved in 300 g of 0.17 M NH3 solution, and 5 mL of dilute TEOS solution (5% v/v TEOS/ethanol) was added with stirring for 5 h. FITC-APTMS solution added before 5 mL of concentrate TEOS solution (25% v/v TEOS/ethanol) was added dropwise with vigorous stirring for 1 h. 900 μL PEG-silane and 40 μL PEI-silane were added and stirring for 30 mins. The solution was then aged at 40° C. for 24 hours to complete the silica condensation. The solution was aged under hydrothermal condition at 90° C. for 24 hours and 70° C. for 24 hours. As-synthesized products was collected by centrifugation and washed with 95% ethanol. The particle was redispersed in 50 mL of 95% ethanol with 1 g of 37 wt % HCl for 1 hour and then the acid solvent was changed to 50 mL of 95% ethanol with 50 μL of 37 wt % HCl to remove the CTAB. FITC conjugated MSN (FMSN)-PEG/PEI particles were collected by centrifugation and washed with 95% ethanol three times.
  • Characterization
  • Transmission electron microscopy (TEM) images were taken on a JEOL JSM-1200 EX II operating at 120 kV. The nickel amount of sample was determined by inductively coupled plasma mass spectrometry (ICP-MS) using Agilent 7700e instrument. Size measurements were performed using dynamic light scattering (DLS) on a Malvern Zetasizer Nano ZS (Malvern, UK). Zeta potential was determined by the electrophoretic mobility and then applying the Henry equation on Malven Zetasizer Nano ZS (Malvern, UK). Table 3 shows dynamic light scattering (DLS) data for average particle size of FMSN-PEG/PEI nanoparticles in different solutions.
  • TABLE 3
    solvent Size (nm)
    H2O 65.04 ± 0.57
    PBS 63.03 ± 0.34
    DMEM 63.71 ± 0.80
    DMEM + FBS 69.41 ± 0.40
  • FIG. 10A shows the conjugation of FMSN-PEG/PEI nanoparticles, while FIG. 10B shows the TEM images of FMSN-PEG/PEI nanoparticles. The TEM images show that these FMSN-PEG/PEI particles possess well-ordered mesoporous structure with an average particle size of about 60-70 nm. DLS-determined size indicates very little aggregation in biological solutions (Table 3).
  • Conjugation of NTA and Ni (II) with FMSN-PEG/PEI
  • 20 mg of FMSN-PEG/PEI was dispersed in 2.5 mL of PBS buffer, and then 6.8 mg of NHS-PEG-MAL(3.4k) was dissolved in 2.5 mL of PBS and then added to FMSN-PEG/PEI solution. The solution was stirred for 2 hours at room temperature. Thiolated Na,Na-Bis(carboxymethyl)-L-lysine hydrate (BCLH) solution was prepared by added 400 μL of Traut's reagent (100 μM) and 5.24 mg of Na,Na-Bis(carboxymethyl)-L-lysine hydrate in 5 mL of PBS buffer and stirred for 30 mins. The thiolated BCLH solution was added to the FMSN-PEG/PEI solution and stirred overnight at 4° C. . Subsequently, hydrolysis of methoxycarbonyl on NTA linker was achieved in the presence of aqueous p-TsOH (0.133 g, pH=2) under stirring at 65° C. for 6 h. After washed by ethanol, the particles were reacted with 50 mM of NiCl2 aqueous for 6 h at room temperature. Followed the same washed procedure described above, the FMSN-PEG/PEI-NTA-Ni were obtained and stored in absolute ethanol.
  • Immobilization of His-TAT-Protein with FMSN-NTA-Ni
  • The lysate of E.coli containing His-TAT-SOD or His-TAT-GPx was mixed with FMSN-NTA-Ni at 4° C. overnight. Based on the metal affinity between the Ni (II) and His-tag protein offered a tight linkage with a very low dissociation constant, the FMSN-NTA-Ni was directly mixed with TAT-SOD or TAT-GPx proteins from the supernatants of pellets of E. coli crude lysates under 8M urea without purifying.The protein-conjugated particles were isolated by centrifugation and washed by ethanol. The protein-functionalized particles were denoted as FMSN-TAT-SOD or FMSN-TAT-GPx.
  • Determination of SOD and GPx Activity
  • In the case of SOD, samples were prepared in 300 μL and monitored using a microplate reader (Bio Tek, Synergy™ H1). Firstly, a stock of cocktail reagents contained EDTA (10−4 M), cytochrome c (10−5 M), and xanthine (5×10−5 M) in 1 mL of 50 mM K3PO4 was prepared. Then, 280 μL of cocktail reagent was added with various samples, xanthine oxidase (10 μL of 58 mU/mL) and completed with D.I. water up to 300 μL total volume. Finally, 200 μL of each sample was transferred to microplate reader and the absorbance at 550 nm was detection. To measure the SOD activity, the inhibition rate of cytochrome c reduction between native SOD and SOD samples were compared using the slopes of absorbance between t=0 sec and t=180 sec. SOD specific activity was expressed as unit per milligram (U/mg) of total lysate proteins (The Journal of Biological Chemistry, 1969, 244, 6049-6055.).
  • GPx activity in HeLa cell was measured using the Glutathione Peroxidase Assay Kit (Cayman Chemical), based on the method of Paglia and Valentine, with hydrogen peroxide as substrate. The method was based on an NADPH-coupled reaction, whereby GPx reduces hydrogen peroxide while oxidizing GSH to GSSG. The generated GSSG is reduced to GSH with consumption of NADPH by GR. Enzyme activity was measured at 340 nm and expressed in units representing oxidation of 1 μmole NADPH per minute per mL sample. GPX specific activity was expressed as unit per milligram (U/mg) of protein.
  • Cell Viability Assay: 3×104 cells per well were seeded in 24-well plates for proliferation assays. After incubation with different amounts of nanoparticles suspended in serum-free medium for 4 h, respectively, then the 500 μM N, N′-dimethyl-4, 4′-bipyridinium dichloride (paraquat) was added to the culture medium for 24 h. Particle-treated cells were then washed twice with PBS and incubated with 200 μL WST-1 (10%) in DMEM. Cells viability was estimated by a formazan dye generated by the live cells and the absorbance at 450 nm was measured using a microplate reader (Bio-Rad, model 680).
  • FIGS. 11A-C show the protection effects of co-delivery of TAT-SOD and TAT-GPx into Hela cells. FIG. 11A shows the enhanced cell viability results for various nanoparticles by using WST-1 assay. FIG. 3B shows the results of ROS detection for various nanoparticles. The levels of ROS were stained by DHE assays and quantified by flow cytometry. FIG. 11C shows the results of Western blotting assays to show the levels of COX II and p-p38. The concentration of PQ and co-delivery of FMSN-TAT-SOD and FMSN-TAT-GPx (1:1 ratio) are 500 μM and 25 μg/mL, respectively.
  • Herein, it is shown that the denatured TAT-SOD or TAT-GPx fusion protein can be co-delivered into Hela cells and the denatured fusion proteins can be refolded and exhibit the specific enzymatic activities after delivering into the cells. Based on the results shown herein, the TAT-SOD or TAT-GPx fusion protein functionalized FMSN, named as FMSN-TAT-SOD or FMSN-TAT-GPx, still has the enzymatic activity by the refolding mechanism of the cells.
  • In conclusion, by using HSN or MSN, the mesoporous carriers of the present disclosure embodiments can deliver peptides, proteins, enzymes or enzymatic mimetics into the cells as needed and the native activities of the peptides, proteins, enzymes or enzymatic mimetics being delivered into the cell are maintained. The mesoporous carriers can function as nanoreactors located within the cells and the delivered peptides, proteins, enzymes or enzymatic mimetics can work together to provide multiple functions.
  • It will be apparent to those skilled in the art that various modifications and variations can be made to the structure of the present invention without departing from the scope or spirit of the invention. In view of the foregoing, it is intended that the present invention cover modifications and variations of this invention provided they fall within the scope of the following claims and their equivalents.

Claims (36)

What is claimed is:
1. Mesoporous carriers, for delivering targets into a cell, comprising:
hollow silica nanospheres (HSN) or mesoporous silica nanoparticles (MSN); and the targets carried by the hollow silica nanospheres or the mesoporous silica nanoparticles, wherein the targets at least includes first targets and second targets, and the first and second targets are different.
2. The carriers of claim 1, wherein the mesoporous silica nanoparticles are surface-functionalized mesoporous silica nanoparticles functionalized with 3-aminopropyl-trimethoxysilane (APTMS).
3. The carriers of claim 2, wherein the surface-functionalized mesoporous silica nanoparticles are bound with crosslinkers containing polyethylene glycol.
4. The carriers of claim 3, wherein the first target is an antibody and the second target is a peptide respectively bound to the crosslinkers on surface-functionalized mesoporous silica nanoparticles.
5. The carriers of claim 4, wherein the antibody is specific to transcriptional factors, mediators or complex in a signalling pathway and the peptide is a nucleus localization sequence (NLS)-peptide or a NLS like-peptide, so that the surface-functionalized mesoporous silica nanoparticles are sequestered outside of a nuclear membrane of the cell.
6. The carriers of claim 4, wherein the antibody is NF-κB p65 antibody and the peptide is TAT transducing peptide.
7. The carriers of claim 2, wherein the first target is superoxide dismutase and the second target is glutathione peroxidase respectively bound to the crosslinkers on surface-functionalized mesoporous silica nanoparticles.
8. The carriers of claim 7, wherein the first target is TAT transducing peptide bound superoxide dismutase and the second target is TAT transducing peptide bound glutathione peroxidase respectively bound to the crosslinkers on surface-functionalized mesoporous silica nanoparticles.
9. The carriers of claim 7, wherein at least one of superoxide dismutase and glutathione peroxidase is bound to the mesoporous silica nanoparticles in a denatured form or a partially active form.
10. The carriers of claim 2, wherein the surface-functionalized mesoporous silica nanoparticles are labelled with a tracking agent or a dye.
11. The carriers of claim 10, wherein the dye is fluorescein isothiocyanate.
12. The carriers of claim 1, wherein the first and second targets are different enzymes or catalytic mimetics capable of catalysing a cascade reaction.
13. The carriers of claim 12, wherein the enzymes or catalytic mimetics are capable of catalysing the cascade reaction involved in scavenges free radicles in the cell, so as to protect the cell against ROS induced stress.
14. The carriers of claim 1, wherein at least one of the first and second targets is bound to the mesoporous silica nanoparticles in a denatured form or a partially active form.
15. The carriers of claim 1, wherein the first and second targets are co-encapsulated within the hollow silica nanospheres.
16. The carriers of claim 15, wherein the first and second targets are different enzymes or catalytic mimetics capable of catalysing a cascade reaction.
17. The carriers of claim 16, wherein the enzymes or catalytic mimetics are capable of catalysing the cascade reaction involved in scavenges free radicles in the cell, so as to protect the cell against ROS induced stress.
18. The carriers of claim 15, the first target is polyethyleneimine-grafted superoxide dismutase and the second target is polyethyleneimine-grafted catalase.
19. A method of delivering targets into a cell, comprising:
providing mesoporous carriers, wherein the mesoporous carriers comprises hollow silica nanospheres (HSN) or mesoporous silica nanoparticles (MSN) and the targets carried by the hollow silica nanospheres or the mesoporous silica nanoparticles, and the targets at least include first targets and second targets, and the first and second targets are different;
contacting the mesoporous carriers with the cell by incubating the cell with the mesoporous carriers; and
delivering the first targets and the second targets into the cell at the same time, wherein the mesoporous carriers and the targets carried by the hollow silica nanospheres or the mesoporous silica nanoparticles enter into the cell.
20. The method of claim 19, wherein providing mesoporous carriers includes surface-functionalizing the mesoporous silica nanoparticles with 3-aminopropyl-trimethoxysilane (APTMS) to form surface-functionalized mesoporous silica nanoparticles.
21. The method of claim 20, wherein the surface-functionalized mesoporous silica nanoparticles are bound with crosslinkers containing polyethylene glycol.
22. The method of claim 21, wherein the first target is an antibody and the second target is a peptide respectively bound to the crosslinkers on surface-functionalized mesoporous silica nanoparticles.
23. The method of claim 22, wherein the antibody is specific to transcriptional factors, mediators or complex in a signalling pathway and the peptide is a nucleus localization sequence (NLS)-peptide or a NLS like-peptide, so that the surface-functionalized mesoporous silica nanoparticles entering into the cell are sequestered outside of a nuclear membrane of the cell.
24. The method of claim 22, wherein the antibody is NF-κB p65 antibody and the peptide is TAT transducing peptide.
25. The method of claim 20, wherein the first target is superoxide dismutase and the second target is glutathione peroxidase respectively bound to the crosslinkers on surface-functionalized mesoporous silica nanoparticles.
26. The method of claim 25, wherein the first target is TAT transducing peptide bound superoxide dismutase and the second target is TAT transducing peptide bound glutathione peroxidase respectively bound to the crosslinkers on surface-functionalized mesoporous silica nanoparticles.
27. The method of claim 25, wherein at least one of superoxide dismutase and glutathione peroxidase is bound to the mesoporous silica nanoparticles in a denatured farm or a partially active form.
28. The method of claim 20, wherein the surface-functionalized mesoporous silica nanoparticles are labelled with a tracking agent or a dye.
29. The method of claim 28, wherein the dye is fluorescein isothiocyanate.
30. The method of claim 19, wherein the first and second targets are different enzymes or catalytic mimetics capable of catalysing a cascade reaction.
31. The method of claim 30, wherein the enzymes or catalytic mimetics are capable of catalysing the cascade reaction involved in scavenges free radicles in the cell, so as to protect the cell against ROS induced stress.
32. The method of claim 19, wherein at least one of the first and second targets is bound to the mesoporous silica nanoparticles in a denatured form or a partially active form.
33. The method of claim 19, wherein the first and second targets are co-encapsulated within the hollow silica nanospheres.
34. The method of claim 33, wherein the first and second targets are different enzymes or catalytic mimetics capable of catalysing a cascade reaction.
35. The method of claim 34, wherein the enzymes or catalytic mimetics are capable of catalysing the cascade reaction involved in scavenges free radicles in the cell, so as to protect the cell against ROS induced stress.
36. The method of claim 34, the first target is polyethyleneimine-grafted superoxide dismutase and the second target is polyethyleneimine-grafted catalase.
US14/819,438 2014-08-07 2015-08-06 Silica-based mesoporous carrier and delivery method of using the same Abandoned US20160038608A1 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
US14/819,438 US20160038608A1 (en) 2014-08-07 2015-08-06 Silica-based mesoporous carrier and delivery method of using the same
JP2018506167A JP2018533543A (en) 2014-08-07 2016-01-20 Silica-based biomolecular carrier, pharmaceutical composition containing the same, method for producing the same, and use thereof
US15/750,759 US11666662B2 (en) 2014-08-07 2016-01-20 Silica-based biomolecule carrier, pharmaceutical composition comprising the same, preparation method and use thereof
AU2016303039A AU2016303039B2 (en) 2014-08-07 2016-01-20 Silica-based biomolecule carrier, pharmaceutical composition comprising the same, preparation method and use thereof
EP16833417.5A EP3331947A4 (en) 2014-08-07 2016-01-20 Silica-based biomolecule carrier, pharmaceutical composition comprising the same, preparation method and use thereof
CA2994809A CA2994809C (en) 2014-08-07 2016-01-20 Silica-based biomolecule carrier, pharmaceutical composition comprising the same, preparation method and use thereof
PCT/US2016/014194 WO2017023358A1 (en) 2014-08-07 2016-01-20 Silica-based biomolecule carrier, pharmaceutical composition comprising the same, preparation method and use thereof
CN201680046902.6A CN108368303A (en) 2014-08-07 2016-01-20 Type silicon oxide biomolecule carrier, the medical component comprising it, preparation method and the usage
TW105103067A TWI611812B (en) 2014-08-07 2016-01-30 Silica-based biomolecule carrier, pharmaceutical composition comprising the same, preparation method and use thereof
IL257337A IL257337A (en) 2014-08-07 2018-02-04 Silica-based biomolecule carrier, pharmaceutical composition comprising the same, preparation method and use thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201462034192P 2014-08-07 2014-08-07
US201462034282P 2014-08-07 2014-08-07
US201462034181P 2014-08-07 2014-08-07
US14/819,438 US20160038608A1 (en) 2014-08-07 2015-08-06 Silica-based mesoporous carrier and delivery method of using the same

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/750,759 Continuation US11666662B2 (en) 2014-08-07 2016-01-20 Silica-based biomolecule carrier, pharmaceutical composition comprising the same, preparation method and use thereof

Publications (1)

Publication Number Publication Date
US20160038608A1 true US20160038608A1 (en) 2016-02-11

Family

ID=55266615

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/819,438 Abandoned US20160038608A1 (en) 2014-08-07 2015-08-06 Silica-based mesoporous carrier and delivery method of using the same
US15/750,759 Active 2035-12-18 US11666662B2 (en) 2014-08-07 2016-01-20 Silica-based biomolecule carrier, pharmaceutical composition comprising the same, preparation method and use thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/750,759 Active 2035-12-18 US11666662B2 (en) 2014-08-07 2016-01-20 Silica-based biomolecule carrier, pharmaceutical composition comprising the same, preparation method and use thereof

Country Status (9)

Country Link
US (2) US20160038608A1 (en)
EP (1) EP3331947A4 (en)
JP (1) JP2018533543A (en)
CN (1) CN108368303A (en)
AU (1) AU2016303039B2 (en)
CA (1) CA2994809C (en)
IL (1) IL257337A (en)
TW (1) TWI611812B (en)
WO (1) WO2017023358A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108743958A (en) * 2018-05-31 2018-11-06 四川大学 Drug molecule and the GSH response type nanometers of valve molecule synergy carry medicine particle and preparation method thereof
EP3766515A1 (en) * 2019-07-18 2021-01-20 Nano Targeting & Therapy Biopharma Inc. Silica nanosphere for immunotherapy
US11541105B2 (en) 2018-06-01 2023-01-03 The Research Foundation For The State University Of New York Compositions and methods for disrupting biofilm formation and maintenance
US11666662B2 (en) * 2014-08-07 2023-06-06 National Taiwan University Silica-based biomolecule carrier, pharmaceutical composition comprising the same, preparation method and use thereof
CN116327979A (en) * 2023-05-25 2023-06-27 西南石油大学 Transition metal-based mesoporous nano catalytic medicine, preparation method and application

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11629198B2 (en) 2017-12-05 2023-04-18 The Trustees Of The University Of Pennsylvania Fusion proteins and antibodies targeting human red blood cell antigens
TWI666176B (en) * 2018-06-11 2019-07-21 亞洲永盛科技有限公司 Hydrogen peroxide-containing wastewater treatment system and applied enzyme carrier
JP7394493B2 (en) * 2019-07-31 2023-12-13 レモネックス インコーポレイテッド Anticancer agent and method for producing porous silica particles

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1523318A4 (en) * 2002-06-24 2007-07-04 Res Dev Foundation Treatment of human multiple myeloma by curcumin
US7265213B2 (en) * 2005-07-28 2007-09-04 Kpl, Inc. Methodology of conjugating chelators to biomolecules
WO2007075680A2 (en) 2005-12-19 2007-07-05 University Of Vermont And State Agricultural College System and method for delivering a desired material to a cell
US20100183504A1 (en) * 2007-06-14 2010-07-22 Fanqing Frank Chen Multimodal imaging probes for in vivo targeted and non-targeted imaging and therapeutics
US20160041153A1 (en) 2008-11-12 2016-02-11 Kirk Brown Biomarker compositions and markers
US20120207795A1 (en) * 2010-07-13 2012-08-16 The Regents Of The University Of California Cationic polymer coated mesoporous silica nanoparticles and uses thereof
ES2397909B1 (en) * 2011-05-05 2014-06-06 Universidad De Zaragoza PROCEDURE FOR OBTAINING MULTIFUNCTIONAL MATERIALS
WO2013006763A1 (en) * 2011-07-06 2013-01-10 The Regents Of The University Of California Oral delivery of enzymes by nanocapsules for targeted metabolism of alcohol or toxic metabolites
BR112014002975A2 (en) 2011-08-08 2017-03-01 Caris Life Sciences Luxembourg Holdings S A R L biomarker compositions and methods
US20130145488A1 (en) * 2011-12-06 2013-06-06 Iowa State University Research Foundation, Inc. Mesoporous silica nanoparticles suitable for co-delivery
TWI472341B (en) * 2013-06-11 2015-02-11 Univ Nat Yang Ming Oligomeric nanoparticle complex release system
WO2015042279A1 (en) 2013-09-18 2015-03-26 Stc. Unm Torroidal mesoporous silica nanoparticles (tmsnps) and related protocells
US20160038608A1 (en) * 2014-08-07 2016-02-11 National Taiwan University Silica-based mesoporous carrier and delivery method of using the same

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
Louzao et al. "Permeability Effects on the Efficiency of Antioxidant Nanoreactors" (2013), Biomacromolecules, vol. 14: 2364-2372. *
Matsuura et al. "Direct visualization of hetero-enzyme co-encapsulated in mesoporous silicas" (2010), Microporous and Mesoporous Materials, vol. 127: 61-66. *
Tang et al. "Mesoporous Silica Nanoparticles: Synthesis, Biocompatibility and Drug Delivery" (2012) Advanced Materials, vol. 24: 1504-1534. *
Zhang et al. "Thin-Layer Polymer Wrapped Enzymes Encapsulated in Hierarchicallly Mesoporous Silica with High Activity and Enhanced Stability" (March 2014) Scientific Reports, vol. 4: 4421, pgs. 1-8. *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11666662B2 (en) * 2014-08-07 2023-06-06 National Taiwan University Silica-based biomolecule carrier, pharmaceutical composition comprising the same, preparation method and use thereof
CN108743958A (en) * 2018-05-31 2018-11-06 四川大学 Drug molecule and the GSH response type nanometers of valve molecule synergy carry medicine particle and preparation method thereof
US11541105B2 (en) 2018-06-01 2023-01-03 The Research Foundation For The State University Of New York Compositions and methods for disrupting biofilm formation and maintenance
EP3766515A1 (en) * 2019-07-18 2021-01-20 Nano Targeting & Therapy Biopharma Inc. Silica nanosphere for immunotherapy
CN116327979A (en) * 2023-05-25 2023-06-27 西南石油大学 Transition metal-based mesoporous nano catalytic medicine, preparation method and application

Also Published As

Publication number Publication date
US20180228918A1 (en) 2018-08-16
TWI611812B (en) 2018-01-21
EP3331947A4 (en) 2019-04-17
CN108368303A (en) 2018-08-03
US11666662B2 (en) 2023-06-06
EP3331947A1 (en) 2018-06-13
CA2994809C (en) 2023-10-31
CA2994809A1 (en) 2017-02-09
TW201705986A (en) 2017-02-16
WO2017023358A1 (en) 2017-02-09
AU2016303039A1 (en) 2018-03-01
AU2016303039B2 (en) 2021-07-01
JP2018533543A (en) 2018-11-15
IL257337A (en) 2018-03-29

Similar Documents

Publication Publication Date Title
US20160038608A1 (en) Silica-based mesoporous carrier and delivery method of using the same
Meng et al. High-performance self-cascade pyrite nanozymes for apoptosis–ferroptosis synergistic tumor therapy
Lin et al. Approach to deliver two antioxidant enzymes with mesoporous silica nanoparticles into cells
Chang et al. Intracellular implantation of enzymes in hollow silica nanospheres for protein therapy: cascade system of superoxide dismutase and catalase
Li et al. A redox-responsive drug delivery system based on RGD containing peptide-capped mesoporous silica nanoparticles
Shahabi et al. Enhancing cellular uptake and doxorubicin delivery of mesoporous silica nanoparticles via surface functionalization: effects of serum
ES2967598T3 (en) Enzyme-driven functionalized nanomotors
Mortera et al. Cell-induced intracellular controlled release of membrane impermeable cysteine from a mesoporous silica nanoparticle-based drug delivery system
CN113499319B (en) Hollow silica nanoparticle encapsulated with bioactive component, preparation method and application thereof
Liu et al. A graphitic hollow carbon nitride nanosphere as a novel photochemical internalization agent for targeted and stimuli-responsive cancer therapy
Mendez et al. Stimulus-responsive controlled release system by covalent immobilization of an enzyme into mesoporous silica nanoparticles
Chen et al. Dual responsive mesoporous silica nanoparticles for targeted co-delivery of hydrophobic and hydrophilic anticancer drugs to tumor cells
Kankala et al. Killing cancer cells by delivering a nanoreactor for inhibition of catalase and catalytically enhancing intracellular levels of ROS
Geisberger et al. Synthesis, characterisation and cytotoxicity of polyoxometalate/carboxymethyl chitosan nanocomposites
ES2684090T3 (en) Biocidal formulations based on selenium and their methods of use
de la Torre et al. ϵ‐Polylysine‐Capped Mesoporous Silica Nanoparticles as Carrier of the C9h Peptide to Induce Apoptosis in Cancer Cells
Su et al. Recent advances in nanozymes for combating bacterial infection
Sun et al. Bioorthogonal catalytic nanozyme-mediated lysosomal membrane leakage for targeted drug delivery
Tolentino et al. Controlled release of small molecules and proteins from DNA-surfactant stabilized metal organic frameworks
Zhang et al. An optimized mesoporous silica nanosphere-based carrier system with chemically removable Au nanoparticle caps for redox-stimulated and targeted drug delivery
Zhang et al. An engineered, self-propelled nanozyme as reactive oxygen species scavenger
Yang et al. Hyaluronic acid and antimicrobial peptide-modified gold/silver hybrid nanocages to combat bacterial multidrug resistance
Llopis-Lorente et al. Au–Mesoporous silica nanoparticles gated with disulfide-linked oligo (ethylene glycol) chains for tunable cargo delivery mediated by an integrated enzymatic control unit
Liu et al. Controllable drug release system in living cells triggered by enzyme–substrate recognition
Yang et al. Biodegradable doxorubicin-loaded ferric phosphate nanosheets for specific tumor elimination through autophagy inhibition-enhanced apoptosis/ferroptosis pathway

Legal Events

Date Code Title Description
AS Assignment

Owner name: NATIONAL TAIWAN UNIVERSITY, TAIWAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MOU, CHUNG-YUAN;CHEN, YI-PING;WU, SI-HAN;REEL/FRAME:036302/0873

Effective date: 20150714

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE AFTER FINAL ACTION FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: ADVISORY ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

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