WO2011130716A2 - A nanostructures containing vitamin b12 for facilitated delivery of drugs across biological barriers - Google Patents

A nanostructures containing vitamin b12 for facilitated delivery of drugs across biological barriers Download PDF

Info

Publication number
WO2011130716A2
WO2011130716A2 PCT/US2011/032793 US2011032793W WO2011130716A2 WO 2011130716 A2 WO2011130716 A2 WO 2011130716A2 US 2011032793 W US2011032793 W US 2011032793W WO 2011130716 A2 WO2011130716 A2 WO 2011130716A2
Authority
WO
WIPO (PCT)
Prior art keywords
nanoparticle
nanoparticles
charged
alternatively
vitamin
Prior art date
Application number
PCT/US2011/032793
Other languages
French (fr)
Other versions
WO2011130716A3 (en
Inventor
Ryszard Zarzycki
Paul Sood
N. Rao Ummaneni
David P. Nowotnik
Original Assignee
Access Pharmaceuticals, Inc.
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 Access Pharmaceuticals, Inc. filed Critical Access Pharmaceuticals, Inc.
Publication of WO2011130716A2 publication Critical patent/WO2011130716A2/en
Publication of WO2011130716A3 publication Critical patent/WO2011130716A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/27Growth hormone [GH] (Somatotropin)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/28Insulins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • A61K47/54Medicinal 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 an organic compound
    • A61K47/55Medicinal 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 an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
    • A61K47/551Medicinal 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 an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds one of the codrug's components being a vitamin, e.g. niacinamide, vitamin B3, cobalamin, vitamin B12, folate, vitamin A or retinoic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5161Polysaccharides, e.g. alginate, chitosan, cellulose derivatives; Cyclodextrin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/88Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation using microencapsulation, e.g. using amphiphile liposome vesicle
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2810/00Vectors comprising a targeting moiety
    • C12N2810/10Vectors comprising a non-peptidic targeting moiety

Definitions

  • the invention relates to the delivery of pharmaceutically-active compounds such as small-molecule drugs, proteins and peptides across biological barriers using naturally-occuring vitamin transport systems.
  • the invention relates to the delivery of pharmaceutically-active compounds utilizing vitamin B12 transport systems with the protection of the active during transport by incorporation in nanostructures, such as, but not limited to, nanostructures containing surface-linked vitamin B12.
  • the nanostructures are made from charged polymers.
  • the invention also relates to processes for preparing the nanostructures, pharmaceutical compositions containing same and methods of drug delivery and treatment of disease involving the nanostructures.
  • cytotoxic drugs in cancer chemotherapy, it may often be necessary to dose patients with high levels of cytotoxic drugs in order to achieve a meaningful therapeutic effect which may also result in damage to normal cells, resulting in significant adverse side-effects. It may be desirable to alter the natural biodistribution of cytotoxic compounds so that more of the drug is delivered to tumor cells, and less to normal tissues.
  • Monoclonal antibodies to tumor-specific antigens have been used as target cytotoxic agents to tumors so as to improve upon the therapeutic index (a ratio of a drug's beneficial effects compared with its adverse side-effects).
  • the use of monoclonal antibodies may generate other issues, such as immunogenicity, whereby the patient's immune systems may develop an immune response to the antibody-drug conjugate.
  • PEC polyelectrolyte complexes
  • the PECs of this invention are able to provide for either or both oral drug delivery of unmodified pharmaceutically-active compounds and other therapeutic agent through transfer from the intestinal lumen into the bloodstream, or targeted to diseased cells in the body that over express the receptors that facilitate vitamin B12 cell uptake.
  • the present invention relates to one or more nanoparticle(s) formed by bringing together in a suitable solvent a synthetic or natural polymer comprising multiple charged and/or ionisable groups, such as, positively charged or negatively charged, with a therapeutic agent which has one or more charged or ionisable groups of the opposite charge to the charge of the polymer, and optionally other components which either assist in nanoparticle formation or in the placement of a vitamin B12 on the surface of the nanoparticle.
  • the polymer and the therapeutically active agent can form a PEC, either alone or in combination with the other components, e.g., the aforementioned components.
  • the present invention relates to nanoparticles formed by bringing together in a suitable solvent two synthetic or natural polymers in which one polymer comprises multiple charged and/or ionisable groups of net positive charge and the other polymer comprises one or multiple charged and/or ionisable groups of net negative charge, together with a therapeutic agent which may or may not possess charged or ionisable groups, and optionally other components which either assist in nanoparticle formation or in the placement of a vitamin B12 on the surface of the nanoparticle.
  • the two polymers can form a PEC, either alone or in combination with the other aforementioned components, and the therapeutically active component is trapped in the PEC during PEC formation.
  • the vitamin B12 is introduced prior to nanoparticle formation either by covalent attachment to the polymer, to the therapeutic agent, and/or to one of the optional additional components.
  • Figure 1 depicts a structure of vitamin B12, in which represents a monodentate axial ligand as defined later.
  • attachment of a vitamin B12 can occur directly to one of the above mentioned components of the nanoparticle or via a suitable linker.
  • Vitamin B12 attachment can occur via either the 2' or 5 '-oxygen atoms on the ribose unit of vitamin B12 (as exemplified by U.S. Pat. No.
  • nanoparticles are formed by bringing together the components of the nanoparticle in an aqueous environment, although other solvent systems known in the art may also be used.
  • the nanoparticles formed may be either soluble or insoluble in the solvent system.
  • Nanoparticles can be isolated by techniques known in the art. For example, soluble nanoparticles can be isolated by precipitation with a cosolvent or by removal of solvent (e.g. lyophilization or spray drying) optionally preceeded by a purification method such as tangential flow filtration (TFF) or centrifugal ultrafiltration. Insoluble nanoparticles might be isolated by centrifugation or filtration, also optionally preceeded by a purification method such as TFF.
  • TFF tangential flow filtration
  • the solid nanoparticles formed and isolated as described above can be formulated for human or veterinary administration by standard methods.
  • the nanoparticles can be formulated into tablets or capsules for oral administration, as lyophilized or dried formulations in vials for subsequent reconstitution with an injection vehicle and administration to humans or animals by injection, or as solutions or suspensions for administration to humans or animals by injection.
  • the pharmaceutical formulations of the nanoparticles of this invention are useful for oral drug delivery and/or disease targeted delivery of a wide variety of therapeutic agents, including, but not limited to, small and large synthetic molecules, proteins, peptides, glycoproteins, humanized and non-humanized monoclonal antibodies and therapeutically relevant fragments thereof, and agents for effecting the delivery of polynucleotides alone or in combination with a gene delivery vector.
  • therapeutic agents including, but not limited to, small and large synthetic molecules, proteins, peptides, glycoproteins, humanized and non-humanized monoclonal antibodies and therapeutically relevant fragments thereof, and agents for effecting the delivery of polynucleotides alone or in combination with a gene delivery vector.
  • the polynucleotides include for example those which are, or that encode RNA
  • RNAi RNA interference
  • siRNA siRNA
  • dsRNA dsRNA
  • mRNA RNA interference
  • antisense RNA DNA
  • DNA DNA
  • compositions of the nanoparticles of this invention are useful to treat a wide variety of diseases including, but not limited to cancer, autoimmune conditions, endocrine disorders, diabetes, genetic conditions,
  • chromosome conditions viral infections, bacterial infections, parasitic infections, mitochondrial diseases, sexually transmitted diseases, immune disorders, balance disorders, pain, systemic disorders, blood conditions, blood vessel conditions, nerve conditions, and conditions of muscles, heart and other organs.
  • a nanoparticle comprising, or alternatively consisting essentially of or alternatively consisting of one or more synthetic or natural polymers comprising one or more charged and/or ionisable groups, a therapeutic agent comprising one or more charged and/or ionisable groups of the opposite charge to the charge of the polymers, and, a vitamin B12 covalently linked to the
  • the one or more charged and/or ionisable groups of the one or more synthetic or natural polymers are positive charge or negative charge.
  • a nanoparticle comprising, or alternatively consisting essentially of or alternatively consisting of one or more synthetic or natural polymers comprising one or more charged and/or ionisable groups which are positively charged, one or more synthetic or natural polymers comprising one or more charged and/or ionisable groups which are negatively charged, a therapeutic agent, and a vitamin B12 covalently linked to the nanoparticle via an optional linker group.
  • the vitamin B12 is attached to one or more of the charged and/or ionisable groups of the one or more synthetic or natural polymers.
  • the vitamin B12 is attached to the therapeutic agent.
  • the nanoparticle of the above noted aspects further comprises one or more of components selected from the group consisting of polyethylene glycol (PEG), PEG block copolymers, polyacrylic, polymethacrylic, polyacrylamide, polymethacrylamide, synthetic polymer, polysaccharide, surfactant, and metal ions.
  • PEG polyethylene glycol
  • PEG block copolymers polyacrylic, polymethacrylic, polyacrylamide, polymethacrylamide, synthetic polymer, polysaccharide, surfactant, and metal ions.
  • the vitamin B12 is attached to one or more of the components.
  • an average nanoparticle diameter is in a range of about 20 nm to about 800 nm.
  • the nanoparticle is configured for oral administration in a subject.
  • the nanoparticle is configured for administration by injection to a subject.
  • the synthetic or natural polymer is selected from the group consisting of poly-Y-glutamic acid ( ⁇ -PGA), succinylated and/or
  • carboxymethylated polysaccharide poly(acrylic acid), poly(p-styrene-sulfonic acid sodium salt), poly(sulfopropylacrylate), negatively-charged polypeptide, chitosan derivatives, polyethyleneimine, positively-charged polypeptide, pharmaceutically- approved polymeric excipients that carry a net negative charge at physiologically- relevant pH levels, and pharmaceutically-approved polymeric excipients that carry a net positive charge at physiologically-relevent pH levels.
  • the synthetic or natural polymer is selected from the group consisting of chitosan derivatives, polyethyleneimine, positively-charged polypeptide, and pharmaceutically-approved polymeric excipients that carry a net positive charge at physiologically-relevent pH levels.
  • the synthetic or natural polymer is selected from the group consisting of poly-Y-glutamic acid ( ⁇ -PGA), succinylated and/or
  • carboxymethylated polysaccharide poly(acrylic acid), poly(p-styrene-sulfonic acid sodium salt), poly(sulfopropylacrylate), negatively-charged polypeptide, and pharmaceutically-approved polymeric excipients that carry a net negative charge at physiologically-relevent pH levels.
  • the succinylated and/or carboxymethylated polysaccharide as noted above is dextran, cellulose, or starch.
  • the negatively-charged polypeptide as noted above is polyglutamic acid.
  • the positively-charged polypeptide as noted above is polylysine.
  • an axial ligand substituent on a cobalt atom of vitamin B 12 is a chemical entity such as cyano (-CN), methyl (CH 3 ), hydro xyl (-OH) or nitrosyl (-NO).
  • the therapeutic agent is selected from the group consisting of a small or large synthetic or semi-synthetic molecule, protein, peptide, glycoprotein, nucleoside, nucleotide, humanized monoclonal antibody, non- humanized monoclonal antibody, therapeutically relevant fragments of humanized and/or non-humanized monoclonal antibody, and agents for effecting RNA interference (RNAi) such as dsRNA, miRNA, siRNA and antisense RNA.
  • RNAi RNA interference
  • polynucleotides includes deoxyribonucleic acid (DNA), and, where appropriate, ribonucleic acid (RNA).
  • RNA or DNA made from nucleotide analogs, and, as applicable to the embodiment being described, single (sense or antisense) and double-stranded polynucleotides.
  • Deoxyribonucleotides include deoxyadenosine, deoxycytidine, deoxyguanosine, and deoxythymidine.
  • a nucleotide of a nucleic acid which can be DNA or an RNA
  • the terms "adenosine”, “cytidine”, “guanosine”, and “thymidine” are used. It is understood that if the nucleic acid is RNA, a nucleotide having a uracil base is uridine.
  • polynucleotide and “oligonucleotide” are used interchangeably and refer to a polymeric form of nucleotides of any length, either
  • Polynucleotides can have any three-dimensional structure and may perform any function, known or unknown.
  • mRNA messenger RNA
  • transfer RNA transfer RNA
  • ribosomal RNA ribozymes
  • cDNA cDNA
  • dsRNA siRNA
  • miRNA miRNA
  • a polynucleotide can comprise modified nucleotides, such as methylated nucleotides and nucleotide analogs. If present, modifications to the nucleotide structure can be imparted before or after assembly of the polynucleotide.
  • the sequence of nucleotides can be interrupted by non-nucleotide components.
  • a polynucleotide can be further modified after polymerization, such as by conjugation with a labeling component.
  • the term also refers to both double- and single-stranded molecules. Unless otherwise specified or required, any embodiment of this invention that is a polynucleotide encompasses both the double-stranded form and each of two complementary single-stranded forms known or predicted to make up the
  • a polynucleotide is composed of a specific sequence of four nucleotide bases: adenine (A); cytosine (C); guanine (G); thymine (T); and uracil (U) for thymine when the polynucleotide is RNA.
  • A adenine
  • C cytosine
  • G guanine
  • T thymine
  • U uracil
  • polynucleotide sequence is the alphabetical representation of a polynucleotide molecule. This alphabetical representation can be input into databases in a computer having a central processing unit and used for bioinformatics applications such as functional genomics and homology searching.
  • polymorphism refers to the coexistence of more than one form of a gene or portion thereof.
  • a polymorphic region can be a single nucleotide, the identity of which differs in different alleles.
  • carrier encompasses any of the standard carriers, such as a phosphate buffered saline solution, buffers, water, and emulsions, such as an oil/water or water/oil emulsion, and various types of wetting agents.
  • compositions also can include stabilizers and preservatives.
  • carriers for examples of carriers, stabilizers and adjuvants, see Sambrook and Russell (2001), supra. Those skilled in the art will know many other suitable carriers for binding polynucleotides, or will be able to ascertain the same by use of routine experimentation.
  • the carrier is a buffered solution such as, but not limited to, a PCR buffer solution.
  • a "gene delivery vehicle” is defined as any molecule that can carry inserted polynucleotides into a host cell.
  • Examples of gene delivery vehicles are liposomes, biocompatible polymers, including natural polymers and synthetic polymers;
  • lipoproteins polypeptides; polysaccharides; lipopolysaccharides; artificial viral envelopes; metal particles; and bacteria, or viruses, such as baculovirus, adenovirus and retrovirus, bacteriophage, cosmid, plasmid, fungal vectors and other
  • recombination vehicles typically used in the art which have been described for expression in a variety of eukaryotic and prokaryotic hosts, and may be used for gene therapy as well as for simple protein expression.
  • Gene delivery are terms referring to the introduction of an exogenous polynucleotide (sometimes referred to as a "transgene") into a host cell, irrespective of the method used for the introduction.
  • exogenous polynucleotide sometimes referred to as a "transgene”
  • transgene an exogenous polynucleotide
  • Such methods include a variety of well-known techniques such as vector-mediated gene transfer (by, e.g., viral infection, sometimes called transduction), transfection, transformation or various other protein-based or lipid-based gene delivery complexes) as well as techniques facilitating the delivery of "naked" polynucleotides (such as electroporation, "gene gun” delivery and various other techniques used for the introduction of polynucleotides).
  • transfected, transduced or transformed may be used interchangeably herein to indicate the presence of exogenous polynucleotides or the expressed polypeptide therefrom in a cell.
  • the introduced polynucleotide may be stably or transiently maintained in the host cell. Stable maintenance typically requires that the introduced polynucleotide either contains an origin of replication compatible with the host cell or integrates into a replicon of the host cell such as an extrachromosomal replicon (e.g., a plasmid) or a nuclear or mitochondrial chromosome.
  • a number of vectors are known to be capable of mediating transfer of genes to mammalian cells, as is known in the art and described herein.
  • RNA interference refers to sequence-specific or gene specific suppression of gene expression (protein synthesis) that is mediated by short interfering RNA (siRNA).
  • siRNA short interfering RNA
  • dsRNA double-stranded RNA molecules
  • RNAi RNA interference
  • 11 nucleotides in length 12 nucleotides in length
  • 13 nucleotides in length 14 nucleotides in length
  • 15 nucleotides in length 16 nucleotides in length
  • 17 nucleotides in length 18 nucleotides in length
  • 19 nucleotides in length 20 nucleotides in length, 21 nucleotides in length, 22 nucleotides in length, 23 nucleotides in length
  • 24 nucleotides in length 25 nucleotides in length
  • 26 nucleotides in length 27 nucleotides in length
  • 28 nucleotides in length or 29 nucleotides in length.
  • siRNA includes short hairpin RNAs (shRNAs).
  • a siRNA directed to a gene or the mRNA of a gene may be a siRNA that recognizes the mRNA of the gene and directs a RNA- induced silencing complex (RISC) to the mRNA, leading to degradation of the mRNA.
  • a siRNA directed to a gene or the mRNA of a gene may also be a siRNA that recognizes the mRNA and inhibits translation of the mRNA.
  • dsRNA double stranded RNA molecules that may be of any length and may be cleaved intracellularly into smaller RNA molecules, such as siRNA.
  • dsRNA In cells that have a competent interferon response, longer dsRNA, such as those longer than about 30 base pair in length, may trigger the interferon response. In other cells that do not have a competent interferon response, dsRNA may be used to trigger specific RNAi.
  • a siRNA can be designed following procedures known in the art. See, e.g., Dykxhoorn, D.M. and Lieberman, J. (2006) “Running Interference: Prospects and Obstacles to Using Small Interfering RNAs as Small Molecule Drugs," Annu. Rev. Biomed. Eng. 8:377-402; Dykxhoorn, D.M. et al. (2006) "The silent treatment:
  • siRNAs as small molecule drugs Gene Therapy, 13:541-52; Aagaard, L. and Rossi, J.J. (2007) "RNAi therapeutics: Principles, prospects and challenges,” Adv. Drug Delivery Rev. 59:75-86; de Fougerolles, A. et al. (2007) “Interfering with disease: a progress report on siRNA-based therapeutics," Nature Reviews Drug Discovery 6:443-53; Krueger, U. et al. (2007) "Insights into effective RNAi gained from large- scale siRNA validation screening," Oligonucleotides 17:237-250; U.S. Patent Application Publication No.: 2008/0188430; and U.S. Patent Application Publication No.:2008/0249055.
  • siRNAs can be made with methods known in the art. See, e.g., Dykxhoorn, D.M. and Lieberman, J. (2006) “Running Interference: Prospects and Obstacles to Using Small Interfering RNAs as Small Molecule Drugs," Annu. Rev. Biomed. Eng. 8:377-402; Dykxhoorn, D.M. et al. (2006) “The silent treatment: siRNAs as small molecule drugs," Gene Therapy, 13:541-52; Aagaard, L. and Rossi, J.J. (2007) “RNAi therapeutics: Principles, prospects and challenges," Adv. Drug Delivery Rev. 59:75- 86; de Fougerolles, A.
  • a siRNA may be chemically modified to increase its stability and safety. See, e.g. Dykxhoorn, D.M. and Lieberman, J. (2006) “Running Interference: Prospects and Obstacles to Using Small Interfering RNAs as Small Molecule Drugs," Annu. Rev. Biomed. Eng. 8:377-402 and U.S. Patent Application Publication No.: 2008/0249055.
  • microRNA or miRNA are single-stranded RNA molecules of 21-23 nucleotides in length, which regulate gene expression. miRNAs are encoded by genes from whose DNA they are transcribed but miRNAs are not translated into protein (non-coding RNA); instead each primary transcript (a pri-miRNA) is processed into a short stem-loop structure called a pre-miRNA and finally into a functional miRNA. Mature miRNA molecules are partially complementary to one or more messenger RNA (mRNA) molecules, and their main function is to down-regulate gene expression.
  • mRNA messenger RNA
  • a siRNA vector, dsRNA vector or miRNA vector as used herein refers to a plasmid or viral vector comprising a promoter regulating expression of the RNA.
  • "siRNA promoters" or promoters that regulate expression of siRNA, dsRNA, or miRNA are known in the art, e.g., a U6 promoter as described in Miyagishi and Taira (2002) Nature Biotech. 20:497-500, and a HI promoter as described in Brummelkamp et al. (2002) Science 296:550-3.
  • the therapeutic agent is selected from the group consisting of analgesic, antiallergenic, antianginal agent, antiarrythmic drug, antibiotic, anticoagulant, antidementia drug, antidepressant, antidiabetic,
  • antihistamine antihypertensive, anti-inflammatory, antineoplastic agent, antiparasitic, antipyretic, antiretroviral drug, antiulcerative agent, antiviral agent, cardiovascular drug, cholesterol-lowering agent, CNS active drug, a hormone, growth hormone inhibitor, growth hormone, hematopoietic drug, hemostatic, hypotensive diuretic, keratolytic, therapeutic for osteoporosis, vaccine, vasoconstrictor, and vasodilator.
  • a process for preparing a nanoparticle composition comprising the nanoparticle of any of the above recited aspects and embodiments, comprising, or alternatively consisting essentially of or alternatively consisting of combining the one or more synthetic or natural polymers, the therapeutic agent, and the vitamin B 12, in a suitable solvent, and isolating, purifying and/or drying the nanoparticles.
  • the solvent is > 50% water.
  • a process for preparing a nanoparticle composition comprising the nanoparticle of any of the above recited aspects and embodiments, comprising, or alternatively consisting essentially of or alternatively consisting of mixing two immiscible solvents and a surfactant to produce an emulsion, optionally cross-linking the nanoparticles, and isolating, purifying, and/or drying resultant nanoparticles.
  • the nanoparticles are isolated by solvent evaporation
  • the nanoparticles are isolated by filtration or centrifugation
  • the nanoparticles are isolated by addition of a cosolvent followed by filtration or centrifugation.
  • the purifying step is effected by washing the nanoparticles with a suitable solvent.
  • the above recited aspects further comprise modifying the nanoparticles to effect cross-linking of the components of the nanoparticle.
  • the above recited aspects further comprise modifying the nanoparticles to add a vitamin B12 analog to a surface of the nanoparticle by physical or covalent attachment.
  • the above recited aspects further comprise modifying the nanoparticles to substitue an axial ligand on a one or more cobalt atoms of attached vitamin B12 with replacement axial ligands, non-limiting examples of which are provided herein.
  • composition comprising the nanoparticle of the above recited aspects, and a pharmaceutically- acceptable excipient.
  • the composition is formulated as a tablet, a capsule, or a liquid.
  • the composition is formulated as a lyophilized powder in a container for subsequent re-suspension or dissolution of the
  • composition in a pharmaceutically-acceptable injection vehicle.
  • the composition is formulated as a suspension or solution in a pharmaceutically-acceptable injection vehicle.
  • a method for treating a subject comprising, or alternatively consisting essentially of, or alternatively consisting of, administring an effective amount of the nanoparticle of any of the above recited aspects or the pharmaceutical composition of any of the above recited aspects.
  • the therapeutic agent is an antidiabetic agent.
  • the therapeutic agent is a hormone.
  • the therapeutic agent is an antineoplastic agent.
  • Figure 1 depicts a structure of vitamin B12.
  • Figure 2 depicts three VB12 drug delivery formats including nanoparticles.
  • Figure 3 depicts a graph showing plasma hGH vs time.
  • Figure 4 shows a reduction of MSI2 gene expression.
  • K562 cells were transfected with 50 nM siRNA targeting MSI2 gene or 50 nM Negative Control siRNA. Forty-eight hours post-transfection, the cells were harvested and analyzed by real-time RT-PCR for MSI2 and 18S rRNA levels. 18S rRNA levels were used to normalize MSI2 expression. "Remaining mRNA (%)” was calculated as a percentage of MSI2 mRNA levels in K562 cells transfected with MSI2 siRNA relative to K562 cells transfected with non-silencing siRNA (Negative Control siRNA). Data are presented as means ⁇ SD.
  • Figure 5 shows 18S rRNA levels. Level of expression of ribosomal RNA (18S) levels is directly associated with number of cells in the sample. Data demonstrate that Lipofectamine 2000 induces significant cytotoxicity at 3 ⁇ ⁇ per well (200 ⁇ ⁇ total volume) and higher volumes. In contrast, CobT-0 and CobT-7 show no toxicity at volumes up to 30 ⁇ ⁇ (5 mg/ml). Bars on the right at each of the noted comparisons show siMS12. Bars on the left show siNC.
  • compositions and methods refers to compounds, compositions and methods including the recited elements, but not exclude others.
  • Consisting essentially of when used to define compounds, compositions or methods, shall mean excluding other elements that would materially affect the basic and novel characteristics of the claimed technology.
  • Consisting of shall mean excluding any element, step, or ingredient not specified in the claim. Embodiments defined by each of these transition terms are within the scope of this technology.
  • nanoparticle or “nanostructure” refers a microscopic particle less than about 1 micron in diameter.
  • the nanoparticles range in size from about 1 nm to about 1 ,000 nm diameter, or alternatively between about 10 nm to about 1000 nm, or alternatively between about 10 nm to about 900 nm, or alternatively between about 10 nm to about 800 nm, or alternatively between about 10 nm to about 700 nm, or alternatively between about 10 nm to about 600 nm, or alternatively between about 10 nm to about 500 nm, or alternatively between about 20 nm to about 1000, or alternatively between about 20 nm to about 800 nm, or alternatively between about 20 nm to about 700 nm, or alternatively between about 20 nm to about 600 nm, or alternatively between about 20 nm to about 500 nm; or alternatively between about 30 nm to about
  • polymer refers to a naturally-occurring, synthetic or semisynthetic large molecule (macromolecule) typically composed of repeating structural units connected by covalent chemical bonds. Polymers useful for the implementation of this invention have molecular weights in the range of 1 to 5000 kDa.
  • random copolymer refers to a polymer comprising two or more repeating structural units in which the sequence of the individual repeating structural units is random and not predetermined or defined.
  • block copolymer refers to a polymer comprising two or more repeating structural units in which individual repeating structural units are connected to each other forming identifyable blocks of repeating structural units within the complete polymer strand.
  • charged group refers to a chemical functional group that is fully ionized resulting in that group having either a positive or a negative charge, or possibly multiple positive or multiple negative charges.
  • Polymers could have multiple charged groups either as components of the polymer chain, and/or as attachments to the polymer, either direct attachment or by way of a linker. Polymer charged groups may be either naturally-occuring or synthetic. A charged group may be part of a therapeutically active compound, either as an intrinsic component of that compound or as a synthetic analog of the therapeutically active compound, for example a prodrug.
  • ionisable group refers to a chemical functional group that is partially ionized at or close to physiological pH resulting in that group having either a partial positive or a partial negative charge.
  • the charge of an ionisable group will vary with pH.
  • Polymers could have multiple ionisable groups either as components of the polymer chain, and/or as attachments to the polymer, either direct attachment or by way of a linker. Polymer ionisable groups may be either naturally-occuring or synthetic. A ionisable group may be part of a therapeutically active compound, either as an intrinsic component of that compound or as a synthetic analog of the
  • therapeutically active compound for example a prodrug.
  • PEC polyelectrolyte complex
  • the diameter of PECs can typically range from 1 nm to several microns, with average particle size and particle size distribution controlled by the chemical and physical nature of the constituent components and method of preparation.
  • PECs can be water soluble (i.e. suspension of nanoparticles in water results in a clear, transparent liquid) or insoluble (i.e.
  • PEC nanoparticles typically can range in size from about 1 nm to about 1,000 nm diameter, or alternatively about 5 nm to about 400 nm or alternatively about 10 nm to about 300 nm.
  • the PECs of the invention range in size from about 1 nm to about 1,000 nm diameter, or alternatively between about 10 nm to about 1000 nm, or alternatively between about 10 nm to about 900 nm, or alternatively between about 10 nm to about 800 nm, or alternatively between about 10 nm to about 700 nm, or alternatively between about 10 nm to about 600 nm, or alternatively between about 10 nm to about 500 nm, or alternatively between about 20 nm to about 1000, or alternatively between about 20 nm to about 800 nm, or alternatively between about 20 nm to about 700 nm, or alternatively between about 20 nm to about 600 nm, or alternatively between about 20 nm to about 500 nm; or alternatively between about 20 nm to about 400 nm; or alternatively between about 20 nm to about 200 nm; or alternatively between about 30 nm to about 1000 nm,
  • alkyl refers to a saturated (containing no multiple carbon- carbon bonds) aliphatic (containing no delocalized ⁇ -electron system), hydrocarbon containing, if otherwise unsubstituted, only carbon and hydrogen atoms.
  • An alkyl group herein may be optionally substituted with one or more entities selected from the group consisting of halo, hydroxy, alkoxy, aryloxy, carbonyl, nitro, cyano, carboxyl and alkoxycarbonyl.
  • linker refers to a group of atoms that is used to couple a polymeric backbone to another function or group to spatially separate the two entities.
  • a linker of this invention has an essentially longitudinal axis, that is, it is essentially linear rather than highly branched or clumped, although the structure will, of course, not be exactly linear due to the angular constraints placed on the structure by required bond angles between covalently bonded atoms.
  • linkers include, but are not limited to, straight and branced alkyl and alkenyl groups containing functional groups such as carboxyl, amino, hydroxyl, and thiol, through which covalent bonds can be formed to connect the linker to the polymer and to other components.
  • a preferred linker is a short peptide chain ( H-[NHCH -CO]n-OH) where n is 1-20, or alternatively from 1-18, or alternatively from 1-16, or alternatively from 1-14, or alternatively from 1-12, or alternatively from 2-14, or alternatively from 2-12, or alternatively from 3-20, or alternatively from 4-18, or alternatively from 5- 20, or alternatively from 5-18, and R is the same or different for each of the n amino acids, and is one of the 22 side groups known to be present in natural amino acids.
  • a peptide linker can be incorporated into the polymer compound by one of the peptide condensation reactions (producing an amide bond) that are known in the art.
  • therapeutic agent refers to a compound, mixture of compounds, or biologic agent that can provide a beneficial effect when administered to a patient.
  • amino acid refers to a compound containing both amino (- NH 2 ) and carboxyl (-COOH) groups generally separated by one carbon atom.
  • the central carbon atom may contain a substituent which can be either charged, ionisable, hydrophilic or hydrophobic.
  • amino acid can optionally include those with an unnatural 'D' stereochemistry and modified forms of the 'D' and 'L' amino acids.
  • peptide refers to a chain of amino acids in which each amino acid is connected to the next by a formation of an amide bond.
  • Peptides are generally considered to consist of up to 30 amino acids, or alternatively upto 25 amino acids, or alternatively upto 20 amino acids, or alternatively upto 15 amino acids, or alternatively upto 10 amino acids, or alternatively up to 5 amino acids, or alternatively between about 5-10 amino acids, or alternatively between about 10-15 amino acids, while the term “protein” is applied to compounds containing longer amino acid chains.
  • glycoprotein refers to a protein which contains a number of carbohydrate substituents.
  • halo or halogen refers to fluorine (F), chlorine (CI), bromine (Br) and iodine (I).
  • a primary, secondary or tertiary alkyl amine refers to an RNH 2 , an RR"NH or an ' "N group, wherein R, R' and R" independently represent, without limitation, alkyl, cycloalkyl, aryl, heteroaryl and heteroalicyclic moieties.
  • vitamin B12 or VB12 refers to the series of compounds otherwise know as cobalamins which are structurally identical and vary only in the nature of the monodentate axial ligand attached to the VB12 cobalt atom, which typically can be cyanide (cyanocobalamin), methyl (methylcobalamin), hydroxyl (hydroxycobalamin), or nitric oxide (nitrosylcobalamin). It is known in the art that these axial ligands can be exchanged under appropriate conditions, and such ligand exchange is incorporated as part of this disclosure.
  • Linkage of the VB12 to the polymer systems to create the delivery systems described herein can be accomplished by converting one or more amide to carboxyl then using the free carboxyl to form a covalent link.
  • formation of a covalent bond to one of the two hydroxyl groups of the ribose unit of VB12 can be employed.
  • VB12 could be linked to the polymer system might also be accomplished by addition of a suitable monodentate ligand to the polymer, via an optional linker, and formation of a metal coordinate bond between the cobalt atom of VB12 and the polymer-attached monodentate ligand.
  • a "disease” or “medical condition” is an abnormal condition of an organism that impairs bodily functions, associated with specific symptoms and signs.
  • cancer refers to various types of malignant neoplasms, most of which can invade surrounding tissues, and may metastasize to different sites, as defined by Stedman's Medical Dictionary, 25th edition (Hensyl ed. 1990).
  • cancers which may be treated using the compounds of the present invention include, but are not limited to, brain, ovarian, colon, prostate, kidney, bladder, breast, lung, oral, skin and blood cancers.
  • a tumor-seeking group refers to an entity that is know to preferentially seek out and bond to surface structures on neoplastic cells that do not occur or are expressed to a substantially lesser degree by normal cells or entitles that preferentially accumulate in tumors over normal tissue.
  • the terms “treat”, “treating” and “treatment” refer to a method of alleviating or abrogating a disease and/or its attendant symptoms.
  • the effect may be prophylactic in terms of completely or partially preventing a disorder or sign or symptom thereof, and/or may be therapeutic in terms of a partial or complete cure for a disorder and/or adverse effect attributable to the disorder.
  • the life expectancy of an individual affected with a cancer will be increased and/or that one or more of the symptoms of the disease will be reduced.
  • administer refers to the delivery of a compound or compounds of this invention or of a pharmaceutical composition containing a compound or compounds of this invention to a patient in a manner suitable for the treatment of a particular disease, such as cancer.
  • administering can be effected in one dose, continuously or intermittently throughout the course of treatment. Methods of determining the most effective means and dosage of administration are known to those of skill in the art and will vary with the composition used for therapy, the purpose of the therapy, the target cell being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician. Suitable dosage formulations and methods of administering the agents are known in the art. Route of administration can also be determined and method of determining the most effective route of administration are known to those of skill in the art and will vary with the composition used for treatment, the purpose of the treatment, the health condition or disease stage of the subject being treated, and target cell or tissue.
  • Non-limiting examples of route of administration include oral administration, nasal administration, injection, and topical application.
  • a "patient” or a “subject” refers to any higher organism that is susceptible to disease. Examples of such higher organisms include, without limitation, mice, rats, rabbits, dogs, cats, horses, cows, pigs, sheep, fish and reptiles. In some embodiments, "patient” or “subject” refers to a human being.
  • the term "therapeutically effective amount” refers to that amount of a compound or combination of compounds of this invention which has the effect of (a) preventing a disorder from occurring in a subject that may be predisposed to a disorder, but may have not yet been diagnosed as having it; (b) inhibiting a disorder, i.e., arresting its development; or (c) relieving or ameliorating the disorder.
  • reducing the size of the tumor For example, but not limited to, (1) reducing the size of the tumor; (2) inhibiting (that is, slowing to some extent, preferably stopping) tumor metastasis; (3) inhibiting to some extent (that is slowing to some extent, preferably stopping) tumor growth; (4) relieving to some extent (or preferably eliminating) one or more symptoms associated with the cancer; and/or (5) extending survival time of the patient.
  • a "pharmaceutical composition” refers to a mixture of one or more of the compounds of this invention with other chemical components such as pharmaceutically acceptable excipients.
  • the purpose of a pharmacological composition is to facilitate administration of a compound of this invention to a patient.
  • “pharmaceutically acceptable carrier” refers to an excipient that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered composition.
  • “Pharmaceutically acceptable carrier” or “pharmaceutically acceptable excipient” refers to any diluents, excipients, or carriers that may be used in the compositions of the invention.
  • excipients or carriers include, without limitation, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances, such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.
  • Suitable pharmaceutical carriers are described in Remington's
  • Enzyme inhibitors are enzyme inhibitors; Mucoadhesive polymeric systems; Particulate carrier delivery systems; and Targeted delivery systems.
  • Absorption or permeation enhancers are molecules which either increase the fluidity of membranes or widen junctions between the cells of membranes thus providing a small transient improvement in paracellular and transcellular drug transport.
  • absorption enhancers for oral drug delivery There are a number of distinct disadvantages to absorption enhancers for oral drug delivery:
  • Enzyme inhibitors slow the rate at which actives, particularly proteins and peptides, are enzymatically degraded in the GI tract. In principle, this provides for a higher concentration of the active at the sites of absorption, resulting in greater passive absorption by virtue of a larger concentration gradient. This effect is only beneficial for actives that are naturally able to diffuse readily across the gut wall, and are only prevented from doing so through enzymatic degradation of the active compound. Additionally, inhibition of enzyme activity in the GI tract can give rise to significant adverse effects as inhibition of protein degradation will be non-selective. For example, enzyme inhibitors will reduce the rate to breakdown (and hence reduced absorption) of food proteins.
  • Peristalsis generates a flow of material down the GI tract. Materials moving along the small intestine, where most pharmaceutical actives are thought to be absorbed, do so in an averge time of about three hours. If were possible to retard the flow of drugs, and provide them with greater contact at the sites of absorption, it should be possible to achieve higher levels of absorption of drugs that are otherwise poorly absorbed in the GI tract. Because of transient 'sticking' of mucoadhesive polymeric systems to the mucosal surface of the GI tract lumen, formulations based upon such polymers have the potential to demonstrate an extended residence on the epithelial cell layer, slowing the flow of these particles relative to other material in the GI tract. .
  • mucoadhesive polymers When formulated into particles, mucoadhesive polymers may also provide some protection to embedded active agents that might otherwise be degraded in the GI tract. Because of the direct contact between the polymer formulation and the GI mucosa, other potential advantages of this oral drug delivery system is the possibility for direct diffusion of actives from the particle into the mucosa and epithelial cell layer, and for pinocytosis of particles into epithelial cells. All of these potential benefits suggest that oral drug delivery systems based upon mucoadhesive polymers should be highly effective, yet results to date in numerous examples in the literature indicate only modest improvements in oral bioavailability of pharmaceutical active compounds using mucoadhesive polymer formulations.
  • Gastrointestinal absorption of many essential nutrients and vitamins can be facilitated by active transport processes. These processes generally require the material to bind to a surface receptor, which initiates a process such as receptor- mediated endocytosis whereby the active is absorbed into the epithelial cell. Dissassociation of the receptor-active complex occurs and other processes then facilitate the transfer of the active material into the blood stream.
  • a surface receptor which initiates a process such as receptor- mediated endocytosis whereby the active is absorbed into the epithelial cell. Dissassociation of the receptor-active complex occurs and other processes then facilitate the transfer of the active material into the blood stream.
  • VB12 vitamin B12
  • VB12 liberated from food binds to intrinsic factor (IF, which is produced in the stomach and passes down the GI tract following a meal), and the VB12-IF complex binds to the Cubulin receptor, primarily located in the ileum.
  • IF intrinsic factor
  • Receptor-mediated endocytosis as described above, then takes place. Dissociation of the receptor-IF-VB12 complex in the epithelial cell results in liberation of VB12, which then binds to transcobalamin II, a protein which facilities the transfer of VB12 to the blood stream.
  • the active is either covalently linked via a degradable linker group to VB12, or covalently linked via a degradable linker group to a polymer which is also linked to VB12, or encapsulated in a nanoparticle to which VB12 is attached (see Figure 2).
  • the active is either covalently linked via a degradable linker group to VB12, or covalently linked via a degradable linker group to a polymer which is also linked to VB12, or encapsulated in a nanoparticle to which VB12 is attached (see Figure 2).
  • multiple drug-linker groups can be attached to a single polymer strand.
  • these constructs will bind IF in the GI tract and be taken up primarily in the ileum by the cubulin receptor and transported to the bloodstream. Breakdown of the degradable linker will then release drug in the bloodstream, completing its oral absorption. Similarly, drug release by diffusion from the nanoparticle and/or breakdown of the nanoparticle structure in the bloodstream will result in
  • the polymer approach allows for multiple drug molecules to be absorbed each time one polymer strand is absorbed as a result of VB12 attached to that polymer strand binding to IF and cubulin. This allows for an 'amplication' of oral uptake when compared with the 1 : 1 conjugate.
  • a VB12 nanoparticle can carry many copies of the drug, also permitting amplification of drug uptake.
  • the formation of a covalent link to connect the drug to VB12 is or may not be the method of utilizing this technology.
  • a covalent link to the drug By formation of a covalent link to the drug, it may be chemically altered.
  • API active pharmaceutical ingredient
  • the release of the drug requires cleigen of the degradable linker, which may leave fragments of the linker still attached to the drug, such that is a different chemical entity.
  • By trapping the drug in a VB12-coated nanoparticle it remains chemically unaltered, so a previously approved drug should not need a full development program for Regulatory approval of the VB12-coated nanoparticle formulation of that drug.
  • formulations described in these patents have advanced from basic research to the clinic as each of these technologies has fundamental technical issues; as examples poor encapsulation / weak binding of the drug to the carrier.
  • the methods of preparation known in the art can also give rise to degradation or denaturing of protein, peptides and other pharmaceutical active ingredients, lowering efficacy and introducing additional impurities.
  • vitamin B12 In many diseases, cells have an increased demand for vitamin B12 which is reflected by an increase in the expression of cell surface receptors which facilitate the uptake, through receptor-mediated endocytosis, of this vitamin.
  • vitamin B12 binds to the circulating protein, transcobalamin II (TC-II), and it is the B12-TC-II complex which is recognized by the cell surface receptors.
  • TC-II transcobalamin II
  • the B12-TC-II complex binding results in receptor-mediated endocytosis and internalization of the complex, followed by release of the vitamin B12.
  • vitamin B12 uptake in the GI tract the process for cell uptake of vitamin B12 can be utilized using the 'Trojan Horse' principle to transport molecules into cells when these molecules are chemically linked to vitamin B12.
  • 'Trojan Horse' principle to transport molecules into cells when these molecules are chemically linked to vitamin B12.
  • Polyelectrolyte complex is a term which relates to two or more compounds binding to each other by virtue of multiple charge interactions.
  • PEC Polyelectrolyte complex
  • This polymer when brought into contact with an compound containing one or more charged (or ionisable) groups of the oppositve charge forms a complex wherein the charged groups on one compound form ionic bonds with the charged groups of the other compound.
  • both compounds possess charged or ionisable groups and form multiple ionic bonds with each other.
  • nanoparticles many molecules of the two or more charged or ionisable compounds come together to form a three-dimensional matrix of nanoparticle size.
  • PECs can be formed simply by bringing both compounds together in an aqueous environment.
  • drug-loaded PECs a simple example might be a pharmacologically-active peptide with either a net positive or net negative overall charge at a suitable pH with a polymer which has charged (or ionisable) groups which have the opposite charge to that of the peptide.
  • PECs results from the formation of PECs from two polymers, one with negatively charged groups and one with positively-charged groups. Bringing these two polymers into contact in an aqueous environment which also contains the drug can result in the formation of PECs in which the drug is trapped in the nanoparticle matrix during PEC formation. In either of these two general examples, controlled drug release can result through slow disassociation of the nanoparticle in the body.
  • the PEC components can be chemically-modified to assist in nanoparticle formation; for example, conversion of a tertiary to quaternary amine or through the addition of hydrophobic groups
  • PECs intended to deliver a therapeutically- beneficial payload can be found in the following publications (all of which are incorporated herein by reference in their entirety): A Jintapattanakit et al, International Journal of Pharmaceutics, 2007, 342, 240-249; Y-Z Du et al, International Journal of Pharmaceutics, 2010 (eprint); Vieira and AM Carmona- ibeiro, Journal of
  • targeting groups have been attached to charged polymers to enhance the delivery of these constructs in vivo to certain target regions, e.g. cancer.
  • targeting groups which have been attached to charged polymers include transferin (M. Kursa et al, Bioconjugate Chemistry, 2003, 14 222) and folate (SH Kim et al, Biotechnol Prog. 2007, 23, 232), During nanoparticle formation such large and/or hydrophobic targeting groups might be expected to migrate towards the surface of nanoparticles in preference to the hydrophilic environment within the internal matrix of the nanoparticle.
  • PEC polyelectrolyte complex
  • the above PEC nanoparticle system provides some protection from degradation or denaturing of the one or more therapeutically-active compounds contained within the nanoparticle in body compartments in body compartments in which one or more therapeutically- active compounds contained within the nanoparticle might otherwise, if unprotected, be caused to degrade, denature or metabolize.
  • the above PEC nanoparticle system has the potential benefit of transportation from one body compartment to another by utilizing the body's natural transportation mechanisms for vitamin B12, including, but not limited to, transportation from the gut lumen to the portal blood vein in the ileum of the GI tract, passage across cell membranes to enter cellular compartments, and traverse major biological barriers such as the blood-brain barrier.
  • the above PEC nanoparticle system can release the one or more therapeutically active compounds contained within the nanoparticle in a controlled manner, and that compound release can result from diffusion of drug through the nanoparticle matrix and/or degradation of the matrix.
  • the above PEC nanoparticle system can release the one or more therapeutically active compounds contained within the nanoparticle at sites within the body to achieve a therapeutically- meaningful effect.
  • the above PEC nanoparticle system can degrade in the body to permit the components of the nanoparticle to be safely metabolized and eliminated from the body.
  • the above PEC nanoparticle system can formulated by methods known in the art to provide pharmaceutical preparations suitable for administration to patients.
  • Examples of pharmaceutical preparations that might be suitable for the PEC nanoparticle system of this invention include, but are not limited to, tablets or capsules for oral
  • the above PEC nanoparticle system be administered to patients for the prevention and treatment of diseases, including, but not limited to cancer, autoimmune conditions, endocrine disorders, diabetes, genetic conditions, chromosome conditions, viral infections, bacterial infections, parasitic infections, mitochondrial diseases, sexually transmitted diseases, immune disorders, balance disorders, pain, systemic disorders, blood conditions, blood vessel conditions, nerve conditions, and conditions of muscles, heart and other organs.
  • diseases including, but not limited to cancer, autoimmune conditions, endocrine disorders, diabetes, genetic conditions, chromosome conditions, viral infections, bacterial infections, parasitic infections, mitochondrial diseases, sexually transmitted diseases, immune disorders, balance disorders, pain, systemic disorders, blood conditions, blood vessel conditions, nerve conditions, and conditions of muscles, heart and other organs.
  • the present invention consists of nanoparticles formed by bringing together in a suitable solvent a synthetic or natural polymer containing multiple charged and/or ionisable groups, predominantly either positive or negative, with a therapeutic agent which has one or more charged or ionisable groups of the opposite charge to the primary charge of the polymer, and optionally other
  • VB12 is an essential component of the nanoparticle, introduced prior to nanoparticle formation either by covalent attachment to the polymer, to the therapeutic agent, and/or to one of the optional additional components.
  • the present invention consists of nanoparticles formed by bringing together in a suitable solvent two synthetic or natural polymers in which one polymer contains multiple charged and/or ionisable groups of net positive charge and the other polymer contains multiple charged and/or ionisable groups of net negative charge, together with a therapeutic agent which may or may not possess charged or ionisable groups, and optionally other components which either assist in nanoparticle formation or in the placement of VB12 on the surface of the nanoparticle.
  • the two polymers can form a PEC, either alone or in combination with the other
  • VB12 is an essential component of the nanoparticle, introduced prior to nanoparticle formation either by covalent attachment to the polymer, to the therapeutic agent, and/or to one of the optional additional components.
  • the present invention consists of nanoparticles formed as described above in which VB12 in not a component or part of a component of the nanoparticle and is introduced to the surface of the nanoparticle after nanoparticle formation either by formation of a covalent bond between the nanoparticle surface and VB12 or VB12 derivative, or by the formation of a physical bonds (ionic, hydrophilic, and/or hydrophobic) between the nanoparticle and VB12 or VB12 derivative.
  • vitamin B12 contains a monodentate axial ligand. It is known in the art that these axial ligands can be exchanged under appropriate conditions, and such ligand exchange is incorporated as part this disclosure. For example, it is known that nitrosyl cobalamin can be effective as an antitumor agent because it serves to deliver nitric oxide to tumors (for example; Bauer, Anti-Cancer Drugs, 1998, 9, 239) and it may be desirable to convert VB12 in the nanoparticles of this invention to the nitrosyl form to enhance the therapeutic effect.
  • nitrosyl cobalamin can be effective as an antitumor agent because it serves to deliver nitric oxide to tumors (for example; Bauer, Anti-Cancer Drugs, 1998, 9, 239) and it may be desirable to convert VB12 in the nanoparticles of this invention to the nitrosyl form to enhance the therapeutic effect.
  • the VB12 may be connected to the linker through the cobalt atom of VB12 by way of a ligand exchange process, as described in (for example; U.S. Patent Publication No.
  • VB12 can be attached using other methods known in the art.
  • one or more of the primary amide groups of VB12 may be selectively hydro lyzed to generate a free carboxyl group or ester, and subsequently the VB12 can be linked to the polymer via an optional linker through the liberated carboxyl group by methods well-known in the art (for example; Wilbur et al, Bioconjugate Chem. 1996, Vol. 7:461-474).
  • the preferred method of attachment of VB12 to the polymer via an optional linker involves the formation of a covalent bond to one of the two hydroxyl groups of the ribose unit of VB12 by methods known in the art (for example; McEwan et al, Bioconjugate Chem. 1999, Vol. 10: 1131-1136).
  • Examples of positively charged or ionizable polymers that can be used to form the PEC nanoparticles of this invention include chitosan, alkylated and acylated chitosan derivatives, DEAE-dextran, polyethylenimine, polyamidoamines, poly(vinyl- N-alkylpyridinium), poly(DMAE-methacrylamide), poly(di- and trialkylamino- methacrylamides), poly(HPMA-co-di- and trialkylamino-methacrylamides), positively-charged polypeptides such as polylysine, polyhistidine, and
  • the chitosan derivative employed is ⁇ , ⁇ , ⁇ -trimethyl chitosan (TMC).
  • TMC ⁇ , ⁇ , ⁇ -trimethyl chitosan
  • the positively charged or ionizable polymers may be optionally modified by covalent linkage of one or more VB12 molecules, either directly or via a suitable linker.
  • Examples of negatively charged or ionizable polymers that can be used to form the PEC nanoparticles of this invention include anionic polysaccharides such as heparin, chondroitin, hyaluronic acid, alginates, pectins, carrageenans, xanthans, neutral polysaccharides such as dextran, cellulose, starch, chitosan that have been derivatized with alkyl- or acyl-carboxylates, sulfates, sulfonates or phosphates, anionic poly-amino acids such as polyglutamic acid, poly-gamma-glutamic acid, polyaspartic acid, synthetic polymers such as poly(acrylate),
  • anionic polysaccharides such as heparin, chondroitin, hyaluronic acid, alginates, pectins, carrageenans, xanthans
  • neutral polysaccharides such as dextran, cellulose, starch,
  • the dextran derivative employed is succinyl-dextran (that is, dextran treated with succinic anhydride providing substituents with one free carboxyl group).
  • the dextran derivative employed is carboxymethyl-dextran (that is, dextran treated with a carboxylmethyl halide providing substituents with one free carboxyl group).
  • the negatively charged or ionizable polymers may be optionally modified by covalent linkage of one or more VB12 molecules, either directly or via a suitable linker.
  • PECs are formed by interacting a therapeutically-active compound which possess a net positive or negative charge with a polymer as described above with the opposite charge. Nanoparticle formation and/or the suitability of the resultant nanoparticle for pharmaceutical use may be facilitated or improved by the use of additional components during nanoparticle formation such that these additional components become incorporated into the nanoparticle.
  • a ratio of the therapeutic agent to the vitamin B12 in the nanoparticles of the present invention is in a range of 1 :20 to about 20: 1 , or alternatively in a range of about 1 : 15 to about 15: 1, or alternatively in a range of about 1 : 10 to about 10: 1, or alternatively in a range of about 1 :5 to about 5: 1, or alternatively in a range of about 1 :2 to about 2: 1 , or alternatively the ratio of the therapeutic agent to the vitamin B12 in the nanoparticles of the present invention is about 1 : 1 , or alternatively about 2: 1 , or alternatively about 1 :2, or alternatively about 3: 1, or alternatively about 1 :3, or alternatively about 4:1, or alternatively about 1 :4, or alternatively about 5: 1, or alternatively about 1:5, or alternatively about 6: 1, or alternatively about 1 :6, or alternatively about 7: 1, or alternatively about 1 :7, or alternatively about 8: 1, or alternatively
  • PECs are formed by interacting a therapeutically-active compound which may or may not possess a net positive or negative charge with two polymers as described above, one with a net positive charge and one with a net negative charge.
  • Nanoparticle formation and/or the suitability of the resultant nanoparticle for pharmaceutical use may be facilitated or improved by the use of additional components during nanoparticle formation such that these additional components become incorporated into the nanoparticle.
  • nanoparticles can also be formed by use of more than one polymer of a particular charge.
  • two or more positively charged polymers can be used when a positively charged polymer is required.
  • two or more negatively charged polymers can be used when a negatively charged polymer is required.
  • pharmaceutically-suitable nanoparticles can also be formed by incorporation of more than one therapeutically-active compound.
  • PEC nanoparticles it may be desirous in the formation of PEC nanoparticles to utilize additional components before, during or after nanoparticle formation in order to control the size of nanoparticles, control stability and/or the drug release profile.
  • additional components include, but are not limited to, polyethylene glycol (PEG) and PEG block copolymers, polyacrylic, polymethacrylic, and other synthetic polymers, starch, cellulose, and other polysaccharides, fatty acids and other surfactants, and metal ions, especially di- and trivalent ions such as zinc,
  • Additional components might also include a crosslinking agent, for example epoxy compounds, dialdehyde starch, glutaraldehyde,
  • VB12 is bound to the nanoparticle after nanoparticle formation, then one of the components used in the formation of the nanoparticle must contain VB12 either covalently of physically linked to that component.
  • VB12 might be linked, directly or via a suitable linker, to either the positively-charged polymer, the negatively-charged polymer, the therapeutically- active compound, or one of the additional components (if employed).
  • the primary purpose of the additional component is to facilitate the introduction of VB12 to the the nanoparticle during its formation.
  • the additional component could be VB12 which contains a fatty acid attached to either the 5 '-0 or 2'-0 position (or both), and the VB12 is incorporated by hydrophobic interaction of the fatty acid portion with other hydrophobic components involved in nanoparticle formation.
  • Other methods of incorporating VB12 as one of the additional components will be obvious to those skilled in the art.
  • the VB12 additional component may be functionalized with a compound that is known to bind strongly to one of the other components of nanoparticle formation (e.g. strepatavidin and biotin are well known to bind strongly to each other; similarly, U.S. Pat. 5,605,890 exemplifies a cyclodextrin- adamantane "lock and key" binding system).
  • the polymers used in this invention can have an average molecular weight in the range of 1-10,000 kDa.
  • the preferred average molecular weights will be determined by the specific requirements of formation and the desired pharmaceutical properties of the PEC nanoparticles.
  • the avergage molecular weight of the polymer of the invention is in a range of about 1-10,000 kDa; or alternatively in a range of about 1-5000 KDa; or alternatively in a range of about 1- 1000 KDa; or alternatively in a range of about 1-500 KDa; or alternatively in a range of about 1-100 KDa; or alternatively in a range of about 10-10,000 KDa; or alternatively in a range of about 10-5000 KDa; or alternatively in a range of about 10- 4000 KDa; or alternatively in a range of about 10-2000 KDa; or alternatively in a range of about 10-1000 KDa; or alternatively in a range of about 10-500 KDa; or alternatively in a range of a range
  • a function of the nanoparticles of this invention is to facilitate or enhance the oral bioavailablity of the therapeutically active compound (or compounds) contained within the nanoparticle.
  • the therapeutically active compound (or compounds) may have poor natural oral bioavailability by virtue of either (or both) degradation or denaturing in the GI tract or an inability to cross the gut wall and enter the bloodstream.
  • a function of the nanoparticles of this invention is to modify the oral bioavailablity of the therapeutically active compound (or compounds) contained within the nanoparticle.
  • the therapeutically active compound (or compounds) may have sufficient oral bioavailability to be therapeutically effective when given orally, and the nanoparticles of this invention either improve oral bioavailability (reducing the amount of drug that needs to be adminstered) and/or alters the pharmacokinetic profile of the drug in a desirable manner.
  • a function of the nanoparticles of this invention is to facilitate targeting of the therapeutically active compound (or compounds) contained within the nanoparticle to sites of disease, especially in diseases in which the demand for VB12 is increased compared with the demand for the vitamin normally.
  • diseases which are known to display increased demand for VB12 include cancer, rheumatoid arthritis, psoriasis, acute leukemia, lymphomas, Crohn's disease, ulcerative colitis, and multiple sclerosis.
  • Pharmaceutical preparations for targeted delivery to sites of disease can be administered by injection.
  • a function of the nanoparticles of this invention is to combine oral drug delivery and targeting; following oral drug delivery as described above, the nanoparticles are then targeted to sites of disease, also as described above.
  • a function of the nanoparticles of this invention is to deliver polynucleotides (e.g. siRNA and antisense RNA) and other RNA interference therapeutics across cell membranes to deliver the actives into the intracellular environment and to the nucleus, where they are effective, and for gene therapy.
  • polynucleotides e.g. siRNA and antisense RNA
  • other RNA interference therapeutics across cell membranes to deliver the actives into the intracellular environment and to the nucleus, where they are effective, and for gene therapy.
  • a function of the nanoparticles of this invention is to deliver therapeutics which are effective in the treatment of CNS disorders across the blood-brain barrier.
  • Therapeutic agents that can be delivered in effective amounts across biological barriers using the PEC nanoparticles of this invention include, but are not limited to small molecules, macromolecules, synthetic drugs, semi-synthetic drugs, naturally-occurring compounds, proteins, peptides, nucleosides, nucleotides, analgesics, antiallergenics, antianginal agents, antiarrythmic drugs, antibiotics, anticoagulants, antidementia drugs, antidepressants, antidiabetics, antihistamines, antihypertensives, anti-inflammatories, antineoplastic agents, antiparasitics, antipyretic, antiretroviral drugs, antiulcerative agents, antiviral agents, cardiovascular drugs, cholesterol-lowering agents, CNS active drugs, growth hormone inhibitors, growth hormones, hematopoietic drugs, hemostatics, hormones, hypotensive diuretics, keratolytics, therapeutics for osteoporosis, vaccines, vasoconstrictors, vaso
  • therapeutic agents that are analgesics are morphine,
  • therapeutic agents that are antiallergic compounds include amlexanox, astemizole, azelastinep, emirolast, alopatadine, cromolyn, fenpiprane, repirinast, tranilast, and traxanox.
  • therapeutic agents that are antianginal agents include nifedipine, atenol, bepridil, carazolol and epanolol.
  • therapeutic agents that are anti-inflammatory analgesic agents include acetaminophen, methyl salicylate, monoglycol salicylate, aspirin, mefenamic acid, flufenamic acid, indomethacin, diclofenac, alclofenac, diclofenac sodium, ibuprofen, ketoprofen, naproxen, pranoprofen, fenoprofen, sulindac, fenclofenac, clidanac, flurbiprofen, fentiazac, bufexamac, piroxicam, phenylbutazone,
  • therapeutic agents that are steroidal anti-inflammatory agents include hydrocortisone, predonisolone, dexamethasone, triamcinolone acetonide, fluocinolone acetonide, hydrocortisone acetate, predonisolone acetate,
  • methylpredonisolone dexamethasone acetate, betamethasone, betamethasone valerate, flumetasone, fiuorometholone, beclomethasone diproprionate, etc.
  • therapeutic agents that are antihistamines include
  • diphenhydramine hydrochloride diphenhydramine salicylate, diphenhydramine, chlorpheniramine hydrochloride, chlorpheniramine maleate isothipendyl
  • hydrochloride tripelennamine hydrochloride, promethazine hydrochloride, methdilazine hydrochloride, etc.
  • Examples of therapeutic agents that are vasoconstrictors include naphazoline nitrate, tetrahydrozoline hydrochloride, oxymetazoline hydrochloride, phenylephrine hydrochloride, tramazoline hydrochloride, etc.
  • hemostatics examples include thrombin, phytonadione, protamine sulfate, aminocaproic acid, tranexamic acid, carbazochrome, carbaxochrome sodium sulfanate, rutin, hesperidin, etc.
  • therapeutic agents that are chemotherapeutic drugs include sulfamine, sulfathiazole, sulfadiazine, homosulfamine, sulfisoxazole, sulfisomidine, sulfamethizole, nitro furazone, taxanes, platinum compounds, topoisomerase I inhibitors, and anthrocycline.
  • antibiotics examples include penicillin, meticillin, oxacillin, cefalotin, cefalordin, erythromycin, lincomycin, tetracycline, chlortetracycline, oxytetracycline, metacycline, chloramphenicol, kanamycin, streptomycin, gentamicin, bacitracin, cycloserine, and clindamycin.
  • therapeutic agents that are growth factors include Autocrine motility factor, Bone morphogenetic proteins (BMPs), Epidermal growth factor
  • EGF Erythropoietin
  • FGF Fibroblast growth factor
  • G-CSF Granulocyte-colony stimulating factor
  • GM- CSF Granulocyte-macrophage colony stimulating factor
  • GDF9 Growth differentiation factor-9
  • HGF Hepatocyte growth factor
  • HDGF Hepatoma derived growth factor
  • IGF Insulin-like growth factor
  • GDF-8 Nerve growth factor
  • PDGF Platelet-derived growth factor
  • TPO Thrombopoietin
  • TGF Transforming growth factor alpha
  • TGF-? Vascular endothelial growth factor
  • VEGF Vascular endothelial growth factor
  • PlGF placental growth factor
  • FBS Foetal Bovine Somatotrophin
  • Examples of therapeutic agents that are growth hormone inhibitors are octreotide and somatostatin.
  • Examples of therapeutic agents that are hormones include Adiponectin, Adrenocorticotropic hormone (or corticotropin), Aldosterone, Androstenedione, Angiotensinogen and angiotensin, Antidiuretic hormone (or vasopressin, arginine vasopressin), Antimullerian hormone (or mullerian inhibiting factor or hormone), Atrial-natriuretic peptide (or atriopeptin), Brain natriuretic peptide, Calcidiol (25- hydroxyvitamin D3), Calcitonin, Calcitriol, Cholecystokinin, Corticotropin-releasing hormone, Cortisol, Dehydroepiandrosterone, Dihydrotestosterone, Dopamine (or prolactin inhibiting hormone), Endothelin, Enkephalin, Epinephrine (or adrenaline), Erythropoietin, Estradiol, Estriol, Estrone, Fol
  • somatomedin Leptin, Leukotrienes, Lipotropin, Luteinizing hormone, Melanocyte stimulating hormone, Melatonin, Neuropeptide Y, Norepinephrine (or noradrenaline), Orexin, Oxytocin, Pancreatic polypeptide, Parathyroid hormone, Progesterone, Prolactin, Prolactin releasing hormone, Prostacyclin, Prostaglandins, elaxin, Renin, Secretin, Serotonin, Somatostatin, Testosterone, Thrombopoietin, Thromboxane, Thyroid-stimulating hormone (or thyrotropin), Thyrotropin-releasing hormone, Thyroxine, Triiodothyronine.
  • therapeutic agents that are antiviral drugs include Abacavir, Aciclovir, Acyclovir, Adefovir,Amantadine, Amprenavir, Ampligen, Arbidol, Atazanavir, Atripla, Boceprevir, Cidofovir, Combivir, Darunavir, Delavirdine,
  • Ibacitabine Imunovir,Idoxuridine, Imiquimod, Indinavir, Inosine, Interferon type III, Interferon type II, Interferon type I, Interferon, Lamivudine, Lopinavir, Loviride, Maraviroc, Moroxydine, Nelfinavir, Nevirapine, Nexavir, Oseltamivir, Peginterferon alfa-2a, Penciclovir, Peramivir, Pleconaril, Podophyllotoxin,Raltegravir, Reverse transcriptase inhibitor, Ribavirin, Rimantadine, Ritonavir, Pyramidine, Saquinavir, Stavudine, Tea tree oil, Tenofovir, Tenofovir disoproxil, Tipranavir, Trifluridine, Trizivir, Tromantadine, Truvada, Valaciclovir , Valganciclovir, Vicriviroc, Vidarabine, Viramidine, Zalcitabine
  • Examples of therapeutic agents that are drugs for the treatment of diabetes or its side effects includes insulin (natural or recombinant; monomer, hexamer, or mixtures thereof), insulin isophane, insulin lispro, insulin glargine, tolbutamide, acetohexamide, tolazamide, chlorpropamide, glipizide, glyburide, glimepiride, gliclazide, repaglinide, nateglinide, metformin, phenformin, buformin, rosiglitazone, pioglitazone, troglitazone, miglitol, acarbose, Glucagon-like peptide-1, Exanatide, Liraglutide, Taspoglutide, Lixisenatide, Albiglutide, vildagliptin, sitagliptin, saxagliptin, pramlintide, muraglitazar, tesaglitazar,
  • Examples of therapeutic agents that are drugs used for the treatment of CNS disorders include memantine hydrochloride, donepezil hydrochloride, rivastigmine tartrate, galantamine hydrochloride, and tacrine hydrochloride.
  • therapeutic agents that are drugs used for the treatment of prostate cancer include Dutasteride, Bicalutamide, Ciprofloxacin, Erythromycin, Tamsulosin, Ofloxacin, Terazosin, Leuprolide, Nilutamide, Finasteride, and
  • Examples of therapeutic agents that are drugs used for the treatment of ovarian cancer include Cisplatin, Carboplatin, Paclitaxel, Melphalan, Doxorubicin, hexamethylmelamine, Toptecan, Ifosfamide, Etoposide, and 5-fluorouracil.
  • Examples of therapeutic agents that are drugs used for the treatment of colorectal cancer include fluorouracil, bevacizumab, irinotecan, oxaliplatin, cetuximab, panitumumab, leucovorin, and capecitabine.
  • Examples of therapeutic agents that are drugs used for the treatment of lung cancer include Carboplatin, Cisplatin, Docetaxel, Erlotinib, Etoposide, Gemcitabine, Gefitinib. Irinotecan, Paclitaxel, Pemetrexed, Topotecan, Vinorelbine, Gefitinib, and Bevacizumab.
  • Examples of therapeutic agents that are drugs used for the treatment of melanoma include dacarbazine, interferon alfa-2b, aldesleukin, and acarbazine.
  • Examples of therapeutic agents that are drugs used for the treatment multiple sclerosis include Interferon Beta la, Glatiramer Acetate, Mitoxantrone, Azathioprine, Cyclophosphamide, Cyclosporine, Methotrexate, Cladribine, MethylPrednisolone, Prednisone, Prednisolone, Dexamethasone, Corticotropin, Carbamazepine,
  • Examples of therapeutic agents that are drugs used for the treatment of Alzheimers disease include donepezil, galantamine, rivastigmine, and memantine.
  • Examples of therapeutic agents that are drugs used for the treatment of arthritis include etanercept, infliximab, adalimumab, celecoxib, ituximab, abatacept, etoricoxib, golimumab, ofatumumab, and certolizumab pegol.
  • therapeutic agents that are drugs used for the treatment of blood deficiencies include pegfilgrastim, GCSF, PEG-GCSF, Darbepoetin alfa, Epoetin, Heparin (including low molecular weight derivatives), and warfarin.
  • Examples of therapeutic agents that are drugs used for the treatment of mucositis include Palifermin.
  • protein therapeutic agents are also monoclonal antibodies, a polyclonal antibodies, humanized antibodies, antibody fragments, and
  • RNA interference examples include, but are not limited to siRNA, dsDNA, miRNA, and antisense RNA.
  • Examples of therapeutic agents that are antibodies or their fragments include Abciximab, Adalimumab, Alemtuzumab, Basiliximab, Bevacizumab, Cetuximab, Certolizumab, Daclizumab, Eculizumab, Efalizumab, Gemtuzumab, Ibritumomab tiuxetan, Infliximab, Muromonab-CD3, Natalizumab, Omalizumab, Palivizumab, Panitumumab, Ranibizumab, Rituximab, Tositumomab, and Trastuzumab.
  • therapeutic agents that are PEGylated drugs include
  • Peginterferon alfa-2a Peginterferon alfa-2b, Pegaspargase, and Pegfilgrastim.
  • therapeutic agents that are small molecules include
  • Atorvastatin Clopidrogel, Aripiprazole, Esomeprazole, Olanzapine, Quetiapine, Rosuvastatin, Monte lukast, Venlafaxine Enoxaparin, and Pioglitazone.
  • the present technology provides compositions comprising or consisting essentially of a nanoparticle of the present technology and a carrier, diluent, or excipient.
  • the carrier, diluent, or excipient is pharmaceutically acceptable.
  • the nanoparticle may comprise an agent or agents which in turn are compounds or isomers, prodrug, tautomer, or pharmaceutically acceptable salts thereof, of the present technology can be formulated in the pharmaceutically acceptable compositions per se, or in the form of a hydrate, solvate, N-oxide, or pharmaceutically acceptable salt, as described herein.
  • such salts are more soluble in aqueous solutions than the corresponding free acids and bases, but salts having lower solubility than the corresponding free acids and bases may also be formed.
  • the present technology includes within its scope solvates of the compounds and salts thereof, for example, hydrates.
  • the present technology provides a pharmaceutically acceptable composition (formulation) comprising a nanoparticle and at least one pharmaceutically acceptable excipient, diluent, preservative, stabilizer, or mixture thereof.
  • the methods can be practiced as a therapeutic approach towards the treatment of the conditions described herein.
  • the compounds of the present technology can be used to treat the conditions described herein in animal subjects, including humans.
  • the methods generally comprise administering to the subject a nanoparticle of the present technology, or a salt, prodrug, hydrate, or N-oxide thereof, effective to treat the condition.
  • prodrug of a compound of the present technology is a compound that is converted in vivo or in vitro to the compound of the present technology. Hydrolysis, oxidation, and/or reduction are some ways that a prodrug is converted to the compound of the present technology.
  • the subject is a non-human mammal, including, but not limited to, bovine, horse, feline, canine, rodent, or primate. In another embodiment, the subject is a human.
  • compositions comprising the nanoparticles described herein (or salts or prodrugs thereof) can be manufactured by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping, or lyophilization processes.
  • the compositions can be formulated in conventional manner using one or more physiologically acceptable carriers, diluents, excipients, or auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically.
  • the nanoparticles of the present technology can be administered by oral, parenteral (e.g., intramuscular, intraperitoneal, intravenous, ICV, intracisternal injection or infusion, subcutaneous injection, or implant), by inhalation spray, nasal, vaginal, rectal, sublingual, urethral (e.g., urethral suppository) or topical routes of administration (e.g., gel, ointment, cream, aerosol, etc.) and can be formulated, alone or together, in suitable dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants, excipients, and vehicles appropriate for each route of administration.
  • parenteral e.g., intramuscular, intraperitoneal, intravenous, ICV, intracisternal injection or infusion, subcutaneous injection, or implant
  • inhalation spray nasal, vaginal, rectal, sublingual, urethral (e.g., urethral suppository) or topical routes of administration
  • compositions for the administration of the compounds can be conveniently presented in unit dosage form and can be prepared by any of the methods well known in the art.
  • the pharmaceutically acceptable compositions can be, for example, prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier, a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation.
  • the active object compound is included in an amount sufficient to produce the desired therapeutic effect.
  • compositions of the present technology may take a form suitable for virtually any mode of administration, including, for example, topical, ocular, oral, buccal, systemic, nasal, injection, transdermal, rectal, and vaginal, or a form suitable for administration by inhalation or insufflation.
  • compositions include those designed for administration by injection (e.g., subcutaneous, intravenous, intramuscular, intrathecal, or intraperitoneal injection) as well as those designed for transdermal, transmucosal, oral, or pulmonary administration.
  • Useful injectable pharmaceutically acceptable compositions include sterile suspensions, solutions, or emulsions of the active compound(s) in aqueous or oily vehicles.
  • the pharmaceutically acceptable compositions may also contain formulating agents, such as suspending, stabilizing, and/or dispersing agents.
  • the formulations for injection can be presented in unit dosage form, e.g., in ampules or in multidose containers, and may contain added preservatives.
  • the injectable pharmaceutically acceptable compositions can be provided in powder form for reconstitution with a suitable vehicle, including but not limited to sterile pyrogen free water, buffer, and dextrose solution, before use.
  • a suitable vehicle including but not limited to sterile pyrogen free water, buffer, and dextrose solution, before use.
  • the active compound(s) can be dried by any art-known technique, such as lyophilization, and reconstituted prior to use.
  • penetrants appropriate to the barrier to be permeated are used in the pharmaceutically acceptable compositions. Such penetrants are known in the art.
  • compositions may take the form of, for example, lozenges, tablets, or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., pregelatinized maize starch, polyvinylpyrrolidone, or hydroxypropyl)
  • pharmaceutically acceptable excipients such as binding agents (e.g., pregelatinized maize starch, polyvinylpyrrolidone, or hydroxypropyl
  • methylcellulose methylcellulose
  • fillers e.g., lactose, micro crystalline cellulose, or calcium hydrogen phosphate
  • lubricants e.g., magnesium stearate, talc, or silica
  • disintegrants e.g., potato starch or sodium starch glycolate
  • wetting agents e.g., sodium lauryl sulfate.
  • the tablets can be coated by methods well known in the art with, for example, sugars, films, or enteric coatings.
  • compositions containing the compounds of the present technology or prodrug thereof in a form suitable for oral use may also include, for example, troches, lozenges, aqueous, or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs.
  • pharmaceutically acceptable compositions intended for oral use can be prepared according to any method known to the art for the manufacture of
  • compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents, and preserving agents in order to provide pharmaceutically elegant and palatable preparations.
  • Tablets contain the active ingredient (including drug and/or prodrug) in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients can be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents (e.g., corn starch or alginic acid); binding agents (e.g.
  • the tablets can be left uncoated or they can be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate can be employed. They may also be coated by the techniques described in the U.S. Pat. Nos. 4,256,108; 4,166,452; and 4,265,874 to form osmotic therapeutic tablets for control release.
  • the pharmaceutically acceptable compositions of the present technology may also be in the form of oil-in-water emulsions.
  • Liquid pharmaceutically acceptable compositions for oral administration may take the form of, for example, elixirs, solutions, syrups, or suspensions, or they can be presented as a dry product for constitution with water or other suitable vehicle before use.
  • Such liquid preparations can be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, cellulose derivatives, or hydrogenated edible fats);
  • emulsifying agents e.g., lecithin, or acacia
  • nonaqueous vehicles e.g., almond oil, oily esters, ethyl alcohol, cremophoreTM, or fractionated vegetable oils
  • preservatives e.g., methyl or propylphydroxybenzoates or sorbic acid
  • the preparations may also contain buffer salts, preservatives, flavoring, coloring, and sweetening agents as appropriate.
  • Preparations for oral administration can be suitably formulated to give controlled release or sustained release of the active compound, as is well known.
  • sustained release formulations (or sustained release pharmaceutically acceptable compositions) of the present technology are preferably in the form of a compressed tablet comprising an intimate mixture of compound of the present technology and a partially neutralized pH-dependent binder that controls the rate of compound dissolution in aqueous media across the range of pH in the stomach (typically approximately 2) and in the intestine (typically approximately about 5.5).
  • one or more pH-dependent binders can be chosen to control the dissolution profile of the sustained release pharmaceutically acceptable compositions so that such pharmaceutically acceptable compositions release compound slowly and continuously as the pharmaceutically acceptable compositions are passed through the stomach and gastrointestinal tract.
  • the pH-dependent binders suitable for use in the present technology are those which inhibit rapid release of drug from a tablet during its residence in the stomach (where the pH is-below about 4.5), and which promotes the release of a therapeutic amount of the compound of the present technology from the dosage form in the lower gastrointestinal tract (where the pH is generally greater than about 4.5).
  • enteric binders and coating agents have a desired pH dissolution properties.
  • the examples include phthalic acid derivatives such as the phthalic acid derivatives of vinyl polymers and copolymers, hydroxyalkylcelluloses, alkylcelluloses, cellulose acetates,
  • One or more pH-dependent binders present in the sustained release formulation of the present technology are in an amount ranging from about 1 to about 30 wt %, about 5 to about 12 wt % and about 10 wt %.
  • pH-independent binders may be in used in oral sustained release pharmaceutically acceptable compositions of the present technology.
  • the pH- independent binders can be present in the pharmaceutically acceptable compositions of the present technology in an amount ranging from about 1 to about 10 wt %, from about 1 to about 3 wt % and about 2 wt %.
  • sustained release pharmaceutically acceptable compositions of the present technology may also contain pharmaceutically acceptable excipients intimately admixed with the compound and the pH-dependent binder.
  • compositions may include, for example, pH-independent binders or film-forming agents such as hydroxypropyl methylcellulose,
  • poly(meth)acrylate esters starch, gelatin, sugars, carboxymethylcellulose, and the like.
  • Other useful pharmaceutical excipients include diluents such as lactose, mannitol, dry starch, microcrystalline cellulose and the like; surface active agents such as polyoxyethylene sorbitan esters, sorbitan esters and the like; and coloring agents and flavoring agents.
  • Lubricants such as talc and magnesium stearate
  • other tableting aids can also be optionally present.
  • the sustained release pharmaceutically acceptable compositions of the present technology have a compound of the present technology in the range of about 50% by weight to about 95% or more by weight, about 70% to about 90%> by weight; a pH-dependent binder content of between 5% and 40%, between 5% and 25%, and between 5% and 15%; with the remainder of the dosage form comprising pH- independent binders, fillers, and other optional excipients.
  • the pharmaceutically acceptable compositions may take the form of tablets or lozenges formulated in the conventional manner.
  • the active compound(s) can be formulated as solutions (for retention enemas), suppositories, or ointments containing conventional suppository bases such as cocoa butter or other glycerides.
  • the active compound(s) or prodrug(s) can be conveniently delivered in the form of an aerosol spray from pressurized packs or a nebulizer with the use of a suitable propellant (e.g., dichlorodifluoromethane, trichlorofluoromethane,
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane,
  • the dosage unit can be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges for use in an inhaler or insufflator can be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
  • the pharmaceutically acceptable compositions can be in the form of a sterile injectable aqueous or oleaginous suspension.
  • This suspension can be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent.
  • the acceptable vehicles and solvents that can be employed are water, Ringer's solution, and isotonic sodium chloride solution.
  • the compounds may also be administered in the form of suppositories for rectal or urethral administration of the drug.
  • compositions for topical use, creams, ointments, jellies, gels, solutions, suspensions, etc., containing the nanoparticles of the present technology, can be employed.
  • the compounds of the present technology can be formulated for topical administration with polyethylene glycol (PEG).
  • PEG polyethylene glycol
  • these formulations may optionally comprise additional pharmaceutically acceptable ingredients such as diluents, stabilizers, and/or adjuvants.
  • nanoparticles of the present technology include those well-known in the art, such as metered dose inhalers, liquid nebulizers, dry powder inhalers, sprayers, thermal vaporizers, and the like.
  • Other suitable technology for administration of particular nanoparticles of the present technology includes electrohydrodynamic aerosolizers.
  • electrohydrodynamic aerosolizers As those skilled in the art will recognize, the formulation of nanoparticles, the quantity of the formulation delivered, and the duration of administration of a single dose depend on the type of inhalation device employed as well as other factors.
  • aerosol delivery systems such as nebulizers, the frequency of administration and length of time for which the system is activated will depend mainly on the concentration of
  • nanoparticles in the aerosol For example, shorter periods of administration can be used at higher concentrations of nanoparticles in the nebulizer solution.
  • Devices such as metered dose inhalers can produce higher aerosol concentrations and can be operated for shorter periods to deliver the desired amount of nanoparticles in some embodiments.
  • Devices such as dry powder inhalers deliver active agent until a given charge of agent is expelled from the device. In this type of inhaler, the amount of nanoparticles in a given quantity of the powder determines the dose delivered in a single administration.
  • compositions of the nanoparticles of the present technology for administration from a dry powder inhaler may typically include a finely divided dry powder containing nanoparticles, but the powder can also include a bulking agent, buffer, carrier, excipient, another additive, or the like.
  • Additives can be included in such a dry powder composition of nanoparticles of the present technology, for example, to dilute the powder as required for delivery from the particular powder inhaler, to facilitate processing of the formulation, to provide advantageous powder properties to the formulation, to facilitate dispersion of the powder from the inhalation device, to stabilize the formulation (e.g., antioxidants or buffers), to provide taste to the formulation, or the like.
  • Typical additives include mono-, di-, and polysaccharides; sugar alcohols and other polyols, such as, for example, lactose, glucose, raffinose, melezitose, lactitol, maltitol, trehalose, sucrose, mannitol, starch, or combinations thereof; surfactants, such as sorbitols,
  • diphosphatidyl choline or lecithin; and the like.
  • the nanoparticle(s) or prodrug(s) of the present technology can be formulated as a depot preparation for administration by
  • the active ingredient can be formulated with suitable polymeric or hydrophobic materials (e.g., as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives (e.g., as a sparingly soluble salt).
  • suitable polymeric or hydrophobic materials e.g., as an emulsion in an acceptable oil
  • ion exchange resins e.g., as an emulsion in an acceptable oil
  • sparingly soluble derivatives e.g., as a sparingly soluble salt
  • transdermal delivery systems manufactured as an adhesive disc or patch which slowly releases the active nanoparticle(s) for percutaneous absorption can be used.
  • permeation enhancers can be used to facilitate transdermal penetration of the active nanoparticle(s).
  • Suitable transdermal patches are described in, for example, U.S. Patent No. 5,407,713.; U.S. Patent No. 5,352,456; U.S. Patent No.
  • Liposomes and emulsions are well-known examples of delivery vehicles that can be used to deliver active nanoparticle(s) or prodrug(s).
  • Certain organic solvents such as dimethylsulfoxide (DMSO) may also be employed, for example for topical administration, although usually at the cost of greater toxicity.
  • DMSO dimethylsulfoxide
  • compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active nanoparticle(s).
  • the pack may, for example, comprise metal or plastic foil, such as a blister pack.
  • the pack or dispenser device can be accompanied by instructions for administration.
  • the nanoparticles described herein, or compositions thereof, will generally be used in an amount effective to achieve the intended result, for example, in an amount effective to treat or prevent the particular condition being treated.
  • the nanoparticles can be administered therapeutically to achieve therapeutic benefit or prophylactically to achieve prophylactic benefit.
  • therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated and/or eradication or amelioration of one or more of the symptoms associated with the underlying disorder such that the patient reports an improvement in feeling or condition, notwithstanding that the patient may still be afflicted with the underlying disorder.
  • Therapeutic benefit also includes halting or slowing the progression of the disease, regardless of whether improvement is realized.
  • the amount of nanoparticle administered will depend upon a variety of factors, including, for example, the particular condition being treated, the mode of administration, the severity of the condition being treated, the age and weight of the patient, the bioavailability of the particular active nanoparticle. Determination of an effective dosage is well within the capabilities of those skilled in the art. As known by those of skill in the art, the preferred dosage of nanoparticles of the present technology will also depend on the age, weight, general health, and severity of the condition of the individual being treated. Dosage may also need to be tailored to the sex of the individual and/or the lung capacity of the individual, where administered by inhalation.
  • Dosage, and frequency of administration of the nanoparticles or prodrugs thereof, will also depend on whether the nanoparticles are formulated for treatment of acute episodes of a condition or for the prophylactic treatment of a disorder. A skilled practitioner will be able to determine the optimal dose for a particular individual.
  • the nanoparticle can be administered to a patient at risk of developing one of the previously described conditions.
  • prophylactic administration can be applied to avoid the onset of symptoms in a patient diagnosed with the underlying disorder.
  • Effective dosages can be estimated initially from in vitro assays.
  • an initial dosage for use in animals can be formulated to achieve a circulating blood or serum concentration of active nanoparticle that is at or above an IC 50 of the particular nanoparticle as measured in as in vitro assay.
  • Calculating dosages to achieve such circulating blood or serum concentrations taking into account the bioavailability of the particular nanoparticle is well within the capabilities of skilled artisans.
  • the reader is referred to Fingl & Woodbury, "General Principles,” GOODMAN AND GILMAN'S THE PHARMACEUTICAL BASIS OF THERAPEUTICS, Chapter 1, pp. 1-46, latest edition, Pergamon Press, and the references cited therein.
  • Initial dosages can also be estimated from in vivo data, such as animal models.
  • Certain animal models useful for testing the efficacy of nanoparticles to treat or prevent the various diseases described above are well-known in the art. Ordinarily skilled artisans can routinely adapt such information to determine dosages suitable for human administration.
  • Dosage amounts will typically be in the range of from about 0.0001 or about 0.001 or about 0.01 mg/kg/day to about 100 mg/kg/day, but can be higher or lower, depending upon, among other factors, the activity of the nanoparticle, its
  • Dosage amount and interval can be adjusted individually to provide levels in the organ system of interest of the nanoparticle(s) which are sufficient to maintain therapeutic or prophylactic effect.
  • the nanoparticles can be
  • the effective local concentration of active nanoparticle(s) may not be related to plasma concentration. Skilled artisans will be able to optimize effective local dosages without undue experimentation.
  • the nanoparticle(s) useful in the treatment methods of the present technology will provide therapeutic or prophylactic benefit without causing substantial toxicity. Toxicity of the nanoparticle(s) can be determined using standard pharmaceutical procedures. The dose ratio between toxic and therapeutic (or prophylactic) effect is the therapeutic index. In certain embodiments, the
  • nanoparticles(s) exhibit high therapeutic indices as pertinent to the disease treated.
  • kits for administration of the nanoparticles of the present technology, prodrug thereof, or pharmaceutical formulations comprising the nanoparticle may include a dosage amount of at least one nanoparticle or a composition comprising at least one nanoparticle, as disclosed herein.
  • Kits may further comprise suitable packaging and/or instructions for use of the nanoparticle.
  • Kits may also comprise a means for the delivery of the at least one nanoparticle or compositions comprising at least one nanoparticle of the present technology, such as an inhaler, spray dispenser (e.g., nasal spray), syringe for injection, or pressure pack for capsules, tablets, suppositories, or other device as described herein.
  • kits provide the nanoparticle and reagents to prepare a composition of the present technology for administration.
  • the composition can be in a dry or lyophilized form or in a solution, particularly a sterile solution.
  • the reagent may comprise a pharmaceutically acceptable diluent for preparing a liquid formulation.
  • the kit may contain a device for administration or for dispensing the compositions, including, but not limited to, syringe, pipette, transdermal patch, or inhalant.
  • kits may include other therapeutic nanoparticles for use in conjunction with the nanoparticles of the present technology described herein.
  • kits will include appropriate instructions for preparation and administration of the composition, side effects of the compositions, and any other relevant information.
  • the instructions can be in any suitable format, including, but not limited to, printed matter, videotape, computer readable disk, or optical disc.
  • the present technology provides a kit comprising a nanoparticle selected from the nanoparticles of the present technology or a salt or prodrug thereof, packaging, and instructions for use.
  • kits comprising the pharmaceutically acceptable composition comprising a nanoparticle selected from the nanoparticles of the present technology or a salt or prodrug thereof and at least one pharmaceutically acceptable excipient, diluent, preservative, stabilizer, or mixture thereof, packaging, and instructions for use.
  • kits for treating an individual who suffers from or is susceptible to the conditions described herein comprising a container comprising a dosage amount of a nanoparticle or composition of the present technology, as disclosed herein, and instructions for use.
  • the container can be any of those known in the art and appropriate for storage and delivery of oral, intravenous, topical, rectal, urethral, or inhaled formulations.
  • Kits may also be provided that contain sufficient dosages of the
  • nanoparticles or composition to provide effective treatment for an individual for an extended period such as a week, 2 weeks, 3, weeks, 4 weeks, 6 weeks, or 8 weeks or more.
  • the aqueous solution was acidified with dilute HC1 to pH 2 and 5x diafiltered with water using a 0.1 m TFF (tangential flow filtration) module with a 5 kDa MWCO membrane.
  • the solution was then concentrated to ⁇ 50 mL by TFF and lyophilized to afford dextran 20% succinate as a white solid (10.2 g).
  • ⁇ ⁇ NMR analysis confirmed that the product contained 0.2 equivalents of succinate per anhydroglucose unit (20% succinylation).
  • N,N-Dimethyl chitosan (1 g) was suspended in N-methyl pyrrolidone
  • 0-Carboxymethyl N,N,N-trimethyl chitosan 180 mg was dissolved in water (10 mL) and the solution adjusted to pH 5. Aminohexyl-VB12 (55 mg), ED AC (50 mg) and NHS (50 mg) were added and the solution was stirred for 20 hours maintained at pH 5.5. The mixture was subjected to centrifugal ultrafiltration in a 3 kDa Amicon-15 centrifugal filter and washed with water. The retentate was lyophilized to afford VB12-CM-TMC (133 mg) as a pale red solid.
  • a solution of recombinant human insulin in pH 7.4 phosphate buffer (11.2 mg/mL; 1.0 mL) was added to a solution of trimethyl chitosan chloride in water (6.7 mg/mL; 5.0 mL) and gently stirred for 30 min forming a light suspension.
  • the mixture was adjusted to pH 7.4 and a solution of VB12-carboxymethyl dextran of example 4 in pH 7.4 phosphate buffer (10.0 mg/mL; 0.1 mL) was added; stirring for 5 min yielded a pale red suspension of the VB12-PEC insulin nanoparticles (z-average diameter 112 nm) containing 47 IU per mL of insulin.
  • a solution of fish sperm DNA in pH 7.4 phosphate buffer (1.0 mg/mL; 0.5 mL) was added to a solution of trimethyl chitosan chloride in pH 7.4 phosphate buffer (0.1 mg/mL; 10.0 mL) and gently stirred for 30 min forming a light suspension of TMC DNA nanoparticles (z-average diameter 131 nm).
  • Example 13 Comparision of Blood Glucose Reduction in Rats by oral and s.c. insulin
  • mice Female Wistar rats (200 g) were housed at room temperature with 12 h light/dark cycle. All animals had ad libitum access to a standard chow diet and water except wherever indicated. The rats were allowed to acclimatize for a period of 7 days in the new environment before initiation of the experiment. After randomization into groups of 4, each rat was marked and followed individually throughout the study.
  • PA AAC oral x Dose sc x 100
  • PA > 80%
  • Example 14 Comparision of Blood hGH levels in Rats following oral and s.c. hGH dosing
  • mice Male Sprague-Dawley JVC-IDC rats each had a surgically implanted jugular vein cannula (JVC) and an intraduodenal cannula (IDC) to bypass the stomach. Rats weighing approximately 100 g (30-35 days old) were housed at room temperature with a 12 h light/dark cycle. All animals had ad libitum access to a standard chow diet and water. The rats were allowed to acclimatize for a period of 4 days in the new environment before initiation of the experiment. All animals were weighed and numbered by ear clip.
  • JVC jugular vein cannula
  • IDC intraduodenal cannula
  • a solution of recombinant human insulin in pH 7.4 phosphate buffer (4.0 mg/mL; 3.0 mL) was added to a solution of Cob-CMTMC (similar to example 8) in water (4.0 mg/mL; 3.0 mL) and gently stirred for 15 min forming a turbid pink suspension of VB12-TMC PEC insulin nanoparticles (z-average diameter 243 nm) containing 53 IU of insulin per mL.
  • Hypromellose phthalate HP-55 500 mg was dissolved in 0.1 M NaOH (20 mL) and the solution adjusted to pH 5.4 with dilute HC1. Aminohexyl-VB12 (50 mg) and NHS (15 mg) were added, followed by ED AC (24 mg), and the mixture was stirred for 72 hours at pH 5.8. The solution was subjected to 5 kDa MWCO TFF diafiltration with water (5x), concentrated by TFF and the retentate was lyophilized to afford VB12-hypromellose phthalate, Cob-HP (420 mg) as a pale red solid. UV-VIS spectrophotometric analysis revealed the product contained 2.6 % w/w of VB12.
  • a solution of recombinant human insulin in pH 7.4 phosphate buffer (5.0 mg/mL; 2.0 mL) was added to a solution of trimethyl chitosan TMC in pH 7.4 phosphate buffer (5.0 mg/mL; 4.0 mL).
  • a solution of Cob-HP in pH 7.4 phosphate buffer (5.0 mg/mL; 1.5 mL) was added to the mixture and gently stirred for 15 min forming a turbid pink suspension of VB12-HP-TMC PEC insulin nanoparticles (z-average diameter 548 nm). The suspension was lyophilized to afford VB12-HP- TMC nanoparticles containing 6.3 IU of insulin per mg.
  • CMTMC CobT-0
  • Cob-CMTMC CobT-7
  • Lipofectamine 2000 as positive control
  • siRNA Ambion Silencer Select Cat#4392420, Lot #AS00L5SB, ID 42755
  • MSI2 gene in serum-free media (RPMI1640, ATCC) for 30 minutes at RT.
  • K562 cells (ATCC, passage 12) were plated in U-bottom tissue culture 96- well plate 24 hour prior to transfection (5,000 cells per well; 180 volume per well).
  • RNAqueous® MAGMax-96 kit Total RNA from siRNA transfected cells was isolated using the RNAqueous® MAGMax- 96 kit (Ambion, Austin, TX). The purified, DNAse-treated RNA was reverse transcribed to make complementary DNA (cDNA) with random decamers using the RETROscript 3 ⁇ 4 Kit (Ambion, Austin, TX). Gene expression levels were determined by real-time PCR on the ABI Prism 7900 SDS real-time PCR machine (Applied Biosystems, Foster City, CA). The MSI2 data was collected via a Taqman Gene Expression Assay specific to amplification of the MSI2 mRNA
  • Fluorescence based real-time reverse transcription PC (qRT-PCR) is a standard tool used for quantification of mRNA levels. This technique has high throughput capabilities with both high sensitivity and specificity for the target of interest.
  • 18S rRNA was also amplified as an internal reference to adjust for well-to- well variances in the amount of starting template into the reverse transcription reaction (cat#Hs99999901_sl; Applied Biosystems).
  • the MSI2 values were corrected to the 18S values (internal reference), and the corrected values were then normalized to a sample transfected with a egative Control siRNA (Ambion, Austin, TX).
  • "% Remaining Expression Levels" were calculated as a percentage of gene expression relative to the negative control siRNA of similar trans fection volume. A value of 100% would represent an untreated sample. Values less than 100% exhibit the percent knockdown of the target.

Abstract

Disclosed herein are compositions and methods for a nanoparticle comprising one or more synthetic or natural polymers comprising one or more charged and/or ionisable groups, a therapeutic agent comprising one or more charged and/or ionisable groups of the opposite charge to the charge of the polymers, and a vitamin B12 covalently linked to the nanoparticle via an optional linker group.

Description

A NANOSTRUCTURES CONTAINING VITAMIN B12 FOR FACILITATED DELIVERY OF DRUGS ACROSS BIOLOGICAL BARRIERS
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit under 35 U.S.C. § 119(e) of U.S.
Provisional Serial No. 61/325,174, filed April 16, 2010, the contents of which is incorporated by reference in its entirety into the current disclosure.
TECHNICAL FIELD
[0002] The invention relates to the delivery of pharmaceutically-active compounds such as small-molecule drugs, proteins and peptides across biological barriers using naturally-occuring vitamin transport systems. In one aspect, the invention relates to the delivery of pharmaceutically-active compounds utilizing vitamin B12 transport systems with the protection of the active during transport by incorporation in nanostructures, such as, but not limited to, nanostructures containing surface-linked vitamin B12. In some embodiments, the nanostructures are made from charged polymers. The invention also relates to processes for preparing the nanostructures, pharmaceutical compositions containing same and methods of drug delivery and treatment of disease involving the nanostructures.
BACKGROUND OF THE INVENTION
[0003] While treatment of disease using pharmaceutically-active compounds is commonplace, the development of medications is challenged by the the need to deliver the drug conveniently to the sites of action in sufficient quantities to achieve the desired pharmacological effect. A convenient route of drug administration is oral delivery. It may be preferred by patients as it is non-invasive and by physicians for patient compliance. Yet this route may be inaccessible for many pharmaceutically- active compounds either because these compounds are broken down in the gasterointestinal tract or fail to be absorbed. Similarly, drugs that may show significant promise in early testing can fail because the compounds may not reach their intended sites of action for failure to cross biological membranes. For example, in cancer chemotherapy, it may often be necessary to dose patients with high levels of cytotoxic drugs in order to achieve a meaningful therapeutic effect which may also result in damage to normal cells, resulting in significant adverse side-effects. It may be desirable to alter the natural biodistribution of cytotoxic compounds so that more of the drug is delivered to tumor cells, and less to normal tissues. Monoclonal antibodies to tumor-specific antigens have been used as target cytotoxic agents to tumors so as to improve upon the therapeutic index (a ratio of a drug's beneficial effects compared with its adverse side-effects). The use of monoclonal antibodies, however, may generate other issues, such as immunogenicity, whereby the patient's immune systems may develop an immune response to the antibody-drug conjugate. It is therefore desirable to have new drug delivery systems that are both safe and effective, and which can facilitate the delivery of drugs across biological membrane such as the gut wall (in oral drug delivery) and cell membranes (in the treatment of disease). This invention satisfies this need and provides related advantages as well.
SUMMARY OF THE INVENTION
[0004] Surprisingly it has been found by the Applicants that polyelectrolyte complexes (PEC) capable of drug delivery can be formed utilizing a small, hydrophilic vitamin B12 molecule as the targeting group. With drug-loading, the PECs of this invention are able to provide for either or both oral drug delivery of unmodified pharmaceutically-active compounds and other therapeutic agent through transfer from the intestinal lumen into the bloodstream, or targeted to diseased cells in the body that over express the receptors that facilitate vitamin B12 cell uptake.
[0005] In one aspect, the present invention relates to one or more nanoparticle(s) formed by bringing together in a suitable solvent a synthetic or natural polymer comprising multiple charged and/or ionisable groups, such as, positively charged or negatively charged, with a therapeutic agent which has one or more charged or ionisable groups of the opposite charge to the charge of the polymer, and optionally other components which either assist in nanoparticle formation or in the placement of a vitamin B12 on the surface of the nanoparticle. The polymer and the therapeutically active agent can form a PEC, either alone or in combination with the other components, e.g., the aforementioned components. [0006] In one aspect, the present invention relates to nanoparticles formed by bringing together in a suitable solvent two synthetic or natural polymers in which one polymer comprises multiple charged and/or ionisable groups of net positive charge and the other polymer comprises one or multiple charged and/or ionisable groups of net negative charge, together with a therapeutic agent which may or may not possess charged or ionisable groups, and optionally other components which either assist in nanoparticle formation or in the placement of a vitamin B12 on the surface of the nanoparticle. The two polymers can form a PEC, either alone or in combination with the other aforementioned components, and the therapeutically active component is trapped in the PEC during PEC formation.
[0007] In one aspect, the vitamin B12 is introduced prior to nanoparticle formation either by covalent attachment to the polymer, to the therapeutic agent, and/or to one of the optional additional components.
[0008] Figure 1 depicts a structure of vitamin B12, in which represents a monodentate axial ligand as defined later. In either of the above embodiments of the present invention, attachment of a vitamin B12 can occur directly to one of the above mentioned components of the nanoparticle or via a suitable linker. Vitamin B12 attachment can occur via either the 2' or 5 '-oxygen atoms on the ribose unit of vitamin B12 (as exemplified by U.S. Pat. No. 6,150,341), or via conversion of one or more of the amide groups to carboxyl and subsequent addition of a linker group (for example, Waibel et al, Cancer Res., 2008, 68, 2904-2911) or by replacement of the axial ligand (R) on the cobalt atom of vitamin B12 with a bifunction ligand; a compound that can bind to cobalt, as exemplified in U.S. Pat. No. 6,262,253.
[0009] In some embodiments, nanoparticles are formed by bringing together the components of the nanoparticle in an aqueous environment, although other solvent systems known in the art may also be used. The nanoparticles formed may be either soluble or insoluble in the solvent system. Nanoparticles can be isolated by techniques known in the art. For example, soluble nanoparticles can be isolated by precipitation with a cosolvent or by removal of solvent (e.g. lyophilization or spray drying) optionally preceeded by a purification method such as tangential flow filtration (TFF) or centrifugal ultrafiltration. Insoluble nanoparticles might be isolated by centrifugation or filtration, also optionally preceeded by a purification method such as TFF.
[0010] The solid nanoparticles formed and isolated as described above, can be formulated for human or veterinary administration by standard methods. For example, optionally with suitable excipients, the nanoparticles can be formulated into tablets or capsules for oral administration, as lyophilized or dried formulations in vials for subsequent reconstitution with an injection vehicle and administration to humans or animals by injection, or as solutions or suspensions for administration to humans or animals by injection.
[0011] The pharmaceutical formulations of the nanoparticles of this invention are useful for oral drug delivery and/or disease targeted delivery of a wide variety of therapeutic agents, including, but not limited to, small and large synthetic molecules, proteins, peptides, glycoproteins, humanized and non-humanized monoclonal antibodies and therapeutically relevant fragments thereof, and agents for effecting the delivery of polynucleotides alone or in combination with a gene delivery vector. The polynucleotides, include for example those which are, or that encode RNA
interference (RNAi) such as siRNA, dsRNA, mRNA and antisense RNA, as well DNA, such as in gene therapy applications.
[0012] The pharmaceutical formulations of the nanoparticles of this invention are useful to treat a wide variety of diseases including, but not limited to cancer, autoimmune conditions, endocrine disorders, diabetes, genetic conditions,
chromosome conditions, viral infections, bacterial infections, parasitic infections, mitochondrial diseases, sexually transmitted diseases, immune disorders, balance disorders, pain, systemic disorders, blood conditions, blood vessel conditions, nerve conditions, and conditions of muscles, heart and other organs.
[0013] In one aspect, there is provided a nanoparticle comprising, or alternatively consisting essentially of or alternatively consisting of one or more synthetic or natural polymers comprising one or more charged and/or ionisable groups, a therapeutic agent comprising one or more charged and/or ionisable groups of the opposite charge to the charge of the polymers, and, a vitamin B12 covalently linked to the
nanoparticle via an optional linker group. [0014] In some embodiments, the one or more charged and/or ionisable groups of the one or more synthetic or natural polymers are positive charge or negative charge.
[0015] In one aspect, there is provided a nanoparticle comprising, or alternatively consisting essentially of or alternatively consisting of one or more synthetic or natural polymers comprising one or more charged and/or ionisable groups which are positively charged, one or more synthetic or natural polymers comprising one or more charged and/or ionisable groups which are negatively charged, a therapeutic agent, and a vitamin B12 covalently linked to the nanoparticle via an optional linker group.
[0016] In some embodiments, the vitamin B12 is attached to one or more of the charged and/or ionisable groups of the one or more synthetic or natural polymers.
[0017] In some embodiments, the vitamin B12 is attached to the therapeutic agent.
[0018] In some embodiments, the nanoparticle of the above noted aspects further comprises one or more of components selected from the group consisting of polyethylene glycol (PEG), PEG block copolymers, polyacrylic, polymethacrylic, polyacrylamide, polymethacrylamide, synthetic polymer, polysaccharide, surfactant, and metal ions.
[0019] In some embodiments, the vitamin B12 is attached to one or more of the components.
[0020] In some embodiments, an average nanoparticle diameter is in a range of about 20 nm to about 800 nm.
[0021] In some embodiments, the nanoparticle is configured for oral administration in a subject.
[0022] In some embodiments, the nanoparticle is configured for administration by injection to a subject. [0023] In some embodiments, the synthetic or natural polymer is selected from the group consisting of poly-Y-glutamic acid (γ-PGA), succinylated and/or
carboxymethylated polysaccharide, poly(acrylic acid), poly(p-styrene-sulfonic acid sodium salt), poly(sulfopropylacrylate), negatively-charged polypeptide, chitosan derivatives, polyethyleneimine, positively-charged polypeptide, pharmaceutically- approved polymeric excipients that carry a net negative charge at physiologically- relevant pH levels, and pharmaceutically-approved polymeric excipients that carry a net positive charge at physiologically-relevent pH levels.
[0024] In some embodiments, the synthetic or natural polymer is selected from the group consisting of chitosan derivatives, polyethyleneimine, positively-charged polypeptide, and pharmaceutically-approved polymeric excipients that carry a net positive charge at physiologically-relevent pH levels.
[0025] In some embodiments, the synthetic or natural polymer is selected from the group consisting of poly-Y-glutamic acid (γ-PGA), succinylated and/or
carboxymethylated polysaccharide, poly(acrylic acid), poly(p-styrene-sulfonic acid sodium salt), poly(sulfopropylacrylate), negatively-charged polypeptide, and pharmaceutically-approved polymeric excipients that carry a net negative charge at physiologically-relevent pH levels.
[0026] In some embodiments, the succinylated and/or carboxymethylated polysaccharide as noted above is dextran, cellulose, or starch.
[0027] In some embodiments, the negatively-charged polypeptide as noted above is polyglutamic acid.
[0028] In some embodiments, the positively-charged polypeptide as noted above is polylysine.
[0029] In some embodiments, an axial ligand substituent on a cobalt atom of vitamin B 12 is a chemical entity such as cyano (-CN), methyl (CH3), hydro xyl (-OH) or nitrosyl (-NO).
[0030] In some embodiments, the therapeutic agent is selected from the group consisting of a small or large synthetic or semi-synthetic molecule, protein, peptide, glycoprotein, nucleoside, nucleotide, humanized monoclonal antibody, non- humanized monoclonal antibody, therapeutically relevant fragments of humanized and/or non-humanized monoclonal antibody, and agents for effecting RNA interference (RNAi) such as dsRNA, miRNA, siRNA and antisense RNA. [0031] As used herein, the term "polynucleotides" includes deoxyribonucleic acid (DNA), and, where appropriate, ribonucleic acid (RNA). The term should also be understood to include, as equivalents, derivatives, variants and analogs of either RNA or DNA made from nucleotide analogs, and, as applicable to the embodiment being described, single (sense or antisense) and double-stranded polynucleotides.
Deoxyribonucleotides include deoxyadenosine, deoxycytidine, deoxyguanosine, and deoxythymidine. For purposes of clarity, when referring herein to a nucleotide of a nucleic acid, which can be DNA or an RNA, the terms "adenosine", "cytidine", "guanosine", and "thymidine" are used. It is understood that if the nucleic acid is RNA, a nucleotide having a uracil base is uridine.
[0032] The terms "polynucleotide" and "oligonucleotide" are used interchangeably and refer to a polymeric form of nucleotides of any length, either
deoxyribonucleotides or ribonucleotides or analogs thereof. Polynucleotides can have any three-dimensional structure and may perform any function, known or unknown. The following are non-limiting examples of polynucleotides: a gene or gene fragment (for example, a probe, primer, EST or SAGE tag), exons, introns, messenger RNA (mRNA), transfer RNA, ribosomal RNA, ribozymes, cDNA, dsRNA, siRNA, miRNA, recombinant polynucleotides, branched polynucleotides, plasmids, vectors, isolated DNA of any sequence, isolated RNA of any sequence, nucleic acid probes and primers. A polynucleotide can comprise modified nucleotides, such as methylated nucleotides and nucleotide analogs. If present, modifications to the nucleotide structure can be imparted before or after assembly of the polynucleotide. The sequence of nucleotides can be interrupted by non-nucleotide components. A polynucleotide can be further modified after polymerization, such as by conjugation with a labeling component. The term also refers to both double- and single-stranded molecules. Unless otherwise specified or required, any embodiment of this invention that is a polynucleotide encompasses both the double-stranded form and each of two complementary single-stranded forms known or predicted to make up the
double-stranded form.
[0033] A polynucleotide is composed of a specific sequence of four nucleotide bases: adenine (A); cytosine (C); guanine (G); thymine (T); and uracil (U) for thymine when the polynucleotide is RNA. Thus, the term "polynucleotide sequence" is the alphabetical representation of a polynucleotide molecule. This alphabetical representation can be input into databases in a computer having a central processing unit and used for bioinformatics applications such as functional genomics and homology searching. The term "polymorphism" refers to the coexistence of more than one form of a gene or portion thereof. A portion of a gene of which there are at least two different forms, i.e., two different nucleotide sequences, is referred to as a "polymorphic region of a gene". A polymorphic region can be a single nucleotide, the identity of which differs in different alleles.
[0034] As used herein, the term "carrier" encompasses any of the standard carriers, such as a phosphate buffered saline solution, buffers, water, and emulsions, such as an oil/water or water/oil emulsion, and various types of wetting agents. The
compositions also can include stabilizers and preservatives. For examples of carriers, stabilizers and adjuvants, see Sambrook and Russell (2001), supra. Those skilled in the art will know many other suitable carriers for binding polynucleotides, or will be able to ascertain the same by use of routine experimentation. In one aspect of the invention, the carrier is a buffered solution such as, but not limited to, a PCR buffer solution.
[0035] A "gene delivery vehicle" is defined as any molecule that can carry inserted polynucleotides into a host cell. Examples of gene delivery vehicles are liposomes, biocompatible polymers, including natural polymers and synthetic polymers;
lipoproteins; polypeptides; polysaccharides; lipopolysaccharides; artificial viral envelopes; metal particles; and bacteria, or viruses, such as baculovirus, adenovirus and retrovirus, bacteriophage, cosmid, plasmid, fungal vectors and other
recombination vehicles typically used in the art which have been described for expression in a variety of eukaryotic and prokaryotic hosts, and may be used for gene therapy as well as for simple protein expression.
[0036] "Gene delivery," "gene transfer," and the like as used herein, are terms referring to the introduction of an exogenous polynucleotide (sometimes referred to as a "transgene") into a host cell, irrespective of the method used for the introduction. Such methods include a variety of well-known techniques such as vector-mediated gene transfer (by, e.g., viral infection, sometimes called transduction), transfection, transformation or various other protein-based or lipid-based gene delivery complexes) as well as techniques facilitating the delivery of "naked" polynucleotides (such as electroporation, "gene gun" delivery and various other techniques used for the introduction of polynucleotides). Unless otherwise specified, the term transfected, transduced or transformed may be used interchangeably herein to indicate the presence of exogenous polynucleotides or the expressed polypeptide therefrom in a cell. The introduced polynucleotide may be stably or transiently maintained in the host cell. Stable maintenance typically requires that the introduced polynucleotide either contains an origin of replication compatible with the host cell or integrates into a replicon of the host cell such as an extrachromosomal replicon (e.g., a plasmid) or a nuclear or mitochondrial chromosome. A number of vectors are known to be capable of mediating transfer of genes to mammalian cells, as is known in the art and described herein.
[0037] "RNA interference" (RNAi) refers to sequence-specific or gene specific suppression of gene expression (protein synthesis) that is mediated by short interfering RNA (siRNA).
[0038] "Short interfering RNA" (siRNA) refers to double-stranded RNA molecules (dsRNA), generally, from about 10 to about 30 nucleotides in length that are capable of mediating RNA interference (RNAi), or 11 nucleotides in length, 12 nucleotides in length, 13 nucleotides in length, 14 nucleotides in length, 15 nucleotides in length, 16 nucleotides in length, 17 nucleotides in length, 18 nucleotides in length, 19 nucleotides in length, 20 nucleotides in length, 21 nucleotides in length, 22 nucleotides in length, 23 nucleotides in length, 24 nucleotides in length, 25 nucleotides in length, 26 nucleotides in length, 27 nucleotides in length, 28 nucleotides in length, or 29 nucleotides in length. As used herein, the term siRNA includes short hairpin RNAs (shRNAs). A siRNA directed to a gene or the mRNA of a gene may be a siRNA that recognizes the mRNA of the gene and directs a RNA- induced silencing complex (RISC) to the mRNA, leading to degradation of the mRNA. A siRNA directed to a gene or the mRNA of a gene may also be a siRNA that recognizes the mRNA and inhibits translation of the mRNA. [0039] "Double stranded RNA" (dsRNA) refer to double stranded RNA molecules that may be of any length and may be cleaved intracellularly into smaller RNA molecules, such as siRNA. In cells that have a competent interferon response, longer dsRNA, such as those longer than about 30 base pair in length, may trigger the interferon response. In other cells that do not have a competent interferon response, dsRNA may be used to trigger specific RNAi.
[0040] A siRNA can be designed following procedures known in the art. See, e.g., Dykxhoorn, D.M. and Lieberman, J. (2006) "Running Interference: Prospects and Obstacles to Using Small Interfering RNAs as Small Molecule Drugs," Annu. Rev. Biomed. Eng. 8:377-402; Dykxhoorn, D.M. et al. (2006) "The silent treatment:
siRNAs as small molecule drugs," Gene Therapy, 13:541-52; Aagaard, L. and Rossi, J.J. (2007) "RNAi therapeutics: Principles, prospects and challenges," Adv. Drug Delivery Rev. 59:75-86; de Fougerolles, A. et al. (2007) "Interfering with disease: a progress report on siRNA-based therapeutics," Nature Reviews Drug Discovery 6:443-53; Krueger, U. et al. (2007) "Insights into effective RNAi gained from large- scale siRNA validation screening," Oligonucleotides 17:237-250; U.S. Patent Application Publication No.: 2008/0188430; and U.S. Patent Application Publication No.:2008/0249055.
[0041] siRNAs can be made with methods known in the art. See, e.g., Dykxhoorn, D.M. and Lieberman, J. (2006) "Running Interference: Prospects and Obstacles to Using Small Interfering RNAs as Small Molecule Drugs," Annu. Rev. Biomed. Eng. 8:377-402; Dykxhoorn, D.M. et al. (2006) "The silent treatment: siRNAs as small molecule drugs," Gene Therapy, 13:541-52; Aagaard, L. and Rossi, J.J. (2007) "RNAi therapeutics: Principles, prospects and challenges," Adv. Drug Delivery Rev. 59:75- 86; de Fougerolles, A. et al. (2007) "Interfering with disease: a progress report on siRNA-based therapeutics," Nature Reviews Drug Discovery 6:443-53; Krueger, U. et al. (2007) "Insights into effective RNAi gained from large-scale siRNA validation screening," Oligonucleotides 17:237-250; U.S. Patent Application Publication No.: 2008/0188430; and U.S. Patent Application Publication No.: 2008/0249055.
[0042] A siRNA may be chemically modified to increase its stability and safety. See, e.g. Dykxhoorn, D.M. and Lieberman, J. (2006) "Running Interference: Prospects and Obstacles to Using Small Interfering RNAs as Small Molecule Drugs," Annu. Rev. Biomed. Eng. 8:377-402 and U.S. Patent Application Publication No.: 2008/0249055.
[0043] microRNA or miRNA are single-stranded RNA molecules of 21-23 nucleotides in length, which regulate gene expression. miRNAs are encoded by genes from whose DNA they are transcribed but miRNAs are not translated into protein (non-coding RNA); instead each primary transcript (a pri-miRNA) is processed into a short stem-loop structure called a pre-miRNA and finally into a functional miRNA. Mature miRNA molecules are partially complementary to one or more messenger RNA (mRNA) molecules, and their main function is to down-regulate gene expression.
[0044] A siRNA vector, dsRNA vector or miRNA vector as used herein, refers to a plasmid or viral vector comprising a promoter regulating expression of the RNA. "siRNA promoters" or promoters that regulate expression of siRNA, dsRNA, or miRNA are known in the art, e.g., a U6 promoter as described in Miyagishi and Taira (2002) Nature Biotech. 20:497-500, and a HI promoter as described in Brummelkamp et al. (2002) Science 296:550-3.
[0045] In some embodiments, the therapeutic agent is selected from the group consisting of analgesic, antiallergenic, antianginal agent, antiarrythmic drug, antibiotic, anticoagulant, antidementia drug, antidepressant, antidiabetic,
antihistamine, antihypertensive, anti-inflammatory, antineoplastic agent, antiparasitic, antipyretic, antiretroviral drug, antiulcerative agent, antiviral agent, cardiovascular drug, cholesterol-lowering agent, CNS active drug, a hormone, growth hormone inhibitor, growth hormone, hematopoietic drug, hemostatic, hypotensive diuretic, keratolytic, therapeutic for osteoporosis, vaccine, vasoconstrictor, and vasodilator.
[0046] In one aspect, there is provided a process for preparing a nanoparticle composition comprising the nanoparticle of any of the above recited aspects and embodiments, comprising, or alternatively consisting essentially of or alternatively consisting of combining the one or more synthetic or natural polymers, the therapeutic agent, and the vitamin B 12, in a suitable solvent, and isolating, purifying and/or drying the nanoparticles. In some embodiments, the solvent is > 50% water. [0047] In another aspect, there is provided a process for preparing a nanoparticle composition comprising the nanoparticle of any of the above recited aspects and embodiments, comprising, or alternatively consisting essentially of or alternatively consisting of mixing two immiscible solvents and a surfactant to produce an emulsion, optionally cross-linking the nanoparticles, and isolating, purifying, and/or drying resultant nanoparticles.
[0048] In some embodiments, the nanoparticles are isolated by solvent evaporation
[0049] In some embodiments, the nanoparticles are isolated by filtration or centrifugation
[0050] In some embodiments, the nanoparticles are isolated by addition of a cosolvent followed by filtration or centrifugation.
[0051] In some embodiments, the purifying step is effected by washing the nanoparticles with a suitable solvent.
[0052] In some embodiments, the above recited aspects further comprise modifying the nanoparticles to effect cross-linking of the components of the nanoparticle.
[0053] In some embodiments, the above recited aspects further comprise modifying the nanoparticles to add a vitamin B12 analog to a surface of the nanoparticle by physical or covalent attachment.
[0054] In some embodiments, the above recited aspects further comprise modifying the nanoparticles to substitue an axial ligand on a one or more cobalt atoms of attached vitamin B12 with replacement axial ligands, non-limiting examples of which are provided herein.
[0055] In another aspect, there is provided a pharmaceutical composition comprising the nanoparticle of the above recited aspects, and a pharmaceutically- acceptable excipient.
[0056] In some embodiments, the composition is formulated as a tablet, a capsule, or a liquid. [0057] In some embodiments, the composition is formulated as a lyophilized powder in a container for subsequent re-suspension or dissolution of the
pharmaceutical composition in a pharmaceutically-acceptable injection vehicle.
[0058] In some embodiments, the composition is formulated as a suspension or solution in a pharmaceutically-acceptable injection vehicle.
[0059] In another aspect, there is provided a method for treating a subject, comprising, or alternatively consisting essentially of, or alternatively consisting of, administring an effective amount of the nanoparticle of any of the above recited aspects or the pharmaceutical composition of any of the above recited aspects.
[0060] In some embodiments, the therapeutic agent is an antidiabetic agent.
[0061] In some embodiments, the therapeutic agent is a hormone.
[0062] In some embodiments, the therapeutic agent is an antineoplastic agent.
BRIEF DESCRIPTION OF THE FIGURES
[0063] Figure 1 depicts a structure of vitamin B12.
[0064] Figure 2 depicts three VB12 drug delivery formats including nanoparticles.
[0065] Figure 3 depicts a graph showing plasma hGH vs time.
[0066] Figure 4 shows a reduction of MSI2 gene expression. K562 cells were transfected with 50 nM siRNA targeting MSI2 gene or 50 nM Negative Control siRNA. Forty-eight hours post-transfection, the cells were harvested and analyzed by real-time RT-PCR for MSI2 and 18S rRNA levels. 18S rRNA levels were used to normalize MSI2 expression. "Remaining mRNA (%)" was calculated as a percentage of MSI2 mRNA levels in K562 cells transfected with MSI2 siRNA relative to K562 cells transfected with non-silencing siRNA (Negative Control siRNA). Data are presented as means ±SD.
[0067] Figure 5 shows 18S rRNA levels. Level of expression of ribosomal RNA (18S) levels is directly associated with number of cells in the sample. Data demonstrate that Lipofectamine 2000 induces significant cytotoxicity at 3 μΐ^ per well (200 μΐ^ total volume) and higher volumes. In contrast, CobT-0 and CobT-7 show no toxicity at volumes up to 30 μΐ^ (5 mg/ml). Bars on the right at each of the noted comparisons show siMS12. Bars on the left show siNC.
DETAILED DESCRIPTION OF THE INVENTION
[0068] All technical and patent publications cited herein are incorporated herein by reference in their entirety.
[0069] All numerical designations, e.g., pH, temperature, time, concentration, and molecular weight, including ranges, are approximations which are varied ( + ) or ( - ) by increments of 0.1 or 1.0, as appropriate. It is to be understood, although not always explicitly stated that all numerical designations are preceded by the term "about". It also is to be understood, although not always explicitly stated, that the reagents described herein are merely exemplary and that equivalents of such are known in the art.
[0070] As used in the specification and claims, the singular form "a", "an" and "the" include plural references unless the context clearly dictates otherwise.
[0071] "Comprising" refers to compounds, compositions and methods including the recited elements, but not exclude others. "Consisting essentially of," when used to define compounds, compositions or methods, shall mean excluding other elements that would materially affect the basic and novel characteristics of the claimed technology. "Consisting of," shall mean excluding any element, step, or ingredient not specified in the claim. Embodiments defined by each of these transition terms are within the scope of this technology.
[0072] As used herein, "nanoparticle" or "nanostructure" refers a microscopic particle less than about 1 micron in diameter. In some embodiments, the nanoparticles range in size from about 1 nm to about 1 ,000 nm diameter, or alternatively between about 10 nm to about 1000 nm, or alternatively between about 10 nm to about 900 nm, or alternatively between about 10 nm to about 800 nm, or alternatively between about 10 nm to about 700 nm, or alternatively between about 10 nm to about 600 nm, or alternatively between about 10 nm to about 500 nm, or alternatively between about 20 nm to about 1000, or alternatively between about 20 nm to about 800 nm, or alternatively between about 20 nm to about 700 nm, or alternatively between about 20 nm to about 600 nm, or alternatively between about 20 nm to about 500 nm; or alternatively between about 30 nm to about 1000 nm, or alternatively between about 30 nm to about 900 nm, or alternatively between about 30 nm to about 800 nm, or alternatively between about 30 nm to about 700 nm, or alternatively between about 100 nm to about 900 nm, or alternatively between about 200 nm to about 1000 nm, or alternatively between about 300 nm to about 1000 nm, or alternatively between about 400 nm to about 1000 nm, or alternatively between about 500 nm to about 1000 nm; or alternatively between about 600 nm to about 1000 nm; or alternatively between about 700 nm to about 1000 nm; or alternatively between about 800 nm to about 1000 nm; or alternatively between about 900 nm to about 1000 nm; or alternatively between about 100 nm to about 300 nm; or alternatively between about 200 nm to about 600 nm; or alternatively between about 300 nm to about 600 nm; or
alternatively between about 500 nm to about 800 nm.
[0073] As used herein, "polymer" refers to a naturally-occurring, synthetic or semisynthetic large molecule (macromolecule) typically composed of repeating structural units connected by covalent chemical bonds. Polymers useful for the implementation of this invention have molecular weights in the range of 1 to 5000 kDa.
[0074] As used herein, "random copolymer" refers to a polymer comprising two or more repeating structural units in which the sequence of the individual repeating structural units is random and not predetermined or defined.
[0075] As used herein, "block copolymer" refers to a polymer comprising two or more repeating structural units in which individual repeating structural units are connected to each other forming identifyable blocks of repeating structural units within the complete polymer strand.
[0076] As used herein, "charged group" refers to a chemical functional group that is fully ionized resulting in that group having either a positive or a negative charge, or possibly multiple positive or multiple negative charges. Polymers could have multiple charged groups either as components of the polymer chain, and/or as attachments to the polymer, either direct attachment or by way of a linker. Polymer charged groups may be either naturally-occuring or synthetic. A charged group may be part of a therapeutically active compound, either as an intrinsic component of that compound or as a synthetic analog of the therapeutically active compound, for example a prodrug.
[0077] As used herein, "ionisable group" refers to a chemical functional group that is partially ionized at or close to physiological pH resulting in that group having either a partial positive or a partial negative charge. The charge of an ionisable group will vary with pH. Polymers could have multiple ionisable groups either as components of the polymer chain, and/or as attachments to the polymer, either direct attachment or by way of a linker. Polymer ionisable groups may be either naturally-occuring or synthetic. A ionisable group may be part of a therapeutically active compound, either as an intrinsic component of that compound or as a synthetic analog of the
therapeutically active compound, for example a prodrug.
[0078] As used herein, "polyelectrolyte complex" or "PEC" refers to a three- dimensional structure resulting from the formation of multiple ionic bonds between two or more compounds having chemical functional groups that are charged and/or ionisable wherein at least one compound possesses a net negative charge and at least one compound has a net positive charge, and at least one compound preferentially is a polymer. The diameter of PECs can typically range from 1 nm to several microns, with average particle size and particle size distribution controlled by the chemical and physical nature of the constituent components and method of preparation. PECs can be water soluble (i.e. suspension of nanoparticles in water results in a clear, transparent liquid) or insoluble (i.e. suspension of nanoparticles in water results in a cloudy liquid). PEC nanoparticles typically can range in size from about 1 nm to about 1,000 nm diameter, or alternatively about 5 nm to about 400 nm or alternatively about 10 nm to about 300 nm.
[0079] In some embodiments, the PECs of the invention range in size from about 1 nm to about 1,000 nm diameter, or alternatively between about 10 nm to about 1000 nm, or alternatively between about 10 nm to about 900 nm, or alternatively between about 10 nm to about 800 nm, or alternatively between about 10 nm to about 700 nm, or alternatively between about 10 nm to about 600 nm, or alternatively between about 10 nm to about 500 nm, or alternatively between about 20 nm to about 1000, or alternatively between about 20 nm to about 800 nm, or alternatively between about 20 nm to about 700 nm, or alternatively between about 20 nm to about 600 nm, or alternatively between about 20 nm to about 500 nm; or alternatively between about 20 nm to about 400 nm; or alternatively between about 20 nm to about 200 nm; or alternatively between about 30 nm to about 1000 nm, or alternatively between about 30 nm to about 900 nm, or alternatively between about 30 nm to about 800 nm, or alternatively between about 30 nm to about 700 nm, or alternatively between about 100 nm to about 300 nm, or alternatively between about 100 nm to about 500 nm, or alternatively between about 100 nm to about 900 nm, or alternatively between about 200 nm to about 1000 nm, or alternatively between about 300 nm to about 1000 nm, or alternatively between about 400 nm to about 1000 nm, or alternatively between about 500 nm to about 1000 nm; or alternatively between about 600 nm to about 1000 nm; or alternatively between about 700 nm to about 1000 nm; or alternatively between about 800 nm to about 1000 nm; or alternatively between about 900 nm to about 1000 nm; or alternatively between about 100 nm to about 300 nm; or alternatively between about 200 nm to about 600 nm; or alternatively between about 300 nm to about 600 nm; or alternatively between about 500 nm to about 800 nm.
[0080] As used herein, "alkyl" refers to a saturated (containing no multiple carbon- carbon bonds) aliphatic (containing no delocalized π-electron system), hydrocarbon containing, if otherwise unsubstituted, only carbon and hydrogen atoms. The designation (nlC- n2C)alkyl, wherein nl and n2 are integers from one to 6, refers to straight or branched chain alkyl groups comprising from nl to and including n2 carbon atoms. An alkyl group herein may be optionally substituted with one or more entities selected from the group consisting of halo, hydroxy, alkoxy, aryloxy, carbonyl, nitro, cyano, carboxyl and alkoxycarbonyl.
[0081] As used herein, "linker" refers to a group of atoms that is used to couple a polymeric backbone to another function or group to spatially separate the two entities. Thus, a linker of this invention has an essentially longitudinal axis, that is, it is essentially linear rather than highly branched or clumped, although the structure will, of course, not be exactly linear due to the angular constraints placed on the structure by required bond angles between covalently bonded atoms. Examples of linkers include, but are not limited to, straight and branced alkyl and alkenyl groups containing functional groups such as carboxyl, amino, hydroxyl, and thiol, through which covalent bonds can be formed to connect the linker to the polymer and to other components. A preferred linker is a short peptide chain ( H-[NHCH -CO]n-OH) where n is 1-20, or alternatively from 1-18, or alternatively from 1-16, or alternatively from 1-14, or alternatively from 1-12, or alternatively from 2-14, or alternatively from 2-12, or alternatively from 3-20, or alternatively from 4-18, or alternatively from 5- 20, or alternatively from 5-18, and R is the same or different for each of the n amino acids, and is one of the 22 side groups known to be present in natural amino acids. A peptide linker can be incorporated into the polymer compound by one of the peptide condensation reactions (producing an amide bond) that are known in the art.
[0082] As used herein, "therapeutic agent" refers to a compound, mixture of compounds, or biologic agent that can provide a beneficial effect when administered to a patient.
[0083] As used herein, "amino acid" refers to a compound containing both amino (- NH 2 ) and carboxyl (-COOH) groups generally separated by one carbon atom. The central carbon atom may contain a substituent which can be either charged, ionisable, hydrophilic or hydrophobic. Any of 22 basic building blocks of proteins having the formula N¾ -CHR-COOH, where R is different for each specific amino acid, and the stereochemistry is in the 'L' configuration. Additionally, amino acid can optionally include those with an unnatural 'D' stereochemistry and modified forms of the 'D' and 'L' amino acids.
[0084] As used herein, "peptide" refers to a chain of amino acids in which each amino acid is connected to the next by a formation of an amide bond. Peptides are generally considered to consist of up to 30 amino acids, or alternatively upto 25 amino acids, or alternatively upto 20 amino acids, or alternatively upto 15 amino acids, or alternatively upto 10 amino acids, or alternatively up to 5 amino acids, or alternatively between about 5-10 amino acids, or alternatively between about 10-15 amino acids, while the term "protein" is applied to compounds containing longer amino acid chains. [0085] As used herein, "glycoprotein" refers to a protein which contains a number of carbohydrate substituents.
[0086] As used herein, "halo" or "halogen"refers to fluorine (F), chlorine (CI), bromine (Br) and iodine (I).
[0087] As used herein, a primary, secondary or tertiary alkyl amine refers to an RNH2, an RR"NH or an ' "N group, wherein R, R' and R" independently represent, without limitation, alkyl, cycloalkyl, aryl, heteroaryl and heteroalicyclic moieties.
[0088] As used herein, "vitamin B12" or VB12" refers to the series of compounds otherwise know as cobalamins which are structurally identical and vary only in the nature of the monodentate axial ligand attached to the VB12 cobalt atom, which typically can be cyanide (cyanocobalamin), methyl (methylcobalamin), hydroxyl (hydroxycobalamin), or nitric oxide (nitrosylcobalamin). It is known in the art that these axial ligands can be exchanged under appropriate conditions, and such ligand exchange is incorporated as part of this disclosure. Linkage of the VB12 to the polymer systems to create the delivery systems described herein can be accomplished by converting one or more amide to carboxyl then using the free carboxyl to form a covalent link. Alternatively, formation of a covalent bond to one of the two hydroxyl groups of the ribose unit of VB12 can be employed. Alternatively, VB12 could be linked to the polymer system might also be accomplished by addition of a suitable monodentate ligand to the polymer, via an optional linker, and formation of a metal coordinate bond between the cobalt atom of VB12 and the polymer-attached monodentate ligand.
[0089] As used herein, a "disease" or "medical condition" is an abnormal condition of an organism that impairs bodily functions, associated with specific symptoms and signs.
[0090] As used herein, the term "cancer" refers to various types of malignant neoplasms, most of which can invade surrounding tissues, and may metastasize to different sites, as defined by Stedman's Medical Dictionary, 25th edition (Hensyl ed. 1990). Examples, without limitation, of cancers which may be treated using the compounds of the present invention include, but are not limited to, brain, ovarian, colon, prostate, kidney, bladder, breast, lung, oral, skin and blood cancers.
[0091] As used herein, a "tumor-seeking" group refers to an entity that is know to preferentially seek out and bond to surface structures on neoplastic cells that do not occur or are expressed to a substantially lesser degree by normal cells or entitles that preferentially accumulate in tumors over normal tissue.
[0092] As used herein, the terms "treat", "treating" and "treatment" refer to a method of alleviating or abrogating a disease and/or its attendant symptoms. The effect may be prophylactic in terms of completely or partially preventing a disorder or sign or symptom thereof, and/or may be therapeutic in terms of a partial or complete cure for a disorder and/or adverse effect attributable to the disorder. For example, the life expectancy of an individual affected with a cancer will be increased and/or that one or more of the symptoms of the disease will be reduced.
[0093] As used herein, "administer," "administering" or "administration" refers to the delivery of a compound or compounds of this invention or of a pharmaceutical composition containing a compound or compounds of this invention to a patient in a manner suitable for the treatment of a particular disease, such as cancer.
"Administration" can be effected in one dose, continuously or intermittently throughout the course of treatment. Methods of determining the most effective means and dosage of administration are known to those of skill in the art and will vary with the composition used for therapy, the purpose of the therapy, the target cell being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician. Suitable dosage formulations and methods of administering the agents are known in the art. Route of administration can also be determined and method of determining the most effective route of administration are known to those of skill in the art and will vary with the composition used for treatment, the purpose of the treatment, the health condition or disease stage of the subject being treated, and target cell or tissue. Non-limiting examples of route of administration include oral administration, nasal administration, injection, and topical application. [0094] A "patient" or a "subject" refers to any higher organism that is susceptible to disease. Examples of such higher organisms include, without limitation, mice, rats, rabbits, dogs, cats, horses, cows, pigs, sheep, fish and reptiles. In some embodiments, "patient" or "subject" refers to a human being.
[0095] As used herein, the term "therapeutically effective amount" refers to that amount of a compound or combination of compounds of this invention which has the effect of (a) preventing a disorder from occurring in a subject that may be predisposed to a disorder, but may have not yet been diagnosed as having it; (b) inhibiting a disorder, i.e., arresting its development; or (c) relieving or ameliorating the disorder. For example, but not limited to, (1) reducing the size of the tumor; (2) inhibiting (that is, slowing to some extent, preferably stopping) tumor metastasis; (3) inhibiting to some extent (that is slowing to some extent, preferably stopping) tumor growth; (4) relieving to some extent (or preferably eliminating) one or more symptoms associated with the cancer; and/or (5) extending survival time of the patient.
[0096] As used herein, a "pharmaceutical composition" refers to a mixture of one or more of the compounds of this invention with other chemical components such as pharmaceutically acceptable excipients. The purpose of a pharmacological composition is to facilitate administration of a compound of this invention to a patient.
[0097] As used herein, a "pharmaceutically acceptable excipient" or
"pharmaceutically acceptable carrier" refers to an excipient that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered composition. "Pharmaceutically acceptable carrier" or "pharmaceutically acceptable excipient" refers to any diluents, excipients, or carriers that may be used in the compositions of the invention. Such excipients or carriers include, without limitation, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances, such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat. Suitable pharmaceutical carriers are described in Remington's
Pharmaceutical Sciences, Mack Publishing Company, a standard reference text in this field. They are preferably selected with respect to the intended form of
administration, that is, oral tablets, capsules, elixirs, syrups and the like, and consistent with conventional pharmaceutical practices.
Description of Embodiments
Oral Drug Delivery
[0098] A number of technologies have been advocated for the enhancement of oral bioavailability of pharmaceutically-active compounds. As an example, one area of particularly active research has been in the development of technologies for the oral delivery of insulin. Khafagy et al (Advanced Drug Delivery Reviews 59 (2007) 1521- 1546) classified the various oral insulin approaches as: Absorption enhancers;
Enzyme inhibitors; Mucoadhesive polymeric systems; Particulate carrier delivery systems; and Targeted delivery systems.
[0099] Absorption or permeation enhancers are molecules which either increase the fluidity of membranes or widen junctions between the cells of membranes thus providing a small transient improvement in paracellular and transcellular drug transport. There are a number of distinct disadvantages to absorption enhancers for oral drug delivery:
[0100] Typically, they should slightly precede the appearance of drug molecules at the absorption site to provide maximum possible drug absorption. Once the concentration of the enhancer molecule decreases at the membrane site (for example, by continued transit in the GI tract, or by virtue of the fact that it is itself absorbed or metabolized), the membrane permability returns to normal.
[0101] Increasing membrane permeability permits increased penetration of all molecules in the vicinity, not just the drug molecules.
[0102] Enzyme inhibitors slow the rate at which actives, particularly proteins and peptides, are enzymatically degraded in the GI tract. In principle, this provides for a higher concentration of the active at the sites of absorption, resulting in greater passive absorption by virtue of a larger concentration gradient. This effect is only beneficial for actives that are naturally able to diffuse readily across the gut wall, and are only prevented from doing so through enzymatic degradation of the active compound. Additionally, inhibition of enzyme activity in the GI tract can give rise to significant adverse effects as inhibition of protein degradation will be non-selective. For example, enzyme inhibitors will reduce the rate to breakdown (and hence reduced absorption) of food proteins.
[0103] Peristalsis generates a flow of material down the GI tract. Materials moving along the small intestine, where most pharmaceutical actives are thought to be absorbed, do so in an averge time of about three hours. If were possible to retard the flow of drugs, and provide them with greater contact at the sites of absorption, it should be possible to achieve higher levels of absorption of drugs that are otherwise poorly absorbed in the GI tract. Because of transient 'sticking' of mucoadhesive polymeric systems to the mucosal surface of the GI tract lumen, formulations based upon such polymers have the potential to demonstrate an extended residence on the epithelial cell layer, slowing the flow of these particles relative to other material in the GI tract. . When formulated into particles, mucoadhesive polymers may also provide some protection to embedded active agents that might otherwise be degraded in the GI tract. Because of the direct contact between the polymer formulation and the GI mucosa, other potential advantages of this oral drug delivery system is the possibility for direct diffusion of actives from the particle into the mucosa and epithelial cell layer, and for pinocytosis of particles into epithelial cells. All of these potential benefits suggest that oral drug delivery systems based upon mucoadhesive polymers should be highly effective, yet results to date in numerous examples in the literature indicate only modest improvements in oral bioavailability of pharmaceutical active compounds using mucoadhesive polymer formulations.
[0104] Gastrointestinal absorption of many essential nutrients and vitamins can be facilitated by active transport processes. These processes generally require the material to bind to a surface receptor, which initiates a process such as receptor- mediated endocytosis whereby the active is absorbed into the epithelial cell. Dissassociation of the receptor-active complex occurs and other processes then facilitate the transfer of the active material into the blood stream. One transport system which has been well documented in the literature is the process for absorption of vitamin B12 (VB12). VB12 liberated from food binds to intrinsic factor (IF, which is produced in the stomach and passes down the GI tract following a meal), and the VB12-IF complex binds to the Cubulin receptor, primarily located in the ileum. Receptor-mediated endocytosis, as described above, then takes place. Dissociation of the receptor-IF-VB12 complex in the epithelial cell results in liberation of VB12, which then binds to transcobalamin II, a protein which facilities the transfer of VB12 to the blood stream.
[0105] It has been documented by Russell- Jones and others that the VB12 uptake mechanism in the GI tract can be used to facilitate the oral absorption of other compounds. Using a 'Trojan Horse' approach, the active is either covalently linked via a degradable linker group to VB12, or covalently linked via a degradable linker group to a polymer which is also linked to VB12, or encapsulated in a nanoparticle to which VB12 is attached (see Figure 2). In the polymer approach, multiple drug-linker groups can be attached to a single polymer strand. For each of these possibilities, provided that VB12 is bound to the linker or particle so as not to prevent binding to IF, these constructs will bind IF in the GI tract and be taken up primarily in the ileum by the cubulin receptor and transported to the bloodstream. Breakdown of the degradable linker will then release drug in the bloodstream, completing its oral absorption. Similarly, drug release by diffusion from the nanoparticle and/or breakdown of the nanoparticle structure in the bloodstream will result in
bioavailability of the active. In the case of single conjugation of the active to the VB12 via a linker, one molecule of the drug is absorbed for each receptor-mediated endocytotic event. By comparision, the polymer approach allows for multiple drug molecules to be absorbed each time one polymer strand is absorbed as a result of VB12 attached to that polymer strand binding to IF and cubulin. This allows for an 'amplication' of oral uptake when compared with the 1 : 1 conjugate. Similarly, a VB12 nanoparticle can carry many copies of the drug, also permitting amplification of drug uptake. [0106] A number of patents which describe either single VB12 conjugates, VB12- polymer conjugates, and VB12-coated naoparticles are known, represented by the following (all of which are incorporated herein by reference in their entirety): US Patent Nos: 5,428,023; 5,449,720; 5,548,064; 5,574,018; 5,589,463; 5,807,832;
5,863,900; 5,869,466; 5,589,463; 6,083,926; 6,150,341; 6,159,502; 6,221,397;
6,262,253 and 6,482,413.
[0107] In some embodiments, the formation of a covalent link to connect the drug to VB12 is or may not be the method of utilizing this technology. By formation of a covalent link to the drug, it may be chemically altered. For a drug which has already received Regulatory approval for its use as a medication, a new active pharmaceutical ingredient (API) would have been created that will require a full drug development program for its approval. The release of the drug requires cleavance of the degradable linker, which may leave fragments of the linker still attached to the drug, such that is a different chemical entity. By trapping the drug in a VB12-coated nanoparticle, it remains chemically unaltered, so a previously approved drug should not need a full development program for Regulatory approval of the VB12-coated nanoparticle formulation of that drug.
[0108] Many methods of forming nanoparticles and utilizing such nanoparticles for drug delivery are reported in the literature. Furthermore, VB12-coated nanoparticles for oral drug delivery have been described (see patent literature: US Pat. Nos.:
6,159,502 and 6,482,413; Patent Publ. No.: WO2007131286). None of the
formulations described in these patents have advanced from basic research to the clinic as each of these technologies has fundamental technical issues; as examples poor encapsulation / weak binding of the drug to the carrier. The methods of preparation known in the art can also give rise to degradation or denaturing of protein, peptides and other pharmaceutical active ingredients, lowering efficacy and introducing additional impurities.
[0109] It is an object of the present invention to overcome or at least alleviate one or more of the above-mentioned disadvantages of the prior art. Disease-targeting
[0110] In many diseases which involve cell proliferation, there is increased demand for certain vitamins compared with normal tissue. This phenomena can be utilized for targeting drugs to the site of disease such as tumors. For example, folic acid (vitamin B9), riboflavin, thiamine, and vitamin B12 has been reported used to target drugs and radioactive materials to tumors for therapy and diagnosis (U.S. Patent Nos.:
5,108,921; 5,416,016; 5,635,382; 5,688,488; 7,128,893; 7,601,332; and publication Waibel et al, Cancer Res., 2008, Vol. 68:2904-2911). In most cases, the drug is covalently linked to the targeting system, thereby altering the drug and potentially altering its pharmacological and toxicological profile. A simple method is required to target the drug to sites of disease without chemical modification of the drug.
[0111] In many diseases, cells have an increased demand for vitamin B12 which is reflected by an increase in the expression of cell surface receptors which facilitate the uptake, through receptor-mediated endocytosis, of this vitamin. Mechanistically, vitamin B12 binds to the circulating protein, transcobalamin II (TC-II), and it is the B12-TC-II complex which is recognized by the cell surface receptors. The B12-TC-II complex binding results in receptor-mediated endocytosis and internalization of the complex, followed by release of the vitamin B12. As was the case for vitamin B12 uptake in the GI tract, the process for cell uptake of vitamin B12 can be utilized using the 'Trojan Horse' principle to transport molecules into cells when these molecules are chemically linked to vitamin B12. For example, R. Waibel et al, Cancer Res., 2008, 68, 2904-2911.
[0112] It is one object of the present invention to provide drug carrier systems and formulations which are effective while requiring no drug modification.
Polyelectrolyte complexes
[0113] Polyelectrolyte complex (PEC) is a term which relates to two or more compounds binding to each other by virtue of multiple charge interactions. For the formation of nanoparticle PECs, it is usual that at least one of the compounds involved in an oligomer or polymer that contains multiple charged (or ionisable) groups, all either positive or negative. This polymer, when brought into contact with an compound containing one or more charged (or ionisable) groups of the oppositve charge forms a complex wherein the charged groups on one compound form ionic bonds with the charged groups of the other compound. Typically, both compounds possess charged or ionisable groups and form multiple ionic bonds with each other. Further interactions such as hydrophibic bonding and H-bonding may serve to increase the strength of binding of one compound to the other. In the formation of nanoparticles, many molecules of the two or more charged or ionisable compounds come together to form a three-dimensional matrix of nanoparticle size. One big advantage for PECs over other methods of nanoparticle formation is that such nanoparticles can be formed simply by bringing both compounds together in an aqueous environment. In the case of drug-loaded PECs, a simple example might be a pharmacologically-active peptide with either a net positive or net negative overall charge at a suitable pH with a polymer which has charged (or ionisable) groups which have the opposite charge to that of the peptide. Another example of PECs results from the formation of PECs from two polymers, one with negatively charged groups and one with positively-charged groups. Bringing these two polymers into contact in an aqueous environment which also contains the drug can result in the formation of PECs in which the drug is trapped in the nanoparticle matrix during PEC formation. In either of these two general examples, controlled drug release can result through slow disassociation of the nanoparticle in the body. The PEC components can be chemically-modified to assist in nanoparticle formation; for example, conversion of a tertiary to quaternary amine or through the addition of hydrophobic groups
[0114] Some representative examples of PECs intended to deliver a therapeutically- beneficial payload can be found in the following publications (all of which are incorporated herein by reference in their entirety): A Jintapattanakit et al, International Journal of Pharmaceutics, 2007, 342, 240-249; Y-Z Du et al, International Journal of Pharmaceutics, 2010 (eprint); Vieira and AM Carmona- ibeiro, Journal of
Nanobiotechnology, May 2008; and H-W Sung and H Tu, U.S. Pat 7,556,826.
[0115] Attempts have been made to attach targeting groups to PECs to enhance the delivery of these constructs in vivo to certain target regions, e.g. cancer. Examples of targeting groups which have been attached to charged polymers include transferin (M. Kursa et al, Bioconjugate Chemistry, 2003, 14 222) and folate (SH Kim et al, Biotechnol Prog. 2007, 23, 232), During nanoparticle formation such large and/or hydrophobic targeting groups might be expected to migrate towards the surface of nanoparticles in preference to the hydrophilic environment within the internal matrix of the nanoparticle.
[0116] It is one object of the present invention to provide a novel nanoparticle system and simple methods of preparation whereupon a nanoparticle of type polyelectrolyte complex (PEC) is formed presenting molecules of vitamin B12 on the surface of said nanoparticle and one or more therapeutically-active compounds are contained within the nanoparticle.
[0117] It is an additional object of the present invention that the above PEC nanoparticle system provides some protection from degradation or denaturing of the one or more therapeutically-active compounds contained within the nanoparticle in body compartments in body compartments in which one or more therapeutically- active compounds contained within the nanoparticle might otherwise, if unprotected, be caused to degrade, denature or metabolize.
[0118] It is an additional object of the present invention that the above PEC nanoparticle system has the potential benefit of transportation from one body compartment to another by utilizing the body's natural transportation mechanisms for vitamin B12, including, but not limited to, transportation from the gut lumen to the portal blood vein in the ileum of the GI tract, passage across cell membranes to enter cellular compartments, and traverse major biological barriers such as the blood-brain barrier.
[0119] It is an additional object of the present invention that the above PEC nanoparticle system can release the one or more therapeutically active compounds contained within the nanoparticle in a controlled manner, and that compound release can result from diffusion of drug through the nanoparticle matrix and/or degradation of the matrix.
[0120] It is an additional object of the present invention that the above PEC nanoparticle system can release the one or more therapeutically active compounds contained within the nanoparticle at sites within the body to achieve a therapeutically- meaningful effect.
[0121] It is an additional object of the present invention that the above PEC nanoparticle system can degrade in the body to permit the components of the nanoparticle to be safely metabolized and eliminated from the body.
[0122] It is an additional object of the present invention that the above PEC nanoparticle system can formulated by methods known in the art to provide pharmaceutical preparations suitable for administration to patients. Examples of pharmaceutical preparations that might be suitable for the PEC nanoparticle system of this invention include, but are not limited to, tablets or capsules for oral
administration, lyophilized powers in vials for subsequent reconstitution with a pharmaceutically-acceptable vehicle for injection into the patient, or liquids comprising the drug containing PEC nanoparticle system in pharmaceutically- acceptable vehicle for injection into the patient.
[0123] It is an additional object of the present invention that the above PEC nanoparticle system be administered to patients for the prevention and treatment of diseases, including, but not limited to cancer, autoimmune conditions, endocrine disorders, diabetes, genetic conditions, chromosome conditions, viral infections, bacterial infections, parasitic infections, mitochondrial diseases, sexually transmitted diseases, immune disorders, balance disorders, pain, systemic disorders, blood conditions, blood vessel conditions, nerve conditions, and conditions of muscles, heart and other organs.
[0124] In one mode, the present invention consists of nanoparticles formed by bringing together in a suitable solvent a synthetic or natural polymer containing multiple charged and/or ionisable groups, predominantly either positive or negative, with a therapeutic agent which has one or more charged or ionisable groups of the opposite charge to the primary charge of the polymer, and optionally other
components which either assist in nanoparticle formation or in the placement of VB12 on the surface of the nanoparticle. The polymer and the therapeutically active agent can form a PEC, either alone or in combination with the other aforementioned components. VB12 is an essential component of the nanoparticle, introduced prior to nanoparticle formation either by covalent attachment to the polymer, to the therapeutic agent, and/or to one of the optional additional components.
[0125] In a further mode, the present invention consists of nanoparticles formed by bringing together in a suitable solvent two synthetic or natural polymers in which one polymer contains multiple charged and/or ionisable groups of net positive charge and the other polymer contains multiple charged and/or ionisable groups of net negative charge, together with a therapeutic agent which may or may not possess charged or ionisable groups, and optionally other components which either assist in nanoparticle formation or in the placement of VB12 on the surface of the nanoparticle. The two polymers can form a PEC, either alone or in combination with the other
aforementioned components, and the therapeutically active component is trapped in the PEC during PEC formation. VB12 is an essential component of the nanoparticle, introduced prior to nanoparticle formation either by covalent attachment to the polymer, to the therapeutic agent, and/or to one of the optional additional components.
[0126] In a further mode, the present invention consists of nanoparticles formed as described above in which VB12 in not a component or part of a component of the nanoparticle and is introduced to the surface of the nanoparticle after nanoparticle formation either by formation of a covalent bond between the nanoparticle surface and VB12 or VB12 derivative, or by the formation of a physical bonds (ionic, hydrophilic, and/or hydrophobic) between the nanoparticle and VB12 or VB12 derivative.
[0127] As described earlier, vitamin B12 contains a monodentate axial ligand. It is known in the art that these axial ligands can be exchanged under appropriate conditions, and such ligand exchange is incorporated as part this disclosure. For example, it is known that nitrosyl cobalamin can be effective as an antitumor agent because it serves to deliver nitric oxide to tumors (for example; Bauer, Anti-Cancer Drugs, 1998, 9, 239) and it may be desirable to convert VB12 in the nanoparticles of this invention to the nitrosyl form to enhance the therapeutic effect. In addition, in order to link the VB12 molecule to a polymer via an optional linker, the VB12 may be connected to the linker through the cobalt atom of VB12 by way of a ligand exchange process, as described in (for example; U.S. Patent Publication No.
20020115595 and Bagnato et al, J. Org. Chem.2004, Vol. 69:8987).
[0128] Alternatively VB12 can be attached using other methods known in the art. For example, one or more of the primary amide groups of VB12 may be selectively hydro lyzed to generate a free carboxyl group or ester, and subsequently the VB12 can be linked to the polymer via an optional linker through the liberated carboxyl group by methods well-known in the art (for example; Wilbur et al, Bioconjugate Chem. 1996, Vol. 7:461-474). The preferred method of attachment of VB12 to the polymer via an optional linker involves the formation of a covalent bond to one of the two hydroxyl groups of the ribose unit of VB12 by methods known in the art (for example; McEwan et al, Bioconjugate Chem. 1999, Vol. 10: 1131-1136).
[0129] Examples of positively charged or ionizable polymers that can be used to form the PEC nanoparticles of this invention include chitosan, alkylated and acylated chitosan derivatives, DEAE-dextran, polyethylenimine, polyamidoamines, poly(vinyl- N-alkylpyridinium), poly(DMAE-methacrylamide), poly(di- and trialkylamino- methacrylamides), poly(HPMA-co-di- and trialkylamino-methacrylamides), positively-charged polypeptides such as polylysine, polyhistidine, and
pharmaceutically-approved polymeric excipients that carry a net positive charge at physiologically-relevant pH levels. In one embodiment, the chitosan derivative employed is Ν,Ν,Ν-trimethyl chitosan (TMC). For the purpose of fulfilling the requirements of this invention, the positively charged or ionizable polymers may be optionally modified by covalent linkage of one or more VB12 molecules, either directly or via a suitable linker.
[0130] Examples of negatively charged or ionizable polymers that can be used to form the PEC nanoparticles of this invention include anionic polysaccharides such as heparin, chondroitin, hyaluronic acid, alginates, pectins, carrageenans, xanthans, neutral polysaccharides such as dextran, cellulose, starch, chitosan that have been derivatized with alkyl- or acyl-carboxylates, sulfates, sulfonates or phosphates, anionic poly-amino acids such as polyglutamic acid, poly-gamma-glutamic acid, polyaspartic acid, synthetic polymers such as poly(acrylate),
poly(hydroxypropylmethacrylamide-co-carboxylates /sulfates /sulfonates /phosphates), poly(styrene-sulfonate), poly(sulfopropylacrylate), and
pharmaceutically-approved polymeric excipients that carry a net negative charge at physiologically-relevant pH levels. In one embodiment, the dextran derivative employed is succinyl-dextran (that is, dextran treated with succinic anhydride providing substituents with one free carboxyl group). In an additional embodiment, the dextran derivative employed is carboxymethyl-dextran (that is, dextran treated with a carboxylmethyl halide providing substituents with one free carboxyl group). For the purpose of fulfilling the requirements of this invention, the negatively charged or ionizable polymers may be optionally modified by covalent linkage of one or more VB12 molecules, either directly or via a suitable linker.
[0131] It is within the scope of this invention that naturally-occuring polymers or readily-available synthetic polymers be used directly for formation of nanoparticles of this invention, or that such polymers can be synthetically-modified. Modifications can include, but are not limited to, the introduction of charged or ionizable groups, attachment of VB12, and the introduction of functional groups (for example, hydrophobic or hydrophilic) which either enhance the nanoparticle formation and/or the pharmaceutical qualities of the resultant nanoparticles.
[0132] In one embodiment of this invention, PECs are formed by interacting a therapeutically-active compound which possess a net positive or negative charge with a polymer as described above with the opposite charge. Nanoparticle formation and/or the suitability of the resultant nanoparticle for pharmaceutical use may be facilitated or improved by the use of additional components during nanoparticle formation such that these additional components become incorporated into the nanoparticle.
[0133] In some embodiments, a ratio of the therapeutic agent to the vitamin B12 in the nanoparticles of the present invention is in a range of 1 :20 to about 20: 1 , or alternatively in a range of about 1 : 15 to about 15: 1, or alternatively in a range of about 1 : 10 to about 10: 1, or alternatively in a range of about 1 :5 to about 5: 1, or alternatively in a range of about 1 :2 to about 2: 1 , or alternatively the ratio of the therapeutic agent to the vitamin B12 in the nanoparticles of the present invention is about 1 : 1 , or alternatively about 2: 1 , or alternatively about 1 :2, or alternatively about 3: 1, or alternatively about 1 :3, or alternatively about 4:1, or alternatively about 1 :4, or alternatively about 5: 1, or alternatively about 1:5, or alternatively about 6: 1, or alternatively about 1 :6, or alternatively about 7: 1, or alternatively about 1 :7, or alternatively about 8: 1, or alternatively about 1:8, or alternatively about 9: 1, or alternatively about 1 :9, or alternatively about 2:3.
[0134] In a further embodiment of this invention, PECs are formed by interacting a therapeutically-active compound which may or may not possess a net positive or negative charge with two polymers as described above, one with a net positive charge and one with a net negative charge. Nanoparticle formation and/or the suitability of the resultant nanoparticle for pharmaceutical use may be facilitated or improved by the use of additional components during nanoparticle formation such that these additional components become incorporated into the nanoparticle.
[0135] It will be obvious to those skilled in the art that pharmaceutically-suitable nanoparticles can also be formed by use of more than one polymer of a particular charge. For example, in forming a PEC nanoparticle two or more positively charged polymers can be used when a positively charged polymer is required. Similarly, two or more negatively charged polymers can be used when a negatively charged polymer is required.
[0136] Furthermore, it will be obvious to those skilled in the art that
pharmaceutically-suitable nanoparticles can also be formed by incorporation of more than one therapeutically-active compound.
[0137] As indicated above, it may be desirous in the formation of PEC nanoparticles to utilize additional components before, during or after nanoparticle formation in order to control the size of nanoparticles, control stability and/or the drug release profile. Possible additional components include, but are not limited to, polyethylene glycol (PEG) and PEG block copolymers, polyacrylic, polymethacrylic, and other synthetic polymers, starch, cellulose, and other polysaccharides, fatty acids and other surfactants, and metal ions, especially di- and trivalent ions such as zinc,
magnesium,and calcium. Additional components might also include a crosslinking agent, for example epoxy compounds, dialdehyde starch, glutaraldehyde,
formaldehyde, dimethyl suberimidate, carbodiimides, succinimidyls, diisocyanates, acyl azide, reuterin, and crosslinking effected by ultraviolet irradiation. [0138] Also as indicated above, unless VB12 is bound to the nanoparticle after nanoparticle formation, then one of the components used in the formation of the nanoparticle must contain VB12 either covalently of physically linked to that component. VB12 might be linked, directly or via a suitable linker, to either the positively-charged polymer, the negatively-charged polymer, the therapeutically- active compound, or one of the additional components (if employed).
[0139] It is within the scope of this invention that the primary purpose of the additional component is to facilitate the introduction of VB12 to the the nanoparticle during its formation. For example, the additional component could be VB12 which contains a fatty acid attached to either the 5 '-0 or 2'-0 position (or both), and the VB12 is incorporated by hydrophobic interaction of the fatty acid portion with other hydrophobic components involved in nanoparticle formation. Other methods of incorporating VB12 as one of the additional components will be obvious to those skilled in the art. As another example, the VB12 additional component may be functionalized with a compound that is known to bind strongly to one of the other components of nanoparticle formation (e.g. strepatavidin and biotin are well known to bind strongly to each other; similarly, U.S. Pat. 5,605,890 exemplifies a cyclodextrin- adamantane "lock and key" binding system).
[0140] The polymers used in this invention can have an average molecular weight in the range of 1-10,000 kDa. The preferred average molecular weights will be determined by the specific requirements of formation and the desired pharmaceutical properties of the PEC nanoparticles. In some embodiments, the avergage molecular weight of the polymer of the invention is in a range of about 1-10,000 kDa; or alternatively in a range of about 1-5000 KDa; or alternatively in a range of about 1- 1000 KDa; or alternatively in a range of about 1-500 KDa; or alternatively in a range of about 1-100 KDa; or alternatively in a range of about 10-10,000 KDa; or alternatively in a range of about 10-5000 KDa; or alternatively in a range of about 10- 4000 KDa; or alternatively in a range of about 10-2000 KDa; or alternatively in a range of about 10-1000 KDa; or alternatively in a range of about 10-500 KDa; or alternatively in a range of about 50-10,000 KDa; or alternatively in a range of about 50-5,000 KDa; or alternatively in a range of about 50-1,000 KDa; or alternatively in a range of about 50-500 KDa; or alternatively in a range of about 100-10,000 KDa; or alternatively in a range of about 100-5,000 KDa; or alternatively in a range of about 100-1,000 KDa; or alternatively in a range of about 100-500 KDa; or alternatively in a range of about 500-10,000 KDa; or alternatively in a range of about 500-1,000 KDa; or alternatively in a range of about 1000-10,000 KDa; or alternatively in a range of about 1000-5,000 KDa; or alternatively in a range of about 2000-10,000 KDa; or alternatively in a range of about 2000-5,000 KDa; or alternatively in a range of about 4000-10,000 KDa; or alternatively in a range of about 4000-5000 KDa; or
alternatively in a range of about 5000-10,000 KDa; or alternatively in a range of about 6000-10,000 KDa; or alternatively in a range of about 7000-10,000 KDa; or alternatively in a range of about 8000-10,000 KDa; or alternatively in a range of about 9000-10,000 KDa.
[0141] In one embodiment, a function of the nanoparticles of this invention is to facilitate or enhance the oral bioavailablity of the therapeutically active compound (or compounds) contained within the nanoparticle. For example, the therapeutically active compound (or compounds) may have poor natural oral bioavailability by virtue of either (or both) degradation or denaturing in the GI tract or an inability to cross the gut wall and enter the bloodstream.
[0142] In a further embodiment, a function of the nanoparticles of this invention is to modify the oral bioavailablity of the therapeutically active compound (or compounds) contained within the nanoparticle. For example, the therapeutically active compound (or compounds) may have sufficient oral bioavailability to be therapeutically effective when given orally, and the nanoparticles of this invention either improve oral bioavailability (reducing the amount of drug that needs to be adminstered) and/or alters the pharmacokinetic profile of the drug in a desirable manner.
[0143] In a further embodiment, a function of the nanoparticles of this invention is to facilitate targeting of the therapeutically active compound (or compounds) contained within the nanoparticle to sites of disease, especially in diseases in which the demand for VB12 is increased compared with the demand for the vitamin normally. Examples of diseases which are known to display increased demand for VB12 include cancer, rheumatoid arthritis, psoriasis, acute leukemia, lymphomas, Crohn's disease, ulcerative colitis, and multiple sclerosis. Pharmaceutical preparations for targeted delivery to sites of disease can be administered by injection.
[0144] In a further embodiment, a function of the nanoparticles of this invention is to combine oral drug delivery and targeting; following oral drug delivery as described above, the nanoparticles are then targeted to sites of disease, also as described above.
[0145] In a further embodiment, a function of the nanoparticles of this invention is to deliver polynucleotides (e.g. siRNA and antisense RNA) and other RNA interference therapeutics across cell membranes to deliver the actives into the intracellular environment and to the nucleus, where they are effective, and for gene therapy.
[0146] In a further embodiment, a function of the nanoparticles of this invention is to deliver therapeutics which are effective in the treatment of CNS disorders across the blood-brain barrier.
[0147] Therapeutic agents that can be delivered in effective amounts across biological barriers using the PEC nanoparticles of this invention include, but are not limited to small molecules, macromolecules, synthetic drugs, semi-synthetic drugs, naturally-occurring compounds, proteins, peptides, nucleosides, nucleotides, analgesics, antiallergenics, antianginal agents, antiarrythmic drugs, antibiotics, anticoagulants, antidementia drugs, antidepressants, antidiabetics, antihistamines, antihypertensives, anti-inflammatories, antineoplastic agents, antiparasitics, antipyretic, antiretroviral drugs, antiulcerative agents, antiviral agents, cardiovascular drugs, cholesterol-lowering agents, CNS active drugs, growth hormone inhibitors, growth hormones, hematopoietic drugs, hemostatics, hormones, hypotensive diuretics, keratolytics, therapeutics for osteoporosis, vaccines, vasoconstrictors, vasodilators. Such therapeutics can be used alone or in combination with other therapeutic agents using dosing regimens effective in providing a beneficial therapeutic effect.
[0148] Examples of therapeutic agents that are analgesics are morphine,
hydromorphone, oxymorphone, lovorphanol, levallorphan, codeine, nalmefene, nalorphine, nalozone, naltrexone, buprenorphine, butorphanol, or nalbufine. Examples of therapeutic agents that are antiallergic compounds include amlexanox, astemizole, azelastinep, emirolast, alopatadine, cromolyn, fenpiprane, repirinast, tranilast, and traxanox.
[0149] Examples of therapeutic agents that are antianginal agents include nifedipine, atenol, bepridil, carazolol and epanolol.
[0150] Examples of therapeutic agents that are anti-inflammatory analgesic agents include acetaminophen, methyl salicylate, monoglycol salicylate, aspirin, mefenamic acid, flufenamic acid, indomethacin, diclofenac, alclofenac, diclofenac sodium, ibuprofen, ketoprofen, naproxen, pranoprofen, fenoprofen, sulindac, fenclofenac, clidanac, flurbiprofen, fentiazac, bufexamac, piroxicam, phenylbutazone,
oxyphenbutazone, clofezone, pentazocine, mepirizole, tiaramide hydrochloride, etc.
[0151] Examples of therapeutic agents that are steroidal anti-inflammatory agents include hydrocortisone, predonisolone, dexamethasone, triamcinolone acetonide, fluocinolone acetonide, hydrocortisone acetate, predonisolone acetate,
methylpredonisolone, dexamethasone acetate, betamethasone, betamethasone valerate, flumetasone, fiuorometholone, beclomethasone diproprionate, etc.
[0152] Examples of therapeutic agents that are antihistamines include
diphenhydramine hydrochloride, diphenhydramine salicylate, diphenhydramine, chlorpheniramine hydrochloride, chlorpheniramine maleate isothipendyl
hydrochloride, tripelennamine hydrochloride, promethazine hydrochloride, methdilazine hydrochloride, etc.
[0153] Examples of therapeutic agents that are vasoconstrictors include naphazoline nitrate, tetrahydrozoline hydrochloride, oxymetazoline hydrochloride, phenylephrine hydrochloride, tramazoline hydrochloride, etc.
[0154] Examples of therapeutic agents that are hemostatics include thrombin, phytonadione, protamine sulfate, aminocaproic acid, tranexamic acid, carbazochrome, carbaxochrome sodium sulfanate, rutin, hesperidin, etc.
[0155] Examples of therapeutic agents that are chemotherapeutic drugs include sulfamine, sulfathiazole, sulfadiazine, homosulfamine, sulfisoxazole, sulfisomidine, sulfamethizole, nitro furazone, taxanes, platinum compounds, topoisomerase I inhibitors, and anthrocycline.
[0156] Examples of therapeutic agents that are antibiotics include penicillin, meticillin, oxacillin, cefalotin, cefalordin, erythromycin, lincomycin, tetracycline, chlortetracycline, oxytetracycline, metacycline, chloramphenicol, kanamycin, streptomycin, gentamicin, bacitracin, cycloserine, and clindamycin.
Examples of therapeutic agents that are keratolytics include salicylic acid, podophyllum resin, podolifox, and cantharidin.
[0157] Examples of therapeutic agents that are growth factors include Autocrine motility factor, Bone morphogenetic proteins (BMPs), Epidermal growth factor
(EGF), Erythropoietin (EPO), Fibroblast growth factor (FGF), Granulocyte-colony stimulating factor (G-CSF), Granulocyte-macrophage colony stimulating factor (GM- CSF), Growth differentiation factor-9 (GDF9), Hepatocyte growth factor (HGF), Hepatoma derived growth factor (HDGF), Insulin-like growth factor (IGF), migration-stimulating factor, Myostatin (GDF-8), Nerve growth factor (NGF) and other neurotrophins, Platelet-derived growth factor (PDGF), Thrombopoietin (TPO), Transforming growth factor alpha, Transforming growth factor beta (TGF-?), Vascular endothelial growth factor (VEGF), placental growth factor (PlGF)and Foetal Bovine Somatotrophin (FBS).
[0158] Examples of therapeutic agents that are growth hormone inhibitors are octreotide and somatostatin.
[0159] Examples of therapeutic agents that are hormones include Adiponectin, Adrenocorticotropic hormone (or corticotropin), Aldosterone, Androstenedione, Angiotensinogen and angiotensin, Antidiuretic hormone (or vasopressin, arginine vasopressin), Antimullerian hormone (or mullerian inhibiting factor or hormone), Atrial-natriuretic peptide (or atriopeptin), Brain natriuretic peptide, Calcidiol (25- hydroxyvitamin D3), Calcitonin, Calcitriol, Cholecystokinin, Corticotropin-releasing hormone, Cortisol, Dehydroepiandrosterone, Dihydrotestosterone, Dopamine (or prolactin inhibiting hormone), Endothelin, Enkephalin, Epinephrine (or adrenaline), Erythropoietin, Estradiol, Estriol, Estrone, Follicle-stimulating hormone, Gastrin, Ghrelin, Glucagon, Gonadotropin-releasing hormone, , Growth hormone-releasing hormone, Histamine, Human chorionic gonadotropin, Human Growth hormone, Human placental lactogen, Inhibin, Insulin, Insulin-like growth factor (or
somatomedin), Leptin, Leukotrienes, Lipotropin, Luteinizing hormone, Melanocyte stimulating hormone, Melatonin, Neuropeptide Y, Norepinephrine (or noradrenaline), Orexin, Oxytocin, Pancreatic polypeptide, Parathyroid hormone, Progesterone, Prolactin, Prolactin releasing hormone, Prostacyclin, Prostaglandins, elaxin, Renin, Secretin, Serotonin, Somatostatin, Testosterone, Thrombopoietin, Thromboxane, Thyroid-stimulating hormone (or thyrotropin), Thyrotropin-releasing hormone, Thyroxine, Triiodothyronine.
[0160] Examples of therapeutic agents that are analgesic narcotics include fentanyl, buprenorphine, codeine sulfate, levorphanol, and morphine hydrochloride.
[0161] Examples of therapeutic agents that are antiviral drugs include Abacavir, Aciclovir, Acyclovir, Adefovir,Amantadine, Amprenavir, Ampligen, Arbidol, Atazanavir, Atripla, Boceprevir, Cidofovir, Combivir, Darunavir, Delavirdine,
Didanosine, Docosanol, Edoxudine, Efavirenz, Emtricitabine, Enfuvirtide, Entecavir, Famciclovir,Fomivirsen, Fosamprenavir, Foscarnet, Fosfonet, Ganciclovir,
Ibacitabine, Imunovir,Idoxuridine, Imiquimod, Indinavir, Inosine, Interferon type III, Interferon type II, Interferon type I, Interferon, Lamivudine, Lopinavir, Loviride, Maraviroc, Moroxydine, Nelfinavir, Nevirapine, Nexavir, Oseltamivir, Peginterferon alfa-2a, Penciclovir, Peramivir, Pleconaril, Podophyllotoxin,Raltegravir, Reverse transcriptase inhibitor, Ribavirin, Rimantadine, Ritonavir, Pyramidine, Saquinavir, Stavudine, Tea tree oil, Tenofovir, Tenofovir disoproxil, Tipranavir, Trifluridine, Trizivir, Tromantadine, Truvada, Valaciclovir , Valganciclovir, Vicriviroc, Vidarabine, Viramidine, Zalcitabine, Zanamivir, and Zidovudine.
[0162] Examples of therapeutic agents that are drugs for the treatment of diabetes or its side effects includes insulin (natural or recombinant; monomer, hexamer, or mixtures thereof), insulin isophane, insulin lispro, insulin glargine, tolbutamide, acetohexamide, tolazamide, chlorpropamide, glipizide, glyburide, glimepiride, gliclazide, repaglinide, nateglinide, metformin, phenformin, buformin, rosiglitazone, pioglitazone, troglitazone, miglitol, acarbose, Glucagon-like peptide-1, Exanatide, Liraglutide, Taspoglutide, Lixisenatide, Albiglutide, vildagliptin, sitagliptin, saxagliptin, pramlintide, muraglitazar, tesaglitazar, and aleglitazar.
[0163] Examples of therapeutic agents that are drugs used for the treatment of CNS disorders include memantine hydrochloride, donepezil hydrochloride, rivastigmine tartrate, galantamine hydrochloride, and tacrine hydrochloride.
[0164] Examples of therapeutic agents that are drugs used for the treatment of prostate cancer include Dutasteride, Bicalutamide, Ciprofloxacin, Erythromycin, Tamsulosin, Ofloxacin, Terazosin, Leuprolide, Nilutamide, Finasteride, and
Goserelin,
[0165] Examples of therapeutic agents that are drugs used for the treatment of ovarian cancer include Cisplatin, Carboplatin, Paclitaxel, Melphalan, Doxorubicin, hexamethylmelamine, Toptecan, Ifosfamide, Etoposide, and 5-fluorouracil.
[0166] Examples of therapeutic agents that are drugs used for the treatment of colorectal cancer include fluorouracil, bevacizumab, irinotecan, oxaliplatin, cetuximab, panitumumab, leucovorin, and capecitabine.
[0167] Examples of therapeutic agents that are drugs used for the treatment of lung cancer include Carboplatin, Cisplatin, Docetaxel, Erlotinib, Etoposide, Gemcitabine, Gefitinib. Irinotecan, Paclitaxel, Pemetrexed, Topotecan, Vinorelbine, Gefitinib, and Bevacizumab.
[0168] Examples of therapeutic agents that are drugs used for the treatment of melanoma include dacarbazine, interferon alfa-2b, aldesleukin, and acarbazine.
[0169] Examples of therapeutic agents that are drugs used for the treatment multiple sclerosis include Interferon Beta la, Glatiramer Acetate, Mitoxantrone, Azathioprine, Cyclophosphamide, Cyclosporine, Methotrexate, Cladribine, MethylPrednisolone, Prednisone, Prednisolone, Dexamethasone, Corticotropin, Carbamazepine,
Gabapentin, Topiramate, Zonisamide, Phenytoin, Desipramine, Amitriptyline, and Imipramine
[0170] Examples of therapeutic agents that are drugs used for the treatment of Alzheimers disease include donepezil, galantamine, rivastigmine, and memantine. Examples of therapeutic agents that are drugs used for the treatment of arthritis include etanercept, infliximab, adalimumab, celecoxib, ituximab, abatacept, etoricoxib, golimumab, ofatumumab, and certolizumab pegol.
[0171] Examples of therapeutic agents that are drugs used for the treatment of blood deficiencies include pegfilgrastim, GCSF, PEG-GCSF, Darbepoetin alfa, Epoetin, Heparin (including low molecular weight derivatives), and warfarin.
[0172] Examples of therapeutic agents that are drugs used for the treatment of mucositis include Palifermin.
[0173] Examples of protein therapeutic agents are also monoclonal antibodies, a polyclonal antibodies, humanized antibodies, antibody fragments, and
immunoglobins.
[0174] Examples of therapeutic agents that are beneficial for RNA interference include, but are not limited to siRNA, dsDNA, miRNA, and antisense RNA.
[0175] Examples of therapeutic agents that are antibodies or their fragments include Abciximab, Adalimumab, Alemtuzumab, Basiliximab, Bevacizumab, Cetuximab, Certolizumab, Daclizumab, Eculizumab, Efalizumab, Gemtuzumab, Ibritumomab tiuxetan, Infliximab, Muromonab-CD3, Natalizumab, Omalizumab, Palivizumab, Panitumumab, Ranibizumab, Rituximab, Tositumomab, and Trastuzumab.
[0176] Examples of therapeutic agents that are PEGylated drugs include
Peginterferon alfa-2a, Peginterferon alfa-2b, Pegaspargase, and Pegfilgrastim.
[0177] Examples of therapeutic agents that are small molecules include
Atorvastatin, Clopidrogel, Aripiprazole, Esomeprazole, Olanzapine, Quetiapine, Rosuvastatin, Monte lukast, Venlafaxine Enoxaparin, and Pioglitazone.
Compositions and Formulations
[0178] In another aspect, the present technology provides compositions comprising or consisting essentially of a nanoparticle of the present technology and a carrier, diluent, or excipient. In another embodiment, the carrier, diluent, or excipient is pharmaceutically acceptable. A variety of carrier, diluent, or excipient,
pharmaceutically acceptable or not, are well known to one of skill in the art. [0179] The nanoparticle may comprise an agent or agents which in turn are compounds or isomers, prodrug, tautomer, or pharmaceutically acceptable salts thereof, of the present technology can be formulated in the pharmaceutically acceptable compositions per se, or in the form of a hydrate, solvate, N-oxide, or pharmaceutically acceptable salt, as described herein. Typically, such salts are more soluble in aqueous solutions than the corresponding free acids and bases, but salts having lower solubility than the corresponding free acids and bases may also be formed. The present technology includes within its scope solvates of the compounds and salts thereof, for example, hydrates.
[0180] In one embodiment, the present technology provides a pharmaceutically acceptable composition (formulation) comprising a nanoparticle and at least one pharmaceutically acceptable excipient, diluent, preservative, stabilizer, or mixture thereof.
[0181] In one embodiment, the methods can be practiced as a therapeutic approach towards the treatment of the conditions described herein. Thus, in a specific embodiment, the compounds of the present technology can be used to treat the conditions described herein in animal subjects, including humans. The methods generally comprise administering to the subject a nanoparticle of the present technology, or a salt, prodrug, hydrate, or N-oxide thereof, effective to treat the condition. As used herein, prodrug of a compound of the present technology is a compound that is converted in vivo or in vitro to the compound of the present technology. Hydrolysis, oxidation, and/or reduction are some ways that a prodrug is converted to the compound of the present technology.
[0182] In some embodiments, the subject is a non-human mammal, including, but not limited to, bovine, horse, feline, canine, rodent, or primate. In another embodiment, the subject is a human.
[0183] The nanoparticles of the present technology can be provided in a variety of formulations and dosages. It is to be understood that reference to the compound of the present technology, or "active" in discussions of formulations is also intended to include, where appropriate as known to those of skill in the art, formulation of the salts and prodrugs of the compounds. [0184] Pharmaceutically acceptable compositions comprising the nanoparticles described herein (or salts or prodrugs thereof) can be manufactured by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping, or lyophilization processes. The compositions can be formulated in conventional manner using one or more physiologically acceptable carriers, diluents, excipients, or auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically.
[0185] The nanoparticles of the present technology can be administered by oral, parenteral (e.g., intramuscular, intraperitoneal, intravenous, ICV, intracisternal injection or infusion, subcutaneous injection, or implant), by inhalation spray, nasal, vaginal, rectal, sublingual, urethral (e.g., urethral suppository) or topical routes of administration (e.g., gel, ointment, cream, aerosol, etc.) and can be formulated, alone or together, in suitable dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants, excipients, and vehicles appropriate for each route of administration.
[0186] The pharmaceutically acceptable compositions for the administration of the compounds can be conveniently presented in unit dosage form and can be prepared by any of the methods well known in the art. The pharmaceutically acceptable compositions can be, for example, prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier, a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation. In the pharmaceutical composition the active object compound is included in an amount sufficient to produce the desired therapeutic effect. For example, pharmaceutically acceptable compositions of the present technology may take a form suitable for virtually any mode of administration, including, for example, topical, ocular, oral, buccal, systemic, nasal, injection, transdermal, rectal, and vaginal, or a form suitable for administration by inhalation or insufflation.
[0187] For topical administration, the compound(s), salt(s) or prodrug(s) can be formulated as solutions, gels, ointments, creams, suspensions, etc., as is well-known in the art. [0188] Systemic pharmaceutically acceptable compositions include those designed for administration by injection (e.g., subcutaneous, intravenous, intramuscular, intrathecal, or intraperitoneal injection) as well as those designed for transdermal, transmucosal, oral, or pulmonary administration.
[0189] Useful injectable pharmaceutically acceptable compositions include sterile suspensions, solutions, or emulsions of the active compound(s) in aqueous or oily vehicles. The pharmaceutically acceptable compositions may also contain formulating agents, such as suspending, stabilizing, and/or dispersing agents. The formulations for injection can be presented in unit dosage form, e.g., in ampules or in multidose containers, and may contain added preservatives.
[0190] Alternatively, the injectable pharmaceutically acceptable compositions can be provided in powder form for reconstitution with a suitable vehicle, including but not limited to sterile pyrogen free water, buffer, and dextrose solution, before use. To this end, the active compound(s) can be dried by any art-known technique, such as lyophilization, and reconstituted prior to use.
[0191] For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the pharmaceutically acceptable compositions. Such penetrants are known in the art.
[0192] For oral administration, the pharmaceutically acceptable compositions may take the form of, for example, lozenges, tablets, or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., pregelatinized maize starch, polyvinylpyrrolidone, or hydroxypropyl
methylcellulose); fillers (e.g., lactose, micro crystalline cellulose, or calcium hydrogen phosphate); lubricants (e.g., magnesium stearate, talc, or silica); disintegrants (e.g., potato starch or sodium starch glycolate); or wetting agents (e.g., sodium lauryl sulfate). The tablets can be coated by methods well known in the art with, for example, sugars, films, or enteric coatings. Additionally, the pharmaceutically acceptable compositions containing the compounds of the present technology or prodrug thereof in a form suitable for oral use may also include, for example, troches, lozenges, aqueous, or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs. [0193] Pharmaceutically acceptable compositions intended for oral use can be prepared according to any method known to the art for the manufacture of
pharmaceutically acceptable compositions, and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents, and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient (including drug and/or prodrug) in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. These excipients can be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents (e.g., corn starch or alginic acid); binding agents (e.g. starch, gelatin, or acacia); and lubricating agents (e.g., magnesium stearate, stearic acid, or talc). The tablets can be left uncoated or they can be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate can be employed. They may also be coated by the techniques described in the U.S. Pat. Nos. 4,256,108; 4,166,452; and 4,265,874 to form osmotic therapeutic tablets for control release. The pharmaceutically acceptable compositions of the present technology may also be in the form of oil-in-water emulsions.
[0194] Liquid pharmaceutically acceptable compositions (or liquid preparations) for oral administration may take the form of, for example, elixirs, solutions, syrups, or suspensions, or they can be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations can be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, cellulose derivatives, or hydrogenated edible fats);
emulsifying agents (e.g., lecithin, or acacia); nonaqueous vehicles (e.g., almond oil, oily esters, ethyl alcohol, cremophore™, or fractionated vegetable oils); and preservatives (e.g., methyl or propylphydroxybenzoates or sorbic acid). The preparations may also contain buffer salts, preservatives, flavoring, coloring, and sweetening agents as appropriate. Preparations for oral administration can be suitably formulated to give controlled release or sustained release of the active compound, as is well known. The sustained release formulations (or sustained release pharmaceutically acceptable compositions) of the present technology are preferably in the form of a compressed tablet comprising an intimate mixture of compound of the present technology and a partially neutralized pH-dependent binder that controls the rate of compound dissolution in aqueous media across the range of pH in the stomach (typically approximately 2) and in the intestine (typically approximately about 5.5).
[0195] To provide for a sustained release of compounds of the present technology, one or more pH-dependent binders can be chosen to control the dissolution profile of the sustained release pharmaceutically acceptable compositions so that such pharmaceutically acceptable compositions release compound slowly and continuously as the pharmaceutically acceptable compositions are passed through the stomach and gastrointestinal tract. Accordingly, the pH-dependent binders suitable for use in the present technology are those which inhibit rapid release of drug from a tablet during its residence in the stomach (where the pH is-below about 4.5), and which promotes the release of a therapeutic amount of the compound of the present technology from the dosage form in the lower gastrointestinal tract (where the pH is generally greater than about 4.5). Many materials known in the pharmaceutical art as "enteric" binders and coating agents have a desired pH dissolution properties. The examples include phthalic acid derivatives such as the phthalic acid derivatives of vinyl polymers and copolymers, hydroxyalkylcelluloses, alkylcelluloses, cellulose acetates,
hydroxyalkylcellulose acetates, cellulose ethers, alkylcellulose acetates, and the partial esters thereof, and polymers and copolymers of lower alkyl acrylic acids and lower alkyl acrylates, and the partial esters thereof. One or more pH-dependent binders present in the sustained release formulation of the present technology are in an amount ranging from about 1 to about 30 wt %, about 5 to about 12 wt % and about 10 wt %.
[0196] One or more pH-independent binders may be in used in oral sustained release pharmaceutically acceptable compositions of the present technology. The pH- independent binders can be present in the pharmaceutically acceptable compositions of the present technology in an amount ranging from about 1 to about 10 wt %, from about 1 to about 3 wt % and about 2 wt %.
[0197] The sustained release pharmaceutically acceptable compositions of the present technology may also contain pharmaceutically acceptable excipients intimately admixed with the compound and the pH-dependent binder.
Pharmaceutically acceptable excipients may include, for example, pH-independent binders or film-forming agents such as hydroxypropyl methylcellulose,
hydroxypropyl cellulose, methylcellulose, polyvinylpyrrolidone, neutral
poly(meth)acrylate esters, starch, gelatin, sugars, carboxymethylcellulose, and the like. Other useful pharmaceutical excipients include diluents such as lactose, mannitol, dry starch, microcrystalline cellulose and the like; surface active agents such as polyoxyethylene sorbitan esters, sorbitan esters and the like; and coloring agents and flavoring agents. Lubricants (such as talc and magnesium stearate) and other tableting aids can also be optionally present.
[0198] The sustained release pharmaceutically acceptable compositions of the present technology have a compound of the present technology in the range of about 50% by weight to about 95% or more by weight, about 70% to about 90%> by weight; a pH-dependent binder content of between 5% and 40%, between 5% and 25%, and between 5% and 15%; with the remainder of the dosage form comprising pH- independent binders, fillers, and other optional excipients.
[0199] For buccal administration, the pharmaceutically acceptable compositions may take the form of tablets or lozenges formulated in the conventional manner.
[0200] For rectal and vaginal routes of administration, the active compound(s) can be formulated as solutions (for retention enemas), suppositories, or ointments containing conventional suppository bases such as cocoa butter or other glycerides.
[0201] For nasal administration or administration by inhalation or insufflation, the active compound(s) or prodrug(s) can be conveniently delivered in the form of an aerosol spray from pressurized packs or a nebulizer with the use of a suitable propellant (e.g., dichlorodifluoromethane, trichlorofluoromethane,
dichlorotetrafluoroethane, fluorocarbons, carbon dioxide, or other suitable gas). In the case of a pressurized aerosol, the dosage unit can be determined by providing a valve to deliver a metered amount. Capsules and cartridges for use in an inhaler or insufflator (for example, capsules and cartridges comprised of gelatin) can be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
[0202] The pharmaceutically acceptable compositions can be in the form of a sterile injectable aqueous or oleaginous suspension. This suspension can be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent. Among the acceptable vehicles and solvents that can be employed are water, Ringer's solution, and isotonic sodium chloride solution. The compounds may also be administered in the form of suppositories for rectal or urethral administration of the drug.
[0203] For topical use, creams, ointments, jellies, gels, solutions, suspensions, etc., containing the nanoparticles of the present technology, can be employed. In some embodiments, the compounds of the present technology can be formulated for topical administration with polyethylene glycol (PEG). These formulations may optionally comprise additional pharmaceutically acceptable ingredients such as diluents, stabilizers, and/or adjuvants.
[0204] Included among the devices which can be used to administer nanoparticles of the present technology, are those well-known in the art, such as metered dose inhalers, liquid nebulizers, dry powder inhalers, sprayers, thermal vaporizers, and the like. Other suitable technology for administration of particular nanoparticles of the present technology includes electrohydrodynamic aerosolizers. As those skilled in the art will recognize, the formulation of nanoparticles, the quantity of the formulation delivered, and the duration of administration of a single dose depend on the type of inhalation device employed as well as other factors. For some aerosol delivery systems, such as nebulizers, the frequency of administration and length of time for which the system is activated will depend mainly on the concentration of
nanoparticles in the aerosol. For example, shorter periods of administration can be used at higher concentrations of nanoparticles in the nebulizer solution. Devices such as metered dose inhalers can produce higher aerosol concentrations and can be operated for shorter periods to deliver the desired amount of nanoparticles in some embodiments. Devices such as dry powder inhalers deliver active agent until a given charge of agent is expelled from the device. In this type of inhaler, the amount of nanoparticles in a given quantity of the powder determines the dose delivered in a single administration.
[0205] Pharmaceutically acceptable compositions of the nanoparticles of the present technology for administration from a dry powder inhaler may typically include a finely divided dry powder containing nanoparticles, but the powder can also include a bulking agent, buffer, carrier, excipient, another additive, or the like. Additives can be included in such a dry powder composition of nanoparticles of the present technology, for example, to dilute the powder as required for delivery from the particular powder inhaler, to facilitate processing of the formulation, to provide advantageous powder properties to the formulation, to facilitate dispersion of the powder from the inhalation device, to stabilize the formulation (e.g., antioxidants or buffers), to provide taste to the formulation, or the like. Typical additives include mono-, di-, and polysaccharides; sugar alcohols and other polyols, such as, for example, lactose, glucose, raffinose, melezitose, lactitol, maltitol, trehalose, sucrose, mannitol, starch, or combinations thereof; surfactants, such as sorbitols,
diphosphatidyl choline, or lecithin; and the like.
[0206] For prolonged delivery, the nanoparticle(s) or prodrug(s) of the present technology can be formulated as a depot preparation for administration by
implantation or intramuscular injection. The active ingredient can be formulated with suitable polymeric or hydrophobic materials (e.g., as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives (e.g., as a sparingly soluble salt). Alternatively, transdermal delivery systems manufactured as an adhesive disc or patch which slowly releases the active nanoparticle(s) for percutaneous absorption can be used. To this end, permeation enhancers can be used to facilitate transdermal penetration of the active nanoparticle(s). Suitable transdermal patches are described in, for example, U.S. Patent No. 5,407,713.; U.S. Patent No. 5,352,456; U.S. Patent No. 5,332,213; U.S. Patent No. 5,336,168; U.S. Patent No. 5,290,561; U.S. Patent No. 5,254,346; U.S. Patent No. 5,164,189; U.S. Patent No. 5,163,899; U.S. Patent No. 5,088,977; U.S. Patent No. 5,087,240; U.S. Patent No. 5,008,110; and U.S. Patent No. 4,921,475.
[0207] Alternatively, other pharmaceutical delivery systems can be employed.
Liposomes and emulsions are well-known examples of delivery vehicles that can be used to deliver active nanoparticle(s) or prodrug(s). Certain organic solvents such as dimethylsulfoxide (DMSO) may also be employed, for example for topical administration, although usually at the cost of greater toxicity.
[0208] The pharmaceutical compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active nanoparticle(s). The pack may, for example, comprise metal or plastic foil, such as a blister pack. The pack or dispenser device can be accompanied by instructions for administration.
[0209] The nanoparticles described herein, or compositions thereof, will generally be used in an amount effective to achieve the intended result, for example, in an amount effective to treat or prevent the particular condition being treated. The nanoparticles can be administered therapeutically to achieve therapeutic benefit or prophylactically to achieve prophylactic benefit. By therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated and/or eradication or amelioration of one or more of the symptoms associated with the underlying disorder such that the patient reports an improvement in feeling or condition, notwithstanding that the patient may still be afflicted with the underlying disorder. Therapeutic benefit also includes halting or slowing the progression of the disease, regardless of whether improvement is realized.
[0210] The amount of nanoparticle administered will depend upon a variety of factors, including, for example, the particular condition being treated, the mode of administration, the severity of the condition being treated, the age and weight of the patient, the bioavailability of the particular active nanoparticle. Determination of an effective dosage is well within the capabilities of those skilled in the art. As known by those of skill in the art, the preferred dosage of nanoparticles of the present technology will also depend on the age, weight, general health, and severity of the condition of the individual being treated. Dosage may also need to be tailored to the sex of the individual and/or the lung capacity of the individual, where administered by inhalation. Dosage, and frequency of administration of the nanoparticles or prodrugs thereof, will also depend on whether the nanoparticles are formulated for treatment of acute episodes of a condition or for the prophylactic treatment of a disorder. A skilled practitioner will be able to determine the optimal dose for a particular individual.
[0211] For prophylactic administration, the nanoparticle can be administered to a patient at risk of developing one of the previously described conditions.
Alternatively, prophylactic administration can be applied to avoid the onset of symptoms in a patient diagnosed with the underlying disorder.
[0212] Effective dosages can be estimated initially from in vitro assays. For example, an initial dosage for use in animals can be formulated to achieve a circulating blood or serum concentration of active nanoparticle that is at or above an IC50 of the particular nanoparticle as measured in as in vitro assay. Calculating dosages to achieve such circulating blood or serum concentrations taking into account the bioavailability of the particular nanoparticle is well within the capabilities of skilled artisans. For guidance, the reader is referred to Fingl & Woodbury, "General Principles," GOODMAN AND GILMAN'S THE PHARMACEUTICAL BASIS OF THERAPEUTICS, Chapter 1, pp. 1-46, latest edition, Pergamon Press, and the references cited therein.
[0213] Initial dosages can also be estimated from in vivo data, such as animal models. Certain animal models useful for testing the efficacy of nanoparticles to treat or prevent the various diseases described above are well-known in the art. Ordinarily skilled artisans can routinely adapt such information to determine dosages suitable for human administration.
[0214] Dosage amounts will typically be in the range of from about 0.0001 or about 0.001 or about 0.01 mg/kg/day to about 100 mg/kg/day, but can be higher or lower, depending upon, among other factors, the activity of the nanoparticle, its
bioavailability, the mode of administration, and various factors discussed above. Dosage amount and interval can be adjusted individually to provide levels in the organ system of interest of the nanoparticle(s) which are sufficient to maintain therapeutic or prophylactic effect. For example, the nanoparticles can be
administered once per week, several times per week (e.g., every other day), once per day, or multiple times per day, depending upon, among other things, the mode of administration, the specific indication being treated, and the judgment of the prescribing physician. In cases of local administration or selective uptake, such as local topical administration, the effective local concentration of active nanoparticle(s) may not be related to plasma concentration. Skilled artisans will be able to optimize effective local dosages without undue experimentation.
[0215] The nanoparticle(s) useful in the treatment methods of the present technology will provide therapeutic or prophylactic benefit without causing substantial toxicity. Toxicity of the nanoparticle(s) can be determined using standard pharmaceutical procedures. The dose ratio between toxic and therapeutic (or prophylactic) effect is the therapeutic index. In certain embodiments, the
nanoparticles(s) exhibit high therapeutic indices as pertinent to the disease treated.
[0216] The foregoing disclosure pertaining to the dosage requirements for the nanoparticles of the present technology is pertinent to dosages required for prodrugs, with the realization, apparent to the skilled artisan, that the amount of prodrug(s) administered will also depend upon a variety of factors, including, for example, the bioavailability of the particular prodrug(s) and the conversation rate and efficiency into active drug nanoparticle under the selected route of administration.
Determination of an effective dosage of prodrug(s) for a particular use and mode of administration is well within the capabilities of those skilled in the art.
[0217] Also provided are kits for administration of the nanoparticles of the present technology, prodrug thereof, or pharmaceutical formulations comprising the nanoparticle that may include a dosage amount of at least one nanoparticle or a composition comprising at least one nanoparticle, as disclosed herein. Kits may further comprise suitable packaging and/or instructions for use of the nanoparticle. Kits may also comprise a means for the delivery of the at least one nanoparticle or compositions comprising at least one nanoparticle of the present technology, such as an inhaler, spray dispenser (e.g., nasal spray), syringe for injection, or pressure pack for capsules, tablets, suppositories, or other device as described herein.
[0218] Other types of kits provide the nanoparticle and reagents to prepare a composition of the present technology for administration. The composition can be in a dry or lyophilized form or in a solution, particularly a sterile solution. When the composition is in a dry form, the reagent may comprise a pharmaceutically acceptable diluent for preparing a liquid formulation. The kit may contain a device for administration or for dispensing the compositions, including, but not limited to, syringe, pipette, transdermal patch, or inhalant.
[0219] The kits may include other therapeutic nanoparticles for use in conjunction with the nanoparticles of the present technology described herein. These
nanoparticles can be provided in a separate form or mixed with the nanoparticles of the present technology. The kits will include appropriate instructions for preparation and administration of the composition, side effects of the compositions, and any other relevant information. The instructions can be in any suitable format, including, but not limited to, printed matter, videotape, computer readable disk, or optical disc.
[0220] In one embodiment, the present technology provides a kit comprising a nanoparticle selected from the nanoparticles of the present technology or a salt or prodrug thereof, packaging, and instructions for use.
[0221] In another embodiment, the present technology provides a kit comprising the pharmaceutically acceptable composition comprising a nanoparticle selected from the nanoparticles of the present technology or a salt or prodrug thereof and at least one pharmaceutically acceptable excipient, diluent, preservative, stabilizer, or mixture thereof, packaging, and instructions for use. In another embodiment, kits for treating an individual who suffers from or is susceptible to the conditions described herein are provided, comprising a container comprising a dosage amount of a nanoparticle or composition of the present technology, as disclosed herein, and instructions for use. The container can be any of those known in the art and appropriate for storage and delivery of oral, intravenous, topical, rectal, urethral, or inhaled formulations. [0222] Kits may also be provided that contain sufficient dosages of the
nanoparticles or composition to provide effective treatment for an individual for an extended period, such as a week, 2 weeks, 3, weeks, 4 weeks, 6 weeks, or 8 weeks or more.
[0223] The technology having been described in summary and in detail is illustrated and not limited by the examples below.
Examples
Example 1 - Synthesis of Dextran Succinate (DS)
[0224] 70 kDa Dextran (10 g) was stirred in dry dimethylsulfoxide (100 mL) and pyridine (15 mL). Succinic anhydride (1.54 g) was added and the mixture, which became a homogenous solution after 1 hour, was stirred at room temperature under argon for 16 hours. The solution was poured into stirred ethyl acetate (400 mL), and then acetone (400 mL) was added and stirring was continued for 16 hours, during which the pasty precipitate eventually became granular. The precipitate was filtered, washed with ethyl acetate and dried under vacuum to afford a white solid, which was dissolved in water (250 mL). The aqueous solution was acidified with dilute HC1 to pH 2 and 5x diafiltered with water using a 0.1 m TFF (tangential flow filtration) module with a 5 kDa MWCO membrane. The solution was then concentrated to ~50 mL by TFF and lyophilized to afford dextran 20% succinate as a white solid (10.2 g). ιΐί NMR analysis confirmed that the product contained 0.2 equivalents of succinate per anhydroglucose unit (20% succinylation).
Example 2 - Synthesis of VB12-Dextran Succinate Conjugate (Cob-DS)
[0225] 70 kDa Dextran 20% succinate (200 mg) and aminohexyl-VB12 (20 mg) were dissolved in water (8 mL). l-Ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride (200 mg) and N-hydroxysuccinimide (200 mg) were added and the solution (pH 5.5) was stirred for 16 hours. The mixture was centrifuged in a 5 kDa Amicon-15 centrifugal filter at 4800 rpm for 45 min. Water (15 mL) was added to the retentate and centrifuged; then the 15 mL wash was repeated once more. The washed retentate was lyophilized to afford Cob-DS (223 mg) as a pale red solid. UV-VIS spectrophotometric analysis revealed the product contained 3.25 % w/w of VB12, which corresponds to -0.5 equivalents of AH-VB12 per 100 anhydroglucose units (0.5 mol% VB12).
Example 3 - Synthesis of Carboxymethyl Dextran (CMD)
[0226] A solution of 70 kDa dextran (4.0 g) in 1 1% sodium hydroxide (20 mL) was added to a solution of chloroacetic acid (2.3 g) in tert-butanol (40 mL) and the biphasic mixture was stirred vigorously at 60°C for 3 hours. After cooling to room temperature, the mixture was poured into stirring acetone (400 mL) and the resulting pasty precipitate was separated by decantation. The paste was dissolved in water (25 mL) and poured into stirring methanol (300 mL) and the resulting white precipitate was filtered, washed with methanol and dried under vacuum. The crude product was dissolved in water and 5x diafiltered with water using a 0.1 m TFF (tangential flow filtration) module with a 5 kDa MWCO membrane. The solution was then concentrated by TFF and lyophilized to afford a white solid (4.6 g). 1H NM analysis revealed that the product contained 0.2 carboxy-methyl equivalents per anhydroglucose unit (20% carboxymethylation).
Example 4 - Synthesis of VB12-Carboxymethyl Dextran Conjugate (Cob-CMD)
[0227] 20% Carboxymethyl 70 kDa dextran (200 mg) and aminohexyl-VB 12 (50 mg) were dissolved in water (10 mL). l-Ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride (ED AC; 60 mg) and N-hydroxysuccinimide (NHS;
15 mg) were added and the solution was stirred for 20 hours at pH 5.5. The mixture was centrifuged in a 5 kDa Amicon-15 centrifugal filter at 3800 rpm for 45 min. Water (15 mL) was added to the retentate and centrifuged, and then the 15 mL wash was repeated once more. The retentate was lyophilized to afford Cob-CMD (224 mg) as a pale red solid. UV-VIS spectrophotometric analysis revealed the product contained 12% w/w of VB12, which corresponds to -1.7 equivalents of AH-VB12 per 100 anhydroglucose units (1.7 mol%> VB12).
Example 5 - Synthesis of N,N-Dimethyl Chitosan (DMC)
[0228] Chitosan (Aldrich Low MW; 10 g) was suspended in water (180 mL) and formaldehyde (40 mL) and formic acid (30 mL) were added. The mixture was heated at 70°C for 24 hours, evolving copious quantities of gas (C02). Further portions of formaldehyde (40 mL) and formic acid (30 mL) were added and the mixture heated at 70°C for another 24 hours, at which time gas evolution had ceased completely. Water (200 mL) was added and the solution was filtered through Celite, then subjected to tangential flow filtration with a 5 kDa MWCO membrane, concentrated by TFF and lyophilized to afford N,N-dimethyl chitosan as a white solid (10 g). 1H NM analysis revealed that all of the non-acetylated amine groups of chitosan had been converted to dimethylamino groups.
Example 6 - Synthesis of N,N,N-Trimethyl Chitosan (TMC)
[0229] N,N-Dimethyl chitosan (1 g) was suspended in N-methyl pyrrolidone
(50 mL) and the mixture was heated at 80°C for 20 min, then cooled. Methyl iodide (3 mL) was added and the mixture maintained at 40°C for 18 hours. The reaction was slowly added to a mixture of ether (100 mL) and ethanol (100 mL) and stirred for 1 hour. The resulting solid was isolated by centrifugation, washed with ether and dried to afford crude TMC iodide (1.4 g) as a brown solid. The solid was dissolved in 1 M NaCl in 0.1 M HC1 (100 mL), filtered, subjected to 5 kDa MWCO diafiltration with 1 M NaCl in 0.1 M HC1 (4x), then water (4x). The solution was concentrated by TFF and lyophilized to afford N,N,N-trimethyl chitosan chloride as a white solid (0.6 g). ιΐί NMR analysis revealed that the product contained 20% acetylated amine groups, 39% dimethylamino groups and 41% trimethylammonium groups (degree of quaternization DQ = 41%).
Example 7 - Synthesis of O-Carboxymethyl N,N,N-Trimethyl Chitosan (CMTMC)
[0230] Sodium hydride 60%> suspension (230 mg) was added to dry DMSO
(20 mL)and the mixture was heated at 70°C for 1 hour. After cooling, crude trimethyl chitosan iodide (500 mg) was added and the mixture stirred at room temperature for 1.5 hours. Chloroacetic acid (238 mg) was added and the reaction stirred for 12 hours, then poured into stirring ether (200 mL). The resulting precipitate was isolated by centrifugation, washed with ether and dried to afford a white solid (1 g). The solid was dissolved in 1 M NaCl in 0.1 M HC1 (40 mL), filtered, subjected to tangential flow filtration with a 5 kDa MWCO membrane, concentrated by TFF and lyophilized to afford O-carboxymethyl N,N,N-trimethyl chitosan (CMTMC) as a white solid (229 mg). 13C NMR analysis confirmed the presence of carboxymethyl groups. Example 8 - Synthesis of VB12 -Carboxymethyl Trimethyl Chitosan (Cob-CMTMC)
[0231] 0-Carboxymethyl N,N,N-trimethyl chitosan (180 mg) was dissolved in water (10 mL) and the solution adjusted to pH 5. Aminohexyl-VB12 (55 mg), ED AC (50 mg) and NHS (50 mg) were added and the solution was stirred for 20 hours maintained at pH 5.5. The mixture was subjected to centrifugal ultrafiltration in a 3 kDa Amicon-15 centrifugal filter and washed with water. The retentate was lyophilized to afford VB12-CM-TMC (133 mg) as a pale red solid.
Example 9 - Preparation of VB12-Dextran Succinate Insulin Nanoparticles
[0232] The dextran derivative of example 2 (145.8 mg) was added to a solution of bovine insulin in dilute HCl (1.8 mg/mL; 2.5 mL) and agitated gently for 1 hour at room temperature, then lyophilized to afford DS Cobalamin insulin nanoparticles as a pale red solid. econstitution of the solid in 2.5 mL of water provided insulin dosages of 50 IU per mL.
Example 10 - Preparation of VB12-PEC Insulin Nanoparticles
[0233] A solution of recombinant human insulin in pH 7.4 phosphate buffer (11.2 mg/mL; 1.0 mL) was added to a solution of trimethyl chitosan chloride in water (6.7 mg/mL; 5.0 mL) and gently stirred for 30 min forming a light suspension. The mixture was adjusted to pH 7.4 and a solution of VB12-carboxymethyl dextran of example 4 in pH 7.4 phosphate buffer (10.0 mg/mL; 0.1 mL) was added; stirring for 5 min yielded a pale red suspension of the VB12-PEC insulin nanoparticles (z-average diameter 112 nm) containing 47 IU per mL of insulin.
Example 11 - Preparation of VB12-PEC hGH Nanoparticles
[0234] A solution of 35 mg/mL Somatropin (47.2 mg) was added to a stirring solution of chitosan (16 mg) in pH 5.5 water (2.0 mL). To this solution was added with stirring a solution of VB12-carboxymethyl dextran of example 4 (16.1 mg) in water (2.0 mL). The resulting pale red VB12-PEC hGH nanoparticle suspension containing 0.4 mg/mL of hGH.
Example 12 - Preparation of PEC Polynucleotide Nanoparticles
[0235] A solution of fish sperm DNA in pH 7.4 phosphate buffer (1.0 mg/mL; 0.5 mL) was added to a solution of trimethyl chitosan chloride in pH 7.4 phosphate buffer (0.1 mg/mL; 10.0 mL) and gently stirred for 30 min forming a light suspension of TMC DNA nanoparticles (z-average diameter 131 nm).
Example 13 - Comparision of Blood Glucose Reduction in Rats by oral and s.c. insulin
[0236] Female Wistar rats (200 g) were housed at room temperature with 12 h light/dark cycle. All animals had ad libitum access to a standard chow diet and water except wherever indicated. The rats were allowed to acclimatize for a period of 7 days in the new environment before initiation of the experiment. After randomization into groups of 4, each rat was marked and followed individually throughout the study.
[0237] All rats were fasted for 1 hour. Streptozotocin obtained from Sigma (98% HPLC) was administered by IV injection at a dose of 55-65 mg/kg in 0.1 M citrate buffer (pH 4.5) followed by an additional 1 hour fast. Blood was collected daily from the tail and blood glucose levels were measured using an Accu-Chek® (Compact Plus-Roche) blood glucose monitor. When all animals in the group had achieved a blood glucose level >250 mg/dl (3-5 days) the insulin metabolism phase was begun.
[0238] All animals were fasted for 1 hour and a blood glucose measurement taken. Groups of rats were dosed s.c. with bovine insulin and (by oral gavage) an aqueous preparation of the nanoparticles of example 9 , or an aqueous solution of bovine insulin, according to the table below. The animals were then fasted for another hour. Additional blood glucose levels were taken at timepoints listed in the table below.
Table 1
Figure imgf000059_0001
[0239] Administration of insulin by subcutaneous injection, or as an oral nanoparticle formulation (groups 2 and 3) resulted in a reduction of blood glucose (BG) levels from an initial BG of approximately 400 μg/ml to less than 200 μg/ml. The results show a significant reduction in BG compared to oral administration of unformulated insulin. Whereas, the reduction of BG from the subcutaneous administration was short-lived (less than 6 hours), the reduction of BG resulting from administration of insulin in a nanoparticle formulation was prolonged to beyond 24 hours. The dose given in the oral nanoparticle formulation was ten-fold higher than that given s.c. Assuming 100% bioavailability of insulin given s.c, then it is possible to estimate the oral bioavailability insulin using the relationship:
[0240] % pharmacological availability (PA) was calculated as follows:
[0241] PA = AAC oral x Dose sc x 100
[0242] AAC sc x Dose oral
[0243] Where AAC is area above the curve (area formed by the curve and a horizontal line extending from the t=0 value). Using simple triangulation to estimate AAC, PA > 80%
Example 14 - Comparision of Blood hGH levels in Rats following oral and s.c. hGH dosing
[0244] Male Sprague-Dawley JVC-IDC rats each had a surgically implanted jugular vein cannula (JVC) and an intraduodenal cannula (IDC) to bypass the stomach. Rats weighing approximately 100 g (30-35 days old) were housed at room temperature with a 12 h light/dark cycle. All animals had ad libitum access to a standard chow diet and water. The rats were allowed to acclimatize for a period of 4 days in the new environment before initiation of the experiment. All animals were weighed and numbered by ear clip. After a period of 4 days, all rats were weighed again and only those rats in good health and those that had not lost more than 10% of their initial body weight were used in the study. Since these were otherwise healthy animals, they had all gained significant weight since arrival (weights -150 to -180 g).
[0245] The rats were randomized into groups of 4. Test articles (exemplified by example 11) were administered in a volume of 3
Figure imgf000060_0001
by intraduodenal cannula or by subcutaneous injection, according to the table below. Blood samples (0.5 mL each) were collected via jugular vein catheter into Na2EDTA tubes and processed for plasma collection by centrifuging at -2,000 rpm for 10 min; samples were collected before administration (t=0) and at 20, 40, 60, 120 and 180 minutes after
administration for i.d. doses and at 10, 20, 30, 40 and 60 minutes after administration for the s.c. dose. The cellular fraction was discarded and the plasma samples stored at -20°C until processed for analysis.
[0246] Levels of hGH in plasma samples were quantitated using a commercial ELISA kit (Bio-Quant, Inc. San Diego, CA; Cat #BQ048H) according to the manufacturer's instructions.
Table 2
Figure imgf000061_0001
[0247] Blood levels of hGH achieved following oral dosing with one of the hGH nanoparticle formulations were significantly greater than obtained with unformulated hGH given orally (see Figure 3).
Example 15 - Preparation of VB12-TMC PEC Insulin Nanoparticles
[0248] A solution of recombinant human insulin in pH 7.4 phosphate buffer (4.0 mg/mL; 3.0 mL) was added to a solution of Cob-CMTMC (similar to example 8) in water (4.0 mg/mL; 3.0 mL) and gently stirred for 15 min forming a turbid pink suspension of VB12-TMC PEC insulin nanoparticles (z-average diameter 243 nm) containing 53 IU of insulin per mL.
Example 16 - Synthesis of VB12-Hypromellose Phthalate (Cob-HP)
[0249] Hypromellose phthalate HP-55 (500 mg) was dissolved in 0.1 M NaOH (20 mL) and the solution adjusted to pH 5.4 with dilute HC1. Aminohexyl-VB12 (50 mg) and NHS (15 mg) were added, followed by ED AC (24 mg), and the mixture was stirred for 72 hours at pH 5.8. The solution was subjected to 5 kDa MWCO TFF diafiltration with water (5x), concentrated by TFF and the retentate was lyophilized to afford VB12-hypromellose phthalate, Cob-HP (420 mg) as a pale red solid. UV-VIS spectrophotometric analysis revealed the product contained 2.6 % w/w of VB12.
Example 17 - Preparation of VB12-HP-TMC PEC Insulin Nanoparticles
[0250] A solution of recombinant human insulin in pH 7.4 phosphate buffer (5.0 mg/mL; 2.0 mL) was added to a solution of trimethyl chitosan TMC in pH 7.4 phosphate buffer (5.0 mg/mL; 4.0 mL). A solution of Cob-HP in pH 7.4 phosphate buffer (5.0 mg/mL; 1.5 mL) was added to the mixture and gently stirred for 15 min forming a turbid pink suspension of VB12-HP-TMC PEC insulin nanoparticles (z-average diameter 548 nm). The suspension was lyophilized to afford VB12-HP- TMC nanoparticles containing 6.3 IU of insulin per mg.
Example 18 - Knockdown of MSI-2 gene expression in K562 leukemia cells with VB12-PEC siRNA Nanoparticles
[0251] CMTMC (CobT-0), Cob-CMTMC (CobT-7) and Lipofectamine 2000 (as positive control) at indicated concentrations were complexed with siRNA (Ambion Silencer Select Cat#4392420, Lot #AS00L5SB, ID 42755) targeting MSI2 gene in serum-free media (RPMI1640, ATCC) for 30 minutes at RT.
[0252] K562 cells (ATCC, passage 12) were plated in U-bottom tissue culture 96- well plate 24 hour prior to transfection (5,000 cells per well; 180 volume per well).
[0253] Complexes (volume = 20 μί) were added to cells cultured in 96-well plates using RPMI 1640 (ATCC) medium supplemented with 10% FBS (ATCC) in 180 volume per well (final volume = 200 μΐ, per well). Cells were incubated at 37°C, 5% C02 for 48 hours.
[0254] Following 48-hour treatment, cells were centrifuged at 2,000xg and cell pellets were lysed in 77 μΐ, of Lysis Buffer (RNAqueous® MAGMax-96 kit). Total RNA from siRNA transfected cells was isolated using the RNAqueous® MAGMax- 96 kit (Ambion, Austin, TX). The purified, DNAse-treated RNA was reverse transcribed to make complementary DNA (cDNA) with random decamers using the RETROscript ¾ Kit (Ambion, Austin, TX). Gene expression levels were determined by real-time PCR on the ABI Prism 7900 SDS real-time PCR machine (Applied Biosystems, Foster City, CA). The MSI2 data was collected via a Taqman Gene Expression Assay specific to amplification of the MSI2 mRNA
(cat#Hs0292670_ml ; Applied Biosystems).
[0255] Fluorescence based real-time reverse transcription PC (qRT-PCR) is a standard tool used for quantification of mRNA levels. This technique has high throughput capabilities with both high sensitivity and specificity for the target of interest.
[0256] 18S rRNA was also amplified as an internal reference to adjust for well-to- well variances in the amount of starting template into the reverse transcription reaction (cat#Hs99999901_sl; Applied Biosystems). The MSI2 values were corrected to the 18S values (internal reference), and the corrected values were then normalized to a sample transfected with a egative Control siRNA (Ambion, Austin, TX). "% Remaining Expression Levels" were calculated as a percentage of gene expression relative to the negative control siRNA of similar trans fection volume. A value of 100% would represent an untreated sample. Values less than 100% exhibit the percent knockdown of the target.
Results:
[0257] 1 and 3 CobT-7 induced 19% and 53% MSI2 gene expression knockdown, respectively (see Figure 4). These values are similar to those induced by 1 μΐ^ and 3 Lipofectamine 2000 positive control (11% and 59%, respectively). However, the Lipofectamine 2000 induced significant toxicity at 3 μΐ^, while CobT-7 exhibited no toxicity up to 30 μΐ, i.e., 10 times the concentration (see Figure 5).
[0258] It is to be understood that while the invention has been described in conjunction with the above embodiments, that the foregoing description and examples are intended to illustrate and not limit the scope of the invention. Other aspects, advantages and modifications within the scope of the invention will be apparent to those skilled in the art to which the invention pertains.
[0259] Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All nucleotide sequences provided herein are presented in the 5' to 3' direction. [0260] The inventions illustratively described herein may suitably be practiced in the absence of any element or elements, limitation or limitations, not specifically disclosed herein. Thus, for example, the terms "comprising", "including,"
containing", etc. shall be read expansively and without limitation. Additionally, the terms and expressions employed herein have been used as terms of description and not of limitation, and there is no intention in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, but it is recognized that various modifications are possible within the scope of the invention claimed.
[0261] Thus, it should be understood that although the present invention has been specifically disclosed by preferred embodiments and optional features, modification, improvement and variation of the inventions embodied therein herein disclosed may be resorted to by those skilled in the art, and that such modifications, improvements and variations are considered to be within the scope of this invention. The materials, methods, and examples provided here are representative of preferred embodiments, are exemplary, and are not intended as limitations on the scope of the invention.
[0262] The invention has been described broadly and generically herein. Each of the narrower species and subgeneric groupings falling within the generic disclosure also form part of the invention. This includes the generic description of the invention with a proviso or negative limitation removing any subject matter from the genus, regardless of whether or not the excised material is specifically recited herein.
[0263] In addition, where features or aspects of the invention are described in terms of Markush groups, those skilled in the art will recognize that the invention is also thereby described in terms of any individual member or subgroup of members of the Markush group.
[0264] All publications, patent applications, patents, and other references mentioned herein are expressly incorporated by reference in their entirety, to the same extent as if each were incorporated by reference individually. In case of conflict, the present specification, including definitions, will control.

Claims

What is claimed is:
1. A nanoparticle comprising:
one or more synthetic or natural polymers comprising one or more charged and/or ionisable groups,
a therapeutic agent comprising one or more charged and/or ionisable groups of the opposite charge to the charge of the polymers, and,
a vitamin B12 covalently linked to the nanoparticle via an optional linker group.
2. The nanoparticle of claim 1, wherein the one or more charged and/or ionisable groups of the one or more synthetic or natural polymers are of positive charge or negative charge.
3. A nanoparticle comprising:
one or more synthetic or natural polymers comprising one or more charged and/or ionisable groups which are positively charged,
one or more synthetic or natural polymers comprising one or more charged and/or ionisable groups which are negatively charged,
a therapeutic agent, and vitamin B12 covalently linked to the nanoparticle via an optional linker group.
4. The nanoparticle of claim 1 or 3, wherein the vitamin B12 is attached to one or more of the charged and/or ionisable groups of the one or more synthetic or natural polymers.
5. The nanoparticle of claim 1 or 3, wherein the vitamin B12 is attached to the therapeutic agent.
6. The nanoparticle of claim 1 or 3, further comprising one or more of components selected from the group consisting of polyethylene glycol (PEG), PEG block copolymers, polyacrylic, polymethacrylic, polyacrylamide,
polymethacrylamide, synthetic polymer, polysaccharide, surfactant, and metal ions.
7. The nanoparticle of claim 6, wherein the vitamin B12 is attached to one or more of the components.
8. The nanoparticle of claim 1 or 3, wherein an average nanoparticle diameter is in a range of about 20 to about 800 nm.
9. The nanoparticle of any of the preceding claims, wherein the nanoparticle is configured for oral administration in a subject.
10. The nanoparticle of any of claims 1-8 , wherein the nanoparticle is configured for administration by injection to a subject.
11. The nanoparticle of any of the preceding claims, wherein the synthetic or natural polymer is selected from the group consisting of poly- -glutamic acid ( - PGA), succinylated and/or carboxymethylated polysaccharide, poly(acrylic acid), poly(p-styrene-sulfonic acid sodium salt), poly(sulfopropylacrylate), negatively- charged polypeptide, chitosan derivatives, polyethyleneimine, positively-charged polypeptide, pharmaceutically-approved polymeric excipients that carry a net negative charge at physiologically-relevant pH levels, and pharmaceutically-approved polymeric excipients that carry a net positive charge at physiologically-relevent pH levels.
12. The nanoparticle of any of claims 1-10, wherein the synthetic or natural polymer is selected from the group consisting of chitosan derivatives,
polyethyleneimine, positively-charged polypeptide, and pharmaceutically-approved polymeric excipients that carry a net positive charge at physiologically-relevent pH levels
13. The nanoparticle of any of claims 1-10, wherein the synthetic or natural polymer is selected from the group consisting of soly- -glutamic acid ( -PGA), succinylated and/or carboxymethylated polysaccharide, poly(acrylic acid), poly(p- styrene-sulfonic acid sodium salt), poly(sulfopropylacrylate), negatively-charged polypeptide, and pharmaceutically-approved polymeric excipients that carry a net negative charge at physiologically-relevent pH levels.
14. The nanoparticle of claim 11, 12, or 13, wherein the succinylated and/or carboxymethylated polysaccharide is dextran, cellulose, or starch.
15. The nanoparticle of claim 11, 12, or 13, wherein the negatively-charged polypeptide is polyglutamic acid.
16. The nanoparticle of claim 11, 12, or 13, wherein the positively-charged polypeptide is polylysine.
17. The nanoparticle of any of the preceding claims, wherein an axial ligand substituent on a cobalt atom of vitamin B12 is CN, Me, OH or NO.
18. The nanoparticle of any of the preceding claims, wherein the therapeutic agent is selected from the group consisting of a small or large synthetic molecule, protein, peptide, glycoprotein, nucleoside, nucleotide, humanized monoclonal antibody, non- humanized monoclonal antibody, therapeutically relevant fragments of humanized and/or non-humanized monoclonal antibody, and agents for effecting RNA
interference (RNAi) such as dsRNA, si NA and antisense RNA.
19. The nanoparticle of any of the preceding claims, wherein the therapeutic agent is selected from the group consisting of analgesic, antiallergenic, antianginal agent, antiarrythmic drug, antibiotic, anticoagulant, antidementia drug, antidepressant, antidiabetic, antihistamine, antihypertensive, anti-inflammatory, antineoplastic agent, antiparasitic, antipyretic, antiretroviral drug, antiulcerative agent, antiviral agent, cardiovascular drug, cholesterol-lowering agent, CNS active drug, a hormone, growth hormone inhibitor, growth hormone, hematopoietic drug, hemostatic, hypotensive diuretic, keratolytic, therapeutic for osteoporosis, vaccine, vasoconstrictor, and vasodilator.
20. A process for preparing a nanoparticle composition comprising the nanoparticle of any of the preceding claims, comprising:
combining the one or more synthetic or natural polymers, the therapeutic agent, and the vitamin B 12, in a suitable solvent, and
isolating, purifying, and/or drying the nanoparticles.
21. The process of claim 20, wherein the solvent is > 50% water.
22. A process for preparing a nanoparticle composition comprising the nanoparticle of any of the preceding claims, comprising: mixing two immiscible solvents and a surfactant to produce an emulsion,
optionally cross-linking the nanoparticles, and
isolating, purifying, and/or drying resultant nanoparticles.
23. The process of claim 20 or 22, wherein the nanoparticles are isolated by solvent evaporation.
24. The process of claim 20 or 22, wherein the nanoparticles are isolated by filtration or centrifugation.
25. The process of claim 20 or 22, wherein the nanoparticles are isolated by addition of a cosolvent followed by filtration or centrifugation.
26. The process of claim 20 or 22, wherein the purifying step is effected by washing the nanoparticles with a suitable solvent.
27. The process of claim 20, further comprising modifying the nanoparticles to effect cross-linking of the components of the nanoparticle.
28. The process of claim 20 or 22, further comprising modifying the nanoparticles to add a vitamin B12 analog to a surface of the nanoparticle by physical or covalent attachment.
29. The process of claim 20 or 22, further comprising modifying the nanoparticles to substitue an axial ligand on a one or more cobalt atoms of attached vitamin B12 with replacement axial ligands.
30. A pharmaceutical composition comprising the nanoparticle of claim 1 or 3, and a pharmaceutically-acceptable excipient.
31. The pharmaceutical composition of claim 30, wherein the composition is formulated as a tablet, a capsule, or a liquid.
32. The pharmaceutical composition of claim 30, wherein the composition is formulated as a lyophilized powder in a container for subsequent re-suspension or dissolution of the pharmaceutical composition in a pharmaceutically-acceptable injection vehicle.
33. The pharmaceutical composition of claim 30, wherein the composition is formulated as a suspension or solution in a pharmaceutically-acceptable injection vehicle.
34. A method for treating a subject, comprising administring an effective amount of the nanoparticle of any of claims 1-19 or the pharmaceutical composition of any of claims 30-33.
35. The method of claim 34, wherein the therapeutic agent is an antidiabetic agent.
36. The method of claim 34, wherein the therapeutic agent is a hormone.
37. The method of claim 34, wherein the therapeutic agent is an antineoplastic agent.
PCT/US2011/032793 2010-04-16 2011-04-15 A nanostructures containing vitamin b12 for facilitated delivery of drugs across biological barriers WO2011130716A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US32517410P 2010-04-16 2010-04-16
US61/325,174 2010-04-16

Publications (2)

Publication Number Publication Date
WO2011130716A2 true WO2011130716A2 (en) 2011-10-20
WO2011130716A3 WO2011130716A3 (en) 2012-06-28

Family

ID=44629084

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/032793 WO2011130716A2 (en) 2010-04-16 2011-04-15 A nanostructures containing vitamin b12 for facilitated delivery of drugs across biological barriers

Country Status (1)

Country Link
WO (1) WO2011130716A2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012030745A1 (en) * 2010-08-30 2012-03-08 Access Pharmaecuticals, Inc MULTIVITAMIN TARGETING OF RNAi THERAPEUTICS
WO2012122313A3 (en) * 2011-03-08 2013-03-14 Access Pharmaceuticals, Inc. Targeted nanocarrier systems for delivery of actives across biological membranes
WO2013123523A1 (en) * 2012-02-19 2013-08-22 Nvigen, Inc. Uses of porous nanostructure in delivery
WO2015126841A1 (en) 2014-02-18 2015-08-27 Plasmatech Biopharmaceuticals, Inc. Nutritional and therapeutic mucoadhesive formulations
WO2017053920A1 (en) 2015-09-25 2017-03-30 Zy Therapeutics Inc. Drug formulation based on particulates comprising polysaccharide-vitamin conjugate
US11160878B1 (en) * 2020-06-10 2021-11-02 Boris Farber Supramolecular systems based on dynamic self-organizing nanostructures with antiviral properties

Citations (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4166452A (en) 1976-05-03 1979-09-04 Generales Constantine D J Jr Apparatus for testing human responses to stimuli
US4256108A (en) 1977-04-07 1981-03-17 Alza Corporation Microporous-semipermeable laminated osmotic system
US4265874A (en) 1980-04-25 1981-05-05 Alza Corporation Method of delivering drug with aid of effervescent activity generated in environment of use
US4921475A (en) 1983-08-18 1990-05-01 Drug Delivery Systems Inc. Transdermal drug patch with microtubes
US5008110A (en) 1988-11-10 1991-04-16 The Procter & Gamble Company Storage-stable transdermal patch
US5087240A (en) 1983-08-18 1992-02-11 Drug Delivery Systems Inc. Transdermal drug patch with conductive fibers
US5088977A (en) 1988-12-21 1992-02-18 Drug Delivery Systems Inc. Electrical transdermal drug applicator with counteractor and method of drug delivery
US5108921A (en) 1989-04-03 1992-04-28 Purdue Research Foundation Method for enhanced transmembrane transport of exogenous molecules
US5163899A (en) 1987-03-20 1992-11-17 Drug Delivery Systems Inc. Transdermal drug delivery system
US5164189A (en) 1989-12-04 1992-11-17 G. D. Searle & Co. Single layer transdermal drug administration system
US5254346A (en) 1988-02-23 1993-10-19 Tucker Mark J Occlusive body for administering a physiologically active substance
US5332213A (en) 1992-01-29 1994-07-26 Franz Volkl Gmbh & Co. Ski Und Tennis Sport-Artkihelfabrik Kg. Ball-game racket, particularly a tennis racket
US5336168A (en) 1987-05-28 1994-08-09 Drug Delivery Systems Inc. Pulsating transdermal drug delivery system
US5352456A (en) 1991-10-10 1994-10-04 Cygnus Therapeutic Systems Device for administering drug transdermally which provides an initial pulse of drug
US5407713A (en) 1991-12-18 1995-04-18 Minnesota Mining And Manufacturing Company Multilayered barrier structures
US5428023A (en) 1985-10-10 1995-06-27 Biotechnology Australia Pty. Ltd. Oral delivery of biologically active substances bound to vitamin B12 or analogues thereof
US5449720A (en) 1993-05-24 1995-09-12 Biotech Australia Pty Limited Amplification of the VB12 uptake system using polymers
US5548064A (en) 1993-05-24 1996-08-20 Biotech Australia Pty Limited Vitamin B12 conjugates with EPO, analogues thereof and pharmaceutical compositions
US5574018A (en) 1994-07-29 1996-11-12 Amgen Inc. Conjugates of vitamin B12 and proteins
US5605890A (en) 1994-06-01 1997-02-25 Hybridon, Inc. Cyclodextrin cellular delivery system for oligonucleotides
US5688488A (en) 1989-04-03 1997-11-18 Purdue Research Foundation Composition and method for tumor imaging
US5807832A (en) 1987-06-09 1998-09-15 Biotech Australia Pty Limited Oral delivery of biologically active substances bound to vitamin B12
US5863900A (en) 1993-05-20 1999-01-26 Biotech Australia Pty., Ltd. Lhrh antagonists
US6083926A (en) 1994-04-08 2000-07-04 The University Of Washington Water soluble vitamin B12 receptor modulating agents and methods related thereto
US6150341A (en) 1998-06-12 2000-11-21 Biotch Australia Pty Limited Vitamin B12 derivatives and methods for their preparation
US6159502A (en) 1991-04-02 2000-12-12 Biotech Australia Pty Ltd Oral delivery systems for microparticles
US6221397B1 (en) 1997-08-29 2001-04-24 Biotech Australia Pty Limited Surface cross-linked particles suitable for controlled delivery
US20020115595A1 (en) 1996-08-27 2002-08-22 Grissom Charles B. Bioconjugates and delivery of bioactive agents
US6482413B1 (en) 2001-02-26 2002-11-19 Council Of Scientific And Industrial Research Vitamin B12 —biodegradable micro particulate conjugate carrier systems for peroral delivery of drugs, therapeutic peptides/proteins and vaccines
US7128893B2 (en) 2002-05-06 2006-10-31 Endocyte, Inc. Vitamin-targeted imaging agents
WO2007131286A1 (en) 2006-05-16 2007-11-22 Apollo Life Sciences Limited Nanostructures suitable for delivery of agents
US20080188430A1 (en) 2001-05-18 2008-08-07 Sirna Therapeutics, Inc. RNA interference mediated inhibition of hypoxia inducible factor 1 (HIF1) gene expression using short interfering nucleic acid (siNA)
US20080249055A1 (en) 2005-08-19 2008-10-09 Hammock Bruce D Use of Seh Inhibitors as Analgesics
US7556826B2 (en) 2005-01-04 2009-07-07 Gp Medical, Inc. Nanoparticles for protein drug delivery
US7601332B2 (en) 2003-01-27 2009-10-13 Endocyte, Inc. Vitamin receptor binding drug delivery conjugates

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009065181A1 (en) * 2007-11-21 2009-05-28 Apollo Life Sciences Limited Nanostructures suitable for delivery of agents

Patent Citations (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4166452A (en) 1976-05-03 1979-09-04 Generales Constantine D J Jr Apparatus for testing human responses to stimuli
US4256108A (en) 1977-04-07 1981-03-17 Alza Corporation Microporous-semipermeable laminated osmotic system
US4265874A (en) 1980-04-25 1981-05-05 Alza Corporation Method of delivering drug with aid of effervescent activity generated in environment of use
US4921475A (en) 1983-08-18 1990-05-01 Drug Delivery Systems Inc. Transdermal drug patch with microtubes
US5087240A (en) 1983-08-18 1992-02-11 Drug Delivery Systems Inc. Transdermal drug patch with conductive fibers
US5589463A (en) 1985-10-10 1996-12-31 Biotechnology Autralia Pty., Ltd. Oral delivery of biologically active substances bound to vitamin B12
US5428023A (en) 1985-10-10 1995-06-27 Biotechnology Australia Pty. Ltd. Oral delivery of biologically active substances bound to vitamin B12 or analogues thereof
US5163899A (en) 1987-03-20 1992-11-17 Drug Delivery Systems Inc. Transdermal drug delivery system
US5336168A (en) 1987-05-28 1994-08-09 Drug Delivery Systems Inc. Pulsating transdermal drug delivery system
US5807832A (en) 1987-06-09 1998-09-15 Biotech Australia Pty Limited Oral delivery of biologically active substances bound to vitamin B12
US5254346A (en) 1988-02-23 1993-10-19 Tucker Mark J Occlusive body for administering a physiologically active substance
US5008110A (en) 1988-11-10 1991-04-16 The Procter & Gamble Company Storage-stable transdermal patch
US5088977A (en) 1988-12-21 1992-02-18 Drug Delivery Systems Inc. Electrical transdermal drug applicator with counteractor and method of drug delivery
US5108921A (en) 1989-04-03 1992-04-28 Purdue Research Foundation Method for enhanced transmembrane transport of exogenous molecules
US5688488A (en) 1989-04-03 1997-11-18 Purdue Research Foundation Composition and method for tumor imaging
US5416016A (en) 1989-04-03 1995-05-16 Purdue Research Foundation Method for enhancing transmembrane transport of exogenous molecules
US5635382A (en) 1989-04-03 1997-06-03 Purdue Research Foundation Method for enhancing transmembrane transport of exogenous molecules
US5164189A (en) 1989-12-04 1992-11-17 G. D. Searle & Co. Single layer transdermal drug administration system
US5290561A (en) 1989-12-04 1994-03-01 G. D. Searle & Co. Single layer transdermal drug administration system
US6159502A (en) 1991-04-02 2000-12-12 Biotech Australia Pty Ltd Oral delivery systems for microparticles
US5352456A (en) 1991-10-10 1994-10-04 Cygnus Therapeutic Systems Device for administering drug transdermally which provides an initial pulse of drug
US5407713A (en) 1991-12-18 1995-04-18 Minnesota Mining And Manufacturing Company Multilayered barrier structures
US5332213A (en) 1992-01-29 1994-07-26 Franz Volkl Gmbh & Co. Ski Und Tennis Sport-Artkihelfabrik Kg. Ball-game racket, particularly a tennis racket
US5863900A (en) 1993-05-20 1999-01-26 Biotech Australia Pty., Ltd. Lhrh antagonists
US6262253B1 (en) 1993-05-24 2001-07-17 Biotech Australia Pty Limited Vitamin B12 conjugates with gcsf, analogues thereof and pharmaceutical compositions
US5548064A (en) 1993-05-24 1996-08-20 Biotech Australia Pty Limited Vitamin B12 conjugates with EPO, analogues thereof and pharmaceutical compositions
US5869466A (en) 1993-05-24 1999-02-09 Biotech Australia Pty Limited Vitamin B12 mediated oral delivery systems for GCSF
US5449720A (en) 1993-05-24 1995-09-12 Biotech Australia Pty Limited Amplification of the VB12 uptake system using polymers
US6083926A (en) 1994-04-08 2000-07-04 The University Of Washington Water soluble vitamin B12 receptor modulating agents and methods related thereto
US5605890A (en) 1994-06-01 1997-02-25 Hybridon, Inc. Cyclodextrin cellular delivery system for oligonucleotides
US5574018A (en) 1994-07-29 1996-11-12 Amgen Inc. Conjugates of vitamin B12 and proteins
US20020115595A1 (en) 1996-08-27 2002-08-22 Grissom Charles B. Bioconjugates and delivery of bioactive agents
US6221397B1 (en) 1997-08-29 2001-04-24 Biotech Australia Pty Limited Surface cross-linked particles suitable for controlled delivery
US6150341A (en) 1998-06-12 2000-11-21 Biotch Australia Pty Limited Vitamin B12 derivatives and methods for their preparation
US6482413B1 (en) 2001-02-26 2002-11-19 Council Of Scientific And Industrial Research Vitamin B12 —biodegradable micro particulate conjugate carrier systems for peroral delivery of drugs, therapeutic peptides/proteins and vaccines
US20080188430A1 (en) 2001-05-18 2008-08-07 Sirna Therapeutics, Inc. RNA interference mediated inhibition of hypoxia inducible factor 1 (HIF1) gene expression using short interfering nucleic acid (siNA)
US7128893B2 (en) 2002-05-06 2006-10-31 Endocyte, Inc. Vitamin-targeted imaging agents
US7601332B2 (en) 2003-01-27 2009-10-13 Endocyte, Inc. Vitamin receptor binding drug delivery conjugates
US7556826B2 (en) 2005-01-04 2009-07-07 Gp Medical, Inc. Nanoparticles for protein drug delivery
US20080249055A1 (en) 2005-08-19 2008-10-09 Hammock Bruce D Use of Seh Inhibitors as Analgesics
WO2007131286A1 (en) 2006-05-16 2007-11-22 Apollo Life Sciences Limited Nanostructures suitable for delivery of agents

Non-Patent Citations (24)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", MACK PUBLISHING COMPANY
A JINTAPATTANAKIT ET AL., INTERNATIONAL JOURNAL OF PHARMACEUTICS, vol. 342, 2007, pages 240 - 249
AAGAARD, L., ROSSI, J.J.: "RNAi therapeutics: Principles, prospects and challenges", ADV. DRUG DELIVERY REV., vol. 59, 2007, pages 75 - 86, XP022087315, DOI: doi:10.1016/j.addr.2007.03.005
AAGAARD, L., ROSSI, J.J.: "RNAi therapeutics: Principles, prospects and challenges", DV. DRUG DELIVERY REV., vol. 59, 2007, pages 75 - 86, XP022087315, DOI: doi:10.1016/j.addr.2007.03.005
BAGNATO ET AL., J. ORG. CHEM., vol. 69, 2004, pages 8987
BAUER, ANTI-CANCER DRUGS, vol. 9, 1998, pages 239
BRUMMELKAMP, SCIENCE, vol. 296, 2002, pages 550 - 3
DE FOUGEROLLES, A. ET AL.: "Interfering with disease: a progress report on siRNA-based therapeutics", NATURE REVIEWS DRUG DISCOVERY, vol. 6, 2007, pages 443 - 53
DYKXHOOM, D.M., LIEBERMAN, J.: "Running Interference: Prospects and Obstacles to Using Small Interfering RNAs as Small Molecule Drugs", ANNU. REV. BIOMED. ENG., vol. 8, 2006, pages 377 - 402, XP009101888, DOI: doi:10.1146/annurev.bioeng.8.061505.095848
DYKXHOORN, D.M. ET AL.: "The silent treatment: siRNAs as small molecule drugs", GENE THERAPY, vol. 13, 2006, pages 541 - 52, XP002562042, DOI: doi:10.1038/sj.gt.3302703
DYKXHOORN, D.M., LIEBERMAN, J.: "Running Interference: Prospects and Obstacles to Using Small Interfering RNAs as Small Molecule Drugs", ANNU. REV. BIOMED. ENG., vol. 8, 2006, pages 377 - 402, XP009101888, DOI: doi:10.1146/annurev.bioeng.8.061505.095848
FINGL, WOODBURY: "GOODMAN AND GILMAN'S THE PHARMACEUTICAL BASIS OF THERAPEUTICS", PERGAMON PRESS, article "General Principles", pages: 1 - 46
KHAFAGY ET AL., ADVANCED DRUG DELIVERY, vol. 59, 2007, pages 1521 - 1546
KMEGER, U. ET AL.: "Insights into effective RNAi gained from large-scale siRNA validation screening", OLIGONUCLEOTIDES, vol. 17, 2007, pages 237 - 250, XP055026573, DOI: doi:10.1089/oli.2006.0065
KRUCGCR, U.: "Insights into effective RNAi gained from largc- scale siRNA validation screening", OLIGONUCLEOTIDES, vol. 17, 2007, pages 237 - 250, XP055026573, DOI: doi:10.1089/oli.2006.0065
M. KURSA ET AL., BIOCONJUGATE CHEMISTRY, vol. 14, 2003, pages 222
MCEWAN ET AL., BIOCONJUGATE CHEM., vol. 10, 1999, pages 1131 - 1136
MIYAGISHI, TAIRA, NATURE BIOTECH., vol. 20, 2002, pages 497 - 500
R. WAIBEL ET AL., CANCER RES., vol. 68, 2008, pages 2904 - 2911
SH KIM ET AL., BIOTECHNOL PROG., vol. 23, 2007, pages 232
VIEIRA, AM CARMONA-RIBEIRO, JOURNAL OF NANOBIOTCCHNOLOGY, May 2008 (2008-05-01)
WAIBEL ET AL., CANCER RES., vol. 68, 2008, pages 2904 - 2911
WILBUR ET AL., BIOCONJUGATE CHEM., vol. 7, 1996, pages 461 - 474
Y-Z DU ET AL., INTERNATIONAL JOURNAL OF PHARMACEUTICS, 2010

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012030745A1 (en) * 2010-08-30 2012-03-08 Access Pharmaecuticals, Inc MULTIVITAMIN TARGETING OF RNAi THERAPEUTICS
WO2012122313A3 (en) * 2011-03-08 2013-03-14 Access Pharmaceuticals, Inc. Targeted nanocarrier systems for delivery of actives across biological membranes
WO2013123523A1 (en) * 2012-02-19 2013-08-22 Nvigen, Inc. Uses of porous nanostructure in delivery
US10398905B2 (en) 2012-02-19 2019-09-03 Nvigen, Inc. Uses of porous nanostructure in delivery
WO2015126841A1 (en) 2014-02-18 2015-08-27 Plasmatech Biopharmaceuticals, Inc. Nutritional and therapeutic mucoadhesive formulations
WO2017053920A1 (en) 2015-09-25 2017-03-30 Zy Therapeutics Inc. Drug formulation based on particulates comprising polysaccharide-vitamin conjugate
EP3352796A4 (en) * 2015-09-25 2019-05-01 ZY Therapeutics Inc. Drug formulation based on particulates comprising polysaccharide-vitamin conjugate
US10517961B2 (en) 2015-09-25 2019-12-31 ZY Therapeutics, Inc. Drug formulation based on particulates comprising polysaccharide-vitamin conjugate
US10994024B2 (en) 2015-09-25 2021-05-04 ZY Therapeutics, Inc. Drug formulation based on particulates comprising polysaccharide-vitamin conjugate
US11160878B1 (en) * 2020-06-10 2021-11-02 Boris Farber Supramolecular systems based on dynamic self-organizing nanostructures with antiviral properties
EP3922347A1 (en) * 2020-06-10 2021-12-15 Boris Slavinovich Farber Supramolecular systems based on dynamic self-organizing nanostructures with antiviral properties

Also Published As

Publication number Publication date
WO2011130716A3 (en) 2012-06-28

Similar Documents

Publication Publication Date Title
US20160175259A1 (en) Targeted Nanocarrier Systems for Delivery of Actives Across Biological Membranes
US10087442B2 (en) Polycation-functionalized nanoporous silicon carrier for systemic delivery of gene silencing agents
EP3315608A1 (en) Sirna, pharmaceutical composition and conjugate which contain sirna, and uses thereof
Luan et al. Anisamide-targeted PEGylated gold nanoparticles designed to target prostate cancer mediate: Enhanced systemic exposure of siRNA, tumour growth suppression and a synergistic therapeutic response in combination with paclitaxel in mice
WO2011130716A2 (en) A nanostructures containing vitamin b12 for facilitated delivery of drugs across biological barriers
EP2289946A1 (en) A polyglycol modified chitosan oligosaccharide fatty acid graft, preparation method thereof and use of the same
US7985426B1 (en) Nanoparticles for targeting hepatoma cells and delivery means
US11110168B2 (en) Nanoparticles, controlled-release dosage forms, and methods for delivering an immunotherapeutic agent
US9061068B2 (en) Polymeric nano-particles for siRNA delivery using charge interaction and covalent bonding
WO2012030745A1 (en) MULTIVITAMIN TARGETING OF RNAi THERAPEUTICS
US20130108686A1 (en) Method for the delivery of oligonucleotides
EP3214178B1 (en) Polyol-based osmotic polydixylitol polymer based gene transporter and use thereof
EP3238727A1 (en) Non-aggregating immunostimulatory oligonucleotides
Wei et al. Role of long non-coding RNAs (lncRNAs) in cancer: from subcellular localization to nanoparticles (NPs)-mediated targeted regulation
US20200369710A1 (en) Rna nanostructures, methods of making, and uses thereof
JP5843086B2 (en) Use of polymerized cyclic nitroxide radical compounds to enhance the action of therapeutically active substances
Fadaka et al. Stage-specific treatment of colorectal cancer: A microRNA-nanocomposite approach
JPWO2018216792A1 (en) Agent for improving stability of RNA in blood and method of administration
US20220372493A1 (en) Rna nanoparticle for liver cancer treatment
Huang et al. DNA-based Nanomaterials in the Immunotherapy
KR20230104196A (en) Non-cationic Soft Polyphenol Nanocapsules for Effective Systemic Delivery of Small Interfering RNA (SiRNA) for Cancer Treatment
US20130259944A1 (en) Methods and compositions for treating cancer with platinum particles
WO2023023662A1 (en) Targeting oncogenic kras with molecular brush-conjugated antisense oligonucleotide
WO2024081511A2 (en) Rna complexes and nanostructures for treatment of cancer metastasis
JP2020193160A (en) Drug carrier for pulmonary delivery and pulmonary disease therapeutic drug containing the same

Legal Events

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

Ref document number: 11736475

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11736475

Country of ref document: EP

Kind code of ref document: A2