[go: up one dir, main page]

HK1116406A - Compositions capable of facilitating penetration across a biological barrier - Google Patents

Compositions capable of facilitating penetration across a biological barrier Download PDF

Info

Publication number
HK1116406A
HK1116406A HK08106131.9A HK08106131A HK1116406A HK 1116406 A HK1116406 A HK 1116406A HK 08106131 A HK08106131 A HK 08106131A HK 1116406 A HK1116406 A HK 1116406A
Authority
HK
Hong Kong
Prior art keywords
composition
group
acid
effector
inhibitor
Prior art date
Application number
HK08106131.9A
Other languages
Chinese (zh)
Inventor
Shmuel A. Ben-Sasson
Original Assignee
克艾思马有限公司
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 克艾思马有限公司 filed Critical 克艾思马有限公司
Publication of HK1116406A publication Critical patent/HK1116406A/en

Links

Description

Composition capable of easily penetrating biological barrier
Technical Field
The present invention relates to novel penetrating compositions that allow effective passage of effectors across biological barriers.
Background
In the field of biotechnology, a technology capable of allowing a target substance to effectively cross a biological barrier is a technology of great interest. For example, these techniques can be used to deliver a variety of different substances across biological barriers formed by tightly-connected tissues (e.g., mucosal epithelium, including intestinal epithelium and respiratory epithelium, and vascular epithelium, including the blood-brain barrier).
The intestinal epithelium is a major obstacle to the absorption of oral compounds into the systemic circulation, and for example, is a major biological obstacle for oral drugs and peptides. This biological barrier is composed of a single layer of columnar epithelial cells (mainly intestinal epithelial cells, goblet cells, endocrine cells and Pan's cells) that are attached to their apical surface by tight junctions. See Madara et al, "gastrointestinal physiology", New York Raven Press, second edition, pages 1251-66 (1987).
Compounds present in the intestine can enter the bloodstream by active or facilitated transport, passive transcellular transport or passive intercellular transport. Active transport or facilitated transport is achieved by cellular delivery of a substance. Active or facilitated transport is limited to the delivery of small molecular weight products such as amino acids, pentoses and hexoses, which are produced by the degradation of complex molecules such as proteins and carbohydrates. Passive transcellular transport is the partitioning of molecules across the apical and basolateral membranes. This process is limited to relatively small hydrophobic compounds. See Jackson, gastrointestinal physiology, New York Raven Press, second edition, p 1597-1621 (1987). Thus, in addition to these molecules that can be transported by active or facilitated transport, the more absorbed, more hydrophilic molecules are largely achieved only by means of intermolecular channels. However, the entry of molecules into tissues through intercellular channels is primarily restricted by tight junctions. See Gumbiner's article in journal of american physiology, 253: C749-C758(1987), see Madara's article in journal of clinical research 83: 1089-94(1989).
Researchers have spent a great deal of time and effort trying to improve cell-to-cell transport by "relaxing" tight junctions. One way to overcome the intercellular transport disorder is to use an absorption enhancer in addition to the bioactive component. In general, intestinal/respiratory absorption enhancers include calcium chelators such as citrate and ethylenediaminetetraacetic acid, surface-active agents such as sodium lauryl sulfate, bile salts, palmitoyl carnitine, and sodium salts of fatty acids; however, the intestinal/respiratory tract absorption enhancer is not limited to these substances. For example, ethylenediaminetetraacetic acid breaks tight junctions by forming a chelating structure with calcium, thereby increasing the efficiency of genes into the airway epithelial tissue of cystic fibrosis patients. See Wang et al, journal of american journal of respiratory cell molecular biology, 22: 129-138(2000). However, one drawback of all of these methods is that they indiscriminately allow penetration of molecules in the nearby gastrointestinal or tracheal tract. In addition, the properties of these intestinal/respiratory absorption enhancers limit their general utility as molecular absorption enhancers for crossing biological disorders.
Furthermore, when caustic surfactants are used, the potential solubility properties of these surfactants can cause safety concerns. In particular, the intestinal epithelial cells and the cells of the respiratory epithelium constitute a barrier, thereby blocking the invasion of toxins, bacteria and viruses from the outside. Therefore, when these surfactants are used as intestinal/respiratory absorption enhancers, there are safety problems caused by the possible exfoliation of epithelial tissues and the potential complications of repairing epithelial tissues.
When the calcium chelating agent is used as an intestinal/respiratory absorption enhancer, the loss of calcium ions does not directly act on tight connection, but causes the cells to change integrally, and the loss of calcium ions can cause the breakage of fibrillin filaments and the breakage of adhesive zonules and reduce the cell adhesion; and activates protein kinases. See the article by Citi in journal of cell biology 117: 169-178(1992). Furthermore, since the calcium chelating agent can only contact mucosal surfaces, the intraluminal calcium ion concentration may vary, and therefore, a sufficient amount of chelating agent to reduce the calcium ion concentration is not usually administered to open the tight junctions in a rapid, reversible or repeatable manner.
Furthermore, certain toxins like Clostridium difficile (Clostridium difficile) toxins a and B appear to irreversibly increase intercellular permeability and are therefore involved in the disassembly of tight junctions. See Hecht et al article in journal of clinical research 82: 1516-24(1988), see Fiorentini and Thelestam, Toxicon, 29: 543-67(1991). Other toxins such as Vibrio cholerae (Vibrio cholerae) zonula occludens toxin can alter the structure of the intermolecular tight junctions. Thus, the intestinal mucosa becomes more permeable, but the penetration is non-selective. See Fasano et al article on the american academy of science, 8: 5242-46(1991), see U.S. Pat. No. 5,857,534. This practice may also lead to diarrhea.
Researchers have conducted special research on orally administered bioactive peptides and proteins because bioactive peptides and proteins are very fragile in the gastrointestinal environment and undergo enzymatic and chemical denaturation. Researchers have used different types of delivery vehicles including liposomes, lipid or polymeric nanoparticles, and microemulsions. These delivery vehicles, in large part, have a protective effect, and thus improve the activity of certain drugs in the case of oral administration. However, these vectors do not address the impermeability of epithelial barriers. Therefore, for most of the related drugs, the absorption rate is not more than 5%, and thus the most basic therapeutic effect cannot be achieved.
Thus, there is a need for an effective, specific, non-invasive, low risk method that overcomes various biological barriers and can be used in the administration of large molecule bioactive drugs, such as polypeptides, polymeric drugs, and other therapeutic agents.
Summary of The Invention
The present invention provides compositions for transporting therapeutically active molecules, which may be, for example, effectors, by including the active molecules in a water-soluble composition and allowing the active molecules to traverse a biological barrier that otherwise would not be able to traverse the biological barrier. In one embodiment, the water-soluble composition is immersed in a hydrophobic medium. Alternatively, the aqueous solution may be first freeze-dried and then resuspended in a hydrophobic medium. The present invention also relates to the use of a membrane fluidizer, wherein the use of a membrane fluidizer is to facilitate the transport process of said at least one effector across a biological barrier.
As used herein, "effective transport" refers to introducing a composition into a biological barrier and allowing at least 5%, desirably 10%, and more desirably 20% of the effector to traverse the biological barrier.
As used herein, a "penetrating composition" includes a water-soluble composition immersed in a hydrophobic medium that facilitates a substance through the use of at least one film fluidizing agent; such as the passage of at least one effector across a biological barrier to form an effective transport. The term "water-soluble composition" as used herein refers to a composition that is soluble in hydrophilic or partially hydrophilic solvents. The hydrophilic or partially hydrophilic solvent may consist of water or a non-aqueous medium such as mono-, di-or triols. Specific suitable monoalcohols include ethanol, propanol, isopropanol, and butanol, but suitable monoalcohols are not limited to these alcohols. Specific glycols include, but are not limited to, propylene glycol. Specific triols include, but are not limited to, glycerol.
According to the methods and compositions of the present invention, the water-soluble composition is immersed in a hydrophobic medium. Alternatively, the aqueous solution may be first freeze-dehydrated and then resuspended in a hydrophobic medium, which may be composed of aliphatic, cyclic or aromatic molecules. Particularly suitable aliphatic hydrophobic media include mineral oils (such as paraffin), fatty acids, monoglycerides, diglycerides, triglycerides, ethers and esters. Particularly suitable triglycerides include long chain triglycerides, medium chain triglycerides and short chain triglycerides. For example, the long chain triglyceride may be castor oil; the short chain triglyceride may be tributyrin. Particularly suitable hydrophobic media of cyclic structure include terpenoids, cholesterol derivatives (such as cholesterol sulfate) and cholesterol esters of fatty acids; however, the hydrophobic medium having a cyclic structure is not limited to these substances. Specific aromatic hydrophobic media include benzyl benzoate, but aromatic hydrophobic media are not limited to benzyl benzoate.
The penetrating composition also includes a film fluidizing agent. The term "membrane fluidizer" as used herein refers to a molecule that can increase the mobility of a biological membrane and can reduce the lipid order in the biological membrane. For example, the membrane fluidizer can be a linear alcohol species, a branched alcohol species, an alcohol species with a cyclic structure, or an aromatic alcohol species. Particularly suitable linear alcohols include butanol, pentanol, hexanol, heptanol, octanol, nonanol, decanol, undecanol, and dodecanol; however, the linear alcohols are not limited to these alcohols. Particularly suitable branched alcohols include geraniol and farnesol; however, the branched alcohol is not limited to these two alcohols. Particularly suitable aromatic alcohols include, but are not limited to, benzyl alcohol, 4-hydroxycinnamic acid and phenolic materials. The phenolic substances comprise phenol, m-cresol and m-chlorocresol; however, the phenolic compounds are not limited to these compounds.
As used herein, the term "biological disorder" refers to biological membranes such as cell membranes, biological structures enclosed by tight connective tissues (or occluding connective tissues) such as mucosal epithelial cells or vascular epithelial cells (including but not limited to gastrointestinal epithelial cells or respiratory epithelial cells), and blood brain disorders. In addition, one skilled in the art will recognize that transport across biological barriers may occur in tissues containing, for example, epithelial and endothelial cells.
The invention also provides a penetrating composition comprising a carrier, an excipient, or a mixture of both, wherein the carrier and excipient are pharmacologically acceptable. In various embodiments, the compositions of the present invention may be packaged in capsules or may be presented in the form of tablets, emulsions, creams, oils, suppositories or nasal sprays.
The penetrating composition includes at least one effector. The effector is a therapeutically active non-penetrating molecule; effectors include nucleic acids, glycosaminoglycans, proteins, peptides or pharmacologically active agents such as hormones, growth factors, incretins, neurotrophic factors, anticoagulants, biologically active molecules, toxins, antibiotics, antifungals, antipathogens, antigens, antibodies, monoclonal antibodies, antibody fragments, soluble receptors, immunomodulators, vitamins, antineoplastic agents, cytokines or other therapeutic agents, but are not limited to these. By way of example, glycosaminoglycans which may be used as non-penetrating compounds include heparin, heparin derivatives, heparan sulfate, chondroitin sulfate, dermatan sulfate and hyaluronic acid; however, the aminodextran which can be used as the non-penetrating composition is not limited to these substances. Specific heparin derivatives include low molecular weight heparins such as enoxaparin, dalteparin, tinzaparin and fondaparin; however, the heparin derivative is not limited to these substances. Nucleic acids as non-penetrating molecules include specific deoxyribonucleic acid sequences (e.g., coding genes), specific ribonucleic acid sequences (e.g., ribonucleic acid aptamers, antisense ribonucleic acids, or specific inhibitory ribonucleic acids (RNAi)), poly-CpG, poly-I of nucleic acids: c, synthesizing a polymer; however, the nucleic acid as the non-penetrating molecule is not limited to these substances. Other suitable proteins include insulin, erythropoietin, glucagon-like peptide 1(GLP-1), melanocyte stimulating hormone (alpha MSH), parathyroid hormone (PTH), thyroid hormone amino acids 1-34(PTH (1-34)), growth hormone, peptide YY amino acids 3-36(PYY (3-36)), calcitonin, leukokine-2 (IL-2), alpha 1-antitrypsin, granulocyte/monocyte colony stimulating factor (GM-CSF), granulocyte colony stimulating factor (G-CSF), T20, anti-tumor necrosis factor antibodies, interferon alpha, interferon beta, interferon gamma, Luteinizing Hormone (LH), Follicle Stimulating Hormone (FSH), brain fibula, dalargin, kyoworphin, basic fibroblast growth factor (bFGF), hirudin beta, hirudin, Luteinizing Hormone Releasing Hormone (LHRH) analogs, Brain Natriuretic Peptide (BNP), glatiramer acetate, and neurotrophic factors; however, other suitable proteins are not limited to these substances.
Suitable effectors also include pharmacologically active agents selected from vitamin B12, hydrogen phosphate, taxol, caspofungin or aminoglycoside antibiotics.
As used herein, "non-penetrating molecules" refers to molecules that do not effectively cross biological barriers, such as cell membranes or tightly-coupled tissues. Generally speaking, the non-penetrating molecule of the present invention is a molecule having a molecular weight greater than 200 daltons. Preferably, the anionic non-penetrating molecule is a polysaccharide such as an aminodextran, a nucleic acid or a net negatively charged protein; in a preferred aspect, the cationic non-penetrating molecule is a protein having a net positive charge.
The net charge of a protein is determined by two factors: 1) total amount of acidic amino acids/total amount of reducing amino acids; 2) the PH environment of a particular solvent will form either positively or negatively charged residues. As used herein, "a net positively or negatively charged protein" refers to a protein that is net positively or negatively charged in an unmodified pH environment. For example, interferon beta contains 23 positively charged residues (lysine and arginine) and 18 negatively charged residues (glutamic acid and aspartic acid). Thus, interferon beta is a net positively charged protein in neutral or acidic PH environments. In contrast, insulin is a 51 amino acid protein that contains two positively charged residues; one is lysine and one is arginine; it also contains four negatively charged glutamate residues. Thus, insulin is a net negatively charged protein in a neutral or alkaline PH environment. In general, one skilled in the art will recognize that regardless of the amino acid composition of a protein, all proteins can be considered as "net negatively charged proteins" or "net positively charged proteins," and specifically whether a "negatively charged protein" or a "positively charged protein" depends on the PH environment and/or solvent environment in which the protein is exposed. For example, solvents of different pH values may form negatively charged side chains or positively charged side chains.
The water-soluble composition of the present invention may further contain a protein structure stabilizer. As used herein, "protein structure stabilizer" refers to a compound capable of stabilizing the structure of a protein under hydrated or non-hydrated conditions; such as polycationic molecules, polyanionic molecules and uncharged polymers. One particular polycationic molecule that may act as a protein stabilizer is a polyamine-based molecule, such as spermine. Specific polyanionic molecules that can act as protein stabilizers include phytic acid and sucrose octasulfate, but polyanionic molecules that can act as protein stabilizers are not limited to these two substances. Non-limiting uncharged polymers that can act as protein stabilizers include polyvinylpyrrolidone and polyvinyl alcohol.
The water-soluble compositions of the present invention may also contain a zwitterionic counterion. For example, the counterion comprises an anionic amphoteric molecule or a cationic amphoteric molecule. In one embodiment, the anionic counterions or cationic counterions of the invention are negatively charged (anionic) or positively charged (cationic) and contain an amphoteric moiety. Under appropriate conditions, anionic counterions or cationic counterions can electrostatically interact with cationic non-penetrating molecules or anionic non-penetrating molecules, respectively. This complexation formation neutralizes the charge, forming new uncharged moieties, which in turn give rise to hydrophobic properties due to the inherent hydrophobicity of the counterion.
Cationic counterions contemplated by the present invention include quaternary amine derivatives, such as benzalkonium derivatives. Suitable quaternary amines may be partially substituted with hydrophobic residues. In general, the quaternary amines contemplated by the present invention have the structure 1-R1-2-R2-3-R3-4-R4-N, wherein R1, R2, R3, and R4 are alkyl or aryl derivatives. In addition, the quaternary amine may be an ionic liquid foaming cation, such as an imidazolium derivative, a pyridinium derivative, a phosphonium compound, or a tetraalkylammonium compound. For example, the imidazolium derivatives have the general structure 1-R1-3-R2-imidazolium, wherein R1 and R2 can be straight-chain or branched alkyl groups with 1 to 12 carbon atoms. A portion of these imidazolium derivatives may be substituted with a halogen or an alkyl group. Specific imidazolium derivatives include 1-ethyl-3-methylimidazolium, 1-butyl-3-methylimidazolium, 1-hexyl-3-methylimidazolium, 1-methyl-3-octylimidazolium, 1-methyl-3- (3, 3, 4, 4, 5, 5, 6, 6, 7, 7, 8, 8, 8-tridecafluorooctyl) -imidazolium, 1, 3-dimethylimidazolium, and 1, 2-dimethyl-3-propylimidazolium; however, specific imidazolium derivatives are not limited to these.
The general structure of the pyridinium derivative is 1-R1-3-R2-pyridinium, wherein R1 is a straight or branched alkyl group with 1-12 carbon atoms; r2 is hydrogen or a straight or branched alkyl group having 1 to 12 carbon atoms. Some of these pyridinium derivatives may be substituted with halogen or alkyl groups. Pyridinium derivatives include 3-methyl-1-propylpyridinium, 1-butyl-3-methylpyridinium, and 1-butyl-4-methylpyridinium; however, the pyridinium derivative is not limited to these substances. The ionic liquid foaming cations described herein may also be part of a water soluble salt.
Suitable anionic counterions are ions having negatively charged residues, such as carboxylate, sulfonate or phosphate ions, and may carry hydrophobic moieties. Specific anionic counterions include sodium lauryl sulfate, cetyl sulfosuccinate and other anionic compounds derived from organic acids; however, the anionic counter ion is not limited to these.
The penetrating compositions of the present invention may also contain a surfactant. Suitable surfactants include ionic detergents and non-ionic detergents. The ionic detergent may be a fatty acid salt, lecithin or bile salt. Specific fatty acid salts include sodium caprylate, sodium caprate and sodium dodecanoate; however, the specific fatty acid salts are not limited to these materials. Non-limiting non-ionic detergents include cremophor, polyethylene glycol fatty alcohol ethers, sorbitan fatty acid esters, polyethylene glycol stearate 15(Solutol HS15), polyols. Specific sorbitan fatty acid esters include sorbitan monolaurate, sorbitan monooleate, and sorbitan monolaurate; however, the specific sorbitan fatty acid ester is not limited to these substances.
The penetrating compositions of the present invention may also include an adhesive polymer, such as methyl cellulose, ethyl cellulose, hydroxypropylmethyl cellulose (HPMC), or carbopol. In addition, the penetrating compositions of the present invention may also contain monoglycerides. Specific monoglycerides include glyceryl monocaprylate, glyceryl monodecanoate, glyceryl monolaurate, glyceryl monomyristate, glyceryl monostearate, glyceryl monolaurate, and glyceryl monooleate; however, the specific monoglyceride is not limited to these substances.
In one embodiment, the penetrating compositions of the present invention comprise at least one effector; and contains arginine, polyvinylpyrrolidone and sodium laurate, which is immersed in a vegetable oil such as castor oil together with octanol and geraniol; or submerged in a medium chain triglyceride or a mixture of tributyrin and castor oil. The composition may also contain sorbitan monolaurate and/or glycerol monooleate and/or methylcellulose and/or cholesterol sulfate.
The penetrating compositions of the present invention may also contain a protectant. One particular protectant is a protease inhibitor. An article by Bernkop-Schnurch et al, journal of controlled release, describes suitable protease inhibitors that may be incorporated into the penetrating composition, 52: 1-16(1998). For example, these protease inhibitors include inhibitors of luminal endocrine proteases; such as aprotinin, Bowman-birk inhibitor, soybean trypsin inhibitor, avian ovomucoid inhibitor, avian egg inhibitor, human trypsin inhibitor, camostat mesylate, flavonoid inhibitor, analgesic, leupeptin, p-aminobenzyl ether, serine endopeptidase inhibitor (AEBSF), tosyl-L-lysine-chloromethyl ketone hydrochloride (TLCK), APMSF, diisopropyl fluorophosphoric acid (DFP), phenylmethylsulfonyl fluoride (PMSF), poly (acrylate) derivatives, chymostatin, benzyloxycarbonyl-proline-phenylalanine-CHO, FK-448, sucrose biphenylboronic acid complex, beta-phenylpropionate, elastase inhibitor, methoxyacyl-alanine-proline-valine-chloromethyl ketone (MPCMK), Ethylenediaminetetraacetic acid and chitosan-ethylenediaminetetraacetic acid conjugates. Suitable protease inhibitors also include membrane-bound protease inhibitors such as amino acids, dipeptides, tripeptides, amastatin, hydroxyaminobutyrylleucine, puromycin, bacitracin, phosphonate dipeptide analogs, alpha-aminoboronic acid derivatives, sodium glycocholate, 1, 10-phenanthroline, acivicin, levoserine borates, membrane metallo-endoproteinase, and amiloride phosphate.
Preferred compositions include enteric coated tablets and gelatin or Hydroxypropylmethylcellulose (HPMC) capsule dosage forms, wherein the capsule dosage forms contain an active ingredient and a) a diluent; such as lactose, glucose, sucrose, mannitol, sorbitol, cellulose and/or glycine; b) a protease inhibitor; such as trypsin inhibitors or aprotinin; c) lubricants, such as silica, talc, stearic acid and its magnesium or calcium salts, polyols and/or polyethylene glycols; binders also used for tablets d) such as magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone; e) ionic surfactants such as polyhydroxy, polyethylene glycol stearate 15(Solutol HS15), cremophor, phospholipids and cholic acid; if desired, f) disintegrating agents, such as starch, agar, alginic acid or sodium alginate, effervescent mixtures; and/or g) absorbents, colorants, flavors, and sweeteners. Suppositories are preferably made of fatty emulsions or suspensions. The composition can be sterilized; and/or contain adjuvants, such as preservatives, reducing agents; such as n-acetyl-1-cysteine, stabilizers, wetting or emulsifying agents, solubility promoters, salts for regulating the osmotic pressure and/or buffers. In addition, the composition may contain other substances having therapeutic effects. The composition can be prepared by conventional mixing, granulating or coating methods; the composition contains about 0.001-75% active ingredient, and preferably the composition contains about 0.01-10% active ingredient.
The composition may also comprise a mixture of at least two substances; the two materials are selected from the group consisting of nonionic detergents, ionic detergents, sticky polymers, monoglycerides, protease inhibitors, mercapto state modifiers, and antioxidants. For example, the non-ionic detergent may be a polyhydroxy body, cremophor, a polyethylene glycol fatty alcohol ether, a sorbitan fatty acid ester, or a polyethylene glycol stearate 15(solutol hs 15); the ionic detergent may be a fatty acid salt, and the adhesive polymer may be methylcellulose, ethylcellulose, Hydroxypropylmethylcellulose (HPMC), or carbopol; the monoglyceride may be glyceryl monocaprylate, glyceryl monocaprate, glyceryl monolaurate, glyceryl monomyristate, glyceryl monostearate, glyceryl monolaurate, and glyceryl monooleate; the protease inhibitor is selected from aprotinin, Bowman-birk inhibitor, soybean trypsin inhibitor, avian ovomucoid inhibitor, avian egg inhibitor, human trypsin inhibitor, camostat mesylate, flavonoid inhibitor, analgesic, leupeptin, p-aminobenzyl ether, serine endopeptidase inhibitor (AEBSF), tosyl-levo lysine-chloromethyl ketone hydrochloride (TLCK), APMSF, diisopropyl fluorophosphoric acid (DFP), phenylmethylsulfonyl fluoride (PMSF), poly (acrylate) derivatives, chymostatin, benzyloxycarbonyl-proline-phenylalanine-CHO, FK-448, sucrose biphenylboronic acid complex, beta-phenylpropionate, elastase inhibitor, methoxysuccinyl-alanine-proline-valine-chloromethyl ketone (MPK) & CMK), Ethylenediaminetetraacetic acid and chitosan-ethylenediaminetetraacetic acid conjugates, amino acids, dipeptides, tripeptides, amastatin, hydroxyaminophenylbutyryl leucine, puromycin, bacitracin, phosphonate dipeptide analogs, alpha-aminoboronic acid derivatives, sodium glycocholate, 1, 10-phenanthroline, acivicin, levoserine borate, membrane metallo-endoproteinase inhibin, and amiloride phosphate. The sulfhydryl group state modifier can be n-acetyl-1-cysteine or diamide; the antioxidant is selected from vitamin E, deferoxamine mesylate, methylparaben, ethylparaben and vitamin C.
The invention also provides a kit with one or more containers containing a therapeutically effective amount or a prophylactically effective amount of a composition of the invention.
Also included within the scope of the present invention are methods of making and using the pharmaceutical compositions of the present invention.
The present invention also relates to methods of using the compositions of the present invention to effectively transport at least one effector across a biological barrier. For example, the water-soluble composition may contain at least one effector, and optionally, after lyophilization, the water-soluble composition is immersed in a hydrophobic medium to form the composition of the present invention. The composition may then be introduced into a biological barrier, thereby effectively causing the effector to traverse the biological barrier, thereby transporting the effector.
Also described herein are methods of treating or preventing a disease or pathological condition using an effective amount of a composition of the invention; the compositions of the present invention are administered to an individual in need of therapeutic or prophylactic measures. For example, diseases or pathological conditions to be treated include endocrine diseases, ophthalmologic diseases, neuropathy, cardiovascular diseases, metabolic diseases, kidney diseases, hematological diseases, diseases of the immune system and rheumatism, infectious diseases, tumor disorders and multi-factorial diseases; wherein the endocrine disorder is selected from diabetes, infertility, hormone deficiency, and osteoporosis; neuropathy includes alzheimer's disease and other forms of dementia, parkinson's disease, multiple sclerosis, and chorea; cardiovascular diseases including atherosclerosis, hypercoagulability, hypocoagulability, coronary heart disease, and cerebrovascular disease; metabolic disorders including obesity and vitamin deficiency; kidney diseases including renal failure; blood disorders include solid anemia; immune system disorders and rheumatic disorders including autoimmune diseases and immunodeficiency; infectious diseases include viral, bacterial, fungal and parasitic infections; multifactorial diseases including impotence, chronic pain, depression, different fibrotic conditions and short stature; however, the conditions to be treated are not limited to these conditions.
Administration of the active compounds and salts described herein can be by any acceptable method of administration. These methods include oral, buccal, anal, rectal, bronchial, pulmonary, nasal, sublingual, intraocular, parenteral, dermal or topical administration.
The invention also includes methods of making the compositions described herein. For example, a water-soluble composition containing an effector substance may be dissolved or suspended in a hydrophilic or partially hydrophilic solvent and then immersed in a hydrophobic medium along with a membrane fluidizer, thereby producing a composition of the present invention. Alternatively, the water-soluble composition containing the effector or any mixture of effector, protein inhibitor and/or counterion may be lyophilized together and then suspended in a hydrophobic medium with the membrane fluidizing agent. In general, the entire water-soluble composition can be first freeze-dehydrated and then resuspended in a hydrophobic medium. Alternatively, other components of the composition may be lyophilized or added during reconstitution of the lyophilized material.
The invention also includes methods of mucosal immunization; i.e. oral, nasal, rectal, vaginal or bronchial administration; these administration procedures include administering to a subject in need thereof an effective amount of a composition of the invention, wherein the effector comprises an antigen required for immunization. In a certain embodiment, the effector is a protective antigen for use in a vaccine against anthrax. In another embodiment, the effector is hepatitis b surface antigen (HBs) for use in a vaccine against hepatitis b.
The details of one or more embodiments of the invention are set forth in the description below. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods and materials are described herein. Other features, objects, and advantages of the invention will be apparent from the description and from the claims. In the specification and the claims appended hereto, the singular forms "a", "an", and "the" include plural referents unless the context clearly dictates otherwise. Unless defined otherwise, 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 patents and publications cited herein are hereby incorporated by reference.
Brief description of the drawings
FIG. 1 shows a gradual and significant decrease in blood glucose concentration in experimental mice following intestinal transit of insulin using a penetrating composition of the invention. The compositions were administered by intramuscular injection or rectally, and then blood glucose concentrations were measured at different times.
Fig. 2 shows that, after the use of the penetrating composition of the present invention, the composition allows interferon α to cross the intestine of mice, and thus interferon α is detected at a high concentration in the blood of mice. In contrast, interferon alpha is present in lower concentrations in the aqueous solution of the comparative phosphate-buffered saline. The agents were administered rectally and serum samples were collected at various times.
FIG. 3 shows that, after the use of the penetrating composition of the present invention, the composition allows interferon alpha to cross the intestine of mice, and thus interferon alpha is detected at a high concentration in the blood of mice. In contrast, interferon alpha is present in lower concentrations in the aqueous solution of the comparative phosphate-buffered saline. The agents were administered nasally and serum samples were collected at different times.
Figure 4 shows that the composition of the invention, after use of the penetrating composition of the invention, caused glucagon-like peptide 1 to cross the intestine, thereby reducing the response of the mice to oral glucose. Glucose was orally administered into mice, and the composition of the present invention was administered intraperitoneally or rectally, and a control formulation without glucagon-like peptide 1 was used for control mice, and then blood glucose concentration was measured at different times.
Detailed description of the invention
The present invention provides traversing compositions that target specifically various tissues, particularly tissues containing epithelial and endothelial cells, to traverse biological disorders with drugs and other therapeutic agents. The existing transport systems in the field are very limited and can only be used for general purposes. These systems alter the biological properties of the active substance due to the ineffectiveness of the existing transport systems, cause damage to the target cells, irreversibly destroy biological barriers and/or pose a great risk to the human body. In one embodiment of the invention, the composition comprises a non-penetrating effector and a film fluidizer in a water soluble composition. Alternatively, the complexed structure may be freeze-dehydrated and then immersed in a hydrophobic medium. Immersion of a water-soluble composition containing at least one effector or a lyophilized product thereof in a hydrophobic medium results in a direct, specific relationship between the effector and the penetration enhancing substance, thereby allowing what was once a non-penetrating effector to be effectively transported across a biological barrier. These compositions of the present invention may be defined by their effectiveness, since these compositions must allow at least 5% (more desirably 10% or even 20%) of at least one effector to cross epithelial tissue barriers. This efficiency is greater than that of other compositions known in the art, which are typically only capable of transporting 1-3% of the effector.
The compositions of the present invention will selectively allow transport of effectors across biological barriers. The hydrophobic medium is a protective layer which can prevent nearby molecules; such as proteins, toxins or other "unrelated" molecules, along with at least one effector, traverse biological barriers, resulting in the co-transport phenomenon.
In recent years, many new drugs have been developed, and these new drugs have been approved, some of which are peptide drugs and protein drugs. Many other drugs are in the advanced stages of clinical trials. However, the development of satisfactory delivery systems for these rapidly emerging drugs has not kept pace. These new drugs have very low gastrointestinal absorption rates and short half-lives in the body, which usually result in administration of these new drugs by infusion or frequent injection.
The use of nanoparticle and microparticle technologies successfully enhanced the biological activity of less absorbable drugs in oral form or successfully induced mucosal immune responses. See Delie's article on advanced drug administration review; 34: 221-233(1998). Nanoparticles are colloidal polymer carriers for drugs, which ensure the effect of oral administration. These polymeric dosage forms have the advantage of allowing continuous delivery to the tissue, preventing the destructive action of degrading enzymes, and enhancing site-specific drug delivery. Macromolecules such as hormones may be occluded inside the polymer particles. See Jiao et al, circulation, 105: 230- & ltCHEM & gt 235(2002), which evaluated heparin-loaded nano-scale polymer particles.
In the development of new oral dosage forms, researchers have focused on the development of lipid-based systems. Researchers have focused on the development of microemulsions, which are used as drug dissolution and absorption enhancement systems. See article by Constantinides et al, pharm. res., 11 (10): 1385-1390(1994).
Microemulsions commonly used are thermodynamically stable dispersions of one liquid phase in another, and microemulsions are mixtures involving at least three components-oil, water and surfactant mixtures. Microemulsions of the water-in-oil and oil-in-water type are used to increase the bioavailability of orally administered drugs. These two microemulsions improve the solubilization of the drug and protect the drug from enzymatic attack, while possibly achieving improved absorption due to the altered membrane permeability of the surfactant. For example, an article by Watnasichakiku et al in the journal of pharmaceuticals describes the oral administration and biological activity of insulin in a water-in-oil microemulsion; 54: 473-480(2002).
As previously mentioned, the penetrating compositions of the present invention comprise at least one effector in a water-soluble composition; the water-soluble composition is immersed in a hydrophobic medium and the penetrating composition of the present invention is effective to transport at least one effector across a biological barrier. In emulsions, water is the basic constituent, and unlike emulsions, the water-soluble compositions of the present invention are soluble in both water and non-aqueous media, such as mono-, di-or tri-alcoholic substances. In addition, the water-soluble composition of the present invention may be subjected to a freeze-dehydration process to evaporate all of the water before suspending in the hydrophobic medium.
In addition, unlike microemulsions of the water-in-oil and oil-in-water type, in which surfactants are necessary, the penetrating compositions of the present invention are an oral delivery system, the addition of surfactants is an option.
Suitable hydrophobic media include, for example, aliphatic molecules, cyclic molecules, or aromatic molecules. Particularly suitable aliphatic hydrophobic media include mineral oils (such as paraffin), fatty acids, monoglycerides, diglycerides, triglycerides, ethers and esters; however, the aliphatic hydrophobic medium to be used in particular is not limited to the above-mentioned substances.
Specific triglycerides include long chain triglycerides, medium chain triglycerides and short chain triglycerides; however, the triglyceride is not limited to these. For example, the long chain triglyceride may be castor oil; the short chain triglyceride may be tributyrin. Particularly suitable hydrophobic media of cyclic structure include terpenoids, cholesterol derivatives (such as cholesterol sulfate) and cholesterol esters of fatty acids; however, the hydrophobic medium having a cyclic structure is not limited to these substances. Non-limiting aromatic hydrophobic media include benzyl benzoate.
One example of a penetrating composition of the invention comprises insulin; insulin is dissolved in water, freeze-dehydrated and then immersed in castor oil or a mixture of castor oil and medium chain triglycerides or glycerol tributyrates. For example, membrane fluidizers such as octanol and geraniol may be included in the hydrophobic medium to facilitate transport of effectors.
In another embodiment, the compositions of the present invention employ a film fluidizing agent. For example, the membrane fluidizer can be a linear alcohol, a branched alcohol, a cyclic alcohol, or an aromatic alcohol. Particularly suitable linear alcohols include butanol, pentanol, hexanol, heptanol, octanol, nonanol, decanol, undecanol, and dodecanol; however, the linear alcohols suitable for use are not limited to these alcohols. Non-limiting branched alcohols include geraniol and farnesol. Specific cyclic alcohols include menthol. Particularly suitable aromatic alcohols include benzyl alcohol, 4-hydroxycinnamic acid and phenolic compounds. Specific phenolic compounds include phenol, m-cresol and m-chlorocresol.
As previously described, the membrane fluidizer may enhance the fluidization of the biofilm and reduce the level of lipids in the biofilm. Changes in membrane kinetics are indicated by a decrease in steady state anisotropy of fluorescent membrane detection species such as 1, 6-biphenyl-1, 3, 5-hexatriene. Known membrane fluidizers include conventional alcohols or alkenols. Because of the amphoteric nature of these alcohols. The hydroxyl moiety of these alcohols near the polar head group of the phospholipid will separate the membrane lipids into two layers, while the fatty bonds in the alcohol molecules will interact with the fatty acyl groups in the phospholipid. The penetration of long-chain alkenols into the bilayer increases the depth and thus influences the order and kinetic properties of the bilayer to a different extent. See article on Zavoico et al, journal of biochemistry and biophysics, 812: 299-312(1985).
Notably, this document does not claim the use of membrane fluidizers to enhance intermolecular transport, as there is no link between the ability to induce membrane fluidization and to expand intermolecular pathways. See article by Ouyang et al in journal of medicinal chemistry, 45: 2857-2866(2002).
In another embodiment, the compositions of the present invention further comprise a protein structure stabilizer. As previously mentioned, protein structure stabilizers are compounds that stabilize protein structure under hydrated or non-aqueous conditions. The protein structure stabilizer may be a polyanionic molecule such as phytic acid and sucrose octasulfate, and the protein structure stabilizer may also be a polycationic molecule such as arginine. An uncharged polymer; such as polyvinylpyrrolidone and polyvinyl alcohol, may also be used as protein structure stabilizers.
Phytate and its derivatives are biologically active compounds which have a high affinity for several proteins. Phytic acid carries six phosphates which are attached to the cyclohexane ring, thus enabling the phytic acid to bind to the guanidino groups of several arginines. See Filikov et al, article on computer-aided molecular design, 12: 229-240(1998).
As described herein, amphoteric cationic counterions or zwitterionic counterions can be used to effect or promote the crossing of a biological barrier by at least one effector organism for effective transport. Cationic counterions in the present invention are positively charged ions and may contain hydrophobic moieties. The anionic counterions in the present invention are negatively charged ions and may contain hydrophobic moieties. Under appropriate conditions, cationic counter-ions or anionic counter-ions can electrostatically interact with anionic non-penetrating molecules or cationic non-penetrating molecules, respectively. This allows the formation of a complex, whereby the charge is neutralized and thus a new uncharged entity is generated, which has hydrophobic properties due to the inherent hydrophobicity of the counter-ion.
The use of the penetrating composition of the present invention has high reproducibility, the penetrating composition can be widely applied to various therapeutic molecules, and the use is simple and easy; the penetrating composition may be used to effect a highly efficient delivery process across biological barriers in a living organism. Thus, these compositions provide a great improvement over conventional vectors such as liposomes or viruses in the efficient delivery of many macromolecules, including nucleic acids. The method of the present invention utilizes effectors that are contained in water-soluble compositions that are optionally freeze-dehydrated and then submerged in a hydrophobic medium to produce a penetrating composition that is effective to allow macromolecules to traverse biological barriers.
The compositions of the present invention can effectively and non-invasively deliver a substance with unchanged biological activity (such as an effector); thus, the compositions of the present invention have a wide range of uses. For example, the compositions of the present invention may be used to treat diabetes. The concentration of insulin in the blood must be tightly controlled. For example, the compositions of the present invention can be used to allow insulin to efficiently traverse mucosal epithelial cells. Other non-invasive methods of insulin delivery previously known in the art typically deliver only 1-4% of the insulin, which results in unacceptable fluctuations in the amount of insulin absorbed. Another method of treating hyperglycemia involves the use of glucagon-like peptide 1. Glucagon-like peptide 1 is a potent hormone that is secreted in the gastrointestinal tract. An important physiological role of glucagon-like peptide 1 is that it can increase insulin secretion levels in a glucose-based manner, thereby treating diabetes.
In addition, these compositions can be used to treat conditions resulting from atherosclerosis and conditions resulting from the formation of blood columns and emboli; such as myocardial infarction and stroke syndrome. In particular, these compositions can be used to deliver heparin or low molecular weight heparin, allowing the heparin and low molecular weight heparin to cross mucosal epithelial cells. Heparin is a proven safe and effective anticoagulant. However, the efficacy of heparin is limited because it requires parenteral administration. To date. Increasing the absorption rate of heparin in the intestine has met with only limited success, and a sustained, systemic anticoagulant effect has not yet been achieved.
The compositions of the present invention may also be used to treat hematological disorders and ischemic disorders that may be treated using hematogenic factors. For example, erythropoietin is a glycoprotein that stimulates erythropoiesis. This glycoprotein is produced in the kidney and stimulates the division and differentiation of committed red cells in the bone marrow. Endogenously, hypoxia and anemia usually increase erythropoietin production, which in turn stimulates erythropoiesis. However, the production of erythropoietin is impaired in patients with chronic renal failure. The deficiency of erythropoietin is the major cause of anemia in these patients. Recombinant erythropoietin stimulates erythropoiesis in anemic patients with renal failure, including patients requiring dialysis and patients who do not require periodic dialysis. Other anemias that erythropoietin can treat include HIV-infected patients treated with zidovudine and cancer patients in chemotherapy. Anemia in cancer patients may be associated with the disease itself, or with the effects of concurrent chemotherapy.
Another major cause of anemia is pernicious anemia, which is caused by a deficiency in vitamin B12. The absorption mechanism of vitamin B12 in the gastrointestinal tract involves the secretion and binding of intrinsic factors. In pernicious anemic patients, this process is abnormal, thus resulting in insufficient vitamin B12 absorption and anemia. The penetrating composition of the present invention is useful for allowing vitamin B12 to pass through mucosal epithelial cells with high efficiency.
Colony stimulating factors act on glycoproteins of hematopoietic cells by binding to specific cell surface receptors to stimulate proliferation, differentiation, commitment, and activation of certain terminal cell functions. Granulocyte colony stimulating factor controls the production of neutrophils in the bone marrow and affects the proliferation, differentiation of neutrophil progenitors and activation of selected apical cell function. These include boosting the ability of macrophages, initiating cellular metabolism associated with paroxysmal breathing, relying on killing by antibodies, and enhancing the expression of certain functions associated with cell surface antigens. For cancer patients, the recombinant granulocyte colony stimulating factor can safely and effectively promote the recovery of the number of neutrophils after various chemotherapies, thereby preventing various dangerous infections. Granulocyte colony stimulating factor also reduces the recovery time of bone marrow after bone marrow transplantation.
The compositions of the invention may also be used to administer monoclonal antibodies to various disorders. For example, antibodies that block tumor necrosis factor signaling can be used to treat pathological inflammation such as rheumatoid arthritis, polyarticular juvenile rheumatoid arthritis, and the resulting joint conditions.
In addition, the composition of the present invention can be used for treating osteoporosis. Recent studies have shown that intermittent administration of parathyroid hormone, as occurs after injection of recombinant parathyroid hormone, results in an anabolic response, rather than the well-known catabolic response, which results from continuous administration of high concentrations of parathyroid hormone, as is the case with hyperthyroidism. Thus, the use of parathyroid hormone by a non-invasive means is beneficial for increasing bone mass in patients with various deficiency disorders, including for increasing bone mass in patients with osteoporosis. See article by Fox in curr in pharmacoe, 2: 338-344(2002).
Today, delivery of effectors (such as delivery of insulin, erythropoietin or heparin into the blood) requires the use of invasive techniques, such as intravenous or intramuscular injection. One advantage of the compositions of the present invention is that they can be delivered by non-invasive administration of the compositions of the present invention to allow the effectors to cross biological barriers. Such non-invasive administration includes, for example, oral, buccal, nasal, rectal, inhalation, insufflation, dermal or deposit administration. Furthermore, another advantage of the compositions of the present invention is that they can traverse blood brain disorders, delivering effectors into the central nervous system.
The compositions of the present invention are effective in facilitating the passage, transport or penetration of a substance (e.g., an effector) through a biological barrier, particularly across a cell enclosed by a tightly-linked tissue. The detection and quantification of the transport can be carried out by methods known to those skilled in the art, including the use of contrast compounds such as radiotracers or the addition of fluorescent probes or pigments to hydrophobic compositions in combination with a transcellular action assay described in the article "infectious disease & immune" by Schifgaarde et al, 68 (8): 4616-23(2000). In general, the transcellular interaction assay is performed by: (a) culturing the cell layer using the composition of the present invention; (b) preparing a cross section of the cell layer; (c) detecting the presence of an effector substance, or detecting the presence of other components of the compositions of the invention. The detecting step may be carried out by culturing the fixed cell sections with labeled antibodies against the components of the composition of the invention; and detecting the immune reaction between the component and the labeled antibody. Another approach is to label the components of the composition with radioactive, fluorescent tracers or pigments so that the location of the components between the cells can be directly observed. In addition, bioassays may be used to detect the transport of the composition. For example, a decrease in blood glucose concentration may be detected after administration of a bioactive molecule, such as insulin, contained in the composition.
As used herein, "effective transport" means that a composition entering a biological barrier can cause at least 5% of the effector to traverse the biological barrier, preferably at least 10% of the effector to traverse the biological barrier, and more desirably at least 20% of the effector to traverse the biological barrier.
As used herein, the term "effector" refers to a molecule or compound that is not penetrating; these molecules or compounds are useful as biological, therapeutic, pharmaceutical or diagnostic agents. Anionic non-penetrating molecules can be composed of nucleic acids (ribonucleic acids, degassed ribonucleic acids) from various sources, in particular from human, viral, animal, eukaryotic or prokaryotic, plant or synthetic substances. The nucleic acid of interest can be of various sizes, e.g., from simple nucleotides to genomic fragments or the entire genome. The nucleic acid may be a viral gene or a plastid.
Alternatively, the effector of interest may be a protein, such as an enzyme, hormone, incretin, aminodextran, cytokine, apoprotein, growth factor, biologically active molecule, antigen or antibody, or the like. Glycosaminoglycans include heparin, heparin derivatives, heparan sulfate, chondroitin sulfate, dermatan sulfate and hyaluronic acid; however, the aminodextran which can be used as the non-penetrating composition is not limited to these substances. Specific heparin derivatives include low molecular weight heparins such as enoxaparin, dalteparin, tinzaparin and fondaparin; however, the heparin derivative is not limited to these substances. As used herein, the term "bioactive molecule" refers to a compound that acts on or causes a response to a living cell, living tissue, or whole organism. Protein is a non-limiting example of a biologically active molecule. Other examples of biologically active molecules include erythropoietin, glucagon-like peptide 1, melanocyte stimulating hormone, parathyroid hormone 1-34, growth hormone, peptide YY amino acids 3-36(PYY (3-36)), calcitonin, interleukin-2 (IL-2), alpha 1-antitrypsin, granulocyte/monocyte colony stimulating factor (GM-CSF), granulocyte colony-stimulating factor (G-CSF), T20, anti-tumor necrosis factor antibodies, interferon alpha, interferon beta, interferon gamma, Luteinizing Hormone (LH), Follicle Stimulating Hormone (FSH), brain peptide, dalargin, kyphospine, basic fibroblast growth factor (bFGF), hirudin, hirulog, Luteinizing Hormone Releasing Hormone (LHRH) analogs, luteinizing hormone (LHRH), and the like, Brain Natriuretic Peptide (BNP), glatiramer acetate, and neurotrophic factors; however, other suitable proteins are not limited to these substances.
Furthermore, the effector may be a pharmacologically active agent, such as a toxin, a therapeutic agent or an antipathogenic agent, for example an antibiotic, antiviral, antifungal or antiparasitic agent. The effector may be active itself, or activated in situ by the composition, the particular substance, or by the environmental conditions to which it is exposed. Particularly suitable pharmacologically active agents include vitamin B12, metaphosphate, taxol, carpoform or an aminoglycoside antibiotic.
The terms "pharmacologically active agent" and "therapeutic agent" are used interchangeably herein and refer to a chemical substance or compound that is administered to an organism and is capable of causing a detectable pharmacological and/or physiological effect.
The compositions of the invention are characterized in that the penetration capacity of these compositions is practically independent of the effectors contained therein.
"counterions" in the context of the present invention also include anionic or cationic amphoteric molecules, i.e., those molecules which have both polar and nonpolar regions or which have both hydrophilic and hydrophobic properties. The anionic counterions or cationic counterions in the present invention are negatively charged (anionic) and positively charged (cationic) and may contain hydrophobic moieties. Under appropriate conditions, anionic counterions or cationic counterions can electrostatically interact with cationic non-penetrating molecules or anionic non-penetrating molecules, respectively. This complexation formation neutralizes the charge, forming new uncharged moieties, which in turn give rise to hydrophobic properties due to the inherent hydrophobicity of the counterion.
Suitable anionic counterions are ions with negatively charged residues, such as carboxylate, sulfonate or phosphate ions, and may have hydrophobic moieties. Specific anionic counterions include sodium lauryl sulfate, cetyl sulfosuccinate and other anionic compounds derived from organic acids; however, the anionic counter ion is not limited to these.
The ionic liquid is prepared from cation such as imidazolium ion, pyridinium ion and other ions such as BF- 4、PF- 6Such anions constitute solutions of salt species and are relatively low temperature liquids. Ionic liquids are characterized by being liquid over a relatively wide temperature range and by having a relatively high ionic conductivity. When an ionic liquid is used as a reaction solvent, a solute is dissolved only by ions, thereby creating an environment completely different from that created by using water or a general organic solvent. The ionic liquid has higher selectivity, and the application range of the ionic liquid is steadily expanded.
Suitable cationic counterions include quaternary amine derivatives, such as benzalkonium derivatives. Suitable quaternary amines may be partially substituted with hydrophobic residues. In general, the quaternary amines contemplated by the present invention have the structure 1-R1-2-R2-3-R3-4-R4-N, wherein R1, R2, R3, and R4 are alkyl or aryl derivatives. In addition, the quaternary amine may be an ionic liquid foaming cation, such as an imidazolium derivative, a pyridinium derivative, a phosphonium compound, or a tetraalkylammonium compound.
For example, the imidazolium derivatives have the general structure 1-R1-3-R2-imidazolium, wherein R1 and R2 can be straight-chain or branched alkyl groups with 1 to 12 carbon atoms. A portion of these imidazolium derivatives may be substituted with a halogen or an alkyl group. Specific imidazolium derivatives include 1-ethyl-3-methylimidazolium, 1-butyl-3-methylimidazolium, 1-hexyl-3-methylimidazolium, 1-methyl-3-octylimidazolium, 1-methyl-3- (3, 3, 4, 4, 5, 5, 6, 6, 7, 7, 8, 8, 8-tridecafluorooctyl) -imidazolium, 1, 3-dimethylimidazolium, and 1, 2-dimethyl-3-propylimidazolium; however, specific imidazolium derivatives are not limited to these.
The general structure of the pyridinium derivative is 1-R1-3-R2-pyridinium, wherein R1 is a straight or branched alkyl group with 1-12 carbon atoms; r2 is hydrogen or a straight or branched alkyl group having 1 to 12 carbon atoms. Some of these pyridinium derivatives may be substituted with halogen or alkyl groups. Pyridinium derivatives include 3-methyl-1-propylpyridinium, 1-butyl-3-methylpyridinium, and 1-butyl-4-methylpyridinium; however, the pyridinium derivative is not limited to these substances.
The penetrating compositions of the present invention may also contain a surfactant. As previously mentioned, suitable surfactants include ionic detergents and non-ionic detergents. Specific ionic detergents may be fatty acid salts, lecithin or bile salts. Specific fatty acid salts include sodium caprylate, sodium caprate, and sodium dodecanoate. The specific nonionic detergent comprises cremophor, polyethylene glycol fatty alcohol ether, sorbitan fatty acid ester, polyethylene glycol stearate 15 and polyhydroxy body. Specific sorbitan fatty acid esters include sorbitan monolaurate, sorbitan monooleate, and sorbitan monolaurate.
The penetrating compositions of the present invention may also include an adhesive polymer, such as methyl cellulose, ethyl cellulose, hydroxypropylmethyl cellulose (HPMC), or carbopol. These adhesive polymers aid in the formation of the penetrating composition of the present invention and/or aid in the adhesion of the composition to mucosal surfaces. In addition, the penetrating compositions of the present invention may also contain monoglycerides. Specific monoglycerides include glyceryl monocaprylate, glyceryl monodecanoate, glyceryl monolaurate, glyceryl monomyristate, glyceryl monostearate, glyceryl monolaurate, and glyceryl monooleate.
The penetrating compositions of the present invention may also contain a protectant. One particular protectant is a protease inhibitor. An article by Bernkop-Schnurch et al, journal of controlled release, describes suitable protease inhibitors that may be incorporated into the penetrating composition, 52: 1-16(1998). For example, these protease inhibitors include inhibitors of luminal endocrine proteases; such as aprotinin, Bowman-birk inhibitor, soybean trypsin inhibitor, avian ovomucoid inhibitor, avian egg inhibitor, human trypsin inhibitor, camostat mesylate, flavonoid inhibitor, analgesic, leupeptin, p-aminobenzyl ether, serine endopeptidase inhibitor (AEBSF), tosyl-L-lysine-chloromethyl ketone hydrochloride (TLCK), APMSF, diisopropyl fluorophosphoric acid (DFP), phenylmethylsulfonyl fluoride (PMSF), poly (acrylate) derivatives, chymostatin, benzyloxycarbonyl-proline-phenylalanine-CHO, FK-448, sucrose biphenylboronic acid complex, beta-phenylpropionate, elastase inhibitor, methoxyacyl-alanine-proline-valine-chloromethyl ketone (MPCMK), Ethylenediaminetetraacetic acid and chitosan-ethylenediaminetetraacetic acid conjugates. Suitable protease inhibitors also include membrane-bound protease inhibitors such as amino acids, dipeptides, tripeptides, amastatin, hydroxyaminobutyrylleucine, puromycin, bacitracin, phosphonate dipeptide analogs, alpha-aminoboronic acid derivatives, sodium glycocholate, 1, 10-phenanthroline, acivicin, levoserine borates, membrane metallo-endoproteinase, and amiloride phosphate.
The invention also includes methods of making the compositions described herein. For example, the water-soluble composition containing the effector can be dissolved or suspended in a hydrophilic or partially hydrophilic solvent and then immersed in a hydrophobic medium along with a membrane fluidizer to form the composition of the present invention. Alternatively, the effector or mixture of effector and protein stabilizing agents forming the water-soluble composition may be lyophilized together and then suspended in a hydrophobic medium together with the membrane fluidizing agent. Alternatively, other components of the composition may be lyophilized or added during reconstitution of the lyophilized material.
It is well known to those skilled in the art that proteins may be further chemically modified to extend the half-life of the protein's circulation in vivo. By way of non-limiting example, polyethylene glycol residues may be attached to effectors of the present invention. Biomolecules can be conjugated to polyethylene glycol through a polyethylene glycol conjugation process, an established method of extending the circulating half-life of proteins. Polyethylene glycol is a non-toxic water-soluble polymer, and because polyethylene glycol has a large hydrodynamic volume, polyethylene glycol forms a barrier around the molecules with which it forms a conjugate, thereby preventing the conjugated molecules from being cleared away in the kidney, and protecting the conjugated molecules from enzymatic degradation and recognition by immune system cells.
In recent years, incentive-specific polyethylene glycol conjugation methods have been used to prepare biologically active conjugated molecules (e.g., drugs, proteins, reagents, enzymes, etc.) that have biological activities that are higher than or equal to the biological activity of the "parent". These formulations have unique pharmacokinetic and pharmacodynamic properties in vivo; the self-regulated elimination of PEG-filgrastim and the prolonged half-life of interferon alpha-2 a absorption conjugated with PEG specifically reflect the unique pharmacokinetic and pharmacodynamic properties of these preparations in vivo. The administration schedule of the polyethylene glycol conjugate molecule is more convenient for the patient and more acceptable to the patient, which may improve the quality of life of the patient. (see Yowlel et al in Cancer TreatRev 28 Supple, A: 3-6 (4 months 2002))
The invention also includes contacting a biological disorder with a composition of the invention in an amount sufficient; thereby effectively crossing biological barriers. The compositions of the present invention may be provided in vitro, ex vivo or in vivo. In addition, the compositions of the present invention may improve the biological activity of the contained substances. It is therefore another object of the present invention to provide methods of using these compositions to increase effector bioactivity.
In addition to the effects of the penetrating compositions of the present invention, the present invention also provides pharmaceutically acceptable basic or acidic addition salts, hydrates, esters, solvates, prodrugs, metabolites, stereoisomers or mixtures of such substances. The invention also provides pharmaceutical formulations comprising a penetrating composition in association with a pharmaceutically acceptable carrier, diluent, enzyme protein inhibitor, surfactant or excipient. For example, surfactants include polyhydroxy body, polyethylene glycol stearate 15, cremophor, phosphosomes, or bile acids/salts.
The term "pharmaceutically acceptable salts" refers to non-toxic salts of the compounds of the present invention, which are generally prepared by reacting the free base with an appropriate organic acid, inorganic acid or solvent, by which reaction a "pharmaceutically acceptable acidic addition salt" of the compound described herein may be prepared. These compounds still retain the biological activity and properties of the free base. Representative examples of such salts include water-soluble and water-insoluble salts such as acetate, astragaloside (4, 4-diaminostilbene-2, 2' -disulfonate), benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, oxide, butyrate, calcium ethylenediaminetetraacetate, camphorsulfonate, carbonate, chloride, citrate, potassium clavulanate, dihydrochloride, edetate, ethanedisulfonate, propionate laurylsulfate, ethanesulfonate, fumarate, erythromycin, gluconate, glutamate, glycoloylp-aminophenylarsenate, hexafluorophosphate, hexylresorcinol, hydrabamine, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isothionate, lactate, lactobionate, hydrabamate, sodium alginate, laurate, hydroxysuccinate, maleate, mandelate, minoxidil, methyl bromide, methyl nitrate, methyl sulfate, mucate, naphthalenesulfonate, nitrate, diatrizoate, 3-hydroxy-2-naphthoate, oleate, oxalate, palmitate, pamoate (1, 1-methylene-bis-2-hydroxy-3-naphthoate), pantothenate, phosphate/diphosphate, picrate, polygalacturonate, propionate, p-toluenesulfonate, salicylate, stearate, hypoacetate, succinate, sulfate, sulfosalicylate, sumamate, tannate, tartrate, 8-chlorotheophylline, tosylate, triiodonium, and valerate.
In accordance with the methods of the present invention, patients, i.e., humans and animals, can be treated with an effective pharmacological dose or an effective therapeutic dose of the compositions of the present invention. As used herein, the term "pharmacologically or therapeutically effective amount" refers to an amount of a drug or therapeutic agent (effector) that elicits a biological or medical response in a tissue, system, animal or human; these biological and medical responses are sought by researchers and clinicians.
The invention also includes pharmaceutical compositions suitable for use in crossing biological disorders with an effector of interest. Preferably, the composition is suitable for internal use and contains an effective amount of the pharmacologically active compound of the present invention; these pharmacologically active compounds may be present alone or in combination with one or more acceptable pharmaceutical carriers. These compounds are very useful and they have very low toxicity.
Preferred compositions include enteric coated tablets and gelatin or Hydroxypropylmethylcellulose (HPMC) capsule dosage forms, wherein the capsule dosage forms contain an active ingredient and a) a diluent; such as lactose, glucose, sucrose, mannitol, sorbitol, cellulose and/or glycine; b) a protease inhibitor; including aprotinin, Bowman-birk inhibitor, soybean trypsin inhibitor, avian ovomucoid inhibitor, avian egg inhibitor, human trypsin inhibitor, camostat mesylate, flavonoid inhibitor, analgesic, leupeptin, p-aminobenzyl ether, serine endopeptidase inhibitor (AEBSF), tosyl-L-lysine-chloromethyl ketone hydrochloride (TLCK), APMSF, diisopropyl fluorophosphoric acid (DFP), phenylmethylsulfonyl fluoride (PMSF), poly (acrylate) derivatives, chymostatin, benzyloxycarbonyl-proline-phenylalanine-CHO, FK-448, sucrose biphenylboronic acid complex, beta-phenylpropionate, elastase inhibitor, methoxyacyl-alanine-proline-valine-chloromethyl ketone (MPCMK), Ethylenediaminetetraacetic acid and chitosan-ethylenediaminetetraacetic acid conjugates. Suitable protease inhibitors also include membrane-bound protease inhibitors such as amino acids, dipeptides, tripeptides, amastatin, hydroxyaminobutyrylleucine, puromycin, bacitracin, phosphonate dipeptide analogs, alpha-aminoboronic acid derivatives, sodium glycocholate, 1, 10-phenanthroline, acivicin, levoserine borates, membrane metallo-endoproteinase, and amiloride phosphate; however, the protease inhibitor is not limited to these substances; c) lubricants, such as silica, talc, stearic acid and its magnesium or calcium salts, polyols and/or polyethylene glycols; binders also used for tablets d) such as magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone; if desired, e) disintegrating agents, such as starch, agar, alginic acid or sodium alginate, effervescent mixtures; and/or f) absorbents, colorants, flavors, and sweeteners. The composition can be sterilized; and/or adjuvants, such as preservatives, stabilizers, wetting agents, emulsifiers, solubility promoters, salts for regulating the osmotic pressure and/or buffers. In addition, the composition may contain other substances having therapeutic effects. The composition can be prepared by conventional mixing, granulating or coating methods, respectively; the composition contains about 0.001-75% active ingredient, and preferably the composition contains about 0.01-10% active ingredient.
Administration of the active compounds and salts described herein can be by any acceptable method of administration. These methods include oral, buccal, anal, rectal, bronchial, pulmonary, nasal, sublingual, intraocular, parenteral, dermal or topical administration. As used herein, "parenteral administration" refers to injection by a route other than the alimentary canal, such as by subcutaneous injection, intramuscular injection, intraocular injection (i.e., into the eye rim or behind the eyeball), intravesicular injection, intraspinal injection, or intravenous injection.
Depending on the mode of administration to be employed, these compositions may be presented in solid, semi-solid or liquid form, for example, the compounds may be presented in the form of tablets, emulsions, creams, ointments, suppositories, pills, sustained release capsules, powders, liquids, suspensions, sprays, aerosols or the like, preferably in unit dosage forms. These compositions contain an effective amount of the active compound or an acceptable pharmaceutical salt; in addition, these compositions may contain conventional pharmaceutical excipients as well as other drugs or agents, carriers, adjuvants, diluents, protease inhibitors and the like, which are conventional in pharmaceutical science.
For compositions in solid form, excipients include pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like. The active compounds described above may also be formulated as suppositories using polyalkylene glycols as carriers, such as propylene glycol.
For example, a liquid composition can be prepared by dissolving, dispersing, emulsifying, and the like. The active compound may be dissolved in or mixed with a pharmaceutically acceptable pure solvent, such as water, saline, hydrated dextrose, glycerol, propylene glycol, ethanol, and the like, to form a solution or suspension.
If desired, the pharmaceutical compositions to be used may also contain minor amounts of non-toxic auxiliary substances such as wetting agents, emulsifying agents, pH buffering agents and other substances such as sodium acetate, triethanolamine oleate, etc.
Those skilled in the art will recognize that the penetrating compositions of the present invention may also be used in mucosal vaccination, i.e., oral, nasal, rectal, vaginal or bronchial vaccination methods; vaccines contain antigens required for vaccination, which are effectors. Such vaccines comprise a composition comprising a desired antigenic sequence including the protective antigen of anthrax or the surface antigen of Hepatitis B (HBs); however, the antigen sequence is not limited to these two substances. The composition can be administered to an individual in need of vaccination by oral or nasal administration. The composition for mucosal vaccination may be administered to humans, but also to other animals. Humans and animals are in the general sense "individuals" or "patients". These animals include agricultural animals such as cattle, sheep, goats, horses, birds as well as cats, dogs and any other animal being cared for.
An "antigen" is a molecule or portion of a molecule that is capable of stimulating an immune response, and in addition, an antigen is capable of inducing the production of antibodies in animals and humans, which antibodies are capable of binding to the antigenic determinants of the antigen. An "epitope" is a portion of a molecule that is recognized and bound by a molecule of the major histocompatibility complex, which is recognized by T cells and bound by an antibody. Typical antigens carry one or more epitopes. Specific recognition indicates that the antigen will react with high selectivity with the corresponding major histocompatibility complex and T cells or antibodies, but not with the numerous other antibodies induced by other antigens.
The peptides immunoreact with the T cell or antibody when the peptide is bound to a major histocompatibility complex and recognized by the T cell or bound to the antibody by virtue of recognition by a particular epitope contained in the peptide. Immunoreactivity can be determined by T cell response or antibody binding in vitro, more specifically, by antibody binding kinetics or by competitive binding using known peptides; the peptides contain the epitopes with which the antibodies and T cells are to interact.
Techniques for determining whether a peptide is immunoreactive with a T cell or an antibody are known in the art. The efficacy of peptide substances can be screened by in vitro or in vivo assays. These assays involve immunizing animals with peptide substances, such as mice, rabbits or primates, and then evaluating the resulting antibody titer.
The invention also includes vaccines that induce the production of secreted antibodies to the corresponding antigens, such that the antibodies serve as a first line of defense against a variety of pathogens. Mucosal vaccination has the advantage that it is achieved by non-invasive administration, and mucosal vaccination is the preferred means of obtaining secreted antibodies, although vaccination may be performed by various means, such as subcutaneous, intraperitoneal, viral, intravascular administration.
The compositions of the present invention may be administered orally, such as in the form of tablets, capsules (containing both sustained release and continuous release components), pills, powders, granules, elixirs, tinctures, suspensions, syrups, creams, sprays, and emulsions. The compositions of the present invention may also be administered nasally in the form of sprays, gels, emulsions or creams.
The dosage regimen of the compositions of the present invention may be selected in accordance with a variety of factors; these determinants include the type, species, age, sex and medical condition of the patient, as well as the severity of the condition to be treated, the route of administration, the liver and kidney function of the patient, and the particular compound or salt used. The effective dosage of the drug required to prevent, treat or arrest the progress of the condition can be readily determined by the ordinarily skilled physician or veterinarian.
In the oral dosage forms of the present invention, these oral dosage forms may be in the form of scored tablets or capsules, as may produce a significant effect; these tablets or capsules contain 0.001, 0.0025, 0.005, 0.01, 0.025, 0.1, 0.25, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100.0, 250.0, 500.0 or 1000.0 mg of the active ingredient.
The compositions of the present invention may be administered in a single daily dose, or the total daily dosage may be divided into two, three or four administrations. Furthermore, preferred compounds of the invention may be administered orally by topical application of suitable buccal agents; bronchial administration by means of a suitable aerosol or inhalant; nasal administration via a suitable nasal vehicle; skin administration is carried out by using a skin patch mode; these modes of administration are well known to those skilled in the art and when using dermal administration, the release of the drug is of course continuous rather than intermittent. Other preferred topical dosage forms include creams, oils, lotions, aerosol sprays, and gels, wherein the active ingredient is present at a concentration of between 0.001% and 50% by weight or volume.
The compounds described in detail herein are generally employed as the active ingredient in admixture with suitable pharmaceutical diluents, excipients or carriers (collectively referred to herein as "carrier" materials) which may be suitably selected with regard to the mode of administration desired, i.e., tablets, capsules, elixirs, syrups and the like, in accordance with conventional pharmaceutical procedures.
For example, for tablets or capsules for oral administration, the active pharmaceutical ingredient may be mixed with a non-toxic pharmaceutically acceptable carrier such as ethanol, ethylene glycol, glycerol, water, or the like; in addition, when necessary or desired, suitable binders, lubricants, protease inhibitors, disintegrating agents, and coloring agents can also be added to the mixture. Suitable binders include starches, gelatin, natural sugars such as glucose or beta-lactose, cereal sweeteners, natural and synthetic gums such as acacia, tragacanth, sodium alginate, hydroxymethyl cellulose, polyols, polyethylene glycol, waxes and the like. Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, and the like. The decomposer comprises starch, methyl cellulose, agar, bentonite, xanthan gum and the like; however, the decomposing agent is not limited to these substances.
The compounds of the present invention may also be combined with soluble polymers as carriers for the drug of interest. These polymers include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropyl-methacrylamide-phenol, polyhydroxyethyl-asparagine-phenol, or polyethylene oxide polylysine substituted with palmitoyl residues. Furthermore, the compounds of the present invention may be combined with biodegradable polymers for achieving controlled release of the drug; these biodegradable polymers may be, for example, polylactic acid, polyepsilon carboxyacetolactone, polycarboxybutyric acid, polyorthoesters, acetal resins, polydihydropyrans, polynitrile propionates, and crosslinked or amphipathic block copolymers of hydrogels.
Any of the above compositions may contain 0.001 to 99% active compound, preferably 0.01 to 50% active compound; these compounds are useful as active ingredients in compositions.
Examples are given below in order to more fully embody the preferred embodiments of the present invention. These examples should not be considered as limiting the scope of the invention; the scope of the invention is defined by the appended claims.
Examples of the invention
EXAMPLE 1 insulin action Using the composition of the invention Transepithelial cell disorder
a) Measurement of blood glucose concentration in mice
The compositions of the present invention are prepared by dissolving human insulin, arginine and phytic acid in a double distilled aqueous solution of sodium hydroxide. The solution was freeze-dehydrated and suspended with sodium laurate, octanol, geraniol in a mixture of mineral oil, medium chain triglyceride oil and castor oil. The specific components and concentrations are listed in table 1.
TABLE 1 compositions for insulin delivery
Human insulin in 7mM sodium hydroxide double distilled aqueous solution Arginine (50 mg/ml in double distilled water) Phytic acid (50 mg/ml in double distilled water) Freeze dehydration 10% sodium dodecaneate polyethylene glycol solution 1: 1 octanol and geraniol Mineral oil, medium chain triglyceride and castor oil 1: 1 Acoustic processing Concentration of insulin
PH9.01mg/985μl 0.5mg(10μl) 0.25mg(5μl) 90μl 90μl 820μl 30 seconds 1mg/ml
8 SD mice are used in the experiment, each weight is 175-200 g, and fasting is carried out 18 hours before the experiment. The mice were divided into two groups and anesthetized with 85% ketimine and 15% xylazine at 0.1ml/100 g body weight. Each formulation was administered either intramuscularly (100 ul/mouse containing 1.11 international units of insulin) or rectally (100 ul/mouse containing 208 international units of insulin). Rectal administration was achieved by gently inserting a small plastic tube protected by a soft coating 2 cm deep into the rectal orifice. Blood glucose concentrations were measured at various time periods after administration (see fig. 1), and blood samples were taken from the tail.
As can be seen in figure 1, there was a gradual, significant decrease in glucose concentration following rectal administration of the composition, indicating absorption of insulin from the intestinal tract into the blood.
b) Measurement of insulin concentration in mouse serum
The compositions of the present invention are prepared by dissolving human insulin, arginine and phytic acid in a double distilled aqueous solution of sodium hydroxide. The solution was freeze-dehydrated and suspended with sodium laurate, octanol, geraniol in a mixture of mineral oil, medium chain triglyceride oil and castor oil. The specific components and concentrations are listed in table 2.
TABLE 2 compositions for insulin delivery
Human insulin in 7mM sodium hydroxide double distilled water solution (pH9.0) Arginine (50 mg/ml in double distilled water) Phytic acid (50 mg/ml in double distilled water) Freeze dehydration 10% sodium dodecaneate polyethylene glycol solution 1: 1 octanol and geraniol Mineral oil, medium chain triglyceride and castor oil 1: 1 Acoustic processing Concentration of insulin
1mg/985μl 0.5mg(10μl) 0.25mg(5μl) 90μl 90μl 820μl 30 seconds 1mg/ml
8 SD mice are used in the experiment, each weight is 175-200 g, and fasting is carried out 18 hours before the experiment. The mice were divided into two groups and anesthetized with 85% ketimine and 15% xylazine at 0.1ml/100 g body weight. Each formulation was administered either intramuscularly (100 ul/mouse containing 1.11 international units of insulin) or rectally (100 ul/mouse containing 208 international units of insulin). Rectal administration was achieved by gently inserting a small plastic tube protected by a soft coating 2 cm deep into the rectal orifice. Blood glucose concentrations were measured at various time periods after administration, and blood samples were taken from the tail. In addition, researchers performed insulin radioimmunoassays to determine the concentration of insulin in serum (see table 3).
TABLE 3
Glucose (mg/dL) and insulin (uU), time after administration
Route of administration 0 5 10 20 60 45 60 90
No. 5 mouse Blood sugar (mg/dL) 75 84 78 56 49 21 18 23
Intramuscular injection Glucose (%) 100 112.00 104.00 74.67 65.33 28.00 24.00 30.67
Pancreatic isletsSu, 25ul 15.49 103.6 81.82 78.41 110.55 86.53 86.08 13.73
No. 6 mouse Blood sugar (mg/dL) 78 89 87 63 48 25 22 26
Intramuscular injection Glucose (%) 100 114.1 111.54 80.77 61.54 32.05 28.21 33.33
Insulin, 25ul 19.37 63.22 80.98 42.75 41.31 49.25 58.54 57.51
No. 7 mouse Blood sugar (mg/dL) 84 90 81 56 39 18 18 18
Intramuscular injection Glucose (%) 100 107.14 96.43 66.67 46.43 21.43 21.43 21.43
Insulin, 25ul 20.36 153.22 135.29 152.57 114.8 133.38 122.7 20.01
No. 8 mouse Blood sugar (mg/dL) 80 79 78 77 63 52 41 38
Intramuscular injection Glucose (%) 101 98.75 97.50 96.25 78.75 65.00 51.25 47.50
Insulin, 25ul 7.17 32.37 31.98 28.49 19.37 19.16 19.52 18.31
No. 1 mouse Blood sugar (mg/dL) 74 85 77 61 43 34 28 42
Rectal administration Glucose (%) 100 114.86 104.05 82.43 58.11 45.95 37.84 56.76
Insulin, 25ul 14.08 119.41 118.49 46.99 25.79 26.36 20 10
No. 2 mouse Blood sugar (mg/dL) 60 82 73 57 41 32 24 36
Rectal administration Glucose (%) 100 136.67 121.67 95.00 68.33 53.33 40.00 60.00
Insulin, 25ul 10.42 99.71 88.98 48.39 35.3 30.32 46.069 19.45
No. 3 mouse Blood sugar (mg/dL) 67 83 81 64 39 30 37 54
Rectal administration Glucose (%) 100 123.88 120.90 95.52 58.21 44.78 55.22 80.60
Insulin, 25ul 19.3 83.38 114.59 32.9 24.56 21.69 13.87 14.63
No. 4 mouse Blood sugar (mg/dL) 63 78 75 61 46 23 18 23
Rectal administration Glucose (%) 101 123.81 119.05 96.83 73.02 36.51 28.57 36.51
Insulin, 25ul 12.98 141.25 210.18 92 53.04 37.29 40.78 16.14
Blood glucose concentration decreases with the amount of insulin absorbed into the blood from the intestinal tract (i.e., blood glucose concentration correlates with the insulin absorbed). Therefore, the drug delivery system can replace an insulin injection mode, thereby providing a safe, effective and convenient drug delivery way for diabetics.
C) Measurement of blood glucose concentration and serum insulin concentration in swine
The composition was prepared by dissolving human insulin and polyvinylpyrrolidone, sodium laurate and methylcellulose in a double distilled aqueous solution of sodium hydroxide. The solution was freeze-dehydrated and suspended with octanol and geraniol in a mixture of medium chain triglyceride oil and castor oil, which also contained sorbitan monopalmitate (span-40). The components and concentrations are listed in table 4.
TABLE 4
Human insulin in 7mM sodium hydroxide double distilled water solution (pH9.0) Arginine (50 mg/ml in double distilled water) Polyvinylpyrrolidone (200 mg/ml in double distilled water) 10% sodium dodecaneate polyethylene glycol solution 0.2% sorbitan monopalmitate Freeze dehydration 1: 1 octanol and geraniol 1% span-40 in a mixture of medium chain triglyceride and castor oil at a ratio of 1: 2 Acoustic processing
1mg/985μl 0.5mg 5mg 9mg 1mg 100μl 900μl 30 seconds
6 gilts are used for the experiment, and each gilt weighs 45-50 kg; the pigs for these experiments were deprived of feed 18 hours prior to the experiment. The test gilts were divided into two groups and anesthetized with 66% ketimine and 33% xylazine at 0.3 ml/kg body weight. A cannula is inserted through the skin over the superior vena cava of the piglet to facilitate blood sample collection. Each formulation was administered either intramuscularly (insulin dose of 0.22 International units/kg) or rectally (insulin dose of 1.1 International units/kg). Rectal administration was achieved by gently inserting a small plastic tube 2 cm deep into the rectal orifice. Blood glucose concentrations were determined at various time periods after administration. Insulin radioimmunoassay was used to determine the concentration of insulin in serum (see table 5).
TABLE 5
Glucose (mg/dL) and insulin (μ U) at time post administration
Piggy number Route of administration 0 5 10 20 30 45 60 90
519 Blood sugar (mg/dL) 87 82 84 71 64 55 48 39
intra-SCD injection Glucose (%) 100 94.25 96.55 81.61 73.56 63.22 55.17 44.83
Insulin, 25ul 14.74 34.95 36.9 31.57 32.81 41.09 32.07 36.71
526 Blood sugar (mg/dL) 47 47 40 30 22 18 18 18
intra-SCD injection Glucose (%) 100 100.00 85.11 63.83 46.81 38.30 38.30 38.30
Insulin, 25ul 31.56 65.51 84.88 54.93 61.47 57.62 52.83 48.07
518 Blood sugar (mg/dL) 54 55 52 48 38 31 21 22
Rectal administration Glucose (%) 11000 101.85 96.30 88.89 70.37 57.41 38.89 40.74
Insulin, 25ul 21.11 71.56 60.92 89.19 64.12 23.29 32.4 21.45
520 Blood sugar (mg/dL) 104 95 95 84 57 31 18 22
Rectal administration Glucose (%) 100 91.35 91.35 80.77 54.81 29.81 17.31 21.15
Insulin, 25ul 8.99 170.96 124.38 189.6 166.58 76.96 68.06 24.67
525 Blood sugar (mg/dL) 73 77 75 51 32 20 18 24
Rectal administration Glucose (%) 11000 105.48 102.74 69.86 43.84 27.40 24.66 32.88
Insulin, 25ul 38.23 63.65 146.43 94.39 51.07 26.99 22.27 15.86
527 Blood sugar (mg/dL) 72 68 68 51 28 18 18 21
Rectal administration Glucose (%) 100 94.44 94.44 70.83 38.89 25.00 25.00 29.17
Insulin, 25ul 11.83 60.06 116.63 95.79 42.2 27.03 25.85 25
As can be seen in table 5, there was a gradual and significant decrease in glucose concentration following rectal administration, while the concentration of insulin in the serum was increasing, indicating absorption of insulin from the intestinal tract into the blood.
d) Measurement of blood glucose concentration and insulin concentration in serum of streptozotocin-induced diabetic mice
The composition was prepared by dissolving human insulin and arginine, polyvinylpyrrolidone, sodium dodecanoate in double distilled aqueous solution of sodium hydroxide. The solution is lyophilized and suspended with octanol and geraniol in a mixture of medium chain triglyceride oil and castor oil, which further comprises sorbitan monopalmitate (span-40), methylcellulose (MC-400) and glycerol monooleate. The components and concentrations are listed in table 6.
TABLE 6
Human insulin in 7mM sodium hydroxide double distilled water solution (pH9.0) Arginine (50 mg/ml in double distilled water) Polyvinylpyrrolidone (200 mg/ml in double distilled water) 10% sodium dodecaneate polyethylene glycol solution Geraniol Octanol (I) Freeze dehydration Geraniol Octanol (I) 1% span-40, 2% glyceryl monooleate, 0.2% methylcellulose in a 1: 2 mixture of medium chain triglyceride and castor oil Acoustic processing Concentration of insulin
4mg/3ml 2mg(40μl) 20mg(100μl) 180μl 20μl 20μl 150μl 20μl 700μl 30 seconds 4mg/ml
Streptozotocin was injected into the tail vein of 6 male SD mice to induce insulin-induced diabetes, and the body weight of the mice was 200-250 g. The fasting blood glucose concentration detected 72 hours after streptozotocin injection was 300-400mg/dL, indicating that the mice were already in a diabetic state.
5 diabetic mice were fasted 18 hours before the experiment, and the experimental mice were divided into two groups and anesthetized with 85% ketimine and 15% xylazine at an amount of 0.1 ml/kg body weight. Each formulation was administered either intramuscularly (100 ul/mouse, 0.56 international units of insulin) or rectally (100 ul/mouse, 11.2 international units of insulin). Rectal administration was achieved by gently inserting a small plastic tube protected by a soft coating 2 cm deep into the rectal orifice. Blood glucose concentrations were determined at various time periods after administration. In addition, insulin radioimmunoassay was used to determine the concentration of insulin in serum. (see Table 7).
TABLE 7
Glucose (mg/dL) and insulin (μ U) at time post administration
Route of administration 0 5 10 20 30 45 60
No. 1 mouse 242 270 223 205 220 20
SCD, rectal administration 100 111.57 92.15 84.71 90.91 0.00 8.26
15.51 124.75 179.89 47.5 342.1
No. 2 mouse 30 49 32 27 32 23 20
SCD, rectal administration 100 163.33 106.67 90.00 106.67 76.67 66.67
23.47 242.59 492.25 664.44 668.93 1687.44 423.36
No. 3 mouse 437 411 411 398 378 377 358
SCD, rectal administration 100 94.05 94.05 91.08 86.50 86.27 81.92
26.35 288.24 408.6 299.75 597.4 387.62 593.73
No. 4 mouse 437 401 402 398 406 380 373
SCD, intramuscular injection 100 91.76 91.99 91.08 92.91 86.96 85.35
18.13 47.46 117.91 149.07 216.61 218.97 252.95
No. 5 mouse 239 288 358 269 306 323 299
SCD, intramuscular injection 100 120.50 149.79 112.55 128.03 135.15 125.10
18.49 50.79 58.61 78.92 113.47 52.93 116.72
As can be seen in table 7, there was a gradual and significant decrease in glucose concentration following rectal administration, while the concentration of insulin in the serum was increasing, indicating absorption of insulin from the intestinal tract into the blood.
Example 2 heparin using the composition of the invention was made effective Transepithelial cell disorder
The compositions used in this study were prepared by dissolving human unfractionated heparin, arginine, and phytic acid in bi-distilled aqueous sodium hydroxide. The solution is freeze-dehydrated and then suspended with octanol and geraniol in a mixture of medium chain triglyceride oil and castor oil; the mixture further comprises sorbitan monopalmitate (span-40), methylcellulose (MC-400), glycerol monooleate, and Pluronic (F-127). The specific components and concentrations are listed in table 8.
TABLE 8 compositions for heparin transport
Heparin Arginine Dodecanoic acid Freeze-drying in 7mM sodium hydroxide solution Geraniol Octanol (I) 1% span-40, 2% glycerol monooleate, 0.1% Pluronic, 0.2% methylcellulose in a 1: 2 mixture of medium chain triglyceride and castor oil
10mg 5mg 180μl 100μl 100μl 800μl
The experiment used 5 male mice of CB6/F1, divided into two groups, and anesthetized with 85% ketimine and 15% xylazine, at 0.01ml per 100 grams of body weight. Each formulation was administered either by intraperitoneal injection (100 ul/mouse, containing 0.2mg heparin) or rectally (100 ul/mouse, containing 1mg heparin). Rectal administration is achieved by gently inserting a small plastic tube protected by a soft coating into the rectal orifice 1 cm deep. Clotting was measured at various time periods after administration (see fig. 1), and blood samples were collected from the tail into the capillary.
TABLE 9
The clotting time (in minutes),time after administration
PH Route of administration 0 5 15 30 45 60 60
No. 1 mouse Abdominal injection 1 1 1 4 7 10 15
No. 2 mouse Abdominal injection 1 6 5 10 14 9 10
No. 3 mouse Rectal means 1 3 4 5 4 4 4
No. 4 mouse Rectal means 1.5 3 6 11 14 16 14
No. 5 mouse Rectal means 1 5 2 13 12 12 12
Clotting time increases with the amount of heparin absorbed by the blood from the intestinal tract (i.e., clotting time correlates with the amount of heparin absorbed). Therefore, the drug delivery system can replace the heparin injection drug delivery system.
Example 3 Interferon alpha Using the compositions of the invention Effective transepithelial cell disorders
The composition was prepared by dissolving human interferon alpha, polyvinylpyrrolidone, sodium laurate in a double distilled aqueous solution of sodium hydroxide. The solution was freeze-dehydrated and suspended with octanol and geraniol in a mixture of medium chain triglyceride oil and castor oil, which also contained sorbitan monopalmitate (span-40), methylcellulose (MC-400) and Glyceryl Monooleate (GMO). The components and concentrations are listed in table 10.
TABLE 10 compositions for transporting interferon alpha
Human interferon alpha phosphate buffered saline solution (200. mu.g/ml) 7mM sodium hydroxide double distilled water solution Arginine (50 mg/ml in double distilled water) Polyvinylpyrrolidone (200 mg/ml in double distilled water) 10% sodium dodecaneate double distilled aqueous solution Freeze dehydration Geraniol Octanol (I) 1% span-40, 0.2% methylcellulose, 2% glycerol monooleate in a 1: 2 mixture of medium chain triglyceride and castor oil Acoustic processing Human interferon alpha concentration
250μl(50μg) 375μl 0.5mg(10μl) 2.5mg(25μl) 45μl 25μl 25μl 450μl 30 seconds 100μg/ml
The experiment was carried out using 6 SD male mice divided into two groups and anesthetized with 85% ketimine and 15% xylazine, at a dose of 0.1ml/100 g body weight. The external jugular vein was exposed by removing the superficial skin. The compositions were administered either nasally (25 ul/mouse, containing 2.5mcg interferon alpha) or rectally (50 ul/mouse, containing 5mcg interferon alpha). Nasal administration is achieved by applying the composition to the external nostrils. Blood samples were taken from the jugular vein at various time periods after dosing (see figures 2-3) and interferon alpha in the serum was detected using an enzyme-linked immunosorbent assay.
As can be seen from figures 2-3, significant concentrations of interferon alpha appear in the blood following nasal and rectal administration of the composition, indicating that interferon alpha is absorbed into the blood from the intestinal tract.
Figure 2 shows a comparison, where interferon alpha solution in phosphate buffered saline was administered rectally, using the same dose per mouse. However, no interferon α was detected in blood, and thus no absorption from the intestinal tract was observed.
Example 4 glucagon-like peptide 1 Using compositions of the invention Effective transepithelial cell disorders
The composition is prepared by dissolving human glucagon-like peptide 1, spermine, polyvinylpyrrolidone, sodium laurate, methylcellulose (MC-400) in a double distilled aqueous solution of sodium hydroxide. The solution was freeze-dehydrated and suspended with octanol and geraniol in a mixture of medium chain triglyceride oil and castor oil, which also contained sorbitan monopalmitate (span-40). The components and concentrations are listed in table 11. Comparative compositions were also prepared as described above except that human glucagon-like peptide 1 was not added.
TABLE 11 compositions for transporting glucagon-like peptide 1
Glucagon-like peptide 1 amide in 7mM sodium hydroxide double distilled water solution Arginine Polyvinylpyrrolidone Sodium dodecanedioate Methyl cellulose Freeze dehydration Geraniol Octanol (I) 1% span-40 in a mixture of medium chain triglyceride and castor oil at a ratio of 1: 2
0.5mg 0.25mg 2.5mg 9mg 2mg 50μl 50μl 450μl
The experiment used 6 SD male mice, and these experimental mice were deprived of food 18 hours prior to the experiment. The mice were divided into three groups, and each mouse was fed with 50% aqueous glucose solution by gavage in an amount of 200mg of glucose. Ten minutes later, administration was by intraperitoneal injection (50 ul/mouse, containing 25mg glucagon-like peptide 1) or by rectal means (200 ul/mouse, containing 100mg glucagon-like peptide 1). Rectal administration was achieved by gently inserting a small plastic tube protected by a soft coating 2 cm deep into the rectal orifice. Blood glucose concentrations were measured at various time periods after dosing (see fig. 4), and blood samples were taken from the tail.
As can be seen from fig. 4, the potential for the rise in blood glucose concentration was alleviated after rectal administration of glucagon-like peptide 1, whereas the blood glucose concentration of control mice was rising and the extent of inhibition of blood glucose rise by rectal administration and intraperitoneal administration was substantially similar, indicating that glucagon-like peptide 1 was absorbed into the blood from the intestinal tract.
EXAMPLE 5 mucosal Vaccination with the composition
Compositions for mucosal vaccination contain the desired antigenic sequence; such as protective antigens against anthrax, and protein stabilizers such as spermine and phytic acid, which may be co-dissolved with surfactants such as polyvinylpyrrolidone and sodium laurate and then freeze-dehydrated, and then suspended in a hydrophobic medium, such as medium chain triglycerides or a mixture of tributyrin and castor oil, together with a membrane fluidizing agent such as octanol and geraniol. The possible additional components of the composition are already described herein. The composition can be administered to an individual in need of vaccination by nasal or oral administration.
The method can be used for simply and rapidly inoculating a plurality of people needing to be inoculated. Another advantage of this method is that it can produce high titers of iga and subsequently form iga antibodies on epithelial mucosa.
The effect of vaccination can be shown by measuring the titer of specific antibodies, in particular the titer of immunoglobulin a, and measuring the immune response; for example, the immune response may be an allergic reaction of the skin to the subcutaneous administration of an antigen.
Other embodiments
As will be clearly understood from the specific embodiments detailed above, the unique method of traversing epithelial and endothelial disorders is described herein. Although specific embodiments have been described herein in detail, this description is for the purpose of illustration only and is not intended to limit the scope of the invention, which is defined by the claims appended hereto. In particular, it is contemplated that any substitutions, alterations and modifications may be made to the invention without departing from the spirit and scope of the invention as defined by the appended claims. For example, the selection of a particular tissue type or the selection of a particular effector to be delivered is within the ordinary skill of those having knowledge of the embodiments described herein.

Claims (93)

1. A composition comprising a therapeutically effective amount of at least one effector comprised in a water-soluble composition, wherein said water-soluble composition is immersed in a hydrophobic medium with at least one membrane fluidizing agent, wherein said composition is effective to allow at least one effect to traverse a biological barrier.
2. The composition of claim 1, wherein at least 5% of said at least one effector traverses a biological barrier.
3. The composition of claim 1, wherein at least 10% of said at least one effector traverses a biological barrier.
4. The composition of claim 1, wherein at least 20% of said at least one effector traverses a biological barrier.
5. The composition of claim 1, wherein the water-soluble composition is dissolved in a hydrophilic or partially hydrophilic solvent selected from the group consisting of water, mono-alcohols, di-alcohols, tri-alcohols, and mixtures thereof.
6. The composition of claim 5 wherein said monol is selected from the group consisting of ethanol, propanol, isopropanol, butanol and mixtures thereof.
7. The composition of claim 5 wherein the glycol is propylene glycol.
8. The composition of claim 5 wherein said triol is glycerol.
9. The composition of claim 1, wherein the hydrophobic medium is selected from the group consisting of aliphatic molecules, cyclic molecules, aromatic molecules, and mixtures thereof.
10. The composition of claim 9 wherein said aliphatic hydrophobic medium is selected from the group consisting of mineral oil, paraffin, fatty acids, monoglycerides, diglycerides, triglycerides, ethers and esters.
11. The composition of claim 10 wherein said triglyceride is selected from the group consisting of long chain triglycerides, medium chain triglycerides, short chain triglycerides and mixtures thereof.
12. The composition of claim 11, wherein said long chain triglyceride is castor oil.
13. The composition of claim 11, wherein said short chain triglyceride is tributyrin.
14. The composition of claim 9, wherein the hydrophobic medium of a cyclic structure is selected from the group consisting of terpenoids, cholesterol derivatives, and fatty acid esters of cholesterol.
15. The composition of claim 14, wherein said cholesterol derivative is cholesterol sulfate.
16. The composition of claim 9, wherein said aromatic hydrophobic medium is benzyl benzoate.
17. The composition of claim 1, wherein said membrane fluidizing agent is selected from the group consisting of linear alcohols, branched alcohols, cyclic alcohols, aromatic alcohols and mixtures thereof.
18. The composition of claim 17 wherein said linear alcohol is selected from the group consisting of butanol, pentanol, hexanol, heptanol, octanol, nonanol, decanol, undecanol, dodecanol, and mixtures thereof.
19. The composition of claim 17 wherein said branched alcohol is geraniol or farnesol.
20. The composition of claim 17, wherein the cyclic alcohol is menthol.
21. The composition of claim 17, wherein said aromatic alcohol is selected from the group consisting of benzyl alcohol, 4-hydroxycinnamic acid and phenolic compounds.
22. The composition of claim 21, wherein said phenolic compound is selected from the group consisting of phenol, m-cresol, m-chlorocresol, and mixtures thereof.
23. The composition of claim 1, wherein said at least one effector comprises an impermeable molecule.
24. The composition of claim 23, wherein said non-penetrating molecule is a protein, peptide, polysaccharide, nucleic acid, or nucleic acid analog.
25. The composition of claim 24, wherein said polysaccharide is an aminodextran; the aminodextran is selected from heparin, heparin derivatives, heparan sulfate, chondroitin sulfate, dermatan sulfate, hyaluronic acid and pharmacologically acceptable salts thereof.
26. The composition of claim 25, wherein the heparin derivative is low molecular weight heparin; these low molecular weight heparins are selected from enoxaparin, dalteparin, fondaparin and tinzaparin.
27. The composition of claim 24, wherein said nucleic acid or nucleic acid analog is selected from the group consisting of a deoxyribonucleic acid, a deoxyribonucleic acid analog, a ribonucleic acid, and a ribonucleic acid analog.
28. The composition of claim 23, wherein said non-penetrating molecule is a biologically active molecule; these bioactive molecules are selected from the group consisting of insulin, erythropoietin, glucagon-like peptide 1(GLP-1), melanocyte stimulating hormone (α MSH), parathyroid hormone (PTH), thyroid hormone amino acids 1-34(PTH (1-34)), growth hormone, peptide YY amino acids 3-36(PYY (3-36)), calcitonin, leukokine-2 (IL-2), α 1-antitrypsin, granulocyte/monocyte colony stimulating factor (GM-CSF), granulocyte colony stimulating factor (G-CSF), T20, anti-tumor necrosis factor antibodies, interferon α, interferon β, interferon γ, Luteinizing Hormone (LH), Follicle Stimulating Hormone (FSH), brain fibula, dalargin, kyenkephalin, basic fibroblast growth factor (b) < FGF >, Hirudin, Luteinizing Hormone Releasing Hormone (LHRH) analogs, Brain Natriuretic Peptide (BNP), glatiramer acetate, and neurotrophic factors.
29. The composition of claim 23, wherein said non-penetrating molecule is a pharmaceutically active agent; these pharmaceutically active agents are selected from hormones, growth factors, incretins, neurotrophic factors, anticoagulants, biologically active molecules, toxins, antibiotics, antifungals, antipathogens, antigens, antibodies, monoclonal antibodies, antibody fragments, soluble receptors, immunomodulators, vitamins, antineoplastic agents, cytokines or other therapeutic agents.
30. The composition of claim 29, wherein said pharmaceutically active agent is selected from the group consisting of vitamin B12, metaphosphate, taxol, caspofungin and aminoglycoside antibiotics.
31. The composition of claim 1, wherein said effector further comprises at least one chemical modifier.
32. The composition of claim 31, wherein said at least one effector is selected from the group consisting of hormones, growth factors, incretins, neurotrophic factors, anticoagulants, biologically active molecules, toxins, antibiotics, antifungals, antipathogens, antigens, antibodies, monoclonal antibodies, antibody fragments, soluble receptors, immunomodulators, vitamins, antineoplastic agents, cytokines, and other therapeutic agents.
33. The composition of claim 31, wherein said chemical modifier comprises one or more polyethylene glycol residues attached to the effector.
34. The composition of claim 1, wherein said water-soluble composition further comprises a protein structure stabilizer; these protein structure stabilizers are selected from the group consisting of polycationic molecules, polyanionic molecules, uncharged polymers and mixtures thereof.
35. The composition of claim 34, wherein said polycation is a polyamine.
36. The composition of claim 35, wherein said polyamine is spermine.
37. The composition of claim 34, wherein said polyanionic molecule is selected from the group consisting of phytic acid and sucrose octasulfate.
38. The composition of claim 34 wherein said non-charged polymer is selected from the group consisting of polyvinylpyrrolidone and polyvinyl alcohol.
39. The composition of claim 1, wherein said water soluble composition further comprises an anionic zwitterionic counterion or a cationic zwitterionic counterion
40. A composition according to claim 39, wherein said anionic zwitterionic balancing molecule comprises an organic acid ion selected from the group consisting of carboxylate, sulfonate and phosphate, and wherein said anionic zwitterionic balancing molecule further comprises a hydrophobic moiety.
41. The composition of claim 40, wherein said anionic counterion is selected from the group consisting of sodium lauryl sulfate and cetyl sulfosuccinate.
42. The composition of claim 39, wherein said polycationic amphiphilic molecule is a quaternary amine having a hydrophobic moiety.
43. The composition of claim 42, wherein the quaternary amine has the general structure:
wherein R is1、R2、R3And R4Is an alkyl or aryl group.
44. The composition of claim 43, wherein the quaternary amine is benzalkonium salt.
45. A composition as in claim 39 wherein said counterion is an ionic liquid foaming cation.
46. The composition of claim 45, wherein said ionic liquid foaming cation is an imidazolium derivative, a pyridinium derivative, a phosphonium compound, or a tetraalkylammonium compound.
47. The composition of claim 46 wherein the imidazolium derivative has the general structure 1-R1-3-R2-imidazolium, wherein R1 and R2 are straight or branched chain alkyl groups having 1 to 12 carbon atoms.
48. The composition of claim 47, wherein the imidazolium derivative further contains a halogen or alkyl group substituent.
49. The composition of claim 46 wherein the imidazolium derivative is selected from the group consisting of 1-ethyl-3-methylimidazolium, 1-butyl-3-methylimidazolium, 1-hexyl-3-methylimidazolium, 1-methyl-3-octylimidazolium, 1-methyl-3- (3, 3, 4, 4, 5, 5, 6, 6, 7, 7, 8, 8, 8-tridecafluorooctyl) -imidazolium, 1, 3-dimethylimidazolium, and 1, 2-dimethyl-3-propylimidazolium.
50. The composition of claim 46 wherein the pyridinium derivative has the general structure 1-R1-3-R2-pyridinium, wherein R1 is a straight or branched chain alkyl group having 1-12 carbon atoms; r2 is hydrogen or a straight or branched alkyl group having 1 to 12 carbon atoms.
51. The composition of claim 50 wherein the pyridinium derivative further comprises a halogen or alkyl group substituent.
52. The composition of claim 46 wherein the pyridinium derivative is selected from the group consisting of 3-methyl-1-propylpyridinium, 1-butyl-3-methylpyridinium, and 1-butyl-4-methylpyridinium.
53. A composition as in claim 45, wherein said ionic liquid foaming cation is part of a water soluble salt.
54. The composition of claim 1, wherein the composition further comprises a surfactant selected from the group consisting of ionic detergents, nonionic detergents, and mixtures thereof.
55. The composition of claim 54, wherein said ionic detergent is selected from the group consisting of fatty acid salts, lecithin, cholate, and mixtures thereof.
56. The composition of claim 55, wherein said fatty acid salt is selected from the group consisting of sodium caprylate, sodium caprate, sodium dodecanoate, and mixtures thereof.
57. A composition as in claim 54 wherein said nonionic detergent is selected from the group consisting of polyhydroxy bodies, polyethylene glycol stearate 15, cremophor, polyethylene glycol fatty alcohol ethers, sorbitan fatty acid esters, and mixtures thereof.
58. The composition of claim 57, wherein said sorbitan fatty acid ester is selected from the group consisting of sorbitan monolaurate, sorbitan monooleate, and sorbitan monopalmitate and mixtures thereof.
59. The composition of claim 1, wherein said composition further comprises an adhesive polymer selected from the group consisting of methylcellulose, ethylcellulose, hydroxypropyl methylcellulose (HPMC), and carbopol.
60. The composition of claim 1, wherein said composition further comprises a monoglyceride; the monoglycerides are selected from the group consisting of glyceryl monocaprylate, glyceryl monocaprate, glyceryl monolaurate, glyceryl monomyristate, glyceryl monostearate, glyceryl monolaurate, and glyceryl monooleate, and mixtures thereof.
61. The composition of claim 1 further comprising at least one protective agent.
62. The composition of claim 61, wherein said protectant protease inhibitor is selected from the group consisting of aprotinin, Bowman-birk inhibitor, soybean trypsin inhibitor, avian ovomucoid inhibitor, avian egg inhibitor, human trypsin inhibitor, camostat mesylate, flavonoid inhibitor, analgesic, leupeptin, p-aminobenzyl ether, serine endopeptidase inhibitor (AEBSF), tosyl-levo-lysine-chloromethyl ketone hydrochloride (TLCK), APMSF, isopropylfluorophosphoric acid (DFP), phenylmethylsulfonyl fluoride (PMSF), poly (acrylate) derivatives, chymatin, benzyloxycarbonyl-proline-phenylalanine-CHO, FK-448, sucrose biphenylboronic acid complex, beta-phenylpropionate, elastase inhibitor, Methoxysuccinyl-alanine-proline-valine-chloromethyl ketone (MPCMK), ethylenediaminetetraacetic acid and chitosan-ethylenediaminetetraacetic acid conjugates. Suitable protease inhibitors also include membrane-bound protease inhibitors such as amino acids, dipeptides, tripeptides, amastatin, hydroxyaminobutyrylleucine, puromycin, bacitracin, phosphonate dipeptide analogs, alpha-aminoboronic acid derivatives, sodium glycocholate, 1, 10-phenanthroline, acivicin, levoserine borates, membrane metallo-endoproteinase, and amiloride phosphate, and mixtures thereof.
63. The composition of claim 1, wherein the composition further comprises a mixture of at least two materials selected from the group consisting of nonionic detergents, ionic detergents, sticky polymers, monoglycerides, protease inhibitors, thiol group status modifiers, and antioxidants.
64. The composition of claim 63, wherein the non-ionic detergent is a polyhydroxy body, cremophor, polyethylene glycol fatty alcohol ether, sorbitan fatty acid ester, polyethylene glycol stearate 15.
65. The composition of claim 63, wherein the adhesive polymer is selected from the group consisting of methylcellulose, ethylcellulose, hydroxypropyl methylcellulose (HPMC), and carbopol.
66. The composition of claim 63, wherein said monoglyceride is selected from the group consisting of glyceryl monocaprylate, glyceryl monocaprate, glyceryl monolaurate, glyceryl monomyristate, glyceryl monostearate, glyceryl monolaurate and glyceryl monooleate, and mixtures thereof.
67. The composition of claim 63, wherein the ionic detergent is a fatty acid salt.
68. The composition of claim 63, wherein the protease inhibitor is selected from the group consisting of aprotinin, Bowman-birk inhibitor, soybean trypsin inhibitor, avian ovomucoid inhibitor, avian ovo inhibitor, human trypsin inhibitor, camostat mesylate, flavonoid inhibitor, analgesic, leupeptin, p-aminobenzyl ether, serine endopeptidase inhibitor (AEBSF), tosyl-levo-lysine-chloromethyl ketone hydrochloride (TLCK), APMSF, Diisopropyl Fluorophosphate (DFP), phenylmethylsulfonyl fluoride (PMSF), poly (acrylate) derivatives, chymatin, benzyloxycarbonyl-proline-phenylalanine-CHO, FK-448, sucrose biphenylboronic acid complex, β -phenylpropionate, elastase inhibitor, methoxysuccinyl-alanine-proline-valine-chloromethyl ketone (MPCMK), Ethylenediaminetetraacetic acid and chitosan-ethylenediaminetetraacetic acid conjugates. Suitable protease inhibitors also include membrane-bound protease inhibitors such as amino acids, dipeptides, tripeptides, amastatin, hydroxyaminobutyrylleucine, puromycin, bacitracin, phosphonate dipeptide analogs, alpha-aminoboronic acid derivatives, sodium glycocholate, 1, 10-phenanthroline, acivicin, levoserine borates, membrane metallo-endoproteinase, and amiloride phosphate, and mixtures thereof.
69. The composition of claim 63, wherein the thiol group state modifier is selected from the group consisting of n-acetyl-1-cysteine and a diamide.
70. The composition of claim 63, wherein the antioxidant is selected from the group consisting of vitamin E, deferoxamine mesylate, methylparaben, ethylparaben, and vitamin C, and mixtures thereof.
71. The composition of claim 1 further comprising an acceptable pharmaceutical excipient, an acceptable pharmaceutical carrier, or a mixture of such materials.
72. The composition of claim 1, wherein optionally said water soluble composition is freeze-dehydrated.
73. A process for preparing a composition according to claim 1, which process comprises dissolving or suspending at least one effector substance in a hydrophilic or partially hydrophilic solvent and immersing the resulting solution or suspension in a hydrophobic medium together with a membrane fluidising agent.
74. A process for the preparation of a composition according to claim 1, which process comprises dissolving or suspending a water-soluble composition in a hydrophilic or partially hydrophilic solvent, subjecting the water-soluble composition to freeze-dehydration, and suspending the freeze-dehydrated material in a hydrophobic medium together with a membrane fluidising agent, thereby preparing the composition.
75. The method of claim 74, wherein the step of lyophilizing comprises first lyophilizing the effector and the stabilizer or other component of the carrier or excipient.
76. A method of transporting at least one effector across a biological barrier, the method comprising introducing the composition of claim 1 into the biological barrier, thereby causing the at least one effector to traverse the biological barrier.
77. The method of claim 76, wherein transport across the biological barrier occurs in a tissue comprising epithelial cells and endothelial cells.
78. The method of claim 77, wherein the biological disorder is selected from the group consisting of tight junction tissue and cell membranes.
79. The method of claim 76, wherein the biological disorder comprises the mucosa of the gastrointestinal tract and the blood-brain barrier.
80. A method of mucosal vaccination comprising administering to a subject in need of vaccination a composition according to claim 1, wherein at least one of the compositions comprises an antigen required for the vaccination.
81. The method of claim 80, wherein the antigen required for vaccination is selected from the group consisting of a protective antigen against anthrax and a hepatitis B surface antigen against hepatitis B.
82. A method of treating or preventing a disease or pathological condition, comprising administering to a subject in need thereof a composition of claim 1 in an amount sufficient to treat or prevent the disease or pathological condition in the subject.
83. The method of claim 82, wherein the disease or pathological condition is selected from the group consisting of endocrine disorders, ophthalmic disorders, neurological disorders, cardiovascular disorders, metabolic disorders, renal disorders, hematological disorders, immune system disorders, rheumatic disorders, infectious diseases, neoplastic disorders, and multi-factorial disorders; wherein the endocrine disorder is selected from diabetes, infertility, hormone deficiency, and osteoporosis; neuropathy includes alzheimer's disease and other forms of dementia, parkinson's disease, multiple sclerosis, and chorea; cardiovascular diseases including atherosclerosis, hypercoagulability, hypocoagulability, coronary heart disease, and cerebrovascular disease; metabolic disorders including obesity and vitamin deficiency; kidney diseases including renal failure; blood disorders include solid anemia; immune system disorders and rheumatic disorders including autoimmune diseases and immunodeficiency; infectious diseases include viral, bacterial, fungal and parasitic infections; multifactorial diseases including impotence, chronic pain, depression, different fibrotic conditions and short stature
84. The method of claim 82, wherein the composition is administered by the following route of administration: oral, nasal, dermal, buccal, sublingual, anal, rectal, bronchial, pulmonary, intraocular, parenteral, and topical administration.
85. A kit comprising one or more containers containing a therapeutically or prophylactically effective amount of the composition of claim 1.
86. The composition of claim 1, wherein said composition is an enteric coating.
87. The composition of claim 1, wherein said composition is contained within a capsule.
88. The composition of claim 1, wherein said composition is a tablet, emulsion, oil, suppository or nasal spray.
89. A composition comprising a therapeutically effective amount of at least one effector; the effector is solubilized with spermine, polyvinylpyrrolidone and sodium dodecanoate, wherein the resulting water-soluble composition is lyophilized and suspended in a mixture of medium-chain triglycerides or glycerol tributyrates and castor oil together with a mixture of octanol and geraniol, wherein said composition is capable of transporting at least one effector across a biological barrier.
90. The composition of claim 89, wherein said composition further comprises sorbitan monopalmitate.
91. The composition of claim 89, wherein said composition further comprises glycerol monooleate.
92. The composition of claim 89, wherein said composition further comprises methylcellulose.
93. The composition of claim 89, wherein said composition further comprises cholesterol sulfate.
HK08106131.9A 2004-04-15 2005-04-14 Compositions capable of facilitating penetration across a biological barrier HK1116406A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US60/562,345 2004-04-15

Publications (1)

Publication Number Publication Date
HK1116406A true HK1116406A (en) 2008-12-24

Family

ID=

Similar Documents

Publication Publication Date Title
CN101084016A (en) Compositions capable of readily penetrating biological barriers
US20170106089A1 (en) Compositions capable of facilitating penetration across a biological barrier
US20070275055A1 (en) Compositions capable of facilitating penetration across a biological barrier
EP3727555B1 (en) Oral delivery of active drug substances
Swaan Recent advances in intestinal macromolecular drug delivery via receptor-mediated transport pathways
JP2019048871A (en) Pharmaceutical composition, and related delivery method
US20110091420A1 (en) Injectable Sustained-Release Pharmaceutical Formulation and the Preparation Method Thereof
RU2002133236A (en) NEW PHARMACEUTICAL COMPOSITION
EP1071407B1 (en) Lipophilic microparticles containing a protein drug or antigen and formulation comprising same
US20080311214A1 (en) Polymerized solid lipid nanoparticles for oral or mucosal delivery of therapeutic proteins and peptides
US20050238716A1 (en) Colloidal drug carrier system
JP6837700B2 (en) How to use dipivefrine
EP2968376B1 (en) Composition containing buffered aminoalkyl glucosaminide phosphate derivatives and its use for enhancing an immune response
KR20180021742A (en) Targeted conjugates and their particles and formulations
EA020257B1 (en) THE NEW STABLE POLYETHYLENE GLYCOL CONJUGATE OF INTERFERON ALPHA, REPRESENTED BY ONE POSITIONAL ISOMER PEF-NαH-IFN AND IMMUNOBIOLOGIC PREPARATION BASED THEREON
WO2005084303A2 (en) Interferon-beta polymer conjugates
Das et al. Chitosan-based systems for oral drug delivery applications
HK1116406A (en) Compositions capable of facilitating penetration across a biological barrier
KR20070030264A (en) Formulations for intranasal administration of interferon beta without protein or polypeptide stabilizer components
Conti et al. Controlled delivery of biotechnological products
CN112601541B (en) Biopolymer formulations for oral administration
Lindner et al. Strategies to Improve the Lipophilicity of Hydrophilic Macromolecular Drugs
WO2024218166A1 (en) Reconstitutable dry powder formulations and methods of use thereof
JP2005537328A (en) Increased absorption of substances through the skin and mucous membranes
HK40038632A (en) Oral delivery of active drug substances