US20070184114A1 - Formulation for topical non-invasive application in vivo - Google Patents

Formulation for topical non-invasive application in vivo Download PDF

Info

Publication number
US20070184114A1
US20070184114A1 US11/638,091 US63809106A US2007184114A1 US 20070184114 A1 US20070184114 A1 US 20070184114A1 US 63809106 A US63809106 A US 63809106A US 2007184114 A1 US2007184114 A1 US 2007184114A1
Authority
US
United States
Prior art keywords
formulation
acid
formulation according
alkyl
penetrants
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/638,091
Inventor
Gregor Cevc
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Idea AG
Original Assignee
Idea AG
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=8167168&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20070184114(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Idea AG filed Critical Idea AG
Priority to US11/638,091 priority Critical patent/US20070184114A1/en
Publication of US20070184114A1 publication Critical patent/US20070184114A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • A61K9/1272Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers with substantial amounts of non-phosphatidyl, i.e. non-acylglycerophosphate, surfactants as bilayer-forming substances, e.g. cationic lipids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • A61K31/573Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/58Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids containing heterocyclic rings, e.g. danazol, stanozolol, pancuronium or digitogenin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics

Definitions

  • the invention relates to formulations comprising molecular arrangements which, owing to penetrant adaptability, are capable of penetrating pores in a barrier, despite the fact that the average diameter of said pores is smaller than the average penetrant diameter.
  • the penetrants can transport agents or else enable agent permeation through the pores after said penetrants have entered said pores.
  • the invention especially relates to new additives to said formulations, such as consistency builders, anti-oxidants or microbicides. It further relates to the preparation and use of such formulations wherein the agent is selected from corticosteroids. Finally, it relates to a method for the preparation of all such formulations.
  • the efficacy of any drug action is a multiparameter function in which the instrinsic potency, the accumulation as well as the elimination kinetics of the drug all play a role. While the former is entirely determined by the chemical composition of the drug the latter two parameters are sensitive to the galenic characteristics of agent formulation and also depend on the site and rate of agent administration.
  • Varying the viscosity of the formulation is thus an appropriate means to avoid a number of successive treatments or else to enable appropriately high agent concentrations.
  • Another storage-related problem lies in the prevention of the formulation against affection with microbes, such as bacteria and fungi, as this may also lead to degradation of carrier components and associated agent.
  • Microbiological affection will not only reduce or eliminate both penetration ability of the carrier and activity of the agent, but can moreover lead to severe side-effects during the application of the drug. Therefore the formulation should not only be prevented from microbiological affection during its storage before use of the formulation, but should also be kept without affection once the vessel has been broken for the purpose of the application of the drug.
  • Corticosteroids Mineralocorticoids and glucocorticoids (hereinafter collectively referred to by the more general term “corticosteroids”) are contained in approximately one third of all dermatics which now may be sold over the counter. Corticosteroids are commonly used, for instance, for the topical treatment of inflammatory diseases, but also are widely used for systemic medication, especially in the treatment of allergy-based syndroms.
  • the present invention therefore aims at the solution of the above discussed problems. It especially addresses the problems with regard to storage and use of the specially optimized, highly adaptable agent carriers.
  • Corticosteroidal formulations moreover are to be adjusted in viscosity, and prevented from oxidative degradation and microbiological affection.
  • Another object of the present invention is to provide a method for the preparation of such formulations for non-invasive applications.
  • FIG. 1 shows the results of biological experiments in which oedema-suppression activity of hydrocortisone in a commercial reference creme (open symbols) was tested against the same amount of identical drug in highly deformable lipid vesicles (Transfersomes) (closed symbols) in mice.
  • the upper panel contains time-dependence (“pharmacodynamic”) data, whereas the lower panel gives the dose dependency measured 16 h after the drug application. Data points give the mean values for 3-4 animals.
  • FIG. 2 illustrates the suppression of the arachidonic acid-induced oedema by dexamethasone in a commercial creme (open symbols) or in Transfersomes (closed symbols) as a function of the time after drug administration (upper panel) or of the epicutaneously applied drug dose (lower panel).
  • FIG. 3 provides information related to that given in FIGS. 1 and 2 , but pertaining to a different glucocorticosteroide, triamcinolone acetonide.
  • FIG. 4 presents the results of dose and time dependence measurements for triamcinolone-acetonide applied in a commercial creme (open symbols) or in Transfersomes (closed symbols) on one forearm of a healthy human volunteer.
  • the read-out was the extent of skin blanching caused by the drug, a the tested doses as given in insets.
  • FIG. 5 shows dexamethasone penetration profile in murine skin in vivo (left panel) or in a pig skin ex vivo (right panel). Open symbols were measured with a commercial creme and closed symbols with the suspension of dexamethasone-loaded Transfersomes.
  • FIG. 7 illustrates (pharmaco)kinetics of transcutaneous transport of various corticosteroids, as assessed by measuring the drug derived radioactivity in the serum, following the topical drug administration with ultradeformable vesicles on (closed symbols) or under (open symbols) intact murine skin. Data points give the mean values for 3-4 animals and vertical bars give standard error of the mean. The applied drug dose in relative units is given in insets.
  • FIG. 8 provides some representative data on biological, anti-oedema activity of triamcinolone-acetonide applied on the skin in a commercial lotion or in conventional lipid vesicles, liposomes (lower panel) or else in highly deformable mixed lipid vesicles, Transfersomes (upper panel). All results were determined in the arachidonic acid induced murine ear oedema model. The topically applied doses are given in the panels.
  • Formulation B was based on oleic acid rather than on phospholipids, as the main carrier ingredient.
  • corticosteroids as the agent associated with highly adaptable agent carriers (cf. PCT/EP96/04526; Claim 15; PCT/EP91/01596 Examples 173-175). But, as for said additives, this disclosure provides no more than a general rule, to add said agent to said agent carriers without any further specification, as is however considered essential for the application of the drug. Consequently said prior art only generally teaches the use of corticosteroids as a test agent for the evaluation of pore penetration rate, rather than teaching the preparation of a usable dermatics product based on highly adaptable agent carriers containing corticosteroids.
  • both topical non-systemic administration, and substantially systemic administration of corticosteroidal dermatics is accompanied by the problem that the more gentle acting agents, like hydrocortisone, only exhibit a rather short and week activity, whereas the more recently developed related agents, such as prednicarbat- or triamcinolone-derivatives are more potent and also act longer, but are also more harmful to the body, as they can evoke severe side-effects if they are applied highly concentrated and/or repeatedly.
  • topical corticosteroid delivery mediated by highly adaptable agent carriers can be varied systematically whereby severe side-effects are dramatically reduced or even avoided.
  • between 100% and less than 5% of the locally administered drug can be deposited into the outermost skin region.
  • Low area-dose favors drug retention in the skin, while larger amounts of a drug shift the distribution towards systemic circulation. It is possible to reach therapeutically meaningful drug concentrations in the blood after a single epicutaneous administration of corticosteroids by said carriers, while one can also keep blood level below a few percent.
  • a formulation comprising molecular arrangements capable of penetrating pores in a barrier, owing to penetrant adaptability, despite the fact that the average diameter of said pores is smaller than the average penetrant diameter, provided that the penetrants can transport agents or else enable agent permeation through the pores after penetrants have entered pores comprises at least one consistency builder in an amount that increases the formulation viscosity above that of the non-thickened corresponding formulation to maximally 5 Nm/s so that spreading over, and retention at, the application area is enabled and/or at least one antioxidant in an amount that reduces the increase of oxidation index to less than 100% per 6 months and/or at least one microbicide in an amount that reduces the bacterial count of 1 million germs added per g of total mass of the formulation to less than 100 in the case of aerobic bacteria, to less than 10 in the case of entero-bacteria, and to less than 1 in the case of Pseudomonas aeruginosa or St
  • said at least one consistency buildner is added in an amount that increases the formulation viscosity to up to 1 Nm/s and more preferably to up to 0.2 Nm/s.
  • said at least one antioxidant is added in an amount that reduces the increase of oxidation index to less than 100% per 12 months and more preferably to less than 50% per 12 months.
  • the increase of the oxidation index is reduced to less than 0.45 units, preferably to less than 0.22 units and even more preferably to less than 0.1 units, per 12 month.
  • said at least one microbicide is added in an amount that reduces the bacterial count of 1 million germs added per g of total mass of the formulation to less than 100 in the case of aerobic bacteria, to less than 10 in the case of entero-bacteria, and to less than 1 in the case of Pseudomonas aeruginosa or Staphilococcus aureus , after a period of 3 days, and more preferably after a period of 1 day.
  • anti-oxidant is selected from synthetic phenolic antioxidants, such as butylated hydroxyanisol (BHA), butylated hydroxytoluene (BHT) and di-tert-butylphenol (LY178002, LY256548, HWA-131, BF-389, CI-986, PD-127443, E-5119, BI-L-239XX, etc.), tertiary butylhydroquinone (TBHQ), propyl gallate (PG), 1-O-hexyl-2,3,5-trimethylhydroquinone (HTHQ); aromatic amines (diphenylamine, p-alkylthio-o-anisidine, ethylenediamine derivatives, carbazol, tetrahydroindenoindol); phenols and phenolic acids (guaiacol, hydroquinone, vanillin, gallic acids and their esters, protocatechuic acid, quinic acid, syringic acid
  • BHA but
  • tocopheryl-POE-succinate acetate, -laurate, myristate, -palmitate, -oleate,-linoleate, etc., or an y other suitable tocopheryl-lipoate), tocopheryl-POE-succinate; trolox and corresponding amide and thiocarboxamide analogues; ascorbic acid and its salts, isoascorbate, (2 or 3 or 6)-o-alkylascorbic acids, ascorbyl esters (e.g. 6-o-lauroyl, myristoyl, palmitoyl-, oleoyl, or linoleoyl-L-ascorbic acid, etc.).
  • NSAIDs non-steroidal anti-inflammatory agents
  • drugs interfering with oxidation including but not limited to non-steroidal anti-inflammatory agents (NSAIDs, such as indomethacine, diclofenac, mefenamic acid, flufenamic acid, phenylbutazone, oxyphenbutazone acetylsalicylic acid, naproxen, diflunisal, ibuprofene, ketoprofene, piroxicam, penicillamine, penicillamine disulphide, primaquine, quinacrine, chloroquine, hydroxychloroquine, azathioprine, phenobarbital, acetaminephen); aminosalicylic acids and derivatives; methotrexate, probucol, antiarrhythmics (amiodarone, aprindine, asocainol), ambroxol, tamoxifene, b-hydroxytamoxifene; calcium antagonists (nifedip
  • oat flour extracts such as avenanthramide 1 and 2; thioethers, dithioethers, sulphoxides, tetralkylthiuram disulphides; phytic acid, steroid derivatives (e.g. U74006F); tryptophan metabolites (e.g. 3-hydroxykynurenine, 3-hydroxyanthranilic acid), and organochalcogenides.
  • a preferred concentration of BHA or BHT is between 0.001 and 2 w-%, more preferably is between 0.0025 and 0.2 w-%, and most preferably is between 0.005 and 0.02 w-%, of TBHQ and PG is between 0.001 and 2 w-%, more preferably is between 0.005 and 0.2 w-%, and most preferably is between 0.01 and 0.02 w-%, of tocopherols is between 0.005 and 5 w-%, more preferably is between 0.01 and 0.5 w-%, and most preferably is between 0.05 and 0.075 w-%, of ascorbic acid esters is between 0.001 and 5, more preferably is between 0.005 and 0.5, and most preferably is between 0.01 and 0.15 w-%, of ascorbic acid is between 0.001 and 5, more preferably is between 0.005 and 0.5 w-%, and most preferably is between 0.01 and 0.1 w-%, of sodium bisulphite or sodium metabis
  • the microbicide is selected from short chain alcohols, including ethyl and isopropyl alcohol, chlorbutanol, benzyl alcohol, chlorbenzyl alcohol, dichlorbenzylalcohol, hexachlorophene; phenolic compounds, such as cresol, 4-chloro-m-cresol, p-chloro-m-xylenol, dichlorophene, hexachlorophene, povidon-iodine; parabenes, especially alkyl-parabenes, such as methyl-, ethyl-, propyl-, or butyl- paraben, benzyl paraben; acids, such as sorbic acid, benzoic acid and their salts; quaternary ammonium compounds, such as alkonium salts, e.g.
  • a bromide such as a chloride or a bromide, cetrimonium salts, e.g. a bromide, phenoalkecinium salts, such as phenododecinium bromide, cetylpyridinium chloride and other salts; furthermore, mercurial compounds, such as phenylmercuric acetate, borate, or nitrate, thiomersal, chlorhexidine or its gluconate, or any antibiotically active compounds of biological origin, or any suitable mixture thereof.
  • mercurial compounds such as phenylmercuric acetate, borate, or nitrate, thiomersal, chlorhexidine or its gluconate, or any antibiotically active compounds of biological origin, or any suitable mixture thereof.
  • the bulk concentration of short chain alcohols in the case of ethyl, propyl, butyl or benzyl alcohol preferably is up to 10 w %, more preferably is up to 5 w-%, and most preferably is in the range between 0.5-3 w-%, and the bulk concentration of chlorobutanol preferably is in the range between 0.3-0.6 w-%; furthermore, the preferred bulk concentration of parabenes is in the range between 0.05-0.2 w-%, in the case of methyl paraben, and is in the range between 0.002-0.02 w-%, in the case of propyl paraben; bulk concentration of sorbic acid preferably is in the range between 0.05-0.2 w-%, and in the case of benzoic acid preferably is in the range between 0.1-0.5 w-%; bulk concentration of phenols, triclosan, is preferably in the range between 0.1-0.3 w-%, and bulk concentration of chlorhexidine preferably is
  • consistency builder is selected from pharmaceutically acceptable hydrophilic polymers, such as partially etherified cellulose derivatives, comprising carboxymethyl-, hydroxyethyl-, hydroxypropyl-, hydroxypropylmethyl- or methyl-cellulose; completely synthetic hydrophilic polymers comprising polyacrylates, polymethacrylates, poly(hydroxyethyl)-, poly(hydroxypropyl)-, poly(hydroxypropylmethyl)methacrylate, polyacrylonitrile, methallyl-sulphonate, polyethylenes, polyoxiethylenes, polyethylene glycols, polyethylene glycol-lactide, polyethylene glycol-diacrylate, polyvinylpyrrolidone, polyvinyl alcohols, poly(propylmethacrylamide), poly(propylene fumarate-co-ethylene glycol), poloxamers, polyaspartamide, (hydrazine cross-linked) hyaluronic acid, silicone; natural gums comprising alginates, carrageenan,
  • the polymer weight fractions preferably are in the range between 0.05% and 10%, more preferably are in the range between 0.1% and 5%, even more preferably are in the range between 0.25% and 3.5% and most preferably are in the range between 0.5% and 2%.
  • viscosity is best suited if the consistency builder is added in an amount that increases the formulation viscosity above that of the non-thickened corresponding formulation, preferably to up to 1 Nm/s and even more preferably to up to 0.2 Nm/s.
  • a formulation comprising molecular arrangements capable of penetrating pores in a barrier, owing to penetrant adaptability, despite the fact that the average diameter of said pores is smaller than the average penetrant diameter, provided that the penetrants can transport agents or else enable agent permeation through the pores after penetrants have entered pores, the agents associated with said penetrants being glucocorticoids or mineralocorticosteroids (corticosteroids), is characterised in that the relative content of corticosteroids is above 0.1 weight-%, relative to total dry mass of the formulation. It then is preferred if at least one consistency builder and/or at least one anti-oxidant and/or at least one microbicide, as described herein, is added to the formulation.
  • That corticosteroid is preferably selected from alclonetasone dipropionate, amcinonide, beclomethasone dipropionate, betamethasone, betamethasone 17-valerate, betamethasone 17,21-divalerate, betamethasone 21-acetate, betamethasone 21-buytrate, betamethasone 21-propionate, betamethasone 21-valerate, betamethasone benzoate, betamethasone dipropionate, betamethasone valerate, budesonide, clobetasol propionate, clobetasone butyrate, cortexolone, corticosterone, cortisone, cortisone 17-acetate, 21-deoxybetamethasone, 21-deoxybetamethasone 17-propionate, deoxycorticosterone, desonide, desoxymethasone, dexamethasone, diflorasone diacetate, diflucortolone valerate, flu
  • the penetrants are suspended or dispersed in a polar liquid in the form of fluid droplets surrounded by a membrane-like coating of one or several layers, said coating comprising at least two kinds or forms of amphiphilic substances with a tendency to aggregate, provided that said at least two substances differ by at least a factor of 10 in solubility in said liquid or else that said substances when in the form of homo-aggregates, for the more soluble substance, or of hetero-aggregates, for any combination of both said substances, have a preferred average diameter smaller than the diameter of the homo-aggregates containing merely the less soluble substance; or else provided that the presence of the more soluble substance lowers the average elastic energy of the membrane-like coating in the vicinity of thermal energy.
  • the more soluble substance tends to solubilise the droplet and the content of such substance is up to 99 mol-% of solubilising concentration or else corresponds to up to 99 mol-% of the saturating concentration in the unsolubilised droplet, whichever is higher. It can be an advantage if the content of the more soluble substance is below 50%, especially below 40% and most preferably below 30%, of the respective solubilising concentration of said substance. It also is often advantageous if the content of the more soluble substance is below 80%, preferably below 65% and most preferably below 50% of the saturation concentration of said substance in the droplet.
  • the less soluble amongst the aggregating substances is a lipid or lipid-like material, especially a polar lipid, whereas the substance which is more soluble in the suspending liquid and which increases the droplet adaptability belongs to the class of surfactants or else has surfactant-like properties.
  • a specific embodiment of the invention is prepared from a lipid or lipid-like material (which may be a lipid or a lipoid from a biological source or a corresponding synthetic lipid or any of its modifications), said lipid preferably belonging to the class of pure phospholipids corresponding to the general formula where R 1 and R 2 is an aliphatic chain, typically a C 10-20 -acyl, or -alkyl or partly unsaturated fatty acid residue, in particular an oleoyl-, palmitoeloyl-, elaidoyl-, linoleyl-, linolenyl-, linolenoyl-, arachidoyl-, vaccinyl-, lauroyl-, myristoyl-, palmitoyl-, or stearoyl chain; and where R 3 is hydrogen, 2-trimethylamino-1-ethyl, 2-amino-1-ethyl, C 1-4 -alky
  • said lipid is selected from the group comprising phosphatidylcholines, phosphatidylethanolamines, phosphatidylglycerols, phosphatidylinositols, phosphatidic acids, phosphatidylserines, sphingomyelins and other sphingophospholipids, glycosphingolipids (including cerebrosides, ceramidepolyhexosides, sulphatides, sphingoplasmalogens), gangliosides and other glycolipids or synthetic lipids, in particular with corresponding sphingosine derivatives, or any other glycolipids, whereby two similar or different chains can be ester-groups-linked to the backbone (as in diacyl and dialkenoyl compound) or be attached to the backbone with ether bonds, as in dialkyl-lipids.
  • phosphatidylcholines phosphatidylethanolamines, phosphatidylglycerols,
  • the surfactant or surfactant-like material is a nonionic, a zwitterionic, an anionic or a cationic surfactant, especially a fatty-acid or -alcohol, an alkyl-tri/di/methyl-ammonium salt, an alkylsulphate salt, a monovalent salt of cholate, deoxycholate, glycocholate, glycodeoxycholate, taurodeoxycholate, taurocholate, etc., an acyl- or alkanoyl-dimethyl-aminoxide, esp.
  • a dodecyl- dimethyl-aminoxide an alkyl- or alkanoyl-N-methylglucamide, N-alkyl-N,N-dimethylglycine, 3-(acyldimethylammonio)-alkanesulphonate, N-acyl-sulphobetaine, a polyethylene-glycol-octylphenyl ether, esp. a nonaethylene-glycol-octylphenyl ether, a polyethylene-acyl ether, esp. a nonaethylen-dodecyl ether, a polyethylene-glycol-isoacyl ether, esp.
  • a octaethylene-glycol-isotridecyl ether polyethylene-acyl ether, esp. octaethylenedodecyl ether, polyethylene-glycol-sorbitane-acyl ester, such as polyethylenglykol-20-monolaurate (Tween 20) or polyethylenglykol-20-sorbitan-monooleate (Tween 80), a polyhydroxyethylene-acyl ether, esp.
  • a sorbitane-monoalkylate e.g. in Arlacel or Span
  • alkyl-sulphate salt
  • lauryl- or oleoyl-sulphate sodium deoxycholate, sodium glycodeoxycholate, sodium oleate, sodium taurate
  • a fatty acid salt such as sodium elaidate, sodium linoleate, sodium laurate
  • Penetration rate of agent carriers is often maximized if the average penetrant diameter is chosen to be between 30 nm and 500 nm, preferably between 40 nm and 250 nm, even more preferably between 50 nm and 200 nm and most preferably between 60 nm and 150 nm.
  • Pore penetration rate of agent carriers is often optimized in terms of the relative ratio between penetrant and pore size, if the average diameter of the penetrant is 2 to 25 times bigger than the average diameter of the pores in the barrier, preferably between 2.25 and 15 times bigger, even more preferably between 2.5 and 8 times bigger and most preferably between 3 and 6 times bigger than said average pore diameter.
  • the dry weight of all carrier droplets in a formulation for the use on human or animal skin is 0.01 weight-% (w-%) to 40 w-% of total formulation mass, in particular between 0.1 w-% and 30 w-%, particularly preferably between 0.5 w-% and 20 w-%, and most preferably between 1 w-% and 10 w-%.
  • the dry weight of all carrier droplets in a formulation is advantageously chosen to be in the range between 0.0001 w-% and 30 w-% of total formulation mass.
  • the pH of the carrier suspension is between 4 and 10, preferably between 5 and 9, and even more often up to 8.5, as required in order to maximise the stability of formulation, depending on the pH of the carrier suspension.
  • a method for the preparation of a formulation for non-invasive application in vivo comprises the use of at least one amphiphilic substance, at least one polar fluid, at least one edge-active substance or surfactant, at least one corticosteroid in an amount of more than 0.1 w-% based on total dry mass of the formulation and, in case, other customary ingredients, which together form said formulation.
  • At least one edge-active substance or surfactant, at least one amphiphilic substance, at least one hydrophilic fluid and the agent are dissolved to form a solution and, if required, are mixed separately, the resulting (partial) mixtures or solutions then being combined to subsequently induce, preferably by action of mechanical energy, such as shaking, stirring, vibrating, homogenising, ultrasonication, shear, freezing and thawing, or filtration using convenient driving pressure, the formation of penetrants that associate with and/or incorporate the agent.
  • mechanical energy such as shaking, stirring, vibrating, homogenising, ultrasonication, shear, freezing and thawing, or filtration using convenient driving pressure
  • amphiphilic substances are either used as such, or dissolved in a physiologically compatible polar fluid, which may be water or miscible with water, or in a solvation-mediating agent, together with a polar solution. It further is preferred that said amphiphilic substances are dissolved in highly volatile alcohols, especially ethanol, or in other pharmaceutically acceptable organic solvents, which are then removed, esp. by evaporation, prior to making the final preparation.
  • said polar solution contains at least one edge-active substance or surfactant.
  • the formation of said penetrants is induced by the addition of the required substances into a fluid phase, evaporation from a reverse phase, by injection or dialysis, if necessary under the influence of mechanical stress, such as shaking, stirring, especially high velocity stirring, vibrating, homogenising, ultrasonication, shearing, freezing and thawing, or filtration using convenient, especially low (1 MPa) or intermediate (up to 10 MPa), driving pressure.
  • mechanical stress such as shaking, stirring, especially high velocity stirring, vibrating, homogenising, ultrasonication, shearing, freezing and thawing, or filtration using convenient, especially low (1 MPa) or intermediate (up to 10 MPa), driving pressure.
  • the filtering material having pores sizes between 0.01 ⁇ m and 0.8 ⁇ m, preferably between 0.02 ⁇ m and 0.3 ⁇ m, and most preferably between 0.05 ⁇ m and 0.15 ⁇ m, whereby several filters may be used sequentially or in parallel.
  • said agents and penetrants are made to associate, at least partly, after the formation of said penetrants, e.g. after injecting a solution of the drug in a pharmaceutically acceptable fluid, such as ethanol, 1-and 2-propanol, benzyl alcohol, propylene glycol, polyethylene glycol (molecular weight: 200-400 D) or glycerol into the suspending medium, said penetrants being formed previously, using the corresponding or some other suitable manufacturing method, or simultaneously with the drug injection, if required using a co-solution of the drug and, at least some, penetrant ingredients.
  • a pharmaceutically acceptable fluid such as ethanol, 1-and 2-propanol, benzyl alcohol, propylene glycol, polyethylene glycol (molecular weight: 200-400 D) or glycerol
  • the penetrants, with which the agent molecules are associated and/or into which the agent molecules are incorporated are prepared just before the application of the formulation, if convenient from a suitable concentrate or a lyophylisate.
  • the content of corticosteroids is between 0.1 relative weight % (rw-%) and 20 rw-%, more preferably between 0.25 rw-% and 10 rw-% and even more preferably between 0.5 rw-% and 5 rw-% with regard to total penetrant dry mass.
  • said corticosteroid is triamcinolone or one of its derivatives, such as acetonide, the relative content thereof is below 2 w-%, relative to total dry mass of the drug-loaded carriers, even more preferably is below 1 w-% and most typically is below 0.5 w-%.
  • the corticosteroid is hydrocortisone or one of its derivatives, the relative content thereof is below 20 w-%, relative to total dry mass of the drug-loaded carriers, even more preferably is below 12.5 w-% and most typically is below 5 w-%.
  • said corticosteroid is dexamethasone or one of its derivatives, the relative content thereof is below 15 w-%, relative to total dry mass of the drug-loaded carriers, even more preferably is below 10 w-% and most typically is below 5 w-%.
  • corticosteroid is clobetasol or one of its derivatives, such as propionate, the relative content thereof is below 15 w-%, relative to total dry mass of the drug-loaded carriers, even more preferably is below 10 w-% and most typically is below 5w-%.
  • the content of said corticosteroid is below the saturation maximum, defined as the point at which the corticosteroid begins to crystallise in or outside the carrier, such maximum depending on interactions between the amphiphilic molecules comprising the carrier and the agent molecules and frequently being reflected in the membrane/or oil/water partition coefficient difference for the main carrier component and said corticosteroid or else relates to the mismatch in molecular size between the carrier and corticosteroid molecules, the drugs with a lower compatibility typically having lower saturation values.
  • a permeation enhancer is added, which preferably is selected from 1-acyl-azacycloheptan-2-ones (azones), 1-acyl-glucosides, 1-acyl-polyoxyethylenes, 1-acyl-saccharides, 2-n-acyl-cyclohexanones, 2-n-acyl-1,3-dioxolanes (SEPA), 1,2,3-triacyl-glycerols, 1-alkanols, 1-alkanoic acids, 1-alkyl-acetates, 1-alkyl-amines, 1-alkyl-n-alkyl-polyoxyethylenes, 1-alkyl-alkylates, n-alkyl-beta-D-thioglucosides, 1-alkyl-glycerides, 1-alkyl-propyleneglycols, 1-alkyl-polyoxyethylenes, (1-alkyl-)2-pyrrolidones, alkyl-
  • permeation enhancers are not comparable to the addition of permeation enhancers as already effected in classical galenic preparations, such as ointments and lotions, as in the art, the permeation enhancers are solely added for the purpose of fluidization of the skin.
  • the permeation enhancers are added to speed up drug action which is to speed up distribution between agent carrier and surroundings.
  • This content of permeation enhancer is not suited to substantially fluidize skin in order to increase the pore penetration rate of agent carriers and therefore is inherently different from prior art.
  • said corticosteroid is added in an amount which enables the formulation to be applied corresponding to an area dose, as expressed by the total dry mass of penetrant applied per unit area, of between 0.1 mg cm ⁇ 2 and 15 mg cm ⁇ 2 , more preferably between 0.5 mg cm ⁇ 2 and 10 mg cm ⁇ 2 , particularly preferably between 0.75 mg cm ⁇ 2 and 5 mg cm ⁇ 2 and most preferably between 1 mg cm ⁇ 2 and 2.5 mg cm ⁇ 2 , if said corticosteroid is desired to exert a therapeutic effect in the deep subcutaneous, e.g. muscle or joints, tissue or else in the remote tissues, including the whole body.
  • an area dose as expressed by the total dry mass of penetrant applied per unit area
  • said corticosteroid is added in an amount which enables the formulation to be applied with an area dose, as expressed by the total dry mass of penetrant applied per unit area, of 1 between ⁇ g cm ⁇ 2 and 250 ⁇ g cm ⁇ 2 , more preferably between 2.5 and 100 ⁇ g cm ⁇ 2 , even more preferably between 5 ⁇ g cm ⁇ 2 and 50 ⁇ g cm ⁇ 2 and most preferably between 7.5 ⁇ g cm ⁇ 2 and 20 ⁇ g cm ⁇ 2 , if said corticosteroid is desired to exert a mainly local, that is, superficial, rather than systemic therapeutic effect.
  • consistency and, if necessary other characteristics of the formulation are appropriately selected to enable spraying, smearing, rolling or sponging of the formulation on the application area in particular by using a sprayer, spender, roller or sponge, as appropriate.
  • the area dose is selected to be between 0.1 mg cm ⁇ 2 and 15 mg cm ⁇ 2 , preferably between 0.5 mg cm ⁇ 2 and 10 mg cm ⁇ 2 , particularly preferably between 0.75 mg cm ⁇ 2 and 5 mg cm ⁇ 2 and most preferably between 1 mg cm ⁇ 2 and 2.5 mg cm ⁇ 2 , if said corticosteroid is desired to exert a substantial therapeutic effect in the deep subcutaneous, e.g. muscle or joints, tissue or else in the remote tissues, including the whole body.
  • the area-dose as expressed by the total dry mass of penetrants applied per unit area, is chosen to be between 1 ⁇ g cm ⁇ 2 and 250 ⁇ g cm ⁇ 2 , preferably between 2.5 ⁇ g cm ⁇ 2 and 100 ⁇ g cm ⁇ 2 , more preferably between 5 ⁇ g cm ⁇ 2 and 50 ⁇ g cm ⁇ 2 and most preferably between 7.5 ⁇ g cm ⁇ 2 and 20 ⁇ g cm ⁇ 2 , to achieve mainly local, that is, superficial, rather than systemic effect of the drug.
  • non-invasive application of corticosteroids associated with or encapsulated into penetrants according to the invention is effected by spraying, smearing, rolling or sponging on the application area in particular by using a sprayer, spender, roller or sponge, as appropriate.
  • a preferred use of a formulation in accordance with the invention is for the treatment of inflammatory disease, dermatosis, kidney or liver failure, adrenal insufficiency, aspiration syndrome, Behcet syndrome, bites and stings, blood disorders, such as cold-haemagglutinin disease, haemolytic anemia, hypereosinophilia, hypoplastic anemia, macroglobulinaemia, trombocytopenic purpura, furthermore, for the management of bone disorders, cerebral oedema, Cogan's syndrome, congenital adrenal hyperplasia, connective tissue disorders, such as lichen, lupus erythematosus, polymyalgia rheumatica, polymyositis and dermatomyositis, epilepsy, eye disorders, such as cataracts, Graves' ophthalmopathy, haemangioma, herpes infections, neuropathies, retinal vasculitis, scleritis, for some gastro-intestinal disorders, such
  • composition 73.2 mg, 64.5 mg, 54.8 mg, Soy bean phosphatidylcholine (SPC) 37.7 mg 26.8 mg, 35.5 mg, 45.2 mg, Polysorbate (Tween 80) 62.3 mg 1 mg/g Triamcinolone acetonide 899 mg Phosphate buffer (10 mM, pH 6.5) Preparation:
  • composition 37.74 mg Soy bean phosphatidylcholine (SPC) 62.26 mg Tween 80 0.4 mg Triamcinolone Acetonide 0, 26.25 mg Benzyl alcohol 4.47 g Phosphate buffer 50 mM pH 6.5 0.3 mg Probucol 0.3 mg Desferal Preparation:
  • composition 37.74 mg SPC 62.26 mg Tween 80 35 mg Ethanol 0.4 mg Triamcinolone acetonide 26.25 mg Benzyl alcohol 4.47 g Phosphate buffer (50 mM pH 6.5) 0.3 mg Probucol 0.3 mg Desferal Preparation:
  • Soy bean phosphatidylcholine (SPC) 11.9 g, 12.6 g, 13.4 g, 14.25 g
  • Sodium cholate (NaChol) 80 g
  • Triamcinolone acetonide 1000 g
  • Phosphate buffer pH 7.1
  • the suspension is either: extruded through a 200 nm membrane and then through a 100 nm membrane under pressure; 2) processed by a high pressure homogeniser (run in the low pressure range, e.g. at 200 psi) to yield an opalescent final suspension.
  • composition 377.4 mg Soy bean phosphatidylcholine (SPC) 622.6 mg Tween 80 50 mg Benzyl alcohol 9000 mg Phosphate buffer 50 mM pH 6.5 a) 12.5 mg, 25 mg, 50 mg, Bethamethasone b) 12.5 mg, 25 mg, 50 mg, Bethamethasone dipropionate c) 12.5 mg, 25 mg, 50 mg Bethamethasone 17 valerate d) 12.5 mg, 25 mg, 50 mg Clobetasol-17-propionate e) 12.5 mg, 25 mg, 50 mg Dexamethasone or f) 25 mg, 50 mg, 75 mg, Hydrocortisone g) 12.5 mg, 25 mg, 50 mg Prednicarbate h) 0.75 mg, 12.5 mg, 25 mg Triamcinolone Preparation:
  • composition 347 mg Soy bean phosphatidylcholine (SPC) 623 mg Tween 80 30 mg Sodium dodecyl sulphate (SDS) 50 mg Benzyl alcohol 9000 mg Phosphate buffer 50 mM pH 6.5 25 mg Clobetasol-17-propionate Preparation:
  • Corticosteroid suspension was prepared as described in previous examples, except in that SDS was added dissolved in buffer to act as drug distribution promotor in the target organ.
  • Formulation containing SDS acts significantly more rapidly in human skin blanching assay than the SDS-free formulation.
  • corticosteroid delivery concept promises to lower the danger of adverse side effects of the topical therapy with such medication. This is possible due to dose-lowering and a different drug delivery mechanism: Corticosteroids in Transfersomes can not penetrate directly into the blood vessels, owing to the prohibitively large size of the carriers. Such drugs are thus confined to the intercellular space, where they can exert their desired biological function. (Only when they are applied in ample amounts such therapeutics are distributed throughout the body, first via the lymphatic and then through blood circulation.)
  • phospholipid suspensions When applied onto the intact skin surface, phospholipid suspensions are not detrimental to the skin. On the contrary: certain phospholipid preparations have been reported to improve the hydration (and thus to a minor extent the optical appearance) of the aging skin. Phospholipid suspensions are also non-irritating to the skin, at least up to the degree of 30% degradation.
  • Corticosteroid preparations on the basis of transfersomes will normally be used in a quantity (around 100 mg per 2 days) that will contain lipid amounts comparable to those used parenterally ( ⁇ 75 mg/injection) or orally ( ⁇ 150 mg/day).
  • the recommended daily dose of transfersomal corticosteroids for human use will be appreciably lower ( ⁇ 25 mg), except in the case of hydrocortisone, where a somewhat higher dosage might be required for a whole body therapy.
  • Total phospholipid amount to be placed on the skin in the form of transfersomes-based corticosteroid formulations will always be less than 0.5 g/day. It is also less than 10% of the natural variability of phosphatidylcholine concentration in the plasma of an average, healthy person. In light of these data given below, one can conclude that the corticosteroidal dermatics based on transfersomes from the carrier point of view will be an extremely safe product.
  • corticosteroids based on transfersomes will cause less side effects, if any, than the currently available commercial formulations of such drugs. This, on the one hand, is due to the more favorable bio-distribution of the drugs from the transfersomes, which is concentrated to the tissue to be treated. On the other hand, drugs from the carriers are likely to be taken in relatively higher proportions by the strongly proliferating cells, which are one of the chief natural targets for the corticosteroid therapy. (It is even possible that very low doses of transfersomal corticosteroids will completely eliminate the problem of skin atrophy after the repeated use of such therapeutics.)
  • Soybean phosphatidylcholine was purchased from Lipoid KG (Ludwigshafen, Germany) or Nattermann Phospholipids—Rhone-Poulenc Rorer (Köln, Germany) and was more than 95% pure.
  • the remaining components which are described in detail in the above cited European patent, were from Henkel (Düsseldorf, Germany) or CPC (Hamburg, Germany).
  • the active ingredients (dexamethasone, hydrocortisone, triamcinolone-acetonide) were purchased from Synopharm (Hamburg, Germany).
  • microbicides, chellators and antioxidants were from Ciba-Geigy (Basel, Switzerland) or Synopharm.
  • the bidistilled water in injectable quality was purchased from the local pharmacy.
  • the commercial drug formulations from the local pharmacy were used for comparison (hydrocortisone: Hydrocortisone-Wolff (Wolff, Bielefeld); dexamethasone: Anemul (Pharmasal, Gräfelfing); triamcinolon-acetonide: Volon A Lotio N (Squibb-Hayden, Ober) and Delphicort-cream (Lederle, Wolfratshausen)).
  • Formulations contained between 0.01 w-% and 0.5 w-% specified corticosteroid per mL of carrier suspension.
  • the latter consisted chiefly of phosphatidylcholine (SPC) in a final concentration between 0.5 w-% and 5 w-%.
  • SPC phosphatidylcholine
  • This lipid was was taken up in a buffer and homogenized (for animal experiments: by sonication with a titanium micro-tip, Heat Systems W 380, USA, 30 min, 4° C.; for human therapeutics by other mechanical means).
  • At least one of the carrier components was characterized by its membrane solubilizing capacity, as is required by the basic rationale of Transfersome design and above-cited patent applications of the applicant. Such a embrane-affecting substance was always incorporated into the carriers in the sub-lytic concentration.
  • the final vesicle size was determined with the photon correlation spectroscopy (90°, ALV-5000 ALV-Lasermaschinesgesellschaft, Langen, Germany) and was typically between 100 nm and 200 nm. For experimental use, lipid suspension was diluted when appropriate. More detailed description and characterization data will be given separately.
  • mice were first anesthetized with an intraperitoneal injection of 10 ⁇ L g ⁇ 1 body weight of a mixture containing 6 mL 0.9% NaCl, 1 mL Ketavet 100 (Parke-Davis, Berlin, FRG), and 0.25 mL Rompun (Bayer, Leverkusen, Germany). The appropriate amount of drug formulation was smeared over the inner side of one ear and left to dry out. When so stated, the ear was wiped free of the superficial formulation with a cotton swab.
  • test mouse was anesthetized and arachidonic acid in ethanol (1/2 V/V, 10 ⁇ muL) was applied to the same ear area.
  • Change in the mouse ear edema was determined, either by measuring the ear thickness with a micro-caliper (our method) or by weighing the ear volume of the killed mouse (original procedure). Both these assays deliver similar results. All values are the means of at least 3 independently measured values and bars give standard deviation of their mean.
  • test formulations were applied to one arm at different doses in parallel rows.
  • individual areas of 1 cm 2 were covered.
  • the vaso-constriction over each such skin domain was then determined by visual inspection (at least once by an independent observer who was unaware of the drug application pattern) and the skin blanching score was identified with the number of well defined square corners or edges.
  • Upper panel of FIG. 1 illustrates the biological edema-suppression activity of hydrocortisone in commercial cream (open symbols) and in the highly adaptable lipid vesicles, Transfersomes, (closed symbols). Data give mean values measured from 3-4 animals and error bars represent the corresponding standard deviations.
  • Lower panel of FIG. 1 shows dose versus action, as assessed in the local edema-suppression tests, of hydrocortisone in a commercial cream (open symbols) and transfersomal suspension (closed symbols) after 16 h of action.
  • the maximum in dose vs. action curve is due to the dose dependence of action kinetics (see also FIG. 2 ).
  • Upper panel of FIG. 2 illustrates suppression of the arachidonic acid-induced edema by dexamethasone in the commercial cream (open symbols) or Transfersomes (closed symbols) as the function of time after drug administration on the intact murine skin. In both cases the excess drug was wiped-away from the application site 8 h after administration.
  • Upper panel of FIG. 3 illustrates the biological anti-edema activity of triamcinolone-acetonide in commercial lotion (open symbols) or in Transfersomes (closed symbols) in the murine ear model whereas lower panel shows dose vs. action curve for triamcinolone-acetonide in Transfersomes (full symbols, two different preparations and test series), commercial cream (open boxes) or commercial lotion (open circles) applied on the intact murine skin.
  • triamcinolone-acetonide in commercial products is thus 10-times lower than that of the drug in a suspension of Transfersomes. The latter also prolong the duration of therapeutic effect by the same order of magnitude.
  • triamcinolone-acetonide in the ultradeformable agent carriers exerts a somewhat stronger but moderately less persistent biological function.
  • the anticipated drug concentration for the commercial formulation on the basis of Transfersomes is between 0.005% and 0.02%.
  • FIG. 4 shows vaso-constriction (blanching-assay) in the intact human skin as a function of time, following an epicutaneous administration of triamcinolone-acetonide in Transfersomes (upper panel) or in commercial cream (lower panel).
  • FIG. 5 different drug penetration profiles in the mammalian skin are shown.
  • the data were measured in vivo in mice (left panel) and ex vivo in porcine skin (right panel).
  • Open symbols represent measurements with a commercial cream and closed symbols with the suspension of dexamethasone-loaded Transfersomes.
  • Transfersomes for carrying corticosteroids in the skin flattens the drug penetration profile in the skin.
  • the relative drug concentration increases in the deeper skin region, when compared to the results achieved with the commercial formulation of similar drug.
  • FIG. 6 illustrates corticosteroid accumulation (retention) in the skin after administration by means of Transfersomes on the intact surface.
  • ⁇ and ⁇ correspond to the inner and outer skin regions and ⁇ gives their sum.
  • FIG. 7 illustrates using of Transfersomes for the transcutaneous corticosteroid delivery into the systemic circulation.
  • FIG. 8 relative efficiency of various triamcinolone acetonide formulations as tested by the murine ear edema assay is shown. Comparison of the biological activity of two different kinds of Transfersomes loaded with this drug (upper panel), a commercial cream (lower panel) and of conventional liposomes (lower panel). The latter two data sets are not statistically significant even at the level of 0.1.

Abstract

A formulation comprising molecular arrangements capable of penetrating pores in a barrier, owing to penetrant adaptability, despite the fact that the average diameter of said pores is smaller than the average penetrant diameter, provided that the prenetrants can transport agents or else enable agent permeation through the pores after penetrants have entered pores, characterized in that the formulation comprises at least one consistency builder in an amount that increases the formulation to maximally 5 Nm/s so that spreading over, and retention at, the application area is enabled and/or at least one antioxidant in an amount that reduces the increase of oxidation index to less than 100% per 6 months and/or at least one microbiocide in an amount that reduces the bacterial count of 1 million germs added per g of total mass of the formulation to less than 100 in the case of aerobic bacteria, to less than 10 in the case of entero-bacteria, and to less than 1 in the case of Pseudomonas aeruginosa or Staphilococcus aureus, after a period of 4 days.

Description

    FIELD OF THE INVENTION
  • The invention relates to formulations comprising molecular arrangements which, owing to penetrant adaptability, are capable of penetrating pores in a barrier, despite the fact that the average diameter of said pores is smaller than the average penetrant diameter. The penetrants can transport agents or else enable agent permeation through the pores after said penetrants have entered said pores. The invention especially relates to new additives to said formulations, such as consistency builders, anti-oxidants or microbicides. It further relates to the preparation and use of such formulations wherein the agent is selected from corticosteroids. Finally, it relates to a method for the preparation of all such formulations.
  • BACKGROUND INFORMATION
  • The efficacy of any drug action is a multiparameter function in which the instrinsic potency, the accumulation as well as the elimination kinetics of the drug all play a role. While the former is entirely determined by the chemical composition of the drug the latter two parameters are sensitive to the galenic characteristics of agent formulation and also depend on the site and rate of agent administration.
  • Choosing the right mode and kind of drug application is thus as important as finding the right agent—in medicine as well as in the pharmaceutical industry. For example, if an epicutaneously administered drug is incapable of getting into and/or across the skin barrier such a drug has no practical value even if it has a high intrinsic potency. The same is true for the drugs that get into the skin easily but are there eliminated too rapidly to fully develop the desired biological action. In either case an optimization of agent formulation may help. Devising an improved galenic formulation is also much faster and more inexpensive than the invention of the corresponding new chemical entity.
  • It is already known in the art that the addition of surfactants to a membrane built from an amphiphilic substance may modify the membrane's adaptability to the pores of a porous barrier. Moreover, it has already been suggested that this fact may be used to provide agent transport into and/or across the skin, by incorporating and/or associating the agent into/on miniature droplets surrounded by the corresponding membranes, of at least one or more layers of amphiphilic molecules or an amphiphilic carrier substance, and suspended in a suitable liquid medium. These formulations are based on self-optimizing agent carriers which can penetrate a porous barrier such as skin by the virtue of their extremely high adaptability to the pores. This is described in greater detail in our earlier applications EP 475 160 B1, PCT/EP96/04526, PCT/EP98/5539 and PCT/EP98/6750, which are incorporated herein for reference.
  • Although the above-cited prior art already teaches a formulation comprising highly adaptable topically administered agent carriers which are suitable to enable agent transport into and/or across barriers, such as the human skin, however, these formulations are still capable of optimization in specific galenic characteristics in order to enhance practicability in storage and use. This holds especially true where certain galenic characteristics such as formulation viskosity, chemical resistance to oxidative degradation and/or microbiological stability of the formulation are concerned.
  • To avoid a repeated treatment, e.g. in view of side-effects possibly evoked, and in order to achieve high local agent concentration, it is necessary to appropriately adjust the viscosity of the formulation as this goal will be reached by enlarging the application area and/or layer thickness of the applied formulation. Varying the viscosity of the formulation is thus an appropriate means to avoid a number of successive treatments or else to enable appropriately high agent concentrations.
  • Storage related problems most often arise through lack of chemical resistance of the formulation against oxidative degradation of its components. This will obviously not only be important during the storage of the formulation inside the vessel before the application, but also during the application on the application site, when the formulation is exposed to ambient oxygen. Any oxidative process involving formulation components may not only degrade carrier and agent molecules and therefore continuously destroy both carrier and agent properties, but may also even lead to the formation of free radicals which then will cause further chemical attack on carrier and agent molecules, and therefore lead to an accelerated degradation of the components in the formulation. Ensuring proper storage and use therefore always involves protection of the formulation against oxidative degradation of its components.
  • Another storage-related problem lies in the prevention of the formulation against affection with microbes, such as bacteria and fungi, as this may also lead to degradation of carrier components and associated agent. Microbiological affection will not only reduce or eliminate both penetration ability of the carrier and activity of the agent, but can moreover lead to severe side-effects during the application of the drug. Therefore the formulation should not only be prevented from microbiological affection during its storage before use of the formulation, but should also be kept without affection once the vessel has been broken for the purpose of the application of the drug.
  • Above mentioned problems relating to poor agent transport into and/or across the skin and further to galenic characteristics are quite common to many corticosteroidal dermatics. Mineralocorticoids and glucocorticoids (hereinafter collectively referred to by the more general term “corticosteroids”) are contained in approximately one third of all dermatics which now may be sold over the counter. Corticosteroids are commonly used, for instance, for the topical treatment of inflammatory diseases, but also are widely used for systemic medication, especially in the treatment of allergy-based syndroms.
  • Administered doses between a few micrograms per square centimeter, for the most potent corticosteroidal agents, and up to a milligram per square centimeter for the less powerful drugs hence are quite common. Supraceding this limit reduces the efficacy of the concentration-driven drug permeation into the skin below the therapeutically accepatable level; superceding such drug amounts may result in intolerable local, or even systemic, side effects or else is simply not achievable by means of the classical galenic formulations.
  • For example, by raising the epidermal drug concentration one can increase the rate of drug transfer into the skin; by creating a local drug depot the problem of too rapid agent elimination may be solved. However, using a highly concentrated drug solution on the skin incurs the danger of agent precipitation on the skin and the greater likeliness of undesired side-effects. High skin irritation potential of many depot formulations, for example, is a serious obstacle for the successful therapeutic application of such medications. One of the chief reasons for this is that the currently used skin ointments or creams typically contain at least 0.1% and sometimes up to 5% of active ingredient as well as, a relative great amount of skin permeation enhancers in order to fluidize, which means to “soften” the skin which are however also very harmful to the skin. This is especially true when such drugs are used repeatedly and/or highly concentrated which often results in severe side effects, such as skin atrophy, which then enforces discontinuation of the therapy. Classical galenic formulations thus are generally lacking in potency and duration of biological functions if undesired severe side-effects evoked by a repeated treatment necessary to obtain sufficient agent concentration are to be avoided.
  • In view of the difficulties and problems cited hereinabove it would be desirable to have a formulation based on highly adaptable agent carriers which is more potent and can exert its desired biological function longer than similar drugs in the classical lotion or cream form, whereas severe side effects evoked by a repeated treatment can be reduced or even eliminated. It moreover is desirable to have a formulation based on highly adaptable agent carriers able to transport corticosteroidal agents into and/or across the skin wherein the viscosity of the formulation can be adjusted to enable enlarged application area and/or layer thickness, in order to avoid repetition in the treatment. It would also be very desirable if this formulation could be prevented from oxidative degradation and microbiological affection during its storage and use.
  • SUMMARY OF THE INVENTION
  • The present invention therefore aims at the solution of the above discussed problems. It especially addresses the problems with regard to storage and use of the specially optimized, highly adaptable agent carriers.
  • It is a further object of the present invention to overcome deficiencies of the prior art in delivering corticosteroidal formulations with regard to a well controlled trans- and/or intra-cutaneous transport of such drugs. Corticosteroidal formulations moreover are to be adjusted in viscosity, and prevented from oxidative degradation and microbiological affection.
  • Another object of the present invention is to provide a method for the preparation of such formulations for non-invasive applications.
  • Solutions to these objects in accordance with present invention are defined in the attached independent claims.
  • Convenient solutions with special properties are provided by the subject-matters of the subclaims.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows the results of biological experiments in which oedema-suppression activity of hydrocortisone in a commercial reference creme (open symbols) was tested against the same amount of identical drug in highly deformable lipid vesicles (Transfersomes) (closed symbols) in mice. The upper panel contains time-dependence (“pharmacodynamic”) data, whereas the lower panel gives the dose dependency measured 16 h after the drug application. Data points give the mean values for 3-4 animals.
  • FIG. 2 illustrates the suppression of the arachidonic acid-induced oedema by dexamethasone in a commercial creme (open symbols) or in Transfersomes (closed symbols) as a function of the time after drug administration (upper panel) or of the epicutaneously applied drug dose (lower panel).
  • FIG. 3 provides information related to that given in FIGS. 1 and 2, but pertaining to a different glucocorticosteroide, triamcinolone acetonide.
  • FIG. 4 presents the results of dose and time dependence measurements for triamcinolone-acetonide applied in a commercial creme (open symbols) or in Transfersomes (closed symbols) on one forearm of a healthy human volunteer. The read-out was the extent of skin blanching caused by the drug, a the tested doses as given in insets.
  • FIG. 5 shows dexamethasone penetration profile in murine skin in vivo (left panel) or in a pig skin ex vivo (right panel). Open symbols were measured with a commercial creme and closed symbols with the suspension of dexamethasone-loaded Transfersomes.
  • FIG. 6 demonstrates the level of corticosteroid accumulation (retention) in the skin after different drugs'application on the organ surface by means of Transfersomes (closed down-arrow: the stratum comeum; closed up-arrow: the skin stripped free of the stratum comeum; open diamond: total drug amount in the entire skin (=the sum of the former two).
  • FIG. 7 illustrates (pharmaco)kinetics of transcutaneous transport of various corticosteroids, as assessed by measuring the drug derived radioactivity in the serum, following the topical drug administration with ultradeformable vesicles on (closed symbols) or under (open symbols) intact murine skin. Data points give the mean values for 3-4 animals and vertical bars give standard error of the mean. The applied drug dose in relative units is given in insets.
  • FIG. 8 provides some representative data on biological, anti-oedema activity of triamcinolone-acetonide applied on the skin in a commercial lotion or in conventional lipid vesicles, liposomes (lower panel) or else in highly deformable mixed lipid vesicles, Transfersomes (upper panel). All results were determined in the arachidonic acid induced murine ear oedema model. The topically applied doses are given in the panels. Formulation B was based on oleic acid rather than on phospholipids, as the main carrier ingredient.
  • DESCRIPTION
  • As mentioned above the preparation and use of a formulation based on highly adaptable agent carriers have already been described in references incorporated herein. From these it is already generally known to add consistency builders and anti-oxidants to some such formulations, (cf. e.g. PCT/EP96/04526; Claim 18). However, this teaching is a general rule without any practical value, as it obviously lacks any specification for the use. This holds especially true, for instance, for the addition of a consistency builder which enables the formulation to be adjusted to the intended dose of the drug. This addition can obviously not be effected by a simple trial-and-error procedure, or accidentally, by the skilled person, since final drug action is essentially concerned. It is moreover essential to appropriately select type and amount of the added anti-oxidant or microbicide, as this obviously affects storage and use of the formulation.
  • It is known from prior art to use corticosteroids as the agent associated with highly adaptable agent carriers (cf. PCT/EP96/04526; Claim 15; PCT/EP91/01596 Examples 173-175). But, as for said additives, this disclosure provides no more than a general rule, to add said agent to said agent carriers without any further specification, as is however considered essential for the application of the drug. Consequently said prior art only generally teaches the use of corticosteroids as a test agent for the evaluation of pore penetration rate, rather than teaching the preparation of a usable dermatics product based on highly adaptable agent carriers containing corticosteroids. This is indicated by the total amount of hydrocortisone which is to be incorporated in the highly adaptable carrier (examples 173-175 of PCT/EP91/01596: 10 Mikrograms per about 100 mg dry weight of agent carrier). The very low relative proportion of about 0.1 per mille of hydrocortisone based on the total dry weight of formulation is far away from any therapeutically useful drug concentration and also far away from any corticosteroid concentration given in this application.
  • Moreover, the prior art does not teach, how the specific application of corticosteroids is to be effected if, more systemic or else more topical drug action is to be achieved. It therefore is necessary to separately adress the problem of both systemic and non-systemic drug action of applied corticosteroidal dermatics based on the highly adaptable agent carriers, as it is done in this application.
  • Furthermore, in general, both topical non-systemic administration, and substantially systemic administration of corticosteroidal dermatics is accompanied by the problem that the more gentle acting agents, like hydrocortisone, only exhibit a rather short and week activity, whereas the more recently developed related agents, such as prednicarbat- or triamcinolone-derivatives are more potent and also act longer, but are also more harmful to the body, as they can evoke severe side-effects if they are applied highly concentrated and/or repeatedly.
  • In contrast to this, topical corticosteroid delivery mediated by highly adaptable agent carriers can be varied systematically whereby severe side-effects are dramatically reduced or even avoided. Depending on the precise application conditions and carrier design, between 100% and less than 5% of the locally administered drug can be deposited into the outermost skin region. Low area-dose favors drug retention in the skin, while larger amounts of a drug shift the distribution towards systemic circulation. It is possible to reach therapeutically meaningful drug concentrations in the blood after a single epicutaneous administration of corticosteroids by said carriers, while one can also keep blood level below a few percent.
  • Unexpectedly, employment of such highly adaptable agent carriers together with an agent selected from corticosteroids provides biologically efficient product at unprecedent small doses per area. As is shown further below, all tested corticosteroids thus gained in potency (by the factor of 2 to 10) and in duration of action (by up to 5-fold) when they were administered on the intact skin by means of highly adaptable agent carriers. In contrast to prior art, ointments and creams containing corticosteroids, minute amounts of corticosteroids in highly adaptable agent carriers, consequently, suffice for good biological drug action.
  • Generally, material abrasion from the surface shortens the therapeutic effect in a conventional cream or lotion. It is another advantage of the present invention that such problems are not obversed with the formulations based on highly adaptable agent carriers which thus exert a much longer biological action than standard corresponding medications. This is due to the fact that such highly adaptable agent carriers generate a drug depot in the viable skin portions, rather than on the skin surface.
  • It is a characteristic feature of the present invention, that a formulation comprising molecular arrangements capable of penetrating pores in a barrier, owing to penetrant adaptability, despite the fact that the average diameter of said pores is smaller than the average penetrant diameter, provided that the penetrants can transport agents or else enable agent permeation through the pores after penetrants have entered pores comprises at least one consistency builder in an amount that increases the formulation viscosity above that of the non-thickened corresponding formulation to maximally 5 Nm/s so that spreading over, and retention at, the application area is enabled and/or at least one antioxidant in an amount that reduces the increase of oxidation index to less than 100% per 6 months and/or at least one microbicide in an amount that reduces the bacterial count of 1 million germs added per g of total mass of the formulation to less than 100 in the case of aerobic bacteria, to less than 10 in the case of entero-bacteria, and to less than 1 in the case of Pseudomonas aeruginosa or Staphilococcus aureus, after a period of 4 days. It thus is possible to prolong storage and use of the formulation and to advantageously increase practicability of the formulation.
  • It then is preferred if said at least one consistency buildner is added in an amount that increases the formulation viscosity to up to 1 Nm/s and more preferably to up to 0.2 Nm/s.
  • It also is preferred if said at least one antioxidant is added in an amount that reduces the increase of oxidation index to less than 100% per 12 months and more preferably to less than 50% per 12 months.
  • For a formulation comprising soy bean phosphatidylcholine as the main degrading species, the increase of the oxidation index is reduced to less than 0.45 units, preferably to less than 0.22 units and even more preferably to less than 0.1 units, per 12 month.
  • In preferred embodiments of the invention said at least one microbicide is added in an amount that reduces the bacterial count of 1 million germs added per g of total mass of the formulation to less than 100 in the case of aerobic bacteria, to less than 10 in the case of entero-bacteria, and to less than 1 in the case of Pseudomonas aeruginosa or Staphilococcus aureus, after a period of 3 days, and more preferably after a period of 1 day.
  • It is preferred if that anti-oxidant is selected from synthetic phenolic antioxidants, such as butylated hydroxyanisol (BHA), butylated hydroxytoluene (BHT) and di-tert-butylphenol (LY178002, LY256548, HWA-131, BF-389, CI-986, PD-127443, E-5119, BI-L-239XX, etc.), tertiary butylhydroquinone (TBHQ), propyl gallate (PG), 1-O-hexyl-2,3,5-trimethylhydroquinone (HTHQ); aromatic amines (diphenylamine, p-alkylthio-o-anisidine, ethylenediamine derivatives, carbazol, tetrahydroindenoindol); phenols and phenolic acids (guaiacol, hydroquinone, vanillin, gallic acids and their esters, protocatechuic acid, quinic acid, syringic acid, ellagic acid, salicylic acid, nordihydroguaiaretic acid (NDGA), eugenol); tocopherols (including tocopherols (alpha, beta, gamma, delta) and their derivatives, such as tocopheryl-acylate (e.g. -acetate, -laurate, myristate, -palmitate, -oleate,-linoleate, etc., or an y other suitable tocopheryl-lipoate), tocopheryl-POE-succinate; trolox and corresponding amide and thiocarboxamide analogues; ascorbic acid and its salts, isoascorbate, (2 or 3 or 6)-o-alkylascorbic acids, ascorbyl esters (e.g. 6-o-lauroyl, myristoyl, palmitoyl-, oleoyl, or linoleoyl-L-ascorbic acid, etc.). It may also be advantageous to use various drugs interfering with oxidation, including but not limited to non-steroidal anti-inflammatory agents (NSAIDs, such as indomethacine, diclofenac, mefenamic acid, flufenamic acid, phenylbutazone, oxyphenbutazone acetylsalicylic acid, naproxen, diflunisal, ibuprofene, ketoprofene, piroxicam, penicillamine, penicillamine disulphide, primaquine, quinacrine, chloroquine, hydroxychloroquine, azathioprine, phenobarbital, acetaminephen); aminosalicylic acids and derivatives; methotrexate, probucol, antiarrhythmics (amiodarone, aprindine, asocainol), ambroxol, tamoxifene, b-hydroxytamoxifene; calcium antagonists (nifedipine, nisoldipine, nimodipine, nicardipine, nilvadipine), beta-receptor blockers (atenolol, propranolol, nebivolol); also useful are the preferentially oxidised compounds, such as sodium bisulphite, sodium metabisulphite, thiourea; chellating agents, such as EDTA, GDTA, desferral; miscellaneous endogenous defence systems, such as transferrin, lactoferrin, ferritin, cearuloplasmin, haptoglobion, heamopexin, albumin, glucose, ubiquinol-10); enzymatic antioxidants, such as superoxide dismutase and metal complexes with a similar activity, including catalase, glutathione peroxidase, and less complex molecules, such as beta-carotene, bilirubin, uric acid; flavonoids (flavones, flavonols, flavonones, flavanonals, chacones, anthocyanins), N-acetylcystein, mesna, glutathione, thiohistidine derivatives, triazoles; tannines, cinnamic acid, hydroxycinnamatic acids and their esters (coumaric acids and esters, caffeic acid and their esters, ferulic acid, (iso-) chlorogenic acid, sinapic acid); spice extracts (e.g. from clove, cinnamon, sage, rosemary, mace, oregano, allspice, nutmeg); camosic acid, carnosol, carsolic acid; rosmarinic acid, rosmaridiphenol, gentisic acid, ferulic acid; oat flour extracts, such as avenanthramide 1 and 2; thioethers, dithioethers, sulphoxides, tetralkylthiuram disulphides; phytic acid, steroid derivatives (e.g. U74006F); tryptophan metabolites (e.g. 3-hydroxykynurenine, 3-hydroxyanthranilic acid), and organochalcogenides.
  • Especially, a preferred concentration of BHA or BHT is between 0.001 and 2 w-%, more preferably is between 0.0025 and 0.2 w-%, and most preferably is between 0.005 and 0.02 w-%, of TBHQ and PG is between 0.001 and 2 w-%, more preferably is between 0.005 and 0.2 w-%, and most preferably is between 0.01 and 0.02 w-%, of tocopherols is between 0.005 and 5 w-%, more preferably is between 0.01 and 0.5 w-%, and most preferably is between 0.05 and 0.075 w-%, of ascorbic acid esters is between 0.001 and 5, more preferably is between 0.005 and 0.5, and most preferably is between 0.01 and 0.15 w-%, of ascorbic acid is between 0.001 and 5, more preferably is between 0.005 and 0.5 w-%, and most preferably is between 0.01 and 0.1 w-%, of sodium bisulphite or sodium metabisulphite is between 0.001 and 5, more preferably is between 0.005 and 0.5 w-%, and most preferably is between 0.01-0.15 w-%, of thiourea is between 0.0001 and 2 w-%, more preferably is between 0.0005 and 0.2, and most preferably is between 0.001-0.01 w-%, most typically 0.005 w-%, of cysteine is between 0.01 and 5, more preferably is between 0.05 and 2 w-%, and most preferably is between 0.1 and 1.0 w-%, most typically 0.5 w-%, of monothioglycerol is between 0.01 and 5 w-%, more preferably is between 0.05 and 2 w-%, and most preferably is between 0.1-1.0 w-%, most typically 0.5 w-%, of NDGA is between 0.0005-2 w-%, more preferably is between 0.001-0.2 w-%, and most preferably is between 0.005-0.02 w-%, most typically 0.01 w-%, of glutathione is between 0.005 and 5 w-%, more preferably is between 0.01 and 0.5 w-%, and most preferably is between 0.05 and 0.2 w-%, most typically 0.1 w-%, of EDTA is between 0.001 and 5 w-%, even more preferably is between 0.005 and 0.5 w-%, and most preferably is between 0.01 and 0.2 w-%, most typically between 0.05 and 0.975 w-%, of citric acid is between 0.001 and 5 w-%, even more preferably is between 0.005 and 3 w-%, and most preferably is between 0.01-0.2, most typically between 0.3 and 2 w-%.
  • In preferred embodiments of the invention, the microbicide is selected from short chain alcohols, including ethyl and isopropyl alcohol, chlorbutanol, benzyl alcohol, chlorbenzyl alcohol, dichlorbenzylalcohol, hexachlorophene; phenolic compounds, such as cresol, 4-chloro-m-cresol, p-chloro-m-xylenol, dichlorophene, hexachlorophene, povidon-iodine; parabenes, especially alkyl-parabenes, such as methyl-, ethyl-, propyl-, or butyl- paraben, benzyl paraben; acids, such as sorbic acid, benzoic acid and their salts; quaternary ammonium compounds, such as alkonium salts, e.g. a bromide, benzalkonium salts, such as a chloride or a bromide, cetrimonium salts, e.g. a bromide, phenoalkecinium salts, such as phenododecinium bromide, cetylpyridinium chloride and other salts; furthermore, mercurial compounds, such as phenylmercuric acetate, borate, or nitrate, thiomersal, chlorhexidine or its gluconate, or any antibiotically active compounds of biological origin, or any suitable mixture thereof.
  • In especially preferrred embodiments, the bulk concentration of short chain alcohols in the case of ethyl, propyl, butyl or benzyl alcohol preferably is up to 10 w %, more preferably is up to 5 w-%, and most preferably is in the range between 0.5-3 w-%, and the bulk concentration of chlorobutanol preferably is in the range between 0.3-0.6 w-%; furthermore, the preferred bulk concentration of parabenes is in the range between 0.05-0.2 w-%, in the case of methyl paraben, and is in the range between 0.002-0.02 w-%, in the case of propyl paraben; bulk concentration of sorbic acid preferably is in the range between 0.05-0.2 w-%, and in the case of benzoic acid preferably is in the range between 0.1-0.5 w-%; bulk concentration of phenols, triclosan, is preferably in the range between 0.1-0.3 w-%, and bulk concentration of chlorhexidine preferably is in the range between 0.01-0.05 w-%.
  • It further is preferred if that consistency builder is selected from pharmaceutically acceptable hydrophilic polymers, such as partially etherified cellulose derivatives, comprising carboxymethyl-, hydroxyethyl-, hydroxypropyl-, hydroxypropylmethyl- or methyl-cellulose; completely synthetic hydrophilic polymers comprising polyacrylates, polymethacrylates, poly(hydroxyethyl)-, poly(hydroxypropyl)-, poly(hydroxypropylmethyl)methacrylate, polyacrylonitrile, methallyl-sulphonate, polyethylenes, polyoxiethylenes, polyethylene glycols, polyethylene glycol-lactide, polyethylene glycol-diacrylate, polyvinylpyrrolidone, polyvinyl alcohols, poly(propylmethacrylamide), poly(propylene fumarate-co-ethylene glycol), poloxamers, polyaspartamide, (hydrazine cross-linked) hyaluronic acid, silicone; natural gums comprising alginates, carrageenan, guar-gum, gelatine, tragacanth, (amidated) pectin, xanthan, chitosan collagen, agarose; mixtures and further derivatives or co-polymers thereof and/or other pharmaceutically, or at least biologically, acceptable polymers. In especially, the polymer weight fractions preferably are in the range between 0.05% and 10%, more preferably are in the range between 0.1% and 5%, even more preferably are in the range between 0.25% and 3.5% and most preferably are in the range between 0.5% and 2%.
  • It has been found that viscosity is best suited if the consistency builder is added in an amount that increases the formulation viscosity above that of the non-thickened corresponding formulation, preferably to up to 1 Nm/s and even more preferably to up to 0.2 Nm/s.
  • In another aspect of the invention, a formulation comprising molecular arrangements capable of penetrating pores in a barrier, owing to penetrant adaptability, despite the fact that the average diameter of said pores is smaller than the average penetrant diameter, provided that the penetrants can transport agents or else enable agent permeation through the pores after penetrants have entered pores, the agents associated with said penetrants being glucocorticoids or mineralocorticosteroids (corticosteroids), is characterised in that the relative content of corticosteroids is above 0.1 weight-%, relative to total dry mass of the formulation. It then is preferred if at least one consistency builder and/or at least one anti-oxidant and/or at least one microbicide, as described herein, is added to the formulation.
  • That corticosteroid is preferably selected from alclonetasone dipropionate, amcinonide, beclomethasone dipropionate, betamethasone, betamethasone 17-valerate, betamethasone 17,21-divalerate, betamethasone 21-acetate, betamethasone 21-buytrate, betamethasone 21-propionate, betamethasone 21-valerate, betamethasone benzoate, betamethasone dipropionate, betamethasone valerate, budesonide, clobetasol propionate, clobetasone butyrate, cortexolone, corticosterone, cortisone, cortisone 17-acetate, 21-deoxybetamethasone, 21-deoxybetamethasone 17-propionate, deoxycorticosterone, desonide, desoxymethasone, dexamethasone, diflorasone diacetate, diflucortolone valerate, fluclorolone acetonide, flumethasone pivalate, fluocinolone acetonide, fluocinonide, fluocortin butyl, fluocortolone, 9-alpha-fluorocortisone, 9-alpha-fluorohydrocortisone, 9-alpha-fluoroprednisolone, fluprednidene acetate, flurandrenolone, halcinonide, hydrocortisone, hydrocortisone 17-acetate, hydrocortisone 17-butyrate, hydrocortisone 17-propionate, hydro cortisone 17-valerate, hydrocortisone 21-acetate, hydrocortisone 21-butyrate, hydrocortisone 21-propionate, hydrocortisone 21-valerate, 17-alpha-hydroxyprogesterone, methylprednisolone acetate, mometasone furoate, prednisolone, prednisone, prednisone 17-acetate, prednisone 17-valerate, progesterone, triamcinolone, triamcinolone acetonide.
  • In a specific embodiment of the invention the penetrants are suspended or dispersed in a polar liquid in the form of fluid droplets surrounded by a membrane-like coating of one or several layers, said coating comprising at least two kinds or forms of amphiphilic substances with a tendency to aggregate, provided that said at least two substances differ by at least a factor of 10 in solubility in said liquid or else that said substances when in the form of homo-aggregates, for the more soluble substance, or of hetero-aggregates, for any combination of both said substances, have a preferred average diameter smaller than the diameter of the homo-aggregates containing merely the less soluble substance; or else provided that the presence of the more soluble substance lowers the average elastic energy of the membrane-like coating in the vicinity of thermal energy.
  • It then is preferred that the more soluble substance tends to solubilise the droplet and the content of such substance is up to 99 mol-% of solubilising concentration or else corresponds to up to 99 mol-% of the saturating concentration in the unsolubilised droplet, whichever is higher. It can be an advantage if the content of the more soluble substance is below 50%, especially below 40% and most preferably below 30%, of the respective solubilising concentration of said substance. It also is often advantageous if the content of the more soluble substance is below 80%, preferably below 65% and most preferably below 50% of the saturation concentration of said substance in the droplet.
  • In many highly preferred embodiments of the invention, the less soluble amongst the aggregating substances is a lipid or lipid-like material, especially a polar lipid, whereas the substance which is more soluble in the suspending liquid and which increases the droplet adaptability belongs to the class of surfactants or else has surfactant-like properties. A specific embodiment of the invention is prepared from a lipid or lipid-like material (which may be a lipid or a lipoid from a biological source or a corresponding synthetic lipid or any of its modifications), said lipid preferably belonging to the class of pure phospholipids corresponding to the general formula
    Figure US20070184114A1-20070809-C00001

    where R1 and R2 is an aliphatic chain, typically a C10-20-acyl, or -alkyl or partly unsaturated fatty acid residue, in particular an oleoyl-, palmitoeloyl-, elaidoyl-, linoleyl-, linolenyl-, linolenoyl-, arachidoyl-, vaccinyl-, lauroyl-, myristoyl-, palmitoyl-, or stearoyl chain; and where R3 is hydrogen, 2-trimethylamino-1-ethyl, 2-amino-1-ethyl, C1-4-alkyl, C1-5-alkyl substituted with carboxy, C2-5-alkyl substituted with hydroxy, C2-5-alkyl substituted with carboxy and hydroxy, or C2-5-alkyl substituted with carboxy and amino, inositol, sphingosine, or salts of said substances, said lipid comprising also glycerides, isoprenoid lipids, steroids, sterines or sterols, of sulphur- or carbohydrate-containing lipids, or any other bilayer forming lipids, in particular half-protonated fluid fatty acids. Preferably, said lipid is selected from the group comprising phosphatidylcholines, phosphatidylethanolamines, phosphatidylglycerols, phosphatidylinositols, phosphatidic acids, phosphatidylserines, sphingomyelins and other sphingophospholipids, glycosphingolipids (including cerebrosides, ceramidepolyhexosides, sulphatides, sphingoplasmalogens), gangliosides and other glycolipids or synthetic lipids, in particular with corresponding sphingosine derivatives, or any other glycolipids, whereby two similar or different chains can be ester-groups-linked to the backbone (as in diacyl and dialkenoyl compound) or be attached to the backbone with ether bonds, as in dialkyl-lipids.
  • It is preferred if the surfactant or surfactant-like material is a nonionic, a zwitterionic, an anionic or a cationic surfactant, especially a fatty-acid or -alcohol, an alkyl-tri/di/methyl-ammonium salt, an alkylsulphate salt, a monovalent salt of cholate, deoxycholate, glycocholate, glycodeoxycholate, taurodeoxycholate, taurocholate, etc., an acyl- or alkanoyl-dimethyl-aminoxide, esp. a dodecyl- dimethyl-aminoxide, an alkyl- or alkanoyl-N-methylglucamide, N-alkyl-N,N-dimethylglycine, 3-(acyldimethylammonio)-alkanesulphonate, N-acyl-sulphobetaine, a polyethylene-glycol-octylphenyl ether, esp. a nonaethylene-glycol-octylphenyl ether, a polyethylene-acyl ether, esp. a nonaethylen-dodecyl ether, a polyethylene-glycol-isoacyl ether, esp. a octaethylene-glycol-isotridecyl ether, polyethylene-acyl ether, esp. octaethylenedodecyl ether, polyethylene-glycol-sorbitane-acyl ester, such as polyethylenglykol-20-monolaurate (Tween 20) or polyethylenglykol-20-sorbitan-monooleate (Tween 80), a polyhydroxyethylene-acyl ether, esp. polyhydroxyethylene-lauryl, -myristoyl, -cetylstearyl, or -oleoyl ether, as in polyhydroxyethylene-4 or 6 or 8 or 10 or 12, etc., -lauryl ether (as in Brij series), or in the corresponding ester, e.g. of polyhydroxyethylen-8-stearate (Myrj 45), -laurate or -oleate type, or in polyethoxylated castor oil 40, a sorbitane-monoalkylate (e.g. in Arlacel or Span), esp. sorbitane-monolaurate, an acyl- or alkanoyl-N-methylglucamide, esp. in or decanoyl- or dodecanoyl-N-methylglucamide, an alkyl-sulphate (salt), e.g. in lauryl- or oleoyl-sulphate, sodium deoxycholate, sodium glycodeoxycholate, sodium oleate, sodium taurate, a fatty acid salt, such as sodium elaidate, sodium linoleate, sodium laurate, a lysophospholipid, such as n-octadecylene(=oleoyl)-glycerophosphatidic acid, -phosphorylglycerol, or -phosphorylserine, n-acyl-, e.g. lauryl or oleoyl-glycero-phosphatidic acid, -phosphorylglycerol, or -phosphorylserine, n-tetradecyl- glycero-phosphatidic acid, -phosphorylglycerol, or -phosphorylserine, a corresponding palmitoeloyl-, elaidoyl-, vaccenyl-lysophospholipid or a corresponding short-chain phospholipid, or else a surface-active polypeptide.
  • Penetration rate of agent carriers is often maximized if the average penetrant diameter is chosen to be between 30 nm and 500 nm, preferably between 40 nm and 250 nm, even more preferably between 50 nm and 200 nm and most preferably between 60 nm and 150 nm.
  • Pore penetration rate of agent carriers is often optimized in terms of the relative ratio between penetrant and pore size, if the average diameter of the penetrant is 2 to 25 times bigger than the average diameter of the pores in the barrier, preferably between 2.25 and 15 times bigger, even more preferably between 2.5 and 8 times bigger and most preferably between 3 and 6 times bigger than said average pore diameter.
  • Specific preferred embodiments of the invention are characterized by the fact, that the dry weight of all carrier droplets in a formulation for the use on human or animal skin is 0.01 weight-% (w-%) to 40 w-% of total formulation mass, in particular between 0.1 w-% and 30 w-%, particularly preferably between 0.5 w-% and 20 w-%, and most preferably between 1 w-% and 10 w-%.
  • If the formulation is to be applied on human or animal mucosa the dry weight of all carrier droplets in a formulation is advantageously chosen to be in the range between 0.0001 w-% and 30 w-% of total formulation mass.
  • For the preparation of a formulation it is preferred if the pH of the carrier suspension is between 4 and 10, preferably between 5 and 9, and even more often up to 8.5, as required in order to maximise the stability of formulation, depending on the pH of the carrier suspension.
  • A method for the preparation of a formulation for non-invasive application in vivo according to the invention comprises the use of at least one amphiphilic substance, at least one polar fluid, at least one edge-active substance or surfactant, at least one corticosteroid in an amount of more than 0.1 w-% based on total dry mass of the formulation and, in case, other customary ingredients, which together form said formulation.
  • It then is preferred if at least one edge-active substance or surfactant, at least one amphiphilic substance, at least one hydrophilic fluid and the agent are dissolved to form a solution and, if required, are mixed separately, the resulting (partial) mixtures or solutions then being combined to subsequently induce, preferably by action of mechanical energy, such as shaking, stirring, vibrating, homogenising, ultrasonication, shear, freezing and thawing, or filtration using convenient driving pressure, the formation of penetrants that associate with and/or incorporate the agent.
  • It is advantageous if said amphiphilic substances are either used as such, or dissolved in a physiologically compatible polar fluid, which may be water or miscible with water, or in a solvation-mediating agent, together with a polar solution. It further is preferred that said amphiphilic substances are dissolved in highly volatile alcohols, especially ethanol, or in other pharmaceutically acceptable organic solvents, which are then removed, esp. by evaporation, prior to making the final preparation.
  • It may also be advantageous if said polar solution contains at least one edge-active substance or surfactant.
  • For the preparation of a formulation according to the invention it further is preferred, that the formation of said penetrants is induced by the addition of the required substances into a fluid phase, evaporation from a reverse phase, by injection or dialysis, if necessary under the influence of mechanical stress, such as shaking, stirring, especially high velocity stirring, vibrating, homogenising, ultrasonication, shearing, freezing and thawing, or filtration using convenient, especially low (1 MPa) or intermediate (up to 10 MPa), driving pressure.
  • It then is convenient if the formation of said penetrants is induced by filtration, the filtering material having pores sizes between 0.01 μm and 0.8 μm, preferably between 0.02 μm and 0.3 μm, and most preferably between 0.05 μm and 0.15 μm, whereby several filters may be used sequentially or in parallel.
  • Furthermore, it is preferred if said agents and penetrants are made to associate, at least partly, after the formation of said penetrants, e.g. after injecting a solution of the drug in a pharmaceutically acceptable fluid, such as ethanol, 1-and 2-propanol, benzyl alcohol, propylene glycol, polyethylene glycol (molecular weight: 200-400 D) or glycerol into the suspending medium, said penetrants being formed previously, using the corresponding or some other suitable manufacturing method, or simultaneously with the drug injection, if required using a co-solution of the drug and, at least some, penetrant ingredients.
  • It may be advantageous if the penetrants, with which the agent molecules are associated and/or into which the agent molecules are incorporated, are prepared just before the application of the formulation, if convenient from a suitable concentrate or a lyophylisate.
  • It is preferred if the content of corticosteroids is between 0.1 relative weight % (rw-%) and 20 rw-%, more preferably between 0.25 rw-% and 10 rw-% and even more preferably between 0.5 rw-% and 5 rw-% with regard to total penetrant dry mass.
  • It then is preferred if said corticosteroid is triamcinolone or one of its derivatives, such as acetonide, the relative content thereof is below 2 w-%, relative to total dry mass of the drug-loaded carriers, even more preferably is below 1 w-% and most typically is below 0.5 w-%.
  • It also is preferred if the corticosteroid is hydrocortisone or one of its derivatives, the relative content thereof is below 20 w-%, relative to total dry mass of the drug-loaded carriers, even more preferably is below 12.5 w-% and most typically is below 5 w-%.
  • Further, it is preferred if said corticosteroid is dexamethasone or one of its derivatives, the relative content thereof is below 15 w-%, relative to total dry mass of the drug-loaded carriers, even more preferably is below 10 w-% and most typically is below 5 w-%.
  • It equally is preferred if said corticosteroid is clobetasol or one of its derivatives, such as propionate, the relative content thereof is below 15 w-%, relative to total dry mass of the drug-loaded carriers, even more preferably is below 10 w-% and most typically is below 5w-%.
  • It is a preferred feature of the invention that the content of said corticosteroid is below the saturation maximum, defined as the point at which the corticosteroid begins to crystallise in or outside the carrier, such maximum depending on interactions between the amphiphilic molecules comprising the carrier and the agent molecules and frequently being reflected in the membrane/or oil/water partition coefficient difference for the main carrier component and said corticosteroid or else relates to the mismatch in molecular size between the carrier and corticosteroid molecules, the drugs with a lower compatibility typically having lower saturation values.
  • It may be advantageous if in order to speed up drug action a permeation enhancer is added, which preferablyis selected from 1-acyl-azacycloheptan-2-ones (azones), 1-acyl-glucosides, 1-acyl-polyoxyethylenes, 1-acyl-saccharides, 2-n-acyl-cyclohexanones, 2-n-acyl-1,3-dioxolanes (SEPA), 1,2,3-triacyl-glycerols, 1-alkanols, 1-alkanoic acids, 1-alkyl-acetates, 1-alkyl-amines, 1-alkyl-n-alkyl-polyoxyethylenes, 1-alkyl-alkylates, n-alkyl-beta-D-thioglucosides, 1-alkyl-glycerides, 1-alkyl-propyleneglycols, 1-alkyl-polyoxyethylenes, (1-alkyl-)2-pyrrolidones, alkyl-acetoacetates, alkylene-glycols, alkyl-methyl-sulphoxides (alkyl-DMSO), alkyl-propionates, alkyl-sulphates, diacyl-succinates, diacyl-N,N-dimethylaminoacetates (DDAA), diacyl-N,N-dimethylaminoisopropionates (DDAIP), phenyl-alkyl-amines.
  • This addition of permeation enhancers is not comparable to the addition of permeation enhancers as already effected in classical galenic preparations, such as ointments and lotions, as in the art, the permeation enhancers are solely added for the purpose of fluidization of the skin. In the present case the permeation enhancers are added to speed up drug action which is to speed up distribution between agent carrier and surroundings. This content of permeation enhancer is not suited to substantially fluidize skin in order to increase the pore penetration rate of agent carriers and therefore is inherently different from prior art.
  • It then is preferred if the bulk concentration range of the preferably used enhancers is up to and around 5% for 1-capryl-propylene glycol, 6-10% for 1-[2-(decylthio)ethyl]azacyclopentan-2-one (=HPE-101), <10% for 1-dodecanol, <10% for 1-dodecyl-azacycloheptan-2-one (=azone), in the range of 10% for 2-n-nonyl-1,3-dioxolane (SEPA), <10% for 2-n-octylcyclohexanone, up to, and preferably around, up to 20% for DMSO, up to, and preferably between 5% and 40% for ethanol, in the range of 10% or higher for ethylene glycol, up to 30% for ethyl acetate, 5-50% for glycerol, up to 75% for isopropanol, 1-20% for isopropyl myristate, between 1 and, preferably, 20% for oleic acid and oleyl-alcohol, of the order of around 1% for oleyl-polyoxyethylene-ether, at least 10% for propylene glycol.
  • The caveat pertaining to these ranges is that the relative and absolute potency of various skin permeation enhancers differs, which makes absolute comparisons difficult. In principle, it is the concentration of an enhancer in the skin which determines the enhancement success. However, it is the nominal enhancer concentration on the skin which typically is considered or is quoted in the literature. The two values often differ by several orders of magnitude, are sensitive to drug-enhancer association, and also may vary with the application conditions. Too small enhancer reservoir on the surface, for example, in the case of a fast enhancer diffusion across the skin or evaporation, leads to substance depletion. It also changes the final system properties. It is a preferred feature of the invention that said corticosteroid is added in an amount which enables the formulation to be applied corresponding to an area dose, as expressed by the total dry mass of penetrant applied per unit area, of between 0.1 mg cm−2 and 15 mg cm−2, more preferably between 0.5 mg cm−2 and 10 mg cm−2, particularly preferably between 0.75 mg cm−2 and 5 mg cm−2 and most preferably between 1 mg cm−2 and 2.5 mg cm−2, if said corticosteroid is desired to exert a therapeutic effect in the deep subcutaneous, e.g. muscle or joints, tissue or else in the remote tissues, including the whole body.
  • It is another preferred feature of the invention that said corticosteroid is added in an amount which enables the formulation to be applied with an area dose, as expressed by the total dry mass of penetrant applied per unit area, of 1 between μg cm−2 and 250 μg cm−2, more preferably between 2.5 and 100 μg cm−2, even more preferably between 5 μg cm−2 and 50 μg cm−2 and most preferably between 7.5 μg cm−2 and 20 μg cm−2, if said corticosteroid is desired to exert a mainly local, that is, superficial, rather than systemic therapeutic effect.
  • It is preferred if consistency and, if necessary other characteristics of the formulation are appropriately selected to enable spraying, smearing, rolling or sponging of the formulation on the application area in particular by using a sprayer, spender, roller or sponge, as appropriate.
  • It is another preferred feature of the invention that for non-invasive application of corticosteroids by means of said penetrants according to the invention the area dose, as expressed by the total dry mass of penetrant applied per unit area, is selected to be between 0.1 mg cm−2 and 15 mg cm−2, preferably between 0.5 mg cm−2 and 10 mg cm−2, particularly preferably between 0.75 mg cm−2 and 5 mg cm−2 and most preferably between 1 mg cm−2 and 2.5 mg cm−2, if said corticosteroid is desired to exert a substantial therapeutic effect in the deep subcutaneous, e.g. muscle or joints, tissue or else in the remote tissues, including the whole body.
  • It otherwise is preferred if for non-invasive application of corticosteroids by means of penetrants according to the invention the area-dose, as expressed by the total dry mass of penetrants applied per unit area, is chosen to be between 1 μg cm−2 and 250 μg cm−2, preferably between 2.5 μg cm−2 and 100 μg cm−2, more preferably between 5 μg cm−2 and 50 μg cm−2 and most preferably between 7.5 μg cm−2 and 20 μg cm−2, to achieve mainly local, that is, superficial, rather than systemic effect of the drug.
  • It may be advantageous if non-invasive application of corticosteroids associated with or encapsulated into penetrants according to the invention is effected by spraying, smearing, rolling or sponging on the application area in particular by using a sprayer, spender, roller or sponge, as appropriate.
  • A preferred use of a formulation in accordance with the invention is for the treatment of inflammatory disease, dermatosis, kidney or liver failure, adrenal insufficiency, aspiration syndrome, Behcet syndrome, bites and stings, blood disorders, such as cold-haemagglutinin disease, haemolytic anemia, hypereosinophilia, hypoplastic anemia, macroglobulinaemia, trombocytopenic purpura, furthermore, for the management of bone disorders, cerebral oedema, Cogan's syndrome, congenital adrenal hyperplasia, connective tissue disorders, such as lichen, lupus erythematosus, polymyalgia rheumatica, polymyositis and dermatomyositis, epilepsy, eye disorders, such as cataracts, Graves' ophthalmopathy, haemangioma, herpes infections, neuropathies, retinal vasculitis, scleritis, for some gastro-intestinal disorders, such as inflammatory bowel disease, nausea and oesophageal damage, for hypercalcaemia, infections, e.g. of the eye (as in infections mononucleosis), for Kawasaki disease, myasthenia gravis, various pain syndromes, such as postherpetic neuralgia, for polyneuropathies, pancreatitis, in respiratory disorders, such as asthma, for the management of rheumatoid disease and osteoarthritis, rhinitis, sarcoidosis, skin diseases, such as alopecia, eczema, erythema multiforme, lichen, pemphigus and pemphigoid, psoriasis, pyoderma gangrenosum, urticaria, in case of thyroid and vascular disorders.
  • The following examples and results of in-vitro and in-vivo studies shown in attached figures should illustrate the scope of the invention without setting or delineating its limits.
  • EXAMPLES 1-4
  • Composition:
    73.2 mg, 64.5 mg, 54.8 mg, Soy bean phosphatidylcholine (SPC)
    37.7 mg
    26.8 mg, 35.5 mg, 45.2 mg, Polysorbate (Tween 80)
    62.3 mg
    1 mg/g Triamcinolone acetonide
    899 mg Phosphate buffer (10 mM, pH 6.5)

    Preparation:
  • Various SPC and triamcinolone acetonide amounts (as specified) are dissolved in 50 mL chloroform and 50 mL methanol. The solvent, which is kept warm (approx. 40 degrees Celsius), is evaporated under a stream of nitrogen and the residue is dried in vacuum at room temperature. Tween 80 in the specified quantity and phosphate buffer is added to the lipid film and the resulting crude suspension is sonicated to prepare smaller mixed lipid vesicles. The resulting suspension should be opalescent and slightly yellow, which requires up to a few minutes of sonication, and is stable for at least 1 day. The test sample is used within 24 h after the preparation.
  • Biological and/or Characterisation Experiments:
  • with this and all following suspensions were done as described further below.
  • EXAMPLES 5a-5b
  • Composition:
    37.74 mg Soy bean phosphatidylcholine (SPC)
    62.26 mg Tween 80
    0.4 mg Triamcinolone Acetonide
    0, 26.25 mg Benzyl alcohol
    4.47 g Phosphate buffer 50 mM pH 6.5
    0.3 mg Probucol
    0.3 mg Desferal

    Preparation:
  • SPC, probucol and triamcinolone acetonide are dissolved in a chloroform/methanol mixture. Dry lipid mixture is prepared as described for example 1. Desferal, Tween 80, and 894.23 mg buffer is added to the dry lipid. The resulting suspension is stirred over night. After adding, if so chosen, 26.25 mg benzyl alcohol in 3.58 g buffer to the suspension, the mixture is extruded through a 200 nm polycarbonate membrane and then through a 50 nm membrane using sufficient excess pressure to give an acceptable flow rate. The resulting particle diameter is below 150 nm
  • EXAMPLE 6
  • Composition:
    37.74 mg SPC
    62.26 mg Tween 80
    35 mg Ethanol
    0.4 mg Triamcinolone acetonide
    26.25 mg Benzyl alcohol
    4.47 g Phosphate buffer (50 mM pH 6.5)
    0.3 mg Probucol
    0.3 mg Desferal

    Preparation:
  • SPC, probucol and triamcinolone Acetonide are dissolved in ethanol. Desferal, Tween 80, 5.25 mg benzyl alcohol and 894.23 mg buffer is added. The resulting suspension is stirred over night. The following day a solution of 21 mg benzyl alcohol in 3.58 g buffer is added to the suspension. The suspension is extruded, first, through a 200 nm pore polycarbonate membrane and then through a 50 nm membrane. This results in particle radius around 60 nm.
  • Analysis of formulation stability by means of HPLS suggests that the presence of probucol and desferal is advantageous to the chemical stability of suspension.
  • EXAMPLES 7-14
  • Composition:
    88.1 g, 87.4 g, 86.6 g, 85.75 g Soy bean phosphatidylcholine (SPC)
    11.9 g, 12.6 g, 13.4 g, 14.25 g Sodium cholate (NaChol)
    80 g Ethanol
    0.5 g Triamcinolone acetonide
    1000 g Phosphate buffer (pH 7.1)

    Preparation:
  • SPC and triamcinolone acetonide are dissolved in ethanol, to which NaChol is also added (which dissolves only partially). After the addition of buffer, the resulting turbid, whitish suspension is stirred over night. To bring the vesicles to final size, the suspension is either: extruded through a 200 nm membrane and then through a 100 nm membrane under pressure; 2) processed by a high pressure homogeniser (run in the low pressure range, e.g. at 200 psi) to yield an opalescent final suspension.
  • From the preparation made as described above, two alternative formulations were made by diluting the suspension with a buffer (containing 0.5 V-% benzyl alcohol) to a final total lipid concentration of 5 w-% and 2 w-%, respectively.
  • EXAMPLES 15-49
  • Composition:
    377.4 mg Soy bean phosphatidylcholine (SPC)
    622.6 mg Tween 80
      50 mg Benzyl alcohol
     9000 mg Phosphate buffer 50 mM pH 6.5
    a) 12.5 mg, 25 mg, 50 mg, Bethamethasone
    b) 12.5 mg, 25 mg, 50 mg, Bethamethasone dipropionate
    c) 12.5 mg, 25 mg, 50 mg Bethamethasone 17 valerate
    d) 12.5 mg, 25 mg, 50 mg Clobetasol-17-propionate
    e) 12.5 mg, 25 mg, 50 mg Dexamethasone or
    f) 25 mg, 50 mg, 75 mg, Hydrocortisone
    g) 12.5 mg, 25 mg, 50 mg Prednicarbate
    h) 0.75 mg, 12.5 mg, 25 mg Triamcinolone

    Preparation:
  • SPC and the corticosteroid of choice is dissolved in ethanol. After the addition of buffer, which also contains Tween 80 and benzyl alcohol, the resulting highly turbid suspension is thoroughly mixed for at least 24 h, and more preferably for several days. The suspension is then extruded through a 200 nm membrane, if required several times. The resulting suspension of vesicles, which are still relatively large, tends to sediment with time, however, but can be re-homogenised easily by swirling-or another gentle mixing method. To get vesicles with smaller final size, and thus a more stable suspension, a final extrusion through a 100 nm membrane is useful. (Vesicles with the highest given drug concentration may contain some drug in the suspension, perhaps in the form of vesicle coated drug crystals.)
  • EXAMPLE 50
  • Composition:
    347 mg Soy bean phosphatidylcholine (SPC)
    623 mg Tween 80
    30 mg Sodium dodecyl sulphate (SDS)
    50 mg Benzyl alcohol
    9000 mg Phosphate buffer 50 mM pH 6.5
    25 mg Clobetasol-17-propionate

    Preparation:
  • Corticosteroid suspension was prepared as described in previous examples, except in that SDS was added dissolved in buffer to act as drug distribution promotor in the target organ.
  • Formulation containing SDS acts significantly more rapidly in human skin blanching assay than the SDS-free formulation.
  • Hereinfurther are described some preclinical experiences with the hydro-lotion-like formulations based on highly adaptable agent carriers, i.e. highly adaptable and flexible lipid vesicles (Transfersomes™; cf. above-cited references) of several corticosteroids in vitro and in vivo. These novel, carrier-based formulations are shown to give rise to the desired drug concentrations in the skin after a single application of the agent in Transfersomes. Depending on amount of carrier used, a localized (intracutaneous) or a local and systemic (whole body) delivery is possible.
  • The new corticosteroid delivery concept promises to lower the danger of adverse side effects of the topical therapy with such medication. This is possible due to dose-lowering and a different drug delivery mechanism: Corticosteroids in Transfersomes can not penetrate directly into the blood vessels, owing to the prohibitively large size of the carriers. Such drugs are thus confined to the intercellular space, where they can exert their desired biological function. (Only when they are applied in ample amounts such therapeutics are distributed throughout the body, first via the lymphatic and then through blood circulation.)
  • Data measured in animals and humans suggest that several widely used corticosteroids can be nearly prevented from reaching the blood if they are placed on the skin in a suspension of Transfersomes. One can argue that this phenomenon relies on the extremely high deformability of transfersome membranes, which permits the drug carriers to pass the skin permeability barrier. The good control over this penetration process and the exclusion of intravasation enables the restriction of the biological effects of transfersomal corticosteroids nearly exclusively to the treated skin. The different vaso-constriction induced by corticosteroids in creams/lotions or Transfersomes indirectly support this conclusion. The use of highly deformable carriers increases the potency of corticosteroids up to one order of magnitude in relation to previous, commercial formulations. This fact also improves the final drug safety. (The area-dose needed for the Transfersomes-mediated therapeutic success of dexamethasone or triamcinolone acetonide in the treated skin surface must reach 1.5 m2 before the total applied drug amount matches that of native hydrocortisone in the blood.)
  • In Vitro Penetration Studies
  • The differential penetration capability of various drug and drug-carrier formulations through an artificial transport barrier clearly demonstrates the relative advantage of ultradeformable Transfersomes in comparison with, for example, standard liposomes. While the latter are nearly completely incapable of crossing such an artificial ‘skin barrier’, Transfersomes pass through the fine openings in such a barrier essentially unhindered. The following table illustrates this behavior.
    TABLE 1
    Capability (relative to water) of the corticosteroid-loaded
    Transfersomes, liposomes and micelles to penetrate through
    the pores 3-4 times smaller than the penetrant size under
    the influence of hydrostatic pressure. a)
    Low pressure High pressure
    Formulation (0.2 MPa) (0.9 MPa)
    Micelles 1.1 ± 0.1 1.1 ± 0.1  
    Liposomes ≦0.0001 ≦0.001
    Transfersomes ≦0.001 1 ± 0.1
    Liposomes with ≦0.0001 ≦0.001
    hydrocortisone
    Transfersomes with ≦0.001 1 ± 0.1
    hydrocortisone
    Transfersomes with ≦0.001 1 ± 0.1
    dexamethasone
    Transfersomes with ≦0.001 1 ± 0.1
    triamcinolone-acetonide

    a) The artificial barrier consisted of a polycarbonate membrane perforated by the pores of 100 nm diameter. Liposomes and Transfersomes had comparable size. The quoted transport efficacy corresponds to the ratio of aggregate-to-water transport rate measured under identical conditions (by HPLC and gravimetry, respectively).
  • When applied onto the intact skin surface, phospholipid suspensions are not detrimental to the skin. On the contrary: certain phospholipid preparations have been reported to improve the hydration (and thus to a minor extent the optical appearance) of the aging skin. Phospholipid suspensions are also non-irritating to the skin, at least up to the degree of 30% degradation.
  • Corticosteroid preparations on the basis of transfersomes will normally be used in a quantity (around 100 mg per 2 days) that will contain lipid amounts comparable to those used parenterally (≦75 mg/injection) or orally (≦150 mg/day). The recommended daily dose of transfersomal corticosteroids for human use will be appreciably lower (≦25 mg), except in the case of hydrocortisone, where a somewhat higher dosage might be required for a whole body therapy.
  • Total phospholipid amount to be placed on the skin in the form of transfersomes-based corticosteroid formulations will always be less than 0.5 g/day. It is also less than 10% of the natural variability of phosphatidylcholine concentration in the plasma of an average, healthy person. In light of these data given below, one can conclude that the corticosteroidal dermatics based on transfersomes from the carrier point of view will be an extremely safe product.
  • From the agent point of view, a maximum corticosteroid amount (1 mg/day for dexamethasone or triamcinolone-acetonide and below 20 mg for hydrocortisone) comparable to that produced in the body (12 mg to 30 mg of hydrocortisone per day) will be applied topically. The area dose will normally be between 0.1 μg cm−2 and 1 μg cm−2, for the high and low potency drugs, respectively. Only a tiny fraction of the epicutaneously drug is likely to appear in the circulation, however, as can be seen from the following table.
    TABLE 2
    The ratio of corticosteroid concentration in the blood and in the
    ‘inner skin’ of mice (measured) and humans (calculated).
    Hydro- Hydro- Dexa- Dexa- Triamcinolone- Triamcinolone-
    Dose cortisone cortisone methasone methasone acetonide acetonide
    (μg/cm−2) (mouse) (human) (mouse) (human) (mouse) (human)
    0.5 0 0 0 0
    1.3 0.02 0.00007
    4.9 0.012 0.000004 0.1 0.00003
    13.2 0.04 0.00011
    20.6 0.03 0.0001
    49.5 0.015 0.000005 0.25 0.00009
  • It can, therefore, be anticipated that corticosteroids based on transfersomes will cause less side effects, if any, than the currently available commercial formulations of such drugs. This, on the one hand, is due to the more favorable bio-distribution of the drugs from the transfersomes, which is concentrated to the tissue to be treated. On the other hand, drugs from the carriers are likely to be taken in relatively higher proportions by the strongly proliferating cells, which are one of the chief natural targets for the corticosteroid therapy. (It is even possible that very low doses of transfersomal corticosteroids will completely eliminate the problem of skin atrophy after the repeated use of such therapeutics.)
  • Even more relevant, for the assessment of practical values of transfersomal corticosteroids, are the results obtained in validated animal trials, which are described in the following section.
  • Preclinical Studies
  • All substances used in this study were of pharmaceutic quality. Soybean phosphatidylcholine (SPC) was purchased from Lipoid KG (Ludwigshafen, Germany) or Nattermann Phospholipids—Rhone-Poulenc Rorer (Köln, Germany) and was more than 95% pure. The remaining components, which are described in detail in the above cited European patent, were from Henkel (Düsseldorf, Germany) or CPC (Hamburg, Germany). The active ingredients (dexamethasone, hydrocortisone, triamcinolone-acetonide) were purchased from Synopharm (Hamburg, Germany). The microbicides, chellators and antioxidants were from Ciba-Geigy (Basel, Switzerland) or Synopharm. The bidistilled water in injectable quality was purchased from the local pharmacy. The commercial drug formulations from the local pharmacy were used for comparison (hydrocortisone: Hydrocortisone-Wolff (Wolff, Bielefeld); dexamethasone: Anemul (Pharmasal, Gräfelfing); triamcinolon-acetonide: Volon A Lotio N (Squibb-Hayden, München) and Delphicort-cream (Lederle, Wolfratshausen)).
  • Drug in the Carrier (Transfersome) Suspensions. The formulations used in the biodistribution studies were labelled with the tritiurated corticosteroids purchased from Amersham or ICN. Preparation of the formulations for the use in animals was done by dissolving all lipoids in methanol/chloroform (1/1 v/v) in the appropriate amounts and preparing a dry mixed lipid film under vacuum (≦10 Pa; ≧12 h). The use of potentially harmful organic solvents or drying was entirely avoided in the manufacturing of human medications.
  • Formulations contained between 0.01 w-% and 0.5 w-% specified corticosteroid per mL of carrier suspension. The latter consisted chiefly of phosphatidylcholine (SPC) in a final concentration between 0.5 w-% and 5 w-%. This lipid was was taken up in a buffer and homogenized (for animal experiments: by sonication with a titanium micro-tip, Heat Systems W 380, USA, 30 min, 4° C.; for human therapeutics by other mechanical means). At least one of the carrier components was characterized by its membrane solubilizing capacity, as is required by the basic rationale of Transfersome design and above-cited patent applications of the applicant. Such a embrane-affecting substance was always incorporated into the carriers in the sub-lytic concentration. This ensured the high carrier deformability without compromising the integrity of transfersome vesicles, since both is necessary for the high efficacy of drug carrier transport across the stratum corneum. The final vesicle size was determined with the photon correlation spectroscopy (90°, ALV-5000 ALV-Laser Vertriebsgesellschaft, Langen, Germany) and was typically between 100 nm and 200 nm. For experimental use, lipid suspension was diluted when appropriate. More detailed description and characterization data will be given separately.
  • In vivo Experiments mainly involved 8-12 weeks old NMRI mice which were kept under standard laboratory conditions (3-5 per suspending cage; standard chew and water ad libitum; 12 h light/dark regime). Stressful or painful manipulations were always carried out under general injection anesthesia.
  • Biodistribution. The hair at the chosen skin site was trimmed with a pair of scissors to the length of ≦2 mm one day before experimentation. The precise application site on the upper back was marked and the appropriate amount (0.5 μL to 25 μL) and drug formulation was applied with a micro-pipette on the skin. After uniform distribution with the side of the same pipette tip, the application was left to dry.
  • Blood samples (20 μL) were taken from the tail end with a glass capillary. After 8 hours the animals were killed by heart puncture and the treated skin area was undermined and carefully excised. The outermost layers of the stratum comeum were collected by five tape-strippings. Subsequently, the residual skin tissue and other organ samples were prepared, destained and used for radioactivity counting. For the experiments with porcine skin, 20×30 cm2 of full thickness organ was excised and fixed on a wet tissue. Several test areas of 1 cm2 were then marked and treated further as in vivo.
  • Biological Action in mice was most often tested by measuring the suppression of a chemically induced edema by the topically administered corticosteroids. For this purpose, the test animals were first anesthetized with an intraperitoneal injection of 10 μL g−1 body weight of a mixture containing 6 mL 0.9% NaCl, 1 mL Ketavet 100 (Parke-Davis, Berlin, FRG), and 0.25 mL Rompun (Bayer, Leverkusen, Germany). The appropriate amount of drug formulation was smeared over the inner side of one ear and left to dry out. When so stated, the ear was wiped free of the superficial formulation with a cotton swab. At a given time the test mouse was anesthetized and arachidonic acid in ethanol (1/2 V/V, 10\muL) was applied to the same ear area. Change in the mouse ear edema (relatively to that of the untreated but challenged ear) was determined, either by measuring the ear thickness with a micro-caliper (our method) or by weighing the ear volume of the killed mouse (original procedure). Both these assays deliver similar results. All values are the means of at least 3 independently measured values and bars give standard deviation of their mean.
  • Human Studies
  • According to the scientific literature, it is customary to test the biological potency of different corticosteroid preparations in humans by a so-called ‘skin blanching’ assay. Such a test is not as adequate for the investigation of carrier-based corticosteroids, as it is for the testing of the corresponding drug solutions for the reasons given below. This notwithstanding, the topical vaso-constriction test was used to compare the kinetics of corticosteroid action on the rodent and human skin.
  • In a pilot trial with three human volunteers, the test formulations were applied to one arm at different doses in parallel rows. By using a high precision micropipette, individual areas of 1 cm2 were covered. The vaso-constriction over each such skin domain was then determined by visual inspection (at least once by an independent observer who was unaware of the drug application pattern) and the skin blanching score was identified with the number of well defined square corners or edges.
  • Human skin was shown to respond similarly to the topical administration of corticosteroids in Transfersomes when compared to murine skin: after approximately 7 h the vaso-constriction (skin blanching) effect reaches 50% of its maximum value and saturates at t≧8 h. A high biological activity is observed for at least 32 h, with final decay to 50% level observed between 36 h and 48 h, provided the administered drug dose is around 1 μg cm−2 (FIG. 4).
  • Similar evolution of skin blanching is observed after the topical administration of triamcinolone-acetonide in a commercial cream, but not before the drug dose has exceeded 10 μg cm−2. Skin palor in the early phase (8≦t/h≦16) after cream application is deeper (whiter) and appears faster than in the case of the cortico-Transfersomes-mediated vaso-constriction. In our opinion, this is due to the restricted ability of transfersome-associated drugs to get into the blood capillaries. This phenomenon is not encountered with the drugs in commercial formulations, which allow diffusiong through and beyond the skin in monomeric (or at least dissociated) form. This explains the faster onset of (the desired) edema-suppression action and the retardation of (the rather undesired) vaso-constriction, which is an indication of drug spill-over into the blood circulation. (The relatively sluggish appearence of Transfersomes-mediate edema-suppression is also due to the poor responsiveness of one test person, who reacted to the drug in Transfersomes slowly and did not react to the low dose of commercial creme at all, the latter lack of effect not being seen in the time-course of average drug action.)
  • Results
  • from the representative experiments are shown in the attached figures.
  • Upper panel of FIG. 1 illustrates the biological edema-suppression activity of hydrocortisone in commercial cream (open symbols) and in the highly adaptable lipid vesicles, Transfersomes, (closed symbols). Data give mean values measured from 3-4 animals and error bars represent the corresponding standard deviations.
  • Lower panel of FIG. 1 shows dose versus action, as assessed in the local edema-suppression tests, of hydrocortisone in a commercial cream (open symbols) and transfersomal suspension (closed symbols) after 16 h of action. (The maximum in dose vs. action curve is due to the dose dependence of action kinetics (see also FIG. 2).)
  • From FIG. 1 can be seen that the biological effect of hydrocortisone in Transfersomes™-based formulation significantly exceeds that of the more conventional cream-like formulation containing similar drug—the lower is the administered drug-dose per area the higher is the resulting therapeutic advantage. These data suggest that it should be possible to make, and sell with an excellent commercial perspective, the (hydro)lotion-like hydrocortisone formulation containing just 0.1% of the drug. This unprecedented low agent content may reduce the danger of side effects.
  • Upper panel of FIG. 2 illustrates suppression of the arachidonic acid-induced edema by dexamethasone in the commercial cream (open symbols) or Transfersomes (closed symbols) as the function of time after drug administration on the intact murine skin. In both cases the excess drug was wiped-away from the application site 8 h after administration.
  • Lower panel shows effect of changing the dose per area on the dexamethasone-mediated suppression of skin edema in the murine ear model. (Different symbols give results from the different experimental series; for further details see FIG. 1.)
  • As a result from FIG. 2 it is obvious that owing to its higher intrinsic potency, dexamethasone exerts a much stronger biological effect than hydrocortisone when tested locally on the challenged skin. The incorporation of dexamethasone into the ultradeformable agent carriers, Transfersomes further improves this therapeutic advantage. The benefit of using Transfersomes is most dramatic when the excess drug is eliminated from the treated skin site (as in real life). It is expected that drug formulations with merely 0.02% dexamethasone (‘strong’) or with just around 0.005% dexamethasone (‘gentle’) in Transfersomes will be needed for an adequate skin treatment.
  • Upper panel of FIG. 3 illustrates the biological anti-edema activity of triamcinolone-acetonide in commercial lotion (open symbols) or in Transfersomes (closed symbols) in the murine ear model whereas lower panel shows dose vs. action curve for triamcinolone-acetonide in Transfersomes (full symbols, two different preparations and test series), commercial cream (open boxes) or commercial lotion (open circles) applied on the intact murine skin.
  • The biological potency of triamcinolone-acetonide in commercial products is thus 10-times lower than that of the drug in a suspension of Transfersomes. The latter also prolong the duration of therapeutic effect by the same order of magnitude. In comparison with transfersomal dexamethasone, triamcinolone-acetonide in the ultradeformable agent carriers exerts a somewhat stronger but moderately less persistent biological function. The anticipated drug concentration for the commercial formulation on the basis of Transfersomes is between 0.005% and 0.02%.
  • FIG. 4 shows vaso-constriction (blanching-assay) in the intact human skin as a function of time, following an epicutaneous administration of triamcinolone-acetonide in Transfersomes (upper panel) or in commercial cream (lower panel).
  • It thus can be seen that the ‘therapeutic effect’ on the human skin of the high potency corticosteroid applied in Transfersomes is dramatically better than that of the conventional triamcinolone-acetonide cream. A single topical drug administration with Transfersomes ensures good biological function for more than a day, with a dose of 1 μg cm−2. While the commercial cream causes a rather short-term ‘deep blanching’, the Transfersomes-based formulations mediates a more gradual and long-lasting superficial vaso-constriction. This is indicative of reduced drug spill-over into the circulation from the carrier-based formulation (see also the figures on the following two pages).
  • In FIG. 5 different drug penetration profiles in the mammalian skin are shown. The data were measured in vivo in mice (left panel) and ex vivo in porcine skin (right panel). Open symbols represent measurements with a commercial cream and closed symbols with the suspension of dexamethasone-loaded Transfersomes.
  • The use of Transfersomes for carrying corticosteroids in the skin flattens the drug penetration profile in the skin. The relative drug concentration increases in the deeper skin region, when compared to the results achieved with the commercial formulation of similar drug.
  • FIG. 6 illustrates corticosteroid accumulation (retention) in the skin after administration by means of Transfersomes on the intact surface. (∇ and Δ correspond to the inner and outer skin regions and ⋄ gives their sum.)
  • As a result transfersomes bring a relatively high proportion of the epicutaneously administered drug into the viable skin.
  • FIG. 7 illustrates using of Transfersomes for the transcutaneous corticosteroid delivery into the systemic circulation.
  • Choosing suitably optimized agent carriers (good Transfersomes) as well as proper dose per area allows for systemic delivery. Lowering the dose per area increases the relative drug concentration at the site of epicutaneous carrier administration.
  • In FIG. 8 relative efficiency of various triamcinolone acetonide formulations as tested by the murine ear edema assay is shown. Comparison of the biological activity of two different kinds of Transfersomes loaded with this drug (upper panel), a commercial cream (lower panel) and of conventional liposomes (lower panel). The latter two data sets are not statistically significant even at the level of 0.1.
  • In order to maximize the efficacy of intracutaneous drug delivery and to achieve good biological effects it is necessary to employ the specially optimized, proprietary agent carriers, Transfersomes. The replacement of such highly deformable Transfersomes by simple, conventional liposomes produces results that are not better than those obtained by the commercial creams (or lotions).

Claims (50)

1. A formulation comprising molecular arrangements capable of penetrating pores in a barrier, owing to penetrant adaptability, despite the fact that the average diameter of said pores is smaller than the average penetrant diameter, provided that the penetrants can transport agents or else enable agent permeation through the pores after penetrants have entered pores,
characterised in that the formulation comprises at least one consistency builder in an amount that increases the formulation viscosity above that of the non-thickened corresponding formulation to maximally 5 Nm/s so that spreading over, and retention at, the application area is enabled and/or at least one antioxidant in an amount that reduces the increase of oxidation index to less than 100% per 6 months and/or at least one microbicide in an amount that reduces the bacterial count of 1 million germs added per g of total mass of the formulation to less than 100 in the case of aerobic bacteria, to less than 10 in the case of entero-bacteria, and to less than 1 in the case of Pseudomonas aeruginosa or Staphilococcus aureus, after a period of 4 days.
2. Formulation according to claim 1, characterised in that said at least one consistency buildner is added in an amount that increases the formulation viscosity to up to 1 Ns/m2 and more preferably to up to 0.2 Ns/m2.
3. Formulation according to claim 1 or 2,
characterised in that said at least one antioxidant is added in an amount that reduces the increase of oxidation index to less than 100% per 12 months and more preferably to less than 50% per 12 months.
4. Formulation according to any one of the preceding claims,
characterised in that said at least one microbicide is added in an amount that reduces the bacterial count of 1 million germs added per g of total mass of the formulation to less than 100 in the case of aerobic bacteria, to less than 10 in the case of entero-bacteria, and to less than 1 in the case of Pseudomonas aeruginosa or Staphilococcus aureus, after a period of 3 days, and more preferably after a period of 1 day.
5. Formulation according to any of the preceding claims,
characterised in that the consistency builder is selected from pharmaceutically acceptable hydrophilic polymers, such as partially etherified cellulose derivatives, comprising carboxymethyl-, hydroxyethyl-, hydroxypropyl-, hydroxypropylmethyl- or methyl-cellulose; completely synthetic hydrophilic polymers comprising polyacrylates, polymethacrylates, poly(hydroxyethyl)-, poly(hydroxypropyl)-, poly(hydroxypropylmethyl)methacrylate, polyacrylonitrile, methallyl-sulphonate, polyethylenes, polyoxiethylenes, polyethylene glycols, polyethylene glycol-lactide, polyethylene glycol-diacrylate, polyvinylpyrrolidone, polyvinyl alcohols, poly(propylmethacrylamide), poly(propylene fumarate-co-ethylene glycol), poloxamers, polyaspartamide, (hydrazine cross-linked) hyaluronic acid, silicone; natural gums comprising alginates, carrageenan, guar-gum, gelatine, tragacanth, (amidated) pectin, xanthan, chitosan collagen, agarose; mixtures and further derivatives or co-polymers thereof and/or other pharmaceutically, or at least biologically, acceptable polymers.
6. Formulation according to claim 5,
characterised in that the polymer weight fractions are in the range between 0.05% and 10%, more preferably are in the range between 0.1% and 5%, even more preferably are in the range between 0.25% and 3.5% and most preferably are in the range between 0.5% and 2%.
7. Formulation according to any one of the preceding claims,
characterised in that the anti-oxidant is selected from synthetic phenolic antioxidants, such as butylated hydroxyanisol (BHA), butylated hydroxytoluene (BHT) and di-tert-butylphenol (LY178002, LY256548, HWA-131, BF-389, CI-986, PD-127443, E-5119, BI-L-239XX, etc.), tertiary butylhydroquinone (TBHQ), propyl gallate (PG), 1-O-hexyl-2,3,5-trimethylhydroquinone (HTHQ); aromatic amines (diphenylamine, p-alkylthio-o-anisidine, ethylenediamine derivatives, carbazol, tetrahydroindenoindol); phenols and phenolic acids (guaiacol, hydroquinone, vanillin, gallic acids and their esters, protocatechuic acid, quinic acid, syringic acid, ellagic acid, salicylic acid, nordihydroguaiaretic acid (NDGA), eugenol); tocopherols (including tocopherols (alpha, beta, gamma, delta) and their derivatives, such as tocopheryl-acylate (e.g. -acetate, -laurate, myristate, -palmitate, -oleate, -linoleate, etc., or an y other suitable tocopheryl-lipoate), tocopheryl-POE-succinate; trolox and corresponding amide and thiocarboxamide analogues; ascorbic acid and its salts, isoascorbate, (2 or 3 or 6)-o-alkylascorbic acids, ascorbyl esters (e.g. 6-o-lauroyl, myristoyl, palmitoyl-, oleoyl, or linoleoyl-L-ascorbic acid, etc.); non-steroidal anti-inflammatory agents (NSAIDs), such as indomethacine, diclofenac, mefenamic acid, flufenamic acid, phenylbutazone, oxyphenbutazone acetylsalicylic acid, naproxen, diflunisal, ibuprofene, ketoprofene, piroxicam, penicillamine, penicillamine disulphide, primaquine, quinacrine, chloroquine, hydroxychloroquine, azathioprine, phenobarbital, acetaminephen); aminosalicylic acids and derivatives; methotrexate, probucol, antiarrhythmics (amiodarone, aprindine, asocainol), ambroxol, tamoxifene, b-hydroxytamoxifene; calcium antagonists (nifedipine, nisoldipine, nimodipine, nicardipine, nilvadipine), beta-receptor blockers (atenolol, propranolol, nebivolol); sodium bisulphite, sodium metabisulphite, thiourea; chellating agents, such as EDTA, GDTA, desferral; miscellaneous endogenous defence systems, such as transferrin, lactoferrin, ferritin, cearuloplasmin, haptoglobion, haemopexin, albumin, glucose, ubiquinol-10); enzymatic antioxidants, such as superoxide dismutase and metal complexes with a similar activity, including catalase, glutathione peroxidase, and less complex molecules, such as beta-carotene, bilirubin, uric acid; flavonoids (flavones, flavonols, flavonones, flavanonals, chacones, anthocyanins), N-acetylcystein, mesna, glutathione, thiohistidine derivatives, triazoles; tannines, cinnamic acid, hydroxycinnamatic acids and their esters (coumaric acids and esters, caffeic acid and their esters, ferulic acid, (iso-) chlorogenic acid, sinapic acid); spice extracts (e.g. from clove, cinnamon, sage, rosemary, mace, oregano, allspice, nutmeg); camosic acid, camosol, carsolic acid; rosmarinic acid, rosmaridiphenol, gentisic acid, ferulic acid; oat flour extracts, such as avenanthramide 1 and 2; thioethers, dithioethers, sulphoxides, tetralkylthiuram disulphides; phytic acid, steroid derivatives (e.g. U74006F); tryptophan metabolites (e.g. 3-hydroxykynurenine, 3-hydroxyanthranilic acid), and organochalcogenides.
8. Formulation according to claim 7,
characterised in that the concentration of BHA or BHT is between 0.001 and 2 w-%, more preferably is between 0.0025 and 0.2 w-%, and most preferably is between 0.005 and 0.02 w-%, of TBHQ and PG is between 0.001 and 2 w-%, more preferably is between 0.005 and 0.2 w-%, and most preferably is between 0.01 and 0.02 w-%, of tocopherols is between 0.005 and 5 w-%, more preferably is between 0.01 and 0.5 w-%, and most preferably is between 0.05 and 0.075 w-%, of ascorbic acid esters is between 0.001 and 5, more preferably is between 0.005 and 0.5, and most preferably is between 0.01 and 0.15 w-%, of ascorbic acid is between 0.001 and 5, more preferably is between 0.005 and 0.5 w-%, and most preferably is between 0.01 and 0.1 w-%, of sodium bisulphite or sodium metabisulphite is between 0.001 and 5, more preferably is between 0.005 and 0.5 w-%, and most preferably is between 0.01-0.15 w-%, of thiourea is between 0.0001 and 2 w-%, more preferably is between 0.0005 and 0.2, and most preferably is between 0.001-0.01 w-%, most typically 0.005 w-%, of cysteine is between 0.01 and 5, more preferably is between 0.05 and 2 w-%, and most preferably is between 0.1 and 1.0 w-%, most typically 0.5 w-%, of monothioglycerol is between 0.01 and 5 w-%, more preferably is between 0.05 and 2 w-%, and most preferably is between 0.1-1.0 w-%, most typically 0.5 w-%, of NDGA is between 0.0005-2 w-%, more preferably is between 0.001-0.2 w-%, and most preferably is between 0.005-0.02 w-%, most typically 0.01 w-%, of glutathione is between 0.005 and 5 w-%, more preferably is between 0.01 and 0.5 w-%, and most preferably is between 0.05 and 0.2 w-%, most typically 0.1 w-%, of EDTA is between 0.001 and 5 w-%, even more preferably is between 0.005 and 0.5 w-%, and most preferably is between 0.01 and 0.2 w-%, most typically between 0.05 and 0.975 w-%, of citric acid is between 0.001 and 5 w-%, even more preferably is between 0.005 and 3 w-%, and most preferably is between 0.01-0.2, most typically between 0.3 and 2 w-%.
9. Formulation according to any one of the preceding claims,
characterised in that the microbicide is selected from short chain alcohols, comprising ethyl and isopropyl alcohol, chlorbutanol, benzyl alcohol, chlorbenzyl alcohol, dichlorbenzylalcohol, hexachlorophene; phenolic compounds, such as cresol, 4-chloro-m-cresol, p-chloro-m-xylenol, dichlorophene, hexachlorophene, povidon-iodine; parabenes, especially alkyl-parabenes, such as methyl-, ethyl-, propyl-, or butyl-paraben, benzyl paraben; acids, such as sorbic acid, benzoic acid and their salts; quaternary ammonium compounds, such as alkonium salts, e.g. a bromide, benzalkonium salts, such as a chloride or a bromide, cetrimonium salts, e.g. a bromide, phenoalkecinium salts, such as phenododecinium bromide, cetylpyridinium chloride and other salts; mercurial compounds, such as phenylmercuric acetate, borate, or nitrate, thiomersal, chlorhexidine or its gluconate, or any antibiotically active compounds of biological origin, or any mixture thereof.
10. Formulation according to claim 9,
characterised in that the bulk concentration of short chain alcohols in the case of ethyl, propyl, butyl or benzyl alcohol is up to 10 w %, more preferably is up to 5 w-%, and most preferably is in the range between 0.5-3 w-%, in the case of chlorobutanol is in the range between 0.3-0.6 w-%; bulk concentration of parabens, especially in the case of methyl paraben is in the range between 0.05-0.2 w-%, and in the case of propyl paraben is in the range between 0.002-0.02 w-%; bulk concentration of sorbic acid is in the range between 0.05-0.2 w-%, and in the case of benzoic acid is in the range between 0.1-0.5 w-%; bulk concentration of phenols, triclosan, is in the range between 0.1-0.3 w-%, and bulk concentration of chlorhexidine is in the range between 0.01-0.05 w-%.
11. Formulation comprising molecular arrangements capable of penetrating pores in a barrier, owing to penetrant adaptability, despite the fact that the average diameter of said pores is smaller than the average penetrant diameter, provided that the penetrants can transport agents or else enable agent permeation through the pores after said penetrants have entered said pores, the agents associated with said penetrants being glucocorticoids or mineralocorticosteroids (corticosteroids),
characterised in that the relative content of corticosteroids is above 0.1 weight-%, relative to total dry mass of the formulation.
12. Formulation according to claim 11,
characterised in that at least one consistency builder and/or at least one anti-oxidant and/or at least one microbicide according to any one of the claims 1 through to 10 is added to the formulation.
13. Formulation according to claim 11 or 12,
characterised in that the corticosteroid is selected from alclonetasone dipropionate, amcinonide, beclomethasone dipropionate, betamethasone, betamethasone 17-valerate, betamethasone 17,21-divalerate, betarnethasone 21-acetate, betamethasone 21-buytrate, betamethasone 21-propionate, betamethasone 21-valerate, betamethasone benzoate, betamethasone dipropionate, betamethasone valerate, budesonide, clobetasol propionate, clobetasone butyrate, cortexolone, corticosterone, cortisone, cortisone 17-acetate, 21-deoxybetamethasone, 21-deoxybetamethasone 17-propionate, deoxycorticosterone, desonide, desoxymethasone, dexamethasone, diflorasone diacetate, diflucortolone valerate, fluclorolone acetonide, flumethasone pivalate, fluocinolone acetonide, fluocinonide, fluocortin butyl, fluocortolone, 9-alpha-fluorocortisone, 9-alpha-fluorohydrocortisone, 9-alpha-fluoroprednisolone, fluprednidene acetate, flurandrenolone, halcinonide, hydrocortisone, hydrocortisone 17-acetate, hydrocortisone 17-butyrate, hydrocortisone 17-propionate, hydro cortisone 17-valerate, hydrocortisone 21-acetate, hydrocortisone 21-butyrate, hydrocortisone 21-propionate, hydrocortisone 21-valerate, 17-alpha-hydroxyprogesterone, methylprednisolone acetate, mometasone furoate, prednisolone, prednisone, prednisone 17-acetate, prednisone 17-valerate, progesterone, triamcinolone, triamcinolone acetonide.
14. Formulation according to any one of the preceding claims,
characterised in that the penetrants are suspended or dispersed in a polar liquid in the form of fluid droplets surrounded by a membrane-like coating of one or several layers, said coating comprising at least two kinds or forms of amphiphilic substances with a tendency to aggregate, provided that said at least two substances differ by at least a factor of 10 in solubility in said liquid or else that said substances when in the form of homo-aggregates, for the more soluble substance, or of hetero-aggregates, for any combination of both said substances, have a preferred average diameter smaller than the diameter of the homo-aggregates containing merely the less soluble substance; or else provided that the presence of the more soluble substance lowers the average elastic energy of the membrane-like coating in the vicinity of thermal energy.
15. Formulation according to claim 14,
characterised in that the more soluble substance tends to solubilise the droplet and the content of such substance is up to 99 mol-% of solubilising concentration or else corresponds to up to 99 mol-% of the saturating concentration in the unsolubilised droplet, whichever is higher.
16. Formulation according to claim 15,
characterised in that the content of the more soluble substance is below 50%, especially below 40% and most preferably below 30%, of said solubilising concentration of said substance.
17. Formulation according to claim 15,
characterised in that the content of the more soluble substance is below 80%, preferably below 65% and most preferably below 50% of said saturation concentration of said substance in the droplet.
18. Formulation according to any one of claims 14 to 17,
characterised in that the less soluble amongst the aggregating substances is a lipid or lipid-like material, especially a polar lipid, whereas the substance which is more soluble in the suspending liquid and which increases the droplet adaptability belongs to surfactants or else has surfactant-like properties.
19. Formulation according to claim 18,
characterised in that the lipid or lipid-like material is a lipid or a lipoid from a biological source or a corresponding synthetic lipid or any of its modifications, said
Figure US20070184114A1-20070809-C00002
 lipid preferably belonging to the class of pure phospholipids with the chemical formula
where R1 and R2 is an aliphatic chain, typically a C10-20-acyl, or -alkyl or partly unsaturated fatty acid residue, in particular, an oleoyl-, palmitoeloyl-, elaidoyl-, linoleyl-, linolenyl-, linolenoyl-, arachidoyl-, vaccinyl-, lauroyl-, myristoyl-, palmitoyl-, or stearoyl chain; and where R3 is hydrogen, 2-trimethylamino-1-ethyl, 2-amino-1-ethyl, C1-4-alkyl, C1-5-alkyl substituted with carboxy, C2-5-alkyl substituted with hydroxy, C2-5-alkyl substituted with carboxy and hydroxy, or C2-5-alkyl substituted with carboxy and amino, inositol, sphingosine, or salts of said substances, said lipid comprising also glycerides, isoprenoid lipids, steroids, sterines or sterols, of sulphur- or carbohydrate-containing lipids, or any other bilayer-forming lipids, in particular half-protonated fluid fatty acids, said lipid is selected from the group comprising phosphatidylcholines, phosphatidylethanolamines, phosphatidylglycerols, phosphatidylinositols, phosphatidic acids, phosphatidylserines, sphingomyelins or other sphingophospholipids, glycosphingolipids (including cerebrosides, ceramidepolyhexosides, sulphatides, sphingoplasmalogens), gangliosides and other glycolipids or synthetic lipids, in particular with corresponding sphingosine derivatives, or any other glycolipids, whereby two similar or different chains can be ester-groups-linked to the backbone (as in diacyl and dialkenoyl compound) or be attached to the backbone with ether bonds, as in dialkyl-lipids.
20. Formulation according to claim 18,
characterised in that the surfactant or surfactant-like material is a nonionic, a zwitterionic, an anionic or a cationic surfactant, especially a fatty-acid or -alcohol, an alkyl-tri/di/methyl-ammonium salt, an alkylsulphate salt, a monovalent salt of cholate, deoxycholate, glycocholate, glycodeoxycholate, taurodeoxycholate, taurocholate, etc., an acyl- or alkanoyl-dimethyl-aminoxide, esp. a dodecyl-dimethyl-aminoxide, an alkyl- or alkanoyl-N-methylglucamide, N-alkyl-N,N-dimethylglycine, 3-(acyldimethylammonio)-alkanesulphonate, N-acyl-sulphobetaine, a polyethylene-glycol-octylphenyl ether, esp. a nonaethylene-glycol-octylphenyl ether, a polyethylene-acyl ether, esp. a nonaethylen-dodecyl ether, a polyethylene-glycol-isoacyl ether, esp. a octaethylene-glycol-isotridecyl ether, polyethylene-acyl ether, esp. octaethylenedodecyl ether, polyethylene-glycol-sorbitane-acyl ester, such as polyethylenglykol-20-monolaurate (Tween 20) or polyethylenglykol-20-sorbitan-monooleate (Tween 80), a polyhydroxyethylene-acyl ether, esp. polyhydroxyethylene-lauryl, -myristoyl, -cetylstearyl, or -oleoyl ether, as in polyhydroxyethylene-4 or 6 or 8 or 10 or 12, etc., -lauryl ether (as in Brij series), or in the corresponding ester, e.g. of polyhydroxyethylen-8-stearate (Myrj 45), -laurate or -oleate type, or in polyethoxylated castor oil 40, a sorbitane-monoalkylate (e.g. in Arlacel or Span), esp. sorbitane-monolaurate, an acyl- or alkanoyl-N-methylglucamide, esp. in or decanoyl- or dodecanoyl-N-methylglucamide, an alkyl-sulphate (salt), e.g. in lauryl- or oleoyl-sulphate, sodium deoxycholate, sodium glycodeoxycholate, sodium oleate, sodium taurate, a fatty acid salt, such as sodium elaidate, sodium linoleate, sodium laurate, a lysophospholipid, such as n-octadecylene(=oleoyl)-glycerophosphatidic acid, -phosphorylglycerol, or -phosphorylserine, n-acyl-, e.g. lauryl or oleoyl-glycero-phosphatidic acid, -phosphorylglycerol, or -phosphorylserine, n-tetradecyl- glycero-phosphatidic acid, -phosphorylglycerol, or -phosphorylserine, a corresponding palmitoeloyl-, elaidoyl-, vaccenyl-lysophospholipid or a corresponding short-chain phospholipid, or else a surface-active polypeptide.
21. Formulation according to any one of claims 14 to 20,
characterised in that the average penetrant diameter is between 30 nm and 500 nm, preferably between 40 nm and 250 nm, even more preferably between 50 nm and 200 nm and most preferably between 60 nm and 150 nm.
22. Formulation according to any one of claims 14 to 20,
characterised in that the average diameter of the penetrant is 2 to 25 times bigger than the average diameter of the pores in the barrier, preferably between 2.25 and 15 times bigger, even more preferably between 2.5 and 8 times bigger and most preferably between 3 and 6 times bigger than said average pore diameter.
23. Formulation according to any one of claims 14 through to 22,
characterised in that the dry weight of all carrier droplets in a formulation for the use on human or animal skin is 0.01 weight-% (w-%) to 40 w-% of total formulation mass, in particular between 0.1 w-% and 30 w-%, particularly preferably between 0.5 w-% and 20 w-%, and most preferably between 1 w-% and 10 w-%.
24. Formulation according to any one of claims 14 through to 22,
characterised in that the dry weight of all carrier droplets in a formulation for the use on human or animal mucosa is 0.0001 w-% to 30 w-% of total formulation mass.
25. Formulation according to any one of the claims 1 through to 24,
characterised in that the pH of carrier suspension is between 4 and 10, preferably between 5 and 9, and even more often up to 8.5, as required to maximise the stability of formulation.
26. A method for preparing a formulation for non-invasive application in vivo, according to any one of the preceding claims,
characterised in that penetrants capable of associating and/or incorporating said agent molecules are formed from at least one amphiphilic substance, at least one polar fluid, at least one edge-active substance or surfactant, at least one corticosteroid in an amount of more than 0.1 w-% based on total dry mass of the formulation, and, in case, other customary ingredients, which together form said formulation.
27. The method of claim 26,
characterised in that at least one edge-active substance or surfactant, at least one amphiphilic substance, at least one hydrophilic fluid and the agent are dissolved to form a solution and, if required, are mixed separately, the resulting (partial) mixtures or solutions then being combined to subsequently induce, preferably by action of mechanical energy, such as shaking, stirring, vibrating, homogenising, ultrasonication, shear, freezing and thawing, or filtration using convenient driving pressure, the formation of penetrants that associate with and/or incorporate the agent.
28. The method of claims 26 or 27,
characterised in that said amphiphilic substances are either used as such, or dissolved in a physiologically compatible polar fluid, which may be water or miscible with water, or in a solvation-mediating agent, together with a polar solution.
29. The method of claims 26 or 27,
characterised in that said amphiphilic substances are dissolved in highly volatile alcohols, in especially ethanol, or in pharmaceutically acceptable organic solvents, which are then removed, esp. by evaporation, prior to making final preparation.
30. The method as claimed in claims 28 or 29,
characterised in that the polar solution contains at least one edge-active substance or surfactant.
31. The method according to any one of claims 26 through to 30,
characterised in that the formation of said penetrants is induced by the addition of required substances into a fluid phase, evaporation from a reverse phase, by injection or dialysis, if necessary under the influence of mechanical stress, such as shaking, stirring, in especially high velocity stirring, vibrating, homogenising, ultrasonication, shearing, freezing and thawing, or filtration using convenient, in especially low (1 MPa) or intermediate (up to 10 MPa), driving pressure.
32. The method of claim 31,
characterised in that the formation of said penetrants is induced by filtration, the filtering material having pores sizes between 0.01 μm and 0.8 μm, preferably between 0.02 μm and 0.3 μm, and most preferably between 0.05 μm and 0.15 μm, whereby several filters may be used sequentially or in parallel.
33. The method according to any one of claims 26 through to 32,
characterised in that said agents and penetrants are made to associate, at least partly, after the formation of said penetrants, e.g. after injecting a solution of the drug in a pharmaceutically acceptable fluid, such as ethanol, 1-and 2-propanol, benzyl alcohol, propylene glycol, polyethylene glycol (molecular weight: 200-400 D) or glycerol into the suspending medium, said penetrants being formed previously, using the corresponding or some other suitable manufacturing method, or simultaneously with the drug injection, if required using a co-solution of the drug and, at least some, penetrant ingredients.
34. The method according to any one of the claims 26 through to 33,
characterised in that said penetrants, with which the agent molecules are associated and/or into which the agent is incorporated, are prepared just before the application of the formulation, if convenient from a suitable concentrate or a lyophylisate.
35. Formulation according to any one of claims 11 through to 25,
characterised in that the content of corticosteroids is between 0.1 w-% and 20 w-%, more preferably between 0.25 w-% and 10 w-% and even more preferably between 0.5 w-% and 5 w-%, relative to total dry mass of drug-loaded carriers.
36. Formulation according to claim 35,
characterised in that the relative content of corticosteroids in the case of triamcinolone or one of its derivatives, such as acetonide, is below 2 w-%, relative to total dry mass of the drug-loaded carriers, even more preferably is below 1 w-% and most preferably is below 0.5 w-%.
37. Formulation according to claim 35,
characterised in that the relative content of corticosteroids in the case of hydrocortisone or one of its derivatives is below 20 w-%, relative to total dry mass of the drug-loaded carriers, even more preferably is below 12.5 w-% and most preferably is below 5 w-%.
38. Formulation according to claim 35,
characterised in that the relative content of corticosteroids in the case of dexamethasone or one of its derivatives is below 15 w-%, relative to total dry mass of the drug-loaded carriers, even more preferably is below 10 w-% and most preferably is below 5 w-%.
39. Formulation according to claim 35,
characterised in that the relative content of corticosteroids in the case of clobetasol or one of its derivatives, such as propionate is below 15 w-%, relative to total dry mass of the drug-loaded carriers, even more preferably is below 10 w-% and most preferably is below 5 w-%.
40. Formulation according to claims 35 to 39,
characterised in that the content of said corticosteroid is below the saturation maximum, defined as the content of corticosteroid at which the corticosteroid begins to crystallise in or outside the carrier.
41. Formulation according to claims 1 through to 25 and 35 through to 38,
characterised in that in order to speed up drug action a permeation enhancer is added.
42. Formulation according to claim 41,
characterised in that the permeation enhancer is selected from 1-acyl-azacycloheptan-2-ones (azones), 1-acyl-glucosides, 1-acyl-polyoxyethylenes, 1-acyl-saccharides, 2-n-acyl-cyclohexanones, 2-n-acyl-1,3-dioxolanes (SEPA), 1,2,3-triacyl-glycerols, 1-alkanols, 1-alkanoic acids, 1-alkyl-acetates, 1-alkyl-amines, 1-alkyl-n-alkyl-polyoxyethylenes, 1-alkyl-alkylates, n-alkyl-beta-D-thioglucosides, 1-alkyl-glycerides, 1-alkyl-propyleneglycols, 1-alkyl-polyoxyethylenes, (1-alkyl-)2-pyrrolidones, alkyl-acetoacetates, alkylene-glycols, alkyl-methyl-sulphoxides (alkyl-DMSO), alkyl-propionates, alkyl-sulphates, diacyl-succinates, diacyl-N,N-dimethylaminoacetates (DDAA), diacyl-N,N-dimethylaminoisopropionates (DDAIP), phenyl-alkyl-amines.
43. Formulation according to claim 42,
characterised in that the bulk concentration range of the used enhancers is up to and around 5% for 1-capryl-propylene glycol, 6-10% for 1-[2-(decylthio)ethyl]azacyclopentan-2-one (=HPE-101), <10% for 1-dodecanol, <10% for 1-dodecyl-azacycloheptan-2-one (=azone), in the range of 10% for 2-n-nonyl-1,3-dioxolane (SEPA), <10% for 2-n-octylcyclohexanone, up to, and preferably around, up to 20% for DMSO, up to, and between 5% and 40% for ethanol, in the range of 10% or higher for ethylene glycol, up to 30% for ethyl acetate, 5-50% for glycerol, up to 75% for isopropanol, 1-20% for isopropyl myristate, between 1 and 20% for oleic acid and oleyl-alcohol, of the order of around 1% for oleyl-polyoxyethylene-ether, at least 10% for propylene glycol.
44. Formulation according to claims 11 through to 25 and 35 through to 43,
characterised in that said corticosteroid is added in an amount which enables the formulation to be applied corresponding to an area dose, as expressed by the total dry mass of penetrant applied per unit area, of between 0.1 mg cm−2 and 15 mg cm−2, more preferably between 0.5 mg cm−2 and 10 mg cm−2, particularly preferably between 0.75 mg cm−2 and 5 mg cm−2 and most preferably between 1 mg cm−2 and 2.5 mg cm−2, if said corticosteroid is desired to exert a therapeutic effect in the deep subcutaneous, e.g. muscle or joints, tissue or else in the remote tissues, including the whole body.
45. Formulation according to claims 11 through to 25 and 35 through to 43,
characterised in that said corticosteroid is added in an amount which enables the formulation to be applied with an area dose, as expressed by the total dry mass of penetrant applied per unit area, of between 1 μg cm−2 and 250 μg cm−2, more preferably between 2.5 and 100 μg cm−2, even more preferably between 5 μg cm−2 and 50 μg cm−2 and most preferably between 7.5 μg cm−2 and 20 μg cm−2, if said corticosteroid is desired to exert a mainly local, that is, superficial, rather than systemic therapeutic effect.
46. Formulation according to claims 11 through to 25 and 35 through to 45,
characterised in that consistency and, if necessary other characteristics of the formulation are appropriately selected to enable spraying, smearing, rolling or sponging of the formulation on the application area in particular by using a sprayer, spender, roller or sponge, as appropriate.
47. A method for non-invasive application of corticosteroids by means of penetrants according to any one of the preceding claims,
characterised in that the area dose, as expressed by the total dry mass of penetrant applied per unit area, is selected to be between 0.1 mg cm−2 and 15 mg cm−2, preferably between 0.5 mg cm−2 and 10 mg cm−2, particularly preferably between 0.75 mg cm−2 and 5 mg cm−2 and most preferably between 1 mg cm−2 and 2.5 mg cm−2, if said corticosteroid is desired to exert a therapeutic effect in the deep subcutaneous, e.g. muscle or joints, tissue or else in the remote tissues, including the whole body.
48. A method for non-invasive application of corticosteroids by means of penetrants according to any one of the preceding claims,
characterised in that the area-dose, as expressed by the total dry mass of penetrants applied per unit area, is between 1 μg cm−2 and 250 μg cm−2, preferably between 2.5 μg cm−2 and 100 μg cm−2, more preferably between 5 μg cm−2 and 50 μg cm−2 and most preferably between 7.5 μg cm−2 and 20 μg cm−2, if said corticosteroid is desired to exert a mainly local, that is, superficial, rather than systemic therapeutic effect.
49. A method for non-invasive application of corticosteroids associated with or encapsulated into said penetrants according to any one of the preceding claims,
characterised in that the formulation is applied by spraying, smearing, rolling or sponging on the application area in particular by using a sprayer, spender, roller or sponge, as appropriate.
50. Use of a formulation in accordance with any one of the preceding claims for the treatment of inflammatory disease, dermatosis, kidney or liver failure, adrenal insufficiency, aspiration syndrome, Behcet syndrome, bites and stings, blood disorders, such as cold-haemagglutinin disease, haemolytic anemia, hypereosinophilia, hypoplastic anemia, macroglobulinaemia, trombocytopenic purpura, furthermore, for the management of bone disorders, cerebral oedema, Cogan's syndrome, congenital adrenal hyperplasia, connective tissue disorders, such as lichen, lupus erythematosus, polymyalgia rheumatica, polymyositis and dermatomyositis, epilepsy, eye disorders, such as cataracts, Graves'ophthalmopathy, haemangioma, herpes infections, neuropathies, retinal vasculitis, scleritis, for some gastro-intestinal disorders, such as inflammatory bowel disease, nausea and oesophageal damage, for hypercalcaemia, infections, e.g. of the eye (as in infections mononucleosis), for Kawasaki disease, myasthenia gravis, various pain syndromes, such as postherpetic neuralgia, for polyneuropathies, pancreatitis, in respiratory disorders, such as asthma, for the management of rheumatoid disease and osteoarthritis, rhinitis, sarcoidosis, skin diseases, such as alopecia, eczema, erythema multiforme, lichen, pemphigus and pemphigoid, psoriasis, pyoderma gangrenosum, urticaria, in case of thyroid and vascular disorders.
US11/638,091 1998-12-23 2006-12-12 Formulation for topical non-invasive application in vivo Abandoned US20070184114A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/638,091 US20070184114A1 (en) 1998-12-23 2006-12-12 Formulation for topical non-invasive application in vivo

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
PCT/EP1998/008421 WO2000038653A1 (en) 1998-12-23 1998-12-23 Improved formulation for topical non-invasive application in vivo
US09/887,493 US7175850B2 (en) 1998-12-23 2001-06-22 Formulation for topical non-invasive application in vivo
US11/638,091 US20070184114A1 (en) 1998-12-23 2006-12-12 Formulation for topical non-invasive application in vivo

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/887,493 Continuation US7175850B2 (en) 1998-12-23 2001-06-22 Formulation for topical non-invasive application in vivo

Publications (1)

Publication Number Publication Date
US20070184114A1 true US20070184114A1 (en) 2007-08-09

Family

ID=8167168

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/887,493 Expired - Fee Related US7175850B2 (en) 1998-12-23 2001-06-22 Formulation for topical non-invasive application in vivo
US11/638,091 Abandoned US20070184114A1 (en) 1998-12-23 2006-12-12 Formulation for topical non-invasive application in vivo

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/887,493 Expired - Fee Related US7175850B2 (en) 1998-12-23 2001-06-22 Formulation for topical non-invasive application in vivo

Country Status (22)

Country Link
US (2) US7175850B2 (en)
EP (1) EP1140021B1 (en)
JP (1) JP2002533379A (en)
KR (1) KR100638150B1 (en)
CN (1) CN1320880C (en)
AT (1) ATE272391T1 (en)
AU (1) AU770803B2 (en)
BR (1) BR9816113A (en)
CA (1) CA2356080C (en)
CZ (1) CZ20012038A3 (en)
DE (1) DE69825495T2 (en)
EE (1) EE200100342A (en)
ES (1) ES2226203T3 (en)
HK (1) HK1040629B (en)
HR (1) HRP20010309B1 (en)
HU (1) HUP0104424A3 (en)
MX (1) MXPA01006424A (en)
NO (1) NO20013164L (en)
PL (1) PL193824B1 (en)
RU (1) RU2207844C2 (en)
TR (1) TR200101790T2 (en)
WO (1) WO2000038653A1 (en)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020048596A1 (en) * 1994-12-30 2002-04-25 Gregor Cevc Preparation for the transport of an active substance across barriers
US20030099694A1 (en) * 1999-07-05 2003-05-29 Gregor Cevc Method for the improvement of transport across adaptable semi-permeable barriers
US20040105881A1 (en) * 2002-10-11 2004-06-03 Gregor Cevc Aggregates with increased deformability, comprising at least three amphipats, for improved transport through semi-permeable barriers and for the non-invasive drug application in vivo, especially through the skin
US20070154403A1 (en) * 2006-01-05 2007-07-05 Thomas Skold Oral, Pulmonary and Transmucosal Delivery Composition
US20080095722A1 (en) * 2004-11-12 2008-04-24 Idea Ag Extended Surface Aggregates in the Treatment of Skin Conditions
US20080159963A1 (en) * 2003-09-22 2008-07-03 Rucinski Paul J Devices and methods for delivering active agents to target sites
US20080279815A1 (en) * 1998-10-23 2008-11-13 Idea Ag Method for developing testing, and using associates of macromolecules and complex aggregates for improved payload and controllable de/association rates
US20090226491A1 (en) * 2002-03-13 2009-09-10 Thomas Skold Water-based delivery systems
US20100040657A1 (en) * 2008-08-15 2010-02-18 Kevin Scott Creevy Gentle, non-irritating, non-alcoholic skin disinfectant
US7867480B1 (en) 1999-01-27 2011-01-11 Gregor Cevc Non-invasive vaccination through the skin
US7927622B1 (en) 1999-01-27 2011-04-19 Gregor Cevc Methods of transnasal transport/immunization with highly adaptable carriers
US20110097372A1 (en) * 2009-10-26 2011-04-28 Rucinski Paul J Devices, methods, and compositions for controlling infections
AU2007333678B2 (en) * 2006-12-19 2013-05-02 Innovation Technologies, Inc. Devices and methods for delivering active agents to target sites
WO2015065795A1 (en) * 2013-10-28 2015-05-07 Wincom, Inc. Purifying liquid azole heteroaromatic compound-containing mixtures
US10265280B2 (en) 2016-11-14 2019-04-23 Mingwu Wang Formulations for the treatment of ocular surface diseases and related methods
US10406088B2 (en) 2015-01-20 2019-09-10 TetraDerm Group LLC Versatile topical drug delivery vehicle and multifactorial tissue moisturizer that provides mucosal and skin barrier restoration

Families Citing this family (93)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2002533379A (en) * 1998-12-23 2002-10-08 イデア アクチェンゲゼルシャフト Improved formulation for locally non-invasive use in vivo
US20030077301A1 (en) * 1999-12-16 2003-04-24 Maibach Howard I. Topical pharmaceutical composition for the treatment of inflammatory dermatoses
JP2004504357A (en) * 2000-07-26 2004-02-12 アルコン,インコーポレイテッド Pharmaceutical suspension compositions containing no polymeric suspending agent
DE60117858T2 (en) 2000-12-07 2006-11-30 Universiteit Utrecht Holding B.V. COMPOSITION FOR THE TREATMENT OF INFLAMMATORY DISEASES
US8246969B2 (en) 2001-11-16 2012-08-21 Skinmedica, Inc. Compositions containing aromatic aldehydes and their use in treatments
US7763663B2 (en) 2001-12-19 2010-07-27 University Of Massachusetts Polysaccharide-containing block copolymer particles and uses thereof
US8367098B2 (en) 2002-04-30 2013-02-05 The Population Council, Inc. Unique combinations of antimicrobial compositions
EP1371362A1 (en) * 2002-06-12 2003-12-17 Universiteit Utrecht Holding B.V. Composition for treatment of inflammatory disorders
IL165910A0 (en) * 2002-07-03 2006-01-15 Pericor Science Inc Compositions of hyaluronic acid and methods of use
ES2298566T3 (en) * 2002-10-11 2008-05-16 Idea Ag ADDED WITH INCREASED DEFORMABILITY, WHICH INCLUDES AT LEAST THREE AMPHIPATIC COMPONENTS, FOR IMPROVED TRANSPORTATION THROUGH SEMIPERMEABLE BARRIERS AND FOR THE APPLICATION OF NON-INVASIVE PHARMACES IN VIVID, ESPECIALLY THROUGH THE SKIN.
AU2003293500A1 (en) * 2002-12-12 2004-07-09 Agennix Incorporated Lactoferrin in the reduction of pain
WO2004103285A2 (en) * 2003-05-14 2004-12-02 Agennix Incorporated Lactoferrin in the treatment of diabetes mellitus
FR2861304B1 (en) * 2003-10-23 2008-07-18 Univ Grenoble 1 CFTR CHANNEL MODULATORS
US9211248B2 (en) 2004-03-03 2015-12-15 Revance Therapeutics, Inc. Compositions and methods for topical application and transdermal delivery of botulinum toxins
US8092788B2 (en) 2004-03-03 2012-01-10 Revance Therapeutics, Inc. Compositions and methods for topical diagnostic and therapeutic transport
CA2562356A1 (en) * 2004-04-08 2005-10-27 Retmed Pty Ltd Treatment of ophthalmic conditions with mineralcorticoids
KR100772327B1 (en) * 2004-04-22 2007-10-31 주식회사유한양행 Nanoemulsion Compositions Containing Hydrocortisone or Its Salts
DE102004021992A1 (en) * 2004-05-03 2005-11-24 Boehringer Ingelheim Pharma Gmbh & Co. Kg Topical preparation containing ambroxol
US20050255154A1 (en) 2004-05-11 2005-11-17 Lena Pereswetoff-Morath Method and composition for treating rhinitis
BRPI0515888A (en) * 2004-09-24 2008-08-12 Lipo Chemicals Inc release system, composition for topical application to mammalian skin or hair, and method for treating mammalian skin
WO2006047744A2 (en) * 2004-10-26 2006-05-04 Agennix Incorporated Compositions of lactoferrin related peptides and uses thereof
EP1835884A2 (en) * 2005-01-14 2007-09-26 Lipo Chemicals Inc Composition and method for treating hyperpigmented skin
US9314416B2 (en) 2005-03-03 2016-04-19 Revance Therapeutics, Inc. Compositions and methods for topical application and transdermal delivery of botulinum toxins
US20090220583A1 (en) 2005-06-09 2009-09-03 Lena Pereswetoff-Morath Method and composition for treating inflammatory disorders
BRPI0613631A8 (en) 2005-07-18 2017-12-26 Univ Massachusetts Lowell nanoemulsion and method,
TWI429452B (en) * 2005-08-31 2014-03-11 Abraxis Bioscience Llc Compositions comprising poorly water soluble pharmaceutical agents and antimicrobial agents
KR101457834B1 (en) * 2005-08-31 2014-11-05 아브락시스 바이오사이언스, 엘엘씨 Compositions and methods for preparation of poorly water soluble drugs with increased stability
PL1919450T3 (en) * 2005-09-01 2014-11-28 Meda Ab Antihistamine- and corticosteroid-containing liposome composition and its use for the manufacture of a medicament for treating rhinitis and related disorders
US20090082321A1 (en) * 2007-09-21 2009-03-26 Allergan, Inc. Steroid containing drug delivery systems
US9486408B2 (en) 2005-12-01 2016-11-08 University Of Massachusetts Lowell Botulinum nanoemulsions
US7998717B2 (en) * 2005-12-02 2011-08-16 Pacific Biosciences Of California, Inc. Mitigation of photodamage in analytical reactions
US8343962B2 (en) 2006-03-06 2013-01-01 Nuvo Research Inc. Topical formulation
US7795309B2 (en) * 2006-03-06 2010-09-14 Fqubed Inc. Topical formulation including diclofenac, or a pharmaceutically acceptable salt thereof
US9642912B2 (en) 2006-03-06 2017-05-09 Crescita Therapeutics Inc. Topical formulations for treating skin conditions
US9308181B2 (en) 2006-03-06 2016-04-12 Nuvo Research Inc. Topical formulations, systems and methods
US7767217B2 (en) * 2006-03-14 2010-08-03 Foresight Biotherapeutics Ophthalmic compositions comprising povidone-iodine
WO2008020032A1 (en) * 2006-08-16 2008-02-21 Action Medicines, S.L. Use of 2,5-dihydroxybenzene derivatives for the treatment of ocular diseases
JP5292304B2 (en) 2006-12-01 2013-09-18 アンテリオス, インコーポレイテッド Peptide nanoparticles and uses thereof
BRPI0719756A2 (en) 2006-12-01 2014-01-21 Anterios Inc Amphiphilic Nanoparticles
CN101032473B (en) * 2007-04-05 2010-04-07 上海交通大学 Sandwich type medical releasing film and preparing method thereof
US10016451B2 (en) 2007-05-31 2018-07-10 Anterios, Inc. Nucleic acid nanoparticles and uses therefor
KR102088949B1 (en) 2007-07-26 2020-03-13 레반스 테라퓨틱스, 아이엔씨. Antimicrobial peptide, compositions, and methods of use
ITRM20070510A1 (en) * 2007-10-02 2009-04-03 Rmfa Trading S A OPHTHALMIC COMPOSITIONS BASED ON TAMARINDO SEED POLYESACCARIDE AND HYALURONIC ACID.
KR101516706B1 (en) * 2008-01-18 2015-05-04 인데나 에스.피.에이 Abnormal intraocular pressure treatment
EP3656385A1 (en) 2008-05-28 2020-05-27 ReveraGen BioPharma, Inc. Non-hormonal steroid modulators of nf-kb for treatment of disease
JP5667060B2 (en) * 2008-10-03 2015-02-12 ネクスメツド・ホールデイングス・インコーポレイテツド Stabilized composition for treating psoriasis
MY170121A (en) * 2009-06-03 2019-07-05 Ulrich Vierl Formulations for the treatment of deep tissue pain
DE112010003355T5 (en) 2009-08-21 2012-07-12 Targeted Delivery Technologies Ltd. Vesicular formulations
US9198921B2 (en) 2010-04-05 2015-12-01 Reveragen Biopharma, Inc. Non-hormonal steroid modulators of NF-κB for treatment of disease
US8834847B2 (en) 2010-08-12 2014-09-16 Pacific Biosciences Of California, Inc. Photodamage mitigation compounds and systems
CA2817206C (en) * 2010-11-10 2020-03-10 Tengion, Inc. Injectable formulations for organ augmentation
CA3043905A1 (en) 2011-01-07 2012-07-12 Allergan, Inc. Compositions comprising substituted benzaldehydes and use thereof for lightening skin or treating hyperpigmenation and hypermelanosis disorders
RU2456979C1 (en) * 2011-04-11 2012-07-27 Общество с ограниченной ответственностью "ЭкоБиоФармДубна" Phospholipid nanoparticle indometacin applicable in ophthalmology
CN104023717B (en) * 2011-06-29 2017-08-04 阿维达斯制药有限责任公司 The topical formulation and its purposes of mediator are delivered including lipid microcapsules
CN102847151B (en) * 2011-07-01 2014-08-13 天津金耀集团有限公司 Inhalant preparation containing sodium 2-mercaptoethanesulfonate and glucocorticoid
CA2856520C (en) 2011-11-23 2021-04-06 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US9301920B2 (en) 2012-06-18 2016-04-05 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
RU2477632C1 (en) * 2011-12-22 2013-03-20 Федеральное государственное бюджетное образовательное учреждение высшего профессионального образования "Российский химико-технологический университет им. Д.И. Менделеева (РХТУ им. Д.И. Менделеева) Method for preparing liposomal form of biologically active substance
GB201205642D0 (en) 2012-03-29 2012-05-16 Sequessome Technology Holdings Ltd Vesicular formulations
US20150196640A1 (en) 2012-06-18 2015-07-16 Therapeuticsmd, Inc. Progesterone formulations having a desirable pk profile
US10806697B2 (en) 2012-12-21 2020-10-20 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US20130338122A1 (en) 2012-06-18 2013-12-19 Therapeuticsmd, Inc. Transdermal hormone replacement therapies
US10806740B2 (en) 2012-06-18 2020-10-20 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
CN102772396B (en) * 2012-07-25 2013-12-18 杭州普瑞美克生物科技有限公司 Cooperatively used combination type preparation for cleaning and sterilizing skin after being bitten or scratched by animals
US10471072B2 (en) 2012-12-21 2019-11-12 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11266661B2 (en) 2012-12-21 2022-03-08 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10537581B2 (en) 2012-12-21 2020-01-21 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10568891B2 (en) 2012-12-21 2020-02-25 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US9180091B2 (en) 2012-12-21 2015-11-10 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US11246875B2 (en) 2012-12-21 2022-02-15 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US20160193147A1 (en) * 2013-07-31 2016-07-07 Sequessome Technology Holdings Limited Vesicles
CA2940494C (en) 2014-03-14 2022-11-08 Gojo Industries, Inc. Hand sanitizers with improved aesthetics and skin-conditioning to encourage compliance with hand hygiene guidelines
MX2016014281A (en) 2014-05-22 2017-02-22 Therapeuticsmd Inc Natural combination hormone replacement formulations and therapies.
US9459201B2 (en) 2014-09-29 2016-10-04 Zyomed Corp. Systems and methods for noninvasive blood glucose and other analyte detection and measurement using collision computing
WO2017004205A1 (en) 2015-06-29 2017-01-05 Reveragen Biopharma, Inc. NON-HORMONAL STEROID MODULATORS OF NF-κB FOR TREATMENT OF DISEASE
EA038208B1 (en) 2015-06-30 2021-07-23 Секвессом Текнолоджи Холдингс Лимитед Multiphasic compositions
US10328087B2 (en) 2015-07-23 2019-06-25 Therapeuticsmd, Inc. Formulations for solubilizing hormones
WO2017035107A1 (en) * 2015-08-24 2017-03-02 Smith & Nephew, Inc. Synergistic antibacterial activity of medium polarity oils in combination with antibacterial agents on bacterial biofilms
CN107477368A (en) * 2015-09-11 2017-12-15 刘玉友 A kind of additive for being used to improve drag reducer for crude oil anti-shear performance
WO2017146602A1 (en) * 2016-02-24 2017-08-31 Arcasiu Nicolae Chlorine or bromine salts of cetylpyridinium for use in the treatment of cutaneous and acute porphyrias and psoriasis
US9554738B1 (en) 2016-03-30 2017-01-31 Zyomed Corp. Spectroscopic tomography systems and methods for noninvasive detection and measurement of analytes using collision computing
WO2017173044A1 (en) 2016-04-01 2017-10-05 Therapeuticsmd Inc. Steroid hormone compositions in medium chain oils
WO2017173071A1 (en) 2016-04-01 2017-10-05 Therapeuticsmd, Inc. Steroid hormone pharmaceutical composition
CN117731591A (en) 2016-11-21 2024-03-22 艾里奥治疗公司 Transdermal delivery of large agents
CN107970257A (en) * 2017-12-22 2018-05-01 成都博创必成医药技术有限公司 A kind of ear pharmaceutical composition, preparation method and applications and ear pharmaceutical preparation
WO2020031065A1 (en) * 2018-08-08 2020-02-13 3M Innovative Properties Company Therapeutic composition and related methods
DE102018133374A1 (en) * 2018-12-21 2020-06-25 Gelita Ag Collagen hydrolyzate for use against skin and intestinal diseases
US11382922B2 (en) 2019-03-07 2022-07-12 Reveragen Biopharma, Inc. Aqueous oral pharmaceutical suspension compositions
CN111904932B (en) * 2019-05-08 2023-06-20 北京德立福瑞医药科技有限公司 Micelle preparation containing glucocorticoid and preparation method thereof
DE102019218241A1 (en) * 2019-11-26 2021-05-27 Beiersdorf Ag Active ingredient combinations of ubiquinol and carrageenan and cosmetic or dermatological preparations containing such active ingredient combinations
US11633405B2 (en) 2020-02-07 2023-04-25 Therapeuticsmd, Inc. Steroid hormone pharmaceutical formulations
CN114831939A (en) * 2021-01-30 2022-08-02 南京星银药业集团有限公司 Triamcinolone acetonide acetate composition spray and preparation method thereof
CN113425845B (en) * 2021-08-10 2022-12-02 成都倍特药业股份有限公司 Carrier preparation for preventing and/or treating alopecia and preparation method and application thereof

Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4333927A (en) * 1980-03-27 1982-06-08 Mitsubishi Chemical Industries, Ltd. Skin preparation
US4619794A (en) * 1982-02-17 1986-10-28 Ciba-Geigy Corporation Spontaneous preparation of small unilamellar liposomes
US4814161A (en) * 1985-01-16 1989-03-21 Riker Laboratories, Inc. Drug-containing chlorofluorocarbon aerosol propellent formulations
US4911928A (en) * 1987-03-13 1990-03-27 Micro-Pak, Inc. Paucilamellar lipid vesicles
US4921706A (en) * 1984-11-20 1990-05-01 Massachusetts Institute Of Technology Unilamellar lipid vesicles and method for their formation
US4937078A (en) * 1988-08-26 1990-06-26 Mezei Associates Limited Liposomal local anesthetic and analgesic products
USRE33273E (en) * 1982-08-18 1990-07-24 Georgia Tech Research Corporation Materials having improved nonfouling characteristics and method of making same
US4944948A (en) * 1989-02-24 1990-07-31 Liposome Technology, Inc. EGF/Liposome gel composition and method
US4954345A (en) * 1985-12-04 1990-09-04 Rohm Pharma Gmbh Dermally acting pharmaceutical preparation with liposomes as vehicle means
US5238613A (en) * 1987-05-20 1993-08-24 Anderson David M Microporous materials
US5322685A (en) * 1989-08-03 1994-06-21 Hisamitsu Pharmaceutical Co., Inc. Skin cream preparation for external use
US5607692A (en) * 1993-12-30 1997-03-04 L'oreal Depigmenting composition for the simultaneous treatment of the surface layers and deep layers of the skin, and use thereof
US5741515A (en) * 1994-10-20 1998-04-21 Bayer Aktiengesellschaft Ketoprofen liposomes
US5985379A (en) * 1997-07-22 1999-11-16 Franklin Mint Company Decorative display plate
US20020048596A1 (en) * 1994-12-30 2002-04-25 Gregor Cevc Preparation for the transport of an active substance across barriers
US6562370B2 (en) * 1999-12-16 2003-05-13 Dermatrends, Inc. Transdermal administration of steroid drugs using hydroxide-releasing agents as permeation enhancers
US6582724B2 (en) * 1999-12-16 2003-06-24 Dermatrends, Inc. Dual enhancer composition for topical and transdermal drug delivery
US6586000B2 (en) * 1999-12-16 2003-07-01 Dermatrends, Inc. Hydroxide-releasing agents as skin permeation enhancers
US6645520B2 (en) * 1999-12-16 2003-11-11 Dermatrends, Inc. Transdermal administration of nonsteroidal anti-inflammatory drugs using hydroxide-releasing agents as permeation enhancers
US7175850B2 (en) * 1998-12-23 2007-02-13 Idea Ag Formulation for topical non-invasive application in vivo

Family Cites Families (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS55153713A (en) 1979-05-02 1980-11-29 Kureha Chem Ind Co Ltd Pharmaceutical preparation of ribosome containing active substance
CA1134276A (en) 1980-03-26 1982-10-26 Alec D. Keith Phosphatide-lower alkanolamine complex for topical application
EP0102324A3 (en) 1982-07-29 1984-11-07 Ciba-Geigy Ag Lipids and surfactants in an aqueous medium
EP0152379A3 (en) 1984-02-15 1986-10-29 Ciba-Geigy Ag Process for preparing pharmaceutical compositions containing unilamellar liposomes
JPS61271204A (en) 1985-05-27 1986-12-01 Shiseido Co Ltd Liposome pharmaceutical
IL79114A (en) 1985-08-07 1990-09-17 Allergan Pharma Method and composition for making liposomes
EP0238554A4 (en) 1985-09-27 1987-10-27 Univ California Liposome transdermal drug delivery system.
JPS6295134A (en) * 1985-10-21 1987-05-01 Nippon Saafuakutanto Kogyo Kk Production of liposome
FR2597367B1 (en) 1986-04-22 1988-07-15 Oreal PROCESS FOR FACILITATING THE FORMATION OF LIPID SPHERULES DISPERSION IN AN AQUEOUS PHASE AND FOR IMPROVING THEIR STABILITY AND THEIR ENCAPSULATION RATE, AND CORRESPONDING DISPERSIONS.
DK86988A (en) 1987-02-25 1988-08-26 Takeda Chemical Industries Ltd LIPOSOM PREPARATION AND APPLICATION THEREOF
CA1323306C (en) 1987-03-05 1993-10-19 Mircea C. Popescu Pharmacological agent-lipid solution preparation
US4855090A (en) 1987-03-13 1989-08-08 Micro-Pak, Inc. Method of producing high aqueous volume multilamellar vesicles
US4828837A (en) 1987-03-30 1989-05-09 Liposome Technology, Inc. Non-crystalline minoxidil composition, its production and application
US4783450A (en) 1987-04-13 1988-11-08 Warner-Lambert Company Use of commercial lecithin as skin penetration enhancer
IL86650A0 (en) 1987-06-30 1988-11-30 Biophor Corp Animal derived cells and liposomes,having an antigenic protein incorporated into their membrane
US5064655A (en) 1989-02-24 1991-11-12 Liposome Technology, Inc. Liposome gel composition and method
EP0393707B1 (en) 1989-04-21 1994-10-26 Otsuka Pharmaceutical Co., Ltd. Bioactive compounds associated with liposomes and their use in pharmaceutical preparations
WO1991004013A1 (en) 1989-09-21 1991-04-04 Micro Vesicular Systems, Inc. Hybrid paucilamellar lipid vesicles
CA2067754C (en) 1990-08-24 2002-06-04 Gregor Cevc Preparation for the application of agents in mini-droplets
SE9003100D0 (en) 1990-09-28 1990-09-28 Kabivitrum Ab LIPID FORMULATION SYSTEM
JP2922017B2 (en) 1991-03-25 1999-07-19 第一製薬株式会社 Oral lipid membrane structure
DE59203307D1 (en) 1991-06-10 1995-09-21 Lohmann Therapie Syst Lts NITROGLYCERIN PLASTER AND METHOD FOR THE PRODUCTION THEREOF.
SE9200951D0 (en) 1992-03-27 1992-03-27 Kabi Pharmacia Ab PHARMACEUTICAL COMPOSITION CONTAINING A DEFINED LIPID SYSTEM
SE9200952D0 (en) 1992-03-27 1992-03-27 Kabi Pharmacia Ab PHARMACEUTICAL CARRIER SYSTEM CONTAINING DEFINED LIPIDS
DE4336557C2 (en) 1993-05-06 1997-07-17 Lohmann Therapie Syst Lts Estradiol-containing transdermal therapeutic system, process for its preparation and its use
US5536263A (en) 1994-03-30 1996-07-16 Lectec Corporation Non-occulusive adhesive patch for applying medication to the skin
US5540934A (en) 1994-06-22 1996-07-30 Touitou; Elka Compositions for applying active substances to or through the skin
US5958379A (en) 1994-09-30 1999-09-28 Mika Pharma Gesellschaft Fuer Die Entwicklung Und Vermarktung Pharmazeutischer Producte Mbh Pharmaceutical composition
DE4447287C1 (en) 1994-12-30 1996-11-07 Cevc Gregor Droplet-in-fluid composition to transport agent e.g. through skin
DE19512181C2 (en) 1995-03-31 2003-11-06 Hexal Pharma Gmbh Transdermal system with ramipril and / or trandolapril as an ACE inhibitor
CA2238263A1 (en) 1995-12-12 1997-06-19 Vesifact Ag Cortisone spray for topical administration
US5837289A (en) 1996-07-23 1998-11-17 Grasela; John C. Transdermal delivery of medications using a combination of penetration enhancers
NZ334077A (en) 1996-08-09 2000-09-29 Keygene Nv Genes having resistance against nematodes and/or aphids
JP2000516244A (en) 1996-08-22 2000-12-05 リサーチ・トライアングル・ファーマシューティカルズ Composition containing fine particles of water-insoluble substance and method for producing the same
AU5777498A (en) 1997-01-13 1998-08-03 Cilag A.G. Liposome-based topical tretinoin formulation
US5891467A (en) 1997-01-31 1999-04-06 Depotech Corporation Method for utilizing neutral lipids to modify in vivo release from multivesicular liposomes
WO1998051281A1 (en) 1997-05-14 1998-11-19 Senju Pharmaceutical Co., Ltd. Aqueous suspension preparations with excellent redispersibility
CA2342628C (en) 1998-09-03 2009-07-07 Loma Linda University Medical Center The use of r-flurbiprofen for the treatment of inflammation
SI1031347T1 (en) 1999-01-27 2002-10-31 Idea Ag Transnasal transport/immunisation with highly adaptable carriers
US6294192B1 (en) 1999-02-26 2001-09-25 Lipocine, Inc. Triglyceride-free compositions and methods for improved delivery of hydrophobic therapeutic agents
US6309663B1 (en) 1999-08-17 2001-10-30 Lipocine Inc. Triglyceride-free compositions and methods for enhanced absorption of hydrophilic therapeutic agents
MXPA02000053A (en) 1999-07-05 2003-07-21 Idea Ag A method for the improvement of transport across adaptable semi-permeable barriers.
JP2004504357A (en) 2000-07-26 2004-02-12 アルコン,インコーポレイテッド Pharmaceutical suspension compositions containing no polymeric suspending agent
US6387383B1 (en) 2000-08-03 2002-05-14 Dow Pharmaceutical Sciences Topical low-viscosity gel composition

Patent Citations (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4427670A (en) * 1980-03-27 1984-01-24 Mitsubishi Chemical Industries Limited Skin preparation
US4333927A (en) * 1980-03-27 1982-06-08 Mitsubishi Chemical Industries, Ltd. Skin preparation
US4619794A (en) * 1982-02-17 1986-10-28 Ciba-Geigy Corporation Spontaneous preparation of small unilamellar liposomes
USRE33273E (en) * 1982-08-18 1990-07-24 Georgia Tech Research Corporation Materials having improved nonfouling characteristics and method of making same
US4921706A (en) * 1984-11-20 1990-05-01 Massachusetts Institute Of Technology Unilamellar lipid vesicles and method for their formation
US4814161A (en) * 1985-01-16 1989-03-21 Riker Laboratories, Inc. Drug-containing chlorofluorocarbon aerosol propellent formulations
US4954345A (en) * 1985-12-04 1990-09-04 Rohm Pharma Gmbh Dermally acting pharmaceutical preparation with liposomes as vehicle means
US4911928A (en) * 1987-03-13 1990-03-27 Micro-Pak, Inc. Paucilamellar lipid vesicles
US5238613A (en) * 1987-05-20 1993-08-24 Anderson David M Microporous materials
US4937078A (en) * 1988-08-26 1990-06-26 Mezei Associates Limited Liposomal local anesthetic and analgesic products
US4944948A (en) * 1989-02-24 1990-07-31 Liposome Technology, Inc. EGF/Liposome gel composition and method
US5322685A (en) * 1989-08-03 1994-06-21 Hisamitsu Pharmaceutical Co., Inc. Skin cream preparation for external use
US5607692A (en) * 1993-12-30 1997-03-04 L'oreal Depigmenting composition for the simultaneous treatment of the surface layers and deep layers of the skin, and use thereof
US5741515A (en) * 1994-10-20 1998-04-21 Bayer Aktiengesellschaft Ketoprofen liposomes
US20020048596A1 (en) * 1994-12-30 2002-04-25 Gregor Cevc Preparation for the transport of an active substance across barriers
US5985379A (en) * 1997-07-22 1999-11-16 Franklin Mint Company Decorative display plate
US7175850B2 (en) * 1998-12-23 2007-02-13 Idea Ag Formulation for topical non-invasive application in vivo
US6562370B2 (en) * 1999-12-16 2003-05-13 Dermatrends, Inc. Transdermal administration of steroid drugs using hydroxide-releasing agents as permeation enhancers
US6582724B2 (en) * 1999-12-16 2003-06-24 Dermatrends, Inc. Dual enhancer composition for topical and transdermal drug delivery
US6586000B2 (en) * 1999-12-16 2003-07-01 Dermatrends, Inc. Hydroxide-releasing agents as skin permeation enhancers
US6645520B2 (en) * 1999-12-16 2003-11-11 Dermatrends, Inc. Transdermal administration of nonsteroidal anti-inflammatory drugs using hydroxide-releasing agents as permeation enhancers
US6835392B2 (en) * 1999-12-16 2004-12-28 Dermatrends, Inc. Dual enhancer composition for topical and transdermal drug delivery

Cited By (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020048596A1 (en) * 1994-12-30 2002-04-25 Gregor Cevc Preparation for the transport of an active substance across barriers
US20080279815A1 (en) * 1998-10-23 2008-11-13 Idea Ag Method for developing testing, and using associates of macromolecules and complex aggregates for improved payload and controllable de/association rates
US7867480B1 (en) 1999-01-27 2011-01-11 Gregor Cevc Non-invasive vaccination through the skin
US7927622B1 (en) 1999-01-27 2011-04-19 Gregor Cevc Methods of transnasal transport/immunization with highly adaptable carriers
US20030099694A1 (en) * 1999-07-05 2003-05-29 Gregor Cevc Method for the improvement of transport across adaptable semi-permeable barriers
US7591949B2 (en) 1999-07-05 2009-09-22 Idea Ag Method for the improvement of transport across adaptable semi-permeable barriers
US7459171B2 (en) 1999-07-05 2008-12-02 Idea Ag Method for the improvement of transport across adaptable semi-permeable barriers
US20090226491A1 (en) * 2002-03-13 2009-09-10 Thomas Skold Water-based delivery systems
US8029810B2 (en) 2002-03-13 2011-10-04 Thomas Skold Water-based delivery systems
US20070031483A1 (en) * 2002-10-11 2007-02-08 Gregor Cevc Aggregates with increased deformability, comprising at least three amphipats, for improved transport through semi-permeable barriers and for the non-invasive drug application in vivo, especially through the skin
US20090042989A1 (en) * 2002-10-11 2009-02-12 Idea Ag Nsaid formulations, based on highly adaptable aggregates, for improved transport through barriers and topical drug delivery
US20090060989A1 (en) * 2002-10-11 2009-03-05 Idea Ag Nsaid formulations, based on highly adaptable aggregates, for improved transport through barriers and topical drug delivery
US20090060990A1 (en) * 2002-10-11 2009-03-05 Idea Ag Nsaid formulations, based on highly adaptable aggregates, for improved transport through barriers and topical drug delivery
US7473432B2 (en) 2002-10-11 2009-01-06 Idea Ag NSAID formulations, based on highly adaptable aggregates, for improved transport through barriers and topical drug delivery
US20040105881A1 (en) * 2002-10-11 2004-06-03 Gregor Cevc Aggregates with increased deformability, comprising at least three amphipats, for improved transport through semi-permeable barriers and for the non-invasive drug application in vivo, especially through the skin
US20110288507A1 (en) * 2003-09-22 2011-11-24 Rucinski Paul J Devices and Methods for Delivering Active Agents to Target Sites
US7959617B2 (en) * 2003-09-22 2011-06-14 Innovation Technologies, Inc. Devices and methods for delivering active agents to target sites
US20100049149A1 (en) * 2003-09-22 2010-02-25 Rucinski Paul J Devices and Methods for Delivering Active Agents to Target Sites
US20100057023A1 (en) * 2003-09-22 2010-03-04 Rucinski Paul J Devices and methods for delivering active agents to target sites
US7662125B2 (en) * 2003-09-22 2010-02-16 Innovation Technologies, Inc. Devices and methods for delivering active agents to target sites
US20080159963A1 (en) * 2003-09-22 2008-07-03 Rucinski Paul J Devices and methods for delivering active agents to target sites
US20080095722A1 (en) * 2004-11-12 2008-04-24 Idea Ag Extended Surface Aggregates in the Treatment of Skin Conditions
US20070154403A1 (en) * 2006-01-05 2007-07-05 Thomas Skold Oral, Pulmonary and Transmucosal Delivery Composition
AU2007333678B2 (en) * 2006-12-19 2013-05-02 Innovation Technologies, Inc. Devices and methods for delivering active agents to target sites
US20100040657A1 (en) * 2008-08-15 2010-02-18 Kevin Scott Creevy Gentle, non-irritating, non-alcoholic skin disinfectant
US8173147B2 (en) * 2008-08-15 2012-05-08 Xttrium Laboratories, Inc. Gentle, non-irritating, non-alcoholic skin disinfectant
US20110097372A1 (en) * 2009-10-26 2011-04-28 Rucinski Paul J Devices, methods, and compositions for controlling infections
WO2015065795A1 (en) * 2013-10-28 2015-05-07 Wincom, Inc. Purifying liquid azole heteroaromatic compound-containing mixtures
US9309205B2 (en) 2013-10-28 2016-04-12 Wincom, Inc. Filtration process for purifying liquid azole heteroaromatic compound-containing mixtures
US9802905B2 (en) 2013-10-28 2017-10-31 Wincom, Inc. Filtration process for purifying liquid azole heteroaromatic compound-containing mixtures
US10406088B2 (en) 2015-01-20 2019-09-10 TetraDerm Group LLC Versatile topical drug delivery vehicle and multifactorial tissue moisturizer that provides mucosal and skin barrier restoration
US10265280B2 (en) 2016-11-14 2019-04-23 Mingwu Wang Formulations for the treatment of ocular surface diseases and related methods

Also Published As

Publication number Publication date
JP2002533379A (en) 2002-10-08
HK1040629B (en) 2005-01-28
EP1140021A1 (en) 2001-10-10
CN1320880C (en) 2007-06-13
EP1140021B1 (en) 2004-08-04
AU2513799A (en) 2000-07-31
CA2356080C (en) 2009-05-12
US20020064524A1 (en) 2002-05-30
NO20013164L (en) 2001-08-22
ES2226203T3 (en) 2005-03-16
HRP20010309A2 (en) 2002-06-30
CA2356080A1 (en) 2000-07-06
KR20010107991A (en) 2001-12-07
HUP0104424A2 (en) 2002-03-28
ATE272391T1 (en) 2004-08-15
MXPA01006424A (en) 2002-06-04
HRP20010309B1 (en) 2005-06-30
EE200100342A (en) 2002-10-15
US7175850B2 (en) 2007-02-13
DE69825495D1 (en) 2004-09-09
WO2000038653A1 (en) 2000-07-06
CZ20012038A3 (en) 2001-09-12
HUP0104424A3 (en) 2002-12-28
HK1040629A1 (en) 2002-06-21
TR200101790T2 (en) 2001-10-22
AU770803B2 (en) 2004-03-04
BR9816113A (en) 2001-10-23
PL349467A1 (en) 2002-07-29
PL193824B1 (en) 2007-03-30
KR100638150B1 (en) 2006-10-26
DE69825495T2 (en) 2005-07-28
CN1327382A (en) 2001-12-19
NO20013164D0 (en) 2001-06-22
RU2207844C2 (en) 2003-07-10

Similar Documents

Publication Publication Date Title
US7175850B2 (en) Formulation for topical non-invasive application in vivo
Cevc et al. Hydrocortisone and dexamethasone in very deformable drug carriers have increased biological potency, prolonged effect, and reduced therapeutic dosage
CN101043875B (en) Liposomal compositions of glucocorticoid and glucocorticoid derivatives
EP1888033B1 (en) Method and composition for treating inflammatory disorders
RU2001120008A (en) Advanced in vivo topical, non-invasive topical preparation
US7591949B2 (en) Method for the improvement of transport across adaptable semi-permeable barriers
EP1919450B1 (en) Antihistamine- and corticosteroid-containing liposome composition and its use for the manufacture of a medicament for treating rhinitis and related disorders
US20090047336A1 (en) novel formulation of dehydrated lipid vesicles for controlled release of active pharmaceutical ingredient via inhalation
EP2405896B1 (en) Glycerosomes and use thereof in pharmaceutical and cosmetic preparations for topical application
EP2231190B1 (en) Particulate drug carriers as desensitizing agents
KR20060088906A (en) Improved formulation for topical non-invasive application in vivo
JPH10316555A (en) Polymer compound-containing liposome external preparation
AU2019301542A1 (en) Methods to reduce complications of intra-articular steroid

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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