CA2644471A1 - Pro-nanodispersion for the delivery of cyclosporin - Google Patents

Pro-nanodispersion for the delivery of cyclosporin Download PDF

Info

Publication number
CA2644471A1
CA2644471A1 CA002644471A CA2644471A CA2644471A1 CA 2644471 A1 CA2644471 A1 CA 2644471A1 CA 002644471 A CA002644471 A CA 002644471A CA 2644471 A CA2644471 A CA 2644471A CA 2644471 A1 CA2644471 A1 CA 2644471A1
Authority
CA
Canada
Prior art keywords
formulation
composition
surfactant
hlb
pharmaceutically effective
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
CA002644471A
Other languages
French (fr)
Inventor
Abraham J. Domb
Avi Avramoff
Victor Pevzner
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.)
Dexcel Ltd Israel
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2644471A1 publication Critical patent/CA2644471A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0048Eye, e.g. artificial tears
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/12Cyclic peptides, e.g. bacitracins; Polymyxins; Gramicidins S, C; Tyrocidins A, B or C
    • A61K38/13Cyclosporins
    • 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
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • A61K9/1075Microemulsions or submicron emulsions; Preconcentrates or solids thereof; Micelles, e.g. made of phospholipids or block copolymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4808Preparations in capsules, e.g. of gelatin, of chocolate characterised by the form of the capsule or the structure of the filling; Capsules containing small tablets; Capsules with outer layer for immediate drug release
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4858Organic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4866Organic macromolecular compounds
    • 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
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • 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
    • A61P11/00Drugs for disorders of the respiratory system
    • 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
    • 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
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • 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
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • 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
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • 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

Abstract

A novel cyclosporine formulation, which is a pro-nanodispersion at room temperature, featuring solid particles of a relatively large particle size (at least about 150 nm) and yet which is a microdispersion at body temperature.

Description

PRO-NANODISPERSION FOR THE DELIVERY OF CYCLOSPORIN
FIELD OF THE INVENTION
The present invention is of a pro-nanodispersion preparation for the delivery of cyclosporin, and in particular, of a pro-nanodispersion preparation which provides a delivery system with high bioavailability of cyclosporin and related substances, for exainple for ophthalmic administration.

BACKGROUND OF THE INVENTION
Many dispersion systems are currently in use as, or being explored for use as, carriers of substa.nces, particularly biologically active compounds. These systems are designed to protect the substance from the environment during delivery and to provide a controlled release of the substance to a targeted area. In some cases, the goal is to target specific sites in the body using the dispersion. In other cases, tlie goal is to prepare a drug carrier system that acts as a reservoir at the site of injection.
Dispersion systems used for pharmaceutical and cosmetic formulations can be categorized as either suspensions or emulsions. Suspensions are defined as solid particles ranging in size from a few naiiometers up to hundreds of microns, dispersed in an aqueous or nonaqueous inedium using suspending agents. Solid particles include microspheres, microcapsules, and nanospheres.
Emulsions can be defined as dispersions of one liquid in another, stabilized by an interfacial film of emulsifiers such as surfactants and lipids. Despite their long history, emulsions are used less often today than many other dosage forms due to the inherent instability. Emulsion formulations include water in oil and oil in water emulsions, multiple water/oil/water emulsions, microemulsions, microdroplets, and liposomes.
A microeinulsion is .a transparent or substantially transparent emulsion which is formed spontaneously or substa.ntially spontaneously wlien its components are brought into contact.
Microemulsions are thermodynamically stable and contain dispersed particles or droplets of a size less than about 200 nm. Generally microemulsions feature droplets or particles having a mean diatneter of less than about 150 mn. These particles may be spherical, although otller structures are feasible, such as liquid crystals with lamellar, hexagonal or isotropic sym.metries. Microemulsions are usually stable over periods in excess of 24 hours.
Microemulsions can also be used as a"micxoemulsion preconcentrate", which is a composition which spontaneously forms a microemulsion in an aqueous medium, for example in water, upon dilution, or in the gastric juices after oral application. Dilution of the microemulsion in water can be for example from about 1: 1 fold to about.l:
10 fold dilution.
As noted above, while emulsion based delivery systems are useful for certain applications, the delivering.vesicles are subject to physical rupture because of the delicate nature of the liquid/membrane/liquid structure. Emulsion based delivery systems also have relatively short release times. Further, it is difficult to isolate emulsion based vesicles from the aqueous media used for storage for subsequent reconstitution.
In spite of these difficulties, microemulsions have been the only successful deliveiy sys"terns fof certain Cypes "of pharmaceutical "compourids,"particularly compousids such as members of the cyclosporin class, which are cyclic oligopeptides. The cyclosporin class includes substances having pharmaceutical utility, for example as immunosuppressive agents, anti-parasitic agents and agents for the reversal of multi-drug resistance, as known and described in the art. Examples of such cyclosporins include, but are not limited to, Cyclosporin A (also known as and referred to herein as "Ciclosporin"), Cyclosporin G, [0- (2-hydroxyethyl)- (D) Ser] 2- Ciclosporin and~[3'-deshydroxy-3'-lcet-MeBmt] '- [Val) 2-Ciclosporin.
The first of the cyclosporins to be isolated was the naturally occurring fungal metabolite Ciclosporin (Cyclosporine). Ciclosporin is the cyclosporin of formula (1):
MeBmt-aAbu-Sar-MeLeu-Val-MeLeu-Ala-(D)Ala-MeLeu-MeLeu-MeVa1 wherein-MeBmt-represents the N-methyl- (4R)-4-but-2E-en-l-y 1-4-methyl- (L) threonyl residue of formula (II):

SUBSTITUTE SHEET (RULE 26) ~

x ~H2 HO`R jCH
CH (R)\CHg I ' -N-CH-CO-CH (s) in which x-y is CH=CH- (trans). Ciclosporin is well known as an immunosupressive agent. In addition, Ciclospoiriri is being examined for the treatment of autoimmune and inflammatory diseases.
Since the original discovery of Ciclosporin, a wide variety of naturally occurring cyclosporins have been isolated and identified. Many further non-natural cyclosporins have been prepared by total-or semi-synthetic means or by the application of modified culture techniques. The class comprising the cyclosporins now includes, for example, the naturally occurring cyclosporins A through Z[c. f. Traber et al. Helv. C'hir.
Acta. 60:
1247-1255.1977; Traber et al.. Hel v. Chim. Acto. 65: 1655-1667.1982: Kobel el nul., Europ.. J. App. Microbio. and Biotech., 14: 273-240 (1982) : and von Wartburg el al., Progress in Allergy, 38: 28-45 (1986)], as well as various non-natural cyclosporin derivatives and artificial or synthetic cyclosporins including: the so-called dihydro-cyclosporins, in which the moiety x-y of the -MeBmt- residue in Formula (II) above is saturated to give x-y of -CH2-CH2- ; derivatized cyclosporins (e. g. in which a further substituent is introduced at the a-carbon atom of the sarcosyl residue at the 3- position of the cyclosporin molecule); cyclosporins in which the -MeBmt- residue is present in isoineric form (e. g. in which the configuration across positions 6'and 7'of the-MeBmt-residue is cis rather than trans); and cyclosporins in which variant amino acids are = 20 incorporated at specific positions within the peptide sequence. Many of these members of the cyclosporin class exhibit pharinaceutical utility which may be comparable to that of Ciclosporin.
Unfortunately, many difficulties have been encountered in the effective administration of Ciclosporin difficulties which appear to be inherent in the nature of the members of the cyclosporin class. Cyclosporins are characteristically highly hydrophobic and thus require a lipophilic carrier. The selection of a suitable carrier is particularly critical for the ad.ininistration of cyclosporins, as the bioavailability of these coinpounds is SUBSTITUTE SHEET (RULE 26) lalown in the art to be highly variable, depending upon the properties of the carrier.
Furtherniore, these coinpounds are lcnown to have bioavailability which may vary significantly between individuals. Such variation is particularly dangerous given the side effects of cyclosporins, such as nephrotoxicity. Thus, the suitable carrier inust provide good bioavailability of cyclosporins wliich is substantially consistent between individuals.
Absorption and metabolism of Cyclosporin are highly variable from patient to patient. Following oral administration, the elimination of Cyclosporin is primarily biliary, with only 6% of the dose (parent drug and metabolites) excreted in urine. The disposition of the orally administered drug from blood is generally biphasic with a terniinal half life in the range of 5-18 hours. The cyclosporine relationship between the administered dose and exposure is linear within the therapeutic dose range.
Following oral administration the Tmax ranges from 1.5 - 2.0 hours and administration of food is known to shows a slight decrease in AUC and Cmax.

The drug is extensively metabolized by cytocllrome P450-3A present in the liver and to a lesser degree by the CYP-3A in the gut and kidney. The drug is also a substrate for the P-glycoprotein (PGP). At least 25 metabolites have been identified in huinan bile, feces, blood and urine. The inununosuppressive activity is primarily due to the parent drug (Physician Desk Reference 59fl' edition. Tliomson NJ, 2005; 2346-2353).
As noted previously, cyclosporins may be administered with a microemulsion carrier. According to formulations of such carriers that are lcnown in the art, the carrier generally contains a hydrophilic solvent, such as liquid PEG200-600 ethylene or propylene glycol, ethanol or propanol, Glycerin, water soluble fatty acid C6-C18 esters of sucrose, dimethylisosorbide, ethyl-acetate, glycofurol (fatty acid derivative of a cyclic poly.ol), PEG derivatives of tocopherol, or PEG-fatty acid esters; a surfactant such as Tween 20, various PEG (polyetliylene glycol) derivatives or-phospholipids; a water insoluble oil such as corn oil aild other oils from plants and mixtures of oils; and Creinophor and similar PEG derivatives of castor oil or other fats which are used as an amphiphilic solvent, einulsifier, surfacta.nt and so forth. Unfortunately, none of these baclcground art formulations provides high bioavailability for cyclosporin.
The currently commercially available forinulation is disclosed in U. S. Patent No.
5,342,625_to Sandoz A. G. This formulation includes a liydrophilic phase, a lipophilic phase and a surfactant. The hydrophilic phase could be a C 1-5 alkyl di-or partial-ether of a inono-or poly-oxy-C2 2alkanediol, for exainple.

PCT Application No. WO 96/13273 to Sandoz describes compositions for cyclosporin and other macrolide drugs such as Rapaniycin, containing a hydrophilic phase which includes diinethylisosorbide and/or a lower alkyl alkanoic ester, a lipophilic phase and a surfactant. The particle size after dispersion can be 200 nm but is preferably 5 100 iun or less. The lZydrophilic phase is PEG, propylene glycol and glycofurol or dimetliylisosorbide (a bicyclic ether). The bioavailability of a coinposition containing cyclosporin and the carrier is not disclosed. PCT Applicatioii No. WO
97/19692, also to Sandoz, describes compositions which are based on PEG-derivatives of saturated hydroxy fatty acids such as PEG-hydroxystearate and a low alcohol such as ethanol or propylene glycol. Again, the bioavailability of such a composition is not disclosed. PCT Application No. WO
98/33512 to Novartis describes compositions for oral administration of cyclosporin which do not contain oil. Instead, these coinpositions contain a surfactant with HLB
10 or higher and a hydrophilic phase whiclz is polyethylene glycol and/or a lower alcohol (not more than 12%). The formulations are preconcentrates which provide a particle size of 10 to 150 nm upon dispersion. The disclosed advantage of these compositions is their ability to be stably contained witliin a hard capsule. However, no specific data is disclosed that is related to the bioavailability of cyclosporin wit11 this composition. As noted above, the .
bioavailability of cyclosporin is known to be highly variable, depending upon the carrier.
PCT Application No. WO 97/04795 to POLI Industria describes compositions - that must contain one polymer,linear, or cross-linlced PEG and poly (acrylic) or mixtures thereof and monoesters of fatty acids with a short alcohol. Again, the bioavailability of such a coinposition is not disclosed.
U. S. Patent Nos. 5,603,951 and 5, 639, 474 to Haruni Pham. describe compositions of dimethylisosorbide as a cosurfactant and a primary alcohol, mediu.m chain triglycerides and a surfactant having a HLB value of 10 to 17 such as Tween 20, formulated in soft gelatin capsule. The particle size is about 100 rmi. Again, the bioavailability of such a composition is not disclosed. U. S. Patent No.
5,583,105 to Biogel describes cyclosporin formulations coinposed of PEG esters of tocopherol and a lipophilic solvent, an amphiphilic solvent and ethanol. Again, the bioavailability of such a composition is not disclosed.
U. S. Patent No. 5,614,491 to Dr. Rentschler GmbH describes formtilations of PEG fatty acid inonoesters as emulsifying agent and a polyol as solvent. U. S.
Patent No.
5,798,333 to Sherman describes formulations composed of Tocophersolan and a polyhydric alcohol. Tocophersolaii is a water soluble surfactant which dissolves cyclosporin only at a 7: 1 ratio. U. S. Patent No. 5,827,822 to Sangstat describes forinulations of alcohol and a PEG surfactant forming particle size between 200 and 400 nm.
Europeaii Patent Application No. EP 0760237 Al to Cipla describes a coniposition containing: vegetable oil triglycerides (castor, peanut, or coconut oil), phospholipid, a surfactant (Tween 20, polyoxyl-40-hydrogenated castor oil) and a 1lydropllilic solvent, propylene glycol.
Japanese Patent Application No. 61-249918 to Yutaka Mizushima describes a cyclosporine formulation featuring eyedrops. The eyedrops use oils such as soy bean oil, cottonseed oil, sesame oil, sunflower oil, corn oil, a squalene, an eicosapetaenoic acid and its ester, azone. The solvents disclosed are glycerol and water.
Swiss Patent No. 641356 describes a cyclosporine forinulation which features a transesterification product of a triglyceride with a polyalkylene glycol.
These products can be transesterification products of two molar fractions of a triglyceride of a natural oil, sucll as corn oil, almond oil, peanut oil, olive oil and/or palm oil, which are again vegetable oil triglycerides.
Again, the bioavailability of cyclosporin administered with such a composition is not disclosed.
None of these disclosed background art caiTier formulations features an organic solvent which is a lower alkyl ester of hydroxyallcanoic acid, such as etllyl lactate.
Moreover, none of these disclosed background art carrier formulations features a combination of a surfactant with higli HLB and a surfactant with low HLB.
Furthermore, none of these baclcground art carrier formulations is disclosed as having high bioavailability. Furthermore, none of these background art carrier formulations is disclosed as having a solid fat as a core component wlizch results in a dispersion when mixed with aqueous media at room temperature.

SUMMARY OF THE INVENTION
There is thus an uiunet need for, and it would be useful to have, a composition for the administration of cyclosporins, particularly for oral administration, which would provide a high bioavailability, and which would preferably contain an organic solvent whicli is a lower allcyl ester of 1lydroxyalkanoic acid and a surfactant which is preferably a combination of a surfactant with high HLB and a surfactant with low HLB and which may contain a solid fat.
The present invention overcoines these deficiencies of the background art by providing a novel formulation for the administration of a cyclosporin.
Unexpectedly, the inventor has found that the novel formulation, wllich is a pro-nanodispersion at room temperature, featuring solid particles of a relatively large particle size as described in greater detail below, can become a microdispersion at body temperature. Furthermore, the inventor has discovered that surprisingly the foi7nulation, when stored in soft gelatin capsules at room temperature, is stable for more than 2 years, maintaining the same coinposition and particle size. Furtherinore, the novel formulation provides excellent bioavailability characteristics as described in greater detail below.
The forinulation according to the present invention features an ainphiphilic solvent which is characterized by being a lower alkyl ester of hydroxyalkanoic acid; a surfactant, preferably a combination of a surfacta.nt with a high HLB
(hydrophilic/lipophilic balaiice) and a surfactant with a low HLB; and optionally and preferably coinprising a solid fat. The solid fat is a fat which is solid at room temperature but which preferably melts at a teznperature of at least about 30 C and wliich more preferably melts at body temperature, such as a fatty acid ester. Preferably, the solid fat comprises a triglyceride such as tricaprin. Otlier non-limiting examples of suitable solid fats incfude trilaurin, fatty acids and fatty alcohols of 10 carbons or more, esters of fatty acids such as ethyl stearate and hydrogenated triglycerides that are solid at room temperature and melt at a temperature of at least about 30 C, more preferably melting at body temperature.
Optionally and more preferably, there is a large difference between the HLB of the low HLB surfactant and that of the high HLB surfactant, such that the low HLB
surfactant preferably has an HLB of,less than about 6, while the high HLB
surfactant preferably has an HLB of greater than about 10. More preferably, the low HLB
surfactailt has an HLB of less than about 4, wliile the high HLB surfactant has an HLB of greater than about 14. The high HLB surfactant preferably comprises Polysorbate 20 (for example Tween 20) and the low HLB surfactant preferably coinprises Sorbitan oleate (for example Span 80). Polysorbate 20 has an HLB of 16.7, while Sorbitan oleate has an HLB
of 4.3.

~
The fornlulation also preferably features a phospholipid and an ethoxylated fat such as Cremophor or Poloxamers wliich are block copolymers of polyethylene glycol and polypropylene glycol.
The preferred mean diameter of the particle of the resultant formulation is preferably greater t11an about 150 nun at room temperature (25 C) but is preferably less than about 100 mil, more preferably less than about 60 nm, and most preferably from about 5 xun to about 50 mn froin at least about 33 C; such that this particle size is achieved at body temperature. Hereinafter, the term "pro-nanodispersion"
includes those compositions featuring droplets or particles having a mean diameter of less than about 150 nm, and which spontaiieously forms a nanodispersion or microdispersion in a1i aqueous medium, for example in water upon dilution, or in the gastric juices after oral intalce. Dilution of the pro-nanodispersion in water can be for example from about 1: 1 fold to about 1: 1000 fold dilution. Hereinafter, the term "nano-dispersion"
refers to a dispersion in an aqueous medium of particles or vesicles of 100 nanometers or less at body temperature.

BRIEF DESCRIPTION OF THE DRAWINGS
The invention is herein described, by way of example oiAy, with reference to the accompanying drawings, wherein:
FIG. 1 shows the mean percent of drug (CsA) released versus time for test (Dexcel Ltd) (o) and reference Neoral 100 mg from Novartis (m) using USP 24 method.
Both test and reference products exhibited very similar dissolution profiles;
and FIG. 2 shows the mean CsA concentration-time profiles after single oral administration of 200 mg of the drug given as soft gelatin capsules test (o) (Dexcel Ltd.) and known drug reference (Novartis Inc.) (a) to 24 healthy volunteers.

DESCRIPTION OF THE PREFERRED EMBODIMENTS
The present invention is of a novel forlnulation for the administration of a cyclosporin.
This forinulation features an amphiphilic solvent which is characterized by being a lower alkyl ester of hydroxyallcanoic acid; and a surfactant, preferably a combination of a surfactant with a high HLB (hydrophilic/lipophilic balance) and a surfactant with a low HLB. Optionally and preferably, the formulation fui-ther features a solid fat that is solid at room temperature but preferably melts at a temperature above 30 C. The high HLB
surfactant preferably comprises Polysorbate 20 (for example Tween 20) and the low HLB
surfactant preferably comprises Sorbitan oleate (for exainple Span 80). The amphiphilic solvent is preferably etliyl lactate. Etllyl lactate is an anlphiphilic solvent characterized by having an octanol/water coefficient of 0.06. The solid fat is preferably a fatty acid ester and is more preferably a triglyceride.
The formulation optionally and preferably comprises a phospholipid. Optionally and more preferably, the forinulation coznprises an ethoxylated fat such as Cremophor or another similar substance.
Hereinafter, the terms "solid fat" and "liquid fat" refer to fats which are solid or liquid, respectively, at room temperature. Preferably, the coinposition of the present invention does not include an alkyl alcohol such as ethanol.
The preferred particle size of the resultant formulatioil from at least about 30 C is less than about 100 nm, more preferably less than about 60 nrn, and most preferably from about 5 zun to about 50 run. In fact, as described in greater detail below, the resultant formulation must have a particle size of less tha.n about 100 iun at body temperature in order to be suitable for the administration of cyclosporin. Surprisingly, it was discovered that the particle size at rooin temperature of the forinulation of the present invention is at least about 150 iun, as the forinulation forms a solid microdispersion, yet contrary to the teachings of the art, is still able to achieve a suitable particle size upon administration to a subj ect.
Furthermore, surprisingly the inventor has discovered that the particle size at room tenlperature does not control bioavailability, wllich is contrary to the teachings of the baclcground art. It is possible to have forinulations with the taugllt particle size at room teinperature that exhibit poor bioavailability, while other formulations (such as the formulation of the present invention) may have particle sizes greater than that taught by the background art, yet may still have excellent bioavailability. Also in direct contradiction to the teachings of the background art, the pro-nanodispersion does not spontaneously form a microdispersion upon dilution in water of the desired particle size without the application of heat (namely the presence in the body of the subject to obtain particle size of less than 100 nanometers, as body ternperature is about 37 C).
As described in greater detail below, the combination of these coinponents has unexpectedly been shown to provide higher bioavailability than had been previously shown for forinulations of cyclosporin. Furtliermore, the forinulations of the present invention have the advantage of not requiring stabilizers, such as anti-oxidants in order to obtain good stability characteristics. Without wishing to be limited to a single mechanism, it is hypothesized that the excellent stability of the formulations of the present invention is 5 due to the use of solvents such as et11y1 lactate as described in greater detail below.
Ethyl lactate, and otlier members of this family of solvents, have unexpectedly good properties for such a forinulation as the forinulations of the present invention. For exainple, ethyl lactate is miscible in both organic and aqueous solvents, since it is more hydrophobic than ethanol. Ethyl lactate has higher storage stability than ethanol. Ethanol 10 is a highly volatile solvent, with correspondingly lower storage stability, such that the use of ethanol in the currently available baclcground art formulations is a clear disadvantage of these forinulations.
Furthermore, these background art formulations require a conlbination of ethanol and propylene glycol in order to stabilize the alcohol, which is another disadvantage of incorporating ethanol into a formulation, a disadvantage which is overcome by the formulations of the present invention.
The present invention may be more readily understood with reference to the following illustrative examples. It should be noted that reference is made generally to "cyclosporin", indicating any member of the cyclosporin class having pharmaceutical efficacy. The particularly preferred meinber of the cyclosporin class is Ciclosporin (Cyclosporin A). The preparation of.the pro-nanodispersion compositions of the present invention is described first with reference to the following general description and then with reference to the following non-limiting exainples of the preparation and application of the compositions of the present invention.
Amphiphilic Solvent First, as noted previously, a suitable organic solvent must be selected. The solvent is preferably selected from the family of lower allcyl esters of hydroxyallcanoic acid.
Hereinafter, the term "lower alkyl" includes C 1 to C4, for example etliyl.
The preferred amphiphilic solvents of the present invention are CI-4 alkyl-hydroxy alkanoic acid ester.
More preferably, the amphiphilic solvent coinprises etlryl lactate.
Ethyl lactate (2-hydroxypropanoic acid ethyl ester), is a colorless liquid which is miscible with water, alcohol and etller. Ethyl lactate is considered to be suitable for hunlan administration with an LD50 whicll was higher than 5 g/lcg in mice when given an oral dose; however, suiprisingly etliyl lactate was not previously taught as a suitable ingredient for pharinaceutical compositions.
Ethyl lactate is amphiphilic and therefore possesses a number of cllaracteristics.
For example, the diffusion of ethyl lactate froin an organic solution into water is much slower and controlled con2pared to highly hydrophilic solvents such as ethanol or propanol. This is an iinportant feature as a fast diffusion may result in inunediate precipitation when exposed to aqueous media and in the other hand, a solvent which diffuses too slowly may not form the desired particle size as the fonned droplets are not spontaneously forined.
Witllout wishing to be limited by a single hypothesis, one important parazneter for the behavior of the formulation of the present invention is the rate of diffusion of the solvent from the hydrophobic mixture of the pro-nanodispersion, as the rate of diffusion determines the particle size and composition of the forined particles. Again without wishing to be limited by a single hypotllesis, a rapid rate of diffusion will result in precipitation of large particles from the aqueous medium, while a slow rate of diffusion from the pro-nanodispersion into the stomach liquid may result in iinproper par-ticle formation and precipitation of large particles, or alternatively may cause the cyclosporin to be poorly integrated within the solid fat carrier of the present forinulation. Also, it may cause one or more components in the stomach fluid to interfere in the process of particle formation. Thus the rate of diffusion from the oil droplet into the aqueous iriedium is a very important feature, as it must be neither too rapid nor too slow. The diffusion rate is related to the partition coefficient.
The partition coefficient of ethyl lactate is exactly on the border between hydrophobicity and hydrophilicity, with a value of 0.06, which means that 50%
of the solvent is in the octanol hydrophobic phase and 50% is in the water phase.
Tlierefore, the diffusion rate is neither slow nor rapid, but in fact is exactly in the middle.
From the chemical view point, ethyl lactate combines the properties of an alcohol and an ester wllere the ester group provides hydrophobicity while fihe hydroxyl provides the hydrophilicity and the ability to forin liydrogen bonding with water.

Surfactant Second, a suitable surfactant is preferably selected, which is preferably a combination of a surfactant with a higll HLB (hydrophilic/lipophilic balance) of at least about 10 and a surfactant wit11 a low HLB of less than about 6. The terin "HLB" refers to the 1lydrophilic/lipophilic balance of a surfactant. A surfactant with high HLB is hydrophilic, while a surfactant with low HLB is liydrophobic.
Therefore, the combination of a surfactant with high HLB and a surfactant with low HLB, as is preferred for the coinpositions of the present invention, is actually a combination of a hydrophilic surfactant and a hydrophobic surfactant. This combination has never been taught or suggested in the baclcground art as being suitable for a pharmaceutical carrier for cyclosporins. Wliere the HLB of the surfactant has been specified in the background art, it has been given in the range of 8 to 20, which is clearly different from the combination of surfactants taught herein. Thus, the compositions of the present invention can be clearly differeiitiated from those taugllt in the background art on the basis of the preferred combination of a surfactant with a low HLB and a surfactant with a high HLB.
Particularly preferred combinations of these surfactants feature a large differeilce between the HLB of the low HLB surfactant and that of the high HLB surfactant.
The high HLB surfactant preferably comprises Polysorbate 20 (for example Tween 20) and the low HLB surfactant preferably comprises Sorbitan oleate (for example Span 80). Of course other such combinations could be also be used.
Span hydrophobic surfactants are a group of sorbitan fatty acid esters suc11 as sorbitan monooleate, sorbitan monopalmitate, sorbitan monostearate, sorbitan tristearate, sorbitan monooleate, sorbitan trioleate and sorbitan monolaurate (Fiedler, H.
P.,"Lexikon der Hilfsstoffe fur Pharmazie, Kosmetic und Angrenzende Gebiete", Editio Cantor, D-7960 Aulendorf, 3rd edition, 1989, pages 1139-1140). Span 80 is an example of a low HLB surfactant, with an HLB of 4. 3, and is sorbitan monooleate. They are cornmercially available froin various producers, which include but are not limited to, Capital City Products, Croda Chem, ICI, Lippo Chem. and Atlas, under various coinniercial names:
ArlacelTM, Arinotan, Crill, Emsorb, Liposorb, Protachem, and Sorbester TM.
Exainples of suitable surfactants froin this group witli HLB values given in parentheses are as follows: Span 60 (4.7), Span 65 (2.1). Span 80 (4.3), Span 85 (1.8), Arlace183 (3.7), Arlacel 85 (1.8), Arlacel 80 (4.3), and Arlacel 60 (4.7). These molecules are generally soluble in oil. They are also soluble in most organic solvents. In water they are generally insoluble but dispersible. Other low HLB surfactants include but are not limited to PEG-6 glyceryl monooleate (HLB of about 3 or 4), and propylene glycol laurate (HLB
of 4).
Tween hydrophilic surfactants (Polysorbates) are a family of PEG sorbitan esters (polyoxyethylene-sorbitan-fatty acid esters), for example mono-and tri-lauryl, palmityl, stearyl and oleyl esters of the type luiown and commercially available under the trade nazne Tween (Fiedler, H. P.,"Lexikon der Hilfsstoffe fur Pharmazie, Kosmetic und Angrenzende Gebiete", Editio Cantor. D-7960 Aulendorf. 3rd edition, 1989, pages 1300-1304). Tween 20 (polyoxyethylene (20) sorbitan monolaurate) has an HLB of 16.7. Other types of Tween surfactants may also be useful for the compositions of the present invention.
Tween surfactants are soluble in water but not in oil. The chemical structure of this family of surfacta.nts features one, two or three short PEG chains, generally of about 5 to 20 ethylene glycol units, connected by an ester bond to sorbitan. These surfactants are produced by various companies (Croda, ICI, Sandoz, Mazer, Atlas) aiid may appear under various trade names, besides Tween: SorlateT"", MonitanTM, CrilletT" and so forth. Members of this fainily which are polysorbates 20, 21, 0, 60, 61, 65, 80 and 85 have an HLB between 11 and 16.7, and therefore would be suitable for the present invention as higll HLB surfactants.
Other suitable high HLB surfactants may be obtained from manufacturers such as Gattefosse Ltd., and include but are not limited to, sucrose fatty acid esters such as saccharose monopahnitate (HLB of 15) and saccharose monostearate (HLB of 11) or PEG-32 glyceryl laurate (HLB of 14). Suitable high HLB nonionic surfactants are polyethylene glycol (PEG) nalkanol esters of the Brij family such as Brij and 99 which have an HLB in the range of 12.4 to 16.9. Brij 56 is polyoxyethylene [10]
cetyl ether and is an example of such a high HLB surfactant which can be substituted for Tween 20. Brij 5 6 has an HLB of 12.9.

Phospholipid According to preferred embodiments of the present invention, the formulation fiu=ther coinprises a phospholipid. A phospholipid is a phosphorylated diacylglyceride molecule or its derivative. The parent structure is diacylglycerol phosphate, or phosphatidic acid. Phosphatidyl choline (lecithin) is the choline ester of phosphorylated diacylglyceride. Synthetic lecitlzins are available with acyl chain lengths ranging from 4 to 19 carbons. The preferred lecithins for biological applications are those witli alkyl chain lengths in the biological range (10 to 18 carbons). Naturally occurring lecitliin can be obtained from a variety of sources such as egg, bovine heart, or soy bean.
Unsaturated lecithins (dioleoyl; dilinoleoyl; alpha-palmitoyl, beta oleoyl; alpha palmitoyl, beta linoleoyl; and alpha oleoyl, beta palmitoyl), dianachidonyl lecithin (highly unsattirated and a prostaglandin precursor), and alpha palinito beta inyristoyl lecithin are also available.
Certain phospholipids, such as phosphatidic acid, phosphatidyl serine, phosphatidyl inositol, cardiolipin (diphosphatidyl glycerol), and phosphatidyl glycerol, can react with calciuin in serum, causing aggregation or the binding of lipospheres to cell membranes.
These unfavorable reactions can be minimized by conibining these phospholipids witli noncalcium binding phospholipids such as phosphatidylcholine.
Phosphatidic acid can be isolated from egg or prepared synthetically (dimyristoyl, dipalmitoyl and distearoyl derivatives are available from Calbiochem). Bovine phosphatidyl serine is also available commercially (Sigma Chemical Co. St. Louis, Mo.). Phosphatidyl inositol can be isolated from plant or bovine sources. Cardiolipin can be purified from bovine or bacterial sources. Phosphatidyl glycerol can also be purified from bacterial ferinentation.
Solid Fat Yet another optional ingredient is a solid fat, preferably a fatty acid ester such as a triglyceride. A non-limiting exa.mple of such a triglyceride is tricaprin.
Tricaprin is a hydrophobic triester of glycerol and caproic acid. Tricaprin does not dissolve in water and thus remains as a component of the dispersed cyclosporin-loaded particles after dispersion in aqueous solution. Tricaprin solubilizes cyclosporin in a fatty inedium which is dispersed by the hydrophilic-hydrophobic dispersing ageiits. Other such fatty components which are suitable as replacement for tricaprin include, but are not limited to, pure and mixed alkyl esters of fatty acids and mixtures thereo Exainples include but are zaot limited to ethyl esters of fatty acids such as etliylstearate and ethylpalmitate triglycerides such as trilaurin and mixtures of solid fatty acid esters.
Mixtures of fats include hydrogenated vegetable oils. The preferred fats are those that solubilize cyclosporin witli a melting point between 25 and 37 C such that the resultant pro-nanodispersion formulation forms a microdispersion of solid particles which melt into a micro-dispersion at body temperature.

The following specific examples illustrate various aspects of the present 5 invention, and are not intending to be limiting in any way. For all experiinents described below, unless otherwise stated, the particle size of the pro-nanodispersion was measured with an N4-Coulter particle size analyzer, suitable for subnlicron particle size determination. Three drops of the pro-nanodispersion were added to five nlilliliters of water. The particle size of the pro-nanodispersion did not change when the pro-10 nanodispersion was dispersed in five milliliters of HCl solution. The member of the cyclosporin class which was used for the experiments described below was Ciclosporin (Cyclosporin A).

Example 1 15 Preferred Foi~rnulation This Example describes a preferred but illustrative formulation according to the present invention. Preferably, the formulation features Ciclosporin as the active ingredient (this Exaanple inchides 100mg of the active ingredient, but as described herein, other dosages may optionally also be provided). The solvent comprises ethyl lactate. In this preferred embodiment, the formulation features a solid fat, which in this Example is triglyceride (such as Tricaprin for example). The formulation also features a combination of a low HLB solvent a.nd a high HLB solvent as described herein; for this Example, the combination comprises Polysorbate 20 and Sorbitan oleate. Preferably, the formulation features a phospholipid (in this Exainple, preferably lecithin). Also preferably, the formulation features an ethoxylated fat such as Cremophor for example.

Table 1 Ingredient _4uantitXper Capsule (ing) Ciclosporin 100.00 Polysorbate 20 168.00 Sorbitan oleate 168.00 Lecithin 84.00 Triglyceride 168.00 Ethoxylated hydrogenated castor oil 168.00 ( olyoxy140 hydro castor oil) Ethyl lactate 332.00 Fill weight 1188 For this formulation, particle size upon mixing 1 ml of the pro-nanodispersion with 10 ml of water at body temperature was found to be 30 mmn. No effect was found upon varying the pH of the water from about 2 to about 10.

Exainple 2 Effect of Solvent on Particle Size An exeinplary coinposition containing Ciclosporin, solvent, TRC (tricaprin), egg phospholipid (Avanti, USA), Tween 20, Span 80 and Cremophor was prepared with increasing ainounts of ethyl lactate, as given in Table 1 (all ainounts of ingredients are given in milligrams). The effect of adding increasing amounts of these ingredients to the composition of the present invention on (mea.n) pa.rticle size is also given in Table 2.
Briefly, all compositions which contained ethyl lactate had a particle size of less than 100 zun at a temperature of 37 C. The particle size decreased as the amount of ethyl lactate was increased.

Table 2: Effect of Solvent on Particle Size Ingredient Formulation Number Ciclosporin 100 100 100 100 Ethyllactate 0 100 200 400 phospholipid 70 70 70 70 Tween 20 270 270 270 270 Span 80 100 100 100 100 Creinophor 300 300 300 300 EL

Particle size 189 92 42 28 Exainple 3 Effect of Surfactant on Particle Size An exeinplary composition containing Ciclosporin, egg phospholipid (95% pure from Avanti, USA), ethyl lactate as a solvent, Tween 20 and Cremophor was prepared with increasing amounts of Span 80, as given in Table 3(a1l amounts of ingredients are given in milligrams). The effect of adding increasing amounts of Span 80 to the composition of the present invention on (inean) par-ticle size is also given in Table 3.
Briefly, the compositions provided a liquid solution. When dispersed in deionized water at 37 C , all compositions which contained Span 80 had a particle size of less than 100 nm. The particle size decreased as the amount of Span 80 was increased.

Table 3: Effect of Surfactant on Particle Size Ingredient Forinulation Nuinber Ciclosporin 100 100 100 100 100 Ethyllactate 300 300 300 300 300 phospholipid 50 50 50 50 50 Tween 20 200 200 200 200 200 Span 80 0 50 100 200 300 Cremophor 400 400 400 400 400 EL

Particle size 155 88 54 32 28 Example 4 Effect of Other Ingredients on Particle Size Different compositions containing Cyclosporin were prepared as described in Table 4 (all amounts of ingredients are given in milligrams). The effect of these ingredients on the particle size of the pro-nanodispersion solution when dispersed in warm water at 37 C is also given in Table 4. Briefly, compositioiis which had both low and high HLB surfacta.nts (such as Tween and Spaii) had a particle size of less than 100 mn at this temperature (which is body teinperature). Tween and Cremophor can be substituted for each otller as high HLB solvents (HLB > 10) but a certain ainount of either surfactant is required to obtain a suitable particle size, dependiiig upon the quantities of the other components. A conlbination of Tween 20 and Cremophor permits the use of reduced arnounts of each ingredient, as described in greater detail below. In addition, the presence of a solvent such as etllyl lactate is required. A solid fat such as tricaprin is also optional. The presence of a phospholipid is also preferred to obtain a particle size in the range of 30 nm, althougll the particle size remained below 100 nin even without the phospholipid as for Fonnulation 3, in wh.ich no phospholipid was added but the particle size was 95 run at 37 C.

Table 4: Effect of Other Ingredients on Particle Size Ingredient Formulation Number Ciclosporin 100 100 100 100 100 100 100 100 100 100 Etliyllactate 400 200 400 400 400 400 400 600 400 400 phospholipid 100 100 0 100 100 100 100 100 100 100 Tween 20 200 200 200 200 0 200 200 200 400 0 Span 80 200 200 200 0 200 200 200 200 200 200 Cremophor 200 200 200 200 200 200 0 200 0 400 EL
Particle size 28 30 95 187 182 230 340 32 78 64 Example 5 Storage Stability of Preferred Formulation The coinposition of Example 1 was prepared at two different total quantities (all amounts of ingredients are given in milligrams), witli the second quantity featuring 10-fold larger amounts of each ingredient. Both compositions were easily prepared by dissolving all components to a liquid solution by mixing with mild heating (about 40 C).
Preferably, the phospholipid was first dissolved in ethyl lactate, and then all other components were added with continuous mixing, apart from Cyclosporin which was added= last. The mean particle size of the composition was measured after dispersion of different amounts of the composition in deionized water at 37 C by using the light scattering technique with a Coulter N4 particle size a.nalyzer. Both volumes of the composition had a particle size in the range of 30 run wliich is preferred This composition was used for human studies, as described in greater detail below.
The stability of the composition was tested by loading doses of 50 mg of Ciclosporin into hard gelatin capsules (size 00) or in glass containers and then storing the coinposition at room tenlperature (25 C) or at refrigeration (4 C). The particle size and the Ciclosporin content were deterznined after 3 and 6 inonths of storage. All sainples were found to have a particle size in the range between 17.2 and 32.6 at any dispersion range (3 to 20 drops per 5 ml) when ineasured at a tenlperature of 37 C. As calculated from the pealc size after analysis by HPLC (high pressure liquid clu=onlatography), the Ciclosporin content for all stored fonnulations was between the required limits of 95 to 105% of the initial concentration even after 2 years of room temperature storage.
5 Example 6 Comparison between Sandiinmune Neoral and the Present Formulation Experiments were perforined to compare the formulation of the present invention to the standard commercially available formulation for Ciclosporin (Neoral", Novartis Inc).
10 The forinulation of the present invention has excellent release properties as described in greater detail below. In addition, structural analyses were performed to conlpare the Neoral formulation with that of the present invention. These analyses showed that surprisingly the structure and behavior of the formulation of the present invention when diluted in water is very different from that of Neoral0.
15 The following approach was talcen to compare the two systems. The formulation of the present invention forms a solid microdispersion at room teinperature while Sandimmune0 Neoral0 forms an oil-in-water einulsion where the oily component is dispersed as droplets in the water phase. Without wishing to be limited by a single liypothesis, it is believed that this different behavior is related to the differences in the 20 compositions, particular in that the formulation of this invention preferably uses tricaprin (a solid fat that has a melting point of 32 C) as the hydrophobic core component of the particles, while Neoral0 formulations use corn oil (liquid at room teinperature) as the core coinponent of the particles.
To examine these differences, the following experiments were conducted. Both concentrated formulations were dispersed in water at 25 C with shaking and the resulted dispersions were analyzed by the following methods: confocal microscopy (Ziess Model 410 confocal scanning microscope) for visualization of the dispersion particles; particle size analysis of the dispersion by a Coulter particle size analyzer; isolation of the dispersed particles; and analysis by DSC for melting point deterinination of the isolated particles. Each of these is discussed in greater detail below.

Confocal Microscopy The formulation of Example 1 was prepared with the addition of 2mg of Nile Red as hydrophobic fluorescent marlcer. As a control, the nanoemulsion forinulation was prepared with the addition of 2 mg Nile red using the NeoralOO (cominercially available) formulation ingredients for the preparation of 1200 mg of forinulation. The formulations were placed on a suitable slide aa.ld examined under the Confocal microscopy at room temperature. Confocal microscopy (lens #20) picture showed particles of various sizes in the range of 0.2 to 1 microns for the forinulation of the preserit invention.
In contrast the NeoralOO formulation did not show any particles but a continuous view which indicates the presence of a nanoemulsion.

Particle Size Analysis Sample preparation: 0.5 ml of both formulations at a temperature in which they are in a liquid or "oily" form was added to 2 ml of double distilled water (25 C) and hand shaken for a few seconds until mixing was uniform. The Neoral forinulation formed a clear solution wllile the formulation of the present invention formed a milky like dispersion. In a second experiment, the formulations in oily form were dispersed in warm water (37 C) and the dispersion was viewed by microscope. Both forinulations formed almost clear solutions that do not show any particles under confocal microcopy due to its low sensitivity. The particle size of the dispersed forinulation at 25 C and at 37 C was determined using Coulter N4 particle size analyzer.
Experimental: 0.2 ml of cyclosporin oil formulations talcen fioin soft gelatin capsules of Neoral or the invented formulation were added to 5 ml double distilled water at 25 C or at 37 C and hand shaken for a few seconds to fornl a uniform dispersion.
The dispersions were analyzed for their particle size. The formulation of the present invention showed a mean particle size of 0.5 microns with most particles in the range of 0.2 to 0.8 microns when dispersed in water at 25 C while the Neoral formulation showed a particle size of 30.2 nanometers. The formulations at 37 C showed a particle size of 33.6 nanometers for the formulation of the present and 32.6 nanometers for the NeoralQ formulation.

Ultracentrifugation of the dispersions and DSC analysis of the preci ~p~tate Experimental: Both formulations, after dispersion in deionized water at 25 C
(0.5 ml in 2 ml water with gentle shaking), were centrifuged at 20 C for 20 minutes using 20,000 rpm centrifugation. The Neoral formulation remained clear and uniform while the formulation of the present invention separated in two layers, a solid layer and a cloudy solution. The solid precipitate was isolated by decantation and a second centrifugation took place under the following conditions: 45,000 rpm for 30 min at 20 C. A
precipitate was obtained and the solution became clear. The Neoral emulsion was also centrifuged a second time under similar conditions and remained clear and homogeneous with no precipitation. This Neoral0 emulsion was farther centrifuged at 100,000 rpm for 30 min at 20 C; again the emulsion remained uniform and clear.
The precipitate from the invented formulation was dried in room air over night and the pellet was analyzed by Differential Scanning Calorimeter (DSC Metier) for melting point. The melting point of the precipitate was 30.4 C which indicates that the dispersion is a suspension of solid particles at temperatures below the melting point.
The cyclosporin formulation of the present invention forms a microdispersion, when dispersed in water at ambient temperature (25 C). The foimned solid particles have a particle size greater than 0.2' microns, and included particles in a range of up to about 0.5 microns, with a melting point of 30 C. By contrast, the Neoral formulation formed a true nano-emulsion of oil droplet size of 30 nm at ambient temperature.
In Vitro Release The comparative drug release rate was assessed using a dissolution apparatus test according to the USP-24 method. Both formulations achieved an in-vitro release rate of more than 90% within 15 minutes. Methods and results are described in greater detail below.

Methods Comparison of the dissolution profiles of Ciclosporin 100 mg capsules (Dexcel, Israel) and Sandimmune NeoralOO 100 mg capsules (Novartis, Sweden) was carried out under the following conditions. The dissolution apparatus was a 2 paddle apparatus, containing 0.1 N HCL, containing 4 mg of lauryldimethylamine-N-oxide per ml as the dissolution medium. The capsules were dissolved in a volume of 1 liter of the dissolution medium, at a temperature of 37 C and a rotation speed of 75rpm. At various time points (0 min, 15 min, 30 min and 60 min), samples were withdrawn from the dissolution apparatus and were assayed with an HPLC apparatus to determine the level of ciclosporin in the sample. A standard, prepared by adding 50 mg of cyclosporine to 30 ml of ethanol and then diluting 10 ml of the resultant solution with 40 ml of the dissolution medium, was used to calibrate the HPLC results. The mobile phase featured a mixture of 0.05 M
phosphoric acid and tetrahydrofuran, at a ratio of 59:41 volume per volume.

Results Table 6 shows the amount of ciclosporin released from each formulation at the various timepoints.

Table 6 - comparison of release rates Dissolved value (%) Time (min) B.N. 161200 B.N. C43 Dexcel Novartis 15' 103 90 Similarly to Table 6, Figure 1 shows the mean percent of drug (CsA) released versus time for test (Deximune ) (o) and reference Neoral'o 100 mg (m) using method. Both test and reference products exhibited very similar dissolution profiles. Both formulations showed a very fast release rate with more than 90% of the drug released and dissolved within 15 minutes (Fig. 1). The release rates for both formulations were superimposable.

Example 7 Analysis of Preferred Formulation - In Vitro Activity The composition of Example 5 was prepared 5 times independently for 400 ing Ciclosporin. The particle size, Ciclosporin content, the morphology of the formed particles and the melting point of the particles was determined. The bioactivity of the Ciclosporin formulation on T-cells was also determined.
The particle size of all formulations ranged between 18 to 29 nm when dispersed at 37 C in deionized water or 0.1 N HCl solution. The particles were viewed by Transmission Electron Microscope (TEM) at high magnification. Spherical particles with a narrow size distribution in the range of 30 nm were observed. The melting point of the particles was determined by differential scanning calorimeter (DSC) and was found to be in a temperature range of from 30 to 35 C. The composition was highly effective at inhibiting the activity of T-cells. The results clearly indicate the superior stability, reproducibility and efficacy of the preferred formulation.
Example 8 Pharmacolcinetic Human Studies A randomized pilot pharmacolcinetic study was undertaken to investigate the pharmacokinetic performance of the composition of the present invention, when compared to the standard commercially available formulation for Ciclosporin (Neoral , Novartis Inc).
The investigation was designed as a randomized, open-labeled, two-period, two-treatment crossover study, in 24 healthy fasted male volunteers. The subjects were administered a single 200mg cyclosporine (CsA) dose of either formulation. The formulation according to the present invention that was tested is given in Example 1.
Serial venous blood samples were obtained over 24 hours after each administration to measure cyclosporin concentrations in the blood. For both treatments, a mean maximum blood concentration (Cmax) of approximately 1200ng/ml was obtained at about 1.6 hours (tmax) after administration; the area under the blood concentration-time curve (AUC) was, on average, 4900ng x hr/ml.
As a summary of these results, bioequivalence was conclusively demonstrated for both rate (Cmax and tmax) and extent (AUC) of CsA absorption, between the two treatments.

The point estimates and its 90% confidence intervals were within the respective equivalence ranges for the pharmacokinetic parameters and were included in the range for narrow therapeutic drugs index.
Methods and results are discussed in more detail below.

Methods:
In Vivo bioequivalence Study:
The study was performed with twenty-four healthy male volunteers who all 10 started and completed the study.
The study protocol was a randomized, two-treatment, two-period, crossover investigation with a washout phase of one week between the two study periods.
The treatments consisted of a single oral 200 mg dose (2 x 100 mg soft gelatin capsules) with 240 ml water.
15 Subjects were confined to the study center from 10-12 hours before unti124 hours after each drug administration. On the days of drug administration, they fasted for 12 hours before dosing until 4 hours after dosing. Thereafter they were given standard, scheduled meals that were identical on both dosing days. The time of day of drug intake was identical for a given subject for each dosing. Fluid intake was also standard, and no 20 alcohol or xanthines were allowed during the periods of confinements. Study participants were asked to refrain from the use of all drugs, including over-the-counter medications, for at least 2 weeks before the first administration, as well as during the entire study.
After each administration, venous blood samples to determine cyclosporin in whole blood were obtained before and then 0.25, 0.5, 0.75, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 6, 9, 12, 15 25 and 24 hours after dosing. Samples were collected in EDTA-containing tubes, gently inverted several times. Two whole blood aliquots were immediately frozen at -36 C.
The following bioanalytical method was used to analyze the level of Cyclosporin in the blood samples. Blood analysis was performed by a pre-validated method of Fluorescence Polarization Immu.noassay - TDx, with TDx/TDxFLx cyclosporin monoclonal whole blood assay kits (Abbott Laboratories), for the determination of drug plasma concentrations having an LOQ of 50ng/ml. The assay was used according to manufacturer instructions.

Statistical Analysis Parametric general linear model procedures are recommended for the analysis of pharmacokinetic' data derived from in vivo bioequivalence studies. An analysis of variance (ANOVA) was performed on the phai7nacolcinetic parameters AUC and Cmax, after logarithmic transformation using General Linear Models (GLM) procedures of SAS
program. Appropriate statistical models pertaining to the design of the bioequivalence study were employed.
The two one-sided hypotheses at the 5% level of significance were tested for AUC and Cmax, by constructing the 90% confidence intervals for the ratio between the test and reference averages. The range for concluding bioequivalence (AUC, Cmax) was set to 80%-125% using the logarithmically transformed data and the Tmax was evaluated by using the nonparametric 'analysis for the median difference were computed, all based on the EU and FDA guidelines.
All statistical computations were performed by the SAS software version 8.0, Cary, NC, USA.

Results and Discussion Figure 2 shows the mean CsA concentration-time profiles after single oral administration of 200 mg of the drug given as soft gelatin capsules test (^) (Dexcel Ltd.) reference (Novartis Inc.) (m) to 24 healthy volunteers.
Superimposable blood CsA concentration profiles were obtained and the mean relative bioavailability of Dexcel's formulation compared with that of Neoral was 101%.
The values for 'the main pharmacokinetic measures and pertinent information on the calculation are summarized in Table 7 below.

Table 7: Pharmacolcinetic Parameters AUC(o_.) AUC(o_t) Cmax Tmax T1/2 N=24 (ng x (ng x hour/ml) (ng/ml) (hours) (hours) hour/ml) Deximune' (Dexcel) 4930 1283 4418 1153 1184 215 1.65 0.48 5.9 2.42 4ean SD

Neoral ovartis 4866 1107 4345 998 1203 231 1.63 0.52 6.05 2.48 ean SD

oint Estimate 101 101 99 (Ratio)*
90%ANOVA 93 93 90 C.I. 109 110 109 ower Limit pper Limit edian 0.00 0.05 ifference -1.50 -5.21 inimum 1.50 8.41 aximum 90% Non- -0.25 -0.99 ara.metric C.I.. 0.25 0.26 ower Limit pper Limit * The presented ratios expressed in percentage (%) are geometric means of the individual ratios between test and reference parameters. Parametric estimators and 90%
Parametric Confidence Intervals, based on the linear model with logarithmic transformation, are brought.

Therefore, it was concluded that these oral CsA formulations are bioequivalent This human study clearly indicates the efficacy of the fonnulation of the present invention as compared to the best coirunercially available formulation.
Sandimmun Neoral (Novartis Inc.).

Example 9 Opthalmic Topical Cyclosporin Fonnulation Effective topical administration of Cyclosporin A to the eye would reduce or eliminate to a large extent the systemic side effects by restricting activity to the locus of the condition being treated. Several formulations for topical delivery of cyclosporine have been reported, as described for example U.S. Pat. No. 4,649,047 and US Pat.
No.
6,5 82,718. However, the utility and effectiveness of Cyclosporin A in treating diseases and conditions of the eye has been hindered until now by the lack of suitable eye-drops which are adapted to the delicate nature of the eye. Eye-drops should not cause patient discomfort and should also permit a convenient administration regimen, by not requiring frequent administration, while providing adequate drug substance delivery to the external and internal regions of the eye. A further difficulty is the very poor solubility of cyclosporin A in water, which frequently leads to precipitation of cyclosporin A from aqueous-based eye-drops, causing major irritation of the eye. For example, the commercially available formulation, Restasis (produced by Allergan), has caused many patients to complain of a burning sensation upon administration.
There is thus an urgent need to develop a topical, ophthalmic formulation wliich causes less irritation to the eyes, is better adapted to evenly distribute cyclosporin A in the eyes and does not cause precipitation of cyclosporin. Also, a stable formulation of cyclosporine that remains in an anhydrous form in storage, while still being capable of dispersion into a microparticulate formulation shortly prior to administration, is preferred.
Such a formulation can be mixed with water to form a microdispersion which is stable for a few weeks of treatment.

According to preferred embodiments thereof, the formulations of the present invention can be mixed with a sterile aqueous medium that is suitable for eye drops to form a dispersion featuring 0.01 to 10 mg/ml of cyclosporin as active agent (after dilution). These clear diluted dispersions can optionally be administered in the eye with a conventional eye dropper. The formulation is optionally and preferably stored as a pro-nanodispersion in the bottle while at the time of initial treatment, the pro-nanodispersion foimulation is diluted with isotonic sterile buffered solution to form a concentration of from about 1 mg/ml to about 0.01 mg/ml. These dispersions require little or no preservative as the for-mulation is preferably mixed with water shortly before use. This also increases the stability of tlie drug.

Alternatively, the pro-nanodispersion is optionally pre-mixed with the buffer solution and packed in single use vesicles or in a multidose bottle which may optionally contain benzyl alcohol or chloramphenicol as preservative for example. Non-limiting examples of other illustrative preservatives with suitable, exemplary amounts (optional ranges) include benzalkonium chloride, 0.004 - 0.01 %; edetate sodium 0.02 -0.05%;
Phenylmercuric acetate or Phenylmercuric nitrate 0.002%; and/or Chlorobutanol and Benzyl alcohol 0.5% (the pH of the solution is preferably buffered to 5.0-5.5) According to preferred embodiments of the present invention, a preferred formulation features cyclosporin in a pro-nanodispersion system comprising polysorbate-20 and polyoxyl 40 hydrogenated castor oil that are approved for ophthalmic solutions.
Sorbitan monooleate is considered as safe for eye formulations ("Handbook of Pharmaceutical Excipients", 4th edition, 2003), while lecithin and solid triglyceride are considered as safe for use in ophthalmic formulations. The solvent ethyl lactate is considered to be safe, as ethyl lactate is a food additive although it has not been used yet in eye formulations.

According to preferred embodiments of the present invention, the ophtllalmic compositions are preferably formulated as pro-nanodispersions, which upon mixing with saline suitable for eye drop formulations, form a microdispersion. With these ophthalmic compositions, very good therapeutic results may be obtained even wlien the cyclosporin is present in low concentrations; for example within the range of from about 0.01 to about 1.0% (w/v), preferably about 0.1% (w/v).

In a typical formulation, Cyclosporin is preferred as the active ingredient (this Example includes 10.00 mg of the active ingredient, but as described herein, other dosages may optionally also be provided). The solvent preferably comprises ethyl lactate, but may optionally comprise N-C 1-4 alicyl pyrrolidone, optionally and preferably N-methyl pyrrolidone, used in a similar amount as for ethyl lactate. The formulation may optionally and preferably comprise a solid fat, which in this Example is a solid triglyceride (such as tricaprin or trilaurin for example). The formulation also preferably features a combination of a low HLB solvent and a high HLB solvent as described herein;
for this Example, the combination comprises Polysorbate 20 and Sorbitan oleate.
Preferably, the formulation features a phospholipid (in this Example, preferably lecithin).
Also preferably, the formulation features an ethoxylated fat such as Creinophor for example. The different examples of ingredients previously described in the specification may also optionally be used for the ophthalmic formulation, as long as tlie ingredients are specially foimulated for use in the eye (and/or are otherwise approved for use in the eye).
Exemplary ranges of amounts of different materials are given in Table 8 below as milligrams of material per l Oml of formulation (the amount in parentheses is for mg per 5 ml formulation).

Table 8: range of components of the eye formulation In redient Quantity per 10 ml 0.1 % cyclosporine final formulation ._(mg), preferred range Ciclosporin 10.00 mg (within the range of from about 0.01 to about 1.0% (w/v), preferably about 0.1 % (w/v)) Hydrophilic surfactant (Polysorbate From about 3 to about 40 (from about 0.3 to 20) about 4.0) Hydrophobic surfactant (Sorbitan From about 3 to.about 40 (from about 0.3 to oleate) about 4.0) Phospholipid (lecithin) From about I to about 20 (from about 0.1 to about 2.0) Solid fat (tricaprin, ethyl stearate) From about 3 to about 30 (from about 0.3 to about 3.0) Ethoxylated hydrogenated castor oil From about 1 to about 20 (from about 0.1 to (polyoxy140 hydro castor oil, about 2.0) Cremophor; or any ethoxylated fat) Ethyl lactate or N-methyl From about 3 to about 30 (from about 0.3 to pyrrolidone , about 3.0) Saline for eye drops 10 ml The amount of each component is preferably determined according to the amounts 10 of the other components. For example, reducing the amount of hydrophilic surfactant (Polysorbate 20) may require increasing the amount of Cremophor.

Table 9 shows an exemplary optical fonnulation according to the present invention.

Table 9 Ingredient Quantity per 10 ml (mg) Ciclosporin 10.00 Polysorbate 20 16.80 Sorbitan oleate 16.80 Lecithin 8.40 Triglyceride 16.80 Macrogolglycerol hydroxystearate 10.00 (polyoxy140 hydro castor oil) Ethyl lactate 10.00 Saline for eye drops 10 ml For this illustrative, non-limiting formulation, particle size upon mixing 100 mg of the pro-nanodispersion with 10 ml of water at body temperature was found to be less than 200 nm. This pro-nanodispersion formulation is stable for more than one year when kept at room temperature in a sealed glass vial. Prior to use, the bottle is opened and 10 ml saline for eye drops is added and shalcen gently to form immediately a translucent dispersion which does not cause irritation to the eye.
Experimental Example I
A diluted formulation of 0.1 mg/ml of the formulation described in Table 9 was instilled in the eye of a rabbit every 30 minutes over a period of 4 hours.
The eye was inspected for any adverse effects. No adverse effects were noted immediately or one week following the treatment.

Additional Examples of Formulations Additional examples of illustrative formulations according to the present invention for ophthalmic administration are described in Table 10 below (the exemplary ingredients are intended for illustrative purposes only and are not meant to be limiting in any way).

Table 10 Ingredient group Exanlple of Concentration prior Concentration in the ingredient to mixing with final composition water (based on 100-200 times dilution) Active ingredient Cyclosporine A 5-10%o 0.05%
Surfactant Tween, SPAN 25-50% 0.125-0.5%
Cremophor Polyoxyethylen Caster Oil, others Triglycerides Tricaprin, Trilaurin, 15-30% 0.075-0.3%
Trimirestin, Lecithin Solvent Propylene Glycol, 20-40% 0.1-0.4%
PEG 400, Ethyl lactate Other Ethylene glycol 0-15% 0-0.15%
palmitostearat carbomer for altering formulation consistency, optional increase in viscosity Osmolarity glycerin 1-3%
maintainer pH stabilizer NaCl As required water Solvent for dilution (diluent) A number of formulations have been tested, resulting in particle size of 40-50nm having very minimal dispersion. Preferred ingredients of those listed above are as follows: surfactants - Tween, SPAN, Cremophor and polyoxyethylene caster oil;
triglycerides - tricaprin, trilaurin; solvent - propylene glycol. Preferably, an osmolarity maintainer, which is more preferably glycerin, is included.
Table 11 shows an exemplary, illustrative preferred'formulation. Percentage of each ingredient is shown before and after dilution 170 times.

Table 11 Pro-nanodispersion % after X170 Materials (percentage) Dilution Cyclosporin 8.50% 0.050 polysorbate 20 14.00% 0.082 sorbitan monooleate 80 14.00% 0.082 Lecithin 7.00% 0.041 Tricaprin 14.00% 0.082 Trilaurin 0.00% 0.000 Polyoxy 40 HCO 14.00% 0.082 Propylene Glycol 28.50% 0.168 TOTAL 100.00% 0.59 Glycerin 2.20%
Water + NaOH to adjust pH to 100%
Animal Testing Exemplary formulations were tested on rabbits. For each rabbit, one eye was tested while the other eye served as control. A formulation according to the present invention as shown in Table 11 above was tested for irritation caused to eye, as compared to the commercially available formulation, Restasis. The experiment was performed on 2 rabbits (one per composition). The formulation according to the present invention was clearly less irritating (6-8 blinks per minute with Restasis vs 2-4 for the inventive formulation). PBS (phosphate buffered saline alone) caused 1-2 blinlcs per minute as a control. Thus, clearly the formulation according to the present invention was much less irritating to the eye than the commercially available formulation.

Example 10 Methods of Administration of Cyclogporins A cyclosporin, such as Ciclosporin, can be administered to a subject in a number of ways, which are well lcnown in the art. Hereinafter, the term "subject"
refers to the human or lower animal to whom cyclosporin was administered. For example achninistration may be done topically (including ophtalmically, vaginally, rectally, intranasally), orally, or parenterally, for example by intravenous drip or intraperitoneal, subcutaneous, or intrainuscular injection.
Formulations for topical administration may include but are not limited to lotions, ointments, gels, creams, suppositories, drops, liquids, sprays and powders. An optional but preferred formulation for administration as eye drops is described above.
Compositions for oral administration include powders or granules, suspensions or solutions in water or non-aqueous media, sachets, capsules or tablets.
Thickeners, diluents, flavorings, dispersing aids, emulsifiers or binders may be desirable.
Coinpositions for oral administration preferably include a soft or hard gelatin capsule.
Formulations for parenteral administration may include but are not limited to sterile aqueous solutions which may also contain buffers, diluents and other suitable additives.
The formulations of the present invention may optionally be administered as a pro-nanodispersion or as a microdispersion in aqueous liquid. Alternatively, these formulations may be lyophilized (dried) after the formation of the microdispersion in aqueous liquid. The lyophilized (dried) dispersion is also optionally administered to the subject. The preferred route of administration is oral administration.
Dosing is dependent on the severity of the syinptoms and on the responsiveness of the subject to cyclosporin. Persons of ordinary skill in the art can easily determine optimum dosages, dosing methodologies and repetition rates.
Example 11 Methods of Treatment with Cyclosporins Cyclosporins are particularly noted for the treatment and prevention of organ or tissue transplant rejection, for the treatment and prevention of autoimmune disease and of inflammatory conditions, and for the treatment of multi-drug resistance (MDR).
With regard to the treatment and prevention of organ or tissue transplant rejection, the compositions of the present invention containing cyclosporin are useful for the treatment of the recipients of heart, lung, combined heart-lung, liver, kidney, pancreatic, bone-marrow, skin or corneal transplants, and in particular allogenic transplants, for example. In addition, the coinpositions of the present invention are useful for the prevention of graft-versus-host-disease, which can sometimes be seen following bone marrow transplantation.

With regard to the treatment and prevention of autoimmune disease and of inflammatory conditions, the compositions of the present invention containing cyclosporin may be useful for the treatment of autoimmune hematological disorder (including hemolytic anemia, aplastic anemia pure red cell anemia and idiopathic 5 thrombocytopenia), systemic lupus erythematosus, polychondritis, scleroderma, Wegener granulamatosis, dermatomyositis, chronic active hepatitis, myasthenia gravis, psoriasis, Steven-Jolinson syndrome, idiopathic sprue, autoimmune inflammatory bowel disease (such as ulcerative colitis and Crohn's disease), endocrine opthalmopathy, Graves disease, sarcoidosis, multiple sclerosis, primary billiary cirrhosis, juvenile diabetes (diabetes 10 mellitus type 1), uveitis (anterior and posterior), keratoconjunctivitis sicca and vernal keratoconjunctivitis, interstitial lung fibrosis, psoriatic arthritis and glomerulonephritis (with and without nephrotic syndrome, such as idiopathic nephrotic syndrome or minimal change nephropathy).

In addition, these compositions may be particularly useful for infla.mmatory 15 conditions with an etiology including an autoimmune component such as arthritis (for example. rheumatoid arthritis, arthritis chronica progrediente and arthritis deformans) and rheumatic diseases.

With regard to multi-drug resistance (MDR), the compositions of the present invention containing cyclosporin may be useful for reversing or abrogating anti-20 neoplastic agent resistance in tumors and the like. The following examples are illustrations only of methods of treating these disorders with the compositions of the present invention containing cyclosporin, and are not intended to be limiting.
The method includes the step of administering the composition of the present invention containing cyclosporin, as described in above, to a subject to be treated. The composition 25 of the present invention is administered according to an effective dosing methodology, preferably until a predefined endpoint is reached (if possible), such as the absence of symptoms of the disorder in the subject. For other disorders, such as organ or tissue transplant rejection, the composition of the preseiit invention may need to be administered continuously without any endpoint.
30 Hereinafter, the term "treatment" includes both pretreatment, before a pathological condition has arisen, and treatment after the condition has arisen. The term "treating" includes both treating the subject after the patllological condition has arisen, and preventing the development of the pathological condition.

It is appreciated that certain features of the _invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination.

Although the invention has been described in conjunction with specific embodiments thereof, it is evident that many alternatives, modifications and variations will be apparent to those skilled in the art. Accordingly, it is intended to embrace all such alternatives, modifications and variations that fall within the spirit and broad scope of the appended claims. All publications, patents and patent applications mentioned in this specification are herein incorporated in their entirety by reference into the specification, to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated herein by reference. In addition, citation or identification of any reference in this application shall not be construed as an admission that such reference is available as prior art to the present invention.

Claims (73)

1. A formulation adapted for ophthalmic administration upon addition of a diluent, comprising:
A pro-nanodispersion comprising a carrier suitable for administration to the eye and cyclosporine.
2. The formulation of claim 1, further comprising a suitable diluent, wherein said pro-nano dispersion forms a dispersion upon dilution with said suitable diluent.
3. The formulation of claim 2, wherein said dispersion comprises particles containing cyclosporine of up to about 500nm.
4. The formulation of any of claims 1-3, wherein said carrier comprises (a) a solid fat;
(b) ethyl lactate;
(c) at least two surfactants comprising a low HLB surfactant and a high HLB
surfactant, wherein a difference between the HLB values for said low and said high HLB
surfactants is at least about 5 units; and (d) an ethoxylated fat.
5. The formulation of any of claims 1-4, wherein said cyclosporine is present in an amount of from about 0.01 to about 1.0% (w/v) upon dilution.
6. The formulation of claim 5, wherein said cyclosporine is present in an amount of about 0.1 % (w/v).
7. The formulation of any of claims 4-6, wherein said hydrophilic surfactant comprises Polysorbate 20.
8. The formulation of claim 7, wherein said polysorbate 20 comprises from about 0.3 to about 4 mg per ml of the final formulation.
9. The formulation of any of claims 4-8, wherein said low HLB surfactant comprises sorbitan oleate.
10. The formulation of claim 9, wherein said sorbitan oleate is present in an amount of from about 0.3 to about 4 mg per ml of the final formulation.
11. The formulation of any of claims 4-10, wherein said solid fat comprises tricaprin or ethyl stearate, or a combination thereof.
12. The formulation of claim 11, wherein said solid fat comprises tricaprin and said tricaprin is present in an amount of from about 0.3 to about 3 mg per ml of the final formulation.
13. The formulation of any of claims 4-12, wherein said ethyoxylated fat comprises ethyoxylated hydrogenated castor oil.
14. The formulation of claim 13, wherein said ethyoxylated hydrogenated castor oil is present in an amount of from about 0.1 to about 10 mg per ml of the final formulation.
15. The formulation of any of claims 4-14, further comprising:
a phospholipid.
16. The formulation of claim 15, wherein said phospholipid comprises lecithin.
17. The formulation of claim 16, wherein said lecithin is present in an amount of from about 0.1 to about 2 mg per ml of the final formulation.
18. Use of the formulation of any of claims 1-17 for manufacture of a medicament for administration of cyclosporine to the eye of a subject in need of administration thereof.
19. A composition for administering a cyclosporin compound, the composition comprising:
(a) a pro-nanodispersion characterized by being capable of forming, upon contact with an aqueous solution, particles of a size of less than about 100 nm at a temperature of at least about 30 C, said pro-nanodispersion comprising:
(i) at least one surfactant; and (ii) an amphiphilic solvent comprising a lower alkyl hydroxyalkanoic acid ester;
and (b) a pharmaceutically effective amount of the cyclosporin compound.
20. The composition of claim 19 wherein said lower alkyl hydroxy alkanoic acid ester comprises ethyl lactate.
21. The composition of claims 19 or 20, wherein said at least one surfactant is a combination of at least two surfactants, at least one surfactant of said combination being a hydrophilic surfactant, and at least one surfactant of said combination being a hydrophobic surfactant.
22. The composition of claim 21, wherein said hydrophilic surfactant has an HLB
(hydrophilic/lipophilic balance) of at least about 10.
23. The composition of claim 22, wherein said hydrophilic surfactant has an HLB
of at least about 14.
24. The composition of claim 22, wherein said hydrophobic surfactant has an HLB of less than about 5.
25. The composition of claim 21, wherein said hydrophobic surfactant comprises a sorbitan fatty acid ester.
26. The composition of claim 21, wherein said combination is a combination of polyoxyethylene (20) sorbitan monolaurate and sorbitan monooleate.
27. The composition of claim 21, further comprising:
(c) an ethoxylated fat.
28. The composition of claim 27, wherein said ethoxylated fat is selected from the group consisting of polyethyleneglycol-hydrogenated castor oils.
29. The composition of claim 28, wherein said ethoxylated fat is selected from the group consisting of Cremophor and Cremophor RH
30. The composition of claim 27, further comprising:
(d) a phospholipid.
31. The composition of claim 30, wherein said phospholipid comprises lecithin.
32. The composition of claim 31, wherein said phospholipid is selected from the group consisting of egg phospholipid, bovine heart phospholipid and soy phospholipid.
33. The composition of claim 30, further comprising:
(e) a fatty acid ester.
34. The composition of claim 33, wherein said fatty acid ester is a solid fat at room temperature.
35. The composition of claim 34, wherein said fatty acid ester comprises triglyceride.
36. The composition of claim 19, wherein said particle size is less than about nm at a temperature of at least about 30 C.
37. The composition of claim 36, wherein said particle size is in a range of from about 5 nm to about 50 nm at a temperature of at least about 30 C.
38. The composition of claim 19, wherein the cyclosporin compound is Ciclosporin.
39. A composition for administering a cyclosporin compound, the composition comprising a pharmaceutically effective amount of the composition of claim 19, and an aqueous solution as a diluent for said pharmaceutically effective amount of the composition of claim 19.
40. A composition for administering a cyclosporin compound, the composition comprising a lyophilized composition, said lyophilized composition being formed from a pharmaceutically effective amount of the composition of claim 19 and an aqueous solution as a diluent for said pharmaceutically effective amount of the composition of claim 19 to form a diluted solution, said diluted solution being lyophilized to form said lyophilized composition.
41. Use of a composition for administering a cyclosporin compound to a subject in need of treatment thereof, the composition comprising a pharmaceutically effective amount of the composition of any of claims 19-40.
42. The use of claim 41, wherein said subject is in need of treatment of a condition selected from the group consisting of autoimmune disease and inflammatory conditions and said composition is adapted thereto.
43. The use of claim 42, wherein said subject is in need of treatment of organ or tissue transplant rejection and said composition is adapted thereto.
44. The use of claim 42, wherein said pharmaceutically effective amount of the composition of any of claims 19-40 is administered to the subject through oral administration and said composition is adapted thereto.
45. The use of claim 42, wherein said pharmaceutically effective amount of the composition of any of claims 19-40 is administered as a dispersion with an aqueous solution as a diluent and said composition is adapted thereto.
46. The use of claim 42, wherein said pharmaceutically effective amount of the composition of any of claims 19-40 is administered to the subject through topical administration and said composition is adapted thereto.
47. The use of claim 42, wherein said pharmaceutically effective amount of the composition of any of claims 19-40 is administered to the subject through parenteral administration and said composition is adapted thereto.
48. The use of claim 42, wherein said pharmaceutically effective amount of the composition of any of claims 19-40 is administered as a capsule and said composition is adapted thereto.
49. The use of claim 42, wherein said pharmaceutically effective amount of the composition of any of claims 19-40 is administered as a tablet and said composition is adapted thereto.
50. The use of claim 42, wherein said pharmaceutically effective amount of the composition of any of claims 19-40 is administered as a powder and said composition is adapted thereto.
51. A method for determining storage stability of a formulation containing a cyclosporin compound, the method comprising the step of analyzing the composition of any of claims 19-40 for particle size, such that if said particle size is less than about 100 nm, the formulation is determined to be stable.
52. A formulation for topical administration to the eye, comprising:
(a) a pro-nanodispersion comprising:
(i) a solid fat;
(ii) an amphiphilic solvent;

(iii) at least one hydrophilic surfactant; and (b) a pharmaceutically effective amount of the cyclosporin compound;
wherein the components are adapted for administration to the eye.
53. The formulation of claim 52, wherein said cyclosporine is present in an amount of from about 0.01 to about 1.0% (w/v).
54. The formulation of claim 53, wherein said cyclosporine is present in an amount of about 0.1% (w/v).
55. The formulation of any claims 52-54, wherein said hydrophilic surfactant comprises Polysorbate 20.
56. The formulation of claim 55, wherein said polysorbate 20 comprises from about 0.3 to about 4 mg per ml of the final formulation.
57. The formulation of any of claims 52-56, further comprising a low HLB
surfactant, wherein a difference between the HLB values for said low and HLB
surfactant and said hydrophilic surfactant is at least about 5 units.
58. The formulation of claim 57, wherein said low HLB surfactant comprises sorbitan oleate.
59. The formulation of claim 58, wherein said sorbitan oleate is present in an amount of from about 0.3 to about 4 mg per ml of the final formulation.
60. The formulation of any of claims 52-59, wherein said solid fat comprises tricaprin or ethyl stearate, or a combination thereof.
61. The formulation of claim 60, wherein said solid fat comprises tricaprin.
62. The formulation of claim 61, wherein said tricaprin is present in an amount of from about 0.3 to about 3 mg per ml of the final formulation.
63. The formulation of any of claims 52-62, wherein said surfactant further comprises an ethoxylated fat.
64. The formulation of any of claims 52-63, wherein said ethyoxylated fat comprises ethoxylated hydrogenated castor oil.
65. The formulation of claim 64, wherein said ethoxylated hydrogenated castor oil is present in an amount of from about 0.1 to about 2 mg per ml of the final formulation.
66. The formulation of any of claims 52-65, wherein said solvent comprises ethyl lactate.
67. The formulation of any of claims 52-65, wherein said solvent comprises N-methyl pyrrolidone.
68. The formulation of any of claims 52-67, further comprising:
a phospholipid.
69. The formulation of claim 68, wherein said phospholipid comprises lecithin.
70. The formulation of claim 69, wherein said lecithin is present in an amount of from about 0.1 to about 2 mg per ml of the final formulation.
71. The formulation of any of claims 52-70, further comprising a suitable diluent.
72. Use of the formulation of any of claims 51-71 for treatment of a condition of the eye.
73. The use of claim 72, wherein said suitable diluent is stored separately from the formulation comprising the active ingredient and wherein prior to said administering said formulation, said suitable diluent is mixed with the formulation.
CA002644471A 2006-02-28 2007-02-28 Pro-nanodispersion for the delivery of cyclosporin Abandoned CA2644471A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US11/363,379 2006-02-28
US11/363,379 US7732404B2 (en) 1999-12-30 2006-02-28 Pro-nanodispersion for the delivery of cyclosporin
PCT/IL2007/000264 WO2007099537A1 (en) 2006-02-28 2007-02-28 Pro-nanodispersion for the delivery of cyclosporin

Publications (1)

Publication Number Publication Date
CA2644471A1 true CA2644471A1 (en) 2007-09-07

Family

ID=38169240

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002644471A Abandoned CA2644471A1 (en) 2006-02-28 2007-02-28 Pro-nanodispersion for the delivery of cyclosporin

Country Status (6)

Country Link
US (1) US7732404B2 (en)
EP (1) EP1988879A1 (en)
JP (1) JP2009529003A (en)
AU (1) AU2007220104A1 (en)
CA (1) CA2644471A1 (en)
WO (1) WO2007099537A1 (en)

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005072701A1 (en) 2004-01-20 2005-08-11 Allergan, Inc. Compositions for localized therapy of the eye, comprising preferably triamcinolone acetonide and hyaluronic acid
GB0623838D0 (en) * 2006-11-29 2007-01-10 Malvern Cosmeceutics Ltd Novel compositions
WO2008095122A2 (en) * 2007-01-31 2008-08-07 Bioactives, Inc. Methods of reducing 15-f2t-isop levels in mammals
US8101274B2 (en) * 2007-06-11 2012-01-24 Spedden Richard H Solid state membranes with surface-embedded glycosylated amphiphilic molecules and micelles formed therefrom
KR20110042107A (en) 2008-08-07 2011-04-22 바이오엑티브 써지컬, 아이엔씨. Stem cell capture and immobilization coatings for medical devices and implants
US20110033527A1 (en) * 2009-06-02 2011-02-10 Nian Wu Opthalmic compositions of cyclosporin
BR112014011744A2 (en) 2011-11-15 2017-05-02 Allergan Inc sustainable action formulation of cyclosporine form 2
EP3701939A1 (en) * 2011-11-15 2020-09-02 Allergan, Inc. Suspensions of cyclosporin a form 2
WO2013074608A1 (en) 2011-11-15 2013-05-23 Allergan, Inc. Cyclosporine a form 2 and method of making same
FR2988297B1 (en) * 2012-03-22 2014-03-28 Thea Lab AQUEOUS OPHTHALMIC SOLUTION FROM CICLOSPORINE WITHOUT PRESERVATIVE
CA2914472C (en) * 2012-08-24 2019-09-03 Ashim K. Mitra Ophthalmic formulation of polyoxyl lipid or polyoxyl fatty acid and treatment of ocular conditions
KR101492447B1 (en) * 2013-05-20 2015-02-23 주식회사태준제약 Eye composition containing a cyclosporine and a method of preparing the same
US10792248B2 (en) * 2013-11-08 2020-10-06 Activus Pharma Co., Ltd. Aqueous suspension preparation comprising nanoparticles of macrolide antibacterial agent
AU2016226224B2 (en) * 2015-03-05 2021-07-08 Allergan, Inc. Self-emulsifying drug delivery system (SEDDS) for ophthalmic drug delivery
CA3015626A1 (en) 2016-02-29 2017-09-08 Sun Pharma Global Fze Topical cyclosporine-containing formulations and uses thereof
US20190380958A1 (en) * 2016-12-28 2019-12-19 Chugai Seiyaku Kabushiki Kaisha Self-emulsifying drug formulation for improving membrane permeability of compound

Family Cites Families (257)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR1274354A (en) 1956-03-10 1961-10-27 Surfactants obtained from triglycerides and polyethylene glycol
GB1171125A (en) 1966-06-08 1969-11-19 Glaxo Lab Ltd Improvements in or relating to Injectable Preparations
US3954967A (en) 1971-08-05 1976-05-04 Vanguard Chemical Company, Inc. Method of producing microcolloidal aqueous emulsions of unsaturated organic insecticidal compounds
US3813345A (en) 1971-08-05 1974-05-28 Vanguard Chem Co Inc Method of producing microcolloidal aqueous emulsions of unsaturated organic compounds
US4073943A (en) 1974-09-11 1978-02-14 Apoteksvarucentralen Vitrum Ab Method of enhancing the administration of pharmalogically active agents
GB1549186A (en) 1975-10-03 1979-08-01 Nat Res Dev Optical connectors
US4117118A (en) 1976-04-09 1978-09-26 Sandoz Ltd. Organic compounds
US4146499A (en) 1976-09-18 1979-03-27 Rosano Henri L Method for preparing microemulsions
JPS53107408A (en) 1977-02-28 1978-09-19 Yamanouchi Pharmaceut Co Ltd Micellar preparation for rectal infusion
FI65914C (en) 1978-03-07 1984-08-10 Sandoz Ag FRAMEWORK FOR PHARMACEUTICAL COMPOSITION OF CYCLOSPORINE A
US4215199A (en) 1978-06-05 1980-07-29 Sandoz Ltd. Antibiotic production
DE3167014D1 (en) 1980-02-14 1984-12-13 Sandoz Ag A method for the total synthesis of cyclosporins and novel cyclosporins
FR2502951B1 (en) 1981-04-06 1985-12-06 Sandoz Sa TOPICAL PHARMACEUTICAL COMPOSITIONS IN THE FORM OF A MICRO-EMULSION
DE3225706C2 (en) 1982-07-09 1984-04-26 A. Nattermann & Cie GmbH, 5000 Köln Liquid active ingredient formulations in the form of concentrates for microemulsions
DE3237814A1 (en) 1982-10-12 1984-04-12 Warner-Lambert Co., 07950 Morris Plains, N.J. WATER-FREE EMULSIONS AND USE THEREOF
SE8301182D0 (en) 1983-03-04 1983-03-04 Haessle Ab NOVEL COMPOUNDS
US4731384A (en) 1983-07-01 1988-03-15 Troponwerke Gmbh & Co, Kg Etofenamate formulation
JPS6061535A (en) 1983-08-24 1985-04-09 エフ・ホフマン・ラ・ロシユ・ウント・コンパニ−・アクチエンゲゼルシヤフト Pharmaceutical composition
DE3406497A1 (en) 1984-02-23 1985-09-05 Mueller Bernhard Willi Werner HIGHLY DISPERSAL PHARMACEUTICAL MULTI-COMPONENT SYSTEMS AND METHOD FOR THEIR PRODUCTION
US4713246A (en) 1984-03-19 1987-12-15 Bristol-Myers Company Etoposide oral dosage form
US4794000A (en) 1987-01-08 1988-12-27 Synthetic Blood Corporation Coacervate-based oral delivery system for medically useful compositions
US4963367A (en) 1984-04-27 1990-10-16 Medaphore, Inc. Drug delivery compositions and methods
US4572915A (en) 1984-05-01 1986-02-25 Bioglan Laboratories Clear micellized solutions of fat soluble essential nutrients
US4871768A (en) 1984-07-12 1989-10-03 New England Deaconess Hospital Corporation Dietary supplement utilizing ω-3/medium chain trigylceride mixtures
US5639724A (en) 1984-07-24 1997-06-17 Sandoz Ltd. Cyclosporin galenic forms
EP0365044A3 (en) 1984-08-02 1990-08-22 Sandoz Ag Novel pharmaceutical use of (nva)2-cyclosporine
JPS6150978A (en) 1984-08-16 1986-03-13 Takeda Chem Ind Ltd Pyridine derivative and preparation thereof
DE3500103A1 (en) 1985-01-04 1986-07-10 R.P. Scherer GmbH, 6930 Eberbach PHARMACEUTICAL PREPARATION WITH AN INTENSIVE SOLUTION IN WATER AND DIGESTIVE JUICES
US4885172A (en) 1985-06-26 1989-12-05 The Liposome Company, Inc. Composition for targeting, storing and loading of liposomes
US5171578A (en) 1985-06-26 1992-12-15 The Liposome Company, Inc. Composition for targeting, storing and loading of liposomes
IL78929A0 (en) 1985-07-29 1986-09-30 Abbott Lab Microemulsion compositions for parenteral administration
US4753963A (en) 1985-09-26 1988-06-28 The Procter & Gamble Company Nutritional fat suitable for enteral and parenteral products
US5041278A (en) 1985-10-15 1991-08-20 The Liposome Company, Inc. Alpha tocopherol-based vesicles
US4861580A (en) 1985-10-15 1989-08-29 The Liposome Company, Inc. Composition using salt form of organic acid derivative of alpha-tocopheral
US4797272A (en) 1985-11-15 1989-01-10 Eli Lilly And Company Water-in-oil microemulsions for cosmetic uses
CA1327010C (en) 1986-02-13 1994-02-15 Tadashi Makino Stabilized solid pharmaceutical composition containing antiulcer benzimidazole compound and its production
GB2189699A (en) 1986-04-30 1987-11-04 Haessle Ab Coated acid-labile medicaments
SE457693B (en) 1986-07-01 1989-01-23 Drilletten Ab COMPOSITION WITH REGULATED RELEASE WAS A BIOLOGICAL MATERIAL LOST OR DISPERSED IN AN L2 PHASE
CA1318589C (en) 1986-08-14 1993-06-01 Bernard Ecanow Drug delivery system
DE3629386A1 (en) 1986-08-29 1988-03-03 Scherer Gmbh R P GELATINE CAPSULES AND METHOD FOR THEIR PRODUCTION
US5071643A (en) 1986-10-17 1991-12-10 R. P. Scherer Corporation Solvent system enhancing the solubility of pharmaceuticals for encapsulation
GB8630273D0 (en) 1986-12-18 1987-01-28 Til Medical Ltd Pharmaceutical delivery systems
US5154930A (en) 1987-03-05 1992-10-13 The Liposome Company, Inc. Pharmacological agent-lipid solution preparation
DE3707711A1 (en) 1987-03-11 1988-09-22 Hoechst Ag OIL-IN-WATER EMULSIONS, METHOD FOR THEIR PRODUCTION AND THEIR USE
CA1301642C (en) 1987-03-30 1992-05-26 Howard Bernard Dawson Chemical formulations
KR880012221A (en) 1987-04-13 1988-11-26 사노 가즈오 Pharmaceutical compositions containing esters or amides as active ingredients
US4798823A (en) 1987-06-03 1989-01-17 Merck & Co., Inc. New cyclosporin analogs with modified "C-9 amino acids"
JP2577049B2 (en) 1987-06-04 1997-01-29 三共株式会社 Cyclosporine preparation
ATE95193T1 (en) 1987-06-17 1993-10-15 Sandoz Ag CYCLOSPORINS AND THEIR USE AS MEDICINAL PRODUCTS.
GB2206119B (en) 1987-06-22 1990-10-31 Merck & Co Inc A new cyclosporin derivative with modified "8-amino acid"
US4835002A (en) 1987-07-10 1989-05-30 Wolf Peter A Microemulsions of oil in water and alcohol
US4963362A (en) 1987-08-07 1990-10-16 Regents Of The University Of Minnesota Freeze-dried liposome mixture containing cyclosporin
HU205010B (en) 1987-09-15 1992-03-30 Sandoz Ag Process for producing pharmaceutical compositions comprising compounds soluble up to 1 per cent and having medical activity
US5756450A (en) 1987-09-15 1998-05-26 Novartis Corporation Water soluble monoesters as solubilisers for pharmacologically active compounds and pharmaceutical excipients and novel cyclosporin galenic forms
GB8729153D0 (en) 1987-12-14 1988-01-27 Efamol Ltd Fatty acid compositions
US5244925A (en) 1987-12-18 1993-09-14 Kabi Pharmacia Aktiebolag Emulsion for parenteral administration
HU203564B (en) 1987-12-21 1991-08-28 Sandoz Ag Process for producing new orthorombos cyclosporin without solvatation
US5081105A (en) 1988-01-15 1992-01-14 New England Deaconess Hospital Corporation Method of treating cancer using structured lipids
US4904474A (en) 1988-01-25 1990-02-27 Alza Corporation Delivery of drug to colon by oral disage form
ES2033086T3 (en) 1988-01-29 1993-03-01 Sankyo Company Limited A PROCEDURE FOR THE PREPARATION OF A PHARMACEUTICAL COMPOSITION.
US4994267A (en) 1988-03-04 1991-02-19 Noven Pharmaceuticals, Inc. Transdermal acrylic multipolymer drug delivery system
US5656286A (en) 1988-03-04 1997-08-12 Noven Pharmaceuticals, Inc. Solubility parameter based drug delivery system and method for altering drug saturation concentration
US4814168A (en) 1988-03-04 1989-03-21 Noven Pharmaceuticals, Inc. Transdermal multipolymer drug delivery system
GB8809616D0 (en) 1988-04-22 1988-05-25 Cancer Res Campaign Tech Further improvements relating to drug delivery systems
JP2677613B2 (en) 1988-06-24 1997-11-17 エーザイ株式会社 Absorption promoting composition of vitamin E or derivative thereof
HU201567B (en) 1988-07-21 1990-11-28 Gyogyszerkutato Intezet Process for production of intravenous medical compositions containing cyclosphorin
US5350741A (en) 1988-07-30 1994-09-27 Kanji Takada Enteric formulations of physiologically active peptides and proteins
US5342625A (en) 1988-09-16 1994-08-30 Sandoz Ltd. Pharmaceutical compositions comprising cyclosporins
KR0148748B1 (en) 1988-09-16 1998-08-17 장 크라메르, 한스 루돌프 하우스 A multiphase cyclosporin composition
US6007840A (en) 1988-09-16 1999-12-28 Novartis Ag Pharmaceutical compositions comprising cyclosporins
CA1337397C (en) 1988-09-29 1995-10-24 Hideo Nakajima Emulsified composition
GB8822857D0 (en) 1988-09-29 1988-11-02 Patralan Ltd Pharmaceutical formulations
SE8804164A0 (en) 1988-11-17 1990-05-18 Per Prisell Pharmaceutical preparation
HU201577B (en) 1988-12-20 1990-11-28 Gyogyszerkutato Intezet Process for producing cyclosporin antibiotics
US4994439A (en) 1989-01-19 1991-02-19 California Biotechnology Inc. Transmembrane formulations for drug administration
US4996193A (en) 1989-03-03 1991-02-26 The Regents Of The University Of California Combined topical and systemic method of administration of cyclosporine
EP0388152B1 (en) 1989-03-15 1994-10-12 Merck & Co. Inc. Process for producing an immunosuppressant agent (demethimmunomycin) using a mutant strain of a microorganism
US5272068A (en) 1989-03-15 1993-12-21 Merck & Co., Inc. Process for producing immunosuppressant agent L-683942 by fermentation
US5364632A (en) 1989-04-05 1994-11-15 Yissum Research Development Company Of The Hebrew University Of Jerusalem Medicinal emulsions
AU614465B2 (en) 1989-04-05 1991-08-29 Yissum Research Development Company Of The Hebrew University Of Jerusalem Medicinal emulsions
DE3919982A1 (en) 1989-06-19 1990-12-20 Liedtke Pharmed Gmbh ORAL LIPID MEDICINE FORM
US5532002A (en) 1989-08-17 1996-07-02 Cortecs Limited Gelatin pharmaceutical formulations
DK469989D0 (en) 1989-09-22 1989-09-22 Bukh Meditec PHARMACEUTICAL PREPARATION
IE904098A1 (en) 1989-11-13 1991-05-22 Nova Pharm Corp Lipospheres for controlled delivery of substances
US5188837A (en) 1989-11-13 1993-02-23 Nova Pharmaceutical Corporation Lipsopheres for controlled delivery of substances
US5227165A (en) 1989-11-13 1993-07-13 Nova Pharmaceutical Corporation Liposphere delivery systems for local anesthetics
KR960005137B1 (en) 1989-11-13 1996-04-22 노바 파머수티컬 코포레이션 Lipospheres for controlled delivery of substances
JP2785981B2 (en) 1989-11-20 1998-08-13 株式会社資生堂 Emulsion composition
US5178866A (en) 1990-03-23 1993-01-12 Alza Corporation Dosage form for delivering drug to the intestine
DE69029909T2 (en) 1990-03-28 1997-09-11 Noven Pharma METHOD AND DEVICE FOR RELEASING MEDICINAL PRODUCTS ON THE SKIN
US5091188A (en) 1990-04-26 1992-02-25 Haynes Duncan H Phospholipid-coated microcrystals: injectable formulations of water-insoluble drugs
IL98087A (en) 1990-05-04 1996-11-14 Perio Prod Ltd Colonic drug delivery system
ATE121618T1 (en) 1990-08-13 1995-05-15 David W Yesair MIXED LIPID-BICARBONATE COLLOIDAL PARTICLES FOR DELIVERING MEDICINAL AND CALORIES.
US5227298A (en) 1990-08-17 1993-07-13 The Trustees Of Columbia University In The City Of New York Method for microencapuslation of cells or tissue
US5156850A (en) 1990-08-31 1992-10-20 Alza Corporation Dosage form for time-varying patterns of drug delivery
US5665379A (en) 1990-09-28 1997-09-09 Pharmacia & Upjohn Aktiebolag Lipid particle forming matrix, preparation and use thereof
DE69124459T3 (en) 1990-11-02 2001-05-31 Novartis Ag Cyclosporine
KR0154343B1 (en) 1990-11-06 1998-11-16 아만 히데아키 Lyophilized fatty emulsions and a process for the production thereof
HU208491B (en) 1990-11-27 1993-11-29 Gyogyszerkutato Intezet Process for producing oral pharmaceutical composition containing cyclosporin
NL9100038A (en) 1991-01-11 1992-08-03 Stamicarbon ENZYME-CATALYZED PREPARATION OF OPTICALLY ACTIVE CARBONIC ACIDS.
EP0568698B1 (en) 1991-01-25 1997-09-10 Fujisawa Pharmaceutical Co., Ltd. Process for producing cyclosporin a and/or c
US5145684A (en) 1991-01-25 1992-09-08 Sterling Drug Inc. Surface modified drug nanoparticles
CZ2190U1 (en) 1991-02-01 1994-07-14 Alexandr Rndr. Csc. Jegorov Targeted polymeric cyclosporin conjugates
US5300529A (en) 1991-02-12 1994-04-05 Isp Investments Inc. Stable, clear, efficacious aqueous microemulsion compositions containing a high loading of a water-insoluble, agriculturally active chemical
US5378475A (en) 1991-02-21 1995-01-03 University Of Kentucky Research Foundation Sustained release drug delivery devices
US5171217A (en) 1991-02-28 1992-12-15 Indiana University Foundation Method for delivery of smooth muscle cell inhibitors
ES2078374T3 (en) 1991-04-06 1995-12-16 Dresden Arzneimittel PROCEDURE FOR THE PRODUCTION BY FERMENTATION AND ISOLATION OF CYCLOSPORINE A, AND NEW STRAINS FORMING CYCLOSPORIN.
AU657961B2 (en) 1991-04-19 1995-03-30 Nexstar Pharmaceuticals, Inc. Pharmaceutical formulation and process
AU668509B2 (en) 1991-04-19 1996-05-09 Affinity Biotech, Inc. Convertible microemulsion formulations
US5683714A (en) 1991-04-19 1997-11-04 Nexstar Pharmaceuticals, Inc. Liposomal cyclosporin pharmaceutical formulation
DE69127275T2 (en) 1991-06-21 1998-03-12 Ilsan Ilac Ve Hammaddeleri San New galenic process for pellets containing omeprazole
GB9113872D0 (en) 1991-06-27 1991-08-14 Sandoz Ag Improvements in or relating to organic compounds
US5206219A (en) 1991-11-25 1993-04-27 Applied Analytical Industries, Inc. Oral compositions of proteinaceous medicaments
US5545423A (en) 1991-11-25 1996-08-13 Vivorx, Inc. Cytoprotective, biocompatible, retrievable macrocapsule containment systems for biologically active materials
US5614219A (en) 1991-12-05 1997-03-25 Alfatec-Pharma Gmbh Oral administration form for peptide pharmaceutical substances, in particular insulin
IL101007A (en) 1992-02-18 1997-08-14 Pharmos Ltd Dry stable compositions prepared by lyophilization
SE9200951D0 (en) 1992-03-27 1992-03-27 Kabi Pharmacia Ab PHARMACEUTICAL COMPOSITION CONTAINING A DEFINED LIPID SYSTEM
US5529785A (en) 1993-05-12 1996-06-25 Dietl; Hans Pharmaceutical preparation containing cyclosporin(s) for oral administration and process for producing same
US5637317A (en) 1992-05-18 1997-06-10 Dietl; Hans Pharmaceutical preparation containing cyclosporin(s) for oral administration and process for producing same
HU213553B (en) 1992-05-25 1997-07-28 Biogal Gyogyszergyar Process for isolating of cyclosporin-a
IT1260505B (en) 1992-06-01 1996-04-09 Poli Ind Chimica Spa ORAL PHARMACEUTICAL SYSTEMS WITH DELAYED DELIVERY FOR THE SPECIFIC RELEASE IN THE COLON
GB9213874D0 (en) 1992-06-30 1992-08-12 Fisons Plc Process to novel medicament form
IT1255449B (en) 1992-06-30 1995-10-31 Fabio Berlati USE OF NOR- AND HOMO-DERIVATIVES OF BILE ACIDS AS DRUGS ABSORPTION PROMOTERS.
US5759566A (en) 1992-07-28 1998-06-02 Poli Industria Chimica Spa Microemulsion pharmaceutical compositions for the delivery of pharmaceutically active agents
EP0616802B1 (en) 1992-08-13 1998-06-17 Teikoku Seiyaku Co., Ltd. Oral preparation for release in lower digestive tracts
CZ278863B6 (en) 1992-09-07 1994-07-13 Galena Medicinal preparations with n-methylated cyclic undecapeptides
DK0661989T3 (en) 1992-09-21 1998-03-02 Upjohn Co Prolonged release protein compositions
DK0589843T3 (en) 1992-09-25 2002-04-02 Novartis Ag Cyclosporin-containing pharmaceutical preparations
ATE175132T1 (en) 1992-10-26 1999-01-15 Sanol Arznei Schwarz Gmbh METHOD FOR PRODUCING MICRO CAPSULES
IT1256386B (en) 1992-11-13 1995-12-04 Luigi Boltri PHARMACEUTICAL COMPOSITIONS INCLUDING A DRUG, A CROSS-LINKED POLYMERIC SUBSTANCE, AN OIL AND A SURFACTIVE AGENT
DE69326015T2 (en) 1992-11-18 2000-02-03 Fujisawa Pharmaceutical Co PHARMACEUTICAL PREPARATION WITH EXTENDED EFFECT
US5376688A (en) 1992-12-18 1994-12-27 R. P. Scherer Corporation Enhanced solubility pharmaceutical solutions
FI92334C (en) 1992-12-30 1994-10-25 Leiras Oy Process for producing cyclosporins and new Tolypocladium strain useful in the process
AU673700B2 (en) 1993-01-27 1996-11-21 Scotia Holdings Plc Triglycerides
EP0684814B1 (en) 1993-02-22 1998-06-17 Alza Corporation Compositions for oral delivery of active agents
US5686105A (en) 1993-10-19 1997-11-11 The Procter & Gamble Company Pharmaceutical dosage form with multiple enteric polymer coatings for colonic delivery
CA2120197A1 (en) 1993-04-02 1994-10-03 Kenji Endo Stable aqueous dispersions containing liposomes
JP3119540B2 (en) 1993-04-12 2000-12-25 松下電器産業株式会社 Light tap
AU686149B2 (en) 1993-04-19 1998-02-05 Institute For Advanced Skin Research Inc. Microemulsion preparation containing difficultly absorbable substance
ES2115231T3 (en) 1993-04-20 1998-06-16 Hexal Ag PLASTER WITH ACTIVE SUBSTANCES.
DE4338086A1 (en) 1993-11-08 1995-05-11 Dietl Hans Cyclosporin (e) containing pharmaceutical preparation for oral administration and process for their preparation
US5576016A (en) 1993-05-18 1996-11-19 Pharmos Corporation Solid fat nanoemulsions as drug delivery vehicles
GB2278780B (en) 1993-05-27 1998-10-14 Sandoz Ltd Macrolide formulations
CH686761A5 (en) 1993-05-27 1996-06-28 Sandoz Ag Pharmaceutical formulations.
US5593691A (en) 1993-06-03 1997-01-14 Marigen S.A. Biotenside solvents for pharmaceuticals and cosmetics
US5478860A (en) 1993-06-04 1995-12-26 Inex Pharmaceuticals Corp. Stable microemulsions for hydrophobic compound delivery
US5639474A (en) 1993-07-01 1997-06-17 Hanmi Pharm. Ind., Ltd. Cyclosporin soft capsule composition
DE4322826A1 (en) 1993-07-08 1995-01-12 Galenik Labor Freiburg Gmbh Pharmaceutical preparation
WO1995002838A1 (en) 1993-07-16 1995-01-26 Luckoff Display Corporation Diffractive display utilizing reflective or transmissive light yielding single pixel full color capability
ES2068762B1 (en) 1993-07-21 1995-12-01 Lipotec Sa A NEW PHARMACEUTICAL PREPARATION TO IMPROVE THE BIOAVAILABILITY OF DRUGS OF DIFFICULT ABSORPTION AND PROCEDURE FOR THEIR OBTAINING.
IL110787A0 (en) 1993-08-27 1994-11-11 Sandoz Ag Biodegradable polymer, its preparation and pharmaceutical composition containing it
DE69426408T2 (en) 1993-09-28 2001-07-12 Scherer Gmbh R P Manufacture of soft gelatin capsules
JP3644543B2 (en) 1993-10-22 2005-04-27 ヘクサル・アーゲー Pharmaceutical composition containing cyclosporin A and α-tocopherol
US6022852A (en) 1993-10-22 2000-02-08 Hexal Ag Pharmaceutical composition containing cyclosporin A
IL107471A (en) 1993-11-02 1997-09-30 Yissum Res Dev Co Pharmaceutical preparation comprising a self- emulsifying formulation and process for its production
WO1995014037A1 (en) 1993-11-17 1995-05-26 Ibah, Inc. Transparent liquid for encapsulated drug delivery
DE4340781C3 (en) 1993-11-30 2000-01-27 Novartis Ag Liquid preparations containing cyclosporin and process for their preparation
DE4402867C1 (en) 1994-01-31 1995-06-14 Rentschler Arzneimittel Liposome(s) contg. encapsulated protein for pharmaceutical or cosmetic use
US5567592A (en) 1994-02-02 1996-10-22 Regents Of The University Of California Screening method for the identification of bioenhancers through the inhibition of P-glycoprotein transport in the gut of a mammal
ATE201981T1 (en) 1994-02-04 2001-06-15 Scotia Lipidteknik Ab OIL-IN-WATER EMULSIONS
US5891846A (en) 1994-02-17 1999-04-06 Shiseido Company, Ltd. Cyclosporin-containing emulsion composition
KR0146671B1 (en) 1994-02-25 1998-08-17 김충환 Cyclosporin-containing powder composition
CA2184834A1 (en) 1994-03-11 1995-09-14 Yoshiyuki Mori Liposome preparation
GB9405304D0 (en) 1994-03-16 1994-04-27 Scherer Ltd R P Delivery systems for hydrophobic drugs
US5430021A (en) 1994-03-18 1995-07-04 Pharmavene, Inc. Hydrophobic drug delivery systems
US5731355A (en) 1994-03-22 1998-03-24 Zeneca Limited Pharmaceutical compositions of propofol and edetate
US5651980A (en) 1994-04-15 1997-07-29 Biohybrid Technologies, Inc. Methods of use of uncoated gel particles
DE4417851C1 (en) 1994-05-20 1995-10-05 Horst Heirler Dietary food with medium-chain fatty acids and its use
FI100692B (en) 1994-05-24 1998-02-13 Leiras Oy A process for the preparation of pharmaceutical compositions, wherein the compositions are based on microemulsion gels and new gels directed to microemulsions.
EP0756489B1 (en) 1994-06-01 1999-08-18 Yuhan Corporation Cyclosporin containing composition and process for the preparation thereof
US5958458A (en) 1994-06-15 1999-09-28 Dumex-Alpharma A/S Pharmaceutical multiple unit particulate formulation in the form of coated cores
US5817320A (en) 1994-06-20 1998-10-06 The United States Of America As Represented By The Secretary Of The Agriculture In ovo immunization of avian embryos with oil-emulsion vaccines
US5536507A (en) 1994-06-24 1996-07-16 Bristol-Myers Squibb Company Colonic drug delivery system
GB2290965A (en) 1994-07-11 1996-01-17 Therapicon Srl Multiple layer capsules for drugs
EP0697214A1 (en) 1994-07-19 1996-02-21 Vestar, Inc. Liposomal cyclosporin pharmaceutical formulations
US5616330A (en) 1994-07-19 1997-04-01 Hemagen/Pfc Stable oil-in-water emulsions incorporating a taxine (taxol) and method of making same
US5599534A (en) 1994-08-09 1997-02-04 University Of Nebraska Reversible gel-forming composition for sustained delivery of bio-affecting substances, and method of use
US5733575A (en) 1994-10-07 1998-03-31 Bpsi Holdings, Inc. Enteric film coating compositions, method of coating therewith, and coated forms
KR100304324B1 (en) 1994-10-13 2001-11-22 김용규 Method for manufacturing cyclosporine a
KR0126610B1 (en) 1994-10-18 1997-12-29 김충환 Producing method of cyclosporina by cell fusioned microorganism
US5603951A (en) 1994-11-09 1997-02-18 Hanmi Pharm. Ind. Co., Ltd. Cyclosporin-containing soft capsule compositions
DE4440337A1 (en) 1994-11-11 1996-05-15 Dds Drug Delivery Services Ges Pharmaceutical nanosuspensions for drug application as systems with increased saturation solubility and dissolution rate
HU215966B (en) 1994-11-21 1999-07-28 BIOGAL Gyógyszergyár Rt. Oral multiple emulsion-preconcentrate containing cyclosporin
DE4446470A1 (en) 1994-12-23 1996-06-27 Basf Ag Process for the production of dividable tablets
KR0167613B1 (en) 1994-12-28 1999-01-15 한스 루돌프 하우스, 니콜 케르커 Cyclosporin-containing soft capsule compositions
US5608278A (en) 1995-01-13 1997-03-04 Eastman Kodak Company Self-pumped fluid bearing with electromagnetic levitation such as for a light beam deflector
EP0725076B1 (en) 1995-02-01 2001-06-06 National Research Development Corporation of India A process for the preparation of cyclosporin A from tolypocladium species
US5571536A (en) 1995-02-06 1996-11-05 Nano Systems L.L.C. Formulations of compounds as nanoparticulate dispersions in digestible oils or fatty acids
US5560931A (en) 1995-02-14 1996-10-01 Nawosystems L.L.C. Formulations of compounds as nanoparticulate dispersions in digestible oils or fatty acids
SE9500478D0 (en) 1995-02-09 1995-02-09 Astra Ab New pharmaceutical formulation and process
US5573783A (en) 1995-02-13 1996-11-12 Nano Systems L.L.C. Redispersible nanoparticulate film matrices with protective overcoats
FI102758B (en) 1995-03-03 1999-02-15 Santen Pharmaceutical Co Ltd Process for producing pure cyclosporine form
JP2740153B2 (en) 1995-03-07 1998-04-15 エフ・ホフマン−ラ ロシユ アーゲー Mixed micelle
US6004583A (en) 1995-03-22 1999-12-21 Orex Pharmaceutical Development Corp. Protein-containing polymer composition for oral administration
IE75744B1 (en) 1995-04-03 1997-09-24 Elan Corp Plc Controlled release biodegradable micro- and nanospheres containing cyclosporin
US5965160A (en) 1995-04-24 1999-10-12 Yissum Research Development Company Of The Hebrew University Of Jerusalem Self-emulsifiable formulation producing an oil-in-water emulsion
US5653987A (en) 1995-05-16 1997-08-05 Modi; Pankaj Liquid formulations for proteinic pharmaceuticals
US5686106A (en) 1995-05-17 1997-11-11 The Procter & Gamble Company Pharmaceutical dosage form for colonic delivery
US5726181A (en) 1995-06-05 1998-03-10 Bionumerik Pharmaceuticals, Inc. Formulations and compositions of poorly water soluble camptothecin derivatives
US5656287A (en) 1995-06-07 1997-08-12 Nexstar Pharmaceuticals, Inc. Liposomal cyclosporin formulations as agents for immunosuppression and multiple drug resistant indications
US5665386A (en) 1995-06-07 1997-09-09 Avmax, Inc. Use of essential oils to increase bioavailability of oral pharmaceutical compounds
US5616595A (en) 1995-06-07 1997-04-01 Abbott Laboratories Process for recovering water insoluble compounds from a fermentation broth
US5716928A (en) 1995-06-07 1998-02-10 Avmax, Inc. Use of essential oils to increase bioavailability of oral pharmaceutical compounds
US5962019A (en) 1995-08-25 1999-10-05 Sangstat Medical Corporation Oral cyclosporin formulations
US5766629A (en) 1995-08-25 1998-06-16 Sangstat Medical Corporation Oral cyclosporin formulations
US5827822A (en) 1996-03-25 1998-10-27 Sangstat Medical Corporation Cyclosporin a formulations as nanoparticles
EP0760237A1 (en) 1995-08-30 1997-03-05 Cipla Limited Oil-in-water microemulsions
US5968972A (en) 1995-10-26 1999-10-19 Baker Norton Pharmaceuticals, Inc. Method for increasing the oral bioactivity of pharmaceutical agents
NZ280689A (en) 1995-12-15 1997-08-22 Bernard Charles Sherma Sherman Pharmaceutical composition comprising a cyclosporipharmaceutical composition comprising a cyclosporin; a tocol, tocopherol or tocotrienol; and propylen; a tocol, tocopherol or tocotrienol; and propylene carbonate or polyethylene glycol ne carbonate or polyethylene glycol
CZ288631B6 (en) 1996-01-18 2001-08-15 Galena, A. S. Therapeutic preparations containing cyclosporin
US5858401A (en) 1996-01-22 1999-01-12 Sidmak Laboratories, Inc. Pharmaceutical composition for cyclosporines
EP0787988B1 (en) 1996-02-05 2004-04-28 Hiroshi Prof. Handa Drug-immobilized particles and a process of purifying proteins
KR970064620A (en) 1996-03-05 1997-10-13 임성기 Cyclosporin-containing external-use pharmaceutical composition
DE19611094C2 (en) 1996-03-21 1999-06-17 Dresden Arzneimittel Process for the purification of cyclosporin A and / or related cyclosporins from a cyclosporin-containing crude extract using chromatographic processes using silica gel as the adsorbent
SE507682C2 (en) 1996-03-29 1998-07-06 Marcin Krotkiewski Pharmaceutical preparation for the treatment of gastritis, reflux oesophagitis, duodenitis, dyspepsia and peptic ulcer disease containing a mixture of ammonium bismuth citrate and an alginate
IL117773A (en) 1996-04-02 2000-10-31 Pharmos Ltd Solid lipid compositions of coenzyme Q10 for enhanced oral bioavailability
US5660858A (en) 1996-04-03 1997-08-26 Research Triangle Pharmaceuticals Cyclosporin emulsions
US6030941A (en) 1996-05-01 2000-02-29 Avi Biopharma, Inc. Polymer composition for delivering substances in living organisms
US5709797A (en) 1996-06-05 1998-01-20 Poli Industria Chimica S.P.A. Method of isolating cyclosporins
US5747330A (en) 1996-06-05 1998-05-05 Poli Industria Chimica Antibiotic producing microbe
EP0914132A1 (en) 1996-06-11 1999-05-12 Cellena Ag Eutrophic drug compositions based on transferrin glycans
US5958876A (en) 1996-06-19 1999-09-28 Novartis Ag Cyclosporin-containing pharmaceutical compositions
US5958378A (en) 1996-07-03 1999-09-28 Research Development Foundation High dose liposomal aerosol formulations containing cyclosporin A or budesonide
DE19629753A1 (en) 1996-07-23 1998-01-29 Basf Ag Process for the production of solid dosage forms
KR980008239A (en) 1996-07-26 1998-04-30 김충환 Cyclosporin-containing pharmaceutical composition
IL128632A (en) * 1996-08-22 2003-03-12 Rtp Pharma Corp Compositions comprising microparticles of water-insoluble substances and method for preparing same
CZ283516B6 (en) 1996-09-12 1998-04-15 Galena A.S. Therapeutical preparations, particularly for internal administration in the form of self-micro-emulsifying therapeutic systems
US5798333A (en) 1996-09-17 1998-08-25 Sherman; Bernard C. Water-soluble concentrates containing cyclosporins
US6048550A (en) 1996-10-03 2000-04-11 Chan; Daniel C. F. Hydrophilic microparticles and methods to prepare same
US6077543A (en) 1996-12-31 2000-06-20 Inhale Therapeutic Systems Systems and processes for spray drying hydrophobic drugs with hydrophilic excipients
JP3632183B2 (en) * 1997-01-28 2005-03-23 東洋電装株式会社 Discharge lamp unit
US6051257A (en) 1997-02-24 2000-04-18 Superior Micropowders, Llc Powder batch of pharmaceutically-active particles and methods for making same
DE19710213A1 (en) 1997-03-12 1998-09-17 Basf Ag Process for the manufacture of solid combination dosage forms
ATE286403T1 (en) 1997-03-12 2005-01-15 Abbott Lab HYDROPHILIC BINARY SYSTEMS FOR ADMINISTRATION OF CYCLOSPORINE
DE19720312A1 (en) 1997-05-15 1998-11-19 Hoechst Ag Preparation with increased in vivo tolerance
JPH1169867A (en) * 1997-08-11 1999-03-09 Matsushita Electric Ind Co Ltd Device and method for controlling and driving sensorless dc brushless motor
US5962522A (en) 1997-09-05 1999-10-05 Avmax, Inc. Propyl gallate to increase bioavailability of orally administered pharmaceutical compounds
US6008191A (en) 1997-09-08 1999-12-28 Panacea Biotec Limited Pharmaceutical compositions containing cyclosporin
US6010719A (en) 1997-09-16 2000-01-04 Universiteit Gent Freeze-dried disintegrating tablets
NZ328751A (en) 1997-09-16 1999-01-28 Bernard Charles Sherman Solid medicament containing an anionic surfactant and cyclosporin
US6004573A (en) 1997-10-03 1999-12-21 Macromed, Inc. Biodegradable low molecular weight triblock poly(lactide-co-glycolide) polyethylene glycol copolymers having reverse thermal gelation properties
US6027747A (en) 1997-11-11 2000-02-22 Terracol; Didier Process for the production of dry pharmaceutical forms and the thus obtained pharmaceutical compositions
US5891845A (en) 1997-11-21 1999-04-06 Fuisz Technologies Ltd. Drug delivery systems utilizing liquid crystal structures
US6028067A (en) 1997-12-05 2000-02-22 Chong Kun Dang Corp. Cyclosporin-containing microemulsion preconcentrate composition
US6063762A (en) 1997-12-05 2000-05-16 Chong Kun Dang Corp. Cyclosporin-containing microemulsion preconcentrate composition
US6013665A (en) 1997-12-16 2000-01-11 Abbott Laboratories Method for enhancing the absorption and transport of lipid soluble compounds using structured glycerides
FR2780061B1 (en) 1998-06-22 2001-09-07 Rhone Poulenc Rorer Sa NOVEL PROCESS FOR THE PREPARATION OF CYCLOSPORIN DERIVATIVES
US6051607A (en) 1998-07-02 2000-04-18 Micro Therapeutics, Inc. Vascular embolizing compositions comprising ethyl lactate and methods for their use
DE69939335D1 (en) * 1998-12-30 2008-09-25 Dexcel Ltd DISPERSIBLE CONCENTRATE FOR THE ADMINISTRATION OF CYCLOSPORIN
ATE249218T1 (en) 1999-01-06 2003-09-15 Tecnimede Sociedade Tecnico Medicinal Sa CYCLODEXTRIN INCLUSION COMPLEXES WITH AMINO ACID SALTS OF BENZIMIDAZOLE DERIVATIVES, THEIR PRODUCTION, AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
US6267985B1 (en) 1999-06-30 2001-07-31 Lipocine Inc. Clear oil-containing pharmaceutical compositions
US6045826A (en) 1999-04-02 2000-04-04 National Research Council Of Canada Water-soluble compositions of bioactive lipophilic compounds
US6057289A (en) 1999-04-30 2000-05-02 Pharmasolutions, Inc. Pharmaceutical composition comprising cyclosporin in association with a carrier in a self-emulsifying drug delivery system
US6388789B1 (en) 2000-09-19 2002-05-14 The Charles Stark Draper Laboratory, Inc. Multi-axis magnetically actuated device
WO2002025810A2 (en) * 2000-09-22 2002-03-28 U.S. Monolithics, L.L.C. Mmic folded power amplifier
BR0210505A (en) 2001-06-21 2004-05-18 Pfizer Prod Inc Self-emulsifying formulations of cholesteryl ester transfer protein inhibitors
US20050277584A1 (en) 2004-06-09 2005-12-15 Allergan, Inc. Pharmaceutical compositions comprising cyclosporins

Also Published As

Publication number Publication date
JP2009529003A (en) 2009-08-13
EP1988879A1 (en) 2008-11-12
US20060205639A1 (en) 2006-09-14
WO2007099537A1 (en) 2007-09-07
AU2007220104A1 (en) 2007-09-07
US7732404B2 (en) 2010-06-08

Similar Documents

Publication Publication Date Title
US7732404B2 (en) Pro-nanodispersion for the delivery of cyclosporin
US5916589A (en) Pharmaceutical compositions comprising cyclosporins
US7026290B1 (en) Dispersible concentrate for the delivery of cyclosprin
US5342625A (en) Pharmaceutical compositions comprising cyclosporins
SK13192000A3 (en) Microemulsion preconcentrate composition containing cyclosporin and use thereof
CA2380951C (en) Pharmaceutical compositions for oral and topical administration
US20020049158A1 (en) Oral drug composition containing a verapamil derivative as a drug-absorption promotor
EP0985412B1 (en) Cyclosporin compositions
EP1151755B1 (en) Pharmaceutical compositions comprising cyclosporin as active ingredient
JP2008518931A (en) Spontaneous dispersible pharmaceutical composition
KR100525234B1 (en) Soft capsule containing cyclosporin and it's manufacturing process
KR0173349B1 (en) Cyclosporin-containing solid dispersion composition
IL143604A (en) Dispersible concentrate for the delivery of cyclosporin
AU2009202703A1 (en) Pharmaceutical compositions for oral and topical administration

Legal Events

Date Code Title Description
FZDE Discontinued