WO2008155141A2 - Novel facultative catonic sterols - Google Patents

Novel facultative catonic sterols Download PDF

Info

Publication number
WO2008155141A2
WO2008155141A2 PCT/EP2008/005221 EP2008005221W WO2008155141A2 WO 2008155141 A2 WO2008155141 A2 WO 2008155141A2 EP 2008005221 W EP2008005221 W EP 2008005221W WO 2008155141 A2 WO2008155141 A2 WO 2008155141A2
Authority
WO
WIPO (PCT)
Prior art keywords
liposomes
chol
acid
sterol
lipid
Prior art date
Application number
PCT/EP2008/005221
Other languages
French (fr)
Other versions
WO2008155141A3 (en
Inventor
Gerold Endert
Cornelia Panzner
Original Assignee
Novosom Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novosom Ag filed Critical Novosom Ag
Priority to AU2008309880A priority Critical patent/AU2008309880B2/en
Priority to US12/682,703 priority patent/US20100330154A1/en
Priority to CA2702103A priority patent/CA2702103A1/en
Priority to JP2010528325A priority patent/JP5711535B2/en
Priority to PCT/EP2008/008621 priority patent/WO2009047006A2/en
Priority to EP08837321.2A priority patent/EP2211840B1/en
Publication of WO2008155141A2 publication Critical patent/WO2008155141A2/en
Publication of WO2008155141A3 publication Critical patent/WO2008155141A3/en
Priority to US14/049,669 priority patent/US20140178462A1/en
Priority to JP2015017794A priority patent/JP6192061B2/en
Priority to JP2017004104A priority patent/JP2017105792A/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/575Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of three or more carbon atoms, e.g. cholane, cholestane, ergosterol, sitosterol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/58Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids containing heterocyclic rings, e.g. danazol, stanozolol, pancuronium or digitogenin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/28Steroids, e.g. cholesterol, bile acids or glycyrrhetinic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • A61K9/1272Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers with substantial amounts of non-phosphatidyl, i.e. non-acylglycerophosphate, surfactants as bilayer-forming substances, e.g. cationic lipids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J41/00Normal steroids containing one or more nitrogen atoms not belonging to a hetero ring
    • C07J41/0033Normal steroids containing one or more nitrogen atoms not belonging to a hetero ring not covered by C07J41/0005
    • C07J41/0055Normal steroids containing one or more nitrogen atoms not belonging to a hetero ring not covered by C07J41/0005 the 17-beta position being substituted by an uninterrupted chain of at least three carbon atoms which may or may not be branched, e.g. cholane or cholestane derivatives, optionally cyclised, e.g. 17-beta-phenyl or 17-beta-furyl derivatives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J43/00Normal steroids having a nitrogen-containing hetero ring spiro-condensed or not condensed with the cyclopenta(a)hydrophenanthrene skeleton
    • C07J43/003Normal steroids having a nitrogen-containing hetero ring spiro-condensed or not condensed with the cyclopenta(a)hydrophenanthrene skeleton not condensed
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J9/00Normal steroids containing carbon, hydrogen, halogen or oxygen substituted in position 17 beta by a chain of more than two carbon atoms, e.g. cholane, cholestane, coprostane
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/582Recycling of unreacted starting or intermediate materials

Definitions

  • the present invention relates to novel facultative cationic sterols and liposomes comprising such sterols.
  • a particular aspect of the invention relates to amphoteric liposomes comprising the novel facultative cationic sterols.
  • Sterol derivatives of the general formula 1 are known in the art and are disclosed in WO 02/066490 by Panzner et al., the content of which is incorporated herein by reference.
  • cation is derived from piperazines, imidazoles, morpholines, purines, pyrimidines and /or pyridines;
  • the spacers 1 and 2 are selected from the group consisting of lower alkyl residues with up to 8 C atoms with a linear, branched or cyclic structure and 0, 1 or 2 ethylenically unsaturated bonds;
  • the steryl is selected from the group consisting of cholesteryl, sitosteryl, campesteryl, desmosteryl, fucosteryl, 22-ketosteryl, 20-hydroxysteryl, stigmasteryl, 22-hydroxycholesteryl, 25-hydroxycholesteryl, lanosteryl, 7-dehydrocholesteryl, dihydrocholesteryl, 19-hydroxycholesteryl, 5 ⁇ -cholest-7-en-3 ⁇ -yl,
  • WO 97/39019 of Deshmukh et al. describes different cationic sterol compounds with a carbamoyl linker group and cyclic N containing bases, such as morpholine. However, these compounds have only one spacer and no second linker group.
  • the object of the present invention was to provide additional, alternative sterol derivative of the general formula 1 having improved stability upon storage.
  • Another object of the present is the provision of liposomes comprising such sterol derivatives.
  • Yet another object of the present invention is to provide liposomes comprising such sterol derivatives and encapsulating an active agent.
  • Yet another object of the invention is to provide liposomes comprising such sterol derivatives and allow effective transfection of cells.
  • Yet another object of the invention is to provide pharmaceutical compositions comprising such liposomes as carriers for the delivery of active agents or ingredients.
  • facultative cationic sterol of general formula 1 cation - spacer 2 - Y - spacer 1 - X - steryl, (1)
  • the spacers 1 is a lower alkyl residue of linear, branched or cyclic structure, which has from 1 to 8 C atoms and includes O 1 1 or 2 or 3 ethylenically unsaturated bonds
  • the spacer 2 is a lower alkyl residue of linear, branched or cyclic structure, which has from 0 to 8 C atoms and includes 0, 1 or 2 or 3 ethylenically unsaturated bonds
  • sterol derivatives comprising the above mentioned linking groups X and/or Y show an improved stability. Therefore liposomes, comprising the novel lipids can be stored in suspension for extended periods of time.
  • the steryls may be selected from the group consisting of cholesteryl, sitosteryl, campesteryl, desmosteryl, fucosteryl, 22-ketosteryl, 20-hydroxysteryl, stigmasteryl, 22-hydroxycholesteryl, 25-hydroxycholesteryl, lanosteryl, 7-dehydrocholesteryl, dihydrocholesteryl, 19-hydroxycholesteryl, 5 ⁇ -cholest-7-en-3 ⁇ -yl, 7-hydroxy- cholesteryl, epicholesteryl, ergosteryl, dehydroergosteryl and derivatives thereof.
  • the nitrogen base may be selected from unsubstituted or substituted piperazines, imidazoles, morpholines, purines, pyrimidines and /or pyridines or may derive from single or multiple substituted nitrogen atoms with lower hydroxyalkyls.
  • inventive sterol derivatives have a pka value of between 5 and 8. In another embodiment of the invention the pka value of inventive sterol derivatives may be between 4 and 7.
  • Another aspect of the present invention relates to liposomes comprising the inventive cationic sterol derivatives.
  • the liposomes may further comprise one or more neutral and/or zwitterionic lipids and/or one or more anionic lipids and/or one or more cationic lipids.
  • the liposomes comprising the inventive cationic sterols are amphoteric liposomes.
  • Said amphoteric liposomes may comprise an anionic lipid selected from the group consisiting of DOGS, DMGS, Chol-C1 , Chol-C2, Chol-C3, Chems, Chol-C5, Chol-C6, Chol-C7 and/or Chol-C8.
  • the liposomes according to the invention may further comprise neutral or zwitterionic lipids, preferably selected from POPC, DPPC, DSPC, DOPC, DMPC, POPE, DPPE, DSPE, DMPE, DOPE, DPhyPE, DLinPE or natural equivalents thereof or cholesterol.
  • neutral or zwitterionic lipids preferably selected from POPC, DPPC, DSPC, DOPC, DMPC, POPE, DPPE, DSPE, DMPE, DOPE, DPhyPE, DLinPE or natural equivalents thereof or cholesterol.
  • said neutral and/or zwitterionic lipid is cholesterol or a mixture of phosphatidylethanolamine and cholesterol or a mixture of phosphatidylcholine and cholesterol.
  • said neutral and/or zwitterionic lipid is a mixture of phosphatidylethanolamine and phosphatidylcholine.
  • the size of the liposomes is of between 20 to 1000 nm.
  • the liposomes comprise an active agent.
  • the active agent is a nucleic acid drug, such as oligonucleotides or plasmids.
  • the inventive liposomes may be used as a vector for in vivo, in vitro or ex vivo transfection.
  • liposomes may be useful for intravenous, subcutaneous, peritoneal, local or topical application.
  • compositions comprising the inventive liposomes for the treatment or prophylaxis of inflammatory, immune or autoimmune disorders, cancer and/or metabolic diseases of humans or non-human animals.
  • the present invention relates to novel facultative cationic sterol derivatives of the general formula 1 :
  • the spacers 1 is a lower alkyl residue of linear, branched or cyclic structure, which has from 1 to 8 C atoms and includes 0, 1 or 2 or 3 ethylenically unsaturated bonds
  • the spacer 2 is a lower alkyl residue of linear, branched or cyclic structure, which has from 0 to 8 C atoms and includes 0, 1 or 2 or 3 ethylenically unsaturated bonds
  • the overall molecule assumes its pH-dependent charge characteristics by one or more nitrogen bases with a pKa value between 3.5 and 8. These nitrogen bases are linked to the 3-position of the sterol skeleton via spacers and coupling groups, thus forming a compound according to the formula of the invention.
  • nitrogen bases are in the form of a ring system
  • positional isomers are existing, wherein the linking spacer is substituted to various positions of the organic cation.
  • Such positional isomers fall within the disclosure of this invention.
  • the pKa values of the organic cation can be influenced via said positional isomerism alone.
  • the relevant fundamental rules are well-known to those skilled in the art. Alternatively, these effects can be estimated from tabular compilations
  • Preferred pka values of the inventive facultative cationic sterol derivatives may depend on the appropriate application of the lipid.
  • the pka value of the sterol derivative may be between 5 and 8.
  • the sterol derivative may have a pKa value of between 4 and 7.
  • these pKa values fall in a range which is of crucial importance for the physiology of numerous organisms.
  • the cations are nitrogen bases.
  • the cations may be derived from piperazines, imidazoles, morpholines, purines, pyrimidines, pyridines, piperidines, anilines, anisidines, toluidines, phenetidines, benzimidazoles, isochinolines, picolines, pterines, purines, pyrazines, pyridazines, chinolines, thiazoles or triazines, pyrazols, imidazolines, imidazolidins, pyrazolines, pyrazolidines or hydrazins or derivatives thereof.
  • the cations preferably can be derived from piperazines, imidazoles, morpholines, purines, pyrimidines and/or pyridines or derivatives thereof.
  • Coupling reactions result in amphiphilic organic cations, e.g. those derived from the following classes of substances: o-, m-, p-anilines; 2-, 3- or 4-substituted anisidines, toluidines or phenetidines; 2-, 3-, 5-, 6-, 7- or 8-substituted benzimidazoles, 2-, 3-, 4- or 5-substituted imidazoles, 1- or 5-substituted isoquinolines, 2-, 3- or 4-substituted morpholines, 2-, 3- or 4-substituted picolines, 2-, 3- or 4-substituted piperidines, 1-, 2- or 3-substituted piperazines, 2-, 5- or 6-modified pterines, 3-, A-, 5-, 6- or 9-substituted purines, 2- or 3-substituted pyrazines, 3- or 4-substituted pyridazines, 2-, 3- or 4-modified pyridine
  • cations are imidazoles and/or morpholines or derivatives thereof.
  • molecule fragments such as occurring in biological systems are preferred, i.e., for example: 4-imidazoles (histamines), 2-, 6- or 9-purines (adenines, guanines, adenosines, or guanosines), 1-, 2- or 4-pyrimidines (uracils, thymines, cytosines, uridines, thymidines, cytidines), or pyridine-3-carboxylic acids (nicotinic esters or amides).
  • 4-imidazoles histamines
  • 2-, 6- or 9-purines adenines, guanines, adenosines, or guanosines
  • 1-, 2- or 4-pyrimidines uracils, thymines, cytosines, uridines, thymidines, cytidines
  • pyridine-3-carboxylic acids nicotinic esters or amides
  • the above-mentioned structural fragments may also have additional substituents.
  • these can be methyl, ethyl, propyl, or isopropyl residues, more preferably in hydroxy lated form, including one or two hydroxy! groups.
  • these can be hydroxyl or keto functions in the ring system.
  • Nitrogen bases with preferred pKa values are also formed by single or multiple substitution of the nitrogen atom with lower alkanehydroxyls such as hydroxymethyl or hydroxyethyl groups.
  • Suitable organic bases from this group are e.g. aminopropanediols, thethanolamines, tris(hydroxymethyl)methylamines, bis(hydroxymethyl)methylamines, tris(hydroxyethyl)methylamines, bis(hydroxyethyl)- methylamines, or the corresponding substituted ethylamines. Coupling of these fragments to the hydrophobic portion of the molecule may proceed either via the nitrogen of the base or via any of the hydroxyl functions.
  • sterol derivatives including a single organic cation
  • those including two or three identical or different groups are also preferred. All of these groups are required to have a pKa value in the above-mentioned range.
  • One suitable complex group is the amide of histamine and histidine or of histamine and histidylhistidine.
  • spacer 1 is a lower alkyl residue of linear, branched or cyclic structure, which has from 1 to 8 C atoms and includes O, 1 or 2 or 3 ethylenically unsaturated bonds. Spacer 1 may have hydroxyl groups so as to increase the polarity of the molecule. In particular, spacer 1 can be a sugar or sugaralcohol.
  • Spacer 2 is a lower alkyl residue of linear, branched or cyclic structure, which has from 0 to 8 C atoms and includes 0, 1 or 2 or 3 ethylenically unsaturated bonds. Spacer 2 may have hydroxy! groups so as to increase the polarity of the molecule.
  • spacer 2 can be a sugar or sugaralcohol.
  • spacer 2 is a lower alkyl residue of linear, branched or cyclic structure, which has from 1 to 8 C atoms and includes 0, 1 or 2 or 3 ethylenically unsaturated bonds.
  • inventive sterol derivatives show an improved stability compared to the lipid MoChol as described in example 5.
  • the steryls are particularly cholesteryl, sitosteryl, campesteryl, desmosteryl, fucosteryl, 22-ketosteryl, 20- hydroxysteryl, stigmasteryl, 22-hydroxycholesteryl, 25-hydroxycholesteryl, lanosteryl, 7-dehydrocholesteryl, dihydrocholesteryl, 19-hydroxycholesteryl, 5 ⁇ -cholest-7-en-3 ⁇ - yl, 7-hydroxycholesteryl, epicholesteryl, ergosteryl, and/or dehydroergosteryl, as well as other related compounds.
  • the steryls that are used may bear various groups in the 3-position thereof, which groups allow for ready and stable coupling or optionally assume the function of a spacer. Particularly suitable for direct coupling are the hydroxyl group which is naturally present, but also, the chlorine of steryl chlorides, or e.g. the amino group of sterylamines, or the thiol group of thiocholesterol.
  • facultative cationic sterols preferably have the following general formula 2:
  • inventive sterol may comprise, but not limited to, one of the following structures:
  • the invention also relates to liposomes comprising the facultative cationic sterols according to the invention.
  • the inventive facultative cationic sterols can be incorporated in high amounts in liposomal membranes.
  • the amount of the inventive sterol derivative in the liposomal membrane is between 3 and 100 mol%, preferred between 5 and 80 mol% and particularly preferred between 7 and 70 mol%.
  • the liposomes may comprise one or more of the inventive lipids and one or more neutral and/or zwitterionic lipid.
  • examples of such lipids include phosphatidyl choline, phosphatidyl ethanolamine, sphingomylein, ceramides, diacylglycerol and/or cholesterol.
  • the phosphatidylcholine may be selected from the group comprising POPC, natural or hydrogenated soya PC, natural or hydrogenated egg PC, DMPC, DPPC, DSPC or DOPC.
  • the phosphatidylethanolamines are preferably selected from the group comprising DOPE, DMPE or DPPE or POPE, DPhyPE, DLinPE, DSPE or natural equivalents thereof.
  • liposomes comprising the inventive lipids may comprise one or more anionic lipids and/or one or more cationic lipids. Examples of anionic and cationic lipids are given below in this disclosure.
  • the liposomes comprising the inventive lipids are amphoteric liposomes.
  • Amphoteric liposomes are a new class of liposomes described in WO 02/066012. Amphoteric liposomes have an anionic or neutral charge at pH 7.5 and a cationic charge at pH 4.
  • These liposomes can be prepared from lipid mixtures comprising either an amphoteric lipid or a mixture of lipids with amphoteric character and optional a neutral and/or zwitterionic lipid.
  • amphoteric liposomes comprising the inventive sterol lipids according to the present invention can be formed from any amphoteric lipid mixture.
  • the amphoteric lipid mixture may comprise a plurality of charged amphiphiles which in combination with one another have amphoteric character.
  • a selection of charged amphiphiles is summarized below in this disclosure.
  • said one or more charged amphiphiles comprise a pH sensitive anionic lipid and a pH sensitive cationic lipid.
  • such a combination of a chargeable cation and chargeable anion is referred to as an "amphoteric II" lipid pair.
  • said chargeable cations have pKa values of between about 5 and about 8.
  • said chargeable cation is a facultative cationic sterol according to the present invention.
  • said one or more charged amphiphiles comprise a stable anion and a chargeable cation and is referred to as "amphoteric III" lipid pair.
  • said chargeable cations have pKa values of between about 4 and about 7.
  • said chargeable cation is a facultative cationic sterol according to the present invention.
  • amphiphiles with multiple charges such as amphipathic dicarboxylic acids, phosphatidic acid, amphipathic piperazine derivatives and the like.
  • Such multicharged amphiphiles might fall into pH sensitive amphiphiles or stable anions or cations or might have mixed character.
  • Suitable anionic lipids for the formation of amphoteric liposomes in combination with the inventive cationic sterols include, but are not limited to diacylglycerolhemisuccinates, e.g. DOGS, DMGS, POGS, DPGS, DSGS; diacylglycerolhemimalonat.es, e.g. DOGM or DMGM; diacylglycerolhemiglutarates, e.g. DOGG, DMGG; diacylglycerolhemiadipates, e.g. DOGA, DMGA; diacylglycerolhemicyclohexane-1 ,4-dicarboxylic acids, e.g.
  • diacylglycerolhemisuccinates e.g. DOGS, DMGS, POGS, DPGS, DSGS
  • diacylglycerolhemimalonat.es e.g. DOGM or DMGM
  • DO-cHA, DM-cHA; (2,3- Diacyl-propyl)amino ⁇ -oxoalkanoic acids e.g. DOAS 1 DOAM, DOAG, DOAA, DMAS, DMAM 1 DMAG, DMAA; Diacyl-alkanoic acids, e.g. DOP 1 DOB, DOS, DOM 1 DOG 1 DOA, DMP 1 DOB, DMS, DMM, DMG 1 DMA; Chems and derivatives therof, e.g. Chol- C2, Chol-C3, Chol-C5 or Chol-C6, Chol-C7, Chol-C8; Chol-C1 , fatty acids, e.g.
  • Oleic acid Linoleic Acid, Myristic Acid, Palmitic acid, Stearic acid, Nervonic Acid, Behenic Acid
  • dialkyl derivatives of these anionic lipids comprising diacyl groups are also within the scope of the present invention.
  • Preferred anionic lipids for the formation of amphoteric liposomes in combination with the inventive cationic sterols may be selected from the group consisting of DOGS, DMGS, Chol-CI , Chol-C2, Chol-C3, Chems, Chol-C5, Chol-C6, Chol-C7 and/or Chol-C8.
  • amphoteric lipid mixtures further comprise one or more neutral and/or zwitterionic lipids.
  • examples of such lipids may include phosphatidyl choline, phosphatidyl ethanolamine, sphingomylein, ceramides, diacylglycerol and/or cholesterol.
  • said neutral lipids may be cholesterol or a mixture of cholesterol and phosphatidylethanolamines.
  • Preferably said lipids are present in the amphoteric liposomes as sole neutral lipids.
  • the molar ratio of the mixtures of cholesterol and phosphatidylethanolamine is between 4 and 0.25, preferred between 3 and 0.5 and most preferred between 2 and 1.
  • phosphatidylethanolamines may include POPE, DPPE, DSPE, DMPE, DOPE, DPhyPE, DLJnPE or natural equivalents thereof. Mixtures of different phosphatidylethanolamines are also within the scope of the present invention. In a preferred embodiment of the invention the phosphatidylethanolamine is DOPE.
  • a mixture of neutral lipids such as phosphatidylcholines (PC), sphingomyelins or ceramides and cholesterol (Choi) may be used as sole neutral lipid components in the amphoteric liposomes.
  • PC phosphatidylcholines
  • Choi cholesterol
  • PC/Choi may be between 4 and 0.25 or between 3 and 0.33. Preferred are molar ratios of PC/Choi between 1.5 and 0,25, more preferred between 1 and 0.25.
  • the phosphatidylcholines may be selected without limitation from the group POPC, DOPC, DMPC, DPPC, DSPC or natural equivalents thereof, such as soy bean PC or egg-PC. Mixtures of different phosphatidylcholines are also within the scope of the present invention. In a preferred embodiment of the invention the phosphatidylcholine is selected from POPC or DOPC.
  • a mixture of phosphatidylcholines, sphingomyelins or ceramides and phosphatidylethanolamines may be used as sole neutral lipid components in the amphoteric liposomes.
  • the phosphatidylcholine is POPC and the phosphatidylethanolamine is DOPE.
  • lipids or compounds which sterically stabilize liposomes and thereby avoid an uptake of the particles by the RES (reticuloendothelial system) upon injection of the particles into the blood stream include for example polyglycerols, dextranes, polysialic acids, hydroxyethyl starches, hyaluronic acids, PEGylated lipids, sugar alcohols, Tween 80 or GM 1 gangliosides.
  • lipids or detergents are known in the art to sterically stabilize liposomes (e.g. Woodle et al., Biochim. Biophys. Acta, 1113(2), 171-179, (1992); Allen et al., Biochim. Biophys. Acta, 981(1), 27-35, (1989)).
  • the liposomes or amphoteric liposomes according to the present invention have an average size of between 50 and 1000 nm, preferably between 50 and 300 nm, and more preferably between 60 and 130 nm. In another embodiment the size of the liposomes or amphoteric liposomes may be less than 50nm, preferably of between 20 and 50 nm.
  • the liposomes comprise active substances.
  • the liposomes according to the invention are suitable for parenteral application. They can be used e.g. in cancer therapy and in the therapy of severe infections.
  • liposome dispersions can be injected, infused or implanted. Thereafter, they are distributed in the blood or lymph or release their active substance in a controlled fashion as a depot. The latter can be achieved by highly concentrated dispersions in the form of gels.
  • the liposomes can also be used for topical application on the skin. In particular, they may contribute to improved penetration of various active substances into the skin or even passage through the skin and into the body. Furthermore, the liposomes can also be used in gene transfer.
  • liposomes of the invention can also be used as model membranes. In their principal structure, liposomes are highly similar to cell membranes. Therefore, they can be used as membrane models to quantify the permeation rate of active substances through membranes or the membrane binding of active substances.
  • the liposomes in the present invention are well suited for use as carriers for active ingredients such as for example proteins, peptides, nucleic acids.
  • the active ingredient is a nucleic acid.
  • the nucleic acid-based therapeutic may comprise a nucleic acid that is capable of being transcribed in a vertebrate cell into one or more RNAs, which RNAs may be mRNAs, shRNAs, miRNAs or ribozymes, wherein such mRNAs code for one or more proteins or polypeptides.
  • nucleic acid therapeutics may be circular DNA plasmids, linear DNA constructs, like MIDGE vectors (Minimalistic lmmunogenically Defined Gene Expression) as disclosed in WO 98/21322 or DE 19753182, or mRNAs ready for translation (e.g., EP 1392341).
  • MIDGE vectors Minimalistic lmmunogenically Defined Gene Expression
  • oligonucleotides may be used that can target existing intracellular nucleic acids or proteins.
  • Said nucleic acids may code for a specific gene, such that said oligonucleotide is adapted to attenuate or modulate transcription, modify the processing of the transcript or otherwise interfere with the expression of the protein.
  • target nucleic acid encompasses DNA encoding a specific gene, as well as all RNAs derived from such DNA, being pre-mRNA or mRNA. A specific hybridisation between the target nucleic acid and one or more oligonucleotides directed against such sequences may result in an inhibition or modulation of protein expression.
  • the oligonucleotide should suitably comprise a continuous stretch of nucleotides that is substantially complementary to the sequence of the target nucleic acid. Oligonucleotides fulfilling the abovementioned criteria may be built with a number of different chemistries and topologies.
  • the oligonucleotides may comprise naturally occurring or modified nucleosides comprising but not limited to DNA, RNA, locked nucleic acids (LNA's), 2'O-methyl RNA (2'Ome), 2' O-methoxyethyl RNA (2 1 MOE) in their phosphate or phosphothioate forms or Morpholinos or peptide nucleic acids (PNA 1 S). Oligonucleotides may be single stranded or double stranded.
  • Oligonucleotides are polyanionic structures having 8-60 charges. In most cases these structures are polymers comprising nucleotides.
  • the present invention is not limited to a particular mechanism of action of the oligonucleotides and an understanding of the mechanism is not necessary to practice the present invention.
  • the mechanisms of action of oligonucleotides may vary and might comprise inter alia effects on splicing, transcription, nuclear-cytoplasmic transport and translation.
  • single stranded oligonucleotides may be used, including, but not limited t ⁇ DNA-based oligonucleotides, locked nucleic acids, 2'-modified oligonucleotides and others, commonly known as antisense oligonucleotides.
  • Backbone or base or sugar modifications may include, but are not limited to, Phosphothioate DNA (PTO), 2'O-methyl RNA (2'Ome), 2'Fluoro RNA (2'F), 2' O- methoxyethyl-RNA (2'MOE), peptide nucleic acids (PNA), N3"-P5' phosphoamidates (NP) 1 2'fluoroarabino nucleic acids (FANA), locked nucleic acids (LNA), Morpholine phosphoamidate (Morpholino), Cyclohexene nucleic acid (CeNA), tricyclo-DNA (tcDNA) and others.
  • PTO Phosphothioate DNA
  • PTO Phosphothioate DNA
  • 2'Ome 2'O-methyl RNA
  • 2'F 2'Fluoro RNA
  • 2'MOE 2' O- methoxyethyl-RNA
  • PNA peptide nucleic acids
  • NP N3"-P5'
  • RNA molecules containing the complementary sequence motifs are known as siRNA molecules in the art (e.g., WO 99/32619 or WO 02/055693).
  • siRNA molecules comprise single stranded siRNAs or double stranded siRNAs having one non-continuous strand.
  • various chemistries were adapted to this class of oligonucleotides.
  • DNA / RNA hybrid systems are known in the art.
  • decoy oligonucleotides can be used. These double stranded DNA molecules and chemical modifications thereof do not target nucleic acids but transcription factors. This means that decoy oligonucleotides bind sequence-specific DNA-binding proteins and interfere with the transcription (e.g., Cho-Chung, et al. in Curr. Opin. MoI. Then, 1999).
  • oligonucleotides that may influence transcription by hybridizing under physiological conditions to the promoter region of a gene may be used. Again various chemistries may adapt to this class of oligonucleotides.
  • DNAzymes may be used. DNAzymes are single-stranded oligonucleotides and chemical modifications therof with enzymatic activity. Typical DNAzymes, known as the "10-23" model, are capable of cleaving single-stranded RNA at specific sites under physiological conditions.
  • the 10-23 model of DNAzymes has a catalytic domain of 15 highly conserved deoxyribonucleotides, flanked by 2 substrate-recognition domains complementary to a target sequence on the RNA. Cleavage of the target mRNAs may result in their destruction and the DNAzymes recycle and cleave multiple substrates.
  • Ribozymes can be used. Ribozymes are single-stranded oligoribonucleotides and chemical modifications thereof with enzymatic activity. They can be operationally divided into two components, a conserved stem-loop structure forming the catalytic core and flanking sequences which are reverse complementary to sequences surrounding the target site in a given
  • RNA transcript Flanking sequences may confer specificity and may generally constitute 14-16 nt in total, extending on both sides of the target site selected.
  • ⁇ aptamers may be used to target proteins.
  • Aptamers are macromolecules composed of nucleic acids, such as RNA or DNA, and chemical modifications thereof that bind tightly to a specific molecular target and are typically 15-60 nt long.
  • the chain of nucleotides may form intramolecular interactions that fold the molecule into a complex three-dimensional shape.
  • the shape of the aptamer allows it to bind tightly against the surface of its target molecule including but not limited to acidic proteins, basic proteins, membrane proteins, transcription factors and enzymes. Binding of aptamer molecules may influence the function of a target molecule.
  • oligonucleotides may vary in length between as little as 5 or 10, preferably 15 and even more preferably 18, and 50, preferably 30 and more preferably 25, nucleotides per strand. More specifically, the oligonucleotides may be antisense oligonucleotides of 8 to 50 nucleotides length that catalyze RNAseH mediated degradation of their target sequence or block translation or re-direct splicing or act as antogomirs; they may be siRNAs of 15 to 30 basepairs length; they may further represent decoy oligonucleotides of 15 to 30 basepairs length; can be complementary oligonucleotides influencing the transcription of genomic DNA of 15 to 30 nucleotides length; they might further represent DNAzymes of 25 to 50 nucleotides length or hbozymes of 25 to 50 nucleotides length or aptamers of 15 to 60 nucleotides length.
  • oligonucleotides are often functionally defined and can be identical or different or share some, but not all features of their chemical nature or architecture without substantially affecting the teachings of this invention.
  • the fit between the oligonucleotide and the target sequence is preferably perfect with each base of the oligonucleotide forming a base pair with its complementary base on the target nucleic acid over a continuous stretch of the abovementioned number of oligonucleotides.
  • the pair of sequences may contain one or more mismatches within the said continuous stretch of base pairs, although this is less preferred.
  • nucleic acids In general the type and chemical composition of such nucleic acids is of little impact for the performance of the inventive liposomes as vehicles be it in vivo or in vitro and the skilled artisan may find other types of oligonucleotides or nucleic acids suitable for combination with the inventive liposomes.
  • liposomes produced using the sterol derivatives of the invention show low non-specific binding to cell surfaces. It is this low non-specific binding which is an essential precondition for achieving specific binding to target cells.
  • Target control of the vehicles is obtained when providing the above-described liposomes with additional ligands.
  • the active substance can be accumulated specifically in such cells or tissues which exhibit a pathological condition.
  • the facultative cationic sterols according to the invention is therefore in the construction of vectors for transfer of active substances in living organisms.
  • the vectors are particularly suited for the transport of therapeutic macromolecules such as proteins or DNA which themselves are incapable of penetrating the cell membrane or undergo rapid degradation in the bloodstream.
  • antibodies, lectins, hormones or other active substances can be coupled to the surface of liposomes under mild conditions in high yields.
  • At least 80% of the active substance is inside the liposome.
  • the liposomes comprise non-encapsulated active agents.
  • liposomes The preparation of liposomes is well known in the art (e.g. Liposomes, Ed. V.P.
  • liposomes include, but are not limited to, the lipid film/hydration procedure with subsequent optional extrusion or sonication or the injection of an ethanolic lipid solution in water or buffer.
  • Amphoteric liposomes offer the distinct advantage that they can encapsulate nucleic acids with high efficiencies as disclosed for example in WO 02/066012 or WO 07/107304 both of Panzner et al..
  • the invention also relates to the use of the liposomes in the production of nanocapsules.
  • the invention also relates to the use of the liposomes in the production of release systems in diagnostics.
  • the liposomes are used for the transport and/or release of active substances.
  • the liposomes conveniently are used as depot formulation and/or as circulative depot.
  • the liposomes can be used in intravenous or peritoneal application.
  • the liposomes are used with advantage as vector to transfect cells in vivo, in vitro and ex vivo.
  • a further aspect of the invention relates to pharmaceutical compositions comprising liposomes comprising the cationic sterol derivatives according to the present invention preferably as a carrier for the delivery of active agents or ingredients, including drugs such as nucleic acid drugs, e.g., oligonucleotides and plasmids.
  • the pharmaceutical composition of the present invention may be formulated in a suitable pharmacologically acceptable vehicle. Vehicles such as water, saline, phosphate buffered saline, D5W and the like are well known to those skilled in the art for this purpose.
  • compositions according to the invention may be used particularly for the treatment or prophylaxis of inflammatory, immune or autoimmune disorders, cancer and/or metabolic diseases of humans or non-human animals.
  • a yet further aspect of the present invention relates to methods for the treatment of human or non-human animals in which said pharmaceutical composition comprising the inventive liposomes or amphoteric liposomes are targeted to a specific organ or organs, tumours or sites of infection or inflammation.
  • amphiphile By “chargeable” is meant that the amphiphile has a pK in the range pH 4 to pH 8. A chargeable amphiphile may therefore be a weak acid or base. A “stable” amphiphile is a strong acid or base, having a substantially stable charge on the range pH 4 to pH 8.
  • amphoteric herein is meant a substance, a mixture of substances or a supra- molecular complex (e.g., a liposome) comprising charged groups of both anionic and cationic character wherein:
  • At least one, and optionally both, of the cation and anionic amphiphiles is chargeable, having at least one charged group with a pK between 4 and 8,
  • the substance or mixture of substances has an isoelectric point of neutral net charge between pH 4 and pH 8.
  • Amphoteric character is by this definition different from zwittehonic character, as zwitterions do not have a pK in the range mentioned above. In consequence, zwitterions are essentially neutrally charged over a range of pH values; phosphatidylcholines and phosphatidylethanolamines are neutral lipids with zwitterionic character.
  • C/A or “C/A ratio” or “CVA molar ratio” herein is meant the molar ratio of cationic amphiphiles to anionic amphiphiles in a mixture of amphiphiles.
  • IC50 herein is meant the inhibitory concentration of an oligonucleotide leading to a 50 % knockdown of a target mRNA or in case of a proliferation assay to a 50 % inhibition of cell viability.
  • fracultative cationic herein is meant that the charge of a cation or a base depends on the pH of the medium.
  • lipids includes specific examples of neutral, zwitterionic, anionic, cationic or amphoteric lipids mentioned in this disclosure.
  • the lipid list by no means limits the scope of this disclosure.
  • abbreviations for the lipids are used herein, the majority of which abbreviations are in standard use in the literature:
  • DlinPE Dilinoleoylphosphatidylethanolamine
  • dialkyl derivatives of the neutral or zwitterionic lipids comprising diacyl groups listed above are also within the scope of the present invention.
  • DMGM Dimyristoylglycerolhemimalonate
  • DMGG Dimyristoylglycerolhemiglutarate
  • DMG-GIuA Dimyristoylglycerol-glucoronic acid (1- or 4-linked) DO-cHA Dioleoylglycerolhemicyclohexane-1 ,4-dicarboxylic acid
  • the diacylglycerols may be dimyristoyl-, dipalmitoyl-, dioleoyl-, distearoyl- or palmitoyloleoylglycerols and R in the table above includes any selection from this group or cholesterol.
  • dialkyl derivatives of the anionic lipids comprising diacyl groups listed above are also within the scope of the present invention.
  • DMTAP 1 2-Dimyristoyl-3-Trimethylammonium-Propane
  • DPTAP 1 2-Dipalmitoyl-3-Trimethylammonium-Propane
  • DODMHEAP 1 2-Dioleoyl-3-dimethylhydroxyethylammonium-Propane
  • DPDMHEAP 1 2-Dipalmitoyl-3-dimethylhydroxyethylammonium-Propane
  • DSDMHEAP 1 2-Distearoyl-3-dimethylhydroxyethylammonium-Propane
  • DOMDHEAP 1 2-Dioleoyl-3-methyldihydroxyethylammonium-Propane
  • DMMDHEAP 1 2-Dimyristoyl-3-methyldihydroxyethylammonium-Propane
  • DODAP 1 2-Dioleoyl-3-Dimethylammonium-Propane
  • DMDAP 1 2-Dimyristoyl-3-Dimethylammonium-PiOpane
  • DPMHEAP 1 2-Dipalmitoyl-3-methylhydroxyethylammonium-Propane
  • DSMHEAP 1 2-Distearoyl-3-methylhydroxyethy!ammonium-Propane
  • DSDHEAP 1 2-Distearoyl-3-dihydroxyethylammonium-Propane
  • DOEPC 1 ⁇ -Dioleoyl-sn-Glycero-S-Ethylphosphocholine
  • DMEPC i ⁇ -Dimyristoyl-sn-Glycero-S-Ethylphosphocholine
  • NeoPhectinTM cationic cardiolipins e.g. [1 , 3-Bis-(1 , 2-bis-tetradecyloxy- propyl-3-dimethylethoxyammonium bromide)-propane-2-ol]
  • the diacylglycerols may be dimyristoyl-, dipalmitoyl-, dioleoyl-, distearoyl- or palmitoyloleoylglycerols and R in the table above includes any selection from this group or R may be a C 6 -C 30 dialkyl, with 0-3 unsaturated bonds.
  • HistDG 1 ,2 Dipalmitoylglycerol-hemisuccinat-N_-Histidinyl- hemisuccinate, & Distearoyl- .Dimyristoyl, Dioleoyl or palmitoyl-oleoylderivatives
  • dialkyl derivatives of the amphoteric lipids comprising diacyl groups listed above are also within the scope of the present invention.
  • Figure 1 shows IC 50 values of amphoteric liposomes encapsulating siRNA targeting Plk-1 derived from the in vitro transfection of HeIa cells as described in example 6. Shown are mixtures of MoChol/DMGS, CHIM/DMGS, CholC4N-Mo2/DMGS and CholC3N-Mo2/DMGS with a C/A ratio of 0.33 and increasing amounts of Choi as neutral lipid (0-60 mol%). The bars marked with an asterisk indicate that the IC 50 values are higher than the tested dose range (75 or 100 nM siRNA, respectively).
  • Figure 2 shows IC 50 values of amphoteric liposomes encapsulating siRNA targeting Plk-1 derived from the in vitro transfection of HeIa cells as described in example 6. Shown are mixtures of MoChol/DMGS, CHIM/DMGS, CholC4N-Mo2/DMGS and CholC3N-Mo2/DMGS with a C/A ratio of 0.5 and increasing amounts of Choi as neutral lipid(0-60 mol%). The bars marked with an asterisk indicate that the IC 50 values are higher than the tested dose range (75 or 100 nM siRNA, respectively).
  • Figure 5 shows the % cell viability (normalized to mock treated cells) of HeIa cells transfected with different amphoteric liposomes encapsulating siRNA targeting Plk-1 (black bars) or non-targeting scrambled siRNA (grey bars). Shown are following amphoteric liposome formulations: MoChol/DMGS/Chol 17.5:52.5:30 (molar ratio), CHIM/DMGS/Chol 17.5:52.5:30 (molar ratio),CholC4N-Mo2/DMGS/Chol 17.5:52.5:30 (molar ratio) and CholC3N-Mo2/DMGS/Chol 17.5:52.5:30 (molar ratio).
  • Figure 6 shows the % cell viability (normalized to mock treated cells) of HeIa cells transfected with different amphoteric liposomes encapsulating siRNA targeting Plk-1 (black bars) or non-targeting scrambled siRNA (grey bars).
  • amphoteric liposome formulations MoChol/DMGS/POPC/Chol 26.7:53.3:6.7:13.3 (molar ratio), CHIM/DMGS/POPC/Chol 26.7:53.3:6.7:13.3 (molar ratio), CholC4N- Mo2/DMGS/POPC/Chol 26.7:53.3:6.7:13.3 (molar ratio), CholC3N- Mo2/DMGS/POPC/Chol 26.7:53.3:6.7:13.3 (molar ratio).
  • Example 1 is given with the understanding of further detailing certain aspects of practising the current invention. The examples by no means limit the scope of this disclosure.
  • Example 1 is given with the understanding of further detailing certain aspects of practising the current invention. The examples by no means limit the scope of this disclosure.
  • Example 1 is given with the understanding of further detailing certain aspects of practising the current invention. The examples by no means limit the scope of this disclosure.
  • Example 1 is given with the understanding of further detailing certain aspects of practising the current invention. The examples by no means limit the scope of this disclosure.
  • Step 1 Synthesis of [(2-Morpholine-4-yl-ethylcarbamoyl)methyl]-carbamic acid tert- butyl ester
  • the crude product was purified by column chromatography at silica gel (eluent: ethyl acetate : methanol 1 :1 + 1% NH 4 OH).
  • the product a colourless oil, was characterized by 1 H-NMR.
  • Step 3 Synthesis of [(2-Morpholine-4-y!-ethy!carbamoy!methy!]-carbarriic acid cholesteryl ester (1)
  • Step 1 Synthesis of [2-(2-Morpholine-4-yl-ethylcarbamoyl)-ethyl]-carbamic acid tert- butyl ester
  • Step 3 Synthesis of [2-(2-Morpholine-4-yl-ethylcarbamoyl)-ethyl]-carbamic acid cholesteryl ester (2)
  • Step 1 Synthesis of [(S-Morpholine ⁇ -yl-propylcarbamoyO-methyll-carbamic acid tert.-butyl ester
  • Step 3 Synthesis of [(3-M ⁇ holine-4-yl-propylcarbamoyl)-methyl]-carbamic acid cholesteryl ester (3) 5.8 g 2-amino-N-(3-morpholine-4-yl-propyl)-acetamid bistrifluoroacetate were dissolved in 150 ml chloroform. Then 13.3 g triethylamine and 3.94 g cholesteryl chloroformiate were added. The reaction mixture was stirred at room temperature overnight. Then the solvent was removed by rotary evaporation and the crude product, a colourless oil, dissolved in 200 ml ethyl acetate and washed twice with water. The organic phase was dried over Na 2 SO 4 and after filtration the solvent was removed by rotary evaporation. The product was dried by lyophilization and characterized by 1 H-NMR and LC-MS.
  • Step 3 Synthesis of [1-Methyl-2-(2-Morpholine-4-yl-ethylcarbamoyl)-propyl]-carbamic acid tert. butyl ester
  • Step 5 Synthesis of [i-Methyl ⁇ -Morpholine ⁇ -yl-ethylcarbamoylJ-propy ⁇ -carbamic acid cholesteryl ester (4)
  • the lipids (see table 6) were dissolved in isopropanol to a final lipid concentration of 30 mM. The lipid solutions were then mixed with PBS resulting in a final lipid concentration of 1 mM and a final isopropanol amount of 3 %. Stability test:
  • Stability of the lipids was tested under stress conditions, including acidic, alkaline and oxidative conditions at room temperature. In addition, one aliquot of the lipid suspensions was incubated at 60 0 C for three days. The conditions of the stability test are summarized in table 5 below.
  • HPTLC High Performance Thin Layer Chromatography
  • Amphoteric liposomes comprising lipids according to the present invention were used for tranfection of HeIa cells.
  • Liposomes were manufactured by an isopropanol-injection method. Lipids were dissolved in isopropanol (30 mM lipid concentration) and mixed. Liposomes were produced by adding siRNA solution in NaAc 2OmM, Sucrose 300 mM, pH 4.0 (pH adjusted with HAc) to the alcoholic lipid mix, resulting in a final alcohol concentration of 30% and a N/P ratio 5. The formed liposomal suspensions were shifted to pH 7.5 with twice the volume of Na2HPO4 136mM, NaCI 10OmM (pH 9), resulting in a final lipid concentration of 3 mM and a final isopropanol concentration of 10%.
  • N/P the ratio cationic charges from the lipids to anionic charges from the siRNA during manufacturing.
  • HeLa cells were obtained from DSMZ (German Collection of Micro Organism and Cell Cultures) and maintained in DMEM. Media were purchased from Gibco- Invitrogen and supplemented with 10% FCS. The cells were plated at a density of 2.5*10 4 cells/ml and cultivated in 100 ⁇ l medium at 37 0 C under 5% CO 2 . After 16 h the liposomes containing siRNA were diluted in the manufacturing buffer system and then 10 ⁇ l were added to the cells (110 ⁇ l final Volume and 9.1 % FCS per well) (0,4 to 100 nM PIkI or scrambled siRNA). 10 ⁇ l dilution buffer were also added to untreated cells and into wells without cells.
  • Cell proliferation/viability was determined by using the CellTiter-Blue Cell Viability assay (Promega, US). In brief, 72 hours after transfection, 100 ⁇ l Medium/CellTiter- Blue reagent (Pre-mix of 80ml Medium and 20 ⁇ l CellTiter-Blue reagent) were added to the wells. Following an incubation at 37 0 C for 2.5 hours, 80 ⁇ l of the medium were transferred into the wells of a black microtiter plate (NUNC, Denmark). Fluorescence was recorded using a fluorescence plate reader (Ex. 550 nm/Em. 590 nm).
  • Figs. 1-6 show the results of the transfection experiments. Compared to the lipids MoChol or CHIM the transfection efficiency can be improved by using the inventive novel sterol lipids (see IC 50 values shown in Figs. 1-4).
  • Figs. 5 and 6 show the dose response curves from specific liposomal formulations of the four tested cationic sterols comprising Plk-1 siRNA or scrambled siRNA.

Abstract

A facultative cationic sterol derivate, said sterol derivate has a pka value of between 3.5 and 8 and the general formula Cation-Spacer 2-Y-Spacer 1-X-Sterol wherein X and Y are linking groups and wherein at least on of X and Y is -NH(C=O)-O- and/or -O-(C=O)-NH. Also disclosed are liposomes comprising said sterol derivatives.

Description

Novel facultative cationic sterols
Field of the invention
The present invention relates to novel facultative cationic sterols and liposomes comprising such sterols. A particular aspect of the invention relates to amphoteric liposomes comprising the novel facultative cationic sterols.
Background of the invention
Sterol derivatives of the general formula 1 are known in the art and are disclosed in WO 02/066490 by Panzner et al., the content of which is incorporated herein by reference.
Cation - Spacer 2 - Y - Spacer 1 - X - Steryl (1),
wherein the cation is derived from piperazines, imidazoles, morpholines, purines, pyrimidines and /or pyridines;
the spacers 1 and 2 are selected from the group consisting of lower alkyl residues with up to 8 C atoms with a linear, branched or cyclic structure and 0, 1 or 2 ethylenically unsaturated bonds;
the linking groups X and/or Y are selected from the group consisting of -(C=O)-O-, -(C=O)-NH-, -(C=O)-S-, -O-, -NH-, -S-, -CH=N-, -0-(O=C)-, -S-(O=C)-, -NH-(O=C)-, and/or -N=CH-;
the steryl is selected from the group consisting of cholesteryl, sitosteryl, campesteryl, desmosteryl, fucosteryl, 22-ketosteryl, 20-hydroxysteryl, stigmasteryl, 22-hydroxycholesteryl, 25-hydroxycholesteryl, lanosteryl, 7-dehydrocholesteryl, dihydrocholesteryl, 19-hydroxycholesteryl, 5α-cholest-7-en-3β-yl,
7-hydroxycholesteryl, epicholesteryl, ergosteryl, and/or dehydroergosteryl; and the pka value of the compounds is between 3.5 and 8.
US 5,965,4343 of Wolff et al. discloses the compound CHIM. This molecule is a cationic sterol derivative with a carbamoyl linker group and an imidazole as base but only one spacer and no second linker group.
WO 97/39019 of Deshmukh et al. describes different cationic sterol compounds with a carbamoyl linker group and cyclic N containing bases, such as morpholine. However, these compounds have only one spacer and no second linker group.
Objects of the invention
The object of the present invention was to provide additional, alternative sterol derivative of the general formula 1 having improved stability upon storage.
Another object of the present is the provision of liposomes comprising such sterol derivatives.
Yet another object of the present invention is to provide liposomes comprising such sterol derivatives and encapsulating an active agent.
Yet another object of the invention is to provide liposomes comprising such sterol derivatives and allow effective transfection of cells.
Yet another object of the invention is to provide pharmaceutical compositions comprising such liposomes as carriers for the delivery of active agents or ingredients.
Summary of the invention
The object of the present invention is accomplished by the technical features of claims 1 , 7, 17, 18 and 19.
Provided are facultative cationic sterol of general formula 1 cation - spacer 2 - Y - spacer 1 - X - steryl, (1)
wherein the cation is a nitrogen base with a pKa value of between 3.5 and 8, the spacers 1 is a lower alkyl residue of linear, branched or cyclic structure, which has from 1 to 8 C atoms and includes O1 1 or 2 or 3 ethylenically unsaturated bonds, the spacer 2 is a lower alkyl residue of linear, branched or cyclic structure, which has from 0 to 8 C atoms and includes 0, 1 or 2 or 3 ethylenically unsaturated bonds, at least one of the linking group X or Y has the structure -NH-(C=O)-O- or -0-(C=O)- NH- and the other linking group X or Y a structure selected from -(C=O)-NH-; -O- (C=O)-NH-; -NH-(C=O)-O-; -(C=O)-S-; -O-; -NH-; -S-; -CH=N-.-O-(O=C)-; -(C=O)-O- ; -S-(O=C)-; -NH-(O=C)- Or -N=CH-, the overall molecule has a pka value of between 3.5 and 8.
Surprisingly the inventors have found that sterol derivatives comprising the above mentioned linking groups X and/or Y show an improved stability. Therefore liposomes, comprising the novel lipids can be stored in suspension for extended periods of time.
The steryls may be selected from the group consisting of cholesteryl, sitosteryl, campesteryl, desmosteryl, fucosteryl, 22-ketosteryl, 20-hydroxysteryl, stigmasteryl, 22-hydroxycholesteryl, 25-hydroxycholesteryl, lanosteryl, 7-dehydrocholesteryl, dihydrocholesteryl, 19-hydroxycholesteryl, 5α-cholest-7-en-3β-yl, 7-hydroxy- cholesteryl, epicholesteryl, ergosteryl, dehydroergosteryl and derivatives thereof.
The nitrogen base may be selected from unsubstituted or substituted piperazines, imidazoles, morpholines, purines, pyrimidines and /or pyridines or may derive from single or multiple substituted nitrogen atoms with lower hydroxyalkyls.
In one embodiment of the invention the inventive sterol derivatives have a pka value of between 5 and 8. In another embodiment of the invention the pka value of inventive sterol derivatives may be between 4 and 7.
Another aspect of the present invention relates to liposomes comprising the inventive cationic sterol derivatives.
The liposomes may further comprise one or more neutral and/or zwitterionic lipids and/or one or more anionic lipids and/or one or more cationic lipids.
In a particular embodiment of the present invention the liposomes comprising the inventive cationic sterols are amphoteric liposomes.
Said amphoteric liposomes may comprise an anionic lipid selected from the group consisiting of DOGS, DMGS, Chol-C1 , Chol-C2, Chol-C3, Chems, Chol-C5, Chol-C6, Chol-C7 and/or Chol-C8.
The liposomes according to the invention may further comprise neutral or zwitterionic lipids, preferably selected from POPC, DPPC, DSPC, DOPC, DMPC, POPE, DPPE, DSPE, DMPE, DOPE, DPhyPE, DLinPE or natural equivalents thereof or cholesterol.
The abbreviations for the lipids used herein can be found below in this disclosure.
In one embodiment said neutral and/or zwitterionic lipid is cholesterol or a mixture of phosphatidylethanolamine and cholesterol or a mixture of phosphatidylcholine and cholesterol.
In another embodiment said neutral and/or zwitterionic lipid is a mixture of phosphatidylethanolamine and phosphatidylcholine.
In one embodiment the size of the liposomes is of between 20 to 1000 nm.
In specific embodiments of the invention the liposomes comprise an active agent. In a preferred embodiment the active agent is a nucleic acid drug, such as oligonucleotides or plasmids.
The inventive liposomes may be used as a vector for in vivo, in vitro or ex vivo transfection.
Furthermore the liposomes may be useful for intravenous, subcutaneous, peritoneal, local or topical application.
Another aspect of the present invention relates to pharmaceutical compositions comprising the inventive liposomes for the treatment or prophylaxis of inflammatory, immune or autoimmune disorders, cancer and/or metabolic diseases of humans or non-human animals.
Detailed description of the invention
Lipids according to the invention
The present invention relates to novel facultative cationic sterol derivatives of the general formula 1 :
Cation - Spacer 2 - Y - Spacer 1 - X - Sterol (1 ).
wherein the cation is a nitrogen base with a pKa value of between 3.5 and 8, the spacers 1 is a lower alkyl residue of linear, branched or cyclic structure, which has from 1 to 8 C atoms and includes 0, 1 or 2 or 3 ethylenically unsaturated bonds, the spacer 2 is a lower alkyl residue of linear, branched or cyclic structure, which has from 0 to 8 C atoms and includes 0, 1 or 2 or 3 ethylenically unsaturated bonds, at least one of the linking group X or Y has the structure -NH-(C=O)-O- or -O-(C=O)- NH- and the other linking group X or Y a structure selected from -(C=O)-NH-; -O- (C=O)-NH-; -NH-(C=O)-O-; -(C=O)-S-; -O-; -NH-; -S-; -CH=N-.-O-(O=C)-; -(C=O)-O- ; -S-(O=C)-; -NH-(O=C)- Or -N=CH-, the steryls may be selected from the group consisting of cholesteryl, sitosteryl, campesteryl, desmosteryl, fucosteryl, 22-ketosteryl, 20-hydroxystery!, stigmasteryl, 22-hydroxycholesteryl, 25-hydroxycholesteryl, lanosteryl, 7-dehydrocholesteryl, dihydrocholesteryl, 19-hydroxycholesteryl, 5α-cholest-7-en-3β-yl, 7-hydroxy- cholesteryl, epicholesteryl, ergosteryl, dehydroergosteryl and derivatives thereof, the overall molecule has a pka value of between 3.5 and 8.
The overall molecule assumes its pH-dependent charge characteristics by one or more nitrogen bases with a pKa value between 3.5 and 8. These nitrogen bases are linked to the 3-position of the sterol skeleton via spacers and coupling groups, thus forming a compound according to the formula of the invention. In many cases, e.g. where the nitrogen bases are in the form of a ring system, positional isomers are existing, wherein the linking spacer is substituted to various positions of the organic cation. Such positional isomers fall within the disclosure of this invention. In many cases, the pKa values of the organic cation can be influenced via said positional isomerism alone. The relevant fundamental rules are well-known to those skilled in the art. Alternatively, these effects can be estimated from tabular compilations
(Handbook of Chemistry and Physics, Vol. 73, pp. 8-37ff.).
Preferred pka values of the inventive facultative cationic sterol derivatives may depend on the appropriate application of the lipid.
Therefore, in one embodiment of the invention the pka value of the sterol derivative may be between 5 and 8.
In another embodiment of the invention, the sterol derivative may have a pKa value of between 4 and 7. Advantageously, these pKa values fall in a range which is of crucial importance for the physiology of numerous organisms.
In a preferred embodiment of the invention, the cations are nitrogen bases.
In one embodiment of the invention the cations may be derived from piperazines, imidazoles, morpholines, purines, pyrimidines, pyridines, piperidines, anilines, anisidines, toluidines, phenetidines, benzimidazoles, isochinolines, picolines, pterines, purines, pyrazines, pyridazines, chinolines, thiazoles or triazines, pyrazols, imidazolines, imidazolidins, pyrazolines, pyrazolidines or hydrazins or derivatives thereof.
The cations preferably can be derived from piperazines, imidazoles, morpholines, purines, pyrimidines and/or pyridines or derivatives thereof.
Coupling reactions result in amphiphilic organic cations, e.g. those derived from the following classes of substances: o-, m-, p-anilines; 2-, 3- or 4-substituted anisidines, toluidines or phenetidines; 2-, 3-, 5-, 6-, 7- or 8-substituted benzimidazoles, 2-, 3-, 4- or 5-substituted imidazoles, 1- or 5-substituted isoquinolines, 2-, 3- or 4-substituted morpholines, 2-, 3- or 4-substituted picolines, 2-, 3- or 4-substituted piperidines, 1-, 2- or 3-substituted piperazines, 2-, 5- or 6-modified pterines, 3-, A-, 5-, 6- or 9-substituted purines, 2- or 3-substituted pyrazines, 3- or 4-substituted pyridazines, 2-, 3- or 4-modified pyridines, 2-, 4-, 5- or 6-substituted pyrimidines, 1-, 2-, 3-, 4-, 5-, 6- or 8-substituted quinolines, 2-, 4- or 5- substituted thiazoles, 2-, 4- or 6-substituted triazines, 1- ,3- ,4-or 5-substituted pyrazols, 2-, 3-, 4- or 5-substituted imidazolines, 1-, 2-, 3- or 4-substituted imidazolidines, 2-, 3- or 4-substituted pyrazolines, 1-, 2- or 3-substituted pyrazolidines or derivatives of tyrosine.
Particularly preferred are piperazines, imidazoles, morpholines, purines, pyrimidines and /or pyridines or derivatives thereof.
Most preferred as cations are imidazoles and/or morpholines or derivatives thereof.
In one embodiment of the invention molecule fragments such as occurring in biological systems are preferred, i.e., for example: 4-imidazoles (histamines), 2-, 6- or 9-purines (adenines, guanines, adenosines, or guanosines), 1-, 2- or 4-pyrimidines (uracils, thymines, cytosines, uridines, thymidines, cytidines), or pyridine-3-carboxylic acids (nicotinic esters or amides).
The above-mentioned structural fragments may also have additional substituents. For example, these can be methyl, ethyl, propyl, or isopropyl residues, more preferably in hydroxy lated form, including one or two hydroxy! groups. Also, these can be hydroxyl or keto functions in the ring system.
Nitrogen bases with preferred pKa values are also formed by single or multiple substitution of the nitrogen atom with lower alkanehydroxyls such as hydroxymethyl or hydroxyethyl groups. Suitable organic bases from this group are e.g. aminopropanediols, thethanolamines, tris(hydroxymethyl)methylamines, bis(hydroxymethyl)methylamines, tris(hydroxyethyl)methylamines, bis(hydroxyethyl)- methylamines, or the corresponding substituted ethylamines. Coupling of these fragments to the hydrophobic portion of the molecule may proceed either via the nitrogen of the base or via any of the hydroxyl functions.
Particularly preferred are β- or γ-hydroxy substituted amines as nitrogen bases.
In addition to sterol derivatives including a single organic cation, those including two or three identical or different groups are also preferred. All of these groups are required to have a pKa value in the above-mentioned range. One suitable complex group is the amide of histamine and histidine or of histamine and histidylhistidine.
In a preferred embodiment of the invention the linking group X has the structure -NH- (C=O)-O- or -0-(C=O)-NH- and the linking group Y a structure selected from -(C=O)- NH-; -0-(C=O)-NH-; -NH-(C=O)-O-; -(C=O)-S-; -O-; -NH-; -S-; -CH=N-.-O-(O=C)-; - (C=O)-O-; -S-(O=C)-; -NH-(O=C)- or -N=CH-.
In another preferred embodiment of the invention the linking group X has a structure selected from -(C=O)-NH-; -0-(C=O)-NH-; -NH-(C=O)-O-; -(C=O)-S-; -O-; -NH-; -S-; -CH=N-.-O-(O=C)-; -(C=O)-O-; -S-(O=C)-; -NH-(O=C)- or -N=CH- and the linking group Y a structure -0-(C=O)-NH- or -NH-(C=O)-O-.
In another preferred embodiment of the invention, spacer 1 is a lower alkyl residue of linear, branched or cyclic structure, which has from 1 to 8 C atoms and includes O, 1 or 2 or 3 ethylenically unsaturated bonds. Spacer 1 may have hydroxyl groups so as to increase the polarity of the molecule. In particular, spacer 1 can be a sugar or sugaralcohol. Spacer 2 is a lower alkyl residue of linear, branched or cyclic structure, which has from 0 to 8 C atoms and includes 0, 1 or 2 or 3 ethylenically unsaturated bonds. Spacer 2 may have hydroxy! groups so as to increase the polarity of the molecule. In particular, spacer 2 can be a sugar or sugaralcohol. In one embodiment spacer 2 is a lower alkyl residue of linear, branched or cyclic structure, which has from 1 to 8 C atoms and includes 0, 1 or 2 or 3 ethylenically unsaturated bonds.
Surprisingly it was found that the inventive sterol derivatives show an improved stability compared to the lipid MoChol as described in example 5.
Methods of performing such coupling reactions are well-known to those skilled in the art and may vary depending on the starting material and coupling component employed. Typical reactions are acylation, esterification, amidation, addition of amines to double bonds, etherification, or reductive amination.
In another preferred embodiment of the invention, the steryls are particularly cholesteryl, sitosteryl, campesteryl, desmosteryl, fucosteryl, 22-ketosteryl, 20- hydroxysteryl, stigmasteryl, 22-hydroxycholesteryl, 25-hydroxycholesteryl, lanosteryl, 7-dehydrocholesteryl, dihydrocholesteryl, 19-hydroxycholesteryl, 5α-cholest-7-en-3β- yl, 7-hydroxycholesteryl, epicholesteryl, ergosteryl, and/or dehydroergosteryl, as well as other related compounds.
The steryls that are used may bear various groups in the 3-position thereof, which groups allow for ready and stable coupling or optionally assume the function of a spacer. Particularly suitable for direct coupling are the hydroxyl group which is naturally present, but also, the chlorine of steryl chlorides, or e.g. the amino group of sterylamines, or the thiol group of thiocholesterol.
In one aspect facultative cationic sterols according to the present invention preferably have the following general formula 2:
Figure imgf000011_0001
wherein
Spacer 1 and Spacer 2 are as defined above and Y is selected from -(C=O)-NH-; -NH-(O=C)-; -0-(C=O)-NH-; -NH-(C=O)-O-; -O-;.-O-(O=C)- Or -(C=O)- O- and the cation is selected from substituted or unsubstituted piperazines, imidazoles, morpholines, purines, pyrimidines , pyridines and/or β- or γ-hydroxy substituted amines
In one embodiment of this aspect the inventive sterol may comprise, but not limited to, one of the following structures:
Table 1 :
Compound #1
CholC3N-Mo2
[^-Morpholine^-yl-ethylcarbamoyOmethylJ-carbamic acid cholesteryl ester
Figure imgf000011_0002
Compound #2
CholC4N-Mo2
[(2-Morpholine-4-yl-ethylcarbamoyl)-ethyl]-carbamic acid cholesteryl ester
Figure imgf000012_0001
Compound #3
CholC3N-Mo3
[(S-Morpholine^-yl-propylcarbamoyO-methyll-carbamic acid cholesteryl ester
Figure imgf000012_0002
Compound #4
CholDMC4N-Mo2
[1 -Methyl-2-(2-morpholine-4-yl-ethylcarbamoyl)-propyl]-carbamic acid cholesteryl ester
Figure imgf000012_0003
Compound #5
Figure imgf000013_0001
Compound #6
Figure imgf000013_0002
Compound #7
Figure imgf000014_0001
Compound #8
Figure imgf000014_0002
Compound #9
Figure imgf000014_0003
Compound #10
Figure imgf000014_0004
Figure imgf000015_0001
Liposomes according to the invention
The invention also relates to liposomes comprising the facultative cationic sterols according to the invention. The inventive facultative cationic sterols can be incorporated in high amounts in liposomal membranes.
In a special embodiment of the invention, the amount of the inventive sterol derivative in the liposomal membrane is between 3 and 100 mol%, preferred between 5 and 80 mol% and particularly preferred between 7 and 70 mol%.
In one embodiment of the invention the liposomes may comprise one or more of the inventive lipids and one or more neutral and/or zwitterionic lipid. Examples of such lipids include phosphatidyl choline, phosphatidyl ethanolamine, sphingomylein, ceramides, diacylglycerol and/or cholesterol.
In a preferred embodiment the phosphatidylcholine may be selected from the group comprising POPC, natural or hydrogenated soya PC, natural or hydrogenated egg PC, DMPC, DPPC, DSPC or DOPC. The phosphatidylethanolamines are preferably selected from the group comprising DOPE, DMPE or DPPE or POPE, DPhyPE, DLinPE, DSPE or natural equivalents thereof.
In another embodiment of the present invention liposomes comprising the inventive lipids may comprise one or more anionic lipids and/or one or more cationic lipids. Examples of anionic and cationic lipids are given below in this disclosure. In a particular aspect of the invention the liposomes comprising the inventive lipids are amphoteric liposomes. Amphoteric liposomes are a new class of liposomes described in WO 02/066012. Amphoteric liposomes have an anionic or neutral charge at pH 7.5 and a cationic charge at pH 4. These liposomes can be prepared from lipid mixtures comprising either an amphoteric lipid or a mixture of lipids with amphoteric character and optional a neutral and/or zwitterionic lipid.
The amphoteric liposomes comprising the inventive sterol lipids according to the present invention can be formed from any amphoteric lipid mixture.
In one embodiment the amphoteric lipid mixture may comprise a plurality of charged amphiphiles which in combination with one another have amphoteric character. A selection of charged amphiphiles is summarized below in this disclosure. In a preferred aspect of this embodiment said one or more charged amphiphiles comprise a pH sensitive anionic lipid and a pH sensitive cationic lipid. Herein, such a combination of a chargeable cation and chargeable anion is referred to as an "amphoteric II" lipid pair. In a preferred embodiment of this aspect said chargeable cations have pKa values of between about 5 and about 8. In a preferred embodiment of this aspect said chargeable cation is a facultative cationic sterol according to the present invention.
In another aspect of this embodiment said one or more charged amphiphiles comprise a stable anion and a chargeable cation and is referred to as "amphoteric III" lipid pair. In a preferred embodiment of this aspect said chargeable cations have pKa values of between about 4 and about 7. In a preferred embodiment of this aspect said chargeable cation is a facultative cationic sterol according to the present invention.
It is of course possible to use amphiphiles with multiple charges such as amphipathic dicarboxylic acids, phosphatidic acid, amphipathic piperazine derivatives and the like. Such multicharged amphiphiles might fall into pH sensitive amphiphiles or stable anions or cations or might have mixed character.
Suitable anionic lipids for the formation of amphoteric liposomes in combination with the inventive cationic sterols include, but are not limited to diacylglycerolhemisuccinates, e.g. DOGS, DMGS, POGS, DPGS, DSGS; diacylglycerolhemimalonat.es, e.g. DOGM or DMGM; diacylglycerolhemiglutarates, e.g. DOGG, DMGG; diacylglycerolhemiadipates, e.g. DOGA, DMGA; diacylglycerolhemicyclohexane-1 ,4-dicarboxylic acids, e.g. DO-cHA, DM-cHA; (2,3- Diacyl-propyl)amino}-oxoalkanoic acids e.g. DOAS1 DOAM, DOAG, DOAA, DMAS, DMAM1 DMAG, DMAA; Diacyl-alkanoic acids, e.g. DOP1 DOB, DOS, DOM1 DOG1 DOA, DMP1 DOB, DMS, DMM, DMG1 DMA; Chems and derivatives therof, e.g. Chol- C2, Chol-C3, Chol-C5 or Chol-C6, Chol-C7, Chol-C8; Chol-C1 , fatty acids, e.g. Oleic acid, Linoleic Acid, Myristic Acid, Palmitic acid, Stearic acid, Nervonic Acid, Behenic Acid; DOPA1 DMPA, DPPA, POPA, DSPA, Chol-SO4, DOPG, DMPG, DPPG, POPG, DSPG or DOPS1 DMPS1 DPPS, POPS, DSPS, Cetyl-phosphate or any of compounds 1-22 of table 3 below.
Any dialkyl derivatives of these anionic lipids comprising diacyl groups are also within the scope of the present invention.
Preferred anionic lipids for the formation of amphoteric liposomes in combination with the inventive cationic sterols may be selected from the group consisting of DOGS, DMGS, Chol-CI , Chol-C2, Chol-C3, Chems, Chol-C5, Chol-C6, Chol-C7 and/or Chol-C8.
In some embodiments of the invention the amphoteric lipid mixtures further comprise one or more neutral and/or zwitterionic lipids. Examples of such lipids may include phosphatidyl choline, phosphatidyl ethanolamine, sphingomylein, ceramides, diacylglycerol and/or cholesterol.
In one aspect of this embodiment said neutral lipids may be cholesterol or a mixture of cholesterol and phosphatidylethanolamines. Preferably said lipids are present in the amphoteric liposomes as sole neutral lipids.
Preferably the molar ratio of the mixtures of cholesterol and phosphatidylethanolamine is between 4 and 0.25, preferred between 3 and 0.5 and most preferred between 2 and 1.
Examples of phosphatidylethanolamines may include POPE, DPPE, DSPE, DMPE, DOPE, DPhyPE, DLJnPE or natural equivalents thereof. Mixtures of different phosphatidylethanolamines are also within the scope of the present invention. In a preferred embodiment of the invention the phosphatidylethanolamine is DOPE.
In another aspect of this embodiment a mixture of neutral lipids, such as phosphatidylcholines (PC), sphingomyelins or ceramides and cholesterol (Choi) may be used as sole neutral lipid components in the amphoteric liposomes.
Preferred are mixtures of phosphatidylcholines and cholesterol. The molar ratio of PC/Choi may be between 4 and 0.25 or between 3 and 0.33. Preferred are molar ratios of PC/Choi between 1.5 and 0,25, more preferred between 1 and 0.25.
The phosphatidylcholines may be selected without limitation from the group POPC, DOPC, DMPC, DPPC, DSPC or natural equivalents thereof, such as soy bean PC or egg-PC. Mixtures of different phosphatidylcholines are also within the scope of the present invention. In a preferred embodiment of the invention the phosphatidylcholine is selected from POPC or DOPC.
In a still further aspect of this embodiment a mixture of phosphatidylcholines, sphingomyelins or ceramides and phosphatidylethanolamines may be used as sole neutral lipid components in the amphoteric liposomes. Preferred are mixtures of phosphatidylcholines and phosphatidylethanolamines. In a preferred embodiment of the invention the phosphatidylcholine is POPC and the phosphatidylethanolamine is DOPE.
Obviously, further modifications of the liposomes according to the present invention are possible. Thus, the use of polyethylene glycol-modified phospholipids or analogous products is particularly advantageous. Other examples of lipids or compounds which sterically stabilize liposomes and thereby avoid an uptake of the particles by the RES (reticuloendothelial system) upon injection of the particles into the blood stream include for example polyglycerols, dextranes, polysialic acids, hydroxyethyl starches, hyaluronic acids, PEGylated lipids, sugar alcohols, Tween 80 or GM 1 gangliosides. Such lipids or detergents are known in the art to sterically stabilize liposomes (e.g. Woodle et al., Biochim. Biophys. Acta, 1113(2), 171-179, (1992); Allen et al., Biochim. Biophys. Acta, 981(1), 27-35, (1989)).
In one embodiment of the invention, the liposomes or amphoteric liposomes according to the present invention have an average size of between 50 and 1000 nm, preferably between 50 and 300 nm, and more preferably between 60 and 130 nm. In another embodiment the size of the liposomes or amphoteric liposomes may be less than 50nm, preferably of between 20 and 50 nm.
In one embodiment of the invention amphoteric liposomes comprising the inventive cationic sterol derivatives may be selected from one of the following formulations:
Table 2:
Figure imgf000019_0001
Figure imgf000020_0001
In another preferred embodiment, the liposomes comprise active substances. For example, the liposomes according to the invention are suitable for parenteral application. They can be used e.g. in cancer therapy and in the therapy of severe infections. To this end, liposome dispersions can be injected, infused or implanted. Thereafter, they are distributed in the blood or lymph or release their active substance in a controlled fashion as a depot. The latter can be achieved by highly concentrated dispersions in the form of gels. The liposomes can also be used for topical application on the skin. In particular, they may contribute to improved penetration of various active substances into the skin or even passage through the skin and into the body. Furthermore, the liposomes can also be used in gene transfer. Due to its size and charge, genetic material is usually incapable of entering cells without an aid. For this purpose, suitable carriers such as liposomes or lipid complexes are required which, together with the DNA, are to be taken up by the respective cells in an efficient and well-directed fashion. To this end, cell-inherent transport mechanisms such as endocytosis are used. Obviously, the liposomes of the invention can also be used as model membranes. In their principal structure, liposomes are highly similar to cell membranes. Therefore, they can be used as membrane models to quantify the permeation rate of active substances through membranes or the membrane binding of active substances.
Without being limited to such use, the liposomes in the present invention are well suited for use as carriers for active ingredients such as for example proteins, peptides, nucleic acids.
In one embodiment of the invention the active ingredient is a nucleic acid.
These drugs are classified into nucleic acids that encode one or more specific sequences for proteins, polypeptides or RNAs and into oligonucleotides that can specifically regulate protein expression levels or affect the protein structure through inter alia interference with splicing and artificial truncation. In some embodiments of the present invention, therefore, the nucleic acid-based therapeutic may comprise a nucleic acid that is capable of being transcribed in a vertebrate cell into one or more RNAs, which RNAs may be mRNAs, shRNAs, miRNAs or ribozymes, wherein such mRNAs code for one or more proteins or polypeptides. Such nucleic acid therapeutics may be circular DNA plasmids, linear DNA constructs, like MIDGE vectors (Minimalistic lmmunogenically Defined Gene Expression) as disclosed in WO 98/21322 or DE 19753182, or mRNAs ready for translation (e.g., EP 1392341).
In another embodiment of the invention, oligonucleotides may be used that can target existing intracellular nucleic acids or proteins. Said nucleic acids may code for a specific gene, such that said oligonucleotide is adapted to attenuate or modulate transcription, modify the processing of the transcript or otherwise interfere with the expression of the protein. The term "target nucleic acid" encompasses DNA encoding a specific gene, as well as all RNAs derived from such DNA, being pre-mRNA or mRNA. A specific hybridisation between the target nucleic acid and one or more oligonucleotides directed against such sequences may result in an inhibition or modulation of protein expression. To achieve such specific targeting, the oligonucleotide should suitably comprise a continuous stretch of nucleotides that is substantially complementary to the sequence of the target nucleic acid. Oligonucleotides fulfilling the abovementioned criteria may be built with a number of different chemistries and topologies. The oligonucleotides may comprise naturally occurring or modified nucleosides comprising but not limited to DNA, RNA, locked nucleic acids (LNA's), 2'O-methyl RNA (2'Ome), 2' O-methoxyethyl RNA (21MOE) in their phosphate or phosphothioate forms or Morpholinos or peptide nucleic acids (PNA1S). Oligonucleotides may be single stranded or double stranded.
Oligonucleotides are polyanionic structures having 8-60 charges. In most cases these structures are polymers comprising nucleotides. The present invention is not limited to a particular mechanism of action of the oligonucleotides and an understanding of the mechanism is not necessary to practice the present invention. The mechanisms of action of oligonucleotides may vary and might comprise inter alia effects on splicing, transcription, nuclear-cytoplasmic transport and translation. In a preferred embodiment of the invention single stranded oligonucleotides may be used, including, but not limited tθτ DNA-based oligonucleotides, locked nucleic acids, 2'-modified oligonucleotides and others, commonly known as antisense oligonucleotides. Backbone or base or sugar modifications may include, but are not limited to, Phosphothioate DNA (PTO), 2'O-methyl RNA (2'Ome), 2'Fluoro RNA (2'F), 2' O- methoxyethyl-RNA (2'MOE), peptide nucleic acids (PNA), N3"-P5' phosphoamidates (NP)1 2'fluoroarabino nucleic acids (FANA), locked nucleic acids (LNA), Morpholine phosphoamidate (Morpholino), Cyclohexene nucleic acid (CeNA), tricyclo-DNA (tcDNA) and others. Moreover, mixed chemistries are known in the art, being constructed from more than a single nucleotide species as copolymers, block- copolymers or gapmers or in other arrangements.
In addition to the aforementioned oligonucleotides, protein expression can also be inhibited using double stranded RNA molecules containing the complementary sequence motifs. Such RNA molecules are known as siRNA molecules in the art (e.g., WO 99/32619 or WO 02/055693). Other siRNAs comprise single stranded siRNAs or double stranded siRNAs having one non-continuous strand. Again, various chemistries were adapted to this class of oligonucleotides. Also, DNA / RNA hybrid systems are known in the art.
In another embodiment of the present invention, decoy oligonucleotides can be used. These double stranded DNA molecules and chemical modifications thereof do not target nucleic acids but transcription factors. This means that decoy oligonucleotides bind sequence-specific DNA-binding proteins and interfere with the transcription (e.g., Cho-Chung, et al. in Curr. Opin. MoI. Then, 1999).
In a further embodiment of the invention oligonucleotides that may influence transcription by hybridizing under physiological conditions to the promoter region of a gene may be used. Again various chemistries may adapt to this class of oligonucleotides. In a still further alternative of the invention, DNAzymes may be used. DNAzymes are single-stranded oligonucleotides and chemical modifications therof with enzymatic activity. Typical DNAzymes, known as the "10-23" model, are capable of cleaving single-stranded RNA at specific sites under physiological conditions. The 10-23 model of DNAzymes has a catalytic domain of 15 highly conserved deoxyribonucleotides, flanked by 2 substrate-recognition domains complementary to a target sequence on the RNA. Cleavage of the target mRNAs may result in their destruction and the DNAzymes recycle and cleave multiple substrates.
In yet another embodiment of the invention, ribozymes can be used. Ribozymes are single-stranded oligoribonucleotides and chemical modifications thereof with enzymatic activity. They can be operationally divided into two components, a conserved stem-loop structure forming the catalytic core and flanking sequences which are reverse complementary to sequences surrounding the target site in a given
RNA transcript. Flanking sequences may confer specificity and may generally constitute 14-16 nt in total, extending on both sides of the target site selected.
In a still further embodiment of the inventionτ aptamers may be used to target proteins. Aptamers are macromolecules composed of nucleic acids, such as RNA or DNA, and chemical modifications thereof that bind tightly to a specific molecular target and are typically 15-60 nt long. The chain of nucleotides may form intramolecular interactions that fold the molecule into a complex three-dimensional shape. The shape of the aptamer allows it to bind tightly against the surface of its target molecule including but not limited to acidic proteins, basic proteins, membrane proteins, transcription factors and enzymes. Binding of aptamer molecules may influence the function of a target molecule.
All of the above-mentioned oligonucleotides may vary in length between as little as 5 or 10, preferably 15 and even more preferably 18, and 50, preferably 30 and more preferably 25, nucleotides per strand. More specifically, the oligonucleotides may be antisense oligonucleotides of 8 to 50 nucleotides length that catalyze RNAseH mediated degradation of their target sequence or block translation or re-direct splicing or act as antogomirs; they may be siRNAs of 15 to 30 basepairs length; they may further represent decoy oligonucleotides of 15 to 30 basepairs length; can be complementary oligonucleotides influencing the transcription of genomic DNA of 15 to 30 nucleotides length; they might further represent DNAzymes of 25 to 50 nucleotides length or hbozymes of 25 to 50 nucleotides length or aptamers of 15 to 60 nucleotides length. Such subclasses of oligonucleotides are often functionally defined and can be identical or different or share some, but not all features of their chemical nature or architecture without substantially affecting the teachings of this invention. The fit between the oligonucleotide and the target sequence is preferably perfect with each base of the oligonucleotide forming a base pair with its complementary base on the target nucleic acid over a continuous stretch of the abovementioned number of oligonucleotides. The pair of sequences may contain one or more mismatches within the said continuous stretch of base pairs, although this is less preferred. In general the type and chemical composition of such nucleic acids is of little impact for the performance of the inventive liposomes as vehicles be it in vivo or in vitro and the skilled artisan may find other types of oligonucleotides or nucleic acids suitable for combination with the inventive liposomes.
Advantageously, liposomes produced using the sterol derivatives of the invention show low non-specific binding to cell surfaces. It is this low non-specific binding which is an essential precondition for achieving specific binding to target cells. Target control of the vehicles is obtained when providing the above-described liposomes with additional ligands. As a result, the active substance can be accumulated specifically in such cells or tissues which exhibit a pathological condition.
One important use of the facultative cationic sterols according to the invention is therefore in the construction of vectors for transfer of active substances in living organisms. The vectors are particularly suited for the transport of therapeutic macromolecules such as proteins or DNA which themselves are incapable of penetrating the cell membrane or undergo rapid degradation in the bloodstream.
Advantageously, antibodies, lectins, hormones or other active substances can be coupled to the surface of liposomes under mild conditions in high yields.
In a particularly preferred embodiment of the invention, at least 80% of the active substance is inside the liposome.
In another embodiments of the invention the liposomes comprise non-encapsulated active agents.
The preparation of liposomes is well known in the art (e.g. Liposomes, Ed. V.P.
Torchilin and V. Weissig, Second Edition, Oxford University Press, USA). Specific examples for the preparation of liposomes include, but are not limited to, the lipid film/hydration procedure with subsequent optional extrusion or sonication or the injection of an ethanolic lipid solution in water or buffer.
Amphoteric liposomes offer the distinct advantage that they can encapsulate nucleic acids with high efficiencies as disclosed for example in WO 02/066012 or WO 07/107304 both of Panzner et al..
The invention also relates to the use of the liposomes in the production of nanocapsules.
The invention also relates to the use of the liposomes in the production of release systems in diagnostics. Advantageously, the liposomes are used for the transport and/or release of active substances.
In another embodiment, the liposomes conveniently are used as depot formulation and/or as circulative depot.
Advantageously, the liposomes can be used in intravenous or peritoneal application.
In another embodiment of the invention, the liposomes are used with advantage as vector to transfect cells in vivo, in vitro and ex vivo.
A further aspect of the invention relates to pharmaceutical compositions comprising liposomes comprising the cationic sterol derivatives according to the present invention preferably as a carrier for the delivery of active agents or ingredients, including drugs such as nucleic acid drugs, e.g., oligonucleotides and plasmids. The pharmaceutical composition of the present invention may be formulated in a suitable pharmacologically acceptable vehicle. Vehicles such as water, saline, phosphate buffered saline, D5W and the like are well known to those skilled in the art for this purpose. In some embodiments the pharmaceutical compositions according to the invention may be used particularly for the treatment or prophylaxis of inflammatory, immune or autoimmune disorders, cancer and/or metabolic diseases of humans or non-human animals. A yet further aspect of the present invention relates to methods for the treatment of human or non-human animals in which said pharmaceutical composition comprising the inventive liposomes or amphoteric liposomes are targeted to a specific organ or organs, tumours or sites of infection or inflammation.
Definitions
By "chargeable" is meant that the amphiphile has a pK in the range pH 4 to pH 8. A chargeable amphiphile may therefore be a weak acid or base. A "stable" amphiphile is a strong acid or base, having a substantially stable charge on the range pH 4 to pH 8. By "amphoteric" herein is meant a substance, a mixture of substances or a supra- molecular complex (e.g., a liposome) comprising charged groups of both anionic and cationic character wherein:
1) at least one, and optionally both, of the cation and anionic amphiphiles is chargeable, having at least one charged group with a pK between 4 and 8,
2) the cationic charge prevails at pH 4, and
3) the anionic charge prevails at pH 8.
As a result the substance or mixture of substances has an isoelectric point of neutral net charge between pH 4 and pH 8. Amphoteric character is by this definition different from zwittehonic character, as zwitterions do not have a pK in the range mentioned above. In consequence, zwitterions are essentially neutrally charged over a range of pH values; phosphatidylcholines and phosphatidylethanolamines are neutral lipids with zwitterionic character.
By "C/A" or "C/A ratio" or "CVA molar ratio" herein is meant the molar ratio of cationic amphiphiles to anionic amphiphiles in a mixture of amphiphiles.
By "IC50" herein is meant the inhibitory concentration of an oligonucleotide leading to a 50 % knockdown of a target mRNA or in case of a proliferation assay to a 50 % inhibition of cell viability.
By "facultative cationic" herein is meant that the charge of a cation or a base depends on the pH of the medium.
The following list of lipids includes specific examples of neutral, zwitterionic, anionic, cationic or amphoteric lipids mentioned in this disclosure. The lipid list by no means limits the scope of this disclosure. The abbreviations for the lipids are used herein, the majority of which abbreviations are in standard use in the literature:
Neutral or zwitterionic lipids:
PC Phosphatidylcholine (unspecified membrane anchor)
PE Phosphatidylethanolamine (unspecified membrane anchor)
SM Sphingomyelin (unspecified membrane anchor)
DMPC Dimyristoylphosphatidylcholine
DPPC Dipalmitoylphosphatidylcholine DSPC Distearoylphosphatidylcholine
POPC 1 -Palmitoyl^-oleoylphosphatidylcholine
DOPC Dioleoylphosphatidylcholine
DOPE Dioleoylphosphatidylethanolamine DMPE Dimyristoylphosphatidylethanolamine
DPPE Dipalmitoylphosphatidylethanolamine
DPhyPE Diphytanoylphosphatidylethanolamine
DlinPE Dilinoleoylphosphatidylethanolamine
Choi Cholesterol
Any dialkyl derivatives of the neutral or zwitterionic lipids comprising diacyl groups listed above are also within the scope of the present invention.
Anionic lipids:
CHEMS Cholesterolhemisuccinate
Chol-COOH or Chol-C1 Cholesteryl-3-carboxylic acid
Chol-C2 Cholesterolhemioxalate
Chol-C3 Cholesterolhemimalonate Chol-C5 Cholesterolhemiglutarate
Chol-C6 Cholesterolhemiadipate
Chol-C7 Cholesterolhemipimelate
Chol-C8 Cholesterolhemisuberate
DGS or DG-Succ Diacylglycerolhemisuccinate (unspecified membrane anchor) DOGS or DOG-Succ Dioleoylglycerolhemisuccinate
DMGS or DMG-Succ Dimyristoylglycerolhemisuccinate
DPGS or DPG-Succ Dipalmitoylglycerolhemisuccinate
DSGS or DSG-Succ Distearoylglycerolhemisuccinate POGS or POG-Succ i-Palmitoyl-2-oleoylglycerolhemisuccinate
DOGM Dioleoylglycerolhemimalonate
DOGG Dioleoylglycerolhemiglutarate
DOGA Dioleoylglycerolhemiadipate
DMGM Dimyristoylglycerolhemimalonate DMGG Dimyristoylglycerolhemiglutarate
DMGA Dimyristoylglycerolhemiadipate
DOAS 4-{(2,3-Dioleoyl-propyl)amino}-4-oxobutanoic acid
DOAM 3-{(2,3-Dioleoyl-propyl)amino}-3-oxopropanoic acid DOAG 5-{(2,3-Dioleoyl-propyl)amino}-5-oxopentanoic acid
DOAA 6-{(2,3-Dioleoyl-propyl)amino}-6-oxohexanoic acid
DMAS 4-{(2,3-Dimyristoyl-propyl)amino}-4-oxobutanoic acid
DMAM 3-{(2,3-Dimyristoyl-propyl)amino}-3-oxopropanoic acid
DMAG 5-{(2,3-Dimyristoyl-propyl)amino}-5-oxopentanoic acid DMAA 6-{(2,3-Dimyristoyl-propyl)amino}-6-oxohexanoic acid
DOP 2,3-Dioleoyl-propanoic acid
DOB 3,4-Dioleoyl-butanoic acid
DOS 5,6-Dioleoyl-hexanoic acid
DOM 4,5-Dioleoyl-pentanoic acid DOG 6,7-Dioleoyl-heptanoic acid
DOA 7,8-Dioleoyl-octanoic acid
DMP 2,3-Dimyristoyl-propanoic acid
DMB 3,4-Dimyristoyl-butanoic acid
DMS 5,6-Dimyristoyl-hexanoic acid DMM 4,5-Dimyristoyl-pentanoic acid
DMG 6,7-Dimyristoyl-heptanoic acid
DMA 7,8-Dimyristoyl-octanoic acid
DOG-GIuA Dioleoylglycerol-glucoronic acid (1- or 4-linked)
DMG-GIuA Dimyristoylglycerol-glucoronic acid (1- or 4-linked) DO-cHA Dioleoylglycerolhemicyclohexane-1 ,4-dicarboxylic acid
DM-cHA Dimyristoylglycerolhemicyclohexane-1 ,4-dicarboxylic acid
DOPS Dioleoylphosphatidylserine
DPPS Dipalmitoylphosphatidylserine
DOPG Dioleoylphosphatidylglycerol DPPG Dipalmitoylphosphatidylglycerol
Chol-SO4 Cholesterol sulphate
DOPA Dioleoylphosphatidic acid
SDS Sodium dodecyl sulphate
Cet-P Cetylphosphate MA Myristic Acid
PA Palmitic Acid
OA Oleic Acid
LA Linoleic Acid
SA Stearic Acid
NA Nervonic Acid
BA Behenic Acid
Table 3:
Figure imgf000030_0001
Figure imgf000031_0001
The diacylglycerols may be dimyristoyl-, dipalmitoyl-, dioleoyl-, distearoyl- or palmitoyloleoylglycerols and R in the table above includes any selection from this group or cholesterol.
Any dialkyl derivatives of the anionic lipids comprising diacyl groups listed above are also within the scope of the present invention.
Cationic lipids:
MoChol 4-(2-Aminoethyl)-Morpholino-Cholesterolhemisuccinate
HisChol Histaminyl-Cholesterolhemisuccinate
CHIM Cholesterol-(3-imidazol-1-yl propyl)carbamate
DmC4Mo2 4-(2-Aminoethyl)-Morpholino-Cholesterol-2,3-dimethylhemisuccinate
DmC3Mo2 4-(2-Aminoethyl)-Morpholino-Cholesterol-2,2-dimethylhemimalonate
C3Mo2 4-(2-Aminoethyl)-Morpholino-Cholesterol-hemimalonate
C3Mo3 4-(2-Aminopropyl)-Moφholino-Cholesterol-hemimalonate
C4Mo4 4-(2-Aminobutyl)-Morpholino-Cholesterol-hemisuccinate
C5Mo2 4-(2-Aminoethyl)-Morpholino-Cholesterol- hemiglutarate
C6Mo2 4-(2-Aminoethyl)-Morpholino-Cholesterol- hemiadipate
C8Mo2 4-(2-Aminoethyl)-Morpholino-Cholesterol- hemiadipate
DDAB Dimethyldioctadecylammonium bromide
1 ,2-Diacyl-3-Trimethylammonium-Propane e.g.
DOTAP 1 ,2-Dioleoyl-3-Trimethylammonium-Propane
DMTAP 1 ,2-Dimyristoyl-3-Trimethylammonium-Propane DPTAP 1 ,2-Dipalmitoyl-3-Trimethylammonium-Propane
DSTAP 1 ,2-Distearoyl-3-Trimethylammonium-Propane
POTAP Palmitoyloleoyl-3-Trimethylammonium-Propane
1 ,2-Diacyl-3-Dimethylhydroxyethylammonium-Propane e.g.
DODMHEAP 1 ,2-Dioleoyl-3-dimethylhydroxyethylammonium-Propane
DMDMHEΞAP 1 ,2-Dimyristoyl-3-dimethylhydroxyethylammonium-Propane
DPDMHEAP 1 ,2-Dipalmitoyl-3-dimethylhydroxyethylammonium-Propane DSDMHEAP 1 ,2-Distearoyl-3-dimethylhydroxyethylammonium-Propane
PODMHEAP Palmitoyloleoyl-3-dimethylhydroxyethyl-ammonium-Propane
1 ,2-Diacyl-3-methyldihydroxyethylammonium-Propane e.g.
DOMDHEAP 1 ,2-Dioleoyl-3-methyldihydroxyethylammonium-Propane DMMDHEAP 1 ,2-Dimyristoyl-3-methyldihydroxyethylammonium-Propane
DPMDHEAP 1 ,2-Dipalmitoyl-3-methyldihydroxyethylammonium-Propane
DSMDHEAP 1 ,2-Distearoyl-3-methyldihydroxyethylammonium-Propane POMDHEAP Palmitoyloleoyl-3-methyldihydroxyethyl-ammonium-Propane
1 ,2-Diacyl-3-Dimethylammonium-Propane e.g.
DODAP 1 ,2-Dioleoyl-3-Dimethylammonium-Propane DMDAP 1 ,2-Dimyristoyl-3-Dimethylammonium-PiOpane
DPDAP 1 ,2-Dipalmitoyl-3-Dimethylammonium-Propane
DSDAP 1 ,2-Distearoyl-3-Dimethylammonium-Propane
PODAP Palmitoyloleoyl-3-Dimethylammonium-Propane
1 ,2-Diacyl-3-methylhydroxyethylammonium-Propane e g-
DOMHEAP 1 ,2-Dioleoyl-3-methylhydroxyethylammonium-Propane
DMMHEAP 1 ,2-Dimyristoyl-3-methylhydroxyethylammonium-Propane
DPMHEAP 1 ,2-Dipalmitoyl-3-methylhydroxyethylammonium-Propane DSMHEAP 1 ,2-Distearoyl-3-methylhydroxyethy!ammonium-Propane
POMHEAP Palmitoyloleoyl-3-methylhydrόxyethylammonium-Propane
1 ,2-Diacyl-3-dihydroxyethylammonium-Propane e.g.
DODHEAP 1 ,2-Dioleoyl-3-dihydroxyethylammonium-Propane
DMDHEAP 1 ,2-Dimyristoyl-3-dihydroxyethylammonium-Propane
DPDHEAP 1 ,2-Dipalmitoyl-3-dihydroxyethylammonium-Propane
DSDHEAP 1 ,2-Distearoyl-3-dihydroxyethylammonium-Propane PODHEAP Palmitoyloleoyl-3-dihydroxyethylammonium-Propane
i ^-Diacyl-sn-Glycero-S-Ethylphosphocholine e.g.
DOEPC 1 ^-Dioleoyl-sn-Glycero-S-Ethylphosphocholine DMEPC i ^-Dimyristoyl-sn-Glycero-S-Ethylphosphocholine
DPEPC 1 ^-Dipalmitoyl-sn-Glycero-S-Ethylphosphocholine
DSEPC 1 ,2-Distearoyl-sn-Glycero-3-Ethylphosphocholine
POEPC Palmitoyloleoyl-sn-Glycero-S-Ethylphosphocholine
DOTMA N-[1-(2,3-dioleyloxy)propyl]-N,N,N-trimethyl ammonium chloride
DOTIM 1 -[2-(oleoyloxy)ethyl]-2-oleyl-3-(2-hydroxyethyl) imidazolinium chloride
TMAG N-(a-trimethylammonioacetyl)-didodecyl-D-glutamate chloride
BCAT O-(2R-1 ,2-di-O-(1 ΘZ.ΘΘZ-octadecadienyO-glyceroO-N- (bis-2-aminoethyl)carbamate
DODAC Dioleyldimethylammonium chloride
DORIE 1 ,2-dioleoyl-3-dimethyl-hydroxyethyl ammonium bromide
DMRIE 1 ,2-dimyristoyl-3-dimethyl-hydroxyethyl ammonium bromide
DOSC 1 ^-dioleoyl-S-succinyl-sn-glycerol choline ester DORI 1 ,2-dioleoyloxypropyl-3-dimethylhydroxyethylammonium chloride
DHMHAC N.N-di-n-hexadecyl-N.N-dihydroxyethylammoniumbromide
DHDEAB N,N-di-n-hexadecyl-N-methyl,N-(2- hydroxyethyl)ammonium chloride DMHMAC N>N-myristyl-N-(1-hydroxyprop-2-yl)-N-methylammonium chloride
DOTB 1 ,2-dioleoyl-3-(4'-trimethylammonio)butanoyl-sn- glycerol DOSPA 2,3-Dioleyloxy-N-[2-(sperminecarboxamido)ethyl]-N,N-dimethyl-1- propanaminium trifluoroacetate
DOGS* Dioctadecylamido-glycylspermine DOGSDSO 1 ^-dioleoyl-sn-glycero-S-succinyl^-hydroxyethyl disulfide ornithine SAINT lipids Synthetic Amphiphiles INTerdisciplinary DPIM1 DOIM 4,(2,3-bis-acyloxy-propyl)-1-methyl-1 H- imidazole (unspecified membrane anchor)
MoDP 1 ,2-Dipalmitoyl-3-N-morpholine-propane
MoDO 1 ,2-Dioleoyl-3-N-morpholine-propane
DPAPy 2,3-bis-palmitoyl-propyl-pyhdin-4-yl-amine
DC-Choi 3b-[N-(N9,N9-dimethylaminoethane)carbamoyl] cholesterol TC-Chol 3b-[N-(N9,N9-trimethylaminoethane) carbamoyl] cholesterol
DAC-Chol 3b(N-(N,N'-Dimethylaminoethan)- carbamoyl)cholesterol
PipC2Chol 4{N-2-ethylamino[(3'-β-cholesteryl) carbamoyl]} piperazine
MoC2Chol {N-2-ethylamino[(3'-β-cholesteryl) carbamoyl]} morpholine
MoC3Chol {N-2-propylamino[(3'-β-cholesteryl) carbamoyl]} morpholine N-methyl-PipChol N-methyl{4-N-amino[(3'-β-cholesteryl) carbamoyl]}piperazine
PyrroC2Chol {N-2-ethylamino[(3'-β-cholesteryl) carbamoyl]}pyrrolidine
PipeC2Chol {N-2-ethylamino[(3'-β-cholesteryl) carbamoyl]}piperidine lmC3Chol {N-2-propylamino[(3'-β-cholesteryl) carbamoyl]}imidazole
PyC2Chol {N-2-ethylamino[(3'-β-cholesteryl) carbamoyl]}pyridine CTAB Cetyltrimethylammonium bromide
NeoPhectin™ cationic cardiolipins (e.g. [1 , 3-Bis-(1 , 2-bis-tetradecyloxy- propyl-3-dimethylethoxyammonium bromide)-propane-2-ol]
Table 4:
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
The diacylglycerols may be dimyristoyl-, dipalmitoyl-, dioleoyl-, distearoyl- or palmitoyloleoylglycerols and R in the table above includes any selection from this group or R may be a C6-C30 dialkyl, with 0-3 unsaturated bonds.
Any dialkyl derivatives of the cationic lipids comprising diacyl groups listed above are also within the scope of the present invention. Amphoteric lipids:
HistChol Nα-Histidinyl-Cholesterol-hemisuccinate
HistDG 1 ,2 — Dipalmitoylglycerol-hemisuccinat-N_-Histidinyl- hemisuccinate, & Distearoyl- .Dimyristoyl, Dioleoyl or palmitoyl-oleoylderivatives
IsoHistSuccDG 1 ^-Dipalmitoylglycerol-O-Histidinyl-Na-hemisuccinat, &
Distearoyl-, Dimyristoyl, Dioleoyl or palmitoyl- oleoylderivatives AC Acylcarnosine, Stearyl- & Palmitoylcarnosine
HCChol Nα-Histidinyl-Cholesterolcarbamate
Any dialkyl derivatives of the amphoteric lipids comprising diacyl groups listed above are also within the scope of the present invention.
Brief description of the drawings
Figure 1 shows IC 50 values of amphoteric liposomes encapsulating siRNA targeting Plk-1 derived from the in vitro transfection of HeIa cells as described in example 6. Shown are mixtures of MoChol/DMGS, CHIM/DMGS, CholC4N-Mo2/DMGS and CholC3N-Mo2/DMGS with a C/A ratio of 0.33 and increasing amounts of Choi as neutral lipid (0-60 mol%). The bars marked with an asterisk indicate that the IC 50 values are higher than the tested dose range (75 or 100 nM siRNA, respectively).
Figure 2 shows IC 50 values of amphoteric liposomes encapsulating siRNA targeting Plk-1 derived from the in vitro transfection of HeIa cells as described in example 6. Shown are mixtures of MoChol/DMGS, CHIM/DMGS, CholC4N-Mo2/DMGS and CholC3N-Mo2/DMGS with a C/A ratio of 0.5 and increasing amounts of Choi as neutral lipid(0-60 mol%). The bars marked with an asterisk indicate that the IC 50 values are higher than the tested dose range (75 or 100 nM siRNA, respectively).
Figure 3 shows IC 50 values of amphoteric liposomes encapsulating siRNA targeting Plk-1 derived from the in vitro transfection of HeIa cells as described in example 6. Shown are mixtures of MoChol/DMGS, CHIM/DMGS, CholC4N-Mo2/DMGS and CholC3N-Mo2/DMGS with a C/A ratio of 0.33 and increasing amounts of POPC/Chol=0.5 (molar ratio) as neutral lipid(0-60 mol%). The bars marked with an asterisk indicate that the IC 50 values are higher than the tested dose range (75 nM siRNA).
Figure 4 shows IC 50 values of amphoteric liposomes encapsulating siRNA targeting Plk-1 derived from the in vitro transfection of HeIa cells as described in example 6. Shown are mixtures of MoChol/DMGS, CHIM/DMGS, CholC4N-Mo2/DMGS and CholC3N-Mo2/DMGS with a C/A ratio of 0.5 and increasing amounts of POPC/Chol=0.5 (molar ratio) as neutral lipid (0-60 mol%). The bars marked with an asterisk indicate that the IC 50 values are higher than the tested dose range (75 or 100 nM siRNA, respectively).
Figure 5 shows the % cell viability (normalized to mock treated cells) of HeIa cells transfected with different amphoteric liposomes encapsulating siRNA targeting Plk-1 (black bars) or non-targeting scrambled siRNA (grey bars). Shown are following amphoteric liposome formulations: MoChol/DMGS/Chol 17.5:52.5:30 (molar ratio), CHIM/DMGS/Chol 17.5:52.5:30 (molar ratio),CholC4N-Mo2/DMGS/Chol 17.5:52.5:30 (molar ratio) and CholC3N-Mo2/DMGS/Chol 17.5:52.5:30 (molar ratio).
Figure 6 shows the % cell viability (normalized to mock treated cells) of HeIa cells transfected with different amphoteric liposomes encapsulating siRNA targeting Plk-1 (black bars) or non-targeting scrambled siRNA (grey bars). Shown are following amphoteric liposome formulations: MoChol/DMGS/POPC/Chol 26.7:53.3:6.7:13.3 (molar ratio), CHIM/DMGS/POPC/Chol 26.7:53.3:6.7:13.3 (molar ratio), CholC4N- Mo2/DMGS/POPC/Chol 26.7:53.3:6.7:13.3 (molar ratio), CholC3N- Mo2/DMGS/POPC/Chol 26.7:53.3:6.7:13.3 (molar ratio).
Examples are given with the understanding of further detailing certain aspects of practising the current invention. The examples by no means limit the scope of this disclosure. Example 1 :
Synthesis of [^-Morpholine^-yl-ethylcarbamoyOmethylJ-carbamic acid cholesteryl ester (D
Reaction scheme:
Figure imgf000040_0001
Step 1 : Synthesis of [(2-Morpholine-4-yl-ethylcarbamoyl)methyl]-carbamic acid tert- butyl ester
6 g N-tert.-butyloxycarbonyl-glycine and 12.65 g TBTU (2-(1 H-benzotriazole-1-yl)- 1 ,1 ,3,3-tetramethyluronium tetrafluoroborate) were suspended in 200 ml THF (tetrahydrofurane) and stirred. Then 4.46 g 4-(2-aminoethyl)morpholine und 10.39 g N-methylmorpholine were added. The reaction mixture was allowed to stir at room temperature overnight. The suspension was filtered and the solvent of the filtrate was removed by rotary evaporation. The crude product was purified by column chromatography at silica gel (eluent: ethyl acetate : methanol 1 :1 + 1% NH4OH). The product, a colourless oil, was characterized by 1H-NMR.
Step 2: Synthesis of 2-Amino-N-(2-morpholine-4-yl-ethyl)-acetamide
Under vacuum 9.8 g [(2-Morpholine-4-yl-ethylcarbamoyl)methyl]-carbamic acid tert- butyl ester were dissolved in 280 ml dry dichloromethane into a round bottom flask.
The mixture was cooled with an ice bath and stirred for 50 min. Subsequently, 52.56 ml trifluoroacetic acid were added drop wise to the mixture. The ice bath was removed and the reaction was allowed to stir for 2 h. The solvent was removed by rotary evaporation and without further purification the product was used for step 3 of the synthesis. Step 3: Synthesis of [(2-Morpholine-4-y!-ethy!carbamoy!)methy!]-carbarriic acid cholesteryl ester (1)
2.56 g 2-Amino-N-(2-morpholine-4-yl-ethyl)-acetamid and 100 ml chloroform were added under vakkum into a round bottom flask and the mixture was cooled with an ice bath. Then 8.65 g triethylamine were added drop wise to the mixture. Subsequently, 4.8 g cholesteryl chloroformiate, dissolved in 25 ml chloroform, were added drop wise. The reaction mixture was stirred for 1.5 days. Then the solvent was removed by rotary evaporation and the crude product purified by column chromatography at silica gel (eluents: CHCI3 + 1 ,5 % MeOH; CHCI3 + 2 % MeOH). The product was characterized by 1H-NMR and LC-MS.
Example 2:
Synthesis of β-^-Morpholine^-yl-ethylcarbamoylJ-ethylJ-carbamic acid cholesteryl ester (2)
Reaction scheme:
Figure imgf000041_0001
Step 1 : Synthesis of [2-(2-Morpholine-4-yl-ethylcarbamoyl)-ethyl]-carbamic acid tert- butyl ester
6 g N-tert.-Butyloxycarbonyl-beta-alanine and 11.71 g TBTU were suspended in 200 ml THF and stirred. Then 4.13 g 4-(2-aminoethyl)morpholine and 9.62 g N- methylmorpholine were added and the reation mixture was allowed to stir at room temperature over night. The suspension was filtered and the solvent of the filtrate was removed by rotary evaporation. The crude product was purified by column chromatography at silica gel (eluent: ethyl acetate : methanol 1 :1 + 1% NH4OH). The product, a colourless oil, was characterized by 1H-NMR.
Step 2: Synthesis of 3-Amino-N-(2-morpholine-4-yl-ethyl)-propionamid ditrifluoroacetate
6.6 g [2-(2-Morpholine-4-yl-ethylcarbamoyl)-ethyl]-carbamic acid tert.-butyl ester were added into a round bottom flask and under vacuum dissolved in 200 ml dry dichloromethane. The mixture was cooled and 33.74 ml thfluoroacetic acid were added drop wise to the mixture. The ice bath was removed and the reaction was allowed to stir for 5 h at room temperature. Then the solvent was removed by rotary evaporation and without further purification the product, a yellow oil, was used for step 3 of the synthesis.
Step 3: Synthesis of [2-(2-Morpholine-4-yl-ethylcarbamoyl)-ethyl]-carbamic acid cholesteryl ester (2)
5.8 g 3-amino-N-(2-morpholine-4-yl-ethyl)-propionamid ditrifluoroacetate were dissolved in 150 ml chloroform. Then 11.09 g triethylamine and 3.94 g cholesteryl chloroformiate were added. The reaction mixture was stirred at room temperature overnight. Then the solvent was removed by rotary evaporation and the crude product, a colourless oil, dissolved in 200 ml ethyl acetate and washed twice with water. The organic phase was dried over Na2SO4 overnight and after filtration the solvent was removed by rotary evaporation. The product was dried by lyophilization and characterized by 1H-NMR and LC-MS.
Example 3:
Synthesis of [(S-Morpholine^-yl-propylcarbamoyO-methylJ-carbamic acid cholesteryl ester (3) Reaction scheme:
Figure imgf000043_0001
3
Step 1 : Synthesis of [(S-Morpholine^-yl-propylcarbamoyO-methyll-carbamic acid tert.-butyl ester
6 g N-tert.-butyloxycarbonyl-glycine and 12.65 g TBTU were suspended in 200 ml THF and stirred. Then 4.94 g N-(3-aminopropyl)morpholine and 10.39 g N- methylmorpholine were added and the reaction mixture was allowed to stir at room temperature over night. The suspension was filtered and the solvent of the filtrate was removed by rotary evaporation. The crude product was purified by column chromatography at silica gel (eluent: ethyl acetate : methanol 1 :1 + 1% NH4OH). The product, a colourless oil, was characterized by 1H-NMR.
Step 2: Synthesis of 2-Amino-N-(3-morpholine-4-yl-propyl)-acetamid bistrifluoroacetate
9.82 g [(S-Morpholine^-yl-propylcarbamoylJ-methylJ-carbamic acid tert.-butyl ester were added into a round bottom flask and under vacuum dissolved in 250 ml dry dichloromethane. The mixture was cooled with an ice bath and 50.2 ml trifluoroacetic acid were added. The ice bath was removed and the reaction was allowed to stir for 3.5 h at room temperature. Then the solvent was removed by rotary evaporation and without further purification the product, a yellow oil, was used for step 3 of the synthesis.
Step 3: Synthesis of [(3-Mθφholine-4-yl-propylcarbamoyl)-methyl]-carbamic acid cholesteryl ester (3) 5.8 g 2-amino-N-(3-morpholine-4-yl-propyl)-acetamid bistrifluoroacetate were dissolved in 150 ml chloroform. Then 13.3 g triethylamine and 3.94 g cholesteryl chloroformiate were added. The reaction mixture was stirred at room temperature overnight. Then the solvent was removed by rotary evaporation and the crude product, a colourless oil, dissolved in 200 ml ethyl acetate and washed twice with water. The organic phase was dried over Na2SO4 and after filtration the solvent was removed by rotary evaporation. The product was dried by lyophilization and characterized by 1H-NMR and LC-MS.
Example 4:
Synthesis of [i-Methyl-Z-^-morpholine^-yl-ethylcarbamoyO-propylj-carbamic acid cholesteryl ester (4)
Reaction scheme:
Figure imgf000044_0001
Step 1 : Synthesis of 3-Amino-2-methyl-butyric acid
In a round bottom flask with a reflux condenser to 3.55 g sodium (in small pieces) 125 ml dry ethanol was added. When all sodium was dissolved and the solution cooled down to room temperature 10.73 g hydroxylamine*HCI in 8 ml H2O were heated to 60 0C and then added to the reaction mixture. The reaction was cooled with an ice bath and then centrifuged. The supernatant was added to 7.73 g (E)-2- methyl-but-2-enoic acid and the yellow solution was refluxed at 100 0C overnight and then cooled down to room temperature. After the addition of a couple of seed crystals the mixture was incubated at -20 0C for 7 h. The formed crystals were filtered, washed with 10 ml cold ethanol and then dried. The product was characterized by thin layer chromatography.
Step 2: Synthesis of S-tert.-Butoxycarbonylamino^-methyl-butyric acid
2.65 g 3-amino-2-methyl-butyric acid were added into a round bottom flask and cooled with an ice bath. Then 2.64 g sodium carbonate (anhydrous) dissolved in 25 ml H2O were added slowly to the 3-amino-2-methyl-butyric acid. In addition, 30.9 g 1.4-dioxane and 4.94 g di-tert-butyl-dicarbonate were added to the mixture. The ice bath was removed and the mixture was allowed to stir at room temperature for 2 days. Then 1 ,4-dioxane was removed by rotary evaporation and subsequently the mixture was diluted with 20 ml H2O. After an extraction with diethyl ether the aqueous phase was overlaid with ethyl acetate and acidified to pH 3 with a KHSO4 solution. The formed white precipitate was removed by filtration and after a separation of the phases the aqueous phase was extracted twice with ethyl acetate. Then the organic phases were combined and the solvent removed by rotary evaporation. The white residue was then combined with the precipitate above and dried. This product was characterized by 1H-NMR.
Step 3: Synthesis of [1-Methyl-2-(2-Morpholine-4-yl-ethylcarbamoyl)-propyl]-carbamic acid tert. butyl ester
Under vacuum 3.3.g 3-tert.-Butoxycarbonylamino-2-methyl-butyhc acid and 5.85 g TBTU were suspended in 100 ml THF and stirred. Then 2.18 g 4-(2- aminoethyl)morpholine und 4.61 g N-methylmorpholine were added and the reaction mixture was allowed to stir at room temperature over night. The suspension was filtered and the solvent of the filtrate was removed by rotary evaporation. The residue was purified twice by column chromatography at silica gel (eluents: ethyl acetate : methanol 1 :1 ; ethyl acetate : methanol 2.5:1). The crude product was dissolved in ethyl acetate and this organic phase was washed with a saturated Na2CO3-solution and with brine. After drying the organic phase over Na2SO4 the solvent was removed by rotary evaporation and a white product remained. Step 4: Synthesis of 3-Amino-2-methy!-N-(2-morpho!ine-4-y!-ethyl)-butyramide bistrifluoroacetate
Under vacuum 6.2 g [1-Methyl-2-(2-Morpholine-4-yl-ethylcarbamoyl)-propyl]- carbamic acid tert. butyl ester and 200 ml dry dichloromethane were added into a round bottom flask. Then the mixture was cooled with an ice bath and 29 ml trifluoroacetic acid were added drop wise. The ice bath was removed and the reaction was allowed to stir for 2 h at room temperature. Then the solvent was removed by rotary evaporation and the product, a yellow oil, characterized by 1H- NMR.
Step 5: Synthesis of [i-Methyl^^-Morpholine^-yl-ethylcarbamoylJ-propy^-carbamic acid cholesteryl ester (4)
Under vacuum 6.95 g 3-amino-2-methyl-N-(2-morpholine-4-yl-ethyl)-butyramide bistrifluoroacetate were dissolved in 90 ml chloroform and cooled with an ice bath. Then 15.3 g triethylamin und von 5.69 g cholesteryl chloroformiate, dissolved in 20 ml chloroform, were added slowly to the mixture. The ice bath was removed and the reaction mixture was allowed to stir at room temperature for 2.5 days. Then the solvent was removed by rotary evaporation and the crude product purified twice by column chromatography at silica gel (eluents: CHCI3 + 1 % methanol; CHCI3 + 2 % methanol). The product was characterized by 1H-NMR and LC-MS.
Example 5:
Stability of the inventive lipids
Preparation of lipid suspensions:
The lipids (see table 6) were dissolved in isopropanol to a final lipid concentration of 30 mM. The lipid solutions were then mixed with PBS resulting in a final lipid concentration of 1 mM and a final isopropanol amount of 3 %. Stability test:
Stability of the lipids was tested under stress conditions, including acidic, alkaline and oxidative conditions at room temperature. In addition, one aliquot of the lipid suspensions was incubated at 60 0C for three days. The conditions of the stability test are summarized in table 5 below.
Table 5:
Figure imgf000047_0001
HPTLC: After the incubation of the samples under the above mentioned conditions samples were analyzed by HPTLC (High Performance Thin Layer Chromatography).
Results:
The results of the stability test are summarized in table 6 as % degradation of the appropriate lipid.
Table 6:
Figure imgf000047_0002
Compared to Mochol the inventive lipids show an improved stability, especially under alkaline conditions and under heat at physiological pH. Example 6:
In vitro transfection of HeIa cells with amphoteric liposomes encapsulating siRNA targeting Plk-1 or non-targeting scrambled (scr) siRNA
Amphoteric liposomes comprising lipids according to the present invention were used for tranfection of HeIa cells.
Preparation of liposomes:
Liposomes were manufactured by an isopropanol-injection method. Lipids were dissolved in isopropanol (30 mM lipid concentration) and mixed. Liposomes were produced by adding siRNA solution in NaAc 2OmM, Sucrose 300 mM, pH 4.0 (pH adjusted with HAc) to the alcoholic lipid mix, resulting in a final alcohol concentration of 30% and a N/P ratio 5. The formed liposomal suspensions were shifted to pH 7.5 with twice the volume of Na2HPO4 136mM, NaCI 10OmM (pH 9), resulting in a final lipid concentration of 3 mM and a final isopropanol concentration of 10%.
N/P = the ratio cationic charges from the lipids to anionic charges from the siRNA during manufacturing.
Plk-1 siRNA as in Haupenthal et al., lnt J Cancer, 121 , 206-210 (2007).
Following liposomal formulations were prepared with Plk-1 siRNA or scrambled siRNA and tested for transfection efficiency on HeIa cells:
Table 7:
Figure imgf000048_0001
Figure imgf000049_0001
Transfection protocol:
HeLa cells were obtained from DSMZ (German Collection of Micro Organism and Cell Cultures) and maintained in DMEM. Media were purchased from Gibco- Invitrogen and supplemented with 10% FCS. The cells were plated at a density of 2.5*104 cells/ml and cultivated in 100 μl medium at 37 0C under 5% CO2. After 16 h the liposomes containing siRNA were diluted in the manufacturing buffer system and then 10 μl were added to the cells (110μl final Volume and 9.1 % FCS per well) (0,4 to 100 nM PIkI or scrambled siRNA). 10μl dilution buffer were also added to untreated cells and into wells without cells. In addition, as control, free siRNA was added to the cells (10 to 80 nM Plk-1 or scrambled siRNA). Cell culture dishes were incubated for 72 h hours at 37 0C under 5% CO2. Transfection efficiency was analyzed using a cell proliferation/viability assay.
Cell proliferation/viability assay:
Cell proliferation/viability was determined by using the CellTiter-Blue Cell Viability assay (Promega, US). In brief, 72 hours after transfection, 100 μl Medium/CellTiter- Blue reagent (Pre-mix of 80ml Medium and 20μl CellTiter-Blue reagent) were added to the wells. Following an incubation at 37 0C for 2.5 hours, 80 μl of the medium were transferred into the wells of a black microtiter plate (NUNC, Denmark). Fluorescence was recorded using a fluorescence plate reader (Ex. 550 nm/Em. 590 nm). On each plate the following controls were included: i) wells without cells but with medium (control for culture medium background fluorescence) and ii) wells with cells (untreated cells = mock-transfected cells). For calculation, the mean fluorescence value of the culture medium background was subtracted from all mean (triplicates) values of experimental wells (transfected and mock-transfected cells). The fluorescence values from each transfection were normalized to the mean fluorescence value from mock-transfected cells, which was set as being 100%.
Results:
Figs. 1-6 show the results of the transfection experiments. Compared to the lipids MoChol or CHIM the transfection efficiency can be improved by using the inventive novel sterol lipids (see IC 50 values shown in Figs. 1-4). Figs. 5 and 6 show the dose response curves from specific liposomal formulations of the four tested cationic sterols comprising Plk-1 siRNA or scrambled siRNA.

Claims

Claims:
1. A sterol derivative according to the general formula (1)
cation - spacer 2 - Y - spacer 1 - X - steryl, (1)
wherein
the cation is a nitrogen base with a pKa value of between 3.5 and 8,
the spacers 1 is a lower alkyl residue of linear, branched or cyclic structure, which has from 1 to 8 C atoms and includes 0, 1 or 2 or 3 ethylenically unsaturated bonds,
the spacer 2 is a lower alkyl residue of linear, branched or cyclic structure, which has from 0 to 8 C atoms and includes 0, 1 or 2 or 3 ethylenically unsaturated bonds,
at least one of the linking group X or Y has the structure -NH-(C=O)-O- or -O- (C=O)-NH- and the other linking group X or Y a structure selected from -
(C=O)-NH-; -0-(C=O)-NH-; -NH-(C=O)-O-; -(C=O)-S-; -O-; -NH-; -S-; -CH=N- .-0-(O=C)-; -(C=O)-O-; -S-(O=C)-; -NH-(O=C)- Or -N=CH-,
the overall molecule has a pka value of between 3.5 and 8.
2. The sterol derivative as claimed in claim 1 wherein said steryl is selected from the group consisting of cholesteryl, sitosteryl, campesteryl, desmosteryl, fuco- steryl, 22-ketosteryl, 20-hydroxysteryl, stigmasteryl, 22-hydroxycholesteryl, 25- hydroxycholesteryl, lanosteryl, 7-dehydrocholesteryl, dihydrocholesteryl, 19-hydroxycholesteryl, 5α-cholest-7-en-3β-yl, 7-hydroxycholesteryl, epicholesteryl, ergosteryl and dehydroergosteryl and derivatives thereof.
3. The sterol derivative as claimed in claim 1 or 2 wherein said nitrogen base is selected from unsubstituted or substituted piperazines, imidazoles, morpholines, purines, pyrimidines and pyridines.
4. The sterol derivative as claimed in any of claim 1 to 3 wherein said nitrogen base is derived from single or multiple substituted nitrogen atoms with lower hydroxyalkyls.
The sterol derivative as claimed in any of claims 1 to 4 wherein said sterol derivate has a pka value of between 5 and 8.
The sterol derivative as claimed in claims 1 ,3-5 wherein said sterol derivative has the following general formula 2
Figure imgf000052_0001
wherein Y is selected from
-(C=O)-NH-; -NH-(O=C)-; -0-(C=O)-NH-; -NH-(C=O)-O-; -O-;.-O-(O=C)- or -
(C=O)-O- and the cation is selected from piperazines, imidazoles, morpholines, purines, pyrimidines , pyridines and β- or γ-hydroxy substituted amines.
Liposomes comprising sterol derivatives as claimed in any of claims 1 to 6.
8. The liposomes as claimed in claim 7, wherein said liposomes comprise one or more neutral or zwitterionic lipid and/or one or more anionic lipid and/or one or more cationic lipid.
9. The liposomes as claimed in any of claim 7 to 8, wherein said liposomes are amphoteric liposomes.
10. The liposomes as claimed in claim 9 wherein said amphoteric liposomes comprise one or more anionic lipids selected from the group consisting of diacylglycerolhemisuccinates, e.g. DOGS, DMGS, POGS1 DPGS, DSGS; diacylglycerolhemimalonates, e.g. DOGM or DMGM; diacylglycerolhemiglutarates, e.g. DOGG, DMGG; diacylglycerolhemiadipates, e.g. DOGA, DMGA; diacylglycerolhemicyclohexane-1 ,4-dicarboxylic acids, e.g. DO-cHA, DM-cHA; (2,3-Diacyl-propyl)amino}-oxoalkanoic acids e.g. DOAS, DOAM, DOAG, DOAA, DMAS, DMAM, DMAG, DMAA; Diacyl-alkanoic acids, e.g. DOP, DOB, DOS, DOM, DOG, DOA, DMP, DOB, DMS, DMM, DMG, DMA; Chems and derivatives thereof, e.g. Chol-C2, Chol-C3, Chol-C5 or Chol-C6, Chol-C7, Chol-C8, Chol-C1 , fatty acids, e.g. Oleic acid, Linoleic acid, Myristic acid, Palmitic acid, Stearic acid, Nervonic Acid, Behenic Acid; DOPA, DMPA, DPPA, POPA, DSPA, Chol-SO4, DOPG, DMPG, DPPG,
POPG, DSPG or DOPS, DMPS, DPPS, POPS, DSPS, Cetyl-phosphate or any of compounds 1-22 of table 3 above.
11. The liposomes as claimed in claim 10 wherein said one or more anionic lipids are selected from the group consisting of DOGS, DMGS, Chol-C1 , Chol-C2,
Chol-C3, Chems, Chol-C5, Chol-C6, Chol-C7 and Chol-C8.
12. The liposomes as claimed in any of claims 7 to 11 , wherein said neutral or zwitterionic lipids are selected from POPC, DPPC, DSPC, DOPC, DMPC, POPE, DPPE, DSPE, DMPE, DOPE, DPhyPE, DLinPE or natural equivalents thereof and/or cholesterol.
13. The liposomes as claimed in claim 12 wherein said neutral or zwittehonic lipid is cholesterol or a mixture of phosphatidylethanolamine and cholesterol or a mixture of phosphatidylcholine and cholesterol.
14. The liposomes as claimed in claim 12 wherein said neutral or zwitterionic lipid is a mixture of phosphatidylcholine and phosphatidylethanolamine.
15. The liposomes as claimed in any of claims 7 to 14, wherein said liposomes have a size of between 20 to 1000 nm.
16. The liposomes as claimed in any of claims 7 to 15, wherein said liposomes comprise an active agent.
17. The liposomes as claimed in claim 16, wherein said active agent is a nucleic acid.
18. Use of liposomes as claimed in any of claim 7 to 17 as a vector for in vivo, in vitro or ex vivo transfection.
19. Use of liposomes as claimed in any of claim 7 to 17 for intravenous, subcutaneous, peritoneal, local or topical application.
20. Pharmaceutical composition comprising liposomes as claimed in any of claims 7 to 17 particularly for the treatment or prophylaxis of inflammatory, immune or autoimmune disorders, cancer and/or metabolic diseases of humans or non- human animals.
PCT/EP2008/005221 2006-10-13 2008-06-20 Novel facultative catonic sterols WO2008155141A2 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
AU2008309880A AU2008309880B2 (en) 2007-10-12 2008-10-12 Amphoteric liposomes comprising neutral lipids
US12/682,703 US20100330154A1 (en) 2006-10-13 2008-10-12 amphoteric liposomes comprising neutral lipids
CA2702103A CA2702103A1 (en) 2007-10-12 2008-10-12 Improvements in or relating to amphotaric liposomes comprising neutral lipids
JP2010528325A JP5711535B2 (en) 2007-10-12 2008-10-12 Amphoteric liposomes containing neutral lipids
PCT/EP2008/008621 WO2009047006A2 (en) 2007-10-12 2008-10-12 Amphoteric liposomes comprising neutral lipids
EP08837321.2A EP2211840B1 (en) 2007-10-12 2008-10-12 Amphoteric liposomes comprising neutral lipids
US14/049,669 US20140178462A1 (en) 2006-10-13 2013-10-09 Amphoteric liposomes comprising neutral lipids
JP2015017794A JP6192061B2 (en) 2007-10-12 2015-01-30 Amphoteric liposomes containing neutral lipids
JP2017004104A JP2017105792A (en) 2007-10-12 2017-01-13 Improvements in or relating to amphoteric liposomes comprising neutral lipids

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
DE102007029471.0 2007-06-20
DE102007029471A DE102007029471A1 (en) 2007-06-20 2007-06-20 New optional cationic sterols

Publications (2)

Publication Number Publication Date
WO2008155141A2 true WO2008155141A2 (en) 2008-12-24
WO2008155141A3 WO2008155141A3 (en) 2009-03-19

Family

ID=40010682

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2008/005221 WO2008155141A2 (en) 2006-10-13 2008-06-20 Novel facultative catonic sterols

Country Status (2)

Country Link
DE (1) DE102007029471A1 (en)
WO (1) WO2008155141A2 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010113172A1 (en) * 2009-03-31 2010-10-07 Council Of Scientific & Industrial Research Amphoteric liposomal compositions for cellular delivery of small rna molecules for use in rna interference
WO2013006825A1 (en) 2011-07-06 2013-01-10 Novartis Ag Liposomes having useful n:p ratio for delivery of rna molecules
CN104744310A (en) * 2009-07-09 2015-07-01 玛瑞纳生物技术有限公司 Amphoteric liposomes comprising imino lipids
EP2590626B1 (en) 2010-07-06 2015-10-28 GlaxoSmithKline Biologicals SA Liposomes with lipids having an advantageous pka-value for rna delivery
WO2021113365A1 (en) * 2019-12-06 2021-06-10 Guide Therapeutics, Inc. Nanomaterials
US11291635B2 (en) 2010-07-06 2022-04-05 Glaxosmithkline Biological Sa Virion-like delivery particles for self-replicating RNA molecules
US11291682B2 (en) 2010-07-06 2022-04-05 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
WO2022241404A1 (en) * 2021-05-11 2022-11-17 Trustees Of Dartmouth College Liver x receptor modulators
US11639370B2 (en) 2010-10-11 2023-05-02 Glaxosmithkline Biologicals Sa Antigen delivery platforms
US11655475B2 (en) 2010-07-06 2023-05-23 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11759422B2 (en) 2010-08-31 2023-09-19 Glaxosmithkline Biologicals Sa Pegylated liposomes for delivery of immunogen-encoding RNA
US11896636B2 (en) 2011-07-06 2024-02-13 Glaxosmithkline Biologicals Sa Immunogenic combination compositions and uses thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0237051A2 (en) * 1986-03-14 1987-09-16 Fujisawa Pharmaceutical Co., Ltd. Prodrug compounds, process for the preparation thereof and sustained release preparation comprising the same
US5215972A (en) * 1989-11-22 1993-06-01 Hoffmann-La Roche Inc. Steroids
WO2003035669A1 (en) * 2001-10-22 2003-05-01 Sterrenbeld Biotechnologie North America, Inc. Amphiphilic cationic lipids derived from cholesterol
WO2007064857A2 (en) * 2005-12-01 2007-06-07 Pronai Therapeutics, Inc. Amphoteric liposome formulation
CN101016325A (en) * 2007-03-08 2007-08-15 复旦大学 Cholesteric organogel compounds and preparing method theroef

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE19648625A1 (en) 1996-11-13 1998-05-14 Soft Gene Gmbh Microprojectile for the introduction of substances into cells by ballistic transfer
DE19753182A1 (en) 1997-11-21 1999-07-29 Claas Junghans Topologically fixed matrix-bound nucleic acid
US6506559B1 (en) 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
DE10160151A1 (en) 2001-01-09 2003-06-26 Ribopharma Ag Inhibiting expression of target gene, useful e.g. for inhibiting oncogenes, by administering double-stranded RNA complementary to the target and having an overhang
DE10109897A1 (en) 2001-02-21 2002-11-07 Novosom Ag Optional cationic liposomes and their use
ES2240745T3 (en) 2001-06-05 2005-10-16 Curevac Gmbh PHARMACEUTICAL COMPOSITION CONTAINING A STABILIZED AND OPTIMIZED mRNA FOR TRANSLATION, APPROPRIATE AS A VACCINE AND AS A REGENERANT OF FABRICS.

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0237051A2 (en) * 1986-03-14 1987-09-16 Fujisawa Pharmaceutical Co., Ltd. Prodrug compounds, process for the preparation thereof and sustained release preparation comprising the same
US5215972A (en) * 1989-11-22 1993-06-01 Hoffmann-La Roche Inc. Steroids
WO2003035669A1 (en) * 2001-10-22 2003-05-01 Sterrenbeld Biotechnologie North America, Inc. Amphiphilic cationic lipids derived from cholesterol
WO2007064857A2 (en) * 2005-12-01 2007-06-07 Pronai Therapeutics, Inc. Amphoteric liposome formulation
CN101016325A (en) * 2007-03-08 2007-08-15 复旦大学 Cholesteric organogel compounds and preparing method theroef

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
TOKUNAGA Y ET AL: "LIPOSOMAL SUSTAINED-RELEASE DELIVERY SYSTEMS FOR INTRAVENOUS INJECTION I. Physicochemical and biological properties of newly synthesized lipophilic derivatives of mitomycin C" CHEMICAL AND PHARMACEUTICAL BULLETIN, PHARMACEUTICAL SOCIETY OF JAPAN, TOKYO, vol. 36, no. 8, 1 January 1988 (1988-01-01), pages 3060-3069, XP002511753 ISSN: 0009-2363 *
TOKUNAGA Y ET AL: "LIPOSOMAL SUSTAINED-RELEASE DELIVERY SYSTEMS FOR INTRAVENOUS INJECTION IV. ANTITUMOR ACTIVITY OF NEWLY SYNTHESIZED LIPOPHILIC 1-BETA-D ARABINOFURANOSYLCYTOSINE PRODRUG-BEARING LIPOSOMES" CHEMICAL AND PHARMACEUTICAL BULLETIN, PHARMACEUTICAL SOCIETY OF JAPAN, TOKYO, vol. 36, no. 9, 1 January 1988 (1988-01-01), pages 3574-3583, XP002349533 ISSN: 0009-2363 *
WU, JUNCHEN ET AL: "Ultrasound switch and thermal self-repair of morphology and surface wettability in a cholesterol-based self-assembly system" ANGEWANDTE CHEMIE, INTERNATIONAL EDITION , ISSN: 1433-7851, vol. 47, no. 6, 20 December 2007 (2007-12-20), pages 1063-1067, XP002511754 *

Cited By (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010113172A1 (en) * 2009-03-31 2010-10-07 Council Of Scientific & Industrial Research Amphoteric liposomal compositions for cellular delivery of small rna molecules for use in rna interference
CN104744310A (en) * 2009-07-09 2015-07-01 玛瑞纳生物技术有限公司 Amphoteric liposomes comprising imino lipids
US11839686B2 (en) 2010-07-06 2023-12-12 Glaxosmithkline Biologicals Sa Lipid formulations with viral immunogens
US11690862B1 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11913001B2 (en) 2010-07-06 2024-02-27 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11690861B2 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11891608B2 (en) 2010-07-06 2024-02-06 Glaxosmithkline Biologicals Sa Immunization of large mammals with low doses of RNA
US11291635B2 (en) 2010-07-06 2022-04-05 Glaxosmithkline Biological Sa Virion-like delivery particles for self-replicating RNA molecules
US11291682B2 (en) 2010-07-06 2022-04-05 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US20220125723A1 (en) 2010-07-06 2022-04-28 Glaxosmithkline Biologicals Sa Lipid formulations with viral immunogens
US11324770B2 (en) 2010-07-06 2022-05-10 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11883534B2 (en) 2010-07-06 2024-01-30 Glaxosmithkline Biologicals Sa Immunisation with lipid formulations with RNA encoding immunogens
US11865080B2 (en) 2010-07-06 2024-01-09 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11857681B2 (en) 2010-07-06 2024-01-02 Glaxosmithkline Biologicals Sa Lipid formulations with RNA encoding immunogens
EP2590626B1 (en) 2010-07-06 2015-10-28 GlaxoSmithKline Biologicals SA Liposomes with lipids having an advantageous pka-value for rna delivery
US11596645B2 (en) 2010-07-06 2023-03-07 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11638694B2 (en) 2010-07-06 2023-05-02 Glaxosmithkline Biologicals Sa Vaccine for eliciting immune response comprising lipid formulations and RNA encoding multiple immunogens
US11638693B2 (en) 2010-07-06 2023-05-02 Glaxosmithkline Biologicals Sa Vaccine for eliciting immune response comprising RNA encoding an immunogen and lipid formulations comprising mole percentage of lipids
US11690863B2 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11655475B2 (en) 2010-07-06 2023-05-23 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11666534B2 (en) 2010-07-06 2023-06-06 Glaxosmithkline Biologicals Sa Methods of administering lipid formulations with viral immunogens
US11851660B2 (en) 2010-07-06 2023-12-26 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11690865B2 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11690864B2 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11696923B2 (en) 2010-07-06 2023-07-11 Glaxosmithkline Biologicals, Sa Delivery of RNA to trigger multiple immune pathways
US11905514B2 (en) 2010-07-06 2024-02-20 Glaxosmithkline Biological Sa Immunisation of large mammals with low doses of RNA
US11850305B2 (en) 2010-07-06 2023-12-26 Glaxosmithkline Biologicals Sa Method of making lipid formulations with RNA encoding immunogens
US11707482B2 (en) 2010-07-06 2023-07-25 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11717529B2 (en) 2010-07-06 2023-08-08 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11730754B2 (en) 2010-07-06 2023-08-22 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11739334B2 (en) 2010-07-06 2023-08-29 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11845925B2 (en) 2010-07-06 2023-12-19 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11759475B2 (en) 2010-07-06 2023-09-19 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11766401B2 (en) 2010-07-06 2023-09-26 Glaxosmithkline Biologicals Sa Methods of administering lipid formulations with immunogens
US11773395B1 (en) 2010-07-06 2023-10-03 Glaxosmithkline Biologicals Sa Immunization of large mammals with low doses of RNA
US11786467B2 (en) 2010-07-06 2023-10-17 Glaxosmithkline Biologicals Sa Lipid formulations with immunogens
US11857562B2 (en) 2010-07-06 2024-01-02 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11759422B2 (en) 2010-08-31 2023-09-19 Glaxosmithkline Biologicals Sa Pegylated liposomes for delivery of immunogen-encoding RNA
US11639370B2 (en) 2010-10-11 2023-05-02 Glaxosmithkline Biologicals Sa Antigen delivery platforms
EP4115876A1 (en) * 2011-07-06 2023-01-11 GlaxoSmithKline Biologicals S.A. Liposomes having useful n:p ratio for delivery of rna molecules
WO2013006825A1 (en) 2011-07-06 2013-01-10 Novartis Ag Liposomes having useful n:p ratio for delivery of rna molecules
EP4115875A1 (en) * 2011-07-06 2023-01-11 GlaxoSmithKline Biologicals S.A. Liposomes having useful n:p ratio for delivery of rna molecules
EP4014966A1 (en) * 2011-07-06 2022-06-22 GlaxoSmithKline Biologicals S.A. Liposomes having useful n:p ratio for delivery of rna molecules
US11896636B2 (en) 2011-07-06 2024-02-13 Glaxosmithkline Biologicals Sa Immunogenic combination compositions and uses thereof
EP3821879A1 (en) * 2011-07-06 2021-05-19 GlaxoSmithKline Biologicals S.A. Liposomes having useful n:p ratio for delivery of rna molecules
EP2729126B1 (en) * 2011-07-06 2020-12-23 GlaxoSmithKline Biologicals SA Liposomes having useful n:p ratio for delivery of rna molecules
WO2021113365A1 (en) * 2019-12-06 2021-06-10 Guide Therapeutics, Inc. Nanomaterials
WO2022241404A1 (en) * 2021-05-11 2022-11-17 Trustees Of Dartmouth College Liver x receptor modulators

Also Published As

Publication number Publication date
DE102007029471A1 (en) 2008-12-24
WO2008155141A3 (en) 2009-03-19

Similar Documents

Publication Publication Date Title
WO2008155141A2 (en) Novel facultative catonic sterols
EP1478652B1 (en) Components for producing amphoteric liposomes
US20200330384A1 (en) Amphoteric liposomes comprising imino lipids
JP6192061B2 (en) Amphoteric liposomes containing neutral lipids
US20080306153A1 (en) Lipids and lipid assemblies comprising transfection enhancer elements
JP2012509273A (en) Releasable fusible lipids for nucleic acid delivery systems
EP2364085A1 (en) Releasable cationic lipids for nucleic acids delivery systems
WO2010014895A2 (en) Nanoparticle compositions for nucleic acids delivery system
EP2350296A1 (en) Branched cationic lipids for nucleic acids delivery system
EP2362728A1 (en) Releasable polymeric lipids for nucleic acids delivery system
EP1938843A1 (en) Lipids and lipid assemblies comrising transfection enhancer elements
JP6774965B2 (en) Compounds as cationic lipids
US20140178462A1 (en) Amphoteric liposomes comprising neutral lipids
AU2014240370B2 (en) Amphoteric liposomes comprising neutral lipids
CN117658848A (en) Lipid compounds for delivery of therapeutic agents and uses thereof

Legal Events

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

Ref document number: 08773697

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08773697

Country of ref document: EP

Kind code of ref document: A2