US20050244501A1 - Water-soluble stabilized self-assembled polyelectrolytes - Google Patents

Water-soluble stabilized self-assembled polyelectrolytes Download PDF

Info

Publication number
US20050244501A1
US20050244501A1 US11/175,796 US17579605A US2005244501A1 US 20050244501 A1 US20050244501 A1 US 20050244501A1 US 17579605 A US17579605 A US 17579605A US 2005244501 A1 US2005244501 A1 US 2005244501A1
Authority
US
United States
Prior art keywords
assemblies
vinyl
hydrophobic
units
group
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/175,796
Inventor
Maxime Ranger
Jean-Christophe Leroux
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Labopharm Inc
Original Assignee
Labopharm Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Labopharm Inc filed Critical Labopharm Inc
Priority to US11/175,796 priority Critical patent/US20050244501A1/en
Publication of US20050244501A1 publication Critical patent/US20050244501A1/en
Assigned to LABOPHARM INC. reassignment LABOPHARM INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LEROUX, JEAN-CHRISTOPHE, RANGER, MAXIME
Priority to US11/558,543 priority patent/US7510731B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08LCOMPOSITIONS OF MACROMOLECULAR COMPOUNDS
    • C08L71/00Compositions of polyethers obtained by reactions forming an ether link in the main chain; Compositions of derivatives of such polymers
    • C08L71/02Polyalkylene oxides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid, pantothenic acid
    • A61K31/198Alpha-aminoacids, e.g. alanine, edetic acids [EDTA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/216Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acids having aromatic rings, e.g. benactizyne, clofibrate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/275Nitriles; Isonitriles
    • A61K31/277Nitriles; Isonitriles having a ring, e.g. verapamil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/282Platinum compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • A61K9/1075Microemulsions or submicron emulsions; Preconcentrates or solids thereof; Micelles, e.g. made of phospholipids or block copolymers
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08FMACROMOLECULAR COMPOUNDS OBTAINED BY REACTIONS ONLY INVOLVING CARBON-TO-CARBON UNSATURATED BONDS
    • C08F293/00Macromolecular compounds obtained by polymerisation on to a macromolecule having groups capable of inducing the formation of new polymer chains bound exclusively at one or both ends of the starting macromolecule
    • C08F293/005Macromolecular compounds obtained by polymerisation on to a macromolecule having groups capable of inducing the formation of new polymer chains bound exclusively at one or both ends of the starting macromolecule using free radical "living" or "controlled" polymerisation, e.g. using a complexing agent
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08GMACROMOLECULAR COMPOUNDS OBTAINED OTHERWISE THAN BY REACTIONS ONLY INVOLVING UNSATURATED CARBON-TO-CARBON BONDS
    • C08G83/00Macromolecular compounds not provided for in groups C08G2/00 - C08G81/00
    • C08G83/008Supramolecular polymers
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08LCOMPOSITIONS OF MACROMOLECULAR COMPOUNDS
    • C08L51/00Compositions of graft polymers in which the grafted component is obtained by reactions only involving carbon-to-carbon unsaturated bonds; Compositions of derivatives of such polymers
    • C08L51/003Compositions of graft polymers in which the grafted component is obtained by reactions only involving carbon-to-carbon unsaturated bonds; Compositions of derivatives of such polymers grafted on to macromolecular compounds obtained by reactions only involving unsaturated carbon-to-carbon bonds
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08LCOMPOSITIONS OF MACROMOLECULAR COMPOUNDS
    • C08L53/00Compositions of block copolymers containing at least one sequence of a polymer obtained by reactions only involving carbon-to-carbon unsaturated bonds; Compositions of derivatives of such polymers

Definitions

  • This invention generally relates to the preparation of water-soluble self-assemblies, particularly to the association of ionizable or permanently-charged copolymers, and most particularly to the inclusion of hydrophobic comonomers to the polyelectrolyte segment forming the assembly core.
  • Polymeric micelles have been the object of growing scientific attention, and have emerged as potential carriers for drugs having poor water solubility because they can solubilize those drugs in their inner core and they offer attractive characteristics such as a generally small size ( ⁇ 300 nm) and a propensity to evade scavenging by the mononuclear phagocyte system.
  • Micelles are often compared to naturally occurring carriers such as viruses or lipoproteins. All three of these carriers demonstrate a similar core-shell structure that allows for their contents to be protected during transportation to the target cell, whether it is DNA for viruses or water-insoluble drugs for lipoproteins and micelles.
  • Polymeric micelles seem to be one of the most advantageous carriers for the delivery of poorly water-soluble drugs as reported by Jones and Leroux, Eur. J. Pharm. Biopharm . (1999) 48, 101-111; Kwon and Okano, Adv. Drug Deliv. Rev . (1996) 21, 107-116 and Allen et al. Colloids Surf. B: Biointerf . (1999) 16, 3-27. They are characterized by a core-shell structure.
  • the hydrophobic inner core generally serves as a microenvironment for the solubilization of poorly water-soluble drugs, whereas the hydrophilic outer shell is responsible for micelle stability, protection against opsonization, and uptake by the mononuclear phagocyte system.
  • Multiblock copolymers such as poly(ethylene oxide)-poly(propylene oxide)-poly(ethylene oxide) (PEO-PPO-PEO) (A-B-A) can also self-organize into micelles, and have been described as potential drug carriers, e.g. Kabanov et al., FEBS Lett . (1989) 258, 343-345.
  • the hydrophobic core which generally consists of a biodegradable polymer such as a poly( ⁇ -benzyl-aspartate) (PBLA), poly(D,L-lactic acid) or poly( ⁇ -caprolactone), serves as a reservoir for a poorly water-soluble drug, protecting it from contact with the aqueous environment.
  • the core may also consist of a water-soluble polymer, such as poly(aspartic acid) (P(Asp)), which is rendered hydrophobic by the chemical conjugation of a hydrophobic drug, or is formed through the association of two oppositely charged polyions (PICM).
  • the hydrophobic inner core can also consist of a highly hydrophobic small chain such as an alkyl chain or a diacyllipid (e.g.
  • the hydrophobic chain can be either attached to one end of a polymer, or randomly distributed within the polymeric structure.
  • the shell usually consists of chains of hydrophilic, non-biodegradable, biocompatible polymers such as poly(ethylene oxide) (PEO) (see Allen et al. Colloids Surf. B: Biointerf . (1999) 16, 3-27 and Kataoka et al. J. Controlled Release (2000) 64, 143-153), poly(N-vinyl-2-pyrrolidone) (PVP) (see Be Spotifyd A et al. Pharm Res (2001) 18, 323-328) or poly(2-ethyl-2-oxazoline) (see Lee et al. Macromolecules (1999) 32, 1847-1852).
  • PEO poly(ethylene oxide)
  • PVP poly(N-vinyl-2-pyrrolidone)
  • the biodistribution of the carrier is mainly dictated by the nature of the hydrophilic shell.
  • Other polymers such as poly(N-isopropylacrylamide) and poly(alkylacrylic acid) impart temperature or pH sensitivity to the micelles, and could eventually be used to confer bioadhesive properties (see U.S. Pat. No. 5,770,627).
  • Micelles presenting functional groups at their surface for conjugation with a targeting moiety have also been described (See, e.g., Scholz, C. et al., Macromolecules (1995) 28, 7295-7297).
  • Such copolymers are positively charged at acidic pH due to the protonation of the nitrogen atoms, and thus exist as unimers in acidic solutions.
  • the unprotonated copolymers self-associate into polymeric micelles.
  • These micelles are primarily intended to deliver their contents at low pH, since the dissociation of the supramolecular assembly under acidic conditions allows a drug to be released. Such conditions may be found, for example in tumors. If intended to be administered by the oral route, these micelles would rapidly release their contents in the stomach because of its acidic pH. Therefore, for oral delivery, they should be formulated using an enteric coating to prevent premature drug leakage.
  • a potential problem with ionizable copolymers is the possibility of forming, at acidic pH, intra and inter-molecular hydrogen bonding between the protonated and the non-ionizable hydrophilic blocks which might lead to the formation of an insoluble complex. This has been recently described by Lele et al. J. Controlled Release (2000) 69, 237-248, between poly(acrylic acid) and poly(ethylene glycol). Precipitation of the micelles at acidic pH could potentially compromise the efficacy of the system when oral delivery is envisaged.
  • PICM have a block or graft copolymer architecture and consist of a polyelectrolyte linked to a non-ionic water-soluble polymer. They bind with charged compounds due to electrostatic interactions with the polyelectrolyte (see, e.g., Kataoka et al. Macromolecules (1996) 29, 8556-8557).
  • the complexes self-assemble into micelle-like structures which have a hydrophobic core from neutralized polyelectrolyte and counterion, and hydrophilic corona.
  • PICM show improved solubility compared with other electrostatic complexes. Furthermore, they show reduced affinity for plasma components and can protect active compounds such as DNA against enzymatic degradation.
  • PICM hold great promise as carriers for a variety of selected from the group consisting of ionizable and permanently-charged diblock copolymers, ionizable and permanently-charged multiblock copolymers, and ionizable and permanently-charged random copolymers with grafted hydrophilic and essentially non-ionic oligomers or polymers compounds, such as charged drugs and nucleic acids, some important issues still remain to be addressed.
  • the stability of the polymeric micelles is influenced by various factors such as concentration, temperature and chemical structure of the polymer.
  • the presence of salts is a key parameter for the dissociation of PICM since Coulombic interactions between charged segments are screened by the added salt.
  • polymeric micelles can be stabilized by cross-linking the core or shell (see, e.g., Kakizawa et al. J. Am. Chem. Soc . (1999) 121, 11247-11248).
  • cross-linking the core or shell can potentially chemically alter the active agent and/or excessively slow down its release from the micelles.
  • U.S. Pat. No. 5,693,751 teaches the preparation of polymeric micelles composed of a water-soluble block copolymer having a hydrophilic segment and a hydrophobic pharmacological-functioning segment to a side chain of which a drug is covalently bonded.
  • U.S. Pat. No. 5,702,717 teaches the preparation of a solution comprising a drug and a block amphiphilic copolymer of poly(ethylene glycol) and poly( ⁇ -hydroxy acids) or poly(ethylene carbonates). These copolymers are not polyelectrolytes.
  • U.S. Pat. No. 5,939,453 teaches the preparation of polymeric micelles and bioerodible drug delivery matrix using poly(ethylene glycol)-poly(orthoester) diblock and triblock copolymers.
  • the principal novelty of this invention relies on the use of poly(orthoester) as the hydrophobic, bioerodible segment.
  • These block copolymers are neither ionizable or permanently charged.
  • U.S. Pat. No. 5,786,387 teaches the preparation of supramolecular assemblies using lipid double chain derivatives containing poly(oxyethylene) for drug delivery applications. These block copolymers are neither ionizable or permanently charged. They efficiently are able to avoid the reticuloendothelial system and possess a long circulation time.
  • U.S. Pat. No. 5,840,319 teaches the preparation of polymeric micelles using polyether block copolymers having a critical micelle concentration of no more than 0.5% (w/w) at 37° C. in an isotonic aqueous solution.
  • the formulation also contains a chemotherapeutic agent.
  • U.S. Pat. No. 5,770,627 teaches the preparation of hydrophobically-modified bioadhesive polyelectrolytes. These polyelectrolytes can form micellar structures in aqueous solutions (example 6) and be loaded with an active agent.
  • the polyelectrolyte can be a graft or block copolymer.
  • the outer shell is ionizable since it contains carboxylic acid groups whereas the inner core consists of a homopolymer, copolymer, unsaturated or saturated alkyl chains, or other hydrophobic moities. Methods of administering such agents to an animal are disclosed.
  • U.S. Pat. No. 5,955,509 relates to the use of poly(vinyl N-heterocycle)-block-poly(alkylene oxide) copolymers in micelle containing pharmaceutical formulations.
  • the copolymers advantageously respond to pH differences in the environment to which they are exposed forming micelles at higher pH values.
  • the micelles which comprise a therapeutic compound and a copolymer, deliver drug in a pH dependent manner.
  • U.S. Pat. No. 5,929,177 provides a block polymer which has functional groups on both ends thereof, and which comprises hydrophilic/hydrophobic segments.
  • the block polymer has amino group, carboxyl group or mercapto group on the alpha terminal, and hydroxyl group, carboxyl group, aldehyde group or vinyl group on the omega terminal.
  • Hydrophilic segment comprises polyethylene oxide, while hydrophobic segment is derived from lactide, lactone or (meth)acrylic acid ester.
  • the block polymer of this invention forms a polymeric micelle which is usable as bio-compatible materials.
  • U.S. Pat. No. 5,925,720 provides a heterotelechelic oligomer or polymer which can be prepared by means of living polymerization and which can form stable micelles in an aqueous solvent. In this invention, there is no reference to stabilization of a polyelectrolyte micelle core.
  • U.S. Pat. No. 5,656,611 relates to compositions for stabilizing polynucleic acids using polyionic complexes.
  • the invention provides a polynucleotide complex between a polynucleotide and a block copolymer comprising a polyether block and a polycation block.
  • U.S. Pat. No. 6,217,912 provides a biodegradable composition suitable for delivering a gene into a cell.
  • U.S. Pat. No. 6,221,959 provides composition for stabilizing polynucleic acids and increasing the ability of polynucleic acid to cross cell membranes and act in the interior of a cell.
  • the invention provides a polynucleotide complex between a polynucleotide and certain polyether block copolymer.
  • U.S. Pat. No. 5,510,103 relates to drug carriers composed of a block copolymer having hydrophilic and hydrophobic segments, a polymeric type drug comprising hydrophobic drugs trapped by physical treatments in said drug carrier and methods for trapping hydrophobic drugs in the carrier.
  • the present invention is directed toward water-soluble supramolecular self-assemblies and a process for their preparation via micellization of polyelectrolytes through the use of hydrophobic monomeric units.
  • the polyelectrolyte segment ultimately forms the core of the supramolecular assembly whereas the shell consists of uncharged hydrophilic polymers or oligomers.
  • the inclusion of the hydrophobic co-monomers to the polyelectrolyte segment forming the micelle core leads to a structure of enhanced stability.
  • Such co-monomers by increasing the attractive forces between the segments of the core, stabilize the micelles and/or decrease the interaction between the ionizable or permanently-charged segment and non-ionizable segment of the copolymer.
  • FIG. 1 is a synthetic route for the preparation of block copolymer, possessing a hydrophobized polyelectrolyte block;
  • FIG. 2 is the 1 H NMR spectrum of PEG-b-P(DMAEMA 30 -co-EMA 70 ) in CDCl 3 ;
  • FIG. 3 shows the variation in light scattering and pyrene fluorescence I 338 /I 333 ratio of a PEG-b-P(EA 50 -co-MAA 50 ) aqueous solution as a function of pH;
  • FIG. 4 shows the 1 H NMR spectrum of PEG-b-P(EA 50 -co-MAA 50 ) in D 2 O at pH 10 (A) and at pH 3 (B).
  • water-soluble self-assemblies and “micelles” are equally employed although the proposed structures may not necessarily correspond to the true definition of micelles.
  • Micelle formation occurs as a result of two forces. One is an attractive force that leads to the association of molecules, while the other is a repulsive force that prevents unlimited growth of the micelles to a distinct macroscopic phase.
  • Amphiphilic copolymers self-associate when placed in a solvent that is selective for either the hydrophilic or hydrophobic polymer.
  • the micellization process of amphiphilic copolymers is similar to that for low molecular weight surfactants.
  • the polymers exist only as single chains.
  • CAC critical association concentration
  • polymer chains start to associate to form micelles in such a way that the hydrophobic part of the copolymer will avoid contact with the aqueous media in which the polymer is diluted.
  • Amphiphilic copolymers usually exhibit a CAC which is much lower than that of low molecular weight surfactants.
  • the CAC of PEO-PBLA and PNIPA-PSt are between 0.0005-0.002%.
  • amphiphilic copolymers exhibit much higher CAC, reaching up to 0.01-10% in the case of poloxamers.
  • Amphiphilic copolymers with high CAC may not be suitable as drug targeting devices since they are unstable in an aqueous environment and are easily dissociated upon dilution.
  • Micelles can be targeted to specific cells or tissues via the inclusion of targeting ligands, e.g. monoclonal antibodies, lectins, sugars, vitamins, peptides or immunologically distinct fragments thereof or the like moieties which provide the micelles with an ability to preferentially concentrate in a particular target area.
  • targeting ligands e.g. monoclonal antibodies, lectins, sugars, vitamins, peptides or immunologically distinct fragments thereof or the like moieties which provide the micelles with an ability to preferentially concentrate in a particular target area.
  • micellization of amphiphilic copolymers can result in two different types of micelles depending on whether the hydrophobic chain is randomly bound to the hydrophilic polymer or grafted to one end of the hydrophilic chain.
  • Micelles formed from randomly modified polymers are generally smaller than end-modified polymers.
  • the micellar size is mainly determined by the hydrophobic forces which sequester the hydrophobic chains in the core, and by the excluded volume repulsion between the chains which limits their size. The difference in the balance of these two forces in random and end-modified copolymers may account for their different size.
  • CAC Critical Association Concentration
  • a preferred method to determine the CAC involves the use of fluorescent probes, among which pyrene is widely used.
  • Pyrene is a condensed aromatic hydrocarbon that is highly hydrophobic and sensitive to the polarity of the surrounding environment. Below the CAC, pyrene is solubilized in water, a medium of high polarity. When micelles are formed, pyrene partitions preferentially toward the hydrophobic domain afforded by the micellar core, and thus experiences a nonpolar environment. Consequently, numerous changes such as an increase in the fluorescence intensity, a change in the vibrational fine structure of the emission spectra, and a red shift of the (0,0) band in the excitation spectra are observed.
  • the apparent CAC can be obtained from the plot of the fluorescence of pyrene, the I 1 /I 3 ratio from emission spectra or the I 338 /I 333 ratio from the excitation spectra versus concentration.
  • a major change in the slope indicates the onset of micellization. Changes in anisotropy of fluorescent probes have also been associated with the onset of micellization. E.g. see Jones and Leroux Eur. J. Pharm. Biopharm . (1999) 48, 101-111.
  • Polymeric micelles such as those of the compositions of the invention are characterized by their small size. Besides being needed for extravasation of the carrier materials, this small size permits the sterilization of the composition to be effected simply by filtration, and minimizes the risks of embolism in capillaries after intravenous injection.
  • Micellar size depends on several factors including copolymer molecular weight, relative proportion of hydrophilic and hydrophobic chains and aggregation number.
  • Micellar diameter and size polydispersity can be obtained directly in water or in an isotonic buffer by dynamic light scattering (DLS).
  • Micelle size can also be estimated by methods such as atomic force microscopy (AFM), transmission electron microscopy (TEM) and scanning electron microscopy (SEM). These methods allow the characterization of the micelle shape and size dispersity. Ultracentrifugation velocity studies are sometimes performed to assess the polydispersity of polymeric micelles.
  • Loading of one or more pharmacological constituents, e.g. various therapeutic agents, drugs, peptides, proteins, genetic material (e.g. oligonucleotides), genetically altered constituents, polyionic constituents and the like, into the micelles can be realized according to techniques well known to one skilled in the art.
  • drugs can be incorporated into the polymeric micelle compositions of the invention by means of chemical conjugation or by physical entrapment through dialysis, emulsification techniques, simple equilibration of the drug and micelles in an aqueous medium or solubilization of a drug/polymer solid dispersion in water.
  • Therapeutic agents which may be used are any compounds, including the ones listed below, which can be entrapped, in a stable manner, in polymeric micelles and administered at a therapeutically effective dose.
  • the therapeutic agents used in accordance with the invention are hydrophobic or polyionic (e.g. DNA).
  • Suitable drugs include antitumor compounds such as phthalocyanines (e.g. aluminum chloride phthalocyanine), anthracyclines (e.g. doxorubicin), poorly soluble antimetabolites (e.g. methotrexate, mitomycin, 5-fluorouracil) and alkylating agents (e.g. carmustine).
  • Micelles may also contain taxanes such as paclitaxel.
  • Additional drugs which can be contained in micelles are conventional hydrophobic antibiotics and antifungal agents such as amphotericin B and itraconazole, poorly water-soluble immunomodulators such as cyclosporin, poorly water-soluble antiviral drugs such as HIV protease inhibitors and poorly water-soluble steroidal (e.g. dexamethasone), and non-steroidal (e.g. indomethacin) anti-inflammatory drugs.
  • conventional hydrophobic antibiotics and antifungal agents such as amphotericin B and itraconazole
  • poorly water-soluble immunomodulators such as cyclosporin
  • poorly water-soluble antiviral drugs such as HIV protease inhibitors and poorly water-soluble steroidal (e.g. dexamethasone)
  • non-steroidal (e.g. indomethacin) anti-inflammatory drugs e.g. indomethacin) anti-inflammatory drugs.
  • Hydrophilic compounds such as proteins may also be incorporated in the polymeric micelle compositions of the invention.
  • the incorporation of such hydrophilic species may, however, require the chemical hydrophobization of the molecule or a particular affinity for the hydrophilic shell.
  • Polyionic compounds e.g. antisense oligonucleotides, genome fragments, peptides
  • the polymeric micelle compositions of the invention are suitable for use in a variety of pharmaceutical fields, such as oral delivery, sustained release and site-specific drug targeting.
  • the micelles of the invention are used as a transport for water-insoluble and polyionic compounds.
  • supramolecular assembly compositions comprising a suitable targeting ligand.
  • block polyelectrolytes useful in the present invention are most preferably prepared by “living”/controlled radical polymerization (LCRP), such as atom transfer radical polymerization (ATRP) (see Coessens et al., Prog. Polym. Sci . (2001) 26, 337-377) or nitroxide-mediated radical polymerization (NMP) (see Benoit et al. J. Am. Chem. Soc . (1999), 121, 3904-3920).
  • LCRP living/controlled radical polymerization
  • ATRP atom transfer radical polymerization
  • NMP nitroxide-mediated radical polymerization
  • any alternative procedure such as other living radical polymerizations or condensation of preformed functionalized polymers could also be used.
  • the radical initiator for the synthesis of the polymer by ATRP can be any appropriately functionalized molecule (e.g. poly(ethylene glycol) (PEG), PVP).
  • the initiator bears an halogeno functionality that can be activated for ATRP (see Coessens et al., Prog. Polym. Sci . (2001) 26, 337-377). Without intending to be limited to any particular substituent, this functionality can be a 2-halogenoisobutyrylate derivative, 2-halogenopropionate derivative, 2-halogenoacetate derivative or 1-(halogenomethyl)benzene derivative.
  • this functionality is appropriately chosen according to the used monomers.
  • the catalyst for the ATRP usually includes a metallic salt and a ligand.
  • the ligand is used for the solubilization of the salt in organic solvent and/or to activate the redox reaction of the metal present in the salt.
  • the salt activates the radical initiator for the ATRP. Without intending to be limited to any particular salt, the latter can be copper(I) bromide, copper(I) chloride or copper(I) thiocyanate, iron(II) and nickel(0 or I) compounds.
  • the ligand can include 2,2′-bipyridine derivatives or bis(dimethylamino) compounds (e.g. N,N,N′,N′,N′′,N′′-pentamethyldiethylene-triamine (PMDETA).
  • Suitable polyelectrolyte compounds useful in the preparation of supramolecular self-assemblies may be selected from the group consisting of diblock copolymers including ionizable units, permanently charged units or mixtures of ionizable and permanently charged units, multiblock copolymers including ionizable units, permanently charged units or mixtures of ionizable and permanently charged units, and random copolymers with grafted hydrophilic and essentially non-ionic oligomers or polymers, said random copolymers including ionizable units, permanently charged units or mixtures of ionizable and permanently charged units.
  • the diblock copolymers generally consist of two blocks, one of which is hydrophilic and generally uncharged and the other containing at least one compound selected from the group consisting of ionizable and permanently-charged repeating units (or combinations thereof) in combination with essentially hydrophobic, e.g. hydrophobic or relatively hydrophobic non-ionic monomers.
  • Ionizable units refers to repeating units that can be transformed from a non-ionic to a charged state via an external stimulus (e.g. pH or chemical reaction).
  • Permanently charged units refers to repeating units that are in fact electrostatically charged irrespective of the external conditions.
  • the ionizable and/or permanently-charged block, bearing hydrophobic repeating units can be synthesized from vinyl monomers, vinyl oligomers or eventually vinyl polymers.
  • These hydrophobic monomers/oligomers/polymers can be acrylate, acrylamide, alkylacrylate, alkylacrylamide, arylacrylate and arylacrylamide derivatives for which the alkyl and aryl term stands for aliphatic or aromatic moieties respectively (e.g. methacrylate, methacrylamide derivatives.)
  • the hydrophobic compound can also be a biodegradable polyester such as vinyl-terminated poly(lactide) and vinyl-terminated poly( ⁇ -caprolactone).
  • the ionizable monomers could be alkylacrylic acid derivatives, (aminoalkyl)acrylate or (aminoalkyl)alkylacrylate derivatives.
  • the acidic or basic units of the polymer chain can be derived from a non-ionizable precursor (e.g. tert-butylmethacrylate), which is cleaved into an acidic moiety.
  • the hydrophilic block can be synthesized from vinyl monomers, vinyl oligomers or eventually vinyl polymers. These hydrophilic monomers/oligomers/polymers can be acrylate, acrylamide, alkylacrylate and alkylacrylamide (e.g. PEG methacrylate and N-(2-hydroxypropyl)acrylamide). On the other hand, the hydrophilic block can also originate from a block radical macroinitiator based on PEG or PVP derivatives.
  • Non biodegradable ionizable and/or permanently-charged copolymers that are intended to be administered parenterally should have a molecular weights not exceeding 40,000. There is no restriction on molecular weights for biodegradable or non-biodegradable ionizable copolymers, which are used orally or locally.
  • the loading of poorly water-soluble and non-ionic drugs should be done in an organic solvent, or in aqueous solutions at a pH where the core is uncharged or mostly uncharged.
  • Charged drugs should be loaded under conditions (e.g. pH) where electrostatic interactions with the ionizable or permanently-charged segment are possible.
  • the subscript text indicates the ratio in a polymeric segment.
  • the letter b features that polymers and/or polymeric arms are based on a diblock copolymeric structure.
  • the term co means the repeating units are disposed randomly along the polymeric segment.
  • the ATRP of monomers was carried out in bulk and in solution, using ⁇ -(2-bromoisobutyrylate)- ⁇ -methyl-PEG as ATRP macroinitiator.
  • the PEG ATRP macroinitiator (1 eq.) was added to a solution containing PMDETA (1.1 eq.), Cu(I)Br (1.1 eq.), EMA (14 eq) and DMAEMA (6 eq) in THF (0.8 M).
  • the mixture was degassed with argon for 15-20 min at room temperature and was then heated to 60° C. overnight. After the polymerization, the mixture was poured in THF, containing 10% of methanol.
  • Copper (I) bromide-pentamethyldiethylenetriamine (CuBr-PMDETA) was used as catalyst and gave yields of polymerization approaching 100% in THF.
  • the reactivities of EMA and DMAEMA in THF were similar, with a k app of 1.95 ⁇ 10 ⁇ 4 Lmol ⁇ 1 s ⁇ 1 . All monomers were completely consumed after 5 h and the Mn obtained experimentally were close to the theoretical values (Table 1).
  • the polydispersity index (PI) was approximately 1.4 and corresponded approximately to the polydispersity of PEG macroinitiator used for the preparation of PEG-b-P(DMAEMA 30 -co-EMA 70 ) TABLE 1 M n and M w of P (DMAEMA 30 -co-EMA 70 ).
  • M n M n M n Copolymers Theo NMR SEC PI PEG-b-P(DMAEMA 30 -co-EMA 70 ) 4690 4900 4700 1.4
  • FIG. 2 shows the 1 H NMR spectrum of PEG-b-P(DMAEMA 30 -co-EMA 70 ).
  • the terminal methoxy group of PEG (signal 1 , 3.35 ppm), was used as a reference to calibrate the integration of the other signals.
  • the integration of signal 2 showed that the degree of polymerization (DP) of ethylene oxide (PEG chains) was approximately 45 in each copolymer and corresponded to the DP of the commercial PEG used.
  • the CAC of PEG-b-P(DMAEMA 30 -co-EMA 70 ) was determined in water and phosphate buffered saline (PBS, pH 7.4) to verify whether salts could interfere with self-assembling.
  • PBS phosphate buffered saline
  • the CAC was not significantly affected by the presence of salts in water. This is an important issue, since a common drawback about PICM is their relative poor stability in physiological media.
  • the stability of PICM can be easily increased by introducing a hydrophobic comonomer in the polymer backbone.
  • the micelle sizes for PEG-b-P(DMAEMA 30 -co-EMA 70 the nature of the aqueous solution (water vs PBS) seemed to influence the proportion of secondary aggregates (Table 2).
  • ester groups bearing a tert-butyl chain (tBMA)
  • tBMA tert-butyl chain
  • the methacrylic acid derivatives were precipitated in diethyl ether and filtered.
  • the polymers were dissolved in ethanol, dialyzed against water and freeze-dried.
  • PEG-b-P Poly(ethylene glycol)-block-poly(methacrylic acid) PEG-b-P (MAA); M n : 3180; Obtained from PEG-b-P (tBMA) after the cleavage of tert-butyl groups
  • Well-defined acidic (ionizable) copolymers containing or not hydrophobic units were prepared by ATRP using a PEGylated ATRP macroinitiator.
  • This macroinitiator was synthesized by coupling PEG monomethyl ether to 2-bromoisobutyryl bromide with a high yield.
  • Copper (I) bromide-pentamethyldiethylenetriamine (CuBr-PMDETA) was used as catalyst and gave yields of polymerization approaching 100% in THF.
  • the Mn estimated by NMR were calculated from the terminal methoxy group of the PEG chain ( ⁇ 3.4 ppm). All monomers were completely consumed after 5 h and the M n obtained experimentally were close to the theoretical values.
  • the PEG-b-P(EA 50 -co-tBMA 50 ) and PEG-b-P(tBMA) precursors were transformed into their respective P(MAA) derivatives by cleaving the tert-butyl groups in presence of hydrochloric acid in dioxane.
  • the monomer ratios of the prepared copolyvinyl polymers corresponded approximately to the proportions of monomers used for the polymerization.
  • the hydrophobic unit EA was incorporated in the polyvinyl segment, to increase hydrophobicity. At high pH values, the copolymers are fully ionized and should be in solution mostly as individual polymeric chains.
  • FIG. 3 shows the ratio of fluorescence intensities (I 338 /I 333 ) versus pH, and the first-order derivative of scattered light as a function of pH for PEG-b-P(EA 50 -co-MAA 50 ).
  • PEG-b-P(EA 50 -co-MAA 50 ) remained soluble at acidic pH, indicating that the protonation of MAA units did not make the diblock copolymers precipitate.
  • PEG-b-P(MAA) a diffuse precipitate appeared when the pH was decreased to around pH 3.5-4. It is known that acrylic acid polymeric derivatives interact through hydrogen bonding with PEG in acidic solutions, resulting in precipitation of the polymers. Without intending to be bound to any specific mechanism, we believe that in the presence of the hydrophobic ethyl acrylate comonomer the attractive forces between the protonated MAA and ethylene oxide units are sterically hindered.
  • PEG-b-P(EA 50 -co-MAA 50 ) remains soluble at acidic pHs, it is possible that the MAA units become sequestered in the inner core of a supramolecular assembly (possibly micelles) which is stabilized by the PEG chains.
  • Chain aggregation leads to the formation of a highly viscous internal core and, thus, to a partial suppression of the signals of EA and MAA units. Supporting these results, analysis by DLS revealed, at pH 3, the presence of a colloid population (215 ⁇ 50 nm), that was absent at pH 10.

Abstract

The present invention is directed toward water-soluble supramolecular self-assemblies and a process for their preparation via micellization of polyelectrolytes through the use of hydrophobic monomeric units. In this invention the polyelectrolyte segment ultimately forms the core of the supramolecular assembly whereas the shell consists of uncharged hydrophilic polymers or oligomers. It has been determined that the inclusion of the hydrophobic co-monomers to the polyelectrolyte segment forming the micelle core leads to a structure of enhanced stability.

Description

    REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of application Ser. No. 09/877,999, filed Jun. 8, 2001, the contents of which are herein incorporated by reference.
  • FIELD OF THE INVENTION
  • This invention generally relates to the preparation of water-soluble self-assemblies, particularly to the association of ionizable or permanently-charged copolymers, and most particularly to the inclusion of hydrophobic comonomers to the polyelectrolyte segment forming the assembly core.
  • BACKGROUND OF THE INVENTION
  • In order to improve the specific delivery of drugs with a low therapeutic index, several drug carriers such as liposomes, microparticles, nano-associates (e.g. polymeric micelles, polyion complex micelles (PICM)) and drug-polymer conjugates have been studied. In recent years, water-soluble supramolecular assemblies such as polymeric micelles and PICM have emerged as promising new colloidal carriers for the delivery of hydrophobic drugs and polyions (e.g. antisense oligonucleotides), respectively.
  • Polymeric micelles have been the object of growing scientific attention, and have emerged as potential carriers for drugs having poor water solubility because they can solubilize those drugs in their inner core and they offer attractive characteristics such as a generally small size (<300 nm) and a propensity to evade scavenging by the mononuclear phagocyte system.
  • Micelles are often compared to naturally occurring carriers such as viruses or lipoproteins. All three of these carriers demonstrate a similar core-shell structure that allows for their contents to be protected during transportation to the target cell, whether it is DNA for viruses or water-insoluble drugs for lipoproteins and micelles.
  • Polymeric micelles seem to be one of the most advantageous carriers for the delivery of poorly water-soluble drugs as reported by Jones and Leroux, Eur. J. Pharm. Biopharm. (1999) 48, 101-111; Kwon and Okano, Adv. Drug Deliv. Rev. (1996) 21, 107-116 and Allen et al. Colloids Surf. B: Biointerf. (1999) 16, 3-27. They are characterized by a core-shell structure. The hydrophobic inner core generally serves as a microenvironment for the solubilization of poorly water-soluble drugs, whereas the hydrophilic outer shell is responsible for micelle stability, protection against opsonization, and uptake by the mononuclear phagocyte system.
  • Pharmaceutical research on polymeric micelles has been mainly focused on copolymers having an AB diblock structure with A, the hydrophilic shell moieties and B the hydrophobic core polymers, respectively. Multiblock copolymers such as poly(ethylene oxide)-poly(propylene oxide)-poly(ethylene oxide) (PEO-PPO-PEO) (A-B-A) can also self-organize into micelles, and have been described as potential drug carriers, e.g. Kabanov et al., FEBS Lett. (1989) 258, 343-345. The hydrophobic core which generally consists of a biodegradable polymer such as a poly(β-benzyl-aspartate) (PBLA), poly(D,L-lactic acid) or poly(□-caprolactone), serves as a reservoir for a poorly water-soluble drug, protecting it from contact with the aqueous environment. The core may also consist of a water-soluble polymer, such as poly(aspartic acid) (P(Asp)), which is rendered hydrophobic by the chemical conjugation of a hydrophobic drug, or is formed through the association of two oppositely charged polyions (PICM). Several studies also describe the use of poorly or non-biodegradable polymers, such as polystyrene (PSt) or poly(methyl methacrylate)(PMMA), as constituents of the inner core. See, e.g., Zhao et al., Langmuir (1990) 6, 514-516; Zhang et al., Science (1995) 268, 1728-1731; Inoue et al., J. Controlled Release (1998) 51, 221-229 and Kataoka J. Macromol. Sci. Pure Appl. Chem. (1994) A31, 1759-1769. The hydrophobic inner core can also consist of a highly hydrophobic small chain such as an alkyl chain or a diacyllipid (e.g. distearoyl phosphatidyl ethanolamine). The hydrophobic chain can be either attached to one end of a polymer, or randomly distributed within the polymeric structure. The shell usually consists of chains of hydrophilic, non-biodegradable, biocompatible polymers such as poly(ethylene oxide) (PEO) (see Allen et al. Colloids Surf. B: Biointerf. (1999) 16, 3-27 and Kataoka et al. J. Controlled Release (2000) 64, 143-153), poly(N-vinyl-2-pyrrolidone) (PVP) (see Benahmed A et al. Pharm Res (2001) 18, 323-328) or poly(2-ethyl-2-oxazoline) (see Lee et al. Macromolecules (1999) 32, 1847-1852).
  • The biodistribution of the carrier is mainly dictated by the nature of the hydrophilic shell. Other polymers such as poly(N-isopropylacrylamide) and poly(alkylacrylic acid) impart temperature or pH sensitivity to the micelles, and could eventually be used to confer bioadhesive properties (see U.S. Pat. No. 5,770,627). Micelles presenting functional groups at their surface for conjugation with a targeting moiety have also been described (See, e.g., Scholz, C. et al., Macromolecules (1995) 28, 7295-7297).
  • At the present time, most polymeric micelles described in the literature are prepared using non-ionizable block polymers or block copolymers where ionizable monomers are used to form the micelle corona whereas the core consists of a hydrophobic neutral homopolymer or copolymer. Ionizable diblock copolymers have been shown to exhibit pH-dependent micellization. Recently, Webber and Martin (U.S. Pat. No. 5,955,509) have described a type of pH-dependent micelles with a polyelectrolyte core. These micelles are composed of the diblock copolymers poly(vinyl N-heterocycle)-block-poly(alkylene oxide). Such copolymers are positively charged at acidic pH due to the protonation of the nitrogen atoms, and thus exist as unimers in acidic solutions. At high pH, the unprotonated copolymers self-associate into polymeric micelles. These micelles are primarily intended to deliver their contents at low pH, since the dissociation of the supramolecular assembly under acidic conditions allows a drug to be released. Such conditions may be found, for example in tumors. If intended to be administered by the oral route, these micelles would rapidly release their contents in the stomach because of its acidic pH. Therefore, for oral delivery, they should be formulated using an enteric coating to prevent premature drug leakage.
  • A potential problem with ionizable copolymers is the possibility of forming, at acidic pH, intra and inter-molecular hydrogen bonding between the protonated and the non-ionizable hydrophilic blocks which might lead to the formation of an insoluble complex. This has been recently described by Lele et al. J. Controlled Release (2000) 69, 237-248, between poly(acrylic acid) and poly(ethylene glycol). Precipitation of the micelles at acidic pH could potentially compromise the efficacy of the system when oral delivery is envisaged.
  • PICM have a block or graft copolymer architecture and consist of a polyelectrolyte linked to a non-ionic water-soluble polymer. They bind with charged compounds due to electrostatic interactions with the polyelectrolyte (see, e.g., Kataoka et al. Macromolecules (1996) 29, 8556-8557). The complexes self-assemble into micelle-like structures which have a hydrophobic core from neutralized polyelectrolyte and counterion, and hydrophilic corona. PICM show improved solubility compared with other electrostatic complexes. Furthermore, they show reduced affinity for plasma components and can protect active compounds such as DNA against enzymatic degradation.
  • Although, PICM hold great promise as carriers for a variety of selected from the group consisting of ionizable and permanently-charged diblock copolymers, ionizable and permanently-charged multiblock copolymers, and ionizable and permanently-charged random copolymers with grafted hydrophilic and essentially non-ionic oligomers or polymers compounds, such as charged drugs and nucleic acids, some important issues still remain to be addressed. For instance, the stability of the polymeric micelles is influenced by various factors such as concentration, temperature and chemical structure of the polymer. In particular, the presence of salts is a key parameter for the dissociation of PICM since Coulombic interactions between charged segments are screened by the added salt. To overcome this problem, polymeric micelles can be stabilized by cross-linking the core or shell (see, e.g., Kakizawa et al. J. Am. Chem. Soc. (1999) 121, 11247-11248). However, cross-linking the core or shell can potentially chemically alter the active agent and/or excessively slow down its release from the micelles.
  • DESCRIPTION OF THE PRIOR ART
  • U.S. Pat. No. 5,693,751 teaches the preparation of polymeric micelles composed of a water-soluble block copolymer having a hydrophilic segment and a hydrophobic pharmacological-functioning segment to a side chain of which a drug is covalently bonded.
  • U.S. Pat. No. 5,702,717 teaches the preparation of a solution comprising a drug and a block amphiphilic copolymer of poly(ethylene glycol) and poly(□-hydroxy acids) or poly(ethylene carbonates). These copolymers are not polyelectrolytes.
  • U.S. Pat. No. 5,939,453 teaches the preparation of polymeric micelles and bioerodible drug delivery matrix using poly(ethylene glycol)-poly(orthoester) diblock and triblock copolymers. The principal novelty of this invention relies on the use of poly(orthoester) as the hydrophobic, bioerodible segment. These block copolymers are neither ionizable or permanently charged.
  • U.S. Pat. No. 5,786,387 teaches the preparation of supramolecular assemblies using lipid double chain derivatives containing poly(oxyethylene) for drug delivery applications. These block copolymers are neither ionizable or permanently charged. They efficiently are able to avoid the reticuloendothelial system and possess a long circulation time.
  • U.S. Pat. No. 5,840,319 teaches the preparation of polymeric micelles using polyether block copolymers having a critical micelle concentration of no more than 0.5% (w/w) at 37° C. in an isotonic aqueous solution. The formulation also contains a chemotherapeutic agent.
  • U.S. Pat. No. 5,770,627 teaches the preparation of hydrophobically-modified bioadhesive polyelectrolytes. These polyelectrolytes can form micellar structures in aqueous solutions (example 6) and be loaded with an active agent. The polyelectrolyte can be a graft or block copolymer. The outer shell is ionizable since it contains carboxylic acid groups whereas the inner core consists of a homopolymer, copolymer, unsaturated or saturated alkyl chains, or other hydrophobic moities. Methods of administering such agents to an animal are disclosed.
  • U.S. Pat. No. 5,955,509 relates to the use of poly(vinyl N-heterocycle)-block-poly(alkylene oxide) copolymers in micelle containing pharmaceutical formulations. The copolymers advantageously respond to pH differences in the environment to which they are exposed forming micelles at higher pH values. The micelles, which comprise a therapeutic compound and a copolymer, deliver drug in a pH dependent manner.
  • U.S. Pat. No. 5,929,177 provides a block polymer which has functional groups on both ends thereof, and which comprises hydrophilic/hydrophobic segments. As for the functional groups on its both ends, the block polymer has amino group, carboxyl group or mercapto group on the alpha terminal, and hydroxyl group, carboxyl group, aldehyde group or vinyl group on the omega terminal. Hydrophilic segment comprises polyethylene oxide, while hydrophobic segment is derived from lactide, lactone or (meth)acrylic acid ester. The block polymer of this invention forms a polymeric micelle which is usable as bio-compatible materials.
  • U.S. Pat. No. 5,925,720 provides a heterotelechelic oligomer or polymer which can be prepared by means of living polymerization and which can form stable micelles in an aqueous solvent. In this invention, there is no reference to stabilization of a polyelectrolyte micelle core.
  • U.S. Pat. No. 5,656,611 relates to compositions for stabilizing polynucleic acids using polyionic complexes. In one aspect the invention provides a polynucleotide complex between a polynucleotide and a block copolymer comprising a polyether block and a polycation block.
  • U.S. Pat. No. 6,217,912 provides a biodegradable composition suitable for delivering a gene into a cell.
  • U.S. Pat. No. 6,221,959 provides composition for stabilizing polynucleic acids and increasing the ability of polynucleic acid to cross cell membranes and act in the interior of a cell. In one aspect the invention provides a polynucleotide complex between a polynucleotide and certain polyether block copolymer.
  • U.S. Pat. No. 5,510,103 relates to drug carriers composed of a block copolymer having hydrophilic and hydrophobic segments, a polymeric type drug comprising hydrophobic drugs trapped by physical treatments in said drug carrier and methods for trapping hydrophobic drugs in the carrier.
  • What is lacking in the art are water-soluble supramolecular assemblies having a physically-stabilized polyelectrolyte core and an uncharged hydrophilic shell and techniques for their preparation.
  • SUMMARY OF THE INVENTION
  • The present invention is directed toward water-soluble supramolecular self-assemblies and a process for their preparation via micellization of polyelectrolytes through the use of hydrophobic monomeric units. In this invention the polyelectrolyte segment ultimately forms the core of the supramolecular assembly whereas the shell consists of uncharged hydrophilic polymers or oligomers. It has been determined that the inclusion of the hydrophobic co-monomers to the polyelectrolyte segment forming the micelle core leads to a structure of enhanced stability. Such co-monomers, by increasing the attractive forces between the segments of the core, stabilize the micelles and/or decrease the interaction between the ionizable or permanently-charged segment and non-ionizable segment of the copolymer.
  • Accordingly, it is an objective of the instant invention to provide a stabilized supramolecular assembly and a process for its production.
  • It is a further objective of the instant invention to provide, also through the use of hydrophobic monomers, pH-dependent polymeric micelles or polyion complexes which exhibit reduced interaction between the ionizable or permanently-charged segment and the non ionizable segments of the copolymer outer shell.
  • It is yet another objective of the instant invention to provide a pH dependent micellar vehicle, suitable as a carrier for pharmacological constituents which is not subject to the untoward formation of insoluble complexes.
  • It is a still further objective of the invention to teach a process for preparing stabilized supramolecular assemblies having a polyelectrolyte core, through the use of hydrophobic monomeric units.
  • Other objects and advantages of this invention will become apparent from the following description taken in conjunction with the accompanying drawings wherein are set forth, by way of illustration and example, certain embodiments of this invention. The drawings constitute a part of this specification and include exemplary embodiments of the present invention and illustrate various objects and features thereof.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 is a synthetic route for the preparation of block copolymer, possessing a hydrophobized polyelectrolyte block;
  • FIG. 2 is the 1H NMR spectrum of PEG-b-P(DMAEMA30-co-EMA70) in CDCl3;
  • FIG. 3 shows the variation in light scattering and pyrene fluorescence I338/I333 ratio of a PEG-b-P(EA50-co-MAA50) aqueous solution as a function of pH;
  • FIG. 4 shows the 1H NMR spectrum of PEG-b-P(EA50-co-MAA50) in D2O at pH 10 (A) and at pH 3 (B).
  • DETAILED DESCRIPTION OF THE INVENTION
  • In the present invention, the terms “water-soluble self-assemblies” and “micelles” are equally employed although the proposed structures may not necessarily correspond to the true definition of micelles.
  • Micelle formation occurs as a result of two forces. One is an attractive force that leads to the association of molecules, while the other is a repulsive force that prevents unlimited growth of the micelles to a distinct macroscopic phase. Amphiphilic copolymers self-associate when placed in a solvent that is selective for either the hydrophilic or hydrophobic polymer.
  • The micellization process of amphiphilic copolymers is similar to that for low molecular weight surfactants. At very low concentrations, the polymers exist only as single chains. As the concentration increases to reach a critical value called the critical association concentration (“CAC”), polymer chains start to associate to form micelles in such a way that the hydrophobic part of the copolymer will avoid contact with the aqueous media in which the polymer is diluted. Amphiphilic copolymers usually exhibit a CAC which is much lower than that of low molecular weight surfactants. For example, the CAC of PEO-PBLA and PNIPA-PSt are between 0.0005-0.002%. Some amphiphilic copolymers, however, exhibit much higher CAC, reaching up to 0.01-10% in the case of poloxamers. Amphiphilic copolymers with high CAC may not be suitable as drug targeting devices since they are unstable in an aqueous environment and are easily dissociated upon dilution.
  • Micelles can be targeted to specific cells or tissues via the inclusion of targeting ligands, e.g. monoclonal antibodies, lectins, sugars, vitamins, peptides or immunologically distinct fragments thereof or the like moieties which provide the micelles with an ability to preferentially concentrate in a particular target area.
  • The micellization of amphiphilic copolymers can result in two different types of micelles depending on whether the hydrophobic chain is randomly bound to the hydrophilic polymer or grafted to one end of the hydrophilic chain. Micelles formed from randomly modified polymers are generally smaller than end-modified polymers. The micellar size is mainly determined by the hydrophobic forces which sequester the hydrophobic chains in the core, and by the excluded volume repulsion between the chains which limits their size. The difference in the balance of these two forces in random and end-modified copolymers may account for their different size.
  • Determination of Critical Association Concentration (CAC):
  • Light scattering is widely used for the determination of the molecular weight and aggregation number of micelles. The onset of micellization can, however, be detected only if the CAC falls within the sensitivity of the scattering method. This is rarely the case for polymers in water. Gel permeation chromatography (GPC) under aqueous conditions can be employed since single chains and micellar fractions of copolymers exhibit different elution volumes. It is also possible to simultaneously determine by GPC the molecular weight of the micelles and their aggregation number.
  • A preferred method to determine the CAC involves the use of fluorescent probes, among which pyrene is widely used. Pyrene is a condensed aromatic hydrocarbon that is highly hydrophobic and sensitive to the polarity of the surrounding environment. Below the CAC, pyrene is solubilized in water, a medium of high polarity. When micelles are formed, pyrene partitions preferentially toward the hydrophobic domain afforded by the micellar core, and thus experiences a nonpolar environment. Consequently, numerous changes such as an increase in the fluorescence intensity, a change in the vibrational fine structure of the emission spectra, and a red shift of the (0,0) band in the excitation spectra are observed. The apparent CAC can be obtained from the plot of the fluorescence of pyrene, the I1/I3 ratio from emission spectra or the I338/I333 ratio from the excitation spectra versus concentration. A major change in the slope indicates the onset of micellization. Changes in anisotropy of fluorescent probes have also been associated with the onset of micellization. E.g. see Jones and Leroux Eur. J. Pharm. Biopharm. (1999) 48, 101-111.
  • Polymeric micelles such as those of the compositions of the invention are characterized by their small size. Besides being needed for extravasation of the carrier materials, this small size permits the sterilization of the composition to be effected simply by filtration, and minimizes the risks of embolism in capillaries after intravenous injection. Micellar size depends on several factors including copolymer molecular weight, relative proportion of hydrophilic and hydrophobic chains and aggregation number.
  • Micellar diameter and size polydispersity can be obtained directly in water or in an isotonic buffer by dynamic light scattering (DLS). Micelle size can also be estimated by methods such as atomic force microscopy (AFM), transmission electron microscopy (TEM) and scanning electron microscopy (SEM). These methods allow the characterization of the micelle shape and size dispersity. Ultracentrifugation velocity studies are sometimes performed to assess the polydispersity of polymeric micelles.
  • Loading of one or more pharmacological constituents, e.g. various therapeutic agents, drugs, peptides, proteins, genetic material (e.g. oligonucleotides), genetically altered constituents, polyionic constituents and the like, into the micelles can be realized according to techniques well known to one skilled in the art. For example, drugs can be incorporated into the polymeric micelle compositions of the invention by means of chemical conjugation or by physical entrapment through dialysis, emulsification techniques, simple equilibration of the drug and micelles in an aqueous medium or solubilization of a drug/polymer solid dispersion in water.
  • Therapeutic agents which may be used are any compounds, including the ones listed below, which can be entrapped, in a stable manner, in polymeric micelles and administered at a therapeutically effective dose. Preferably, the therapeutic agents used in accordance with the invention are hydrophobic or polyionic (e.g. DNA). Suitable drugs include antitumor compounds such as phthalocyanines (e.g. aluminum chloride phthalocyanine), anthracyclines (e.g. doxorubicin), poorly soluble antimetabolites (e.g. methotrexate, mitomycin, 5-fluorouracil) and alkylating agents (e.g. carmustine). Micelles may also contain taxanes such as paclitaxel.
  • Additional drugs which can be contained in micelles are conventional hydrophobic antibiotics and antifungal agents such as amphotericin B and itraconazole, poorly water-soluble immunomodulators such as cyclosporin, poorly water-soluble antiviral drugs such as HIV protease inhibitors and poorly water-soluble steroidal (e.g. dexamethasone), and non-steroidal (e.g. indomethacin) anti-inflammatory drugs.
  • Hydrophilic compounds such as proteins may also be incorporated in the polymeric micelle compositions of the invention. The incorporation of such hydrophilic species may, however, require the chemical hydrophobization of the molecule or a particular affinity for the hydrophilic shell. Polyionic compounds (e.g. antisense oligonucleotides, genome fragments, peptides) can be incorporated into micelles through the formation of PICM via electrostatic interaction with an oppositely charged block polyelectrolyte.
  • The polymeric micelle compositions of the invention are suitable for use in a variety of pharmaceutical fields, such as oral delivery, sustained release and site-specific drug targeting. Preferably, the micelles of the invention are used as a transport for water-insoluble and polyionic compounds. Included within the scope of the invention are supramolecular assembly compositions comprising a suitable targeting ligand.
  • Without intending to be limited to a particular synthesis procedure, block polyelectrolytes useful in the present invention are most preferably prepared by “living”/controlled radical polymerization (LCRP), such as atom transfer radical polymerization (ATRP) (see Coessens et al., Prog. Polym. Sci. (2001) 26, 337-377) or nitroxide-mediated radical polymerization (NMP) (see Benoit et al. J. Am. Chem. Soc. (1999), 121, 3904-3920). However, any alternative procedure such as other living radical polymerizations or condensation of preformed functionalized polymers could also be used. On the other hand, (i) ionizable and permanently-charged multiblock copolymers, (ii) ionizable and permanently-charged amphiphilic random copolymers with grafted hydrophilic oligomers (or polymers) could be used instead of block copolymers for diverse applications within the scope of the presently disclosed invention.
  • The radical initiator for the synthesis of the polymer by ATRP can be any appropriately functionalized molecule (e.g. poly(ethylene glycol) (PEG), PVP). The initiator bears an halogeno functionality that can be activated for ATRP (see Coessens et al., Prog. Polym. Sci. (2001) 26, 337-377). Without intending to be limited to any particular substituent, this functionality can be a 2-halogenoisobutyrylate derivative, 2-halogenopropionate derivative, 2-halogenoacetate derivative or 1-(halogenomethyl)benzene derivative. For other types of polymerizations (e.g. NMP), this functionality is appropriately chosen according to the used monomers.
  • The catalyst for the ATRP usually includes a metallic salt and a ligand. The ligand is used for the solubilization of the salt in organic solvent and/or to activate the redox reaction of the metal present in the salt. The salt activates the radical initiator for the ATRP. Without intending to be limited to any particular salt, the latter can be copper(I) bromide, copper(I) chloride or copper(I) thiocyanate, iron(II) and nickel(0 or I) compounds. The ligand can include 2,2′-bipyridine derivatives or bis(dimethylamino) compounds (e.g. N,N,N′,N′,N″,N″-pentamethyldiethylene-triamine (PMDETA).
  • Suitable polyelectrolyte compounds useful in the preparation of supramolecular self-assemblies may be selected from the group consisting of diblock copolymers including ionizable units, permanently charged units or mixtures of ionizable and permanently charged units, multiblock copolymers including ionizable units, permanently charged units or mixtures of ionizable and permanently charged units, and random copolymers with grafted hydrophilic and essentially non-ionic oligomers or polymers, said random copolymers including ionizable units, permanently charged units or mixtures of ionizable and permanently charged units.
  • The diblock copolymers generally consist of two blocks, one of which is hydrophilic and generally uncharged and the other containing at least one compound selected from the group consisting of ionizable and permanently-charged repeating units (or combinations thereof) in combination with essentially hydrophobic, e.g. hydrophobic or relatively hydrophobic non-ionic monomers. Ionizable units refers to repeating units that can be transformed from a non-ionic to a charged state via an external stimulus (e.g. pH or chemical reaction). Permanently charged units refers to repeating units that are in fact electrostatically charged irrespective of the external conditions.
  • The ionizable and/or permanently-charged block, bearing hydrophobic repeating units, can be synthesized from vinyl monomers, vinyl oligomers or eventually vinyl polymers. These hydrophobic monomers/oligomers/polymers can be acrylate, acrylamide, alkylacrylate, alkylacrylamide, arylacrylate and arylacrylamide derivatives for which the alkyl and aryl term stands for aliphatic or aromatic moieties respectively (e.g. methacrylate, methacrylamide derivatives.) The hydrophobic compound can also be a biodegradable polyester such as vinyl-terminated poly(lactide) and vinyl-terminated poly(□-caprolactone). The ionizable monomers could be alkylacrylic acid derivatives, (aminoalkyl)acrylate or (aminoalkyl)alkylacrylate derivatives. The acidic or basic units of the polymer chain can be derived from a non-ionizable precursor (e.g. tert-butylmethacrylate), which is cleaved into an acidic moiety.
  • The hydrophilic block can be synthesized from vinyl monomers, vinyl oligomers or eventually vinyl polymers. These hydrophilic monomers/oligomers/polymers can be acrylate, acrylamide, alkylacrylate and alkylacrylamide (e.g. PEG methacrylate and N-(2-hydroxypropyl)acrylamide). On the other hand, the hydrophilic block can also originate from a block radical macroinitiator based on PEG or PVP derivatives.
  • Non biodegradable ionizable and/or permanently-charged copolymers that are intended to be administered parenterally, should have a molecular weights not exceeding 40,000. There is no restriction on molecular weights for biodegradable or non-biodegradable ionizable copolymers, which are used orally or locally.
  • The loading of poorly water-soluble and non-ionic drugs should be done in an organic solvent, or in aqueous solutions at a pH where the core is uncharged or mostly uncharged. Charged drugs should be loaded under conditions (e.g. pH) where electrostatic interactions with the ionizable or permanently-charged segment are possible.
  • EXAMPLES
  • Abbreviations:
  • The subscript text indicates the ratio in a polymeric segment. The letter b features that polymers and/or polymeric arms are based on a diblock copolymeric structure. The term co means the repeating units are disposed randomly along the polymeric segment.
  • Example 1 Synthesis of Poly(Ethylene Glycol)-Block-Poly(N,N-Dimethylaminoethanemethacrylate-Co-Ethylmethacrylate) with a Ratio for DMAEMA/EMA of 30/70
  • PEG-b-P(DMAEMA30-co-EMA70)
  • Materials:
  • All products were purchased from Aldrich (Milwaukee, Wis.). Copper(I) bromide (99.99% Grade), 2-bromoisobutyryl bromide, anhydrous triethylamine and N,N,N′,N′,N″,N″-pentamethyldiethylenetriamine (PMDETA) were used without further purification. Poly(ethylene glycol) monomethyl ether (MeO-PEG-OH, Mn: 2000) was dried with toluene by an azeotropic distillation before use. Ethyl methacrylate (EMA) and 2-(N,N-dimethylamino)ethyl methacrylate (DMAEMA) were used as vinyl monomers and distilled before polymerization. Prior to use, tetrahydrofuran (THF) was distilled over sodium, using benzophenone as drying indicator.
  • Synthesis of PEG ATRP macroinitiator (α-(2-bromoisobutyrylate)-ω-methylPEG)
  • As exemplified in FIG. 1, to a solution of MeO-PEG-OH (Mn: 2000, 10 g, 0.005 mol) and triethylamine (1.0 g, 0.01 mol) in 70 mL of anhydrous THF, slightly cooled in a water-ice bath, was slowly added 2-bromoisobutyryl bromide (4.3 mL, 0.035 mol). The solution was then warmed to room temperature and stirred for 24 h. The mixture was poured into water and extracted with methylene chloride. The organic extracts were washed successively with a HCl 1M and NaOH 1M solution (containing NaCl), and dried over magnesium sulfate. The solvent was removed under reduced pressure. The crude was dissolved in a minimum of methylene chloride, and then precipitated in diethyl ether. The title compound was recovered by simple filtration. Yield: 70% after precipitation. White solid. M.P. 60-65° C.
  • 1H NMR (δ, ppm, CDCl3): 4.18 (2H); 3.50 (188H); 3.23 (3H, s); 1.80 (6H, s).
  • ATRP:
  • The ATRP of monomers was carried out in bulk and in solution, using α-(2-bromoisobutyrylate)-ω-methyl-PEG as ATRP macroinitiator. The PEG ATRP macroinitiator (1 eq.) was added to a solution containing PMDETA (1.1 eq.), Cu(I)Br (1.1 eq.), EMA (14 eq) and DMAEMA (6 eq) in THF (0.8 M). The mixture was degassed with argon for 15-20 min at room temperature and was then heated to 60° C. overnight. After the polymerization, the mixture was poured in THF, containing 10% of methanol. The resulting polymers were filtered on silica gel, with THF as eluent, to remove copper bromide. Finally, polymers were dialyzed (SPECTRA/POR No. 1, molecular weight cutoff 6000-8000) against water during 48 h and then freeze-dried. Yield: 98%. (FIG. 1)
  • Poly(ethylene glycol)-block-poly(N,N-dimethylaminoethanemethacrylate-co-ethylmethacry-late)
  • PEG-b-P(DMAEMA30-co-EMA70)
  • 1H NMR (δ, ppm, CDCl3): 4.30 (18H); 4.04 (32H); 3.60 (182H); 3.38 (3H); 2.69 (54H), 2.05-1.87 (42H); 1.43 (6H); 1.26 (56H); 1.05 and 0.88 (73H).
  • Polymer and Micelle Characterization:
  • 1H and 13C NMR spectra were recorded on a Bruker AMX300 and ARX400 in deuterated chloroform (CDCl3) and methanol (CD3OD) (CDN Isotopes, Canada) at 25° C. Number- (Mn) and weight-average (Mw) molecular weights were determined by size exclusion chromatography (SEC) with an Alliance GPCV2000 (Waters, Milford, Mass.) and by nuclear magnetic resonance spectroscopy (1H-NMR). Particle sizes were evaluated by dynamic light-scattering. The apparent CAC was measured by a steady-state pyrene fluorescence method.
  • Results:
  • Copper (I) bromide-pentamethyldiethylenetriamine (CuBr-PMDETA) was used as catalyst and gave yields of polymerization approaching 100% in THF. The reactivities of EMA and DMAEMA in THF were similar, with a kapp of 1.95×10−4 Lmol−1s−1. All monomers were completely consumed after 5 h and the Mn obtained experimentally were close to the theoretical values (Table 1). Moreover, the polydispersity index (PI) was approximately 1.4 and corresponded approximately to the polydispersity of PEG macroinitiator used for the preparation of PEG-b-P(DMAEMA30-co-EMA70)
    TABLE 1
    Mn and Mw of P (DMAEMA30-co-EMA70).
    Mn Mn Mn
    Copolymers Theo NMR SEC PI
    PEG-b-P(DMAEMA30-co-EMA70) 4690 4900 4700 1.4
  • FIG. 2 shows the 1H NMR spectrum of PEG-b-P(DMAEMA30-co-EMA70). The terminal methoxy group of PEG (signal 1, 3.35 ppm), was used as a reference to calibrate the integration of the other signals. The integration of signal 2 showed that the degree of polymerization (DP) of ethylene oxide (PEG chains) was approximately 45 in each copolymer and corresponded to the DP of the commercial PEG used. The narrow signal of the PEG ATRP macroinitiator at 1.80 ppm, assigned to the methyl of the bromoisobutyryl group, decreased rapidly at the beginning of the polymerization and shifted within 10 min to about 1.4 ppm (signal 3), confirming that all PEG chains were coupled to the polyvinyl segments. The signals assigned to the methylene groups in the backbone were observed at 1.8 ppm (signals 4 and 4′) and are represented by 3 successive peaks of decreasing intensity. The polymethacrylate derivatives presented two signals assigned to the methyl attached to the backbone (signals 5 and 5′). The ratios were determined using the signals 7, 8 and 9.
  • The CAC of PEG-b-P(DMAEMA30-co-EMA70) was determined in water and phosphate buffered saline (PBS, pH 7.4) to verify whether salts could interfere with self-assembling. A remarkable fact was that the PEG-b-P(DMAEMA30-co-EMA70) showed a low CAC in water and PBS, despite the presence of DMAEMA which is ionized in these aqueous solutions (Table 2). This could be explained by the presence of EMA in the polymer. Interestingly, the CAC was not significantly affected by the presence of salts in water. This is an important issue, since a common drawback about PICM is their relative poor stability in physiological media. Accordingly, the stability of PICM can be easily increased by introducing a hydrophobic comonomer in the polymer backbone. With regard to the micelle sizes for PEG-b-P(DMAEMA30-co-EMA70) the nature of the aqueous solution (water vs PBS) seemed to influence the proportion of secondary aggregates (Table 2).
    TABLE 2
    Micellar properties of PEG-b-P(DMAEMA30-co-EMA70)
    DP Micelle
    of the CAC size Size
    polyvinyl (mg/L) ± (nm) Peak
    Copolyer block 10% SD ± 25% amount
    PEG-b-P 20 2 392 79%
    (DMAEMA30-co-EMA70)
    In water  22 21%
    PEG-b-P 20 3 280 24%
    (DMAEMA30-co-EMA70)
    In PBS  79 76%
  • Example 2
  • Synthesis of a Diblock Copolymer Containing Methacrylic Acid Units as Ionizable Units
    • PEG-b-P(EA50-co-tBMA50) (precursor)
    • PEG-b-P(EA50-co-MAA50)
      Materials, Synthesis of PEG-ATRP Macroinitiator and ATRP:
  • Carried out as described in Example 1. However, the only difference was 5 eq. of EMA and 5 eq. of tert-butyl methacrylate (tBMA) versus 1 eq. of PEG-ATRP macroinitiator were used for the polymerization of PEG-b-P(EA50-co-tBMA50). In the case of PEG-b-P(MAA), only the tBMA monomer (18 eq.) was used. (FIG. 1)
  • Transformation of tBMA into MAA:
  • The ester groups, bearing a tert-butyl chain (tBMA), were transformed into carboxylic acid groups, by the cleavage of tert-butyl in acidic conditions. To a solution of the polymers having tBMA units (7.7 mmol) in dioxane (2.6 M) was added concentrated HCl (32 mmol) for 5 h. The methacrylic acid derivatives were precipitated in diethyl ether and filtered. The polymers were dissolved in ethanol, dialyzed against water and freeze-dried.
  • Polymer and Micelle Characterization:
  • 1H and 13C NMR spectra were recorded on a Bruker AMX300 and ARX400 in CDCl3, in CD3OD and in D2O (CDN Isotopes) at different pH, using very small amounts of HCl and NaOH. Mn, and Mw, were determined by SEC with an Alliance GPVC2000 (Waters, Milford, Mass.) and by NMR spectroscopy. Hydrodynamic mean diameter and size distribution were determined at a 90° angle by DLS using differential size distribution processor intensity analysis (N4Plus, Coulter Electronics, Miami, Fla.). The apparent CAC was measured by a steady-state pyrene fluorescence method. The pH of self-association was determined by static light scattering at 480 nm, using a Series 2 Aminco fluorimeter. The pH of association was also determined by the steady-state pyrene fluorescence method.
  • Poly(ethylene glycol)-block-poly(tert-butyl methacrylate) PEG-b-P (tBMA); Mn: 4560; Using tBMA as monomer
  • 1H NMR (δ, ppm, CDCl3): 4.10 (36H); 3.64 (181H); 3.37 (3H, s); 2.02-1.80 (36H); 1.44 (6H, s); 1.40 (162H, s); 1.13 (18H, s); 1.02 (36H, s)
  • Poly(ethylene glycol)-block-poly(methacrylic acid) PEG-b-P (MAA); Mn: 3180; Obtained from PEG-b-P (tBMA) after the cleavage of tert-butyl groups
  • 1H NMR (δ, ppm, MeOD): 4.19 (36H); 3.66 (186H); 3,38 (3H); 2.02 (24H); 1.57 (2H); 1.16 (43H).
  • Poly(ethylene glycol)-block-poly(ethyl acrylate-co-tert-butyl methacrylate); PEG-b-P(EA50-co-tBMA50); Mn: 3600; Using tBMA and EMA as monomers
  • 1H NMR (δ, ppm, CDCl3): 4.10 (12H); 3.66 (181H); 3.39 (3H); 2.10-1.70 (22H); 1.44 (54H); 1.28 (18H); 1.00 (22H).
  • Poly(ethylene glycol)-block-poly(ethyl acrylate-co-methacrylic acid); PEG-b-P(EA50-co-MAA50); Mn: 3400; Obtained from PEG-b-P(EMA50-co-tBMA50) after the cleavage of tert-butyl groups:
  • 1H NMR (δ, ppm, MeOD): 4.10 (12H); 3.63 (181H); 3.36 (3H); 2.20-1.70 (22H); 1.65 (6H); 1.26 (18H); 1.14 (22H).
  • Results:
  • Well-defined acidic (ionizable) copolymers, containing or not hydrophobic units were prepared by ATRP using a PEGylated ATRP macroinitiator. This macroinitiator was synthesized by coupling PEG monomethyl ether to 2-bromoisobutyryl bromide with a high yield. Copper (I) bromide-pentamethyldiethylenetriamine (CuBr-PMDETA) was used as catalyst and gave yields of polymerization approaching 100% in THF. The Mn estimated by NMR were calculated from the terminal methoxy group of the PEG chain (˜3.4 ppm). All monomers were completely consumed after 5 h and the Mn obtained experimentally were close to the theoretical values. For all copolymers, the polydispersity was within the range of about 1.3 to about 1.5 (Table 3).
    TABLE 3
    Molecular weights of precursors of P(MAA)
    diblock copolymer derivatives
    Mn Mn
    Mn Mn SEC SEC
    Copolymer Theo NMR relative universal Mw/Mn
    PEG ATRP macroinitiator 2150 2250 2100 2200 1.3
    PEG-b-P(tBMA) 4460 4560 3600 3900 1.5
    PEG-b-P(EA50-co-tBMA50) 3360 3600 3700 4000 1.4
  • The PEG-b-P(EA50-co-tBMA50) and PEG-b-P(tBMA) precursors were transformed into their respective P(MAA) derivatives by cleaving the tert-butyl groups in presence of hydrochloric acid in dioxane. The monomer ratios of the prepared copolyvinyl polymers corresponded approximately to the proportions of monomers used for the polymerization. The hydrophobic unit EA was incorporated in the polyvinyl segment, to increase hydrophobicity. At high pH values, the copolymers are fully ionized and should be in solution mostly as individual polymeric chains. As the pH is decreased, the protonation of the carboxylic groups should increase the hydrophobic character of the copolymer and induce chain aggregation. The pH of interchain association was determined at body temperature (37° C.) by static light scattering and by spectrofluorimetry, using pyrene as a probe (Table 2). FIG. 3 shows the ratio of fluorescence intensities (I338/I333) versus pH, and the first-order derivative of scattered light as a function of pH for PEG-b-P(EA50-co-MAA50). The polymeric chains associated at a pH value close to 6, as determined by both techniques (FIG. 3).
  • Interestingly, PEG-b-P(EA50-co-MAA50) remained soluble at acidic pH, indicating that the protonation of MAA units did not make the diblock copolymers precipitate. In the case of PEG-b-P(MAA), a diffuse precipitate appeared when the pH was decreased to around pH 3.5-4. It is known that acrylic acid polymeric derivatives interact through hydrogen bonding with PEG in acidic solutions, resulting in precipitation of the polymers. Without intending to be bound to any specific mechanism, we believe that in the presence of the hydrophobic ethyl acrylate comonomer the attractive forces between the protonated MAA and ethylene oxide units are sterically hindered. Accordingly, since PEG-b-P(EA50-co-MAA50) remains soluble at acidic pHs, it is possible that the MAA units become sequestered in the inner core of a supramolecular assembly (possibly micelles) which is stabilized by the PEG chains.
  • To confirm the pH-dependent associative behavior of PEG-b-P(EA-co-MAA), its 1H NMR spectra were recorded in D2O at pH 10 and 3 (FIG. 4). At pH 10 (FIG. 4A), the copolymer demonstrated all peaks proper to the PEG and the ionized parts, presenting integrations in accordance with the molecular weight (Mn). This suggests that all polymer chains were isolated from each other in water at high pH. However, at low pH (˜3), the decreased peaks assigned to P(EA-co-MAA) segments indicated the presence of chain aggregation (FIG. 4B). Chain aggregation (into polymeric micelles) leads to the formation of a highly viscous internal core and, thus, to a partial suppression of the signals of EA and MAA units. Supporting these results, analysis by DLS revealed, at pH 3, the presence of a colloid population (215±50 nm), that was absent at pH 10.
    TABLE 4
    Determination of the aggregation pH of polymethacrylic acid
    derivatives
    Aggregation
    Aggregation pH
    pH determined Number of
    determined by by light MAA per
    Copolymer fluorimetry scattering chains*
    PEG-b-P(MAA) 5.6 4.9 18
    PEG-b-P(EA50-co-MAA50) 5.8 6.0  5

    *Number of MAA units in the polyvinyl segment, evaluated by 1H NMR spectroscopy from the corresponding copolymers having tert-butyl groups.
  • All patents and publications mentioned in this specification are indicative of the levels of those skilled in the art to which the invention pertains. All patents and publications are herein incorporated by reference to the same extent as if each individual publication was specifically and individually indicated to be incorporated by reference.
  • It is to be understood that while a certain form of the invention is illustrated, it is not to be limited to the specific form or arrangement of parts herein described and shown. It will be apparent to those skilled in the art that various changes may be made without departing from the scope of the invention and the invention is not to be considered limited to what is shown and described in the specification and drawings.
  • One skilled in the art will readily appreciate that the present invention is well adapted to carry out the objects and obtain the ends and advantages mentioned, as well as those inherent therein. The compounds, compositions, biologically related compounds, methods, procedures and techniques described herein are presently representative of the preferred embodiments, are intended to be exemplary and are not intended as limitations on the scope. Changes therein and other uses will occur to those skilled in the art which are encompassed within the spirit of the invention and are defined by the scope of the appended claims. Although the invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention which are obvious to those skilled in the art are intended to be within the scope of the following claims.

Claims (16)

1. Supra molecular self-assemblies of a polyelectrolyte for use in combination with a pharmacological ingredient, comprising:
at least one polyelectrolyte compound selected from the group consisting of diblock copolymers, multiblock copolymers and graft copolymers,
said diblock and multiblock copolymers including at least two blocks one of which is hydrophilic and essentially uncharged, and another of which contains at least one repeating unit selected from the group consisting of anionizable or permanently-negatively charged repeating units in combination with essentially hydrophobic repeating units,
said graft copolymers comprising grafted hydrophilic and essentially non-ionic oligomers or polymers, and including anionizable units, permanently-negatively charged units or mixtures of anionizable and permanently-negatively charged units, in combinaison with essentially hydrophobic repeating units,
wherein a polyelectrolyte segment forms a core of the assembly and chemically bears non-ionic hydrophobic repeating units.
2. The supramolecular self-assemblies of claim 1, wherein:
said anionizable units are repeating units that can be transformed from a non-ionic to a negatively charged state via an external stimulus.
3. The supramolecular self-assemblies of claim 2, wherein:
said external stimulus is selected from the group consisting of a change in pH, ionic strength, temperature or other physico-chemical changes.
4. The supramolecular self-assemblies of claim 1, wherein:
said anionizable or permanently-negatively charged block, bearing hydrophobic repeating units, is synthesized from at least one hydrophobic compound selected from the group consisting of vinyl monomers, vinyl oligomers, and vinyl polymers.
5. The supramolecular self-assemblies of claim 4, wherein:
said hydrophobic compound is at least one compound selected from the group consisting of acrylate, acrylamide, alkylacrylate, alkylacrylamide, arylacrylate and arylacrylamide derivatives.
6. The supramolecular self-assemblies of claim 5, wherein:
said alkyl or aryl derivatives include at least one aliphatic or aromatic moiety selected from the group consisting of acrylate, acrylamide, methacrylate and methacrylamide derivatives.
7. The supramolecular self-assemblies of claim 4, wherein:
said hydrophobic compound is at least one vinyl-terminated biodegradable polyester.
8. The supramolecular self-assemblies of claim 7, wherein:
said vinyl terminated biodegradable polyester is selected from the group consisting of vinyl-terminated poly(lactide) and vinyl-terminated poly ε-caprolactone).
9. The supramolecular self-assemblies of claim 1, wherein:
said anionizable units include at least one functional group selected from carboxylate, phosphate and sulfate.
10. The supramolecular self-assemblies of claim 1, wherein:
said hydrophilic block is synthesized from at least one hydrophilic compound selected from vinyl monomers, vinyl oligomers and vinyl polymers.
11. The supramolecular self-assemblies of claim 10, wherein:
said hydrophilic compound is at least one compound selected from the group consisting of acrylate derivatives, acrylamide derivatives, alkylacrylate derivatives, alkylacrylamide, and N-vinyl-2-pyrrolidone derivatives.
12. The supramolecular self-assemblies of claim 1, wherein:
said hydrophilic block originates from a macroinitiator based on poly (ethylene glycol) or poly (N-vinyl-2-pyrrolidone).
13. A pharmaceutical formulation comprising supramolecular self-assemblies of claim 1 in combination with a effective amount of at least one pharmacological constituent.
14. The pharmaceutical formulation of claim 13, wherein the pharmacological constituent is a drug.
15. The pharmaceutical formulation of claim 13, wherein the pharmacological constituent is a peptide, protein or genetic material.
16. The pharmaceutical formulation of claim 13, including a targeting ligand.
US11/175,796 2001-06-08 2005-07-05 Water-soluble stabilized self-assembled polyelectrolytes Abandoned US20050244501A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US11/175,796 US20050244501A1 (en) 2001-06-08 2005-07-05 Water-soluble stabilized self-assembled polyelectrolytes
US11/558,543 US7510731B2 (en) 2001-06-08 2006-11-10 Water-soluble stabilized self-assembled polyelectrolytes

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US09/877,999 US6939564B2 (en) 2001-06-08 2001-06-08 Water-soluble stabilized self-assembled polyelectrolytes
US11/175,796 US20050244501A1 (en) 2001-06-08 2005-07-05 Water-soluble stabilized self-assembled polyelectrolytes

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/877,999 Continuation US6939564B2 (en) 2001-06-08 2001-06-08 Water-soluble stabilized self-assembled polyelectrolytes

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/558,543 Continuation US7510731B2 (en) 2001-06-08 2006-11-10 Water-soluble stabilized self-assembled polyelectrolytes

Publications (1)

Publication Number Publication Date
US20050244501A1 true US20050244501A1 (en) 2005-11-03

Family

ID=25371168

Family Applications (3)

Application Number Title Priority Date Filing Date
US09/877,999 Expired - Lifetime US6939564B2 (en) 2001-06-08 2001-06-08 Water-soluble stabilized self-assembled polyelectrolytes
US11/175,796 Abandoned US20050244501A1 (en) 2001-06-08 2005-07-05 Water-soluble stabilized self-assembled polyelectrolytes
US11/558,543 Expired - Lifetime US7510731B2 (en) 2001-06-08 2006-11-10 Water-soluble stabilized self-assembled polyelectrolytes

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/877,999 Expired - Lifetime US6939564B2 (en) 2001-06-08 2001-06-08 Water-soluble stabilized self-assembled polyelectrolytes

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/558,543 Expired - Lifetime US7510731B2 (en) 2001-06-08 2006-11-10 Water-soluble stabilized self-assembled polyelectrolytes

Country Status (13)

Country Link
US (3) US6939564B2 (en)
EP (1) EP1465665B1 (en)
JP (1) JP4302509B2 (en)
AT (1) ATE313335T1 (en)
AU (1) AU2002315598B2 (en)
BR (1) BR0210284A (en)
CA (1) CA2452806C (en)
DE (1) DE60208237T2 (en)
DK (1) DK1465665T3 (en)
ES (1) ES2257556T3 (en)
MX (1) MXPA03011296A (en)
PT (1) PT1465665E (en)
WO (1) WO2002100439A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040197360A1 (en) * 2001-08-27 2004-10-07 Kazunori Kataoka Composition containing fine particles for supporting biologically active substance thereon or having the same supported thereon and method for preparing these
US20080081075A1 (en) * 2006-10-02 2008-04-03 National Tsing Hua University Multifunctional mixed micelle of graft and block copolymers and preparation thereof

Families Citing this family (71)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6780428B2 (en) * 2001-06-08 2004-08-24 Labopharm, Inc. Unimolecular polymeric micelles with an ionizable inner core
US6939564B2 (en) * 2001-06-08 2005-09-06 Labopharm, Inc. Water-soluble stabilized self-assembled polyelectrolytes
US7094810B2 (en) * 2001-06-08 2006-08-22 Labopharm, Inc. pH-sensitive block copolymers for pharmaceutical compositions
EP1465933B1 (en) 2002-01-16 2007-08-29 Biocompatibles UK Limited Polymer conjugates
US8105652B2 (en) * 2002-10-24 2012-01-31 Massachusetts Institute Of Technology Methods of making decomposable thin films of polyelectrolytes and uses thereof
GB0301014D0 (en) * 2003-01-16 2003-02-19 Biocompatibles Ltd Conjugation reactions
US7217776B1 (en) * 2003-02-14 2007-05-15 Iowa State University Research Foundation pH-sensitive methacrylic copolymer gels and the production thereof
US7807722B2 (en) * 2003-11-26 2010-10-05 Advanced Cardiovascular Systems, Inc. Biobeneficial coating compositions and methods of making and using thereof
US20070293539A1 (en) * 2004-03-18 2007-12-20 Lansbury Peter T Methods for the treatment of synucleinopathies
JP2007529555A (en) * 2004-03-18 2007-10-25 ザ ブライハム アンド ウイメンズ ホスピタル, インコーポレイテッド How to treat synucleinopathy
EP1744751A4 (en) * 2004-03-18 2010-03-10 Brigham & Womens Hospital Methods for the treatment of synucleinopathies
US20060106060A1 (en) * 2004-03-18 2006-05-18 The Brigham And Women's Hospital, Inc. Methods for the treatment of synucleinopathies (Lansbury)
US20060198891A1 (en) 2004-11-29 2006-09-07 Francois Ravenelle Solid formulations of liquid biologically active agents
US7727554B2 (en) 2004-12-21 2010-06-01 Board Of Regents Of The University Of Nebraska By And Behalf Of The University Of Nebraska Medical Center Sustained-release nanoparticle compositions and methods for using the same
TW200624464A (en) * 2004-12-31 2006-07-16 Ind Tech Res Inst Amphiphilic block copolymer and pharmaceutical formulation comprising the same
US7534756B2 (en) * 2005-02-25 2009-05-19 Solutions Biomed, Llc Devices, systems, and methods for dispensing disinfectant solutions comprising a peroxygen and transition metal
US7504369B2 (en) * 2005-02-25 2009-03-17 Solutions Biomed, Llc Methods and compositions for decontaminating surfaces exposed to chemical and/or biological warfare compounds
US7553805B2 (en) * 2005-02-25 2009-06-30 Solutions Biomed, Llc Methods and compositions for treating viral, fungal, and bacterial infections
CA2599010C (en) 2005-02-25 2014-02-04 Solutions Biomed, Llc Aqueous disinfectants and sterilants
US7507701B2 (en) * 2005-02-25 2009-03-24 Solutions Biomed, Llc Aqueous disinfectants and sterilants including transition metals
US7473675B2 (en) * 2005-02-25 2009-01-06 Solutions Biomed, Llc Disinfectant systems and methods comprising a peracid, alcohol, and transition metal
US7462590B2 (en) 2005-02-25 2008-12-09 Solutions Biomed, Llc Aqueous disinfectants and sterilants comprising a peroxide/peracid/transition metal mixture
US7511007B2 (en) * 2005-02-25 2009-03-31 Solutions Biomed, Llc Aqueous sanitizers, disinfectants, and/or sterilants with low peroxygen content
KR100626767B1 (en) * 2005-07-07 2006-09-25 한국과학기술연구원 Chain-end functionalized poly(ethylene oxide) and method for the preparation of nano-sized transition metals and metal salts using the same
KR101306150B1 (en) * 2005-10-24 2013-09-10 삼성전자주식회사 Polymer for top coating layer, top coating solution compositions and immersion lithography process using the same
US9738745B2 (en) 2006-06-29 2017-08-22 Life Technologies As Particles containing multi-block polymers
CA2699184A1 (en) * 2006-09-22 2008-03-27 Labopharm Inc. Compositions and methods for ph targeted drug delivery
JP5340956B2 (en) * 2006-12-20 2013-11-13 アーケマ・インコーポレイテッド Encapsulation and / or binding of polymers
WO2008157372A2 (en) 2007-06-14 2008-12-24 Massachusetts Institute Of Technology Self assembled films for protein and drug delivery applications
US8464910B2 (en) * 2008-03-14 2013-06-18 Solutions Biomed, Llc Multi-chamber container system for storing and mixing fluids
US20110129921A1 (en) * 2008-05-13 2011-06-02 University Of Washington Targeted polymer bioconjugates
AU2009246329B8 (en) 2008-05-13 2013-12-05 Phaserx, Inc. Micellic assemblies
CN102083878B (en) 2008-05-13 2013-07-10 华盛顿大学 Diblock copolymers and polynucleotide complexes thereof for delivery into cells
BRPI0911988A2 (en) * 2008-05-13 2015-10-13 Phaserx Inc micelles for intracellular release of therapeutic agents.
KR20110026424A (en) * 2008-05-13 2011-03-15 유니버시티 오브 워싱톤 Polymeric carrier
NZ589287A (en) * 2008-05-16 2012-06-29 Univ Sydney Administrable compositions
CA2635187A1 (en) 2008-06-05 2009-12-05 The Royal Institution For The Advancement Of Learning/Mcgill University Oligonucleotide duplexes and uses thereof
WO2010021973A2 (en) * 2008-08-17 2010-02-25 Massachusetts Institute Of Technology Controlled delivery of bioactive agents from decomposable films
US9211250B2 (en) * 2008-08-22 2015-12-15 University Of Washington Heterogeneous polymeric micelles for intracellular delivery
KR20110095292A (en) 2008-11-06 2011-08-24 유니버시티 오브 워싱톤 Multiblock copolymers
US8822213B2 (en) 2008-11-06 2014-09-02 University Of Washington Bispecific intracellular delivery vehicles
US8716339B2 (en) 2008-11-12 2014-05-06 Solutions Biomed, Llc Two-part disinfectant system and related methods
US8789716B2 (en) * 2008-11-12 2014-07-29 Solutions Biomed, Llc Multi-chamber container system for storing and mixing liquids
US20100120913A1 (en) * 2008-11-12 2010-05-13 Larson Brian G Resin catalyzed and stabilized peracid compositions and associated methods
CA2745926A1 (en) 2008-12-08 2010-07-08 Phaserx, Inc. Omega-functionalized polymers, junction-functionalized block copolymers, polymer bioconjugates, and radical chain extension polymerization
CN101701068B (en) * 2009-10-30 2011-09-21 北京化工大学 Y-type drug delivery material with pH responsiveness and preparation method thereof
US9415113B2 (en) 2009-11-18 2016-08-16 University Of Washington Targeting monomers and polymers having targeting blocks
US9474269B2 (en) 2010-03-29 2016-10-25 The Clorox Company Aqueous compositions comprising associative polyelectrolyte complexes (PEC)
US9309435B2 (en) 2010-03-29 2016-04-12 The Clorox Company Precursor polyelectrolyte complexes compositions comprising oxidants
US20110236582A1 (en) 2010-03-29 2011-09-29 Scheuing David R Polyelectrolyte Complexes
EP2425817A1 (en) * 2010-08-09 2012-03-07 Commissariat à l'Énergie Atomique et aux Énergies Alternatives Polymerized micelles for diagnosis
WO2012039741A1 (en) * 2010-09-22 2012-03-29 The Board Of Regents Of The University Of Texas System Novel block copolymer and micelle compositions and methods of use thereof
CN102070756B (en) * 2010-12-10 2012-03-28 北京化工大学 Sugar-responsive medicament delivery material and preparation method thereof
EP2841056A4 (en) 2012-04-23 2015-09-16 Massachusetts Inst Technology Stable layer-by-layer coated particles
WO2014134029A1 (en) 2013-02-26 2014-09-04 Massachusetts Institute Of Technology Nucleic acid particles, methods and use thereof
US9463244B2 (en) 2013-03-15 2016-10-11 Massachusetts Institute Of Technology Compositions and methods for nucleic acid delivery
US9867885B2 (en) 2013-07-30 2018-01-16 Phaserx, Inc. Block copolymers
US9084735B2 (en) 2013-08-01 2015-07-21 International Business Machines Corporation Self-assembling bis-urea compounds for drug delivery
NL2013317B1 (en) * 2014-08-11 2016-09-21 Univ Utrecht Holding Bv Amphiphilic block copolymers for delivery of active agents.
US8975220B1 (en) 2014-08-11 2015-03-10 The Clorox Company Hypohalite compositions comprising a cationic polymer
FR3030530B1 (en) * 2014-12-23 2017-01-27 Arkema France COPOLYMER DIBLOC WATER SOLUBLE
FR3030529B1 (en) 2014-12-23 2017-01-27 Arkema France WATER-SOLUBLE BLOCK COPOLYMER AND USE THEREOF AS A MATERIAL SUPPORT FOR 3D PRINTING
CN107530436B (en) 2015-01-21 2022-03-29 菲泽尔克斯公司 Methods, compositions and systems for delivering therapeutic and diagnostic agents into cells
US10792477B2 (en) 2016-02-08 2020-10-06 Orbusneich Medical Pte. Ltd. Drug eluting balloon
EP3496732A4 (en) 2016-08-12 2020-05-06 Purdue Research Foundation Polymer lung surfactants
KR102071912B1 (en) * 2016-10-31 2020-01-31 주식회사 엘지화학 Polymer Electolyte, an Electrochromic Device Comprising the Same and Method for Preparing thereof
EP3562510A4 (en) 2016-12-30 2021-01-06 Genevant Sciences GmbH Branched peg molecules and related compositions and methods
US11419947B2 (en) 2017-10-30 2022-08-23 Massachusetts Institute Of Technology Layer-by-layer nanoparticles for cytokine therapy in cancer treatment
CN107857846B (en) * 2017-11-23 2019-10-11 常州大学 A kind of preparation method of polyethylene glycol grafting polybutyl methacrylate amphipathic graft copolymer
US11786464B2 (en) 2020-04-24 2023-10-17 The Board Of Regents Of The University Of Texas System PH responsive block copolymer compositions and micelles that inhibit MCT 1 and related proteins
CN114163591B (en) * 2021-11-29 2023-08-01 南方医科大学 Block copolymer, block copolymer drug-loaded micelle and preparation method and application thereof

Citations (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5510103A (en) * 1992-08-14 1996-04-23 Research Development Corporation Of Japan Physical trapping type polymeric micelle drug preparation
US5620850A (en) * 1994-09-26 1997-04-15 President And Fellows Of Harvard College Molecular recognition at surfaces derivatized with self-assembled monolayers
US5656611A (en) * 1994-11-18 1997-08-12 Supratek Pharma Inc. Polynucleotide compositions
US5693751A (en) * 1989-05-11 1997-12-02 Research Development Corporation Of Japan Water soluble high molecular weight polymerized drug preparation
US5702717A (en) * 1995-10-25 1997-12-30 Macromed, Inc. Thermosensitive biodegradable polymers based on poly(ether-ester)block copolymers
US5770627A (en) * 1995-08-16 1998-06-23 University Of Washington Hydrophobically-modified bioadhesive polyelectrolytes and methods relating thereto
US5786387A (en) * 1994-03-23 1998-07-28 Meiji Seika Kabushiki Kaisha Lipid double-chain derivative containing polyoxyethylene
US5840319A (en) * 1992-10-08 1998-11-24 Alakhov; Valery Yu Biological agent compositions
US5908777A (en) * 1995-06-23 1999-06-01 University Of Pittsburgh Lipidic vector for nucleic acid delivery
US5925720A (en) * 1995-04-19 1999-07-20 Kazunori Kataoka Heterotelechelic block copolymers and process for producing the same
US5929177A (en) * 1995-08-10 1999-07-27 Kazunori Kataoka Block polymer having functional groups at both ends
US5939453A (en) * 1998-06-04 1999-08-17 Advanced Polymer Systems, Inc. PEG-POE, PEG-POE-PEG, and POE-PEG-POE block copolymers
US5955509A (en) * 1996-05-01 1999-09-21 Board Of Regents, The University Of Texas System pH dependent polymer micelles
US6217912B1 (en) * 1998-07-13 2001-04-17 Expression Genetics, Inc. Polyester analogue of poly-L-lysine as a soluble, biodegradable gene delivery carrier
US6221959B1 (en) * 1994-11-18 2001-04-24 Supratek Pharma, Inc. Polynucleotide compositions
US6372203B1 (en) * 1999-04-30 2002-04-16 Wella Aktiengesellschaft Hair treatment compositions with polymers made from unsaturated saccharides, unsaturated saccharic acids or their derivatives
US20020187199A1 (en) * 2001-06-08 2002-12-12 Maxime Ranger Unimolecular polymeric micelles with an ionizable inner core
US6939564B2 (en) * 2001-06-08 2005-09-06 Labopharm, Inc. Water-soluble stabilized self-assembled polyelectrolytes

Family Cites Families (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4016332A (en) 1972-05-01 1977-04-05 Ppg Industries, Inc. Mercaptan blocked thermosetting copolymers
US3933940A (en) 1973-02-08 1976-01-20 Ppg Industries, Inc. Mercaptan blocked thermosetting copolymers
GB1411843A (en) 1973-02-28 1975-10-29 Univ Strathclyde Method of modifying the properties of a polymer
GB1575343A (en) 1977-05-10 1980-09-17 Ici Ltd Method for preparing liposome compositions containing biologically active compounds
DE3001013A1 (en) 1980-01-12 1981-07-23 Basf Ag, 6700 Ludwigshafen VINYLPYRROLIDONE POLYMERISATES, THEIR PRODUCTION AND THEIR USE FOR THE PRODUCTION OF BLOOD REPLACEMENT LIQUIDS
JPS5767860A (en) 1980-10-15 1982-04-24 Fuji Photo Film Co Ltd Material for multilayer analysis
ATE37983T1 (en) 1982-04-22 1988-11-15 Ici Plc DELAYED RELEASE AGENT.
US4565854A (en) 1983-04-07 1986-01-21 Kuraray Co., Ltd. Polymer having thiol end group
US4826689A (en) 1984-05-21 1989-05-02 University Of Rochester Method for making uniformly sized particles from water-insoluble organic compounds
GB8416234D0 (en) 1984-06-26 1984-08-01 Ici Plc Biodegradable amphipathic copolymers
US6312679B1 (en) 1986-08-18 2001-11-06 The Dow Chemical Company Dense star polymer conjugates as dyes
US5019400A (en) 1989-05-01 1991-05-28 Enzytech, Inc. Very low temperature casting of controlled release microspheres
US5041516A (en) 1989-06-21 1991-08-20 Cornell Research Foundation, Inc. Dendritic molecules and method of production
US5206410A (en) 1989-08-31 1993-04-27 University Of South Florida Multifunctional synthons as used in the preparation of cascade polymers or unimolecular micelles
US5154853A (en) 1991-02-19 1992-10-13 University Of South Florida Unimolecular micelles and method of making the same
FR2678168B1 (en) 1991-06-28 1993-09-03 Rhone Poulenc Rorer Sa NANOPARTICLES HAVING CAPTURE TIME BY THE EXTENDED RETICULO ENDOTHELIAL DYSTEM.
US5543158A (en) 1993-07-23 1996-08-06 Massachusetts Institute Of Technology Biodegradable injectable nanoparticles
EP0639592A1 (en) 1993-08-18 1995-02-22 Nippon Shokubai Co., Ltd. Water-soluble graft polymers and production process thereof
ES2104518T1 (en) 1994-03-07 1997-10-16 Dow Chemical Co CONJUGATES BIOACTIVE DENDRIMEROS AND / OR DIRECTORS TO DIANA.
NL9401886A (en) 1994-05-27 1996-01-02 Dsm Nv Composition consisting of a dendrimer and an active substance contained in the dendrimer, a method of preparing such a composition and a method of releasing the active substance.
US5863919A (en) 1994-07-25 1999-01-26 University Of South Florida Lock and key micelles and monomer building blocks therefor
US5997861A (en) 1994-10-31 1999-12-07 Burstein Laboratories, Inc. Antiviral supramolecules containing target-binding molecules and therapeutic molecules bound to spectrin
WO1996021036A2 (en) 1994-12-30 1996-07-11 Chiron Viagene, Inc. Nucleic acid condensing agents with reduced immunogenicity
US5492996A (en) 1995-02-21 1996-02-20 The United States Of America As Represented By The Secretary Of The Air Force Alcohol soluble benzazole polymers
US6143211A (en) 1995-07-21 2000-11-07 Brown University Foundation Process for preparing microparticles through phase inversion phenomena
JPH11510837A (en) 1995-07-28 1999-09-21 フォーカル,インコーポレイテッド Multi-block biodegradable hydrogels for use as controlled release and tissue treatment agents for drug delivery
WO1997010814A1 (en) 1995-09-18 1997-03-27 Vesifact Ag Propofol nanodispersions
KR0180334B1 (en) 1995-09-21 1999-03-20 김윤 Drug messenger using el-2l-2 micelle and method for sealing drug to it
KR100413029B1 (en) 1995-09-29 2004-04-30 가가쿠 기쥬츠 신코 지교단 New anthracycline compound derivatives and medicinal preparations containing the same
ES2166987T3 (en) 1996-03-15 2002-05-01 Harvard College METHOD OF FORMING ARTICLES AND MODELED SURFACES THROUGH CAPILLARY MICROMOLDING.
CA2258851A1 (en) 1996-06-27 1997-12-31 G.D. Searle & Co. Particles comprising amphiphilic copolymers, having a cross-linked shell domain and an interior core domain, useful for pharmaceutical and other applications
US6284267B1 (en) 1996-08-14 2001-09-04 Nutrimed Biotech Amphiphilic materials and liposome formulations thereof
US6060518A (en) 1996-08-16 2000-05-09 Supratek Pharma Inc. Polymer compositions for chemotherapy and methods of treatment using the same
TW520297B (en) 1996-10-11 2003-02-11 Sequus Pharm Inc Fusogenic liposome composition and method
GB9623051D0 (en) 1996-11-06 1997-01-08 Schacht Etienne H Delivery of DNA to target cells in biological systems
WO1998037902A1 (en) 1997-02-28 1998-09-03 The Regents Of The University Of California Inhibition of cell-cell binding by lipid assemblies
EP0963758A3 (en) 1998-05-07 2000-03-22 Universiteit Gent Synthetic polyaminoacid complexes for delivery of nucleic acids to target cells
IN191203B (en) 1999-02-17 2003-10-04 Amarnath Prof Maitra
JP3523821B2 (en) 2000-02-09 2004-04-26 ナノキャリア株式会社 Method for producing polymer micelle in which drug is encapsulated and polymer micelle composition
DE10008895A1 (en) 2000-02-25 2001-08-30 Beiersdorf Ag Stabilization of active substances that are sensitive to oxidation and / or UV
GB2359747B (en) 2000-02-29 2002-04-24 Maelor Pharmaceuticals Ltd Anaesthetic formulations
EP1286643A2 (en) 2000-05-17 2003-03-05 Labopharm Inc. Drug containing polymeric micelles
US6338859B1 (en) 2000-06-29 2002-01-15 Labopharm Inc. Polymeric micelle compositions
IN188917B (en) 2000-12-07 2002-11-23 Bharat Surums & Vaccines Ltd
JP2004532277A (en) 2001-06-08 2004-10-21 パウダージェクト ワクチンズ,インコーポレーテッド Spray freeze-dried composition
US7094810B2 (en) * 2001-06-08 2006-08-22 Labopharm, Inc. pH-sensitive block copolymers for pharmaceutical compositions
US20060003012A9 (en) 2001-09-26 2006-01-05 Sean Brynjelsen Preparation of submicron solid particle suspensions by sonication of multiphase systems
US6756449B2 (en) 2002-02-27 2004-06-29 Medtronic, Inc. AnB block copolymers containing poly (vinyl pyrrolidone) units, medical devices, and methods
US6780324B2 (en) 2002-03-18 2004-08-24 Labopharm, Inc. Preparation of sterile stabilized nanodispersions
US7018655B2 (en) 2002-03-18 2006-03-28 Labopharm, Inc. Amphiphilic diblock, triblock and star-block copolymers and their pharmaceutical compositions
EP1555984B1 (en) 2002-10-21 2008-03-12 L'oreal Process for dissolving lipophilic compounds, and cosmetic composition.
US7262253B2 (en) 2003-12-02 2007-08-28 Labopharm, Inc. Process for the preparation of amphiphilic poly (N-vinyl-2-pyrrolidone) block copolymers
US20060198891A1 (en) 2004-11-29 2006-09-07 Francois Ravenelle Solid formulations of liquid biologically active agents

Patent Citations (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5693751A (en) * 1989-05-11 1997-12-02 Research Development Corporation Of Japan Water soluble high molecular weight polymerized drug preparation
US5510103A (en) * 1992-08-14 1996-04-23 Research Development Corporation Of Japan Physical trapping type polymeric micelle drug preparation
US5840319A (en) * 1992-10-08 1998-11-24 Alakhov; Valery Yu Biological agent compositions
US5786387A (en) * 1994-03-23 1998-07-28 Meiji Seika Kabushiki Kaisha Lipid double-chain derivative containing polyoxyethylene
US5620850A (en) * 1994-09-26 1997-04-15 President And Fellows Of Harvard College Molecular recognition at surfaces derivatized with self-assembled monolayers
US6221959B1 (en) * 1994-11-18 2001-04-24 Supratek Pharma, Inc. Polynucleotide compositions
US5656611A (en) * 1994-11-18 1997-08-12 Supratek Pharma Inc. Polynucleotide compositions
US6440743B1 (en) * 1994-11-18 2002-08-27 Supratek Pharma Inc. Methods of using polynucleotide compositions
US5925720A (en) * 1995-04-19 1999-07-20 Kazunori Kataoka Heterotelechelic block copolymers and process for producing the same
US5908777A (en) * 1995-06-23 1999-06-01 University Of Pittsburgh Lipidic vector for nucleic acid delivery
US5929177A (en) * 1995-08-10 1999-07-27 Kazunori Kataoka Block polymer having functional groups at both ends
US5770627A (en) * 1995-08-16 1998-06-23 University Of Washington Hydrophobically-modified bioadhesive polyelectrolytes and methods relating thereto
US5702717A (en) * 1995-10-25 1997-12-30 Macromed, Inc. Thermosensitive biodegradable polymers based on poly(ether-ester)block copolymers
US5955509A (en) * 1996-05-01 1999-09-21 Board Of Regents, The University Of Texas System pH dependent polymer micelles
US5939453A (en) * 1998-06-04 1999-08-17 Advanced Polymer Systems, Inc. PEG-POE, PEG-POE-PEG, and POE-PEG-POE block copolymers
US6217912B1 (en) * 1998-07-13 2001-04-17 Expression Genetics, Inc. Polyester analogue of poly-L-lysine as a soluble, biodegradable gene delivery carrier
US6372203B1 (en) * 1999-04-30 2002-04-16 Wella Aktiengesellschaft Hair treatment compositions with polymers made from unsaturated saccharides, unsaturated saccharic acids or their derivatives
US20020187199A1 (en) * 2001-06-08 2002-12-12 Maxime Ranger Unimolecular polymeric micelles with an ionizable inner core
US6939564B2 (en) * 2001-06-08 2005-09-06 Labopharm, Inc. Water-soluble stabilized self-assembled polyelectrolytes

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040197360A1 (en) * 2001-08-27 2004-10-07 Kazunori Kataoka Composition containing fine particles for supporting biologically active substance thereon or having the same supported thereon and method for preparing these
US7897176B2 (en) 2001-08-27 2011-03-01 Toudai Tlo, Ltd. Composition containing fine particles for supporting biologically active substance thereon or having the same supported thereon and method for preparing these
US20080081075A1 (en) * 2006-10-02 2008-04-03 National Tsing Hua University Multifunctional mixed micelle of graft and block copolymers and preparation thereof

Also Published As

Publication number Publication date
DE60208237T2 (en) 2007-01-11
JP4302509B2 (en) 2009-07-29
PT1465665E (en) 2006-05-31
BR0210284A (en) 2004-07-20
AU2002315598B2 (en) 2006-08-24
CA2452806A1 (en) 2002-12-19
WO2002100439A1 (en) 2002-12-19
ES2257556T3 (en) 2006-08-01
ATE313335T1 (en) 2006-01-15
DK1465665T3 (en) 2006-05-08
DE60208237D1 (en) 2006-01-26
CA2452806C (en) 2011-12-13
US20070110709A1 (en) 2007-05-17
US6939564B2 (en) 2005-09-06
US7510731B2 (en) 2009-03-31
EP1465665A1 (en) 2004-10-13
MXPA03011296A (en) 2004-10-28
JP2004534879A (en) 2004-11-18
US20030059398A1 (en) 2003-03-27
EP1465665B1 (en) 2005-12-21

Similar Documents

Publication Publication Date Title
US7510731B2 (en) Water-soluble stabilized self-assembled polyelectrolytes
AU2002315598A1 (en) Water-soluble stabilized self-assembled polyelectrolytes
CA2414241C (en) Polymeric micelle compositions
US6780428B2 (en) Unimolecular polymeric micelles with an ionizable inner core
US7018655B2 (en) Amphiphilic diblock, triblock and star-block copolymers and their pharmaceutical compositions
US7094810B2 (en) pH-sensitive block copolymers for pharmaceutical compositions
Dufresne et al. Study of the micellization behavior of different order amino block copolymers with heparin
AU2001270405A1 (en) Polymeric micelle compositions
US20050158271A1 (en) Pharmaceutical applications of hydrotropic polymer micelles
AU2003301690B2 (en) Tri-block polymers for nanosphere-based drug or gene delivery
Kosakowska et al. Star-Shaped Amphiphilic Polymers as Soluble Carriers for Drug Delivery
US20060182752A1 (en) Tri-block polymers for nanosphere-based drug or gene delivery

Legal Events

Date Code Title Description
AS Assignment

Owner name: LABOPHARM INC., CANADA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:RANGER, MAXIME;LEROUX, JEAN-CHRISTOPHE;REEL/FRAME:017420/0297

Effective date: 20010605

STCB Information on status: application discontinuation

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