WO2010143972A2 - Particules sensibles à l'énergie pour l'administration de médicament comprenant des lipides formant une structure non lamellaire - Google Patents

Particules sensibles à l'énergie pour l'administration de médicament comprenant des lipides formant une structure non lamellaire Download PDF

Info

Publication number
WO2010143972A2
WO2010143972A2 PCT/NO2010/000218 NO2010000218W WO2010143972A2 WO 2010143972 A2 WO2010143972 A2 WO 2010143972A2 NO 2010000218 W NO2010000218 W NO 2010000218W WO 2010143972 A2 WO2010143972 A2 WO 2010143972A2
Authority
WO
WIPO (PCT)
Prior art keywords
liposomes
lipid
dspe
mol
anyone
Prior art date
Application number
PCT/NO2010/000218
Other languages
English (en)
Other versions
WO2010143972A3 (fr
Inventor
Cecilia Leal Lauten
Karen Sibylla RØGNVALDSSON
Sigrid L. Fossheim
Esben A. Nilssen
Tove J. Evjen
Original Assignee
Epitarget As
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 Epitarget As filed Critical Epitarget As
Publication of WO2010143972A2 publication Critical patent/WO2010143972A2/fr
Publication of WO2010143972A3 publication Critical patent/WO2010143972A3/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0002Galenical forms characterised by the drug release technique; Application systems commanded by energy
    • A61K9/0009Galenical forms characterised by the drug release technique; Application systems commanded by energy involving or responsive to electricity, magnetism or acoustic waves; Galenical aspects of sonophoresis, iontophoresis, electroporation or electroosmosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • A61K9/1272Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers with substantial amounts of non-phosphatidyl, i.e. non-acylglycerophosphate, surfactants as bilayer-forming substances, e.g. cationic lipids

Definitions

  • Acoustically sensitive drug delivery particles comprising non-lamellar forming lipids
  • the present invention is related to particles comprising non-lamellar forming amphiphilic lipids for controlled drug delivery and release at a defined volume in an animal.
  • the invention relates to acoustically sensitive drug carrying particles, e.g. liposomes, as well as compositions, methods and uses thereof.
  • Ultrasound has been suggested as a method to trigger specific drug release (Pitt, Husseini et al. 2004). This may allow the engineering of robust particles protecting healthy tissue while in circulation, accumulating in the diseased volume and releasing the payload on exposure to acoustic energy. Also, US is known to increase cell permeability thus providing a twofold effect: drug carrier disruption and increased intracellular drug uptake (Larina, Evers et al. 2005; Larina, Evers et al. 2005).
  • micelles are non-covalently self-assembled particles typically formed by molecules containing one part that is water-soluble and one that is fat soluble.
  • the monomer aqueous solubility is typically in the mM range and at a critical concentration; micelles are formed shielding the fat soluble part from the aqueous phase. Micelle formation and disruption is therefore an equilibrium process controlled by concentration, making these particles rather unstable and less suitable for drug delivery.
  • limited drug types can be encapsulated.
  • Gas- filled liposomes and microbubbles are highly US responsive but too large (-1 ⁇ m) for efficient accumulation in e.g. tumour tissue.
  • liposomes or other lipid dispersions may encapsulate a broad range of water soluble and fat soluble drugs, as well as efficiently accumulate in e.g. tumour tissue.
  • reports on ultrasound sensitive liposomes are scarce.
  • Long-chain alcohols may also be incorporated in phospholipid bilayers.
  • the alcohol has one part with affinity for water (hydroxyl group) and another with affinity for oily or lipidic environments (hydrocarbon moiety).
  • hydrocarbon moiety When added to a liposome dispersion some alcohol molecules remain in the aqueous phase, whilst others are incorporated in the phospholipid membrane.
  • the extent of incorporation depends on the alcohol chain length. The longer the chain length, the more molecules will be captured within the membrane (Aagaard, Kristensen et al. 2006). The fact that organic alcohols can penetrate membranes also has an implication on local and general anaesthesia in animals (Lee 1976).
  • the effect of alcohols on the liposomal membrane properties is remarkably different depending on the alcohol chain length.
  • the membrane can be made "thinner” by inclusion of short chain alcohols (Rowe and Campion 1994; Tierney, Block et al. 2005) and the gel-to-liquid crystalline phase transition temperature of the membrane lowered by the addition of decanol (Thewalt and Cushley 1987).
  • decanol Thewalt and Cushley 1987.
  • octanol which has a shorter chain is even more efficient to lower the phase transition temperature.
  • Phosphatidyletanolamine is the main constituent of one important class of pH sensitive liposomes (for a review see Drummond et al, Prog Lipid Res 2000; 39(5): 409-460). pH sensitive liposomes are designed to release its payload when exposed to acidic environments.
  • liposomal doxorubicin (Caelyx®)
  • doxorubicin (Caelyx® or Doxil®)
  • Myhr and Moan 2006 liposomal doxorubicin
  • Doxil® is not engineered for ultrasound mediated drug release and shows a rather low drug release in vitro (see e.g. WO2008120998, incorporated herein in its entirety by reference).
  • US 2005/0019266 discloses lipid based ultrasound sensitive vesicles comprising a lipid, targeting ligand, gas or gas precursor, and, optionally, an oil. Due to the gass bubble, such microbubbles are too large for passive in target tissues and are therefore less suited for e.g. cancer treatment.
  • the current inventors have found that the sonosensitivity of drug delivery particles is surprisingly improved by the incorporation of non-lamellar forming lipids, more particularly, inverted structure forming lipids (ISF lipids). Even more surprising, the combination of ISF lipids and alcohols further improved sonosensitivity synergistically.
  • the current invention may be used to efficiently deliver drugs in a defined tissue volume to combat localized diseases. Such particles may passively or actively accumulate in the target tissue and the drug payload may be dumped in the tissue by means of ultrasound thereby increasing the therapeutic-to-toxicity ratio. Definitions
  • DOPE herein means 1 ⁇ -Dioleoyl-sn-Glycero-S-Phosphoethanolamine
  • DSPC means 1 ,2-distearoyl-sn-glycero-3 phosphocholine or, in short, distearoylphosphatidylcholine.
  • DSPE means i ⁇ -distearoyl-sn-glycero-S-phosphoethanolamine or distearoylphosphatidylethanolamine.
  • DSPE-PEGXXXX means 1 ,2-distearoyl-sn-glycero-3-phosphoethanolarnine-N-[meth- oxy(polyethylene glycol)-XXXX, wherein XXXX signifies the molecular weight of the polyethylene glycol moiety, e.g. DSPE-PEG2000 or DSPE-PEG5000.
  • ISF herein mean Inverted Structure Forming.
  • n-alcohol means any alcohol with n carbon atoms.
  • PC herein means phosphatidylcholine with any composition of acyl chain.
  • PE means phosphatidylethanolamine with any composition of acyl chain length.
  • PEG means polyethylene glycol or a derivate thereof.
  • PEGXXXX means polyethylene glycol or a derivate thereof, wherein XXXX signifies the molecular weight of the polyethylene glycol moiety.
  • POPE herein means i-palmitoyl ⁇ -oleoyl-sn-glycero-S-phosphoethanolamine.
  • SOPE herein means i-stearoyl- ⁇ -oleoyl-sn-glycero-S-phosphoethanolamine.
  • 'US' herein means ultrasound.
  • 'US sensitive', 'sonosensitive' or 'acoustically sensitive' herein means the ability of an entity, e.g. a particle, to release its payload upon exposure to acoustic energy.
  • Nominal concentration means the initial (weighed amounts per given volume) concentration of a constituent in the liposome membrane or in the hydration medium.
  • Inverted Structure Forming Lipid (ISF lipid) herein means an amphiphilic lipid with a spontaneous curvature of H ⁇ 1 , that is, with conical-like geometry.
  • ISF inverted structure forming
  • the current invention relates to a sonosensitive particulate material comprising an ISF lipid.
  • the particulate material may be arranged in any form of dispersion of a given internal structure.
  • preferred structures are hexagonal structures (e.g. Hexosome®), cubic structures (e.g. Cubosomes®), emulsion, microemulsions, liquid crystalline particles and liposomes.
  • the particulate material is a membrane structure, more preferably a liposome.
  • a liposome normally consists of a lipid bilayer with an aqueous interior.
  • the ISF lipid may be any amphiphilic lipid naturally prone to form so-called inverted structures.
  • the lipid may be e.g. glycerol based (e.g. phospholipids), or a sphingolipid (e.g. ceramides).
  • Lipid phase behaviour can be understood in terms of molecular shape, also known as packing parameter (P) or spontaneous curvature (H). Packing parameter may be described as
  • Lipids with a parameter P ⁇ 1 normally form hexagonal (Hi) phases or micelles, while lipids P>1 form inverted structures, like e.g. cubic, inverted hexagonal (Hn) or inverted micelles.
  • An ISF lipid is preferably an amphiphilic lipid with a packing parameter of P>1.
  • the ISF lipid may have symmetric or asymmetric acyl chains. 5
  • at least one of the acyl chains of the ISF lipid is 16 carbon atoms or longer, more preferably at least one of said chains is 18 carbon atoms or longer, and most preferably none of the acyl chains are shorter than 18 carbon atoms.
  • the particulate material may carry any concentration of ISF lipid sufficient to facilitate the sonosensitive effect, although the sonosensitivity generally increase with increasing io ISF lipid content.
  • the particulate material of the invention preferably comprises more than 10 mol%, more preferably more than 20 mol%, even more preferably more than 25 mol%, even more preferably more than 30 mol%, even more preferably more than 40 mol%, even more preferably more than 50 mol%, even more preferably more than 60 mol%, and yet even more preferably more than 70 mol% ISF lipid.
  • the ISF lipid concentration is 25, 47, 52, 54.5, 58, 62, 67, 72, or 77 mol%. Accordingly, the ISF lipid concentration is preferably within any of the possible ranges constituted by the mentioned embodiment concentrations.
  • the ISF lipid is preferably a glycerol based amphiphilic lipid, more preferably a phospholipid, even more preferably a phosphatidylethanolamine (PE) or a long chain
  • PC phosphatidylcholine
  • ISF PE may be saturated or non saturated; and of any suitable length. Examples of symmetric and asymmetric ISF PEs are shown in Table 1 and 2, respectively.
  • One or both acyl chains of the PE should preferably be 16 carbon atoms or longer, like 5 dipalmitoleoyl-, diheptadecanoyl-, distearoly-, dioleoly-, dielaidoyl-, dilinoeoyl-, dilinolenoyl-, diarachidonoyl-, docosa-hexaenoyl-, 1-palmitoyl-2-oleoyl-, 1-palmitoyl-2- linoleoyl-, 1-palmitoyl-2-arachidonoyl-, 1-palmitoyl-2-docosahexaenoyl-, 1-stearoyl-2- oleoyl-, i-stearoyl-2-linoleoyl-
  • At least one of the PE acyl chains is 18 carbon atoms or longer, most preferably none of the acyl chains are shorter than 18 carbon atoms. Also, it is preferred that at least one of the acyl chains is unsaturated. Even more preferred, both chains are unsaturated. More specifically, DSPE, DOPE, POPE, and/or SOPE are preferred. In preferred embodiments of the current invention the inverted structure5 forming phospholipid is DSPE, SOPE and/or DOPE. In a most preferred embodiment the inverted structure forming phospholipid is DOPE. The latter ISF lipid show surprisingly high sonosensitivity and stability.
  • At least one of the acyl chains of the ISF PC should preferably be 18 carbon atoms or longer, more preferably both acyl chains are 18 carbon atoms or longer, even more preferably at least one of the acyl chains is 20 carbon atoms or longer, and even more preferably the acyl chain is 22 carbons or longer. Furthermore, at least one of the ISF PC acyl chains is preferably unsaturated. Examples of preferred symmetric and asymmetric inverted structure forming PC are found in Table 3 and 4, respectively.
  • ISF PCs 1 ,2-dibehenoyl-sn-glycero-3-phosphocholine (DBPC), eicosenoyl, erucoyl, or nervonoyl, alone or in combination, are preferred ISF PCs.
  • Both ISF PE and PC may harbour additional groups on the acyl chain to make it more bulky as in e.g. diphytanoyl PE.
  • the current particulate material may comprise a suitable ISF PE and/or ISF PC phospholipid as the sole phospholipid or in combination with other lipids or phospholipids.
  • the particulate material comprises 47 mol % or more ISF PE and/or PC, more preferably 52 mol % or more, even more preferably 54.5 mol % or more, even more preferably 58 mol % or more, even more preferably 62 mol % or more, even more preferably 67 mol % or more, and yet even more preferably 77 mol % or more ISF PE and/or PC lipid.
  • a higher concentration of ISF lipid yields higher sonosensitivity.
  • an ISF lipid e.g. an ISF PE or PC
  • an ISF lipid will change properties, in particular spontaneous curvature, if the head group is modified. Conjugation of e.g. PEG to PE will make it prone to form micelles (P ⁇ 1 ) and it will consequently loose its capacity to form inverted structures.
  • e.g. DSPE-PEG is not an ISF lipid.
  • double bonds should exist in the cis conformation.
  • said ISF lipid is c/s-monounsaturated.
  • the material of the invention may further comprise an alcohol.
  • the alcohol may be any 5 alcohol, however, primary alcohols are preferred.
  • the alcohol or primary alcohol is hexanol. Any concentration of alcohol, e.g. hexanol, may beo employed in the hydration liquid used to hydrate the lipid film and generate liposomes.
  • the nominal alcohol concentration is at least 1 mM, preferably at least 10 mM, more preferably above 25mM, more preferably above 50 mM, even more preferably above 60 mM, and most preferably around 75 mM.
  • the inventors prefer that the concentrations is within the range 50 mM to 80 mM, more preferably within the range 60 mM to 75 mM.
  • the hexanol concentration is 25, 50, 60 or 75 mM.
  • the alcohol should be incorporated into the membrane to modulate the membrane sonosensitivity properties; in particular, the alkyl group of the alcohol should be embedded in the lipophilic part of the membrane.
  • membranes e.g. coatedo with an alcohol like polyvinyl alcohol, are not an essential part of the invention, neither are emulgating or solubilising alcohols like e.g. lanolin alcohol and octadecanol.
  • Sonosensitivity is not the sole parameter in selecting the optimal liposomal formulation.
  • Other key aspects are chemical stability, blood stability, blood clearance, 5 biodistribution, target tissue accumulation, and toxicity. The final goal is of course high therapeutic effect and/or reduced toxicity.
  • ISF lipids or alcohols are not alone in modulating these aspects and other components of the particle may be important in this respect.
  • Components for improving blood circulation time and/or further modulate sonosensitivity may be included in the material, like e.g. polyvinyl alcohols, polyethylene glycols (PEG), dextrans, or polymers. PEG or a derivate thereof, at any suitable concentration, is preferred.
  • PEG concentrations are preferably up to 15 mol %, more preferably within the range 3 to 10 mol %, even more preferably in the range 3 to 8 mol %, and even more preferably within the range 5.5 to 8 mol%. In embodiments of the current invention the PEG concentration is 3, 5.5, 8, or 10 mol%.
  • the PEG moiety may be of any molecular weight or type, however, it is preferred that the molecular weight is within the range 350 to 5000 Da, more preferably within 1000 - 3000 Da. In a preferred embodiment the molecular weight is 2000 Da.
  • the PEG moiety may be associated with any molecule allowing it to form part of the particulate material.
  • the PEG moiety is conjugated to a sphingolipid (e.g. ceramide), a glycerol based lipid (e.g. phospholipid), or a sterol (e.g. cholesterol), more preferably to a ceramide and/or PE, and even more preferably to PE, like DMPE, DPPE, or DSPE.
  • a sphingolipid e.g. ceramide
  • a glycerol based lipid e.g. phospholipid
  • a sterol e.g. cholesterol
  • the acyl chain length should be the same as that of the main phospholipid of the membrane.
  • the lipid-grafted PEG is preferably DPPE-PEG 2000 and/or DPPE-PEG 5000. In a particularly preferred embodiment lipid-grafted PEG is DSPE-PEG 2000.
  • lipids phospholipids, sphingolipids (e.g. ceramides), sterols, polyethyleneglycol, peptides, etc.
  • sphingolipids e.g. ceramides
  • sterols e.g. sterols
  • polyethyleneglycol e.g. ethylene glycol
  • peptides e.g. aminoethyl-N-(2-aminoethyl)
  • the size of the particulate material may be varied.
  • the particulate material may, in addition to the ISF lipids, further comprise any lipid.
  • the lipid is an amphiphilic lipid such as a sphingolipid and/or a phospholipid.
  • the amphiphilic lipids are phospholipids of any type or source.
  • the phospholipids may be saturated or unsaturated, or a combination thereof, although saturated phospholipids are preferred.
  • the selected phospholipids will have an acyl chain length longer than 12 carbon atoms, more often longer than 14 carbon atoms, and even more often longer than 16 carbon atoms.
  • the acyl chain length is within the range 14 to 24 carbon atoms, more preferably within 16 to 22 carbon atoms, even more preferably within 18 to 22.
  • Acyl chain of different lengths may be mixed in the material of the invention or all acyl chains may have similar or identical length.
  • the acyl chain length of the phospholipid is 18 carbon atoms.
  • the polar head of the phospholipid may be of any type, e.g. phosphatidylethanolamine (PE), phosphatidylcholine (PC), phosphatidic acid (PA), phosphatidyl serine (PS), or phosphatidylglycerol (PG).
  • the material of the invention may comprise mixtures of phospholipids with different polar heads.
  • Neutral phospholipid components of the lipid bilayer are preferably a phosphatidylcholine, most preferably chosen from diarachidoylphosphatidylcholine (DAPC), hydrogenated egg phosphatidylcholine (HEPC), hydrogenated soya phosphatidylcholine (HSPC), distearoylphosphatidylcholine (DSPC), dipalmitoylphosphatidylcholine (DPPC) and dimyristoylphosphatidylcholine (DMPC).
  • DAPC diarachidoylphosphatidylcholine
  • HEPC hydrogenated egg phosphatidylcholine
  • HSPC hydrogenated soya phosphatidylcholine
  • DSPC distearoylphosphatidylcholine
  • DPPC dipalmitoylphosphatidylcholine
  • DMPC dimyristoylphosphatid
  • Negatively charged phospholipid components of the lipid bilayer may be a phosphatidylglycerol, phosphatidylserine, phosphatidylinositol, phosphatidic acid or phosphatidylethanolamine compound, preferably a phosphatidylglycerol.
  • the additional or modulating phospholipid is PC, in particular DSPC.
  • the DSPC concentrations are within the range 5 to 30 mol %. The level of PC is important to modulate e.g. blood clearance rates.
  • the particulate material may also comprise a sterol, wherein the sterol may be cholesterol, a secosterol, or a combination thereof.
  • the secosterol is preferably vitamin D or a derivate thereof, more particularly calcidiol or a calcidiol derivate.
  • the particulate material may also comprise a sterol, wherein the sterol may be cholesterol, a secosterol, or a combination thereof.
  • the secosterol is preferably vitamin D or a derivate thereof, more particularly calcidiol or a calcidiol derivate.
  • the particulate material may comprise any suitable sterol concentration, preferably cholesterol, depending on the specific particle properties. In general, 50 mol% sterol is considered the upper concentration limit in liposome membranes.
  • the particulate material preferably comprises up to 20 mol % cholesterol, more preferably up to 30 mol %, and even more preferably up to 40 mol % cholesterol, and most preferably within the range 20 to 40 mol%.
  • the particulate material comprises 20, 26, 30, 35, or 40 mol % cholesterol. Accordingly, the cholesterol concentration is preferably within any of the possible ranges constituted by the mentioned embodiment concentrations. Higher concentration ranges are, however, preferred. Sterols may have a therapeutic effect, as well as improve stability and reduce blood clearance rates.
  • the particulate material of the invention may be of any suitable size. However, the material should preferably be less than 1000 nm, preferably less than 500 nm, more preferably less than 200 nm, more preferably 150 nm or less. In preferred embodiments the size falls within the range 50 to 200 nm, more preferably 50 to 150 nm more preferably 50 to 95 nm, even more preferably 80 to 90 nm.
  • the size is around 85 nm or 85 nm.
  • the current inventors' data show that size may be a parameter modulating the sonosensitivity of the particulate material. More specifically, size appears to be positively correlated with sonosensitivity. Hence, the optimal size range is predicted to be within the range 85 nm to 150 nm.
  • the particulate material of the invention typically comprises a drug or a functional molecule of any sort.
  • the drug may be any drug suitable for the purpose.
  • anti-bacterial drugs, anti-inflammatory drugs, anti cancer drugs, or any combination thereof are preferred.
  • anti cancer drugs are preferred.
  • Anti cancer drugs includes any chemotherapeutic, cytostatic or radiotherapeutic drug. It may be of special interest to load the current particulate material with deoxyribonucleic acid (DNA) or ribonucleic acid (RNA), in particular small interfering RNA (siRNA).
  • cytostatics are alkylating agents (L01A), anti-metabolites (L01B), plant alkaloids and terpenoids (L01C), vinca alkaloids (L01CA), podophyllotoxin (L01CB), taxanes (L01CD), topoisomerase inhibitors (L01CB and L01XX), antitumour antibiotics (L01D), hormonal therapy.
  • cytostatics are daunorubicin, cisplatin, docetaxel, 5-fluorouracil, vincristine, methotrexate, cyclophosphamide and doxorubicin.
  • the drug may include alkylating agents, antimetabolites, anti-mitotic agents, epipodophyllotoxins, antibiotics, hormones and hormone antagonists, enzymes, platinum coordination complexes, anthracenediones, substituted ureas, methylhydrazine derivatives, imidazotetrazine derivatives, cytoprotective agents, DNA topoisomerase inhibitors, biological response modifiers, retinoids, therapeutic antibodies, differentiating agents, immunomodulatory agents, and angiogenesis inhibitors.
  • the drug may also be alpha emitters like radium-223 (223Ra) and/or thorium-227 (227Th) or beta emitters. Other alpha emitting isotopes currently used in preclinical and clinical research include astatine-211 (211At), bismuth-213 (213Bi) and actinium-225 (225Ac).
  • the drug may further comprise anti-cancer peptides, like telomerase or fragments of telomerase, like hTERT; or proteins, like monoclonal or polyclonal antibodies, scFv, tetrabodies, Vaccibodies, Troybodies, etc.
  • the material of the invention may comprise collagenases or other enzymes. In particular proteins or molecules improving the uptake and distribution of particulate material in target tissues.
  • therapeutic agents that may be included in the particulate material include abarelix, aldesleukin, alemtuzumab, alitretinoin, allopurinol, altretamine, amifostine, anastrozole, arsenic trioxide, asparaginase, BCG live, bevaceizumab, bexarotene, bleomycin, bortezomib, busulfan, calusterone, camptothecin, capecitabine, carboplatin, carmustine, celecoxib, cetuximab, chlorambucil, cinacalcet, cisplatin, cladribine, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, darbepoetin alfa, daunorubicin, denileukin diftitox, dexrazoxane, docetaxel, doxorubicin,
  • doxorubicin epirubicin, or mitoxantrone
  • cisplatin etoposide, vinblastine, mitomycin, vindesine, gemcitabine, paclitaxel, docetaxel, carboplatin, ifosfamide, estramustine, or any combination thereof
  • doxorubicin methotrexate, 5-FU, cisplatin, siRNA, or any combination thereof.
  • the drug is a water soluble drug.
  • the drug is doxorubicin.
  • the particle of the invention may also comprise an imaging contrast agent, like e.g. an MR, X-ray, or optical imaging contrast agent, to render tracking and monitoring possible.
  • an imaging contrast agent like e.g. an MR, X-ray, or optical imaging contrast agent
  • Examples of MR and X-ray contrast agents, as well as fluorescent and bioluminescent probes may be found in the literature.
  • the particulate material as described anywhere supra does not comprise air bubbles of perfluorobutane or perfluoropropane gas, or any non-dissolved gasses.
  • heat sensitive or pH sensitive particles are not part of the current invention. More particularly, components making the particles heat sensitive, that is, releasing their payload below or above physiological temperature, like e.g. lysolipids, are not part of the current inventive particles. Accordingly, components like cholesterolhemisuccinate (CHEMS) or fatty acids (long chain fatty acids like e.g. oleic acid (OA)), N-palmitoyl homocysteine (PHC). diplamitoyl succinyl glycerol (DSPG), or similar components making the membrane sensitive to pH below or above physiological pH are not part of the current invention.
  • CHEMS cholesterolhemisuccinate
  • OA oleic acid
  • PLC N-palmitoyl homocysteine
  • DSPG diplamitoyl succinyl glycerol
  • Preparation of liposomes are well known within the art and a number of methods may be used to prepare the current particles.
  • the current invention also comprises the use of a sonosensitive particulate material comprising an ISF lipid for manufacturing a medicament for treating a condition or disease.
  • a sonosensitive particulate material comprising an ISF lipid for manufacturing a medicament for treating a condition or disease.
  • the particulate material is the material of the invention as described supra
  • Another aspect of the current invention is a therapeutic method for delivering a drug to a predefined tissue volume comprising administering a particulate material comprising an ISF lipid to a patient in need thereof. More particularly, the particular material is the particle of the invention, as described supra.
  • Yet another aspect is a method for treating a disease or condition comprising administering a particulate material comprising an ISF lipid as defined supra to a patient in need thereof. More particularly, the particulate material is the particle of the invention, as described supra.
  • the use or methods further comprise the step of administering or activating said particulate material by means of acoustic energy or ultrasound.
  • the active drug is released or administrated from the particulate material by means of acoustic energy.
  • the patient is protected against potential toxic effects of the drug en route to the target tissue and high local concentrations of the drug are obtainable in short time.
  • acoustic energy or ultrasound should preferably have a frequency below 3 MHz, more preferably below 1.5 MHz, more preferably below 1 MHz, more preferably below 0.5 MHz, more preferably below 0.25 MHz, and even more preferably below 0.1 MHz.
  • the frequency is 1.17 MHz, 40 kHz or 20 kHz. It should, however, be noted that focused ultrasound transducers may be driven at significantly higher frequencies than non-focused transducers and still induce efficient drug release from the current sonosensitive material. Without being limited to prevailing scientific theories, the current inventors believe that the level of ultrasound induced cavitation in the target tissue is the primary physical factor inducing drug release from the particulate material of the invention. A person skilled in the art of acoustics would know that ultrasound at any frequency may induce so-called inertial or transient cavitation.
  • the disease to be treated is typically of localised nature, although disseminated disease may also be treated.
  • the disease may be neoplastic disease, cancer, inflammatory conditions, immune disorders, and/or infections, preferably localised variants.
  • the methods described are particularly well suited to treat cancers, in particular solid tumours. Cancers readily available for ultrasound energy are preferred like e.g. cancers of head and neck, breast, cervix, kidney, liver, ovaries, prostate, skin, pancreas, as well as sarcomas.
  • the current sonosensitive particles are well suited to treat all above conditions as they naturally accumulate in such disease volumes.
  • the current invention further comprises a composition comprising the above sonosensitive particulate material, as well as a pharmaceutical composition comprising the above sonosensitive particulate material.
  • the current invention comprises a kit comprising the material of the invention.
  • the invention also comprises a process or method of producing the sonosensitiveo particulate material of the invention. Said method or process comprising the steps of producing a thin film of the constituents, except membrane embedded alcohols like e.g. hexanol, of the membrane as described above, and then hydrating the film with a suitable hydration liquid.
  • the hydration liquid may contain alcohol like e.g. hexanol.
  • the method or process may further comprise a freeze-thaw cycle followed by ans extrusion process.
  • the drug may be included in the hydration liquid or actively loaded at the end of the process or method. Embodiments of method or process are described in detail in the Examples section.
  • the current invention also comprises a product produced by the process or methodo described supra.
  • FIG. 3 Percent calcein release from liposomes (3 mol % DSPE-PEG 2000, 20 mol % cholesterol, 50 mM hexanol) containing two different main phospholipids (both at 77 is mol%): DSPC (open circles) and DSPE (closed squares) during exposure to 20 kHz ultrasound up to 6 minutes (see example 6). DSPE-based liposomes show superior sonosensitivity.
  • FIG. 5 2D surface plot of release extent (post 6 min US) vs. DSPE and hexanol levels (see example 7). High levels of hexanol and DSPE show positive synergy, while 25 low level of DSPE and high level of hexanol interact negatively.
  • Figure 8. 3D surface plot of release extent (post 6 min US) vs. DSPE and DSPE-PEG 35 2000 levels (see example 11).
  • Figure 9. Ultrasound mediated release of DOPE based liposomes in 20% serum. Release curve for Caelyx® given as reference.
  • Figure 10 40 kHz ultrasound mediated drug release of DEPC based liposomes in 20% serum. Release curve for Caelyx® given as reference.
  • Example 1 Preparation of liposomes containing fluorescent drug marker calcein DSPC 1 DSPE, DOPE and DSPE-PEG 2000 were purchased from Genzyme Pharmaceuticals (Liestal, Switzerland). Cholesterol, calcein, HEPES, TRITON-X100 (10% solution), sodium azide and sucrose were obtained from Sigma Aldrich. Hexanol was supplied by BDH Chemicals Ltd. (Poole, England).
  • Calcein carrying liposomes (liposomal calcein) of different membrane composition were prepared using the thin film hydration method (Lasic 1993). The nominal lipid concentration was 16 mg/ml. Liposomes were loaded with calcein via passive loading, the method being well known within the art.
  • the hydration liquid consisted of 10 mM HEPES (pH 7.4) and 50 mM calcein. For the preparation of liposomal calcein containing hexanol, the hydration liquid was supplemented with a given amount of hexanol 2 days prior to usage in the lipid film hydration step.
  • the liposomes were down-sized to 80-90 nm by extrusion (Lipex, Biomembrane Inc. Canada) at 65 0 C (DSPC liposomes), 23°C (DOPE liposomes) and 68 0 C (DSPE liposomes) through polycarbonate (Nuclepore) filters of consecutive smaller size.
  • Extraliposomal calcein was removed by extensive dialysis.
  • the dialysis was performed by placing disposable dialysers (MW cut off 100 000 D) containing the liposome dispersion, in a large volume of an isosmotic sucrose solution containing 10 mM HEPES and 0.02 % (w/v) sodium azide solution.
  • the setup was protected from light and the dialysis ended until the trace of calcein in the dialysis minimum was negligible.
  • the liposome dispersion was then, until further use, stored in the fridge protected from light.
  • Example 2 Characterisation of calcein containing liposomes Liposomes were characterised with respect to key physicochemical properties like particle size, pH and osmolality by use of well-established methodology.
  • the average particle size (intensity weighted) and size distribution were determined by photon correlation spectroscopy (PCS) at a scattering angle of 173° and 25 deg C (Nanosizer, Malvern Instruments, Malvern, UK).
  • the width of the size distribution is 5 defined by the polydispersity index. Prior to sample measurements the instruments was tested by running a latex standard (60 nm).
  • Osmolality was determined on non-diluted liposome dispersions by freezing point depression analysis (Fiske 210 Osmometer, Advanced Instruments, MA, US). Prior to sample measurements, a reference sample with an osmolality of 290 mosmol/kg was measured; if not within specifications, a three step calibration was performed. s Duplicates of liposome samples were analysed.
  • Example 3 US mediated release methodology and quantification for calcein containing liposomes o Liposome samples were exposed to 20 or 40 kHz ultrasound up to 6 min in a custom built sample chamber as disclosed in Huang and MacDonald (Huang and Macdonald 2004).
  • the US power supply and converter system was one of two systems: (1) 'Vibra- CeII' ultrasonic processor, VC 750, 20 kHz unit with a 6.35 cm diameter transducer or (2) 'Vibra-Ceir ultrasonic processor, VC754, 40 kHz unit with a 19mm cup horn probe,5 both purchased from Sonics and Materials, Inc. (USA). Pressure measurements were conducted with a Bruel and Kjaer hydrophone type 8103.
  • liposome dispersions were diluted in a 1 :500 volume ratio, with isosmotic sucrose solution containing 10 mM HEPES (pH 7.4) and 0.02 % (w/v) sodium azide. Duplicates were analysed.
  • the release assessment of calcein is based on the following well-established methodology: Intact liposomes containing calcein will display low fluorescence intensity due to self-quenching caused by the high intraliposomal concentration of calcein (here 50 mM). Ultrasound mediated release of calcein into the extraliposomal phase can be detected by an increase in fluorescence intensity due to a reduced overall quenching effect. The following equation is used for release quantification:
  • F b and F 11 are, respectively, the fluorescence intensities of the liposomal calcein sample before and after ultrasound application.
  • F ⁇ is the fluorescence intensity of the liposomal calcein sample after solubilisation with the surfactant (to mimic 100% release). Studies have shown that for calcein containing liposomes the solubilisation step must be performed at high temperature, above the phase transition temperature of is the phospholipid mixture.
  • Fluorescence measurements were either carried out with a Luminescence spectrometer model LS50B (Perkin Elmer, Norwalk, CT) equipped with a photomultiplier tube R3896 (Hamamatsu, Japan) or a QE6500 spectrometer with scientific grade detector (Ocean Optics B.V., Duiven, The Netherlands). Fluorescence
  • Two liposome formulations composed of 90 mol% DSPC and 10 mol% DSPE-PEG
  • Example 2 For the liposomes containing hexanol, the calcein solution (hydration liquid) was doped with hexanol at 60 mM concentration. The size of the hexanol containing liposomes was measured to 82 nm, while non-hexanol containing liposomes measured 95 nm (see Example 2 for size measurement methodology). The liposomes (diluted 1 :500 v/v)
  • Figure 1 shows that for the liposome formulation containing hexanol (full dots), the sonosensitivity was improved giving an increase in calcein release of 20% (in absolute value) compared to the liposome formulation containing no hexanol (open squares) this after 4 minutes of ultrasound treatment.
  • Example 5 Hexanol improves the sonosensitivitv of cholesterol containing liposomes
  • the development of a stable liposome formulation often requires the inclusion of a sterol in the membrane.
  • liposome size is known to affect ultrasound sensitivity. Therefore, the effect of incorporating hexanol on the sonosensitivity was evaluated for similar sized liposomes consisting of 50 mol% DSPC, 10 mol% DSPE-PEG2000 and 40 mol% cholesterol.
  • the liposomes were loaded with calcein as previously described and the size of hexanol and non-hexanol containing liposomes was measured to 88 nm and 89 nm, respectively.
  • the calcein solution hydrolysis liquid
  • liposomes composed of either 77 mol% DSPC or 77 mol% DSPE were investigated. Both formulations further consisted of 20 mol % cholesterol and 3 mol% DSPE-PEG 2000.
  • the calcein solution (hydration liquid) contained 50 mM hexanol.
  • the size of the calcein solution (hydration liquid) contained 50 mM hexanol.
  • DSPC-based and DSPE-based liposomes was 80 and 84 nm, respectively.
  • the ultrasound experiment was performed at 20 kHz and the percentage of calcein release was estimated by fluorescence measurements after 0.5, 1 , 1.5, 2 and 6 minutes of ultrasound exposure.
  • Figure 3 shows that for the DSPE-based liposomes (full dots), the sonosensitivity was increased compared to DSPC-based liposomes (open squares). We conclude that the inclusion of PE increases the sonosensitivity and drug release properties of liposomes.
  • Example 7 PE and hexanol svnerqisticallv improve sonosensitivity of liposomes As disclosed above the liposome sensitivity vis-a-vis US is affected by the inclusion of hexanol and/or PE lipids. To further investigate the effect of alcohols and/or PE lipids on liposomal sonosensitivity a multivariate study design was conducted. The initial study design comprised 11 different formulations where the amount of DSPE and hexanol was varied at different levels (see Table 5). For all formulations the level of cholesterol and DSPE-PEG 2000 was kept constant at 20 and 3 mol%, respectively.
  • Liposomes were prepared and analysed as previously described. Release experiments were performed at 40 kHz ultrasound. Results from the study are listed in Table 6.
  • Example 7 The study in Example 7 was extended to include DSPE liposome formulations s containing no hexanol. DSPE-PEG 2000 and cholesterol levels were held constant at 3 mol % and 20 mol %, respectively, whilst the target size was 85 nm. DSPC functioned as additional phospholipid. Liposomes were prepared and tested at 40 kHz ultrasound. Release data are listed in Table 7. o Table " 7 Batch data
  • Example 9 High levels of PEG do not reduce sonosensitivitv of DSPE liposomes
  • the DSPE-PEG 2000 level was increased from 3 to 8 mol %. Cholesterol was kept at 20 mol %, while DSPC functioned as additional phospholipid. Release data (at 40 kHz) are listed in Table 8.
  • Example 10 DOPE improves sonosensitivitv of liposomes Two liposomal calcein formulations containing DOPE as the main lipid were investigated. DSPE-PEG 2000 and cholesterol levels were kept constant at 8 mol % and 20 mol %, respectively. DSPC functioned as additional phospholipid. Release data (at 40 kHz) are given in Table 9.
  • DOPE-based liposomes have good sonosensitivity in the absence of any alcohols.
  • DOPE liposomes have a higher sonosensitivity compared to DSPE-based liposomes (Exp 2 vs. Exp 16).
  • Example 11 Effect of DSPE-PEG 2000 and cholesterol level on sonosensitibitv of DOPE-based liposomes.
  • the study design comprised 11 different formulations where the amount of DOPE, cholesterol and DSPE-PEG 2000 was varied at different levels (see Table 10).
  • Liposomes were prepared and analysed as previously described. Release experiments were performed at 40 kHz ultrasound. Results from the study are listed in Table 11.
  • Example 12 Preparation and characterisation of doxorubicin- containing liposomes DSPC 1 DSPE, DOPE and DSPE-PEG 2000 were purchased from Genzyme
  • Doxorubicin HCI was obtained from Nycomed, Norway. Cholesterol, citrate tri-sodium salt, Triton X-100 (10% solution), HEPES, ammonium sulphate, sodium azide, and sucrose were obtained from Sigma Aldrich. Hexanol was supplied by BDH Chemicals Ltd. (Poole. England).
  • Liposomes of different membrane composition were prepared using the thin film hydration method (Lasic 1993). The dry lipid film was hydrated with either 300 mM ammonium sulphate (pH 5.5 unbuffered) or 300 mM citrate (pH 4), see Table 12. The nominal lipid concentration was 20 mg/ml after hydration. In liposomes containing hexanol, the hydration solution was doped with a given amount of hexanol.
  • the liposome preparations were submitted to 3 freeze thaw cycles in a dry ice/acetone/methanol mixture.
  • the liposomes were downsized to small unilamellar vesicles of 80-90 nm by stepwise extrusion (Lipex. Biomembrane Inc. Canada) through polycarbonate (Nuclepore) filters. During extrusion the temperature was kept constant around the transition temperature for the respective liposome formulations.
  • Formation of an ammonium sulphate gradient or a pH citrate gradient was obtained by extensive dialysis.
  • the dialysis was performed by placing disposable dialysers (MW cut off 100 000 D) containing the liposome dispersion. Three consecutive dialysis exchanges against a large volume of either an isotonic sucrose solution (pH 5.5 unbuffered) or an isotonic 20 mM HEPES buffered NaCI solution (pH 7.4) (Table 12).
  • the liposome dispersions were then mixed with a given volume of doxorubicin HCI solution to give a final drug to lipid ratio of 1 :8 or 1 :16 and a final nominal lipid concentration of 16 mg/ml.
  • Vz- ⁇ h incubation at 23-75 0 C dependent on the membrane composition
  • the liposome sample was cooled down to room temperature.
  • the percent drug loading was determined by fluorescence measurements after separating free drug by dialysis or by using Sephadex G-50 columns. After loading the extraliposomal phase was exchanged with an isotonic 10 mM HEPES buffered sucrose solution (pH 7.4) or 20 mM HEPES buffered NaCI solution (pH 7.4) (Table 12).
  • DOPE-liposomes (Table 14) show very good stability in 20% serum (1 :125 dilution); no leakage of doxorubicin could be detected after 6 hours incubation at 37 deg C.
  • DEPC (Erucoyl or13-cis-docosenoic) is a long chain PC phospholipid with an acyl chain length of 22 carbon atoms and with one unsaturated bond. Liposomes with
  • Table 15 shows characterisation data, including sonosenitivity, for several long chain PC based liposomes.
  • DNPC 1 ,2-dinervonoyl-sn-glycero-3- phosphocholine
  • DNPC Nervonoyl or 15-cis-tetracosenoic; comprising acyl chain length of 24 carbon atoms with one unsaturated bond.
  • Liposomes with composition DNPC:DSPC:DSPE-PEG2000:CHOL of mol% 52:5:8:35 were produced and doxorubicin loaded as described supra. The formulation showed only 1% leakage after 6 hours of incubation in 20% serum at 37°C.
  • DNPC liposomes were reformulated to also comprise 1 ,2- dibehenoyl-s ⁇ -glycero-3-phosphocholine (DBPC), DBPC is a saturated long chain PC with an acyl chain length of 22 carbon atoms, with the composition DNPC:DBPC:DSPE-PEG2000:CHOL of molar percentage 25:27:8.40.
  • DBPC dibehenoyl-s ⁇ -glycero-3-phosphocholine
  • the formulation was loaded successfully with doxorubicin as described supra and tested with respect to serum stability and sonosensitivity: the formulation showed only 2% leakage after 6 hours of incubation in 20% serum at 37°C, while 83.0% and 57.5% of the drug load was released after 6 minutes of 40 kHz ultrasound exposure in HEPES buffered sucrose solution and 20% serum, respectively.
  • Example 15 Liposomes comprising low concentrations of long chain unsaturated PC show improved sonosensitivitv.
  • Liposomes with composition DEPC:DSPC:DSPE-PEG2000:CHOL of molar percentage 25:27:8:40 were manufactured and doxorubicin loaded as described above.
  • In ultrasound experiments almost 18% of the drug load was released after 6 minutes of 40 kHz ultrasound exposure in HEPES buffered sucrose, while the equivalent release in 20 % serum was 13%.
  • the experiment was conducted as described supra.
  • the release from hydrogenated soy PC based liposomes (Caelyx®) is only 3% after 6 min ultrasound exposure in 20 % serum.
  • Example 15 Animal blood clearance kinetics For anaesthesia a mixture of 2.4 mg/ml tiletamine/2.4 mg/ml zolazepam (Zoletil ® vet, Virbac Laboratories, Carros, France), 3.8 mg/ml xylazine (Narcoxyl ® vet, Roche, Basel, Switzerland) and 0,1 mg/ml butorphanol (Torbugesic ® , Fort Dodge Laboratories, Fort Dodge, IA) was administered at a dose of 0.1 ml s.c. Healthy mice received 7 mg/kg liposomal doxorubicin (DXR) under anaesthesia as a single i.v. bolus through the tail vein.
  • DXR liposomal doxorubicin
  • the total blood volume was collected by cardiac puncture using heparinized syringes and stored in heparinized tubes. The samples were kept on ice bath until storage at -80 0 C.
  • DXR Quantification of DXR was done as described by Gabizon et al. 1989. In brief, 0,1 ml whole blood samples (lysed due to freezing), was mixed with 1.9 ml 50 % acidified ethanol (equal parts of distilled water and cone, ethanol), creating a 1 :20 dilution. Duplicate samples were prepared. Tissue samples were added acidified ethanol in a 1 :10 dilution and homogenized using a Polytron ® Benchtop Homogenizer. The samples were incubated for 24 hrs at 4 0 C in the dark.
  • Liposomes comprising long chain unsaturated PCs eicosenoyl or nervonoyl will be produced and tested vis-a-vis sonosensitivity and in vitro stability. The data will show sonosensitivity comparable to DOPE liposomes disclosed supra.
  • Liposomes of composition SOPE:DSPE-PEG2000:CHOL of molar percentage 52:8:40 were manufactured and doxorubicin loaded as described above. In ultrasound experiments almost 62% of the drug load was released after 6 minutes of 40 kHz ultrasound exposure in HEPES buffered sucrose. The experiment was conducted as described supra.
  • liposomes of composition SOPE:DSPC:DSPE-PEG2000:CHOL of molar percentage 62:10:8:20 were manufactured and doxorubicin loaded as described above. Not more than 3% doxorubicin leakage was observed after 24hrs incubation in 20% serum at 37°C. In ultrasound experiments almost around 70% of the drug load was released after 6 minutes of 40 kHz ultrasound exposure in both HEPES buffered sucrose and 20% serum. The experiment was conducted as described supra. In contrast, release from hydrogenated soy PC based liposomes (Caelyx®) is only 3% after 6 min ultrasound exposure in 20 % serum (see figure 9). References

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Dispersion Chemistry (AREA)
  • Molecular Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • Engineering & Computer Science (AREA)
  • Dermatology (AREA)
  • Medicinal Preparation (AREA)

Abstract

La présente invention concerne des nouvelles particules sensibles à l'énergie acoustique transportant un médicament et comprenant des lipides formant une structure non lamellaire. L'invention porte également sur l'utilisation de ces particules et les procédés correspondants. Les particules transportant un médicament s'accumulent dans le tissu cible malade et libèrent efficacement leur charge utile à l'exposition d'une énergie acoustique.
PCT/NO2010/000218 2009-06-08 2010-06-08 Particules sensibles à l'énergie pour l'administration de médicament comprenant des lipides formant une structure non lamellaire WO2010143972A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
NO20092196 2009-06-08
NO20092196 2009-06-08

Publications (2)

Publication Number Publication Date
WO2010143972A2 true WO2010143972A2 (fr) 2010-12-16
WO2010143972A3 WO2010143972A3 (fr) 2011-09-29

Family

ID=42536416

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/NO2010/000218 WO2010143972A2 (fr) 2009-06-08 2010-06-08 Particules sensibles à l'énergie pour l'administration de médicament comprenant des lipides formant une structure non lamellaire

Country Status (1)

Country Link
WO (1) WO2010143972A2 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011078695A3 (fr) * 2009-12-22 2011-09-22 Epitarget As Particules d'administration de médicament acoustiquement sensibles comprenant de faibles concentrations de phosphatidyléthanolamine
WO2018136002A1 (fr) 2017-01-18 2018-07-26 Temasek Life Sciences Laboratory Limited Liposomes hyperstabilisés augmentant le ciblage de cellules mitotiques

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050019266A1 (en) 1997-05-06 2005-01-27 Unger Evan C. Novel targeted compositions for diagnostic and therapeutic use
WO2008120998A2 (fr) 2007-03-30 2008-10-09 Epitarget As Particules administrant des médicaments acoustiquement sensibles

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2002953285A0 (en) * 2002-12-12 2003-01-02 Protech Research Pty Ltd Yeast treatment
WO2009075582A2 (fr) * 2007-12-10 2009-06-18 Epitarget As Particules acoustiquement sensibles administrant des médicaments comprenant des lipides formant des structures non lamellaires
WO2009075583A1 (fr) * 2007-12-10 2009-06-18 Epitarget As Utilisation de particules comprenant un alcool

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050019266A1 (en) 1997-05-06 2005-01-27 Unger Evan C. Novel targeted compositions for diagnostic and therapeutic use
WO2008120998A2 (fr) 2007-03-30 2008-10-09 Epitarget As Particules administrant des médicaments acoustiquement sensibles

Non-Patent Citations (15)

* Cited by examiner, † Cited by third party
Title
AAGAARD. T..; M. KRISTENSEN. ET AL.: "Packing properties of 1-alkanols and alkanes in a phospholipid membrane.", BIOPHYS. CHEM., vol. 119, 2006, pages 61 - 68, XP025017215, DOI: doi:10.1016/j.bpc.2005.09.003
ANDRESEN. T. L.; S. S. JENSEN. ET AL.: "Advanced Startegies in Liposomal Cancer Therapy: problems and prospects of active and tumor specific drug release.", PROG. LIPID RES., vol. 44, no. 1, 2005, pages 68 - 97
DRUMMOND ET AL., PROG LIPID RES, vol. 39, no. 5, 2000, pages 409 - 460
DRUMMOND. D. C.; O. MEYER. ET AL.: "Optimizing Liposomes for Delivery of Chemotheraoeutic Agents to Solid Tumors.", PHARMACOL. REV., vol. 51, no. 4, 1999, pages 691 - 743, XP009059957
HOLLAND. J. W..; P. R. CULLIS. ET AL.: "Poly(ethylene glycol)-Lipid Conjugates Promote Bilayer Formation in Mixtures of Non-Bilayer-Forming Lipids.", BIOCHEMISTRY, vol. 35, 1996, pages 2610 - 2617
HUANG. S.; R. C. MACDONALD: "Acoustically active liposomes for drug encapsulation and ultrasound-triggered release.", BIOCHIM. BIOPHYS. ACTA, vol. 1665, 2004, pages 134 - 141, XP004592242, DOI: doi:10.1016/j.bbamem.2004.07.003
LARINA. I. V..; B. M. EVERS. ET AL.: "Optimal drug and gene delivery in cancer cells by ultrasound-induced cavitation.", ANTICANCER RES, vol. 25, no. 1A, 2005, pages 149 - 156, XP008117329
LARINA. I. V.; B. M. EVERS. ET AL.: "Enhancement of drug delivery in tumors by using interaction of nanoparticles with ultrasound radiation.", TECHNOL CANCER RES TREAT, vol. 4, no. 2, 2005, pages 217 - 226
LEE. A. G.: "Interactions between anesthetics and lipid mixtures. Normal alcohols.", BIOCHEMISTRY, vol. 15, 1976, pages 2448 - 2454
LIN. H. Y.; J. L. THOMAS: "PEG-Lipids and Oligo(ethylene glycol) Surfactants Enhance the Ultrasonic Permeabilizability of Liposomes.", LANGMUIR, vol. 19, no. 4, 2003, pages 1098 - 1105, XP002479875, DOI: doi:10.1021/la026604t
MYHR. G.; J. MOAN: "Synergistic and tumor selective effects of chemotherapy and ultrasound treatment.", CANCER LETT., vol. 232, 2006, pages 206 - 213
PITT. W. G.; G. A. HUSSEINI. ET AL.: "Ultrasonic drug delivery--a general review.", EXPERT OPIN DRUG DELIV, vol. 1, no. 1, 2004, pages 37 - 56, XP003004111, DOI: doi:10.1517/17425247.1.1.37
ROWE. E. S.; J. M. CAMPION: "Alcohol Induction of Interdigitation in Distearoylphosphatidylcholine: Fluorescence Studies of Alcohol Chain Length Requirements.", BIOPHYS. J., vol. 67, 1994, pages 1888 - 1895
THEWALT. J. L.; R. J. CUSHLEY: "Phospholipid/cholesterol membranes containing n-alkanols: a 2H-NMR study.", BIOCHIM. BIOPHVS. ACTA, vol. 905, 1987, pages 329 - 338, XP023618109, DOI: doi:10.1016/0005-2736(87)90461-5
TIERNEY. K. J..; D. E. BLOCK. ET AL.: "Elasticity and Phase Behavior of DPPC Membrane Modulated by Cholesterol. Ergosterol. and Ethanol", BIOPHVS. J., vol. 89, 2005, pages 2481 - 2493

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011078695A3 (fr) * 2009-12-22 2011-09-22 Epitarget As Particules d'administration de médicament acoustiquement sensibles comprenant de faibles concentrations de phosphatidyléthanolamine
WO2018136002A1 (fr) 2017-01-18 2018-07-26 Temasek Life Sciences Laboratory Limited Liposomes hyperstabilisés augmentant le ciblage de cellules mitotiques
CN110381922A (zh) * 2017-01-18 2019-10-25 淡马锡生命科学研究院有限公司 超稳定脂质体增加对有丝分裂细胞的靶向作用
EP3570818A4 (fr) * 2017-01-18 2020-11-18 Temasek Life Sciences Laboratory Limited Liposomes hyperstabilisés augmentant le ciblage de cellules mitotiques
RU2765736C2 (ru) * 2017-01-18 2022-02-02 Темасек Лайф Сайенсиз Лаборатори Лимитед Гиперстабилизированные липосомы, повышающие направленное таргетирование митотических клеток
US11331272B2 (en) 2017-01-18 2022-05-17 Temasek Life Sciences Laboratory Limited Hyperstabilized liposomes increase targeting of mitotic cells
AU2018210748B2 (en) * 2017-01-18 2023-09-28 Temasek Life Sciences Laboratory Limited Hyperstabilized liposomes increase targeting of mitotic cells

Also Published As

Publication number Publication date
WO2010143972A3 (fr) 2011-09-29

Similar Documents

Publication Publication Date Title
US9034374B2 (en) Acoustically sensitive drug delivery particles comprising non-lamellar forming phosphatidylethanolamine
US8765172B2 (en) Acoustically sensitive drug delivery particles comprising non-lamellar forming lipids
US20090098212A1 (en) Acoustically sensitive drug delivery particle
Large et al. Liposome composition in drug delivery design, synthesis, characterization, and clinical application
Mondal et al. Hybrid exosomes, exosome-like nanovesicles and engineered exosomes for therapeutic applications
Wang et al. Enhanced drug delivery using sonoactivatable liposomes with membrane-embedded porphyrins
Kataria et al. Stealth liposomes: a review.
Kotla et al. Biomimetic lipid-based nanosystems for enhanced dermal delivery of drugs and bioactive agents
US20110020429A1 (en) Use of particles comprising an alcohol
Zhu et al. Bubble-generating polymersomes loaded with both indocyanine green and doxorubicin for effective chemotherapy combined with photothermal therapy
Yang et al. Recent advances in liposome formulations for breast cancer therapeutics
US8906409B2 (en) Acoustically sensitive drug delivery particles comprising non-lamellar forming phosphatidylcholine
Evjen et al. Sonosensitive dioleoylphosphatidylethanolamine-containing liposomes with prolonged blood circulation time of doxorubicin
Li et al. Nanotechnology-enabled immunogenic cell death for improved cancer immunotherapy
CN108619096A (zh) 声动力敏感脂质体、药物组合物及其用途
US20120148663A1 (en) Lipophilic drug carrier
KR20150047336A (ko) 나노입자, 이를 제조하는 방법, 및 이의 용도
US20120288557A1 (en) Acoustically sensitive drug delivery particles comprising low concentrations of phosphatidylethanolamine
WO2010143972A2 (fr) Particules sensibles à l'énergie pour l'administration de médicament comprenant des lipides formant une structure non lamellaire
WO2014030601A1 (fr) Méthode de fabrication d'un nouveau lipoplexe à nanobulles constitué de polymères et de liposomes aux propriétés anioniques
Li et al. Lipid Nanocarrier‐Based mRNA Therapy: Challenges and Promise for Clinical Transformation
Reddy et al. Liposomes–a novel drug delivery system: A review
Evjen Sonosensitive liposomes for ultrasound-mediated drug delivery
Nogueira et al. Topical Drug Delivery Using Liposomes and Liquid Crystalline Phases for Skin Cancer Therapy
NO328851B1 (no) Ultralydsensitivt partiklaert material og anvendelse av nevnte material til fremstilling av et medikament for behandling av lokalisert sykdom, hvor legemiddelet frigjores ved ultralyd

Legal Events

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

Ref document number: 10727166

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 10727166

Country of ref document: EP

Kind code of ref document: A2