WO2000064484A2 - Conjugate having a cleavable linkage for use in a liposome - Google Patents

Conjugate having a cleavable linkage for use in a liposome Download PDF

Info

Publication number
WO2000064484A2
WO2000064484A2 PCT/US2000/010922 US0010922W WO0064484A2 WO 2000064484 A2 WO2000064484 A2 WO 2000064484A2 US 0010922 W US0010922 W US 0010922W WO 0064484 A2 WO0064484 A2 WO 0064484A2
Authority
WO
WIPO (PCT)
Prior art keywords
conjugate
therapeutic drug
mitomycin
drug
dithiobenzyl
Prior art date
Application number
PCT/US2000/010922
Other languages
English (en)
French (fr)
Other versions
WO2000064484A3 (en
Inventor
Samuel Zalipsky
Alberto A. Gabizon
Original Assignee
Alza Corporation
Hadasit Medical Research Services & Development Ltd.
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
Priority to AU46577/00A priority Critical patent/AU769425B2/en
Priority to IL14605500A priority patent/IL146055A0/xx
Priority to KR1020017013571A priority patent/KR100669053B1/ko
Priority to MXPA01010750A priority patent/MXPA01010750A/es
Application filed by Alza Corporation, Hadasit Medical Research Services & Development Ltd. filed Critical Alza Corporation
Priority to DE60030965T priority patent/DE60030965T2/de
Priority to CA2369595A priority patent/CA2369595C/en
Priority to DK00928321T priority patent/DK1173222T3/da
Priority to EP00928321A priority patent/EP1173222B1/en
Priority to HU0201425A priority patent/HUP0201425A3/hu
Priority to JP2000613474A priority patent/JP4558952B2/ja
Publication of WO2000064484A2 publication Critical patent/WO2000064484A2/en
Priority to IL146055A priority patent/IL146055A/en
Priority to NO20015144A priority patent/NO20015144L/no
Publication of WO2000064484A3 publication Critical patent/WO2000064484A3/en
Priority to HK02103601.3A priority patent/HK1041820B/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • 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
    • 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
    • A61K31/196Carboxylic acids, e.g. valproic acid having an amino group the amino group being directly attached to a ring, e.g. anthranilic acid, mefenamic acid, diclofenac, chlorambucil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/407Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with other heterocyclic ring systems, e.g. ketorolac, physostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/14Peptides containing saccharide radicals; Derivatives thereof, e.g. bleomycin, phleomycin, muramylpeptides or vancomycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/542Carboxylic acids, e.g. a fatty acid or an amino acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/543Lipids, e.g. triglycerides; Polyamines, e.g. spermine or spermidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/543Lipids, e.g. triglycerides; Polyamines, e.g. spermine or spermidine
    • A61K47/544Phospholipids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/554Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being a steroid plant sterol, glycyrrhetic acid, enoxolone or bile acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6905Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion
    • A61K47/6911Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion the form being a liposome
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/10Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and singly-bound oxygen atoms bound to the same carbon skeleton
    • C07C323/18Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and singly-bound oxygen atoms bound to the same carbon skeleton having the sulfur atom of at least one of the thio groups bound to a carbon atom of a six-membered aromatic ring of the carbon skeleton
    • C07C323/19Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and singly-bound oxygen atoms bound to the same carbon skeleton having the sulfur atom of at least one of the thio groups bound to a carbon atom of a six-membered aromatic ring of the carbon skeleton with singly-bound oxygen atoms bound to acyclic carbon atoms of the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/52Two oxygen atoms
    • C07D239/54Two oxygen atoms as doubly bound oxygen atoms or as unsubstituted hydroxy radicals
    • C07D239/545Two oxygen atoms as doubly bound oxygen atoms or as unsubstituted hydroxy radicals with other hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/553Two oxygen atoms as doubly bound oxygen atoms or as unsubstituted hydroxy radicals with other hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms with halogen atoms or nitro radicals directly attached to ring carbon atoms, e.g. fluorouracil
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains three hetero rings
    • C07D487/14Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H17/00Compounds containing heterocyclic radicals directly attached to hetero atoms of saccharide radicals
    • C07H17/04Heterocyclic radicals containing only oxygen as ring hetero atoms
    • C07H17/08Hetero rings containing eight or more ring members, e.g. erythromycins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/12Triazine radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T428/00Stock material or miscellaneous articles
    • Y10T428/29Coated or structually defined flake, particle, cell, strand, strand portion, rod, filament, macroscopic fiber or mass thereof
    • Y10T428/2982Particulate matter [e.g., sphere, flake, etc.]
    • Y10T428/2984Microcapsule with fluid core [includes liposome]

Definitions

  • the present invention relates to a conjugate comprised of a hydrophobic moiety, a cleavable linkage, and a therapeutic agent. More particularly, the present invention relates to conjugates comprised of a lipid, a cleavable linkage and a drug inco ⁇ orated into a liposomal formulation. The conjugates are cleavable under mild thiolytic conditions in vivo for release of the drug in an unmodified state.
  • Liposomes are closed lipid vesicles used for a variety of therapeutic purposes, a d in particular, for carrying therapeutic agents to a target region or cell by systemic administration of liposomes.
  • Liposomes having a surface grafted with chains of water- soluble, biocompatible polymer, in particular polyethylene glycol, have become important drug carries. These liposomes offer an extended blood circulation lifetime over liposomes lacking the polymer coating. The grafted polymer chains shield or mask the liposome, thus minimizing nonspecific interaction by plasma proteins. This in turn slows the rate at which the liposomes are cleared or eliminated in vivo since the liposome circulate unrecognized by macrophages and other cells of the reticuloendothelial system. Furthermore, due to the so-called enhanced permeability and retention effect, the liposomes tend to accumulate in sites of damaged or expanded vasculature, e.g. , tumors, sites of inflammation.
  • An extended blood circulation time is often desired to allow systemically administered liposomes to reach a target region, cell or site.
  • a blood circulation lifetime of greater than about 12 hours is preferred for liposomal-therapy to a tumor region, as the liposomes must systemically distribute and then extravasate into the tumor region.
  • liposome-based therapy One problem associated with liposome-based therapy is retention of drug within the liposome for a time sufficient for systemic distribution. This problem is of particular concern when long-circulating liposomes, i.e., liposomes with grafted polymer chains, are administered. Relatively few drugs can be efficiently loaded and retained for a long duration and subsequently released.
  • lipid bilayer components that render the bilayer less permeable to entrapped drug.
  • the lipid bilayer should be sufficiently fluidic such that the drug is released, for example by transport across the lipid bilayer or by lipid vesicle breakdown, at the desired time, e.g. , after localization at a target site or sufficient biodistribution.
  • Another approach to improving drug retention is to covalently attach the drug to a lipid in the liposomal lipid bilayer (Waalkes, et al., Selective Cancer Therap., 6: 15-22 (1990); Asai, et al, Biol. Pharm. Bull , 21:766-771 (1998)). It would be desirable to formulate a liposome composition having a long blood circulation lifetime and capable of retaining an entrapped drug for a desired time, yet able to release the drug on demand.
  • a liposome from a non-vesicle-forming lipid, such as dioleoylphosphatidylethanolamine (DOPE), and a lipid bilayer stabilizing lipid, such as methoxy-polyethylene glycol-distearoyl phosphatidylethanolamine (mPEG-DSPE) (Kirpotin, D, et al, FEBS Lett. 388:115-118 (1996)).
  • DOPE dioleoylphosphatidylethanolamine
  • mPEG-DSPE methoxy-polyethylene glycol-distearoyl phosphatidylethanolamine
  • the mPEG is attached to the DSPE via a cleavable linkage. Cleavage of the linkage destabilizes the liposome for a quick release of the liposome contents.
  • Labile bonds for linking PEG polymer chains to liposomes has been described (U.S. Patent Nos. 5,013,556, 5,891,468; WO 98/16201).
  • the labile bond in these liposome compositions releases the PEG polymer chains from the liposomes, for example, to expose a surface attached targeting ligand or to trigger fusion of the liposome with a target cell.
  • Senter describes a drug-antibody prodrug, where the antibody is linked to a drug using a disulfide benzyl carbamate or carbonate linker, and reduction of the disulfide bond effects release of the drug.
  • Senter' s teaching is specific to cleavage of a drug-ligand prodrug molecule, under the action of reducing agents such as 1,4- dithiothreitol, glutathione, NADH and NADPH.
  • an extended blood circulation time is a desirable feature of PEG- coated liposomes, with blood circulation lifetimes of greater than about 12 hours being preferred for liposomal-therapy to a tumor region.
  • the disclosure of Senter provides no guidance as to the release kinetics of a conjugate incorporated into a liposome under endogenous reducing conditions, such as during blood circulation of the liposome.
  • the invention includes a conjugate for use in a liposomal drug- delivery vehicle, the conjugate having the general structural formula:
  • L is a hydrophobic moiety suitable for incorporation into a liposomal lipid bilayer
  • R 1 represents a therapeutic drug covalently attached to the dithiobenzyl moiety, and where orientation of the CH 2 R' group is selected from the ortho position and the para position.
  • the therapeutic drug is covalently attached by a linkage selected from the group consisting of urethane, amine, amide, carbonate, thio- carbonate, ether and ester.
  • L is selected from the group consisting of cholesterol, a diacylglycerol, a phospholipid and derivatives thereof.
  • L is a diacylglycerol derivative to yield a conjugate having the general structural formula:
  • R 2 and R 3 are hydrocarbons having between about 8 to about 24 carbon atoms, or in another embodiment, from about 12 to about 22 carbon atoms. In still another embodiment, R 2 and R 3 are hydrocarbon chains of the same length.
  • the drug is selected from the group consisting of mitomycin C, mitomycin A, bleomycin, doxorubicin, daunorubicin, fluorodeoxyuridine, iododeoxyuridine, etoposide, AZT, acyclovir, vidarabine, arabinosyl cytosine, pentostatin, quinidine, atropine, chlorambucil, methotrexate, mitoxantrone and 5-fluorouracil.
  • the therapeutic drug is covalently linked to the dithiobenzyl moiety to form a conjugate having the structure:
  • R 4 represents a residue of the therapeutic drug.
  • R 4 in one embodiment is a therapeutic drug residue containing a primary or a secondary amine moiety thereby forming a urethane linkage between the dithiobenzyl and the therapeutic drug.
  • the therapeutic drug can be, for example, mitomycin A, mitomycin C, bleomycin or a polypeptide.
  • R 4 is a residue of a carboxyl-containing therapeutic drug, which forms an ester linkage between the dithiobenzyl and the therapeutic drug.
  • Examplary drugs in this embodiment include chlorambucil or methotrexate.
  • R 4 is a therapeutic drug residue containing a hydroxyl moiety thereby to form a carbonate linkage between the dithiobenzyl and the therapeutic drug.
  • exemplary drugs in this embodiment include fluorodeoxyuridine, iododeoxyuridine, etoposide, AZT, acyclovir, vidarabine, arabinosyl cytosine, pentostatin, quinidine, mitoxantrone and atropine.
  • the invention includes a liposome composition, comprising liposomes composed of vesicle-forming lipids including from about 1 to about 30 mole percent of a conjugate having the general structural described above.
  • the therapeutic drug is released from the conjugate in vivo in response to a physiologic condition or an artificially induced condition.
  • the invention includes a method for retaining a drug in a liposome, comprising preparing liposomes comprised of a vesicle-forming lipid and of between about 1 to about 30 mole percent of a conjugate described above.
  • the liposomes effectively retain the drug in the liposomes until release from the conjugate in response to a physiologic condition or an artificially induced condition.
  • Fig. 1 shows a synthetic reaction scheme for preparation of /r ⁇ ra-diacyldiglycerol- dithiobenzylalcohol for further reaction with amine-, hydroxy- or carboxyl-containing drugs;
  • Fig. 2 A shows a general reaction scheme for attachment of an amino-containing drug to a reactive diacyldiglycerol-dithiobenzylcarbonate
  • Fig. 2B shows the products after thiolytic cleavage of the conjugate in Fig. 2A;
  • Fig. 3 A shows a synthetic reaction scheme for preparation of a diacyldiglycerol- dithiobenzyl-5-fluorouracil conjugate ;
  • Fig. 3B shows the products after thiolytic cleavage of the conjugate in Fig. 3 A;
  • Fig. 4 shows an alternative synthetic reaction scheme for preparation of a diacyldiglycerol-dithiobenzyl-5-fluorouracil conjugate and the products after thiolytic cleavage of the conjugate;
  • Fig. 5 shows a synthetic reaction scheme for preparation of a diacyldiglycerol- dithiobenzyl-chlorambucil conjugate and the products after thiolytic cleavage of the conjugate;
  • Fig. 6 A shows a synthetic reaction scheme for preparation of a diacyldiglycerol- difhiobenzy 1-mitomycin-C conjugate ;
  • Fig. 6B shows the products after thiolytic cleavage of the conjugate in Fig. 6A
  • Fig. 7 shows a synthetic reaction scheme for preparation of a cholesterol- dithiobenzyl-mitomycin-C conjugate ;
  • Fig. 8 shows another synthetic reaction scheme for preparation of a cholesterol- dithiobenzy 1-mitomycin-C conjugate
  • Figs. 9A-9C show the structures of three lipid-dithiobenzy 1-mitomycin-C conjugates, ⁇ ra-distearoyl-DTB-mitomycin-C (Fig. 9A), /? ⁇ r ⁇ -dipalmitoyl-DTB- mitomycin-C (Fig. 9B) and ⁇ rtb ⁇ -dipalmitoyl-DTB- mitomycin-C (Fig. 9C);
  • Figs. 10A-10B are HPLC chromatograms for liposomes comprised of HSPC/mPEG-DSPE/lipid-DTB-mitomycin C (Fig. 10A) and HSPC/cholesterol/mPEG- DSPE/lipid-DTB-mitomycin C (Fig. 10B), where each figure shows a series of chromatograms as a function of time of incubation of the liposomes in the presence of cysteine;
  • Fig. 11 is a plot showing the percent of mitomycin C released from liposomes comprised of HSPC/mPEG-DSPE/lipid-DTB-mitomycin C (closed diamonds) and HSPC/cholesterol/mPEG-DSPE/lipid-DTB-mitomycin C (closed circles) as a function of time of incubation in the presence of cysteine;
  • Figs. 12A-12B are plots showing the percent of mitomycin C released from liposomes comprised of HSPC/mPEG-DSPE/lipid-DTB-mitomycin C (Fig. 12 A) and HSPC/cholesterol/mPEG-DSPE/lipid-DTB-mitomycin C (Fig. 12B) as a function of time of incubation in the presence of cysteine at concentrations of 150 ⁇ M (closed symbols) and at 1.5 mM (open symbols);
  • Fig. 13 is a plot of growth rate of Ml 09 cells, expressed as a percentage based on growth of M 109 cells in the absence of drug and cysteine, as a function of mitomycin C amount, in nM, for free mitomycin c (open triangles), liposomes comprised of HSPC/mPEG-DSPE/lipid-DTB-mitomycin C (closed squares), and liposomes comprised of HSPC/cholesterol/mPEG-DSPE/lipid-DTB-mitomycin C (open circles); Fig.
  • 14A is a plot of growth rate of Ml 09 cells, expressed as a percentage based on growth of M109 cells in the absence of drug or cysteine, as a function of mitomycin C concentration in nM. Shown are cells treated mitomycin C in free form (open triangles) and with mitomycin C in free form plus 1000 ⁇ M cystein (closed triangles). Also shown are cells treated with the liposome formulation comprised of HSPC/PEG- DSPE/lipid-DTB-mitomycin C (open circles) and with the liposome formulation with additional cysteine added at concentrations of 150 ⁇ M (open diamonds), 500 ⁇ M (closed circles) and 1000 ⁇ M (open squares);
  • Fig. 14B is a plot of growth rate of Ml 09 cells, expressed as a percentage based on growth of M109 cells in the absence of drug or cysteine, as a function of mitomycin C concentration in nM. Shown are cells treated mitomycin C in free form (open triangles) and with mitomycin C in free form plus 1000 ⁇ M cystein (closed triangles). Also shown are cells treated with the liposome formulation comprised of HSPC/cholesterol/mPEG-DSPE/lipid-DTB-mitomycin C (open circles) and with the liposome formulation with additional cysteine added at concentrations of 150 ⁇ M (open diamonds), 500 ⁇ M (closed circles) and 1000 ⁇ M (open squares); Fig. 15 is a plot showing the percent increase in cytotoxicity (as determined by
  • Fig. 16A is a plot showing the concentration of mitomycin C in the blood of rats as a function of time in hours following intravenous injection of free mitomycin C (open squares), liposomes comprised of HSPC/cholesterol/mPEG-DSPE/lipid-DTB- mitomycin C (closed diamonds), and liposomes comprised of HSPC/mPEG-DSPE/lipid- DTB-mitomycin C (closed circles); and
  • Fig. 16B is a plot showing the percent of injected dose remaining in the blood of rats as a function of time in hours following intravenous injection of free mitomycin C (open squares), liposomes comprised of HSPC/cholesterol/mPEG-DSPE/lipid-DTB- mitomycin C (closed diamonds), and liposomes comprised of HSPC/mPEG-DSPE/lipid- DTB-mitomycin C (closed circles).
  • hydrophobic moiety suitable for incorporation into a liposomal lipid bilayer intends any material comprising a hydrophobic portion capable of being integrated with the hydrophobic bilayer region of a liposomal lipid bilayer.
  • Such hydrophobic moieties are typically lipids, including amphipathic lipids having a hydrophobic lipid tail and a hydrophilic polar head, such as phospholipids and diacylglycerols.
  • Triglycerides, sterols, derivatives of phospholipids, diacylglyerols, sterols and triglycerides and other lipids derived from a natural source or synthetically prepared are also contemplated.
  • the term “residue” as in "therapeutic drug residue” intends a drug molecule that has been reacted to form an linkage with another molecule where at least one atom of the drug molecule is replaced or has been sacrificed to from the linkage.
  • Polypeptide refers to a polymer of amino acids and does not refer to a specific length of a polymer of amino acids. Thus, for example, the terms peptide, oligopeptide, protein, and enzyme are included within the definition of polypeptide. This term also includes post-expression modifications of the polypeptide, for example, glycosylations, acetylations, phosphorylations, and the like.
  • PEG poly(ethylene glycol); mPEG, methoxy-PEG; DTB, dithiobenzyl; DSPE, distearoyl phosphatidylethanolamine; HSPC, hydrogenated soy phosphatidylcholine; MMC, mitomycin C.
  • the invention includes a conjugate of the form:
  • L is a hydrophobic moiety suitable for incorporation into a liposomal lipid bilayer
  • R 1 represents a therapeutic drug residue covalently attached to the dithiobenzyl moiety, and where orientation of the CH 2 R' group is selected from the ortho position and the para position.
  • the hydrophobic moiety, L is typically a lipid such as a diacylglycerol, a sterol, a phospholipid, derivatives of these lipids, other naturally-occurring lipids and their synthetic analogs.
  • a therapeutic drug is attached to the dithiobenzyl moiety by a covalent linkage, thereby forming a drug residue, represented by R 1 in the structure.
  • the linkage will vary according to the drug and the reaction chemistry, as will be appreciated by those of skill in the art.
  • the therapeutic drug is covalently attached to the diithiobenzyl moiety by a linkage selected from the group consisting of urethane, amine, amide, carbonate, thio-carbonate, ether and ester.
  • a drug containing a primary or secondary amine such as mitomycin C, mitomycin A, bleomycin and therapeutic polypeptides to name a few, is reacted to from a urethane linkage with the amine moiety in the drug.
  • Exemplary drugs having such a moiety for reaction with dithiobenzyl alcohol to form a carbonate linkage include fluorodeoxyuridine, iododeoxyuridine, etoposide, AZT, acyclovir, vidarabine, arabinosyl cytosine, pentostatin, quinidine, mitoxantrone and atropine.
  • the linkage derives from reaction with a carboxylic acid moiety in the therapeutic drug, and an example of a conjugate having an ester linkage between chlorambucil and dithiobenzyl is described below.
  • Methotrexate is another example of a drug capable of forming an ester linkage with the dithiobenzyl moiety of the conjugate.
  • Conjugates having a urethane, carbonate or ester linkage attaching the drug to the dithiobenzyl moiety can generally be represented by the following structure:
  • R 4 represents a residue of the therapeutic drug.
  • the conjugate includes an ether linkage, which takes the form of O-R 4 , where R 4 represents the therapeutic drug residue.
  • the linkage typically derives from reaction with an alcohol functionality on the drug.
  • a conjugate with the drug 5-fluorouracil where an amine linkage is formed will be described below.
  • An amide linkage can also be formed with a peptide as the therapeutic agent, where the free carboxyl of an amino acid residue, such as an aspartic acid or glutamic acid, is condensed with dithiobenzylamine.
  • Fig. 1 shows a synthetic reaction scheme for preparation of exemplary conjugates in accord with the invention.
  • synthesis of an intermediate compound, ⁇ r ⁇ -diacyldiglyceroldithiobenzalcohol (Compound IV) is prepared for further reaction with a selected therapeutic drug.
  • Compound IV is prepared, as described in Example 1 , by reacting 3-mercapto-l ,2-propanediol (Compound I) with hydrogen peroxide to form r ⁇ c-3,3'-dithiobis(l ,2-propanediol) (Compound II).
  • Rac- 3,3'-dithiobis(l ,2-propanediol) is acylated with a hydrophobic moiety R.
  • R can be a fatty acid having from about 8 to about 24 carbon atoms.
  • Example 1 details the reaction procedure where R is stearic acid.
  • R is a fatty acid having from about 12 to about 22 carbon atoms.
  • Acylation of Compound II yields Rac- 3,3'-dithiobis(l ,2-propanedistearoyl) (Compound III), which is reacted with sulfuryl chloride and 4-mercaptobenzalcohol to form the desired intermediate product, para- diacyldiglycerol-dithiobenzalcohol (Compound IV).
  • Compound IV is readily reacted with a drug containing a reactive carboxyl moiety (R'C0 2 H) to form a lipid- dithiobenzyl (DTB)-drug conjugate where the drug is joined to the DTB via an ester linkage (Compound V).
  • Compound IV is also readily reacted with a drug containing a reactive amine moiety (R'-NH 2 ) to yield a lipid-DTB-drug conjugate where the drug is joined to the DTB by a urethane linkage (Compound VI).
  • Compound IV is also readily reacted with a drug containing a reactive hydroxyl moiety (R'OH) to form a lipid-DTB- drug conjugate where the drug is joined to the DTB by a carbonate linkage (Compound VII).
  • drugs are contemplated for use in the conjugate of the invention.
  • the invention contemplates drugs having an amine (NH or NH 2 ), carboxyl, sulfhydryl or hydroxyl moiety suitable for reaction.
  • "suitable for reaction” implies that the drug has one of the recited moieties capable of reacting with the dithiobenzyl moiety, in the form of, for example, dithiobenzyl alcohol.
  • Exemplary drugs include 5-fluorouracil, which has an NH group suitable for reaction, chlorambucil, which has a reactive carboxyl and mitomycin C, which has a reactive amine (aziridine group).
  • exemplary drugs contemplated for use include mitomycin C, mitomycin A, bleomycin, doxorubicin, daunorubicin, fluorodeoxyuridine, iododeoxyuridine, etoposide, AZT, acyclovir, vidarabine, arabinosyl cytosine, pentostatin, quinidine, atropine, chlorambucil, methotrexate, mitoxantrone and 5-fluorouracil.
  • polypeptides, aminoglycosides, alkaloids are all also suitable for use in the invention.
  • Example 1 also details the reaction conditions for preparation of ortho- diacyldiglyceroldithiobenzalcohol, which can serve as a intermediary compound to form the conjugate.
  • Figs. 2A-2B show preparation of a lipid-DTB-drug conjugate (Fig. 2A), and thiolytic cleavage of the conjugate in the presence of a reducing agent (Fig. 2B).
  • Fig. 2A Compound VII of Fig.
  • hydrophobic moiety R is derived from a fatty acid R"(CO)OH, such as stearic acid (CH 3 (CH 2 ) 16 CO 2 H), is reacted with an amine-containing drug, H 2 N-drug, in the presence of phosgene (COCl 2 ).
  • This reaction yields the lipid-DTB-drug conjugate illustrated in Fig. 2A.
  • the conjugate upon exposure to reducing conditions, i.e., a reducing agent such as cysteine or glutathione, decomposes to yield the products shown in Fig. 2B.
  • thiolytic cleavage of the conjugate results in regeneration of the drug in an unmodified, natural state.
  • the drug in conjugate can be readily incorporated into liposomes for administration in vivo to a subject. Further, the drug in the form of the conjugate is not toxic, as will also be shown below. After administration and upon exposure to endogeneous reducing agents or exposure to an exogeneous reducing agent, the conjugate decomposes to yield the drug in its native state and with biological activity.
  • Fig. 3 A a synthetic reaction scheme for preparation of a conjugate of 5- fluorouracil is illustrated.
  • Compound IV /r ⁇ ra-diacyl-diglycerol-dithiobenzalalcohol
  • /r ⁇ ra-toluenesulfonyl chloride to form the intermediate compound IX.
  • Reaction with 5-fluorouracil anion or sodium salt (Compound X) yields the desired lipid-DTB-5-fluorouracil conjugate (Compound XI).
  • Decomposition of the lipid-DTB- 5-fluorouracil conjugate (Compound XI) upon exposure to a reducing agent, R'-SH is shown in Fig. 3B. Thiolytic cleavage of the conjugate results in regeneration of 5- fluorouracil in an unmodified form.
  • Fig. 4 shows an alternative synthetic reaction scheme for preparation of a diacyldiglycerol-DTB-5-fluorouracil conjugate.
  • 1-chloroethyl chloroformate (Compound XII) is reacted with /? ⁇ r ⁇ -diacyl-diglycerol-dithiobenzalalcohol (Compound IV) to form a reactive chloroethyl carbonate-DTB-diacyldiglycerol intermediate (Compound XII).
  • the intermediate is subsequently reacted with 5-fluorouracil in the presence of ttiemanolamine (TEA) to yield a lipid-DTB-5-fluorouracil conjugate (Compound XIV).
  • TAA ttiemanolamine
  • a drug containing a reactive carboxyl moiety chlorambucil (Compound XV)
  • Compound IV ⁇ ra-diacyl-diglycerol-dithiobenzalalcohol
  • DCC 1,3- dicyclohexycarbodiimide
  • DMAP dimethylaminopyridine
  • a lipid- DTB-chlorambucil conjugate Compound XVI
  • DCC 1,3- dicyclohexycarbodiimide
  • DMAP dimethylaminopyridine
  • the conjugate thiolytically decomposes to the products shown. Chlorambucil is subsequently regenerated in an unmodified state.
  • Fig. 6 A shows the synthesis of a conjugate in accord with another embodiment of the invention.
  • mitomycin C Compound XVII, Fig. 6B
  • a drug containing a reactive amine moiety is reacted with /r ⁇ ra-diacyl-diglycerol- dithiobenzalalcohol (Compound IV) in the presence of phosgene to form a diacyldiglycerol-dithiobenzy 1-mitomycin-C conjugate (Compound XVIII).
  • Compound IV /r ⁇ ra-diacyl-diglycerol- dithiobenzalalcohol
  • phosgene diacyldiglycerol-dithiobenzy 1-mitomycin-C conjugate
  • Fig. 6B shows the thiolytic decomposition of a diacyldiglycerol-DTB-mitomycin-
  • hydrophobic moiety in the conjugate can be selected from any number of hydrophobic moieties, e.g. , lipids.
  • lipids e.g. lipids
  • a diacyldiglycerol lipid was used to form conjugates having the structure:
  • R 2 and R 3 are hydrocarbons having between about 8 to about 24 carbon atoms.
  • Fig. 7 shows another embodiment where cholesterol is used as the hydrophobic moiety in the conjugate.
  • Cholesterol (Compound XIV) is reacted with methanesulfonyl chloride in dichloromethane in the presence of triemylamine (TEA). The resulting intermediate is then converted into the thiol derivative and ultimately into the principal dithiobenzyl alcohol, which is used to link mitomycin C in a similar fashion as described above for diacylglycerol.
  • the invention includes a liposome composition comprised of a vesicle-forming lipid and a conjugate as described above.
  • Liposomes are closed lipid vesicles used for a variety of therapeutic purposes, and in particular, for carrying therapeutic agents to a target region or cell by systemic administration of liposomes.
  • liposomes having a surface coating of hydrophilic polymer chains, such as polyethylene glycol (PEG), are desirable as drug carries as these liposomes offer an extended blood circulation lifetime over liposomes lacking the polymer coating.
  • the polymer acts as a barrier to blood proteins thereby preventing binding of the protein and recognition of the liposomes for uptake and removal by macrophages and other cells of the reticuloendothelial system.
  • Liposomes include a conjugate in combination with a lipid, which in one embodiment is a vesicle-forming lipid, and, optionally, other bilayer components.
  • a lipid which in one embodiment is a vesicle-forming lipid, and, optionally, other bilayer components.
  • "Vesicle-forming lipids” are lipids that spontaneously form bilayer vesicles in water.
  • the vesicle-forming lipids preferably have two hydrocarbon chains, typically acyl chains, and a polar head group.
  • Examples include the phospholipids, such as phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidic acid (PA), phosphatidylinositol (PI), and sphingomyelin (SM).
  • a preferred lipid for use in the present invention is hydrogenated soy phosphatidylcholine (HSPC).
  • Another preferred family of lipids are diacylglycerols. These lipids can be obtained commercially or prepared according to published methods.
  • the vesicle-forming lipid may be selected to achieve a degree of fluidity or rigidity, to control the stability of the liposome in serum, and to control the rate of release of an entrapped agent in the liposome.
  • Liposomes having a more rigid lipid bilayer, or a liquid crystalline bilayer can be prepared by incorporation of a relatively rigid lipid, e.g., a lipid having a relatively high phase transition temperature, e.g. , up to about 80°C.
  • Rigid lipids i.e. , saturated, contribute to greater membrane rigidity in the lipid bilayer.
  • Other lipid components, such as cholesterol are also known to contribute to membrane rigidity in lipid bilayer structures.
  • Lipid fluidity is achieved by incorporation of a relatively fluid lipid, typically one having a lipid phase with a relatively low liquid to liquid-crystalline phase transition temperature, e.g. , at or below room temperature (about 20-25°C).
  • the liposome can also include other components that can be inco ⁇ orated into lipid bilayers, such as sterols. These other components typically have a hydrophobic moiety in contact with the interior, hydrophobic region of the bilayer membrane, and a polar head group moiety oriented toward the exterior, polar surface of the membrane.
  • Another lipid component in the liposomes of the present invention is a vesicle- forming lipid derivatized with a hydrophilic polymer. In this lipid component, a derivatized lipid results in formation of a surface coating of hydrophilic polymer chains on both the inner and outer lipid bilayer surfaces. Typically, between about 1-20 mole percent of the derivatized lipid is included in the lipid composition.
  • Hydrophilic polymers suitable for derivatization with a vesicle-forming lipid include polyvinylpyrrolidone, polyvinylmethylether, polymethyloxazoline, polyethyloxazoline, polyhydroxypropyloxazoline, polyhydroxypropylmethacrylamide, polymethacrylamide, polydimethylacrylamide, poly hydroxypropylmethacry late, polyhydroxyethylacrylate, hydroxymethylcellulose, hydroxyethylcellulose, polyethyleneglycol, and polyaspartamide.
  • the polymers may be employed as homopolymers or as block or random copolymers.
  • a preferred hydrophilic polymer chain is polyethyleneglycol (PEG), preferably as a PEG chain having a molecular weight between about 500 to about 10,000 Daltons, preferably between about 1,000 to about 5,000 Daltons.
  • PEG polyethyleneglycol
  • Methoxy or ethoxy-capped analogues of PEG are also preferred hydrophilic polymers. These polymers are commercially available in a variety of polymer sizes, e.g. , from about 12 to about 220,000 Daltons.
  • Liposomes of the present invention include typically between about 1 and about 30 mole percent of the lipid-DTB-drug conjugate, preferably between about 5 and about 30 mole percent, more preferably between about 5 and about 20 mole percent.
  • liposomes comprised of the vesicle- forming lipid hydrogenated soy phosphatidylcholine (HSPC), distearoyl phosphatidylethanolamine derivatized with methoxy-polyethylene glycol (mPEG-DSPE) and the conjugate shown in Fig. 9A, ⁇ ra-distearoyl-DTB-mitomycin C (Compound XVIII) were prepared as described in Examples 4A-4B.
  • HSPC vesicle- forming lipid hydrogenated soy phosphatidylcholine
  • mPEG-DSPE distearoyl phosphatidylethanolamine derivatized with methoxy-polyethylene glycol
  • Fig. 9A ⁇ ra-d
  • One of the liposome formulations included cholesterol (Example 4A), with the lipids HSCP/cholesterol/mPEG-DSPE//r ⁇ r ⁇ -distearoyl-DTB-mitomycin C (Compound XVIII) present at a molar ratio of 60/30/5/5.
  • the lipids HSCP/mPEG-DSPE/ ⁇ ra-distearoyl-DTB-mitomycin C (Compound XVII) were present at a molar ratio of 90/5/5.
  • the liposome formulations e.g., HSPC/cholesterol/mPEG-DSPE/conjugate Compound XVffl (hereinafter the "cholesterol-containing formulation”) and HSPC/mPEG- DSPE/conjugate Compound XVffl (hereinafter the "cholesterol-free liposome formulation”) were incubated at 37°C in the presence of 150 ⁇ M cysteine for 24 hours. Samples were withdrawn at selected time points and analyzed by high performance liquid chromatography (HPLC) to quantify the amount of conjugate and of free mitomycin C. The HPLC conditions are described in Example 5.
  • HPLC high performance liquid chromatography
  • Figs. 10A-10B show HPLC chromatograms for two liposome formulations.
  • Fig. 10A the results for the cholesterol-free liposome formulation are shown.
  • time zero there is no detectable free mitomycin C and all measurable drug is in the form of a lipid-DTB-drug conjugate that is liposome bound.
  • Fig. 10B shows the results for the liposome formulation containing cholesterol.
  • Fig. 11 is a plot showing the percent of mitomycin C released from the two liposome formulations, as determined from the chromatograms in Figs. 10A-10B.
  • the cholesterol-free liposomes (closed diamonds) had a higher rate of release than the liposomes containing cholesterol (closed circles). More than 50% of the mitomycin C was released from the liposome-bound conjugate after 2 hours for the cholesterol-free formulation. For both formulations, greater than 80% of the drug was released at the end of the 24 hour incubation period.
  • Figs. 12A-12B show the results of mitomycin C released from the lipid-DTB-drug conjugate inco ⁇ orated into the cholesterol-free liposomes (HSPC/PEG- DSPE/lipid-DTB-mitomycin C). The percent release during incubation with 150 ⁇ M are also shown (closed diamonds) for comparison. As seen, incubation at a higher concentration of reducing agent (1.5 mM, open diamonds) causes an increase in the rate of conjugate decomposition and rate of drug release.
  • Fig. 12B shows the results for the liposome formulation containing cholesterol. Liposomes incubated in 1.5 mM (open circles) have a significantly higher decomposition rate than the same liposomes incubated in 150 ⁇ M cysteine (closed circles).
  • Liposomes prepared as described in Examples 4A-4B with the molar ratios specified in Example 6A were tested. Cysteine at concentrations of 150 ⁇ M, 500 ⁇ M and 1000 ⁇ m was added to some of the test cells to effect thioytic decomposition of the conjugate and release of mitomycin C.
  • IC50 values were taken as the drug concentration which caused a 50% inhibition of the control growth rate (IC 50 ), as described in Example 6. The results are shown in Table 1.
  • the percent growth rate of M109 mouse carcinoma cells determined from the cytotoxicity studies is shown in Fig. 13.
  • the percent growth rate is expressed as a percentage based on growth rate of Ml 09 cells in the absence of mitomycin C and of cysteine and is shown as a function of mitomycin C concentration, in nM.
  • the growth rate of cells was determined as described in Example 6. As seen, the percent of cell growth rate decreases as the cysteine concentration is increased for both the liposomes containing cholesterol (open circles) and the cholesterol-free liposome formulation (closed squares). It can also be seen that cysteine has no effect on the activity of free mitomycin c and that mitomycin C is released from the conjugate to effectively inhibit cell growth.
  • Figs. 14A-14B the results for the liposome formulation containing no cholesterol are shown.
  • the growth rate of M109 cells is expressed as a percentage based on growth of M109 cells in the absence of drug and cysteine and is shown as a function of mitomycin C concentration in nM.
  • the cells treated with mitomycin C in free form (open triangles) and with mitomycin C in free form plus 1000 ⁇ M cysteine (closed triangles) exhibit a decrease in growth rate due tl e toxicity of the drug in free form.
  • Fig. 14B is a similar plot for the liposome formulation containing cholesterol.
  • Cytotoxicity of free mitomycin C (closed squares) is not effected by the presence of cysteine.
  • the cytotoxicity data shows that the cholesterol-free liposome formulation is more affected by cysteine.
  • the IC50 of the cholesterol-free liposome formulation at certain cysteine concentrations is only 2-fold lower than that of the free drug alone.
  • the liposome formulation containing cholesterol is less cytotoxic than the cholesterol-free liposome formulation.
  • the data also shows that cysteine has no cytotoxic effect of the tumor cells and no effect on the cytotoxicity of free mitomycin C. It is also apparent from the data that cysteine increases in a dose-dependent fashion the cytotoxcity of liposome-bound mitomycin C. Thus, the cytotoxic effects observed for the liposomal formulations are mostly accounted for by cysteine-mediated release of mitomycin C from the lipid-DTB-drug conjugate.
  • the in vivo pharmacokinetics of the liposomes containing cholesterol and the cholesterol-free liposome formulation was determined in rats. As described in Example 7, the animals were treated with a single bolus intravenous injection of approximately 0.1 mg/mL mitomycin C in free form or inco ⁇ orated into liposomes in the form of the lipid-DTB-mitomycin C conjugate in accord with the invention. After injection, blood samples were taken and analyzed for amount of mitomycin C. The results are shown in Figs. 16A-16B.
  • Fig. 16A shows the concentration ( ⁇ g/mL) of mitomycin C in the blood of rats as a function of time in hours following intravenous injection.
  • free mitomycin C (open squares) administered intravenously in free form is rapidly cleared from the blood.
  • Mitomycin C in the form of a liposome-bound lipid-DTB-drug conjugate remains in circulation for a substantially longer period of time.
  • Mitomycin C associated with liposomes containing cholesterol (closed diamonds) and with cholesterol-free liposomes (closed circles) was detected in the blood at greater than 10 ⁇ g/mL for 20-25 hours.
  • Fig. 16B shows the percent of injected dose remaining in the blood as a function of time in hours following intravenous injection of the test formulations.
  • the rac-3,3'-dithiobis(l,2-propanediol) product (Compound U) was acylated by adding the compound (980 mg, 4.6 mmol) to an oven-dried 100 mL round bottom flask and dissolving in dry methylene chloride (40 mL). To this, stearic acid (4.92 g, 17.1 mmol) and 4-dimethylamino)pyridinium 4-toluenesulfonate (1.38 g, 4.6 mmol) as the catalyst was and stirred at room temperature (25 °C) for 20 minutes.
  • the mitomycin C solution was added drop-wise the acyl chloride solution. After 1 hour, the toluene was evaporated off and the crude product was chromatographed (1: 1 hexane: ethyl acetate) on silica. The purified product was then taken up in t-BuOH (50 mL) and lyophilized. The product was a pu ⁇ le solid (183 mg, 53%).
  • Liposomes Containing Cholesterol 1. Liposome Preparation 59 mg HSPC, 14.4 mg cholesterol, 17.4 mg mPEG-DSPE, and 7.4 ⁇ ngpara- distearoyl-DTB-mitomycin C (molar ratio of 60/30/5/5) were added to 1 mL dehydrated ethanol at 60-65 °C and mixed until dissolved, approximately 10 minutes. A hydration medium composed of 10 mM histidine and 150 mM NaCl in distilled water was warmed to 70 °C.
  • the warm lipid solution was rapidly added to the warm (63-67 °C) hydration medium, with mixing, to form a suspension of liposomes having heterogeneous sizes.
  • the suspension was mixed for one hour at 63-67 °C.
  • the liposomes were sized to the desired mean particle diameter by controlled extrusion through polycarbonate filter cartridges housed in Teflon-lined stainless steel vessels.
  • the liposome suspension was maintained at 63-65 °C throughout the extrusion process, a period of 6-8 hours.
  • Ethanol was removed from the liposome suspension by diafiltration.
  • a histidine/sodium chloride solution was prepared by dissolving histidine (10 mM) and sodium chloride (150 mM) in sterile water. The pH of the solution was adjusted to approximately 7. The solution was filtered through a 0.22 ⁇ m Durapore filter. The liposome suspension was diluted in approximately a 1 : 1 (v/v) ratio with the histidine/sodium chloride solution and diafiltered through a polysulfone hollow-fiber ultrafilter. Eight volume exchanges were performed against the histidine/sodium chloride solution to remove the ethanol. The process fluid temperature was maintained at about 20-30 °C. Total diafiltration time was approximately 4.5 hours.
  • lipid concentration and conjugate/drug concentration were determined by HPLC.
  • Liposome particle size was measured by dynamic light scattering and the amount of "free" , unbound mitomycin C in the external suspension medium was measured by HPLC.
  • Liposomes were prepared as described above with a lipid composition of HSPC, mPEG-DSPE and ⁇ ra-distearoyl-DTB-mitomycin C in a molar ratio of 90/5/5. Specifically, 88.5 mg HPSC, 17.9 mg mPEG-DSPE (PEG MW 2000 Daltons) and 7.3 mg of the conjugate were dissolved in 1 mL ethanol. Liposome size, lipid and drug concentration and free mitomycin C concentration in the external suspension medium were determined after each processing step.
  • Liposomes prepared as described in Examples 4A-4B were diluted in 0.6 M octaylglucopyranoside. The liposomes were incubated in the presence of 150 mM cysteine at 37 °C. Samples with withdrawn at time zero, 30 minutes, 1 hour, 2 hours, 4 hours and 24 hours. A 20 ⁇ L volume was analyzed by HPLC using a Water Symmetry C 8 3.5 x 5 cm column. The flow rate was 1 mL/min and the mobile phase gradient as follows:
  • Liposomes prepared as described in Example 4A-4B, were composed of
  • HSPC/cholesterol/mPEG-DSPE/distearoyl-DTB-mitomycin C (90/45/5/5).
  • the liposome preparations were sterile filtered through 0.45 ⁇ m cellulose membranes and were not downsized via extrusion. After liposome formation, mitomycin C concentration was determined by absorbance at 360 nm in liposomes solubilized by 10- 20 fold dilution in isopropanol and the phospholipid concentration was determined by inorganic phosphate assay.
  • the liposomes containing cholesterol had an average diameter of 275 ⁇ 90 nm.
  • the cholesterol-free liposomes had an average diameter of 150 + 50 nm.
  • the phospholipid concentration in both liposome formulations was 10 ⁇ M/mL and the concentration of mitomycin C in both formulations was 120 ⁇ g/mL.
  • the cytotoxic effect of free mitomycin C or mitomycin C in the form of a distearoyl-DTB-mitomycin C conjugate inco ⁇ orated into liposomes was assayed colorimetrically by a methylene blue staining method described previously (Horowitz, A.T. et l , Biochim. Biophys. Acta, 1 109:203-209 (1992)) with slight modifications. Upon completion of the assay, the cells were fixed and evaluated using the methylene blue staining assay.
  • the plates were washed three times with deionized water, once with 0.1 M borate buffer (pH 8.5) and then stained for 60 minutes with 100 ⁇ l methylene blue (1 % in 0.1 M buffer borate, pH 8.5) at room temperature (20-25°C) .
  • the plates were rinsed in five baths of deionized water to remove non-cell bound dye and then dried.
  • the dye was extracted with 200 ⁇ l 0.1 N HC1 for 60 minutes at 37°C and the optical density was determined using a microplate spectrophotometer .
  • the cell number determined by counting cells with a hemocytometer correlated well with the spectrophotometric absorbance.
  • the percent growth inhibition or percent of control growth rate was obtained by dividing the growth rate of drug-treated cells by the growth rate of the untreated, control cells.
  • the drug concentration which caused a 50% inhibition of the control growth rate (IC 50 ) was calculated by inte ⁇ olation of the two closest values of the growth inhibition curve.
  • Mitomycin C was assayed in the range 10 "8 -10 "5 M.
  • the liposomal formulations with conjugate-bound were assayed in the range 10 "8 - 3 x 10 "5 M.
  • For interaction stodies cysteine (SIGMA, St. Louis, MO) was added together with the mitomycin C or liposome formulations to final concentration of 150, 500, or 1000 ⁇ M. The results are shown in Table 1 and in Figs. 13, 14 and 15A-15B.
  • Liposomes containing cholesterol and cholesterol-free liposomes were prepared as described in Example 5 A and 5B.
  • a solution of mitomycin C in free form was prepared by dissolving 11.9 mg of mitomycin C in 119 ⁇ L ethanol. After dissolution, approximately 11.8 ⁇ L of a solution of 10 mM histidine/ 150 mM saline was added. Prior to use, the mitomycn C solution was diluted to 100 ⁇ g/mL with the histidine/saline solution and filtered.
  • a single intravenous injection of the test formulation was administered as a bolus dose.
  • Blood samples were taken from each animal at the following times after injection: 30 seconds, 15 minutes, 30 minutes, 1 hour, 2 hours, 4 hours, 8 hours, 24 hours, 48 hours, 72 hours and 96 hours.
  • the quantity of mitomycin C in the blood samples was determined by the HPLC procedure given below.
  • a 200 mM iodoacetamine solution was prepared by placing 199.3 mg of iodoacetamide in 5.1 mL of 7.5% EDTA. 15 ⁇ L of the 200 mM iodoacetamide solution was placed in each 1 ⁇ L of blood sample.
  • a mobile phase of methanol and the aqueous buffer were mixed via a gradient program using a Waters Alliance binary pump.
  • the concentration ranges were 0.05-5.0 ⁇ g/mL and 0.1-5 ⁇ g/mL for mitomycin C and mitomycin C conjugate, respectively.
  • the final volume was adjusted to 1 mL with methanol.
  • a similar procedure was followed to prepare quality control samples.
  • the concentrations of quality control samples was 0.1, 0.5 and 5 ⁇ g/mL for mitomycin C and 0.1 , 1 and 5 ⁇ g/mL for mitomycin C conjugate in rat plasma.
  • the samples were spun down at 3,000 ⁇ m for 10 minutes at room temperature. 300 ⁇ L of supernatant was transferred to HPLC vials containing 300 ⁇ L insert for injection. 3.
  • a Supelco ® C-8, 5 ⁇ , 4.6mm x 5 cm column was used.
  • the mobile phase A was 10 mM ammonium phosphate, pH 7.
  • Mobil phase B was methanol.
  • the flow rate was 1 mL/min and detection was by UV at 360 nm.
  • the injection volume was 40 ⁇ L and the typical run time was 15 minutes.
  • the gradient program was as follows:
PCT/US2000/010922 1999-04-23 2000-04-21 Conjugate having a cleavable linkage for use in a liposome WO2000064484A2 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
CA2369595A CA2369595C (en) 1999-04-23 2000-04-21 Conjugate having a cleavable linkage for use in a liposome
KR1020017013571A KR100669053B1 (ko) 1999-04-23 2000-04-21 리포좀에 사용하기 위한 절단가능 결합을 갖는 콘쥬게이트
MXPA01010750A MXPA01010750A (es) 1999-04-23 2000-04-21 Conjugado que tiene un enlace dividible para utilizarse en un liposoma.
EP00928321A EP1173222B1 (en) 1999-04-23 2000-04-21 Conjugate having a cleavable linkage for use in a liposome
DE60030965T DE60030965T2 (de) 1999-04-23 2000-04-21 Konjugate enthaltend eine spaltbare bindung zur anwendung in einem liposom
IL14605500A IL146055A0 (en) 1999-04-23 2000-04-21 Conjugate having a cleavable linkage for use in a liposome
DK00928321T DK1173222T3 (da) 1999-04-23 2000-04-21 Konjugat med en spaltelig binding til brug i et liposom
AU46577/00A AU769425B2 (en) 1999-04-23 2000-04-21 Conjugate having a cleavable linkage for use in a liposome
HU0201425A HUP0201425A3 (en) 1999-04-23 2000-04-21 Conjugate having a cleavable linkage for use in a liposome
JP2000613474A JP4558952B2 (ja) 1999-04-23 2000-04-21 リポソームにおいての使用のための開裂可能な結合を有する複合体
IL146055A IL146055A (en) 1999-04-23 2001-10-18 A conjugate with a link can be fissured for liposome use
NO20015144A NO20015144L (no) 1999-04-23 2001-10-22 Konjugat med en spaltbar binding for anvendelse i et liposom
HK02103601.3A HK1041820B (zh) 1999-04-23 2002-05-13 用於脂質體的有可裂鍵的合成物

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US13089799P 1999-04-23 1999-04-23
US60/130,897 1999-04-23

Publications (2)

Publication Number Publication Date
WO2000064484A2 true WO2000064484A2 (en) 2000-11-02
WO2000064484A3 WO2000064484A3 (en) 2001-11-15

Family

ID=22446872

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/010922 WO2000064484A2 (en) 1999-04-23 2000-04-21 Conjugate having a cleavable linkage for use in a liposome

Country Status (18)

Country Link
US (8) US6365179B1 (US06605299-20030812-C00002.png)
EP (3) EP1579874A3 (US06605299-20030812-C00002.png)
JP (1) JP4558952B2 (US06605299-20030812-C00002.png)
KR (2) KR100669053B1 (US06605299-20030812-C00002.png)
CN (2) CN1244376C (US06605299-20030812-C00002.png)
AT (1) ATE340592T1 (US06605299-20030812-C00002.png)
AU (1) AU769425B2 (US06605299-20030812-C00002.png)
CA (1) CA2369595C (US06605299-20030812-C00002.png)
DE (1) DE60030965T2 (US06605299-20030812-C00002.png)
DK (1) DK1173222T3 (US06605299-20030812-C00002.png)
ES (1) ES2272280T3 (US06605299-20030812-C00002.png)
HK (1) HK1041820B (US06605299-20030812-C00002.png)
HU (2) HUP0200797A3 (US06605299-20030812-C00002.png)
IL (2) IL146055A0 (US06605299-20030812-C00002.png)
MX (2) MXPA01010750A (US06605299-20030812-C00002.png)
NO (1) NO20015144L (US06605299-20030812-C00002.png)
WO (1) WO2000064484A2 (US06605299-20030812-C00002.png)
ZA (2) ZA200108726B (US06605299-20030812-C00002.png)

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1385479A1 (en) * 2001-03-26 2004-02-04 Alza Corporation Liposome composition for improved intracellular delivery of a therapeutic agent
WO2004110497A2 (en) 2003-04-30 2004-12-23 Alza Corporation Mitomycin conjugates cleavable by thiols
WO2005053749A2 (en) * 2003-11-26 2005-06-16 Alza Corporation Thiol-cleavable linkage between polymer and ligand
US7211440B2 (en) 2002-03-08 2007-05-01 Wallac Oy Dissociative fluorescence enhancement assay
US7276248B2 (en) 1999-04-23 2007-10-02 Alza Corporation Conjugate having a cleavable linkage for use in a liposome
CN101284136A (zh) * 2002-12-03 2008-10-15 布朗歇特洛克菲勒神经科学研究所 传输物质穿过血脑屏障的人工低密度脂蛋白载体
US9181321B2 (en) 2013-03-14 2015-11-10 Shire Human Genetic Therapies, Inc. CFTR mRNA compositions and related methods and uses
US9308281B2 (en) 2011-06-08 2016-04-12 Shire Human Genetic Therapies, Inc. MRNA therapy for Fabry disease
US9522176B2 (en) 2013-10-22 2016-12-20 Shire Human Genetic Therapies, Inc. MRNA therapy for phenylketonuria
US9629804B2 (en) 2013-10-22 2017-04-25 Shire Human Genetic Therapies, Inc. Lipid formulations for delivery of messenger RNA
US9717690B2 (en) 2011-06-08 2017-08-01 Rana Therapeutics, Inc. Cleavable lipids
US9850269B2 (en) 2014-04-25 2017-12-26 Translate Bio, Inc. Methods for purification of messenger RNA
GB2552301A (en) * 2016-07-11 2018-01-24 Evox Therapeutics Ltd Metabolic drug loading of EVs
US9877919B2 (en) 2012-03-29 2018-01-30 Translate Bio, Inc. Lipid-derived neutral nanoparticles
US9957499B2 (en) 2013-03-14 2018-05-01 Translate Bio, Inc. Methods for purification of messenger RNA
US10022455B2 (en) 2014-05-30 2018-07-17 Translate Bio, Inc. Biodegradable lipids for delivery of nucleic acids
US10138213B2 (en) 2014-06-24 2018-11-27 Translate Bio, Inc. Stereochemically enriched compositions for delivery of nucleic acids
WO2019016208A1 (en) * 2017-07-17 2019-01-24 Technische Universiteit Eindhoven APPLICABLE CHEMICAL COMPOSITION COMPRISING AN AGENT CONJUGATED TO A HYDROPHOBIC PART AND A SUPPORT
US10576166B2 (en) 2009-12-01 2020-03-03 Translate Bio, Inc. Liver specific delivery of messenger RNA
US11173190B2 (en) 2017-05-16 2021-11-16 Translate Bio, Inc. Treatment of cystic fibrosis by delivery of codon-optimized mRNA encoding CFTR
US11174500B2 (en) 2018-08-24 2021-11-16 Translate Bio, Inc. Methods for purification of messenger RNA
US11224642B2 (en) 2013-10-22 2022-01-18 Translate Bio, Inc. MRNA therapy for argininosuccinate synthetase deficiency
US11253605B2 (en) 2017-02-27 2022-02-22 Translate Bio, Inc. Codon-optimized CFTR MRNA
US11254936B2 (en) 2012-06-08 2022-02-22 Translate Bio, Inc. Nuclease resistant polynucleotides and uses thereof

Families Citing this family (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7422902B1 (en) * 1995-06-07 2008-09-09 The University Of British Columbia Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer
US5981501A (en) * 1995-06-07 1999-11-09 Inex Pharmaceuticals Corp. Methods for encapsulating plasmids in lipid bilayers
DE69841002D1 (de) 1997-05-14 2009-09-03 Univ British Columbia Hochwirksame verkapselung von nukleinsäuren in lipidvesikeln
US7112337B2 (en) * 1999-04-23 2006-09-26 Alza Corporation Liposome composition for delivery of nucleic acid
GB9915074D0 (en) * 1999-06-28 1999-08-25 Cortecs Plc Ligand-binding composition
US7094423B1 (en) * 1999-07-15 2006-08-22 Inex Pharmaceuticals Corp. Methods for preparation of lipid-encapsulated therapeutic agents
WO2002085908A1 (en) * 2001-04-24 2002-10-31 Purdue Research Foundation Folate mimetics and folate-receptor binding conjugates thereof
WO2003039595A2 (en) * 2001-11-07 2003-05-15 Inex Pharmaceuticals Corporation Mucosal adjuvants comprising an oligonucleotide and a cationic lipid
US7842498B2 (en) * 2001-11-08 2010-11-30 Bio-Rad Laboratories, Inc. Hydrophobic surface chip
US6737524B2 (en) * 2002-03-25 2004-05-18 Paul K. Smith Activated polyethylene glycol compounds
US20040009944A1 (en) * 2002-05-10 2004-01-15 Inex Pharmaceuticals Corporation Methylated immunostimulatory oligonucleotides and methods of using the same
CA2752143C (en) 2002-05-15 2014-09-23 Sutter West Bay Hospitals Delivery of nucleic acid-like compounds
KR20040106547A (ko) * 2002-05-15 2004-12-17 엔도사이트, 인코포레이티드 비타민-마이토마이신 공액체
US7601332B2 (en) 2003-01-27 2009-10-13 Endocyte, Inc. Vitamin receptor binding drug delivery conjugates
JP2007509040A (ja) * 2003-10-11 2007-04-12 イネックス ファーマシューティカルズ コーポレイション 先天性免疫及び抗体依存性細胞傷害を強化するための方法及び組成物
JP2007520481A (ja) * 2004-01-15 2007-07-26 アルザ・コーポレーシヨン 治療薬を送達するためのリポソーム組成物
US7282590B2 (en) * 2004-02-12 2007-10-16 The Research Foundation Of State University Of New York Drug conjugates
US7931693B2 (en) * 2004-02-26 2011-04-26 Endosphere, Inc. Method and apparatus for reducing obesity
JP2007532666A (ja) * 2004-04-14 2007-11-15 アヴィリッド インコーポレーテッド ウイルス核酸を対象とした修飾ヌクレアーゼを用いた組成物及びウイルス性疾患の予防並びに治療方法
CA2562527A1 (en) * 2004-04-21 2005-11-10 Alza Corporation Polymer conjugate releasable under mild thiolytic conditions
JP2007533750A (ja) * 2004-04-21 2007-11-22 アルザ コーポレイション 緩和なチオール分解条件下で遊離可能なポリマー結合体
WO2006012527A1 (en) 2004-07-23 2006-02-02 Endocyte, Inc. Bivalent linkers and conjugates thereof
US20060058236A1 (en) * 2004-09-03 2006-03-16 Hutchins Maria U Endogenously-formed conjugate of albumin
TW200612993A (en) * 2004-10-08 2006-05-01 Alza Corp Lipopolymer conjugates
CA2584583A1 (en) * 2004-10-28 2006-05-11 Alza Corporation Lyophilized liposome formulations and method
JP5289935B2 (ja) * 2005-03-16 2013-09-11 エンドサイト,インコーポレイテッド プテロイン酸およびその結合体の合成と精製
RU2470668C2 (ru) * 2005-08-19 2012-12-27 Эндосайт, Инк. Конъюгаты лиганда с несколькими лекарственными средствами
EP2382995A3 (en) * 2005-08-19 2013-09-25 Endocyte, Inc. Ligand conjugates of Vinca alkaloids, analogs and derivatives
MX2009000658A (es) * 2006-07-19 2009-01-30 Univ Texas Preparaciones de fosfolipidos y componentes farmaceuticos que contienen acido 5-amino salicilico para el tratamiento de una enfermedad inflamatoria del intestino delgado.
US20100329664A1 (en) * 2007-01-23 2010-12-30 Lim Dae-Soon Shutter device for camera
US20100104626A1 (en) * 2007-02-16 2010-04-29 Endocyte, Inc. Methods and compositions for treating and diagnosing kidney disease
US9555139B2 (en) * 2007-03-14 2017-01-31 Endocyte, Inc. Binding ligand linked drug delivery conjugates of tubulysins
ITRM20070327A1 (it) * 2007-06-11 2008-12-12 Univ Palermo Vettori colloidali a struttura poliamminoacidica per il rilascio orale di peptidi e proteine e relativo metodo di produzione.
US9877965B2 (en) 2007-06-25 2018-01-30 Endocyte, Inc. Vitamin receptor drug delivery conjugates for treating inflammation
US9138484B2 (en) 2007-06-25 2015-09-22 Endocyte, Inc. Conjugates containing hydrophilic spacer linkers
WO2009006311A2 (en) * 2007-06-29 2009-01-08 Wisconsin Alumni Research Foundation Structuring effect of cholesterol in peg-phospholipid micelles, drug delivery of amphotericin b, and combination antifungals
CN101854917A (zh) * 2007-07-20 2010-10-06 巴斯夫欧洲公司 包含跨膜转运触发体系的小泡
AU2008317383B2 (en) 2007-10-23 2013-11-14 Nektar Therapeutics Hydroxyapatite-targeting multiarm polymers and conjugates made therefrom
CA2703491C (en) 2007-10-25 2017-06-13 Endocyte, Inc. Tubulysins and processes for preparing
US9259398B1 (en) * 2007-11-26 2016-02-16 Abbott Cardiovascular Systems Inc. Bioactive agent-loaded targeting micelles
WO2009141738A2 (en) * 2008-05-23 2009-11-26 The University Of British Columbia Modified drugs for use in liposomal nanoparticles
EP2393854B1 (en) * 2009-02-04 2019-12-04 The Brigham and Women's Hospital, Inc. Nanoscale platinum compounds and methods of use thereof
KR20180026572A (ko) 2009-05-27 2018-03-12 셀렉타 바이오사이언시즈, 인크. 방출 속도가 상이한 성분을 갖는 나노운반체
CN107617110A (zh) * 2009-08-26 2018-01-23 西莱克塔生物科技公司 诱导t细胞辅助的组合物
US20120219538A1 (en) * 2009-11-02 2012-08-30 Therapeomic Ag Stabilized protein formulations and use thereof
WO2011119995A2 (en) 2010-03-26 2011-09-29 Cerulean Pharma Inc. Formulations and methods of use
DE102010042338A1 (de) * 2010-10-12 2012-04-12 Bayer Technology Services Gmbh Zusammensetzung zur Behandlung von Krebs mit kontrollierter Freisetzung des Wirkstoffes
EA201390660A1 (ru) 2010-11-05 2013-11-29 Селекта Байосайенсиз, Инк. Модифицированные никотиновые соединения и связанные способы
BR112013027500A2 (pt) 2011-04-29 2017-01-10 Selecta Biosciences Inc liberação controlada de imunossupressores de nanotransportadores sintéticos
EP2606884A1 (en) 2011-12-21 2013-06-26 Ecole Polytechnique Fédérale de Lausanne (EPFL) Inhibitors of notch signaling pathway and use thereof in treatment of cancers
WO2013126797A1 (en) 2012-02-24 2013-08-29 Purdue Research Foundation Cholecystokinin b receptor targeting for imaging and therapy
US20140080175A1 (en) 2012-03-29 2014-03-20 Endocyte, Inc. Processes for preparing tubulysin derivatives and conjugates thereof
BR112015008365A2 (pt) 2012-10-16 2017-07-04 Endocyte Inc composto da fórmula b-l(d)x, ou um sal farmaceuticamente aceitável do mesmo, composição farmacêutica, uso de um composto, composição de forma de dosagem unitária ou dose unitária, composição para tratar um câncer em um paciente, e método para tratar um câncer em um paciente
US9827552B2 (en) 2013-07-17 2017-11-28 Clemson University Functionalized lipid modification of solid phase surfaces for use in chromatography
EP3151864B1 (en) * 2014-06-09 2020-08-05 Lipomedix Pharmaceuticals Ltd. Combination therapy comprising a liposomal prodrug of mitomycin c and radiotherapy
US20180071253A1 (en) 2015-03-17 2018-03-15 Lipomedix Pharmaceuticals Ltd. Methods for the treatment of bladder cancer
CN106519211B (zh) * 2015-09-10 2018-10-09 中国科学院高能物理研究所 一种两亲性聚合物以及由其形成的磁性胶束纳米载体和其用途
CN106519221B (zh) * 2015-09-10 2019-04-26 中国科学院高能物理研究所 一种聚乙二醇/聚酰胺胺共聚物、其制备方法及包含该共聚物的两亲性siRNA载体
CN105866311B (zh) * 2016-05-25 2017-05-31 福建出入境检验检疫局检验检疫技术中心 测定鸡肉中抗病毒药物残留的uplc‑ms/ms方法
US20200079785A1 (en) 2016-11-08 2020-03-12 Mallinckrodt Llc Mitomycin c prodrug liposome formulations and uses thereof
EP3672969B1 (en) 2017-08-22 2023-07-05 Dynavax Technologies Corporation Alkyl chain modified imidazoquinoline derivatives as tlr7/8 agonists and uses thereof
US10722591B2 (en) 2017-11-14 2020-07-28 Dynavax Technologies Corporation Cleavable conjugates of TLR7/8 agonist compounds, methods for preparation, and uses thereof
CN113453668A (zh) * 2019-01-11 2021-09-28 利普麦迪克斯制药有限公司 包含丝裂霉素c的脂质体前药的脂质体组合物及其制备方法
CA3134791A1 (en) 2019-04-10 2020-10-15 Cellestia Biotech Ag Inhibitors of notch signalling pathway and use thereof in treatment of cancers
EP4008324A1 (en) 2020-12-07 2022-06-08 Cellestia Biotech AG Combinations comprising an inhibitor of an anti-apoptotic protein, such as bcl-2, bcl-xl, bclw or mcl-1, and a notch signaling pathway inhibitor for treating cancer
US20240041884A1 (en) 2020-12-07 2024-02-08 Cellestia Biotech Ag Pharmaceutical Combinations for Treating Cancer
CN117794536A (zh) 2021-06-02 2024-03-29 塞莱斯蒂亚生物技术股份公司 在治疗自身免疫性和炎症性疾病的方法中使用的化合物
WO2023079132A1 (en) 2021-11-08 2023-05-11 Cellestia Biotech Ag Pharmaceutical combinations for treating cancer
EP4223292A1 (en) 2022-02-07 2023-08-09 Cellestia Biotech AG Pharmaceutical combinations for treating cancer

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2254336A1 (US06605299-20030812-C00002.png) * 1973-12-17 1975-07-11 Kyowa Hakko Kogyo Kk
EP0317957A2 (en) * 1987-11-23 1989-05-31 Bristol-Myers Squibb Company Drug-monoclonal antibody conjugates
EP0317956A2 (en) * 1987-11-23 1989-05-31 Bristol-Myers Squibb Company Anti-tumor prodrugs
EP0510197A1 (en) * 1990-01-11 1992-10-28 Nippon Shinyaku Company, Limited Fat emulsion
US5169934A (en) * 1990-05-14 1992-12-08 Anergen, Inc. Intracellularly cleavable compounds
WO1997036904A1 (fr) * 1996-04-02 1997-10-09 Sagami Chemical Research Center Derive de la mitomycine c et inhibiteur des tyrosine kinases non receptrices
WO1999029302A1 (en) * 1997-12-05 1999-06-17 Katarina Edwards Drug delivery system with two-step targeting

Family Cites Families (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
GB8430252D0 (en) 1984-11-30 1985-01-09 Beecham Group Plc Compounds
US5059421A (en) 1985-07-26 1991-10-22 The Liposome Company, Inc. Preparation of targeted liposome systems of a defined size distribution
US4766106A (en) 1985-06-26 1988-08-23 Cetus Corporation Solubilization of proteins for pharmaceutical compositions using polymer conjugation
US4917888A (en) 1985-06-26 1990-04-17 Cetus Corporation Solubilization of immunotoxins for pharmaceutical compositions using polymer conjugation
JPH0615532B2 (ja) * 1986-02-03 1994-03-02 テルモ株式会社 5−フルオロウラシル誘導体およびこれを含有する医薬製剤
US4766105A (en) * 1986-10-31 1988-08-23 Shell Oil Company Ethylene oxide catalyst and process for preparing the catalyst
JPH01113391A (ja) * 1987-10-24 1989-05-02 Kyowa Hakko Kogyo Co Ltd マイトマイシン誘導体
US5103556A (en) 1988-05-05 1992-04-14 Circon Corporation Method of manufacturing an electrohydraulic probe
US4902502A (en) 1989-01-23 1990-02-20 Cetus Corporation Preparation of a polymer/interleukin-2 conjugate
US5585112A (en) 1989-12-22 1996-12-17 Imarx Pharmaceutical Corp. Method of preparing gas and gaseous precursor-filled microspheres
CA2132711C (en) 1992-03-23 2005-02-08 Aquilar Rahman Liposome encapsulated paclitaxel and a method of using the same
JP2813511B2 (ja) * 1992-06-10 1998-10-22 大日本スクリーン製造株式会社 ロールコータによる基板表面への塗液塗布方法
AU5092893A (en) 1992-09-02 1994-03-29 Georgetown University Method of encapsulating anthracycline glycosides in liposomes
US5395619A (en) 1993-03-03 1995-03-07 Liposome Technology, Inc. Lipid-polymer conjugates and liposomes
US5820873A (en) * 1994-09-30 1998-10-13 The University Of British Columbia Polyethylene glycol modified ceramide lipids and liposome uses thereof
CA2210500A1 (en) * 1995-01-16 1996-07-25 Commonwealth Scientific And Industrial Research Organisation Therapeutic compound - fatty acid conjugates
AU4857796A (en) 1995-01-20 1996-08-07 Florida State University Sex-specific dna probe for parrots, methods and kits
JP2001503396A (ja) 1996-10-11 2001-03-13 アルザ コーポレイション 治療用リポソーム組成物および方法
TW520297B (en) * 1996-10-11 2003-02-11 Sequus Pharm Inc Fusogenic liposome composition and method
JPH1160499A (ja) 1997-08-22 1999-03-02 Hiroshi Maeda 抗腫瘍剤
US6180095B1 (en) * 1997-12-17 2001-01-30 Enzon, Inc. Polymeric prodrugs of amino- and hydroxyl-containing bioactive agents
PL196533B1 (pl) * 1998-04-28 2008-01-31 Applied Research Systems Sposób stopniowego przyłączania ugrupowań poli (glikolu etylenowego) (PEG) szeregowo do polipeptydu
JP2002542386A (ja) 1999-04-23 2002-12-10 アルザ・コーポレーション 放出可能な結合及びこれを含む組成物
EP1579874A3 (en) * 1999-04-23 2006-01-25 ALZA Corporation Conjugate having a cleavable linkage for use in a liposome
US7303760B2 (en) 1999-04-23 2007-12-04 Alza Corporation Method for treating multi-drug resistant tumors
US7112337B2 (en) 1999-04-23 2006-09-26 Alza Corporation Liposome composition for delivery of nucleic acid
US7238368B2 (en) 1999-04-23 2007-07-03 Alza Corporation Releasable linkage and compositions containing same
AU7868400A (en) 1999-10-08 2001-04-23 Alza Corporation Neutral-cationic lipid for nucleic acid and drug delivery
ES2367891T3 (es) 2000-09-29 2011-11-10 Schering Corporation Interleucina-10 pegilada.
US7260330B2 (en) * 2002-11-04 2007-08-21 The Boeing Company Optical communication system using correlation receiver

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2254336A1 (US06605299-20030812-C00002.png) * 1973-12-17 1975-07-11 Kyowa Hakko Kogyo Kk
EP0317957A2 (en) * 1987-11-23 1989-05-31 Bristol-Myers Squibb Company Drug-monoclonal antibody conjugates
EP0317956A2 (en) * 1987-11-23 1989-05-31 Bristol-Myers Squibb Company Anti-tumor prodrugs
EP0510197A1 (en) * 1990-01-11 1992-10-28 Nippon Shinyaku Company, Limited Fat emulsion
US5169934A (en) * 1990-05-14 1992-12-08 Anergen, Inc. Intracellularly cleavable compounds
WO1997036904A1 (fr) * 1996-04-02 1997-10-09 Sagami Chemical Research Center Derive de la mitomycine c et inhibiteur des tyrosine kinases non receptrices
WO1999029302A1 (en) * 1997-12-05 1999-06-17 Katarina Edwards Drug delivery system with two-step targeting

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
DATABASE WPI Section Ch, Week 198923 Derwent Publications Ltd., London, GB; Class B02, AN 1989-170530 XP002159819 & JP 01 113391 A (KYOWA HAKKO KOGYO KK), 2 May 1989 (1989-05-02) *
DIAZ C ET AL: "Synthesis of disulfide-containing phospholipid analogs for the preparation of head group-specific lipid antigens: generation of phosphatidylserine antibodies" BIOCONJUGATE CHEMISTRY,US,AMERICAN CHEMICAL SOCIETY, WASHINGTON, vol. 9, no. 2, March 1998 (1998-03), pages 250-254, XP002125967 ISSN: 1043-1802 *
VAAGE J ET AL: "THERAPY OF PRIMARY AND METASTATIC MOUSE MAMMARY CARCINOMAS WITH DOXORUBICIN ENCAPSULATED IN LONG CIRCULATING LIPOSOMES" INTERNATIONAL JOURNAL OF CANCER, vol. 51, no. 6, 1992, pages 942-948, XP000979548 ISSN: 0020-7136 *

Cited By (81)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7608687B2 (en) 1999-04-23 2009-10-27 Alza Corporation Releasable linkage and compositions containing same
US7285622B2 (en) 1999-04-23 2007-10-23 Alza Corporation Releasable linkage and compositions containing same
US7238368B2 (en) 1999-04-23 2007-07-03 Alza Corporation Releasable linkage and compositions containing same
US7303760B2 (en) 1999-04-23 2007-12-04 Alza Corporation Method for treating multi-drug resistant tumors
US7592307B2 (en) 1999-04-23 2009-09-22 Alza Corporation Releasable linkage and compositions containing same
US7276248B2 (en) 1999-04-23 2007-10-02 Alza Corporation Conjugate having a cleavable linkage for use in a liposome
US7108863B2 (en) 2001-03-26 2006-09-19 Alza Corporation Liposome composition for improved intracellular delivery of a therapeutic agent
EP1385479A4 (en) * 2001-03-26 2006-12-06 Alza Corp LIPOSOMIC COMPOSITION FOR IMPROVED INTRA-CELLULAR RELEASE OF A THERAPEUTIC AGENT
EP1385479A1 (en) * 2001-03-26 2004-02-04 Alza Corporation Liposome composition for improved intracellular delivery of a therapeutic agent
US7211440B2 (en) 2002-03-08 2007-05-01 Wallac Oy Dissociative fluorescence enhancement assay
US7682627B2 (en) * 2002-12-03 2010-03-23 Blanchette Rockefeller Neurosciences Institute Artificial low-density lipoprotein carriers for transport of substances across the blood-brain barrier
JP2012131799A (ja) * 2002-12-03 2012-07-12 Blanchette Rockefeller Neurosciences Inst 物質を血液−脳関門を渡って輸送するための人工低密度リポタンパク質キャリア
CN101284136A (zh) * 2002-12-03 2008-10-15 布朗歇特洛克菲勒神经科学研究所 传输物质穿过血脑屏障的人工低密度脂蛋白载体
US7576055B2 (en) * 2002-12-03 2009-08-18 Blanchette Rockefeller Neurosciences Institute Artificial low-density lipoprotein carriers for transport of substances across the blood-brain barrier
AU2004247004B2 (en) * 2003-04-30 2010-10-14 Alza Corporation Mitomycin conjugates cleavable by thiols
WO2004110497A2 (en) 2003-04-30 2004-12-23 Alza Corporation Mitomycin conjugates cleavable by thiols
WO2004110497A3 (en) * 2003-04-30 2005-03-24 Alza Corp Mitomycin conjugates cleavable by thiols
WO2005053749A3 (en) * 2003-11-26 2006-11-09 Alza Corp Thiol-cleavable linkage between polymer and ligand
WO2005053749A2 (en) * 2003-11-26 2005-06-16 Alza Corporation Thiol-cleavable linkage between polymer and ligand
US10576166B2 (en) 2009-12-01 2020-03-03 Translate Bio, Inc. Liver specific delivery of messenger RNA
US10507183B2 (en) 2011-06-08 2019-12-17 Translate Bio, Inc. Cleavable lipids
US10413618B2 (en) 2011-06-08 2019-09-17 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US9597413B2 (en) 2011-06-08 2017-03-21 Shire Human Genetic Therapies, Inc. Pulmonary delivery of mRNA
US11291734B2 (en) 2011-06-08 2022-04-05 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11234936B2 (en) 2011-06-08 2022-02-01 Translate Bio, Inc. Cleavable lipids
US9717690B2 (en) 2011-06-08 2017-08-01 Rana Therapeutics, Inc. Cleavable lipids
US11185595B2 (en) 2011-06-08 2021-11-30 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11951179B2 (en) 2011-06-08 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US11547764B2 (en) 2011-06-08 2023-01-10 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US11052159B2 (en) 2011-06-08 2021-07-06 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11951180B2 (en) 2011-06-08 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US10888626B2 (en) 2011-06-08 2021-01-12 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11951181B2 (en) 2011-06-08 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US10702478B2 (en) 2011-06-08 2020-07-07 Translate Bio, Inc. Cleavable lipids
US10507249B2 (en) 2011-06-08 2019-12-17 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11338044B2 (en) 2011-06-08 2022-05-24 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11730825B2 (en) 2011-06-08 2023-08-22 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US10350303B1 (en) 2011-06-08 2019-07-16 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US10238754B2 (en) 2011-06-08 2019-03-26 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US9308281B2 (en) 2011-06-08 2016-04-12 Shire Human Genetic Therapies, Inc. MRNA therapy for Fabry disease
US10137087B2 (en) 2012-03-29 2018-11-27 Translate Bio, Inc. Lipid-derived neutral nanoparticles
US11497716B2 (en) 2012-03-29 2022-11-15 Translate Bio, Inc. Lipid-derived neutral nanoparticles
US9877919B2 (en) 2012-03-29 2018-01-30 Translate Bio, Inc. Lipid-derived neutral nanoparticles
US10786455B2 (en) 2012-03-29 2020-09-29 Translate Bio, Inc. Lipid-derived neutral nanoparticles
US10137086B2 (en) 2012-03-29 2018-11-27 Translate Bio, Inc. Lipid-derived neutral nanoparticles
US11254936B2 (en) 2012-06-08 2022-02-22 Translate Bio, Inc. Nuclease resistant polynucleotides and uses thereof
US10876104B2 (en) 2013-03-14 2020-12-29 Translate Bio, Inc. Methods for purification of messenger RNA
US11510937B2 (en) 2013-03-14 2022-11-29 Translate Bio, Inc. CFTR MRNA compositions and related methods and uses
US9713626B2 (en) 2013-03-14 2017-07-25 Rana Therapeutics, Inc. CFTR mRNA compositions and related methods and uses
US9181321B2 (en) 2013-03-14 2015-11-10 Shire Human Genetic Therapies, Inc. CFTR mRNA compositions and related methods and uses
US11820977B2 (en) 2013-03-14 2023-11-21 Translate Bio, Inc. Methods for purification of messenger RNA
US10420791B2 (en) 2013-03-14 2019-09-24 Translate Bio, Inc. CFTR MRNA compositions and related methods and uses
US9957499B2 (en) 2013-03-14 2018-05-01 Translate Bio, Inc. Methods for purification of messenger RNA
US11692189B2 (en) 2013-03-14 2023-07-04 Translate Bio, Inc. Methods for purification of messenger RNA
US10052284B2 (en) 2013-10-22 2018-08-21 Translate Bio, Inc. Lipid formulations for delivery of messenger RNA
US10959953B2 (en) 2013-10-22 2021-03-30 Translate Bio, Inc. Lipid formulations for delivery of messenger RNA
US9629804B2 (en) 2013-10-22 2017-04-25 Shire Human Genetic Therapies, Inc. Lipid formulations for delivery of messenger RNA
US11890377B2 (en) 2013-10-22 2024-02-06 Translate Bio, Inc. Lipid formulations for delivery of messenger RNA
US9522176B2 (en) 2013-10-22 2016-12-20 Shire Human Genetic Therapies, Inc. MRNA therapy for phenylketonuria
US10493031B2 (en) 2013-10-22 2019-12-03 Translate Bio, Inc. Lipid formulations for delivery of messenger RNA
US10208295B2 (en) 2013-10-22 2019-02-19 Translate Bio, Inc. MRNA therapy for phenylketonuria
US11377642B2 (en) 2013-10-22 2022-07-05 Translate Bio, Inc. mRNA therapy for phenylketonuria
US11224642B2 (en) 2013-10-22 2022-01-18 Translate Bio, Inc. MRNA therapy for argininosuccinate synthetase deficiency
US11884692B2 (en) 2014-04-25 2024-01-30 Translate Bio, Inc. Methods for purification of messenger RNA
US9850269B2 (en) 2014-04-25 2017-12-26 Translate Bio, Inc. Methods for purification of messenger RNA
US11059841B2 (en) 2014-04-25 2021-07-13 Translate Bio, Inc. Methods for purification of messenger RNA
US10155785B2 (en) 2014-04-25 2018-12-18 Translate Bio, Inc. Methods for purification of messenger RNA
US10286083B2 (en) 2014-05-30 2019-05-14 Translate Bio, Inc. Biodegradable lipids for delivery of nucleic acids
US11433144B2 (en) 2014-05-30 2022-09-06 Translate Bio, Inc. Biodegradable lipids for delivery of nucleic acids
US10912844B2 (en) 2014-05-30 2021-02-09 Translate Bio, Inc. Biodegradable lipids for delivery of nucleic acids
US10493166B2 (en) 2014-05-30 2019-12-03 Translate Bio, Inc. Biodegradable lipids for delivery of nucleic acids
US10293057B2 (en) 2014-05-30 2019-05-21 Translate Bio, Inc. Biodegradable lipids for delivery of nucleic acids
US10286082B2 (en) 2014-05-30 2019-05-14 Translate Bio, Inc. Biodegradable lipids for delivery of nucleic acids
US10022455B2 (en) 2014-05-30 2018-07-17 Translate Bio, Inc. Biodegradable lipids for delivery of nucleic acids
US10138213B2 (en) 2014-06-24 2018-11-27 Translate Bio, Inc. Stereochemically enriched compositions for delivery of nucleic acids
US11104652B2 (en) 2014-06-24 2021-08-31 Translate Bio, Inc. Stereochemically enriched compositions for delivery of nucleic acids
GB2552301A (en) * 2016-07-11 2018-01-24 Evox Therapeutics Ltd Metabolic drug loading of EVs
US11253605B2 (en) 2017-02-27 2022-02-22 Translate Bio, Inc. Codon-optimized CFTR MRNA
US11173190B2 (en) 2017-05-16 2021-11-16 Translate Bio, Inc. Treatment of cystic fibrosis by delivery of codon-optimized mRNA encoding CFTR
WO2019016208A1 (en) * 2017-07-17 2019-01-24 Technische Universiteit Eindhoven APPLICABLE CHEMICAL COMPOSITION COMPRISING AN AGENT CONJUGATED TO A HYDROPHOBIC PART AND A SUPPORT
US11174500B2 (en) 2018-08-24 2021-11-16 Translate Bio, Inc. Methods for purification of messenger RNA

Also Published As

Publication number Publication date
US7285622B2 (en) 2007-10-23
HUP0201425A2 (hu) 2002-12-28
MXPA01010751A (es) 2002-05-14
ZA200108724B (en) 2002-10-23
CA2369595C (en) 2010-10-05
KR100669053B1 (ko) 2007-01-15
CA2369595A1 (en) 2000-11-02
ES2272280T3 (es) 2007-05-01
JP2002542302A (ja) 2002-12-10
US6342244B1 (en) 2002-01-29
EP1173222B1 (en) 2006-09-27
CN100512879C (zh) 2009-07-15
KR100642955B1 (ko) 2006-11-10
US20080152702A1 (en) 2008-06-26
DK1173222T3 (da) 2007-01-29
HK1041820B (zh) 2007-03-30
WO2000064484A3 (en) 2001-11-15
NO20015144L (no) 2001-12-11
US20030054028A1 (en) 2003-03-20
HUP0200797A2 (hu) 2002-07-29
ZA200108726B (en) 2003-03-05
EP1579874A2 (en) 2005-09-28
US20050271715A1 (en) 2005-12-08
HUP0200797A3 (en) 2010-01-28
IL146055A (en) 2006-12-31
IL146055A0 (en) 2002-07-25
CN1355713A (zh) 2002-06-26
US20030211079A1 (en) 2003-11-13
KR20020005006A (ko) 2002-01-16
AU4657700A (en) 2000-11-10
ATE340592T1 (de) 2006-10-15
HK1041820A1 (en) 2002-07-26
US7608687B2 (en) 2009-10-27
CN1244376C (zh) 2006-03-08
DE60030965D1 (de) 2006-11-09
US6365179B1 (en) 2002-04-02
NO20015144D0 (no) 2001-10-22
US20020128195A1 (en) 2002-09-12
CN1399561A (zh) 2003-02-26
MXPA01010750A (es) 2003-08-20
KR20010114247A (ko) 2001-12-31
US6605299B2 (en) 2003-08-12
EP1579874A3 (en) 2006-01-25
AU769425B2 (en) 2004-01-29
DE60030965T2 (de) 2007-05-24
US6984396B2 (en) 2006-01-10
US6849270B2 (en) 2005-02-01
EP1880736A1 (en) 2008-01-23
US20050123597A1 (en) 2005-06-09
US7276248B2 (en) 2007-10-02
HUP0201425A3 (en) 2006-07-28
EP1173222A2 (en) 2002-01-23
JP4558952B2 (ja) 2010-10-06

Similar Documents

Publication Publication Date Title
US6365179B1 (en) Conjugate having a cleavable linkage for use in a liposome
US7238368B2 (en) Releasable linkage and compositions containing same
AU2002305094B2 (en) Liposome composition for improved intracellular delivery of a therapeutic agent
KR100617921B1 (ko) 리포솜 조성물 및 퀴놀론의 투여 방법
CA2067178C (en) Solid tumor treatment method and composition
US20040022842A1 (en) Liposome preparations containing oxaliplatin
Karathanasis et al. Preparation of in vivo cleavable agglomerated liposomes suitable for modulated pulmonary drug delivery
AU2004247004B2 (en) Mitomycin conjugates cleavable by thiols
AU770390B2 (en) Releasable linkage and compositions containing same
JPH06228012A (ja) リポソーム製剤
WO2005069750A2 (en) Therapeutic composition with nanoscale activation agents

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 00807901.3

Country of ref document: CN

AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
ENP Entry into the national phase

Ref document number: 2369595

Country of ref document: CA

Ref document number: 2369595

Country of ref document: CA

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2000 613474

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2000928321

Country of ref document: EP

Ref document number: 2001/08726

Country of ref document: ZA

Ref document number: 514991

Country of ref document: NZ

Ref document number: PA/a/2001/010750

Country of ref document: MX

Ref document number: 46577/00

Country of ref document: AU

Ref document number: 1020017013571

Country of ref document: KR

Ref document number: 200108726

Country of ref document: ZA

AK Designated states

Kind code of ref document: A3

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

WWP Wipo information: published in national office

Ref document number: 1020017013571

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2000928321

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWG Wipo information: grant in national office

Ref document number: 46577/00

Country of ref document: AU

WWG Wipo information: grant in national office

Ref document number: 2000928321

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 1020017013571

Country of ref document: KR