US20030215490A1 - Therapeutic liposome composition and method of preparation - Google Patents

Therapeutic liposome composition and method of preparation Download PDF

Info

Publication number
US20030215490A1
US20030215490A1 US10/115,566 US11556602A US2003215490A1 US 20030215490 A1 US20030215490 A1 US 20030215490A1 US 11556602 A US11556602 A US 11556602A US 2003215490 A1 US2003215490 A1 US 2003215490A1
Authority
US
United States
Prior art keywords
receptor
composition
group
growth factor
targeting
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/115,566
Inventor
Theresa Allen
Paul Uster
Francis Martin
Samuel Zalipsky
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Alza Corp
Original Assignee
Sequus Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=46203431&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20030215490(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from US08/949,046 external-priority patent/US5891468A/en
Application filed by Sequus Pharmaceuticals Inc filed Critical Sequus Pharmaceuticals Inc
Priority to US10/115,566 priority Critical patent/US20030215490A1/en
Publication of US20030215490A1 publication Critical patent/US20030215490A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • 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
    • A61K47/56Medicinal 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
    • A61K47/61Medicinal 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 the organic macromolecular compound being a polysaccharide or a derivative thereof
    • 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
    • A61K47/62Medicinal 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 a protein, peptide or polyamino 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
    • A61K47/68Medicinal 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 antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal 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 antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal 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 antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • 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
    • 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
    • A61K47/6913Medicinal 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 the liposome being modified on its surface by an antibody
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/12Preparations containing radioactive substances for use in therapy or testing in vivo characterised by a special physical form, e.g. emulsion, microcapsules, liposomes, characterized by a special physical form, e.g. emulsions, dispersions, microcapsules
    • A61K51/1217Dispersions, suspensions, colloids, emulsions, e.g. perfluorinated emulsion, sols
    • A61K51/1234Liposomes
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • A61K9/1272Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers with substantial amounts of non-phosphatidyl, i.e. non-acylglycerophosphate, surfactants as bilayer-forming substances, e.g. cationic lipids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2851Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the lectin superfamily, e.g. CD23, CD72
    • C07K16/2854Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the lectin superfamily, e.g. CD23, CD72 against selectins, e.g. CD62
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • 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
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S424/00Drug, bio-affecting and body treating compositions
    • Y10S424/812Liposome comprising an antibody, antibody fragment, antigen, or other specific or nonspecific immunoeffector
    • 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
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S436/00Chemistry: analytical and immunological testing
    • Y10S436/829Liposomes, e.g. encapsulation

Definitions

  • the present invention relates to a target-cell sensitized therapeutic liposome composition and to a method of preparing the composition.
  • a library for preparation of the composition is also described.
  • Liposomes spherical, self-enclosed vesicles composed of amphipathic lipids, have been widely studied and are employed as vehicles for in vivo administration of therapeutic agents.
  • the so-called long circulating liposomes formulations which avoid uptake by the organs of the mononuclear phagocyte system, primarily the liver and spleen, have found commercial applicability.
  • Such long-circulating liposomes include a surface coat of flexible water soluble polymer chains, which act to prevent interaction between the liposome and the plasma components which play a role in liposome uptake.
  • targeting ligands such as an antibody
  • This approach where the targeting ligand is bound to the polar head group residues of liposomal lipid components, results in interference by the surface-grafted polymer chains, inhibiting the interaction between the bound ligand and its intended target (Klibanov, A. L., et al., Biochim. Biophys. Acta., 1062:142-148 (1991); Hansen, C. B., et al., Biochim. Biophys. Acta, 1239:133-144 (1995)).
  • the targeting ligand is attached to the free ends of the polymer chains forming the surface coat on the liposomes (Allen. T. M., et al., Biochim. Biophys. Acta, 1237:99-108 (1995); Blume, G., et al., Biochim. Biophys. Acta, 1149:180-184 (1993)).
  • Two approaches have been described for preparing a liposome having a targeting ligand attached to the distal end of the surface polymer chains.
  • One approach involves preparation of lipid vesicles which include an end-functionalized lipid-polymer derivative; that is, a lipid-polymer conjugate where the free polymer end is reactive or “activated”.
  • Such an activated conjugate is included in the liposome composition and the activated polymer ends are reacted with a targeting ligand after liposome formation.
  • the disadvantage to this approach is the difficulty in reacting all of the activated ends with a ligand.
  • the approach also requires a subsequent step for separation of the unreacted ligand from the liposome composition.
  • the lipid-polymer-ligand conjugate is included in the lipid composition at the time of liposome formation.
  • This approach has the disadvantage that some of the valuable ligand faces the inner aqueous compartment of the liposome and is unavailable for interaction with the intended target.
  • the invention includes a therapeutic liposome composition sensitized to a target cell, comprising
  • a liposomal composition composed of pre-formed liposomes having an entrapped therapeutic agent; and (ii) a plurality of conjugates, each conjugate composed of (a) a lipid having a polar head group and a hydrophobic tail, (b) a hydrophilic polymer having a proximal end and a distal end, where the polymer is attached at its proximal end to the head group of the lipid, and (c) a targeting ligand attached to the distal end of the polymer.
  • the therapeutic, target-cell sensitized liposome composition is formed by combining the liposomal composition with a conjugate selected from the plurality of conjugates.
  • the targeting ligand is an antibody or an antibody fragment.
  • the antibody or antibody fragment is of mouse origin and is humanized to remove murine epitopes.
  • the targeting ligand specifically binds to an extracellular domain of a growth factor receptor.
  • a growth factor receptor is selected from c-erbB-2 protein product of the HER2/neu oncogene, epidermal growth factor receptor, basic fibroblast growth factor receptor, and vascular endothelial growth factor receptor.
  • the targeting ligand binds a receptor selected from E-selectin receptor, L-selectin receptor, P-selectin receptor, folate receptor, CD4 receptor, CD19 receptor, ⁇ integrin receptors and chemokine receptors.
  • the targeting ligand can also be folic acid, pyridoxal phosphate, vitamin B12, sialyl Lewis x , transferrin, epidermal growth factor, basic fibroblast growth factor, vascular endothelial growth factor, VCAM-1, ICAM-1, PECAM-1, an RGD peptide or an NGR peptide.
  • the hydrophilic polymer surrounding the preformed liposomes is selected from the group consisting of polyvinylpyrrolidone, polyvinylmethylether, polymethyloxazoline, polyethyloxazoline, polyhydroxypropyloxazoline, polyhydroxypropylmethacrylamide, polymethacrylamide, polydimethylacrylamide, polyhydroxypropylmethacrylate, polyhydroxyethylacrylate, hydroxymethylcellulose, hydroxyethylcellulose polyethyleneglycol, polyaspartamide and hydrophilic peptide sequences.
  • the hydrophilic polymer is polyethylene glycol of molecular weight between 500-5,000 daltons.
  • the entrapped therapeutic agent is, in one embodiment, a cytotoxic drug.
  • the drug can be an anthracycline antibiotic selected from doxorubicin, daunorubicin, epirubicin and idarubicin and analogs thereof.
  • the cytotoxic agent can also be a platinum compound selected from cisplatin, carboplatin, ormaplatin, oxaliplatin, zeniplatin, enloplatin, lobaplatin, spiroplatin, (( ⁇ )-(R)-2-aminomethylpyrrolidine (1,1-cyclobutane dicarboxylato)platinum), (SP-4-3(R)-1,1-cyclobutane-dicarboxylato(2-)-(2-methyl-1,4-butanediamine-N,N′)platinum), nedaplatin and (bis-acetato-ammine-dichloro-cyclohexylamine-platinum(IV)).
  • platinum compound selected from cisplatin, carboplatin, ormaplatin, oxaliplatin, zeniplatin, enloplatin, lobaplatin, spiroplatin, (( ⁇ )-(R)-2-aminomethylpyrrolidine (1,1-cycl
  • the cytotoxic agent is a topoisomerase 1 inhibitor selected from the group consisting of topotecan, irinotecan, (7-(4-methylpiperazino-methylene)-10,11-ethylenedioxy-20(S)-camptothecin), 7-(2-(N-isopropylamino)ethyl)-(20S)-camptothecin, 9-aminocamptothecin and 9-nitrocamptothecin.
  • the cytotoxic agent is a vinca alkaloid selected from the group consisting of vincristine, vinblastine, vinleurosine, vinrodisine, vinorelbine and vindesine.
  • the entrapped agent is a nucleic acid.
  • the nucleic acid can be an antisense oligonucleotide or ribozyme or a plasmid containing a therapeutic gene which when internalized by the target cells achieves expression of the therapeutic gene to produce a therapeutic gene product.
  • the invention includes a plurality of targeting conjugates for use in preparing a targeted, therapeutic liposome composition.
  • Each conjugate is composed of a (i) a lipid having a polar head group and a hydrophobic tail, (ii) a hydrophilic polymer having a proximal end and a distal end, the polymer attached at its proximal end to the head group of the lipid, and (iii) a targeting ligand attached to the distal end of the polymer.
  • the lipid in the conjugates is, in one embodiment, distearoyl phosphatidylethanolamine, distearoyl-phosphatidylcholine, monogalactosyl diacylglycerols or digalactosyl diacylglycerols.
  • the hydrophilic polymer in the conjugates is selected from the group consisting of polyvinylpyrrolidone, polyvinylmethylether, polymethyloxazoline, polyethyloxazoline, polyhydroxypropyloxazoline, polyhydroxypropylmethacrylamide, polymethacrylamide, polydimethylacrylamide, polyhydroxypropylmethacrylate, polyhydroxyethylacrylate, hydroxymethylcellulose, hydroxyethylcellulose, polyethyleneglycol, polyaspartamide and hydrophilic peptide sequences.
  • the targeting ligand of the conjugates can be any of those recited above.
  • the invention includes a method of formulating a therapeutic liposome composition having sensitivity to a target cell.
  • the method includes the steps of (i) selecting a liposome formulation composed of pre-formed liposomes having an entrapped therapeutic agent; (ii) selecting from a plurality of targeting conjugates a targeting conjugate composed of (a) a lipid having a polar head group and a hydrophobic tail, (b) a hydrophilic polymer having a proximal end and a distal end, where the polymer is attached at its proximal end to the head group of the lipid, and (c) a targeting ligand attached to the distal end of the polymer; and (iii) combining the liposome formulation and the selected targeting conjugate to form said therapeutic, target-cell sensitive liposome composition.
  • combining includes incubating under conditions effective to achieve insertion of the selected targeting conjugate into the liposomes of the selected liposome formulation.
  • selecting a liposome formulation includes determining the sensitivity of the target cell to the therapeutic activity of the entrapped therapeutic agent.
  • selecting a targeting conjugate includes determining the ability of the targeting ligand to bind cell surface receptors expressed on the target cell.
  • selecting a targeting conjugate is based on (i) the ability of a targeting ligand to bind to cell surface receptors expressed on the target cell and (ii) the ability of the target cell to internalize liposomes bound to the target cell by binding between the target cell and the targeting ligand.
  • FIG. 1 illustrates a library composed of a plurality of therapeutic pre-formed liposomes and a plurality of targeting conjugates
  • FIGS. 2 A- 2 D are plots showing the fraction of liposomes (peak centered at fraction 10 ) and the fraction of micellular targeting conjugates (peak centered at fraction 20 ) by size exclusion chromatography from samples taken during incubation of a targeting conjugate sialyl-Lewis x -PEG-DSPE with preformed liposomes at times of 0 hours (FIG. 2A), 1 hour (FIG. 2B), 3 hours (FIG. 2C) and 5 hours (FIG. 2D);
  • FIG. 3 is a plot showing the time course for insertion of the targeting conjugate sialyl-Lewis x -PEG-DSPE into pre-formed liposomes when incubated at 25° C. (closed circles) and 37° C. (open squares);
  • FIG. 4 is a plot showing the blood circulation lifetime of target-cell sensitized liposome prepared in accordance with the invention, where the percent of injected dose in vivo for liposomes having E-selectin Fab fragments targeting ligands (30 ligands per liposome represented by solid triangles, 70 ligands per liposome represented by solid squares) and for liposomes having a surface coating of polyethyleneglycol chains (open circles) as a function of time after dosing; and
  • FIGS. 5 A- 5 B are scanned images of micrographs of blood vessels in a window chamber of a mouse dorsal fold, where FIG. 5A is the control of the untreated blood vessels under transmitted light, and FIG. 5B is a fluorescence micrograph showing binding of fluorsecin-labeled liposomes bearing an E-selectin Fab fragments to endothelial cells in the blood vessels.
  • “Incubating” or “incubating under conditions effective to achieve insertion” refer to conditions of time, temperature and liposome lipid composition which allow for penetration and entry of a selected component, such as a lipid or lipid conjugate, into the lipid bilayer of a liposome.
  • Pre-formed liposomes refers to intact, previously formed unilamellar or multilamellar lipid vesicles.
  • Target-cell sensitized refers to a liposome which includes a ligand or moiety covalently bound to the liposome and having binding affinity for a receptor expressed on a particular cell.
  • “Therapeutic liposome composition” refers to liposomes which include a therapeutic agent entrapped in the aqueous spaces of the liposomes or in the lipid bilayers of the liposomes.
  • Vehicle-forming lipid refers to any lipid capable of forming part of a stable micelle or liposome composition and typically including one or two hydrophobic acyl hydrocarbon chains or a steroid group and may contain a chemically reactive group, such as an amine, acid, ester, aldehyde or alcohol, at its polar head group.
  • the invention includes a kit or “library” for preparation of a therapeutic, target-cell sensitized liposome composition.
  • FIG. 1 shows such a library 10 , where a plurality 12 of targeting conjugates 12 ( a ), 12 ( b ), 12 ( c ), etc. and a plurality 14 of pre-formed therapeutic liposome compositions, such compositions 14 ( a ), 14 ( b ), 14 ( c ) are shown.
  • the targeting conjugates and pre-formed liposome pluralities are shown in suspension form in vials ready for use, however it will be appreciated that other storage forms are contemplated, such as lyophilized or freeze-dried.
  • the targeting conjugates in the library are lipid-polymer-ligand conjugates and will be described in more detail below.
  • the conjugates in the library differ in the targeting ligand attached to the lipid-polymer, as well as in the lipid and polymer components. Exemplary ligands and lipid and polymer components will be set forth below.
  • the pre-formed liposomes in the library are either conventional liposomes containing an entrapped therapeutic agent or are liposomes having a surface coating of hydrophilic polymer chains, as will be described below.
  • the pre-formed liposomes in the library differ from one another generally in the entrapped therapeutic agent and exemplary agents will be set forth below.
  • the pre-formed liposomes can also differ from one another in the liposome lipid components.
  • a therapeutic, target-cell sensitized liposome composition is prepared from the library as follows.
  • a composition specific for a subject suffering from a particular condition for example a solid tumor of the lung, a bacterial infection or a viral infection, is prepared by selecting a targeting conjugate from the library.
  • the targeting conjugate is selected either according to knowledge of those of skill in the art of ligand-receptor binding pairs or by obtaining a suitable patient sample, e.g., a fluid sample, a biopsy or the like.
  • the sample is tested by means known to those in the art for expression of a variety of receptors to determine the appropriate targeting ligand.
  • a pre-formed therapeutic liposome composition is selected based on knowledge of those of skill in the art of the therapeutic agents appropriate for treatment of the particular condition.
  • the therapeutic liposome composition is selected after performing chemosensitivity tests to determine the effect of the entrapped agent on cells of concern obtained from the patient biopsy or fluid sample.
  • the target-cell sensitized, therapeutic liposome composition for the subject is prepared by combining the two components. As will be described, the components are combined under conditions effective to achieve insertion of the targeting conjugate into the liposome bilayer to create the target-cell sensitized liposomes. After insertion is complete, the composition is administered to patient.
  • one component of the kit or library for preparing the composition of the invention is a plurality of pre-formed liposomes having an entrapped therapeutic or diagnostic agent.
  • the liposome lipid components, exemplary agents and methods of preparing the liposomes are described.
  • Liposomes suitable for use in the composition of the present invention include those composed primarily of vesicle-forming lipids.
  • a vesicle-forming lipid is one which (a) can form spontaneously into bilayer vesicles in water, as exemplified by the phospholipids, or (b) is stably incorporated into lipid bilayers, with its hydrophobic moiety in contact with the interior, hydrophobic region of the bilayer membrane, and its head group moiety oriented toward the exterior, polar surface of the membrane.
  • the vesicle-forming lipids of this type are preferably ones having two hydrocarbon chains, typically acyl chains, and a head group, either polar or nonpolar.
  • the vesicle-forming lipids and naturally-occurring vesicle-forming lipids including the phospholipids, such as phosphatidylcholine, phosphatidylethanolamine, phosphatidic acid, phosphatidylinositol, and sphingomyelin, where the two hydrocarbon chains are typically between about 14-22 carbon atoms in length, and have varying degrees of unsaturation.
  • the above-described lipids and phospholipids whose acyl chains have varying degrees of saturation can be obtained commercially or prepared according to published methods.
  • Other suitable lipids include glycolipids, cerebrosides and sterols, such as cholesterol.
  • Cationic lipids are also suitable for use in the liposomes of the invention, where the cationic lipid can be included as a minor component of the lipid composition or as a major or sole component.
  • Such cationic lipids typically have a lipophilic moiety, such as a sterol, an acyl or diacyl chain, and where the lipid has an overall net positive charge.
  • the head group of the lipid carries the positive charge.
  • Exemplary cationic lipids include 1,2-dioleyloxy-3-(trimethylamino) propane (DOTAP); N-[1-(2,3,-ditetradecyloxy)propyl]-N,N-dimethyl-N-hydroxyethylammonium bromide (DMRIE); N-[1-(2,3,-dioleyloxy)propyl]-N,N-dimethyl-N-hydroxy ethylammonium bromide (DORIE); N-[1-(2,3-dioleyloxy) propyl]-N,N,N-trimethylammonium chloride (DOTMA); 3 [N-(N′,N′-dimethylaminoethane) carbamoly] cholesterol (DC-Chol); and dimethyldioctadecylammonium (DDAB).
  • DOTAP 1,2-dioleyloxy-3-(trimethylamino) propane
  • DMRIE N-[1-(2,3,-ditetrade
  • the cationic vesicle-forming lipid may also be a neutral lipid, such as dioleoylphosphatidyl ethanolamine (DOPE) or an amphipathic lipid, such as a phospholipid, derivatized with a cationic lipid, such as polylysine or other polyamine lipids.
  • DOPE dioleoylphosphatidyl ethanolamine
  • amphipathic lipid such as a phospholipid
  • a cationic lipid such as polylysine or other polyamine lipids.
  • DOPE dioleoylphosphatidyl ethanolamine
  • an amphipathic lipid such as a phospholipid
  • a cationic lipid such as polylysine or other polyamine lipids.
  • the neutral lipid (DOPE) can be derivatized with polylysine to form a cationic lipid.
  • the vesicle-forming lipid is selected to achieve a specified degree of fluidity or rigidity, to control the stability of the liposome in serum, to control the conditions effective for insertion of the targeting conjugate, as will be described, and to control the rate of release of the entrapped agent in the liposome.
  • Liposomes having a more rigid lipid bilayer, or a liquid crystalline bilayer are achieved by incorporation of a relatively rigid lipid, e.g., a lipid having a relatively high phase transition temperature, e.g., up to 60° C.
  • a relatively rigid lipid e.g., a lipid having a relatively high phase transition temperature, e.g., up to 60° C.
  • Rigid, i.e., saturated, lipids 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.
  • the targeted, therapeutic liposome composition of the invention is prepared using pre-formed liposomes and a targeting conjugate, which are incubated together under conditions effective to achieve insertion of the conjugate into the liposome bilayer. More specifically, the two components are incubated together under conditions which achieve insertion of the conjugate in such a way that the targeting ligand is oriented outwardly from the liposome surface, and therefore available for interaction with its cognate receptor.
  • Vesicle-forming lipids having phase transition temperatures from approximately 2° C.-80° C. are suitable for use in the pre-formed liposome component of the present composition.
  • the lipid distearyl phosphatidylcholine (DSPC) has a phase transition temperature of 62° C.
  • the lipid hydrogenated soy phosphatidylcholine (HSPC) has a phase transition temperature of 58° C.
  • Phase transition temperatures of many lipids are tabulated in a variety of sources, such as Avanti Polar Lipids catalogue and Lipid Thermotropic Phase Transition Database (LIPIDAT, NIST Standard Reference Database 34).
  • a vesicle-forming lipid having a phase transition temperature between about 30-70° C. is employed.
  • the lipid used in forming the liposomes is one having a phase transition temperature within about 20° C., more preferably 10° C., most preferably 5° C., of the temperature to which the ligand in the targeting conjugate can be heated without affecting its binding activity.
  • the conditions effective to achieve insertion of the targeting conjugate into the liposome are determined based on several variables, including, the desired rate of insertion, where a higher incubation temperature may achieve a faster rate of insertion, the temperature to which the ligand can be safely heated without affecting its activity, and to a lesser degree the phase transition temperature of the lipids and the lipid composition. It will also be appreciated that insertion can be varied by the presence of solvents, such as amphipathic solvents including polyethyleneglycol and ethanol, or detergents.
  • the preformed liposomes also include a vesicle-forming lipid derivatized with a hydrophilic polymer.
  • a vesicle-forming lipid derivatized with a hydrophilic polymer As has been described, for example in U.S. Pat. No. 5,013,556, including such a derivatized lipid in the liposome composition forms a surface coating of hydrophilic polymer chains around the liposome. The surface coating of hydrophilic polymer chains is effective to increase the in vivo blood circulation lifetime of the liposomes when compared to liposomes lacking such a coating.
  • Vesicle-forming lipids suitable for derivatization with a hydrophilic polymer include any of those lipids listed above, and, in particular phospholipids, such as distearoyl phosphatidylethanolamine (DSPE).
  • DSPE distearoyl phosphatidylethanolamine
  • Hydrophilic polymers suitable for derivatization with a vesicle-forming lipid include polyvinylpyrrolidone, polyvinylmethylether, polymethyloxazoline, polyethyloxazoline, polyhydroxypropyloxazoline, polyhydroxypropylmethacrylamide, polymethacrylamide, polydimethylacrylamide, polyhydroxypropylmethacrylate, polyhydroxyethylacrylate, hydroxymethylcellulose, hydroxyethylcellulose, polyethyleneglycol, polyaspartamide and hydrophilic peptide sequences.
  • 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 500-10,000 daltons, more preferably between 1,000-5,000 daltons.
  • PEG polyethyleneglycol
  • Methoxy or ethoxy-capped analogues of PEG are also preferred hydrophilic polymers, commercially available in a variety of polymer sizes, e.g., 120-20,000 daltons.
  • vesicle-forming lipids derivatized with hydrophilic polymers has been described, for example in U.S. Pat. No. 5,395,619.
  • liposomes including such derivatized lipids has also been described, where typically, between 1-20 mole percent of such a derivatized lipid is included in the liposome formulation.
  • the pre-formed liposomes include an agent entrapped in the liposome. Entrapped is intended to include encapsulation of an agent in the aqueous core and aqueous spaces of liposomes as well as entrapment of an agent in the lipid bilayer(s) of the liposomes.
  • Agents contemplated for use in the composition of the invention are widely varied, and include both therapeutic applications and those for use in diagnostic applications.
  • Therapeutic agents include natural and synthetic compounds having the following therapeutic activities:
  • the entrapped agent is a cytotoxic drug, that is, a drug having a deleterious or toxic effect on cells.
  • cytotoxic agents include the anthracycline antibiotics such as doxorubicin, daunorubicin, epirubicin and idarubicin, and analogs of these, such as epirubidin and mitoxantrone; platinum compounds, such as cisplatin, carboplatin, ormaplatin, oxaliplatin, zeniplatin, enloplatin, lobaplatin, spiroplatin, (( ⁇ )-(R)-2-aminomethylpyrrolidine (1,1-cyclobutane dicarboxylato)platinum)(DWA2114R), (SP-4-3(R)-1,1-cyclobutane-dicarboxylato(2-)-(2-methyl-1,4-butanediamine-N,N′)platinum) (CI-973), n
  • vinca alkaloids such as vincristine, vinblastine, vinleurosine, vinrodisine, vinorelbine (navelbine) and vindesine.
  • cytotoxic agents is a topoisomerase I inhibitor, such as camptothecin and its analogues, including SN-38 ((+)-(4S)-4,11-diethyl-4,9-dihydroxy-1H-pyrano[3′,4′:6,7]-indolizino[1,2-b]quinoline-3,14(4H,12H)-dione); 9-aminocamptothecin; 9-nitrocamptothecin, topotecan (hycamtin; 9-dimethyl-aminomethyl-10-hydroxycamptothecin); irinotecan (CPT-11; 7-ethyl-10-[4-(1-piperidino)-1-piperidino]-carbonyloxy-camptothecin), which is hydrolyzed in vivo to SN-38); 7-ethylcamptothecin and its derivatives (Sawada, S.
  • camptothecin and its analogues including
  • the entrapped therapeutic agent is an angiogenesis inhibitor, such as angiostatin, endostatin and TNF ⁇ .
  • the entrapped therapeutic agent in a nucleic acid selected from a variety of DNA and RNA based nucleic acids, including fragments and analogues of these.
  • a variety of genes for treatment of various conditions have been described, and coding sequences for specific genes of interest can be retrieved from DNA sequence databanks, such as GenBank or EMBL.
  • DNA sequence databanks such as GenBank or EMBL.
  • polynucleotides for treatment of viral, malignant and inflammatory diseases and conditions such as, cystic fibrosis, adenosine deaminase deficiency and AIDS.
  • Treatment of cancers by administration of tumor suppressor genes such as APC, DPC4, NF-1, NF-2, MTS1, RB, p53, WT 1 , BRCA1, BRCA2 and VHL, are contemplated.
  • HLA-B7 tumors, colorectal carcinoma, melanoma
  • IL-2 cancers, especially breast cancer, lung cancer, and tumors
  • IL-4 cancer
  • TNF cancer
  • IGF-1 antisense brain tumors
  • IFN neuroblastoma
  • GM-CSF renal cell carcinoma
  • MDR-1 cancer, especially advanced cancer, breast and ovarian cancers
  • HSV thymidine kinase brain tumors, head and neck tumors, mesothelioma, ovarian cancer.
  • the polynucleotide can be an antisense DNA oligonucleotide composed of sequences complementary to its target, usually a messenger RNA (mRNA) or an mRNA precursor.
  • mRNA messenger RNA
  • the mRNA contains genetic information in the functional, or sense, orientation and binding of the antisense oligonucleotide inactivates the intended mRNA and prevents its translation into protein.
  • antisense molecules are determined based on biochemical experiments showing that proteins are translated from specific RNAs and once the sequence of the RNA is known, an antisense molecule that will bind to it through complementary Watson-Crick base pairs can be designed.
  • Such antisense molecules typically contain between 10-30 base pairs, more preferably between 10-25, and most preferably between 15-20.
  • the antisense oligonucleotide can be modified for improved resistance to nuclease hydrolysis, and such analogues include phosphorothioate, methylphosphonate, phosphodiester and p-ethoxy oligonucleotides (WO 97/07784).
  • the entrapped agent can also be a ribozyme or catalytic RNA.
  • the liposomes may be prepared by a variety of techniques, such as those detailed in Szoka, F., Jr., et al., Ann. Rev. Biophys. Bioeng. 9:467 (1980), and specific examples of liposomes prepared in support of the present invention will be described below.
  • the liposomes are multilamellar vesicles (MLVs), which can be formed by simple lipid-film hydration techniques. In this procedure, a mixture of liposome-forming lipids of the type detailed above dissolved in a suitable organic solvent is evaporated in a vessel to form a thin film, which is then covered by an aqueous medium. The lipid film hydrates to form MLVs, typically with sizes between about 0.1 to 10 microns.
  • the preformed liposomes include a vesicle-forming lipid derivatized with a hydrophilic polymer to form a surface coating of hydrophilic polymer chains on the liposomes surface.
  • a coating is preferably prepared by including between 1-20 mole percent of the derivatized lipid with the remaining liposome forming components, e.g., vesicle-forming lipids.
  • Exemplary methods of preparing derivatized lipids and of forming polymer-coated liposomes have been described in co-owned U.S. Pat. Nos. 5,013,556, 5,631,018 and 5,395,619, which are incorporated herein by reference.
  • the hydrophilic polymer may be stably coupled to the lipid, or coupled through an unstable linkage which allows the coated liposomes to shed the coating of polymer chains as they circulate in the bloodstream or in response to a stimulus.
  • the therapeutic or diagnostic agent of choice can be incorporated into liposomes by standard methods, including (i) passive entrapment of a water-soluble compound by hydrating a lipid film with an aqueous solution of the agent, (ii) passive entrapment of a lipophilic compound by hydrating a lipid film containing the agent, and (iii) loading an ionizable drug against an inside/outside liposome pH gradient.
  • Other methods such as reverse evaporation phase liposome preparation, are also suitable.
  • Polynucleotides, oligonucleotides, other nucleic acids, such as a DNA plasmid can be entrapped in the liposome by condensing the nucleic acid in single-molecule form.
  • the nucleic acid is suspended in an aqueous medium containing protamine sulfate, spermine, spermidine, histone, lysine, mixtures thereof, or other suitable polycationic condensing agent, under conditions effective to condense the nucleic acid into small particles.
  • the solution of condensed nucleic acid molecules is used to rehydrate a dried lipid film to form liposomes with the condensed nucleic acid in entrapped form.
  • a similar approach to condensing nucleic acids for entrapment in liposomes is described in co-pending U.S. patent application Ser. No. 09/103,341.
  • the pre-formed liposomes of the invention are preferably prepared to have substantially homogeneous sizes in a selected size range, typically between about 0.01 to 0.5 microns, more preferably between 0.03-0.40 microns.
  • One effective sizing method for REVs and MLVs involves extruding an aqueous suspension of the liposomes through a series of polycarbonate membranes having a selected uniform pore size in the range of 0.03 to 0.2 micron, typically 0.05, 0.08, 0.1, or 0.2 microns.
  • the pore size of the membrane corresponds roughly to the largest sizes of liposomes produced by extrusion through that membrane, particularly where the preparation is extruded two or more times through the same membrane.
  • Homogenization methods are also useful for down-sizing liposomes to sizes of 100 nm or less (Martin, F. J., in SPECIALIZED DRUG DELIVERY SYSTEMS - MANUFACTURING AND PRODUCTION TECHNOLOGY, (P. Tyle, Ed.) Marcel Dekker, New York, pp. 267-316 (1990)).
  • the kit or library of the invention also includes a targeting conjugate, now to be described.
  • the targeting conjugate is composed of (i) a lipid having a polar head group and a hydrophobic tail, e.g., a vesicle-forming lipid and any of those described above are suitable; (ii) a hydrophilic polymer attached to the head group of the vesicle-forming lipid, and any of the polymers recited above are suitable; and (iii) a targeting ligand attached to the polymer.
  • the targeting ligand for use in the conjugate can be selected from a wide variety of moieties capable of targeting the pre-formed liposomes to a selected cell or tissue.
  • suitable ligands suitable are listed in Table 1.
  • TABLE 1 LIGAND-RECEPTOR PAIRS AND ASSOCIATED TARGET CELL LIGAND RECEPTOR CELL TYPE Folate folate receptor epithelial carcinomas, bone arrow stem cells water soluble vitamins vitamin receptor various cells Pyridoxyl phosphate CD4 CD4 + lymphocytes Apolipoproteins LDL liver hepatocytes, vascular endothelial cells Insulin insulin receptor pancreatic islet cells Transferrin Transferrin receptor endothelial cells (brain) Galactose Asialoglycoprotein liver hepatocytes receptor Sialyl-Lewis x E, P selectin activated endothelial cells Mac-1 L selectin neutrophils, leukocytes VEGF Flk-1, 2 tumor epithelial cells basic FGF FGF
  • One preferred ligand is an antibody or an antibody fragment. It will be appreciated that the antibody or antibody fragment can be of mouse origin and humanized to remove murine surface recognition features.
  • the targeting ligand binds to an extracellular domain of a growth factor receptor.
  • exemplary receptors include the c-erbB-2 protein product of the HER2/neu oncogene, epidermal growth factor (EGF) receptor, basic fibroblast growth receptor (basic FGF) receptor and vascular endothelial growth factor receptor, E-, L- and P-selectin receptors, folate receptor, CD4 receptor, CD19 receptor, ⁇ integrin receptors and chemokine receptors.
  • the targeting ligand is covalently attached to the free distal end of the hydrophilic polymer chain, which is attached at its proximal end to a vesicle-forming lipid.
  • the hydrophilic polymer polyethyleneglycol (PEG) has been widely studied (Allen, T.
  • the PEG chains are functionalized to contain reactive groups suitable for coupling with, for example, sulfhydryls, amino groups, and aldehydes or ketones (typically derived from mild oxidation of carbohydrate portions of an antibody) present in a wide variety of ligands (see Table 1).
  • PEG-terminal reactive groups examples include maleimide (for reaction with sulfhydryl groups), N-hydroxysuccinimide (NHS) or NHS-carbonate ester (for reaction with primary amines), hydrazide or hydrazine (for reaction with aldehydes or ketones), iodoacetyl (preferentially reactive with sulfhydryl groups) and dithiopyridine (thiol-reactive).
  • Synthetic reaction schemes for activating PEG with such groups are set forth in U.S. Pat. Nos. 5,631,018, 5,527,528, 5,395,619, and the relevant sections describing synthetic reaction procedures are expressly incorporated herein by reference.
  • hydrophilic polymers recited above in combination with any of the vesicle-forming lipids recited above can be employed for the targeting conjugate and suitable reaction sequences can be determined by those of skill in the art.
  • a pre-formed therapeutic liposome composition and a targeting conjugate are selected from the library.
  • the two components are combined under conditions effective to achieve insertion of the targeting conjugate into the liposome lipid bilayer to form the target-cell sensitized composition.
  • Sialyl-Lewis x can be used to target liposomes to cells expressing endothelial leukocyte adhesion molecule-1 (ELAM-1 or E-selectin) for delivery of a therapeutic agent to a site of inflammation.
  • ELAM-1 endothelial leukocyte adhesion molecule-1
  • ELAM-1 is expressed on the surface of endothelial cells of blood vessels adjacent to sites of inflammation.
  • ELAM-1 recognizes and binds the polysaccharide moiety sialyl-Lewis x which is present on surfaces of neutrophils, and recruits neutrophils to sites of inflammation.
  • Pre-formed liposomes were prepared as described in Example 1 and were composed of partially hydrogenated soy-bean phosphatidylcholine (PHPC), cholesterol and MPEG-DSPE in a molar ratio of 55:40:3.
  • the liposomes were sized to a diameter of about 100 nm.
  • the liposomes were incubated at 37° C. with 1.2 mole percent sialyl-Lewis x -PEG-DSPE targeting conjugate to achieve insertion of the conjugate into the pre-formed liposomes.
  • FIGS. 2 A- 2 D the liposome fraction of the incubation mixture is represented by the peak centered around fraction 10 and the micellular targeting conjugate is represented by the peak centered around fraction 20 .
  • FIG. 2A shows the initial composition mixture, at time zero and FIGS. 2 B- 2 D show the composition after 1, 3 and 5 hours incubation, respectively. Disappearance as a function of incubation time of the conjugate micelles (peak at fraction 20 ) is apparent, indicating insertion of the targeting conjugate into the pre-formed liposomes.
  • FIG. 3 shows the time-course of insertion of sialyl-Lewis x -PEG-DSPE targeting conjugate into PHPC:Chol:mPEG-DSPE (55:40:3) pre-formed liposomes at 37° C. (open squares) and 25° C. (closed circles). Insertion of the conjugate into the pre-formed liposomes proceeded more rapidly at 37° C., however insertion at ambient temperature was also substantial.
  • Table 2 shows the average particle size determined by dynamic light scattering after a 5 hour incubation period at 37° C. of pre-formed liposomes and a targeting conjugate, either YIGSRG-PEG-DSPE or sialyl-Lewis x -PEG-DSPE.
  • the average particle size after incubation and insertion of the targeting conjugate increased only slightly.
  • Liposomes having an E-selectin Fab fragment targeting ligand were prepared in accordance with the invention for in vivo administration to rodents.
  • an anti-E-selectin Fab fragment was conjugated to PEG-DSPE to form an E-selectin Fab-PEG-DSPE targeting conjugate.
  • the targeting conjugate was incubated with pre-formed 111 In-labelled-liposomes composed of partially hydrogenated soy phosphatidylcholine (PHPC), PEG-DSPE and cholesterol in a 55:40:3 molar ratio in an amount sufficient to obtain 12, 20, 33, 40 and 70 Fab residues per 100 nm liposome (Example 2B).
  • the insertion procedure resulted in greater than 95% of the targeting conjugates being inserted into the preformed liposomes.
  • the insertion efficiency is greater than 90%, more preferably greater than 95%.
  • the liposomes containing 30 Fab residues per liposome and 70 Fab residues per liposome were administered to rats to determine the blood circulation lifetime of the liposomes.
  • 111 In-labelled-liposomes of PHPC, cholesterol and PEG-DSPE (molar ratio of 55:40:3) were administered.
  • the results are shown in FIG. 4, where the liposomes having 70 Fab residues per liposome (solid squares) and 30 Fab residues per liposomes (solid triangles) have a pharmacokinetic profile similar to that of the control liposomes (open circles). As seen, 24 hours after administration, nearly 25% of the injected dose remains in circulation in the bloodstream.
  • FIGS. 5 A- 5 B are scanned images of photomicrographs of the blood vessels under transmitted light prior to liposome administration (FIG. 5A) and 5 hours after administration of the target-cell sensitized, fluorescein-labeled liposomes (FIG. 5B).
  • E-selectin Fab liposomes target the endothelial cells along the blood vessels.
  • the appearance of E-selectin antigen peak was around 5 hours after endotoxin treatment, indicating that the binding activity of the E-selectin antibody was retained.
  • a plurality of targeting conjugates and a plurality of liposome formulations with a variety of entrapped therapeutic agents are available for selection according to the indication to be treated.
  • preparation and use of the library will be further demonstrated by describing suitable library components for treatment of an exemplary indication, breast cancer.
  • the library of the invention includes a plurality of targeting conjugates in the form of pre-filled vials containing the conjugate as a purified, sterile micellar suspension in an appropriate buffer.
  • the plurality of targeting conjugates can include the following.
  • the c-erbB-2 receptor of the HER2-neu oncogene is over-expressed in many human breast cancer cells.
  • Humanized monoclonal antibodies have been developed which bind with high affinity to the c- erbB-2 receptor (Baselga J., et al., J. Clin Oncol., 14(3):737-44 (1996)).
  • Single chain sFv fragments of the anti c-erbB-2 C6.5 antibody into which a terminal cysteine group is inserted are obtained as described by Schier et al. ( Immunotechnology, 1(1):73-81 (1995)).
  • the whole c-erbB-2 antibody is conjugated to PEG-DSPE having a reactive hydrazide moiety.
  • the sFv fragment containing the terminal cysteine (and thus a free thiol group) is conjugated to PEG-DSPE-maleimide, under conditions like those described for the conjugation of the anti-E selected Fab′ antibody fragment to the same compound in Example 2.
  • EGFR Epidermal Growth Factor Receptor
  • EGFRvIII a deletion-mutant form of EGFR
  • EGFRvIII a deletion-mutant form of EGFR
  • the RGD sequence is the cell attachment site for proliferating vascular endothelial cells which form the blood supply to tumors (during angiogenesis). Such attachments are mediated by a v integrins expressed by these endothelial cells.
  • the integrin-binding activity of matrix adhesion proteins can be reproduced by short synthetic peptides containing the RGD sequence.
  • Reagents that bind selectively to only one or a few of the RGD-directed integrins can be designed by cyclizing peptides with selected sequences around the RGD and by synthesizing RGD mimics.
  • RGD peptides can be isolated by using phage display peptide libraries (Pasqualini, R., and Ruoslahti, E., Nature, 380(6572):364-6 (1996)). Two of these peptides—one containing an a v integrin-binding Arg-Gly-Asp motif and the other an Asn-Gly-Arg motif—have been identified that bind selectively to tumor vasculature. These can be linked to liposomes using the methods described herein.
  • a plurality of other PEG-DSPE conjugates of ligands such as folate or transferin, which may bind to receptors on human breast cancer cells are prepared according to the examples set herein and by methods known in the art.
  • the library further includes a therapeutic liposome composition or a plurality of liposome compositions containing encapsulated agents appropriate for treating human breast cancer cells in vivo.
  • the pre-formed liposomes are in the form of pre-filled vials containing the liposomes as a sterile suspension in appropriate buffers is created.
  • Liposome containing the following entrapped agents are exemplary for the human breast cancer example: doxorubicin, cisplatin, water-soluble camptothecin derivatives (e.g. topotecan, navelbine, vincristine, antisense oligonucleotides, p53 gene, HSVtk gene, a radiation sensitizer and an angiogenesis inhibitor.
  • a targeting conjugate and a therapeutic liposome composition are selected. Selection of the targeting conjugate is based upon the expression of the conjugate's cognate receptor on individual patient's breast cancer cells. For example, it is common to test for the expression of a variety of receptors on cancer cells obtained from patients during biopsy. Clinical reference laboratories routinely screen biopsy specimens for estrogen receptor status and c-erbB-2 expression status is becoming routine with the clinical development of HERCEPTIN an anti-tumor therapeutic antibody product described by Baselga, et al, ( J. Clin Oncol., ( 3):737-44 (1996)). Exemplary methods for determining c-erbB-2 receptor status are given by Sjogren, et al. ( J.
  • a pre-formed therapeutic liposome composition is selected from the library.
  • a variety of methods exist to screen for the sensitivity of breast cancer cells taken at biopsy to the cell killing effects of drugs in vitro and in vivo chemosensitivity testing
  • exemplary methods are described by Tomikawa, et al. ( Anticancer Res., 18(2A):1059-62 (1998)) and by Coley, et al. ( Anticancer Res. 17(1A):231-6 (1997)) and by Andreotti, et al. i Cancer Res., 55(22):5276-82 (1995)).
  • In vitro cytotoxicity is often expressed as the concentration of a particular cancer drug needed to inhibit cancer cell proliferation by 50% in culture (IC 50 )
  • IC 50 50% in culture
  • cells obtained from a patient's biopsy specimen are teased apart (mechanically and/or by enzyme treatment), suspended in a medium which supports their growth and placed in wells of a culture plate.
  • Drugs at various dilutions are added and any growth inhibition of the cells caused by the drug is measured.
  • IC 50 values are derived from these measurements. Drugs that kill the cells or inhibit growth at concentrations at or below IC 50 values that can be achieved in vivo are considered as candidates for therapeutic intervention.
  • a therapeutic agent can be selected on the basis of historical information and accepted clinical practice (see for example, Handbook of Cancer Chemotherapy, 3 rd edition, R. T. Skeell, editor, A, Little Brown, Boston, 1991, pp 77-138.).
  • doxorubicin is known to be one of the most active agents against human breast cancer. Therefore, in a plurality of liposome-encapsulated cancer drugs, doxorubicin would represent an obvious selection for the treatment of breast cancer based on accepted clinical practice.
  • the two reagents are combined to create target cell-sensitized therapeutic liposome composition tailored to an individual patient's cancer.
  • Aseptic technique is used, preferably in a hospital pharmacy or other appropriate setting.
  • the liposomes are typically in suspension form and are administered parenterally, preferably intravenously.
  • Other routes of administration are suitable, including subcutaneous, intramuscular, interlesional (to tumors), intertracheal by inhalation, topical, internasal, intraocular, via direct injection into organs and intravenous.
  • a hematological disease e.g. a B-cell or T-cell malignancy, such as B-cell leukemias/lymphomas, multiple myeloma, T-cell lymphoma and acute lymphocytic leukemia.
  • the library includes a plurality of targeting conjugates.
  • Targeting conjugates suitable for selection include lipid-polymer-antibody conjugates, where the antibody is a monoclonal antibody or antibody fragment having a specific recognition to a B-cell or a T-cell epitope, as has been described in U.S. Pat. No. 5,620,689, which is incorporated herein by reference.
  • the antibody can be one that recognizes the B-cell epitopes CD19, CD20, CD22 or CD77.
  • the antibody can be one that recognizes the T-cell epitopes CD4, CD7 or CD8.
  • the library further includes liposomes having entrapped agents.
  • liposomes having the following entrapped agents are potential candidates for selection from the library: doxorubicin, vincristine, lomustine, interferon, melphalan, cyclophosphamide, prednisone, chlorambucil, carmustin and dexamethasone.
  • a blood or tissue sample is taken from the patient suffering from the hematological disorder for determination of the expression of various receptors, such as CD19, CD20, CD22, CD4, CD7, CD8. If the origin of the disorder is known to be either B-cell or T-cell, the receptor screening can of course be more selective, e.g., if the disorder is B-cell related, then the sample can be tested for expression of CD19, CD20 and CD22. Based on the results of the screening, a suitable targeting conjugate is chosen.
  • a therapeutic agent for treatment of the disorder is selected from the library using the procedures described in the breast cancer example above.
  • the selected conjugate and liposome composition are incubated together as described above to form the target-cell sensitized, therapeutic liposome composition specific for the patient.
  • Suitable dosages for the composition can be initially based on the standard chemotherapeutic dose and adjusted accordingly over the course of treatment by monitoring the disease progression.
  • Liposomes were prepared by mixing partially hydrogenated soy-bean phosphatiylcholine (PHPC, iodine value of 35, Lipoid (Ludwigshafen, Germany)), cholesterol (Croda (Fullerton, Calif.)) and mPEG-DSPE (prepared as described in Zalipsky, S., et al., Bioconjugate Chemistry, 4:296-299 (1993)) at a molar ratio of 55:40:3 in chloroform and/or methanol in a round bottom flask.
  • PHPC soy-bean phosphatiylcholine
  • cholesterol Choid (Ludwigshafen, Germany)
  • mPEG-DSPE prepared as described in Zalipsky, S., et al., Bioconjugate Chemistry, 4:296-299 (1993)
  • the solvents were removed by rotary evaporation, and the dried lipid film produced was hydrated with either sodium phosphate buffer (10 mM, 140 mM NaCl, pH 7) or HEPES buffer (25 mM, 150 mM NaCl, pH 7) to produce large multilamellar vesicles.
  • the resulting vesicles were passed repeatedly under pressure through 0.2, 0.1 and 0.05 m pore size polycarbonate membranes, until the average size distribution for the diameter (monitored by dynamic light scattering using a Coulter N4MD (Hialeah, Fla.)) was approximately 100 nm.
  • the mean particle diameter measured from 12 different batches ranged form 92 to 111 nm with an average 98 nm.
  • Targeting conjugates of and YIGSRG-PEG-DSPE were prepared according to Zalipsky, S., et al., Bioconjugate Chemistry, 8(2):111-118 (1997).
  • the pre-formed liposomes were incubated at either 25° C. or 37° C. with 1.2 mole percent of one of the targeting conjugates.
  • targeting conjugates micelles
  • liposomes liposomes
  • size exclusion chromatography For the sialyl-Lewis x -PEG-DSPE conjugate, a Biogel A50M column equilibrated with 10 mM sodium phosphate, 140 mM sodium chloride, and 0.02% NaN 3 at pH 6.5 was used.
  • YIGSRG-PEG-DSPE conjugate a Sepharose 4B column was used with 10% sucrose and 10 mM HEPES at pH 7.0 as eluent.
  • An anti-E-selectin Fab fragment was conjugated to PEG-DSPE to form a targeting conjugate as follows.
  • the excess reducing agent was removed on a 10DG-column (Bio-Rad) equilibrated with 25 mM HEPES/0.9% saline buffer. The collected fractions were analyzed spectrophotometrically to determine the fractions containing the Fab fragments. These fractions were pooled and diluted 1:50 in phosphate buffered saline to determine the protein concentration.
  • the Fab fragments (molecular weight of 3,000 Daltons) were mixed in a 1:1 molar ratio with PEG-DSPE (molecular weight 50,000 daltons) having an active maleimide end group (prepared as described in U.S. Pat. No. 5,527,528). The two components were incubated overnight at room temperature. The unreacted maleimide was quenched with 2 mM ⁇ -mercaptoethanol for 30 minutes at room temperature. The free Fab fragments and ⁇ -mercaptoethanol were separated from the Fab-PEG-DSPE conjugate on an S-200 column equilibrated in 25 mM HEPES/0.9% saline at pH 7.2.
  • Fractions of 1 ml were collected and read on the spectrophotometer at 280 nm to determine the fractions containing the conjugate and the free Fab fragments. The fractions were pooled accordingly and the concentration of the Fab-PEG-DSPE micellular solution is determined spectrophotometrically (280 nm). The efficiency of conjugation of the Fab fragment to the maleimide-PEG-DSPE was approximately 40%.
  • Liposomes of partially hydrogenated soy phosphatidylcholine (PHPC), PEG-DSPE and cholesterol in a 55:40:3 molar ratio were prepared as described in Example 1.
  • an amount of the Fab-PEG-DSPE conjugate was added to a suspension of liposomes and incubated overnight at room temperature.
  • a 100 ⁇ l aliquot of the insertion mixture was taken and placed on a SEPHAROSE 4B column (0.7 ⁇ 30 cm) to separate the free Fab conjugate from the liposomes. 1 ml fractions were collected and read on the spectrophotometer to determine the amount of conjugate inserted into the pre-formed liposomes. Greater than 95% of the conjugates were inserted into the pre-formed liposomes.
  • 111 In-labelled-liposomes containing 30 Fab residues per liposome and 70 Fab residues per liposome were administered to rats to determine the blood circulation lifetime of the liposomes.
  • 111 In-labelled-liposomes of PHPC, cholesterol and PEG-DSPE were administered. The results are shown in FIG. 4.
  • E-selectin Fab-PEG-DSPE targeting conjugate was inserted into pre-formed liposomes as follows.
  • the pre-formed liposomes were composed of hydrogenated soy phosphatidylcholine (HSPC), cholesterol and PEG-DSPE in a molar ratio of 53.5/40/4.
  • the liposomes included 2.5 mole percent of the lipid marker of fluorescein-DHPE (Molecular Probes, Inc.).
  • the preformed liposomes were incubated with the micellular solution of the targeting conjugate at 37° C. for 1 hour.
  • the insertion mixture was placed on a Bio-Rad A50 m column equilibrated with 25 mM HEPES/saline pH 7.2 and 0.5 ml fractions were collected. Spectrophotometric analysis of the fractions indicated that the insertion efficiency of the Fab targeting conjugate into the liposomes was approximately 100% after 2 hours at 37° C.
  • FIGS. 5 A- 5 B are photomicrographs (scanned images) of the blood vessels under transmitted light prior to liposome administration (FIG. 5A) and 5 hours after administration of the target-cell sensitized, fluorescein-labeled liposomes (FIG. 5B).

Abstract

Reagents for use in preparing a therapeutic liposome composition sensitized to a target cell are described. The reagents include a liposomal composition composed of pre-formed liposomes having an entrapped therapeutic agent and a plurality of targeting conjugates composed of a lipid, a hydrophilic polymer and a targeting ligand. The therapeutic, target-cell sensitized liposome composition is formed by incubating the liposomal composition with a selected conjugate.

Description

  • This application is a division of U.S. application Ser. No. 09/138,480 filed Aug. 21, 1998, now pending; which is a continuation-in-part of U.S. application Ser. No. 08/949,046, filed Oct. 10, 1997, now U.S. Pat. No. 5,891,468; which claims the priority of U.S. Provisional Application No. 60/028,269, filed Oct. 11, 1996, now abandoned, which are all incorporated herein by reference.[0001]
  • FIELD OF THE INVENTION
  • The present invention relates to a target-cell sensitized therapeutic liposome composition and to a method of preparing the composition. A library for preparation of the composition is also described. [0002]
  • BACKGROUND OF THE INVENTION
  • Liposomes, spherical, self-enclosed vesicles composed of amphipathic lipids, have been widely studied and are employed as vehicles for in vivo administration of therapeutic agents. In particular, the so-called long circulating liposomes formulations which avoid uptake by the organs of the mononuclear phagocyte system, primarily the liver and spleen, have found commercial applicability. Such long-circulating liposomes include a surface coat of flexible water soluble polymer chains, which act to prevent interaction between the liposome and the plasma components which play a role in liposome uptake. [0003]
  • More recently, efforts have focused on ways to achieve site specific delivery of long-circulating liposomes. In one approach, targeting ligands, such as an antibody, are attached to the liposomes' surfaces. This approach, where the targeting ligand is bound to the polar head group residues of liposomal lipid components, results in interference by the surface-grafted polymer chains, inhibiting the interaction between the bound ligand and its intended target (Klibanov, A. L., et al., [0004] Biochim. Biophys. Acta., 1062:142-148 (1991); Hansen, C. B., et al., Biochim. Biophys. Acta, 1239:133-144 (1995)).
  • In another approach, the targeting ligand is attached to the free ends of the polymer chains forming the surface coat on the liposomes (Allen. T. M., et al., [0005] Biochim. Biophys. Acta, 1237:99-108 (1995); Blume, G., et al., Biochim. Biophys. Acta, 1149:180-184 (1993)). Two approaches have been described for preparing a liposome having a targeting ligand attached to the distal end of the surface polymer chains. One approach involves preparation of lipid vesicles which include an end-functionalized lipid-polymer derivative; that is, a lipid-polymer conjugate where the free polymer end is reactive or “activated”. Such an activated conjugate is included in the liposome composition and the activated polymer ends are reacted with a targeting ligand after liposome formation. The disadvantage to this approach is the difficulty in reacting all of the activated ends with a ligand. The approach also requires a subsequent step for separation of the unreacted ligand from the liposome composition.
  • In another approach; the lipid-polymer-ligand conjugate is included in the lipid composition at the time of liposome formation. This approach has the disadvantage that some of the valuable ligand faces the inner aqueous compartment of the liposome and is unavailable for interaction with the intended target. [0006]
  • Both approaches suffer from a lack of flexibility in designing a therapeutic composition that is specific for a target cell for a specific patient. There is then a need for a liposome composition which provides flexibility in choice of the entrapped agent and the targeting ligand. [0007]
  • SUMMARY OF THE INVENTION
  • Accordingly, it is an object of the invention to provide a therapeutic liposome composition that is readily tailored and designed for a particular patient. [0008]
  • It is another object of the invention to provide a kit for formation of a therapeutic, target-cell sensitive liposome composition. [0009]
  • In one aspect, the invention includes a therapeutic liposome composition sensitized to a target cell, comprising [0010]
  • (i) a liposomal composition composed of pre-formed liposomes having an entrapped therapeutic agent; and (ii) a plurality of conjugates, each conjugate composed of (a) a lipid having a polar head group and a hydrophobic tail, (b) a hydrophilic polymer having a proximal end and a distal end, where the polymer is attached at its proximal end to the head group of the lipid, and (c) a targeting ligand attached to the distal end of the polymer. The therapeutic, target-cell sensitized liposome composition is formed by combining the liposomal composition with a conjugate selected from the plurality of conjugates. [0011]
  • In one embodiment, the targeting ligand is an antibody or an antibody fragment. In one embodiment, the antibody or antibody fragment is of mouse origin and is humanized to remove murine epitopes. [0012]
  • In another embodiment, the targeting ligand specifically binds to an extracellular domain of a growth factor receptor. Such receptors are selected from c-erbB-2 protein product of the HER2/neu oncogene, epidermal growth factor receptor, basic fibroblast growth factor receptor, and vascular endothelial growth factor receptor. [0013]
  • In another embodiment, the targeting ligand binds a receptor selected from E-selectin receptor, L-selectin receptor, P-selectin receptor, folate receptor, CD4 receptor, CD19 receptor, αβ integrin receptors and chemokine receptors. [0014]
  • The targeting ligand can also be folic acid, pyridoxal phosphate, vitamin B12, sialyl Lewis[0015] x, transferrin, epidermal growth factor, basic fibroblast growth factor, vascular endothelial growth factor, VCAM-1, ICAM-1, PECAM-1, an RGD peptide or an NGR peptide.
  • The hydrophilic polymer surrounding the preformed liposomes is selected from the group consisting of polyvinylpyrrolidone, polyvinylmethylether, polymethyloxazoline, polyethyloxazoline, polyhydroxypropyloxazoline, polyhydroxypropylmethacrylamide, polymethacrylamide, polydimethylacrylamide, polyhydroxypropylmethacrylate, polyhydroxyethylacrylate, hydroxymethylcellulose, hydroxyethylcellulose polyethyleneglycol, polyaspartamide and hydrophilic peptide sequences. [0016]
  • In one embodiment, the hydrophilic polymer is polyethylene glycol of molecular weight between 500-5,000 daltons. [0017]
  • The entrapped therapeutic agent is, in one embodiment, a cytotoxic drug. The drug can be an anthracycline antibiotic selected from doxorubicin, daunorubicin, epirubicin and idarubicin and analogs thereof. [0018]
  • The cytotoxic agent can also be a platinum compound selected from cisplatin, carboplatin, ormaplatin, oxaliplatin, zeniplatin, enloplatin, lobaplatin, spiroplatin, ((−)-(R)-2-aminomethylpyrrolidine (1,1-cyclobutane dicarboxylato)platinum), (SP-4-3(R)-1,1-cyclobutane-dicarboxylato(2-)-(2-methyl-1,4-butanediamine-N,N′)platinum), nedaplatin and (bis-acetato-ammine-dichloro-cyclohexylamine-platinum(IV)). [0019]
  • In another embodiment, the cytotoxic agent is a topoisomerase 1 inhibitor selected from the group consisting of topotecan, irinotecan, (7-(4-methylpiperazino-methylene)-10,11-ethylenedioxy-20(S)-camptothecin), 7-(2-(N-isopropylamino)ethyl)-(20S)-camptothecin, 9-aminocamptothecin and 9-nitrocamptothecin. [0020]
  • In another embodiment, the cytotoxic agent is a vinca alkaloid selected from the group consisting of vincristine, vinblastine, vinleurosine, vinrodisine, vinorelbine and vindesine. [0021]
  • In another embodiment, the entrapped agent is a nucleic acid. The nucleic acid can be an antisense oligonucleotide or ribozyme or a plasmid containing a therapeutic gene which when internalized by the target cells achieves expression of the therapeutic gene to produce a therapeutic gene product. [0022]
  • In another aspect, the invention includes a plurality of targeting conjugates for use in preparing a targeted, therapeutic liposome composition. Each conjugate is composed of a (i) a lipid having a polar head group and a hydrophobic tail, (ii) a hydrophilic polymer having a proximal end and a distal end, the polymer attached at its proximal end to the head group of the lipid, and (iii) a targeting ligand attached to the distal end of the polymer. [0023]
  • The lipid in the conjugates is, in one embodiment, distearoyl phosphatidylethanolamine, distearoyl-phosphatidylcholine, monogalactosyl diacylglycerols or digalactosyl diacylglycerols. [0024]
  • The hydrophilic polymer in the conjugates is selected from the group consisting of polyvinylpyrrolidone, polyvinylmethylether, polymethyloxazoline, polyethyloxazoline, polyhydroxypropyloxazoline, polyhydroxypropylmethacrylamide, polymethacrylamide, polydimethylacrylamide, polyhydroxypropylmethacrylate, polyhydroxyethylacrylate, hydroxymethylcellulose, hydroxyethylcellulose, polyethyleneglycol, polyaspartamide and hydrophilic peptide sequences. [0025]
  • The targeting ligand of the conjugates can be any of those recited above. [0026]
  • In another aspect, the invention includes a method of formulating a therapeutic liposome composition having sensitivity to a target cell. The method includes the steps of (i) selecting a liposome formulation composed of pre-formed liposomes having an entrapped therapeutic agent; (ii) selecting from a plurality of targeting conjugates a targeting conjugate composed of (a) a lipid having a polar head group and a hydrophobic tail, (b) a hydrophilic polymer having a proximal end and a distal end, where the polymer is attached at its proximal end to the head group of the lipid, and (c) a targeting ligand attached to the distal end of the polymer; and (iii) combining the liposome formulation and the selected targeting conjugate to form said therapeutic, target-cell sensitive liposome composition. [0027]
  • In one embodiment, combining includes incubating under conditions effective to achieve insertion of the selected targeting conjugate into the liposomes of the selected liposome formulation. [0028]
  • In another embodiment, selecting a liposome formulation includes determining the sensitivity of the target cell to the therapeutic activity of the entrapped therapeutic agent. [0029]
  • In another embodiment, selecting a targeting conjugate includes determining the ability of the targeting ligand to bind cell surface receptors expressed on the target cell. [0030]
  • In another embodiment, selecting a targeting conjugate is based on (i) the ability of a targeting ligand to bind to cell surface receptors expressed on the target cell and (ii) the ability of the target cell to internalize liposomes bound to the target cell by binding between the target cell and the targeting ligand. [0031]
  • These and other objects and features of the invention will be more fully appreciated when the following detailed description of the invention is read in conjunction with the accompanying drawings.[0032]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 illustrates a library composed of a plurality of therapeutic pre-formed liposomes and a plurality of targeting conjugates; [0033]
  • FIGS. [0034] 2A-2D are plots showing the fraction of liposomes (peak centered at fraction 10) and the fraction of micellular targeting conjugates (peak centered at fraction 20) by size exclusion chromatography from samples taken during incubation of a targeting conjugate sialyl-Lewisx-PEG-DSPE with preformed liposomes at times of 0 hours (FIG. 2A), 1 hour (FIG. 2B), 3 hours (FIG. 2C) and 5 hours (FIG. 2D);
  • FIG. 3 is a plot showing the time course for insertion of the targeting conjugate sialyl-Lewis[0035] x-PEG-DSPE into pre-formed liposomes when incubated at 25° C. (closed circles) and 37° C. (open squares);
  • FIG. 4 is a plot showing the blood circulation lifetime of target-cell sensitized liposome prepared in accordance with the invention, where the percent of injected dose in vivo for liposomes having E-selectin Fab fragments targeting ligands (30 ligands per liposome represented by solid triangles, 70 ligands per liposome represented by solid squares) and for liposomes having a surface coating of polyethyleneglycol chains (open circles) as a function of time after dosing; and [0036]
  • FIGS. [0037] 5A-5B are scanned images of micrographs of blood vessels in a window chamber of a mouse dorsal fold, where FIG. 5A is the control of the untreated blood vessels under transmitted light, and FIG. 5B is a fluorescence micrograph showing binding of fluorsecin-labeled liposomes bearing an E-selectin Fab fragments to endothelial cells in the blood vessels.
  • DETAILED DESCRIPTION OF THE INVENTION
  • I. Definitions [0038]
  • Unless otherwise indicated, the terms below have the following meaning: [0039]
  • “Incubating” or “incubating under conditions effective to achieve insertion” refer to conditions of time, temperature and liposome lipid composition which allow for penetration and entry of a selected component, such as a lipid or lipid conjugate, into the lipid bilayer of a liposome. [0040]
  • “Pre-formed liposomes” refers to intact, previously formed unilamellar or multilamellar lipid vesicles. [0041]
  • “Sensitized to a target cell” or “target-cell sensitized” refers to a liposome which includes a ligand or moiety covalently bound to the liposome and having binding affinity for a receptor expressed on a particular cell. [0042]
  • “Therapeutic liposome composition” refers to liposomes which include a therapeutic agent entrapped in the aqueous spaces of the liposomes or in the lipid bilayers of the liposomes. [0043]
  • “Vesicle-forming lipid” refers to any lipid capable of forming part of a stable micelle or liposome composition and typically including one or two hydrophobic acyl hydrocarbon chains or a steroid group and may contain a chemically reactive group, such as an amine, acid, ester, aldehyde or alcohol, at its polar head group. [0044]
  • I. Liposome and Conjugate Library [0045]
  • In one aspect, the invention includes a kit or “library” for preparation of a therapeutic, target-cell sensitized liposome composition. FIG. 1 shows such a [0046] library 10, where a plurality 12 of targeting conjugates 12(a), 12(b), 12(c), etc. and a plurality 14 of pre-formed therapeutic liposome compositions, such compositions 14(a), 14(b), 14(c) are shown. The targeting conjugates and pre-formed liposome pluralities are shown in suspension form in vials ready for use, however it will be appreciated that other storage forms are contemplated, such as lyophilized or freeze-dried.
  • The targeting conjugates in the library are lipid-polymer-ligand conjugates and will be described in more detail below. The conjugates in the library differ in the targeting ligand attached to the lipid-polymer, as well as in the lipid and polymer components. Exemplary ligands and lipid and polymer components will be set forth below. [0047]
  • The pre-formed liposomes in the library are either conventional liposomes containing an entrapped therapeutic agent or are liposomes having a surface coating of hydrophilic polymer chains, as will be described below. The pre-formed liposomes in the library differ from one another generally in the entrapped therapeutic agent and exemplary agents will be set forth below. The pre-formed liposomes can also differ from one another in the liposome lipid components. [0048]
  • A therapeutic, target-cell sensitized liposome composition is prepared from the library as follows. A composition specific for a subject suffering from a particular condition, for example a solid tumor of the lung, a bacterial infection or a viral infection, is prepared by selecting a targeting conjugate from the library. The targeting conjugate is selected either according to knowledge of those of skill in the art of ligand-receptor binding pairs or by obtaining a suitable patient sample, e.g., a fluid sample, a biopsy or the like. The sample is tested by means known to those in the art for expression of a variety of receptors to determine the appropriate targeting ligand. [0049]
  • A pre-formed therapeutic liposome composition is selected based on knowledge of those of skill in the art of the therapeutic agents appropriate for treatment of the particular condition. Alternatively, the therapeutic liposome composition is selected after performing chemosensitivity tests to determine the effect of the entrapped agent on cells of concern obtained from the patient biopsy or fluid sample. [0050]
  • Following selection of the targeting conjugate and of the pre-formed liposome composition, the target-cell sensitized, therapeutic liposome composition for the subject is prepared by combining the two components. As will be described, the components are combined under conditions effective to achieve insertion of the targeting conjugate into the liposome bilayer to create the target-cell sensitized liposomes. After insertion is complete, the composition is administered to patient. [0051]
  • The therapeutic pre-formed liposomes and the targeting conjugate will now be described in more detail. [0052]
  • A. Therapeutic Pre-formed Liposome Component [0053]
  • As discussed above, one component of the kit or library for preparing the composition of the invention is a plurality of pre-formed liposomes having an entrapped therapeutic or diagnostic agent. In this section, the liposome lipid components, exemplary agents and methods of preparing the liposomes are described. [0054]
  • 1. Liposome Components [0055]
  • Liposomes suitable for use in the composition of the present invention include those composed primarily of vesicle-forming lipids. Such a vesicle-forming lipid is one which (a) can form spontaneously into bilayer vesicles in water, as exemplified by the phospholipids, or (b) is stably incorporated into lipid bilayers, with its hydrophobic moiety in contact with the interior, hydrophobic region of the bilayer membrane, and its head group moiety oriented toward the exterior, polar surface of the membrane. [0056]
  • The vesicle-forming lipids of this type are preferably ones having two hydrocarbon chains, typically acyl chains, and a head group, either polar or nonpolar. There are a variety of synthetic vesicle-forming lipids and naturally-occurring vesicle-forming lipids, including the phospholipids, such as phosphatidylcholine, phosphatidylethanolamine, phosphatidic acid, phosphatidylinositol, and sphingomyelin, where the two hydrocarbon chains are typically between about 14-22 carbon atoms in length, and have varying degrees of unsaturation. The above-described lipids and phospholipids whose acyl chains have varying degrees of saturation can be obtained commercially or prepared according to published methods. Other suitable lipids include glycolipids, cerebrosides and sterols, such as cholesterol. [0057]
  • Cationic lipids are also suitable for use in the liposomes of the invention, where the cationic lipid can be included as a minor component of the lipid composition or as a major or sole component. Such cationic lipids typically have a lipophilic moiety, such as a sterol, an acyl or diacyl chain, and where the lipid has an overall net positive charge. Preferably, the head group of the lipid carries the positive charge. Exemplary cationic lipids include 1,2-dioleyloxy-3-(trimethylamino) propane (DOTAP); N-[1-(2,3,-ditetradecyloxy)propyl]-N,N-dimethyl-N-hydroxyethylammonium bromide (DMRIE); N-[1-(2,3,-dioleyloxy)propyl]-N,N-dimethyl-N-hydroxy ethylammonium bromide (DORIE); N-[1-(2,3-dioleyloxy) propyl]-N,N,N-trimethylammonium chloride (DOTMA); 3 [N-(N′,N′-dimethylaminoethane) carbamoly] cholesterol (DC-Chol); and dimethyldioctadecylammonium (DDAB). [0058]
  • The cationic vesicle-forming lipid may also be a neutral lipid, such as dioleoylphosphatidyl ethanolamine (DOPE) or an amphipathic lipid, such as a phospholipid, derivatized with a cationic lipid, such as polylysine or other polyamine lipids. For example, the neutral lipid (DOPE) can be derivatized with polylysine to form a cationic lipid. [0059]
  • In another embodiment, the vesicle-forming lipid is selected to achieve a specified degree of fluidity or rigidity, to control the stability of the liposome in serum, to control the conditions effective for insertion of the targeting conjugate, as will be described, and to control the rate of release of the entrapped agent in the liposome. [0060]
  • Liposomes having a more rigid lipid bilayer, or a liquid crystalline bilayer, are achieved by incorporation of a relatively rigid lipid, e.g., a lipid having a relatively high phase transition temperature, e.g., up to 60° C. Rigid, i.e., saturated, lipids 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. [0061]
  • On the other hand, 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. [0062]
  • As will be described below, the targeted, therapeutic liposome composition of the invention is prepared using pre-formed liposomes and a targeting conjugate, which are incubated together under conditions effective to achieve insertion of the conjugate into the liposome bilayer. More specifically, the two components are incubated together under conditions which achieve insertion of the conjugate in such a way that the targeting ligand is oriented outwardly from the liposome surface, and therefore available for interaction with its cognate receptor. [0063]
  • Vesicle-forming lipids having phase transition temperatures from approximately 2° C.-80° C. are suitable for use in the pre-formed liposome component of the present composition. By way of example, the lipid distearyl phosphatidylcholine (DSPC) has a phase transition temperature of 62° C. and the lipid hydrogenated soy phosphatidylcholine (HSPC) has a phase transition temperature of 58° C. Phase transition temperatures of many lipids are tabulated in a variety of sources, such as Avanti Polar Lipids catalogue and Lipid Thermotropic Phase Transition Database (LIPIDAT, NIST Standard Reference Database 34). [0064]
  • In one embodiment of the invention, a vesicle-forming lipid having a phase transition temperature between about 30-70° C. is employed. In another embodiment, the lipid used in forming the liposomes is one having a phase transition temperature within about 20° C., more preferably 10° C., most preferably 5° C., of the temperature to which the ligand in the targeting conjugate can be heated without affecting its binding activity. [0065]
  • It will be appreciated that the conditions effective to achieve insertion of the targeting conjugate into the liposome are determined based on several variables, including, the desired rate of insertion, where a higher incubation temperature may achieve a faster rate of insertion, the temperature to which the ligand can be safely heated without affecting its activity, and to a lesser degree the phase transition temperature of the lipids and the lipid composition. It will also be appreciated that insertion can be varied by the presence of solvents, such as amphipathic solvents including polyethyleneglycol and ethanol, or detergents. [0066]
  • In one embodiment of the invention, the preformed liposomes also include a vesicle-forming lipid derivatized with a hydrophilic polymer. As has been described, for example in U.S. Pat. No. 5,013,556, including such a derivatized lipid in the liposome composition forms a surface coating of hydrophilic polymer chains around the liposome. The surface coating of hydrophilic polymer chains is effective to increase the in vivo blood circulation lifetime of the liposomes when compared to liposomes lacking such a coating. [0067]
  • Vesicle-forming lipids suitable for derivatization with a hydrophilic polymer include any of those lipids listed above, and, in particular phospholipids, such as distearoyl phosphatidylethanolamine (DSPE). [0068]
  • Hydrophilic polymers suitable for derivatization with a vesicle-forming lipid include polyvinylpyrrolidone, polyvinylmethylether, polymethyloxazoline, polyethyloxazoline, polyhydroxypropyloxazoline, polyhydroxypropylmethacrylamide, polymethacrylamide, polydimethylacrylamide, polyhydroxypropylmethacrylate, polyhydroxyethylacrylate, hydroxymethylcellulose, hydroxyethylcellulose, polyethyleneglycol, polyaspartamide and hydrophilic peptide sequences. The polymers may be employed as homopolymers or as block or random copolymers. [0069]
  • A preferred hydrophilic polymer chain is polyethyleneglycol (PEG), preferably as a PEG chain having a molecular weight between 500-10,000 daltons, more preferably between 1,000-5,000 daltons. Methoxy or ethoxy-capped analogues of PEG are also preferred hydrophilic polymers, commercially available in a variety of polymer sizes, e.g., 120-20,000 daltons. [0070]
  • Preparation of vesicle-forming lipids derivatized with hydrophilic polymers has been described, for example in U.S. Pat. No. 5,395,619. Preparation of liposomes including such derivatized lipids has also been described, where typically, between 1-20 mole percent of such a derivatized lipid is included in the liposome formulation. [0071]
  • 2. Therapeutic Agent [0072]
  • The pre-formed liposomes include an agent entrapped in the liposome. Entrapped is intended to include encapsulation of an agent in the aqueous core and aqueous spaces of liposomes as well as entrapment of an agent in the lipid bilayer(s) of the liposomes. [0073]
  • Agents contemplated for use in the composition of the invention are widely varied, and include both therapeutic applications and those for use in diagnostic applications. [0074]
  • Therapeutic agents include natural and synthetic compounds having the following therapeutic activities: [0075]
  • anti-arthritic, anti-arrhythmic, anti-bacterial, anticholinergic, anticoagulant, antidiuretic, antidote, antiepileptic, antifungal, anti-inflammatory, antimetabolic, antimigraine, antineoplastic, antiparasitic, antipyretic, antiseizure, antisera, antispasmodic, analgesic, anesthetic, beta-blocking, biological response modifying, bone metabolism regulating, cardiovascular, diuretic, enzymatic, fertility enhancing, growth-promoting, hemostatic, hormonal, hormonal suppressing, hypercalcemic alleviating, hypocalcemic alleviating, hypoglycemic alleviating, hyperglycemic alleviating, immunosuppressive, immunoenhancing, muscle relaxing, neurotransmitting, parasympathomimetic, sympathominetric plasma extending, plasma expanding, psychotropic, thrombolytic and vasodilating. [0076]
  • In a preferred embodiment, the entrapped agent is a cytotoxic drug, that is, a drug having a deleterious or toxic effect on cells. Exemplary cytotoxic agents include the anthracycline antibiotics such as doxorubicin, daunorubicin, epirubicin and idarubicin, and analogs of these, such as epirubidin and mitoxantrone; platinum compounds, such as cisplatin, carboplatin, ormaplatin, oxaliplatin, zeniplatin, enloplatin, lobaplatin, spiroplatin, ((−)-(R)-2-aminomethylpyrrolidine (1,1-cyclobutane dicarboxylato)platinum)(DWA2114R), (SP-4-3(R)-1,1-cyclobutane-dicarboxylato(2-)-(2-methyl-1,4-butanediamine-N,N′)platinum) (CI-973), nedaplatin (254-S) and (bis-acetato-ammine-dichloro-cyclohexylamine-platinum(IV)) (JM-216) (Weiss, R. B., et al., [0077] Drugs, 46(3):360-377 (1993)); and vinca alkaloids, such as vincristine, vinblastine, vinleurosine, vinrodisine, vinorelbine (navelbine) and vindesine.
  • Another preferred group of cytotoxic agents is a topoisomerase I inhibitor, such as camptothecin and its analogues, including SN-38 ((+)-(4S)-4,11-diethyl-4,9-dihydroxy-1H-pyrano[3′,4′:6,7]-indolizino[1,2-b]quinoline-3,14(4H,12H)-dione); 9-aminocamptothecin; 9-nitrocamptothecin, topotecan (hycamtin; 9-dimethyl-aminomethyl-10-hydroxycamptothecin); irinotecan (CPT-11; 7-ethyl-10-[4-(1-piperidino)-1-piperidino]-carbonyloxy-camptothecin), which is hydrolyzed in vivo to SN-38); 7-ethylcamptothecin and its derivatives (Sawada, S. et al., [0078] Chem. Pharm. Bull., 41(2):310-313 (1993)); 7-chloromethyl-10,11-methylene-dioxy-camptothecin; and others (SN-22, Kunimoto, T. et al., J. Pharmacobiodyn., 10(3):148-151: (1987); DX-8951f and GG-211 ((7-(4-methylpiperazino-methylene)-10,11-ethylenedioxy-20(S)-camptothecin)) (Rothenberg, M. L., Ann. Oncol., 8(9):837-855 (1997)), and 7-(2-(N-isopropylamino)ethyl)-(20S)-camptothecin (Chong Kun Dang Corp., Seoul Dorea, CKD602).
  • In another embodiment, the entrapped therapeutic agent is an angiogenesis inhibitor, such as angiostatin, endostatin and TNFα. [0079]
  • In another embodiment, the entrapped therapeutic agent in a nucleic acid, selected from a variety of DNA and RNA based nucleic acids, including fragments and analogues of these. A variety of genes for treatment of various conditions have been described, and coding sequences for specific genes of interest can be retrieved from DNA sequence databanks, such as GenBank or EMBL. For example, polynucleotides for treatment of viral, malignant and inflammatory diseases and conditions, such as, cystic fibrosis, adenosine deaminase deficiency and AIDS, have been described. Treatment of cancers by administration of tumor suppressor genes, such as APC, DPC4, NF-1, NF-2, MTS1, RB, p53, WT[0080] 1, BRCA1, BRCA2 and VHL, are contemplated.
  • Administration of the following nucleic acids for treatment of the indicated conditions are also contemplated: HLA-B7, tumors, colorectal carcinoma, melanoma; IL-2, cancers, especially breast cancer, lung cancer, and tumors; IL-4, cancer; TNF, cancer; IGF-1 antisense, brain tumors; IFN, neuroblastoma; GM-CSF, renal cell carcinoma; MDR-1, cancer, especially advanced cancer, breast and ovarian cancers; and HSV thymidine kinase, brain tumors, head and neck tumors, mesothelioma, ovarian cancer. [0081]
  • The polynucleotide can be an antisense DNA oligonucleotide composed of sequences complementary to its target, usually a messenger RNA (mRNA) or an mRNA precursor. The mRNA contains genetic information in the functional, or sense, orientation and binding of the antisense oligonucleotide inactivates the intended mRNA and prevents its translation into protein. Such antisense molecules are determined based on biochemical experiments showing that proteins are translated from specific RNAs and once the sequence of the RNA is known, an antisense molecule that will bind to it through complementary Watson-Crick base pairs can be designed. Such antisense molecules typically contain between 10-30 base pairs, more preferably between 10-25, and most preferably between 15-20. [0082]
  • The antisense oligonucleotide can be modified for improved resistance to nuclease hydrolysis, and such analogues include phosphorothioate, methylphosphonate, phosphodiester and p-ethoxy oligonucleotides (WO 97/07784). [0083]
  • The entrapped agent can also be a ribozyme or catalytic RNA. [0084]
  • 3. Liposome Preparation [0085]
  • The liposomes may be prepared by a variety of techniques, such as those detailed in Szoka, F., Jr., et al., [0086] Ann. Rev. Biophys. Bioeng. 9:467 (1980), and specific examples of liposomes prepared in support of the present invention will be described below. Typically, the liposomes are multilamellar vesicles (MLVs), which can be formed by simple lipid-film hydration techniques. In this procedure, a mixture of liposome-forming lipids of the type detailed above dissolved in a suitable organic solvent is evaporated in a vessel to form a thin film, which is then covered by an aqueous medium. The lipid film hydrates to form MLVs, typically with sizes between about 0.1 to 10 microns.
  • As described above, in one embodiment, the preformed liposomes include a vesicle-forming lipid derivatized with a hydrophilic polymer to form a surface coating of hydrophilic polymer chains on the liposomes surface. Such a coating is preferably prepared by including between 1-20 mole percent of the derivatized lipid with the remaining liposome forming components, e.g., vesicle-forming lipids. Exemplary methods of preparing derivatized lipids and of forming polymer-coated liposomes have been described in co-owned U.S. Pat. Nos. 5,013,556, 5,631,018 and 5,395,619, which are incorporated herein by reference. It will be appreciated that the hydrophilic polymer may be stably coupled to the lipid, or coupled through an unstable linkage which allows the coated liposomes to shed the coating of polymer chains as they circulate in the bloodstream or in response to a stimulus. [0087]
  • The therapeutic or diagnostic agent of choice can be incorporated into liposomes by standard methods, including (i) passive entrapment of a water-soluble compound by hydrating a lipid film with an aqueous solution of the agent, (ii) passive entrapment of a lipophilic compound by hydrating a lipid film containing the agent, and (iii) loading an ionizable drug against an inside/outside liposome pH gradient. Other methods, such as reverse evaporation phase liposome preparation, are also suitable. [0088]
  • Polynucleotides, oligonucleotides, other nucleic acids, such as a DNA plasmid, can be entrapped in the liposome by condensing the nucleic acid in single-molecule form. The nucleic acid is suspended in an aqueous medium containing protamine sulfate, spermine, spermidine, histone, lysine, mixtures thereof, or other suitable polycationic condensing agent, under conditions effective to condense the nucleic acid into small particles. The solution of condensed nucleic acid molecules is used to rehydrate a dried lipid film to form liposomes with the condensed nucleic acid in entrapped form. A similar approach to condensing nucleic acids for entrapment in liposomes is described in co-pending U.S. patent application Ser. No. 09/103,341. [0089]
  • The pre-formed liposomes of the invention are preferably prepared to have substantially homogeneous sizes in a selected size range, typically between about 0.01 to 0.5 microns, more preferably between 0.03-0.40 microns. One effective sizing method for REVs and MLVs involves extruding an aqueous suspension of the liposomes through a series of polycarbonate membranes having a selected uniform pore size in the range of 0.03 to 0.2 micron, typically 0.05, 0.08, 0.1, or 0.2 microns. The pore size of the membrane corresponds roughly to the largest sizes of liposomes produced by extrusion through that membrane, particularly where the preparation is extruded two or more times through the same membrane. Homogenization methods are also useful for down-sizing liposomes to sizes of 100 nm or less (Martin, F. J., in [0090] SPECIALIZED DRUG DELIVERY SYSTEMS-MANUFACTURING AND PRODUCTION TECHNOLOGY, (P. Tyle, Ed.) Marcel Dekker, New York, pp. 267-316 (1990)).
  • B. Targeting Conjugates [0091]
  • The kit or library of the invention also includes a targeting conjugate, now to be described. The targeting conjugate is composed of (i) a lipid having a polar head group and a hydrophobic tail, e.g., a vesicle-forming lipid and any of those described above are suitable; (ii) a hydrophilic polymer attached to the head group of the vesicle-forming lipid, and any of the polymers recited above are suitable; and (iii) a targeting ligand attached to the polymer. [0092]
  • The targeting ligand for use in the conjugate can be selected from a wide variety of moieties capable of targeting the pre-formed liposomes to a selected cell or tissue. Examples of suitable ligands suitable are listed in Table 1. [0093]
    TABLE 1
    LIGAND-RECEPTOR PAIRS AND ASSOCIATED TARGET CELL
    LIGAND RECEPTOR CELL TYPE
    Folate folate receptor epithelial carcinomas, bone
    arrow stem cells
    water soluble vitamins vitamin receptor various cells
    Pyridoxyl phosphate CD4 CD4 + lymphocytes
    Apolipoproteins LDL liver hepatocytes, vascular
    endothelial cells
    Insulin insulin receptor pancreatic islet cells
    Transferrin Transferrin receptor endothelial cells (brain)
    Galactose Asialoglycoprotein liver hepatocytes
    receptor
    Sialyl-Lewisx E, P selectin activated endothelial cells
    Mac-1 L selectin neutrophils, leukocytes
    VEGF Flk-1, 2 tumor epithelial cells
    basic FGF FGF receptor tumor epithelial cells
    EGF EGF receptor epithelial cells
    VCAM-1 α4β1 integrin vascular endothelial cells
    ICAM-1 αLβ2 integrin vascular endothelial cells
    PECAM-1/CD31 αvβ3 integrin vascular endothelial cells
    Fibronectin αvβ3 integrin activated platelets
    Osteopontin αvβ1 and αvβ5 endothelial cells and
    integrins smooth muscle cells in
    atherosclerotic plaques
    RGD peptides and αvβ3 integrin tumor endothelial cells,
    peptide mimetics (i.e. vascular smooth muscle
    amino acid, sequences cells
    of matrix proteins)
    HIV GP 120/41 or CD4 CD4 + lymphocytes
    GP120 C4 domain
    peptomers
    HIV GP120/41 Chemokine receptor macrophages, dendritic
    (Macrophage tropic CC-CRK-5 cells
    isolates)
    Anti-cell surface re- Cell surface erythrocytes, platelets,
    ceptor antibodies (or receptors endothelial cells,
    fragments thereof), lymphocytes, tumors
    such as anit-HER2/
    neu, anti-selectin,
    anti-VEGF
    Anti-cell surface re- Cell surface recep- bone marrow stem cells,
    ceptor antibodies (or tors such as CD34, malignant B and T cells
    fragments thereof) CD19, CD4, CD7,
    CD8, CD20, CD22
  • One preferred ligand is an antibody or an antibody fragment. It will be appreciated that the antibody or antibody fragment can be of mouse origin and humanized to remove murine surface recognition features. [0094]
  • In another preferred embodiment, the targeting ligand binds to an extracellular domain of a growth factor receptor. Exemplary receptors include the c-erbB-2 protein product of the HER2/neu oncogene, epidermal growth factor (EGF) receptor, basic fibroblast growth receptor (basic FGF) receptor and vascular endothelial growth factor receptor, E-, L- and P-selectin receptors, folate receptor, CD4 receptor, CD19 receptor, αβ integrin receptors and chemokine receptors. [0095]
  • 1. Preparation of Targeting Conjugates [0096]
  • As described above, the targeting ligand is covalently attached to the free distal end of the hydrophilic polymer chain, which is attached at its proximal end to a vesicle-forming lipid. There are a wide variety of techniques for attaching a selected hydrophilic polymer to a selected lipid and activating the free, unattached end of the polymer for reaction with a selected ligand, and in particular, the hydrophilic polymer polyethyleneglycol (PEG) has been widely studied (Allen, T. M., et al., [0097] Biochemicia et Biophysica Acta 1237:99-108 (1995); Zalipsky, S., Bioconjugate Chem., 4(4):296-299 (1993); Zalipsky, S., et al., FEBS Lett. 353:71-74 (1994); Zalipsky, S., et al., Bioconjugate Chemistry, 705-708 (1995); Zalipsky, S., in STEALTH LIPOSOMES (D. Lasic and F. Martin, Eds.) Chapter 9, CRC Press, Boca Raton, Fla. (1995)).
  • Generally, the PEG chains are functionalized to contain reactive groups suitable for coupling with, for example, sulfhydryls, amino groups, and aldehydes or ketones (typically derived from mild oxidation of carbohydrate portions of an antibody) present in a wide variety of ligands (see Table 1). Examples of such PEG-terminal reactive groups include maleimide (for reaction with sulfhydryl groups), N-hydroxysuccinimide (NHS) or NHS-carbonate ester (for reaction with primary amines), hydrazide or hydrazine (for reaction with aldehydes or ketones), iodoacetyl (preferentially reactive with sulfhydryl groups) and dithiopyridine (thiol-reactive). Synthetic reaction schemes for activating PEG with such groups are set forth in U.S. Pat. Nos. 5,631,018, 5,527,528, 5,395,619, and the relevant sections describing synthetic reaction procedures are expressly incorporated herein by reference. [0098]
  • It will be appreciated that any of the hydrophilic polymers recited above in combination with any of the vesicle-forming lipids recited above can be employed for the targeting conjugate and suitable reaction sequences can be determined by those of skill in the art. [0099]
  • II. Preparation of the Liposome Composition [0100]
  • The section above described preparation of the components in the library of the invention, namely the pre-formed liposomes and the targeting conjugates. This section describes preparation of the target-cell sensitized, therapeutic liposome composition using these two components. [0101]
  • As discussed briefly above, a pre-formed therapeutic liposome composition and a targeting conjugate are selected from the library. The two components are combined under conditions effective to achieve insertion of the targeting conjugate into the liposome lipid bilayer to form the target-cell sensitized composition. [0102]
  • In studies performed in support of the invention, a targeting conjugate of the ligand sialyl-Lewis[0103] x was attached to PEG-DSPE according to known methods (DeFrees, S. A., et al., J. Am. Chem. Soc., 118:6101-6104 (1996)). Sialyl-Lewisx can be used to target liposomes to cells expressing endothelial leukocyte adhesion molecule-1 (ELAM-1 or E-selectin) for delivery of a therapeutic agent to a site of inflammation. ELAM-1 is expressed on the surface of endothelial cells of blood vessels adjacent to sites of inflammation. ELAM-1 recognizes and binds the polysaccharide moiety sialyl-Lewisx which is present on surfaces of neutrophils, and recruits neutrophils to sites of inflammation.
  • Pre-formed liposomes were prepared as described in Example 1 and were composed of partially hydrogenated soy-bean phosphatidylcholine (PHPC), cholesterol and MPEG-DSPE in a molar ratio of 55:40:3. The liposomes were sized to a diameter of about 100 nm. The liposomes were incubated at 37° C. with 1.2 mole percent sialyl-Lewis[0104] x-PEG-DSPE targeting conjugate to achieve insertion of the conjugate into the pre-formed liposomes.
  • Insertion of the conjugate into the liposomes was monitored by sampling the mixture and tracking the relative amounts of micellular conjugate and liposomes by size exclusion chromatography, and the results are shown in FIGS. [0105] 2A-2D.
  • In FIGS. [0106] 2A-2D, the liposome fraction of the incubation mixture is represented by the peak centered around fraction 10 and the micellular targeting conjugate is represented by the peak centered around fraction 20. FIG. 2A shows the initial composition mixture, at time zero and FIGS. 2B-2D show the composition after 1, 3 and 5 hours incubation, respectively. Disappearance as a function of incubation time of the conjugate micelles (peak at fraction 20) is apparent, indicating insertion of the targeting conjugate into the pre-formed liposomes.
  • FIG. 3 shows the time-course of insertion of sialyl-Lewis[0107] x-PEG-DSPE targeting conjugate into PHPC:Chol:mPEG-DSPE (55:40:3) pre-formed liposomes at 37° C. (open squares) and 25° C. (closed circles). Insertion of the conjugate into the pre-formed liposomes proceeded more rapidly at 37° C., however insertion at ambient temperature was also substantial.
  • Other experiments in support of the invention were performed using the targeting conjugate YIGSRG-PDG-DSPE, prepared as described in Zalipsky, S., et al., [0108] Bioconjugate Chemistry, 8(2):111-118 (1997). The pentapeptide YIGSRG (Tyr-Ile-Gly-Ser-Arg) is the shortest fragment of the basement membrane glycoprotein laminin which retains binding activity to laminin cell surface receptors. In these studies, essentially the same time course of insertion was observed (data not shown) upon incubation with pre-formed liposomes of PHPC:cholesterol:mPEG-DSPE.
  • Table 2 shows the average particle size determined by dynamic light scattering after a 5 hour incubation period at 37° C. of pre-formed liposomes and a targeting conjugate, either YIGSRG-PEG-DSPE or sialyl-Lewis[0109] x-PEG-DSPE. The average particle size after incubation and insertion of the targeting conjugate increased only slightly.
    TABLE 2
    Liposome Size
    Targeting Conjugate Before insertion After insertion
    YIGSRG-PEG-DSPE 100 nm 105 nm
    Sialyl-Lewisx-PEG-DSPE  98 nm  99 nm
  • The studies above illustrate preparation of a target-cell sensitized liposome composition by incubating pre-formed liposomes with a ligand-polymer-lipid targeting conjugate. It will be appreciated that liposomes having any composition and any selected entrapped therapeutic agent can be used in conjunction with the desired targeting conjugate. The ligand-polymer-lipid conjugate readily inserts into pre-formed liposomes in a time and temperature dependent fashion, and, as will be appreciated, is variable according to the liposome and ligand compositions. [0110]
  • III. In vivo Administration of the Composition [0111]
  • Liposomes having an E-selectin Fab fragment targeting ligand were prepared in accordance with the invention for in vivo administration to rodents. As described in Example 2, an anti-E-selectin Fab fragment was conjugated to PEG-DSPE to form an E-selectin Fab-PEG-DSPE targeting conjugate. The targeting conjugate was incubated with pre-formed [0112] 111In-labelled-liposomes composed of partially hydrogenated soy phosphatidylcholine (PHPC), PEG-DSPE and cholesterol in a 55:40:3 molar ratio in an amount sufficient to obtain 12, 20, 33, 40 and 70 Fab residues per 100 nm liposome (Example 2B). The insertion procedure resulted in greater than 95% of the targeting conjugates being inserted into the preformed liposomes. In one embodiment of the invention, the insertion efficiency is greater than 90%, more preferably greater than 95%.
  • The liposomes containing 30 Fab residues per liposome and 70 Fab residues per liposome were administered to rats to determine the blood circulation lifetime of the liposomes. As a control, [0113] 111In-labelled-liposomes of PHPC, cholesterol and PEG-DSPE (molar ratio of 55:40:3) were administered. The results are shown in FIG. 4, where the liposomes having 70 Fab residues per liposome (solid squares) and 30 Fab residues per liposomes (solid triangles) have a pharmacokinetic profile similar to that of the control liposomes (open circles). As seen, 24 hours after administration, nearly 25% of the injected dose remains in circulation in the bloodstream.
  • As described in Example 2C, pre-formed liposomes composed of hydrogenated soy phosphatidylcholine (HSPC), cholesterol, PEG-DSPE and fluorescein-labelled DHPE, in a molar ratio of 53.5/40/4/2.5, were incubated with the E-selectin-PEG-DSPE targeting conjugate at 37° C. for 1 hour. The fluorescein-labeled liposomes were administered to mice equipped with a dorsal skin fold window chamber. Endotoxin was applied topically in the [0114] window chamber 10 minutes after intravenous injection of the liposomes. FIGS. 5A-5B are scanned images of photomicrographs of the blood vessels under transmitted light prior to liposome administration (FIG. 5A) and 5 hours after administration of the target-cell sensitized, fluorescein-labeled liposomes (FIG. 5B).
  • As can be seen in FIG. 5B, the E-selectin Fab liposomes target the endothelial cells along the blood vessels. The appearance of E-selectin antigen peak was around 5 hours after endotoxin treatment, indicating that the binding activity of the E-selectin antibody was retained. [0115]
  • IV. Method of Using the Library [0116]
  • In accordance with the invention, a plurality of targeting conjugates and a plurality of liposome formulations with a variety of entrapped therapeutic agents are available for selection according to the indication to be treated. In this section, preparation and use of the library will be further demonstrated by describing suitable library components for treatment of an exemplary indication, breast cancer. [0117]
  • For treatment of human breast cancer, the library of the invention includes a plurality of targeting conjugates in the form of pre-filled vials containing the conjugate as a purified, sterile micellar suspension in an appropriate buffer. The plurality of targeting conjugates can include the following. [0118]
  • 1. Anti c-erbB-2-PEG-DSPE [0119]
  • The c-erbB-2 receptor of the HER2-neu oncogene is over-expressed in many human breast cancer cells. Humanized monoclonal antibodies have been developed which bind with high affinity to the c- erbB-2 receptor (Baselga J., et al., [0120] J. Clin Oncol., 14(3):737-44 (1996)). Single chain sFv fragments of the anti c-erbB-2 C6.5 antibody into which a terminal cysteine group is inserted are obtained as described by Schier et al. (Immunotechnology, 1(1):73-81 (1995)). The whole c-erbB-2 antibody is conjugated to PEG-DSPE having a reactive hydrazide moiety. The sFv fragment containing the terminal cysteine (and thus a free thiol group) is conjugated to PEG-DSPE-maleimide, under conditions like those described for the conjugation of the anti-E selected Fab′ antibody fragment to the same compound in Example 2.
  • 2. Anti-EGFR-PEG-DSPE Targeting Conjugate [0121]
  • Epidermal Growth Factor Receptor (EGFR) and a deletion-mutant form of EGFR (EGFRvIII) are over-expressed in certain breast cancers, gliomas and lung tumors (Beckmann, M. W., [0122] Geburtshilfe Frauenheilkd, 55(5):258-65 (1995)). Whole mouse monoclonal antibodies which bind this receptor are obtained as described by Wikstrand et al (Cancer Res., 55(14):3140-8 (1995)). These whole antibodies are conjugated to DSPE-PEG having an active hydrazide end as has been described in the art.
  • It will be appreciated that other antibody or antibody fragments which are known to bind receptors over-expressed in breast cancers cells including integrins such as a[0123] vB5 and interlukin-8 are available and can be linked to PEG-DSPE for preparation of targeting conjuates for use in the library.
  • 3. PEG-DSPE Targeting Conjugates Including a[0124] v Integrin-Binding RGD Peptides
  • Proteins that contain the Arg-Gly-Asp (RGD) attachment site, together with the integrins that serve as receptors for them, constitute a major recognition system for cell adhesion. The RGD sequence is the cell attachment site for proliferating vascular endothelial cells which form the blood supply to tumors (during angiogenesis). Such attachments are mediated by a[0125] v integrins expressed by these endothelial cells. The integrin-binding activity of matrix adhesion proteins can be reproduced by short synthetic peptides containing the RGD sequence. Reagents that bind selectively to only one or a few of the RGD-directed integrins can be designed by cyclizing peptides with selected sequences around the RGD and by synthesizing RGD mimics. Such RGD peptides can be isolated by using phage display peptide libraries (Pasqualini, R., and Ruoslahti, E., Nature, 380(6572):364-6 (1996)). Two of these peptides—one containing an av integrin-binding Arg-Gly-Asp motif and the other an Asn-Gly-Arg motif—have been identified that bind selectively to tumor vasculature. These can be linked to liposomes using the methods described herein.
  • As can be appreciated, a plurality of other PEG-DSPE conjugates of ligands, such as folate or transferin, which may bind to receptors on human breast cancer cells are prepared according to the examples set herein and by methods known in the art. [0126]
  • Continuing with the example of using the library for treatment of human breast cancer, the library further includes a therapeutic liposome composition or a plurality of liposome compositions containing encapsulated agents appropriate for treating human breast cancer cells in vivo. The pre-formed liposomes are in the form of pre-filled vials containing the liposomes as a sterile suspension in appropriate buffers is created. Liposome containing the following entrapped agents are exemplary for the human breast cancer example: doxorubicin, cisplatin, water-soluble camptothecin derivatives (e.g. topotecan, navelbine, vincristine, antisense oligonucleotides, p53 gene, HSVtk gene, a radiation sensitizer and an angiogenesis inhibitor. [0127]
  • To use the library, a targeting conjugate and a therapeutic liposome composition are selected. Selection of the targeting conjugate is based upon the expression of the conjugate's cognate receptor on individual patient's breast cancer cells. For example, it is common to test for the expression of a variety of receptors on cancer cells obtained from patients during biopsy. Clinical reference laboratories routinely screen biopsy specimens for estrogen receptor status and c-erbB-2 expression status is becoming routine with the clinical development of HERCEPTIN an anti-tumor therapeutic antibody product described by Baselga, et al, ([0128] J. Clin Oncol., (3):737-44 (1996)). Exemplary methods for determining c-erbB-2 receptor status are given by Sjogren, et al. (J. Clin Oncol., 16(2):462-9 (1998)). Patients whose tumors overexpress c-erbB-2 receptor are identified by this approach. EGFR receptor status is determined by similar methodology (Newby, J. C. et al., Br J Cancer., 71(6):1237-42 (1995)). Expression of other receptors is determined by similar methodology.
  • Next, a pre-formed therapeutic liposome composition is selected from the library. A variety of methods exist to screen for the sensitivity of breast cancer cells taken at biopsy to the cell killing effects of drugs in vitro and in vivo (chemosensitivity testing) and exemplary methods are described by Tomikawa, et al. ([0129] Anticancer Res., 18(2A):1059-62 (1998)) and by Coley, et al. (Anticancer Res. 17(1A):231-6 (1997)) and by Andreotti, et al. i Cancer Res., 55(22):5276-82 (1995)). In vitro cytotoxicity is often expressed as the concentration of a particular cancer drug needed to inhibit cancer cell proliferation by 50% in culture (IC50) In a typical screening test, cells obtained from a patient's biopsy specimen are teased apart (mechanically and/or by enzyme treatment), suspended in a medium which supports their growth and placed in wells of a culture plate. Drugs at various dilutions are added and any growth inhibition of the cells caused by the drug is measured. IC50 values are derived from these measurements. Drugs that kill the cells or inhibit growth at concentrations at or below IC50 values that can be achieved in vivo are considered as candidates for therapeutic intervention.
  • In an alternative approach, a therapeutic agent can be selected on the basis of historical information and accepted clinical practice (see for example, Handbook of Cancer Chemotherapy, 3[0130] rd edition, R. T. Skeell, editor, A, Little Brown, Boston, 1991, pp 77-138.). For example, doxorubicin is known to be one of the most active agents against human breast cancer. Therefore, in a plurality of liposome-encapsulated cancer drugs, doxorubicin would represent an obvious selection for the treatment of breast cancer based on accepted clinical practice.
  • After selection of the targeting conjugate and the therapeutic pre-formed liposome composition, the two reagents are combined to create target cell-sensitized therapeutic liposome composition tailored to an individual patient's cancer. The contents of the vial containing the conjugate and the vial containing the pre-formed therapeutic liposome composition, selected as described above and based upon the expression of the appropriate cell surface receptor and the sensitivity of the cell to growth inhibitory action of the encapsulated agent, are combined under the conditions described effective to achieve insertion of the conjugate into the liposome bilayer. Aseptic technique is used, preferably in a hospital pharmacy or other appropriate setting. Once the target-sensitized therapeutic liposome composition is formed, it is administered to the patient for which it was created. The liposomes are typically in suspension form and are administered parenterally, preferably intravenously. Other routes of administration are suitable, including subcutaneous, intramuscular, interlesional (to tumors), intertracheal by inhalation, topical, internasal, intraocular, via direct injection into organs and intravenous. [0131]
  • As can be appreciated, tailoring the formulation in this way to the individual patient maximizes the likelihood of therapeutic benefit provided by the targeting component and the encapsulated drug. [0132]
  • It will be appreciated that the dosage will depend on the liposome composition and the condition to be treated. Suitable dosages can be readily determined by those of skill in the art. [0133]
  • Use of the library will now be demonstrated for treatment of a patient suffering from a hematological disease, e.g. a B-cell or T-cell malignancy, such as B-cell leukemias/lymphomas, multiple myeloma, T-cell lymphoma and acute lymphocytic leukemia. [0134]
  • As described above, the library includes a plurality of targeting conjugates. Targeting conjugates suitable for selection include lipid-polymer-antibody conjugates, where the antibody is a monoclonal antibody or antibody fragment having a specific recognition to a B-cell or a T-cell epitope, as has been described in U.S. Pat. No. 5,620,689, which is incorporated herein by reference. For example, the antibody can be one that recognizes the B-cell epitopes CD19, CD20, CD22 or CD77. The antibody can be one that recognizes the T-cell epitopes CD4, CD7 or CD8. [0135]
  • The library further includes liposomes having entrapped agents. For treatment of hematological disorders, liposomes having the following entrapped agents are potential candidates for selection from the library: doxorubicin, vincristine, lomustine, interferon, melphalan, cyclophosphamide, prednisone, chlorambucil, carmustin and dexamethasone. [0136]
  • A blood or tissue sample is taken from the patient suffering from the hematological disorder for determination of the expression of various receptors, such as CD19, CD20, CD22, CD4, CD7, CD8. If the origin of the disorder is known to be either B-cell or T-cell, the receptor screening can of course be more selective, e.g., if the disorder is B-cell related, then the sample can be tested for expression of CD19, CD20 and CD22. Based on the results of the screening, a suitable targeting conjugate is chosen. [0137]
  • A therapeutic agent for treatment of the disorder is selected from the library using the procedures described in the breast cancer example above. [0138]
  • The selected conjugate and liposome composition are incubated together as described above to form the target-cell sensitized, therapeutic liposome composition specific for the patient. Suitable dosages for the composition can be initially based on the standard chemotherapeutic dose and adjusted accordingly over the course of treatment by monitoring the disease progression. [0139]
  • V. EXAMPLES
  • The following examples illustrate methods of preparing the composition of the present invention. The examples are in no way intended to limit the scope of the invention. [0140]
  • Example 1 Preparation of Pre-Formed Liposomes and Insertion of Targeting Conjugate
  • Liposomes were prepared by mixing partially hydrogenated soy-bean phosphatiylcholine (PHPC, iodine value of 35, Lipoid (Ludwigshafen, Germany)), cholesterol (Croda (Fullerton, Calif.)) and mPEG-DSPE (prepared as described in Zalipsky, S., et al., [0141] Bioconjugate Chemistry, 4:296-299 (1993)) at a molar ratio of 55:40:3 in chloroform and/or methanol in a round bottom flask. The solvents were removed by rotary evaporation, and the dried lipid film produced was hydrated with either sodium phosphate buffer (10 mM, 140 mM NaCl, pH 7) or HEPES buffer (25 mM, 150 mM NaCl, pH 7) to produce large multilamellar vesicles. The resulting vesicles were passed repeatedly under pressure through 0.2, 0.1 and 0.05 m pore size polycarbonate membranes, until the average size distribution for the diameter (monitored by dynamic light scattering using a Coulter N4MD (Hialeah, Fla.)) was approximately 100 nm. The mean particle diameter measured from 12 different batches ranged form 92 to 111 nm with an average 98 nm.
  • Targeting conjugates of and YIGSRG-PEG-DSPE were prepared according to Zalipsky, S., et al., [0142] Bioconjugate Chemistry, 8(2):111-118 (1997).
  • The pre-formed liposomes were incubated at either 25° C. or 37° C. with 1.2 mole percent of one of the targeting conjugates. At various time points, targeting conjugates (micelles) were separated from inserted targeting conjugates (liposomes) by size exclusion chromatography. For the sialyl-Lewis[0143] x-PEG-DSPE conjugate, a Biogel A50M column equilibrated with 10 mM sodium phosphate, 140 mM sodium chloride, and 0.02% NaN3 at pH 6.5 was used. For YIGSRG-PEG-DSPE conjugate, a Sepharose 4B column was used with 10% sucrose and 10 mM HEPES at pH 7.0 as eluent. The results for the sialyl-Lewisx-PEG-DSPE conjugate are shown in FIGS. 2A-2D for the 0, 1, 3 and 5 hour time points, where the peak centered around fraction 10 corresponds to the liposomes and the peak centered around fraction 20 corresponds to the micellular, targeting conjugate.
  • The collected fractions (1 mL) from the size exclusion chromatograph were diluted 1:10 in methanol, and analyzed for ligand content by HPLC (Shimadzu and Rainin systems), with the results shown in FIG. 3. [0144]
  • Example 2 Preparation of Anti-E-Selectin Fab Conjugate and Insertion into Pre-Formed Liposomes
  • A. Preparation of the Targeting Conjugate [0145]
  • An anti-E-selectin Fab fragment was conjugated to PEG-DSPE to form a targeting conjugate as follows. An aqueous solution of 750 mM 2-mercaptoethylamine as a reducing agent was prepared. 10 μl of the mercaptoethylamine was added to 1 ml of 5 mg/ml anti E-selectin Fab fragment in 50 μM sodium acetate and 125 mM NaCl, pH=5.0. The final concentration of reducing agent was 7.5 mM. The solution was incubated at 37° C. for 30 minutes. The excess reducing agent was removed on a 10DG-column (Bio-Rad) equilibrated with 25 mM HEPES/0.9% saline buffer. The collected fractions were analyzed spectrophotometrically to determine the fractions containing the Fab fragments. These fractions were pooled and diluted 1:50 in phosphate buffered saline to determine the protein concentration. [0146]
  • The Fab fragments (molecular weight of 3,000 Daltons) were mixed in a 1:1 molar ratio with PEG-DSPE (molecular weight 50,000 daltons) having an active maleimide end group (prepared as described in U.S. Pat. No. 5,527,528). The two components were incubated overnight at room temperature. The unreacted maleimide was quenched with 2 mM β-mercaptoethanol for 30 minutes at room temperature. The free Fab fragments and β-mercaptoethanol were separated from the Fab-PEG-DSPE conjugate on an S-200 column equilibrated in 25 mM HEPES/0.9% saline at pH 7.2. Fractions of 1 ml were collected and read on the spectrophotometer at 280 nm to determine the fractions containing the conjugate and the free Fab fragments. The fractions were pooled accordingly and the concentration of the Fab-PEG-DSPE micellular solution is determined spectrophotometrically (280 nm). The efficiency of conjugation of the Fab fragment to the maleimide-PEG-DSPE was approximately 40%. [0147]
  • B. Insertion of the Conjugate into Pre-Formed Liposomes [0148]
  • Liposomes of partially hydrogenated soy phosphatidylcholine (PHPC), PEG-DSPE and cholesterol in a 55:40:3 molar ratio were prepared as described in Example 1. Depending on the desired number of targeting ligands per liposomes, an amount of the Fab-PEG-DSPE conjugate was added to a suspension of liposomes and incubated overnight at room temperature. A 100 μl aliquot of the insertion mixture was taken and placed on a SEPHAROSE 4B column (0.7×30 cm) to separate the free Fab conjugate from the liposomes. 1 ml fractions were collected and read on the spectrophotometer to determine the amount of conjugate inserted into the pre-formed liposomes. Greater than 95% of the conjugates were inserted into the pre-formed liposomes. [0149]
  • Following insertion of the targeting conjugate, an aliquiot of the liposomes were analyzed by amino acid analysis to determine the protein concentration. Another aliquot was analyzed for phosphorus content. Based on these values, the amount of protein per liposome was determined. [0150]
  • Using this insertion procedure, liposomes containing 12, 20, 33, 40 and 70 Fab residues per 100 nm liposome, as determined by amino acid analysis, were prepared. [0151]
  • [0152] 111In-labelled-liposomes containing 30 Fab residues per liposome and 70 Fab residues per liposome were administered to rats to determine the blood circulation lifetime of the liposomes. As a control, 111In-labelled-liposomes of PHPC, cholesterol and PEG-DSPE (molar ratio of 55:40:3) were administered. The results are shown in FIG. 4.
  • C. In vivo Targeting [0153]
  • E-selectin Fab-PEG-DSPE targeting conjugate was inserted into pre-formed liposomes as follows. The pre-formed liposomes were composed of hydrogenated soy phosphatidylcholine (HSPC), cholesterol and PEG-DSPE in a molar ratio of 53.5/40/4. The liposomes included 2.5 mole percent of the lipid marker of fluorescein-DHPE (Molecular Probes, Inc.). The preformed liposomes were incubated with the micellular solution of the targeting conjugate at 37° C. for 1 hour. The insertion mixture was placed on a Bio-Rad A50 m column equilibrated with 25 mM HEPES/saline pH 7.2 and 0.5 ml fractions were collected. Spectrophotometric analysis of the fractions indicated that the insertion efficiency of the Fab targeting conjugate into the liposomes was approximately 100% after 2 hours at 37° C. [0154]
  • The fluorescein-labeled liposomes were administered to mice equipped with a window chamber in a dorsal skin fold. Endotoxin was applied topically in the [0155] window chamber 10 minutes after intravenous injection of the liposomes. FIGS. 5A-5B are photomicrographs (scanned images) of the blood vessels under transmitted light prior to liposome administration (FIG. 5A) and 5 hours after administration of the target-cell sensitized, fluorescein-labeled liposomes (FIG. 5B).
  • Although the invention has been described with respect to particular embodiments, it will be apparent to those skilled in the art that various changes and modifications can be made without departing from the invention. [0156]

Claims (56)

It is claimed:
1. Reagents for use in preparing a therapeutic liposome composition sensitized to a target cell, said reagents comprising
a liposomal composition composed of pre-formed liposomes having an entrapped therapeutic agent; and
a plurality of conjugates, each conjugate composed of (i) a lipid having a polar head group and a hydrophobic tail, (ii) a hydrophilic polymer having a proximal end and a distal end, said polymer attached at its proximal end to the head group of the lipid, and (iii) a targeting ligand attached to the distal end of the polymer;
wherein the therapeutic, target-cell sensitized liposome composition is formed by incubating the liposomal composition with a selected conjugate.
2. The composition of claim 1, wherein the targeting ligand is an antibody or an antibody fragment.
3. The composition of claim 2, wherein the antibody or antibody fragment is of mouse origin and is humanized to remove murine epitopes.
4. The composition of claim 2, wherein the targeting ligand specifically binds to an extracellular domain of a growth factor receptor.
5. The composition of claim 4, wherein the receptors are selected from the group consisting of c-erbB-2 protein product of the HER2/neu oncogene, epidermal growth factor receptor, basic fibroblast growth factor receptor, and vascular endothelial growth factor receptor.
6. The composition of claim 2, wherein the targeting ligand binds a receptor selected from the group consisting of E-selectin receptor, L-selectin receptor, P-selectin receptor, folate receptor, CD4 receptor, CD19 receptor, αβ integrin receptors and chemokine receptors.
7. The composition of claim 1, wherein the targeting ligand is selected from the group consisting of folic acid, pyridoxal phosphate, vitamin B12, sialyl Lewisx, transferrin, epidermal growth factor, basic fibroblast growth factor, vascular endothelial growth factor, VCAM-1, ICAM-1, PECAM-1, RGD peptides and NGR peptides.
8. The composition of claim 1, wherein the targeting ligand binds a receptor on a malignant B-cell or T-cell, said receptor selected from the group consisting of CD19, CD20, CD22, CD4, CD7 and CD8.
9. The composition of claim 1, wherein the hydrophilic polymer is selected from the group consisting of polyvinylpyrrolidone, polyvinylmethylether, polymethyloxazoline, polyethyloxazoline, polyhydroxypropyloxazoline, polyhydroxypropylmethacrylamide, polymethacrylamide, polydimethylacrylamide, polyhydroxypropylmethacrylate, polyhydroxyethylacrylate, hydroxymethylcellulose, hydroxyethylcellulose, polyethyleneglycol, polyaspartamide and hydrophilic peptide sequences.
10. The composition of claim 1, wherein the hydrophilic polymer is polyethylene glycol.
11. The composition of claim 10, wherein the polyethylene glycol has a molecular weight between 500-5,000 daltons.
12. The composition of claim 1, wherein the liposomes further contain a cationic lipid.
13. The composition of claim 1, wherein the entrapped therapeutic agent is a cytotoxic drug.
14. The composition of claim 13, wherein the cytotoxic drug is an anthracycline antibiotic selected from the group consisting of doxorubicin, daunorubicin, epirubicin and idarubicin and analogs thereof.
15. The composition of claim 13, wherein the cytotoxic agent is a platinum compound selected from cisplatin, carboplatin, ormaplatin, oxaliplatin, zeniplatin, enloplatin, lobaplatin, spiroplatin, ((−)-(R)-2-aminomethylpyrrolidine (1,1-cyclobutane dicarboxylato)platinum), (SP-4-3(R)-1,1-cyclobutane-dicarboxylato(2-)-(2-methyl-1,4-butanediamine-N,N′)platinum), nedaplatin and (bis-acetato-ammine-dichloro-cyclohexylamine-platinum(IV)).
16. The composition of claim 13, wherein the cytotoxic agent is a topoisomerase 1 inhibitor selected from the group consisting of topotecan, irinotecan, (7-(4-methylpiperazino-methylene)-10,11-ethylenedioxy-20(S)-camptothecin), 7-(2-(N-isopropylamino)ethyl)-(20S)-camptothecin, 9-aminocamptothecin and 9-nitrocamptothecin.
17. The composition of claim 13, wherein the cytotoxic agent is a vinca alkaloid selected from the group consisting of vincristine, vinblastine, vinleurosine, vinrodisine, vinorelbine and vindesine.
18. The composition of claim 1, wherein the entrapped agent is a nucleic acid.
19. The composition of claim 18, wherein the nucleic acid is an antisense oligonucleotide or ribozyme.
20. The composition of claim 18, wherein the nucleic acid is a plasmid containing a therapeutic gene which when internalized by the target cells achieves expression of the therapeutic gene to produce a therapeutic gene product.
21. A plurality of targeting conjugates for use in preparing a targeted, therapeutic liposome composition, each conjugate composed of a (i) a lipid having a polar head group and a hydrophobic tail, (ii) a hydrophilic polymer having a proximal end and a distal end, said polymer attached at its proximal end to the head group of the lipid, and (iii) a targeting ligand attached to the distal end of the polymer.
22. The conjugates of claim 21, wherein the lipid is selected from the group consisting of distearoyl phosphatidylethanolamine, distearoyl-phosphatidylcholine, monogalactosyl diacylglycerols and digalactosyl diacylglycerols.
23. The conjugates of claim 21, wherein the hydrophilic polymer is selected from the group consisting of polyvinylpyrrolidone, polyvinylmethylether, polymethyloxazoline, polyethyloxazoline, polyhydroxypropyloxazoline, polyhydroxypropylmethacrylamide, polymethacrylamide, polydimethylacrylamide, polyhydroxypropylmethacrylate, polyhydroxyethylacrylate, hydroxymethylcellulose, hydroxyethylcellulose, polyethyleneglycol, polyaspartamide and hydrophilic peptide sequences.
24. The conjugates of claim 21, wherein the hydrophilic polymer is polyethylene glycol.
25. The conjugates of claim 24, wherein the polyethylene glycol has a molecular weight between 500-5,000 daltons.
26. The conjugates of claim 21, wherein the targeting ligand is an antibody or an antibody fragment.
27. The conjugates of claim 26, wherein the antibody or antibody fragment is of mouse origin and is humanized to remove murine epitopes.
28. The conjugates of claim 21, wherein the targeting ligand specifically binds to an extracellular domain of a growth factor receptor.
29. The conjugates of claim 28, wherein the receptors are selected from the group consisting of c-erbB-2 protein product of the HER2/neu oncogene, epidermal growth factor receptor, basic fibroblast growth factor receptor, and vascular endothelial growth factor receptor.
30. The conjugates of claim 21, wherein the targeting ligand binds a receptor selected from the group consisting of E-selectin receptor, L-selectin receptor, P-selectin receptor, folate receptor, CD4 receptor, CD19 receptor, αβ integrin receptors and chemokine receptors.
31. The conjugates of claim 21, wherein the targeting ligand binds a receptor on a malignant B-cell or T-cell, said receptor selected from the group consisting of CD19, CD20, CD22, CD4, CD7 and CD8.
32. The conjugates of claim 21, wherein the targeting ligand is selected from the group consisting of folic acid, pyridoxal phosphate, vitamin B12, sialyl Lewisx, transferrin, epidermal growth factor, basic fibroblast growth factor, vascular endothelial growth factor, VCAM-1, ICAM-1, PECAM-1, RGD peptides and NGR peptides.
33. A method of formulating a therapeutic liposome composition having sensitivity to a target cell, comprising
selecting a liposomal composition composed of pre-formed liposomes having an entrapped therapeutic agent;
selecting from a plurality of targeting conjugates a targeting conjugate composed of (i) a lipid having a polar head group and a hydrophobic tail, (ii) a hydrophilic polymer having a proximal end and a distal end, said polymer attached at its proximal end to the head group of the lipid, and (iii) a targeting ligand attached to the distal end of the polymer; and
combining the selected liposome formulation and the selected targeting conjugate to form said therapeutic, target-cell sensitive liposome composition.
34. The method of claim 33, wherein said combining includes incubating under conditions effective to achieve insertion of the selected targeting conjugate into the liposomes of the selected liposome formulation.
35. The method of claim 33, wherein said selecting a liposome formulation includes determining the sensitivity of the target cell to the therapeutic activity of the entrapped therapeutic agent.
36. The method of claim 33, wherein said selecting a targeting conjugate includes determining the ability of the targeting ligand to bind cell surface receptors expressed on the target cell.
37. The method of claim 36, wherein said selecting a targeting conjugate is based on (i) the ability of a targeting ligand to bind to cell surface receptors expressed on the target cell and (ii) the ability of the target cell to internalize liposomes bound to the target cell by binding between the target cell and the targeting ligand.
38. The method of claim 33, wherein the targeting ligand is an antibody or an antibody fragment.
39. The method of claim 38, wherein the antibody or antibody fragment is of mouse origin and is humanized to remove murine epitopes.
40. The method of claim 38, wherein the targeting ligand specifically binds to an extracellular domain of a growth factor receptor.
41. The method of claim 40, wherein the receptors are selected from the group consisting of c-erbB-2 protein product of the HER2/neu oncogene, epidermal growth factor receptor, basic fibroblast growth factor receptor, and vascular endothelial growth factor receptor.
42. The method of claim 38, wherein the targeting ligand binds a receptor selected from the group consisting of E-selectin receptor, L-selectin receptor, P-selectin receptor, folate receptor, CD4 receptor, CD19 receptor, αβ integrin receptors and chemokine receptors.
43. The method of claim 33, wherein the targeting ligand binds a receptor on a malignant B-cell or T-cell, said receptor selected from the group consisting of CD19, CD20, CD22, CD4, CD7 and CD8.
44. The method of claim 33, wherein the targeting ligand is selected from the group consisting of folic acid, pyridoxal phosphate, vitamin B12, sialyl Lewisx, transferrin, epidermal growth factor, basic fibroblast growth factor, vascular endothelial growth factor, VCAM-1, ICAM-1, PECAM-1, RGD peptides and NGR peptides.
45. The method of claim 33, wherein the hydrophilic polymer is selected from the group consisting of polyvinylpyrrolidone, polyvinylmethylether, polymethyloxazoline, polyethyloxazoline, polyhydroxypropyloxazoline, polyhydroxypropylmethacrylamide, polymethacrylamide, polydimethylacrylamide, polyhydroxypropylmethacrylate, polyhydroxyethylacrylate, hydroxymethylcellulose, hydroxyethylcellulose, polyethyleneglycol, polyaspartamide and hydrophilic peptide sequences.
46. The method of claim 33, wherein the hydrophilic polymer is polyethylene glycol.
47. The method of claim 46, wherein the polyethylene glycol has a molecular weight between 500-5,000 daltons.
48. The method of claim 33, wherein the liposomes further contain a cationic lipid.
49. The method of claim 33, wherein the entrapped therapeutic agent is a cytotoxic drug.
50. The method of claim 49 wherein the cytotoxic drug is an anthracycline antibiotic selected from the group consisting of doxorubicin, daunorubicin, epirubicin and idarubicin and analogs thereof.
51. The method of claim 49, wherein the cytotoxic agent is a platinum compound selected from cisplatin, carboplatin, ormaplatin, oxaliplatin, zeniplatin, enloplatin, lobaplatin, spiroplatin, ((−)-(R)-2-aminomethylpyrrolidine (1,1-cyclobutane dicarboxylato)platinum), (SP-4-3(R)-1,1-cyclobutane-dicarboxylato(2-)-(2-methyl-1,4-butanediamine-N,N′)platinum), nedaplatin and (bis-acetato-ammine-dichloro-cyclohexylamine-platinum(IV)).
52. The method of claim 49, wherein the cytotoxic agent is a topoisomerase 1 inhibitor selected from the group consisting of topotecan, irinotecan, (7-(4-methylpiperazino-methylene)-10,11-ethylenedioxy-20(S)-camptothecin), 7-(2-(N-isopropylamino)ethyl)-(20S)-camptothecin, 9-aminocamptothecin and 9-nitrocamptothecin.
53. The method of claim 49, wherein the cytotoxic agent is a vinca alkaloid selected from the group consisting of vincristine, vinblastine, vinleurosine, vinrodisine, vinorelbine and vindesine.
54. The method of claim 33, wherein the entrapped agent is a nucleic acid.
55. The method of claim 54, wherein the nucleic acid is an antisense oligonucleotide or ribozyme.
56. The method of claim 54, wherein the nucleic acid is a plasmid containing a therapeutic gene which when internalized by the target cells achieves expression of the therapeutic gene to produce a therapeutic gene product.
US10/115,566 1996-10-11 2002-04-02 Therapeutic liposome composition and method of preparation Abandoned US20030215490A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/115,566 US20030215490A1 (en) 1996-10-11 2002-04-02 Therapeutic liposome composition and method of preparation

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US2826996P 1996-10-11 1996-10-11
US08/949,046 US5891468A (en) 1996-10-11 1997-10-10 Fusogenic liposome compositions and method
US09/138,480 US6056973A (en) 1996-10-11 1998-08-21 Therapeutic liposome composition and method of preparation
US09/517,224 US6316024B1 (en) 1996-10-11 2000-03-02 Therapeutic liposome composition and method of preparation
US09/876,707 US7122202B2 (en) 1996-10-11 2001-06-07 Therapeutic liposome composition and method of preparation
US10/115,566 US20030215490A1 (en) 1996-10-11 2002-04-02 Therapeutic liposome composition and method of preparation

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/876,707 Continuation US7122202B2 (en) 1996-10-11 2001-06-07 Therapeutic liposome composition and method of preparation

Publications (1)

Publication Number Publication Date
US20030215490A1 true US20030215490A1 (en) 2003-11-20

Family

ID=46203431

Family Applications (10)

Application Number Title Priority Date Filing Date
US09/138,480 Expired - Lifetime US6056973A (en) 1996-10-11 1998-08-21 Therapeutic liposome composition and method of preparation
US09/517,224 Expired - Lifetime US6316024B1 (en) 1996-10-11 2000-03-02 Therapeutic liposome composition and method of preparation
US09/876,707 Expired - Fee Related US7122202B2 (en) 1996-10-11 2001-06-07 Therapeutic liposome composition and method of preparation
US10/016,324 Expired - Lifetime US6936272B2 (en) 1996-10-11 2001-12-10 10139483Therapeutic liposome composition and method of preparation
US10/115,566 Abandoned US20030215490A1 (en) 1996-10-11 2002-04-02 Therapeutic liposome composition and method of preparation
US10/821,021 Abandoned US20040191307A1 (en) 1996-10-11 2004-04-07 Therapeutic liposome composition and method of preparation
US10/821,018 Abandoned US20040191250A1 (en) 1996-10-11 2004-04-07 Therapeutic liposome composition and method of preparation
US11/049,848 Abandoned US20050136064A1 (en) 1996-10-11 2005-02-02 Therapeutic liposome composition and method of preparation
US11/050,012 Abandoned US20050169980A1 (en) 1996-10-11 2005-02-02 Therapeutic liposome composition and method of preparation
US11/479,437 Abandoned US20060246126A1 (en) 1996-10-11 2006-06-30 Therapeutic liposome composition and method of preparation

Family Applications Before (4)

Application Number Title Priority Date Filing Date
US09/138,480 Expired - Lifetime US6056973A (en) 1996-10-11 1998-08-21 Therapeutic liposome composition and method of preparation
US09/517,224 Expired - Lifetime US6316024B1 (en) 1996-10-11 2000-03-02 Therapeutic liposome composition and method of preparation
US09/876,707 Expired - Fee Related US7122202B2 (en) 1996-10-11 2001-06-07 Therapeutic liposome composition and method of preparation
US10/016,324 Expired - Lifetime US6936272B2 (en) 1996-10-11 2001-12-10 10139483Therapeutic liposome composition and method of preparation

Family Applications After (5)

Application Number Title Priority Date Filing Date
US10/821,021 Abandoned US20040191307A1 (en) 1996-10-11 2004-04-07 Therapeutic liposome composition and method of preparation
US10/821,018 Abandoned US20040191250A1 (en) 1996-10-11 2004-04-07 Therapeutic liposome composition and method of preparation
US11/049,848 Abandoned US20050136064A1 (en) 1996-10-11 2005-02-02 Therapeutic liposome composition and method of preparation
US11/050,012 Abandoned US20050169980A1 (en) 1996-10-11 2005-02-02 Therapeutic liposome composition and method of preparation
US11/479,437 Abandoned US20060246126A1 (en) 1996-10-11 2006-06-30 Therapeutic liposome composition and method of preparation

Country Status (1)

Country Link
US (10) US6056973A (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040191250A1 (en) * 1996-10-11 2004-09-30 Alza Corporation Therapeutic liposome composition and method of preparation
US20080063621A1 (en) * 2006-09-07 2008-03-13 Canadian Blood Services Surface cross-linked lipidic particles, methods of production and uses therefor
US7829113B2 (en) 2005-03-10 2010-11-09 Mebiopharm Co., Ltd. Liposome compositions
WO2014015027A1 (en) 2012-07-18 2014-01-23 Onyx Therapeutics, Inc. Liposomal compositions of epoxyketone-based proteasome inhibitors
US9878044B2 (en) 2012-03-16 2018-01-30 Merck Patent Gmbh Targeting aminoacid lipids
WO2018087720A1 (en) 2016-11-14 2018-05-17 Novartis Ag Compositions, methods, and therapeutic uses related to fusogenic protein minion

Families Citing this family (352)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6008202A (en) * 1995-01-23 1999-12-28 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US5795587A (en) * 1995-01-23 1998-08-18 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US5739313A (en) * 1995-11-13 1998-04-14 Regents Of The University Of Minnesota Radionuclide labeling of vitamin B12 and coenzymes thereof
US6210707B1 (en) 1996-11-12 2001-04-03 The Regents Of The University Of California Methods of forming protein-linked lipidic microparticles, and compositions thereof
US6506783B1 (en) * 1997-05-16 2003-01-14 The Procter & Gamble Company Cancer treatments and pharmaceutical compositions therefor
US6537520B1 (en) * 1998-03-31 2003-03-25 Bristol-Myers Squibb Pharma Company Pharmaceuticals for the imaging of angiogenic disorders
US6548663B1 (en) 1998-03-31 2003-04-15 Bristol-Myers Squibb Pharma Company Benzodiazepine vitronectin receptor antagonist pharmaceuticals
US6524553B2 (en) * 1998-03-31 2003-02-25 Bristol-Myers Squibb Pharma Company Quinolone vitronectin receptor antagonist pharmaceuticals
KR20010042288A (en) 1998-03-31 2001-05-25 블레어 큐. 퍼거슨 Pharmaceuticals For The Imaging Of Angiogenic Disorders
US20040022788A1 (en) * 1998-05-19 2004-02-05 Moser Tammy L. Compositions and methods for promoting or inhibiting angiogenesis
US20040247662A1 (en) * 1998-06-25 2004-12-09 Dow Steven W. Systemic immune activation method using nucleic acid-lipid complexes
US20030022854A1 (en) * 1998-06-25 2003-01-30 Dow Steven W. Vaccines using nucleic acid-lipid complexes
US6297245B1 (en) * 1998-08-04 2001-10-02 Unitech Pharmaceuticals Cisplatin and folic acid administered to treat breast cancer
CA2346879A1 (en) * 1998-09-16 2000-04-27 Alza Corporation Liposome-entrapped topoisomerase inhibitors
US6569402B1 (en) * 1998-12-18 2003-05-27 Bristol-Myers Squibb Pharma Company Vitronectin receptor antagonist pharmaceuticals
BR9917079A (en) 1998-12-18 2001-10-30 Du Pont Pharm Co Devitronectin receptor antagonist compounds, kit, diagnostic or therapeutic metallopharmaceutical composition, ultrasound contrast agent composition, therapeutic radiopharmaceutical composition, diagnostic pharmaceutical composition, treatment method for rheumatoid arthritis, cancer and darthenosis in a patient, angiogenesis formation method therapy, cancer, new blood vessels, arteriosclerosis, darthenosis, ischemia and myocardial reperfusion injury in a patient
US6511649B1 (en) * 1998-12-18 2003-01-28 Thomas D. Harris Vitronectin receptor antagonist pharmaceuticals
DE69938148T2 (en) * 1998-12-18 2009-02-26 Alza Corp., Mountain View METHOD FOR ADMINISTERING A CONNECTION TO MULTIPLE RESISTANT CELLS
US6794518B1 (en) * 1998-12-18 2004-09-21 Bristol-Myers Squibb Pharma Company Vitronectin receptor antagonist pharmaceuticals
AU2371400A (en) 1998-12-18 2000-07-03 Du Pont Pharmaceuticals Company Vitronectin receptor antagonist pharmaceuticals
US7311924B2 (en) * 1999-04-01 2007-12-25 Hana Biosciences, Inc. Compositions and methods for treating cancer
IT1306129B1 (en) * 1999-04-13 2001-05-30 Sigma Tau Ind Farmaceuti ESTERS OF L-CARNITINE OR ALCANOYL L-CARNITINE USABLE CATIONIC COMELIPIDS FOR INTRACELLULAR PLACING OF COMPOUNDS
US6806363B1 (en) * 1999-04-16 2004-10-19 Mayo Foundation For Medical Education & Research Cobalamin conjugates useful as antitumor agents
US6852334B1 (en) * 1999-04-20 2005-02-08 The University Of British Columbia Cationic peg-lipids and methods of use
WO2001026625A2 (en) * 1999-10-08 2001-04-19 Alza Corp Neutral-cationic lipid for nucleic acid and drug delivery
US7591995B2 (en) * 1999-10-15 2009-09-22 Mayo Foundation For Medical Education And Research Cobalamin conjugates useful as imaging and therapeutic agents
US20040009229A1 (en) * 2000-01-05 2004-01-15 Unger Evan Charles Stabilized nanoparticle formulations of camptotheca derivatives
WO2001064743A1 (en) * 2000-03-02 2001-09-07 Mitsubishi Pharma Corporation GPib-LIPID BOND CONSTRUCT AND USE THEREOF
US7189705B2 (en) * 2000-04-20 2007-03-13 The University Of British Columbia Methods of enhancing SPLP-mediated transfection using endosomal membrane destabilizers
US10293056B1 (en) * 2000-05-24 2019-05-21 Board Of Regents, The University Of Texas System Methods and compositions for non-viral gene therapy for treatment of hyperproliferative diseases
US20030059461A1 (en) * 2000-06-06 2003-03-27 Sibtech, Inc. Molecular delivery vehicle for delivery of selected compounds to targets
US7355019B2 (en) * 2000-06-06 2008-04-08 Sibtech, Inc. Cysteine-containing peptide tag for site-specific conjugation of proteins
EP1299085B1 (en) * 2000-06-30 2005-11-16 Inex Pharmaceuticals Corp. Improved liposomal camptothecins and uses thereof
US20060177416A1 (en) 2003-10-14 2006-08-10 Medivas, Llc Polymer particle delivery compositions and methods of use
US6462062B1 (en) * 2000-09-26 2002-10-08 The Procter & Gamble Company Compounds and methods for use thereof in the treatment of cancer or viral infections
EP1323415B1 (en) * 2000-10-04 2010-01-13 Kyowa Hakko Kirin Co., Ltd. Method of coating fine particle with lipid film
AU2001297913A1 (en) * 2000-10-13 2002-12-23 Ligocyte Pharmaceuticals, Inc. Polyvalent nanoparticles
TWI228512B (en) * 2000-10-25 2005-03-01 Mayo Foundation Transcobalamin binding conjugates useful for treating abnormal cellular proliferation
WO2002036073A2 (en) * 2000-11-02 2002-05-10 Smithkline Beecham Corporation Receptor antagonist-lipid conjugates and delivery vehicles containing same
WO2002058622A2 (en) * 2000-11-09 2002-08-01 Neopharm, Inc. Sn-38 lipid complexes and methods of use
US6497896B2 (en) 2001-02-12 2002-12-24 Supergen, Inc. Method for administering camptothecins via injection of a pharmaceutical composition comprising microdroplets containing a camptothecin
US6509027B2 (en) 2001-02-12 2003-01-21 Supergen, Inc. Injectable pharmaceutical composition comprising coated particles of camptothecin
IL158160A0 (en) * 2001-03-26 2004-03-28 Alza Corp Liposome composition for improved intracellular delivery of a therapeutic agent
EP1389089B1 (en) * 2001-03-27 2009-08-26 Phares Pharmaceutical Research N.V. Method and composition for solubilising a biologically active compound with low water solubility
WO2002085908A1 (en) 2001-04-24 2002-10-31 Purdue Research Foundation Folate mimetics and folate-receptor binding conjugates thereof
EP1383480A4 (en) * 2001-04-30 2006-05-24 Targeted Genetics Corp Lipid-comprising drug delivery complexes and methods for their production
WO2003030864A1 (en) * 2001-05-29 2003-04-17 Neopharm, Inc. Liposomal formulation of irinotecan
CN100384480C (en) * 2001-05-30 2008-04-30 斯克里普斯研究学院 Delivery system for nucleic acids
US20060074034A1 (en) * 2001-09-17 2006-04-06 Collins Douglas A Cobalamin mediated delivery of nucleic acids, analogs and derivatives thereof
US20030144198A1 (en) * 2001-09-28 2003-07-31 Collins Douglas A. Coadministration of transport protein with conjugated cobalamin to deliver agents
US6476068B1 (en) 2001-12-06 2002-11-05 Pharmacia Italia, S.P.A. Platinum derivative pharmaceutical formulations
US20060193906A1 (en) * 2002-01-30 2006-08-31 National Institute Of Advanced Industrial Science And Technology Sugar-modified liposome and products comprising the liposome
CA2503094A1 (en) * 2002-02-27 2003-09-04 The Ohio State University Research Foundation Therapeutic methods for acute myeloid leukemia
US9770517B2 (en) 2002-03-01 2017-09-26 Immunomedics, Inc. Anti-Trop-2 antibody-drug conjugates and uses thereof
AU2003233396B2 (en) * 2002-03-13 2007-05-24 Thomas Skold Water-based delivery systems
MXPA04008937A (en) 2002-03-15 2005-06-17 Schering Corp Methods of modulating cd200 receptors.
US20050209252A1 (en) * 2002-03-29 2005-09-22 Che-Ming Teng Cancer treatment
US20040009216A1 (en) * 2002-04-05 2004-01-15 Rodrigueza Wendi V. Compositions and methods for dosing liposomes of certain sizes to treat or prevent disease
KR20040106547A (en) * 2002-05-15 2004-12-17 엔도사이트, 인코포레이티드 Vitamin-mitomycin conjugates
FR2840532B1 (en) * 2002-06-11 2005-05-06 Ethypharm Sa FURENT LIPID NANOCAPSULES, PROCESS FOR THEIR PREPARATION AND USE AS VECTOR OF ACTIVE (S) PRINCIPLES
US20050129769A1 (en) * 2002-06-03 2005-06-16 Barry Stephen E. Polymeric articles for carrying therapeutic agents
JP3415131B1 (en) * 2002-06-03 2003-06-09 メビオファーム株式会社 Liposome preparation
US20040022842A1 (en) * 2002-06-03 2004-02-05 Mebiopharm Co., Ltd. Liposome preparations containing oxaliplatin
KR100440725B1 (en) * 2002-06-20 2004-07-15 주식회사 그린진 바이오텍 A Method for Increasing an Abiotic-Resistance in Monocot Plants
EP1393719A1 (en) * 2002-08-23 2004-03-03 Munich Biotech AG Camptothecin-carboxylate formulations
AU2003241598B2 (en) * 2002-07-02 2009-11-05 Nanotx Corp. Radiolabeled compounds and liposomes and their methods of making and using the same
US20040224986A1 (en) 2002-08-16 2004-11-11 Bart De Corte Piperidinyl targeting compounds that selectively bind integrins
AU2003296897A1 (en) * 2002-08-20 2004-05-04 Neopharm, Inc. Pharmaceutical formulations of camptothecine derivatives
US20060030578A1 (en) * 2002-08-20 2006-02-09 Neopharm, Inc. Pharmaceutically active lipid based formulation of irinotecan
WO2004017943A2 (en) * 2002-08-23 2004-03-04 Medigene Oncology Gmbh Non-vesicular cationic lipid formulations
EP1565165A2 (en) * 2002-11-26 2005-08-24 Gilead Sciences, Inc. Liposomal formulations
JP4959136B2 (en) 2002-12-13 2012-06-20 イミューノメディクス、インコーポレイテッド Immunoconjugates with cleavable linkages in cells
US8420086B2 (en) * 2002-12-13 2013-04-16 Immunomedics, Inc. Camptothecin conjugates of anti-CD22 antibodies for treatment of B cell diseases
KR20050090987A (en) * 2002-12-19 2005-09-14 알자 코포레이션 Method of treating angiogenic tissue growth
TWI309571B (en) * 2002-12-31 2009-05-11 Ind Tech Res Inst Delivery carrier for targeting cells haring over-expressed estrogen
US8980310B2 (en) * 2002-12-31 2015-03-17 Bharat Serums and Vaccines, Ltd. Non-pegylated long-circulating liposomes
EP1583562B1 (en) 2003-01-06 2011-06-15 Angiochem Inc. Angiopep-1, related compounds, and uses thereof
WO2004069159A2 (en) * 2003-01-27 2004-08-19 Endocyte, Inc. Vitamin receptor binding drug delivery conjugates
WO2004070009A2 (en) * 2003-01-31 2004-08-19 Targesome Inc. Targeted multivalent macromolecules
ATE538776T1 (en) 2003-02-14 2012-01-15 Childrens Hosp & Res Ct Oak CARRIER FOR RELEASE OF LIPOPHILIC ACTIVE INGREDIENTS AND METHOD OF USE THEREOF
US20040171175A1 (en) * 2003-02-28 2004-09-02 Swanson Basil I. Process for conjugating biomolecules to hydrophobic membrane-incorporated molecules
US7160908B2 (en) * 2003-03-04 2007-01-09 Unitech Pharmaceuticals, Inc. Dynamic anticancer platinum compounds
AU2004219625B9 (en) 2003-03-10 2010-12-23 Merck Sharp & Dohme Corp. Uses of IL-23 agonists and antagonists; related reagents
WO2004082626A2 (en) * 2003-03-18 2004-09-30 Ethicon, Inc. Aromatase inhibitor diagnosis and therapy
WO2004089339A2 (en) 2003-03-31 2004-10-21 Alza Corporation Lipid particles having asymmetric lipid coating and method of preparing same
WO2004098569A1 (en) * 2003-04-18 2004-11-18 Northeastern University Micelle delivery system loaded with a pharmaceutical agent
EP1643971A2 (en) * 2003-05-22 2006-04-12 Neopharm, Inc. Liposomal formulations comprising a combination of two or more active agents
GB0312309D0 (en) * 2003-05-29 2003-07-02 Gaslini Children S Hospital G Targeted liposome
WO2004110493A2 (en) * 2003-05-30 2004-12-23 Alza Corporation Method of pulmonary administration of an agent
US20040247624A1 (en) * 2003-06-05 2004-12-09 Unger Evan Charles Methods of making pharmaceutical formulations for the delivery of drugs having low aqueous solubility
TWI262192B (en) * 2003-07-01 2006-09-21 Univ Nat Taiwan Labeling peptide for nasopharyngeal carcinoma (NPC) cells
US20050266066A1 (en) * 2003-10-20 2005-12-01 Nof Corporation Phospholipid membrane preparation
EP1547580A1 (en) * 2003-12-23 2005-06-29 MediGene Oncology GmbH Loading of a camptothecin drug into colloidal nanoparticles
TWI317286B (en) * 2003-12-31 2009-11-21 Targeting delivery system
JP2007520481A (en) * 2004-01-15 2007-07-26 アルザ・コーポレーシヨン Liposome composition for delivering therapeutic agents
NZ549040A (en) 2004-02-17 2009-07-31 Schering Corp Use for interleukin-33 (IL33) and the IL-33 receptor complex
JP4903061B2 (en) 2004-02-17 2012-03-21 シェーリング コーポレイション Methods of modulating IL-23 activity; related reagents
CA3006109A1 (en) 2004-05-03 2005-11-17 Ipsen Biopharm Ltd. Drug delivery liposomes containing anionic polyols or anionic sugars
US8658203B2 (en) 2004-05-03 2014-02-25 Merrimack Pharmaceuticals, Inc. Liposomes useful for drug delivery to the brain
WO2005108382A1 (en) * 2004-05-04 2005-11-17 Merck & Co., Inc. 1,2,4-oxadiazole derivatives as dipeptidyl peptidase-iv inhibitors for the treatment or prevention of diabetes
US7449196B2 (en) * 2004-07-09 2008-11-11 Robert Sabin Anti tumor compositions and methods of use
EP3153159A1 (en) 2004-07-09 2017-04-12 Robert Sabin Compositions comprising a copper compound for treatment of mammalian diseases
JP4433918B2 (en) * 2004-07-15 2010-03-17 コニカミノルタエムジー株式会社 Image forming method
WO2006012527A1 (en) 2004-07-23 2006-02-02 Endocyte, Inc. Bivalent linkers and conjugates thereof
US8119153B2 (en) * 2004-08-26 2012-02-21 Boston Scientific Scimed, Inc. Stents with drug eluting coatings
JP2008515929A (en) * 2004-10-08 2008-05-15 アルザ コーポレイション A method for inserting a lipid assembly into which a lipid linking part is preformed using microwaves
WO2006044660A2 (en) * 2004-10-14 2006-04-27 Vanderbilt University Functionalized solid lipid nanoparticles and methods of making and using same
JP4990786B2 (en) * 2004-11-05 2012-08-01 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 Compositions and methods for stabilizing drug liposome formulations
TW200618820A (en) * 2004-11-05 2006-06-16 Alza Corp Liposome formulations of boronic acid compounds
KR100651728B1 (en) * 2004-11-10 2006-12-06 한국전자통신연구원 Compounds having anchoring group and electronic device including the same and methods for producing the same
US7820168B2 (en) 2004-12-20 2010-10-26 Schering Corporation Treatment of diabetes using antibodies to IL-23, IL-23 receptor and IL-17
PT2727583T (en) 2004-12-22 2021-12-27 Nitto Denko Corp Drug carrier and drug carrier kit for inhibiting fibrosis
US9393315B2 (en) * 2011-06-08 2016-07-19 Nitto Denko Corporation Compounds for targeting drug delivery and enhancing siRNA activity
US20120269886A1 (en) 2004-12-22 2012-10-25 Nitto Denko Corporation Therapeutic agent for pulmonary fibrosis
DK2360258T3 (en) * 2005-02-18 2015-01-12 Angiochem Inc Aprotininpolypeptider to transport a compound of blood-brain-barrier
US20090016959A1 (en) * 2005-02-18 2009-01-15 Richard Beliveau Delivery of antibodies to the central nervous system
US10058621B2 (en) 2015-06-25 2018-08-28 Immunomedics, Inc. Combination therapy with anti-HLA-DR antibodies and kinase inhibitors in hematopoietic cancers
US9707302B2 (en) 2013-07-23 2017-07-18 Immunomedics, Inc. Combining anti-HLA-DR or anti-Trop-2 antibodies with microtubule inhibitors, PARP inhibitors, bruton kinase inhibitors or phosphoinositide 3-kinase inhibitors significantly improves therapeutic outcome in cancer
JP5289935B2 (en) * 2005-03-16 2013-09-11 エンドサイト,インコーポレイテッド Synthesis and purification of pteroic acid and its conjugates
BRPI0610026A2 (en) * 2005-04-22 2010-05-18 Alza Corp immunoliposome composition for targeting a her2 cell receptor
EP1893038A1 (en) * 2005-05-17 2008-03-05 Cargill, Incorporated Granular lecithins, granular lysolecithins, process for their production and compositions containing them
WO2007005754A2 (en) * 2005-07-01 2007-01-11 Alza Corporation Liposomal delivery vehicle for hydrophobic drugs
ES2424242T3 (en) 2005-07-15 2013-09-30 Angiochem Inc. Use of aprotinin polypeptides as carriers in pharmaceutical conjugates
WO2007018759A2 (en) * 2005-07-25 2007-02-15 Centocor, Inc. Ligand-binding reagents for quenching and improved purification of lipidated proteins
US20070055199A1 (en) 2005-08-10 2007-03-08 Gilbert Scott J Drug delivery device for buccal and aural applications and other areas of the body difficult to access
US20070059318A1 (en) * 2005-08-15 2007-03-15 Balu-Iyer Sathy V Lipid nano particulates containing antigens as cancer vaccines
EP2382995A3 (en) * 2005-08-19 2013-09-25 Endocyte, Inc. Ligand conjugates of Vinca alkaloids, analogs and derivatives
JP5475992B2 (en) * 2005-08-19 2014-04-16 エンドサイト,インコーポレイテッド Multidrug ligand conjugate
CA2621086A1 (en) 2005-09-01 2007-03-08 Schering Corporation Use of il-23 and il-17 antagonists to treat autoimmune ocular inflammatory disease
US20110020434A1 (en) * 2005-09-02 2011-01-27 O'halloran Thomas V Nanoparticle arsenic-platinum compositions
JP2009507919A (en) * 2005-09-12 2009-02-26 アルザ・コーポレーシヨン Liposomes for treating multiple myeloma
JP5178520B2 (en) 2005-09-22 2013-04-10 メディバス エルエルシー Solid polymer delivery compositions and methods of use thereof
EP1926780B1 (en) 2005-09-22 2013-08-14 Medivas, LLC Bis-( -amino)-diol-diester-containing poly(ester amide) and poly(ester urethane) compositions and methods of use
US7846445B2 (en) * 2005-09-27 2010-12-07 Amunix Operating, Inc. Methods for production of unstructured recombinant polymers and uses thereof
JP2009509535A (en) * 2005-09-27 2009-03-12 アムニクス, インコーポレイテッド Proteinaceous drugs and their use
US7855279B2 (en) * 2005-09-27 2010-12-21 Amunix Operating, Inc. Unstructured recombinant polymers and uses thereof
US20090099031A1 (en) * 2005-09-27 2009-04-16 Stemmer Willem P Genetic package and uses thereof
WO2007038862A1 (en) * 2005-10-03 2007-04-12 Seneca College Of Applied Arts & Technology Nucleic acid immunological composition for human metapneumovirus
DK2628794T3 (en) 2005-10-18 2016-08-15 Prec Biosciences RATIONALE CONSTRUCTED MECHANUCLEAS WITH CHANGED SEQUENCE SPECIFICITY AND DNA BINDING EFFICIENCY
CA2896083A1 (en) 2005-12-08 2007-06-14 Insmed Incorporated Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US9572886B2 (en) 2005-12-22 2017-02-21 Nitto Denko Corporation Agent for treating myelofibrosis
CA2841386A1 (en) 2005-12-30 2007-07-12 Zensun (Shanghai) Science & Technology Limited Extended release of neuregulin for improved cardiac function
US20070154403A1 (en) * 2006-01-05 2007-07-05 Thomas Skold Oral, Pulmonary and Transmucosal Delivery Composition
WO2007092944A2 (en) * 2006-02-08 2007-08-16 Introgen Therapeutics, Inc. Compositions and methods involving gene therapy and proteasome modulation
WO2007095175A2 (en) * 2006-02-15 2007-08-23 Massachusetts Institute Of Technology Thermo-responsive materials
AU2007243370A1 (en) * 2006-04-24 2007-11-08 The Cbr Institute For Biomedical Research, Inc. Method of producing immunoliposomes and compositions thereof
JP5196498B2 (en) * 2006-05-09 2013-05-15 メディバス エルエルシー Biodegradable water-soluble polymer
US20070264322A1 (en) * 2006-05-10 2007-11-15 Huang Ken S Method for making liposomes conjugated with temperature-sensitive ligands
JP5072275B2 (en) * 2006-07-03 2012-11-14 テルモ株式会社 Method for separating closed vesicles, method for producing preparation and evaluation method
US20090202620A1 (en) * 2006-09-05 2009-08-13 Medivas, Llc Polymer-stabilized liposomal compositions and methods of use
US20080213349A1 (en) * 2006-09-11 2008-09-04 Deepak Ramesh Thakker Liposome Complexes Containing Pharmaceutical Agents and Methods
US20080081051A1 (en) * 2006-09-28 2008-04-03 Robert Sabin Method of manufacturing anti-tumor and anti-viral compositions
US8268347B1 (en) 2006-10-24 2012-09-18 Aradigm Corporation Dual action, inhaled formulations providing both an immediate and sustained release profile
US8119156B2 (en) * 2006-10-24 2012-02-21 Aradigm Corporation Dual action, inhaled formulations providing both an immediate and sustained release profile
US8071127B2 (en) * 2006-10-24 2011-12-06 Aradigm Corporation Dual action, inhaled formulations providing both an immediate and sustained release profile
US20080118500A1 (en) * 2006-11-16 2008-05-22 Taiwan Liposome Company Sustained releasing composition via local injection for treating eye diseases
US8278415B2 (en) 2006-12-21 2012-10-02 Centocor, Inc. Dimeric high affinity EGFR constructs and uses thereof
US8834920B2 (en) 2006-12-21 2014-09-16 Alza Corporation Liposome composition for targeting egfr receptor
WO2008079973A2 (en) * 2006-12-21 2008-07-03 Centocor, Inc. Egfr binding peptides and uses thereof
CA2676986A1 (en) * 2007-02-01 2008-08-07 Sigma-Tau Industrie Farmaceutiche Riunite S.P.A. Pharmaceutical composition comprising a campothecin derivative
WO2008101231A2 (en) 2007-02-16 2008-08-21 Endocyte, Inc. Methods and compositions for treating and diagnosing kidney disease
CA2679400A1 (en) 2007-02-28 2008-09-04 Schering Corporation Combination therapy for treatment of immune disorders
AU2008224988A1 (en) * 2007-03-14 2008-09-18 Endocyte, Inc. Binding ligand linked drug delivery conjugates of tubulysins
TWI407971B (en) 2007-03-30 2013-09-11 Nitto Denko Corp Cancer cells and tumor-related fibroblasts
US9119783B2 (en) 2007-05-07 2015-09-01 Insmed Incorporated Method of treating pulmonary disorders with liposomal amikacin formulations
US20090196913A1 (en) * 2007-05-11 2009-08-06 Ken Shi Kun Huang Anti-Alpha-V Immunoliposome Composition, Methods, and Uses
US9365634B2 (en) * 2007-05-29 2016-06-14 Angiochem Inc. Aprotinin-like polypeptides for delivering agents conjugated thereto to tissues
CN104383553A (en) * 2007-06-25 2015-03-04 恩多塞特公司 Conjugates containing hydrophilic spacer linkers
US9877965B2 (en) 2007-06-25 2018-01-30 Endocyte, Inc. Vitamin receptor drug delivery conjugates for treating inflammation
US20100210575A1 (en) * 2007-06-29 2010-08-19 Wisconsin Alumni Research Foundation Structuring effect of cholesterol in peg-phospholipid micelles, drug delivery of amphotericin b, and combination antifungals
US8889622B2 (en) * 2007-07-25 2014-11-18 Washington University Methods of inhibiting seizure in a subject
CN101952312A (en) 2007-07-31 2011-01-19 米迪缪尼有限公司 Multispecific epitope binding proteins and uses thereof
KR100979462B1 (en) * 2007-08-03 2010-09-02 한국화학연구원 Anthracycline anticancer drug-encapsulated liposomes and preparation method thereof
JP2010536341A (en) * 2007-08-15 2010-12-02 アムニクス, インコーポレイテッド Compositions and methods for altering properties of biologically active polypeptides
CA2713185A1 (en) * 2007-08-23 2009-02-26 Medivas, Llc Cationic alpha-amino acid-containing biodegradable polymer gene transfer compositions
WO2009036368A2 (en) 2007-09-14 2009-03-19 Nitto Denko Corporation Drug carriers
WO2009055562A1 (en) 2007-10-25 2009-04-30 Endocyte, Inc. Tubulysins and processes for preparing
ES2732735T3 (en) 2007-10-31 2019-11-25 Prec Biosciences Inc Single-chain meganucleases designed rationally with non-palindromic recognition sequences
WO2009074569A1 (en) * 2007-12-11 2009-06-18 Bracco International Bv Targeting and therapeutic compounds with a polyproline-comprising spacer and gas-filled microvesicles comprising said compounds
CN101485629B (en) * 2008-01-16 2013-01-23 沈阳药科大学 Drug delivery system and preparation method thereof
AU2009205995B2 (en) 2008-01-18 2014-04-03 Medimmune, Llc Cysteine engineered antibodies for site-specific conjugation
US8710013B2 (en) * 2008-04-18 2014-04-29 Angiochem Inc. Pharmaceutical compositions of paclitaxel, paclitaxel analogs or paclitaxel conjugates and related methods of preparation and use
WO2009158678A1 (en) 2008-06-27 2009-12-30 Children's Hospital & Research Center At Oakland Lipophilic nucleic acid delivery vehicle and methods of use therefor
EP2310440A4 (en) * 2008-07-10 2013-06-05 Serina Therapeutics Inc Polyoxazolines with inert terminating groups, polyoxazolines prepared from protected initiating groups and related compounds
CA2740317A1 (en) 2008-10-15 2010-04-22 Angiochem Inc. Etoposide and doxorubicin conjugates for drug delivery
AU2009304560A1 (en) 2008-10-15 2010-04-22 Angiochem Inc. Conjugates of GLP-1 agonists and uses thereof
BRPI0921357A2 (en) 2008-11-06 2016-06-21 Phaserx Inc multiblock copolymers
US20100151573A1 (en) * 2008-11-17 2010-06-17 King Michael R Compositions and methods for delivery of molecules to selectin-ligand-expressing and selectin-expressing cells
EP2370451B1 (en) 2008-12-02 2016-11-16 Wave Life Sciences Japan, Inc. Method for the synthesis of phosphorus atom modified nucleic acids
AU2009322043A1 (en) * 2008-12-05 2011-07-07 Angiochem Inc. Conjugates of neurotensin or neurotensin analogs and uses thereof
CA2747220A1 (en) 2008-12-17 2010-06-24 Richard Beliveau Membrane type-1 matrix metalloprotein inhibitors and uses thereof
US20100233270A1 (en) 2009-01-08 2010-09-16 Northwestern University Delivery of Oligonucleotide-Functionalized Nanoparticles
US8957021B2 (en) 2009-02-03 2015-02-17 Amunix Operating Inc. Glucose-regulating polypeptides and methods of making and using same
NZ593833A (en) 2009-02-03 2013-10-25 Amunix Operating Inc Extended recombinant polypeptides and compositions comprising same
JP5392707B2 (en) * 2009-03-31 2014-01-22 株式会社Nttドコモ Membrane vesicle division system
MX2011011023A (en) 2009-04-20 2012-01-20 Angiochem Inc Treatment of ovarian cancer using an anticancer agent conjugated to an angiopep-2 analog.
CN106916229A (en) 2009-06-08 2017-07-04 阿穆尼克斯运营公司 Growth hormone polypeptides and its preparation and application
RU2012101999A (en) 2009-06-22 2013-07-27 МЕДИММЬЮН, ЭлЭлСи Fc RECOMBINED SITES FOR SITE-SPECIFIC CONJUGATION
IN2012DN00248A (en) 2009-07-02 2015-05-01 Angiochem Inc
SG177564A1 (en) 2009-07-06 2012-02-28 Ontorii Inc Novel nucleic acid prodrugs and methods of use thereof
GB0913442D0 (en) * 2009-07-31 2009-09-16 Univ Ramot Cell-targeting nanoparticles comprising polynucleotide agents and uses thereof
EP3222287A1 (en) 2009-08-24 2017-09-27 Amunix Operating Inc. Coagulation factor ix compositions and methods of making and using same
NZ599543A (en) 2009-10-30 2014-02-28 Ntf Therapeutics Inc Improved neurturin molecules
US9415113B2 (en) 2009-11-18 2016-08-16 University Of Washington Targeting monomers and polymers having targeting blocks
WO2011084610A1 (en) * 2009-12-16 2011-07-14 Children Medical Center Corporation Liposomes for preventing the spread of hiv
RU2012151823A (en) 2010-05-04 2014-06-10 Мерримак Фармасьютикалз, Инк. ANTIBODIES AGAINST EPIDERMAL GROWTH FACTOR (EGFR) RECEPTOR AND THEIR APPLICATION
PE20130207A1 (en) 2010-05-06 2013-02-28 Novartis Ag ANTIBODIES ANTAGONISTS TO LRP6 (LOW DENSITY LIPOPROTEIN-RELATED PROTEIN 6) AND COMPOSITIONS
SG185415A1 (en) 2010-05-06 2012-12-28 Novartis Ag Compositions and methods of use for therapeutic low density lipoprotein - related protein 6 (lrp6) multivalent antibodies
KR101198715B1 (en) * 2010-05-14 2012-11-13 한국생명공학연구원 Asymmetric liposomes with higher encapsulation efficiency of nucleic acids and hydrophilic anion chemicals
US20130071321A1 (en) * 2010-05-28 2013-03-21 Purdue Research Foundation Delivery of agents to inflamed tissues using folate-targeted liposomes
CN103080125A (en) 2010-07-02 2013-05-01 安吉奥开米公司 Short and D-amino acid-containing polypeptides for therapeutic conjugates and uses thereof
MY162825A (en) 2010-08-20 2017-07-31 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3)
JP5868324B2 (en) 2010-09-24 2016-02-24 株式会社Wave Life Sciences Japan Asymmetric auxiliary group
US20130245233A1 (en) 2010-11-24 2013-09-19 Ming Lei Multispecific Molecules
KR20140026396A (en) * 2011-03-08 2014-03-05 어섹스 팔마큐티칼스 인코포레이티드 Targeted nanocarrier systems for delivery of actives across biological membranes
US8691231B2 (en) 2011-06-03 2014-04-08 Merrimack Pharmaceuticals, Inc. Methods of treatment of tumors expressing predominantly high affinity EGFR ligands or tumors expressing predominantly low affinity EGFR ligands with monoclonal and oligoclonal anti-EGFR antibodies
US10196637B2 (en) 2011-06-08 2019-02-05 Nitto Denko Corporation Retinoid-lipid drug carrier
TWI658830B (en) 2011-06-08 2019-05-11 日東電工股份有限公司 Retinoid-liposomes for enhancing modulation of hsp47 expression
US9873765B2 (en) 2011-06-23 2018-01-23 Dsm Ip Assets, B.V. Biodegradable polyesteramide copolymers for drug delivery
JP6045575B2 (en) 2011-06-23 2016-12-14 ディーエスエム アイピー アセッツ ビー.ブイ. Novel biodegradable polyesteramide copolymers for drug delivery
EP2731970B1 (en) 2011-07-15 2018-11-28 MorphoSys AG Antibodies that are cross-reactive for macrophage migration inhibitory factor (mif) and d-dopachrome tautomerase (d-dt)
RU2014105311A (en) 2011-07-19 2015-08-27 Уэйв Лайф Сайенсес Пте. Лтд. METHODS FOR SYNTHESIS OF FUNCTIONALIZED NUCLEIC ACIDS
US20130115270A1 (en) * 2011-11-09 2013-05-09 Henry John Smith Anti-interleukin-1 (IL-1) antibody used as a targeting agent to treat arthritis and other diseases
BR112014013568A8 (en) 2011-12-05 2017-06-13 Novartis Ag epidermal growth factor 3 (her3) receptor antibodies directed to her3 domain ii
AU2012349735B2 (en) 2011-12-05 2016-05-19 Novartis Ag Antibodies for epidermal growth factor receptor 3 (HER3)
WO2013093891A1 (en) 2011-12-22 2013-06-27 Nuvo Research Gmbh Liposomal chlorite or chlorate compositions
EP3539982A3 (en) 2011-12-23 2020-01-15 Pfizer Inc Engineered antibody constant regions for site-specific conjugation and methods and uses therefor
KR101347833B1 (en) 2011-12-29 2014-01-16 광주과학기술원 CD7-BPB Capable of Binding Specifically to CD7
DK2822577T3 (en) 2012-02-15 2019-04-01 Bioverativ Therapeutics Inc RECOMBINANT FACTOR VIII PROTEINS
ES2771208T3 (en) 2012-02-15 2020-07-06 Bioverativ Therapeutics Inc Factor VIII compositions and methods of preparation and use thereof
WO2013126797A1 (en) 2012-02-24 2013-08-29 Purdue Research Foundation Cholecystokinin b receptor targeting for imaging and therapy
CN104168887B (en) 2012-03-16 2018-09-04 默克专利股份有限公司 amino acid lipids
US20140080175A1 (en) 2012-03-29 2014-03-20 Endocyte, Inc. Processes for preparing tubulysin derivatives and conjugates thereof
US9925143B2 (en) 2012-04-18 2018-03-27 Ramot At Tel-Aviv University Ltd. Lipidated glycosaminoglycan particles for the delivery of nucleic acids
MX353567B (en) * 2012-05-23 2018-01-18 Univ Ohio State Lipid nanoparticle compositions for antisense oligonucleotides delivery.
KR20150027072A (en) 2012-06-04 2015-03-11 아이알엠 엘엘씨 Site-specific labeling methods and molecules produced thereby
NZ630849A (en) 2012-06-08 2016-04-29 Alkermes Inc Ligands modified by circular permutation as agonists and antagonists
SG11201500239VA (en) 2012-07-13 2015-03-30 Wave Life Sciences Japan Asymmetric auxiliary group
US9982257B2 (en) 2012-07-13 2018-05-29 Wave Life Sciences Ltd. Chiral control
SG11201500243WA (en) 2012-07-13 2015-04-29 Shin Nippon Biomedical Lab Ltd Chiral nucleic acid adjuvant
KR20150070318A (en) 2012-10-16 2015-06-24 엔도사이트, 인코포레이티드 Drug delivery conjugates containing unnatural amino acids and methods for using
RU2015115956A (en) 2012-11-09 2017-01-10 Пфайзер Инк. ANTIBODIES SPECIFIC TO THE THROMBOCYTE B GROWTH FACTOR, AND THEIR COMPOSITION AND APPLICATION
KR102186116B1 (en) 2012-11-20 2020-12-03 스펙트럼 파마슈티컬즈 인크 Improved method for the preparation of liposome encapsulated vincristine for therapeutic use
RU2675859C2 (en) 2012-11-29 2018-12-25 Инсмед Инкорпорейтед Stabilised vancomycin formulations
KR20150095809A (en) 2012-12-12 2015-08-21 템플 유니버시티-오브 더 커먼웰쓰 시스템 오브 하이어 에듀케이션 Compositions and methods for treatment of cancer
US10206918B2 (en) 2012-12-13 2019-02-19 Immunomedics, Inc. Efficacy of anti-HLA-DR antiboddy drug conjugate IMMU-140 (hL243-CL2A-SN-38) in HLA-DR positive cancers
US10413539B2 (en) 2012-12-13 2019-09-17 Immunomedics, Inc. Therapy for metastatic urothelial cancer with the antibody-drug conjugate, sacituzumab govitecan (IMMU-132)
WO2015012904A2 (en) 2012-12-13 2015-01-29 Immunomedics, Inc. Antibody-sn-38 immunoconjugates with a cl2a linker
US9492566B2 (en) 2012-12-13 2016-11-15 Immunomedics, Inc. Antibody-drug conjugates and uses thereof
US9931417B2 (en) 2012-12-13 2018-04-03 Immunomedics, Inc. Antibody-SN-38 immunoconjugates with a CL2A linker
US10137196B2 (en) 2012-12-13 2018-11-27 Immunomedics, Inc. Dosages of immunoconjugates of antibodies and SN-38 for improved efficacy and decreased toxicity
HUE057977T2 (en) 2012-12-13 2022-06-28 Immunomedics Inc Dosages of immunoconjugates of antibodies and sn-38 for improved efficacy and decreased toxicity
US10744129B2 (en) 2012-12-13 2020-08-18 Immunomedics, Inc. Therapy of small-cell lung cancer (SCLC) with a topoisomerase-I inhibiting antibody-drug conjugate (ADC) targeting Trop-2
US9107947B2 (en) * 2013-01-31 2015-08-18 The Penn State Research Foundation Anti-cancer compositions and methods
WO2014124258A2 (en) 2013-02-08 2014-08-14 Irm Llc Specific sites for modifying antibodies to make immunoconjugates
KR102447350B1 (en) 2013-02-08 2022-09-23 노파르티스 아게 Specific sites for modifying antibodies to make immunoconjugates
US9498532B2 (en) 2013-03-13 2016-11-22 Novartis Ag Antibody drug conjugates
JP2016514130A (en) 2013-03-14 2016-05-19 ノバルティス アーゲー Antibody against Notch3
NZ710929A (en) 2013-03-15 2018-02-23 Novartis Ag Antibody drug conjugates
AU2014271207B2 (en) * 2013-05-23 2019-11-07 The University Of Newcastle Targeted delivery of drugs to the myometrium
US11253606B2 (en) 2013-07-23 2022-02-22 Immunomedics, Inc. Combining anti-HLA-DR or anti-Trop-2 antibodies with microtubule inhibitors, PARP inhibitors, Bruton kinase inhibitors or phosphoinositide 3-kinase inhibitors significantly improves therapeutic outcome in cancer
US10894963B2 (en) 2013-07-25 2021-01-19 Exicure, Inc. Spherical nucleic acid-based constructs as immunostimulatory agents for prophylactic and therapeutic use
US10568898B2 (en) 2013-08-13 2020-02-25 Northwestern University Lipophilic nanoparticles for drug delivery
US10548953B2 (en) 2013-08-14 2020-02-04 Bioverativ Therapeutics Inc. Factor VIII-XTEN fusions and uses thereof
CA3191031A1 (en) 2013-09-27 2015-04-02 Massachusetts Institute Of Technology Carrier-free biologically-active protein nanostructures
EP3060198A4 (en) 2013-10-22 2017-06-28 Aradigm Corporation Inhaled surfactant-modified liposomal formulations providing both an immediate and sustained release profile
US10287572B2 (en) 2013-11-01 2019-05-14 Regents Of The University Of Minnesota Protein scaffolds and methods of use
US9683998B2 (en) 2013-11-13 2017-06-20 Pfizer Inc. Tumor necrosis factor-like ligand 1A specific antibodies and compositions and uses thereof
WO2015126502A2 (en) 2013-12-03 2015-08-27 Northwestern University Liposomal particles, methods of making same and uses thereof
US10149905B2 (en) 2014-01-15 2018-12-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having antitumor effect and antitumor agent
JPWO2015108047A1 (en) 2014-01-15 2017-03-23 株式会社新日本科学 Chiral nucleic acid adjuvant having immunity induction activity and immunity induction activator
JPWO2015108046A1 (en) 2014-01-15 2017-03-23 株式会社新日本科学 Chiral nucleic acid adjuvant and antiallergic agent having antiallergic action
DK3094728T3 (en) 2014-01-16 2022-05-16 Wave Life Sciences Ltd KIRALT DESIGN
WO2015109212A1 (en) 2014-01-17 2015-07-23 Pfizer Inc. Anti-il-2 antibodies and compositions and uses thereof
WO2015110930A1 (en) 2014-01-24 2015-07-30 Pfizer Inc. Modified interleukin 21 receptor proteins
US20170021033A1 (en) 2014-03-12 2017-01-26 Novartis Ag Specific sites for modifying antibodies to make immunoconjugates
WO2015148648A1 (en) 2014-03-25 2015-10-01 The Government Of The United States Of America As Represented By The Secretary Of The Army Non-toxic adjuvant formulation comprising a monophosphoryl lipid a (mpla)-containing liposome composition and a saponin
HUE052968T2 (en) 2014-04-30 2021-05-28 Fujifilm Corp Liposome composition and production method therefor
EP3138558B1 (en) * 2014-04-30 2023-06-07 FUJIFILM Corporation Liposome composition and production method therefor
ES2926985T3 (en) 2014-05-15 2022-10-31 Insmed Inc Methods for treating nontuberculous mycobacterial lung infections
JP6581604B2 (en) 2014-06-04 2019-09-25 イグジキュア, インコーポレーテッドExicure, Inc. Multivalent delivery of immunomodulators with liposomal globular nucleic acids for prophylactic or therapeutic applications
JP2017520549A (en) 2014-06-26 2017-07-27 ラモット アット テル アビブ ユニバーシティ, リミテッド Liposome formulation for delivery of nucleic acids
CN107257691B (en) 2014-07-16 2021-09-21 达娜-法勃肿瘤研究所公司 HER3 inhibition in low-grade serous ovarian cancer
US20160030343A1 (en) * 2014-08-01 2016-02-04 Texas Tech University System Preparation and characterization of bone-targeted vancomycin-loaded liposomes for osteomyelitis treatment
US10786578B2 (en) 2014-08-05 2020-09-29 Novartis Ag CKIT antibody drug conjugates
WO2016022547A1 (en) 2014-08-06 2016-02-11 Indiana University Research And Technology Corporation Tuneable delivery of nanoparticle bound active plasmin for the treatment of thrombosis
KR20170040249A (en) 2014-08-12 2017-04-12 노파르티스 아게 Anti-cdh6 antibody drug conjugates
US10245393B2 (en) 2014-08-13 2019-04-02 Elwha Llc Methods, systems, and devices related to a supplemental inhaler
US10987048B2 (en) 2014-08-13 2021-04-27 Elwha Llc Systems, methods, and devices to incentivize inhaler use
US10765817B2 (en) 2014-08-13 2020-09-08 Elwha, Llc Methods, systems, and devices related to delivery of alcohol with an inhaler
KR20170063949A (en) 2014-10-06 2017-06-08 엑시큐어, 인크. Anti-tnf compounds
SG11201703464UA (en) 2014-11-14 2017-05-30 Novartis Ag Antibody drug conjugates
CN107106493A (en) 2014-11-21 2017-08-29 西北大学 The sequence-specific cellular uptake of spherical nucleic acid nano particle conjugate
WO2016085986A1 (en) 2014-11-24 2016-06-02 Northwestern University High density lipoprptein nanoparticles for inflammation
ES2784900T3 (en) 2014-12-12 2020-10-01 Massachusetts Gen Hospital Treatment of brain metastases from breast cancer
JP6720447B2 (en) 2014-12-18 2020-07-08 ディーエスエム アイピー アセッツ ビー.ブイ.Dsm Ip Assets B.V. Drug delivery system for delivery of acid sensitive drugs
CA2974196C (en) 2015-01-20 2023-12-12 TetraDerm Group LLC Versatile topical drug delivery vehicle and multifactorial tissue moisturizer that provides mucosal and skin barrier restoration
TW201632202A (en) 2015-01-30 2016-09-16 諾華公司 Treatment of breast cancer
CA2981543A1 (en) 2015-04-22 2016-10-27 Immunomedics, Inc. Isolation, detection, diagnosis and/or characterization of circulating trop-2-positive cancer cells
US10980892B2 (en) 2015-06-15 2021-04-20 Angiochem Inc. Methods for the treatment of leptomeningeal carcinomatosis
US10975112B2 (en) * 2015-06-16 2021-04-13 Hangzhou Dac Biotech Co., Ltd. Linkers for conjugation of cell-binding molecules
EP3310813A1 (en) 2015-06-17 2018-04-25 Novartis AG Antibody drug conjugates
US10195175B2 (en) 2015-06-25 2019-02-05 Immunomedics, Inc. Synergistic effect of anti-Trop-2 antibody-drug conjugate in combination therapy for triple-negative breast cancer when used with microtubule inhibitors or PARP inhibitors
TWI678213B (en) 2015-07-22 2019-12-01 美商史倍壯製藥公司 A ready-to-use formulation for vincristine sulfate liposome injection
CA3025896A1 (en) 2015-07-23 2017-01-26 The Regents Of The University Of California Antibodies to coagulation factor xia and uses thereof
KR20180029262A (en) 2015-08-03 2018-03-20 바이오버라티브 테라퓨틱스 인크. Factor IX fusion proteins and methods for their manufacture and use
CA2995212A1 (en) 2015-08-12 2017-02-16 Massachusetts Institute Of Technology Cell surface coupling of nanoparticles
US9862760B2 (en) 2015-09-16 2018-01-09 Novartis Ag Polyomavirus neutralizing antibodies
SG10201912568PA (en) 2015-10-16 2020-02-27 Ipsen Biopharm Ltd Stabilizing camptothecin pharmaceutical compositions
TW201722989A (en) 2015-10-23 2017-07-01 輝瑞大藥廠 Anti-IL-2 antibodies and compositions and uses thereof
MA44334A (en) 2015-10-29 2018-09-05 Novartis Ag ANTIBODY CONJUGATES INCLUDING A TOLL-TYPE RECEPTOR AGONIST
KR20180077180A (en) 2015-11-06 2018-07-06 아쥬반스 테크놀로지스 인코포레이티드 Triterpene saponin analog
WO2017125897A1 (en) 2016-01-21 2017-07-27 Novartis Ag Multispecific molecules targeting cll-1
US20200297867A1 (en) * 2016-02-01 2020-09-24 Exicure, Inc. Surface functionalization of liposomes and liposomal spherical nucleic acids (snas)
US20170224837A1 (en) 2016-02-10 2017-08-10 Immunomedics, Inc. Combination of abcg2 inhibitors with sacituzumab govitecan (immu-132) overcomes resistance to sn-38 in trop-2 expressing cancers
RU2725292C2 (en) 2016-04-27 2020-06-30 Иммьюномедикс, Инк. Efficacy of conjugates of anti-trop-2 antibody with drug sn-38 for therapy of recurrent/refractory to inhibitors of tumor control point
WO2017193087A1 (en) 2016-05-06 2017-11-09 Exicure, Inc. Liposomal spherical nucleic acid (sna) constructs prsenting antisense oligonucleotides(aso) for specific knockdown of interleukin 17 receptor mrna
CN109715196A (en) 2016-06-13 2019-05-03 转矩医疗股份有限公司 For promoting the composition and method of immune cell function
JOP20190155A1 (en) 2016-12-21 2019-06-23 Novartis Ag Antibody drug conjugates for ablating hematopoietic stem cells
JOP20190187A1 (en) 2017-02-03 2019-08-01 Novartis Ag Anti-ccr7 antibody drug conjugates
CA3056797A1 (en) 2017-03-16 2018-09-20 Children's Medical Center Corporation Engineered liposomes as cancer-targeted therapeutics
JP2020512314A (en) 2017-03-27 2020-04-23 イミューノメディクス、インコーポレイテッドImmunomedics, Inc. Treatment of Trop-2-expressing triple-negative breast cancer with sacituzumab gobitecan and RAD51 inhibitor
EP3606964A4 (en) 2017-04-03 2020-12-09 Immunomedics, Inc. Subcutaneous administration of antibody-drug conjugates for cancer therapy
WO2018185618A1 (en) 2017-04-03 2018-10-11 Novartis Ag Anti-cdh6 antibody drug conjugates and anti-gitr antibody combinations and methods of treatment
CA3060442A1 (en) * 2017-04-19 2018-10-25 Apa- Advanced Technologies Ltd. Fusogenic liposomes, compositions, kits and use thereof for treating cancer
AR111651A1 (en) 2017-04-28 2019-08-07 Novartis Ag CONJUGATES OF ANTIBODIES THAT INCLUDE TOLL TYPE RECEIVER AGONISTS AND COMBINATION THERAPIES
KR20200018407A (en) * 2017-04-28 2020-02-19 텍사스 칠드런스 하스피탈 Targeting Nanoparticles
US11696954B2 (en) 2017-04-28 2023-07-11 Exicure Operating Company Synthesis of spherical nucleic acids using lipophilic moieties
EP3652186A4 (en) 2017-07-13 2021-03-31 Northwestern University General and direct method for preparing oligonucleotide-functionalized metal-organic framework nanoparticles
US11524033B2 (en) 2017-09-05 2022-12-13 Torque Therapeutics, Inc. Therapeutic protein compositions and methods of making and using the same
SG11202001826YA (en) 2017-10-16 2020-03-30 Adjuvance Technologies Inc Triterpene saponin analogues
EP3717510A2 (en) 2017-12-01 2020-10-07 Novartis AG Polyomavirus neutralizing antibodies
WO2019108639A1 (en) 2017-12-01 2019-06-06 Pfizer Inc. Anti-cxcr5 antibodies and compositions and uses thereof
SG11202009539XA (en) 2018-03-29 2020-10-29 Pfizer Lfa3 variants and compositions and uses thereof
WO2019191627A1 (en) 2018-03-30 2019-10-03 Insmed Incorporated Methods for continuous manufacture of liposomal drug products
EP3801766A1 (en) 2018-05-31 2021-04-14 Novartis AG Hepatitis b antibodies
US11492409B2 (en) 2018-06-01 2022-11-08 Novartis Ag Binding molecules against BCMA and uses thereof
TW202014209A (en) 2018-06-20 2020-04-16 瑞士商諾華公司 Antibody drug conjugates for ablating hematopoietic stem cells
US20210268070A1 (en) 2018-07-10 2021-09-02 Rush University Medical Center Use of Immunomodulators to Control Infection and Stimulate Healing in Normal and Diabetic Wounds
TW202024131A (en) 2018-09-07 2020-07-01 美商輝瑞大藥廠 Anti-αvβ8 antibodies and compositions and uses thereof
WO2020053742A2 (en) 2018-09-10 2020-03-19 Novartis Ag Anti-hla-hbv peptide antibodies
WO2020128863A1 (en) 2018-12-19 2020-06-25 Novartis Ag Anti-tnf-alpha antibodies
JOP20210159A1 (en) 2018-12-21 2023-01-30 Novartis Ag Antibodies to pmel17 and conjugates thereof
EP3936501A4 (en) 2019-03-08 2022-11-09 ABTIS Co., Ltd. Site-specific antibody conjugation and antibody-drug conjugate as specific example thereof
CN113853219A (en) 2019-05-20 2021-12-28 诺华股份有限公司 Antibody drug conjugates with linkers comprising hydrophilic groups
US20230181756A1 (en) 2020-04-30 2023-06-15 Novartis Ag Ccr7 antibody drug conjugates for treating cancer
CN116472288A (en) 2020-11-06 2023-07-21 诺华股份有限公司 Antibody Fc variants
AU2021373366A1 (en) 2020-11-06 2023-06-01 Novartis Ag Cd19 binding molecules and uses thereof
EP4240494A1 (en) 2020-11-06 2023-09-13 Novartis AG Anti-cd19 agent and b cell targeting agent combination therapy for treating b cell malignancies
CR20230283A (en) 2020-11-24 2023-07-27 Novartis Ag Anti-cd48 antibodies, antibody drug conjugates, and uses thereof
EP4323526A1 (en) 2021-04-16 2024-02-21 Novartis AG Antibody drug conjugates and methods for making thereof
WO2023175454A1 (en) 2022-03-14 2023-09-21 Pfizer Inc. Methods for producing an adjuvant

Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3539177A (en) * 1968-06-13 1970-11-10 Jacobs Machine Corp Delivery system for cloth
US4913902A (en) * 1987-11-10 1990-04-03 North Carolina State University Purification by affinity binding to liposomes
US5013556A (en) * 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5370933A (en) * 1992-01-31 1994-12-06 Ppg Industries, Inc. Soil release composition for use with polyester textiles
US5395619A (en) * 1993-03-03 1995-03-07 Liposome Technology, Inc. Lipid-polymer conjugates and liposomes
US5426112A (en) * 1984-04-09 1995-06-20 Scully, Scott, Murphy & Presser, P.C. Growth regulation and related applications of opioid antagonists
US5527528A (en) * 1989-10-20 1996-06-18 Sequus Pharmaceuticals, Inc. Solid-tumor treatment method
US5534241A (en) * 1993-07-23 1996-07-09 Torchilin; Vladimir P. Amphipathic polychelating compounds and methods of use
US5603872A (en) * 1991-02-14 1997-02-18 Baxter International Inc. Method of binding recognizing substances to liposomes
US5620689A (en) * 1989-10-20 1997-04-15 Sequus Pharmaceuuticals, Inc. Liposomes for treatment of B-cell and T-cell disorders
US5719032A (en) * 1992-01-31 1998-02-17 University Of British Columbia Melanoma and prostate cancer specific antibodies for immunodetection and immunotherapy
US5840687A (en) * 1995-02-02 1998-11-24 Schering Aktiengesellschaft Modified ligands for receptor tyrosine kinases
US5891468A (en) * 1996-10-11 1999-04-06 Sequus Pharmaceuticals, Inc. Fusogenic liposome compositions and method
US5932462A (en) * 1995-01-10 1999-08-03 Shearwater Polymers, Inc. Multiarmed, monofunctional, polymer for coupling to molecules and surfaces
US5985852A (en) * 1996-03-01 1999-11-16 The Regents Of The University Of California Inhibition of selectin binding
US6056973A (en) * 1996-10-11 2000-05-02 Sequus Pharmaceuticals, Inc. Therapeutic liposome composition and method of preparation
US6120798A (en) * 1997-06-23 2000-09-19 Alza Corporation Liposome-entrapped polynucleotide composition and method
US6129916A (en) * 1991-04-19 2000-10-10 Tanox, Inc. Method of Increasing activation on proliferation of T cells using antibody-microbead conjugates
US6224903B1 (en) * 1996-10-11 2001-05-01 Sequus Pharmaceuticals, Inc. Polymer-lipid conjugate for fusion of target membranes
US20050169880A1 (en) * 1999-11-08 2005-08-04 Water Journey Ltd. Antibacterial compositions and method of using same

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5800815A (en) * 1903-05-05 1998-09-01 Cytel Corporation Antibodies to P-selectin and their uses
ATE78158T1 (en) 1985-05-22 1992-08-15 Liposome Technology Inc METHOD AND SYSTEM FOR INHALATION OF LIPOSOMES.
US5136095A (en) 1987-05-19 1992-08-04 Syntex (U.S.A.) Inc. Reversible agglutination mediators
JPH0720857B2 (en) 1988-08-11 1995-03-08 テルモ株式会社 Liposome and its manufacturing method
IL91664A (en) * 1988-09-28 1993-05-13 Yissum Res Dev Co Ammonium transmembrane gradient system for efficient loading of liposomes with amphipathic drugs and their controlled release
US4906476A (en) * 1988-12-14 1990-03-06 Liposome Technology, Inc. Novel liposome composition for sustained release of steroidal drugs in lungs
JP3220180B2 (en) 1991-05-23 2001-10-22 三菱化学株式会社 Drug-containing protein-bound liposomes
JPH07173079A (en) 1992-12-22 1995-07-11 Nippon Oil & Fats Co Ltd Amphiphatic polyethylene glycol derivative and its use
WO1994021281A1 (en) 1993-03-23 1994-09-29 Liposome Technology, Inc. Polymer-polypeptide composition and method
JPH08510729A (en) * 1993-05-14 1996-11-12 サイテル コーポレイション Sialyl Le <ab> x analogues as inhibitors of cell adhesion
DE69407292T2 (en) * 1993-06-30 1998-06-25 Genentech Inc METHOD FOR PRODUCING LIPOSOME
US5885613A (en) * 1994-09-30 1999-03-23 The University Of British Columbia Bilayer stabilizing components and their use in forming programmable fusogenic liposomes
US5939401A (en) * 1994-12-09 1999-08-17 Genzyme Corporation Cationic amphiphile compositions for intracellular delivery of therapeutic molecules
US5686114A (en) * 1995-06-02 1997-11-11 University Of Iowa Research Foundation Uses of inorganic pyrophosphates
CA2250219C (en) * 1996-03-28 2008-10-07 Hayat Onyuksel Materials and methods for making improved liposome compositions
US6210707B1 (en) * 1996-11-12 2001-04-03 The Regents Of The University Of California Methods of forming protein-linked lipidic microparticles, and compositions thereof

Patent Citations (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3539177A (en) * 1968-06-13 1970-11-10 Jacobs Machine Corp Delivery system for cloth
US5426112A (en) * 1984-04-09 1995-06-20 Scully, Scott, Murphy & Presser, P.C. Growth regulation and related applications of opioid antagonists
US4913902A (en) * 1987-11-10 1990-04-03 North Carolina State University Purification by affinity binding to liposomes
US5013556A (en) * 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5620689A (en) * 1989-10-20 1997-04-15 Sequus Pharmaceuuticals, Inc. Liposomes for treatment of B-cell and T-cell disorders
US5527528A (en) * 1989-10-20 1996-06-18 Sequus Pharmaceuticals, Inc. Solid-tumor treatment method
US5603872A (en) * 1991-02-14 1997-02-18 Baxter International Inc. Method of binding recognizing substances to liposomes
US6129916A (en) * 1991-04-19 2000-10-10 Tanox, Inc. Method of Increasing activation on proliferation of T cells using antibody-microbead conjugates
US5719032A (en) * 1992-01-31 1998-02-17 University Of British Columbia Melanoma and prostate cancer specific antibodies for immunodetection and immunotherapy
US5370933A (en) * 1992-01-31 1994-12-06 Ppg Industries, Inc. Soil release composition for use with polyester textiles
US5395619A (en) * 1993-03-03 1995-03-07 Liposome Technology, Inc. Lipid-polymer conjugates and liposomes
US5631018A (en) * 1993-03-03 1997-05-20 Sequus Pharmaceuticals, Inc. Lipid-polymer conjugates and liposomes
US5534241A (en) * 1993-07-23 1996-07-09 Torchilin; Vladimir P. Amphipathic polychelating compounds and methods of use
US5932462A (en) * 1995-01-10 1999-08-03 Shearwater Polymers, Inc. Multiarmed, monofunctional, polymer for coupling to molecules and surfaces
US5840687A (en) * 1995-02-02 1998-11-24 Schering Aktiengesellschaft Modified ligands for receptor tyrosine kinases
US5985852A (en) * 1996-03-01 1999-11-16 The Regents Of The University Of California Inhibition of selectin binding
US6056973A (en) * 1996-10-11 2000-05-02 Sequus Pharmaceuticals, Inc. Therapeutic liposome composition and method of preparation
US5891468A (en) * 1996-10-11 1999-04-06 Sequus Pharmaceuticals, Inc. Fusogenic liposome compositions and method
US6224903B1 (en) * 1996-10-11 2001-05-01 Sequus Pharmaceuticals, Inc. Polymer-lipid conjugate for fusion of target membranes
US6316024B1 (en) * 1996-10-11 2001-11-13 Sequus Pharmaceuticals, Inc. Therapeutic liposome composition and method of preparation
US20040191307A1 (en) * 1996-10-11 2004-09-30 Alza Corporation Therapeutic liposome composition and method of preparation
US20040191250A1 (en) * 1996-10-11 2004-09-30 Alza Corporation Therapeutic liposome composition and method of preparation
US20050136064A1 (en) * 1996-10-11 2005-06-23 Alza Corporation Therapeutic liposome composition and method of preparation
US6936272B2 (en) * 1996-10-11 2005-08-30 Alza Corporation 10139483Therapeutic liposome composition and method of preparation
US7122202B2 (en) * 1996-10-11 2006-10-17 Alza Corporation Therapeutic liposome composition and method of preparation
US6120798A (en) * 1997-06-23 2000-09-19 Alza Corporation Liposome-entrapped polynucleotide composition and method
US20050169880A1 (en) * 1999-11-08 2005-08-04 Water Journey Ltd. Antibacterial compositions and method of using same

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040191250A1 (en) * 1996-10-11 2004-09-30 Alza Corporation Therapeutic liposome composition and method of preparation
US20040191307A1 (en) * 1996-10-11 2004-09-30 Alza Corporation Therapeutic liposome composition and method of preparation
US20050136064A1 (en) * 1996-10-11 2005-06-23 Alza Corporation Therapeutic liposome composition and method of preparation
US20050169980A1 (en) * 1996-10-11 2005-08-04 Alza Corporation Therapeutic liposome composition and method of preparation
US20060246126A1 (en) * 1996-10-11 2006-11-02 Alza Corporation Therapeutic liposome composition and method of preparation
US8758810B2 (en) 2005-03-10 2014-06-24 Mebiopharm Co., Ltd. Liposome compositions
US7829113B2 (en) 2005-03-10 2010-11-09 Mebiopharm Co., Ltd. Liposome compositions
US20080213369A1 (en) * 2006-09-07 2008-09-04 Canadian Blood Services Synthetic platelets
US20080063621A1 (en) * 2006-09-07 2008-03-13 Canadian Blood Services Surface cross-linked lipidic particles, methods of production and uses therefor
US9878044B2 (en) 2012-03-16 2018-01-30 Merck Patent Gmbh Targeting aminoacid lipids
US11510988B2 (en) 2012-03-16 2022-11-29 Merck Patent Gmbh Targeting aminoacid lipids
WO2014015027A1 (en) 2012-07-18 2014-01-23 Onyx Therapeutics, Inc. Liposomal compositions of epoxyketone-based proteasome inhibitors
US10022326B2 (en) 2012-07-18 2018-07-17 Onyx Therapeutics, Inc. Liposomal compositions of epoxyketone-based proteasome inhibitors
WO2018087720A1 (en) 2016-11-14 2018-05-17 Novartis Ag Compositions, methods, and therapeutic uses related to fusogenic protein minion

Also Published As

Publication number Publication date
US20050169980A1 (en) 2005-08-04
US20010038851A1 (en) 2001-11-08
US20020172711A1 (en) 2002-11-21
US20060246126A1 (en) 2006-11-02
US7122202B2 (en) 2006-10-17
US20050136064A1 (en) 2005-06-23
US6316024B1 (en) 2001-11-13
US6056973A (en) 2000-05-02
US20040191307A1 (en) 2004-09-30
US20040191250A1 (en) 2004-09-30
US6936272B2 (en) 2005-08-30

Similar Documents

Publication Publication Date Title
US6316024B1 (en) Therapeutic liposome composition and method of preparation
Mamot et al. Epidermal growth factor receptor–targeted immunoliposomes significantly enhance the efficacy of multiple anticancer drugs in vivo
EP0932391B1 (en) Fusogenic liposome composition and method
US20060269542A1 (en) Immunoliposome composition for targeting to a HER2 cell receptor
US6224903B1 (en) Polymer-lipid conjugate for fusion of target membranes
US7465716B2 (en) Targeted drug delivery with a hyaluronan ligand
US6043094A (en) Therapeutic liposome composition and method
US20070092558A1 (en) Methods of Preparing Targeted Immunoliposomes
US20100239652A1 (en) Immunoliposomes for treatment of cancer
CN108926719B (en) Long-circulating liposomes modified with c (RGD-ACP-K)
US20070292497A1 (en) Method for treating micrometastatic tumors
EP1214935A2 (en) Fusogenic liposome composition and method
WO2007133627A2 (en) Method for making liposomes conjugated with temperature-sensitive ligands
AU736055B2 (en) Fusogenic liposome composition and method
AU761204B2 (en) Fusogenic liposome composition and method
CA2505445A1 (en) Fusogenic liposome composition and method

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- AFTER EXAMINER'S ANSWER OR BOARD OF APPEALS DECISION