US20130071321A1 - Delivery of agents to inflamed tissues using folate-targeted liposomes - Google Patents

Delivery of agents to inflamed tissues using folate-targeted liposomes Download PDF

Info

Publication number
US20130071321A1
US20130071321A1 US13/700,358 US201113700358A US2013071321A1 US 20130071321 A1 US20130071321 A1 US 20130071321A1 US 201113700358 A US201113700358 A US 201113700358A US 2013071321 A1 US2013071321 A1 US 2013071321A1
Authority
US
United States
Prior art keywords
folate
agent
targeted
entrapped
liposomal composition
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
US13/700,358
Inventor
Philip Stewart Low
Scott Poh
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.)
Purdue Research Foundation
Original Assignee
Purdue Research Foundation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Purdue Research Foundation filed Critical Purdue Research Foundation
Priority to US13/700,358 priority Critical patent/US20130071321A1/en
Assigned to PURDUE RESEARCH FOUNDATION reassignment PURDUE RESEARCH FOUNDATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: POH, SCOTT YEW TAT, LOW, PHILIP S.
Publication of US20130071321A1 publication Critical patent/US20130071321A1/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
    • 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
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • A61K31/573Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
    • A61K47/48215
    • 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
    • A61K47/55Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
    • A61K47/551Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds one of the codrug's components being a vitamin, e.g. niacinamide, vitamin B3, cobalamin, vitamin B12, folate, vitamin A or retinoic 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
    • A61K47/69Medicinal 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
    • A61K49/00Preparations for testing in vivo
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/0019Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules
    • A61K49/0021Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules the fluorescent group being a small organic molecule
    • A61K49/0032Methine dyes, e.g. cyanine dyes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/0019Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules
    • A61K49/0021Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules the fluorescent group being a small organic molecule
    • A61K49/0041Xanthene dyes, used in vivo, e.g. administered to a mice, e.g. rhodamines, rose Bengal
    • A61K49/0043Fluorescein, used in vivo
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/005Fluorescence in vivo characterised by the carrier molecule carrying the fluorescent agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/005Fluorescence in vivo characterised by the carrier molecule carrying the fluorescent agent
    • A61K49/0054Macromolecular compounds, i.e. oligomers, polymers, dendrimers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/04X-ray contrast preparations
    • A61K49/0433X-ray contrast preparations containing an organic halogenated X-ray contrast-enhancing agent
    • A61K49/0438Organic X-ray contrast-enhancing agent comprising an iodinated group or an iodine atom, e.g. iopamidol
    • 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
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61BDIAGNOSIS; SURGERY; IDENTIFICATION
    • A61B5/00Measuring for diagnostic purposes; Identification of persons
    • A61B5/05Detecting, measuring or recording for diagnosis by means of electric currents or magnetic fields; Measuring using microwaves or radio waves 
    • A61B5/055Detecting, measuring or recording for diagnosis by means of electric currents or magnetic fields; Measuring using microwaves or radio waves  involving electronic [EMR] or nuclear [NMR] magnetic resonance, e.g. magnetic resonance imaging
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61BDIAGNOSIS; SURGERY; IDENTIFICATION
    • A61B6/00Apparatus for radiation diagnosis, e.g. combined with radiation therapy equipment
    • A61B6/48Diagnostic techniques
    • A61B6/481Diagnostic techniques involving the use of contrast agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61BDIAGNOSIS; SURGERY; IDENTIFICATION
    • A61B8/00Diagnosis using ultrasonic, sonic or infrasonic waves
    • A61B8/48Diagnostic techniques
    • A61B8/481Diagnostic techniques involving the use of contrast agent, e.g. microbubbles introduced into the bloodstream

Definitions

  • folate-receptor targeted agents comprising therapeutic agents useful for the treatment of inflammatory disease, including folate-receptor targeted liposomes (folate-targeted liposomes) containing entrapped therapeutic agents and folate-receptor targeted dendrimers conjugated to therapeutic agents (folate-targeted dendrimer conjugates), useful for the treatment of inflammatory disease, including auto-immune disease, as well as to folate-targeted liposomes containing entrapped imaging agents and dendrimer conjugates conjugated to imaging agents, for use in the diagnosis and monitoring of treatment in such disease.
  • therapeutic agents useful for the treatment of inflammatory disease including folate-receptor targeted liposomes (folate-targeted liposomes) containing entrapped therapeutic agents and folate-receptor targeted dendrimers conjugated to therapeutic agents (folate-targeted dendrimer conjugates)
  • folate-targeted liposomes containing entrapped therapeutic agents
  • Liposomes are closed, spherical vesicles comprising amphiphilic lipids such as phospholipids, sphingolipids, and/or other lipids, including sterols such as cholesterol and cholesterol ester salts, in proportions such that they arrange themselves into multiple concentric bilayers when hydrated in aqueous solutions.
  • amphiphilic lipids such as phospholipids, sphingolipids, and/or other lipids, including sterols such as cholesterol and cholesterol ester salts
  • sterols such as cholesterol and cholesterol ester salts
  • liposomes as carriers for therapeutic agents has been recognized and has lead to the development of long-circulating liposomes.
  • altering the composition of the lipid bilayer in such liposomes by, for example, the inclusion of a surface coat of flexible biocompatible hydrophilic chains the resulting liposomes have improved stability.
  • the surface coat serves to protect the liposome from uptake by organs of the mononuclear phagocyte system, especially the liver, lung and spleen, and from the plasma components which are involved in liposome uptake.
  • hydrophilic chain constituent of the surface coat is polyethylene glycol (PEG), in which the terminal hydroxy group may be capped with a methyl group to form a methyl ether group (denoted as mPEG or MPEG).
  • PEG polyethylene glycol
  • MPEG MPEG
  • PEG will include the hydroxy terminated form, the methyl ether terminated form, and any other similarly terminated form in the context of the surface coat of a liposome.
  • Inflamed tissue regions in the body have been imaged using folate-targeted imaging agents.
  • the folate-receptor targeted, radionuclide conjugate imaging agent EC20 (folate-Tc99m), has been used to image rheumatoid arthritis sites in vivo.
  • WO 94/07466 there is disclosed a PEG coated liposomal composition containing an entrapped therapeutic agent for concentrating therapeutics in an inflamed tissue region in the body.
  • the beneficial effects of the liposomes of WO 94/07466 (EP-0662820) are questioned in WO 03/105805, which discloses PEG-coated liposomes composed of non-charged vesicle-forming lipids, optionally containing not more than 5 mol percent of charged vesicle-forming lipids, and containing a water soluble corticosteroid for the site-specific treatment of inflammatory disorders.
  • folate-targeted liposomes carrying the entrapped fluorescent dye calcein have been shown to unload the dye in ovarian cancer cells and tumor-asssociated macrophages within tumor ascites fluid associated with an intraperitoneal cancer both ex vivo and after in vivo administration and collection of the ascites fluid for analysis.
  • the same reference discloses folate targeted liposomes in which the lipid phase includes tritiated cholesterol-oleoyl ether.
  • folate-targeted liposomal compositions comprising an anti-inflammatory agent as an entrapped agent
  • pharmaceutical compositions comprising such liposomal compositions
  • use of such compositions in the treatment of an inflammatory disease such as in an inflamed tissue region in the body.
  • Entrapped agents can be encapsulated within the aqueous interior of the liposomes and/or dissolved into the hydrocarbon regions of their bilayers.
  • folate-targeted liposomal compositions comprising an imaging or visualizing agent as an entrapped agent, as well as the use of such liposomal compositions for diagnosis or monitoring the treatment of an inflammatory disease in an inflamed tissue region in the body.
  • dendrimers conjugated to active agents such as anti-inflammatory agents
  • pharmaceutical compositions comprising such dendrimer conjugates
  • use of such compositions in the treatment of disease such as inflammatory disease are known classes of compounds which are generally regarded as repeatedly branched, approximately spherical molecules and are sometimes referred to as arborols or cascade molecules.
  • folate-targeted dendrimer conjugate compositions comprising an imaging or visualizing agent, as well as the use of such compositions for diagnosis or monitoring the treatment of a disease such as inflammatory disease in an inflamed tissue region in the body.
  • FIG. 1 shows the relative biodistribution of uptake of 3 H-cholesterol labeled folate-targeted liposomes in the aortas (1) and hearts (2) of Normal Chow (N) and Western Diet (W) mice; the biodistribution in a number of organs (aorta (1), heart (2), liver (3), kidney (4), spleen (5), intestine (6), muscle (7), skin (8) and lung (9)) is shown in FIG. 1 (bottom).
  • FIG. 2 shows whole body imaging of the atherosclerotic sites in Apo E KO mice fed a western diet for one week using a folate-targeted liposomal composition, with the entrapped fluorescent dye DiD, preinjection (top) and 2 hours post injection (bottom) in the absence (left) and presence (right) of pre-administered folic acid.
  • the scale shows low (bottom) to high (top) intensity of fluorescence.
  • FIG. 3 shows the imaging of the atherosclerotic sites in the aortic arch in Apo E KO mice fed a western diet for one week using a folate-targeted liposomal composition, with the entrapped fluorescent dye DiD. Atherosclerotic plaques showed a 5-fold increase in mean fluorescence.
  • FIG. 4 shows whole body imaging of the atherosclerotic sites in Apo E KO mice fed a western diet for four weeks before imaging under four conditions: (A) control with no imaging agent, (B) DiD-entrapped folate-targeted liposomal composition, (C) non-targeted DiD-entrapped liposomal composition, and (D) DiD-entrapped folate-targeted liposomal composition administered with an excess of folic acid.
  • FIG. 5 shows radiographic (X-ray) imaging at preinjection (0) and 2, 4, 24 and 48 hours post injection of an Apo E KO mouse dosed with an iobitridol-entrapped folate-targeted liposomal composition.
  • FIG. 6 shows radioisotopic imaging in Apo E KO mice fed a western diet for four weeks before dosing using a 99 Tc-entrapped folate-targeted liposomal composition, administered either alone (A) or with an excess of folic acid (B).
  • FIG. 7 shows radioisotopic imaging of the whole animal (top) and excised hearts (bottom) using the folate-receptor targeted, radionuclide conjugate imaging agent EC20 (folate-Tc99m) in Apo E KO mice fed a western diet for three weeks and dosed as (A) a control (Untreated) group which received a folate-targeted liposomal composition which contained only phospate buffered saline as the entrapped agent, (B) a group (NT Liposome) which received a non-targeted liposomal composition which contained betamethasone as the entrapped agent, and (C) a group (Fol-Liposome) which received a folate-targeted liposomal composition which contained betamethasone as the entrapped agent.
  • A a control (Untreated) group which received a folate-targeted liposomal composition which contained only phospate buffered saline as the en
  • FIG. 8 shows the relative EC20 heart uptake of the groups of FIG. 7 .
  • FIG. 9 shows the biodistribution of EC20 uptake in the organs (heart (1), lung (2), liver (3), spleen (4), kidney (5), muscle (6), skin (7) and intestine (8) of the groups of FIG. 7 .
  • FIG. 10 shows radioisotopic imaging (top) of the hearts (1) and aortas (8) and the biodistribution (bottom) of the organs (heart (1), lung (2), liver (3), intestine (4), kidney (5), spleen (6), muscle (7), aorta (8) and skin(9)) using the folate-receptor targeted, radionuclide conjugate imaging agent EC20 (folate-Tc99m) in Apo E KO mice fed a western diet for 4 weeks and dosed as (A) a control (Untreated) group which received a folate-targeted liposomal composition which contained only phospate buffered saline as the entrapped agent, (B) a group (NT Liposome) which received a non-targeted liposomal composition which contained betamethasone as the entrapped agent, and (C) a group (Fol-Liposome) which received a folate-targeted liposomal
  • FIG. 11 shows the imaging of the intestines of mice in an intestinal inflammation model using imaging with (A) a DiD-entrapped folate-targeted liposomal composition, (B) a DiD-entrapped folate-targeted liposomal composition administered with an excess of folic acid, and (C) a non-targeted DiD-entrapped liposomal composition.
  • FIG. 12 shows the imaging of the paws of rats in an arthritis model using imaging with (A) a DiD-entrapped folate-targeted liposomal composition, (B) a DiD-entrapped folate-targeted liposomal composition administered with an excess of folic acid, and (C) a non-targeted DiD-entrapped liposomal composition.
  • FIG. 13 shows the imaging of mice in an inflammatory muscle injury model showing a healthy mouse imaged with a folate-targeted DiD-entrapped liposomal composition (Healthy), a cardiotoxin treated mouse imaged with a non-targeted DiD-entrapped liposomal composition (NT Liposome), and a cardiotoxin treated mouse imaged with a folate-targeted DiD-entrapped liposomal composition (Fol Liposome).
  • the scale shows low (left) to high (right) intensity of uptake of the label.
  • FIG. 14 shows the cell bound fluorescence measured by flow cytometry in thioglycolate recruited macrophages for control non-targeted calcein-entrapped liposomal compositions (shaded curve), and for folate-targeted calcein-entrapped liposomal compositions in the presence (middle curve) and absence (right curve) of excess free folic acid.
  • FIG. 15 shows the imaging of the paws of rats in an arthritis model showing a diseased rat imaged with a non-targeted DiD-entrapped liposomal composition (NT Liposome), a diseased rat imaged with a folate-targeted DiD-entrapped liposomal composition (Fol-Liposome), and a healthy rat with a folate-targeted DiD-entrapped liposomal composition (Healthy).
  • NT Liposome non-targeted DiD-entrapped liposomal composition
  • Fol-Liposome a diseased rat imaged with a folate-targeted DiD-entrapped liposomal composition
  • Healthy a healthy rat with a folate-targeted DiD-entrapped liposomal composition
  • FIG. 16 shows the imaging in mice implanted with (M109) tumors which express the folate receptor at high levels on the surface and concomitantly used in the muscle injury model or the ulcerative colitis model when imaged with a folate-targeted DiD-entrapped liposomal composition (Fol-Liposome).
  • FIG. 17 shows the imaging in mice implanted with (M109) tumors which express the folate receptor at high levels on the surface and concomitantly used in the muscle injury model when imaged with a non-targeted DiD-entrapped liposomal composition (NT Liposome), with a folate-targeted DiD-entrapped liposomal composition (Fol-Liposome), and with empty folate-targeted liposome (injected empty folate-targeted liposome first, then dosed with a folate-targeted DiD-entrapped liposomal composition) (Competition with Folate Liposome).
  • NT Liposome non-targeted DiD-entrapped liposomal composition
  • Fol-Liposome folate-targeted DiD-entrapped liposomal composition
  • empty folate-targeted liposome injected empty folate-targeted liposome first, then dosed with a folate-targeted DiD-entrapped
  • FIG. 18 shows the imaging in mice implanted with (M109) tumors which express the folate receptor at high levels on the surface and concomitantly used in an intestinal inflammation model when imaged with a non-targeted DiD-entrapped liposomal composition (NT Liposome), with a folate-targeted DiD-entrapped liposomal composition (Fol-Liposome), and with empty folate-targeted liposome (injected empty folate-targeted liposome first, then dosed with a folate-targeted DiD-entrapped liposomal composition) (Fol-Liposome with Competition).
  • NT Liposome non-targeted DiD-entrapped liposomal composition
  • Fol-Liposome folate-targeted DiD-entrapped liposomal composition
  • empty folate-targeted liposome injected empty folate-targeted liposome first, then dosed with a folate-targeted DiD-entrap
  • FIG. 19 shows schematically the types of dendrimer conjugates used in the imaging studies.
  • FIG. 20 shows a synthetic scheme for the preparation of a folate-targeted dendrimer conjugate conjugated to a dye.
  • Folate-targeted dendrimer conjugates conjugated to active agents, such as anti-inflammatory agents, may be prepared similarly.
  • FIG. 21 shows imaging of cells which express the folate receptor at high levels on the surface (control) and in the presence of a folate-targeted dendrimer conjugate conjugated to FITC dye (FolDendFITC) alone and in the presence of excess folic acid (FolDendFITC w Excess Folic Acid).
  • FIG. 22 shows whole body imaging of the atherosclerotic sites in Apo E KO mice fed a western diet with a folate-targeted dendrimer conjugate conjugated to Cy5.5 dye (FolDend(G3)Cy5.5), with a non-targeted dendrimer conjugate conjugated to Cy5.5 dye, and in competition with a folate-targeted dendrimer conjugate which lacks a conjugated dye (FolDend(G3)Cy5.5 W Competion).
  • FIG. 23 shows the imaging of the atherosclerotic sites in the aortic arch in Apo E KO mice fed a western diet with a folate-targeted dendrimer conjugate conjugated to Cy5.5 dye (FolDend(G3)Cy5.5), with a non-targeted dendrimer conjugate conjugated to Cy5.5 dye, and in competition with a folate-targeted dendrimer conjugate which lacks a conjugated dye (FolDend(G3)Cy5.5 W Competion).
  • FIG. 24 shows the imaging of (the intestines of) mice in an intestinal inflammation model using imaging with a folate-targeted dendrimer conjugate conjugated to Cy5.5 dye (FolDend(G3)Cy5.5), with a non-targeted dendrimer conjugate conjugated to Cy5.5 dye, and in competition with a folate-targeted dendrimer conjugate which lacks a conjugated dye (FolDend(G3)Cy5.5 W Competion).
  • a folate-receptor targeted agent comprising a therapeutic agent useful for the treatment of inflammatory disease.
  • the folate-receptor targeted agent is a folate-targeted liposomal composition comprising an anti-inflammatory agent as an entrapped agent.
  • the folate-receptor targeted agent is a folate-targeted dendrimer conjugate comprising a folate-targeted dendrimer conjugated to an anti-inflammatory agent.
  • a folate-targeted liposomal composition comprising an anti-inflammatory agent as an entrapped agent.
  • the embodiment of the liposome may be as described herein or of any conventional composition and may be constructed as described herein or by any conventional methodology with the folate-targeting ligand attached as described herein or by any conventional linkage structure or methodology.
  • the lipid components used in forming the embodiments of the liposomes may be any of the variety of vesicle-forming lipids, including phospholipids, sphingolipids, and/or other lipids, including sterols such as cholesterol and cholesterol ester salts.
  • Suitable lipids for the embodiments include those having two hydrocarbon chains, typically acyl chains, and a polar head group, such as phospholipids and glycolipids.
  • phospholipid includes any one phospholipid or combination of phospholipids capable of forming liposomes.
  • Phospholipids include phosphatidylcholines (PC), phosphatidylethanolamine (PE), phosphatidic acid (PA), phosphatidylinositol (PI), and sphingomyelin (SM), where the two hydrocarbon chains are typically between about 14-22 carbons in length, and have varying degrees of unsaturation.
  • the glycolipids include cerebrosides and gangliosides.
  • Phosphatidylcholines including those obtained from natural sources or those that are partially or wholly synthetic, or of variable chain length and unsaturation, are suitable.
  • Phospholipids which contain saturated alkyl chains, yielding a relatively high transition temperature, are suitable for the embodiments.
  • DSPE distearoylphosphatidylethanolamine
  • DSPC distearoylphosphatidylcholine
  • HSPC hydrogenated soy phosphatidylcholine
  • the folate-targeting components used in forming the embodiments of the liposomes may be any of those known in the preparation of folate-targeted liposomes or those described herein.
  • preparation of folate-targeted liposomal compositions see, for example, R. J. Lee et al., J. Biological Chemistry, 269(5) (1994) 3198-3204; and R. J. Lee et al., Biochimica et Biophysica Acta, 1233 (1995) 134-144.
  • lipid bilayer comprises a comprises a folate targeting conjugate composed of (a) a lipid having a polar head group and a hydrophobic tail, (b) a hydrophilic polymer having a first end and a second end, said polymer attached at its first end to the head group of the lipid, and (c) a folate ligand (Fol) attached to the second end of the polymer, and wherein the folate ligand is a folic acid residue or an analog or derivative thereof.
  • a folate targeting conjugate composed of (a) a lipid having a polar head group and a hydrophobic tail, (b) a hydrophilic polymer having a first end and a second end, said polymer attached at its first end to the head group of the lipid, and (c) a folate ligand (Fol) attached to the second end of the polymer, and wherein the folate ligand is a folic acid residue or an analog or derivative thereof.
  • Folic acid analogs or derivatives thereof include the (unnatural) D-isomer of folic acid and derivatives other related folic acid receptor binding molecules, such as folinic acid, pteroic acid, pteropolyglutamic acid, receptor-binding pteridines such as tetrahydropterins, dihydrofolates, tetrahydrofolates, and their deaza and dideaza analogs.
  • the terms “deaza” and “dideaza” analogs refer to the art-recognized folate analogs having a carbon atom substituted for one or two nitrogen atoms in the naturally occurring folic acid structure.
  • Other folate analogs or derivatives include the folate receptor-binding analogs aminopterin, amethopterin (methotrexate), and derivatives thereof.
  • the hydrophilic polymer of the folate targeting conjugate is a PEG having an average molecular weight of about 200-5000 in which the end groups, prior to attachment, are independently amino, hydroxy, thiol or carboxy. In one embodiment the hydrophilic polymer of the folate targeting conjugate is a PEG having an average molecular weight of about 3200-3400 in which the end groups, prior to attachment, are independently amino or thiol.
  • the hydrophilic polymer of the folate targeting conjugate is a PEG having an average molecular weight of about 3200-3400 in which the end groups, prior to attachment, are each amino, such as for example, NH 2 PEG 3200 NH 2 or NH 2 PEG 3400 NH 2 , which also may be denoted as a polyoxyethylene bis-amine or a PEG bis-amine, such as for example PEG bis-amine, M r ⁇ 3350.
  • the hydrophilic polymer of the folate targeting conjugate is a PEG having an average molecular weight of about 3200-3400 in which the end groups, prior to attachment, are amino and thiol, which also may be denoted amino-PEG-SH.
  • An amino-PEG-SH may be obtained from the corresponding PEG bis-amine by a conventional method, such as by the addition of Traut's reagent.
  • the folate-targeted liposomal composition is a phosphatidyl group as defined herein.
  • the phosphatidyl group comprises two saturated acyl groups each of which is between about 14-22 carbons in length.
  • the phosphatidyl group comprises two saturated acyl groups each of which is between about 16-18 carbons in length.
  • the phosphatidyl group is a distearoyl (DS) derivative.
  • the polar head group of the lipid is attached to the first end of the hydrophilic polymer by any conventional method.
  • the polar head group is part of a phosphatidylethanolamine, wherein the phosphatidyl group has any of the meanings herein, and the amino group of the ethanolamine portion is linked to an amino group of the first end of the hydrophilic polymer by a bis-acyl group, such as a succinyl group.
  • a succinyl group also may be denoted as a 1,4-dioxobutane-1,4-diyl group.
  • the polar head group is part of a phosphatidylethanolamine, wherein the phosphatidyl group has any of the meanings herein, and the amino group of the ethanolamine portion is linked to a thiol group of the first end of the hydrophilic polymer via addition of the thiol group to an N-maleiimidocaproylphospatidylethanolamine.
  • the phosphatidylethanolamine is distearoylphosphatidylethanolamine (DSPE).
  • the folate ligand is a folic acid residue which is attached to an amino group at the second end of the hydrophilic polymer by acylation with the a-carbonyl group or the ⁇ -carbonyl group of the glutamic acid portion of folic acid.
  • the folate ligand is a folic acid residue which is attached to an amino group at the second end of the hydrophilic polymer by acylation with the ⁇ -carbonyl group of the glutamic acid portion of folic acid.
  • the folate targeting conjugate comprises distearoylphosphatidylethanolamine linked to the first end of a bis-amino PEG and a folic acid residue which is attached to the amino group at the second end of the bis-amino PEG by acylation with the ⁇ -carbonyl group of the glutamic acid portion of folic acid.
  • the distearoylphosphatidylethanolamine is linked to the first end of the bis-amino PEG by a succinyl group.
  • a further embodiment of the above liposomal composition is one wherein the folate targeting conjugate comprises distearoylphosphatidylethanolamine wherein the amino group of the ethanolamine portion is linked by a succinyl group to the amino group of the first end of a PEG having an average molecular weight of about 3200-3400 in which the end groups, prior to attachment, are each amino, and wherein the folate ligand is a folic acid residue which is attached to the amino group at the second end of the PEG by acylation with the ⁇ -carbonyl group of the glutamic acid portion of folic acid.
  • the folate targeting conjugate is present at about 0.01 mol % to about 1 mol % in the lipid bilayer. In another embodiment, the folate targeting conjugate is present at about 0.05 mol % to about 0.5 mol % in the lipid bilayer. In another embodiment, the folate targeting conjugate is present at about 0.1 mol % in the lipid bilayer.
  • One embodiment is a liposomal composition as described herein wherein the lipid bilayer further comprises a hydrophilic coating composed of (a) a lipid having a polar head group and a hydrophobic tail and (b) a hydrophilic polymer having a first end and a second end, said polymer attached at its first end to the head group of the lipid and optionally capped at its second end.
  • the hydrophilic coating components used in forming the embodiments of the liposomes may be any of those known in the preparation of hydrophilic coated liposomes or those described herein.
  • the lipid of the hydrophilic coating having a polar head group and a hydrophobic tail can be any of those disclosed herein for the preparation of a folate targeting conjugate.
  • the lipid having a polar head group and a hydrophobic tail is a phosphatidyl group as defined herein.
  • the phosphatidyl group comprises two saturated acyl groups each of which is between about 14-22 carbons in length.
  • the phosphatidyl group comprises two saturated acyl groups each of which is between about 16-18 carbons in length.
  • the phosphatidyl group is a distearoyl (DS) derivative.
  • the hydrophilic polymer of the hydrophilic coating can be any of a number of biocompatible hydrophilic polymers.
  • the hydrophilic polymer is a polyethylene glycol (PEG), a polylactic acid (PLA), a polyglycolic acid (PGA) or a polyvinyl alcohol (PVA), as well as copolymers of lactic and glycolic acids, in which the first end group, prior to attachment, is amino, hydroxy or thiol.
  • the hydrophilic polymer is optionally capped with methyl, ethyl, or another unreactive group.
  • the hydrophilic polymer is a polyethylene glycol with an average molecular weight of about 2000, which is uncapped or capped with a methyl or ethyl group.
  • the polar head group of the lipid is attached to the first end of the hydrophilic polymer of the hydrophilic coating by any conventional method.
  • the polar head group is part of a phosphatidylethanolamine, wherein the phosphatidyl group has any of the meanings herein, and the amino group of the ethanolamine portion is linked directly to a carbon of the first end of the hydrophilic polymer, or via a linker to an amino, hydroxy or thiol group of the first end of the hydrophilic polymer by a linking group.
  • the amino group of the ethanolamine portion may be linked directly to a carbon of the first end of the hydrophilic polymer, for example by converting a hydroxy group to a leaving group, such as a triflate, and alkylating the amino group of the ethanolamine.
  • the amino group of the ethanolamine portion of a phosphatidylethanolamine may be linked to a hydroxy group of the first end of the hydrophilic polymer, for example, by a linking group such as a carbonyl group or a 4-chloro-1,3,5-triazinyl group; or the amino group of the ethanolamine portion may be linked to an amino group of the first end of the hydrophilic polymer, for example, by a linking group such as a carbonyl group, a succinyl group or a 4-chloro-1,3,5-triazinyl group.
  • the amino group of the ethanolamine portion of a phosphatidylethanolamine may be linked to a thiol group of the first end of the hydrophilic polymer by formation of a disulfide bond between an alkyl thiol group on the amino group of the ethanolamine and the thiol group of the hydrophilic polymer.
  • the group at the second end may be protected using a conventional protecting group while the first end is attached to the polar head group of the lipid of the hydrophilic coating having a polar head group and a hydrophobic tail; then the protecting group is removed.
  • a hydroxy group at the second end of the hydrophilic polymer may be protected using a trimethylsilyl group, which is removed following the attachment of the first end of the hydrophilic polymer to the polar head group.
  • One embodiment is a liposomal composition as described herein wherein the hydrophilic coating is present at about 1 to 20 mol % in the lipid bilayer.
  • Another embodiment is a liposomal composition as described herein wherein the hydrophilic coating is present at about 4 mol % in the lipid bilayer.
  • One embodiment is a liposomal composition as described herein wherein the liposome bilayer comprises a mol ratio of DSPC/cholesterol/mPEG2000-DSPE of about 56:40:4.
  • Another embodiment is a liposomal composition as described herein wherein the liposome bilayer comprises a mol ratio of DSPC/Chol/mPEG 2000 -DSPE/Fol-PEG 3400 DSPE of about 56:40:4:0.1.
  • One embodiment is a liposomal composition as described herein wherein the average particle size of the liposome is about 30 to 200 nM. Another embodiment is a liposomal composition as described herein wherein the average particle size of the liposome is about 40 to 120 nM. A further embodiment is a liposomal composition as described herein wherein the average particle size of the liposome is about 50 to 100 nM.
  • a further embodiment of folate-targeted liposomal compositions is the ability to deliver more of the entrapped agent to an inflamed site than corresponding non-targeted liposomal compositions, which are the same as the folate-targeted liposomal compositions, except the folate targeting conjugate, such as folate-PEG-DSPE, is omitted.
  • One embodiment is a liposomal composition, or folate-targeted dendrimer conjugate, as described herein wherein the entrapped anti-inflammatory agent is an anti-inflammatory steroid.
  • the anti-inflammatory steroid is a sytemically administered (lipophilic) anti-inflammatory steroid.
  • the anti-inflammatory steroid is betamethasone, dexamethasone, flumethasone, methylprednisolone, paramethasone, prednisolone, prednisone, triamcinolone, hydrocortisone or cortisone.
  • a further embodiment is a liposomal composition as described herein wherein the entrapped agent is betamethasone.
  • the anti-inflammatory steroid is a topically administered anti-inflammatory steroid.
  • topically administered includes administration by any of a number of nonsystemic means, including by inhalation, suppository, topical cream ointment, foam, lotion or gel, etc.
  • the anti-inflammatory steroid is alcomethasone dipropionate, amcinonide, betamethasone dipropionate, betamethasone monopropionate, betamethasone 17-valerate, budesonide, budesonide disodium phosphate, ciclomethasone, clobetasol-17-propionate, clobetasone-17-butyrate, cortisone acetate, deprodone propionate, desonide, desoxymethasone, dexamethasone acetate, diflucortolone valerate, diflurasone diacetate, diflucortolone, difluprednate, flumetasone pivalate, flunisolide, fluocinolone acetonide acetate, fluocinonide, fluocortolone, fluocortolone caproate, fluocortolone hexanoate, fluocortolone pivalate, fluor
  • the anti-inflammatory steroid is a water soluble anti-inflammatory steroid.
  • Another embodiment is a liposomal composition as described herein wherein the anti-inflammatory steroid is betamethasone sodium phosphate, desonide sodium phosphate, dexamethasone sodium phosphate, hydrocortisone sodium phosphate, hydrocortisone sodium succinate, cortisone sodium phosphate, cortisone sodium succinate, methylprednisolone disodium phosphate, methylprednisolone sodium succinate, methylprednisone disodium phosphate, methylprednisone sodium succinate, prednisolone sodium phosphate, prednisolone sodium succinate, prednisone sodium phosphate, prednisone sodium succinate, prednisolamate hydrochloride, triamcinolone acetonide disodium phosphate or triamcinolone acetonide dipotassium phosphate.
  • One embodiment is a liposomal composition, or folate-targeted dendrimer conjugate, as described herein wherein the entrapped anti-inflammatory agent is a non-steroidal anti-inflammatory drug (NSAID), which also may be denoted as a non-steroidal anti-inflammatory agent (NSAIA) or as a non-steroidal anti-inflammatory medicine (NSAIM).
  • NSAID non-steroidal anti-inflammatory drug
  • the NSAID comprises a propionic acid derivative such as, for example, ibuprofen, naproxen, fenoprofen, ketoprofen, flurbiprofen or oxaprozin.
  • the NSAID comprises an acetic acid derivative, such as, for example, indomethacin, sulindac, etodolac or diclofenac.
  • the NSAID comprises an oxicam derivative, such as, for example, piroxicam, meloxicam, tenoxicam, droxicam, lornoxicam or isoxicam.
  • the NSAID comprises a fenamic acid derivative, such as, for example, mefenamic acid, meclofenamic acid, flufenamic acid or tolfenamic acid.
  • the NSAID comprises a selective COX-2 (cyclooxygenase-2) inhibitor (coxib), such as, for example, celecoxib, rofecoxib, valdecoxib, parecoxib, lumiracoxib or etoricoxib.
  • COX-2 cyclooxygenase-2
  • coxib selective COX-2 (cyclooxygenase-2) inhibitor
  • Another embodiment is a liposomal composition, or folate-targeted dendrimer conjugate, as described herein wherein the entrapped anti-inflammatory agent comprises another drug useful in the treatment of rheumatoid arthritis or other autoimmune disease including an antiproliferative, immunomodulator or immunosuppresant agent, comprising, for example, aspirin, methotrexate, sulfasalazine, D-penicillamine, nambumetone, aurothioglucose, auranofin, other gold-containing compound, colloidal gold, cyclosporin, tacrolimus, pimecrolimus or sirolimus.
  • an antiproliferative, immunomodulator or immunosuppresant agent comprising, for example, aspirin, methotrexate, sulfasalazine, D-penicillamine, nambumetone, aurothioglucose, auranofin, other gold-containing compound, colloidal gold, cycl
  • liposomes are useful carriers both of hydrophilic, lipophobic agents, which can reside entrapped in the aqueous interior of the liposome, and of hydrophobic, lipophilic agents, which can reside entrapped in the lipid bilayer, it is possible to load a lipsome as disclosed herein simultaneously with more than one anti-inflammatory agent, any of which may be hydrophilic or lipophilic. Similarly, it is possible to load a lipsome as disclosed herein simultaneously with an anti-inflammatory agent and a further therapeutic agent, any of which may be hydrophilic or lipophilic.
  • a liposomal composition as described herein comprising more than one anti-inflammatory agent as an entrapped agent.
  • Another embodiment is a liposomal composition as described herein wherein there are two entrapped anti-inflammatory agents.
  • a further embodiment is a liposomal composition as described herein comprising an anti-inflammatory agent and a further therapeutic agent.
  • a pharmaceutical composition comprising a liposomal composition comprising an anti-inflammatory agent as an entrapped agent, or folate-targeted dendrimer conjugate, as described herein and further comprising at least one pharmaceutically acceptable carrier or excipient.
  • the pharmaceutical composition may be formulated in a conventional manner as known for the formulation of liposomes for administration to patients.
  • the pharmaceutical composition may be a sterile dispersion in a buffered aqueous vehicle, optionally containing a tonicity adjusting agent.
  • the pharmaceutical composition may be a sterile, non-pyrogenic lyophilized product comprising a buffer and/or a tonicity adjusting agent, which is suitable for reconstitution with sterile water for injection to form a sterile dispersion.
  • the sterile dispersion may diluted in 5% dextrose to an appropriate concentration prior to intravenous infusion.
  • a pharmaceutical composition includes a veterinary composition.
  • a liposomal composition comprising an anti-inflammatory agent as an entrapped agent, or folate-targeted dendrimer conjugate, as described herein for use as a medicament.
  • a liposomal composition comprising an anti-inflammatory agent as an entrapped agent, or folate-targeted dendrimer conjugate, as described herein for use in the treatment of an inflammatory disease.
  • a liposomal composition comprising an anti-inflammatory agent as an entrapped agent, or folate-targeted dendrimer conjugate, as described herein for the treatment of an inflammatory disease.
  • a liposomal composition comprising an anti-inflammatory agent as an entrapped agent, or folate-targeted dendrimer conjugate, as described herein for the manufacture of a medicament for the treatment of an inflammatory disease.
  • a method of treatment of an inflammatory disease in a subject in need thereof comprising administering an effective amount of a liposomal composition comprising an anti-inflammatory agent as an entrapped agent, or folate-targeted dendrimer conjugate, as described in any of claims.
  • a liposomal composition comprising an anti-inflammatory agent as an entrapped agent, or folate-targeted dendrimer conjugate, as described in any of claims.
  • the subject is a mammal.
  • the subject is a human.
  • the inflammatory disease comprises an inflamed tissue region in the body.
  • the use or method is one wherein the inflammatory disease is arthritis, arteriosclerosis, graft-versus-host disease, multiple sclerosis, osteomyelitis, psoriasis or inflammatory bowel disease, such as Crohn's disease or ulcerative colitis.
  • the use or method one wherein the inflammatory disease is arthritis.
  • the use or method one wherein the inflammatory disease is rheumatoid arthritis.
  • the use or method one wherein the inflammatory disease is osteoarthritis.
  • the use or method one wherein the inflammatory disease is arteriosclerosis.
  • the use or method one wherein the inflammatory disease is atherosclerosis.
  • the use or method one wherein the inflammatory disease is inflammatory bowel disease, such as Crohn's disease or ulcerative colitis.
  • a folate-targeted liposomal composition, or folate- targeted dendrimer conjugate comprising an imaging or visualizing agent as an entrapped agent for diagnosis or monitoring the treatment of an inflammatory disease in an inflamed tissue region in the body.
  • a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate comprising an imaging or visualizing agent as an entrapped agent for the manufacture of an agent for diagnosis or monitoring the treatment of an inflammatory disease in an inflamed tissue region in the body.
  • Another embodiment is an agent for diagnosis or monitoring the treatment of an inflammatory disease, comprising a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate, comprising an imaging or visualizing agent as an entrapped agent.
  • a pharmaceutical composition comprising a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate, comprising an imaging or visualizing agent as an entrapped agent and further comprising at least one pharmaceutically acceptable carrier or excipient.
  • a further embodiment is a method of using a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate, comprising an imaging or visualizing agent as an entrapped agent for diagnosis or monitoring the treatment of an inflammatory disease in an inflamed tissue region in the body in a subject in need thereof.
  • a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate comprising an imaging or visualizing agent as an entrapped agent for diagnosis or monitoring the treatment of an inflammatory disease in an inflamed tissue region in the body in a subject in need thereof.
  • the subject is a mammal.
  • Another embodiment of the method is wherein the subject is a human.
  • agent, pharmaceutical composition or method of using a folate-targeted liposomal composition wherein the entrapped agent is a folate-targeted liposomal composition comprising an imaging or visualizing agent, other than the difference in the entrapped agent embodiments of the folate-targeted liposomal composition may be characterized in the same manner as embodiments of a folate-targeted liposomal composition comprising an anti-inflammatory agent as an entrapped agent as described herein.
  • embodiments may be characterized in the same manner as embodiments comprising an anti-inflammatory agent.
  • the entrapped or conjugated agent is a pharmaceutically acceptable fluorescent dye.
  • the fluorescent dye is DiD (1,1′-dioctadecyl-3,3,3′,3′-tetramethyl-indodicarbocyanine perchlorate), calcein or fluorescein.
  • agent, pharmaceutical composition or method is one wherein the entrapped or conjugated agent is a contrast agent for X-ray, MRI (magnetic resonance imaging) or ultrasound.
  • MRI magnetic resonance imaging
  • ultrasound ultrasound
  • the X-ray contrast agent is iobitridol.
  • the entrapped or conjugated agent comprises a radionuclide.
  • the radionuclide is an isotope of gallium, indium, copper, technitium or rhenium. In another embodiment, the radionuclide is an isotope of technitium.
  • kits comprising a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate, comprising an imaging or visualizing agent as an entrapped agent as described herein and instructions for diagnosis or monitoring the treatment of an inflammatory disease.
  • a folate-targeted liposomal composition comprising the fluorescent dye calcein has been described.
  • a folate-targeted liposomal composition comprising an imaging or visualizing agent other than a fluorescent dye as an entrapped agent provides a novel embodiment.
  • embodiments of the folate-targeted liposomal composition may be characterized in the same manner as embodiments of a folate-targeted liposomal composition comprising an anti-inflammatory agent as an entrapped agent as described herein.
  • the folate-targeted liposomal composition, or folate-targeted dendrimer conjugate, comprising an imaging or visualizing agent other than a fluorescent dye as an entrapped agent is one wherein the entrapped agent is an X-ray contrast agent.
  • the X-ray contrast agent is iobitridol.
  • the folate-targeted liposomal composition, or folate-targeted dendrimer conjugate, comprising an imaging or visualizing agent other than a fluorescent dye as an entrapped agent is one wherein the entrapped agent comprises a radionuclide.
  • the radionuclide is an isotope of gallium, indium, copper, technitium or rhenium. In one embodiment, the radionuclide is an isotope of technitium.
  • One embodiment is a pharmaceutical composition comprising a liposomal composition, or folate-targeted dendrimer conjugate, comprising an imaging or visualizing agent other than a fluorescent dye as an entrapped agent and further comprising at least one pharmaceutically acceptable carrier or excipient.
  • a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate comprising an imaging or visualizing agent other than a fluorescent dye as an entrapped agent for diagnosis or monitoring the treatment of an inflammatory disease in an inflamed tissue region in the body.
  • a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate comprising an imaging or visualizing agent other than a fluorescent dye as an entrapped agent for the manufacture of an agent for diagnosis or monitoring the treatment of an inflammatory disease in an inflamed tissue region in the body.
  • an agent for diagnosis or monitoring the treatment of an inflammatory disease comprising a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate, comprising an imaging or visualizing agent other than a fluorescent dye as an entrapped agent.
  • a further embodiment is a method of using a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate, comprising an imaging or visualizing agent other than a fluorescent dye as an entrapped agent for diagnosis or monitoring the treatment of an inflammatory disease in an inflamed tissue region in the body in a subject in need thereof.
  • a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate comprising an imaging or visualizing agent other than a fluorescent dye as an entrapped agent for diagnosis or monitoring the treatment of an inflammatory disease in an inflamed tissue region in the body in a subject in need thereof.
  • the subject is a mammal.
  • Another embodiment of the method is wherein the subject is a human.
  • kits comprising a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate, comprising an anti-inflammatory agent as an entrapped agent, or a pharmaceutical composition thereof, as described herein, and an imaging agent for the inflammatory condition.
  • the imaging agent is a folate-targeted imaging agent.
  • the folate-targeted imaging agent is a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate, as described herein.
  • the folate-targeted imaging agent is a folate-targeted conjugate of an imaging agent.
  • the folate-targeted conjugate is EC20 (folate-Tc99m).
  • kits further comprising instructions for diagnosis or monitoring the treatment of an inflammatory disease.
  • An additional embodiment of any of the above described kits is a kit further comprising instructions for the treatment of an inflammatory disease.
  • a folate-receptor targeted agent comprising a therapeutic agent useful for the treatment of inflammatory disease.
  • the folate-receptor targeted agent of clause 1 which is a folate-targeted liposomal composition comprising an anti-inflammatory agent as an entrapped agent.
  • lipid bilayer comprises a comprises a folate-targeting conjugate composed of (a) a lipid having a polar head group and a hydrophobic tail, (b) a hydrophilic polymer having a first end and a second end, said polymer attached at its first end to the head group of the lipid, and (c) a folate ligand (Fol) attached to the second end of the polymer, and wherein the folate ligand is a folic acid residue or an analog or derivative thereof.
  • a folate-targeting conjugate composed of (a) a lipid having a polar head group and a hydrophobic tail, (b) a hydrophilic polymer having a first end and a second end, said polymer attached at its first end to the head group of the lipid, and (c) a folate ligand (Fol) attached to the second end of the polymer, and wherein the folate ligand is a folic acid residue or an analog or derivative thereof.
  • hydrophilic polymer of the folate targeting conjugate is a PEG having an average molecular weight of about 200-5000 in which the end groups, prior to attachment, are independently amino, hydroxy, thiol or carboxy.
  • lipid bilayer further comprises a hydrophilic coating composed of (a) a lipid having a polar head group and a hydrophobic tail and (b) a hydrophilic polymer having a first end and a second end, said polymer attached at its first end to the head group of the lipid and optionally capped at its second end.
  • hydrophilic coating is a polyethylene glycol (PEG), a polylactic acid (PLA), a polyglycolic acid (PGA) or a polyvinyl alcohol (PVA).
  • PEG polyethylene glycol
  • PLA polylactic acid
  • PGA polyglycolic acid
  • PVA polyvinyl alcohol
  • a method of treatment of an inflammatory disease in a subject in need thereof comprising administering an effective amount of a liposomal composition as described in any of clauses 2-12.
  • a pharmaceutical composition comprising a liposomal composition comprising an anti-inflammatory agent as an entrapped agent as described in any of clauses 2-12 and further comprising at least one pharmaceutically acceptable carrier or excipient.
  • a composition comprising a folate-targeted liposomal composition comprising an imaging or visualizing agent as an entrapped agent and further comprising at least one pharmaceutically acceptable carrier or excipient.
  • a method of using a folate-targeted liposomal composition comprising an imaging or visualizing agent as an entrapped agent for diagnosis or monitoring the treatment of an inflammatory disease in an inflamed tissue region in the body in a subject in need thereof.
  • kits comprising a folate-targeted liposomal composition comprising an imaging or visualizing agent as an entrapped agent as described in clause 14 and instructions for diagnosis or monitoring the treatment of an inflammatory disease.
  • kits comprising a folate-targeted liposomal composition comprising an anti-inflammatory agent as an entrapped agent, as described in any of clauses 2-11, or a pharmaceutical composition thereof, as described in clause 13, and an imaging agent for the inflammatory condition.
  • the folate-receptor targeted agent of clause 1 which is a folate-targeted dendrimer conjugate comprising a folate-targeted dendrimer conjugated to an anti-inflammatory agent.
  • the folate-targeting ligand Fol
  • the folate-targeting ligand is a folic acid residue or an analog or derivative thereof, a hydrophilic coating, and a conjugated anti-inflammatory agent.
  • a method of treatment of an inflammatory disease in a subject in need thereof comprising administering an effective amount of a folate-targeted dendrimer conjugate as described in any of clauses 18-24.
  • a pharmaceutical composition comprising a folate-targeted dendrimer conjugate comprising an anti-inflammatory agent as described in any of clauses 18-24 and further comprising at least one pharmaceutically acceptable carrier or excipient.
  • a composition comprising a folate-targeted dendrimer conjugate comprising an imaging or visualizing agent as a conjugated agent and further comprising at least one pharmaceutically acceptable carrier or excipient.
  • kits comprising a composition comprising a folate-targeted dendrimer conjugate comprising an imaging or visualizing agent as a conjugated agent as described in clause 27 and instructions for diagnosis or monitoring the treatment of an inflammatory disease.
  • kits comprising a folate-targeted dendrimer conjugate comprising an anti-inflammatory agent as described in clause 18-24, or a pharmaceutical composition thereof, as described in clause 26, and an imaging agent for the inflammatory condition.
  • Arteriosclerosis has traditionally been viewed to simply reflect the deposition of lipids within the vessel wall of medium-sized and large arteries. Upregulation of cell adhesion molecules facilitates adherence of leukocytes to the dysfunctional endothelium and their subsequent transmigration into the vessel wall. Evolving inflammatory reaction results in the initiation of atherosclerotic plaques.
  • activated macrophages constitute the key effector cells in atherosclerosis; and it was reported that the folate receptor, FR ⁇ , the nonepithelial isoform of the folate receptor is expressed on activated (but not resting) macrophages.
  • Imaging Example 1 That the instant folate-targeted liposomal composition can deliver its entrapped agent to inflammation induced activated macrophages is demonstrated in Imaging Example 1, below.
  • the uptake of calcein from a calcein-entrapped folate-targeted liposomal composition by cells from rat peritoneal fluid in vitro is demonstrated in a thioglycolate-induced inflammation model using flow cytometry ( FIG. 14 ).
  • the calcein uptake is competitively inhibited by incubation in the presence of excess folic acid; and the comparable nontargeted (NT) liposomal composition delivers significantly less calcein.
  • the dye within the liposomes is quenched at high concentration, the fluorescence is observed only following endocytosis and calcein unloading inside the target cells. Accordingly, such imaging experiments support the unloading of therapeutic agents in similar situations.
  • Apo E mice with the knock out mutation show a marked increase in total plasma cholesterol levels that are unaffected by age or sex. Fatty streaks in the proximal aorta are found at 3 month of age. The lesions increase with age and progress to lesions with less lipid but more elongated cells, typical of a more advanced stage of pre-atherosclerotic lesions.
  • mice fed a normal diet show obvious atherosclerotic lesions in the aortic sinus and the ascending aorta. The number of atherosclerotic lesions in the aortic sinus and the ascending aorta is significantly increased in homozygotes fed an atherosclerotic western diet vs a normal diet.
  • Imaging Example 2 demonstrates the increased uptake of of 3 H-cholesterol labeled folate-targeted liposomes in the aorta and hearts of mice fed a high fat western diet compared to a normal diet, indicating the increase in atheroscerotic lesions in the western diet animal.
  • Imaging Examples 3 through 6 demonstrate the ability of folate-targeted liposomal compositions to deliver entrapped imaging agents to atherosclerotic sites in an atherosclerosis model employing Apo E knock out (Apo E KO) mice.
  • Therapeutic Examples 1 and 2 demonstrate the ability of betamethasone-entrapped folate-targeted liposomal compositions to reduce the evidence of atherosclerotic inflammation in Apo E KO mice.
  • Imaging Example 3 ability of a folate-targeted liposomal composition, with the entrapped fluorescent dye DiD, to label the atherosclerotic sites in Apo E KO mice fed a western diet for one week before dosing is demonstrated along with the competitive blocking of the labeling by pre-administered free folic acid. Imaging of both the whole animals ( FIG. 2 ) and the excised aortic arch ( FIG. 3 ) show the labeling of the sites and competitive blocking.
  • Imaging Example 4 provides a similar example in Apo E KO mice fed a western diet for four weeks before imaging under four conditions: (A) control with no imaging agent, (B) DiD-entrapped folate-targeted liposomal composition, (C) non-targeted DiD-entrapped liposomal composition, and (D) DiD-entrapped folate-targeted liposomal composition administered with an excess of folic acid.
  • mice of each of the four treatment groups demonstrate superior uptake of the dye from the DiD-entrapped folate-targeted liposomal composition compared to the non-targeted DiD-entrapped liposomal composition, as well as the competitive inhibition of uptake of the dye from the DiD-entrapped folate-targeted liposomal composition by excess folic acid.
  • Imaging Example 5 provides an example of an Apo E KO mouse dosed with an iobitridol-entrapped folate-targeted liposomal composition which demonstrates the use of an entrapped CT agent for radiographic imaging ( FIG. 5 ).
  • Imaging Example 6 provides an example using radioisotopic imaging in Apo E KO mice fed a western diet for four weeks before dosing using a 99 Tc-entrapped folate-targeted liposomal composition, adminisered either alone or with an excess of folic acid ( FIG. 6 ), demonstrating the competitive blockage of uptake of the folate-targeted liposomal composition by excess folic acid.
  • mice were imaged using the folate-receptor targeted, radionuclide conjugate imaging agent EC20 (folate-Tc99m) and the biodistribution of EC20 uptake was determined.
  • the reduction in inflammatory cells imaged by EC20 in the Fol Liposome group compared to the NT Liposome and Untreated groups is demonstrated by the imaged mice ( FIG. 7 , top) and excised hearts ( FIG. 7 , bottom), particulary the heart uptake of EC20 ( FIG. 8 ), and biodistribution ( FIG. 9 ).
  • mice were imaged using the folate-receptor targeted, radionuclide conjugate imaging agent EC20 (folate-Tc99m) and the biodistribution of EC20 uptake was determined.
  • the reduction in inflammatory cells imaged by EC20 in the Fol Liposome group compared to the NT Liposome and Untreated groups is demonstrated by the measures of EC20 uptake in the hearts and the aortas of the groups FIG. 10 (top); FIG. 10 (bottom) shows the relative biodistribution of EC20 uptake in the organs of the groups.
  • Imaging Examples 7 through 9 demonstrate the ability of folate-targeted liposomal compositions to deliver entrapped imaging agents to sites of inflammation in other tissues and support the use of folate-targeted liposomal compositions comprising an anti-inflammatory agent as an entrapped agent, as disclosed herein, in the treatment of an inflammatory disease, such as in an inflamed tissue region in the body.
  • Imaging Example 7 demonstrates ( FIG. 11 ) the ability of the folate-targeted liposomal compositions to deliver entrapped agents to inflamed intestinal tissues, for example in inflammatory bowel disease, such as Crohn's disease or ulcerative colitis, using imaging under the following conditions: (A) DiD-entrapped folate-targeted liposomal composition, (B) DiD-entrapped folate-targeted liposomal composition administered with an excess of folic acid, and (C) non-targeted DiD-entrapped liposomal composition.
  • A DiD-entrapped folate-targeted liposomal composition
  • B DiD-entrapped folate-targeted liposomal composition administered with an excess of folic acid
  • C non-targeted DiD-entrapped liposomal composition.
  • Imaging Example 8 demonstrates ( FIG. 12 ) the ability of the folate-targeted liposomal compositions to deliver entrapped agents to inflamed arthritic tissues, for example as in rheumatoid arthritis, using imaging under the following conditions: (A) DiD-entrapped folate-targeted liposomal composition, (B) DiD-entrapped folate-targeted liposomal composition administered with an excess of folic acid, and (C) non-targeted DiD-entrapped liposomal composition.
  • Imaging Example 8A also demonstrates ( FIG. 15 ) the ability of the folate-targeted liposomal compositions to deliver entrapped agents to inflamed arthritic tissues, for example as in rheumatoid arthritis, using imaging under the following conditions: a non-targeted DiD-entrapped liposomal composition (NT Liposome), a diseased rat imaged with a folate-targeted DiD-entrapped liposomal composition (Fol-Liposome), and a healthy rat with a folate-targeted DiD-entrapped liposomal composition (Healthy).
  • NT Liposome non-targeted DiD-entrapped liposomal composition
  • Fol-Liposome a diseased rat imaged with a folate-targeted DiD-entrapped liposomal composition
  • Healthy a healthy rat with a folate-targeted DiD-entrapped liposomal composition
  • Imaging Example 9 demonstrates ( FIG. 13 ) the ability of the folate-targeted liposomal compositions to deliver entrapped agents to inflamed skeletal muscle tissues using imaging with (A) a DiD-entrapped folate-targeted liposomal composition, and (B) a DiD-entrapped folate-targeted liposomal composition administered with an excess of folic acid.
  • Imaging Examples 10 and 11 demonstrate ( FIG. 16 , FIG. 17 and FIG. 18 ) that the folate-targeted liposomal compositions preferentially deliver entrapped agents to inflammatory tissues as compared to tumors which express a high level of folate receptors on their surface.
  • the examples demonstrate that the increase in uptake of the folate-targeted liposomal compositions compared to the non-targeted liposomal compositions is greater in the inflammatory tissues than the increase in the tumors.
  • Imaging Example 12 demonstrates uptake of a folate-targeted dendrimer conjugate conjugated to FITC dye and the competitive inhibition of the uptake by excess folic acid in the cells which express the folate receptor at high levels on the surface.
  • Imaging Example 13 ability of a dendrimer conjugate conjugated to Cy5.5 dye to label the atherosclerotic sites in Apo E KO mice fed a western diet before dosing is demonstrated along with a comparison with a non-targeted dendrimer conjugate conjugated to Cy5.5 dye and the competitive blocking of the labeling by competition with a folate-targeted dendrimer conjugate which lacks a conjugated dye.
  • Imaging of both the whole animals ( FIG. 22 ) and the excised aortic arch ( FIG. 23 ) show the labeling of the sites and competitive blocking.
  • Imaging Example 14 demonstrates ( FIG. 24 ) the ability of a dendrimer conjugate to deliver conjugated agents to inflamed intestinal tissues in an ulcerative colitis model using using imaging with a folate-targeted dendrimer conjugate conjugated to Cy5.5 dye, with a non-targeted dendrimer conjugate conjugated to Cy5.5 dye, and in competition with a folate-targeted dendrimer conjugate which lacks a conjugated dye.
  • Folate-PEG-NH 2 is synthesized using 500 mg of NH 2 PEG 3200 NH 2 (PEG-bis amine) with an equimolar quantity of folic acid in 5 mL DMSO containing 1 molar equivalent of DCC or EDC and 10 ⁇ l pyridine, triethylamine or DIPEA. The reaction mixture is stirred overnight in the dark at room temperature. The reaction is quenched with water. The by-product urea and the trace amount of unreacted folate and PEG-bis amine are removed by both dialysis and by using a G-15 size exclusion column in deionized water. Folate-PEG-NH 2 is analyzed using HPLC with monitoring by absorbance at 363 nm.
  • N-succinyl-DSPE is synthesized by reacting overnight 1.1 molar equivalent of succinic anhydride with 100 mg DSPE in 5 mL of dry chloroform or dichloromethane containing 10 ⁇ L pyridine, triethylamine or DIPEA.
  • the product N-succinyl-DSPE runs as a spot with a higher Rf value ( ⁇ 0.5) than DSPE and is ninhydrin negative on TLC, using chloroform/methanol (3:2) as the solvent phase.
  • the structure of the product N-succinyl-DSPE is confirmed by MS.
  • the synthesis of the folate-PEG-DSPE construct is illustrated above. Under nitrogen, N-succinyl-DSPE is dissolved in chloroform and its carboxyl-group is activated by reacting with 0.95 molar equivalent of DCC or EDC. Then an equimolar amount of folate-PEG-DSPE is also dissolved in the above mentioned reaction mixture and the reaction is continued overnight at room temperature. The solvent is then removed under reduced pressure. The unreacted products are then removed from the mixture by membrane dialysis. The product folate-PEG-DSPE is analyzed on a HPLC system chromatography (water/methanol (1.5:1)) using a C-18 reverse phase column.
  • the liposomal suspension is transferred to an extruder and extruded under nitrogen pressure, 10 times each through 200 nm, 100 nm, and 50 nm pore filters, respectively.
  • the mean particle size of the liposome as determined by light scattering is 60-70 nm.
  • a total mass of 100 mg of the lipid mixture; [DSPC/Chol/mPEG2000-DSPE/folate-PEG-DSPE (56:40:4:0.1) mol %] is dissolved in chloroform and dried to a thin film in a round bottom flask under reduced pressure overnight.
  • the lipid mixture is rehydrated in 10 mM calcein dissolved in PBS (pH 7.4).
  • the resulting lipid suspension is subjected to 10-15 cycles of freezing and thawing.
  • nm and100 nm folate-PEG-liposomes of different size distributions are generated by extruding the lipid suspension 10 times each through polycarbonate membranes having pore sizes of sequentially 800 nm, 400 nm, 200 nm and 100 nm, followed by 50 nm, if needed, respectively. Unentrapped dye is separated using membrane dialysis. Particle size is determined by Dynamic Light Scattering-DynaPro99. Phosphate assay (Steward assay) is performed to determined phospholipid concentration.
  • the mixture is subjected to freeze-thaw-vortex cycle 10 times.
  • the liposomal suspension is transferred to an extruder and extruded under nitrogen pressure, 10 times each through 200 nm, 100 nm, and 50 nm pore filters, respectively.
  • the mean particle size of the liposome as determined by light scattering is about 60 nm.
  • Lipid mixture consisting of DSPC/Chol/mPEG 2000 DSPE/FolPEG 3200 DSPE in a 56:40:4:0.1 molar ratio was dissolved in ethanol at 70° C. The ethanol solution was then hydrated with iobitridol (350 mg I/mL) for 2 h. The suspension of liposomes obtained was submitted to a filtration through 400 nm, 200 nm, 100 nm, and 50 nm (10 cycles each) polycarbonate membranes using an extruder. Liposomes were then dialyzed overnight in a 300K Mw cutoff dialysis bag against PBS solution.
  • Folate-targeted liposomes encapsulating glutathione were prepared using a procedure based on polycarbonate membrane extrusion. Briefy, unilamellar vesicles were prepared from DSPC/Chol/mPEG 2000 DSPE/FolPEG 3200 DSPE (56:40:4:0.1) by thin film hydration method. Lipids at indicated ratios were dissolved in chloroform, and the solvent was evaporated under vacuum with a stream of N 2 gas to remove all organic solvent. The resultant film was then hydrated with 50 mM reduced glutathione in PBS (pH 7.4) at 60° C. with constant stirring.
  • the suspension of liposomes obtained was submitted to a filtration through 400 nm, 200 nm, 100 nm, and 50 nm (10 cycles each) polycarbonate membranes using an extruder. Untrapped residual reduced glutathione was removed by passing the liposome suspension through a Sephadex G50 column.
  • the commercially available kit of HMPAO, hexamethylpropylene amine oxime (Ceretec®, UK) was labeled with Tc-99m.
  • the kit was reconstituted with 15 mCi sodium pertechnetate in 0.9% NaCl solution at room temperature for 8 minutes.
  • the 99mTc-HMPAO complex was then mixed with the preformed liposome (part1) and incubated at 37° C. with intermittent vortexing for 30 minutes. Uncapsulated 99mTc-HMPAO was removed by gel filtration on a Sephadex G50 column using PBS as an eluent.
  • liposomal compositions are prepared as disclosed for the preparation of the corresponding folate-targeted liposomal compositions, except the folate-PEG-DSPE conjugate is omitted.
  • Folate-PEG-COOH is synthesized using 500 mg of NH 2 PEGCOOH (Mw′3400) with equimolar quantity of folic acid in 5 mL DMSO containing 1 molar equivalent of dicyclohexylcarbodiimide (DCC) or 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride (EDC) and 10 ⁇ L pyridine, triethylamine or diisopropylethylamine (DIPEA).
  • DCC dicyclohexylcarbodiimide
  • EDC 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride
  • DIPEA diisopropylethylamine
  • FolatePEG-Dendrimer is synthesized by reacting overnight 6 molar equivalent of FolNHPEGCOOH and EDC (6 eq) with Dendrimer (1 eq) in 5 mL of dry DMSO containing 10 ⁇ L pyridine, triethylamine or DIPEA. The trace amount of unreacted materials is also removed by both dialysis and G-15 size exclusion column in dH 2 O. Folate-PEG-Dendrimer is analyzed using UV/Vis and IR.
  • the step of coupling with a dye is omitted to provide folate-dendrimer conjugates.
  • Betamethasone-Entrapped Folate-Targeted Liposomal Composition Treatment with a Betamethasone-Entrapped Folate-Targeted Liposomal Composition in a 3-Week Model of Atherosclerosis in the Apo E Knock-Out Mouse
  • the Fol Liposome Treated group was injected i.v., twice per week, with a betamethasone-entrapped folate-targeted liposomal composition.
  • the NT Liposome group was injected similarly with a non-targeted betamethasone-entrapped liposomal composition.
  • the Untreated group was similarly injected with a folate-targeted liposomal composition with only encapsulated phosphate buffer solution.
  • Betamethasone-Entrapped Folate-Targeted Liposomal Composition Treatment with a Betamethasone-Entrapped Folate-Targeted Liposomal Composition in a 4-Week Model of Atherosclerosis in the Apo E Knock-Out Mouse
  • the Fol Liposome group was injected i.v. twice per week with a folate-targeted betamethasone-entrapped liposomal composition.
  • the NT Liposome group was similarly injected with a non-targeted betamethasone-entrapped liposomal composition.
  • the Untreated group was similarly injected with a folate-targeted liposomal composition with only encapsulated phosphate buffer solution.
  • FIG. 10 shows the relative EC20 uptake of the hearts and the aortas of the groups;
  • FIG. 10 (bottom) shows the relative biodistribution of EC20 uptake in the organs of the groups.
  • Thioglycolate recruited macrophages were isolated by peritoneal lavage 3 days after intraperitoneal injection of 3.0 mL 3% sterile medium.
  • the Lewis rats were sacrificed by CO 2 asphyxiation, and the peritoneal macrophages were harvested by injecting 60 mL of PBS into the peritoneal cavity. After centrifugal separation of the cells collected in the PBS, the cells were resuspended into Eppendorf tubes with FDRPMI 1640 medium. The medium was replaced with fresh medium containing Folate-targeted liposome encapsulating dye (DiD or calcein). For competition studies, excess free folic acid is preintroduced to the peritoneal medium for 30 min before addition of folate targeted liposome. For control, empty folate targeted liposome (PBS) were used. All media were incubated for 2 h at 37° C. . The cells were washed 3 times with PBS and resuspended in fresh PBS. The cell bound fluorescence was analyzed using flow cytometry.
  • Folate-targeted liposome encapsulating dye DiD or calcein
  • FIG. 14 are shown the results of the flow cytometry for control and calcein-entrapped folate-targeted liposomal compositions in the presence and absence of excess free folic acid.
  • One set of Apo E KO mice (Normal Chow) was fed a normal diet and one set of Apo E KO mice (Western Diet) was fed a high fat western diet for two weeks. Each mouse then received a folate-targeted liposome composition (2 mg total phospholipid, i.v) containing 10 ⁇ Ci 3 H cholesterol-oleoyl-ether. The animals were sacrificed 24 h later, and organs were removed. Tissue samples were solubilized using Soluene 350 (1 mL/100 mg tissue) at 60° C. for 48 h. Tissue solutions were then bleached to uniform color using 30% hydrogen peroxide, followed by 6 mL Hionic-Fluor scintillation cocktail.
  • FIG. 1 The relative biodistribution of uptake of the labeled liposomes in the aorta and hearts of Normal Chow and Western Diet mice is shown in FIG. 1 (top); the biodistribution in a number of organs is shown in FIG. 1 (bottom).
  • DiD is the imaging agent
  • approximately 50 nm DiD-entrapped liposomes of DSPC/Chol/mPEG 2000 DSPE/FolPEG 3400 DSPE (56:40:4:0.1) were used, along with the corresponding non-targeted liposomes.
  • excess 0.1 mM folic acid was used.
  • Administration was i.p.
  • Apo E knockout mice are fed with western diet for 1 week. After 7 days, mice received the DiD-entrapped folate-targeted liposomal composition (fol-PEG-liposome-DiD) (0.2 ⁇ mol lipid) i.p.(total volume injected: 100 ⁇ l) in each mouse. For competition studies, mice were pre-administered free folic acid (50 ⁇ l) before i.p injection of DiD liposome. Images were taken 2 h postinjection. The images are shown in FIG. 2 and FIG. 3 .
  • Apo E knockout mice are fed with western diet for 4 weeks. DiD-entrapped liposomes were administered. The animals were imaged after 4 hours. The images are shown in FIG. 4 .
  • An Apo E KO mouse (7 weeks old) was subjected to i.p. injection of the iobitridol-entrapped folate-targeted liposomal composition described above (3.0 ⁇ 10 ⁇ 6 mol total lipids).
  • mice Three C57 mice were fed with a 2% dextran sodium sulfate for 7 days. After 7 days, DiD-entrapped liposomes were administered. Intestines were removed and imaged after 4 h. The images are shown in FIG. 11 .
  • Adjuvant-induced arthritis in Lewis rats was induced by injection of butyricum (0.15 mg) into a hind paw. After 17 days, DiD-entrapped liposomes were administered. The hind paws of the rats were imaged. The images are shown in FIG. 12 .
  • Adjuvant-induced arthritis in Lewis rats was induced by injection of heat killed mycoplasmia butyricum (0.5 mg), suspended in mineral oil (5 mg/mL), on day 1 into the left hind foot of Lewis rats. Disease was allowed to progress for 17 days. 100% of animals developed arthritis as evidenced by gross swelling in the injected paw and progressive swelling in all extremities. DiD-entrapped liposomes were administered (i.v.). The hind paws of the rats were imaged after 12 hours. The images are shown in FIG. 15 .
  • mice Four mice were injected with 100 ⁇ L of a 10 ⁇ M solution of a cardiotoxin (from Naja mossambica mossambica), and two mice were used as controls. Four days later, the two healthy mice were injected with folate-targeted DiD-entrapped liposomes, as were two mice with cardiotixin-induced muscle injury (chosen randomly). The two remaining cardiotoxin-injected mice (chosen randomly) were treated with non-targeted DiD-entrapped liposome. 300 ⁇ g of phospholipid were injected per mouse. The dye used was 1% DiD. Images were taken eight hours after a tail vein injection. The images of three of the mice are shown in FIG. 13 .
  • mice One million M109 cells were injected subcutaneously into a shoulder of Balb-C mice. On day 14 post tumor cell inoculation, the mice were injected in the opposite thigh with 100 ⁇ L of a 10 ⁇ M solution of a cardiotoxin (from Naja mossambica mossambica). Four days later, on day 18, mice were treated i.v.
  • mice were imaged 8 hours later. The images of the mice are shown in FIG. 17 .
  • mice One million M109 cells were injected subcutaneously into a shoulder of Balb-C mice. On day 11 post tumor cell inoculation, the mice were administered 2% dextran sodium sulfate (DSS) for 5 days. The mice were then treated i.v. with a non-targeted DiD-entrapped liposomal composition (NT Liposome), with a folate-targeted DiD-entrapped liposomal composition (Fol-Liposome), and with empty folate-targeted liposome (injected empty folate-targeted liposome first, then dosed with a folate-targeted DiD-entrapped liposomal composition) (Folate Liposome W Competion). The mice were imaged 8 hours later. The images of the mice are shown in FIG. 18 .
  • NT Liposome non-targeted DiD-entrapped liposomal composition
  • Fol-Liposome folate-targeted DiD-entrapped liposomal
  • Raw cells are plated in each well and treated with 10 ⁇ L of the indicated dendrimers (1.0 mg/mL). After 1 hour of incubation, confocal imaging is carried out. The images are shown in FIG. 21 .
  • Apo E knockout mice are fed with western diet are treated with the indicated dendrimer conjugates and imaged 4 hours post injection. The images are shown in FIG. 22 . The images of the excised aortas are shown in FIG. 23 .
  • mice were fed with a 2% dextran sodium sulfate, and the indicated dendrimer conjugates were administered.
  • the mice were imaged after 6 h. The images are shown in FIG. 24 .

Abstract

The invention described herein pertains to folate-receptor targeted agents comprising therapeutic agents useful for the treatment of inflammatory disease, including folate-receptor targeted liposomes (folate-targeted liposomes) containing entrapped therapeutic agents and folate-receptor targeted dendrimers conjugated to therapeutic agents (folate-targeted dendrimer conjugates), useful for the treatment of inflammatory disease, including auto-immune disease, as well as to folate-targeted liposomes containing entrapped imaging agents and dendrimer conjugates conjugated to imaging agents, for use in the diagnosis and monitoring of treatment in such disease.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims the benefit of U.S. provisional application 61/349,434, filed 28 May 2010, which is incorporated by reference herein.
  • TECHNICAL FIELD
  • The invention described herein pertains to folate-receptor targeted agents comprising therapeutic agents useful for the treatment of inflammatory disease, including folate-receptor targeted liposomes (folate-targeted liposomes) containing entrapped therapeutic agents and folate-receptor targeted dendrimers conjugated to therapeutic agents (folate-targeted dendrimer conjugates), useful for the treatment of inflammatory disease, including auto-immune disease, as well as to folate-targeted liposomes containing entrapped imaging agents and dendrimer conjugates conjugated to imaging agents, for use in the diagnosis and monitoring of treatment in such disease.
  • BACKGROUND AND SUMMARY OF THE INVENTION
  • Liposomes are closed, spherical vesicles comprising amphiphilic lipids such as phospholipids, sphingolipids, and/or other lipids, including sterols such as cholesterol and cholesterol ester salts, in proportions such that they arrange themselves into multiple concentric bilayers when hydrated in aqueous solutions. Using any of a number of methods, such liposomes can be converted into single bilayer liposomes. These single bilayer liposomes are useful carriers both of hydrophilic (lipophobic) agents, which can reside entrapped in the aqueous interior of the liposome, and of hydrophobic (lipophilic) agents, which can reside entrapped in the lipid bilayer. The utility of liposomes as carriers for therapeutic agents has been recognized and has lead to the development of long-circulating liposomes. By altering the composition of the lipid bilayer in such liposomes by, for example, the inclusion of a surface coat of flexible biocompatible hydrophilic chains, the resulting liposomes have improved stability. The surface coat serves to protect the liposome from uptake by organs of the mononuclear phagocyte system, especially the liver, lung and spleen, and from the plasma components which are involved in liposome uptake. One example of the hydrophilic chain constituent of the surface coat is polyethylene glycol (PEG), in which the terminal hydroxy group may be capped with a methyl group to form a methyl ether group (denoted as mPEG or MPEG). (Unless specifically defined otherwise, in this specification PEG will include the hydroxy terminated form, the methyl ether terminated form, and any other similarly terminated form in the context of the surface coat of a liposome.)
  • Inflamed tissue regions in the body have been imaged using folate-targeted imaging agents. For example, the folate-receptor targeted, radionuclide conjugate imaging agent EC20 (folate-Tc99m), has been used to image rheumatoid arthritis sites in vivo.
  • In WO 94/07466 there is disclosed a PEG coated liposomal composition containing an entrapped therapeutic agent for concentrating therapeutics in an inflamed tissue region in the body. The beneficial effects of the liposomes of WO 94/07466 (EP-0662820) are questioned in WO 03/105805, which discloses PEG-coated liposomes composed of non-charged vesicle-forming lipids, optionally containing not more than 5 mol percent of charged vesicle-forming lipids, and containing a water soluble corticosteroid for the site-specific treatment of inflammatory disorders.
  • In a folate receptor positive mouse model of ovarian cancer, folate-targeted liposomes carrying the entrapped fluorescent dye calcein have been shown to unload the dye in ovarian cancer cells and tumor-asssociated macrophages within tumor ascites fluid associated with an intraperitoneal cancer both ex vivo and after in vivo administration and collection of the ascites fluid for analysis. (M. J. Turk et al., Cancer Letters 213 (2004) 165-172.) The same reference discloses folate targeted liposomes in which the lipid phase includes tritiated cholesterol-oleoyl ether.
  • Disclosed herein are folate-targeted liposomal compositions comprising an anti-inflammatory agent as an entrapped agent, pharmaceutical compositions comprising such liposomal compositions, and the use of such compositions in the treatment of an inflammatory disease, such as in an inflamed tissue region in the body. Entrapped agents can be encapsulated within the aqueous interior of the liposomes and/or dissolved into the hydrocarbon regions of their bilayers. Also disclosed are folate-targeted liposomal compositions comprising an imaging or visualizing agent as an entrapped agent, as well as the use of such liposomal compositions for diagnosis or monitoring the treatment of an inflammatory disease in an inflamed tissue region in the body.
  • Further disclosed herein are dendrimers conjugated to active agents (folate-targeted dendrimer conjugates), such as anti-inflammatory agents, pharmaceutical compositions comprising such dendrimer conjugates, and the use of such compositions in the treatment of disease such as inflammatory disease. Dendrimers are known classes of compounds which are generally regarded as repeatedly branched, approximately spherical molecules and are sometimes referred to as arborols or cascade molecules. Also, disclosed are folate-targeted dendrimer conjugate compositions comprising an imaging or visualizing agent, as well as the use of such compositions for diagnosis or monitoring the treatment of a disease such as inflammatory disease in an inflamed tissue region in the body.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 (top) shows the relative biodistribution of uptake of 3H-cholesterol labeled folate-targeted liposomes in the aortas (1) and hearts (2) of Normal Chow (N) and Western Diet (W) mice; the biodistribution in a number of organs (aorta (1), heart (2), liver (3), kidney (4), spleen (5), intestine (6), muscle (7), skin (8) and lung (9)) is shown in FIG. 1 (bottom).
  • FIG. 2 shows whole body imaging of the atherosclerotic sites in Apo E KO mice fed a western diet for one week using a folate-targeted liposomal composition, with the entrapped fluorescent dye DiD, preinjection (top) and 2 hours post injection (bottom) in the absence (left) and presence (right) of pre-administered folic acid. The scale shows low (bottom) to high (top) intensity of fluorescence.
  • FIG. 3 shows the imaging of the atherosclerotic sites in the aortic arch in Apo E KO mice fed a western diet for one week using a folate-targeted liposomal composition, with the entrapped fluorescent dye DiD. Atherosclerotic plaques showed a 5-fold increase in mean fluorescence.
  • FIG. 4 shows whole body imaging of the atherosclerotic sites in Apo E KO mice fed a western diet for four weeks before imaging under four conditions: (A) control with no imaging agent, (B) DiD-entrapped folate-targeted liposomal composition, (C) non-targeted DiD-entrapped liposomal composition, and (D) DiD-entrapped folate-targeted liposomal composition administered with an excess of folic acid.
  • FIG. 5 shows radiographic (X-ray) imaging at preinjection (0) and 2, 4, 24 and 48 hours post injection of an Apo E KO mouse dosed with an iobitridol-entrapped folate-targeted liposomal composition.
  • FIG. 6 shows radioisotopic imaging in Apo E KO mice fed a western diet for four weeks before dosing using a 99Tc-entrapped folate-targeted liposomal composition, administered either alone (A) or with an excess of folic acid (B).
  • FIG. 7 shows radioisotopic imaging of the whole animal (top) and excised hearts (bottom) using the folate-receptor targeted, radionuclide conjugate imaging agent EC20 (folate-Tc99m) in Apo E KO mice fed a western diet for three weeks and dosed as (A) a control (Untreated) group which received a folate-targeted liposomal composition which contained only phospate buffered saline as the entrapped agent, (B) a group (NT Liposome) which received a non-targeted liposomal composition which contained betamethasone as the entrapped agent, and (C) a group (Fol-Liposome) which received a folate-targeted liposomal composition which contained betamethasone as the entrapped agent.
  • FIG. 8 shows the relative EC20 heart uptake of the groups of FIG. 7.
  • FIG. 9 shows the biodistribution of EC20 uptake in the organs (heart (1), lung (2), liver (3), spleen (4), kidney (5), muscle (6), skin (7) and intestine (8) of the groups of FIG. 7.
  • FIG. 10 shows radioisotopic imaging (top) of the hearts (1) and aortas (8) and the biodistribution (bottom) of the organs (heart (1), lung (2), liver (3), intestine (4), kidney (5), spleen (6), muscle (7), aorta (8) and skin(9)) using the folate-receptor targeted, radionuclide conjugate imaging agent EC20 (folate-Tc99m) in Apo E KO mice fed a western diet for 4 weeks and dosed as (A) a control (Untreated) group which received a folate-targeted liposomal composition which contained only phospate buffered saline as the entrapped agent, (B) a group (NT Liposome) which received a non-targeted liposomal composition which contained betamethasone as the entrapped agent, and (C) a group (Fol-Liposome) which received a folate-targeted liposomal composition which contained betamethasone as the entrapped agent.
  • FIG. 11 shows the imaging of the intestines of mice in an intestinal inflammation model using imaging with (A) a DiD-entrapped folate-targeted liposomal composition, (B) a DiD-entrapped folate-targeted liposomal composition administered with an excess of folic acid, and (C) a non-targeted DiD-entrapped liposomal composition.
  • FIG. 12 shows the imaging of the paws of rats in an arthritis model using imaging with (A) a DiD-entrapped folate-targeted liposomal composition, (B) a DiD-entrapped folate-targeted liposomal composition administered with an excess of folic acid, and (C) a non-targeted DiD-entrapped liposomal composition.
  • FIG. 13 shows the imaging of mice in an inflammatory muscle injury model showing a healthy mouse imaged with a folate-targeted DiD-entrapped liposomal composition (Healthy), a cardiotoxin treated mouse imaged with a non-targeted DiD-entrapped liposomal composition (NT Liposome), and a cardiotoxin treated mouse imaged with a folate-targeted DiD-entrapped liposomal composition (Fol Liposome). The scale shows low (left) to high (right) intensity of uptake of the label.
  • FIG. 14 shows the cell bound fluorescence measured by flow cytometry in thioglycolate recruited macrophages for control non-targeted calcein-entrapped liposomal compositions (shaded curve), and for folate-targeted calcein-entrapped liposomal compositions in the presence (middle curve) and absence (right curve) of excess free folic acid.
  • FIG. 15 shows the imaging of the paws of rats in an arthritis model showing a diseased rat imaged with a non-targeted DiD-entrapped liposomal composition (NT Liposome), a diseased rat imaged with a folate-targeted DiD-entrapped liposomal composition (Fol-Liposome), and a healthy rat with a folate-targeted DiD-entrapped liposomal composition (Healthy).
  • FIG. 16 shows the imaging in mice implanted with (M109) tumors which express the folate receptor at high levels on the surface and concomitantly used in the muscle injury model or the ulcerative colitis model when imaged with a folate-targeted DiD-entrapped liposomal composition (Fol-Liposome).
  • FIG. 17 shows the imaging in mice implanted with (M109) tumors which express the folate receptor at high levels on the surface and concomitantly used in the muscle injury model when imaged with a non-targeted DiD-entrapped liposomal composition (NT Liposome), with a folate-targeted DiD-entrapped liposomal composition (Fol-Liposome), and with empty folate-targeted liposome (injected empty folate-targeted liposome first, then dosed with a folate-targeted DiD-entrapped liposomal composition) (Competition with Folate Liposome).
  • FIG. 18 shows the imaging in mice implanted with (M109) tumors which express the folate receptor at high levels on the surface and concomitantly used in an intestinal inflammation model when imaged with a non-targeted DiD-entrapped liposomal composition (NT Liposome), with a folate-targeted DiD-entrapped liposomal composition (Fol-Liposome), and with empty folate-targeted liposome (injected empty folate-targeted liposome first, then dosed with a folate-targeted DiD-entrapped liposomal composition) (Fol-Liposome with Competition).
  • FIG. 19 shows schematically the types of dendrimer conjugates used in the imaging studies.
  • FIG. 20 shows a synthetic scheme for the preparation of a folate-targeted dendrimer conjugate conjugated to a dye. Folate-targeted dendrimer conjugates conjugated to active agents, such as anti-inflammatory agents, may be prepared similarly.
  • FIG. 21 shows imaging of cells which express the folate receptor at high levels on the surface (control) and in the presence of a folate-targeted dendrimer conjugate conjugated to FITC dye (FolDendFITC) alone and in the presence of excess folic acid (FolDendFITC w Excess Folic Acid).
  • FIG. 22 shows whole body imaging of the atherosclerotic sites in Apo E KO mice fed a western diet with a folate-targeted dendrimer conjugate conjugated to Cy5.5 dye (FolDend(G3)Cy5.5), with a non-targeted dendrimer conjugate conjugated to Cy5.5 dye, and in competition with a folate-targeted dendrimer conjugate which lacks a conjugated dye (FolDend(G3)Cy5.5 W Competion).
  • FIG. 23 shows the imaging of the atherosclerotic sites in the aortic arch in Apo E KO mice fed a western diet with a folate-targeted dendrimer conjugate conjugated to Cy5.5 dye (FolDend(G3)Cy5.5), with a non-targeted dendrimer conjugate conjugated to Cy5.5 dye, and in competition with a folate-targeted dendrimer conjugate which lacks a conjugated dye (FolDend(G3)Cy5.5 W Competion).
  • FIG. 24 shows the imaging of (the intestines of) mice in an intestinal inflammation model using imaging with a folate-targeted dendrimer conjugate conjugated to Cy5.5 dye (FolDend(G3)Cy5.5), with a non-targeted dendrimer conjugate conjugated to Cy5.5 dye, and in competition with a folate-targeted dendrimer conjugate which lacks a conjugated dye (FolDend(G3)Cy5.5 W Competion).
  • DETAILED DESCRIPTION
  • In one embodiment, there is provided a folate-receptor targeted agent comprising a therapeutic agent useful for the treatment of inflammatory disease. In one embodiment, the folate-receptor targeted agent is a folate-targeted liposomal composition comprising an anti-inflammatory agent as an entrapped agent. In one embodiment, the folate-receptor targeted agent is a folate-targeted dendrimer conjugate comprising a folate-targeted dendrimer conjugated to an anti-inflammatory agent.
  • In one embodiment, there is provided a folate-targeted liposomal composition comprising an anti-inflammatory agent as an entrapped agent. In general, the embodiment of the liposome may be as described herein or of any conventional composition and may be constructed as described herein or by any conventional methodology with the folate-targeting ligand attached as described herein or by any conventional linkage structure or methodology.
  • The lipid components used in forming the embodiments of the liposomes may be any of the variety of vesicle-forming lipids, including phospholipids, sphingolipids, and/or other lipids, including sterols such as cholesterol and cholesterol ester salts. Suitable lipids for the embodiments include those having two hydrocarbon chains, typically acyl chains, and a polar head group, such as phospholipids and glycolipids. As used herein, unless otherwise defined or clear from the context, phospholipid includes any one phospholipid or combination of phospholipids capable of forming liposomes. Phospholipids include phosphatidylcholines (PC), phosphatidylethanolamine (PE), phosphatidic acid (PA), phosphatidylinositol (PI), and sphingomyelin (SM), where the two hydrocarbon chains are typically between about 14-22 carbons in length, and have varying degrees of unsaturation. The glycolipids include cerebrosides and gangliosides. Phosphatidylcholines, including those obtained from natural sources or those that are partially or wholly synthetic, or of variable chain length and unsaturation, are suitable. Phospholipids which contain saturated alkyl chains, yielding a relatively high transition temperature, are suitable for the embodiments. These include, for example, distearoylphosphatidylethanolamine (DSPE), distearoylphosphatidylcholine (DSPC) and hydrogenated soy phosphatidylcholine (HSPC). One embodiment of the liposomal composition disclosed herein is one wherein the lipid bilayer is primarily composed of DSPC/cholesterol with a mol ratio of about 56:40.
  • The folate-targeting components used in forming the embodiments of the liposomes may be any of those known in the preparation of folate-targeted liposomes or those described herein. For examples of the preparation of folate-targeted liposomal compositions, see, for example, R. J. Lee et al., J. Biological Chemistry, 269(5) (1994) 3198-3204; and R. J. Lee et al., Biochimica et Biophysica Acta, 1233 (1995) 134-144.
  • One embodiment is a liposomal composition as described herein wherein the lipid bilayer comprises a comprises a folate targeting conjugate composed of (a) a lipid having a polar head group and a hydrophobic tail, (b) a hydrophilic polymer having a first end and a second end, said polymer attached at its first end to the head group of the lipid, and (c) a folate ligand (Fol) attached to the second end of the polymer, and wherein the folate ligand is a folic acid residue or an analog or derivative thereof. Folic acid analogs or derivatives thereof include the (unnatural) D-isomer of folic acid and derivatives other related folic acid receptor binding molecules, such as folinic acid, pteroic acid, pteropolyglutamic acid, receptor-binding pteridines such as tetrahydropterins, dihydrofolates, tetrahydrofolates, and their deaza and dideaza analogs. The terms “deaza” and “dideaza” analogs refer to the art-recognized folate analogs having a carbon atom substituted for one or two nitrogen atoms in the naturally occurring folic acid structure. Other folate analogs or derivatives include the folate receptor-binding analogs aminopterin, amethopterin (methotrexate), and derivatives thereof.
  • For example, in one embodiment, the hydrophilic polymer of the folate targeting conjugate is a PEG having an average molecular weight of about 200-5000 in which the end groups, prior to attachment, are independently amino, hydroxy, thiol or carboxy. In one embodiment the hydrophilic polymer of the folate targeting conjugate is a PEG having an average molecular weight of about 3200-3400 in which the end groups, prior to attachment, are independently amino or thiol. In one embodiment the hydrophilic polymer of the folate targeting conjugate is a PEG having an average molecular weight of about 3200-3400 in which the end groups, prior to attachment, are each amino, such as for example, NH2PEG3200NH2 or NH2PEG3400NH2, which also may be denoted as a polyoxyethylene bis-amine or a PEG bis-amine, such as for example PEG bis-amine, Mr˜3350. In another embodiment, the hydrophilic polymer of the folate targeting conjugate is a PEG having an average molecular weight of about 3200-3400 in which the end groups, prior to attachment, are amino and thiol, which also may be denoted amino-PEG-SH. An amino-PEG-SH may be obtained from the corresponding PEG bis-amine by a conventional method, such as by the addition of Traut's reagent.
  • In one embodiment, the folate-targeted liposomal composition, the lipid having a polar head group and a hydrophobic tail, is a phosphatidyl group as defined herein. In one embodiment, the phosphatidyl group comprises two saturated acyl groups each of which is between about 14-22 carbons in length. In one embodiment, the phosphatidyl group comprises two saturated acyl groups each of which is between about 16-18 carbons in length. In one embodiment, the phosphatidyl group is a distearoyl (DS) derivative.
  • In the folate targeting conjugate the polar head group of the lipid is attached to the first end of the hydrophilic polymer by any conventional method. In one embodiment, the polar head group is part of a phosphatidylethanolamine, wherein the phosphatidyl group has any of the meanings herein, and the amino group of the ethanolamine portion is linked to an amino group of the first end of the hydrophilic polymer by a bis-acyl group, such as a succinyl group. A succinyl group also may be denoted as a 1,4-dioxobutane-1,4-diyl group. In another embodiment, the polar head group is part of a phosphatidylethanolamine, wherein the phosphatidyl group has any of the meanings herein, and the amino group of the ethanolamine portion is linked to a thiol group of the first end of the hydrophilic polymer via addition of the thiol group to an N-maleiimidocaproylphospatidylethanolamine. In one embodiment of any of the above, the phosphatidylethanolamine is distearoylphosphatidylethanolamine (DSPE).
  • In one embodiment of the folate-targeted liposomal composition, the folate ligand is a folic acid residue which is attached to an amino group at the second end of the hydrophilic polymer by acylation with the a-carbonyl group or the γ-carbonyl group of the glutamic acid portion of folic acid. In another embodiment of the folate-targeted liposomal composition, the folate ligand is a folic acid residue which is attached to an amino group at the second end of the hydrophilic polymer by acylation with the γ-carbonyl group of the glutamic acid portion of folic acid.
  • In one embodiment of the folate-targeted liposomal composition, the folate targeting conjugate comprises distearoylphosphatidylethanolamine linked to the first end of a bis-amino PEG and a folic acid residue which is attached to the amino group at the second end of the bis-amino PEG by acylation with the γ-carbonyl group of the glutamic acid portion of folic acid. In a further embodiment, the distearoylphosphatidylethanolamine is linked to the first end of the bis-amino PEG by a succinyl group.
  • A further embodiment of the above liposomal composition is one wherein the folate targeting conjugate comprises distearoylphosphatidylethanolamine wherein the amino group of the ethanolamine portion is linked by a succinyl group to the amino group of the first end of a PEG having an average molecular weight of about 3200-3400 in which the end groups, prior to attachment, are each amino, and wherein the folate ligand is a folic acid residue which is attached to the amino group at the second end of the PEG by acylation with the γ-carbonyl group of the glutamic acid portion of folic acid.
  • In one embodiment of the folate-targeted liposomal composition, as described herein, the folate targeting conjugate is present at about 0.01 mol % to about 1 mol % in the lipid bilayer. In another embodiment, the folate targeting conjugate is present at about 0.05 mol % to about 0.5 mol % in the lipid bilayer. In another embodiment, the folate targeting conjugate is present at about 0.1 mol % in the lipid bilayer.
  • One embodiment is a liposomal composition as described herein wherein the lipid bilayer further comprises a hydrophilic coating composed of (a) a lipid having a polar head group and a hydrophobic tail and (b) a hydrophilic polymer having a first end and a second end, said polymer attached at its first end to the head group of the lipid and optionally capped at its second end. The hydrophilic coating components used in forming the embodiments of the liposomes may be any of those known in the preparation of hydrophilic coated liposomes or those described herein.
  • In general, the lipid of the hydrophilic coating having a polar head group and a hydrophobic tail can be any of those disclosed herein for the preparation of a folate targeting conjugate. Thus, in one embodiment, the lipid having a polar head group and a hydrophobic tail is a phosphatidyl group as defined herein. In one embodiment, the phosphatidyl group comprises two saturated acyl groups each of which is between about 14-22 carbons in length. In one embodiment, the phosphatidyl group comprises two saturated acyl groups each of which is between about 16-18 carbons in length. In one embodiment, the phosphatidyl group is a distearoyl (DS) derivative.
  • The hydrophilic polymer of the hydrophilic coating can be any of a number of biocompatible hydrophilic polymers. In one embodiment, the hydrophilic polymer is a polyethylene glycol (PEG), a polylactic acid (PLA), a polyglycolic acid (PGA) or a polyvinyl alcohol (PVA), as well as copolymers of lactic and glycolic acids, in which the first end group, prior to attachment, is amino, hydroxy or thiol. The hydrophilic polymer is optionally capped with methyl, ethyl, or another unreactive group. In another embodiment, the hydrophilic polymer is a polyethylene glycol with an average molecular weight of about 2000, which is uncapped or capped with a methyl or ethyl group.
  • The polar head group of the lipid is attached to the first end of the hydrophilic polymer of the hydrophilic coating by any conventional method. In one embodiment, the polar head group is part of a phosphatidylethanolamine, wherein the phosphatidyl group has any of the meanings herein, and the amino group of the ethanolamine portion is linked directly to a carbon of the first end of the hydrophilic polymer, or via a linker to an amino, hydroxy or thiol group of the first end of the hydrophilic polymer by a linking group. The amino group of the ethanolamine portion may be linked directly to a carbon of the first end of the hydrophilic polymer, for example by converting a hydroxy group to a leaving group, such as a triflate, and alkylating the amino group of the ethanolamine. The amino group of the ethanolamine portion of a phosphatidylethanolamine may be linked to a hydroxy group of the first end of the hydrophilic polymer, for example, by a linking group such as a carbonyl group or a 4-chloro-1,3,5-triazinyl group; or the amino group of the ethanolamine portion may be linked to an amino group of the first end of the hydrophilic polymer, for example, by a linking group such as a carbonyl group, a succinyl group or a 4-chloro-1,3,5-triazinyl group. The amino group of the ethanolamine portion of a phosphatidylethanolamine may be linked to a thiol group of the first end of the hydrophilic polymer by formation of a disulfide bond between an alkyl thiol group on the amino group of the ethanolamine and the thiol group of the hydrophilic polymer. For the preparation of a hydrophilic coating in which the hydrophilic polymer is not capped, the group at the second end may be protected using a conventional protecting group while the first end is attached to the polar head group of the lipid of the hydrophilic coating having a polar head group and a hydrophobic tail; then the protecting group is removed. For example, a hydroxy group at the second end of the hydrophilic polymer may be protected using a trimethylsilyl group, which is removed following the attachment of the first end of the hydrophilic polymer to the polar head group. One embodiment is a liposomal composition as described herein wherein the lipid bilayer further comprises a hydrophilic coating comprising distearoylphosphatidyl-ethanolamine and polyethylenegycol 2000 (PEG2000)or methoxypolyethyleneglycol 200 (mPEG2000).
  • One embodiment is a liposomal composition as described herein wherein the hydrophilic coating is present at about 1 to 20 mol % in the lipid bilayer. Another embodiment is a liposomal composition as described herein wherein the hydrophilic coating is present at about 4 mol % in the lipid bilayer.
  • One embodiment is a liposomal composition as described herein wherein the liposome bilayer comprises a mol ratio of DSPC/cholesterol/mPEG2000-DSPE of about 56:40:4. Another embodiment is a liposomal composition as described herein wherein the liposome bilayer comprises a mol ratio of DSPC/Chol/mPEG2000-DSPE/Fol-PEG3400DSPE of about 56:40:4:0.1.
  • One embodiment is a liposomal composition as described herein wherein the average particle size of the liposome is about 30 to 200 nM. Another embodiment is a liposomal composition as described herein wherein the average particle size of the liposome is about 40 to 120 nM. A further embodiment is a liposomal composition as described herein wherein the average particle size of the liposome is about 50 to 100 nM.
  • A further embodiment of folate-targeted liposomal compositions, as described herein, is the ability to deliver more of the entrapped agent to an inflamed site than corresponding non-targeted liposomal compositions, which are the same as the folate-targeted liposomal compositions, except the folate targeting conjugate, such as folate-PEG-DSPE, is omitted.
  • One embodiment is a liposomal composition, or folate-targeted dendrimer conjugate, as described herein wherein the entrapped anti-inflammatory agent is an anti-inflammatory steroid. In one embodiment, the anti-inflammatory steroid is a sytemically administered (lipophilic) anti-inflammatory steroid. In one embodiment, the anti-inflammatory steroid is betamethasone, dexamethasone, flumethasone, methylprednisolone, paramethasone, prednisolone, prednisone, triamcinolone, hydrocortisone or cortisone. A further embodiment is a liposomal composition as described herein wherein the entrapped agent is betamethasone.
  • In another embodiment, the anti-inflammatory steroid is a topically administered anti-inflammatory steroid. In this sense, topically administered includes administration by any of a number of nonsystemic means, including by inhalation, suppository, topical cream ointment, foam, lotion or gel, etc. In one embodiment, the anti-inflammatory steroid is alcomethasone dipropionate, amcinonide, betamethasone dipropionate, betamethasone monopropionate, betamethasone 17-valerate, budesonide, budesonide disodium phosphate, ciclomethasone, clobetasol-17-propionate, clobetasone-17-butyrate, cortisone acetate, deprodone propionate, desonide, desoxymethasone, dexamethasone acetate, diflucortolone valerate, diflurasone diacetate, diflucortolone, difluprednate, flumetasone pivalate, flunisolide, fluocinolone acetonide acetate, fluocinonide, fluocortolone, fluocortolone caproate, fluocortolone hexanoate, fluocortolone pivalate, fluormetholone acetate, fluprednidene acetate, fluticasone propionate, halcinonide, halometasone, hydrocortisone acetate, hydrocortisone-17-butyrate, hydrocortisone-17-valerate, medrysone, methylprednisolone acetate, mometasone furoate, parametasone acetate, prednicarbate, prednisolone acetate, prednylidene, rimexolone, tixocortol pivalate, triamcinolone acetonide, triamcinolone alcohol or triamcinolone hexacetonide. In one embodiment, the anti-inflammatory steroid is budesonide, flunisolide or fluticasone propionate.
  • In another embodiment, the anti-inflammatory steroid is a water soluble anti-inflammatory steroid. Another embodiment is a liposomal composition as described herein wherein the anti-inflammatory steroid is betamethasone sodium phosphate, desonide sodium phosphate, dexamethasone sodium phosphate, hydrocortisone sodium phosphate, hydrocortisone sodium succinate, cortisone sodium phosphate, cortisone sodium succinate, methylprednisolone disodium phosphate, methylprednisolone sodium succinate, methylprednisone disodium phosphate, methylprednisone sodium succinate, prednisolone sodium phosphate, prednisolone sodium succinate, prednisone sodium phosphate, prednisone sodium succinate, prednisolamate hydrochloride, triamcinolone acetonide disodium phosphate or triamcinolone acetonide dipotassium phosphate. In one embodiment, the anti-inflammatory steroid is budesonide disodium phosphate.
  • One embodiment is a liposomal composition, or folate-targeted dendrimer conjugate, as described herein wherein the entrapped anti-inflammatory agent is a non-steroidal anti-inflammatory drug (NSAID), which also may be denoted as a non-steroidal anti-inflammatory agent (NSAIA) or as a non-steroidal anti-inflammatory medicine (NSAIM). In one embodiment, the NSAID comprises a propionic acid derivative such as, for example, ibuprofen, naproxen, fenoprofen, ketoprofen, flurbiprofen or oxaprozin. In one embodiment, the NSAID comprises an acetic acid derivative, such as, for example, indomethacin, sulindac, etodolac or diclofenac. In one embodiment, the NSAID comprises an oxicam derivative, such as, for example, piroxicam, meloxicam, tenoxicam, droxicam, lornoxicam or isoxicam. In one embodiment, the NSAID comprises a fenamic acid derivative, such as, for example, mefenamic acid, meclofenamic acid, flufenamic acid or tolfenamic acid. In one embodiment, the NSAID comprises a selective COX-2 (cyclooxygenase-2) inhibitor (coxib), such as, for example, celecoxib, rofecoxib, valdecoxib, parecoxib, lumiracoxib or etoricoxib.
  • Another embodiment is a liposomal composition, or folate-targeted dendrimer conjugate, as described herein wherein the entrapped anti-inflammatory agent comprises another drug useful in the treatment of rheumatoid arthritis or other autoimmune disease including an antiproliferative, immunomodulator or immunosuppresant agent, comprising, for example, aspirin, methotrexate, sulfasalazine, D-penicillamine, nambumetone, aurothioglucose, auranofin, other gold-containing compound, colloidal gold, cyclosporin, tacrolimus, pimecrolimus or sirolimus.
  • Because liposomes are useful carriers both of hydrophilic, lipophobic agents, which can reside entrapped in the aqueous interior of the liposome, and of hydrophobic, lipophilic agents, which can reside entrapped in the lipid bilayer, it is possible to load a lipsome as disclosed herein simultaneously with more than one anti-inflammatory agent, any of which may be hydrophilic or lipophilic. Similarly, it is possible to load a lipsome as disclosed herein simultaneously with an anti-inflammatory agent and a further therapeutic agent, any of which may be hydrophilic or lipophilic. Thus, one embodiment is a liposomal composition as described herein comprising more than one anti-inflammatory agent as an entrapped agent. Another embodiment is a liposomal composition as described herein wherein there are two entrapped anti-inflammatory agents. A further embodiment is a liposomal composition as described herein comprising an anti-inflammatory agent and a further therapeutic agent.
  • As a further embodiment, there is provided a pharmaceutical composition comprising a liposomal composition comprising an anti-inflammatory agent as an entrapped agent, or folate-targeted dendrimer conjugate, as described herein and further comprising at least one pharmaceutically acceptable carrier or excipient. The pharmaceutical composition may be formulated in a conventional manner as known for the formulation of liposomes for administration to patients. For example, the pharmaceutical composition may be a sterile dispersion in a buffered aqueous vehicle, optionally containing a tonicity adjusting agent. Alternatively, for example, the pharmaceutical composition may be a sterile, non-pyrogenic lyophilized product comprising a buffer and/or a tonicity adjusting agent, which is suitable for reconstitution with sterile water for injection to form a sterile dispersion. In either case, the sterile dispersion may diluted in 5% dextrose to an appropriate concentration prior to intravenous infusion. A pharmaceutical composition includes a veterinary composition.
  • As a further embodiment, there is provided a liposomal composition comprising an anti-inflammatory agent as an entrapped agent, or folate-targeted dendrimer conjugate, as described herein for use as a medicament. As a further embodiment, there is provided a liposomal composition comprising an anti-inflammatory agent as an entrapped agent, or folate-targeted dendrimer conjugate, as described herein for use in the treatment of an inflammatory disease. As a further embodiment, there is provided the use of a liposomal composition comprising an anti-inflammatory agent as an entrapped agent, or folate-targeted dendrimer conjugate, as described herein for the treatment of an inflammatory disease. As a further embodiment, there is provided the use of a liposomal composition comprising an anti-inflammatory agent as an entrapped agent, or folate-targeted dendrimer conjugate, as described herein for the manufacture of a medicament for the treatment of an inflammatory disease.
  • As a further embodiment, there is provided a method of treatment of an inflammatory disease in a subject in need thereof, comprising administering an effective amount of a liposomal composition comprising an anti-inflammatory agent as an entrapped agent, or folate-targeted dendrimer conjugate, as described in any of claims. In one embodiment the subject is a mammal. In one embodiment the subject is a human.
  • For any of the above uses or methods of treatment, in one embodiment the inflammatory disease comprises an inflamed tissue region in the body. In one embodiment the use or method is one wherein the inflammatory disease is arthritis, arteriosclerosis, graft-versus-host disease, multiple sclerosis, osteomyelitis, psoriasis or inflammatory bowel disease, such as Crohn's disease or ulcerative colitis. In one embodiment the use or method one wherein the inflammatory disease is arthritis. In one embodiment the use or method one wherein the inflammatory disease is rheumatoid arthritis. In one embodiment the use or method one wherein the inflammatory disease is osteoarthritis. In one embodiment the use or method one wherein the inflammatory disease is arteriosclerosis. In one embodiment the use or method one wherein the inflammatory disease is atherosclerosis. In one embodiment the use or method one wherein the inflammatory disease is inflammatory bowel disease, such as Crohn's disease or ulcerative colitis.
  • One embodiment is the use of a folate-targeted liposomal composition, or folate- targeted dendrimer conjugate, comprising an imaging or visualizing agent as an entrapped agent for diagnosis or monitoring the treatment of an inflammatory disease in an inflamed tissue region in the body. Another embodiment is the use of a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate, comprising an imaging or visualizing agent as an entrapped agent for the manufacture of an agent for diagnosis or monitoring the treatment of an inflammatory disease in an inflamed tissue region in the body. Another embodiment is an agent for diagnosis or monitoring the treatment of an inflammatory disease, comprising a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate, comprising an imaging or visualizing agent as an entrapped agent. Another embodiment is a pharmaceutical composition comprising a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate, comprising an imaging or visualizing agent as an entrapped agent and further comprising at least one pharmaceutically acceptable carrier or excipient. A further embodiment is a method of using a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate, comprising an imaging or visualizing agent as an entrapped agent for diagnosis or monitoring the treatment of an inflammatory disease in an inflamed tissue region in the body in a subject in need thereof. One embodiment of the method is wherein the subject is a mammal. Another embodiment of the method is wherein the subject is a human.
  • For any use, agent, pharmaceutical composition or method of using a folate-targeted liposomal composition wherein the entrapped agent is a folate-targeted liposomal composition comprising an imaging or visualizing agent, other than the difference in the entrapped agent, embodiments of the folate-targeted liposomal composition may be characterized in the same manner as embodiments of a folate-targeted liposomal composition comprising an anti-inflammatory agent as an entrapped agent as described herein. Similarly for folate-targeted dendrimer conjugates comprising an imaging or visualizing agent, embodiments may be characterized in the same manner as embodiments comprising an anti-inflammatory agent.
  • One embodiment of the above use, agent, pharmaceutical composition or method is one wherein the entrapped or conjugated agent is a pharmaceutically acceptable fluorescent dye. In one embodiment, the fluorescent dye is DiD (1,1′-dioctadecyl-3,3,3′,3′-tetramethyl-indodicarbocyanine perchlorate), calcein or fluorescein.
  • Another embodiment of the above use, agent, pharmaceutical composition or method is one wherein the entrapped or conjugated agent is a contrast agent for X-ray, MRI (magnetic resonance imaging) or ultrasound. One embodiment is the X-ray contrast agent is iobitridol.
  • Another embodiment of the above use, agent, pharmaceutical composition or method is one wherein the entrapped or conjugated agent comprises a radionuclide. In one embodiment, the radionuclide is an isotope of gallium, indium, copper, technitium or rhenium. In another embodiment, the radionuclide is an isotope of technitium.
  • One embodiment comprises a kit comprising a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate, comprising an imaging or visualizing agent as an entrapped agent as described herein and instructions for diagnosis or monitoring the treatment of an inflammatory disease.
  • As described above, a folate-targeted liposomal composition comprising the fluorescent dye calcein has been described. (M. J. Turk et al., Cancer Letters 213 (2004) 165-172.) However, a folate-targeted liposomal composition comprising an imaging or visualizing agent other than a fluorescent dye as an entrapped agent provides a novel embodiment. For a folate-targeted liposomal composition comprising an imaging or visualizing agent other than a fluorescent dye as an entrapped agent, embodiments of the folate-targeted liposomal composition may be characterized in the same manner as embodiments of a folate-targeted liposomal composition comprising an anti-inflammatory agent as an entrapped agent as described herein.
  • In one embodiment the folate-targeted liposomal composition, or folate-targeted dendrimer conjugate, comprising an imaging or visualizing agent other than a fluorescent dye as an entrapped agent is one wherein the entrapped agent is an X-ray contrast agent. In one embodiment, the X-ray contrast agent is iobitridol.
  • In another embodiment the folate-targeted liposomal composition, or folate-targeted dendrimer conjugate, comprising an imaging or visualizing agent other than a fluorescent dye as an entrapped agent is one wherein the entrapped agent comprises a radionuclide. In one embodiment, the radionuclide is an isotope of gallium, indium, copper, technitium or rhenium. In one embodiment, the radionuclide is an isotope of technitium.
  • One embodiment is a pharmaceutical composition comprising a liposomal composition, or folate-targeted dendrimer conjugate, comprising an imaging or visualizing agent other than a fluorescent dye as an entrapped agent and further comprising at least one pharmaceutically acceptable carrier or excipient. Another embodiment is the use of a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate, comprising an imaging or visualizing agent other than a fluorescent dye as an entrapped agent for diagnosis or monitoring the treatment of an inflammatory disease in an inflamed tissue region in the body. Another embodiment is the use of a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate, comprising an imaging or visualizing agent other than a fluorescent dye as an entrapped agent for the manufacture of an agent for diagnosis or monitoring the treatment of an inflammatory disease in an inflamed tissue region in the body. Another embodiment is an agent for diagnosis or monitoring the treatment of an inflammatory disease, comprising a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate, comprising an imaging or visualizing agent other than a fluorescent dye as an entrapped agent. A further embodiment is a method of using a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate, comprising an imaging or visualizing agent other than a fluorescent dye as an entrapped agent for diagnosis or monitoring the treatment of an inflammatory disease in an inflamed tissue region in the body in a subject in need thereof. One embodiment of the method is wherein the subject is a mammal. Another embodiment of the method is wherein the subject is a human.
  • An embodiment is a kit comprising a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate, comprising an anti-inflammatory agent as an entrapped agent, or a pharmaceutical composition thereof, as described herein, and an imaging agent for the inflammatory condition. In one embodiment of the kit, the imaging agent is a folate-targeted imaging agent. In another embodiment of the kit, the folate-targeted imaging agent is a folate-targeted liposomal composition, or folate-targeted dendrimer conjugate, as described herein. In a further embodiment of the kit, the folate-targeted imaging agent is a folate-targeted conjugate of an imaging agent. In one embodiment of the kit, the folate-targeted conjugate is EC20 (folate-Tc99m).
  • An additional embodiment of any of the above described kits is a kit further comprising instructions for diagnosis or monitoring the treatment of an inflammatory disease. An additional embodiment of any of the above described kits is a kit further comprising instructions for the treatment of an inflammatory disease.
  • Embodiments of the invention are further described by the following enumerated clauses:
  • 1. A folate-receptor targeted agent comprising a therapeutic agent useful for the treatment of inflammatory disease.
  • 2. The folate-receptor targeted agent of clause 1 which is a folate-targeted liposomal composition comprising an anti-inflammatory agent as an entrapped agent.
  • 3. The liposomal composition of clause 2 wherein the lipid bilayer is primarily composed of DSPC/cholesterol with a mol ratio of about 56:40.
  • 4. The liposomal composition of clause 2 or 3 wherein the lipid bilayer comprises a comprises a folate-targeting conjugate composed of (a) a lipid having a polar head group and a hydrophobic tail, (b) a hydrophilic polymer having a first end and a second end, said polymer attached at its first end to the head group of the lipid, and (c) a folate ligand (Fol) attached to the second end of the polymer, and wherein the folate ligand is a folic acid residue or an analog or derivative thereof.
  • 5. The liposomal composition of clause 4 wherein the hydrophilic polymer of the folate targeting conjugate is a PEG having an average molecular weight of about 200-5000 in which the end groups, prior to attachment, are independently amino, hydroxy, thiol or carboxy.
  • 6. The liposomal composition of any of clauses 2-5 wherein the folate targeting conjugate is present at about 0.01 mol % to about 1 mol % in the lipid bilayer.
  • 7. The liposomal composition of any of clauses 4-6 wherein the lipid bilayer further comprises a hydrophilic coating composed of (a) a lipid having a polar head group and a hydrophobic tail and (b) a hydrophilic polymer having a first end and a second end, said polymer attached at its first end to the head group of the lipid and optionally capped at its second end.
  • 8. The liposomal composition of clause 7 wherein the hydrophilic coating is a polyethylene glycol (PEG), a polylactic acid (PLA), a polyglycolic acid (PGA) or a polyvinyl alcohol (PVA).
  • 9. The liposomal composition of clause 7 or 8 wherein the hydrophilic polymer is a polyethylene glycol with an average molecular weight of about 2000, which is uncapped or capped with a methyl or ethyl group.
  • 10. The liposomal composition of any of clauses 2-9 wherein the entrapped agent is an anti-inflammatory steroid.
  • 11. The liposomal composition of any of clauses 2-10 wherein the entrapped agent is a drug useful in the treatment of rheumatoid arthritis or other autoimmune disease including an antiproliferative, immunomodulator or immunosuppresant agent.
  • 12. A method of treatment of an inflammatory disease in a subject in need thereof, comprising administering an effective amount of a liposomal composition as described in any of clauses 2-12.
  • 13. A pharmaceutical composition comprising a liposomal composition comprising an anti-inflammatory agent as an entrapped agent as described in any of clauses 2-12 and further comprising at least one pharmaceutically acceptable carrier or excipient.
  • 14. A composition comprising a folate-targeted liposomal composition comprising an imaging or visualizing agent as an entrapped agent and further comprising at least one pharmaceutically acceptable carrier or excipient.
  • 15. A method of using a folate-targeted liposomal composition comprising an imaging or visualizing agent as an entrapped agent for diagnosis or monitoring the treatment of an inflammatory disease in an inflamed tissue region in the body in a subject in need thereof.
  • 16. A kit comprising a folate-targeted liposomal composition comprising an imaging or visualizing agent as an entrapped agent as described in clause 14 and instructions for diagnosis or monitoring the treatment of an inflammatory disease.
  • 17. A kit comprising a folate-targeted liposomal composition comprising an anti-inflammatory agent as an entrapped agent, as described in any of clauses 2-11, or a pharmaceutical composition thereof, as described in clause 13, and an imaging agent for the inflammatory condition.
  • 18. The folate-receptor targeted agent of clause 1 which is a folate-targeted dendrimer conjugate comprising a folate-targeted dendrimer conjugated to an anti-inflammatory agent.
  • 19. The folate-targeted dendrimer conjugate of clause 18 wherein the dendrimer comprises a folate-targeting conjugate wherein the folate-targeting ligand (Fol) is a folic acid residue or an analog or derivative thereof, a hydrophilic coating, and a conjugated anti-inflammatory agent.
  • 20. The folate-targeted dendrimer conjugate of clause 19 wherein the folate targeting conjugate is present at about 12-25% of the dendrimeric termini; the hydrophilic coating residue is present at about 25-40% of the dendrimeric termini; and the conjugated to anti-inflammatory agent is present at about 3-13% of the dendrimeric termini.
  • 21. The folate-targeted dendrimer conjugate of clause 19 or 20 wherein the hydrophilic coating is a polyethylene glycol (PEG), a polylactic acid (PLA), a polyglycolic acid (PGA) or a polyvinyl alcohol (PVA).
  • 22. The liposomal composition of any of clauses 19-21 wherein the hydrophilic polymer is a polyethylene glycol with an average molecular weight of about 2000, which is uncapped or capped with a methyl or ethyl group.
  • 23. The folate-targeted dendrimer conjugate of any of clauses 19-22 wherein the conjugated anti-inflammatory agent is an anti-inflammatory steroid.
  • 24. The folate-targeted dendrimer conjugate of any of clauses 19-23 wherein the conjugated anti-inflammatory agent is a drug useful in the treatment of rheumatoid arthritis or other autoimmune disease including an antiproliferative, immunomodulator or immunosuppresant agent.
  • 25. A method of treatment of an inflammatory disease in a subject in need thereof, comprising administering an effective amount of a folate-targeted dendrimer conjugate as described in any of clauses 18-24.
  • 26. A pharmaceutical composition comprising a folate-targeted dendrimer conjugate comprising an anti-inflammatory agent as described in any of clauses 18-24 and further comprising at least one pharmaceutically acceptable carrier or excipient.
  • 27. A composition comprising a folate-targeted dendrimer conjugate comprising an imaging or visualizing agent as a conjugated agent and further comprising at least one pharmaceutically acceptable carrier or excipient.
  • 28. A method of using a folate-targeted dendrimer conjugate comprising an imaging or visualizing agent as a conjugated agent for diagnosis or monitoring the treatment of an inflammatory disease in an inflamed tissue region in the body in a subject in need thereof.
  • 29. A kit comprising a composition comprising a folate-targeted dendrimer conjugate comprising an imaging or visualizing agent as a conjugated agent as described in clause 27 and instructions for diagnosis or monitoring the treatment of an inflammatory disease.
  • 30. A kit comprising a folate-targeted dendrimer conjugate comprising an anti-inflammatory agent as described in clause 18-24, or a pharmaceutical composition thereof, as described in clause 26, and an imaging agent for the inflammatory condition.
  • Exemplary preparations of folate-targeted liposomal compositions comprising an anti-inflammatory agent or an imaging agent as an entrapped agent, as well as exemplary preparations of the corresponding non-folate-targeted liposomal compositions, are provided below in the examples.
  • Arteriosclerosis has traditionally been viewed to simply reflect the deposition of lipids within the vessel wall of medium-sized and large arteries. Upregulation of cell adhesion molecules facilitates adherence of leukocytes to the dysfunctional endothelium and their subsequent transmigration into the vessel wall. Evolving inflammatory reaction results in the initiation of atherosclerotic plaques. A large number of recent studies demonstrate that activated macrophages constitute the key effector cells in atherosclerosis; and it was reported that the folate receptor, FRβ, the nonepithelial isoform of the folate receptor is expressed on activated (but not resting) macrophages.
  • That the instant folate-targeted liposomal composition can deliver its entrapped agent to inflammation induced activated macrophages is demonstrated in Imaging Example 1, below. In that example the uptake of calcein from a calcein-entrapped folate-targeted liposomal composition by cells from rat peritoneal fluid in vitro is demonstrated in a thioglycolate-induced inflammation model using flow cytometry (FIG. 14). The calcein uptake is competitively inhibited by incubation in the presence of excess folic acid; and the comparable nontargeted (NT) liposomal composition delivers significantly less calcein. Because the dye within the liposomes is quenched at high concentration, the fluorescence is observed only following endocytosis and calcein unloading inside the target cells. Accordingly, such imaging experiments support the unloading of therapeutic agents in similar situations.
  • Apo E mice with the knock out mutation show a marked increase in total plasma cholesterol levels that are unaffected by age or sex. Fatty streaks in the proximal aorta are found at 3 month of age. The lesions increase with age and progress to lesions with less lipid but more elongated cells, typical of a more advanced stage of pre-atherosclerotic lesions. At 24 weeks of age, mice fed a normal diet show obvious atherosclerotic lesions in the aortic sinus and the ascending aorta. The number of atherosclerotic lesions in the aortic sinus and the ascending aorta is significantly increased in homozygotes fed an atherosclerotic western diet vs a normal diet. Imaging Example 2 demonstrates the increased uptake of of 3H-cholesterol labeled folate-targeted liposomes in the aorta and hearts of mice fed a high fat western diet compared to a normal diet, indicating the increase in atheroscerotic lesions in the western diet animal.
  • Imaging Examples 3 through 6 demonstrate the ability of folate-targeted liposomal compositions to deliver entrapped imaging agents to atherosclerotic sites in an atherosclerosis model employing Apo E knock out (Apo E KO) mice. Therapeutic Examples 1 and 2 demonstrate the ability of betamethasone-entrapped folate-targeted liposomal compositions to reduce the evidence of atherosclerotic inflammation in Apo E KO mice.
  • In Imaging Example 3, ability of a folate-targeted liposomal composition, with the entrapped fluorescent dye DiD, to label the atherosclerotic sites in Apo E KO mice fed a western diet for one week before dosing is demonstrated along with the competitive blocking of the labeling by pre-administered free folic acid. Imaging of both the whole animals (FIG. 2) and the excised aortic arch (FIG. 3) show the labeling of the sites and competitive blocking.
  • Imaging Example 4 (FIG. 4) provides a similar example in Apo E KO mice fed a western diet for four weeks before imaging under four conditions: (A) control with no imaging agent, (B) DiD-entrapped folate-targeted liposomal composition, (C) non-targeted DiD-entrapped liposomal composition, and (D) DiD-entrapped folate-targeted liposomal composition administered with an excess of folic acid. The images of the mice of each of the four treatment groups demonstrate superior uptake of the dye from the DiD-entrapped folate-targeted liposomal composition compared to the non-targeted DiD-entrapped liposomal composition, as well as the competitive inhibition of uptake of the dye from the DiD-entrapped folate-targeted liposomal composition by excess folic acid.
  • Imaging Example 5 provides an example of an Apo E KO mouse dosed with an iobitridol-entrapped folate-targeted liposomal composition which demonstrates the use of an entrapped CT agent for radiographic imaging (FIG. 5).
  • Imaging Example 6 provides an example using radioisotopic imaging in Apo E KO mice fed a western diet for four weeks before dosing using a 99Tc-entrapped folate-targeted liposomal composition, adminisered either alone or with an excess of folic acid (FIG. 6), demonstrating the competitive blockage of uptake of the folate-targeted liposomal composition by excess folic acid.
  • In Therapeutic Example 1, Apo E KO mice were fed a western diet for 3 weeks and dosed as (A) a control (Untreated) group which received a folate-targeted liposomal composition which contained only phospate buffered saline as the entrapped agent, (B) a group (NT Liposome) which received a non-targeted liposomal composition which contained betamethasone as the entrapped agent, and (C) a group (Fol-Liposome) which received a folate-targeted liposomal composition which contained betamethasone as the entrapped agent. The mice were imaged using the folate-receptor targeted, radionuclide conjugate imaging agent EC20 (folate-Tc99m) and the biodistribution of EC20 uptake was determined. The reduction in inflammatory cells imaged by EC20 in the Fol Liposome group compared to the NT Liposome and Untreated groups is demonstrated by the imaged mice (FIG. 7, top) and excised hearts (FIG. 7, bottom), particulary the heart uptake of EC20 (FIG. 8), and biodistribution (FIG. 9).
  • In Therapeutic Example 2, Apo E KO mice were fed a western diet for 4 weeks before dosing as (A) a control (Untreated) group which received a folate-targeted liposomal composition which contained only phospate buffered saline as the entrapped agent, (B) a group (NT Liposome) which received a non-targeted liposomal composition which contained betamethasone as the entrapped agent, and (C) a group (Fol Liposome) which received a folate-targeted liposomal composition which contained betamethasone as the entrapped agent. The mice were imaged using the folate-receptor targeted, radionuclide conjugate imaging agent EC20 (folate-Tc99m) and the biodistribution of EC20 uptake was determined. The reduction in inflammatory cells imaged by EC20 in the Fol Liposome group compared to the NT Liposome and Untreated groups is demonstrated by the measures of EC20 uptake in the hearts and the aortas of the groups FIG. 10 (top); FIG. 10 (bottom) shows the relative biodistribution of EC20 uptake in the organs of the groups.
  • Imaging Examples 7 through 9 demonstrate the ability of folate-targeted liposomal compositions to deliver entrapped imaging agents to sites of inflammation in other tissues and support the use of folate-targeted liposomal compositions comprising an anti-inflammatory agent as an entrapped agent, as disclosed herein, in the treatment of an inflammatory disease, such as in an inflamed tissue region in the body.
  • Imaging Example 7 demonstrates (FIG. 11) the ability of the folate-targeted liposomal compositions to deliver entrapped agents to inflamed intestinal tissues, for example in inflammatory bowel disease, such as Crohn's disease or ulcerative colitis, using imaging under the following conditions: (A) DiD-entrapped folate-targeted liposomal composition, (B) DiD-entrapped folate-targeted liposomal composition administered with an excess of folic acid, and (C) non-targeted DiD-entrapped liposomal composition.
  • Imaging Example 8 demonstrates (FIG. 12) the ability of the folate-targeted liposomal compositions to deliver entrapped agents to inflamed arthritic tissues, for example as in rheumatoid arthritis, using imaging under the following conditions: (A) DiD-entrapped folate-targeted liposomal composition, (B) DiD-entrapped folate-targeted liposomal composition administered with an excess of folic acid, and (C) non-targeted DiD-entrapped liposomal composition.
  • Imaging Example 8A also demonstrates (FIG. 15) the ability of the folate-targeted liposomal compositions to deliver entrapped agents to inflamed arthritic tissues, for example as in rheumatoid arthritis, using imaging under the following conditions: a non-targeted DiD-entrapped liposomal composition (NT Liposome), a diseased rat imaged with a folate-targeted DiD-entrapped liposomal composition (Fol-Liposome), and a healthy rat with a folate-targeted DiD-entrapped liposomal composition (Healthy).
  • Imaging Example 9 demonstrates (FIG. 13) the ability of the folate-targeted liposomal compositions to deliver entrapped agents to inflamed skeletal muscle tissues using imaging with (A) a DiD-entrapped folate-targeted liposomal composition, and (B) a DiD-entrapped folate-targeted liposomal composition administered with an excess of folic acid.
  • Imaging Examples 10 and 11 demonstrate (FIG. 16, FIG. 17 and FIG. 18) that the folate-targeted liposomal compositions preferentially deliver entrapped agents to inflammatory tissues as compared to tumors which express a high level of folate receptors on their surface. In addition, the examples demonstrate that the increase in uptake of the folate-targeted liposomal compositions compared to the non-targeted liposomal compositions is greater in the inflammatory tissues than the increase in the tumors.
  • Imaging Example 12 demonstrates uptake of a folate-targeted dendrimer conjugate conjugated to FITC dye and the competitive inhibition of the uptake by excess folic acid in the cells which express the folate receptor at high levels on the surface.
  • In Imaging Example 13, ability of a dendrimer conjugate conjugated to Cy5.5 dye to label the atherosclerotic sites in Apo E KO mice fed a western diet before dosing is demonstrated along with a comparison with a non-targeted dendrimer conjugate conjugated to Cy5.5 dye and the competitive blocking of the labeling by competition with a folate-targeted dendrimer conjugate which lacks a conjugated dye. Imaging of both the whole animals (FIG. 22) and the excised aortic arch (FIG. 23) show the labeling of the sites and competitive blocking.
  • Imaging Example 14 demonstrates (FIG. 24) the ability of a dendrimer conjugate to deliver conjugated agents to inflamed intestinal tissues in an ulcerative colitis model using using imaging with a folate-targeted dendrimer conjugate conjugated to Cy5.5 dye, with a non-targeted dendrimer conjugate conjugated to Cy5.5 dye, and in competition with a folate-targeted dendrimer conjugate which lacks a conjugated dye.
  • The following examples further illustrate specific embodiments of the invention; however, the following illustrative examples should not be interpreted in any way to are to limit invention. Abbreviations used in the examples include the following: Chol, cholesterol; DCC, dicyclohexylcarbodiimide; DiD, 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindodicarbocyanine perchlorate; DIPEA, diisopropylethylamine; DMSO, dimethyl sulfoxide; DSPC, distearoyl-phosphatidylcholine; DSPE, distearoylphosphatidylethanolamine; EDC, 1-ethyl-3-(3-dimethyl-aminopropyl)carbodiimide hydrochloride; mPEG, methyl capped PEG; MS, mass spectrometry; PBS, phosphate buffered saline; PEG, poly(ethylene glycol).
  • EXAMPLES Example Preparations Exemplary Preparation 1 Preparation of Folate-Targeted Liposomal Compositions Synthesis of Folate-PEG-NH2
  • Figure US20130071321A1-20130321-C00001
  • Under N2, Folate-PEG-NH2 is synthesized using 500 mg of NH2PEG3200NH2 (PEG-bis amine) with an equimolar quantity of folic acid in 5 mL DMSO containing 1 molar equivalent of DCC or EDC and 10 μl pyridine, triethylamine or DIPEA. The reaction mixture is stirred overnight in the dark at room temperature. The reaction is quenched with water. The by-product urea and the trace amount of unreacted folate and PEG-bis amine are removed by both dialysis and by using a G-15 size exclusion column in deionized water. Folate-PEG-NH2 is analyzed using HPLC with monitoring by absorbance at 363 nm.
  • Synthesis of N-succinyl-DSPE
  • Figure US20130071321A1-20130321-C00002
  • Under N2, N-succinyl-DSPE is synthesized by reacting overnight 1.1 molar equivalent of succinic anhydride with 100 mg DSPE in 5 mL of dry chloroform or dichloromethane containing 10 μL pyridine, triethylamine or DIPEA. The product N-succinyl-DSPE runs as a spot with a higher Rf value (˜0.5) than DSPE and is ninhydrin negative on TLC, using chloroform/methanol (3:2) as the solvent phase. The structure of the product N-succinyl-DSPE is confirmed by MS.
  • Synthesis of Folate-PEG-DSPE
  • Figure US20130071321A1-20130321-C00003
  • The synthesis of the folate-PEG-DSPE construct is illustrated above. Under nitrogen, N-succinyl-DSPE is dissolved in chloroform and its carboxyl-group is activated by reacting with 0.95 molar equivalent of DCC or EDC. Then an equimolar amount of folate-PEG-DSPE is also dissolved in the above mentioned reaction mixture and the reaction is continued overnight at room temperature. The solvent is then removed under reduced pressure. The unreacted products are then removed from the mixture by membrane dialysis. The product folate-PEG-DSPE is analyzed on a HPLC system chromatography (water/methanol (1.5:1)) using a C-18 reverse phase column.
  • Preparation of Betamethasone-Entrapped Folate-Targeted Liposomal Composition:
  • 126.4 mg of DSPC, 44 mg of cholesterol, 32 mg of mPEG2000DSPE and 1 mg of FolatePEG3200DSPE are weighed and mixed in a 50 mL round bottom flask. The lipid mixture is dissolved in chloroform before evaporating to dryness in a Rotavapor and kept under vacuum overnight. 200 mg of betamethasone phosphate is weighed and dissolved in 6 mL of phosphate buffered saline. The betamethasone solution is added to the dry lipid film, and the mixing is further enhanced at 70 ° C. The mixture is subjected to freeze-thaw cycle 10 times. The liposomal suspension is transferred to an extruder and extruded under nitrogen pressure, 10 times each through 200 nm, 100 nm, and 50 nm pore filters, respectively. The mean particle size of the liposome as determined by light scattering is 60-70 nm.
  • Preparation of calcein entrapped-folate-targeted liposomal compositions:
  • In order to prepare calcein entrapped folate-PEG-liposomal compositions, a total mass of 100 mg of the lipid mixture; [DSPC/Chol/mPEG2000-DSPE/folate-PEG-DSPE (56:40:4:0.1) mol %] is dissolved in chloroform and dried to a thin film in a round bottom flask under reduced pressure overnight. The lipid mixture is rehydrated in 10 mM calcein dissolved in PBS (pH 7.4). The resulting lipid suspension is subjected to 10-15 cycles of freezing and thawing. Then 50 nm and100 nm folate-PEG-liposomes of different size distributions are generated by extruding the lipid suspension 10 times each through polycarbonate membranes having pore sizes of sequentially 800 nm, 400 nm, 200 nm and 100 nm, followed by 50 nm, if needed, respectively. Unentrapped dye is separated using membrane dialysis. Particle size is determined by Dynamic Light Scattering-DynaPro99. Phosphate assay (Steward assay) is performed to determined phospholipid concentration.
  • Preparation of DiD-Entrapped Folate-Targeted Liposomal Composition (1% Dye with Respect to Phospholipid):
  • 126.4 mg of DSPC, 44 mg of cholesterol, 32 mg of mPEG2000DSPE and 1 mg of FolatePEG3200DSPE are weighed and mixed in a 50 mL round bottom flask. The lipid mixture is dissolved in chloroform before evaporating to dryness in a Rotavapor and kept under vacuum overnight. 15 μl of 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindodicarbocyanine perchlorate (DiD) is added in 6 mL of (pH 7.4) phosphate buffered saline. The dye solution is added to the dry lipid film, and the mixing is further enhanced at 70° C. The mixture is subjected to freeze-thaw-vortex cycle 10 times. The liposomal suspension is transferred to an extruder and extruded under nitrogen pressure, 10 times each through 200 nm, 100 nm, and 50 nm pore filters, respectively. The mean particle size of the liposome as determined by light scattering is about 60 nm.
  • Preparation of Iobitridol-Entrapped Folate-Targeted Liposomal Compositions:
  • Lipid mixture consisting of DSPC/Chol/mPEG2000DSPE/FolPEG3200DSPE in a 56:40:4:0.1 molar ratio was dissolved in ethanol at 70° C. The ethanol solution was then hydrated with iobitridol (350 mg I/mL) for 2 h. The suspension of liposomes obtained was submitted to a filtration through 400 nm, 200 nm, 100 nm, and 50 nm (10 cycles each) polycarbonate membranes using an extruder. Liposomes were then dialyzed overnight in a 300K Mw cutoff dialysis bag against PBS solution.
  • Preparation of Chelated 99Tc-Entrapped Folate-Targeted Liposomal Compositions by Remote Loading:
  • (Part 1) Preparation of folate-targeted liposome encapsulating glutathione.
  • Folate-targeted liposomes encapsulating glutathione were prepared using a procedure based on polycarbonate membrane extrusion. Briefy, unilamellar vesicles were prepared from DSPC/Chol/mPEG2000DSPE/FolPEG3200DSPE (56:40:4:0.1) by thin film hydration method. Lipids at indicated ratios were dissolved in chloroform, and the solvent was evaporated under vacuum with a stream of N2 gas to remove all organic solvent. The resultant film was then hydrated with 50 mM reduced glutathione in PBS (pH 7.4) at 60° C. with constant stirring. The suspension of liposomes obtained was submitted to a filtration through 400 nm, 200 nm, 100 nm, and 50 nm (10 cycles each) polycarbonate membranes using an extruder. Untrapped residual reduced glutathione was removed by passing the liposome suspension through a Sephadex G50 column.
  • (Part 2) Radiolabeling of Tc-99m into Liposomes
  • The commercially available kit of HMPAO, hexamethylpropylene amine oxime (Ceretec®, UK) was labeled with Tc-99m. The kit was reconstituted with 15 mCi sodium pertechnetate in 0.9% NaCl solution at room temperature for 8 minutes. The 99mTc-HMPAO complex was then mixed with the preformed liposome (part1) and incubated at 37° C. with intermittent vortexing for 30 minutes. Uncapsulated 99mTc-HMPAO was removed by gel filtration on a Sephadex G50 column using PBS as an eluent.
  • Exemplary Preparation 2 Preparation of NonTargeted (NT) Liposomal Compositions
  • For comparison experiments, liposomal compositions are prepared as disclosed for the preparation of the corresponding folate-targeted liposomal compositions, except the folate-PEG-DSPE conjugate is omitted.
  • Exemplary Preparation 3 Preparation of Folate-Targeted Dendrimer Conjugates
  • An exemplary preparation of a folate-targeted dendrimer conjugate conjugaged to a fluorescent dye (Cy 5.5 NHS or FITC) using a Dendritech Generation 3 (G3) poly(amido amine) (PAMAM), denoted as “Dendrimer,” having a molecular weight of 6,909 Daltons, a measured diameter of 36 Å, and 32 surface primary amino groups follows: On the average, each resulting dendrimer conjugate contains six folate-PEG residues, ten mPEG residues and two Cy 5.5 or FITC residues.
  • Synthesis of Folate-PEG-NH2
  • Figure US20130071321A1-20130321-C00004
  • Under N2, Folate-PEG-COOH is synthesized using 500 mg of NH2PEGCOOH (Mw′3400) with equimolar quantity of folic acid in 5 mL DMSO containing 1 molar equivalent of dicyclohexylcarbodiimide (DCC) or 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride (EDC) and 10 μL pyridine, triethylamine or diisopropylethylamine (DIPEA). The reaction mixture is stirred overnight in the dark at room temp. The reaction is quenched with H2O. The trace amount of unreacted folate is also removed by both dialysis and G-15 size exclusion column in dH2O. Folate-PEG-COOH is analyzed using HPLC by absorbance at 363 nm.
  • Synthesis of Folate-PEG-Dendrimer
  • Figure US20130071321A1-20130321-C00005
  • Under N2, FolatePEG-Dendrimer is synthesized by reacting overnight 6 molar equivalent of FolNHPEGCOOH and EDC (6 eq) with Dendrimer (1 eq) in 5 mL of dry DMSO containing 10 μL pyridine, triethylamine or DIPEA. The trace amount of unreacted materials is also removed by both dialysis and G-15 size exclusion column in dH2O. Folate-PEG-Dendrimer is analyzed using UV/Vis and IR.
  • Synthesis of Folate-PEG-Dendrimer with mPEG
  • Figure US20130071321A1-20130321-C00006
  • The synthesis of the pegylated folate—PEG-Dendrimer construct is illustrated above, briefly under nitrogen, mPEG-NHS (10 eq,Mw ˜2000) is dissolved in dry DMSO, and 40 μL pyridine, triethylamine or DIPEA is added. Then 1 equivalent amount of folate-PEG-Dendrimer is also dissolved with the above mentioned reaction mixture overnight at room temperature. The unreacted mixture is then removed by membrane dialysis, confirmed by UV/Vis and IR.
  • Loading of FITC or Cy 5.5 into Folate-PEG-Dendrimer:
  • Figure US20130071321A1-20130321-C00007
  • In order to prepare folate-PEG-Dendrimer loaded with dye (FITC or Cy 5.5), 1 equivalent of Folate-PEG-Dendrimer and Cy 5.5 NHS (2 eq) or FITC (2 eq) are dissolved in dry DMSO. 40 μL pyridine, triethylamine or DIPEA is added. The reaction is carried out overnight at room temperature. The unreacted mixture is then removed by membrane dialysis. The remaining product is freeze drid and confirmed by UV/Vis and IR.
  • Exemplary Preparation 4 Preparation of Non-Targeted Dendrimer Conjugates
  • For non-targeted dendrimer conjugates, the coupling with FolNHPEGCOOH is omitted.
  • Exemplary Preparation 5: Preparation of Folate-Targeted Dendrimer Conjugates without Conjugated Dye
  • For competition studies, the step of coupling with a dye is omitted to provide folate-dendrimer conjugates.
  • THERAPEUTIC EXAMPLES Therapeutic Example 1 Treatment with a Betamethasone-Entrapped Folate-Targeted Liposomal Composition in a 3-Week Model of Atherosclerosis in the Apo E Knock-Out Mouse
  • Three groups of Apo E KO mice (Untreated (A), NT Liposome (B) and Fol Liposome (C)) were fed with a western diet for 3 weeks. The Fol Liposome Treated group was injected i.v., twice per week, with a betamethasone-entrapped folate-targeted liposomal composition. The NT Liposome group was injected similarly with a non-targeted betamethasone-entrapped liposomal composition. The Untreated group was similarly injected with a folate-targeted liposomal composition with only encapsulated phosphate buffer solution. One day after the last liposome injection, all groups were injected with the folate-receptor targeted, radionuclide conjugate imaging agent EC20 (folate-Tc99m) before radioisotopic imaging and monitoring of EC20 uptake. The images are shown in FIG. 7 (top) of the whole animals and in FIG. 7 (bottom) of the hearts of the three groups. The relative EC 20 uptake in the heart for each of the three groups is shown if FIG. 8. The relative EC 20 uptake in various organs is shown in FIG. 9.
  • Therapeutic Example 2 Treatment with a Betamethasone-Entrapped Folate-Targeted Liposomal Composition in a 4-Week Model of Atherosclerosis in the Apo E Knock-Out Mouse
  • Three groups (Untreated (A), NT Liposome (B) and Fol-Liposome (C)) of Apo E KO mice (3 mice per group) were fed a western diet for 4 weeks. The Fol Liposome group was injected i.v. twice per week with a folate-targeted betamethasone-entrapped liposomal composition. The NT Liposome group was similarly injected with a non-targeted betamethasone-entrapped liposomal composition. The Untreated group was similarly injected with a folate-targeted liposomal composition with only encapsulated phosphate buffer solution. One day after the last liposome injection, the groups were injected with the folate-receptor targeted, radionuclide conjugate imaging agent EC20 (folate-Tc99m) before radioisotopic imaging and monitoring of EC20 uptake. FIG. 10 (top) shows the relative EC20 uptake of the hearts and the aortas of the groups; FIG. 10 (bottom) shows the relative biodistribution of EC20 uptake in the organs of the groups.
  • IMAGING EXAMPLES Imaging Example 1 Uptake of Calcein from a Calcein-Entrapped Folate-Targeted Liposomal Composition by Rat Peritoneal Cells in a Thioglycolate-Induced Inflammation Model
  • 30 g of dehydrated brewer thioglycolate medium powder was dissolved in 1 L of deionized water and autoclaved for 25 minutes at 15 pounds pressure (124° C.). The autoclaved medium was kept in the dark under sterile conditions at room temperature for at least 3 months before use.
  • Thioglycolate recruited macrophages were isolated by peritoneal lavage 3 days after intraperitoneal injection of 3.0 mL 3% sterile medium.
  • After 3 days, the Lewis rats were sacrificed by CO2 asphyxiation, and the peritoneal macrophages were harvested by injecting 60 mL of PBS into the peritoneal cavity. After centrifugal separation of the cells collected in the PBS, the cells were resuspended into Eppendorf tubes with FDRPMI 1640 medium. The medium was replaced with fresh medium containing Folate-targeted liposome encapsulating dye (DiD or calcein). For competition studies, excess free folic acid is preintroduced to the peritoneal medium for 30 min before addition of folate targeted liposome. For control, empty folate targeted liposome (PBS) were used. All media were incubated for 2 h at 37° C. . The cells were washed 3 times with PBS and resuspended in fresh PBS. The cell bound fluorescence was analyzed using flow cytometry.
  • In FIG. 14 are shown the results of the flow cytometry for control and calcein-entrapped folate-targeted liposomal compositions in the presence and absence of excess free folic acid.
  • Imaging Example 2 Labeling and Biodistribution of Atheroscerotic Lesions in Apo E Knock Out Mice Fed Normal and Western Diets
  • One set of Apo E KO mice (Normal Chow) was fed a normal diet and one set of Apo E KO mice (Western Diet) was fed a high fat western diet for two weeks. Each mouse then received a folate-targeted liposome composition (2 mg total phospholipid, i.v) containing 10 μCi 3H cholesterol-oleoyl-ether. The animals were sacrificed 24 h later, and organs were removed. Tissue samples were solubilized using Soluene 350 (1 mL/100 mg tissue) at 60° C. for 48 h. Tissue solutions were then bleached to uniform color using 30% hydrogen peroxide, followed by 6 mL Hionic-Fluor scintillation cocktail. 1 mL of tissue solution was counted on scintillation counter. The relative biodistribution of uptake of the labeled liposomes in the aorta and hearts of Normal Chow and Western Diet mice is shown in FIG. 1 (top); the biodistribution in a number of organs is shown in FIG. 1 (bottom).
  • In the following examples, when DiD is the imaging agent, approximately 50 nm DiD-entrapped liposomes of DSPC/Chol/mPEG2000DSPE/FolPEG3400DSPE (56:40:4:0.1) were used, along with the corresponding non-targeted liposomes. For competition studies excess 0.1 mM folic acid was used. Administration was i.p.
  • Imaging Example 3 Imaging with a DiD-Entrapped Folate-Targeted Liposomal composition in a 1-week model of atherosclerosis in the Apo E Knock-Out Mouse.
  • Apo E knockout mice are fed with western diet for 1 week. After 7 days, mice received the DiD-entrapped folate-targeted liposomal composition (fol-PEG-liposome-DiD) (0.2 μmol lipid) i.p.(total volume injected: 100 μl) in each mouse. For competition studies, mice were pre-administered free folic acid (50 μl) before i.p injection of DiD liposome. Images were taken 2 h postinjection. The images are shown in FIG. 2 and FIG. 3.
  • Imaging Example 4 Imaging with a DiD-Entrapped Folate-Targeted Liposomal Composition in a 4-Week Model of Atherosclerosis in the Apo E Knock-Out Mouse
  • Apo E knockout mice are fed with western diet for 4 weeks. DiD-entrapped liposomes were administered. The animals were imaged after 4 hours. The images are shown in FIG. 4.
  • Imaging Example 5 Imaging with Iobitridol-Entrapped Folate-Targeted Liposomal Composition in a Model of Atherosclerosis in the Apo E Knock-Out Mouse
  • An Apo E KO mouse (7 weeks old) was subjected to i.p. injection of the iobitridol-entrapped folate-targeted liposomal composition described above (3.0×10−6 mol total lipids). The mouse was imaged by X-ray: preinjection, T=2 , 3, 24, 48 hours interval. The images are shown in FIG. 5.
  • Imaging Example 6 Imaging with a 99Tc-Entrapped Folate-Targeted Liposomal Composition in an 8-Week Model of Atherosclerosis in the Apo E Knock-Out Mouse
  • Two groups of Apo E KO mice were fed with western diet for 8 weeks. One group of Apo E KO mice (competition group) was subjected to 0.1 mM free folic acid thru i.p. injection. After 1 hour, i.p. injection of the 99Tc-entrapped folate-targeted liposomal composition was preformed on both groups of mice. Images were taken 4 hours postinjection. The images are shown in FIG. 6.
  • Imaging Example 7 Imaging with a DiD-Entrapped Folate-Targeted Liposomal Composition in a Murine Intestinal Inflammation (Ulcerative Colitis) Model
  • Three C57 mice were fed with a 2% dextran sodium sulfate for 7 days. After 7 days, DiD-entrapped liposomes were administered. Intestines were removed and imaged after 4 h. The images are shown in FIG. 11.
  • Imaging Example 8 Imaging with a DiD-Entrapped Folate-Targeted Liposomal Composition in an Adjuvant-Induced Arthritis Model in the Rat
  • Adjuvant-induced arthritis (AIA) in Lewis rats was induced by injection of butyricum (0.15 mg) into a hind paw. After 17 days, DiD-entrapped liposomes were administered. The hind paws of the rats were imaged. The images are shown in FIG. 12.
  • Imaging Example 8A Imaging with a DiD-Entrapped Folate-Targeted Liposomal Composition in an Adjuvant-Induced Arthritis Model in the Rat
  • Adjuvant-induced arthritis (AIA) in Lewis rats was induced by injection of heat killed mycoplasmia butyricum (0.5 mg), suspended in mineral oil (5 mg/mL), on day 1 into the left hind foot of Lewis rats. Disease was allowed to progress for 17 days. 100% of animals developed arthritis as evidenced by gross swelling in the injected paw and progressive swelling in all extremities. DiD-entrapped liposomes were administered (i.v.). The hind paws of the rats were imaged after 12 hours. The images are shown in FIG. 15.
  • Imaging Example 9 Imaging with a DiD-Entrapped Folate-Targeted Liposomal Composition in a Cardiotoxin-Induced Muscle Injury Model in the Rat
  • Four mice were injected with 100 μL of a 10 μM solution of a cardiotoxin (from Naja mossambica mossambica), and two mice were used as controls. Four days later, the two healthy mice were injected with folate-targeted DiD-entrapped liposomes, as were two mice with cardiotixin-induced muscle injury (chosen randomly). The two remaining cardiotoxin-injected mice (chosen randomly) were treated with non-targeted DiD-entrapped liposome. 300 μg of phospholipid were injected per mouse. The dye used was 1% DiD. Images were taken eight hours after a tail vein injection. The images of three of the mice are shown in FIG. 13.
  • Imaging Example 10 Imaging with a DiD-Entrapped Folate-Targeted Liposomal Composition in a Cardiotoxin-Induced Muscle Injury Model in in Mice Implanted with M109 Tumor Cells
  • One million M109 cells were injected subcutaneously into a shoulder of Balb-C mice. On day 14 post tumor cell inoculation, the mice were injected in the opposite thigh with 100 μL of a 10 μM solution of a cardiotoxin (from Naja mossambica mossambica). Four days later, on day 18, mice were treated i.v. with a non-targeted DiD-entrapped liposomal composition (NT Liposome), with a folate-targeted DiD-entrapped liposomal composition (Fol-Liposome), and with empty folate-targeted liposome (injected empty folate-targeted liposome first, then dosed with a folate-targeted DiD-entrapped liposomal composition) (Competition with Folate Liposome). The mice were imaged 8 hours later. The images of the mice are shown in FIG. 17.
  • Imaging Example 11 Imaging with a DiD-Entrapped Folate-Targeted Liposomal Composition in a Murine Intestinal Inflammation (Ulcerative Colitis) Model in Mice Implanted with M109 Tumor Cells
  • One million M109 cells were injected subcutaneously into a shoulder of Balb-C mice. On day 11 post tumor cell inoculation, the mice were administered 2% dextran sodium sulfate (DSS) for 5 days. The mice were then treated i.v. with a non-targeted DiD-entrapped liposomal composition (NT Liposome), with a folate-targeted DiD-entrapped liposomal composition (Fol-Liposome), and with empty folate-targeted liposome (injected empty folate-targeted liposome first, then dosed with a folate-targeted DiD-entrapped liposomal composition) (Folate Liposome W Competion). The mice were imaged 8 hours later. The images of the mice are shown in FIG. 18.
  • Imaging Example 12 Uptake of Folate-Targeted Dendrimer Conjugate by Cells which Express the Folate Receptor at High Levels on the Surface (Control) and in the Presence of a Folate-Targeted Dendrimer Conjugate Conjugated to FITC dye (FolDendFITC) Alone and in the Presence of Excess Folic Acid (FolDendFITC w Excess Folic Acid)
  • Raw cells are plated in each well and treated with 10 μL of the indicated dendrimers (1.0 mg/mL). After 1 hour of incubation, confocal imaging is carried out. The images are shown in FIG. 21.
  • Imaging Example 13 Imaging in the Apo E Knock-Out Mouse model of Atherosclerosis with a Folate-Targeted Dendrimer Conjugate Conjugated to Cy5.5 dye (FolDend(G3)Cy5.5), with a Non-Targeted Dendrimer Conjugate Conjugated to Cy5.5 Dye, and in Competition with a Folate-Targeted Dendrimer Conjugate which Lacks a Conjugated Dye (FolDend(G3)Cy5.5 W Competion)
  • Apo E knockout mice are fed with western diet are treated with the indicated dendrimer conjugates and imaged 4 hours post injection. The images are shown in FIG. 22. The images of the excised aortas are shown in FIG. 23.
  • Imaging Example 14 Imaging in a Murine Intestinal Inflammation (Ulcerative Colitis) Model with a Folate-Targeted Dendrimer Conjugate Conjugated to Cy5.5 Dye (FolDend(G3)Cy5.5), with a Non-Targeted Dendrimer Conjugate Conjugated to Cy5.5 dye, and in Competition with a Folate-Targeted Dendrimer Conjugate which Lacks a Conjugated Dye (FolDend(G3)Cy5.5 W Competion)
  • C57 mice were fed with a 2% dextran sodium sulfate, and the indicated dendrimer conjugates were administered. The mice were imaged after 6 h. The images are shown in FIG. 24.

Claims (26)

1. A folate-receptor targeted agent comprising a therapeutic agent useful for the treatment of inflammatory disease which is a folate-targeted liposomal composition comprising an anti-inflammatory agent as an entrapped agent wherein the lipid bilayer is primarily composed of DSPC/cholesterol with a mol ratio of about 56:40.
2-3. (canceled)
4. The liposomal composition of claim 1 wherein the lipid bilayer comprises a comprises a folate-targeting conjugate composed of (a) a lipid having a polar head group and a hydrophobic tail, (b) a hydrophilic polymer having a first end and a second end, said polymer attached at its first end to the head group of the lipid, and (c) a folate ligand (Fol) attached to the second end of the polymer, and wherein the folate ligand is a folic acid residue or an analog or derivative thereof, wherein the hydrophilic polymer of the folate targeting conjugate is a PEG having an average molecular weight of about 200-5000 in which the end groups, prior to attachment, are independently amino, hydroxy, thiol or carboxy; and wherein the folate targeting conjugate is present at about 0.01 mol % to about 1 mol % in the lipid bilayer.
5-6. (canceled)
7. The liposomal composition of claim 4 wherein the lipid bilayer further comprises a hydrophilic coating composed of (a) a lipid having a polar head group and a hydrophobic tail and (b) a hydrophilic polymer having a first end and a second end, said polymer attached at its first end to the head group of the lipid and optionally capped at its second end, wherein the hydrophilic coating is a polyethylene glycol (PEG), a polylactic acid (PLA), a polyglycolic acid (PGA) or a polyvinyl alcohol (PVA).
8-9. (canceled)
10. The liposomal composition of claim 4 wherein the entrapped agent is an anti-inflammatory steroid.
11. The liposomal composition of claim 1 wherein the entrapped agent is a drug useful in the treatment of rheumatoid arthritis or other autoimmune disease including an antiproliferative, immunomodulator or immunosuppresant agent.
12. A method of treatment of an inflammatory disease in a subject in need thereof, comprising administering an effective amount of a liposomal composition as described in claim 1.
13. A pharmaceutical composition comprising a liposomal composition comprising an anti-inflammatory agent as an entrapped agent as described in claim 1 and further comprising at least one pharmaceutically acceptable carrier or excipient.
14. A composition comprising a folate-targeted liposomal composition comprising an imaging or visualizing agent as an entrapped agent and further comprising at least one pharmaceutically acceptable carrier or excipient, wherein the lipid bilayer of the liposomal composition is primarily composed of DSPC/cholesterol with a mol ratio of about 56:40 and, wherein the hydrophilic polymer of the folate targeting conjugate is a PEG having an average molecular weight of about 200-5000 in which the end groups, prior to attachment, are independently amino, hydroxy, thiol or carboxy; and wherein the folate targeting conjugate is present at about 0.01 mol % to about 1 mol % in the lipid bilayer.
15. A method of using a folate-targeted liposomal composition comprising an imaging or visualizing agent as an entrapped agent as described in claim 14 for diagnosis or monitoring the treatment of an inflammatory disease in an inflamed tissue region in the body in a subject in need thereof.
16. A kit comprising a folate-targeted liposomal composition comprising an imaging or visualizing agent as an entrapped agent as described in claim 14 and instructions for diagnosis or monitoring the treatment of an inflammatory disease.
17-18. (canceled)
19. A folate-targeting dendrimer conjugate comprising a folate-targeted dendrimer conjugated to an anti-inflammatory agent wherein the folate-targeting ligand (Fol) is a folic acid residue or an analog or derivative thereof, a hydrophilic coating, and a conjugated anti-inflammatory agent; and wherein the folate targeting conjugate is present at about 12-25% of the dendrimeric termini; the hydrophilic coating residue is present at about 25-40% of the dendrimeric termini; and the conjugated to anti-inflammatory agent is present at about 3-13% of the dendrimeric termini.
20. (canceled)
21. The folate-targeted dendrimer conjugate of claim 19 wherein the hydrophilic coating is a polyethylene glycol (PEG), a polylactic acid (PLA), a polyglycolic acid (PGA) or a polyvinyl alcohol (PVA).
22. (canceled)
23. The folate-targeted dendrimer conjugate of claim 19 wherein the conjugated anti-inflammatory agent is an anti-inflammatory steroid.
24. The folate-targeted dendrimer conjugate of claim 19 wherein the conjugated anti-inflammatory agent is a drug useful in the treatment of rheumatoid arthritis or other autoimmune disease including an antiproliferative, immunomodulator or immunosuppresant agent.
25. A method of treatment of an inflammatory disease in a subject in need thereof, comprising administering an effective amount of a folate-targeted dendrimer conjugate as described in claim 19.
26. A pharmaceutical composition comprising a folate-targeted dendrimer conjugate comprising an anti-inflammatory agent as described in claim 19 and further comprising at least one pharmaceutically acceptable carrier or excipient.
27. A composition comprising a folate-targeted dendrimer conjugate comprising an imaging or visualizing agent as a conjugated agent, wherein the folate-targeting ligand (Fol) is a folic acid residue or an analog or derivative thereof, a hydrophilic coating, and a conjugated anti-inflammatory agent; and wherein the folate targeting conjugate is present at about 12-25% of the dendrimeric termini; the hydrophilic coating residue is present at about 25-40% of the dendrimeric termini; and further comprising at least one pharmaceutically acceptable carrier or excipient.
28. A method of using a folate-targeted dendrimer conjugate comprising an imaging or visualizing agent as a conjugated agent as described in claim 27 for diagnosis or monitoring the treatment of an inflammatory disease in an inflamed tissue region in the body in a subject in need thereof.
29. A kit comprising a composition comprising a folate-targeted dendrimer conjugate comprising an imaging or visualizing agent as a conjugated agent as described in claim 27 and instructions for diagnosis or monitoring the treatment of an inflammatory disease.
30. A kit comprising a folate-targeted dendrimer conjugate comprising an anti-inflammatory agent as described in claim 19, or a pharmaceutical composition thereof, and an imaging agent for the inflammatory condition.
US13/700,358 2010-05-28 2011-05-27 Delivery of agents to inflamed tissues using folate-targeted liposomes Abandoned US20130071321A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/700,358 US20130071321A1 (en) 2010-05-28 2011-05-27 Delivery of agents to inflamed tissues using folate-targeted liposomes

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US34943410P 2010-05-28 2010-05-28
US13/700,358 US20130071321A1 (en) 2010-05-28 2011-05-27 Delivery of agents to inflamed tissues using folate-targeted liposomes
PCT/US2011/038437 WO2011150392A1 (en) 2010-05-28 2011-05-27 Delivery of therapeutic agents to inflamed tissues using folate-targeted agents

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/038437 A-371-Of-International WO2011150392A1 (en) 2010-05-28 2011-05-27 Delivery of therapeutic agents to inflamed tissues using folate-targeted agents

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/894,287 Division US11185505B2 (en) 2010-05-28 2020-06-05 Delivery of agents to inflamed tissues using folate-targeted liposomes

Publications (1)

Publication Number Publication Date
US20130071321A1 true US20130071321A1 (en) 2013-03-21

Family

ID=45004436

Family Applications (3)

Application Number Title Priority Date Filing Date
US13/700,358 Abandoned US20130071321A1 (en) 2010-05-28 2011-05-27 Delivery of agents to inflamed tissues using folate-targeted liposomes
US16/894,287 Active US11185505B2 (en) 2010-05-28 2020-06-05 Delivery of agents to inflamed tissues using folate-targeted liposomes
US17/519,171 Pending US20220054417A1 (en) 2010-05-28 2021-11-04 Delivery of agents to inflamed tissues using folate-targeted liposomes

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/894,287 Active US11185505B2 (en) 2010-05-28 2020-06-05 Delivery of agents to inflamed tissues using folate-targeted liposomes
US17/519,171 Pending US20220054417A1 (en) 2010-05-28 2021-11-04 Delivery of agents to inflamed tissues using folate-targeted liposomes

Country Status (3)

Country Link
US (3) US20130071321A1 (en)
CA (1) CA2800693A1 (en)
WO (1) WO2011150392A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015077303A1 (en) * 2013-11-19 2015-05-28 Purdue Research Foundation Patient selection method for inflammation
US20160158152A1 (en) * 2013-07-17 2016-06-09 Council Of Scientific & Industrial Research Pharmaceutical composition for the treatment of diminution of bone tissue
CN108430457A (en) * 2015-12-17 2018-08-21 南洋理工大学 The nano liposomes and preparation method thereof comprising corticosteroid as drug

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2854769B1 (en) 2012-07-05 2023-06-07 Taiwan Liposome Company, Ltd. Methods of treating arthritis
JP6192799B2 (en) 2013-03-15 2017-09-06 パーデュー・リサーチ・ファウンデーションPurdue Research Foundation Synthesis and composition of amino acid linking groups conjugated to compounds used for targeted imaging of tumors
MA40428A (en) * 2014-08-14 2016-02-18 L E A F Holdings Group Llc Liposome encapsulated affinity drug
KR20230146097A (en) 2016-08-12 2023-10-18 엘.이.에이.에프. 홀딩스 그룹 엘엘씨. Alpha and gamma-d polyglutamated antifolates and uses thereof
CN111954531A (en) 2018-02-07 2020-11-17 L.E.A.F.控股集团公司 Alpha polyglutamated pemetrexed and its use
CA3090387A1 (en) 2018-02-07 2019-08-15 L.E.A.F. Holdings Group Llc Alpha polyglutamated pralatrexate and uses thereof
EP3752157A4 (en) 2018-02-14 2022-07-06 L.E.A.F Holdings Group LLC Gamma polyglutamated lometrexol and uses thereof
BR112022013478A2 (en) * 2020-01-07 2022-09-13 Purdue Research Foundation TARGETED STEROID COMPOUNDS

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030026831A1 (en) * 2001-04-20 2003-02-06 Aparna Lakkaraju Anionic liposomes for delivery of bioactive agents
US6852334B1 (en) * 1999-04-20 2005-02-08 The University Of British Columbia Cationic peg-lipids and methods of use
US20110237686A1 (en) * 2010-03-26 2011-09-29 Cerulean Pharma Inc Formulations and methods of use
US20140179761A1 (en) * 2007-12-04 2014-06-26 Tekmira Pharmaceuticals Corporation Targeting lipids

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4721612A (en) 1984-04-12 1988-01-26 The Liposome Company, Inc. Steroidal liposomes
JPS6252188A (en) 1985-08-27 1987-03-06 エスケ−化研株式会社 Composition with high refractory properties
GB2304438A (en) 1995-08-17 1997-03-19 Kenneth Austin Re-configurable application specific device
AU2316997A (en) 1996-02-27 1997-09-16 David H Thompson Liposomal delivery system
US6056973A (en) 1996-10-11 2000-05-02 Sequus Pharmaceuticals, Inc. Therapeutic liposome composition and method of preparation
HUP0401127A3 (en) 2001-05-02 2006-03-28 Purdue Research Foundation Treatment and diagnosis of macrophage disease
US20050152962A1 (en) 2002-06-12 2005-07-14 Metselaar Josbert M. Composition for treatment of inflammatory disorders
EP1371362A1 (en) 2002-06-12 2003-12-17 Universiteit Utrecht Holding B.V. Composition for treatment of inflammatory disorders
CA2524441C (en) 2003-05-06 2012-03-20 Purdue Research Foundation Treatment of lupus targeting the macrophages or the folate receptor
JP5175723B2 (en) * 2005-07-05 2013-04-03 パーデュー・リサーチ・ファウンデーション Preparation of compositions for treating monocyte-mediated diseases
US20070041934A1 (en) * 2005-08-12 2007-02-22 Regents Of The University Of Michigan Dendrimer based compositions and methods of using the same
US20090311191A1 (en) 2008-06-13 2009-12-17 Ananth Annapragada Imaging of atherosclerotic plaques using liposomal imaging agents

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6852334B1 (en) * 1999-04-20 2005-02-08 The University Of British Columbia Cationic peg-lipids and methods of use
US20030026831A1 (en) * 2001-04-20 2003-02-06 Aparna Lakkaraju Anionic liposomes for delivery of bioactive agents
US20140179761A1 (en) * 2007-12-04 2014-06-26 Tekmira Pharmaceuticals Corporation Targeting lipids
US20110237686A1 (en) * 2010-03-26 2011-09-29 Cerulean Pharma Inc Formulations and methods of use

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Al-Muhammad et al., J. Microencapsulation, 1996, vol. 13, no. 2, 123-130. *
Rayburn et al., Mol Cell Pharmacol. 2009; 1(1):29-43 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160158152A1 (en) * 2013-07-17 2016-06-09 Council Of Scientific & Industrial Research Pharmaceutical composition for the treatment of diminution of bone tissue
US10596115B2 (en) * 2013-07-17 2020-03-24 Council Of Scientific & Industrial Research Pharmaceutical composition for the treatment of diminution of bone tissue
WO2015077303A1 (en) * 2013-11-19 2015-05-28 Purdue Research Foundation Patient selection method for inflammation
US11162937B2 (en) 2013-11-19 2021-11-02 Purdue Research Foundation Patient selection method for inflammation
CN108430457A (en) * 2015-12-17 2018-08-21 南洋理工大学 The nano liposomes and preparation method thereof comprising corticosteroid as drug
US20180360758A1 (en) * 2015-12-17 2018-12-20 Nanyang Technological University Nanoliposomes comprising corticosteroid as medicaments and methods to prepare them

Also Published As

Publication number Publication date
WO2011150392A1 (en) 2011-12-01
CA2800693A1 (en) 2011-12-01
US11185505B2 (en) 2021-11-30
US20200297632A1 (en) 2020-09-24
US20220054417A1 (en) 2022-02-24

Similar Documents

Publication Publication Date Title
US11185505B2 (en) Delivery of agents to inflamed tissues using folate-targeted liposomes
JP6612905B2 (en) Nanoparticles for mitochondrial transport of drugs
Lee et al. Nano-sized metabolic precursors for heterogeneous tumor-targeting strategy using bioorthogonal click chemistry in vivo
CA2834918C (en) Liposomes comprising polymer-conjugated lipids and related uses
Sou et al. Selective uptake of surface-modified phospholipid vesicles by bone marrow macrophages in vivo
ES2301871T3 (en) CORTICOSTEROIDS ENCAPSULATED IN VESICLES FOR THE TREATMENT OF CANCER.
BR112013005202B1 (en) TARGET LEGUMAIN LIPIDE, TARGET LEGUMAIN NANOPARTICLES COMPOSITION AND USES
US20240066144A1 (en) Bone marrow-, reticuloendothelial system-, and/or lymph node-targeted radiolabeled liposomes and methods of their diagnostic and therapeutic use
Aranda-Lara et al. Radiolabeled liposomes and lipoproteins as lipidic nanoparticles for imaging and therapy
Gouveia et al. Non-biologic nanodelivery therapies for rheumatoid arthritis
WO1997042166A1 (en) Amidine derivatives and drug carriers comprising them
US20150086611A1 (en) Liposome including complex of hydrophobic active ingredient and polypeptide and use of the liposome
US20030170299A1 (en) Therapeutic methods for acute myeloid leukemia
Isaac-Olivé et al. [99m Tc-HYNIC-N-dodecylamide]: a new hydrophobic tracer for labelling reconstituted high-density lipoproteins (rHDL) for radioimaging
ES2314210T3 (en) COMPOSITION FOR THE TREATMENT OF INFLAMMATORY DISORDERS.
JP2010505880A (en) pH-sensitive liposome composition
Ahmed et al. Synthesis, Characterization, and In Vivo Distribution of 99mTc Radiolabelled Docetaxel Loaded Folic Acid-Thiolated Chitosan Enveloped Liposomes
US20230414795A1 (en) Psma targeted compounds and uses thereof
Cvjetinović et al. 177Lu–labeled micro liposomes as a potential radiosynoviorthesis therapeutic agent
CN117860702A (en) Nucleic acid drug administration system and preparation method and application thereof
Lecommandoux et al. Polymeric micelles and vesicles: Biological behavior evaluation using radiolabeling techniques

Legal Events

Date Code Title Description
AS Assignment

Owner name: PURDUE RESEARCH FOUNDATION, INDIANA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LOW, PHILIP S.;POH, SCOTT YEW TAT;SIGNING DATES FROM 20121210 TO 20130115;REEL/FRAME:029677/0615

STCV Information on status: appeal procedure

Free format text: ON APPEAL -- AWAITING DECISION BY THE BOARD OF APPEALS

STCV Information on status: appeal procedure

Free format text: BOARD OF APPEALS DECISION RENDERED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STCB Information on status: application discontinuation

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