WO2007139815A2 - Imaging and therapeutic method using progenitor cells - Google Patents

Imaging and therapeutic method using progenitor cells Download PDF

Info

Publication number
WO2007139815A2
WO2007139815A2 PCT/US2007/012269 US2007012269W WO2007139815A2 WO 2007139815 A2 WO2007139815 A2 WO 2007139815A2 US 2007012269 W US2007012269 W US 2007012269W WO 2007139815 A2 WO2007139815 A2 WO 2007139815A2
Authority
WO
WIPO (PCT)
Prior art keywords
progenitor cells
folate
group
cells
endothelial progenitor
Prior art date
Application number
PCT/US2007/012269
Other languages
French (fr)
Other versions
WO2007139815A3 (en
Inventor
Philip Stewart Low
Andrew Richard Hilgenbrink
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 US12/301,864 priority Critical patent/US20100226967A1/en
Publication of WO2007139815A2 publication Critical patent/WO2007139815A2/en
Publication of WO2007139815A3 publication Critical patent/WO2007139815A3/en
Priority to US13/910,306 priority patent/US20130266964A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • 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
    • 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
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/6415Toxins or lectins, e.g. clostridial toxins or Pseudomonas exotoxins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/65Peptidic linkers, binders or spacers, e.g. peptidic enzyme-labile linkers
    • 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
    • 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
    • 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
    • A61K49/0052Small organic molecules
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/82Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving vitamins or their receptors
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/10Screening for compounds of potential therapeutic value involving cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases

Definitions

  • This invention relates to methods for treating and diagnosing disease states worsened by progenitor cells. More particularly, ligands that bind to progenitor cells are complexed with a quantifiable marker for use in diagnosis or to an antigen, a cytotoxin, or an agent for altering progenitor cell function for use in the treatment of disease states worsened by progenitor cells.
  • Applicants have also undertaken to determine whether a quantifiable marker linked to a ligand capable of binding to progenitor cells, such as CDl 33 + FIkI + endothelial progenitor cells or common precursor cells for both endothelial progenitor cells and macrophages, may be useful for diagnosing inflammatory pathologies, and other pathologies that involve vasculogenesis.
  • a quantifiable marker linked to a ligand capable of binding to progenitor cells such as CDl 33 + FIkI + endothelial progenitor cells or common precursor cells for both endothelial progenitor cells and macrophages.
  • a method for diagnosing a disease state worsened by CDl 33 + FIkI + endothelial progenitor cells.
  • the method comprises the steps of isolating CDl 33 + FIkI + endothelial progenitor cells from a patient suffering from a disease state worsened by CDl 33 + FIkI + endothelial progenitor cells, contacting the endothelial progenitor cells with a composition comprising a conjugate or complex of the general formula
  • the progenitor cells can be common progenitor cells for both endothelial progenitor cells and macrophages.
  • the quantifiable marker can be, for example, a radioactive probe, a fluorescent probe, an enzyme capable of amplifying a signal, an antibody capable of assisting in amplifying a signal, or other agents for use in amplifying a signal, such as oligonucleotides.
  • a method is provided for treating a disease state worsened by CD133 + FIkI + endothelial progenitor cells.
  • a compound for diagnosing or treating a disease state worsened by progenitor cells such as CDl 33 + FIkI + endothelial progenitor cells or common progenitor cells for both endothelial progenitor cells and macrophages.
  • the compound is selected from the following group of compounds:
  • a method of quantifying endothelial progenitor cells comprises the steps of isolating the progenitor cells from a patient suffering from a disease state mediated by the progenitor cells, contacting the progenitor cells with a composition comprising a conjugate or complex of the general formula Ab-X where the group Ab comprises a vitamin, or an analog thereof, that binds to endothelial progenitor cells and the group X comprises a quantifiable marker, and quantifying the percentage of progenitor cells that expresses a receptor for the vitamin.
  • a use is provided of a composition comprising a conjugate or complex of the general formula Ab-X where the group A b comprises a vitamin, or an analog thereof, that binds to the progenitor cells and the group X comprises an antigen, a cytotoxin, or a compound capable of altering progenitor cell function in the manufacture of a medicament for use in treating a disease state worsened by progenitor cells.
  • a method of quantifying endothelial progenitor cells comprises the steps of contacting the progenitor cells in a patient suffering from a disease state mediated by the progenitor cells with a composition comprising a conjugate or complex of the general formula Ab-X where the group Ab comprises a vitamin, or an analog thereof, that binds to endothelial progenitor cells and the group X comprises a quantifiable marker, and quantifying the percentage of progenitor cells that expresses a receptor for the vitamin.
  • Fig. 1 shows flow cytometry analysis, using Flkl (A), CDl 15 (B), CD69 (C) 5 CDl Ib (D), CD8a (E), and CD25 (F) antibodies and folate-FITC, of markers that are co-expressed with the folate receptor on CD133 + FIkI + endothelial progenitor cells or on common precursor cells to both endothelial progenitor cells and macrophages.
  • Fig. 2 shows flow cytometry analysis, using CD62L (A), CD80 (B), CD86 (C), CD44 (D), CD23 (E), and CD14 (F) antibodies and folate-FITC, of markers that are co-expressed with the folate receptor on CDl 33 + FIkI + endothelial progenitor cells.
  • Fig. 3 shows flow cytometry analysis, using Ly-6 (A), F4/80 (B), CD49d (C), CDl 6.2/32.2 (D), and MHC Class II (E) antibodies and folate-FITC, of markers that are co-expressed with the folate receptor on CDl 33 + FIkI + endothelial progenitor cells or on common precursor cells to both endothelial progenitor cells and macrophages.
  • Fig. 4 shows folate-fluorescein (folate-FITC) binding, quantified by flow cytometry, to CD133 + endothelial progenitor cells (panel A), to FIk-I + endothelial progenitor cells (Panel B), and to CD44 + endothelial progenitor cells (panel C) without excess unlabeled folic acid (top panels) or preincubated with an excess of unlabeled folic acid (bottom panels (Competed Samples)) to compete with folate-FITC for binding.
  • fluorescein folate-FITC
  • Fig. 5 shows folate-fluorescein (folate-FITC) binding, quantified by flow cytometry, to Ly-6 + endothelial progenitor cells (panel A), to CD25 + endothelial progenitor cells (Panel B), and to CD62-L + endothelial progenitor cells (panel C) without excess unlabeled folic acid (top panels) or preincubated with an excess of unlabeled folic acid (bottom panels (Competed Samples)) to compete with folate- FITC for binding.
  • folate-FITC folate-fluorescein
  • progenitor cells such as CD133 4" FIk-I + endothelial progenitor cells or common precursor cells to both endothelial progenitor cells and macrophages (i.e., referred to in this application as "common precursor cells").
  • Exemplary disease states include fibromyalgia, rheumatoid arthritis, osteoarthritis, ulcerative colitis, Crohn's disease, psoriasis, osteomyelitis, multiple sclerosis, atherosclerosis, pulmonary fibrosis, sarcoidosis, systemic sclerosis, organ transplant rejection (GVHD), lupus erythematosus, Sjogren's syndrome, glomerulonephritis, inflammations of the skin (e.g., psoriasis), cancer, proliferative retinopathy, restenosis, and chronic inflammations.
  • GVHD organ transplant rejection
  • lupus erythematosus e.g., Sjogren's syndrome
  • glomerulonephritis inflammations of the skin (e.g., psoriasis), cancer, proliferative retinopathy, restenosis, and chronic inflammations.
  • Such disease states can be diagnosed by isolating the progenitor cells from a patient suffering from such disease state, contacting the cells with a composition comprising a conjugate of the general formula A b -X wherein the group A b comprises a ligand that binds to the progenitor cells, and the group X comprises a quantifiable marker, and quantifying the percentage of the progenitor cells expressing a receptor for the ligand.
  • progenitor cells includes CD 133 and/or FIkI + endothelial progenitor cells and common precursor cells for both endothelial progenitor cells and macrophages.
  • common precursor cells refers to common precursor cells for both endothelial progenitor cells and macrophages. These cells have CDl 33 and/or Flkl markers and also have a marker selected from the group consisting of CDl Ib, F4/80, and CDl 15.
  • the terms “eliminated” and “eliminating” in reference to the disease state mean reducing the symptoms or eliminating the symptoms of the disease state or preventing the progression or the reoccurrence of disease.
  • the terms “elimination” and “deactivation” of the progenitor cell population that expresses the ligand receptor mean that this progenitor cell population is killed or is completely or partially inactivated which reduces the pathogenesis characteristic of the disease state being treated.
  • endothelial progenitor cells can directly cause disease or can augment disease states such as by stimulating other immune cells to secrete factors that worsen disease states, such as by stimulating T-cells to secrete TNF-o; or by increasing the blood supply (e.g., by vasculogenesis) to pathologic tissues, such as cancer tissues.
  • endothelial progenitor cells themselves may also harbor infections and cause disease and infected progenitor cells may cause other immune cells to secrete factors that cause disease such as TNF- ⁇ secretion by T-cells.
  • Such disease states can also be diagnosed by administering parenterally to a patient a composition comprising a conjugate or complex of the general formula A b -X where the group A b comprises a ligand that binds to progenitor cells and the group X comprises a quantifiable marker, and quantifying the percentage of the cells that expresses a receptor for the ligand.
  • CD133 + FIk-I + endothelial progenitor cell-worsened disease states can be treated in accordance with the methods disclosed herein by administering an effective amount of a composition A b -X wherein Ab comprises a ligand that binds to CD133 + FIk-I + endothelial progenitor cells and wherein the group X comprises an antigen, a cytotoxin, or a compound capable of altering the function of the endothelial progenitor cells.
  • Such targeting conjugates when administered to a patient suffering from a disease state augmented by the endothelial progenitor cells, work to concentrate and associate the conjugated cytotoxin, antigen, or compound capable of altering endothelial progenitor cell function with the population of endothelial progenitor cells to kill the cells or alter cell function.
  • the conjugate is typically administered parenterally, but can be delivered by any suitable method of administration (e.g., orally), as a composition comprising the conjugate and a pharmaceutically acceptable carrier therefor.
  • Conjugate administration is typically continued until symptoms of the disease state are reduced or eliminated, or administration is continued after this time to prevent progression or reappearance of the disease.
  • the cells may be common precursor cells in similar embodiments and in these embodiments ligands that bind to common precursor cells can be used.
  • disease states worsened by progenitor cells are diagnosed in a patient by isolating the cells from the patient, contacting the progenitor cells with a conjugate A b -X wherein Ab comprises a ligand that binds to the progenitor cells and X comprises a quantifiable marker, and quantifying the percentage of progenitor cells expressing the receptor for the ligand.
  • the diagnostic conjugates can be administered to the patient as a diagnostic composition comprising a conjugate and a pharmaceutically acceptable carrier and thereafter the progenitor cells can be collected from the patient to quantify the percentage of cells expressing the receptor for the ligand A b .
  • the composition is typically formulated for parenteral administration and is administered to the patient in an amount effective to enable quantification of the progenitor cells.
  • disease states can also be diagnosed by administering parenterally to a patient a composition comprising a conjugate or complex of the general formula A b -X where the group A b comprises a ligand that binds to progenitor cells and the group X comprises a quantifiable marker, and quantifying the percentage of the cells that expresses a receptor for the ligand.
  • the quantifiable marker e.g., a reporter molecule
  • the quantifiable marker can comprise a radiolabeled compound such as a chelating moiety and an element that is a radionuclide, for example a metal cation that is a radionuclide.
  • the radionuclide is selected from the group consisting of technetium, gallium, indium, and a positron emitting radionuclide (PET imaging agent).
  • Diagnosis typically occurs before treatment.
  • diagnosis can also mean monitoring of the disease state before, during, or after treatment to determine the progression of the disease state.
  • the monitoring can occur before, during, or after treatment, or combinations thereof, to determine the efficacy of therapy, or to predict future episodes of disease.
  • the quantification can be performed by any suitable method known in the art, including imaging methods, such as intravital imaging.
  • the method disclosed herein can be used for both human clinical medicine and veterinary applications.
  • the host animal afflicted with the disease state worsened by progenitor cells and in need of diagnosis or therapy can be a human, or in the case of veterinary applications, can be a laboratory, agricultural, domestic or wild animal.
  • the conjugates can be administered parenterally to the animal or patient suffering from the disease state, for example, intradermally, subcutaneously, intramuscularly, intraperitoneally, or intravenously.
  • the conjugates can be administered to the animal or patient by other medically useful procedures and effective doses can be administered in standard or prolonged release dosage forms, such as a slow pump.
  • the therapeutic method described herein can be used alone or in combination with other therapeutic methods recognized for the treatment of inflammatory disease states, or disease states augmented by vasculogenesis.
  • the group Ab is a ligand that binds to CD133 + FIk-I + endothelial progenitor cells or common precursor cells when the conjugates are used to diagnose or treat disease states.
  • Any of a wide number of binding ligands can be employed.
  • Acceptable ligands include particularly folate receptor binding ligands, and analogs thereof, and antibodies or antibody fragments capable of recognizing and binding to surface moieties expressed or presented on CDl 33 + FIk-I + endothelial progenitor cells or on common precursor cells.
  • Antagonists and agonists for CD133, Flkl, CDl Ib, F4/80, or CDl 15 may be acceptable ligands.
  • the binding ligand can be folic acid, a folic acid analog, or another folate receptor-binding molecule.
  • Analogs of folate that can be used include folinic acid, pteropolyglutamic acid, and folate receptor-binding pteridines such as tetrahydropterins, dihydrofolates, tetrahydrofolates, and their deaza and dideaza analogs.
  • the terms “deaza” and “dideaza” analogs refers to the art recognized analogs having a carbon atom substituted for one or two nitrogen atoms in the naturally occurring folic acid structure.
  • the deaza analogs include the 1 -deaza, 3-deaza, 5-deaza, 8-deaza, and 10-deaza analogs.
  • the dideaza analogs include, for example, 1,5 dideaza, 5,10-dideaza, 8,10-dideaza, and 5,8-dideaza analogs.
  • the foregoing folic acid analogs are conventionally termed "folates," reflecting their capacity to bind to folate receptors.
  • folate receptor-binding analogs include ammopterin, amethopterin (methotrexate), N 10 -methylfolate, 2- deamino-hydroxyfolate, deaza analogs such as 1 -deazamethopterin or 3- deazamethopterin, and 3',5'-dichloro-4-amino-4-deoxy-N 10 -methylpteroylglutamic acid (dichloromethotrexate).
  • vitamins can be used as the binding ligand.
  • the vitamins that can be used in accordance with the methods described herein include niacin, pantothenic acid, folic acid, riboflavin, thiamine, biotin, vitamin B i 2 , vitamins A, D, E and K, other related vitamin molecules, analogs and derivatives thereof, and combinations thereof.
  • the binding ligand can be any ligand that binds to a receptor expressed or overexpressed on endothelial progenitor cells or common precursor cells including CD133, Flkl, CDl Ib, CDl 15, CD69, CD8a, CD25, CD62L, CD80, CD86, CD44, CD23, CD14, Ly-6, F4/80, CD49d, CDl 6.2/32.2, and the like.
  • ligands include both antagonists and agonists for each of the above membrane-spanning proteins.
  • the targeted conjugates used for diagnosing or treating disease states mediated by progenitor cells have the formula Ab-X, wherein Ab is a ligand capable of binding to the progenitor cells, and the group X comprises a quantifiable marker or an antigen (such as an immunogen), cytotoxin, or a compound capable of altering progenitor cell function.
  • the group A b is folic acid, a folic acid analog, or another folic acid receptor binding ligand
  • these conjugates are described in detail in U.S. Patent No. 5,688,488, the specification of which is incorporated herein by reference. That patent, as well as related U.S. Patents Nos.
  • the progenitor cells can be activated cells and the group Ab can be any of the ligands described above.
  • cytotoxic moieties useful for forming conjugates for use in accordance with the methods described herein are clodronate, anthrax, Pseudomonas exotoxin, typically modified so that these cytotoxic moieties do not bind to normal cells, and other toxins or cytotoxic agents including art- recognized chemotherapeutic agents such as adrenocorticoids, alkylating agents, antiandrogens, antiestrogens, androgens, estrogens, antimetabolites such as cytosine arabinoside, purine analogs, pyrimidine analogs, and methotrexate, busulfan, carboplatin, chlorambucil, cisplatin and other platinum compounds, tamoxiphen, taxol, cyclophosphamide, plant alkaloids, prednisone, hydroxyurea, teniposide
  • Such toxins or cytotoxic compounds can be directly conjugated to the targeting ligand, for example, folate or another folate receptor- binding ligand, or they can be formulated in liposomes or other small particles which themselves are targeted as conjugates of the progenitor cell-binding ligand typically by covalent linkages to component phospholipids.
  • the group X comprises a compound capable of altering progenitor cell function, for example, a cytokine such as IL-IO or IL-11
  • the compound can be covalently linked to the targeting ligand Ab, for example, a folate receptor-binding ligand or a progenitor cell-binding antibody or antibody fragment directly, or the function altering compound can be encapsulated in a liposome which is itself targeted to progenitor cells by pendent targeting ligands A b covalently linked to one or more liposome components.
  • conjugates Ab-X where X is an antigen or a compound capable of altering progenitor cell function can be administered in combination with a cytotoxic compound.
  • cytotoxic compounds listed above are among the compounds suitable for this purpose.
  • the group X in the targeted conjugate Ab-X comprises an antigen (i.e., a compound that elicits an immune response in vivo), the ligand-antigen conjugates being effective to "label" the population of progenitor cells responsible for disease pathogenesis in the patient suffering from the disease for specific elimination by an endogenous immune response or by co-administered antibodies.
  • an antigen i.e., a compound that elicits an immune response in vivo
  • the ligand-antigen conjugates being effective to "label" the population of progenitor cells responsible for disease pathogenesis in the patient suffering from the disease for specific elimination by an endogenous immune response or by co-administered antibodies.
  • the endogenous immune response can include a humoral response, a cell-mediated immune response, and any other immune response endogenous to the host animal, including complement-mediated cell lysis, antibody-dependent cell- mediated cytotoxicity (ADCC), antibody opsonization leading to phagocytosis, clustering of receptors upon antibody binding resulting in signaling of apoptosis, antiproliferation, or differentiation, and direct immune cell recognition of the delivered antigen (e.g., a hapten). It is also contemplated that the endogenous immune response may employ the secretion of cytokines that regulate such processes as the multiplication, differentiation, and migration of immune cells.
  • the endogenous immune response may include the participation of such immune cell types as B cells, T cells, including helper and cytotoxic T cells, macrophages, natural killer cells, neutrophils, LAK cells, and the like.
  • the humoral response can be a response induced by such processes as normally scheduled vaccination, or active immunization with a natural antigen or an unnatural antigen or hapten, e.g., fluorescein isothiocyanate (FITC), with the unnatural antigen inducing a novel immunity.
  • Active immunization involves multiple injections of the unnatural antigen or hapten scheduled outside of a normal vaccination regimen to induce the novel immunity.
  • the humoral response may also result from an innate immunity where the host animal has a natural preexisting immunity, such as an immunity to ⁇ -galactosyl groups.
  • a passive immunity may be established by administering antibodies to the host animal such as natural antibodies collected from serum or monoclonal antibodies that may or may not be genetically engineered antibodies, including humanized antibodies.
  • antibodies to the host animal such as natural antibodies collected from serum or monoclonal antibodies that may or may not be genetically engineered antibodies, including humanized antibodies.
  • the utilization of a particular amount of an antibody reagent to develop a passive immunity, and the use of a ligand-antigen conjugate wherein the passively administered antibodies are directed to the antigen, would provide the advantage of a standard set of reagents to be used in cases where a patient's preexisting antibody titer to potential antigens is not therapeutically useful.
  • the passively administered antibodies may be "co-administered" with the ligand- antigen conjugate, and co-administration is defined as administration of antibodies at a time prior to, at the same time as, or at a time following administration of the ligand-antigen conjugate.
  • the preexisting antibodies, induced antibodies, or passively administered antibodies will be redirected to the progenitor cells by preferential binding of the ligand-antigen conjugates to the progenitor cell populations, and such pathogenic cells are killed by complement-mediated lysis, ADCC, antibody- dependent phagocytosis, or antibody clustering of receptors.
  • the cytotoxic process may also involve other types of immune responses, such as cell-mediated immunity.
  • Acceptable antigens for use in preparing the conjugates used in the method of treatment described herein are antigens that are capable of eliciting antibody production in a host animal or that have previously elicited antibody production in a host animal, resulting in a preexisting immunity, or that constitute part of the innate immune system.
  • antibodies directed against the antigen may be administered to the host animal to establish a passive immunity.
  • Suitable antigens for use in the invention include antigens or antigenic peptides against which a preexisting immunity has developed via normally scheduled vaccinations or prior natural exposure to such agents such as polio virus, tetanus, typhus, rubella, measles, mumps, pertussis, tuberculosis and influenza antigens, and ⁇ -galactosyl groups.
  • the ligand-antigen conjugates will be used to redirect a previously acquired humoral or cellular immunity to a population of progenitor cells in the host animal for elimination of the progenitor cells.
  • suitable immunogens include antigens or antigenic peptides to which the host animal has developed a novel immunity through immunization against an unnatural antigen or hapten, for example, fluorescein isothiocyanate (FITC) or dinitrophenyl, and antigens against which an innate immunity exists, for example, super antigens and muramyl dipeptide.
  • FITC fluorescein isothiocyanate
  • FITC fluorescein isothiocyanate
  • dinitrophenyl for example, antigens against which an innate immunity exists, for example, super antigens and muramyl dipeptide.
  • the progenitor cell-binding ligands and antigens, cytotoxic agents, compounds capable of altering progenitor cell function, or imaging agents, as the case may be in forming conjugates for use in accordance with the methods described herein can be conjugated by using any art-recognized method for forming a complex. This can include covalent, ionic, or hydrogen bonding of the ligand to the antigen, either directly or indirectly via a linking group such as a divalent linker.
  • the conjugate is typically formed by covalent bonding of the ligand to the targeted entity through the formation of amide, ester or imino bonds between acid, aldehyde, hydroxy, amino, or hydrazo groups on the respective components of the complex or, for example, by the formation of disulfide bonds.
  • Methods of linking binding ligands to antigens, cytotoxic agents, compounds capable of altering progenitor cell function, or quantifiable markers are described in U.S. Patent Application Publication No. US 2005/0002942-A1 and PCT Publication No. WO 2006/012527, each incorporated herein by reference.
  • the ligand complex can be one comprising a liposome wherein the targeted entity (that is, the quantifiable marker, or the antigen, cytotoxic agent or progenitor cell function-altering agent) is contained within a liposome which is itself covalently linked to the binding ligand.
  • the targeted entity that is, the quantifiable marker, or the antigen, cytotoxic agent or progenitor cell function-altering agent
  • Other nanoparticles, dendrimers, derivatizable polymers or copolymers that can be linked to therapeutic or quantifiable markers useful in the treatment and diagnosis of progenitor cell-worsened diseases can also be used in targeted conjugates.
  • the ligand is folic acid, an analog of folic acid, or any other folate receptor binding molecule
  • the folate ligand is conjugated to the targeted entity by a procedure that utilizes trifiuoroacetic anhydride to prepare ⁇ -esters of folic acid via a pteroyl azide intermediate.
  • This procedure results in the synthesis of a folate ligand, conjugated to the targeted entity only through the ⁇ -carboxy group of the glutamic acid groups of folate.
  • folic acid analogs can be coupled through the ⁇ -carboxy moiety of the glutamic acid group or both the o: and ⁇ carboxylic acid entities.
  • the therapeutic methods described herein can be used to slow the progress of disease completely or partially. Alternatively, the therapeutic methods described herein can eliminate or prevent reoccurrence of the disease state.
  • the conjugates used in accordance with the methods described herein of the formula A b -X are used in one aspect to formulate therapeutic or diagnostic compositions, for administration to a patient, wherein the compositions comprise effective amounts of the conjugate and an acceptable carrier therefor. Typically such compositions are formulated for parenteral use.
  • the amount of the conjugate effective for use in accordance with the methods described herein depends on many parameters, including the nature of the disease being treated or diagnosed, the molecular weight of the conjugate, its route of administration and its tissue distribution, and the possibility of co-usage of other therapeutic or diagnostic agents.
  • the effective amount to be administered to a patient is typically based on body surface area, patient weight and physician assessment of patient condition.
  • An effective amount can range from about to 1 ng/kg to about 1 mg/kg, more typically from about 1 ⁇ g/kg to about 500 ⁇ g/kg, and most typically from about 1 ⁇ g/kg to about 100 ⁇ g/kg.
  • the ligand conjugates can be administered as single doses, or they can be divided and administered as a multiple-dose daily regimen.
  • a staggered regimen for example, one to three days per week can be used as an alternative to daily treatment, and such an intermittent or staggered daily regimen is considered to be equivalent to every day treatment and within the scope of this disclosure.
  • the patient is treated with multiple injections of the ligand conjugate wherein the targeted entity is an antigen or a cytotoxic agent or a compound capable of altering progenitor cell function to eliminate the population of pathogenic progenitor cells.
  • the patient is treated, for example, injected multiple times with the ligand conjugate at, for example, 12-72 hour intervals or at 48-72 hour intervals. Additional injections of the ligand conjugate can be administered to the patient at intervals of days or months after the initial injections, and the additional injections prevent recurrence of disease. Alternatively, the ligand conjugates may be administered prophylactically to prevent the occurrence of disease in patients known to be disposed to development of disease states worsened by progenitor cells.
  • more than one type of ligand conjugate can be used, for example, the host animal may be pre-immunized with fluorescein isothiocyanate and dinitrophenyl and subsequently treated with fluorescein isothiocyanate and dinitrophenyl linked to the same or different targeting ligands in a co-dosing protocol.
  • the progenitor cells can be activated cells or other cell populations that augment or cause disease states.
  • the following examples are illustrative embodiments only and are not intended to be limiting.
  • EXAMPLE 1 MATERIALS Fmoc-protected amino acid derivatives, trityl-protected cysteine 2- chlorotrityl resin (H-Cys(Trt)-2-ClTrt resin #04-12-2811), Fmoc-lysine(4- methyltrityl) wang resin, 2-(lH-benzotriaxol-l-yl)-l,l,3,3-tetramethyluronium hexafluorophosphage (HBTU) and N-hydroxybenzotriazole were purchased from Novabiochem (La Jolla, CA). N 10 -trifluoroacetylpteroic acid was purchased from Sigma, St. Louis, MO.
  • Piperidine, DIPEA (diisopropylethylamine), Rhodamine B isothiocyanate (Rd-ITC) and triisopropyl saline (TIPS) were from Aldrich (Milwaukee).
  • Anti-mouse antibodies were purchased from Caltag Laboratories, Burlingame, CA. The following anti-mouse antibodies were purchased from Caltag Laboratories: CDlIb, CD16.2/32.2, CD23, CD44, CD49d, CD62L, CD69, CD80, CD86, F4/80, Ly-6C/G, I-A b MHC Class II and Streptavidin secondary fluorescence tag.
  • Fmoc peptide chemistry was used to synthesize folate- cysteine with the cysteine attached to the 7-COOH of folic acid.
  • the sequence Cys- Glu-Pteroic acid (Folate-Cys) was constructed by Fmoc chemistry with HBTU and N- hydroxybenzotriazole as the activating agents along with diisopropyethylamine as the base and 20% piperidine in dimethylformamide (DMF) for deprotection of the Fmoc groups.
  • DMF dimethylformamide
  • N- ⁇ -Fmoc-L-glutamic acid was linked to a trityl- protected Cys linked to a 2-Chlorotrityl resin.
  • N I0 -trifluoroacetylpteroic acid was then attached to the 7-COOH of GIu.
  • the Folate-Cys was cleaved from the resin using a 92.5% trifluoroacetic acid-2.5% water-2.5% triisopropylsilane- 2.5% ethanedithio solution. Diethyl ether was used to precipitate the ⁇ product, and the precipitant was collected by centrifugation.
  • Folate-FITC was synthesized as described by Kennedy, M.D. et al. in Pharmaceutical Research, Vol. 20(5); 2003.
  • Fmoc peptide chemistry was used to synthesize folate- aspartate-arginine-aspartate-aspartate-cysteine (Folate-Asp-Arg-Asp-Asp-Cys, Folate- D-R-D-D-C) with the amino acid spacer attached to the 7-COOH of folic acid.
  • the sequence Cys-Asp-Asp-Arg-Asp-Glu-Pteroic acid was constructed by Fmoc chemistry with HBTU and N-hydroxybenzotriazole as the activating agents along with diisopropyethylamine as the base and 20% piperidine in dimethylforrnamide (DMF) for deprotection of the Fmoc groups.
  • Fmoc-D- Asp(OtBu)-OH was linked to a trityl-protected Cys linked to a 2-Chlorotrityl resin.
  • the mobile phase consisting of 1OmM NH4HCO3 buffer, pH 7.0 (eluent A) and acetonitrile (eluent B), was maintained at a 99: 1 A:B ratio for the first five minutes and then changed to 70:30 A:B in a linear gradient over the next 30 minutes.
  • the recovered final product was confirmed by mass spectrometry.
  • Example 9 except that diclofenac was used in place of indomethicin.
  • n 1, 2, or 3, and where n is illustratively 2.
  • the folate glucocorticoid conjugate of prednisolone was prepared as follows. A 1.1 molar equivalent of prednisone was dissolved in tetrahydrofuran (THF). In a separate vial, a 0.7 molar equivalent of dimethylaminopyridine, 1 molar equivalent of tri(hydroxyethyl)amine and 1 molar equivalent of the linker (synthesis described in PCT Publication No. WO 2006/012527, incorporated herein by reference) were dissolved in dichloromethane. An approximately equal volume of both solutions were combined, mixed and reacted at room temperature for 4 hours.
  • THF tetrahydrofuran
  • the reaction was monitored by thin layer chromatography using 40: 10: 1 (Dichloromethane: Acetonitrile: Methanol).
  • the product was purified on a silica column (Silica 32-63, 6 ⁇ A) using the same ratio of solvents.
  • the recovered product was dried in preparation for conjugation to a folate- peptide.
  • the derivatized glucocorticoid was dissolved in DMSO, to which was added a 1.5 molar equivalent of either the folate-cys or folate- Asp- Arg- Asp- Asp-Cys peptide.
  • the resulting solution was reacted for 3 hours at room temperature followed by purification using a HPLC reverse-phase Cl 8 column at a flow rate of lml/min.
  • the mobile phase consisting of 1OmM NH 4 HCO3 buffer, pH 7.0 (eluent A) and acetonitrile (eluent B), was maintained at a 99: 1 A:B ratio for the first minute and then changed to 1:99 A:B in a linear gradient over the next 39 minutes.
  • the folate- glucocorticoid conjugate eluted at approximately 26 minutes.
  • the recovered final product was confirmed by mass spectrometry.
  • Folate-cys-dexamethasone was synthesized by a procedure similar to that described in Example 11 except that the glucocorticoid was dexamethasone.
  • Folate-cys-flumethasone was synthesized by a procedure similar to that described in Example 11 except that the glucocorticoid was flumethasone.
  • mice Female 6- to 8-week-old BALB/c mice were injected in the peritoneal cavity with either Complete Freund's Adjuvant (CFA; 50-100 ⁇ L), Pseudomonas aeruginosa (IxIO 7 CFU (colony forming units)), or Yersinia enterocolitica (IxIO 6 CFU). Cells were isolated from the peritoneal cavity by lavage with 8 mL of sterile phosphate-buffered saline (PBS) 2-4 days later.
  • CFA Complete Freund's Adjuvant
  • IxIO 7 CFU colony forming units
  • IxIO 6 CFU Yersinia enterocolitica
  • the cells were pelleted by centrifugation (400 x g, 10 minutes at room temperature) and resuspended in folate- deficient RPMI-1640 media (FD-RPMI; Gibco) containing 10% heat-inactivated fetal bovine serum (FBS), penicillin (100 IU/mL) and streptomycin (100 ⁇ g/mL). Peritoneal extracted cells were seeded at densities of 1 x 10 6 cells/microcentrifuge tube for antibody and folate conjugate studies.
  • FD-RPMI-1640 media containing 10% heat-inactivated fetal bovine serum (FBS), penicillin (100 IU/mL) and streptomycin (100 ⁇ g/mL).
  • FBS heat-inactivated fetal bovine serum
  • penicillin 100 IU/mL
  • streptomycin 100 ⁇ g/mL
  • the desired product was isolated from the reaction mixture by preparative HPLC as described above.
  • the final conjugation was performed by mixing excess DIPEA with 2 (in DMSO) followed by addition of BODIPY FL NHS ester (Molecular Probes, Eugene, OR).
  • Compound 3 was then isolated from this reaction mixture by preparative HPLC.
  • Two laser sources with 543nm (He-Ne) and 488nm (Argon) wavelength can be used to excite BODIPY FL and rhodamine separately to obtain two color images when needed.
  • Confocal images can be acquired with a size of 512 x 512 pixels at 2.7 second scan time and images can be processed using FluoView (Olympus) software.
  • the proteins momordin and gelonin were purchased from Sigma (St. Louis, MO). Folate-cys pyridyldisulfide was prepared by reacting folate-cys with Aldrithiol (Sigma, St. Louis, MO). Both proteins were dissolved in 0.1M HEPPS buffer, pH 8.2. A 6-fold molar excess of Trouts reagent (Aldrich St. Louis, MO) dissolved in DMSO (16 mM) was added to each protein solution. The solutions were allowed to react for 1 hour at room temperature. Unreacted material was separated from the protein using a Sephadex G-25 column equilibrated in 0.1M phosphate buffer, pH 7.0.
  • FOLATE-FITC BINDING TO ENDOTHELIAL PROGENITOR CELLS Folate-FITC binding to CD133 + FIkI + endothelial progenitor cells and binding of antibodies to CD62L, CD80, CD86, CD44, CD23, and CD14 markers on endothelial progenitor cells was quantified. Endothelial progenitor cells were isolated as described in Example 14 and folate-FITC and antibody binding and flow cytometry were performed as described in Example 15. As shown in Fig. 2, CD62L, CD80, CD86, CD23, and CD 14 markers are co-expressed with the folate receptor on CD133 + FIkI + endothelial progenitor cells.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Molecular Biology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Analytical Chemistry (AREA)
  • Pathology (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • General Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Toxicology (AREA)
  • Dispersion Chemistry (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The invention relates to a method of treating or diagnosing a disease state worsened by progenitor cells. The method utilizes a composition comprising a conjugate or complex of the general formula Ab-X wherein the group Ab comprises a vitamin, or an analog thereof, that binds to progenitor cells, and when the conjugate is being used for treatment of the disease state, the group X comprises an antigen, a cytotoxin, or a compound capable of altering progenitor cell function, and when the conjugate is being used for diagnosing the disease state, the group X comprises a quantifiable marker.

Description

IMAGING AND THERAPEUTIC METHOD USING PROGENITOR CELLS
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims priority under 35 U.S.C. § 119(e) to U.S. Provisional Application No. 60/802,648, filed May 23, 2006, incorporated herein by reference in its entirety.
FIELD OF THE INVENTION
This invention relates to methods for treating and diagnosing disease states worsened by progenitor cells. More particularly, ligands that bind to progenitor cells are complexed with a quantifiable marker for use in diagnosis or to an antigen, a cytotoxin, or an agent for altering progenitor cell function for use in the treatment of disease states worsened by progenitor cells.
BACKGROUND
The mammalian immune system provides a means for the recognition and elimination of foreign pathogens. While the immune system normally provides a line of defense against foreign pathogens, there are many instances where the immune response itself is involved in the progression of disease. Exemplary of diseases caused or worsened by the host's own immune response are autoimmune diseases and other diseases in which the immune response contributes to pathogenesis. For example, macrophages are generally the first cells to encounter foreign pathogens, and accordingly, they play an important role in the immune response, but activated macrophages can also contribute to the pathophysiology of disease in some instances. The folate receptor is a 38 KD GPI-anchored protein that binds the vitamin folic acid with high affinity (< 1 nM). Following receptor binding, rapid endocytosis delivers a substantial fraction of the vitamins into the cell, where they are unloaded in an endosomal compartment at low pH. Importantly, covalent conjugation of small molecules, proteins, and even liposomes to folic acid does not block the vitamin's ability to bind the folate receptor, and therefore, folate-drug conjugates can readily be delivered to and can enter cells by receptor-mediated endocytosis.
Because most cells use an unrelated reduced folate carrier to acquire the necessary folic acid, expression of the folate receptor is restricted to a few cell types. With the exception of kidney, choroid plexus, and placenta, normal tissues express low or nondetectable levels of the folate receptor. However, many malignant tissues, including ovarian, breast, bronchial, and brain cancers express significantly elevated levels of the receptor. In fact, it is estimated that 95% of all ovarian carcinomas overexpress the folate receptor. It has been reported that the folate receptor β, the nonepithelial isoform of the folate receptor, is expressed on activated (but not resting) synovial macrophages. Thus, folate receptors are expressed on a subset of macrophages (i.e., activated macrophages).
SUMMARY
It is unknown, however, whether folate receptors are expressed on progenitor cells such as CD133+ FIkI+ cells commonly referred to as endothelial progenitor cells, or on common progenitor cells for both endothelial progenitor cells and macrophages. Thus, Applicants have undertaken to determine whether folate receptors are expressed on these progenitor cells and whether progenitor cell targeting, using a ligand such as folate, to deliver cytotoxic or other inhibitory compounds to these cells, is useful therapeutically. Applicants have also undertaken to determine whether a quantifiable marker linked to a ligand capable of binding to progenitor cells, such as CDl 33+ FIkI+ endothelial progenitor cells or common precursor cells for both endothelial progenitor cells and macrophages, may be useful for diagnosing inflammatory pathologies, and other pathologies that involve vasculogenesis.
A method is provided for treating and diagnosing disease states worsened by progenitor cells. In one embodiment, the progenitor cells are CDl 33+ FIkI+ endothelial progenitor cells. In another embodiment, the CD133+ FIkI+ cells are activated progenitor cells. In one embodiment, disease states worsened by CDl 33+ FIkI+ endothelial progenitor cells are treated by delivering an antigen to the cells, by linking the antigen to a ligand that binds to these cells, to redirect host immune responses to CDl 33+ FIkI+ endothelial progenitor cells. In another embodiment, CD133 FIkI+ endothelial progenitor cells can be inactivated or killed by other methods such as by the delivery to these cells of cytotoxins or other compounds capable of altering their function. In similar embodiments, the progenitor cells can be common progenitor cells for both endothelial progenitor cells and macrophages.
In one embodiment, an antigen is delivered to CD133+ FIkI+ endothelial progenitor cells to inactivate or kill these cells. In this embodiment, ligands that bind to CD133+ FIkI+ endothelial progenitor cells can be conjugated with an antigen to redirect host immune responses to the these cells, or the ligand can be conjugated to a cytotoxin for killing of these cells. Ligands that can be used in the conjugates of the present invention include those that bind to receptors expressed on CDl 33+ FIkI+ endothelial progenitor cells, such as the folate receptor, or ligands such as monoclonal antibodies directed to cell surface markers expressed on CD133 FIkI+ endothelial progenitor cells or other ligands that bind to these cells. In another embodiment, ligands that bind to CDl 33+ FIkI+ endothelial progenitor cells are conjugated to a quantifiable marker and the conjugate is used to diagnose diseases worsened by CDl 33 Flkl endothelial progenitor cells. In similar embodiments, the progenitor cells can be common progenitor cells for both endothelial progenitor cells and macrophages. In this embodiment, ligands that bind to the common precursor cells can be used.
In another embodiment, a method is provided for diagnosing a disease state worsened by CDl 33+ FIkI+ endothelial progenitor cells. The method comprises the steps of isolating CDl 33+ FIkI+ endothelial progenitor cells from a patient suffering from a disease state worsened by CDl 33+ FIkI+ endothelial progenitor cells, contacting the endothelial progenitor cells with a composition comprising a conjugate or complex of the general formula
A0-X where the group Ab comprises a vitamin, or an analog thereof, that binds to the progenitor cells and the group X comprises a quantifiable marker, and quantifying the percentage of CD133+ FIkI+ endothelial progenitor cells that expresses a receptor for the vitamin. In another embodiment, Ab comprises folate, or an analog thereof. In yet another embodiment, Ab comprises a CDl 33+ FIkI+ endothelial progenitor cell-binding antibody or antibody fragment or other ligands that bind to CD133+ FIkI+ endothelial progenitor cells. In another embodiment, the quantifiable marker comprises a metal chelating moiety that binds an element that is a radionuclide. In still another embodiment, the quantifiable marker comprises a chromophore selected from the group consisting of fluorescein, Oregon Green, rhodamine, phycoerythrin, Texas Red, and AlexaFluor 488, or another appropriate fluorescent chromophore. In similar embodiments, the progenitor cells can be common progenitor cells for both endothelial progenitor cells and macrophages. In this embodiment, ligands that bind to the common precursor cells can be used.
In another embodiment, a method is provided for diagnosing a disease state worsened by CD133+ FIkI+ endothelial progenitor cells. The method comprises the steps of administering parenterally to a patient a composition comprising a conjugate or complex of the general formula AB-X where the group Ab comprises a vitamin, or an analog thereof, that binds to CDl 33+ FIkI+ endothelial progenitor cells and the group X comprises a quantifiable marker, and quantifying the percentage of CD133+ FIkI+ endothelial progenitor cells that expresses a receptor for the vitamin. In similar embodiments, the progenitor cells can be common progenitor cells for both endothelial progenitor cells and macrophages. The quantifiable marker can be, for example, a radioactive probe, a fluorescent probe, an enzyme capable of amplifying a signal, an antibody capable of assisting in amplifying a signal, or other agents for use in amplifying a signal, such as oligonucleotides. In another embodiment, a method is provided for treating a disease state worsened by CD133+ FIkI+ endothelial progenitor cells. The method comprises the steps of administering to a patient suffering from a disease state worsened by CDl 33+ FIkI+ endothelial progenitor cells an effective amount of a composition comprising a conjugate or complex of the general formula AB-X where the group Ab comprises a vitamin, or an analog thereof, that binds to CDl 33+ FIkI+ endothelial progenitor cells and the group X comprises an antigen, a cytotoxin, or a compound capable of altering the function of the progenitor cells, and eliminating the disease state. In similar embodiments, the progenitor cells can be common progenitor cells for both endothelial progenitor cells and macrophages.
In yet another embodiment, a compound for diagnosing or treating a disease state worsened by progenitor cells, such as CDl 33+ FIkI+ endothelial progenitor cells or common progenitor cells for both endothelial progenitor cells and macrophages is provided. The compound is selected from the following group of compounds:
Figure imgf000007_0001
Alexa Fluor 488 - Cys-γ-Glu-Pteroic Acid MW 1242.21
Figure imgf000007_0002
MW 1507.5
Figure imgf000007_0003
Indomethacin-Cys-Asp-Asp-Arg-AspHy-Glu-Reroic Acid MW 1462
Figure imgf000008_0001
Prednisolone-Cys— g-Glu-Pteroic Acid MW 1007.10
Figure imgf000008_0002
Figure imgf000008_0003
Dexamethasone-Cys~g-Glu-Pteroic Acid MW 1039.11
Figure imgf000009_0001
Figure imgf000009_0002
Figure imgf000010_0001
Folate-Oregon Green 488
Figure imgf000010_0002
Figure imgf000011_0001
Folate-CW800
Figure imgf000011_0002
Figure imgf000011_0003
Folate-EDA-Rhodamine
Figure imgf000011_0004
Folate-EDA-Tetramethylrhodamine
Figure imgf000012_0001
Folate-Lys-Rhodamine
Figure imgf000012_0002
Folate-AlexaFluor 488
In another embodiment, a method of quantifying endothelial progenitor cells is provided. The method comprises the steps of isolating the progenitor cells from a patient suffering from a disease state mediated by the progenitor cells, contacting the progenitor cells with a composition comprising a conjugate or complex of the general formula Ab-X where the group Ab comprises a vitamin, or an analog thereof, that binds to endothelial progenitor cells and the group X comprises a quantifiable marker, and quantifying the percentage of progenitor cells that expresses a receptor for the vitamin. In another embodiment, a use is provided of a composition comprising a conjugate or complex of the general formula Ab-X where the group Ab comprises a vitamin, or an analog thereof, that binds to the progenitor cells and the group X comprises an antigen, a cytotoxin, or a compound capable of altering progenitor cell function in the manufacture of a medicament for use in treating a disease state worsened by progenitor cells.
In yet another embodiment, a method of quantifying endothelial progenitor cells is provided. The method comprises the steps of contacting the progenitor cells in a patient suffering from a disease state mediated by the progenitor cells with a composition comprising a conjugate or complex of the general formula Ab-X where the group Ab comprises a vitamin, or an analog thereof, that binds to endothelial progenitor cells and the group X comprises a quantifiable marker, and quantifying the percentage of progenitor cells that expresses a receptor for the vitamin.
BRIEF DESCRIPTION OF THE DRAWINGS
Fig. 1 shows flow cytometry analysis, using Flkl (A), CDl 15 (B), CD69 (C)5 CDl Ib (D), CD8a (E), and CD25 (F) antibodies and folate-FITC, of markers that are co-expressed with the folate receptor on CD133+ FIkI+ endothelial progenitor cells or on common precursor cells to both endothelial progenitor cells and macrophages.
Fig. 2 shows flow cytometry analysis, using CD62L (A), CD80 (B), CD86 (C), CD44 (D), CD23 (E), and CD14 (F) antibodies and folate-FITC, of markers that are co-expressed with the folate receptor on CDl 33+ FIkI+ endothelial progenitor cells.
Fig. 3 shows flow cytometry analysis, using Ly-6 (A), F4/80 (B), CD49d (C), CDl 6.2/32.2 (D), and MHC Class II (E) antibodies and folate-FITC, of markers that are co-expressed with the folate receptor on CDl 33+ FIkI+ endothelial progenitor cells or on common precursor cells to both endothelial progenitor cells and macrophages.
Fig. 4 shows folate-fluorescein (folate-FITC) binding, quantified by flow cytometry, to CD133+ endothelial progenitor cells (panel A), to FIk-I+ endothelial progenitor cells (Panel B), and to CD44+ endothelial progenitor cells (panel C) without excess unlabeled folic acid (top panels) or preincubated with an excess of unlabeled folic acid (bottom panels (Competed Samples)) to compete with folate-FITC for binding.
Fig. 5 shows folate-fluorescein (folate-FITC) binding, quantified by flow cytometry, to Ly-6+ endothelial progenitor cells (panel A), to CD25+ endothelial progenitor cells (Panel B), and to CD62-L+ endothelial progenitor cells (panel C) without excess unlabeled folic acid (top panels) or preincubated with an excess of unlabeled folic acid (bottom panels (Competed Samples)) to compete with folate- FITC for binding.
DETAILED DESCRIPTION
Methods are provided for treating and diagnosing disease states worsened (e.g., caused or augmented) by progenitor cells, such as CD1334" FIk-I+ endothelial progenitor cells or common precursor cells to both endothelial progenitor cells and macrophages (i.e., referred to in this application as "common precursor cells"). Exemplary disease states include fibromyalgia, rheumatoid arthritis, osteoarthritis, ulcerative colitis, Crohn's disease, psoriasis, osteomyelitis, multiple sclerosis, atherosclerosis, pulmonary fibrosis, sarcoidosis, systemic sclerosis, organ transplant rejection (GVHD), lupus erythematosus, Sjogren's syndrome, glomerulonephritis, inflammations of the skin (e.g., psoriasis), cancer, proliferative retinopathy, restenosis, and chronic inflammations. Such disease states can be diagnosed by isolating the progenitor cells from a patient suffering from such disease state, contacting the cells with a composition comprising a conjugate of the general formula Ab-X wherein the group Ab comprises a ligand that binds to the progenitor cells, and the group X comprises a quantifiable marker, and quantifying the percentage of the progenitor cells expressing a receptor for the ligand.
As used herein, the phrase "progenitor cells" includes CD 133 and/or FIkI+ endothelial progenitor cells and common precursor cells for both endothelial progenitor cells and macrophages. As used herein, the phrase "common precursor cells" refers to common precursor cells for both endothelial progenitor cells and macrophages. These cells have CDl 33 and/or Flkl markers and also have a marker selected from the group consisting of CDl Ib, F4/80, and CDl 15. As used herein, the terms "eliminated" and "eliminating" in reference to the disease state, mean reducing the symptoms or eliminating the symptoms of the disease state or preventing the progression or the reoccurrence of disease.
As used herein, the terms "elimination" and "deactivation" of the progenitor cell population that expresses the ligand receptor mean that this progenitor cell population is killed or is completely or partially inactivated which reduces the pathogenesis characteristic of the disease state being treated.
As used herein, "worsened by" in reference to diseases worsened by progenitor cells means caused by or augmented by. For example, endothelial progenitor cells can directly cause disease or can augment disease states such as by stimulating other immune cells to secrete factors that worsen disease states, such as by stimulating T-cells to secrete TNF-o; or by increasing the blood supply (e.g., by vasculogenesis) to pathologic tissues, such as cancer tissues. Illustratively, endothelial progenitor cells themselves may also harbor infections and cause disease and infected progenitor cells may cause other immune cells to secrete factors that cause disease such as TNF-α secretion by T-cells.
Such disease states can also be diagnosed by administering parenterally to a patient a composition comprising a conjugate or complex of the general formula Ab-X where the group Ab comprises a ligand that binds to progenitor cells and the group X comprises a quantifiable marker, and quantifying the percentage of the cells that expresses a receptor for the ligand.
CD133+ FIk-I+ endothelial progenitor cell-worsened disease states can be treated in accordance with the methods disclosed herein by administering an effective amount of a composition Ab-X wherein Ab comprises a ligand that binds to CD133+ FIk-I+ endothelial progenitor cells and wherein the group X comprises an antigen, a cytotoxin, or a compound capable of altering the function of the endothelial progenitor cells. Such targeting conjugates, when administered to a patient suffering from a disease state augmented by the endothelial progenitor cells, work to concentrate and associate the conjugated cytotoxin, antigen, or compound capable of altering endothelial progenitor cell function with the population of endothelial progenitor cells to kill the cells or alter cell function. The conjugate is typically administered parenterally, but can be delivered by any suitable method of administration (e.g., orally), as a composition comprising the conjugate and a pharmaceutically acceptable carrier therefor. Conjugate administration is typically continued until symptoms of the disease state are reduced or eliminated, or administration is continued after this time to prevent progression or reappearance of the disease. The cells may be common precursor cells in similar embodiments and in these embodiments ligands that bind to common precursor cells can be used.
In one embodiment, disease states worsened by progenitor cells are diagnosed in a patient by isolating the cells from the patient, contacting the progenitor cells with a conjugate Ab-X wherein Ab comprises a ligand that binds to the progenitor cells and X comprises a quantifiable marker, and quantifying the percentage of progenitor cells expressing the receptor for the ligand. In another embodiment, the diagnostic conjugates can be administered to the patient as a diagnostic composition comprising a conjugate and a pharmaceutically acceptable carrier and thereafter the progenitor cells can be collected from the patient to quantify the percentage of cells expressing the receptor for the ligand Ab. In this embodiment, the composition is typically formulated for parenteral administration and is administered to the patient in an amount effective to enable quantification of the progenitor cells. In another embodiment, disease states can also be diagnosed by administering parenterally to a patient a composition comprising a conjugate or complex of the general formula Ab-X where the group Ab comprises a ligand that binds to progenitor cells and the group X comprises a quantifiable marker, and quantifying the percentage of the cells that expresses a receptor for the ligand.
In one embodiment, for example, the quantifiable marker (e.g., a reporter molecule) can comprise a radiolabeled compound such as a chelating moiety and an element that is a radionuclide, for example a metal cation that is a radionuclide. In another embodiment, the radionuclide is selected from the group consisting of technetium, gallium, indium, and a positron emitting radionuclide (PET imaging agent). In another embodiment, the quantifiable marker can comprise a fluorescent chromophore such as, for example, fluorescein, rhodamine, Texas Red, phycoerythrin, Oregon Green, AlexaFluor 488 (Molecular Probes, Eugene, Oregon), Cy3, Cy5, Cy7, and the like.
Diagnosis typically occurs before treatment. However, in the diagnostic methods described herein, the term "diagnosis" can also mean monitoring of the disease state before, during, or after treatment to determine the progression of the disease state. The monitoring can occur before, during, or after treatment, or combinations thereof, to determine the efficacy of therapy, or to predict future episodes of disease. The quantification can be performed by any suitable method known in the art, including imaging methods, such as intravital imaging. The method disclosed herein can be used for both human clinical medicine and veterinary applications. Thus, the host animal afflicted with the disease state worsened by progenitor cells and in need of diagnosis or therapy can be a human, or in the case of veterinary applications, can be a laboratory, agricultural, domestic or wild animal. In embodiments where the conjugates are administered to the patient or animal, the conjugates can be administered parenterally to the animal or patient suffering from the disease state, for example, intradermally, subcutaneously, intramuscularly, intraperitoneally, or intravenously. Alternatively, the conjugates can be administered to the animal or patient by other medically useful procedures and effective doses can be administered in standard or prolonged release dosage forms, such as a slow pump. The therapeutic method described herein can be used alone or in combination with other therapeutic methods recognized for the treatment of inflammatory disease states, or disease states augmented by vasculogenesis.
In the ligand conjugates of the general formula Ab-X, the group Ab is a ligand that binds to CD133+ FIk-I+ endothelial progenitor cells or common precursor cells when the conjugates are used to diagnose or treat disease states. Any of a wide number of binding ligands can be employed. Acceptable ligands include particularly folate receptor binding ligands, and analogs thereof, and antibodies or antibody fragments capable of recognizing and binding to surface moieties expressed or presented on CDl 33+ FIk-I+ endothelial progenitor cells or on common precursor cells. Antagonists and agonists for CD133, Flkl, CDl Ib, F4/80, or CDl 15 may be acceptable ligands. In one embodiment, the binding ligand is folic acid, a folic acid analog, or another folate receptor binding molecule. In another embodiment the binding ligand is a specific monoclonal or polyclonal antibody or an Fab or an scFv (i.e., a single chain variable region) fragment of an antibody capable of binding to CD1334" FIk-I+ endothelial progenitor cells or to common precursor cells.
In one embodiment, the binding ligand can be folic acid, a folic acid analog, or another folate receptor-binding molecule. Analogs of folate that can be used include folinic acid, pteropolyglutamic acid, and folate receptor-binding pteridines such as tetrahydropterins, dihydrofolates, tetrahydrofolates, and their deaza and dideaza analogs. The terms "deaza" and "dideaza" analogs refers to the art recognized analogs having a carbon atom substituted for one or two nitrogen atoms in the naturally occurring folic acid structure. For example, the deaza analogs include the 1 -deaza, 3-deaza, 5-deaza, 8-deaza, and 10-deaza analogs. The dideaza analogs include, for example, 1,5 dideaza, 5,10-dideaza, 8,10-dideaza, and 5,8-dideaza analogs. The foregoing folic acid analogs are conventionally termed "folates," reflecting their capacity to bind to folate receptors. Other folate receptor-binding analogs include ammopterin, amethopterin (methotrexate), N10-methylfolate, 2- deamino-hydroxyfolate, deaza analogs such as 1 -deazamethopterin or 3- deazamethopterin, and 3',5'-dichloro-4-amino-4-deoxy-N10-methylpteroylglutamic acid (dichloromethotrexate).
In another embodiment, other vitamins can be used as the binding ligand. The vitamins that can be used in accordance with the methods described herein include niacin, pantothenic acid, folic acid, riboflavin, thiamine, biotin, vitamin B i2, vitamins A, D, E and K, other related vitamin molecules, analogs and derivatives thereof, and combinations thereof.
In other embodiments, the binding ligand can be any ligand that binds to a receptor expressed or overexpressed on endothelial progenitor cells or common precursor cells including CD133, Flkl, CDl Ib, CDl 15, CD69, CD8a, CD25, CD62L, CD80, CD86, CD44, CD23, CD14, Ly-6, F4/80, CD49d, CDl 6.2/32.2, and the like. Examples of such ligands include both antagonists and agonists for each of the above membrane-spanning proteins.
The targeted conjugates used for diagnosing or treating disease states mediated by progenitor cells have the formula Ab-X, wherein Ab is a ligand capable of binding to the progenitor cells, and the group X comprises a quantifiable marker or an antigen (such as an immunogen), cytotoxin, or a compound capable of altering progenitor cell function. In such conjugates wherein the group Ab is folic acid, a folic acid analog, or another folic acid receptor binding ligand, these conjugates are described in detail in U.S. Patent No. 5,688,488, the specification of which is incorporated herein by reference. That patent, as well as related U.S. Patents Nos.
5,416,016 and 5,108,921, and related U.S. Patent Publication Serial No.
US 2005/0002942 Al, each incorporated herein by reference, describe methods and examples for preparing conjugates useful in accordance with the methods described herein. The present targeted diagnostic and therapeutic agents can be prepared and used following general protocols described in those earlier patents and patent applications, and by the protocols described herein. In accordance with another embodiment, there is provided a method of treating disease states worsened by progenitor cells by administering to a patient suffering from such disease state an effective amount of a composition comprising a conjugate of the general formula Ab-X wherein Ab is as defined above and the group X comprises a cytotoxin, an antigen (i.e., a compound that elicits an immune response in vivo), or a compound capable of altering progenitor cell function. In these embodiments, the progenitor cells can be activated cells and the group Ab can be any of the ligands described above. Exemplary of cytotoxic moieties useful for forming conjugates for use in accordance with the methods described herein are clodronate, anthrax, Pseudomonas exotoxin, typically modified so that these cytotoxic moieties do not bind to normal cells, and other toxins or cytotoxic agents including art- recognized chemotherapeutic agents such as adrenocorticoids, alkylating agents, antiandrogens, antiestrogens, androgens, estrogens, antimetabolites such as cytosine arabinoside, purine analogs, pyrimidine analogs, and methotrexate, busulfan, carboplatin, chlorambucil, cisplatin and other platinum compounds, tamoxiphen, taxol, cyclophosphamide, plant alkaloids, prednisone, hydroxyurea, teniposide, and bleomycin, nitrogen mustards, nitrosureas, vincristine, vinblastine, MEK kinase inhibitors, MAP kinase pathway inhibitors, PI-3 -kinase inhibitors, mitochondrial perturbants, NFKB pathway inhibitors, proteosome inhibitors, pro-apoptotic agents, glucocorticoids, such as prednisolone, flumethasone, dexamethasone, and betamethasone, indomethacin, diclofenac, non-steroidal anti-inflammatory agents, cyclooxygenase inhibitors, lipooxygenase inhibitors, apoptosis-inducing agents, proteins such as pokeweed, saporin, momordin, and gelonin, non-steroidal antiinflammatory drugs (NSAIDs), protein synthesis inhibitors, didemnin B, verrucarin A, geldanamycin, and the like. Such toxins or cytotoxic compounds can be directly conjugated to the targeting ligand, for example, folate or another folate receptor- binding ligand, or they can be formulated in liposomes or other small particles which themselves are targeted as conjugates of the progenitor cell-binding ligand typically by covalent linkages to component phospholipids. Similarly, when the group X comprises a compound capable of altering progenitor cell function, for example, a cytokine such as IL-IO or IL-11, the compound can be covalently linked to the targeting ligand Ab, for example, a folate receptor-binding ligand or a progenitor cell-binding antibody or antibody fragment directly, or the function altering compound can be encapsulated in a liposome which is itself targeted to progenitor cells by pendent targeting ligands Ab covalently linked to one or more liposome components.
In another embodiment, conjugates Ab-X where X is an antigen or a compound capable of altering progenitor cell function, can be administered in combination with a cytotoxic compound. The cytotoxic compounds listed above are among the compounds suitable for this purpose.
In another method of treatment embodiment, the group X in the targeted conjugate Ab-X, comprises an antigen (i.e., a compound that elicits an immune response in vivo), the ligand-antigen conjugates being effective to "label" the population of progenitor cells responsible for disease pathogenesis in the patient suffering from the disease for specific elimination by an endogenous immune response or by co-administered antibodies. The use of ligand-antigen conjugates in the method of treatment described herein works to enhance an immune response- mediated elimination of the progenitor cell population that expresses the ligand receptor. Such elimination can be effected through an endogenous immune response or by a passive immune response effected by co-administered antibodies.
The methods of treatment involving the use of ligand-antigen conjugates are described in U.S. Patent Application Publications Nos. US 2001/0031252 Al and US 2002/0192157 Al and PCT Publication No. PCT/US2004/014097, each incorporated herein by reference.
The endogenous immune response can include a humoral response, a cell-mediated immune response, and any other immune response endogenous to the host animal, including complement-mediated cell lysis, antibody-dependent cell- mediated cytotoxicity (ADCC), antibody opsonization leading to phagocytosis, clustering of receptors upon antibody binding resulting in signaling of apoptosis, antiproliferation, or differentiation, and direct immune cell recognition of the delivered antigen (e.g., a hapten). It is also contemplated that the endogenous immune response may employ the secretion of cytokines that regulate such processes as the multiplication, differentiation, and migration of immune cells. The endogenous immune response may include the participation of such immune cell types as B cells, T cells, including helper and cytotoxic T cells, macrophages, natural killer cells, neutrophils, LAK cells, and the like. The humoral response can be a response induced by such processes as normally scheduled vaccination, or active immunization with a natural antigen or an unnatural antigen or hapten, e.g., fluorescein isothiocyanate (FITC), with the unnatural antigen inducing a novel immunity. Active immunization involves multiple injections of the unnatural antigen or hapten scheduled outside of a normal vaccination regimen to induce the novel immunity. The humoral response may also result from an innate immunity where the host animal has a natural preexisting immunity, such as an immunity to α-galactosyl groups.
Alternatively, a passive immunity may be established by administering antibodies to the host animal such as natural antibodies collected from serum or monoclonal antibodies that may or may not be genetically engineered antibodies, including humanized antibodies. The utilization of a particular amount of an antibody reagent to develop a passive immunity, and the use of a ligand-antigen conjugate wherein the passively administered antibodies are directed to the antigen, would provide the advantage of a standard set of reagents to be used in cases where a patient's preexisting antibody titer to potential antigens is not therapeutically useful. The passively administered antibodies may be "co-administered" with the ligand- antigen conjugate, and co-administration is defined as administration of antibodies at a time prior to, at the same time as, or at a time following administration of the ligand-antigen conjugate. The preexisting antibodies, induced antibodies, or passively administered antibodies will be redirected to the progenitor cells by preferential binding of the ligand-antigen conjugates to the progenitor cell populations, and such pathogenic cells are killed by complement-mediated lysis, ADCC, antibody- dependent phagocytosis, or antibody clustering of receptors. The cytotoxic process may also involve other types of immune responses, such as cell-mediated immunity.
Acceptable antigens for use in preparing the conjugates used in the method of treatment described herein are antigens that are capable of eliciting antibody production in a host animal or that have previously elicited antibody production in a host animal, resulting in a preexisting immunity, or that constitute part of the innate immune system. Alternatively, antibodies directed against the antigen may be administered to the host animal to establish a passive immunity. Suitable antigens for use in the invention include antigens or antigenic peptides against which a preexisting immunity has developed via normally scheduled vaccinations or prior natural exposure to such agents such as polio virus, tetanus, typhus, rubella, measles, mumps, pertussis, tuberculosis and influenza antigens, and α-galactosyl groups. In such cases, the ligand-antigen conjugates will be used to redirect a previously acquired humoral or cellular immunity to a population of progenitor cells in the host animal for elimination of the progenitor cells.
Other suitable immunogens include antigens or antigenic peptides to which the host animal has developed a novel immunity through immunization against an unnatural antigen or hapten, for example, fluorescein isothiocyanate (FITC) or dinitrophenyl, and antigens against which an innate immunity exists, for example, super antigens and muramyl dipeptide.
The progenitor cell-binding ligands and antigens, cytotoxic agents, compounds capable of altering progenitor cell function, or imaging agents, as the case may be in forming conjugates for use in accordance with the methods described herein can be conjugated by using any art-recognized method for forming a complex. This can include covalent, ionic, or hydrogen bonding of the ligand to the antigen, either directly or indirectly via a linking group such as a divalent linker. The conjugate is typically formed by covalent bonding of the ligand to the targeted entity through the formation of amide, ester or imino bonds between acid, aldehyde, hydroxy, amino, or hydrazo groups on the respective components of the complex or, for example, by the formation of disulfide bonds. Methods of linking binding ligands to antigens, cytotoxic agents, compounds capable of altering progenitor cell function, or quantifiable markers are described in U.S. Patent Application Publication No. US 2005/0002942-A1 and PCT Publication No. WO 2006/012527, each incorporated herein by reference. Alternatively, as mentioned above, the ligand complex can be one comprising a liposome wherein the targeted entity (that is, the quantifiable marker, or the antigen, cytotoxic agent or progenitor cell function-altering agent) is contained within a liposome which is itself covalently linked to the binding ligand. Other nanoparticles, dendrimers, derivatizable polymers or copolymers that can be linked to therapeutic or quantifiable markers useful in the treatment and diagnosis of progenitor cell-worsened diseases can also be used in targeted conjugates.
In one embodiment of the invention the ligand is folic acid, an analog of folic acid, or any other folate receptor binding molecule, and the folate ligand is conjugated to the targeted entity by a procedure that utilizes trifiuoroacetic anhydride to prepare γ-esters of folic acid via a pteroyl azide intermediate. This procedure results in the synthesis of a folate ligand, conjugated to the targeted entity only through the γ-carboxy group of the glutamic acid groups of folate. Alternatively, folic acid analogs can be coupled through the α-carboxy moiety of the glutamic acid group or both the o: and γ carboxylic acid entities.
The therapeutic methods described herein can be used to slow the progress of disease completely or partially. Alternatively, the therapeutic methods described herein can eliminate or prevent reoccurrence of the disease state. The conjugates used in accordance with the methods described herein of the formula Ab-X are used in one aspect to formulate therapeutic or diagnostic compositions, for administration to a patient, wherein the compositions comprise effective amounts of the conjugate and an acceptable carrier therefor. Typically such compositions are formulated for parenteral use. The amount of the conjugate effective for use in accordance with the methods described herein depends on many parameters, including the nature of the disease being treated or diagnosed, the molecular weight of the conjugate, its route of administration and its tissue distribution, and the possibility of co-usage of other therapeutic or diagnostic agents. The effective amount to be administered to a patient is typically based on body surface area, patient weight and physician assessment of patient condition. An effective amount can range from about to 1 ng/kg to about 1 mg/kg, more typically from about 1 μg/kg to about 500 μg/kg, and most typically from about 1 μg/kg to about 100 μg/kg.
Any effective regimen for administering the ligand conjugates can be used. For example, the ligand conjugates can be administered as single doses, or they can be divided and administered as a multiple-dose daily regimen. Further, a staggered regimen, for example, one to three days per week can be used as an alternative to daily treatment, and such an intermittent or staggered daily regimen is considered to be equivalent to every day treatment and within the scope of this disclosure. In one embodiment, the patient is treated with multiple injections of the ligand conjugate wherein the targeted entity is an antigen or a cytotoxic agent or a compound capable of altering progenitor cell function to eliminate the population of pathogenic progenitor cells. Tn one embodiment, the patient is treated, for example, injected multiple times with the ligand conjugate at, for example, 12-72 hour intervals or at 48-72 hour intervals. Additional injections of the ligand conjugate can be administered to the patient at intervals of days or months after the initial injections, and the additional injections prevent recurrence of disease. Alternatively, the ligand conjugates may be administered prophylactically to prevent the occurrence of disease in patients known to be disposed to development of disease states worsened by progenitor cells. In one embodiment, more than one type of ligand conjugate can be used, for example, the host animal may be pre-immunized with fluorescein isothiocyanate and dinitrophenyl and subsequently treated with fluorescein isothiocyanate and dinitrophenyl linked to the same or different targeting ligands in a co-dosing protocol.
The ligand conjugates are administered in one aspect parenterally and most typically by intraperitoneal injections, subcutaneous injections, intramuscular injections, intravenous injections, intradermal injections, or intrathecal injections. The ligand conjugates can also be delivered to a patient using an osmotic pump.
Examples of parenteral dosage forms include aqueous solutions of the conjugate, for example, a solution in isotonic saline, 5% glucose or other well-known pharmaceutically acceptable liquid carriers such as alcohols, glycols, esters and amides. The parenteral compositions for use in accordance with this invention can be in the form of a reconstitutable lyophilizate comprising the one or more doses of the ligand conjugate. In another aspect, the ligand conjugates can be formulated as one of any of a number of prolonged release dosage forms known in the art such as, for example, the biodegradable carbohydrate matrices described in U.S. Patent Nos. 4,713,249; 5,266,333; and 5,417,982, the disclosures of which are incorporated herein by reference. The ligand conjugates can also be administered topically such as in an ointment or a lotion, for example, for treatment of inflammations of the skin.
In any of the embodiments discussed above, the progenitor cells can be activated cells or other cell populations that augment or cause disease states. The following examples are illustrative embodiments only and are not intended to be limiting.
EXAMPLE 1 MATERIALS Fmoc-protected amino acid derivatives, trityl-protected cysteine 2- chlorotrityl resin (H-Cys(Trt)-2-ClTrt resin #04-12-2811), Fmoc-lysine(4- methyltrityl) wang resin, 2-(lH-benzotriaxol-l-yl)-l,l,3,3-tetramethyluronium hexafluorophosphage (HBTU) and N-hydroxybenzotriazole were purchased from Novabiochem (La Jolla, CA). N10-trifluoroacetylpteroic acid was purchased from Sigma, St. Louis, MO. Piperidine, DIPEA (diisopropylethylamine), Rhodamine B isothiocyanate (Rd-ITC) and triisopropyl saline (TIPS) were from Aldrich (Milwaukee). Anti-mouse antibodies were purchased from Caltag Laboratories, Burlingame, CA. The following anti-mouse antibodies were purchased from Caltag Laboratories: CDlIb, CD16.2/32.2, CD23, CD44, CD49d, CD62L, CD69, CD80, CD86, F4/80, Ly-6C/G, I-Ab MHC Class II and Streptavidin secondary fluorescence tag. The anti-mouse CD8a, CD 133, and FIk-I antibodies were purchased from eBioscience (San Diego, CA). The anti-mouse CDl 15 antibody was purchased from Serotec (Raleigh, NC). The anti-mouse CD 14 and CD25 antibodies were purchased from Becton Dickinson (Franklin Lakes, NJ). Folate-R-Phycoerytherin, Folate- AlexaFluor 488, Folate-Texas Red, and Folate-Fluorescein and Folate-cysteine were synthesized as described. Folate-FITC was provided by Endocyte, Inc.
EXAMPLE 2
SYNTHESIS OF FOLATE-CYSTEINE Standard Fmoc peptide chemistry was used to synthesize folate- cysteine with the cysteine attached to the 7-COOH of folic acid. The sequence Cys- Glu-Pteroic acid (Folate-Cys) was constructed by Fmoc chemistry with HBTU and N- hydroxybenzotriazole as the activating agents along with diisopropyethylamine as the base and 20% piperidine in dimethylformamide (DMF) for deprotection of the Fmoc groups. An oj-t-Boc-protected N-α-Fmoc-L-glutamic acid was linked to a trityl- protected Cys linked to a 2-Chlorotrityl resin. NI0-trifluoroacetylpteroic acid was then attached to the 7-COOH of GIu. The Folate-Cys was cleaved from the resin using a 92.5% trifluoroacetic acid-2.5% water-2.5% triisopropylsilane- 2.5% ethanedithio solution. Diethyl ether was used to precipitate the^product, and the precipitant was collected by centrifugation. The product was washed twice with diethyl ether and dried under vacuum overnight. To remove the N10-trifluoracetyl protecting group, the product was dissolved in a 10% ammonium hydroxide solution and stirred for 30 min at room temperature. The solution was kept under a stream of nitrogen the entire time in order to prevent the cysteine from forming disulfides. After 30 minutes, hydrochloric acid was added to the solution until the compound precipitated. The product was collected by centrifugation and lyophilized. The product was analyzed and confirmed by mass spectroscopic analysis (MW 544, M+ 545).
Figure imgf000026_0001
Cys— g-Glu-Pteroic Acid MW 544.54
EXAMPLE 3 SYNTHESIS OF FOLATE-CYS-ALEXAFLUOR 488 AlexaFluor 488 Cs-maleimide (Molecular Probes, Eugene, OR) was dissolved in dimethyl sulfoxide (DMSO) (0.5mg in 50μl DMSO). A 1.5 molar equivalent (0.57mg) of Folate-Cys was added to the solution and mixed for 4 hours at room temperature. Folate-Cys-AlexaFluor 488 (Folate- AlexaFluor) was purified by reverse-phase HPLC on a Cl 8 column at a flow rate of lml/min. The mobile phase, consisting of 1OmM NH4HCO3 buffer, pH 7.0 (eluent A) and acetonitrile (eluent B), was maintained at a 99:1 A:B ratio for the first minute and then changed to 1 :99 A:B in a linear gradient over the next 29 minutes. Folate-Cys- AlexaFluor 488 eluted at 20 minutes. The product was confirmed by mass spectroscopy and the biologic activity was confirmed by fluorescence measurement of its binding to cell surface folate receptors on folate receptor positive M 109 cells in culture.
Figure imgf000027_0001
Alexa Fluor 488 - Cys-^y-Glu-Pteroic Acid MW 1242.21
EXAMPLE 4
SYNTHESIS OF FOLATE-CYS-TEXAS RED Texas Red C2-maleimide (Molecular Probes, Eugene, OR) was dissolved in dimethyl sulfoxide (DMSO) (lmg in 200μl DMSO). A 1.4 molar equivalent (lmg) of Folate-Cys was added to the solution and mixed for 4 hours at room temperature. Folate-Cys-Texas Red (Folate-Texas Red) was purified by reverse-phase HPLC on a Cl 8 column at a flow rate of lml/min. The mobile phase, consisting of 1OmM NH4HCO3 buffer, pH 7.0 (eluent A) and acetonitrile (ehient B), was maintained at a 99: 1 A:B ratio for the first five minutes and then changed to 70:30 A:B in a linear gradient over the next 30 minutes followed by a 1 :99 A:B linear gradient over the last 15 minutes. Folate-Cys-Texas Red eluted as two isomer peaks at 44.5 and 45.8 minutes. The product was confirmed by mass spectroscopy and the biologic activity was confirmed by fluorescence measurement of its binding to cell surface folate receptors on folate receptor positive Ml 09 cells in culture.
Figure imgf000027_0002
EXAMPLE 5
SYNTHESIS OF FOLATE-LYS-OREGON GREEN 514 Standard Fmoc peptide chemistry was used to synthesize a folate peptide linked to Oregon Green (Molecular Probes, Eugene, OR) attached to the y- COOH of folic acid. The sequence Lys-Glu-Pteroic acid (Folate-Cys) was constructed by Fmoc chemistry with HBTU and N-hydroxybenzotriazole as the activating agents along with diisopropyethylamine as the base and 20% piperidine in dimethylformamide (DMF) for deprotection of the Fmoc groups. An ot-Boc- protected N-α-Fmoc-L-glutamic acid followed by a N10-trifluoroacetylpteroic acid was linked to a Fmoc-protected lysine wang resin containing a 4-methyltrityl protecting group on the e-amine. The methoxytrityl protecting group on the 6-amine of lysine was removed with 1% trifluoroacetic acid in dichloromethane to allow attachment of Oregon Green (Folate-Oregon Green). A 1.5 molar equivalent of Oregon Green carboxylic acid, succinimidyl ester was reacted overnight with the peptide and then washed thoroughly from the peptide resin beads. The Folate-Oregon Green was then cleaved from the resin with a 95% trifluoroacetic acid-2.5% water- 2.5% triisopropylsilane solution. Diethyl ether was used to precipitate the product, and the precipitant was collected by centrifugation. The product was washed twice with diethyl ether and dried under vacuum overnight. To remove the N10- trifiuoracetyl protecting group, the product was dissolved in a 10% ammonium hydroxide solution and stirred for 30 min at room temperature. The product was precipitated with combined isopropanol and ether, and the precipitant was collected by centrifugation.
Figure imgf000029_0001
EXAMPLE 6
SYNTHESIS OF FOLATE-R-PHYCOERYTHRIN Folate-phycoerythrin was synthesized by following a procedure published by Kennedy M.D. et al. in Pharmaceutical Research, Vol. 20(5); 2003. Briefly, a 10-fold excess of folate-cysteine was added to a solution of R- phycoerythrin pyridyldisulfide (Sigma, St. Louis, MO) in phosphate buffered saline (PBS), pH 7.4. The solution was allowed to react overnight at 4°C and the labeled protein (Mr -260 kDa) was purified by gel filtration chromatography using a G-15 desalting column. The folate labeling was confirmed by fluorescence microscopy of M 109 cells incubated with folate-phycoerythrin in the presence and absence of 100- fold excess of folic acid. After a 1-h incubation and 3 cells washes with PBS, the treated cells were intensely fluorescent, while the sample in the presence of excess folic acid showed little cellular fluorescence.
EXAMPLE 7
SYNTHESIS OF FOLATE-FLUORESCEIN
Folate-FITC was synthesized as described by Kennedy, M.D. et al. in Pharmaceutical Research, Vol. 20(5); 2003.
Figure imgf000030_0001
Folate-EDA-FITC
MW 888.90
EXAMPLE 8
SYNTHESIS OF FOLATE-D-R-D-D-C--PREDNISOLONE Standard Fmoc peptide chemistry was used to synthesize folate- aspartate-arginine-aspartate-aspartate-cysteine (Folate-Asp-Arg-Asp-Asp-Cys, Folate- D-R-D-D-C) with the amino acid spacer attached to the 7-COOH of folic acid. The sequence Cys-Asp-Asp-Arg-Asp-Glu-Pteroic acid (Folate- Asp-Arg-Asp-Asp-Cys) was constructed by Fmoc chemistry with HBTU and N-hydroxybenzotriazole as the activating agents along with diisopropyethylamine as the base and 20% piperidine in dimethylforrnamide (DMF) for deprotection of the Fmoc groups. Fmoc-D- Asp(OtBu)-OH was linked to a trityl-protected Cys linked to a 2-Chlorotrityl resin. A second Fmoc-D-Asp(OtBu)-OH followed by Fmoc-Arg(Pbf)-OH, Fmoc-D- Asp(OtBu)-OH and Fmoc-Glu-OtBu were added successively to the resin. N10- trifluoroacetylpteroic acid was then attached to the 7-COOH of GIu. The Folate-Asp- Arg-Asp-Asp-Cys was cleaved from the resin using a 92.5% trifluoroacetic acid-2.5% water-2.5% triisopropylsilane- 2.5% ethanedithio solution. Diethyl ether was used to precipitate the product, and the precipitant was collected by centrifugation. The product was washed twice with diethyl ether and dried under vacuum overnight. To remove the Nl0-trifluoracetyl protecting group, the product was dissolved in a 10% ammonium hydroxide solution and stirred for 30 min at room temperature. The solution was kept under a stream of nitrogen the entire time in order to prevent the cysteine from forming disulfides. After 30 minutes, hydrochloric acid was added to the solution until the compound precipitated. The product was collected by centrifugation and lyophilized. The product was analyzed and confirmed by mass spectroscopic analysis (MW 1046).
Figure imgf000031_0001
EXAMPLE 9 SYNTHESIS OF FOLATE-INDOMETHACIN
Figure imgf000031_0002
I ndomethaciπ-Cys-Asp-Asp-Arg-Asp-γ-GI u-Pteroic Acid MW 1462 2-(2-Pyridyldithio)ethanol was synthesized by dissolving 1.5 equivalents of Aldri thiol (Sigma, St. Louis, MO) with 6 equivalents of 4- dimethylaminopyridine (DMAP) in dichloromethane (DCM). The solution was purged with nitrogen and 1 equivalent of mercaptoethanol was added dropwise to the Aldrithiol solution over the course of 15 minutes. The reaction proceeded at room temperature for 30 minutes at which time no odor of mercaptoethanol remained. The reaction was diluted 100-fold with DCM and 5g of activated carbon was added per gram of Aldrithiol. The reaction mixture was filtered and the solvent removed. The mixture was resuspended in 70:30 (Petroleum etheπEthylacetate (EtOAc)) and purified by flash chromatography on a 6θA silica gel column. The product was monitored by thin layer chromatography and collected.
Folate-indomethacin was synthesized following a modified method published by Kalgutkar et al. in the Journal of Med. Chem. 2000, 43; 2860-2870 where the anti-inflammatory (indomethacin) was linked through an ester bond with the 2-(2-Pyridyldithio)ethanol. Briefly, 1 equivalent of indomethacin was dissolved in DCM along with 0.08 equivalents DMAP, 1.1 equivalents 2-(2-Pyridyldithio) ethanol and 1.1 equivalents 1,3-dicyclohexyl-carbodiimide. The reaction proceeded at room temperature for 5 hours. The reaction was purified by chromatography on silica gel (EtOAc :hexanes, 20:80). One equivalent of the purified compound was dissolved in DMSO and to it were added 1.5 equivalents of the folate- Asp- Arg-Asp- Asp-Cys peptide. The resulting solution was reacted for 3 hours at room temperature followed by purification using a HPLC reverse-phase Cl 8 column at a flow rate of lml/min. The mobile phase, consisting of 1OmM NH4HCO3 buffer, pH 7.0 (eluent A) and acetonitrile (eluent B), was maintained at a 99: 1 A:B ratio for the first five minutes and then changed to 70:30 A:B in a linear gradient over the next 30 minutes. The recovered final product was confirmed by mass spectrometry.
EXAMPLE 10 SYNTHESIS OF FOLATE-DICLOFENAC
Figure imgf000032_0001
Diclofenac-Cys-Asp-Asp-Arg-Asp-γ-Glu-Pteroic Acid
MW 1386
Folate-diclofenac was synthesized by the method described in
Example 9 except that diclofenac was used in place of indomethicin. In various embodiments, n = 1, 2, or 3, and where n is illustratively 2.
EXAMPLE 11 SYNTHESIS OF FOLATE-CYS-PREDNISOLONE
The folate glucocorticoid conjugate of prednisolone was prepared as follows. A 1.1 molar equivalent of prednisone was dissolved in tetrahydrofuran (THF). In a separate vial, a 0.7 molar equivalent of dimethylaminopyridine, 1 molar equivalent of tri(hydroxyethyl)amine and 1 molar equivalent of the linker (synthesis described in PCT Publication No. WO 2006/012527, incorporated herein by reference) were dissolved in dichloromethane. An approximately equal volume of both solutions were combined, mixed and reacted at room temperature for 4 hours. The reaction was monitored by thin layer chromatography using 40: 10: 1 (Dichloromethane: Acetonitrile: Methanol). The product had an Rf= 0.52. The product was purified on a silica column (Silica 32-63, 6θA) using the same ratio of solvents. The recovered product was dried in preparation for conjugation to a folate- peptide. The derivatized glucocorticoid was dissolved in DMSO, to which was added a 1.5 molar equivalent of either the folate-cys or folate- Asp- Arg- Asp- Asp-Cys peptide. The resulting solution was reacted for 3 hours at room temperature followed by purification using a HPLC reverse-phase Cl 8 column at a flow rate of lml/min. The mobile phase, consisting of 1OmM NH4HCO3 buffer, pH 7.0 (eluent A) and acetonitrile (eluent B), was maintained at a 99: 1 A:B ratio for the first minute and then changed to 1:99 A:B in a linear gradient over the next 39 minutes. The folate- glucocorticoid conjugate eluted at approximately 26 minutes. The recovered final product was confirmed by mass spectrometry.
Figure imgf000033_0001
Prednisolonc-Cys— g-Glu-Pteroic Acid MW 1007.10
EXAMPLE 12 SYNTHESIS OF FOLATE-CYS-DEXAMETHASONE
Figure imgf000034_0001
Dexamethasone-Cys~g-Glu-Pteroic Acid MW 1039.11
Folate-cys-dexamethasone was synthesized by a procedure similar to that described in Example 11 except that the glucocorticoid was dexamethasone.
EXAMPLE 13 SYNTHESIS OF FOLATE-CYS-FLUMETHASONE
Figure imgf000034_0002
Folate-cys-flumethasone was synthesized by a procedure similar to that described in Example 11 except that the glucocorticoid was flumethasone.
EXAMPLE 14 ISOLATION OF FOLATE-RECEPTOR-POSITIVE ENDOTHELIAL
PROGENITOR CELLS
Female 6- to 8-week-old BALB/c mice were injected in the peritoneal cavity with either Complete Freund's Adjuvant (CFA; 50-100μL), Pseudomonas aeruginosa (IxIO7 CFU (colony forming units)), or Yersinia enterocolitica (IxIO6 CFU). Cells were isolated from the peritoneal cavity by lavage with 8 mL of sterile phosphate-buffered saline (PBS) 2-4 days later. The cells were pelleted by centrifugation (400 x g, 10 minutes at room temperature) and resuspended in folate- deficient RPMI-1640 media (FD-RPMI; Gibco) containing 10% heat-inactivated fetal bovine serum (FBS), penicillin (100 IU/mL) and streptomycin (100 μg/mL). Peritoneal extracted cells were seeded at densities of 1 x 106 cells/microcentrifuge tube for antibody and folate conjugate studies.
EXAMPLE 15 LIGAND BINDING
All binding experiments were conducted on ice or in a 4°C cold room unless indicated otherwise. All antibody labeling was optimized by titration. Optimal labeling was most often achieved with a 1/1000-1/10,000 dilution of the manufacture's stock antibody solution. After cells were labeled with antibodies, the samples were washed twice with PBS to remove non-specific binding. The samples were then incubated with a 100 nM concentration of folate-FITC for 45 minutes. Competition samples were prepared by pre-incubating the appropriate samples with a 100-fold excess concentration of folic acid (lOμM) for five minutes prior to adding the folate dye conjugate. All samples were analyzed by flow cytometry using a Becton Dickinson FACS Calibur (BD, Franklin Lakes, NJ).
EXAMPLE 16
SYNTHESIS OF FOLATE RESONANCE ENERGY TRANSFER REPORTER Compound 1 was prepared by following standard Fmoc chemistry on an acid-sensitive trityl resin loaded with Fmoc-L-Cys (Trt)-OH, as described previously (adapted to the shown peptide sequence). The crude compound 1 was purified by HPLC using a VYDAC protein and peptide Cl 8 column. The HPLC- purifϊed 1 was then reacted with tetraethylrhodamine methanethiosulfonate (Molecular Probes, Eugene, OR) in DMSO to afford compound 2, in the presence of diisopropylethylamine (DIPEA). The desired product was isolated from the reaction mixture by preparative HPLC as described above. The final conjugation was performed by mixing excess DIPEA with 2 (in DMSO) followed by addition of BODIPY FL NHS ester (Molecular Probes, Eugene, OR). Compound 3 was then isolated from this reaction mixture by preparative HPLC.
Figure imgf000036_0001
EXAMPLE 17 LASER IMAGING
Fluorescence resonance energy transfer (FRET) imaging of progenitor cells to determine uptake of folate-linked markers will be carried out using a confocal microscopy. An Olympus IX-70 inverted microscopy (Olympus, USA) equipped with an Olympus FW300 scanning box and an Olympus 6OX/ 1.2 NA water objective will be used to image the cells. Separate excitation lines and emission filters will be used for each fluorochrorne (BODIPY FL, 488 nm (excitation) and 520/40 nm (emission); rhodamine, 543 nm (excitation) and 600/70 nm (emission)). Two laser sources with 543nm (He-Ne) and 488nm (Argon) wavelength can be used to excite BODIPY FL and rhodamine separately to obtain two color images when needed. Confocal images can be acquired with a size of 512 x 512 pixels at 2.7 second scan time and images can be processed using FluoView (Olympus) software.
EXAMPLE 18
LIPOSOME PREPARATION
Liposomes were prepared following methods by Leamon et al. in Bioconjugate Chemistry 2003, 14, 738-747. Briefly, lipids and cholesterol were purchased from Avanti Polar Lipids (Alabaster, AL). Folate-targeted liposomes consisted of 40 mole % cholesterol, either 4 mole % or 6 mole % polyethyleneglycol (Mr~2000)-derivatized phosphatidylethanolamine (PEG2000-PE, Nektar AL, Huntsville, AL), either 0.03 mole % or 0.1 mole % folate-cysteine-PEG3400-PE and the remaining mole% was composed of egg phosphatidylcholine. Non-targeted liposomes were prepared identically with the absence of folate-cysteme-PEG3400-PE. Lipids in chloroform were dried to a thin film by rotary evaporation and then rehydrated in PBS containing the drug. Rehydration was accomplished by vigorous vortexing followed by 10 cycles of freezing and thawing. Liposomes were then extruded 10 times through a 50nm pore size polycarbonate membrane using a high- pressure extruder (Lipex Biomembranes, Vancouver, Canada).
EXAMPLE 19 SYNTHESIS OF FOLATE-POKEWEED Pokeweed antiviral protein was purchased from Worthington
Biochemical Corporation (Lakewood, NJ). iV-succinimidyl-3-[2-pyridyldithio] propionate (SPDP; Pierce, Rockford, IL) was dissolved in dimethylformamide (9.6mM). While on ice, a 5 fold molar excess of SPDP (—170 nmoles) was added to the pokeweed solution (lmg/ml PBS, MW~29,000). The resulting solution was gently mixed and allowed to react for 30 minutes at room temperature. The non- conjugated SPDP was removed using a centrifuge molecular weight concentrator (MWCO 10,000) (Millipore, Billerica, MA). The resulting protein solution was resuspended in PBS containing 1OmM EDTA to a final volume of 1 mL. Approximately a 60 fold molar excess of folate-Asp-Arg-Asp-Asp-Cys peptide (2000 nmoles) was added to the protein solution and allowed to react for 1 hour. The non- reacted folate-Asp-Arg-Asp-Asp-Cys peptide was removed using the centrifuge concentrators as previously described. The protein was washed twice by resuspending the protein in PBS and repeating the protein concentration by centrifugation. EXAMPLE 20
SYNTHESIS OF FOLATE-SAPORIN
The protein saporin was purchased from Sigma (St. Louis, MO). Folate-saporin was prepared following folate-protein conjugation methods published by Leamon and Low in The Journal of Biological Chemistry 1992, 267(35); 24966- 24971. Briefly, folic acid was dissolved in DMSO and incubated with a 5 fold molar excess of l-ethyl-3-(3-dimethylaminopropyl)carbodiimide for 30 minutes at room temperature. The saporin was dissolved in 100 mM KH2PO4, 100 mM boric acid, pH 8.5. A 10-fold molar excess of the "activated" vitamin was added to the protein solution and the labeling reaction was allowed to proceed for 4 hours. Unreacted material was separated from the labeled protein using a Sephadex G-25 column equilibrated in phosphate-buffered saline, pH 7.4.
EXAMPLE 21 SYNTHESIS OF FOLATE-MOMORDIN AND FOLATE-GELONIN
The proteins momordin and gelonin were purchased from Sigma (St. Louis, MO). Folate-cys pyridyldisulfide was prepared by reacting folate-cys with Aldrithiol (Sigma, St. Louis, MO). Both proteins were dissolved in 0.1M HEPPS buffer, pH 8.2. A 6-fold molar excess of Trouts reagent (Aldrich St. Louis, MO) dissolved in DMSO (16 mM) was added to each protein solution. The solutions were allowed to react for 1 hour at room temperature. Unreacted material was separated from the protein using a Sephadex G-25 column equilibrated in 0.1M phosphate buffer, pH 7.0. Ellmans test for the presence of free thiols were positive for both proteins. While the protein solution was on ice, a 5-fold molar excess of folate-cys pyridyldisulfide dissolved in DMSO was added. The resulting solution was warmed up to room temperature and reacted for 30 minutes. Unreacted material was separated from the labeled protein using a Sephadex G-25 column equilibrated in phosphate- buffered saline, pH 7.4.
EXAMPLE 22 PREPARATION OF FOLATE-TARGETED CLODRONATE OR PREDNISOLONE
PHOSPHATE LIPOSOMES
Liposomes were prepared following methods by Leamon et al. in Bioconjugate Chemistry 2003, 14; 738-747. Briefly, lipids and cholesterol were purchased from Avanti Polar Lipids (Alabaster, AL). Folate-targeted liposomes consisted of 40 mole % cholesterol, 5 mole % polyethyleneglycol (Mr~2000)- derivatized phosphatidylethanolamine (PEG2000-PE, Nektar AL, Huntsville, AL), 0.03 mole % folate-cysteine-PEG3400-PE and 54.97 mole % egg phosphatidylcholine. Lipids in chloroform were dried to a thin film by rotary evaporation and then rehydrated in PBS containing either clodronate (250 mg/ml) or prednisolone phosphate (100 mg/ml). Rehydration was accomplished by vigorous vortexing followed by 10 cycles of freezing and thawing. Liposomes were then extruded 10 times through a 50 nm pore size polycarbonate membrane using a high- pressure extruder (Lipex Biomembranes, Vancouver, Canada). The liposomes were separated from unencapsulated clodronate or prednisolone phosphate by passage through a CL4B size exclusion column (Sigma, St. Louis, MO) in PBS. Average particle size was between 70 and 100 nm.
EXAMPLE 23 FOLATE-FITC BINDING TO ENDOTHELIAL PROGENITOR CELLS
Folate-FITC binding to CDl 33+ FIkI+ endothelial progenitor cells and binding of antibodies to Flkl, CDl 15, CD69, Cl Ib, CD8a, and CD25 markers on endothelial progenitor cells was quantified. Endothelial progenitor cells were isolated as described in Example 14 and folate-FITC and antibody binding and flow cytometry were performed as described in Example 15. As shown in Fig. 1, Flkl, CDl 15, CD69, CD8a, and CD25 markers are co-expressed with the folate receptor on the progenitor cells.
EXAMPLE 24
FOLATE-FITC BINDING TO ENDOTHELIAL PROGENITOR CELLS Folate-FITC binding to CD133+ FIkI+ endothelial progenitor cells and binding of antibodies to CD62L, CD80, CD86, CD44, CD23, and CD14 markers on endothelial progenitor cells was quantified. Endothelial progenitor cells were isolated as described in Example 14 and folate-FITC and antibody binding and flow cytometry were performed as described in Example 15. As shown in Fig. 2, CD62L, CD80, CD86, CD23, and CD 14 markers are co-expressed with the folate receptor on CD133+ FIkI+ endothelial progenitor cells.
EXAMPLE 25 FOLATE-FITC BINDING TO ENDOTHELIAL PROGENITOR CELLS
Folate-FITC binding to CD133+ FIkI+ endothelial progenitor cells and binding of antibodies to Ly-6, F4/80, CD49d, CD 16.2/32.2, and MHC Class II markers on endothelial progenitor cells was quantified. Endothelial progenitor cells were isolated as described in Example 14 and folate-FITC and antibody binding and flow cytometry were performed as described in Example 15. As shown in Fig. 3, Ly- 6, F4/80, CD49d, and CDl 6.2/32.2 markers are co-expressed with the folate receptor on the progenitor cells. EXAMPLE 26 FOLATE-FITC BINDING TO ENDOTHELIAL PROGENITOR CELLS
Folate-FITC binding to CD133+ FIkI+ endothelial progenitor cells and binding of antibodies to CDl 33, FIk-I, and CD44 markers on endothelial progenitor cells was quantified. Endothelial progenitor cells were isolated as described in Example 14 and folate-FITC and antibody binding and flow cytometry were performed as described in Example 15. As shown in Fig. 4, CD133, FIk-I , and CD44 markers are co-expressed with the folate receptor CDl 33+ FIkI+ endothelial progenitor cells. As also shown in Fig. 4, folate-FITC bound to CD133+ FIkI+ endothelial progenitor cells in the absence of unlabeled folic acid and binding was competed in the presence of a 100-fold excess of unlabeled folic acid.
EXAMPLE 27 FOLATE-FITC BINDING TO ENDOTHELIAL PROGENITOR CELLS
Folate-FITC binding to CDl 33+ FIkI+ endothelial progenitor cells and binding of antibodies to Ly-6, CD25, and CD62-L markers on endothelial progenitor cells was quantified. Endothelial progenitor cells were isolated as described in Example 14 and folate-FITC and antibody binding and flow cytometry were performed as described in Example 15. As shown in Fig. 5, Ly-6, CD25, and CD62- L markers are co-expressed with the folate receptor on CD133+ FIkI+ endothelial progenitor cells. As also shown in Fig. 5, folate-FITC bound to CDl 33+ FIkI+ endothelial progenitor cells in the absence of unlabeled folic acid and binding was competed in the presence of a 100-fold excess of unlabeled folic acid.
EXAMPLE 28
SOLID PHASE SYNTHESIS OF FOLATE CONJUGATES The precursor of folate, N10-TFA-Pteroic acid was synthesized according to standard procedures. Fmoc-Lys(Mtt)-Wang resin was soaked in DMF for 20 minutes with nitrogen bubbling before the reaction. 20% piperidine was added to cleave the Fmoc protective group. 2.5 e.q. Fmoc-Glu-OtBu, HOBT and HBTU, dissolved in DMF, as well as 4 e.q. DIPEA were added to the reaction funnel. After 2 hours of nitrogen bubbling at room temperature, the Fmoc cleavage step was repeated with 20% piperidine. 1.5 e.q. NI0-TFA-Pteroic acid and 2.5 e.q. HOBT and HBTU, dissolved in 1:1 DMF/DMSO (dimethylformamide/dimethylsulfoxide), as well as 4 e.q. DIPEA were then added to the reaction for 4 hours with bubbling with nitrogen. The product was then washed with DMF, DCM (dichloromethane), methanol and isopropyl alcohol thoroughly and dried under nitrogen. 1% TFA/DCM (trifluoroacetic acid/dichloromethane) was used to cleave the Mtt (Mtt = 4-methyl- trityl) group. 2.5 e.q. Rd-ITC, dissolved in DMF, and 4 e.q. DIPEA were added to the resin and reaction was carried out at room temperature overnight under reduced light conditions. Cleavage of the conjugates was achieved by TFA:TIPS:H2O (95:2.5:2.5). The crude product was collected by precipitation with cool ether. The crude product was lyophilized overnight. On the second day, the crude product was hydrolyzed using 10% ammonium hydroxide (pH=10) for 45 minutes with nitrogen bubbling. The product was collected by lyophilization. Purification was carried out using preparative HPLC (Rigel).
EXAMPLE 29 SYNTHESIS OF FOLATE OREGON GREEN 488
Figure imgf000042_0001
DIPEA/ DMF
Figure imgf000042_0002
1M HOBt in DCMH-FE (1:1) to resin swollen in DCM
Figure imgf000042_0003
1) Oregon Green 488 (HOBt), DIPEAfDMF
Figure imgf000042_0004
2) TFA/TIPS/H2O (95:2.5:2.5)
3) NH4OH(aq)
Figure imgf000042_0005
N10 TFA- Pteroic acid was synthesized as follows. A universal folate resin was synthesized using Universal NovaTag™ resin (Novabiochem; Catalog # 04- 12-3910). After swelling the resin in DCM (Dichloromethane) for one hour and then with DMF (N,N-Dimethylformaτnide) for thirty minutes, deprotection of the Fmoc (Fluorenlmethyloxycarbonyl) protecting group was achieved by using a solution of 20% piperidine in DMF. Then Fmoc-Glu-OtBu (three-fold molar excess) was coupled to the deprotected secondary amine using HATU [2-(lH-7-Azabenzotriazol- 1-yl) — 1 ,1,3,3-tetramethyl uronium hexafluorophosphate] (three-fold molar excess) and DIPEA (N,N-Diisopropylethylamine) (ten-fold molar excess) in DMF. After thorough washing of this resin, the Fmoc on GIu was removed as described above and N10-TFA Pteroic acid was coupled using standard Fmoc solid phase peptide synthesis (SPPS) procedures. Next, the pendant Mmt (4-Methoxytrityl) was removed with IM HOBT (1-Hyroxybenzotriazole) in DCM/ TFE (Trifluoroethanol). At this point the resin can be washed with DMF and used immediately for further synthesis or washed sequentially with DCM5DMF and MeOH (Methanol), and dried for later use. To the deprotected, amine reactive universal folate resin, a 1.5-fold molar excess of Oregon Green 488 carboxylic acid succinimidyl ester 6 isomer (O-6149) and a 3-fold molar excess of DIPEA was allowed to react for 12 h at room temperature. The resin was next exhaustively rinsed with DMF3 DCM, and methanol and dried for 2 hours. The Folate-Oregon Green 488 was then cleaved from the resin with a 95% trifluoroacetic acid-2.5% water-2'.5% triisopropylsilane solution. Diethyl ether was used to precipitate the product, and the precipitant was collected by centrifugation. The product was washed twice with diethyl ether and dried under vacuum overnight. To remove the N10-trifluoracetyl protecting group, the product was dissolved in a 10% ammonium hydroxide solution and stirred for 30 min at room temperature. The product was precipitated with combined isopropanol and ether, and the precipitant was collected by centrifugation. The product was purified by reverse-phase HPLC on a Cl 8 column at a flow rate of 1 ml/min. The mobile phase, consisting of 10 mM NH4HCO3 buffer, pH 7.0 (eluent A) and acetonitrile (eluent B), was maintained at a 99: 1 A:B ratio for the first minute and then changed to 1 :99 A:B in a linear gradient over the next 29 minutes. The product was confirmed by MS and NMR. EXAMPLE 30 SYNTHESIS OF FOLATE DYLIGHT 680
Figure imgf000044_0001
Dylight 680 Maleimide (Pierce) was dissolved in dimethyl sulfoxide
(DMSO) (1 mg in 100 uL DMSO). A 3-fold molar excess of Folate-Asp-Arg-Asp- Asp-Cys (synthesized as previously described: Bioorganic & Medicinal Chemistry Letters, Volume 16, Issue 20, 15 October 2006, Pages 5350-5355) was added to the solution and mixed for 4 hours at room temperature. Folate-Dylight 680 was purified by reverse-phase HPLC on a C 18 column at a flow rate of 1 ml/min. The mobile phase, consisting of 10 mM NH4HCO3 buffer, pH 7.0 (eluent A) and acetonitrile (eluent B), was maintained at a 99: 1 A:B ratio for the first minute and then changed to 1 :99 A:B in a linear gradient over the next 29 minutes. The product was confirmed by MS and NMR ((C83H103Ni9O3OS4)2", exact Mass: 1973.60; molecular weight: 1975.08; C, 50.47; H, 5.26; N, 13.47; 0, 24.30; S, 6.49). EXAMPLE 31 SYNTHESIS OF RHODAMINE PEG CONJUGATES
Figure imgf000045_0001
a) piperidine, DMF; b) Fmoc-Glu-OtBu (1), HOBt, HBTU, DIPEA, DMF; c) Pteroic acid (2), HOBt, HBTU5 DIPEA, DMF; d) 1% TFA, CH2Cl2; e) Fmoc-Lys(Mtt)-OH
(3), HOBT, HBTU, DIPEA, DMF; f) S-Trityl-3-mercaptopropionic acid (4), HOBT, HBTU, DIPEA, DMF; g) Rhodamine-ITC (5), DIPEA; h) 94.5% TFA, 2.5 % H2O5
2.5 % EDT; 1% TIS; i) NH4OH.
In the preceding scheme, R represents the following:
Figure imgf000045_0002
Synthesis of Folate-Rhodamine-SH as a PEG- Anchor
Standard Fmoc peptide chemistry was used to synthesize a folate linked to Rhodamine B-isothiocyanate via a spacer composed of two lysines attached to the γ-COOH terminal of folic acid. The sequence Lys-Lys-(γ)Glu-pteroic acid was constructedby Fmoc chemistry with HBTU and HOBT (Novabiochem, San Diego, CA) as the activating agents along with diisopropyethylamine (DIPEA) as the base. The Fmoc groups were deprotected with 20% piperidine in dimethylformamide (DMF). ct-Fmoc-protected lysine-loaded Wang resin, containing a 4-methyltrityl protecting group on the ε-amine, was used as an anchor for folate. An Fmoc-Glu- O/Bu was linked to the α-amine of the lysine to provide a γ-linked conjugate of folate after N10-trifluoroacetylpteroic acid (SIGMA, St. Louis, MO) was attached to the glutamic acid amine. The methoxytrityl (Mtt) protecting group on the ε-amine of lysine was removed with 1% trifluoroacetic acid in dichloromethane to allow attachment of a second Fmoc-Lys(Mtt)-OH. After removing the Mtt-protecting group of the second lysine, S-Trityl-protected 3-mecaptopropionic acid was coupled to the cx-amine of the second lysine, using the coupling reagents, HOBT and HBTU as described above. Finally, the Mtt-protecting group of the second lysine was removed and Rhodamine-B isothiocyanate (SIGMA, St. Louis, MO) dissolved in DMF was reacted overnight with the peptide in the presence of DIPEA, and then washed thoroughly from the peptide resin beads. The resin was washed several times with dichloromethane, and methanol and left to dry under N2 for several hours. The folate- Lys-Lys-mercaptopropionic acid-rhodamine peptide was then cleaved from the resin with 95% TFA / 2.5 H2O / 2.5% TIS / 2.5% EDT solution for 3-4 hours. Ice cold diethyl ether was used to precipitate the product, and the precipitant was collected by centrifugation. The product was then washed three times with diethyl ether and dried under vacuum. To remove the N10-trifiuoracetyl protecting group from the folate moiety, the product was dissolved in 10% ammonium hydroxide solution and stirred for 30 min at room temperature under argon to prevent disulfide bonds from forming. The product was then lyophilized until dry and stored under argon. The product was confirmed by mass spectroscopic analysis ([JWT] calculated, 1286.5; found, 1285.08).
Synthesis of Folate-PEG(5k)-Rhodamine, Folate-PEG(20k)-Rhodamine, and Folate- PEG(60k)-Rhodamine The folate-rhodamine-SH anchor, synthesized as described above, was used to react with maleimide-activated PEG(5k), PEG(20k), or PEG(60k) (Nektar Therapeutics, San Carlos, CA). The PEG-MAL molecules were dissolved in PBS and a 5 -fold molar excess of folate-rhodamine-SH was added to the solution and stirred ovemight, at room temperature, under nitrogen. The non-reacted folate-rhodamine was then separated from the folate-PEG-rhodamine conjugate by gel filtration chromatography, using a coarse Sephadex G-50 column equilibrated in water, (fractionation range for globular proteins: 1,500-30,000, SIGMA, St. Louis, MO), and using gravity for running the samples. The folate-PEG-rhodamine peak was collected, lyophilized and re-suspended in phosphate buffered saline (PBS) for animal studies.
Characterization of the Molecular Weight of Folate-PEG-Rhodamine Conjugates In order to characterize the apparent molecular weight of the folate-
PEG-rhodamine conjugates, their Ve/Vo ratio was compared with the Ve/Vo of protein standards of known molecular weight (Ve is the elution volume, and V0 is the void volume). Columns were run in phosphate buffered saline (PBS, pH 7.4), at room temperature, at a flow rate of 5 ml/min. The void volume of the column (V0) was determined spectrophotometrically by the elution volume for blue dextran (molecular weight approx. 2,000,000, SIGMA, St. Loius, MO) at 610 ran, by measuring the volume of effluent collected from the point of sample application to the center of the effluent peak. Individual protein standards were dissolved in the PBS and their elution time was followed by absorbance readings at 280 nm. The elution volume (Ve) of the protein standards was determined by measuring the volume of effluent collected from the point of sample application to the center of the effluent peak. In order to determine the Veof the folate-PEG-rhodamine conjugates, samples were applied on the column and ran at the same flow rate as used for blue dextran and the protein standards. The Ve of the folate-PEG-rhodamine conjugates was determined using the same method applied to the standards. Plotting the logarithms of the known molecular weights of protein standards versus their respective Ve/Vo values produces a linear calibration curve. Two different Sephacryl HR columns were used for the purpose of resolving all the folate-PEG-conjugates. A 24 cm x 1.0 cm Sephacryl 100- HR (MW range 1000-10,000 Da) was able to resolve the folate-PEG(5k)-rhodamine conjugate, but not the folate-PEG(20k)-rhodamine and folate-PEG(60k)-rhodamine conjugates. The latter two conjugates were resolved on a 22 cm x 1.0 cm Sephacryl 200-HR (MW range 5-250 kDa). The protein standards used on the Sephacryl 100- HR were: bradykinin fragment 2-9 (MW -904), aprotinin from bovine lung (MW 6,511.44), myoglobin from horse heart (MW -17,000), carbonic anhydrase from bovine erythrocytes (MW -29,000), albumin (MW -66,000), aldolase (MW -161,000). The protein standards used on Sephacryl 200-HR were: myoglobin from horse heart (MW ~17,000), carbonic anhydrase from bovine erythrocytes (MW -29,000), albumin (MW -66,000), alcohol dehydrogenase from yeast (MW
-150,000), /3-amylase from sweet potato (MW -200,000), apoferritin from horse spleen (MW -443,000), bovine thyroglobulin (MW -669,000).
Characterization of Folate/Rhodamine Ratio for Folate-PEG-Rhodamine Conjugates In order to determine the ratio of folate to rhodamine on all the folate-
PEG-rhodamine conjugates, first the extinction coefficients of folic acid and rhodamine-isothiocyanate in water were determined at two different wavelengths, 280 nm and 560 nra, by constructing standard curves at both these wavelengths. The slopes of these standard curves correspond to the extinction coefficients of folic acid and rhodamine-isothiocyanate in water. Samples of folate-PEG-rhodamine conjugates were then dissolved in water and their absorbances 280 nm and 560 nm were measured. The absorbances of folate-PEG-rhodamine conjugates at these wavelengths are due to both, the absorbance of folic acid (FA) and rhodamine (Rhod), therefore: A280 = A280(FA) + A280(RhOd) and A560 = A560(FA) + A560(RhOd)
By using the extinction coefficients of folic acid (FA) and rhodamine (Rhod), determined by the standard curves, the concentrations of folate and rhodamine, and thus their ratio, in each folate-PEG-rhodamine conjugate sample can be determined by simultaneously solving for their respective concentrations in the following equations:
A280 = ε28o(FA) 1 c(FA) + ε2go(Rhod) 1 c(Rhod) A56O = S560(FA) 1 c(FA) + ε56o(Rhod) 1 c(Rhod)
EXAMPLE 32
SYNTHESIS OF FOLATE CW800
N -TFA-Pteroic acid was synthesized as reported elsewhere. First, Fmoc-Lys(Mtt)-Wang resin was swelled in DMF for 20 min. The deprotection of Fmoc group on the resin was achieved by 20% piperidine in DMF. 2.5 e.q. Fmoc- (γ)Glu-OtBu, HOBT, HBTU and 4 e.q. DIPEA were added to the reaction. Two hours later, the Fmoc group on glutamic acid was deprotected with 20% piperidine. Then, 2.5 e.q. N10-TFA-Pteroic acid, HOBT and HBTU were dissolved in 3:1 DMF/DMSO and 4 e.q. DIPEA were added to the reaction and reacted for 4 h. The product was washed with DMF, DCM and methanol. 1% TFA/DCM was used to cleave the Mtt protection group. Cleavage of the conjugates was achieved by TFA:TIPS:H2O (95:2.5:2.5). The crude product was then precipitated with cool ether. The crude product was then hydrolyzed with ammonium hydroxide (pH=10) for 20 min. Folate-lysine was purified by HPLC and characterized by MS and NMR. Folate-lysine and CW 800 succinimidyl ester (1 :1) were stirred in 0.1 M carbonate buffer (pH 9.0) in the dark for 18 h. The folate-CW800 conjugates was purified by HPLC and characterized by MS and NMR.
Figure imgf000049_0001
Figure imgf000049_0002
I a) 20% piperidine/DMF. b) Fmoc-GluO/8u( J)/HOBt/HBTU/DlPEA/DMF c> Nlo-;/ff-l«eroic acid(2)/
HOBI/HBTU/DIPF.A/DMF J) I% TFA/DCM c) 95% TFA/2.5% H2O/2.3%TlPS 0 10% NH/OH, μH-lOg) CWSOOyDlPLA/DMSO EXAMPLE 33
SYNTHESIS OF FOLATE ALEXAFLUOR 647
Figure imgf000050_0001
First, H-Cys(Trt)-2-Cl Trt resin was swelled in DMF for 20 min. The deprotection of Fmoc group on the resin was achieved with 20% piperidine in DMF. 2.5 e.q. Fmoc-(γ)Glu-OtBu, HOBT, HBTU and 4 e.q. DIPEA were added to the reaction. Two hours later, the Fmoc group on glutamic acid was deprotected with 20% piperidine. Then, 2.5 e.q. N10-TFA-Pteroic acid, HOBT and HBTU were dissolved in 3 : 1 DMF/DMSO and 4 e.q. DIPEA were added to the reaction and reacted for 4 h. The product was washed with DMF, DCM and methanol. Cleavage of the conjugates was achieved with TFArTIPS=H2O (95:2.5:2.5). The crude product was then precipitated with cool ether. The crude product was hydrolyzed with ammonium hydroxide (pH=10) for 20 min. Folate-cysteine was purified by HPLC and characterized by MS and NMR. Folate-cysteine and AlexaFluor 647 maleimide (Invitrogen, Carlsbad, CA; 1:1) were coupled in DMSO in the dark for 18 h. The folate-AlexaFluor 647 conjugate was purified by HPLC and characterized by MS and NMR.
It is to be understood that the foregoing Examples are merely illustrative of the compounds described herein and additional compounds may be prepared as described herein by the appropriate selection the starting materials, including the dye or fluorescent agent. For example, the following additional compounds are described. EXAMPLE 34 SYNTHESIS OF FOLATE-EDA-RHODAMINE
Figure imgf000051_0001
EXAMPLE 35 FOLATE-EDA-TETRAMETHYLRHODAMINE
Figure imgf000051_0002
Folate-EDA-tetramethylrhodamine was prepared according to the process described above for Example 34.
EXAMPLE 36 SYNTHESIS OF FOLATE-LYS-RHODAMINE
Figure imgf000052_0001
a) piperidine, DMF; b) Fmoc-Glu-OtBu, HOBt, HBTU, DIPEA, DMF; c) Pteroic acid, HOBt, HBTU, DIPEA, DMF; d) 1% TFA, DCM.
EXAMPLE 37 SYNTHESIS OF FOLATE-ALEXAFLUOR 488
Figure imgf000052_0002
HATU, DIPEA/ DMF
1) 20% piperidino in DMF NH 2) N10 TFA-Ptβroic ""
HATU, DIPEA/ DMF
Figure imgf000052_0003
Figure imgf000052_0004

Claims

CLAIMS:
1. A method of quantifying endothelial progenitor cells, said method comprising the steps of isolating the progenitor cells from a patient suffering from a disease state mediated by the progenitor cells; contacting the progenitor cells with a composition comprising a conjugate or complex of the general formula
Ab-X where the group Ab comprises a vitamin, or an analog thereof, that binds to endothelial progenitor cells and the group X comprises a quantifiable marker; and quantifying the percentage of progenitor cells that expresses a receptor for the vitamin.
2. The method of claim 1 wherein the progenitor cells are CD133
FIkI+ endothelial progenitor cells.
3. The method of claim 1 wherein the progenitor cells are common precursor cells.
4. The method of claim 1 wherein Ab comprises folate, or an analog thereof.
5. The method of claim 1 wherein Ab comprises a CDl 33+ FIkI+ endothelial progenitor cell-binding antibody or antibody fragment.
6. The method of claim 1 wherein the quantifiable marker comprises a metal chelating moiety.
7. The method of claim 6 wherein the quantifiable marker further comprises a metal cation.
8. The method of claim 7 wherein the metal cation is a radionuclide.
9. The method of claim 1 wherein the quantifiable marker comprises a radionuclide.
10. The method of claim 9 wherein the radionuclide is selected from the group consisting of technetium, gallium, indium, and a positron emitting radionuclide.
11. The method of claim 1 wherein the quantifiable marker comprises a fluorescent chromophore.
12. The method of claim 11 wherein the fluorescent chromophore comprises a compound selected from the group consisting of fluorescein, Oregon Green, rhodamine, phycoerythrin, Texas Red, and AlexaFluor 488.
13. The method of claim 1 wherein the patient is suffering from a disease state selected from the group consisting of rheumatoid arthritis, osteoarthritis, ulcerative colitis, Crohn's disease, inflammatory lesions, infections of the skin, osteomyelitis, organ transplant rejection, pulmonary fibrosis, sarcoidosis, systemic sclerosis, lupus erythematosus, glomerulonephritis, restenosis, proliferative retinopathy, cancer, inflammations of the skin, and any chronic inflammation.
14. A use of a composition comprising a conjugate or complex of the general formula
Ab-X where the group Ab comprises a vitamin, or an analog thereof, that binds to the progenitor cells and the group X comprises an antigen, a cytotoxin, or a compound capable of altering progenitor cell function in the manufacture of a medicament for use in treating a disease state worsened by progenitor cells.
15. The use of claim 14 wherein the progenitor cells are CD133+ FIkI+ endothelial progenitor cells.
16. The use of claim 14 wherein the progenitor cells are common precursor cells.
17. The use of claim 14 wherein Ab comprises folate, or an analog thereof.
18. The use of claim 14 wherein Ab comprises a CD133+ FIkI+ endothelial progenitor cell-binding antibody or antibody fragment.
19. The use of claim 14 wherein the group X comprises an antigen.
20. The use of claim 17 wherein the group X comprises an antigen.
21. The use of claim 14 wherein the group X comprises a cytotoxin.
22. The use of claim 21 wherein the group X further comprises a liposome.
23. The use of claim 17 wherein the group X comprises a cytotoxin.
24. The use of claim 23 wherein the group X further comprises a liposome.
25. The use of claim 14 wherein X comprises a compound capable of altering progenitor cell function.
26. The use of claim 25 wherein the compound capable of altering progenitor cell function is a cytokine.
27. The use of claim 17 wherein the patient is suffering from a disease state selected from the group consisting of rheumatoid arthritis, osteoarthritis, ulcerative colitis, Crohn's disease, inflammatory lesions, infections of the skin, osteomyelitis, organ transplant rejection, pulmonary fibrosis, sarcoidosis, systemic sclerosis, lupus erythematosus, restenosis, proliferative retinopathy, cancer, glomerulonephritis, inflammations of the skin, and any chronic inflammation.
28. The use of claim 17 wherein X comprises a compound capable of altering progenitor cell function.
29. The use of claim 28 wherein the compound capable of altering progenitor cell function is a cytokine.
30. The use of claim 14 wherein the patient is suffering from a disease state selected from the group consisting of rheumatoid arthritis, osteoarthritis, ulcerative colitis, Crohn's disease, inflammatory lesions, infections of the skin, osteomyelitis, organ transplant rejection, restenosis, proliferative retinopathy, cancer, pulmonary fibrosis, sarcoidosis, systemic sclerosis, lupus erythematosus, glomerulonephritis, inflammations of the skin, and any chronic inflammation.
31. A method of quantifying endothelial progenitor cells, said method comprising the steps of contacting the progenitor cells in a patient suffering from a disease state mediated by the progenitor cells with a composition comprising a conjugate or complex of the general formula
Ab-X where the group Ab comprises a vitamin, or an analog thereof, that binds to endothelial progenitor cells and the group X comprises a quantifiable marker; and quantifying the percentage of progenitor cells that expresses a receptor for the vitamin.
32. The method of claim 31 wherein the progenitor cells are CD133+ FIkI+ endothelial progenitor cells.
33. The method of claim 31 wherein the progenitor cells are common precursor cells.
PCT/US2007/012269 2006-05-23 2007-05-23 Imaging and therapeutic method using progenitor cells WO2007139815A2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US12/301,864 US20100226967A1 (en) 2006-05-23 2007-05-23 Imaging and therapeutic method using progenitor cells
US13/910,306 US20130266964A1 (en) 2006-05-23 2013-06-05 Method of detecting endothelial progenitor cells

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US80264806P 2006-05-23 2006-05-23
US60/802,648 2006-05-23

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/910,306 Division US20130266964A1 (en) 2006-05-23 2013-06-05 Method of detecting endothelial progenitor cells

Publications (2)

Publication Number Publication Date
WO2007139815A2 true WO2007139815A2 (en) 2007-12-06
WO2007139815A3 WO2007139815A3 (en) 2008-06-19

Family

ID=38657845

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/012269 WO2007139815A2 (en) 2006-05-23 2007-05-23 Imaging and therapeutic method using progenitor cells

Country Status (2)

Country Link
US (2) US20100226967A1 (en)
WO (1) WO2007139815A2 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009016181A2 (en) * 2007-07-31 2009-02-05 Ge Healthcare Limited Optical imaging agents
WO2013084208A3 (en) * 2011-12-07 2013-11-07 Universidade Do Minho Liposomes and corresponding production method
WO2014143309A1 (en) * 2013-03-15 2014-09-18 Purdue Research Foundation Fluorescence imaging of inflammatory diseases
US9315506B2 (en) 2007-04-11 2016-04-19 Merck & Cie 18F-labelled folates
CN106279212A (en) * 2016-08-01 2017-01-04 康宏耀源(天津)科技有限公司 With folic acid be the photosensitive drug of targeting group synthesis and application
CN108586506A (en) * 2018-05-23 2018-09-28 山西大学 A kind of glimmering vinyl coumarin derivative of fluorine boron and its synthetic method and application
CN109776788A (en) * 2017-11-14 2019-05-21 博瑞生物医药(苏州)股份有限公司 Folacin receptor targets multi-arm conjugate
US10377756B2 (en) 2007-04-11 2019-08-13 Merck & Cie 18F-labelled folates

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160082126A1 (en) * 2013-05-13 2016-03-24 Tufts University Nanocomplexes for delivery of saporin
MX2018000840A (en) * 2015-07-23 2018-05-04 Purdue Research Foundation Rapid concentration, recovery and detection of pathogens in food samples.
WO2018026965A1 (en) 2016-08-02 2018-02-08 Isi Life Sciences, Inc. Compositions and methods for detecting cancer cells in a tissue sample
US20180036312A1 (en) * 2016-08-02 2018-02-08 ISI Life Sciences Inc. Novel Scaffolds for Intracellular Compound Delivery for the Detection of Cancer Cells
WO2018101473A1 (en) * 2016-12-02 2018-06-07 国立大学法人東京大学 Compound, folate receptor visualization fluorescent probe, and use of these
US10753942B2 (en) 2017-05-15 2020-08-25 Indicator Systems International, Inc. Methods to detect remnant cancer cells
WO2023192653A1 (en) * 2022-04-02 2023-10-05 The General Hospital Corporation Probes for fluorescence imaging

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004110250A2 (en) * 2003-05-30 2004-12-23 Purdue Research Foundation Diagnostic method for atherosclerosis

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4713249A (en) * 1981-11-12 1987-12-15 Schroeder Ulf Crystallized carbohydrate matrix for biologically active substances, a process of preparing said matrix, and the use thereof
US5266333A (en) * 1985-03-06 1993-11-30 American Cyanamid Company Water dispersible and water soluble carbohydrate polymer compositions for parenteral administration of growth hormone
US5108921A (en) * 1989-04-03 1992-04-28 Purdue Research Foundation Method for enhanced transmembrane transport of exogenous molecules
US5688488A (en) * 1989-04-03 1997-11-18 Purdue Research Foundation Composition and method for tumor imaging
US5417982A (en) * 1994-02-17 1995-05-23 Modi; Pankaj Controlled release of drugs or hormones in biodegradable polymer microspheres
US7033594B2 (en) * 2000-03-31 2006-04-25 Purdue Research Foundation Method of treatment using ligand-immunogen conjugates
WO2002087424A2 (en) * 2001-05-02 2002-11-07 Purdue Research Foundation Treatment and diagnosis of macrophage mediated disease
EP2529758A3 (en) * 2003-01-27 2013-01-02 Endocyte, Inc. Vitamin receptor binding drug delivery conjugates

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004110250A2 (en) * 2003-05-30 2004-12-23 Purdue Research Foundation Diagnostic method for atherosclerosis

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CHENSUE S W ET AL: "BIOLOGIC AND IMMUNOHISTOCHEMICAL ANALYSIS OF MACROPHAGE INTERLEUKIN-1-ALPHA INTERLEUKIN-1-BETA AND TUMOR NECROSIS FACTOR PRODUCTION DURING THE PERITONEAL EXUDATIVE RESPONSE" JOURNAL OF LEUKOCYTE BIOLOGY, vol. 46, no. 6, 1989, pages 529-537, XP002468231 ISSN: 0741-5400 *
RIBATTI DOMENICO: "The discovery of endothelial progenitor cells - An historical review" LEUKEMIA RESEARCH, vol. 31, no. 4, April 2007 (2007-04), pages 439-444, XP002468230 ISSN: 0145-2126 *
SHIBUYA MASABUMI: "Differential roles of vascular endothelial growth factor receptor-1 and receptor-2 in angiogenesis" JOURNAL OF BIOCHEMISTRY AND MOLECULAR BIOLOGY, vol. 39, no. 5, September 2006 (2006-09), pages 469-478, XP002468233 ISSN: 1225-8687 *
SHMELKOV SERGEY V ET AL: "AC133/CD133/Prominin-1" INTERNATIONAL JOURNAL OF BIOCHEMISTRY & CELL BIOLOGY, vol. 37, no. 4, April 2005 (2005-04), pages 715-719, XP002468232 ISSN: 1357-2725 *

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9315506B2 (en) 2007-04-11 2016-04-19 Merck & Cie 18F-labelled folates
US10377756B2 (en) 2007-04-11 2019-08-13 Merck & Cie 18F-labelled folates
US9771368B2 (en) 2007-04-11 2017-09-26 Merck & Cie 18F-labelled folates
WO2009016181A3 (en) * 2007-07-31 2010-02-25 Ge Healthcare Limited Optical imaging agents
WO2009016181A2 (en) * 2007-07-31 2009-02-05 Ge Healthcare Limited Optical imaging agents
WO2013084208A3 (en) * 2011-12-07 2013-11-07 Universidade Do Minho Liposomes and corresponding production method
US9393320B2 (en) 2011-12-07 2016-07-19 Universidade Do Minho Liposomes and its production method
CN105120903A (en) * 2013-03-15 2015-12-02 目标实验室有限责任公司 Methods of manufacture and synthesis of amino acid linking groups conjugated to compounds used for targeted imaging of tumors
EP2968335A4 (en) * 2013-03-15 2016-10-26 Purdue Research Foundation Synthesis and composition of amino acid linking groups conjugated to compounds used for the targeted imaging of tumors
CN105492905A (en) * 2013-03-15 2016-04-13 普渡研究基金会 Fluorescence imaging of inflammatory diseases
CN105228628A (en) * 2013-03-15 2016-01-06 普渡研究基金会 Synthesis and the compositions of base is connected with the aminoacid for making the compound of cancer target imaging put together
JP2016512814A (en) * 2013-03-15 2016-05-09 オン ターゲット ラボラトリーズ エルエルシー Methods for the production and synthesis of amino acid linking groups conjugated to compounds used for targeted imaging of tumors
JP2016512813A (en) * 2013-03-15 2016-05-09 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
WO2014149069A1 (en) 2013-03-15 2014-09-25 Purdue Research Foundation Synthesis and composition of amino acid linking groups conjugated to compounds used for the targeted imaging of tumors
EP2968614A4 (en) * 2013-03-15 2016-10-19 On Target Lab Llc Methods of manufacture and synthesis of amino acid linking groups conjugated to compounds used for targeted imaging of tumors
EP2972320A4 (en) * 2013-03-15 2016-10-26 Purdue Research Foundation Methods of imaging inflammatory diseases by ligands conjugated to fluorescent compounds
US9233175B2 (en) 2013-03-15 2016-01-12 Purdue Research Foundation Methods of imaging inflammatory diseases by ligands conjugated to fluorescent compounds
WO2014143309A1 (en) * 2013-03-15 2014-09-18 Purdue Research Foundation Fluorescence imaging of inflammatory diseases
US20140271484A1 (en) * 2013-03-15 2014-09-18 Purdue Research Foundation Methods of Imaging Inflammatory Diseases by Ligands Conjugated to Fluorescent Compounds
US9782497B2 (en) 2013-03-15 2017-10-10 Purdue Research Foundation Synthesis and composition of amino acid linking groups conjugated to compounds used for the targeted imaging of tumors
US9789208B2 (en) 2013-03-15 2017-10-17 Purdue Research Foundation Synthesis and composition of amino acid linking groups conjugated to compounds used for the targeted imaging of tumors
CN106279212B (en) * 2016-08-01 2019-06-18 康宏耀源(天津)科技有限公司 It is the synthesis and application for targeting the photosensitive drug of group with folic acid
CN106279212A (en) * 2016-08-01 2017-01-04 康宏耀源(天津)科技有限公司 With folic acid be the photosensitive drug of targeting group synthesis and application
CN109776788A (en) * 2017-11-14 2019-05-21 博瑞生物医药(苏州)股份有限公司 Folacin receptor targets multi-arm conjugate
CN109776788B (en) * 2017-11-14 2021-07-30 博瑞生物医药(苏州)股份有限公司 Folate receptor targeted multi-arm conjugates
CN108586506A (en) * 2018-05-23 2018-09-28 山西大学 A kind of glimmering vinyl coumarin derivative of fluorine boron and its synthetic method and application
CN108586506B (en) * 2018-05-23 2020-04-17 山西大学 Fluoroboron fluorovinylcoumarin derivative and synthetic method and application thereof

Also Published As

Publication number Publication date
US20100226967A1 (en) 2010-09-09
WO2007139815A3 (en) 2008-06-19
US20130266964A1 (en) 2013-10-10

Similar Documents

Publication Publication Date Title
US20100226967A1 (en) Imaging and therapeutic method using progenitor cells
US9731035B2 (en) Method of imaging osteoarthritis using a folate conjugate
US20120270791A1 (en) Methods and Compositions for Treating and Diagnosing Kidney Disease
Kue et al. Small molecules for active targeting in cancer
Leamon et al. Synthesis and biological evaluation of EC72: a new folate-targeted chemotherapeutic
US7740854B2 (en) Treatment of macrophage mediated disease
US8795633B2 (en) Multiphoton in vivo flow cytometry method and device
CN102272159B (en) SPARC binding ScFc
US11730817B2 (en) Pro-cyclic dinucleotides and pro-cyclic dinucleotide conjugates for cytokine induction
Leamon et al. Synthesis and biological evaluation of EC140: a novel folate-targeted vinca alkaloid conjugate
Rana et al. Advancements in folate receptor targeting for anti-cancer therapy: A small molecule-drug conjugate approach
JP5021152B2 (en) Integrin targeting compounds
US20090012009A1 (en) Composition and Method for Treating Inflammatory Disease
ES2759999T3 (en) Minimally toxic prodrugs
US20210330819A1 (en) Design and development of neurokinin-1 receptor-binding agent delivery conjugates
WO2021016082A1 (en) Compositions and methods for treating autoimmune disorders
CA3073631A1 (en) Castration resistant prostate cancer
Lu et al. Folate-targeted dinitrophenyl hapten immunotherapy: effect of linker chemistry on antitumor activity and allergic potential
Samadian et al. Anti-tumor effect of folate-binding protein: in vitro and in vivo studies
Rizvi et al. Peptide-Drug Conjugates: Design, Chemistry, and Drug Delivery System as a Novel Cancer Theranostic
Wang et al. Small Molecule–Drug Conjugates Emerge as a New Promising Approach for Cancer Treatment
US20240115717A1 (en) Chlorotoxin derivatives and use thereof
Rizvi et al. Engineering peptide drug therapeutics through chemical conjugation and implication in clinics
CA3232979A1 (en) Fibronectin-binding peptides for use in tumor or fibrosis diagnosis and therapy
RAPOSO MOREIRA DIAS Synthesis of RGD Peptidomimetic-Drug Conjugates for the Tumor-Targeted Delivery of Cytotoxic Agents

Legal Events

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

Ref document number: 07795218

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 12301864

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 07795218

Country of ref document: EP

Kind code of ref document: A2