US20160136292A1 - Metap-2 inhibitor polymersomes for therapeutic administration - Google Patents

Metap-2 inhibitor polymersomes for therapeutic administration Download PDF

Info

Publication number
US20160136292A1
US20160136292A1 US15/005,184 US201615005184A US2016136292A1 US 20160136292 A1 US20160136292 A1 US 20160136292A1 US 201615005184 A US201615005184 A US 201615005184A US 2016136292 A1 US2016136292 A1 US 2016136292A1
Authority
US
United States
Prior art keywords
metap
fumagillol
inhibitor
acetyl
block copolymer
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/005,184
Inventor
Ofra Benny-Ratsaby
Robert D'Amato
Judah Folkman
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Childrens Medical Center Corp
Original Assignee
Childrens Medical Center Corp
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 Childrens Medical Center Corp filed Critical Childrens Medical Center Corp
Priority to US15/005,184 priority Critical patent/US20160136292A1/en
Publication of US20160136292A1 publication Critical patent/US20160136292A1/en
Priority to US15/262,143 priority patent/US9789199B2/en
Assigned to CHILDREN'S MEDICAL CENTER CORPORATION reassignment CHILDREN'S MEDICAL CENTER CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BENNY-RATSABY, OFRA, D'AMATO, ROBERT
Assigned to CHILDREN'S MEDICAL CENTER CORPORATION reassignment CHILDREN'S MEDICAL CENTER CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FOLKMAN, JUDAH
Assigned to NIH-DEITR reassignment NIH-DEITR CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: BOSTON CHILDREN'S HOSPITAL
Assigned to THE GOVERNMENT OF THE UNITED STATES, AS REPRESENTED BY THE SECRETARY OF THE ARMY reassignment THE GOVERNMENT OF THE UNITED STATES, AS REPRESENTED BY THE SECRETARY OF THE ARMY CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: BOSTON CHILDREN'S HOSPITAL
Abandoned legal-status Critical Current

Links

Images

Classifications

    • A61K47/48215
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/336Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having three-membered rings, e.g. oxirane, fumagillin
    • A61K47/482
    • A61K47/488
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/593Polyesters, e.g. PLGA or polylactide-co-glycolide
    • 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
    • A61K47/6907Medicinal 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 microemulsion, nanoemulsion or micelle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6905Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion
    • A61K47/6911Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion the form being a liposome
    • A61K47/6915Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion the form being a liposome the form being a liposome with polymerisable or polymerized bilayer-forming substances, e.g. polymersomes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • A61K9/1075Microemulsions or submicron emulsions; Preconcentrates or solids thereof; Micelles, e.g. made of phospholipids or block copolymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • 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
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/10Antioedematous agents; Diuretics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers
    • CCHEMISTRY; METALLURGY
    • C01INORGANIC CHEMISTRY
    • C01BNON-METALLIC ELEMENTS; COMPOUNDS THEREOF; METALLOIDS OR COMPOUNDS THEREOF NOT COVERED BY SUBCLASS C01C
    • C01B32/00Carbon; Compounds thereof
    • C01B32/15Nano-sized carbon materials
    • C01B32/158Carbon nanotubes
    • C01B32/16Preparation
    • C01B32/164Preparation involving continuous processes
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates generally to compositions and methods for the treatment of angiogenic diseases and disorders.
  • MetAP-2 methionine aminopeptidase type 2
  • VEGF/VPF vascular endothelial growth factor
  • bFGF basic fibroblast growth factor
  • IL-8 interleukin-8
  • TNP-470 O-(chloracetyl-carbamoyl)fumagillol
  • Fumagillol is the alcohol derived from the hydrolysis of fumagillin 3 , a compound secreted by the fungus Aspergillus fumigatus fresenius .
  • TNP-470 is a potent endothelial inhibitor in vitro 30 . Recently, TNP-470 has been tested as a potential new anticancer agent.
  • TNP-470 is also referred to as AGM-1470, an analog of fumagillin, is among the most potent inhibitors of angiogenesis.
  • AGM-1470 an analog of fumagillin
  • the anti-angiogenic properties of the TNP-470 were tested on different tumor models in animals, and in clinical trials with a variety of malignancies such as prostate, breast, lung and cervical cancer.
  • TNP-470 has the broadest anticancer spectrum of any known agent 32, 31 .
  • TNP-470 inhibited the growth of murine tumors up to 91%, human tumors up to 100% and metastatic tumors up to 100% in mice (reviewed in ref. 31). In most studies, mice were treated at the same optimal dose of 30 mg/kg subcutaneously every other day. In clinical trials TNP-470 has shown evidence of antitumor activity when used as a single agent, with a number of objective responses reported with relapsed and refractory malignancies 17,19,23 .
  • TNP-470 In addition to its anti-angiogenic activities, TNP-470 also inhibits or prevents vascular leakage.
  • Methionine aminopeptidase-2 (MetAP-2) has been identified as the target of anti-angiogenic fumagillol derivatives (e.g., TNP-470). 41 The crystal structure of fumagillin complexed with MetAP-2 was reported by Liu et al. 42
  • TNP-470 is generally administered via injections, for example intramuscular or intravenous delivery systems or by a continuous intravenous infusion given every few days. TNP-470 is also typically administered at high doses for a specific period of time at infrequent time intervals, for example TNP-470 is administered at high doses as a chemotherapeutic. TNP-470 is not generally administered orally.
  • the present invention relates, in part, to compositions and formulations comprising a MetAP-2 inhibitor and to compositions and formulations comprising a MetAP-2 inhibitor associated with a block copolymer, wherein the block copolymer comprises a hydrophobic and hydrophilic moiety.
  • the MetAP-2 inhibitor is associated with a block copolymer comprising a hydrophobic moiety, wherein the hydrophobic moiety is part of a block copolymer comprising hydrophilic and hydrophobic moieties.
  • the block copolymer comprising hydrophilic and hydrophobic moieties forms a micelle.
  • the MetAP-2 inhibitor-comprising compositions and formulations can be used in the treatment of a disease or disorder involving or associated with MetAP-2 expression, including, but not necessarily limited to MetAP-2 overexpression or over-activity. Accordingly, the present invention relates to a method to treat a condition wherein the condition involves or requires MetAP-2 activity for its pathology, comprising administering a composition comprising a MetAP-2 inhibitor formulated as described herein.
  • One aspect of the present invention relates to a composition
  • a composition comprising a formulation of an irreversible MetAP-2 inhibitor, for example a fumagillol analog or derivative that has anti-proliferative activity, where the formulation comprises the MetAP-2 inhibitor associated with a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • the irreversible inhibitor is, for example, a member of the bengamide class of MetAP-2 inhibitors, where the formulation is associated with a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • compositions comprising a formulation of reversible inhibitors, for example bestatin class inhibitors or 3-amino-2-hydroxyamides and related hydroxyamides and acylhydrazines, where the formulation is associated with a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • reversible inhibitors for example bestatin class inhibitors or 3-amino-2-hydroxyamides and related hydroxyamides and acylhydrazines
  • the block copolymer is a diblock copolymer, for example, the fumagillin analogs/derivatives or bengamide analogs thereof are associated with the hydrophobic moiety of a block copolymer such as a diblock copolymer.
  • the fumagillin analog/derivative or bengamide analog is associated with the hydrophobic moiety of such diblock copolymer.
  • the formulation comprises a micelle comprising the block copolymer associated with the MetAP-2 reversible or irreversible inhibitors.
  • composition comprising a methionine aminopeptidase-2 (MetAP-2) inhibitor formulation having anti-proliferative activity, wherein the formulation comprises a MetAP-2 inhibitor covalently linked to a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • MetAP-2 methionine aminopeptidase-2
  • the MetAP-2 inhibitor is a fumagillol derivative.
  • the MetAP-2 inhibitor is an irreversible MetAP-2 inhibitor.
  • MetAP-2 inhibitor is a reversible MetAP-2 inhibitor.
  • block copolymer is a diblock copolymer.
  • MetAP-2 inhibitor is covalently linked with the hydrophobic moiety of the block copolymer.
  • the hydrophobic polymer moiety of the block copolymer is selected from the group consisting of poly(d,L-lactic acid), poly(caprolactone) (PCL), and poly(propylene oxide).
  • the hydrophobic moiety is a poly(d,L-lactic acid) (PLA) polymer.
  • the hydrophobic polymer moiety is 1-15 kDa.
  • the hydrophobic polymer moiety can be between 1-10 kDa, 1-8 kDa, 1-5 kDa, 1-3 kDa, 3-15 kDa, 5-15 kDa, 8-15 kDa, 10-15 kDa, 12-15 kDa, 2-12 kDa, or 4-10 kDa, 6-8 kDa in size.
  • the hydrophobic polymer is approximately 2 kDa.
  • the hydrophilic moiety is a poly(ethylene glycol) (PEG) polymer.
  • the hydrophilic polymer moiety is between 1-15 kDa.
  • a hydrophilic polymer moiety useful in the compositions as described herein can be between 1-10 kDa, 1-8 kDa, 1-5 kDa, 1-3 kDa, 3-15 kDa, 5-15 kDa, 8-15 kDa, 10-15 kDa, 12-15 kDa, 2-12 kDa, 4-10 kDa, 6-8 kDa in size.
  • the hydrophilic polymer is approximately 2 kDa.
  • the polymer is a diblock copolymer comprising a PEG-PLA diblock copolymer having hydrophilic PEG and hydrophobic PLA moieties.
  • the formulation is formulated for oral administration.
  • the formulation is formulated for topical administration.
  • the formulation is formulated for administration by oral, IV, peritoneal, injected (e.g., subcutaneous, intramuscular, etc.), eyedrop or ocular, suppository, topical, pulmonary, including inhaled, and nasal routes, among others, and includes a reversible or irreversible MetAP-2 inhibitor.
  • the anti-proliferative activity is an anti-tumor activity or an activity which normalizes vascular permeability.
  • the MetAP-2 inhibitor has anti-angiogenic activity and is useful in the preparation of a medicament for the treatment of an angiogenesis-mediated condition and/or in a method of treating an angiogenesis-mediated condition.
  • the fumagillol derivative comprises a derivative selected from the group consisting of 6-O—(N-chloroacetylcarbamoyl)fumagillol (TNP-470), 6-O-(4-methoxyaniline)acetyl fumagillol; 6-O-(3,4,5-trimethexyaniline)acetyl fumagillol; 6-O-(4-(N,N-dimethylethoxy)aniline)acetyl fumagillol; 6-O-(cyclopropylamino)acetyl fumagillol; 6-O-(cyclobutylamino)acetyl fumagillol; 4-((cyclopropylamino)acetyl)oxy-2-(1,2-epoxy-1,5-dimethyl-4-hexenyl)-3-methoxy
  • the fumagillol derivative comprises 6-O—(N-chloroacetylcarbamoyl)fumagillol (i.e., TNP-470).
  • the MetAP-2 inhibitor is selected from the group consisting of a bengamide, a sulphonamide MetAP-2 inhibitor compound, a bestatin, a 3-amino-2-hydroxyamide MetAP-2 inhibitor compound, a hydroxyamide MetAP-2 inhibitor compound, an acylhydrazine MetAP-2 inhibitor compound, ovacillin, a reversible MetAP-2 inhibitor and an irreversible MetAP-2 inhibitor.
  • a method of treating a condition in which MetAP-2 activity is required for or involved in the pathology of the condition comprising administering a MetAP-2 inhibitor formulation comprising a MetAP-2 inhibitor having anti-proliferative activity, covalently linked to a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • the condition is an angiogenesis-mediated condition.
  • Angiogenesis mediated conditions include, as non-limiting examples, cancer, metastatic tumors, ascites formation, psoriasis, age-related macular degeneration (AMD), thyroid hyperplasia, preeclampsia, rheumatoid arthritis and osteo-arthritis, Alzheimer's disease, obesity, pleural effusion, atherosclerosis, endometriosis, diabetic/other retinopathies, ocular neovascularizations such as neovascular glaucoma and corneal neovascularization, and IL-2 therapy associated edema and abnormal vascular proliferation of other types.
  • condition involving MetAP-2 activity comprises a tumor activity.
  • the condition is selected from the group consisting of cancer, metastatic tumors, psoriasis, age-related macular degeneration (AMD), thyroid hyperplasia, preeclampsia, rheumatoid arthritis and osteo-arthritis, Alzheimer's disease, obesity, pleural effusion, atherosclerosis, endometriosis, diabetic/other retinopathies, ocular neovascularizations, IL-2 therapy associated edema and other edemas, malaria, SARS, HIV, herpes, lupus, IPF, COPD, asthma, cystic fibrosis, transplant rejection, allergic reaction, multiple sclerosis, bacterial infection, viral infection, and conditions involving or characterized by vascular hyperpermeability, inflammation, and spinal injury.
  • ALD age-related macular degeneration
  • a method of making a diblock copolymer composition comprising a MetAP-2 inhibitor that has anti-proliferative activity, the method comprising conjugating the MetAP-2 inhibitor to a diblock copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • the MetAP-2 inhibitor is a fumagillol derivative that has anti-proliferative and/or anti-angiogenic activity.
  • a diblock copolymer composition comprising a MetAP-2 inhibitor, the composition produced by the methods described herein.
  • the MetAP-2 inhibitor is a fumagillol derivative that has anti-proliferative activity and/or anti-angiogenic activity.
  • the fumagillol derivative comprises 6-O—(N-chloroacetylcarbamoyl)fumagillol (TNP-470) and its metabolites.
  • described herein is the use of any of the MetAP-2 inhibitor block copolymer formulations described herein in the manufacture of a medicament for the treatment of a condition that relies upon or involves MetAP-2 activity for its pathology.
  • the condition comprises a tumor activity.
  • the condition is selected from the group consisting of cancer, metastatic tumors, psoriasis, age-related macular degeneration (AMD), thyroid hyperplasia, preeclampsia, rheumatoid arthritis and osteo-arthritis, Alzheimer's disease, obesity, pleural effusion, atherosclerosis, endometriosis, diabetic/other retinopathies, ocular neovascularizations, IL-2 therapy associated edema and other edemas, malaria, SARS, HIV, herpes, lupus, IPF, COPD, asthma, cystic fibrosis, transplant rejection, allergic reaction, multiple sclerosis, bacterial infection, viral infection, fungal infection and conditions involving or characterized by abnormal vasculature or vascular hyperpermeability, inflammation and spinal injury.
  • ALD age-related macular degeneration
  • a method of treating ocular neovascularization in a subject in need thereof comprising administering a MetAP-2 inhibitor formulation comprising a MetAP-2 inhibitor having anti-angiogenic activity, associated with a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • a method of treating ocular neovascularization in a subject in need thereof comprising administering a composition comprising a formulation of a fumagillol derivative that retains anti-angiogenic activity, the formulation comprising the fumagillol derivative is associated with a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • ocular neovascularization include but are not limited to AMD, ARMD, proliferative diabetic retinopathy (PDR), and retinopathy of prematurity (ROP).
  • PDR proliferative diabetic retinopathy
  • ROP retinopathy of prematurity
  • eye diseases which involves neovascularization, infection and edema.
  • a method of making a diblock copolymer micelle comprising a MetAP-2 inhibitor comprising conjugating the MetAP-2 inhibitor to a diblock copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety and forming micelles of the resulting conjugate.
  • a diblock copolymer micelle comprising a MetAP-2 inhibitor.
  • FIG. 1 shows NMR spectra for a block copolymer and a block copolymer conjugate with TNP-470. Differences in NMR pattern verify the binding of the ethylendiamine and the TNP-470 to the polymer.
  • FIG. 2 shows amine levels of mPEG-PLA-en polymer before and after TNP-470 binding.
  • TNBSA reaction was performed as described in the Examples; levels of 450 nm absorption indicate the levels of free amines.
  • FIG. 3 shows TNP-470 polymersomes' size ranges.
  • a typical DLS graph showing diameter distribution of TNP-470 polymersomes.
  • FIG. 4A shows inhibition of human umbilical vein epithelial cell (HUVEC) proliferation by MetAP-2 inhibitor polymersomes.
  • TNP-470 micelles comprising between 62.5-1000 nM of TNP-470 equivalent
  • WST-1 *p ⁇ 0.0005
  • FIG. 4B shows that HUVEC proliferation was not affected by polymeric micelles without MetAP-2 inhibitor. No significant differences in proliferation were detected, even with a double concentration of carrier than that of the MetAP-2 inhibitor polymersomes.
  • FIG. 5A shows the effect of MetAP-2 inhibitor polymersomes on tumor volume. Tumor volume, measured during 14-18 days of daily oral administration of TNP-470 polymeric micelles to C57/BL mice (*p ⁇ 0.05).
  • FIG. 5B shows the effect of TNP-470 polymersomes on mice weights. Mice were weighed q.o.d, the results are an average weight of mice in each group.
  • FIG. 6A shows a diagram of a conjugation reaction between TNP-470 and modified mPEG-PLA.
  • FIG. 7A shows flow cytometry analysis of uptake of Rhodamine labeled mPEG-PLA micelles by HUVEC (FL2high) at 0 min, 20 min and 24 h of incubation with micelle. Incubation after 2, 4 and 7 h showed a similar pattern as after 24 h of incubation (not shown).
  • FIG. 7C shows the HUVEC growth curve ( ⁇ ) of cells treated q.o.d. with Lodamin (60 nM TNP-470 equivalent) ( ⁇ ) Untreated cells and ( ⁇ ) Vehicle treated cells.
  • FIG. 8A shows the fluorescent signal of tissue extracts and in serum of mice treated with Lodamin.
  • Data show the values of the three different mice, autofluorescence was omitted by subtracting the fluorescent signal of tissue extracts from an untreated mouse. The percent of labeled cells was measured for each organ.
  • FIG. 8B shows the levels of fluorescent signals in mouse serum as measured after different time points post oral administration. The results are presented as the concentration of micelles calculated by standard calibration curve.
  • FIG. 8C shows the percentage of FL2 high positive cells as isolated from the designated organs (ratio of numbers FL2 high cells of treated tumors to those of control mouse).
  • FIG. 8D shows the FACS analysis graphs of single-cell suspensions from three representative organs (tumor, liver, brain) taken from a mouse bearing Lewis lung carcinoma after controlled enzymatic degradation.
  • the FL2 high represents those cells that contain the mPEG-PLA-rhodamine micelles.
  • FIG. 9E shows some representative Lewis lung carcinoma and B16/F10 tumors removed from mice at day 18 post treatment with oral Lodamin at 30 mg/kg q.o.d., and 15 mg/kg e.d. respectively, and from control untreated mice.
  • FIG. 9F shows the neurotoxcity of Lodamin on treated mice (10 d, 30 mg/kg q.o.d.) compared to mice treated with subcutaneous (30 mg/kg q.o.d.) free TNP-470 or water given by gavage.
  • FIG. 9G shows the mice's weight on day 1 and day 18 post treatment with oral polymeric micelle TNP-470 (Lodamin).
  • FIG. 10A shows the livers removed from Lodamin-treated or untreated mice with B16/F10 tumors, 20 d post cell injection. Control livers were enlarged with wide-spread macroscopic malignant nodules and extensive cirrhosis.
  • FIG. 10B shows the spleens removed from Lodamin-treated or untreated mice with B16/F10 tumors. Control spleens had large masses compared to treated mice with normal spleen morphology.
  • FIG. 11B shows the effect of Lodamin given in drinking water at a dose of 15 mg/kg q.d. (solid squares), water with Lodamin was changed every 3 days. *p ⁇ 0.05 (results are mean ⁇ SE). The mice had established human glioblastoma tumors (U87-MG tumors) growth.
  • FIG. 13A shows the effects of Lodamin on angiogenesis in Matrigel implants, the implants were removed 8 days post injection from mice.
  • FIG. 13B shows FACS analysis of the Matrigel incorporated cells in Lodamin-treated and non-treated mice, CD45-CD31+ represents the endothelial cells.
  • FIG. 14 shows schematic drawings of diblock-copolymer that is part hydrophilic and part hydrophobic conjugated to MetAP-2 inhibitors and diblock-copolymer micelles (polymersomes) formed.
  • FIG. 15 shows schematics of exemplary approaches for functionalizing MetAP-2 inhibitors and coupling copolymers to them.
  • FIG. 16A shows that Lodamin inhibits angiogenesis and the inflammatory response in the Matrigel plug angiogenesis assay and DTH in mice.
  • FIG. 16B shows FACS dot plots of endothelial cells (CD31+/CD45) or macrophages (CD45+F4/80+) from Matrigel plugs (marked in the inner squares).
  • FIG. 16E shows the ear swelling is represented as change in thickness (in ⁇ m) compared to original ear thickness.
  • FIG. 17A shows the quantification of vessel area (mm 2 ) in different corneal assays performed with Lodamin which were administered either orally (30 mg/kg q.d), by eye drops (30 mg/ml 1 gtt q.d) or by subconjunctional injections (30 mg/ml, q.d).
  • FIG. 17B shows representative images taken from the different groups at day 5, bFGF pellet is detected as a white spot in the center of the cornea, blood vessels growing from limbal periphery are reduced in Lodamin treated group compared to controls. Vessel area in mm 2 is calculated by the following formula: ⁇ clock hours ⁇ vessel length (mm) ⁇ 0.2 mm.
  • FIG. 18A shows the CNV lesion size of Lodamin-treated mice compared to controls. 15 mg/kg/day was given for 7 or 14 d and dose of 30 mg/kg/day was given for 7 d.
  • FIG. 18C shows the FACS analysis of single-cell suspension from retinas after 3 and 7 d of oral Lodamin treatment. Quantification of macrophage infiltration in mouse retinas originated from Lodamin treated (30 mg/kg, daily, oral) or mice which were treated with equivalent dose of vehicle. Macrophage population was detected as a double positive CD45+ and F4/80+ staining.
  • FIG. 19B shows representative full-field flash ERG responses from the eyes of mice 14 days after intra-ocular injection of vehicle (left) or 300 m/eye Lodamin (right).
  • FIG. 19C shows representative responses to 8 Hz flicker.
  • FIG. 19D shows the 5th to 95th prediction limits for ERG parameters in vehicle treated rats are shown; the dashed line represents the normal mean of all the parameters. Symbols are parameter values of individual Lodamin-treated mice, and represent the same mouse in each column. Data are A Log Normal (eq. 3). Only the saturating amplitude of the rod photoresponse (RmP3) differed significantly after Lodamin.
  • FIG. 20B shows the ELISA quantification of secreted MCP-1 from RPE to the medium post 24 h incubation with Lodamin at different doses (0.1-100 nM TNP-470 equivalent).
  • Factors detected are TNF ⁇ (26 kDa), RAGE-two bands of 35 kDa and ⁇ 50 kDa, HMGB-1 (29 kDa), NFidB (detected as two bands ⁇ 65 kDa), MAPK or phosphorylated MAPK (ERK/p-ERK, bands of 42 kDa) and 3-actin (42 kDa).
  • the samples were analyzed separately for choroids and retinas. (each sample is originated from 6 eyes).
  • FIG. 20D is a zymogram showing MMP-2 and MMP-9 activity in ocular tissue following oral Lodamin treatment of mice (30 mg/kg/day 7 d).
  • FIG. 20E shows the levels of RAGE receptor protein (50 kDa) and its ligand HMGB-1 (29 kDa) in HMVEC cells post TNP-470 treatment at different doses (0.1 nM-100 nM). A dose dependent effect on RAGE expression was detected.
  • FIG. 21B shows that MCP-1 induces vessel permeability in a dose dependent manner. At 10 pg/ml, MCP-1 induced significant vessel leak compared with PBS.
  • FIG. 21C shows that Lodamin reduces whole eye vessel permeability as determined by Evans blue extraction method.
  • Mouse eyes treated with Lodamin (3 h before conducting the assay) were 65% less than the control.
  • compositions and formulations comprising a MetAP-2 inhibitor associated with a block copolymer, wherein the block copolymer comprises a hydrophobic and hydrophilic moiety.
  • the composition comprises associating a MetAP-2 inhibitor with a block copolymer comprising a hydrophobic moiety, wherein the hydrophobic moiety is part of a block copolymer comprising a hydrophilic and a hydrophobic moiety.
  • the block copolymer comprising hydrophilic and hydrophobic moiety forms a micelle.
  • the present invention relates to a method to treat a condition in which MetAP-2 activity is involved in or required for the pathology of the condition, the method comprising administering a composition comprising a MetAP-2 inhibitor formulated as disclosed herein.
  • the composition is formulated for oral administration, although other routes are specifically contemplated.
  • the condition is a proliferative condition and/or an angiogenesis-mediated condition, and the method comprises orally administering a composition comprising a MetAP-2 inhibitor formulated as disclosed herein.
  • Other routes of administration are specifically contemplated, as discussed herein below.
  • Methionine aminopeptidases are essential for removal of the N-terminal methionine from nascent proteins. Following this removal, a variety of molecules are attached to the unstable, open N-terminus, including myristic acid. This acid changes the hydrophobicity of the nascent protein, and causes specific cellular localization and function. In some instances, this function may lead to pathological diseases, as with the oncogene Src. 46 Recently, MetAP-2, one of the 2 major types of aminopeptidases, has itself been identified as a possible oncogene. 47 Its importance in cell proliferation is well documented. 45
  • MetAP-2 inhibitors particularly those in the fumagillol class, are poorly soluble in water (1.9 mg/ml; see U.S. Pat. No. 5,536,623), and as such have low absorption and bioavailability if given orally. Therefore, modification of these MetAP-2 inhibitors is highly desirable from therapeutic, administration and safety perspectives.
  • MetAP-2 inhibitors In order to administer a MetAP-2 inhibitor one must overcome several issues associated with the chemical properties of the small molecule MetAP-2 inhibitors such as their poor solubility in water, low absorption, non-specific bio-distribution and poor bioavailability if administered orally, topically, inhaled, or given via eyedrop. Due to these properties, the vast majority of MetAP-2 inhibitors are currently formulated for administration exclusively via injections (i.e. intramuscularly and intravenously).
  • the inventors have discovered a composition that formulates MetAP-2 inhibitors such as TNP-470, among others, for alternative routes of administration other than injections, by binding the MetAP-2 inhibitor to a diblock copolymer, for example a diblock co-polymer comprising PEG-PLA.
  • a diblock copolymer for example a diblock co-polymer comprising PEG-PLA.
  • block copolymer-MetAP-2 inhibitor conjugates is a highly suitable formulation for oral, topical, inhaled, eyedrop or peritoneal administration. While having advantages for oral or other non-injected routes of administration, is is also contemplated herein that the block copolymer compositions described herein can also be administered by injection (e.g., intramuscular, subcutaneous, etc.).
  • the inventors demonstrate that a MetAP-2 inhibitor was successfully conjugated to a modified PEG-PLA polymer through its amine, and formed nano-size polymersomes.
  • the inventors also demonstrate, using images taken by TEM, that spherical micelles were formed, and size measurement with DLS showed a low range of size distribution around 10 nanometers.
  • the inventors demonstrate rapid uptake by Human Umbilical Vein Endothelial Cells (HUVECs), and when the MetAP-2 inhibitor polymersomes were added, a significant inhibition of HUVEC proliferation was shown (as compared to no effect of the carrier itself).
  • HUVECs Human Umbilical Vein Endothelial Cells
  • mice In-vivo studies done on mice showed a significant inhibition of subcutaneous Lewis Lung Carcinoma tumors with C57/BL mice given a daily oral administration of TNP470 micelles. A dose of 15 mg/kg TNP-470 equivalent showed 63% inhibition without any weight loss to the mice.
  • a MetAP-2 inhibitor such as TNP-470 is useful for formulation as an oral administration formulation and that such a MetAP-2 inhibitor-diblock copolymer conjugate is useful in a method to treat conditions that involve or require MetAP-2 activity for their pathologies, such as cancer, RA, viral infection, bacterial or fungal infection among others, and those associated with abnormal or hyperpermeable vasculature such as edema, age-related macular degeneration, diabetic retinopathy, inflammation or other conditions involving or requiring MetAP-2 activity as described herein or as known in the art.
  • pathologies such as cancer, RA, viral infection, bacterial or fungal infection among others, and those associated with abnormal or hyperpermeable vasculature such as edema, age-related macular degeneration, diabetic retinopathy, inflammation or other conditions involving or requiring MetAP-2 activity as described herein or as known in the art.
  • Lodamin an oral anti-prolierative/anti-angiogenic MetAP-2 inhibitor polymeric drug with potent anti-tumor and anti-metastatic efficacy named Lodamin.
  • the inventors characterized its physicochemical properties and show that this polymeric pro-drug derived from TNP 470 successfully overcomes the drug limitations while retaining its anti-proliferative and antiangiogenic activities.
  • Lodamin is produced by covalent conjugation of TNP 470 to a di-block copolymer: PEG PLA.
  • PEG PLA di-block copolymer
  • the amphiphilic nature of this polymeric drug enables self-assembly of micelles in an aqueous medium 26 .
  • the TNP-470 is located in the core, where it is protected from the acidic environment of the stomach, thus enabling oral availability.
  • advantage is taken of using biocompatible and well characterized polymers 27,28 .
  • Lodamin administered orally, is effectively absorbed in the intestine and accumulates in tumor tissue.
  • the drug significantly inhibits cell proliferation and angiogenesis, as demonstrated by inhibition of HUVEC proliferation, in the corneal micropocket assay, and in mouse tumor models.
  • Lodamin significantly inhibited primary tumor growth as demonstrated in models of melanoma and lung cancer.
  • oral Lodamin successfully prevented liver metastasis of melanoma tumor cells without causing liver-toxicity or other side-effects and led to prolonged mouse survival.
  • Lodamin does not penetrate the blood-brain barrier, and accordingly did not cause neurotoxicity in mice.
  • conjugating MetAP-2 inhibitors to diblock copolymers is useful for formulation such as an oral administration formulation, among others, and that such a fumagillol derivative-diblock copolymer conjugate is useful in a method to treat proliferative disorders, including angiogenesis-mediated conditions and disorders such as cancer and AMD, among many others.
  • Lodamin a polymeric conjugate of TNP-470, is a potent broad spectrum antiangiogenic ophthalmic drug. Lodamin significantly reduced angiogenesis, vascular leakage and inflammation in the murine models of choroidal neovascularization (CNV) and corneal neovascularization. Lodamin was found to suppress angiogenic and inflammatory signals including monocyte chemotactic protein-1 (MCP-1). Additionally, Lodamin reduced levels of the receptor for advanced glycation end products (RAGE) which likely contributes to its broad spectrum of activity (see Example 7, FIGS. 16-21 ).
  • CNV choroidal neovascularization
  • MCP-1 monocyte chemotactic protein-1
  • RAGE receptor for advanced glycation end products
  • a method of treating a condition involving or relying upon MetAP-2 activity for its pathology comprising administering a MetAP-2 inhibitor formulation comprising a MetAP-2 inhibitor having anti-proliferative activity, covalently linked to a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • the condition is selected from the group consisting of cancer, metastatic tumors, psoriasis, age-related macular degeneration (AMD), thyroid hyperplasia, preeclampsia, rheumatoid arthritis and osteo-arthritis, Alzheimer's disease, obesity, pleural effusion, atherosclerosis, endometriosis, diabetic/other retinopathies, ocular neovascularizations, IL-2 therapy associated edema and other edemas, malaria, SARS, HIV, herpes, lupus, IPF, COPD, asthma, cystic fibrosis, transplant rejection, allergic reaction, multiple sclerosis, bacterial infection, viral infection, conditions involving or characterized by vascular hyperpermeability, inflammation, and spinal injury.
  • ALD age-related macular degeneration
  • described herein is a method of treating an angiogenesis-mediated condition, the method comprising orally administering a composition comprising a formulation of a fumagillol derivative that retains anti-angiogenic activity, the formulation comprising the fumagillol derivative is associated with a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • the angiogenesis-mediated condition is selected from a group comprising cancer, metastatic tumors, psoriasis, age-related macular degeneration (AMD), thyroid hyperplasia, preclampsia, rheumatoid arthritis and osteo-arthritis, Alzheimer's disease, obesity, pleural effusion, atherosclerosis, endometriosis, diabetic/other retinopathies, ocular neovascularizations, IL-2 therapy associated edema and other edemas.
  • ALD age-related macular degeneration
  • described herein is a method of treating ocular neovascularization in a subject in need thereof, the method comprising administering a MetAP-2 inhibitor formulation comprising a MetAP-2 inhibitor having anti-proliferative activity, covalently linked to a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • the MetAP-2 inhibitor is a fumagillol derivative.
  • the MetAP-2 inhibitor formulation described herein can be applied to an eye disease or disorder which involves neovascularization, and/or edema.
  • a method of treating ocular neovascularization in a subject in need thereof comprising administering a composition comprising a formulation of a fumagillol derivative that retains anti-angiogenic activity, wherein the formulation comprising the fumagillol derivative is associated with a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • ocular neovascularization diseases or disorders include but are not limited to AMD, ARMD, proliferative diabetic retinopathy (PDR), and retinopathy of prematurity (ROP).
  • composition comprising a methionine aminopeptidase-2 (MetAP-2) inhibitor formulation having anti-proliferative activity, wherein the formulation comprises a MetAP-2 inhibitor covalently linked to a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • MetAP-2 methionine aminopeptidase-2
  • a method of making a diblock copolymer composition comprising a MetAP-2 inhibitor that has anti-proliferative activity, the method comprising conjugating the MetAP-2 inhibitor to a diblock copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • a diblock copolymer composition comprising a MetAP-2 inhibitor, the composition produced by the methods described herein.
  • composition comprising a formulation of a fumagillol derivative that has anti-angiogenic activity, the formulation comprising the derivative associated with a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • a method of making a diblock copolymer micelle comprising a fumagillol derivative that has anti-angiogenic activity comprising conjugating the fumagillol derivative to a diblock copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety and forming micelles of the resulting conjugate.
  • MetAP-2 inhibitor is an irreversible MetAP-2 inhibitor.
  • MetAP-2 inhibitor is a reversible MetAP-2 inhibitor.
  • the block copolymer is a diblock copolymer.
  • the formulation comprises a micelle comprising the block copolymer associated with the fumagillol derivative.
  • the fumagillol derivative thereof is associated with the hydrophobic moiety of the block copolymer.
  • the hydrophobic polymer moiety of the block copolymer is selected from the group consisting of poly(d,L-lactic acid), poly(L-lysine), poly(aspartic acid), poly(caprolactone) (PCL), and poly(propylene oxide).
  • the hydrophobic moiety is a poly(d,L-lactic acid) (PLA) polymer.
  • MetAP-2 inhibitor is associated with the PLA moiety of the diblock copolymer.
  • the hydrophilic polymer moiety of the block copolymer is polyethylene glycol (PEG).
  • PEG polyethylene glycol
  • the PEG polymer is a capped PEG polymer.
  • the block copolymer is a diblock copolymer comprising a PEG-PLA diblock copolymer having hydrophilic PEG and hydrophobic PLA moieties.
  • the anti-angiogenic activity is an anti-tumor activity.
  • the fumagillol derivative comprises a derivative selected from the group consisting of 6-O—(N-chloroacetylcarbamoyl)fumagillol (TNP-470), 6-O-(4-methoxyaniline)acetyl fumagillol; 6-O-(3,4,5-trimethexyaniline)acetyl fumagillol; 6-O-(4-(N,N-dimethylethoxy)aniline)acetyl fumagillol; 6-O-(cyclopropylamino)acetyl fumagillol; 6-O-(cyclobutylamino)acetyl fumagillol; 4-((cyclopropylamino)acetyl)oxy-2-(1,2-epoxy-1,5 20 dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-1 cyclohexanol; and 4-((cyclobutylamin
  • the fumagillol derivative comprises 6-O—(N-chloroacetylcarbamoyl)fumagillol (TNP-470).
  • the formulation comprises a diblock copolymer micelle formed with the diblock copolymer described herein.
  • the fumagillol derivative is associated with the PLA moiety of the diblock copolymer.
  • composition or formulation is administered orally.
  • the composition or formulation is administered by intravitreous injection.
  • the composition or formulation is for topical administration, IV administration, peritoneal administration, injection, ocular administration, suppository administration, pulmonary administration or inhalation, and nasal administration.
  • the composition is administered by intravitreous injection.
  • Metal-2 inhibitor refers to an agent that, at a minimum, inhibits the activity of MetAP-2 by at least 20% in a MetAP-2 assay as described in U.S. Pat. No. 6,548,477 or in U.S. Pat. No. 7,030,262 (which are both incorporated herein by reference), preferably at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or more, up to and including complete or 100% inhibition relative to the absence of such an agent.
  • MetAP-2 inhibitors Reversible and irreversible MetAP-2 inhibitors are known in the art and are encompassed by the term “MetAP-2 inhibitor.” Fumagillol derivatives as described herein that have anti-proliferative activity are MetAP-2 inhibitors as the term is used herein.
  • the fumagillol and ovacilin classes of MetAP-2 inhibitors have a reactive spiroepoxide moiety that reacts to form a covalent bond with the imidazole nitrogen of histidine 231 in the catalytic site of MetAP-2.
  • Molecular modeling with potential irreversible MetAP-2 inhibitors will permit the skilled artisan to readily predict whether a given candidate inhibitor will react with histidine 231 of the enzyme and therefore have the desired inhibitory activity.
  • a rational design approach can be used to prepare reversible MetAP-2 inhibitors, as demonstrated by Wang et al. 43
  • fluagillol derivative encompasses compounds represented by the Formula I below:
  • X is —OH and Y is halogen, or X and Y are linked together to form a oxyrane ring
  • B represents —(C ⁇ O)— or —CH2-
  • R1 represents hydrogen, hydroxyl, —CN; NO2, —CF3; formyl; C 1 -C 4 thioalkyl, acetamido; acetoxy; C 1 -C 6 alkyl, C 1 -C 4 aminoalkyl, C 1 -C 4 alkylaminoalkyl; C 1 -C 4 dialkylaminoalkyl; C 1 -C 6 alkyloxy, C 1 -C 6 aminoalkyloxy; C 1 -C 4 alkylaminoalkoyy, C 1 -C 4 dialkylaminoalkoxy, amino; C 1 -C 6 alkylamino; C 1 -C 4 dialkylamino; C1-C 4 dialkylamino; C1-C4 hyrd
  • Fumagillol derivatives are disclosed in European Patent Application 0354787, 0357061, and 0415294 and Japanese patent application JP-A01-233275 and U.S. Pat. No. 5,290,807, which are incorporated herein in their entirety by reference.
  • Other MetAP-2 inhibitory fumagillol derivatives, such as PPI-2458, are described by Bernier et al., Proc. Natl. Acad. Sci. U.S.A. 101: 10768-10773 (2004).
  • Fumagillol derivatives as described herein have, at a minimum, anti-angiogenic activity when tested, for example, in a HUVEC proliferation assay as known in the art and described herein below.
  • fumagillol derivative refers to the derivatives of fumagillol of Formula I
  • a “fumagillol derivative block copolymer conjugate” refers to such derivatives conjugated to a block copolymer.
  • a “MetAP-2 inhibitor” refers to the inhibitor itself
  • a “MetAP-2 inhibitor block copolymer conjugate” refers to the MetAP-2 inhibitor conjugated to a block copolymer.
  • the term “proliferation” refers to the development of cells that result in unwanted or undesirable physiological consequences, such as with a tumor or inflammation or hyperpermeable, abnormal vasculature, viral infections, bacterial infections and fungal infections.
  • Angiogenesis should be understood to be a proliferative process.
  • the term “proliferation” can apply to the development of blood vessels.
  • Such development is also referred to herein as “angiogenesis.”
  • the term “angiogenesis”, as used herein refers to the sprouting of new blood vessels from pre-existing blood vessels, characterized by endothelial cell proliferation and migration triggered by certain pathological conditions, such as the growth of tumors, metastasis, AMD and arthritis, among others.
  • the term “proliferation” can also apply to the proliferation of viruses, bacteria, fungi, microsporidia, etc. The context of the term will make it clear which type of “proliferation” is being referred to.
  • anti-proliferative activity refers to the property of an agent that inhibits, suppresses or reduces the rate of growth or creation of new, undesired cells or blood vessels in the body in order to combat disease.
  • a compound or agent with anti-proliferative activity as used herein is an agent that inhibits MetAP-2 and leads to the suppression of unwanted proliferating cells, e.g., in unwanted angiogenesis.
  • anti-proliferative activity is an agent that inhibits MetAP-2 and leads to cell death.
  • anti-angiogenesis activity refers to an agent which inhibits or suppresses or reduces the rate of growth or creation of new blood vessels in the body in order to combat disease.
  • a compound or agent with anti-angiogenesis activity as used herein is an agent capable of inhibiting the formation of blood vessels or the formation of vasculature with abnormal or hyperpermeable properties.
  • a disease associated with vascular permeability includes vascular complications of diabetes such as non-proliferative diabetic retinopathy and diabetic nephropathy, nephrotic syndrome, pulmonary hypertension or fibrosis, burn edema, tumor edema, brain tumor edema, IL-2 therapy-associated edema, and other edema-associated diseases, inflammatory disorders, complications from spinal injury, fibrotic disorders, and infections, including viral infections, bacterial infections and fungal infections.
  • diabetes such as non-proliferative diabetic retinopathy and diabetic nephropathy, nephrotic syndrome, pulmonary hypertension or fibrosis, burn edema, tumor edema, brain tumor edema, IL-2 therapy-associated edema, and other edema-associated diseases, inflammatory disorders, complications from spinal injury, fibrotic disorders, and infections, including viral infections, bacterial infections and fungal infections.
  • the term “retains anti-proliferative activity” means that a given compound derived from a MetAP-2 inhibitor has at least 20% of the anti-proliferative activity of that MetAP-2 inhibitor.
  • a given fumagillol derivative will “retain anti-proliferative activity” if it retains at least 20% of the anti-proliferative activity of fumagillol.
  • the term “retains anti-angiogenesis activity” means that a given fumagillol derivative has at least 20% of the anti-angiogenic activity of TNP-470 in a HUVEC assay of angiogenesis as described herein.
  • MetAP-2 inhibitor formulations as described herein can also be used to prevent the leakage of cell proliferation stimulators from blood vessels. That is, MetAP-2 inhibitors can reduce or prevent vascular hyperpermeability, and prevent the accumulation of stimulatory factors at sites of such permeability.
  • the term “inhibit” or “inhibition” means the reduction or prevention of tumor growth and/or tumor metastasis in cancers. Inhibition includes slowing the rate of tumor growth and metastasis.
  • the tumor growth rate can be reduced by about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 100%, about 125%, about 150% or more compared to a control, untreated tumor of the same type.
  • Inhibition also means a reduction in the size of the tumor (including, but not limited to a reduction caused by cytotoxic activity) of at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, up to and including 100% (no tumor) when compared to a control, untreated tumor of the same type.
  • the prevention of tumor growth and/or metastasis means no further increase in the size of the tumors from the time of start of treatment administration.
  • Prevention also means status quo of no new metastatic tumors detected (ie. no further spread of cancer) and/or no increased amount of tumor markers detected by methods known in the art.
  • MetAP-2-mediated condition refers to diseases or disorders in which MetAP-2 activity contributes to the pathology of the condition, whether it involves inappropriate cell proliferation, e.g., inappropriate angiogenesis, or, for example, vascular hyperpermeability resulting from the MetAP-2 activity. While MetAP-2 over-expression or over-activity can be involved, the expression or activity involved in a condition is not necessarily over-expression or over-activity—that is, normal levels of MetAP-2 activity can also contribute to pathological conditions and should be considered a target for therapy in such instances.
  • MetAP-2 activity is involved in or required for the pathology
  • conditions in which MetAP-2 activity is involved in or required for the pathology include, but are not limited to cancer, metastatic tumors, psoriasis, age-related macular degeneration (AMD), thyroid hyperplasia, preeclampsia, rheumatoid arthritis and osteo-arthritis, Alzheimer's disease, obesity, pleural effusion, atherosclerosis, endometriosis, diabetic/other retinopathies, ocular neovascularizations, IL-2 therapy associated edema and other edemas, malaria, SARS, HIV, herpes, lupus, IPF, COPD, asthma, cystic fibrosis, transplant rejection, allergic reaction, multiple sclerosis, bacterial infection, viral infection, and conditions involving or characterized by vascular hyperpermeability.
  • Angiogenesis-mediated conditions are a sub-set of MetAP-2-mediated conditions, diseases or disorders that are the direct result of aberrant blood vessel proliferation (e.g. diabetic retinopathy and hemangiomas, among others known in the art and/or discussed herein).
  • angiogenesis-mediated condition refers to diseases or disorders that are dependent on a rich blood supply and blood vessel proliferation for the disease pathological progression (eg. metastatic tumors) or diseases or disorders that are the direct result of aberrant blood vessel proliferation (e.g. diabetic retinopathy and hemangiomas).
  • Non-limiting examples include abnormal vascular proliferation, neovascularization, hyperpermeability, ascites formation, psoriasis, age-related macular degeneration, retinopathy, thyroid hyperplasia, preeclampsia, rheumatoid arthritis and osteo-arthritis, Alzheimer's disease, obesity, pleural effusion, atherosclerosis, endometriosis, diabetic/other retinopathies, ocular neovascularizations such as neovascular glaucoma and corneal neovascularization.
  • tumor means a mass of transformed cells that are characterized, at least in part, by containing angiogenic vasculature.
  • the transformed cells are characterized by neoplastic uncontrolled cell multiplication which is rapid and continues even after the stimuli that initiated the new growth has ceased.
  • the term “tumor” is used broadly to include the tumor parenchymal cells as well as the supporting stroma, including the angiogenic blood vessels that infiltrate the tumor parenchymal cell mass.
  • a tumor generally is a malignant tumor, i.e., a cancer having the ability to metastasize (i.e. a metastatic tumor), a tumor also can be nonmalignant (i.e. non-metastatic tumor).
  • Tumors are hallmarks of cancer, a neoplastic disease the natural course of which is fatal. Cancer cells exhibit the properties of invasion and metastasis and are highly anaplastic. Tumors are among the angiogenesis-mediated diseases encompassed by the therapeutic methods described herein.
  • tumor is also used in reference to specific types of tumors, e.g., brain tumors including neuroblastoma, medulloblastoma, meningioma and glioblastoma; head and neck cancer, thyroid carcinoma, endocrine tumors, esophageal cancer, small cell and non-small cell lung cancer, colon cancer, rectal cancer, pancreatic cancer, gastric cancer, bladder cancer, hepatic cancer, malignant lymphoma, acute and chronic leukemia, Kaposi's sarcoma, glioma, hemangioma, osteosarcoma, soft tissue sarcoma, malignant melanoma, skin cancer, prostate cancer, breast carcinoma, choriocarcinoma, ovarian cancer, cervical cancer, uterine cancer and mesenchymal tumors, among others.
  • brain tumors including neuroblastoma, medulloblastoma, meningioma and glioblastoma
  • head and neck cancer thyroid
  • non-solid “tumors” can also benefit from the administration of MetAP-2 inhibitors formulated as described herein.
  • the term “metastases” or “metastatic tumor” refers to a secondary tumor that grows separately elsewhere in the body from the primary tumor and has arisen from detached, transported cells, wherein the primary tumor is a solid tumor.
  • the primary tumor refers to a tumor that originated in the location or organ in which it is present and did not metastasize to that location from another location.
  • a “malignant tumor” is one having the properties of invasion and metastasis and generally showing a high degree of anaplasia. Anaplasia is the reversion of cells to an immature or a less differentiated form, and it occurs in most malignant tumors.
  • polymersomes and “polymeric micelles” are used interchangeably herein to refer to the same block copolymer compositions.
  • copolymer also known as “heteropolymer” as used herein refers to a polymer derived from two (or more) monomeric species, as opposed to a homopolymer where only one monomer is used. Copolymerization refers to methods used to chemically synthesize a copolymer.
  • block copolymer refers to the polymer comprising more than one subunit (or oligomer) type, wherein the copolymer comprises regions of a polymer comprising one subunit type adjoined to a polymer region comprising a second subunit type, for example the term block copolymer refers to a copolymer comprised of two or more homopolymer subunits linked by covalent bonds. The union of the homopolymer subunits may require an intermediate non-repeating subunit, known as a junction block.
  • Block copolymers are made up of blocks of different polymerized monomers. Block copolymers are interesting because they can “microphase separate” to form periodic nanostructures. Block copolymers are described in further detail in the section “Copolymers” herein below.
  • diblock copolymer refers to a block copolymer with two distinct blocks.
  • a block copolymer with three distinct blocks is called a triblock copolymers. It is also possible to have tetrablocks, multiblocks, etc.
  • hydrophilic refers to a molecule or portion of a molecule that is typically charge-polarized and capable of hydrogen bonding, enabling it to dissolve more readily in water than in oil or other hydrophobic solvents. Hydrophilic molecules are also known as polar molecules and are molecules that readily absorb moisture, are hygroscopic, and have strong polar groups that readily interact with water. A “hydrophilic” polymer as the term is used herein, has a solubility in water of at least 100 mg/ml at 25° C.
  • hydrophobic refers molecules tend to be non-polar and prefer other neutral molecules and non-polar solvents. Hydrophobic molecules in water often cluster together. Water on hydrophobic surfaces will exhibit a high contact angle. Examples of hydrophobic molecules include the alkanes, oils, fats, and greasy substances in general. Hydrophobic materials are used for oil removal from water, the management of oil spills, and chemical separation processes to remove non-polar from polar compounds. Hydrophobic molecules are also known as non-polar molecules. Hydrophobic molecules do not readily absorb water or are adversely affected by water, e.g., as a hydrophobic colloid.
  • a “hydrophobic” polymer as the term is used herein has a solubility in water less than 10 mg/ml at 25° C., preferably less than 5 mg/ml, less than 1 mg/ml or lower.
  • hydrophobic drug refers to any organic or inorganic compound or substance having biological or pharmacological activity and adapted or used for a therapeutic purpose having a water solubility less than 10 mg/ml.
  • MetAP-2 inhibitors in particular fumagillol derivatives, tend to be hydrophobic drugs.
  • micelle refers to an arrangement of surfactant molecules (surfactants comprise a non-polar, lipophilic “tail” and a polar, hydrophilic “head”).
  • surfactants comprise a non-polar, lipophilic “tail” and a polar, hydrophilic “head”).
  • a micelle has the arrangement in aqueous solution in which the non-polar tails face inward and the polar heads face outward.
  • Micelles are typically colloid particles formed by an aggregation of small molecules and are usually microscopic particles suspended in some sort of liquid medium, e.g., water, and are between one nanometer and one micrometer in size.
  • a typical micelle in aqueous solution forms an aggregate with the hydrophilic “head” regions in contact with surrounding solvent, sequestering the hydrophobic tail regions in the micelle center.
  • This type of micelle is known as a normal phase micelle (oil-in-water micelle).
  • Inverse micelles have the headgroups at the centre with the tails extending out (water-in-oil micelle).
  • Micelles are approximately spherical in shape. Other phases, including shapes such as ellipsoids, cylinders, and bilayers are also possible.
  • the shape and size of a micelle is a function of the molecular geometry of its surfactant molecules and solution conditions such as surfactant concentration, temperature, pH, and ionic strength.
  • the process of forming micellae is known as micellization.
  • terapéuticaally effective amount refers to an amount that is sufficient to effect a therapeutically or prophylactically significant reduction or measurable suppression of a marker or symptom associated with a disease, condition or disorder dependent upon MetAP-2 activity for its pathology when that amount is administered to a typical subject who has such a condition.
  • a therapeutically or prophylatically significant reduction in a marker or symptom is, e.g. about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, up to and including 100%, i.e., no symptoms or a marker at levels characteristic of non-diseased individuals, as compared to a control or non-treated subject.
  • a “therapeutically effective amount” can refer to an amount which directly or indirectly provides a reduction (i.e., at least a 10% reduction, preferably at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95% or more) in the expression or activity of MetAP-2.
  • the term “therapeutically effective amount” refers to the amount that is safe and sufficient to prevent or delay the development and further spread of metastases in cancer patients.
  • the amount can also cure or cause the cancer to go into remission, slow the course of cancer progression, slow or inhibit tumor growth, slow or inhibit tumor metastasis, slow or inhibit the establishment of secondary tumors at metastatic sites, or inhibit the formation of new tumor metastasis.
  • treat refers to both therapeutic treatment and prophylactic or preventative measures (prevention is also understood to refer to a reduction in the likelihood of developing disease, e.g., at least a 20% reduced likelihood), wherein the object is to prevent or slow down the development or spread of disease.
  • beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable.
  • Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • Those in need of treatment include those already diagnosed with disease. Where the disease is, e.g., cancer, those in need of treatment include those likely to develop metastases.
  • compositions or “pharmaceutical composition” are used interchangeably herein and refers to compositions or formulations that usually comprise an excipient, such as a pharmaceutically acceptable carrier that is conventional in the art and that is suitable for administration to mammals, and preferably humans or human cells.
  • excipient such as a pharmaceutically acceptable carrier that is conventional in the art and that is suitable for administration to mammals, and preferably humans or human cells.
  • Such compositions can be specifically formulated for administration via one or more of a number of routes, including but not limited to oral, IV, peritoneal, injected (e.g., subcutaneous, intramuscular, etc.), eyedrop or ocular, suppository, topical, pulmonary, including inhaled, and nasal routes, among others.
  • polymeric drug delivery composition refers to the combination of drug, and block copolymer.
  • the term “medicament” refers to an agent that promotes the recovery from and/or alleviates a symptom of a relevant condition.
  • the “pharmaceutically acceptable carrier” means a pharmaceutically acceptable means to mix and/or deliver the targeted delivery composition to a subject.
  • pharmaceutically acceptable carrier as used herein means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material which solubilizes, stabilizes or otherwise provides a drug formulation with properties necessary to deliver the drug to an individual in a controlled format.
  • a “carrier” can also be involved in carrying or transporting the subject agents from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and is compatible with administration to a subject, for example a human.
  • a diblock copolymer as described herein is a pharmaceutically acceptable carrier as the term is used herein.
  • Other pharmaceutically acceptable carriers can be used in combination with the block copolymer carriers as described herein.
  • a pharmaceutically acceptable carrier does not promote the raising of an immune response to the drug.
  • polymer solution when used in reference to a block copolymer contained in such a solution, refer to water, i.e. aqueous, based composition having such block copolymer, or particularly to copolymer conjugates as described herein, such as a MetAP-2 inhibitor-PLA-PEG conjugate dissolved therein at a functional concentration.
  • Polymer solution includes all free flowing forms of the composition comprising the conjugated copolymers as described herein and water. Polymer solutions act to solubilize the drug in a form that is acceptable for administration at physiologically relevant temperatures (temperatures ⁇ 45° C.).
  • aqueous solution includes water without additives, or aqueous solutions containing additives or excipients such as pH buffers, components for tonicity adjustment, antioxidants, preservatives, drug stabilizers, etc., as commonly used in the preparation of pharmaceutical formulations.
  • additives or excipients such as pH buffers, components for tonicity adjustment, antioxidants, preservatives, drug stabilizers, etc., as commonly used in the preparation of pharmaceutical formulations.
  • drug formulation refers to all combinations of drug with polymer, for example polymer solutions that are mixed with drug to form drug solutions, as well as mixtures of undissolved polymer with drug, i.e. polymeric drug delivery compositions, that are subsequently dissolved into an aqueous environment to form a drug solution.
  • administration refers to the presentation of formulations to humans and animals in effective amounts, and includes all routes for dosing or administering drugs, whether self-administered or administered by medical practitioners. Oral administration is preferred.
  • biodegradable as used herein means the block copolymer can chemically break down or degrade within the body to form nontoxic components.
  • the rate of degradation can be the same or different from the rate of drug release.
  • PLA refers to a polymer derived from the condensation of lactic acid or by the ring opening polymerization of lactide.
  • biodegradable polyesters refers to any biodegradable polyesters, which are preferably synthesized from monomers selected from the group consisting of D, L-lactide, D-lactide, L-lactide, D, L-lactic acid, D-lactic acid, L-lactic acid, glycolide, glycolic acid, ⁇ -caprolactone, ⁇ -hydroxy hexanoic acid, ⁇ -butyrolactone, ⁇ -hydroxy butyric acid, ⁇ -valerolactone, ⁇ -hydroxy valeric acid, hydroxybutyric acids, malic acid, and copolymers thereof.
  • the term “patient” refers to a mammal, including a human, in need of the treatment to be administered.
  • the term “subject” and “individual” are used interchangeably herein, and refer to an animal, for example a human, to whom treatment, including prophylactic treatment, with a composition as described herein, is provided.
  • the term “mammal” is intended to encompass a singular “mammal” and plural “mammals,” and includes, but is not limited: to humans, primates such as apes, monkeys, orangutans, and chimpanzees; canids such as dogs and wolves; felids such as cats, lions, and tigers; equids such as horses, donkeys, and zebras, food animals such as cows, pigs, and sheep; ungulates such as deer and giraffes; rodents such as mice, rats, hamsters and guinea pigs; and bears.
  • the mammal is a human subject.
  • a “subject” refers to a mammal, preferably a human
  • MetAP-2 inhibitors there are a number of MetAP-2 inhibitors known in the art, and MetAP-2 inhibitors of any kind are contemplated for formulation and use as described herein. That is, it is specifically contemplated herein that any of them can beneficially be prepared as block copolymer conjugates for delivery to a subject to treat a disease, disorder or condition related to, associated with or requiring MetAP-2 activity for its pathology.
  • MetAP-2 inhibitors formulated as described herein retain anti-proliferative activity—that is, a MetAP-2 inhibitor formulated with a block copolymer conjugate as described herein will retain a meaningful portion of the anti-proliferative (including, but not limited to anti-angiogenic) activity of the inhibitor alone as measured in a MetAP-2 assay as described herein.
  • MetAP-2 inhibitors including, but not limited to fumagillol derivatives, ovalicin, anthranilic acid sulfonamides (Wang et al., Proc. Natl. Acad. Sci. U.S.A. 105: 1838-1843 (2008)) e.g., A-800141, bengamides, bestatins (including, but not limited to reversible MetAP-2 inhibitors such as A-357300 and others described by Wang et al., Cancer Res. 63: 7861-7869), triazoles (see, e.g., compounds described in U.S. Pat. No.
  • MetAP-2 inhibitors of various classes are described, for example, in U.S. Pat. Nos. 7,348,307, 7,268,111, 7,157,420, 7,105,482, 7,084,108, 7,037,890, 6,919,307, 6,548,477, 6,242,494, 6,288,228, 6,849,757, 6,887,863, 4,831,135, 7,122,345 and 7,030,262 and in U.S. published Patent Applications 20070254843, 20070161570, 20070117758, 20070010452, 20060223758, 20060069028, 20050239878, 20050059585, 20030109671, 20020193298, and 20020151493, the disclosures of each of which are incorporated herein by reference.
  • Each of the MetAP-2 inhibitors described is potentially suitable for formulation as a MetAP-2 inhibitor block copolymer conjugate as described herein, for the treatment of MetAP-2 associated diseases, disorders or conditions.
  • MetAP-2 inhibitors with alcohol functionality can be coupled to PEG-PLA using the same type of synthesis used to functionalize fumagillol (e.g., reaction with chloroacetylisocyanate, followed by reaction with a polymer bearing primary amine functionality).
  • the compound A-357300 can be coupled in this manner after amine protection. See, e.g., FIG. 15 .
  • Similar coupling for can also be used, for example, to couple compounds such as (1-hydroxymethyl-2-methyl-propyl)-carbamic acid -(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methyl-but-2-enyl)-oxiranyl]-1-oxa-spiro[2.5]oct-6-yl ester (see U.S. Pat. No. 6,548,477). Additional fumagillin derivatives with alcohol functionalities permitting such conjugation are described, for example, in U.S. Pat. No. 7,087,768 (Han et al.)—see, e.g., Example 35 therein.
  • MetAP-2 activity are also known in the art. Assays are described, for example in U.S. Pat. Nos. 6,548,477 and 7,030,262. Any such assay can be used to evaluate the MetAP-2 inhibitory activity of a given compound or the activity of a given MetAP-2 inhibitor block copolymer conjugate as described herein. For the avoidance of doubt, however, a MetAP-2 inhibitor will demonstrate MetAP-2 inhibition (by at least 20%, preferably by at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, up to and including 100% (complete inhibition)) at a dosage or concentration that is substantially not toxic to the cells or subject to which it is to be administered, in either of the following two MetAP-2 assays.
  • Recombinant human MetAP-2 is expressed and purified from insect cells as described in Li and Chang, (1996) Biochem. Biophys. Res. Commun. 227:152-159.
  • Various amounts of candidate MetAP-2 inhibitor compound are then added to buffer H (10 mM Hepes, pH 7.35, 100 mM KC 1, 10% glycerol, and 0.1 M Co.sup.2+) containing 1 nM purified recombinant human MetAP-2 and incubated at 37° C. for 30 minutes.
  • buffer H 10 mM Hepes, pH 7.35, 100 mM KC 1, 10% glycerol, and 0.1 M Co.sup.2+
  • a peptide containing a methionine residue e.g., Met-Gly-Met
  • MetAP-2 inhibition is scored relative to a control lacking the candidate inhibitor, and can also be scored relative to a known MetAP-2 inhibitor, e.g. TNP-470 or, for example, TNP-470 block copolymer conjugate as described herein.
  • Assays for the inhibition of catalytic activity of MetAP-2 can be performed in 96-well microtiter plates. Compounds to be tested are dissolved in dimethyl sulfoxide at 10 mM and diluted ten-fold in assay buffer (50 mM HEPES, pH 7.4, 100 mM NaCl). Ten microliters of solution of each compound to be tested for inhibition are introduced into each cell of the plate. Zero inhibition of enzyme activity is taken to be the result obtained in cells in which 10 mL of assay buffer was placed. A mixture totaling 90 mL per well and made up of 84 mL of assay buffer containing 100 mM MnCl 2 , 1 mL of L-amino acid oxidase (Sigma Catalog No.
  • the absorbence at 450 nanometers is measured every 20 seconds over a period of twenty minutes using an automatic plate reader (e.g., from Molecular Devices, California, USA).
  • the Vmax in mOD/min, calculated for each well, is used to represent MetAP-2 activity.
  • the IC 50 for each candidate inhibitor is obtained by plotting the remaining activity versus inhibitor concentrations.
  • pro-drugs may require pre-treatment (e.g., enzymatic or non-specific hydrolysis) before assaying for MEtAP-2 inhibitory activity.
  • derivatives of fumagillol are formulated for oral administration in compositions and methods described herein. It should be understood that anywhere in which the instant description refers to a MetAP-2 inhibitor, it can also be said to be referring to a fumagillol derivative having anti-proliferative and anti-angiogenic activity. Fumagillol derivatives useful in the compositions and methods described herein retain anti-proliferative, including, but not limited to anti-angiogenic, activity—i.e., at least 50% of the anti-proliferative activity of TNP-470, as measured in a HUVEC assay (see below). Numerous fumagillol derivatives meeting this criterion are known in the art. In one embodiment, suitable fumagillol derivatives for use in the compositions and formulations as disclosed herein are described in U.S. Pat. No. 5,290,807 which is incorporated herein in its entirety by reference.
  • suitable fumagillol derivatives for use in the compositions and formulations as disclosed herein are representative of general Formula II as disclosed in U.S. Pat. Nos. 5,166,172; 5,290,807; 5,180,738 and 5,164,410, which are hereby incorporated by reference.
  • fumagillol derivatives for use in the compositions and formulations as disclosed herein are listed in International Patent Application WO03/027104 which is incorporated herein in its entirety by reference.
  • a fumagillol derivative is 6-O—(N-chloroacetylcarbamoyl)fumagillol, also known as TNP-470, a derivative of the fumagillol of Formula III of International Patent Application WO03/027104 shown below.
  • TNP-470 6-O—(N-chloroacetylcarbamoyl)fumagillol
  • TNP-470 The synthesis of TNP-470 is disclosed in U.S. Pat. Nos. 5,180,738 and 5,290,807 which are hereby incorporated herein in their entirety by reference.
  • Fumagillin has been disclosed to be anti-proliferative and to have anti-angiogenic activity in U.S. Pat. No. 5,135,919 which is incorporated herein in its entirety by reference.
  • various fumagillol derivatives have been disclosed to be anti-proliferative and to have anti-angiogenic activity in U.S. Pat. Nos. 5,180,738; 5,164,410; 5,196, 406; 5,166,172; and 5,290,807 which are incorporated herein in their entirety by reference.
  • Other MetAP-2 inhibitory fumagillol derivatives, such as PPI-2458 are described by Bernier et al., Proc. Natl. Acad. Sci. U.S.A.
  • fumagillol derivatives include, for example, but not limited to, O-(3,4-dimethoxycinnamoyl)fumagillol; O-(4-methoxycinnamoyl)fumagillol; O-(3,4,5-trimethoxycinnamoyl)fumagillol; O-(4-Chlorocinnamoyl)fumagillol; 4-(3,4,5-trimethoxycinnamoyl)oxy-2-(1,2-epoxy-1,5-dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-1-cyclohexanol; O-(4-trifluoromethylcinnamoyl)fumagillol; O-(4-nitrocinnamoyl)fumagillol; O-(3, 4-dimethoxy-6-nitrocinnamoyl)fumagillol; O-(4-acetoxycin
  • fumagillol derivatives include derivatives of the formula I, for example, but are not limited to: 1) 6-O-(4-methoxyaniline)acetyl fumagillol; 2) 6-O-(3,4,5-trimethoxyaniline)acetyl fumagillol; 3) 6-O-(2,4-dimethexyaniline)acetyl fumagillol; 4) 6-O-(3,4-dimethoxyaniline)acetyl fumagillol; 5) 6-O-(3,4-dimethoxy-6-nitroaniline)acetyl 5 fumagillol; 6) 6-O-(3,4-dimethexy-6-cyaneaniline)acetyl fumagillol; 7) 6-O-(4-allyloxyaniline)acetyl fumagillol; 8) 6-O-(4-(2-acetoxyethexy)aniline)acetyl fumagillol; 9) 6-O-(3-cyan
  • fumagillol derivatives useful in the methods and compositions as disclosed herein include, for example, but are not limited to 1) 6-O-(4-methoxyaniline)acetyl fumagillol; 2) 6-O-(3,4,5-trimethexyaniline)acetyl fumagillol; 3) 6-O-(4-(N,N-dimethylethoxy)aniline)acetyl fumagillol; 4) 6-O-(cyclopropylamino)acetyl fumagillol; 5) 6-O-(cyclobutylamino)acetyl fumagillol; 6) 4-((cyclopropylamino)acetyl)oxy-2-(1,2-epoxy-1,5 20 dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-1 cyclohexanol; and 7) 4-((cyclobutylamino)acetyl)oxy-2-(1,2-e
  • compositions as disclosed herein including a MetAP-2 inhibitor block copolymer conjugate in conjunction with other agents is encompassed in the present invention for treatment of diseases, disorders or conditions associated with, characterized by, or otherwise requiring MetAP-2 activity for their pathologies.
  • diseases, disorders or conditions associated with, characterized by, or otherwise requiring MetAP-2 activity for their pathologies include, but are not limited to diseases involving abnormally stimulated neovascularization, such as inflammatory diseases (rheumatism and psoriasis, among others), diabetic retinopathy and cancer.
  • a copolymer is a polymer comprising subunits of more than one type, i.e a copolymer can comprise subunits of A and subunits of B etc.
  • a copolymer is an alternating copolymer, for example, having a repeating structure comprising the different types of subunits, for example an alternating copolymer can have the formula: -A-B-A-B-A-B-A-B-A-B-, or -(-A-B-) n -.
  • MetAP-2 inhibitors as disclosed herein are associated with a block copolymer to form a MetAP-2 inhibitor-block copolymer conjugate.
  • a block copolymer comprises subunits of more than one type, but instead of an alternating copolymer, a block copolymer comprises at a minimum, a block of subunits of one subunit type followed by a block of subunits of another subunit type.
  • a block copolymer comprising two subunits, for example A and B can have the formula of: -(A-A-) n -(B-B-) n - or -A-A-A-A-A-A-B-B-B-B-B-.
  • the number of the different subunits can be different, for example; A-A-A-A-B-B-B-B.
  • a diblock copolymer is a block copolymer comprising two different blocks of polymer subunits
  • a triblock copolymer is a block copolymer comprising three different blocks of polymer subunits
  • a tretrablock copolymer is a block copolymer comprising four different blocks of copolymer subunits, etc.
  • a block copolymer is a polymer comprising at least a first block comprising a polymer of hydrophobic monomers, and at least a second block comprising a polymer comprised of hydrophilic monomers (see FIG. 14 ).
  • a “block” copolymer differs from a non-block copolymer in that a block copolymer comprises blocks of polymer of one type (e.g. a block of hydrophilic monomers) that are joined to a block of polymer of another type (e.g. a block of hydrophobic monomers), as opposed to a non-block copolymer in which the different monomers are not joined together in blocks.
  • the block copolymer would have, e.g., the structure AAAAABBBBB.
  • a non-block copolymer of the same monomer subunits would have, for example, the structure ABAABABBABAABB or the structure ABABABABABAB, for example.
  • the blocks of a block copolymer as the term is used herein will have at least 5 monomers per block (i.e., for a block copolymer of A and B monomers, the A block will be at least 5 A monomers long, and the B block will be at least 5 B monomers long). In some embodiments the homopolymer blocks will be at least 10 monomers long, 15 monomers long, 20 monomers long or more.
  • a block copolymer as the term is used herein can have different block lengths (but each block will be at least 5 monomers long)—differences in block lengths can influence the ability of the block copolymer to form certain structures, e.g., micelles.
  • the monomers making up the hydrophilic block of a block copolymer can comprise different hydrophilic monomer subunits, and similarly, in some embodiments, monomers making up the hydrophobic block of a block copolymer can comprise different hydrophobic monomer subunits.
  • Diblock copolymers can be made using living polymerization techniques, such as atom transfer free radical polymerization (ATRP), reversible addition fragmentation chain transfer (RAFT), ring-opening metathesis polymerization (ROMP), and living cationic or living anionic polymerizations.
  • ATRP atom transfer free radical polymerization
  • RAFT reversible addition fragmentation chain transfer
  • REP ring-opening metathesis polymerization
  • living cationic or living anionic polymerizations such as atom transfer free radical polymerization (ATRP), reversible addition fragmentation chain transfer (RAFT), ring-opening metathesis polymerization (ROMP), and living cationic or living anionic polymerizations.
  • Block copolymers can “microphase separate” to form periodic nanostructures, for example, when one block is hydrophobic and the other hydrophilic. Microphase separation is a situation similar to that of oil and water. Oil and water don't mix together—they macrophase separate. If you have an “oil-like” first block and a “water-like” second block, the block copolymers undergo microphase separation. The blocks want to get as far from each other as possible, but they are covalently bonded, so they're not going to get very far.
  • the “oil” and “water” blocks form nanometer-sized structures, including micelles, which comprise essentially spherical arrangement with the hydrophilic blocks arrange to the outside of the sphere, in contact with an aqueous solution and the hydrophobic blocks forming an inner hydrophobic core.
  • Thermodynamic terms can describe how the different blocks interact.
  • the interaction parameter, “chi” gives an indication of how different, chemically, the two blocks are and whether or not they will microphase separate. Generally, if the product of chi and the molecular weight is large (greater than 10.5), the blocks will microphase separate. Conversely, if the product of chi and the molecular weight is too small (less than 10.5), the different blocks are able to mix, rather than microphase separate.
  • amphiphilic block copolymers are useful in the formulations and compositions of the present invention, for example as effective drug carriers that solubilize hydrophobic drugs into an aqueous environment.
  • amphiphilic block copolymers exhibiting self-association properties are disclosed in EP No. 0397 307 and EP0583955 and EP0552802, which are incorporated herein by reference.
  • useful biodegradable polyesters comprised by the hydrophobic block of copolymers described herein are, for example, biodegradable polyester oligomers or polymers synthesized from monomers selected from, e.g., D, L-lactide, D-lactide, L-lactide, D, L-lactic acid, D-lactic acid, L-lactic acid, glycolide, glycolic acid, ⁇ -caprolactone, ⁇ -hydroxy hexanoic acid, ⁇ -butyrolactone, ⁇ -hydroxy butyric acid, ⁇ -valerolactone, ⁇ -hydroxy valeric acid, hydroxybutyric acids, malic acid, and copolymers thereof.
  • monomers selected from, e.g., D, L-lactide, D-lactide, L-lactide, D, L-lactic acid, D-lactic acid, L-lactic acid, glycolide, glycolic acid, ⁇ -caprolactone, ⁇ -hydroxy hexanoic acid, ⁇
  • the biodegradable polyester is synthesized from monomers selected from the group consisting of D, L-lactide, D-lactide, L-lactide, D, L-lactic acid, D-lactic acid, L-lactic acid, glycolide, glycolic acid, ⁇ -caprolactone, ⁇ -hydroxy hexanoic acid, and copolymers thereof.
  • the biodegradable polyester is synthesized from monomers selected from the group consisting of D, L-lactide, D-lactide, L-lactide, D, L-lactic acid, D-lactic acid, L-lactic acid, glycolide, glycolic acid, and copolymers thereof.
  • the block copolymers comprise hydrophobic polyesters containing polyester bonds, for example but not limited to polylactic acid (PLA), polyglycolic acid (PGA), poly(D,L-lactic-co-glycolic acid)(PLGA), poly(caprolactone), poly(valerolactone), poly(hydroxybutyrate) and poly(hydroxyvalerate).
  • PLA polylactic acid
  • PGA polyglycolic acid
  • PLGA poly(D,L-lactic-co-glycolic acid)
  • PLGA poly(caprolactone), poly(valerolactone), poly(hydroxybutyrate) and poly(hydroxyvalerate).
  • the block copolymers comprise hydrophobic polymers, for example of the block copolymer is selected from the group consisting of poly(d,L-lactic acid), poly(L-lysine), poly(aspartic acid), poly(caprolactone) (PCL), poly(propylene oxide).
  • a hydrophobic polymer moiety is 1-15 kDa.
  • a hydrophobic polymer moiety is between 0.5-10 kDa, 1-8 kDa, 1-5 kDa, 1-3 kDa, 3-15 kDa, 5-15 kDa, 8-15 kDa, 10-15 kDa, 12-15 kDa, 2-12 kDa, 4-10 kDa, 6-8 kDa in size.
  • a hydrophobic polymer is approximately 2 kDa, for example 1.5 kDa, 2 kDa, or 2.5 kDa.
  • a hydrophobic moiety useful in the composition as disclosed herein is a poly(d,L-lactic acid) (PLA) polymer, for example a poly(d,L-lactic acid) (PLA) polymer of 1kDa (1000 Da).
  • the copolymers comprise hydrophilic polymers, for example but not limited to polyethene glycol (PEG) polymer (which is also referred to a poly(ethylene oxide) (PEO) or poly(oxyethelene) in the art).
  • the hydrophillic polymer may be capped at one end.
  • the capping group is alkoxy.
  • a hydrophilic polymer useful in the composition as disclosed by be capped with a methoxy group.
  • a hydrophilic polymer is between 1-15 kDa.
  • a hydrophilic polymer moiety useful in the composition as disclosed is between 1-10 kDa, 1-8 kDa, 1-5 kDa, 1-3 kDa, 3-15 kDa, 5-15 kDa, 8-15 kDa, 10-15 kDa, 12-15 kDa, 2-12 kDa, 4-10 kDa, 6-8 kDa in size.
  • a hydrophilic polymer is approximately 2 kDa, for example 1.5 kDa, 2 kDa, or 2.5 kDa.
  • a hydrophilic polymer is a poly(ethylene glycol) (PEG) polymer, for example a poly(ethylene glycol) (PEG) polymer of 2 kDa (2000 Da).
  • the copolymers comprise a diblock copolymer comprising a PEG-PLA diblock copolymer, where block copolymer comprises blocks of the hydrophilic PEG monomers and blocks of the hydrophobic monomer PLA.
  • hydrophilic blocks of a copolymer can be coupled to the hydrophobic blocks by covalent bonds, for example by ester or urethane links and the like.
  • Condensation polymerization and ring opening polymerization procedures may be utilized as may the coupling of a monofunctional hydrophilic block to either end of a difunctional hydrophobic block in the presence of coupling agents such as isocyanates.
  • coupling reactions may follow activation of functional groups with activating agents, such as carbonyl diimidazole, succinic anhydride, N-hydroxy succinimide and p-nitrophenyl chloroformate and the like.
  • hydrophilic blocks of a copolymer can comprise PEG or derivatized PEG monomers of an appropriate molecular weight.
  • PEG has particularly favorable biocompatibility, nontoxic properties, hydrophilicity, solubilization properties, and rapid clearance from a patient's body.
  • the hydrophobic blocks of a copolymer should also comprise biodegradable polyester monomers that are biodegradable and biocompatible.
  • biodegradable polyester monomers that are biodegradable and biocompatible.
  • the in vitro and in vivo degradation of hydrophobic, biodegradable polyester blocks of a copolymer are well understood and the degradation products are readily metabolized and/or eliminated from the patient's body.
  • MetAP-2 inhibitors including fumagillol derivatives, such as TNP-470
  • block copolymers as disclosed herein.
  • One advantage of using the block copolymers as disclosed herein is that the MetAP-2 inhibitors such as TNP-470 that have limited solubility or dispersibility in an aqueous or hydrophilic environment have enhanced solubility and/or dispersibility and can be administered via oral administration.
  • Another advantage of using the block copolymer is improved biodistribution versus the bengamide class of MetAP-2 inhibitors, or the sulphonamide class, or the bestatin class of inhibitors.
  • one can prepare block copolymer micelles for sustained release of a MetAP-2 inhibitor using the method is disclosed in U.S. Pat. No. 6,623,729, which is incorporated in its entirety herein by reference and is further illustrated in the following steps:
  • Step 1 Preparation of Block Copolymer.
  • a block copolymer containing a hydrophobic part having hydroxyl group at one end and a hydrophilic part at the other end is prepared by copolymerization of a biodegradable polyester polymer and a polyethylene glycol (PEG) polymer in the presence of stannous octate as a catalyst: The copolymerization is performed at 160-200.quadrature for 2-6 hours under a vacuum condition.
  • PEG polyethylene glycol
  • the polyester polymer includes polylactic acid (PLA), polyglycolic acid (PGA), poly(D,L-lactic-co-glycolic acid)(PLGA), poly(caprolactone), poly(valerolactone), poly(hydroxy butyrate) or poly(hydroxy valerate), and in some embodiments PLA and methoxypolyethyleneglycol is used as the polyethyleneglycol polymer.
  • Step 2 Binding of Linker by an Activation of Functional Groups of Block Copolymer.
  • the block copolymer is dissolved in an organic solvent and reacted with a linker at room temperature in the presence of pyridine and nitrogen:
  • the organic solvent includes, but without limitation, methylenechloride, and the linker includes p-nitrophenyl chloroformate, carbonyldiimidazole (CDI), N,N′-disuccinimidyl carbonate (DSC), or a mixture of these compounds, preferably p-nitrophenyl chloroformate.
  • the reaction is carried out for 2 to 6 hours, with a molar ratio of block copolymer:linker:pyridine ranging from 1:2:2 to 1:2:6.
  • Step 3 Preparation of a Conjugate of Drug and Biodegradable Polymer.
  • the linker-bound block copolymer is conjugated to a drug by covalent linkage to obtain a micelle monomer of a conjugate of drug and block copolymer, where the block copolymer obtained by reacting with hydrazine may be used:
  • the block copolymer reacted with hydrazine forms a micelle monomer by binding the linker to a ketone group of a MetAP-2 inhibitor, such as a fumagillol derivative, while the block copolymer without hydrazine reaction forms a micelle monomer by binding the linker to an amine group of a MetAP-2 inhibitor, such as a fumagillol derivative.
  • the MetAP-2 inhibitor is TNP-470.
  • MetAP-2 inhibitors are equally suitable for copolymer conjugation.
  • other drugs or chemotherapeutic agents are used in addition to a MetAP-2 inhibitor, for example anti-cancer agents such as, but without limitation, doxorubicin, adriamycin, cisplatin, taxol and 5-fluorouracil.
  • Step 4 Preparation of Sustained Release Micelle.
  • the micelle monomers prepared in Step 3 are dispersed in an aqueous solution to prepare sustained release micelles.
  • micelles When micelle monomers are dispersed in a certain concentration, micelles are formed spontaneously by thermodynamic equilibrium. Sustained release micelles thus prepared release a drug by way of hydrolysis and enzymatic action in vivo, and the released drug exerts the same effect as free drug does.
  • a MetAP-2 inhibitor for example TNP-470, is associated with a block copolymer.
  • association means that one entity is in physical association or contact with another.
  • a MetAP-2 inhibitor “associated with” a block copolymer can be either covalently or non-covalently joined to the block copolymer. It is preferred that the association be covalent.
  • the association can be mediated by a linker moiety, particularly where the association is covalent.
  • association or “interaction” or “associated with” are used interchangeably herein and as used in reference to the association or interaction of a MetAP-2 inhibitor, e.g., TNP-470 with a block copolymer, refers to any association between the MetAP-2 inhibitor and the block copolymer, for example a diblock copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety, either by a direct linkage or an indirect linkage.
  • An indirect linkage includes an association between a MetAP-2 inhibitor and the block copolymer, wherein the MetAP-2 inhibitor and block copolymer are attached via a linker moiety, e.g., they are not directly linked.
  • Linker moieties include, but are not limited to, chemical linker moieties.
  • a linker between the MetAP-2 inhibitor and the copolymer is formed by reacting the polymer and a linker selected e.g., from the group consisting of p-nitrophenyl chloroformate, carbonyldiimidazole (CDI), N,N′-disuccinimidyl carbonate (DSC), cis-aconitic anhydride, and a mixture of these compounds.
  • a direct linkage includes any linkage wherein a linker moiety is not required.
  • a direct linkage includes a chemical or a physical interaction wherein the two moieties, i.e. the a MetAP-2 inhibitor and the block copolymer interact such that they are attracted to each other.
  • Examples of direct interactions include covalent interactions, non-covalent interactions, hydrophobic/hydrophilic, ionic (e.g., electrostatic, coulombic attraction, ion-dipole, charge-transfer), Van der Waals, or hydrogen bonding, and chemical bonding, including the formation of a covalent bond.
  • a a MetAP-2 inhibitor such as TNP-470 and the block copolymer are not linked via a linker, e.g., they are directly linked.
  • a MetAP-2 inhibitor and the block copolymer are electrostatically associated with each other.
  • the linker is a peptide linker.
  • the peptide linker is enzymatically cleavable, e.g., by a protease enzyme that cleaves the linkage to release the MetAP-2 inhibitor.
  • the peptide linkage is capable of being cleaved by preselected cellular enzymes, for instance, those found in lysosomes of cancerous cells or proliferating endothelial cells.
  • an acid hydrolysable linker could comprise an ester or amide linkage and be for instance, a cis-aconityl linkage.
  • a pH sensitive linker can also be used.
  • the linker preferably comprises at least one cleavable peptide bond.
  • the linker is an enzyme cleavable oligopeptide group preferably comprising sufficient amino acid units to allow specific binding and cleavage by a selected cellular enzyme.
  • the linker is at least two amino acids long, more preferably at least three amino acids long.
  • Cleavable linkers are described, e.g., in U.S. Pat. No. 7,332,523, which is incorporated herein by reference.
  • An example of a preferred peptide linker is Gly-Phe-Leu-Gly (see U.S. Pat. No. 7,332,523).
  • Other linker are known to those of skill in the art.
  • the MetAP-2 inhibitor in the formulations and compositions as disclosed herein is particularly useful in methods of treating conditions in which MetAP-2 activity is involved in or required for the pathology of the condition, including, but not necessarily limited to conditions in which MetAp-2 is either over-expressed or over-active in a mammal, for example a human.
  • Such conditions herein also referred to as a “MetAP-2-dependent disease or disorder” are selected from a group including, but not necessarily limited to cancer, ascites formation, psoriasis, age-related macular degeneration, thyroid hyperplasia, preeclampsia, rheumatoid arthritis and osteoarthritis, Alzheimer's disease, obesity, pleural effusion, atherosclerosis, endometriosis, diabetic/other retinopathies, neovascular glaucoma, age-related macular degeneration, hemangiomas, and corneal neovascularization, HIV, HPV, herpes and other viral infections, anthrax, IPF, COPD, multiple sclerosis, other sclerotic diseases, transplant rejection, lupus, asthma and other conditions described herein as associated with MetAP-2 activity.
  • a “MetAP-2-dependent disease or disorder” are selected from a group including, but not necessarily limited to cancer, ascites formation, psoria
  • a MetAP-2-dependent disease or disorder is cancer, where the cells are rapidly dividing neoplastic cancer cells, and where the neoplastic cells require an efficient blood supply to maintain continued growth of the tumor.
  • cancer refers to any of various malignant neoplasms characterized by the proliferation of anaplastic cells that tend to invade surrounding tissue and metastasize to new body sites and also refers to the pathological condition characterized by such malignant neoplastic growths.
  • the blood vessels provide conduits to metastasize and spread elsewhere in the body. Upon arrival at the metastatic site, the cancer cells then work on establishing a new blood supply network.
  • a MetAP-2 inhibitor in the formulations and compositions as disclosed herein is useful to inhibit proliferation at the primary disease site, and in the case of cancer, at secondary tumor sites; embodiments of the invention serve to prevent and limit the progression of the disease. Any disease that requires a continuous proliferation of cells, either primary cells, endothelial cells, adjacent cells or others in order to perpetuate the disease is a candidate target.
  • candidates for the treatment of cancer as described herein include, but are not limited to carcinomas and sarcomas found in the anus, bladder, bile duct, bone, brain, breast, cervix, colon/rectum, endometrium, esophagus, eye, gallbladder, head and neck, liver, kidney, larynx, lung, mediastinum (chest), mouth, ovaries, pancreas, penis, prostate, skin, small intestine, stomach, spinal marrow, tailbone, testicles, thyroid and uterus.
  • carcinomas include papilloma/carcinoma, choriocarcinoma, endodermal sinus tumor, teratoma, adenoma/adenocarcinoma, melanoma, fibroma, lipoma, leiomyoma, rhabdomyoma, mesothelioma, angioma, osteoma, chondroma, glioma, lymphoma/leukemia, squamous cell carcinoma, small cell carcinoma, large cell undifferentiated carcinomas, basal cell carcinoma and sinonasal undifferentiated carcinoma.
  • sarcomas include soft tissue sarcoma such as alveolar soft part sarcoma, angiosarcoma, dermatofibrosarcoma, desmoid tumor, desmoplastic small round cell tumor, extraskeletal chondrosarcoma, extraskeletal osteosarcoma, fibrosarcoma, hemangiopericytoma, hemangiosarcoma, Kaposi's sarcoma, leiomyosarcoma, liposarcoma, lymphangiosarcoma, lymphosarcoma, malignant fibrous histiocytoma, neurofibrosarcoma, rhabdomyosarcoma, synovial sarcoma, and Askin's tumor, Ewing's sarcoma (primitive neuroectodermal tumor), malignant hemangioendothelioma, malignant schwannoma, osteosarcoma, and chondrosarcoma.
  • a MetAP-2-dependent disease or disorder is age-related macular degeneration.
  • VEGF contributes to abnormal blood vessel growth from the choroid layer of the eye into the retina, similar to what occurs during the wet or neovascular form of age-related macular degeneration.
  • Macular degeneration often called AMD or ARMD (age-related macular degeneration)
  • AMD age-related macular degeneration
  • endothelial cells must proliferate. This proliferation is uneven, and results in the leakage described above, which causes permanent damage to light-sensitive retinal cells, which die off and create blind spots in central vision or the macula.
  • a MetAP-2-dependent disease or disorder is diabetic retinopathy-abnormal blood vessel growth associated with diabetic eye diseases.
  • diabetic retinopathy and retinopathy of prematurity (ROP) VEGF is released which promotes blood vessel formation—thus, the disease or disorder is an angiogenic disease or disorder.
  • ROP diabetic retinopathy of prematurity
  • VEGF releases which promotes blood vessel formation—thus, the disease or disorder is an angiogenic disease or disorder.
  • the retina light-sensitive tissue in back of the eye
  • VEGF turns on its receptor, igniting a chain reaction that culminates in new blood vessel growth.
  • the backup blood vessels having proliferated too quickly and unevenly, are faulty, so they leak, bleed and encourage scar tissue that detaches the retina, resulting in severe loss of vision.
  • Such growth is the hallmark of diabetic retinopathy, the leading cause of blindness among young people in developed countries.
  • the subject in need of treatment can be a ma
  • a MetAP-2-dependent disease or disorder is rheumatoid arthritis.
  • RA Rheumatoid arthritis
  • the disease is thought to occur as an immunological response to an as yet unidentified antigen.
  • the expansion of the synovial lining of joints in rheumatoid arthritis (RA) and the subsequent invasion by the pannus of underlying cartilage and bone necessitate an increase in the vascular supply to the synovium, to cope with the increased requirement for oxygen and nutrients.
  • Endothelial cell proliferation is now recognized as a key event in the formation and maintenance of the pannus in RA (Paleolog, E.
  • RA is an angiogenesis-related disease or disorder.
  • some of the earliest histological observations are blood vessels.
  • a mononuclear infiltrate characterizes the synovial tissue along with a luxuriant vasculature.
  • Endothelial cell proliferation is integral to formation of the inflammatory pannus and without it, leukocyte ingress could not occur (Koch, A. E., 2000).
  • Disruption of the formation of new blood vessels would not only prevent delivery of nutrients to the inflammatory site, it could also reduce joint swelling due to the additional activity of VEGF, a potent proangiogenic factor in RA, as a vascular permeability factor.
  • a MetAP-2-dependent disease or disorder is Alzheimer's disease.
  • AD Alzheimer's disease
  • statins histamine H2-receptor blockers
  • calcium-channel blockers all of which are cardiovascular drugs with an anti-proliferative effects, seem to prevent Alzheimer's disease and/or influence the outcome of AD patients.
  • AD endothelial cell proliferation in the brain vasculature may play an important role in AD, that is, AD can be an angiogenesis-related or -mediated disease.
  • AD can be an angiogenesis-related or -mediated disease.
  • the brain endothelium secretes the precursor substrate for the beta-amyloid plaque and a neurotoxic peptide that selectively kills cortical neurons.
  • amyloid deposition in the vasculature leads to endothelial cell apoptosis and endothelial cell activation which leads to neovascularization.
  • Vessel formation could be blocked by the VEGF antagonist SU 4312 as well as by statins, indicating that anti-proliferative or anti-angiogenic strategies can interfere with endothelial cell activation in AD (Schultheiss C., el. al., 2006; Grammas P., et. al., 1999) and can be used for preventing and/or treating AD.
  • a MetAP-2-dependent disease or disorder is obesity. It has been shown that the MetAP-2 inhibitor TNP-470 was able to prevent diet-induced and genetic obesity in mice (Ebba Br ⁇ kenhielm et. al., Circulation Research, 2004; 94:1579). TNP-470 reduced vascularity in the adipose tissue, thereby inhibiting the rate of growth of the adipose tissue and obesity development. Obesity is thus an angiogenesis-related or -mediated disease or disorder.
  • a MetAP-2-dependent disease or disorder is endometriosis.
  • Excessive endometrial angiogenesis is proposed as an important mechanism in the pathogenesis of endometriosis (Healy, D L., et. al., 1998).
  • the endometrium of patients with endometriosis shows enhanced endothelial cell proliferation.
  • Strategies that inhibit endothelial cell proliferation can be used to treat endometriosis.
  • the method of treating the MetAP-2-dependent disease or disorder is applicable to the treatment of cancer; the method as disclosed herein is applicable to all carcinomas, blood-borne cancers and sarcomas.
  • the cancer is selected from the group consisting of papilloma/carcinoma, choriocarcinoma, endodermal sinus tumor, teratoma, adenoma/adenocarcinoma, melanoma, fibroma, lipoma, leiomyoma, rhabdomyoma, mesothelioma, angioma, osteoma, chondroma, glioma, lymphoma/leukemia, squamous cell carcinoma, small cell carcinoma, large cell undifferentiated carcinomas, basal cell carcinoma, sinonasal undifferentiated carcinoma, soft tissue sarcoma such as alveolar soft part sarcoma, angiosarcoma, dermatofibrosarcoma,
  • the MetAP-2 inhibitor in the formulations and compositions as disclosed herein is particularly useful in methods of inhibiting cell proliferation, including angiogenesis at a site of tumorigenesis in a mammal. 45
  • the MetAP-2 inhibitor in the formulations and compositions as disclosed herein administered at such sites and in such varied ways prevents or inhibits endothelial cell proliferation and blood vessel formation at the site thereby inhibiting the development and growth of the tumor.
  • Tumors which may be prevented or inhibited by preventing or inhibiting proliferation with the conjugate include but are not limited to melanoma, adenocarcinoma, sarcomas, thymoma, lymphoma, lung tumors, liver tumors, colon tumors, kidney tumors, non-Hodgkins lymphoma, Hodgkins lymphoma, leukemias, uterine tumors, breast tumors, prostate tumors, renal tumors, ovarian tumors, pancreatic tumors, brain tumors, testicular tumors, bone tumors, muscle tumors, tumors of the placenta, gastric tumors, metastases and the like.
  • compositions as disclosed herein comprising a MetAP-2 inhibitor can inhibit proliferation of blood vessel endothelial cells, thus having anti-angiogenesis effect or inhibition of angiogenesis.
  • the compositions as disclosed herein comprising MetAP-2 inhibitors can be used in methods for treating angiogenesis-mediated conditions in which MetAP-2 is involved in the pathology, such as inhibiting growth and metastasis of cancer as well as treating other various diseases where inappropriate angiogenesis occurs or proliferation of blood vessel endothelial cells occur, for example but not limited to, inflammatory diseases, diabetic retinopathy, rheumatoid arthritis, psoriasis.
  • compositions comprising MetAP-2 inhibitors as disclosed herein can be used as a cancer metastasis inhibitor or therapeutic agent against cancer, inflammatory diseases, diabetic retinopathy, rheumatoid arthritis, psoriasis and other retinopathies such as retinopathy of prematurity.
  • Actual dosage levels of active ingredients in the pharmaceutical compositions as described herein can be varied so as to obtain an amount of the active compound(s) which is effective to achieve the desired therapeutic response for a particular subject or patient.
  • the selected dosage level will depend upon the activity of the particular MetAP-2 inhibitor, the type of administration composition (i.e. tablet versus liquid oral administration versus ocular versus topical versus inhaled, for example), the severity of the condition being treated and the condition and prior medical history of the patient being treated.
  • a therapeutically effective amount of a compound e.g., a MetAP-2 inhibitor as described herein means a sufficient amount of the compound to treat disorders, at a reasonable benefit/risk ratio applicable to any medical treatment. It will be understood, however, that the total daily usage of the compositions and formulations as disclosed herein will be decided by the attending physician within the scope of sound medical judgment.
  • a “therapeutically effective amount” as the term is used herein need not eradicate a disease. Rather, a therapeutically effective amount will at least slow progression of a disease (as non-limiting example, the growth of a tumor or neoplasm) relative to progression without the therapeutic agent, for example the composition as disclosed herein comprising a MetAP-2-block copolymer conjugate. Thus, it is preferred, but not required that the therapeutic agent actually eliminate the disease.
  • the specific therapeutically effective dose level for any particular subject will depend upon a variety of factors including the disorder being treated and the severity of the disorder; activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the compositions and formulations as disclosed herein which are employed; and like factors well known in the medical arts.
  • compositions as disclosed herein can also be administered in prophylatically or therapeutically effective amounts.
  • the formulations and compositions as disclosed herein can be administered along with a pharmaceutically acceptable carrier.
  • a prophylatically or therapeutically effective amount means that amount necessary, at least partly, to attain the desired effect, or to delay the onset of, inhibit the progression of, or halt altogether, the onset or progression of the particular disease or disorder being treated. Such amounts will depend, of course, on the particular condition being treated, the severity of the condition and individual patient parameters including age, physical condition, size, weight and concurrent treatment. These factors are well known to those of ordinary skill in the art and can be addressed with no more than routine experimentation. It is preferred generally that a maximum dose be used, that is, the highest safe dose according to sound medical judgment. It will be understood by those of ordinary skill in the art, however, that a lower dose or tolerable dose can be administered for medical reasons, psychological reasons or for virtually any other reasons.
  • the term “effective amount” as used herein refers to the amount of therapeutic agent of pharmaceutical composition to alleviate at least some of the symptoms of the disease or disorder.
  • the term “effective amount” includes within its meaning a sufficient amount of pharmacological composition to provide the desired effect. The exact amount required will vary depending on factors such as the type of tumor to be treated, the severity of the tumor, the drug resistance level of the tumor, the species being treated, the age and general condition of the subject, the mode of administration and so forth. Thus, it is not possible to specify the exact “effective amount”. However, for any given case, an appropriate “effective amount” can be determined by one of ordinary skill in the art using only routine experimentation.
  • efficacy of treatment can be judged by an ordinarily skilled practitioner. As disclosed in the Examples, efficacy can be assessed in animal models of cancer and tumor, for example treatment of a rodent with a cancer, and any treatment or administration of the compositions or formulations that leads to a decrease of at least one symptom of the cancer, for example a reduction in the size of the tumor or a slowing or cessation of the rate of growth of the tumor indicates effective treatment.
  • Efficacy for any given formulation can also be judged using an experimental animal model of cancer, e.g., wild-type mice or rats, or preferably, transplantation of tumor cells akin to that described in the Examples herein below.
  • an experimental animal model of cancer e.g., wild-type mice or rats, or preferably, transplantation of tumor cells akin to that described in the Examples herein below.
  • efficacy of treatment is evidenced when a reduction in a symptom of the cancer, for example a reduction in the size of the tumor or a slowing or cessation of the rate of growth of the tumor occurs earlier in treated, versus untreated animals.
  • “earlier” is meant that a decrease, for example in the size of the tumor occurs at least 5% earlier, but preferably more, e.g., one day earlier, two days earlier, 3 days earlier, or more.
  • the amount of each component to be administered also depends upon the frequency of administration, such as whether administration is once a day, twice a day, 3 times a day or 4 times a day, once a week; or several times a week, for example 2 or 3, or 4 times a week.
  • TNP-470 administration will be described in greater detail as a representative example of the administration procedures for all MetAP-2 inhibitors in general.
  • 6-O—(N-chloroacetylcarbamoyl)fumagillol (TNP-470) the following information may serve as a general guideline for administration.
  • the formulations and compositions as disclosed herein are administered from once a day to several times a day, for example 2 times a day, three times a day, or four times a day.
  • the formulations and compositions as disclosed herein can be administered, for example three to five times a week, if it is to be plurally administered in a given week.
  • a suitable dose of the MetAP-2 inhibitor in the formulations and compositions as disclosed herein for a subject in need of treatment can be used according to conventionally used dose ranges of about 1 mg to about 2000 mg TNP-470 equivalent per kilogram of body weight.
  • conventional doses of fumagillol derivatives are about 0.1 mg/kg to 40 mg/kg body weight, preferably about 0.5 mg/kg to 20 mg/kg body weight as disclosed in U.S. Pat. No. 5,290,807.
  • a dose below the threshold used for chemotherapy can be used.
  • a suitable dose could be less than the conventionally used chemotherapeutic dose, for example, dose ranges of about 1 ⁇ g to 1 mg or 0.1 m to 1 mg, or 1 mg to 10 mg TNP-470 per kilogram of body weight can be used.
  • TNP-470 if TNP-470 is administered once a week, it can be administered in an amount of from about 20 to about 200 mg/m 2 /week; preferably in an amount of from about 40 to about 180 mg/m 2 /week; and most preferably in an amount of from about 135 to about 175 mg/m 2 /week. In some embodiments, if TNP-470 is administered daily, it may be administered in an amount of from about 1 to about 10 mg/m 2 /day; for example in an amount of from about 1.25 to about 5 mg/m 2 /day; or in an amount of from about 1 to about 3 mg/m 2 /day. For continuous administration, the component is usually administered for at least five consecutive days of the week.
  • the effective amount of a composition as disclosed herein comprising a fumagillol derivative can be determined using an anti-angiogenesis assay as disclosed herein, and in some embodiments, the effective amount is less than the amount used as the conventionally effective dose. Similar dosage regimes can be applied for formulations of other MetAP-2 inhibitor compositions as disclosed herein.
  • higher dosages can be used, provided there is not unacceptable toxicity.
  • dosages in the range of about 50 to about 500 mg/m 2 /week or more, and sub-ranges within this range are specifically contemplated.
  • dosages in the range of about 120 to 350 mg/m 2 /week, 200 to 400 mg/m 2 /week, etc. are specifically contemplated.
  • compositions and formulations as disclosed herein are oral administration.
  • Solid dosage forms for oral administration include, for example but not limited to capsules, tablets, pills, powders and granules.
  • the compositions as disclosed herein may be mixed with at least one inert, pharmaceutically acceptable excipient or carrier, such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol and silicic acid; b) binders such as carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; c) humectants such as glycerol; d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates and sodium carbonate; e) solution retarding agents such as paraffin; f) absorption accelerators such as quaternary
  • compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • the active components can also be in micro-encapsulated form, if appropriate, with one or more of the above-mentioned excipients.
  • the compound selected can be mixed with solid, powdered ingredients, such as lactose, saccharose, sorbitol, mannitol, starch, arnylopectin, cellulose derivatives, gelatin, or another suitable ingredient, as well as with disintegrating agents and lubricating agents such as magnesium stearate, calcium stearate, sodium stearyl fumarate and polyethylene glycol waxes.
  • solid, powdered ingredients such as lactose, saccharose, sorbitol, mannitol, starch, arnylopectin, cellulose derivatives, gelatin, or another suitable ingredient, as well as with disintegrating agents and lubricating agents such as magnesium stearate, calcium stearate, sodium stearyl fumarate and polyethylene glycol waxes.
  • disintegrating agents and lubricating agents such as magnesium stearate, calcium stearate, sodium stearyl fumarate and polyethylene glycol waxes.
  • compositions for topical e.g., oral mucosa, respiratory mucosa
  • oral administration can form solutions, suspensions, tablets, pills, capsules, sustained-release formulations, oral rinses, or powders, as known in the art are described herein.
  • the compositions also can include stabilizers and preservatives. For examples of carriers, stabilizers and adjuvants, University of the Sciences in Philadelphia (2005) Remington: The Science and Practice of Pharmacy with Facts and Comparisons , 21st Ed.
  • the compositions can also be inhaled (pulmonary, nasal), ocular (eyedrop), sub-lingual, suppository, or topical (e.g., an ointment).
  • MetAP-2 inhibitors such as TNP-470 have been tested in conjunction with treatment with various other drugs to enhance efficacy for treatment of diseases induced by abnormally stimulated neovascularization, such as inflammatory diseases (rheumatism and psoriasis among others), diabetic retinopathy, cancer and other diseases and conditions as discussed elsewhere herein.
  • Soft gelatin capsules can be prepared with capsules containing a mixture of the active compound or compounds of the invention in vegetable oil, fat, or other suitable vehicle for soft gelatin capsules.
  • Hard gelatin capsules can contain granules of the active compound.
  • Hard gelatin capsules can also contain the targeted delivery composition including the targeting moiety and the carrier particle as well as the therapeutic agent in combination with solid powdered ingredients such as lactose, saccharose, sorbitol, mannitol, potato starch, corn starch, amylopectin, cellulose derivatives or gelatin.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents that are compatible with the maintenance of a micelle of a diblock copolymer as described herein.
  • Liquid preparations for oral administration can also be prepared in the form of syrups or suspensions, e.g. solutions or suspensions containing from 0.2% to 20% by weight of the active ingredient and the remainder consisting of sugar or sugar alcohols and a mixture of ethanol, water, glycerol, propylene glycol and polyethylene glycol provided that such solvent is compatible with maintaining the micelle form.
  • liquid preparations can contain coloring agents, flavoring agents, saccharin and carboxymethyl cellulose or other thickening agents.
  • Liquid preparations for oral administration can also be prepared in the form of a dry powder to be reconstituted with a suitable solvent prior to use.
  • the oral compositions may also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring and perfuming agents
  • Transdermal patches may also be used to provide controlled delivery of the formulations and compositions as disclosed herein to specific regions of the body. Such dosage forms can be made by dissolving or dispensing the component in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate-controlling membrane or by dispersing the compound in a polymer matrix or gel.
  • Such transdemal patches are useful for treating parts of the body where abnormally stimulated neovascularization occurs, such as inflammatory diseases, for example rheumatism and psoriasis among others, diabetic retinopathy and cancer, for example skin cancer or other skin related neovascular conditions (such as psoriasis) or malignancies.
  • a further form of topical administration is to the eye, for example as a treatment for retinopathy, such as diabetic retinopathy or retinopathy of prematurity (ROP) or for the treatment of immune-mediated conditions of the eye such as autoimmune diseases, allergic or inflammatory conditions, and corneal transplants.
  • Components of the invention may be delivered in a pharmaceutically acceptable ophthalmic vehicle, such that the component is maintained in contact with the ocular surface for a sufficient time period to allow the component to penetrate the corneal and internal regions of the eye, as for example the anterior chamber, posterior chamber, vitreous body, aqueous humor, vitreous humor, cornea, iris/cilary, lens, choroid/retina and sclera.
  • the pharmaceutically acceptable ophthalmic vehicle may, for example, be an ointment or an encapsulating material.
  • compositions and formulations as disclosed herein can be also administered via rectal or vaginal administration.
  • the compositions and formulations as disclosed herein can be in the form of suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at room temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active component.
  • Dosage units for rectal or vaginal administration can be prepared (i) in the form of suppositories which contain the active substance mixed with a neutral fat base; (ii) in the form of a gelatin rectal capsule which contains the active substance in a mixture with a vegetable oil, paraffin oil or other suitable vehicle for gelatin rectal capsules; (iii) in the form of a ready-made micro enema; or (iv) in the form of a dry micro enema formulation to be reconstituted in a suitable solvent just prior to administration.
  • MetAP-2 inhibitor-copolymer conjugates described herein can be used in admixture or in combination with a gastric acid secretion-inhibitor and/or an antacid.
  • Gastric acid secretion inhibitors include, for example H 2 blockers (e.g. famotidine, cimetidine, ranitidine hydrochloride, etc.) and proton pump inhibitors (e.g. lansoprazole, omeprazole, etc.).
  • H 2 blockers e.g. famotidine, cimetidine, ranitidine hydrochloride, etc.
  • proton pump inhibitors e.g. lansoprazole, omeprazole, etc.
  • compounds which elevate the intragastric pH level such as magnesium carbonate, sodium hydrogen carbonate, magnesium hydroxide, magnesium oxide and magnesium hydroxide can be employed.
  • the oral dosage forms of the compositions and formulations as disclosed herein can be administered after the intragastric pH has been increased to alleviate the influence of gastric acid by the administration of a gastric acid secretion inhibitor and/or antacid.
  • compositions and formulations as disclosed herein can be in a form of enteric-coated preparation for oral administration comprising a MetAP-2 inhibitor-block copolymer conjugate.
  • a MetAP-2 inhibitor containing core for coating with an enteric coating film can be prepared using an oleaginous base or by other known formulation methods without using an oleaginous base.
  • the compositions and formulations as disclosed herein in the form of the drug-containing core for coating with a coating agent may be, for example, tablets, pills and granules.
  • the excipient contained in the core is exemplified by saccharides, such as sucrose, lactose, mannitol and glucose, starch, crystalline cellulose and calcium phosphate.
  • Useful binders include polyvinyl alcohol, hydroxypropyl cellulose, macrogol, Pluronic F-68, gum arabic, gelatin and starch.
  • Useful disintegrants include carboxymethyl cellulose calcium (ECG505), crosslinked carboxymethylcellulose sodium (Ac-Di-Sol), polyvinylpyrrolidone and low-substituted hydroxypropyl cellulose (L-HPC).
  • Useful lubricants and antiflocculants include talc and magnesium stearate.
  • the enteric coating agent is an enteric polymer which is substantially insoluble in the acidic pH and is at least partially soluble at weaker acidic pH through the basic pH range.
  • the range of acidic pH is about 0.5 to about 4.5, preferably about 1.0 to about 2.0.
  • the range of weaker acidic pH through basic pH is about 5.0 to about 9.0, preferably about 6.0 to about 7.5.
  • cellulose acetate phthalate, hydroxypropylmethylcellulose phthalate, hydroxypropylmethyl acetate succinate (Shin-Etsu Chemicals), methacrylic copolymers (Rhon-Pharma, Eudragit® L-30D-55, L100-55, L100, S100, etc.), etc. can be mentioned as examples of the enteric coating agent. These materials are effective in terms of stability, even if they are directly used as enteric compositions.
  • micelles for example, spherical microparticles having particle diameters of about 0.01 ⁇ m to about 1000 ⁇ m are generated, or more preferably about 0.01 to about 5 ⁇ m (about 10 nm to about 5,000 nm). This formation can be performed by the methods as disclosed in the Examples or as disclosed in Japanese Patent Application JP-A-223533/1991.
  • micelles between 50 and 500 nm are useful in the compositions as disclosed herein, for example about 50-100 nm, 100-150 nm, 150-200 nm, 200-250 nm, 250-300 nm, 300-350 nm, 350-400 nm, 450-500 nm.
  • the micelles are about 130 nm as disclosed in the examples.
  • the concentration or content of the MetAP-2 inhibitor in the composition can be appropriately selected according to the physicochemical properties of the composition.
  • the concentration is about 0.0005 to about 30% (w/v) and preferably about 0.005 to about 25% (w/v).
  • the content is about 0.01 to about 90% (w/w) and preferably about 0.1 to about 50% (w/w).
  • additives such as a preservative (e.g. benzyl alcohol, ethyl alcohol, benzalkonium chloride, phenol, chlorobutanol, etc.), an antioxidant (e.g. butylhydroxyanisole, propyl gallate, ascorbyl palmitate, alpha-tocopherol, etc.), and a thickener (e.g. lecithin, hydroxypropylcellulose, aluminum stearate, etc.) can be used in the compositions and formulations as disclosed herein.
  • a preservative e.g. benzyl alcohol, ethyl alcohol, benzalkonium chloride, phenol, chlorobutanol, etc.
  • an antioxidant e.g. butylhydroxyanisole, propyl gallate, ascorbyl palmitate, alpha-tocopherol, etc.
  • a thickener e.g. lecithin, hydroxypropylcellulose, aluminum stearate, etc.
  • diblock copolymer conjugates as described generally need no further emulsifiers. Nonetheless, if necessary, one can use an additional emulsifier with the compositions and formulations as disclosed herein.
  • emulsifiers that might be used include pharmaceutically acceptable phospholipids and nonionic surfactants.
  • the emulsifiers can be used individually or in combinations of two or more.
  • the phospholipid includes naturally occurring phospholipids, e.g. egg yolk lecithin, soya lecithin, and their hydrogenation products, and synthetic phospholipids, e.g. phosphatidylcholine, phosphatidylethanolamine, etc.
  • the nonionic surfactant includes macro-molecular surfactants with molecular weights in the range of about 800 to about 20000, such as polyethylene-propylene copolymer, polyoxyethylene alkyl ethers, polyoxyethylene alkylarylethers, hydrogenated castor oil-polyoxyethylene derivatives, polyoxyethylene sorbitan derivatives, polyoxyethylene sorbitol derivatives, polyoxyethylene alkyl ether sulfate, and so on.
  • the proportion of the emulsifier is selected so that the concentration in a final administrable composition will be in the range of about 0.1 to about 10%, preferably about 0.5 to about 5%.
  • a stabilizer for further improving the stability of the compositions and formulations as disclosed herein such as an antioxidant or a chelating agent, an isotonizing agent for adjusting the osmolarity, an auxiliary emulsifier for improving the emulsifying power, and/or an emulsion stabilizer for improving the stability of the emulsifying agent can be incorporated.
  • the isotonizing agent that can be used includes, for example, gylycerin, sugar alcohols, monosaccharides, disaccharides, amino acids, dextran, albumin, etc. These isotonizing agents can be used individually or in combination, with two or more.
  • An emulsion stabilizer that can be used which includes cholesterol, cholesterol esters, tocopherol, albumin, fatty acid amide derivatives, polysaccharides, polysaccharide fatty acid ester derivatives, etc.
  • compositions and formulations as disclosed herein can further comprise a viscogenic substance which can adhere to the digestive tract mucosa due to its viscosity expressed on exposure to water.
  • the examples of the viscogenic substance include, but are not particularly limited as long as it is pharmaceutically acceptable, such as polymers (e.g. polymers or copolymers of acrylic acids and their salts) and natural-occurring viscogenic substances (e.g. mucins, agar, gelatin, pectin, carrageenin, sodium alginate, locust bean gum, xanthan gum, tragacanth gum, arabic gum, chitosan, pullulan, waxy starch, sucralfate, curdlan, cellulose, and their derivatives).
  • polymers e.g. polymers or copolymers of acrylic acids and their salts
  • natural-occurring viscogenic substances e.g. mucins, agar, gelatin, pectin, carrageenin, sodium alginate
  • the additives conventionally used for preparing the oral compositions can be added.
  • the additives include excipients (e.g. lactose, corn starch, talc, crystalline cellulose, sugar powder, magnesium stearate, mannitol, light anhydrous silicic acid, magnesium carbonate, calcium carbonate, L-cysteine, etc.), binders (e.g. starch, sucrose, gelatin, arabic gum powder, methylcellulose, carboxymethylcellulose, carboxymethylcellulose sodium, hydroxypropylcellulose, hydroxypropylmethylcellulose, polyvinylpyrrolidone, pullulan, dextrin, etc.), disintegrators (e.g.
  • anionic surfactants e.g. sodium alkylsulfates etc.
  • nonionic surfactants e.g. polyoxyethylene sorbitan fatty acid esters, polyoxyethylene fatty acid esters, polyoxyethylene-castor oil derivatives, etc.
  • maltosyl-beta-cyclodextrin maltosyl-beta-cyclodextrin, maltosyl-beta-cyclodextrin-carboxylic acid, etc.
  • colorants corrigents, adsorbents, antiseptics, moistening agents, antistatic agents, disintegration retardants, and so on.
  • the proportion of these additives can be appropriately selected from the range that can keep the stability and absorption of the basis.
  • compositions and formulations as disclosed herein for oral administration of the present invention may also include flavoring agents.
  • flavoring agents include, for example, anise oil, lavender oil, lemon oil, orange essence, rose oil, powder green tea, bergamot oil, (alpha[litre]) borneol, Natural Peal Extract AH-10, Sugar, bitter essence, pine flavor etc.
  • micelles for example, spherical microparticles having particle diameters of about 0.1 nm to about 1000 nm
  • this formation can be achieved by methods known in the art (e.g. JP-A-223533/1991).
  • micelles between 50 and 500 nm are useful in the compositions as disclosed herein, for example about 50-100 nm, 100-150 nm, 150-200 nm, 200-250 nm, 250-300 nm, 300-350 nm, 350-400 nm, 450-500 nm.
  • the micelles are about 130 nm as disclosed in the examples.
  • compositions and formulations as disclosed herein can exhibit potent pharmacological activity with low toxicity such that they are useful as a medicament for prevention and treatment for, interalia, MetAP-2-associated disease in mammals (e.g. mouse, rat, monkey, bovine, canine, human, etc.), the MetAP-2-associated disease including, for example, (1) inflammatory diseases such as rheumatoid arthritis, (2) diabetic retinopathy, and (3) benign and malignant tumors (e.g.
  • gastric cancer cancer of the esophagus, duodenal cancer, cancer of the tongue, pharyngeal cancer, brain tumors, neurilemoma, colorectal cancer, non-small-cell lung cancer, small cell carcinoma of the lung, hepatic carcinoma, renal cancer, cancer of the breast, biliary tract cancer, cancer of the pancreas, cancer of the prostate, cancer of the uterus, carcinoma of the uterine cervix, ovarian cancer, cancer of the urinary bladder, cancer of the skin, malignant melanoma, cancer of the thyroid, sarcomas of bone, hemangioma, hemangiofibroma, retinal sarcoma, cancer of the penis, solid tumors of childhood, Kaposi's sarcoma in AIDS, etc., inclusive of recurrencies and metastases to other organs), fibrotic diseases such as idiopathic pulmonary fibrosis, or cystic fibrosis.
  • compositions and formulations as disclosed herein insures an effective blood concentration within the range not causing expression of the side effects of the active substance in prolonged time, or not contributing to new side effects due to prolonged blood circulation or depoting in organ tissue for lack of renal or other clearance mechanism.
  • compositions and formulations as disclosed herein are filled into capsule shells coated with an enteric coating agent as mentioned above for use as an enteric composition.
  • an enteric coating agent as mentioned above for use as an enteric composition.
  • the capsule shell for example, soft capsules (e.g. the product of R. P. Sealer) and hard gelatin capsules are used.
  • liquid or solid compositions and formulations as disclosed herein can be administered orally.
  • the liquid form it can be directly administered e.g., by drinking an elixir or suspension of the composition, or alternatively, into the digestive tract via a catheter or sonde for oral administration or administered in the usual manner in the unit dosage form of a hard capsule or a soft capsule.
  • the solid form it can be administered orally as powders, capsules, tablets, or the like in the usual manner. It can also be redispersed in a suitable dispersion medium and administered in a liquid form.
  • the oral dose of the composition as disclosed herein is about 1 mg to about 3 g/day, preferably about 10 mg to about 1 g/day, of a MetAP-2 inhibitor. In some embodiments, the oral dose of the composition as disclosed herein is between the ranges of about 25 mg to about 1 g/day, and in some embodiments less than 25 mg to 1 g/day, for example about 10 mg to about 0.5 g/day, of a MetAP-2 inhibitor.
  • compositions and formulations as disclosed herein enhance biodistribution, stability and uptake properties of MetAP-2 inhibitors and increase the pharmacological activity thereof, so that better assurance of therapeutic efficacy is achieved without the need for parenteral administration.
  • the dosage form of the compositions and formulations as disclosed herein is stable and exhibits remarkable inhibiting activity on proliferative activity in oral administration so that it can be used as clinically advantageous oral medicine.
  • Suspensions in addition to the active components, may contain suspending agents, as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like.
  • suspending agents as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like.
  • Proper fluidity can be maintained, for example, by the use of coating materials such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.
  • the delivery is by intranasal administration of the composition, especially for use in therapy of the brain and related organs (e.g., meninges and spinal cord).
  • intraocular administration is also possible.
  • Suitable formulations can be found in Remington's Pharmaceutical Sciences, 16th and 18th Eds., Mack Publishing, Easton, Pa. (1980 and 1990), and Introduction to Pharmaceutical Dosage Forms, 4th Edition, Lea & Febiger, Philadelphia (1985), each of which is incorporated herein by reference.
  • the present invention encompasses combination therapy in which the formulations and compositions as disclosed herein are used in combination with a chemotherapeutic agent such as Taxol, cyclophosphamide, cisplatin, gancyclovir and the like.
  • a chemotherapeutic agent such as Taxol, cyclophosphamide, cisplatin, gancyclovir and the like.
  • the chemotherapeutic agent may also be included within a micelle as described herein.
  • Such a therapy is particularly useful in situations in which the subject or patient to be treated has a large preexisting tumor mass which is well vascularized.
  • the chemotherapeutic agent serves to reduce the tumor mass and the conjugate prevents or inhibits neovascularization within or surrounding the tumor mass.
  • the chemotherapeutic agent may also be administered at lower doses than normally used and at such doses may act as an anti-proliferative agent.
  • the second therapy can be administered to the subject before, during, after or a combination thereof relative to the administration of the compositions as disclosed herein.
  • Anti-proliferative therapies are well known in the art and are encompassed for use in the methods of the present invention. Therapies includes, but are not limited to an alkylating agent, mitotic inhibitor, antibiotic, or antimetabolite, anti-angliogenic agents etc. Such chemotherapy can comprise administration of CPT-11, temozolomide, or a platin compound. Radiotherapy can include, for example, x-ray irradiation, w-irradiation, ⁇ -irradiation, or microwaves.
  • chemotherapeutic agent or “chemotherapy agent” are used interchangeably herein and refers to an agent that can be used in the treatment of cancers and neoplasms, for example brain cancers and gliomas and that is capable of treating such a disorder.
  • a chemotherapeutic agent can be in the form of a prodrug which can be activated to a cytotoxic form.
  • Chemotherapeutic agents are commonly known by persons of ordinary skill in the art and are encompassed for use in the present invention.
  • chemotherapeutic drugs for the treatment of tumors and gliomas include, but are not limited to: temozolomide (Temodar), procarbazine (Matulane), and lomustine (CCNU).
  • Chemotherapy given intravenously includes vincristine (Oncovin or Vincasar PFS), cisplatin (Platinol), carmustine (BCNU, BiCNU), and carboplatin (Paraplatin), Mexotrexate (Rheumatrex or Trexall), irinotecan (CPT-11); erlotinib; oxalipatin; anthracyclins-idarubicin and daunorubicin; doxorubicin; alkylating agents such as melphalan and chlorambucil; cis-platinum, methotrexate, and alkaloids such as vindesine and vinblastine.
  • the present invention encompasses combination therapy in which the formulations and compositions as disclosed herein are used in combination with, a cytostatic agent, anti-VEGF and/or p53 reactivation agent.
  • a cytostatic agent is any agent capable of inhibiting or suppressing cellular growth and multiplication. Examples of cytostatic agents used in the treatment of cancer are paclitaxel, 5-fluorouracil, 5-fluorouridine, mitomycin-C, doxorubicin, and zotarolimus.
  • Other cancer therapeutics include inhibitors of matrix metalloproteinases such as marimastat, growth factor antagonists, signal transduction inhibitors and protein kinase C inhibitors.
  • anti-VEGF agents include bevacizumab (AvastinTM) VEGF Trap, CP-547,632, AG13736, AG28262, SU5416, SU11248, SU6668, ZD-6474, ZD4190, CEP-7055, PKC 412, AEE788, AZD-2171, sorafenib, vatalanib, pegaptanib octasodium, IM862, DC101, angiozyme, Sirna-027, caplostatin, neovastat, ranibizumab, thalidomide, and AGA-1470, a synthetic analog of fumagillin (alternate names: Amebacilin, Fugillin, Fumadil B, Fumadil) (A. G. Scientific, catalog #F1028), an angio-inhibitory compound secreted by Aspergillus fumigates
  • anti-VEGF agent refers to any compound or agent that produces a direct effect on the signaling pathways that promote growth, proliferation and survival of a cell by inhibiting the function of the VEGF protein, including inhibiting the function of VEGF receptor proteins.
  • agent or “compound” as used herein means any organic or inorganic molecule, including modified and unmodified nucleic acids such as antisense nucleic acids, RNAi agents such as siRNA or shRNA, peptides, peptidomimetics, receptors, ligands, and antibodies.
  • Preferred VEGF inhibitors include for example, AVASTIN® (bevacizumab), an anti-VEGF monoclonal antibody of Genentech, Inc.
  • VEGF Trap (Regeneron/Aventis). Additional VEGF inhibitors include CP-547,632 (3-(4-Bromo-2,6-difluoro-benzyloxy)-5-[3-(4-pyrrolidin 1-yl-butyl)-ureido]-isothiazole-4-carboxylic acid amide hydrochloride; Pfizer Inc., NY), AG13736, AG28262 (Pfizer Inc.), SU5416, SU11248, & SU6668 (formerly Sugen Inc., now Pfizer, New York, N.Y.), ZD-6474 (AstraZeneca), ZD4190 which inhibits VEGF-R2 and -R1 (AstraZeneca), CEP-7055 (Cephalon Inc., Frazer, Pa.), PKC 412 (Novartis), AEE788 (Novartis), AZD-2171), NEXAVAR®
  • VEGFR2-selective monoclonal antibody DC101 ImClone Systems, Inc.
  • angiozyme a synthetic ribozyme from Ribozyme (Boulder, Colo.) and Chiron (Emeryville, Calif.)
  • Sirna-027 an siRNA-based VEGFR1 inhibitor, Sirna Therapeutics, San Francisco, Calif.
  • Neovastat terna Zentaris Inc; Quebec City, Calif.
  • the present invention encompasses combination therapy in which the formulations and compositions as disclosed herein are used in combination or conjunction with therapeutics, physiotherapy and/or behavioral psychotherapy used in the treatment of rheumatoid arthritis, obesity, endometriosis, idiopathic pulmonary fibrosis (IPF), lupus, and Alzheimer's disease.
  • therapeutics physiotherapy and/or behavioral psychotherapy used in the treatment of rheumatoid arthritis, obesity, endometriosis, idiopathic pulmonary fibrosis (IPF), lupus, and Alzheimer's disease.
  • IPF idiopathic pulmonary fibrosis
  • NSAIDS non-steroidal anti-inflammatory drugs
  • corticosteroids such as prednisone or cortisone
  • anti-malarial medications such as hydroxychloroquine
  • gold such as hydroxychloroquine
  • sulfasalazine gold
  • penicillamine a member of the group consisting of hydroxychloroquine
  • cyclophosphamide a member of the group consisting of hydroxychloroquine
  • cyclosporine a member of the group consisting of hydroxychloroquine
  • minocycline such as hydroxychloroquine
  • interleukin receptor antagonist such as interleukin receptor antagonist
  • the formulations and compositions as disclosed herein are particularly useful for administration in conjunction therapies used for treatment of diseases associated with vascular permeability, such as vascular complications of diabetes such as non-proliferative diabetic retinopathy and nephropathy, nephrotic syndrome, pulmonary hypertension, burn edema, tumor edema, brain tumor edema, IL-2 therapy-associated edema, and other edema-associated diseases, as disclosed in International Application No: WO2003/086178 and U.S. Patent Applications US2005/0203013 and US2005/0112063 which are incorporated herein in their entirety by reference.
  • diseases associated with vascular permeability such as vascular complications of diabetes such as non-proliferative diabetic retinopathy and nephropathy, nephrotic syndrome, pulmonary hypertension, burn edema, tumor edema, brain tumor edema, IL-2 therapy-associated edema, and other edema-associated diseases, as disclosed in International Application No
  • the formulations and compositions as disclosed herein are particularly useful for administration in conjunction with IL-2 therapy, where the limiting factor of IL-2 therapy is IL-2 therapy-associated edema as disclosed in International Application No: WO2003/086178 and U.S. Patent Applications US2005/0203013 and US2005/0112063.
  • Treatment for Alzheimer's disease include, but are not be limited to, nonsteroidal anti-inflammatory drugs (NSAIDs), estrogen, steroids such as prednisone, vitamin E, menantine, donepezil, rivastigmine, tacrine, and galantamine.
  • NSAIDs nonsteroidal anti-inflammatory drugs
  • steroids such as prednisone, vitamin E, menantine, donepezil, rivastigmine, tacrine, and galantamine.
  • Holistic medicine include example such as gingko nuts extracts.
  • Treatment of endometrosis include, but should not be construed as limited to, a combination oral contraceptives (estrogen plus a progestin), progestins (such as medroxyprogesterone, danazol (a synthetic hormone related to testosterone, gonadotropin-releasing hormone agonists (GnRH agonists such as buserelin, goserelin, leuprolide and nafarelin), and nonsteroidal anti-inflammatory drugs (NSAIDs) for pain control.
  • a combination oral contraceptives estrogen plus a progestin
  • progestins such as medroxyprogesterone, danazol (a synthetic hormone related to testosterone, gonadotropin-releasing hormone agonists (GnRH agonists such as buserelin, goserelin, leuprolide and nafarelin
  • NSAIDs nonsteroidal anti-inflammatory drugs
  • Examples of treatment options for obesity include dieting and nutritional counseling, exercise regime, gastric-bypass surgery, and drugs such as a combination of fenfluramine and phentermine (often called fen-phen), orlistat, sibutramine, phentermine, benzphetamine, diethylpropion, mazindol, and phendimetrazine.
  • the formulations and compositions can comprise a plurality of micelles, wherein some micelles comprise MetAP-2 inhibitor, and other micelles comprise other therapeutic agents, for example chemotherapeutic agents and antineoplastic agents.
  • the amount of the pharmaceutical composition as disclosed herein when administered to a subject will, of course, be dependent on the subject being treated, the severity of the affliction, the manner of administration, the judgment of the prescribing physician, etc.
  • the formulations and pharmaceutical composition can, if desired, be presented in a suitable container (e.g., a pack or dispenser device), such as an FDA approved kit, which can contain one or more unit dosage forms containing the carrier portion containing the targeting and immune response triggering portions.
  • a suitable container e.g., a pack or dispenser device
  • FDA approved kit which can contain one or more unit dosage forms containing the carrier portion containing the targeting and immune response triggering portions.
  • a given agent for example a fumagillol derivative
  • a given agent for example a fumagillol derivative
  • an anti-proliferative or anti-angiogenic agent for example a fumagillol derivative
  • a composition or formulation comprising a MetAP-2 inhibitor block copolymer conjugate in accordance with the present invention can treat or prevent diseases associated with an angiogenesis-mediated condition can be determined by its effect in the mouse model as shown in the Examples below.
  • a MetAP-2 inhibitor block copolymer as described herein will have anti-proliferative activity (i.e., at least 50% of the anti-proliferative activity of TNP-470) in a HUVEC assay as described in the Examples herein.
  • Another useful assay for determining if the compositions and formulations has disclosed herein have anti-proliferative or anti-angiogenesis activity is the CAM assay, which is frequently used to evaluate the effects of endothelial cell proliferation regulating factors because it is relatively easy and provides relatively rapid results.
  • a proliferation-regulating factor useful in the methods and compositions described herein will modify the number of microvessels in the modified CAM assay described by Iruela-Arispe et al., 1999, Circulation 100: 1423-1431. The method is based on the vertical growth of new capillary vessels into a collagen gel pellet placed on the CAM.
  • the collagen gel is supplemented with a proliferative factor such as FGF-2 (50 ng/gel) or VEGF (250 ng/gel) in the presence or absence of test agents.
  • a proliferative factor such as FGF-2 (50 ng/gel) or VEGF (250 ng/gel) in the presence or absence of test agents.
  • the extent of the proliferative response is measured using FITC-dextran (50 ⁇ g/mL) (Sigma) injected into the circulation of the CAM.
  • the degree of fluorescence intensity parallels variations in capillary density; the linearity of this correlation can be observed with a range of capillaries between 5 and 540. Morphometric analyses are performed, for example, by acquisition of images with a CCD camera.
  • Images are then analyzed by imported into an analysis package, e.g., NHImage 1.59, and measurements of fluorescence intensity are obtained as positive pixels.
  • Each data point is compared with its own positive and negative controls present in the same CAM and interpreted as a percentage of inhibition, considering the positive control to be 100% (VEGF or FGF-2 alone) and the negative control (vehicle alone) 0%.
  • Statistical evaluation of the data is performed to check whether groups differ significantly from random, e.g., by analysis of contingency with Yates' correction.
  • Additional proliferative assays including additional assays specifically designed to monitor angiogenic proliferation are known in the art and can be used to evaluate MetAP-2 inhibitors for use in the methods and compositions described herein. These include, for example, the corneal micropocket assay, hamster cheek pouch assay, the Matrigel assay and modifications thereof, and co-culture assays. Donovan et al. describe a comparison of three different in vitro assays developed to evaluate angiogenesis regulators in a human background (Donovan et al., 2001, Angiogenesis 4: 113-121, incorporated herein by reference).
  • the assays examined include: 1) a basic Matrigel assay in which low passage human endothelial cells (Human umbilical vein endothelial cells, HUVEC) are plated in wells coated with Matrigel (Becton Dickinson, Cedex, France) with or without angiogenesis regulator(s); 2) a similar Matrigel assay using “growth factor reduced” or GFR Matrigel; and 3) a co-culture assay in which primary human fibroblasts and HUVEC are co-cultured with or without additional angiogenesis regulator(s), the fibroblasts produce extracellular matrix and other factors that support HUVEC differentiation and tubule formation.
  • HUVEC Human umbilical vein endothelial cells
  • the co-culture assay provided microvessel networks that most closely resembled microvessel networks in vivo.
  • the basic Matrigel assay and the GFR Matrigel assay can also be used by one of skill in the art to evaluate whether a given fumagillol derivative is an angiogenesis-inhibiting agent as necessary for the methods and compositions described herein.
  • an in vitro angiogenesis assay kit is marketed by Chemicon (Millipore).
  • the Fibrin Gel In Vitro Angiogenesis Assay Kit is Chemicon Catalog No. ECM630.
  • proliferative assays are disclosed in International Application No: WO2003/086178 and U.S. Patent Applications US2005/0203013 and US2005/0112063, and involve assaying endothelial cells on a permeable substrate (e.g., a collagen coated inserts of “Transwells”), contacting the assay with a test compound (e.g., a fumagillol derivative block copolymer conjugate), treating the assay with a marker (e.g., FITC label) and a permeability-inducing agent (e.g., vascular endothelial growth factor (VEGF) and platelet-activating factor (PAP) among others), and measuring the rate of diffusion of the marker compare to control.
  • a permeable substrate e.g., a collagen coated inserts of “Transwells”
  • a test compound e.g., a fumagillol derivative block copolymer conjugate
  • a marker e.g., FIT
  • MetAP-2 inhibitor block copolymer conjugate as an anti-proliferative or angiogenic agent in a patient being treated with such a block copolymer conjugate can be assessed by methods commonly known by person skilled in the art, for example, as disclosed in International Application No: WO2003/086178 and U.S. Patent Applications US2005/0203013 and US2005/0112063.
  • One approach involves administering to the patient an intradermal injection of histamine before treating the subject with the a MetAP-2 block copolymer conjugate and measuring a histamine-induced local edema.
  • composition or formulation as disclosed herein capable of preventing or treating non-proliferative diabetic retinopathy can be tested by in vitro studies of endothelial cell proliferation and in other models of diabetic retinopathy, such as Streptozotocin.
  • color Doppler imaging can be used to evaluate the action of a drug in ocular pathology (Valli et al., Ophthalmologica 209(13): 115-121 (1995)).
  • Color Doppler imaging is a recent advance in ulkasonography, allowing simultaneous two-dimension imaging of structures and the evaluation of blood flow. Accordingly, retinopathy can be analyzed using such technology.
  • compositions and formulations as disclosed herein are administered is dependent, in part, upon whether the treatment of a disease associated with vascular hyperpermeability, including non-proliferative retinopathy is prophylactic or therapeutic.
  • the manner in which compositions and formulations as disclosed herein are administered for treatment of retinopathy is dependent, in part, upon the cause of the retinopathy.
  • the compositions and formulations as disclosed herein can be administered preventatively as soon as the pre-diabetic retinopathy state is detected.
  • mice C57/B16J, 8 weeks old were purchased from Jackson Laboratories (Bar Harbor, Me., USA).
  • non pigmented Lewis rats 8-10 wk old, Charles River Laboratories were used. All protocols were approved by the Institutional Animal Care and Use Committee at Children's Hospital Boston and were conducted in accordance with the Association for Research in Vision and Ophthalmology's Statement for the Use of Animals in Ophthalmic and Vision Research.
  • Aqueous solubility is one of the important chemical properties affecting oral absorption of a drug.
  • Log-D value is a parameter of hydrophobicity determined by the ratio of drug concentration in octanol to that in water at 25° C. (Analiza, Cleveland Ohio).
  • High Log-D values (>2) indicate low water solubility and hence a poor oral availability of a drug.
  • TNP-470 (Takeda) was chosen for its activity, hydrophobicity and poor pharmacokinetic profile (see Placidi, Cancer Res. 55, 3036-3042 (1995)) and was bound to a diblock co-polymer using a two-step conjugation ( FIG. 6A ).
  • succinated monomethoxy-poly(ethylene glycol)-poly (lactic acid) mPEG2000-PLA1000
  • free carboxic acid end-groups Advanced Polymers Materials Inc.
  • ethylendiamine A five fold molar excess of ethylendiamine was added and reacted for 4 h at 25° C.
  • the polymer solution was then dialyzed (MWCO 1000, Spectra/Por Biotech Regenerated Cellulose VWR) against DMSO leading to a 65% reaction efficiency.
  • the amine containing polymer was mixed with TNP-470 (350 gr), dissolved in DMSO and the solution was stirred for 4 h at 25° C.
  • the polymeric micelles were formed by dialyzing the DMSO solution of the conjugate against double distilled water using a regenerated cellulose dialysis bag (MWCO 1000, Spectra/Por Biotech Regenerated Cellulose, VWR) to obtain micelles with high incorporation efficiency (>90%) and 0.8-1% free drug (w/w). The micelles were then lyophilized and stored at ⁇ 20° C. in a dry environment until use.
  • MWCO 1000 Spectra/Por Biotech Regenerated Cellulose, VWR
  • Rhodamine labeled polymeric-micelles were formed using a similar protocol.
  • the mPEG-PLA was conjugated via the N-terminal amine group with Lissamine rhodamine B sulfonyl chloride (Molecular Probes, Eugene, Oreg.) in DMSO.
  • Lissamine rhodamine B sulfonyl chloride Molecular Probes, Eugene, Oreg.
  • DMSO Lissamine rhodamine B sulfonyl chloride
  • a commonly used hydrophobic marker 6-coumarin Sigma-Aldrich, MO, USA
  • NMR Nuclear Magnetic Resonance
  • NERCE/BEID Nuclear Magnetic Resonance
  • Mass spectrometry Mass spectrometry
  • TNBSA 2,4,6-Trinitrobenozene Sulfonic Acid
  • TNP-470 concentration was measured using a high-performance liquid chromatography system (HPLC, System Gold® Microbore, Beckman Coulter).
  • TNP-470 binding to amine was also measured using TNBSA reagent and was confirmed by subtracting the non-bound drug from the total drug added to the reaction.
  • TNBSA (2,4,6-Trinitrobenozene Sulfonic Acid
  • the reaction was carried out after the addition of 0.01% (w/v) TNBSA in 0.1M Sodium bicarbonate buffer.
  • a series of PEG-PLA polymer concentrations before and after TNP-470 conjugation were allowed to react for 2 hr in 37° C. with TNBSA and the yellow color was measured using a plate reader.
  • the correlation between the molar amine concentration and color absorption at 450 nm was determined according to a standard curve done for aspartic acid.
  • Murine Lewis lung carcinoma (LLC) and B16/F10 melanoma cells were obtained from American Type Culture Collection (ATCC, Manassas, Va., USA). Human Umbilical Vascular Endothelial cells (HUVEC) were purchased from Cambrex (USA). The cells were grown and maintained in medium as recommended by the manufacturers. Dulbecco's Modified Eagle's Medium with 10% fetal bovine serum was used for tumor cells and EMB-2 (Cambrex Bio Science Walkersville, Inc.) containing 2% fetal bovine serum and EGM-2 supplements was used for HUVEC.
  • EMB-2 fetal bovine serum
  • RPE cells were cultivated as has been previously described 59 . As determined by flow cytometry, the primary RPE cultures were found to be more than 98% cytokeratin positive (Clone PCK-26, Sigma). HMVECs, were purchased from Cambrex Bio Science Inc. (Rockland, Me.) and were maintained on collagen I coated plates with endothelial cell growth medium containing 2% FBS (EGM-2; Cambrex, Inc.) at 37° C. with humidified 95% air/5% CO 2 .
  • HUVEC were exposed to different concentrations of Lodamin equivalent to 50-1,000 nM free TNP-470 (0.12-2.4 mg/ml micelles) and incubated in a low serum medium for 48 h at 37° C.
  • Lodamin equivalent to 50-1,000 nM free TNP-470 (0.12-2.4 mg/ml micelles) and incubated in a low serum medium for 48 h at 37° C.
  • empty micelles were added to HUVECs as a control (4.8 mg/ml).
  • a WST-1 proliferation assay (Roche Diagnostics) was used. Cell viability was calculated as the percentage of formazan absorbance at 450 nm of treated versus untreated cells. Data were derived from quadruplicate samples in two separate experiments.
  • the effect of Lodamin (60 nM TNP-470 equivalent q.o.d. on HUVEC growth rate was evaluated by daily counting of HUVEC cells up to 5 d and compared to the number of untreated cells or cells treated with Vehicle (same concentration as
  • HUVEC 6-coumarin labeled PEG-PLA micelles or rhodamine conjugated to mPEG-PLA were used.
  • HUVEC were seeded in a 24-well plate (2 ⁇ 10 4 per well) in EGM-2 medium (Cambrex) and were allowed to attach overnight.
  • Fluorescent-labeled micelles (10 mg/ml) were suspended over a bath sonicator for 5 min, and 20 ⁇ l of the suspension was added to the cultured cells. After the designated time points (20 min, 2, 4, 7 and 24 h), the cells were washed three-times with PBS and analyzed by flow cytometry, or alternatively fixed with 4% parformaldehyde.
  • confocal microscopy cells were mounted using DAPI containing Vectashild (Vector, Laboratories, Burlingame, Calif.). Optical sections were scanned using Leica TCS SP2 AOBS a ⁇ 40 objective equipped with 488-nm argon, 543-nm HeNe, and 405-nm diode lasers. To study Lodamin internalization into endothelial cells, confocal microscopy was used to co-localize 6-coumarin labeled polymeric-micelles with endo-lysozome. Live HUVEC cells were imaged in different time points after addition of labeled micelles to cell media (15 ⁇ g/ml) up to 1 h.
  • LysoTracker Red® (Molecular Probes, Eugene, Oreg.) was added to the medium for the detection of acidic intracellular vesicles: Endosomes and Lysosomes. After 20 min of incubation, cells were imaged by confocal using optical sections with 488-nm argon, 543-nm HeNe, and 405-nm diode lasers.
  • TEM Transmission Electron Microscopy
  • the average size and size distribution of polymeric micelles were determined by Dynamic Light Scatterer (DLS, DynaPro, Wyatt Technology). The measurements were done at 25° C. using Dynamic V6 software. Lodamin (TNP-470 polymersomes) at 1.5 or 2 mg/ml dispersed in double-distilled water were measured by 20 successive readings with DLS. Micelles' size was also measured after 24 hr to evaluate their structural stability.
  • mice Animal procedures were performed in the animal facility at Children's Hospital. Eight-week-old male C57BL/6 mice (Jackson Laboratories, Bar Harbor, Mass.) were injected subcutaneously with 1 ⁇ 10 6 Lewis Lung Carcinoma cells (LLC) at the back. When tumors reached a volume of 100 mm 3 the mice were divided into 5 groups, and treated every day for 12 days with TNP-470 polymeric micelles, administrated orally using a gavage needle. Different doses of polymersomes were given daily: 15 mg/kg (5 mice), 30 mg/kg (3 mice) and 60 mg/kg (1 mice) TNP-470 equivalent, respectively (5, 10, 20 mg polymeric drug per mouse, respectively). The control groups were five mice that were given the same dose of polymersomes without the drug, and five mice that were given drinking water.
  • LLC Lewis Lung Carcinoma cells
  • mice Every two days the weight of the mice was monitored and tumors dimensions were measured using a caliper. Tumor volume was calculated according to an ellipsoid formula.
  • a non-invasive IVIS 200 in-vivo imaging system (Xenogen system 3.0) was used in order to track the polymeric micelles labeled with fluorescent marker in a manner similar to the cell uptake study. Fluorescent polymersomes were given orally to three nude mice, the mice were anesthetized using an Isofluran chamber, and images were taken after different time periods. All the images were taken under identical conditions.
  • the corneal micropocket angiogenesis assay was performed as previously detailed 29 .
  • Pellets containing 80 ng carrier-free recombinant human bFGF or 160 ng VEGF (R&D Systems, Minneapolis, Minn.) were implanted into micropockets created in the cornea of anesthetized mice. Mice were treated daily with 15 mg/kg TNP-470 equivalent of Lodamin for 6 d, and then the vascular growth area was measured using a slit lamp.
  • Tissue autofluorescence was corrected by subtracting the fluorescent signal of non-treated mouse organs from the respective readings in treated mice.
  • levels of fluorescent signal in mouse sera were measured in different time points (1, 2, 4, 8, 24, 48 and 72 h) using excitation/emission readings at 488 nm/530 nm.
  • PEG-PLA-rhodamine micelles 100 ⁇ l of 1.5 mg/ml or water were orally administered to C57B1/6J mice bearing LLC tumors (200 mm 3 ) for 3 d. Organs were removed, incubated for 50 min in collagenase (Liberase Blendzyme 3; Roche Diagnostics Corp., IN) in 37° C. to obtain a single cell suspension. These suspensions were analyzed by flow cytometry in order to quantify the uptake of micelles into different tissue cells when compared to those in the untreated mouse.
  • mPEG-PLA-rhodamine micelles were orally administered to C57B1/6J mice after 8 h of food fasting. After 2 h mice were sacrificed and 2.5 cm segments of the small intestine were removed, washed, and analyzed by histology and confocal microscopy. The rhodamine-labeled polymeric micelles were detected by confocal microscopy (Leica TCS SP2 AOBS) with a 488-nm argon laser line. Actin filaments were stained with phalloidin-FITC (Sigma) and nuclei were stained by DAPI (Sigma).
  • LLC cells (1 ⁇ 10 6 ) or B16/F10 melanoma cells (1 ⁇ 10 6 ) were implanted subcutaneously in 8 week old C57B1/6J male mice (Jackson Laboratories, Bar Harbor, Me.). Oral availability of free TNP-470 was compared to that of Lodamin. A dose of 30 mg/kg q.o.d. of free TNP-470 and an equivalent dose of Lodamin (Lodamin, 6 mg in 100 ⁇ l/d) were administered to LLC tumor bearing mice ( ⁇ 100 mm 3 ) and tumor growth was followed for 18 d. Free drug was given as a suspension in double distilled water and freshly prepared before each dose.
  • Lodamin was administered to B16/F10 melanoma bearing mice.
  • tumor size and animal weight were monitored every 2 d.
  • Tumor volume was measured with calipers in two diameters as follows: (width) 2 ⁇ (length) ⁇ 0.52. Note that all the above Lodamin doses are presented as TNP-470 equivalent.
  • liver metastases were generated by spleen injection.
  • mice were killed and autopsied, livers and spleens were removed by surgical dissection, imaged, and histology was carried out. Liver and spleen tissues were stained with Hematoxylin & Eosin to evaluate tissue morphology and detect metastasis. Immunohistochemistry was carried out to specifically detect B16/F10 cells in the liver using anti-mouse melanoma antibody (HMB45, abCAM) and using DAB staining.
  • HMB45 anti-mouse melanoma antibody
  • mice At the end of the training trials, no freezing behavior was observed, and the mice would start to walk within 4 seconds of being placed on the beam.
  • the mice were then videotaped as they performed in 3 test trials of 3 beam crossings each—a total of 9 crossings per mouse.
  • the 3 trials were separated by at least 1 h to avoid fatigue of the mice.
  • Videotaped crossings were scored for number of foot-slip errors and time to cross. All experiments and scoring of the different groups were performed by an investigator who has been no knowledge regarding the treatment regime of the mice.
  • Histologic evaluation of tissue was performed on 8 ⁇ m thick frozen sections of LLC tumors that were removed from two random Lodamin-treated or untreated mice 14 d post treatment (15 mg/kg q.d. TNP-470 equivalent). Tumors were sectioned and analyzed for cell markers, 20 microscope fields ( ⁇ 400) were imaged.
  • Tissues were stained with Hematoxylin & Eosin to detect tissue morphology.
  • Immunohistochemistry was carried out using Vectastain Elite ABC kit (Vector Laboratories, Burlingame, Calif.). Primary antibodies included CD31 (BD Biosciences, San Jose, Calif.) for microvessel staining and anti-Ki-67 (DAKO, CA, USA) detection of proliferation. Detection was carried out using a 3,3′-diaminobenzidine chromogen, which results in a positive brown staining. Apoptotic cells were detected by reacting the tissues with Terminal deoxynucleotide transferase mediated dUTP-biotin nick end labeling (TUNEL) using a kit (Roche) following the company's protocol. Vessels were detected in the same tissues by anti-CD31 and secondary FITC anti-mouse antibody (Jackson ImmunoResearch) conjugated antibody (green) and nuclei were detected by DAPI (blue).
  • TUNEL Terminal deoxynucleotide transferase mediated dUTP-biot
  • DTH reactions were induced in the skin of 8-week-old C57B1/6J male mice as previously reviewed 62 .
  • Laser-induced CNV was generated by a technique described previously with some modifications 63 . Only lesions in which a subretinal bubble or focal serous detachment of the retina developed were used for the experiments. Lodamin treatments were started at the day of CNV induction and the sizes of CNV lesions were determined after 7 or 14 d. For this purpose, four burns were performed per eye while leaving a space around the optic disc. After 7 or 14 d of laser induction, the retinas were analyzed for blood vessel formation.
  • Miles assay was performed in C57B1/6J mice 5 d after oral Lodamin treatment (30 mg/kg/day). MCP-1 and VEGF-165 were used as inducers.
  • CNV lesions of the same size were used to compare blood vessel permeability in mice and rats. Two different assays were performed; Angiography in live rats and modified Miles assay in mice. A modified Miles assay was performed as described in the Examples. Angiography was performed on rats using Fluorescein imaging as previously described 64 .
  • Single cell suspensions were prepared from C57BL/6J mouse retinas with CNV lesions. In order to collect a sufficient number of ocular-infiltrating cells for flow cytometry, 20 separate burns were delivered similar to the panretinal photocoagulation procedure in humans. The quantity of macrophages in single-cell suspensions from mouse retinas from days 3 and 7 post laser procedure was evaluated for Lodamin treated (oral, 30 mg/kg q.d for 3 days) or vehicle or untreated mice. In the third day of treatment mice were euthanized and analyses of their retinas were performed.
  • In-vitro data are presented as mean ⁇ SD, whereas in-vivo data are presented as mean ⁇ SE. Differences between groups were assessed using unpaired two-tailed Student's t-test, and P ⁇ 0.05 was considered as statistically significant.
  • Block copolymer micelles have been proposed for the delivery of hydrophobic drugs with low aqueous solubility (such as Paclitaxel).
  • the amphiphilic nature of diblock copolymers such as poly (lactide)-poly(ethylene glycol) (PLA-PEG), enables the formation of micelles in aqueous media.
  • the diblock copolymer In an aqueous environment, the diblock copolymer is self-assembled into a structure of hydrophobic core, formed by the association of the hydrophobic polymer and the drug, and a hydrophilic shell, formed by the hydrophilic polymer, commonly PEG.
  • Block copolymers as disclosed in U.S. Pat. No. 4,745,160 have been used form water insoluble, amphiphilic, non-crosslinked linear, branched or graft block copolymers having polyethylene glycol as the hydrophilic component and poly(D-, L-, or D, L-lactic acids) as the hydrophobic components.
  • Block copolymers as disclosed in U.S. Pat. No. 5,543,158 have been described to form nanoparticles or microparticles that are solid particles that are suspended in water that are formed from a water-insoluble block copolymer consisting essentially of poly(alkylene glycol) and poly(lactic acid). The molecular weight of the block copolymer is high and the copolymer is insoluble in water.
  • the biodegradable moieties of the copolymer are in the core of the nanoparticle or microparticle and the poly(alkylene glycol) moieties are on the surface of the nanoparticle or microparticle in an amount effective enough to decrease uptake of the nanoparticle or microparticle by the reticuloendothelial system.
  • Nanoparticles are prepared by dissolving the block copolymer and drug in an organic solvent, forming an oil and water emulsion by sonication or stirring, and collecting the nanoparticles containing the drug following precipitation. It does not provide for the solubilization of hydrophobic drugs.
  • the inventors demonstrate that MetAP-2 inhibitor TNP-470 was successfully conjugated to a modified PEG-PLA polymer through its amine, and formed nano-size polymersomes.
  • a schematic diagram of the conjugation process is shown in FIG. 6A .
  • Step I demonstrates the reaction between succinated mPEG-PLA and ethylendiamine that results in an amine terminated polymer.
  • Step II demonstrates the reaction between the amine reactive polymer and the terminal chlorine of TNP-470.
  • the conjugate is then dialyzed against water in an excess of TNP-470 to form polymeric micelles.
  • the inventors demonstrate, using images taken by TEM, that spherical micelles were formed and size measurement with DLS showed a low range of size distribution around 10 nanometers.
  • HUVECs Human Umbilical Vein Endothelial Cells
  • mice In-vivo studies done on mice showed a significant inhibition of subcutaneous Lewis Lung Carcinoma tumors with C57/BL mice given a daily oral administration of TNP470 micelles. A dose of 15 mg/kg TNP-470 equivalent showed 63% inhibition without any weight loss to the mice.
  • NMR results from the different reaction steps are shown in FIG. 1 .
  • the results show the binding of the ethylendiamine through the carboxic acid group at the original polymer, and the TNP-470 binding to the polymer through the amine.
  • TNP-470 To further verify the binding of TNP-470 to the modified PEG-PLA through the amine in the polymer and the terminal chlorine in the drug, a TNBSA reagent reaction for the detection of free amino groups was performed. According to the results ( FIG. 2 ) the polymer was saturated with ethylendiamine. Correlation between molar amine groups in the modified polymer and absorption in 450 nm were determined by a theoretical value from a standard curve done for aspartic acid (not shown). Moreover, after TNP-470 conjugation, no free amines were detected, indicating that the polymer was saturated with TNP-470 conjugated through the amine group at the polymer.
  • Polymersomes dispersed in double distilled water were imaged with cryo-TEM (JEOL 2100 TEM, Harvard university—CNS). The images show that the self-assembled polymersomes had a uniform spherical morphology.
  • FIG. 4 shows the inhibition in proliferation after 48 hr of incubation.
  • FIG. 4A shows the effect of TNP-470 micelles
  • FIG. 4B shows the effect of micelles without drug on HUVECs.
  • a significant inhibition of HUVEC proliferation was detected from 0.3 mg/ml and up to 2.4 mg/ml micelles, which are 62.5 nM-1000 nM TNP-470 equivalent (88% and 95% inhibition respectively).
  • the same concentrations of polymeric micelles without TNP-470, as well as double concentration (4.8 mg/ml) showed no significant effect on HIJVEC proliferation.
  • HUVECs were incubated with fluorescent polymersomes, washed and imaged by Confocal microscopy. HUVECs were incubated with the micelles for 20 min, 4 hr and 24 hr. After 20 min. micelles were already uptaken by the cells and located at the cytoplasm (data not shown). After 2 hr, 4 hr, 7 hr and 24 hr the uptake increased and the micelles were detected as concentrated spots inside cell cytoplasm (data not shown).
  • C57/BL mice bearing s.c LLC tumors showed inhibition in tumor growth when treated with PEG-PLA-TNP-470 micelles given orally. All doses showed a significant effect already after 14 days of daily treatment, as shown in FIG. 5A . The controlled groups had to be scarified on day 14, and the treatment continued up to day 18. The lowest dose (15 mg/kg TNP-470 equivalent) showed a significant inhibition of tumor growth (63%) after 14 days (*p ⁇ 0.05), without any weight loss to the mice. whereas higher doses of 30 mg/kg and 60 mg/kg TNP-470 equivalent showed 74%, 75% inhibition respectively, but also caused almost 20% weight loss ( FIG. 5B ).
  • the non-invasive Xenogen system was used to track the absorption kinetics of the polymersomes after oral administration.
  • the micelles were concentrated at the gastrointestinal tract within 10 min post administration (GI track) (data not shown). Apparently the absorption was rapid and after 1 hr fluorescent signal was found near the bladder and the prostate glands. After 24 hr the signal was still detectable.
  • Lodamin and the conjugation of TNP-470 are described in detail in the Methods.
  • the chemical and physical properties of Lodamin was characterized.
  • the binding of TNP-470 to mPEG-PLA was confirmed by 3 H NMR (data not shown) and mass spectrometry showing an average m/z of 3687 for mPEG-PLA-TNP-470.
  • FIG. 6A demonstrates the reaction of Lodamin preparation. Using amine detection reagent, the incorporation efficiency of ethylenediamine to mPEG-PLA was determined as 65%, and in the second step TNP-470 was shown to be bound in high efficiency of >90%. Lodamin contained 0.8-1% (w/w) free TNP-470 as determined HPLC.
  • the majority of micelles (90%) at day of preparation were 7.8-8 nm in diameter, with a small population of larger particles (200-400 nm). The size remained almost unchanged after 10 days.
  • the morphology of Lodamin was characterized by TEM. ( FIG. 6B ).
  • the images showed that the polymeric micelles had acquired a uniform spherical structure, which remained stable after two weeks of incubation in water at 37° C. Since the drug is located in the PLA core of the micelle structure and PLA is spontaneously hydrolyzed in an aqueous environment, we studied the release kinetics of TNP-470 from Lodamin.
  • the TNP-470 was released over a period of 28 d with an early peak burst of ⁇ 30% after the first day of incubation ( FIG. 6C ).
  • Endothelial Cells Take Up MetAP-2 Inhibitor Polymersomes by Endocytosis
  • HUVEC The uptake of polymeric micelles by HUVEC and its kinetics was evaluated initially.
  • HUVEC were incubated with 6-coumarin labeled mPEG-PLA micelles for 20 min, 2, 4 and 7 h and were imaged by confocal microscopy. Confocal images show HUVEC uptake of polymeric micelles labeled with 6-coumarin after 20 min and 7 h incubation time periods (data not shown).
  • LysoTracker Red® was used to detect Endosomes and Lysosomes. Overlay between micelles and Endo-lysosomes is represented in yellow/orange color (data not shown).
  • Lodamin (62.5 nM-1,000 nM TNP-470 equivalent) inhibited HUVEC proliferation by 88% to 95% respectively after 48 h ( FIG. 7B ).
  • MetAP-2 Inhibitor Polymersomes Inhibit VEGF and bFGF-Induced Angiogenesis In Vivo
  • Lodamin The anti-proliferative and antiangiogenic properties of Lodamin were evaluated in-vivo by the corneal micropocket assay 29 .
  • mice were administered oral polymeric-micelles. After 2 h mice were euthanized and isolated segments of the small intestine were fixed and imaged using confocal microscopy. A histological section of gut epithelial cells of Lodamin treated mouse was observed with TEM. The polymeric micelles were intensively taken up by columnar epithelium lining the luminal side of the small intestine. In a high magnification of small intestine villi, micelles are clearly detected in the lamina intestinal and in the vicinity of the blood vessels, indicating transepithelial absorption.
  • Lodamin The biological efficacy of Lodamin was evaluated as an anti-cancer agent in tumor bearing mice.
  • mice were given oral free TNP-470 at a dose of (30 mg/kg q.o.d.) no inhibition in the growth of subcutaneous LLC tumors was observe ( FIG. 9A ).
  • the equivalent dose of Lodamin resulted in a significant inhibition of tumor growth ( FIG. 9A ).
  • This inhibition was observed after 12 d of Lodamin treatment, and at day 18 tumor growth was inhibited by 83%.
  • Different dosing strategies of Lodamin were tested: 15 mg/kg q.d., 30 mg/kg q.o.d. and 15 mg/kg q.o.d., resulting 87%, 77%, and 74% of tumor volume inhibition respectively ( FIG. 9B ).
  • the vehicle (PEG-PLA) showed no effect on tumor growth and was similar to untreated control mice ( FIG. 9C ).
  • FIG. 9E shows representative tumors of treated or untreated LLC or B16/F10 tumors.
  • mice were subjected to a sensitive test of motor coordination: crossing a narrow (4 mm) balance beam 30 .
  • Lodamin-treated mice (30 mg/kg q.o.d. for 14 d) performed in this challenge similar to control (water treated) mice, whereas mice injected with free TNP-470 (30 mg/kg q.o.d.) committed over twice as many errors (P ⁇ 0.0001).
  • Oral MetAP-2 Inhibitor Polymersomes Inhibit Angiogenesis and Cell Proliferation in Tumors and Induce Tumor Cell Apoptosis
  • LLC Lewis lung carcinoma
  • H&E Hematoxylin & Eosin
  • TUNEL staining was used for the detection of apoptotic cells.
  • Cell nuclei were stained with DAPI. Sections were counterstained with Eosin (nuclei).
  • the tumors of untreated mice had a net organization of small and large vessels with apparent lumen structure as demonstrated by CD-31 immunostaining.
  • Lodamin treated tumors formed very small undeveloped vessels (data not shown).
  • Lodamin treated tumors showed less cellular proliferation than untreated tumors, as detected by the nuclear marker KI-67.
  • TUNEL staining for the detection of apoptosis in the tissues indicated an enhanced apoptosis in Lodamin treated tumors.
  • Lodamin treated tumor had less vessels but high levels of apoptosis predominantly of tumor cells. In the control tissue, apoptotic cells were found in the capsule of the tumor but less in the center of the tissue.
  • TNP-470 a nontoxic oral formulation of the MetAP-2 inhibitor TNP-470, named Lodamin, as a potent anti-proliferative and anti-metastatic drug.
  • TNP-470 a highly potent anti-proliferative/antiangiogenesis agent, is a leading candidate for chronic administration for cancer maintenance therapy and metastasis prevention.
  • TNP-470 has very poor oral availability as illustrated by its high Log D values, indicating low water solubility 35 .
  • this property of TNP-470 was altered while retaining its activity by conjugating it with mPEG-PLA to form mPEG-PLA-TNP-470 polymeric micelles i.e. Lodamin.
  • Lodamin can be suspended in water to form polymeric micelles caused by the amphiphilic nature of PEG-PLA 26 .
  • the drug is located in the core of the micelle protected from the harsh gastrointestinal environment 36 .
  • Polymer micelles have previously been used for the delivery of hydrophobic drugs 37, 38 and gene delivery 39 .
  • Lodamin acquired a stable spherical morphology of nanomicelles, as imaged by TEM.
  • PLA which is a biodegradable and biocompatible polymer, hydrolyzes in an aqueous environment and allows a slow-release of TNP-470.
  • In-vitro studies showed a continuous release of TNP-470 from Lodamin over almost a month period, where the majority of the drug was released after 4 d (in both gastric and plasma pH conditions), although acidic environment is known to accelerate degradation of PLA we observed only a minor effect on day 15.
  • PEG-PLA micelles penetrated the gut epithelial layer into the submucosa as shown by using fluorescent marker.
  • the mechanism of Lodamin penetration to gut epithelial cells seems to be via endocytosis as detected by high resolution TEM imaging.
  • labeled micelles had long blood circulation time of at least 72 h post administration, a significant increase compared to free drug which was detected in mice sera up to 2 h post-administration.
  • Biodistribution showed relatively high concentration in the liver because oral administration delivers the drug directly from the intestine to the liver.
  • no liver toxicity was observed by histology and by liver enzymes profile after 20 d of daily Lodamin treatment.
  • Lodamin given orally to mice showed substantial anti-proliferative effects (83% reduction), while free TNP-470 given orally had no effect.
  • the effect on LLC growth was dose dependent, as 30 mg/kg q.o.d. was more effective than 15 mg/kg. q.o.d., and a dose of daily 15 mg/kg q.d. was more effective than 30 mg/kg q.o.d. (a double dose given every 2 d).
  • a similar anti-proliferative effect of Lodamin was also observed with melanoma B16/F10 tumors, confirming the broad biological effect of Lodamin, very much like the original free TNP-470.
  • liver metastasis is very common in many tumor types and has often been associated with poor prognosis and survival rate.
  • the inventors chose the intrasplenic model for induction of liver metastasis, in which the transition time of B16/F10 from spleen to liver microvasculature post spleen injection in mice was found to be very fast (20% of the injected cells after 15 min) 40. Mice injected with B16/F10 cells into the spleen had a low survival rate. They developed ascites, macroscopic malignant nodules, and extensive cirrhosis in their livers 20 d post injection.
  • oral Lodamin is therapeutically effective in the treatment of solid tumors and metastasis in mice. It captures the antiproliferative properties of free TNP-470 while adding important advantages: oral availability, improved biodistribution, tumor accumulation, continual release, improved solubility, proper clearance, and low related side-effects.
  • its use is contemplated, among others, for cancer patients as a long-term maintenance angiogenesis inhibitor to prevent tumor recurrence.
  • it is also contemplated for use as maintenance therapy for chronic MetAP-2-related diseases such as age-related macular degeneration, endometriosis and rheumatoid arthritis. Other contemplated uses are described elsewhere herein.
  • Lodamin The effect of Lodamin on human brain tumor (U87MG, Glioblastoma) was studied using subcutaneous tumor-bearing nude mice. Treatment with Lodamin started when tumors were in volume of ⁇ 100 mm 3 . Lodamin that was given by gavage ( FIG. 11A ) in a daily dose of 15 mg/kg TNP-470 equivalent, inhibited tumor volume by 70% after 30 days. When Lodamin was given in drinking water at the same dose ( FIG. 11B ) a 71% inhibition of tumor volume was obtained after 42 days.
  • mice C57/B1 mice were injected into their tail-veins with 2.5 ⁇ 105 B16/F10 cells. Day after injection the mice were divided into 3 groups: 10 mice were given a daily oral administration of Lodamin (15 mg/kg TNP-470 equivalent), 9 mice were given IP injection with the same Lodamin dose, and 10 mice remained untreated. Mice survival was followed up to 40 days post treatment.
  • Lodamin 15 mg/kg TNP-470 equivalent
  • Lodamin treated mice showed a prolonged survival as can be seen in Kaplan-Meier survival curve ( FIG. 12 ). After 32 days all control mice died, compared to 6 and 5 mice that lived at day 40 in the IP Lodamin treated group and Oral Lodamin group respectively.
  • In-Vivo Matrigel Angiogenesis Assay was conducted in order to assess the antiangiogenic properties of Lodamin.
  • Matrigel (BD bioscience) which was remained in 4° C. overnight was mixed with fibroblast growth factor-2 (FGF-2) to obtain a final FGF-2 concentration of 1 ⁇ g/ml.
  • FGF-2 fibroblast growth factor-2
  • Three mice got a daily oral treatment with Lodamin (15 mk/kg/day TNP-470 equivalent).
  • the Matrigel was mixed with Lodamin (5 mg/ml) and no further treatment was performed and three untreated mice with Matrigel implant were used as a control.
  • mice were euthanized and the Matrigel implant harvested, washed twice with PBS, and then one of the implant was immediately frozen on dry ice and used for histology assessment of blood vessels using CD-31 antibody.
  • the second implant was enzymaticaly digested using collagenase (Librease, Roche) to obtain single cell suspension which again was analyzed by FACS to quantify the number of endothelial cells using CD-31 and CD-45 antibodies (endothelial cells are CD31+CD45 ⁇ ).
  • Ocular neovascular-related diseases are associated with vision impairment or vision loss in millions worldwide.
  • the growth of abnormal, leaky blood vessels is a prominent component of several debilitating eye diseases including AMD, proliferative diabetic retinopathy (PDR), and retinopathy of prematurity (ROP).
  • PDR proliferative diabetic retinopathy
  • ROP retinopathy of prematurity
  • antiangiogenic strategies could be particularly beneficial in preventing and treating the progression of these diseases.
  • Corneal neovascularization is often the result of inflammation, chemical burns, and conditions related to hypoxia 48, 49 . These conditions are currently treated by indirect angiogenesis inhibitors such as steroids and immunosuppressants 48 .
  • VEGF Vascular Endothelial Growth Factor
  • Efforts to inhibit retinal neovascular disease such as ARMD have focused on the angiogenic factor Vascular Endothelial Growth Factor (VEGF) and such therapies are now FDA approved.
  • the most prominent class of anti-VEGF therapy is represented by ranibizumab and bevacizumab. These molecules are derived from a humanized anti-VEGF antibody which has the ability to reduce the retinal edema in ARMD 50 .
  • ranibizumab and bevacizumab These molecules are derived from a humanized anti-VEGF antibody which has the ability to reduce the retinal edema in ARMD 50 .
  • the effects of these therapies are short lived and thus repeated intravitreal injections every 4 weeks is necessary 51 . While clinical trials have shown a beneficial effect in 30-40% of treated patients 50, 51 , in many cases these therapies become less effective over time 52 and the long-term safety is still debated.
  • Lodamin is a novel polymeric antiangiogenic drug and the first oral formulation for TNP-470 55 .
  • Lodamin is a conjugate of TNP-470 and monomethoxy polyethyleneglycol-polylactic acid (mPEG-PLA) polymer that forms a micelle-like structure when introduced in aqueous solution.
  • the active molecule, TNP-470 is a synthetic analogue of fumagillin and a methionine aminopeptidase-2 (MetAP2) inhibitor.
  • the broad spectrum activity of TNP-470 was demonstrated in more than a hundred preclinical animal tumor studies. In clinical trials, some cancer patients demonstrated partial or complete responses 56, 57 .
  • TNP-470 also demonstrated poor pharmaceutical properties and dose-limiting reversible cerebellar side effects 58 . Therefore Lodamin was rationally designed to overcome these limitations. It acquires superior properties such as: water solubility, improved oral availability, prolonged half life, slow-release properties and minimal TNP-470-releated cerebellar side effects 55 . Lodamin had potent antiangiogenic effects in murine tumor and metastases models.
  • Lodamin is a first-in-its-class polymeric broad-spectrum antiangiogenic ophthalmic drug that targets numerous molecular pathways in addition to VEGF. Lodamin's unique chemical properties, combined with its mechanism of action, place it as a leading candidate for treating corneal and retinal neovascular diseases.
  • FIGS. 16A and 16G Angiogenesis in the Matrigel was visually assessed after removal of the plugs 7 days post Matrigel injection ( FIGS. 16A and 16G ) and by immunohistochemical staining of blood vessels using CD31 antibodies ( FIG. 16A ).
  • Infiltrated endothelial cells and macrophages in Matrigel matrices were quantified in a single-cell suspension by fluorescence-activated cell sorting (FACS) ( FIG. 16C ).
  • FACS fluorescence-activated cell sorting
  • FIG. 17B shows representative images taken from the different mouse groups.
  • Lodamin reduces time course of delayed-type hypersensitivity DTH ear-swelling
  • Lodamin's effect on angiogenesis, inflammation, vascular hyperpermeability and edema were evaluated in the DTH reaction.
  • C57B1/6J mice were sensitized with oxazolone and treated with Lodamin orally (15 mg/kg q.d.) starting from 5 days after the initial exposure to oxazolone.
  • a significant reduction of ear erythema and swelling was obtained in Lodamin-treated mice compared with vehicle treated mice ( FIGS. 16D & 16F ).
  • H&E Hematoxillin and Eosin
  • Lodamin The effect of Lodamin on CNV progression in mice was determined after oral or intravitreal administrations. Oral Lodamin was administered in different doses and duration: 15 mg/kg/day (for 7 days or 14 d of treatment) and 30 mg/kg dose (7 days). CNV lesions in choroidal flat-mounts were evaluated after staining for blood vessels. CNV area was significantly reduced by Lodamin treatment in all treatment groups. Higher doses and longer treatment led to an enhanced effect: 14 day treatment (15 mg/kg q.d.) led to 70.6% inhibition whereas 7 day of treatment at the same concentration led to a 27.2% inhibition. At 30 mg/kg/day a more dramatic inhibition was obtained (50.5%) after 7 day.
  • FIG. 18A summarizes the CNV area of all treatment groups.
  • FIG. 18B H&E staining revealed that Lodamin substantially reduced the formation of fibrovascular response from the choroid induced by the spot rupture of Bruch's membrane by the laser.
  • FIG. 18C FACS in single-cell suspension originated from retina tissues following Lodamin treatment (30 mg/kg/day, oral). Retinal infiltrated macrophages were reduced after Lodamin treatment in mice (control group: 2% and 3.6%, Lodamin treated group: 1.8% and 2.3%, on day 3 and 7 respectively) ( FIG. 18C ).
  • Lodamin was introduced by a single intravitreal injection using 100 ⁇ g and 300 ⁇ g Lodamin, a 56% or 75% suppression was obtained respectively when measured after 14 days. No significant difference was found when injecting PBS compared to vehicle (data not shown).
  • FIG. 19 shows representative CNV lesions from the different groups and their size measurements. No toxicity was observed in histological H&E whole eye samples (data not shown).
  • retinal function was assessed by electroretinography (ERG) after Lodamin treatment.
  • ERG electroretinography
  • Responses to an ⁇ 6.6 log unit range of intensities of full-field flashes presented to the dark-adapted eye were recorded ( FIG. 19B ) as was the response to an 8 Hz flickering light presented in the presence of an adapting background ( FIG. 19C ).
  • Rod photoreceptor response sensitivity (Srod) and saturating amplitude (RmP3) were derived from the ERG a-wave and postreceptor sensitivity (kP2) and saturating amplitude (RmP2) were derived from the ERG b-wave ( FIG. 19D ).
  • the saturating energy in the ERG oscillatory potentials (Em), and the trough-to-peak amplitude of the flicker response (R8) were also determined.
  • Em ERG oscillatory potentials
  • R8 trough-to-peak amplitude of the flicker response
  • VEGF baseline levels in na ⁇ ve mouse eyes were almost as high as in CNV bearing mouse (313 pg/mg, 320 pg/mg respectively) compared to Lodamin treated mice that reduced VEGF levels 24% below the baseline (243 pg/mg).
  • MCP-1 levels in primary RPE cultures were also measured by ELISA and was found to be reduced by Lodamin in a dose dependant manner after 24 h treatment (39%, 55%, 83% reduction was obtained with 1, 10, 100 nM Lodamin, respectively). All ELISA data were normalized to total protein in each sample ( FIG. 20B ).
  • MMP-2 and 9 activities in CNV lesions were determined following oral Lodamin treatment (30 mg/kg q.d for 7 d) using zymograms ( FIG. 20D ).
  • the enzymatic activity was accelerated in tissues with CNV compared to na ⁇ ve mouse tissues whereas both MMP-2 and 9 was reduced by Lodamin to the levels of na ⁇ ve mice.
  • the levels of TNF ⁇ a major inflammatory cytokine, were higher in choroids than in retina and were reduced in the choroid by Lodamin ( FIG. 20C ).
  • NF-kB a pro-inflammatory transcription factor induced by TNF ⁇ , was detected in both retina and choroid tissues and was also reduced in the choroid by Lodamin.
  • Lodamin reduced mitogen-activated kinases (MAPK) and its phosphorylated form in the choroid with no effect in the retina ( FIG. 20C ).
  • MCP-1 has a role in Lodamin's effect on vessel permeability.
  • Representative mouse skin patches showing Evans blue leak in MCP-1 injection spots are presented in FIG. 21B .
  • the effect of Lodamin on MCP-1-induced vessel leakage was measured after 5 d of oral treatment with 30 mg/kg/day in C57B1/6 mice. Lodamin significantly reduced vessel permeability by 87%.
  • a standard Miles assay was performed with VEGF demonstrating 65% reduction of vascular leakage after the same oral treatment of 5 d.
  • Lodamin comprises many of the requirements for an advanced ocular delivery: it can be introduced into an aqueous solution, is nontoxic, biocompatible, biodegradable, and has slow-release properties 55 . This is the first report of Lodamin treatment for retinal neovascular disorders.
  • RPE cells play a central role in the progression of AMD disease. These cells are responsible for the generation and maintenance of the extracellular and photoreceptor matrices, as well as for the integrity of Bruch's membrane. Impaired function of RPE, occurs in ARMD and causes lipid and protein accumulation in the vicinity of Bruch's membrane and formation of drusen 70 . Furthermore, RPE cells secrete chemoattractant and inflammatory cytokines such as interleukin-8 (IL-8), MCP-1, and TNF ⁇ . As a result, neutrophils and macrophages are recruited and stimulate angiogenesis which contributes to ARMD progression 71 .
  • IL-8 interleukin-8
  • MCP-1 MCP-1
  • TNF ⁇ TNF ⁇
  • MCP-1 is a small cytokine of the C-C chemokine family and is a potent chemoattractant for neutrophils and macrophages with proangiogenic properties. MCP-1 has been shown to be elevated in the vitreous of patients with retinal neovascularization 71, 72 and to be involved in other systemic inflammatory and autoimmune diseases such as diabetes, rheumatoid arthritis and multiple sclerosis 73, 74 .
  • Lodamin effects were not limited to the suppression of MCP-1.
  • Other pro-angiogenesis and pro-inflammatory factors including VEGF, nuclear factor k-B (NF ⁇ B), TNF ⁇ and matrix metalloproteinases (MMPs), were also suppressed in situ by Lodamin treatment. Since NF ⁇ B regulates MCP-1 and is involved in the immune response 77 stimulated by TNF- ⁇ and IL-6, suppressing this pathway can potentially lead to a substantial reduction in inflammation and angiogenesis and thereby prevent the progression of ocular neovascular diseases.
  • RAGE a multiligand receptor of the immunoglobulin superfamily, and its ligands: advanced glycolyzation proteins (AGE), high-mobility group box 1 (HMGB-1) and S100 protein family
  • AGE advanced glycolyzation proteins
  • HMGB-1 high-mobility group box 1
  • S100 protein family are directly involved in inflammation, vascular and autoimmune diseases, diabetes, Alzheimer, cancer and angiogenesis 82-88 .
  • Protein analysis of CNV tissues revealed a dramatic reduction of the 35 kDa isoform of RAGE (the secreted isoform of RAGE described previously 89, 90 ) in retina and choroid tissues from CNV lesion treated by Lodamin.
  • HMGB-1 Amphoterin
  • MAPK activation e.g.
  • ERK/p-ERK enhance MCP-1 and TNF ⁇ production, upregulate VEGF in RPE, and increase MMPs activation 84, 92-99 . All these factors were shown to be suppressed by Lodamin in this CNV model, suggesting the possibility of additional indirect effects following the suppression RAGE activation.
  • S100A4 which is a RAGE ligand and a member of S100 family, can covalently bind MetAp2 100 , a molecular target of TNP-470.
  • S100 is a family of nonubiquitous cytoplasmic Ca2+-binding proteins that are expressed in a wide variety of cell types and are linked to many human pathologies 101-103.
  • the reduction of RAGE by TNP-470 provides a link between the known interactions of S 100-RAGE 104, 105 , S100-MetAp2 103 and TNP-470-MetAp2 106 . It is likely that MetAp2 is associated with RAGE signal and TNP-470 suppresses this signal in a direct or indirect way.
  • Lodamin can target broader molecular pathways, it may potentially improve therapeutic outcomes. It is well established that many factors are playing a central role in the progression of the diseases in addition to VEGF.
  • the specific structure of Lodamin as a polymeric nanomicelle, enhances its accumulation in CNV lesions compared to the injection of free small molecule (as seen in FIG. 21 ) suggesting an ideal biodistribution via systemic administration. This effect may be due to the Enhanced Permeability and Retention effect (EPR) 107 caused by the leaky vasculature in neovascular sites. Lodamin was well tolerated and showed minimal toxicity in the retina as indicated by histological analysis and ERG. Lastly, the slow release properties of Lodamin may minimize the dose and frequency of treatments, which is particularly important for intravitreal therapies. The current study therefore supports the fact that Lodamin is an excellent candidate for therapeutic treatment of ocular neovascular diseases in humans.

Abstract

Described herein are MetAP-2 inhibitors and compositions and formulations thereof, and more particularly compositions and formulations of MetAP-2 inhibitors wherein the MetAP-2 inhibitor is associated with a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety. The present invention also relates to compositions and formulations comprising MetAP-2 inhibitors for oral administration or administration via routes such as topical or ocular administration. The present invention also provides methods to treat conditions associated with or related to the over-expression or over-activity of MetAP-2 by administering the compositions and formulations comprising MetAP-2 inhibitors as disclosed herein.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation application of U.S. application Ser. No. 14/495,725 filed on Sep. 24, 2014, which is a continuation application of U.S. application Ser. No. 12/648,078 filed on Dec. 28, 2009, which issued as U.S. Pat. No. 8,865,151 on Oct. 21, 2014, which is a continuation-in-part of the International Application Serial No. PCT/US2008/068367 filed on Jun. 26, 2008, which claims benefit under 35 U.S.C. §119(e) of U.S. Provisional Application No. 60/937,198 filed Jun. 26, 2007 and U.S. Provisional Application No. 61/054,595 filed May 20, 2008, the contents of each of which are incorporated herein by reference in their entireties.
  • GOVERNMENT SUPPORT
  • This invention was made with Government support under Grant No.: W81XWH-05-1-0115 awarded by the Department of Defense. The Government has certain rights in the invention.
  • FIELD OF THE INVENTION
  • The present invention relates generally to compositions and methods for the treatment of angiogenic diseases and disorders.
  • BACKGROUND
  • Emerging data strongly implicates a ubiquitous eukaryotic enzyme “methionine aminopeptidase type 2” (MetAP-2) in many diseases (Kass 2007, Yeh 2005, Bo 2004, Bringardner 2008, Zhang 2001, Watanabe 2006). While MetAP-2's role is not yet fully understood, it is clear that expression and activity of MetAP-2, including, but not necessarily limited to over-expression or over-activity of MetAP-2, is correlated with disease progression and stage of disease (Salvukumar 2006).
  • It is a widely accepted hypothesis that tumor growth is angiogenesis-dependent, which is supported by both biological and pharmacological evidence and confirmed by genetic evidence which provides a scientific basis for current clinical trials of angiogenesis inhibitors. Increased tumor angiogenesis and elevated levels of proangiogenic factors such as vascular endothelial growth factor (VEGF/VPF), basic fibroblast growth factor (bFGF), and interleukin-8 (IL-8) correlate with decreased survival and increased risk of relapse in studies of patients with malignant solid tumors. The importance of angiogenesis is further supported by the observation that antiangiogenic agents inhibit tumor growth in a variety of animal models.
  • In the U.S. there are currently more than 30 angiogenesis inhibitors in various clinical trials for late-stage cancer. One of these angiogenesis inhibitors, O-(chloracetyl-carbamoyl)fumagillol (TNP-470), is a low molecular weight synthetic compound. Fumagillol is the alcohol derived from the hydrolysis of fumagillin3, a compound secreted by the fungus Aspergillus fumigatus fresenius. TNP-470 is a potent endothelial inhibitor in vitro30. Recently, TNP-470 has been tested as a potential new anticancer agent. TNP-470 is also referred to as AGM-1470, an analog of fumagillin, is among the most potent inhibitors of angiogenesis. The anti-angiogenic properties of the TNP-470 were tested on different tumor models in animals, and in clinical trials with a variety of malignancies such as prostate, breast, lung and cervical cancer.
  • In animal models, TNP-470 has the broadest anticancer spectrum of any known agent32, 31. TNP-470 inhibited the growth of murine tumors up to 91%, human tumors up to 100% and metastatic tumors up to 100% in mice (reviewed in ref. 31). In most studies, mice were treated at the same optimal dose of 30 mg/kg subcutaneously every other day. In clinical trials TNP-470 has shown evidence of antitumor activity when used as a single agent, with a number of objective responses reported with relapsed and refractory malignancies17,19,23. It has also shown promise when used in combination with conventional chemotherapy20,34, although many patients experience neurotoxicity (malaise, rare seizures, asthenia, anxiety and dysphoria)20,21,22, 23 at high chemotherapeutic doses where antitumor activity has been seen. In addition to its anti-angiogenic activities, TNP-470 also inhibits or prevents vascular leakage.
  • Methionine aminopeptidase-2 (MetAP-2) has been identified as the target of anti-angiogenic fumagillol derivatives (e.g., TNP-470).41 The crystal structure of fumagillin complexed with MetAP-2 was reported by Liu et al.42
  • TNP-470 is generally administered via injections, for example intramuscular or intravenous delivery systems or by a continuous intravenous infusion given every few days. TNP-470 is also typically administered at high doses for a specific period of time at infrequent time intervals, for example TNP-470 is administered at high doses as a chemotherapeutic. TNP-470 is not generally administered orally.
  • SUMMARY
  • The present invention relates, in part, to compositions and formulations comprising a MetAP-2 inhibitor and to compositions and formulations comprising a MetAP-2 inhibitor associated with a block copolymer, wherein the block copolymer comprises a hydrophobic and hydrophilic moiety. In some embodiments, the MetAP-2 inhibitor is associated with a block copolymer comprising a hydrophobic moiety, wherein the hydrophobic moiety is part of a block copolymer comprising hydrophilic and hydrophobic moieties. In some embodiments the block copolymer comprising hydrophilic and hydrophobic moieties forms a micelle.
  • The MetAP-2 inhibitor-comprising compositions and formulations can be used in the treatment of a disease or disorder involving or associated with MetAP-2 expression, including, but not necessarily limited to MetAP-2 overexpression or over-activity. Accordingly, the present invention relates to a method to treat a condition wherein the condition involves or requires MetAP-2 activity for its pathology, comprising administering a composition comprising a MetAP-2 inhibitor formulated as described herein.
  • One aspect of the present invention relates to a composition comprising a formulation of an irreversible MetAP-2 inhibitor, for example a fumagillol analog or derivative that has anti-proliferative activity, where the formulation comprises the MetAP-2 inhibitor associated with a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety. In one embodiment, the irreversible inhibitor is, for example, a member of the bengamide class of MetAP-2 inhibitors, where the formulation is associated with a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • Another aspect of the present invention relates to a composition comprising a formulation of reversible inhibitors, for example bestatin class inhibitors or 3-amino-2-hydroxyamides and related hydroxyamides and acylhydrazines, where the formulation is associated with a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • In some embodiments, the block copolymer is a diblock copolymer, for example, the fumagillin analogs/derivatives or bengamide analogs thereof are associated with the hydrophobic moiety of a block copolymer such as a diblock copolymer. In such instances, the fumagillin analog/derivative or bengamide analog is associated with the hydrophobic moiety of such diblock copolymer.
  • In some embodiments, the formulation comprises a micelle comprising the block copolymer associated with the MetAP-2 reversible or irreversible inhibitors.
  • The invention thus encompasses at least the following aspects and embodiments.
  • In one aspect, described herein is a composition comprising a methionine aminopeptidase-2 (MetAP-2) inhibitor formulation having anti-proliferative activity, wherein the formulation comprises a MetAP-2 inhibitor covalently linked to a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • In one embodiment of this and all other aspects described herein, the MetAP-2 inhibitor is a fumagillol derivative.
  • In another embodiment of this and all other aspects described herein, the MetAP-2 inhibitor is an irreversible MetAP-2 inhibitor.
  • In another embodiment of this and all other aspects described herein, MetAP-2 inhibitor is a reversible MetAP-2 inhibitor.
  • In another embodiment of this and all other aspects described herein, block copolymer is a diblock copolymer.
  • In another embodiment of this and all other aspects described herein, MetAP-2 inhibitor is covalently linked with the hydrophobic moiety of the block copolymer.
  • In another embodiment of this and all other aspects described herein, the hydrophobic polymer moiety of the block copolymer is selected from the group consisting of poly(d,L-lactic acid), poly(caprolactone) (PCL), and poly(propylene oxide).
  • In another embodiment of this and all other aspects described herein, the hydrophobic moiety is a poly(d,L-lactic acid) (PLA) polymer.
  • In another embodiment of this and all other aspects described herein, the hydrophobic polymer moiety is 1-15 kDa. For example, the hydrophobic polymer moiety can be between 1-10 kDa, 1-8 kDa, 1-5 kDa, 1-3 kDa, 3-15 kDa, 5-15 kDa, 8-15 kDa, 10-15 kDa, 12-15 kDa, 2-12 kDa, or 4-10 kDa, 6-8 kDa in size. In another embodiment of this and all other aspects described herein, the hydrophobic polymer is approximately 2 kDa.
  • In another embodiment of this and all other aspects described herein, the hydrophilic moiety is a poly(ethylene glycol) (PEG) polymer.
  • In another embodiment of this and all other aspects described herein, the hydrophilic polymer moiety is between 1-15 kDa. For example, a hydrophilic polymer moiety useful in the compositions as described herein can be between 1-10 kDa, 1-8 kDa, 1-5 kDa, 1-3 kDa, 3-15 kDa, 5-15 kDa, 8-15 kDa, 10-15 kDa, 12-15 kDa, 2-12 kDa, 4-10 kDa, 6-8 kDa in size. In another embodiment of this and all other aspects described herein, the hydrophilic polymer is approximately 2 kDa.
  • In another embodiment of this and all other aspects described herein, the polymer is a diblock copolymer comprising a PEG-PLA diblock copolymer having hydrophilic PEG and hydrophobic PLA moieties.
  • In another embodiment of this and all other aspects described herein, the formulation is formulated for oral administration.
  • In another embodiment of this and all other aspects described herein, the formulation is formulated for topical administration.
  • In another embodiment of this and all other aspects described herein, the formulation is formulated for administration by oral, IV, peritoneal, injected (e.g., subcutaneous, intramuscular, etc.), eyedrop or ocular, suppository, topical, pulmonary, including inhaled, and nasal routes, among others, and includes a reversible or irreversible MetAP-2 inhibitor.
  • In another embodiment of this and all other aspects described herein, the anti-proliferative activity is an anti-tumor activity or an activity which normalizes vascular permeability.
  • In another embodiment of this and all other aspects described herein, the MetAP-2 inhibitor has anti-angiogenic activity and is useful in the preparation of a medicament for the treatment of an angiogenesis-mediated condition and/or in a method of treating an angiogenesis-mediated condition.
  • In another embodiment of this and all aspects described herein relating to fumagillol derivatives having anti-proliferative and/or anti-angiogenic activity, the fumagillol derivative comprises a derivative selected from the group consisting of 6-O—(N-chloroacetylcarbamoyl)fumagillol (TNP-470), 6-O-(4-methoxyaniline)acetyl fumagillol; 6-O-(3,4,5-trimethexyaniline)acetyl fumagillol; 6-O-(4-(N,N-dimethylethoxy)aniline)acetyl fumagillol; 6-O-(cyclopropylamino)acetyl fumagillol; 6-O-(cyclobutylamino)acetyl fumagillol; 4-((cyclopropylamino)acetyl)oxy-2-(1,2-epoxy-1,5-dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-1 cyclohexanol; 4-((cyclobutylamino)acetyl)oxy-2-(1,2-epoxy-1,5 dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-1-cyclohexanol.
  • In another embodiment of this and all aspects described herein relating to fumagillol derivatives having anti-proliferative and/or anti-angiogenic activity, the fumagillol derivative comprises 6-O—(N-chloroacetylcarbamoyl)fumagillol (i.e., TNP-470).
  • In another embodiment of this and all aspects described herein, the MetAP-2 inhibitor is selected from the group consisting of a bengamide, a sulphonamide MetAP-2 inhibitor compound, a bestatin, a 3-amino-2-hydroxyamide MetAP-2 inhibitor compound, a hydroxyamide MetAP-2 inhibitor compound, an acylhydrazine MetAP-2 inhibitor compound, ovacillin, a reversible MetAP-2 inhibitor and an irreversible MetAP-2 inhibitor.
  • In another aspect, described herein is a method of treating a condition in which MetAP-2 activity is required for or involved in the pathology of the condition, the method comprising administering a MetAP-2 inhibitor formulation comprising a MetAP-2 inhibitor having anti-proliferative activity, covalently linked to a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • In one embodiment of this and all aspects described herein, the condition is an angiogenesis-mediated condition. Angiogenesis mediated conditions include, as non-limiting examples, cancer, metastatic tumors, ascites formation, psoriasis, age-related macular degeneration (AMD), thyroid hyperplasia, preeclampsia, rheumatoid arthritis and osteo-arthritis, Alzheimer's disease, obesity, pleural effusion, atherosclerosis, endometriosis, diabetic/other retinopathies, ocular neovascularizations such as neovascular glaucoma and corneal neovascularization, and IL-2 therapy associated edema and abnormal vascular proliferation of other types.
  • In one embodiment of this and all aspects described herein, the condition involving MetAP-2 activity comprises a tumor activity.
  • In one embodiment of this and all aspects described herein, the condition is selected from the group consisting of cancer, metastatic tumors, psoriasis, age-related macular degeneration (AMD), thyroid hyperplasia, preeclampsia, rheumatoid arthritis and osteo-arthritis, Alzheimer's disease, obesity, pleural effusion, atherosclerosis, endometriosis, diabetic/other retinopathies, ocular neovascularizations, IL-2 therapy associated edema and other edemas, malaria, SARS, HIV, herpes, lupus, IPF, COPD, asthma, cystic fibrosis, transplant rejection, allergic reaction, multiple sclerosis, bacterial infection, viral infection, and conditions involving or characterized by vascular hyperpermeability, inflammation, and spinal injury.
  • In another aspect, described herein is a method of making a diblock copolymer composition comprising a MetAP-2 inhibitor that has anti-proliferative activity, the method comprising conjugating the MetAP-2 inhibitor to a diblock copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • In one embodiment of this and all aspects described herein, the MetAP-2 inhibitor is a fumagillol derivative that has anti-proliferative and/or anti-angiogenic activity.
  • In another aspect, described herein is a diblock copolymer composition comprising a MetAP-2 inhibitor, the composition produced by the methods described herein. In one embodiment, the MetAP-2 inhibitor is a fumagillol derivative that has anti-proliferative activity and/or anti-angiogenic activity. In one embodiment, the fumagillol derivative comprises 6-O—(N-chloroacetylcarbamoyl)fumagillol (TNP-470) and its metabolites.
  • In another aspect, described herein is the use of any of the MetAP-2 inhibitor block copolymer formulations described herein in the manufacture of a medicament for the treatment of a condition that relies upon or involves MetAP-2 activity for its pathology.
  • In one embodiment of this and all aspects described herein, the condition comprises a tumor activity.
  • In another embodiment of this and all aspects described herein, the condition is selected from the group consisting of cancer, metastatic tumors, psoriasis, age-related macular degeneration (AMD), thyroid hyperplasia, preeclampsia, rheumatoid arthritis and osteo-arthritis, Alzheimer's disease, obesity, pleural effusion, atherosclerosis, endometriosis, diabetic/other retinopathies, ocular neovascularizations, IL-2 therapy associated edema and other edemas, malaria, SARS, HIV, herpes, lupus, IPF, COPD, asthma, cystic fibrosis, transplant rejection, allergic reaction, multiple sclerosis, bacterial infection, viral infection, fungal infection and conditions involving or characterized by abnormal vasculature or vascular hyperpermeability, inflammation and spinal injury.
  • In one embodiment of this and all aspects described herein, is a method of treating ocular neovascularization in a subject in need thereof, the method comprising administering a MetAP-2 inhibitor formulation comprising a MetAP-2 inhibitor having anti-angiogenic activity, associated with a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • In another embodiment of this and all aspects described herein, is a method of treating ocular neovascularization in a subject in need thereof, the method comprising administering a composition comprising a formulation of a fumagillol derivative that retains anti-angiogenic activity, the formulation comprising the fumagillol derivative is associated with a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety. Examples of ocular neovascularization include but are not limited to AMD, ARMD, proliferative diabetic retinopathy (PDR), and retinopathy of prematurity (ROP). Encompasseh herein are eye diseases which involves neovascularization, infection and edema.
  • In another aspect, described herein is a method of making a diblock copolymer micelle comprising a MetAP-2 inhibitor, the method comprising conjugating the MetAP-2 inhibitor to a diblock copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety and forming micelles of the resulting conjugate.
  • In another aspect, described herein is the production of a diblock copolymer micelle comprising a MetAP-2 inhibitor.
  • Other aspects of the invention are disclosed infra.
  • BRIEF DESCRIPTION OF FIGURES
  • FIG. 1 shows NMR spectra for a block copolymer and a block copolymer conjugate with TNP-470. Differences in NMR pattern verify the binding of the ethylendiamine and the TNP-470 to the polymer.
  • FIG. 2 shows amine levels of mPEG-PLA-en polymer before and after TNP-470 binding. TNBSA reaction was performed as described in the Examples; levels of 450 nm absorption indicate the levels of free amines.
  • FIG. 3 shows TNP-470 polymersomes' size ranges. A typical DLS graph showing diameter distribution of TNP-470 polymersomes.
  • FIG. 4A shows inhibition of human umbilical vein epithelial cell (HUVEC) proliferation by MetAP-2 inhibitor polymersomes. Different concentrations of TNP-470 micelles (comprising between 62.5-1000 nM of TNP-470 equivalent) showed a cytostatic effect on HUVECs as measured by WST-1 (*p<0.0005).
  • FIG. 4B shows that HUVEC proliferation was not affected by polymeric micelles without MetAP-2 inhibitor. No significant differences in proliferation were detected, even with a double concentration of carrier than that of the MetAP-2 inhibitor polymersomes.
  • FIG. 5A shows the effect of MetAP-2 inhibitor polymersomes on tumor volume. Tumor volume, measured during 14-18 days of daily oral administration of TNP-470 polymeric micelles to C57/BL mice (*p<0.05).
  • FIG. 5B shows the effect of TNP-470 polymersomes on mice weights. Mice were weighed q.o.d, the results are an average weight of mice in each group.
  • FIG. 6A shows a diagram of a conjugation reaction between TNP-470 and modified mPEG-PLA.
  • FIG. 6B TEM images of Lodamin dispersed in water, the spherical structure of the micelles are shown at different time points post incubation in water bar=10 nm.
  • FIG. 6C TNP-470 release from Lodamin during a 30 d period as determined by HPLC; Micelles were incubated in gastric fluid pH=1.2 (▪) or PBS pH=7.4 (□) and analysis of the released TNP-470 was done in duplicate. The results are presented as means±SD.
  • FIG. 7A shows flow cytometry analysis of uptake of Rhodamine labeled mPEG-PLA micelles by HUVEC (FL2high) at 0 min, 20 min and 24 h of incubation with micelle. Incubation after 2, 4 and 7 h showed a similar pattern as after 24 h of incubation (not shown).
  • FIG. 7B shows inhibition of HUVEC proliferation by different concentrations of Lodamin 50-1,000 nM TNP-470 equivalent. Empty polymeric micelles were also added as a control (n=8, *P<0.0005).
  • FIG. 7C shows the HUVEC growth curve () of cells treated q.o.d. with Lodamin (60 nM TNP-470 equivalent) (□) Untreated cells and (⋄) Vehicle treated cells.
  • FIG. 7D shows shows the quantification of neovascularization area in the cornea in a Corneal Micropocket assay (n=10, mean±SD). Inhibition of angiogenesis in Lodamin treated mouse (15 mg/kg q.d.) with respect to control.
  • FIG. 8A shows the fluorescent signal of tissue extracts and in serum of mice treated with Lodamin. Mice (n=3) were given a single dose of oral 6-coumarin labeled polymeric micelles (150 μl 30 mg/ml). Data show the values of the three different mice, autofluorescence was omitted by subtracting the fluorescent signal of tissue extracts from an untreated mouse. The percent of labeled cells was measured for each organ.
  • FIG. 8B shows the levels of fluorescent signals in mouse serum as measured after different time points post oral administration. The results are presented as the concentration of micelles calculated by standard calibration curve.
  • FIG. 8C shows the percentage of FL2high positive cells as isolated from the designated organs (ratio of numbers FL2high cells of treated tumors to those of control mouse).
  • FIG. 8D shows the FACS analysis graphs of single-cell suspensions from three representative organs (tumor, liver, brain) taken from a mouse bearing Lewis lung carcinoma after controlled enzymatic degradation. The FL2high represents those cells that contain the mPEG-PLA-rhodamine micelles.
  • FIG. 9A shows the effect of free or conjugated TNP-470 on established primary tumor-Lewis lung carcinoma tumors: effect of 30 mg/kg. q.o.d. of free TNP-470 (▪) given orally, compared to equivalent dose of Lodamin (x) or water (□), (n=5 mice per group, *P<0.05).
  • FIG. 9B shows the Lewis lung carcinoma volume during 18 d of different frequencies and doses of Lodamin: 30 mg/kg. q.o.d. (x), 15 mg/kg q.o.d. (◯), 15 mg/kg q.d. (▪), and water (□) by gavage (n=5 mice per group, *P<0.05.
  • FIG. 9C shows the effect of the vehicle, empty mPEG-PLA micelles, on Lewis lung carcinoma (n=5 mice per group, *P<0.05).
  • FIG. 9D shows the effect of Lodamin given at a dose of 15 mg/kg q d. () on B16/F10 murine melanoma tumor in C57B1/6J; water was given as control (□) (n=5 mice per group, *P<0.05).
  • FIG. 9E shows some representative Lewis lung carcinoma and B16/F10 tumors removed from mice at day 18 post treatment with oral Lodamin at 30 mg/kg q.o.d., and 15 mg/kg e.d. respectively, and from control untreated mice.
  • FIG. 9F shows the neurotoxcity of Lodamin on treated mice (10 d, 30 mg/kg q.o.d.) compared to mice treated with subcutaneous (30 mg/kg q.o.d.) free TNP-470 or water given by gavage. Balance beam test was quantified by foot-slip errors and the numbers of slips per meter are presented (n=4-5 mice per group). *P<0.05, **P<0.01, ***P<0.0001 (results are mean±SE).
  • FIG. 9G shows the mice's weight on day 1 and day 18 post treatment with oral polymeric micelle TNP-470 (Lodamin).
  • FIG. 10A shows the livers removed from Lodamin-treated or untreated mice with B16/F10 tumors, 20 d post cell injection. Control livers were enlarged with wide-spread macroscopic malignant nodules and extensive cirrhosis.
  • FIG. 10B shows the spleens removed from Lodamin-treated or untreated mice with B16/F10 tumors. Control spleens had large masses compared to treated mice with normal spleen morphology.
  • FIG. 10C shows the survival curve of treated versus control mice with B16/F10 tumors (n=7). Treatment started at day three (TX) after cell injection (arrow).
  • FIG. 11A shows the effect of Lodamin administered orally by gavage on established human glioblastoma tumors (U87-MG tumors) growth in nude mice: 15 mg/kg. q.d of Lodmain (solid diamonds) given orally, water (solid squares), (n=5-10 mice per group, *p<0.05).
  • FIG. 11B shows the effect of Lodamin given in drinking water at a dose of 15 mg/kg q.d. (solid squares), water with Lodamin was changed every 3 days. *p<0.05 (results are mean±SE). The mice had established human glioblastoma tumors (U87-MG tumors) growth.
  • FIG. 12 shows the Kaplan-Meier survival curve of C57/B1 mice treated with Lodamin given at a dose of 15 mg/kg q.d TNP-470 equivalent IP (n=9, white squares), oral (n=10, solid line) or without any treatment (n=10, diamond black).
  • FIG. 13A shows the effects of Lodamin on angiogenesis in Matrigel implants, the implants were removed 8 days post injection from mice.
  • FIG. 13B shows FACS analysis of the Matrigel incorporated cells in Lodamin-treated and non-treated mice, CD45-CD31+ represents the endothelial cells.
  • FIG. 14 shows schematic drawings of diblock-copolymer that is part hydrophilic and part hydrophobic conjugated to MetAP-2 inhibitors and diblock-copolymer micelles (polymersomes) formed.
  • FIG. 15 shows schematics of exemplary approaches for functionalizing MetAP-2 inhibitors and coupling copolymers to them.
  • FIG. 16A shows that Lodamin inhibits angiogenesis and the inflammatory response in the Matrigel plug angiogenesis assay and DTH in mice. Matrigel containing VEGF and bFGF were injected subcutaneously (s.c., n=5) to determine the effect of Lodamin on angiogenesis and macrophage infiltration. Upper panel shows representative plugs removed from Lodamin treated or untreated mice (bar=1 cm). Bottom panel shows vessel staining with anti-CD31 antibody and nuclei staining with Hematoxilin Gill's, bar=100 μm; arrows point to large vessel with open lumans.
  • FIG. 16B shows FACS dot plots of endothelial cells (CD31+/CD45) or macrophages (CD45+F4/80+) from Matrigel plugs (marked in the inner squares).
  • FIG. 16C shows the quantification of infiltrating endothelial cell or macrophages in Matrigel plugs. Data are presented as a percent of specific cell population out of the total cell population (n=3-4, p<0.05).
  • FIG. 16D shows that Lodamin suppressed ear swelling in DTH reactions elicited by oxazolone, photos of representative ears of Lodamin treated or untreated mice are circled. Bar=1 cm.
  • FIG. 16E shows the ear swelling is represented as change in thickness (in μm) compared to original ear thickness. Mice treated with Lodamin (white squares) showed a significant reduction of ear thickness compared to control mice (black squares). The differences were statistically significant from day 2 to day 12 post challenge, excluding day 5 (P<0.05) data are presented as an average±SEM, n=5.
  • FIG. 16F shows the immunohistological analysis of mice ears post DTH reaction: H&E staining and staining of CD-31 endothelial cell marker. Control ears exhibited an excessive inflammation and edema, including fibroblast proliferation and spongiosis pointed by arrows. Bar=50 μm.
  • FIG. 16G shows the representative Matrigel plugs exposed under mouse skin (bar=1 cm). Vehicle treated mice presented bloody plugs surrounded by massive blood vessels, compared to Lodamin treated mice which had poor vasculature. Bar=100 μm.
  • FIG. 16H shows immunohistological analysis of mice ears post DTH reaction: staining of macrophages using F4/80 marker. Control ears exhibited an excessive inflammation. Bar=50 μm. No obvious difference was found in macrophage distribution, however, the total number of macrophages present in control mice was elevated due to greater amount of tissue associated with increased swelling.
  • FIG. 17A shows the quantification of vessel area (mm2) in different corneal assays performed with Lodamin which were administered either orally (30 mg/kg q.d), by eye drops (30 mg/ml 1 gtt q.d) or by subconjunctional injections (30 mg/ml, q.d). Graphs show the significant inhibition of vessel formation after 5 d of treatment. Vessel area was reduced by 38% after subconjunctional injection (P=0.0002), by 30% and for eye drops (P=0.003), and by 37% (P=0.04) for oral administration. In each group n=10.
  • FIG. 17B shows representative images taken from the different groups at day 5, bFGF pellet is detected as a white spot in the center of the cornea, blood vessels growing from limbal periphery are reduced in Lodamin treated group compared to controls. Vessel area in mm2 is calculated by the following formula: η×clock hours×vessel length (mm)×0.2 mm.
  • FIG. 18A shows the CNV lesion size of Lodamin-treated mice compared to controls. 15 mg/kg/day was given for 7 or 14 d and dose of 30 mg/kg/day was given for 7 d. CNV lesions in choroidal flat mounts were evaluated after staining of blood vessels post blood vessel staining using isolectin-IB4 conjugated with Alexa® Fluor 488. Data are presented as a percent of blood vessels area in choroidal flat mounts (pixels) of treatment group out of controls. Data are expressed as mean±SD, (T-test, * P<0.05, ** P<0.005, n=10).
  • FIG. 18B shows H&E stained histological side sections of CNV site in Lodamin treated or untreated mice. Substantial differences in fibrous tissue thickness and choridal vessel invasion to CNV site are detected, Bars=50 p.m.
  • FIG. 18C shows the FACS analysis of single-cell suspension from retinas after 3 and 7 d of oral Lodamin treatment. Quantification of macrophage infiltration in mouse retinas originated from Lodamin treated (30 mg/kg, daily, oral) or mice which were treated with equivalent dose of vehicle. Macrophage population was detected as a double positive CD45+ and F4/80+ staining.
  • FIG. 19A shows the size of CNV lesions after intravitreal injections of Lodamin (100 μg Lodamin, 12.5 μg equivalent to TNP-470 or 300 μg Lodamin equivalent to 37.5 μg TNP-470). Data is presented as mean pixel number±SD (n=7-17, *P<0.05, t-test). Lower panels show representative images of retinal flat mounts stained with a lectin-FITC, Bars=10 μm.
  • FIG. 19B shows representative full-field flash ERG responses from the eyes of mice 14 days after intra-ocular injection of vehicle (left) or 300 m/eye Lodamin (right).
  • FIG. 19C shows representative responses to 8 Hz flicker.
  • FIG. 19D shows the 5th to 95th prediction limits for ERG parameters in vehicle treated rats are shown; the dashed line represents the normal mean of all the parameters. Symbols are parameter values of individual Lodamin-treated mice, and represent the same mouse in each column. Data are A Log Normal (eq. 3). Only the saturating amplitude of the rod photoresponse (RmP3) differed significantly after Lodamin.
  • FIG. 19E shows representative images of retinal tissue taken 14 d post intravitreal injection of Lodamin (300 μg/eye) and control naïve mouse retinas. (Bars=50 μm). No apparent retinal tissue changes were detected; both eyes presented normal structures of (1) choroid (2) photoreceptors (3) outer nuclear layer (4) outer plexiform layer (5) inner nuclear layer (6) inner plexiform layer and (7) ganglion cell layer. The total retinal thickness remained unchanged.
  • FIG. 20A shows the levels of VEGF and MCP-1 extracted from whole eye tissues from naïve mice, mice treated with Lodamin (30 mg/kg, oral every day for 7 d) or Vehicle (same equivalent amount of mPEG-PLA micelles). Data of ELISA assay are presented as a mean of pg/ml concentration±SD (n=6, * P<0.05, t-test).
  • FIG. 20B shows the ELISA quantification of secreted MCP-1 from RPE to the medium post 24 h incubation with Lodamin at different doses (0.1-100 nM TNP-470 equivalent).
  • FIG. 20C shows the Western blot protein analysis of samples of tissue lysis of retina or choroid of the different treatment groups: N=naïve mice, V=vehicle treated mice, L=Lodamin treated mice. Factors detected are TNFα (26 kDa), RAGE-two bands of 35 kDa and ˜50 kDa, HMGB-1 (29 kDa), NFidB (detected as two bands ˜65 kDa), MAPK or phosphorylated MAPK (ERK/p-ERK, bands of 42 kDa) and 3-actin (42 kDa). The samples were analyzed separately for choroids and retinas. (each sample is originated from 6 eyes).
  • FIG. 20D is a zymogram showing MMP-2 and MMP-9 activity in ocular tissue following oral Lodamin treatment of mice (30 mg/kg/day 7 d).
  • FIG. 20E shows the levels of RAGE receptor protein (50 kDa) and its ligand HMGB-1 (29 kDa) in HMVEC cells post TNP-470 treatment at different doses (0.1 nM-100 nM). A dose dependent effect on RAGE expression was detected.
  • FIG. 21A shows the quantification of Miles assays performed in mice after induction of vessel permeability using VEGF (50 ng) and MCP-1 (50 pg); extracted dye contents were quantified by measuring at 620 nm (Histograms). Data are expressed as mean±SD (n=10, *P<0.05, t-test). Also shown are representative photos of mouse skin showing diminished dye in mice treated with oral Lodamin 1 and 3 h before conducting the assay.
  • FIG. 21B shows that MCP-1 induces vessel permeability in a dose dependent manner. At 10 pg/ml, MCP-1 induced significant vessel leak compared with PBS.
  • FIG. 21C shows that Lodamin reduces whole eye vessel permeability as determined by Evans blue extraction method. Mouse eyes treated with Lodamin (3 h before conducting the assay) were 65% less than the control. Data are presented as mean±SD (n=5, *P<0.05, t-test).
  • DETAILED DESCRIPTION
  • The present invention relates in part to compositions and formulations comprising a MetAP-2 inhibitor associated with a block copolymer, wherein the block copolymer comprises a hydrophobic and hydrophilic moiety. Described herein are compositions comprising MetAP-2 inhibitors formulated for oral administration. In some embodiments, the composition comprises associating a MetAP-2 inhibitor with a block copolymer comprising a hydrophobic moiety, wherein the hydrophobic moiety is part of a block copolymer comprising a hydrophilic and a hydrophobic moiety. In some embodiments the block copolymer comprising hydrophilic and hydrophobic moiety forms a micelle.
  • Accordingly, the present invention relates to a method to treat a condition in which MetAP-2 activity is involved in or required for the pathology of the condition, the method comprising administering a composition comprising a MetAP-2 inhibitor formulated as disclosed herein. In one embodiment, the composition is formulated for oral administration, although other routes are specifically contemplated. In one embodiment, the condition is a proliferative condition and/or an angiogenesis-mediated condition, and the method comprises orally administering a composition comprising a MetAP-2 inhibitor formulated as disclosed herein. Other routes of administration are specifically contemplated, as discussed herein below.
  • Methionine aminopeptidases are essential for removal of the N-terminal methionine from nascent proteins. Following this removal, a variety of molecules are attached to the unstable, open N-terminus, including myristic acid. This acid changes the hydrophobicity of the nascent protein, and causes specific cellular localization and function. In some instances, this function may lead to pathological diseases, as with the oncogene Src.46 Recently, MetAP-2, one of the 2 major types of aminopeptidases, has itself been identified as a possible oncogene.47 Its importance in cell proliferation is well documented.45
  • As a result of this knowledge, numerous classes of therapeutics have been synthesized to target and inhibit MetAP-2 activity (including, but not necessarily limited to over-activity) including the fumagillin and ovacillin classes, bengamides, bestatins, pyrimidines, sulfonamides, among others (Bernier 2005). None have clinically succeeded, due partly to the non-specific nature of their bio-distribution and partly due to a poor PK (pharmacokinetic) profile, such as too rapid clearance, short half life, unacceptable levels of interaction with the nervous system, or crossing the blood-brain barrier. These issues are manifested in unwanted toxic side effects.
  • Many attempts have been made to overcome these issues associated with both the reversible and irreversible MetAP-2 classes of inhibitors largely through small molecule formulation, synthesis or high molecular weight (>60 kDa) polymeric conjugation. However, each formulation has shown insurmountable short-comings including too-short plasma half-life (a few minutes for the drug and less than 2 hr for the metabolites in the case of the fumagillin class compound TNP-470), weight loss with the sulfonamides and bestatins, unpredictable cardiovascular events with the bengamides, difficulty in manufacturing, questionable stability, the inability to dose patients over long periods of time, and lack of clearance of too slow clearance as with higher molecular weight polymer conjugated drugs. Again, the majority of these failures can be attributed to their non-specific biodistribution. It is therefore desirable to administer a MetAP-2 inhibitor to patients in doses and methods that are limited in overall toxicities, are stable, and can provide multiple routes of administration depending on the presentation of the disease.
  • Further, many MetAP-2 inhibitors, particularly those in the fumagillol class, are poorly soluble in water (1.9 mg/ml; see U.S. Pat. No. 5,536,623), and as such have low absorption and bioavailability if given orally. Therefore, modification of these MetAP-2 inhibitors is highly desirable from therapeutic, administration and safety perspectives.
  • In order to administer a MetAP-2 inhibitor one must overcome several issues associated with the chemical properties of the small molecule MetAP-2 inhibitors such as their poor solubility in water, low absorption, non-specific bio-distribution and poor bioavailability if administered orally, topically, inhaled, or given via eyedrop. Due to these properties, the vast majority of MetAP-2 inhibitors are currently formulated for administration exclusively via injections (i.e. intramuscularly and intravenously).
  • As demonstrated herein, the inventors have discovered a composition that formulates MetAP-2 inhibitors such as TNP-470, among others, for alternative routes of administration other than injections, by binding the MetAP-2 inhibitor to a diblock copolymer, for example a diblock co-polymer comprising PEG-PLA. The inventors have discovered that such conjugation of a MetAP-2 inhibitor with block copolymers (herein referred to as “block copolymer-MetAP-2 inhibitor conjugates”) is a highly suitable formulation for oral, topical, inhaled, eyedrop or peritoneal administration. While having advantages for oral or other non-injected routes of administration, is is also contemplated herein that the block copolymer compositions described herein can also be administered by injection (e.g., intramuscular, subcutaneous, etc.).
  • The inventors demonstrate that a MetAP-2 inhibitor was successfully conjugated to a modified PEG-PLA polymer through its amine, and formed nano-size polymersomes. The inventors also demonstrate, using images taken by TEM, that spherical micelles were formed, and size measurement with DLS showed a low range of size distribution around 10 nanometers. By using confocal microscopy images of fluorescently-labeled polymersomes, the inventors demonstrate rapid uptake by Human Umbilical Vein Endothelial Cells (HUVECs), and when the MetAP-2 inhibitor polymersomes were added, a significant inhibition of HUVEC proliferation was shown (as compared to no effect of the carrier itself).
  • In-vivo studies done on mice showed a significant inhibition of subcutaneous Lewis Lung Carcinoma tumors with C57/BL mice given a daily oral administration of TNP470 micelles. A dose of 15 mg/kg TNP-470 equivalent showed 63% inhibition without any weight loss to the mice.
  • Accordingly, the inventors have discovered that conjugating a MetAP-2 inhibitor such as TNP-470 to diblock copolymers is useful for formulation as an oral administration formulation and that such a MetAP-2 inhibitor-diblock copolymer conjugate is useful in a method to treat conditions that involve or require MetAP-2 activity for their pathologies, such as cancer, RA, viral infection, bacterial or fungal infection among others, and those associated with abnormal or hyperpermeable vasculature such as edema, age-related macular degeneration, diabetic retinopathy, inflammation or other conditions involving or requiring MetAP-2 activity as described herein or as known in the art.
  • One embodiment, for the first time, is an oral anti-prolierative/anti-angiogenic MetAP-2 inhibitor polymeric drug with potent anti-tumor and anti-metastatic efficacy named Lodamin. The inventors characterized its physicochemical properties and show that this polymeric pro-drug derived from TNP 470 successfully overcomes the drug limitations while retaining its anti-proliferative and antiangiogenic activities. Lodamin is produced by covalent conjugation of TNP 470 to a di-block copolymer: PEG PLA. The amphiphilic nature of this polymeric drug enables self-assembly of micelles in an aqueous medium26. In this structure, the TNP-470 is located in the core, where it is protected from the acidic environment of the stomach, thus enabling oral availability. Furthermore, advantage is taken of using biocompatible and well characterized polymers27,28.
  • Lodamin, administered orally, is effectively absorbed in the intestine and accumulates in tumor tissue. The drug significantly inhibits cell proliferation and angiogenesis, as demonstrated by inhibition of HUVEC proliferation, in the corneal micropocket assay, and in mouse tumor models. Lodamin significantly inhibited primary tumor growth as demonstrated in models of melanoma and lung cancer. Notably, oral Lodamin successfully prevented liver metastasis of melanoma tumor cells without causing liver-toxicity or other side-effects and led to prolonged mouse survival.
  • Moreover, unlike the neurological impairments caused by free TNP-470, Lodamin does not penetrate the blood-brain barrier, and accordingly did not cause neurotoxicity in mice. These results indicate that Lodamin is a good candidate for a safe maintenance drug with effective anti-tumor and anti-metastatic properties.
  • Accordingly, the inventors have discovered that conjugating MetAP-2 inhibitors to diblock copolymers is useful for formulation such as an oral administration formulation, among others, and that such a fumagillol derivative-diblock copolymer conjugate is useful in a method to treat proliferative disorders, including angiogenesis-mediated conditions and disorders such as cancer and AMD, among many others.
  • The inventors also showed that Lodamin, a polymeric conjugate of TNP-470, is a potent broad spectrum antiangiogenic ophthalmic drug. Lodamin significantly reduced angiogenesis, vascular leakage and inflammation in the murine models of choroidal neovascularization (CNV) and corneal neovascularization. Lodamin was found to suppress angiogenic and inflammatory signals including monocyte chemotactic protein-1 (MCP-1). Additionally, Lodamin reduced levels of the receptor for advanced glycation end products (RAGE) which likely contributes to its broad spectrum of activity (see Example 7, FIGS. 16-21).
  • Accordingly, described herein is a method of treating a condition involving or relying upon MetAP-2 activity for its pathology, the method comprising administering a MetAP-2 inhibitor formulation comprising a MetAP-2 inhibitor having anti-proliferative activity, covalently linked to a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety. In one embodiment, the condition is selected from the group consisting of cancer, metastatic tumors, psoriasis, age-related macular degeneration (AMD), thyroid hyperplasia, preeclampsia, rheumatoid arthritis and osteo-arthritis, Alzheimer's disease, obesity, pleural effusion, atherosclerosis, endometriosis, diabetic/other retinopathies, ocular neovascularizations, IL-2 therapy associated edema and other edemas, malaria, SARS, HIV, herpes, lupus, IPF, COPD, asthma, cystic fibrosis, transplant rejection, allergic reaction, multiple sclerosis, bacterial infection, viral infection, conditions involving or characterized by vascular hyperpermeability, inflammation, and spinal injury.
  • In one embodiment, described herein is a method of treating an angiogenesis-mediated condition, the method comprising orally administering a composition comprising a formulation of a fumagillol derivative that retains anti-angiogenic activity, the formulation comprising the fumagillol derivative is associated with a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety. In one embodiment, the angiogenesis-mediated condition is selected from a group comprising cancer, metastatic tumors, psoriasis, age-related macular degeneration (AMD), thyroid hyperplasia, preclampsia, rheumatoid arthritis and osteo-arthritis, Alzheimer's disease, obesity, pleural effusion, atherosclerosis, endometriosis, diabetic/other retinopathies, ocular neovascularizations, IL-2 therapy associated edema and other edemas.
  • In one embodiment, described herein is a method of treating ocular neovascularization in a subject in need thereof, the method comprising administering a MetAP-2 inhibitor formulation comprising a MetAP-2 inhibitor having anti-proliferative activity, covalently linked to a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety. In one embodiment, the MetAP-2 inhibitor is a fumagillol derivative. In some aspects, the MetAP-2 inhibitor formulation described herein can be applied to an eye disease or disorder which involves neovascularization, and/or edema.
  • In another embodiment, provided herein is a method of treating ocular neovascularization in a subject in need thereof, the method comprising administering a composition comprising a formulation of a fumagillol derivative that retains anti-angiogenic activity, wherein the formulation comprising the fumagillol derivative is associated with a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety. Examples of ocular neovascularization diseases or disorders include but are not limited to AMD, ARMD, proliferative diabetic retinopathy (PDR), and retinopathy of prematurity (ROP).
  • In one embodiment, provided herein is a composition comprising a methionine aminopeptidase-2 (MetAP-2) inhibitor formulation having anti-proliferative activity, wherein the formulation comprises a MetAP-2 inhibitor covalently linked to a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • In another embodiment, provided herein is a method of making a diblock copolymer composition comprising a MetAP-2 inhibitor that has anti-proliferative activity, the method comprising conjugating the MetAP-2 inhibitor to a diblock copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • In another embodiment, provided herein is a diblock copolymer composition comprising a MetAP-2 inhibitor, the composition produced by the methods described herein.
  • In another embodiment, provided herein is a composition comprising a formulation of a fumagillol derivative that has anti-angiogenic activity, the formulation comprising the derivative associated with a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
  • In one embodiment, provided herein is a method of making a diblock copolymer micelle comprising a fumagillol derivative that has anti-angiogenic activity, the method comprising conjugating the fumagillol derivative to a diblock copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety and forming micelles of the resulting conjugate.
  • In one embodiment of all aspects described herein, MetAP-2 inhibitor is an irreversible MetAP-2 inhibitor.
  • In one embodiment of all aspects described herein, MetAP-2 inhibitor is a reversible MetAP-2 inhibitor.
  • In one embodiment of this and all other aspects described herein, the block copolymer is a diblock copolymer.
  • In one embodiment of this and all other aspects described herein, the formulation comprises a micelle comprising the block copolymer associated with the fumagillol derivative.
  • In one embodiment of all aspects described herein, the fumagillol derivative thereof is associated with the hydrophobic moiety of the block copolymer.
  • In one embodiment of all aspects described herein, the hydrophobic polymer moiety of the block copolymer is selected from the group consisting of poly(d,L-lactic acid), poly(L-lysine), poly(aspartic acid), poly(caprolactone) (PCL), and poly(propylene oxide).
  • In one embodiment of all aspects described herein, the hydrophobic moiety is a poly(d,L-lactic acid) (PLA) polymer.
  • In one embodiment of all aspects described herein, MetAP-2 inhibitor is associated with the PLA moiety of the diblock copolymer.
  • In one embodiment of this and all other aspects described herein, the hydrophilic polymer moiety of the block copolymer is polyethylene glycol (PEG). In one embodiment of all aspects described herein, the PEG polymer is a capped PEG polymer.
  • In one embodiment of this and all other aspects described herein, the block copolymer is a diblock copolymer comprising a PEG-PLA diblock copolymer having hydrophilic PEG and hydrophobic PLA moieties.
  • In one embodiment of this and all other aspects described herein, the anti-angiogenic activity is an anti-tumor activity.
  • In one embodiment of this and all other aspects described herein, the fumagillol derivative comprises a derivative selected from the group consisting of 6-O—(N-chloroacetylcarbamoyl)fumagillol (TNP-470), 6-O-(4-methoxyaniline)acetyl fumagillol; 6-O-(3,4,5-trimethexyaniline)acetyl fumagillol; 6-O-(4-(N,N-dimethylethoxy)aniline)acetyl fumagillol; 6-O-(cyclopropylamino)acetyl fumagillol; 6-O-(cyclobutylamino)acetyl fumagillol; 4-((cyclopropylamino)acetyl)oxy-2-(1,2-epoxy-1,5 20 dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-1 cyclohexanol; and 4-((cyclobutylamino)acetyl)oxy-2-(1,2-epoxy-1,5 dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-1-cyclohexanol.
  • In one embodiment of this and all other aspects described herein, the fumagillol derivative comprises 6-O—(N-chloroacetylcarbamoyl)fumagillol (TNP-470).
  • In one embodiment of this and all other aspects described herein, the formulation comprises a diblock copolymer micelle formed with the diblock copolymer described herein.
  • In one embodiment of this and all other aspects described herein, the fumagillol derivative is associated with the PLA moiety of the diblock copolymer.
  • In one embodiment of this and all other aspects described herein, the composition or formulation is administered orally.
  • In one embodiment of this and all other aspects described herein, the composition or formulation is administered by intravitreous injection. In other embodiments, the composition or formulation is for topical administration, IV administration, peritoneal administration, injection, ocular administration, suppository administration, pulmonary administration or inhalation, and nasal administration.
  • In one embodiment, the composition is administered by intravitreous injection.
  • DEFINITIONS
  • The term “MetAP-2 inhibitor” refers to an agent that, at a minimum, inhibits the activity of MetAP-2 by at least 20% in a MetAP-2 assay as described in U.S. Pat. No. 6,548,477 or in U.S. Pat. No. 7,030,262 (which are both incorporated herein by reference), preferably at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or more, up to and including complete or 100% inhibition relative to the absence of such an agent. Reversible and irreversible MetAP-2 inhibitors are known in the art and are encompassed by the term “MetAP-2 inhibitor.” Fumagillol derivatives as described herein that have anti-proliferative activity are MetAP-2 inhibitors as the term is used herein. The fumagillol and ovacilin classes of MetAP-2 inhibitors have a reactive spiroepoxide moiety that reacts to form a covalent bond with the imidazole nitrogen of histidine 231 in the catalytic site of MetAP-2. Molecular modeling with potential irreversible MetAP-2 inhibitors will permit the skilled artisan to readily predict whether a given candidate inhibitor will react with histidine 231 of the enzyme and therefore have the desired inhibitory activity. Similarly, a rational design approach can be used to prepare reversible MetAP-2 inhibitors, as demonstrated by Wang et al.43
  • The term “fumagillol derivative” encompasses compounds represented by the Formula I below:
  • Figure US20160136292A1-20160519-C00001
  • wherein, X is —OH and Y is halogen, or X and Y are linked together to form a oxyrane ring, B represents —(C═O)— or —CH2-, R1 represents hydrogen, hydroxyl, —CN; NO2, —CF3; formyl; C1-C4 thioalkyl, acetamido; acetoxy; C1-C6 alkyl, C1-C4 aminoalkyl, C1-C4 alkylaminoalkyl; C1-C4 dialkylaminoalkyl; C1-C6 alkyloxy, C1-C6 aminoalkyloxy; C1-C4 alkylaminoalkoyy, C1-C4 dialkylaminoalkoxy, amino; C1-C6 alkylamino; C1-C4 dialkylamino; C1-C4 hyrdoalkyl; C1-C4 alkyloxycarboxylic acid etc. Fumagillol derivatives are disclosed in European Patent Application 0354787, 0357061, and 0415294 and Japanese patent application JP-A01-233275 and U.S. Pat. No. 5,290,807, which are incorporated herein in their entirety by reference. Other MetAP-2 inhibitory fumagillol derivatives, such as PPI-2458, are described by Bernier et al., Proc. Natl. Acad. Sci. U.S.A. 101: 10768-10773 (2004). Fumagillol derivatives as described herein have, at a minimum, anti-angiogenic activity when tested, for example, in a HUVEC proliferation assay as known in the art and described herein below. It should be understood that the term “fumagillol derivative” refers to the derivatives of fumagillol of Formula I, whereas a “fumagillol derivative block copolymer conjugate” refers to such derivatives conjugated to a block copolymer. Similarly, a “MetAP-2 inhibitor” refers to the inhibitor itself, while a “MetAP-2 inhibitor block copolymer conjugate” refers to the MetAP-2 inhibitor conjugated to a block copolymer.
  • As used herein, the term “proliferation” refers to the development of cells that result in unwanted or undesirable physiological consequences, such as with a tumor or inflammation or hyperpermeable, abnormal vasculature, viral infections, bacterial infections and fungal infections. Angiogenesis should be understood to be a proliferative process. Thus, in certain instances, the term “proliferation” can apply to the development of blood vessels. Such development is also referred to herein as “angiogenesis.” In some instances, the term “angiogenesis”, as used herein refers to the sprouting of new blood vessels from pre-existing blood vessels, characterized by endothelial cell proliferation and migration triggered by certain pathological conditions, such as the growth of tumors, metastasis, AMD and arthritis, among others. It should be noted that the term “proliferation” can also apply to the proliferation of viruses, bacteria, fungi, microsporidia, etc. The context of the term will make it clear which type of “proliferation” is being referred to.
  • The term “anti-proliferative activity” refers to the property of an agent that inhibits, suppresses or reduces the rate of growth or creation of new, undesired cells or blood vessels in the body in order to combat disease. A compound or agent with anti-proliferative activity as used herein is an agent that inhibits MetAP-2 and leads to the suppression of unwanted proliferating cells, e.g., in unwanted angiogenesis. Also encompassed by the term “anti-proliferative activity” is an agent that inhibits MetAP-2 and leads to cell death.
  • The term “anti-angiogenesis activity” as used herein refers to an agent which inhibits or suppresses or reduces the rate of growth or creation of new blood vessels in the body in order to combat disease. A compound or agent with anti-angiogenesis activity as used herein is an agent capable of inhibiting the formation of blood vessels or the formation of vasculature with abnormal or hyperpermeable properties. A disease associated with vascular permeability includes vascular complications of diabetes such as non-proliferative diabetic retinopathy and diabetic nephropathy, nephrotic syndrome, pulmonary hypertension or fibrosis, burn edema, tumor edema, brain tumor edema, IL-2 therapy-associated edema, and other edema-associated diseases, inflammatory disorders, complications from spinal injury, fibrotic disorders, and infections, including viral infections, bacterial infections and fungal infections.
  • As used herein, the term “retains anti-proliferative activity” means that a given compound derived from a MetAP-2 inhibitor has at least 20% of the anti-proliferative activity of that MetAP-2 inhibitor. Thus, a given fumagillol derivative will “retain anti-proliferative activity” if it retains at least 20% of the anti-proliferative activity of fumagillol.
  • As used herein, the term “retains anti-angiogenesis activity” means that a given fumagillol derivative has at least 20% of the anti-angiogenic activity of TNP-470 in a HUVEC assay of angiogenesis as described herein.
  • MetAP-2 inhibitor formulations as described herein can also be used to prevent the leakage of cell proliferation stimulators from blood vessels. That is, MetAP-2 inhibitors can reduce or prevent vascular hyperpermeability, and prevent the accumulation of stimulatory factors at sites of such permeability.
  • As used herein, the term “inhibit” or “inhibition” means the reduction or prevention of tumor growth and/or tumor metastasis in cancers. Inhibition includes slowing the rate of tumor growth and metastasis. The tumor growth rate can be reduced by about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 100%, about 125%, about 150% or more compared to a control, untreated tumor of the same type. Inhibition also means a reduction in the size of the tumor (including, but not limited to a reduction caused by cytotoxic activity) of at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, up to and including 100% (no tumor) when compared to a control, untreated tumor of the same type. The prevention of tumor growth and/or metastasis means no further increase in the size of the tumors from the time of start of treatment administration. Prevention also means status quo of no new metastatic tumors detected (ie. no further spread of cancer) and/or no increased amount of tumor markers detected by methods known in the art.
  • As used herein, the term “MetAP-2-mediated condition” refers to diseases or disorders in which MetAP-2 activity contributes to the pathology of the condition, whether it involves inappropriate cell proliferation, e.g., inappropriate angiogenesis, or, for example, vascular hyperpermeability resulting from the MetAP-2 activity. While MetAP-2 over-expression or over-activity can be involved, the expression or activity involved in a condition is not necessarily over-expression or over-activity—that is, normal levels of MetAP-2 activity can also contribute to pathological conditions and should be considered a target for therapy in such instances. Examples of conditions in which MetAP-2 activity is involved in or required for the pathology include, but are not limited to cancer, metastatic tumors, psoriasis, age-related macular degeneration (AMD), thyroid hyperplasia, preeclampsia, rheumatoid arthritis and osteo-arthritis, Alzheimer's disease, obesity, pleural effusion, atherosclerosis, endometriosis, diabetic/other retinopathies, ocular neovascularizations, IL-2 therapy associated edema and other edemas, malaria, SARS, HIV, herpes, lupus, IPF, COPD, asthma, cystic fibrosis, transplant rejection, allergic reaction, multiple sclerosis, bacterial infection, viral infection, and conditions involving or characterized by vascular hyperpermeability. Angiogenesis-mediated conditions are a sub-set of MetAP-2-mediated conditions, diseases or disorders that are the direct result of aberrant blood vessel proliferation (e.g. diabetic retinopathy and hemangiomas, among others known in the art and/or discussed herein).
  • As used herein, the term “angiogenesis-mediated condition” refers to diseases or disorders that are dependent on a rich blood supply and blood vessel proliferation for the disease pathological progression (eg. metastatic tumors) or diseases or disorders that are the direct result of aberrant blood vessel proliferation (e.g. diabetic retinopathy and hemangiomas). Non-limiting examples include abnormal vascular proliferation, neovascularization, hyperpermeability, ascites formation, psoriasis, age-related macular degeneration, retinopathy, thyroid hyperplasia, preeclampsia, rheumatoid arthritis and osteo-arthritis, Alzheimer's disease, obesity, pleural effusion, atherosclerosis, endometriosis, diabetic/other retinopathies, ocular neovascularizations such as neovascular glaucoma and corneal neovascularization.
  • As used herein, the term “tumor” means a mass of transformed cells that are characterized, at least in part, by containing angiogenic vasculature. The transformed cells are characterized by neoplastic uncontrolled cell multiplication which is rapid and continues even after the stimuli that initiated the new growth has ceased. The term “tumor” is used broadly to include the tumor parenchymal cells as well as the supporting stroma, including the angiogenic blood vessels that infiltrate the tumor parenchymal cell mass. Although a tumor generally is a malignant tumor, i.e., a cancer having the ability to metastasize (i.e. a metastatic tumor), a tumor also can be nonmalignant (i.e. non-metastatic tumor). Tumors are hallmarks of cancer, a neoplastic disease the natural course of which is fatal. Cancer cells exhibit the properties of invasion and metastasis and are highly anaplastic. Tumors are among the angiogenesis-mediated diseases encompassed by the therapeutic methods described herein.
  • As used herein, the term “tumor” is also used in reference to specific types of tumors, e.g., brain tumors including neuroblastoma, medulloblastoma, meningioma and glioblastoma; head and neck cancer, thyroid carcinoma, endocrine tumors, esophageal cancer, small cell and non-small cell lung cancer, colon cancer, rectal cancer, pancreatic cancer, gastric cancer, bladder cancer, hepatic cancer, malignant lymphoma, acute and chronic leukemia, Kaposi's sarcoma, glioma, hemangioma, osteosarcoma, soft tissue sarcoma, malignant melanoma, skin cancer, prostate cancer, breast carcinoma, choriocarcinoma, ovarian cancer, cervical cancer, uterine cancer and mesenchymal tumors, among others. In the context of the methods and compositions disclosed herein, and as highlighted by the inclusion of lymphomas and leukemias on the list above, it should also be understood that non-solid “tumors” can also benefit from the administration of MetAP-2 inhibitors formulated as described herein.
  • As used herein, the term “metastases” or “metastatic tumor” refers to a secondary tumor that grows separately elsewhere in the body from the primary tumor and has arisen from detached, transported cells, wherein the primary tumor is a solid tumor. The primary tumor, as used herein, refers to a tumor that originated in the location or organ in which it is present and did not metastasize to that location from another location. As used herein, a “malignant tumor” is one having the properties of invasion and metastasis and generally showing a high degree of anaplasia. Anaplasia is the reversion of cells to an immature or a less differentiated form, and it occurs in most malignant tumors.
  • The terms “polymersomes” and “polymeric micelles” are used interchangeably herein to refer to the same block copolymer compositions.
  • The term “copolymer” also known as “heteropolymer” as used herein refers to a polymer derived from two (or more) monomeric species, as opposed to a homopolymer where only one monomer is used. Copolymerization refers to methods used to chemically synthesize a copolymer.
  • The term “block copolymer” as used herein refers to the polymer comprising more than one subunit (or oligomer) type, wherein the copolymer comprises regions of a polymer comprising one subunit type adjoined to a polymer region comprising a second subunit type, for example the term block copolymer refers to a copolymer comprised of two or more homopolymer subunits linked by covalent bonds. The union of the homopolymer subunits may require an intermediate non-repeating subunit, known as a junction block. Block copolymers are made up of blocks of different polymerized monomers. Block copolymers are interesting because they can “microphase separate” to form periodic nanostructures. Block copolymers are described in further detail in the section “Copolymers” herein below.
  • The term “diblock copolymer” as used herein refers to a block copolymer with two distinct blocks. A block copolymer with three distinct blocks is called a triblock copolymers. It is also possible to have tetrablocks, multiblocks, etc.
  • The term “hydrophilic” as used herein refers to a molecule or portion of a molecule that is typically charge-polarized and capable of hydrogen bonding, enabling it to dissolve more readily in water than in oil or other hydrophobic solvents. Hydrophilic molecules are also known as polar molecules and are molecules that readily absorb moisture, are hygroscopic, and have strong polar groups that readily interact with water. A “hydrophilic” polymer as the term is used herein, has a solubility in water of at least 100 mg/ml at 25° C.
  • The term “hydrophobic” as used herein refers molecules tend to be non-polar and prefer other neutral molecules and non-polar solvents. Hydrophobic molecules in water often cluster together. Water on hydrophobic surfaces will exhibit a high contact angle. Examples of hydrophobic molecules include the alkanes, oils, fats, and greasy substances in general. Hydrophobic materials are used for oil removal from water, the management of oil spills, and chemical separation processes to remove non-polar from polar compounds. Hydrophobic molecules are also known as non-polar molecules. Hydrophobic molecules do not readily absorb water or are adversely affected by water, e.g., as a hydrophobic colloid. A “hydrophobic” polymer as the term is used herein has a solubility in water less than 10 mg/ml at 25° C., preferably less than 5 mg/ml, less than 1 mg/ml or lower.
  • The term “hydrophobic drug” as used herein refers to any organic or inorganic compound or substance having biological or pharmacological activity and adapted or used for a therapeutic purpose having a water solubility less than 10 mg/ml. MetAP-2 inhibitors, in particular fumagillol derivatives, tend to be hydrophobic drugs.
  • The term “micelle” as used herein refers to an arrangement of surfactant molecules (surfactants comprise a non-polar, lipophilic “tail” and a polar, hydrophilic “head”). As the term is used herein, a micelle has the arrangement in aqueous solution in which the non-polar tails face inward and the polar heads face outward. Micelles are typically colloid particles formed by an aggregation of small molecules and are usually microscopic particles suspended in some sort of liquid medium, e.g., water, and are between one nanometer and one micrometer in size. A typical micelle in aqueous solution forms an aggregate with the hydrophilic “head” regions in contact with surrounding solvent, sequestering the hydrophobic tail regions in the micelle center. This type of micelle is known as a normal phase micelle (oil-in-water micelle). Inverse micelles have the headgroups at the centre with the tails extending out (water-in-oil micelle). Micelles are approximately spherical in shape. Other phases, including shapes such as ellipsoids, cylinders, and bilayers are also possible. The shape and size of a micelle is a function of the molecular geometry of its surfactant molecules and solution conditions such as surfactant concentration, temperature, pH, and ionic strength. The process of forming micellae is known as micellization.
  • The term “therapeutically effective amount” refers to an amount that is sufficient to effect a therapeutically or prophylactically significant reduction or measurable suppression of a marker or symptom associated with a disease, condition or disorder dependent upon MetAP-2 activity for its pathology when that amount is administered to a typical subject who has such a condition. A therapeutically or prophylatically significant reduction in a marker or symptom is, e.g. about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, up to and including 100%, i.e., no symptoms or a marker at levels characteristic of non-diseased individuals, as compared to a control or non-treated subject. Alternatively, or in addition, a “therapeutically effective amount” can refer to an amount which directly or indirectly provides a reduction (i.e., at least a 10% reduction, preferably at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95% or more) in the expression or activity of MetAP-2. In some embodiments where the condition is, for example, cancer, the term “therapeutically effective amount” refers to the amount that is safe and sufficient to prevent or delay the development and further spread of metastases in cancer patients. The amount can also cure or cause the cancer to go into remission, slow the course of cancer progression, slow or inhibit tumor growth, slow or inhibit tumor metastasis, slow or inhibit the establishment of secondary tumors at metastatic sites, or inhibit the formation of new tumor metastasis.
  • The term “treat” or “treatment” refer to both therapeutic treatment and prophylactic or preventative measures (prevention is also understood to refer to a reduction in the likelihood of developing disease, e.g., at least a 20% reduced likelihood), wherein the object is to prevent or slow down the development or spread of disease. Beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable. “Treatment” can also mean prolonging survival as compared to expected survival if not receiving treatment. Those in need of treatment include those already diagnosed with disease. Where the disease is, e.g., cancer, those in need of treatment include those likely to develop metastases.
  • The terms “composition” or “pharmaceutical composition” are used interchangeably herein and refers to compositions or formulations that usually comprise an excipient, such as a pharmaceutically acceptable carrier that is conventional in the art and that is suitable for administration to mammals, and preferably humans or human cells. Such compositions can be specifically formulated for administration via one or more of a number of routes, including but not limited to oral, IV, peritoneal, injected (e.g., subcutaneous, intramuscular, etc.), eyedrop or ocular, suppository, topical, pulmonary, including inhaled, and nasal routes, among others.
  • The term “polymeric drug delivery composition” as used herein refers to the combination of drug, and block copolymer.
  • As used herein, the term “medicament” refers to an agent that promotes the recovery from and/or alleviates a symptom of a relevant condition.
  • The “pharmaceutically acceptable carrier” means a pharmaceutically acceptable means to mix and/or deliver the targeted delivery composition to a subject. The term “pharmaceutically acceptable carrier” as used herein means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material which solubilizes, stabilizes or otherwise provides a drug formulation with properties necessary to deliver the drug to an individual in a controlled format. A “carrier” can also be involved in carrying or transporting the subject agents from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and is compatible with administration to a subject, for example a human. A diblock copolymer as described herein is a pharmaceutically acceptable carrier as the term is used herein. Other pharmaceutically acceptable carriers can be used in combination with the block copolymer carriers as described herein. A pharmaceutically acceptable carrier does not promote the raising of an immune response to the drug.
  • The terms “polymer solution,” “aqueous solution” and the like, when used in reference to a block copolymer contained in such a solution, refer to water, i.e. aqueous, based composition having such block copolymer, or particularly to copolymer conjugates as described herein, such as a MetAP-2 inhibitor-PLA-PEG conjugate dissolved therein at a functional concentration. Polymer solution includes all free flowing forms of the composition comprising the conjugated copolymers as described herein and water. Polymer solutions act to solubilize the drug in a form that is acceptable for administration at physiologically relevant temperatures (temperatures <45° C.).
  • The term “aqueous solution” as used herein includes water without additives, or aqueous solutions containing additives or excipients such as pH buffers, components for tonicity adjustment, antioxidants, preservatives, drug stabilizers, etc., as commonly used in the preparation of pharmaceutical formulations.
  • The term “drug formulation” as used herein refers to all combinations of drug with polymer, for example polymer solutions that are mixed with drug to form drug solutions, as well as mixtures of undissolved polymer with drug, i.e. polymeric drug delivery compositions, that are subsequently dissolved into an aqueous environment to form a drug solution.
  • The term “administration” as used herein refers to the presentation of formulations to humans and animals in effective amounts, and includes all routes for dosing or administering drugs, whether self-administered or administered by medical practitioners. Oral administration is preferred.
  • The term “biodegradable” as used herein means the block copolymer can chemically break down or degrade within the body to form nontoxic components. The rate of degradation can be the same or different from the rate of drug release.
  • The term “PLA” as used herein refers to a polymer derived from the condensation of lactic acid or by the ring opening polymerization of lactide.
  • The term “biodegradable polyesters” as used herein refers to any biodegradable polyesters, which are preferably synthesized from monomers selected from the group consisting of D, L-lactide, D-lactide, L-lactide, D, L-lactic acid, D-lactic acid, L-lactic acid, glycolide, glycolic acid, ε-caprolactone, ε-hydroxy hexanoic acid, γ-butyrolactone, γ-hydroxy butyric acid, δ-valerolactone, δ-hydroxy valeric acid, hydroxybutyric acids, malic acid, and copolymers thereof.
  • As used herein, the term “patient” refers to a mammal, including a human, in need of the treatment to be administered.
  • The term “subject” and “individual” are used interchangeably herein, and refer to an animal, for example a human, to whom treatment, including prophylactic treatment, with a composition as described herein, is provided. The term “mammal” is intended to encompass a singular “mammal” and plural “mammals,” and includes, but is not limited: to humans, primates such as apes, monkeys, orangutans, and chimpanzees; canids such as dogs and wolves; felids such as cats, lions, and tigers; equids such as horses, donkeys, and zebras, food animals such as cows, pigs, and sheep; ungulates such as deer and giraffes; rodents such as mice, rats, hamsters and guinea pigs; and bears. Preferably, the mammal is a human subject. As used herein, a “subject” refers to a mammal, preferably a human. The term “individual”, “subject”, and “patient” are used interchangeably
  • In this specification and the appended claims, the singular forms “a,” “an,” and “the” include plural references unless the context clearly dictates otherwise. Thus, for example, reference to a composition for delivering “a drug” includes reference to two or more drugs. In describing and claiming the present invention, the following terminology will be used in accordance with the definitions set out below.
  • MetAP-2 Inhibitors
  • There are a number of MetAP-2 inhibitors known in the art, and MetAP-2 inhibitors of any kind are contemplated for formulation and use as described herein. That is, it is specifically contemplated herein that any of them can beneficially be prepared as block copolymer conjugates for delivery to a subject to treat a disease, disorder or condition related to, associated with or requiring MetAP-2 activity for its pathology. MetAP-2 inhibitors formulated as described herein retain anti-proliferative activity—that is, a MetAP-2 inhibitor formulated with a block copolymer conjugate as described herein will retain a meaningful portion of the anti-proliferative (including, but not limited to anti-angiogenic) activity of the inhibitor alone as measured in a MetAP-2 assay as described herein.
  • More specifically, however, MetAP-2 inhibitors including, but not limited to fumagillol derivatives, ovalicin, anthranilic acid sulfonamides (Wang et al., Proc. Natl. Acad. Sci. U.S.A. 105: 1838-1843 (2008)) e.g., A-800141, bengamides, bestatins (including, but not limited to reversible MetAP-2 inhibitors such as A-357300 and others described by Wang et al., Cancer Res. 63: 7861-7869), triazoles (see, e.g., compounds described in U.S. Pat. No. 7,303,082, and compounds described, e.g., by Garrabrant et al., Angiogenesis 7: 91-96 (2004)), 3-amino-2-hydroxyamides, hydroxyamides and acylhydrazines are contemplated for use in the formulations and methods described herein.
  • MetAP-2 inhibitors of various classes are described, for example, in U.S. Pat. Nos. 7,348,307, 7,268,111, 7,157,420, 7,105,482, 7,084,108, 7,037,890, 6,919,307, 6,548,477, 6,242,494, 6,288,228, 6,849,757, 6,887,863, 4,831,135, 7,122,345 and 7,030,262 and in U.S. published Patent Applications 20070254843, 20070161570, 20070117758, 20070010452, 20060223758, 20060069028, 20050239878, 20050059585, 20030109671, 20020193298, and 20020151493, the disclosures of each of which are incorporated herein by reference. Each of the MetAP-2 inhibitors described is potentially suitable for formulation as a MetAP-2 inhibitor block copolymer conjugate as described herein, for the treatment of MetAP-2 associated diseases, disorders or conditions.
  • Standard chemical approaches known to those of skill in the art can be used to conjugate a MetAP-2 inhibitor to block copolymer to generate a composition as described herein; the specific approach will depend upon the type of MetAP-2 inhibitor selected. For example, MetAP-2 inhibitors with alcohol functionality can be coupled to PEG-PLA using the same type of synthesis used to functionalize fumagillol (e.g., reaction with chloroacetylisocyanate, followed by reaction with a polymer bearing primary amine functionality). As an example, the compound A-357300 can be coupled in this manner after amine protection. See, e.g., FIG. 15. Similar coupling for can also be used, for example, to couple compounds such as (1-hydroxymethyl-2-methyl-propyl)-carbamic acid -(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methyl-but-2-enyl)-oxiranyl]-1-oxa-spiro[2.5]oct-6-yl ester (see U.S. Pat. No. 6,548,477). Additional fumagillin derivatives with alcohol functionalities permitting such conjugation are described, for example, in U.S. Pat. No. 7,087,768 (Han et al.)—see, e.g., Example 35 therein.
  • Assays for MetAP-2 activity are also known in the art. Assays are described, for example in U.S. Pat. Nos. 6,548,477 and 7,030,262. Any such assay can be used to evaluate the MetAP-2 inhibitory activity of a given compound or the activity of a given MetAP-2 inhibitor block copolymer conjugate as described herein. For the avoidance of doubt, however, a MetAP-2 inhibitor will demonstrate MetAP-2 inhibition (by at least 20%, preferably by at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, up to and including 100% (complete inhibition)) at a dosage or concentration that is substantially not toxic to the cells or subject to which it is to be administered, in either of the following two MetAP-2 assays.
  • Recombinant human MetAP-2 is expressed and purified from insect cells as described in Li and Chang, (1996) Biochem. Biophys. Res. Commun. 227:152-159. Various amounts of candidate MetAP-2 inhibitor compound are then added to buffer H (10 mM Hepes, pH 7.35, 100 mM KC 1, 10% glycerol, and 0.1 M Co.sup.2+) containing 1 nM purified recombinant human MetAP-2 and incubated at 37° C. for 30 minutes. To start the enzymatic reaction, a peptide containing a methionine residue, e.g., Met-Gly-Met, is added to the reaction mixture to a concentration of 1 mM. Released methionine is subsequently quantified at different time points (e.g., at 0, 2, 3, and 5 minutes) using the method of Zou et al. (1995) Mol. Gen Genetics 246:247-253). MetAP-2 inhibition is scored relative to a control lacking the candidate inhibitor, and can also be scored relative to a known MetAP-2 inhibitor, e.g. TNP-470 or, for example, TNP-470 block copolymer conjugate as described herein.
  • Assays for the inhibition of catalytic activity of MetAP-2 can be performed in 96-well microtiter plates. Compounds to be tested are dissolved in dimethyl sulfoxide at 10 mM and diluted ten-fold in assay buffer (50 mM HEPES, pH 7.4, 100 mM NaCl). Ten microliters of solution of each compound to be tested for inhibition are introduced into each cell of the plate. Zero inhibition of enzyme activity is taken to be the result obtained in cells in which 10 mL of assay buffer was placed. A mixture totaling 90 mL per well and made up of 84 mL of assay buffer containing 100 mM MnCl2, 1 mL of L-amino acid oxidase (Sigma Catalog No. A-9378, ˜11 mg/mL), 1 mL of horseradish peroxidase (Sigma Catalog No. P-8451, dissolved in assay buffer at a concentration of 10 mg/mL), 1 mL of the tripeptide Met-Ala-Ser (Bachem) dissolved in assay buffer at concentration of 50 mM, 1 mL of ortho-dianisidine (Sigma Catalog No. D-1954, freshly made solution in water at a concentration of 10 mg/mL), and MetAP-2 at a final concentration of 1.5 mg/mL are rapidly mixed and added to each cell containing test or control compound. The absorbence at 450 nanometers is measured every 20 seconds over a period of twenty minutes using an automatic plate reader (e.g., from Molecular Devices, California, USA). The Vmax in mOD/min, calculated for each well, is used to represent MetAP-2 activity. The IC50 for each candidate inhibitor is obtained by plotting the remaining activity versus inhibitor concentrations.
  • It is noted that some pro-drugs may require pre-treatment (e.g., enzymatic or non-specific hydrolysis) before assaying for MEtAP-2 inhibitory activity.
  • Fumagillol Derivatives
  • As one class of MetAP-2 inhibitors, derivatives of fumagillol are formulated for oral administration in compositions and methods described herein. It should be understood that anywhere in which the instant description refers to a MetAP-2 inhibitor, it can also be said to be referring to a fumagillol derivative having anti-proliferative and anti-angiogenic activity. Fumagillol derivatives useful in the compositions and methods described herein retain anti-proliferative, including, but not limited to anti-angiogenic, activity—i.e., at least 50% of the anti-proliferative activity of TNP-470, as measured in a HUVEC assay (see below). Numerous fumagillol derivatives meeting this criterion are known in the art. In one embodiment, suitable fumagillol derivatives for use in the compositions and formulations as disclosed herein are described in U.S. Pat. No. 5,290,807 which is incorporated herein in its entirety by reference.
  • In other embodiments, suitable fumagillol derivatives for use in the compositions and formulations as disclosed herein are representative of general Formula II as disclosed in U.S. Pat. Nos. 5,166,172; 5,290,807; 5,180,738 and 5,164,410, which are hereby incorporated by reference. In further embodiments, fumagillol derivatives for use in the compositions and formulations as disclosed herein are listed in International Patent Application WO03/027104 which is incorporated herein in its entirety by reference.
  • In one embodiment, a fumagillol derivative is 6-O—(N-chloroacetylcarbamoyl)fumagillol, also known as TNP-470, a derivative of the fumagillol of Formula III of International Patent Application WO03/027104 shown below.
  • Figure US20160136292A1-20160519-C00002
  • The structures of fumagillin and 6-O—(N-chloroacetylcarbamoyl)fumagillol (TNP-470) are shown below:
  • Figure US20160136292A1-20160519-C00003
  • The synthesis of TNP-470 is disclosed in U.S. Pat. Nos. 5,180,738 and 5,290,807 which are hereby incorporated herein in their entirety by reference.
  • Figure US20160136292A1-20160519-C00004
  • Fumagillin has been disclosed to be anti-proliferative and to have anti-angiogenic activity in U.S. Pat. No. 5,135,919 which is incorporated herein in its entirety by reference. Moreover, various fumagillol derivatives have been disclosed to be anti-proliferative and to have anti-angiogenic activity in U.S. Pat. Nos. 5,180,738; 5,164,410; 5,196, 406; 5,166,172; and 5,290,807 which are incorporated herein in their entirety by reference. Other MetAP-2 inhibitory fumagillol derivatives, such as PPI-2458, are described by Bernier et al., Proc. Natl. Acad. Sci. U.S.A. 101: 10768-10773 (2004), which is incorporated herein by reference. In particular, one fumagillol derivative (3R,4S,5S,6R)-5-methoxy-4-(2R,3R)-2-methyl-3-(3-methyl-2-butenyl)-oxiranyl)-1-oxaspiro(2,5)oct-6-yl(chloroacetyl) carbamate, also known as 6-O—(N-chloroacetylcarbamoyl)fumagillol or TNP-470 (available from Takeda Chemical Industries, Ltd. of Japan) is a particularly potent anti-proliferative and anti-angiogenic compound. Bhargava et al. review TNP-470 in Chapter 26 of Angiogenesis in Health and Disease, G. M. Rubanyi, ed., Marcel/Dekkker: 2000, pp. 387-406. One can determine if a fumagillol derivative has anti-proliferative activity using a proliferation assay as described herein. Similarly, where the proliferation involves angiogenesis, anti-angiogenic activity can be measured using an angiogenesis assay as shown in the Examples or as disclosed herein.
  • In some embodiments, fumagillol derivatives include, for example, but not limited to, O-(3,4-dimethoxycinnamoyl)fumagillol; O-(4-methoxycinnamoyl)fumagillol; O-(3,4,5-trimethoxycinnamoyl)fumagillol; O-(4-Chlorocinnamoyl)fumagillol; 4-(3,4,5-trimethoxycinnamoyl)oxy-2-(1,2-epoxy-1,5-dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-1-cyclohexanol; O-(4-trifluoromethylcinnamoyl)fumagillol; O-(4-nitrocinnamoyl)fumagillol; O-(3, 4-dimethoxy-6-nitrocinnamoyl)fumagillol; O-(4-acetoxycinnamoyl)nimagillol; O-(4-hydroxycinnamoyl)fumagillol; O-(4-acetoxy-3,5-dimethoxycinnamoyl)fumagillol; O-(3,5-dimethoxy-4-hydroxycinnamoyl)fumagillol; 4-(4-methoxycinnamoyl)oxy-2-(1,2-epoxy-1,5-dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-1-cyclohexanol; O-(4-dimethylaminocinnamoyl)fumagillol; O-(4-aminocinnamoyl)fumagillol; O-(4-cyanocinnamoyl)fumagillol; O-(3,4,5-trimethoxycinnamyl)fumagillol; O-(4-dimethylaminoethoxycinnamoyl)fumagillol; O-(3-dimethylaminomethyl-4-methoxycinnamoyl)fumagillol; O-(3,4-methylenedioxycinnamoyl)fumagillol; O-(3,4-dimethoxy-6-aminocinnamoyl)fumagillol; O-(4-ethylaminocinnamoyl)fumagillol; O-(4-ethylaminoethoxycinnamoyl)fumagillol; O-(4-dimethylaminocinnamyl)fumagillol; and 4-(4-dimethylaminocinnamoyl)oxy-2-(1,2-epoxy-1,5-dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-1-cyclohexanol.
  • In some embodiments, fumagillol derivatives include derivatives of the formula I, for example, but are not limited to: 1) 6-O-(4-methoxyaniline)acetyl fumagillol; 2) 6-O-(3,4,5-trimethoxyaniline)acetyl fumagillol; 3) 6-O-(2,4-dimethexyaniline)acetyl fumagillol; 4) 6-O-(3,4-dimethoxyaniline)acetyl fumagillol; 5) 6-O-(3,4-dimethoxy-6-nitroaniline)acetyl 5 fumagillol; 6) 6-O-(3,4-dimethexy-6-cyaneaniline)acetyl fumagillol; 7) 6-O-(4-allyloxyaniline)acetyl fumagillol; 8) 6-O-(4-(2-acetoxyethexy)aniline)acetyl fumagillol; 9) 6-O-(3-cyano-4-methoxyaniline)acetyl fumagillol; 10) 6-O-(3-(dimethylaminomethyl)-4-methexyaniline)acetyl fumagillol; 11) 6-O-(4-(2-methylpropoxyaniline)acetyl fumagillol; 12) 6-O-(3-isopropoxy-4-methoxyaniline)acetyl 15 fumagillol; 13) 6-O-(4-(N,N-dimethylethoxy)aniline)acetyl fumagillol; 14) 6-O-(3,5-diisopropyl-4-methoxyaniline)acetyl fumagillol; 15) 6-O-(3,5-dimethyl-4-methoxyaniline)acetyl fumagillol; 16) 6-O-(3-isopropyl-4-ethoxy-6-methylaniline)acetyl fumagillol; 17) 6-O-(4-propyloxyaniline)acetyl fumagillol; 18) 6-c-(aniline)acetyl fumagillol; 19) 6-O-(4-chloroaniline)acetyl fumagillol; 20) 6-O-(4-dimethylaminoaniline)acetyl fumagillol; 21) 6-O-(4-hydroxyaniline)acetyl fumagillol; 22) 6-O-(4-aminoaniline)acetyl fumagillol; 23) 6-O-(3,4-methylenedioxyaniline)acetyl fumagillol; 24) 6-O-(4-nitroaniline)acetyl fumagillol; 25) 6-O-(2,3,4-trimethoxy-6-aminoaniline)acetyl fumagillol; 26) 6-O-(4-acetoxy-3,5-dimethoxyaniline)acetyl fumagillol; 27) 6-O-(3,4-dimethoxy-5-hydroxyaniline)acetyl fumagillol; 28) 6-O-(4-dimethylaminoethoxyaniline)acetyl fumagillol; 29) 6-O-(4-ethylaminoaniline)acetyl fumagillol; 30) 6-O-(4-ethylaminoethoxyaniline)acetyl fumagillol; 31) 6-O-(3-dimethylaminomethyl-4 methoxyaniline)acetyl fumagillol; 32) 6-O-(4-trifluoromethylaniline)acetyl fumagillol; 33) 6-O-(4-acetoxy aniline)acetyl fumagillol; 34) 6-O-(4-cyanoaniline)acetyl fumagillol; 35) 6-O-(4-hydroxyethoxyaniline)acetyl fumagillol; 36) 6-O-(5-amino-2-methoxypyridine)acetyl fumagillol; 37) 6-O-(5-methoxypyrimidine-2-amino)acetyl fumagillol; 38) 6-o-(3-methoxy-6-aminopyridazine)acetyl fumagillol; 39) 4-((4-methoxyaniline)acetyl)oxy-2-(1,2-epoxy-1,5 dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-1 cyclohexanol; 40) 4-((3,4,5-trimethoxyaniline)acetyl)oxy-2-(1,2 5 epoxy-1,5-dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-1 cyclohexanol; 41) 6-O-(ethylamino)acetyl fumagillol; 42) 6-O-(isopropyl amino)acetyl fumagillol; 43) 6-O-(1-propyl amino)acetyl fumagillol; 44) 6-O-(1-butyl amino)acetyl fumagillol; 45) 6-O-(sec-butyl amino)acetyl fumagillol; 46) 6-O-(2-methyl-butylamino)acetyl fumagillol; 47) 6-O-(t-butyl amino)acetyl fumagillol; 48) 6-O-(pentyl amino)acetyl fumagillol; 49) 6-O-(1-methyl-butyl amino)acetyl fumagillol; 50) 6-O-(1-ethyl-propyl amino)acetyl-fumagilloli; 51) 6-O-(1-methyl-pentylamino)acetyl fumagillol; 52) 6-O-(1,2-dimethyl-butylamino)acetyl fumagillol; 53) 6-O-(1,2,2-trimethyl-propylamino)acetyl 20 fumagillol; 54) 6-0-(1-isopropyl-2-methylpropylamino)acetyl fumagillol; 55) 6-O-(3-methylbutylamino)acetyl fumagillol; 56) 6-O-(2-methylallylamino)acetyl fumagillol; 57) 6-O-(4-methyl-hepta-2,4-dienylamino)acetyl fumagillol; 58) 6-O-(1,5-dimethyl-4-hexenylamino)acetyl fumagillol; 59) 6-O-(1,1-dimethyl-2-propynylamino)acetyl fumagillol; 60) 6-O-(prop-2-enylamino)acetyl fumagillol; 61) 6-O-(2-bromo-ethylamino)acetyl fumagillol; 62) 6-O-(chloroethynylamino)acetyl fumagillol; 63) 6-O-(cyclopropylamino)acetyl fumagillol; 64) 6-O-(cyclobutylamino)acetyl fumagillol; 65) 6-0-(cyclopentylamino)acetyl fumagillol; 66) 6-O-(cyclohexylamino)acetyl fumagillol; 67) 6-O-(4-tert-butylcyclohexylamino)acetyl fumagillol; 68) 6-O-(2-dimethylamino-1-methylethylamino)acetyl 15 fumagillol; 69) 6-O-(2-dimethylamino-propylamino)acetyl fumagillol; 70) 6-O-(2-methexy-2-methyl-propylamino)acetyl fumagillol; 71) 6-O-(2-oxo-propylamine)acetyl fumagillol; 72) 6-O-(1,1-dimethyl-3-oxobutylamino)acetyl fumagillol; 73) 6-0-(ethyl-2-aminoacetate)acetyl fumagillol; 74) 6-O-(alanine-methylesteramino)acetyl fumagillol; 75) 6-O-(methyl-2-amino-3,3-dimethylbutanoate)acetyl fumagillol; 76) 6-O-(allylglycine-methylester)acetyl fumagillol; 77) 6-O-(2,2-dimethexy-ethylamino)acetyl fumagillol; 78) 4-((cyclopropylamino)acetyl)oxy-2-(1,2-epoxy 1,5-dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-15 cyclohexanol; 79) 4-((cyclobutylamino)acetyl)oxy-2-(1,2-epoxy-1,5-: dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-1-1 cyclohexanol; and 80) 6-O-(chloro)acetyl fumagillol.
  • Among the compounds of the Formula I, fumagillol derivatives useful in the methods and compositions as disclosed herein include, for example, but are not limited to 1) 6-O-(4-methoxyaniline)acetyl fumagillol; 2) 6-O-(3,4,5-trimethexyaniline)acetyl fumagillol; 3) 6-O-(4-(N,N-dimethylethoxy)aniline)acetyl fumagillol; 4) 6-O-(cyclopropylamino)acetyl fumagillol; 5) 6-O-(cyclobutylamino)acetyl fumagillol; 6) 4-((cyclopropylamino)acetyl)oxy-2-(1,2-epoxy-1,5 20 dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-1 cyclohexanol; and 7) 4-((cyclobutylamino)acetyl)oxy-2-(1,2-epoxy-1,5 dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-1-cyclohexanol. Structural formulas of the above compounds are shown in Tables in International Patent Application No: WO03/027104, which is incorporated herein by reference.
  • To enhance the activity of anti-proliferative or anti-angiogenic treatments, use of adjunct treatments with MetAP-2 inhibitors, including fumagillol derivatives, can be performed. Thus in one embodiment, use of the compositions as disclosed herein including a MetAP-2 inhibitor block copolymer conjugate in conjunction with other agents is encompassed in the present invention for treatment of diseases, disorders or conditions associated with, characterized by, or otherwise requiring MetAP-2 activity for their pathologies. These include, but are not limited to diseases involving abnormally stimulated neovascularization, such as inflammatory diseases (rheumatism and psoriasis, among others), diabetic retinopathy and cancer.
  • Copolymers
  • One aspect of the invention relates to the use of copolymers as carriers for the solubilization and formulation of a MetAP-2 inhibitor, for example TNP-470, as disclosed herein. A copolymer is a polymer comprising subunits of more than one type, i.e a copolymer can comprise subunits of A and subunits of B etc. In some embodiments, a copolymer is an alternating copolymer, for example, having a repeating structure comprising the different types of subunits, for example an alternating copolymer can have the formula: -A-B-A-B-A-B-A-B-A-B-, or -(-A-B-)n-.
  • In some embodiments, MetAP-2 inhibitors as disclosed herein are associated with a block copolymer to form a MetAP-2 inhibitor-block copolymer conjugate.
  • The polymers useful in the compositions and methods described herein are block copolymers. A block copolymer comprises subunits of more than one type, but instead of an alternating copolymer, a block copolymer comprises at a minimum, a block of subunits of one subunit type followed by a block of subunits of another subunit type. For example, a block copolymer comprising two subunits, for example A and B can have the formula of: -(A-A-)n-(B-B-)n- or -A-A-A-A-A-B-B-B-B-B-. The number of the different subunits can be different, for example; A-A-A-A-B-B-B-B-B. A diblock copolymer is a block copolymer comprising two different blocks of polymer subunits, whereas a triblock copolymer is a block copolymer comprising three different blocks of polymer subunits, and a tretrablock copolymer is a block copolymer comprising four different blocks of copolymer subunits, etc.
  • As used herein, a block copolymer is a polymer comprising at least a first block comprising a polymer of hydrophobic monomers, and at least a second block comprising a polymer comprised of hydrophilic monomers (see FIG. 14). A “block” copolymer differs from a non-block copolymer in that a block copolymer comprises blocks of polymer of one type (e.g. a block of hydrophilic monomers) that are joined to a block of polymer of another type (e.g. a block of hydrophobic monomers), as opposed to a non-block copolymer in which the different monomers are not joined together in blocks. For example, for a block copolymer of monomers A and B, the block copolymer would have, e.g., the structure AAAAABBBBB. A non-block copolymer of the same monomer subunits would have, for example, the structure ABAABABBABAABB or the structure ABABABABABAB, for example. The blocks of a block copolymer as the term is used herein will have at least 5 monomers per block (i.e., for a block copolymer of A and B monomers, the A block will be at least 5 A monomers long, and the B block will be at least 5 B monomers long). In some embodiments the homopolymer blocks will be at least 10 monomers long, 15 monomers long, 20 monomers long or more. A block copolymer as the term is used herein can have different block lengths (but each block will be at least 5 monomers long)—differences in block lengths can influence the ability of the block copolymer to form certain structures, e.g., micelles. In some instances, the monomers making up the hydrophilic block of a block copolymer can comprise different hydrophilic monomer subunits, and similarly, in some embodiments, monomers making up the hydrophobic block of a block copolymer can comprise different hydrophobic monomer subunits.
  • Diblock copolymers can be made using living polymerization techniques, such as atom transfer free radical polymerization (ATRP), reversible addition fragmentation chain transfer (RAFT), ring-opening metathesis polymerization (ROMP), and living cationic or living anionic polymerizations.
  • Block copolymers can “microphase separate” to form periodic nanostructures, for example, when one block is hydrophobic and the other hydrophilic. Microphase separation is a situation similar to that of oil and water. Oil and water don't mix together—they macrophase separate. If you have an “oil-like” first block and a “water-like” second block, the block copolymers undergo microphase separation. The blocks want to get as far from each other as possible, but they are covalently bonded, so they're not going to get very far. In “microphase separation” the “oil” and “water” blocks form nanometer-sized structures, including micelles, which comprise essentially spherical arrangement with the hydrophilic blocks arrange to the outside of the sphere, in contact with an aqueous solution and the hydrophobic blocks forming an inner hydrophobic core. Thermodynamic terms can describe how the different blocks interact. The interaction parameter, “chi” gives an indication of how different, chemically, the two blocks are and whether or not they will microphase separate. Generally, if the product of chi and the molecular weight is large (greater than 10.5), the blocks will microphase separate. Conversely, if the product of chi and the molecular weight is too small (less than 10.5), the different blocks are able to mix, rather than microphase separate.
  • In some embodiments, amphiphilic block copolymers are useful in the formulations and compositions of the present invention, for example as effective drug carriers that solubilize hydrophobic drugs into an aqueous environment. For example, amphiphilic block copolymers exhibiting self-association properties are disclosed in EP No. 0397 307 and EP0583955 and EP0552802, which are incorporated herein by reference.
  • In some embodiments, useful biodegradable polyesters comprised by the hydrophobic block of copolymers described herein are, for example, biodegradable polyester oligomers or polymers synthesized from monomers selected from, e.g., D, L-lactide, D-lactide, L-lactide, D, L-lactic acid, D-lactic acid, L-lactic acid, glycolide, glycolic acid, ε-caprolactone, ε-hydroxy hexanoic acid, γ-butyrolactone, γ-hydroxy butyric acid, δ-valerolactone, δ-hydroxy valeric acid, hydroxybutyric acids, malic acid, and copolymers thereof. More preferably, the biodegradable polyester is synthesized from monomers selected from the group consisting of D, L-lactide, D-lactide, L-lactide, D, L-lactic acid, D-lactic acid, L-lactic acid, glycolide, glycolic acid, ε-caprolactone, ε-hydroxy hexanoic acid, and copolymers thereof. Most preferably, the biodegradable polyester is synthesized from monomers selected from the group consisting of D, L-lactide, D-lactide, L-lactide, D, L-lactic acid, D-lactic acid, L-lactic acid, glycolide, glycolic acid, and copolymers thereof.
  • In some embodiments, the block copolymers comprise hydrophobic polyesters containing polyester bonds, for example but not limited to polylactic acid (PLA), polyglycolic acid (PGA), poly(D,L-lactic-co-glycolic acid)(PLGA), poly(caprolactone), poly(valerolactone), poly(hydroxybutyrate) and poly(hydroxyvalerate).
  • In some embodiments, the block copolymers comprise hydrophobic polymers, for example of the block copolymer is selected from the group consisting of poly(d,L-lactic acid), poly(L-lysine), poly(aspartic acid), poly(caprolactone) (PCL), poly(propylene oxide). In some embodiments, a hydrophobic polymer moiety is 1-15 kDa. For example, a hydrophobic polymer moiety is between 0.5-10 kDa, 1-8 kDa, 1-5 kDa, 1-3 kDa, 3-15 kDa, 5-15 kDa, 8-15 kDa, 10-15 kDa, 12-15 kDa, 2-12 kDa, 4-10 kDa, 6-8 kDa in size. In some embodiments, a hydrophobic polymer is approximately 2 kDa, for example 1.5 kDa, 2 kDa, or 2.5 kDa. In particular embodiments, a hydrophobic moiety useful in the composition as disclosed herein is a poly(d,L-lactic acid) (PLA) polymer, for example a poly(d,L-lactic acid) (PLA) polymer of 1kDa (1000 Da).
  • In some embodiments, the copolymers comprise hydrophilic polymers, for example but not limited to polyethene glycol (PEG) polymer (which is also referred to a poly(ethylene oxide) (PEO) or poly(oxyethelene) in the art). In some embodiments, the hydrophillic polymer may be capped at one end. In some embodiments, the capping group is alkoxy. For example, a hydrophilic polymer useful in the composition as disclosed by be capped with a methoxy group. In some embodiments, a hydrophilic polymer is between 1-15 kDa. For example, a hydrophilic polymer moiety useful in the composition as disclosed is between 1-10 kDa, 1-8 kDa, 1-5 kDa, 1-3 kDa, 3-15 kDa, 5-15 kDa, 8-15 kDa, 10-15 kDa, 12-15 kDa, 2-12 kDa, 4-10 kDa, 6-8 kDa in size. In some embodiments, a hydrophilic polymer is approximately 2 kDa, for example 1.5 kDa, 2 kDa, or 2.5 kDa. In particular embodiments, a hydrophilic polymer is a poly(ethylene glycol) (PEG) polymer, for example a poly(ethylene glycol) (PEG) polymer of 2 kDa (2000 Da).
  • In some embodiments, the copolymers comprise a diblock copolymer comprising a PEG-PLA diblock copolymer, where block copolymer comprises blocks of the hydrophilic PEG monomers and blocks of the hydrophobic monomer PLA.
  • The hydrophilic blocks of a copolymer can be coupled to the hydrophobic blocks by covalent bonds, for example by ester or urethane links and the like. Condensation polymerization and ring opening polymerization procedures may be utilized as may the coupling of a monofunctional hydrophilic block to either end of a difunctional hydrophobic block in the presence of coupling agents such as isocyanates. Furthermore, coupling reactions may follow activation of functional groups with activating agents, such as carbonyl diimidazole, succinic anhydride, N-hydroxy succinimide and p-nitrophenyl chloroformate and the like.
  • In some embodiments, hydrophilic blocks of a copolymer can comprise PEG or derivatized PEG monomers of an appropriate molecular weight. PEG has particularly favorable biocompatibility, nontoxic properties, hydrophilicity, solubilization properties, and rapid clearance from a patient's body.
  • The hydrophobic blocks of a copolymer should also comprise biodegradable polyester monomers that are biodegradable and biocompatible. The in vitro and in vivo degradation of hydrophobic, biodegradable polyester blocks of a copolymer are well understood and the degradation products are readily metabolized and/or eliminated from the patient's body.
  • MetAP-2 inhibitors, including fumagillol derivatives, such as TNP-470, can be solubilized or dispersed using block copolymers as disclosed herein. One advantage of using the block copolymers as disclosed herein is that the MetAP-2 inhibitors such as TNP-470 that have limited solubility or dispersibility in an aqueous or hydrophilic environment have enhanced solubility and/or dispersibility and can be administered via oral administration. Another advantage of using the block copolymer is improved biodistribution versus the bengamide class of MetAP-2 inhibitors, or the sulphonamide class, or the bestatin class of inhibitors.
  • One can prepare block copolymer, MetAP-2 conjugates as micelles as disclosed in the Examples herein. In another embodiment, one can prepare block copolymer micelles for sustained release of a MetAP-2 inhibitor using the method is disclosed in U.S. Pat. No. 6,623,729, which is incorporated in its entirety herein by reference and is further illustrated in the following steps:
  • Step 1: Preparation of Block Copolymer.
  • A block copolymer containing a hydrophobic part having hydroxyl group at one end and a hydrophilic part at the other end is prepared by copolymerization of a biodegradable polyester polymer and a polyethylene glycol (PEG) polymer in the presence of stannous octate as a catalyst: The copolymerization is performed at 160-200.quadrature for 2-6 hours under a vacuum condition. The polyester polymer includes polylactic acid (PLA), polyglycolic acid (PGA), poly(D,L-lactic-co-glycolic acid)(PLGA), poly(caprolactone), poly(valerolactone), poly(hydroxy butyrate) or poly(hydroxy valerate), and in some embodiments PLA and methoxypolyethyleneglycol is used as the polyethyleneglycol polymer.
  • Step 2: Binding of Linker by an Activation of Functional Groups of Block Copolymer.
  • The block copolymer is dissolved in an organic solvent and reacted with a linker at room temperature in the presence of pyridine and nitrogen: The organic solvent includes, but without limitation, methylenechloride, and the linker includes p-nitrophenyl chloroformate, carbonyldiimidazole (CDI), N,N′-disuccinimidyl carbonate (DSC), or a mixture of these compounds, preferably p-nitrophenyl chloroformate. The reaction is carried out for 2 to 6 hours, with a molar ratio of block copolymer:linker:pyridine ranging from 1:2:2 to 1:2:6.
  • Step 3: Preparation of a Conjugate of Drug and Biodegradable Polymer.
  • The linker-bound block copolymer is conjugated to a drug by covalent linkage to obtain a micelle monomer of a conjugate of drug and block copolymer, where the block copolymer obtained by reacting with hydrazine may be used: The block copolymer reacted with hydrazine forms a micelle monomer by binding the linker to a ketone group of a MetAP-2 inhibitor, such as a fumagillol derivative, while the block copolymer without hydrazine reaction forms a micelle monomer by binding the linker to an amine group of a MetAP-2 inhibitor, such as a fumagillol derivative. Preferably, the MetAP-2 inhibitor is TNP-470. In alternative embodiments, other MetAP-2 inhibitors are equally suitable for copolymer conjugation. In yet another embodiment, other drugs or chemotherapeutic agents are used in addition to a MetAP-2 inhibitor, for example anti-cancer agents such as, but without limitation, doxorubicin, adriamycin, cisplatin, taxol and 5-fluorouracil.
  • Step 4: Preparation of Sustained Release Micelle.
  • The micelle monomers prepared in Step 3 are dispersed in an aqueous solution to prepare sustained release micelles. When micelle monomers are dispersed in a certain concentration, micelles are formed spontaneously by thermodynamic equilibrium. Sustained release micelles thus prepared release a drug by way of hydrolysis and enzymatic action in vivo, and the released drug exerts the same effect as free drug does.
  • Conjugation of MetAP-2 Inhibitors with Copolymers
  • In the composition and methods disclosed herein, a MetAP-2 inhibitor, for example TNP-470, is associated with a block copolymer.
  • As used herein, the term “associated with” means that one entity is in physical association or contact with another. Thus, a MetAP-2 inhibitor “associated with” a block copolymer can be either covalently or non-covalently joined to the block copolymer. It is preferred that the association be covalent. The association can be mediated by a linker moiety, particularly where the association is covalent. The term “association” or “interaction” or “associated with” are used interchangeably herein and as used in reference to the association or interaction of a MetAP-2 inhibitor, e.g., TNP-470 with a block copolymer, refers to any association between the MetAP-2 inhibitor and the block copolymer, for example a diblock copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety, either by a direct linkage or an indirect linkage.
  • An indirect linkage includes an association between a MetAP-2 inhibitor and the block copolymer, wherein the MetAP-2 inhibitor and block copolymer are attached via a linker moiety, e.g., they are not directly linked. Linker moieties include, but are not limited to, chemical linker moieties. In some embodiments, a linker between the MetAP-2 inhibitor and the copolymer is formed by reacting the polymer and a linker selected e.g., from the group consisting of p-nitrophenyl chloroformate, carbonyldiimidazole (CDI), N,N′-disuccinimidyl carbonate (DSC), cis-aconitic anhydride, and a mixture of these compounds.
  • A direct linkage includes any linkage wherein a linker moiety is not required. In one embodiment, a direct linkage includes a chemical or a physical interaction wherein the two moieties, i.e. the a MetAP-2 inhibitor and the block copolymer interact such that they are attracted to each other. Examples of direct interactions include covalent interactions, non-covalent interactions, hydrophobic/hydrophilic, ionic (e.g., electrostatic, coulombic attraction, ion-dipole, charge-transfer), Van der Waals, or hydrogen bonding, and chemical bonding, including the formation of a covalent bond. Accordingly, in one embodiment, a a MetAP-2 inhibitor, such as TNP-470 and the block copolymer are not linked via a linker, e.g., they are directly linked. In a further embodiment, a MetAP-2 inhibitor and the block copolymer are electrostatically associated with each other.
  • In one embodiment, the linker is a peptide linker. In another embodiment, the peptide linker is enzymatically cleavable, e.g., by a protease enzyme that cleaves the linkage to release the MetAP-2 inhibitor. Most preferably, the peptide linkage is capable of being cleaved by preselected cellular enzymes, for instance, those found in lysosomes of cancerous cells or proliferating endothelial cells. Alternatively, an acid hydrolysable linker could comprise an ester or amide linkage and be for instance, a cis-aconityl linkage. A pH sensitive linker can also be used. Cleavage of the linker of the conjugate results in release of active MetAP-2 inhibitor. Thus the MetAP-2 inhibitor must be conjugated with the polymer in a way that does not alter the activity of the agent. The linker preferably comprises at least one cleavable peptide bond. Preferably the linker is an enzyme cleavable oligopeptide group preferably comprising sufficient amino acid units to allow specific binding and cleavage by a selected cellular enzyme. Preferably the linker is at least two amino acids long, more preferably at least three amino acids long. Cleavable linkers are described, e.g., in U.S. Pat. No. 7,332,523, which is incorporated herein by reference. An example of a preferred peptide linker is Gly-Phe-Leu-Gly (see U.S. Pat. No. 7,332,523). Other linker are known to those of skill in the art.
  • Pharmaceutical Compositions and Administration:
  • The MetAP-2 inhibitor in the formulations and compositions as disclosed herein is particularly useful in methods of treating conditions in which MetAP-2 activity is involved in or required for the pathology of the condition, including, but not necessarily limited to conditions in which MetAp-2 is either over-expressed or over-active in a mammal, for example a human. Such conditions, herein also referred to as a “MetAP-2-dependent disease or disorder” are selected from a group including, but not necessarily limited to cancer, ascites formation, psoriasis, age-related macular degeneration, thyroid hyperplasia, preeclampsia, rheumatoid arthritis and osteoarthritis, Alzheimer's disease, obesity, pleural effusion, atherosclerosis, endometriosis, diabetic/other retinopathies, neovascular glaucoma, age-related macular degeneration, hemangiomas, and corneal neovascularization, HIV, HPV, herpes and other viral infections, anthrax, IPF, COPD, multiple sclerosis, other sclerotic diseases, transplant rejection, lupus, asthma and other conditions described herein as associated with MetAP-2 activity.
  • In one embodiment, a MetAP-2-dependent disease or disorder is cancer, where the cells are rapidly dividing neoplastic cancer cells, and where the neoplastic cells require an efficient blood supply to maintain continued growth of the tumor. As used herein, cancer refers to any of various malignant neoplasms characterized by the proliferation of anaplastic cells that tend to invade surrounding tissue and metastasize to new body sites and also refers to the pathological condition characterized by such malignant neoplastic growths. The blood vessels provide conduits to metastasize and spread elsewhere in the body. Upon arrival at the metastatic site, the cancer cells then work on establishing a new blood supply network. Administration of a MetAP-2 inhibitor in the formulations and compositions as disclosed herein is useful to inhibit proliferation at the primary disease site, and in the case of cancer, at secondary tumor sites; embodiments of the invention serve to prevent and limit the progression of the disease. Any disease that requires a continuous proliferation of cells, either primary cells, endothelial cells, adjacent cells or others in order to perpetuate the disease is a candidate target. For example, candidates for the treatment of cancer as described herein include, but are not limited to carcinomas and sarcomas found in the anus, bladder, bile duct, bone, brain, breast, cervix, colon/rectum, endometrium, esophagus, eye, gallbladder, head and neck, liver, kidney, larynx, lung, mediastinum (chest), mouth, ovaries, pancreas, penis, prostate, skin, small intestine, stomach, spinal marrow, tailbone, testicles, thyroid and uterus. The types of carcinomas include papilloma/carcinoma, choriocarcinoma, endodermal sinus tumor, teratoma, adenoma/adenocarcinoma, melanoma, fibroma, lipoma, leiomyoma, rhabdomyoma, mesothelioma, angioma, osteoma, chondroma, glioma, lymphoma/leukemia, squamous cell carcinoma, small cell carcinoma, large cell undifferentiated carcinomas, basal cell carcinoma and sinonasal undifferentiated carcinoma. The types of sarcomas include soft tissue sarcoma such as alveolar soft part sarcoma, angiosarcoma, dermatofibrosarcoma, desmoid tumor, desmoplastic small round cell tumor, extraskeletal chondrosarcoma, extraskeletal osteosarcoma, fibrosarcoma, hemangiopericytoma, hemangiosarcoma, Kaposi's sarcoma, leiomyosarcoma, liposarcoma, lymphangiosarcoma, lymphosarcoma, malignant fibrous histiocytoma, neurofibrosarcoma, rhabdomyosarcoma, synovial sarcoma, and Askin's tumor, Ewing's sarcoma (primitive neuroectodermal tumor), malignant hemangioendothelioma, malignant schwannoma, osteosarcoma, and chondrosarcoma. Abnormal build up and growth of blood vessels in the skin or internal organs in the form of hemangiomas can also be treated according to the methods described herein.
  • In one embodiment, a MetAP-2-dependent disease or disorder is age-related macular degeneration. It is known that VEGF contributes to abnormal blood vessel growth from the choroid layer of the eye into the retina, similar to what occurs during the wet or neovascular form of age-related macular degeneration. Macular degeneration, often called AMD or ARMD (age-related macular degeneration), is the leading cause of vision loss and blindness in Americans aged 65 and older. In order for new blood vessels to grow (neovascularization) beneath the retina and leak blood and fluid, endothelial cells must proliferate. This proliferation is uneven, and results in the leakage described above, which causes permanent damage to light-sensitive retinal cells, which die off and create blind spots in central vision or the macula.
  • In one embodiment, a MetAP-2-dependent disease or disorder is diabetic retinopathy-abnormal blood vessel growth associated with diabetic eye diseases. In diabetic retinopathy and retinopathy of prematurity (ROP) VEGF is released which promotes blood vessel formation—thus, the disease or disorder is an angiogenic disease or disorder. Released by the retina (light-sensitive tissue in back of the eye) when normal blood vessels are damaged by tiny blood clots due to diabetes, VEGF turns on its receptor, igniting a chain reaction that culminates in new blood vessel growth. However, the backup blood vessels, having proliferated too quickly and unevenly, are faulty, so they leak, bleed and encourage scar tissue that detaches the retina, resulting in severe loss of vision. Such growth is the hallmark of diabetic retinopathy, the leading cause of blindness among young people in developed countries. In one embodiment, the subject in need of treatment can be a mammal, such as a dog or a cat, preferably a human.
  • In one embodiment, a MetAP-2-dependent disease or disorder is rheumatoid arthritis.44 Rheumatoid arthritis (RA) is characterized by synovial tissue swelling, leukocyte ingress and new blood vessel growth from existing vessels. The disease is thought to occur as an immunological response to an as yet unidentified antigen. The expansion of the synovial lining of joints in rheumatoid arthritis (RA) and the subsequent invasion by the pannus of underlying cartilage and bone necessitate an increase in the vascular supply to the synovium, to cope with the increased requirement for oxygen and nutrients. Endothelial cell proliferation is now recognized as a key event in the formation and maintenance of the pannus in RA (Paleolog, E. M., 2002). Thus, RA is an angiogenesis-related disease or disorder. Even in early RA, some of the earliest histological observations are blood vessels. A mononuclear infiltrate characterizes the synovial tissue along with a luxuriant vasculature. Endothelial cell proliferation is integral to formation of the inflammatory pannus and without it, leukocyte ingress could not occur (Koch, A. E., 2000). Disruption of the formation of new blood vessels would not only prevent delivery of nutrients to the inflammatory site, it could also reduce joint swelling due to the additional activity of VEGF, a potent proangiogenic factor in RA, as a vascular permeability factor.
  • In one embodiment, a MetAP-2-dependent disease or disorder is Alzheimer's disease. Alzheimer's disease (AD) is the most common cause of dementia worldwide. AD is characterized by an excessive cerebral amyloid deposition leading to degeneration of neurons and eventually to dementia. The exact cause of AD is still unknown. It has been shown by epidemiological studies that long-term use of non-steroidal anti-inflammatory drugs, statins, histamine H2-receptor blockers, or calcium-channel blockers, all of which are cardiovascular drugs with an anti-proliferative effects, seem to prevent Alzheimer's disease and/or influence the outcome of AD patients. Therefore, it has been speculated that in AD endothelial cell proliferation in the brain vasculature may play an important role in AD, that is, AD can be an angiogenesis-related or -mediated disease. In Alzheimer's disease, the brain endothelium secretes the precursor substrate for the beta-amyloid plaque and a neurotoxic peptide that selectively kills cortical neurons. Moreover amyloid deposition in the vasculature leads to endothelial cell apoptosis and endothelial cell activation which leads to neovascularization. Vessel formation could be blocked by the VEGF antagonist SU 4312 as well as by statins, indicating that anti-proliferative or anti-angiogenic strategies can interfere with endothelial cell activation in AD (Schultheiss C., el. al., 2006; Grammas P., et. al., 1999) and can be used for preventing and/or treating AD.
  • In one embodiment, a MetAP-2-dependent disease or disorder is obesity. It has been shown that the MetAP-2 inhibitor TNP-470 was able to prevent diet-induced and genetic obesity in mice (Ebba Bråkenhielm et. al., Circulation Research, 2004; 94:1579). TNP-470 reduced vascularity in the adipose tissue, thereby inhibiting the rate of growth of the adipose tissue and obesity development. Obesity is thus an angiogenesis-related or -mediated disease or disorder.
  • In one embodiment, a MetAP-2-dependent disease or disorder is endometriosis. Excessive endometrial angiogenesis is proposed as an important mechanism in the pathogenesis of endometriosis (Healy, D L., et. al., 1998). The endometrium of patients with endometriosis shows enhanced endothelial cell proliferation. Moreover there is an elevated expression of the cell adhesion molecule integrin vβ3 in more blood vessels in the endometrium of women with endometriosis when compared with normal women. Strategies that inhibit endothelial cell proliferation can be used to treat endometriosis.
  • In one embodiment, the method of treating the MetAP-2-dependent disease or disorder is applicable to the treatment of cancer; the method as disclosed herein is applicable to all carcinomas, blood-borne cancers and sarcomas. Preferably, the cancer is selected from the group consisting of papilloma/carcinoma, choriocarcinoma, endodermal sinus tumor, teratoma, adenoma/adenocarcinoma, melanoma, fibroma, lipoma, leiomyoma, rhabdomyoma, mesothelioma, angioma, osteoma, chondroma, glioma, lymphoma/leukemia, squamous cell carcinoma, small cell carcinoma, large cell undifferentiated carcinomas, basal cell carcinoma, sinonasal undifferentiated carcinoma, soft tissue sarcoma such as alveolar soft part sarcoma, angiosarcoma, dermatofibrosarcoma, desmoid tumor, desmoplastic small round cell tumor, extraskeletal chondrosarcoma, extraskeletal osteosarcoma, fibrosarcoma, hemangiopericytoma, hemangiosarcoma, Kaposi's sarcoma, leiomyosarcoma, liposarcoma, lymphangiosarcoma, lymphosarcoma, malignant fibrous histiocytoma, neurofibrosarcoma, rhabdomyosarcoma, synovial sarcoma, and Askin's tumor, Ewing's sarcoma (primitive neuroectodermal tumor), malignant hemangioendothelioma, malignant schwannoma, osteosarcoma, and chondrosarcoma, that are found in the anus, bladder, bile duct, bone, brain, breast, cervix, colon/rectum, endometrium, esophagus, eye, gallbladder, head and neck, liver, kidney, larynx, lung, mediastinum (chest), mouth, ovaries, pancreas, penis, prostate, skin, small intestine, stomach, spinal marrow, tailbone, testicles, thyroid and uterus.
  • The MetAP-2 inhibitor in the formulations and compositions as disclosed herein is particularly useful in methods of inhibiting cell proliferation, including angiogenesis at a site of tumorigenesis in a mammal.45 The MetAP-2 inhibitor in the formulations and compositions as disclosed herein administered at such sites and in such varied ways prevents or inhibits endothelial cell proliferation and blood vessel formation at the site thereby inhibiting the development and growth of the tumor. Tumors which may be prevented or inhibited by preventing or inhibiting proliferation with the conjugate include but are not limited to melanoma, adenocarcinoma, sarcomas, thymoma, lymphoma, lung tumors, liver tumors, colon tumors, kidney tumors, non-Hodgkins lymphoma, Hodgkins lymphoma, leukemias, uterine tumors, breast tumors, prostate tumors, renal tumors, ovarian tumors, pancreatic tumors, brain tumors, testicular tumors, bone tumors, muscle tumors, tumors of the placenta, gastric tumors, metastases and the like.
  • In some embodiments, the compositions as disclosed herein comprising a MetAP-2 inhibitor can inhibit proliferation of blood vessel endothelial cells, thus having anti-angiogenesis effect or inhibition of angiogenesis. Accordingly, the compositions as disclosed herein comprising MetAP-2 inhibitors can be used in methods for treating angiogenesis-mediated conditions in which MetAP-2 is involved in the pathology, such as inhibiting growth and metastasis of cancer as well as treating other various diseases where inappropriate angiogenesis occurs or proliferation of blood vessel endothelial cells occur, for example but not limited to, inflammatory diseases, diabetic retinopathy, rheumatoid arthritis, psoriasis. Accordingly, the compositions comprising MetAP-2 inhibitors as disclosed herein can be used as a cancer metastasis inhibitor or therapeutic agent against cancer, inflammatory diseases, diabetic retinopathy, rheumatoid arthritis, psoriasis and other retinopathies such as retinopathy of prematurity.
  • Actual dosage levels of active ingredients in the pharmaceutical compositions as described herein can be varied so as to obtain an amount of the active compound(s) which is effective to achieve the desired therapeutic response for a particular subject or patient. The selected dosage level will depend upon the activity of the particular MetAP-2 inhibitor, the type of administration composition (i.e. tablet versus liquid oral administration versus ocular versus topical versus inhaled, for example), the severity of the condition being treated and the condition and prior medical history of the patient being treated.
  • The phrase “therapeutically effective amount” of a compound, e.g., a MetAP-2 inhibitor as described herein means a sufficient amount of the compound to treat disorders, at a reasonable benefit/risk ratio applicable to any medical treatment. It will be understood, however, that the total daily usage of the compositions and formulations as disclosed herein will be decided by the attending physician within the scope of sound medical judgment. A “therapeutically effective amount” as the term is used herein need not eradicate a disease. Rather, a therapeutically effective amount will at least slow progression of a disease (as non-limiting example, the growth of a tumor or neoplasm) relative to progression without the therapeutic agent, for example the composition as disclosed herein comprising a MetAP-2-block copolymer conjugate. Thus, it is preferred, but not required that the therapeutic agent actually eliminate the disease.
  • The specific therapeutically effective dose level for any particular subject will depend upon a variety of factors including the disorder being treated and the severity of the disorder; activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the compositions and formulations as disclosed herein which are employed; and like factors well known in the medical arts. For example, it is well within the skill of the art to either start doses of the compound at levels lower than required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved, or start doses of the compound at high levels and to gradually decrease the dosage until the desired effect is achieved, as appropriate for the care of the individual patient.
  • The compositions as disclosed herein can also be administered in prophylatically or therapeutically effective amounts. The formulations and compositions as disclosed herein can be administered along with a pharmaceutically acceptable carrier. A prophylatically or therapeutically effective amount means that amount necessary, at least partly, to attain the desired effect, or to delay the onset of, inhibit the progression of, or halt altogether, the onset or progression of the particular disease or disorder being treated. Such amounts will depend, of course, on the particular condition being treated, the severity of the condition and individual patient parameters including age, physical condition, size, weight and concurrent treatment. These factors are well known to those of ordinary skill in the art and can be addressed with no more than routine experimentation. It is preferred generally that a maximum dose be used, that is, the highest safe dose according to sound medical judgment. It will be understood by those of ordinary skill in the art, however, that a lower dose or tolerable dose can be administered for medical reasons, psychological reasons or for virtually any other reasons.
  • The term “effective amount” as used herein refers to the amount of therapeutic agent of pharmaceutical composition to alleviate at least some of the symptoms of the disease or disorder. The term “effective amount” includes within its meaning a sufficient amount of pharmacological composition to provide the desired effect. The exact amount required will vary depending on factors such as the type of tumor to be treated, the severity of the tumor, the drug resistance level of the tumor, the species being treated, the age and general condition of the subject, the mode of administration and so forth. Thus, it is not possible to specify the exact “effective amount”. However, for any given case, an appropriate “effective amount” can be determined by one of ordinary skill in the art using only routine experimentation.
  • Efficacy of treatment can be judged by an ordinarily skilled practitioner. As disclosed in the Examples, efficacy can be assessed in animal models of cancer and tumor, for example treatment of a rodent with a cancer, and any treatment or administration of the compositions or formulations that leads to a decrease of at least one symptom of the cancer, for example a reduction in the size of the tumor or a slowing or cessation of the rate of growth of the tumor indicates effective treatment.
  • Efficacy for any given formulation (e.g., MetAP-2 inhibitor associated with block copolymer) can also be judged using an experimental animal model of cancer, e.g., wild-type mice or rats, or preferably, transplantation of tumor cells akin to that described in the Examples herein below. When using an experimental animal model, efficacy of treatment is evidenced when a reduction in a symptom of the cancer, for example a reduction in the size of the tumor or a slowing or cessation of the rate of growth of the tumor occurs earlier in treated, versus untreated animals. By “earlier” is meant that a decrease, for example in the size of the tumor occurs at least 5% earlier, but preferably more, e.g., one day earlier, two days earlier, 3 days earlier, or more.
  • In addition, the amount of each component to be administered also depends upon the frequency of administration, such as whether administration is once a day, twice a day, 3 times a day or 4 times a day, once a week; or several times a week, for example 2 or 3, or 4 times a week.
  • As an example only, TNP-470 administration will be described in greater detail as a representative example of the administration procedures for all MetAP-2 inhibitors in general. For 6-O—(N-chloroacetylcarbamoyl)fumagillol (TNP-470), the following information may serve as a general guideline for administration. Usually, the formulations and compositions as disclosed herein are administered from once a day to several times a day, for example 2 times a day, three times a day, or four times a day. In alternative embodiments, the formulations and compositions as disclosed herein can be administered, for example three to five times a week, if it is to be plurally administered in a given week. In some modes of administration, e.g., IV administration, it is desirable to dose less frequently, e.g., weekly or biweekly; block copolymer conjugate compounds can be useful in such a regimen.
  • For example, in one embodiment a suitable dose of the MetAP-2 inhibitor in the formulations and compositions as disclosed herein for a subject in need of treatment can be used according to conventionally used dose ranges of about 1 mg to about 2000 mg TNP-470 equivalent per kilogram of body weight. Generally however, conventional doses of fumagillol derivatives are about 0.1 mg/kg to 40 mg/kg body weight, preferably about 0.5 mg/kg to 20 mg/kg body weight as disclosed in U.S. Pat. No. 5,290,807. In alternative embodiments, where maintenance of tumor growth is the goal (i.e. the goal is to attenuate the growth of the tumor), a dose below the threshold used for chemotherapy can be used. For example, a suitable dose could be less than the conventionally used chemotherapeutic dose, for example, dose ranges of about 1 μg to 1 mg or 0.1 m to 1 mg, or 1 mg to 10 mg TNP-470 per kilogram of body weight can be used.
  • In some embodiments, if TNP-470 is administered once a week, it can be administered in an amount of from about 20 to about 200 mg/m2/week; preferably in an amount of from about 40 to about 180 mg/m2/week; and most preferably in an amount of from about 135 to about 175 mg/m2/week. In some embodiments, if TNP-470 is administered daily, it may be administered in an amount of from about 1 to about 10 mg/m2/day; for example in an amount of from about 1.25 to about 5 mg/m2/day; or in an amount of from about 1 to about 3 mg/m2/day. For continuous administration, the component is usually administered for at least five consecutive days of the week. In some embodiments, the effective amount of a composition as disclosed herein comprising a fumagillol derivative can be determined using an anti-angiogenesis assay as disclosed herein, and in some embodiments, the effective amount is less than the amount used as the conventionally effective dose. Similar dosage regimes can be applied for formulations of other MetAP-2 inhibitor compositions as disclosed herein.
  • In an alternative embodiment, higher dosages can be used, provided there is not unacceptable toxicity. For example, dosages in the range of about 50 to about 500 mg/m2/week or more, and sub-ranges within this range are specifically contemplated. Thus, dosages in the range of about 120 to 350 mg/m2/week, 200 to 400 mg/m2/week, etc. are specifically contemplated.
  • The preferred route of administration of the compositions and formulations as disclosed herein is oral administration. Solid dosage forms for oral administration include, for example but not limited to capsules, tablets, pills, powders and granules. In such solid dosage forms, the compositions as disclosed herein may be mixed with at least one inert, pharmaceutically acceptable excipient or carrier, such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol and silicic acid; b) binders such as carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; c) humectants such as glycerol; d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates and sodium carbonate; e) solution retarding agents such as paraffin; f) absorption accelerators such as quaternary ammonium compounds; g) wetting agents such as cetyl alcohol and glycerol monostearate; h) absorbents such as kaolin and bentonite clay and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The active components can also be in micro-encapsulated form, if appropriate, with one or more of the above-mentioned excipients. In the preparation of pharmaceutical formulations as disclosed herein in the form of dosage units for oral administration the compound selected can be mixed with solid, powdered ingredients, such as lactose, saccharose, sorbitol, mannitol, starch, arnylopectin, cellulose derivatives, gelatin, or another suitable ingredient, as well as with disintegrating agents and lubricating agents such as magnesium stearate, calcium stearate, sodium stearyl fumarate and polyethylene glycol waxes. The mixture is then processed into granules or pressed into tablets.
  • In addition, compositions for topical (e.g., oral mucosa, respiratory mucosa) and/or oral administration can form solutions, suspensions, tablets, pills, capsules, sustained-release formulations, oral rinses, or powders, as known in the art are described herein. The compositions also can include stabilizers and preservatives. For examples of carriers, stabilizers and adjuvants, University of the Sciences in Philadelphia (2005) Remington: The Science and Practice of Pharmacy with Facts and Comparisons, 21st Ed. The compositions can also be inhaled (pulmonary, nasal), ocular (eyedrop), sub-lingual, suppository, or topical (e.g., an ointment).
  • To enhance the activity of anti-proliferative treatments, use of adjunct treatments is contemplated. In particular, MetAP-2 inhibitors such as TNP-470 have been tested in conjunction with treatment with various other drugs to enhance efficacy for treatment of diseases induced by abnormally stimulated neovascularization, such as inflammatory diseases (rheumatism and psoriasis among others), diabetic retinopathy, cancer and other diseases and conditions as discussed elsewhere herein.
  • Soft gelatin capsules can be prepared with capsules containing a mixture of the active compound or compounds of the invention in vegetable oil, fat, or other suitable vehicle for soft gelatin capsules. Hard gelatin capsules can contain granules of the active compound. Hard gelatin capsules can also contain the targeted delivery composition including the targeting moiety and the carrier particle as well as the therapeutic agent in combination with solid powdered ingredients such as lactose, saccharose, sorbitol, mannitol, potato starch, corn starch, amylopectin, cellulose derivatives or gelatin.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups and elixirs. In addition to the active components, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents that are compatible with the maintenance of a micelle of a diblock copolymer as described herein. Liquid preparations for oral administration can also be prepared in the form of syrups or suspensions, e.g. solutions or suspensions containing from 0.2% to 20% by weight of the active ingredient and the remainder consisting of sugar or sugar alcohols and a mixture of ethanol, water, glycerol, propylene glycol and polyethylene glycol provided that such solvent is compatible with maintaining the micelle form. If desired, such liquid preparations can contain coloring agents, flavoring agents, saccharin and carboxymethyl cellulose or other thickening agents. Liquid preparations for oral administration can also be prepared in the form of a dry powder to be reconstituted with a suitable solvent prior to use.
  • Besides inert diluents, the oral compositions may also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring and perfuming agents
  • Transdermal patches may also be used to provide controlled delivery of the formulations and compositions as disclosed herein to specific regions of the body. Such dosage forms can be made by dissolving or dispensing the component in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate-controlling membrane or by dispersing the compound in a polymer matrix or gel. Such transdemal patches are useful for treating parts of the body where abnormally stimulated neovascularization occurs, such as inflammatory diseases, for example rheumatism and psoriasis among others, diabetic retinopathy and cancer, for example skin cancer or other skin related neovascular conditions (such as psoriasis) or malignancies.
  • A further form of topical administration is to the eye, for example as a treatment for retinopathy, such as diabetic retinopathy or retinopathy of prematurity (ROP) or for the treatment of immune-mediated conditions of the eye such as autoimmune diseases, allergic or inflammatory conditions, and corneal transplants. Components of the invention may be delivered in a pharmaceutically acceptable ophthalmic vehicle, such that the component is maintained in contact with the ocular surface for a sufficient time period to allow the component to penetrate the corneal and internal regions of the eye, as for example the anterior chamber, posterior chamber, vitreous body, aqueous humor, vitreous humor, cornea, iris/cilary, lens, choroid/retina and sclera. The pharmaceutically acceptable ophthalmic vehicle may, for example, be an ointment or an encapsulating material.
  • In an alternative embodiment, the compositions and formulations as disclosed herein can be also administered via rectal or vaginal administration. In such embodiments, the compositions and formulations as disclosed herein can be in the form of suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at room temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active component.
  • Dosage units for rectal or vaginal administration can be prepared (i) in the form of suppositories which contain the active substance mixed with a neutral fat base; (ii) in the form of a gelatin rectal capsule which contains the active substance in a mixture with a vegetable oil, paraffin oil or other suitable vehicle for gelatin rectal capsules; (iii) in the form of a ready-made micro enema; or (iv) in the form of a dry micro enema formulation to be reconstituted in a suitable solvent just prior to administration.
  • To further protect the active ingredient, the MetAP-2 inhibitor-copolymer conjugates described herein can be used in admixture or in combination with a gastric acid secretion-inhibitor and/or an antacid.
  • Gastric acid secretion inhibitors include, for example H2 blockers (e.g. famotidine, cimetidine, ranitidine hydrochloride, etc.) and proton pump inhibitors (e.g. lansoprazole, omeprazole, etc.). As an antacid, compounds which elevate the intragastric pH level, such as magnesium carbonate, sodium hydrogen carbonate, magnesium hydroxide, magnesium oxide and magnesium hydroxide can be employed. The oral dosage forms of the compositions and formulations as disclosed herein can be administered after the intragastric pH has been increased to alleviate the influence of gastric acid by the administration of a gastric acid secretion inhibitor and/or antacid.
  • Alternatively, compositions and formulations as disclosed herein can be in a form of enteric-coated preparation for oral administration comprising a MetAP-2 inhibitor-block copolymer conjugate. In some embodiments, a MetAP-2 inhibitor containing core for coating with an enteric coating film can be prepared using an oleaginous base or by other known formulation methods without using an oleaginous base. In some embodiments, the compositions and formulations as disclosed herein in the form of the drug-containing core for coating with a coating agent may be, for example, tablets, pills and granules.
  • The excipient contained in the core is exemplified by saccharides, such as sucrose, lactose, mannitol and glucose, starch, crystalline cellulose and calcium phosphate. Useful binders include polyvinyl alcohol, hydroxypropyl cellulose, macrogol, Pluronic F-68, gum arabic, gelatin and starch. Useful disintegrants include carboxymethyl cellulose calcium (ECG505), crosslinked carboxymethylcellulose sodium (Ac-Di-Sol), polyvinylpyrrolidone and low-substituted hydroxypropyl cellulose (L-HPC). Useful lubricants and antiflocculants include talc and magnesium stearate.
  • The enteric coating agent is an enteric polymer which is substantially insoluble in the acidic pH and is at least partially soluble at weaker acidic pH through the basic pH range. The range of acidic pH is about 0.5 to about 4.5, preferably about 1.0 to about 2.0. The range of weaker acidic pH through basic pH is about 5.0 to about 9.0, preferably about 6.0 to about 7.5. Specifically, cellulose acetate phthalate, hydroxypropylmethylcellulose phthalate, hydroxypropylmethyl acetate succinate (Shin-Etsu Chemicals), methacrylic copolymers (Rhon-Pharma, Eudragit® L-30D-55, L100-55, L100, S100, etc.), etc. can be mentioned as examples of the enteric coating agent. These materials are effective in terms of stability, even if they are directly used as enteric compositions.
  • In the case of forming micelles, for example, spherical microparticles having particle diameters of about 0.01 μm to about 1000 μm are generated, or more preferably about 0.01 to about 5 μm (about 10 nm to about 5,000 nm). This formation can be performed by the methods as disclosed in the Examples or as disclosed in Japanese Patent Application JP-A-223533/1991. In some embodiments, micelles between 50 and 500 nm are useful in the compositions as disclosed herein, for example about 50-100 nm, 100-150 nm, 150-200 nm, 200-250 nm, 250-300 nm, 300-350 nm, 350-400 nm, 450-500 nm. In some embodiments, the micelles are about 130 nm as disclosed in the examples.
  • The concentration or content of the MetAP-2 inhibitor in the composition can be appropriately selected according to the physicochemical properties of the composition. When the composition is in a liquid form, the concentration is about 0.0005 to about 30% (w/v) and preferably about 0.005 to about 25% (w/v). When the composition is a solid, the content is about 0.01 to about 90% (w/w) and preferably about 0.1 to about 50% (w/w).
  • If necessary, additives such as a preservative (e.g. benzyl alcohol, ethyl alcohol, benzalkonium chloride, phenol, chlorobutanol, etc.), an antioxidant (e.g. butylhydroxyanisole, propyl gallate, ascorbyl palmitate, alpha-tocopherol, etc.), and a thickener (e.g. lecithin, hydroxypropylcellulose, aluminum stearate, etc.) can be used in the compositions and formulations as disclosed herein.
  • It is noted that diblock copolymer conjugates as described generally need no further emulsifiers. Nonetheless, if necessary, one can use an additional emulsifier with the compositions and formulations as disclosed herein. Examples of emulsifiers that might be used include pharmaceutically acceptable phospholipids and nonionic surfactants. The emulsifiers can be used individually or in combinations of two or more. The phospholipid includes naturally occurring phospholipids, e.g. egg yolk lecithin, soya lecithin, and their hydrogenation products, and synthetic phospholipids, e.g. phosphatidylcholine, phosphatidylethanolamine, etc. Among them, egg yolk lecithin, soya lecithin, and phosphatidylcholine derived from egg yolk or soybean are preferred. The nonionic surfactant includes macro-molecular surfactants with molecular weights in the range of about 800 to about 20000, such as polyethylene-propylene copolymer, polyoxyethylene alkyl ethers, polyoxyethylene alkylarylethers, hydrogenated castor oil-polyoxyethylene derivatives, polyoxyethylene sorbitan derivatives, polyoxyethylene sorbitol derivatives, polyoxyethylene alkyl ether sulfate, and so on. The proportion of the emulsifier is selected so that the concentration in a final administrable composition will be in the range of about 0.1 to about 10%, preferably about 0.5 to about 5%.
  • In addition to the above-mentioned components, a stabilizer for further improving the stability of the compositions and formulations as disclosed herein, such as an antioxidant or a chelating agent, an isotonizing agent for adjusting the osmolarity, an auxiliary emulsifier for improving the emulsifying power, and/or an emulsion stabilizer for improving the stability of the emulsifying agent can be incorporated. The isotonizing agent that can be used includes, for example, gylycerin, sugar alcohols, monosaccharides, disaccharides, amino acids, dextran, albumin, etc. These isotonizing agents can be used individually or in combination, with two or more. An emulsion stabilizer that can be used, which includes cholesterol, cholesterol esters, tocopherol, albumin, fatty acid amide derivatives, polysaccharides, polysaccharide fatty acid ester derivatives, etc.
  • The compositions and formulations as disclosed herein can further comprise a viscogenic substance which can adhere to the digestive tract mucosa due to its viscosity expressed on exposure to water. The examples of the viscogenic substance include, but are not particularly limited as long as it is pharmaceutically acceptable, such as polymers (e.g. polymers or copolymers of acrylic acids and their salts) and natural-occurring viscogenic substances (e.g. mucins, agar, gelatin, pectin, carrageenin, sodium alginate, locust bean gum, xanthan gum, tragacanth gum, arabic gum, chitosan, pullulan, waxy starch, sucralfate, curdlan, cellulose, and their derivatives). Furthermore, for controlling the release of the active drug or for formulation purposes, the additives conventionally used for preparing the oral compositions can be added. Example of the additives include excipients (e.g. lactose, corn starch, talc, crystalline cellulose, sugar powder, magnesium stearate, mannitol, light anhydrous silicic acid, magnesium carbonate, calcium carbonate, L-cysteine, etc.), binders (e.g. starch, sucrose, gelatin, arabic gum powder, methylcellulose, carboxymethylcellulose, carboxymethylcellulose sodium, hydroxypropylcellulose, hydroxypropylmethylcellulose, polyvinylpyrrolidone, pullulan, dextrin, etc.), disintegrators (e.g. carboxymethylcellulose calcium, low-substituted hydroxypropylcellulose, croscarmellose sodium, etc.), anionic surfactants (e.g. sodium alkylsulfates etc.), nonionic surfactants (e.g. polyoxyethylene sorbitan fatty acid esters, polyoxyethylene fatty acid esters, polyoxyethylene-castor oil derivatives, etc.), antacids and mucous membrane protectants (e.g. magnesium hydroxide, magnesium oxide, aluminum hydroxide, aluminum sulfate, magnesium metasilicate aluminate, magnesium silicate aluminate, sucralfate, etc.), cyclodextrin and the corresponding carboxylic acid (e.g. maltosyl-beta-cyclodextrin, maltosyl-beta-cyclodextrin-carboxylic acid, etc.), colorants, corrigents, adsorbents, antiseptics, moistening agents, antistatic agents, disintegration retardants, and so on. The proportion of these additives can be appropriately selected from the range that can keep the stability and absorption of the basis.
  • The compositions and formulations as disclosed herein for oral administration of the present invention may also include flavoring agents. Such agents include, for example, anise oil, lavender oil, lemon oil, orange essence, rose oil, powder green tea, bergamot oil, (alpha[litre]) borneol, Natural Peal Extract AH-10, Sugar, bitter essence, pine flavor etc.
  • In the case of forming micelles, for example, spherical microparticles having particle diameters of about 0.1 nm to about 1000 nm, this formation can be achieved by methods known in the art (e.g. JP-A-223533/1991). In some embodiments, micelles between 50 and 500 nm are useful in the compositions as disclosed herein, for example about 50-100 nm, 100-150 nm, 150-200 nm, 200-250 nm, 250-300 nm, 300-350 nm, 350-400 nm, 450-500 nm. In some embodiments, the micelles are about 130 nm as disclosed in the examples.
  • The compositions and formulations as disclosed herein can exhibit potent pharmacological activity with low toxicity such that they are useful as a medicament for prevention and treatment for, interalia, MetAP-2-associated disease in mammals (e.g. mouse, rat, monkey, bovine, canine, human, etc.), the MetAP-2-associated disease including, for example, (1) inflammatory diseases such as rheumatoid arthritis, (2) diabetic retinopathy, and (3) benign and malignant tumors (e.g. gastric cancer, cancer of the esophagus, duodenal cancer, cancer of the tongue, pharyngeal cancer, brain tumors, neurilemoma, colorectal cancer, non-small-cell lung cancer, small cell carcinoma of the lung, hepatic carcinoma, renal cancer, cancer of the breast, biliary tract cancer, cancer of the pancreas, cancer of the prostate, cancer of the uterus, carcinoma of the uterine cervix, ovarian cancer, cancer of the urinary bladder, cancer of the skin, malignant melanoma, cancer of the thyroid, sarcomas of bone, hemangioma, hemangiofibroma, retinal sarcoma, cancer of the penis, solid tumors of childhood, Kaposi's sarcoma in AIDS, etc., inclusive of recurrencies and metastases to other organs), fibrotic diseases such as idiopathic pulmonary fibrosis, or cystic fibrosis. It is particularly useful when the dosage form of the compositions and formulations as disclosed herein insures an effective blood concentration within the range not causing expression of the side effects of the active substance in prolonged time, or not contributing to new side effects due to prolonged blood circulation or depoting in organ tissue for lack of renal or other clearance mechanism.
  • Generally, no enteric coating is required for the fumagillol-derivative diblock copolymer formulations described herein to be orally effective. It can be advantageous, however, from the standpoint of stability, that the compositions and formulations as disclosed herein are filled into capsule shells coated with an enteric coating agent as mentioned above for use as an enteric composition. As the capsule shell, for example, soft capsules (e.g. the product of R. P. Sealer) and hard gelatin capsules are used.
  • The liquid or solid compositions and formulations as disclosed herein can be administered orally. In the case of the liquid form, it can be directly administered e.g., by drinking an elixir or suspension of the composition, or alternatively, into the digestive tract via a catheter or sonde for oral administration or administered in the usual manner in the unit dosage form of a hard capsule or a soft capsule. In the case of the solid form, it can be administered orally as powders, capsules, tablets, or the like in the usual manner. It can also be redispersed in a suitable dispersion medium and administered in a liquid form. Taking a patient of breast cancer (body weight: 50 kg) as an example, the oral dose of the composition as disclosed herein is about 1 mg to about 3 g/day, preferably about 10 mg to about 1 g/day, of a MetAP-2 inhibitor. In some embodiments, the oral dose of the composition as disclosed herein is between the ranges of about 25 mg to about 1 g/day, and in some embodiments less than 25 mg to 1 g/day, for example about 10 mg to about 0.5 g/day, of a MetAP-2 inhibitor.
  • The compositions and formulations as disclosed herein enhance biodistribution, stability and uptake properties of MetAP-2 inhibitors and increase the pharmacological activity thereof, so that better assurance of therapeutic efficacy is achieved without the need for parenteral administration. The dosage form of the compositions and formulations as disclosed herein is stable and exhibits remarkable inhibiting activity on proliferative activity in oral administration so that it can be used as clinically advantageous oral medicine.
  • Suspensions, in addition to the active components, may contain suspending agents, as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like.
  • Proper fluidity can be maintained, for example, by the use of coating materials such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.
  • In one embodiment, the delivery is by intranasal administration of the composition, especially for use in therapy of the brain and related organs (e.g., meninges and spinal cord). Along these lines, intraocular administration is also possible. Suitable formulations can be found in Remington's Pharmaceutical Sciences, 16th and 18th Eds., Mack Publishing, Easton, Pa. (1980 and 1990), and Introduction to Pharmaceutical Dosage Forms, 4th Edition, Lea & Febiger, Philadelphia (1985), each of which is incorporated herein by reference.
  • In one embodiment, the present invention encompasses combination therapy in which the formulations and compositions as disclosed herein are used in combination with a chemotherapeutic agent such as Taxol, cyclophosphamide, cisplatin, gancyclovir and the like. The chemotherapeutic agent may also be included within a micelle as described herein. Such a therapy is particularly useful in situations in which the subject or patient to be treated has a large preexisting tumor mass which is well vascularized. The chemotherapeutic agent serves to reduce the tumor mass and the conjugate prevents or inhibits neovascularization within or surrounding the tumor mass. The chemotherapeutic agent may also be administered at lower doses than normally used and at such doses may act as an anti-proliferative agent. The second therapy can be administered to the subject before, during, after or a combination thereof relative to the administration of the compositions as disclosed herein. Anti-proliferative therapies are well known in the art and are encompassed for use in the methods of the present invention. Therapies includes, but are not limited to an alkylating agent, mitotic inhibitor, antibiotic, or antimetabolite, anti-angliogenic agents etc. Such chemotherapy can comprise administration of CPT-11, temozolomide, or a platin compound. Radiotherapy can include, for example, x-ray irradiation, w-irradiation, γ-irradiation, or microwaves.
  • The term “chemotherapeutic agent” or “chemotherapy agent” are used interchangeably herein and refers to an agent that can be used in the treatment of cancers and neoplasms, for example brain cancers and gliomas and that is capable of treating such a disorder. In some embodiments, a chemotherapeutic agent can be in the form of a prodrug which can be activated to a cytotoxic form. Chemotherapeutic agents are commonly known by persons of ordinary skill in the art and are encompassed for use in the present invention. For example, chemotherapeutic drugs for the treatment of tumors and gliomas include, but are not limited to: temozolomide (Temodar), procarbazine (Matulane), and lomustine (CCNU). Chemotherapy given intravenously (by IV, via needle inserted into a vein) includes vincristine (Oncovin or Vincasar PFS), cisplatin (Platinol), carmustine (BCNU, BiCNU), and carboplatin (Paraplatin), Mexotrexate (Rheumatrex or Trexall), irinotecan (CPT-11); erlotinib; oxalipatin; anthracyclins-idarubicin and daunorubicin; doxorubicin; alkylating agents such as melphalan and chlorambucil; cis-platinum, methotrexate, and alkaloids such as vindesine and vinblastine.
  • In another embodiment, the present invention encompasses combination therapy in which the formulations and compositions as disclosed herein are used in combination with, a cytostatic agent, anti-VEGF and/or p53 reactivation agent. A cytostatic agent is any agent capable of inhibiting or suppressing cellular growth and multiplication. Examples of cytostatic agents used in the treatment of cancer are paclitaxel, 5-fluorouracil, 5-fluorouridine, mitomycin-C, doxorubicin, and zotarolimus. Other cancer therapeutics include inhibitors of matrix metalloproteinases such as marimastat, growth factor antagonists, signal transduction inhibitors and protein kinase C inhibitors.
  • In another embodiment, the methods described herein are administered in conjunction with an anti-VEGF agent. Some examples of anti-VEGF agents include bevacizumab (Avastin™) VEGF Trap, CP-547,632, AG13736, AG28262, SU5416, SU11248, SU6668, ZD-6474, ZD4190, CEP-7055, PKC 412, AEE788, AZD-2171, sorafenib, vatalanib, pegaptanib octasodium, IM862, DC101, angiozyme, Sirna-027, caplostatin, neovastat, ranibizumab, thalidomide, and AGA-1470, a synthetic analog of fumagillin (alternate names: Amebacilin, Fugillin, Fumadil B, Fumadil) (A. G. Scientific, catalog #F1028), an angio-inhibitory compound secreted by Aspergillus fumigates.
  • As used herein the term “anti-VEGF agent” refers to any compound or agent that produces a direct effect on the signaling pathways that promote growth, proliferation and survival of a cell by inhibiting the function of the VEGF protein, including inhibiting the function of VEGF receptor proteins. The term “agent” or “compound” as used herein means any organic or inorganic molecule, including modified and unmodified nucleic acids such as antisense nucleic acids, RNAi agents such as siRNA or shRNA, peptides, peptidomimetics, receptors, ligands, and antibodies. Preferred VEGF inhibitors, include for example, AVASTIN® (bevacizumab), an anti-VEGF monoclonal antibody of Genentech, Inc. of South San Francisco, Calif., VEGF Trap (Regeneron/Aventis). Additional VEGF inhibitors include CP-547,632 (3-(4-Bromo-2,6-difluoro-benzyloxy)-5-[3-(4-pyrrolidin 1-yl-butyl)-ureido]-isothiazole-4-carboxylic acid amide hydrochloride; Pfizer Inc., NY), AG13736, AG28262 (Pfizer Inc.), SU5416, SU11248, & SU6668 (formerly Sugen Inc., now Pfizer, New York, N.Y.), ZD-6474 (AstraZeneca), ZD4190 which inhibits VEGF-R2 and -R1 (AstraZeneca), CEP-7055 (Cephalon Inc., Frazer, Pa.), PKC 412 (Novartis), AEE788 (Novartis), AZD-2171), NEXAVAR® (BAY 43-9006, sorafenib; Bayer Pharmaceuticals and Onyx Pharmaceuticals), vatalanib (also known as PTK-787, ZK-222584: Novartis & Schering: AG), MACUGEN® (pegaptanib octasodium, NX-1838, EYE-001, Pfizer Inc./Gilead/Eyetech), IM862 (glufanide disodium, Cytran Inc. of Kirkland, Wash., USA), VEGFR2-selective monoclonal antibody DC101 (ImClone Systems, Inc.), angiozyme, a synthetic ribozyme from Ribozyme (Boulder, Colo.) and Chiron (Emeryville, Calif.), Sirna-027 (an siRNA-based VEGFR1 inhibitor, Sirna Therapeutics, San Francisco, Calif.) Caplostatin, soluble ectodomains of the VEGF receptors, Neovastat (
    Figure US20160136292A1-20160519-P00001
    terna Zentaris Inc; Quebec City, Calif.) and combinations thereof.
  • In yet another embodiment, the present invention encompasses combination therapy in which the formulations and compositions as disclosed herein are used in combination or conjunction with therapeutics, physiotherapy and/or behavioral psychotherapy used in the treatment of rheumatoid arthritis, obesity, endometriosis, idiopathic pulmonary fibrosis (IPF), lupus, and Alzheimer's disease.
  • For examples of treatments of rheumatoid arthritis, there are therapeutic drugs that decrease pain and local inflammation including aspirin and non-steroidal anti-inflammatory drugs or NSAIDS (such as ibuprofen or naproxen) and other immunosuppressive drugs that decrease pain and inflammation while decreasing the growth of abnormal synovial tissue (the tissue that lines the inside of the joint). These drugs include methotrexate and low doses of corticosteroids (such as prednisone or cortisone). Other medications used to treat rheumatoid arthritis include: anti-malarial medications (such as hydroxychloroquine), gold, sulfasalazine, penicillamine, cyclophosphamide, cyclosporine, minocycline, interleukin receptor antagonist and anti-IL2 antibodies.
  • In particular embodiments, the formulations and compositions as disclosed herein are particularly useful for administration in conjunction therapies used for treatment of diseases associated with vascular permeability, such as vascular complications of diabetes such as non-proliferative diabetic retinopathy and nephropathy, nephrotic syndrome, pulmonary hypertension, burn edema, tumor edema, brain tumor edema, IL-2 therapy-associated edema, and other edema-associated diseases, as disclosed in International Application No: WO2003/086178 and U.S. Patent Applications US2005/0203013 and US2005/0112063 which are incorporated herein in their entirety by reference. In a particular embodiment, the formulations and compositions as disclosed herein are particularly useful for administration in conjunction with IL-2 therapy, where the limiting factor of IL-2 therapy is IL-2 therapy-associated edema as disclosed in International Application No: WO2003/086178 and U.S. Patent Applications US2005/0203013 and US2005/0112063.
  • Treatment for Alzheimer's disease include, but are not be limited to, nonsteroidal anti-inflammatory drugs (NSAIDs), estrogen, steroids such as prednisone, vitamin E, menantine, donepezil, rivastigmine, tacrine, and galantamine. Holistic medicine include example such as gingko nuts extracts.
  • Treatment of endometrosis include, but should not be construed as limited to, a combination oral contraceptives (estrogen plus a progestin), progestins (such as medroxyprogesterone, danazol (a synthetic hormone related to testosterone, gonadotropin-releasing hormone agonists (GnRH agonists such as buserelin, goserelin, leuprolide and nafarelin), and nonsteroidal anti-inflammatory drugs (NSAIDs) for pain control.
  • Examples of treatment options for obesity include dieting and nutritional counseling, exercise regime, gastric-bypass surgery, and drugs such as a combination of fenfluramine and phentermine (often called fen-phen), orlistat, sibutramine, phentermine, benzphetamine, diethylpropion, mazindol, and phendimetrazine.
  • In alternative embodiments, the formulations and compositions can comprise a plurality of micelles, wherein some micelles comprise MetAP-2 inhibitor, and other micelles comprise other therapeutic agents, for example chemotherapeutic agents and antineoplastic agents.
  • The amount of the pharmaceutical composition as disclosed herein when administered to a subject will, of course, be dependent on the subject being treated, the severity of the affliction, the manner of administration, the judgment of the prescribing physician, etc.
  • The formulations and pharmaceutical composition can, if desired, be presented in a suitable container (e.g., a pack or dispenser device), such as an FDA approved kit, which can contain one or more unit dosage forms containing the carrier portion containing the targeting and immune response triggering portions.
  • Proliferative Assays
  • In general, one of skill in the art will know whether a given agent, for example a fumagillol derivative, is an anti-proliferative or anti-angiogenic agent. For the avoidance of doubt, one can use any of a number of in vitro or in vivo assays to evaluate the influence of a given agent on proliferation or angiogenesis. Whether or not a composition or formulation comprising a MetAP-2 inhibitor block copolymer conjugate in accordance with the present invention, can treat or prevent diseases associated with an angiogenesis-mediated condition can be determined by its effect in the mouse model as shown in the Examples below. However, at a minimum, a MetAP-2 inhibitor block copolymer as described herein will have anti-proliferative activity (i.e., at least 50% of the anti-proliferative activity of TNP-470) in a HUVEC assay as described in the Examples herein.
  • Another useful assay for determining if the compositions and formulations has disclosed herein have anti-proliferative or anti-angiogenesis activity is the CAM assay, which is frequently used to evaluate the effects of endothelial cell proliferation regulating factors because it is relatively easy and provides relatively rapid results. A proliferation-regulating factor useful in the methods and compositions described herein will modify the number of microvessels in the modified CAM assay described by Iruela-Arispe et al., 1999, Circulation 100: 1423-1431. The method is based on the vertical growth of new capillary vessels into a collagen gel pellet placed on the CAM. In the assay as described by Iruela-Arispe et al., the collagen gel is supplemented with a proliferative factor such as FGF-2 (50 ng/gel) or VEGF (250 ng/gel) in the presence or absence of test agents. The extent of the proliferative response is measured using FITC-dextran (50 μg/mL) (Sigma) injected into the circulation of the CAM. The degree of fluorescence intensity parallels variations in capillary density; the linearity of this correlation can be observed with a range of capillaries between 5 and 540. Morphometric analyses are performed, for example, by acquisition of images with a CCD camera. Images are then analyzed by imported into an analysis package, e.g., NHImage 1.59, and measurements of fluorescence intensity are obtained as positive pixels. Each data point is compared with its own positive and negative controls present in the same CAM and interpreted as a percentage of inhibition, considering the positive control to be 100% (VEGF or FGF-2 alone) and the negative control (vehicle alone) 0%. Statistical evaluation of the data is performed to check whether groups differ significantly from random, e.g., by analysis of contingency with Yates' correction.
  • Additional proliferative assays, including additional assays specifically designed to monitor angiogenic proliferation are known in the art and can be used to evaluate MetAP-2 inhibitors for use in the methods and compositions described herein. These include, for example, the corneal micropocket assay, hamster cheek pouch assay, the Matrigel assay and modifications thereof, and co-culture assays. Donovan et al. describe a comparison of three different in vitro assays developed to evaluate angiogenesis regulators in a human background (Donovan et al., 2001, Angiogenesis 4: 113-121, incorporated herein by reference). Briefly, the assays examined include: 1) a basic Matrigel assay in which low passage human endothelial cells (Human umbilical vein endothelial cells, HUVEC) are plated in wells coated with Matrigel (Becton Dickinson, Cedex, France) with or without angiogenesis regulator(s); 2) a similar Matrigel assay using “growth factor reduced” or GFR Matrigel; and 3) a co-culture assay in which primary human fibroblasts and HUVEC are co-cultured with or without additional angiogenesis regulator(s), the fibroblasts produce extracellular matrix and other factors that support HUVEC differentiation and tubule formation. In the Donovan et al. paper the co-culture assay provided microvessel networks that most closely resembled microvessel networks in vivo. However, the basic Matrigel assay and the GFR Matrigel assay can also be used by one of skill in the art to evaluate whether a given fumagillol derivative is an angiogenesis-inhibiting agent as necessary for the methods and compositions described herein. Finally, an in vitro angiogenesis assay kit is marketed by Chemicon (Millipore). The Fibrin Gel In Vitro Angiogenesis Assay Kit is Chemicon Catalog No. ECM630.
  • Other proliferative assays are disclosed in International Application No: WO2003/086178 and U.S. Patent Applications US2005/0203013 and US2005/0112063, and involve assaying endothelial cells on a permeable substrate (e.g., a collagen coated inserts of “Transwells”), contacting the assay with a test compound (e.g., a fumagillol derivative block copolymer conjugate), treating the assay with a marker (e.g., FITC label) and a permeability-inducing agent (e.g., vascular endothelial growth factor (VEGF) and platelet-activating factor (PAP) among others), and measuring the rate of diffusion of the marker compare to control. Compounds that are found to affect vascular permeability can be further tested for anti-proliferative activity using existing methods. The bioeffectiveness of MetAP-2 inhibitor block copolymer conjugate as an anti-proliferative or angiogenic agent in a patient being treated with such a block copolymer conjugate can be assessed by methods commonly known by person skilled in the art, for example, as disclosed in International Application No: WO2003/086178 and U.S. Patent Applications US2005/0203013 and US2005/0112063. One approach involves administering to the patient an intradermal injection of histamine before treating the subject with the a MetAP-2 block copolymer conjugate and measuring a histamine-induced local edema. Then, treating the subject with the a MetAP-2 inhibitor block copolymer conjugate, and again administering to the subject an intradermal injection of histamine subsequent to treating the subject with the MetAP-2 inhibitor block copolymer conjugate and measuring the histamine-induced local edema. A decrease in the measurement of the histamine-induced local edema compared to that seen before the treatment with the MetAP-2 inhibitor block copolymer conjugate indicates that the compound is bioeffective.
  • Composition or formulation as disclosed herein capable of preventing or treating non-proliferative diabetic retinopathy can be tested by in vitro studies of endothelial cell proliferation and in other models of diabetic retinopathy, such as Streptozotocin. In addition, color Doppler imaging can be used to evaluate the action of a drug in ocular pathology (Valli et al., Ophthalmologica 209(13): 115-121 (1995)). Color Doppler imaging is a recent advance in ulkasonography, allowing simultaneous two-dimension imaging of structures and the evaluation of blood flow. Accordingly, retinopathy can be analyzed using such technology. The manner in which the compositions and formulations as disclosed herein are administered is dependent, in part, upon whether the treatment of a disease associated with vascular hyperpermeability, including non-proliferative retinopathy is prophylactic or therapeutic. For example, the manner in which compositions and formulations as disclosed herein are administered for treatment of retinopathy is dependent, in part, upon the cause of the retinopathy. Specifically, given that diabetes is the leading cause of retinopathy, the compositions and formulations as disclosed herein can be administered preventatively as soon as the pre-diabetic retinopathy state is detected.
  • It should be understood that this invention is not limited to the particular methodology, protocols, and reagents, etc., described herein and as such may vary. The terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention, which is defined solely by the claims.
  • Other than in the operating examples, or where otherwise indicated, all numbers expressing quantities of ingredients or reaction conditions used herein should be understood as modified in all instances by the term “about.” The term “about” when used in connection with percentages may mean±1%. The present invention is further explained in detail by the following examples, but the scope of the invention should not limit thereto.
  • EXAMPLES
  • The examples presented herein relate to methods, compositions and formulations of MetAP-2 inhibitors for oral or alternative administration. Throughout this application, various publications are referenced. The disclosures of all of the publications and those references cited within those publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art to which this invention pertains. The following examples are not intended to limit the scope of the claims to the invention, but are rather intended to be exemplary of certain embodiments. Any variations in the exemplified methods which occur to the skilled artisan are intended to fall within the scope of the present invention.
  • Methods Abbreviations
  • mPEG-PLA=methoxy-poly(ethylene glycol)-poly(lactic acid), Suc=succinated; EDC=ethyl(diethylaminopropyl) carbodiimide; NHS=N-hydroxysuccinimide; en=ethylenamine; DDW=Double distilled water; DMF=Dimethylformamide; DMSO=Dimethyl sulfoxide.
  • Animals
  • All animal procedures were performed in compliance with Boston Children's Hospital guidelines, and protocols were approved by the Institutional Animal Care and Use Committee.
  • Mice (C57/B16J, 8 weeks old) were purchased from Jackson Laboratories (Bar Harbor, Me., USA). For rat studies, non pigmented Lewis rats (8-10 wk old, Charles River Laboratories) were used. All protocols were approved by the Institutional Animal Care and Use Committee at Children's Hospital Boston and were conducted in accordance with the Association for Research in Vision and Ophthalmology's Statement for the Use of Animals in Ophthalmic and Vision Research.
  • Log D Measurement for TNP-470
  • Aqueous solubility is one of the important chemical properties affecting oral absorption of a drug. In order to predict the intestinal absorption of TNP-470, we measured its Log-D value which is a parameter of hydrophobicity determined by the ratio of drug concentration in octanol to that in water at 25° C. (Analiza, Cleveland Ohio). High Log-D values (>2) indicate low water solubility and hence a poor oral availability of a drug. For this study Log-D values of TNP-470 (30 mM) were measured at plasma and stomach pHs: pH=7.4 and pH=2 respectively.
  • Preparation of PEG-PLA-MetAP-2 polymersomes(polymeric-micelles).
  • A known MetAP-2 inhibitor, TNP-470 (Takeda) was chosen for its activity, hydrophobicity and poor pharmacokinetic profile (see Placidi, Cancer Res. 55, 3036-3042 (1995)) and was bound to a diblock co-polymer using a two-step conjugation (FIG. 6A). In the first step succinated monomethoxy-poly(ethylene glycol)-poly (lactic acid) (mPEG2000-PLA1000) with free carboxic acid end-groups (Advanced Polymers Materials Inc.), was reacted with ethylenediamine (Sigma-Aldrich). Succinated mPEG-b-PLA-OOCCH2CH2COOH (500 mg) was dissolved in DMSO and reacted with ethyl(diethylaminopropyl) carbodiimide (EDC) and N-hydroxysuccinimide (NHS) in a molar ratio of 1:10:20 (Polymer:EDC:NHS respectively). In an alternative approach, the same amount of succinated mPEG-b-PLA-OOCCH2CH2COOH is dissolved in 10 ml MES buffer at pH 6 and the catalyst is N-hydroxysuccinimide (sulfo-NHS)). A five fold molar excess of ethylendiamine was added and reacted for 4 h at 25° C. The polymer solution was then dialyzed (MWCO 1000, Spectra/Por Biotech Regenerated Cellulose VWR) against DMSO leading to a 65% reaction efficiency. In the second step, the amine containing polymer was mixed with TNP-470 (350 gr), dissolved in DMSO and the solution was stirred for 4 h at 25° C. The polymeric micelles were formed by dialyzing the DMSO solution of the conjugate against double distilled water using a regenerated cellulose dialysis bag (MWCO 1000, Spectra/Por Biotech Regenerated Cellulose, VWR) to obtain micelles with high incorporation efficiency (>90%) and 0.8-1% free drug (w/w). The micelles were then lyophilized and stored at −20° C. in a dry environment until use.
  • Rhodamine labeled polymeric-micelles were formed using a similar protocol. The mPEG-PLA was conjugated via the N-terminal amine group with Lissamine rhodamine B sulfonyl chloride (Molecular Probes, Eugene, Oreg.) in DMSO. For green fluorescent polymer micelles, a commonly used hydrophobic marker 6-coumarin (Sigma-Aldrich, MO, USA) at 0.1% w/w was added to the polymeric solution before the final dialysis step.
  • For fluorescent labeled Lodamin, a commonly used hydrophobic marker 6-coumarin (Sigma-Aldrich) at 0.1% wt/wt was added to the polymeric solution before the final dialysis step. Characterization of conjugation by NMR analysis
  • Nuclear Magnetic Resonance (NMR) spectrometer analysis (NERCE/BEID, Harvard Medical School) was conducted for each reaction step and Mass spectrometry (Proteomic core, Harvard Medical School) was performed on the conjugate. In order to evaluate the efficiency of ethylendiamine binding to mPEG-PLA (first reaction step, FIG. 6A) and TNP-470 binding to the polymer via the amine (second step, FIG. 6A), we used the colorimetric amine detection reagent: 2,4,6-Trinitrobenozene Sulfonic Acid (TNBSA) (Pierce, Rockford Ill.) was used. TNBSA reacts with primary amines to produce a yellow product whose intensity was measured at 450 nm. To calculate amine concentration in the polymer a linear calibration curve of amino acid was used. To measure TNP-470 loading into polymeric-micelles, we incubated 10 mg/ml Lodamin in 500 μl NaOH (0.1 N) to accelerate PLA degradation. After an overnight incubation with shaking (100 r.p.m) at 37° C., we added Acetonitrile to the samples (1:1 NaOH:Acetonitrile) and analyzed for TNP-470 concentration. TNP-470 concentration was measured using a high-performance liquid chromatography system (HPLC, System Gold® Microbore, Beckman Coulter). A 20-μl portion of each sample was injected into a Nova-pak C18 column (3.9-×150-mm i.d.; Waters), and analyzed using a calibration curve of TNP-470. TNP-470 binding to amine was also measured using TNBSA reagent and was confirmed by subtracting the non-bound drug from the total drug added to the reaction.
  • Amine Determination
  • The first step of ethylenediamine binding to the polymer, and the second step of TNP470 conjugation reaction through the free amine, were further verified using (2,4,6-Trinitrobenozene Sulfonic Acid (TNBSA) reagent (Pierce, Rockford Ill.). TNBSA is a sensitive assay reagent which can couple to free amino groups and form a highly chromogenic derivate. The reaction was carried out after the addition of 0.01% (w/v) TNBSA in 0.1M Sodium bicarbonate buffer. A series of PEG-PLA polymer concentrations before and after TNP-470 conjugation were allowed to react for 2 hr in 37° C. with TNBSA and the yellow color was measured using a plate reader.
  • The correlation between the molar amine concentration and color absorption at 450 nm was determined according to a standard curve done for aspartic acid.
  • Cell Culture.
  • Murine Lewis lung carcinoma (LLC) and B16/F10 melanoma cells were obtained from American Type Culture Collection (ATCC, Manassas, Va., USA). Human Umbilical Vascular Endothelial cells (HUVEC) were purchased from Cambrex (USA). The cells were grown and maintained in medium as recommended by the manufacturers. Dulbecco's Modified Eagle's Medium with 10% fetal bovine serum was used for tumor cells and EMB-2 (Cambrex Bio Science Walkersville, Inc.) containing 2% fetal bovine serum and EGM-2 supplements was used for HUVEC.
  • RPE cells were cultivated as has been previously described59. As determined by flow cytometry, the primary RPE cultures were found to be more than 98% cytokeratin positive (Clone PCK-26, Sigma). HMVECs, were purchased from Cambrex Bio Science Inc. (Rockland, Me.) and were maintained on collagen I coated plates with endothelial cell growth medium containing 2% FBS (EGM-2; Cambrex, Inc.) at 37° C. with humidified 95% air/5% CO2.
  • HUVEC Proliferation Assay
  • HUVEC were exposed to different concentrations of Lodamin equivalent to 50-1,000 nM free TNP-470 (0.12-2.4 mg/ml micelles) and incubated in a low serum medium for 48 h at 37° C. To rule out a possible cytotoxic effect of the carrier, empty micelles were added to HUVECs as a control (4.8 mg/ml). A WST-1 proliferation assay (Roche Diagnostics) was used. Cell viability was calculated as the percentage of formazan absorbance at 450 nm of treated versus untreated cells. Data were derived from quadruplicate samples in two separate experiments. The effect of Lodamin (60 nM TNP-470 equivalent q.o.d. on HUVEC growth rate was evaluated by daily counting of HUVEC cells up to 5 d and compared to the number of untreated cells or cells treated with Vehicle (same concentration as Lodamin).
  • Uptake of Polymeric Micelles by HUVEC and their Localization in Cells.
  • To evaluate the uptake of polymeric micelles by HUVEC, 6-coumarin labeled PEG-PLA micelles or rhodamine conjugated to mPEG-PLA were used. HUVEC were seeded in a 24-well plate (2×104 per well) in EGM-2 medium (Cambrex) and were allowed to attach overnight. Fluorescent-labeled micelles (10 mg/ml) were suspended over a bath sonicator for 5 min, and 20 μl of the suspension was added to the cultured cells. After the designated time points (20 min, 2, 4, 7 and 24 h), the cells were washed three-times with PBS and analyzed by flow cytometry, or alternatively fixed with 4% parformaldehyde. For confocal microscopy, cells were mounted using DAPI containing Vectashild (Vector, Laboratories, Burlingame, Calif.). Optical sections were scanned using Leica TCS SP2 AOBS a ×40 objective equipped with 488-nm argon, 543-nm HeNe, and 405-nm diode lasers. To study Lodamin internalization into endothelial cells, confocal microscopy was used to co-localize 6-coumarin labeled polymeric-micelles with endo-lysozome. Live HUVEC cells were imaged in different time points after addition of labeled micelles to cell media (15 μg/ml) up to 1 h. At this point, LysoTracker Red® (Molecular Probes, Eugene, Oreg.) was added to the medium for the detection of acidic intracellular vesicles: Endosomes and Lysosomes. After 20 min of incubation, cells were imaged by confocal using optical sections with 488-nm argon, 543-nm HeNe, and 405-nm diode lasers.
  • To further verify that polymeric micelle internalization occurs through endocytosis, cell uptake was measured in cold conditions (4° C.) in comparison to cell uptake at 37° C. HUVEC were plated at a concentration of 15,000 cells/ml in two 24-well plates for 24 h. Fluorescent labeled polymeric micelles (15 μg/ml) were added and incubated at different time points: 20, 30, 40 and 60 min (n=3) in cold room (4° C.) and in 37° C. After the designated time points the cells were washed 3 times with PBS and lysed with 100 μl lysis buffer (BD Biosciences). Cell extracts were measured for fluorescent signal in a Wallac 1420 VICTOR plate-reader (Perkin-Elmer Life Sciences) with excitation/emission at 488 nm/530 nm.
  • Morphology of MetAP-2 Inhibitor Polymeric Micelles by Transmission Electron Microscopy (TEM).
  • In order to study the morphology of MetAP-2 inhibitor polymersomes, Transmission Electron Microscopy (TEM) images were taken at the day of preparation and one week post preparation. Polymer micelles dispersed in double distilled water were imaged with cryo-TEM (JEOL 2100 TEM, Harvard University—CNS).
  • In-Vitro TNP-470 Release from MetAP-2 Inhibitor-Polymersomes.
  • To study the kinetic release of TNP-470, Lodamin (20 mg) was incubated with either 1 ml PBS pH=7.4 or simulated gastric fluid (HCL:ddw pH=1.2). Every few days, supernatant was taken and analyzed for TNP-470 concentration and a cumulative release graph of TNP-470 was determined. TNP-470 concentration was measured using HPLC. TNP-470 was detected as a peak at 6 min with 50% acetonitrile in water at the mobile phase. The flow rate was 1 ml/min, and the detection was monitored at 210 nm wavelength.
  • Size of MetAP-2 Inhibitor Polymeric Micelles
  • The average size and size distribution of polymeric micelles were determined by Dynamic Light Scatterer (DLS, DynaPro, Wyatt Technology). The measurements were done at 25° C. using Dynamic V6 software. Lodamin (TNP-470 polymersomes) at 1.5 or 2 mg/ml dispersed in double-distilled water were measured by 20 successive readings with DLS. Micelles' size was also measured after 24 hr to evaluate their structural stability.
  • Evaluation of Antitumor Activity of MetAP-2 Inhibitor Polymersomes In-Vivo
  • Animal procedures were performed in the animal facility at Children's Hospital. Eight-week-old male C57BL/6 mice (Jackson Laboratories, Bar Harbor, Mass.) were injected subcutaneously with 1×106 Lewis Lung Carcinoma cells (LLC) at the back. When tumors reached a volume of 100 mm3 the mice were divided into 5 groups, and treated every day for 12 days with TNP-470 polymeric micelles, administrated orally using a gavage needle. Different doses of polymersomes were given daily: 15 mg/kg (5 mice), 30 mg/kg (3 mice) and 60 mg/kg (1 mice) TNP-470 equivalent, respectively (5, 10, 20 mg polymeric drug per mouse, respectively). The control groups were five mice that were given the same dose of polymersomes without the drug, and five mice that were given drinking water.
  • Every two days the weight of the mice was monitored and tumors dimensions were measured using a caliper. Tumor volume was calculated according to an ellipsoid formula.
  • Absorption Kinetics of Polymeric Micelles in the Gastrointestinal Tract
  • A non-invasive IVIS 200 in-vivo imaging system (Xenogen system 3.0) was used in order to track the polymeric micelles labeled with fluorescent marker in a manner similar to the cell uptake study. Fluorescent polymersomes were given orally to three nude mice, the mice were anesthetized using an Isofluran chamber, and images were taken after different time periods. All the images were taken under identical conditions.
  • Corneal Micropocket Assay
  • In order to evaluate the antiangiogenic properties of Lodamin, the corneal micropocket angiogenesis assay was performed as previously detailed29. Pellets containing 80 ng carrier-free recombinant human bFGF or 160 ng VEGF (R&D Systems, Minneapolis, Minn.) were implanted into micropockets created in the cornea of anesthetized mice. Mice were treated daily with 15 mg/kg TNP-470 equivalent of Lodamin for 6 d, and then the vascular growth area was measured using a slit lamp. The area of neovascularization was calculated as vessel area by the product of vessel length measured from the limbus and clock hours around the cornea, using the following equation: Vessel area (mm2)=[π×clock hours×vessel length (mm)×0.2 mm].
  • Body Distribution, Intestinal Absorption and Toxicity of Lodamin
  • For all biodistribution studies we used a fluorescent marker for tracking Lodamin. Mice were administered 6-coumarin labeled mPEG-PLA by oral gavage for 3 d (100 μl of 1.5 mg/ml). On the third day of treatment, after 8 h of fasting, animals were sacrificed and spleen, kidney, brain, lungs, liver, intestine, stomach, and bladder were collected. The fluorescent 6-coumarin was extracted from the tissues by incubation with formamide for 48 h at 25° C. Samples were centrifuged and signal intensity of fluorescence of supernatants was detected with a Wallac 1420 VICTOR plate-reader (Perkin-Elmer Life Sciences) with excitation/emission at 488 nm/530 nm. The results were normalized to protein levels in the corresponding tissues. Tissue autofluorescence was corrected by subtracting the fluorescent signal of non-treated mouse organs from the respective readings in treated mice. Similarly, levels of fluorescent signal in mouse sera were measured in different time points (1, 2, 4, 8, 24, 48 and 72 h) using excitation/emission readings at 488 nm/530 nm.
  • In order to analyze cell uptake in the different tissues in tumor bearing mice, PEG-PLA-rhodamine micelles (100 μl of 1.5 mg/ml) or water were orally administered to C57B1/6J mice bearing LLC tumors (200 mm3) for 3 d. Organs were removed, incubated for 50 min in collagenase (Liberase Blendzyme 3; Roche Diagnostics Corp., IN) in 37° C. to obtain a single cell suspension. These suspensions were analyzed by flow cytometry in order to quantify the uptake of micelles into different tissue cells when compared to those in the untreated mouse.
  • In order to evaluate intestinal absorption, mPEG-PLA-rhodamine micelles were orally administered to C57B1/6J mice after 8 h of food fasting. After 2 h mice were sacrificed and 2.5 cm segments of the small intestine were removed, washed, and analyzed by histology and confocal microscopy. The rhodamine-labeled polymeric micelles were detected by confocal microscopy (Leica TCS SP2 AOBS) with a 488-nm argon laser line. Actin filaments were stained with phalloidin-FITC (Sigma) and nuclei were stained by DAPI (Sigma). To further study the uptake of Lodamin in the intestine, high resolution images were imaged with cryo-TEM. Intestines from treated (as above) and untreated mice were excised and immersed immediately in a freshly prepared 4% paraformaldehyde in PBS pH 7.4 for 2 h at 25° C. The samples were washed in PBS, transferred to a 30% sucrose solution overnight at 4° C. and embedded in OCT and kept at −80° C. until processing. Fifteen sections with 10 μm each were prepared and processed for Confocal microscopy or TEM. Four TEM samples were fixed for 30 min in freshly prepared 2% paraformaldehyde, 2.5% glutaraldehyde, 0.025% CaCl2 in 0.1 M sodium cacodylate buffer, pH 7.4 and subsequently postfixed for 30 min in 1% osmium tetroxide in 0.1 M sym collidine buffer, pH 7.4 at 25° C., stained en bloc in 2% uranyl acetate, dehydrated, and embedded under inverted plastic capsules. Samples were snapped free of the glass coverslips by a cycle of rapid freezing and thawing. Thin sections were cut en face with diamond knives using a LEICA UCT Ultramicrotome. Specimens were examined using a JEOL 2100 TEM.
  • To exclude tissue toxicity, histological analysis (H&E) of liver, intestine, lung and kidneys was conducted (Beth-Israel pathology department, BIDMC). To further exclude liver toxicity we analyzed serum levels of liver enzymes: ALT, AST (Done at Shriners Burns Hospital, Boston, Mass.). These studies were performed on 20 d-long Lodamin treated mice (15 mg/kg TNP-470 equivalent q.d.) and compared to untreated mice (n=3-4).
  • Oral Administration of Lodamin In-Vivo and Primary Tumor Experiments
  • Animal procedures were performed in the animal facility at Children's Hospital Boston using 8 week old C57B1/6J male mice (Jackson Laboratories, Bar Harbor, Me.).
  • For tumor experiments: LLC cells (1×106) or B16/F10 melanoma cells (1×106) were implanted subcutaneously in 8 week old C57B1/6J male mice (Jackson Laboratories, Bar Harbor, Me.). Oral availability of free TNP-470 was compared to that of Lodamin. A dose of 30 mg/kg q.o.d. of free TNP-470 and an equivalent dose of Lodamin (Lodamin, 6 mg in 100 μl/d) were administered to LLC tumor bearing mice (˜100 mm3) and tumor growth was followed for 18 d. Free drug was given as a suspension in double distilled water and freshly prepared before each dose. Additionally, we compared different doses and frequencies of Lodamin treatment: 15 mg/kg q.d., 15 mg/kg q.o.d. and 30 mg/kg q.o.d. To eliminate any possible effect of the vehicle (polymer without drug) one group of mice was given micelles without drug and tumor progression was compared to water treated mice.
  • For the melanoma tumor experiment, a dose of 15 mg/kg q.d. Lodamin was administered to B16/F10 melanoma bearing mice. In all experiments, tumor size and animal weight were monitored every 2 d. Tumor volume was measured with calipers in two diameters as follows: (width)2×(length)×0.52. Note that all the above Lodamin doses are presented as TNP-470 equivalent.
  • Oral Administration of Lodamin and Liver Metastasis Experiments
  • To examine the effect of oral Lodamin treatment on metastasis development and prevention, liver metastases were generated by spleen injection. C57B1/6J mice (n=14) were anaesthetized with Isoflurane and prepared for surgery. A small abdominal incision was made in the left flank and the spleen was isolated. B16/F10 tumor cells in suspension (50 μl, 5×105 in DMEM medium without serum) were injected into the spleen with a 30-gauge needle, and the spleen was returned to the abdominal cavity. The wound was closed with stitches and metal clips. After 2 d mice were divided into two groups, one was treated daily with oral Lodamin (15 mg/kg) using gavage and the second group was administered water by gavage. After 20 d, we terminated the experiment. Mice were killed and autopsied, livers and spleens were removed by surgical dissection, imaged, and histology was carried out. Liver and spleen tissues were stained with Hematoxylin & Eosin to evaluate tissue morphology and detect metastasis. Immunohistochemistry was carried out to specifically detect B16/F10 cells in the liver using anti-mouse melanoma antibody (HMB45, abCAM) and using DAB staining.
  • Evaluation of Neurotoxicity with Balance Beam Motor Coordination Test
  • A slightly modified balance beam motor coordination test was performed on three groups of mice: Oral Lodamin treated mice (30 mg/kg eq. q.o.d.), free TNP-470 (30 mg/kg) subcutaneously injected mice and water treated mice (administered by gavage). The mice were pretreated for 14 d (n=4-5 per group) and then the mice were allowed to acclimate to the procedure room for 1 h, after which they were trained in 3 trials to cross a wide (20 mm width×1 m length) balance beam. All the mice crossed the wide beam without making foot-slip errors. The mice were then trained on a narrow (4 mm width×1 m length) beam for 3 trials. At the end of the training trials, no freezing behavior was observed, and the mice would start to walk within 4 seconds of being placed on the beam. The mice were then videotaped as they performed in 3 test trials of 3 beam crossings each—a total of 9 crossings per mouse. The 3 trials were separated by at least 1 h to avoid fatigue of the mice. Videotaped crossings were scored for number of foot-slip errors and time to cross. All experiments and scoring of the different groups were performed by an investigator who has been no knowledge regarding the treatment regime of the mice.
  • MetAP-2 Inhibitor Polymersome Effect on Angiogenesis, Proliferation and Apoptosis in Tumor Tissues
  • Histologic evaluation of tissue was performed on 8 μm thick frozen sections of LLC tumors that were removed from two random Lodamin-treated or untreated mice 14 d post treatment (15 mg/kg q.d. TNP-470 equivalent). Tumors were sectioned and analyzed for cell markers, 20 microscope fields (×400) were imaged.
  • Tissues were stained with Hematoxylin & Eosin to detect tissue morphology.
  • Immunohistochemistry was carried out using Vectastain Elite ABC kit (Vector Laboratories, Burlingame, Calif.). Primary antibodies included CD31 (BD Biosciences, San Jose, Calif.) for microvessel staining and anti-Ki-67 (DAKO, CA, USA) detection of proliferation. Detection was carried out using a 3,3′-diaminobenzidine chromogen, which results in a positive brown staining. Apoptotic cells were detected by reacting the tissues with Terminal deoxynucleotide transferase mediated dUTP-biotin nick end labeling (TUNEL) using a kit (Roche) following the company's protocol. Vessels were detected in the same tissues by anti-CD31 and secondary FITC anti-mouse antibody (Jackson ImmunoResearch) conjugated antibody (green) and nuclei were detected by DAPI (blue).
  • Delayed Type Hypersensitivity (DTH) Reactions
  • DTH reactions were induced in the skin of 8-week-old C57B1/6J male mice as previously reviewed62.
  • Induction of CNV and Flat-Mount Preparation
  • Laser-induced CNV was generated by a technique described previously with some modifications63. Only lesions in which a subretinal bubble or focal serous detachment of the retina developed were used for the experiments. Lodamin treatments were started at the day of CNV induction and the sizes of CNV lesions were determined after 7 or 14 d. For this purpose, four burns were performed per eye while leaving a space around the optic disc. After 7 or 14 d of laser induction, the retinas were analyzed for blood vessel formation.
  • Analysis of Cell Population in Retina by Flow Cytometry
  • Cell populations in retina tissues were analyzed by FACS.
  • In-Vivo Miles Assay for Vessel Permeability
  • Miles assay was performed in C57B1/6J mice 5 d after oral Lodamin treatment (30 mg/kg/day). MCP-1 and VEGF-165 were used as inducers.
  • Laser Induced Retinal Vascular Permeability and Biodistribution Studies
  • In order to evaluate specifically the effect of Lodamin on laser-induced retinal leakage while excluding the anti-angiogenic activity of Lodamin, CNV lesions of the same size (same day post laser injury) were used to compare blood vessel permeability in mice and rats. Two different assays were performed; Angiography in live rats and modified Miles assay in mice. A modified Miles assay was performed as described in the Examples. Angiography was performed on rats using Fluorescein imaging as previously described64.
  • In order to evaluate biodistribution of Lodmain in CNV site, fluorescent labeled drug was used.
  • Electroretinography
  • Dark-adapted full-field ERGs were obtained as previously described65. By fitting the Hood and Birch formulation66 of the Lamb and Puch model67, 68 of phototransduction to the a-wave, we obtained the photoreceptor's kinetics (Srod) and saturated amplitude (RmP3). From the response vs. intensity relationship of P2, the putatively-pure postreceptor response derived from the b-wave, we determined bipolar sensitivity (log kP2) and saturated amplitude (RmP2). The oscillatory potentials (OPs) obtained after passing P2 through a 5th-order Butterworth filter (bandpass 50-250 Hz); were evaluated in the frequency domain69 to estimate maximal OP energy (Em). Finally, the retinal response to light-adapted 8 Hz flicker was measured (R8). ERG data were expressed as A Log Normal.
  • Analysis of Cell Population in Retina by Flow Cytometry
  • Single cell suspensions were prepared from C57BL/6J mouse retinas with CNV lesions. In order to collect a sufficient number of ocular-infiltrating cells for flow cytometry, 20 separate burns were delivered similar to the panretinal photocoagulation procedure in humans. The quantity of macrophages in single-cell suspensions from mouse retinas from days 3 and 7 post laser procedure was evaluated for Lodamin treated (oral, 30 mg/kg q.d for 3 days) or vehicle or untreated mice. In the third day of treatment mice were euthanized and analyses of their retinas were performed.
  • Statistical Analysis
  • In-vitro data are presented as mean±SD, whereas in-vivo data are presented as mean±SE. Differences between groups were assessed using unpaired two-tailed Student's t-test, and P<0.05 was considered as statistically significant.
  • Example 1
  • Block copolymer micelles (polymersomes) have been proposed for the delivery of hydrophobic drugs with low aqueous solubility (such as Paclitaxel). The amphiphilic nature of diblock copolymers such as poly (lactide)-poly(ethylene glycol) (PLA-PEG), enables the formation of micelles in aqueous media. In an aqueous environment, the diblock copolymer is self-assembled into a structure of hydrophobic core, formed by the association of the hydrophobic polymer and the drug, and a hydrophilic shell, formed by the hydrophilic polymer, commonly PEG.
  • Block copolymers as disclosed in U.S. Pat. No. 4,745,160 (which is incorporated herein by reference) have been used form water insoluble, amphiphilic, non-crosslinked linear, branched or graft block copolymers having polyethylene glycol as the hydrophilic component and poly(D-, L-, or D, L-lactic acids) as the hydrophobic components.
  • Block copolymers as disclosed in U.S. Pat. No. 5,543,158 (which is incorporated herein by reference) have been described to form nanoparticles or microparticles that are solid particles that are suspended in water that are formed from a water-insoluble block copolymer consisting essentially of poly(alkylene glycol) and poly(lactic acid). The molecular weight of the block copolymer is high and the copolymer is insoluble in water. In the nanoparticle or microparticle, the biodegradable moieties of the copolymer are in the core of the nanoparticle or microparticle and the poly(alkylene glycol) moieties are on the surface of the nanoparticle or microparticle in an amount effective enough to decrease uptake of the nanoparticle or microparticle by the reticuloendothelial system. Nanoparticles are prepared by dissolving the block copolymer and drug in an organic solvent, forming an oil and water emulsion by sonication or stirring, and collecting the nanoparticles containing the drug following precipitation. It does not provide for the solubilization of hydrophobic drugs.
  • The inventors demonstrate that MetAP-2 inhibitor TNP-470 was successfully conjugated to a modified PEG-PLA polymer through its amine, and formed nano-size polymersomes. A schematic diagram of the conjugation process is shown in FIG. 6A. Step I demonstrates the reaction between succinated mPEG-PLA and ethylendiamine that results in an amine terminated polymer. Step II demonstrates the reaction between the amine reactive polymer and the terminal chlorine of TNP-470. The conjugate is then dialyzed against water in an excess of TNP-470 to form polymeric micelles. The inventors demonstrate, using images taken by TEM, that spherical micelles were formed and size measurement with DLS showed a low range of size distribution around 10 nanometers. By using confocal microscopy images of fluorescent-labeled polymersomes, the inventors demonstrate rapid uptake by Human Umbilical Vein Endothelial Cells (HUVECs), and when TNP-470 polymersomes were added, a significant inhibition of HUVECs proliferation was shown (as compared to no effect of the carrier itself).
  • In-vivo studies done on mice showed a significant inhibition of subcutaneous Lewis Lung Carcinoma tumors with C57/BL mice given a daily oral administration of TNP470 micelles. A dose of 15 mg/kg TNP-470 equivalent showed 63% inhibition without any weight loss to the mice.
  • NMR results from the different reaction steps are shown in FIG. 1. The results show the binding of the ethylendiamine through the carboxic acid group at the original polymer, and the TNP-470 binding to the polymer through the amine.
  • Amine Determination.
  • To further verify the binding of TNP-470 to the modified PEG-PLA through the amine in the polymer and the terminal chlorine in the drug, a TNBSA reagent reaction for the detection of free amino groups was performed. According to the results (FIG. 2) the polymer was saturated with ethylendiamine. Correlation between molar amine groups in the modified polymer and absorption in 450 nm were determined by a theoretical value from a standard curve done for aspartic acid (not shown). Moreover, after TNP-470 conjugation, no free amines were detected, indicating that the polymer was saturated with TNP-470 conjugated through the amine group at the polymer.
  • Morphology of MetAP-2 Inhibitor Polymeric Micelles by Transmission Electron Microscopy (TEM).
  • In order to study the morphology of TNP-470-micelles, Transmission Electron Microscopy (TEM) images were taken at the day of preparation (data not shown).
  • Polymersomes dispersed in double distilled water were imaged with cryo-TEM (JEOL 2100 TEM, Harvard university—CNS). The images show that the self-assembled polymersomes had a uniform spherical morphology.
  • HUVEC Proliferation.
  • The effect of different concentrations of TNP-470 polymersomes on the proliferation of HUVECs was evaluated using a WST-1 reagent. FIG. 4 shows the inhibition in proliferation after 48 hr of incubation. FIG. 4A shows the effect of TNP-470 micelles, and FIG. 4B shows the effect of micelles without drug on HUVECs. A significant inhibition of HUVEC proliferation was detected from 0.3 mg/ml and up to 2.4 mg/ml micelles, which are 62.5 nM-1000 nM TNP-470 equivalent (88% and 95% inhibition respectively). The same concentrations of polymeric micelles without TNP-470, as well as double concentration (4.8 mg/ml) showed no significant effect on HIJVEC proliferation.
  • Example 2 Cellular Uptake Studies
  • In order to evaluate the kinetics of polymersomes uptake by HUVECs in in-vitro condition, HUVECs were incubated with fluorescent polymersomes, washed and imaged by Confocal microscopy. HUVECs were incubated with the micelles for 20 min, 4 hr and 24 hr. After 20 min. micelles were already uptaken by the cells and located at the cytoplasm (data not shown). After 2 hr, 4 hr, 7 hr and 24 hr the uptake increased and the micelles were detected as concentrated spots inside cell cytoplasm (data not shown).
  • In Vivo Effects of PEG-PLA-MetAP-2 Inhibitor Micelles.
  • C57/BL mice bearing s.c LLC tumors showed inhibition in tumor growth when treated with PEG-PLA-TNP-470 micelles given orally. All doses showed a significant effect already after 14 days of daily treatment, as shown in FIG. 5A. The controlled groups had to be scarified on day 14, and the treatment continued up to day 18. The lowest dose (15 mg/kg TNP-470 equivalent) showed a significant inhibition of tumor growth (63%) after 14 days (*p<0.05), without any weight loss to the mice. whereas higher doses of 30 mg/kg and 60 mg/kg TNP-470 equivalent showed 74%, 75% inhibition respectively, but also caused almost 20% weight loss (FIG. 5B).
  • Absorption of Polymeric Micelles in the Gastrointestinal Tract.
  • The non-invasive Xenogen system was used to track the absorption kinetics of the polymersomes after oral administration. The micelles were concentrated at the gastrointestinal tract within 10 min post administration (GI track) (data not shown). Apparently the absorption was rapid and after 1 hr fluorescent signal was found near the bladder and the prostate glands. After 24 hr the signal was still detectable.
  • Example 3 Chemical and Physical Characterization of MetAP-2 Inhibitor Polymersomes
  • In an effort to predict the oral availability of TNP-470, the hydrophobicity of Lodamin was measured using log-D parameter. The measured Log-D values were 2.39 at pH=2 and 2.57 at pH=7.4. The high Log-D values (>2) indicate a very low solubility in water. This property indicate that the designed Lodamin had with improved solubility and oral availability.
  • The formulation of Lodamin and the conjugation of TNP-470 are described in detail in the Methods. The chemical and physical properties of Lodamin was characterized. First, the binding of TNP-470 to mPEG-PLA was confirmed by 3H NMR (data not shown) and mass spectrometry showing an average m/z of 3687 for mPEG-PLA-TNP-470. FIG. 6A demonstrates the reaction of Lodamin preparation. Using amine detection reagent, the incorporation efficiency of ethylenediamine to mPEG-PLA was determined as 65%, and in the second step TNP-470 was shown to be bound in high efficiency of >90%. Lodamin contained 0.8-1% (w/w) free TNP-470 as determined HPLC. The average size and size distribution of Lodamin was determined by a DLS (FIG. 3) on the day of preparation and after 10 d of incubation in aqueous medium to evaluate Lodamin stability (n=4). The majority of micelles (90%) at day of preparation were 7.8-8 nm in diameter, with a small population of larger particles (200-400 nm). The size remained almost unchanged after 10 days.
  • The morphology of Lodamin was characterized by TEM. (FIG. 6B). The images showed that the polymeric micelles had acquired a uniform spherical structure, which remained stable after two weeks of incubation in water at 37° C. Since the drug is located in the PLA core of the micelle structure and PLA is spontaneously hydrolyzed in an aqueous environment, we studied the release kinetics of TNP-470 from Lodamin. A slow-release kinetic of TNP-470 was obtained following incubation in PBS (pH=7.4) or in gastric liquid (pH=1.2). The TNP-470 was released over a period of 28 d with an early peak burst of ˜30% after the first day of incubation (FIG. 6C).
  • Endothelial Cells Take Up MetAP-2 Inhibitor Polymersomes by Endocytosis
  • The uptake of polymeric micelles by HUVEC and its kinetics was evaluated initially. HUVEC were incubated with 6-coumarin labeled mPEG-PLA micelles for 20 min, 2, 4 and 7 h and were imaged by confocal microscopy. Confocal images show HUVEC uptake of polymeric micelles labeled with 6-coumarin after 20 min and 7 h incubation time periods (data not shown). In Live HUVEC cells as were imaged 1 h post the addition of labeled micelles to cell media (15 μg/ml, in green). LysoTracker Red® was used to detect Endosomes and Lysosomes. Overlay between micelles and Endo-lysosomes is represented in yellow/orange color (data not shown). As soon as 20 min after incubation, micelles were taken up by the cells and was located in their cytoplasm. After 2 h the uptake was maximal and after 4 to 7 h micelles were detected as defined aggregates inside the cytoplasm. Flow cytometry analysis of HUVEC incubated with rhodamine-labeled polymeric micelles for the same incubation times confirmed a maximal uptake after 2 h (FIG. 7A), while, no difference was observed between 2 to 24 h of incubation. In live-cell analysis, co-localization of Lyso-tracker staining with the micelles suggests endocytosis mechanism of uptake. Incubation of the micelles with HUVEC in cold conditions reduced micelle's uptake by up to 55%, confirming the endocytosis process (data not shown).
  • MetAP-2 Inhibitor Polymersomes Inhibit Proliferation of Endothelial Cells
  • Following polymeric-micelle uptake studies, the effect of Lodamin on the proliferation of endothelial cells was evaluated. Lodamin (62.5 nM-1,000 nM TNP-470 equivalent) inhibited HUVEC proliferation by 88% to 95% respectively after 48 h (FIG. 7B). The growth of HUVEC treated with Lodamin (60 nM TNP-470 equivalent) was completely inhibited compared to untreated cells or cells treated by vehicle only. No substantial cytotoxic effect was found (FIG. 7C).
  • MetAP-2 Inhibitor Polymersomes Inhibit VEGF and bFGF-Induced Angiogenesis In Vivo
  • The anti-proliferative and antiangiogenic properties of Lodamin were evaluated in-vivo by the corneal micropocket assay29. Mice were treated with daily oral Lodamin (15 mg/kg q.d.) or vehicle for 6 d. Newly formed blood vessels grew towards the bFGF pellet (data not shown) in representative eyes of untreated mice. Treated mice show the inhibition of bFGF induced angiogenesis in representative eyes. Quantification of the proliferative area (FIG. 7D) showed 31% inhibition of angiogenesis, compared to vehicle (P=0.00016, n=10). Similar results were obtained with VEGF165 (160 ng) induced angiogenesis in the cornea (data not shown), in this case Lodamin resulted in 40% inhibition of vessel area.
  • Polymeric Micelles: Absorption by the Intestine
  • To study the intestinal absorption of mPEG-PLA-rhodamine micelles, mice were administered oral polymeric-micelles. After 2 h mice were euthanized and isolated segments of the small intestine were fixed and imaged using confocal microscopy. A histological section of gut epithelial cells of Lodamin treated mouse was observed with TEM. The polymeric micelles were intensively taken up by columnar epithelium lining the luminal side of the small intestine. In a high magnification of small intestine villi, micelles are clearly detected in the lamina propria and in the vicinity of the blood vessels, indicating transepithelial absorption. In high resolution TEM images of single gut epithelial cells, many endosomes loaded with the drug were detected (data not shown). Microvilli (MV) structures and endosomes loaded with polymeric micelles (data not shown) are detected. Bar=500 nm (left) and 200 nm (right). Confocal microscopy image also confirms the TEM observations. Note the polymeric micelles can be detected in the lamina propria in the vicinity of blood vessels. The actin filaments were stained with phalloidin-FITC, nuclei were labeled with DAPI and mPEG-PLA were labeled with rhodamine, bars=5 μm. These vesicles were different in contrast and number than in an intestine of untreated mouse. These data support the premise that Lodamin was absorbed by the villous structure of the intestine by endocytosis.
  • Body Distribution of Polymeric-Micelles, Accumulations in Tumors and Toxicity
  • The biodistribution and tissue uptake of orally administered labeled MetAP-2 inhibitor polymersomes were studied by treating mice with fluorescent labeled PEG-PLA micelles for 3 d. After harvesting the tissue, tissue drug concentration was quantified by dye extraction or by flow cytometry. The results of the fluorescent dye extraction method (FIG. 8A) showed that as expected, a high concentration of fluorescent signal was detected in the stomach and the small intestine, where the liver showed the highest level. Importantly, the brain showed no presence of fluorescent signal. In the serum, labeled micelles were already detected after 1 h post oral administration, peaking after 2 h and were still detected up to 72 h. Accordingly, in tumor bearing mice, flow cytometry analysis of enzymatically digested tissues (FIGS. 8C and D) demonstrated a large uptake of labeled micelles by the liver, and no uptake by brain cells. Importantly, the highest uptake of micelles was detected in tumor cells. In FIG. 8C the percentage of FL2high cells, i.e. cells that absorbed rhodamine-labeled micelles is demonstrated. Taken together, these results indicate that the drug is mostly concentrated in the tumor and in the gastrointestinal organs (although to a lower extent) and not in the brain.
  • No tissue abnormalities were detected in histological analysis (H&E) of liver, intestine, lung and kidneys of MetAP-2 inhibitor polymersome treated mice (15 mg/kg TNP-470 equivalent q.d., for 20 d) when compared to untreated mice (data not shown). In addition, no significant differences were found in mouse serum liver-enzyme profile of MetAP-2 inhibitor polymersome treated mice compared to untreated mice. In Lodamin treated group ALT concentration was 41±9 u/l and AST was 120±39 u/l whereas in the untreated group the ALT concentration was 37.5±4 u/l and AST concentration was 152±131 u/l.
  • MetAP-2 Inhibitor Polymersomes Inhibit Primary Tumor Growth
  • The biological efficacy of Lodamin was evaluated as an anti-cancer agent in tumor bearing mice. When mice were given oral free TNP-470 at a dose of (30 mg/kg q.o.d.) no inhibition in the growth of subcutaneous LLC tumors was observe (FIG. 9A). The equivalent dose of Lodamin, however, resulted in a significant inhibition of tumor growth (FIG. 9A). This inhibition was observed after 12 d of Lodamin treatment, and at day 18 tumor growth was inhibited by 83%. Different dosing strategies of Lodamin were tested: 15 mg/kg q.d., 30 mg/kg q.o.d. and 15 mg/kg q.o.d., resulting 87%, 77%, and 74% of tumor volume inhibition respectively (FIG. 9B). The vehicle (PEG-PLA) showed no effect on tumor growth and was similar to untreated control mice (FIG. 9C).
  • In another tumor model, B16/F10 melanoma tumors were induced subcutaneously and their growth was also inhibited by oral Lodamin (15 mg/kg q.d.). This treatment was effective as of day 4, and after 13 d 77% volume inhibition was obtained (FIG. 9D). No apparent side-effects or weight loss were detected in either tumor models. Higher doses of Lodamin: 30 mg/kg q.d., and 60 mg/kg. q.o.d., showed substantial tumor inhibition, however, it was accompanied by weight loss (data not shown). FIG. 9E shows representative tumors of treated or untreated LLC or B16/F10 tumors.
  • MetAP-2 Inhibitor Polymersomes Did not Cause Neurotoxicity in Mice
  • Since the biodistribution study indicated that Lodamin does not cross the blood brain barrier, we further tested if the possible penetration of escaped free drug into the brain might still result in neurotoxicty and cerebellar dysfunction. To examine this, mice were subjected to a sensitive test of motor coordination: crossing a narrow (4 mm) balance beam 30. As shown in FIG. 9F Lodamin-treated mice (30 mg/kg q.o.d. for 14 d) performed in this challenge similar to control (water treated) mice, whereas mice injected with free TNP-470 (30 mg/kg q.o.d.) committed over twice as many errors (P<0.0001). These results indicate that Lodamin treatment avoids the cerebellar neurotoxicity observed with unconjugated TNP-470 treatment.
  • Oral MetAP-2 Inhibitor Polymersomes Inhibit Angiogenesis and Cell Proliferation in Tumors and Induce Tumor Cell Apoptosis
  • The effect of Lodamin on the histological structure of the LLC tumors was tested. Both treated and untreated tumors showed a dense cellular structure (data not shown). Lewis lung carcinoma (LLC) tumors were removed from Lodamin treated or untreated mice and sectioned. Tissues were stained with Hematoxylin & Eosin (H&E) to detect tissue morphology. Immunostaining with anti-CD31 was used to detect microvesselsand anti-Ki67 nuclear antigen for cell proliferation. TUNEL staining was used for the detection of apoptotic cells. Cell nuclei were stained with DAPI. Sections were counterstained with Eosin (nuclei). The tumors of untreated mice had a net organization of small and large vessels with apparent lumen structure as demonstrated by CD-31 immunostaining. In contrast, Lodamin treated tumors formed very small undeveloped vessels (data not shown). Lodamin treated tumors showed less cellular proliferation than untreated tumors, as detected by the nuclear marker KI-67. TUNEL staining for the detection of apoptosis in the tissues indicated an enhanced apoptosis in Lodamin treated tumors. Lodamin treated tumor had less vessels but high levels of apoptosis predominantly of tumor cells. In the control tissue, apoptotic cells were found in the capsule of the tumor but less in the center of the tissue.
  • In addition, the new oral formulation of the MetAP-2 inhibitor TNP-470 did not cause any weight loss or other apparent side-effects in the mice (FIG. 9G).
  • MetAP-2 Inhibitor Polymersomes Prevent Development of Liver Metastasis
  • The effect of oral Lodamin administration on liver metastases was tested after spleen injection of B16/F10. Oral Lodamin treatment dramatically affected development of liver metastasis. After 18 d of treatment mice were autopsied. All untreated mice had ascites while their enlarged livers had macroscopic malignant nodules and extensive cirrhosis, (FIG. 10A). In contrast, Lodamin-treated mice had no macroscopic metastasis in the abdomen and in the liver (FIG. 10A). Their organs had normal morphology and no weight-loss or other apparent side-effects were found. Immunohistology showed only a few sporadic B16/F10 cells in the liver, which had not developed into lesions (data not shown). Only one treated mouse out of seven had malignant nodules in its liver, but the liver was smaller than in the untreated control and had less cirrhosis. The spleens of all Lodamin treated mice had normal morphology compared to the congestion found in the enlarged spleens of control mice (FIG. 10B). Twenty days post B16/F10 cell injection into the spleen, 4 out of 7 control mice had died while all treated mice survived (FIG. 10C).
  • In this study, the inventors describe for the first time the development of a nontoxic oral formulation of the MetAP-2 inhibitor TNP-470, named Lodamin, as a potent anti-proliferative and anti-metastatic drug. TNP-470, a highly potent anti-proliferative/antiangiogenesis agent, is a leading candidate for chronic administration for cancer maintenance therapy and metastasis prevention.
  • The challenge that the inventors faced was based on the fact that TNP-470 has very poor oral availability as illustrated by its high Log D values, indicating low water solubility35. In order to make the drug suitable for oral administration, this property of TNP-470 was altered while retaining its activity by conjugating it with mPEG-PLA to form mPEG-PLA-TNP-470 polymeric micelles i.e. Lodamin. Unlike TNP-470 which is only dissolvable in organic solvents, Lodamin can be suspended in water to form polymeric micelles caused by the amphiphilic nature of PEG-PLA26. In this structure the drug is located in the core of the micelle protected from the harsh gastrointestinal environment36. Polymer micelles have previously been used for the delivery of hydrophobic drugs37, 38 and gene delivery39. Lodamin acquired a stable spherical morphology of nanomicelles, as imaged by TEM. In addition, PLA which is a biodegradable and biocompatible polymer, hydrolyzes in an aqueous environment and allows a slow-release of TNP-470. In-vitro studies showed a continuous release of TNP-470 from Lodamin over almost a month period, where the majority of the drug was released after 4 d (in both gastric and plasma pH conditions), although acidic environment is known to accelerate degradation of PLA we observed only a minor effect on day 15. While not wishing to be bound by theory, one possible explanation may be the masking effect of the PEG shell which delays the water penetration to the PLA core and slows the release of the drug through diffusion of this layer. In culture, Lodamin was rapidly taken up by endothelial cells via endocytosis and retained the original antiangiogenic activity of the free TNP-470 as demonstrated by the inhibition of HUVEC proliferation and growth rate.
  • PEG-PLA micelles penetrated the gut epithelial layer into the submucosa as shown by using fluorescent marker. The mechanism of Lodamin penetration to gut epithelial cells seems to be via endocytosis as detected by high resolution TEM imaging. In serum, labeled micelles had long blood circulation time of at least 72 h post administration, a significant increase compared to free drug which was detected in mice sera up to 2 h post-administration. Biodistribution showed relatively high concentration in the liver because oral administration delivers the drug directly from the intestine to the liver. However, no liver toxicity was observed by histology and by liver enzymes profile after 20 d of daily Lodamin treatment.
  • Lodamin given orally to mice showed substantial anti-proliferative effects (83% reduction), while free TNP-470 given orally had no effect. The effect on LLC growth was dose dependent, as 30 mg/kg q.o.d. was more effective than 15 mg/kg. q.o.d., and a dose of daily 15 mg/kg q.d. was more effective than 30 mg/kg q.o.d. (a double dose given every 2 d). A similar anti-proliferative effect of Lodamin was also observed with melanoma B16/F10 tumors, confirming the broad biological effect of Lodamin, very much like the original free TNP-470. Immunohistochemical studies carried out on LLC tumor tissues showed a reduction of proliferation and angiogenesis induced by Lodamin. Using TUNEL staining we detected a high level of tumor cell apoptosis following the Lodamin treatment, whereas in the controls much of the apoptosis occurred on the capsule of the tumor. These results indicate that the prevention of angiogenesis by Lodamin leads to tumor cell apoptosis, thus making Lodamin a cytotoxic drug, in addition to an antiangiogenic drug.
  • One of the most notable effects of oral Lodamin is the prevention of liver metastasis in mice. Liver metastasis is very common in many tumor types and has often been associated with poor prognosis and survival rate. The inventors chose the intrasplenic model for induction of liver metastasis, in which the transition time of B16/F10 from spleen to liver microvasculature post spleen injection in mice was found to be very fast (20% of the injected cells after 15 min) 40. Mice injected with B16/F10 cells into the spleen had a low survival rate. They developed ascites, macroscopic malignant nodules, and extensive cirrhosis in their livers 20 d post injection. However, all oral Lodamin treated mice survived up to this point and had normal liver (except one mouse) and spleen morphology without any other apparent side-effects. Immunohistology of treated mice's livers showed some rare sporadic B16/F10 cells which remained dormant over the 18 d of oral Lodamin treatment, compared to large metastasic tumors found in the untreated mice. It should be noted that Lodamin's effect on secondary tumor growth is in addition to its effect on the primary tumor.
  • While not wishing to be bound by theory, this dramatic effect may be due to the oral route of administration in which Lodamin is absorbed in the gastrointestinal tract and concentrated in large quantities in the liver via the portal vein. These results indicate that Lodamin can prevent the development of metastasis in the liver associated with different tumor types. Importantly, this property of the polymeric micelles might be used for enabling oral availability of other antiangiogenic or anti-cancer drugs to target liver metastasis.
  • In summary, oral Lodamin is therapeutically effective in the treatment of solid tumors and metastasis in mice. It captures the antiproliferative properties of free TNP-470 while adding important advantages: oral availability, improved biodistribution, tumor accumulation, continual release, improved solubility, proper clearance, and low related side-effects. In addition to use for the treatment or suppression of known, existing tumors, its use is contemplated, among others, for cancer patients as a long-term maintenance angiogenesis inhibitor to prevent tumor recurrence. Furthermore, it is also contemplated for use as maintenance therapy for chronic MetAP-2-related diseases such as age-related macular degeneration, endometriosis and rheumatoid arthritis. Other contemplated uses are described elsewhere herein.
  • Example 4 MetAP-2 Inhibitor Polymersomes Inhibit Glioblastoma Tumor Growth
  • The effect of Lodamin on human brain tumor (U87MG, Glioblastoma) was studied using subcutaneous tumor-bearing nude mice. Treatment with Lodamin started when tumors were in volume of ˜100 mm3. Lodamin that was given by gavage (FIG. 11A) in a daily dose of 15 mg/kg TNP-470 equivalent, inhibited tumor volume by 70% after 30 days. When Lodamin was given in drinking water at the same dose (FIG. 11B) a 71% inhibition of tumor volume was obtained after 42 days.
  • Example 5 MetAP-2 Inhibitor Polymersomes Prolong Survival of Mice Bearing Lung Metastasis
  • C57/B1 mice were injected into their tail-veins with 2.5×105 B16/F10 cells. Day after injection the mice were divided into 3 groups: 10 mice were given a daily oral administration of Lodamin (15 mg/kg TNP-470 equivalent), 9 mice were given IP injection with the same Lodamin dose, and 10 mice remained untreated. Mice survival was followed up to 40 days post treatment.
  • Lodamin treated mice showed a prolonged survival as can be seen in Kaplan-Meier survival curve (FIG. 12). After 32 days all control mice died, compared to 6 and 5 mice that lived at day 40 in the IP Lodamin treated group and Oral Lodamin group respectively.
  • Example 6 MetAP-2 Inhibitor Polymersomes Inhibit Angiogenesis in Matrigel Angiogenesis
  • In-Vivo Matrigel Angiogenesis Assay was conducted in order to assess the antiangiogenic properties of Lodamin. Matrigel (BD bioscience) which was remained in 4° C. overnight was mixed with fibroblast growth factor-2 (FGF-2) to obtain a final FGF-2 concentration of 1 μg/ml. Nine C57/B1 mice were injected with Matrigel (500 μl) in two sites subcutaneously. Three mice got a daily oral treatment with Lodamin (15 mk/kg/day TNP-470 equivalent). In three mice the Matrigel was mixed with Lodamin (5 mg/ml) and no further treatment was performed and three untreated mice with Matrigel implant were used as a control.
  • After 8 days, mice were euthanized and the Matrigel implant harvested, washed twice with PBS, and then one of the implant was immediately frozen on dry ice and used for histology assessment of blood vessels using CD-31 antibody. The second implant was enzymaticaly digested using collagenase (Librease, Roche) to obtain single cell suspension which again was analyzed by FACS to quantify the number of endothelial cells using CD-31 and CD-45 antibodies (endothelial cells are CD31+CD45−).
  • The implants of Lodamin treated mice, both the one with the mixture of matrigel and Lodamin or the mice treated orally with Lodamin, showed a significant reduction in blood vessels inside the implants (FIG. 13A). Histology showed less vessels in Lodamin treated mice and less dense tissue. FACS analysis suggests 70% reduction in the percent of endothelial cells which were attracted to the implant (FIG. 13B).
  • Example 7 Broad Spectrum Antiangiogenic Treatment for Ocular Neovascular Diseases Introduction
  • Ocular neovascular-related diseases are associated with vision impairment or vision loss in millions worldwide. The growth of abnormal, leaky blood vessels is a prominent component of several debilitating eye diseases including AMD, proliferative diabetic retinopathy (PDR), and retinopathy of prematurity (ROP). Thus antiangiogenic strategies could be particularly beneficial in preventing and treating the progression of these diseases. Corneal neovascularization is often the result of inflammation, chemical burns, and conditions related to hypoxia48, 49. These conditions are currently treated by indirect angiogenesis inhibitors such as steroids and immunosuppressants48. Efforts to inhibit retinal neovascular disease such as ARMD have focused on the angiogenic factor Vascular Endothelial Growth Factor (VEGF) and such therapies are now FDA approved. The most prominent class of anti-VEGF therapy is represented by ranibizumab and bevacizumab. These molecules are derived from a humanized anti-VEGF antibody which has the ability to reduce the retinal edema in ARMD50. However, the effects of these therapies are short lived and thus repeated intravitreal injections every 4 weeks is necessary51. While clinical trials have shown a beneficial effect in 30-40% of treated patients50, 51, in many cases these therapies become less effective over time52 and the long-term safety is still debated.
  • In the recent years it has become clear that diverse cellular processes have been implicated in pathogenesis of retinal diseases, such as ARMD or PDR. These processes include oxidative stress, vascular permeability, fibrosis, inflammation and impaired function of RPE53. Given the multifactorial nature of neovascularization and the contribution of the microenvironment, the focus on more effective therapies to block CNV has shifted toward targeting multiple processes rather than a single pathway54.
  • In this study it was demonstrated that a broad spectrum antiangiogenic drug can be especially beneficial in inhibiting CNV by blocking vessel growth, reducing vascular leakage and modifying the neovascular stroma via suppression of inflammation. This type of therapy thus offers an improvement over current treatments and an alternative where existing therapies fail.
  • Lodamin is a novel polymeric antiangiogenic drug and the first oral formulation for TNP-47055. Lodamin is a conjugate of TNP-470 and monomethoxy polyethyleneglycol-polylactic acid (mPEG-PLA) polymer that forms a micelle-like structure when introduced in aqueous solution. The active molecule, TNP-470, is a synthetic analogue of fumagillin and a methionine aminopeptidase-2 (MetAP2) inhibitor. The broad spectrum activity of TNP-470 was demonstrated in more than a hundred preclinical animal tumor studies. In clinical trials, some cancer patients demonstrated partial or complete responses56, 57. However, TNP-470 also demonstrated poor pharmaceutical properties and dose-limiting reversible cerebellar side effects58. Therefore Lodamin was rationally designed to overcome these limitations. It acquires superior properties such as: water solubility, improved oral availability, prolonged half life, slow-release properties and minimal TNP-470-releated cerebellar side effects55. Lodamin had potent antiangiogenic effects in murine tumor and metastases models.
  • Using the CNV model in mice and rats, the inventors show that Lodamin is a first-in-its-class polymeric broad-spectrum antiangiogenic ophthalmic drug that targets numerous molecular pathways in addition to VEGF. Lodamin's unique chemical properties, combined with its mechanism of action, place it as a leading candidate for treating corneal and retinal neovascular diseases.
  • Results Lodamin Inhibits Angiogenesis and Macrophage Recruitment In Vivo
  • To investigate the effect of Lodamin on angiogenesis and macrophage recruitment, the mouse Matrigel angiogenesis assay was first performed. Angiogenesis in the Matrigel was visually assessed after removal of the plugs 7 days post Matrigel injection (FIGS. 16A and 16G) and by immunohistochemical staining of blood vessels using CD31 antibodies (FIG. 16A). Immunohistology revealed that Lodamin dramatically reduced angiogenesis compared with untreated mice which presented numerous large blood vessels with open lumen structures (n=5). Infiltrated endothelial cells and macrophages in Matrigel matrices were quantified in a single-cell suspension by fluorescence-activated cell sorting (FACS) (FIG. 16C). After 7 days of Lodamin treatment, significant reductions of infiltrating endothelial cells and macrophages were seen in the Lodamin treated group compared to vehicle treated mice (endothelial cells: 0.11±0.06% versus 0.49±0.11%; macrophages: 30±7% versus 43±9% in Lodamin and control groups, respectively).
  • FIG. 16H shows the immunohistological analysis of mice ears post DTH reaction: staining of macrophages using F4/80 marker. Control ears exhibited an excessive inflammation. Bar=50 p.m. No obvious difference was found in macrophage distribution; however, the total number of macrophages present in control mice was elevated due to greater amount of tissue associated with increased swelling.
  • Systemic and Local Lodamin Treatment Inhibit Corneal Neovascularization
  • Antiangiogenic activity of Lodamin following topical or systemic administration was evaluated in the mouse corneal micropocket assay using bFGF-induced neovascularization. In Lodamin treated mice, corneal vessel area was reduced by 38% relative to the control group after subconjunctional injection (30 mg/ml q.d for 5 days), 30% after once daily eye drops (30 mg/ml q.d for 5 days) and 37% after oral administration (30 mg/kg/d q.d for 5 days). FIG. 17B shows representative images taken from the different mouse groups.
  • Lodamin reduces time course of delayed-type hypersensitivity DTH ear-swelling
  • Lodamin's effect on angiogenesis, inflammation, vascular hyperpermeability and edema were evaluated in the DTH reaction. C57B1/6J mice were sensitized with oxazolone and treated with Lodamin orally (15 mg/kg q.d.) starting from 5 days after the initial exposure to oxazolone. A significant reduction of ear erythema and swelling was obtained in Lodamin-treated mice compared with vehicle treated mice (FIGS. 16D & 16F). Histology sections stained by Hematoxillin and Eosin (H&E) confirmed a markedly reduced swelling and edema in treated mice compared to untreated mice that presented thick epidermis with extensive edema demonstrated by cell spongiosis, high cellularity and infiltrating monocytes (FIG. 16D). A chronic response consisting of neutrophil accumulation and enhanced fibroblast proliferation was also detected in the untreated tissues. In some samples, acute inflammation associated with focal polynuclear cell accumulations was found. Immunohistochemistry of blood vessels (CD31) revealed a significant reduction of angiogenesis by Lodamin. Some reduction of infiltrated leukocytes and macrophages was observed in Lodamin treated mouse ears stained with CD45 and F4/80 (data not shown).
  • Oral and Intravitreal Treatment of Lodamin Reduces CNV Progression
  • The effect of Lodamin on CNV progression in mice was determined after oral or intravitreal administrations. Oral Lodamin was administered in different doses and duration: 15 mg/kg/day (for 7 days or 14 d of treatment) and 30 mg/kg dose (7 days). CNV lesions in choroidal flat-mounts were evaluated after staining for blood vessels. CNV area was significantly reduced by Lodamin treatment in all treatment groups. Higher doses and longer treatment led to an enhanced effect: 14 day treatment (15 mg/kg q.d.) led to 70.6% inhibition whereas 7 day of treatment at the same concentration led to a 27.2% inhibition. At 30 mg/kg/day a more dramatic inhibition was obtained (50.5%) after 7 day. FIG. 18A summarizes the CNV area of all treatment groups. H&E staining revealed that Lodamin substantially reduced the formation of fibrovascular response from the choroid induced by the spot rupture of Bruch's membrane by the laser (FIG. 18B). Recruitment of macrophages into CNV lesions over time was evaluated by FACS in single-cell suspension originated from retina tissues following Lodamin treatment (30 mg/kg/day, oral). Retinal infiltrated macrophages were reduced after Lodamin treatment in mice (control group: 2% and 3.6%, Lodamin treated group: 1.8% and 2.3%, on day 3 and 7 respectively) (FIG. 18C). When Lodamin was introduced by a single intravitreal injection using 100 μg and 300 μg Lodamin, a 56% or 75% suppression was obtained respectively when measured after 14 days. No significant difference was found when injecting PBS compared to vehicle (data not shown). FIG. 19 shows representative CNV lesions from the different groups and their size measurements. No toxicity was observed in histological H&E whole eye samples (data not shown).
  • Lodamin has Minimal Adverse Effect on Retinal Function
  • To further evaluate possible adverse side effects of Lodamin treatment, retinal function was assessed by electroretinography (ERG) after Lodamin treatment. Normal C57BL/6J mice were injected intravitreal with the highest dose studied of Lodamin (300 μg; n=10) or vehicle (n=10) and were tested 14 days after injection. Responses to an ˜6.6 log unit range of intensities of full-field flashes presented to the dark-adapted eye were recorded (FIG. 19B) as was the response to an 8 Hz flickering light presented in the presence of an adapting background (FIG. 19C). Rod photoreceptor response sensitivity (Srod) and saturating amplitude (RmP3) were derived from the ERG a-wave and postreceptor sensitivity (kP2) and saturating amplitude (RmP2) were derived from the ERG b-wave (FIG. 19D). The saturating energy in the ERG oscillatory potentials (Em), and the trough-to-peak amplitude of the flicker response (R8) were also determined. For each of the six ERG parameters, in log scale, the mean and 5th and 95th prediction limits for normal were calculated and compared to those of Lodamin-treated mice (FIG. 19D); Nearly every observation (56 of 60) in Lodamin-treated mice fell within the normal range. Only RmP3 differed significantly (P=0.46) between groups, by <0.15 log unit. These ERG data confirm the safety of Lodamin with respect to retina function. Lodamin drug function was further confirmed by histological analysis (FIG. 19E). No apparent retinal tissue changes were detected, both eyes presented normal structures of (1) choroid (2) photoreceptors (3) outer nuclear layer (4) outer plexiform layer (5) inner nuclear layer (6) inner plexiform layer and (7) ganglion cell layer. The total retinal thickness remained unchanged.
  • Lodamin Reduces Pro-Angiogenic and Inflammatory Factors in CNV Bearing Mice
  • Levels of different pro-inflammatory and pro-angiogenic factors were measured using specific antibody by enzyme-linked immunosorbent assay (ELISA) or western blot analysis. Significant reductions of MCP-1 and VEGF in retina-choroids tissues were found in Lodamin treated mice by ELISA. MCP-1 was not detectable in naïve mice but was found in high concentrations in CNV bearing mice: 486±3 pg/mg in control CNV mice and 61% lower level in Lodamin treated mice (FIG. 20A). Unlike MCP-1, VEGF baseline levels in naïve mouse eyes were almost as high as in CNV bearing mouse (313 pg/mg, 320 pg/mg respectively) compared to Lodamin treated mice that reduced VEGF levels 24% below the baseline (243 pg/mg). MCP-1 levels in primary RPE cultures were also measured by ELISA and was found to be reduced by Lodamin in a dose dependant manner after 24 h treatment (39%, 55%, 83% reduction was obtained with 1, 10, 100 nM Lodamin, respectively). All ELISA data were normalized to total protein in each sample (FIG. 20B).
  • MMP-2 and 9 activities in CNV lesions were determined following oral Lodamin treatment (30 mg/kg q.d for 7 d) using zymograms (FIG. 20D). The enzymatic activity was accelerated in tissues with CNV compared to naïve mouse tissues whereas both MMP-2 and 9 was reduced by Lodamin to the levels of naïve mice. The levels of TNFα, a major inflammatory cytokine, were higher in choroids than in retina and were reduced in the choroid by Lodamin (FIG. 20C). NF-kB, a pro-inflammatory transcription factor induced by TNFα, was detected in both retina and choroid tissues and was also reduced in the choroid by Lodamin. Lodamin reduced mitogen-activated kinases (MAPK) and its phosphorylated form in the choroid with no effect in the retina (FIG. 20C).
  • A dramatic reduction of RAGE levels (35 kDa isoform) were detected in both choroids and retinas of Lodamin treated mice. HMGB1 (RAGE ligand) was detected in the retina (29 kDa) but no difference was found between the groups. A ˜50 kDa band, possibly corresponding to a multiprotein complex, was detected in the choroid and was reduced by Lodamin. In vitro RAGE and HMGB-1 levels in microvascular endothelial cells (HMVEC) were reduced in a dose dependent manner by TNP-470 treatment (0.1-100 nM) (FIG. 20E).
  • Lodamin Reduces MCP-1 and VEGF Induced Vessel Permeability
  • The investigators further investigated whether MCP-1 has a role in Lodamin's effect on vessel permeability. In the in vivo Miles assay, MCP-1 enhanced vessel permeability at concentrations of 10 pg/ml (n=15, FIG. 21B). Representative mouse skin patches showing Evans blue leak in MCP-1 injection spots are presented in FIG. 21B. The effect of Lodamin on MCP-1-induced vessel leakage was measured after 5 d of oral treatment with 30 mg/kg/day in C57B1/6 mice. Lodamin significantly reduced vessel permeability by 87%. Similarly, a standard Miles assay was performed with VEGF demonstrating 65% reduction of vascular leakage after the same oral treatment of 5 d.
  • Lodamin Reduces Ocular Vascular Permeability
  • In order to evaluate the effect of Lodamin on ocular vascular leak while excluding direct angiogenesis inhibitory effects, CNV lesions of the same stage were used to compare vessel leakage in mice and in rats. Laser-induced CNV in rat retinas were imaged by fluorescein angiography (n=6). Rats with established CNV lesions (7 days after laser injury) were treated with Lodamin once 3 h before measuring the leakage (60 mg/kg). The lesions were graded by levels of leakage in laser spots and showed a significant reduction of retinal vessel permeability by Lodamin (data not shown). A modified Miles assay was also performed to evaluate Lodamin's effect on ocular vessel permeability in mice with an established CNV lesion. Similarly, when Lodamin (60 mg/kg, n=5) was administered once 3 h before Evans blue injection, 65% reduction in vessel leakage was obtained by this short-term Lodamin treatment (FIG. 21C).
  • Polymeric Micelles Accumulate in CNV Site
  • The biodistribution of fluorescent-labeled Lodamin in CNV lesions was determined 3 h post tail vein injection in C57B1/6J mice. A model fluorescent hydrophobic small molecule (6-coumarin, MW=350 gr/mole, Sigma) was used to determine differences between accumulations of the free molecule and its encapsulated form. Blood vessels stained with rhodamine concanavalin A are found in the periphery of the lesion. 6-coumarin dye is detected in the center of the lesion that is devoid of blood vessels. Compared with the free molecule, labeled Lodamin was detected in greater extent in CNV sites confirming a passive targeting (n=3) (data not shown).
  • Many small-molecule angiogenic inhibitors, such as TNP-470, have poor water solubility necessitating the development of unique formulations to facilitate their use. In general, particulate drugs offer improved ocular drug delivery since they are usually more stable than other colloidal systems. Lodamin comprises many of the requirements for an advanced ocular delivery: it can be introduced into an aqueous solution, is nontoxic, biocompatible, biodegradable, and has slow-release properties55. This is the first report of Lodamin treatment for retinal neovascular disorders.
  • RPE cells play a central role in the progression of AMD disease. These cells are responsible for the generation and maintenance of the extracellular and photoreceptor matrices, as well as for the integrity of Bruch's membrane. Impaired function of RPE, occurs in ARMD and causes lipid and protein accumulation in the vicinity of Bruch's membrane and formation of drusen70. Furthermore, RPE cells secrete chemoattractant and inflammatory cytokines such as interleukin-8 (IL-8), MCP-1, and TNFα. As a result, neutrophils and macrophages are recruited and stimulate angiogenesis which contributes to ARMD progression71.
  • The data demonstrate the suppressive effects of Lodamin on vascular leakage and inflammation, in addition to direct antiangiogenic effects. This was found to be partly due to the substantial reduction of MCP-1 production in CNV lesions and in RPE cells. MCP-1 is a small cytokine of the C-C chemokine family and is a potent chemoattractant for neutrophils and macrophages with proangiogenic properties. MCP-1 has been shown to be elevated in the vitreous of patients with retinal neovascularization71, 72 and to be involved in other systemic inflammatory and autoimmune diseases such as diabetes, rheumatoid arthritis and multiple sclerosis73, 74. The data show that the suppression of MCP-1 in the CNV site by Lodamin was accompanied by a reduction of infiltrating macrophages. This is very important for ARMD treatment since macrophages express TNFα in CNV which stimulates further secretion of MCP-1, IL-8, and TNFα from RPE cells, thus leading to a feedback loop with additional recruitment of macrophages75 and ARMD progression71, 76.
  • The Miles assay data demonstrated that MCP-1 induced vascular leak in a dose dependant manner. Surprisingly, Lodmain was able to antagonize MCP-1 mediated vascular leak, similar to its antagonism of VEGF induced leakage. These results provide an explanation for the significant reduction of edema demonstrated in DTH reaction, and of the reduction in retinal edema as analyzed by angiography and Evans blue extraction. Moreover, the reduction of retinal edema by Lodamin was very rapid, where only one pretreatment with oral Lodamin 3 h before performing the assay was sufficient to substantially reduce vessel permeability.
  • Lodamin effects were not limited to the suppression of MCP-1. Other pro-angiogenesis and pro-inflammatory factors, including VEGF, nuclear factor k-B (NFκB), TNFα and matrix metalloproteinases (MMPs), were also suppressed in situ by Lodamin treatment. Since NFκB regulates MCP-1 and is involved in the immune response77 stimulated by TNF-α and IL-6, suppressing this pathway can potentially lead to a substantial reduction in inflammation and angiogenesis and thereby prevent the progression of ocular neovascular diseases.
  • It is shown here for the first time that TNP-470 is linked to the RAGE pathway, which plays a pivotal role in retinal diseases such as diabetic retinopathy and AMD78, 79-81. RAGE, a multiligand receptor of the immunoglobulin superfamily, and its ligands: advanced glycolyzation proteins (AGE), high-mobility group box 1 (HMGB-1) and S100 protein family, are directly involved in inflammation, vascular and autoimmune diseases, diabetes, Alzheimer, cancer and angiogenesis82-88. Protein analysis of CNV tissues revealed a dramatic reduction of the 35 kDa isoform of RAGE (the secreted isoform of RAGE described previously89, 90) in retina and choroid tissues from CNV lesion treated by Lodamin. No detectable difference in the full size RAGE in the retina was detected. In HMVEC cells, a dose dependant reduction of the full size receptor RAGE and its ligand HMGB-1 were obtained with TNP-470 (0.01-100 nM). HMGB-1 (Amphoterin) is a low molecular weight protein with intracellular and extracellular regulatory activities that is known to have pro-inflammatory and pro-angiogenic properties87, 91. It has been demonstrated, in different models, that RAGE ligands can induce a sustained activation of NFκB via MAPK activation (e.g. ERK/p-ERK), enhance MCP-1 and TNFα production, upregulate VEGF in RPE, and increase MMPs activation84, 92-99. All these factors were shown to be suppressed by Lodamin in this CNV model, suggesting the possibility of additional indirect effects following the suppression RAGE activation.
  • In previous reports it has been shown that S100A4, which is a RAGE ligand and a member of S100 family, can covalently bind MetAp2100, a molecular target of TNP-470. S100 is a family of nonubiquitous cytoplasmic Ca2+-binding proteins that are expressed in a wide variety of cell types and are linked to many human pathologies101-103. The reduction of RAGE by TNP-470 provides a link between the known interactions of S 100-RAGE104, 105, S100-MetAp2103 and TNP-470-MetAp2106. It is likely that MetAp2 is associated with RAGE signal and TNP-470 suppresses this signal in a direct or indirect way.
  • The current treatment for ARMD is primarily based on VEGF targeting. Because Lodamin can target broader molecular pathways, it may potentially improve therapeutic outcomes. It is well established that many factors are playing a central role in the progression of the diseases in addition to VEGF. In addition, the specific structure of Lodamin, as a polymeric nanomicelle, enhances its accumulation in CNV lesions compared to the injection of free small molecule (as seen in FIG. 21) suggesting an ideal biodistribution via systemic administration. This effect may be due to the Enhanced Permeability and Retention effect (EPR)107 caused by the leaky vasculature in neovascular sites. Lodamin was well tolerated and showed minimal toxicity in the retina as indicated by histological analysis and ERG. Lastly, the slow release properties of Lodamin may minimize the dose and frequency of treatments, which is particularly important for intravitreal therapies. The current study therefore supports the fact that Lodamin is an excellent candidate for therapeutic treatment of ocular neovascular diseases in humans.
  • The references cited herein and throughout the application are incorporated herein by reference.
  • Although the foregoing invention has been described in some detail by way of illustration and example for the purposes of clarity of understanding, one skilled in the art will easily ascertain that certain changes and modifications may be practiced without departing from the spirit and scope of the appended claims.
  • REFERENCES
    • 1. Holmgren, L., O'Reilly, M. & Folkman, J. Dormancy of micrometastases: balanced proliferation and apoptosis in the presence of angiogenesis suppression. Nat. Med. 1, 149-153 (1995).
    • 2. Naumov, G., Folkman, J. Strategies to prolong the nonangiogenic dormant state of human cancer. in Antiangiogenic cancer therapy, Edn. 1. (ed. W. Darren, Herbst, R S, Abbruzzese, J L.) 3-23 (CRS press, Taylor and Francis group, 2007).
    • 3. Ingber, D. et al. Synthetic analogue of fumagillin that inhibit angiogenesis and suppress tumour growth. Nature 348, 555-557 (1990).
    • 4. Folkman, J., Kalluri, R. Tumor angiogenesis. in Cancer Medicine Vol. 1. (ed. D. Kufe, Pollock, R E., Weischselbaum, R R., Bast, R C., Gansler, T S., Holland, J F., Frei, E.) 161-194 (Hamilton, Ontario: BC Decker Inc, 2003).
    • 5. Yamaoka, M., Yamamoto, T., Ikeyama, S., Sudo, K., Fujita, T. Angiogenesis inhibitor TNP-470 (AGM-1470) potently inhibits the tumor growth of hormone-independent human breast and prostate carcinoma cell lines. Cancer Res. 53, 5233-5236 (1993).
    • 6. Shusterman, S., Grupp, S A., Barr, R., Carpentieri, D., Zhao, H., Maris, J M. The angiogenesis inhibitor tnp-470 effectively inhibits human neuroblastoma xenograft growth, especially in the setting of subclinical disease. Clin. Cancer Res. 7, 977-984 (2001).
    • 7. Yanase, T., Tamura, M., Fujita, K., Kodama, S., Tanaka, K. Inhibitory effect of angiogenesis inhibitor TNP-470 on tumor growth and metastasis of human cell lines in vitro and in vivo. Cancer Res. 53, 2566-2570 (1993).
    • 8. Takamiya, Y., Brem, H., Ojeifo, J., Mineta, T. & Martuza, R. AGM-1470 inhibits the growth of human glioblastoma cells in vitro and in vivo. Neurosurgery 34, 869-875 (1994).
    • 9. Takamiya, Y., Friedlander, R M., Brem, H., Malick, A., Martuza, R L Inhibition of angiogenesis and growth of human nerve-sheath tumors by AGM-1470. J Neurosurg 78, 470-476 (1993).
    • 10. Emoto, M., Tachibana, K., Iwasaki, H., Kawarabayashi, T. Antitumor effect of TNP-470, an angiogenesis inhibitor, combined with ultrasound irradiation for human uterine sarcoma xenografts evaluated using contrast color Doppler ultrasound. Cancer Sci. 98, 929-935 (2007).
    • 11. Nahari, D. et al. Tumor cytotoxicity and endothelial Rac inhibition induced by TNP-470 in anaplastic thyroid cancer. Mol. Cancer Ther. 6, 1329-1337 (2007).
    • 12. Kanamori, M., Yasuda, T., Ohmori, K., Nogami, S., Aoki, M. Genetic analysis of high-metastatic clone of RCT sarcoma in mice, and its growth regression in vivo in response to angiogenesis inhibitor TNP-470. J. Exp. Clin. Cancer Res. 26, 101-107 (2007).
    • 13. Sin, N., Meng, L., Wang, M Q., Wen, J J., Bornmann, W G., Crews, C M. The anti-angiogenic agent fumagillin covalently binds and inhibits the methionine aminopeptidase, MetAP-2. Proc. Natl. Acad. Sci. USA 94, 6099-6103 (1997).
    • 14. Zhang, Y., Griffith, E., Sage, J., Jacks, T. & Liu, J. Cell cycle inhibition by the anti-angiogenic agent TNP-470 is mediated by p53 and p21WAF1/CIP1. Proc Natl. Acad. Sci. USA 97, 6427-6432 (2000).
    • 15. Mauriz, J. et al. Cell-cycle inhibition by TNP-470 in an in vivo model of hepatocarcinoma is mediated by a p53 and p21WAF1/CIP1 mechanism. Transl. Res. 149, 46-53 (2007).
    • 16. Kruger, E., Figg, W D. TNP-470: an angiogenesis inhibitor in clinical development for cancer. Expert. Opin. Investig. Drugs 9, 1383-1396 (2000).
    • 17. Kudelka, A., Verschraegen, C. & Loyer, E. Complete remission of metastatic cervical cancer with the angiogenesis inhibitor TNP-470. N. Engl. J Med. 338, 991-992 (1998).
    • 18. Tran, H. et al. Clinical and pharmacokinetic study of TNP-470, an angiogenesis inhibitor, in combination with paclitaxel and carboplatin in patients with solid tumors. Cancer Chemother. Pharmacol. 54, 308-314 (2004).
    • 19. Kudelka, A. et al. A phase I study of TNP-470 administered to patients with advanced squamous cell cancer of the cervix. Clin. Cancer Res. 3, 1501-1505 (1997).
    • 20. Herbst, R. et al. Safety and pharmacokinetic effects of TNP-470, an angiogenesis inhibitor, combined with paclitaxel in patients with solid tumors: evidence for activity in non-small-cell lung cancer. J. Clin. Oncol. 20, 4440-4447 (2002).
    • 21. Stadler, W., Kuzel, T., Shapiro, C., Sosman, J., Clark, J., Vogelzang, N J. Multi-institutional study of the angiogenesis inhibitor TNP-470 in metastatic renal carcinoma. J. Clin. Oncol. 17, 2541-2545 (1999).
    • 22. Logothetis, C. et al. Phase I trial of the angiogenesis inhibitor TNP-470 for progressive androgen-independent prostate cancer. Clin. Cancer Res. 7, 1198-1203 (2001).
    • 23. Bhargava, P. et al. A phase I and pharmacokinetic study of TNP-470 administered weekly to patients with advanced cancer Clin. Cancer Res. 5, 1989-1995 (1999).
    • 24. Satchi-Fainaro, R. et al. Targeting angiogenesis with a conjugate of HPMA copolymer and TNP-470. Nat Med 10, 255-261 (2004).
    • 25. Cretton-Scott, E., Placidi, L., McClure, H., Anderson, D. & Sommadossi, J. Pharmacokinetics and metabolism of O-(chloroacetyl-carbamoyl)fumagillol (TNP-470, AGM-1470) in rhesus monkeys. Cancer Chemother. Pharmacol. 38, 117-122 (1996).
    • 26. Kataoka, K., Harada, A., Nagasaki, Y. Block copolymer micelles for drug delivery: design, characterization and biological significance. Adv. Drug Deliv. Rev. 47, 113-131 (2001).
    • 27. Harris, J. & Chess, R. Effect of pegylation on pharmaceuticals. Nat. Rev. Drug Discov. 2, 214-221 (2003).
    • 28. Edlund, U. & Albertsson, A. Degradable polymer microspheres for controlled drug delivery. Adv. Polym. Sci. 157, 67-112 (2001).
    • 29. Rogers, M., Birsner, A. & D'Amato, R. The mouse cornea micropocket angiogenesis assay. Nat. Protoc. 2, 2545-2550 (2007).
    • 30. Antoine, N. et al. AGM-1470, a potent angiogenesis inhibitor, prevents the entry of normal but not transformed endothelial cells into the G1 phase of the cell cycle. Cancer Res 54, 2073-6 (1994).
    • 31. Folkman, J. Tumor angiogenesis. in Accomplishments in cancer research (eds. Wells, S. J. & Sharp, P.) 32-44 (Lippincott Williams & Wilkins, New York, 1998).
    • 32. Folkman, J. Tumor angiogenesis. in Cancer Medicine (eds. Holland, J. et al.) 132-152 (B. C. Decker Inc., Ontario, Canada, 2000).
    • 33. Folkman, J., Klement, G. Platelet biomarkers for the detection of disease. US and International Patent 20060204951(2006).
    • 34. Kim, E. S. & Herbst, R. S. Angiogenesis inhibitors in lung cancer. Curr Oncol Rep 4, 325-33 (2002).
    • 35. Kwon, Y. Handbook of Essential Pharmacokinetics, Pharmacodynamics and Drug Metabolism for Industrial Scientists. (Plenum Pub New York; 2001).
    • 36. Pierri, E. & Avgoustakis, K. Poly(lactide)-poly(ethylene glycol) micelles as a carrier for griseofulvin. J. Biomed. Mater. Res. A 75, 639-647 (2005).
    • 37. Kakizawa, Y., Kataoka, K. Block copolymer micelles for delivery of gene and related compounds. Adv. Drug Deliv. Rev. 54, 203-222 (2002).
    • 38. Torchilin, V. Targeted polymeric micelles for delivery of poorly soluble drugs. Cell. Mol. Life Sci. 61, 2549-2559 (2004).
    • 39. Nishiyama, N., Kataoka, K. Current state, achievements, and future prospects of polymeric micelles as nanocarriers for drug and gene delivery. Pharmacol. Ther. 112, 630-648 (2006).
    • 40. Duncan, R. Polymer conjugates as anticancer nanomedicines. Nat. Rev. Cancer 6, 688-701 (2006).
    • 41. Bernier, S. G., Westlin, W. F. & Hannig, G., Fumagillin class inhibitors of methionine aminopeptidase-2. Drugs of the Future 30, 497-508 (2005).
    • 42. Lui, S., Widom, J., Kemp, C. W., Crews, C. M., & Clardy, J., Structure of human methionine aminopeptidase-2 complexed with fumagillin. Science 282, 1324-1327 (1998).
    • 43. Wang et al., Tumor suppression by a rationally designed reversible inhibitor of methionine aminopeptidase-2. Cancer Res. 63, 7861-7869 (2003).
    • 44. Bainbridge et al., Arthritis Res. and Therapy, vol. 9, No. 6.
    • 45. Wang et al., Correlation of tumor growth suppression and methionine aminopetidase-2 activity blockade using an orally active inhibitor, Proc. Natl. Acad. Sci. U.S.A., 105, 1838-1843 (2008).
    • 46. Roskoski, Src protein-tyrosine kinase structure and regulation, Biochemical and Biophysical Research Communications 324 (2004) 1155-1164
    • 47. Tucker, et al., Ectopic expression of methionine aminopeptidase-2 causes cell transformation and stimulates proliferation; Oncogene (2008), 1-10.
    • 48. Regenfuss, B., Bock, F., Parthasarathy, A. & Cursiefen, C. Corneal (lymph)angiogenesis—from bedside to bench and back: a tribute to Judah Folkman. Lymphat. Res. Biol. 6, 191-201 (2008).
    • 49. Clark, A. & Yorio, T. Ophthalmic drug discovery. Nat. Rev. Drug. Discov. 2, 448-459 (2003).
    • 50. Campochiaro, P. Targeted pharmacotherapy of retinal diseases with ranibizumab. Drugs Today 43, 529-537 (2007).
    • 51. Narayanan, R., Kuppermann, B., Jones, C. & Kirkpatrick, P. Ranibizumab. Nat Rev. Drug Discov. 5, 815-816 (2006).
    • 52. ClinicalTrials.gov. Detection of neutralizing antibodies in patients treated with bevacizumab or ranibizumab. National Institutes of Health Clinical Center (CC) NCT00605943(2009).
    • 53. Augustin, A. & Kirchhof, J. Inflammation and the pathogenesis of age-related macular degeneration. Expert. Opin. Ther. Targets 13, 641-651 (2009).
    • 54. Adamis, A. The rationale for drug combinations in age-related macular degeneration.
    • Retina 29, S42-44 (2009).
    • 55. Benny, O., et al. An orally delivered small-molecule formulation with antiangiogenic and anticancer activity. Nat. Biotechnol. 26, 799-807 (2008).
    • 56. Kudelka, A., et al. A phase I study of TNP-470 administered to patients with advanced squamous cell cancer of the cervix. Clin. Cancer Res. 3, 1501-1505 (1997).
    • 57. Kudelka, A., Verschraegen, C. & Loyer, E. Complete remission of metastatic cervical cancer with the angiogenesis inhibitor TNP-470. N. Engl. J. Med. 338, 991-992 (1998).
    • 58. Bhargava, P., et al. A phase I and pharmacokinetic study of TNP-470 administered weekly to patients with advanced cancer Clin. Cancer Res. 5, 1989-1995 (1999).
    • 59. Sugita, S. & Streilein, J. Iris pigment epithelium expressing CD86 (B7-2) directly suppresses T cell activation in vitro via binding to cytotoxic T lymphocyte-associated antigen J. Exp. Med. 198, 161-171 (2003).
    • 60. Rogers, M., Birsner, A. & D'Amato, R. The mouse cornea micropocket angiogenesis assay. Nat. Protoc. 2, 2545-2550 (2007).
    • 61. Adini, A., et al. Matrigel cytometry: A novel method for quantifying angiogenesis in vivo. J. Immunol. Methods 342, 78-81 (2009).
    • 62. Black, C. Delayed type hypersensitivity: current theories with an historic perspective. Dermatol. Online J. 5, 7 (1999).
    • 63. Tobe, T., et al. Targeted disruption of the FGF2 gene does not prevent choroidal neovascularization in a murine model. Am. J. Pathol. 153, 1641-1646 (1998).
    • 64. Marneros, A., et al. Endogenous endostatin inhibits choroidal neovascularization. FASEB J. 21, 3809-3818 (2007).
    • 65. Akula, J., et al. The neurovascular relation in oxygen-induced retinopathy. Mol. Vis. 14, 2499-2508 (2008).
    • 66. Hood, D. & Birch, D. Rod phototransduction in retinitis pigmentosa: estimation and interpretation of parameters derived from the rod a-wave. Invest. Ophthalmol. Vis. Sci. 35, 2948-2961 (1994).
    • 67. Lamb, T. & Pugh, E. J. A quantitative account of the activation steps involved in phototransduction in amphibian photoreceptors. J. Physiol. 449, 719-758 (1992).
    • 68. Pugh, E. J. & Lamb, T. Amplification and kinetics of the activation steps in phototransduction. Biochim. Biophys. Acta. 1141, 111-149 (1993).
    • 69. Akula, J., Mocko, J., Moskowitz, A., Hansen, R. & Fulton, A. The oscillatory potentials of the dark-adapted electroretinogram in retinopathy of prematurity. Invest. Ophthalmol. Vis. Sci. 48, 5788-5797 (2007).
    • 70. de Jong, P. Age-related macular degeneration. N. Engl. J. Med. 355, 1474-1485 (2006).
    • 71. Higgins, G., Wang, J., Dockery, P., Cleary, P. & Redmond, H. Induction of angiogenic cytokine expression in cultured RPE by ingestion of oxidized photoreceptor outer segments. Invest. Ophthalmol. Vis. Sci. 44, 1775-1782 (2003).
    • 72. Abu el-Asrar, A., et al. Monocyte chemotactic protein-1 in proliferative vitreoretinal disorders. Am. J. Ophthalmol. 123, 599-606 (1997).
    • 73. Hartge, M., Unger, T. & Kintscher, U. The endothelium and vascular inflammation in diabetes. Diab. Vasc. Dis. Res. 4, 84-88 (2007).
    • 74. Rollins, B. Monocyte chemoattractant protein 1: a potential regulator of monocyte recruitment in inflammatory disease. Mol. Med. Today 2, 198-204 (1996).
    • 75. Oh, H., et al. The potential angiogenic role of macrophages in the formation of choroidal neovascular membranes. Invest. Ophthalmol. Vis. Sci. 40, 1891-1898 (1999).
    • 76. Jaffe, G., Roberts, W., Wong, H., Yurochko, A. & Cianciolo, G. Monocyte-induced cytokine expression in cultured human retinal pigment epithelial cells. Exp. Eye. Res. 60, 533-543 (1995).
    • 77. Vallabhapurapu, S. & Karin, M. Regulation and function of NF-kappaB transcription factors in the immune system. Annu. Rev. Immunol. 27, 693-733 (2009).
    • 78. Howes, K., et al. Receptor for advanced glycation end products and age-related macular degeneration. Invest. Ophthalmol. Vis. Sci. 45, 37 13-3720 (2004).
    • 79. Yamagishi, S., Ueda, S., Matsui, T., Nakamura, K. & Okuda, S. Role of advanced glycation end products (AGEs) and oxidative stress in diabetic retinopathy. Curr. Pharm. Des. 14, 962-968 (2008).
    • 80. Pachydaki, S., et al. Upregulation of RAGE and its ligands in proliferative retinal disease. Exp. Eye. Res. 82, 807-815 (2006).
    • 81. Barile, G. & Schmidt, A. RAGE and its ligands in retinal disease. Curr. Mol. Med. 7, 758-765 (2007).
    • 82. Goldin, A., Beckman, J., Schmidt, A. & Creager, M. Advanced glycation end products: sparking the development of diabetic vascular injury. Circulation 114,597-605 (2006).
    • 83. Schmidt, A. & Stern, D. Atherosclerosis and diabetes: the RAGE connection. Curr. Atheroscler. Rep. 2, 430-436 (2000).
    • 84. Chavakis, T., Bierhaus, A. & Nawroth, P. RAGE (receptor for advanced glycation end products): a central player in the inflammatory response. Microbes. Infect. 6, 1219-1225 (2004).
    • 85. Takeuchi, M. & Yamagishi, S. Possible involvement of advanced glycation end-products (AGEs) in the pathogenesis of Alzheimer's disease. Curr. Pharm. Des. 14, 973-978 (2008).
    • 86. Riehl, A., Németh, J., Angel, P. & Hess, J. The receptor RAGE: Bridging inflammation and cancer. Cell. Commun. Signal 7, 12 (2009).
    • 87. Logsdon, C., Fuentes, M., Huang, E. & Arumugam, T. RAGE and RAGE ligands in cancer. Curr. Mol. Med. 7, 777-789 (2007).
    • 88. Bierhaus, A., et al. Understanding RAGE, the receptor for advanced glycation end products. J. Mol. Med. 83, 876-886 (2005).
    • 89. Yonekura, H., et al. Novel splice variants of the receptor for advanced glycation end-products expressed in human vascular endothelial cells and pericytes, and their putative roles in diabetes-induced vascular injury. Biochem. J. 370, 1097-1109 (2003).
    • 90. Malherbe, P., et al. cDNA cloning of a novel secreted isoform of the human receptor for advanced glycation end products and characterization of cells co-expressing cell-surface scavenger receptors and Swedish mutant amyloid precursor protein. Brain Res. Mol. Brain Res. 71, 159-170 (1999).
    • 91. Dougan, M. & Dranoff, G. Inciting inflammation: the RAGE about tumor promotion. J. Exp. Med. 205, 267-270 (2008).
    • 92. Stern, D., Yan, S., Yan, S. & Schmidt, A. Receptor for advanced glycation endproducts: a multiligand receptor magnifying cell stress in diverse pathologic settings. Adv. Drug. Deliv. Rev. 54, 1615-1625 (2002).
    • 93. Aggarwal, B., et al. From traditional Ayurvedic medicine to modern medicine: identification of therapeutic targets for suppression of inflammation and cancer. Expert. Opin. Ther. Targets 10, 87-118 (2006).
    • 94. Huttunen, H., Fages, C. & Rauvala, H. Receptor for advanced glycation end products (RAGE)-mediated neurite outgrowth and activation of NF-kappaB require the cytoplasmic domain of the receptor but different downstream signaling pathways. J. Biol. Chem. 274, 199 19-19924 (1999).
    • 95. Ma, W., et al. RAGE ligand upregulation of VEGF secretion in ARPE-19 cells. Invest. Ophthalmol. Vis. Sci. 48, 1355-1361 (2007).
    • 96. Gu, L., et al. Role of receptor for advanced glycation end-products and signalling events in advanced glycation end-product-induced monocyte chemoattractant protein-1 expression in differentiated mouse podocytes. Nephrol. Dial. Transplant. 21, 299-313 (2006).
    • 97. Tomino, Y., et al. Basic research in progressive glomerulopathies: the role of fibrosing factors in IgA nephropathy and diabetic nephropathy. Kidney Int. Suppl. 94, S92-95 (2005).
    • 98. Fiuza, C., et al. Inflammation-promoting activity of HMGB 1 on human microvascular endothelial cells. Blood 101, 2652-2660 (2003).
    • 99. Bierhaus, A., et al. Diabetes-associated sustained activation of the transcription factor nuclear factor-kappaB. Diabetes 50, 2792-2808 (2001).
    • 100. Endo, H., Takenaga, K., Kanno, T., Satoh, H. & Mori, S. Methionine aminopeptidase 2 is a new target for the metastasis-associated protein, S100A4. J. Biol. Chem. 277, 26396-26402 (2002).
    • 101. Björk, P., et al. Identification of human S100A9 as a novel target for treatment of autoimmune disease via binding to quinoline-3-carboxamides. PLoS Biol. 7(2009).
    • 102. Schafer, B. & Heizmann, C. The S100 family of EF-hand calcium-binding proteins: functions and pathology. Trends Biochem. Sci. 21, 134-140 (1996).
    • 103. Nacken, W., Roth, J., Sorg, C. & Kerkhoff, C. S100A9/S100A8:myeloid representatives of the S100 protein family as prominent players in innate immunity. Microsc. Res. Tech. 60, 569-5 80 (2003).
    • 104. Heizmann, C., Ackermann, G. & Galichet, A. Pathologies involving the S100 proteins and RAGE. Subcell. Biochem. 45, 93-13 8 (2007).
    • 105. Donato, R. RAGE: a single receptor for several ligands and different cellular responses: the case of certain S100 proteins. Curr. Mol. Med. 7, 711-724 (2007).
    • 106. Sin, N., Meng, L., Wang, M Q., Wen, J J., Bornmann, W G., Crews, C M. The anti-angiogenic agent fumagillin covalently binds and inhibits the methionine aminopeptidase, MetAP-2. Proc. Natl. Acad. Sci. USA. 94, 6099-6103 (1997).
    • 107. Greish, K. Enhanced permeability and retention of macromolecular drugs in solid tumors: a royal gate for targeted anticancer nanomedicines. J. Drug. Target. 15, 457-464 (2007).

Claims (51)

1. A composition comprising a methionine aminopeptidase-2 (MetAP-2) inhibitor formulation having anti-proliferative activity, wherein said formulation comprises a MetAP-2 inhibitor covalently linked to a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
2. The composition of claim 1, wherein said MetAP-2 inhibitor is a fumagillol derivative.
3. The composition of claim 1, wherein said MetAP-2 inhibitor is covalently linked with the hydrophobic moiety of said block copolymer.
4. The composition of claim 1, wherein said hydrophobic polymer moiety of said block copolymer is selected from the group consisting of poly(d,L-lactic acid), poly(caprolactone) (PCL), and poly(propylene oxide).
5. The composition of claim 1, wherein said hydrophilic moiety is a poly(ethylene glycol) (PEG) polymer.
6. The composition of claim 1, wherein said block copolymer is a diblock copolymer comprising a PEG-PLA diblock copolymer having hydrophilic PEG and hydrophobic PLA moieties.
7. The composition of claim 1 wherein said formulation is formulated for oral administration, topical administration, IV administration, peritoneal administration, injection, ocular administration, suppository administration, pulmonary administration or inhalation, and nasal administration.
8. The composition of claim 1, wherein said anti-proliferative activity is an anti-tumor activity.
9. The composition of claim 2, wherein said fumagillol derivative comprises a derivative selected from the group consisting of 6-O—(N-chloroacetylcarbamoyl)fumagillol (TNP-470), 6-O-(4-methoxyaniline)acetyl fumagillol, 6-O-(3,4,5-trimethexyaniline)acetyl fumagillol, 6-O-(4-(N,N-dimethylethoxy)aniline)acetyl fumagillol, 6-O-(cyclopropylamino)acetyl fumagillol, 6-O-(cyclobutylamino)acetyl fumagillol, 4-((cyclopropylamino)acetyl)oxy-2-(1,2-epoxy-1,5 20 dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-1 cyclohexanol and 4-((cyclobutylamino)acetyl)oxy-2-(1,2-epoxy-1,5 dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-1-cyclohexanol.
10. The composition of claim 1, wherein said MetAP-2 inhibitor is selected from the group consisting of a bengamide, a sulphonamide, a bestatin, a 3-amino-2-hydroxyamide, a hydroxyamide, an acylhydrazine, ovacillin, a reversible MetAP-2 inhibitor and an irreversible MetAP-2 inhibitor.
11. A method of treating a condition involving or relying upon MetAP-2 activity for its pathology, the method comprising administering a MetAP-2 inhibitor formulation comprising a MetAP-2 inhibitor having anti-proliferative activity, covalently linked to a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
12. The method of claim 11, wherein said MetAP-2 inhibitor is a fumagillol derivative.
13. The method of claim 11, wherein said MetAP-2 inhibitor is associated with the hydrophobic moiety of said block copolymer.
14. The method of claim 11, wherein said hydrophobic polymer moiety of said block copolymer is selected from the group consisting of poly(d,L-lactic acid), poly(caprolactone) (PCL) and poly(propylene oxide).
15. The method of claim 11, wherein said hydrophilic moiety is a poly(ethylene glycol) (PEG) polymer.
16. The method of claim 11, wherein said block copolymer is a diblock copolymer comprising a PEG-PLA diblock copolymer having hydrophilic PEG and hydrophobic PLA moieties.
17. The method of claim 11, wherein said anti-proliferative composition has anti-tumor activity.
18. The method of claim 12, wherein said fumagillol derivative comprises a derivative selected from the group consisting of 6-O—(N-chloroacetylcarbamoyl)fumagillol (TNP-470), 6-O-(4-methoxyaniline)acetyl fumagillol, 6-O-(3,4,5-trimethexyaniline)acetyl fumagillol, 6-O-(4-(N,N-dimethylethoxy)aniline)acetyl fumagillol, 6-O-(cyclopropylamino)acetyl fumagillol, 6-O-(cyclobutylamino)acetyl fumagillol, 4-((cyclopropylamino)acetyl)oxy-2-(1,2-epoxy-1,5 20 dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-1 cyclohexanol and 4-((cyclobutylamino)acetyl)oxy-2-(1,2-epoxy-1,5 dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-1-cyclohexanol.
19. The method of claim 11, wherein said MetAP-2 inhibitor is selected from the group consisting of a bengamide, a sulphonamide MetAP-2 inhibitor, a bestatin, a 3-amino-2-hydroxyamide MetAP-2 inhibitor, a hydroxyamide MetAP-2 inhibitor, an acylhydrazine MetAP-2 inhibitor, ovacillin, a reversible MetAP-2 inhibitor and an irreversible MetAP-2 inhibitor.
20. The method of claim 11, wherein said condition comprises a tumor activity.
21. The method of claim 11, wherein the condition is selected from the group consisting of cancer, metastatic tumors, psoriasis, age-related macular degeneration (AMD), thyroid hyperplasia, preeclampsia, rheumatoid arthritis and osteo-arthritis, Alzheimer's disease, obesity, pleural effusion, atherosclerosis, endometriosis, diabetic/other retinopathies, ocular neovascularizations, IL-2 therapy associated edema and other edemas, malaria, SARS, HIV, herpes, lupus, IPF, COPD, asthma, cystic fibrosis, transplant rejection, allergic reaction, multiple sclerosis, bacterial infection, viral infection, conditions involving or characterized by vascular hyperpermeability, inflammation, and spinal injury.
22. A method of making a diblock copolymer composition comprising a MetAP-2 inhibitor that has anti-proliferative activity, the method comprising conjugating said MetAP-2 inhibitor to a diblock copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
23. The method of claim 22, wherein said MetAP-2 inhibitor is a fumagillol derivative that has anti-angiogenic activity.
24. The method of claim 22, wherein said MetAP-2 inhibitor comprises 6-O—(N-chloroacetylcarbamoyl)fumagillol (TNP-470).
25. A diblock copolymer composition comprising a MetAP-2 inhibitor, said composition produced by the method of claim 22.
26. The diblock copolymer composition of claim 25, wherein said MetAP-2 inhibitor is a fumagillol derivative that has anti-proliferative activity.
27. The diblock copolymer composition of claim 26, wherein said fumagillol derivative comprises 6-O—(N-chloroacetylcarbamoyl)fumagillol (TNP-470).
28. A composition comprising a formulation of a fumagillol derivative that has anti-angiogenic activity, said formulation comprising said derivative covalently associated with a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
29. The composition of claim 28, wherein said formulation comprises a micelle comprising said block copolymer associated with said fumagillol derivative.
30. The composition of claim 28, wherein said fumagillol derivative thereof is associated with the hydrophobic moiety of said block copolymer.
31. The composition of claim 28, wherein said hydrophobic polymer moiety of said block copolymer is selected from the group consisting of poly(d,L-lactic acid), poly(L-lysine), poly(aspartic acid), poly(caprolactone) (PCL) and poly(propylene oxide).
32. The composition of claim 28, wherein said hydrophilic polymer moiety of said block copolymer is a polyethylene glycol (PEG).
33. The composition of claim 28, wherein said block copolymer is a diblock copolymer comprising a PEG-PLA diblock copolymer having hydrophilic PEG and hydrophobic PLA moieties.
34. The composition of claim 28, wherein said formulation is formulated for oral administration.
35. The composition of claim 28, wherein said anti-angiogenic activity is an anti-tumor activity.
36. The composition of claim 28, wherein said fumagillol derivative comprises a derivative selected from the group consisting of 6-O—(N-chloroacetylcarbamoyl)fumagillol (TNP-470), 6-O-(4-methoxyaniline)acetyl fumagillol, 6-O-(3,4,5-trimethexyaniline)acetyl fumagillol, 6-O-(4-(N,N-dimethylethoxy)aniline)acetyl fumagillol, 6-O-(cyclopropylamino)acetyl fumagillol, 6-O-(cyclobutylamino)acetyl fumagillol, 4-((cyclopropylamino)acetyl)oxy-2-(1,2-epoxy-1,5 20 dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-1 cyclohexanol and 4-((cyclobutylamino)acetyl)oxy-2-(1,2-epoxy-1,5 dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-1-cyclohexanol.
37. A method of treating an angiogenesis-mediated condition, the method comprising orally administering a composition comprising a formulation of a fumagillol derivative that retains anti-angiogenic activity, said formulation comprising said fumagillol derivative is covalently associated with a block copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety.
38. The method of claim 37, wherein said formulation comprises a micelle comprising said block copolymer associated with said fumagillol derivative.
39. The method of claim 37, wherein said fumagillol derivative thereof is associated with the hydrophobic moiety of said block copolymer.
40. The method of claim 37, wherein said hydrophobic polymer moiety of said block copolymer is selected from the group consisting of poly(d,L-lactic acid), poly(L-lysine), poly(aspartic acid), poly(caprolactone) (PCL) and poly(propylene oxide).
41. The method of claim 37, wherein said hydrophilic polymer moiety of said block copolymer is polyethylene glycol (PEG).
42. The method of claim 37, wherein said block copolymer is a diblock copolymer comprising a PEG-PLA diblock copolymer having hydrophilic PEG and hydrophobic PLA moieties.
43. The method of claim 37 wherein said anti-angiogenic activity is an anti-tumor activity.
44. The method of claim 37 wherein said fumagillol derivative comprises a derivative selected from the group consisting of 6-O—(N-chloroacetylcarbamoyl)fumagillol (TNP-470), 6-O-(4-methoxyaniline)acetyl fumagillol, 6-O-(3,4,5-trimethexyaniline)acetyl fumagillol, 6-O-(4-(N,N-dimethylethoxy)aniline)acetyl fumagillol, 6-O-(cyclopropylamino)acetyl fumagillol, 6-O-(cyclobutylamino)acetyl fumagillol, 4-((cyclopropylamino)acetyl)oxy-2-(1,2-epoxy-1,5 20 dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-1 cyclohexanol and 4-((cyclobutylamino)acetyl)oxy-2-(1,2-epoxy-1,5 dimethyl-4-hexenyl)-3-methoxy-1-chloromethyl-1-cyclohexanol.
45. The method of claim 37 wherein said formulation comprises a diblock copolymer micelle formed with said diblock copolymer.
46. The method of claim 37 wherein said angiogenesis-mediated condition comprises a tumor activity.
47. The method of claim 37, wherein the angiogenesis-mediated condition is selected from the group consisting of cancer, metastatic tumors, psoriasis, age-related macular degeneration (AMD), thyroid hyperplasia, preclampsia, rheumatoid arthritis and osteo-arthritis, Alzheimer's disease, obesity, pleural effusion, atherosclerosis, endometriosis, diabetic/other retinopathies, ocular neovascularizations, IL-2 therapy associated edema and other edemas.
48. A method of making a diblock copolymer micelle comprising a fumagillol derivative that has anti-angiogenic activity, the method comprising conjugating said fumagillol derivative to a diblock copolymer comprising a hydrophilic polymer moiety and a hydrophobic polymer moiety and forming micelles of the resulting conjugate.
49. The method of claim 48, wherein said fumagillol derivative comprises 6-O—(N-chloroacetylcarbamoyl)fumagillol (TNP-470).
50. A diblock copolymer micelle comprising a fumagillol derivative, said micelle produced by the method of claim 48.
51. The diblock copolymer micelle of claim 48, wherein said fumagillol derivative comprises 6-O—(N-chloroacetylcarbamoyl)fumagillol (TNP-470).
US15/005,184 2007-06-26 2016-01-25 Metap-2 inhibitor polymersomes for therapeutic administration Abandoned US20160136292A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US15/005,184 US20160136292A1 (en) 2007-06-26 2016-01-25 Metap-2 inhibitor polymersomes for therapeutic administration
US15/262,143 US9789199B2 (en) 2007-06-26 2016-09-12 MetAP-2 inhibitor polymersomes for therapeutic administration

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US93719807P 2007-06-26 2007-06-26
US5459508P 2008-05-20 2008-05-20
PCT/US2008/068367 WO2009003110A2 (en) 2007-06-26 2008-06-26 Metap-2 inhibitor polymersomes for therapeutic administration
US12/648,078 US8865151B2 (en) 2007-06-26 2009-12-28 MetAP-2 inhibitor polymersomes for therapeutic administration
US14/495,725 US9272050B2 (en) 2007-06-26 2014-09-24 MetAP-2 inhibitor polymersomes for therapeutic administration
US15/005,184 US20160136292A1 (en) 2007-06-26 2016-01-25 Metap-2 inhibitor polymersomes for therapeutic administration

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US14/495,725 Continuation US9272050B2 (en) 2007-06-26 2014-09-24 MetAP-2 inhibitor polymersomes for therapeutic administration

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/262,143 Continuation US9789199B2 (en) 2007-06-26 2016-09-12 MetAP-2 inhibitor polymersomes for therapeutic administration

Publications (1)

Publication Number Publication Date
US20160136292A1 true US20160136292A1 (en) 2016-05-19

Family

ID=39810218

Family Applications (7)

Application Number Title Priority Date Filing Date
US12/648,155 Expired - Fee Related US8790634B2 (en) 2007-06-26 2009-12-28 MetAP-2 inhibitor polymersomes for therapeutic administration
US12/648,078 Active 2029-02-01 US8865151B2 (en) 2007-06-26 2009-12-28 MetAP-2 inhibitor polymersomes for therapeutic administration
US14/313,020 Active US9446140B2 (en) 2007-06-26 2014-06-24 MetAP-2 inhibitor polymersomes for therapeutic administration
US14/495,725 Expired - Fee Related US9272050B2 (en) 2007-06-26 2014-09-24 MetAP-2 inhibitor polymersomes for therapeutic administration
US15/005,184 Abandoned US20160136292A1 (en) 2007-06-26 2016-01-25 Metap-2 inhibitor polymersomes for therapeutic administration
US15/248,043 Expired - Fee Related US9782489B2 (en) 2007-06-26 2016-08-26 MetAP-2 inhibitor polymersomes for therapeutic administration
US15/262,143 Expired - Fee Related US9789199B2 (en) 2007-06-26 2016-09-12 MetAP-2 inhibitor polymersomes for therapeutic administration

Family Applications Before (4)

Application Number Title Priority Date Filing Date
US12/648,155 Expired - Fee Related US8790634B2 (en) 2007-06-26 2009-12-28 MetAP-2 inhibitor polymersomes for therapeutic administration
US12/648,078 Active 2029-02-01 US8865151B2 (en) 2007-06-26 2009-12-28 MetAP-2 inhibitor polymersomes for therapeutic administration
US14/313,020 Active US9446140B2 (en) 2007-06-26 2014-06-24 MetAP-2 inhibitor polymersomes for therapeutic administration
US14/495,725 Expired - Fee Related US9272050B2 (en) 2007-06-26 2014-09-24 MetAP-2 inhibitor polymersomes for therapeutic administration

Family Applications After (2)

Application Number Title Priority Date Filing Date
US15/248,043 Expired - Fee Related US9782489B2 (en) 2007-06-26 2016-08-26 MetAP-2 inhibitor polymersomes for therapeutic administration
US15/262,143 Expired - Fee Related US9789199B2 (en) 2007-06-26 2016-09-12 MetAP-2 inhibitor polymersomes for therapeutic administration

Country Status (6)

Country Link
US (7) US8790634B2 (en)
EP (1) EP2170402B1 (en)
JP (2) JP2010531896A (en)
AU (1) AU2008268262C1 (en)
CA (1) CA2690244C (en)
WO (1) WO2009003110A2 (en)

Families Citing this family (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SMP200800060B (en) 2006-04-07 2009-07-14 Procter & Gamble Antibodies that bind the human protein tyrosine phosphatase beta (hptbeta) and their uses
US8642650B2 (en) 2008-12-04 2014-02-04 Zafgen, Inc. Methods of treating an overweight or obese subject
US20120004162A1 (en) 2008-12-04 2012-01-05 Vath James E Methods of Treating an Overweight or Obese Subject
WO2013055385A2 (en) 2011-10-03 2013-04-18 Zafgen Corporation Methods of treating age related disorders
CA2777108A1 (en) 2009-10-09 2011-04-14 Zafgen Corporation Sulphone compounds and methods of making and using same
WO2011085198A1 (en) 2010-01-08 2011-07-14 Zafgen Corporation Metap-2 inhibitor for use in treating benign prostatic hypertrophy (bph)
JP5827239B2 (en) 2010-01-08 2015-12-02 ザフゲン,インコーポレイテッド Fumagillol type compound and method for producing and using the same
WO2011127304A2 (en) 2010-04-07 2011-10-13 Zafgen Corporation Methods of treating an overweight subject
CN107158398B (en) 2010-05-25 2022-02-18 辛德弗雷克斯公司 Polymer-conjugated MetAP2 inhibitors and methods of treatment using the same
US9895449B2 (en) 2010-05-25 2018-02-20 Syndevrx, Inc. Polymer-conjugated MetAP2 inhibitors, and therapeutic methods of use thereof
US20110294952A1 (en) * 2010-05-25 2011-12-01 SynDevRX Optimized Drug Conjugates
WO2011150338A1 (en) * 2010-05-27 2011-12-01 Zafgen Corporation Methods of treating obesity
WO2012012642A1 (en) 2010-07-22 2012-01-26 Zafgen Corporation Tricyclic compounds and methds of making and using same
US20140073691A1 (en) * 2010-11-10 2014-03-13 Zafgen, Inc. Methods and composition for Treating Thyroid Hormone Related Disorders
CN103379905A (en) 2010-11-29 2013-10-30 扎夫根股份有限公司 Treatment of obesity using non-daily administration of 6-0-(4-dimethylaminoethoxy) cinnamoyl fumagillol
KR20140006888A (en) 2011-01-26 2014-01-16 자프겐 인크. Tetrazole compounds and methods of making and using same
KR101328660B1 (en) * 2011-02-28 2013-11-14 부산대학교 산학협력단 Sorafenib-incorporated poly(ε-caprolactone) film and its drug-eluting stent
CA2829204C (en) 2011-03-08 2019-12-24 Zafgen, Inc. Oxaspiro[2.5]octane derivatives and analogs
MX343687B (en) 2011-05-06 2016-11-16 Zafgen Inc Tricyclic sulfonamide compounds and methods of making and using same.
BR112013028666A2 (en) 2011-05-06 2017-08-08 Zafgen Inc tricyclic pyrazolo sulfonamide compounds and methods for making and using the same
SG194812A1 (en) 2011-05-06 2013-12-30 Zafgen Inc Partially saturated tricyclic compounds and methods of making and using same
CA2850830A1 (en) 2011-10-13 2013-04-18 Aerpio Therapeutics, Inc. Methods for treating vascular leak syndrome and cancer
CN107080843A (en) * 2011-10-13 2017-08-22 爱尔皮奥治疗有限公司 The treatment of illness in eye
MX2014008706A (en) 2012-01-18 2015-03-05 Zafgen Inc Tricyclic sulfonamide compounds and methods of making and using same.
WO2013109735A1 (en) 2012-01-18 2013-07-25 Zafgen, Inc. Tricyclic sulfone compounds and methods of making and using same
CA2867381C (en) * 2012-03-16 2016-09-20 The Johns Hopkins University Controlled release formulations for the delivery of hif-1 inhibitors
EA030318B1 (en) * 2012-03-16 2018-07-31 Дзе Джонс Хопкинс Юниверсити Non-linear multiblock copolymer-drug conjugates for the delivery of active agents
KR20150013222A (en) 2012-05-08 2015-02-04 자프겐 인크. Treating hypothalamic obesity with metap2 inhibitors
KR20150016534A (en) 2012-05-09 2015-02-12 자프겐 인크. Fumagillol type compounds and methods of making and using same
JP2014051478A (en) * 2012-09-10 2014-03-20 Panasonic Corp Functional substance, bio-active pharmaceutical drug and method for permeation of functional substance into living body, and functional substance dissipating apparatus air cleaning device
KR20150079952A (en) 2012-11-05 2015-07-08 자프겐 인크. Tricyclic compounds and methods of making and using same
MX2015005734A (en) 2012-11-05 2016-02-10 Zafgen Inc Methods of treating liver diseases.
WO2014071369A1 (en) 2012-11-05 2014-05-08 Zafgen, Inc. Tricyclic compounds for use in the treatment and/or control of obesity
CA2904353A1 (en) 2013-03-14 2014-09-25 Zafgen, Inc. Methods of treating renal disease and other disorders
EA033912B1 (en) 2013-04-10 2019-12-09 Синдевркс, Инк. MetAP2 INHIBITORS AND METHODS OF TREATING OBESITY
AR105671A1 (en) 2015-08-11 2017-10-25 Zafgen Inc HUMEROCYCLIC COMPOUNDS OF FUMAGILLOL AND ITS METHODS OF ELABORATION AND USE
CN106432255A (en) 2015-08-11 2017-02-22 扎夫根公司 Fumigillol spiro-compound, preparation and use method thereof
WO2017100553A1 (en) 2015-12-10 2017-06-15 Syndevrx, Inc. Fumagillol derivatives and polymorphs thereof
KR20180100663A (en) 2016-01-11 2018-09-11 신데브알엑스, 인크. Treatment of tumors induced by metabolic dysfunction
MX2019000727A (en) 2016-07-20 2019-05-02 Aerpio Therapeutics Inc Humanized monoclonal antibodies that target ve-ptp (hptp-ãŸ).
CN109414037A (en) 2016-12-15 2019-03-01 雀巢产品技术援助有限公司 Adjust the composition and method of the bacterium in companion animals
US20190336535A1 (en) * 2017-01-17 2019-11-07 Tufts Medical Center, Inc. Adoptive transfer of plasmacytoid dendritic cells to prevent or treat ocular diseases and conditions
CN109963570A (en) 2017-01-21 2019-07-02 宁波知明生物科技有限公司 Application of-the O- of the Paeoniflorin -6 ' benzene sulfonate in treatment Sjogren syndrome
US10894824B2 (en) 2018-09-24 2021-01-19 Aerpio Pharmaceuticals, Inc. Multispecific antibodies that target HPTP-β (VE-PTP) and VEGF
US11612577B2 (en) 2018-10-26 2023-03-28 Syndevrx, Inc. Biomarkers of METAP2 inhibitors and applications thereof

Family Cites Families (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5190989A (en) * 1987-06-23 1993-03-02 Mitsubishi Petrochemical Company Limited Hydrophilic group-containing AB-type block copolymer
JP2517760B2 (en) * 1989-05-11 1996-07-24 新技術事業団 Water-soluble polymerized pharmaceutical preparation
US5384333A (en) * 1992-03-17 1995-01-24 University Of Miami Biodegradable injectable drug delivery polymer
WO1994005203A1 (en) * 1992-09-04 1994-03-17 The General Hospital Corporation Biocompatible polymers containing diagnostic or therapeutic moieties
JP2003510359A (en) * 1999-10-01 2003-03-18 スミスクライン・ビーチャム・コーポレイション Compounds and methods
EP1253150B1 (en) * 2000-01-26 2005-03-30 Japan Science and Technology Agency Polymeric micellar structure
US6919307B2 (en) * 2000-11-01 2005-07-19 Praecis Pharmaceuticals, Inc. Therapeutic agents and methods of use thereof for the modulation of angiogenesis
US6548477B1 (en) * 2000-11-01 2003-04-15 Praecis Pharmaceuticals Inc. Therapeutic agents and methods of use thereof for the modulation of angiogenesis
US20070161570A1 (en) * 2000-11-01 2007-07-12 Praecis Pharmaceuticals, Inc. Methionine aminopeptidase-2 inhibitors and methods of use thereof
PL366216A1 (en) * 2000-11-01 2005-01-24 Praecis Pharmaceuticals Inc. Therapeutic agents and methods of use thereof for the modulation of angiogenesis
US7105482B2 (en) * 2000-11-01 2006-09-12 Praecis Pharmaceuticals, Inc. Methionine aminopeptidase-2 inhibitors and methods of use thereof
US6623729B2 (en) * 2001-07-09 2003-09-23 Korea Advanced Institute Of Science And Technology Process for preparing sustained release micelle employing conjugate of anticancer drug and biodegradable polymer
ATE440094T1 (en) 2001-09-27 2009-09-15 Equispharm Co Ltd FUMAGILLOL DERIVATIVES AND METHOD FOR THE PRODUCTION THEREOF
US6592899B2 (en) * 2001-10-03 2003-07-15 Macromed Incorporated PLA/PLGA oligomers combined with block copolymers for enhancing solubility of a drug in water
CA2480809A1 (en) * 2002-04-11 2003-10-23 Children's Medical Center Corporation Methods for inhibiting vascular hyperpermeability
EP1494662B1 (en) * 2002-04-11 2012-11-28 Children's Medical Center Corporation Tnp-470-hpma-methacrylic acid copolymer conjugates and use thereof
US20040265917A1 (en) * 2003-04-07 2004-12-30 Dennis Benjamin Methods of measuring the ability of a test compound to inactivate a biological target in cells of a subject
EP1633367A2 (en) * 2003-05-28 2006-03-15 EntreMed, Inc. Antiangiogenic agents
JP4235226B2 (en) * 2003-10-10 2009-03-11 サムヤン コーポレイション Amphiphilic block copolymer and polymer composition for drug delivery comprising the same
US20050208095A1 (en) * 2003-11-20 2005-09-22 Angiotech International Ag Polymer compositions and methods for their use
WO2005058376A1 (en) * 2003-12-17 2005-06-30 Smithkline Beecham Corporation Polymeric micellar complexes and drug delivery vehicles thereof
AU2004312512A1 (en) * 2003-12-29 2005-07-21 Praecis Pharmaceuticals, Inc. Inhibitors of methionine aminopeptidase-2 and uses thereof
EP1940484A1 (en) * 2005-08-25 2008-07-09 Medtronic Vascular, Inc. Controlled radical polymerization-derived block copolymer compositions for medical device coatings
WO2007124005A2 (en) * 2006-04-18 2007-11-01 Praecis Pharmaceuticals Incorporated Methods for treating bone associated diseases by the use of methionine aminopeptidase-2 inhibitors
WO2009036108A1 (en) * 2007-09-13 2009-03-19 Children's Medical Center Corporation Methods and compositions for inhibiting vascular leakage
CN101336890A (en) * 2008-05-30 2009-01-07 济南基福医药科技有限公司 Anticancer sustained-release gel injection

Also Published As

Publication number Publication date
US9789199B2 (en) 2017-10-17
JP2014208679A (en) 2014-11-06
AU2008268262C1 (en) 2015-02-19
EP2170402A2 (en) 2010-04-07
US20100111894A1 (en) 2010-05-06
JP2010531896A (en) 2010-09-30
US9782489B2 (en) 2017-10-10
US20160361428A1 (en) 2016-12-15
AU2008268262A1 (en) 2008-12-31
US20160375144A1 (en) 2016-12-29
AU2008268262B2 (en) 2014-11-13
US9446140B2 (en) 2016-09-20
EP2170402B1 (en) 2015-03-25
US20100158855A1 (en) 2010-06-24
US8865151B2 (en) 2014-10-21
US20150104406A1 (en) 2015-04-16
CA2690244C (en) 2016-08-09
WO2009003110A3 (en) 2009-10-22
US8790634B2 (en) 2014-07-29
US20140301969A1 (en) 2014-10-09
CA2690244A1 (en) 2008-12-31
WO2009003110A2 (en) 2008-12-31
JP5881779B2 (en) 2016-03-09
US9272050B2 (en) 2016-03-01

Similar Documents

Publication Publication Date Title
US9789199B2 (en) MetAP-2 inhibitor polymersomes for therapeutic administration
WO2009036108A1 (en) Methods and compositions for inhibiting vascular leakage
Yang et al. Estrone-modified pH-sensitive glycol chitosan nanoparticles for drug delivery in breast cancer
Lin et al. Overcoming the anatomical and physiological barriers in topical eye surface medication using a peptide-decorated polymeric micelle
Li et al. Efficacious delivery of protein drugs to prostate cancer cells by PSMA-targeted pH-responsive chimaeric polymersomes
Torchilin Micellar nanocarriers: pharmaceutical perspectives
Ernsting et al. Synthetic modification of carboxymethylcellulose and use thereof to prepare a nanoparticle forming conjugate of docetaxel for enhanced cytotoxicity against cancer cells
Peng et al. Dual chemotherapy and photodynamic therapy in an HT-29 human colon cancer xenograft model using SN-38-loaded chlorin-core star block copolymer micelles
Yan et al. Stepwise targeted drug delivery to liver cancer cells for enhanced therapeutic efficacy by galactose-grafted, ultra-pH-sensitive micelles
US8916206B2 (en) Nanoparticles containing water-soluble non-peptide low-molecular weight drug
Zhao et al. Application of star poly (ethylene glycol) derivatives in drug delivery and controlled release
Liu et al. A novel FK506 loaded nanomicelles consisting of amino-terminated poly (ethylene glycol)-block-poly (D, L)-lactic acid and hydroxypropyl methylcellulose for ocular drug delivery
Shen et al. A self-assembly nanodrug delivery system based on amphiphilic low generations of PAMAM dendrimers-ursolic acid conjugate modified by lactobionic acid for HCC targeting therapy
Salmanpour et al. Nanoparticulate delivery of irinotecan active metabolite (SN38) in murine colorectal carcinoma through conjugation to poly (2-ethyl 2-oxazoline)-b-poly (L-glutamic acid) double hydrophilic copolymer
Wadhwa et al. Polymer-drug conjugates for anticancer drug delivery
Jagur‐Grodzinski Polymers for targeted and/or sustained drug delivery
JP2017524712A (en) Nanoparticles loaded with glucocorticoids to prevent corneal allograft rejection and neovascularization
Zhang et al. Zwitterionic targeting doxorubicin-loaded micelles assembled by amphiphilic dendrimers with enhanced antitumor performance
Lei et al. Nanoscale platform for delivery of active IRINOX to combat pancreatic cancer
Qiu et al. Dendritic Oligoethylenimine Decorated Liposome with Augmented Corneal Retention and Permeation for Efficient Topical Delivery of Antiglaucoma Drugs
Yu Development of Chloroquine-containing HPMA Copolymers for Drug Delivery
Gouveia pH sensitive liposomes for the treatment of rheumatoid arthritis
Jafarzadeh-Holagh et al. Self-Assembled and pH-Sensitive Mixed Micelles as An
Sutton PH Sensitive RNA and Drug Delivery Systems
Kim et al. Polymeric carriers for anticancer drugs

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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

AS Assignment

Owner name: CHILDREN'S MEDICAL CENTER CORPORATION, MASSACHUSET

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BENNY-RATSABY, OFRA;D'AMATO, ROBERT;REEL/FRAME:042721/0156

Effective date: 20100107

Owner name: CHILDREN'S MEDICAL CENTER CORPORATION, MASSACHUSET

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:FOLKMAN, JUDAH;REEL/FRAME:042721/0162

Effective date: 20100908

AS Assignment

Owner name: NIH-DEITR, MARYLAND

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:BOSTON CHILDREN'S HOSPITAL;REEL/FRAME:051355/0745

Effective date: 20191223

AS Assignment

Owner name: THE GOVERNMENT OF THE UNITED STATES, AS REPRESENTED BY THE SECRETARY OF THE ARMY, MARYLAND

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:BOSTON CHILDREN'S HOSPITAL;REEL/FRAME:053767/0605

Effective date: 20191124