NZ550088A - Angiogenesis-affecting compounds and methods of use thereof - Google Patents

Angiogenesis-affecting compounds and methods of use thereof

Info

Publication number
NZ550088A
NZ550088A NZ550088A NZ55008805A NZ550088A NZ 550088 A NZ550088 A NZ 550088A NZ 550088 A NZ550088 A NZ 550088A NZ 55008805 A NZ55008805 A NZ 55008805A NZ 550088 A NZ550088 A NZ 550088A
Authority
NZ
New Zealand
Prior art keywords
angiogenesis
perhexiline
compounds
use according
medicament
Prior art date
Application number
NZ550088A
Inventor
Helmut Augustin
Mats Hellstrom
Original Assignee
Angiogenetics Sweden Ab
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 Angiogenetics Sweden Ab filed Critical Angiogenetics Sweden Ab
Publication of NZ550088A publication Critical patent/NZ550088A/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4402Non condensed pyridines; Hydrogenated derivatives thereof only substituted in position 2, e.g. pheniramine, bisacodyl
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7048Compounds having saccharide radicals and heterocyclic rings having oxygen as a ring hetero atom, e.g. leucoglucosan, hesperidin, erythromycin, nystatin, digitoxin or digoxin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • 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
    • A61P35/00Antineoplastic 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • 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

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Dermatology (AREA)
  • Urology & Nephrology (AREA)
  • Hospice & Palliative Care (AREA)
  • Rheumatology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Vascular Medicine (AREA)
  • Molecular Biology (AREA)
  • Pain & Pain Management (AREA)
  • Endocrinology (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Hydrogenated Pyridines (AREA)
  • Saccharide Compounds (AREA)

Abstract

Disclosed is the use of perhexiline, or a pharmaceutically acceptable salt, solvate, hydrate or combination thereof, for the preparation of a medicament for inhibiting angiogenesis in an angiogenesis related condition chosen from cancer, sarcoma, retinopathy, macular degeneration, corneal ulceration, scleroderma, Berger's disease, proliferative viteoretinopathy, chronic inflammation, inflammatory bowel disease, psoriasis, sarcoidosis and rheumatoid arthritis.

Description

5 00 88 *10056775195* 1 Angiogenesis-Affecting Compounds and Methods of Use Thereof Field of the Invention The invention relates to compounds useful for promoting or inhibiting angiogenesis and 5 to pharmaceutical compositions containing a compound that affects angiogenesis, which are useful in a number of angiogenesis-related conditions. Further, the invention relates to methods of using such angiogenesis-affecting compounds.
Background of the Invention -10 Angiogenesis is the fundamental process by which new blood vessels are formed. The process involves the migration of vascular endothelial cells into tissue, followed by the condensation of such endothelial cells into vessels. Angiogenesis may be induced by an angiogenic agent or be the result of a natural condition. Angiogenesis, or its absence, plays an important role in the maintenance of a variety of pathological states. Some of 15 these states are characterized by neovascularization, e.g., cancer, diabetic retinopathy, glaucoma, and age related macular degeneration. Others, e.g., stroke, infertility, heart disease, ulcers, and scleroderma, are diseases of angiogenic insufficiency.
Angiogenesis has a number of stages (see, e.g., Zhu and Witte, Invest New Drugs 20 17:195-212, 1999). The early stages of angiogenesis include endothelial cell protease ^ production, migration of cells, and proliferation. The early stages also appear to require some growth factors. Later stages of angiogenesis include population of the vessels with mural cells, basement membrane production, and the induction of vessel bed specializations. The final stages of vessel formation include remodelling, wherein a 25 forming vasculature becomes a stable, mature vessel bed. Thus, the process is highly dynamic, often requiring coordinated spatial and temporal waves of gene expression.
The complex angiogenesis process is subject to disruption through interference with one or more critical steps, and numerous disease states can result from or be exacerbated by 30 the disruption. Unregulated angiogenesis can cause or worsen disease, for example, ocular neovascularization has been implicated as the most common cause of blindness 2 and underlies the pathology of approximately 20 eye diseases. In certain previously existing conditions such as arthritis, newly formed capillary blood vessels invade the joints and destroy cartilage. In diabetes, new capillaries formed in the retina invade the vitreous humour, causing bleeding and blindness, In addition to pathologies linked to unregulated angiogenesis, insufficient angiogenesis can also lead to undesirable results. Dead or damaged tissue can lead to numerous pathologies, revascularization of damaged tissues through a healthy, normal angiogenic process is essential to preventing further complications, Thus, it has been recognized in the medical art that compounds which affect angiogenesis have potential both as treatments for a number of disease states and as research tools. Investigators have made some progress in identifying and utilizing natural angiogenesis-affecting molecules. Some progress has also been made toward identifying compounds 15 which could be used to affect angiogenesis. Often it is revealed that those compounds which have promising effects on angiogenesis have unacceptable toxicity profiles, are prohibitively difficult or expensive to make, or both. Despite the current level of knowledge and available treatments, there remains a need in the art for further medicaments or compounds that can be utilized for affecting angiogenesis or preventing 20 or treating an angiogenesis-related condition. It is therefore an object of the present invention to go some way towards meeting this need; or to at least provide the public with a useful choice.
Perhexiline was initially used for management of angina pectoris. Despite some positive effects, adverse hepatic and neurological effects were also associated with Perhexiline administration. The original classification as a coronary vasodilator was revised to a 25 calcium channel antagonist, but recent data suggest it may be a cardiac metabolic agent acting through the inhibition of the enzyme carnitine palmitoyltransferase-1 (CPT-1). Killalea S.M. and Krum H. describe an evaluation of Perhexiline for managing severe myocardial ischemia in Am. J. Cardiocasc. Drugs, 2001; l(3):193-204.
Erythromycin is produced by a strain of Streptornyces eiythraeus belonging to the macrolide group of antibiotics. It is basic and readily forms salt it 1 1 MM r e c eiy; 3 is the base which is microbiologically active. It is primarily given topically or orally to treat bacterial infections. Given the substantial history for use of this drug in humans, it would be desirable to elucidate any further applications for which it would be a candidate. Some recent research has indicated that erythromycin may have new 5 applications, see, for example, Yatsunami J, et al., Inhibitory effects of roxithromycin on tumor angiogenesis, growth and metastasis of mouse B16 melanoma cells, Clin Exp Metastasis. 1999 Mar; 17(2): 119-24; Yatsunami J, et al., Inhibition of tumor angiogenesis by roxithromycin, a 14-membered ring macrolide antibiotic, Cancer Lett. 1998 Sep 25;131(2):137-43; Yatsunami J, et al., Clarithromycin is a potent inhibitor of tumor-10 induced angiogenesis, Res Exp Med (Berl). 1997; 197(4): 189-97.
Dopamine regulators are known and many have been effectively used in patients. For example, Bromocriptine mesylate (Bromocriptine) is an ergot derivative with potent dopamine receptor antagonist activity. It can be used in various other applications, such 15 as infertility, but its full range of potential uses has not yet been elucidated. Similarly, Eticlopride and Lisuride are described in the relevant literature for their ability to regulate dopamine. Dopamine and Dopamine receptor-2 selective agonists such as Bromocriptine have been shown to prevent angiogenesis in Basu S et al., The neurotransmitter dopamine inhibits angiogenesis induced by vascular permeability 20 factor/vascular endothelial growth factor, Nat Med. 2001 May;7(5):569-74. Eticlopride was shown to have not effect on angiogenesis.
It is also known in the art that anti-mycotics exist. For example, miconazole, sulconazole and econazole are known substances that have been utilized in various treatment settings 25 for their anti-fungal activity. Miconazole is perhaps the best known, but sulconazole is also an anti-mycotic, an imidazole derivative with known broad-spectrum antifungal activity. Its ability to act against other microbes is being explored. Econazole, like other anti-fungals, is typically administered topically. When this is done very low doses of the active substance are absorbed. This can be a detriment both in elucidating other uses and 30 in utilizing econazole for those prospective other uses. 4 (followed by page 4a) Further, anti-androgens have been provided, examples of which include flutamide and danazol. Flutamide has been successful!)' used to inhibit androgen, however, there is a serious and substantial risk of hepatic injury in conjunction with its use. Liver failure and death have been reported, typically within the initial few months of flutamide use. Despite this, determining any other potentially beneficial uses of flutamide is warranted. The synthetic hormone danazol, derived from ethisterone, could be a desirable medicament due to its rapid and extensive metabolization following oral administration. Unlike flutamide which is primarily used in male patients, danazol also offers the advantage of having clinical data in both male and female subjects.
Summary of the Invention In one aspect, the present invention provides use of perhexiline, or a pharmaceutically acceptable salt, solvate, hydrate or combination thereof, for the preparation of a medicament for inhibiting angiogenesis in an angiogenesis-related condition chosen from cancer, sarcomas, retinopathy, macular degeneration, corneal ulceration, scleroderma, Berger's disease, proliferative vitreoretinopathy, chronic inflammation, inflammatory bowel disease, psoriasis, sarcoidosis, and rheumatoid arthritis.
In another aspect, the present invention provides a use of: a) perhexiline or a pharmaceutically acceptable salt, solvate, hydrate or combination thereof; and b) at least one anti-VEGF compound; in the manufacture of a medicament for inhibiting angiogenesis, in an angiogenesis-related condition chosen from cancer, sarcoma, retinopathy, macular degeneration, corneal ulceration, scleroderma, Berger's disease, proliferative vitreoretinopathy, chronic inflammation, inflammatory bowel disease, psoriasis, sarcoidosis, and rheumatoid arthritis wherein a) and b) are formulated for separate, simultaneous or sequential administration.
In another aspect, the present invention provides an in vitro method of inhibiting angiogenesis comprising contacting a cell, group of cells, or tissue with a therapeutic; perhexiline or a pharmaceutically acceptable salt, solvate, hydrate, or con: lliyT«SQbefiF<J/^diifit :ofi • OFFICE OF *1,2 R E C E I v . 4a (followed by page 5) Described herein is a method of inhibiting angiogenesis in a group of cells, a tissue or an organism which comprises administering a therapeutically effective amount of perhexiline or a combination of erythromycin and at least one anti-VEGF treatment or pharmaceutically acceptable prodrugs, salts, solvates, hydrates, active metabolites, or combinations thereof to the group of cells, tissue or organism. The organism can be a mammal, and the mammal can have a condition selected from the group consisting of cancer, sarcomas, retinopathy, macular degeneration, corneal ulceration, scleroderma, Berger's disease, proliferative vitreoretinopathy, chronic inflammation, inflammatory bowel disease, psoriasis, sarcoidosis, and rheumatoid arthritis. The compound can be administered locally.
Also described is a method of inhibiting angiogenesis in a group of cells, a tissue or an organism is provided which comprises administering a therapeutically effective amount of a pharmaceutical composition comprising perhexiline or a combination of erythromycin and at least one anti-VEGF treatment or pharmaceutically acceptable prodrugs, salts, solvates, hydrates, active metabolites, or combinations thereof to the group of cells, tissue or organism. The organism can be a mammal, and the mammal can have a condition selected from the group consisting of cancer, sarcomas, retinopathy, macular degeneration, corneal ulceration, scleroderma, Berger's disease, 5 proliferative vitreoretinopathy, chronic inflammation, inflammatory bowel disease, psoriasis, sarcoidosis, and rheumatoid arthritis. The compound can be administered locally. The pharmaceutical composition can be formulated as a salve, gel, ointment, patch, injection, oral solution or suspension and can be in a controlled release matrix. The pharmaceutical composition can further comprise a pharmaceutically acceptable carrier, 10 diluent, vehicle, or excipient. Where the pharmaceutical composition comprises perhexiine the method can further comprise administering at least one anti-VEGF treatment to the group of cells, tissue, or organism. An anti-VEGF treatment can be AVASTIN or MACUGEN.
Also described is a method of stimulating angiogenesis in a group of cells, a tissue or an organism which comprises administering a therapeutically effective amount of a compound selected from the group consisting of Bromocriptine, Eticlopride, Lisuride (S)(-), Miconazole, Sulconazole, Econazole, Flutamide and Danazol or pharmaceutically acceptable prodrugs, salts, solvates, hydrates, active metabolites, or 20 combinations thereof to the group of cells, tissue or organism. The organism can be a mammal. The mammal can have a condition selected from the group consisting of stroke, ischemic heart disease, wound healing, ischemia, myocardial infarction, myocardosis, angina pectoris, unstable angina, coronary arteriosclerosis, arteriosclerosis obliterans, and cerebral infarction. The compound may be administered locally.
Also described is a method of stimulating angiogenesis in a group of cells, a tissue or an organism which comprises administering a therapeutically effective amount of a pharmaceutical composition comprising a compound selected from the group consisting of Bromocriptine, Eticlopride, Lisuride (S)(-), Miconazole, 30 Sulconazole, Econazole, Flutamide and Danazol or pharmaceutically acceptable prodrugs, salts, solvates, hydrates, active metabolites, or combinat OFFICE OF N.Z " I 1 t MAY 2009 ! RECEIVED 6 group of cells, tissue or organism. The organism can be a mammal. The mammal can have a condition selected from the group consisting of stroke, ischemic heart disease, wound healing, ischemia, myocardial infarction, myocardosis, angina pectoris, unstable angina, coronary arteriosclerosis, arteriosclerosis obliterans, and cerebral infarction.
Also described is a method of increasing blood flow to a tissue which comprises administering a pharmaceutical composition comprising a compound selected from the group consisting of Bromocriptine, Eticlopride, Lisuride (S)(-), Miconazole, Sulconazole, Econazole, Flutamide and Danazol to the tissue in an 10 amount effective to promote angiogenesis.
For convenience, certain terms employed in the specification, examples, and appended claims are collected here.
The term "comprising" as used in this specification means "consisting at least in part of'. When 15 interpreting each statement in this specification that includes the term "comprising", features other than that or those prefaced by the term may also be present. Related terms such as "comprise" and "comprises" are to be interpreted in the same manner.
The term "patient" refers to the subject of the novel treatment disclosed herein.
The term "tissue" refers to a group of living cells. The term should be read to encompass single cells and groups of cells where appropriate. The term can also be read to include organs. The term, refers not only to living material in situ but also to living material that has been cultured in vitro or extracted from an organism.
The term "animal" or "organism" refers to mammals, preferably humans.
The term "angiogenesis" means the process by which living cells, tissues, or organisms form new blood "vessels.
The term "angiogenesis-affecting" means a compound or process whictojiffi^^iOeB pk ,. , i >■ angiogenesis in a treated tissue or organism. The effect could be, for examp Le, in^ijpi^j^ ^'{jg j or promotion of angiogenesis. R E C ^ I V P rv' WO 2005/097121 PCT/SE2005/000506 7 The phrase "angiogenesis-related condition" refers to any one of the medical conditions or disease states recognized to be influenced by angiogenesis or by an increase/decrease in angiogenesis of by the lack thereof, including conditions which may be linked to angiogenesis in the future. Examples of" such conditions include cancer, sarcoma, 5 retinopathy, macular degeneration, corneal ulceration, stroke, ischemic heart disease, infertility, ulcers, scleradoma, wound healing, ischemia, ischemic heart disease, myocardial infarction, myocardosis, angina pectoris, unstable angina, coronary arteriosclerosis, arteriosclerosis obliterans, Berger's disease, arterial embolism, arterial thrombosis, cerebrovascular occlusion, cerebral infarction, cerebral thrombosis, cerebral 10 embolism, rubeosis proliferative vitreoretinopathy, chronic inflammation, inflammatory ^ bowel disease, psoriasis, sarcoidosis, and rheumatoid arthritis.
The term "stimulant" refers to molecules or compounds which initiate, promote, or increase in speed, duration, or degree the natural or typical (i.e., unaffected) angiogenesis 15 in the treated tissue or patient.
The term "inhibitor" refers to molecules or compounds which stop, prevent, reduce, hinder, or delay the natural or typical (i.e., unaffected) angiogenesis in the treated tissue or patient.
The term "pharmaceutical," "pharmaceutical compound," or "drug" as used herein, refers ^^to any medicinal substance used in living cells, tissues, humans or other animals. Encompassed within this definition are compound analogs, naturally occurring and synthetic compounds.
The phrase "pharmaceutically acceptable" refers to molecular entities and compositions that are physiologically tolerable and do not typically produce an allergic or similar untoward reaction when administered to a patient.
The phrase "therapeutically effective amount" is used herein to mean an amount which, when applied as part of a desired dosage regimen, is sufficient to affect angiogenesis to 8 an extent that results in some clinically significant change. When used in conjunction with angiogenesis inhibitors, the term means an amount which will prevent, or preferably reduce by at least about 30 percent, more preferably by at least about 50 percent, most preferably by at least about 90 percent, angiogenesis in the tissue or patient treated. When 5 used in conjunction with angiogenesis stimulators or promoters, the term means an amount which will start, or preferably increase by at least about 30 percent, more preferably by at least about 50 percent, most preferably by at least about 90 percent, angiogenesis in the tissue or patient treated. The amount of a given compound described herein that will correspond to a "therapeutically effective amount" will vary depending 10 upon factors such as the particular compound, the disease condition and the severity thereof, the identity of the mammal in need thereof, but it can nevertheless be readily determined by one of skill in the art.
"Treating" or "treatment" is intended- to mean at least the mitigation of a disease 15 condition in a mammal, such as a human, that is alleviated by affecting angiogenesis, and includes: (a) prophylactic treatment in a mammal, particularly when the mammal is found to be predisposed to having the disease; condition but not yet diagnosed as having it; (b) inhibiting the disease condition; and/or (c) alleviating, in whole or in part, the disease condition.
^ A "pharmaceutically acceptable prodrug" means a compound that may be converted under physiological conditions or by solvolysis to a compound of one of the formulas disclosed herein.
A "pharmaceutically acceptable active metabolite" means a pharmacologically active product produced through biological metabolism, such as in the body of a patient, of a compound of one of the formulas disclosed herein.
A "pharmaceutically acceptable solvate" means a solvate that retains the biological 30 effectiveness and properties of the biologically active compounds of one of the formulas disclosed herein. Examples of pharmaceutically acceptable solvates include, but are not 9 limited to, one or more of the compounds disclosed herein in combination with water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic a_cid, or ethanolamine.
A "pharmaceutically acceptable salt" means a salt that retains the biological effectiveness 5 and properties of the free acids and bases of compounds disclosed herein and that is not biologically or otherwise undesirable. Examples of pharmaceutically acceptable salts include, but are not limited to, sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogenphosphates, diliy dro genpho sph ates , metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, 10 caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-l,4-dioates, hexyne-1,6-dioates. benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydrox}^benzoates, methoxyenzoates, phthalates, sulfonates, xylenesulfonates, phenylacetates, phenylpropionates, phenylbutyrates, citrates, lactates, .gamma.-15 hydroxybutyrates, glycollates, tartrates, methane-sulfonates, propanesulfonates, naphthalene-1 -sulfonates, naphthalene-2-sulfonates, and mandelates.
If the described compound is a base, the desired salt may be prepared by any suitable method known to the art, including treatment of the free base -with an inorganic acid, such 20 as hydrochloric acid; hydrobromic acid; sulfuric acid; nitric acid; phosphoric acid; and the like, or with an organic acid, such as acetic acid; maleic a-cid; succinic acid; mandelic acid; fumaric acid; malonic acid; pyruvic acid; oxalic acid; glycolic acid; salicylic acid; pyranosidyl acids such as glucuronic acid and galacturonic acid; alpha-hydroxy acids such as citric acid and tartaric acid; amino acids such as aspartic acid and glutamic acid; 25 aromatic acids such as benzoic acid and cinnamic acid; sulfonic acids such as p-toluenesulfonic acid or ethanesulfonic acid; or the like.
If the described compound is an acid, the desired salt may fee prepared by any suitable method known to the art, including treatment of the free acid "with an inorganic or organic base, such as an amine (primary, secondary or tertiary); an alkali mqEal INTELLECTUAL PRO- metal hydroxide; or the like. Illustrative examples of suitable salts i icludfiF&§arfi€ Salts 11 nay 2009 |R EC EIV e r derived from amino acids such as glycine and arginkie; ammonia; primary, secondary and tertiary amines; and cyclic amines such as piperidine, morpholine, and piperazine, and inorganic salts derived from sodium, calcium, potassium, magnesium., manganese, iron, copper, zinc, aluminum, and lithium.
In the case of compounds, salts, or solvates that are solids, it is understood by those skilled in the art that the described compounds, salts, solvates may exist in different crystal forms, all of which are intended to be within the scope of the present invention.
The described compounds may exist as single stereoisomers, racemates, and/or mixtures of enantiomers and/or diastereomers. All such single stereoisomers, racemates, and mixtures thereof are intended to be within the scope of the present invention. Preferably, the described compounds are used in optionally pure form.
As generally understood by those skilled in the art, an optically pure compound is one that is enantiomerically pure. As used herein, the term "optically pure" is intended to mean a compound which comprises at least a sufficient amount of a single enantiomer to yield a compound having the desired pharmacological activity. Preferably, "optically pure" means a compound that comprises at least 90% of a single isomer, preferably at least 95%, more preferably 97.5%, and most preferably 99%.
Described are methods of affecting angiogenesis and methods of treating or preventing angiogenesis-related conditions, which methods or treatments comprise the use of a compound disclosed herein or a pharmaceuticalLy acceptable prodrug, salt active metabolite, or solvate thereof.
The activity of the described compounds as angiogenesis-effectors may be measured by any of the methods available to those skilled in the art, including in vivo and in vitro assays. , INTELLECTUAL PROPERTY j OFFICE OF N.2. 1 \ MAY 2009 I received! 11 Administration of the compounds disclosed herein, or their pharmaceutically acceptable prodrugs, salts, active metabolites, and solvates, may be performed according to any of the accepted modes of administration available to those skilled in the art. Illustrative examples of suitable modes of administration include, but are not limited to, oral, nasal, 5 parenteral, topical, and transdermal.
The compounds disclosed herein may be administered as a pharmaceutical composition in any suitable pharmaceutical form recognizable to the skilled artisan. Suitable pharmaceutical forms include, but are not limited to, solid, semisolid, liquid, or 0 lyopholized formulations, such as tablets, powders, capsules, suspensions, and aerosols. The pharmaceutical composition may also include suitable excipients, diluents, vehicles, and carriers, as well as other pharmaceutically active agents, depending upon the intended use.
Acceptable methods of preparing suitable pharmaceutical forms of the pharmaceutical compositions are known to those skilled in the art. For example, pharmaceutical preparations may be prepared following conventional techniques of the pharmaceutical chemist involving steps such as mixing, granulating, and compressing when necessary for tablet forms, or mixing, filling, and dissolving the ingredients as appropriate, to give the 0 desired products for, for example, oral, parenteral, topical, intranasal, intrabronchial ^ and/or intraocular administration.
Solid or liquid pharmaceutically acceptable carriers, diluents, vehicles, or excipients may be employed in the pharmaceutical compositions. Illustrative solid carriers iaelude starch, 5 lactose, calcium sulphate dihydrate, terra alba, sucrose, talc, gelatin, pectin, acacia, magnesium stearate, and stearic acid. Illustrative liquid carriers may include alcohol, oil, saline, and water. The carrier or diluent may include a suitable proloznged-release material, such as glyceryl monostearate or glyceryl distearate, alone or with a wax. When a liquid carrier is used, the preparation may be in the form of a syrup, elixir, emulsion, 0 soft gelatin capsule, sterile injectable liquid (e.g. solution), or a nonaqueous or aqueous liquid suspension. 12 A dose of the pharmaceutical composition contains at least a therapeutically effective amount of the active compound (i.e., a compound described herein or a pharmaceutically acceptable prodrug, salt, active metabolite, or solvate thereof) and preferably is made up 5 of one or more pharmaceutical dosage units. The selected dose may be administered to a mammal, for example, a human patient, in need of angiogenesis-effecting treatment, by any known method of administering the dose including topical, for example, as an ointment or cream; orally; parenterally by injection; or continuously by intranasal, intrabronchial, intraaural, or intraocular infusion.
Brief Description of the Drawing Figures Figure 1 shows exemplary results of primary angiogenesis screening assays; Figure 2 shows the chemical structure of perhexiline maleate; Figure 3 shows the chemical structure of erythromycin; Figures 4A, 4B, and 4C depict cell proliferation data for perhexiline; Figures 5 A, 5B, and 5C depict cell proliferation data for erythromycin; Figures 6A, 6B, and 6C show cytoviability assay results for perhexiline; Figures 7A, 7B, and 7C show cytoviability assay results for erythromycin; Figure 8 depicts cell viability data for B16F10 cells treated with perhexiline; Figure 9 depicts cell viability data for B16F10 cells treated with erythromycin; ^ Figure 10 shows IC50 values for two VEGFR-1 inhibitors; Figures 11A and 1 IB show IC50 values for perhexiline alone and in conjunction with VEGFR-1 inhibitors, respectively; Figures 12A and 12B show IC50 values for erythromycin alone and in conjunction with 25 VEGFR-1 inhibitors, respectively; Figure 13 shows the chemical structure of lisuride (S)(-); Figure 14 shows the chemical structure of miconazole; Figure 15 shows the chemical structure of sulconazole; Figure 16 shows the chemical structure of flutamide; Figure 17 shows the chemical structure of danazol; Figure 18 shows cell proliferation data for lisuride; (followed by page 13a) 13 Figure 19 shows cell proliferation data for miconazole; Figure 20 shows cell proliferation data for flutamide; Figure 21 shows cell proliferation data for danazol; Figure 22 quantifies measured HUVEC sprouting after treatment with select compounds; Figure 23 quantifies measured HUVEC sprouting after treatment with select compounds in conjunction with VEGF; and Figure 24 quantifies measured HUVEC sprouting after treatment with select compounds and a VEGFR-1 inhibitor.
Detailed Description In one aspect, the present invention provides use of perhexiline, or a pharmaceutically acceptable salt, solvate, hydrate or combination thereof, for the preparation of a medicament for inhibiting angiogenesis in an angiogenesis-related condition chosen from cancer, sarcomas, retinopathy, macular degeneration, corneal ulceration, scleroderma, Berger's disease, proliferative vitreoretinopathy, chronic inflammation, inflammatory bowel disease, psoriasis, sarcoidosis, and rheumatoid arthritis.
In another aspect, the present invention provides a use of: a) perhexiline or a pharmaceutically acceptable salt, solvate, hydrate or combination thereof; and b) at least one anti-VEGF compound; in the manufacture of a medicament for inhibiting angiogenesis, in an angiogenesis-related condition chosen from cancer, sarcoma, retinopathy, macular degeneration, corneal ulceration, scleroderma, Berger's disease, proliferative vitreoretinopathy, chronic inflammation, inflammatory bowel disease, psoriasis, sarcoidosis, and rheumatoid arthritis wherein a) and b) are formulated for separate, simultaneous or sequential administration.
In another aspect, the present invention provides an in vitro method of inhibiting angiogenesis comprising contacting a cell, group of cells, or tissue with a therapeutically effective amount of perhexiline or a pharmaceutically acceptable salt, solvate, hydrate, or combination thereof.
INTELLECTUAL PROPERTY OFFICE OF N.Z. 1 t MAY 2009 received 13a (followed by page 14) The following examples serve only to illustrate the invention and are not intended to limit the same. If sources are not specifically described materials are known and commercially available.
It should be understood that the described compounds may have chemical and structural properties which are different from those specified hereinafter at some point during their manufacture and use. For example, during use, the compounds may undergo a series of metabolic changes that result in intermediaries with properties outside of the ranges set forth hereinafter for certain parameters. However, such intermediates and metabolic end products are nevertheless still within the scope of this invention if they at one time before, during, or after administration have the requisite values specified hereinafter.
As indicated above, the described compounds can be used to affect angiogenesis. Such uses could be confined to cell or tissue culture application, as well as administration to a patient. In order to ensure the invention is fully disclosed to skilled workers, details related to the identification of the described compounds are provided by way of introduction herein. Where a description provides the purpose of a particular step or material, such purpose is based on the current understanding of the mechanism of 14 action. Such description is provided in the interest of full disclosure; however, the invention is not bound to the theories described herein.
In the description in this specification reference is made to compounds and other subject matter which is not within the scope of the claims of the current application. That subject matter should be readily identifiable by a person skilled in the art and may assist in putting into practice the invention as defined in the claims of this application.
Primary Angiogenesis Screens HUVEC Spheroid Evaluation A test substance was selected for evaluation in angiogenesis screens. The test substance was synthesized or obtained as appropriate and stored at -20 °C in DMSO, Prior to use, the test substance was thawed and 5 mM of the substance was diluted to a 10 fold concentrated working solution (100 uM) in endothelial jell basal medium. The basal medium fraction was supplemented with 25 ng VEGF/100 pi for the stimulation of HUVEC sprouting.
Hie screening procedures were modified versions of those described by Korff and Augustin (J Cell Sci 112: 3249-58, 1999). By way of summary, spheroids were prepared by pipetting 400 human umbilical vein endothelial cells (HUVEC) into each well of nonadhesive 96-well plates. Prior to use the HUVEC were cultured according to the supplier's instructions (PromoCell, Heidelberg, Germany). The cells were allowed to aggregate overnight into spheroids (Korff and Augustin: J Cell Biol 143: 1341-52, 1998). After aggregation, 48 HUVEC spheriods were harvested. Each harvested spheroid was seeded in 900pl of methocel-collagen solution and pipetted into an individual well of a 24-well plate to allow collagen gel polymerization.
Thirty (30) minutes after seeding, the test substance was added. The working dilution (10 fold concentration) was utilized, 100 fil was pipetted on top of the polymerized collagen gel in each well. The plates were then incubated at 37°C for 24 hours. At the end of the incubation period the plates were fixed by adding 1 ml paraformaldehyde (10 %) to each well.
The HUVEC spheriods were microscopically assayed for endothelial eel (EC)|Sjjr^yi^QQg intensity. The degree of EC sprouting was classified into one of comparing the degree of sprouting to that observed in VEGF control a INTELLECTUAL Wpw OFFICE OF ^ yw c W SWSlir were: strong inhibition of sprouting (indicating the test compound is a strong angiogenesis inhibitor), sprouting inhibited to basal sprouting (indicating that the test compound is an angiogenesis inhibitor), excessively above the VEGF control sprouting (indicating the test compound is an angiogenesis stimulator) or indistinguishable to the 5 VEGF control sprouting (where the test compound appears to have no effect). For exemplary results, see Figure 1.
NHDF Spheroid Evaluation Where the test compound was classified as an inhibitor or stimulator, a second screening 10 . process was performed. In the second screen, the test substance was thawed and 5 mM of ^ the substance was diluted to a 10 fold concentrated working solution (100 (J,M) in endothelial cell basal medium. No supplements were used in the basal medium.
Instead of HUVEC, spheroids were prepared by pipetting 400 human dermal fibroblasts 15 (NHDF) into each well of nonadhesive 96-well plates. Prior to use the NHDF were cultured according to the supplier's instructions (PromoCell, Heidelberg, Germany). The cells were allowed to aggregate overnight into spheroids (Korff and Augustin: J Cell Biol 143: 1341-52, 1998). After aggregation, 48 NHDF spheroids were harvested. Each of the harvested spheroids was seeded in 900|il of methocel-collagen solution and pipetted into 20 an individual well of a 24-well plate to allow collagen gel polymerization.
^ Thirty (30) minutes after seeding, the test substance was added. The working dilution (10 fold concentration) was utilized, 100 fxl was pipetted on top of the polymerized collagen gel in each well. The plates were then incubated at 37°C for 24 hours. At the end of the 25 incubation period the plates were fixed by adding 1 ml paraformaldehyde (10 %) to each well.
The NHDF spheroids were microscopically assayed for fibroblast scattering intensity. The degree of fibroblast scattering intensity was classified into one of four groups: strong 30 inhibition of scattering (indicating the test compound is a non-specific inhibitor), scattering inhibited or only mildly inhibited (indicating the test compound is an EC 16 specific inhibitor), scattering unaffected (indicating the test compound is an EC specific stimulator) or scattering increased (indicating the test compound is a non-specific stimulator). For exemplary results, see Figure 1.
Angiogenesis Inhibitors Where a test compound reduced HUVEC sprouting to basal sprouting or there was no sprouting at all, and where NHDF scattering was not affected or mildly affected, the compound was deemed an inhibitor of angiogenesis.
Perhexiline maleate Perhexiline maleate is one such inhibitor. Its' chemical structure is shown in Figure 2. Its' CAS No. is 6724-53-4, formulaic structure is C23H39NO4, and molecular weight is 393.6. Perhexiline maleate exhibited sprouting inhibition but no fibroblast invasion effect, making it a specific angiogenesis inhibitor.
Erythromycin Erythromycin is one such inhibitor. Its' chemical structure is shown in Figure 3. Its' CAS No. is 114-07-8, formulaic structure is C37H67NO13, and molecular weight is 733.9. Erythromycin exhibited sprouting inhibition but no fibroblast invasion effect, making it a specific angiogenesis inhibitor. This finding is surprising in light of the historical uses of erythromycin and even in light of more recent work with alternative applications of the compound.
By identifying the above-noted compounds as angiogenesis inhibitors, the present inventors make available new compounds that will inhibit or prevent angiogenesis. It may be desirable to prevent, decrease, or stop angiogenesis in a patient with certain disease states, such as cancer. Alternatively, it may be desirable to prevent angiogenesis in an otherwise healthy animal or cell/tissue culture model to further study the mechanisms of angiogenesis.
In vitro tests which confirm inhibition WO 2005/097121 PCT/SE2005/000506 17 Cell Proliferation Assay In order to further confirm the surprising results described above, the compounds were tested in a cell proliferation assay. Crystal violet was used to measure cell proliferation; this basic dye stains cell nuclei and provides a sensitive, rapid evaluation. Light 5 absorption of unstained or destained cell layers is negligible, making it possible to perform cell number measurements in wells.
Six cell types (HUVEC, HDMEC, SMC, Fibroblast, A375 and RENCA) were plated and treated with the compounds in a log2 titration for 72 hours. The optimal cell number for 10 each cell type was determined and optical density (OD) was measured at 595 nm. Each data point was measured in quadruplicate. The assay was stopped after 72 hours and subsequently analyzed.
The OD-value of the plated cells before addition of the compounds was subtracted from 15 all OD-values obtained after treatment. The percentage in relation to control proliferation was then calculated. Negative values therefore represent a count of fewer cells than were plated. The IC50 is given below in Table 1. Results are shown as a percentage in relation to the control proliferation. The IC50 were calculated using GraphPad Prism software (GraphPad Software, Inc.). Perhexiline results are depicted graphically in Figures 4A -20 4C and Erythromycin results in Figures 5A - 5C.
^ Table 1: Summary of die IC50 obtained in the proliferation assay.
Compound IC50 values for each cell type HUVEC HDMEC SMC Fibroblast A3 75 RENCA Perhexiline 1.0 x 10'5 1.1 x 105 1.4x10' 1.2 x 10"5 2.5 x 10" 7.6 x 1(T6 Erythromycin >1 x 10'4 ~1 x 10"4 >1 x ltr4 >1 x 10-4 >1 x 10"4 .6 x 10"^ Cytoviability Assay WO 2005/097121 PCT/SE2005/000506 18 A cell viability assay based on cellular reduction of resazurin to the fluorescent product resorufin was performed. Viable cells can metabolize and reduce the dye, whereas dead cells rapidly lose the capacity to do so, making the increase in fluorescence directly proportional to the number of viable cells. The tested compounds were added in a log2 5 titration and viable cells were analyzed 24 hours after treatment. The fluorescence was determined at an excitation wavelength of 560 nm and an emission wavelength of 590 nm. The percentage of viable cells in relation to the untreated control proliferation was calculated. Table 2 shows the IC5o values obtained in the cell viability assay. Perhexiline results are depicted graphically in Figures 6A - 6C and Erythromycin results in Figures 10 7A-7C.
Table 2: Summary of selected IC50 results obtained in the cell viability assay Compound IC50 values for each cell type HUVEC HDMEC SMC Fibroblast A375 RENCA Perhexiline 8.4 x lO-6 8.0 x 10"6 3.2 xlO"5 .2 x lO-6 1.0 x 10° .0 x 10"6 Through the two aforementioned assays it was determined that RENCA cells are more sensitive when treated with Perhexiline than the other cell types in the proliferation assay 15 and in the viability assay. Therefore, the B16F10 cells were subsequently tested. The ^^B16F10 tumor model, like the RENCA model, is an orthotopic tumor model. The IC50 after 24 and 48 hours incubation for Perhexiline- and Erythromycin-treated B16F10 cells was determined at 1.0 x 10"5 M; results are given in Figures 8 and 9.
Evaluation of synergistic effects in combination with VEGF inhibitors The 3D angiogenesis assay is suitable for IC50 determinations with a narrow variation. Therefore, the assay was applied to test compounds alone or in combination with a VEGFR-2 inhibitor to address changes in IC50. The inhibitors tested were SU5614 and PTK787/ZK222584. The VEGFR-2 inhibitor was applied at a concentration close to the 25 IC50.
WO 2005/097121 PCT/SE2005/000506 19 SU5614 has an IC50 = 4.2 x 10"6 M and has been described in, inter alia, Sun L, et al., Synthesis and biological evaluations of 3-substituted indolin-2-ones: a novel class of tyrosine kinase inhibitors that exhibit selectivity toward particular receptor tyrosine kinases. J Med Chem. 1998 Jul 2;41(14):2588-603. 5 and available from, e.g. Calbiochem. PTK787/ZK222584 (at times abbreviated AG1 in the figures) has an IC50 = 1.0 x 10"7 M and has been described in, inter alia, Bold, G. et al., New anilinophthalazines as potent and orally well absorbed inhibitors of the VEGF receptor tyrosine kinases useful as antagonists or tumor-driven angiogenesis, J. Med. Chem. 2000, 43, 2310-2323.
^ The IC50 value of the VEGFR-2 inhibitors was determined in the spheroid-based 3D assay, see Figure 10. The perhexiline or erythromycin was added on top of the gel where tested in combination with AG1 or directly in the gel when tested in combination with SU5614, at the concentration shown in Figures 11A and B and 12A and B, respectively. 15 Data are summarized in Table 3.
Table 3: Summary of the IC5o obtained in the synergistic 3D angiogenesis assay Compound without VEGFR-2 inhibitor with VEGFR-2 inhibitor Perhexiline .4 x 10"" 3.3 x lO'6 Erythromycin - 8.6 x lO"3 Further assays can be performed to demonstrate the useful effects of the claimed compounds. For example, the Spherogenex angiogenesis assay can be used to elucidate the effects of specific enantiomers of the test compounds. As angiogenesis is known to be driven at times by VEGF, the assay can also be employed to ascertain bFGF-driven angiogenesis, thereby allowing determination of the effects of the compounds on non-25 VEGF-driven angiogenesis. mRNA expression profiling of the Spherogenex angiogenesis assay comparing treatment with the angiogenesis modulating compounds WO 2005/097121 PCT/SE2005/000506 and nontreated controls could be used to determine which signalling pathways are affected by the test compounds.
One example of a further analysis is a Hepato-Cellular Carcinoma Model (HCC) in vivo 5 study. Such a study can evaluate the anti-angiogenic effect of Perhexiline and Erythromycin in a subcutaneous and highly vascularized tumor model such as Alexander cells, Perhexiline has been shown, above, to affect in vitro 3D sprouting of HUVEC (IC50 = 1.5 x 10"6 M) more potently than fibroblast scattering (IC50 = 1.9 x 10"5 M). Perhexiline has an influence on the viability of cells (Endothelial cells, Fibroblast, Alexander cells) 10 with an IC5o around 10 jiM which could interfere with potential anti-angiogenic effects. Erythromycin's effect on HUVEC sprouting was only observed when it was combined with VEGFR-2 inhibitors. In such a further study the anti-angiogenic effect of Erythromycin alone could be excluded.
Alexander cells (PLC/PRF/5) are available from, e.g. American Type Culture Collection (Manassas, Virginia, USA). Monolayers of Alexander cells can be grown in 85% Dulbecco's Modified Eagle's Medium (DMEM), 15% Fetal Bovine Serum (FBS), 4mM L-glutamine, 100 units penicillinG/ml, and 100jig of streptomycin-sulfate/ml. The cells can be cultured in a humidified atmosphere of 90% air and 10% carbon dioxide at 37°C.
Media may preferably be changed every 4 days.
£ ^^Six to eight week old female NMRTnu/mi mice could be utilized. The Alexander cells can be implanted in the left flank region of the mice, for example, by injecting 4xl07 cells in 0.2 ml aliquots into the subcutaneous space of the left flank using a 29G needle syringe. 25 After inoculation the appearing subcutaneous tumours can be measured by callipering and multiplying the distances of all three dimensions.
Therapy can be initiated after the tumor size has reached a volume of approximately 100 mm3 (day 1 of the study), and continue for about 21 days. Four experimental groups 30 could be used as detailed in Table 4, below.
WO 2005/097121 PCT/SE2005/000506 21 Table 4: Optional dosing scheme for in vitro analysis Group Treatment Application Route 1 Control - - 2 Perhexiline 40 mg/lcg (1/2 MTD) p.o. 3 Erythromycin mg/kg i.p. 4 PTK787/ZK222584 50 mg/kg p.o.
As shown, Group 1 would serve as control, where the animals would receive no further treatment after injection of Alexander cells. Group 2 could evaluate perhexiline's effects on the tumor through daily doses of 40 mg/kg via oral gavage. Group 3 could receive erythromycin once daily in standard dose levels, such as 10 mg/kg i.p., preferably given in conjunction with a low dose of PTK787/ZK222584. Group 4 could be administered PTK787/ZK222584 orally at doses of 50 mg/kg twice daily, optionally a sub-group of Group 4 or a separate Group 5 could receive a low dose of PTK787/ZK222584.
After approximately 21 days of treatment, mice could be sacrificed and tumors collected and optionally stored appropriately at -80°C. For histological examination of the tumor vasculature, cryosections of the tissues (thickness of 5-10|xm) can be taken. For visualization of the blood vessels, immunohistochemical staining for CD 31 (PECAM-1) can be performed, and vessels counted microscopically using a defined magnification (x200). Examinations of all tissue sections at low magnification and can be carried out, including photography if desired. The proliferation index of the tumor tissue could be examined by BrdU labeling of cryosections. If that is the case, BrdU (500mg/kg) should be administered to the animals 12 hours before sacrifice. To investigate the apoptotic index a TUNEL staining could be performed on cryosections.
Based on the significant results already acquired, such a study would be expected to reveal a reduced tumor size in Groups 2-4 as compared to control Group 1. If variable amounts (low and high doses) of PTK787/ZK222584 were given in Group 4, those receiving the high dose amount would be expected to have a smaller tumor than those WO 2005/097121 PCT/SE2005/000506 22 with the low amount. Both the Group 2 animals treated with perhexiline and the Group 3 animals receiving erythromycin combined with a low dose of PTK787/ZK222584 would be expected to have reduced tumors of the scale seen with the high dose PTK787/ZK222584 animals. As both perhexiline and erythromycin/VEGFR-2 inhibitor 5 have been shown to have surprising angiogenesis-inhibiting activity, the tumors in animals receiving these compounds would be considerably smaller than those observed in control animals. Tumor apoptosis could be increased and a decrease in microvascular density could be observed for the Group 2 and 3 animals.
Angiogenesis Stimulators ^ Where a test compound stimulated HUVEC sprouting excessively above the VEGF control and where NHDF scattering was unaffected or only mildly increased, the compound was deemed a stimulator of angiogenesis.
Dopamine regulators Bromocriptine is an angiogenesis stimulator with the molecular formula CaaH^BrNsCVCBLtSOj and a molecular weight of 750.70. Eticlopride is also known as a dopamine receptor blocker, as is Lisuride (S)(-). Lisuride's chemical structure is shown in Figure 13. Its' CAS No. is 18016-80-3, formulaic structure is C2oH26N40, and molecular weight is 338.5. Lisuride (S)(-) exhibited increased sprouting stimulation but no fibroblast invasion effect, making it a specific angiogenesis stimulator. This is in surprising contrast to that described previously and noted above, where research indicated that certain of these dopamine regulators had no effect on angiogenesis or actually works to prevent angiogenesis.
Anti-Mycotics Miconazole is one such stimulator. Its' chemical structure is shown in Figure 14. Its' CAS No. is 22916-47-8, formulaic structure is C18H14Q4N2O, and molecular weight is 416.1. Miconazole exhibited increased sprouting stimulation but no fibroblast invasion effect, making it a specific angiogenesis stimulator. Sulconazole, whose chemical structure is shown in Figure 15, is (+)-l-[2.4-dichloro-& -[(p-chlorobenzyl)-thio]- 23 phenethyl]imidazole mononitrate. Econazole is l-[2-{(4-chloro-phenyl)methoxy}-2-(2,4-dichlorophenyl)ethyl]-lH-imidazole mononitrate, both are known anti-mycotics and have surprisingly been shown to be angiogenesis stimulators as well Anti-Androgens Flutamide is one such stimulator. Its' chemical structure is shown in Figure 16. Its' CAS No, is 13311-84-7, formulaic structure is C11H11F3N2O3, and molecular weight is 276.2. Flutamide exhibited increased sprouting stimulation but no fibroblast invasion effect, making it a specific angiogenesis stimulator. Danazol is also an anti-androgen that has surprisingly been shown to be an angiogenesis stimulator, Its' chemical structure is shown in Figure 17. Its' CAS No. is 17230-88-5, formulaic structure is C22H27NO2, and molecular weight is 337.5. Danazol exhibited increased sprouting stimulation but no fibroblast invasion effect, making it a specific angiogenesis stimulator.
By identifying the above-noted compounds as angiogenesis stimulators, the present inventors make possible the possibility of new compounds that will stimulate or begin angiogenesis. It may be desirable to stimulate angiogenesis in a patient with certain disease states, such as ischemia. Alternatively, it may be desirable to stimulate angiogenesis in an animal or cell/tissue culture model to further study the mechanisms of angiogenesis. For example, by administering a compound of the present invention, angiogenesis could be stimulated in an animal model expressing a tumor, providing additional research opportunities into the growth of the tumor and possibly improved animal models for studying tumor treatments.
In vitro tests which confirm stimulation Cell Proliferation Assay In order to further confirm the surprising results described above, the compounds were tested in a cell proliferation assay. The assay was performed as described, above. The OD value at day 0 was subtracted from the OD value after 72 hours of treatment.
Negative values therefore represent observation of fewer cells than originally plated and WO 2005/097121 PCT/SE2005/000506 24 further indicate cytotoxic effects of the compound on HUVEC. The compounds inhibit proliferation, results are shown in Figures 18-21.
HUVEC Spheroid Results 5 As described above with regard to angiogenesis inhibitors, the compounds newly found to be angiogenesis stimulators were also evaluated in a 3D angiogenesis assay. They were all consistently found to independently stimulate EC sprouting or further increase VEGF-induced sprouting.
The HUVEC monolayer was treated for 6 hours with the compounds then the induced transcriptional changes, when compared to untreated HUVEC, were determined. The compounds were tested according to their ability to induce sprouting without addition of VEGF, in combination with VEGF and together with the VEGFR-2 inhibitor PTK787/ZK222584 (1 |jM). The VEGFR-2 inhibitor completely prevents VEGF-induced 15 sprouting at a concentration of 1 jjM.
Figure 22 shows the effects of the compounds alone on HUVEC sprouting, after 24 hours of treatment the basal sprouting was subtracted and the resultant sprouting compared to VEGF-induced sprouting. Both bromocriptine and eticlopride stimulated sprouting in 20 excess of the VEGF-induced sprouting. m The compounds given in combination with VEGF (25 ng/ml) were also evaluated, data shown in Figure 23. The compounds can enhance VEGF-driven angiogenesis even further. Results from the evaluation of the compounds administered in conjunction with 25 PTK787/ZK222584 are shown in Figure 24, where the percentage is given as compared to untreated control. The results show that the sprouting is not VEGF-dependent since it can not be inhibited by VEGFR inhibitor.
In vivo tests to further elucidate stimulation 30 In addition to the in vitro procedures noted above, in vivo tests could also be conducted to further qualify and quantify the action of the test compounds. The test compounds could be evaluated in conjunction with or without angiogenic stimulators (for example, growth factors such as VEGF or bFGF) in a matrigel angiogenesis assay as described in Passaniti, A., et al Lab Invest. 1992 Oct;67(4):519-28. Alternatively or in addition, the modified matrigel angiogenesis procedure as in Guedez, L., et al., American Journal of 5 Pathology, 2003, vol. 162, no. 5, 1431-39 could be used.
Information could also be compiled by analysing the effects of test compound treatment on the angiogenesis process in disease models of myocardial infarction and tissue ischemias. Such models are known to the skilled worker and include, for example, those 10 described in W003/000009 (PCT/US02/19568) and Witzenbichler B., Am J Pathol. 1998 0 Aug;153(2):381-94.
A further in vitro evaluation that could be performed would be to evaluate the retinal vascularization Mid vessel density in mice treated with the compounds as described in 15 WO 05/008250.
Dosing and guidelines for use The compounds described herein can be used to affect angiogenesis. The amount of compound, route of administration and other related factors could well be based on the 20 regimen advised for the compound's original indication. Such information is available to the skilled worker; see, for example, Goodman & Gilman, The Pharamacological Basis of Therapeutics. Dosing and delivery could be modified based on the needs of the patient and the availability of improved dosing techniques as they develop in the art.
The preferred method of treatment may further include a step of detecting angiogenesis in the affected tissue or organism following treatment. Methods of detecting angiogenesis are known in the art, see for example, U.S. Patent No. 6,689,807.
It is contemplated that a compound of the present invention could be metabolised by 30 enzymes in the body of the organism such as a human being to generate a metabolite that can affect angiogenesis. Such metabolites are within the scope of the present invention. It WO 2005/097121 PCT/SE2005/000506 26 is also contemplated that precursor compounds could be administered which, after undergoing processing such as enzyme metabolism, would result in the compound of the present invention.
In certain circumstances, a skilled worker may choose to employ one or more of the compounds discussed above in conjunction with another active compound. Alternatively, two or more of the compounds disclosed herein may be administered in conjunction to affect angiogenesis to a different degree. The compounds may be administered at the same time or in a sequential fashion. One way to determine whether particular 10 combinations of compounds interact in a preferred fashion is by preparing a linear isobologram (Tallarida RJ., et al, Statistical analysis of drug-drug and site-site interactions with isobolograms, Life Sciences. 45(11):947-61, 1989). Such a diagram helps elucidate where the additional compound(s) have merely an additive effect, or where they have a synergistic effect. In such an evaluation, at least nine experimental 15 groups are evaluated; three groups define the dose-response of a first compound, three groups define the dose-response for a second compound, and three groups define the dose-response for a fixed ratio combination of those two compounds.
The selected dose response values, ED50 values, for the single dose groups are plotted as 20 a line which assumes an addititive effect. This line is based on the compound ratio tested; a 1:2 ratio of first test compound to second test compound is shown. When data measured for the group with combined compound administration is superimposed on the hypothetical addititive line, an observed dose response which lies to the left (i.e., below) of the additive line represents a synergistic effect of the compounds used together, 25 whereas a observed dose response to the right (i.e., above) of the additive line represents an antagonistic effect of the compounds in combination.
The compounds disclosed herein may be particularly effective when combined with treatments which block VEGF activity or signalling. Such anti-VEGF treatments include 30 inhibitory anti-VEGF receptor antibodies, soluble receptor constructs, antisense 27 strategies, RNA aptamers against VEGF and low molecular weight VEGF receptor tyrosine kinase (RTK) inhibitors.
Examples of specific compounds which may preferably be used in combination with the above-noted compounds are AVASTIN (bevacizumab), a recombinant human antibody to VEGF, and MACUGEN (pegaptanib sodium), an aptamer which selectively binds to and neutralizes VEGF. Research in the anti-VEGF art is ongoing, future compounds would be expected to show similar positive effects when used in conjunction with one or more of the compounds disclosed hererin.
The effective dosage amount will vary with the particular condition being treated, the age and physical condition of the subject being treated, the severity of the condition, the duration of treatment, the nature of concurrent therapy (if any), the specific route of administration and factors within the knowledge and expertise of a health care practitioner. For example, in connection with occlusive or obstructive vascular disorders, an effective amount is that amount which engenders sufficient angiogenesis so as to provide an increase in blood flow in the ischemic region.
The foregoing disclosure has been set forth merely to illustrate the invention and is not intended to be limiting. Since modifications of the disclosed embodiments incorporating the spirit and substance of the invention may occur to persons skilled in the art, the invention should be construed to include everything within the scope of the appended claims and equivalents thereof. 28

Claims (12)

WHAT WE CLAIM IS:
1. Use of perhexiline, or a pharmaceutically acceptable salt, solvate, hydrate or combination thereof, for the preparation of a medicament for inhibiting angiogenesis in an angiogenesis-related condition chosen from cancer, sarcoma, retinopathy, macular degeneration, corneal ulceration, scleroderma, Berger's disease, proliferative vitreoretinopathy, chronic inflammation, inflammatory bowel disease, psoriasis, sarcoidosis, and rheumatoid arthritis.
2. A use according to claim 1, wherein the medicament is formulated for local administration.
3. A use according to claim 1 or 2, wherein the perhexiline is in the form of a composition.
4. A use according to any one of claims 1-3, wherein the medicament is formulated as a salve, gel, ointment, patch, injection, oral solution or suspension.
5. A use according to any one of claims 1-4, wherein the medicament is in a controlled release matrix.
6. A use according to any one of claims 1-5, wherein the medicament further comprises a pharmaceutically acceptable carrier, diluent, vehicle, or excipient.
7. A use according to any one claims 1-6, wherein the medicament further comprises at least one anti-VEGF compound.
8. A use according to claim 7, wherein said at least one anti-VEGF compound is selected from the group consisting of AVASTIN and MACUGEN.
9. Use of: a) perhexiline or a pharmaceutically acceptable salt, solvate, hydrate or combination thereof; and a) at least one anti-VEGF compound: in the manufacture of a medicament for inhibiting angiogenesis, in an angiogenesis- related condition chosen from cancer, sarcoma, retinopathy, macular degeneration, corneal ulceration, scleroderma, Berger's disease, proUferati^^tt®r®tWK>path.yr I vJFFICE Of rv i " I 1 1 MAr 2009 [RECEIVE '29 chronic inflammation, inflammatory bowel disease, psoriasis, sarcoidosis, and rheumatoid arthritis; wherein a) and b) are formulated for separate, simultaneous or sequential administration.
10. An in vitro method of inhibiting angiogenesis comprising contacting a cell, group of cells or tissue with a therapeutically effective amount of perhexiline or a pharmaceutically acceptable salt, solvate, hydrate, or combination thereof.
11. A use according to claim 1 or 9, substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.
12. A method according to claim 10, substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.
NZ550088A 2004-04-06 2005-04-06 Angiogenesis-affecting compounds and methods of use thereof NZ550088A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US52134604P 2004-04-06 2004-04-06
PCT/SE2005/000506 WO2005097121A1 (en) 2004-04-06 2005-04-06 Angiogenesis-affecting compounds and methods of use thereof

Publications (1)

Publication Number Publication Date
NZ550088A true NZ550088A (en) 2009-08-28

Family

ID=35124826

Family Applications (1)

Application Number Title Priority Date Filing Date
NZ550088A NZ550088A (en) 2004-04-06 2005-04-06 Angiogenesis-affecting compounds and methods of use thereof

Country Status (7)

Country Link
EP (1) EP1737452A1 (en)
JP (1) JP2007532536A (en)
CN (1) CN1938025A (en)
AU (1) AU2005230811A1 (en)
CA (1) CA2560416A1 (en)
NZ (1) NZ550088A (en)
WO (1) WO2005097121A1 (en)

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0405381D0 (en) 2004-03-10 2004-04-21 Univ Wales Medicine A method and means for treating heart failure
CN103169714A (en) 2005-07-12 2013-06-26 Dmi生物科学公司 Methods and products for treatment of diseases
CA2619654A1 (en) * 2005-08-24 2007-03-01 Cell Matrix Combination therapies for inhibiting integrin-extracellular matrix interactions
WO2007087457A2 (en) * 2006-01-30 2007-08-02 (Osi) Eyetech, Inc. Combination therapy for the treatment of neovascular disorders
GB0908193D0 (en) 2009-05-13 2009-06-24 Albright Patents Treatment of disease state
CA2816448A1 (en) 2009-05-20 2010-11-25 Heart Metabolics Limited Treatment of heart failure with normal ejection fraction
CA2751002C (en) 2009-06-22 2012-09-11 Dmi Acquisition Corp. Method for treatment of diseases
SG10201705044YA (en) 2012-12-19 2017-07-28 Ampio Pharmaceuticals Inc Method for treatment of diseases
WO2015131231A1 (en) * 2014-03-03 2015-09-11 Adelaide Research & Innovation Pty Ltd Methods for using (-)-perhexiline
US20170114323A1 (en) * 2014-06-19 2017-04-27 Whitehead Institute For Biomedical Research Uses of kinase inhibitors for inducing and maintaining pluripotency
US10124008B2 (en) 2014-06-23 2018-11-13 Wisconsin Alumni Research Foundation Use of inhibitors of acid sphingomyelinase to treat acquired and inherited retinal degenerations
CN113546172A (en) * 2020-04-24 2021-10-26 山东大学齐鲁医院 Application of VEGF inhibitor in preparation of medicine for treating hypoxia-related diseases

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5512591A (en) 1993-02-18 1996-04-30 President And Fellows Of Harvard College Treatments for diseases characterized by neovascularization
US6413967B1 (en) * 1995-03-30 2002-07-02 The University Of Virginia Patents Foundation Inhibition of novel calcium entry pathway in electrically non-excitable cells acting as an anti-proliferative therapy
AUPN380695A0 (en) * 1995-06-23 1995-07-20 Queen Elizabeth Hospital, The Methods related to the treatment of and isolation of compounds for treatment of ischaemic conditions
CA2295098A1 (en) * 1997-06-30 1999-01-07 Allergan Sales, Inc. Calcium blockers to treat proliferative vitreoretinopathy
GB0008269D0 (en) * 2000-04-05 2000-05-24 Astrazeneca Ab Combination chemotherapy
US20020137755A1 (en) * 2000-12-04 2002-09-26 Bilodeau Mark T. Tyrosine kinase inhibitors
US20030162824A1 (en) * 2001-11-12 2003-08-28 Krul Elaine S. Methods of treating or preventing a cardiovascular condition using a cyclooxygenase-1 inhibitor
AU2003262515A1 (en) 2002-09-19 2004-04-08 Nieland, John Mr Inhibitors of the fatty acid oxidation for the prophylaxis and/or the treatment of chronic and/or atopic skin diseases

Also Published As

Publication number Publication date
WO2005097121A1 (en) 2005-10-20
CA2560416A1 (en) 2005-10-20
AU2005230811A1 (en) 2005-10-20
EP1737452A1 (en) 2007-01-03
JP2007532536A (en) 2007-11-15
CN1938025A (en) 2007-03-28

Similar Documents

Publication Publication Date Title
NZ550088A (en) Angiogenesis-affecting compounds and methods of use thereof
KR102647026B1 (en) Fluorinated tetrahydronaphthyridinyl nonanoic acid derivatives and uses thereof
RU2753740C2 (en) Treatment of hyperkinetic motor disorders
EP2588110B1 (en) Methods of targeting pten mutant diseases and compositions therefor
JP6238969B2 (en) New method
JP5341933B2 (en) Preventive and therapeutic agent for stress urinary incontinence and screening method thereof
KR20180064373A (en) Diaryl and aryl heteroaryl urea derivatives as modulators of 5-HT2A serotonin receptors useful for the prevention and treatment of hallucinations associated with neurodegenerative diseases
JP5840324B2 (en) New uses of PAI-1 inhibitors
KR20190101365A (en) Method of treating Alport syndrome using bardoxolone methyl or an analog thereof
JP2010523682A (en) Administration of carboline derivatives useful for the treatment of cancer and other diseases
CN115154470A (en) Methods of treating depression using orexin-2 receptor antagonists
EA019919B1 (en) Method of inhibiting the growth or metastasis of an angiogenesis-dependent tumor
WO2008115443A1 (en) Kinase protein binding inhibitors
MX2013005705A (en) Therapeutic treatment for metabolic syndrome, type 2 diabetes, obestiy or prediabetes.
CA3027469C (en) Treatment of vascular calcification
US20070142308A1 (en) Angiogenesis-affecting compounds and methods for use thereof
KR20160049435A (en) Composition for reducing cell senescence comprising Atm inhibitor and use thereof
US20070142433A1 (en) Angiogenesis-affecting compounds and methods of use thereof
Fossa et al. Electrical alternans and hemodynamics in the anesthetized guinea pig can discriminate the cardiac safety of antidepressants
US20230042703A1 (en) Cardioprotective Lipid and Method of Use
KR20130064162A (en) Pharmaceutical composition for inhibiting cancer growth, recurrent or metastasis
TW201304774A (en) Pharmaceutical compositions comprising (3-(1-(1H-imidazol-4-yl)ethyl)-2-methylphenyl)methanol
KR20140145939A (en) Method for administration of a gamma secretase inhibitor
US20240342199A1 (en) Combination therapies targeting mitochondria for cancer therapy
TW202417005A (en) Cardioprotective lipid and method of use

Legal Events

Date Code Title Description
PSEA Patent sealed