WO2010006189A2 - Small-molecule inhibitors of hif and angiogenesis - Google Patents

Small-molecule inhibitors of hif and angiogenesis Download PDF

Info

Publication number
WO2010006189A2
WO2010006189A2 PCT/US2009/050137 US2009050137W WO2010006189A2 WO 2010006189 A2 WO2010006189 A2 WO 2010006189A2 US 2009050137 W US2009050137 W US 2009050137W WO 2010006189 A2 WO2010006189 A2 WO 2010006189A2
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
formula
pharmaceutically acceptable
acceptable salt
compound according
Prior art date
Application number
PCT/US2009/050137
Other languages
French (fr)
Other versions
WO2010006189A3 (en
Inventor
Erwin G. Van Meir
Kyriacos Nicolaou
Original Assignee
Emory University
The Scripps Research Institute
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 Emory University, The Scripps Research Institute filed Critical Emory University
Publication of WO2010006189A2 publication Critical patent/WO2010006189A2/en
Publication of WO2010006189A3 publication Critical patent/WO2010006189A3/en

Links

Classifications

    • 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
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • 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
    • A61P35/00Antineoplastic agents
    • 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

Definitions

  • Inducible Factor for example 2,2-dimethylbenzopyran compounds, derivatives thereof, and methods of their use including treatment of cancer, other disorders leading to ischemia (e.g., stroke and ischemic heart disease), and non-cancerous angiogenic diseases (e.g., rheumatoid arthritis and macular degeneration).
  • ischemia e.g., stroke and ischemic heart disease
  • non-cancerous angiogenic diseases e.g., rheumatoid arthritis and macular degeneration
  • Cancers are among the most common causes of death in developed countries. Despite continuing advances, existing treatments exhibit undesirable side effects and limited efficacy. Cancer is now primarily treated with one or a combination of three types of therapies: surgery; radiation; and chemotherapy.
  • Surgery involves the bulk removal of diseased tissue. While surgery is sometimes effective in removing tumors located at certain sites, for example, in the breast, colon, and skin, it cannot be used in the treatment of tumors located in other areas, such as the backbone or brainstem, nor in the treatment of disseminated neoplastic conditions such as leukemia.
  • Radiation therapy involves the exposure of living tissue to ionizing radiation causing death or damage to exposed cells. Side effects from radiation therapy may be acute and temporary, while others may be irreversible.
  • Chemotherapy involves the disruption of cell replication or cell metabolism. It is used most often in the treatment of breast, lung, and testicular cancer.
  • One of the main causes of failure in this treatment of cancer is the development of drug resistance by cancer cells, a serious problem that may lead to recurrence of disease or even death.
  • Hypoxia can pose a major hindrance to effective solid tumor therapy.
  • the microenvironment of rapidly growing solid tumors is associated with increased energy demand and diminished vascular supply, resulting in focal areas of prominent hypoxia (e.g., regions with reduced oxygen).
  • Tissue oxygen electrode measurements taken in cancer patients have shown a median range of oxygen partial pressure of 10 to 30 mmHg, with a significant proportion of readings below 2.5 mmHg, whereas those in normal tissues range from 24 to 66 mmHg.
  • radiotherapy is severely compromised in its ability to kill hypoxic tumor cells.
  • hypoxia hypoxia
  • anticancer drugs are more effective against rapidly proliferating cells than slowly or non-proliferating cells, this slowing of cell proliferation leads to decreased cell killing.
  • Chemotherapy is further affected by hypoxia as chemotherapeutic drugs are delivered systemically. The diffusion of these drugs into the tumor decreases the exposure of the hypoxic regions to the drug as compared to oxygenated cells proximal to the vessels. Hypoxia also drives genetic changes in tumors such as loss or mutation of the p53 tumor suppressor gene.
  • hypoxic regions are expected to be less amenable to immunotherapy due to their distance from nearby vessels and compromised lymphocyte function in a hypoxic environment. Tumor cells in this aberrant environment are therefore often resistant to radio- and chemotherapy.
  • Angiogenesis is a process by which new blood vessels are formed, and is essential in reproduction, development, and wound repair. Under these conditions, angiogenesis is highly regulated, so that it is turned on only as necessary, usually for brief periods of days, then completely inhibited. However, many diseases are driven by persistent unregulated angiogenesis. For example, in tumor formation, angiogenesis is a critical step for tumor growth beyond a few mm 2 and is associated with vascular leakiness and edema; in arthritis, new capillary blood vessels can invade the joint and destroy cartilage; and in diabetes, new capillaries can invade the vitreous humor, bleed, and cause blindness.
  • HIF small molecule inhibitors of HIF
  • angiogenesis for example, 2,2-dimethylbenzopyran derivatives
  • R 1 , R 2 , and R 5 are each independently H, OH, or OCi_6 alkyl;
  • R 3 is Ci_6 alkyl;
  • R 4 is H or R 4 comes together with R 3 to form a fused dioxolane or dioxane ring; and wherein at least one of R 1 or R 2 is not H; and wherein if R 1 is OH and R 2 is H then R 4 comes together with R 3 to form a fused dioxolane or dioxane ring; or a pharmaceutically acceptable salt form thereof.
  • R 1 is OH.
  • R 2 is OH.
  • R 3 is CH 3 .
  • R 3 and R 4 come together to form a fused dioxolane ring.
  • a compound according to formula I is selected from:
  • R 1 , R 2 , R 3 , and R 5 are each independently H, OH, or OCi_6 alkyl; R 4 is H or Ci_6 alkyl; and R 6 is H or OH; or a pharmaceutically acceptable salt form thereof.
  • R 1 and R 2 are H.
  • R 6 is OH.
  • R 3 and R 5 are independently an OCi_6 alkyl, and R 4 is a Ci_6 alkyl.
  • R 3 and R 5 are OCH3, and R 4 is CH3.
  • a compound according to formula II is:
  • HIF is HIF-I.
  • the cell is a non-cancerous cell.
  • a method of treating cancer in a subject comprising administering to the subject a therapeutically effective amount of a compound according to formula I or II or a pharmaceutically acceptable salt form thereof.
  • the cancer is selected from: bladder cancer, brain cancer, breast cancer, colorectal cancer, cervical cancer, gastrointestinal cancer, genitourinary cancer, head and neck cancer, lung cancer, ovarian cancer, pancreatic cancer, prostate cancer, renal cancer, skin cancer, and testicular cancer.
  • This disclosure also provides a method of inhibiting angiogenesis in a subject, the method comprising administering to the subject an effective amount of a compound according to formula I or II or a pharmaceutically acceptable salt form
  • the angiogenesis is associated with non-cancerous pathologies.
  • the noncancerous disease is selected from: atherosclerotic plaque growth and hemorrhage; chronic cystitis; Crohn's disease; diabetic retinopathy; dystrophic epidermolysis bullosa; infantile hemangiomas; intraperitoneal bleeding in endometriosis; macular degeneration; prostate growth in benign prostatic hypertrophy; psoriasis; rheumatoid arthritis; verruca vulgaris; surgical adhesions; keloids; non-cancerous lesions; aneurysms and vascular malformations in the brain; varicose veins; hemorrhoids; and rosacea.
  • the non-cancerous disease is macular degeneration.
  • a method of treating macular degeneration in a subject comprising administering to the subject a therapeutically effective amount of a compound according to formula I or II or a pharmaceutically acceptable salt form thereof.
  • This disclosure also provides a method of treating a hypoxia-related pathology in a subject, the method comprising administering to the subject a therapeutically effective amount of a compound according to formula I or II or a pharmaceutically acceptable salt form thereof.
  • a method of modulating transcription and/or translation of a nucleic acid sequence in a cell comprising contacting the cell with an effective amount of a compound according to formula I or II or a pharmaceutically acceptable salt form thereof.
  • the cell is a cancer cell.
  • the nucleic acid sequence encodes for VEGF, erythropoietin, a glucose transporter, a glycolytic enzyme, or tyrosine hydroxylase.
  • a method of modulating a basic-helix-loop-helix transcription factor in a cell comprising administering to the cell an effective amount of a compound according to formula I or II or a pharmaceutically acceptable salt form thereof.
  • a method of modulating mRNA translation in a cell comprising contacting the cell with an effective amount of a compound according to formula I or II or a pharmaceutically acceptable salt form thereof.
  • compositions comprising a pharmaceutically acceptable carrier and a compound according to formula I or II or a pharmaceutically acceptable salt form thereof.
  • the composition is administered to the eye.
  • the pharmaceutical composition is selected from ophthalmic drops, creams, ointments, installations, mucosal inserts, saturated contact lenses, diffusion release implants, and injectable solutions and suspensions.
  • Fig. 2A is a bar graph illustrating the efficacy of Compound H-A on LN229V6R cells.
  • Fig. 2B is a line graph illustrating the efficacy of Compound H-A on
  • contacting means bringing at least two moieties together, whether in an in vitro system or an in vivo system.
  • an amount of compound applied in a method refers to the amount of a compound that achieves the desired pharmacological effect or other effect, for example an amount that inhibits HIF activity resulting in a useful effect.
  • treating and “treatment” mean causing a therapeutically beneficial effect, such as ameliorating existing symptoms, preventing additional symptoms, ameliorating or preventing the underlying causes of symptoms, postponing or preventing the further development of a disorder and/or reducing the severity of symptoms that will or are expected to develop.
  • modulating means changing, adjusting, or varying a property of a molecule or pathway including increasing, decreasing, inhibiting, or activating the activity or quantity of the molecule, or activity or inhibition of a pathway.
  • subject means both mammals and non-mammals.
  • Mammals include, for example, humans; non-human primates, e.g. apes and monkeys; cattle; horses; sheep; rats; mice; pigs; and goats.
  • Non-mammals include, for example, fish and birds.
  • alkyl carbon chains if not specified, should be broadly interpreted, for example to encompass substituted or unsubstituted, straight, branched, unsaturated, and cyclic "chains.”
  • R 1 , R 2 , and R 5 are each independently H, OH, or OCi_ 6 alkyl
  • R is Ci_ 6 alkyl; R 4 is H or R 4 comes together with R 3 to form a fused dioxolane or dioxane ring; and wherein at least one of R 1 or R 2 is not H; and wherein if R 1 is OH and R 2 is H then R 4 comes together with R 3 to form a fused dioxolane or dioxane ring; or a pharmaceutically acceptable salt form thereof.
  • R 1 is OH.
  • R 2 is OH.
  • R 3 is CH 3 .
  • R 3 and R 4 come together to form a fused dioxolane ring.
  • the compound according to formula I is selected from:
  • R 1 , R 2 , R 3 , and R 5 are each independently H, OH, or OCi_ 6 alkyl; R 4 is H or Ci 6 alkyl; and R 6 is H or OH; or a pharmaceutically acceptable salt form thereof.
  • R 1 and R 2 are H.
  • R ⁇ is OH.
  • R 3 and R 5 are independently an OCi_6 alkyl, and R 4 is a Ci_6 alkyl.
  • ssoommee eemmbbooddiimmeennttss RR 33 aanndd RR 55 aarree OOCCH3, and R 4 is CH3.
  • the compound according to formula II is:
  • Compounds according to formulas I and II can be synthesized by conventional techniques using readily available starting materials.
  • a compound of formula I or II is conveniently obtained and isolated via standard organic chemistry methods.
  • compounds according to formulas I and II can be defined by three regions: region I (the right-hand aryl substituents), region II (e.g., the acyl/sulfanyl/iminyl substituent), and region III (the benzopyran aromatic ring system).
  • a 2,2-dimethylbenzopyran scaffold (region III) can be prepared using solid- phase and solution-phase methodologies (Nicolaou, K. C, Pfefferkorn, J. A.,
  • pharmaceutically acceptable salt refers to salts which possess toxicity profiles within a range that affords utility in pharmaceutical applications. Pharmaceutically unacceptable salts may nonetheless possess properties such as high crystallinity, which may render them useful, for example in processes of synthesis, purification, or formulation of compounds described herein. In general, the useful properties of the compounds described herein do not depend critically on whether the compound is or is not in a salt form. Unless clearly indicated otherwise (such as specifying that the compound should be in "free base” or “free acid” form), reference in the specification to a compound of formula I or II should be understood as encompassing salt forms of the compound, whether or not this is explicitly stated.
  • Suitable pharmaceutically-acceptable acid addition salts may be prepared from an inorganic acid or from an organic acid.
  • inorganic acids include hydrochloric, hydrobromic, hydriodic, nitric, carbonic, sulfuric, and phosphoric acids.
  • Appropriate organic acids may be selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic, and sulfonic classes of organic acids, examples of which include formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic, benzoic, anthranilic, 4-hydroxybenzoic, phenylacetic, mandelic, embonic (pamoic), methanesulfonic, ethanesulfonic, benzenesulfonic, pantothenic, trifluoromethanesulfonic, 2-hydroxyethanesulfonic, p-toluenesulfonic, sulfanilic, cyclohexylaminosulfonic, stearic, alginic, ⁇ -hydroxybutyric
  • Suitable pharmaceutically acceptable base addition salts include, for example, metallic salts including alkali metal, alkaline earth metal, and transition metal salts such as, for example, calcium, magnesium, potassium, sodium and zinc salts.
  • Pharmaceutically acceptable base addition salts also include organic salts made from basic amines such as, for example, N ⁇ -dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine ( ⁇ -methylglucamine) and procaine.
  • Examples of pharmaceutically unacceptable base addition salts include lithium salts and cyanate salts. All of these salts may be prepared by conventional means from the corresponding compound according to formula I or II by reacting, for example, the appropriate acid or base with a compound according to formula I or II.
  • the salts are in crystalline form, and preferably prepared by crystallization of the salt from a suitable solvent.
  • suitable salt forms for example, as described in Handbook of Pharmaceutical Salts: Properties, Selection, and Use By P. H. Stahl and C. G. Wermuth (Wiley-VCH 2002).
  • prodrug is meant, for example, any compound (whether itself active or inactive) that is converted chemically in vivo into a biologically active compound of formula I or II following administration of the prodrug to a subject.
  • a "prodrug” is a covalently bonded carriers which releases the active parent drug when administered to a mammalian subject.
  • Prodrugs can be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compounds.
  • Prodrugs include, for example, compounds wherein a hydroxyl group is bonded to any group that, when administered to a mammalian subject, cleaves to form a free hydroxyl group.
  • Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol functional groups in the compounds according to formula I or II.
  • prodrugs are well known by those skilled in the art. Preparation and use of prodrugs is discussed in T. Higuchi and V. Stella, "Pro-drugs as Novel Delivery Systems," Vol. 14 of the ACS
  • compositions The compounds according to formula I or II may be administered in the form of a pharmaceutical composition, in combination with a pharmaceutically acceptable carrier.
  • the amount of compound in such formulations may comprise from 0.1 to 99.99 weight percent of the composition.
  • “Pharmaceutically acceptable carrier” means any carrier, diluent or excipient which is compatible with the other ingredients of the formulation and not deleterious to the recipient.
  • the compound may be administered with a pharmaceutically acceptable carrier selected on the basis of the selected route of administration and standard pharmaceutical practice.
  • the compound may be formulated into dosage forms according to standard practices in the field of pharmaceutical preparations. See Alphonso Gennaro, ed., Remington: The Science and Practice of Pharmacy, 20th Edition (2003), Mack Publishing Co., Easton, PA.
  • Suitable dosage forms may comprise, for example, tablets, capsules, solutions, parenteral solutions, troches, suppositories, or suspensions.
  • the compound can be mixed with a suitable carrier or diluent such as water, an oil (particularly a vegetable oil), ethanol, saline solution, aqueous dextrose (glucose) and related sugar solutions, glycerol, or a glycol such as propylene glycol or polyethylene glycol.
  • Solutions for parenteral administration can contain a water soluble salt of the compound.
  • Stabilizing agents, antioxidant agents and preservatives may also be added. Suitable antioxidant agents include sulfite, ascorbic acid, citric acid and its salts, and sodium EDTA. Suitable preservatives include benzalkonium chloride, methyl- or propyl-paraben, and chlorbutanol.
  • the composition for parenteral administration can take the form of an aqueous or non-aqueous solution, dispersion, suspension, or emulsion.
  • the compound can be combined with one or more solid inactive ingredients for the preparation of tablets, capsules, pills, powders, granules, or other suitable oral dosage forms.
  • the active agent can be combined with at least one excipient such as fillers, binders, humectants, disintegrating agents, solution retarders, absorption accelerators, wetting agents absorbents, or lubricating agents.
  • the active agent can be combined with carboxymethylcellulose calcium, magnesium stearate, mannitol, and starch, and then formed into tablets by conventional tableting methods.
  • the compound can be in the form of, for example, ophthalmic drops, creams, ointments, installations, mucosal inserts, saturated contact lenses and the like, diffusion release implants, as well as solutions and suspensions suitable for injection.
  • the compound can be combined with one or more suitable carriers (e.g., a polymer or glyceride) for the preparation of a suitable ocular dosage form.
  • suitable carriers e.g., a polymer or glyceride
  • Ophthalmic solutions and suspensions can contain an aqueous vehicle or an oily vehicle.
  • the total concentration of solutes can be such that the resulting solution is isotonic with the lacrimal fluid and has a pH in the range of 6-8.
  • Ophthalmic formulations are typically sterile, and if intended for multiple dosing regimens, can be antimicrobially effective for their minimum reasonable shelf life, e.g., at least one year, and preferably two to three years or more.
  • the ingredients used in ophthalmic formulations are typically commercially available or can be made by methods readily known to those skilled in the art.
  • Pharmaceutical ophthalmic formulations can contain an effective amount, e.g., 0.001% to 10% wt/vol., most preferably 0.005% to
  • a compound of formula I or II 1% wt/vol. of a compound of formula I or II.
  • the amount of active ingredient will vary with the particular formulation and the disease state for which it is intended.
  • the compound may be administered as part of a drug delivery device, e.g., a microsphere, liposome, or silicon-based drug delivery device.
  • a drug delivery device e.g., a microsphere, liposome, or silicon-based drug delivery device.
  • the specific dose of a compound according to according to formula I or II required to obtain therapeutic benefit in the methods of treatment described herein will, of course, be determined by the particular circumstances of the individual patient including the size, weight, age, and sex of the patient, the nature and stage of the disease being treated, the aggressiveness of the disease disorder, and the route of administration of the compound.
  • a daily dosage from about 0.05 to about 50 mg/kg/day may be utilized, for example a dosage from about 0.1 to about 10 mg/kg/day. Higher or lower doses are also contemplated as it may be necessary to use dosages outside these ranges in some cases.
  • the daily dosage may be divided, such as being divided equally into two to four times per day daily dosing.
  • the compositions may be formulated in a unit dosage form, each dosage containing from about 1 to about 500 mg, more typically, about 10 to about 100 mg of compound per unit dosage.
  • unit dosage form refers to physically discrete units suitable as a unitary dosage for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
  • compositions described herein may also be formulated so as to provide slow or controlled release of the compound therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, and/or microspheres.
  • a controlled-release preparation is a pharmaceutical composition capable of releasing the compound at the required rate to maintain constant pharmacological activity for a desirable period of time.
  • dosage forms provide a supply of a drug to the body during a predetermined period of time and thus maintain drug levels in the therapeutic range for longer periods of time than conventional non- controlled formulations.
  • the controlled-release of the compound may be stimulated by various inducers, for example pH, temperature, enzymes, water, or other physiological conditions or compounds.
  • the controlled-release component may swell and form porous openings large enough to release the active ingredient after administration to a patient.
  • controlled-release component means a compound or compounds, such as polymers, polymer matrices, gels, permeable membranes, liposomes, and/or microspheres that facilitate the controlled-release of the active ingredient in the pharmaceutical composition.
  • the controlled-release component is biodegradable, induced by exposure to the aqueous environment, pH, temperature, or enzymes in the body.
  • sol-gels may be used, wherein the active ingredient is incorporated into a sol-gel matrix that is a solid at room temperature. This matrix is implanted into a patient, preferably a mammal, having a body temperature high enough to induce gel formation of the sol-gel matrix, thereby releasing the active ingredient into the patient.
  • the components used to formulate the pharmaceutical compositions are of high purity and are substantially free of potentially harmful contaminants (e.g., at least National Food grade, generally at least analytical grade, and more typically at least pharmaceutical grade). Particularly for human consumption, the composition is preferably manufactured or formulated under Good Manufacturing Practice standards as defined in the applicable regulations of the U.S. Food and Drug Administration.
  • suitable formulations may be sterile and/or substantially isotonic and/or in full compliance with all Good Manufacturing Practice regulations of the U.S. Food and Drug Administration.
  • This disclosure provides a method of modulating a basic-helix-loop-helix transcription factor comprising contacting an effective amount of compound according to formula I or II, or a salt form thereof, with a basic-helix-loop-helix transcription factor.
  • the modulation of a basic-helix-loop-helix transcription factor includes inhibition of the transcription factor.
  • the basic-helix- loop-helix transcription factor can be any basic-helix-loop-helix transcription factor, or a heterodimeric structure basic-helix-loop-helix transcription factor.
  • the basic-helix-loop-helix transcription factor can be selected from ATOHl; AhR; AHRR; ARNT; ASCLl; BHLHB2; BMAL (e.g., ARNTL, ARNTL2); CLOCK; EPASl; HAND (e.g, HAND-I and HAND-2); HES (e.g., HES-5 and HES-
  • HEY e.g., HEY-I, HEY-2, and HEY-L
  • HES-I HIF (e.g., HIF-l ⁇ and HIF-3 ⁇ ); ID (e.g., ID-I, ID-2, ID-3, ID-4); LYLl; MXD4; MYCLl; MYCN; Myogenic regulatory factors (e.g., MyoD, Myogenin, MYF-5, MYF-6); Neurogenins; NeuroD (e.g., NeuroD-1 and NeuroD-2); NPAS (NPAS-I, NPAS-2, and NPAS-3); OLIG (e.g., OLIG-I and OLIG-2); Scleraxis; TAL-I; Twist; and USF-I.
  • HIF e.g., HIF-l ⁇ and HIF-3 ⁇
  • ID e.g., ID-I, ID-2, ID-3, ID-4
  • LYLl MXD4; MYCL
  • the basic-helix-loop-helix transcription factor can be a HIF transcription factor (e.g., HIF-I, HIF-l ⁇ , HIF-l ⁇ , HIF-2, and HIF-3 ⁇ ).
  • the basic-helix-loop-helix transcription factor can be HIF-I (e.g., HIF- l ⁇ and HIF-I ⁇ ).
  • a method of modulating HIF activity comprising contacting an effective amount of compound according to formula I or II, or a salt form thereof, with HIF.
  • modulating HIF activity includes inhibition of the transcription factor.
  • modulating HIF activity includes interfering, inhibiting, or blocking signal transduction through the HIF pathway.
  • modulating HIF activity includes inhibiting HIF activity.
  • Inhibition of HIF activity can be accomplished by binding HIF, molecules associated with HIF, or molecules needed for proper HIF folding with the disclosed compounds or their derivatives to render HIF inactive or unavailable.
  • the HIF pathway can be inhibited, in whole or in part, by preventing the expression of HIF in a cell (through preventing HIF mRNA transcription, post-transcriptional modification of HIF mRNA, translation of HIF mRNA, posttranslational modification of HIF protein and HIF stability).
  • HIF inhibition can also be achieved by interfering with the binding of HIF or HIF complexes to a hypoxia responsive element (HRE).
  • HIF can be HIF-I or HIF-2.
  • HIF-I can be HIF- l ⁇ or HIF-I ⁇ .
  • HIF-2 can be HIF-2 ⁇ or HIF-2 ⁇ .
  • This disclosure also provides a method of modulating transcription and/or translation of a nucleic acid sequence (e.g., present in the genome or isolated therefrom) comprising contacting an effective amount of compound according to formula I or II, or a salt form thereof, with a cell.
  • the modulation of nucleic acid transcription or translation includes inhibition of the activity of a HIF transcription factor.
  • the inhibition of HIF activity with the disclosed compounds and compositions can occur at transcriptional, translational, and/or post-translational levels.
  • the disclosed compounds can modulate nucleic acid transcription by binding to HIF and preventing HIF from forming complexes with other molecules including DNA and proteins.
  • the disclosed compounds and compositions can bind to HIF and induce conformational changes that prevent HIF from interacting with its biological targets.
  • the disclosed compounds can bind HIF and form aggresomes or other complexes that sequester HIF or otherwise physically prevent HIF from interacting with other biological molecules.
  • the disclosed compounds and compositions can inhibit or interfere with HIF folding including, but not limited to, the inhibiting or interfering with intracellular transport of chaperone species (e.g., HSP90) from the cytoplasm to the nucleus.
  • the nucleic acid sequence can be any nucleic acid sequence, or a mixture of sequences.
  • the nucleic acid sequence can be selected from those encoding VEGF, erythropoietin, a glucose transporter (e.g., glucose transporter- 1), a glycolytic enzyme, or tyrosine hydroxylase.
  • a method of modulating mRNA translation comprising contacting an effective amount of a compound according to formula I or
  • the modulating of mRNA translation includes inhibition of a HIF transcription factor.
  • This disclosure also provides a method of inhibiting angiogenesis in a subject, comprising contacting an effective amount of a compound according to formula I or II, or a salt form thereof, with a cell.
  • the method includes inhibiting angiogenesis in a non-cancerous cell.
  • the methods described above may be performed in vitro or in vivo.
  • the methods can be performed by contacting a cell with a compound according to formula I or II, or a salt form thereof, in vitro. Contacting can be performed in the presence of cells or alternatively may be performed in a cell free medium.
  • Uses of such in vitro methods include, but are not limited to, use in a screening assay (for example, wherein the compound according to formula I or II is used as a positive control or standard compared to compounds of unknown activity or potency).
  • the methods can be performed by contacting a cell with a compound according to formula I or II, or a salt form thereof, in vivo. Contacting can be achieved by causing the compound according to formula I or II, or a salt form thereof, to be present in the subject in an amount effective to achieve the desired result. In some embodiments, an effective amount of a compound according to formula I or II, or a pharmaceutically acceptable salt form thereof, can be administered to the subject, or a prodrug of a compound according to formula I or II, or a pharmaceutically acceptable salt form thereof, can be administered to the subject.
  • the methods include, but are not limited to, use in methods of treating a disease or condition.
  • the methods may be used in a cancer cell, for example in a patient suffering from cancer.
  • the method is preferably performed by administering an effective amount of a compound according to formula I or II, or a pharmaceutically acceptable salt form thereof, to a subject who is suffering from cancer.
  • the cell can be a non-cancer cell. In any of the above described methods, in some embodiments the cell can be a cancer cell. Further provided herein is a method for treating cancer comprising administering a therapeutically effective amount of a compound according to formula I or II, or a salt form or prodrug thereof, to the subject.
  • the compounds according to formula I or II are believed effective against a broad range of cancers and tumor types, including but not limited to bladder cancer, brain cancer, breast cancer, colorectal cancer, cervical cancer, gastrointestinal cancer, genitourinary cancer, head and neck cancer, lung cancer, ovarian cancer, pancreatic cancer, prostate cancer, renal cancer, skin cancer, and testicular cancer.
  • cancers that may be treated by the compounds, compositions and methods described herein include, but are not limited to, the following: cardiac cancers, including, for example sarcoma, e.g., angiosarcoma, fibrosarcoma, rhabdomyosarcoma, and liposarcoma; myxoma; rhabdomyoma; fibroma; lipoma and teratoma; lung cancers, including, for example, bronchogenic carcinoma, e.g., squamous cell, undifferentiated small cell, undifferentiated large cell, and adenocarcinoma; alveolar and bronchiolar carcinoma; bronchial adenoma; sarcoma; lymphoma; chondromatous hamartoma; and mesothelioma; gastrointestinal cancer, including, for example, cancers of the esophagus, e.g., squamous cell carcinoma, adenos
  • neuroblastoma and cancers of the spinal cord e.g., neurofibroma, meningioma, glioma, and sarcoma
  • gynecological cancers including, for example, cancers of the uterus, e.g., endometrial carcinoma; cancers of the cervix, e.g., cervical carcinoma, and pre tumor cervical dysplasia; cancers of the ovaries, e.g., ovarian carcinoma, including serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma, granulosa thecal cell tumors, Sertoli Leydig cell tumors, dysgerminoma, and malignant teratoma; cancers of the vulva, e.g., squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, and melanoma; cancers of the vagina, e.g
  • Cancers may be solid tumors that may or may not be metastatic. Cancers may also occur, as in leukemia, as a diffuse tissue.
  • tumor cell includes a cell afflicted by any one of the above identified disorders, as well as cancer stem cells.
  • the compounds according to formula I or II can also be administered in combination with existing methods of treating cancers, for example by chemotherapy, irradiation, or surgery.
  • a method of treating cancer comprising administering a therapeutically effective amount of a compound according to formula I or II, or a salt thereof, to a subject in need of such treatment, wherein a therapeutically effective amount of at least one further cancer chemotherapeutic agent is administered to the subject.
  • chemotherapeutic agents include any of: abarelix, aldesleukin, alemtuzumab, alitretinoin, allopurinol, altretamine, anastrozole, arsenic trioxide, asparaginase, azacitidine, bevacizumab, bexarotene, bleomycin, bortezombi, bortezomib, busulfan intravenous, busulfan oral, calusterone, capecitabine, carboplatin, carmustine, cetuximab, chlorambucil, cisplatin, cladribine, clofarabine, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, dalteparin sodium, dasatinib, daunorubicin, decitabine, denileukin, denileukin diftitox, dexrazoxane, docetaxe
  • hypoxia-related pathology comprising administering to the subject a therapeutically effective amount of a compound according to formula I or II.
  • the compounds according to formula I or II are believed effective against a broad range of hypoxia-related pathologies, including but not limited to hypoxemic hypoxia, such as the hypoxia caused by sleep apnea or hypopnea, chronic obstructive pulmonary disease or respiratory arrest, and shunts; anemic hypoxia; hypemic hypoxia, for example, as the result of carbon monoxide poisoning and methaemoglobinaemia; histotoxic hypoxia; and ischemic, or stagnant hypoxia (e.g., cerebral ischemia, ischemic heart disease and intrauterine hypoxia).
  • hypoxemic hypoxia such as the hypoxia caused by sleep apnea or hypopnea, chronic obstructive pulmonary disease or respiratory arrest, and shunts
  • hypoxemic hypoxia such as the hypoxia caused by sleep apnea or
  • hypooxia-related pathology can include a pathology that is caused in part, either directly or indirectly, by conditions of below typical physiological amounts of oxygen.
  • hyperoxia-related pathology also means a pathology caused by a non-hypoxic stimuli.
  • the term includes cancer, cancer metastasis, ischemia, stroke and related conditions, diseases, or syndromes.
  • non-cancerous angiogenic diseases refers to non-cancerous diseases or conditions wherein inappropriate angiogenesis is observed as a symptom of the disease.
  • Non-limiting examples include, atherosclerotic plaque growth and hemorrhage; chronic cystitis; Crohn's disease; diabetic retinopathy; dystrophic epidermolysis bullosa; infantile hemangiomas; intraperitoneal bleeding in endometriosis; macular degeneration; prostate growth in benign prostatic hypertrophy; psoriasis; rheumatoid arthritis; verruca vulgaris; surgical adhesions; keloids; non-cancerous lesions; aneurysms and vascular malformations in the brain; varicose veins; hemorrhoids; and rosacea.
  • a compound according to formula I or II can be used to treat macular degeneration in a subject, comprising administering to the subject a therapeutically effective amount of a compound according to formula I or II, or a salt form thereof.
  • Macular degeneration can include age-related macular degeneration (AMD), dry macular degeneration, wet macular degeneration (e.g., classic choroidal neovascularization and occult choroidal neovascularization), and juvenile macular degeneration or macular dystrophy (e.g., Best's disease, Doyne's honeycomb retinal dystrophy, Sorsby's disease, and Stargardt's disease).
  • AMD age-related macular degeneration
  • dry macular degeneration e.g., wet macular degeneration
  • wet macular degeneration e.g., classic choroidal neovascularization and occult choroidal neovascularization
  • juvenile macular degeneration or macular dystrophy e.g.
  • the compounds according to formula I or II are believed effective against a broad range of pathologies associated with excessive vascularization pathologies, including those of the eye such as age-related macular degeneration (AMD) and Diabetic retinopathy.
  • the compounds according to formula I or II may be administered to subjects (mammals, including animals and humans) afflicted with a disease such as cancer or non-cancerous angiogenesis.
  • the subject treated is a human.
  • the compounds may be administered by any route, including oral, rectal, sublingual, ocular, and parenteral administration.
  • Parenteral administration includes, for example, intrathecal, intravenous, intramuscular, intraarterial, intraperitoneal, intranasal, intravaginal, intraocular, intravesical (e.g., to the bladder), intradermal, transdermal, topical, or subcutaneous administration.
  • the instillation of a drug in the body of the patient in a controlled formulation with systemic or local release of the drug to occur at a later time.
  • the drug may be localized in a depot for controlled release to the circulation, or for release to a local site, e.g., at the site of tumor growth.
  • the compounds are administered in the form of a pharmaceutical composition.
  • One or more compounds useful in the practice of the methods described herein may be administered simultaneously, by the same or different routes, or at different times during treatment.
  • the compounds may be administered before, along with, or after other medications, including other compounds.
  • the treatment using methods described herein may be carried out for as long a period as necessary, either in a single, uninterrupted session, or in discrete sessions.
  • the treating physician will know how to increase, decrease, or interrupt treatment based on patient response.
  • treatment is carried out from about four to about sixteen weeks.
  • the treatment schedule may be repeated as required.
  • kits typically include a compound of formula I or II.
  • a kit can include one or more delivery systems, e.g., for a compound of formula I or II, and directions for use of the kit (e.g., instructions for treating a subject).
  • a kit can include a compound of formula I or II and one or more additional anticancer agents.
  • a kit can include a compound of formula I or II and one or more antinausea agents.
  • the kit can include a compound of formula
  • kits can include a compound of formula I or II and one or more pain relief agents.
  • a kit can include a compound of formula I or II and a label that indicates that the contents are to be administered with an anticancer agent.
  • a kit can include a compound of formula I or II and a label that indicates that the contents are to be administered with an antinausea agent.
  • a kit can include a compound of formula I or II and a label that indicates that the contents are to be administered with a pain relief agent.
  • a kit can include a compound of formula I or II and a label that indicates that the contents are to be administered to the eye.
  • a cell-based assay was established by stably transfecting a glioma cell line with a hypoxia-inducible alkaline phosphatase (AlkPhos) expression vector (LN229-HRE-AP). Exposure of the cells to hypoxia (1% O 2 ) induced reporter gene expression, which could be detected and quantitated by a colorimetric reaction. This bioassay was used to screen 10,000 natural product-like compounds built upon a 2,2-dimethylbenzopyran scaffolding motif.
  • AlkPhos alkaline phosphatase
  • the 2,2-dimethylbenzopyran motif was chosen as a preferential synthetic scaffold for drug design because it is present in >4,000 natural products, is sufficiently lipophilic to ensure good cell membrane penetration and will generate compounds on average of less than 500 Da which are likely to cross the blood brain barrier (BBB) and reach hypoxic tumors (Nicolaou, K.C. et al. Natural Product-like Combinatorial Libraries Based on Privileged Structures 1. General Principles and
  • LN-229-HRE-luciferase glioblastoma cells containing a stably integrated reporter construct were prepared.
  • the reporter construct was made of six copies of the HIF responsive element derived from the VEGF gene cloned in front of a luciferase gene as described in Post, D.E. and Van Meir, E.G. Gene Then 8(23): 1801- 1807 (2001), which is hereby incorporated by reference in its entirety.
  • the cells were split into 48-well plates with 30,000 cells per well. The cells were then treated for one hour with either Compound I-A, Compound I-B, or Compound H-A, along with DMSO (Sigma; St. Louis, MO), DMEM media with 10% FBS (Media Tech;
  • Compound I-A was analyzed using the above assay at concentrations of 2 ⁇ M, 10 ⁇ M, 20 ⁇ M, 50 ⁇ M, and 100 ⁇ M.
  • Compound H-A was analyzed using the above assay at concentrations of 1 ⁇ M, 2 ⁇ M 5 ⁇ M, 10 ⁇ M, 20 ⁇ M, 25 ⁇ M, 50 ⁇ M, and 100 ⁇ M.
  • the values in Tables 2A and 3A illustrate the luciferase activity measured in relative light units for each compound in separate trials. The data is also expressed as percentage of control (see Tables 2B and 3B) due to the highly sensitive nature of the luciferase assay.
  • Compound H-A Table 2A. Luciferase Activity for Compounds I-A and H-A

Abstract

Inhibitors of Hypoxia Inducible Factor (HIF) and angiogenesis, for example 2,2-dimethylbenzopyran compounds and derivatives thereof, and methods of their use including treatment of cancer, disorders leading to ischemia (e.g., stroke and ischemic heart disease), and non-cancerous angiogenic diseases are provided.

Description

Small-Molecule Inhibitors of HIF and Angiogenesis
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims priority to U.S. Provisional Application No. 61/079,947, filed July 11, 2008, incorporated by reference in its entirety herein. STATEMENT REGARDING FEDERALLY FUNDED RESEARCH
This invention was made with government support under Grant Nos. ROl CAl 16804 and ROl CA46446 awarded by the National Institutes of Health. The government has certain rights in the invention.
TECHNICAL FIELD The present disclosure relates to small-molecule inhibitors of Hypoxia
Inducible Factor (HIF) and angiogenesis, for example 2,2-dimethylbenzopyran compounds, derivatives thereof, and methods of their use including treatment of cancer, other disorders leading to ischemia (e.g., stroke and ischemic heart disease), and non-cancerous angiogenic diseases (e.g., rheumatoid arthritis and macular degeneration).
BACKGROUND
Cancers are among the most common causes of death in developed countries. Despite continuing advances, existing treatments exhibit undesirable side effects and limited efficacy. Cancer is now primarily treated with one or a combination of three types of therapies: surgery; radiation; and chemotherapy. Surgery involves the bulk removal of diseased tissue. While surgery is sometimes effective in removing tumors located at certain sites, for example, in the breast, colon, and skin, it cannot be used in the treatment of tumors located in other areas, such as the backbone or brainstem, nor in the treatment of disseminated neoplastic conditions such as leukemia. Radiation therapy involves the exposure of living tissue to ionizing radiation causing death or damage to exposed cells. Side effects from radiation therapy may be acute and temporary, while others may be irreversible. Chemotherapy involves the disruption of cell replication or cell metabolism. It is used most often in the treatment of breast, lung, and testicular cancer. One of the main causes of failure in this treatment of cancer is the development of drug resistance by cancer cells, a serious problem that may lead to recurrence of disease or even death. Hypoxia can pose a major hindrance to effective solid tumor therapy. The microenvironment of rapidly growing solid tumors is associated with increased energy demand and diminished vascular supply, resulting in focal areas of prominent hypoxia (e.g., regions with reduced oxygen). Tissue oxygen electrode measurements taken in cancer patients have shown a median range of oxygen partial pressure of 10 to 30 mmHg, with a significant proportion of readings below 2.5 mmHg, whereas those in normal tissues range from 24 to 66 mmHg. In the absence of oxygen, which is the most electroaffϊnic molecule in cells and reacts chemically with the fundamental biological lesion produced by ionizing radiation, radiotherapy is severely compromised in its ability to kill hypoxic tumor cells. On the other hand, hypoxia
(and possibly hypoxia-associated deficiencies in other nutrients such as glucose) can cause tumor cells to stop or slow their rate of progression through the cell cycle. Because most anticancer drugs are more effective against rapidly proliferating cells than slowly or non-proliferating cells, this slowing of cell proliferation leads to decreased cell killing. Chemotherapy is further affected by hypoxia as chemotherapeutic drugs are delivered systemically. The diffusion of these drugs into the tumor decreases the exposure of the hypoxic regions to the drug as compared to oxygenated cells proximal to the vessels. Hypoxia also drives genetic changes in tumors such as loss or mutation of the p53 tumor suppressor gene. Finally, hypoxic regions are expected to be less amenable to immunotherapy due to their distance from nearby vessels and compromised lymphocyte function in a hypoxic environment. Tumor cells in this aberrant environment are therefore often resistant to radio- and chemotherapy.
Angiogenesis is a process by which new blood vessels are formed, and is essential in reproduction, development, and wound repair. Under these conditions, angiogenesis is highly regulated, so that it is turned on only as necessary, usually for brief periods of days, then completely inhibited. However, many diseases are driven by persistent unregulated angiogenesis. For example, in tumor formation, angiogenesis is a critical step for tumor growth beyond a few mm2 and is associated with vascular leakiness and edema; in arthritis, new capillary blood vessels can invade the joint and destroy cartilage; and in diabetes, new capillaries can invade the vitreous humor, bleed, and cause blindness. SUMMARY
Provided herein are small molecule inhibitors of HIF (e.g., HIF-I) and angiogenesis, for example, 2,2-dimethylbenzopyran derivatives, and the use of these compounds and derivatives in the treatment of cancer, hypoxia-related pathologies, and non-cancerous angiogenic diseases.
This disclosure provides a compound according to formula I:
Figure imgf000004_0001
wherein R1, R2, and R5 are each independently H, OH, or OCi_6 alkyl; R3 is Ci_6 alkyl; R4 is H or R4 comes together with R3 to form a fused dioxolane or dioxane ring; and wherein at least one of R1 or R2 is not H; and wherein if R1 is OH and R2 is H then R4 comes together with R3 to form a fused dioxolane or dioxane ring; or a pharmaceutically acceptable salt form thereof. In some embodiments, R1 is OH. In some embodiments, R2 is OH. In some embodiments, R3 is CH3. In some embodiments, R3 and R4 come together to form a fused dioxolane ring. In some embodiments, a compound according to formula I is selected from:
Figure imgf000004_0002
or a pharmaceutically acceptable salt form thereof.
Also provided herein is a compound according to formula II:
Figure imgf000005_0001
wherein R1, R2, R3, and R5 are each independently H, OH, or OCi_6 alkyl; R4 is H or Ci_6 alkyl; and R6 is H or OH; or a pharmaceutically acceptable salt form thereof. In some embodiments, R1 and R2 are H. In some embodiments, R6 is OH. In some embodiments, R3 and R5 are independently an OCi_6 alkyl, and R4 is a Ci_6 alkyl. In some embodiments, R3 and R5 are OCH3, and R4 is CH3. In some embodiments, a compound according to formula II is:
Figure imgf000005_0002
or a pharmaceutically acceptable salt form thereof. Provided herein is a method of modulating HIF activity in a cell comprising contacting the cell with an effective amount of a compound according to formula I or II or a or a pharmaceutically acceptable salt form thereof. In some embodiments, HIF is HIF-I. In some embodiments, the cell is a non-cancerous cell.
A method of treating cancer in a subject is also provided, the method comprising administering to the subject a therapeutically effective amount of a compound according to formula I or II or a pharmaceutically acceptable salt form thereof. In some embodiments, the cancer is selected from: bladder cancer, brain cancer, breast cancer, colorectal cancer, cervical cancer, gastrointestinal cancer, genitourinary cancer, head and neck cancer, lung cancer, ovarian cancer, pancreatic cancer, prostate cancer, renal cancer, skin cancer, and testicular cancer.
This disclosure also provides a method of inhibiting angiogenesis in a subject, the method comprising administering to the subject an effective amount of a compound according to formula I or II or a pharmaceutically acceptable salt form
-4- thereof. In some embodiments, the angiogenesis is associated with non-cancerous pathologies.
Also provided herein is a method of treating a non-cancerous angiogenic disease in a subject, the method comprising administering to the subject a therapeutically effective amount of a compound according to formula I or II or a pharmaceutically acceptable salt form thereof. In some embodiments, the noncancerous disease is selected from: atherosclerotic plaque growth and hemorrhage; chronic cystitis; Crohn's disease; diabetic retinopathy; dystrophic epidermolysis bullosa; infantile hemangiomas; intraperitoneal bleeding in endometriosis; macular degeneration; prostate growth in benign prostatic hypertrophy; psoriasis; rheumatoid arthritis; verruca vulgaris; surgical adhesions; keloids; non-cancerous lesions; aneurysms and vascular malformations in the brain; varicose veins; hemorrhoids; and rosacea. In some embodiments, the non-cancerous disease is macular degeneration.
Further provided herein is a method of treating macular degeneration in a subject, the method comprising administering to the subject a therapeutically effective amount of a compound according to formula I or II or a pharmaceutically acceptable salt form thereof.
This disclosure also provides a method of treating a hypoxia-related pathology in a subject, the method comprising administering to the subject a therapeutically effective amount of a compound according to formula I or II or a pharmaceutically acceptable salt form thereof.
A method of modulating transcription and/or translation of a nucleic acid sequence in a cell is provided, the method comprising contacting the cell with an effective amount of a compound according to formula I or II or a pharmaceutically acceptable salt form thereof. In some embodiments, the cell is a cancer cell. In some embodiments, the nucleic acid sequence encodes for VEGF, erythropoietin, a glucose transporter, a glycolytic enzyme, or tyrosine hydroxylase.
Provided herein is a method of modulating a basic-helix-loop-helix transcription factor in a cell, the method comprising administering to the cell an effective amount of a compound according to formula I or II or a pharmaceutically acceptable salt form thereof. Also provided herein is a method of modulating mRNA translation in a cell comprising contacting the cell with an effective amount of a compound according to formula I or II or a pharmaceutically acceptable salt form thereof.
A method of treating excessive vascularization in a subject is provided, the method comprising administering to the subject a therapeutically effective amount of a compound according to formula I or II or a pharmaceutically acceptable salt form thereof. In some embodiments, the excessive vascularization is associated with noncancerous pathologies.
This disclosure further provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound according to formula I or II or a pharmaceutically acceptable salt form thereof. In some embodiments, the composition is administered to the eye. In some embodiments, the pharmaceutical composition is selected from ophthalmic drops, creams, ointments, installations, mucosal inserts, saturated contact lenses, diffusion release implants, and injectable solutions and suspensions.
The details of one or more embodiments of the invention are set forth in the accompanying drawings and the description below. Other features, objects, and advantages of the invention will be apparent from the description and drawings, and from the claims. DESCRIPTION OF DRAWINGS
Fig. 1 is a bar graph illustrating the efficacy of Compounds I-A and H-A on LN229V6R cells.
Fig. 2A is a bar graph illustrating the efficacy of Compound H-A on LN229V6R cells. Fig. 2B is a line graph illustrating the efficacy of Compound H-A on
LN229V6R cells.
DETAILED DESCRIPTION I. Definitions
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art to which this disclosure belongs. All patents, applications, published applications, and other publications are incorporated by reference in their entirety. In the event that there is a plurality of definitions for a term herein, those in this section prevail unless stated otherwise.
The term "contacting" means bringing at least two moieties together, whether in an in vitro system or an in vivo system.
The expression "effective amount," when used to describe an amount of compound applied in a method, refers to the amount of a compound that achieves the desired pharmacological effect or other effect, for example an amount that inhibits HIF activity resulting in a useful effect.
The terms "treating" and "treatment" mean causing a therapeutically beneficial effect, such as ameliorating existing symptoms, preventing additional symptoms, ameliorating or preventing the underlying causes of symptoms, postponing or preventing the further development of a disorder and/or reducing the severity of symptoms that will or are expected to develop.
The term "modulating" as used herein means changing, adjusting, or varying a property of a molecule or pathway including increasing, decreasing, inhibiting, or activating the activity or quantity of the molecule, or activity or inhibition of a pathway.
As used herein, "subject" (as in the subject of the treatment) means both mammals and non-mammals. Mammals include, for example, humans; non-human primates, e.g. apes and monkeys; cattle; horses; sheep; rats; mice; pigs; and goats. Non-mammals include, for example, fish and birds.
As used herein, "alkyl" carbon chains, if not specified, should be broadly interpreted, for example to encompass substituted or unsubstituted, straight, branched, unsaturated, and cyclic "chains."
II. Compounds
Provided herein are compounds according to formula I:
Figure imgf000008_0001
wherein:
R1, R2, and R5 are each independently H, OH, or OCi_6 alkyl;
R is Ci_6 alkyl; R4 is H or R4 comes together with R3 to form a fused dioxolane or dioxane ring; and wherein at least one of R1 or R2 is not H; and wherein if R1 is OH and R2 is H then R4 comes together with R3 to form a fused dioxolane or dioxane ring; or a pharmaceutically acceptable salt form thereof.
In some embodiments, R1 is OH. In some embodiments, R2 is OH. In some embodiments, R3 is CH3. In some embodiments, R3 and R4 come together to form a fused dioxolane ring. In some embodiments, the compound according to formula I is selected from:
Figure imgf000009_0001
(I-A); and
Figure imgf000009_0002
or a pharmaceutically acceptable salt form thereof.
Further provided herein are compounds according to formula II:
Figure imgf000009_0003
wherein:
R1, R2, R3, and R5 are each independently H, OH, or OCi_6 alkyl; R4 is H or Ci 6 alkyl; and R6 is H or OH; or a pharmaceutically acceptable salt form thereof. In some embodiments, R1 and R2 are H. In some embodiments, R^ is OH. In some embodiments, R3 and R5 are independently an OCi_6 alkyl, and R4 is a Ci_6 alkyl. In ssoommee eemmbbooddiimmeennttss,, RR33 aanndd RR55 aarree OOCCH3, and R4 is CH3. In some embodiments, the compound according to formula II is:
Figure imgf000010_0001
(H-A) or a pharmaceutically acceptable salt form thereof.
Compounds according to formulas I and II can be synthesized by conventional techniques using readily available starting materials. In general, a compound of formula I or II is conveniently obtained and isolated via standard organic chemistry methods. For example, compounds according to formulas I and II can be defined by three regions: region I (the right-hand aryl substituents), region II (e.g., the acyl/sulfanyl/iminyl substituent), and region III (the benzopyran aromatic ring system). A 2,2-dimethylbenzopyran scaffold (region III) can be prepared using solid- phase and solution-phase methodologies (Nicolaou, K. C, Pfefferkorn, J. A.,
Mitchell, H. J., Roecker, A. J., Barluenga, S., Cao, G.-Q., Affleck, R. L. & Lillig, J. E. Natural product- like combinatorial libraries based on privileged structures. 2. Construction of a 10,000-membered benzopyran library by directed split-and-pool chemistry using NanoKans and optical encoding. J Am Chem Soc 122, 9954-67 (2000), which is incorporated herein by reference). These scaffolds, containing an aldehyde functionality, can be subsequently modified using short synthetic sequences which can provide for regions I and II: Grignard addition followed by acylation; or reductive animation followed by imine formation. Further diversification on region III can be achieved through further reactivity at the carbon-carbon double bond of the pyran ring. Compounds can be synthesized using parallel synthesis, purified by common chromatographic techniques, and characterized by LC- or GC-MS.
The term "pharmaceutically acceptable salt" refers to salts which possess toxicity profiles within a range that affords utility in pharmaceutical applications. Pharmaceutically unacceptable salts may nonetheless possess properties such as high crystallinity, which may render them useful, for example in processes of synthesis, purification, or formulation of compounds described herein. In general, the useful properties of the compounds described herein do not depend critically on whether the compound is or is not in a salt form. Unless clearly indicated otherwise (such as specifying that the compound should be in "free base" or "free acid" form), reference in the specification to a compound of formula I or II should be understood as encompassing salt forms of the compound, whether or not this is explicitly stated.
Suitable pharmaceutically-acceptable acid addition salts may be prepared from an inorganic acid or from an organic acid. Examples of inorganic acids include hydrochloric, hydrobromic, hydriodic, nitric, carbonic, sulfuric, and phosphoric acids. Appropriate organic acids may be selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic, and sulfonic classes of organic acids, examples of which include formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic, benzoic, anthranilic, 4-hydroxybenzoic, phenylacetic, mandelic, embonic (pamoic), methanesulfonic, ethanesulfonic, benzenesulfonic, pantothenic, trifluoromethanesulfonic, 2-hydroxyethanesulfonic, p-toluenesulfonic, sulfanilic, cyclohexylaminosulfonic, stearic, alginic, β-hydroxybutyric, salicylic, galactaric, and galacturonic acid. Examples of pharmaceutically unacceptable acid addition salts include, for example, perchlorates and tetrafluoroborates.
Suitable pharmaceutically acceptable base addition salts include, for example, metallic salts including alkali metal, alkaline earth metal, and transition metal salts such as, for example, calcium, magnesium, potassium, sodium and zinc salts. Pharmaceutically acceptable base addition salts also include organic salts made from basic amines such as, for example, N^-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (Ν-methylglucamine) and procaine. Examples of pharmaceutically unacceptable base addition salts include lithium salts and cyanate salts. All of these salts may be prepared by conventional means from the corresponding compound according to formula I or II by reacting, for example, the appropriate acid or base with a compound according to formula I or II. Preferably the salts are in crystalline form, and preferably prepared by crystallization of the salt from a suitable solvent. A person skilled in the art will know how to prepare and select suitable salt forms for example, as described in Handbook of Pharmaceutical Salts: Properties, Selection, and Use By P. H. Stahl and C. G. Wermuth (Wiley-VCH 2002).
The compounds according to formula I or II, and salts thereof may be administered in the form of prodrugs. By "prodrug" is meant, for example, any compound (whether itself active or inactive) that is converted chemically in vivo into a biologically active compound of formula I or II following administration of the prodrug to a subject.
Generally a "prodrug" is a covalently bonded carriers which releases the active parent drug when administered to a mammalian subject. Prodrugs can be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compounds. Prodrugs include, for example, compounds wherein a hydroxyl group is bonded to any group that, when administered to a mammalian subject, cleaves to form a free hydroxyl group. Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol functional groups in the compounds according to formula I or II.
The suitability and techniques involved in making and using prodrugs are well known by those skilled in the art. Preparation and use of prodrugs is discussed in T. Higuchi and V. Stella, "Pro-drugs as Novel Delivery Systems," Vol. 14 of the ACS
Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are hereby incorporated by reference in their entirety.
III. Pharmaceutical Compositions The compounds according to formula I or II may be administered in the form of a pharmaceutical composition, in combination with a pharmaceutically acceptable carrier. The amount of compound in such formulations may comprise from 0.1 to 99.99 weight percent of the composition. "Pharmaceutically acceptable carrier" means any carrier, diluent or excipient which is compatible with the other ingredients of the formulation and not deleterious to the recipient.
The compound may be administered with a pharmaceutically acceptable carrier selected on the basis of the selected route of administration and standard pharmaceutical practice. The compound may be formulated into dosage forms according to standard practices in the field of pharmaceutical preparations. See Alphonso Gennaro, ed., Remington: The Science and Practice of Pharmacy, 20th Edition (2003), Mack Publishing Co., Easton, PA. Suitable dosage forms may comprise, for example, tablets, capsules, solutions, parenteral solutions, troches, suppositories, or suspensions.
For parenteral administration, the compound can be mixed with a suitable carrier or diluent such as water, an oil (particularly a vegetable oil), ethanol, saline solution, aqueous dextrose (glucose) and related sugar solutions, glycerol, or a glycol such as propylene glycol or polyethylene glycol. Solutions for parenteral administration can contain a water soluble salt of the compound. Stabilizing agents, antioxidant agents and preservatives may also be added. Suitable antioxidant agents include sulfite, ascorbic acid, citric acid and its salts, and sodium EDTA. Suitable preservatives include benzalkonium chloride, methyl- or propyl-paraben, and chlorbutanol. The composition for parenteral administration can take the form of an aqueous or non-aqueous solution, dispersion, suspension, or emulsion.
For oral administration, the compound can be combined with one or more solid inactive ingredients for the preparation of tablets, capsules, pills, powders, granules, or other suitable oral dosage forms. For example, the active agent can be combined with at least one excipient such as fillers, binders, humectants, disintegrating agents, solution retarders, absorption accelerators, wetting agents absorbents, or lubricating agents. According to one tablet embodiment, the active agent can be combined with carboxymethylcellulose calcium, magnesium stearate, mannitol, and starch, and then formed into tablets by conventional tableting methods. For ocular administration, the compound can be in the form of, for example, ophthalmic drops, creams, ointments, installations, mucosal inserts, saturated contact lenses and the like, diffusion release implants, as well as solutions and suspensions suitable for injection.. The compound can be combined with one or more suitable carriers (e.g., a polymer or glyceride) for the preparation of a suitable ocular dosage form. For example, see U.S. Patent No. 5,766,619 and 6,378,526. Ophthalmic solutions and suspensions can contain an aqueous vehicle or an oily vehicle. In some embodiments, the total concentration of solutes can be such that the resulting solution is isotonic with the lacrimal fluid and has a pH in the range of 6-8. Ophthalmic formulations are typically sterile, and if intended for multiple dosing regimens, can be antimicrobially effective for their minimum reasonable shelf life, e.g., at least one year, and preferably two to three years or more. The ingredients used in ophthalmic formulations are typically commercially available or can be made by methods readily known to those skilled in the art. Pharmaceutical ophthalmic formulations can contain an effective amount, e.g., 0.001% to 10% wt/vol., most preferably 0.005% to
1% wt/vol. of a compound of formula I or II. The amount of active ingredient will vary with the particular formulation and the disease state for which it is intended. In some embodiments, the compound may be administered as part of a drug delivery device, e.g., a microsphere, liposome, or silicon-based drug delivery device. The specific dose of a compound according to according to formula I or II required to obtain therapeutic benefit in the methods of treatment described herein will, of course, be determined by the particular circumstances of the individual patient including the size, weight, age, and sex of the patient, the nature and stage of the disease being treated, the aggressiveness of the disease disorder, and the route of administration of the compound.
For example, a daily dosage from about 0.05 to about 50 mg/kg/day may be utilized, for example a dosage from about 0.1 to about 10 mg/kg/day. Higher or lower doses are also contemplated as it may be necessary to use dosages outside these ranges in some cases. The daily dosage may be divided, such as being divided equally into two to four times per day daily dosing. The compositions may be formulated in a unit dosage form, each dosage containing from about 1 to about 500 mg, more typically, about 10 to about 100 mg of compound per unit dosage. The term "unit dosage form" refers to physically discrete units suitable as a unitary dosage for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
The pharmaceutical compositions described herein may also be formulated so as to provide slow or controlled release of the compound therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, and/or microspheres.
In general, a controlled-release preparation is a pharmaceutical composition capable of releasing the compound at the required rate to maintain constant pharmacological activity for a desirable period of time. Such dosage forms provide a supply of a drug to the body during a predetermined period of time and thus maintain drug levels in the therapeutic range for longer periods of time than conventional non- controlled formulations. The controlled-release of the compound may be stimulated by various inducers, for example pH, temperature, enzymes, water, or other physiological conditions or compounds. Various mechanisms of drug release exist. For example, in one embodiment, the controlled-release component may swell and form porous openings large enough to release the active ingredient after administration to a patient. The term "controlled-release component" means a compound or compounds, such as polymers, polymer matrices, gels, permeable membranes, liposomes, and/or microspheres that facilitate the controlled-release of the active ingredient in the pharmaceutical composition. In another embodiment, the controlled-release component is biodegradable, induced by exposure to the aqueous environment, pH, temperature, or enzymes in the body. In another embodiment, sol-gels may be used, wherein the active ingredient is incorporated into a sol-gel matrix that is a solid at room temperature. This matrix is implanted into a patient, preferably a mammal, having a body temperature high enough to induce gel formation of the sol-gel matrix, thereby releasing the active ingredient into the patient. The components used to formulate the pharmaceutical compositions are of high purity and are substantially free of potentially harmful contaminants (e.g., at least National Food grade, generally at least analytical grade, and more typically at least pharmaceutical grade). Particularly for human consumption, the composition is preferably manufactured or formulated under Good Manufacturing Practice standards as defined in the applicable regulations of the U.S. Food and Drug Administration.
For example, suitable formulations may be sterile and/or substantially isotonic and/or in full compliance with all Good Manufacturing Practice regulations of the U.S. Food and Drug Administration.
IV. Methods of Use Provided herein are methods of using the compounds according to formula I or
II comprising administering a compound according to formula I or II, or a pharmaceutically acceptable salt form thereof, to a subject. This disclosure provides a method of modulating a basic-helix-loop-helix transcription factor comprising contacting an effective amount of compound according to formula I or II, or a salt form thereof, with a basic-helix-loop-helix transcription factor. In some embodiments, the modulation of a basic-helix-loop-helix transcription factor includes inhibition of the transcription factor. The basic-helix- loop-helix transcription factor can be any basic-helix-loop-helix transcription factor, or a heterodimeric structure basic-helix-loop-helix transcription factor. In some embodiments, the basic-helix-loop-helix transcription factor can be selected from ATOHl; AhR; AHRR; ARNT; ASCLl; BHLHB2; BMAL (e.g., ARNTL, ARNTL2); CLOCK; EPASl; HAND (e.g, HAND-I and HAND-2); HES (e.g., HES-5 and HES-
6); HEY (e.g., HEY-I, HEY-2, and HEY-L); HES-I; HIF (e.g., HIF-lα and HIF-3α); ID (e.g., ID-I, ID-2, ID-3, ID-4); LYLl; MXD4; MYCLl; MYCN; Myogenic regulatory factors (e.g., MyoD, Myogenin, MYF-5, MYF-6); Neurogenins; NeuroD (e.g., NeuroD-1 and NeuroD-2); NPAS (NPAS-I, NPAS-2, and NPAS-3); OLIG (e.g., OLIG-I and OLIG-2); Scleraxis; TAL-I; Twist; and USF-I. In some embodiments, the basic-helix-loop-helix transcription factor can be a HIF transcription factor (e.g., HIF-I, HIF-lα, HIF-lβ, HIF-2, and HIF-3α). In some embodiments, the basic-helix-loop-helix transcription factor can be HIF-I (e.g., HIF- lα and HIF-I β). Also provided herein is a method of modulating HIF activity comprising contacting an effective amount of compound according to formula I or II, or a salt form thereof, with HIF. In some embodiments, modulating HIF activity includes inhibition of the transcription factor. In some embodiments, modulating HIF activity includes interfering, inhibiting, or blocking signal transduction through the HIF pathway. In some embodiments, modulating HIF activity includes inhibiting HIF activity. Inhibition of HIF activity can be accomplished by binding HIF, molecules associated with HIF, or molecules needed for proper HIF folding with the disclosed compounds or their derivatives to render HIF inactive or unavailable. Alternatively, the HIF pathway can be inhibited, in whole or in part, by preventing the expression of HIF in a cell (through preventing HIF mRNA transcription, post-transcriptional modification of HIF mRNA, translation of HIF mRNA, posttranslational modification of HIF protein and HIF stability). HIF inhibition can also be achieved by interfering with the binding of HIF or HIF complexes to a hypoxia responsive element (HRE). In some embodiments, HIF can be HIF-I or HIF-2. In some embodiments, HIF-I can be HIF- lα or HIF-I β. In some embodiments, HIF-2 can be HIF-2α or HIF-2β.
This disclosure also provides a method of modulating transcription and/or translation of a nucleic acid sequence (e.g., present in the genome or isolated therefrom) comprising contacting an effective amount of compound according to formula I or II, or a salt form thereof, with a cell. In some embodiments, the modulation of nucleic acid transcription or translation includes inhibition of the activity of a HIF transcription factor. The inhibition of HIF activity with the disclosed compounds and compositions can occur at transcriptional, translational, and/or post-translational levels. The disclosed compounds can modulate nucleic acid transcription by binding to HIF and preventing HIF from forming complexes with other molecules including DNA and proteins. For example, the disclosed compounds and compositions can bind to HIF and induce conformational changes that prevent HIF from interacting with its biological targets. Alternatively, the disclosed compounds can bind HIF and form aggresomes or other complexes that sequester HIF or otherwise physically prevent HIF from interacting with other biological molecules. Finally, the disclosed compounds and compositions can inhibit or interfere with HIF folding including, but not limited to, the inhibiting or interfering with intracellular transport of chaperone species (e.g., HSP90) from the cytoplasm to the nucleus. The nucleic acid sequence can be any nucleic acid sequence, or a mixture of sequences. In some embodiments, the nucleic acid sequence can be selected from those encoding VEGF, erythropoietin, a glucose transporter (e.g., glucose transporter- 1), a glycolytic enzyme, or tyrosine hydroxylase.
A method of modulating mRNA translation is provided, the method comprising contacting an effective amount of a compound according to formula I or
II, or a salt form thereof, with a cell. In some embodiments, the modulating of mRNA translation includes inhibition of a HIF transcription factor.
This disclosure also provides a method of inhibiting angiogenesis in a subject, comprising contacting an effective amount of a compound according to formula I or II, or a salt form thereof, with a cell. In some embodiments, the method includes inhibiting angiogenesis in a non-cancerous cell.
The methods described above may be performed in vitro or in vivo. In some embodiments, the methods can be performed by contacting a cell with a compound according to formula I or II, or a salt form thereof, in vitro. Contacting can be performed in the presence of cells or alternatively may be performed in a cell free medium. Uses of such in vitro methods include, but are not limited to, use in a screening assay (for example, wherein the compound according to formula I or II is used as a positive control or standard compared to compounds of unknown activity or potency).
In some embodiments, the methods can be performed by contacting a cell with a compound according to formula I or II, or a salt form thereof, in vivo. Contacting can be achieved by causing the compound according to formula I or II, or a salt form thereof, to be present in the subject in an amount effective to achieve the desired result. In some embodiments, an effective amount of a compound according to formula I or II, or a pharmaceutically acceptable salt form thereof, can be administered to the subject, or a prodrug of a compound according to formula I or II, or a pharmaceutically acceptable salt form thereof, can be administered to the subject.
Uses of such in vivo methods include, but are not limited to, use in methods of treating a disease or condition. In some embodiments, the methods may be used in a cancer cell, for example in a patient suffering from cancer. The method is preferably performed by administering an effective amount of a compound according to formula I or II, or a pharmaceutically acceptable salt form thereof, to a subject who is suffering from cancer.
In any of the above described methods, in some embodiments the cell can be a non-cancer cell. In any of the above described methods, in some embodiments the cell can be a cancer cell. Further provided herein is a method for treating cancer comprising administering a therapeutically effective amount of a compound according to formula I or II, or a salt form or prodrug thereof, to the subject.
The compounds according to formula I or II are believed effective against a broad range of cancers and tumor types, including but not limited to bladder cancer, brain cancer, breast cancer, colorectal cancer, cervical cancer, gastrointestinal cancer, genitourinary cancer, head and neck cancer, lung cancer, ovarian cancer, pancreatic cancer, prostate cancer, renal cancer, skin cancer, and testicular cancer. More particularly, cancers that may be treated by the compounds, compositions and methods described herein include, but are not limited to, the following: cardiac cancers, including, for example sarcoma, e.g., angiosarcoma, fibrosarcoma, rhabdomyosarcoma, and liposarcoma; myxoma; rhabdomyoma; fibroma; lipoma and teratoma; lung cancers, including, for example, bronchogenic carcinoma, e.g., squamous cell, undifferentiated small cell, undifferentiated large cell, and adenocarcinoma; alveolar and bronchiolar carcinoma; bronchial adenoma; sarcoma; lymphoma; chondromatous hamartoma; and mesothelioma; gastrointestinal cancer, including, for example, cancers of the esophagus, e.g., squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, and lymphoma; cancers of the stomach, e.g., carcinoma, lymphoma, and leiomyosarcoma; cancers of the pancreas, e.g., ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, and vipoma; cancers of the small bowel, e.g., adenocarcinoma, lymphoma, carcinoid tumors, Kaposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, and fibroma; cancers of the large bowel, e.g., adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, and leiomyoma; genitourinary tract cancers, including, for example, cancers of the kidney, e.g., adenocarcinoma, Wilm's tumor (nephroblastoma), lymphoma, and leukemia; cancers of the bladder and urethra, e.g., squamous cell carcinoma, transitional cell carcinoma, and adenocarcinoma; cancers of the prostate, e.g., adenocarcinoma, and sarcoma; cancer of the testis, e.g., seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, and lipoma; liver cancers, including, for example, hepatoma, e.g., hepatocellular carcinoma; cholangiocarcinoma; hepatoblastoma; angiosarcoma; hepatocellular adenoma; and hemangioma; bone cancers, including, for example, osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochrondroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors; nervous system cancers, including, for example, cancers of the skull, e.g., osteoma, hemangioma, granuloma, xanthoma, and osteitis deformans; cancers of the meninges, e.g., meningioma, meningiosarcoma, and gliomatosis; cancers of the brain, e.g., astrocytoma, medulloblastoma, glioma, ependymoma, germinoma (pinealoma), glioblastoma multiforme, oligodendroglioma, PNET, schwannoma, retinoblastoma, neuroma and congenital tumors; neural crest derived cancers, e.g. neuroblastoma and cancers of the spinal cord, e.g., neurofibroma, meningioma, glioma, and sarcoma; gynecological cancers, including, for example, cancers of the uterus, e.g., endometrial carcinoma; cancers of the cervix, e.g., cervical carcinoma, and pre tumor cervical dysplasia; cancers of the ovaries, e.g., ovarian carcinoma, including serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma, granulosa thecal cell tumors, Sertoli Leydig cell tumors, dysgerminoma, and malignant teratoma; cancers of the vulva, e.g., squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, and melanoma; cancers of the vagina, e.g., clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma, and embryonal rhabdomyosarcoma; and cancers of the fallopian tubes, e.g., carcinoma; hematologic cancers, including, for example, cancers of the blood, e.g., acute myeloid leukemia, chronic myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, and myelodysplastic syndrome, Hodgkin's lymphoma, non Hodgkin's lymphoma (malignant lymphoma) and Waldenstrom's macroglobulinemia; skin cancers, including, for example, malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Kaposi's sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis; and adrenal gland cancers, including, for example, neuroblastoma. Cancers may be solid tumors that may or may not be metastatic. Cancers may also occur, as in leukemia, as a diffuse tissue. Thus, the term "tumor cell", as provided herein, includes a cell afflicted by any one of the above identified disorders, as well as cancer stem cells.
The compounds according to formula I or II can also be administered in combination with existing methods of treating cancers, for example by chemotherapy, irradiation, or surgery. Thus, there is further provided a method of treating cancer comprising administering a therapeutically effective amount of a compound according to formula I or II, or a salt thereof, to a subject in need of such treatment, wherein a therapeutically effective amount of at least one further cancer chemotherapeutic agent is administered to the subject. Examples of suitable chemotherapeutic agents include any of: abarelix, aldesleukin, alemtuzumab, alitretinoin, allopurinol, altretamine, anastrozole, arsenic trioxide, asparaginase, azacitidine, bevacizumab, bexarotene, bleomycin, bortezombi, bortezomib, busulfan intravenous, busulfan oral, calusterone, capecitabine, carboplatin, carmustine, cetuximab, chlorambucil, cisplatin, cladribine, clofarabine, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, dalteparin sodium, dasatinib, daunorubicin, decitabine, denileukin, denileukin diftitox, dexrazoxane, docetaxel, doxorubicin, dromostanolone propionate, eculizumab, epirubicin, erlotinib, estramustine, etoposide phosphate, etoposide, exemestane, fentanyl citrate, filgrastim, floxuridine, fludarabine, fluorouracil, fulvestrant, gefϊtinib, geldanamycin derivatives (e.g., 17-AAG or 17-DMAG), gemcitabine, gemtuzumab ozogamicin, goserelin acetate, histrelin acetate, ibritumomab tiuxetan, idarubicin, ifosfamide, imatinib mesylate, interferon alfa 2a, irinotecan, lapatinib ditosylate, lenalidomide, letrozole, leucovorin, leuprolide acetate, levamisole, lomustine, meclorethamine, megestrol acetate, melphalan, mercaptopurine, methotrexate, methoxsalen, mitomycin C, mitotane, mitoxantrone, nandrolone phenpropionate, nelarabine, nofetumomab, oxaliplatin, paclitaxel, pamidronate, panitumumab, pegaspargase, pegfϊlgrastim, pemetrexed disodium, pentostatin, pipobroman, plicamycin, procarbazine, quinacrine, radicicol (Hsp90 inhibitor), rasburicase, rituximab, sorafenib, streptozocin, sunitinib, sunitinib maleate, tamoxifen, temozolomide, teniposide, testolactone, thalidomide, thioguanine, thiotepa, topotecan, toremifene, tositumomab, trastuzumab, tretinoin, uracil mustard, valrubicin, vinblastine, vincristine, vinorelbine, vorinostat, and zoledronate.
Further provided herein is a method of treating a hypoxia-related pathology in a subject, comprising administering to the subject a therapeutically effective amount of a compound according to formula I or II. The compounds according to formula I or II are believed effective against a broad range of hypoxia-related pathologies, including but not limited to hypoxemic hypoxia, such as the hypoxia caused by sleep apnea or hypopnea, chronic obstructive pulmonary disease or respiratory arrest, and shunts; anemic hypoxia; hypemic hypoxia, for example, as the result of carbon monoxide poisoning and methaemoglobinaemia; histotoxic hypoxia; and ischemic, or stagnant hypoxia (e.g., cerebral ischemia, ischemic heart disease and intrauterine hypoxia). The term "hypoxia-related pathology" can include a pathology that is caused in part, either directly or indirectly, by conditions of below typical physiological amounts of oxygen. The term "hypoxia-related pathology" also means a pathology caused by a non-hypoxic stimuli. The term includes cancer, cancer metastasis, ischemia, stroke and related conditions, diseases, or syndromes.
This disclosure also provides a method of treating non-cancerous angiogenic diseases in a subject, comprising administering to the subject a therapeutically effective amount of a compound according to formula I or II, or a salt form thereof. As used herein, "non-cancerous angiogenic diseases" refers to non-cancerous diseases or conditions wherein inappropriate angiogenesis is observed as a symptom of the disease. Non-limiting examples include, atherosclerotic plaque growth and hemorrhage; chronic cystitis; Crohn's disease; diabetic retinopathy; dystrophic epidermolysis bullosa; infantile hemangiomas; intraperitoneal bleeding in endometriosis; macular degeneration; prostate growth in benign prostatic hypertrophy; psoriasis; rheumatoid arthritis; verruca vulgaris; surgical adhesions; keloids; non-cancerous lesions; aneurysms and vascular malformations in the brain; varicose veins; hemorrhoids; and rosacea.
In some embodiments, a compound according to formula I or II can be used to treat macular degeneration in a subject, comprising administering to the subject a therapeutically effective amount of a compound according to formula I or II, or a salt form thereof. Macular degeneration can include age-related macular degeneration (AMD), dry macular degeneration, wet macular degeneration (e.g., classic choroidal neovascularization and occult choroidal neovascularization), and juvenile macular degeneration or macular dystrophy (e.g., Best's disease, Doyne's honeycomb retinal dystrophy, Sorsby's disease, and Stargardt's disease).
Also provided herein is a method of treating excessive vascularization in a subject, comprising administering to the subject a therapeutically effective amount of a compound according to formula I or II. The compounds according to formula I or II are believed effective against a broad range of pathologies associated with excessive vascularization pathologies, including those of the eye such as age-related macular degeneration (AMD) and Diabetic retinopathy. In the methods of treatment described herein, the compounds according to formula I or II may be administered to subjects (mammals, including animals and humans) afflicted with a disease such as cancer or non-cancerous angiogenesis. In particular embodiments, the subject treated is a human.
The compounds may be administered by any route, including oral, rectal, sublingual, ocular, and parenteral administration. Parenteral administration includes, for example, intrathecal, intravenous, intramuscular, intraarterial, intraperitoneal, intranasal, intravaginal, intraocular, intravesical (e.g., to the bladder), intradermal, transdermal, topical, or subcutaneous administration. Also contemplated is the instillation of a drug in the body of the patient in a controlled formulation, with systemic or local release of the drug to occur at a later time. For example, the drug may be localized in a depot for controlled release to the circulation, or for release to a local site, e.g., at the site of tumor growth. Advantageously, the compounds are administered in the form of a pharmaceutical composition.
One or more compounds useful in the practice of the methods described herein may be administered simultaneously, by the same or different routes, or at different times during treatment. The compounds may be administered before, along with, or after other medications, including other compounds.
The treatment using methods described herein may be carried out for as long a period as necessary, either in a single, uninterrupted session, or in discrete sessions. The treating physician will know how to increase, decrease, or interrupt treatment based on patient response. According to one embodiment, treatment is carried out from about four to about sixteen weeks. The treatment schedule may be repeated as required. There is additionally provided a compound according to formula I or II, or any of the embodiments thereof, or a salt thereof, for use in any of the aforementioned methods of treatment, or for use in treatment of any of the aforementioned diseases or conditions. Also provided is a use of a compound according to formula I or II, or any of the embodiments thereof, or a salt thereof, for use in the manufacture of a medicament, for use in any of the aforementioned methods of treatment, or for use in of any of the aforementioned diseases or conditions.
V. Kits
Also provided herein are kits. Typically, a kit includes a compound of formula I or II. In certain embodiments, a kit can include one or more delivery systems, e.g., for a compound of formula I or II, and directions for use of the kit (e.g., instructions for treating a subject). In certain embodiments, a kit can include a compound of formula I or II and one or more additional anticancer agents. In another embodiment, a kit can include a compound of formula I or II and one or more antinausea agents. In some embodiments, the kit can include a compound of formula
I or II and one or more pain relief agents. In a further embodiment, a kit can include a compound of formula I or II and a label that indicates that the contents are to be administered with an anticancer agent. In another embodiment, a kit can include a compound of formula I or II and a label that indicates that the contents are to be administered with an antinausea agent. In some embodiments, a kit can include a compound of formula I or II and a label that indicates that the contents are to be administered with a pain relief agent. In some embodiments, a kit can include a compound of formula I or II and a label that indicates that the contents are to be administered to the eye. EXAMPLES
Example 1: Alkaline Phosphatase Assay
To screen for small molecule HIF-I pathway inhibitors, a cell-based assay was established by stably transfecting a glioma cell line with a hypoxia-inducible alkaline phosphatase (AlkPhos) expression vector (LN229-HRE-AP). Exposure of the cells to hypoxia (1% O2) induced reporter gene expression, which could be detected and quantitated by a colorimetric reaction. This bioassay was used to screen 10,000 natural product-like compounds built upon a 2,2-dimethylbenzopyran scaffolding motif. The 2,2-dimethylbenzopyran motif was chosen as a preferential synthetic scaffold for drug design because it is present in >4,000 natural products, is sufficiently lipophilic to ensure good cell membrane penetration and will generate compounds on average of less than 500 Da which are likely to cross the blood brain barrier (BBB) and reach hypoxic tumors (Nicolaou, K.C. et al. Natural Product-like Combinatorial Libraries Based on Privileged Structures 1. General Principles and
Solid-Phase Synthesis of Benzopyrans. J. Am. Chem. Soc. 122:9939-9953 (2000); Nicolaou, K.C. et al. Natural Product-like Combinatorial Libraries Based on Privileged Structures. 2. Construction of a 10,000-Membered Benzopyran Library by Directed Split-and-Pool Chemistry Using NanoKans and Optical Encoding. J. Am. Chem. Soc. 122:9954-9967 (2000); Nicolaou, K.C. et al. Natural Product-like
Combinatorial Libraries Based on Privileged Structures Natural Product-like Combinatorial Libraries Based on Privileged Structures 3. The "Libraries from Libraries" Principle for Diversity Enhancement of Benzopyran Libraries. J. Am. Chem. Soc. 122:9968-9976 (2000)). Genetically engineered LN229 cells (a human malignant glioma cell line) which stably expresses the alkaline phosphatase reporter gene under the control of six copies of a hypoxia-responsive element was used to identify small-molecule inhibitors of the HIF- 1/HRE pathway. The engineered LN229 cell line (LN229-HRE-AP) is disclosed in U.S. Provisional Patent Application No. 60/235,283, which is incorporated by reference in its entirety herein. Cells were seeded at 40,000 cells per well in 96-well plates. Compounds of interest (at a concentration range, for example 0.1-100 μM) were added and the plates were incubated under hypoxic conditions (1 % O2, 5% CO2 and 94% N2) at 37°C for 24 h. Cells were then washed with phosphate- buffered saline and incubated with the alkaline phosphatase substrate p-nitrophenyl phosphate at 370C for 30 min. The reaction was terminated by adding 3 N NaOH and the plates were read for absorbance at 405 nm. The anti-HIF- 1/HRE activity of each compound was quantified as the decrease in percentage of alkaline phosphatase (AP) activity compared to the untreated control cells.
Compounds I-A, I-B, and H-A were analyzed using the above assay at a concentration of 10 μM. The values in Table 1 illustrate the remaining percent AP activity measured for each compound. Table 1.
Figure imgf000026_0001
Example 2: Luciferase Assay
LN-229-HRE-luciferase glioblastoma cells containing a stably integrated reporter construct were prepared. The reporter construct was made of six copies of the HIF responsive element derived from the VEGF gene cloned in front of a luciferase gene as described in Post, D.E. and Van Meir, E.G. Gene Then 8(23): 1801- 1807 (2001), which is hereby incorporated by reference in its entirety. The cells were split into 48-well plates with 30,000 cells per well. The cells were then treated for one hour with either Compound I-A, Compound I-B, or Compound H-A, along with DMSO (Sigma; St. Louis, MO), DMEM media with 10% FBS (Media Tech;
Manassas, VA), Trypsin EDTA (Media Tech), and Penicillin/Streptomycin with Fungizone (Gibco, Invitrogen; Carlsbad, CA). The plates were then transferred to a hypoxia incubator (1% hypoxia). After 24 hours, the plates were removed from the incubator and the media was aspirated from the wells. Passive lysis buffer (50μL; IX) was added to each well. The plates were shaken for 10 minutes, and 20μL of protein and 25 μL of luciferase buffer were added. Measurement of luciferase activity was performed according to the protocol of the Luciferase Assay System (Promega; Madison, Wisconsin).
Compound I-A was analyzed using the above assay at concentrations of 2 μM, 10 μM, 20 μM, 50 μM, and 100 μM. Compound H-A was analyzed using the above assay at concentrations of 1 μM, 2 μM 5 μM, 10 μM, 20 μM, 25 μM, 50 μM, and 100 μM. The values in Tables 2A and 3A illustrate the luciferase activity measured in relative light units for each compound in separate trials. The data is also expressed as percentage of control (see Tables 2B and 3B) due to the highly sensitive nature of the luciferase assay. Compound H-A Table 2A. Luciferase Activity for Compounds I-A and H-A
Figure imgf000027_0001
Graphical representation in Figure 1.
Table 2B. Luciferase Activity for Compounds I-A and H-A
Figure imgf000027_0002
Table 3A. Luciferase Activity for Compound H-A
Figure imgf000027_0003
Graphical representation in Figures 2A and 2B.
Table 3B. Luciferase Activity for Compound H-A
Figure imgf000027_0004
A number of embodiments of the invention have been described. Nevertheless, it will be understood that various modifications may be made without departing from the spirit and scope of the invention. Accordingly, other embodiments are within the scope of the following claims.

Claims

WHAT IS CLAIMED IS:
1. A method of modulating HIF activity in a cell comprising contacting the cell with an effective amount of a compound according to formula I:
Figure imgf000028_0001
wherein:
R1, R2, and R5 are each independently H, OH, or OCi_6 alkyl;
R3 is C1-6 alkyl;
R4 is H or R4 comes together with R3 to form a fused dioxolane or dioxane ring; and wherein at least one of R1 or R2 is not H; and wherein if R1 is OH and R2 is H then R4 comes together with R3 to form a fused dioxolane or dioxane ring; or a pharmaceutically acceptable salt form thereof.
2. The method of claim 1 , wherein the HIF is HIF- 1.
3. The method of claim 1, wherein the cell is a non-cancerous cell.
4. The method of claim 1, wherein R1 is OH.
5. The method of claim 1 , wherein R2 is OH.
6. The method of claim 1 , wherein R3 is CH3.
7. The method of claim 1 , wherein R3 and R4 come together to form a fused dioxolane ring.
8. The method of claim 1, wherein the compound according to formula I is selected from:
Figure imgf000028_0002
or a pharmaceutically acceptable salt form thereof.
9. A method of modulating HIF activity in a cell comprising contacting the cell with an effective amount of a compound according to formula II:
Figure imgf000029_0002
wherein:
R1, R2, R3, and R5 are each independently H, OH, or OCi_6 alkyl;
R4 is H or Ci_6 alkyl; and
R6 is H or OH; or a pharmaceutically acceptable salt form thereof.
10. The method of claim 9, wherein the HIF is HIF-I .
11. The method of claim 9, wherein the cell is a non-cancerous cell.
12. The method of claim 9, wherein R1 and R2 are H.
13. The method of claim 9, wherein R6 is OH.
14. The method of claim 9, wherein R3 and R5 are independently an OCi_β alkyl, and R4 is a Ci_6 alkyl.
15. The method of claim 9, wherein R3 and R5 are OCH3, and R4 is CH3.
16. The method of claim 9, wherein the compound according to formula II is:
Figure imgf000030_0001
or a pharmaceutically acceptable salt form thereof.
17. A method of treating cancer in a subject, the method comprising administering to the subject a therapeutically effective amount of a compound according to formula I:
Figure imgf000030_0002
wherein:
R1, R2, and R5 are each independently H, OH, or OCi_6 alkyl;
R3 is Ci_6 alkyl;
R4 is H or R4 comes together with R3 to form a fused dioxolane or dioxane ring; and wherein at least one of R1 or R2 is not H; and wherein if R1 is OH and R2 is H then R4 comes together with R3 to form a fused dioxolane or dioxane ring; or a pharmaceutically acceptable salt form thereof.
18. A method of treating cancer in a subject, the method comprising administering to the subject a therapeutically effective amount of a compound according to formula II:
Figure imgf000031_0001
wherein:
R1, R2, R3, and R5 are each independently H, OH, or OCi_6 alkyl;
R4 is H or C i_6 alkyl; and
R6 is H or OH; or a pharmaceutically acceptable salt form thereof.
19. The method of claim 17 or 18, wherein the cancer is selected from: bladder cancer, brain cancer, breast cancer, colorectal cancer, cervical cancer, gastrointestinal cancer, genitourinary cancer, head and neck cancer, lung cancer, ovarian cancer, pancreatic cancer, prostate cancer, renal cancer, skin cancer, and testicular cancer.
20. A method of inhibiting angiogenesis in a subject, the method comprising administering to the subject an effective amount of a compound according to formula I:
Figure imgf000031_0002
wherein:
R1, R2, and R5 are each independently H, OH, or OCi_6 alkyl;
.3 •
R' is C1-6 alkyl;
R4 is H or R4 comes together with R3 to form a fused dioxolane or dioxane ring; and wherein at least one of R1 or R2 is not H; and wherein if R1 is OH and R2 is H then R4 comes together with R3 to form a fused dioxolane or dioxane ring; or a pharmaceutically acceptable salt form thereof.
21. The method of claim 20, wherein R1 is OH.
22. The method of claim 20, wherein R2 is OH.
23. The method of claim 20, wherein R3 is CH3.
24. The method of claim 20, wherein R3 and R4 come together to form a fused dioxolane ring.
25. The method of claim 20, wherein the compound according to formula I is selected from:
Figure imgf000032_0001
26. A method of inhibiting angiogenesis in a subject, the method comprising administering to the subject an effective amount of a compound according to formula II:
Figure imgf000032_0002
wherein:
R1, R2, R3, and R5 are each independently H, OH, or OCi_6 alkyl;
R ,4 i •s H or Ci_6 alkyl; and R6 is H or OH; or a pharmaceutically acceptable salt form thereof.
27. The method of claim 26, wherein R1 and R2 are H.
28. The method of claim 26, wherein R6 is OH.
29. The method of claim 26, wherein R3 and R5 are independently an OCi_6 alkyl, and R4 is a Ci_6 alkyl.
30. The method of claim 26, wherein R3 and R5 are OCH3, and R4 is CH3.
31. The method of claim 26, wherein the compound according to formula II is:
Figure imgf000033_0001
or a pharmaceutically acceptable salt form thereof.
32. The method of any one of claims 20 or 26, wherein the angiogenesis is associated with non-cancerous pathologies.
33. A method of treating a non-cancerous angiogenic disease in a subject, the method comprising administering to the subject a therapeutically effective amount of a compound according to formula I:
Figure imgf000033_0002
wherein:
R1, R2, and R5 are each independently H, OH, or OCi_6 alkyl;
R3 is C1-6 alkyl;
R4 is H or R4 comes together with R3 to form a fused dioxolane or dioxane ring; and wherein at least one of R1 or R2 is not H; and wherein if R1 is OH and R2 is H then R4 comes together with R3 to form a fused dioxolane or dioxane ring; or a pharmaceutically acceptable salt form thereof.
34. A method of treating a non-cancerous angiogenic disease in a subject, the method comprising administering to the subject a therapeutically effective amount of a compound according to formula II:
Figure imgf000034_0001
wherein:
R1, R2, R3, and R5 are each independently H, OH, or OCi_6 alkyl;
R4 is H or C 1-6 alkyl; and
R6 is H or OH; or a pharmaceutically acceptable salt form thereof.
35. The method of any one of claims 33 or 34, wherein the non-cancerous disease is selected from: atherosclerotic plaque growth and hemorrhage; chronic cystitis; Crohn's disease; diabetic retinopathy; dystrophic epidermolysis bullosa; infantile hemangiomas; intraperitoneal bleeding in endometriosis; macular degeneration; prostate growth in benign prostatic hypertrophy; psoriasis; rheumatoid arthritis; verruca vulgaris; surgical adhesions; keloids; non-cancerous lesions; aneurysms and vascular malformations in the brain; varicose veins; hemorrhoids; and rosacea.
36. The method of claim 35, wherein the non-cancerous disease is macular degeneration.
37. A method of treating macular degeneration in a subject, the method comprising administering to the subject a therapeutically effective amount of a compound according to formula I:
Figure imgf000035_0001
wherein:
R1, R2, and R5 are each independently H, OH, or OCi_6 alkyl;
R3 is Ci_6 alkyl;
R4 is H or R4 comes together with R3 to form a fused dioxolane or dioxane ring; and wherein at least one of R1 or R2 is not H; and wherein if R1 is OH and R2 is H then R4 comes together with R3 to form a fused dioxolane or dioxane ring; or a pharmaceutically acceptable salt form thereof.
38. A method of treating macular degeneration in a subject, the method comprising administering to the subject a therapeutically effective amount of a compound according to formula II:
Figure imgf000035_0002
wherein:
R1, R2, R3, and R5 are each independently H, OH, or OCi_6 alkyl;
R4 is H or Ci_6 alkyl; and
R6 is H or OH; or a pharmaceutically acceptable salt form thereof.
39. A method of treating a hypoxia-related pathology in a subject, the method comprising administering to the subject a therapeutically effective amount of a compound according to formula I:
Figure imgf000036_0001
wherein:
R1, R2, and R5 are each independently H, OH, or OCi_6 alkyl;
R3 is Ci_6 alkyl;
R4 is H or R4 comes together with R3 to form a fused dioxolane or dioxane ring; and wherein at least one of R1 or R2 is not H; and wherein if R1 is OH and R2 is H then R4 comes together with R3 to form a fused dioxolane or dioxane ring; or a pharmaceutically acceptable salt form thereof.
40. A method of treating a hypoxia-related pathology in a subject, the method comprising administering to the subject a therapeutically effective amount of a compound according to formula II:
Figure imgf000036_0002
wherein:
R1, R2, R3, and R5 are each independently H, OH, or OCi_6 alkyl;
R4 is H or Ci_6 alkyl; and
R6 is H or OH; or a pharmaceutically acceptable salt form thereof.
41. A method of modulating transcription and/or translation of a nucleic acid sequence in a cell comprising contacting the cell with an effective amount of a compound according to formula I:
Figure imgf000037_0001
wherein:
R1, R2, and R5 are each independently H, OH, or OCi_6 alkyl;
R3 is Ci_6 alkyl;
R4 is H or R4 comes together with R3 to form a fused dioxolane or dioxane ring; and wherein at least one of R1 or R2 is not H; and wherein if R1 is OH and R2 is H then R4 comes together with R3 to form a fused dioxolane or dioxane ring; or a pharmaceutically acceptable salt form thereof.
42. A method of modulating transcription and/or translation of a nucleic acid sequence in a cell comprising contacting the cell with an effective amount of a compound according to formula II:
Figure imgf000037_0002
wherein:
R1, R2, R3, and R5 are each independently H, OH, or OCi_6 alkyl;
R4 is H or Ci_6 alkyl; and
R6 is H or OH; or a pharmaceutically acceptable salt form thereof.
43. The method of claim 41 or 42, wherein the cell is a cancer cell.
44. The method of claim 41 or 42, wherein the nucleic acid sequence encodes for VEGF, erythropoietin, a glucose transporter, a glycolytic enzyme, or tyrosine hydroxylase.
45. A method of modulating a basic-helix-loop-helix transcription factor in a cell, the method comprising administering to the cell an effective amount of a compound according to formula I:
Figure imgf000038_0001
wherein:
R1, R2, and R5 are each independently H, OH, or OCi_6 alkyl;
R3 is C1-6 alkyl;
R4 is H or R4 comes together with R3 to form a fused dioxolane or dioxane ring; and wherein at least one of R1 or R2 is not H; and wherein if R1 is OH and R2 is H then R4 comes together with R3 to form a fused dioxolane or dioxane ring; or a pharmaceutically acceptable salt form thereof.
46. A method of modulating a basic-helix-loop-helix transcription factor in a cell, the method comprising administering to the cell an effective amount of a compound according to formula II:
Figure imgf000038_0002
wherein:
R1, R2, R3, and R5 are each independently H, OH, or OCi_6 alkyl;
R4 is H or Ci_6 alkyl; and
R6 is H or OH; or a pharmaceutically acceptable salt form thereof.
47. A method of modulating mRNA translation in a cell comprising contacting the cell with an effective amount of a compound according to formula I:
Figure imgf000039_0001
wherein:
R1, R2, and R5 are each independently H, OH, or OCi_6 alkyl;
R3 is C1-6 alkyl;
R4 is H or R4 comes together with R3 to form a fused dioxolane or dioxane ring; and wherein at least one of R1 or R2 is not H; and wherein if R1 is OH and R2 is H then R4 comes together with R3 to form a fused dioxolane or dioxane ring; or a pharmaceutically acceptable salt form thereof.
48. A method of modulating mRNA translation in a cell comprising contacting the cell with an effective amount of a compound according to formula II:
Figure imgf000039_0002
wherein:
R1, R2, R3, and R5 are each independently H, OH, or OCi_6 alkyl;
R4 is H or Ci_6 alkyl; and
R6 is H or OH; or a pharmaceutically acceptable salt form thereof.
49. A method of treating excessive vascularization in a subject, the method comprising administering to the subject a therapeutically effective amount of a compound according to formula I:
Figure imgf000040_0001
wherein:
R1, R2, and R5 are each independently H, OH, or OCi_6 alkyl;
R3 is Ci_6 alkyl;
R4 is H or R4 comes together with R3 to form a fused dioxolane or dioxane ring; and wherein at least one of R1 or R2 is not H; and wherein if R1 is OH and R2 is H then R4 comes together with R3 to form a fused dioxolane or dioxane ring; or a pharmaceutically acceptable salt form thereof.
50. A method of treating excessive vascularization in a subject, the method comprising administering to the subject a therapeutically effective amount of a compound according to formula II:
Figure imgf000040_0002
wherein:
R1, R2, R3, and R5 are each independently H, OH, or OCi_6 alkyl;
R4 is H or Ci_6 alkyl; and
R6 is H or OH; or a pharmaceutically acceptable salt form thereof.
51. The method of any one of claims 49 or 50, wherein the excessive vascularization is associated with non-cancerous pathologies.
52. A compound of the following formula:
Figure imgf000041_0001
or a pharmaceutically acceptable salt form thereof.
53. A compound of the following formula:
Figure imgf000041_0002
or a pharmaceutically acceptable salt form thereof.
54. A compound of the following formula:
Figure imgf000041_0003
or a pharmaceutically acceptable salt form thereof.
55. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound according to formula I:
Figure imgf000041_0004
wherein:
R1, R2, and R5 are each independently H, OH, or 0Ci_6 alkyl;
.3 •
R' is C1-6 alkyl; R4 is H or R4 comes together with R3 to form a fused dioxolane or dioxane ring; and wherein at least one of R1 or R2 is not H; and wherein if R1 is OH and R2 is H then R4 comes together with R3 to form a fused dioxolane or dioxane ring; or a pharmaceutically acceptable salt form thereof.
56. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound according to formula II:
Figure imgf000042_0001
wherein:
R1, R2, R3, and R5 are each independently H, OH, or OCi_6 alkyl;
R4 is H or Ci_6 alkyl; and
R6 is H or OH; or a pharmaceutically acceptable salt form thereof.
57. The pharmaceutical composition of any one or claims 52 or 56, wherein the composition is administered to the eye.
58. The pharmaceutical composition of claim 57, wherein the pharmaceutical composition is selected from ophthalmic drops, creams, ointments, installations, mucosal inserts, saturated contact lenses, diffusion release implants, and injectable solutions and suspensions.
PCT/US2009/050137 2008-07-11 2009-07-09 Small-molecule inhibitors of hif and angiogenesis WO2010006189A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US7994708P 2008-07-11 2008-07-11
US61/079,947 2008-07-11

Publications (2)

Publication Number Publication Date
WO2010006189A2 true WO2010006189A2 (en) 2010-01-14
WO2010006189A3 WO2010006189A3 (en) 2010-04-29

Family

ID=41507743

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/050137 WO2010006189A2 (en) 2008-07-11 2009-07-09 Small-molecule inhibitors of hif and angiogenesis

Country Status (1)

Country Link
WO (1) WO2010006189A2 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7981420B2 (en) 2000-12-22 2011-07-19 Max-Planck-Gesellschaft Zur Foederung Der Wissenschaften E.V. Therapeutic use of antibodies directed against repulsive guidance molecule (RGM)
US8906864B2 (en) 2005-09-30 2014-12-09 AbbVie Deutschland GmbH & Co. KG Binding domains of proteins of the repulsive guidance molecule (RGM) protein family and functional fragments thereof, and their use
US8962803B2 (en) 2008-02-29 2015-02-24 AbbVie Deutschland GmbH & Co. KG Antibodies against the RGM A protein and uses thereof
US9062072B2 (en) 2010-04-20 2015-06-23 Emory University Inhibitors of HIF and angiogenesis
US9102722B2 (en) 2012-01-27 2015-08-11 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of diseases associated with neurite degeneration
US9175075B2 (en) 2009-12-08 2015-11-03 AbbVie Deutschland GmbH & Co. KG Methods of treating retinal nerve fiber layer degeneration with monoclonal antibodies against a retinal guidance molecule (RGM) protein
US9381260B2 (en) 2011-12-27 2016-07-05 Emory University Hypoxia inducible factor-1 pathway inhibitors and uses as anticancer and imaging agents
WO2016164412A1 (en) * 2015-04-07 2016-10-13 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Hypoxia-inducible factor 1 (hif-1) inhibitors
WO2020205801A1 (en) * 2019-03-29 2020-10-08 Judith Boston Treating ophthalmic disease using hypoxia-inducible factor inhibitors

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002020008A1 (en) * 2000-09-06 2002-03-14 The Scripps Research Institute Inhibitors of nadh:ubiquinone oxidoreductase

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002020008A1 (en) * 2000-09-06 2002-03-14 The Scripps Research Institute Inhibitors of nadh:ubiquinone oxidoreductase

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
K.C.NICOLAOU ET AL: 'Natural Product-like Combinatorial Libraries Based on Privileged Structures. 1. General Principles and Solid-Phase Synthesis of Benzopyrans' JOURNAL OF THE AMERICAN CHEMICAL SOCIETY vol. 122, 30 September 2000, pages 9939 - 9953 *
KARSTEN KROHN ET AL: 'Isolation and synthesis of chalcones with different degrees of saturation' PHYTOCHEMISTRY vol. 61, 20 November 2002, pages 931 - 936 *

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7981420B2 (en) 2000-12-22 2011-07-19 Max-Planck-Gesellschaft Zur Foederung Der Wissenschaften E.V. Therapeutic use of antibodies directed against repulsive guidance molecule (RGM)
US8906864B2 (en) 2005-09-30 2014-12-09 AbbVie Deutschland GmbH & Co. KG Binding domains of proteins of the repulsive guidance molecule (RGM) protein family and functional fragments thereof, and their use
US8962803B2 (en) 2008-02-29 2015-02-24 AbbVie Deutschland GmbH & Co. KG Antibodies against the RGM A protein and uses thereof
US9605069B2 (en) 2008-02-29 2017-03-28 AbbVie Deutschland GmbH & Co. KG Antibodies against the RGM a protein and uses thereof
US9175075B2 (en) 2009-12-08 2015-11-03 AbbVie Deutschland GmbH & Co. KG Methods of treating retinal nerve fiber layer degeneration with monoclonal antibodies against a retinal guidance molecule (RGM) protein
US9062072B2 (en) 2010-04-20 2015-06-23 Emory University Inhibitors of HIF and angiogenesis
US9381260B2 (en) 2011-12-27 2016-07-05 Emory University Hypoxia inducible factor-1 pathway inhibitors and uses as anticancer and imaging agents
US9365643B2 (en) 2012-01-27 2016-06-14 AbbVie Deutschland GmbH & Co. KG Antibodies that bind to repulsive guidance molecule A (RGMA)
US9102722B2 (en) 2012-01-27 2015-08-11 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of diseases associated with neurite degeneration
US10106602B2 (en) 2012-01-27 2018-10-23 AbbVie Deutschland GmbH & Co. KG Isolated monoclonal anti-repulsive guidance molecule A antibodies and uses thereof
WO2016164412A1 (en) * 2015-04-07 2016-10-13 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Hypoxia-inducible factor 1 (hif-1) inhibitors
US10246463B2 (en) 2015-04-07 2019-04-02 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Hypoxia-inducible factor 1 (HIF-1) inhibitors
WO2020205801A1 (en) * 2019-03-29 2020-10-08 Judith Boston Treating ophthalmic disease using hypoxia-inducible factor inhibitors

Also Published As

Publication number Publication date
WO2010006189A3 (en) 2010-04-29

Similar Documents

Publication Publication Date Title
WO2010006189A2 (en) Small-molecule inhibitors of hif and angiogenesis
US9062072B2 (en) Inhibitors of HIF and angiogenesis
RU2449996C2 (en) COMPOUNDS AND METHODS FOR INHIBITING Bcl PROTEIN INTERACTION WITH BINDING PARTNERS
WO2010006184A2 (en) Small-molecule inhibitors of hif and angiogenesis
US20090318526A1 (en) Use of cannabinoid receptor agonists as hypothermia inducing drugs for the treatment of ischemia
US20100029739A1 (en) Use of a combination of hypothermia inducing drugs
TW201105677A (en) Enantiomers of spiro-oxindole compounds and their uses as therapeutic agents
WO2008040360A2 (en) Use of hypothermia inducing drugs to treat ischemia
ES2655455T3 (en) Compositions for the treatment of hypertension and / or fibrosis
PT100602A (en) USE OF TETRA-HYDROBENZAZEPIN DERIVATIVES, TETRA-HYDROBENZAZEPINES DERIVATIVES, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS THAT CONTAIN THEM
US20030176496A1 (en) N-substituted Dithiocarbamates for the Treatment of Biological Disorders
CN105616400A (en) Use of arctigenin carbamate derivatives in preparation of drug for treating Alzheimer disease
KR20190115011A (en) Sulfinylpyridine and its use in the treatment of cancer
CN102245174A (en) Synergistic effects between sphingosine-1-phosphate receptor antagonists and antimicrotubule agents
KR102011105B1 (en) pharmaceutical composition for prevention or treatment of pancreatic cancer comprising a gossypol and a phenformin
JP2021523934A (en) Aminopyrazine and related compounds useful as mitochondrial deconjugation agents
CN107698470A (en) Substituted diaryl sulfonamide and application thereof
CN101654427A (en) Anticoagulant compound, composition and application thereof
CN105163732B (en) Methods for treating ophthalmic diseases and disorders
US20100137421A1 (en) Small molecule therapeutics, synthesis of analogues and derivatives and methods of use
TW200829261A (en) Method for controlling angiogenesis in animals
EA029157B1 (en) Baclofen and acamprosate based therapy of macular degeneration
JP2007516289A (en) 5-HT2 receptor modulators for the treatment of cardiovascular and muscular diseases, their pharmaceutical compositions and their use
JP7170341B2 (en) New low-molecular-weight compounds
CN102775596B (en) D51-7059 and its production and use

Legal Events

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

Ref document number: 09795203

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09795203

Country of ref document: EP

Kind code of ref document: A2