WO2018068131A1 - Substituted hydroxystilbenes and their therapeutic applications - Google Patents

Substituted hydroxystilbenes and their therapeutic applications Download PDF

Info

Publication number
WO2018068131A1
WO2018068131A1 PCT/CA2017/051182 CA2017051182W WO2018068131A1 WO 2018068131 A1 WO2018068131 A1 WO 2018068131A1 CA 2017051182 W CA2017051182 W CA 2017051182W WO 2018068131 A1 WO2018068131 A1 WO 2018068131A1
Authority
WO
WIPO (PCT)
Prior art keywords
disease
compound
group
subject
bromo
Prior art date
Application number
PCT/CA2017/051182
Other languages
French (fr)
Inventor
Liren Tang
Junling Zhang
Tao Guo
Michael Patrick Allen Lyle
Original Assignee
Resolvex Pharmaceuticals Inc.
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 Resolvex Pharmaceuticals Inc. filed Critical Resolvex Pharmaceuticals Inc.
Priority to CN201780062107.0A priority Critical patent/CN109890784B/en
Priority to EP17860520.0A priority patent/EP3526186B1/en
Priority to ES17860520T priority patent/ES2924065T3/en
Priority to JP2019520644A priority patent/JP6936319B2/en
Priority to CA3038575A priority patent/CA3038575C/en
Priority to AU2017344103A priority patent/AU2017344103B2/en
Priority to KR1020197009817A priority patent/KR102419043B1/en
Publication of WO2018068131A1 publication Critical patent/WO2018068131A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C39/00Compounds having at least one hydroxy or O-metal group bound to a carbon atom of a six-membered aromatic ring
    • C07C39/205Compounds having at least one hydroxy or O-metal group bound to a carbon atom of a six-membered aromatic ring polycyclic, containing only six-membered aromatic rings as cyclic parts with unsaturation outside the rings
    • C07C39/21Compounds having at least one hydroxy or O-metal group bound to a carbon atom of a six-membered aromatic ring polycyclic, containing only six-membered aromatic rings as cyclic parts with unsaturation outside the rings with at least one hydroxy group on a non-condensed ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • A61K31/047Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates having two or more hydroxy groups, e.g. sorbitol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • A61K31/05Phenols
    • A61K31/055Phenols the aromatic ring being substituted by halogen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/275Nitriles; Isonitriles
    • A61K31/277Nitriles; Isonitriles having a ring, e.g. verapamil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C253/00Preparation of carboxylic acid nitriles
    • C07C253/30Preparation of carboxylic acid nitriles by reactions not involving the formation of cyano groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C255/00Carboxylic acid nitriles
    • C07C255/49Carboxylic acid nitriles having cyano groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton
    • C07C255/53Carboxylic acid nitriles having cyano groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton containing cyano groups and hydroxy groups bound to the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C37/00Preparation of compounds having hydroxy or O-metal groups bound to a carbon atom of a six-membered aromatic ring
    • C07C37/001Preparation of compounds having hydroxy or O-metal groups bound to a carbon atom of a six-membered aromatic ring by modification in a side chain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C39/00Compounds having at least one hydroxy or O-metal group bound to a carbon atom of a six-membered aromatic ring
    • C07C39/24Halogenated derivatives
    • C07C39/373Halogenated derivatives with all hydroxy groups on non-condensed rings and with unsaturation outside the aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C39/00Compounds having at least one hydroxy or O-metal group bound to a carbon atom of a six-membered aromatic ring
    • C07C39/24Halogenated derivatives
    • C07C39/42Halogenated derivatives containing six-membered aromatic rings and other rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/02Systems containing only non-condensed rings with a three-membered ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/06Systems containing only non-condensed rings with a five-membered ring
    • C07C2601/08Systems containing only non-condensed rings with a five-membered ring the ring being saturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/12Systems containing only non-condensed rings with a six-membered ring
    • C07C2601/14The ring being saturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/12Systems containing only non-condensed rings with a six-membered ring
    • C07C2601/16Systems containing only non-condensed rings with a six-membered ring the ring being unsaturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/18Systems containing only non-condensed rings with a ring being at least seven-membered

Definitions

  • the present invention pertains to substituted hydroxystilbenes and their therapeutic applications.
  • the inflammatory response is mediated by cytokines, and cytokine activity and expression in turn are regulated by kinases.
  • Aberrant kinase and cytokine activity play pivotal roles in chronic and acute inflammatory diseases.
  • Aberrant kinase and cytokine activity also underly autoimmune diseases and different types of cancer.
  • Kinase inhibitors are an established class of anticancer drugs (Chen, J Nat Prod, 2012, 75 (12):2269-2269) and anti-inflammation drugs (Wang et al, J Immunol, 2013,
  • Janus Kinase 2 (JAK2) and Janus Kinase 3 (JAK3) are tyrosine kinases that regulate transcription of various target genes involved in growth and proliferation (Reiter et al, Cancer Res, 2005, 65(7):2662-2667; Takemoto et al, Proc Natl Acad Sci USA, 1997, 94(25): 13897-13902). JAK2 and JAK3 are also involved in mediating the signaling of many inflammatory cytokines involved in inflammation (Wang et al, J Immunol, 2013, 191(3): 1164-1174).
  • JAK2 and JAK 3 inhibitors are therefore useful in treating cancers, and autoimmune and inflammatory diseases (Fridman et al, J Invest Dermatol, 2011, 131(9): 1838-1844).
  • p38a p38 mitogen-activated protein kinase alpha
  • MAP Mitogen-activated protein
  • Glycogen synthase kinase 3 beta ( ⁇ 8 ⁇ 3 ⁇ ) is a serine-threonine kinase that regulates p53 function in proliferating cells such as cancer cells (WO2006/006939) and its inhibition down regulates inflammatory cytokines (Li et al, Cell Physiol Biochem, 2013, 32(6): 1720- 1728).
  • Lymphocyte-specific protein tyrosine kinase is a Src tyrosine kinase involved in T cell activation and proliferation (Hanke et al, Inflamm Res, 1995, 44:357; Bolen et al, Ann Rev Immunol, 1997, 15:371). LCK inhibition has been successful in treatment of inflammatory diseases (Brisslert et al, Biochem Biophys Acta, 2014, 1842(11):2049-2059).
  • Cytokines play important roles in initiating and regulating immune responses, and therefore their inhibition is a well-established approach to controlling autoimmune and inflammatory diseases (Kopf et al, Nature Reviews Drug Discovery, September 2010, 9:703- 718).
  • exemplary cytokine targets include Interleukin- ⁇ (IL- ⁇ ) (Xu et al, Clin Exp Pharmacol Physiol, 2015, 42(10): 1075-83), Interleukin-2 (IL-2) (Roediger et al., J Allergy Clin Immunol, 2015, 136(6): 1653-63), Interleukin-6 (IL-6) (Scheller et al, Biochem
  • Interleukin-8 (IL-8) (Aghazarian et al., Urology, 2015, 86(l):52-56), Interferon
  • IFN- ⁇ gamma
  • TNF-a Tumor necrosis factor alpha
  • MIP-la Macrophage Inflammatory Protein la
  • PDE4 inhibition is known to improve therapeutic treatment of a number of inflammatory, respiratory and allergic diseases and conditions (US6649633B2; Keren et al, J Dermatol Sci, January 2015, 77(l):74-76).
  • FIG. 1 describes the constituents of compounds according to embodiments of the invention.
  • FIG. 2 shows the structures of compounds according to embodiments of the invention.
  • FIG. 3 shows the synthetic process for making compounds according to embodiments of the invention.
  • FIG. 4 shows the efficacy of a compound according to an embodiment of the invention in dextran sodium sulphate (DSS)-induced model of inflammatory bowel disease (IBD)/colitis in mice.
  • DSS dextran sodium sulphate
  • IBD inflammatory bowel disease
  • autoimmune disease refers to the physiological condition in mammals that is typically characterized by the immune system's reaction to inappropriate targets such as normal tissues.
  • the term "autoimmune disease” includes Addison's disease, ankylosing spondylitis, celiac disease, Hashimoto's thyroiditis, multiple sclerosis, myasthenia gravis, pemphigus, rheumatoid arthritis, Sjogren syndrome, systemic lupus erythematosus, Type 1 diabetes, IBD, psoriasis, vitiligo, alopecia areata and vasculitis.
  • cancer refers to the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • cancer includes cancer of any origin, including benign and malignant cancers, metastatic and non-metastatic cancers, and primary and secondary cancers.
  • cancer includes reference to cancer cells.
  • cancers include, but are not limited to, cancers of the bladder, bone, brain/CNS, breast, cervix, colon, duodenum, esophagus, eye, gall bladder, heart, kidney, larynx, liver, lung, mouth, ovary, pancreas, pharynx, prostate, rectum, stomach, testis, uterus, as well as AIDS-related cancers, Hodgkin's disease, non-Hodgkin's lymphoma, multiple myeloma, melanoma, leukemia (including lymphocytic leukemia, hairy cell leukemia, and acute myelogenous leukemia), choriocarcinoma, rhabdomyosarcoma, and neuroblastoma.
  • AIDS-related cancers Hodgkin's disease, non-Hodgkin's lymphoma, multiple myeloma, melanoma
  • leukemia including lymphocytic leukemia
  • an "effective amount” as used herein refers to the amount of active agent sufficient to elicit a desired biological response or, equivalently, to inhibit an undesired biological response.
  • An amount of a particular active agent that is effective may vary depending on such factors as the desired biological response, severity of the disease, the activity of the active agent to be delivered, the route of administration, the rate of excretion of the active agent being employed, the duration of the treatment, other drugs, compounds or materials used in combination with the particular active agent employed, the subject's age, sex, weight, condition, general health and prior medical history of the subject, and like factors well known in the medical arts.
  • an "effective amount” will be that amount of the active agent that is the lowest dose effective to produce the desired biological response.
  • an “effective amount” will generally depend upon the factors described above. Generally, an “effective amount” will range from about 1 to about 400 mg per kilogram of body weight per day, more preferably from about 10 to about 50 mg per kg per day. If desired, daily dosage in an “effective amount” may be administered as one dose, or two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms.
  • inflammatory disease refers to the physiological condition in mammals that is typically characterized by the immune system's excessive reaction to innocuous targets or reaction to inappropriate targets.
  • inflammatory disease includes Alzheimer's disease, amyotrophic lateral sclerosis, asthma, atherosclerosis, atopic dermatitis, autoimmune diseases, eczema, inflammatory bowel disease, interstitial cystitis, irritable bowel syndrome, osteoarthritis, Parkinson's disease, periodontal disease, psoriasis and sun burn.
  • a "package insert" as used herein refers to instructions customarily included in commercial packages of medicaments that contain information about the indications, usage, dosage, administration, contraindications, other medicaments to be combined with the packaged product, and/or warnings concerning the use of such medicaments, and the like.
  • pharmaceutically acceptable carrier refers to one or more excipients, stabilizers, fillers, binders, humectants, disintegrating agents, solution retarding agents, absorption accelerators, wetting agents, absorbents, lubricants, coloring agents, diluents, emulsifiers, preservatives, solubilizing agents, suspending agents and the like that are suitable for use with the subject being exposed thereto at the dosages and concentrations employed without undue adverse side effects (such as toxicity, irritation, and allergic response) commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable carrier refers to one or more excipients, stabilizers, fillers, binders, humectants, disintegrating agents, solution retarding agents, absorption accelerators, wetting agents, absorbents, lubricants, coloring agents, diluents, emulsifiers, preservatives, solubilizing agents, suspending agents and the like that are suitable for use with the subject being exposed thereto
  • Examples of pharmaceutically acceptable carriers include water, citrate or phosphate buffers, starches, lactose, sucrose, glucose, mannitol, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, glycerol, agar, calcium carbonate, alginic acid, sodium carbonate, paraffin, quaternary ammonium compounds, cetyl alcohol, glycerol monostearate, kaolin and bentonite clay, talc, calcium stearate, magnesium stearate, polyethylene glycols, sodium lauryl sulfate, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils, tetrahydrofuryl alcohol, fatty acid esters, thioxylated isostearyl alcohols, polyoxyethylene sorbitol and
  • salts refers to toxicologically compatible organic or inorganic salts of the active agent.
  • exemplary salts include, but are not limited, to sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucuronate, saccharate, formate, benzoate, glutamate, methanesulfonate
  • a pharmaceutically acceptable salt can have one or more charged atoms and/or one or more counter ion.
  • the desired pharmaceutically acceptable salt may be prepared by any suitable method available in the art, for example, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, methanesulfonic acid, phosphoric acid and the like, or with an organic acid, such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, a pyranosidyl acid, such as glucuronic acid or galacturonic acid, an alpha hydroxy acid, such as citric acid or tartaric acid, an amino acid, such as aspartic acid or glutamic acid, an aromatic acid, such as benzoic acid or cinnamic acid, a sulfonic acid, such as p-toluenesulfonic acid or ethanesulfonic acid, or
  • suitable salts include, but are not limited to, organic salts derived from amino acids, such as glycine and arginine, ammonia, primary, secondary, and tertiary amines, and cyclic amines, such as piperidine, morpholine and piperazine, and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.
  • proliferative disease refers to the physiological condition in mammals that is characterized by the excessive proliferation of cells and turnover of cellular matrix.
  • proliferative disease includes cancer, graft-versus-host disease, restenosis, hamartoma syndromes (e.g., tuberous sclerosis or Cowden Syndrome), encephalomyelitis, arthritis, scleroderma, pulmonary fibrosis, renal fibrosis, cystic fibrosis, pulmonary hypertension, hypertrophic cardiomyopathy, Parkinson-White syndrome and wet and dry macular degeneration.
  • subject refers to an individual to whom an active agent is to be delivered, e.g., for treatment purposes.
  • the term “subject” includes mammals, in particular humans, and other mammals including companion animals such as cats and dogs, livestock animals such as cows, pigs, horses, sheep and goats, zoo animals, and research animals, such as rodents.
  • the "subject” also includes in vitro cultured cells.
  • Desired clinical results can include, but are not limited to, reduction or alleviation of at least one symptom of a disease.
  • treatment can be diminishment of at least one symptom of disease, diminishment of extent of disease, stabilization of disease state, prevention of spread of disease, delay or slowing of disease progression, palliation of disease, diminishment of disease reoccurrence, remission of disease, prolonging survival with disease, or complete eradication of disease.
  • AKT1 RAC-alpha serine/threonine -protein kinase
  • G-CSF Granulocyte colony stimulating factor
  • GM-CSE Granulocyte macrophage colony stimulating factor
  • GSK3 Glycogen synthase kinase 3 beta
  • IBD Inflammatory bowel disease
  • IFN- ⁇ Interferon gamma
  • IL- ⁇ Interleukin-1 beta
  • IL-2 Interleukin-2
  • IL-8 Interleukin-8
  • JAK3 Janus Kinase 3
  • LCK Lymphocyte specific protein tyrosine kinase
  • MIP- ⁇ Macrophage inflammatory protein 1 alpha
  • PBMC Peripheral blood mononuclear cell
  • p38a p38 mitogen activated protein kinase alpha
  • ⁇ 38 ⁇ p38 mitogen activated protein kinase beta
  • PHA Phytohaemagglutinin
  • TNF-a Tumor necrosis factor alpha
  • R 1 is selected from H, halogen and CN
  • R 2 is selected from H, halogen and CN
  • R 3 is selected from H, halogen and CN
  • R 4 is H
  • R 5 is selected from H, halogen and CN
  • R 6 is alkyl or cycloalkyl
  • the compound may be one of exemplary Compounds 1 to 27 set out in FIG. 1.
  • FIG. 2 shows the structure of exemplary Compounds 1. 2. 7, 10, 12, 18, 20. 24 and 27.
  • the compound may have Formula I wherein R 1 is selected from H, F and CN; R 2 is H or CN; R 3 is selected from H, F and CN; R 4 is H; R 5 is H or F; and R 6 is selected from iso-propyl, cyclopropyl, cyclopentyl. cyclohexyl and cycloheptyl.
  • the compound may have Formula I wherein R 1 is H; R 2 is H or CN; R 3 is selected from H, F and CN; R 4 is H; R 5 is H; and R 6 is selected from iso-propyl, cyclopropyl, cyclopentyl, cyclohexyl and cycloheptyl.
  • the compound may have Formula I wherein R 1 is H; R 2 is H; R 3 is F; R 4 is H; R 5 is H; and R 6 is selected from iso-propyl, cyclopropyl and cyclopentyl.
  • the compound may be 3-hydroxy-4-isopropyl-4'-fluoro-(£)- stilbene.
  • the compound having Formula I may be provided as a pharmaceutically acceptable salt.
  • compositions comprising a compound having Formula I and a pharmaceutically acceptable carrier.
  • compositions comprising compounds of Formula I may be provided in unit dosage form.
  • the amounts of compounds of Formula I that can be combined with a pharmaceutically acceptable carrier to produce a single dosage form will generally be the amount that produces a therapeutic effect. In some embodiments, out of one hundred percent, this amount will range from about 0.1 percent to about 99 percent of active agent, from about 5 percent to about 70 percent of active agent, or from about 10 percent to about 30 percent of active agent.
  • aspects relate to methods of treating a condition by administration to a subject of an effective amount of a compound having Formula I. While it is possible for compounds of Formula I to be administered alone, it may be preferable to administer them in combination with one or more pharmaceutically acceptable carriers as a composition. In some
  • the methods comprise administering to the subject at least one compound having Formula I either alone in or in combination with at least one additional therapeutic agent.
  • compounds having Formula I inhibit cell proliferation, inhibit PDE4, inhibit a range of kinases, and inhibit a range of cytokines.
  • Suitable diseases may be treated with compounds having Formula I. Suitable diseases that may be treated include cancers, autoimmune diseases and inflammatory diseases.
  • the subject to be treated may be any subject diagnosed as having one of the indicated conditions.
  • the subject to be treated may be in need of treatment for one of the indicated conditions because of a diagnosis of the condition or because of an assessment of risk for developing the condition.
  • the subject may be diagnosed with the condition using diagnostic or clinical tests that are well known.
  • the administration route of compounds of Formula I, alone or in a composition, in terms of effect may be local or systemic (enteral or parenteral), and in terms of location may for example be buccal, epi cutaneous, epidural, intraartciular, intracardiac, intracavemous, intracerebral, intracerebroventricular, intradermal, intramuscular, intraosseous, intraperitoneal, intrathecal, intrauterine, intravaginal, intravenous, intravesical, intravitreal, nasal, oral, rectal, subcutaneous, sublingual, sublabial, transdermal, transmucosal, and the like.
  • oral administration may be in the form of capsules, cachets, pills, tablets, lozenges, pastes, powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles or as mouth washes and the like.
  • compositions in solid dosage forms for oral administration include capsules, tablets, pills, dragees, powders, granules and the like.
  • the solid dosage forms may be scored or prepared with coatings and shells, such as enteric coatings and other coatings.
  • compositions in liquid dosage forms for oral administration include emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • topical or transdermal administration may be in the form of powders, sprays, ointments, pastes, creams, lotions, gels, solutions, controlled-release patches and inhalants.
  • parenteral administration e.g. intravenous administration
  • compounds of Formula I are formulated into pharmaceutically acceptable dosage forms by conventional methods.
  • kits comprising compounds of Formula I, and a package insert comprising instructions for using compounds of Formula I to treat a condition in a subject.
  • Suitable conditions that may be treated include cancers, autoimmune diseases and inflammatory diseases.
  • FIG. 3 illustrates a synthetic method used to prepare compounds of Formula I according to an embodiment of the invention.
  • the compounds are synthesized in six steps from the commercially available starting material, 3-methoxybenzyl alcohol, as follows.
  • Step 1 3-Methoxybenzyl alcohol was brominated with one molar equivalent of N- bromosuccinimide in tetrahydrofuran (THF) at room temperature to afford 4-bromo-3- methoxybenzyl alcohol.
  • Step 2 4-bromo-3-methoxybenzyl alcohol was converted to the corresponding benzyl chloride with an excess of thionyl chloride in dimethylformamide (DMF) at room
  • Step 3 The benzyl chloride from Step 2 was converted to diethyl 4-bromo-3- methoxybenzylphosphonate with an excess of tri ethyl phosphite upon heating at 150 °C for 16 hours.
  • Step 4 The diethyl 4-bromo-3-methoxybenzylphosphonate from Step 3 was reacted with an excess of sodium hydride and a substituted benzaldehyde in THF to afford the substituted trans-4-bromo-3-methoxystilbene (Horner-Wadsworth-Emmons Reaction).
  • Step 5 The substituted trans-4-bromo-3-methoxystilbene from Step 4 was reacted with an alkyl Grignard reagent in the presence of a catalytic amount of N1CI2DPPE
  • Step 6 The substituted trans-4-alkyl-3-methoxystilbene from Step 5 was reacted with an excess of boron tribromide in dichloromethane (DCM) at room temperature to afford the substituted trans-4-alkyl-3-hydroxystilbene.
  • DCM dichloromethane
  • reaction mixture was quenched with water (5 mL), diluted with additional ether (60 mL) and the resultant mixture was washed first with a saturated aqueous solution of sodium thiosulfate and then with water. The organic layer was dried over anhydrous magnesium sulfate and concentrated in vacuo to afford the crude product which was purified by recrystallization from hexanes to afford the title compound as a light yellow crystalline solid (874 mg, 66%).
  • DMEM containing 10% FBS (A549 and MDA-MB-435).
  • l-5xl0 4 cells per well in 100 of the cell culture medium containing 0.5% FBS were seeded into a 96-well flat bottom plate.
  • the test compounds were added to the culture at different concentrations in triplicates.
  • the controls include triplicates of no compound treatment and cell culture medium without cells.
  • the cells were incubated at 5% C0 2 , 37 °C for 48 hours.
  • MTS reagent was added to each well and incubated at 37 °C for 4 hours.
  • the absorbance was read at 490 nm.
  • the percentage of inhibition was calculated using the formula: [1 -(experiment reading-background reading)/(negative control reading-background reading)] x 100.
  • the determined cell viability IC50 values are listed in Table 1.
  • Cells were cultured in RPMI-1640 containing 10% FBS (Jurkat) or DMEM containing 10% FBS (MDA-MB-435).
  • lxlO 4 cells were seeded into one well of a 96-well white ISOPLATETM with 0.5% FBS.
  • the cells were cultured at 5% C0 2 , 37 °C overnight.
  • the diluted test compounds were added into each well in a total volume of 100 of cell culture medium.
  • For each cell line were set up three wells containing cells and only diluted DMSO as a negative (vehicle) control, and three wells without cells but containing the same volume and type of cell culture medium as background control.
  • the cells were incubated at 5% C0 2 , 37 °C for 24 hours. On Day 3, 1 of [3 H ] -thymidine was added into each well and incubation at 5% C0 2 , 37 °C was continued for another 24 hours. On Day 4, 30 of 50% TCA was added into each well and was incubated at 4 °C for 4 hours. Plates were washed with ddH 2 0 four times and then air dried for 30 minutes followed by the addition of 100 ⁇ . of scintillation fluid into each well. The radioactivity was read using
  • kinases profiling was conducted using different recombinant kinase targets against the compounds at 5 uM ATP.
  • the kinases tested included AKTl (RAC-alpha serine/threonine- protein kinas 1), JAK2, JAK3, ⁇ 38 ⁇ , ⁇ 38 ⁇ (p38 mitogen-activated protein kinase beta), GSK3fi, SYK (Spleen tyrosine kinase), LCK, IKKa and ⁇ and CLK4.
  • the test compounds were tested at 5 ⁇ and 50 ⁇ in triplicates. The results are shown in Table 3.
  • Assays were performed using recombinant human PDE4B1 enzyme expressed in a baculoviral system.
  • the radiometric assay is a modification of the 2-step method of Thompson and Appleman (Biochemistry 10, 1971, 311-316). The reactions were performed at 1 ⁇ of cAMP.
  • the test articles were each tested at 0.2 ⁇ and 20 ⁇ in triplicates.
  • the percentage inhibition values for the reference inhibitor, rolipram were also determined and compared to historical assay values to ensure it was in an acceptable range. The results are shown in Table 4.
  • PBMC cells were used to test the effects of the compounds on the production multiple cytokines.
  • PBMC human PBMC
  • FBS fetal bovine serum
  • Cytokine production was induced with a combination of PMA and PHA 30 minutes post the compound treatments.
  • Cells were treated with various concentrations of the compounds for 24 hours at 37 °C.
  • the culture supernatants were collected from drug-treated or control samples.
  • Cytokines tested include: IL- la, IL-1 ⁇ , IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12, IL-13, IL-17A, IFN- ⁇ , TNF-a, G- CSF(Granulocyte -colony -stimulating factor), GM-CSF(Granulocyte-macrophage colony- stimulating factor), TGF- ⁇ 1 and ⁇ .
  • EXAMPLE 8 Effect on inflammatory bowel disease (IBD)/colitis in mice
  • DSS dextran sodium sulphate
  • IBD inflammatory bowel disease
  • Rolipram was used as a positive control in this experiment.
  • Colitis was induced in the mice by providing drinking water containing DSS (3.5%, M. Wt. 30,000-50,000) from Day 0 to Day 8.
  • the test compounds were administered as per their respective groups at specific dose levels starting from Day 0 through to Day 8.
  • Compound 2 was tested at 10 and 30 mg/kg and Rolipram was tested at 5 mg/kg.
  • Colitis was consistently induced in most of the mice, with onset of disease (stool consistency) from Day 2 and drastic reduction in body weight, diarrhea and severe rectal bleeding at Day 5-7 demonstrating the severity of disease.
  • Compound 2 was dissolved in vehicle (1.5% Cremophor EL) at 3 mg/mL and 1 mg/mL for the 30 and 10 mg/kg doses, respectively.
  • Rolipram was dissolved in 1% DMSO/99% PBS at 0.5 mg/mL for the 5 mg/kg dose. All doses were administered by oral gavage.
  • DAI disease activity index
  • EXAMPLE 9 Efficacy against the inflammatory skin conditions psoriasis and eczema
  • a topical cream containing compound 2 was prepared as follows: Compound 2 was dissolved in propylene glycol at a concentration of 11.0% w/w and this solution was then mixed with the appropriate amount of Glaxal base cream to afford a 1.0% w/w cream of compound 2.
  • 10 subjects with psoriasis and 5 subjects with eczema were treated with the cream twice daily for a period of 2-12 weeks.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Dermatology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

The present invention provides substituted hydroxystilbenes compounds of the general formula I that inhibit the activities of numerous of protein kinases involving the signaling of inflammatory cytokines, therefore, the compounds can be used for treating cancers, autoimmune diseases and inflammatory diseases.

Description

SUBSTITUTED HYDROXYSTILBENES AND THEIR THERAPEUTIC APPLICATIONS
Technical Field
[0001] The present invention pertains to substituted hydroxystilbenes and their therapeutic applications.
Background
[0002] The inflammatory response is mediated by cytokines, and cytokine activity and expression in turn are regulated by kinases. Aberrant kinase and cytokine activity play pivotal roles in chronic and acute inflammatory diseases. Aberrant kinase and cytokine activity also underly autoimmune diseases and different types of cancer.
[0003] Kinase inhibitors are an established class of anticancer drugs (Chen, J Nat Prod, 2012, 75 (12):2269-2269) and anti-inflammation drugs (Wang et al, J Immunol, 2013,
191(3): 1164-1174). For example, Janus Kinase 2 (JAK2) and Janus Kinase 3 (JAK3) are tyrosine kinases that regulate transcription of various target genes involved in growth and proliferation (Reiter et al, Cancer Res, 2005, 65(7):2662-2667; Takemoto et al, Proc Natl Acad Sci USA, 1997, 94(25): 13897-13902). JAK2 and JAK3 are also involved in mediating the signaling of many inflammatory cytokines involved in inflammation (Wang et al, J Immunol, 2013, 191(3): 1164-1174). JAK2 and JAK 3 inhibitors are therefore useful in treating cancers, and autoimmune and inflammatory diseases (Fridman et al, J Invest Dermatol, 2011, 131(9): 1838-1844). p38a (p38 mitogen-activated protein kinase alpha) is a MAP (Mitogen-activated protein) kinase that also mediates the signaling of many
inflammatory cytokines involved in inflammation. Inhibitors of the kinase activity of p38 a are therefore useful anti-inflammatory agents (Daniele et al, Cell Signal, 2015, 27(8): 1609- 1629). Glycogen synthase kinase 3 beta (ϋ8Κ3β) is a serine-threonine kinase that regulates p53 function in proliferating cells such as cancer cells (WO2006/006939) and its inhibition down regulates inflammatory cytokines (Li et al, Cell Physiol Biochem, 2013, 32(6): 1720- 1728). Lymphocyte-specific protein tyrosine kinase (LCK) is a Src tyrosine kinase involved in T cell activation and proliferation (Hanke et al, Inflamm Res, 1995, 44:357; Bolen et al, Ann Rev Immunol, 1997, 15:371). LCK inhibition has been successful in treatment of inflammatory diseases (Brisslert et al, Biochem Biophys Acta, 2014, 1842(11):2049-2059). Inhibition of the ΙκΒ kinase (IKK) β subunit of the IKK kinase enzyme complex has also been associated with anti-inflammatory effects (Novoselova et al, Mediators Inflamm, 2014, 2014:724838). CLK (CDC2-like kinase) plays important roles in gene splicing and is a potential therapeutic target for Alzheimer's disease (Jain et al, Curr Drug Targets, 2014, 15(5):539-550).
[0004] Cytokines play important roles in initiating and regulating immune responses, and therefore their inhibition is a well-established approach to controlling autoimmune and inflammatory diseases (Kopf et al, Nature Reviews Drug Discovery, September 2010, 9:703- 718). Exemplary cytokine targets include Interleukin-ΐβ (IL-Ιβ) (Xu et al, Clin Exp Pharmacol Physiol, 2015, 42(10): 1075-83), Interleukin-2 (IL-2) (Roediger et al., J Allergy Clin Immunol, 2015, 136(6): 1653-63), Interleukin-6 (IL-6) (Scheller et al, Biochem
Biophysica Acta (BBA) - Molecular Cell Research, May 2011, 1813(5): 878-888),
Interleukin-8 (IL-8) (Aghazarian et al., Urology, 2015, 86(l):52-56), Interferon
gamma (IFN-γ) (Di Bari, Clin Ter, 2015, 166(3)), Tumor necrosis factor alpha (TNF-a) (Roubille et al, Ann Rheum Dis, 2015, 74(3):480-489), and Macrophage Inflammatory Protein la (MIP-la) (Dapunt, Mediators Inflamm, 2014, 2014:728619).
[0005] PDE4 inhibition is known to improve therapeutic treatment of a number of inflammatory, respiratory and allergic diseases and conditions (US6649633B2; Keren et al, J Dermatol Sci, January 2015, 77(l):74-76).
[0006] Novel and improved compounds and compositions for treating cancers, autoimmune diseases and inflammatory diseases are desirable. Brief Description of Drawings
[0007] The accompanying drawings illustrate non-limiting example embodiments of the invention.
[0008] FIG. 1 describes the constituents of compounds according to embodiments of the invention. [0009] FIG. 2 shows the structures of compounds according to embodiments of the invention.
[0010] FIG. 3 shows the synthetic process for making compounds according to embodiments of the invention.
[0011] FIG. 4 shows the efficacy of a compound according to an embodiment of the invention in dextran sodium sulphate (DSS)-induced model of inflammatory bowel disease (IBD)/colitis in mice. Detailed Description
[0012] Throughout the following description, specific details are set forth in order to provide a more thorough understanding of the invention. However, the invention may be practiced without these particulars. In other instances, well known elements have not been shown or described in detail to avoid unnecessarily obscuring the invention. Accordingly, the specification and drawings are to be regarded in an illustrative, rather than in a restrictive sense.
[0013] The term "autoimmune disease" as used herein refers to the physiological condition in mammals that is typically characterized by the immune system's reaction to inappropriate targets such as normal tissues. The term "autoimmune disease" includes Addison's disease, ankylosing spondylitis, celiac disease, Hashimoto's thyroiditis, multiple sclerosis, myasthenia gravis, pemphigus, rheumatoid arthritis, Sjogren syndrome, systemic lupus erythematosus, Type 1 diabetes, IBD, psoriasis, vitiligo, alopecia areata and vasculitis. [0014] The term "cancer" as used herein refers to the physiological condition in mammals that is typically characterized by unregulated cell growth. The term "cancer" includes cancer of any origin, including benign and malignant cancers, metastatic and non-metastatic cancers, and primary and secondary cancers. The term "cancer" includes reference to cancer cells. Examples of cancers include, but are not limited to, cancers of the bladder, bone, brain/CNS, breast, cervix, colon, duodenum, esophagus, eye, gall bladder, heart, kidney, larynx, liver, lung, mouth, ovary, pancreas, pharynx, prostate, rectum, stomach, testis, uterus, as well as AIDS-related cancers, Hodgkin's disease, non-Hodgkin's lymphoma, multiple myeloma, melanoma, leukemia (including lymphocytic leukemia, hairy cell leukemia, and acute myelogenous leukemia), choriocarcinoma, rhabdomyosarcoma, and neuroblastoma.
[0015] The term an "effective amount" as used herein refers to the amount of active agent sufficient to elicit a desired biological response or, equivalently, to inhibit an undesired biological response. An amount of a particular active agent that is effective may vary depending on such factors as the desired biological response, severity of the disease, the activity of the active agent to be delivered, the route of administration, the rate of excretion of the active agent being employed, the duration of the treatment, other drugs, compounds or materials used in combination with the particular active agent employed, the subject's age, sex, weight, condition, general health and prior medical history of the subject, and like factors well known in the medical arts. In general, an "effective amount" will be that amount of the active agent that is the lowest dose effective to produce the desired biological response. Such an "effective amount" will generally depend upon the factors described above. Generally, an "effective amount" will range from about 1 to about 400 mg per kilogram of body weight per day, more preferably from about 10 to about 50 mg per kg per day. If desired, daily dosage in an "effective amount" may be administered as one dose, or two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms.
[0016] The term "inflammatory disease" as used herein refers to the physiological condition in mammals that is typically characterized by the immune system's excessive reaction to innocuous targets or reaction to inappropriate targets. The term "inflammatory disease" includes Alzheimer's disease, amyotrophic lateral sclerosis, asthma, atherosclerosis, atopic dermatitis, autoimmune diseases, eczema, inflammatory bowel disease, interstitial cystitis, irritable bowel syndrome, osteoarthritis, Parkinson's disease, periodontal disease, psoriasis and sun burn.
[0017] The term a "package insert" as used herein refers to instructions customarily included in commercial packages of medicaments that contain information about the indications, usage, dosage, administration, contraindications, other medicaments to be combined with the packaged product, and/or warnings concerning the use of such medicaments, and the like. [0018] The term "pharmaceutically acceptable carrier" as used herein refers to one or more excipients, stabilizers, fillers, binders, humectants, disintegrating agents, solution retarding agents, absorption accelerators, wetting agents, absorbents, lubricants, coloring agents, diluents, emulsifiers, preservatives, solubilizing agents, suspending agents and the like that are suitable for use with the subject being exposed thereto at the dosages and concentrations employed without undue adverse side effects (such as toxicity, irritation, and allergic response) commensurate with a reasonable benefit/risk ratio. Examples of pharmaceutically acceptable carriers include water, citrate or phosphate buffers, starches, lactose, sucrose, glucose, mannitol, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, glycerol, agar, calcium carbonate, alginic acid, sodium carbonate, paraffin, quaternary ammonium compounds, cetyl alcohol, glycerol monostearate, kaolin and bentonite clay, talc, calcium stearate, magnesium stearate, polyethylene glycols, sodium lauryl sulfate, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils, tetrahydrofuryl alcohol, fatty acid esters, thioxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, and tragacanth, and mixtures thereof and other ingredients that are well known to those skilled in the art.
[0019] The term "pharmaceutically acceptable salt" as used herein, refers to toxicologically compatible organic or inorganic salts of the active agent. Exemplary salts include, but are not limited, to sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucuronate, saccharate, formate, benzoate, glutamate, methanesulfonate
"mesylate", ethanesulfonate, benzenesulfonate, alkali metal (e.g., sodium and potassium) salts, alkaline earth metal (e.g., magnesium) salts, and ammonium salts. A pharmaceutically acceptable salt can have one or more charged atoms and/or one or more counter ion. If the active agent is a base, the desired pharmaceutically acceptable salt may be prepared by any suitable method available in the art, for example, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, methanesulfonic acid, phosphoric acid and the like, or with an organic acid, such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, a pyranosidyl acid, such as glucuronic acid or galacturonic acid, an alpha hydroxy acid, such as citric acid or tartaric acid, an amino acid, such as aspartic acid or glutamic acid, an aromatic acid, such as benzoic acid or cinnamic acid, a sulfonic acid, such as p-toluenesulfonic acid or ethanesulfonic acid, or the like. If the active agent is an acid, the desired pharmaceutically acceptable salt may be prepared by any suitable method, for example, treatment of the free acid with an inorganic or organic base, such as an amine
(primary, secondary or tertiary), an alkali metal hydroxide or alkaline earth metal hydroxide, or the like. Illustrative examples of suitable salts include, but are not limited to, organic salts derived from amino acids, such as glycine and arginine, ammonia, primary, secondary, and tertiary amines, and cyclic amines, such as piperidine, morpholine and piperazine, and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.
[0020] The term "proliferative disease" as used herein, refers to the physiological condition in mammals that is characterized by the excessive proliferation of cells and turnover of cellular matrix. The term "proliferative disease" includes cancer, graft-versus-host disease, restenosis, hamartoma syndromes (e.g., tuberous sclerosis or Cowden Syndrome), encephalomyelitis, arthritis, scleroderma, pulmonary fibrosis, renal fibrosis, cystic fibrosis, pulmonary hypertension, hypertrophic cardiomyopathy, Parkinson-White syndrome and wet and dry macular degeneration.
[0021] The term "subject" as used herein, refers to an individual to whom an active agent is to be delivered, e.g., for treatment purposes. The term "subject" includes mammals, in particular humans, and other mammals including companion animals such as cats and dogs, livestock animals such as cows, pigs, horses, sheep and goats, zoo animals, and research animals, such as rodents. The "subject" also includes in vitro cultured cells.
[0022] The terms "treat", "treating" and "treatment" as used herein refers to an approach for obtaining desired clinical results. Desired clinical results can include, but are not limited to, reduction or alleviation of at least one symptom of a disease. For example, treatment can be diminishment of at least one symptom of disease, diminishment of extent of disease, stabilization of disease state, prevention of spread of disease, delay or slowing of disease progression, palliation of disease, diminishment of disease reoccurrence, remission of disease, prolonging survival with disease, or complete eradication of disease. Abbreviations:
AKT1 : RAC-alpha serine/threonine -protein kinase
CLK4: CDC2-like kinase 4
DCM: Dichloromethane
DMF: Dimethylformamide
DPPE : Dipheny lphosphinoethane
G-CSF: Granulocyte colony stimulating factor
GM-CSE: Granulocyte macrophage colony stimulating factor
GSK3 : Glycogen synthase kinase 3 beta
IBD: Inflammatory bowel disease
IFN-γ: Interferon gamma
ΙΚΚβ: Inhibitor of nuclear factor kappa-B kinase subunit beta
IL-Ι : Interleukin-1 beta
IL-2: Interleukin-2
IL-8: Interleukin-8
JAK2: Janus Kinase 2
JAK3: Janus Kinase 3
LCK: Lymphocyte specific protein tyrosine kinase
MAP: Mitogen-activated protein
MIP-Ια: Macrophage inflammatory protein 1 alpha
PBMC: Peripheral blood mononuclear cell
p38a: p38 mitogen activated protein kinase alpha ρ38β: p38 mitogen activated protein kinase beta
PDE4: Phosphodiesterase 4
PDE4B1 : Phosphodiesterase 4B1
PHA: Phytohaemagglutinin
PMA: Phorbol 12-myristate 13-acetate SYK: Spleen tyrosine kinase
THF: Tetrahydrofuran
TNF-a: Tumor necrosis factor alpha
[0024] Aspects relate to a compound having Formula I:
Figure imgf000009_0001
Formula I wherein:
R1 is selected from H, halogen and CN;
R2 is selected from H, halogen and CN;
R3 is selected from H, halogen and CN;
R4 is H;
R5 is selected from H, halogen and CN; and
R6 is alkyl or cycloalkyl,
or a pharmaceutically acceptable salt thereof.
[0025] In some embodiments, the compound may be one of exemplary Compounds 1 to 27 set out in FIG. 1. FIG. 2 shows the structure of exemplary Compounds 1. 2. 7, 10, 12, 18, 20. 24 and 27. [0026] In some embodiments, the compound may have Formula I wherein R1 is selected from H, F and CN; R2 is H or CN; R3 is selected from H, F and CN; R4 is H; R5 is H or F; and R6 is selected from iso-propyl, cyclopropyl, cyclopentyl. cyclohexyl and cycloheptyl. [0027] In some embodiments, the compound may have Formula I wherein R1 is H; R2 is H or CN; R3 is selected from H, F and CN; R4 is H; R5 is H; and R6 is selected from iso-propyl, cyclopropyl, cyclopentyl, cyclohexyl and cycloheptyl. [0028] In some embodiments, the compound may have Formula I wherein R1 is H; R2 is H; R3 is F; R4 is H; R5 is H; and R6 is selected from iso-propyl, cyclopropyl and cyclopentyl.
[0029] In some embodiments, the compound may be 3-hydroxy-4-isopropyl-4'-fluoro-(£)- stilbene.
[0030] In some embodiments, the compound having Formula I may be provided as a pharmaceutically acceptable salt.
[0031] Aspects relate to compositions comprising a compound having Formula I and a pharmaceutically acceptable carrier. In some embodiments, compositions comprising compounds of Formula I may be provided in unit dosage form. The amounts of compounds of Formula I that can be combined with a pharmaceutically acceptable carrier to produce a single dosage form will generally be the amount that produces a therapeutic effect. In some embodiments, out of one hundred percent, this amount will range from about 0.1 percent to about 99 percent of active agent, from about 5 percent to about 70 percent of active agent, or from about 10 percent to about 30 percent of active agent.
[0032] Aspects relate to methods of treating a condition by administration to a subject of an effective amount of a compound having Formula I. While it is possible for compounds of Formula I to be administered alone, it may be preferable to administer them in combination with one or more pharmaceutically acceptable carriers as a composition. In some
embodiments, the methods comprise administering to the subject at least one compound having Formula I either alone in or in combination with at least one additional therapeutic agent. As demonstrated below, compounds having Formula I inhibit cell proliferation, inhibit PDE4, inhibit a range of kinases, and inhibit a range of cytokines. As such, a variety of diseases may be treated with compounds having Formula I. Suitable diseases that may be treated include cancers, autoimmune diseases and inflammatory diseases. [0033] The subject to be treated may be any subject diagnosed as having one of the indicated conditions. The subject to be treated may be in need of treatment for one of the indicated conditions because of a diagnosis of the condition or because of an assessment of risk for developing the condition. The subject may be diagnosed with the condition using diagnostic or clinical tests that are well known. Different diagnostic or clinical tests may be used to diagnose different diseases. The diagnostic tools include, without limit, physical examination, patient and family history, screening tests, laboratory tests, imaging tools, physical tests, cognitive tests, and the like. [0034] In some embodiments, the administration route of compounds of Formula I, alone or in a composition, in terms of effect may be local or systemic (enteral or parenteral), and in terms of location may for example be buccal, epi cutaneous, epidural, intraartciular, intracardiac, intracavemous, intracerebral, intracerebroventricular, intradermal, intramuscular, intraosseous, intraperitoneal, intrathecal, intrauterine, intravaginal, intravenous, intravesical, intravitreal, nasal, oral, rectal, subcutaneous, sublingual, sublabial, transdermal, transmucosal, and the like.
[0035] In some embodiments, oral administration may be in the form of capsules, cachets, pills, tablets, lozenges, pastes, powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles or as mouth washes and the like. In some embodiments, compositions in solid dosage forms for oral administration include capsules, tablets, pills, dragees, powders, granules and the like. The solid dosage forms may be scored or prepared with coatings and shells, such as enteric coatings and other coatings. They may also be formulated so as to provide slow or controlled release of compounds of Formula I. In some embodiments, compositions in liquid dosage forms for oral administration include emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In some embodiments, topical or transdermal administration may be in the form of powders, sprays, ointments, pastes, creams, lotions, gels, solutions, controlled-release patches and inhalants. In some embodiments, parenteral administration (e.g. intravenous administration) may be in the form of solutions in physiologically compatible buffers. [0036] Regardless of the route of administration selected, in some embodiments compounds of Formula I are formulated into pharmaceutically acceptable dosage forms by conventional methods. [0037] Aspects relate to a kit comprising compounds of Formula I, and a package insert comprising instructions for using compounds of Formula I to treat a condition in a subject. Suitable conditions that may be treated include cancers, autoimmune diseases and inflammatory diseases. EXAMPLES
[0038] The invention can be further understood by reference to the following examples, which are provided by way of illustration and are not meant to be limiting. EXAMPLE 1
[0039] FIG. 3 illustrates a synthetic method used to prepare compounds of Formula I according to an embodiment of the invention. The compounds are synthesized in six steps from the commercially available starting material, 3-methoxybenzyl alcohol, as follows.
[0040] Step 1 : 3-Methoxybenzyl alcohol was brominated with one molar equivalent of N- bromosuccinimide in tetrahydrofuran (THF) at room temperature to afford 4-bromo-3- methoxybenzyl alcohol. [0041] Step 2: 4-bromo-3-methoxybenzyl alcohol was converted to the corresponding benzyl chloride with an excess of thionyl chloride in dimethylformamide (DMF) at room
temperature.
[0042] Step 3: The benzyl chloride from Step 2 was converted to diethyl 4-bromo-3- methoxybenzylphosphonate with an excess of tri ethyl phosphite upon heating at 150 °C for 16 hours. [0043] Step 4: The diethyl 4-bromo-3-methoxybenzylphosphonate from Step 3 was reacted with an excess of sodium hydride and a substituted benzaldehyde in THF to afford the substituted trans-4-bromo-3-methoxystilbene (Horner-Wadsworth-Emmons Reaction). [0044] Step 5: The substituted trans-4-bromo-3-methoxystilbene from Step 4 was reacted with an alkyl Grignard reagent in the presence of a catalytic amount of N1CI2DPPE
(diphenylphosphinoethane) in diethyl ether at room temperature to afford the corresponding substituted trans-4-alkyl-3-methoxystilbene (Nickel(II)-catalyzed Kumada Cross Coupling Reaction).
[0045] Step 6: The substituted trans-4-alkyl-3-methoxystilbene from Step 5 was reacted with an excess of boron tribromide in dichloromethane (DCM) at room temperature to afford the substituted trans-4-alkyl-3-hydroxystilbene. EXAMPLE 2
[0046] The synthetic procedure for 3-hydroxy-4-isopropyl-4'-fluoro-(£) -stilbene
(Compound 2) according to an embodiment of the invention is presented below. All compounds of the invention may be prepared using this process.
4-Bromo-3-methoxybenzyl alcohol
[0047] To a solution of 3-methoxybenzyl alcohol (20.0 g, 145 mmol) in THF (400 mL) was added N-bromosuccinimide (26 g, 146 mmol) and the resultant mixture was stirred at room temperature for 8 h. Ether (500 mL) was added and the mixture was washed with water (3 x 100 mL). The solution was dried over magnesium sulfate and concentrated to afford the title compound (31.2 g, 99%) which was used as such for the next step.
Diethyl 4-bromo-3-methoxybenzylphosphonate
[0048] To a solution of 4-bromo-3-methoxybenzyl alcohol (31.0 g, 143 mmol) in DMF (200 mL) at 0 °C was added thionyl chloride (17.5 mL, 241 mmol) dropwise over the course of 20 minutes. The resultant mixture was allowed to warm to room temperature and stirring was continued for 24 hours. The reaction was quenched with water and extracted with ether. The combined organic extracts were washed with water and dried over magnesium sulfate before being concentrated in vacuo. The residue was taken up in triethylphosphite (200 mL) and the solution was heated to 150 °C for 4 hours. Excess triethylphosphite was removed in vacuo to afford the crude product which was purified by vacuum distillation to afford the title compound as a colorless oil which solidified upon standing (34.7 g, 72%).
4-Bromo-5-methoxy-4'-fluoro-(E)-stilbene [0049] To a stirred solution of diethyl (4-bromo-3-methoxybenzyl) phosphonate (13.50 g, 40.0 mmol) and 4-fluorobenzaldehyde (5.68 g, 40.0 mmol) in THF (125 mL) at room temperature was added an aqueous solution of sodium hydroxide (50%, 8.0 mL) drop-wise over the course of 30 minutes. The resultant solution was allowed to stir overnight. The reaction mixture was treated with water (200 mL) and extracted with ethyl acetate. The combined organic extracts were dried over magnesium sulfate and concentrated in vacuo to afford the crude product which was purified by recrystallization from hexanes to afford the title compound as a white crystalline solid (9.8 g, 80%).
3 -Methoxy-4-isopropyl-4 ' -fluoro-(E)-stilbene
[0050] To a solution of 4-bromo-5-methoxy-4'-fluoro-(E)-stilbene (1.5 g, 4.9 mmol) in ether (30 mL) was added [l,2-bis(diphenylphosphino)ethane] dichloronickel(II) (52 mg, 0.098 mmol). A solution of isopropylmagnesium chloride in THF (2.0 M, 2.7 mL, 5.4 mmol) was then added drop-wise over the course of 5 minutes and the resultant solution was allowed to stir for 24 hours. The reaction mixture was quenched with water (5 mL), diluted with additional ether (60 mL) and the resultant mixture was washed first with a saturated aqueous solution of sodium thiosulfate and then with water. The organic layer was dried over anhydrous magnesium sulfate and concentrated in vacuo to afford the crude product which was purified by recrystallization from hexanes to afford the title compound as a light yellow crystalline solid (874 mg, 66%).
3 -Hy droxy-4-isopropyl-4 ' -fluoro-^-stilbene [0051] To a solution of 3-methoxy-4-isopropyl-4'-fluoro-(£')-stilbene (500 mg, 1.85 mmol) in DCM (45 niL) was added boron tribromide (1.4 mL, 15 mmol) and the resultant mixture was stirred for 2 hours. The reaction was quenched by the slow addition of water and was extracted with DCM. The combined organic extracts were dried over magnesium sulfate and concentrated in vacuo to afford the crude product which was purified by recrystallization from hexanes to afford the title compound as a white crystalline solid (327 mg, 69%).
EXAMPLE 3 - Effects on Cell Viability [0052] Cells were cultured in RPMI-1640 containing 10% FBS (Jurkat and THP-1) or
DMEM containing 10% FBS (A549 and MDA-MB-435). On Day 1, l-5xl04 cells per well in 100 of the cell culture medium containing 0.5% FBS were seeded into a 96-well flat bottom plate. On Day 2, the test compounds were added to the culture at different concentrations in triplicates. The controls include triplicates of no compound treatment and cell culture medium without cells. The cells were incubated at 5% C02, 37 °C for 48 hours. On Day 4, MTS reagent was added to each well and incubated at 37 °C for 4 hours. The absorbance was read at 490 nm. The percentage of inhibition was calculated using the formula: [1 -(experiment reading-background reading)/(negative control reading-background reading)] x 100. The determined cell viability IC50 values are listed in Table 1.
Table 1. Cell Viability IC50 Values of Compounds against MB435, Jurkat, THP1 and A549
Cells.
Figure imgf000015_0001
EXAMPLE 4 - Effects on Cell Proliferation
[0053] Cells were cultured in RPMI-1640 containing 10% FBS (Jurkat) or DMEM containing 10% FBS (MDA-MB-435). On Day 1, lxlO4 cells were seeded into one well of a 96-well white ISOPLATE™ with 0.5% FBS. The cells were cultured at 5% C02, 37 °C overnight. On Day 2, the diluted test compounds were added into each well in a total volume of 100 of cell culture medium. For each cell line were set up three wells containing cells and only diluted DMSO as a negative (vehicle) control, and three wells without cells but containing the same volume and type of cell culture medium as background control. The cells were incubated at 5% C02, 37 °C for 24 hours. On Day 3, 1 of [3H] -thymidine was added into each well and incubation at 5% C02, 37 °C was continued for another 24 hours. On Day 4, 30 of 50% TCA was added into each well and was incubated at 4 °C for 4 hours. Plates were washed with ddH20 four times and then air dried for 30 minutes followed by the addition of 100 μΐ. of scintillation fluid into each well. The radioactivity was read using
MICROBETA TRILUX™. The percentage of inhibition was calculated using the formula [1- (experiment reading-background reading)/(negative control reading-background reading)] x 100. The determined anti -proliferation IC50 values are shown in Table 2. Table 2. Anti-proliferation IC50 Values of Compounds against MB435 and Jurkat Cells.
Figure imgf000016_0001
EXAMPLE 5 -Effects on Protein Kinase Activities
[0054] Kinase profiling was conducted using different recombinant kinase targets against the compounds at 5 uM ATP. The kinases tested included AKTl (RAC-alpha serine/threonine- protein kinas 1), JAK2, JAK3, ρ38α, ρ38β (p38 mitogen-activated protein kinase beta), GSK3fi, SYK (Spleen tyrosine kinase), LCK, IKKa and ΙΚΚβ and CLK4. The test compounds were tested at 5 μΜ and 50 μΜ in triplicates. The results are shown in Table 3.
Table 3. Effects of Compounds on Tested Kinase Activities.
Figure imgf000017_0001
. Where "-" is less than 20% inhibition, "+" is 20-30% inhibition, "++" is 31-50% inhibition, "+++" is greater than 50% inhibition.
EXAMPLE 6 - Effects on phosphodiesterase 4 (PDE4) Activity
[0055] Assays were performed using recombinant human PDE4B1 enzyme expressed in a baculoviral system. The radiometric assay is a modification of the 2-step method of Thompson and Appleman (Biochemistry 10, 1971, 311-316). The reactions were performed at 1 μΜ of cAMP. The test articles were each tested at 0.2 μΜ and 20 μΜ in triplicates. The percentage inhibition values for the reference inhibitor, rolipram, were also determined and compared to historical assay values to ensure it was in an acceptable range. The results are shown in Table 4. Table 4. Inhibitory Activities on PDE4B1 Enzyme.
Figure imgf000018_0001
Where "-" is less than 15% inhibition, "+" is 15-30% inhibition, "++" is 31-50% inhibition and "+++" is greater than 50% inhibition
EXAMPLE 7 - Effects on Human Cytokine Production
[0056] Commercially available PBMC cells were used to test the effects of the compounds on the production multiple cytokines. Around one hundred million human PBMC (lxlO8) were thawed in a 37 °C water bath and recovered by culturing for 3 hours in RPMI 1640 with 10% fetal bovine serum (FBS) at 37 °C in a C02 incubator. Cytokine production was induced with a combination of PMA and PHA 30 minutes post the compound treatments. Cells were treated with various concentrations of the compounds for 24 hours at 37 °C. To determine the cytokine production secreted into the culture medium by PBMC cells, the culture supernatants were collected from drug-treated or control samples. Commercial ELISA kits (QIAGEN™ ELISAarray Kits) were used for the quantitation. Cytokines tested include: IL- la, IL-1 β, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12, IL-13, IL-17A, IFN-τ, TNF-a, G- CSF(Granulocyte -colony -stimulating factor), GM-CSF(Granulocyte-macrophage colony- stimulating factor), TGF-β 1 and ΜΙΡΙ . The effects of the compounds on those cytokines demonstrated to be consistently stimulated by phorbol 12-myristate 13-acetate (PMA) and polyhydroxy fatty acid (PHA) are shown in Table 5. Table 5. Inhibition of PMA- and PHA-induced Cytokine Production in Human PBMC Cells.
Figure imgf000019_0001
Where "-" is no inhibition, "+" is >50% inhibition at 10 μΜ, "++" is >50% inhibition at 1 μΜ
EXAMPLE 8 - Effect on inflammatory bowel disease (IBD)/colitis in mice [0057] The efficacy of Compound 2 was tested in a dextran sodium sulphate (DSS) induced model of inflammatory bowel disease (IBD)/colitis in female Balb/c mice. Rolipram was used as a positive control in this experiment. Colitis was induced in the mice by providing drinking water containing DSS (3.5%, M. Wt. 30,000-50,000) from Day 0 to Day 8. The test compounds were administered as per their respective groups at specific dose levels starting from Day 0 through to Day 8. Compound 2 was tested at 10 and 30 mg/kg and Rolipram was tested at 5 mg/kg. Colitis was consistently induced in most of the mice, with onset of disease (stool consistency) from Day 2 and drastic reduction in body weight, diarrhea and severe rectal bleeding at Day 5-7 demonstrating the severity of disease. For dosing, Compound 2 was dissolved in vehicle (1.5% Cremophor EL) at 3 mg/mL and 1 mg/mL for the 30 and 10 mg/kg doses, respectively. Rolipram was dissolved in 1% DMSO/99% PBS at 0.5 mg/mL for the 5 mg/kg dose. All doses were administered by oral gavage.
[0058] There was a considerable increase in disease activity index (DAI) (cumulative scores of body weight, stool consistency and rectal bleeding) in the DSS control group (24.2 + 5.7) when compared to normal control group (1.9 + 2.4) which indicated very good disease induction. In treatment groups, Compound 2 at both 10 mg/kg (17.4 + 12.0) and 30 mg/kg (15.4 + 4.9) showed very good reduction in DAI when compared to DSS control group (24.2 + 5.7). The positive control rolipram at 5 mg/kg dose (13.8 + 8.1) demonstrated very good inhibition on DAI as compared to DSS control group (24.2 + 5.7). Results are shown in FIG. 4.
EXAMPLE 9 - Efficacy against the inflammatory skin conditions psoriasis and eczema [0059] A topical cream containing compound 2 was prepared as follows: Compound 2 was dissolved in propylene glycol at a concentration of 11.0% w/w and this solution was then mixed with the appropriate amount of Glaxal base cream to afford a 1.0% w/w cream of compound 2. [0060] To test the efficacy of this cream against the inflammatory skin conditions psoriasis and eczema, 10 subjects with psoriasis and 5 subjects with eczema were treated with the cream twice daily for a period of 2-12 weeks. Amongst the 10 subjects with psoriasis, 8 of them showed a significant reduction in the skin thickness (induration), erythema and scaling after 8 weeks of treatment. The majority of the subject's lesions were cleared after 12 weeks of treatment. All 5 subjects with eczema responded to the treatment with significantly reduced skin inflammation and pruritis beginning as early as after 2 weeks of treatment.
[0061] As those skilled in the art will appreciate, the methods and uses described herein are only examples of many conditions where the invention may be applied to produce therapeutic benefit. Specific pharmacological responses observed may vary according to and depending on the particular active agent(s) and/or pharmaceutical acceptable carrier(s) selected, as well as the type of formulation and mode of administration employed, and such expected variations or differences in the results are contemplated in accordance with practice of the present invention.

Claims

What is claimed is:
1. A compound having general formula I, the general formula I is shown as following
Figure imgf000022_0001
wherein R1 is selected from H, halogen and CN; R2 is selected from H, halogen and CN; R3 is selected from H, halogen and CN; R4 is H; R5 is selected from H, halogen and CN; and R6 is alkyl or cycloalkyl.
2. The compound according to claim 1, wherein R1 is selected from H, F and CN; R2 is H or CN; R3 is selected from H, F and CN; R4 is H; R5 is H or F; and R6 is selected from iso- propyl, cyclopropyl, cyclopentyl, cyclohexyl and cycloheptyl.
3. The compound according to claim 1, wherein R1 is H; R2 is H or CN; R3 is selected from H, F, CN and OCH3; R4 is H; R5 is H; and R6 is selected from iso-propyl, cyclopropyl, cyclopentyl, cyclohexyl and cycloheptyl.
4. The compound according to claim 1, wherein R1 is H; R2 is H; R3 is F; R4 is H; R5 is H; and R6 is selected from iso-propyl, cyclopropyl and cyclopentyl.
5. The compound according to claim 1, wherein the compound is 3-hydroxy-4- isopropyl-4'-fluoro-(£)-stilbene.
6. A composition comprising a compound according to claim 1, and a pharmaceutically acceptable carrier.
7. A method of treating a disease in a subject comprising administering to the subject an effective amount of a compound according to claim 1.
8. The method according to claim 7, wherein the disease is selected from a group consisting of an autoimmune disease,an inflammatory disease or a proliferative disease.
9. The method according to claim 8, wherein the proliferative disease is a cancer.
10. The method according to claim 8, wherein the inflammatory disease is inflammatory bowel disease, psoriasis or eczema
11. The method according to claim 7, wherein the disease is characterized by enhanced activity of one or more kinases selected from the group consisting of JAK2, JAK3, p38a, ρ38β, GSK3fi, LCK, ΙΚΚβ and CLK4
12. The method according to claim 7, wherein the disease is characterized by enhanced expression of one or more cytokines selected from the group consisting of IL-Ιβ, IL-2, IL-6, IFN-γ, TNF-a and MIP-1A.
13. The method according to claim 7, wherein the disease is characterized by enhanced activity of PDE4.
14. A method for inhibiting kinase activity in a subject with an effective amount of a compound of claim 1.
15. The method according to claim 14, wherein the kinase is selected from the group consisting of JAK2, JAK3, ρ38α, ρ38β, GSK3fi, LCK, ΙΚΚβ and CLK4.
16. A method for inhibiting production of a cytokine in a subject with an effective amount of a compound of claim 1.
17. The method according to claim 16, wherein the cytokine is selected from the group consisting of IL-Ιβ, IL-2, IL-6, IFN-γ, TNF-a and MIP-1A.
18. A method for inhibiting PDE4 activity in a subject with an effective amount of a compound of claim 1.
19. A process for using a compound according to claim 1 for treating a disease in a subject.
20. The process according to claim 19, wherein the disease is selecting from a group consisting of an autoimmune disease, an inflammatory disease, or a proliferative disease.
21. The process according to claim 20, wherein the proliferative disease is a cancer.
22. The process according to claim 20, wherein the inflammatory disease is an inflammatory bowel disease, psoriasis or eczema
23. The process according to claim 19, wherein the disease is characterized by enhanced activity of one or more kinases selected from the group consisting of JAK2, JAK3, p38a, ρ38β, GSK3fi, LCK, ΙΚΚβ and CLK4.
24. The process according to claim 19, wherein the disease is characterized by enhanced expression of one or more cytokines selected from the group consisting of IL-Ιβ, IL-2, IL-6, IFN-γ, TNF-a and MIP-IA.
25. The process according to claim 19, wherein the disease is characterized by enhanced activity of PDE4.
26. A kit comprising an active agent comprising a compound of claim 1, and a package insert comprising instructions for using the active agent to treat a disease in a subject.
27. The kit according to claim 26, wherein the disease is selecting from a group consisting of , an autoimmune disease, an inflammatory disease or a proliferative disease.
28. A method for manufacturing a compound according to claim 1, comprising the following steps:
i) 3-Methoxybenzyl alcohol is brominated with a molar equivalent of N- bromosuccinimide in THF at room temperature to yield 4-bromo-3- methoxybenzyl alcohol;
ii) 4-bromo-3-methoxybenzyl alcohol is converted into the corresponding benzyl
chloride with an excess of thionyl chloride in DMF at room temperature; iii) the benzyl chloride from step ii) is converted into diethyl 4-bromo-3- methoxybenzylphosphonate with an excess of triethyl phosphite upon heating at 150 °C for 16 hours;
iv) the diethyl 4-bromo-3-methoxybenzylphosphonate from step iii) is reacted with an excess of sodium hydride and a substituted benzaldehyde in THF to yield a substituted trans-4-bromo-3-methoxystilbene (Horner- Wadsworth-Emmons Reaction);
v) the substituted trans-4-bromo-3-methoxystilbene from step iv) is reacted with an alkyl
Grignard reagent in the presence of a catalytic amount of N1CI2DPPE in a diethyl ether at room temperature to yield a substituted trans-4-alkyl-3-methoxystilbene (Nickel(II)-catalyzed Kumada Cross Coupling Reaction);
vi) the substituted trans-4-alkyl-3-methoxystilbene from step v) is reacted with an excess of boron tribromide in DCM at room temperature to yield the substituted trans-4- alkyl-3-hydroxystilbene.
PCT/CA2017/051182 2016-10-13 2017-10-04 Substituted hydroxystilbenes and their therapeutic applications WO2018068131A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
CN201780062107.0A CN109890784B (en) 2016-10-13 2017-10-04 Substituted hydroxystyrenes and therapeutic uses thereof
EP17860520.0A EP3526186B1 (en) 2016-10-13 2017-10-04 Substituted hydroxystilbenes and their therapeutic applications
ES17860520T ES2924065T3 (en) 2016-10-13 2017-10-04 Substituted hydroxystilbenes and their therapeutic applications
JP2019520644A JP6936319B2 (en) 2016-10-13 2017-10-04 Substituted hydroxystilbenes and their therapeutic applications
CA3038575A CA3038575C (en) 2016-10-13 2017-10-04 Substituted hydroxystilbenes and their therapeutic applications
AU2017344103A AU2017344103B2 (en) 2016-10-13 2017-10-04 Substituted hydroxystilbenes and their therapeutic applications
KR1020197009817A KR102419043B1 (en) 2016-10-13 2017-10-20 Substituted hydroxystilbenes and therapeutic uses thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201662407886P 2016-10-13 2016-10-13
US62/407,886 2016-10-13
US15/338,232 2016-10-28
US15/338,232 US9725406B1 (en) 2016-10-13 2016-10-28 Substituted hydroxystilbenes and their therapeutic applications

Publications (1)

Publication Number Publication Date
WO2018068131A1 true WO2018068131A1 (en) 2018-04-19

Family

ID=59411070

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2017/051182 WO2018068131A1 (en) 2016-10-13 2017-10-04 Substituted hydroxystilbenes and their therapeutic applications

Country Status (9)

Country Link
US (2) US9725406B1 (en)
EP (1) EP3526186B1 (en)
JP (1) JP6936319B2 (en)
KR (1) KR102419043B1 (en)
CN (1) CN109890784B (en)
AU (1) AU2017344103B2 (en)
CA (1) CA3038575C (en)
ES (1) ES2924065T3 (en)
WO (1) WO2018068131A1 (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9725406B1 (en) * 2016-10-13 2017-08-08 Resolvex Pharmaceuticals Inc. Substituted hydroxystilbenes and their therapeutic applications
SG11202008361YA (en) * 2018-03-27 2020-09-29 Neuropore Therapies Inc Compounds as modulators of tlr2 signaling
CN110183311B (en) * 2019-07-25 2019-10-29 江西中医药大学 A kind of isopentene group stilbene and its purposes in preparation treatment diseases associated with inflammation drug
CN110496117B (en) * 2019-09-30 2022-09-06 中国科学院微生物研究所 New application of small molecular compound in resisting African swine fever virus infection
WO2023175177A1 (en) * 2022-03-18 2023-09-21 Institut Curie Differential diagnosis of crohn's disease and ulcerative colitis
CN117229208B (en) * 2023-11-13 2024-02-23 上海泽德曼医药科技有限公司 Condensed ring compound, preparation method and medical application thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6649633B2 (en) 2001-01-31 2003-11-18 Pfizer Inc Nicotinamide biaryl derivatives useful as inhibitors of PDE4 isozymes
US20080255245A1 (en) 1999-12-06 2008-10-16 Welichem Biotech Inc. Anti-Inflammatory and Psoriasis Treatment and Protein Kinase Inhibition by Hydroxystilbenes and Novel Stilbene Derivatives and Analogues

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7321050B2 (en) * 1999-12-06 2008-01-22 Welichem Biotech Inc. Anti-inflammatory and psoriasis treatment and protein kinase inhibition by hydroxy stilbenes and novel stilbene derivatives and analogues
JP4880847B2 (en) * 1999-12-06 2012-02-22 ウェリケム,バイオテック インコーポレーテッド Anti-inflammatory and psoriasis treatments and protein kinase inhibition with hydroxylstilbene and novel stilbene derivatives and analogs
CN1955153A (en) * 2005-10-24 2007-05-02 南京莱因医药科技有限公司 Synthetic method of pterostilbene
US9725406B1 (en) * 2016-10-13 2017-08-08 Resolvex Pharmaceuticals Inc. Substituted hydroxystilbenes and their therapeutic applications

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080255245A1 (en) 1999-12-06 2008-10-16 Welichem Biotech Inc. Anti-Inflammatory and Psoriasis Treatment and Protein Kinase Inhibition by Hydroxystilbenes and Novel Stilbene Derivatives and Analogues
US6649633B2 (en) 2001-01-31 2003-11-18 Pfizer Inc Nicotinamide biaryl derivatives useful as inhibitors of PDE4 isozymes

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
AGHAZARIAN ET AL., UROLOGY, vol. 86, no. 1, 2015, pages 52 - 56
BAUER ET AL.: "Studies in Relation to Biosynthesis", AUSTRALIAN JOURNAL OF CHEMISTRY, 1955, pages 534 - 538, XP009514079, ISSN: 0004-9425 *
DI BARI, CLIN TER, vol. 166, no. 3, 2015
JAIN ET AL., CURR DRUG TARGETS, vol. 15, no. 5, 2014, pages 539 - 550
KEREN ET AL., J DERMATOL SCI, vol. 77, no. 1, January 2015 (2015-01-01), pages 74 - 76
KOPF ET AL., NATURE REVIEWS DRUG DISCOVERY, vol. 9, September 2010 (2010-09-01), pages 703 - 718
NOVOSELOVA ET AL., MEDIATORS INFLAMM, vol. 2014, 2014, pages 728619
ROEDIGER ET AL., J ALLERGY CLIN IMMUNOL, vol. 136, no. 6, 2015, pages 1653 - 63
ROUBILLE ET AL., ANN RHEUM DIS, vol. 74, no. 3, 2015, pages 480 - 489
SCHELLER ET AL., BIOCHEM BIOPHYSICA ACTA (BBA) - MOLECULAR CELL RESEARCH, vol. 1813, no. 5, May 2011 (2011-05-01), pages 878 - 888
See also references of EP3526186A4
XU ET AL., CLIN EXP PHARMACOL PHYSIOL, vol. 42, no. 10, 2015, pages 1075 - 83

Also Published As

Publication number Publication date
ES2924065T3 (en) 2022-10-04
CA3038575C (en) 2023-08-01
EP3526186A1 (en) 2019-08-21
EP3526186A4 (en) 2020-04-29
US10166201B2 (en) 2019-01-01
CN109890784A (en) 2019-06-14
CA3038575A1 (en) 2018-04-19
US9725406B1 (en) 2017-08-08
AU2017344103A1 (en) 2019-04-04
JP2019532076A (en) 2019-11-07
JP6936319B2 (en) 2021-09-15
US20180118667A1 (en) 2018-05-03
KR20190101953A (en) 2019-09-02
CN109890784B (en) 2022-04-29
KR102419043B1 (en) 2022-07-07
AU2017344103B2 (en) 2021-07-01
EP3526186B1 (en) 2022-05-04

Similar Documents

Publication Publication Date Title
AU2017344103B2 (en) Substituted hydroxystilbenes and their therapeutic applications
JP7085566B2 (en) Apoptosis inducer
US20130281396A1 (en) Treatment of diseases by epigenetic regulation
EP3705480B1 (en) Class of amino-substituted nitrogen-containing fused ring compounds, preparation method therefor, and use thereof
CN102603743A (en) Anti-tumor benzazepine[f]azulene derivative, preparation method thereof, and purpose thereof
DE60117835T2 (en) Amino-substituted tetracyclic compounds useful as anti-inflammatory agents and drugs containing them
CN111116469A (en) HDAC inhibitor, preparation method, pharmaceutical composition and application thereof
US11000485B2 (en) Substituted hydroxystilbenes as AhR and Nrf2 modulators and their therapeutic applications
US20150299185A1 (en) Novel imidazopyridine derivatives as a tyrosine kinase inhibitor
EA003941B1 (en) 2-aminopyridines containing fused ring substituents
CN110054627A (en) A kind of novel IDO inhibitor, preparation method, medical composition and its use
JP7117029B2 (en) 2-Substituted pyrazoleamino-4-substituted amino-5-pyrimidineformamide compounds, compositions and uses thereof
JP2022527279A (en) Quinoline derivatives and their use for the treatment of cancer
CN111116551A (en) 1-azaspiro [5.5] undecan-3-ones and 1-azaspiro [5.5] undecan-3-ols
JP2009507858A (en) Quinoline derivatives and use as antitumor agents
CN113549046B (en) Bisbecklonin S derivative and preparation method and application thereof
CN113801110B (en) 1,2, 4-oxadiazole heterocyclic compound and application thereof
CN111039940B (en) Aurora A kinase inhibitor, preparation method, pharmaceutical composition and application thereof
CN107281180A (en) Application of the 8- alkyl berberine salts in prevention and treatment lung-cancer medicament is prepared
CN101317845A (en) Pharmaceutical use of 6-aryl substituted pyridine compounds
CN106986821B (en) A kind of aminomethyl pyridine analog derivative and its preparation method and application
WO2020228463A1 (en) Preparation method for and use of anti-leukemia selenium-substituted noscapine derivatives
NZ199887A (en) Phenanthroline derivatives and pharmaceutical compositions
CN101284007B (en) Pharmaceutical use of aryl group substituted pyridone derivates in preparing medicine for tumour
CN117304148A (en) 3, 4-disubstituted gamma-butyrolactone derivative, preparation method thereof and application of 3, 4-disubstituted gamma-butyrolactone derivative in preparation of antitumor drugs

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: 17860520

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3038575

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2017344103

Country of ref document: AU

Date of ref document: 20171004

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20197009817

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2019520644

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2017860520

Country of ref document: EP

Effective date: 20190513