WO2021087127A1 - Cannabinoids and uses thereof - Google Patents

Cannabinoids and uses thereof Download PDF

Info

Publication number
WO2021087127A1
WO2021087127A1 PCT/US2020/057985 US2020057985W WO2021087127A1 WO 2021087127 A1 WO2021087127 A1 WO 2021087127A1 US 2020057985 W US2020057985 W US 2020057985W WO 2021087127 A1 WO2021087127 A1 WO 2021087127A1
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
compound
fibrosis
disease
membered heterocyclyl
Prior art date
Application number
PCT/US2020/057985
Other languages
French (fr)
Other versions
WO2021087127A8 (en
Inventor
Hongfeng Deng
Tama Evron
Clifton David LEIGH
Cesar Reiriz ARMENTAL
Original Assignee
Corbus 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 Corbus Pharmaceuticals, Inc. filed Critical Corbus Pharmaceuticals, Inc.
Publication of WO2021087127A1 publication Critical patent/WO2021087127A1/en
Publication of WO2021087127A8 publication Critical patent/WO2021087127A8/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/04Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicines Containing Plant Substances (AREA)

Abstract

The invention relates to cannabinoid compounds, pharmaceutical compositions including one or more cannabinoid compounds, and the use of pharmaceutical compositions including one or more cannabinoid compounds for the treatment of a disease or condition (e.g., a fibrotic disease or an inflammatory disease) in a subject in need thereof.

Description

CANNABINOIDS AND USES THEREOF
Background of the Invention
Cannabinoids are a class of chemicals found in Cannabis sativa L (Cannabis) and related derivatives that have been shown to exhibit various pharmacologic activities. Tetrahydrocannabinol (THC) is the major psychoactive cannabinoid of cannabis. In addition to mood-altering effects, THC has been reported to exhibit other activities, some of which may have therapeutic value. The potential therapeutic value of THC has led to a search for related compounds which minimize the psychoactive effects, while retaining the activities of potential medicinal value.
Cannabinoids in current therapeutic use, such as nabilone, activate both the cannabinoid type 1 receptor (CBi) and the cannabinoid type 2 receptor (CB2). Selective CB2 modulations or peripherally restricted CB2 modulation may provide some of the therapeutic effects of cannabinoids, such as their immuno-modulatory properties, without the psychoactive effects of CBi activation associated with central neuron system. Therefore, cannabinoid CB2 receptors represent an attractive target for drug development.
(6aR,10aR)-1 -hydroxy-6, 6-dimethyl-3-(2-methyl-2-octanyl)-6a, 7, 10,10a-tetrahydro-6H- benzo[c]chromene-9-carboxylic acid (also known as ajulemic acid, AJA, JBT-101 , resunab, anabasum, or lenabasum) has been investigated for its potential therapeutic benefits in a number of diseases, including fibrotic diseases and inflammatory diseases, for which there is a need for new therapies with improved safety and efficacy profiles. Ajulemic acid has been shown to exhibit receptor selectivity for CB2 over CBi as well as preferential peripheral distribution.
There is a continued need for the development of cannabinoids with improved potency and selectivity for the CB2 receptor.
Summary of the Invention
The invention relates to cannabinoid compounds, pharmaceutical compositions including one or more cannabinoid compounds, and the use of pharmaceutical compositions including one or more cannabinoid compounds for the treatment of a disease or condition (e.g., a fibrotic disease or an inflammatory disease) in a subject in need thereof. In particular, the invention features compounds sharing structural features with (6aR,10aR)-1-hydroxy-6,6-dimethyl-3-(2-methyl-2-octanyl)-6a,7,10,10a- tetrahydro-6H-benzo[c]chromene-9-carboxylic acid (ajulemic acid). In some embodiments, the invention features compounds which are modulators (e.g., agonists, inverse agonists, or antagonists) of the CB2 receptor. In preferred embodiments of the invention, the invention features compounds that have increased affinity for the CB2 receptor (e.g., increased affinity for the CB2 receptor compared to ajulemic acid), increased selectivity for the CB2 receptor (e.g., increased selectivity for the CB2 receptor over the CBi receptor compared to ajulemic acid), or both increased affinity and increased selectivity for the CB2 receptor. In some embodiments, the invention features compounds with an increased safety or efficacy profile in the treatment of a disease or condition (e.g., a fibrotic disease or an inflammatory disease), as compared to other cannabinoids, such as ajulemic acid. In some embodiments, the invention features compounds having improved pharmacokinetic properties or improved stability (e.g., improved pharmacokinetic properties or improved stability as compared to ajulemic acid). In a first aspect, the invention features a compound described by formula (I):
Figure imgf000004_0001
wherein Ri is optionally substituted 3-to-8 membered heterocyclyl; R2 IS H, OH, F, Cl, Br, NH2, or optionally substituted C1-C3 alkoxy; R3 is optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C1-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C1-C20 heteroalkynyl, optionally substituted C5-C15 aryl, optionally substituted C2-C15 heterocyclyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C1-C20 alkoxy, or optionally substituted amide; R4 and Rs are each independently H, -CH3, -CF3, -CH2OH, -CH2F, or -CHF2; and R6 is -CH3 or -CH2OH, or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (II):
Figure imgf000004_0002
wherein Ri is optionally substituted 3-to-8 membered heterocyclyl; R2 IS H, OH, F, Cl, Br, NH2, or optionally substituted C1-C3 alkoxy; and R3 is optionally substituted C1-C20 alkyl, optionally substituted Ci- C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C1-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C1-C20 heteroalkynyl, optionally substituted C5- C15 aryl, optionally substituted C2-C15 heterocyclyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C1-C20 alkoxy, or optionally substituted amide; or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (III):
Figure imgf000004_0003
wherein Ri is optionally substituted 3-to-8 membered heterocyclyl; and R3 optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C1-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C1-C20 heteroalkynyl, optionally substituted C5-C15 aryl, optionally substituted C2-C15 heterocyclyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C1-C20 alkoxy, or optionally substituted amide; or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by any one of formulas (111-1), (III-2), or (III-3):
Figure imgf000005_0001
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (IV):
Figure imgf000005_0002
wherein Ri is optionally substituted 3-to-8 membered heterocyclyl; and R3 optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C1-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C1-C20 heteroalkynyl, optionally substituted C5-C15 aryl, optionally substituted C2-C15 heterocyclyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C1-C20 alkoxy, or optionally substituted amide; or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by any one of formulas (IV-1), (IV-2), or (IV-3):
Figure imgf000006_0001
or a pharmaceutically acceptable salt thereof.
In some embodiments of any of the aspects described herein (e.g., any one of formulas (I), (II), (III), (MI-1), (III-2), (III-3), (IV), (IV-1), (IV-2), or (IV-3)), Ri is optionally substituted 3-to-8 membered heterocyclyl (e.g., an optionally substituted 5-membered heterocyclyl or optionally substituted 6- membered heterocyclyl). In preferred embodiments, the compound is described by any one of formulas (III-1), (IM-2), or (MI-3) and Ri is optionally substituted 3-to-8 membered heterocyclyl (e.g., an optionally substituted 5-membered heterocyclyl or optionally substituted 6-membered heterocyclyl). In preferred embodiments, the compound is described by any one of formulas (IV-1), (IV-2), or (IV-3) and Ri is optionally substituted 3-to-8 membered heterocyclyl (e.g., an optionally substituted 5-membered heterocyclyl or optionally substituted 6-membered heterocyclyl).
In some embodiments of any of the aspects described herein (e.g., any one of formulas (I), (II), (III), (MI-1), (MI-2), (MI-3), (IV), (IV-1), (IV-2), or (IV-3)), Ri is optionally substituted pyrrole, optionally substituted pyrazole, optionally substituted isoxazole, optionally substituted pyrrolidine, optionally substituted imidazole, optionally substituted thiazole, optionally substituted thiophene, optionally substituted thiolane, optionally substituted furan, optionally substituted tetrahydrofuran, optionally substituted diazole, optionally substituted triazole, optionally substituted tetrazole, optionally substituted oxazole, optionally substituted 1 ,3,4-oxadiazole, optionally substituted 1 ,3,4-thiadiazole, optionally substituted 1 ,2,3,4-oxatriazole, or optionally substituted 1 ,2,3,4-thiatriazole. In some embodiments of any of the aspects described herein (e.g., any one of formulas (I), (II),
Figure imgf000007_0001
In some embodiments of any of the aspects described herein (e.g., any one of formulas (I), (II), (III), (MI-1), (MI-2), (MI-3), (IV), (IV-1), (IV-2), or (IV-3)), Ri is preferably
Figure imgf000007_0004
(e.g., any one of formulas (I), (II), (III), (MI-1), (MI-2), (MI-3), (IV), (IV-1), (IV-2), or (IV-3)), Ri is optionally substituted pyridine, optionally substituted piperidine, optionally substituted piperazine, optionally substituted pyrimidine, optionally substituted pyrazine, optionally substituted pyridazine, optionally substituted triazine, optionally substituted azepine, optionally substituted 1 ,4-diazepine, optionally substituted 2/-/-pyran, optionally substituted 4/-/-pyran, optionally substituted tetrahydropyran, optionally substituted thiepine, or optionally substituted 1 ,4-thiazepine.
In some embodiments of any of the aspects described herein (e.g., any one of formulas (I), (II), (III), (MI-1), (ill-2), (ill-3), (IV), (IV-1), (IV-2), or (IV-3)), Ri is
Figure imgf000007_0002
In some embodiments of any of the aspects described herein (e.g., any one of formulas (I), (II), (III), (ill-1), (ill-2), (ill-3), (IV), (IV-1), (IV-2), or (IV-3)), Ri is preferably
Figure imgf000007_0003
In some embodiments of any of the aspects described herein (e.g., any one of formulas (I), (II), (III), (ill-1), (ill-2), (ill-3), (IV), (IV-1), (IV-2), or (IV-3)), Ri is preferably N N
Figure imgf000008_0001
.In some embodiments of any of the aspects described herein (e.g., any one of formulas (I) or (II)), R2 is OH, H, F, Cl, Br, NH2, or optionally substituted C1-C3 alkoxy (e.g., -OCH3, -OCH2CH3, or -OCH2CH2CH3). In preferred embodiments, R2 is OH or -OCH3.
In some embodiments of any of the aspects described herein (e.g., any one of formulas (I), (II), (III), (IV), (IV-1), (IV-2), or (IV-3)), R3 is an optionally substituted C3-C7 alkyl.
In some embodiments of any of the aspects described herein (e.g., any one of formulas (I), (II),
Figure imgf000008_0002
In some embodiments of any of the aspects described herein (e.g., any one of formulas (I), (II),
(III), (IV), (IV-1), (IV-2), or (IV-3)), R3 is
Figure imgf000008_0003
In some embodiments of any of the aspects described herein (e.g., any one of formulas (I), (II),
(III), (IV), (IV-1), (IV-2), or (IV-3)), R3 is optionally substituted C2-C7 heteroalkyl.
In some embodiments of any of the aspects described herein (e.g., any one of formulas (I), (II),
Figure imgf000009_0001
In some embodiments of any of the aspects described herein (e.g., any one of formulas (I), (II), (III), (IV), (IV-1), (IV-2), or (IV-3)), R3 is optionally substituted C3-C7 alkenyl.
In some embodiments of any of the aspects described herein (e.g., any one of formulas (I), (II),
Figure imgf000009_0002
In some embodiments of any of the aspects described herein (e.g., any one of formulas (I), (II), (III), (IV), (IV-1), (IV-2), or (IV-3)), R3 is optionally substituted phenyl.
In some embodiments of any of the aspects described herein (e.g., any one of formulas (I), (II), (III), (IV), (IV-1), (IV-2), or (IV-3)), R3 is
Figure imgf000009_0003
In some embodiments of any of the aspects described herein (e.g., any one of formulas (I), (II), (III), (IV), (IV-1), (IV-2), or (IV-3)), R3 is optionally substituted 5- to 7-membered heterocyclyl.
In some embodiments of any of the aspects described herein (e.g., any one of formulas (I), (II), (III), (IV), (IV-1), (IV-2), or (IV-3)), R3 is optionally substituted pyridine, optionally substituted piperidine, optionally substituted piperazine, optionally substituted pyrimidine, optionally substituted pyrazine, optionally substituted pyridazine, optionally substituted triazine, optionally substituted azepine, optionally substituted 1 ,4-diazepine, optionally substituted 2/-/-pyran, optionally substituted 4/-/-pyran, optionally substituted tetrahydropyran, optionally substituted thiepine, or optionally substituted 1 ,4-thiazepine.
In some embodiments of any of the aspects described herein (e.g., any one of formulas (I), (II), (III), (IV), (IV-1), (IV-2), or (IV-3)), R3 is
Figure imgf000009_0004
Figure imgf000010_0001
In some embodiments of any of the aspects described herein (e.g., any one of formulas (I), (II), (III), (IV), (IV-1), (IV-2), or (IV-3)), R3 is an optionally substituted amide of formula -C(0)NHR8, wherein Rs is H, optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2- C20 alkenyl, optionally substituted C1-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C1-C20 heteroalkynyl, optionally substituted C5-C15 aryl, optionally substituted C2-C15 heterocyclyl, optionally substituted C3-C20 cycloalkyl, or optionally substituted C1-C20 alkoxy. In some embodiments of any of the aspects described herein (e.g., any one of formulas (I), (II),
(III), (IV), (IV-1), (IV-2), or (IV-3)), R3 is
Figure imgf000010_0002
In some embodiments of any of the aspects described herein (e.g., a compound of formulas (I)), R4 is -CH3, -CH2OH, -CF3, -CH2F, or -CHF2. In preferred embodiments, R4 is -CH3. In some embodiments of any of the aspects described herein (e.g., a compound of formulas (I)),
R5 is -CH3, -CH2OH, -CF3, -CH2F, or -CHF2. In preferred embodiments, Rs is -CH3.
In some embodiments of any of the aspects described herein (e.g., a compound of formulas (I)), R6 is -CH3 or -CH2OH In preferred embodiments, R6 is -CH3.
In some embodiments, the compound is a compound of Table 1 (e.g., any one of compounds 1- 40):
Table 1
Figure imgf000010_0003
Figure imgf000011_0001
Figure imgf000012_0001
Figure imgf000013_0001
In another aspect, the invention provides a pharmaceutical composition including a compound of the invention (e.g., a compound of any one of formulas (l)-(IV) or any one of compounds 1-40), or a salt thereof, and a pharmaceutically acceptable excipient.
In another aspect, the invention provides a method of treating an inflammatory disease in a subject in need thereof. The method includes administering to the subject a pharmaceutical composition including a compound of the invention (e.g., a compound of any one of formulas (l)-(IV) or any one of compounds 1-40), or a salt thereof, and a pharmaceutically acceptable excipient, in an amount sufficient to treat the condition.
In some embodiments, the inflammatory disease is selected from the group consisting of scleroderma, dermatomyositis, systemic lupus erythematosus, acquired immune deficiency syndrome (AIDS), multiple sclerosis, rheumatoid arthritis, psoriasis, diabetes, cancer, asthma, atopic dermatitis, an autoimmune thyroid disorders, ulcerative colitis, Crohn’s disease, stroke, ischemia, a neurodegenerative disease, amyotrophic lateral sclerosis (ALS), chronic traumatic encephalopathy (CTE), chronic inflammatory demyelinating polyneuropathy, an autoimmune inner ear disease, uveitis, iritis, and peritonitis. In some embodiments, the inflammatory disease is scleroderma (e.g., systemic sclerosis, localized scleroderma, or sine scleroderma). In some embodiments, the inflammatory disease is dermatomyositis.
In another aspect, the invention provides a method of treating a fibrotic disease in a subject in need thereof. The method includes administering to the subject the pharmaceutical composition including a compound of the invention (e.g., a compound of any one of formulas (l)-(IV) or any one of compounds 1-40), or a salt thereof, and a pharmaceutically acceptable excipient, in an amount sufficient to treat the condition.
In some embodiments, the fibrotic disease is selected from the group consisting of scleroderma, cystic fibrosis, liver cirrhosis, interstitial pulmonary fibrosis, idiopathic pulmonary fibrosis, Dupuytren’s contracture, keloids, chronic kidney disease, chronic graft rejection, scarring, wound healing, postoperative adhesions, reactive fibrosis, polymyositis, ANCA vasculitis, Behcet's disease, anti-phospholipid syndrome, relapsing polychondritis, Familial Mediterranean Fever, giant cell arteritis, Graves ophthalmopathy, discoid lupus, pemphigus, bullous pemphigoid, hydradenitis suppuritiva, sarcoidosis, bronchiolitis obliterans, primary sclerosing cholangitis, primary biliary cirrhosis, and organ fibrosis. In some embodiments, the fibrotic disease is scleroderma (e.g., systemic sclerosis, localized scleroderma, or sine scleroderma). In some embodiments, the fibrotic disease is organ fibrosis (e.g., dermal fibrosis, lung fibrosis, liver fibrosis, kidney fibrosis, or heart fibrosis).
In some embodiments, the compound has increased affinity for the CB2 receptor compared to affinity for the CBi receptor. In some embodiments, the compound has 10%, 20%, 30% 40%, 50%, 60% 70%, 80%, 90%, 100%, 200%, 300%, 400%, 500%, 600%, 700%, 800%, 900%, or 1000% or more greater affinity for the CB2 receptor compared to the CBi receptor. In some embodiments, the compound has 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 11-fold, 12-fold, 13-fold, 14-fold, 15- fold, 16-fold, 17-fold, 18-fold, 19-fold, 20-fold, 25-fold, 30-fold, 40-fold, or 50-fold or more greater affinity for the CB2 receptor compared to the CBi receptor. Receptor selectivity may be determined by receptor binding or by functional assay (e.g., cAMP or b-arrestin), as described here, for example in Examples 4-6 In some embodiments, the compound has greater CB2 receptor selectivity compared to the CB2 receptor selectivity of ajulemic acid.
Definitions
To facilitate the understanding of this invention, a number of terms are defined below. Terms defined herein have meanings as commonly understood by a person of ordinary skill in the areas relevant to the invention. Terms such as "a", "an," and "the" are not intended to refer to only a singular entity, but include the general class of which a specific example may be used for illustration. The terminology herein is used to describe specific embodiments of the invention, but their usage does not limit the invention, except as outlined in the claims.
As used herein, the term “about” refers to a value that is within 10% above or below the value being described.
As used herein, any values provided in a range of values include both the upper and lower bounds, and any values contained within the upper and lower bounds.
As used herein, the term "treat" or "treatment" includes administration of a compound to a subject, e.g., by any route, e.g., orally, topically, by inhalation, by ex-vivo contact with one or more cells of the subject. The compound can be administered alone or in combination with one or more additional compounds. Treatments may be sequential, with the present compound being administered before or after the administration of other agents. Alternatively, compounds may be administered concurrently.
The subject, e.g., a patient, can be one having a disorder (e.g., a disease or condition described herein), a symptom of a disorder, or a predisposition toward a disorder. Treatment is not limited to curing or complete healing, but can result in one or more of alleviating, relieving, altering, partially remedying, ameliorating, improving or affecting the disorder, reducing one or more symptoms of the disorder or the predisposition toward the disorder. In an embodiment the treatment (at least partially) alleviates or relieves symptoms related to a fibrotic disease. In an embodiment the treatment (at least partially) alleviates or relieves symptoms related to an inflammatory disease. In one embodiment, the treatment reduces at least one symptom of the disorder or delays onset of at least one symptom of the disorder.
The effect is beyond what is seen in the absence of treatment.
The terms “therapeutically effective amount” or “amount sufficient to treat” as used interchangeably herein, refer to an amount, e.g., pharmaceutical dose, effective in inducing a desired effect in a subject or in treating a subject having a condition or disorder described herein (e.g., a fibrotic disease of an inflammatory disease). It is also to be understood herein that a “therapeutically effective amount” may be interpreted as an amount giving a desired therapeutic and/or preventative effect, taken in one or more doses or in any dosage or route, and/or taken alone or in combination with other therapeutic agents. The term “subject,” as used herein, can be a human, non-human primate, or other mammal, such as but not limited to dog, cat, horse, cow, pig, turkey, goat, fish, monkey, chicken, rat, mouse, and sheep.
The term “pharmaceutical composition” refers to the combination of an active agent with an excipient, inert or active, making the composition especially suitable for diagnostic or therapeutic use in vivo or ex vivo. A “pharmaceutically acceptable excipient,” after being administered to or upon a subject, does not cause undesirable physiological effects. The excipient in the pharmaceutical composition must be “acceptable” also in the sense that it is compatible with the active ingredient. The excipient may also be capable of stabilizing the active ingredient. One or more solubilizing agents can be utilized as pharmaceutical excipients for delivery of an active compound. Examples of pharmaceutically acceptable excipients include, but are not limited to, biocompatible vehicles, adjuvants, additives, and diluents to achieve a composition usable as a dosage form. Examples of other excipients include colloidal silicon oxide, magnesium stearate, cellulose, and sodium lauryl sulfate.
As used herein, the term "carrier" refers to a diluent, adjuvant, excipient, or vehicle with which the active compound is administered. Such pharmaceutical vehicles can be liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. The pharmaceutical vehicles can be saline, gum acacia, gelatin, starch paste, talc, keratin, colloidal silica, urea, and the like. In addition, auxiliary, stabilizing, thickening, lubricating and coloring agents can be used. When administered to a subject, the pharmaceutically acceptable vehicles are preferably sterile. Water can be the vehicle when the active compound is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid vehicles, particularly for injectable solutions. Suitable pharmaceutical vehicles also include excipients such as starch, glucose, lactose, sucrose, gelatin, sodium stearate, glycerol monostearate, talc, sodium chloride, glycerol, propylene glycol, water, and ethanol. The present compositions, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
As used herein, the term “pharmaceutically acceptable salt” represents a salt of a compound of the invention (e.g., a compound of any one of formulas (l)-(IV) or any one of compounds 1-40) that is within the scope of sound medical judgment, suitable for use in methods described herein without undue toxicity, irritation, and/or allergic response. Pharmaceutically acceptable salts are well known in the art.
For example, pharmaceutically acceptable salts are described in: Handbook of Pharmaceutical Salts: Properties, Selection, and Use (Eds. P.H. Stahl and C.G. Wermuth), Wiley-VCH, 2008. The salts can be prepared in situ during the final isolation and purification of a compound described herein or separately by reacting the free base group with a suitable organic acid.
The terms “alkyl,” “alkenyl,” and “alkynyl,” as used herein, include straight-chain and branched- chain monovalent substituents, as well as combinations of these, containing only C and H when unsubstituted. When the alkyl group includes at least one carbon-carbon double bond or carbon-carbon triple bond, the alkyl group can be referred to as an “alkenyl” or “alkynyl” group respectively. The monovalency of an alkyl, alkenyl, or alkynyl group does not include the optional substituents on the alkyl, alkenyl, or alkynyl group. For example, if an alkyl, alkenyl, or alkynyl group is attached to a compound, monovalency of the alkyl, alkenyl, or alkynyl group refers to its attachment to the compound and does not include any additional substituents that may be present on the alkyl, alkenyl, or alkynyl group. In some embodiments, the alkyl group may contain, e.g., 1-20. 1-18, 1-16, 1-14, 1-12, 1-10, 1-8, 1-6, 1-4, or 1-2 carbon atoms (e.g., C1-C20, C1-C18, C1-C16, C1-C14, C1-C12, C1-C10, C1-C8, C1-C6, C1-C4, or C1- C2). In some embodiments, the alkenyl or alkynyl group may contain, e.g., 2-20, 2-18, 2-16, 2-14, 2-12, 2-10, 2-8, 2-6, or 2-4 carbon atoms (e.g., C2-C20, C2-C18, C2 C16, C2-C14, C2-C12, C2-C10, C2-C8, C2-C6, or C2-C4). Examples include, but are not limited to, methyl, ethyl, isobutyl, sec-butyl, tert-butyl, 2- propenyl, and 3-butynyl.
The terms “heteroalkyl,” “heteroalkenyl,” and “heteroalkynyl,” as used herein, include alkyl, alkenyl, or alkynyl groups, as defined above, but which include one or more heteroatoms (e.g., oxygen, nitrogen, and/or sulfur atoms) in the alkyl, alkenyl, or alkynyl main chain. In some embodiments, the heteroalkyl, heteroalkenyl, or heteroalkynyl group may contain, e.g., 1-20. 1-18, 1-16, 1-14, 1-12, 1-10, 1- 8, 1-6, 1-4, or 1-2 carbon atoms (e.g., C1-C20, C1-C18, C1-C16, C1-C14, C1-C12, C1-C10, C1-C8, C1- C6, C1-C4, or C1-C2) and one or more (e.g., one, two, three, four, or five) heteroatoms atoms.
The term “alkoxy,” as used herein, represents a chemical substituent of formula -OR, where R is a C1-20 alkyl group (e.g., Ci-6 or CMO alkyl), unless otherwise specified. Exemplary alkoxy groups include methoxy, ethoxy, propoxy (e.g., n-propoxy and isopropoxy), t-butoxy, and the like. In some embodiments, the alkyl group can be further substituted with 1 , 2, 3, 4, or more substituent groups as defined herein (e.g., hydroxy or alkoxy).
The term “aryl,” as used herein, refers to any monocyclic or fused ring bicyclic or tricyclic system which has the characteristics of aromaticity in terms of electron distribution throughout the ring system, e.g., phenyl, naphthyl, or phenanthrene. In some embodiments, a ring system contains 5-15 ring member atoms or 5-10 ring member atoms. An aryl group may have, e.g., five to fifteen carbons (e.g., a C5-C6, C5-C7, C5-C8, C5-C9, C5-C10, C5-C11 , C5-C12, C5-C13, C5-C14, or C5-C15 aryl). The term “heteroaryl” also refers to such monocyclic or fused bicyclic ring systems containing one or more, e.g., 1 - 4, 1-3, 1 , 2, 3, or 4, heteroatoms selected from O, S and N. A heteroaryl group may have, e.g., two to fifteen carbons (e.g., a C2-C3, C2-C4, C2-C5, C2-C6, C2-C7, C2-C8, C2-C9. C2-C10, C2-C11 , C2-C12, C2-C13, C2-C14, or C2-C15 heteroaryl). The inclusion of a heteroatom permits inclusion of 5 membered rings to be considered aromatic as well as 6 membered rings. Thus, typical heteroaryl systems include, e.g., pyridyl, pyrimidyl, indolyl, benzimidazolyl, benzotriazolyl, isoquinolyl, quinolyl, benzothiazolyl, benzofuranyl, thienyl, furyl, pyrrolyl, thiazolyl, oxazolyl, isoxazolyl, benzoxazolyl, benzoisoxazolyl, and imidazolyl. Because tautomers are possible, a group such as phthalimido is also considered heteroaryl.
In some embodiments, the aryl or heteroaryl group is a 5- or 6-membered aromatic ring system optionally containing 1-2 nitrogen atoms. In some embodiments, the aryl or heteroaryl group is an optionally substituted phenyl, pyridyl, indolyl, pyrimidyl, pyridazinyl, benzothiazolyl, benzimidazolyl, pyrazolyl, imidazolyl, isoxazolyl, thiazolyl, or imidazopyridinyl. In some embodiments, the aryl group is phenyl. In some embodiments, an aryl group may be optionally substituted with a substituent such an aryl substituent, e.g., biphenyl.
The term “heterocyclyl,” as used herein represents a ring have 2 or more carbon atoms and at least one heteroatom. For example, a heterocyclyl ring may have, e.g., two to fifteen carbons ring atoms (e.g., a C2-C3, C2-C4, C2-C5, C2-C6, C2-C7, C2-C8, C2-C9. C2-C10, C2-C11 , C2-C12, C2-C13, C2- C14, or C2-C15 heterocyclyl) and one or more (e.g., one, two, three, four, or five) ring heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur. Heterocyclyl groups include both non-aromatic and aromatic rings (e.g., includes heteroaryl groups, as previously defined). In preferred embodiments of the invention, a heterocyclyl group is a 3- to 8-membered ring, a 3- to 6- membered ring, a 4- to 6-membered ring, most preferably a 5-membered ring or a 6-membered ring. Heterocyclyls include aromatic and non-aromatic rings. Exemplary 5-membered heterocyclyl groups may have zero to two double bonds, and exemplary 6-membered heterocyclyl groups may have zero to three double bonds. Exemplary 5-membered heterocyclyl groups include, for example, optionally substituted pyrrole, optionally substituted pyrazole, optionally substituted isoxazole, optionally substituted pyrrolidine, optionally substituted imidazole, optionally substituted thiazole, optionally substituted thiophene, optionally substituted thiolane, optionally substituted furan, optionally substituted tetrahydrofuran, optionally substituted diazole, optionally substituted triazole, optionally substituted tetrazole, optionally substituted oxazole, optionally substituted 1 ,3,4-oxadiazole, optionally substituted 1 ,3,4-thiadiazole, optionally substituted 1 ,2,3,4-oxatriazole, and optionally substituted 1 ,2,3,4-thiatriazole. Exemplary 6- membered heterocyclyl groups include, for example, optionally substituted pyridine, optionally substituted piperidine, optionally substituted piperazine, optionally substituted pyrimidine, optionally substituted pyrazine, optionally substituted pyridazine, optionally substituted triazine, optionally substituted 2/-/-pyran, optionally substituted 4/-/-pyran, and optionally substituted tetrahydropyran. Exemplary 7-membered heterocyclyl groups include, for example, optionally substituted azepine, optionally substituted 1 ,4- diazepine, optionally substituted thiepine, and optionally substituted 1 ,4-thiazepine.
The term “cycloalkyl,” as used herein, represents a monovalent saturated or unsaturated nonaromatic cyclic alkyl group. A cycloalkyl may have, e.g., three to twenty carbons (e.g., a C3-C7, C3-C8, C3-C9, C3-C10, C3-C11 , C3-C12, C3-C14, C3-C16, C3-C18, or C3-C20 cycloalkyl). Examples of cycloalkyls include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl. When the cycloalkyl group includes at least one carbon-carbon double bond, the cycloalkyl group can be referred to as a “cycloalkenyl” group. A cycloalkenyl may have, e.g., four to twenty carbons (e.g., a C4- C7, C4-C8, C4-C9, C4-C10, C4-C11 , C4-C12, C4-C14, C4-C16, C4-C18, or C4-C20 cycloalkenyl). Exemplary cycloalkenyl groups include, but are not limited to, cyclopentenyl, cyclohexenyl, and cycloheptenyl. When the cycloalkyl group includes at least one carbon-carbon triple bond, the cycloalkyl group can be referred to as a “cycloalkynyl” group. A cycloalkynyl may have, e.g., eight to twenty carbons (e.g., a C8-C9, C8-C10, C8-C11 , C8-C12, C8-C14, C8-C16, C8-C18, or C8-C20 cycloalkynyl). The term “cycloalkyl” also includes a cyclic compound having a bridged multicyclic structure in which one or more carbons bridges two non-adjacent members of a monocyclic ring, e.g., bicyclo[2.2.1 .Jheptyl and adamantyl. The term “cycloalkyl” also includes bicyclic, tricyclic, and tetracyclic fused ring structures, e.g., decalin and spiro-cyclic compounds.
The term “alkaryl,” refers to an aryl group that is connected to an alkylene, alkenylene, or alkynylene group. In general, if a compound is attached to an alkaryl group, the alkylene, alkenylene, or alkynylene portion of the alkaryl is attached to the compound. In some embodiments, an alkaryl is C6- C35 alkaryl (e.g., C6-C16, C6-C14, C6-C12, C6-C10, C6-C9, C6-C8, C7, or C6 alkaryl), in which the number of carbons indicates the total number of carbons in both the aryl portion and the alkylene, alkenylene, or alkynylene portion of the alkaryl. Examples of alkaryls include, but are not limited to, (C1 - C8)alkylene(C6-C12)aryl, (C2-C8)alkenylene(C6-C12)aryl, or (C2-C8)alkynylene(C6-C12)aryl. In some embodiments, an alkaryl is benzyl or phenethyl. In a heteroalkaryl, one or more heteroatoms selected from N, O, and S may be present in the alkylene, alkenylene, or alkynylene portion of the alkaryl group and/or may be present in the aryl portion of the alkaryl group. In an optionally substituted alkaryl, the substituent may be present on the alkylene, alkenylene, or alkynylene portion of the alkaryl group and/or may be present on the aryl portion of the alkaryl group.
The term “carboxyl,” as used herein, represents a -COOH group. An optionally substituted carboxyl includes, for example, a -COOR group, wherein R is H or any substituent group described herein.
The term “amine,” as used herein, represents an -Nhh group. An optionally substituted amine includes, for example, a -NHR or a -NR1R2 group, wherein R, Ri, and R2 are each independently H or any substituent group described herein. In some embodiments, Ri and R2form cyclic ring (e.g., a 5- or 6- membered ring), such that -NR1R2 is an optionally substituted heterocyclyl or heteroaryl.
The term “amide,” as used herein, represents a -C(=0)NH2 group. An optionally substituted amide includes, for example, a -C(=0)NHR or a -C(=0)NRIR2 group, wherein R, Ri, and R2 are each independently H or any substituent group described herein.
The term “imino” as used herein, represents a -C(=NRI)R2 group. An optionally substituted imino includes, for example, a -C(=NRI)R2 group, wherein each of Ri and R2 are independently selected from H or any substituted group described herein.
The term “thioester,” as used herein, represents a -C(=0)SH group. An optionally substituted thioester includes, for example, a -C(=0)SR group, wherein R is H or any substituent group described herein.
The term “thioamide,” as used herein, represents a -C(=S)NH2 group. An optionally substituted thioamide includes, for example, a -C(=S)NHR or a -C(=S)NRIR2 group, wherein R, Ri, and R2 are each independently H or any substituent group described herein.
The term “sulfonamide,” as used herein, represents a -S(=0)2NH2 group. An optionally substituted sulfonamide includes, for example, a -S(=0)2NHR or a -S(=0)2NRIR2 group, wherein R, Ri, and R2 are each independently H or any substituent group described herein.
The term “sulfonyl,” as used herein, represents a -S(=0)2R group. An optionally substituted sulfonyl includes, for example, a S(=0)2R, wherein R is an H or any substituent group described herein.
The term “cyano,” as used herein, represents a -CN group.
The term “hydroxyl,” as used herein, represents an -OH group.
The term “oxo,” as used herein, refers to a substituent having the structure =0, where there is a double bond between an atom and an oxygen atom.
The term “optionally substituted,” as used herein, refers to having 0, 1 , or more substituents, such as 0-25, 0-20, 0-10 or 0-5 substituents. Substituents include, but are not limited to, alkyl, alkenyl, alkynyl, aryl, alkaryl, acyl, heteroaryl, heteroalkyl, heteroalkenyl, heteroalkynyl, heteroalkaryl, halogen, oxo, cyano, nitro, amino, alkamino, hydroxy, alkoxy, alkanoyl, carbonyl, carbamoyl, guanidinyl, ureido, amidinyl, any of the groups or moieties described above, and hetero versions of any of the groups or moieties described above. Substituents include, but are not limited to, F, Cl, Br, methyl, ethyl, propyl, butyl, phenyl, benzyl, OR, NR2, SR, SOR, SO2R, OCOR, NRCOR, NRCONR2, NRCOOR, OCONR2,
RCO, COOR, alkyl-OOCR, SO3R, CONR2, S02NR2, NRSO2NR2, CN, CF3, OCF3, SiR3, and NO2, wherein each R is, independently, H, alkyl, alkenyl, aryl, heteroalkyl, heteroalkenyl, or heteroaryl, and wherein two of the optional substituents on the same or adjacent atoms can be joined to form a fused, optionally substituted aromatic or nonaromatic, saturated or unsaturated ring which contains 3-8 members, or two of the optional substituents on the same atom can be joined to form an optionally substituted aromatic or nonaromatic, saturated or unsaturated ring which contains 3-8 members. An optionally substituted group or moiety refers to a group or moiety (e.g., any one of the groups or moieties described above) in which one of the atoms (e.g., a hydrogen atom) is optionally replaced with another substituent. For example, an optionally substituted alkyl may be an optionally substituted methyl, in which a hydrogen atom of the methyl group is replaced by, e.g., OH. As another example, a substituent on a heteroalkyl or its divalent counterpart, heteroalkylene, may replace a hydrogen on a carbon or a hydrogen on a heteroatom such as N.
Brief Description of the Figures
FIGs. 1A-1J are a series of graphs showing the biological activity of Compound 2 as determined by an inflammation bioassay in human Peripheral Blood Mononuclear Cells (PBMCs). Compound 2 was assayed for its effect on inflammatory cytokine release from lipopolysaccharides (LPS)-induced PBMCs, isolated from a human blood sample: MCP-1 (FIG. 1A), IL-1 b (FIG. 1 B), IL-8 (FIG. 1C), IL-6 (FIG. 1 D), IL- 12(p70) (FIG. 1 E), IL-17A (FIG. 1F), IL-4 (FIG. 1G), IL-31 (FIG. 1H), IL-10 (FIG. 11), and TNFa (FIG. 1J).. The assay was performed as described in Example 17.
FIGs. 2A-2D are a series of graphs showing biological activity of Compound 2 as determined by an T cell proliferation bioassay in human T cells isolated from PBMCs. The results are presented as percent CD4+ and CD8+ proliferation from three donors: donor 1 (FIG. 2A), donor 2 (FIG. 2B), and donor 3 (FIG. 3C). Representative results of cyclosporin A from 2 donors are also presented (FIG. 2D). The assay was performed as described in Example 19.
FIG. 3 is a set of graphs showing the biological activity of Compound 2 as determined by a fibroblast-to-myofibroblast transition (FMT) assay in human lung fibroblast from healthy donors. Compound 2 of the invention was assayed for its effect on fibroblast activation. The assay was performed as described in Example 20.
Detailed Description of the Invention
The invention relates to cannabinoid compounds, pharmaceutical compositions including one or more cannabinoid compounds, and the use of pharmaceutical compositions including one or more cannabinoid compounds for the treatment of a disease or condition (e.g., a fibrotic disease or an inflammatory disease) in a subject in need thereof.
Compounds
The disclosure provides compounds (e.g., a cannabinoid compound, a compound described by any one of formulas (l)-(IV), or any one of compounds 1-40) useful for the treatment of disease (e.g., a fibrotic disease or an inflammatory disease).
In particular, the invention features compounds sharing structural features with (6aR,10aR)-1- hydroxy-6,6-dimethyl-3-(2-methyl-2-octanyl)-6a,7,10,10a-tetrahydro-6H-benzo[c]chromene-9-carboxylic acid (ajulemic acid):
Figure imgf000020_0001
In some embodiments, the invention features compounds which are modulators (e.g., agonists, inverse agonists, or antagonists) of the CB2 receptor. In preferred embodiments of the invention, the invention features compounds that have increased affinity for the CB2 receptor (e.g., increased affinity for the CB2 receptor compared to ajulemic acid), increased selectivity for the CB2 receptor (e.g., increased selectivity for the CB2 receptor over the CBi receptor compared to ajulemic acid), or both increased affinity and increased selectivity for the CB2 receptor.
In some embodiments, the invention features compounds with an increased safety or efficacy profile in the treatment of a disease or condition (e.g., a fibrotic disease or an inflammatory disease), as compared to other cannabinoids, such as ajulemic acid. In some embodiments, administration of a compound of the invention to a subject (e.g., a subject having a disease or condition described herein) results in a decrease in treatment-associated adverse events relative to treatment with one or more other cannabinoids (e.g., treatment with an equivalent dose and method of administration of ajulemic acid). In some embodiments, administration of a compound of the invention to a subject (e.g., a subject having a disease or condition described herein) results in a decrease in CBi-associated adverse events relative to treatment with one or more other cannabinoids (e.g., ajulemic acid). In some embodiments, administration of a compound of the invention to a subject (e.g., a subject having a disease or condition described herein) results in a decrease in the rate of occurrence, severity, or risk of one or more of the following adverse events: dizziness, dry mouth, disorientation, euphoria, headache, nausea, pallor, somnolence, vomiting, tremor, abnormal feeling, tachycardia, fatigue, feeling drunk, paresthesia, muscle spasms, muscle tightness, disturbance in attention, deja vu, altered mood, anorexia, and cardiovascular events such as orthostatic hypotension, or QTc prolongation. The reduction in adverse events may be a reduction of 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or more in the occurrence or severity of any one of the above-described adverse events (e.g., compared to a subject or subjects treated with an equivalent dose and method of administration of another cannabinoid, such as ajulemic acid).
In some embodiments, the invention features compounds having improved pharmacokinetic properties or improved stability (e.g., improved pharmacokinetic properties or improved stability as compared to ajulemic acid).
In some embodiments, a compound of the invention is described by any one of formulas (l)-(IV) (e.g., any one of formulas (I), (II), (III), (MI-1), (III-2), (ill-3), (IV), (IV-1), (IV-2), or (IV-3).
In some embodiments, the compound of the invention is a compound of Table 1 (e.g., a compound selected from any one of compounds 1-40 of Table 1). Pharmaceutical compositions
Compounds of the invention (e.g., a cannabinoid compound, such as a compound described by any one of formulas (l)-(IV), or any one of compounds 1-40) prepared by any of the methods described herein may be formulated as a pharmaceutical composition for the treatment of disease. As described above, the pharmaceutical compositions of the invention additionally include a pharmaceutically acceptable excipient, which, as used herein, includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, and lubricants, as suited to the particular dosage form desired. Remington’s Pharmaceutical Sciences, Sixteenth Edition, E. W. Martin (Mack Publishing Co., Easton,
Pa., 1980) discloses various excipients used in formulating pharmaceutical compositions and known techniques for the preparation thereof. Some examples of materials which can serve as pharmaceutically acceptable excipients include, but are not limited to, sugars such as lactose, glucose, mannitol, and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatine; talc; excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil; safflower oil, sesame oil; olive oil; corn oil and soybean oil; glycols; such as propylene glycol; esters such as ethyl oleate and ethyl laurate; agar; natural and synthetic phospholipids, such as soybean and egg yolk phosphatides, lecithin, hydrogenated soy lecithin, dimyristoyl lecithin, dipalmitoyl lecithin, distearoyl lecithin, dioleoyl lecithin, hydroxylated lecithin, lysophosphatidylcholine, cardiolipin, sphingomyelin, phosphatidylcholine, phosphatidyl ethanolamine, diastearoyl phosphatidylethanolamine (DSPE) and its pegylated esters, such as DSPE-PEG750 and, DSPE-PEG2000, phosphatidic acid, phosphatidyl glycerol and phosphatidyl serine. Commercial grades of lecithin which are preferred include those which are available under the trade name Phosal® or Phospholipon® and include Phosal 53 MCT, Phosal 50 PG, Phosal 75 SA, Phospholipon 90H, Phospholipon 90G and Phospholipon 90 NG; soy-phosphatidylcholine (SoyPC) and DSPE-PEG2000 are particularly preferred; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer’s solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator.
Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention also include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS), self- microemulsifying drug delivery systems (SMEDDS), such as d-E-tocopherol polyethylene-glycol 1000 succinate; surfactants used in pharmaceutical compositions such as Tweens or other similar polymeric delivery matrices; serum proteins such as human serum albumin; buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts; or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, magnesium trisilicate, polyvinyl pyrrolidone, cellulose- based substances, polyethylene glycol, sodium carboxmethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat. Cyclodextrins such as alpha, beta and .gamma. -cyclodextrin, or chemically modified derivatives such as hydroxyalkylcyclodextrins, including 2- and 3-hydroxypropyl-beta cyclodextrins, or other solubilized derivatives may also be advantageously used to enhance delivery of compounds of the formulae described herein that can be used in the methods of the invention for preventing and/or treating fibrotic conditions. In certain embodiments, unit dosage formulations are compounded for immediate release, though unit dosage formulations compounded for delayed or prolonged release of one or both agents are also disclosed.
Viscosity modifiers that may be used in pharmaceutical compositions of the present invention include, but are not limited to, caprylic/capric triglyceride (Migliol 810), isopropyl myristate (IPM), ethyl oleate, triethyl citrate, dimethyl phthalate, benzyl benzoate and various grades of polyethylene oxide.
High viscosity liquid carriers used in sustained release pharmaceutical compositions include, but are not limited to, sucrose acetate isobutyrate (SAIB) and cellulose acetate butyrate (CAB 381-20).
Non-limiting examples of binding agents that may be used in pharmaceutical compositions of the present invention include but are not limited to hydroxyalkyl cellulose, a hydroxyalkylalkyl cellulose, hydroxypropyl methyl cellulose, or a polyvinylpyrrolidone.
Non-limiting examples of osmotic agents that may be used in pharmaceutical compositions of the present invention include, but are not limited to, sorbitol, mannitol, sodium chloride, or other salts. Nonlimiting examples of biocompatible polymers employed in the contemplated pharmaceutical compositions include but are not limited to poly(hydroxy acids), polyanhydrides, polyorthoesters, polyamides, polycarbonates, polyalkylenes, polyalkylene glycols, polyalkylene oxides, polyalkylene terephthalates, polyvinyl alcohols, polyvinyl ethers, polyvinyl esters, polyvinyl halides, polyvinylpyrrolidone, polysiloxanes, poly(vinyl alcohols), poly (vinyl acetate), polystyrene, polyurethanes and co-polymers thereof, synthetic celluloses, polyacrylic acids, poly(butyric acid), poly(valeric acid), and poly(lactide-co-caprolactone), ethylene vinyl acetate, copolymers and blends thereof.
Non-limiting examples of hygroscopic polymers that may be employed in the contemplated pharmaceutical compositions include, but are not limited to, polyethylene oxide (e.g., Polyox® with MWs from 4,000,000 to 10,000,000), cellulose, hydroxymethylcellulose, hydroxyethylcellulose, crosslinked polyacrylic acids, and xanthan gum.
Non-limiting examples of rate-controlling polymers the may be employed in the contemplated pharmaceutical compositions include but are not limited to polymeric acrylate, methacrylate lacquer or mixtures thereof, polymeric acrylate lacquer, methacrylate lacquer, an acrylic resin including a copolymer of acrylic and methacrylic acid esters, or an ammonium methacrylate lacquer with a plasticizer.
The above-described compositions, in any of the forms described herein, can be used for treating disease (e.g., fibrotic disease, inflammatory disease, or any other disease or condition described herein). An effective amount refers to the amount of an active compound/agent that is required to confer a therapeutic effect on a treated subject. Effective doses will vary, as recognized by those skilled in the art, depending on the types of diseases treated, route of administration, excipient usage, and the possibility of co-usage with other therapeutic treatment.
A pharmaceutical composition of this invention can be administered by any suitable route, e.g., parenterally, orally, nasally, rectally, topically, buccally, by ophthalmic administration, or by inhalation.
The term “parenteral” as used herein refers to subcutaneous, intracutaneous, intravenous, intramuscular, intraarticular, intraarterial, intrasynovial, intrasternal, intrathecal, intralesional, or intracranial injection, as well as any suitable infusion technique.
A sterile injectable composition can be a solution or suspension in a non-toxic parenterally acceptable diluent or solvent. Such solutions include, but are not limited to, 1 ,3-butanediol, mannitol, water, Ringer’s solution, and isotonic sodium chloride solution. In addition, fixed oils are conventionally employed as a solvent or suspending medium (e.g., synthetic mono- or diglycerides). Fatty acids, such as, but not limited to, oleic acid and its glyceride derivatives, are useful in the preparation of injectables, as are natural pharmaceutically acceptable oils, such as, but not limited to, olive oil or castor oil, or polyoxyethylated versions thereof. These oil solutions or suspensions also can contain a long chain alcohol diluent or dispersant such as, but not limited to, carboxymethyl cellulose, or similar dispersing agents. Other commonly used surfactants, such as, but not limited to, Tweens or Spans or other similar emulsifying agents or bioavailability enhancers, which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other pharmaceutical compositions also can be used for the purpose of formulation.
A composition for oral administration can be any orally acceptable dosage form including capsules, tablets, emulsions and aqueous suspensions, dispersions, and solutions. In some embodiments, the dosage form is an oral dosage form such as a pressed tablet, hard or soft gel capsule, enteric coated tablet, osmotic release capsule, or unique combination of excipients. In the case of tablets, commonly used excipients include, but are not limited to, lactose, mannitol, and corn starch. Lubricating agents, such as, but not limited to, magnesium stearate, also are typically added. For oral administration in a capsule form, useful diluents include, but are not limited to, lactose, mannitol, glucose, sucrose, corn starch, potato starch, or cellulose. In additional embodiments, the dosage form includes a capsule wherein the capsule contains a mixture of materials to provide a desired sustained release formulation. When aqueous suspensions or emulsions are administered orally, the active ingredient can be suspended or dissolved in an oily phase combined with emulsifying or suspending agents. If desired, certain sweetening, flavoring, or coloring agents can be added.
The pharmaceutical compositions can include a tablet coated with a semipermeable coating. In certain embodiments, the tablet includes two layers, a layer containing a compound of the invention and a second layer referred to as a "push" layer. The semi-permeable coating is used to allow a fluid (e.g., water) to enter the tablet and erode a layer or layers. In certain embodiments, this sustained release dosage form further includes a laser hole drilled in the center of the coated tablet. The compound containing layer may include a compound described herein, a disintegrant, a viscosity enhancing agent, a binding agent, and an osmotic agent. The push layer includes a disintegrant, a binding agent, an osmotic agent, and a viscosity enhancing agent. Non-limiting examples of materials that make up preferred semipermeable layers include, but are not limited to cellulosic polymers such as cellulose acetate, cellulose acylate, cellulose diacylate, cellulose triacylate, cellulose diacetate, cellulose triacetate or any mixtures thereof; ethylene vinyl acetate copolymers, polyethylene, copolymers of ethylene, polyolefins including ethylene oxide copolymers (e.g., Engage® Dupont Dow Elastomers), polyamides, cellulosic materials, polyurethanes, polyether blocked amides, and copolymers (e.g., PEBAX®, cellulosic acetate butyrate and polyvinyl acetate). Non-limiting examples of disintegrants that may be employed in the above sustained release pharmaceutical compositions include but are not limited to croscarmellose sodium, crospovidone, sodium alginate or similar excipients.
In further embodiments, the dosage form includes a tablet including a biocompatible matrix and a compound described herein. The dosage form may also include a hard-shell capsule containing biopolymer microspheres that contains the therapeutically active agent. The biocompatible matrix and biopolymer microspheres each contain pores for drug release and delivery. These pores are formed by mixing the biocompatible matrix of bio-polymer microsphere with a pore forming agent. Each biocompatible matrix or bio-polymer microsphere is made up of a biocompatible polymer or mixture of biocompatible polymers. The matrix and microspheres can be formed by dissolving the biocompatible polymer and active agent (compound described herein) in a solvent and adding a pore-forming agent (e.g., a volatile salt). Evaporation of the solvent and pore forming agent provides a matrix or microsphere containing the active compound. In additional embodiments, the dosage form includes a tablet, wherein the tablet contains a compound of the invention and one or more polymers and wherein the tablet can be prepared by compressing the compound and one or more polymers. In some embodiments, the one or more polymers may include a hygroscopic polymer formulated with a compound of the invention. Upon exposure to moisture, the tablet dissolves and swells. This swelling allows the sustained release dosage form to remain in the upper Gl tract. The swelling rate of the polymer mixture can be varied using different grades of polyethylene oxide.
Pharmaceutical compositions for topical administration according to the described invention can be formulated as solutions, ointments, creams, suspensions, lotions, powders, pastes, gels, sprays, aerosols, or oils. Alternatively, topical formulations can be in the form of patches or dressings impregnated with active ingredients), which can optionally include one or more excipients or diluents. In some preferred embodiments, the topical formulations include a material that would enhance absorption or penetration of the active agent(s) through the skin or other affected areas.
A topical composition contains a safe and effective amount of a dermatologically acceptable excipient suitable for application to the skin. A “cosmetically acceptable” or “dermatologically-acceptable” composition or component refers a composition or component that is suitable for use in contact with human skin without undue toxicity, incompatibility, instability, or allergic response. The excipient enables an active agent and optional component to be delivered to the skin at an appropriate concentration^).
The excipient thus can act as a diluent, dispersant, solvent, or the like to ensure that the active materials are applied to and distributed evenly over the selected target at an appropriate concentration. The excipient can be solid, semi-solid, or liquid. The excipient can be in the form of a lotion, a cream, or a gel, in particular one that has a sufficient thickness or yield point to prevent the active materials from sedimenting. The excipient can be inert or possess dermatological benefits. It also should be physically and chemically compatible with the active components described herein, and should not unduly impair stability, efficacy, or other use benefits associated with the composition.
The present compositions may be formulated for sustained release (e.g., over a 6 hour period, over a 12 hour period, over a 24 hour period, or over a 48 hour period). In some embodiments, the sustained release dosage form includes a tablet or a capsule including particle cores coated with a suspension of active agent and a binding agent which is subsequently coated with a polymer. The polymer may be a rate-controlling polymer. In general, the delivery rate of the rate-controlling polymer is determined by the rate at which the active agent is dissolved.
In another embodiment, the composition is formulated to provide extended release. For example, the agent is formulated with an enteric coating. In an alternative embodiment, the agent is formulated using a biphasic controlled release delivery system, thereby providing prolonged gastric residence. For example, in some embodiments, the delivery system includes (1) an inner solid particulate phase formed of substantially uniform granules containing a pharmaceutical having a high water solubility, and one or more hydrophilic polymers, one or more hydrophobic polymers and/or one or more hydrophobic materials such as one or more waxes, fatty alcohols and/or fatty acid esters, and (2) an outer solid continuous phase in which the above granules of inner solid particulate phase are embedded and dispersed throughout, the outer solid continuous phase including one or more hydrophobic polymers, one or more hydrophobic polymers and/or one or more hydrophobic materials such as one or more waxes, fatty alcohols and/or fatty acid esters, which may be compressed into tablets or filled into capsules. In some embodiments, the agent is incorporated into polymeric matrices included of hydrophilic polymers that swell upon imbibition of water to a size that is large enough to promote retention of the dosage form in the stomach during the fed mode.
The active compound in the formulation may be formulated as a combination of fast-acting and controlled release forms. For example, the active compound is formulated with a single release property. For example, it is present in a modified release form, e.g., a controlled release form.
The pharmaceutical composition can be administered alone or in combination with one or more additional compounds. Treatments may be sequential, with the present compound being administered before or after the administration of other agents. Alternatively, compounds may be administered concurrently. Exemplary additional agents include an analgesic agent such as an opiate, an antiinflammatory agent, or a natural agent such as a triglyceride containing unsaturated fatty acid, or isolated pure fatty acids such as eicosapentaenoic acid (EPA), dihomogamma linolenic acid (DGLA), docosahexaenoic acid (DHA) and others. In some embodiments, the therapeutic agents that can be used in the present methods are formulated in a single unit dose such that the agents are released from the dosage at different times.
The pharmaceutical composition can be administered at a dose determined by one of skill in the art (e.g., an effective therapeutic dose, for example, to treat a disorder described herein). For example, pharmaceutical composition including a compound of the present invention can be administered at a dose of 0.001-0.01 mg/kg, 0.01-0.5 mg/kg, 0.5-2 mg/kg, 2-5 mg/kg, 5-10 mg/kg, 10-20 mg/kg, 20-40 mg/kg, 40- 60 mg/kg, 60-80 mg/kg, 80-100 mg/kg, 100-200 mg/kg or200-500mg/kg. A unit of a pharmaceutical dosage form may include, for example, 0.001-0.01 mg, 0.01-0.5 mg, 0.5-2 mg, 2-5 mg, 5-10 mg, 10-20 mg, 20-40 mg, 40-60 mg, 60-80 mg, 80-100 mg, 100-200 mg or 200-500mg of a compound of the invention.
Methods of treatment
In some embodiments of the invention, any of the above-described compositions (e.g., compositions including a cannabinoid compound, a compound described by any one of formulas (l)-(IV), or any one of compounds 1-40 prepared according to the methods of the invention), including any of the above-described pharmaceutical compositions, may be administered to a subject (e.g., a mammal, such as a human, cat, dog, horse, cow, or pig) having a disease (e.g., a fibrotic disease or an inflammatory disease) in order to treat, prevent, or ameliorate the disease.
Inflammation
A therapeutically effective amount of any of the compositions described herein (e.g. a pharmaceutical composition comprising a compound described by any one of formulas (l)-(IV), or any one of compounds 1-40) may be used to treat or prevent inflammatory disease.
Inflammatory diseases include, for example, (e.g., systemic sclerosis, localized scleroderma, or sine scleroderma), dermatomyositis, systemic lupus erythematosus, acquired immune deficiency syndrome (AIDS), multiple sclerosis, rheumatoid arthritis, psoriasis, diabetes, cancer, asthma, atopic dermatitis, an autoimmune thyroid disorders, ulcerative colitis, Crohn’s disease, stroke, ischemia, a neurodegenerative disease, amyotrophic lateral sclerosis (ALS), chronic traumatic encephalopathy (CTE), chronic inflammatory demyelinating polyneuropathy, an autoimmune inner ear disease, uveitis, iritis, and peritonitis.
In some embodiments, inflammation can be assayed by measuring the chemotaxis and activation state of inflammatory cells. In some embodiments, inflammation can be measured by examining the production of specific inflammatory mediators such as interleukins, cytokines and eicosanoids. In some embodiments, in vivo inflammation is measured by swelling and edema of a localized tissue or migration of leukocytes. Inflammation may also be measured by organ function such as in the lung or kidneys and by the production of pro-inflammatory factors. Inflammation may also be assessed by other suitable methods. Other methods known to one skilled in the art may also be suitable methods for the assessment of inflammation and may be used to evaluate or score the response of the subject to treatment with one or more therapeutic agents of the invention (e.g., a cannabinoid compound, a compound described by any one of formulas (l)-(IV), or any one of compounds 1-40).
Fibrotic diseases
A therapeutically effective amount of any of the compositions described herein (e.g. a pharmaceutical composition comprising a compound described by any one of formulas (l)-(IV), or any one of compounds 1-40) may be used to treat or prevent inflammatory disease.
Fibrotic diseases include, for example, scleroderma (e.g., systemic sclerosis, localized scleroderma, or sine scleroderma), cystic fibrosis, liver cirrhosis, interstitial pulmonary fibrosis, idiopathic pulmonary fibrosis, Dupuytren’s contracture, keloids, chronic kidney disease, chronic graft rejection, scarring, wound healing, post-operative adhesions, reactive fibrosis, polymyositis, ANCA vasculitis, Behcet's disease, anti-phospholipid syndrome, relapsing polychondritis, Familial Mediterranean Fever, giant cell arteritis, Graves ophthalmopathy, discoid lupus, pemphigus, bullous pemphigoid, hydradenitis suppuritiva, sarcoidosis, bronchiolitis obliterans, primary sclerosing cholangitis, primary biliary cirrhosis, and organ fibrosis (e.g., dermal fibrosis, lung fibrosis, liver fibrosis, kidney fibrosis, or heart fibrosis).
Non-limiting examples of fibrosis include liver fibrosis, lung fibrosis (e.g., silicosis, asbestosis, idiopathic pulmonary fibrosis), oral fibrosis, endomyocardial fibrosis, retroperitoneal fibrosis, deltoid fibrosis, kidney fibrosis (including diabetic nephropathy), cystic fibrosis, and glomerulosclerosis. Liver fibrosis, for example, occurs as a part of the wound-healing response to chronic liver injury. Fibrosis can occur as a complication of haemochromatosis, Wilson's disease, alcoholism, schistosomiasis, viral hepatitis, bile duct obstruction, exposure to toxins, and metabolic disorders. Endomyocardial fibrosis is an idiopathic disorder that is characterized by the development of restrictive cardiomyopathy. In endomyocardial fibrosis, the underlying process produces patchy fibrosis of the endocardial surface of the heart, leading to reduced compliance and, ultimately, restrictive physiology as the endomyocardial surface becomes more generally involved. Oral submucous fibrosis is a chronic, debilitating disease of the oral cavity characterized by inflammation and progressive fibrosis of the submucosal tissues (lamina propria and deeper connective tissues). The buccal mucosa is the most commonly involved site, but any part of the oral cavity can be involved, even the pharynx. Retroperitoneal fibrosis is characterized by the development of extensive fibrosis throughout the retroperitoneum, typically centered over the anterior surface of the fourth and fifth lumbar vertebrae.
Treatment of fibrosis may be assessed by suitable methods known to one of skill in the art including the improvement, amelioration, or slowing the progression of one or more symptoms associated with the particular fibrotic disease being treated.
Scleroderma
Scleroderma is a disease of the connective tissue characterized by inflammation and fibrosis of the skin and internal organs. Scleroderma has a spectrum of manifestations and a variety of therapeutic implications. It includes localized scleroderma, systemic sclerosis, scleroderma-like disorders, and sine scleroderma. Systemic sclerosis can be diffuse or limited. Limited systemic sclerosis is also called CREST (calcinosis, Raynaud's esophageal dysfunction, sclerodactyly, telangiectasia). Systemic sclerosis includes: scleroderma lung disease, scleroderma renal crisis, cardiac manifestations, muscular weakness including fatigue or limited CREST, gastrointestinal dysmotility and spasm, and abnormalities in the central, peripheral and autonomic nervous system.
The major symptoms or manifestations of scleroderma, and in particular of systemic sclerosis, are inappropriate excessive collagen synthesis and deposition, endothelial dysfunction, vasospasm, collapse and obliteration of vessels by fibrosis. In terms of diagnosis, an important clinical parameter may be skin thickening proximal to the metacarpophalangeal joints. Raynaud's phenomenon may be a component of scleroderma. Raynaud’s may be diagnosed by color changes of the skin upon cold exposure. Ischemia and skin thickening may also be symptoms of Raynaud's disease.
A therapeutically effective amount of any of the compositions described herein (e.g. a cannabinoid compound, a compound described by any one of formulas (l)-(IV), or any one of compounds 1-40 prepared by any of the methods described herein) may be used to treat or prevent fibrosis. Fibrosis may be assessed by suitable methods known to one of skill in the art.
Examples
The following examples are put forth so as to provide those of ordinary skill in the art with a description of how the compositions and methods described herein may be used, made, and evaluated, and are intended to be purely exemplary of the invention and are not intended to limit the scope of what the inventors regard as their invention. General Methods
Liquid Chromatography-Mass Spectrometry (LC-MS) Methods
Method Tacc50-6 and Method Tacc50-6_AP_AMAC: The analyses were performed using an Agilent Technology 6120 LC/MSD quadrupole coupled to an Agilent 1260 Infinity series liquid chromatography (LC) system consisting of a binary pump with degasser, autosampler, thermostat column compartment and diode array detector. The mass spectrometer (MS) was operated with an atmospheric pressure electro-spray ionization (API-ES) source in positive ion mode. The capillary voltage was set to 3000 V, the fragmentor voltage to 70 V and the quadrupole temperature was maintained at 100°C. The drying gas flow and temperature values were 12.0 L/min and 350 °C, respectively. Nitrogen was used as the nebulisergas, at a pressure of 35 psig. Data acquisition was performed with Agilent Chemstation software.
Method Tacc50-6: Analyses were carried out on a Thermo Scientific Accucore aQ C18 column (50 mm long x 4.6 mm I.D.; 2.6 pm particles) at 35 °C, with a flow rate of 3 mL/min. A gradient elution was performed from 90% (Water + 0.1% Formic acid) / 10% Acetonitrile to 5% (Water + 0.1% Formic acid) / 95% Acetonitrile in 1.50 minutes; the resulting composition was held for 0.90 min; then the final mobile phase composition; from 10% (Water + 0.1 % Formic acid) / 90% Acetonitrile to 90% (Water + 0.1 % Formic acid) / 10% Acetonitrile in 0.10 minutes. The injection volume was 2 pL. MS acquisition range and DAD detector were set to 100-1000 m/z and 200-400 nm respectively.
Method Tacc50-6_AP_AMAC: Analyses were carried out on a Thermo Scientific Accucore aQ C18 column (50 mm long x 4.6 mm I.D.; 2.6 pm particle size) at 35 °C, with a flow rate of 3 mL/min. A gradient elution was performed from 50% (Water + 50 mM NH4OAc)/50% Acetonitrile to 5% (Water + 50 mM NH40Ac)/95% Acetonitrile in 1.5 min; the resulting composition was held for 0.9 min; from 5% (Water + 50 mM NH40Ac)/95% Acetonitrile to 95% (Water + 50 mM NH40Ac)/5% Acetonitrile in 0.2 min. Acquisition ranges were set to 190-400 nm for the UV-PDA detector and 100-1400 m/z for the MS detector.
Method VILLA and Method ZVilla_amonico_21 : The analyses were performed using an Agilent G1956A LC/MSD quadrupole coupled to an Agilent 1100 series liquid chromatography (LC) system consisting of a binary pump with degasser, autosampler, thermostat column compartment and diode array detector. The mass spectrometer (MS) was operated with an atmospheric pressure electro-spray ionization (API-ES) source in positive ion mode. The capillary voltage was set to 3000 V, the fragmentor voltage to 70 V and the quadrupole temperature was maintained at 100°C. The drying gas flow and temperature values were 12.0 L/min and 350 °C, respectively. Nitrogen was used as the nebuliser gas, at a pressure of 35 psig. Data acquisition was performed with Agilent Chemstation software.
Method VILLA: It is a long routine analysis for Quality Control of final compounds. Analyses were carried out on a YMC pack ODS-AQ C18 column (50 mm long x4.6 mm I.D.; 3 pm particle size) at 35 °C, with a flow rate of 2.6 mL/min. A gradient elution was performed from 95% (Water + 0.1 % Formic acid)/5% Acetonitrile to 5% (Water + 0.1% Formic acid)/95% Acetonitrile in 4.8 min; the resulting composition was held for 1 .0 min; from 5% (Water + 0.1 % formic acid)/95% Acetonitrile to 95% (Water + 0.1 % formic acid)/5% Acetonitrile in 0.2 min. Acquisition ranges were set to 190-400 nm for the UV-PDA detector and 100-1400 m/z for the MS detector.
Method ZVilla_amonico_21 : Analyses were carried out on a Phenomenex Kinetex C18 column (50 mm long x 2.1 mm; 2.6 pm particle size) at 35 °C, with a flow rate of 0.7 mL/min. A gradient elution was performed from 95% (Water, 50 mM NH40Ac)/5% Acetonitrile to 5% (Water, 50 mM NH40Ac)/95% Acetonitrile in 4.8 min; the resulting composition was held for 1 .0 min and to 95% (Water, 50 mM NH40Ac)/5% Acetonitrile in 0.2 min. Acquisition ranges were set to 190-400 nm for the UV-PDA detector and 100-1400 m/z for the MS detector.
Synthesis of ajulemic acid
Ajulemic acid (AJA) may be synthesized as known in the art. Preferably, ajulemic acid is an ultrapure formulation of ajulemic acid including more than 99% ajulemic acid and less than 1% highly-active CB-1 impurities, e.g., HU-210. Ajulemic acid may be synthesized as described in U.S. Patent Publication No. 2015/0141501 , which is incorporated herein by reference.
Example 1. Synthesis of Compound 1
Step a. (6aR, 10aR)-1 -hydroxy-6, 6-dimethyl-3-(2-methyloctan-2-yl)-6a, 7, 10, 10a-tetrahydro-6H- benzo[c]chromene-9-carboxamide
Figure imgf000029_0001
(1-Chloro-2-methyl-propenyl)-dimethyl-amine (1.81 g, 13.73 mmol, 1 ,1 eq) was added to a stirred solution of AJA (5.0 g, 12.48 mmol, 1 eq) in dry CH2Cl2 (50 ml_) under N2 atmosphere. The mixture reaction was stirred at rt for 2 hours. Then, a solution of Ammonia in dioxane [0.5M] (75 ml_, 37.45 mmol, 3.0 eq) was added dropwise and the mixture was stirred at rt overnight. The mixture was diluted with CH2CI2 and washed with an aqueous solution of HCI [1 M] and brine, the organic layer was dried over anhydrous MgS04, filtered and concentrated under reduced pressure. The crude product was purified by chromatography on silica gel (Heptane: EtOAc; from 100:0 to 80:20) to yield the desired product as a foam. (3.47 g; 69.3 %). 1H NMR (300 MHz, DMSO-de) d 9.20 (s, 1 H), 7.23 (s, 1H), 6.80 (s, 1 H), 6.54 (s,
1 H), 6.31 (s, 1 H), 6.12 (s, 1 H), 3.69 (d, J = 17.49 Hz, 1 H), 2.44 (m, 1 H), 2.28 (1 H, J = 15.81 Hz, 1 H), 1.95 (m, 1 H), 1.66 (m, 2H), 1.44 (bs, 2H), 1.31 (s, 3H), 1.16 (s, 5H), 1.14 (s, 7H), 1.01 (s, 5H), 0.81 (t, J = 6.15 Hz, 3H). LC-MS (ESI+): 400.3 (M+H+); R.T.: 1.602 min. (Method Tacc50-6_AP_AMAC). Step b. ( 6aR, 10aR)-9-cyano-6, 6-dimethyl-3-(2-methyloctan-2-yl)-6a, 7,10,10a-tetrahydro-6H- benzo[c]chromen-1-yl acetate
Figure imgf000030_0001
Acetic anhydride (1 .63 ml_, 17.30 mmol, 2.0 eq) was added to a stirred solution of the product from step a. (3.457 g, 8.65 mmol, 1eq) in pyridine (26 ml_). The mixture was stirred at rt for 1 hour. The solution was diluted with EtOAc and washed with an aqueous solution of HCI [1 M], and brine, the organic layer was dried over anhydrous MgS04, filtered and concentrated under reduced pressure. The oil was dissolved in pyridine (26 ml_) and cooled at 0°C, then trifluoromethanesulfonic anhydride (2.33 ml_, 13.84 mmol, 1 .6 eq) was added dropwise. The reaction mixture was allowed to warm to rt and stirred 1 h. The mixture was diluted with EtOAc and washed with an aqueous solution of HCI [1 M], and brine, the organic layer was dried over anhydrous MgS04, filtered and concentrated under reduced pressure. The product was purified by chromatography on silica gel (Heptane: EtOAc; from 100:0 to 90:10) to yield the desired product as an oil (1 .77 g; 48.3 %). 1H NMR (300 MHz, DMSO-de) d 6.82 (s, 1 H), 6.58 (d, J = 8.73 Hz, 2H), 2.97 (d, J = 16.92 Hz, 1 H), 2.56 (m, 1 H), 2.39 (d, J = 17.91 Hz, 1 H), 2.29 (s, 3H), 2.14 - 1.98 (m, 2H),
1.76 (t, J = 9.60 Hz, 1 H), 1.48 (m, 2H), 1.33 (s, 3H), 1.17 (s, 12H), 1.02 (s, 5H), 0.81 (t, J = 5.73 Hz, 3H). LC-MS (ESI+): 424.3 (M+H+); R.T.: 1.986 min. (Method Tacc50-6_AP_AMAC).
Step c. (6aR, 10aR)-6, 6-dimethyl-3-(2-methyloctan-2-yl)-9-( 1H-tetrazol-5-yl)-6a, 7, 10, 10a-tetrahydro-6H- benzo[c]chromen-1-ol (Compound 1)
Figure imgf000030_0002
Compound 1
Tributyltin azide (0.097 mg, 0.354 mmol, 1.5 eq) was added to a stirred solution of the product from step b. (0.1 g, 0.236 mmol, 1 eq) in dioxane (0.7 ml_) in a sealed tube under nitrogen atmosphere. The mixture was stirred at 90°C for 15 hours. The solution was cooled at room temperature, then an aqueous solution of NaOH [1 M] was added and the mixture was stirred at rt for 30 minutes. EtOAc was added and the aqueous layer was separated, diluted with water and acidified to pH 3 with HCI [6 M], the product was extracted with EtOAc. The combined organic layers were dried over anhydrous MgS04, filtered and concentrated under reduced pressure. The product was purified by chromatography on silica gel (Heptane: EtOAc; from 100:0 to 70:30) to yield Compound 1 as a white solid (7 mg, 7.0 %). 1H NMR (300 MHz, DMSO-de) d 9.31 (s, 1H), 6.79 (s, 1 H), 6.35 (s, 1 H), 6.15 (s, 1 H), 4.02 (d, J = 16.77 Hz, 1 H), 2.66 (t, J = 6.72 Hz, 1 H), 2.41 (s, 1 H), 2.06 (q, J = 11.97 Hz, 2H), 1.79 (t, J = 10.17 Hz, 1 H), 1.46 (bs, 2H), 1.34 (s, 3H), 1.15 (s, 12H), 1.06 (s, 5H), 0.81 (s, 3H). LC-MS (ESI+): 425.3 (M+H+); R.T.: 4.230 min. (Method Villa). Example 2. Synthesis of Compound 2
Figure imgf000031_0001
AJA
Pyridine (10.5 ml_, 131 .07 mmol, 2.1 eq) and n-heptane (225 ml_) were charged to round bottom flask, and the solution was heated at 50 °C, then AJA (25 g, 62.41 mmol, 1 eq) was added to the stirred solution, followed by acetic anhydride (10.62 ml_, 112.34 mmol, 1 .8 eq) added dropwise. The reaction was stirred at 50° C for 18 hours. Water (35 ml_) was added and the mixture was stirred at 50°C for 5 hours. The product was extracted with EtOAc (3x50 ml_), the combined organic layers were washed with brine and water, dried over anhydrous MgSC , filtered and concentrated under reduced pressure. The product was purified by chromatography on silica gel (Heptane: EtOAc; from 100:0 to 75:25) to yield the desired product as a white solid. (20.14 g; 72.9%). 1H NMR (300 MHz, DMSO-de) d 12.22 (s, 1 H), 6.89 (s, 1 H), 6.59 (s, 1 H), 6.55 (s, 1 H), 3.26 (d, J = 17.53 Hz, 1 H), 2.47 - 2.31 (m, 2H), 2.24 (s, 3H), 2.08 - 1 .95 (m, 1 H), 1 .84 - 1 .67 (m, 2H), 1 .52 - 1 .45 (m, 2H), 1 .34 (s, 3H), 1 .17 (s, 12H), 1 .03 (s, 5H), 0.81 (t, J = 6.84 Hz, 3H). LC-MS (ESI+): 443.3 (M+H+); R.T.: 1.563 min. (Method Tacc50-6_AP_AMAC).
Step b. ( 6aR, 10aR)-6, 6-dimethyl-3-(2-methyloctan-2-yl)-9-oxo-6a, 7, 8, 9, 10,10a-hexahydro-6H- benzo[c]chromen-1-yl acetate
Figure imgf000031_0002
Diphenylphosphorylazide (1 .07 ml_, 4.97 mmol, 1.1 eq) and triethylamine (0.92 ml_, 6.78 mmol, 1 .5 eq) were added to a stirred solution of the product from step a. (2.0 g, 4.52 mmol, 1 eq) in THF under nitrogen. The reaction mixture was stirred at 70 °C for 2 hours. The solvent was removed under reduced pressure. The yellow oil was diluted in dioxane (10 ml_), then 10 ml_ of citric acid aqueous solution (10%) was added and the resulted mixture was stirred at 60°C for 2 hours. The dioxane was distilled under reduced pressure, and the product was extracted with EtOAc (3x 15 ml_). The combined organic layers were dried over anhydrous MgS04, filtered and concentrated under reduced pressure. The product was purified by chromatography on silica gel (Heptane: EtOAc: from 100: 0 to 90:10) to yield the product as a light-yellow solid. (1.24 g; 66.2%). 1H NMR (300 MHz, CDCI3) d 6.69 (s, 1 H), 6.51 (s, 1 H), 3.27 (d, J = 14.34 Hz, 1H), 2.71 (t, J = 12.39 Hz, 1 H), 2.57 (d, J = 15.36 Hz, 1 H), 2.47 - 2-40 (m, 1 H), 2.32 (s, 3H), 2.26 (d, J = 14.43 Hz, 1 H), 2.19 - 2.13 (m, 2H), 1.95 (t, J = 11.34 Hz, 1 H), 1.57 (s, 1 H), 1.47 (s, 6H), 1.21 (s, 12H), 1.11 (s, 3H), 1.06 (bs, 2H), 0.84 (s, 3H). LC-MS (ESI+): 415.1 (M+H+); R.T.: 1.915 min. (Method T acc50-6_AP_AM AC) .
Figure imgf000032_0001
Lithium bis(trimethylsilyl)amide solution [1.0 M] in THF (2.47 mL, 2.47 mmol, 1.2 eq) was added to a stirred solution of the product from step b. (0.86 g, 2.06 mmol, 1 eq) in dry THF (7.0 mL) under nitrogen at -78° C. The mixture was stirred at -78°C for 1 h, then a solution of A/-Phenyl-bis(trifluomethansulfonimide) (1 .03 g, 2.89 mmol, 1 .4 eq) in dry THF (6.5 mL) was added dropwise. The mixture reaction was allowed to warm up to room temperature over a period of 4 hours and stirred at room temperature for 12 hours more. The solution was treated with an aqueous saturated solution of NH4CI and extracted with Et20. The combined organic layers were dried over anhydrous MgSC , filtered and concentrated under reduced pressure. The product was purified by chromatography on silica gel (Heptane: EtOAc) to yield the product as an oil. (0.52 g; 46.1%). 1H NMR (300 MHz, CDCI3) 66.69 (s, 1 H), 6.54 (s, 1H), 5.82 (s, 1 H), 3.21 (d, J = 17.55 Hz, 1H), 2.78 (m, 1H), 2.31 (s, 5H), 1.99 (t, J = 14.94 Hz, 1 H), 1.84 (dt, J = 11 .28 - 3.2 Hz, 1 H), 1.56 - 1.48 (m, 3H), 1.42 (s, 3H), 1.22 (s, 12H), 1.13 (s, 3H), 1.05 (bs, 2H), 0.84 (t, J = 5.61 Hz, 3H). LC- MS (ESI+): 547.2 (M+H+); R.T.: 2.172 min. (Method Tacc50-6_AP_AMAC).
Step d. ( 6aR, 10aR)-6, 6-dimethyl-3-(2-methyloctan-2-yl)-9-(4, 4, 5, 5-tetramethyl-1 ,3, 2-dioxaborolan-2-yl)- 6a, 7, 10, 10a-tetrahydro-6H-benzo[c]chromen-1-yl acetate
Figure imgf000032_0002
The product from step c. (0.5 g, 0.92 mmol, 1 eq), bis(pinacolato)diboron (0.28 g, 1.10 mmol, 1.2 eq), Pd(dppf) (38 mg, 0.046 mmol, 0.05 eq) and potassium acetate (0.27 g, 2.75 mmol, 3.0 eq) were dissolved in dioxane (12-5 mL) in a sealed tube under nitrogen atmosphere. The reaction mixture was stirred at 90°C for 14 hours. The mixture was diluted with EtOAc and washed with a saturated aqueous solution of NaHC03, the organic layer was dried over MgS04, filtered and concentrated under reduced pressure.
The product was purified by chromatography on silica gel (Heptane: EtOAc; from 100:0 to 95:5) to yield the product as a white solid. (0.198 g; 41.2 %). 1H NMR CDCI3 66.69 (m, 1 H), 6.52 (m, 2H), 3.12 (dd, J = 15.60 - 3.24 Hz, 1 H), 2.50 (dt, J = 10.92 - 3.84 Hz, 1 H), 2.29 (s, 3H), 2.25 (s, 2H), 1 .95 - 1 .79 (m, 3H), 1.52 - 1.46 (m, 2H), 1.40 (s, 1 H), 1.37 (s, 2H), 1.25 - 1.13 (m, 24H), 1.11 (s, 3H), 1.03 (bs, 2H), 0.84 (t, J = 6.90 Hz, 3H) LC-MS (ESI+): 525.4 (M+H+); R.T.: 2.343 min. (Method Tacc50-6_AP_AMAC).
Figure imgf000033_0001
Compound 2
The product from step d. (0.593 g, 1.13 mmol, 1 eq), 2-Bromo-1 /-/-imidazole (0.199 g, 1 .356 mmol, 1 .2 eq), Pd(Pph3)4 (66 mg, 0.057 mmol, 0.05 eq), xPhos (54 mg, 0.113 mmol, 0.1 eq) and potassium carbonate (0.625 g, 4.52 mmol, 4.0 eq) were dissolved in a mixture dioxane: water (4:1) (22 ml_) in a sealed tube under nitrogen atmosphere. The reaction mixture was stirred at 100°C for overnight. The mixture was diluted with EtOAc and washed with a saturated aqueous solution of NaHCC>3, the organic layer was dried over MgSC , filtered and concentrated under reduced pressure. The product was purified by chromatography on silica gel (Heptane: EtOAc; from 100:0 to 70:30) to yield Compound 2 as a white solid. (53 mg; 11.2 %). 1H NMR (300 MHz, DMSO-de) 11.96 (s, 1 H), 9.25 (s, 1 H), 6.95 (s, 2H), 6.39 (s, 1 H), 6.33 (s, 1 H), 6.14 (s, 1 H), 4.06 (d, J = 14.85 Hz, 1 H), 2.57 (dt, J = 13.20 - 4.08 Hz, 1 H), 2.37 - 2.27 (m, 1 H), 2.07 - 1 .79 (m, 3H), 1 .72 (dt, J = 11 .28 - 4.20, 1 H, 1 .49 - 1 .43 (m, 2H), 1 .33 (s, 3H), 1 .17 (bs, 5H), 1.15 (s, 7H), 1.04 (s, 5H), 0.81 (t, J = 6.78 Hz, 3H). LC-MS (ESI+): 423.3 (M+H+); R.T.: 3.155 min. (Method Villa).
Example 3. Synthesis of Compound 3
Figure imgf000033_0002
Compound 3
The starting material was synthesized as indicated in Example 2. The product from step d. of Example 2 (0.447 g, 0.852 mmol, 1 eq), 2-Bromo-1 ,3-oxazole (0.151 g, 1.022 mmol, 1.2 eq), Pd(Pphi3)4 (50 mg,
0.043 mmol, 0.05 eq), xPhos (41 mg, 0.085 mmol, 0.1 eq) and potassium carbonate (0.471 g, 3.41 mmol, 4.0 eq) were dissolved in a mixture dioxane: water (4:1) (17 ml_) in a sealed tube under nitrogen atmosphere. The reaction mixture was stirred at 100°C for overnight. The mixture was diluted with EtOAc and washed with a saturated aqueous solution of NaHC03, the organic layer was dried over MgS04, filtered and concentrated under reduced pressure. The product was purified by chromatography on silica gel (Heptane: EtOAc; from 100:0 to 50:50) to yield Compound 3 as a white solid. (16 mg; 4.5 %). 1H NMR (300 MHz, CDCb) 8 8.77 (s, 1H), 7.59 (s, 1 H), 7.16 (s, 1 H), 6.85 (bs, 1 H), 6.29 (d, J = 1.50 Hz, 1 H), 6.25 (d, J = 1.47, 1 H), 4.31 (dd, J = 17.79 - 2.25 Hz, 1 H), 3.64 (s, 0.5H), 2.80 (dt, J = 11 .16 - 4.47 Hz, 1 H), 2.47 (dt, J = 17.46 - 4.80, 1 H), 2.26 - 2.03 (m, 2H), 1 .91 (dt, J = 11 .46 - 4.41 Hz, 1 H), 1 .60 (bs, 1 H), 1 .41 (s, 5H), 1 .16 (s, 6H), 1 .11 (s, 8H), 1 .03 (bs, 1 H), 0.83 (t, J = 7.02 Hz, 3H) . LC-MS (ESI+): 424.3 (M+H+); R.T.: 5.107 min (Method Villa).
Example 4. Synthesis of 6aR,10aR)-6,6-dimethyl-3-(2-methyloctan-2-yl)-9-(pyrimidin-5-yl)- 6a,7,10,10a-tetrahydro-6H-benzo[c]chromen-1-ol (Compound 31)
Figure imgf000034_0001
The starting material was synthesized as indicated in Example 2. The starting material (0.095 g, 0.181 mmol, 1 eq), 5-bromopyrimidine (0.035 g, 0.217 mmol, 1.2 eq), Pd(PPh3)4 (10 mg, 0.009 mmol, 0.05 eq), XPhos (9 mg, 0.018 mmol, 0.1 eq), and potassium carbonate (0.106 g, 0.764 mmol, 4.0 eq) were dissolved in dioxane:water (4:1) (2 ml_) in a sealed tube under nitrogen atmosphere. The reaction mixture was stirred at 100 °C for overnight. The mixture was diluted with EtOAc and washed with a saturated aqueous solution of NaHCC>3, the organic layer was dried over MgSC , filtered and concentrated under reduced pressure. The product was purified by reverse phase HPLC (Gemini C18 column (100 x 30 mm, 5 pm), gradient: 32% [Aqueous phase] - 68% [organic phase] to 4% [Aqueous phase] - 96% [organic phase]. Aqueous phase: 65 mM NhUOAc; Organic phase: Acetonitrile:MeOH (1 :1), run time = 30 min). The desired fractions were collected and concentrated to the smallest possible volume. The residue was diluted with CH2CI2 and washed with brine. The organic layer was dried over anhydrous MgS04, filtered, and concentrated under reduced pressure to yield Compound 31 as a white solid (0.022 g, 28%). 1H NMR (300 MHz, CDCI3) d 9.15 (s, 1 H), 8.89 (s, 2H), 6.43 - 6.31 (m, 3H), 4.02 (dd, J = 16.6, 2.7 Hz, 1 H), 2.87 (td, J = 11.0, 4.3 Hz, 1 H), 2.55 - 2.41 (m, 1 H), 2.35 - 2.18 (m, 1 H), 2.18 - 2.01 (m, 1 H), 1.95 (td, J = 11.6, 4.1 Hz, 1 H), 1.54 - 1.46 (m, 2H), 1.45 (s, 3H), 1.21 (s, 6H), 1.24 - 1.12 (m, 6H), 1 .19 (s, 3H), 1 .12 - 1 .02 (m, 2H), 0.83 (t, J = 6.7 Hz, 3H); LC-MS (ESI+): 435.3 (M+H+); HPLC RT: 4.331 min (Method Villa). Example 5. Synthesis of (6a/?,10a/?)-6,6-dimethyl-3-(2-methyloctan-2-yl)-9-(pyrazin-2-yl)-6a,7,10,10a- tetrahydro-6H-benzo[c]chromen-1-ol (Compound 32)
Figure imgf000035_0001
The starting material was synthesized as indicated in Example 2. The starting material (0.228 g, 0.435 mmol, 1 eq), 2-bromopyrazine (0.047 ml_, 0.522 mmol, 1.2 eq), Pd(PPh3)4 (25 mg, 0.022 mmol, 0.05 eq), XPhos (21 mg, 0.044 mmol, 0.1 eq), and potassium carbonate (0.145 g, 1.052 mmol, 4.0 eq) were dissolved in dioxane:water (4:1) (2 ml_) in a sealed tube under nitrogen atmosphere. The reaction mixture was stirred at 100 °C for overnight. The mixture was diluted with EtOAc and washed with a saturated aqueous solution of NaHCC>3, the organic layer was dried over MgSC , filtered and concentrated under reduced pressure. The product was purified by reverse phase HPLC (Gemini C18 column (100 x 30 mm, 5 pm), gradient: 32% [Aqueous phase] - 68% [organic phase] to 4% [Aqueous phase] - 96% [organic phase]. Aqueous phase: 65mM NhUOAc; Organic phase: acetonitrile:MeOH (1 :1), run time = 30 min). The desired fractions were collected and concentrated to the smallest possible volume. The residue was diluted with CH2CI2 and washed with brine. The organic layer was dried over anhydrous MgS04, filtered and concentrated under reduced pressure to yield Compound 32 as a white solid (0.036 g, 19%). 1H NMR (300 MHz, CDCI3) d 8.69 (s, 1 H), 8.50 (s, 1 H), 8.35 (s, 1H), 6.89 - 6.82 (m, 1 H), 6.33 (d, J = 1 .7 Hz, 1 H), 6.22 (d, J = 1 .7 Hz, 1 H), 4.06 (dd, J = 17.1 , 2.0 Hz, 1 H), 2.79 (td, J = 11 .1 , 4.5 Hz, 1 H), 2.52 - 2.40 (m, 1H), 2.30 - 2.15 (m, 1 H), 2.12 - 2.00 (m, 1 H), 1.90 (td, J = 11.7, 4.4 Hz, 1 H), 1.45 - 1.39 (m, 2H), 1.38 (s, 3H), 1.20 - 1.06 (m, 6H), 1.13 (s, 6H), 1.11 (s, 3H), 1.04 - 0.94 (m, 2H),
0.77 (t, J = 6.9 Hz, 3H); LC-MS (ESI+): 435.3 (M+H+); HPLC RT: 4.334 min (Method Villa).
Example 6. Synthesis of (6a/?,10a/?)-6,6-dimethyl-3-(2-methyloctan-2-yl)-9-(pyrimidin-4-yl)- 6a,7,10,10a-tetrahydro-6H-benzo[c]chromen-1-ol (Compound 33)
Figure imgf000036_0001
The starting material was synthesized as indicated in Example 2. The starting material (0.100 g, 0.191 mmol, 1 eq), 2-bromopyrazine (0.045 ml_, 0.221 mmol, 1.2 eq), Pd(PPh3)4 (12 mg, 0.010 mmol, 0.05 eq), XPhos (9 mg, 0.019 mmol, 0.1 eq), and potassium carbonate (0.106 g, 0.764 mmol, 4.0 eq) were dissolved in dioxane:water (4:1) (2 ml_) in a sealed tube under nitrogen atmosphere. The reaction mixture was stirred at 100 °C for overnight. The mixture was diluted with EtOAc and washed with a saturated aqueous solution of NaHCC>3, the organic layer was dried over MgSC , filtered and concentrated under reduced pressure. The product was purified by normal phase chromatography on silica gel (heptane:EtOAc, from 100:0 to 40:60). The desired fractions were collected and concentrated in vacuo to yield a 7:3 mixture of (6aR,10aR)-6,6-dimethyl-3-(2-methyloctan-2-yl)-9-(pyrimidin-4-yl)- 6a,7,10,10a-tetrahydro-6H-benzo[c]chromen-1-yl acetate (0.041g, 46%) and Compound 33 (0.017 g, 20%). The product was used in the next step without further purification. LC-MS (ESI+): 477.3 (71 % area, M+H+), 435.3 (29% area, M+H+); HPLC RT: 2.453 min (71% area), 2.167 min. (29% area) (Method Villa).
Step b. (6aR, 10aR)-6, 6-dimethyl-3-(2-methyloctan-2-yl)-9-(pyrimidin-4-yl)-6a, 7, 10, 10a-tetrahydro-6H- benzo[c]chromen-1-ol (Compound 33)
Figure imgf000036_0002
To a solution of (6aR, 10aR)-6, 6-dimethyl-3-(2-methyloctan-2-yl)-9-(pyrimidin-4-yl)-6a, 7, 10, 10a-tetrahydro- 6H-benzo[c]chromen-1-yl acetate (0.058 g, 0.122 mmol, 1 eq) in methanol (1 ml_) at 0 °C was added dropwise a solution of sodium hydroxide in H2O (0.2 ml_). The reaction mixture was stirred at room temperature for overnight. The mixture was acidified to pH = 5 and the product was extracted with CH2CI2. The organic layers were dried over MgSC , filtered, and concentrated under reduced pressure. The product was purified by normal phase chromatography on silica gel (Heptane:EtOAc, from 100:0 to 40:60). The desired fractions were collected and concentrated in vacuo to yield Compound 33 as a white solid (0.040 g, 75%). 1H NMR (400 MHz, CDCI3) d 9.14 (s, 1 H), 8.65 (d, J = 4.8 Hz, 1 H), 7.40 (d, J = 5.0 Hz, 1 H), 7.15-7.09 (m, 1H), 6.39 (d, J= 1.7 Hz, 1H), 6.30 (d, J= 1.7 Hz, 1H), 4.05 (dd, J= 17.2, 2.1 Hz, 1 H), 2.82 (td, J= 11.2, 4.6 Hz, 1H), 2.57-2.47 (m, 1H), 2.28-2.19 (m, 1H), 2.17-2.07 (m, 1H), 1.94 (td, J = 11.6, 4.4 Hz, 1 H), 1.53 - 1.46 (m, 2H), 1.44 (s, 3H), 1.25 - 1.14 (m, 6H), 1.20 (s, 3H), 1.17 (s,
3H), 1.10 - 1.01 (m, 2H), 0.84 (t, J = 6.9 Hz, 3H); LC-MS (ESI+): 435.3 (M+H+); HPLC RT: 4.355 min (Method Villa).
Example 7. Synthesis of (6a/?,10a/?)-6,6-dimethyl-3-(2-methyloctan-2-yl)-9-(pyridin-2-yl)-6a,7,10,10a- tetrahydro-6H-benzo[c]chromen-1-ol (Compound 34)
Figure imgf000037_0001
The starting material was synthesized as indicated in Example 2. The starting material (0.036 g, 0.069 mmol, 1 eq), 2-bromopyridine (0.008 ml_, 0.083 mmol, 1.2 eq), Pd(PPh3)4 (3 mg, 0.003 mmol, 0.05 eq), XPhos (3 mg, 0.006 mmol, 0.1 eq), and potassium carbonate (0.019 g, 0.136 mmol, 4.0 eq) were dissolved in dioxane:water (4:1) (0.7 ml_) in a sealed tube under nitrogen atmosphere. The reaction mixture was stirred at 100 °C for overnight. The mixture was diluted with EtOAc and washed with a saturated aqueous solution of NaHCC>3, the organic layer was dried over MgSC , filtered, and concentrated under reduced pressure. The product was purified by normal phase chromatography on silica gel (Heptane:EtOAc, from 100:0 to 80:20). The desired fractions were collected and concentrated in vacuo to yield Compound 34 as a white solid (0.015 g, 50%). 1H NMR (400 MHz, CDCI3) d 8.57 (dd, J = 4.9, 0.9 Hz, 1 H), 7.67 (td, J= 7.8, 1.8 Hz, 1H), 7.49 (d, J= 8.1 Hz, 1H), 7.15 (dd, J = 6.9, 5.2 Hz, 1H), 6.77-6.68 (m, 1H), 6.35 (d, J= 1.7 Hz, 1H), 6.23 (d, J= 1.8 Hz, 1H), 4.16 (dd, J= 17.3, 2.2 Hz, 1H), 2.84 (td, J= 11.1,4.5 Hz, 1H), 2.53-2.42 (m, 1H), 2.36-2.24 (m, 1H), 2.16-2.04 (m, 1H), 1.94 (td , J = 11.7,4.5 Hz, 1 H), 1.48-1.39 (m, 2H), 1.43 (s,3H), 1.27 - 1.10 (m, 6H), 1.17 (s,3H), 1.16 (s,3H), 1.15 (s, 3H), 1.09 - 0.96 (m, 2H), 0.84 (t, J = 7.0 Hz, 3H); LC-MS (ESI+): 434.0 (M+H+); HPLC RT: 4.489 min (Method Villa). Example 8. Synthesis of (6a/?,10a/?)-6,6-dimethyl-3-(2-methyloctan-2-yl)-9-(pyridin-3-yl)-6a,7,10,10a- tetrahydro-6H-benzo[c]chromen-1-ol (Compound 35)
Figure imgf000038_0001
The starting material was synthesized as indicated in Example 2. The starting material (0.100 g, 0.191 mmol, 1 eq), 3-bromopyridine (0.022 ml_, 0.229 mmol, 1.2 eq), Pd(PPh3)4 (12 mg, 0.010 mmol, 0.05 eq), XPhos (09 mg, 0.019 mmol, 0.1 eq) , and potassium carbonate (0.106 g, 0.764 mmol, 4.0 eq) were dissolved in dioxane:water (4:1) (2 ml_) in a sealed tube under nitrogen atmosphere. The reaction mixture was stirred at 100 °C for overnight. The mixture was diluted with EtOAc and washed with a saturated aqueous solution of NaHCC>3, the organic layer was dried over MgSC , filtered, and concentrated under reduced pressure. The product was purified by normal phase chromatography on silica gel (Heptane:EtOAc, from 100:0 to 80:20). The desired fractions were collected and concentrated in vacuo to yield Compound 35 as a white solid (0.039 g, 47%). 1H NMR (400 MHz, CDCI3) d 9.65 (s 1 H), 8.79 (s, 1 H), 8.51 (d, J = 3.9 Hz, 1H), 7.92 (d, J = 8.1 Hz, 1 H), 7.37 (dd, J = 8.0, 4.8 Hz, 1 H), 6.48 (d, J =
I .7 Hz, 1 H), 6.36 (d, J = 1 .6 Hz, 1 H), 6.31 (d, J = 2.6 Hz, 1 H), 4.21 (dd, J = 17.2, 2.5 Hz, 1 H), 2.88 (td, J =
I I .1 , 4.5 Hz, 1 H), 2.50 - 2.40 (m, 1H), 2.33 - 2.22 (m, 1 H), 2.14 - 2.04 (m, 1 H), 1.96 (td, J = 11.6, 4.3 Hz, 1 H), 1.54 - 1.46 (m, 2H), 1.45 (s, 3H), 1.21 (s, 3H), 1.20 (s, 3H), 1.19 (s, 3H), 1.25 - 1.12 (m, 6H),
1 .11 - 1 .02 (m, 2H), 0.82 (t, J = 6.9 Hz, 3H); LC-MS (ESI+): 434.2 (M+H+); HPLC RT: 4.412 min (Method Villa).
Example 9. Synthesis of (6a/?,10a/?)-6,6-dimethyl-3-(2-methyloctan-2-yl)-9-(pyridin-4-yl)-6a,7,10,10a- tetrahydro-6H-benzo[c]chromen-1-ol (Compound 36)
Figure imgf000038_0002
The starting material was synthesized as indicated in Example 2. The starting material (0.100 g, 0.191 mmol, 1 eq), 4-bromopyridine hydrochloride (0.045 g, 0.229 mmol, 1.2 eq), Pd(PPhi3)4 (12 mg, 0.010 mmol, 0.05 eq), XPhos (9 mg, 0.019 mmol, 0.1 eq), and potassium carbonate (0.106 g, 0.764 mmol, 4.0 eq) were dissolved in dioxane:water (4:1) (2 ml_) in a sealed tube under nitrogen atmosphere. The reaction mixture was stirred at 100 °C for overnight. The mixture was diluted with EtOAc and washed with a saturated aqueous solution of NaHCC>3, the organic layer was dried over MgSC , filtered, and concentrated under reduced pressure. The product was purified by normal phase chromatography on silica gel (Heptane:EtOAc, from 100:0 to 80:20). The desired fractions were collected and concentrated in vacuo to yield Compound 36 as a white solid (0.030 g, 36%). 1H NMR (400 MHz, CDCI3) d 8.52 (d, J = 6.0 Hz, 2H), 7.37 (d, J = 6.2 Hz, 2H), 6.44 - 6.40 (m, 2H), 6.39 (d, J = 1 .6 Hz, 1 H), 3.93 (dd, J = 17.0, 2.3 Hz, 1 H), 2.85 (td, J = 11.1 , 4.4 Hz, 1 H), 2.52 - 2.41 (m, 1 H), 2.35 - 2.22 (m, 1 H), 2.14 - 2.02 (m, 1 H),
1 .95 (td, J = 11 .6, 4.3 Hz, 1 H), 1 .49 (dd, J = 9.8, 6.6 Hz, 2H), 1 .44 (s, 3H), 1 .19 (s, 6H), 1 .25 - 1 .13 (m, 6H), 1.18 (s, 3H), 1.12 - 1 .02 (m, 2H), 0.84 (t, J = 6.9 Hz, 3H); LC-MS (ESI+): 434.2 (M+H+); HPLC RT: 4.408 min (Method Villa)..
Example 10. Synthesis of (6a/?,10a/?)-6,6-dimethyl-9-(5-methyl-1,3,4-oxadiazol-2-yl)-3-(2- methyloctan-2-yl)-6a,7,10,10a-tetrahydro-6H-benzo[c]chromen-1-ol (Compound 37)
Figure imgf000039_0001
The starting material was synthesized as indicated in Example 2. The starting material (0.100 g, 0.191 mmol, 1 eq), 2-bromo-5-methyl-1 ,3,4-oxadiazole (0.037 ml_, 0.229 mmol, 1.2 eq), Pd(PPhi3)4 (12 mg, 0.010 mmol, 0.05 eq), XPhos (9 mg, 0.019 mmol, 0.1 eq), and potassium carbonate (0.106 g, 0.764 mmol, 4.0 eq) were dissolved in dioxane:water (4:1) (2 ml_) in a sealed tube under nitrogen atmosphere. The reaction mixture was stirred at 100 °C for overnight. The mixture was diluted with EtOAc and washed with a saturated aqueous solution of NaHC03, the organic layer was dried over MgS04, filtered, and concentrated under reduced pressure. The product was purified by normal phase chromatography on silica gel (Heptane:EtOAc, from 100:0 to 80:20). The desired fractions were collected and concentrated in vacuo to yield Compound 37 as a white solid (0.037 g, 44%). 1H NMR (400 MHz, CDCI3) d 6.86 - 6.80 (m, 1 H), 6.44 (d, J = 1 .7 Hz, 1 H), 6.32 (d, J = 1 .6 Hz, 1 H), 4.29 (d, J = 15.7 Hz, 1 H), 2.82 (td, J = 11.2, 4.5 Hz, 1 H), 2.54 (s, 3H), 2.52 - 2.43 (m, 1 H), 2.31 - 2.20 (m, 1 H), 2.14 - 2.03 (m, 1 H), 1.93 (td, J = 11.6, 4.3 Hz, 1 H), 1.50 - 1.43 (m, 2H), 1.42 (s, 3H), 1.16 (s, 3H), 1.16 (s, 3H), 1.24 - 1 .12 (m,
6H), 1.15 (s, 3H), 1.11 - 1 .02 (m, 2H), 0.83 (t, J = 6.9 Hz, 3H); LC-MS (ESI+): 439.3 (M+H+); HPLC RT: 4.105 min (Method Villa). Example 11. Synthesis of (6a/?,10a/?)-9-(isoxazol-4-yl)-6,6-dimethyl-3-(2-methyloctan-2-yl)- 6a,7,10,10a-tetrahydro-6H-benzo[c]chromen-1-ol (Compound 38)
Figure imgf000040_0001
Compound 38
The starting material was synthesized as indicated in Example 2. The starting material (0.195 g, 0.372 mmol, 1.1 eq), 4-bromoisoxazole (0.050 ml_, 0.338 mmol, 1 eq), PdChdppf (25 mg, 0.034 mmol, 0.1 eq), and potassium carbonate (0.187 g, 1 .352 mmol, 4.0 eq) were dissolved in dioxane:water (3:1) (3.3 ml_) in a microwave sealed tube under nitrogen atmosphere. The reaction mixture was stirred under microwave irradiation at 100 °C for 1 hour. The mixture was diluted with EtOAc and washed with a saturated aqueous solution of NaHCC>3, the organic layer was dried over MgSC , filtered, and concentrated under reduced pressure. The product was purified by normal phase chromatography on silica gel (Heptane:EtOAc, from 100:0 to 40:60). The desired fractions were collected and concentrated in vacuo to yield Compound 38 as a white solid (0.035 g, 25%). 1H NMR (400 MHz, CDCI3) d 8.37 (d, J = 11 .0 Hz, 2H), 6.42 (d, J = 1 .7 Hz, 1 H), 6.25 (d, J = 1 .7 Hz, 1 H), 6.11 - 6.06 (m, 1 H), 4.79 (s, 1 H), 3.65 (dd, J = 16.9, 3.2 Hz, 1 H), 2.83 (td, J = 11 .1 , 4.7 Hz, 1 H), 2.41 - 2.32 (m, 1 H), 2.21 - 2.11 (m, 1 H), 2.07 - 1.97 (m, 1 H), 1 .91 (td, J = 11 .6, 4.2 Hz, 1 H), 1 .54 - 1 .48 (m, 2H), 1 .42 (s, 3H), 1 .22 (s, 6H), 1 .26 - 1 .17 (m, 6H), 1.15 (s, 3H), 1.12 - 1.03 (m, 2H), 0.85 (t, J = 6.9 Hz, 3H); LC-MS (ESI+): 424.2 (M+H+); HPLC RT: 4.427 min (Method Villa).
Example 12. (6a/?,10a/?)-6,6-dimethyl-3-(2-methyloctan-2-yl)-9-(1 H-1 ,2,3-triazol-4-yl)-6a,7,10,10a- tetrahydro-6H-benzo[c]chromen-1-ol (Compound 39)
Figure imgf000040_0002
To a solution of 4-bromo-1 H-1 ,2,3-triazole (0.146 g, 0.987 mmol, 1 eq) in dry DMF (9.8 ml_) at 0 °C was added 2-(trimethylsilyl)ethoxymethyl chloride (SEMCI, 0.260 ml_, 1.48 mmol, 1.5 eq). The reaction mixture was stirred at rt for overnight. The mixture was diluted with EtOAc and washed with brine, the organic layer was dried over MgS04, filtered, and concentrated under reduced pressure. The product was purified by normal phase chromatography on silica gel (Heptane:EtOAc, from 100:0 to 80:20). The desired fractions were collected and concentrated in vacuo to yield 4-bromo-1-((2- (trimethylsilyl)ethoxy)methyl)-1 H-1 , 2, 3-triazole as a colourless oil (0.107 g, 39%). 1H NMR (300 MHz, CDCb) d 7.62 (s, 1H), 5.62 (s, 2H), 3.63 (dd, J = 16.9, 8.6 Hz, 2H), 0.93 (dd, J = 15.5, 7.2 Hz, 2H), -0.02 (s, 9H); LC-MS (ESI+): 280.0 (M+H+); HPLC RT: 1.565 min (Method Tacc50-6).
Figure imgf000041_0001
The starting material was synthesized as indicated in Example 2. The starting material (0.114 g, 0.217 mmol, 1 eq), 4-bromo-1-((2-(trimethylsilyl)ethoxy)methyl)-1/-/-1 ,2,3-triazole (0.072 g, 0.260 mmol, 1.2 eq), Pd(PPh3)4 (13 mg, 0.013 mmol, 0.05 eq), XPhos (10 mg, 0.022 mmol, 0.1 eq), and potassium carbonate (0.120 g, 0.868 mmol, 4.0 eq) were dissolved in dioxane:water (4:1) (2.2 ml_) in a sealed tube under nitrogen atmosphere. The reaction mixture was stirred at 100 °C for overnight. The mixture was diluted with EtOAc and washed with a saturated aqueous solution of NaHCC>3, the organic layer was dried over MgSC>4, filtered, and concentrated under reduced pressure. The product was purified by normal phase chromatography on silica gel (Heptane: EtOAc, from 100:0 to 60:40). The desired fractions were collected and concentrated in vacuo to yield (6aR,10aR)-6,6-dimethyl-3-(2-methyloctan-2-yl)-9-(1-((2- (trimethylsilyl)ethoxy)methyl)-1H-1 ,2,3-triazol-4-yl)-6a,7,10,10a-tetrahydro-6H-benzo[c]chromen-1-ol as a colourless oil (0.120 g, 98%). LC-MS (ESI+): 554.4 (M+H+); HPLC RT: 2.784 min (Method Tacc50-6).
Figure imgf000041_0002
To a solution of (6aR,10aR)-6,6-dimethyl-3-(2-methyloctan-2-yl)-9-(1-((2-(trimethylsilyl)ethoxy)methyl)-1H- 1 ,2,3-triazol-4-yl)-6a,7,10,10a-tetrahydro-6H-benzo[c]chromen-1-ol (0.120 g, 0.217 mmol, 1 eq) in THF (0.65 mL) was added tetrabutylammonium fluoride hydrate (0.303 g, 1.085 mmol, 5 eq) at 60 °C for overnight. The mixture was diluted with EtOAc and washed with water. The organic layer was dried over MgS04, filtered, and concentrated under reduced pressure. The product was purified by reverse phase HPLC (Gemini C18 column (100 x 30 mm, 5 pm), gradient: 47% [Aqueous phase] - 53% [organic phase] to 18% [Aqueous phase] - 82% [organic phase]. Aqueous phase: 25mM NH4HCO3; Organic phase: 100% Acetonitrile, run time = 30 min). The desired fractions were collected and concentrated to the smallest possible volume. The residue was diluted with CH2CI2 and washed with brine. The organic layer was dried over anhydrous MgS04, filtered and concentrated under reduced pressure to yield Compound 39 as a white solid (0.008 g, 9%). 1H NMR (400 MHz, CDCI3) d 7.75 (s, 1 H), 6.47 (s, 1 H), 6.39 (s, 1 H), 6.33 (s,
1 H), 3.92 (d, J = 15.7 Hz, 1 H), 2.88 - 2.74 (m, 1 H), 2.46 - 2.32 (m, 1 H), 2.24 - 2.10 (m, 1 H), 2.09 - 1.96 (m, 1 H), 1.95 - 1.84 (m, 1 H), 1.53 - 1.46 (m, 2H), 1.41 (s, 3H), 1 .25 - 1 .13 (m, 5H), 1.19 (s, 3H), 1.14 (s, 3H), 1 .10 - 1 .02 (m, 2H), 0.83 (t, J = 6.8 Hz, 3H). LC-MS (ESI+): 424.3 (M+H+); HPLC RT: 3.607 min (Method Villa).
Example 13: Synthesis of 6aR,10aR)-6,6-dimethyl-3-(2-methyloctan-2-yl)-9-(1H-1,2,4-triazol-5-yl)- 6a,7,10,10a-tetrahydro-6H-benzo[c]chromen-1-ol (Compound 40)
Step a: 5-Bromo-1-(2-trimethylsilanyl-ethoxymethyl)-1H-[ 1,2,3]triazole
Figure imgf000042_0001
SEMCI (0.5 mL, 3.0 mmol, 1 .5 eq) was added to a solution of 3-bromo-1 ,2,4-triazole (300 mg, 2.0 mmol, 1 eq) and cesium carbonate (2.6 g, 8.1 mmol, 4 eq) in anhydrous DMF (20 mL) at 0 °C, and the mixture was stirred at room temperature for 16 hours. The reaction mixture was subsequently diluted with ethyl acetate and washed with water and brine. The organic layer was dried over MgSC , filtered, concentrated in vacuo, and chromatographed in heptane/ethyl acetate mixtures to give the product as an oil (248 mg, 44%). 1H NMR (400 MHz, CDCI3) d 7.92 (s, 1 H), 5.51 (s, 2H), 3.86 - 3.48 (m, 2H), 1.13 - 0.75 (m, 2H), 0.01 (d, J = 9.0 Hz, 9H).
Step b: (6aR, 10aR)-6, 6-dimethyl-3-(2-methyloctan-2-yl)-9-( 1-((2-(trimethylsilyl)ethoxy)methyl)-1 H-1 ,2,4- triazol-5-yl)-6a, 7, 10, 10a-tetrahydro-6H-benzo[c]chromen-1-ol
Figure imgf000042_0002
The starting material, (6aR,10aR)-6,6-dimethyl-3-(2-methyloctan-2-yl)-9-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)-6a,7,10,10a-tetrahydro-6/-/-benzo[c]chromen-1-yl acetate, was synthesized as indicated in Example 2. Pd(PPh3)4 (12 mg, 0.01 mmol, 0.05 eq) and XPhos (9 mg, 0.02 mmol, 0.1 eq) were added to a degassed solution of the starting material (100 mg, 0.2 mmol, 1 eq), 5-bromo-1-(2-trimethylsilanyl- ethoxymethyl)-1 H-[1 ,2,3]triazole (64 mg, 0.2 mmol, 1.2 eq), and 1.5 M potassium phosphate (aq.) (0.6 ml_, 0.6 mmol, 3.0 eq) in dioxane:water (4:1) (3 ml_) and the mixture was stirred at 90 °C in a sealed tube under nitrogen atmosphere for 12 hours. The reaction mixture was subsequently diluted with ethyl acetate and washed with NaHCC>3 (aq., sat.). The organic layer was dried over MgSC , filtered, concentrated under reduced pressure, and chromatographed with heptane/ethyl acetate mixtures to give the product as a white solid (70 mg, 66%). LC-MS (ESI+): 554.4 (M+H+); HPLC RT: 2.44 min (Method TACC50-6).
Step c : (6aR, 10aR)-6,6-dimethyl-3-(2-methyloctan-2-yl)-9-(1 H-1 ,2,4-triazol-5-yl)-6a, 7, 10, 10a-tetrahydro-6H- benzo[c]chromen-1-ol (Compound 40)
Figure imgf000043_0001
Compound 40
TBAF (176 mg, 0.6 mmol, 5 eq) was added to a solution of (6aR,10aR)-6,6-dimethyl-3-(2-methyloctan-2-yl)- 9-(1-((2-(trimethylsilyl)ethoxy)methyl)-1 H-1 ,2,4-triazol-5-yl)-6a,7,10,10a-tetrahydro-6H-benzo[c]chromen-1-ol (70 mg, 0.1 mmol, 1 eq) in THF (0.65 ml_) and the reaction was stirred at 60 °C for 12 hours. The reaction mixture was subsequently diluted with ethyl acetate and washed with water. The organic layer was dried over MgSC>4, filtered, concentrated, and purified first by normal phase chromatography eluting with heptane/ethyl acetate and then by reverse phase HPLC (Gemini C18 column (100 x 30 mm, 5 pm) using the following gradient: from 59% aqueous [0.1% formic acid] - 41% acetonitrile to 17% aqueous [0.1 % formic acid] - 83% acetonitrile; run time: 30 min). All the fractions containing the product were directly lyophilized to yield Compound 40 as a white powder (7 mg, 13%). 1H NMR (400 MHz, CDCI3) d 8.18 (s, 1 H), 6.83 (s, 1 H), 6.28 (d, J = 11 .9 Hz, 2H), 3.99 (d, J = 15.7 Hz, 1 H), 3.66 (q, J = 7.0 Hz, 1 H), 2.73 (td, J = 10.9, 4.1 Hz, 1 H), 2.36 (d, J = 17.9 Hz, 1 H), 2.20 - 2.05 (m, 1 H), 2.05 - 1.91 (m, 1 H), 1.82 (td, J = 11.5, 4.1 Hz, 1 H), 1.38 (dd, J = 10.1 , 6.6 Hz, 2H), 1.33 (s, 3H), 1.18 (dd, J = 9.1 , 4.9 Hz, 10H), 1.11 (d, J = 22.2 Hz, 3H), 1.05 (s, 3H), 0.96 (s, 2H), 0.75 (t, J = 6.9 Hz, 3H); LC-MS (ESI+): 424.3 (M+H+); HPLC RT: 3.87 min (Method VILLA).
Example 14. Affinity for CBi and CB2 Receptors as determined by a radioligand binding assay
The binding affinity (% inhibition, Ki) of compounds of the invention for the CBi and CB2 receptors was determined by a competitive radioligand binding assay, the results of which are provided in Table 2. Exemplary methods for the determination of binding affinity for a cannabinoid receptor by competitive radioligand binding can be found in the literature, for example, in Rinaldi-Carmona M, et al. Characterization of two cloned human CB1 cannabinoid receptors isoform. J. Pharmacol. Exp. Ther. 278: 871 (1996); and Munro S., etal. Molecular characterization of a peripheral receptor for cannabinoids. Nature 365:61-65 (1993).
CBi Radioligand Binding Assay: Cell membrane homogenates (5 pg protein) prepared from Chem cells expressing human recombinant CB1 receptor were incubated for 30 min at 22°C with 2 nM [3H]CP 55940 (CB1/CB2 radioligand) in the absence or presence of the test compound in a buffer containing 50 mM Tris-HCI (pH 7.4), 5 mM MgCh, 2.5 mM EDTA and 0.3% BSA. Nonspecific binding is determined in the presence of 10 pM WIN 55212-2. Each compound was tested in 8 concentrations. For each concentration, % Inhibition was determined as a function of radioligand specific binding to the CBi receptor. Inhibition constant (Ki) was determined from the concentration-response curve of each tested compound.
CB2 Radioligand Binding Assay: Cell membrane homogenates (12 pg protein) prepared from CHO cells expressing human recombinant CB2 receptor were incubated for 120 min at 37°C with 0.8 nM [3H]WIN 55212-2 (CB1/CB2 radioligand) in the absence or presence of the test compound in a buffer containing 50 mM Hepes/Tris (pH 7.4), 5 mM MgC , 2.5 mM EGTA and 0.1% BSA. Nonspecific binding was determined in the presence of 5 pM WIN 55212-2. Each compound was tested in 8 concentrations. For each concentration, % Inhibition was determined as a function of radioligand specific binding to the CB2 receptor. Inhibition constant (Ki) was determined from the concentration-response curve of each tested compound.
Table 2
Figure imgf000044_0001
Example 15. CBi- and CB2-mediated activity as determined by cyclic adenosine monophosphate (cAMP) assay
Compounds of the invention were assayed in the Hit Hunter® cAMP assay to determine Gi-coupled agonist activity on the CB1 and CB2 receptors, the results of which are provided in Table 3.
The Hit Hunter® cAMP assay monitors the activation of a GPCR via Gi and Gs secondary messenger signaling in a homogenous, non-imaging assay format using a technology developed by DiscoverX called Enzyme Fragment Complementation (EFC) with b-galactosidase (b-Gal) as the functional reporter. The enzyme is split into two inactive complementary portions: EA for Enzyme Acceptor and ED for Enzyme Donor. ED is fused to cAMP and in the assay competes with cAMP generated by cells for binding to a cAMP-specific antibody. Active b-Gal is formed by complementation of exogenous EA to any unbound ED cAMP. Active enzymes can then convert a chemiluminescent substrate, generating an output signal detectable on a standard microplate reader. cAMP Hunter cell lines were expanded from freezer stocks according to standard procedures. Cells were seeded in a total volume of 20 pL into white walled, 384-well microplates and incubated at 37°C for the appropriate time prior to testing. cAMP modulation was determined using the DiscoverX Hit Hunter cAMP XS+ assay. For Gi agonist activity determination, cells were incubated with sample in the presence of EC80 forskolin to induce response (20 pM and 25 pM in the CB1 and CB2 assays, respectively). Media was aspirated from cells and replaced with 15 pL 2:1 HBSS/10 mM HEPES : cAMP XS+ Ab reagent. Intermediate dilution of sample stocks was performed to generate 4X sample in assay buffer. 5 pL of 4X compound was added to cells and incubated at 37°C or room temperature for 30 or 60 minutes. Final vehicle concentration was 1%. Assay signal was generated through incubation with 20 pL cAMP XS+ ED/CL lysis cocktail for one hour at room temperature. Microplates were read following signal generation with a PerkinElmer Envision™ instrument for chemiluminescent signal detection. Compound activity was analyzed using CBIS data analysis suite (Chemlnnovation, CA). For agonist assays, percentage activity was calculated using the following formula: % Activity = 100% x (1 - (mean RLU of test sample - mean RLU of Max control ligand) / (mean RLU of vehicle control - mean RLU of Max control ligand). Control ligand was the non-selective CB1/CB2 agonist CP55,940.
Table 3
Figure imgf000045_0001
Example 16. CBi- and CB2-mediated activity as determined by b-Arrestin assay
Compounds of the invention were assayed in the PathHunter® b-Arrestin assay to determine agonist activity on the CBi and CB2 receptors, the results of which are provided in Table 4.
The PathHunter® b-Arrestin assay monitors the activation of a GPCR in a homogenous, non-imaging assay format using a technology developed by DiscoverX called Enzyme Fragment Complementation (EFC) with b-galactosidase (b-Gal) as the functional reporter. The enzyme is split into two inactive complementary portions (EA for Enzyme Acceptor and PK for ProLink) expressed as fusion proteins in the cell. EA is fused to b-Arrestin and PK is fused to the GPCR of interest.
PathHunter cell lines were expanded from freezer stocks according to standard procedures. Cells were seeded in a total volume of 20 pL into white walled, 384-well microplates and incubated at 37°C for the appropriate time prior to testing. For agonist activity determination, cells were incubated with sample to induce response. Intermediate dilution of sample stocks was performed to generate 5X sample in assay buffer. 5 pl_ of 5X sample was added to cells and incubated at 37°C or room temperature for 90 to 180 minutes. Vehicle concentration was 1%. Assay signal was generated through a single addition of 12.5 or 15 mI_ (50% v/v) of PathHunter Detection reagent cocktail, followed by a one hour incubation at room temperature. Microplates were read following signal generation with a PerkinElmer Envision™ instrument for chemiluminescent signal detection. Compound activity was analyzed using CBIS data analysis suite (Chemlnnovation, CA). For agonist assays, percentage activity was calculated using the following formula: % Activity = 100% x (mean RLU of test sample - mean RLU of vehicle control) / (mean MAX control ligand - mean RLU of vehicle control).
Table 4
Figure imgf000046_0001
Example 17. Biological activity as determined by an inflammation bioassay in human Peripheral Blood Mononuclear Cells (PBMCs)
Compounds of the invention were assayed for their effect on inflammatory cytokine release from lipopolysaccharides (LPS)-induced PBMCs, isolated from a human blood sample, the results of which are provided in Table 5 and FIGs. 1A-1J. Exemplary methods for quantifying secretion of inflammatory cytokines in PBMCs are known in the art, for example, as described in Haller et al., Infection and Immunity. 68(2)752-759 (2000); and Merlini et al., Frontiers in Immunology. 7:614 (2016); each of which is incorporated herein by reference with respect to methods for evaluating cytokine release in PBMCs. In brief, human blood samples from healthy volunteers were collected and PBMCs were isolated. PBMCs were cultured and incubated with the compounds 2 hours. In experiment 1 , compounds were added at final concentrations of 1 and 10 pM and in experiment 2, at final concentrations of 0.37, 1 .1 , 3.3 and 10 pM. 1 pg/ml dexamethasone (DEX) served as a positive control. LPS was added at a final concentration of 0.1 pg/ml and further incubated for 24 hours. At the end of the incubation, the supernatants were collected and the levels of a panel of secreted cytokines were measured by a Human Magnetic Luminex® assay (R&D).
The results of experiment 1 are presented in Table 5 as a fold change from LPS-treated PBMCs.
Reduced levels are expressed by negative values according to the formula: (-)1 /fold change). OOR denotes values that are out of the standard curve range for the specific cytokine and thus, could not be accurately measured. Cytotoxicity was also determined. We observed no cytotoxic effect with the compounds presented in Table 5 as compared to the vehicle control (DMSO).
Table 5
Figure imgf000047_0001
The results of experiment 2 are presented in FIGs. 1 A-1 J as raw data (pg/ml of each specific cytokine) from two donors. We observed no cytotoxic effect with the compounds presented in FIGs. 1 A-1 J as compared to the vehicle control (DMSO).
Example 18. Biological activity as determined by a phenotypic screen in the BioMap Diversity Phenotypic Screen platform
Compounds of the invention were assayed for their effects on 148 biomarkers of inflammation, immune modulation and tissue remodeling in the BioMAP® Diversity PLUS® (DiscoverX) platform designed to model aspects of human diseases in vitro (Shah et al, 2017). Example results are provided in Table 6. The BioMap platform consists of 12 systems of human primary cells: 3C: venular endothelial cells stimulated with IL1 -b, TNFa and INFy; 4H: venular endothelial cells stimulated with IL-4 and histamine; LPS: PBMCs co-cultured with venular endothelial cells and stimulated with TLR4 ligand; SAg: PBMCs cocultured with venular endothelial cells and stimulated with TCR ligands; BT: PBMCs co-cultured with B cells and stimulated with a-IGM and TCR ligands; BF4T: bronchial epithelial cells co-cultured with dermal fibroblast and stimulated with TNFa and IL-4; BE3C: bronchial epithelial cells stimulated with IL1 -b, TNFa and INFy; CASM3C: coronary artery smooth muscle cells stimulated with ILI-b, TNFa and INFy; HDF3CGF: dermal fibroblasts stimulated with IL1-p, TNFa, INFy, EGF, bFGF and PDGF-BB; KF3CT: keratinocytes co-cultured with dermal fibroblasts and stimulated with IL1 -b, TNFa, INFy and TGFp; MyoF: lung fibroblasts stimulated with TNFa and TGFp; /Mphg: venular endothelial cells co-cultured with macrophages and stimulated with TLR2 ligand. The example compounds in Table 6 (3.3 pM) induced increases or decreases in the levels of the indicated biomarkers, which are at least 1.5-fold change from vehicle control and are outside the significance prediction envelop. All compounds presented in Table 6 were not cytotoxic.
Table 6
Figure imgf000048_0001
Example 19. Biological activity as determined by an T cell proliferation bioassay in human T cells isolated from Peripheral Blood Mononuclear Cells (PBMCs)
Compounds of the invention were assayed for their effect on T cell proliferation, the results of which are provided in FIGs. 2A-2D. Exemplary methods for quantifying the percent of T cell proliferation are known in the art, for example, as described in Trickett & Kwan., Journal of immunological Methods. 275: 251-255 (2003).
In brief, human blood samples from healthy volunteers were collected and CD3+ T cells were isolated from PBMCs. The T cells were cultured with anti- human CD3/CD28 beads in the presence of the compound and incubated at 37 °C for 72 hours. Compounds were added at final concentration of 0.37, 1.1 , 3 and 10 pM. At the end of the incubation, the percent of proliferative cells were determined by FACS analysis (CD45+, CD4+ and CD8+ T cells). Cell viability was also evaluated, and Cyclosporin A served as a positive control in all 3 donors. The results are presented in FIGs. 2A-2C as percent CD4+ and CD8+ proliferation from 3 donors. Representative results of cyclosporin A from 2 donors are presented (FIG. 2D). We observed no cytotoxic effect with the compounds as compared to the vehicle control (with or without beads).
Example 20. Biological activity as determined by a fibroblast-to-myofibroblast transition (FMT) assay in human lung fibroblast from healthy donors.
Compounds of the invention were assayed for their effect on fibroblast activation, the results of which are provided in FIG. 3. Exemplary methods for quantifying the levels of alpha smooth muscle actin (aSMA) as a measure of fibroblast to myofibroblast transition and activation are known in the art, for example, as described in Zhao et al, Medical Science Monitor. 24: 6280-6288 (2018) and Jung et al, Marine Drugs.
16: 323 (2018).
In brief, human lung fibroblast from 3 healthy donors were cultured and stimulated with 1.25 ng/ml TGF to induce FMT 1-hour post compound addition. Compounds of the inventions were added at 8 concentrations in duplicates and the cells were incubated for 72 hours. 0.1% DMSO served as vehicle control and 1 pM SB525334 served as a positive control. At the end of the incubation, the cells were fix with 4% formaldehyde and stained with aSMA antibody and DAPI for nuclei counts. aSMA levels were calculated using a high content imaging algorithm, with signal density X area output. The results are presented as the percent of aSMA levels following compound treatment and TGF stimulation (3 concentrations of the compounds are presented) as, percent of unstimulated, vehicle treated fibroblasts. Remaining nuclei are also presented as percent of unstimulated, vehicle treated fibroblasts. We did not observe nuclei loss with the compound tested.
Other Embodiments
While the invention has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the invention that come within known or customary practice within the art to which the invention pertains and may be applied to the essential features hereinbefore set forth, and follows in the scope of the claims. Other embodiments are within the claims.
This application claims the benefit of U.S. serial no. 62/938,080, filed on 20 November 2019, and EP19382942.1 , filed on 29 October 2019, each of which is incorporated herein in its entirety.

Claims

What is claimed is: CLAIMS
1 . A compound described by formula (I):
Figure imgf000050_0001
wherein Ri is optionally substituted 3-to-8 membered heterocyclyl;
R2 IS H, OH, F, Cl, Br, NH2, or optionally substituted C1-C3 alkoxy;
R3 is optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C1-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C1-C20 heteroalkynyl, optionally substituted C5-C15 aryl, optionally substituted C2-C15 heterocyclyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C1-C20 alkoxy, or optionally substituted amide;
R4 and R5 are each independently H, -CH3, -CF3, -CH2OH, -CH2F, or -CHF2; and
Re is -CH3 or -CH2OH, or a pharmaceutically acceptable salt thereof.
2. The compound of claim 1 , wherein Ri is optionally substituted 3-to-6 membered heteroaryl.
3. The compound of claim 1 , wherein Ri is optionally substituted 5-membered heterocyclyl.
4. The compound of claim 3, wherein Ri is optionally substituted pyrrole, optionally substituted pyrazole, optionally substituted isoxazole, optionally substituted pyrrolidine, optionally substituted imidazole, optionally substituted thiazole, optionally substituted thiophene, optionally substituted thiolane, optionally substituted furan, optionally substituted tetrahydrofuran, optionally substituted diazole, optionally substituted triazole, optionally substituted tetrazole, optionally substituted oxazole, optionally substituted 1 ,3,4-oxadiazole, optionally substituted 1 ,3,4-thiadiazole, optionally substituted 1 , 2,3,4- oxatriazole, or optionally substituted 1 ,2,3,4-thiatriazole.
5. The compound of claim 3, wherein Ri is
Figure imgf000051_0001
6. The compound of claim 5, wherein Ri is
Figure imgf000051_0002
7. The compound of claim 3, wherein
Figure imgf000051_0003
8. The compound of claim 1 , wherein Ri is optionally substituted 6- or 7-membered heterocyclyl.
9. The compound of claim 8, wherein Ri is optionally substituted pyridine, optionally substituted piperidine, optionally substituted piperazine, optionally substituted pyrimidine, optionally substituted pyrazine, optionally substituted pyridazine, optionally substituted triazine, optionally substituted azepine, optionally substituted 1 ,4-diazepine, optionally substituted 2/-/-pyran, optionally substituted 4/-/-pyran, optionally substituted tetrahydropyran, optionally substituted thiepine, or optionally substituted 1 ,4- thiazepine.
10. The compound of claim 8, wherein Ri is
Figure imgf000051_0004
11 . The compound of claim 10, wherein Ri is
Figure imgf000051_0005
12. The compound of claim 8, wherein Ri is
Figure imgf000052_0001
13. The compound of any one of claims 1-12, wherein R2 is OH.
14. The compound of any one of claims 1-12, wherein R2 is H.
15. The compound of any one of claims 1-12, wherein R2 is F.
16. The compound of any one of claims 1-12, wherein R2 is Cl.
17. The compound of any one of claims 1-12, wherein R2 is Br.
18. The compound of any one of claims 1-12, wherein R2 is NH2.
19. The compound of any one of claims 1-12, wherein R2 is -OCH3, -OCH2CH3, or
-OCH2CH2CH3.
20. The compound of any one of claims 1 -19, wherein R3 is an optionally substituted C3-C7 alkyl.
21 . The compound of claim 20, wherein R3 is
Figure imgf000052_0002
22. The compound of claim 20, wherein R3 is
Figure imgf000053_0001
23. The compound of any one of claims 1 -19, wherein R3 is optionally substituted C2-C7 heteroalkyl.
24. The compound of claim 23, wherein R3 is
Figure imgf000053_0002
25. The compound of any one of claim 1 -19, wherein R3 is optionally substituted C3-C7 alkenyl.
26. The compound of claim 25, wherein R3 is
Figure imgf000053_0003
27. The compound of any one of claims 1 -19, wherein R3 is optionally substituted phenyl.
28. The compound of claim 27, wherein R3 is
Figure imgf000053_0004
29. The compound of any one of claim 1 -19, wherein R3 is optionally substituted 5- to 7- membered heterocyclyl.
30. The compound of claim 29, wherein R3 is optionally substituted pyridine, optionally substituted piperidine, optionally substituted piperazine, optionally substituted pyrimidine, optionally substituted pyrazine, optionally substituted pyridazine, optionally substituted triazine, optionally substituted azepine, optionally substituted 1 ,4-diazepine, optionally substituted 2 H- pyran, optionally substituted 4/-/-pyran, optionally substituted tetrahydropyran, optionally substituted thiepine, or optionally substituted 1 ,4-thiazepine.
31 . The compound of claim 29, wherein R3 is
Figure imgf000054_0001
32. The compound of any one of claims 1 -19, wherein R3 is an optionally substituted amide of formula -C(0)NHR8, wherein Rs is H, optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C1-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C1-C20 heteroalkynyl, optionally substituted C5-C15 aryl, optionally substituted C2-C15 heterocyclyl, optionally substituted C3-C20 cycloalkyl, or optionally substituted C1-C20 alkoxy.
33. The compound of claim 32, wherein R3 is
Figure imgf000054_0002
34. The compound of any one of claims 1 -33, wherein R4 is -CH3
35. The compound of any one of claims 1 -33, wherein F is -CH2OH
36. The compound of any one of claims 1 -33, wherein R4 is -CF3
37. The compound of any one of claims 1 -33, wherein R4 is -CH2F.
38. The compound of any one of claims 1 -33, wherein R4 is -CHF2.
39. The compound of any one of claims 1 -38, wherein Rs is -CH3
40. The compound of any one of claims 1 -38, wherein Rs is -CH2OH
41 . The compound of any one of claims 1 -38, wherein Rs is -CF3
42. The compound of any one of claims 1 -38, wherein Rs is -CH2F.
43. The compound of any one of claims 1 -38, wherein Rs is -CHF2.
44. The compound of any one of claims 1 -43, wherein R6 is -CH3
45. The compound of any one of claims 1 -43, wherein R6 is -CH2OH
46. The compound of claim 1 , wherein the compound is described by formula (II):
Figure imgf000055_0001
wherein Ri is optionally substituted 3-to-8 membered heterocyclyl;
R2 IS H, OH, F, Cl, Br, NH2, or optionally substituted C1-C3 alkoxy; and R3 is optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C1-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C1-C20 heteroalkynyl, optionally substituted C5-C15 aryl, optionally substituted C2-C15 heterocyclyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C1-C20 alkoxy, or optionally substituted amide; or a pharmaceutically acceptable salt thereof.
47. The compound of claim 46, wherein the compound is described by formula (III):
Figure imgf000056_0001
wherein Ri is optionally substituted 3-to-8 membered heterocyclyl; and R3 optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C1-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C1-C20 heteroalkynyl, optionally substituted C5-C15 aryl, optionally substituted C2-C15 heterocyclyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C1-C20 alkoxy, or optionally substituted amide; or a pharmaceutically acceptable salt thereof.
48. The compound of claim 47, wherein the compound is described by formula (ill-1):
Figure imgf000056_0002
wherein Ri is optionally substituted 3-to-8 membered heterocyclyl, or a pharmaceutically acceptable salt thereof.
49. The compound of claim 47, wherein the compound is described by formula (III-2):
Figure imgf000056_0003
wherein Ri is optionally substituted 3-to-8 membered heterocyclyl, or a pharmaceutically acceptable salt thereof.
50. The compound of claim 47, wherein the compound is described by formula (ill-3):
Figure imgf000057_0001
wherein Ri is optionally substituted 3-to-8 membered heterocyclyl, or a pharmaceutically acceptable salt thereof.
51 . The compound of claim 46, wherein the compound is described by formula (IV):
Figure imgf000057_0002
wherein Ri is optionally substituted 3-to-8 membered heterocyclyl; and R3 optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C1-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C1-C20 heteroalkynyl, optionally substituted C5-C15 aryl, optionally substituted C2-C15 heterocyclyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C1-C20 alkoxy, or optionally substituted amide; or a pharmaceutically acceptable salt thereof.
52. The compound of claim 51 , wherein the compound is described by formula (IV-1):
Figure imgf000057_0003
wherein Ri is optionally substituted 3-to-8 membered heterocyclyl, or a pharmaceutically acceptable salt thereof.
53. The compound of claim 51 , wherein the compound is described by formula (IV-2):
Figure imgf000058_0001
(IV-2) wherein Ri is optionally substituted 3-to-8 membered heterocyclyl, or a pharmaceutically acceptable salt thereof.
54. The compound of claim 51 , wherein the compound is described by formula (IV-3):
Figure imgf000058_0002
wherein Ri is optionally substituted 3-to-8 membered heterocyclyl, or a pharmaceutically acceptable salt thereof.
55. The compound of any one of claims 46-54, wherein Ri is optionally substituted 3-to-6 membered heteroaryl.
56. The compound of any one of claims 46-54, wherein Ri is optionally substituted 5-membered heterocyclyl.
57. The compound of claim 56, wherein Ri is optionally substituted pyrrole, optionally substituted pyrazole, optionally substituted isoxazole, optionally substituted pyrrolidine, optionally substituted imidazole, optionally substituted thiazole, optionally substituted thiophene, optionally substituted thiolane, optionally substituted furan, optionally substituted tetrahydrofuran, optionally substituted diazole, optionally substituted triazole, optionally substituted tetrazole, optionally substituted oxazole, optionally substituted 1 ,3,4-oxadiazole, optionally substituted 1 ,3,4-thiadiazole, optionally substituted 1 , 2,3,4- oxatriazole, or optionally substituted 1 ,2,3,4-thiatriazole.
58. The compound of claim 56, wherein Ri is
Figure imgf000059_0001
59. The compound of claim 58, wherein Ri is
Figure imgf000059_0002
60. The compound of claim 56, wherein
Figure imgf000059_0003
61 . The compound of any one of claims 46-54, wherein Ri is optionally substituted 6- or 7- membered heterocyclyl.
62. The compound of claim 61 , wherein Ri is optionally substituted pyridine, optionally substituted piperidine, optionally substituted piperazine, optionally substituted pyrimidine, optionally substituted pyrazine, optionally substituted pyridazine, optionally substituted triazine, optionally substituted azepine, optionally substituted 1 ,4-diazepine, optionally substituted 2/-/-pyran, optionally substituted 4/-/-pyran, optionally substituted tetrahydropyran, optionally substituted thiepine, or optionally substituted 1 ,4-thiazepine.
63. The compound of claim 61 , wherein Ri is
Figure imgf000059_0004
64. The compound of claim 63, wherein Ri is
Figure imgf000059_0005
65. The compound of claim 61 , wherein Ri is
Figure imgf000060_0001
66. The compound of claim 1 , wherein the compound is any one of compounds 1 -40.
67. A pharmaceutical composition comprising the compound of any claims 1-66, or a salt thereof, and a pharmaceutically acceptable excipient.
68. A method of treating an inflammatory disease in a subject in need thereof, the method comprising administering to the subject a pharmaceutical composition of claim 67 in an amount sufficient to treat the condition.
69. The method of claim 68, wherein the inflammatory disease is selected from the group consisting of scleroderma, dermatomyositis, systemic lupus erythematosus, acquired immune deficiency syndrome (AIDS), multiple sclerosis, rheumatoid arthritis, psoriasis, diabetes, cancer, asthma, atopic dermatitis, an autoimmune thyroid disorders, ulcerative colitis, Crohn’s disease, stroke, ischemia, a neurodegenerative disease, amyotrophic lateral sclerosis (ALS), chronic traumatic encephalopathy (CTE), chronic inflammatory demyelinating polyneuropathy, an autoimmune inner ear disease, uveitis, iritis, and peritonitis.
70. The method of claim 69, wherein the inflammatory disease is scleroderma.
71. The method of claim 70, wherein the scleroderma is selected from systemic sclerosis, localized scleroderma, or sine scleroderma.
72. The method of claim 69, wherein the inflammatory disease is dermatomyositis.
73. A method of treating a fibrotic disease in a subject in need thereof, the method comprising administering to the subject the pharmaceutical composition of claim 67 in an amount sufficient to treat the condition.
74. The method of claim 73, wherein the fibrotic disease is selected from the group consisting of scleroderma, cystic fibrosis, liver cirrhosis, interstitial pulmonary fibrosis, idiopathic pulmonary fibrosis, Dupuytren’s contracture, keloids, chronic kidney disease, chronic graft rejection, scarring, wound healing, post-operative adhesions, reactive fibrosis, polymyositis, ANCA vasculitis, Behcet's disease, antiphospholipid syndrome, relapsing polychondritis, Familial Mediterranean Fever, giant cell arteritis, Graves ophthalmopathy, discoid lupus, pemphigus, bullous pemphigoid, hydradenitis suppuritiva, sarcoidosis, bronchiolitis obliterans, primary sclerosing cholangitis, primary biliary cirrhosis, or organ fibrosis.
75. The method of claim 74, wherein the fibrotic disease is scleroderma.
76. The method of claim 75, wherein the scleroderma is selected from systemic sclerosis, localized scleroderma, or sine scleroderma.
77. The method of claim 74, wherein the fibrotic disease is organ fibrosis.
78. The method of claim 77, wherein the organ fibrosis is selected from dermal fibrosis, lung fibrosis, liver fibrosis, kidney fibrosis, or heart fibrosis.
PCT/US2020/057985 2019-10-29 2020-10-29 Cannabinoids and uses thereof WO2021087127A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP19382942.1 2019-10-29
EP19382942 2019-10-29
US201962938080P 2019-11-20 2019-11-20
US62/938,080 2019-11-20

Publications (2)

Publication Number Publication Date
WO2021087127A1 true WO2021087127A1 (en) 2021-05-06
WO2021087127A8 WO2021087127A8 (en) 2021-07-15

Family

ID=75715603

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2020/057985 WO2021087127A1 (en) 2019-10-29 2020-10-29 Cannabinoids and uses thereof

Country Status (1)

Country Link
WO (1) WO2021087127A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040186166A1 (en) * 2002-12-19 2004-09-23 Burstein Sumner H. Cannabinoid analogs as peroxisome proliferator activated nuclear receptor gamma activators
WO2007125049A1 (en) * 2006-04-27 2007-11-08 Solvay Pharmaceuticals Gmbh Use of cbx cannabinoid receptor modulators as potassium channel modulators
US20170022179A1 (en) * 2010-10-05 2017-01-26 Corbus Pharmaceuticals, Inc. Compositions, dosages, and methods of using tetrahydrocannabinol derivatives

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040186166A1 (en) * 2002-12-19 2004-09-23 Burstein Sumner H. Cannabinoid analogs as peroxisome proliferator activated nuclear receptor gamma activators
WO2007125049A1 (en) * 2006-04-27 2007-11-08 Solvay Pharmaceuticals Gmbh Use of cbx cannabinoid receptor modulators as potassium channel modulators
US20170022179A1 (en) * 2010-10-05 2017-01-26 Corbus Pharmaceuticals, Inc. Compositions, dosages, and methods of using tetrahydrocannabinol derivatives

Also Published As

Publication number Publication date
WO2021087127A8 (en) 2021-07-15

Similar Documents

Publication Publication Date Title
JP6970802B2 (en) Methods of Inducing Targeted Proteolysis by Bifunctional Molecules
US20210284621A1 (en) Cannabinoids and uses thereof
CN106456795A (en) Targeted therapeutics
JP2016515530A (en) Intravenous emulsion of triptolide as an immunomodulator and anticancer agent
US10370400B2 (en) 4-methylumbelliferone derivatives for treatment for immune modulation
Bettayeb et al. N-&-N, a new class of cell death-inducing kinase inhibitors derived from the purine roscovitine
US8642612B2 (en) Nicotinic desensitizers and methods of selecting, testing, and using them
Fujisawa Therapeutic application of apoptosis signal-regulating kinase 1 inhibitors
JP4028888B2 (en) Novel uses of bis-heterocyclic compounds and pharmaceutical compositions containing them
ITMI20130647A1 (en) COMPOUNDS WITH INHABITING ACTIVITIES ON SIRTUINES
CA2728766A1 (en) Compositions for the treatment of fibrotic diseases or conditions
WO2021087127A1 (en) Cannabinoids and uses thereof
Qi et al. Discovery of NAFLD-improving agents by promoting the degradation of Keap1
Wang et al. Small-molecule inhibitors targeting apoptosis signal-regulated kinase 1
TWI649082B (en) Method for treating cancer using an AURORA kinase inhibitor
EP2217242A1 (en) Methods of use
WO2021067834A1 (en) Cannabinoids and uses thereof
CA3117666A1 (en) Biomarkers of metap2 inhibitors and applications thereof
WO2021113656A1 (en) Cannabinoids and uses thereof
US9233949B2 (en) Treatment for oxidative stress and/or hypertension
WO2021113669A1 (en) Cannabinoids and uses thereof
CN117222407A (en) Small molecule activators of YAP transcriptional activity for regenerative organ repair
WO2014039452A1 (en) Methods of treating a bruton's tyrosine kinase disease or disorder
DE10250870A1 (en) Composition containing activators of IK potassium channels and calcineurin antagonists and their use
AU2020211994A1 (en) Non-immunosuppressive FK506 analogs and use thereof

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: 20880939

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20880939

Country of ref document: EP

Kind code of ref document: A1