专利摘要:
COMPOUNDS DERIVED FROM METHYL BARDOXOLONE 2,2-DIFLOROPROPIONAMIDE, POLYMORPHIC FORMS AND PHARMACEUTICAL COMPOSITION FOR USE INCLUDING THEM. The present invention generally relates to the compound: N - ((4aS ,6aR ,6bS ,8aR ,12aS ,14aR ,14bS)-11-cyano-2,2,6a,6b,9,9,12a-heptamethyl- 10,14- dioxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a, 9,10,12a,14,14a, 14b-octadecahydropicen-4a-yl) -2,2-difluoropropanamide and polymorphic forms thereof. The present invention also relates to methods of preparation and use thereof, pharmaceutical compositions, and kits and articles of manufacture thereof.
公开号:BR112014026640B1
申请号:R112014026640-9
申请日:2013-04-24
公开日:2021-05-18
发明作者:Eric Anderson;Andrea Decker;Xiaofeng Liu
申请人:Reata Pharmaceuticals, Inc;
IPC主号:
专利说明:

BACKGROUND OF THE INVENTION
[001] The present application claims the benefit of priority to US Provisional Application No. 61/780444, filed March 13, 2013, US Provisional Application No. 61/775288, filed March 8, 2013, and US Provisional Application No. 61/687669, filed on April 27, 2012; the entire contents of each are hereby incorporated by reference.
[002] In accordance with 37 CFR 1.821(c), a string list is attached as an ASCII compliant text file named "REATP0073WO_ST25", created on April 24, 2013 and which has a size of ~ 6 kilobytes. The contents of that file are incorporated herein by reference in their entirety. I. Field of Invention
[003] The present invention relates generally to the compound:
[004] N-((4aS,6aR,6bS,8aR,12aS,14aR,14bS)-11-cyano-2,2,6a,6b,9,9,12a-heptamethyl-10,14-dioxo-1, 2,3,4,4a,5,6,6a,6b,7,8,8a,9,10, 12a,14,14a,14b-octadecahydropicen-4a-yl)-2,2-difluoropropanamide, also referred to herein as RTA 408, 63415, or PP415. The present invention also concerns polymorphic forms thereof, methods for the preparation and use thereof, their pharmaceutical compositions and kits and articles of manufacture thereof. II. Description of Related Art
[005] The anti-inflammatory and antiproliferative activity of the naturally occurring triterpenoid, oleanolic acid, has been improved by chemical modifications. For example, 2-cyano-3,12-diooxoolean 1,9-(11)-dien-28-oic acid (CDDO) and related compounds have been developed. See Honda et al., 1997; Honda et al., 1998; Honda et al., 1999; Honda et al., 2000a; Honda et al., 2000b; Honda et al., 2002; Suh et al., 1998; Suh et al., 1999; Place et al., 2003; Liby et al., 2005; and US Patents 8129429, 7915402, 8124799, and 7943778, all of which are incorporated herein by reference. The methyl ester, methyl bardoxolone (CDDO-Me), was evaluated in phase II and III clinical trials for the treatment and prevention of diabetic nephropathy and chronic kidney disease. See Pergola et al. 2011, which is incorporated herein by reference.
[006] Synthetic triterpenoid analogues of oleanolic acid have also been shown to be inhibitors of cellular inflammatory processes, such as the induction by IFN-g of nitric oxide synthase (iNOS) and COX-2 in mouse macrophages. See Honda et al., (2000a), Honda et al. (2000b), Honda et al. (2002), and US Patents 8,129,429, 7,915,402, 8,124,799, and 7,943,778, all of which are incorporated herein by reference. . Compounds derived from oleanolic acid have been shown to affect the function of several target proteins and thereby modulate the activity of several important cell signaling pathways related to oxidative stress, cell cycle control, and inflammation (eg, Dinkova- Kostova et al., 2005; Ahmad et al., 2006; Ahmad et al., 2008; Liby et al., 2007a, and US Patents 8129429, 7915402, 8124799, and 7943778).
[007] Since the biological activity profiles of known triterpenoid derivatives vary, and taking into account the wide variety of diseases that can be treated or prevented with compounds having potent antioxidant and anti-inflammatory effects, and the high degree of medical necessity not represented in this variety of diseases, it is desirable to synthesize new compounds with different biological activity profiles for the treatment or prevention of one or more indications. SUMMARY OF THE INVENTION
In some aspects of the present invention, a compound of formula (also referred to herein as RTA 408, 63415, or PP415) is envisioned:

[009] or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is in the form of a pharmaceutically acceptable salt. In some embodiments, the compound is not in the form of a salt.
In another aspect of the present invention, polymorphic forms of the above compound are envisioned. In some embodiments, the polymorphic form has an X-ray diffraction (CuKa) pattern that comprises a halogen peak at about 14o2θ. In some embodiments, the X-ray diffraction (CuKa) pattern further comprises a shoulder at the peak of about 8°2θ. In some embodiments, the X-ray diffraction (CuKa) pattern is substantially as shown in FIG. 59. In some embodiments, the polymorphic form has a Tg of about 150°C to about 155°C, including, for example, a Tg of about 153°C or a Tg of about 150°C. In some embodiments, the polymorphic form has a differential scanning calorimetry (DSC) curve, which comprises a centered endotherm of about 150°C to about 155°C. In some embodiments, the endotherm is centered at about 153°C. In some embodiments, the endotherm is centered at about 150°C. In some embodiments, the differential scanning calorimetry (DSC) curve is substantially as shown in FIG. 62.
[0012] In some embodiments, the polymorphic form is a solvate with an X-ray diffraction (CuKa) pattern comprising peaks at about 5.6, 7.0, 10.6, 12.7, and 14.6 o2θ . In some embodiments, the X-ray diffraction (CuKa) pattern is substantially as shown in FIG. 75, top standard.
[0013] In some embodiments, the polymorphic form is a solvate with an X-ray diffraction pattern (CuKa) comprising peaks at about 7.0, 7.8, 8.6, 11.9, 13.9 (peak double), 14.2, and 16.0 o2θ. In some embodiments, the X-ray diffraction (CuKa) pattern is substantially as shown in FIG. 75, the second top pattern.
[0014] In some embodiments, the polymorphic form is an acetonitrile hemissolvate with an X-ray diffraction pattern (CuKa) comprising peaks at about 7.5, 11.4, 15.6 and 16.6 o2θ. In some embodiments, the X-ray diffraction (CuKa) pattern is substantially as shown in FIG. 75, the second lowest pattern. In some embodiments, the polymorphic form has a Tg of about 196°C. In some embodiments, the polymorphic form has a differential scanning calorimetry (DSC) curve, which comprises an endotherm centered at about 196°C. In some embodiments, the differential scanning calorimetry (DSC) curve substantially as shown in FIG. 116.
[0015] In some embodiments, the polymorphic form is a solvate with an X-ray diffraction (CuKa) pattern comprising peaks at about 6.8, 9.3, 9.5, 10.5, 13.6, and 15.6 the 2. In some embodiments, the X-ray diffraction (CuKa) pattern is substantially as shown in FIG. 75, lower standard.
[0016] In another aspect of the present invention, pharmaceutical compositions comprising an active ingredient consisting of the above compound or a polymorphic form thereof (such as, for example, any of the polymorphic forms described herein above and below), and a pharmaceutically acceptable carrier. In some modalities, the pharmaceutical composition is formulated for administration: orally, intra-adipose, intra-arterial, intra-articular, intracranial, intradermal, intralesional, intramuscularly, intranasally, intraocularly, intrapericardially, intraperitoneally, intrapleurally, intraprostatic, intrarectally, intrathecally, intratracheal, intratumoral, intraumbilical, intravaginally, intravenously, intravesicular, intravitreal, in liposomes, locally, mucosally, parenterally, rectal, subconjunctival, subcutaneously, by sublingually, topically, transbuccally, transdermally, vaginally, in creams, in lipid compositions, by means of a catheter, by means of a wash, by means of continuous infusion, by infusion, by inhalation , via injection, through local delivery, or through localized perfusion. In some embodiments, the pharmaceutical composition is formulated for oral, intra-arterial, intravenous or topical administration. In some embodiments, the pharmaceutical composition is formulated for oral administration.
In some embodiments, the pharmaceutical composition is formulated as a hard or soft capsule, a lozenge, a syrup, a suspension, an emulsion, a solution, a solid dispersion, a wafer, or an elixir. In some embodiments, the pharmaceutical composition according to the invention further comprises an agent that enhances solubility and dispersibility. (For example, agents that improve solubility and dispersibility include, but are not limited to, PEGs, cyclodextranes, and cellulose derivatives.) In some embodiments, the polymorphic form or compound is suspended in sesame oil.
[0018] In other embodiments, the pharmaceutical composition is formulated for topical administration. In other embodiments, the pharmaceutical composition is formulated as a lotion, cream, gel, oil, ointment, ointment, emulsion, solution, or suspension. In some embodiments, the pharmaceutical composition is formulated as a lotion, cream, or gel. In some embodiments, the amount of active ingredient is from about 0.01% to about 5% by weight, about 0.01% to about 3% by weight, or 0.01%, 0.1%, 1%, or 3% by weight.
[0019] In another aspect of the present invention, methods of treating or preventing a condition associated with inflammation or oxidative stress in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of the pharmaceutical composition, such as as described above or below. The invention also relates to the compound N -((4a S,6a R,6b S,8a R,12a S,14a R,14bS)-11-cyano-2,2,6a,6b,9,9,12a -heptamethyl-10,14-dioxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,12a,14,14a,14b-octadecahydropicen-4a -yl)-2,2-difluoropropanamide (RTA or 408) or a pharmaceutically acceptable salt thereof, or a polymorphic form of such a compound (such as, for example, any of the polymorphic forms described herein above or below), or a A pharmaceutical composition comprising any of the above entities and a pharmaceutically acceptable carrier (including, for example, the pharmaceutical compositions described above), for use in treating or preventing a condition associated with inflammation or oxidative stress. The invention also relates to the use of the above-mentioned compound, polymorphic form or pharmaceutical composition for the preparation of a medicament for the treatment or prevention of a condition associated with inflammation or oxidative stress. In some modalities, the condition is associated with inflammation. In other modalities, the condition is associated with oxidative stress. In some modalities, the condition is a skin disease or disorder, sepsis, dermatitis, osteoarthritis, cancer, inflammation, autoimmune disease, inflammatory bowel disease, a complication of localized or total body exposure to ionizing radiation, mucositis, acute or chronic organ failure, liver disease, pancreatitis, an eye disorder, a lung disease, or diabetes.
The present invention further relates to the compound N -((4a S,6a R,6b S,8a R,12a S,14a R,14b S)-11-cyano-2,2,6a,6b, 9,9,12a- heptamethyl-10,14-dioxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,12a,14,14a,14b- octadecahydropicen-4a-yl)-2,2-difluoropropanamide (RTA or 408) or a pharmaceutically acceptable salt thereof, or a polymorphic form of such a compound (such as, for example, any of the polymorphic forms described herein above or below), or a pharmaceutical composition comprising any of the above entities and a pharmaceutically acceptable carrier (including, for example, the pharmaceutical compositions described above), for use in treating or preventing a condition selected from a disease or skin disorder, sepsis, dermatitis, osteoarthritis, cancer, inflammation, autoimmune disease, inflammatory bowel disease, a complication of localized or total body exposure to ionizing radiation, mucositis, acute or chronic organ failure , liver disease, pancreatitis, an eye disease, lung disease, or diabetes. Therefore, the invention relates to the use of the aforementioned compound, polymorphic form or pharmaceutical composition for the preparation of a medicament for the treatment or prevention of a condition selected from a skin disease or disorder, sepsis, dermatitis , osteoarthritis, cancer, inflammation, an autoimmune disease, inflammatory bowel disease, a complication of localized or total body exposure to ionizing radiation, mucositis, acute or chronic organ failure, liver disease, pancreatitis, an eye disorder, a lung disease, or diabetes. The invention also relates to a method of treating or preventing a condition selected from a skin disease or disorder, septicemia, dermatitis, osteoarthritis, cancer, inflammation, autoimmune disease, inflammatory bowel disease, a complication of localized exposure or total body to ionizing radiation, mucositis, acute or chronic organ failure, liver disease, pancreatitis, an eye disorder, a lung disease, or diabetes in a patient in need thereof, comprising the method of administering to the patient a therapeutically effective amount of the aforementioned compound, the polymorphic form or a pharmaceutical composition. In some modalities, the condition is a skin disease or disorder, such as dermatitis, thermal or chemical burn, a chronic wound, acne, alopecia, other hair follicle disorders, bullous epidermolysis, sunburn, complications of sunburn, skin disorders. skin pigmentation, an aging-related condition of the skin; a post-surgical wound, a scar from a skin lesion or burns, psoriasis, a dermatological manifestation of an autoimmune disease or a host versus graft disease, skin cancer, or a disorder involving hyperproliferation of skin cells. In some modalities, the skin disorder or disease is dermatitis. In some modalities, the dermatitis is allergic dermatitis, atopic dermatitis, dermatitis due to exposure to chemicals, or radiation-induced dermatitis. In other modalities, the skin disease or disorder is a chronic wound. In some modalities, the chronic wound is a diabetic ulcer, a pressure ulcer, or a venous ulcer. In other modalities, the skin disease or disorder is alopecia. In some embodiments, alopecia is selected from baldness or drug-induced alopecia. In other embodiments, the skin disease or disorder is a skin pigmentation disorder. In some modalities, the skin pigmentation disorder is vitiligo. In other embodiments, the skin disease or disorder is a disorder that involves hyperproliferation of skin cells. In some embodiments, the disorder involving hyperproliferation of skin cells is hyperkeratosis.
In other embodiments, the condition is an autoimmune disease, such as rheumatoid arthritis, lupus, Crohn's disease, or psoriasis. In other embodiments, the condition is a liver disease, such as fatty liver disease or hepatitis.
[0022] In other embodiments, the condition is an eye disease, such as uveitis, macular degeneration, glaucoma, diabetic macular edema, blepharitis, diabetic retinopathy, a disease or disorder of the corneal endothelium, postoperative inflammation , dry eye, allergic conjunctivitis or a form of conjunctivitis. In some modalities, the eye disorder is macular degeneration. In some modalities, macular degeneration is the dry form. In other embodiments, macular degeneration is the wet form. In some embodiments, the corneal endothelial disease or disorder is corneal endothelial Fuchs dystrophy.
[0023] In other embodiments, the condition is a lung disease, such as lung inflammation, pulmonary fibrosis, COPD, asthma, cystic fibrosis, or idiopathic pulmonary fibrosis. In some modalities, COPD is induced by cigarette smoking.
[0024] In other embodiments, the condition is sepsis. In other modalities, the condition is mucositis resulting from radiotherapy or chemotherapy. In some modalities, mucositis presents orally. In other modalities, the condition is associated with exposure to radiation. In some modalities, exposure to radiation leads to dermatitis. In some modalities, radiation exposure is acute. In other modalities, radiation exposure is fractional.
[0025] In other modalities, the disease is cancer. In some modalities, the cancer is a carcinoma, sarcoma, lymphoma, leukemia, melanoma, mesothelioma, multiple myeloma, or seminoma. In other modalities, cancer is of the bladder, blood, bone, brain, breast, central nervous system, cervix, colon, endometrium, esophagus, gallbladder, genital organs, genitourinary tract, head, kidney, larynx, liver, lung, muscle tissue, neck, oral or nasal mucosa, ovary, pancreas, prostate, skin, spleen, small intestine, large intestine, stomach, testis, or thyroid.
In some embodiments, the pharmaceutical composition is administered before or immediately after an individual is treated with radiation therapy or chemotherapy, wherein the chemotherapy does not comprise RTA 408 or its polymorphic forms. In some embodiments, the pharmaceutical composition is administered either before or after the individual is treated with radiation therapy or chemotherapy, or both. In some modalities, treatment reduces a side effect of radiation therapy or chemotherapy. In some modalities, the side effect is mucositis and dermatitis. In some modalities, treatment increases the effectiveness of radiation therapy or chemotherapy. In some embodiments, chemotherapy comprises administering to the patient a therapeutically effective amount of 5-fluorouracil or docetaxel.
[0027] Additional combination treatment therapy is also contemplated by this description. For example, in some embodiments, methods of treating cancer in a subject, comprising administering to the subject a pharmaceutically effective amount of a compound of the present disclosure, the methods may further comprise one or more treatments selected from from the group consisting of administering a pharmaceutically effective amount of a second drug, radiotherapy, immunotherapy, gene therapy, and surgery. In some embodiments, the methods may further comprise (1) contacting a tumor cell with the compound prior to contacting the tumor cell with the second drug, (2) contacting a tumor cell with the second drug prior to contacting the tumor cell with the second drug. contacting the tumor cell with the compound, or (3) contacting a tumor cell with the compound and the second drug at the same time. The second drug may, in certain embodiments, be an antibiotic, anti-inflammatory, anti-neoplastic, anti-proliferative, anti-viral, immunomodulatory, or immunosuppressive. In other modalities, the second drug may be an alkylating agent, androgen receptor modulator, cytoskeleton disruptors, estrogen receptor modulator, histone deacetylase inhibitor, HMG-CoA-reductase inhibitor, prenyl-protein inhibitor transferase, retinoid receptor modulator, topoisomerase inhibitor, or tyrosine kinase inhibitor. In certain embodiments, the second drug is 5-azacytidine, 5-fluorouracil, 9-cis-retinoic acid, actinomycin D, alitretinoin, all-trans-retinoic, Annamycin, Axitinib, belinostat, bevacizumab, bexarotene, bosutinib , busulfan, capecitabine, carboplatin, carmustine, CD437, cediranib, cetuximab, chlorambucil, cisplatin, cyclophosphamide, cytarabine, dacarbazine, dasatinib, daunorubicin, decitabine, docetaxel, dolastatin-10, dox-oricin, doxfluridine , epirubicin, erlotinib, etoposide, gefitinib, gemcitabine, gemtuzumab ozogamycin, hexamethylmelamine, Zavedos®, ifosfamide, imatinib, irinotecan, isotretinoin, ixabepilone, la-patinib, LBH589, lomustine, na- metho- trimethothamine, melomustine, mitomycin, mitoxantrone, MS-275, neratinib, nilotinib, nitrosourea, oxaliplatin, paclitaxel, plicamycin, procarbazine, semaxanib, semustine, sodium butyrate, sodium phenylacetate, streptozotocin, hydroxylic acid suberoylani lido, sunitinib, tamoxifen, teniposide, thiopeta, tioguanine, topotecan, TRAIL, trastuzumab, tretinoin, tricostine A, valproic acid, valrubicin, vandetanib, vinblastine, vincristine, vindesine, or vinorelbine.
[0028] Methods of treating or preventing a disease with an inflammatory component in a subject, comprising administering to the subject a pharmaceutically effective amount of a compound of the present description are also contemplated. In some modalities, the disease can be, for example, lupus or rheumatoid arthritis. In other modalities, the disease can be an inflammatory bowel disease, such as Crohn's disease or ulcerative colitis. In other embodiments, the disease with an inflammatory component can be cardiovascular disease. In other embodiments, the disease with an inflammatory component can be diabetes, such as type 1 or type 2 diabetes. In other embodiments, RTA 408, its polymorphs, and pharmaceutical compositions can also be used to treat complications associated with diabetes. . Such complications are well known to a person skilled in the art and include, but are not limited to, for example, obesity, hypertension, atherosclerosis, coronary heart disease, stroke, peripheral vascular disease, hypertension, nephropathy, neuropathy, myonecrosis, retinopathy and metabolic syndrome (syndrome X). In other embodiments, the disease with an inflammatory component can be a skin disease such as psoriasis, acne, or atopic dermatitis. Administration of RTA 408, its polymorphs, and pharmaceutical compositions in methods of treating such skin diseases can be, but are not limited to, for example, orally or topically.
[0029] In other modalities, the disease with an inflammatory component may be metabolic syndrome (syndrome X). A patient with this syndrome is characterized as having three or more symptoms selected from the following set of five symptoms: (1) abdominal obesity; (2) hypertriglyceridemia; (3) low high density lipoprotein (HDL); (4) high blood pressure; and (5) elevated fasting glucose, which may be in the range characteristic of type 2 diabetes if the patient is also diabetic. Each of these symptoms is defined in the Third Report of the National Cholesterol Education Program Expert Panel on Detection, Assessment and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III, or ATP III), National Institute of Health, 2001, NIH Publication No. 01-3670, which is incorporated herein by reference. Patients with metabolic syndrome, whether or not they have developed overt diabetes mellitus, are at increased risk of developing the micro and macrovascular complications listed above that occur with type 2 diabetes, such as atherosclerosis and coronary artery disease.
[0030] Another general method of the present description involves a method of treating or preventing a cardiovascular disease in a person, which comprises administering to the subject a pharmaceutically effective amount of a compound of the present description. In some modalities, cardiovascular disease can be, but is not limited to, for example, atherosclerosis, cardiomyopathy, congenital heart disease, congestive heart failure, myocarditis, rheumatic heart disease, valve disease, coronary artery disease, endocarditis , or myocardial infarction. Combination therapy is also contemplated for methods of treating or preventing a cardiovascular disease in a person. For example, such methods can further comprise administering a pharmaceutically effective amount of one or more cardiovascular drugs. The cardiovascular drug can be, but is not limited to, for example, a cholesterol-lowering drug, an antihyperlipidemic agent, a calcium channel blocker, an antihypertensive, or an HMG-CoA reductase inhibitor. In some embodiments, non-limiting examples of cardiovascular medications include amlodipine, aspirin, ezetimibe, felodipine, lacidipine, lercanidipine, nicardipine, nifedipine, nimodipine, nisoldipine and nitrendipine. In other embodiments, other non-limiting examples of cardiovascular medications include atenolol, bucindolol, carvedilol, clonidine, doxazosin, indoramine, labetalol, methyldopa, metoprolol, nadolol, oxprenolol, phenoxybenzamine, phentolamine, pindolol, prazosin, propranolol, terazoloin, or thiazoline . In other embodiments, the cardiovascular drug can be, for example, a statin such as atorvastatin, cerivastatin, fluvastatin, lovastatin, mevastatin, pitavastatin, pravastatin, rosuvastatin or simvastatin.
[0031] Methods of treating or preventing a neurodegenerative disease in a subject, comprising administering to the subject a pharmaceutically effective amount of a compound of the present description are also contemplated. In some modalities, neurodegenerative disease perhaps selected, for example, from the group consisting of Parkinson's disease, Alzheimer's disease, multiple sclerosis (MS), Huntington's disease, and amyotrophic lateral sclerosis. In particular embodiments, the neurodegenerative disease is Alzheimer's disease. In certain modalities, the neurodegenerative disease is MS, such as progressive, secondary relapsing progressive MS, or primary relapsing-remitting progressive. In some modalities, the individual can be, for example, a primate. In some modalities, the individual can be a human being.
In particular embodiments of methods of treating or preventing a neurodegenerative disease in an individual, comprising administering to the individual a pharmaceutically effective amount of a compound of the present description, the treatment suppresses demyelination of neurons in the brain. of the individual, or spinal cord. In certain modalities, the treatment suppresses inflammatory demyelination. In certain modalities, the treatment suppresses the transect of axons from neurons in the individual's brain, or spinal cord. In certain modalities, the treatment suppresses the transection of neurites into the individual's brain or spinal cord. In certain modalities, the treatment inhibits neuronal apoptosis in the individual's brain, or spinal cord. In certain modalities, the treatment stimulates the remyelination of axons from neurons in the individual's brain, or spinal cord. In certain modalities, the treatment restores function lost after an MS attack. In certain modalities, the treatment prevents a new MS attack. In certain modalities, treatment prevents a disability resulting from an MS attack.
[0033] A general aspect of the present description contemplates a method of treating or preventing a disorder characterized by overexpression of iNOS genes in an individual, comprising administering to the individual a pharmaceutically effective amount of RTA 408, polymorphic forms, or a pharmaceutical composition of the present description.
[0034] Another general aspect of the present description contemplates a method of inhibiting IFN-Y-induced nitric oxide production in cells of an individual, comprising administering to said individual a pharmaceutically effective amount of RTA 408, polymorphic forms, or a pharmaceutical composition of the present description.
Yet another general method of the present description comprises a method of treating or preventing a disorder characterized by overexpression of COX-2 genes in an individual, comprising administering to the individual a pharmaceutical amount. effective RTA 408, polymorphic forms, or a pharmaceutical composition of the present description.
[0036] Methods of treating kidney/kidney disease (RKD) in a subject, comprising administering to the subject a pharmaceutically effective amount of a compound of the present description are also contemplated. See US Patent 8,129,429, which is incorporated herein by reference. RKD can result, for example, in a toxic assault. Toxic aggression can result from, but is not limited to, for example, an imaging agent or a drug. The drug can be a chemotherapeutic agent, for example. RKD can result from an ischemia/reperfusion injury in certain modalities. In certain modalities, RKD results from diabetes or hypertension. In some modalities, RKD can result from an autoimmune disease. RKD can be further defined as acute RKD or chronic RKD.
In certain methods of treating kidney/kidney disease (RKD) in a subject, comprising administering to the subject a pharmaceutically effective amount of a compound of the present disclosure, the subject has undergone or is undergoing dialysis. In certain embodiments, the individual has undergone or is a candidate for kidney transplantation. The individual could be a primate. The primate can be a human being. The object of this or any other method can be, for example, a cow, horse, dog, cat, pig, rat, mouse or guinea pig.
[0038] Also contemplated by the present description is a method for improving glomerular filtration rate or creatinine clearance in an individual, comprising administering to the individual a pharmaceutically effective amount of RTA 408, polymorphic forms, or a pharmaceutical composition thereof description.
[0039] In some embodiments, the pharmaceutical composition is administered in a single dose per day. In other embodiments, the pharmaceutical composition is administered in more than one dose per day. In some embodiments, the pharmaceutical composition is administered in a pharmaceutically effective amount.
In some embodiments, the dose is from about 1 mg/kg to about 2000 mg/kg. In other embodiments, the dose is from about 3 mg/kg to about 100 mg/kg. In other embodiments, the dose is about 3, 10, 30, or 100 mg/kg.
[0041] In other modalities, the pharmaceutical composition is administered topically. In some embodiments, topical administration is administered to the skin. In other modalities, topical administration is administered to the eye.
[0042] In other modalities, the pharmaceutical composition is administered orally. In other embodiments, the pharmaceutical composition is administered intraocularly.
[0043] Other objects, features and advantages of the present description will become apparent from the following detailed description. It should be understood, however, that the detailed description and specific examples, while indicating specific embodiments of the invention, are given by way of illustration only, as various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description. Note that just because a particular compound is assigned to a particular generic formula does not mean that it cannot also belong to another generic formula. BRIEF DESCRIPTION OF THE DRAWINGS
[0044] The following figures form part of the present specification and are included to better demonstrate certain aspects of the present description. One or more German words can be found in the figures, including "Masseanderung" and "temperatur", which means "change in mass" and "temperature", respectively. The invention may be better understood by reference to one of these figures in combination with the detailed description of specific embodiments presented herein.
[0045] FIG. 1 - Effect of RTA 408 on IFNY-induced nitric oxide production and cell viability in RAW264.7 cells.
[0046] FIGS. 2a & b - Effect of RTA 408 on antioxidant response element (ARE) activation: (a) NQO1-ARE luciferase activity; (B) GSTA2-ARE luciferase activity.
[0047] FIGS. 3a-f - Nrf2 GST ARE higher relative after cell phone treatment (a) RTA 402; (B) 63,415 (RTA 408); (C) 63170; (D) 63171; (E) 63179; and (f) 63189. The graphs also show cell viability as tested using WST1 cell proliferation reagent and measuring absorbance after 1 hour. All drugs were administered in DMSO and cells were cultured at 10,000 cells/well in 384-well plates in low glucose DMEM supplemented with 10% FBS, 1% Penicillin Streptomycin, and 0.8 mg/mL geneticin .
[0048] FIGS. 4a-d - Effect of RTA 408 on Nrf2 target gene expression in HFL1 lung fibroblasts. (A) NQO1; (B) HMOX1; (C) GCLM; (D) TXNRD1.
[0049] FIGS. 5a-d - Effect of RTA 408 on Nrf2 target gene expression in BEAS-2B bronchial epithelial cells. (A) NQO1; (B) HMOX1; (C) GCLM; (D) TXNRD1.
[0050] FIGS. 6 a & b - Effect of RTA 408 on NRF2 target protein levels. (A), SH-SY5Y cells; cells (b) BV2.
[0051] FIG. 7 - Effect of RTA 408 on NQO1 enzymatic activity in RAW264.7 cells.
[0052] FIG. 8 - Effect of RTA 408 on total glutathione levels in the AML-12 hepatocyte cell line.
[0053] FIG. 9 - Effect of RTA 408 on WST-1 as a NADPH absorbance marker.
[0054] FIGS. 10 a-d - Effect of RTA 408 on the expression of genes involved in NADPH synthesis. (A) H6PD; (B) PGD; (C) TKT; (D) ME1.
[0055] FIGS. 11a & b - (a) Effect of RTA 408 on TNF-α induced activation of an NF-kB luciferase reporter in the NIH3T3 mouse cell line with overlapping WST1 viability and WST1/2 viability. (B) Activation of an NF-kB luciferase reporter in the TNF-α-induced NIH3T3 mouse cell line. The graph shows the relative double change as a function of the change of record in Concentration RTA 408.
[0056] FIG. 12 - Effect of RTA 408 on TNF-α induced activation of luciferase reporter construct NF-kB.
[0057] FIGS. 13a & b - (a) Effect of RTA 408 on TNF-α induced activation of an NF-kB luciferase reporter in the human cell line A549 with overlapping WST1 viability and WST1/2 viability. (B) of TNF-α induced by activation of an NF-kB luciferase reporter in the human A549 cell line. The graph shows the relative double change as a function of the change of record in Concentration RTA 408.
[0058] FIG. 14 - Effect of RTA 408 on TNF-α induced phosphorylation of iKBα.
[0059] FIGS. 15a-d - Effect of RTA 408 on transaminase gene expression: (a) ALT1 (GPT1); (B) ALT2 (GPT2); (C) AST1 (GOT1); (D) AST1 (GOT2). Asterisks indicate a statistically significant difference between the control group (*P < 0.05; ** P < 0.01).
[0060] FIG. 16 - Effect of RTA 408 on pyruvate levels in cultured muscle cells (*P < 0.05).
[0061] FIG. 17 - Effect of 63415 in a model of LPS-mediated pulmonary inflammation (% change in pro-inflammatory cytokines in relation to LPS treatment). Compound 63415 was administered QD x 3 at time 0, 24, and 48 h, followed by LPS one hour after the last dose of 63415 in female BALB/c mice. Animals were sacrificed 20 hours after LPS administration. BAL was analyzed for the expression of proinflammatory cytokines. Compound 63415 reduced proinflammatory cytokines in a dose-dependent manner, with maximum reductions ranging between 50% - 80% in TNF-α, IL-6 and IL-12.
[0062] FIGS. 18a & b - Effect of RTA 408 on LPS-induced pulmonary inflammation in mice. (A) inflammatory cytokines; goals (b) Nrf2. RTA 408 was administered to female BALB/c mice (n = 10) QD x 6 at time 0, 24, 48, 72, 96 and 120 h followed by LPS in 121 h with animals sacrificed in 141 h. Pro-inflammatory cytokine protein expression assayed in FLBA. Nrf2 biomarkers tested in the lung. Asterisks indicate a statistically significant difference from the saline control group (*P<0.05; **P<0.01; ***P<0.001).
[0063] FIGS. 19a & b - Effect of 63,415 on BALF infiltrates on bleomycin-induced lung inflammation: (a) BAL cell fluid count; (B) body weight. Compound 63415 was administered QD x 39 on days -10 to 28 in C57BL/6 mice. Bleomycin was given on day 0. Daily weights were obtained. Cell counts were obtained from the BAL sacrifice. A notable reduction in the inflammatory infiltrate was observed. No significant improvements were seen in the chronic inflammation score, interstitial fibrosis, or the number of fibrotic foci.
[0064] FIGS. 20a & b - Effect of RTA 408 on bleomycin-induced pulmonary fibrosis in rats: (a) PMN; (B) Hydroxyproline. Asterisks indicate a statistically significant difference from the bleomycin control group (*P < 0.05).
[0065] FIG. 21 - Effect of RTA 408 on Nrf2 target enzymes in lungs of rats with bleomycin-induced pulmonary fibrosis. Asterisks indicate a statistically significant difference from the saline control group (*P<0.05; **P<0.01; ***P<0.001).
[0066] FIGS. 22a-e - Effect of RTA 408 on smoke-induced COPD cigarette in mice. (A), KC; (B) IL-6; (C) TNF-α; (D) IFN-Y; (E) to RANTES. RTA 408 (63415) was tested at dose levels of 3 mg/kg (low), 10 mg/kg (half), and 30 mg/kg (high). An AIM analogue (63355) was tested in the same study for comparison. Asterisks indicate a statistically significant difference between the CS control group.
[0067] FIG. 23 - Effect of RTA 408 on Nrf2 target enzymes in lungs of rats with tobacco-induced COPD cigarette. Asterisks indicate a statistically significant difference from the saline control group (*P<0.05; **P<0.01; ***P<0.001). Daggers represent a statistically significant difference in rats exposed to cigarette smoke and administered vehicle (f P < 0.05).
[0068] FIGS. 24a-d - Effects of 63415 (RTA 408) on body weight in a BALB/c rat model of sepsis. LPS was administered to all animals on Day 0. (a) Body Weight: 63,415 (RTA 408); (B) Body Weight: RTA 405; (C) Systemic LPS: % Survivor: 63,415 (RTA 408); (D) Systemic LPS: % Survivor: RTA 405 Both RTA 405 and 63,415 (RTA 408) were administered QD x 5 on days -2 to 2 Compound 63,415 (RTA 408) improved survival. Body weight as a function of time in animals treated 63415 BALB mice serves as a model for sepsis.
[0069] FIG. 25 - Effect of 63415 in a radiation-induced oral mucositis model. RTA 405 or 63415 (RTA 408) was administered BID x 20 on days -5 to -1 and days 1 to 15 of male golden Syrian hamsters. Radiation occurred on day 0 Mucositis grades range from 0 to 5 based on clinical manifestations (0: completely healthy; 12: light to severe erythema, 3-5: different degrees of ulceration). 63415 improved mucositis by 30 mg/kg and 100 mg/kg, with up to a 36% reduction in ulcerations.
[0070] FIG. 26 - Effect of 63415 on Nrf2 target gene induction in a 14-day rat toxicity study in C57BL/6 mice. NRF2 target gene mRNAs were evaluated in livers of PO QDx14 treated rats. Substantial increases in mRNA expression for several NRF2 target genes were observed and were consistent with tissue exposure.
[0071] FIGS. 27a & b - Effect of 63,415 on induction of the Nrf2 target gene in rat livers: (a) The target genes; (B) the negative regulators. NRF2 target gene mRNAs were evaluated in livers of PO QD x 14 treated rats.
[0072] FIGS. 28a & b - Effect of 63,415 on NRF2 target genes in monkey tissues: (a) liver; (B) pulmonary. NRF2 gene target mRNAs were evaluated in monkeys treated PO QD x 14 using Panomics QuantiGene® 2.0 Plex technology.
[0073] FIGS. 29 a & b - Effect of 63,415 on Nrf2 target enzyme activity in rat liver: (a) NQO1 activity; (B) GST activity. Nrf2 target enzyme activity was evaluated in livers of PO QD x 14 treated rats. NQO1 and GST enzyme activities were induced in a dose-dependent manner.
[0074] FIGS. 30a & b - Effect of 63415 on Nrf2 target enzyme activity in rat liver: (a) NQO1 activity; (B) GST activity. Nrf2 target enzyme activity was evaluated in livers of PO QD x 14 treated rats. NQO1 and GST enzyme activities were induced in a dose-dependent manner.
[0075] FIG. 31a & b - Effects of 63,415 on Nrf2 inducing target enzyme activity in various tissues of cynomolgus monkeys: (a) NQO1 activity; (B) GSR activity.
[0076] FIGS. 32a & b - RTA 408 concentration in rat liver, lung and brain, and NQO1 activity in mouse liver after 14 days of daily oral administration. (A) Tissue distribution of RTA 408 in rats after 14 days of daily oral administration. Data represent the mean ± SD of RTA 408 tissue concentrations collected 4 h after the final dose of the study. The numbers above the error bars represent the mean of. (B) Correlation of mouse liver RTA 408 content with NQO1 enzymatic activity. Individual mouse liver RTA 408 liver content was plotted against individual enzyme activity from this report.
[0077] FIGS. 33a & b - RTA 408 concentration in rat plasma, liver, lung, and brain, and NQO1 activity in rat liver after 14 days of daily oral administration. (A) Tissue distribution of RTA 408 in rats after 14 days of daily oral administration. Data represent the mean ± SD of RTA 408 tissue concentrations collected 4 h after the final dose of the study. The numbers above the error bars represent the mean of. *Two values were excluded from the mean calculation due to being outliers, defined as values causing the dataset to fail the Shapiro-Wilk normality test. (B) Correlation of rat liver RTA 408 content with NQO1 enzymatic activity. Individual rat liver RTA 408 content was plotted against individual enzyme activity from this report. Tissues from the RTA 408/kg 100 mg dose group were collected on day 6, and the toxicities observed in this group prevented NQO1 liver enzyme activity assessments.
[0078] FIGS. 34a & b - Effect of treatment on 63,415 Nrf2 activation in monkey PBMC: (a) PBMC NQO1 versus Plasma Concentration; (B) pulmonary NQO1 against PBMC NQO1.
[0079] FIG. 35 - Summary of the 63,415 14-day toxicity study in monkeys. All doses were well tolerated with no adverse clinical signs. Clinical chemistry data suggests no obvious toxicity.
[0080] FIG. 36 - Effect of dosing time on plasma concentration of RTA 408 after topical and oral ocular. The plasma concentration of RTA 408 was also measured after 5 days of daily topical ocular administration of RTA 408 and determined to remain relatively constant from measurements taken after the first day.
[0081] FIGS. 37a & b - Correlation of exposure to RTA 408 in monkey plasma with expression of NQO1 and SRXN1 mRNA in PBMC: (a) NQO1; (B) SRXN1.
[0082] FIG. 38 - Concentration of RTA 408 in various tissues and fluids within the eye as a function of time after 5 days of topical ocular application. RTA 408 plasma concentration was also measured after topical ocular administration.
[0083] FIG. 39 - Effect of RTA 408 on the incidence of grade 3 dermatitis caused by acute radiation exposure to different concentrations of topically administered RTA 408.
[0084] FIG. 40 - Effect of RTA 408 on the incidence of grade 2 dermatitis caused by exposure to acute radiation during a 30-day course of treatment to different concentrations of topically administered RTA 408.
[0085] FIG. 41 - Effect of RTA 408 on the incidence of grade 2 dermatitis caused by exposure to acute radiation during a 28-day course of treatment at different concentrations of orally administered RTA 408.
[0086] FIGS. 42a & b - (a) Area under the analysis curve of clinical dermatitis score as a function of time for each of the different control groups, including all animals used in the trial. (B) Area under analysis of the clinical dermatitis score curve as a function of duration than the score for each of the different control groups, including only those animals that completed the entire 30 day trial.
[0087] FIG. 43 - Mean acute radiation dermatitis score as a function of time for untreated, treated without radiation exposure, vehicle only and three oral amounts of RTA 408 at 3, 10 and 30 mg/kg. The dermatitis score was based on a scale that 0 is completely healthy, 1-2 exhibited mild to moderate erythema with minimal to slight scaling, 3-4 exhibited moderate to severe erythema and scaling, and 5 exhibited a frank ulcer.
[0088] FIG. 44 - The mean acute radiation dermatitis score as a function of time for untreated, untreated with no radiation exposure, vehicle only, and three oral values of RTA 408 at 3, 10, and 30 mg/kg, measured each two days from day 4 to day 30. Dermatitis score was based on a scale that 0 is completely healthy, 1-2 exhibited mild to moderate erythema with slight scaling, 3-4 exhibited moderate to severe erythema and scaling, and 5 exhibited a frank ulcer.
[0089] FIG. 45 - mean acute radiation dermatitis score as a function of time for untreated, treated without radiation exposure, single vehicle, and three current RTA 408 values at 0.01%, 0.1% and 1% measured every two days from day 4 to day 30. Dermatitis score was based on a scale that 0 is completely healthy, 1-2 exhibited mild to moderate erythema with minimal to slight scaling, 3-4 exhibited moderate to severe erythema and scaling, and 5 exhibited a frank ulcer.
[0090] FIG. 46 - Clinical fractional radiation dermatitis scores plotted as a function of time and changes in dermatitis score for each test group. The dermatitis score was based on a scale that 0 is completely healthy, 1-2 exhibited mild to moderate erythema with minimal to slight scaling, 3-4 exhibited moderate to severe erythema and scaling, and 5 exhibited a frank ulcer.
[0091] FIG. 47 - AUC analysis data shows the dermatitis score (severity x days) for each of the test groups over the entire observation period. Dermatitis scores were assessed every other day from day 4 to day 30 of the study.
[0092] FIGS. 48a & b - (a) Absorbance graph at 595 nm for the LNCaP treated prostate cancer cell line showing cytotoxic effect towards cells treated with a chemotherapeutic agent and RTA 408 versus RTA 408 alone. (B) Absorbance graph at 595 nm for the DU-145 prostate cancer cell treatment line, showing the cytotoxic effect against cells treated with a chemotherapeutic agent and RTA 408 versus RTA 408 alone.
[0093] FIG. 49 - black and white versions of color photographs of photographed mice showing tumor luciferase activity for three mice: an untreated control animal, an animal subjected to ionizing radiation only (single dose of 18 Gy), and an animal who received ionizing radiation (single dose of 18 Gy, day 0) and RTA 408 (17.5 mg/kg ip, once a day on days -3 to -1, then single doses on days 1, 3 and 5) . The colors indicated by the arrows are indicative of intensity with the intensities being represented by red, yellow, green and blue in order from highest to lowest intensity.
[0094] FIG. 50 - Reduction of aqueous humor protein concentrations for different formulations of RTA 408 (dark bars) compared to literature values for MaxiDex® (0.1% dexamethasone) and mapracorat (light bars), after paracend induction - thesis.
[0095] FIG. 51 - Dose-dependent suppression of NO in vivo by 63415. CD-1 mice (n = 6) were treated with DMSO or MA by oral gavage. LPS (5 mg/kg) was administered 24 hours later. Twenty-four hours after LPS administration, whole blood was collected for NO assay. NO inhibition was determined by the Griess reaction of reduced deproteinated plasma.
[0096] FIG. 52 - Wide distribution of 63415 (RTA 408) in mouse tissues. Mice received PO QD x 3 doses with 25 mg/kg 63415 (RTA 408) or 25 mg/kg RTA 405 blood (plasma and whole blood) and tissues (brain, liver, lung, and kidney) were collected 6 hours later at last dose. Semiquantitative analysis of drug content was performed. Significant levels were observed in the CNS.
[0097] FIG. 53 - induction of NQO1 activity in rat liver, lung, kidney and by 63415. Mice received PO QD x 3 doses at 25 mg/kg. Tissues were collected 6 hours after the last dose, and analysis of NQO1 activity was performed. Significant activation of NQO1 was observed in several tissues.
[0098] FIG. 54 - Summary of 14-day mouse toxicity study 63,415. C57BL/6 mice were administered PO QD x 14. Endpoints included survival, weight and clinical biochemical analyses. All animals survived to day 14. There are no significant weight changes compared to the vehicle group, and there was no evidence of toxicity at any dose based on clinical biochemistry.
[0099] FIG. 55 tissue distribution of 63,415 from 14-day mouse toxicity study in C57BL/6 mice -. Brain, lung, liver and samples were collected 4 h after the final dose and quantified to 63415 content using sensitive LC/MS/MS. Exposures at 10 and 100 mg/kg in the lung exceeded the in vitro IC 50 for the induction of NO and 55 to 1138-fold, respectively. The exposure at 10 and 100 mg/kg in the brain exceeded the in vitro IC 50 for NO by induction and 29 and 541 times, respectively.
[00100] FIG. 56 tissue distribution of 63,415 in Sprague Dawley rats -. Tissues were collected four hours after final dosing, on day 14 or day 6 (100 mg/kg), extracted and quantified for 63,415 content using a sensitive LC/MS/MS method. Compound 63415 distributes well to target tissues. Exposures at 10 mg/kg in lung and brain exceed the in vitro IC 50 for NO inhibition by 294- and 240-fold, respectively.
[00101] FIG. 57 - Target tissue distribution of compound 63415 in cynomolgus monkeys. Tissues were collected four hours after final dosing, on day 14 Compound 63415 content was extracted and quantified using a sensitive LC/MS/MS.
[00102] FIG. 58 - FT-Raman spectrum (3400-50 cm -1) of sample PP415-P1, which corresponds to the amorphous form (class 1).
[00103] FIG. 59 - PXRD (1.5-55.5 °2θ) standard from sample PP415-P1, which corresponds to the amorphous form (class 1).
[00104] FIG. 60 - Thermogram TG-FTIR (25-350°C) of sample PP415-P1, which corresponds to the amorphous form (class 1).
[00105] FIG. 61 - 1H-NMR spectrum in DMSO-d6 of sample PP415-P1, which corresponds to the amorphous form (class 1).
[00106] FIG. 62 - DSC thermogram of sample PP415-P1, which corresponds to the amorphous form (class 1).
[00107] FIG. 63 - Isothermal DVS of sample PP415-P1, which corresponds to the amorphous form (class 1).
[00108] FIG. 64 - FT-Raman spectrum of sample PP415-P1, which corresponds to the amorphous form (class 1), after the DVS measurement (upper) is unchanged compared to the material before the DVS measurement (lower part). Spectra were scaled and shifted in the y direction for comparison purposes.
[00109] FIG. 65 - PXRD pattern of sample PP415-P1, which corresponds to the amorphous form (class 1), after the DVS measurement (upper) is unchanged compared to the material before the DVS measurement (lower part). Patterns are not scaled, but are offset in the y direction for comparison purposes.
[00110] FIG. 66 - PXRD pattern of sample PP415-P40 (top) corresponds to pattern of solvate form (Class 2) (bottom, sample PP415-P19). Patterns have been scaled and offset in the y direction for comparison purposes.
[00111] FIG. 67 PXRD patterns from stability samples PP415-P2a (top), PP415-P3a (2nd from top), PP415-P4a (middle), and PP415-P5a (2nd from bottom), which corresponds to the shape amorphous (Class 1), after one week show no differences compared to the starting material, at time point t0 (bottom, sample PP415-P1). Patterns are not scaled, but are offset in the y direction for comparison purposes.
[00112] FIG. 68 PXRD patterns of stability samples PP415-P2b (top), PP415-P3b (2nd from stop), PP415-P4b (middle), and PP415-P5b (2nd from bottom), which corresponds to the shape amorphous (Class 1), after two weeks show no differences compared to the starting material at the t0 time point (bottom, sample PP415-P1). Patterns are not scaled, but are offset in the y direction for comparison purposes.
[00113] FIG. 69 PXRD patterns from stability samples PP415-P2c (top), PP415-P3c (2nd from top), PP415-P4C (middle), and PP415-P5c (2nd from bottom), which corresponds to the shape amorphous (Class 1), after four weeks show no differences compared to the starting material at the t0 time point (bottom, sample PP415-P1). Patterns are not scaled, but are offset in the y direction for comparison purposes.
[00114] FIG. 70 - FT-Raman spectra (2400-50 cm -1) of the samples in solvate form (Class 2) (PP415-P7: top; PP415-P8: 2nd from top; PP415-P9: 3rd from top; PP415 -P10 : 4th from above; PP415-P11: medium; PP415-P15: 4th from below; PP415-P17: 3rd from below; PP415-P21: 2nd from below; PP415-P24: low). Spectra were scaled and shifted in the y direction for comparison purposes.
[00115] FIG. 71 - FT-Raman spectrum (1750-1000 cm -1) of the solvate form (Class 2) (PP415-P7: top) clearly differs from the spectrum of the amorphous form (class 1) (PP415-P1: lower). Spectra were scaled and shifted in the y direction for comparison purposes.
[00116] FIG. 72 FT-Raman spectra (1750-1000 cm - -1) of class 2 (sample PP415-P19: upper), class 3 (sample PP415-P6: 2nd from above), class 4 (sample PP415-P13: 2nd from below), and class 5 (sample PP415-P14: bottom) differ significantly from each other. Spectra were scaled and shifted in the y direction for comparison purposes.
[00117] FIG. 73 - PXRD standards (2-32 °2θ) of solvate form samples (Class 2) (PP415-P7: top; PP415-P8: 2nd from top; PP415- P10: 3rd from top; PP415-P15: 4th from above; PP415-P17: medium; PP415-P18: 4th from below; PP415-P19: 3rd from below; PP415-P21: 2nd from below; PP415-P24: low). Patterns have been scaled and offset in the y direction for comparison purposes.
[00118] FIG. 74 - PXRD patterns (11-21 °2θ) of some samples of solvate form (class 2) (PP415-P7: top; PP415-P8: 2° from top; PP415-P10: medium; PP415-P21: 2° from below; PP415-P24: low). Patterns have been scaled and offset in the y direction for comparison purposes.
[00119] FIG. 75 PXRD standards (2-32 °2θ) of class 2 (sample PP415-P19: top), class 3 (sample PP415-P6: 2nd from top), class 4 (sample PP415-P13: 2nd from bottom) and class 5 (sample PP415- P14: from the bottom) are very different. Patterns have been scaled and offset in the y direction for comparison purposes.
[00120] FIG. 76 - TG-FTIR thermogram of sample PP415-P7, which corresponds to a solvate form (Class 2).
[00121] FIG. 77 - TG-FTIR thermogram of sample PP415-P21, which corresponds to a solvate form (Class 2).
[00122] FIG. 78 - TG-FTIR thermogram of sample PP415-P24, which corresponds to a solvate form (Class 2).
[00123] FIG. 79 - TG-FTIR thermogram of sample PP415-P29, which corresponds to a solvate form (Class 2).
[00124] FIG. 80 - TG-FTIR thermogram of sample PP415-P47, which corresponds to a solvate form (Class 2).
[00125] FIG. 81 - TG-FTIR thermogram of sample PP415-P48, which corresponds to a solvate form (Class 2).
[00126] FIG. 82 - The FT-Raman (1800-700 cm -1) spectra of the solvate form (Class 2) (below, the sample PP415-P7) and the dry solvate form (Class 2) (above, the sample PP415-P30) are similar and show only small differences that can hardly be distinguished within the graph. Spectra are scaled for comparison purposes.
[00127] FIG. 83 PXRD pattern of dry solvate form (class 2), sample PP415-P30 (top), compared to standard solvate form (class 2), sample PP415-P7 (bottom) -. Patterns are not scaled, but are offset in the y direction for comparison purposes.
[00128] FIG. 84 - TG-FTIR thermogram of dry sample PP415-P30, which corresponds to a solvate form (Class 2).
[00129] FIG. 85 - FT-Raman spectrum of dry sample PP415-P18 (light gray) is identical to spectrum of original sample PP415-P15 (dark grey) are similar and show only small differences that cannot be distinguished in the graph. Spectra were scaled for comparison purposes.
[00130] FIG. 86 - PXRD pattern from dry sample PP415-P18 (top) shows small differences from original sample pattern PP415-P15 (bottom), despite both forms of solvates (Class 2). Patterns have been scaled and offset in the y direction for comparison purposes.
[00131] FIG. 87 - TG-FTIR thermogram of sample PP415-P18, which corresponds to a solvate form (Class 2).
[00132] FIG. 88 - FT-Raman spectrum of sample PP415-P17 (top) is almost identical to the spectrum of dry samples PP415-P19 (middle) and PP415-P32 (bottom) and show only small differences that cannot be distinguished inside the graph. Spectra were scaled and shifted in the y direction for comparison purposes.
[00133] FIG. 89 - PXRD pattern of dry sample PP415-P19 (middle) is different from the pattern of original sample PP415-P17 (top), but still corresponds to the class 2 shape. The pattern of sample PP415-P32 is still dry (bottom ) shows larger peaks with a low S/N ratio. The crystalline material is smaller, but still matches the Class 2 shape. Patterns have been scaled and shifted in the y direction for comparison purposes.
[00134] FIG. 90 - TG-FTIR thermogram of sample PP415-P19, which corresponds to a solvate form (Class 2).
[00135] FIG. 91 - TG-FTIR thermogram of sample PP415-P32A, which corresponds to a solvate form (Class 2).
[00136] FIG. 92 - FT-Raman spectrum of sample PP415-P21 (top) is identical to the spectra of dry samples PP415-P28 (middle) and PP415-P34 (bottom). Spectra were scaled and shifted in the y direction for comparison purposes.
[00137] FIG. 93 PXRD patterns from dried samples PP415-P28 (middle) and PP415-P34 (bottom) show larger peaks with a lower S/N ratio, indicating a lower crystallinity of the samples compared to the original sample PP415-P21 pattern - (top). The patterns are slightly different, but still correspond to form class 2. They have been scaled and offset in the y direction for comparison purposes.
[00138] FIG. 94 - TG-FTIR thermogram of dry sample PP415-P28, which corresponds to a solvate form (Class 2).
[00139] FIG. 95 - TG-FTIR thermogram of dry sample PP415-P34, which corresponds to a solvate form (Class 2).
[00140] FIG. 96 - FT-Raman spectra (2400-50 cm-1) of the samples in solvate form (Class 3) (PP415-P6: top; PP415-P12: medium; PP415-P20: low). Spectra were scaled and shifted in the y direction for comparison purposes.
[00141] FIG. 97 - FT-Raman spectra (1750-1000 cm-1) of the samples in solvate form (Class 3) (PP415-P6: top; PP415-P12: 2nd from top; PP415-P20: 2nd from bottom) are very similar to each other with only small differences, eg in ~1,690 cm-1, but is clearly different from class 1 (PP415-P1: lower). Spectra were scaled and shifted in the y direction for comparison purposes.
[00142] FIG. 98 PXRD standards (2-32 °2θ) from the solvate form samples (Class 3) (PP415-P6: top; PP415-P12: medium; PP415-P20: low). Patterns have been scaled and offset in the y direction for comparison purposes.
[00143] FIG. 99 - PXRD standards (13.5-18.5 °2θ) of the solvate form samples (Class 3) (PP415-P6: top; PP415-P12: medium; PP415-P20: bottom) show small differences. Patterns have been scaled and offset in the y direction for comparison purposes.
[00144] FIG. 100 - TG-FTIR thermogram of sample PP415-P6, which corresponds to the solvate form (class 3).
[00145] FIG. 101 - TG-FTIR thermogram of sample PP415-P12, which corresponds to the solvate form (class 3).
[00146] FIG. 102 - TG-FTIR thermogram of dry solvate form (class 3), sample PP415-P25.
[00147] FIG. 103 - TG-FTIR thermogram of the additionally dry solvate form (class 3), sample PP415-P33.
[00148] FIG. 104 - The FT-Raman spectra (1800-700 cm-1) of the solvate form (class 3) (above, sample PP415-P6), of the dry solvate form (class 3) (medium, sample PP415- P25), and otherwise the dry solvate (class 3) (below, sample PP415-P33) are identical. Spectra were scaled and shifted in the y direction for comparison purposes.
[00149] FIG. 105 PXRD standards (4-24 °2θ) of solvate form (class 3) (top, sample PP415-P6), dry solvate form (class 3) (medium, sample PP415-P25), and still dry solvate form (class 3) (below, sample PP415-P33). Patterns have been scaled and offset in the y direction for comparison purposes.
[00150] FIG. 106 - TG-FTIR thermogram of sample PP415-P13, which corresponds to a form of acetonitrile solvate (class 4).
[00151] FIG. 107 - FT-Raman spectra (1800-700 cm-1) of the acetonitrile solvate form (class 4) (dark gray, sample PP415-P13) and of the dry material of an acetonitrile solvate form (class 4) (light grey, sample) PP415-P26) are identical and overlap perfectly. Spectra were scaled for comparison purposes.
[00152] FIG. 108 PXRD standard of dry acetonitrile solvate form (grade 4), sample PP415-P26 (bottom), compared to reference standard of acetonitrile solvate form (grade 4), sample PP415-P13 (top) . Patterns have not been scaled but have been offset in the y direction for comparison purposes.
[00153] FIG. 109 - TG-FTIR thermogram of dry acetonitrile solvate form (class 4), sample PP415-P26.
[00154] FIG. 110 - FT-Raman spectra (1800-700 cm-1) of the acetonitrile solvate form (class 4) (upper, sample PP415-P35), and of the dry acetonitrile solvate form (class 4) (medium, sample PP415-P36 and lower , sample PP415-P37) correspond to each other. Spectra were scaled and shifted in the y direction for comparison purposes.
[00155] FIG. 111 PXRD standards (4-24 °2θ) of the acetonitrile solvate form (grade 4) (top, sample PP415-P35) and of the dry acetonitrile solvate form (grade 4) (medium, sample PP415-P36 and bottom , PP415-P37 of the sample) agree with each other. Patterns have been scaled and offset in the y direction for comparison purposes.
[00156] FIG. 112 - TG-FTIR thermogram of dry acetonitrile solvate form (class 4), sample PP415-P36.
[00157] FIG. 113 - TG-FTIR thermogram of dry acetonitrile solvate form (class 4), sample PP415-P37.
[00158] FIG. 114 - Isothermal DVS of the desolvated acetonitrile solvate form (class 4) sample PP415-P37).
[00159] FIG. 115 - PXRD standard from sample PP415-P37, a form of acetonitrile solvate (class 4), after DVS measurement (bottom) remains unchanged compared to the material before the DVS measurement (top). Patterns are not scaled, but are offset in the y direction for comparison purposes.
[00160] FIG. 116 - DSC thermogram of the desolvated acetonitrile solvate form (class 4) (sample PP415-P37).
[00161] FIG. 117 - DSC thermogram of the amorphous form ~ 1:1 (Class 1), sample PP415-P1, with the desolvated acetonitrile solvate form (class 4), sample PP415-P36.
[00162] FIG. 118 - DSC thermogram of a 1:1 mixture of the amorphous form (Class 1), sample PP415-P1, with the desolvated acetonitrile solvate form (Class 4), sample PP415-P36 (experiment number: PP415-P39). The warm-up check (Step 1) was stopped for 30 minutes at 173°C (Step 2) and then resumed (Step 3).
[00163] FIG. 119 - TG-FTIR thermogram of sample PP415-P14, which corresponds to a form of THF solvate (class 5).
[00164] FIG. 120 - FT-Raman spectra (1800-1100 cm-1) of a THF solvatar form (Class 5) (dashed line, sample PP415-P14), dried material of a THF solvatar form (Class 5) (dotted line, sample PP415 -P27), and of the amorphous form (class 1) (solid line, sample PP415-P1). The spectra have been scaled for comparison purposes and show small changes in magnitude but little corresponding change in spectral form.
[00165] FIG. 121 - PXRD standard of dry THF solvate form (class 5), sample PP415-P27 (top), compared to THF solvate standard (class 5), sample PP415-P14 (bottom). Patterns are not scaled, but are offset in the y direction for comparison purposes.
[00166] FIG. 122 - TG-FTIR thermogram of sample PP415-P27, which corresponds to a dry THF solvate (class 5).
[00167] FIG. 123 - PXRD pattern of sample PP415-P41 (top) corresponds to the pattern of the solvate form of THF (Class 5) (middle, sample PP415-P14) and not for the pattern of heptane solvate form, (Class 2) ( background, try PP415-P19). The patterns were scaled and shifted in the y direction for comparison purposes.
[00168] FIG. 124 - PXRD pattern of sample PP415-P45 (top) corresponds to the pattern of the THF solvate form (Class 5) (middle, sample PP415-P14) and not for the heptane pattern of the solvate form (Class 2) (Bottom, PP415-P19 of the sample). Patterns have been scaled and offset in the y direction for comparison purposes.
[00169] FIG. 125 - PXRD pattern of sample PP415-P41 (top) corresponds to a form of THF solvate (class 5). After drying the PP415-P41 sample for 1 day (2° from above, the sample: PP415-P44), the material is essentially amorphous. Some broad peaks with low intensity remain. After more drying overnight (2° from bottom, sample PP415-P44a) the intensity of these broad peaks is further reduced. The amorphous form (class 1) is shown as a reference (bottom, sample: PP415-P42). Patterns are not scaled, but are offset in the y direction for comparison purposes.
[00170] FIG. 126 - TG-FTIR thermogram of sample PP415-P44a, which corresponds to the amorphous form (class 1).
[00171] FIG. 127 - PXRD pattern of sample PP415-P45 (top) corresponds to a form of THF solvate (class 5). After drying the PP415-P45 sample for 1 day (2° from above, the PP415-P46 sample), the material is essentially amorphous. Some broad peaks with low intensity remain. After a total of 4 days of drying (2° from below, from sample PP415-P46a), the standard remains unchanged. The amorphous form (class 1) is shown as a reference (bottom part, sample PP415-P42). Patterns are not scaled, but are offset in the y direction for comparison purposes.
[00172] FIG. 128 - TG-FTIR thermogram of sample PP415-P46a, which corresponds to the amorphous form (class 1).
[00173] FIG. 129 - PXRD pattern of sample PP415-P42 (top) corresponds to pattern of amorphous form (class 1) (bottom, sample PP415-P1). Patterns have been scaled and offset in the y direction for comparison purposes.
[00174] FIG. 130 - PXRD pattern of sample PP415-P43 (top) corresponds to the pattern of the isostructural solvate form (Class 2) (below, sample PP415-P19) and not to the pattern of THF solvate (class 5) (middle, PP415-P14 of the sample). Patterns have been scaled and offset in the y direction for comparison purposes.
[00175] FIG. 131 - PXRD patterns from PP415-P47 (top) and PP415-P48 (middle) samples essentially correspond to the pattern of the isostructural solvate forms (Class 2) (bottom, PP415-P19 sample), although there are some differences. Patterns have been scaled and offset in the y direction for comparison purposes.
[00176] FIG. 132 - PXRD pattern of sample PP415-P49 (top) corresponds to pattern of amorphous form (class 1) (bottom, sample PP415-P1). Patterns have been scaled and offset in the y direction for comparison purposes. DESCRIPTION OF ILLUSTRATIVE MODALITIES
[00177] The present invention provides, in one aspect, the compound:
N -((4a S,6a R,6b S,8a R,12a S,14a R,14b S)-11-cyano-2,2,6a,6b,9,9,12a-heptamethyl-10 ,14-dioxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,12a,14,14a,14b-octadecahydropicen-4a-yl)- 2,2-difluoropropanamide, which is also referred to herein as RTA 408, 63415, or PP415. In other non-limiting aspects, the present invention also provides polymorphic forms thereof, including solvates thereof. In other non-limiting aspects, the invention also provides pharmaceutically acceptable salts thereof. In other non-limiting aspects, there are also methods provided for the preparation, pharmaceutical compositions and kits and articles of manufacture of these compounds and polymorphic forms thereof. I. Definitions
[00179] When used in the context of a chemical group: "hydrogen" means H; "Hydroxy" means -OH; "Oxo" means =O; "Carbonyl" means -C (=O) -;"Carboxy" means -C (=O) OH (also written as -COOH or -CO 2 H); "Halo" independently means -F, -Cl, Br or I; "Amino" means -NH 2; "Hydroxyamino" means -NHOH; "Nitro" means -NO2; imino means = NH; "Cyan" means -CN; "Isocyanate" means N=C=O; "Azide" means N3; in a monovalent context "phosphate" means -OP(O)(OH)2 or a deprotonated form thereof; in a bivalent context "phosphate" means -OP(O)(OH)O- or a deprotonated form thereof; "Mercapto" means -SH; and "thio" means = S; "Sulfonyl" means -S(O)2 -; and "sulfinyl" means -S(O) -. Any undefined valence in an atom of a structure presented in this application implicitly represents a hydrogen atom bonded to the atom.
[00180] In the context of this description, the formulas:


[00181] represent the same structures. When a dot is drawn on a carbon, the dot indicates the hydrogen atom attached to the carbon that is leaving the plane of the page.
[00182] The use of the word "a" or "an" when used in conjunction with the term "comprising" in the claims and/or the specification may mean "one", but is also consistent with the meaning of "one or more , "," at least one "and" one or more than one. "
[00183] Throughout this application, the term "about" is used to indicate that a value includes the inherent variation of error for the device, being the method used to determine the value, or the variation that exists between study subjects . When used in the context of X-ray powder diffraction, the term "about" is used to indicate a value of 0.2 ± o2θ of the reported value, preferably a value of 0.1 ± o2θ of the reported value. When used in the context of differential scanning or glass transition calorimetry temperatures, the term "about" is used to indicate a value of ± 10°C relative to the peak maximum, preferably a value of ± 2°C in relation to the peak maximum. When used in other contexts, the term "about" is used to indicate a value of ±10% of the reported value, preferably a value of ±5% of the reported value. It is to be understood that whenever the term "about" is used, a specific reference to the exact numerical value indicated is also included.
[00184] The terms "comprise", "have" and "include" are open connecting verbs. All forms or tenses of one or more of these verbs, such as "comprises", "comprising", "has", "with", "includes" and "including", are also open. For example, any method that "comprises", "has", or "includes" one or more steps is not limited to just those that have one or more steps and also covers other steps not listed.
The term "effective", as this term is used in the specification and/or claims, means adequate to achieve a desired, expected or intended result. "Effective amount", "therapeutically effective amount" or "pharmaceutically effective amount" when used in the context of treating a patient or individual with a compound means the amount of the compound when administered to an individual or patient for the treatment of a disease, is sufficient to effect this treatment for the disease.
[00186] The term "halogen peak" in the context of X-ray powder diffraction would mean a broad peak, often measuring >10°2θ in an X-ray diffractogram, typically characteristic of a solid or amorphous system.
The term "hydrate" when used as a modifier of a compound means that the compound has less than one (e.g., hemihydrate), one (e.g., monohydrate), or more than one ( for example, dihydrate) molecules of water associated with each molecule of the compound, such as in solid forms of the compound.
[00188] As used herein, the term "IC 50 "refers to an inhibitory dose that is 50% of the maximum response obtained. This quantitative measure indicates how much of a particular drug or other substance (inhibitor) is needed to inhibit a given biochemical, chemical or biological process (or component of a process, i.e. an enzyme, the cell, the receptor of the cell or microorganism) by half.
[00189] An "isomer" of a first compound is a separate compound, in which each molecule contains the same constituent atoms as the first compound, but in which the configuration of the atoms differs in three dimensions.
As used herein, the term "patient" or "individual" refers to a living mammalian organism, such as a human, monkey, cow, sheep, goat, dog, cat, mouse, rat, guinea pig, or its transgenic species. In certain embodiments, the patient or individual is a non-human animal. In certain embodiments, the patient or individual is a primate. In certain embodiments, the patient or individual is a human being. Non-limiting examples of human beings are adults, juveniles, children and fetuses.
As generally used herein "pharmaceutically acceptable" refers to those compounds, materials, compositions, and/or dosage forms that are, within the scope of good medical judgment, suitable for use in contact with tissues, organs, and/or bodily fluids from humans and animals without excessive toxicity, irritation, allergic response, or other problems or complications commensurate with a reasonable benefit/risk ratio.
"Pharmaceutically acceptable salts" means salts of compounds of the present invention which are pharmaceutically acceptable, as defined above, and which possess the desired pharmacological activity. Such salts include acid addition salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or with organic acids such as 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, 2-naphthalenesulfonic acid, 3-phenylpropionic acid, 4,4'-methylenebis (3-hydroxy-2-ene-1-carboxylic acid), 4 -methylbicyclo [2.2.2] oct-2-ene-1-carboxylic acid, acetic acid, aliphatic mono and dicarboxylic acids, aliphatic sulfuric acids, aromatic sulfuric acids, benzenesulfonic acid, benzoic acid, camphorsulfonic acid, carbonic acid, acid cinnamic acid, citric acid, cyclopentanepropionic acid, ethanesulfonic acid, fumaric acid, glucoheptonic acid, gluconic acid, glutamic acid, glycolic acid, heptanoic acid, hexanoic acid, hydroxynaphthoic acid, lactic acid, lauryl sulfuric acid, maleic acid, malic acid, acid malonic, mandelic, methanesulfonic acid, muconic acid, o-(4-hydroxybenzoyl) benzoic acid, oxalic acid, p -chlorobenzenesulfonic acid, phenyl substituted alkanoic acids, propionic acid, p -toluenesulfonic acid, pyruvic acid, acid salicylic acid, stearic acid, succinic acid, tartaric acid, tertbutylacetic acid, trimethylacetic acid and the like. Pharmaceutically acceptable salts also include base addition salts which may be formed when the acids present are capable of reacting with inorganic or organic bases. Acceptable inorganic bases include sodium hydroxide, sodium carbonate, potassium hydroxide, aluminum hydroxide and calcium hydroxide. Acceptable organic bases include ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine and the like. It should be recognized that the particular anion or cation that forms a part of any salt of this invention is not critical, so long as the salt, as a whole, is pharmacologically acceptable. Additional examples of pharmaceutically acceptable salts and their methods of preparation and use are presented in Handbook of Pharmaceutical salts: Proprerties and Use (P.H. Stahl & C.G. Wermuth eds., Verlag Helvetica Chimica Acta, 2002.).
[00193] "Prevention" or "preventing" includes: (1) inhibiting the onset of a disease in an individual or patient who may be at risk for and/or a predisposition to the disease, but who does not yet experience or exhibit any or all of the disease pathology or symptomatology, and/or (2) delay the onset of the disease pathology or symptomatology in an individual or patient who may be at risk and/or a predisposition to the disease, but not yet experience or show any one or all of the pathology or symptomatology of the disease.
"Prodrug" means a compound that is metabolically convertible in vivo to an inhibitor according to the present invention. The prodrug itself may or may not also have activity against a given target protein. For example, a compound comprising a hydroxyl group can be administered as an ester, which is converted by hydrolysis in vivo to the hydroxy compound. Suitable esters that can be converted in vivo to the hydroxyl compounds include acetates, citrates, lactates, phosphates, tartrates, malonates, oxalates, salicylates, propionates, succinates, fumarates, maleates, methylene-bis-b-hydroxynaphthoate, gentisates, isethionates, p -di-toluoyltartrates, methanesulfonates, ethanesulfonates, benzenesulfonates, p-toluenesulfonates, cyclohexylsulfamates, quinates, amino acid esters, and the like. Likewise, a compound comprising an amine group can be administered as an amide which is converted by hydrolysis in vivo to the amine compound.
[00195] A "stereoisomer" or "optical isomer" is an isomer of a particular compound, in which the same atoms are attached to the same atoms of others, but in which the configuration of the atoms differs in three dimensions. "Enantiomers" are stereoisomers of a particular compound that are mirror images of each other, such as the left and right hands. "Diastereomers" are stereoisomers of a given compound that are not enantiomers. Chiral molecules contain a chiral center, also referred to as a stereocenter or estrogenic center, which is any point, though not necessarily, an atom, a molecule in which such groups an interchangeability of any two groups leads to a rolling stereoisomer. . In organic compounds, the chiral center is typically a carbon, sulfur or phosphorus atom, although it is also possible for other atoms to be stereocenters in organic and inorganic compounds. The molecule can have multiple stereocenters, giving it many stereoisomers. In compounds whose stereoisomerism is due to tetrahedral estrogen centers (eg carbon tetrahedral), the total number of hypothetically possible stereoisomers will not exceed 2n, where n is the number of tetrahedral stereocenters. Symmetrical molecules often have fewer than the maximum possible number of stereoisomers. A 50:50 mixture of enantiomers is referred to as a racemic mixture. Alternatively, a mixture of enantiomers can be enantiomerically enriched so that one enantiomer is present in an amount greater than 50%. Typically, enantiomers and/or diastereomers can be separated or resolved using techniques known in the art. It is contemplated that for any stereocenter or axis of chirality for which the stereochemistry is not defined, that stereocenter or axis of chirality may be present in its R form, S form, or as a mixture of the R and S forms, including racemic and non-racemic mixtures. As used herein, the phrase "substantially free of other stereoisomers" means that the composition contains <15%, more preferably <10%, even more preferably <5%, or most preferably <1% of another stereoisomer(s).
[00196] "Treatment" or "treating" includes (1) inhibiting a disease in an individual or patient suffering or exhibiting the pathology or symptomatology of the disease (for example, arresting further development of the pathology and/or symptomatology), (2 ) ameliorating a disease in an individual or patient who suffers or exhibits the pathology or symptomatology of the disease (eg, reversing the pathology and/or symptomatology), and/or (3) effecting any measurable decrease in a disease in an individual or patient who experiences or presents the pathology or symptomatology of the disease.
[00197] The above definitions supersede any conflicting definition in any of the reference which is incorporated herein by reference. The fact that some terms are defined, however, should not be taken as indicating that any term that is not defined is indefinite. Rather, all terms used are believed to describe the invention in terms such that one of ordinary skill will appreciate the scope and practice of the present invention. II. RTA 408 and Synthetic Methods
[00198] RTA 408 can be prepared according to the methods described in the Examples section below. These methods can be further modified and optimized to apply the principles and techniques of organic chemistry as applied by one of ordinary skill in the art. Such principles and techniques are taught, for example, in March's Advanced Organic Chemistry: Reations, Mechanisms and Structure (2007), which is incorporated herein by reference.
[00199] It should be recognized that the particular anion or cation that forms a part of any salt of this invention is not critical, so long as the salt, as a whole, is pharmacologically acceptable. Additional examples of pharmaceutically acceptable salts and their methods of preparation and use are set forth in the Handbook of Pharmaceutical Salts: Properties and Use (2002), which is incorporated herein by reference.
[00200] RTA 408 may also exist in the form of a prodrug. Since prodrugs are known to improve numerous desirable qualities of pharmaceuticals, e.g. solubility, bioavailability, manufacture, etc., the compounds used in some methods of the invention may, if desired, be presented in prodrug form. Thus, the invention contemplates prodrugs of the compounds of the present invention, as well as methods of delivering prodrugs. Prodrugs of the compounds used in the invention can be prepared by modifying the functional groups present in the compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound. Thus, prodrugs include, for example, the compounds described herein, in which a hydroxy, amino or carboxyl group is attached to any group which, when the prodrug is administered to a patient, cleaves to form a hydroxyl, amino or carboxylic acid group. , respectively.
[00201] RTA 408 may contain one or more asymmetrically substituted carbon or nitrogen atoms and may be isolated in an optically active or racemic form. Thus, all chiral, diastereomeric, racemic form, epimeric form, and all geometric isomeric forms of a structure are intended, unless the specific stereochemistry or isomeric form is specifically indicated. RTA 408 can occur as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. In some embodiments, a single diastereomer is obtained. The chiral centers of RTA 408 in accordance with the present invention may have the S or the P configuration.
[00202] Furthermore, the atoms constituting RTA 408 of the present invention are intended to include all isotopic forms of such atoms. Isotopes, as used herein, include atoms that have the same atomic number but different mass numbers. By way of general example and without limitation, hydrogen isotopes include tritium and deuterium, and carbon isotopes include 13C and 14C. Likewise, it is contemplated that one or more carbon atom(s) of a compound of the present invention may be replaced by a silicon atom(s). Furthermore, it is contemplated that one or more oxygen atom(s) of RTA 408 may be replaced by a sulfur or selenium atom(s).
[00203] RTA 408 and its polymorphic form, may also have the advantage of being more effective than, being less toxic than, being longer acting than, being more potent than, producing fewer side effects than, be more easily absorbed than, and/or have a better pharmacokinetic profile (eg, greater oral bioavailability and/or lower clearance) than, and/or have other useful pharmacological, physical, chemical or more advantages, the compounds known in the state of the art to be used in the indications described herein. III. Polymorphic forms of RTA 408
[00204] In some embodiments, the present invention provides different solid forms of RTA 408, including its solvates. A preliminary preformulation and polymorphism study was performed, and RTA 408 was found to have a high tendency for solvate formation. Class 2, 3, 4 and 5 crystalline forms are consistent with solvates. For a description of the classes, see Table 1 below. Attempts at dry grades 2 and 3 (two groups of isostructural solvates) were unsuccessful, consistent with tightly bound solvent molecules. In some embodiments, drying of a class 4 solid (acetonitrile solvate) has led to an isostructural desolvated form. In some embodiments, drying of a solid class 5 (THF solvate) resulted in amorphous form class 1. Unsolvated forms of RTA 408 include the amorphous form (class 1), and in the desolvated solvate lens class 4 (isostructural to class 4 acetonitrile solvate). In some embodiments, the amorphous form has a high glass transition with Tg ~ 153°C (ΔCp = 0.72 J/g°C) and is only slightly hygroscopic (Δm = +0.4% 50% 85% ^ rh). In some embodiments, the amorphous form is stable for at least four weeks under conditions of high temperature and humidity (ie, open at 40°C/~75% relative humidity or closed at 80°C). In some embodiments, the amorphous form (class 1) has been successfully prepared from class 2 materials in a two-step process (transformation to class 5 and subsequent drying of class 5 to obtain the amorphous form), as well as in a code for a direct step method (precipitation from an acetone solution in a cold water bath). Class 4 solvated crystalline solvate (isostructural for class 4 solvate) is slightly hygroscopic (mass gain ~0.7 wt.-% 50% relative humidity at 85%) and has a possible melting point at 196 0.1°C (ΔH = 29.31 J/g).
[00205] A sample of the amorphous form of 63415, class 1, was characterized by FT-Raman, PXRD, TG-FTIR, Karl Fischer titration, 1 H-NMR, DSC, and DVS spectroscopy (see Examples section for details additional). The sample was found to contain ~0.9% by weight. - EtOH with traces of H 2 O (according to TG-FTIR). A water content of 0.5% by weight. - was determined by Karl Fischer titration. DSC shows a high glass transition temperature with Tg ~ 153°C (Δ Cp = 0.72 J/g°C). According to DVS, the material is slightly hygroscopic (Δm = +0.4% 50% 85% ^rh). No crystallization was observed in the DSC or DVS experiments.
[00206] The chemical stability of the amorphous form was investigated in organic solvents, including acetone, EtOAc, MeOH, and MeCN, as well as different aqueous media (eg, 1% aq. Tween 80, 1% aq. SDS, 1% aq. CTAB) at a concentration of 1 mg/ml at 6 hr, 24 hr, 2 d, and 7 day time points. >1% decomposition was observed only for solutions in MeCN after 7 days and for suspensions in 1% aqueous medium of Tween 80 (at all times points at 254 nm and after 24 h, 2 d, and 7 days at 242 nm).
[00207] Furthermore, the stability of the amorphous form was investigated by storage under conditions of high temperature and humidity (open at 25°C/62% rh and 40°C/75% rh and closed at 60°C and 80°C ). After one week, two weeks and four weeks, stored samples were analyzed by PXRD. None of the samples differed from the amorphous starting material.
[00208] More than 30 crystallization and drying experiments were performed, including suspension equilibrium, slow cooling, evaporation and precipitation. Four new crystalline forms were obtained (classes 2, 3, 4 and 5), in addition to the amorphous form (class 1).
[00209] The four new forms (classes 2, 3, 4 and 5) were characterized by FT-Raman, PXRD and TG-FTIR spectroscopy. All forms of solvate matching (Table 1). Drying tests under vacuum or N2 flow were carried out in order to obtain a crystalline solid, non-solvated form of 63415. Table 1. Summary of the classes obtained

[00210] Class 2: Most of the crystallization experiments that were carried out resulted in class 2 solid material (see Examples section below). Its members may correspond to isostructural, non-stoichiometric (<0.5 eq.) Solvates (of heptane, cyclohexane, isopropyl ether, 1-butanol, triethylamine, and possibly other solvents such as hexane , other ethers, etc.) with tightly bound solvent molecules. The standard Raman and PXRD spectra within this class are very similar to each other, so the structures may be essentially identical, with only minor differences due to the different solvents that have been incorporated.
[00211] Drying experiments on class 2 samples did not result in a crystalline, unsolvated form. Even at high temperatures (80°C) and a high vacuum (<1x10 -3 mbar), it was not possible to remove the tightly bound solvent molecules completely; a solvent content of >2% by weight always remained. The crystallinity of the partially dried samples is reduced, but no transformation to a different structure or substantial amorphization was observed.
[00212] Class 3: Class 3 solid material can be obtained from various crystallizations (see Examples section below). Class 3 samples are likely isostructural solvates of 2PrOH, EtOH, and probably acetone with tightly bound solvent molecules. They can correspond to any stoichiometric hemisolvates or non-stoichiometric solvates, with a solvent content of ~0.5 eq. As with class 2, the Raman and PXRD pattern spectra within this class are very similar to each other, indicating similar structures that incorporate different solvents.
[00213] Similar to class 2, drying experiments were not successful. Very tightly bound solvent molecules could only be partially removed (i.e. ~5.4% by weight.- to ~4.8% by weight.- then up to 3 d at 1x10 -3 mbar and 80°C). PXRD patterns remained unchanged.
[00214] Class 4 can be obtained from a 7:3 MeCN/H2O solvent system (see Examples section below). It is more likely to correspond to a crystalline acetonitrile hemisolvate. By drying (under vacuum or N2 flow at elevated temperatures), most solvent molecules can be removed without altering or destroying the crystal structure (PXRD remained unchanged). Thus, a crystalline, unsolvated form (or rather desolvated solvate) was obtained. It is slightly hygroscopic (mass gain ~0.7 wt.-% 50% relative humidity at 85%) and has a possible melting point at 196.1°C (ΔH = 29.31 J/g).
[00215] Class 5 can be obtained from a ~ 1:1 THF/H2O solvent system. Class 5 contains THF (and maybe H2O) binding. As the content of the two components cannot be easily quantified separately, the exact nature of this crystalline solvate has not been determined.
[00216] Drying of class 5 resulted in significant desolvation and transformation towards the amorphous form (class 1). In some embodiments, the amorphous form of RTA 408 can be prepared by suspending class 2 in 1:1 heptane solvate of THF/H2 O to form a class 5 solid, followed by drying and amorphization.
[00217] Experiments aiming to prepare the amorphous form (class 1) were carried out using class 2 starting material. Mainly amorphous material (class 1), it was prepared from class 2 materials in a two-step process through class 5 on a scale of 100 mg and 3 g (after drying at 100 mbar, 80°C, several days ). The preparation of fully amorphous material (class 1) was found to be possible in a one-step process avoiding the THF solvent by direct precipitation of the amorphous form (class 1) from an acetone solution of the class 2 material in a cold water bath. IV. Diseases associated with inflammation and/or oxidative stress
[00218] Inflammation is a biological process that provides resistance to infectious organisms or parasites and repair of damaged tissue. Inflammation is usually characterized by localized vasodilation, redness, swelling and pain, recruitment of leukocytes to the site of infection or injury, production of inflammatory cytokines such as TNF-a and IL-1, and production of species reactive oxygen or nitrogen, such as hydrogen peroxide, the superoxide radical, and peroxynitrite. In the later stages of inflammation, tissue remodeling, angiogenesis, and scarring (fibrosis) can occur as part of the wound healing process. Under normal circumstances, the inflammatory response is regulated, temporary, and is resolved in an orchestrated fashion once the infection or injury has been adequately treated. However, acute inflammation can become excessive and life-threatening if regulatory mechanisms fail. Alternatively, inflammation can become chronic and cause cumulative tissue damage or systemic complications. Based on at least the elements presented herein, RTA 408 can be used in the treatment or prevention of inflammation or diseases associated with inflammation.
[00219] Many serious and incurable illnesses involve dysregulation of inflammatory processes, including diseases such as cancer, atherosclerosis and diabetes, which were not traditionally seen as inflammatory conditions. In the case of cancer, inflammatory processes are associated with processes that include tumor formation, progression, metastasis, and resistance to therapy. In some modalities, RTA 408 can be used in the treatment or prevention of cancers, including a carcinoma, sarcoma, lymphoma, leukemia, melanoma, mesothelioma, multiple myeloma, or seminoma, or cancer of the bladder, blood, bone, brain, breast, breast, central nervous system, cervix, colon, endometrium, esophagus, gallbladder, genital organs, genitourinary tract, head, kidney, larynx, liver, lung, muscle tissue, neck, oral or nasal mucosa , ovary, pancreas, prostate, skin, spleen, small intestine, large intestine, stomach, testis, or thyroid. Atherosclerosis, long seen as a disorder of lipid metabolism, is now understood to be essentially an inflammatory condition, with activated macrophages playing an important role in the formation and eventual rupture of atherosclerotic plaques. Activation of inflammatory signaling pathways has also been shown to play a role in the development of insulin resistance as well as peripheral tissue damage associated with diabetes hyperglycemia. Overproduction of reactive oxygen species and reactive nitrogen species, such as superoxide, hydrogen peroxide, nitric oxide, and peroxynitrite, is a hallmark of inflammatory conditions. Evidence of deregulated peroxynitrite production has been reported in a wide range of diseases (Szabo et al., 2007; Schulz. et al., 2008; Forstermann, 2006; Pall, 2007).
[00220] Autoimmune diseases such as rheumatoid arthritis, lupus, psoriasis, multiple sclerosis and involve inappropriate and chronic activation of inflammatory processes in affected tissues, resulting from auto dysfunction versus non-independent immune system recognition and response mechanisms. In neurodegenerative diseases such as Alzheimer's and Parkinson's disease, nerve damage is correlated with microglial activation and elevated levels of pro-inflammatory proteins such as nitric oxide synthase (iNOS). Chronic organ failure, such as kidney failure, heart failure, liver failure, chronic obstructive pulmonary disease, is closely related to the presence of chronic oxidative stress and inflammation, leading to the development of fibrosis and eventual loss of organ function. Oxidative stress on vascular endothelial cells, which line the larger and smaller blood vessels, can lead to endothelial dysfunction and is believed to be an important contributing factor to the development of systemic cardiovascular diseases, complications of diabetes, chronic kidney disease, and other forms organ failure; and a number of other age-related diseases, including degenerative diseases of the central nervous system and the retina.
[00221] Many other disorders involve oxidative stress and inflammation in the affected tissues, including inflammatory bowel disease; inflammatory skin diseases; mucositis and dermatitis caused by radiotherapy and chemotherapy; eye diseases such as uveitis, glaucoma, macular degeneration, and various forms of retinopathy; transplant failure and rejection; ischemia-reperfusion injury; chronic pain; degenerative conditions of bones and joints, including osteoarthritis and osteoporosis; asthma and cystic fibrosis; seizure disorders; and neuropsychiatric conditions, including schizophrenia, depression, bipolar disorder, post-traumatic stress disorder, attention deficit disorders, autism spectrum disorders, and eating disorders such as anorexia nervosa. The dysregulation of inflammatory signaling pathways is believed to be an important factor in the pathology of muscle wasting diseases, including muscular dystrophy and various forms of cachexia.
A variety of life-threatening acute diseases also involve dysregulated inflammatory signaling, including acute organ failure involving pancreas, kidneys, liver or lungs, myocardial infarction or acute coronary syndrome, stroke, septic shock, trauma, burns severe, and anaphylaxis.
[00223] Many complications of infectious diseases also involve dysregulation of inflammatory responses. While the inflammatory response can kill invading pathogens, an excessive inflammatory response can also be quite destructive and, in some cases, can be a major source of damage to infected tissue. Furthermore, an excessive inflammatory response can also lead to systemic complications due to the excessive production of inflammatory cytokines, such as TNF-α and IL-1. This is believed to be a factor in mortality arising from severe flu, severe acute respiratory syndrome and sepsis.
[00224] Aberrant or excessive expression of either iNOS or cyclooxygenase-2 (COX-2) has been implicated in the pathogenesis of many disease processes. For example, it is evident that NO is a potent mutagenic agent (Tamir and Tannebaum, 1996), and that nitric oxide can also activate COX-2 (Salvemini et al., 1994). In addition, there is a marked increase in iNOS in rat colon tumors induced by the carcinogen, azoxymethane (Takahashi et al., 1997). A series of synthetic analogues of the oleanolic acid triterpenoids have been shown to be potent inhibitors of cellular inflammatory processes, such as the induction by IFN-g of induced nitric oxide synthase (iNOS) and COX-2 in mouse macrophages. See Honda et al. (2000a), Honda et al. (2000b), and Honda et al (2002), all of which are incorporated herein by reference.
[00225] In one aspect, RTA 408 described herein is, in part, characterized by its ability to inhibit nitric oxide production in macrophage-derived RAW 264.7 cells induced by exposure to interferon-Y. RTA 408 is further characterized by the ability to induce the expression of antioxidant proteins, such as NQO1, and reduce the expression of pro-inflammatory proteins, such as COX-2 and nitric oxide synthase (iNOS). These properties are important for the treatment of a wide variety of diseases and disorders involving oxidative stress and the dysregulation of inflammatory processes, including cancer, complications from localized or total body exposure to ionizing radiation, mucositis and dermatitis resulting from radiotherapy or chemotherapy, autoimmune diseases, cardiovascular disease, including atherosclerosis, ischemia-reperfusion injury, acute and chronic organ failure, including kidney failure and heart failure, respiratory disease, diabetes and diabetes complications, severe allergies, transplant rejection, graft-versus-host disease, neurodegenerative diseases, eye and retinal diseases, acute and chronic pain, degenerative bone diseases including osteoarthritis and osteoporosis, inflammatory bowel diseases, dermatitis and other skin diseases , sepsis, burns, seizure disorders, and neuropsychiatric disorders.
[00226] In another aspect, RTA 408 can be used to treat an individual with a condition such as eye diseases. For example, uveitis, macular degeneration (both dry and wet form), glaucoma, diabetic macular edema, blepharitis, diabetic retinopathy, corneal endothelial diseases and disorders such as corneal endothelial Fuchs' dystrophy, post-inflammation. operative, dry eye, allergic conjunctivitis, and other forms of conjunctivitis are non-limiting examples of eye diseases that can be treated with RTA 408.
[00227] In another aspect, RTA 408 can be used to treat an individual with a condition such as skin diseases or disorders. For example, dermatitis, including allergic dermatitis, atopic dermatitis, dermatitis due to exposure to chemicals, and radiation-induced dermatitis; thermal or chemical burns; chronic wounds, including diabetic ulcers, pressure ulcers, and venous ulcers; acne; alopecia, including baldness and drug-induced alopecia; other hair follicle disorders; bullous epidermolysis; sunburn and its complications; skin pigmentation disorders including vitiligo; aging-related skin problems; post-surgical scarring; prevention or reduction of scarring from skin lesions, surgery or burns; psoriasis; dermatological manifestations of autoimmune diseases or graft versus host; prevention or treatment of skin cancer; disorders involving hyperproliferation of skin cells such as hyperkeratosis is a non-limiting example of skin disorders that can be treated with RTA 408.
[00228] Without being bound by theory, activation of the antioxidant/anti-inflammatory pathway KEAP1/Nrf2/ARE is believed to be involved in both the anti-inflammatory and anti-cancer properties of the compounds disclosed herein.
[00229] In another aspect, RTA 408 can be used to treat an individual with a condition caused by elevated levels of oxidative stress in one or more tissues. Oxidative stress results from abnormally high and prolonged levels of reactive oxygen species such as superoxide, hydrogen peroxide, nitric oxide, and peroxynitrite (formed by the reaction of nitric oxide and superoxide). Oxidative stress can be accompanied by any acute or chronic inflammation. Oxidative stress can be caused by mitochondrial dysfunction, by activation of immune system cells such as macrophages and neutrophils, by acute exposure to an external agent such as radiation or a cytotoxic chemotherapeutic agent (ionizing eg doxorubicin ), trauma or other acute tissue injury, ischemia/reperfusion, poor circulation or anemia, localized or systemic hypoxia or hyperoxia, high levels of inflammatory cytokines and other proteins related to inflammation, and/or other physiological states abnormalities, such as hyperglycaemia or hypoglycaemia.
[00230] In animal models of many of these conditions, stimulating the inducible expression of heme oxygenase (HO-1), a target gene of the Nrf2 pathway, has been shown to have a significant therapeutic effect, including in models of myocardial infarction, renal failure , transplant failure and rejection, stroke, cardiovascular disease, and autoimmune disease (eg, Sacerdoti et al., 2005; Abraham & Kappas, 2005; Bach, 2006; Araujo et al., 2003; Liu et al., 2006; Ishikawa, and others from 2001; Kruger and others from 2006; Satoh. et al. 2006; Zhou. et al. 2005; Morse and Choi, 2005; Morse and Choi, 2002). This enzyme breaks free heme down to iron, carbon monoxide (CO), and biliverdin (which is subsequently converted to the potent antioxidant molecule, bilirubin).
[00231] In another aspect, RTA 408 can be used in the prevention or treatment of tissue damage or organ failure, both acute and chronic, that results from oxidative stress exacerbated by inflammation. Examples of diseases that fall into this category include heart failure, liver failure, transplant failure and rejection, kidney failure, pancreatitis, fibrotic lung diseases (cystic fibrosis, COPD, and idiopathic pulmonary fibrosis, among others), diabetes (including complications) , atherosclerosis, injury, glaucoma, stroke, autoimmune disease, autism, macular degeneration, and ischemia-reperfusion muscular dystrophy. For example, in the case of autism, studies suggest that increased oxidative stress on the central nervous system may contribute to the development of the disease (Chauhan and Chauhan, 2006).
[00232] Evidence also links oxidative stress and inflammation in the development and pathology of many other diseases of the central nervous system, including psychiatric disorders such as psychoses, depression and bipolar disorder; seizure disorders such as epilepsy; pain and sensory syndromes, such as migraine, neuropathic pain, or tinnitus; and behavioral syndromes such as attention deficit disorders. See, for example, Dickerson et al. 2007; Hanson et al., 2005; Kendall-Tackett, 2007; Lencz et al., 2007; Dudhgaonkar et al. 2006; Lee et al. 2007; Morris et al., 2002; Ruster et al. 2005; McIver et al. 2005; Sarchielli et al., 2006; Kawakami et al. 2006; Ross et al. 2003, all of which are incorporated herein by reference. For example, elevated levels of inflammatory cytokines, including TNF-α, interferon-Y and IL-6, are associated with severe mental illness (Dickerson et al., 2007). Microglial activation has also been linked to severe mental illness. Therefore, downregulating inflammatory cytokines and inhibiting excessive microglial activation could be beneficial in patients with schizophrenia, depression, bipolar disorder, autism spectrum disorders, and other neuropsychiatric disorders.
Thus, in conditions involving oxidative stress alone or oxidative stress exacerbated by inflammation, treatment may comprise administering to a subject a therapeutically effective amount of a compound of the present invention, such as those described above or to throughout this specification. Treatment can be administered preventively, prior to a predictable state of oxidative stress (eg, organ transplantation or administration of radiation therapy to a cancer patient), or may be administered therapeutically in settings which involve the oxidative stress created and inflammation. In some embodiments, when a compound of the present invention is used to treat a patient receiving radiation therapy and/or chemotherapy, the compound of the invention may be administered before, at the same time, and/or after the chemotherapy or radiation therapy. , or the compound can be administered in combination with the other therapies. In some embodiments, the compound of the present invention can prevent and/or reduce the severity of side effects associated with radiation therapy or chemotherapy (using a different agent) without reducing the anticancer effects of radiation therapy or chemotherapy. Because such side effects can be dose limiting for radiation therapy and/or chemotherapy, in some embodiments, the compound of the present invention can be used to allow for higher dosage and/or more frequent radiation therapy and/or chemotherapy. radiation, for example, which results in greater treatment effectiveness. In some embodiments, the compound of the invention when administered in combination with radiotherapy and/or chemotherapy can increase the effectiveness of a given dose of radiation and/or chemotherapy. In some embodiments, the compound of the invention, when administered in combination with radiotherapy and/or chemotherapy can increase the effectiveness of a given dose of radiation and/or chemotherapy and reduce (or at least not add to) the side to the effects of radiation and/or chemotherapy. In some embodiments, and without being limited by theory, this combinatorial efficacy may result from inhibition of the activity of the pro-inflammatory transcription factor NF-KB by the compound of the invention. NF-KB is often chronically activated in cancer cells, and this activation is associated with resistance to therapy and promotion of tumor progression (eg, Karin, 2006; Aghajan. et al., 2012). Other transcription factors that promote inflammation and cancer, such as STAT3 (eg, he and Karin 2011; Grivennikov and Karin, 2010), can also be inhibited by the compound of the invention, in some modalities.
[00234] RTA 408 can be used to treat or prevent inflammatory diseases such as sepsis, dermatitis, autoimmune disease, and osteoarthritis. RTA 408 can also be used to treat or prevent inflammatory pain and/or neuropathic pain, for example, by inducing Nrf2 and/or inhibiting NF-KB
[00235] RTA 408 can also be used to treat or prevent diseases such as cancer, inflammation, Alzheimer's disease, Parkinson's disease, multiple sclerosis, autism, amyotrophic lateral sclerosis, Huntington's disease, autoimmune diseases such as rheumatoid arthritis, lupus, Crohn's disease, and psoriasis, inflammatory bowel disease, all other diseases whose pathogenesis is believed to involve excessive nitric oxide production or both prostaglandins, and pathologies involving oxidative stress alone or oxidative stress exacerbated by inflammation. RTA 408 can be used to treat or prevent cancers include carcinoma, sarcoma, lymphoma, leukemia, melanoma, mesothelioma, multiple myeloma, or seminoma, or cancer of the bladder, blood, bone, brain, breast, central nervous system, cervix , colon, endometrium, esophagus, gallbladder, genital organs, genitourinary tract, head, kidney, larynx, liver, lung, muscle tissue, neck, oral or nasal mucosa, ovary, pancreas, prostate, skin, spleen, small intestine, intestine, stomach, testis, or thyroid.
[00236] Another aspect of inflammation is the production of inflammatory prostaglandins, such as prostaglandin E. RTA 408 can be used to promote vasodilation, plasma leakage, localized pain, elevated temperature, and other symptoms of inflammation . The inducible form of the COX-2 enzyme is associated with its production, and elevated levels of COX-2 are found in inflamed tissues. Therefore, inhibition of COX-2 can alleviate many symptoms of inflammation and a number of important anti-inflammatory drugs (eg ibuprofen and celecoxib) act by inhibiting the activity of COX-2. A class of prostaglandins (cyclopentenonic cyPGs) (eg, 15-deoxy prostaglandin J2, or PGJ2) has been shown to play a role in stimulating the orchestrated resolution of inflammation (eg, Rajakariar et al., 2007). COX-2 is also associated with the production of cyclopentenonic prostaglandins. Therefore, COX-2 inhibition can interfere with the complete resolution of inflammation, potentially promoting the persistence of activated immune cells in tissues and leading to chronic, "latent" inflammation. This effect may be responsible for the increased incidence of cardiovascular disease in patients using selective COX-2 inhibitors for long periods of time.
[00237] In one aspect, RTA 408 can be used to control the production of pro-inflammatory cytokines within the cell by selectively activating regulatory cysteine residues (RCR values) on proteins that regulate the activity of redox sensitive transcription factors. Activation of RCR values by cyPGs has been shown to initiate a pro-resolution program, in which potently induced antioxidant and cytoprotective Nrf2 transcription factor activity and pro-inflammatory activities of pro-oxidant and NF-pro-inflammatory transcription factors kB and statistics are suppressed. In some modalities, RTA 408 can be used to increase the production of antioxidant and reducing molecules (NQO1, HO-1, SOD1, g -GCS) and to decrease oxidative stress and the production of pro-oxidant and pro-inflammatory molecules ( iNOS COX-2, TNF-α). In some embodiments, RTA 408 can be used to cause cells that host the inflammatory event to revert to a non-inflammatory state by promoting inflammation resolution and limiting excessive tissue damage to the host. A. Cancer
[00238] In some embodiments, RTA 408, polymorphic forms, and methods of the present description can be used to induce apoptosis in tumor cells, to induce cell differentiation, to inhibit cancer cell proliferation, to inhibit an inflammatory response, and/or to function in a chemopreventive capacity. For example, the invention provides novel polymorphic forms that have one or more of the following properties: (1) the ability to induce apoptosis and differentiate both malignant and non-malignant cells, (2) an activity at submicromolar or nanomolar levels as an inhibitor of proliferation of many pre-malignant or malignant cells, (3) the ability to suppress the de novo synthesis of the inflammatory enzyme nitric oxide synthase (iNOS), (4) an ability to inhibit NF-kB activation, and (5 ) the ability to induce the expression of heme oxygenase-1 (HO-1).
[00239] iNOS and COX-2 levels are elevated in certain cancers and have been implicated in carcinogenesis and COX-2 inhibitors have been shown to reduce the incidence of colon adenomas in primary humans (Rostom et al. 2007; Brown and DuBois, 2005; Crowel et al., 2003). iNOS is expressed in myeloid-derived suppressor cells (MDSCs) (Angulo et al., 2000) and COX-2 activity in cancer cells has been shown to result in the production of prostaglandin E2 (PGE 2), which has been shown to induce the expression of arginase in MDSCs (Sinha et al., 2007). Arginase and iNOS are enzymes that use L-arginine as a substrate and produce L-ornithine and urea, and L-citrulline and NO, respectively. The depletion of arginine from the tumor microenvironment by MDSCs, combined with the production of NO and peroxynitrite has been shown to inhibit proliferation and induce T-cell apoptosis (Bronte et al., 2003). Inhibition of COX-2 and iNOS has been shown to reduce the accumulation of MDSCs, restore the cytotoxic activity of tumor-associated T cells, and retard tumor growth (Sinha et al., 2007; Mazzoni et al., 2002; Zhou et al., , 2007).
[00240] Inhibition of the NF-kB and JAK/STAT signaling pathways has been implicated as a strategy to inhibit the proliferation of cancer epithelial cells and induce their apoptosis. Activation of NF-kB and STAT3 kB has been shown to result in suppression of apoptosis in cancer cells, and promotion of proliferation, invasion and metastasis. Many of the target genes involved in these processes have been shown to be transcriptionally regulated by both NF- kB and STAT3 (Yu et al., 2007).
[00241] In addition to their direct roles in cancer epithelial cells, NF-kB and STAT3 also have important roles in other cells found within the tumor microenvironment. Experiments in animal models have shown that NF-kB is required in both cancer cells and hematopoeitic cells to propagate the effects of inflammation on cancer onset and progression (Greten et al., 2004). Inhibition of NF-kB in myeloid and cancer cells reduces the number and size, respectively, of the resulting tumors. Activation of STAT3 in cancer cells results in the production of several cytokines (IL-6, IL-10) that suppress the maturation of tumor-associated dendritic cells (CC). Furthermore, STAT3 is activated by these cytokines in the dendritic cells themselves. Inhibition of STAT3 in mouse models of cancer restores DC maturation, promotes antitumor immunity, and inhibits tumor growth (Kortilewski et al., 2005). In some modalities, RTA 408 and its polymorphic forms can be used to treat cancer, including, for example, prostate cancer. In some embodiments, RTA 408 and its polymorphic forms can be used in a combination therapy to treat cancer including, for example, prostate cancer. See, for example, Example H below. B. Multiple sclerosis and other neurodegenerative diseases
In some embodiments, RTA 408, the polymorphic forms, and methods of the present invention can be used to treat patients for multiple sclerosis (MS) or other neurodegenerative diseases such as Alzheimer's disease, Alzheimer's disease. Parkinson's or amyotrophic lateral sclerosis. MS is known to be an inflammatory disease of the central nervous system (Williams et al., 1994; Merrill and Benvenist, 1996; Genain and NAUser, 1997). Based on several investigations, evidence suggests that oxidative and/or inflammatory or immune mechanisms are involved in the pathogenesis of Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS) and MS (Bagasra et al., 1995; McGeer and McGeer, 1995; Simonian and Coile, 1996; Kaltschmidt. et al., 1997). Epidemiological data indicate that the chronic use of NSAIDs that synthesize prostaglandins from arachidonate block, markedly reduces the risk of developing AD (McGeer et al., 1996; Stewart. et al., 1997). Thus, agents that block the formation of NO and prostaglandins, can be used in methods to prevent and treat neurodegenerative diseases. Successful therapeutic candidates for the treatment of such a disease typically require an ability to penetrate the blood-brain barrier. See, for example, US Patent Publication 2009/0060873 which is incorporated herein by reference. C. Neuroinflammation
[00243] In some embodiments, RTA 408, the polymorphic forms, and methods of the present invention can be used to treat patients with neuroinflammation. Neuroinflammation encapsulates the idea that microglial and astrocytic responses and actions in the central nervous system are fundamentally inflammation-like in character, and that these responses are fundamental to the pathogenesis and progression of a wide variety of neurologic disorders. - cos. These ideas have been extended from Alzheimer's disease to other neurodegenerative diseases (Eikelenboom et al., 2002; Ishizawa and Dickson, 2001), ischemic/toxic diseases (Gehrmann et al., 1995;. Touzani et al., 1999.), to tumor biology (Graeber et al. 2002) and for normal brain development. Neuroinflammation incorporates a broad spectrum of complex cellular responses, which include microglia and astrocyte activation and the induction of cytokines, chemokines, complement proteins, acute phase proteins, oxidative damage, and related molecular processes, and events can have detrimental effects on neuronal function, leading to neuronal injury, glial activation, and finally neurodegeneration. D. Kidney Diseases
[00244] In some modalities, RTA 408, as well as polymorphic forms thereof, can be used for the treatment of patients with kidney diseases and disorders, including renal failure and chronic kidney disease (CRF), based, for example, on the methods taught by US 8129429, which is incorporated by reference. Kidney failure, resulting in inadequate clearance of metabolic waste from blood and abnormal concentrations of electrolytes in the blood, is a significant medical problem worldwide, especially in developed countries. Diabetes and hypertension are among the most important causes of chronic kidney failure, also known as chronic kidney disease (CKD), but it is also associated with other conditions such as lupus. Acute kidney failure can result from exposure to certain drugs (eg, acetaminophen) or toxic chemicals, or from ischemia-reperfusion injury associated with shock or surgical procedures, such as transplantation, and can result in chronic kidney failure. In many patients, kidney failure progresses to a stage where the patient needs regular kidney dialysis or transplantation to continue to live. Both procedures are highly invasive and associated with significant side effects and quality of life issues. Although there are effective treatments for some complications of acute renal failure, such as hyperparathyroidism and hyperphosphatemia, no available treatment has been shown to stop or reverse the progression of underlying renal failure. Thus, agents that can improve kidney function would represent a significant advance in the treatment of kidney failure.
Inflammation contributes significantly to the pathology of CKD. There is also a strong mechanical link between oxidative stress and kidney dysfunction. The NF-KB signaling pathway plays an important role in CKD progression as NF-KB regulates the transcription of MCP-1, a chemokine, which is responsible for monocyte/macrophage recruitment, resulting in an inflammatory response that ultimately analysis hurts the kidney (Wardle, 2001). The KE-AP1/Nrf2/ARE pathway controls the transcription of several genes that encode antioxidant enzymes, including heme oxygenase-1 (HO-1). Ablation of the Nrf2 gene in female mice results in the development of glomerular lupus nephritis (Yoh et al., 2001). Furthermore, several studies have shown that HO-1 expression is induced in response to kidney damage and inflammation and that this enzyme and its products - bilirubin and carbon monoxide - play a protective role in the kidney (Nath et al., 2006 ).
[00246] Acute kidney injury (AKI) may follow ischemia-reperfusion injury, treatment with certain pharmacological agents such as cisplatin and rapamycin, and intravenous injection of radiocontrast medium used in medical imaging. As in CKD, inflammation and oxidative stress contribute to AKI pathology. The underlying molecular mechanisms induced by radiological contrast-induced nephropathy (RCN) are not well understood; However, it is likely that a combination of events, including prolonged vasoconstriction, kidney autoregulation, and direct contrast medium toxicity contribute to renal failure (Tumlin et al., 2006). Vasoconstriction results in decreased renal blood flow and causes ischemia-reperfusion and the production of reactive oxygen species. HO-1 is strongly induced under these conditions and has been shown to prevent ischemia-reperfusion injury in several different organs, including the kidneys (Nath et al., 2006). Specifically, induction of HO-1 has been shown to be protective in a mouse model of RCN (Goodman et al., 2007). Reperfusion also induces an inflammatory response, in part, though activation of NF-kB signaling (Nichols, 2004). NF-k B targeting has been proposed as a therapeutic strategy to prevent organ damage (Zingarelli et al., 2003). E. Cardiovascular Disease
[00247] In some embodiments, RTA 408, the polymorphic forms and methods of the present invention can be used to treat patients with cardiovascular disease. The etiology of cardiovascular disease is complex, but most causes are related to inadequate or completely interrupted blood supply to a critical organ or tissue. Often, such a condition is due to the rupture of one or more atherosclerotic plaques, which leads to the formation of a thrombus that blocks blood flow in a critical vessel.
[00248] In some incidents, atherosclerosis can be so extensive in critical blood vessels that stenosis (narrowing of the arteries) and develops blood flow to critical organs (including the heart) is chronic failure. This chronic ischemia can lead to target organ damage of various types, including cardiac hypertrophy associated with congestive heart failure.
[00249] Atherosclerosis, the underlying defect that leads to many forms of cardiovascular disease, when a physical defect or injury to the lining (endothelium) of an artery occurs, it triggers an inflammatory response involving vascular smooth muscle cell proliferation and infiltration. of leukocytes to the affected area. Ultimately, a complex lesion known as an atherosclerotic plaque can form, composed of the aforementioned cells combined with lipoprotein-bearing cholesterol deposits, and other materials (eg., Hansson et al., 2006). Despite the considerable advantages offered by current therapeutic treatments, mortality from cardiovascular disease remains unmet and significant needs in the treatment of cardiovascular disease remain.
[00250] Induction of HO-1 has been shown to be beneficial in a variety of models of cardiovascular disease, and low levels of HO-1 expression have been clinically correlated with increased risk of cardiovascular disease. RTA 408, the polymorphic forms and methods of the invention, therefore, can be used in the treatment or prevention of a variety of cardiovascular disorders, including but not limited to atherosclerosis, hypertension, myocardial infarction, chronic heart failure, stroke , subarachnoid hemorrhage, and restenosis. In some embodiments, RTA 408, the polymorphic forms and methods of the invention can be used as a combination therapy with other known cardiovascular therapies, such as, but not limited to, anticoagulants, thrombolytics, streptokinase, plasminogen activators. tissue surgery, coronary artery bypass grafting, balloon angioplasty, the use of stents, drugs that inhibit cell proliferation, or drugs that lower cholesterol levels. F. Diabetes
[00251] In some embodiments, RTA 408, as well as polymorphic forms thereof, can be used for the treatment of patients with diabetes, based, for example, on the methods taught by US 8129429, which is incorporated herein by reference. Diabetes is a complex disease characterized by the body's failure to regulate circulating glucose levels. This insufficiency can result from a lack of insulin, a peptide hormone that regulates both the production and absorption of glucose in various tissues. Insulin deficient compromises the ability of muscle, fat, and other tissues to properly absorb glucose, leading to hyperglycemia (abnormally high blood glucose levels). More commonly, such insulin deficiency results from inadequate production in the islet cells of the pancreas. In most cases, this arises from autoimmune destruction of these cells, a condition known as type 1 or juvenile-onset diabetes, but it can also be due to physical trauma or some other cause.
[00252] Diabetes can also arise when muscle and fat cells become less sensitive to insulin and do not absorb glucose properly, resulting in hyperglycemia. This phenomenon is known as insulin resistance, and the resulting condition is known as type 2 diabetes. Type 2 diabetes, the most common type, is highly associated with obesity and hypertension. Obesity is associated with an inflammatory state of adipose tissue, thought to play an important role in the development of insulin resistance (eg, Hotamisligil, 2006; Guilherme et al. 2008).
[00253] Diabetes is associated with damage to various tissues, mainly because hyperglycemia (and hypoglycemia, which can result from excessive or poorly timed doses of insulin) is an important cause of oxidative stress. Due to their ability to protect against oxidative stress, in particular by inducing expression of HO-1, LAR 408, the polymorphic forms, and methods of the present invention can be used in treatments for many complications of diabetes. As mentioned above (Cai et al., 2005), chronic inflammation and oxidative stress in the liver are suspected to be major factors in the development of type 2 diabetes. In addition, PPAR g agonists such as thiazolidinediones are able to reduce insulin resistance. and are known to be effective treatments for type 2 diabetes. In some embodiments, RTA 408, the polymorphic forms, and methods of the present invention can be used as combination therapies with PPARY agonists, such as thiazolidinediones. G. Arthritis
In some embodiments, RTA 408, the polymorphic forms, and methods of the present invention can be used to treat patients with a form of arthritis. In some embodiments, the forms of arthritis that can be treated with RTA 408 and the polymorphic forms of this invention are rheumatoid arthritis (RA), psoriatic arthritis (PAs), spondyloarthropathies (ZPE), including ankylosing spondylitis (AS), reactive arthritis ( ARPE), and enteropathic arthritis (EA), juvenile rheumatoid arthritis (JRA), and early inflammatory arthritis.
[00255] In rheumatoid arthritis, the first signs usually appear in the synovial lining layer, with the proliferation of synovial fibroblasts and their attachment to the articular surface at the articular margin (Lipsky, 1998). Subsequently, macrophages, T cells and other inflammatory cells are recruited into the pool, where they produce a number of mediators, including cytokine interleukin-1 (IL-1), which contributes to chronic sequelae leading to bone and cartilage destruction, and tumor necrosis factor (TNF-a), which plays a role in inflammation (Dinarello, 1998; Arend and Dayer, 1995; van den Berg, 2001). The plasma IL-1 concentration is significantly higher in RA patients than in healthy individuals, and in particular plasma IL-1 levels correlate with RA disease activity (Eastgate et al., 1988). In addition, IL-1 synovial fluid levels are correlated with various radiographic and histological features of RA (Kahle et al., 1992; Rooney et al., 1990).
[00256] Other forms of arthritis include psoriatic arthritis (PA), which is a chronic inflammatory arthropathy characterized by the association of arthritis and psoriasis. Studies have revealed that PSA shares a number of genetic, pathogenic, and clinical characteristics with other spondyloarthropathies (ZPE), a group of diseases that comprise ankylosing spondylitis, reactive arthritis, and enteropathic arthritis (Wright, 1979). The notion that PSA belongs to the SpA group has recently gained further support in imaging studies that demonstrate generalized enthesitis in AP but not AR (McGonagle et al. 1999; McGonagle et al. 1998). More specifically, enthesitis has been thought to be one of the first events to occur in SPAs, leading to bone remodeling and spinal ankylosis, as well as articular synovitis when inflamed entheses are close to peripheral joints. Increased amounts of TNF-α have been reported in both psoriatic skin (Ettehadi et al., 1994) and synovial fluid (Partsch et al., 1997). Recent trials have demonstrated a positive benefit of anti-TNF treatment in both PA (Mease et al., 2000) and ankylosing spondylitis (Brandt et al., 2000).
[00257] Juvenile rheumatoid arthritis (JRA), a term for the most prevalent form of arthritis in children, is applied to a family of diseases characterized by chronic inflammation and hypertrophy of synovial membranes. The term overlaps with, but is not fully synonymous with, the family of diseases referred to as juvenile chronic arthritis and/or juvenile idiopathic arthritis in Europe.
[00258] Polyarticular JRA is a distinct clinical subtype characterized by inflammation and synovial proliferation in multiple joints (four or more), including the small joints of the hands (Jarvis, 2002). This JRA subtype can be serious because of both its multiple joint involvement and its ability to progress rapidly over time. Although clinically distinct, polyarticular JRA is not homogeneous, and patients vary in disease manifestations, age at onset, prognosis, and therapeutic response. These differences most likely reflect a spectrum of variations in the nature of immune and inflammatory attack that can occur in this disease (Jarvis, 1998).
[00259] Ankylosing spondylitis (AS) is a subset of the disease within a broader disease classification of spondyloarthropathy. Patients affected with the various subsets of spondyloarthropathy have disease etiologies that are often very different, ranging from bacterial infections to inheritance. However, in all subgroups, the end result of the disease process is axial arthritis.
[00260] AS is a chronic systemic inflammatory rheumatic disease of the axial skeleton, with or without extraskeletal manifestations. Sacroiliac joints and the spine are mainly affected, but hip and shoulder joints, and less commonly peripheral joints or certain extra-articular structures, such as the eye, vascular system, nervous system, gastrointestinal system, and may also be involved. Aetiology of the disease is not yet fully understood (Wordsworth, 1995; Calin and Taurog, 1998). The etiology is strongly associated with the major histocompatibility class I (MHC I) HLA-B27 allele (Calin and Taurog, 1998). AS affects individuals in the prime of their lives and is feared because of its potential to cause chronic pain and irreversible damage to tendons, ligaments, joints, and bones (Brewerton et al., 1973a; Brewerton. et al., 1973b; Schlosstein et al., 1973). H. Ulcerative Colitis
[00261] In some embodiments, RTA 408, the polymorphic forms and methods of the present invention can be used to treat patients with ulcerative colitis. Ulcerative colitis is a disease that causes inflammation and sores, called ulcers, in the lining of the large intestine. Inflammation usually occurs in the rectum and lower part of the large intestine, but it can affect the entire colon. Ulcerative colitis can also be called colitis or proctitis. Inflammation of the colon becomes empty often, causing diarrhea. Ulcers form where inflammation has killed the cells lining the colon and ulcers bleed and produce pus.
[00262] Ulcerative colitis is an inflammatory bowel disease (IBD), the generic name for diseases that cause inflammation in the small intestine and colon. Ulcerative colitis can be difficult to diagnose because its symptoms are similar to other bowel disorders and to another type of inflammatory bowel disease, Crohn's disease. Crohn's disease differs from ulcerative colitis in that it causes inflammation deeper in the intestinal wall. In addition, Crohn's disease usually occurs in the small intestine, although the disease can also occur in the mouth, esophagus, stomach, duodenum, large intestine, appendix, and anus. I. Crohn's Disease
In some embodiments, RTA 408, the polymorphic forms, and methods of the present invention can be used to treat patients with Crohn's disease. Symptoms of Crohn's disease include intestinal inflammation and the development of intestinal strictures and fistulas; neuropathy often accompanies these symptoms. Anti-inflammatory drugs such as 5-aminosalicylates (eg, mesalamine) or corticosteroids are typically prescribed but not always effective (reviewed in Botoman et al., 1998). Cyclosporine immunosuppression is sometimes beneficial for corticosteroid-resistant or intolerant patients (Brynskov et al., 1989).
[00264] In active cases of Crohn's disease, elevated concentrations of TNF-α and IL-6 are secreted into the blood circulation, and TNF-α, IL-1, IL-6 and IL-8 are overproduced locally by mucosal cells (ID.; Funakoshi. et al., 1998). These cytokines can have far-reaching effects on physiological systems, including bone development, hematopoiesis and liver, thyroid, and neuropsychiatric function. Furthermore, an imbalance of IL-1 b ratio/IL-1ra in favor of the pro-inflammatory IL-1 b has been observed in patients with Crohn's disease (Rogler and Andus, 1998; Saiki et al., 1998 ; Dionne et al., 1998; but see Kuboyama, 1998).
Treatments that have been proposed for Crohn's disease include the use of various cytokine antagonists (eg IL-1ra), inhibitors (eg IL-1b converting enzyme and antioxidants) and antibodies anti-cytokines (Rogler and Andus, 1998; van Hogezand and Verspaget, 1998; Reimund. et al., 1998; Lugering et al., 1998; McAlindon. et al., 1998). In particular, monoclonal antibodies against TNF-α have been tried with some success in the treatment of Crohn's disease (Targan et al., 1997; Pile. et al., 1997; van Dullemen. et al., 1995). These compounds can be used in combination therapy with RTA 408, the polymorphic forms, and the methods of the present disclosure. J. Systemic Lupus Erythematosus
[00266] In some embodiments, RTA 408, the polymorphic forms and methods of the present invention can be used to treat patients with SLE. Systemic lupus erythematosus (SLE) is an autoimmune rheumatic disease characterized by tissue deposition of autoantibodies and from immune complexes that lead to tissue damage (Kotzin, 1996). Unlike autoimmune diseases such as multiple sclerosis and type 1 diabetes mellitus, systemic lupus erythematosus potentially involves multiple organ systems directly, and its clinical manifestations are diverse and variable (reviewed by Kotzin and O'Dell, 1995). For example, some patients may primarily demonstrate rash and joint pain, show spontaneous remissions, and require little medication. At the other end of the spectrum are patients who present with severe and progressive renal involvement that require therapy with high doses of steroids and cytotoxic drugs such as cyclophosphamide (Kotzin, 1996).
[00267] One of the antibodies produced by SLE, IgG anti-edDNA, plays an important role in the development of lupus glomerulonephritis (GN) (Hahn and Tsao, 1993; Ohnishi et al., 1994). Glomerulonephritis is a serious condition, in which the capillary walls of glomeruli clearing blood from the kidney become thickened by additions on the epithelial side of the glomerular basement membrane. The disease is often chronic and progressive and can lead to eventual kidney failure. K. Irritable Bowel Syndrome
[00268] In some embodiments, RTA 408, the polymorphic forms, and methods of the present invention can be used to treat patients with irritable bowel syndrome (IBS). IBS is a functional disorder characterized by abdominal pain and altered bowel habits. This syndrome can begin in young adulthood and can be associated with significant disability. This syndrome is not a homogeneous disease. Instead, subtypes of IBS have been described on the basis of the predominant symptom - diarrhea, constipation, or pain. In the absence of "alarm" symptoms such as fever, weight loss, and gastrointestinal bleeding, limited examination is necessary.
[00269] Increasingly, evidence for the origins of IBS suggests a relationship between infectious enteritis and later development of IBS. Inflammatory cytokines may play a role. In a survey of patients with a confirmed history of bacterial gastroenteritis (Neal et al., 1997), 25% reported persistent change in bowel habits. The persistence of symptoms may be due to psychological stress at the time of acute infection (Gwee et al., 1999).
[00270] Recent data suggest that bacterial overgrowth in the small intestine may also play a role in IBS symptoms. In one study (Pimentel et al., 2000), 157 (78%) of 202 patients with IBS referred for hydrogen breath testing had test results that were positive for bacterial overgrowth. Of the 47 individuals who had follow-up examinations, 25 (53%) reported improvement in symptoms (ie, abdominal pain and diarrhea) with antibiotic treatment. L. Sjogren's Syndrome
[00271] In some embodiments, RTA 408, the polymorphic forms, and methods of the present invention can be used to treat patients with Sjogren's syndrome. Primary Sjogren's Syndrome (SS) is a chronic, autoimmune, slowly progressive, systemic disease that predominantly affects middle-aged women (female to male ratio 9:1), although it can be seen at all ages, including childhood. (Jonsson et al., 2002). The disease is characterized by lymphocyte infiltration and destruction of exocrine glands, which are infiltrates of mononuclear cells, including CD4+, CD8+ lymphocytes, and B cells (Jonsson et al., 2002). In addition, extraglandular (systemic) manifestations are seen in one third of patients (Jonsson et al., 2001).
[00272] In other systemic autoimmune diseases such as RA, critical factors for ectopic germinal centers (GCs) have been identified. Rheumatoid synovial tissues with GCs have been shown to produce chemokines CXCL13, CCL21 and lymphotoxin (LT)-β (detected in cells from the follicular center and mantle zone B). Multivariate analysis of these analytes identified CXCL13 and LT-β as the solitary cytokines predicting GCs in arthritis synovitis (Weyand and Goronzy, 2003). Recently, CXCL13 and CXCR5 in salivary glands have been shown to play a key role in the inflammatory process through the recruitment of B and T cells, thus contributing to lymphoid neogenesis and the formation of ectopic CG in SS (Salomonsson et al., 2002) . M. Psoriasis
[00273] In some embodiments, RTA 408, the polymorphic forms, and methods of the present invention can be used to treat patients with psoriasis. Psoriasis is a chronic skin disease of desquamation and inflammation that affects between 2 and 2.6 percent of the US population, or between 5.8 and 7.5 million people. Psoriasis occurs when skin cells quickly rise from their origin below the skin's surface and accumulate on the surface before they have a chance to mature. Typically, this movement (also called bulking) takes about a month, but bulking psoriasis can occur in just a few days. In its typical form, psoriasis results in patches of red (inflammation) thick skin, covered in silvery scales. These spots, which are sometimes referred to as plaques, usually itch or feel sore. Plaques most commonly occur on the elbows, knees, other parts of the legs, scalp, back, face, palms, and soles of the feet, but they can occur on the skin anywhere on the body. The disease can also affect the nails, toenails, and soft tissue in the genitals and inside the mouth.
[00274] Psoriasis is a skin disease driven by the immune system, especially involving a type of white blood cell called a T cell. Normally, T cells help protect the body against infection and disease. In the case of psoriasis, the T cells are mistakenly put into action and become so active that they trigger other immune responses that lead to inflammation and rapid turnover of skin cells. N. Infectious Diseases
[00275] In some embodiments, RTA 408, the polymorphic forms, and methods of the present disclosure may be useful in the treatment of infectious diseases, including viral and bacterial infections. As noted above, these infections can be associated with severe localized or systemic inflammatory responses. For example, influenza can provoke severe inflammation from lung infection and bacterial can trigger the systemic hyperinflammatory response, including the excessive production of multiple inflammatory cytokines, which is the hallmark of sepsis. Furthermore, compounds of the invention may be useful in inhibiting direct replication of viral pathogens. Previous studies have shown that related compounds such as CDDO can inhibit HIV replication in macrophages (Vazquez et al., 2005). Other studies have indicated that inhibition of NF-kB signaling can inhibit influenza virus replication, and that cyclopentenonic prostaglandins can inhibit viral replication (eg, Mazur. et al., 2007; Pica et al., 2000).
[00276] The present invention relates to the treatment or prevention of each of the diseases/disorders/conditions referred to above in section IV, using the compound RTA 408 or a pharmaceutically acceptable salt thereof, or a polymorphic form of such compound ( such as, for example, any of the polymorphic forms described herein above or below), or a pharmaceutical composition comprising any of the aforementioned entities and a pharmaceutically acceptable carrier (including, for example, the pharmaceutical compositions described above ). V. Pharmaceutical formulations and administration routes
[00277] RTA 408 can be administered by a variety of methods, eg, orally or by injection (eg, subcutaneously, intravenously, intraperitoneally, etc.). Depending on the route of administration, the active compounds can be coated with a material to protect the compound from the action of acids and other natural conditions that can inactivate the compound. They can also be administered by continuous infusion/infusion of a disease or wound site.
[00278] To administer RTA 408 by other than parenteral administration, it may be necessary to coat the compound with, or co-administer the compound with, a material to prevent its inactivation. For example, the therapeutic compound can be administered to a patient in an appropriate carrier, for example, liposomes, or a diluent. Pharmaceutically acceptable diluents include saline and aqueous buffer solutions. Liposomes include water-in-oil-in-water CGF emulsions as well as conventional liposomes (Strejan et al., 1984).
[00279] RTA 408 can also be administered parenterally, intraperitoneally, intraspinally, intracerebrally or. Dispersions can be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under normal conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms.
Sterile injectable solutions can be prepared by incorporating RTA 408 in the required amount in an appropriate solvent with one or a combination of the ingredients enumerated above, as required, followed by filter sterilization. Generally, dispersions are prepared by incorporating the therapeutic compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for preparing sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze drying, which produce a powder of the active ingredient (ie, the therapeutic compound) plus any additional ingredients from a previously filter-sterilized solution.
[00281] RTA 408 can be rendered fully amorphous using a direct spray drying procedure. RTA 408 can be administered orally, for example, with an inert diluent or an assimilable edible carrier. The therapeutic compound and other ingredients may also be enclosed in a hard or soft gelatin capsule, compressed into tablets, or incorporated directly into the patient's diet. For oral therapeutic administration, the therapeutic compound can be incorporated, for example, with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, including hard or soft capsules, elixirs, emulsions, solid dispersions, suspensions, syrups , cookies, and the like. The percentage of the therapeutic compound in the compositions and preparations can, of course, be varied. The amount of the therapeutic compound in such therapeutically useful compositions is such that a suitable dosage will be obtained.
It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suitable as unitary dosages for patients to be treated, each unit containing a predetermined quantity of therapeutic compound calculated to produce the desired therapeutic effect in association with the pharmaceutical carrier required. The specification for the unit dosage forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the therapeutic compound and the particular therapeutic effect to be achieved, and (b) the inherent limitations in the art of composing such a therapeutic compound for the treatment of a selected condition of a patient.
[00283] RTA 408 can also be administered topically to the skin, eyes or mucosa. In some embodiments, the compound can be prepared from a lotion, cream, gel, oil, ointment, ointment, solution, suspension, or emulsion. Alternatively, if delivery site to the lungs is desired the therapeutic compound can be administered by inhalation in dry powder or aerosol formulation.
[00284] RTA 408 will typically be administered at a therapeutically effective dosage sufficient to treat a condition associated with a given patient. For example, the efficacy of a compound can be evaluated in an animal model system that can predict efficacy in treating disease in humans, such as the model systems shown in the examples and figures.
[00285] The actual dosage amount of RTA 408 or composition comprising RTA 408 administered to a patient can be determined by physical and physiological factors such as age, sex, body weight, severity of the condition, the type of disease to be treated, the therapeutic interventions previously or simultaneously, idiopathic of the patient, and the route of administration. These factors can be determined by one skilled in the art. The administering physician will typically determine the concentration of active ingredient(s) in an appropriate composition and dose(s) for the individual patient. Dosage can be adjusted by the individual physician in the event of any complications.
An effective amount will typically range from about 0.001 mg/kg to about 1000 mg/kg, from about 0.01 mg/kg to about 750 mg/kg, from about 100 mg/kg to about from 500 mg/kg, from about 1.0 mg/kg to about 250 mg/kg, from about 10.0 mg/kg to about 150 mg/kg in one or more daily administration doses for one or several days (depending on the course of the mode of administration and the factors discussed above). Other suitable ranges include doses of 1 mg to 10,000 mg per day, 100 mg and 10,000 mg per day, 500 mg and 10,000 mg per day, and 500 mg to 1000 mg per day. In some particular embodiments, the amount is less than 10,000 mg per day, with a range of 750 mg to 9000 mg per day.
[00287] The effective amount may be less than 1 mg/kg/day, less than 500 mg/kg/day, less than 250 mg/kg/day, less than 100 mg/kg/day, less than 50 mg/day kg/day, less than 25 mg/kg/day, or less than 10 mg/kg/day. Alternatively, it can range from 1 mg/kg/day to 200 mg/kg/day. In some embodiments, the amount can be 10, 30, 100, or 150 mg/kg, formulated as a suspension in sesame oil, as described below in Example C1. In some embodiments, the amount may be 3, 10, 30, or 100 mg/kg, administered daily by oral gavage as described below in Examples C2 and C3. In some embodiments, the amount may be 10, 30, or 100 mg µg/kg administered orally as described below in Example C6. For example, with respect to treating diabetic patients, the unit dosage may be an amount that reduces blood glucose by at least 40% compared to an untreated patient. In another embodiment, the unit dosage is an amount that reduces blood glucose to a level that is ±10% of the blood glucose level of a non-diabetic patient.
[00288] In other non-limiting examples, a dose may also comprise from about 1 µg/kg body weight, about 5 µg/kg body weight, about 10 µg/kg body weight, about 50 µg/ kg body weight, about 100 μg/kg body weight, about 200 ug/kg body weight, about 350 μg/kg body weight, about 500 μg/kg body weight, about 1 mg/kg of body weight, about 5 mg/kg of body weight, about 10 mg/kg of body weight, about 50 mg/kg of body weight, about 100 mg/kg of body weight, about 200 mg/kg of body weight, about 350 mg/kg body weight, about 500 mg/kg body weight, to about 1000 mg/kg body weight or more per administration, and any range derivable therefrom. In non-limiting examples of range derivable from the numbers given herein, a range from about 5 mg/kg body weight to about 100 mg/kg body weight, about 5 µg/kg body weight to about 500 mg/kg body weight, etc. can be administered, based on the numbers described above.
[00289] In certain embodiments, a pharmaceutical composition of the present disclosure may comprise, for example, at least about 0.01% RTA 408. In other embodiments, RTA 408 may comprise between about 0.01% to about 75% of the weight of the unit, or between about 0.01% to about 5%, for example, and any range derivable therefrom. In some embodiments, RTA 408 can be used in a formulation, such as a suspension in 0.01%, 0.1%, or 1% sesame oil, as described below in Examples F and G. In some embodiments, RTA 408 they can be formulated for topical administration to the skin or eye, using a suitable pharmaceutical carrier or as a suspension, emulsion, or solution, in concentrations ranging from about 0.01% to 10%. In some embodiments the concentration ranges from about 0.1% to about 5%. The optimal concentration can vary depending on the target organ, the specific preparation, and the condition being treated.
Single or multiple doses of the agent comprising RTA 408 are contemplated. Desired time intervals for multiple dose delivery can be determined by one skilled in the art who have no more than routine experimentation. As an example, patients may be given two doses per day at approximately 12 hour intervals. In some embodiments, the agent is administered once a day. The agent(s) may be administered in a routine pattern. As used herein refers to a routine pattern for a designated predetermined period of time. The routine pattern can encompass periods of time that are identical or that differ in length, as long as the time is predetermined. For example, the routine calendar may involve administration twice a day, every day, every two days, every three days, every four days, every five days, every six days, weekly, monthly, or whatever. number of days or weeks there are between them. Alternatively, the pre-terminated routine pattern may involve administration on a daily basis twice for the first week, followed daily for several months, etc. In other embodiments, the invention provides that the agent(s) can be taken orally and that the timing of it is or is not dependent on food intake. So, for example, the agent can be taken every morning and/or every night, regardless of when the patient has eaten or is going to eat. SAW. combined therapy
[00291] In addition to being used as a monotherapy, RTA 408 and the polymorphic forms described in the present invention may also find use in combination therapies. Effective combination therapy can be achieved with a single pharmaceutical composition or formulation that includes both agents, or with two distinct compositions or formulations, administered at the same time, wherein one composition includes RTA 408 or its polymorphic forms, and the other includes the second agent (s). The other therapeutic modality can be administered before, concurrently with, or after administration of RTA 408 or its polymorphic forms. Therapy using RTA 408 or its polymorphic forms may precede or follow administration of the other agent(s) at intervals ranging from minutes to weeks. In modalities where the other agent and RTA 408 or its polymorphic forms are administered separately, it can generally be ensured that a significant period of time has not expired between the time of each delivery, such that each of the agents would still be able to exert an advantageously combined effect. In such cases, it is considered that one would typically administer RTA 408 or the polymorphic forms and the other therapeutic agent within about 12-24 hours of each other, and more preferably within about 6-12 hours of each other, with a delay time of only about 12 hours being most preferred. In some situations it may be desirable to extend the time period for treatment significantly, however, where several days (2, 3, 4, 5, 6 or 7) to several weeks (1, 2, 3, 4 , 5, 6, 7 or 8) lapse between the respective administrations.
[00292] It is also conceivable that more than one administration of RTA 408 or its polymorphic forms, or other agent will be desired. In this regard, various combinations can be employed. By way of illustration, where RTA 408 or its polymorphic forms is "A" and the other agent is "B", the following permutations based on 3 and 4 total administrations are exemplary: A / B / AB / A / BB / B / AA / A / BB / D / AA / B / BB / B / B / AB / B / A / BA / A / B / BA / B / A / BA / B / B / AB / B / A / AB / A / B / AB / A / A / BB / B / B / AA / A / A / BB / A / D / AA / B / A / AA / A / B / AA / B / B / BB / A / B / BB / B / A / B
[00293] Other combinations are also contemplated. Non-limiting examples of pharmacological agents that can be used in the present invention include any pharmacological agent known to be beneficial in the treatment of cancer. In some embodiments, combinations of RTA 408 or its polymorphic forms with targeted cancer immunotherapy, gene therapy, radiotherapy, chemotherapeutic agent, or surgery are contemplated. Also contemplated is the combination of RTA 408 or its polymorphic forms with more than one of the aforementioned methods, including more than one type of a specific therapy. In some modalities, immunotherapy may be targeted by other cancer antibodies, such as, but not limited to, trastuzumab (Herceptin), alemtuzumab (Campath®), bevacizumab (Avastin), cetuximab (Eribitux®), and panitumumab (Vectibix®) or conjugated antibodies such as ibritumomab tiuxetan (Zevalin), tositumomab (Bexxar®), brentuximab vedotin (Adcetris®), emtansine ado-trastuzumab (Kadcila™), or dititox denileucine (Ontak®). Furthermore, in some embodiments, RTA 408 or its polymorphic forms are envisioned to be used in combination therapies with dendritic cell-based immunotherapy, such as Sipuleucel-T (Provenge®) or adoptive T-cell immunotherapy.
[00294] Furthermore, it is contemplated that RTA 408 or its polymorphic forms are used in combination with a chemotherapeutic agent, such as, but not limited to, anthracyclines, taxanes, methotrexate, mitoxantrone, estramustine, doxorubicin, etoposide, vinblas - tin, carboplatin, vinorelbine, 5-fluorouracil, cisplatin, topotecan, ifosfamide, cyclophosphamide, epirubicin, gemcitabine, vinorelbine, irinotecan, etoposide, vinblastine, pemetrexed, melphalan, capecitabine and oxaliplatin. In some embodiments, RTA 408 or its polymorphic forms are used in combination with radiation therapy, including but not limited to the use of ionizing radiation. In some modalities, the effects of the cancer therapeutic agent are synergistically enhanced through administration with RTA 408 and its polymorphic forms. In some modalities, combination therapies that include RTA 408 are used to treat cancer, including, for example, prostate cancer. See, for example, Example H below.
[00295] In some embodiments, the methods may further comprise (1) contacting a tumor cell with the compound prior to contacting the tumor cell with the second chemotherapeutic agent, (2) contacting a tumor cell with the second chemotherapeutic agent prior to contacting the tumor cell with the compound, or (3) contacting a tumor cell with the compound and the second chemotherapeutic agent at the same time. The second chemotherapeutic agent can, in certain embodiments, be an antibiotic, anti-inflammatory, anti-neoplastic, anti-proliferative, anti-viral, immunomodulatory, or immunosuppressive. In other modalities, the second chemotherapeutic agent may be an alkylating agent, androgen receptor modulator, cytoskeletal disruptors, estrogen receptor modulator, histone deacetylase inhibitor, HMG-CoA reductase inhibitors, inhibitors of prenyl-protein transferase, retinoid receptor modulator, topoisomerase inhibitor, or tyrosine kinase inhibitor. In certain embodiments, the second chemotherapeutic agent is 5-azacytidine, 5-fluorouracil, 9-cis-retinoic acid, actinomycin D, alitretinoin, all-trans-retinoic, Annamicin, Axitinib, belinostat, bevacizumab, bexarotene, bosutinib , busulfan, capecitabine, carboplatin, carmustine, CD437, cediranib, cetuximab, chlorambucil, cisplatin, cyclophosphamide, cytarabine, dacarbazine, dasatinib, daunorubicin, decitabine, docetaxel, dolastatin-10, doxypyrtinicin, doxortinicin, dox , etoposide, gefitinib, gemcitabine, ozogamicin ozogamycin, hexamethylmelamine, Zavedos®, ifosfamide, imatinib, irinotecan, isotretinoin, ixabepilone, lapatinib, LBH589, lomustine, mechlorethamine, melphalan, mercaptoxa- mine, irinotecan 275, neratinib, nilotinib, nitrosourea, oxaliplatin, paclitaxel, plicamycin, procarbazine, semaxanib, semustine, sodium butyrate, sodium phenylacetate, streptozotocin, hydroxamic acid co suberoylanilide, sunitinib, tamoxifen, teniposide, thiopeta, thioguanine, topotecan, TRAIL, trastuzumab, tretinoin, trichostatin A, valproic acid, valrubicin, vandetanib, vinblastine, vincristine, vindesine, or vino.
[00296] Furthermore, combination therapies for the treatment of cardiovascular disease using RTA 408, polymorphic forms, and pharmaceutical compositions of the present description are contemplated. For example, such methods can further comprise administering a pharmaceutically effective amount of one or more cardiovascular drugs in addition to RTA 408, polymorphic forms, and the pharmaceutical compositions of the present disclosure. The cardiovascular drug can be, but is not limited to, for example, a cholesterol-lowering medication, an antihyperlipidemic agent, a calcium channel blocker, an antihypertensive, or an HMG-inhibitor. CoA reductase. In some embodiments, non-limiting examples of cardiovascular medications include amlodipine, aspirin, ezetimibe, felodipine, lacidipine, lercanidipine, nicardipine, nifedipine, nimodipine, nisoldipine and nitrendipine. In other embodiments, other non-limiting examples of cardiovascular drugs include atenolol, bucindolol, carvedilol, clonidine, doxazosin, indoramine, labetalol, methyldopa, metoprolol, nadolol, oxprenolol, phenoxybenzamine, phentolamine, pindolol, prazosin, propranolol, terazosin, tolazoline. In other embodiments, the cardiovascular drug can be, for example, a statin such as atorvastatin, cerivastatin, fluvastatin, lovastatin, mevastatin, pitavastatin, pravastatin, rosuvastatin or simvastatin. VII. Examples
[00297] The following examples are included to demonstrate preferred embodiments of the invention. It should be appreciated by those skilled in the art that the techniques disclosed in the examples which follow represent techniques discovered by the inventor to function well in the practice of the invention, and thus can be considered to constitute preferred modes for its practice. However, those skilled in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments that are disclosed and still obtain a similar or similar result without departing from the spirit and scope of the invention. A. Synthesis of RTA 408 (63415)
RTA 408 (63415)
[00298] Reagents and conditions: (a) (PhO)2 PON3 (DPPA), Et3 N, toluene, at 0°C for 5 min, then at room temperature overnight, ~94%; (b) benzene, 80°C for 2 h; (c) HCl, CH3 CN, room temperature for 1 h; (d) CH3CF2CO2H, DCC, DMAP, CH2Cl2, room temperature overnight, 73% from 401 RTA (4 steps).
[00299] Compound 1: to a toluene solution (400 mL), RTA 401 (which can be prepared according to the methods described, for example, by Honda, et al., 1998; Honda. et al., 2000b; Honda . et al., 2002; Yates et al., 2007, and US Patents 6,326,507 and 6,974,801, which are hereby incorporated by reference) (20.0 g, 40.6 mmol) and Et3N (17.0 mL, 122.0 mmol) were added to a reactor and cooled to 0°C with stirring. Diphenyl-phosphoryl-azide (DPPA) (13.2 mL, 61.0 mmol) was added with stirring at 0°C over 5 min and the mixture was continuously stirred at room temperature overnight (control by HPLC-MS shows no RTA 401 left). The reaction mixture was directly loaded onto a silica gel column and purified by column chromatography (silica gel, 0% to 5% EtOAc in CH 2 Cl 2 ) to give compound 1 (19.7 g, ~ 94%, partially converted to compound 2) as a white foam.
[00300] Compound 2: Compound 1 (19.7 g, ~ 38.1 mmol) and benzene (250 mL) were added to a reactor and heated at 80°C with stirring for 2 h (HPLC-MS control shows none compound 1 on the left). The reaction mixture was concentrated under reduced pressure to obtain crude compound 2 as a solid residue, which was used for the next step without purification.
Compound 3: crude compound 2 (<38.1 mmol) and CH 3 CN (200 mL) were added to a reactor and cooled to 0°C with stirring. HCl (12N, 90 mL) was added at 0°C over 1 min and the mixture was continuously stirred at room temperature for 1 h (HPLC-MS control shows no compound 2 left). The reaction mixture was cooled to 0°C and 10% NaOH (~500 mL) was added with stirring. Then saturated NaHCO3 (1 L) was added with stirring. The aqueous phase was extracted with EtOAc (2 x 500 ml). The combined organic phase was washed with H2O (200 mL), saturated NaCl (200 mL), dried over Na2SO4, and concentrated to give crude compound 3 (16.62 g) as a light yellow foam. which was used for the next step, without purification.
[00302] RTA 408: crude amine 3 (16.62 g, 35.9 mmol), CH3CF2CO2H (4.7388 g, 43.1 mmol), and CH2Cl2 (360 mL) were added to a stirred reactor at temperature environment. Then, dicyclohexylcarbodiimide (DCC) (11.129 g, 53.9 mmol) and 4(dimethylamino)pyridine (DMAP) (1.65 g, 13.64 mmol) were added and the mixture was continuously stirred. at room temperature overnight (HPLC-MS shows no compound 3 check on the left). The reaction mixture was filtered to remove solid by-products and the filtrate was directly loaded onto a silica gel column and purified by column chromatography (silica gel, 0% to 20% EtOAc in Hexanes) twice to give the compound RTA 408 (16.347 g, 73% from 401 RTA over 4 steps) as a white foam: 1H NMR (400 MHz, CD 3 Cl) δ 8.04 (s, 1H), 6.00 (s, 1H) ), 5.94 (s, br, 1H), 3.01 (d, 1H, J = 4.8 Hz), 2.75-2.82 (m, 1H), 1.92-2.18 ( m, 4H), 1.69-1.85 (m, 7H), 1.53-1.64 (m, 1H), 1.60 (s, 3H), 1.50 (s, 3H), 1 1.42 (s, 3H), 1.11-1.38 (m, 3H), 1.27 (s, 3H), 1.18 (s, 3H), 1.06 (s, 3H), 1. 04 (s, 3H), 0.92 (s, 3H); m/z 555 (M + 1). B. Pharmacodynamics
[00303] A summary of in vitro and in vivo studies to assess the primary pharmacodynamic effects of RTA 408 is provided below. 1. Effects of RTA-408 on KEAP1 Nrf2 and NF-kB in Vitro
[00304] The inhibition of IFN-y-induced NO production by targets is Nrf2-dependent (Dinkova-Kostova, 2005). RAW264.7 macrophages were plated in 96-well plates at 30,000 cells/well, in triplicate, in RPMI 1640 medium supplemented with 0.5% FBS and incubated at 37°C with 5% CO2. The next day, cells were pretreated with DMSO (vehicle) or RTA 408 for 2 h, followed by treatment with 20 ng/ml mouse IFN-y for 24 h. Nitrite (NO2-) levels in the media were measured as an indicator for nitric oxide using the Griess reagent system (cat No. G2930, Promega) according to the manufacturer's instructions, since nitrite is a primary, stable NO degradation product. Cell viability was assessed using the WST-1 Cell Proliferation Reagent (cat No. 11644807001, Roche Applied Science) according to the manufacturer's instructions. IC50 values were determined based on suppression of IFN-Y-induced nitric oxide production normalized to cell viability. Treatment with RTA 408 resulted in a dose-dependent suppression of in-duction of IFN-y NO production, with a mean IC50 value of 3.8 ± 1.2 nM. The results of a representative experiment are shown in FIG. 1. The IC50 for the RTA 408 value was found to be 45% to 65% lower than the IC50 values for compounds 63170 (8 ± 3 nM), 63171 (6.9 ± 0.6 nM), 63179 ( 11 ± 2 nM), and 63189 (7 ± 2 nM). 63170, 63171, 63179, and 63189 are the compounds of the formulas:

2. Effect of RTA 408 on Nrf2 Target Genes
[00305] RTA 408 was tested in two different reporter luciferase assays to assess ARE activation. The first reporter luciferase was tested under the control of a single promoter derivative of the human NQO1 gene, which allows quantitative assessment of the endogenous activity of the transcription factor Nrf2 in cultured mammalian cells. Luciferase Firefly expression of NQO1-ARE controlled luciferase reporter plasmid by binding Nrf2 to a specific enhancer sequence corresponding to the antioxidant response element (ARE) that has been identified in the promoter region of human NADPH: quinone oxidoreductase 1 (NQO1 gene ) (Xie et al., 1995). The NQO1-ARE-luciferase reporter plasmid was constructed by inserting the human NQO1-ARE (5'-CAGTCACAGTGACTCAGCAGAATCTG-3') into the pluc-MCS vector using HindIII/Xhol cloning sites (GenScript Corp, Piscataway, NJ). The human HuH-7 hepatoma cell line, maintained in DMEM (Invitrogen) supplemented with 10% FBS and 100 U/ml (each) of penicillin and streptomycin, was transiently transfected using Lipofectamine 2000 (Invitrogen) with the NQO1-ARE luciferase from the reporter plasmid and the plasmid pRL-TK, which constitutively expresses Renilla luciferase and is used as an internal control for normalization of transfection levels. Thirty hours after transfection, cells were treated with RTA 408 for 18 h. Firefly and Renilla Luciferase activity was assayed by Dual-Glo Luciferase Assay (cat No. E2920, Promega), and the luminescence signal was measured in an L-Max II luminometer (Molecular Devices). Firefly luciferase activity was normalized to Renilla activity, and double induction via a vehicle control (DMSO) of normalized Firefly activity was calculated. FIG. 2a shows a dose-dependent induction of luciferase activity by RTA 408 in this cell line. Values represent the mean of three independent experiments. Twenty percent less RTA 408 (12 nM) than 63189 (14.9 nM) was needed to increase transcription from the NQO1 ARE in Huh-7 cells twice. Similarly, 2.1-2.4 times less than 63170 RTA 408 (25.2 nM) and 63179 (29.1 nM), respectively, was required to increase transcription from the NQO1 ARE in cells Huh-7 twice.
[00306] The activating effect on the luciferase reporter RTA 408 was also evaluated in the AREc32 reporter cell line. This cell line is derived from MCF-7 human breast carcinoma and the cells are stably transfected with a Firefly luciferase reporter gene under the transcriptional control of eight copies of the mouse ARE GSTA2 sequence (Wang, et al. 2006, which is here by incorporating reference). Following treatment with RTA 408 for 18 h, Firefly luciferase activity was measured using the ONE-Glo Luciferase Assay System (Promega, Catalog No. E6110) according to the manufacturer's instructions. A dose-dependent response was observed in the AREc32 reporter cell line (FIG. 2b). A ~2-fold induction of luciferase activity was evident following treatment with 15.6 nM RTA 408 in the NQO1-ARE and GSTA2-ARE reporter assay system. By looking at the results of the GSTA2-ARE (AREc32) luciferase activity study, the effects of 63,415 (RTA 408) on GSTA2-ARE induction can be directly compared to that of RTA 402, 63170, 63171, 63179, 63189 and together with WST1 Feasibility Studies (Figs. 3a-f). Compared to RTA values 402, 63415 showed the rapid induction of GSTA2-ARE-mediated transcription of the five comparison compounds at a concentration of 93 nM required to achieve 4-fold induction in the luciferase reporter assay. All other compounds showed a similar induction only at much higher concentrations with 63170 needing a concentration of 171 nM, 63171 needing a concentration of 133 nM, 63179 needing a concentration of 303 nM and 63189 needing a concentration of 174 nM to achieve a 4-fold induction of luciferase activity. These values correspond to a 1.86 (63415), 3.40 (63170), 2.65 (63171), 6.05 (63179) and 3.47 (63189) double increase in the amount of active compound needed compared to RTA 402 to take the same amount of activity.

[00307] RTA 408 has also been shown to increase transcription levels of known Nrf2 target genes in the human fetal lung fibroblast of HFL1 and human bronchial epithelial cell lines BEAS-2B. HFL1 cells were cultured in F-12K medium supplemented with 10% fetal bovine serum and 1% penicillin-streptomycin. BEAS-2B cells were cultured in DMEM/F-12 media supplemented with 10% fetal bovine serum and 1% penicillin-streptomycin. Cells were plated in 6-well plates at a density of 2.5 x 10 5 cells/well. The next day, cells were treated with DMSO (vehicle) or RTA 408 (7.8, 15.6, 31.3, 62.5, or 125 nM) for 18 h. Each well received the same volume of vehicle. After treatment, the medium was removed and cells were harvested using RLT buffer (Qiagen). Lysates were homogenized using QIAShredder columns (Qiagen, Catalog # 79654) and RNA was isolated using RNeasy Mini kits (Qiagen, Catalog # 74104). For reverse transcription, RNA (1 ng) was combined with oligo (dT) 12-18 primer and H 2 O in a final volume of 23.25 mL. The mixture was heated to 70°C for 10 min and then placed on ice. A master mix containing 8 µl 5X 1 r chain buffer, 2 µl 1 mg/ml BSA, 2 µl 20 mM DTT, 4 µl 5 mM dNTP mix, 0.25 ml RNaseOUT™ and 0.5 ul SuperScript ® II reverse transcriptase was added to the RNA mixture and incubated at 42°C for 1 h. The reaction was inactivated by heating at 70°C for 10 min. The reaction mixture was diluted 1:3 with H 2 O, prior to use, in qPCR. 2.5 ul of the diluted reverse transcription reaction was combined with a set of PCR primers (0.36 uM final concentration), 2X iQ ™ SYBR® Green Supermix (Bio-Rad, Catalog # 170-8885) and H 2 0 for a final volume of 20 ul The sequences for the PCR primers are as follows: Cysteine glutamate ligase, subunit modifier (GCLM), sense primer 5'-GCTGTGGCTACTGCGGTATT-3' (SEQ ID NO : 1) antisense primer 5'-ATCTGCCTCAATGACACCAT-3' (SEQ ID NO: 2); heme oxygenase-1 (HMOX1) sense primer 5'-TCCGATGGGTCCTTACACTC-3' (SEQ ID NO: 3), reverse primer 5'-TAGGCTCCTTCCTCCTTTCC-3' (SEQ ID NO: 4); NAD(P)H dehydrogenase, a quinone (NQO1) sense primer 5'-AAAACACTGCCCTCTTGTGG-3' (SEQ ID NO: 5), reverse primer 5'-GTGCCAGTCAGCATCTGGTA-3' (SEQ ID NO: 6); ribosomal protein S9 (RPS9) sense primer 5'-GATGAGAAGGACCCCACGGCGTCTG-3' (SEQ ID NO: 7), reverse primer 5'-GAGACAATCCAGCAGCCAGGAGGG-3' (SEQ ID NO: 8);Thioredoxin Reductase 1 (TXNRD1 ) sense primer 5'-ATTGCCACTGGTGAAAGACC-3' (SEQ ID NO: 9), reverse primer 5'- ACCAATTTTGTTGGCCATGT-3' (SEQ ID NO: 10). All primers previously validated for specificity and amplification efficiency. The cDNA was amplified using the following cycling conditions: (95°C for 3 min, 44 cycles of 95°C for 30 sec, 60°C for 15 sec, 72°C for 15 sec, followed by a curve of melting from 55°C to 95°C in 0.5°C increments). The relative abundance of each Nrf2 target gene was determined using the comparative CT (ΔΔC T) method. PCR reactions were performed in triplicate wells for each sample. Two independent experiments were performed using the conditions described above. Treatment of HFL1 lung fibroblasts with RTA 408 for 18 h resulted in increased expression of several genes, including the Nrf2 target NQO1, HMOX1, GCLM, and TXNRD1, as measured by quantitative PCR (FIGS. 4a-d) . For all genes tested, induction by RTA 408 was dose-dependent and evident at concentrations as low as 15.6 nM. Treatment of BEAS-2B bronchial epithelial cells with RTA 408 for 18 hours resulted in a dose-dependent increase similar to all Nrf2 target genes evaluated (figures 5a d.). RTA 408 also increased target gene expression in normal human mesangial Nrf2 cells (nHMC), the mouse BV2 microglia cell line, and human SH-SY5Y neuroblastoma cell line at similar concentrations.
Nrf2s Target protein levels of NQO1 and HMOX1 were measured in SH-5Y5Y and BV-2 cells by Western blotting after treatment with RTA 408 SH-SY5Y cells were cultured in 6-well plates at a density of 4 x 10 5 cells per well. BV-2 were plated in 6-well plates at a density of 2.5 x 10 4 cells per well. Twenty-four (BV-2) or 48 (SH-SY5Y) h after plating, cells were treated with RTA 408 for 24 h. After treatment, cells were washed twice with cold PBS and harvested in lysis buffer. Cells were sonicated and debris cleared by centrifugation (10 min @ 18,000 rcf, Beckman Coulter, microfuge 18 centrifuge). Total protein in the supernatant was determined using the Bio-Rad protein reagent, with BSA as a standard. Equal amounts of total cellular protein were separated on SDS-PAGE, and proteins were transferred to nitrocellulose membrane. Membranes were blocked for 1 hour in TBST (1 x TBS with 0.1% Tween-20) containing 5% milk, washed 3 times with TBST and incubated with primary antibodies overnight at 4°C. Antibody was from NQO1 Abcam (#AB2346); HMOX1 (HO-1) was from antibody from Santa Cruz (# sc-10789); actin antibody was from Millipore (#MAB 1501). After washing with TBST, secondary antibodies were added in TBST + 5% milk for 1 h at room temperature. AffiniPure goat anti-rabbit or anti-mouse IgG secondary antibodies were from Jackson ImmunoResearch (catalog #111-035-144 and #115-035-146, respectively). Membranes were washed in TBST, developed using ECL, and exposed to X-ray film. Treatment with RTA 408 also increased NQO1 protein levels in SH-SY5Y cells in a dose-dependent manner (Fig. 6a). HMOX1 protein was not detected in RTA-408 treated or untreated SH-SY5Y cells. In BV2 cells, treatment with RTA 408 NQO1 and increased levels of HMOX1 proteins in concentrations up to 125 nM (Fig. 6b). The EC50 value for induction of Nrf2 protein expression in SK-N-SH cells by RTA 408 (56.4 nM) was 45%-65% lower than the EC50 values for 63171 (122 nM), 63189 (102 nM), and 63179 (126 nM). The same amount of 63170 (54.6 nM) was required.
The EC 50 was measured using an NQO1 Western cell assay where the cells were incubated with the compound under evaluation for three days. After incubation with the compound of interest, cells were reacted with NQO1 mouse antibody and then the next day, cells were reacted with IRDye-800CW-anti-mouse IgG antibody. Target signals were visualized and then analyzed.
Consistent with the induction of target genes and corresponding Nrf2 protein products, treatment of rat RAW264.7 macrophage cells for 24 h increased NQO1 enzymatic activity in a dose-dependent manner, with an increase evident at the time 7.8 nM (figure 7.). NQO1 enzymatic activity was measured by a modified Prochaska assay (Prochaska and Santamaria, Anal Biochem. 169: 328-336, 1988, which is incorporated herein by reference).
[00311] Taken together, these data from several cell lines demonstrate that treatment with RTA 408 increases transcriptional activity controlled by antioxidant response elements, increases expression of Nrf2 target genes, and increases the activity of NQO1, a product of the Nrf2 target gene. 2. Effect of RTA 408 on Cell Redox Capacity Markers
[00312] Glutathione and NADPH are critical factors necessary for the maintenance of cellular redox capacity. Several genes involved in glutathione synthesis (eg, GCLC and GLCM) and NADPH [eg, hexose-6-phosphate dehydrogenase (H6PD) and malic enzyme 1 (ME1)] have been shown to be regulated by Nrf2 (Wu, 2011). The effect of the RTA 408 treatment on total glutathione levels was evaluated in the rat AML-12 hepatocyte cell line using the GSH-Glo™ Glutathione Assay kit (Promega, Catalog # V6912) according to the manufacturer's instructions. Treatment of AML-12 cells for 24 h with RTA 408 increased total cellular glutathione levels in a dose-dependent manner (figure 8.). Data presented are representative of two independent experiments. A >2-fold increase in total glutathione was observed at RTA 408 concentrations as low as 15.6 nM. The EC50 value using a RAW264.7 mouse model for induction of glutathione levels by RTA 408 (9.9 nM) was 22% to 57% lower than the EC50 values for 63,170 (12.1 nM), 63171 ( 23.2 nM) and 63189 (16 nM).
[00313] The effect of RTA 408 treatment on NADPH levels, as measured by the absorbance of a redox sensitive dye, WST-1 (Roche Applied Science, Catalog #11644807001), was evaluated in HCT-116 cells. Absorbance WST-1 is used to assess cell viability by measuring the glycolytic production of NAD(P)H in viable cells. Therefore, in situations where NADPH production increases in the absence of any effect on cell viability, WST-1 also increases uptake (Berridge et al. 1996, which is incorporated herein by reference). Several genes involved in NADPH production have also been shown to be regulated by Nrf2 (Thimmulappa et al., 2002; Wu, et al., 2011, both of which are incorporated herein by reference). RTA 408 during 24 h of treatment increased WST-1 absorbance in a dose-dependent manner (figure 9.), suggesting that NADPH levels were increased.
[00314] The effect of RTA 408 on the expression of genes involved in NADPH synthesis pathways was also evaluated in this study. HCT-116 cells were treated with RTA 408 for 24 h, and mRNA levels of H6PD, phosphogluconate dehydrogenase (PGD), transketolase (TKT), and ME1 were measured using quantitative PCR. 6-well plates at a density of 3 x 10 5 cells/well. The next day, cells were treated with DMSO (vehicle), 10 nM RTA 408, or 50 nM RTA 408 for 24 h. Each well received the same volume of vehicle. After treatment, the medium was removed and cells were harvested using RLT buffer (Qiagen). Lysates were homogenized using QIAShredder columns (Qiagen, Catalog # 79654) and RNA was isolated using RNeasy Mini kits (Qiagen, Catalog # 74104). For reverse transcription, RNA (1 ng) was combined with oligo (dT) 12-18 primer and H 2 O in a final volume of 23.25 mL. The mixture was heated to 70°C for 10 min and then placed on ice. A master mix containing 8 μl 5X first strand buffer, 2 μl 1 mg/ml BSA, 2 μl 20 mM DTT, 4 μl 5 mM dNTP mix, 0.25 ml RNaseOUT™ and 0.5 ml Superscript® II reverse transcriptase was added to the RNA mixture and incubated at 42°C for 1 h. The reaction was inactivated by heating at 70°C for 10 min. The reaction mixture was diluted 1:3 with H 2 O, prior to use, in qPCR. 2.5 μl of the diluted reverse transcription reaction was combined with a set of PCR primers (0.36 uM final concentration), 2X iQ ™ SYBRÒ green Supermix (Bio-Rad, Catalog # 170-8885) and H 2 O to a final volume of 20 ml. The sequences for the PCR primers are as follows: ribosomal protein S9 (RPS9) sense primer 5'-GATGAGAAGGACCCCACGGCGTCTG-3' (SEQ ID NO: 7), reverse primer 5'-GAGACAATCCAGCAGCCCAGGAGGG-3' (SEQ ID NO: 7) : 8); Hexose-6-phosphate dehydrogenase (H6PD) forward primer 5'-GAGGCCGTGTACACCAAGAT-3' (SEQ ID NO: 11), reverse primer 5'-AGCAGTGGGGTGAAAATACG-3' (SEQ ID NO: 12), Phosphogluconate dehydrogenase (PGD) 5' sense primer - AAGGCACTCTACGCTTCCAA-3' (SEQ ID NO: 13), reverse primer 5'-AGGAGTCCTGGCAGTTTTCA-3' (SEQ ID NO: 14), Transketolase (TKT) sense primer forward 5'-CATCTCCGAGAGCAACATCA-3' (SEQ ID NO: 15), reverse primer 5'-TTGTATTGGCGGCTAGTTCC-3' (SEQ ID NO: 16); malic enzyme 1 (ME1) sense primer 5'- TATATCCTGGCCAAGGCAAC-3' (SEQ ID NO: 17) antisense primer 5'-GGATAAAGCCGACCCTCTTC-3' (SEQ ID NO: 18). All primers previously validated for specificity and amplification efficiency. The cDNA was amplified using the following cycling conditions: (95°C for 3 min, 44 cycles of 95°C for 30 sec, 60°C for 15 sec, 72°C for 15 sec, followed by a curve of melting from 55°C to 95°C in 0.5°C increments). The relative abundance of each target gene was determined using the comparative CT method (ΔΔCT). PCR reactions were performed in triplicate wells for each sample. Two independent experiments were carried out using the conditions described above. Treatment with RTA 408 resulted in a dose-dependent increase in the expression of genes involved in NADPH synthesis (FIGS. 10a-d).
[00315] In summary, treatment with RTA 408 increased total glutathione levels in AML-12 hepatocytes and increased WST-1 absorbance, a marker of NADPH production, in HCT-116 cells. This observation correlated with an increase in the expression of several genes that encode key enzymes involved in NADPH synthesis. 3. Effect of RTA 408 on TNFα-induced NF-kB signaling
[00316] NF-kB is a transcription factor that plays a central role in the regulation of many immune and inflammatory responses. RTA 402 and other goals have been shown to inhibit pro-inflammatory NF-kB signaling from a variety of cell lines (Shishodia, 2006; Ahmad, 2006; Yore, 2006). Using the NIH3T3/NF-kB-luc mouse cell line (Panomics), the effects of RTA 408 and compounds 63171, 63179, 63170, and 63189 on the NF-kB-Luc reporter were explored. The NIH3T3/NF-kB-luc cell line maintains a chromosomal integration of a Firefly luciferase reporter construct regulated by eight copies of the NF-kB response element. The effects of these compounds can be quantified by measuring the NF-kB IC 50 value. RTA 408 showed a 1.2 uM IC 50, which when normalized to viability showed an IC 50 of 1.4 uM. The other four compounds showed NIH3T3/NF-kB IC 50 values of 1.7, 0.2, 1.1, and 1.1 uM, which when viability normalized showed IC 50 values of 1.8, 0, 6, 1.1, and 1.0 µM, respectively. RTA 408 and its effects on NF-kB are represented as a dose shift function and times relatively well as WST1 and WST1/2 are shown in Figs. 11a & b. The effect of RTA 408 on TNFα-induced NF-kB signaling was evaluated in He-La/NF-kB-Luc cells, a human cervical adenocarcinoma cell line stably transfected with a luciferase reporter under the control of a multi-element construct. NF-kB transcriptional response. HeLa/NF-kB-Luc cells were pretreated for 1 h with RTA 408, followed by treatment with TNF-α (10 ng/ml) for an additional 5 h. After treatment, luminescence was measured, and the effect of pretreatment RTA 408 on TNF-α-induced luciferase activity was determined. The mean and standard deviation results of three independent experiments are shown in FIG. 12 TNF-α induced LAR 408 dose-dependently inhibited NF-kB activation, with an IC 50 value of 517 ± 83 nM. Similar results were seen in another NF-kB reporter cell line (A549/NF-kB-Luc) where RTA 408 inhibited TNF-α-induced NF-kB activation, with an IC50 value of 627 nM (range 614- 649 nM). RTA 408 was 1.61.8-fold more efficient in reducing the expression of the NF-kB promoter reporter in HeLa/NF-kB-Luc cells than 63189 (854 nM) and 63170 (953 nM), respectively. Other experiments with the human A549 cell line showed an IC 50 for RTA 408 as 1.7 pM and a value that has been normalized to viability to 1.7 pM. The IC 50 of RTA 408 showed activity similar to 63189, 63179, 63171, and 63170, which showed IC 50 values of 1.1, 1.4, 2.0, and 1.0, respectively, that when these values were normalized for viability, the assay showed 1.2, 1.5, 2.1 and 1.1 uM IC 50, respectively. The folding shift for NF-kB as a function of RTA 408 concentration along with the WST1 and WST1/2 curves were plotted and are shown in FIG. 13a & b.
[00317] The effect of RTA 408 on TNF-α induced phosphorylation of iKBα, a key step in the activation of the NF-kB pathway, was also evaluated in HeLa cells. HeLa cells were pretreated with RTA 408 for 6 hours, followed by treatment with TNF-α (20 ng/ml) for 5 min. Total phosphorylated and IKBα levels were assessed by Western blot. Primary IKBα antibodies were from Santa Cruz-(sc-371), pIKBα antibodies were from Cell Signaling (9246), actin antibody was from Millipore (MAB 1501). Affini-pure goat anti-rabbit (IgG) peroxidase conjugated peroxidase-conjugated and Affini-pure goat anti-mouse IgG secondary antibodies were purchased from Jackson ImmunoResearch. Protein spots were developed using ECL, and exposed to X-ray film. Consistent with the results of the luciferase reporter assay, RTA 408 inhibited TNF-α induced phosphorylation of IKBα in a dose-dependent manner (Figure 14.).
[00318] RTA 408 has also been shown to inhibit other pro-inflammatory signaling pathways, such as the IL-6-induced signal transducer and the transcriptional phosphorylation activator 3 (STAT3) and ligand-induced osteoclastogenesis receptor activator NF-kB (RANKL). In HeLa cells, pretreatment with 1 µM RTA 408 for 6 h inhibited IL-6-induced STAT3 phosphorylation. STAT3 (124H6) and phospho-STAT3 (Tyr705) monoclonal antibodies were from Cell Signaling Technology. Peroxidase-conjugated pure Affini goat anti-rabbit IgG and peroxidase-conjugated pure Affini goat anti-mouse IgG were from Jackson ImmunoResearch. Osteoclastogenesis is a multi-step differentiation process that results from the binding of RANKL to its receptor, RANK, in cells of hematopoietic origin. This results in the activation of NF-kB and MAPK, which in turn increases transcription of osteoclast-specific target genes, including tartrate-resistant acid phosphatase (TRAP). The effect of RTA 408 on RANKL-induced osteoclastogenesis was evaluated in the rat macrophage cell line RAW264.7. RAW 264.7 were plated in 24-well plates at a density of 5000 cells/well. The next day, cells were treated with RTA 408 for 2 h and then treated with 50 ng/ml of recombinant mouse RANKL (R & D Systems). Treated cells were incubated for four days to allow differentiation into osteoclasts. Differentiation into osteoclasts was assessed by measuring the activity of TRAP. Briefly, 90 µl of conditioned cell culture medium was removed from each test well and aliquoted into triplicate wells (30 µl/well) of a 96-well plate. 170 ml of TRAP assay buffer (Kamiya Biomedical) was then added to each well and the plate was incubated at 37°C for 3 hours. After incubation, the absorbance at 540 nm was determined using a Spectramax M2 plate-reading spectrophotometer. RTA 408 dose-dependently inhibited RANKL-induced TRAP activity and osteoclast formation, with an IC 50 of ~5-10 nM. 4. Effect of RTA 408 on Expression of Transaminase Enzymes Encoding Genes
Increased transaminases were observed in the 28-day toxicity studies with RTA 408 in rats and, to a much lesser extent, in monkeys. Similar results were observed after oral administration of a related MA (methyl bardoxolone) in humans (Pergola, 2011). One hypothesis for this effect is that it aims directly or indirectly to increase the expression of the transaminase gene in the absence of cell toxicity. To assess whether treatment with RTA 408 affects transaminase mRNA levels, mouse AML-12 hepatocytes were treated with RTA 408 for 18 h, and mRNA levels of genes encoding transaminases were measured using quantitative PCR. AML-12 cells were plated in 6 wells in 3 x 10 5 cell culture plates per well, using 2 ml of medium per well. The following days cells were treated with DMSO (vehicle) or 250 nM and 500 nM RTA 408 for 18 h at 37°C. Each well received 0.1% DMSO. Three independent duplicate experiments were performed. After treatment, the medium was removed and cells were harvested using RLT buffer (Qiagen). Lysates were homogenized using QIAShredder columns (Qiagen, Catalog # 79654) and RNA was isolated using RNeasy Mini kits (Qiagen, Catalog # 74104). For reverse transcription, RNA (1 ng) was combined with oligo (dT) 12-18 primer and H2O in a final volume of 23.25 mL. The mixture was heated to 70°C for 10 min and then placed on ice. A master mix containing 8 μl 5X first strand buffer, 2 μl 1 mg/ml BSA, 2 μl 20 mM DTT, 4 μl 5 mM dNTP mix, 0.25 ml RNaseOUT™ and 0.5 ml Superscript® II reverse transcriptase was added to the RNA mixture and incubated at 42°C for 1 h. The reaction was inactivated by heating at 70°C for 10 min. The reaction mixture was diluted 1:3 with H2O, prior to use, in qPCR. 2.5 μl of the diluted reverse transcription reaction was combined with a set of PCR primers (0.36 uM final concentration), 2X iQ ™ SYBRÒ green Supermix (Bio-Rad, Catalog # 170-8885) and H 2 O to a final volume of 20 ml. The sequences for the PCR primers are as follows: ribosomal protein L19 (Rp19) sense primer 5'-TCAGGCTACAGAAGAGGCTTGC-3' (SEQ ID NO: 19), reverse primer 5'-ACAGTCACAGGCTTGCGGATG-3' (SEQ ID NO: 20); NAD(P)H-dehydrogenase, a quinone (NQO1) sense primer 5'-TCGGGCTAGTCCCAGTTAGA-3' (SEQ ID NO:21), reverse primer 5'-AAAGAGCTGGAGAGCCAACC-3' (SEQ ID NO:22); glutamic pyruvic transaminase 1 (GPT1 or Alt1) forward primer 5'-CACGGAGCAGGTCTTCAACG-3' (SEQ ID NO: 23), reverse primer 5'-AGAATGGTCATCCGGAAATG-3' (SEQ ID NO: 24); glutamic pyruvic transaminase 2 (GPT2 or Alt2) forward primer 5'-CGCGGTGCAGGTCAACTACT-3' (SEQ ID NO: 25), reverse primer 5'-CCTCATCAGCCAGGAGAAAA-3' (SEQ ID NO: 26); glutamate-oxaloacetate transaminase 1 (Got1 or AST1) sense primer 5'-GGCTATTCGCTATTTTGTGT-3' (SEQ ID NO: 27), reverse primer 5'-GACCAGGTGATTCGTACAAT-3' (SEQ ID NO: 28); glutamate-oxaloacetate transaminase 2 (Got2 or ast2) sense primer 5'-AGAGTCCTCTTCAGTCATTG-3' (SEQ ID NO: 29), reverse primer 5'-ATGATTAGAGCAGATGGTGG-3' (SEQ ID NO: 30). All primers previously validated for specificity and amplification efficiency. The cDNA was amplified using the following cycling conditions: (95°C for 3 min, 44 cycles of 95°C for 30 sec, 60°C for 15 sec, 72°C for 15 sec, followed by a cur- melting range from 55°C to 95°C in 0.5°C increments). The relative abundance of each target gene was determined using the comparative CT method (ΔΔCT). PCR reactions were performed in triplicate wells for each sample. Treatment with RTA 408 increased the mRNA levels of alanine transaminase 1 (Alt1 or GPT1) and aspartate transaminase 1 (AST1 or Got1) (FIGS. 15a, c). RTA 408 had no effect on alanine transaminase 2 mRNA (ast2 or Got2) levels and reduced aspartate transaminase 2 mRNA levels (FIG. 15b, d) (GPT2 or Alt2). These results demonstrate that RTA 408, at the concentrations tested (250 nm or 500 nm), affects the expression of the transaminase gene in vitro. 6. Effect of RTA 408 on levels of glycolytic intermediates
[00320] Studies in diabetic mice have shown that bardoxolone methyl increases muscle-specific insulin-stimulated glucose uptake (Saha, 2010). In humans, a greater percentage of patients receiving bardoxolone methyl reported experiencing muscle cramps compared to patients receiving placebo (Pergola, 2011). Muscle spasms have also been reported in diabetic patients after insulin administration, suggesting a possible association with muscle glucose metabolism. The effect of RTA 408 on glycolytic metabolism was evaluated by evaluating the levels of lactate and pyruvate in C2C12 muscle cells of bred rodents. To measure lactate levels, differentiated C2C12 myotubes were treated with 1 mM or 2 µM RTA 408 or insulin for 3 h at 37°C. Buffer was removed and saved for measuring extracellular lactate levels. Cell debris was pelleted by centrifugation (10 min at 14000 rpm) before measuring lactate. To measure intracellular lactate, cells were suspended in 0.1% Triton X-100 in PBS and lysed by cutting with a 25-gauge needle. The cell lysate was centrifuged (10 min at 14,000 rpm, 4°C) and lactate was measured in the supernatant. Intracellular and extracellular lactate was measured using the Lactate Assay Kit (BioVision, Catalog # K607-100). Similar to insulin treatment, treatment of differentiated C2C12 myotubes with 1 µM or 2 µM RTA 408 for 3 h significantly increased intracellular and extracellular lactate levels in a dose-dependent manner.
To measure pyruvate levels, differentiated C2C12 myotubes were treated with 250 or 500 nM RTA 408 or 100 nM insulin for 18 h. After drug treatment, the medium was removed and cells were washed with PBS. Cells were lysed in Pyruvate Assay Buffer (Pyruvate Assay Kit, BioVision, Catalog # K609-100). Cell lysates were centrifuged (10 min at 14,000 rpm, 4°C) and pyruvate levels were measured in the supernatant. Treatment of differentiated C2C12 myotubes with 250 nM or 500 nM RTA 408 for 18 h also significantly increased (P<0.0001, marked by asterisks) intracellular pyruvate levels in a dose-dependent manner (Fig. 16.). Together, these results demonstrate that RTA 408, at the concentrations tested, can affect glycolytic intermediates in vitro from muscle; however, it is unclear how the results of this in vitro system at the concentrations tested RTA 408 relate to the potential effects on glucose metabolism at clinically relevant doses in humans. 7. In Vitro Evaluation of RTA 408 efflux by MRP-1
[00322] One of the characteristics of a drug candidate is the efflux ratio of the compound. The efflux ratio measures how easily the compound is transported across a membrane. The MRP-1 protein, or multi-drug resistance-assistance protein 1, is one of a family of proteins that contribute to facilitating the transport of organic anions and other small molecules across cell membranes. The higher efflux ratio usually means that the drug candidate is more easily transported out of the membrane and less available to modulate intracellular processes. Similar proteins also regulate the transport of compounds across the blood-brain barrier. The efflux ratio of MRP-1 to RTA 408 (1.3) was experimentally determined to be about ten times less than 63170 (10) and 63171 (11.2) and more than 40 times less than 63179 (56.5) and 63.189 (57.1). Without being bound by theory, RTA 408 may not be a good substrate for MRP-1 and/or a candidate for p-glycoprotein mediated efflux at the blood-brain barrier. In some modalities, RTA 408 can be used to treat central nervous system (CNS) disorders. C. Protective effects of RTA 408 in animal models of lung disease
[00323] RTA 408 has been tested in various animal models of lung disease to assess its potential efficacy in the lung. For all studies, RTA 408 was administered orally daily in sesame oil at dose levels ranging from 3 to 150 mg/kg. In most cases, RTA 408 was administered starting several days before the induction of the lung injury response. 1. LPS-induced pulmonary inflammation in mice
[00324] RTA 408 was tested in two studies of LPS-induced lung inflammation in mice. In the first study, intended to be a preliminary dose range locator, RTA 408 (30, 100, or 150 mg/kg) was administered orally once daily for three days, followed by LPS administration 1 h after the final dose. . Bronchoalveolar lavage (BAL) fluid was collected 20 h after LPS administration (21 h after the final dose of RTA 408) and assessed for levels of pro-inflammatory markers (eg IL-6, IL-12p40, TNF-α and RANTES) using LuminexTM technology. RTA 408 treatment resulted in a significant reduction in IL-12p40 production at all doses and in TNF-α at doses of 100 and 150 mg/kg (figure 17.). In the second study, RTA 408 (10, 30, or 100 mg/kg) was administered daily for six days, followed by LPS administration 1 h after the final dose. In this study, significant reductions in body weight were observed at the 100 mg/kg dose level starting on Day 3. Significant reductions in TNF-α were observed at the 10 mg/kg dose, and significant reductions in IL-12p40 levels, TNF α, and RANTES were observed at a dose of 30 mg/kg (Fig. 18a). Further evaluation of the rat lungs in this study revealed significant involvement of relevant Nrf2 target genes, including significant induction of NQO1 enzyme activity (by measuring the rate of 2,6-dicholorphenol-indophenol reduction) and increases in total GSH (GSH -Glo TM, Promega, Madison, WI), at 10 and 30 mg/kg (Fig. 18b). 2. Bleomycin-Induced Pulmonary Fibrosis
[00325] The effect of RTA 408 was also evaluated in models of bleomycin-induced pulmonary fibrosis in mice and rats. In the first preliminary study, RTA 408 (10, 30 or 100 mg/kg) was administered to mice daily via oral gavage for 39 days, with the bleomycin challenge (intranasal) on day 10. On the last day of dosing, lung tissue was collected and histology was performed to assess the degree of inflammation and interstitial fibrosis. In this model, statistically significant effects were not observed at the doses of RTA 408 tested (FIGS. 19a & b). Additional evaluation was performed using a mouse model of pulmonary fibrosis that has been extensively characterized at the Lovelace Respiratory Research Institute. In this study, rats were challenged with bleomycin or saline by intratracheal administration on day 0. After the challenge, the animals received RTA 408 (3, 10 or 30 mg/kg) daily via oral gavage for 28 days. Administration of the 30 mg/kg dose was interrupted on day 14, due to excessive dehydration and diarrhea in the animals. For the remaining animals, bronchoalveolar lavage fluid was collected on day 28 for evaluation of pro-inflammatory infiltrates by flow cytometry, and lung tissue was analyzed for hydroxyproline levels by LC-MS and histopathology. Bleomycin sulfate challenge induced a substantial neutrophil release and an increase in soluble collagen in the BALF, as well as an increase in hydroxyproline in the lung. Treatment with 3 and 10 mg/kg of RTA 408 significantly suppressed polymorphonuclear cell (PMN) infiltration into the lungs and also produced a significant reduction (~10% - 20%) in hydroxyproline deposition (Figs. 20a & b).
[00326] Importantly, histopathological evaluation revealed a significant decrease in collagen deposition, assessed by trichrome staining, in rats treated with RTA 408. While bleomycin control animals mainly exhibited moderate staining, animals treated with 10 mg/kg of RTA 408 had predominantly minimal to mild coloration (Table 2). Table 2: Effect of RTA 408 on collagen deposition in rat lung, assessed by trichrome staining intensity
a Values represent the intensity of staining in animals with interstitial trichrome staining in the areas of bleomycin-induced lung changes.
Further evaluation of rat lungs in this study also revealed significant involvement of the relevant Nrf2 target genes as analyzed by the Quantigene Plex 2.0 Multiplex Assay (Affymetrix, Santa Clara, CA) (FIG. 21). RTA 408 dose-dependent and significantly increased the enzymatic activity of Gst, Gsr, Txnrd and NQO1 in the lungs of rats exposed to bleomycin, demonstrating activation of Nrf2 by RTA 408 in this disease setting. NQO1 enzyme activity was assessed by measuring the rate of reduction of DCPIP. Enzyme activities of Txnrd, Gst and Gst were measured using commercially available kits from Cayman Chemical (Ann Arbor, MI). 3. Cigarette Smoke-Induced COPD in Mice
[00328] RTA 408 was also tested in a mouse model of cigarette smoke-induced DMFT. Mice received RTA 408 (3, 10 or 30 mg/kg) daily via oral gavage for two weeks and were exposed to cigarette smoke five days a week during the RTA 408 dosing period. At the end of the study, lung tissue and BALF were collected for analysis of inflammatory infiltrates and cytokines. In this experiment, administration of multiple doses of RTA 408 at doses as low as 3 mg/kg of RTA 408 resulted in significant suppression of pro-inflammatory cytokines, including KC (mouse functional homologue of human IL-8) and TNF-α as measured using LuminexTM Technology. A summary of the results of this study is shown in FIGS. 22a-e. An AIM analogue (63355) was tested in the same study for comparison. 63355 is a compound of the formula:

[00329] Further evaluation of mouse lungs in this study also revealed significant involvement of the relevant Nrf2 target genes (FIG. 23). Lung NQO1 enzyme activity, measured as the rate of DCPIP reduction, was significantly decreased by exposure to cigarette smoke; administration of RTA 408 redeemed this loss. Txnrd enzymatic activity was also induced by the 30 mg/kg dose of RTA 408. In general the enzymatic activity of Gsr was not altered, and the enzymatic activity of Gst was decreased with treatment - both of which were probably the consequence of a temporal response to these enzymes. Enzymatic activities of Txnrd, Gst and Gst were measured using commercially available kits from Cayman Chemical (Ann Arbor, MI). 4. Ovalbumin-Induced Asthma in Mice
[00330] The potential activity of RTA 408 was also evaluated in a pilot study in a mouse model of ovalbumin-induced asthma. Mice were sensitized with an IP injection of ovalbumin and aluminum hydroxide on day 0 and day 14 and challenged intranasally with ovalbumin in saline solution on days 14, 25, 26 and 27. Mice received RTA 408 (3, 10 or 30 mg/ kg) daily via oral gavage on days 1-13 and 15-27. After sensitization and challenge with ovalbumin, vehicle-treated mice had a significant increase in the total number of leukocytes compared to mice treated with positive control (dexamethasone). An increase in the number of T cells and B cells was also observed in vehicle treated rats. Treatment with RTA 408 at 30 mg/kg significantly reduced the number and percentage of B cells within the airways. RTA 408 (3 and 30 mg/kg) also significantly reduced the number of macrophages, but not the mean percentage of macrophages, detected in the airways. These observations are suggestive of potential effectiveness in this model. 5. Effects of RTA 408 on LPS-Induced Sepsis in Mice
[00331] Sepsis was induced on day 0 with an IP injection of LPS (21 mg/kg), and survival was followed until day 4. RTA 408 (10, 30 or 100 mg/kg) was administered daily via oral gavage from day -2 to day 2. In the vehicle control group, 60% of animals survived to day 4 (greater than the ~40% survival rate expected in this model). In the RTA 408 treatment groups, 80% of the animals in the 10 mg/kg dose group and 90% of the animals in the 30 mg/kg dose group survived to day 4 (FIGS. 24 c & d). For the 100 mg/kg dose group, 90% of the animals survived to day 4, with only a single death occurring on day 4. Although these RTA 408-induced effects are indicative of profound efficacy in this model, the survival rate relatively high in the vehicle control group prevented a statistically significant difference between the RTA 408 and control groups. Results obtained using the compound RTA 405 are also presented (Fig. 24a & b). RTA 405 is a compound of the formula:
6. Effects of RTA 408 against Radiation-Induced Oral Mucositis
[00332] Exposure to acute radiation directed to the buccal buccal pouch of hamsters produces effects similar to those observed in oral ulcerative mucositis in humans. These effects include moderate to severe mucositis, characterized by severe erythema and vasodilation, erosion of the superficial mucosa, and ulcer formation. A single study was conducted to assess the effects of RTA 408 in this model. On day 0, each hamster was given an acute radiation dose of 40 Gy directed to the left buccal sac. RTA 408 (10, 30 or 100 mg/kg) was administered orally twice daily from day -5 to day -1 and from day 1 to day 15. Starting on day 6 and continuing through day 28, every other day , oral mucositis was assessed using a standard 6-point scoring scale. Both the 30 and 100 mg/kg doses of RTA 408 caused a significant reduction in the duration of ulcerative mucositis (FIG. 25). Furthermore, a dose-dependent decrease in the percentage of animals with mucositis scores >3 was also observed. However, administration of RTA 408 at 30 or 100 mg/kg caused significant dose-dependent reductions in weight gain in irradiated hamsters. Due to greater than 20% weight loss, two of eight hamsters in the 100 mg/kg dose group were sacrificed on day 2. 7. Effect of RTA 408 on induction of Nrf2 Biomarkers in Vivo
[00333] As described above, a key molecular target of RTA 408 is Nrf2, a central transcriptional regulator of cellular antioxidant protection. Activation of Nrf2 induces upregulation of a battery of cytoprotective genes, including NQO1, enzymes involved in GSH synthesis [ie, catalytic glutamate-cysteine ligase and modifying subunits (Gclc and Gclm)], enzymes involved in detoxification (ie, glutathione S-transferases [GST]) and efflux transporters [ie, multiple drug resistance-associated proteins (Mrps)]. Induction of these genes results in a coordinated cellular effort to protect against oxidative insult, highlighted by increased antioxidant capacity, induction of glutathione synthesis, and the conjugation and export of potentially harmful molecules from the cell. In addition to the Nrf2 target gene expression and efficacy endpoints evaluated in various animal models described above, the ability of RTA 408 to induce Nrf2 target gene expression was also evaluated using tissues collected from healthy monkeys, rats and mice treated with RTA 408.
[00334] As part of 14-day non-BPL toxicity studies of RTA 408 in mice, rats and monkeys, tissues were collected for the purposes of measuring mRNA and enzyme activity levels of selected Nrf2 target genes. For mice and rats, liver samples were collected 4h after the last dose on day 14. For monkeys, blood (for PBMC isolation), liver, lung and brain tissue were collected 24h after the final dose on day 14. Activity enzymes for NQO1, Gst glutathione reductase (Gsr), as described above, were measured in tissue homogenates. mRNA levels were determined using Quantigene Plex 2.0 technology according to the manufacturer's protocol, which involves a hybridization-based assay using xMAP® Luminex® magnetic beads for direct quantification of mRNA targets. In addition, RTA 408 concentrations were measured in plasma and tissues by LC/MS/MS methods on a TQD mass spectrometer (Waters, Milford, MA).
[00335] RTA 408 generally increases the expression of several Nrf2 target genes in a dose-dependent manner at doses of 10, 30 and 100 mg/kg (FIG. 26, FIG. 27A, Fig. 28a & b). Transcriptional upregulation of Nrf2 target genes by RTA 408 also resulted in functional increases in the antioxidant response, manifested by a dose-dependent increase in NQO1, Gst and Gsr enzyme activity in rodent liver as well as monkey liver and lung (FIGS). 29a & b, Figures 30A & b, Figures 31a & b). Furthermore, in rodents, liver exposure of RTA 408 correlated with the level of enzyme activity of NQO1, the prototypical target gene for Nrf2 (FIG. 32b, FIG. 33b). In monkeys, the PBMC mRNA expression level of NQO1 and sulfiredoxin 1 (SRXN1) correlated with plasma exposure to RTA 408 (FIGS. 37A & b). In general, RTA 408 increased mRNA levels and activity of Nrf2 targets, and such increases generally correlated with plasma and tissue exposures, suggesting that Nrf2 targets may serve as viable biomarkers for Nrf2 activation (FIGS. 34a & b) and may be useful to assess the pharmacological activity of RTA 408 in healthy humans. D. Safety Pharmacology
A GLP compliant safety pharmacology program was completed using RTA 408. This included in vitro and in vivo (monkey) studies on the cardiovascular system as well as studies on the respiratory system and central nervous system in rats. 1. Evaluation of the Effects of RTA 408 on Cloned hERG Channels Expressed in HEK293 Cells
[00337] This study was carried out to evaluate the effects of RTA 408 on the rapid activation inward rectifying potassium current (IKr) driven by hERG (human ether-a-go-go related gene) channels stably expressed in the human embryonic kidney cell line (HEK293). The effects of RTA 408 on hERG-related potassium current were evaluated using whole-cell patch clamp electrophysiological methods. RTA 408 was determined to have an IC50 value of 12.4 µM in a QPatch_Kv11.1 hERG assay. This value was 2.5-3 times higher than the values for 63170 (4.9 μM) and 63189 (3.8 μM), respectively. The RTA 408 IC50 value was similar to the 63171 value (15.7 µM). 2. Cardiovascular Evaluation of RTA 408 in the Cynomolgus Monkey
[00338] A single study was conducted to assess the potential cardiovascular effects of RTA 408 in conscious, free-moving cynomolgus monkeys. The same four male and female cynomolgus monkeys were administered vehicle (sesame oil) and RTA 408 at doses of 10, 30 and 100 mg/kg, according to a Latin square design, with one animal/sex/treatment dosed each week followed by a 14-day washout period between administrations, until each animal has received all treatments. Vehicle and RTA 408 were administered to all animals via oral gavage at a dose volume of 5 mL/kg.
[00339] Animals were instrumented with telemetry transmitters for the measurement of body temperature, blood pressure, heart rate and evaluation of the electrocardiogram (ECG). Body temperature, systolic, diastolic and mean blood pressure, heart rate, and ECG parameters (QRS duration and QT, PR and RR intervals) were continuously monitored at least 2 h pre-dose until at least 24 h post-dose. ECG tracings were printed at designated time points from cardiovascular monitoring data and qualitatively evaluated by a certified veterinary cardiologist. Prior to the first administration in the study, untreated animals were continuously monitored for cardiovascular endpoints for at least 24h, and this data was used in calculating the corrected QT interval throughout the study.
[00340] Observations of mobility, mortality, injury and availability of food and water were performed at least twice a day for all animals. Clinical observations were conducted pre-dose, approximately 4 h post-dose and following completion of the cardiovascular monitoring period. Body weights were measured and recorded the day before each treatment administration.
[00341] RTA 408 at doses of 10, 30, and 100 mg/kg did not produce mortality, adverse clinical signs or resulted in significant changes in body weight, body temperature, blood pressure, or qualitative or quantitative ECG parameters (PR, RR, QRS, QT intervals) (FIG. 35; Table 45). In the 100 mg/kg dose group, a small (1.6% on average) but statistically significant increase in the corrected QT interval was observed; however, data from individual animals did not demonstrate consistent increases in QTc that would indicate a test article-related effect. Consequently, due to the small magnitude of change and the lack of a consistent response in individual animals, these slight increases in QTc were not considered to be related to RTA 408 treatment. Therefore, oral administration of RTA 408 had no effect on function cardiovascular disease in cynomolgus monkeys at doses up to and including 100 mg/kg. 3. Neurobehavioral Assessment of RTA 408 in rats
The potential acute neurobehavioral toxicity of RTA 408 was evaluated in rats. Three treatment groups of 10 male and 10 female CD® [Crl:CD® (SD)] rats received RTA 408 at doses of 3, 10 or 30 mg/kg. An additional group of 10 animals/sex served as a control and received the vehicle (sesame oil). Vehicle or RTA 408 was administered to all groups by oral gavage, once a day 1, with a dose volume of 10 mL/kg.
[00343] Observations of mobility, mortality, injury and availability of food and water were performed at least twice a day for all animals. Observations for clinical signs were performed before dosing on day 1 and after each observational battery functional assessment (FOB). FOB pre-dose (day -1) and approximately 4 and 24h post-dose evaluations were performed. Body weights were measured and recorded pre-dose on day 1.
[00344] RTA 408 at doses of 3, 10 and 30 mg/kg did not produce mortality, adverse clinical observations or effects on any of the neurobehavioral measures tested. Slight decreases in weight gain were observed about 24h post-dose in the 30 mg/kg group that could potentially be related to a test article. With respect to basic neurobehavioral endpoints evaluated in this study, RTA 408 did not produce any adverse effects in rats at doses up to and including 30 mg/kg. 4. Pulmonary Evaluation of RTA 408 in Rats
[00345] The potential effect of RTA 408 on lung function was evaluated in rats. Three treatment groups of eight male and eight female CD® [Crl:CD® (SD)] rats received RTA 408 at doses of 3, 10 or 30 mg/kg. An additional group of 8 animals/sex served as a control and received the vehicle (sesame oil). Vehicle or RTA 408 was administered to all groups by oral gavage, once a day 1, with a dose volume of 10 mL/kg.
[00346] Observations of mobility, mortality, injury and availability of food and water were performed at least twice a day for all animals. Clinical observations were conducted pre-dose, approximately 4 h post-dose, and following completion of the 8-h pulmonary monitoring period. Body weights were measured and recorded on the day of RTA 408 administration. Lung function (respiratory rate, tidal volume, and minute volume) was monitored at least 1h before dosing to establish a baseline and for at least 8h post-dose.
[00347] RTA 408 at doses of 3, 10 and 30 mg/kg did not produce mortality, adverse clinical observations or effects on any of the pulmonary parameters tested. Thus, with respect to the pulmonary basal endpoints evaluated in this study, RTA 408 did not produce any adverse effects in rats at doses up to and including 30 mg/kg. E. Non-Clinical Overview 5. Pharmacokinetics
[00348] RTA 408 was investigated both in vitro and in vivo to assess its PK and metabolism properties. In vitro studies were carried out to determine the binding of plasma proteins and RTA 408 and blood/plasma partitioning, inhibition and induction of cytochrome P450 (CYP450) and to identify the metabolites formed by the liver microsomes of mice, rats, monkeys and human beings. Data regarding in vivo absorption and distribution after repeated administration of RTA 408 were obtained primarily by monitoring drug levels in plasma and selected tissues from toxicology studies. Sensitive and selective bioanalytical methods (LC/MS/MS) based on liquid chromatography-mass spectrometry were used to measure concentrations of RTA 408 in plasma, blood and tissues with adequate precision and accuracy. Measurements were performed using TQD and QToF (Waters) mass spectrometers. The. Absorption
[00349] The absorption and systemic pharmacokinetic behavior of RTA 408 was studied in mice, rats and monkeys after single and repeated oral administration (daily). After oral administration of a suspension formulation at doses of 10 to 100 mg/kg, maximum concentrations were observed within 1 to 2h in mice and within 1 to 24h in rats and monkeys. Systemic exposure to RTA 408 tended to be higher in rats, with lower levels seen in rats and monkeys. Estimates of the apparent terminal half-life of RTA 408 observed after oral administration were generally in the range of 6 to 26-h, although the apparent prolonged absorption phase in some instances precluded calculation of a definitive half-life estimate.
[00350] Systemic exposure to RTA 408 was generally similar in males and females. Exposure to RTA 408 followed by repeated daily oral administration tended to be slightly higher (< 2-fold) than the exposure observed after a single dose. Administration of RTA 408 over a dose range of 3 to 100 mg/kg in a suspension formulation generally resulted in dose-proportional increases in systemic exposure. However, administration of higher doses (100 to 800 mg/kg in monkeys; 500 to 2000 mg/kg in rats) did not result in similar increases in exposure, suggesting saturation of absorption at doses greater than 100 mg/kg. Following oral administration of a non-optimized (loose-filled) capsule formulation of RTA 408 (3 mg/kg) to monkeys, dose-normalized systemic exposure tended to be somewhat lower than that observed with a suspension formulation.
[00351] The absorption and systemic pharmacokinetic behavior of RTA 408 was studied in rats using single and repeated topical administration. Administration of RTA 408 on a 0.01% to 3% scale showed lower plasma concentrations compared to oral dosing. Systemic exposure to RTA 408 generally increased in a dose-dependent manner. Topical administration was formulated as a suspension in sesame oil.
[00352] Using rabbits, the ocular absorption and systemic pharmacokinetic behavior of RTA 408 were evaluated. RTA 408 was administered topically into the eye once daily for five days. Ocular administration showed low plasma concentration of RTA 408 relative to when RTA 408 is administered orally (FIG. 36). The amount of RTA 408 in plasma even after five consecutive days showed only a small change compared to the concentration after the first dose compared to when RTA 408 was administered orally, where plasma concentrations were almost 100-fold superiors (Fig. 36). B. Distribution
Plasma protein binding of RTA 408 was evaluated in mouse, rat, rabbit, dog, mini pig, monkey and human plasma at RTA 408 concentrations of 10-2000 ng/ml using ultracentrifugation methodology. RTA 408 has been extensively bound to plasma proteins. Plasma protein binding in the appropriate species ranged from 93% (mouse) to >99% (mini pig), with 95% binding in toxicology species (rat and monkey) and 97% in humans. There was no evidence of protein concentration-dependent binding in any species tested. Results from blood-to-plasma partitioning experiments indicate that RTA 408 tended to distribute primarily in the blood plasma fraction in a linear fashion, with blood:plasma ratios <1.0 for all species and all concentrations tested.
The tissue distribution of RTA 408 was investigated after oral administration in mice, rats and monkeys. In the 14-day non-GLP toxicity studies, selected tissues (liver, lung, and brain) were collected at a single time point (4h for rat and mouse; 24h for monkey) after the last dose of the study was administered and were analyzed for RTA 408 content using LC/MS/MS. RTA 408 is easily distributed in the lung, liver and brain. In the lung, 4h RTA 408 concentrations in mice and rats were similar or slightly higher (< two-fold) than plasma concentrations, whereas at 24h in monkeys, lung RTA 408 concentrations were 6- to 16-fold higher than plasma concentrations. A similar pattern was observed for the brain. In contrast, RTA 408 concentrations in liver were 5- to 17-fold higher than plasma for mice and rats at 4h, and 2-to-5-fold higher than plasma at 24h in monkeys.
[00355] The pharmacodynamic effects of RTA 408 on tissues were evaluated in mice, rats and monkeys by monitoring the induction of Nrf2 target genes in the same tissues collected for drug exposure from the 14-day toxicity studies. Induction of Nrf2 target genes by RTA 408 resulted in increases in the antioxidant response as manifested by dose-dependent increases in NQO1 enzymatic activity, glutathione S-transferase (Gst) and glutathione reductase (Gsr) in the tissues examined. Enzyme activities were measured as described above. Furthermore, in rodents, liver content of RTA 408 correlated with the level of enzyme activity of NQO1, the prototypical target gene for Nrf2. In monkeys, the level of mRNA expression in peripheral blood mononuclear cells (PBMC) for NQO1 and sulfiredoxin 1 (SRXN1) correlated with plasma exposure of RTA 408 (FIGS. 37A & b). In general, RTA 408 induced Nrf2 biomarkers in rodents and monkeys, and such inductions generally correlated with tissue and plasma exposure to RTA 408.
The highest concentrations of the compound when RTA 408 was administered to rabbits via topical ocular administration were found in the cornea, retina or iris while the vitreous humor, aqueous humor and plasma showed significantly lower concentrations of RTA 408 (FIG. .38). ç. Metabolism
[00357] The metabolism of RTA 408 was investigated after in vitro incubation of RTA 408 for 60 min with liver microsomes from mice, rats, monkeys and humans in the presence of a nicotinamide adenine dinucleotide phosphate (NADPH)-regenerating system and a uridine diphosphate glucuronosyltransferase (UGT) reaction mixture. Extensive turnover of RTA 408 was observed with primate microsomes, with <10% of the parent molecule remaining at the end of the 60 min incubation in monkey and human microsomes. In contrast, the extent of metabolism was smaller in rodent microsomes, with >65% of the parent molecule remaining at the end of the incubation. The lack of available authentic standards for the various potential metabolites of RTA 408 precluded quantitative assessment of the observed metabolites. From a qualitative perspective, a similar pattern of RTA 408 metabolites was observed across the species and included peaks with masses consistent with the reduction and hydroxylation of RTA 408, as well as the glucuronidation of RTA 408 or its reduction/hydroxylation metabolites . No single human metabolite was observed, with all peaks in human microsome incubations also being observed in one or more of the preclinical species. In particular, based on in vitro microsome data, all human metabolites were present in rat or monkey, selected rodent and non-rodent toxicity species. d. Pharmacokinetic Drug Interactions
The potential of RTA 408 to inhibit cytochrome P450 (CYP450)-mediated metabolism was evaluated using pooled human liver microsomes and standard substrates for specific CYP450 enzymes. RTA 408 directly inhibited CYP2C8 and CYP3A4/5 with Ki values of approximately 0.5 μM for each enzyme. No significant inhibition was observed for the other enzymes tested (CYP1A2, CYP2B6, CYP2C9, CYP2C19 or CYP2D6), with <50% inhibition at the highest concentration tested (3 μM). Furthermore, there was little or no evidence of metabolism-dependent inhibition of any of the enzymes tested. Future studies investigating the potential for CYP3A4/5-mediated drug-drug interactions are warranted based on these data and the potentially high concentrations that can be reached locally in the gastrointestinal (GI) tract after oral administration.
The potential of RTA 408 to induce CYP450 enzyme expression was evaluated using cultured human hepatocytes. Under conditions where prototypic inducers caused the expected increases in CYP activity, RTA 408 (up to 3 µM) was not an inducer of CYP1A2, CYP2B6, or CYP3A4 enzyme activity in cultured human hepatocytes. Enzyme activity was measured by monitoring the substrate conversion of phenacetin, bupropion and testosterone to CYP1A2, CYP2B6 and CYP3A4, respectively, in isolated microsomes. F. Effects of RTA 408 on Acute Radiation Dermatitis
[00360] The effects of RTA 408 as a topical or oral preventive for acute radiation dermatitis were examined. Using male BALB/c mice, a dose of 30 Gy of radiation was administered on day 0 (Table 3). Sesame oil vehicle or RTA 408 was administered to rats on day -5 to -1 and days 1 to 30. RTA 408 was administered either orally at 3, 10 and 30 mg/kg in sesame oil and topically in sesame oil. percentage composition of 0.01%, 0.1% and 1% sesame oil. Dermatitis was blindly assessed every day from day 4 to day 30. On day 12, the typical peak of dermatitis was observed and 4 mice were sacrificed 4 hours after dose administration. The remaining mice were sacrificed on day 30, 4h post-dose. Plasma was collected on days 12 and 30, as well as skin samples irradiated for mRNA and histological examination. Table 3: Study Design for Acute Radiation Dermatitis Model

[00361] In groups of this where rats were treated with RTA 408, the incidence of dermatitis appeared to be slightly decreased in severity when RTA 408 was given in oral or topical administration (FIGS. 39-42). Furthermore, curves plotting the mean clinical dermatitis score for the test groups as a function of time show some change with administration of RTA 408 in oral or topical form from the untreated test groups (FIGS. 43-45) particularly in the case where RTA 408 was given via an oral administration. Furthermore, as can be seen in Tables 4 and 5 below, the percentage of mice suffering from dermatitis with a clinical score above 3 was significantly lower for mice treated with RTA 408 via an oral administration, while the percentage of rats suffering from dermatitis with a clinical score above 2 was slightly lower for test groups that received a topical administration of RTA 408. Table 4: Percentage of mice per test group that scored above 2 on their clinical dermatitis exam and received a topical treatment containing RTA 408 (Part 1)

G. Effects of RTA 408 on Fractional Radiation Dermatitis
[00362] Using RTA 408 via topical administration, the effects of RTA 408 towards attenuating the effects of fractional radiation dermatitis were measured. Using Balb/c mice, RTA 408 in a topical preparation was administered to rats daily from day -5 to day 30 in three doses from 0.01% to 1%. Mice were irradiated on days 0-2 and 5-7 with six doses of 10-Gy per day. Clinical dermatitis scores for mice were assessed blindly every other day from day 4 until the end of the study. In Figure 46, the graph shows the change in mean clinical score for each group plotted as a function of time. The graph shows a statistically significant improvement in scores for mice treated with topical formulations from 0.1 to 1% of RTA 408. Study and treatment parameters can be found in Table 6. Table 6: Study Conditions for Radiation-Induced Dermatitis fractioned

[00363] Analyzing the average cnic scores that were shown in FIG. 46, an area under the curve (AUC) analysis was performed, which yielded the severity of dermatitis in relation to how long the dermatitis persisted. This AUC analysis allowed direct comparison between the different groups of mice and the effect of different percentage compositions of RTA 408 (FIG. 47 and Table 7). Administration of topical formulations of RTA 408 reduced grade 2 and grade 3 lesions by 60% and 33% when mice were exposed to vehicle alone to 21% and 6% with RTA 408 at 1% concentration, respectively. The other RTA composition showed some activity but was not as significant as manifested by the 1% formulation. Table 7: Dermatitis Score Percentage for Each Treatment Group
H. Synergistic Effects of RTA 408 and Cancer Therapeutic Agents on Tumor Growth
[00364] A study of the effects of RTA 408 used in combination with traditional chemotherapeutic agents was performed to determine the effectiveness of potential treatment. In vitro studies were performed to determine the effects of RTA 408 on two different prostate cancer cell lines, LNCaP and DU-145. As can be seen in Fig. 48a, treatment of prostate cancer cell lines (LNCaP) in vitro with 5-fluorouracil shows a statistically significant increase in cytotoxicity when combined with RTA 408 at doses ranging from 0.125 to 0.5 μM . Using the DU-145 prostate cell line and docetaxel, RTA 408 amplified the cytotoxicity of the chemotherapeutic agent statistically significantly for RTA 408 dosage from 0.125 to 0.75 μM, as shown in Fig. 48b. This evidence supports the concept that RTA 408 could act synergistically with cancer therapeutic agents and can be used in some modalities to provide greater efficacy in the treatment of cancer patients.
[00365] Following the successful results of the in vitro assay, a pilot in vivo assay was performed using LNCaP/C4-2B and DU145 human prostate cancer designed to express firefly luciferase (hereinafter referred to as C4-2B-Luc and DU145-Luc, respectively). It should be noted that both of these cell lines grow in an androgen-independent manner. Cells were cultured in RPMI 1640 supplemented with 10% FBS. Cells were harvested using TrypLE Express (Invitrogen) and washed with PBS and counted. Cells were reconstituted in PBS to a final concentration of 3 x 106 cells per 30 µL (unless otherwise indicated) and aliquoted into separate tubes. Matrigel (BD Bioscience) of reduced growth factor was thawed overnight at +4°C and transferred to tubes in 30μL aliquots. Cell/Matrigel solutions were transferred to the vivarium and mixed just before injection in a 1:1 ratio. Each mouse (n = 1 per group, for a total of three animals) received a single subcutaneous injection of tumor cells. Tumors were previously established for 4 weeks. Then, one animal was treated with RTA 408 (17.5 mg/kg, i.p.) once a day for 3 days (days -3 to -1). The following day (day 0), the animals treated with RTA 408 and one other animal were treated with a single dose of 18 Gy IR, located in the pelvic region, where the tumors were implanted. The mouse that was previously treated with RTA 408 received three additional doses of RTA 408 (17.5 mg/kg, i.p.) once every other day for the following week. The third animal received no treatment and served as a positive control. Tumor progression was monitored weekly via live imaging. To detect tumor cells expressing luciferase, mice were injected IP with D-Luciferin 5 min before imaging according to the manufacturer's protocol (Caliper LifeScience). Prior to imaging, rats were anesthetized by isoflurane inhalation and imaged in the IVIS Lumina XR system (Caliper LifeScience). For standardization, the minimum exposure time required to image a control tumor was determined and all animals were imaged under these conditions. On day 7, no apparent reduction in tumor size was visible in the IR-treated animal compared to the control, whereas the animal receiving both RTA 408 and IR showed a smaller tumor image. On day 14 and day 21, the control animal showed continuous tumor development and growth, while the animal treated with ionizing radiation showed some improvement, most notably on day 21. On the other hand, animals treated with RTA 408 and ionizing radiation did not show no progression from day 7 to day 14 and they had no visible tumor by day 21. The tumor progress per week can be seen in FIG. 49. Both in vitro and in vivo data show that RTA 408 appears to complement the activity of different cancer therapeutic agents, thus increasing the agent's efficacy. I. Effects of RTA 408 in an Ocular Inflammation Model
[00366] A study of the effects of RTA 408 on ocular inflammation was carried out using rabbits of the New Zealand strain albino. The rabbits were divided into 5 groups of 12 rabbits which received three different concentrations of RTA 408 (0.01%, 0.1% and 1%), Voltarene® collyre in 0.1% and the vehicle (sesame oil). Each rabbit was given three instillations within 60 minutes before induction of paracentesis and two instillations within 30 minutes after induction of paracentesis. Each instillation was 50 µL and given to both eyes. Aqueous humor for 6 animals per time point was collected 30 min and again 2 h after paracentesis induction. The amount of inflammation was determined by the concentration of proteins in the aqueous humor. As shown in FIG. 50, RTA 408 showed a reduction in aqueous humor protein similar to that of the highest concentration of any of the other reference compounds (MaxiDex or mapracorat) at only 0.01% of RTA 408 in the formulation. The effects of increasing RTA 408 concentration appeared to be insignificant as all RTA 408 concentrations appeared to show relatively similar effects within error in reducing aqueous humor protein concentration. J. Polymorph Screening
[00367] A preformulation and polymorphism study was performed for compound 63415. As part of this study, a preliminary polymorphism program was performed with the aim of identifying the most stable anhydrous form at room temperature and possible hydrates with a reasonably high probability. A total of 30 crystallization experiments, including phase equilibrium, drying experiments and other techniques, were performed. All solids obtained were characterized by FT-Raman spectroscopy. All new forms were characterized by PXRD and TG-FTIR and optionally by DSC and DVS.
[00368] Furthermore, the amorphous form was prepared and characterized. Several experiments were carried out using different techniques and approaches to prepare the amorphous form. The amorphous form was characterized by Karl-Fischer titration, DSC, TG-FTIR, DVS, PXRD and FT-Raman spectroscopy. The stability of the amorphous form was tested under conditions of high temperature and humidity over four weeks. B. Raw Material and Nomenclature
[00369] Two batches of 63415 were used as raw materials (table 8). 63415 is also referred to as PP415 in this description. All samples received or generated during this project were given a unique identification code of the form PP415-Px, where Px refers to the sample/experiment number (x = 1, 2,..., n). Table 8: Raw Materials
7. Compound 63415, lot # 0414-66-1 (PP415-P1): The Amorphous Form
[00370] Raw material 63415, batch # 0414-66-1, was characterized by FT-Raman spectroscopy, PXRD, TG-FTIR, Karl-Fischer titration, 1H-NMR, DSC, DVS and approximate solubility measurements . The results are summarized in table 9. Table 9: Characterization of raw material 63415 (PP415-P1)

[00371] FT-Raman spectrum (FIG. 58) will be used as the reference spectrum for the raw material. PXRD (FIG. 59) does not show the sharp peak pattern. The wide halo of ~10-20 °2θ is characteristic for amorphous materials.
The TG-FTIR thermogram (FIG. 60) shows the gradual loss of ~0.9 wt-% EtOH (ie ~0.1 eq.) with traces of H2O between 25 and 200°C. Decomposition starts at T > 290°C.
[00373] A water content of 0.5 wt-% was determined by Karl Fischer titration.
[00374] O1H-NMR spectrum (FIG. 61) is in agreement with the structure and shows ~0.08 eq. EtOH, according to the thermogram of TG-FTIR.
[00375] The DSC thermogram (FIG. 62) shows in a first heating scan a glass transition of the amorphous material at Tg = 152.7°C (ΔCp = 0.72 J/g°C). In a second scan after cooling, the glass transition occurs at Tg = 149.7°C (ΔCp = 0.45 J/g°C).
[00376] The DVS isotherm (FIG. 63) shows that a gradual mass loss of 1.0 wt-% occurred after the relative humidity of 50% r.u. down to 0% r.u.; equilibrium was achieved at 0% r.u. After increasing the relative humidity to 95% r.u. a gradual mass gain of 2.1 weight-% (relative to mass at 0% r.u.) occurred; equilibrium was achieved at 95% r.u. Lowering the relative humidity to 95% r.u. to 50% u.r. the final mass was 0.2 wt-% below the initial mass. The increase in mass of 0.4 wt-% by 85% r.u. (in relation to the initial mass) classifies the sample as slightly hygroscopic.
[00377] The FT-Raman spectrum (FIG. 64) and the PXRD standard (FIG. 65) of the sample after the DVS measurement are unchanged compared to the spectrum and the standard of the sample before the measurement.
[00378] The approximate solubility of the PP415-P1 raw materials was measured in twelve solvents and four solvent mixtures in rt by manual dilution combined with visual observation (table 10). Due to the experimental error inherent in this method, solubility values are intended to be considered as rough estimates and should be used exclusively for the design of crystallization experiments. All solvent mixtures are listed as ratios by volume (v/v). Table 10: Approximate solubility of raw material PP415-P1 (amorphous)
8. Compound 63415, lot # 2083-69-DC (PP415-P40): Class 2
[00379] 63415, lot # 2083-69-DC, is a solvate of heptane. This material (PP415-P40) was characterized by PXRD and found to correspond to class 2 (FIG. 66).
[00380] Class 2 probably corresponds to isostructural, non-stoichiometric (< 0.5 eq.) solvates (of heptane, cyclohexane, isopropyl ether, 1-butanol, triethyl amine and possibly other solvents such as hexane and other ethers ) with tightly bound solvent.
The small peaks visible in the PP415-P40 pattern at 7.9°2θ and 13.8°2θ do not correspond to the peaks of classes 3, 4 or 5. Their origin is not clear at this point. 9. Chemical Stability of the Amorphous Form
[00382] The chemical stability of the amorphous form was investigated in different solvents over seven days.
Solutions/suspensions with a concentration of 1 mg/mL were prepared in four organic solvents (acetone, MeOH, MeCN, EtOAc) and three aqueous surfactant media (1% aq. SDS, 1% aq. Tween 80, 1 % aq. CTAB).
[00384] Four separate solutions/suspensions were elaborated for each solvent, equilibrated for 6 h, 24 h, d 2 and d 7 and later analyzed by HPLC.
[00385] Relative area-% obtained from the HPLC chromatograms are given in table 11. The compound appears to be somewhat unstable in the diluent (0.1% formic acid in MeCN); along the sequence (ie within ~24 hours) the area-% of a reference sample (PP415-P1, run at the beginning and end of the sequence) decreased from 99.9% to 99.3% at 254 nm and from 99.9% to 99.5% at 242 nm. Due to this effect, samples measured at the end of the sequence (set up in the following order: 7d, 2d, 24h, 6h) may be affected and the area-% obtained may be underestimated. Table 11: Chemical stability experiments with the amorphous form of 63415 (PP415-P1)a
a At the third wavelength (210 nm), the signal intensity was weak and the signal-to-noise ratio large, thus integration was not performed b suspensions, not all material dissolved for all time points c suspensions, not all dissolved solids for 24 h and 6 h time points
Decomposition >1% was observed for solutions in MeCN after seven days and for suspensions in aqueous media of 1% Tween 80 (at all time points at 254 nm and after 24h, 2d and 7d at 242 nm). 10. Amorphous Form Storage Stability
[00387] To learn more about its fundamental properties and physical stability, the amorphous form of 63415 was highlighted by storage at high temperatures and high relative humidity.
[00388] Samples of the amorphous form (the raw material PP415-P1) were stored open at 25°C/~62% ur (over saturated aqueous solution of NH4NO3) and 40°C/~75% ur (over saturated aqueous solution NaCl) and closed at 60°C and 80°C (Table 12). At the 0 s, 1 s, 2 s and 4 s time points the samples were examined by PXRD and compared to the raw material, PP415-P1. Table 12. Storage stability experiments with the amorphous form of 63415 (PP415-P1)

After one week (time point 1 s, FIG. 67), two weeks (time point 2 s, Fig. 68) and four weeks (time point 4 s, FIG. 69) all four samples they were still amorphous, as X-ray powder diffractograms show no difference compared to raw materials at the 0 s time point. 11. Crystallization and Drying Experiments a. Crystallization Experiments
[00390] Phase equilibriums, hot solution crystallizations and evaporation experiments were carried out from the amorphous form to identify with reasonably high probability the most stable anhydrous form in r.t. and possible hydrates. All materials obtained were characterized by FT-Raman spectroscopy; selected samples were also characterized by PXRD.
[00391] FT-Raman spectra were grouped into classes according to the similarity of their peak positions. The original sample (PP415-P1, see table 8) was classified together with the crystallization products. Spectra within a class, however, are not strictly identical, but similar. There may be small differences and peak shifts. Considering the FT-Raman spectra alone, it is difficult to determine whether the spectra of a class belong to the same polymorphic form.
The peaks of the PXRD patterns were determined and the patterns were then classified into clusters using the PANalytical X'Pert software (Highscore Plus). These clusters identify patterns of high similarity. However, small but significant differences exist within a cluster. Thus, patterns within a cluster do not necessarily correspond to the same polymorphs, but represent different shapes with very similar molecular structures. The FT-Raman classes correspond in all cases to the PXRD clusters. B. Suspension Equilibrium Experiments
[00393] Suspension equilibrium experiments were carried out in one solvent and eleven solvent mixtures (Table 13). Suspensions of ~100 mg PP415-P1 in 0.2-2.0 mL of selected solvents were made and stirred for 4-15 days at 22-24°C. Solids were recovered and characterized by FT-Raman spectroscopy; most were also characterized by PXRD. Table 13. Suspension equilibrium experiments from the amorphous form (PP415-P1)

water activities: aa(H2O) ~ 0.5 to 50°C; ba(H2O) ~ 0.85 at 50°C; ca(H2O) > 0.99 at 64°C; of spectrum contains solvent signals c. Hot Solution Crystallizations
[00394] Hot solutions of PP415-P1 were prepared in one solvent and four solvent mixtures (table 14). After slow cooling at 5°C at a rate of ~0.2 K/min, precipitation was observed in three cases (-P20, -P21, -P24). In two cases (-P22, -P23) no solids precipitated, even after storage at 4-5°C for two days. Here, the solvent was evaporated under N2 flow at rt. The solids were recovered and characterized by FT-Raman spectroscopy and for those with different spectra of the amorphous raw material, FT-Raman class 1, also by PXRD. Table 14: Slow cooling experiments from the amorphous form (PP415-P1)
to no precipitation after slow cooling and stirring at 5°C for 2 days; solvent evaporated under N2 flow in rt b spectrum contains solvent signals d. Evaporation/Precipitation Experiments
[00395] Clear solutions of PP415-P1 were prepared in three solvent mixtures (table 15). Solvents were then slowly evaporated at rt under ambient conditions. However, in two of the three experiments (-P15 and -P17) white solid precipitated before evaporation started. The solids obtained were analyzed by FT-Raman and PXRD spectroscopy. Table 15. Slow evaporation experiments with the amorphous form (PP415-P1)
the spectrum contains solvent signals e. Drying Experiments
[00396] At least one sample of each class was vacuum dried in order to dissolve the solvates and to obtain unsolvated crystalline forms of 63415 (Table 16). The dry materials were characterized by FT-Raman, PXRD and TG-FTIR. Table 16. Drying experiments performed on samples obtained from crystallization experiments

successful dissolution, significant reduction in solvent content, mostly amorphous sample; only few, broad peaks in PXRD b less crystalline sample, as indicated by broader peaks in PXRD c successful dissolution, significant reduction in solvent content, sample still crystalline; no change in structure 12. Characterization of New Forms (Classes) a. New Classes Summary
[00397] In addition to the amorphous form of 63415, four new crystalline forms were obtained in this study (table 17). Table 17: Summary of obtained classes

[00398] Class 2: Most crystallization experiments resulted in solid class 2 material. These samples probably correspond to isostructural, non-stoichiometric (< 0.5 eq.) solvates (of heptane, cyclohexane, isopropyl ether, 1-butanol, triethylamine and possibly hexane and other ethers, etc.) with tightly bound solvent molecules. The Raman spectra and PXRD standards within this class are very similar to each other, so the structures can be essentially identical with small differences due to the different solvents that have been incorporated.
[00399] Drying experiments on class 2 samples did not result in a crystalline, unsolvated form. Even high temperatures (80°C) and a high vacuum (<1x10-3mbar) did not remove the tightly bound solvent molecules completely; a solvent content of >2 wt-% was always maintained. The crystallinity of these samples is reduced, but neither transformation to a different structure nor substantial amorphization was observed.
[00400] Class 3: Class 3 solid material was obtained from several crystallization experiments. Class 3 samples are likely isostructural solvates of 2PrOH, EtOH, and likely acetone with tightly bound solvent molecules. They could correspond to stoichiometric hemisolvates or non-stoichiometric solvates with a solvent content of ~0.5 eq. As with class 2, the Raman spectra and PXRD patterns within this class are very similar to each other, indicating similar structures that incorporate different solvents.
[00401] Similar to class 2, drying experiments were also unsuccessful. Only partially tightly bound solvent molecules could be removed (~5.4 wt-% to ~4.8 wt-%, up to 3 d at 1xio-3 mbar and 80°C). The PXRD pattern remained unchanged.
[00402] Class 4: Class 4 material was obtained only from a solvent system7:3 MeCN/H2O It probably corresponds to a crystalline acetonitrile hemisolvate.
[00403] By drying (under vacuum or N2 flow at elevated temperatures), most of the solvent could be removed without altering or destroying the crystal structure (PXRD remained unchanged). Thus, an unsolvated, crystalline form (or rather, dissolved solvate) was obtained. It is slightly hygroscopic (mass gain ~0.7 wt%-% from 50% r.u. to 85% r.u.) and has a possible melting point of 196.1°C (ΔH = 29.31 J/g).
[00404] Class 5: Class 5 also obtained only one solvent system (~1:1 THF/H2O) and contains bound THF (and perhaps H2O). As the content of the two components cannot be quantified separately, the exact nature of this crystalline solvate cannot be determined.
[00405] Class 5 drying resulted in complete dissolution and transformation to the amorphous form (class 1). A possible process to prepare the amorphous form of class 2 material is a transformation from class 2 to class 5, followed by drying and amorphization. B. Class 1 - The Amorphous Form
[00406] Class 1, the amorphous form of 63415, was obtained from some crystallization experiments (table 18). Most crystallization experiments resulted in class 2, 3, 4 or 5 crystalline material.
[00407] The raw material, PP415-P1, is amorphous and belongs to class 1. Additional experiments were carried out, exclusively intended to prepare the amorphous form (class 1). Table 18. Crystallization experiments resulting in class 1 solid material
to no precipitation after slow cooling and stirring at 5°C for 2 days; solvent evaporated under N2 flow in rtc Class 2 - Isostructural Solvates (eg heptane)
[00408] Most crystallization experiments resulted in solid class 2 material (Table 19). In addition, a batch of a class 2 heptane solvent, PP415-P40, was used as a raw material (see table 8).
[00409] Class 2 FT-Raman spectra are clearly similar to each other (FIG. 70) but show small differences. They differ significantly from the spectrum of the amorphous raw material, class 1 (FIG. 71) and from the spectra of classes 3, 4, and 5 (FIG. 72).
[00410] Class 2 PXRD standards (FIG. 73) confirm the crystallinity of the materials. The patterns of the samples are very similar to each other, but show small differences (FIG. 74). Class 2 patterns clearly differ from patterns in classes 3, 4, and 5 (Fig. 75).
[00411] TG-FTIR thermogram of sample PP415-P7 (FIG. 76) shows loss of ~7.5 wt-% EtOAc and heptane in two steps from ~100°C to 290°C and decomposition at temperatures T >290 °C. Prior to the TG-FTIR experiments, samples were dried briefly (for ~5 min) under vacuum (10-20 mbar) to remove excess, unbound solvent. The loss of both EtOAc and heptane occurs together in the same temperature range; the two solvents appear to be tightly bound within the structure. The theoretical EtOAc content (bp = 76°C) of a hemisolvate is 7.4 wt-%, the theoretical heptane content (bp = 98°C) of a hemisolvate is 8.3 wt-%. Unfortunately, the content of the two components cannot be quantified separately.
[00412] TG-FTIR thermogram of sample PP415-P21 (FIG. 77) shows the loss of ~5.8 wt-% cyclohexane in two steps of ~ 140°C ~ 250°C and decomposition at temperatures T > 250°C. With the boiling point of cyclohexane at 81°C, the solvent appears tightly bound within the structure. The theoretical cyclohexane content of a hemisolvate is 7.1 wt-%. Thus, sample PP415-P18 possibly corresponds to a non-stoichiometric cyclohexane solvate (with solvent content <0.5 eq.).
[00413] TG-FTIR thermogram of sample PP415-P24 (FIG. 78) shows ~16.6 wt-% loss of 1BuOH in one step of ~50 C to 160°C, plus loss of 1BuOH (6.6 in weight-%) in a second step at 160°C to 230°C and decomposition at temperatures T > 230°C. With the boiling point of 1BuOH 117°C, the solvent from at least the second stage appears tightly bound within the structure. The theoretical 1BuOH content of a hemisolvate is 6.3 wt-%.
[00414] TG-FTIR thermogram of sample PP415-P29 (FIG. 79) shows loss of ~5.1 wt-% EtOAc and TEA from ~50°C ~220°C, mostly in a 180° step C to 210°C. Decomposition takes place at temperatures T > 220°C. The loss of both EtOAc and TEA occurs together in the same temperature range; the two solvents appear to bond tightly within the structure (boiling point of EtOAc at 77°C and TEA at 89°C).
[00415] TG-FTIR thermogram of sample PP415-P47 (FIG. 80) shows typical class 2 two-step mass loss (total ~7.9 wt-% EtOAc) at temperatures up to 240°C, indicating very tightly bound solvent molecules.
[00416] TG-FTIR thermogram of sample PP415-P48 (FIG. 81) shows mass loss of ~3.5 wt-% ethyl formate and water, first gradually and then in one clear step between 180°C and 200°C. There may be further loss of ethyl formate concomitant with decomposition at T > 240°C.
[00417] Thus, class 2 samples can all correspond to non-stoichiometric (< 0.5 eq.), isostructural solvates with tightly bound solvent molecules. As the Raman spectra and PXRD patterns within this class are very similar to each other, the structures can be essentially identical to each other with only small distortions of cell unit dimensions or small changes in atomic positions within the unit cell due to different sizes and forms of the incorporated solvent molecules. Table 19: Crystallization experiments resulting in class 2 solid material

the raw material: PP415-P40, class 2; in all other experiments PP415-P1, class 1, was used as raw material. d. Drying Experiments on Class 2 Samples
[00418] Several class 2 samples were dried under vacuum (and some at elevated temperatures) and in an attempt to dissolve them in order to obtain an anhydrous form of 63415. Details and characterization of the dried samples are provided below in table 20.
[00419] However, even drying for three days at 80°C and a vacuum < 1x10-3 mbar could remove the tightly bound solvent molecules completely; a solvent content of >wt-%2 remained (see samples -P32 and -P34). The PXRD standards show a reduced crystallinity of these samples, but no transformation to a different structure was observed. Table 20. Drying Experiments on Class 2 Samples

a according to PXRD, a little less crystalline
[00420] Thus, class 2 solvates appear to have very tightly bound solvent molecules. They are difficult to dissolve or transform/amorphize. and. PP415-P7 ^ PP415-P30
[00421] The solid material of sample PP415-P7, class 2, obtained from a suspension equilibrium experiment in 1:2 EtO-Ac/heptane was dried (as PP415-P30) under vacuum for several days (1- 10 mbar, 50-70°C).
[00422] FT-Raman spectrum of dry class 2 material (PP415-P30) shows small differences from the original spectrum (sample PP415-P7, FIG. 82) but still corresponds to class 2.
[00423] The PXRD pattern of dry class 2 material (PP415-P30) shows slightly wider, less intense peaks (FIG. 83) but still corresponds to class 2.
[00424] TG-FTIR thermogram of the dry sample of PP415-P30 (FIG. 84) shows the loss of ~2.5 wt-% heptane (and some EtOAc) in two steps of ~50°C ~ 250°C and decomposition at temperatures T > 250°C. Compared to the TG-FTIR of the PP415-P7 sample (FIG. 76), the two solvent loss steps are preserved, but the total amount of solvent in the sample decreased from ~7.5 wt-% in PP415-P7 to ~ 2.5 wt-% in PP415-P30.
Thus, the attempt to dissolve this solvent at elevated temperatures (50-70°C) and a vacuum of 1-10 mbar) caused only a partial loss of solvent. f. PP415-P15 ^ PP415-P18
[00426] The solid sample material PP415-P15, class 2, obtained from a precipitation experiment in 1:2 DCM/IPE was dry (as PP415-P18) under vacuum (~ 2-20 mbar) at r.t. for ~2 h.
[00427] The FT-Raman spectrum of PP415-P18 is identical to the spectrum of sample PP415-P15 (FIG. 85), both correspond to class 2.
[00428] PXRD pattern of PP415-P18 shows small differences to the pattern of PP415-P15 (FIG. 86). PP415-P18 still corresponds to class 2.
[00429] The thermogram of TG-FTIR (FIG. 87) shows the loss of ~7.0 wt-% IPE in two steps from ~140°C to ~250°C and decomposition at temperatures T >250°C. With the boiling point of IPE at 67°C, the solvent appears tightly bound within the structure. The theoretical IPE content of a hemisolvate is 8.4 wt-%.
[00430] Unfortunately, no TG-FTIR was noted of the material prior to the drying step. However, as the solvent appears so tightly bound in structure and no (or only small) changes are observed in the FT-Raman spectra and PXRD standards, it is assumed that drying had no significant effect on the structure or solvent content. . g. PP415-P17 ^ PP415-P19 ^ PP415-P32
The solid sample material PP415-P17, class 2, obtained from a precipitation experiment in 1:3 EtOAc/heptane was dried (as PP415-P19) under vacuum (~2-20 mbar) at r.t. for ~2 h.
The FT-Raman spectrum of PP415-P19 is identical to the spectrum of sample PP415-P17 (FIG. 88); no changes can be observed, and both correspond to class 2.
[00433] PXRD pattern of PP415-P19 is slightly different from pattern of PP415-P17 (FIG. 89) but still corresponds to class 2.
The TG-FTIR thermogram (FIG. 90) shows the loss of ~7.6 wt-% heptane in two steps of ~140°C to ~270°C and decomposition at temperatures T >270°C. With the boiling point of heptane at 98°C, the solvent appears tightly bound within the structure. The theoretical heptane content of a hemisolvate is 8.3 wt-%.
[00435] An additional drying experiment (80°C, <1xio-3 mbar, 3 days) was performed on the same sample as PP415-P32.
[00436] FT-Raman spectrum remained unchanged (FIG. 88). The PXRD pattern still corresponded to class 2 (FIG. 89), but the sample was less crystalline (as the peaks were broader and had a low S/N ratio).
[00437] The TG-FTIR thermogram (FIG. 90) shows the loss of ~2.2 wt-% heptane, mostly in a step of 170°C to 200°C and decomposition at temperatures T > 250°C.
[00438] Thus, the heptane content was reduced only from 7.6 wt-% to 2.2 wt-%, confirming the tight binding of the solvent molecules. H. PP415-P21 ^ PP415-P28 ^ PP415-P34
[00439] Sample solid material PP415-P21, class 2, obtained from a slow cooling experiment in ~1:5 EtOH/cyclohexane was dried (as PP415-P28) under vacuum for several days (2-20 mbar , rt at 60°C).
[00440] FT-Raman spectrum of dry class 2 material (PP415-P28) shows small differences from class 2 spectrum (sample PP415-P21, FIG. 92) but still corresponds to class 2.
[00441] The PXRD pattern of the class 2 dry material (PP415-P28) shows slightly broader, less intense peaks compared to the PP415-P21 pattern (FIG. 93), indicating that the dry sample is less crystalline. However, the pattern still corresponds to class 2.
[00442] TG-FTIR thermogram of the dry sample of PP415-P28 (FIG. 94) shows the loss of ~3.0 wt-% cyclohexane in two steps of ~140°C ~ 250°C and decomposition at temperatures T > 250°C. Compared to the TG-FTIR of the PP415-P21 sample (FIG. 77), the two solvent loss steps are preserved, but the total amount of solvent in the sample decreased from ~5.8 wt-% in PP415-P21 to ~ 3.0 wt-% in PP415-P28.
[00443] Thus, the dissolution of this solvate appears to have caused only a partial loss of solvent, parallel to a partial loss of crystallinity.
Further drying of this sample (at 80°C, < 1 *10-3 mbar, 3 days) was performed as PP415-P34.
[00445] FT-Raman spectrum remained unchanged (FIG. 92). The PXRD pattern still corresponded to class 2 (FIG. 93), but the sample was less crystalline (as the peaks were broader and had a low S/N ratio).
[00446] The thermogram of TG-FTIR (FIG. 95) shows the loss of ~2.3 wt-% cyclohexane in two steps of ~25°C ~ 270°C and decomposition at temperatures T > 270° Ç.
[00447] Thus, the cyclohexane content was reduced only from 3.0 wt-% to 2.3 wt-%, confirming the tight binding of the solvent molecules. i. Class 3 - Isostructural Solvates (eg ethanol)
[00448] Several crystallization experiments resulted in solid material of class 3 and were characterized by FT-Raman, PXRD and TG-FTIR spectroscopy (table 21).
[00449] Class 3 FT-Raman spectra are clearly similar to each other (FIG. 96) but show small differences (FIG. 97). The class 3 spectra differ significantly from the spectrum of the amorphous raw material, class 1 (FIG. 98) and from the spectra of classes 2, 4, and 5 (FIG. 72).
[00450] Class 3 PXRD standards (FIG. 99) confirm the crystallinity of the materials. The patterns of the three samples are very similar to each other, but show small but significant differences (FIG. 100). The class 3 pattern differs from the crystalline patterns of classes 2, 4 and 5 (Fig. 75).
[00451] TG-FTIR thermogram of sample PP415-P6 (FIG. 100) shows the loss of ~5.4 wt-% 2PrOH from 25°C to 250°C, mostly in one step from ~ 170°C to 190°C. Decomposition starts at temperatures T > 250°C. Prior to the TG-FTIR experiments, samples were dried briefly (for ~5 min) under vacuum (10-20 mbar) to remove excess, unbound solvent. The theoretical 2PrOH content (bp = 82°C) of a hemisolvate is 5.1 wt-%.
[00452] TG-FTIR thermogram of sample PP415-P12 (FIG. 101) shows the loss of ~4.9 wt-% EtOH (with traces of water) from 25°C to 250°C, mostly in one step from approximately 160°C to 190°C. Decomposition starts at temperatures T > 250°C. The theoretical EtOH content (bp = 78°C) of a hemisolvate is 4.0 wt-%.
[00453] Thus, class 3 samples appear to be isostructural solvates of 2PrOH, EtOH and probably acetone with tightly bound solvent content. They can correspond to stoichiometric hemisolvates. It cannot be ruled out, however, that these forms are non-stoichiometric solvates.
[00454] As the Raman spectra and PXRD patterns within this class are very similar to each other, the structures can be essentially identical to each other with only small distortions of cell unit dimensions or small changes of atomic positions within the unit cell, due to to incorporated solvent molecules. Table 21. Crystallization experiments resulting in class 3 solid material
j. Class 3 Samples Drying Experiments
[00455] One of the class 3 samples (PP415-P6), obtained from a suspension equilibrium experiment in 2PrOH, was dried (as PP415-P25) under vacuum for several days (2-20 mbar, rt to 60 °C, table 22).
[00456] TG-FTIR thermogram of this class 3 dry material, sample PP415-P25 (Fig. 102), shows the loss of ~5.4 wt-% 2PrOH from 50°C to 250°C, mostly in one step 170°C to 190°C, another loss of ~1.0 wt-% 2PrOH from 290°C to 320°C and decomposition at temperatures T > 320°C. Compared to the TG-FTIR of the original sample fr class 3 PP415-P6 (FIG. 103), with a solvent content of ~5.4 wt-% 2PrOH, the solvent content does not appear to have decreased significantly.
[00457] This material was further dried (as PP415-P33, table 22) for three days under high vacuum and high temperatures (<1*10-3 mbar, 80°C) in order to dissolve the solvate and obtain a anhydrous unsolvated form of 63415.
[00458] The TG-FTIR thermogram of this even drier class 3 material, sample PP415-P33 (FIG. 103) shows ~4.2 wt-% 2PrOH loss from 50°C to 210°C, most in one step of 160°C to 190°C, another loss of ~0.5 wt-% 2PrOH from 210°C to 290°C and decomposition at temperatures T > 290°C.
[00459] Compared to the solvent content of samples PP415-P6 and PP415-P25, the solvent content decreased only from ~5.4 wt-% to ~4.8 wt-%. Table 22. Drying Experiments on Class 3 Samples

[00460] The spectra of FT-Raman of class 3 (sample PP415-P6), of dry material of class 3 (sample PP415-P25) and of the driest material of class 3 (sample PP415-P33) are identical and do not show amendments (FIG. 104).
[00461] The PXRD standards of class 3 (sample PP415-P6) and the further dried material of class 3 (sample PP415-P33) do not show significant differences, although there are some small changes and differences in the pattern of the dried raw material from class 3 (sample PP415-P25, FIG. 105). All patterns correspond to class 3.
[00462] As drying had no major effect on solvent content, it is not surprising that the FT-Raman spectra and PXRD patterns of the dried materials do not show differences compared to the undried material.
[00463] Thus, class 3 is a class of isostructural solvates (2PrOH, EtOH and probably acetone) with very tightly bound solvent molecules that could only be partially removed (~5.4 wt-% to ~4.8 wt-% ) with the drying conditions applied here (up to 3 da 1xio-3 mbar and 80°C). k. Class 4 - Acetonitrile Solvate
[00464] Class 4 was obtained only from 7:3 MeCN/H2O solvent mixture (table 23). The experiment that resulted in class 4 (PP415-P13) was repeated as PP415-P35 to prepare more material for further drying studies.
[00465] FT-Raman spectrum (FIG. 72) and the PXRD standard (FIG. 75) of class 4 (sample PP415-P13) differ significantly from the spectra and standards of classes 2, 3 and 5.
[00466] Class 4 TG-FTIR thermogram (sample PP415-P13, FIG. 106) shows the loss of ~3.4 wt-% MeCN (with traces of water) from 25°C to 270°C, most in one step from ~180°C to 210°C. Decomposition starts at temperatures T > 270°C. Prior to the TG-FTIR experiments, samples were dried briefly (for ~5 min) under vacuum (10-20 mbar) to remove excess, unbound solvent. The theoretical MeCN content (bp = 81°C) of a hemisolvate is 3.6 wt-%. Table 23. Crystallization experiments resulting in class 4 solid material
l. Class 4 Drying Experiments
[00467] Class 4 samples obtained from suspension equilibration experiments in ~ 7:3 MeCN/H2O were dried under vacuum for several days or under N2 flow (table 24). Table 24. Drying Experiments on Class 4 Samples
to a solvent content possibly MeCN and H2O, but it is difficult to determine as the amounts are small
[00468] The FT-Raman spectrum of the dry class 4 material (PP415-P26) is identical to the class 4 spectrum (PP415-P13, FIG. 107).
[00469] The PXRD standard of class 4 dry material (PP415-P26) shows small differences from the class 4 standard, sample PP415-P13 (FIG. 108). Some peaks seem better resolved, and peak intensities have changed. No amorphization is observed. The standard of PP415-P26 corresponds to class 4.
[00470] TG-FTIR thermogram of class 4 dry material, PP415-P26 (Fig. 109) shows the loss of ~2.8 wt-% MeCN from 170°C to 250°C and decomposition at temperatures T > 300° Ç. Compared to the TG-FTIR of sample PP415-P13 (FIG. 106), the solvent content of the sample has dropped from 3.4 wt-% to 2.8 wt-%.
[00471] Thus, the sample appears to be a partially dissolved solvate. As sufficient material did not remain for a second drying experiment with subsequent characterization, experiment PP415-P13 was repeated (as PP415-P35). More class 4 material was prepared and two drying experiments were performed with this prepared material: • PP415-P36: drying under vacuum (<1x10-3 mbar) at 80°C for three days • PP415-P37: drying under flow of N2 at 80°C for three days
[00472] FT-Raman spectra of these class 4 dry samples (PP415-P36 and P37) correspond to the class 4 spectrum (ie, PP415-P35, FIG. 110).
[00473] The PXRD standards (FIG. 111) of the class 4 material (sample P35-PP415) and the dry class 4 samples (samples PP415-P36 and P37-PP415) are identical. Dry samples are crystal clear.
[00474] TG-FTIR thermograms of these class 4 dry samples (FIG. 112 for PP415-P36 and FIG. 113 for PP415-P37) show only a small solvent content (MeCN and/or H2O) of ~0.6 by weight -% and ~0.9 wt-% for PP415-P36 and P37-PP415, respectively, in two steps from 25°C to 280°C. Solvent content is possibly MeCN and H2O, but it is difficult to determine since amounts are small. Decomposition starts at temperatures T > 280°C.
[00475] Thus, most of the solvent from this solvate can be removed without destroying the crystal structure. A crystalline, unsolvated form (or rather, dissolved solvate) was obtained. m. Additional Characterization of Dried and Dissolved Class 4
[00476] Drying of class 4 (MeCN solvate) resulted in a dissolved solvate with the solvent content reduced to <1 wt-% (TG-FTIR).
[00477] No change in structure occurred after dissolution (FT-Raman and PXRD). There was no significant loss of crystallinity.
[00478] Thus, an unsolvated crystalline form of 63415 was obtained, the only one known to date.
[00479] This dissolved class 4 material was characterized by DVS and DSC.
[00480] The DVS isotherm (FIG. 114) shows that during time the initial equilibrium at 50% r.u. a mass gain of ~0.4 wt-% occurred. During measurement, a gradual reversible mass loss of ~1.3 wt-% occurred after the relative humidity dropped to 50% r.u. to 0% r.u. Balance has been reached. By increasing the relative humidity by 95% r.u., a gradual mass gain of ~0.8 wt-% (relative to the equilibrium mass at 50% r.u.) was observed. Balance has been reached. After lowering the relative humidity to 50% r.u., the final mass remained 0.1 wt-% below the initial balanced mass. Mass gain ~0.7 wt-% when increasing relative humidity by 50% r.u. to 85% r.u. classified the sample as slightly hygroscopic.
[00481] The PXRD standard of the sample after measurement is unchanged compared to the standard before measurement (FIG. 115).
[00482] DSC thermogram of a sample of dissolved class 4 material (FIG. 116) shows no transition to glass attributable to the amorphous form, which would have been expected at ~150 °C, but rather a sharp endothermic peak with a maximum at T = 196.1°C (ΔH = 29.31 J/g), probably corresponding to melting and not decomposition to 270°C.
[00483] In addition, a DSC experiment was performed with a ~1:1 mixture of the amorphous material, class 1, with the dissolved class 4 material to investigate whether the amorphous material would transform and crystallize into the dissolved class 4, an event predicted to occur (if it occurred) above the glass transition temperature of the amorphous form (Tg ~ 150°C) and below the melting of the dissolved class 4 (T.=196°C).
[00484] DSC thermogram of the mixture (FIG. 117) shows an endothermic event, with a peak at T = 156.7 C (ΔH = 1.47 J/g) and a second endothermic event, with a peak at 197, 0°C (ΔH = 14.1 J/g). The first event could be attributed to amorphous material (glass transition at Tg ~ 150°C). The second event could correspond to the fusion of class 4 dissolved at Tm ~196°C. The heat of fusion (ΔH =14.1 J/g) of the mixture correlates well with half the heat of fusion (ΔH = 29.3 J/g) of pure dissolved class 4.
[00485] No exothermic event in the temperature range between the glass transition and melting corresponding to a possible crystallization of the amorphous material can be observed. Thus, no transformation from the amorphous form to the dissolved class 4 form seemed to have occurred on this time scale.
[00486] In another DSC experiment with a ~1:1 mixture of the amorphous material, class 1, with the dissolved class 4 material, heating was stopped at 173°C (between the glass transition and melting) to allow time for possible crystallization.
[00487] DSC thermogram of the mixture (FIG. 118) shows an endothermic event, with a peak at T = 161.4 C (ΔH = 0.31 J/g) and a second endothermic event, with a peak at 201, 4°C (ΔH = 11.4 J/g). As in the first experiment, it did not increase the heat of fusion of the second peak; there are no visible indications of a transformation from the amorphous form to the dissolved class 4 form.
[00488] The curved baseline (-50°C to 150°C) is most likely an artifact (due to a twisted sample holder cap). n. Class 5- THF Solvate
[00489] Class 5 was obtained only from a 1:1 THF/H2O solvent mixture (table 25).
[00490] FT-Raman spectrum (FIG. 71) and the PXRD standard (FIG. 75) of class 5 differ significantly from the spectra and standards of classes 2, 3 and 4.
[00491] Class 5 TG-FTIR thermogram (sample PP415-P14, FIG. 119) shows the loss of ~36.1 wt-% THF and H2O from 25 to 200°C, mostly in one step of ~100°C to 130°C. Prior to the TG-FTIR experiments, samples were dried briefly (for ~5 min) under vacuum (10-20 mbar) to remove excess, unbound solvent. The losses of both THF and H2O occur together in the same temperature range. Decomposition starts at temperatures T > 300°C. The theoretical THF content (bp = 66°C) of a trisolvate is 28.1 wt-%. Unfortunately, as the content of the two components cannot be quantified separately, the exact solvation state cannot be determined.
[00492] Information about the experiments and characterizations of samples PP415-P41 and P45-PP415 is presented. Table25. Crystallization experiments resulting in class 5 solid material
b raw material: PP415-P40, class 2; in all other experiments in this table PP415-P1, class 1, was used as the starting material for the 3-g scale experiment instead of the 100-mg o scale. Drying Experiments on Class 5 Samples
[00493] The class 5 sample (PP415-P14), obtained from a suspension equilibrium experiment in ~1:1 THF/H2O, was dried (as PP415-P27) under vacuum for several days (2-20 mbar, rt at 60°C, table 26). Table 26. Class 5 Samples Drying Experiments
the mostly amorphous, only a few broad peaks with a low Y/N ratio
[00494] The FT-Raman spectrum of the dry material (PP415-P27) is different from the spectrum of class 5 (PP415-P14, FIG. 120) and, with its amplified peaks, it more resembles the spectrum of class 1 , the amorphous raw materials, PP415-P1.
[00495] PXRD pattern of class 5 dry material (PP415-P27) shows only a few broad peaks of low intensity, with a low S/N ratio, indicating the poor crystallinity of the sample (FIG. 121). Some of the peaks may correspond to class 5, while others, ie at 7.35 °2θ, are new or displaced.
[00496] TG-FTIR thermogram of dry class 5 material (FIG. 122) shows a mass loss of ~0.3 wt-% from 25°C to 290°C and decomposition at temperatures T > 290°C. The sample is anhydrous.
[00497] Thus, by drying under vacuum, the material lost its solvent content and also much of its crystallinity. 13. Experiments to Prepare the Amorphous Form
[00498] Experiments aiming to prepare the amorphous form, class 1, were carried out using class 2 material (PP415-P40, table 8) as raw materials. Several strategies and methods have been tried: • Transformation from class 2 to class 5, followed by drying from class 5 to obtain the amorphous form, class 1. • Preparation of the amorphous form, class 1, directly from class 2, if possible, using solvents class 3 of ICH.
[00499] Mainly amorphous material was prepared from class 2 material in a two-step process via class 5 in the 100-mg and 3-g scale.
[00500] Other experiments were carried out in order to simplify the procedure for a one-step process, to avoid the ICH class 2 solvent THF and to obtain totally amorphous material. The most promising method was found to be the precipitation of an acetone solution in a cold water bath. This direct method gives much better results than the two-step method via class 5. a. Preparation of Amorphous Form through Class 5
[00501] Crystallization experiments using class 2, PP415-P40, as raw materials were carried out in order to transform this heptane solvate into class 5 (probably THF solvate), followed by drying of class 5 to obtain the material amorphous (table 27).
[00502] Class 5 is thought to be a good intermediate step, as it is easier to dissolve and amorphize than classes 2 or 3. Table 27. Summary of experiences aimed at preparing amorphous form, class 1, through class 5 material
a mostly amorphous, only a few broad peaks with a low Y/N ratio b. Step 1: Transformation from Class 2 to Class 5
[00503] Transformation of the heptane solvate, class 2, into the THF solvate, class 5, was successfully carried out by suspending the material PP415-P40 (heptane solvate) in a mixture of (1:1) THF/H2O and equilibrium of the suspension in rt (PP415-P41, 100 mg-scale). The resulting solid material corresponds to THF solvate, class 5 (Fig. 123).
[00504] A first experiment of increasing from mg-scale to g-scale (*30, ie 3-g scale) was carried out analogous to PP415-P41: the raw material of class 2 heptane solvate ( PP415-P40) was equilibrated in THF/H2O (1:1) for one day and successfully transformed into class 5, the THF solvate (PP415-P45, FIG. 124). ç. Step 2: Amorphization of Class 5 Material by Drying
The class 5 material (THF solvate) was dried at elevated temperature (80°C) under vacuum (~100 mbar) taking into account the conditions that can be used at the API MFG site.
[00506] After drying the material from the 100mg scale experiment, PP415-P41, for one day at 80°C and 100 mbar, it turned into mainly amorphous material (PP415-P44, Fig. 125). The PXRD pattern shows only a few broad peaks with low intensity. After additional drying (80 C, 100 mbar) overnight, the intensity of these broad peaks is further reduced (PP415-P44a). The TG-FTIR of this material shows the loss of ~0.9 wt-% THF (with traces of water) gradually from 25°C to 280°C and decomposition at temperatures T >300°C (FIG. 126).
The material from the 3-g scale experiment, PP415-P45, was also dried at 80°C and 100 mbar (as PP415-P46). It turned at night into mostly amorphous material with only a few broad peaks with low intensity (FIG. 127). After a total of four days of drying (80°C, 100 mbar), these broad peaks are still present (-P46a, Fig. 128). The TG-FTIR of this material shows no THF loss but ~0.4 wt-% water loss gradually from 25°C to 250°C and decomposition at temperatures T > 250°C (FIG. 129). d. Getting the Amorphous Form Directly
[00508] The preparation of the amorphous form from class 2 material in the two-step process via class 5 was largely, but not entirely, successful. Thus, further experiments were carried out in order to simplify the procedure for a one-step process, to avoid the use of THF of ICH class 2 solvent, and to obtain totally amorphous material (table 28).
[00509] The amorphous form, class 1, was prepared directly with the class 2 material in an evaporation experiment of a class 2 solution in THF under N2 flow (PP415-P42, Fig. 129).
[00510] In an attempt to simulate an incompletely dry heptane/hexane solvent with a significant amount of solvent remaining, an evaporation of a class 2 solution into an 8:2 THF/hexane solution was performed (hexane was used instead of heptane to have similar boiling points in the solvent mixture). However, the resulting solid corresponds to class 2, class of isostructural solvates, not class 5 (PP415-P43, FIG. 130).
[00511] To avoid the THF of ICH class 2 solvent, evaporation experiments were performed on ICH class 3 solvents.
[00512] Evaporation of a class 2 solution in EtOAc under N2 flow resulted in crystalline material with a PXRD pattern corresponding to class 2 (PP415-P47, Fig. 130). The TG-FTIR (FIG. 80) shows typical class 2 two-stage mass loss (~7.9 wt-% EtOAc total) at temperatures up to 240°C, indicating very tightly bound solvent molecules.
[00513] Evaporation into ethyl formate also gave crystalline material of class 2 and not the amorphous form (PP415-P48, FIG. 131). The TG-FTIR (FIG. 78) shows the mass loss of ~3.5 wt-% ethyl formate, first gradually and then in a clear step between 180°C and 200°C. There may be further loss of ethyl formate concomitant with decomposition at T > 240°C.
[00514] However, class 2 material successfully transformed into the amorphous form, class 1 by adding an acetone solution to a cold (5°C) water bath (PP415-P49, FIG. 132) .
[00515] This direct method for preparing the amorphous form gives better results and is a more promising route than the two-step process. Table 28. Summary experiments aiming to obtain the amorphous form directly from class 2 raw material
14. Instrumental - Typical Measurement Conditions
[00516] FT-Raman spectroscopy: Bruker RFS100 with OPUS 6.5 software; 1064 nm Nd:YAG excitation, Ge detector, range 3500-100 cm-1; typical measurement conditions: 100-300 mW rated laser power, 64-128 scans, 2 cm-1 resolution.
[00517] PXRD: Stoe Stadi P; MynIK Detector; Cu-Kα radiation; standard measurement conditions: transmission; 40 kV and 40 mA tube power; Ge curved monochromator; 0.02 °2θ step size, 12 s or 60 step time, 1.5-50.5 °2θ or 1.0-55 °2θ scan interval; detector mode: step scan; 1 °2θ or 6 °2θ step detector; standard sample preparation: 10 to 20 mg sample was placed between two acetate sheets; Sample support: Stoe broadcast sample support; the sample was rotated during measurement.
TG-FTIR: Netzsch rmo-Microbalance TG 209 with Bruker FT-IR Spectrometer Vector 22; aluminum crucible (with microhole), N2 atmosphere, heating rate 10 K/min, range 25250 °C or 25-350°C.
[00519] DSC: Perkin Elmer DSC 7; gold crucibles (closed or microhole), sample filled in an N2 environment, heating rate 10 K/min, range -50 to 250°C or 350°C, sometimes cooling (to -200 K/min ) down to -50°C between scans.
[00520] DVS: Projekt Messtechnik Sorptions Prüfsystem SPS 11 - 100n or Surface Measurement Systems DVS-1. The sample was placed in an aluminum or platinum holder on top of a microbalance and allowed to equilibrate for 2 h in 50% r.u. before starting the pre-set humidity program: (1) 50 ^ 0% r.u. (5%/h); 5 h at 0% r.u. (2) 0^95% r.u. (5%/h); 5 h at 95% r.u. (3) 95^50% r.u. (5%/h); 2 h at 50% r.u.
Hygroscopicity was classified based on mass gain at 85% r.u. in relation to the initial mass as follows: deliquescent (sufficient water adsorbed to form a liquid), very hygroscopic (mass increase > 15%), hygroscopic (mass increase <15% but >2%), slightly hygroscopic (mass increase <2% but >0.2%), or non-hygroscopic (mass increase <0.2%).
[00522] Solvents: For all experiments, Fluka, Merck or ABCR analytical grade solvents were used.
Determination of approximate solubility: Approximate solubilities were determined by gradually diluting a suspension of about 10 mg of substance in 0.05 ml of solvent. If the substance was not dissolved by adding a total of >10 mL of solvent, the solubility is reported as <1 mg/mL. Due to the experimental error inherent in this method, the solubility values are intended to be considered as rough estimates and should be used exclusively for the design of crystallization experiments.
[00524] Determination of Chemical Stability: Four 1.0 mg samples of the material PP415-P1 were prepared in 1.0 mL of the respective solvent. The resulting suspensions/solutions were equilibrated on an Eppendorf rmomixer Comfort temperature-controlled shaker for 7 d, 2 d, 24 h and 6 h at 25°C, at an agitation rate of 500 rpm. If necessary, the solid phase was separated by filter centrifugation (0.5-μm from PVDF membrane). The filtrates were diluted in the diluent (0.1% formic acid in MeCN) to concentrations < 0.2 mg/mL (unknown and probably lower for suspensions) and examined using the HPLC method, given in table 29. As reference, the material PP415-P1 was diluted in the diluent to a concentration of 0.25 mg/ml and added to the beginning and end of the HPLC sequence. HPLC results Table 29: HPLC method used for chemical stability determinations



























[00525] All compounds, polymorphs, formulations and methods disclosed and claimed in this document can be made and executed without undue experimentation, taking into account the present description. Although the compounds, polymorphs, formulations and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those skilled in the art that variations can be applied to the compounds, polymorphs, formulations and methods, as well as in the steps or sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents that are chemically and physiologically related can be substituted for the agents described herein, while achieving similar or the same results. All such similar substitutions and modifications apparent to those skilled in the art are considered to be within the spirit, scope and concept of the invention as defined by the appended claims. REFERENCES
The following references, insofar as they provide exemplary procedures or other details in addition to those set forth herein, are specifically incorporated herein by reference. US Patent 6,326,507 US Patent 6,974,801 US Patent 7,915,402 US Patent 7,943,778 US Patent 8,124,799 US Patent 8,129,429 U.S. Patent Publication 2009/0060873 Abraham and Kappas, Free Radical Biol. Med., 39:1-25, 2005. Aghajan et al., J Gastroenterol Hepatol., Suppl 2:10-14, 2012. Ahmad et. al., Cancer Res., 68:2920-2926, 2008. Ahmad et. al., J. Biol. Chem., 281:35764-9, 2006. Angulo et al., Eur. J. Immunol., 30:1263-1271, 2000. Araujo et. al., J. Immunol., 171(3):1572-1580, 2003. Arend and Dayer, Arthritis Rheum., 38:151-160, 1995. Bach, Hum. Immunol., 67(6):430-432 , 2006. Bagasra et al., Proc. Natl. Academic Sci. USA, 92:1204112045, 1995. Blake et. al. Am J Respir Cell Mol Biol, 42:524-36, 2010. Berridge et al., Biochemica, 4: 14-19, 1996. Botoman et al., Am. Fam. Physician, 57(1):57-68, 1998. Brandt et al., Arthritis Rheum., 43:1346-1352, 2000. Brewerton et al., Lancet., 1:904-907, 1973a. Brewerton et al., Lancet., 1:956-957, 1973b. Bronte et al., Trends Immunol., 24:302-306, 2003. Brown and DuBois J. Clin. Oncol., 23:2840-2855, 2005. Brynskov et al., N. Engl. J. Med., 321(13):845-850, 1989. Cantin et. al., J Clin Invest, 79:1665-73, 1987. Cai et al., Nat. Med., 11(2):183-190, 2005. Calin and Taurog, In: The Spondylarthritides, Calin et al. (Eds.), Oxford, UK. Oxford University Press, 179, 1998. Car et. al., Am J Respir Crit Care Med, 149:655-9, 1994. Cernuda-Morollon et. al., J Biol Chem, 276:35530-6, 2001. Chan and Kan, Proc Natl Acad Sci USA, 96:12731-6, 1999. 2006. Chauhan and Chauhan, Pathophysiology, 13(3):171-181. Chen and Kunsch, Curr Pharm Des, 10:879-91, 2004. Cho et. al., Am J Respir Cell Mol Biol, 26:175-82, 2002. Crowell et al., Mol. Cancer Ther., 2:815-823, 2003. Cuzzocrea et. al., Mol Pharmacol, 61:997-1007, 2002. Dickerson et. al., Prog Neuropsychopharmacol Biol. Psychiatry, March 6, 2007. Dinarello, Int. Rev. Immunol., 16:457-499, 1998. Dinkova-Kostova et. al., Proc. Natl. Academic Sci. USA, 102(12):4584-4589, 2005. Dionne et al., Clin. Exp. Immunol., 112(3):435-442, 1998. Douglas et. al., Am J Respir Crit Care Med, 158:220-5, 1998. Dudhgaonkar et. al., Eur. J. Pain, 10(7):573-9, 2006. Eastgate et al., Lancet, 2(8613):706-9, 1988. Eikelenboom et al., Glia, 40(2): 232-239, 2002. Ettehadi et al., Clin. Exp. Immunol., 96(1):146-151, 1994. Fischer et. al.,. Int J Chron Obstruct Pulmon Dis, 6:413-21, 2011. Forstermann, Biol. Chem., 387:1521, 2006. Funakoshi et al., Digestion, 59(1):73-78, 1998. Gebel et. al., Toxicol Sci, 115:238-52, 2010. Gehrmann et al., Glia, 15(2):141-151, 1995. Genain and Nauser, J. Mol. Med., 75:187-197, 1997 Goodman et al., Kidney Int., 72(8):945-953, 2007. Graeber et al., Glia, 40(2):252-259, 2002. Greten et al., Cell, 118:285- 296, 2004. Grivennikov and Karin, Cytokine Growth Factor Rev., 21(1):11-19, 2010. Guilherme et al., Nat. Rev. Mol. Cell Biol., 9(5):367-77, 2008 Gwee et al., Gut., 44(3):400-406., 1999. Hallgren et. al., Am Rev Respir Dis, 139:373-7, 1989. Hahn and Tsao, In: Dubois' Lupus Erythematosus, 4th Ed, Wallace and Hahn (Eds.), Lea and Febiger, Philadelphia, 195-201, 1993. Handbook of Pharmaceutical Salts: Properties, and Use, Stahl and Wermuth Eds.), Verlag Helvetica Chimica Acta, 2002. Hanson et. al., BMC Medical Genetics, 6(7), 2005. Hansson et al., Annu. Rev. Pathol. Mech. Dis., 1:297-329, 2006. Hayden and Ghosh, Cell, 132:344-62, 2008. He and Karin, Cell Res., 21(1):159-168, 2011. Honda et. al. Bioorg. Med. Chem. Lett., 12:1027-1030, 2002. Honda et. al., Bioorg. Med. Chem. Lett., 16(24):6306-6309, 2006. Honda et. al., Bioorg. Med. Chem. Lett., 7:1623-1628, 1997. Honda et. al., Bioorg. Med. Chem. Lett., 8(19):2711-2714, 1998. Honda et. al., Bioorg. Med. Chem. Lett., 9(24):3429-3434, 1999. Honda et. al., J. Med. Chem., 43:4233-4246, 2000a. Honda et. al., J. Med. Chem., 43:1866-1877, 2000b. Hotamisligil, Nature, 444(7121):860-7, 2006. Iizuka et. al., Genes Cells, 10:1113-25, 2005. Ikezaki et. al., Food Chem Toxicol, 34:327-35, 1996. Ishii et. al., J Immunol, 175:6968-75, 2005. Ishikawa et. al., Circulation, 104(15):1831-1836, 2001. Ishizawa and Dickson, J. Neuropathol. Exp. Neurol., 60(6):647-657, 2001. Jarvis, Curr. Opinion Rheumatol., 10(5):459-467, 1998. Jarvis, Pediatr. Ann., 31(7):437-446, 2002. Jonsson et al., Oral Dis., 8(3):130-140, 2002. Jonsson et al., Trends Immunol., 22(12):653- 654, 2001. Kahle et al., Ann. Rheum. Dis., 51:731-734, 1992. Kaltschmidt et al., Proc. Natl. Academic Sci. USA, 94:26422647, 1997. Karin, Nature, 441(7092):431-436, 2006. Kawakami et. al., Brain Dev., 28(4):243-246, 2006. Kawamoto et. al., J Biol Chem, 275:11291-9, 2000. Kendall-Tackett, Trauma Violence Abuse, 8(2):117-126, 2007. Kensler et. al., Annu Rev Pharmacol Toxicol, 47:89-116, 2007. Kikuchi et. al., Respir Res, 11:31, 2010. Kim et. al., Mutat Res, 690:12-23, 2010. King et. al., Am J Respir Crit Care Med, 184:92-99, 2011. Kinnula et. al., Am J Respir Crit Care Med, 172:417-22, 2005. Klingsberg et. al., Respirology, 15:19-31, 2010. Kortylewski et al., Nat. Med., 11:1314-1321, 2005. Kotzin and O'Dell, In: Samler's Immunologic Diseases, 5th Ed., Frank et al. . (Eds.), Little Brown & Co., Boston, 667-697, 1995. Kotzin, Cell, 85:303-306, 1996. Kruger et. al., J. Pharmacol. Exp. Ther., 319(3):1144-1152, 2006. Kuboyama, Kurume Med. J., 45(1):33-37, 1998. Lee et. al., Mol Cell, 36:131-40, 2009. Lee et. al., Glia., 55(7):712-22, 2007. Lencz et. al., Mol. Psychiatry, 12(6):572-80, 2007. Levonen et. al., Biochem J, 378:373-82, 2004. Li and Kong, Mol Carcinog, 48:91-104, 2009. Liby et. al., Cancer Res., 65(11):4789-4798, 2005. Liby et. al., Mol. Cancer Ther., 6(7):2113-9, 2007b. Liby et. al., Nat. Rev. Cancer, 7(5):357-356, 2007a. Lipsky, In: Harrison's Principles of Internal Medicine, Fauci et al. (Eds.), 14th Ed., NY, McGraw-Hill, 1880-1888, 1998. Liu et. al., FASEB J., 20(2):207-216, 2006. Lu et. al., J. Clin. Invest., 121(10):4015-29, 2011. Lugering et al., Ital. J. Gastroenterol. Hepatol., 30(3):338-344, 1998. Malhotra et. al., Am J Respir Crit Care Med, 178:592-604, 2008. March's Advanced Organic Chemistry: Reactions, Mecha-nisms, and Structure, 2007. Mazur et al., Cell Microbiol., 9(7):1683- 94, 2007. Mazzoni et al., J. Immunol., 168:689-695, 2002. McAlindon et al., Gut, 42(2):214-219, 1998. McGeer and McGeer, Brain Res. Brain Res. Rev., 21:195218, 1995. McGeer et al., Neurology, 19:331-338, 1996. McGonagle et al., Arthritis Rheum., 41:694-700, 1998. McGonagle et al., Curr. Opinion Rheumatol., 11:244-250, 1999. McIver et. al., Pain, 120(1-2):161-9, 2005. Mease et al., Lancet, 356:385-390, 2000. Merrill and Benvenist, Trends Neurosci., 19:331-338, 1996. Mochizuki et. al., Am J Respir Crit Care Med, 171:1260-6, 2005. Morbidity & Mortality: 2009 Chart Book on Cardiovascular, Lung, and Blood Diseases. National Heart, Lung, and Blood Institute, 2009. Morris et. al., J. Mol. Med., 80(2):96-104, 2002. Morse and Choi, Am. J. Respir. Crit. Care Med., 172(6):660-670, 2005. Morse and Choi, Am. J. Respir. Crit. Care Med., 27(1):8-16, 2002. Nath et al., Neurology, 66(1):149-150, 2006. Neal et al., BMJ., 314(7083):779-782, 1997. Nichols, Drug News Perspect., 17(2):99-104, 2004. Ohnishi et al., Int. Immunol., 6:817-830, 1994. Ogushi et. al., J Med Invest, 44:53-8, 1997. Pall, Med. Hypoth., 69:821-825, 2007. Parambil et. al., Chest, 128:3310-5, 2005. Partsch et al., Br. J. Rheumatol., 24:518-523, 1997. Pergola et. al., N Engl J Med, 365:327-336, 2011. Pica et al., Antimicrob Agents Chemother., 44(1):200-4, 2000. Pimentel et al., Am. J. Gastroenterol., 95 (12):3503-3506, 2000. Place et. al., Clin. Cancer Res., 9(7):2798-806, 2003. Prochaska and Santamaria, Anal Biochem., 169:328-336, 1988. Rajakariar et. al., Proc. Natl. Academic Sci. USA, 104(52):20979-84, 2007. Rangasamy et. al., J Clin Invest, 114:1248-59, 2004. Rangasamy et. al., J Exp Med, 202:47-59, 2005. Reimund et al., Eur. J. Clin. Invest., 28(2):145-150, 1998. Renauld, J Clin Pathol, 54:577-89, 2001. Rogler and Andus, World J. Surg., 22(4):382-389, 1998. Rooney et al., Rheumatol. Int., 10:217-219, 1990. Ross et. al., Am.J.Clin. Pathol., 120(Suppl):S53-71, 2003. Ross et. al., Expert Rev. Mol. Diagn., 3(5):573-585, 2003. Rossi et. al., Nature, 403:103-8, 2000. Rostom et al., Ann. Intern. Med., 146, 376-389, 2007. Ruster et. al., Scand. J. Rheumatol., 34(6):460-3, 2005. Sacerdoti et. al., Curr Neurovasc Res., 2(2):103-111, 2005. Saha et. al., J Biol Chem, 285:40581-92, 2010. Saiki et al., Scand. J. Gastroenterol., 33(6):616-622, 1998. Salomonsson et al., Scand. J. Immunol., 55(4):336-342, 2002. Salvemini et. al., J. Clin. Invest., 93(5):1940-1947, 1994. Sarchielli et. al., Cephalalgia, 26(9):1071-1079, 2006. Satoh et. al., Proc. Natl. Academic Sci. USA, 103(3):768-773, 2006. Schlosstein et al., NE J. Medicine, 288:704-706, 1973. Schulz et al. al., Antioxid. Redox. Sig., 10:115, 2008. Selman et. al., Ann Intern Med, 134:136-51, 2001. Sheppard, J Clin Invest, 107:1501-2, 2001. Shishodia et. al., Clin Cancer Res, 12:1828-38, 2006. Simonian and Coyle, Annu. Rev. Pharmacol. Toxicol., 36:83-106, 1996. Singh et. al., Free Rad Biol Med, 46:376-386, 2009. Sinha et al., Cancer Res., 67:4507-4513, 2007. Sporn et. al., J Nat Prod, 74:537-45, 2011. Sriram et. al., Pulm Pharmacol Ther, 22:221-36, 2009. Stack et al., Lancet, 349(9051):521-524, 1997. Standiford et. al., Chest, 103:121S, 1993. Standiford et. al., J Immunol, 151:2852-63, 1993. Stewart et al., Neurology, 48:626-632, 1997. Straus et. al., Proc Natl Acad Sci USA, 97:4844-9, 2000. Strejan et. al., J. Neuroimmunol., 7:27, 1984. Strieter, Am J Respir Crit Care Med, 165:1206-7, 2002. Suh et. al., Cancer Res., 58:717-723, 1998. Suh et. al., Cancer Res., 59(2):336-341, 1999. Sussan et al., Proc Natl Acad Sci USA, 106:250-5, 2009. Szabo et. al., Nature Rev. Drug Disc., 6:662-680, 2007. Takahashi et. al., Cancer Res., 57:1233-1237, 1997. Tamir and Tannebaum, Biochim. Biophys. Acta, 1288:F31-F36, 1996. Taniguchi et. al., Eur Respir J, 35:821-9, 2010. Targan et al., N. Engl. J. Med., 337(15):1029-1035, 1997. Thimmulappa et al., Cancer Research, 62: 5196-5203, 2002. Third Report of the National Cholesterol Education Program Expert Panel on Detection, Evaluation and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III, or ATP III), National Institutes of Health, 2001, NIH Publication No. 01-3670. Touzani et al., J. Neuroimmunol., 100(1-2):203-215, 1999. Tumlin et al., Am. J. Cardiol., 98(6A):14K-20K, 2006. van den Berg, Seminar Arthritis Rheum., 30(5S-2):7-16, 2001. van Dullemen et al., Gastroenterol., 109(1):129-135, 1995. van Hogezand and Verspaget, Drugs, 56(3):299 -305, 1998. Vazquez et al., J. Virol., 79(7):4479-91, 2005. Wakabayashi, et. al., Antioxid Redox Signal, 13:1649-63, 2010. Wang et al., Cancer Res., 66:10983-10994, 2006. Wardle, Nephrol. Dial. Transplant., 16(9):1764-8, 2001. Weyand and Goronzy, Ann. NY Acad. Sci., 987:140-149, 2003. Williams et al., Clin. Neurosci., 2(3-4):229-245, 1994. Wordsworth, In: Genes and Arthritis, Brit. Medical Bulletin, 51:249-266, 1995. Wright, Clin. Orthop. Related Res., 143:8-14, 1979. Wu et. al., Toxicol Sci, 123:590-600, 2011. Xie et. al., J. Biol. Chem., 270(12):6894-6900, 1995. Yates et al., Mol. Cancer Ther, 6(1):154-162, 2007. Yore et. al., Mol Cancer Ther, 5:3232-9, 2006. Yoh et al., Kidney Int., 60(4):1343-1353, 2001. Yu et al., Nat. Rev. Immunol., 7:41 -51, 2007. Zhou et al., Am. J. Pathol., 166(1):27-37, 2005. Zhou et al., Cancer Sci., 98:882-889, 2007. Zingarelli et al., J. Immunol., 171(12):6827-6837, 2003.
权利要求:
Claims (20)
[0001]
1. Compound, characterized by the fact that it has the formula:
[0002]
2. Polymorphic form, characterized in that it is a compound as defined in claim 1, wherein the polymorphic form has an X-ray powder diffraction pattern (CuKα) comprising a halo peak at 14 o2θ; or wherein the polymorphic form is a solvate having an X-ray powder diffraction pattern (CuKα) comprising peaks at 5.6, 7.0, 10.6, 12.7 and 14.6 o2θ; or wherein the polymorphic form is a solvate having an X-ray powder diffraction pattern (CuKα) comprising peaks at 7.0, 7.8, 8.6, 11.9, 13.9 (double peak), 14 .2 and 16.0 o2θ; or wherein the polymorphic form is an acetonitrile hemisolvate having an X-ray powder diffraction pattern (CuKα) comprising peaks at 7.5, 11.4, 15.6 and 16.6o2θ; or wherein the polymorphic form is a solvate having an X-ray powder diffraction pattern (CuKα) comprising peaks at 6.8, 9.3, 9.5, 10.5, 13.6, and 15.6 o2θ .
[0003]
3. Compound according to claim 1 or the polymorphic form according to claim 2, characterized in that it is for use in a medicine.
[0004]
4. Pharmaceutical composition, characterized in that it comprises: an active ingredient consisting of a compound, as defined in claim 1, or a polymorphic form, as defined in claim 2, and a pharmaceutically acceptable carrier.
[0005]
5. Pharmaceutical composition according to claim 4, characterized in that it is formulated for oral, intra-adiposal, intra-arterial, intra-articular, intracranial, intradermal, intralesional, intramuscular, intranasal administration , intraocular, intrapericardial, intraperitoneal, intrapleural, intraprostatic, intrarectal, intrathecal, intratracheally, intratumoral, intraumbilical, intravaginal, intravenously, intravesicular, intravitreal, liposomal, locally, mucosal, parenterally, rectally, subconjunctival , subcutaneously, sublingually, topically, transbuccal, transdermally, vaginally, in creams, in lipid compositions, through a catheter, through a wash, through continuous infusion, through infusion, through inhalation, through injection, via local delivery or via localized perfusion.
[0006]
6. Pharmaceutical composition according to claim 5, characterized in that it is formulated as a hard or soft capsule, a tablet, a syrup, a suspension, emulsion, a solution, a dispersion of solids, a wafer or an elixir.
[0007]
7. Pharmaceutical composition according to any one of claims 4 to 6, characterized in that it is formulated for topical administration in a form selected from among a lotion, a cream, a gel, an oil, an ointment, an ointment , an emulsion, a solution, and a suspension.
[0008]
8. Pharmaceutical composition according to any one of claims 4 to 7, characterized in that the amount of active ingredient is from 0.01% to 5% by weight.
[0009]
9. Use of a compound as defined in claim 1, or, characterized in that it is in the manufacture of a medicine to treat or prevent a condition associated with inflammation or oxidative stress, wherein said condition is a disease or disorder of skin, sepsis, dermatitis, arthrosis, cancer, inflammation, an autoimmune disease, inflammatory bowel disease, a complication of localized or whole-body exposure to ionizing radiation, mucositis, acute or chronic organ failure, liver disease, pancreatitis, a eye disease, a lung disease, or diabetes.
[0010]
10. Use according to claim 9, characterized in that the condition is a skin disease or disorder, selected from dermatitis, a chemical or thermal burn, a chronic wound, acne, alopecia, other hair follicle disorders, bullous epidermolysis, sunburn, complications of sunburn, a skin pigmentation disorder, an aging-related skin condition, a post-surgical wound, a scar from a skin lesion or burn , psoriasis, a dermatological manifestation of autoimmune disease or a graft versus host disease, skin cancer, and a disorder involving hyperproliferation of skin cells.
[0011]
11. Use according to claim 9, characterized in that dermatitis is allergic dermatitis, atopic dermatitis, dermatitis due to exposure to chemical substances, or radiation-induced dermatitis.
[0012]
12. Use according to claim 9, characterized by the fact that the condition is an autoimmune disease, selected from rheumatoid arthritis, lupus, Crohn's disease, and psoriasis.
[0013]
13. Use according to claim 9, characterized by the fact that the condition is a liver disease, selected from fatty liver disease and hepatitis.
[0014]
14. Use according to claim 9, characterized in that the condition is an ocular disorder, selected from uveitis, macular degeneration, glaucoma, diabetic macular edema, blepharitis, diabetic retinopathy, a disease or disorder of the corneal endothelium , postsurgical inflammation, dry eye, allergic conjunctivitis and a form of conjunctivitis.
[0015]
15. Use according to claim 9, characterized by the fact that the condition is a lung disease, selected from pulmonary inflammation, pulmonary fibrosis, COPD, asthma, cystic fibrosis and idiopathic pulmonary fibrosis.
[0016]
16. Use according to claim 9, characterized by the fact that the condition is mucositis resulting from radiotherapy or chemotherapy.
[0017]
17. Use according to claim 9, characterized by the fact that the condition is cancer, selected from a carcinoma, sarcoma, lymphoma, leukemia, melanoma, mesothelioma, multiple myeloma and seminoma.
[0018]
18. Use according to claim 9, characterized by the fact that the condition is cancer, selected from bladder cancer, blood, bone, brain, breast, central nervous system, cervix, colon , endometrium, esophagus, gallbladder, genitalia, genitourinary, head, kidney, larynx, liver, lung, muscle tissue, neck, oral or nasal mucosa, ovary, pancreas, prostate, skin, spleen, small intestine, large intestine , stomach, testicle and thyroid.
[0019]
19. Use of a compound as defined in claim 1, characterized in that it is in the manufacture of a drug to treat cancer in a patient.
[0020]
20. Use according to claim 19, characterized in that the patient is still being treated with an immunotherapy.
类似技术:
公开号 | 公开日 | 专利标题
BR112014026640B1|2021-05-18|methyl bardoxolone 2,2-difluoropropionamide derivative compounds, polymorphic forms, pharmaceutical composition, and use thereof
US11117927B2|2021-09-14|2,2-difluoropropionamide derivatives of bardoxolone methyl, polymorphic forms and methods of use thereof
NZ749873B2|2021-01-06|2,2-Difluoropropionamide derivatives of bardoxolone methyl, polymorphic forms and methods of use thereof
同族专利:
公开号 | 公开日
DK3444261T3|2021-03-08|
EP2841445B1|2017-06-14|
LT2841445T|2017-09-11|
AU2013251602B2|2017-09-28|
CY1119333T1|2018-02-14|
EP3444261B1|2021-01-13|
BR112014026640A2|2017-07-18|
NZ630222A|2016-06-24|
DK2841445T3|2017-09-11|
KR102101774B1|2020-04-20|
US20130324599A1|2013-12-05|
HK1207085A1|2016-01-22|
RS56154B1|2017-11-30|
PT2841445T|2017-09-01|
PL3444261T3|2021-07-19|
HRP20171275T1|2017-10-20|
TW201726701A|2017-08-01|
US20180009839A1|2018-01-11|
CN104395332A|2015-03-04|
RS61544B1|2021-04-29|
HUE035673T2|2018-05-28|
CL2014002886A1|2015-02-06|
CO7170182A2|2015-01-28|
AR092823A1|2015-05-06|
ZA201407326B|2016-10-26|
CN104395332B|2017-01-18|
SG11201406868XA|2014-11-27|
PT3444261T|2021-03-12|
ME02926B|2018-04-20|
CA2869783A1|2013-10-31|
US11078230B2|2021-08-03|
MX357060B|2018-06-25|
HUE053113T2|2021-06-28|
ES2634315T3|2017-09-27|
EP2841445A1|2015-03-04|
EA030468B1|2018-08-31|
SI2841445T1|2017-12-29|
LT3444261T|2021-05-10|
SI3444261T1|2021-04-30|
AU2013251602A1|2014-10-23|
US8993640B2|2015-03-31|
US9701709B2|2017-07-11|
US20210355156A1|2021-11-18|
TW201348245A|2013-12-01|
KR20150003875A|2015-01-09|
JP2015521166A|2015-07-27|
EA201491972A1|2015-02-27|
PE20150160A1|2015-02-19|
ES2861393T3|2021-10-06|
MX2014013076A|2015-05-11|
EP3444261A1|2019-02-20|
HK1207086A1|2016-01-22|
IL235275A|2018-03-29|
UA116209C2|2018-02-26|
JP6637136B2|2020-01-29|
PL2841445T3|2017-10-31|
JP6410710B2|2018-10-24|
UY34764A|2013-11-29|
US20150259377A1|2015-09-17|
DOP2014000236A|2015-04-30|
WO2013163344A1|2013-10-31|
JP2019011349A|2019-01-24|
TWI623548B|2018-05-11|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题

JPS5555153Y2|1975-03-19|1980-12-20|
NZ191586A|1978-10-10|1981-10-19|Sterling Drug Inc|Cyanoketones derived from glycyrrhetinic acid and pharmaceutical compositions|
US4395423A|1978-10-10|1983-07-26|Sterling Drug Inc.|Polycyclic cyanoketones|
US5064823A|1988-08-24|1991-11-12|Research Triangle Institute|Pentacyclic triterpenoid compounds as topoisomerase inhibitors or cell differentiation inducers|
US6025395A|1994-04-15|2000-02-15|Duke University|Method of preventing or delaying the onset and progression of Alzheimer's disease and related disorders|
US6326507B1|1998-06-19|2001-12-04|Trustees Of Dartmouth College|Therapeutic compounds and methods of use|
US6369101B1|1999-02-26|2002-04-09|Regents Of The University Of Minnesota|Therapeutic method to treat herpes virus infection|
JP5059996B2|1999-05-14|2012-10-31|ネレアスファーマシューティカルズインコーポレーテッド|Novel interleukin-1 and tumor necrosis factor-α modulator, synthesis of the modulator, and method of using the modulator|
US6649654B1|1999-11-23|2003-11-18|The Regents Of The University Of California|Methods for identifying and using IKK inhibitors|
JP2001240573A|2000-03-01|2001-09-04|Meiji Seika Kaisha Ltd|Triterpene derivative and liver disease medical treatment agent|
WO2002003996A1|2000-07-12|2002-01-17|RAJKUMAR, Sujatha|Use of dammarane-type tritepenoid saporins|
US6689767B2|2000-09-29|2004-02-10|Regents Of The University Of Minnesota|Triterpenes having antibacterial activity|
US6951847B2|2000-09-29|2005-10-04|Regents Of The University Of Minnesota|Methods of treating fungal infections using lupeol|
US6642217B2|2000-09-29|2003-11-04|Naturtek, Llc|Triterpenes having human antifungal and antiyeast activity|
US6878751B1|2000-10-19|2005-04-12|Imperial College Of Science Technology And Medicine|Administration of resveratrol to treat inflammatory respiratory disorders|
US7435755B2|2000-11-28|2008-10-14|The Trustees Of Dartmouth College|CDDO-compounds and combination therapies thereof|
CA2430454A1|2000-11-28|2002-06-20|Board Of Regents, The University Of Texas System|Cddo-compounds and combination therapies thereof|
CN1636058B|2001-05-14|2011-06-08|龚大为|Novel alanine transaminase enzyme and methods of use|
WO2006029221A2|2004-09-07|2006-03-16|Pacific Arrow Limited|Anti-tumor compounds with angeloyl groups|
US7176237B2|2002-01-15|2007-02-13|The Trustees Of Dartmouth College|Tricyclic-bis-enone derivatives and methods of use thereof|
DE60325329D1|2002-01-18|2009-01-29|Univ Minnesota|Quaternary terpene salts as biologically active surfactants|
JP2006515859A|2002-05-13|2006-06-08|トラスティーズオブダートマスカレッジ|Inhibitors and uses thereof|
WO2004089357A2|2003-04-02|2004-10-21|Regents Of The University Of Minnesota|Anti-fungal formulation of triterpene and essential oil|
WO2005042002A2|2003-10-30|2005-05-12|Entelos, Inc.|Treatment of rhematoid arthritis with flip antagonists|
US8288439B2|2003-11-04|2012-10-16|The United States Of America, As Represented By The Secretary, Department Of Health And Human Services|Methods and compositions for the inhibition of HIV-1 replication|
US20060258752A1|2004-02-12|2006-11-16|Vander Jagt David L|Method and compounds for cancer treatment utilizing NFkB as a direct or ultimate target for small molecule inhibitors|
JP2005314381A|2004-03-30|2005-11-10|Anges Mg Inc|Prophylactic/therapeutic/ameliorating agent for proliferative nephropathy|
CA2614110A1|2005-07-01|2007-01-11|The Johns Hopkins University|Compositions and methods for the treatment or prevention of disorders relating to oxidative stress|
US20070232577A1|2006-03-23|2007-10-04|Advanced Life Sciences, Inc.|Synthetic pentacyclic triterpenoids and derivatives of betulinic acid and betulin|
WO2007127791A2|2006-04-25|2007-11-08|The Administrators Of The Tulane Educational Fund|New pharmacological method for treatment of neuropathic pain|
US8389573B2|2006-06-27|2013-03-05|Wellington Laboratories Inc.|Glycyrrhetinic acid derivatives|
WO2008016095A1|2006-08-02|2008-02-07|Santen Pharmaceutical Co., Ltd.|PREVENTIVE OR REMEDY FOR KERATOCONJUNCTIVAL DISORDERS CONTAINING Nrf2 ACTIVATOR AS THE ACTIVE INGREDIENT|
JP2008110962A|2006-08-02|2008-05-15|Santen Pharmaceut Co Ltd|PREVENTING OR TREATING AGENT OF KERATOCONJUNCTIVAL TROUBLE, CONTAINING Nrf2-ACTIVATING SUBSTANCE AS ACTIVE INGREDIENT|
US8299046B2|2006-11-17|2012-10-30|Trustees Of Dartmouth College|Synthetic triterpenoids and tricyclic-bis-enones for use in stimulating bone and cartilage growth|
CA2670099A1|2006-11-17|2008-05-29|Trustees Of Dartmouth College|Synthesis and biological activities of new tricyclic-bis-enones |
WO2008097596A2|2007-02-08|2008-08-14|Biogen Idec Ma Inc.|Nrf2 screening assays and related methods and compositions|
WO2008111497A1|2007-03-08|2008-09-18|Santen Pharmaceutical Co., Ltd.|Prophylactic or therapeutic agent for ophthalmic disease associated with oxidative stress, comprising triterpenoid as active ingredient|
US20080254055A1|2007-04-11|2008-10-16|John Erich Oblong|Compositions for Regulation of Hair Growth|
WO2008136838A1|2007-05-04|2008-11-13|Trustees Of Dartmouth College|Novel amide derivatives of cddo and methods of use thereof|
US20090048205A1|2007-08-15|2009-02-19|Colin Meyer|Combination therapy with synthetic triterpenoids and gemcitabine|
US8088824B2|2007-08-15|2012-01-03|Reata Pharmaceuticals Inc.|Forms of CDDO methyl ester|
WO2009058849A1|2007-10-29|2009-05-07|University Of Rochester|Use of electrophilic compounds for inducing platelet production or maintaining platelet function|
BRPI0907423A2|2008-01-11|2020-10-27|Reata Pharmaceuticals, Inc.|synthetic triterpenoid compound for use in a method of improving kidney function in an individual, and use of that compound|
PT2271658T|2008-04-18|2017-02-13|Reata Pharmaceuticals Inc|Antioxidant inflammation modulators: c-17 homologated oleanolic acid derivatives|
NZ588708A|2008-04-18|2012-09-28|Reata Pharmaceuticals Inc|2-cyano steroid derivatives including an anti-inflammatory pharmacore|
WO2009129545A1|2008-04-18|2009-10-22|Reata Pharmaceuticals, Inc.|Antioxidant inflammation modulators: oleanolic acid derivatives with saturation in the c-ring|
DK2276493T3|2008-04-18|2019-01-02|Reata Pharmaceuticals Inc|ANTIOXIDATIVE INFLAMMATION MODULATORS: OLEANOLIC ACID DERIVATIVES WITH AMINO AND OTHER MODIFICATIONS BY C-17|
US8071632B2|2008-04-18|2011-12-06|Reata Pharmaceuticals, Inc.|Antioxidant inflammation modulators: novel derivatives of oleanolic acid|
JP5758801B2|2008-07-22|2015-08-05|トラスティーズ・オブ・ダートマス・カレッジTrustees of Dartmouth College|Monocyclic cyanoenone and description of how to use it|
WO2010053817A1|2008-11-04|2010-05-14|Trustees Of Dartmouth College|Betulinic acid derivatives and methods of use thereof|
US20120029071A1|2008-11-21|2012-02-02|The Johns Hopkins University|Compositions and methods for treating or preventing radiation injury|
KR20140016441A|2009-02-13|2014-02-07|리아타 파마슈티컬즈, 아이엔씨.|Delayed release, oral dosage compositions that contain amorphous cddo-me|
EP3165606A1|2009-05-01|2017-05-10|Ophthotech Corporation|Methods for treating or preventing ophthalmological diseases|
PT2558105T|2010-04-12|2020-01-30|Reata Pharmaceuticals Inc|Method of treating obesity using antioxidant inflammation modulators|
WO2011140078A1|2010-05-04|2011-11-10|Concert Pharmaceuticals, Inc.|Synthetic triterpenoid derivatives|
US20130288985A1|2010-07-15|2013-10-31|The Schepens Eye Research Institute Inc.|Compositions and methods of treatment of corneal endothelium disorders|
CN102079772A|2010-12-09|2011-06-01|中国药科大学|Pentacyclic triterpene-13,28-lactone compounds as well as preparation method and application thereof|
CN102070697A|2010-12-09|2011-05-25|中国药科大学|Oleanolic acid derivative, and preparation method and purpose thereof|
ES2653717T3|2010-12-17|2018-02-08|Reata Pharmaceuticals, Inc.|Pyrazolyl- and pyrimidinyl-tricyclic enones as antioxidant modulators of inflammation|
MY162186A|2011-01-31|2017-05-31|Viiv Healthcare Uk Ltd|C-17 and c-3 modified triterpenoids with hiv maturation inhibitory activity|
CN102093462B|2011-02-28|2012-11-21|贵州省中国科学院天然产物化学重点实验室|Preparation method and application of 1alpha, 2alpha-dyhydroxyl oleanolic acid|
CN102153613B|2011-02-28|2013-08-14|贵州省中国科学院天然产物化学重点实验室|Method for preparing prunellin and application|
MX348862B|2011-03-11|2017-07-03|Reata Pharmaceuticals Inc|C4-monomethyl triterpenoid derivatives and methods of use thereof.|
WO2012154554A1|2011-05-06|2012-11-15|Catabasis Pharmaceuticals, Inc.|Fatty acid triterpene derivatives and their uses|
CN102250189B|2011-05-20|2012-08-29|中国药科大学|Glycyrrhetic acid derivative with 1, 12-diene-3-ketone skeleton, its preparation method and medicinal uses|
DK2841445T3|2012-04-27|2017-09-11|Reata Pharmaceuticals Inc|2,2-DIFLUOROPROPIONAMIDE DERIVATIVES OF BARDOXOLONMETHYL, POLYMORPHE FORMS THEREOF AND PROCEDURES FOR USING IT|
US8921419B2|2012-05-08|2014-12-30|Trustees Of Dartmouth College|Triterpenoids and compositions containing the same|
WO2013169740A2|2012-05-08|2013-11-14|Trustees Of Dartmouth College|Synthetic triterpenoids and methods for modulating stem/progenitor cell gene expression|
US8981144B2|2012-05-08|2015-03-17|Trustees Of Dartmouth College|Method for synthesizing 2-cyano-3,12-dioxoolean-1, 9-dien-28-oic acid methyl ester and derivatives thereof|
WO2013188818A1|2012-06-15|2013-12-19|Reata Pharmaceuticals, Inc.|A-ring epoxidized triterpenoid-based anti-inflammation modulators and methods of use thereof|
RU2487884C1|2012-07-13|2013-07-20|Общество с ограниченной ответственностью "Дакор" |Agent having anti-oxidant, anti-inflammatory, neuroprotective, hypolipidemic, hypocholesterolemic, hypoglycemic, hepatoprotective and immunosuppressive activity|
CN103665087A|2012-09-03|2014-03-26|上海源力生物技术有限公司|Terpenoid and application thereof in medicine|
WO2014040060A1|2012-09-10|2014-03-13|Reata Pharmaceuticals, Inc.|C17-alkanediyl and alkenediyl derivatives of oleanolic acid and methods of use thereof|
EA029069B1|2012-09-10|2018-02-28|Рита Фармасьютикалз, Инк.|C17-heteroaryl derivatives of oleanolic acid and methods of use thereof|
WO2014040073A1|2012-09-10|2014-03-13|Reata Pharmaceuticals, Inc.|C13-hydroxy derivatives of oleanolic acid and methods of use thereof|
US9512094B2|2012-09-10|2016-12-06|Reata Pharmaceuticals, Inc.|C17-heteroaryl derivatives of oleanolic acid and methods of use thereof|
EP2892910B1|2012-09-10|2016-09-07|AbbVie Inc.|Glycyrrhetinic acid derivatives with anti-inflammatory activity|
US9656951B2|2012-09-28|2017-05-23|Applied Pharmaceutical Science, Inc.|Polymorphs of CDDO ethyl ester and uses thereof|
CN102875634B|2012-10-24|2014-08-20|苏州晶云药物科技有限公司|Crystal forms of 2-cyano-3, 12-dioxooleana-1, 9-diene-28-oic acid methyl ester|
CN102887936A|2012-10-24|2013-01-23|苏州晶云药物科技有限公司|Crystal forms of 2-cyano-3,12-dioxoolean-1,9-diene-28-oic acid methyl ester|
TWI649330B|2013-04-24|2019-02-01|艾伯維有限公司|2,2-difluoropropionamide derivative of methyl bardoxolone, polymorph thereof and use thereof|
TWI716341B|2013-08-23|2021-01-21|美商瑞塔醫藥有限責任公司|Methods of treating and preventing endothelial dysfunction using bardoxolone methyl or analogs thereof|
WO2015112792A1|2014-01-24|2015-07-30|Abbvie Inc.|Aryl and arylalkyl substituted pyrazolyl and pyrimidinyl tricyclic enones as antioxidant inflammation modulators|
WO2016130927A1|2015-02-12|2016-08-18|Reata Pharmaceuticals, Inc.|Imidazolyl tricyclic enones as antioxidant iflammation modulators|
EP3353190B1|2015-09-23|2021-03-17|Reata Pharmaceuticals, Inc.|C4-modified oleanolic acid derivatives for inhibition of il-17 and other uses|
MA46749A|2016-11-08|2019-09-18|Reata Pharmaceuticals Inc|METHODS OF TREATMENT OF ALPORT SYNDROME USING BARDOXOLONE METHYL OR ANALOGUES OF THE LATTER|
TW201835051A|2016-12-16|2018-10-01|美商瑞塔醫藥有限責任公司|Pyrimidine tricyclic enone derivatives for inhibition of ROR[gamma] and other uses|BRPI0907423A2|2008-01-11|2020-10-27|Reata Pharmaceuticals, Inc.|synthetic triterpenoid compound for use in a method of improving kidney function in an individual, and use of that compound|
DK2276493T3|2008-04-18|2019-01-02|Reata Pharmaceuticals Inc|ANTIOXIDATIVE INFLAMMATION MODULATORS: OLEANOLIC ACID DERIVATIVES WITH AMINO AND OTHER MODIFICATIONS BY C-17|
NZ588708A|2008-04-18|2012-09-28|Reata Pharmaceuticals Inc|2-cyano steroid derivatives including an anti-inflammatory pharmacore|
ES2653717T3|2010-12-17|2018-02-08|Reata Pharmaceuticals, Inc.|Pyrazolyl- and pyrimidinyl-tricyclic enones as antioxidant modulators of inflammation|
DK2841445T3|2012-04-27|2017-09-11|Reata Pharmaceuticals Inc|2,2-DIFLUOROPROPIONAMIDE DERIVATIVES OF BARDOXOLONMETHYL, POLYMORPHE FORMS THEREOF AND PROCEDURES FOR USING IT|
WO2013188818A1|2012-06-15|2013-12-19|Reata Pharmaceuticals, Inc.|A-ring epoxidized triterpenoid-based anti-inflammation modulators and methods of use thereof|
WO2014040060A1|2012-09-10|2014-03-13|Reata Pharmaceuticals, Inc.|C17-alkanediyl and alkenediyl derivatives of oleanolic acid and methods of use thereof|
US9512094B2|2012-09-10|2016-12-06|Reata Pharmaceuticals, Inc.|C17-heteroaryl derivatives of oleanolic acid and methods of use thereof|
TWI649330B|2013-04-24|2019-02-01|艾伯維有限公司|2,2-difluoropropionamide derivative of methyl bardoxolone, polymorph thereof and use thereof|
EP3212195A4|2014-10-31|2018-06-06|The Regents of The University of California|Compositions and methods for treating hiv-associated cognitive dysfunction|
WO2016130927A1|2015-02-12|2016-08-18|Reata Pharmaceuticals, Inc.|Imidazolyl tricyclic enones as antioxidant iflammation modulators|
MA46749A|2016-11-08|2019-09-18|Reata Pharmaceuticals Inc|METHODS OF TREATMENT OF ALPORT SYNDROME USING BARDOXOLONE METHYL OR ANALOGUES OF THE LATTER|
EP3956342A1|2019-02-15|2022-02-23|Triterpenoid Therapeutics, Inc.|Methods and compositions for inhibiting the nlrp3 inflammasome and/or lon protease|
WO2021113551A1|2019-12-03|2021-06-10|Baylor College Of Medicine|Therapeutic compounds for methods of use in insulin resistance|
法律状态:
2017-12-12| B07D| Technical examination (opinion) related to article 229 of industrial property law [chapter 7.4 patent gazette]|Free format text: DE ACORDO COM O ARTIGO 229-C DA LEI NO 10196/2001, QUE MODIFICOU A LEI NO 9279/96, A CONCESSAO DA PATENTE ESTA CONDICIONADA A ANUENCIA PREVIA DA ANVISA. CONSIDERANDO A APROVACAO DOS TERMOS DO PARECER NO 337/PGF/EA/2010, BEM COMO A PORTARIA INTERMINISTERIAL NO 1065 DE 24/05/2012, ENCAMINHA-SE O PRESENTE PEDIDO PARA AS PROVIDENCIAS CABIVEIS. |
2018-03-06| B06F| Objections, documents and/or translations needed after an examination request according [chapter 6.6 patent gazette]|
2019-04-16| B07E| Notification of approval relating to section 229 industrial property law [chapter 7.5 patent gazette]|Free format text: NOTIFICACAO DE ANUENCIA RELACIONADA COM O ART 229 DA LPI |
2020-02-04| B09A| Decision: intention to grant [chapter 9.1 patent gazette]|
2020-04-14| B09X| Republication of the decision to grant [chapter 9.1.3 patent gazette]|
2021-05-18| B16A| Patent or certificate of addition of invention granted [chapter 16.1 patent gazette]|Free format text: PRAZO DE VALIDADE: 20 (VINTE) ANOS CONTADOS A PARTIR DE 24/04/2013, OBSERVADAS AS CONDICOES LEGAIS. |
优先权:
申请号 | 申请日 | 专利标题
US201261687669P| true| 2012-04-27|2012-04-27|
US61/687,669|2012-04-27|
US201361775288P| true| 2013-03-08|2013-03-08|
US61/775,288|2013-03-08|
US201361780444P| true| 2013-03-13|2013-03-13|
US61/780,444|2013-03-13|
PCT/US2013/038064|WO2013163344A1|2012-04-27|2013-04-24|2.2-difluoropropionamide derivatives of bardoxolone methyl, polymorphic forms and methods of use thereof|
[返回顶部]