US20140179779A1 - Deuterium Substituted Fumarate Derivatives - Google Patents

Deuterium Substituted Fumarate Derivatives Download PDF

Info

Publication number
US20140179779A1
US20140179779A1 US14/136,990 US201314136990A US2014179779A1 US 20140179779 A1 US20140179779 A1 US 20140179779A1 US 201314136990 A US201314136990 A US 201314136990A US 2014179779 A1 US2014179779 A1 US 2014179779A1
Authority
US
United States
Prior art keywords
compound
formula
deuterium
disease
compounds
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/136,990
Other languages
English (en)
Inventor
Jianhua Chao
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Biogen MA Inc
Original Assignee
Biogen Idec MA Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Biogen Idec MA Inc filed Critical Biogen Idec MA Inc
Priority to US14/136,990 priority Critical patent/US20140179779A1/en
Assigned to BIOGEN IDEC MA INC. reassignment BIOGEN IDEC MA INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHAO, JIANHUA
Publication of US20140179779A1 publication Critical patent/US20140179779A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C69/00Esters of carboxylic acids; Esters of carbonic or haloformic acids
    • C07C69/52Esters of acyclic unsaturated carboxylic acids having the esterified carboxyl group bound to an acyclic carbon atom
    • C07C69/593Dicarboxylic acid esters having only one carbon-to-carbon double bond
    • C07C69/60Maleic acid esters; Fumaric acid esters
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/22Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
    • A61K31/225Polycarboxylic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/235Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids having an aromatic ring attached to a carboxyl group
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/235Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids having an aromatic ring attached to a carboxyl group
    • A61K31/24Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids having an aromatic ring attached to a carboxyl group having an amino or nitro group
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/02Muscle relaxants, e.g. for tetanus or cramps
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • C07B59/001Acyclic or carbocyclic compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/02Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C229/34Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton containing six-membered aromatic rings
    • C07C229/36Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton containing six-membered aromatic rings with at least one amino group and one carboxyl group bound to the same carbon atom of the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C69/00Esters of carboxylic acids; Esters of carbonic or haloformic acids
    • C07C69/52Esters of acyclic unsaturated carboxylic acids having the esterified carboxyl group bound to an acyclic carbon atom
    • C07C69/533Monocarboxylic acid esters having only one carbon-to-carbon double bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C69/00Esters of carboxylic acids; Esters of carbonic or haloformic acids
    • C07C69/76Esters of carboxylic acids having a carboxyl group bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/14Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D295/145Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals with the ring nitrogen atoms and the carbon atoms with three bonds to hetero atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings

Definitions

  • BG-12 (dimethyl fumarate) is currently undergoing regulatory review as a potential oral treatment for multiple sclerosis (MS), following the completion of two large Phase 3 studies in patients with relapsing-remitting MS (RRMS).
  • CONFIRM which additionally evaluated subcutaneous glatiramer acetate as an active reference treatment (rater-blind). See, e.g., Fox R J, et al. N Engl J Med 2012; 367: 1087-97.
  • DMF dimethyl fumarate
  • Nrf2 is expressed ubiquitously in a range of tissues and, under normal basal conditions, is sequestered in the cytoplasm in a complex with Keap1 protein. However, when cells are under oxidative stress and overloaded with reactive oxygen or nitrogen species (ROS or RNS), or electrophilic entities, Nrf2 rapidly translocates to the nucleus, forms heterodimer with small protein Maf, then binds to the antioxidant response element, resulting in increased transcription of several antioxidant and detoxifying genes including NQO-1, HO-1, and SRXN1. See, e.g., Nguyen et al., Annu Rev Pharmacol Toxicol 2003; 43:233-260; McMahon et al., Cancer Res 2001; 61:3299-3307.
  • Sustained oxidative stress has been implicated in the pathogenesis of a variety of neurodegenerative diseases such as MS, amyotrophic lateral sclerosis (ALS), Alzheimer's disease, and Parkinson's disease.
  • ALS amyotrophic lateral sclerosis
  • Parkinson's disease For reviews, see, e.g., van Muiswinkel et al., Curr. Drug Targets CNS—Neurol. Disord, 2005; 4:267-281; Burton N. C. et al., Comprehensive Toxicology, 2010, 59-69.
  • DMF quickly gets absorbed in vivo and converted to MMF.
  • the half-life of MMF was shown to be approximately 1 hour (0.9 h in rat at 100 mg/Kg oral dose).
  • Both DMF and MMF are metabolized by esterases which are ubiquitous in the GI tract, blood and tissues.
  • BG-12 has demonstrated an acceptable safety profile in the DEFINE and CONFIRM studies.
  • tolerability issues such as flushing and gastrointestinal events were observed. While these events are generally mild to moderate in severity, there remains a desire to reduce such events to further increase patient compliance and improve patient's quality of life.
  • These mild adverse events could be the result of off-target events induced either by DMF or MMF and or the metabolites derived from them.
  • recent reports (Hanson et al., J. Clin. Invest. 2010, 120, 2910-2919; Hanson et al., Pharmacol. Ther. 2012, 136, 1-7.) indicate that MMF induced flushing is due to the activation of the G-protein-coupled receptor HCA2 (Hydroxy-carboxylic acid receptor 2, GPR109A).
  • each of R 1 and R 2 is hydrogen, deuterium, deuterated methyl, deuterated ethyl, C 1-6 aliphatic, phenyl, 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or a 5-6 membered heteroaryl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and
  • each of R 3 and R 4 is hydrogen or deuterium, provided that the compound of formula (I) contains at least one deuterium atom and that R 1 and R 2 are not hydrogen at the same time.
  • Also provided is a method of treating a neurodegenerative disease comprising administering to a subject in need of treatment for the neurodegenerative disease an effective amount of a compound of formula (I) described herein.
  • FIGS. 1( a )-( c ) describe the PD response of DMF and compounds of Examples 1 and 2.
  • aliphatic or “aliphatic group”, as used herein, means a straight-chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation. Unless otherwise specified, aliphatic groups are optionally substituted and contain 1-6 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-5 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-4 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-3 aliphatic carbon atoms, and in yet other embodiments, aliphatic groups contain 1-2 aliphatic carbon atoms. Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, alkynyl groups and hybrids thereof.
  • carbocyclic means a monocyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic (also referred to herein as “ ”), that has a single point of attachment to the rest of the molecule.
  • carbocyclic groups are optionally substituted. Examples include cycloalkyl and cycloalkenyl.
  • aliphatic groups contain 3-7 aliphatic carbon atoms.
  • aliphatic groups contain 4-6 aliphatic carbon atoms.
  • aliphatic groups contain 5-6 aliphatic carbon atoms, and in yet other embodiments, aliphatic groups contain 6 aliphatic carbon atoms.
  • heteroaryl refers to groups having 5 to 6 ring atoms, sharing ⁇ electrons in a cyclic array; and having, in addition to carbon atoms, from 1-3 heteroatoms.
  • heteroatom refers to nitrogen, oxygen, or sulfur, and includes any oxidized form of nitrogen or sulfur, and any quaternized form of a basic nitrogen.
  • Heteroaryl groups include, without limitation, thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, and pyrazinyl. Unless otherwise specified, heteroaryl groups are optionally substituted.
  • heterocycle As used herein, the terms “heterocycle,” “heterocyclyl,” “heterocyclic radical,” and “heterocyclic ring” are used interchangeably and refer to a stable 5- to 7-membered monocyclic moiety that is either saturated or partially unsaturated, and having, in addition to carbon atoms, 1-3 heteroatoms, as defined above.
  • nitrogen includes a substituted nitrogen.
  • the nitrogen may be N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl), or + NR (as in N-substituted pyrrolidinyl).
  • a heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure and any of the ring atoms can be optionally substituted.
  • saturated or partially unsaturated heterocyclic radicals include, without limitation, tetrahydrofuranyl, tetrahydrothiophenyl pyrrolidinyl, piperidinyl, pyrrolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, and morpholinyl.
  • compounds of the invention may, when specified, contain “optionally substituted” moieties.
  • substituted whether preceded by the term “optionally” or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent.
  • an “optionally substituted” group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position.
  • Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds.
  • stable refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein.
  • optionally substituted groups include halogen, —NO 2 , —CN, —OR, —SR, —N(R) 2 , —C(O)R, —CO 2 R, —N(R)C(O)OR, —C(O)N(R) 2 , —OC(O)R, —N(R)C(O)R, —S(O)R, —S(O) 2 R, or —S(O) 2 N(R) 2 .
  • Each R is independently hydrogen or C 1-6 aliphatic; or two R groups attached to the same nitrogen are taken together with their intervening atoms to form an optionally substituted 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms, independently selected from nitrogen, oxygen, or sulfur.
  • Optionally substituted groups of aliphatic can further include phenyl, 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered heteroaryl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • cycloalkyl)alkyl For example, cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl, heterocyclylalkyl.
  • Optionally substituted groups of phenyl, heterocycle, carbocycle, and heteroaryl can further include optionally substituted aliphatic groups.
  • deuterium enrichment factor means the ratio between the isotopic abundance and the natural abundance of deuterium.
  • a position designated as having deuterium when a particular position is designated as having deuterium, it is understood that the abundance of deuterium at that position is substantially greater than the natural abundance of deuterium, which is 0.015%.
  • a position designated as having deuterium typically has a minimum isotopic enrichment factor of at least 3340 (50.1% deuterium incorporation) at each atom designated as deuterium in said compound.
  • a compound of this invention has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation).
  • deuterated methyl and “deuterated ethyl,” as used herein, means that the methyl group and the ethyl group contain at least one deuterium atom.
  • deuterated methyl include —CDH 2 , —CD 2 H, and —CD 3 .
  • deuterated ethyl include —CHDCH 3 , —CD 2 CH 3 , —CHDCDH 2 ,
  • the term “pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1-19, incorporated herein by reference.
  • the neutral forms of the compounds are regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner.
  • the parent form of the compound differs from the various salt forms in certain physical properties, such as solubility in polar solvents.
  • a “pharmaceutically acceptable carrier,” as used herein refers to pharmaceutical excipients, for example, pharmaceutically, physiologically, acceptable organic or inorganic carrier substances suitable for enteral or parenteral application that do not deleteriously react with the active agent.
  • suitable pharmaceutically acceptable carriers include water, salt solutions (such as Ringer's solution), alcohols, oils, gelatins, and carbohydrates such as lactose, amylose or starch, fatty acid esters, hydroxymethycellulose, and polyvinyl pyrrolidine.
  • Such preparations can be sterilized and, if desired, mixed with auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like that do not deleteriously react with the compounds of the invention.
  • auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like that do not deleteriously react with the compounds of the invention.
  • activation and “upregulation,” when used in reference to the Nrf2 pathway, are used interchangeably herein.
  • a drug for treating a neurological disease refers to a compound that has a therapeutic benefit in a specified neurological disease as shown in at least one animal model of a neurological disease or in human clinical trials for the treatment of a neurological disease.
  • treatment refers to therapeutic as well as prophylactic/preventative measures.
  • those in need of treatment may include individuals already having a specified disease and those who are at risk for acquiring that disease.
  • terapéuticaally effective dose and “therapeutically effective amount” refer to that amount of a compound which results in prevention or delay of onset or amelioration of symptoms of a neurological disorder in a subject or an attainment of a desired biological outcome, such as reduced neurodegeneration (e.g., demyelination, axonal loss, and neuronal death), reduced inflammation of the cells of the CNS, or reduced tissue injury caused by oxidative stress and/or inflammation in a variety of cells.
  • a desired biological outcome such as reduced neurodegeneration (e.g., demyelination, axonal loss, and neuronal death), reduced inflammation of the cells of the CNS, or reduced tissue injury caused by oxidative stress and/or inflammation in a variety of cells.
  • each of R 1 and R 2 is hydrogen, deuterium, deuterated methyl, deuterated ethyl, C 1-6 aliphatic, phenyl, 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or a 5-6 membered heteroaryl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and
  • each of R 3 and R 4 is hydrogen or deuterium, provided that the compound of formula (I) contains at least one deuterium atom and that R 1 and R 2 are not hydrogen at the same time.
  • each of R 1 and R 2 is hydrogen, deuterium, deuterated methyl, deuterated ethyl, or C 1-6 aliphatic, and
  • each of R 3 and R 4 is hydrogen or deuterium, provided that the compound of formula (I) contains at least one deuterium atom and that R 1 and R 2 are not hydrogen at the same time.
  • R 1 is hydrogen or —CH 3 . In some embodiments, R 1 is —CD 3 . In some embodiments, R 1 is —CD 2 CD 3 .
  • R 2 is —CH 2 D, —CHD 2 , or —CD 3 . In some embodiments, R 2 is H, —CH 3 , —CH 2 D, —CHD 2 , or —CD 3 .
  • R 1 is hydrogen or —CH 3 and R 2 is —CH 2 D,
  • R 1 is —CD 3 and R 2 is —CH 2 D, —CHD 2 , or —CD 3 .
  • At least one of R 3 and R 4 is deuterium. In some embodiments, both of R 3 and R 4 are deuterium.
  • R 3 and R 4 are deuterium and R 2 is hydrogen, —CH 3 , —CH 2 D, —CHD 2 , or —CD 3 . In some embodiments, both of R 3 and R 4 are deuterium and R 2 is hydrogen, —CH 3 , —CH 2 D, —CHD 2 , or —CD 3 .
  • R 1 is —CD 2 CD 3 and R 2 is H, —CH 3 , —CH 2 D,
  • the compound of formula (I) is ( 2 H 6 )dimethyl fumaric acid ester, ( 2 H 3 )methyl fumaric acid ester, ( 2 H 3 )dimethyl fumaric acid ester, or dimethyl fumaric(2,3- 2 H 2 ) acid ester.
  • the compound of formula (I) is more resistant to CYP450 enzymes as compared with compounds of similar structure but lacking deuterium substitution.
  • the compound of formula (I) will have slightly altered and slower metabolism as compared with compounds of similar structure but lacking deuterium substitution.
  • the compound of formula (I) will have longer duration of action and/or improved side effect profile as compared with compounds of similar structure but lacking deuterium substitution.
  • fumaric acid 1 can be converted to the monohydrogen ( 2 H 3 )methyl fumaric acid ester 2 by reacting with deuterated methanol under the catalytic condition of p-toluenesulfonic acid at room temperature.
  • ester 2 can react with a variety of alcohols R 1 OH (e.g., CH 3 OH, CH 2 DOH, CHD 2 OH, CD 3 OH, CD 3 CH 2 OH, or other deuterated alcohols) to provide deuterated fumaric acid esters of Formula Ia.
  • compounds of Formula 1b can be prepared in two steps by first reacting with CD 3 OD at room temperature under, e.g., the catalytic condition of p-toluene sulfonic acid followed by subsequent esterification with alcohol R 1 OH (e.g., CH 3 OH, CH 2 DOH, CHD 2 OH, CD 3 OH, CD 3 CH 2 OH, or other deuterated alcohols) using either Method A or B.
  • CD 3 OD e.g., the catalytic condition of p-toluene sulfonic acid
  • R 1 OH e.g., CH 3 OH, CH 2 DOH, CHD 2 OH, CD 3 OH, CD 3 CH 2 OH, or other deuterated alcohols
  • Compounds of Formula Ic can be prepared by reacting 3 with a variety of alcohol R 1 OH under catalytic acid condition at ambient temperature. Treatment of compounds of Formula Ic under the conditions of HOBT, EDCI, and DiPEA will generate diester compounds of Formula Id.
  • compositions comprising a compound of formula (I) or a compound of formula (I) in combination with a pharmaceutically acceptable excipient (e.g., carrier).
  • a pharmaceutically acceptable excipient e.g., carrier
  • Compounds of formula (I) can be administered by any method that permits the delivery of the compound to a subject in need thereof.
  • compounds of formula (I) can be administered by orally, intranasally, transdermally, subcutaneously, intradermally, vaginally, intraaurally, intraocularly, intramuscularly, buccally, rectally, transmucosally, or via inhalation, or intravenous administration.
  • compounds of formula (I) can be administered via pills, tablets, microtablets, pellets, micropellets, capsules (e.g., containing microtablets), suppositories, liquid formulations for oral administration, and in the form of dietary supplements.
  • Oral formulations e.g., tablets and microtablets
  • the mean diameter of a microtablet is about 1-5 mm, e.g., about 1-3 mm or about 2 mm.
  • compositions can include well-known pharmaceutically acceptable excipients, e.g., if the composition is an aqueous solution containing the active agent, it can be an isotonic saline, 5% glucose, or others. Solubilizing agents such as cyclodextrins, or other solubilizing agents well known to those familiar with the art, can be utilized as pharmaceutical excipients for delivery of the therapeutic compound. See, e.g., U.S. Pat. Nos. 6,509,376 and 6,436,992 for some formulations containing DMF and/or MMF.
  • Solid form preparations include powders, tablets, pills, capsules, cachets, suppositories, and dispersible granules.
  • a solid carrier can be one or more substance that may also act as diluents, flavoring agents, binders, preservatives, tablet disintegrating agents, or an encapsulating material.
  • Carriers or excipients generally serve as fillers, disintegrants, lubricants, and glidants.
  • suitable carriers include magnesium carbonate, magnesium stearate, croscarmellose sodium, microcrystalline cellulose, talc, silica, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, a low melting wax, cocoa butter, and the like.
  • co-solvents include: Polysorbate 20, 60, and 80; Pluronic F-68, F-84, and P-103; cyclodextrin; and polyoxyl 35 castor oil.
  • co-solvents can be employed at a level between about 0.01% and about 2% by weight.
  • Viscosity greater than that of simple aqueous solutions may be desirable to decrease variability in dispensing the formulations, to decrease physical separation of components of a suspension or emulsion of formulation, and/or otherwise to improve the formulation.
  • Such viscosity building agents include, for example, polyvinyl alcohol, polyvinyl pyrrolidone, methyl cellulose, hydroxy propyl methylcellulose, hydroxyethyl cellulose, carboxymethyl cellulose, hydroxy propyl cellulose, chondroitin sulfate and salts thereof, hyaluronic acid and salts thereof, and combinations of the foregoing.
  • Such agents can be employed at a level between about 0.01% and about 2% by weight.
  • compositions of formula (I) can be administered in the form of a sustained or controlled release pharmaceutical formulation.
  • a sustained or controlled release pharmaceutical formulation can be prepared by various technologies by a skilled person in the art.
  • the formulation can contain the therapeutic compound, a rate-controlling polymer (i.e., a material controlling the rate at which the therapeutic compound is released from the dosage form) and optionally other excipients.
  • rate-controlling polymers are hydroxy alkyl cellulose, hydroxypropyl alkyl cellulose (e.g., hydroxypropyl methyl cellulose, hydroxypropyl ethyl cellulose, hydroxypropyl isopropyl cellulose, hydroxypropyl butyl cellulose and hydroxypropyl hexyl cellulose), poly(ethylene)oxide, alkyl cellulose (e.g., ethyl cellulose and methyl cellulose), carboxymethyl cellulose, hydrophilic cellulose derivatives, and polyethylene glycol, and compositions as described in WO 2006/037342, WO 2007/042034, WO 2007/042035, WO 2007/006308, WO 2007/006307, and WO 2006/050730.
  • Nrf2 Nuclear Factor-Erythroid 2-related factor 2; for sequence of the Nrf2, see Accession No. AAB32188
  • ARE antioxidant response element
  • the Nrf2/ARE pathway a major determinant of phase II gene induction, has been well characterized for its role in hepatic detoxification and chemoprevention through the activation of phase II gene expression.
  • ARE-regulated genes may also contribute to the maintenance of redox homeostasis by serving as endogenous anti-oxidant systems.
  • Nrf2-regulated genes contains over 200 genes encoding proteins and enzymes involved in detoxification and antioxidant response (Kwak et al., J. Biol. Chem., 2003, 278:8135) such as, e.g., glutathione peroxidase, glutathione-S-transferases (GSTs), NAD(P)H:quinone oxidoreductases, now commonly known as nicotinamide quinone oxidoreductase 1 (NQO1; EC 1.6.99.2; also known as DT diaphorase and menadione reductase), NQO2, g-glutamylcysteine synthase (g-GCS), glucuronosyltransferase, ferritin, and heme oxygenase-1 (HO-1), as well as any one of the enzymes proteins listed in Table 1 in Chen & Kunsch, Curr. Pharm. Designs, 2004, 10:879-8
  • the Nrf2-regulated gene which is used to assess the activation of the Nrf2 pathway is a phase II detoxification enzyme, an anti-oxidant enzyme, an enzyme of the NADPH generating system, and/or Nrf2 itself.
  • phase II detoxification enzymes include NQO1, NQO2, GST-Ya, GST-pi, GST-theta 2, GST-mu (1,2,3), microsomal GST 3, catalytic y-GCS, regulatory-GCS, microsomal epoxide hydrolase, UDP-glucuronosyltransferase, transaldolase, transketolase, and drug-metabolizing enzyme.
  • anti-oxidant enzymes examples include HO-1, ferritin (L), glutathione reductase, glutathione peroxidase, metallothionein I, thioredoxin, thioredoxin reductase, peroxiredoxin MSP23, Cu/Zn superoxide dismutase, and catalase.
  • the enzymes of the NADPH generating system include malic enzyme, UDP-glucose dehydrogenase, malate oxidoreductase, and glucose-6-phosphate dehydrogenase.
  • Nrf2 is sequestered in the cytoplasm to the actin-bound Kelch-like ECH-associated protein 1 (Keap1; Accession No. NP — 987096 for human Keap1), a Cullin3 ubiquitin ligase adaptor protein. More specifically, the N-terminal domain of Nrf2, known as Neh2 domain, is thought to interact with the C-terminal Kelch-like domain of Keap1. In response to xenobiotics or oxidative stress, Nrf2 is released from the Keap1/Nrf2 complex, thereby promoting nuclear translocation of Nrf2 and concomitant activation of ARE-mediated gene transcription.
  • Kelch-like ECH-associated protein 1 Kelch-like ECH-associated protein 1
  • Keap1 function requires association with Cullin3, a scaffold protein that positions Keap1 and its substrate in proximity to the E3 ligase Rbx1, allowing the substrate (Nrf2) to be polyubiquitinated and thus targeted for degradation.
  • the exact mechanism of how the Keap1/Nrf2 complex senses oxidative stress remains poorly understood.
  • Human Keap1 contains 25 cysteine residues that were hypothesized to function as sensors of oxidative stress; 9 of the cysteines are thought to be highly reactive (Dinkova-Kostova et al., PNAS, 2005, 102(12):4584-9).
  • DMF is neuroprotective in animal models of neuroinflammation and neurodegeneration and that it defends against oxidative stress-induced injury.
  • Linker R. A. et al. and Scannevin R. H. et al., supra see also Ellrichmann C, et al. PLoS One 2011; 6:e16172.
  • the neuroprotective effects of compounds of formula (I) can be evaluated in similar studies.
  • the neuroprotective effects of compounds of formula (I) can be investigated in the malonate striatal lesion model of excitotoxicity.
  • Malonate is a succinate dehydrogenase inhibitor, which is a mitochondrial enzyme that plays a central role in neuronal energy metabolism.
  • Injection of malonate into the striatal region of the brain produces a lesion that is excitotoxic in character, as it can be blocked by systemic administration of N-methyl-D-aspartate (NMDA) receptor antagonists and has little inflammatory involvement.
  • NMDA N-methyl-D-aspartate
  • Intrastriatal malonate injection has been used as a model for acute neurodegeneration, and the potential therapeutic effects of test compounds of formula (I) can be explored in this setting. See, e.g., Scannevin R. H.
  • cuprizone is a neurotoxicant that when administered chronically to mice results in demyelination in the central nervous system, and has been used as a model to investigate modulation of remyelination.
  • rapamycin in addition to cuprizone results in more robust and consistent demyelination, presumably due to the anti-proliferative effect of stimulating the mammalian target of rapamycin (mTOR) receptor and pathway.
  • mTOR mammalian target of rapamycin
  • ROS/RNS are most damaging in the brain and neuronal tissue, where they attack post-mitotic (i.e., non-dividing) cells such as glial cells and neurons, which are particularly sensitive to free radicals, leading to neuronal damage.
  • Oxidative stress has been implicated in the pathogenesis of a variety of neurodegenerative diseases, including MS, ALS, Alzheimer's disease, Huntington disease, and Parkinson's disease.
  • MS post-mitotic
  • glial cells and neurons which are particularly sensitive to free radicals, leading to neuronal damage.
  • Oxidative stress has been implicated in the pathogenesis of a variety of neurodegenerative diseases, including MS, ALS, Alzheimer's disease, Huntington disease, and Parkinson's disease.
  • van Muiswinkel et al. Curr. Drug Targets CNS—Neurol. Disord., 2005, 4:267-281.
  • Nrf2 An anti-oxidative enzyme under control of Nrf2, NQO1 (NAD(P)H dehydrogenase, quinone 1), was reported to be substantially upregulated in the brain tissues of AD and PD subjects (Muiswinkel et al., Neurobiol. Aging, 2004, 25:1253). Similarly, increased expression of NQO1 was reported in the ALS subjects' spinal cord (Muiswinkel et al., Curr. Drug Targets—CNS. Neurol. Disord., 2005, 4:267-281) and in active and chronic lesions in the brains of patients suffering from MS (van Horssen et al., Free Radical Biol . & Med., 2006, 41:311-311). These observations indicate that the Nr12 pathway may be activated in neurodegenerative and neuroinflammatory diseases as an endogenous protective mechanism.
  • the invention provides methods of treating, slowing, or preventing a neurological disease by treating (e.g., orally administering to) a subject in need thereof one or more compounds of formula (I).
  • neurological diseases include MS, ALS, Alzheimer's disease, Parkinson's disease and Huntington's disease.
  • the subject is mammalian, and can be a rodent or another laboratory animal, e.g., a non-human primate. In preferred embodiments, the subject is human.
  • neurodegenerative diseases include acute haemorrhagic leucoencephalomyelitis, Hurst's disease, acute disseminated encephalomyelitis, optic neuritis, spinal cord lesions, acute necrotizing myelitis, transverse myelitis, chronic progressive myelopathy, progressive multifocal leukoencephalopathy (PML), radiation myelopathy, HTLV-1 associated myelopathy, monophasic isolated demyelination, central pontine myelinolysis, and leucodystrophy (e.g., adrenoleucodystrophy, metachromatic leucodystrophy, Krabbe's disease, Canavan's disease, Alexander's disease, Pelizaeus-Merbacher disease, vanishing white matter disease, oculodentodigital syndrome), chronic inflammatory demyelinating polyneuritis (CIDP), and acute inflammatory demyelinating polyneuropathy (AIDP).
  • CIDP chronic inflammatory
  • GBS Guillain-Barre syndrome
  • MG myasthenia gravis
  • ELS Eaton Lambert Syndrome
  • encephalomyelitis e.g., encephalomyelitis
  • Other diseases for which compounds of formula (I) may be therapeutically effective include inflammatory bowel disease, Crohn's disease, lupus, asthma, and other inflammatory diseases.
  • Nrf2 pathway Activation of the Nrf2 pathway has demonstrated protective benefits in several neurodegenerative disease models. See, e.g., Calkins M J et al., Toxicoi Sci 2010; 115:557-568.
  • EAE Experimental Autoimmune Encephalomyelitis
  • MS animal model such as Experimental Autoimmune Encephalomyelitis (EAE) (Tuohy et al., J. Immunol., 1988, 141:1126-1130, Sobel et al. J. Immunol., 1984, 132:2393-2401, and Traugott, Cell Immunol., 1989 119:114-129).
  • EAE Experimental Autoimmune Encephalomyelitis
  • Chronic relapsing EAE provides a well established experimental model for testing agents that would be useful for the treatment of MS.
  • the mouse EAE is an induced autoimmune demyelinating disease with many similarities to human MS in its clinical manifestations.
  • TMEV Thieler's murine encephalomyelitis virus
  • MHV murine hepatitis virus
  • Sindbis virus Sindbis virus as described in, e.g., Ercoli et al., J. immunol., 2006, 175:3293-3298.
  • ALS is a progressive neurodegenerative disease characterized by loss of both upper and lower motor neurons leading to body and facial muscle weakness. Life expectancy is approximately 3 years post diagnosis. The unmet need is extremely high as Rilutek (riluzole) is the only approved disease modifier and offers only a modest benefit (extends survival by about 3 months).
  • a commonly used animal model is the mouse model with ALS-linked SOD1 G93A mutation. It has been shown recently that activation of the Nrf2 pathway via genetic overexpression or pharmacological induction conferred benefit in an hSOD1 G93A animal model. See Vargas M. R., et al., J. Neurosci., 2008, 28(50):1357-13581.
  • AD Alzheimer's disease
  • Ab extracellular amyloid-beta
  • NFT neurofibrillary tangles
  • AD patients display prominent cognitive deficits such as memory loss, executive dysfunctioning, and behavior and psychological symptoms associated with dementia including paranoid and elusional behavior, hallucinations, anxieties and phobias.
  • AD animal models commonly used include spontaneous models in various species, including senescence-accelerated mice, chemical and lesion-induced rodent models, and genetically modified models developed in Drosophila melanogaster, Caenorhabditis elegans, Danio rerio and rodents.
  • spontaneous models in various species including senescence-accelerated mice, chemical and lesion-induced rodent models, and genetically modified models developed in Drosophila melanogaster, Caenorhabditis elegans, Danio rerio and rodents.
  • Parkinson's disease is characterized by the loss of ⁇ 50-70% of the dopaminergic neurons in the substantia nigra pars compacta (SNc), a profound loss of dopamine (DA) in the striatum, and the presence of intracytoplasmic inclusions called Lewy bodies (LB), which are composed mainly of ⁇ -synuclein and ubiquitin.
  • SNc substantia nigra pars compacta
  • DA dopamine
  • LB Lewy bodies
  • PD animal models can typically be divided into toxin-based (those produced by 6-hydroxydopamine (6-OHDA), 1-methyl-1,2,3,6-tetrahydropiridine (MPTP) rotenone, and paraquat) or genetic models such as those utilizing the in vivo expression of PD-related mutations (e.g., those related to alpha-synuclein, PINK1, Parkin and LRRK2).
  • 6-OHDA 6-hydroxydopamine
  • MPTP 1-methyl-1,2,3,6-tetrahydropiridine
  • paraquat paraquat
  • genetic models such as those utilizing the in vivo expression of PD-related mutations (e.g., those related to alpha-synuclein, PINK1, Parkin and LRRK2).
  • HD Huntington's disease
  • IT15 cognitive dysfunction and abnormal body movements
  • HD is characterized by progressive neurodegeneration of the striatum but also involves other regions, primarily the cerebral cortex.
  • HD animal models are typically either toxin-induced models or genetic models.
  • Toxin-induced models e.g., those based on 3-nitropropionic acid and quinolinic acid
  • mitochondrial impairment and excitotoxicity-induced cell death are both mechanisms of degeneration seen in the HD brain.
  • transgenic and knock-out mice include transgenic and knock-out mice, as well as a model that uses a viral vector to encode the gene mutation in certain areas of the brain.
  • a model that uses a viral vector to encode the gene mutation in certain areas of the brain See, e.g., Ramaswamy et al., ILAR J 2007; 48(9):356-373.
  • a compound may be optionally tested in at least one additional animal model (see, generally, Immunologic Defects in Laboratory Animals, eds. Gershwin et al., Plenum Press, 1981), for example, such as the following: the SWR X NZB (SNF1) mouse model (Uner et al., J. Autoimmune Disease, 1998, 11(3):233-240), the KRN transgenic mouse (K/BxN) model (Ji et al., Immunol.
  • SNF1 SWR X NZB
  • K/BxN KRN transgenic mouse
  • NZB X NZW mice a model for SLE (Riemekasten et al., Arthritis Rheum., 2001,) 44(10):2435-2445); the NOD mouse model of diabetes (Baxter et al., Autoimmunity, 1991, 9(1):61-67), etc.).
  • the invention further includes treating a subject having a neurodegenerative disease by combination therapy.
  • the method includes treating (e.g., orally administering to) a subject having or at risk of developing a neurodegenerative disease with a compound of formula (I) and one or more other compounds of formula (I) or one or more other therapeutic agents.
  • the one or more other therapeutic agents is a disease modifying agent.
  • the one or more other therapeutic agents alleviate the side effects of the compound of formula (I).
  • the one or more other therapeutic agent can be a therapeutic agent that can reduce the flushing (e.g., aspirin) or GI disturbance (e.g., loperamide).
  • the combination therapy requires orally administering two compounds wherein at least one of the two compounds is a compound of formula (I).
  • the subject can be treated with two compounds of formula (I).
  • the subject can be treated with a compound of formula (I) and DMF, MMF, or a DMF/MMF prodrug
  • the first compound and the second compound may be administered concurrently (as separate compositions or together in a single dosage form) or consecutively over overlapping or non-overlapping intervals.
  • the first compound and the second compound can be administered in any order.
  • the length of an overlapping interval is more than 2, 4, 6, 12, 24, 48 weeks or longer.
  • the compound of formula (I) and the one or more other therapeutic agents can be used to treat MS.
  • the one or more other therapeutic agents can be, e.g., interferon beta-1a (Avonex®, Rebif®), glatiramer (Copaxone®), modafinil, azathioprine, predisolone, mycophenolate, mofetil, mitoxantrone, natalizumab (Tysabri®), sphinogosie-1 phosphate modulator e.g., fingolimod (Gilenya®), and other drugs useful for MS treatment such as teriflunomide (Aubagio®), piroxicam, and phenidone.
  • interferon beta-1a Avonex®, Rebif®
  • glatiramer Copaxone®
  • modafinil modafinil
  • azathioprine predisolone
  • mycophenolate mofetil
  • mitoxantrone natalizuma
  • the compound of formula (I) and the one or more other therapeutic agents can be used to treat ALS.
  • the one or more other therapeutic agents is an agent or agents known or believe to be effective for ALS treatment, e.g., riluzole and dexpramipexole.
  • the compound of formula (I) and the one or more other therapeutic agents can be used to treat AD.
  • the one or more other therapeutic agents is an agent or agents known or believe to be effective for AD treatment, e.g., rosiglitazone, roloxifene, vitamin E, donepezil, tacrine, rivastigmine, galantamine, and memantine.
  • the compound of formula (I) and the one or more other therapeutic agents can be used to treat PD.
  • the one or more other therapeutic agents is an agent or agents known or believe to be effective for PD treatment include dopamine precursors such levodopa, dopamine agonists such as bromocriptine, pergolide, pramipexole, and ropinirole, MAO-B inhibitors such as selegiline, anticholinergic drugs such as benztropine, trihexyphenidyl, tricyclic antidepressants such as amitriptyline, amoxapine, clomipramine, desipramine, doxepin, imipramine, maprotiline, nortriptyline, protriptyline, amantadine, and trimipramine, some antihistamines such as diphenhydramine; antiviral drugs such as amantadine.
  • dopamine precursors such levodopa
  • dopamine agonists such as bromocriptine
  • Useful drugs for treating symptoms of Huntington's disease further include selective serotonin reuptake inhibitors (SSRI) such as fluoxetine, paroxetine, sertraline, escitalopram, citalopram, fluvosamine; norepinephrine and serotoiun reuptake inhibitors (NSR1) such as venlafaxine and duloxetine.
  • SSRI selective serotonin reuptake inhibitors
  • NSR1 serotonin reuptake inhibitors
  • compositions provided by the present invention include compositions wherein the active ingredient is contained in a therapeutically effective amount, i.e., in an amount effective to achieve its intended purpose (reduce or prevent neurodegeneration or neuroinflammation).
  • a therapeutically effective amount i.e., in an amount effective to achieve its intended purpose (reduce or prevent neurodegeneration or neuroinflammation).
  • the actual amount effective for a particular application will depend, inter alia, on the condition being treated.
  • the dosage and frequency (single or multiple doses) of compound administered can vary depending upon a variety of factors, including route of administration; size, age, sex, health, body weight, body mass index, and diet of the recipient; nature and extent of symptoms of the disease being treated (e.g., the disease responsive to Nrf2 activation); presence of other diseases or other health-related problems; kind of concurrent treatment; and complications from any disease or treatment regimen.
  • Other therapeutic regimens or agents, as mentioned above, can be used in conjunction with the methods and compounds of the invention.
  • At least one compound of formula (I) or pharmaceutically acceptable salt thereof is administered in an amount and for a period of time sufficient to reduce or prevent neurodegeneration and neuroinflammation in the subject. In one embodiment, at least one compound is administered in an amount and for a period of time sufficient to reduce astrogliosis, demyelination, axonal loss, and/or neuronal death in the subject. In one embodiment, the at least one compound or pharmaceutically acceptable salt thereof is administered in an amount and for a period of time sufficient to provide neuroprotection (e.g., restoring or increasing myelin content) to the subject.
  • neuroprotection e.g., restoring or increasing myelin content
  • Methods of the invention may include treating the subject having a neurodegenerative disease with a therapeutically effective amount of at least one compound of formula (I), which can range from about 1 mg/kg to about 50 mg/kg (e.g., from about 2.5 mg/kg to about 20 mg/kg or from about 2.5 mg/kg to about 15 mg/kg). Effective doses will also vary, as recognized by those skilled in the art, dependent on route of administration, excipient usage, and the possibility of co-usage with other therapeutic treatments including use of other therapeutic agents.
  • a therapeutically effective amount of at least one compound of formula (I) can range from about 1 mg/kg to about 50 mg/kg (e.g., from about 2.5 mg/kg to about 20 mg/kg or from about 2.5 mg/kg to about 15 mg/kg).
  • Effective doses will also vary, as recognized by those skilled in the art, dependent on route of administration, excipient usage, and the possibility of co-usage with other therapeutic treatments including use of other therapeutic agents.
  • an effective dose of a compound of formula (I) to be administered to a subject can be from about 0.1 g to about 1 g per day, for example, from about 200 mg to about 800 mg per day (e.g., from about 240 mg to about 720 mg per day; or from about 480 mg to about 720 mg per day; or about 480 mg per day; or about 720 mg per day).
  • the daily doses may be administered in separate administrations of 2, 3, 4, or 6 equal doses.
  • the effective daily dose is about 720 mg per day and is administered in 3 equal doses to a subject in need thereof (i.e., three times a day, TID).
  • the effective daily dose is about 480 mg per day and is administered in 2 equal doses to a subject in need thereof (i.e., two times a day, BID).
  • the therapeutically effective dose of a compound of formula (I) is administered to a subject in need thereof for a period of time sufficient to reduce neurodegeneration and/or neuroinflammation, e.g., by at least 30%, 50%, 100% or higher over a control over a period of at least 5, 10, 12, 20, 40, 52, 100, or 200 weeks.
  • the pharmaceutical composition is administered at least one hour before or after food is consumed by the subject in need thereof.
  • the subject experiences side effects (e.g., flushing or GI discomfort)
  • the subject can consume food shortly (e.g., 30 mins to an hour) before administered the pharmaceutical composition.
  • the subject administered a compound of formula (I) may take one or more non-steroidal anti-inflammatory drugs (e.g., aspirin) before (for example, 10 minutes to an hour, e.g., 30 minutes before) taking the pharmaceutical composition.
  • the subject administered the pharmaceutical composition takes the one or more non-steroidal anti-inflammatory drugs (e.g., aspirin) to control side effects (e.g., flushing).
  • the one or more non-steroidal anti-inflammatory drugs is selected from a group consisting of aspirin, ibuprofen, naproxen, ketoprofen, celecoxib, MK-0524, and combinations thereof.
  • the one or more non-steroidal anti-inflammatory drugs can be administered in an amount of about 50 mg to about 500 mg before taking the dosage form described above.
  • a patient takes 325 mg aspirin before taking each dosage form described above.
  • the pharmaceutical preparation further comprises administering to the patient a first dose of the pharmaceutical preparation for a first dosing period; and administering to the patient a second dose of the pharmaceutical preparation for a second dosing period.
  • the first dose is lower than the second dose (e.g., the first dose is about half of the second dose).
  • the first dosing period is at least one week (e.g., 1-4 weeks).
  • the first dose of the pharmaceutical preparation comprises about 120 mg of a compound of formula (I) and the pharmaceutical preparation is administered to the patient BID or TID (e.g., BID) for the first dosing period.
  • the second dose of the first pharmaceutical preparation comprises about 240 mg of a compound of formula (I) and the first pharmaceutical preparation is administered to the patient BID or TID (e.g., BID) for the second dosing period.
  • the subject after being administered the dose at the second dosing period, experiences more than expected level of side effects (e.g., flushing or a gastrointestinal disturbance), the subject can use a lower dose (e.g., the dose at the first dosing period) for a period (e.g., 1-4 weeks or more) sufficient to allow the side effects to decrease before returning to the dose at the second dosing period.
  • Example 1 was dosed in 0.5% HPMC suspension @ 104 mg/kg (90 mg/kg MMF-eq) to male SD rats via oral gavage. Plasma samples were collected at 5 minutes, 15 minutes, 30 minutes, 1 hour, 2 hours, 4 hours, 6 hours and 12 hours and processed according to the above procedure. The concentration of D-MMF was determined by LC/MS/MS. Brain and CSF samples were collected at 30 min, 2 hr, 4 hr and 6 hr. The concentration of D-MMF was determined by LC/MS/MS.
  • Human cancer cell line DLD-1 and breast cancer cell line MCF7 reporter stable cell lines were generated by transfection with a firefly luciferase reporter construct harboring the luciferase cDNA cloned downstream of eight catenated copies of the antioxidant response element (ARE) (GACAAAGCACCCGT; SEQ ID NO.:1). See Wang et al. Cancer Res. 2006; 66:10983-94.
  • ARE antioxidant response element
  • the cells were plated in 96-well plates at 20-50 k cells/well 24 hours prior to stimulation with the test compounds.
  • the test compounds were prepared in dimethylsulfoxide (DMSO) and diluted with culture media to required concentrations (final DMSO concentrations ⁇ 0.3%).
  • DMSO dimethylsulfoxide
  • the reporter cells were harvested 24 hours-48 hours after addition of the compounds and lysed for detection of luciferase activity. Luciferase activity in the lysates was monitored using the Bright-Glo Luciferase Assay System of Promega and Tecan Genios Pro plate reader.
  • Luciferase induction in the compound-treated cells was calculated as fold change over the baseline activity detected in control cultures treated with DMSO-containing media.
  • Nrf2 Activation in DLD-1 and MCF-7 ARE-Luc Reporter Cell Line
  • Test compounds were dosed either in suspension of 0.8% HPMC or corn oil via oral gavage to male SD rats (average weight of 250 mg, 6 animals per group, two groups), at a dose of 100 mg/kg equivalent of DMF (dosing volume: 5 ml/kg). After 30 minutes, the first group of animals was sacrificed via CO2 asphyxiation. 1.0 mL blood sample via cardiac bleed pipetted into chilled lithium heparin tubes with 10 mg sodium fluoride. Samples were centrifuged within 30 minutes at 4° C. for 15 minutes at 1500 G and plasma was transferred to chilled tubes and immediately frozen on dry ice, further kept at ⁇ 70° C. until shipment for analysis. Brain was removed; sections were weighed and frozen until analysis.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Emergency Medicine (AREA)
  • Diabetes (AREA)
  • Cardiology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Dermatology (AREA)
  • Psychology (AREA)
  • Rheumatology (AREA)
  • Hematology (AREA)
  • Pulmonology (AREA)
  • Hospice & Palliative Care (AREA)
  • Pain & Pain Management (AREA)
  • Obesity (AREA)
  • Psychiatry (AREA)
  • Communicable Diseases (AREA)
  • Endocrinology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Oncology (AREA)
  • Transplantation (AREA)
US14/136,990 2012-12-21 2013-12-20 Deuterium Substituted Fumarate Derivatives Abandoned US20140179779A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/136,990 US20140179779A1 (en) 2012-12-21 2013-12-20 Deuterium Substituted Fumarate Derivatives

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261745115P 2012-12-21 2012-12-21
US14/136,990 US20140179779A1 (en) 2012-12-21 2013-12-20 Deuterium Substituted Fumarate Derivatives

Publications (1)

Publication Number Publication Date
US20140179779A1 true US20140179779A1 (en) 2014-06-26

Family

ID=49943593

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/136,990 Abandoned US20140179779A1 (en) 2012-12-21 2013-12-20 Deuterium Substituted Fumarate Derivatives
US14/201,380 Active 2034-06-27 US10399924B2 (en) 2012-12-21 2014-03-07 Deuterium substituted fumarate derivatives

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/201,380 Active 2034-06-27 US10399924B2 (en) 2012-12-21 2014-03-07 Deuterium substituted fumarate derivatives

Country Status (8)

Country Link
US (2) US20140179779A1 (de)
EP (2) EP2934503B1 (de)
JP (2) JP6506174B2 (de)
CN (1) CN105142628A (de)
AR (1) AR094277A1 (de)
ES (1) ES2733961T3 (de)
TW (1) TW201436790A (de)
WO (1) WO2014100728A1 (de)

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9302977B2 (en) 2013-06-07 2016-04-05 Xenoport, Inc. Method of making monomethyl fumarate
US9326965B2 (en) 2014-02-28 2016-05-03 Banner Life Sciences Llc Controlled release fumarate esters
US9326947B1 (en) 2014-02-28 2016-05-03 Banner Life Sciences Llc Controlled release fumarate esters
US9416096B2 (en) 2013-09-06 2016-08-16 Xenoport, Inc. Crystalline forms of (N,N-Diethylcarbamoyl)methyl methyl (2E)but-2-ene-1,4-dioate, methods of synthesis and use
US9422226B2 (en) 2011-06-08 2016-08-23 Biogen Ma Inc. Process for preparing high purity and crystalline dimethyl fumarate
US9421182B2 (en) 2013-06-21 2016-08-23 Xenoport, Inc. Cocrystals of dimethyl fumarate
US9452972B2 (en) 2008-08-19 2016-09-27 Xenoport, Inc. Methods of using prodrugs of methyl hydrogen fumarate and pharmaceutical compositions thereof
WO2016153957A2 (en) 2015-03-20 2016-09-29 Biogen Ma Inc. Methods and compositions for the intravenous administration of fumarates for the treatment of neurological diseases
US9566259B1 (en) 2015-08-31 2017-02-14 Banner Life Sciences Llc Fumarate ester dosage forms
US9597292B2 (en) 2012-08-22 2017-03-21 Xenoport, Inc. Oral dosage forms of methyl hydrogen fumarate and prodrugs thereof
US20180064653A1 (en) * 2015-03-17 2018-03-08 Hetero Labs Limited Pharmaceutical compositions of dimethyl fumarate
US9999672B2 (en) 2014-03-24 2018-06-19 Xenoport, Inc. Pharmaceutical compositions of fumaric acid esters
US10098863B2 (en) 2014-02-28 2018-10-16 Banner Life Sciences Llc Fumarate esters
US10179118B2 (en) 2013-03-24 2019-01-15 Arbor Pharmaceuticals, Llc Pharmaceutical compositions of dimethyl fumarate
US10945984B2 (en) 2012-08-22 2021-03-16 Arbor Pharmaceuticals, Llc Methods of administering monomethyl fumarate and prodrugs thereof having reduced side effects
US10959972B2 (en) 2014-11-17 2021-03-30 Biogen Ma Inc. Methods of treating multiple sclerosis
WO2021144478A2 (en) 2020-05-06 2021-07-22 Imcyse Sa Combination treatment for fumarate-related diseases
US11903918B2 (en) 2020-01-10 2024-02-20 Banner Life Sciences Llc Fumarate ester dosage forms with enhanced gastrointestinal tolerability

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AR094277A1 (es) * 2012-12-21 2015-07-22 Biogen Idec Inc Derivados de fumarato sustituidos con deuterio
PT2970101T (pt) 2013-03-14 2018-10-04 Alkermes Pharma Ireland Ltd Pró-fármacos de fumaratos e seu uso no tratamento de várias doenças
US8669281B1 (en) 2013-03-14 2014-03-11 Alkermes Pharma Ireland Limited Prodrugs of fumarates and their use in treating various diseases
CN104415026A (zh) * 2013-08-31 2015-03-18 成都渊源生物科技有限公司 氘代富马酸衍生物在治疗多发性硬化症中的应用
NZ723269A (en) 2014-02-24 2017-04-28 Alkermes Pharma Ireland Ltd Sulfonamide and sulfinamide prodrugs of fumarates and their use in treating various diseases
MA40990A (fr) * 2014-11-19 2017-09-26 Biogen Ma Inc Formulations de matrice pharmaceutique comprenant du fumarate de diméthyle
WO2017060400A1 (en) 2015-10-07 2017-04-13 Neurovive Pharmaceutical Ab Protected carboxylic acid-based metabolites for the treatment of disesases related to mitochondrial dysfunctions
ES2846886T3 (es) * 2016-02-03 2021-07-30 Rigel Pharmaceuticals Inc Compuestos activadores de Nrf2 y usos de los mismos
CA3042123A1 (en) 2016-11-08 2018-05-17 Reata Pharmaceuticals, Inc. Methods of treating alport syndrome using bardoxolone methyl or analogs thereof
CN106946701B (zh) 2017-03-28 2020-05-15 重庆纽源生物科技有限公司 一种水杨酸基富马酸盐衍生物及在治疗帕金森症和其他神经退行性疾病应用
WO2020094767A1 (en) 2018-11-08 2020-05-14 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of nrf2 activators for the treatment of staphylococcus aureus infections
CN109364058A (zh) * 2018-12-05 2019-02-22 中南大学湘雅医院 富马酸酯及其可药用盐在制备治疗铁死亡相关疾病的药物中的应用
CA3175830A1 (en) * 2019-04-17 2020-10-22 Sundeep Dugar Prodrugs of monomethyl fumarate
CN112675159A (zh) * 2021-01-12 2021-04-20 杭州师范大学 L-苹果酸在制备防治肝脏缺血再灌注损伤药物中的应用
WO2024047248A1 (en) 2022-09-02 2024-03-07 Institut National de la Santé et de la Recherche Médicale Use of nrf2 activators for the treatment of cerebral small vessel disease

Family Cites Families (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3819738A1 (de) * 1988-06-10 1989-12-14 Stockhausen Chem Fab Gmbh Drei funktionelle estergruppen enthaltende verbindungen, zwischenprodukte zu ihrer herstellung, ihre herstellung und verwendung
DE19721099C2 (de) 1997-05-20 1999-12-02 Fumapharm Ag Muri Verwendung von Fumarsäurederivaten
DE19814358C2 (de) 1998-03-31 2002-01-17 Fumapharm Ag Muri Verwendung von Alkylhydrogenfumaraten zur Behandlung von Psoriasis, psoriatischer Arthritis, Neurodermitis und Enteritis regionalis Crohn
DE19839566C2 (de) 1998-08-31 2002-01-17 Fumapharm Ag Muri Verwendung von Fumarsäurederivaten in der Transplantationsmedizin
DE19848260C2 (de) 1998-10-20 2002-01-17 Fumapharm Ag Muri Fumarsäure-Mikrotabletten
DE19853487A1 (de) 1998-11-19 2000-05-25 Fumapharm Ag Muri Verwendung von Dialkylfumaraten
DE10000577A1 (de) 2000-01-10 2001-07-26 Fumapharm Ag Muri Verwendung von Fumarsäurederivaten zur Behandlung mitochondrialer Krankheiten
RS52083B (sr) 2001-01-12 2012-06-30 Fumapharm Ag Amidi fumarne kiseline
DE10101307A1 (de) 2001-01-12 2002-08-01 Fumapharm Ag Muri Fumarsäurederivate als NF-kappaB-Inhibitor
DE10217314A1 (de) 2002-04-18 2003-11-13 Fumapharm Ag Muri Carbocyclische und Oxacarboncyclische Fumarsäure-Oligomere
EP1663197B1 (de) 2003-09-09 2007-12-05 Fumapharm AG Verwendung von fumarsäure-derivaten zur behandlung von herzinsuffizienz und asthma
LT2801354T (lt) 2004-10-08 2017-06-26 Forward Pharma A/S Kontroliuojamo atpalaidavimo farmacinės kompozicijos, apimančios fumaro rūgšties esterį
EP1812374A1 (de) 2004-11-10 2007-08-01 Aditech Pharma AB Neue salze von fumarsäuremonoalkylesternund deren pharmazeutische verwendung
DE102005022845A1 (de) 2005-05-18 2006-11-23 Fumapharm Ag Thiobernsteinsäurederivate und deren Verwendung
EP2186819A1 (de) 2005-07-07 2010-05-19 Aditech Pharma AG Neuartige Glucopyranoseester und Glucofuranoseester von Alkylfumaraten und ihre pharmazeutische Verwendung
WO2007006307A2 (en) 2005-07-07 2007-01-18 Aditech Pharma Ab Novel salts of fumaric acid monoalkylesters and their pharmaceutical use
WO2007042035A2 (en) 2005-10-07 2007-04-19 Aditech Pharma Ab Combination therapy with fumaric acid esters for the treatment of autoimmune and/or inflammatory disorders
JP2009510137A (ja) 2005-10-07 2009-03-12 アディテック・ファルマ・アクチボラゲット フマル酸エステルを含む制御放出医薬組成物
HUE032251T2 (en) 2007-02-08 2017-09-28 Biogen Ma Inc Neuroprotection in demyelinating diseases
HRP20220902T3 (hr) 2007-02-08 2022-10-14 Biogen Ma Inc. Pripravci i njihova upotreba u liječenju multiple skleroze
US20110160253A1 (en) 2008-05-28 2011-06-30 Harbeson Scott L Deuterated tizanidine
MX2011001341A (es) * 2008-08-19 2011-03-29 Xenoport Inc Prodrogas de metil hidrogeno fumarato, sus composiciones farmaceuticas, y metodos de uso.
EP3466420A1 (de) * 2009-04-29 2019-04-10 Biogen MA Inc. Dimethylfumarat zur behandlung von friedreich-ataxie
NZ601698A (en) * 2010-01-08 2015-04-24 Catabasis Pharmaceuticals Inc Fatty acid fumarate derivatives and their uses
HUE025878T2 (en) 2010-02-12 2016-05-30 Biogen Ma Inc Neuroprotection in demyelinating diseases
SG195049A1 (en) 2011-05-26 2013-12-30 Biogen Idec Inc Methods of treating multiple sclerosis and preserving and/or increasing myelin content
US9422226B2 (en) 2011-06-08 2016-08-23 Biogen Ma Inc. Process for preparing high purity and crystalline dimethyl fumarate
EP2791181A4 (de) 2011-12-16 2015-08-05 Biogen Ma Inc Siliciumhaltige fumarsäureester
CN104220061A (zh) 2012-02-07 2014-12-17 比奥根艾迪克依蒙菲利亚公司 含有富马酸二甲酯的药物组合物
JP5918395B2 (ja) * 2012-02-07 2016-05-18 ゼノポート,インコーポレイティド モルホリノアルキルフマレート化合物、医薬組成物及び使用方法
EP2934507B1 (de) * 2012-12-21 2017-05-31 ratiopharm GmbH Prodrugs von monomethylfumarat (mmf)
AR094277A1 (es) * 2012-12-21 2015-07-22 Biogen Idec Inc Derivados de fumarato sustituidos con deuterio
CN104415026A (zh) 2013-08-31 2015-03-18 成都渊源生物科技有限公司 氘代富马酸衍生物在治疗多发性硬化症中的应用

Cited By (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9452972B2 (en) 2008-08-19 2016-09-27 Xenoport, Inc. Methods of using prodrugs of methyl hydrogen fumarate and pharmaceutical compositions thereof
US9422226B2 (en) 2011-06-08 2016-08-23 Biogen Ma Inc. Process for preparing high purity and crystalline dimethyl fumarate
US10940117B2 (en) 2012-08-22 2021-03-09 Arbor Pharmaceuticals, Llc Oral dosage forms of methyl hydrogen fumarate and prodrugs thereof
US10945984B2 (en) 2012-08-22 2021-03-16 Arbor Pharmaceuticals, Llc Methods of administering monomethyl fumarate and prodrugs thereof having reduced side effects
US9597292B2 (en) 2012-08-22 2017-03-21 Xenoport, Inc. Oral dosage forms of methyl hydrogen fumarate and prodrugs thereof
US10716760B2 (en) 2012-08-22 2020-07-21 Arbor Pharmaceuticals, Llc Oral dosage forms of methyl hydrogen fumarate and prodrugs thereof
US11938111B2 (en) 2013-03-24 2024-03-26 Arbor Pharmaceuticals, Llc Pharmaceutical compositions of dimethyl fumarate
US10179118B2 (en) 2013-03-24 2019-01-15 Arbor Pharmaceuticals, Llc Pharmaceutical compositions of dimethyl fumarate
US9302977B2 (en) 2013-06-07 2016-04-05 Xenoport, Inc. Method of making monomethyl fumarate
US9421182B2 (en) 2013-06-21 2016-08-23 Xenoport, Inc. Cocrystals of dimethyl fumarate
US9416096B2 (en) 2013-09-06 2016-08-16 Xenoport, Inc. Crystalline forms of (N,N-Diethylcarbamoyl)methyl methyl (2E)but-2-ene-1,4-dioate, methods of synthesis and use
US9682057B2 (en) 2013-09-06 2017-06-20 Xenoport, Inc. Crystalline forms of (N,N-Diethylcarbamoyl)methyl methyl (2E)but-2-ene-1,4-dioate, methods of synthesis and use
US9814691B2 (en) 2014-02-28 2017-11-14 Banner Life Sciences Llc Fumarate ester pharmaceutical compositions
US9820960B2 (en) 2014-02-28 2017-11-21 Banner Life Sciences Llc Fumarate ester pharmaceutical compositions
US9326965B2 (en) 2014-02-28 2016-05-03 Banner Life Sciences Llc Controlled release fumarate esters
US9517209B2 (en) 2014-02-28 2016-12-13 Banner Life Sciences Llc Fumarate ester pharmaceutical compositions
US10918615B2 (en) 2014-02-28 2021-02-16 Banner Life Sciences Llc Fumarate esters
US9511043B2 (en) 2014-02-28 2016-12-06 Banner Life Sciences Llc Fumarate ester pharmaceutical compositions
US10918616B2 (en) 2014-02-28 2021-02-16 Banner Life Sciences Llc Fumarate ester pharmaceutical compositions
US9326947B1 (en) 2014-02-28 2016-05-03 Banner Life Sciences Llc Controlled release fumarate esters
US10918617B2 (en) 2014-02-28 2021-02-16 Banner Life Sciences Llc Fumarate ester pharmaceutical compositions
EP3501510A1 (de) 2014-02-28 2019-06-26 Banner Life Sciences LLC Enterische weichkapseln von fumaratestern mit kontrollierter freisetzung
US10098863B2 (en) 2014-02-28 2018-10-16 Banner Life Sciences Llc Fumarate esters
US10105336B2 (en) 2014-02-28 2018-10-23 Banner Life Sciences Llc Fumarate ester pharmaceutical compositions
EP3766487A1 (de) 2014-02-28 2021-01-20 Banner Life Sciences LLC Enterische weichkapseln von fumaratestern mit kontrollierter freisetzung
US10105337B2 (en) 2014-02-28 2018-10-23 Banner Life Sciences Llc Fumarate ester pharmaceutical compositions
US11135296B2 (en) 2014-03-24 2021-10-05 Arbor Pharmaceuticals, Llc Pharmaceutical compositions of fumaric acid esters
US9999672B2 (en) 2014-03-24 2018-06-19 Xenoport, Inc. Pharmaceutical compositions of fumaric acid esters
US10959972B2 (en) 2014-11-17 2021-03-30 Biogen Ma Inc. Methods of treating multiple sclerosis
US11129806B2 (en) 2014-11-17 2021-09-28 Biogen Ma Inc. Methods of treating multiple sclerosis
EP4215191A1 (de) 2014-11-17 2023-07-26 Biogen MA Inc. Zusammensetzung enthaltend ein fumarat zur verwendung in einem verfahren zur behandlung von multipler sklerose
US11246850B2 (en) 2014-11-17 2022-02-15 Biogen Ma Inc. Methods of treating multiple sclerosis
US11007166B2 (en) 2014-11-17 2021-05-18 Biogen Ma Inc. Methods of treating multiple sclerosis
US11007167B2 (en) 2014-11-17 2021-05-18 Biogen Ma Inc. Methods of treating multiple sclerosis
EP3804711A1 (de) 2014-11-17 2021-04-14 Biogen MA Inc. Verfahren zur behandlung von multipler sklerose
US20180064653A1 (en) * 2015-03-17 2018-03-08 Hetero Labs Limited Pharmaceutical compositions of dimethyl fumarate
WO2016153957A3 (en) * 2015-03-20 2016-11-10 Biogen Ma Inc. Methods and compositions for the intravenous administration of fumarates for the treatment of neurological diseases
WO2016153957A2 (en) 2015-03-20 2016-09-29 Biogen Ma Inc. Methods and compositions for the intravenous administration of fumarates for the treatment of neurological diseases
US10105335B2 (en) 2015-08-31 2018-10-23 Banner Life Sciences Llc Fumarate ester dosage forms
US9814692B2 (en) 2015-08-31 2017-11-14 Banner Life Sciences Llc Fumarate ester dosage forms
US10945985B2 (en) 2015-08-31 2021-03-16 Banner Life Sciences Llc Fumarate ester dosage forms
US9636318B2 (en) 2015-08-31 2017-05-02 Banner Life Sciences Llc Fumarate ester dosage forms
US9820961B2 (en) 2015-08-31 2017-11-21 Banner Life Sciences Llc Fumarate ester dosage forms
US11590095B2 (en) 2015-08-31 2023-02-28 Banner Life Sciences Llc Fumarate ester dosage forms
US9636319B1 (en) 2015-08-31 2017-05-02 Banner Life Sciences Llc Fumarate ester dosage forms
US9566259B1 (en) 2015-08-31 2017-02-14 Banner Life Sciences Llc Fumarate ester dosage forms
US11903918B2 (en) 2020-01-10 2024-02-20 Banner Life Sciences Llc Fumarate ester dosage forms with enhanced gastrointestinal tolerability
WO2021144478A2 (en) 2020-05-06 2021-07-22 Imcyse Sa Combination treatment for fumarate-related diseases

Also Published As

Publication number Publication date
JP6506174B2 (ja) 2019-04-24
US10399924B2 (en) 2019-09-03
AR094277A1 (es) 2015-07-22
ES2733961T3 (es) 2019-12-03
EP2934503A1 (de) 2015-10-28
EP2934503B1 (de) 2019-04-10
WO2014100728A1 (en) 2014-06-26
JP2016510317A (ja) 2016-04-07
TW201436790A (zh) 2014-10-01
CN105142628A (zh) 2015-12-09
EP3566701A1 (de) 2019-11-13
JP2019077711A (ja) 2019-05-23
US20140194427A1 (en) 2014-07-10

Similar Documents

Publication Publication Date Title
US20140179779A1 (en) Deuterium Substituted Fumarate Derivatives
CN111039944B (zh) Mst1激酶抑制剂及其用途
JP7042548B2 (ja) Magl阻害剤の結晶形態
TW200843778A (en) Pterin analogs
JP2022105159A (ja) 統合失調症を治療するための2-((1-(2(4-フルオロフェニル)-2-オキソエチル)ピペリジン-4-イル)メチル)イソインドリン-1-オン
AU2013359315B2 (en) Methods and compositions comprising Akt inhibitors and/or phospholipase D inhibitors
CN112638866B (zh) 索拉非尼衍生物的共晶体及其制备方法
US11938122B2 (en) 5-[[4-[2-[5-(1-hydroxyethyl)pyridin-2-yl]ethoxy]phenyl]methyl]-1,3- thiazolidine-2,4-dione for treating nonalcoholic fatty liver disease
US20130079417A1 (en) Highly Selective 5-HT(2C) Receptor Agonists That Show Anti-Psychotic Effects with Antagonist Activity at the 5-HT(2B) Receptor
US20200069674A1 (en) Targeted drug rescue with novel compositions, combinations, and methods thereof
AU2015231076B2 (en) Carboxy-cyclopropyl undecanol compounds for treatment of liver disease and other medical disorders
WO2023028091A1 (en) Deuterated empathogens
WO2023038039A1 (ja) 抗肥満作用の関与する疾患の予防及び治療用医薬
US20090176835A1 (en) Pharmaceutical composition for suppression of the expression of atp citrate lyase and use thereof
EP1547614B1 (de) Medizinische zusammensetzungen zur hemmung der expression von atp-citrat-lyase und ihre verwendung
US20230365521A1 (en) Compounds for increasing the nicotinamide adenine dinucleotide in a subject and methods of use thereof
WO2005118521A1 (en) Monomethylphytosphingosine and the composition for anti-cancer containing the same
RU2537227C1 (ru) Гепатопротектор для лечения неалкогольного жирового гепатоза при сахарном диабете 2 типа
WO2024072778A1 (en) Methods and compositions for treating amyotrophic lateral sclerosis
WO2023028092A2 (en) Fluorinated empathogens

Legal Events

Date Code Title Description
AS Assignment

Owner name: BIOGEN IDEC MA INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CHAO, JIANHUA;REEL/FRAME:032163/0494

Effective date: 20140124

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION