throbber
Table of Contents For Ex. 1015A
`Certified Copy of US Provisional 60/888,921 as received by the International
`Bureau
`
`WIPO Cover Letter---------------------------------------------------- page 2
`
`Front page -------------------------------------------------------------- page 3
`
`Provisional Application Cover Sheet ------------------------------- page 4
`
`Specification ----------------------------------------------------------- pages 5 - 40
`
`Claims ------------------------------------------------------------------ pages 41 - 43
`
`Abstract ---------------------------------------------------------------- page 44
`
`
`Drawings --------------------------------------------------------------- pages 45 - 46
`
`Electronic Acknowledgment Receipt ------------------------------ pages 47 - 49
`
`
`
`Page 1 of 49
`
`

`
`Document made available under
`Patent Cooperation Treaty (PCT)
`
`the
`
`International application number: PCT IUS2008/00 1602
`
`International filing date:
`
`07 February 2008 (07.02.2008)
`
`Document type:
`
`Certified copy of priority document
`
`Document details:
`
`Country/Office: US
`Number:
`60/888,921
`08 February 2007 (08.02.2007)
`Filing date:
`
`Date of receipt at the International Bureau:
`
`26 March 2008 (26.03.2008)
`
`Remark:
`
`Priority document submitted or transmitted to the International Bureau in
`compliance with Rule 17 .1 (a) or (b)
`
`World Intellectual Property Organization (WIPO) - Geneva, Switzerland
`Organisation Mondiale de la Propriete Intellectuelle (OMPI) - Geneve, Suisse
`
`Page 2 of 49
`
`

`
`LN ITEU STAlES DEP\RT'-U:N'f OF ('O\i:\IEI{'CE
`
`Cnikd States I'l't~nl and TradNllurk Ot1'kc
`
`March 26, 2008
`
`THIS IS TO CERTIFY THAT ANNEXED HERETO IS A TRUE COPY FROM
`THE RECORDS OF THE UNITED STATES PATENT AND TRADEMARK
`OFFICE OF THOSE PAPERS OF THE BELOW IDENTIFIED PATENT
`APPLICATION THAT MET THE REQUIREMENTS TO BE GRANTED A
`FILING DATE.
`
`APPLICATION NUMBER: 601888,921
`FILING DATE: February 08, 2007
`RELA TED PCT APPLICATION NUMBER: PCTIUS08101602
`
`THE COUNTRY CODE AND NUMBER OF YOUR PRIORITY
`APPLICATION, TO BE USED FOR FILING ABROAD UNDER THE PARIS
`CONVENTION, IS US601888,921
`
`Lmkr Sf'cretal':>" of CUllIllIt'fCt'
`for lntdh:!l'tllill Pnll)l~rfy
`afld Dirertor of nlc t'nitNl Statc.'>
`I'atenl and TnH-{em:Ir!, Oflk.'
`
`Page 3 of 49
`
`

`
`PROVISIONAL APPLICATION COVER SHEET
`
`This is a request for filing a PROVISIONAL APPLICATION under 37 CFR 1.53(c).
`I Docket Number 08201.6042-00000
`I
`I
`Type a plus sign (+) inside this box '"
`INVENTOR(s)/APPLlCANT(s)
`
`I
`
`+
`
`LAST NAME
`
`FIRST
`NAME
`
`MIDDLE
`INITIAL
`
`RESIDENCE (CITY AND EITHER STATE OR FOREIGN
`COUNTRY)
`
`..
`
`..
`
`..
`
`. .
`
`TITLE OF INVENTION (500 characters max)
`
`Nrf2 SCREENING ASSAYS AND RELATED METHODS AND COMPOSITIONS
`
`CORRESPONDENCE ADDRESS
`
`BIOGEN IDEC I FINNEGAN HENDERSON, LLP
`
`Customer Number 65,779
`
`ENCLOSED APPLICATION PARTS (check all that apply)
`
`[8J Specification: 40 Pages
`
`[8J Drawing(s): 2 Sheets/2 Figures
`
`0 Other: [Number] Pages; [Description]
`
`METHOD OF PAYMENT
`
`0
`
`[8J
`
`A check or money order is
`enclosed to cover the Provisional
`Application filing fees
`
`The Commissioner is hereby
`authorized to charge any
`deficiency or credit any
`overpayment in fees to Deposit
`Account Number 06-0916.
`
`PROVISIONAL FILING FEE
`
`$200.00
`
`Total Number of Pages of specification, drawings,
`sequence or computer listing, or other papers 42. If more
`than 100 pages, add $250 for each additional 50 pages or
`fraction thereof.
`
`TOTAL FILING FEE
`
`$200.00
`
`The invention was made by an agency of the United States Government or under a contract with an
`agency of the United States Government.
`[8J No.
`o Yes, the name of the U.S. Government agency and the Government contract number are:
`[Text]
`
`Respectfully submitted on b alf of the patent practitioners associated with Customer Number 65,779.
`
`SIGNATURE
`
`f(
`
`Date February 8, 2007
`
`TYPED OR PRINTED NAME Konstantin M. Linnik
`
`REGISTRATION NO. 56,309
`
`o Additional inventors are being named on separately numbered sheets attached hereto.
`
`PROVISIONAL APPLICATION FILING ONLY
`
`Page 4 of 49
`
`

`
`Attorney Docket No. 08201.6042-00000
`
`Nrf2 SCREENING ASSAYS
`AND RELATED METHODS AND COMPOSITIONS
`
`[0001]
`
`The invention relates to the field of cell and molecular biology and
`
`to the development and use of therapeutic compounds, more particularly,
`
`compounds for treating neurological diseases, including demyelinating
`
`neurological diseases, such as, e.g., multiple sclerosis.
`
`[0002] Multiple sclerosis (MS) is an autoimmune disease with the
`
`autoimmune activity directed against central nervous system (eNS) antigens.
`
`The disease is characterized by inflammation in parts of the eNS, leading to the
`
`loss of the myelin sheathing around neuronal axons (demyelination), loss of
`
`axons, and the eventual death of neurons, oligodenrocytes and glial cells.
`
`[0003] An estimated 2,500,000 people in the world suffer from MS. It is
`
`one of the most common diseases of the eNS in young adults. MS is a chronic,
`
`progressing, disabling disease, which generally strikes its victims some time after
`
`adolescence, with diagnosis generally made between 20 and 40 years of age,
`
`although onset may occur earlier. The disease is not directly hereditary,
`
`although genetiC susceptibility plays a part in its development.
`
`Relapsing-remitting MS presents in the form of recurrent attacks of focal or
`
`multifocal neurologic dysfunction. Attacks may occur, remit, and recur,
`
`seemingly randomly over many years. Remission is often incomplete and as one
`
`attack follows another, a stepwise downward progression ensues with increasing
`
`permanent neurological deficit.
`
`[0004] Although various immunotherapeutic drugs can provide relief in
`
`patients with MS, none is capable of reversing disease progression, and some
`
`can cause serious adverse effects. Most current therapies for MS are aimed at
`
`the reduction of inflammation and suppression or modulation of the immune
`
`system. As of 2006, the available treatments for MS reduce inflammation and
`
`the number of new episodes but not all have an effect on disease progression. A
`
`number of clinical trials have shown that the suppression of inflammation in
`
`chronic MS rarely significantly limits the accumulation of disability through
`
`1
`
`Page 5 of 49
`
`

`
`Attorney Docket No. 08201.6042-00000
`
`sustained disease progression, suggesting that neuronal damage and
`
`inflammation are independent pathologies. Promoting CNS remyelination as a
`
`repair mechanism and otherwise preventing axonal loss and neuronal death are
`
`some of the important goals for the treatment of MS. For a comprehensive
`
`review of MS and its current therapies, see, e.g., McAlpine's Multiple Sclerosis,
`
`by Alastair Compston et aL, 4th edition, Churchill Livingstone Elsevier, 2006.
`
`[0005]
`
`"Phase 2 enzymes" serve as a protection mechanism in
`
`mammalian cells against oxygen/nitrogen species (ROS/RNS), electrophiles and
`
`xenobiotics. These enzymes are not normally expressed at their maximal levels
`
`and, their expression can be induced by a variety of natural and synthetic agents.
`
`Nuclear factor E2-related factor 2 (Nrf2) is a transcription factor responsible for
`
`the induction of a variety of important antioxidant and detoxification enzymes that
`
`coordinate a protective cellular response to metabolic and toxic stress.
`
`[0006] ROS/RNS are most damaging in the brain and neuronal tissue,
`
`where they attack post-mitotic (Le., non-dividing) cells such as glial cells,
`
`oligodendocytes, and neurons, which are particularly sensitive to free radicals.
`
`This process leads to neuronal damage. Oxidative stress has been implicated in
`
`the pathogenesis of a variety of neurodegenerative diseases, including ALS,
`
`Alzheimer's disease (AD), and Parkinson's disease (PO). For review, see, e.g.,
`
`van Muiswinkel et aL, Curr. Drug Targets CNS--Neurol. Disord., 2005, 4:267-281.
`
`An anti-oxidative enzyme under control of Nrf2, N001 (NAD(P)H
`
`dehydrogenase, quinone 1), was recently reported to be substantially
`
`upregulated in the brain tissues of AD and PO subjects (Muiswinkel et aL,
`
`Neurobiol. Aging, 2004, 25: 1253). Similarly, increased expression of N001 was
`
`reported in the ALS subjects' spinal cord (Muiswinkel et aI., Curro 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 BioI. & Med., 2006, 41 311-311). These observations indicate that the
`
`Nrf2 pathway may be activated in neurodegenerative and neuroinflammatory
`
`diseases as an endogenous protective mechanism. Indeed, most recently, it has
`
`2
`
`Page 6 of 49
`
`

`
`Attorney Docket No. 08201.6042-00000
`
`been reported that induced activation of Nrf2-dependent genes by certain
`
`cyclopenanone-based compounds (NEPP) counters the toxic effects of metabolic
`
`inhibition and ROS/RNS production in the brain and protects neurons from death
`
`in vitro and in vivo (see Satoh et aI., PNAS, 2006, 103(3):768-773).
`
`[0007] Additionally, many publications have reported neuroprotective
`
`effects of compounds in natural plant-derived compounds ("phytochemicals"),
`
`including a-tocopherol (vitamin E), Iycopene (tomatoes), resveratrol (red grapes),
`
`sulforaphane (broccoli), EGCG (green tea), etc. For review, see Mattson and
`
`Cheng, Trends in Neurosci., 2006, 29(11 ):632-639. Originally, the action of
`
`these compounds was attributed to their anti-oxidant properties. However, while
`
`most anti-oxidants are effective only at high concentrations, at least some of
`
`these compounds appear to exert neuroprotective effects at much lower doses.
`
`Emerging evidence suggests that these compounds may exert their
`
`neuroprotective effects by activating cellular stress-response pathways, including
`
`the Nrf2 pathway, resulting in the upregulation of neuroprotective genes.
`
`However, the exact mechanism of action of these compounds remains poorly
`
`understood.
`
`[0008] To date, more than 10 different chemical classes of inducers of
`
`Nrf2 pathway have been identified including isothiocyanates and their thiol
`
`addition products, dithiocarbamates, as well as 1 ,2-dithiole-3-thiones, trivalent
`
`arsenic derivatives (e.g., phenyl arsenoxide), heavy metals, certain conjugated
`
`cyclic and acyclic polyenes (including porphyrins, chlorophyllins, and chlorophyll),
`
`and vicinal dimercaptans. These inducers have few structural similarities. They
`
`are mostly electrophiles, and all can react chemically with thiol groups by
`
`alkylation, oxidation, or reduction, suggesting that the intracellular sensor for
`
`inducers is likely to contain very highly reactive (cysteine) thiols. The inducers
`
`can modify thiol groups by a variety of mechanisms including: alkylation (Michael
`
`addition acceptors, isothiocyanates, quinones); oxidation (e.g., peroxides and
`
`hydroperoxides); and direct reaction with thiol/disulfide linkages (e.g., vicinal
`
`dithiols such as 1,2-dimercaptopropanol, lipoic acid). These diverse response
`
`3
`
`Page 7 of 49
`
`

`
`Attorney Docket No. 08201.6042-00000
`
`mechanisms provide plasticity for cellular responses to a variety of electrophilic
`
`and oxidant stressors.
`
`[0009] There is a need to develop new treatments, and in particular,
`
`compounds for treating MS that provide neuroprotection. The development of
`
`cell-based and in vitro assays to identify and characterize existing drug
`
`candidates serves this goal.
`
`[0010]
`
`In some embodiments, the invention provides the following
`
`methods:
`
`1) methods of screening for new candidate compounds that may be
`
`useful for treating a neurological disease;
`
`2) methods of evaluating neuroprotective properties of drugs and drug
`
`candidates for treating a neurological disease;
`
`3) methods of comparing (e.g., for bioequivalence) two or more
`pharmaceutical compositions which contain fumaric acid derivatives;
`
`4) methods of treating a neurological disease by administering to the
`
`subject in need thereof compounds that are partially structurally
`
`similar to DMF or MMF; and
`
`5) methods of treating a neurological disorder by a combination therapy
`
`that includes administration of a first compound that upregulates the
`
`Nrf2 pathway and a second compound that does not upregulate the
`
`Nrf2 pathway.
`
`[0011] A neurological disease in methods 1-5 above is preferrably a
`
`neurodegenerative disease such as, for example, ALS, Parkinson's disease,
`
`Alzheimer's disease, and Huntington's disease. More preferably, the
`
`neurological disease is MS or another demyelinating neurological disease.
`
`[0012] Methods 1-3 of the invention may comprise:
`
`a) contacting a cell with the test compound, and
`
`b) determining whether the Nrf2 pathway is upregulated in the cell.
`
`In some embodiments, the methods may further comprise:
`
`4
`
`Page 8 of 49
`
`

`
`Attorney Docket No. 08201.6042-00000
`
`c) determining whether the test compound slows or prevents
`
`demyelination, axonal loss, and/or neuronal death, and/or
`
`d) selecting the test compound as a candidate for treating
`
`neurodegeneration in a neurological disease if 1) the Nrf2 pathway is
`
`upregulated and 2) demyelination, axonal loss, and/or neuronal death
`
`are/is prevented or slowed.
`
`[0013] Methods 1-3 of the invention comprise contacting a cell with test
`
`compound(s) and determining whether the Nrf2 pathway is upregulated in the
`
`cell. In such methods, an upregulation of the Nrf2 pathway above a threshold
`
`(e.g., by at least 30% over a control) indicates that the compound(s) has/have
`
`certain biological properties beneficial in treating a neurological disease (e.g.,
`
`neuroprotective properties). In some embodiments, the upregulation of the Nrf2
`
`pathway is assessed (in vivo and/or in vitro) by one or more of the following:
`
`i) expression levels of endogenously produced or exogenously
`
`introduced Nrf2;
`
`ii) subcellular localization and/or nuclear translocation of Nrf2;
`
`iii) expression levels and/or activity of one or more genes under control
`
`of Nrf2 (e.g., endogenous NQ01) or an Nrf2-regulated reporter gene
`
`in an artificial reporter construct;
`
`iv)
`
`levels of Nrf2 binding to the Nrf2-binding DNA element ARE;
`
`v) stability of Nrf2/Keap1 complexes; and
`
`vi) modification (e.g., alkylation) levels of Keap1 and other
`
`Nrf2/Keap1-associated proteins.
`
`[0014]
`
`In some embodiments of methods 1-3, the compounds that are
`
`being screened, evaluated, or compared are mild alkylating agents, and more
`
`specifically, Michael addition acceptors, or compounds that are metabolized upon
`
`administration to Michael addition acceptors. In some embodiments, such
`
`Michael addition acceptors have the structure of Formula I, II, III, or IV set forth in
`
`the Detailed Description.
`
`5
`
`Page 9 of 49
`
`

`
`Attorney Docket No. 08201.6042-00000
`
`[0015]
`
`In certain embodiments of method 1, the method of screening for
`
`a candidate compound for treating a neurological disease comprises:
`
`a) contacting a cell with a plurality of test compounds,
`
`b) determining whether the Nrf2 pathway is upregulated in the cell, and
`
`c) selecting from the plurality of compounds at least one compound that
`
`upregulates the Nrf2 pathway,
`
`wherein an upregulation of the Nrf2 pathway by the selected compound(s)
`
`indicates that the selected compound(s) may be useful for treating a neurological
`
`disease. The plurality of compounds may be represented, e.g., by a
`
`combinatorial chemical library, and the method may be performed, e.g., by
`
`high-throughput screening.
`
`[0016]
`
`In certain embodiments of method 2, the method of evaluating
`
`neuroprotective properties of a drug or drug candidate for treating a neurological
`
`disease comprises:
`
`a) contacting a cell with the drug or drug candidate, and
`
`b) determining whether the Nrf2 pathway is upregulated in the cell,
`
`wherein an upregulation of the Nrf2 pathway by the drug or drug candidate
`
`indicates that the drug or drug candidate is useful for neuroprotection in treating
`
`a human having a neurological disease.
`
`[0017]
`
`In certain embodiments of method 3, a method of comparing two
`
`or more pharmaceutical compositions (e.g., for bioequivalence) comprises:
`
`a) contacting a cell with a first composition comprising a test compound,
`
`and
`
`b) comparing the level of Nrf2 pathway upregulation in the cell by the
`
`test compound to the corresponding level of the Nrf2 pathway
`
`upregulation in a control cell treated with a second composition
`
`comprising DMF, MMF, or both.
`
`[0018]
`
`In some embodiments of method 3, the test compound is fumaric
`
`acid, a salt thereof, or a fumaric acid derivative. In some embodiments, the first
`
`composition comprises DMF, MMF, or both. In some embodiments, the dose
`
`6
`
`Page 10 of 49
`
`

`
`Attorney Docket No. 08201.6042-00000
`
`and/or the formulation of the first composition differs from the dose and/or the
`
`formulation of the second composition.
`
`[0019]
`
`In some embodiments, method 3 further comprises:
`
`c) comparing at least one pharmacokinetic parameter (e.g.,
`
`serum-half-life) of the first and the second compositions.
`
`[0020]
`
`In some embodiments of method 4, the method of treating a
`
`mammal having a neurological disease comprises administering to the mammal
`
`a therapeutically effective amount of a neuroprotective compound having
`
`Formula I, II, III, or IV, e.g., a fumaric acid derivative (e.g., DMF or MMF).
`
`[0021]
`
`In some embodiments of method 4, the invention provides a
`
`method of slowing or preventing neurodegeneration (more specifically, e.g.,
`
`demyelination, axonal loss, and/or neuronal death) in a patient in need thereof,
`
`by administering the compound in an amount and for a period of time sufficient to
`
`slow or prevent demyelination, axonal loss, and/or neuronal death, e.g., by at
`
`least 30% relative to a control.
`
`[0022]
`
`In certain embodiments of method 5, the method of treating a
`
`mammal having a neurological disease by combination therapy comprises:
`
`a) administering to the mammal a therapeutically effective amount of a
`
`first compound that upregulates the Nrf2 pathway, and
`
`b) administering a therapeutically effective amount of a second
`
`compound that does not upregulate the Nrf2 pathway.
`
`[0023]
`
`In some of embodiments of method 5, the first compound, used in
`
`step (a), is a compound of Formula I, II, III, or IV, e.g., a fumaric acid derivative
`
`(e.g., DMF or MMF); and the second compound, which is used in step (b), is an
`
`immunosuppressive or an immunomodulatory compound that does not
`
`upregulate the Nrf2 pathway (e.g., by more than 30% over a control).
`
`[0024]
`
`In some embodiments of method 5, the method of treating a
`
`neurological disease in a mammal comprises administering to the mammal a
`
`therapeutically effective amount of a compound of Formula I, II, III, or IV.
`
`7
`
`Page 11 of 49
`
`

`
`Attorney Docket No. 08201.6042-00000
`
`[0025]
`
`In some of the embodiments of methods 1-5, the compound being
`
`screened, identified, evaluated, or used for treating a neurological disorder is not
`
`fumaric acid or its salt, or a fumaric acid derivative (e.g., DMF or MMF).
`
`[0026] Other features and embodiments of the invention will be apparent
`
`from the following description and the claims.
`
`BRIEF DESCRIPTION OF THE FIGURES
`[0027] Figure 1 demonstrates that DMF and MMF are potent activators
`
`of Nrf2 at concentrations within clinical exposure range (cells in culture).
`
`[0028] Figure 2 shows results of RNAi experiments.
`
`DETAILED DESCRIPTION
`
`[0029] The present invention is based, in part, on the discovery that
`
`dimethyl fumarate (DMF) and monomethyl fumarate (MMF) are potent activators
`
`of the Nrf2 pathway, a major neuroprotective and anti-inflammatory mechanism.
`
`The invention is further based, at least in part, on the finding that DMF and MMF
`
`are neuroprotective (myelinoprotective and axonoprotective) in a mouse model of
`
`autoimmune neurodegenerative disease.
`
`[0030] Due to the involvement of Nrf2 in the regulation of cellular
`
`response to metabolic stress, survival and inflammation, DMF, MMF, and other
`
`Nrf2 activators may be useful for therapeutic management of a variety of
`
`inflammatory, ischemiC, and neurodegenerative processes.
`
`[0031] Fumaric acid esters, such as DMF, have been proposed for
`treatment of MS (see, e.g., Schimrigk et aI., Eur. J. Neurol., 2006, 13(6):604-10;
`Drugs R&D, 2005, 6(4):229-30).
`
`[0032] DMF activates a major cytoprotective (neuroprotective) and
`
`anti-inflammatory mechanism not targeted by current therapies. Thus, the
`
`findings that DMF activates the Nrf2 pathway and has a neuroprotective effect,
`
`offer a rationale for the use of DMF in combination with immunosuppressive or
`
`8
`
`Page 12 of 49
`
`

`
`Attorney Docket No. 08201.6042-00000
`
`immunodulatory therapeutics that do not upregulate the Nrf2 pathway. The
`
`invention further provides means for identifying compounds with a new
`
`therapeutic modality useful in multiple neurological indications and
`
`complementary to other drugs for the treatment of a neurological disease,
`
`including a number of currently used immunomodulators.
`
`[0033] DMF is a member of a large group of anti-oxidant molecules
`
`known for their cytoprotective and anti-inflammatory properties. These
`
`molecules also share the property of the Nrf2 pathway activation. Thus, the
`
`finding that DMF activates the Nrf2 pathway in conjunction with the
`
`neuroprotective effects of DMF further offers a rationale for identification of
`
`structurally and/or mechanistically related molecules that would be expected to
`
`be therapeutically effective for the treatment of neurological disorders, such as,
`
`e.g., MS.
`
`I. Definitions
`[0034] Certain terms are defined in this section; additional definitions are
`
`provided throughout the description.
`
`[0035] The terms "activation" and "upregulation," when used in reference
`
`to the Nrf2 pathway, are used interchangeably herein.
`
`[0036] The terms "disease" and "disorder" are used interchangeably
`
`herein.
`
`[0037] The term "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.
`
`[0038] The term "neuroprotection" and its cognates refer to prevention or
`
`a slowing in neuronal degeneration, including, for example, demyelination and/or
`
`axonal loss, and optionally, neuronal and oligodendrocyte death. Neuroprotection
`
`may occur through several mechanisms, e.g., through reducing inflammation,
`
`providing neurotrophic factors, scavenging free radicals, etc. As used herein, a
`
`compound is considered neuroprotective if it (1) upregulates the Nrf2 pathway
`
`9
`
`Page 13 of 49
`
`

`
`Attorney Docket No. 08201.6042-00000
`
`above a certain threshold and (2) provides neuroprotection, regardless of
`
`possible other mechanisms of action.
`
`[0039] The terms "treatment," "therapeutic method," "therapeutic
`
`benefits," and the like refer to therapeutic as well as prophylactic/preventative
`
`measures. Thus, those in need of treatment may include individuals already
`
`having a specified disease and those who are at risk for acquiring that disease.
`
`[0040] The terms "therapeutically 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) or
`
`reduced inflammation of the cells of the eNS.
`
`II.
`
`Methods of Evaluating Compounds
`[0041]
`In one aspect, the invention provides method of evaluating
`neuroprotective properties of test compounds, including the following methods:
`
`1) methods of screening for new candidate compounds that may be
`
`useful for treating a neurological disease;
`
`2) methods of evaluating neuroprotective properties of drugs and
`
`candidates that are used or proposed for treating a neurological
`
`disease;
`
`3) methods of comparing (e.g., for bioequivalence) two or more
`
`pharmaceutical compositions which contain fumaric acid
`
`derivatives;
`
`[0042] Methods 1-3 of the invention may comprise:
`
`a) contacting a cell with the test compound,
`
`b) determining whether the Nrf2 pathway is upregulated in the cell,
`
`and, in some embodiments, additionally performing the following step(s):
`
`c) determining whether the test compound slows or prevents
`
`demyelination, axonal loss, and/or neuronal death, and/or
`
`10
`
`Page 14 of 49
`
`

`
`Attorney Docket No. 08201.6042-00000
`
`d) selecting the test compound as a candidate for treating
`
`neurodegeneration in a neurological disease if 1) the Nrf2 pathway is
`
`upregulated and 2) demyelination, axonal loss, and/or neuronal death
`
`are/is prevented or slowed.
`
`[0043] Methods 1-3 are described in detail below.
`
`Method 1: Methods of screening
`
`[0044] The invention provides methods of screening for a candidate
`
`compound for treating a neurological disease. Such methods comprise:
`
`a) contacting a cell with a plurality of test compounds,
`
`b) determining whether the Nrf2 pathway is upregulated in the cell, and
`
`c) selecting from the plurality of compounds at least one compound that
`
`upregulates the Nrf2 pathway,
`
`wherein an upregulation of the Nrf2 pathway by the selected compound(s)
`
`indicates that the selected compound(s) may be useful for treating a neurological
`
`disease. For example, the plurality of compounds may be represented by a
`
`combinatorial chemical library, and the screening method may be performed by a
`
`high-throughput screening as described in, e.g., High-Throughput Screening in
`
`Drug Discovery (Methods and Principles in Medicinal Chemistry), by Jorg Huser
`
`(ed.), John Wiley & Sons (2006).
`
`[0045] Combinatorial libraries of compounds are also described in, e.g.,
`
`Solid-Supported Combinatorial and Parallel Synthesis of Small-Molecular-Weight
`
`Compound Libraries (Tetrahedron Organic Chemistry) Ian Salusbury (ed.),
`
`Elsevier (1998); Combinatorial Libraries: Synthesis, Screening and Application
`
`Potential (Library Binding), by Riccardo Cortese (ed.), Walter de Gruyter (1995).
`
`The libraries of compounds may be, for example, quinone libraries and other
`
`libraries as described in Mittoo, Comb. Chem. & High Throughput Screen, 2006,
`
`9:421-423.
`
`[0046]
`
`In some embodiments, the compounds that are being screened
`
`and/or selected comprise at least one or a plurality of mild alkylating agents, and
`
`11
`
`Page 15 of 49
`
`

`
`Attorney Docket No. 08201.6042-00000
`
`more particularly, Michael addition acceptors or compounds that are metabolized
`
`to Michael addition acceptors, including compounds of Formulas I, II, III, or IV.
`
`[0047]
`
`In some of the embodiments, the compounds comprise fumaric
`
`acid, its salt(s), and/or fumaric acid derivative(s).
`
`Methods 2: Methods of evaluating drugs and drug candidates
`
`[0048] The invention further provides methods of evaluating
`
`neuroprotective properties of a drug or drug candidate for treating a neurological
`
`disease. Such methods comprise:
`
`a) contacting a cell with the drug or drug candidate, and
`
`b) determining whether the Nrf2 pathway is upregulated in the cell,
`
`wherein the upregulation of the Nrf2 pathway by the drug or drug candidate
`
`indicates that the drug or drug candidate is neuroprotective in treating a human
`
`having a neurological disease.
`
`[0049]
`
`In some embodiments, the upregulation of the Nrf2 pathway by
`
`the drug or drug candidate indicates that the drug or drug candidate has the
`
`ability to slow demyelination, the loss ofaxons, and/or neuronal death.
`
`[0050]
`
`In some embodiments, the method of evaluating a drug or drug
`
`candidate comprise an additional step:
`
`c) evaluating demyelination, loss ofaxons, and/or neuronal death.
`
`[0051]
`
`In some embodiments, steps a) and c) are performed in vivo in at
`
`least one model of a neurological disease, e.g., as described below.
`
`[0052]
`
`In other embodiments, particularly those in which the neurological
`
`disease is multiple sclerosis or another demyelinating disease, the evaluated
`
`drug or drug candidate for a neurological disease is chosen from the following:
`
`FTY720 (2-(4-octylphenethyl)-2-aminopropane-1 ,3-diol; Novartis); anti-IL 12
`
`antibody (e.g., ABT-874; Abbott Laboratories); GSK683699 (GSK/Tanabe);
`
`NeuroVax (Immune Response Corp.; Darlington, Curro Opin. Mol. Ther., 2005,
`
`7(6):598-603); anti-CCR2 antibody (e.g., MLN 1202; Millennium); interferon J3-1a
`
`(e.g., Avonex®; Biogen Idec); anti-a4-integrin antibody (e.g., Tysabri®; Biogen
`
`Idec/Elan); anti-CD20 antibody (e.g., Rituxan® (Biogen Idec/Genentech);
`
`12
`
`Page 16 of 49
`
`

`
`Attorney Docket No. 08201.6042-00000
`
`TV 5010 (Teva); NBI-788 (Neurocrine); MBP8298 (BioMS (see Warren et aI.,
`Eur. J. Neurol., 2006, 13(8):887-95); Mylinax (Oral Cladribine;
`2-chlorodeoxyadenosine; Serono/IVAX); Teriflunomide
`
`((Z)-2-cyano-N-( 4-( trifl uoromethyl)phenyl)-3-hyd roxybut -2-enamide;
`
`Sanofi-Aventis); Temsirolimus (Wyeth); Laquinimod
`
`(5-chloro-N-ethyl-1 ,2-dihydro-4-hydroxy-1-methyl-2-oxo-N-phenylquinoline-3-car
`
`boxamide; Active BiotechlTeva); and interferon tau (Tauferon; Pepgen).
`
`[0053]
`
`In some embodiments, the drug or drug candidate being
`
`evaluated is a mild alkylating agent, and more specifically, a Michael addition
`
`acceptor, or a compound that is metabolized to a Michael addition acceptor,
`
`including compounds of Formulas I, II, III, or IV.
`
`[0054]
`
`In some of the embodiments, the compound is fumaric acid, its
`
`salt, or a fumaric acid derivative.
`
`Method 3: Methods of methods of evaluating neuroprotective properties
`
`[0055] The invention further provides methods of comparing (e.g., for
`
`bioequivalence) two or more pharmaceutical compositions. Such methods
`
`comprise:
`
`a) contacting a cell with a first composition comprising a test compound,
`
`and
`
`b) comparing the level of the Nrf2 pathwayupregulation in the cell by
`
`the test compound to the corresponding level of the Nrf2 pathway
`
`upregulation in a cell treated with a second composition ("comparator
`
`composition") comprising DMF, MMF, or both.
`
`[0056]
`
`In some embodiments, substantially dissimilar levels of
`
`upregulation by the first and second compositions indicate that the compositions
`
`are not bioequivalent.
`
`[0057]
`
`In some embodiments, the test compound is fumaric acid, its salt
`
`thereof, a fumaric acid derivative, or mixtures thereof. In some embodiments, the
`
`first composition comprises DMF, MMF, or both. In some embodiments, the
`
`dose and/or the formulation of the first composition differs from the dose and/or
`
`13
`
`Page 17 of 49
`
`

`
`Attorney Docket No. 08201.6042-00000
`
`the formulation of the second composition. The first composition may be a
`
`controlled release composition such as, e.g., compositions described in
`
`WO 2006/037342.
`
`[0058]
`
`In some embodiments, the method further comprises and
`
`additional step:
`
`c) comparing at least one pharmacokinetic parameter of the first and
`
`the second compositions.
`[0059] Pharmacokinetic parameters and methods for evaluating the
`
`same are well known and are described in, e.g., Pharmacokinetics, Second
`
`Edition (Drugs and the Pharmaceutical Sciences) by Milo Gibaldi et al. (eds.),
`
`Informa Healthcare (1982). Examples of such pharmacokinetic parameters that
`
`can be evaluated include serum half-life, clearance, and volume distribution.
`
`[0060]
`
`In some embodiments, substantially dissimilar pharmacokinetic
`
`parameter(s) of the first and second compositions indicate that the compositions
`
`are not bioequivalent.
`[0061]
`In some embodiments, the test compound being evaluated is a
`
`mild alkylating agent, and more specifically, a Michael addition acceptor, or a
`
`compound that is metabolized to a Michael addition acceptor.
`
`[0062]
`
`In some of the embodiments, the test compound is fumaric acid or
`
`its salt, or a fumariC acid derivative.
`
`III. Methods of Treatment
`
`[0063] The invention provides methods of treating a mammal who has or
`
`is at risk for developing a neurological disease, including the following methods:
`
`4) methods of treating a neurological disease by administering to the
`
`subject in need thereof compounds that are partially structurally
`
`similar to DMF or MMF (including compounds selected using
`
`methods 1-3 described above) ; and
`
`5) methods of treating a neurological disorder by a combination
`
`therapy that includes administration of a first compound that does
`
`14

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket