WO2015189534A1 - Inhibiteurs de vap-1 pour le traitement de la dystrophie musculaire - Google Patents

Inhibiteurs de vap-1 pour le traitement de la dystrophie musculaire Download PDF

Info

Publication number
WO2015189534A1
WO2015189534A1 PCT/GB2014/053677 GB2014053677W WO2015189534A1 WO 2015189534 A1 WO2015189534 A1 WO 2015189534A1 GB 2014053677 W GB2014053677 W GB 2014053677W WO 2015189534 A1 WO2015189534 A1 WO 2015189534A1
Authority
WO
WIPO (PCT)
Prior art keywords
pyridin
chlorophenyl
vap
imidazo
muscle
Prior art date
Application number
PCT/GB2014/053677
Other languages
English (en)
Inventor
Kenneth MULVANY
Martyn Pritchard
Peter Richardson
Original Assignee
Proximagen Limited
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
Priority claimed from GB201410529A external-priority patent/GB201410529D0/en
Priority claimed from GB201421812A external-priority patent/GB201421812D0/en
Application filed by Proximagen Limited filed Critical Proximagen Limited
Publication of WO2015189534A1 publication Critical patent/WO2015189534A1/fr

Links

Classifications

    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid or pantothenic acid
    • A61K31/198Alpha-amino acids, e.g. alanine or edetic acid [EDTA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • A61K31/573Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
    • 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

Definitions

  • This invention relates to the use of a compound which inhibits VAP-1 /SSAO activity for the treatment of muscular dystrophy.
  • the invention also relates to the use of pharmaceutical compositions comprising these compounds for the treatment of muscular dystrophy.
  • the invention also relates to combined preparations, and their use for the treatment of muscular dystrophy.
  • SSAO Semicarbazide-sensitive amine oxidase
  • VAP-1 Vascular Adhesion Protein-1
  • AOC3 Amine Oxidase, Copper Containing 3
  • TPQ cupric ion and protein-derived topa quinone
  • Known substrates for human SSAO include endogenous methylamine and aminoacetone as well as some xenobiotic amines such as benzylamine [Lyles, Int. J. Biochem. Cell Biol. 1996, 28, 259-274; Klinman, Blochim. Biophys. Acta 2003, 1647(1-2), 131 -137; Matyus et al affect Curr. Med. Chem. 2004, 11(10), 1285-1298; O'Sullivan et al., Neurotoxicology 2004, 25(1-2), 303-315].
  • tissue-bound human SSAO is a homodimeric glycoprotein consisting of two 90-100 kDa subunits anchored to the plasma membrane by a single N-terminal membrane spanning domain [Morris et al., J. Biol. Chem. 1997, 272, 9388-9392; Smith et al., J. Exp. Med. 1998, 188, 17-27; Airenne et al., Protein Science 2005, 14, 1964-1974; Jakobsson et al., Acta Crystallogr. D Biol. Crystallogr. 2005, 61 (Pt 11), 1550-1562].
  • SSAO activity has been found in a variety of tissues including vascular and non-vascular smooth muscle tissue, endothelium, and adipose tissue [Lewinsohn, Braz. J. Med. Biol. Res. 1984, 17, 223- 256; Nakos & Gossrau, Folia Histochem. Cytobiol. 1994, 32, 3-10; Yu et al., Biochem. Pharmacol. 1994, 47, 1055-1059; Castillo et al., Neurochem. Int. 1998, 33, 415-423; Lyles & Pino, J. Neural. Transm. Suppl. 1998, 52, 239-250; Jaakkola et al., Am. J. Pathol.
  • SSAO protein is found in blood plasma and this soluble form appears to have similar properties as the tissue-bound form [Yu et al., Biochem. Pharmacol. 1994, 47, 1055-1059; Kurkijarvi et al., J. Immunol. 1998, 161, 1549-1557]. It has recently been shown that circulating human and rodent SSAO originates from the tissue-bound form [Gokturk et al., Am. J. Pathol.
  • SSAO plays a role in both GLUT4-mediated glucose uptake [Enrique-Tarancon et al., J. Biol. Chem. 1998, 273, 8025-8032; Morin et al., J. Pharmacol. Exp. Ther. 2001 , 297, 563-572] and adipocyte differentiation [Fontana et al., Biochem. J. 2001, 356, 769-777; Mercier et al., Biochem. J. 2001 , 358, 335-342].
  • SSAO has been shown to be involved in inflammatory processes where it acts as an adhesion protein for leukocytes [Salmi & Jalkanen, Trends Immunol. 2001, 22, 211 -216; Salmi & Jalkanen, in "Adhesion Molecules: Functions and Inhibition” K. Ley (Ed.), 2007, pp. 237-251], and might also play a role in connective tissue matrix development and maintenance [Langford et al., Cardiovasc. Toxicol. 2002, 2(2), 141-150; Gokturk et al., Am. J. Pathol. 2003, 163(5), 1921-1928]. Moreover, a link between SSAO and angiogenesis has recently been discovered [Noda et al., FASEB J. 2008, 22(8), 2928-2935].
  • SSAO activity in blood plasma is elevated in conditions such as congestive heart failure, diabetes mellitus, Alzheimer's disease, and inflammation [Lewinsohn, Braz. J. Med. Biol. Res. 1984, 17, 223-256; Boomsma et al., Cardiovasc. Res. 1997, 33, 387-391 ; Ekblom, Pharmacol. Res. 1998, 37, 87-92; Kurkijarvi et al., J. Immunol. 1998, 161, 1549- 1557; Boomsma et al., Diabetologia 1999, 42, 233-237; Meszaros et al., Eur. J. Drug Metab. Pharmacokinet.
  • SSAO knockout animals are phenotypically overtly normal but exhibit a marked decrease in the inflammatory responses evoked in response to various inflammatory stimuli [Stolen et al., Immunity 2005, 22(1), 105-1 15].
  • antagonism of its function in wild type animals in multiple animal models of human disease e.g.
  • carrageenan-induced paw inflammation, oxazolone-induced colitis, lipopolysaccharide-induced lung inflammation, collagen-induced arthritis, endotoxin-induced uveitis) by the use of antibodies and/or small molecules has been shown to be protective in decreasing the leukocyte infiltration, reducing the severity of the disease phenotype and reducing levels of inflammatory cytokines and chemokines [Kirton et al., Eur. J. Immunol. 2005, 35(11), 3119-3130; Salter-Cid et al., J. Pharmacol. Exp. Ther.
  • Fibrosis can result from chronic tissue inflammation when the resolution of the inflammation is partly abrogated by the chronic nature of the inflammatory stimulus.
  • the result can be inappropriate repair of the tissue with excessive extracellular matrix deposition (including collagen) with tissue scarring.
  • myofibroblast activation by stimuli including fibronectin and reactive oxygen species as well as growth factors such as transforming growth factor- ⁇ - ⁇ (TGFB-1 ), insulin-like growth factor-l (IGF-I), platelet-derived growth factor (PDGF) and connective tissue growth factor (CTGF) resulting in increased production of collagen, elastin, hyaluronan, glycoproteins and proteoglycans.
  • IGF-I insulin-like growth factor-l
  • PDGF platelet-derived growth factor
  • CTGF connective tissue growth factor
  • the activity of invading macrophages plays a crucial part In regulating the repair and fibrotic processes.
  • VAP-1 has also been implicated in the progression and maintenance of fibrotic diseases especially in the liver.
  • Weston and Adams J Neural Transm. 2011, 118(7), 1055-614 have summarised the experimental data implicating VAP-1 in liver fibrosis.
  • Weston et al (EASL Poster 2010) showed highly increased expression of VAP-1 in human fibrotic liver, particularly associated with the activated myofibroblasts and collagen fibrils. This anatomical association with fibrosis was consistent with the observation that blockade of VAP-1 accelerated the resolution of carbon tetrachloride induced fibrosis, and suggested a role for the VAP-1 /SSAO enzyme product H202 in the activation of the myofibroblasts.
  • the same authors also showed that the pro-fibrotic growth factor TGF increased the expression of VAP-1 in liver cells by approximately 50-fold.
  • the muscle tissue then suffers from repeated cycles of cell death and aberrant repair, resulting In fibrosis and the replacement of muscle tissue by fat tissue (Mann et al., 2011 , Skeletal Muscle. 1(1):2 Klinger et al. 2012 Acta Myol. 31(3): 184-189).
  • the symptoms of these diseases include pain and muscle weakness.
  • Other dystrophies arising from similar causes include limb girdle muscular dystrophy, congenital muscular dystrophy and distal muscular dystrophy. All of these appear to have defects in cell attachment to the extracellular matrix. Fibrosis is therefore a major issue in the muscular dystrophies and a therapeutic capable of reducing or reversing the fibrosis would be extremely beneficial to patients suffering from muscular dystrophy.
  • the invention described herein relates to the expression of VAP-1 in dystrophic muscle, which VAP-1 expression is expected to increase during the fibrotic disease process.
  • VAP-1 expression is low, and largely restricted to the blood vessels (Salmi et al., 1993, J. Exp. Med. 178, 2255-2260) but increases in inflamed and fibrotic tissues. This increase in expression in the diseased state makes VAP-1 a promising therapeutic target in dystrophic muscle.
  • VAP-1/SSAO Inhibition of VAP-1/SSAO is expected to reduce the concentration of pro-inflammatory and pro-fibrotic enzyme products (such as aldehydes, hydrogen peroxide and ammonia) whilst also decreasing the adhesive capacity of immune and myofibroblast cells and correspondingly their activation and invasion of the muscle tissue.
  • pro-inflammatory and pro-fibrotic enzyme products such as aldehydes, hydrogen peroxide and ammonia
  • inhibition of VAP-1 /SSAO is expected to be therapeutically beneficial in the treatment of muscle fibrosis and therefore muscular dystrophy.
  • VAP-1/SSAO inhibitors are known to reduce leukocyte and monocyte incursion into tissues. It is known from the mdx mouse model, a murine model of Duchenne Muscular Dystrophy, that partial inhibition of macrophage incursion into the muscle tissue has a beneficial effect on muscle tissue maintenance. Therefore VAP-1/SSAO inhibitors are expected to have therapeutic effects in dystrophic muscle by reducing leukocyte, and particularly monocyte, incursion into the tissue.
  • VAP-1/SSAO inhibitors will reduce inflammation and muscle loss through inhibition of leukocyte invasion, and reduce muscular fibrosis and scarring through reduced VAP-1 activity in the muscle tissue, and reduce inflammatory and fibrotic cell activation in muscle tissue through reduced production of pro-inflammatory and pro-fibrotic enzyme products such as aldehydes, hydrogen peroxide and ammonia.
  • VAP-1 inhibitor for use in the treatment of muscular dystrophy.
  • the invention provides the use of a VAP-1 inhibitor in the manufacture of a medicament for treatment of muscular dystrophy.
  • the invention provides a method of treating muscular dystrophy comprising administering to a subject suffering such disease an effective amount of a VAP-1 inhibitor.
  • VAP-1 inhibitors especially the VAP-1 inhibitor carbidopa
  • a steroid for example a glucocorticoid, such as prednisolone.
  • a combined preparation which comprises: (a) a VAP-1 inhibitor compound; and (b) a steroid.
  • a combined preparation of the invention may be provided as a pharmaceutical combination for administration to a mammal, preferably a human.
  • a pharmaceutical composition which comprises: (a) a VAP-1 inhibitor compound; and (b) a steroid; and (c) a pharmaceutically acceptable carrier, excipient, or diluent.
  • the VAP-1 inhibitor may optionally be provided together with a pharmaceutically acceptable carrier, excipient, or diluent, and/or the steroid may be provided together with a pharmaceutically acceptable carrier, excipient, or diluent.
  • the invention provides a combined preparation of the invention, or a pharmaceutical composition of the invention, for use in the treatment of muscular dystrophy.
  • the invention provides use of a combined preparation of the invention, or a pharmaceutical composition of the invention, in the manufacture of a medicament for the treatment of muscular dystrophy.
  • the invention provides a method of treating muscular dystrophy comprising administering to a subject suffering such disease an effective amount of a VAP-1 inhibitor compound and a steroid.
  • the steroid may be a glucocorticoid, such as prednisolone, or a pharmaceutically acceptable salt thereof, or prednisone, or a pharmaceutically acceptable salt thereof.
  • treatment refers to obtaining a desired pharmacologic and/or physiologic effect.
  • the effect can be prophylactic in terms of completely or partially preventing muscular dystrophy or a symptom thereof and/or can be therapeutic in terms of a partial or complete cure for muscular dystrophy and/or an adverse effect attributable to the disease.
  • Treatment covers any treatment of muscular dystrophy in a mammal, particularly in a human, and includes: (a) preventing the disease from occurring in a subject which can be predisposed to the disease but has not yet been diagnosed as having it; (b) inhibiting the disease, i.e., arresting its development; and (c) relieving the disease, i.e., causing regression of the disease.
  • an “effective amount” of a VAP-1 inhibitor refers to the amount of a VAP-1 inhibitor that, when administered to a mammal or other subject for treating muscular dystrophy, is sufficient to effect such treatment for the disease.
  • the “effective amount” will vary depending on the VAP-1 inhibitor, the disease and its severity and the age, weight, etc., of the subject to be treated.
  • VAP-1 inhibitor or "VAP-1 inhibitor compound” includes both non-biological small molecule inhibitors of VAP-1 and biological inhibitors of VAP-1 , including but not limited to RNA, antibodies, polypeptidic or proteinaceous inhibitors of VAP-1.
  • VAP-1 inhibitor or “VAP-1 inhibitor compound” is one which has an IC50 value of less than 1000nM in the VAP-1 Assay described below.
  • VAP-1 inhibitors Small molecules of different structural classes have previously been disclosed as VAP-1 inhibitors, for example in WO 02/38153 (tetrahydroimidazo[4,5-c]pyridine derivatives), in WO 03/006003 (2- indanylhydrazine derivatives), in WO 2005/014530 (allylhydrazine and hydroxylamine (aminooxy) compounds) and in WO 2007/120528 (allylamino compounds), WO2011034078 (N-[3-(heterocyclyl or phenyl)benzyl]-2-aminoglycinamides), and WO2012120195 (Pyridazinones), and WO2012124696 (Guanidines), and Bioorganic & Medicinal Chemistry (2013), 21 (13), 3873-3881 (1 H-imidazol-2-amine derivatives), and Bioorganic & Medicinal Chemistry (2013), 21(5), 1219-1233 (Thiazoles).
  • VAP-1 inhibitors are known, for example, haloallyl amines of WO2009066152; imidazopyridines of WO2010064020; dihydralazine (WO2010015870); pyrazolo[4,3- cjpyridines of WO2010031791 ; 4,5,6,7-tetrahydroimidazo[4,5-c]pyridines of US2002198189, WO0238153 and WO2010031789; oximes of WO2010029379; allyl hydrazine, hydroxylamine and other compounds of US2005096360, WO2006094201 and WO2005014530; amine, amide and allylamino compounds of WO2007120528, US2007078157, WO2005082343 and WO2009055002; hydroxamic acids of WO2006013209; vitamin B1 , vitamin B1 derivatives and vitamin B1 precursors of WO2008025870; 2,3,4,6,8-
  • VAP-1 Biological inhibitors of VAP-1 include but are not limited to antibodies to VAP-1 , RNAi, siRNA (examples of siRNAs suitable for targeting VAP-1 are described, for example, in WO2006134203), anti-sense oligonucleotides, anti-sense peptidyl nucleic acids, and aptamers.
  • VAP-1 antibodies include but are not limited to anti-VAP-1 neutralizing antibody (available, for example, from R&D systems, Minneapolis, MN, catalogue numbers.
  • VAP-1 inhibitors disclosed specifically or generically in the above publications are expected to have utility in the treatment of muscular dystrophy according to the present invention.
  • VAP-1 inhibitor compounds suitable for use in the present invention are provided below. Any pharmaceutically acceptable salt form of the Examples is suitable for use in the present invention.
  • Specific examples of inhibitors of VAP-1 include the compounds speficially disclosed as Examples in WO 2010/031789, namely: 2,2,2-Trichloroethyl 4-isopropyl-1 ,4,6,7- Pyridin-3-ylmethyl 4-isopropyl-1 ,4,6,7- tetrahydro-5H-imidazo[4,5-c]pyridine-5- tetrahydro-5H-imidazo[4,5-c]pyridine-5- carboxylat carboxyla
  • inhibitors of VAP-1 include the following, which are Examples from WO2011/113798:
  • VAP-1 compounds include the Examples of WO2013/037411 , namely:
  • VAP-1 compounds include the Examples of WO2013/038189, namely:
  • inhibitors of VAP-1 include the compounds speficially disclosed as Examples in WO 2010/031791 , namely: 3-(4-Fluorophenyl)-1-(tetrahydro-2H-pyran-4- 3-(4-Chlorophenyl)-1-piperidin
  • inhibitors of VAP-1 include the compounds speficlally disclosed as Examples in WO 2010/064020, namely:
  • VAP-1 compounds include; ferf-Butyl N-(3- ⁇ 4-[1 -(4-chlorophenyl)-1 H- 2-Amino-1 - ⁇ 4-[1 -(4-chlorophenyl)-1 H- pyrrolo[2,3-c]pyridin-3-yl]piperidin-1 -yl ⁇ -3- pyrrolo[2,3-c]pyridin-3-yl]piperidin-1-yl ⁇ ethan-1- oxopropyl)carbamate one
  • VAP-1 inhibitor compounds suitable for use in the present invention are provided below. Any pharmaceutically acceptable salt form of the Examples is suitable for use in the present invention.
  • Specific examples of inhibitors of VAP-1 include: the substituted 3-haloallylamine inhibitors specifically disclosed as Examples in WO 2013/163675, in particular compounds 1-39 in Table 1 of that document; the IMIDAZO[4,5-C]PYRIDINE AND PYRROLO[2,3-C]PYRIDINE DERIVATIVES specifically disclosed as Examples in WO 2014/140592, namely:
  • VAP-1 inhibitor suitable for use in the present invention is selected from the group consisiting of:
  • Racemic Carbidopa is useful in the present invention.
  • the Carbidopa for use in the invention is the (R) enantiomer or the (S) enantiomer.
  • Benserazide is preferred for use in the present invention.
  • the Benserazide for use in the present invention is the (R) enantiomer or the (S) enantiomer.
  • benserazide or a pharmaceutically acceptable salt thereof, for use in the treatment of muscular dystrophy, particularly Duchenne muscular dystrophy, in a human subject.
  • the VAP-1 inhibitor compounds of the invention are formulated into pharmaceutical formulations for various modes of administration. It will be appreciated that compounds may be administered together with a physiologically acceptable carrier, excipient, or diluent.
  • the pharmaceutical compositions of the invention may be administered by any suitable route, preferably by oral, rectal, nasal, topical (including buccal and sublingual), sublingual, transdermal, intrathecal, transmucosal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration.
  • Formulations may conveniently be presented in unit dosage form, e.g., tablets and sustained release capsules, and in liposomes, and may be prepared by any method known in the art of pharmacy.
  • Pharmaceutical formulations are usually prepared by mixing the active substance, or a pharmaceutically acceptable salt thereof, with conventional pharmaceutically acceptable carriers, diluents or excipients.
  • excipients are water, gelatin, gum arabicum, lactose, microcrystalline cellulose, starch, sodium starch glycolate, calcium hydrogen phosphate, magnesium stearate, talcum, colloidal silicon dioxide, and the like.
  • Such formulations may also contain other pharmacologically active agents, and conventional additives, such as stabilizers, wetting agents, emulsifiers, flavouring agents, buffers, and the like.
  • the amount of active compounds is between 0.1-95% by weight of the preparation, preferably between 0.2-20% by weight in preparations for parenteral use and more preferably between 1-50% by weight in preparations for oral administration.
  • the formulations can be further prepared by known methods such as granulation, compression, microencapsulation, spray coating, etc.
  • the formulations may be prepared by conventional methods in the dosage form of tablets, capsules, granules, powders, syrups, suspensions, suppositories or injections.
  • Liquid formulations may be prepared by dissolving or suspending the active substance in water or other suitable vehicles. Tablets and granules may be coated in a conventional manner. To maintain therapeutically effective plasma concentrations for extended periods of time, compounds of the invention may be incorporated Into slow release formulations.
  • the dose level and frequency of dosage of the specific compound will vary depending on a variety of factors including the potency of the specific compound employed, the metabolic stability and length of action of that compound, the patient's age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the condition to be treated, and the patient undergoing therapy.
  • the daily dosage may, for example, range from about 0.001 mg to about 100 mg per kilo of body weight, administered singly or multiply in doses, e.g. from about 0.01 mg to about 25 mg each. Such a dosage may be given orally or parenterally.
  • Multiple doses may be administered over a period of time, such as at least a week, a month, several months, a year, or several years, or throughout the course of the condition.
  • the frequency of dosage may be at least once per month, once per week, or once per day.
  • the components of a combined preparation of the invention may be for simultaneous, separate, or sequential use.
  • combined preparation refers to a "kit of parts" in the sense that the combination components (a) and (b) can be dosed independently or by use of different fixed combinations with distinguished amounts of the combination components (a) and (b).
  • the components can be administered simultaneously or one after the other. If the components are administered one after the other, preferably the time interval between administration is chosen such that the effect on the treated disorder or disease in the combined use of the components is greater than the effect which would be obtained by use of only any one of the combination components (a) and (b).
  • the components of the combined preparation may be present in one combined unit dosage form, or as a first unit dosage form of component (a) and a separate, second unit dosage form of component (b).
  • the ratio of the total amounts of the combination component (a) to the combination component (b) to be administered in the combined preparation can be varied, for example in order to cope with the needs of a patient sub-population to be treated, or the needs of the single patient, which can be due, for example, to the particular disease, age, sex, or body weight of the patients.
  • there is at least one beneficial effect for example an enhancing of the effect of the VAP-1 inhibitor, or a mutual enhancing of the effect of the combination components (a) and (b), for example a more than additive effect, additional advantageous effects, fewer side effects, less toxicity, or a combined therapeutic effect compared with a non-effective dosage of one or both of the combination components (a) and (b), and very preferably a synergism of the combination components (a) and (b).
  • beneficial effect for example an enhancing of the effect of the VAP-1 inhibitor, or a mutual enhancing of the effect of the combination components (a) and (b), for example a more than additive effect, additional advantageous effects, fewer side effects, less toxicity, or a combined therapeutic effect compared with a non-effective dosage of one or both of the combination components (a) and (b), and very preferably a synergism of the combination components (a) and (b).
  • the VAP-1 inhibitor and the steroid may be administered sequentially to the subject, i.e. the VAP-1 inhibitor may be administered before, with, or after the steroid.
  • the VAP-1 inhibitor and the steroid may be administered to the subject within 96 hours, 72 hours, 48 hours, 24 hours, or 12 hours, of each other.
  • the VAP-1 inhibitor and the steroid may be co-administered to the subject, for example as a composition comprising the VAP-1 inhibitor and the steroid, or by simultaneous administration of separate doses of the VAP-1 inhibitor and the steroid.
  • a plurality of doses of the VAP-1 inhibitor, and/or a plurality of doses of the steroid is administered to the subject.
  • a dose of the VAP-1 inhibitor is administered before, with, or after each administration of two or more doses of the steroid.
  • a dose of VAP-1 inhibitor may be administered within 96 hours, 72 hours, 48 hours, 24 hours, or 12 hours, of each administration of two or more doses of the steroid.
  • the choice of appropriate dosages of the components used in combination therapy according to the present invention can be determined and optimized by the skilled person, for example, by observation of the patient, including the patient's overall health, and the response to the combination therapy. Optimization, for example, may be necessary if it is determined that a patient is not exhibiting the desired therapeutic effect or conversely, if the patient is experiencing undesirable or adverse side effects that are too many in number or are of a troublesome severity.
  • an effective amount of the combination therapy is an amount that results in a reduction of at least one pathological parameter associated with muscular dystrophy.
  • an effective amount of the combination therapy is an amount that is effective to achieve a reduction of at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90%, in the parameter, compared to the expected reduction in the parameter associated with the muscular dystrophy without the combination therapy.
  • the parameter may be: muscle pathology; muscle degeneration; muscle necrosis; inflammation of muscle; infiltration of inflammatory cells into muscle, especially inflammatory myeloid cells such as monocytes, macrophages, neutrophils, or eosinophils; infiltration of lymphoid cells into muscle, especially T helper lymphocytes or double negative T cells; inflammatory cell activation in muscle; muscle fibrosis; or fibrotic cell activation in muscle.
  • muscle pathology muscle degeneration
  • muscle necrosis inflammation of muscle
  • infiltration of inflammatory cells into muscle especially inflammatory myeloid cells such as monocytes, macrophages, neutrophils, or eosinophils
  • lymphoid cells especially T helper lymphocytes or double negative T cells
  • inflammatory cell activation in muscle muscle fibrosis
  • muscle fibrosis or fibrotic cell activation in muscle.
  • combination treatment may be employed to increase the therapeutic effect of the VAP-1 inhibitor or steroid, compared with the effect of the VAP-1 Inhibitor or steroid as a monotherapy, or to decrease the doses of the individual components in the resulting combinations while preventing or further reducing the risk of unwanted or harmful side effects of the individual components.
  • a typically prescribed dose range for a steroid as a monotherapy is 0.3-1 mg/kg/day (suitably 0.7 or 0.75mg/kg/day), or 0.3mg/kg/day to 10mg/kg/week, in humans.
  • a typically prescribed dose range for a VAP-1 inhibitor as a monotherapy in humans is 20-200mg/day for carbidopa (suitably 30mg/day or 75 mg/day), and 25-300mg/day (suitably 25mg/day or 50mg/day) for benserazide.
  • the VAP-1 inhibitor and the steroid are each prescribed at a dose that is within a typically prescribed dose range for each compound as a monotherapy.
  • the compounds may be prescribed as separate dosages or as a combination dosage. Such combinations provide increased efficacy compared with the effect of either compound as a monotherapy.
  • the VAP-1 inhibitor and the steroid are each prescribed at a dose that is below a typically prescribed dose for each component as a monotherapy, but at doses that have therapeutic efficacy in combination.
  • the components may be prescribed as separate dosages or as a combination dosage.
  • the dosages of the components in combination may be selected to provide a similar level of therapeutic efficacy as the VAP-1 inhibitor or the steroid as a monotherapy, but with the advantage that the lower doses of the VAP-1 inhibitor and/or the steroid reduce the risk of adverse side effects compared to the prescribed dosages of each compound as a monotherapy.
  • the prescribed dosage of the VAP-1 inhibitor is within a typically prescribed dose range for monotherapy, and the steroid is prescribed at a dosage that is below a typically prescribed dose for monotherapy.
  • the prescribed dosage of the VAP-1 inhibitor is below a typically prescribed dose for monotherapy, and the steroid is prescribed at a dosage that is within a typically prescribed dose range for monotherapy.
  • Preferred dosages below the typically prescribed dose for monotherapy are doses that are up to 50%, or up to 25%, of the typically prescribed dose.
  • the VAP-1 inhibitor and the steroid may be administered substantially simultaneously (for example, within about 60 minutes, about 50 minutes, about 40 minutes, about 30 minutes, about 20 minutes, about 10 minutes, about 5 minutes, or about 1 minute of each other) or separated in time by about 1 hour, about 2 hours, about 4 hours, about 6 hours, about 10 hours, about 12 hours, about 24 hours, about 36 hours, about 72 hours, or about 96 hours, or more.
  • the skilled person will be able to determine, and optimise, a suitable time course for sequential administration, depending on the particular combination of the VAP-1 inhibitor and the steroid.
  • the time course is preferably selected such that there is at least one beneficial effect, for example an enhancing of the effect of the VAP-1 inhibitor or the steroid, or a mutual enhancing of the effect of the combination components, for example a more than additive effect, additional advantageous effects, fewer side effects, less toxicity, or a combined therapeutic effect compared with a non-effective dosage of one or both of the combination components, and very preferably a synergism of the combination components.
  • the optimum time course will depend on factors such as the time taken for the peak plasma concentration of the compound to be reached after administration, and the elimination half-life of each compound.
  • the time difference is less than the half-life of the first component to be administered.
  • the VAP-1 inhibitor may be administered in the morning, and the steroid administered at least once later in the day. In other embodiments, the VAP-1 inhibitor and the steroid may be administered at substantially the same time.
  • the subject may receive doses of the VAP-1 inhibitor and the steroid over a period of weeks, months, or years. For example, 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year, 2 years, 3 years, 4 years, 5 years, or more.
  • the components of a combination of the invention may be administered by known means, in any suitable formulation, by any suitable route.
  • Suitable routes of administration may include by oral, rectal, nasal, topical (including buccal and sublingual), sublingual, transdermal, intrathecal, transmucosal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration.
  • the VAP-1 inhibitor and the steroid are administered orally.
  • Suitable pharmaceutical compositions and dosage forms may be prepared using conventional methods known to those in the field of pharmaceutical formulation and described in the relevant texts and literature, for example, in Remington: The Science and Practice of Pharmacy (Easton, Pa.: Mack Publishing Co., 1995).
  • unit dosage forms refers to physically discrete units suited as unitary dosages for the individuals to be treated. That is, the compositions are formulated into discrete dosage units each containing a predetermined, "unit dosage” quantity of an active agent calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specifications of unit dosage forms of the invention are dependent on the unique characteristics of the active agent to be delivered. Dosages can further be determined by reference to the usual dose and manner of administration of the ingredients.
  • two or more individual dosage units in combination provide a therapeutically effective amount of the active agent, for example, two tablets or capsules taken together may provide a therapeutically effective dosage, such that the unit dosage in each tablet or capsule is approximately 50% of the therapeutically effective amount.
  • Preparations according to the invention for parenteral administration include sterile aqueous and nonaqueous solutions, suspensions, and emulsions.
  • Injectable aqueous solutions contain the active agent in water-soluble form.
  • non-aqueous solvents or vehicles include fatty oils, such as olive oil and corn oil, synthetic fatty acid esters, such as ethyl oleate or triglycerides, low molecular weight alcohols such as propylene glycol, synthetic hydrophilic polymers such as polyethylene glycol, liposomes, and the like.
  • Parenteral formulations may also contain adjuvants such as solubilizers, preservatives, wetting agents, emulsifiers, dispersants, and stabilizers, and aqueous suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, and dextran.
  • Injectable formulations may be rendered sterile by incorporation of a sterilizing agent, filtration through a bacteria-retaining filter, irradiation, or heat. They can also be manufactured using a sterile injectable medium.
  • the active agent may also be in dried, e.g., lyophilized, form that may be rehydrated with a suitable vehicle immediately prior to administration via injection.
  • the active agent may be formulated as a depot preparation for controlled release of the active agent, preferably sustained release over an extended time period.
  • sustained release dosage forms are generally administered by implantation (for example, subcutaneously or intramuscularly or by intramuscular injection).
  • Combined preparations of the invention may be packaged with instructions for administration of the components on the combination.
  • the instructions may be recorded on a suitable recording medium or substrate.
  • the instructions may be printed on a substrate, such as paper or plastic.
  • the instructions may be present as a package insert, in the labeling of the container or components thereof (i.e., associated with the packaging or sub-packaging).
  • the instructions are present as an electronic storage data file present on a suitable computer readable storage medium, for example, CD-ROM, diskette.
  • This assay is performed at room temperature with purified recombinantly expressed human VAP-1 (SSAO).
  • Enzyme was prepared essentially as described in Ohman et al. (Protein Expression and Purification 46 (2006) 321-331 ).
  • the enzyme activity is assayed with benzylamine as substrate by measuring either benzaldehyde production, using 14C-labeled substrate, or by utilizing the production of hydrogen peroxide in a horseradish peroxidise (HRP) coupled reaction. Briefly, test compounds are dissolved in dimethyl sulfoxide (DMSO) to a concentration of 10 m .
  • DMSO dimethyl sulfoxide
  • Dose-response measurements are assayed by either creating 1 :10 serial dilutions in DMSO to produce a 7 point curve or by making 1 :3 serial dilutions in DMSO to produce 11 point curves.
  • the top concentrations are adjusted depending on the potency of the compounds and subsequent dilution in reaction buffer yielded a final DMSO concentration ⁇ 2%.
  • Hydrogen peroxide detection In a horseradish peroxidise (HRP) coupled reaction, hydrogen peroxide oxidation of 10- acetyl-3,7-dihydroxyphenoxazine produces resorufin, which is a highly fluorescent compound (Zhout and Panchuk-Voloshina. Analytical Biochemistry 253 (1997) 169-174; AmplexR Red Hydrogen Peroxide/peroxidise Assay kit, Invitrogen A22188). Enzyme and compounds in 50 mM sodium phosphate, pH 7.4 are set to pre-incubate in flat-bottomed microtiter plates for approximately 15 minutes before initiating the reaction by addition of a mixture of HRP, benzylamine and Amplex reagent.
  • HRP horseradish peroxidise
  • Benzylamine concentration is fixed at a concentration corresponding to the Michaelis constant, determined using standard procedures. Fluorescence intensity is then measured at several time points during 1 - 2 hours, exciting at 544 nm and reading the emission at 590 nm.
  • final concentrations of the reagents in the assay wells are: SSAO enzyme 1 mg/ml, benzylamine 100 ⁇ , Amplex reagent 20 ⁇ , HRP 0.1 U/mL and varying concentrations of test compound.
  • the inhibition is measured as % decrease of the signal compared to a control without inhibitor (only diluted DMSO).
  • the background signal from a sample containing no SSAO enzyme is subtracted from all data points. Data is fitted to a four parameter logistic model and IC50 values are calculated, for example by using the GraphPad Prism 4 or XLfit 4 programs.
  • SSAO activity is assayed using 14C-labeled benzylamine and analysed by measuring radioactive benzaldehyde.
  • 20 ⁇ of diluted test compound is pre-incubated at rt with 20 ⁇ _ SSAO enzyme for approximately 15 minutes with continuous agitation. All dilutions are made with PBS.
  • the reaction is initiated by adding 20 ⁇ _ of the benzylamine substrate solution containing [7-14C] Benzylamine hydrochloride (CFA589, GE Healthcare). The plate is incubated for 1 hour as above after which the reaction is stopped by acidification (10 ⁇ 1 M HCI).
  • Figure 1 shows: (a) VAP-1 expression in a muscle tissue section of a boy with Duchenne Muscular Dystrophy (DMD); and (b) VAP-1 expression in a muscle tissue section of an age-matched boy with normal muscles;
  • Figure 2 shows, at ten times and twenty times magnification, hematoxylin and eosin (H & E) staining of sections of diaphragms of mdx mice treated with: (a) vehicle; or (b) benserazide;
  • Figure 3 shows, at twenty times magnification, staining of murine F4/80 antigen in sections of diaphragms of mdx mice treated with: (a) vehicle; or (b) benserazide;
  • Figure 4 shows the effect of prednisolone, benserazide, and carbidopa, on recruitment of CD1 1 b + myeloid cells into muscles of mdx mice (*P ⁇ 0 05 ** P ⁇ 0 005, compared to vehicle);
  • Figure 5 shows the effect of prednisolone, benserazide, and carbidopa, on recruitment of: (A) CD1 1 b + Ly6C h ' 9h profibrotic myeloid cells; and (B) CD1 1 b + Ly6C l0 anti-fibrotic myeloid cells; into muscles of mdx mice (*P ⁇ 0.05, ** PO.005 compared to vehicle);
  • Figure 6 shows the effect of benserazide, carbidopa, and a combination of carbidopa and prednisolone, on recruitment of: (A) lymphoid CD4 + CD8 " T cells; and (B) lymphoid CD4 CD8 " T cells; into muscles of mdx mice (*P ⁇ 0.05, ** P ⁇ 0.005 compared to vehicle);
  • Figure 7 shows the effect of prednisolone, benserazide, carbidopa, and a combination of carbidopa and prednisolone, on mRNA levels of the following inflammatory markers in muscles of mdx mice: (A) TNF; (B) Lgals3; (C) CD53; (D) CD48; (E) CD1 1 b (*P ⁇ 0.05, ** PO.005 compared to vehicle); and Figure 8 shows the effect of prednisolone, benserazide, carbidopa, and a combination of carbidopa and prednisolone, on mRNA levels of the pro-fibrotic growth factor TGF in muscles of mdx mice ( * P ⁇ 0.05 compared to vehicle).
  • VAP-1 VAP-1 in the tissue section (detected with a goat anti-human VAP-1 antibody (Everest) followed by Cy3 labelled anti-goat IgG and imaged using a confocal microscope) and a monoclonal rat anti mouse antibody followed by a Cy3 labelled anti-rat antibody is revealed when compared to non-dystrophic control tissue.
  • VAP-1 /SSAO inhibitors including carbidopa is being examined in the mdx and dy/dy mouse models of muscular dystrophy.
  • mice were dosed once per day with carbidopa (25 mg/kg p.o.) for up to 12 weeks. The degree of inflammation and fibrosis in the muscle was then examined.
  • VAP-1 expression is increased in the muscle of a patient with Duchenne Muscular Dystrophy (DMD)
  • VAP-1 expression in a muscle tissue section of a boy with Duchenne Muscular Dystrophy was compared with VAP-1 expression in a muscle tissue section of an age-matched boy with normal muscles as a control.
  • VAP-1 expression was detected with a monoclonal rat anti-mouse VAP- 1 antibody, followed by a Cy3-labelled anti-rat IgG antibody, and imaged using a confocal microscope. The results are shown in Figure 1.
  • Figure 1(a) shows VAP-1 expression in the DMD tissue section
  • Figure 1 (b) shows VAP-1 expression in the age-matched control. VAP-1 expression is greatly increased in the DMD tissue section.
  • VAP-1 inhibitor benserazide Effect of the VAP-1 inhibitor benserazide on diaphragm muscle in a mouse model of muscular dystrophy
  • Duchenne muscular dystrophy is an X-linked muscle disease. Patients develop progressive weakness of skeletal and respiratory muscles and dilated cardiomyopathy. Clinical onset is usually between 2 and 5 years of age. Most patients loose independent ambulation in their teens, after which scoliosis develops. Death usually occurs before forty years of age and is most often the result of respiratory or cardiac failure.
  • the biochemical cause of DMD is a severe deficiency of dystrophin, an essential component of the sarcolemmal dystrophin-associated glycoprotein complex. When complex assembly is disturbed, the linkage between the muscle cell's cytoskeleton and the extracellular matrix is compromised, leading to sarcolemmal instability and increased vulnerability to mechanical stress. Fibres undergo necrosis by excessive Ca 2+ influx and are progressively replaced by connective and adipose tissue.
  • the immune system plays a pivotal role in the pathogenesis of DMD. Contraction of dystrophin deficient myofibres produces severe damage and generates cycles of muscle fibre necrosis and regeneration. Necrotizing myofibres are attacked by macrophages; inflammatory cells are present throughout the endomysial, perimysial, and perivascular areas. Macrophages are the most abundant immune cells observed in DMD muscle and both proinflammatory M1 phenotype macrophages and regeneration-focussed M2 phenotype macrophages are present. Within the inflammatory areas, few T cells, B cells, and dendritic cells are also present. Infiltrating T cells are predominantly CD4+, and smaller numbers of CD8+Tcells can be found.
  • T cell receptor repertoire of CD4+ and CD8+ T cells does not display dominant Vet or V ? rearrangements, which points toward a nonspecific cell recruitment to sites of muscle fibre destruction.
  • T cells also play an important role in the fibrotic processes present in dystrophic muscle.
  • T cell deficiency significantly reduces collagen matrix accumulation in the murine model.
  • the build up of the inflammatory response is regulated through interactions between adhesion molecules, receptors, and soluble factors, recruiting immune cells from the blood stream to the muscle tissue.
  • the most studied animal model for DMD is the mdx mouse. This was first described by Bulfield er al (Proc. Natl. Acad. Sci. USA, 1984, 81 :1189-1 192). It has a point mutation within its dystrophin gene, and as a result the mouse has no functional dystrophin in its muscles. Early in life, the mdx mouse exhibits phases of marked skeletal muscle degeneration and subsequent regeneration. As it ages, certain muscle types (including the diaphragm) show weakness and increased fibrosis. The mdx mouse diaphragm reproduces the degenerative changes of DMD, exhibiting a pattern of degeneration, fibrosis and severe functional deficit comparable to that of DMD limb muscle. This provides a quantitative framework for studying the pathogenesis of dystrophy (Stedman er al, Nature, 1991 , 352, 536-539).
  • mice 12 week old mdx mice were treated with benserazide (20mg/kg, po, once per day) or vehicle (water, once per day), in groups of 8 mice. After 6 weeks of treatment, diaphragms of the mice were collected and flash frozen in liquid nitrogen-cooled isopentane. The sections were stored on slides at -20°C until required.
  • Hematoxylin and eosin (H & E) staining was used to show cytoplasmic, nuclear, and extracellular matrix features. Hematoxylin stains nucleic acids, and eosin stains proteins nonspecifically. Staining of F4/80 antigen (a glycoprotein expressed by murine macrophages) was used to show macrophages. The results of H & E staining are shown in Figure 2, and the results of staining of murine F4/80 antigen are shown in Figure 3.
  • the H & E staining in Figure 2 shows an approximate 50% reduction in inflammatory infiltrates in mice treated with benserazide compared to vehicle.
  • the F4/80 staining in Figure 3 also shows an approximate 50% reduction in macrophage infiltration in mice treated with benserazide compared to vehicle.
  • VAP-1 inhibitor benserazide reduces the inflammatory response to muscle damage in dystrophic mice. It is known from the mdx mouse model that partial inhibition of macrophage incursion into the muscle tissue has a beneficial effect on muscle tissue maintenance. Thus, this example shows that the VAP-1 inhibitor benserazide can be used for the treatment of dystrophic muscle, and muscular dystrophy.
  • Glucocorticoids are the only medication currently clinically available that slows the decline in muscle strength and function in DMD, which in turn reduces the risk of scoliosis and stabilises pulmonary function.
  • the glucocorticoid prednisolone is often used in Europe to treat DMD.
  • This example describes the results of a comparison of the effects of prednisolone, and the VAP-1 inhibitors carbidopa and benserazide, on infiltration of myeloid and lymphoid cells into muscles of mdx mice.
  • Immune cells secrete diffusible factors, such as growth factors, IL-6, globular adiponectin, extracellular matrix (ECM) components, and ECM remodeling matrix metalloproteinases (MMPs). These diffusible factors generate ECM chemoattractive fragments, which help satellite cells escape from the basal lamina of myofibres, and promote satellite cell proliferation. In addition, cell-to- cell contact between immune and satellite cells protects satellite cells from apoptosis. Disruption of these events lead to impaired regeneration, increased muscle wasting, and deposition of fibrotic tissue, as occurs in muscular dystrophies, such as Duchenne muscular dystrophy (D D).
  • D D D Duchenne muscular dystrophy
  • Dystrophin is a critical component of the dystrophin glycoprotein complex (DGC), acting as a link between the cytoskeleton and extracellular matrix in skeletal and cardiac muscles.
  • DGC dystrophin glycoprotein complex
  • Inefficiency of the DGC in DMD causes muscle fragility, contraction-induced damage, necrosis, and inflammation.
  • satellite cells compensate for muscle fibre loss in the early stages of disease, eventually these progenitors become exhausted.
  • aberrant intracellular signalling cascades that regulate both inflammatory and immune processes contribute substantially to the degenerative process.
  • fibrous and fatty connective tissue overtakes the functional myofibres.
  • the mdx mouse model of DMD exhibits extensive limb muscle degeneration and inflammation, as well as myocardial lesions. Although lack of dystrophin makes myofibres susceptible to fragility and degeneration when contracting, this mechanical defect hypothesis for dystrophic muscle death has been unable to explain many aspects of the pathophysiology of DMD. Early immune cell infiltration in DMD patients and mdx mice is believed to represent an important aspect of dystrophic muscle pathology.
  • DMD muscle is characterized by continuous cycles of necrosis and repair of myofibers.
  • Myofibers undergoing degeneration/necrosis independently of the injury insult, release Th1 inflammatory stimuli, which recruit neutrophils and monocytes/macrophages required to clear cell debris, followed by a Th2 immune response which promotes muscle healing.
  • neutrophils are the first cells to invade injured muscle, followed by macrophages.
  • they In acutely injured muscle in mice, they begin to appear at elevated numbers within 2 hours of muscle damage, typically peaking in concentration between 6 and 24 hours after injury, and then rapidly decline in numbers.
  • Their function mostly involves phagocytic activity to remove debris but also release of TNF , as a Th1 stimuli, and production of myeloperoxidase (MPO), inducing muscle membrane damage and increasing macrophage proinflammatory activity.
  • MPO myeloperoxidase
  • neutrophils As in acute injury, neutrophils, together with macrophages, invade mdx dystrophic muscle as early as 2 weeks of age. Initial muscle injury and membrane lysis are caused by superoxide production mediated by these early infiltrating neutrophils.
  • Monocytes can differentiate into inflammatory or anti-inflammatory subsets. Inflammatory monocytes selectively traffic to the sites of inflammation, produce inflammatory cytokines and contribute to local and systemic inflammation. They are highly infiltrative and can be differentiated into inflammatory macrophages, which remove pathogen-associated molecular patterns (PAMPs) and cell debris. Antiinflammatory monocytes patrol the vasculature to monitor PAMPs and become tissue resident macrophages. During inflammation, they differentiate into anti-inflammatory macrophages, which repair damaged tissues.
  • PAMPs pathogen-associated molecular patterns
  • Mouse monocyte subsets are characterized by differential expression of an inflammatory monocyte marker Ly6C (Gr1 ) (Yang et a/, Biomarker Research 2014, 2:1 -9). Mouse monocyte subsets are grouped as pro-inflammatory Ly6C + (further divided as Ly6C high and Ly6C middle ) and anti-inflammatory Ly6C " (also called Ly6C l0W ) monocyte subsets based on expression levels of Ly6C on the cell surface.
  • Ly6C inflammatory monocyte marker
  • Mouse Ly6C + monocytes have a high antimicrobial capability due to their potent capacity for phagocytosis, secrete ROS, TNFa, nitric oxide, IL- ⁇ ⁇ , a little amount of IL-10 upon bacterial Infection, and a large amount of type 1 interferon (IFN) in response to viral ligands.
  • CCR2-CCL2 signaling in Ly6C + monocytes alters the conformational change of VLA-4 ( ⁇ 4 ⁇ 1 integrin), the ligand for VCAM-1 , leading to high affinity interaction and monocyte transmigration.
  • Ly6C + monocytes are preferentially recruited into inflamed tissue via interaction of chemokine receptor CCR2 and are more likely to mature to inflammatory M1 macrophages, which are distinguished by secretion of pro-inflammatory cytokine, TNFa, and IL-6 and contribute to tissue degradation and T cell activation.
  • Ly6C + monocytes differentiate into Ly6C " monocytes in the circulation.
  • This subset patrols the luminal side of endothelium of small blood vessels and bind to endothelium by chemokine receptor CX3CR1 via LAF-1/ICAM1-dependent manner.
  • the patrolling behavior of monocytes may be due to low-level expression of adhesion molecules, Ly6C " monocytes secrete anti-inflammatory cytokine, IL-10 upon in vivo bacterial infection.
  • Ly6C " monocytes are recruited to tissue and are more likely to differentiate into M2 macrophages, which secrete antiinflammatory cytokine and contribute to tissue repair.
  • the M1 population are proinflammatory, characterized by the expression of iNOS and secretion of proinflammatory cytokines (e.g., TNFa, IL-1/?, and IL-6), and promote muscle cell lysis.
  • proinflammatory cytokines e.g., TNFa, IL-1/?, and IL-6
  • M2 population is characterized by the expression of arginase-1 , CD163, and CD206 mannose receptor (usually in noninflammatory, repair conditions) and/or anti-inflammatory cytokines (e.g., IL- 10). They are believed to enhance muscle regeneration, by inducing satellite cell proliferation.
  • M2b macrophages exhibit a wide variety of intermediate phenotypes, including M2b and M2c, M1 and M2 being the extremes of a continuum in activation states.
  • M2b macrophages are known to release large amounts of IL-10, which promotes the proliferation of nonmyeloid cells, although, like M1 macrophages, they can also release proinflammatory cytokines, such as IL-1 ? and TNFa.
  • IL-10 can also induce M2c macrophages, which have anti-inflammatory functions.
  • Eosinophil invasion has been found in both DMD and mdx dystrophies. Previous studies observed that eosinophils increased within mdx dystrophic muscle at about 4 weeks of age, together with cytotoxic T cells invasion, and, although their number decreases during the regenerative phase, their concentration remains higher at 30-32 weeks of age, as compared to healthy muscle of age-matched wild type mice, depending on the muscle examined. Prednisone treatment has been shown to reduce eosinophil infiltration.
  • lymphocytes do not play a relevant role in healthy regenerating muscle, due to the inability of skeletal muscle to activate a T cell response.
  • MHC class I nor class II molecules has been detected on muscle fibers from healthy muscle tissues.
  • appearance of MHC class I and/or II was observed in muscle tissue of patients with idiopathic inflammatory myopathies (MM), where an autoimmune pathogenesis is now recognized, but also in regenerating fibers of patients with DMD.
  • MM idiopathic inflammatory myopathies
  • T cells are found in degenerating muscle after acute injury, but their recruitment is more robust and persistent in chronic diseases, such as muscular dystrophies.
  • T cells in dystrophic muscle may actually modulate inflammatory milieu and immune cell activity, but may also directly interfere with muscle cell function through lymphocyte-released cytokines and chemokines.
  • Very early studies correlated the reduction in T cells observed in prednisone-treated DMD patients, with reduction in muscle necrosis and fibrosis.
  • Further studies identified T cells in muscles of several DMD patients, characterized by a specific T-cell receptor (TCR) rearrangement.
  • TCR T-cell receptor
  • the over- representation of a T-cell population expressing a restricted set of TCR variable genes might indicate a selective T-cell response directed to a muscle-specific antigen.
  • Their persistence in DMD muscle could derive from either clonal expansion or conserved antigen recognition, or from the emergence of a regulatory population.
  • cytotoxic CD8 + and helper CD4 + T cells are present in affected muscles of mdx mice aged 4-8 weeks but rapidly decrease in concentration by 14 weeks of age.
  • CD8 + T cells are the first to invade dystrophic muscle, peaking at 4 weeks of age; their activation is generally driven by a Th1 cellular immune response to kill their target cells through perforin- mediated processes.
  • CD4 + T cells also invade dystrophic muscle; T helper CD4 + T cells can generally differentiate into Th effector inflammatory cells, mainly Th1 and Th2, or into regulatory T cells (Treg), both of which participate in immune responses.
  • Th1 cells are known to support macrophage M1 polarization by producing IL-1 , IL-2, TNF-a, and INF-y, while Th2 produce IL- 4, IL-13, and IL-6 sustaining the M2 macrophage polarization.
  • Treg cells are required for the resolution of the immune response. These cells produce anti-inflammatory cytokines such as IL-10.
  • T cells represent approximately 3% of all infiltrating cells in mdx muscle, with over half present as double-negative T cells (lacking both CD4 and CD8 expression), 8%-10% of which were recently identified as being NKT-like cells, which express both T and NK markers.
  • CD11 b is a transmembrane glycoprotein which is expressed on the surface of granulocytes (including neutrophils and eosinophils), monocytes, NK cells, dendritic cells, tissue macrophages and subsets of T and B cells.
  • Labelled antibody to CD11 b was used to assess infiltration of inflammatory myeloid cells into muscles of mdx mice.
  • Labelled antibody to Ly6C was used to assess infiltration of pro- and antiinflammatory monocytes into muscles of mdx mice.
  • Labelled antibody to CD4 and CD8a antigen was used to assess infiltration of Th cells (CD4 + CD8 ⁇ ) and double negative (CD4 CD8 ) T cells into muscles of mdx mice.
  • mice A total of a ninety C57BL/10ScSn-Dmd mdx /J male mice, 3 weeks of age (+/- 3days), were transferred to the in vivo research laboratory in Bar Harbor, ME. Additional ten C57BL/10ScSnJ male mice 3 weeks of age (+/- 3 days) were also transferred and served as a wild-type control group. The mice were ear notched for identification and housed in ventilated polysulfone cages with HEPA filtered air at a density of 3-4 mice per cage. The animal room was lit entirely with artificial fluorescent lighting on controlled 12 hour light/dark cycle (6 a.m. to 6 p.m. light).
  • the normal temperature and relative humidity ranges in the animal rooms were maintained at 22 ⁇ 4°C and 50 ⁇ 15%, respectively.
  • the animal room was set for 15 air exchanges per hour.
  • Filtered tap water acidified to a pH of 2.8 to 3.2 was provided ad libitum.
  • LabDiet 5K52 was provided ad libitum.
  • mice Prior to study initiation, mice were acclimated for 3 days. Mice were tested for grip strength and in the open field at ages 25 and 26 days, and drug regimen was initiated at age 27 days.
  • mice were monitored twice weekly for clinical observations and body weights. Mice in each group were treated as follows, for 4 weeks:
  • Prednisolone 0.2 and 1 mg/kg ip once per day;
  • Carbidopa plus prednisolone 100mg/kg carbidopa po twice per day, and 1 mg/kg prednisolone ip
  • mice were tested again for grip test and in the open field. Sample collection
  • mice 24 hours after the second open field tests, blood was collected by retro-orbital bleeding, and mice were humanely euthanized by C0 2 asphyxiation.
  • One triceps surae was collected fresh and processed for immune cell sorting, one tibialis anterior was preserved in RNALater for RNA extraction, one whole hind limb and the diaphragm were fixed in 2% paraformaldehyde overnight at 4°C for paraffin embedding and histology stains.
  • Serum was prepared immediately after collection and frozen at -20°C. At the end of the study, all serum samples were dosed for Creatine Kinase on a Beckman Coulter AU Clinical Chemistry analyzer following manufacturer instructions (a modification of the International Federation of Clinical Chemistry method).
  • muscle samples were enzymatically and mechanically dissociated following manufacturer instructions (Skeletal Muscle Dissociation KitCatalog no. 130-098-305, Miltenyi Biotec). Cells were stained with the following antibodies:
  • RNAs were extracted with a modification of the Trizol method, reverse transcribed, and mRNAs of TGF , and the following inflammation markers were quantified by the SYBR green method with GAPDH as a normalizer:
  • VAP-1 inhibitors benserazide and carbidopa reduced recruitment of inflammatory myeloid cells into the muscle of mdx mice.
  • CD11 b+Ly6C hi9 profibrotic myeloid cells The ratio of CD11 b+Ly6C hi9 profibrotic myeloid cells to CD11 b+Ly6C l0W anti-fibrotic myeloid cells in mdx mouse muscle following treatment of mdx mice with prednisolone, benserazide, and carbidopa is shown below:
  • Carbidopa dramatically inhibited recruitment of profibrotic myeloid cells into the muscles of mdx mice.
  • Carbidopa reduced profibrotic myeloid cell recruitment by over 80% compared with vehicle at both doses tested (compare columns 5, 6 of Figure 5A with column 7).
  • Carbidopa also inhibited recruitment of anti-fibrotic myeloid cells into the muscles of mdx mice, but by less than 50% compared with vehicle (compare columns 5, 6 of Figure 5B with column 7).
  • the measured mRNA levels of inflammatory markers in muscles of mdx mice treated with prednisolone, benserazide, carbidopa, or a combination of carbidopa and prednisolone, are shown in the Table below, and in Figure 7.
  • the inflammatory markers are: (A) TNF; (B) Lgals3; (C) CD53; (D) CD48; (E) CD11 b.
  • the level of CD53 and CD11 b mRNA was not significantly affected by treatment with prednisolone or benserazide alone. However, carbidopa alone reduced the level of CD53 mRNA by 52.6%, and the level of CD11 b mRNA by 42%. Treatment with a combination of carbidopa and prednisolone was even more effective, reducing the level of CD53 mRNA by 68%, and the level of CD11 b mRNA by 59%.
  • VAP-1 inhibitor in particular benserazide or carbidopa
  • a VAP-1 inhibitor in particular benserazide or carbidopa
  • VAP-1 inhibitor in particular carbidopa
  • VAP-1 inhibitor in particular carbidopa
  • the effects of a VAP-1 inhibitor, in particular carbidopa on reducing infiltration of dystrophic muscle by T helper cells, and DN T cells, were significantly enhanced by co-administration with a glucocorticoid; • the expression levels of inflammatory markers in dystrophic muscle were reduced by treatment with a VAP-1 inhibitor, in particular carbidopa;
  • VAP-1 inhibitor in particular carbidopa, and a glucocorticoid in combination
  • VAP-1 inhibitor in particular carbidopa or benserazide
  • VAP-1 inhibitors in particular benserazide or carbidopa
  • a steroid in particular, a glucocorticoid such as prednisolone
  • VAP-1 inhibitors such as benserazide or carbidopa, or a pharmaceutically acceptable salt thereof
  • combined treatment with VAP-1 inhibitors and steroids in particular, treatment with carbidopa, or a pharmaceutically acceptable salt thereof, and a glucocorticoid, such as prednisolone, or a pharmaceutically acceptable salt thereof, in combination
  • inflammatory myeloid cells such as monocytes, macrophages, neutrophils, or eosinophils
  • lymphoid cells • infiltration of lymphoid cells into muscle, especially T helper lymphocytes or double negative T cells;

Landscapes

  • Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Neurology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

L'invention concerne l'utilisation de composés inhibant l'activité de VAP-1/SSAO pour le traitement de la dystrophie musculaire. L'invention concerne également des préparations combinées comprenant des composés qui inhibent l'activité de VAP-1/SSAO, et leur utilisation pour le traitement de la dystrophie musculaire.
PCT/GB2014/053677 2014-06-12 2014-12-11 Inhibiteurs de vap-1 pour le traitement de la dystrophie musculaire WO2015189534A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
GB1410529.0 2014-06-12
GB201410529A GB201410529D0 (en) 2014-06-12 2014-06-12 New therapeutic uses of enzyme inhibitors
GB201421812A GB201421812D0 (en) 2014-12-08 2014-12-08 New therapeutic uses of enzyme inhibitors
GB1421812.7 2014-12-08

Publications (1)

Publication Number Publication Date
WO2015189534A1 true WO2015189534A1 (fr) 2015-12-17

Family

ID=52282752

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2014/053677 WO2015189534A1 (fr) 2014-06-12 2014-12-11 Inhibiteurs de vap-1 pour le traitement de la dystrophie musculaire

Country Status (1)

Country Link
WO (1) WO2015189534A1 (fr)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9580415B2 (en) 2013-03-13 2017-02-28 Proximagen Limited Compounds
US9676769B2 (en) 2013-03-13 2017-06-13 Proximagen Limited Imidazo[4,5-C]pyridine and pyrrolo[2,3-C]pyridine derivatives as SSAO inhibitors
WO2017098237A1 (fr) * 2015-12-07 2017-06-15 Proximagen Limited Nouvelles utilisations thérapeutiques d'inhibiteurs enzymatiques
US10065954B2 (en) 2014-09-17 2018-09-04 Proximagen Limited Substituted imidazo[4,5-c]pyridines as SSAO inhibitors
CN112004533A (zh) * 2018-02-20 2020-11-27 艾知怀斯治疗学公司 治疗运动障碍的方法和组合物
US11530208B2 (en) 2014-09-17 2022-12-20 Proximagen, Llc Imidazo[4,5-C]pyridine derived SSAO inhibitors

Citations (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993023023A1 (fr) 1992-05-15 1993-11-25 University Of Saskatchewan Procede pour prevenir les lesions de l'endothelium chez les mammiferes et pour soulager la douleur provoquee par la goutte et l'arthrite
WO2002002541A2 (fr) 2000-07-05 2002-01-10 Biotie Therapies Corp. Inhibiteur d'amine oxydases contenant du cuivre
WO2002038153A1 (fr) 2000-11-09 2002-05-16 Biovitrum Ab Nouvelle utilisation de derives de 4, 5, 6, 7-tetrahydroimidazo-[4,5-c]pyridine
US20020198189A1 (en) 2000-11-09 2002-12-26 Olivier Besencon New use
WO2003006003A1 (fr) 2001-07-12 2003-01-23 Biotie Therapies Corporation Inhibiteurs hydrazino carbocycliques des amine oxydases a teneur en cuivre
WO2003093319A1 (fr) 2002-04-29 2003-11-13 Biotie Therapies Corporation Nouveaux anticorps monoclonaux humanises contre la proteine d'adhesion vasculaire 1
WO2004067521A1 (fr) 2003-01-27 2004-08-12 Astellas Pharma Inc. Derives de thiazole et leur utilisation en tant qu'inhibiteurs de vap-1
WO2004087138A1 (fr) 2003-03-31 2004-10-14 Sucampo Ag Procede de traitement de maladie vasculaire hyperpermeable
WO2004104191A1 (fr) 2003-05-26 2004-12-02 Biotie Therapies Corporation Vap-1 cristalline et ses utilisations
WO2005014530A2 (fr) 2003-08-08 2005-02-17 La Jolla Pharmaceutical Co. Inhibiteurs d'amine oxydase sensible aux semicarbazides (ssao) et adherence mediee par la vap-1 utile pour le traitement de maladies
WO2005063261A1 (fr) 2003-12-31 2005-07-14 Faron Pharmaceuticals Oy Antibiotiques aminoglycosides utilises comme inhibiteurs de la vap-1/ssao
WO2005080319A1 (fr) 2004-02-20 2005-09-01 Biotie Therapies Corporation Derives d'hydrazino alcool utiles en tant qu'inhibiteurs d'amine-oxydases contenant du cuivre
WO2005082343A2 (fr) 2004-02-25 2005-09-09 La Jolla Pharmaceutical Company Activité enzymatique d'inhibiteurs d'amine oxidase à base d'amine et d'amide sensible aux semicarbazides (ssao), et adhésion induite par vap-1 utile pour le traitement de maladies
WO2005089755A1 (fr) 2004-03-18 2005-09-29 R-Tech Ueno, Ltd. Composition aqueuse comprenant un dérivé de thiazole
WO2006011631A2 (fr) 2004-07-27 2006-02-02 Astellas Pharma Inc. Derives de thiazole presentant une activite d'inhibition de la vap-1
WO2006013209A2 (fr) 2004-08-02 2006-02-09 Genmedica Therapeutics Sl Composes inhibant les amine oxydases contenant du cuivre, et utilisations correspondantes
WO2006028269A2 (fr) 2004-09-09 2006-03-16 Astellas Pharma Inc. Derives du thiazole presentant une activite inhibitrice de la proteine 1 d'adhesion vasculaire
WO2006094201A2 (fr) 2005-03-02 2006-09-08 La Jolla Pharmaceutical Company Inhibiteurs de l'amine oxydase sensible aux semicarbazides (ssao) et adhesion induite par vap-1 utilisee pour le traitement de maladies
WO2006134203A1 (fr) 2005-06-16 2006-12-21 Faron Pharmaceuticals Oy Composés pour le traitement ou la prévention de pathologies ou de troubles liés à l’amine oxydase
US20070066646A1 (en) 2005-08-02 2007-03-22 Genmedica Therapeutics Sl Compounds for Inhibiting Copper-Containing Amine Oxidases and Uses Thereof
WO2007120528A2 (fr) 2006-03-31 2007-10-25 La Jolla Pharmaceutical Company Inhibiteurs d'adhérence médiée par l'amine oxydase sensible aux semi-carbazides (ssao) et la vap-1 utilisés dans le traitement et la prévention de maladies
WO2008025870A1 (fr) 2006-08-28 2008-03-06 Faron Pharmaceuticals Oy Compositions utiles en particulier dans le traitement ou la prévention du syndrome métabolique
WO2008129124A1 (fr) 2007-04-20 2008-10-30 Biotie Therapies Corporation Nouveaux anticorps monoclonaux anti-vap-1 entièrement humains
WO2009055002A1 (fr) 2007-10-24 2009-04-30 La Jolla Pharmaceutical Company Inhibiteurs combinés de la cyclooxygénase et de l'amine oxydase sensible au semicarbazide (ssao) (protéine d'adhésion vasculaire, vap-1)
CN100486971C (zh) 2006-10-31 2009-05-13 北京理工大学 2,3,4,6,8-五甲氧基-二苯并呋喃及其用途
WO2009066152A2 (fr) 2007-11-21 2009-05-28 Pharmaxis Ltd. Inhibiteurs de ssao/vap-1 de type haloallylamines et leurs utilisations
WO2009096609A1 (fr) 2008-01-31 2009-08-06 R-Tech Ueno, Ltd. Dérivé de thiazole et son utilisation en tant qu'inhibiteur de la vap-1
WO2009145360A1 (fr) 2008-05-30 2009-12-03 R-Tech Ueno, Ltd. Dérivé de benzène ou de thiophène et son utilisation en tant qu'inhibiteur de la vap-1
WO2010015870A1 (fr) 2008-08-06 2010-02-11 Semmelweis Egyetem Utilisation de dihydralazine pour traiter des maladies associées à une activité amine-oxidase sensible au semi-carbazide (ssao) élevée
WO2010029379A1 (fr) 2008-09-11 2010-03-18 Semmelweis Egyetem Composés pour inhiber l’amine oxydase sensible à la semi-carbazide (ssao) / protéine d’adhésion vasculaire 1 (vap-1) et utilisations de ceux-ci pour le traitement et la prévention de maladies
WO2010031789A1 (fr) 2008-09-16 2010-03-25 Biovitrum Ab (Publ) Composés inédits i
WO2010031791A1 (fr) 2008-09-16 2010-03-25 Biovitrum Ab (Publ) Composés inédits ii
WO2010064020A1 (fr) 2008-12-04 2010-06-10 Proximagen Ltd. Composés imidazopyridines
WO2011029996A1 (fr) * 2009-09-08 2011-03-17 Biotie Therapies Corp. Utilisation d'inhibiteurs de vap-1 pour traiter des lésions fibreuses
WO2011034078A1 (fr) 2009-09-16 2011-03-24 アステラス製薬株式会社 Composé de glycine
WO2012120195A1 (fr) 2011-03-08 2012-09-13 Biotie Therapies Corporation Nouveaux composés de pyridazinone et pyridone
WO2012124696A1 (fr) 2011-03-15 2012-09-20 アステラス製薬株式会社 Composé de guanidine
WO2013163675A1 (fr) 2012-05-02 2013-11-07 Pharmaxis Ltd. Inhibiteurs de ssao 3-halogénoallylamines substituées et leurs utilisations
WO2014140592A1 (fr) 2013-03-13 2014-09-18 Proximagen Limited Dérivés d'imidazo[4,5-c]pyridine et de pyrrolo[2,3-c]pyridine en tant qu'inhibiteurs ssao
WO2014140591A1 (fr) 2013-03-13 2014-09-18 Proximagen Limited Nouveaux composés
WO2014199171A1 (fr) * 2013-06-12 2014-12-18 Proximagen Limited Nouvelles utilisations thérapeutiques d'inhibiteurs enzymatiques

Patent Citations (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993023023A1 (fr) 1992-05-15 1993-11-25 University Of Saskatchewan Procede pour prevenir les lesions de l'endothelium chez les mammiferes et pour soulager la douleur provoquee par la goutte et l'arthrite
WO2002002541A2 (fr) 2000-07-05 2002-01-10 Biotie Therapies Corp. Inhibiteur d'amine oxydases contenant du cuivre
WO2002002090A2 (fr) 2000-07-05 2002-01-10 Biotie Therapies Corp. Inhibiteur d'amines oxydases contenant du cuivre
US20040106654A1 (en) 2000-07-05 2004-06-03 Smith David John Inhibitors of copper-containing amine oxidases
WO2002038153A1 (fr) 2000-11-09 2002-05-16 Biovitrum Ab Nouvelle utilisation de derives de 4, 5, 6, 7-tetrahydroimidazo-[4,5-c]pyridine
US20020198189A1 (en) 2000-11-09 2002-12-26 Olivier Besencon New use
WO2003006003A1 (fr) 2001-07-12 2003-01-23 Biotie Therapies Corporation Inhibiteurs hydrazino carbocycliques des amine oxydases a teneur en cuivre
WO2003093319A1 (fr) 2002-04-29 2003-11-13 Biotie Therapies Corporation Nouveaux anticorps monoclonaux humanises contre la proteine d'adhesion vasculaire 1
US20040259923A1 (en) 2003-01-27 2004-12-23 Fujisawa Pharmaceutical Co. Ltd. Thiazole derivatives
WO2004067521A1 (fr) 2003-01-27 2004-08-12 Astellas Pharma Inc. Derives de thiazole et leur utilisation en tant qu'inhibiteurs de vap-1
WO2004087138A1 (fr) 2003-03-31 2004-10-14 Sucampo Ag Procede de traitement de maladie vasculaire hyperpermeable
WO2004104191A1 (fr) 2003-05-26 2004-12-02 Biotie Therapies Corporation Vap-1 cristalline et ses utilisations
WO2005014530A2 (fr) 2003-08-08 2005-02-17 La Jolla Pharmaceutical Co. Inhibiteurs d'amine oxydase sensible aux semicarbazides (ssao) et adherence mediee par la vap-1 utile pour le traitement de maladies
US20050096360A1 (en) 2003-08-08 2005-05-05 Salter-Cid Luisa M. Inhibitors of semicarbazide-sensitive amine oxidase (SSAO) and VAP-1 mediated adhesion useful for treatment of diseases
WO2005063261A1 (fr) 2003-12-31 2005-07-14 Faron Pharmaceuticals Oy Antibiotiques aminoglycosides utilises comme inhibiteurs de la vap-1/ssao
WO2005080319A1 (fr) 2004-02-20 2005-09-01 Biotie Therapies Corporation Derives d'hydrazino alcool utiles en tant qu'inhibiteurs d'amine-oxydases contenant du cuivre
WO2005082343A2 (fr) 2004-02-25 2005-09-09 La Jolla Pharmaceutical Company Activité enzymatique d'inhibiteurs d'amine oxidase à base d'amine et d'amide sensible aux semicarbazides (ssao), et adhésion induite par vap-1 utile pour le traitement de maladies
US20060025438A1 (en) 2004-02-25 2006-02-02 Salter-Cid Luisa M Amine-based and amide-based inhibitors of semicarbazide-sensitive amine oxidase (SSAO) enzyne activity and VAP-1 mediated adhesion useful for treatment of diseases
US20070078157A1 (en) 2004-02-25 2007-04-05 La Jolla Pharmaceutical Company Amine-based and amide-based inhibitors of semicarbazide-sensitive amine oxidase (SSAO)enzyme activity and VAP-1 mediated adhesion useful for treatment of diseases
WO2005089755A1 (fr) 2004-03-18 2005-09-29 R-Tech Ueno, Ltd. Composition aqueuse comprenant un dérivé de thiazole
WO2006011631A2 (fr) 2004-07-27 2006-02-02 Astellas Pharma Inc. Derives de thiazole presentant une activite d'inhibition de la vap-1
WO2006013209A2 (fr) 2004-08-02 2006-02-09 Genmedica Therapeutics Sl Composes inhibant les amine oxydases contenant du cuivre, et utilisations correspondantes
WO2006028269A2 (fr) 2004-09-09 2006-03-16 Astellas Pharma Inc. Derives du thiazole presentant une activite inhibitrice de la proteine 1 d'adhesion vasculaire
WO2006094201A2 (fr) 2005-03-02 2006-09-08 La Jolla Pharmaceutical Company Inhibiteurs de l'amine oxydase sensible aux semicarbazides (ssao) et adhesion induite par vap-1 utilisee pour le traitement de maladies
WO2006134203A1 (fr) 2005-06-16 2006-12-21 Faron Pharmaceuticals Oy Composés pour le traitement ou la prévention de pathologies ou de troubles liés à l’amine oxydase
US20070066646A1 (en) 2005-08-02 2007-03-22 Genmedica Therapeutics Sl Compounds for Inhibiting Copper-Containing Amine Oxidases and Uses Thereof
WO2007120528A2 (fr) 2006-03-31 2007-10-25 La Jolla Pharmaceutical Company Inhibiteurs d'adhérence médiée par l'amine oxydase sensible aux semi-carbazides (ssao) et la vap-1 utilisés dans le traitement et la prévention de maladies
WO2008025870A1 (fr) 2006-08-28 2008-03-06 Faron Pharmaceuticals Oy Compositions utiles en particulier dans le traitement ou la prévention du syndrome métabolique
CN100486971C (zh) 2006-10-31 2009-05-13 北京理工大学 2,3,4,6,8-五甲氧基-二苯并呋喃及其用途
WO2008129124A1 (fr) 2007-04-20 2008-10-30 Biotie Therapies Corporation Nouveaux anticorps monoclonaux anti-vap-1 entièrement humains
WO2009055002A1 (fr) 2007-10-24 2009-04-30 La Jolla Pharmaceutical Company Inhibiteurs combinés de la cyclooxygénase et de l'amine oxydase sensible au semicarbazide (ssao) (protéine d'adhésion vasculaire, vap-1)
WO2009066152A2 (fr) 2007-11-21 2009-05-28 Pharmaxis Ltd. Inhibiteurs de ssao/vap-1 de type haloallylamines et leurs utilisations
WO2009096609A1 (fr) 2008-01-31 2009-08-06 R-Tech Ueno, Ltd. Dérivé de thiazole et son utilisation en tant qu'inhibiteur de la vap-1
WO2009145360A1 (fr) 2008-05-30 2009-12-03 R-Tech Ueno, Ltd. Dérivé de benzène ou de thiophène et son utilisation en tant qu'inhibiteur de la vap-1
WO2010015870A1 (fr) 2008-08-06 2010-02-11 Semmelweis Egyetem Utilisation de dihydralazine pour traiter des maladies associées à une activité amine-oxidase sensible au semi-carbazide (ssao) élevée
WO2010029379A1 (fr) 2008-09-11 2010-03-18 Semmelweis Egyetem Composés pour inhiber l’amine oxydase sensible à la semi-carbazide (ssao) / protéine d’adhésion vasculaire 1 (vap-1) et utilisations de ceux-ci pour le traitement et la prévention de maladies
WO2010031789A1 (fr) 2008-09-16 2010-03-25 Biovitrum Ab (Publ) Composés inédits i
WO2010031791A1 (fr) 2008-09-16 2010-03-25 Biovitrum Ab (Publ) Composés inédits ii
WO2010064020A1 (fr) 2008-12-04 2010-06-10 Proximagen Ltd. Composés imidazopyridines
WO2011029996A1 (fr) * 2009-09-08 2011-03-17 Biotie Therapies Corp. Utilisation d'inhibiteurs de vap-1 pour traiter des lésions fibreuses
WO2011034078A1 (fr) 2009-09-16 2011-03-24 アステラス製薬株式会社 Composé de glycine
WO2012120195A1 (fr) 2011-03-08 2012-09-13 Biotie Therapies Corporation Nouveaux composés de pyridazinone et pyridone
WO2012124696A1 (fr) 2011-03-15 2012-09-20 アステラス製薬株式会社 Composé de guanidine
WO2013163675A1 (fr) 2012-05-02 2013-11-07 Pharmaxis Ltd. Inhibiteurs de ssao 3-halogénoallylamines substituées et leurs utilisations
WO2014140592A1 (fr) 2013-03-13 2014-09-18 Proximagen Limited Dérivés d'imidazo[4,5-c]pyridine et de pyrrolo[2,3-c]pyridine en tant qu'inhibiteurs ssao
WO2014140591A1 (fr) 2013-03-13 2014-09-18 Proximagen Limited Nouveaux composés
WO2014199171A1 (fr) * 2013-06-12 2014-12-18 Proximagen Limited Nouvelles utilisations thérapeutiques d'inhibiteurs enzymatiques

Non-Patent Citations (81)

* Cited by examiner, † Cited by third party
Title
A. COZZOLI ET AL: "Evaluation of potential synergistic action of a combined treatment with alpha-methyl-prednisolone and taurine on the mdx mouse model of Duchenne muscular dystrophy", NEUROPATHOLOGY AND APPLIED NEUROBIOLOGY, vol. 37, no. 3, 22 April 2011 (2011-04-22), pages 243 - 256, XP055172408, ISSN: 0305-1846, DOI: 10.1111/j.1365-2990.2010.01106.x *
ABELLA ET AL., DIABETOLOGIA, vol. 47, no. 3, 2004, pages 429 - 438
AIRENNE ET AL., PROTEIN SCIENCE, vol. 14, 2005, pages 1964 - 1974
ARVILOMMI ET AL., EUR J IMMUNOL, 1996, pages 25
BERTINI ET AL., J. MED. CHEM., 2005, pages 664
BIOORGANIC & MEDICINAL CHEMISTRY, vol. 21, no. 13, 2013, pages 3873 - 3881
BIOORGANIC & MEDICINAL CHEMISTRY, vol. 21, no. 5, 2013, pages 1219 - 1233
BOOMSMA ET AL., CARDIOVASC. RES., vol. 33, 1997, pages 387 - 391
BOOMSMA ET AL., DIABETOLOGIA, vol. 42, 1999, pages 233 - 237
BULFIELD ET AL., PROC. NATL. ACAD. SCI. USA, vol. 81, 1984, pages 1189 - 1192
BUSHBY ET AL.: "Diagnosis and management of Duchenne muscular dystrophy, part 1: diagnosis, and pharmacological and psychosocial management", THE LANCET, 30 November 2009 (2009-11-30)
CALLINGHAM ET AL., PROG. BRAIN RES., vol. 106, 1995, pages 305 - 321
CASTILLO ET AL., NEUROCHEM. INT., vol. 33, 1998, pages 415 - 423
DEL MAR HERNANDEZ ET AL., NEUROSCI. LETT., vol. 384, no. 1-2, 2005, pages 183 - 187
DOSTERT ET AL., J. PHARMACY & PHARMACOL., 1984, pages 782
EKBLOM, PHARMACOL. RES., vol. 37, 1998, pages 87 - 92
ENRIQUE-TARANCON ET AL., J. BIOL. CHEM., vol. 273, 1998, pages 8025 - 8032
FONTANA ET AL., BIOCHEM. J., vol. 356, 2001, pages 769 - 777
FRANS BOOMSMA ET AL: "Contrasting effects of peripheral decarboxylase inhibitors on plasma activity of aromatic-L-amino acid decarboxylase and semicarbazide-sensitive amine oxidase in Parkinson's disease", LIFE SCIENCES, vol. 57, no. 19, September 1995 (1995-09-01), pages 1753 - 1759, XP055170609, ISSN: 0024-3205, DOI: 10.1016/0024-3205(95)02153-A *
GOKTURK ET AL., AM. J. PATHOL., vol. 163, no. 5, 2003, pages 1921 - 1928
JAAKKOLA ET AL., AM. J. PATHOL., vol. 155, 1999, pages 1953 - 1965
JAKOBSSON ET AL., ACTA CRYSTALLOGR. D BIOL. CRYSTALLOGR., vol. 61, 2005, pages 1550 - 1562
JIANG ET AL., NEUROPATHOL APPL NEUROBIOL., vol. 34, no. 2, 2008, pages 194 - 204
KATHARINE BUSHBY ET AL: "Diagnosis and management of Duchenne muscular dystrophy, part 1: diagnosis, and pharmacological and psychosocial management", THE LANCET NEUROLOGY, vol. 9, 30 November 2009 (2009-11-30), pages 77 - 93, XP055170518, DOI: 10.1016/S1474- *
KIRTEN ET AL., EUR. J. IMMUNOL., 2005, pages 3119
KIRTON ET AL., EUR. J. IMMUNOL., vol. 35, no. 11, 2005, pages 3119 - 3130
KLINGER ET AL., ACTA MYOL., vol. 31, no. 3, 2012, pages 184 - 189
KLINMAN, BIOCHIM. BIOPHYS. ACTA, vol. 1647, no. 1-2, 2003, pages 131 - 137
KOSKINEN ET AL., BLOOD, 2004, pages 3388
KURKIJARVI ET AL., J. IMMUNOL., vol. 161, 1998, pages 1549 - 1557
KURKIJARVI ET AL., J. LMMUNOL., vol. 161, 1998, pages 1549 - 1557
LANGFORD ET AL., CARDIOVASC. TOXICOL., vol. 2, no. 2, 2002, pages 141 - 150
LEWINSOHN, BRAZ. J. MED. BIOL. RES., vol. 17, 1984, pages 223 - 256
LEWINSOHN, BRAZ., J. MED. BIOL. RES., vol. 17, 1984, pages 223 - 256
LYLES ET AL., BIOCHEM. PHARMACOL., 1987, pages 2847
LYLES, INT. J. BIOCHEM. CELL BIOL., vol. 28, 1996, pages 259 - 274
LYLES; PINO, J. NEURAL. TRANSM. SUPPL., vol. 52, 1998, pages 239 - 250
MAITÉ CARRE-PIERRAT ET AL: "Blocking of Striated Muscle Degeneration by Serotonin in C. elegans", JOURNAL OF MUSCLE RESEARCH AND CELL MOTILITY, KLUWER ACADEMIC PUBLISHERS, DO, vol. 27, no. 3-4, 22 June 2006 (2006-06-22), pages 253 - 258, XP019402695, ISSN: 1573-2657, DOI: 10.1007/S10974-006-9070-9 *
MANN ET AL., SKELETAL MUSCLE, vol. 1, no. 1, 2011, pages 21
MARQUES M J ET AL: "EM.P.1.01 Prevention of muscle fibrosis and myonecrosis in mdx mice by suramin, a TGF-beta 1 blocker", NEUROMUSCULAR DISORDERS, PERGAMON PRESS, GB, vol. 19, no. 8-9, September 2009 (2009-09-01), pages 549, XP026395052, ISSN: 0960-8966, [retrieved on 20090730] *
MARTTILA-ICHIHARA ET AL., JI, 2010, pages 2741
MAT CARRE-PIERRAT ET AL: "Pre-clinical study of 21 approved drugs in themouse", NEUROMUSCULAR DISORDERS, PERGAMON PRESS, GB, vol. 21, no. 5, 26 January 2011 (2011-01-26), pages 313 - 327, XP028198602, ISSN: 0960-8966, [retrieved on 20110201], DOI: 10.1016/J.NMD.2011.01.005 *
MATYUS ET AL., CURR. MED. CHEM., vol. 11, no. 10, 2004, pages 1285 - 1298
MCDONALD ET AL., ANNUAL REPORTS IN MEDICINAL CHEMISTRY, vol. 42, 2007, pages 229 - 243
MCDONALD ET AL., J. MED. CHEM., 1985, pages 186
MERCIER ET AL., BIOCHEM. J., vol. 358, 2001, pages 335 - 342
MESZAROS ET AL., EUR. J. DRUG METAB. PHARMACOKINET., vol. 24, 1999, pages 299 - 302
MORIN ET AL., J. PHARMACAL. EXP. THER., vol. 297, 2001, pages 563 - 572
MORIN ET AL., J. PHARMACOL. EXP. THER., vol. 297, 2001, pages 563 - 572
MORRIS ET AL., J. BIOL. CHEM., vol. 272, 1997, pages 9388 - 9392
NAKOS; GOSSRAU, FOLIA HISTOCHEM. CYTOBIOL., vol. 32, 1994, pages 3 - 10
NODA ET AL., FASEB J., vol. 22, no. 4, 2008, pages 1094 - 1103
NODA ET AL., FASEB J., vol. 22, no. 8, 2008, pages 2928 - 2935
OHMAN ET AL., PROTEIN EXPRESSION AND PURIFICATION, vol. 46, 2006, pages 321 - 331
O'ROURKE ET AL., JPET, 2008, pages 867
O'SULLIVAN ET AL., NEUROTOXICOLOGY, vol. 25, no. 1-2, 2004, pages 303 - 315
REMINGTON: "The Science and Practice of Pharmacy", 1995, MACK PUBLISHING CO.
SALMI ET AL., IMMUNITY, vol. 14, no. 3, 2001, pages 265 - 276
SALMI ET AL., J. EXP. MED., 1993, pages 2255
SALMI ET AL., J. EXP. MED., vol. 178, 1993, pages 2255 - 2260
SALMI; JALKANEN, TRENDS IMMUNOL., vol. 22, 2001, pages 211 - 216
SALMI; JALKANEN, TRENDS LMMUNOL., vol. 22, 2001, pages 211 - 216
SALMI; JALKANEN: "Adhesion Molecules: Functions and Inhibition", 2007, pages: 237 - 251
SALTER-CID ET AL., J. PHARMACOL. EXP. THER., vol. 315, no. 2, 2005, pages 553 - 562
SAYRE ET AL., BIOCHEM., BIOPHYS., RES. COMMUN, 2003, pages 788
SAYRE ET AL., BIOORG. MED. CHEM., 2006, pages 1444
SAYRE ET AL., BIOORG. MED. CHEM., 2007, pages 1868
SAYRE ET AL., EUR. J. BIOCHEM., 2002, pages 3645
SAYRE ET AL., J. AM. CHEM. SOC., 2002, pages 12135
SCHWELBERGER, J., NEURAL. TRANSM., vol. 1 14, no. 6, 2007, pages 757 - 762
SMITH ET AL., J. EXP. MED., vol. 188, 1998, pages 17 - 27
STEDMAN, NATURE, vol. 352, 1991, pages 536 - 539
STOLEN ET AL., CIRC. RES., vol. 95, no. 1, 2004, pages 50 - 57
STOLEN ET AL., IMMUNITY, vol. 22, no. 1, 2005, pages 105 - 115
WESTON ET AL., EASL POSTER, 2010
WESTON; ADAMS, J NEURAL TRANSM., vol. L18, no. 7, 2011, pages 1055 - 64
YANG ET AL., BIOMARKER RESEARCH, vol. 2, 2014, pages 1 - 9
YEGUTKIN, EUR. J. IMMUNOL., 2004, pages 2276
YU ET AL., BIOCHEM. PHARMACOL., vol. 47, 1994, pages 1055 - 1059
YU ET AL., BIOCHIM. BIOPHYS. ACTA, vol. 1647, no. 1-2, 2003, pages 193 - 199
ZHOUT; PANCHUK-VOLOSHINA, ANALYTICAL BIOCHEMISTRY, vol. 253, 1997, pages 169 - 174

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9580415B2 (en) 2013-03-13 2017-02-28 Proximagen Limited Compounds
US9676769B2 (en) 2013-03-13 2017-06-13 Proximagen Limited Imidazo[4,5-C]pyridine and pyrrolo[2,3-C]pyridine derivatives as SSAO inhibitors
US10590125B2 (en) 2013-03-13 2020-03-17 Proximagen, Llc Compounds
US10766897B2 (en) 2013-03-13 2020-09-08 Proximagen, Llc Imidazo[4,5-c]pyridine and pyrrolo[2,3-c]pyridine derivatives as SSAO inhibitors
US10968223B2 (en) 2013-03-13 2021-04-06 Proximagen, Llc Imidazo[4,5-c]pyridine and pyrrolo[2,3-c]pyridine derivatives as SSAO inhibitors
US11512082B2 (en) 2013-03-13 2022-11-29 Proximagen, Llc Substituted imidazo[4,5-c]pyridine compounds and compositions thereof
US10065954B2 (en) 2014-09-17 2018-09-04 Proximagen Limited Substituted imidazo[4,5-c]pyridines as SSAO inhibitors
US11530208B2 (en) 2014-09-17 2022-12-20 Proximagen, Llc Imidazo[4,5-C]pyridine derived SSAO inhibitors
WO2017098237A1 (fr) * 2015-12-07 2017-06-15 Proximagen Limited Nouvelles utilisations thérapeutiques d'inhibiteurs enzymatiques
CN112004533A (zh) * 2018-02-20 2020-11-27 艾知怀斯治疗学公司 治疗运动障碍的方法和组合物

Similar Documents

Publication Publication Date Title
US11851434B2 (en) Substituted pyrazolo[1,5-A]pyrazine compounds as ret kinase inhibitors
US20160113893A1 (en) New therapeutic uses of enzyme inhibitors
JP7129420B6 (ja) Hpk1阻害剤としてのイソキノリン
WO2015189534A1 (fr) Inhibiteurs de vap-1 pour le traitement de la dystrophie musculaire
US10138243B2 (en) Substituted pyrazolo[1,5-a]pyridine compounds as RET kinase inhibitors
AU2011212927B2 (en) Identification of LKB1 mutation as a predictive biomarker for sensitivity to TOR kinase inhibitors
WO2018106729A1 (fr) Préservation de la réponse immunitaire lors de régimes chimiothérapeutiques
EP3875078A1 (fr) Des composes pour pour le traitement de covid-19
JP2020203908A (ja) 疼痛の治療のためのvap−1阻害剤
WO2005065691A1 (fr) Traitement de gliomes malins au moyen d'inhibiteurs de tgf-beta
JP2010535220A (ja) 抗精神病用の併用療法剤を使用する統合失調症の治療のための方法および組成物
US10537560B2 (en) P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
KR20180064403A (ko) 신규한 fyn 키나제 억제제의 방법, 조성물, 및 용도
US11446274B2 (en) Use of dianhydrogalactitol or derivatives or analogs thereof for treatment of pediatric central nervous system malignancies
EP3630106A1 (fr) Composés destinés à être utilisés dans la régulation de la maturation folliculaire
WO2020206035A1 (fr) Traitement de troubles néoplasiques résistant aux inhibiteurs de cdk4/6
TW201815395A (zh) 二去水半乳糖醇或其衍生物或類似物於治療小兒中樞神經系統惡性病之用途
US20160101072A1 (en) Therapeutic uses of enzyme inhibitors
KR20120079163A (ko) 피리미딜아미노벤즈아미드와 mtor 키나제 억제제의 조합물
RU2779478C2 (ru) Сохранение иммунного ответа во время химиотерапевтических схем
EA043297B1 (ru) Применение ингибитора cdk4/6 в сохранении иммунного ответа во время химиотерапевтических схем

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14824067

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14824067

Country of ref document: EP

Kind code of ref document: A1