Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
NEW THERAPEUTIC COMPOUND AND USE IN THERAPY
Document Type and Number:
WIPO Patent Application WO/2017/017414
Kind Code:
A1
Abstract:
{4-[3-(Dimethylamino)propoxy]phenyl}methyl (4S)-4-(propan-2-yl)- 3H,4H,5H,6H,7H-imidazo[4,5-c]pyridine-5-carboxylate, and pharmaceutically or veterinarily acceptable salts thereof. Formula (I)

Inventors:
PATIENT LEE (GB)
Application Number:
PCT/GB2016/052214
Publication Date:
February 02, 2017
Filing Date:
July 21, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
PROXIMAGEN LTD (GB)
International Classes:
C07D471/04; A61K31/437; A61P29/00
Domestic Patent References:
WO2010031789A12010-03-25
Attorney, Agent or Firm:
WALLS, Steven (GB)
Download PDF:
Claims:
CLAIMS

1 . {4-[3-(Dimethylamino)propoxy]phenyl}methyl (4S)-4-(propan-2-yl)- 3H,4H,5H,6H,7H-imidazo[4,5-c]pyridine-5-carboxylate, and pharmaceutically or veterinarily acceptable salts thereof.

2. A pharmaceutical composition comprising {4-[3- (Dimethylamino)propoxy]phenyl}methyl (4S)-4-(propan-2-yl)-3H,4H,5H,6H,7H- imidazo[4,5-c]pyridine-5-carboxylate or pharmaceutically acceptable salt thereof, and one or more suitable excipients.

3. A veterinary composition comprising {4-[3- (Dimethylamino)propoxy]phenyl}methyl (4S)-4-(propan-2-yl)-3H,4H,5H,6H,7H- imidazo[4,5-c]pyridine-5-carboxylate or veterinarily acceptable salt thereof, and one or more suitable excipients.

4. A compound according to claim 1 or a pharmaceutical composition according to claim 2 for use in the treatment of, or in the manufacture of a medicament for treatment of, inflammation, an inflammatory disease, an immune or an autoimmune disorder, or inhibition of tumour growth.

5. A method for the treatment of inflammation, an inflammatory disease, an immune or an autoimmune disorder, or inhibition of tumour growth, which comprises administering to a mammal suffering such disease an effective amount of a compound as claimed in claim 1 or a pharmaceutical composition as claimed in claim 2.

6. A method according to claim 5 wherein the mammal is a human. 7. A compound according to claim 4 or a method according to claim 5 or 6 wherein the inflammation or inflammatory disease or immune or autoimmune disorder is arthritis (including rheumatoid arthritis, juvenile rheumatoid arthritis, osteoarthritis and psoriatic arthritis), synovitis, vasculitis, Sjogren's disease, a condition associated with inflammation of the bowel (including Crohn's disease, ulcerative colitis, inflammatory bowel disease and irritable bowel syndrome), atherosclerosis, multiple sclerosis, Alzheimer's disease, vascular dementia, Parkinson's disease, cerebral amyloid angiopathy, cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy, a pulmonary inflammatory disease (including asthma, chronic obstructive pulmonary disease and acute respiratory distress syndrome), a fibrotic disease (including idiopathic pulmonary fibrosis, cardiac fibrosis, liver fibrosis and systemic sclerosis (scleroderma)), an inflammatory disease of the skin (including contact dermatitis, atopic dermatitis and psoriasis), an inflammatory disease of the eye (including age related macular degeneration, uveitis and diabetic retinopathy), systemic inflammatory response syndrome, sepsis, an inflammatory and/or autoimmune condition of the liver (including autoimmune hepatitis, primary biliary cirrhosis, alcoholic liver disease, sclerosing cholangitis, and autoimmune cholangitis), diabetes (type I or II) and/or the complications thereof, chronic heart failure, congestive heart failure, an ischemic disease (including stroke and ischemia- reperfusion injury) or myocardial infarction and/or the complications thereof, or epilepsy.

8. A compound according to claim 1 or a veterinary composition according to claim 3, or a method according to claim 5, wherein the animal to be treated is a non- human mammal selected from the group consisting of rat, mouse, gerbil, guinea pig, hampster, chinchilla, cat, dog, and horse.

AMENDED CLAIMS

received by the International Bureau on 24 November 2016 (24.11.2016)

CLAIMS

1. {4-[3-(Dimethylamino)propoxy]phenyl}methyl (4S)-4-(propan-2-yl)- 3H,4H,5H,6H,7H-imidazo[4,5-c]pyridine-5-carboxylate, and pharmaceutically or veterinarily acceptable salts thereof.

2. A pharmaceutical composition comprising {4-[3- (Dimethylamino)propoxy]phenyl}methyl (4S)-4-(propan-2-yl)-3H,4H,5H,6H,7H- imidazo[4,5-c]pyridine-5-carboxylate or a pharmaceutically acceptable salt thereof, and one or more suitable excipients.

3. A veterinary composition comprising {4-[3- (Dimethylamino)propoxy]phenyl}methyl (4S)-4-(propan-2-yl)-3H,4H,5H,6H,7H- imidazo[4,5-c]pyridine-5-carboxylate or a veterinarily acceptable salt thereof, and one or more suitable excipients.

4. A compound according to claim 1 or a pharmaceutical composition according to claim 2 for use in the treatment of, or in the manufacture of a medicament for treatment of, inflammation, an inflammatory disease, an immune or an autoimmune disorder, or inhibition of tumour growth.

5. A method for the treatment of inflammation, an inflammatory disease, an immune or an autoimmune disorder, or inhibition of tumour growth, which comprises administering to a mammal suffering such disease an effective amount of a compound as claimed in claim 1 or a pharmaceutical composition as claimed in claim 2.

6. A method according to claim 5 wherein the mammal is a human.

7. A compound according to claim 4 or a method according to claim 5 or 6 wherein the inflammation or inflammatory disease or immune or autoimmune disorder is arthritis (including rheumatoid arthritis, juvenile rheumatoid arthritis, osteoarthritis and psoriatic arthritis), synovitis, vasculitis, Sjogren's disease, a condition associated with inflammation of the bowel (including Crohn's disease, ulcerative colitis, inflammatory bowel disease and irritable bowel syndrome), atherosclerosis, multiple sclerosis, Alzheimer's disease, vascular dementia, Parkinson's disease, cerebral amyloid angiopathy, cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy, a pulmonary inflammatory disease (including asthma, chronic obstructive pulmonary disease and acute respiratory distress syndrome), a fibrotic disease (including idiopathic pulmonary fibrosis, cardiac fibrosis, liver fibrosis and systemic sclerosis (scleroderma)), an inflammatory disease of the skin (including contact dermatitis, atopic dermatitis and psoriasis), an inflammatory disease of the eye (including age related macular degeneration, uveitis and diabetic retinopathy), systemic inflammatory response syndrome, sepsis, an inflammatory and/or autoimmune condition of the liver (including autoimmune hepatitis, primary biliary cirrhosis, alcoholic liver disease, sclerosing cholangitis, and autoimmune cholangitis), diabetes (type I or II) and/or the complications thereof, chronic heart failure, congestive heart failure, an ischemic disease (including stroke and ischemia- reperfusion injury) or myocardial infarction and/or the complications thereof, or epilepsy.

8. A compound according to claim 1 or a veterinary composition according to claim 3, for use in, or for use in the manufacture of a medicament for, the treatment of an animal selected from the group consisting of rat, mouse, gerbil, guinea pig, hampster, chinchilla, cat, dog, and horse.

9. A method according to claim 5, wherein the mammal to be treated is an animal selected from the group consisting of rat, mouse, gerbil, guinea pig, hampster, chinchilla, cat, dog, and horse.

Description:
NEW THERAPEUTIC COMPOUND AND USE IN THERAPY

Field of the Invention

This invention relates to a new SSAO inhibitor compound, and the use of that compound in medicine, and in particular to the use of the compound in the treatment of humans and animals suffering from a condition susceptible to modulation by an SSAO inhibitor.

Background of the Invention

Semicarbazide-sensitive amine oxidase (SSAO) activity is an enzyme activity expressed by Vascular Adhesion Protein-1 (VAP-1) or Amine Oxidase, Copper Containing 3 (AOC3), belongs to the copper-containing amine oxidase family of enzymes (EC.1.4.3.6). Therefore inhibitors of the SSAO enzyme may also modulate the biological functions of the VAP-1 protein.

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. 1999, 155, 1953-1965; Morin et al., J. Pharmacol. Exp. Ther. 2001 , 297, 563-572; Salmi & Jalkanen, Trends Immunol. 2001 , 22, 211-216]. In addition, 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].

The precise physiological role of this abundant enzyme has yet to be fully determined, but it appears that SSAO and its reaction products may have several functions in cell signalling and regulation. For example, recent findings suggest that 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]. In addition, 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], and based on this link it is expected that inhibitors of SSAO have an anti-angiogenic effect.

Several studies in humans have demonstrated that 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. 1999, 24, 299-302; Yu et al., Biochim. Biophys. Acta 2003, 1647(1-2), 193-199; Matyus et al., Curr. Med. Chem. 2004, 1 1(10), 1285- 1298; O'Sullivan et al., Neurotoxicology 2004, 25(1-2), 303-315; del Mar Hernandez et al., Neurosci. Lett. 2005, 384(1-2), 183-187]. It has been suggested that reactive aldehydes and hydrogen peroxide produced by endogenous amine oxidases contribute to the progression of cardiovascular diseases, diabetic complications and Alzheimer's disease [Callingham et al., Prog. Brain Res. 1995, 106, 305-321 ; Ekblom, Pharmacol. Res. 1998, 37, 87-92; Yu et al., Biochim. Biophys. Acta 2003, 1647(1-2), 193-199; Jiang et al., Neuropathol Appl Neurobiol. 2008, 34(2), 194-204]. Furthermore, the enzymatic activity of SSAO is involved in the leukocyte extravasation process at sites of inflammation where SSAO has been shown to be strongly expressed on the vascular endothelium [Salmi et al., Immunity 2001 , 14(3), 265-276; Salmi & Jalkanen, in "Adhesion Molecules: Functions and Inhibition" K. Ley (Ed.), 2007, pp. 237-251]. Accordingly, inhibition of SSAO has been suggested to have a therapeutic value in the prevention of diabetic complications and in inflammatory diseases [Ekblom, Pharmacol. Res. 1998, 37, 87-92; Salmi et al., Immunity 2001 , 14(3), 265-276; Salter-Cid et al., J. Pharmacol. Exp. Ther. 2005, 315(2), 553-562].

WO2007146188 teaches that blocking SSAO activity inhibits leucocyte recruitment, reduces the inflammatory response, and is expected to be beneficial in prevention and treatment of seizures, for example, in epilepsy.

O'Rourke et al (J Neural Transm. 2007; 1 14(6) :845-9) examined the potential of SSAO inhibitors in neurological diseases, having previously demonstrated the efficacy of SSAO inhibition in a rat model of stroke. An SSAO inhibitor is tested on relapsing-remitting experimental autoimmune encephalomyelitis (EAE), a mouse model that shares many characteristics with human multiple sclerosis. The data demonstrates the potential clinical benefit of small molecule anti-SSAO therapy in this model and therefore in treatment of human multiple sclerosis.

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), 105-115]. In addition, 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(1 1), 31 19-3130; Salter-Cid et al., J. Pharmacol. Exp. Ther. 2005, 315(2), 553-562; McDonald et al., Annual Reports in Medicinal Chemistry 2007 , 42, 229-243; Salmi & Jalkanen, in "Adhesion Molecules: Functions and Inhibition" K. Ley (Ed.), 2007, pp. 237-251 ; Noda et al., FASEB J. 2008 22(4), 1094-1 103; Noda et al., FASEB J. 2008, 22(8), 2928-2935]. This anti- inflammatory protection seems to be afforded across a wide range of inflammatory models all with independent causative mechanisms, rather than being restricted to one particular disease or disease model. This would suggest that SSAO may be a key nodal point for the regulation of the inflammatory response, and it is therefore likely that SSAO inhibitors will be effective anti-inflammatory drugs in a wide range of human and animal diseases. VAP-1 has also been implicated in the progression and maintenance of fibrotic diseases including those of the liver and lung. Weston and Adams (J Neural Transm. 2011 , 118(7), 1055-64) have summarised the experimental data implicating VAP-1 in liver fibrosis, and Weston et al (EASL Poster 2010) reported that blockade of VAP-1 accelerated the resolution of carbon tetrachloride induced fibrosis. In addition VAP-1 has been implicated in inflammation of the lung (e.g. Singh et al., 2003, Virchows Arch 442:491^195) suggesting that VAP-1 blockers would reduce lung inflammation and thus be of benefit to the treatment of cystic fibrosis by treating both the pro-fibrotic and proinflammatory aspects of the disease. SSAO (VAP-1) is up regulated in gastric cancer and has been identified in the tumour vasculature of human melanoma, hepatoma and head and neck tumours (Yoong KF, McNab G, Hubscher SG, Adams DH. (1998), J Immunol 160, 3978-88.; Irjala H, Salmi M, Alanen K, Gre ' nman R, Jalkanen S (2001), Immunol. 166, 6937- 6943; Forster-Horvath C, Dome B, Paku S, et al. (2004), Melanoma Res. 14, 135- 40.). One report (Marttila-lchihara F, Castermans K, Auvinen K, Oude Egbrink MG, Jalkanen S, Griffioen AW, Salmi M. (2010), J Immunol. 184, 3164-3173.) has shown that mice bearing enzymically inactive VAP-1 grow melanomas more slowly, and have reduced tumour blood vessel number and diameter. The reduced growth of these tumours was also reflected in the reduced (by 60-70%) infiltration of myeloid suppressor cells. Encouragingly VAP-1 deficiency had no effect on vessel or lymph formation in normal tissue.

For the above reasons, it is expected that inhibition of SSAO will reduce the levels of pro-inflammatory enzyme products (aldehydes, hydrogen peroxide and ammonia) whilst also decreasing the adhesive capacity of immune cells and correspondingly their activation and final extra-vasation. Diseases where such an activity is expected to be therapeutically beneficial include all diseases where immune cells play a prominent role in the initiation, maintenance or resolution of the pathology, such inflammatory diseases and immune/autoimmune diseases. Examples of such diseases include multiple sclerosis, arthritis and vasculitis.

An unmet medical need exists for new inhibitors of SSAO having utility in medicine, including veterinary medicine, in the treatment of humans and animals suffering from a condition susceptible to modulation by an SSAO inhibitor. Summary of the Invention

The present invention makes available the new SSAO inhibitor compound {4-[3-(Dimethylamino)propoxy]phenyl}methyl (4S)-4-(propan-2-yl)-3H,4H,5H,6H,7H- imidazo[4,5-c]pyridine-5-carboxylate, and pharmaceutically acceptable salts thereof.

The invention relates also to pharmaceutical compositions comprising {4-[3- (Dimethylamino)propoxy]phenyl}methyl (4S)-4-(propan-2-yl)-3H,4H,5H,6H,7H- imidazo[4,5-c]pyridine-5-carboxylate, and one or more pharmaceutically acceptable excipients and/or carriers.

The invention relates also to the use of {4-[3- (Dimethylamino)propoxy]phenyl}methyl (4S)-4-(propan-2-yl)-3H,4H,5H,6H,7H- imidazo[4,5-c]pyridine-5-carboxylate in medicine, including veterinary medicine. The invention relates also to the use of {4-[3- (Dimethylamino)propoxy]phenyl}methyl (4S)-4-(propan-2-yl)-3H,4H,5H,6H,7H- imidazo[4,5-c]pyridine-5-carboxylate in the treatment of a human or an animal suffering from a disease or condition susceptible to modulation of SSAO.

In an embodiment, the disease or condition susceptible to modulation of

SSAO is selected from inflammation, an inflammatory disease, an immune or an autoimmune disorder, or inhibition of tumour growth.

In an embodiment the inflammation or inflammatory disease or immune or autoimmune disorder is selected from arthritis (including rheumatoid arthritis, juvenile rheumatoid arthritis, osteoarthritis and psoriatic arthritis), synovitis, vasculitis, Sjogren's disease, a condition associated with inflammation of the bowel (including Crohn's disease, ulcerative colitis, inflammatory bowel disease and irritable bowel syndrome), atherosclerosis, multiple sclerosis, Alzheimer's disease, vascular dementia, Parkinson's disease, cerebral amyloid angiopathy, cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy, a pulmonary inflammatory disease (including asthma, chronic obstructive pulmonary disease and acute respiratory distress syndrome), a fibrotic disease (including idiopathic pulmonary fibrosis, cardiac fibrosis, liver fibrosis and systemic sclerosis (scleroderma)), an inflammatory disease of the skin (including contact dermatitis, atopic dermatitis and psoriasis), an inflammatory disease of the eye (including age related macular degeneration, uveitis and diabetic retinopathy), systemic inflammatory response syndrome, sepsis, an inflammatory and/or autoimmune condition of the liver (including autoimmune hepatitis, primary biliary cirrhosis, alcoholic liver disease, sclerosing cholangitis, and autoimmune cholangitis), diabetes (type I or II) and/or the complications thereof, chronic heart failure, congestive heart failure, an ischemic disease (including stroke and ischemia- reperfusion injury) or myocardial infarction and/or the complications thereof, or epilepsy.

In an embodiment, {4-[3-(Dimethylamino)propoxy]phenyl}methyl (4S)-4- (propan-2-yl)-3H,4H,5H,6H,7H-imidazo[4,5-c]pyridine-5-carbox ylate has utility in the treatment of an animal suffering from a disease or condition susceptible to modulation of SSAO. In an embodiment the animal is a non-human mammal. In an embodiment the animal is a companion animal. In an embodiment, the animal is selected from the group consisting of cat, dog, rodent including mouse, rabbit, gerbil, chinchilla, rat, guinea pig, hamster, horse, pony, donkey, livestock including pig, cow, bull, and sheep.

Brief Description of the Drawings

Figure 1 : Effect of {4-[3-(Dimethylamino)propoxy]phenyl}methyl (4S)-4-

(propan-2-yl)-3H,4H,5H,6H,7H-imidazo[4,5-c]pyridine-5-car boxylate (referred to herein as Example 1) on Arthritic Index in the Collagen-Induced Arthritis Mouse Model. Data shows a dose-dependent decrease in the rate of disease development.

Figure 2: Histological assessment of joints following treatment with Example 1 in the Collagen-Induced Arthritis Mouse Model. Data shows a dose-dependent inhibition of inflammation, pannus formation, cartilage damage, and bone resorption.

Figures 3-6: Effect of Example 1 in the DSS-lnduced Colitis Mouse Model. Data shows a dose-dependent improvement in the DSS colitis model, with the dose of 100mg/kg reaching statistically significant effects on overall body weight (Figure 3), endoscopy scores on day 12 (Figure 4), colon weight (Figure 5) and diarrhoea (Figure 6).

Figure 7: Effect of Example 1 in the mouse PLP relapsing-remitting EAE model. Data shows a dose-dependent decrease in the time to disease remission, increased time to disease relapse and decrease in disease severity.

Figure 8: Effect of Example 1 in a carbon tetrachloride induced Liver

Fibrosis Model. Data shows treatment with Example 1 significantly reduced the deposition of collagen in the liver.

Figure 9: Effect of Example 1 in a murine model of LPS-induced cytokine release. Data shows a statistically significant dose-dependent decrease in serum TN Fa levels.

Figure 10: Effect of Example 1 in the mouse choroidal neovascularisation model. Data shows a significant reduction in lesion size.

Figure 11 : Effect of Example 1 in a murine model of Duchenne's muscular dystrophy. Data shows a significant reduction in pro-inflammatory monocytes, cytokines, macrophages, B cells, T cells and TGF in mdx mice.

Description of the Invention

Compositions and Formulations

{4-[3-(Dimethylamino)propoxy]phenyl}methyl (4S)-4-(propan-2-yl)-

3H,4H,5H,6H,7H-imidazo[4,5-c]pyridine-5-carboxylate may be used as such or in the form of a pharmaceutically acceptable salt. For the avoidance of doubt, the term 'pharmaceutically acceptable salt(s)' as used herein includes veterinarily acceptable salt(s); and the term 'pharmaceutical composition(s)' as used herein includes veterinary composition(s). Pharmaceutically acceptable salts include, for example, acid addition salts derived from inorganic or organic acids, such as hydrochlorides, hydrobromides, p-toluenesulphonates, methansulphonates phosphates, sulphates, perchlorates, acetates, trifluoroacetates, propionates, citrates, malonates, succinates, lactates, oxalates, tartrates and benzoates.

A typical dosage is 1 to 200 mg/kg, administered one or more times per day or by continuous infusion. The drug is preferably administered via the intravenous route, or the oral route. In an embodiment the typical dosage is 1 to 100 mg/kg orally twice a day, or 1 to 200 mg/kg orally once per day. It will be understood, however, that the specific dose level for any particular patient will depend upon a variety of factors including the age, body weight, general health, sex, diet, time of administration, drug combination and the severity of the particular condition undergoing therapy.

A pharmaceutical or veterinary composition containing the active ingredient may be in any suitable form, for example aqueous or non-aqueous solutions or suspensions, dispersible powders or granules, transdermal or transmucosal patches, creams, ointments or emulsions.

The pharmaceutical or veterinary composition may be in the form of a sterile injectable aqueous or non-aqueous (e.g. oleaginous) solution or suspension. The sterile injectable preparation may also be in a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1 ,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, phosphate buffer solution, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed, including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables. Suspensions may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned elsewhere.

Aqueous suspensions contain the active ingredient in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents such as a naturally occurring phosphatide, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such a polyoxyethylene with partial esters derived from fatty acids and hexitol anhydrides, for example polyoxyethylene sorbitan monooleate. The aqueous suspensions may also contain one or more preservatives, for example ethyl or n-propyl p- hydroxy benzoate, one or more colouring agents, one or more flavouring agents, and one or more sweetening agents, such as sucrose or saccharin.

Non-aqueous (i.e. oily) suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.

Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are known.

The pharmaceutical or veterinary compositions of the invention may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil, for example olive oil or arachis oil, or a mineral oil, for example liquid paraffin or mixtures of these. Suitable emulsifying agents may be naturally occurring gums, for example gum acacia or gum tragacanth, naturally occurring phosphatides, for example soya bean, lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, for example sorbitan monooleate and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate.

The active agent may also be administered in the form of suppositories for rectal administration of the drug. These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials include cocoa butter and polyethylene glycols.

For topical delivery, transdermal and transmucosal patches, creams, ointments, jellies, solutions or suspensions may be employed. For sub-lingual delivery, fast dissolving tablet formulations may be used, as well as a number of the presentations described above. For oral administration, the drug may be administered as tablets, capsules or liquids.

Synthesis of {4-[3-(Dimethylamino)propoxy]phenyl}methyl (4S)-4-(propan-2- yl)-3H,4H,5H,6H,7H-imidazo[4,5-c]pyridine-5-carboxylate (Example 1)

The following abbreviations have been used:

CFA Complete Freund's adjuvant

DCM Dichloromethane

DIPEA Diisopropylethylamine

DMSO Dimethylsulfoxide

(-)-DPTT (-)-Di-0,a-p-toluyl-L-tartaric acid

DSC N,N-Disuccinimidylcarbonate

ee enantiomeric excess

ES + Electrospray

EtOAc Ethyl acetate

EtOH Ethanol

h hour

HPLC High Performance Liquid Chromatography

lUPAC International Union of Pure and Applied Chemistry

LCMS Liquid Chromatography Mass Spectrometry

LPS Lipopolysaccharide

MeCN Acetonitrile

[MH] + Protonated molecular ion

min minute

RP Reverse phase

Rt Retention time

TFA Trifluoroacetic acid Experimental Methods

Reactions were conducted at room temperature unless otherwise specified. Preparative chromatography was performed using a CombiFlash Companion system equipped with GraceResolv silica column. Reverse Phase HPLC was performed on a Gilson system with a UV detector equipped with Phenomenex Synergi Hydro RP 150x10mm, or YMC ODS-A 100/150x20mm columns. The purest fractions were collected, concentrated and dried under vacuum. Compounds were typically dried in a vacuum oven at 40°C prior to purity analysis. Compound analysis was performed by HPLC/LCMS using an Agilent 1 100 HPLC system / Waters ZQ mass spectrometer connected to an Agilent 1 100 HPLC system with a Phenomenex Synergi, RP-Hydro column (150x4.6mm, 4um, 1.5mL per min, 30°C, gradient 5- 100% MeCN (+0.085% TFA) in water (+0.1 % TFA) over 7min, 200-300nm). Enantiomeric excesses were determined by Chiral HPLC performed on an Agilent 1200 system using an Astec Chirobiotic V 100 x 4.6mm 5um column. The compounds prepared were named using lUPAC nomenclature.

INTERMEDIATE 1

(4S)-4-(Propan-2-yl)-3H,4H,5H,6H,7H-imidazo[4,5-c]pyridine

Histamine dihydrochloride (47.7g, 259mmol) was suspended in EtOH (150mL) and water (5mL), NaOH (20.7g, 518mmol) was added and the reaction mixture was heated to 75°C. Isobutyraldehyde (28.6mL, 314mmol) was added drop-wise over 50min and the reaction mixture was stirred at 75°C for 18h, cooled to 0°C for 1 h and filtered, washing with EtOH (250mL). The combined filtrates were diluted with EtOH (400mL) and (-)-DPTT (200g, 518mmol), water (1.2L) and EtOH (400mL) were added. The reaction mixture was heated to 58°C until dissolution was complete and was then allowed to cool to room temperature over 16h. The precipitate was collected by filtration and re-crystallised three times from 1 : 1 EtOH:water. This process was repeated on the same scale (47.7g) and the products were combined and re-crystallised six times from 1 : 1 EtOH:water at 80°C. The residue (146g, 155mmol) was suspended in EtOAc (285mL) and a solution of H 2 S0 4 (9.00mL, 168mmol) in water (107mL) was added. The reaction mixture was stirred at room temperature for 1.5h and the aqueous layer was separated, washed with EtOAc (177ml_), cooled to 0°C and a solution of KOH (19.2g, 342mmol) in water (16ml_) and EtOH (73ml_) was added cautiously. The reaction mixture was diluted with EtOH (177ml_) and allowed to warm to room temperature for 72h. The reaction mixture was filtered, washing with EtOH (100ml_) and concentrated in vacuo to give the title compound (21. Og, 24.5%) as a light yellow oil. LCMS (ES+): 166.1 [MH]+. HPLC: Rt 0.51 min, 98.8% purity. Chiral HPLC: Rt 8.79min, >99% ee.

EXAMPLE 1

{4-[3-(Dimethylamino)propoxy]phenyl}methyl (4S)-4-(propan-2-yl)- 3H,4H,5H,6H,7H-imidazo 4,5-c]pyridine-5-carboxylate

{4-[3-(Dimethylamino)propoxy]phenyl}methanol (13.9g, 66.6mmol) was dissolved in MeCN (200ml_) and cooled to 0°C. DSC (17.1 g, 66.6mmol) and DIPEA (11.7ml_, 90.8mmol) were added and the reaction mixture was stirred at 0°C for 1 h. A suspension of Intermediate 1 (10. Og, 60.5mmol) in DCM (100ml_) was added and the reaction mixture was stirred for 17h and concentrated in vacuo. The residue was partitioned between EtOAc (200ml_) and sat aq NaHC03 (250ml_) and the aqueous layer was extracted with EtOAc (5 x 100ml_). The combined organic fractions were concentrated in vacuo and the residue was purified by normal phase column chromatography to give the title compound (7.52g, 31 %) as a colourless gum. This process was repeated with 2.00g Intermediate 1 , and the products were combined and purified by reverse phase column chromatography to give the title compound (11.5g, 39.5%) as a colourless gum. LCMS (ES+): 401.0 [MH]+. HPLC: Rt 3.91 min, 99.7% purity.

Biological tests

In vitro SSAO enzyme inhibition assays

The in vitro SSAO enzyme inhibition assay was performed at room temperature with purified recombinantly expressed human SSAO. Enzyme was prepared essentially as described in Ohman et al. (Protein Expression and Purification 46 (2006) 321- 331). The enzyme activity was assayed with benzylamine as substrate by measuring the production of hydrogen peroxide in a horseradish peroxidase (HRP) coupled reaction. Briefly, the test compound was dissolved in dimethyl sulfoxide (DMSO) to a concentration of 10mM. Dose-response measurements were 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 1 1 point curves. The top concentration was adjusted depending on the potency of the compound and subsequent dilution in reaction buffer yielded a final DMSO concentration≤ 2%. In a horseradish peroxidase (HRP) coupled reaction, hydrogen peroxide oxidation of 10-acetyl-3,7-dihydroxyphenoxazine produced resorufin, which is a highly fluorescent compound (Zhout and Panchuk-Voloshina. Analytical Biochemistry 253 (1997) 169-174; Amplex ® Red Hydrogen Peroxide/peroxidase Assay kit, Invitrogen A22188). Enzyme and test compound in 50mM sodium phosphate, pH 7.4 were set to pre-incubate in flat-bottomed microtiter plates for approximately 15min before initiating the reaction by addition of a mixture of HRP, benzylamine and Amplex reagent. Benzylamine concentration was fixed at a concentration corresponding to the Michaelis constant, determined using standard procedures. Fluorescence intensity was then measured at several time points during 1-2h, exciting at 544nm and reading the emission at 590nm. The final concentrations of the reagents in the assay wells were: SSAO enzyme 1 ug/ml, benzylamine 100uM, Amplex reagent 20uM, HRP 0.1 U/mL and varying concentrations of test compound. The inhibition was 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 was subtracted from all data points. Data was fitted to a four parameter logistic model and an IC 50 value was calculated using the GraphPad Prism 4 or XLfit 4 programs.

Study 1

Effect of Example 1 in the established mouse collagen-induced arthritis model

Example 1 was assessed in an established mouse collagen-induced arthritis model. Disease was induced in male DBA/1J mice by means of a subcutaneous injection of a collagen (bovine)/CFA emulsion at the base of the tail. Each paw was scored daily and the sum of all four scores was recorded as the Arthritic Index (Al). The maximum possible Al was 16 (0 = no visible effects of arthritis, 1 = edema and/or erythema of one digit, 2 = edema and/or erythema of 2 joints, 3 = edema and/or erythema of more than 2 joints, 4 = severe arthritis of the entire paw and digits including limb deformation and ankylosis of the joint). As animals developed disease, they were sorted into treatment groups with Al in the range of 2-6 and an average group Al of 3.6 prior to initiation of the dosing regimen. Example 1 (10, 50 and 100 mg/kg) was administered orally twice daily from day 30 post-inoculation. The treatment resulted in a dose-dependent decrease in the rate of disease development such that at the termination of the study after two weeks of therapy the highest dose (100 mg/kg) had yielded a 46% reduction in disease severity (Figure 1).

Histological evaluation of the rear right hind limbs was also carried out. The joints were scored for severity of arthritis (inflammation, pannus, cartilage damage and bone damage) by a board certified pathologist, who was blind to the assigned treatment. Assessment of the effect of Example 1 revealed a dose-dependent effect of the joints such that at the highest dose (100 mg/kg) a significant inhibition of inflammation (60%), pannus formation (84%), cartilage damage (60%), and bone resorption (84%) was recorded (Figure 2).

Study 2

Effect of Example 1 in the DSS mouse colitis model

Example 1 was assessed in the treatment of established disease in the chronic Dextran Sulphate (DSS)-induced colitis model. DSS was administered in the drinking water as a 3% solution for five days. DSS-treated mice were orally administered vehicle, 6-Thioguanine (0.5mg/kg) or Example 1 (10mg/kg, 30mg/kg or 100mg/kg) b.i.d. from Days 6 to 19. A no treatment control group of 5 mice was also used. Body weights were measured daily for 19 days and animals were assessed visually for the presence of diarrhoea and/or bloody stool. All animals underwent video endoscopy on Days 5 (baseline), 12 and 19 to assess the extent of colitis and whether any beneficial treatment effects could be observed. All animals were euthanized on Day 19 to obtain colon tissues for pathology examination.

The therapeutic administration of Example 1 resulted in a dose-dependent improvement in the DSS colitis model, with the dose of 100mg/kg reaching statistically significant effects on overall body weight (Figure 3), endoscopy scores on day 12 (Figure 4), colon weight (Figure 5) and diarrhoea (Figure 6). The magnitude of the effect at this dose was comparable to the effect seen with the positive control, 6- Thioguanine.

Study 3

Effect of Example 1 in the mouse PLP relapsing-remitting EAE model

Example 1 was assessed in a PLP 139-151 remitting and relapsing model of experimental allergic encephalomyelitis (EAE) in female SJL mice as an indicator of a potential treatment for multiple sclerosis in humans.

Adjuvant was prepared by homogenizing 100mg Mycobacterium tuberculosis H37 RA in 50mL Incomplete Freund's Adjuvant to prepare a 2mg/mL suspension which was stored at 4-8°C. 50ug pertussis toxin from Bordetella pertussis was reconstituted in 50mL sterile PBS to prepare a 1 ug/mL solution which was kept in an ice bath. 10.35mg ΡίΡ 39 . 5 was dissolved in 10.35mL sterile PBS to prepare a 1 mg/mL solution. 10mL of PLP 139-151 was emulsified with 10mL adjuvant in ten batches in an ice bath to prepare a 0.5mg/mL PLP 139-151 / 1 mg/mL M. tuberculosis H37 RA emulsion which was kept in an ice bath. A drop of the emulsion was floated on water and did not disperse. On day 0, 70 mice were injected subcutaneously in both flanks with 0.1 mL of the PLPi 39 .i 5 i/CFA emulsion. Two hours later the mice were injected by intraperitoneal route with 0.2mL pertussis toxin (200ng/mouse). The mice were returned to routine maintenance with no adverse effects. On day 2, the mice were injected IP with 0.2mL of pertussis toxin (200ng/mouse). From day 7 the mice were weighed and scored for signs of disease as follows: 0 = no obvious signs of motor dysfunction compared to non-immunized control; 1 = limp or floppy tail; 2=limp tail and weakness in hind legs; 3 = limp tail and complete paralysis of legs or limp tail with paralysis of one front and one hind leg; 4 = limp tail, complete hind leg and partial front leg paralysis; 5 = complete hind and complete front leg paralysis. On days 10-12, the mice were sorted into treatment groups based upon average score and twice daily oral administration of Example 1 (3, 10, 30 or 100mg/kg), dexamethasone or vehicle (n=10 per group) was initiated. The disease score was monitored for 20 days.

Twice daily therapeutic oral therapy with Example 1 resulted in a dose-dependent decrease in the time to disease remission, increased time to disease relapse and decrease in disease severity (in Figure 7, day 0 has been re-set as the onset of therapy). Study 4

Effect of Example 1 in a carbon tetrachloride induced Liver Fibrosis Model

Example 1 was assessed in a carbon tetrachloride (CCU)-induced Liver Fibrosis Model. 7 week old female C57BL/6 mice were divided into four groups (n=5/group for one Control group to act as non-disease comparator and n=10/group for three CCU-treated groups based on their body weight the day before the start of the treatment. Thirty mice were intraperitoneally administered 5% CCI 4 in mineral oil at a volume of 100uL twice a week (days 0, 4, 1 1 , 14, 18, 21 and 25). Five mice intraperitoneally administered mineral oil instead of the 5% CCI 4 , served as the non- diseased control group. Example 1 (100mg/kg bid), Valsartan (20mg/kg qd, positive control) and Vehicle were administered orally (dose volume = 10 mL/kg) for 28 days (days -1 to 27). Mice in all groups were sacrificed after 16-20 hours from last dosing. Bouin's fixed left lateral liver sections were stained using picro-Sirius red solution. For quantitative analysis of fibrosis area, bright field images of Sirius red-stained section was blindly captured using a digital camera at 100-fold magnification, and the Sirius red-stained positive areas in 5 fields/ 4um section/ mouse were measured using ImageJ software (National Institute of Health, USA) and in accordance with the following expression: (collagen area/total area-vascular lumen area) x 100. The operator was blinded to treatments. Statistical analyses were performed using one- way ANOVA followed by Bonferroni multiple comparison test on GraphPad Prism 4 (GraphPad Software, USA). P values < 0.05 were considered statistically significant. A trend or tendency was assumed when a one-tailed t-test returned P values < 0.10. Results were expressed as mean ± SD. Comparison of the Valsartan group and Vehicle group in fibrosis area was assessed with student's t-test to confirm whether Valsartan worked as a positive control.

Treatment with Example 1 significantly reduced the deposition of collagen in the liver as demonstrated by Sirius red staining and thereby showed anti-fibrosis efficacy (see Figure 8). Study 5

Effect of Example 1 in a murine model of LPS-induced cytokine release

Example 1 was assessed in an LPS-induced cytokine release assay. One hour after administration of Example 1 , female Swiss Webster mice (n=5 per group) were injected intraperitoneally with 2mL/kg LPS (1.5mg/mL). One hour after LPS injection the mice were anesthetized and exsanguinated into pre-chilled serum separator microtainer tubes. The blood was processed to serum which was stored at - 80°C.The frozen serum aliquots were thawed to room temperature, diluted 1 :5 with sterile saline and assayed by ELISA for TNFa.

Oral administration of Example 1 one hour prior to LPS challenge resulted in a statistically significant dose-dependent decrease in serum TNFa levels, relative to the vehicle control (Group 1) (see Figure 9).

Study 6

Effect of Example 1 in the mouse choroidal neovascularisation model

Example 1 was assessed in a mouse choroidal neovascularisation model. Pigmented mice (C57BL6 strain, n=15 per group) each were induced with 3 choroidal burns in the right eyes using a 532nm photocoagulator. Example 1 and vehicle were dosed topically using 5uL instillations tid from days 0 to 12. On day 12, lesion size was determined by isolectine B4 immunostaining on flatmount choroids from the treated eyes and confocal microscopy for determining the size of the lesion. Example 1 showed a significant reduction in lesion size versus vehicle treated animals (see Figure 10).

Study 7

Effect of Example 1 in a murine model of Duchenne's muscular dystrophy

C57BL/10ScSn-Dmd mdx /J male mice were dosed from 27 days for 4 weeks with either vehicle (n=5), prednisolone (1 mg/kg ip qd, n=8) or Example 1 (100mg/kg po bid, n=8). At 8 weeks of age, 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).

For immune cell sorting, 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:

Stained cells were analyzed on a BD LSR II cytometer and counts of different populations were expressed in percentage of the number of live cells counted. Total RNAs were extracted with a modification of the Trizol method, reverse transcribed, and mRNAs of TGFa, and the following inflammation markers were quantified by the SYBR green method with GAPDH as a normalizer:

Example 1 showed a significant reduction in pro-inflammatory monocytes, cytokines, macrophages, B cells, T cells and TGF in the mdx mice (see Figure 11).




 
Previous Patent: A CONNECTOR

Next Patent: CONTAINER AND CLOSURE