WO2023194841A1 - Substituted tricyclic compounds and their use in ulcerative colitis - Google Patents

Substituted tricyclic compounds and their use in ulcerative colitis Download PDF

Info

Publication number
WO2023194841A1
WO2023194841A1 PCT/IB2023/052975 IB2023052975W WO2023194841A1 WO 2023194841 A1 WO2023194841 A1 WO 2023194841A1 IB 2023052975 W IB2023052975 W IB 2023052975W WO 2023194841 A1 WO2023194841 A1 WO 2023194841A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
pyrrolo
dihydroimidazo
pyridine
Prior art date
Application number
PCT/IB2023/052975
Other languages
French (fr)
Inventor
Dhananjay Sathe
Dnyaneshwar Gawas
Saravanakumar IYAPPAN
Original Assignee
Unichem Laboratories 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
Application filed by Unichem Laboratories Limited filed Critical Unichem Laboratories Limited
Publication of WO2023194841A1 publication Critical patent/WO2023194841A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system

Definitions

  • the present invention relates to compounds of Formula (I) and/or its pharmaceutically acceptable salts thereof for use in prevention, treatment and/or cure of a diseased condition in a subject suffering from Ulcerative colitis.
  • IBD Inflammatory Bowel Disease
  • UC Ulcerative colitis
  • CD Crohn’s disease
  • UC ulcerative colitis
  • Ulcerative colitis is a chronic inflammatory disease affecting the large bowel and altered regulation of cell-mediated and humoral factor-mediated immune responses against intestinal constituents which have been suggested to play a role in the development of the disease.
  • UC/IBD is associated with chronic inflammation, defined by a dysregulated response of the innate and adaptive immune systems.
  • Chronic inflammation in Crohn’s disease is characterized by a response of helper T cells type 1 (Thl) and helper T cells type 17 (Thl7), with inadequate activity of regulatory T cells (Treg), whereas UC has generally been considered a type 2 T helper cell cytokine profile (Th2).
  • Thl helper T cells type 1
  • Thl7 helper T cells type 17
  • Reg regulatory T cells
  • Th2 type 2 T helper cell cytokine profile
  • cytokines produced by these T cells signal through JAK receptors; therefore, JAK proteins have an important place in the signaling of inflammation in Inflammatory Bowel Disease.
  • Inflammation in UC occurs typically in the colon and rectum. Symptoms include the development of bloody diarrhea with or without mucus, rectal urgency, tenesmus, abdominal pain, weight loss, fatigue and extra intestinal manifestations.
  • the key risk factors for UC include genetics, environmental factors, autoimmunity and gut microbiota.
  • Cytokines play a major role in pathogenesis of UC/IBD, mainly belonging to Type I and Type II cytokines receptors [i.e., IL-6, IL-5, IL-9, IL-10, IL-13, IL-12/23, IL-22, granulocyte-macrophage colony-stimulating factor (GM-CSF), IFN-gamma],
  • Type I and Type II cytokines receptors i.e., IL-6, IL-5, IL-9, IL-10, IL-13, IL-12/23, IL-22, granulocyte-macrophage colony-stimulating factor (GM-CSF), IFN-gamma
  • Interleukin-6 along with oncostatin M (OSM) and IL-11 signal through the gpl30- associated receptor family.
  • IL-6 activates JAK1, JAK2, and TYK2 leading to STAT3 transduction, which promotes T cell proliferation, favoring the polarization of Thl7 cells.
  • IL-6 can also promote Th2 differentiation in addition, IL-6 has other functions relevant to UC/IBD, such as regulating intestinal permeability, by its effects on tight junctions, regulating the proliferation of epithelium, and healing of wounds.
  • Interferon-gamma activates JAK1 and JAK2, inducing STAT1 activation, resulting in macrophage activation, Thl polarization, and increased expression of several proinflammatory cytokines.
  • IFN-g also has a protective function in epithelial healing.
  • IFN-gamma protects from tissue destruction by inhibiting the expression of genes that code for tissue destructive factors such as matrix metalloproteinases (MMPs), serine proteases, coagulation factors, complement components, and enzymes involved in the metabolism of prostaglandin.
  • MMPs matrix metalloproteinases
  • serine proteases serine proteases
  • coagulation factors coagulation factors
  • complement components and enzymes involved in the metabolism of prostaglandin.
  • chemokines such as IL-8 and pro-inflammatory cytokines such as TNF-a in the intestinal tissue of UC patients.
  • UC ulcerative colitis .
  • Ileocolonoscopy with biopsy is one of the means to diagnoseUC.
  • a pathognomonic finding of UC is the presence of continuous colonic inflammation characterized by erythema, loss of normal vascular pattern, granularity, erosions, friability, bleeding, and ulcerations, with distinct demarcation between inflamed and non-inflamed bowel. Histopathology is the definitive tool in diagnosing UC, assessing the diseaseseverity and identifying intraepithelial neoplasia (dysplasia) or cancer.
  • the classical histological changes in UC include decreased crypt density, crypt architectural distortion, irregular mucosal surface and heavy diffuse transmucosal inflammation, in the absence of genuine granulomas.
  • CT computed tomographic
  • Nonspecific immunomodulatory drugs such as glucocorticoids, sulfasalazine/5-aminosalicylates, methotrexate, and thiopurines were among the first drugs used to treat IBD/UC.
  • Some of the medications often used to treat ulcerative colitis include Janus kinase inhibitors such as Tofacitinib; Biologies such as Adalimumab, 5 -aminosalicylic acid (5-ASA) such as Balsalazide, Mesalamine, Olsalazine, and Sulfasalazine; Sphingosine 1 -phosphate (SIP) receptor modulators such as Ozanimod.
  • Janus kinase inhibitors such as Tofacitinib
  • Biologies such as Adalimumab, 5 -aminosalicylic acid (5-ASA) such as Balsalazide, Mesalamine, Olsalazine, and Sulfasalazine
  • SIP Sphingosine 1 -phosphate
  • mAbs anti-TNF monoclonal antibodies
  • infliximab adalimumab
  • certolizumab pegol certolizumab pegol
  • golimumab antibodies with a different mechanism of action
  • mAbs have limitations in terms of safety, cost, and sustained efficacy. In fact, around 10-30% of patients treated with anti-TNF are primary non-responders to therapy, and 23-46% are secondary nonresponders.
  • JAKs Janus Kinases
  • An object of the present invention is to provide a means to prevent, cure, treat and/or to ameliorate the diseased condition in a subject suffering from inflammatory disease and/or Ulcerative colitis.
  • An object of the present invention is to provide a compound of Formula (I) or a pharmaceutically acceptable salt thereof, for use in prophylactic and/or therapeutic treatment of diseased condition in a subject suffering from Ulcerative colitis.
  • An object of the present invention is to provide a compound of Formula (I) or a pharmaceutically acceptable salt thereof, for use in the manufacturing of medicament for prophylactic and/or therapeutic treatment of diseased condition in a subject suffering from Ulcerative colitis.
  • An object of the present invention is to provide a composition comprising of a compound of Formula (I) or a pharmaceutically acceptable salt thereof, for use in prophylactic and/or therapeutic treatment of Ulcerative colitis.
  • An object of the present invention is to provide a means for treatment of Ulcerative colitis, by administering a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • Present disclosure provides a means to prevent, cure, treat and/or to ameliorate the diseased condition in a subject suffering from Ulcerative colitis. More specifically, the said invention provides a compound of Formula (I) or a pharmaceutically acceptable salt thereof, for use in the manufacturing of medicament for treatment of Ulcerative Colitis. The said invention also provides a means for treatment of Ulcerative Colitis by administering to the subject a therapeutically effective amount of compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • Figure 1 shows Percentage Change in Body Weight (g) of the Groups
  • Figure 2 depicts Cumulative Feed Intake of the groups
  • Figure 3 illustrates DAI score of the groups
  • Figure 4 depicts Colon length of the groups
  • Figure 5 relates to Colon weight of the groups
  • Figure 6 shows Histopathology scoring of the Groups
  • Figure 7 illustrates Effect of Test compound on Serum IL6 levels
  • Figure 8 depicts Effect of Test compound on Colon IL6 levels
  • Figure 9 relates to Effect of Test compound on Serum IFN-Gamma level
  • Figure 10 shows Effect of Test compound on colon IFN-Gamma level
  • Figure 11 illustrates Effect of Test compound on Colon IL8 levels
  • a compound for use or "a compound as described herein” means the compound or a pharmaceutically acceptable salt, thereof.
  • a compound of Formula (I) means a compound of that formula and pharmaceutically acceptable salts, thereof.
  • composition as used herein in the present disclosure may comprise a compound of Formula (I) or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable excipient.
  • excipient or ‘excipients’ as used herein refers to inactive or usually inert substances that are added to the formulation which do not affect the therapeutic action of the active ingredient, but serve as a vehicle or medium for the active ingredient. It may be used to provide a desired consistency, to improve stability, and/or to adjust osmolality of the composition or adjust permeability.
  • the excipients may be selected from the substances that are known to the skilled person for use in the form of compositions that are dependent on the route of administration.
  • excipients include diluents, carriers, binding agents, fillers lubricants, disintegrants, wetting agents, suitable coatings, stabilizers, sterilized water, physiological saline, suitable propellant cocoa butter, glycerides, suspending agents, emulsifying agents, preservatives polymers, solubilizers, cryoprotectants, lyoprotectants, bulking agent/s and/or pharmaceutically acceptable buffers or a mixture thereof.
  • suitable excipients are listed in standard references such as Handbook of Pharmaceutical Excipients (Rowe RC, Sheskey P, Quinn M.
  • composition ‘composition’, ‘medicament’, ‘pharmaceutical formulation’ and ‘pharmaceutical composition’ are used interchangeably and refer to preparations which are in such a form as to permit the biological activity of the active ingredients to be effective, and therefore may be administered to a subject for therapeutic use, wherein the subject is a mammal, preferably human.
  • administer refers to implanting, absorbing, ingesting, injecting, inhaling, or otherwise introducing an inventive compound, or a pharmaceutical composition thereof.
  • an “effective amount” or a “therapeutically effective amount” of a compound or any active ingredient as described herein are used interchangeably and refers to an amount sufficient to elicit a desired biological response, i.e., treating the condition.
  • the effective amount of a compound may vary depending on such factors as the desired biological endpoint, the pharmacokinetics of the compound, the condition being treated, the mode of administration, and the age and health of the subject.
  • An effective amount encompasses therapeutic and prophylactic treatment.
  • inflammation or “inflammatory disease” as used herein relates to any type of inflammatory immune response in a subject.
  • the inflammation may be associated with a chronic inflammatory disease.
  • treatment comprises substantially curing (i.e. eliminating) the inflammation or an underlying chronic inflammatory disease, or reducing (either permanently or temporarily) the symptoms associated with the inflammation.
  • symptoms may include swelling, pain, itching, heat, redness (e.g. of the skin), loss of function (e.g. of joints or limbs), and difficulty in breathing.
  • a means to prevent, cure, treat and/or to ameliorate the diseased condition in a subject suffering from inflammatory disease particularly there is provided a means to prevent, cure, treat and/or to ameliorate the diseased condition in a subject suffering from Ulcerative Colitis.
  • a compound of Formula (I) and/or a pharmaceutically acceptable salt thereof for use in the manufacturing of medicament for prophylactic and/or therapeutic treatment of diseased condition in a subject suffering from Ulcerative Colitis.
  • the medicament having compound of Formula (I) or a pharmaceutically acceptable salt thereof may be administered in a subject for prophylactic and/or therapeutic treatment ofdiseased condition in a subject suffering from Ulcerative Colitis.
  • the compound of Formula I is wherein Q is a group of formula QI or Q2;
  • R 2 is hydrogen or a Ci-Cio alkyl group
  • R a and R b independently represent hydrogen or a Ci-Cio alkyl group.
  • alkyl is intended to include branched and straight-chain saturated aliphatic hydrocarbon groups and cycloalkyl group having the specified number of carbon atoms.
  • Cyno alkyl is intended to include Ci, C 2 , C 3 , C 4 , C 5 , C 6 , C 7 , C 8 , C 9 , and Cio alkyl groups.
  • Preferred alkyl groups have from 1-6, especially 1-4, carbon atoms.
  • Example alkyl groups include, but are not limited to, methyl (Me), ethyl (Et), propyl (e.g., n- propyl and isopropyl), butyl (e.g., n-butyl, isobutyl, t-butyl), pentyl (e.g., n-pentyl, isopentyl, neopentyl).
  • the said alkyl may be further substituted by alkyl, halogen, amides, esters, acids, cyanide, amines.
  • cycloalkyl refers to cyclized alkyl groups, including monocyclic ring systems.
  • C3- 13cycloalkyl is intended to include C3, C4, C5, C6, and C7cycloalkyl groups.
  • Preferred cycloalkyl groups have from 3-8, especially 3-6, carbon atoms.
  • Example cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like.
  • the compounds of formula (I) may form salts with alkali metals such as sodium, potassium and lithium, with alkaline earth metals such as calcium and magnesium, with organic bases such as dicyclohexylamine, tributylamine, pyridine and amino acids such as arginine, lysine and the like.
  • alkali metals such as sodium, potassium and lithium
  • alkaline earth metals such as calcium and magnesium
  • organic bases such as dicyclohexylamine, tributylamine, pyridine and amino acids such as arginine, lysine and the like.
  • amino acids such as arginine, lysine and the like.
  • the compounds of formula (I) may form salts with a variety of organic and inorganic acids.
  • pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric, borates and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicyclic, sulfanilic, 2-acetoxybenzoic, fumaric, benzenesulfonic , toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, and the like.
  • zwitterions may be formed.
  • the compounds of present invention have more than one basic site, they have the capacity to form a salt with more than one molecule of acid.
  • the present disclosure embodies mono di or tri salts of the compounds of this disclosure.
  • the compound derived from Formula (I) includes but are not limited to one or more compounds selected from Table 1 :
  • the compound of Formula (I) including compounds given in Table 1.
  • the skilled person in the art may prepare the said compounds by using any of the known methods in chemistry.
  • the said compounds may be prepared using the process as described in below scheme.
  • Scheme I illustrates the preparation of a compound of formula (I) wherein Q, Ri and R2 are defined as above and Li and L2 represent X or leaving groups.
  • X may be a leaving group which is either the same as that of Li or L2 or other than that of Li and L2.
  • X may also be a group that can be easily substituted by or converted to -CORi.
  • the leaving group L b L 2 or X is one which can be easily replaced by the desired group or atom.
  • the leaving group may be selected from halogen atoms, alkoxy and sulfonyloxy groups.
  • sulfonyloxy groups include, but are not limited to, alkylsulfonyloxy groups (for example methyl sulfonyloxy (mesylate group) and trifluorom ethyl sulfonyl oxy (triflate group)) and arylsulfonyloxy groups (for example /- toluenesulfonyloxy (tosylate group) and /-nitrosulfonyloxy (nosylate group)).
  • L 2 and X may be particularly selected from halogens such as bromo, chloro or iodoand a triflate group.
  • halogens such as bromo, chloro or iodoand a triflate group.
  • the selection of X is well within the understanding and knowledge of the skilled person.
  • a compound of formula 1-1 is converted into a compound of formula 1-2 by a displacement reaction of a compound of formula 1-1 with ammonia solution in a suitable solvent, such as water, THF, 1,4-Di oxane, Dimethyl formamide (DMF), Dimethyl sulfoxide (DMSO) or Acetonitrile (ACN), or mixture(s) therefore at a temperature ranging from 45°C to 120°C for 0.5 hours to 20 hours to form a compound of formula 1-2.
  • a suitable solvent such as water, THF, 1,4-Di oxane, Dimethyl formamide (DMF), Dimethyl sulfoxide (DMSO) or
  • a compound of formula 1-2 is converted to a compound of formula 1-3 by reacting a compound of formula 1-2 with a tritiating agent such as trifluoromethanesulfonic anhydride or a halogenating agent in a suitable solvent such as acetonitrile, chloroform or tetrahydrofuran at a temperature ranging from -20° C to the refluxing temperature for a time period between about 1 hour to about 10 hours.
  • a tritiating agent such as trifluoromethanesulfonic anhydride or a halogenating agent
  • a suitable solvent such as acetonitrile, chloroform or tetrahydrofuran
  • a halogenating agent according to the present invention is a reagent that is a source of halogen.
  • the agent may be a chlorinating agent such as chlorine, thionyl chloride, N-Chlorosuccinimide, Oxalyl Chloride or a brominating agent such as bromine, NBromosuccinimide, Carbon Tetrabromide or an iodinating agent such as Iodine, Hydriodic acid or N-Iodosuccinimide.
  • the halogenating agent may be selected according to the knowledge and understanding of skilled person.
  • a Sonogashira reaction with a compound of formula 1-3 and an acetylene derivative using a suitable catalyst provides a compound of formula 1-4.
  • the reaction conditions for a Sonogashira reaction vary depending on the starting material, the solvent and the transition metal catalyst. The reaction conditions are not limited in particular as long as they are similar to the conditions of the present reactions, and the methods well known to those skilled in the art can be used.
  • Examples of preferred solvents include acetonitrile, tetrahydrofuran, 1,4- dioxane, 1,2-dimethoxy ethane, benzene, toluene, xylene, 1 -methyl -2-pyrrolidone, N,Ndimethylformamideand dimethylsulfoxide, dichloromethane or mixture thereof.
  • the reaction temperature should be a temperature that is sufficient to complete the coupling reaction, and is preferably from room temperature to 100°C.
  • the present reaction can be carried out under an inert gas atmosphere, and also under a nitrogen or an argon gas atmosphere. Under the preferred reaction conditions, this reaction is completed in 1 hour to 24 hours.
  • the transition metal catalyst is preferably a palladium complex.
  • palladium complexes examples include, but not limited to palladium(II) acetate, dichlorobis(triphenylphosphine)palladium(II), tris(dibenzylideneacetone) dipalladium (0) and tetrakis(triphenylphosphine)palladium(0).
  • a phoshorous chelating agent such as triphenylphosphine, tri-o-tolylphosphine or tri-tert-butylphosphine may be added in order to obtain satisfactory results.
  • reaction may be accelerated using a metal halide or a quaternary ammonium salt or other such salts, preferably copper(I) iodide, lithium chloride, tetrabutylammoniumfluoride or silver(I) oxide.
  • a metal halide or a quaternary ammonium salt or other such salts preferably copper(I) iodide, lithium chloride, tetrabutylammoniumfluoride or silver(I) oxide.
  • Preferred results can also be obtained in the presence of a base; the base used is not limited in particular as long as it is used in a coupling reaction similar to the present reaction Examples of such bases include, but not limited to diethylamine, triethylamine, N,N-diisopropylethylamine, piperidine and pyridine.
  • a compound of formula 1-4 can readily undergo 5-endo-dig cyclization in the presence of a base or transition metal catalyst in the presence of a suitable solvent such as alcoholic solvents or THF or DMA to provide a compound of formula 1-5.
  • a base may be selected from Potassium tert-butoxide, Lithium hydride, Lithium Aluminium hydride and n-butyl lithium and the transition metal catalyst may be selected from Palladium and a copper catalyst.
  • a compound of formula 1-5 can be optionally protected by treating it with a protecting group to provide a compound of formula 1-6.
  • Exemplarily a compound of formula 1-5 is converted to the corresponding compound of formulaI-6, wherein R 3 is benzenesulfonyl or benzyl, by treating the compound of formula I- 5 with benzenesulfonyl chloride, benzyl chloride or benzylbromide in the presence of a base, such as sodium hydride or potassium carbonate, and a polar aprotic solvent, such as dimethylformamide or tetrahydrofuran.
  • the reaction mixture is stirred at a temperature between about 0°C. to about 70°C, preferably about 30°C, for a time period between about 1 hour to about 3 hours, preferably about 2 hours.
  • R 3 is a protecting group such as benzenesulfonyl, substituted benzenesulfonyl, methylsulfonyl, benzyl or carbamate protecting groups such as Boc (t-Butyloxycarbonyl) and CBz (carboxybenzyl) or other groups such as benzoyl, iso-butanoyl, acetyl, phenoxyacetyl, 4- (t-butyl)benzoyl, 4-(t-butyl)phenoxyacetyl, 4-(methoxy)benzoyl, 2-(4-nitrophenyl) ethyl oxycarbonyl, 2-(2,4-dinitrophenyl)ethyloxy-carbonyl, 9-fluorenylmethoxycarbonyl, diphenylcarbamoyl or formamidine groups.
  • Boc t-Butyloxycarbonyl
  • CBz carboxybenzyl
  • benzoyl iso-
  • benzoyl isobutanoyl, 4-(t-butyl)benzoyl, 2-(4-nitro-i5 phenyl)ethyloxycarbonyl, 2-(2,4- dinitrophenyl)ethyl-oxycarbonyl, 9-fluorenylmethoxycarbonyl, 4-(methoxy)-benzoyl or para- (t-butyl)phenoxyacetyl, para-nitrophenyl-2-ethyloxycarbonyl group or2-N-acetyl with the 6- O-diphenylcarbamoyl group.
  • Compounds of formula 1-5 and 1-6 can be converted to a compound of formula 1-8 and 1-7, respectively in a similar way as the process described for the preparation of a compound of formula 1-3.
  • Compounds of formula 1-8 can be converted into compounds of formula (I) by a process known to the person skilled in the art. Such process may include converting X of formula 1-8 directly to an amide group or via formation of ester, anhydride, aldehyde, ketone, cyanide, acid or any such group which can be converted to an amide group which is well within the understanding and knowledge of the skilled person.
  • compounds of formula 1-8 when X is converted to an ester group and successively converted to an amide, compounds of formula 1-8 can be treated with an esterifying agent in the presence of a base in a polar aprotic solvent like THF, 1,4-Dioxane, DMF, DMSO and ACN at -75°C to 100°C temperature for 0.5 hours to 20 hours which leads to formation of ester derivative.
  • a polar aprotic solvent like THF, 1,4-Dioxane, DMF, DMSO and ACN
  • the ester derivative on reaction with a trialkylaluminium (like, trimethylaluminium) and required amine derivatives or ammonia solution in the presence of solvents like Toluene, chloroform, methanol, ethanol, THF, 1,4-Dioxane, DMF, DMSO and ACN at -10°C to 100°C temperature for 0.5 hours to 20 hours gives an amide having formula I.
  • a compound of formula 1-7 can be converted to a compound of formula I-9using a similar process that may be used for conversion of a compound of formula 1-8 to a compound of formula I.
  • a compound of formula 1-9 can be converted into a compound of formula I by cleaving the protecting group R 3 .
  • Protecting groups of a compound of formula 1-9 can be cleaved by deprotecting agents as understood by the skilled person to obtain a compound of formula I.
  • deprotecting agents for an amino protective group are acids such as trifluoroacetic acid, trichloroacetic acid, dichloroacetic acid p-toluenesulfonic acid or bases such as alkali or alkaline bases.
  • the deprotection is carried out by treating 1-9 with an alkali base, such as sodium hydroxide or potassium hydroxide, sodium carbonate, potassium carbonate, potassium tert-butoxide, sodiumtert-butoxide in an alcohol solvent, such as methanol or ethanol, or mixed solvents, such as alcohol/tetrahydrofuran or alcohol/water.
  • an alkali base such as sodium hydroxide or potassium hydroxide, sodium carbonate, potassium carbonate, potassium tert-butoxide, sodiumtert-butoxide in an alcohol solvent, such as methanol or ethanol, or mixed solvents, such as alcohol/tetrahydrofuran or alcohol/water.
  • R 3 is benzyl
  • deprotection is eitherconducted by treating 1-9 with sodium in ammonia at a temperature of about -78° C for a time period between about 15 minutes to about 1 hour or by using hydrogen and a catalyst, such as palladium hydroxide on carbon, Pd/C, Raney Nickel, Raney Nickel in combination with NH2-NH2 or Hydrogen.
  • a catalyst such as palladium hydroxide on carbon, Pd/C, Raney Nickel, Raney Nickel in combination with NH2-NH2 or Hydrogen.
  • Other suitable deprotecting agents are Lewis acids, such as, for example boron trifluorideetherate or zinc bromide in dichloromethane/isopropanol, aq. HC1, aq. HBr, HBr in acetic acid, sulfuric acid.
  • Scheme II illustrates the preparation of a compound of formula (I) wherein Q, Ri R 2 , R3 and X as defined in aforementioned embodiment.
  • R represents alkoxy (-OR) or CX3, Z is NO2.
  • a compound of formula I- 10 can be converted to the corresponding compound of formula 1-11, by treating the compound of formula 1-10 with protecting group R3 such as benzenesulfonyl chloride, benzyl chloride or benzylbromide in the presence of a base, such as sodium hydride, potassium carbonate, sodium hydroxide, potassium hydroxide or cesium carbonate or alkyl lithium such as n-butyl lithium, secondary butyl lithium, tertiary butyl lithium or lithium diisopropyl amide.
  • protecting group R3 such as benzenesulfonyl chloride, benzyl chloride or benzylbromide in the presence of a base, such as sodium hydride, potassium carbonate, sodium hydroxide, potassium hydroxide or cesium carbonate or alkyl lithium such as n-butyl lithium, secondary butyl lithium, tertiary butyl lithium or lithium diisopropyl amide.
  • reaction may be carried out in solvent such as dimethylformamide, dimethylacetamide, tetrahydrofuran, hexamethyl phosphoramide, dimethyl sulfoxide, 1,4-Dioxane, acetonitrile, water, dichloromethane, Toluene, DMSO or mixture(s) therefore.
  • solvent such as dimethylformamide, dimethylacetamide, tetrahydrofuran, hexamethyl phosphoramide, dimethyl sulfoxide, 1,4-Dioxane, acetonitrile, water, dichloromethane, Toluene, DMSO or mixture(s) therefore.
  • the reaction mixture is stirred at a temperature between about 0°C. to about 70°C., preferably about 10°C, for a time period between about 1 hour to about 10 hours, preferably about 4 hours.
  • R3 is a protecting group defined as above.
  • Compounds of formula 1-11 can be converted to a compound of formula 1-12 by reacting a compound of formula I- 11 with an acylating agent such as trifluoroacetic anhydride, tri chloroacetyl chloride , acid halides, acid anhydrides in a suitable solvent such as acetonitrile, chloroform, n-methyl pyrrolidone, toluene, tetrahydrofuran, dimethylformamide, dimethylsulfoxide, dimethylacetamide 1,4-Dioxane chlorinated alkyl or aryl solvents such as dichloromethane or chlorobenzene, di chlorobenzene or di chloroethane or mixture(s) therefore at a temperature ranging from -20°C to the refluxing temperature for a time period between about 1 hour to about 15 hours preferably at 65-75°Cfor 4-5 hours.
  • an acylating agent such as trifluoroacetic anhydride, tri chloroacetyl chloride
  • a compound of formula 1-12 can be converted to a compound of formula 1-13 by treating compound of formula 1-12 with nitrating agents such as alkyl ammonium nitrate for example, tetrabutyl ammonium nitrate or tetramethyl ammonium nitrate and using trifluoroacetic anhydride in solvents such as dichloromethane, toluene, acetonitrile, tetrahydrofuran, chlorobenzene, nitrobenzene, di chloroethane 1,4-Dioxane, acetonitrile, water, dimethylsulfoxide or mixture(s) therefore, at a temperature ranging from -10°C to 100°C for a time period between about 1 hour to about 30 hours preferably for 5 hours.
  • nitrating agents such as alkyl ammonium nitrate for example, tetrabutyl ammonium nitrate or tetramethyl ammonium nitrate and using tri
  • a compound of formula I- 13 can be converted to a compound of formula 1-14 by reaction with ammonia or with primary amines such as methyl amine, ethyl amine, isopropyl amine, npropyl amine, isobutylamine or n-butylamine in suitable solvents such as tetrahydrofuran, dichloromethane, 1,4 dioxane, toluene, dimethylformamide, water, alcoholic solvents, DMSO, acetonitrile or mixture(s) thereof at a temperature ranging from -10°C to the refluxing temperature for a time period between about 1 hour to about 25 hours, preferably for 8-10 hours.
  • primary amines such as methyl amine, ethyl amine, isopropyl amine, npropyl amine, isobutylamine or n-butylamine in suitable solvents such as tetrahydrofuran, dichloromethane, 1,4 dioxane
  • a compound of formula I-14 can be converted to compound of formula 1-15 by reduction of nitro group using metal catalyst such as palladium on carbon, Raney nickel in combination with NH2-NH2 or Hydrogen, iron/ammonium chloride, platinum on carbon, zinc/ammonium chloride, Fe/AcOH or sodium dithionite in suitable alcoholic solvents such as methanol, ethanol or water or cyclic/acyclic ethers such as tetrahydrofuran or 1,4-dioxane or acetonitrile and water or in mixture of suitable alcoholic solvents such as methanol, ethanol, or cyclic/acyclic ethers such as tetrahydrofuran or 1,4-dioxane or acetonitrile and water at temperature ranging from -10°C to reflux temperature, preferably at room temperature for time period of 1 to 10 hours.
  • a compound of formula 1-15 is optionally converted to compound of formula I- 15a by treating compound of formula 1-15 with alkylating agents or treating
  • a compound of formula 1-15 or 1-15 a can be converted to compound of formula 1-16 by cyclization methods using reagents such as triethylorthoformate and acid catalyst viz para toluene sulphonic acid or dimethylformamide or formic acid and metal catalyst such as zinc acetate, using solvents such as toluene, halobenzene such as chlorobenzene, 1,2 di chlorobenzene, dimethylformamide, dimethylacetamide, tetrahydrofuran, acetonitrile, 1,4- dioxane, water, acetic acid, formic acid, formamide or mixture(s) thereof at a temperature ranging from room temperature to reflux temperature preferably at 0°C-100°C for period of 1 to 10 hours preferably for 5 hours.
  • reagents such as triethylorthoformate and acid catalyst viz para toluene sulphonic acid or dimethylformamide or formic acid and metal catalyst such as zinc acetate
  • a compound of formula 1-16 can be converted to compound of formula 1-17 by hydrolysis using alkali hydroxide such as sodium hydroxide, potassium hydroxide or lithium hydroxide or aqueous solution thereof or any other reagents as understood by the skilled person in suitable alcoholic solvents such as methanol or ethanol or water or in mixture of suitable alcoholic solvents such as methanol, ethanol, propanol, butanol, iso-butanol or cyclic/acyclic ethers such as tetrahydrofuran or 1,4-Dioxane or acetonitrile and water to obtain a compound of formula 1-17, at a temperature ranging from room temperature to reflux temperature preferably at a temperature 80°C for time period of 30 minutes to 10 hours.
  • suitable alcoholic solvents such as methanol or ethanol or water
  • suitable alcoholic solvents such as methanol or ethanol or water or in mixture of suitable alcoholic solvents such as methanol, ethanol, propanol, butanol
  • a compound of formula 1-17 can be converted into a compound of formula 1-18 by cleaving the protecting group R 3 .
  • Protecting groups of a compound of formula 1-17 can be cleaved by deprotecting agents as understood by the skilled person to obtain a compound of formula (I).
  • deprotecting agents for an amino protective group are acids such astrifluoroacetic acid, trichloroacetic acid, dichloroacetic acid p-toluenesulfonic acid, HC1, HBr, H2SO4 or bases such as alkali or alkaline bases.
  • the deprotection is carried out by treating 1-17 with an alkali base, such as sodium hydroxide or potassium hydroxide, sodium carbonate, potassium carbonate, cesium carbonate in an alcohol solvent, such as methanol or ethanol, or mixed solvents, such as alcohol/tetrahydrofuran or alcohol/water, MDC, THF, toluene, CAN, water or mixture(s) thereof.
  • an alkali base such as sodium hydroxide or potassium hydroxide, sodium carbonate, potassium carbonate, cesium carbonate
  • an alcohol solvent such as methanol or ethanol, or mixed solvents, such as alcohol/tetrahydrofuran or alcohol/water, MDC, THF, toluene, CAN, water or mixture(s) thereof.
  • the reaction is carried out at room temperature to reflux temperature for a time period between about 15 minutes to about 1 hour, preferably 30 minutes.
  • R 3 is benzyl
  • deprotection is either conducted by treating 1-17 with sodium in ammonia at a temperature of about -78°C for a time period between about 15 minutes to about 10 hour or by using hydrogen and a catalyst, such as palladium hydroxide on carbon, Pd/C in ether solvents such as tetrahydrofuran and alcohol such as tert-butanol, MDC, THF, toluene, CAN, water or mixture(s) thereof.
  • a catalyst such as palladium hydroxide on carbon, Pd/C in ether solvents such as tetrahydrofuran and alcohol such as tert-butanol, MDC, THF, toluene, CAN, water or mixture(s) thereof.
  • a catalyst such as palladium hydroxide on carbon, Pd/C in ether solvents such as tetrahydrofuran and alcohol such as tert-butanol, MDC, THF, toluen
  • a compound of formula 1-18 can be converted to compound of formula I by reaction of acid derivative ( Formula 1-18) with chlorinating agent such as thionyl chloride, Oxalylchloride using mixture of solvents such as dimethylformamide, dimethyl acetamide, dichloromethane, di chloroethane, tetrahydrofuran, benzene, toluene, halobenzene viz. 1,2 di chlorobenzene or acetonitrile, at a temperature ranging from 0°C to reflux temperature preferably at 70-80°C for a time period of 0.5 hours to 15 hours preferably for 5 hours to form acid chloride derivative.
  • chlorinating agent such as thionyl chloride, Oxalylchloride using mixture of solvents such as dimethylformamide, dimethyl acetamide, dichloromethane, di chloroethane, tetrahydrofuran, benzene, toluene, halobenzene viz. 1,
  • This acid chloride derivative can be converted to desired amide compound of formula (I) by reaction with ammonia or suitable primary, secondary amine such as methylamine, ethylamine, n-propylamine, isopropylamine, isobutylamine, n-butylamine, Cyclopropyl amine, cyclopentyl amine, cyclohexyl amine.
  • ammonia or suitable primary, secondary amine such as methylamine, ethylamine, n-propylamine, isopropylamine, isobutylamine, n-butylamine, Cyclopropyl amine, cyclopentyl amine, cyclohexyl amine.
  • Amine can be any primary or secondary alkyl amines for example, "Cl-10 alkyl” is intended to include Cl, C2, C3, C4, C5, C6, C7, C8, C9, and CIO alkyl groups, in solvents such as di chloromethane, di chloroethane, tetrahydrofuran, acetonitrile, 1,4-Dioxane, dimethylformamide, dimethylacetamide or mixture(s) thereof at temperature ranging from 0°C to reflux temperature preferably at room temperature for a time period of 0.5 hours to 10 hours preferably for 5 hours.
  • solvents such as di chloromethane, di chloroethane, tetrahydrofuran, acetonitrile, 1,4-Dioxane, dimethylformamide, dimethylacetamide or mixture(s) thereof at temperature ranging from 0°C to reflux temperature preferably at room temperature for a time period of 0.5 hours to 10 hours preferably for 5 hours.
  • a compound of formula 1-18 can be converted to a compound of formula (I) by treating compound of formula 1-18 with ammonia or suitable primary, secondary amine such as methylamine, ethylamine, n-propylamine, isopropylamine, isobutylamine, n-butylamine, Cyclopropyl, cyclopentyl, cyclohexyl, using coupling agents such as PyBOP, EDC. HC1,
  • Amine can be primary or secondary alkylalkyl amines for example , "Cl-10 alkyl” is intended to include Cl, C2, C3, C4, C5, C6, C7, C8, C9, and CIO alkyl groups, in solvents such as di chloromethane, di chloroethane, tetrahydrofuran, acetonitrile, 1,4-Dioxane, dimethylformamide, dimethylacetamide or mixture(s) thereof at temperature ranging from 0°C to reflux temperature preferably at room temperature for a time period of 0.5 hours to 15 hours preferably for 10 hours.
  • solvents such as di chloromethane, di chloroethane, tetrahydrofuran, acetonitrile, 1,4-Dioxane, dimethylformamide, dimethylacetamide or mixture(s) thereof at temperature ranging from 0°C to reflux temperature preferably at room temperature for a time period of 0.5 hours to 15 hours preferably for 10 hours.
  • a compound of formula (I) or its pharmaceutically acceptable salts can be prepared with or without isolation of intermediates. Isolation of a compound of formula (I) or its pharmaceutically acceptable salts and its intermediates may be carried out by any method known in the art such as cooling, filtration, centrifugation, washing, drying and combination thereof.
  • compound of Formula (I) or a pharmaceutically acceptable salt or a pharmaceutical composition thereof for prophylactic and/or therapeutic treatment of diseased condition in a subject suffering from Ulcerative Colitis.
  • the compound of Formula (I) or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof is envisaged to provide a subject (suffering from ulcerative colitis) with significant protection in % body weight loss, reduced DAI score, inhibited shortening of colon, decrease in cytokine levels of IL6, IL8 and IFN gamma in colon and serum tissue and reduced histopathology score.
  • compound of Formula (I) or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof for prophylactic and/or therapeutic treatment of diseased condition in a subject suffering from Ulcerative Colitis.
  • the compound of Formula (I) and its pharmaceutically acceptable salts may be formulated with pharmaceutically acceptable stabilizers, diluentsand otherexcipients.
  • suitable form of the composition may be determined by the route of administration of the composition.
  • the suitable form of the composition may include but is not limited to, injection for intravenous (bolus or infusion), intra-arterial, intraperitoneal, subcutaneous (bolus or infusion), intraventricular, intramuscular, or subarachnoidal route; tablet, capsule, gel, lozenge or liquid for oral ingestion; a solution, suspension or aerosol as sprays for inhalation; gel, spray or cream for topical application; transmucosal composition for administration via oral, nasal or rectal mucosa; by delivery in the form of a transdermal patch, subcutaneous implant, or in the form of a suppository.
  • the compounds may also be formulated in rectal compositions such as suppositories or retention enemas.
  • compositions may take the form of tablets or lozenges formulated in conventional manner.
  • the composition may be a vesicular drug delivery system such as, but not limited to bilosomes, liposomes, niosomes, transferosome, ethosomes, sphingosomes, pharmacosomes, multilamellar vesicles, microspheres and the like.
  • the compounds of Formula (I), or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof may be administered in a subject via any of the known suitable routes of drug administration. It is further envisaged that the administration of said compounds may provide a means to prevent, treat, cure and/or amelioratethe effect of diseased condition in a subject suffering fromUlcerative Colitis.
  • the routes of drug administration include but not limited to intravenous injection (bolus or infusion), intra-arterial, intra-peritoneal, subcutaneous, intraventricular, intramuscular, subarachnoidal, surgical implants, oral ingestion, inhalation, topical, and mucosal (oral, nasal and/or rectal), etc.
  • drug may be administered enterally, parenterally and/or topically.
  • Liquid dosage forms for oral administration may include but not limited to emulsions, microemulsions, solutions, suspensions, syrups and elixirs pharmaceutically acceptable.
  • the liquid dosage forms may contain inert diluents commonly used in the art.
  • Injectable preparations such as aqueous or oleaginous sterile injectable suspensions may be formulated according to known techniques by suitable dispersing or wetting agents and suspending agents commonly used in the art.
  • subject may be an animal.
  • the subject is mammalian subject.
  • the mammalian subject may be a human subject suffering from Ulcerative Colitis.
  • the human subject may be male or female coming from different ethnic groups or race, geographical regions, countries, continents and races.
  • the human subject may be selected from a group comprising of an Asian (e.g., far-east Asian, middle-east Asian, south-east Asian, north-east Asian, or Asian Indian), a Caucasian (Canadian, American, European, or Mediterranean), an African, a pacific islander, a Hispanic, etc or a general mixed population.
  • the said subject may be of any age, including newborn, neonate, infant, child, adolescent, adult, and geriatric, etc.
  • the compounds envisaged to be administered in a subject may comprise of compound of Formula I or combination of one or more salts as disclosed in Tablet.
  • the compounds envisaged by the present disclosure may be given in form of a prodrug.
  • “Prodrugs” may include any covalently bonded carriers that release an active compound of the present disclosure in vivo when such prodrug is administered to mammalian subject.
  • Prodrugs of the present invention are prepared by modifying functional groups present in the compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to give the parent compound.
  • the skilled person in the art is well aware that the prodrugs are known to enhance numerous desirable qualities of pharmaceuticals (e.g. solubility, bioavailability, manufacturing, etc.) and the compounds of the present disclosure may be delivered in prodrug form.
  • the compounds envisaged by the present disclosure may be given at a concentration sufficient to elicit a desired biological response.
  • concentration of the compound envisaged by the present disclosure may vary depending upon such factors as desired biological endpoint; the pharmacokinetics of the compound, the condition being treated, the mode of administration, age, bodyweight and health of the subject.
  • prophylactic and/or therapeutic treatment of a subject may include administration of an effective concentration of compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • the effective concentration of the compound of Formula (I) or a pharmaceutically acceptable salt thereof may be determined using pharmacokinetics and pharmacodynamic studies.
  • the compound of Formula (I) or a pharmaceutically acceptable salt thereof may be administered at a concentration in the range of 0.01 to 1000 milligram per kilogram of subject’s body weight.
  • the compounds of the disclosure may be administered as such or in a pharmaceutically acceptable form orally or parenterally at dosage levels of about 0.01 to 1000 milligram per kilogram, from 0.1 to 100 milligram per kilogram, from 0.5 to 100 milligram per kilogram or from 1 to 50 milligram per kilogram of the subject's body weight per day, once or several times per day, to obtain the desired therapeutic effect.
  • dosage levels of about 0.01 to 1000 milligram per kilogram, from 0.1 to 100 milligram per kilogram, from 0.5 to 100 milligram per kilogram or from 1 to 50 milligram per kilogram of the subject's body weight per day, once or several times per day, to obtain the desired therapeutic effect.
  • Step A 6-benzyl-l-methyl-l,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine
  • Step B 6-benzyl-8-bromo-l-methyl-l,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine
  • 6-benzyl-l-methyl-l,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine (1 mmol) was dissolved in THF (25 mL) at room temperature and to the resulting solution was added Nbromosuccinimide (1.2 mmol). The resulting suspension was stirred at room temperature for 14 hours, then quenched with aqueous saturated sodium thiosulfate solution (20 mL). The reaction was concentrated in vacuo, and the resulting residue was diluted with ethyl acetate (75 mL).
  • aqueous layer was extracted with ethyl acetate (2*100 mL) and the combined organic layers were washed with aqueous IN sodium bicarbonate solution (50 mL) and brine (50 mL), then dried over magnesium sulfate, filtered and concentrated in vacuo to provide title compound (87%), which was used further with or without purification.
  • Step C ethyl 6-benzyl-l-methyl-l,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8- carboxylate
  • 6-benzyl-8-bromo-l -methyl- l,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine (173 mmol) was added in dry tetrahydrofuran (500 mL) at -78°C and n-butyl lithium (2.5 M solution in hexane, 487 mmol) was added over a period of 2 hours. The reaction mixture was stirred for another 30 minutes at -78°C. Ethyl chloroformate (186 mmol) was added over 30 minutes and the reaction mixture was stirred for 2 hours at -60°C. The temperature was slowly increased to 30°C and mixture was allowed to stir for 12 hours at 30°C.
  • reaction mixture was then quenched with saturated solution of ammonium chloride (150 mL) at 0°C and the reaction mixture was extracted with ethyl acetate (3X300 mL). The combined organic layers were washed with water, dried over anhydrous sodium sulfate (50 g), filtered and concentrated under reduced pressure to afford a crude reaction mixture. The residue was purified by chromatography to provide the title compound (50%).
  • Step D 6-benzyl-N,l-dimethyl-l,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8- carboxamide
  • Step A l-(l-benzyl-4-chloro-lH-pyrrolo[2,3-b]pyridin-3-yl)-2,2,2-trifluoroethanone
  • Reaction mixture was filtered and purified by using Isopropanol to obtain beige to light yellow colour solid l-(l-benzyl-4- chloro-lH-pyrrolo[2,3-b]pyridin-3-yl)-2,2,2-trifluoroethanone, (125.0 g 89.6%).
  • Step B l-(4-amino-l-benzyl-5-nitro-lH-pyrrolo[2,3-b]pyridin-3-yl)-2,2,2- tri fluoroethanone
  • Step C l-(4,5-diamino-l-benzyl-lH-pyrrolo[2,3-b]pyridin-3-yl)-2,2,2-trifluoroethanone
  • Step D l-(6-benzyl-3,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridin-8-yl)-2,2,2- tri fluoroethanone
  • Step E 6-benzyl-3,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-carboxylic acid
  • reaction mass was heated at 80-85°C for 5 hours. After completion, reaction mixture was diluted with water followed by dilute HC1 and filtered.
  • Step G N-(propan-2-yl)-3,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8- carboxamide
  • Step H N-(propan-2-yl)-3,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-
  • DSS Dextran sodium sulphate
  • Dextran sodium sulphate (DSS) induced Ulcerative Colitis was used as a test compound for evaluation.
  • Test compound was evaluated on Dextran sodium sulphate (DSS) induced Ulcerative Colitis in mice by measuring, Disease activity index, proinflammatory cytokine levels in serum and colon tissue and histopathological investigation.
  • DSS Dextran sodium sulphate
  • Test compound treated groups showed reduced Disease Activity Index (DAI) score (body weight loss, stool score & bleeding score), inhibited shortening of colon length, decrease cytokine (IL-6, IL-8, IFN-Gamma) levels and reduce Histopathology scoring compared to Disease/Colitis group.
  • DAI Disease Activity Index
  • test compound(6mg/kg):3mg of Test compound was weighed and suspended in 2.5mL of water to get final concentration of 1.2mg/mL.
  • test compound (12mg/kg): 6mg of test compound was weighed and suspended in 2.5mL of water to get final concentration of 2.4 mg/mL.
  • test compound 24mg/kg 12mg of test compound was weighed and suspended with in 2.5mL of water to get final concentration of 4.8 mg/mL.
  • Reference drug-sulfasalazine 15 mg was suspended in 2.5 ml of 0.5% Na-CMC to get final concentration of 6mg/mL (Dose:30 mg/kg; Dose Volume: 05 mL/kg).
  • Group G1 and G2 animals received distilled water vehicle at dose volume of 5ml/kg.
  • Group G4, G5 and G6 animals received test compoundorally at dose of 6mg/kg, 12mg/kg and 24mg/kg respectively.
  • Colitis was induced in all the animals (except normal control) by providing 3% DSS solution in drinking water for 7 days from day 4 to day 11.
  • sulfasalazine at the dose of 30mg/kg.
  • Test compound treatment for groups G4, G5 and G6 was continued during the disease induction period.
  • DAI disease activity index
  • animals were kept for overnight fasting. Day 11, respective treatment was given.
  • serum was separated and kept in deep freezer for future analysis. After blood collection animals were humanely sacrificed, colon tissue was collected in all the animals. The colon length was measured and weighed.
  • Proximal and distal part of colon was fixed in 10% NBF, embedded in paraffin and sectioned at 4-5pm thickness used for histopathological investigation using H&E staining.
  • the middle part colon segment lOOmg of colon tissue was collected in 1ml of ice cold 1XPBS. Colon tissue was homogenized and stored in -20°C for overnight. After two freeze-thaw cycles were performed to break the cell membranes. The homogenate was centrifuged for 5 minutes at 5000 gyration. The supernatant was initially stored at -20°C and after freeze was transferred to -80°C. Serum and tissue homogenate samples were used for estimation of biomarker/cytokine analysis as for ELISA kit procedure. Body weight
  • Feed intake was calculated by formula
  • Feed intake Feed kept (gram) -Feed left (gram)
  • feed intake was calculated appropriately for each group.
  • the cumulative feed intake of groups was calculated by adding feed intake of groups during the disease induction period.
  • Group G1 normal control animals showed healthy/normal feed intake pattern.
  • Administration of 3% DSS in drinking water for group G2 showed marked decrease in cumulative feed intake.
  • Reference standard and test compound treatment groups showed marked improvement in cumulative feed intake compared to G2 disease control.
  • DAI was calculated as a composite of body weight loss, stool consistency and stool blood, was scored to analyze the potential of test compound.
  • the 3% DSS administration to G2-DSS (3%) control group was associated with significant clinical changes including weight loss, appearance of occult faecal blood and diarrhoea in mice and DAI score of was significantly elevated from day 7 to dayl 1.
  • DAI scoring of the Groups is mentioned in Table 6.
  • test compound treatment at dose of 6 and 12 mg/kg significantly reducec the DAI score from day 8 to day 11 & day 9 to day 11 respectively.
  • Test compound treatment at dose 6 mg/kg and dose 12 mg/kg groups delayed or reduced the appearance of the colitis symptoms like appearance of hemoccult and diarrhoea, which resulted in a significant reduction of DAI (Refer Figure 3).
  • DSS Dextran sodium sulphate
  • the middle part colon segment washed with PBS, lOOmg of colon tissue was weighed in 1ml of ice cold 1XPBS. Colon tissue was homogenized and stored in -20°C for overnight. After two freeze-thaw cycles were performed to break the cell membranes. The homogenate was centrifuged for 5 minutes at 5000 gyration. The supernatant was initially stored at -20°C and after freeze was transferred to -80°C.
  • Colon Length Administration of 3% DSS in drinking water for group G2 showed significant decrease p ⁇ 0.001 in colon length. Reference standard and treatment groups showed significant improvement in colon length when compared to G2; disease control (Refer Figure 4).
  • Colon Weight Administration of 3% DSS in drinking water for group G2 showed marked decrease in Colon weight. Reference standard and treatment groups showed marginal improvement in colon weight when compared to G2; disease control (Refer Figure 5).
  • DSS Dextran sodium sulphate
  • Proximal and Distal part of colon was inflated and fixed in 10% NBF, embedded in paraffin and sectioned at 4-5pm thickness used for histopathological investigation using H&E staining.
  • scoring system which is reliable to human is adapted for scoring as mentioned in Table 8. From all the parameters total score was calculated.
  • Table 9 Histopathology of colon-Colitis parameters score and Total Score Conclusion: Administration of DSS to group G2 showed the Total score 5.29 ⁇ 2.35 compared to G1 Normal control 0.43 ⁇ 0.43 indicating the mild colitis Disease induction.
  • DSS Dextran sodium sulphate
  • IL-6 was 6.30 ⁇ 1.21 pg/mL in the serum of DSS treated mice (Group 2), the IFN-Gamma concentration in the serum of TBStreated mice was391.4 ⁇ 52.4 pg/mL, (Refer Table No. 10 and 11).
  • a non- significant increase in the serum level of IL-6 and IFN-Gamma was observed in DSS group with percentage change of 45.4% and 43.1% when compared to Normal control group (Gl) which was 4.337 ⁇ 0.95 pg/mL and 273.5 ⁇ 57.3 pg/mL respectively.
  • Table 12 Effect of Test compound on Colon IL6 levels
  • Table 13 Effect of Test Compound on colon IFN-Gamma level
  • IL-6 was 94.2 ⁇ 18.6 pg/Ml in the colon of DSS treated mice(Group 2), IFN-Gamma concentration in colon of DSS treated mice was 924.9 ⁇ 157.3 pg/mL, while the IL-8 levels was 308.4 ⁇ 52.6 pg/mL of the DSS treated mice in colon tissue (Refer Table No.12, 13 and 14).
  • the compounds of formula (I) or a pharmaceutically acceptable salt thereof such as test compound showed a promising anti-inflammatory activity due to its ability to suppress the pro-inflammatory cytokines, IL-6 and IFN-Gamma of colon and serum tissue and IL-8 levels in colon tissue in a DSS-induced mice inflammation model.

Abstract

Present invention provides a means to prevent, cure, treat and/or to ameliorate the diseased condition in a subject suffering from ulcerative colitis. The said invention further provides a means for treatment of diseased condition in a subject suffering from ulcerative colitis using a compound of Formula (I) or a pharmaceutically acceptable salt thereof; wherein the compound of Formula (I) is wherein Q is a group of formula Q1 or Q2; (a)(wavy bond) represents the points of attachment; wherein R1 is -NRaRb; R2 is hydrogen or a C1-C10 alkyl group; Ra and Rbindependently represent hydrogen or a C1-C10 alkyl group.

Description

SUBSTITUTED TRICYCLIC COMPOUNDS AND THEIR USE IN ULCERATIVE COLITIS
FIELD OF INVENTION
The present invention relates to compounds of Formula (I) and/or its pharmaceutically acceptable salts thereof for use in prevention, treatment and/or cure of a diseased condition in a subject suffering from Ulcerative colitis.
BACKGROUND
Inflammatory Bowel Disease (IBD) is a broad term referring to conditions with chronic inflammation of the gastrointestinal tract. Ulcerative colitis (UC) and Crohn’s disease (CD) are two common forms of IBD that share some characteristics.
An estimated 1.2 million people in major industrialized countries suffer from ulcerative colitis and the numbers are expected to increase along with population growth. UC was first described by Samuel Wilks in 1859 and it is more common than Crohn's disease worldwide. The overall incidence and prevalence of UC is reported to be 1.2-20.3 and 7.6-245 cases per 100,000 persons/year respectively. UC has a bimodal age distribution with an incidence peak in the 2nd or 3rd decades and followed by second peak between 50 and 80 years of age. About 3-14% of patients with UC will have a family history of IBD, more commonly UC. (Ref. Mahesh Gajendran, et. al., 2019)
Ulcerative colitisis a chronic inflammatory disease affecting the large bowel and altered regulation of cell-mediated and humoral factor-mediated immune responses against intestinal constituents which have been suggested to play a role in the development of the disease. UC/IBD is associated with chronic inflammation, defined by a dysregulated response of the innate and adaptive immune systems. Chronic inflammation in Crohn’s disease is characterized by a response of helper T cells type 1 (Thl) and helper T cells type 17 (Thl7), with inadequate activity of regulatory T cells (Treg), whereas UC has generally been considered a type 2 T helper cell cytokine profile (Th2). In both diseases, many of the cytokines produced by these T cells signal through JAK receptors; therefore, JAK proteins have an important place in the signaling of inflammation in Inflammatory Bowel Disease.
Inflammation in UC occurs typically in the colon and rectum. Symptoms include the development of bloody diarrhea with or without mucus, rectal urgency, tenesmus, abdominal pain, weight loss, fatigue and extra intestinal manifestations. The key risk factors for UC include genetics, environmental factors, autoimmunity and gut microbiota.
Cytokines play a major role in pathogenesis of UC/IBD, mainly belonging to Type I and Type II cytokines receptors [i.e., IL-6, IL-5, IL-9, IL-10, IL-13, IL-12/23, IL-22, granulocyte-macrophage colony-stimulating factor (GM-CSF), IFN-gamma],
Interleukin-6 (IL-6) along with oncostatin M (OSM) and IL-11 signal through the gpl30- associated receptor family. IL-6 activates JAK1, JAK2, and TYK2 leading to STAT3 transduction, which promotes T cell proliferation, favoring the polarization of Thl7 cells. Notably, IL-6 can also promote Th2 differentiation in addition, IL-6 has other functions relevant to UC/IBD, such as regulating intestinal permeability, by its effects on tight junctions, regulating the proliferation of epithelium, and healing of wounds.
Interferon-gamma activates JAK1 and JAK2, inducing STAT1 activation, resulting in macrophage activation, Thl polarization, and increased expression of several proinflammatory cytokines. However, IFN-g also has a protective function in epithelial healing. Moreover, IFN-gamma protects from tissue destruction by inhibiting the expression of genes that code for tissue destructive factors such as matrix metalloproteinases (MMPs), serine proteases, coagulation factors, complement components, and enzymes involved in the metabolism of prostaglandin.
In past, studies have demonstrated elevated concentrations of chemokines such as IL-8 and pro-inflammatory cytokines such as TNF-a in the intestinal tissue of UC patients.
UC is diagnosed based on the combination of clinical presentation, endoscopic findings, and histology. In addition to confirming the diagnosis of UC, it is also important to define the extent and severity of inflammation, which aids in the selection of appropriate treatment and for predicting the patient's prognosis. Ileocolonoscopy with biopsy is one of the means to diagnoseUC.
A pathognomonic finding of UC is the presence of continuous colonic inflammation characterized by erythema, loss of normal vascular pattern, granularity, erosions, friability, bleeding, and ulcerations, with distinct demarcation between inflamed and non-inflamed bowel. Histopathology is the definitive tool in diagnosing UC, assessing the diseaseseverity and identifying intraepithelial neoplasia (dysplasia) or cancer. The classical histological changes in UC include decreased crypt density, crypt architectural distortion, irregular mucosal surface and heavy diffuse transmucosal inflammation, in the absence of genuine granulomas. Abdominal computed tomographic (CT) scanning is the preferred initial radiographic imaging study in UC patients with acute abdominal symptoms. As per medical reports available, the hallmark CT finding of UC is mural thickening with a mean wall thickness of 8 mm, as opposed to a 2-3 mm mean wall thickness of the normal colon. The Mayo scoring system is a commonly used index to assess disease severity and monitor patients during therapy.
For UC/IBD, currently no curative therapies are available, and in most cases, lifelong treatment is required. Nonspecific immunomodulatory drugs such as glucocorticoids, sulfasalazine/5-aminosalicylates, methotrexate, and thiopurines were among the first drugs used to treat IBD/UC. Some of the medications often used to treat ulcerative colitis include Janus kinase inhibitors such as Tofacitinib; Biologies such as Adalimumab, 5 -aminosalicylic acid (5-ASA) such as Balsalazide, Mesalamine, Olsalazine, and Sulfasalazine; Sphingosine 1 -phosphate (SIP) receptor modulators such as Ozanimod.
The introduction of biologies during the last 20 years has revolutionized the treatment of IBD, and several anti-TNF monoclonal antibodies (mAbs) (including infliximab, adalimumab, certolizumab pegol, and golimumab) are commonly used. More recently, antibodies with a different mechanism of action, such as anti-integrin a4b7 (vedolizumab) and anti-IL12/IL23 (ustekinumab), are available for clinical use. However, mAbs have limitations in terms of safety, cost, and sustained efficacy. In fact, around 10-30% of patients treated with anti-TNF are primary non-responders to therapy, and 23-46% are secondary nonresponders. For these reasons, orally available drugs are still in great need and are being developed to treat UC/IBD. Among these, the Janus Kinases (JAKs) inhibition has emerged as a therapeuticstrategy to modulate downstream cytokine signaling during immune-mediated diseases. These drugs target various cytokine signaling pathways that participate in the pathogenesis of IBD.
OBJECT OF THE INVENTION
An object of the present invention is to provide a means to prevent, cure, treat and/or to ameliorate the diseased condition in a subject suffering from inflammatory disease and/or Ulcerative colitis. An object of the present invention is to provide a compound of Formula (I) or a pharmaceutically acceptable salt thereof, for use in prophylactic and/or therapeutic treatment of diseased condition in a subject suffering from Ulcerative colitis.
An object of the present invention is to provide a compound of Formula (I) or a pharmaceutically acceptable salt thereof, for use in the manufacturing of medicament for prophylactic and/or therapeutic treatment of diseased condition in a subject suffering from Ulcerative colitis.
An object of the present invention is to provide a composition comprising of a compound of Formula (I) or a pharmaceutically acceptable salt thereof, for use in prophylactic and/or therapeutic treatment of Ulcerative colitis.
An object of the present invention is to provide a means for treatment of Ulcerative colitis, by administering a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
SUMMARY OF THE INVENTION
Present disclosure provides a means to prevent, cure, treat and/or to ameliorate the diseased condition in a subject suffering from Ulcerative colitis. More specifically, the said invention provides a compound of Formula (I) or a pharmaceutically acceptable salt thereof, for use in the manufacturing of medicament for treatment of Ulcerative Colitis. The said invention also provides a means for treatment of Ulcerative Colitis by administering to the subject a therapeutically effective amount of compound of Formula (I) or a pharmaceutically acceptable salt thereof.
Other and further aspects, features, benefits, and advantages of the present disclosure will be apparent from the following description of the presently preferred embodiments of the invention given for the purpose of disclosure.
FIGURES
Figure 1 shows Percentage Change in Body Weight (g) of the Groups
Figure 2 depicts Cumulative Feed Intake of the groups
Figure 3 illustrates DAI score of the groups
Figure 4 depicts Colon length of the groups
Figure 5 relates to Colon weight of the groups Figure 6 shows Histopathology scoring of the Groups
Figure 7 illustrates Effect of Test compound on Serum IL6 levels
Figure 8 depicts Effect of Test compound on Colon IL6 levels
Figure 9 relates to Effect of Test compound on Serum IFN-Gamma level
Figure 10 shows Effect of Test compound on colon IFN-Gamma level
Figure 11 illustrates Effect of Test compound on Colon IL8 levels
DESCRIPTION
Unless stated otherwise, the following terms and phrases as used herein are intended to have the following meanings:
The term, "a compound for use" or "a compound as described herein" means the compound or a pharmaceutically acceptable salt, thereof. Similarly, the phrase "a compound of Formula (I)" means a compound of that formula and pharmaceutically acceptable salts, thereof.
The term ‘composition’ as used herein in the present disclosure may comprise a compound of Formula (I) or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable excipient. The term ‘excipient’ or ‘excipients’ as used herein refers to inactive or usually inert substances that are added to the formulation which do not affect the therapeutic action of the active ingredient, but serve as a vehicle or medium for the active ingredient. It may be used to provide a desired consistency, to improve stability, and/or to adjust osmolality of the composition or adjust permeability. The excipients may be selected from the substances that are known to the skilled person for use in the form of compositions that are dependent on the route of administration. Exemplary excipients include diluents, carriers, binding agents, fillers lubricants, disintegrants, wetting agents, suitable coatings, stabilizers, sterilized water, physiological saline, suitable propellant cocoa butter, glycerides, suspending agents, emulsifying agents, preservatives polymers, solubilizers, cryoprotectants, lyoprotectants, bulking agent/s and/or pharmaceutically acceptable buffers or a mixture thereof. The selection of excipients for preparation of a composition of the present invention is well within the scope and understanding of the skilled person, and suitable excipients are listed in standard references such as Handbook of Pharmaceutical Excipients (Rowe RC, Sheskey P, Quinn M. Pharmaceutical Press; 2009); The Theory And Practice Of Industrial Pharmacy (Lachman, L., Lieberman, H. A., & Kanig, J. L. 1976). The Science and Practice of Pharmacy (Remington JP 2006) and Pharmaceutical Dosage Forms and Drug Delivery Systems (Allen L, Ansel HC 2013 Dec 23).
The terms ‘formulation’, ‘composition’, ‘medicament’, ‘pharmaceutical formulation’ and ‘pharmaceutical composition’ are used interchangeably and refer to preparations which are in such a form as to permit the biological activity of the active ingredients to be effective, and therefore may be administered to a subject for therapeutic use, wherein the subject is a mammal, preferably human.
The terms “administer,” “administering,” or “administration,” herein are used interchangeably and refers to implanting, absorbing, ingesting, injecting, inhaling, or otherwise introducing an inventive compound, or a pharmaceutical composition thereof.
An “effective amount” or a “therapeutically effective amount” of a compound or any active ingredient as described herein are used interchangeably and refers to an amount sufficient to elicit a desired biological response, i.e., treating the condition. As will be appreciated by those of ordinary skill in art, the effective amount of a compound may vary depending on such factors as the desired biological endpoint, the pharmacokinetics of the compound, the condition being treated, the mode of administration, and the age and health of the subject. An effective amount encompasses therapeutic and prophylactic treatment.
The term “inflammation” or “inflammatory disease” as used herein relates to any type of inflammatory immune response in a subject. The inflammation may be associated with a chronic inflammatory disease.
The term “treatment” comprises substantially curing (i.e. eliminating) the inflammation or an underlying chronic inflammatory disease, or reducing (either permanently or temporarily) the symptoms associated with the inflammation. Such symptoms may include swelling, pain, itching, heat, redness (e.g. of the skin), loss of function (e.g. of joints or limbs), and difficulty in breathing.
The terms such as “about”, “generally”, “substantially,” and the like are to be construed as modifying a term or value such that it is not an absolute. Such terms will be defined by the circumstances and the terms that they modify as those terms are understood by those skilled in the art. This includes, at the very least, the degree of expected experimental error, technique error and instrument error for a given technique used to measure a value. In an aspect of the present disclosure, there is provided a means to prevent, cure, treat and/or to ameliorate the diseased condition in a subject suffering from inflammatory disease, particularly there is provideda means to prevent, cure, treat and/or to ameliorate the diseased condition in a subject suffering from Ulcerative Colitis. In an embodiment of the present disclosure, there is provided a compound of Formula (I) and/or a pharmaceutically acceptable salt thereof, for use in the manufacturing of medicament for prophylactic and/or therapeutic treatment of diseased condition in a subject suffering from Ulcerative Colitis. The medicament having compound of Formula (I) or a pharmaceutically acceptable salt thereof may be administered in a subject for prophylactic and/or therapeutic treatment ofdiseased condition in a subject suffering from Ulcerative Colitis.
In an embodiment of the present disclosure, the compound of Formula I is
Figure imgf000008_0001
wherein Q is a group of formula QI or Q2;
Figure imgf000008_0002
/v w' (wavy bond) represents the points of attachment; wherein R| is -NRaRb;
R2 is hydrogen or a Ci-Cio alkyl group; and
Ra and Rb independently represent hydrogen or a Ci-Cio alkyl group. As used herein, the term "alkyl" is intended to include branched and straight-chain saturated aliphatic hydrocarbon groups and cycloalkyl group having the specified number of carbon atoms. For example, "Cno alkyl” is intended to include Ci, C2, C3, C4, C5, C6, C7, C8, C9, and Cio alkyl groups. Preferred alkyl groups have from 1-6, especially 1-4, carbon atoms.
Example alkyl groups include, but are not limited to, methyl (Me), ethyl (Et), propyl (e.g., n- propyl and isopropyl), butyl (e.g., n-butyl, isobutyl, t-butyl), pentyl (e.g., n-pentyl, isopentyl, neopentyl). The said alkyl may be further substituted by alkyl, halogen, amides, esters, acids, cyanide, amines.
The term "cycloalkyl" refers to cyclized alkyl groups, including monocyclic ring systems. C3- 13cycloalkyl is intended to include C3, C4, C5, C6, and C7cycloalkyl groups. Preferred cycloalkyl groups have from 3-8, especially 3-6, carbon atoms. Example cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like. The compounds of formula (I) may form salts with alkali metals such as sodium, potassium and lithium, with alkaline earth metals such as calcium and magnesium, with organic bases such as dicyclohexylamine, tributylamine, pyridine and amino acids such as arginine, lysine and the like. Such salts can be formed as known to those skilled in the art.
The compounds of formula (I) may form salts with a variety of organic and inorganic acids. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric, borates and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicyclic, sulfanilic, 2-acetoxybenzoic, fumaric, benzenesulfonic , toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, and the like. In addition, zwitterions ("inner salts") may be formed.
The skilled person would appreciate that since the compounds of present invention have more than one basic site, they have the capacity to form a salt with more than one molecule of acid. The present disclosure embodies mono di or tri salts of the compounds of this disclosure. In another embodiment of the present disclosure, it is envisaged that the compound derived from Formula (I) includes but are not limited to one or more compounds selected from Table 1 :
Figure imgf000010_0001
Figure imgf000011_0001
Figure imgf000012_0001
In an embodiment of the present disclosure, it is envisaged to prepare the compound of Formula (I) including compounds given in Table 1. The skilled person in the art may prepare the said compounds by using any of the known methods in chemistry. The said compounds may be prepared using the process as described in below scheme.
Scheme I:
Scheme I illustrates the preparation of a compound of formula (I) wherein Q, Ri and R2 are defined as above and Li and L2 represent X or leaving groups. X may be a leaving group which is either the same as that of Li or L2 or other than that of Li and L2. X may also be a group that can be easily substituted by or converted to -CORi.
Figure imgf000012_0002
In the present embodiment, the leaving group Lb L2or X is one which can be easily replaced by the desired group or atom. The leaving group may be selected from halogen atoms, alkoxy and sulfonyloxy groups. Examples of sulfonyloxy groups include, but are not limited to, alkylsulfonyloxy groups (for example methyl sulfonyloxy (mesylate group) and trifluorom ethyl sulfonyl oxy (triflate group)) and arylsulfonyloxy groups (for example /- toluenesulfonyloxy (tosylate group) and /-nitrosulfonyloxy (nosylate group)). For the purpose of the present invention L2 and X may be particularly selected from halogens such as bromo, chloro or iodoand a triflate group. The selection of X is well within the understanding and knowledge of the skilled person. In the above reactions of Scheme I, a compound of formula 1-1 is converted into a compound of formula 1-2 by a displacement reaction of a compound of formula 1-1 with ammonia solution in a suitable solvent, such as water, THF, 1,4-Di oxane, Dimethyl formamide (DMF), Dimethyl sulfoxide (DMSO) or Acetonitrile (ACN), or mixture(s) therefore at a temperature ranging from 45°C to 120°C for 0.5 hours to 20 hours to form a compound of formula 1-2.
A compound of formula 1-2 is converted to a compound of formula 1-3 by reacting a compound of formula 1-2 with a tritiating agent such as trifluoromethanesulfonic anhydride or a halogenating agent in a suitable solvent such as acetonitrile, chloroform or tetrahydrofuran at a temperature ranging from -20° C to the refluxing temperature for a time period between about 1 hour to about 10 hours.
A halogenating agent according to the present invention is a reagent that is a source of halogen. For example, the agent may be a chlorinating agent such as chlorine, thionyl chloride, N-Chlorosuccinimide, Oxalyl Chloride or a brominating agent such as bromine, NBromosuccinimide, Carbon Tetrabromide or an iodinating agent such as Iodine, Hydriodic acid or N-Iodosuccinimide. The halogenating agent may be selected according to the knowledge and understanding of skilled person.
A Sonogashira reaction with a compound of formula 1-3 and an acetylene derivative using a suitable catalyst provides a compound of formula 1-4. The reaction conditions for a Sonogashira reaction vary depending on the starting material, the solvent and the transition metal catalyst. The reaction conditions are not limited in particular as long as they are similar to the conditions of the present reactions, and the methods well known to those skilled in the art can be used. Examples of preferred solvents include acetonitrile, tetrahydrofuran, 1,4- dioxane, 1,2-dimethoxy ethane, benzene, toluene, xylene, 1 -methyl -2-pyrrolidone, N,Ndimethylformamideand dimethylsulfoxide, dichloromethane or mixture thereof. The reaction temperature should be a temperature that is sufficient to complete the coupling reaction, and is preferably from room temperature to 100°C. The present reaction can be carried out under an inert gas atmosphere, and also under a nitrogen or an argon gas atmosphere. Under the preferred reaction conditions, this reaction is completed in 1 hour to 24 hours. The transition metal catalyst is preferably a palladium complex. Examples of palladium complexes include, but not limited to palladium(II) acetate, dichlorobis(triphenylphosphine)palladium(II), tris(dibenzylideneacetone) dipalladium (0) and tetrakis(triphenylphosphine)palladium(0). Furthermore, in the present reaction, a phoshorous chelating agent such as triphenylphosphine, tri-o-tolylphosphine or tri-tert-butylphosphine may be added in order to obtain satisfactory results. Further the reaction may be accelerated using a metal halide or a quaternary ammonium salt or other such salts, preferably copper(I) iodide, lithium chloride, tetrabutylammoniumfluoride or silver(I) oxide. Preferred results can also be obtained in the presence of a base; the base used is not limited in particular as long as it is used in a coupling reaction similar to the present reaction Examples of such bases include, but not limited to diethylamine, triethylamine, N,N-diisopropylethylamine, piperidine and pyridine.
A compound of formula 1-4 can readily undergo 5-endo-dig cyclization in the presence of a base or transition metal catalyst in the presence of a suitable solvent such as alcoholic solvents or THF or DMA to provide a compound of formula 1-5. Exemplarily the base may be selected from Potassium tert-butoxide, Lithium hydride, Lithium Aluminium hydride and n-butyl lithium and the transition metal catalyst may be selected from Palladium and a copper catalyst.
A compound of formula 1-5 can be optionally protected by treating it with a protecting group to provide a compound of formula 1-6.
Exemplarily a compound of formula 1-5 is converted to the corresponding compound of formulaI-6, wherein R3 is benzenesulfonyl or benzyl, by treating the compound of formula I- 5 with benzenesulfonyl chloride, benzyl chloride or benzylbromide in the presence of a base, such as sodium hydride or potassium carbonate, and a polar aprotic solvent, such as dimethylformamide or tetrahydrofuran. The reaction mixture is stirred at a temperature between about 0°C. to about 70°C, preferably about 30°C, for a time period between about 1 hour to about 3 hours, preferably about 2 hours.
R3 is a protecting group such as benzenesulfonyl, substituted benzenesulfonyl, methylsulfonyl, benzyl or carbamate protecting groups such as Boc (t-Butyloxycarbonyl) and CBz (carboxybenzyl) or other groups such as benzoyl, iso-butanoyl, acetyl, phenoxyacetyl, 4- (t-butyl)benzoyl, 4-(t-butyl)phenoxyacetyl, 4-(methoxy)benzoyl, 2-(4-nitrophenyl) ethyl oxycarbonyl, 2-(2,4-dinitrophenyl)ethyloxy-carbonyl, 9-fluorenylmethoxycarbonyl, diphenylcarbamoyl or formamidine groups. Particularly preferred are the benzoyl, isobutanoyl, 4-(t-butyl)benzoyl, 2-(4-nitro-i5 phenyl)ethyloxycarbonyl, 2-(2,4- dinitrophenyl)ethyl-oxycarbonyl, 9-fluorenylmethoxycarbonyl, 4-(methoxy)-benzoyl or para- (t-butyl)phenoxyacetyl, para-nitrophenyl-2-ethyloxycarbonyl group or2-N-acetyl with the 6- O-diphenylcarbamoyl group. Compounds of formula 1-5 and 1-6 can be converted to a compound of formula 1-8 and 1-7, respectively in a similar way as the process described for the preparation of a compound of formula 1-3.
Compounds of formula 1-8 can be converted into compounds of formula (I) by a process known to the person skilled in the art. Such process may include converting X of formula 1-8 directly to an amide group or via formation of ester, anhydride, aldehyde, ketone, cyanide, acid or any such group which can be converted to an amide group which is well within the understanding and knowledge of the skilled person.
For example, when X is converted to an ester group and successively converted to an amide, compounds of formula 1-8 can be treated with an esterifying agent in the presence of a base in a polar aprotic solvent like THF, 1,4-Dioxane, DMF, DMSO and ACN at -75°C to 100°C temperature for 0.5 hours to 20 hours which leads to formation of ester derivative. The ester derivative on reaction with a trialkylaluminium (like, trimethylaluminium) and required amine derivatives or ammonia solution in the presence of solvents like Toluene, chloroform, methanol, ethanol, THF, 1,4-Dioxane, DMF, DMSO and ACN at -10°C to 100°C temperature for 0.5 hours to 20 hours gives an amide having formula I.
A compound of formula 1-7 can be converted to a compound of formula I-9using a similar process that may be used for conversion of a compound of formula 1-8 to a compound of formula I.
A compound of formula 1-9 can be converted into a compound of formula I by cleaving the protecting group R3. Protecting groups of a compound of formula 1-9 can be cleaved by deprotecting agents as understood by the skilled person to obtain a compound of formula I. Examples of deprotecting agents for an amino protective group are acids such as trifluoroacetic acid, trichloroacetic acid, dichloroacetic acid p-toluenesulfonic acid or bases such as alkali or alkaline bases. For example, fora compound of formula 1-9 wherein R3 is benzenesulfonyl, the deprotection is carried out by treating 1-9 with an alkali base, such as sodium hydroxide or potassium hydroxide, sodium carbonate, potassium carbonate, potassium tert-butoxide, sodiumtert-butoxide in an alcohol solvent, such as methanol or ethanol, or mixed solvents, such as alcohol/tetrahydrofuran or alcohol/water. The reaction is carried out at room temperature or to reflux temperature for a time period between about 15 minutes to about 1 hour, preferably 30 minutes. When R3 is benzyl, deprotection is eitherconducted by treating 1-9 with sodium in ammonia at a temperature of about -78° C for a time period between about 15 minutes to about 1 hour or by using hydrogen and a catalyst, such as palladium hydroxide on carbon, Pd/C, Raney Nickel, Raney Nickel in combination with NH2-NH2 or Hydrogen. Other suitable deprotecting agents are Lewis acids, such as, for example boron trifluorideetherate or zinc bromide in dichloromethane/isopropanol, aq. HC1, aq. HBr, HBr in acetic acid, sulfuric acid.
In another embodiment of the present disclosure, it is envisaged to prepare the compound of Formula (I) including compounds given in Table 1. The skilled person in the art may prepare the said compound by using any of the known methods in chemistry. The said compounds may be prepared using the process as described in below scheme.
Scheme II
Scheme II illustrates the preparation of a compound of formula (I) wherein Q, Ri R2, R3 and X as defined in aforementioned embodiment. R represents alkoxy (-OR) or CX3, Z is NO2.
Figure imgf000016_0001
In the above reactions of Scheme II, a compound of formula I- 10 can be converted to the corresponding compound of formula 1-11, by treating the compound of formula 1-10 with protecting group R3 such as benzenesulfonyl chloride, benzyl chloride or benzylbromide in the presence of a base, such as sodium hydride, potassium carbonate, sodium hydroxide, potassium hydroxide or cesium carbonate or alkyl lithium such as n-butyl lithium, secondary butyl lithium, tertiary butyl lithium or lithium diisopropyl amide. Such reaction may be carried out in solvent such as dimethylformamide, dimethylacetamide, tetrahydrofuran, hexamethyl phosphoramide, dimethyl sulfoxide, 1,4-Dioxane, acetonitrile, water, dichloromethane, Toluene, DMSO or mixture(s) therefore. The reaction mixture is stirred at a temperature between about 0°C. to about 70°C., preferably about 10°C, for a time period between about 1 hour to about 10 hours, preferably about 4 hours. R3 is a protecting group defined as above.
Compounds of formula 1-11 can be converted to a compound of formula 1-12 by reacting a compound of formula I- 11 with an acylating agent such as trifluoroacetic anhydride, tri chloroacetyl chloride , acid halides, acid anhydrides in a suitable solvent such as acetonitrile, chloroform, n-methyl pyrrolidone, toluene, tetrahydrofuran, dimethylformamide, dimethylsulfoxide, dimethylacetamide 1,4-Dioxane chlorinated alkyl or aryl solvents such as dichloromethane or chlorobenzene, di chlorobenzene or di chloroethane or mixture(s) therefore at a temperature ranging from -20°C to the refluxing temperature for a time period between about 1 hour to about 15 hours preferably at 65-75°Cfor 4-5 hours.
A compound of formula 1-12 can be converted to a compound of formula 1-13 by treating compound of formula 1-12 with nitrating agents such as alkyl ammonium nitrate for example, tetrabutyl ammonium nitrate or tetramethyl ammonium nitrate and using trifluoroacetic anhydride in solvents such as dichloromethane, toluene, acetonitrile, tetrahydrofuran, chlorobenzene, nitrobenzene, di chloroethane 1,4-Dioxane, acetonitrile, water, dimethylsulfoxide or mixture(s) therefore, at a temperature ranging from -10°C to 100°C for a time period between about 1 hour to about 30 hours preferably for 5 hours. A compound of formula I- 13 can be converted to a compound of formula 1-14 by reaction with ammonia or with primary amines such as methyl amine, ethyl amine, isopropyl amine, npropyl amine, isobutylamine or n-butylamine in suitable solvents such as tetrahydrofuran, dichloromethane, 1,4 dioxane, toluene, dimethylformamide, water, alcoholic solvents, DMSO, acetonitrile or mixture(s) thereof at a temperature ranging from -10°C to the refluxing temperature for a time period between about 1 hour to about 25 hours, preferably for 8-10 hours.
A compound of formula I-14can be converted to compound of formula 1-15 by reduction of nitro group using metal catalyst such as palladium on carbon, Raney nickel in combination with NH2-NH2 or Hydrogen, iron/ammonium chloride, platinum on carbon, zinc/ammonium chloride, Fe/AcOH or sodium dithionite in suitable alcoholic solvents such as methanol, ethanol or water or cyclic/acyclic ethers such as tetrahydrofuran or 1,4-dioxane or acetonitrile and water or in mixture of suitable alcoholic solvents such as methanol, ethanol, or cyclic/acyclic ethers such as tetrahydrofuran or 1,4-dioxane or acetonitrile and water at temperature ranging from -10°C to reflux temperature, preferably at room temperature for time period of 1 to 10 hours. A compound of formula 1-15 is optionally converted to compound of formula I- 15a by treating compound of formula 1-15 with alkylating agents or treating with aldehydes, ketones followed by reduction by the methods known to person skilled in the art.
A compound of formula 1-15 or 1-15 a can be converted to compound of formula 1-16 by cyclization methods using reagents such as triethylorthoformate and acid catalyst viz para toluene sulphonic acid or dimethylformamide or formic acid and metal catalyst such as zinc acetate, using solvents such as toluene, halobenzene such as chlorobenzene, 1,2 di chlorobenzene, dimethylformamide, dimethylacetamide, tetrahydrofuran, acetonitrile, 1,4- dioxane, water, acetic acid, formic acid, formamide or mixture(s) thereof at a temperature ranging from room temperature to reflux temperature preferably at 0°C-100°C for period of 1 to 10 hours preferably for 5 hours.
A compound of formula 1-16 can be converted to compound of formula 1-17 by hydrolysis using alkali hydroxide such as sodium hydroxide, potassium hydroxide or lithium hydroxide or aqueous solution thereof or any other reagents as understood by the skilled person in suitable alcoholic solvents such as methanol or ethanol or water or in mixture of suitable alcoholic solvents such as methanol, ethanol, propanol, butanol, iso-butanol or cyclic/acyclic ethers such as tetrahydrofuran or 1,4-Dioxane or acetonitrile and water to obtain a compound of formula 1-17, at a temperature ranging from room temperature to reflux temperature preferably at a temperature 80°C for time period of 30 minutes to 10 hours.
A compound of formula 1-17 can be converted into a compound of formula 1-18 by cleaving the protecting group R3. Protecting groups of a compound of formula 1-17 can be cleaved by deprotecting agents as understood by the skilled person to obtain a compound of formula (I).
Examples of deprotecting agents for an amino protective group are acids such astrifluoroacetic acid, trichloroacetic acid, dichloroacetic acid p-toluenesulfonic acid, HC1, HBr, H2SO4 or bases such as alkali or alkaline bases. For example, for a compound of formula 1-17 wherein R3 is benzenesulfonyl, the deprotection is carried out by treating 1-17 with an alkali base, such as sodium hydroxide or potassium hydroxide, sodium carbonate, potassium carbonate, cesium carbonate in an alcohol solvent, such as methanol or ethanol, or mixed solvents, such as alcohol/tetrahydrofuran or alcohol/water, MDC, THF, toluene, CAN, water or mixture(s) thereof. The reaction is carried out at room temperature to reflux temperature for a time period between about 15 minutes to about 1 hour, preferably 30 minutes. When R3 is benzyl, deprotection is either conducted by treating 1-17 with sodium in ammonia at a temperature of about -78°C for a time period between about 15 minutes to about 10 hour or by using hydrogen and a catalyst, such as palladium hydroxide on carbon, Pd/C in ether solvents such as tetrahydrofuran and alcohol such as tert-butanol, MDC, THF, toluene, CAN, water or mixture(s) thereof. Other suitable deprotecting agents are Lewis acids, such as, for example boron trifluorideetherate or zinc bromide in dichloromethane/isopropanol. HC1, HBr, H2SO4.
A compound of formula 1-18 can be converted to compound of formula I by reaction of acid derivative ( Formula 1-18) with chlorinating agent such as thionyl chloride, Oxalylchloride using mixture of solvents such as dimethylformamide, dimethyl acetamide, dichloromethane, di chloroethane, tetrahydrofuran, benzene, toluene, halobenzene viz. 1,2 di chlorobenzene or acetonitrile, at a temperature ranging from 0°C to reflux temperature preferably at 70-80°C for a time period of 0.5 hours to 15 hours preferably for 5 hours to form acid chloride derivative. This acid chloride derivative can be converted to desired amide compound of formula (I) by reaction with ammonia or suitable primary, secondary amine such as methylamine, ethylamine, n-propylamine, isopropylamine, isobutylamine, n-butylamine, Cyclopropyl amine, cyclopentyl amine, cyclohexyl amine. Amine can be any primary or secondary alkyl amines for example, "Cl-10 alkyl” is intended to include Cl, C2, C3, C4, C5, C6, C7, C8, C9, and CIO alkyl groups, in solvents such as di chloromethane, di chloroethane, tetrahydrofuran, acetonitrile, 1,4-Dioxane, dimethylformamide, dimethylacetamide or mixture(s) thereof at temperature ranging from 0°C to reflux temperature preferably at room temperature for a time period of 0.5 hours to 10 hours preferably for 5 hours.
A compound of formula 1-18 can be converted to a compound of formula (I) by treating compound of formula 1-18 with ammonia or suitable primary, secondary amine such as methylamine, ethylamine, n-propylamine, isopropylamine, isobutylamine, n-butylamine, Cyclopropyl, cyclopentyl, cyclohexyl, using coupling agents such as PyBOP, EDC. HC1,
DCC, HoBt or coupling agents known to person skilled in the art. Amine can be primary or secondary alkylalkyl amines for example , "Cl-10 alkyl” is intended to include Cl, C2, C3, C4, C5, C6, C7, C8, C9, and CIO alkyl groups, in solvents such as di chloromethane, di chloroethane, tetrahydrofuran, acetonitrile, 1,4-Dioxane, dimethylformamide, dimethylacetamide or mixture(s) thereof at temperature ranging from 0°C to reflux temperature preferably at room temperature for a time period of 0.5 hours to 15 hours preferably for 10 hours. A compound of formula (I) or its pharmaceutically acceptable salts can be prepared with or without isolation of intermediates. Isolation of a compound of formula (I) or its pharmaceutically acceptable salts and its intermediates may be carried out by any method known in the art such as cooling, filtration, centrifugation, washing, drying and combination thereof.
In an embodiment of the present disclosure, it is envisaged to use compound of Formula (I) or a pharmaceutically acceptable salt or a pharmaceutical composition thereof for prophylactic and/or therapeutic treatment of diseased condition in a subject suffering from Ulcerative Colitis.
The compound of Formula (I) or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof is envisaged to provide a subject (suffering from ulcerative colitis) with significant protection in % body weight loss, reduced DAI score, inhibited shortening of colon, decrease in cytokine levels of IL6, IL8 and IFN gamma in colon and serum tissue and reduced histopathology score.
In an embodiment of the present disclosure, it is envisaged to use compound of Formula (I) or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof for prophylactic and/or therapeutic treatment of diseased condition in a subject suffering from Ulcerative Colitis.
In an embodiment of the present disclosure, the compound of Formula (I) and its pharmaceutically acceptable salts may be formulated with pharmaceutically acceptable stabilizers, diluentsand otherexcipients. As understood by the skilled person in art the suitable form of the composition may be determined by the route of administration of the composition. Therefore the suitable form of the composition may include but is not limited to, injection for intravenous (bolus or infusion), intra-arterial, intraperitoneal, subcutaneous (bolus or infusion), intraventricular, intramuscular, or subarachnoidal route; tablet, capsule, gel, lozenge or liquid for oral ingestion; a solution, suspension or aerosol as sprays for inhalation; gel, spray or cream for topical application; transmucosal composition for administration via oral, nasal or rectal mucosa; by delivery in the form of a transdermal patch, subcutaneous implant, or in the form of a suppository. The compounds may also be formulated in rectal compositions such as suppositories or retention enemas. For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner. The composition may be a vesicular drug delivery system such as, but not limited to bilosomes, liposomes, niosomes, transferosome, ethosomes, sphingosomes, pharmacosomes, multilamellar vesicles, microspheres and the like.
In an embodiment of the present disclosure, the compounds of Formula (I), or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof may be administered in a subject via any of the known suitable routes of drug administration. It is further envisaged that the administration of said compounds may provide a means to prevent, treat, cure and/or amelioratethe effect of diseased condition in a subject suffering fromUlcerative Colitis.
The routes of drug administration include but not limited to intravenous injection (bolus or infusion), intra-arterial, intra-peritoneal, subcutaneous, intraventricular, intramuscular, subarachnoidal, surgical implants, oral ingestion, inhalation, topical, and mucosal (oral, nasal and/or rectal), etc. In a preferred embodiment of the present disclosure, drug may be administered enterally, parenterally and/or topically. Liquid dosage forms for oral administration may include but not limited to emulsions, microemulsions, solutions, suspensions, syrups and elixirs pharmaceutically acceptable. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art. Injectable preparations such as aqueous or oleaginous sterile injectable suspensions may be formulated according to known techniques by suitable dispersing or wetting agents and suspending agents commonly used in the art.
In an embodiment of the present disclosure, subject may be an animal. In preferred embodiment, the subject is mammalian subject. In particular, the mammalian subject may be a human subject suffering from Ulcerative Colitis. The human subject may be male or female coming from different ethnic groups or race, geographical regions, countries, continents and races. For example, the human subject may be selected from a group comprising of an Asian (e.g., far-east Asian, middle-east Asian, south-east Asian, north-east Asian, or Asian Indian), a Caucasian (Canadian, American, European, or Mediterranean), an African, a pacific islander, a Hispanic, etc or a general mixed population. The said subject may be of any age, including newborn, neonate, infant, child, adolescent, adult, and geriatric, etc.
In an embodiment of the present disclosure, the compounds envisaged to be administered in a subject may comprise of compound of Formula I or combination of one or more salts as disclosed in Tablet. In an embodiments, it is further envisaged to use one or more compounds selected from ‘N-(propan-2-yl)-l,6-dihydroimidazo[4,5-d]pyrrolo[2,3- b]pyridine-8-carboxamide hydrochloride’ and ‘N-(propan-2-yl)-3,6-dihydroimidazo[4,5- d]pyrrolo[2,3-b]pyridine-8-10carboxamide hydrochloride’ independently or in combination thereof.
In an embodiment of the present disclosure, the compounds envisaged by the present disclosure may be given in form of a prodrug. “Prodrugs” may include any covalently bonded carriers that release an active compound of the present disclosure in vivo when such prodrug is administered to mammalian subject. Prodrugs of the present invention are prepared by modifying functional groups present in the compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to give the parent compound. The skilled person in the art is well aware that the prodrugs are known to enhance numerous desirable qualities of pharmaceuticals (e.g. solubility, bioavailability, manufacturing, etc.) and the compounds of the present disclosure may be delivered in prodrug form.
In an embodiment of the present disclosure, the compounds envisaged by the present disclosure may be given at a concentration sufficient to elicit a desired biological response. As understood by the skilled person in art, the concentration of the compound envisaged by the present disclosure may vary depending upon such factors as desired biological endpoint; the pharmacokinetics of the compound, the condition being treated, the mode of administration, age, bodyweight and health of the subject.
In an embodiment of the present disclosure, prophylactic and/or therapeutic treatment of a subject may include administration of an effective concentration of compound of Formula (I) or a pharmaceutically acceptable salt thereof. As understood by the skilled person in art, the effective concentration of the compound of Formula (I) or a pharmaceutically acceptable salt thereof may be determined using pharmacokinetics and pharmacodynamic studies. Particularly, the compound of Formula (I) or a pharmaceutically acceptable salt thereof may be administered at a concentration in the range of 0.01 to 1000 milligram per kilogram of subject’s body weight. In certain ways of embodiment, the compounds of the disclosure may be administered as such or in a pharmaceutically acceptable form orally or parenterally at dosage levels of about 0.01 to 1000 milligram per kilogram, from 0.1 to 100 milligram per kilogram, from 0.5 to 100 milligram per kilogram or from 1 to 50 milligram per kilogram of the subject's body weight per day, once or several times per day, to obtain the desired therapeutic effect. EXAMPLES
The following examples are provided to illustrate the invention. Examples provided herein are exemplary in nature and do not limit the scope of invention in any manner.
Example 1: Preparation of N-(propan-2-yl)-l,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b] pyridine-8-carboxamide hydrochloride
Figure imgf000023_0001
Ethanolic hydrochloride solution was added to the solution of N-(propan-2-yl)-l,6- dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-carboxamide (10 mmol) in 50mL ethanol. The reaction was stirred for 3-4 hours at ambient temperature. Reaction mass was concentrated under reduced pressure. lOmL ethanol was added to the residue, stirred and reaction mass was concentrated under reduced pressure. Obtained solid was dried under vacuum to afford white to off while solid of the title compound (Yield = 95%).
15 1H NMR (400 MHz, DMSO-D6) 8: 1.25 (d, 6 H), 4.23 (m, 1 H), 7.99 (s, 1 H), 8.34 (s, 1H), 8.57 (s, 1H),8 8.7 (bslH), 11.2 (br. S, 1H), 12.9 (br. S, 2H).
Preparation of an Intermediate:6-benzyl-N,l-dimethyl-l,6-dihydroimidazo[4,5- d]pyrrolo[2,3-b]pyridine-8-carboxamide
Figure imgf000023_0002
Step A: 6-benzyl-l-methyl-l,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine
Figure imgf000024_0002
In a round bottom flask, sodium hydride (0.3 mole) was added in a DMF (5volume) solvent, 1- methyl-l,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine (0.1 mole) was slowly added at 5-15°C to a flask, the resulting suspension was stirred at room temperature for 1 hour, then benzyl bromide (0.12 mole) was slowly added at 5-15°C. The reaction mass was warmed to room temperature and stirred for 1-3 hours. The reaction was monitored on TLC. The reaction mass was cooled after completion of the reaction, methanol (1 volume) was added in the reaction mass at 5-15°C and stirred for 10 minutes. Ammonium chloride (25 volume) solution was added in the reaction mass and stirred for 30 minutes. The reaction mass was extracted with ethyl acetate (3* 5 volume). Combined organic layers were washed with water (3* 5 volume) and brine (1 volume), then dried over magnesium sulfate, filtered and concentrated in vacuo to provide the title compound (86%).
Step B: 6-benzyl-8-bromo-l-methyl-l,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine
Figure imgf000024_0001
6-benzyl-l-methyl-l,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine (1 mmol) was dissolved in THF (25 mL) at room temperature and to the resulting solution was added Nbromosuccinimide (1.2 mmol). The resulting suspension was stirred at room temperature for 14 hours, then quenched with aqueous saturated sodium thiosulfate solution (20 mL). The reaction was concentrated in vacuo, and the resulting residue was diluted with ethyl acetate (75 mL). The aqueous layer was extracted with ethyl acetate (2*100 mL) and the combined organic layers were washed with aqueous IN sodium bicarbonate solution (50 mL) and brine (50 mL), then dried over magnesium sulfate, filtered and concentrated in vacuo to provide title compound (87%), which was used further with or without purification.
Step C: ethyl 6-benzyl-l-methyl-l,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8- carboxylate
Figure imgf000025_0001
6-benzyl-8-bromo-l -methyl- l,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine (173 mmol) was added in dry tetrahydrofuran (500 mL) at -78°C and n-butyl lithium (2.5 M solution in hexane, 487 mmol) was added over a period of 2 hours. The reaction mixture was stirred for another 30 minutes at -78°C. Ethyl chloroformate (186 mmol) was added over 30 minutes and the reaction mixture was stirred for 2 hours at -60°C. The temperature was slowly increased to 30°C and mixture was allowed to stir for 12 hours at 30°C. The progress of the reaction was monitored by TLC. The reaction mixture was then quenched with saturated solution of ammonium chloride (150 mL) at 0°C and the reaction mixture was extracted with ethyl acetate (3X300 mL). The combined organic layers were washed with water, dried over anhydrous sodium sulfate (50 g), filtered and concentrated under reduced pressure to afford a crude reaction mixture. The residue was purified by chromatography to provide the title compound (50%).
Step D: 6-benzyl-N,l-dimethyl-l,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8- carboxamide
Figure imgf000026_0001
A solution of trimethylaluminium (2 M in toluene, 1.2 mmol) was added dropwise (exothermic) to a solution of methylamine (2 M in toluene, 1.2 mmol) in dioxane (7.5 mL) and the resulting mixture was stirred at room temperature for 1 hour. Then a solution of ethyl 6-benzyl-l -methyl- l,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-carboxylate (0.3 mmol) in dioxane (4 mL) was added. The resulting mixture was then heated at 85-95°C for 3hours and then cooled to room temperature and then poured into water and extracted with MDC which was then washed with brine, dried over sodium sulfate and evaporated. Purification by chromatography (SiO2, MDC:MeOH=90: 10) afforded the title compound as a white solid. (70%).
Example 2: Preparation of N-(propan-2-yl)-3,6-dihydroimidazo[4,5-d]pyrrolo[2,3- b]pyridine-8-carboxamide hydrochloride
Figure imgf000026_0002
Step A: l-(l-benzyl-4-chloro-lH-pyrrolo[2,3-b]pyridin-3-yl)-2,2,2-trifluoroethanone
Figure imgf000027_0001
To a stirred suspension of sodium hydride (39.3 g, 1638.5 mmol, 60%) in dimethylacetamide (500mL) was added a solution of 4-chloro-7-aza indole (100g, 655.4 mmol) in dimethylacetamide (150 mL) at 0-5°C followed by benzyl bromide (134.5 g, 786.5mmol). The resultant reaction mixture was stirred for 4 hours and then quenched with 100 mL of methanol followed by saturated ammonium chloride (500mL) and extracted with ethylacetate. The organic layer was evaporated under reduced pressure to afford brown to yellow colour liquid, l-benzyl-4-chloro-lH-pyrrolo [2,3 -b]pyri dine (180g). The above compound was dissolved in in dimethylformamide (700 mL) and then was added trifluoroacetic anhydride (129.8 g, 618.0 mmol). The resulting reaction mixture was heated at 70-75°C for 3 hours. Reaction mixture was cooled to 10-15°C and was added ice cold water (500 mL) followed by saturated aqueous sodium bicarbonate. Reaction mixture was filtered and purified by using Isopropanol to obtain beige to light yellow colour solid l-(l-benzyl-4- chloro-lH-pyrrolo[2,3-b]pyridin-3-yl)-2,2,2-trifluoroethanone, (125.0 g 89.6%).
1HNMR (400 MHz, DMSO-d6): 8 9.03 (S,1H), 8 8.38 (m 1H), 8 7.46 (m, 1H), 8 7.34(m,5H), 8 5.66 (S,2H).
Step B: l-(4-amino-l-benzyl-5-nitro-lH-pyrrolo[2,3-b]pyridin-3-yl)-2,2,2- tri fluoroethanone
Figure imgf000027_0002
To a stirred solution of l-(l-benzyl-4-chloro-lH-pyrrolo[2,3-b]pyridin-3-yl)-2,2,2- trifluoroethanone (100 g, 295.2 mmol) in dichloromethane (2500mL) was added tetrabutylammonium nitrate (224.7g, 738.0 mmol) in portions followed by dropwise addition of trifluoroacetic anhydride (155 g, 738.0 mmol) at 0°C. The reaction mixture was stirred for 5 hours at room temperature. Organic layer was washed with water and concentrated under reduced pressure to afford yellowsolid, l-(l-benzyl-4-chloro-5-nitro-lHpyrrolo[2,3- b]pyridin-3-yl)-2,2,2 trifluoroethanonene (100 g 88.5%).
To a stirred solution of l-(l-benzyl-4-chloro-5-nitro-lH-pyrrolo [2,3 -b]pyri din-3 -yl)-2, 2,2- trifluoroethanone (100 g, 260.6mmol) in di chloromethane (500 mL) was purged ammonia gas till completion of reaction on TLC. Solvent was removed under reduced pressure. The residue was dissolved in dichloromethane (300 mL), cooled to 5-10°C and filtered. The obtained wet solid was dried under vacuum to afford yellow solid, l-(4-amino-l-benzyl-5- nitro-lHpyrrolo[2,3-b]pyridin-3-yl)-2,2,2-trifluoroethanone (85 g,89.5%) 1HNMR (400 MHz, DMSO-d6): 8 9.04 (S,1H), 8 8.94(m 1H), 8 8.73 (S, 1H), 8 7.30 (m,6H), 8 5.57 (S,2H).
Step C: l-(4,5-diamino-l-benzyl-lH-pyrrolo[2,3-b]pyridin-3-yl)-2,2,2-trifluoroethanone
Figure imgf000028_0001
To a stirred solution of l-(4-amino-l-benzyl-5-nitro-lH-pyrrolo[2,3-b]pyridin-3-yl)-2,2,2- trifluoroethanone (85 g, 233.3 mmol) in mixture of methanol: tetrahydrofuran (1500 mL, 1 :0.5) was added Raney Nickel (21.2 g, 25.0 % w/w) followed by dropwise addition of hydrazine hydrate ( 59.5 mL, 0.70 w/v) and reaction mixture was stirred for 1 hour at room temperature. After completion, reaction mixture was filtered through hyflo bed and washed with methanol (400 mL). The filtrate was concentrated under reduced pressure and obtained was purified by water (500 mL), filtered and dried under reduced pressure to afford brown colour solid l-(4,5-diamino-l-benzyl-lH-pyrrolo[2,3-b]pyridin-3-yl)-2,2,2-trifluoroethanone (71.0gm 90.8%) 1HNMR (400 MHz, DMSO-d6): 8 8.59 (d,lH), 8 7.66 (s 1H), 8 7.33 (m, 4H), 8 7.26 (m,lH), 8 6.56 (s,2H), 8 5.45 (s,2H), 8 4.47 (s,2H).
Step D: l-(6-benzyl-3,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridin-8-yl)-2,2,2- tri fluoroethanone
Figure imgf000029_0001
To stirred suspension of l-(4,5-diamino-l-benzyl-lH-pyrrolo[2,3-b]pyridin-3-yl)-2,2,2- trifluoroethanone (70g, 209.4 mmol) in toluene (700 mL) was added triethylorthoformate (96.7 mL, 418.8 mmol) and para-toulenesulfonic acid monohydrate (8.0 g, 41.88 mmol). The resulting reaction mixture was heated at 80-85°C for 5hours. After completion, reaction mixture was concentrated under reduced pressure. To the obtained residue was added water (700 mL), stirred at room temperature and filtered an dried to afford l-(6-benzyl-3,6- dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridin-8-yl)-2,2,2-trifluoroethanone (65g, 90.1%). 1HNMR (400 MHz, DMSO-d6): 8 12.51(bs,lH), 8 8.89 (m 2H), 8 8.29 (t, 1H), 8 7.31 (m,5H), 8 5.72(s,2H).
Step E: 6-benzyl-3,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-carboxylic acid
Figure imgf000029_0002
To the solution of sodium hydroxide (151g, 3775 mmol) in water (945 mL) was added l-(6- benzyl-3,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridin-8-yl)-2,2,2 trifluoroethanone (65 g, 188.8 mmol), reaction mass was heated at 80-85°C for 5 hours. After completion, reaction mixture was diluted with water followed by dilute HC1 and filtered. The obtained wet cake was dried under vacuum to afford beige to light brown colour solid 6-benzyl-3,6- dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8- carboxylic acid, (50g, 90.5% ) 1HNMR (400 MHz, DMSO-d6): 8 12.2(s,lH), , 8 8.75(s, 1H), 8 8.22(s,lH),8 8.17 (s, 1H) 8 7.27(m,5H), 8 5.61(s,2H) Step F: 3,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-carboxylic acid
Figure imgf000030_0001
To the solution of liquid ammonia (750 mL) was added sodium metal (32.8 g, 1368.5mmol) lot wise. To the resulting reaction mixture, was added tertiary butanol (50 mL), Tetrahydrofuran (500 mL) followed by 6-benzyl-3,6-dihydroimidazo[4,5-d] pyrrolo [2,3- b]pyridine-8-carboxylic acid (50 g, 171.1 mmol). Then reaction mixture was stirred at -60°C to -30°C for 4 hours and quenched with methanol (50 mL) and water (50 mL). Solvent was evaporated under reduced pressure. Then was added water (100 mL) to the residue followed by HC1 and stirred. The reaction mixture was filtered and wet solid was dried under reduced pressure to afford beige colour solid 3,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8- carboxylic acid, (32g, 91.4%).
1HNMR (400 MHz, DMSO-d6): 8 12.39 (bs,lH), 8 12.07 (bs 1H), 8 8.66(s, 1H), 8 8.14(d,lH), 8 7.94 (s,lH).
Step G: N-(propan-2-yl)-3,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8- carboxamide
Figure imgf000030_0002
To a solution of 3,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-carboxylic acid (30.0 g, 148.4 mmol) in dimethylformamide (15 mL) was added thionyl chloride (300 mL) and reaction mixture was heated to 65-70°C and stirred for 4 hours. After completion, reaction mixture was concentrated under reduced pressure to obtain acid chloride (30 g) which was used as such for further reaction. Above acid chloride derivative(30 g) was taken into dichloromethane (300 mL), cooled to 5-10°C and added isopropyl amine (300 mL). The resulting reaction mixture was stirred for 5hours at room temperature. After completion, reaction mass was concentrated under reduced pressure, added water (150 mL) and filtered. The obtained wet solid was dried under vacuum to afford beige to off white color solid N- (propan-2-yl)-3,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-carboxamide
(29.0g, 80.3%) 1HNMR (400 MHz, DMSO-d6): 8 13.04 (bs,lH), 8 12.19 (t, 1H), 8 10.03(d,lH), 8 8.59(d,2H), 8 8.07(m,lH), 8 4.19(m,lH), 8 1.22(td,6H).
Step H: N-(propan-2-yl)-3,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-
Figure imgf000031_0001
To a solution of N-(propan-2-yl)-3,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8- carboxamide (25.0 g, 102.8 mmol) in isopropanol (175 mL) was added solution of HC1 in isopropanol at 10-15°C. The resultant reaction mixture was stirred at 50-55°C 2hours. After completion, reaction mixture was concentrated under reduced pressure. To the residue obtained was added water (250 mL) and stirred for 1 hour at room temperature, filtered and dried under vacuumto afford beige colour solid N-(propan-2-yl)-3,6-dihydroimidazo[4,5- d]pyrrolo[2,3- b]pyridine-8-carboxamide hydrochloride, (25 g, 87.0%)
1HNMR (400 MHz, DMSO-d6): 8 13.66 (bs,lH), 8 12.87(bs, 1H), 8 9.26(s,lH), 8 8.86(s,lH), 8 8.57(d,2H),84.81 (bs 1H), 8 4.22(qd,lH), 8 1.25(d,6H).
Example 3
Evaluation of therapeutic activity of test compound on Dextran sodium sulphate (DSS) induced Ulcerative Colitis in mice by measuring, Disease activity index, proinflammatory cytokine levels in serum and colon tissue and histopathological investigation.
N-(propan-2-yl)-3,6-dihydroimidazo[4,5-d]pyrrolo[2,3- b]pyridine-8-carboxamide hydrochloride was used as a test compound for evaluation. Test compound was evaluated on Dextran sodium sulphate (DSS) induced Ulcerative Colitis in mice by measuring, Disease activity index, proinflammatory cytokine levels in serum and colon tissue and histopathological investigation.
Test compound treated groups showed reduced Disease Activity Index (DAI) score (body weight loss, stool score & bleeding score), inhibited shortening of colon length, decrease cytokine (IL-6, IL-8, IFN-Gamma) levels and reduce Histopathology scoring compared to Disease/Colitis group.
Preparation of Test Compound (3 Doses)
Dose preparation of test compound(6mg/kg):3mg of Test compound was weighed and suspended in 2.5mL of water to get final concentration of 1.2mg/mL.
Dose preparation of test compound (12mg/kg): 6mg of test compound was weighed and suspended in 2.5mL of water to get final concentration of 2.4 mg/mL.
Dose preparation of test compound (24mg/kg): 12mg of test compound was weighed and suspended with in 2.5mL of water to get final concentration of 4.8 mg/mL.
Preparation of Reference Drug-Sulfasalazine
Reference drug-sulfasalazine 15 mg was suspended in 2.5 ml of 0.5% Na-CMC to get final concentration of 6mg/mL (Dose:30 mg/kg; Dose Volume: 05 mL/kg).
A study was conducted to check the effect of test compound on inflammatory cytokines (IL6, IFN-Gamma in serum and colon tissueand IL8 levels in colon tissue)and in a DSS-induced cytokine release model in mice.
48 C57BL/6 mice were randomized based on body weight into six groups each containing 8 animals per group (n=8)as mentioned in the Grouping Table 2. During acclimatization, animals were observed daily once for clinical signs by cage side observation, morbidity and mortality. Body weight was recorded on first and last day of acclimatization period.
Table 2: Grouping of animals
Figure imgf000032_0001
Figure imgf000033_0001
Pre-treatment
Animals were given pre-treatment for 3 days from dayl to day3. Group G1 and G2 animals received distilled water vehicle at dose volume of 5ml/kg. Group G4, G5 and G6 animals received test compoundorally at dose of 6mg/kg, 12mg/kg and 24mg/kg respectively.
Experimental Induction of Ulcerative colitis and co-treatment
Colitis was induced in all the animals (except normal control) by providing 3% DSS solution in drinking water for 7 days from day 4 to day 11. During induction period of day 4 to day 11, for group G3 standard treatment was administered sulfasalazine at the dose of 30mg/kg. Test compound treatment for groups G4, G5 and G6 was continued during the disease induction period. During experimental period, feed intake, daily body weight, stool consistency and bleeding score was observed, disease activity index (DAI) was calculated. On day 10, animals were kept for overnight fasting. Day 11, respective treatment was given. Approximately 30 minutes later blood was collected, serum was separated and kept in deep freezer for future analysis. After blood collection animals were humanely sacrificed, colon tissue was collected in all the animals. The colon length was measured and weighed. Proximal and distal part of colon was fixed in 10% NBF, embedded in paraffin and sectioned at 4-5pm thickness used for histopathological investigation using H&E staining. The middle part colon segment lOOmg of colon tissue was collected in 1ml of ice cold 1XPBS. Colon tissue was homogenized and stored in -20°C for overnight. After two freeze-thaw cycles were performed to break the cell membranes. The homogenate was centrifuged for 5 minutes at 5000 gyration. The supernatant was initially stored at -20°C and after freeze was transferred to -80°C. Serum and tissue homogenate samples were used for estimation of biomarker/cytokine analysis as for ELISA kit procedure. Body weight
Body weight of each animal was recorded daily till the end of experiment. Mean body weight was calculated and represented in tabular (Table 3) and graphical form (Figure 1). Percentage change in body weight of the animals calculated by using formula with respect to day 4. % Body Weight Change = {(Mean B.wt at day x - Initial Mean B.wt)/ Initial Mean B.wt} 100
Table 3: Percentage Change in Body Weight (g) of the Groups
Figure imgf000034_0001
Figure imgf000034_0002
Conclusion: After 5 days of 3% DSS administration i.e.day 9 to day 11, G2 disease control group showed significant decrease (p<0.001) in body weight. The reference standard sulfasalazine showed significant improvement (p<0.05) in body weight on day 11 and test compound dose group showed significant improvement (p<0.05) of body weight on day 9 to day 11. Feed consumption
Animals were offered weighed quantity of feed (gram) for cages. Feed left (gram) was measured daily during experimental period. Cumulative feed intake of the Groupsis mentioned in Table 4 and Figure 2. Feed intake was calculated by formula
Feed intake =Feed kept (gram) -Feed left (gram)
From day 4 to day 11, feed intake was calculated appropriately for each group. The cumulative feed intake of groups was calculated by adding feed intake of groups during the disease induction period.
Table 4: Cumulative feed intake of the Groups
Figure imgf000035_0001
Conclusion: Group G1 normal control animals showed healthy/normal feed intake pattern. Administration of 3% DSS in drinking water for group G2 showed marked decrease in cumulative feed intake. Reference standard and test compound treatment groups showed marked improvement in cumulative feed intake compared to G2 disease control.
Example 3.1
Evaluation of therapeutic activity of test compound on Dextran sodium sulphate (DSS) induced Ulcerative Colitis in mice by measuring, Disease activity index
Disease Activity Index
Clinical score of colitis for each mouse was recorded daily during the treatment period, i.e. day 4 to day 11. Percentagebody weight loss, stool consistency and bleeding score observed during the experiment is mentioned in the Table 5. The composite score of 3 parameters % change in body weight, stool consistency and bleeding were considered as the DAI score. Table 5: Disease activity index scoring
Figure imgf000036_0001
DAI was calculated as a composite of body weight loss, stool consistency and stool blood, was scored to analyze the potential of test compound. The 3% DSS administration to G2-DSS (3%) control group was associated with significant clinical changes including weight loss, appearance of occult faecal blood and diarrhoea in mice and DAI score of was significantly elevated from day 7 to dayl 1. DAI scoring of the Groups is mentioned in Table 6.
Table 6: DAI scoring of the Groups
Figure imgf000036_0002
Figure imgf000037_0002
Conclusion: Treatment with test compound at dose of 6 and 12 mg/kg significantly reducec the DAI score from day 8 to day 11 & day 9 to day 11 respectively. Test compound treatment at dose 6 mg/kg and dose 12 mg/kg groups delayed or reduced the appearance of the colitis symptoms like appearance of hemoccult and diarrhoea, which resulted in a significant reduction of DAI (Refer Figure 3).
Example 3.2
Evaluation of therapeutic activity of test compound on Dextran sodium sulphate (DSS) induced Ulcerative Colitis in mice by measuring, blood and colon collection.
Blood Collection
On day 10, all mice wasfasted for 12 hours (overnight). On day 11, Ihr after dosing, blood was collected under mild Isoflurane anesthesia. Serum was separated by centrifugation and stored deep freezer initially -20°C and after freezing samples were stored in -80°C until biomarkers estimation.
Colon Collection
On day 11, after blood collection animals were sacrificed humanely by CO2 anaesthesia. The colon from anus to the ileocecum will be dissected out. Colon length was measured and weighed as mentioned in Table 7.
The middle part colon segment washed with PBS, lOOmg of colon tissue was weighed in 1ml of ice cold 1XPBS. Colon tissue was homogenized and stored in -20°C for overnight. After two freeze-thaw cycles were performed to break the cell membranes. The homogenate was centrifuged for 5 minutes at 5000 gyration. The supernatant was initially stored at -20°C and after freeze was transferred to -80°C.
Table 7: Colon length and weight of the groups
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000038_0003
Figure imgf000038_0004
Conclusion: Colon Length: Administration of 3% DSS in drinking water for group G2 showed significant decrease p<0.001 in colon length. Reference standard and treatment groups showed significant improvement in colon length when compared to G2; disease control (Refer Figure 4). Colon Weight: Administration of 3% DSS in drinking water for group G2 showed marked decrease in Colon weight. Reference standard and treatment groups showed marginal improvement in colon weight when compared to G2; disease control (Refer Figure 5).
Example 3.3
Evaluation of therapeutic activity of test compound on Dextran sodium sulphate (DSS) induced Ulcerative Colitis in mice by measuring, histopathological investigation.
Histopathology
Proximal and Distal part of colon was inflated and fixed in 10% NBF, embedded in paraffin and sectioned at 4-5pm thickness used for histopathological investigation using H&E staining. For histopathological investigation of proximal colon, following scoring system which is reliable to human is adapted for scoring as mentioned in Table 8. From all the parameters total score was calculated.
Table 8: Scoring system for histopathological investigation
Figure imgf000038_0002
Figure imgf000039_0001
In histopathological investigation, each colon was scored or detail using parameters- Inflammatory infiltrate, Goblet cell loss, Crypt density, Crypt hyperplasia, Mucosal thickening, Submucosal inflammation, Crypt abcess, Ulceration. After scoring all parameters total score was calculated. Histopathology of colon-Colitis parameters score and Total Score is mentioned in Table 9.
Table 9: Histopathology of colon-Colitis parameters score and Total Score
Figure imgf000039_0002
Conclusion: Administration of DSS to group G2 showed the Total score 5.29±2.35 compared to G1 Normal control 0.43±0.43 indicating the mild colitis Disease induction.
G3 Reference Standard-Sulfasalazine-30mg/kg Showed marginal reduction in Total score i.e. 4.29±2.23 compared to disease control G2, indicating treatment effect. Treatment group’s test compound 6 mg G4 treatment group showed better reduction of Total score i.e. 4.57±1.89 (Refer Figure 6).
Example 3.4
Evaluation of therapeutic activity of test compound on Dextran sodium sulphate (DSS) induced Ulcerative Colitis in mice by measuring, proinflammatory cytokine levels in serum and colon tissue.
Serum and Colon cytokine levels
Table 10: Effect of Test compound on Serum IL6 levels
Figure imgf000040_0001
Table 11 : Effect of Test compound on Serum IFN-Gamma level
Figure imgf000040_0002
Figure imgf000041_0003
IL-6 was 6.30± 1.21 pg/mL in the serum of DSS treated mice (Group 2), the IFN-Gamma concentration in the serum of TBStreated mice was391.4±52.4 pg/mL, (Refer Table No. 10 and 11). A non- significant increase in the serum level of IL-6 and IFN-Gamma was observed in DSS group with percentage change of 45.4% and 43.1% when compared to Normal control group (Gl) which was 4.337±0.95 pg/mL and 273.5 ±57.3 pg/mL respectively.
Table 12: Effect of Test compound on Colon IL6 levels
Figure imgf000041_0001
Table 13 : Effect of Test Compound on colon IFN-Gamma level
Figure imgf000041_0002
Figure imgf000042_0002
Table 14: Effect of test compound on colon IL8 levels
Figure imgf000042_0001
IL-6 was 94.2±18.6 pg/Ml in the colon of DSS treated mice(Group 2), IFN-Gamma concentration in colon of DSS treated mice was 924.9±157.3 pg/mL, while the IL-8 levels was 308.4± 52.6 pg/mL of the DSS treated mice in colon tissue (Refer Table No.12, 13 and 14).
A significant increase in the colon level of IL-6 and IFN-Gamma was observed in DSS group with percentage change of 198.8% and 262.1% when compared to Normal control group (Gl) which was 31.5±8.2 pg/mL, 255.4±30.7 pg/Ml respectively. A non-significant increase in the colon IL-8 level was observed in DSS group with percentage change of 47.3% when compared to Normal control group (Gl) which was 209.3±51.7 pg/mL.
The compounds of formula (I) or a pharmaceutically acceptable salt thereof such as test compound showed a promising anti-inflammatory activity due to its ability to suppress the pro-inflammatory cytokines, IL-6 and IFN-Gamma of colon and serum tissue and IL-8 levels in colon tissue in a DSS-induced mice inflammation model.

Claims

We Claim:
1. A compound of Formula (I)
Figure imgf000043_0002
or a pharmaceutically acceptable salt thereof; wherein Q is a group of formula QI or Q2;
Figure imgf000043_0001
Q1 Q2
(wavy bond) represents the points of attachment; wherein Ri is -NRaRb;
R2 is hydrogen or a C1-C10 alkyl group; and
Ra and Rb independently represent hydrogen or a C1-C10 alkyl group; for use in the treatment or prevention of Ulcerative colitis.
2. The compound for use according to claim 1, wherein Q is QI and R2 represents hydrogen or a C1-C10 alkyl group.
3. The compound for use according to claim 1, wherein Q is Q2 and R2 represents hydrogen or a C1-C10 alkyl group.
4. The compound for use according to claim 1, wherein Riis -NHRa.
5. The compound for use according to claim 1, wherein Riis -NHRb.
6. The compound for use according to any one of claims 1 to 3, wherein Riis methyl.
7. The compound for use according to any one of claims 1 to 3, wherein Riis hydrogen.
8. The compound for use according to any one of claims 1 to 5, wherein Rais methyl, Rb is hydrogen and Ri methyl.
9. The compound for use according to any one of claims 1 to 5, wherein Ra is ethyl, Rb is hydrogen and Ri methyl.
10. The compound for use according to any one of claims 1 to 5, wherein Ra is propyl, Rb is hydrogen and Ri methyl.
11. The compound for use according to any one of claims 1 to 5, wherein Ra is isopropyl, R is hydrogen and Ri methyl.
12. The compound for use according to any one of claims 1 to 5, wherein Rais methyl, Rb and Ri are hydrogen.
13. The compound for use according to any one of claims 1 to 5, wherein Ra is ethyl, Rb and Ri are hydrogen.
14. The compound for use according to any one of claims 1 to 5, wherein Ra is propyl, Rb and Ri are hydrogen.
15. The compound for use according to any one of claims 1 to 5, wherein Ra is isopropyl, Rb and R2 are hydrogen.
16. The compound for use according to claim 1 wherein the compound of Formula (I) is selected from the group consisting of:
N, 1 -dimethyl- 1 ,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-carboxamide;
N-ethyl-l-methyl-l,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-carboxamide; l-methyl-N-propyl-l,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-carboxamide; l-methyl-N-(propan-2-yl)-l,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-carboxamide;
N-methyl-l,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-carboxamide;
N-ethyl-l,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-carboxamide;
N-(propan-2-yl)-l,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-carboxamide; N,3-dimethyl-3,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-carboxamide;
N-ethyl-3-methyl-3,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-carboxamide;
3-methyl-N-propyl-3,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-carboxamide;
3-methyl-N-(propan-2-yl)-3,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-carboxamide;
N-methyl-3,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-carboxamide;
N-ethyl-3,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-carboxamide; and
N-(propan-2-yl)-3,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-carboxamide; or a pharmaceutically acceptable salt thereof.
17. The compound for use according to claim 1 wherein the compound of Formula (I) is selected from the group consisting of:
N-(propan-2-yl)-l,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-carboxamide hydrochloride; and
N-(propan-2-yl)-3,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-8-carboxamide hydrochloride.
18. A pharmaceutical composition comprising a compound of Formula (I) as defined in any one of the preceding claims or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable excipient for use in the treatment or prevention of Ulcerative colitis.
19. Use of a compound as defined in any one of claims 1 to 17 or a pharmaceutical composition as defined in claim 18 for the manufacture of a medicament for use in the treatment or prevention of Ulcerative colitis.
20. A compound for use according to any one of claims 1 to 17 or a pharmaceutical composition for use according to claim 18, wherein the compound of formula (I) as defined above inhibits the cytokine levels of IL6, IFN-Gamma and IL8.
21. A method of treating or preventing Ulcerative colitis in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound as defined in any one of claims 1 to 17 or a pharmaceutical composition as defined in claim 18.
22. A method according to claim 21, wherein the compound of formula (I) as defined above inhibits cytokine levels of IL6, IFN-Gamma and IL8.
23. Use according to claim 19, wherein the compound of formula (I) as defined above inhibits cytokine levels of IL6, IFN-Gamma and IL8.
24. The compound for use according to any one of claims 1 to 17 or a composition for use according to claim 18, wherein the concentration of said compound is between O.Olmg/kg to 1000 mg/kg of subject’s body weight.
PCT/IB2023/052975 2022-04-05 2023-03-25 Substituted tricyclic compounds and their use in ulcerative colitis WO2023194841A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IN202221020506 2022-04-05
IN202221020506 2022-04-05

Publications (1)

Publication Number Publication Date
WO2023194841A1 true WO2023194841A1 (en) 2023-10-12

Family

ID=86271779

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2023/052975 WO2023194841A1 (en) 2022-04-05 2023-03-25 Substituted tricyclic compounds and their use in ulcerative colitis

Country Status (1)

Country Link
WO (1) WO2023194841A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011086053A1 (en) * 2010-01-12 2011-07-21 F. Hoffmann-La Roche Ag Tricyclic heterocyclic compounds, compositions and methods of use thereof
WO2018112382A1 (en) * 2016-12-16 2018-06-21 Janssen Pharmaceutica Nv Imidazopyrrolopyridine as inhibitors of the jak family of kinases
WO2022074572A1 (en) * 2020-10-08 2022-04-14 Unichem Laboratories Ltd Substituted tricyclic compounds

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011086053A1 (en) * 2010-01-12 2011-07-21 F. Hoffmann-La Roche Ag Tricyclic heterocyclic compounds, compositions and methods of use thereof
WO2018112382A1 (en) * 2016-12-16 2018-06-21 Janssen Pharmaceutica Nv Imidazopyrrolopyridine as inhibitors of the jak family of kinases
WO2022074572A1 (en) * 2020-10-08 2022-04-14 Unichem Laboratories Ltd Substituted tricyclic compounds

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
ALLEN LANSEL HC, PHARMACEUTICAL DOSAGE FORMS AND DRUG DELIVERY SYSTEMS, 23 December 2013 (2013-12-23)
KRISTI A. LEONARD ET AL: "Discovery of a Gut-Restricted JAK Inhibitor for the Treatment of Inflammatory Bowel Disease", JOURNAL OF MEDICINAL CHEMISTRY, vol. 63, no. 6, 5 March 2020 (2020-03-05), US, pages 2915 - 2929, XP055711298, ISSN: 0022-2623, DOI: 10.1021/acs.jmedchem.9b01439 *
LACHMAN, L.LIEBERMAN, H. A.KANIG, J. L., THE THEORY AND PRACTICE OF INDUSTRIAL PHARMACY, 1976
REMINGTON JP, THE SCIENCE AND PRACTICE OF PHARMACY, 2006
ROWE RCSHESKEY PQUINN M.: "Handbook of Pharmaceutical Excipients", 2009, PHARMACEUTICAL PRESS

Similar Documents

Publication Publication Date Title
TW490466B (en) Imidazo pyridine derivatives which inhibit gastric acid secretion
JP6266639B2 (en) Alkylamide-substituted pyrimidine compounds useful for the modulation of IL-12, IL-23 and / or IFNα
JPH0586391B2 (en)
KR20180042356A (en) How to treat cancer
US9353113B2 (en) Tetrahydrocarboline derivative
US20100056637A1 (en) Treatment methods using triaryl methane compounds
US20220298112A1 (en) N-(PHENYL)-Indole-3-Sulfonamide Derivatives And Related Compounds As GPR17 Modulators For Treating CNS Disorders Such As Multiple Sclerosis
US20230374011A1 (en) Substituted tricyclic compounds
WO1998022442A2 (en) Pharmaceutical compositions comprising diaryl-cyclomethylenpyrazole compounds and their use as cyclooxygenase i (cox 1) inhibitors
US20140350099A1 (en) Method of Treating or Preventing Pain
JPH0249306B2 (en)
US20140179919A1 (en) Compounds, compositions, methods of synthesis, and methods of treatment
US10759809B2 (en) Deuterated compound and medical use thereof
WO2023194841A1 (en) Substituted tricyclic compounds and their use in ulcerative colitis
US20040097539A1 (en) Hsp inductor
JP3034604B2 (en) Benzazapine compounds for therapy
US20190290620A1 (en) Applications of novel thiazole derivative in treating inflammatory bowel diseases
US9133103B2 (en) N-substituted benzenepropanamide and benzenepropenamide for use in the prevention or the treatment of affective disorders
AU2006331653A1 (en) Treatment methods using triaryl methane compounds
WO2023194842A1 (en) Substituted tricyclic compounds and their use in rheumatoid arthritis
WO2023194840A1 (en) Substituted tricyclic compounds and their use in covid-19
WO2021257173A1 (en) Antibiotic adjuvant compounds
US8049014B2 (en) Aminoquinoline derivatives, preparation method thereof and pharmaceutical composition comprising the same
JP2021534090A (en) Substituted benzamide and its use in treatment
WO2019088057A1 (en) Anilide derivative and medicinal use thereof

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: 23720360

Country of ref document: EP

Kind code of ref document: A1