WO2009110008A1 - Anti-ulcer activity of flavone analogs - Google Patents

Anti-ulcer activity of flavone analogs Download PDF

Info

Publication number
WO2009110008A1
WO2009110008A1 PCT/IN2009/000154 IN2009000154W WO2009110008A1 WO 2009110008 A1 WO2009110008 A1 WO 2009110008A1 IN 2009000154 W IN2009000154 W IN 2009000154W WO 2009110008 A1 WO2009110008 A1 WO 2009110008A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
body weight
induced
ulcer
ulceration
Prior art date
Application number
PCT/IN2009/000154
Other languages
French (fr)
Inventor
Janaswamy Madhusudhana Rao
Muralidhar Gurachar Purohit
Manjulatha Khanapur
Devappa Satyanarayan Nayak
Venkata Srinivas Pullela
Jhillu Singh Yadav
Original Assignee
Council Of Scientific & Industrial Research
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 Council Of Scientific & Industrial Research filed Critical Council Of Scientific & Industrial Research
Publication of WO2009110008A1 publication Critical patent/WO2009110008A1/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
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants

Definitions

  • This invention relates to anti-ulcer activity of substituted chrysin or substituted 7-hydroxy flavone analogs.
  • Chrysin or substituted 7-hydroxy flavone extracted from Acalypha indica a medicinal plant known for its antigastric ulcer property.
  • Invention identifies flavonoid in substantial yields from ethanol extract.
  • the «thanol extract was fractionated, purified and identified the compounds as chrysin, compound I and compound II.
  • Invention of potent antigastric ulcer compounds were accompanied with synthesis of few analogues derived from the chrysin, which was isolated from this plant.
  • derivative of 7-hydroxy flavone compound showed potent activity against gastric ulcers induced by aspirin, ethanol, stress and pylorus ligation at lower dose level.
  • Gastric or peptic ulcer constitutes a major disease that affects human gastrointestinal tract and major, health problem both in terms of morbidity and mortality.
  • the common clinical features of peptic ulcers are hyperacid s ⁇ retion and ulcer formation in. the stomach and duodenal part of the intestine*.
  • Peptic ulcer disease primarily effects the adult population in developed and developing countries.
  • the risk for peptic ulcer was highest in generations born before the turn of the century and has declined in all subsequent generations.
  • Low family income, old age, smoking lower educational attainment, ethnicity, increased gastric acid output, Helicobacter pylori, NSATDs and stress are that act as significant and independent basic risk factors in PUD risk factor.
  • the prevalence of upper GI diseases is increasing in subjects aged 65 years and over. Almost 40% of GU (gastric ulcer) and 25% of DU (duodenal ulcer) in the elderly patients are associated with the use of non-steroidal anti-inflammatory drugs (NSAIDs).
  • NSAIDs non-steroidal anti-inflammatory drugs
  • Gastrointestinal (GI) side effects include ulcers (found at endoscopy in 15-SO % of patients using NSAIDs regularly), complications such as upper GI bleeding (annual incidence of 1.0-1.5 %) and development of upper GI symptoms such as dyspepsia (occurring in up to 60 % patients taking NSAIDs).
  • NSAIDs are among most widely used prescribed drugs world wide for anti-inflammatory, analgesic and antipyretic effects, whereas low dose aspirin (also a NSAID) is used for cardiovascular prophylaxis. Although the therapeutic benefits of these drugs are substantial, their use is limited by their gastro duodenal toxicity, some of which can be serious or even fatal. Established risk factors for NSAIDs induced GI complications are age, ulcer history, heavy alcohol consumption, individual NSAEDs, dose association with corticoid or aspirin or anticoagulant (ulcer hemorrhage). The therapeutically acquisition of PUD of the year 2004 is the use of COX-2 inhibito reduced significantly the GI side effects of antiinflammatory treatments.
  • Aspirin is a very useful medication for the prevention of cardiovascular thrombotic events in patients with or those at risk for cardiovascular disease (CVD). Patients being treated with aspirin, even at 81 mg/day for cardio protection, should be assessed for factors that increase the risk for GI injury.
  • Stress has wide spread effects on various body systems. Stress has long been implicated as one of the risk factors for coronary diseases. Stress, defined as an acute threat to homeostasis, evokes an adaptive or allostatic response and can have both a short and long term influence on the function of the gastrointestinal tract. Stress ulceration of the stomach is associated with clinical conditions like trauma * head injury, burns, shock, sepsis and neurological disorders; and is now regarded as a multifactorial phenomenon. It is reported to result from interaction between mucosal, vascular and neuro-humoral factors and the autonomic nervous system plays a crucial role. Circulatory disturbances and the nutritional deficiency are thus induced in the local tissue, which are then followed by a rapid appearance of a deep ulcer.
  • the prophylactic regimen chosen to prevent stress ulcer bleeding should take into account the risk factors and underlying disease state of individual patients to provide the best therapy to those most likely to benefit.
  • Ethanol is common cause of acute gastric mucosal injury in both human and animals. This gastritis may produce life-threatening hemorrhage that requires surgical intervention. The mortality rate of such an intervention . at least 30%. In the rat persistence of gastric mucosal ischemia produces chronic ulceration of the stomach. Several other factors areassociated with ulcer formation although this may be an indirect relationship such factors include hereditary, smoking, elevated calcium level, corticosteroids in high dose.
  • Prophylactic options for patients suffering with gastrointestinal ulceration include antacids, sucralfate, histamme2-receptor antagonists (H2RAs), prostaglandins, muscarinic Ml- antagonists and proton pump inhibitors. Therapy has been and still is largely empirical.
  • Natural product drugs although are highly effective and free from toxic side effects, have a disadvantage with respect to short supply and chemical structure, which makes their manufacture difficult or impossible. Natural product drugs have been a source of lead structure in drug design and development. Semi synthetic analogues or synthetic analogues closely related to the natural product drug of lead are synthesized and screened to disorder their action.
  • fiavonoids have been isolated which are potent antiulcer agents increasing the gastric pH, mucosal lining of stomach and related disorders, led to the identification of Acalypha indica, which contained in substantial yields potent antiulcer flavonoid for the first time.
  • Acalypha indica Linn has been advocated in traditional medical practice of India for several diseases and many of homeopathic formulations. In folklore medicine in India, the powdered plant material is used to treat bronchitis, asthma and pneumonia.
  • Present invention relates to the identification of isolation of potent antiulcer molecules from extracts of Acalypha indica, which may find preventive as well as therapeutic applications for the control of gastrointestinal toxicity along with other complications further use in disorders where gastrointestinal toxicity inhibition play an important role in prevention and treatment of diseases not mentioned in this description.
  • Acalypha indica an Indian medicinal plant to possess potent where gastrointestinal toxicity inhibitors.
  • the ethanol extract of dried whole plant material of Acalypha indica constitutes of active principles identified as Chrysin, compound I and compound II and synthesized analogues of Chrysin and 7- hydroxy flavone in substantial yields. These mixtures and molecules may find preventive as well as therapeutic application in controlling disorders of gastrointestinal disorders and diseases.
  • the main object of the present invention is to provide the activity of analogs of 7-hydroxy flavone of general formula 1 comprising substituted Chrysin or 7-hydroxy flavone analogs, showing potent activity against ulcer at a dose level of l-25mg/kg body weight.
  • Yet another object of the present in ' ntion is to provide anti-ulcer activity of pharmaceutical .
  • composition comprising analogs of general formula ⁇ , optionally along with a pharmaceutically acceptable carrier or additives.
  • Still another object of the preset invention is to provide a method of treating ulcer in a subject wherein the said method comprising the steps of administering to the subject a compound of general formula 1 or a pharmaceutical composition of the compound of general formula 1 optionally along with pharmaceutically acceptable carrier or additives.
  • the present invention provides the use of analogs of substituted chrysin or substituted 7-hydroxy flavone analogs useful for anti-ulcer activity.
  • the said analogs were found to provide gastro-protection at lower dose level, following oral administration in aspirin induced model, pylorus ligation, ethanol induced and stress induced models in experimental rats for antiulcer potential of peptic ulcers.
  • the standard ranitidine at dose of 50 mg/kg body weight, Omeprazole at dose of 30mg/kg body weight and sucralfate at dose of 400mg/kg are taken for comparison. All three standards have shown good percentage of protection, ranitidine and Omeprazole treated groups are comparable to each other, sucralfate group is higher.
  • FIG.I Derivates of Chrysin and 7-hydroxy flavone
  • Jl [(5-hydroxy-2-phenyl-4H-l -benzopyran-4-one-7-yloxy) acetic acid ethyl ester]
  • J2 [(5-hydroxy-2-phenyl-4H-l-benzopyran-4-one-7-yloxy) acetic acid methyl ester]
  • J3 [2,2-dimethyl propionic acid 5-hydroxy-2-phenyl-4H-l-benzopyran-4-one-7yl ester]
  • J4 [7- allyloxy-5-hydroxy-2-phenyl-4H-l-benzopyran-4-one]
  • J5 [ 5,7 -bisallyloxy- ⁇ -phenyl- ⁇ H-l- benzopyran-4-one].
  • J6 [ 7-butoxy-5-hydroxy-2-phenyl-4H-l -benzopyran-4-one].
  • J7 [7-propyloxy— 5- hydroxy-2- phenyl- 4H-1 - benzopyran- 4-one]
  • J8 [ (2-phenyl-4H-l-benzopyran-4-one-7 yloxy) acetic acid ethyl ester] 15
  • FIG:2 Derivates of Chrysin and 7-hydroxy flavone J9: [7- allyloxy-2-phenyl-4H-l -benzopyran-4-one] JlO: [1 (7-oxy-2-phenyl-4H-l -benzopyran-4-one) acetamide] Jl 1: [ (2- ⁇ henyl— 4H- 1 -benzopyran— 4-one-7 yloxy) acetic acid hydrazide].
  • the present invention relates to the identification of synthetic analogues of 7-hydroxy flavone, to provide gastro protection following oral administration in aspirin induced model, pylorus ligation, ethanol induced and stress induced models in experimental rats for •0 antiulcer potential of peptic ulcers and comparing with the standard ranitidine at dose of 50 mg/kg body weight, omeprazole at dose of 30mg/kg body weight and sucralfate at dose of 400mg/kg body weight.
  • the ethanol extract of dried whole plant material of Acalypha indica constitutes of active principles identified as Chrysin, compound I and compound II .
  • Chrysin was purified from ethanol extract and was used as follows:
  • J 14 is a reactant in the above reaction for preparing Jl 5 in scheme 4.
  • Jl 6 is reactant for preparation of J13 in scheme 4
  • These antigastric ulcer molecule(s) may be administrated by any suitable conventional method prevalent in pharmaceutical practice for the treatment of gastrointestinal toxicity, control gastric pH and reduction ulcers risk factors in GI toxicity, and also in disease condition such as inflammation, stress conditions, NSAID therapy requiring inhibition of gastric acid output, formation of mucosal lining, elevate the gastric acid pH for prevention and treatment of diseases mentioned and not mentioned in this invention.
  • the potent antiulcer J 12 molecule in this invention augment defensive factors to protect the gastric mucosal from injury and for aggressive factors of gastric lesions its contributions in lesser, which provides as a good mucosal protectant and finds itself not in any other major aggressive biochemical process that are interlinked in one and another thus presents itself as more protectant than blocker.
  • Application as the case of antigastric ulcer molecules may preferably be taken orally and potentiate the mechanism of action and hence impart better therapeutic action.
  • the antigastric ulcer molecules present in pharmaceutical preparation in this invention may be formulated with any of the suitable pharmaceutically acceptable additive, carrier, vehicle, food preparations etc., suitable for human application.
  • the materials should be selected such that they should not interfere with the potency and the property of the mixture or the molecule but materials that can add to or improve the activity, may be preferred and may be decided by the conventional art and the skills available in formulary.
  • Effect r e dose- Effective dose level and duration of drug administration may be decided by the skill of ordinary art in order to bring therapeutic parameter of the disease under consideration under the control.
  • the actual rate, amount of applications, and the time of administration may vary depending upon the disease condition and severity and may be irrespective of the concentration and duration as described in the examples of this invention.
  • the compound of general Fo ⁇ ila 1 is use for anti-ulcer activity, wherein the said compound comprising substituted chrysin or substituted 7-hydroxy flavone analogs,
  • R' is selected from the group consisting of alkyl, alkenyl, alkoyl, benzoyl, hydrogen carboxymethyl, acetamide and R" is selected from the group consisting of H, OH, alkoxy, acetoxy and propenyl.
  • the compound of general formula 1 are selected from the group comprising: i. [7- allyloxy-5-hydroxy-2-phenyl-4H-l -benzopyran-4-one] - (J4) ii. [ 7-benzoyloxy-2-phenyl- 4H-1 -benzopyran-4-one] - (J12)
  • the compound is administered through oral. route at a dose ranging between lmg/lcg-25mg/kg body weight.
  • the compound is effective in treating gastrointestinal toxicity diseases induced by NSAIDs medication such as aspirin, stress, alcohol consumption and related conditions administered at a dose ranging between lmg/kg- 25mg/kg body weight.
  • the compound Jl 2 provides ulcer inhibition at a range of 47.06 - 87.75% induced by aspirin ulceration at a dose ranging between lmg/kg-25mg/kg body weight.
  • the compound Jl 2 provides glandular ulcer inhibition at a range of 51.21-95.86 % induced by cold stress ulceration at a dose ranging between lmg/kg ⁇ 25mg/kg body weight.
  • the compound J12 provides ulcer inhibition at a range of 57.63- 96.45 % induced by ethanol ulceration at a dose ranging between lmg/kg-25mg/kg body weight.
  • the compound provides elevation of pH in the range of 3.2-3.9 in ethanol induced ulceration at a dose ranging between Img/kg- 25mg/kg body weight
  • the compound provides reduction in acidity level in the range of 31.66-46.33mEq in ethanol induced ulceration at a dose ranging between lmg/kg-25mg/kg body weight.
  • the compound provides reduction in gastric juice secretion at a range of 3-5 ml in ethanol induced ulceration at a dose ranging between lmg/kg-25mg/kg body weight.
  • the compound provides ulcer inhibition in the range of 40.48-92% induced by pylorus ligated ulceration at a dose ranging between lmg/kg-25mg/kg body weight.
  • the compound provides elevation of pH in the range of 3-4.4 in pylorus ligated induced ulceration at a dose ranging between lmg/kg- 25mg/kg body weight.
  • the compound provides reduction in acidity level in the range of 48.33-31.33mEq in pylorus ligated induced ulceration- at a dose ranging between lmg/kg-25mg/kg body weight.
  • the compound provides reduction in gastric juice secretion at a range 1-2 ml in pylorus ligated induced ulceration at a dose ranging between lmg/kg-25mg/kg body weight.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising compound of general formula 1, optionally along with a pharmaceutically acceptable carrier, additives, vehicle, and food preparation, useful for anti-ulcer activity.
  • the composition is administered through oral route at a dose ranging between l-25mg/kg body weight.
  • the present invention provides a method of treating ulcer in a subject wherein the said method comprising the steps of administering to the subject a compound of general formula 1 or a pharmaceutical composition.
  • the method comprises the steps of administering to the subject the compound or the pharmaceutical composition through oral route at a dose ranging between l-25mg/kg body weight.
  • the method is effective in treating gastrointestinal toxicity diseases induced by NSAIDs medication such as aspirin, stress, consumption of alcohol and related conditions.
  • Example 1 is given by way of illustration of the present invention and should • not he construed to limit the scope of the present invention.
  • the dried powdered whole plant ( 5 Kg) was first defatted with petrol , then extracted successively with chloroform and ethanol in a soxhlet apparatus.
  • the ethanol extract was partition with n-Butanol and this on concentration obtained brown semisolid n-butanol extract.
  • This extract (100 g) was chromatographed over silica gel (60-120 mesh).
  • the column was successively eluted with 20 % ethyl acetate in hexane to afford Chrysin.
  • the yield of Chrysin is around 310 mg.
  • Further elution of the column with 2% methanol in chloroform afforded compound I the yield of compound I is around 5g.
  • Further elution of the column with 3%methanol in chloroform afforded compound I the yield of compound II is around 7g.
  • Example 2 Experimental method for gastric ulcer The compounds taken under, study for antigastric ulcer screening by four different models were selected using experimental albino rats:
  • the antiulcer activity of the compounds under taken was studied.
  • the animals were divided into 18 groups of 6 animals each.
  • Group 1 received the vehicle Tween 80 (1%, 1 ml) which served as the control.
  • Group 2 received ranitidine at a dose of 50-m.g/kg body weight, which served as standard for comparison.
  • Group 3 to 18 received Jl to J16 compounds respectively at a dose of 25 mg / kg body weight. Rats were administered per orally with a daily dose of the compounds and the drug ranitidine for a period of five days and then fasted for 24 hours.
  • narcotizing agent acetyl salicylic acid (aspirin) at a dose of 200 mg /kg body weight was administered as a suspension in tween-80 (1%), 30rnin after the drug administration each day. All drugs were administered orally on the 6th day after the last administration of the drugs and the ulcer inducing agent aspirin, the rats were killed by cervical dislocation and their stomach were opened along the greater curvature and washed with luke warm saline and examined under a dissecting microscope. The ulcer index was calculated for each stomach. The results are tabulated below in table no. 1.
  • the antiulcer activity of the compounds was studied.
  • the .animals were divided into 19 groups of 6 animals each.
  • Group 1 received the vehicle Tween 80 (1%, 1 ml) which served as the control.
  • Group 2 received ranitidine at a dose of 50-mg/kg body weight, which served as standard for comparison.
  • Group 3 to 19 received Jl to Jl € compounds respectively at a dose of 25 mg / kg body weight.
  • Animals were deprived of food 48 hours before the experiment. The water was allowed for free access. Rats were administered per orally with compounds and the drug ranitidine. The water was removed 1 hour before restraint and exposed to a temperature of 4 0 C for 2 hours. Two hours after stress, the animals were sacrificed. The stressed animals were opened along the greater curvature and the severity of gastric ulcer was assessed in terms of mean ulcer index. Results are tabulated below in table no.2.
  • the animals were divided into 20 groups of 6 animals each. Animals were deprived of food for 48 hours but had free access to water.
  • Group 1 received the vehicle Tween 80 (1%, 1 ml) which served as the control.
  • Group 2 received ranitidine at a dose of 50-mg/kg-body weight, which served as standard for comparison.
  • Group 3 received Omeprazole at a dose of 30- mg/kg body weight, which served as standard for comparison and administered i.p.
  • Group 4 received sucralfate at a dose of 400-mg/kg body weight, which served as standard for comparison.
  • Group 5 to 16 received Jl to J12 compounds respectively at a dose of 25 mg / kg body weight and group 17 to 20 received Jl 3 to J20 compounds respectively at a dose of 50 mg/kg body weight.
  • Group 1 received the vehicle Tween 80 (1%, 1 ml) which served as the control.
  • Group 2 received ranitidine at a dose of 50-mg/kg-body weight, which served as standard for comparison.
  • Group 3 to 14 received Jl to J12 compounds respectively at a dose of 25 mg / kg body weight and group 15 to 18 received Jl 3 to Jl 6 compounds respectively at a dose of 50 mg / kg body weight.
  • the stomach was ligated at the pylorus under ether anesthesia. 4hours after pylorus ligation, the animals were sacrificed and the contents drained and centrifuges at 5000 rpm for lOminutes. Aliquots of supernatant were used for determination of total acid by titrating with 0.01N NaOH using topfers reagent and phenolphthalein indicators. Results are tabulated below in table no.4
  • the antiulcer activity of the compound J12 was studied.
  • the animals were divided into 7 groups of 6 animals each.
  • Group 1 received the vehicle Tween 80 (1%, 1 ml) which served as the control.
  • Group 1 received ranitidine at a dose of 50-mg/kg-body weight, which served as standard for comparison.
  • Group 3 to 7 received J12 compound at a dose of 25, 10,-5, 2, 1 mg / kg body weight respectively. Rats were administered per orally with a daily dose of the compounds and the drug ranitidine to respective groups for a period of five days and then fasted for 24 hours.
  • acetyl salicylic acid (aspirin) at a dose of 200 mg /kg body weight was administered as a suspension in tween-80 (1%), 30min after the drug administration each day. All drugs were administered orally on the 6th day after the last administration of the drugs and the ulcer inducing agent aspirin, the rats were killed by cervical dislocation and their stomach were opened along the greater curvature and washed with luke warm saline and examined under a dissecting microscope. The ulcer index was calculated for each stomach. Results are tabulated below in table no. 5 Table 5 Ulcer protective effect of JK compound in acetyl salicyclic acid induced gastric lesions.
  • the antiulcer activity of the compound Jl 2 was studied.
  • the animals were divided into 8 groups of 6 animals each.
  • Group 1 received the vehicle Tween 80 (1%, 1 ml) which served as the control.
  • Group 2 received ranitidine at a dose of 50-mg/kg body weight
  • group 3 received diazepam at a dose of lmg/kg body weight, which served as standard for comparison.
  • Group 4 to 8 received J12 compound at a dose of 25, 10, 5, 2 and 1 mg / kg body weight respectively.
  • Animals were deprived of food 48 hours before the experiment. The water was allowed for free access. Rats were administered per orally with compounds and the drug ranitidine. The water was removed 1 hour before restraint and exposed to a temperature of 4 0 C for 2 hours.
  • the animals ⁇ vere divided into 10 groups of 6 animals each. Animals were deprived of food for 48 hours but had free access to water. Group 1 received the vehicle Tween 80 (1%, 1 ml) which served as the control. Group 2 received ranitidine at a dose of 50-mg/kg-body weight, which served as standard for comparison. Group 3 received Omeprazole at a dose of 30- mg/kg body weight, which served as standard for comparison. Group 4 received sucralfate at a .dose of 400-mg/kg body weight, which served as standard for comparison. Group 5 to 10 received J12 compound at a dose of 25, 10, 5, 3, 2 and 1 mg / kg body weight respectively. Lesions were induced 1 hour after ethanol challenge animals.
  • the animals were divided into 7 groups of 6 animals each. Animals were deprived of food for 48 hours but had free access to water. Group 1 received the vehicle Tween 80 (1%, 1 ml) which served as the control. Group 2 received ranitidine at a dose of 50-mg/kg-body weight, which served as standard for comparison. Group 3 to 7 received Jl 2 compound at a dose of 25, 10, 5, 2 and 1 mg / kg body weight respectively. After one hour of administration of drug the stomach was ligated at the pylorus under ether anesthesia. 4hours after pylorus ligation, the animals were sacrificed and the contents drained and centrifuges at 5000 rpm for lOminutes.
  • Ulcers are thought to be imbalances in gastric offensive and defensive mucosal factors. While acid and pepsin make up the offensive factors, the defensive factors include mucin secretion, mucosal glycoprotein, cell proliferation etc. To regain the balance, different therapeutic agents are used to inhibit the gastric acid secretion or to boost the mucosal defense mechanisms by increasing mucus production, stabilizing the surface epithelial cells. The number of lesions present on the gastric mucosa is indicative of the severity of the ulcer disease.
  • J12 (benzoyloxy group substitution in 7-hydroxy flavone) at a dose level of 5mg/kg and J4 (allyloxy group substitution in chrysin) at dose level 25mg/kg found to provide gastro-protection following oral administration in aspirin induced model, pylorus ligation, ethanol induced and stress induced models induced in experimental rats for antiulcer potential of peptic ulcer.
  • the standard ranitidine at dose of 50mg/kg body weight, omeprazole at dose of 30mg/kg body weight and sucralfate at dose of 400mg/kg body weight are taken for comparison. All three standard have shown good percentage of protection, ranitidine and omeprazole treated groups are comparable to each other, sucralfate treated group are higher.
  • Jl 2 and J4 compounds are effective in protection against gastric ulcers far lower than the standard drugs dose levels.
  • Gastric mucosal damages induced in different experimental ulcer models have different mechanisms. A favorable result here would indicate, it could provide protection from NSAID- induced darn ⁇ e, ethanol ulceration, stress induced ulceration, pylorus ligation on oral administration.
  • this compound are effective against gastric ulcer induced by different ways, probably this can be efficient drug for the protection of gastric ulcer caused due to various factors, low family income, old age, smoking, lower educational attainment, ethnicity, increase gastric acids output, Helicobacter pylori infection, NSAIDs and stress that act as significant and independent basic risk factors in PUD risk factors.
  • Gastric or peptic ulcer constitutes a major disease that affects human gastrointestinal tract and major health problem both in term of morbidity and motility.
  • the common clinical features of peptic ulcer are hyperacid secretion and ulcer formation in the stomach and duodenal part of the intestine.

Abstract

The present invention relates to the use of a compound of general formula (1), useful for anti- ulcer activity. The compounds comprising substituted chrysin or substituted 7-hydroxy flavone analogs of the formula (1) wherein R' is selected from the group consisting of alkyl, alkenyl, alkoyl,benzoyl, hydrogen, carboxymethyl, acetamjde and R" is selected from the group consisting of H, OH, alkoxy, acetoxy and propenyl. More particularly, the present invention provides the use of analogs of 7-hydroxy flavone, to provide anti-ulcer against aspirin induced ulceration, pylorus ligation induced ulceration, ethanol induced ulceration and stress induced ulceration at a dose ranging between.1 -25mg/kg body weight.

Description

"ANTI-ULCER ACTIVITY OF FLAVONE ANALOGS"
Field of invention:
This invention relates to anti-ulcer activity of substituted chrysin or substituted 7-hydroxy flavone analogs. Chrysin or substituted 7-hydroxy flavone extracted from Acalypha indica, a medicinal plant known for its antigastric ulcer property. As a result of literature survey and in our present study we have identified mucoprotective and antiulcer properties in the flavone class of compounds isolated. Invention identifies flavonoid in substantial yields from ethanol extract. The «thanol extract was fractionated, purified and identified the compounds as chrysin, compound I and compound II. Invention of potent antigastric ulcer compounds were accompanied with synthesis of few analogues derived from the chrysin, which was isolated from this plant. As per the results, derivative of 7-hydroxy flavone compound showed potent activity against gastric ulcers induced by aspirin, ethanol, stress and pylorus ligation at lower dose level.
Background of invention:
Gastric or peptic ulcer constitutes a major disease that affects human gastrointestinal tract and major, health problem both in terms of morbidity and mortality. The common clinical features of peptic ulcers are hyperacid s ■ retion and ulcer formation in. the stomach and duodenal part of the intestine*.
Peptic ulcer disease (PUD) primarily effects the adult population in developed and developing countries. The risk for peptic ulcer was highest in generations born before the turn of the century and has declined in all subsequent generations. Low family income, old age, smoking lower educational attainment, ethnicity, increased gastric acid output, Helicobacter pylori, NSATDs and stress are that act as significant and independent basic risk factors in PUD risk factor. The prevalence of upper GI diseases is increasing in subjects aged 65 years and over. Almost 40% of GU (gastric ulcer) and 25% of DU (duodenal ulcer) in the elderly patients are associated with the use of non-steroidal anti-inflammatory drugs (NSAIDs). Gastrointestinal (GI) side effects include ulcers (found at endoscopy in 15-SO % of patients using NSAIDs regularly), complications such as upper GI bleeding (annual incidence of 1.0-1.5 %) and development of upper GI symptoms such as dyspepsia (occurring in up to 60 % patients taking NSAIDs).
NSAIDs are among most widely used prescribed drugs world wide for anti-inflammatory, analgesic and antipyretic effects, whereas low dose aspirin (also a NSAID) is used for cardiovascular prophylaxis. Although the therapeutic benefits of these drugs are substantial, their use is limited by their gastro duodenal toxicity, some of which can be serious or even fatal. Established risk factors for NSAIDs induced GI complications are age, ulcer history, heavy alcohol consumption, individual NSAEDs, dose association with corticoid or aspirin or anticoagulant (ulcer hemorrhage). The therapeutically acquisition of PUD of the year 2004 is the use of COX-2 inhibito reduced significantly the GI side effects of antiinflammatory treatments. Since cardiac adverse effects of certain COX-2 inhibitors (NSAIDs) had been reported, the treatments with COX-2 inhibitors came widely into question. Aspirin is a very useful medication for the prevention of cardiovascular thrombotic events in patients with or those at risk for cardiovascular disease (CVD). Patients being treated with aspirin, even at 81 mg/day for cardio protection, should be assessed for factors that increase the risk for GI injury.
Stress has wide spread effects on various body systems. Stress has long been implicated as one of the risk factors for coronary diseases. Stress, defined as an acute threat to homeostasis, evokes an adaptive or allostatic response and can have both a short and long term influence on the function of the gastrointestinal tract. Stress ulceration of the stomach is associated with clinical conditions like trauma* head injury, burns, shock, sepsis and neurological disorders; and is now regarded as a multifactorial phenomenon. It is reported to result from interaction between mucosal, vascular and neuro-humoral factors and the autonomic nervous system plays a crucial role. Circulatory disturbances and the nutritional deficiency are thus induced in the local tissue, which are then followed by a rapid appearance of a deep ulcer.
Gastrointestinal complications frequently occur in patients admitted to the intensive care unit. Of this ulceration and bleeding related to stress-related mucosal disease can lengthen hospitalization and increases mortality. The prophylactic regimen chosen to prevent stress ulcer bleeding should take into account the risk factors and underlying disease state of individual patients to provide the best therapy to those most likely to benefit.
Ethanol is common cause of acute gastric mucosal injury in both human and animals. This gastritis may produce life-threatening hemorrhage that requires surgical intervention. The mortality rate of such an intervention . at least 30%. In the rat persistence of gastric mucosal ischemia produces chronic ulceration of the stomach. Several other factors areassociated with ulcer formation although this may be an indirect relationship such factors include hereditary, smoking, elevated calcium level, corticosteroids in high dose.
The majority of peptic ulcers causing growing burning or aching pain in the region of the stomach made worse by or unrelated to food. Pain tends to be worse at night and occurs usually 1 to 3 hours after food during the day. Additionally there may be food aversion, weight loss, nausea, belching or bloating. There is great individual variation and occasionally the pain may be referred to the back or the upper quadrant of the abdomen. Complications include bleeding, obstruction, perforation or intractable pain.
Prophylactic options for patients suffering with gastrointestinal ulceration include antacids, sucralfate, histamme2-receptor antagonists (H2RAs), prostaglandins, muscarinic Ml- antagonists and proton pump inhibitors. Therapy has been and still is largely empirical.
The prostaglandin's fulfilled their early promise and muscarinic Mi -antagonists, although more selective than the earlier anti-cholinergic agents, have limited application. Inhibition of the H+/ K+ ATPase by non-competitive agents is limited to short-term administration and the development of a potent selective gastrin antagonist has yet be realized.
Reduction of symptoms, nullifying the side effects and improvement in quality of life are among the top priorities of diseases for the suffering persons. Although these factors need to be considered and balanced in evaluating new therapies for widespread use. The reduction in risk in a specific patient population should be considered before a particular regimen is deemed ineffective or too costly.
The plants create unexpected and novel structure to protect themselves from predator organism. By trail and error, several plants and plant products are identified as drugs. Natural product drugs although are highly effective and free from toxic side effects, have a disadvantage with respect to short supply and chemical structure, which makes their manufacture difficult or impossible. Natural product drugs have been a source of lead structure in drug design and development. Semi synthetic analogues or synthetic analogues closely related to the natural product drug of lead are synthesized and screened to disorder their action. In the light of above descriptions, in our isolation work fiavonoids have been isolated which are potent antiulcer agents increasing the gastric pH, mucosal lining of stomach and related disorders, led to the identification of Acalypha indica, which contained in substantial yields potent antiulcer flavonoid for the first time. Acalypha indica Linn has been advocated in traditional medical practice of India for several diseases and many of homeopathic formulations. In folklore medicine in India, the powdered plant material is used to treat bronchitis, asthma and pneumonia.
Present invention relates to the identification of isolation of potent antiulcer molecules from extracts of Acalypha indica, which may find preventive as well as therapeutic applications for the control of gastrointestinal toxicity along with other complications further use in disorders where gastrointestinal toxicity inhibition play an important role in prevention and treatment of diseases not mentioned in this description.
In this invention, we identify Acalypha indica an Indian medicinal plant to possess potent where gastrointestinal toxicity inhibitors. The ethanol extract of dried whole plant material of Acalypha indica constitutes of active principles identified as Chrysin, compound I and compound II and synthesized analogues of Chrysin and 7- hydroxy flavone in substantial yields. These mixtures and molecules may find preventive as well as therapeutic application in controlling disorders of gastrointestinal disorders and diseases. (N.D. Satyanarayan and M. G. Purohit Antiulcer activity of Acalypha indica L. (Euphorbiaceae) on ethanol induced gastric ulcers "Journal of Ethanopharmacology" Articles in Press). Synthesis of Compound J4 is performed by known methods mentioned in (V K Ahluwalia, G P Sachdev & T R Seshadri., Nuclear allylation of Chrysin., Ind J Chem VoI 5, 1967, 97-1 OφSynthesis of J12 is performed by known methods mentioned in (Baker, J.Chem.Soc, 1933, 1381,1387. Rangaswam. Seshadri., Proc-Indian Acad. Sci. SectA 10, 1939, 6 Trivedi eta al., J In Chem Soc, 20, 1943, 171 Dorofeenko, Tkachenko., J Org Chem USSR (Eng. Transl), 8, 1972, 2250.
Objects of invention:
The main object of the present invention is to provide the activity of analogs of 7-hydroxy flavone of general formula 1 comprising substituted Chrysin or 7-hydroxy flavone analogs, showing potent activity against ulcer at a dose level of l-25mg/kg body weight.
Yet another object of the present in ' ntion is to provide anti-ulcer activity of pharmaceutical . composition comprising analogs of general formula \, optionally along with a pharmaceutically acceptable carrier or additives.
Still another object of the preset invention is to provide a method of treating ulcer in a subject wherein the said method comprising the steps of administering to the subject a compound of general formula 1 or a pharmaceutical composition of the compound of general formula 1 optionally along with pharmaceutically acceptable carrier or additives.
Summary of the invention:
Accordingly, the present invention provides the use of analogs of substituted chrysin or substituted 7-hydroxy flavone analogs useful for anti-ulcer activity.
The said analogs were found to provide gastro-protection at lower dose level, following oral administration in aspirin induced model, pylorus ligation, ethanol induced and stress induced models in experimental rats for antiulcer potential of peptic ulcers. In the present invention the standard ranitidine at dose of 50 mg/kg body weight, Omeprazole at dose of 30mg/kg body weight and sucralfate at dose of 400mg/kg are taken for comparison. All three standards have shown good percentage of protection, ranitidine and Omeprazole treated groups are comparable to each other, sucralfate group is higher.
Brief description of the drawings and tables:
The invention is illustrated by the accompanying drawings where in 5 FIGtI: Derivates of Chrysin and 7-hydroxy flavone
Jl : [(5-hydroxy-2-phenyl-4H-l -benzopyran-4-one-7-yloxy) acetic acid ethyl ester] J2: [(5-hydroxy-2-phenyl-4H-l-benzopyran-4-one-7-yloxy) acetic acid methyl ester] J3: [2,2-dimethyl propionic acid 5-hydroxy-2-phenyl-4H-l-benzopyran-4-one-7yl ester]
10 J4 : [7- allyloxy-5-hydroxy-2-phenyl-4H-l-benzopyran-4-one] J5 : [ 5,7 -bisallyloxy-^-phenyl-^H-l- benzopyran-4-one]. J6: [ 7-butoxy-5-hydroxy-2-phenyl-4H-l -benzopyran-4-one]. J7: [7-propyloxy— 5- hydroxy-2- phenyl- 4H-1 - benzopyran- 4-one] J8: [ (2-phenyl-4H-l-benzopyran-4-one-7 yloxy) acetic acid ethyl ester] 15
FIG:2: Derivates of Chrysin and 7-hydroxy flavone J9: [7- allyloxy-2-phenyl-4H-l -benzopyran-4-one] JlO: [1 (7-oxy-2-phenyl-4H-l -benzopyran-4-one) acetamide] Jl 1: [ (2-ρhenyl— 4H- 1 -benzopyran— 4-one-7 yloxy) acetic acid hydrazide]. iO Jl 2: [ 7-benzoyloxy-2-phenyl- 4H-1— benzopyran-4-one] J13: [ 5,7 -diacetoxy^-phenyl-^H-l- benzopyran-4-one] Jl 4 : [5-hydroxy ,7 -methoxy -2-phenyl-4H-l - benzopyran-4-one] Jl 5 : [7-methoxy-5-acetoxy-2-phenyl- 4H-l-benzopyran-4-one] Jl 6: Chrysin [5,7-dihydroxy-2-phenyl-chromen-4-one] •5
Detail description of the invention:
The present invention relates to the identification of synthetic analogues of 7-hydroxy flavone, to provide gastro protection following oral administration in aspirin induced model, pylorus ligation, ethanol induced and stress induced models in experimental rats for •0 antiulcer potential of peptic ulcers and comparing with the standard ranitidine at dose of 50 mg/kg body weight, omeprazole at dose of 30mg/kg body weight and sucralfate at dose of 400mg/kg body weight.
The ethanol extract of dried whole plant material of Acalypha indica constitutes of active principles identified as Chrysin, compound I and compound II . Chrysin was purified from ethanol extract and was used as follows:
i) General procedure for the preparation of 7-O- substituted derivatives of chrysin and 7-O- substituted derivatives of 7-hydroxy flavone:
To Ά mixture of .chrysin or 7-hydroxy flavone (1 g, 3.93 mmol), anhydrous potassium carbonate (0.81 g, 5.8 mmol) and corresponding halide in 20ml acetone. The mixture was refiuxed for 6-12 h. After completion of the reaction potassium carbonate was filtered off and washed with -excess of acetone (2 x 50 ml). The combined filtrate was concentrated under vacuum. The residue was purified by recrystallisation to yield 7-0-substituted derivatives of chrysin and 7-0-substituted derivatives of flavone in pure form ( Jl to Jl 6 except J5, J135J11,J15) of Scheme-1.
Schem e - 1
Figure imgf000009_0001
R1"
R' R X
OH Cl O H
J1 -CH2COOC2H5 OH Cl OH
J2 -CH2COOCH3 OH Cl OH
J3 -COC(CH3)3 OH Br OH
J4
OH Br OH
J6
OH
J7 Br OH
H
J8 -CH2COOC2H5 Cl H
^^ H H
J9 Br
J10 Cl H
-CH2CO NH2 H
Cl H
J12 -CO Ph H
I -OH
J14 -CH3 OH
R=OH=JlO
R=H= 7-hydroxy flavone Jl 6 is reactant in the above reaction
ϋ) Synthesis of 5,7-bisallyIoxy-2-phenyl-4H~l-benzopyran-4-one (J5)
7-allylαxy-5-hydroxy~2-phenyl-4H-l-benzopyran-4-one (J4) (2.4g, 0.01 mol) and allyl bromide (0.01 mol) in equimolar taken in round bottom flask along with 50 ml acetone and activated potassium carbonate (5g). The reaction was refluxed for 6 hours. The reaction mixture was cooled and filtered. The filtrate was concentrated and poured onto crushed ice and solid separated. The solid was crystallized with hexane and alcohol to obtain colorless crystals (J5).
Scheme - 2
Figure imgf000010_0001
iii) Synthesis of (2-phenyl-4H-l-benzopyran-4-one-7-yloxy) acetic acid hydrazide (JlI)
Compound J8 (O.Olmol) and hydrazine hydtate (99%, 0.01 mol) in absolute ethanol was refluxed for 20 hr and concentrated. The solid, thus obtained, was filtered and crystallized from ethanol and methanol mixture to get (2-phenyl-4H-l-benzopyran-4-one-7- yloxy) acetic acid hydrazide (Jl 1).
Scheme - 3
Figure imgf000010_0002
iv) General procedure for the preparation of 5, 7-O-substituted derivatives of chrysin
To a mixture of 7-0- substituted derivatives of chrysin (1 g, 3.93 mmol) , anhydrous potassium carbonate (0.81 g, 5.8 mmol) and corresponding methylating agent in 20ml acetone. The mixture was refluxed for 6 h. After completion of the reaction potassium carbonate was filtered off and washed with excess of acetone (2 x 50 ml). The combined filtrate was concentrated under vacuum. The residue was purified by recrystallisation to yield 5, 7-O-substituted derivatives of chrysin in pure form (J 13 and Jl 5). Scheme - 4
Figure imgf000011_0001
J13 , J15
R- =H R=OH=JI 6 R-- =H R=OCH3=014
R R1 R"
J13 (CH3CO)2O COCH3 COCH3
OH J14 OCH3 C H3I CH3 CH3
COCH3 JI 5 OCH, (CH3CO)2O CH3
J 14 is a reactant in the above reaction for preparing Jl 5 in scheme 4. Jl 6 is reactant for preparation of J13 in scheme 4
.5 Chrysin (Fig ': 16) obtained from Acalypha indica has the following spectral, chemical and physical properties
MP: 285-2860C. IR (KBr)vmax 3450, 2925, 1626, 1024 cm"1. 1H NMR (400 MHz, CDCl3+ MeOH-dj) (δ) 7.82-7 /92 (2H, m, H-2', 6'), 7.44-7.58 (3H, m, H-3', 4', 5'), 6.64 (IH, s, H-8), 6.44 (IH, s, H-3), 6.24 (IH, s, H-6). 13C NMR (50 MHz, DMSO d6) 5163.0 (C-2), 105.0 (C- O 3), 181.6 (C-4), 161.5 (C-5), 99.0 (C-6), 164.3 (C-I), 94.0 (C-8), 157.3 (C-9), 104.0 (C-IO), 138.7 (C-I' ), 126.1 (C-2' ), 128.8 (C-3' ), 131.6 (C-4'), 128.8 (C-5' ), 126.1 (C-6' ). EIMS: M+ 254.
Analogs from chrysin obtained from Acalypha indica as derivatives namely Jl (Fig:l) [(5- hydroxy-2-phenyl-4H-l-benzopyran-4-one-7-yloxy) acetic acid ethyl ester] following 5 spectraichemical and physical properties
MP: 145-1460C. IR (KBr) vmax 1690 51737, 3446 cm"1. 1H NMR (400 MHz5 CDCl3) (δ) 7.8 - 7.9 (2H, m, H-2', 6'), 7.4 -7.6 (2H, m, H-3', 5'), 7.2 (IH, s, H-4'), 6.6 (IH, s, H-8), 6.7 (IH, s, H-3), 6.4 (IH, s, H-6), 12.8 (IH, s, OH-5), 4.8 (2H, s, H-I"), 4.4 (3H, q, H-3'"), 1.4(2H, t, H-4'")
Analogs from chrysin obtained from Acafypha indica as derivatives namely J2 (Fig:2) [(5- hydroxy-2-phenyl-4H-l-benzopyran-4-one-7-yloxy) acetic acid methyl ester] following spectral chemical and physical properties
MP: 158 - 1600C. IR (KBr) vraax 1660 ,1741, 3542.12 cm'1. 1H NMR (400 MHz3 DMSO-de ) (δ) 8.0 - 8.2 (2H, m, H-2', 6'), 7.4-7.6 (3H, m, H-3',4', 5'), 6.9 (IH, s, H-8), 6.8 (IH, s, H- 3), 6.4 (IH, s, H-6), 12.8 (IH, s, OH-5), 4.9 (2H, s, H-I'") , 3.75 (3H, s, H-3'")
Analogs from chrysin obtained from Acafypha indica, as derivatives namely J3 (Fig:3) [ 2,2- dimethyl propionic acid 5-hydroxy-2-phenyl-4H-l-bc" pyrari-4-one-7yl ester] following spectral chemical and physical properties
MP: 130-1310C. IR (KBr) vmax 1657.4 ,1749.9, 3445.12 cm"1. 1H NMR (400 MHz, CDCl3) (δ) 7.8 - 7.9(2H, m, H-2', 6'), 7.6 -7.7 (3H, m, H-3', 4', 5'), 7.4 (IH, s, H-8), 7.3 (IH, s, H-3), 7.1 (IH, s, H-6), 12.6 (IH, s, OH-5), IA (9H, s, H-I"') , EIMS: M+SS?.
Analogs from chrysin obtained from Acafypha indica as derivatives namely J4 (Fig:4) [7- allyloxy-5-hydroxy-2-phenyl-4H-l-benzopyran-4-one] following spectral chemical and physical properties
MP138-135°C. IR (KBr) vmax 1665.45, 3448.3 cm"1. 1H NMR (400 MHz, CDCl3) (δ)7.9-8.0 (2H, d, H-2', 6'), 7.5 -7.6 (3H, m, H-3',4', 5'), 7.3(1H, s, H-8), 6.55(1H, s, H-3), 6.4 (IH, s, H-6), 12.75 (IH, s, OH-5), 4.75 (2H, m, H-I") , 6.1 (IH, m, H-2'"), 5.3-5.5 (2H, m, H-3'") EIMS: M+SSS. ' •
Analogs from chrysin obtained from Acafypha indica as derivatives namely J5 (Fig:5) [5,7
-bisallyloxy-2-phenyl-4H-l- benzopyran-4—one] following spectral chemical and physical properties
MP 100 - 1010C. IR (KBr)vmax 1658.49 cm 1. 1H NMR (400 MHz, CDCl3) (δ) 7.9-8.0 (2H, d, H-2', 6'), 7.5 -7.6 (3H, m, H-3',4', 5'), 6.7 (IH, s, H-8), 6.6 (IH, s, H-3), 6.4 (IH, s, H-6), 4.65-4.75 (4H, m. H-I "M"") , 6.0-6.2 (2H, m, H-2'", 2""), 5.35-5.7 (4H, m, H-3'",-3"")
Analogs from chrysin obtained from Acalypha indica as derivatives namely J6 (Fig:6) [ 7- butoxy— 5-hydroxy— 2-phenyl-4H— l-benzopyran-4-one] following spectral chemical and physical properties
MP: 115 - 1170C. IR (KBr) Vn13x 1666.32, 3443.73 cm"1. 1H NMR (400 MHz, DMSOd6) (δ) 8.1-82 (2H, m, H-2', 61), 1£ -7.7 (2H, m, H-3',4', 5'), 7.1(1H5 s, H-8), 6.8 (IH, s, H-3), 6.4 (IH, s, H-6), 12.6 (IH, s, OH-5), 3.9 (2H,m, 1"), 3.4 (4H,m, 2"), 2.2 (2H,m, 3"), 1.0 (3H, m5 4").
Analogs from chrysin obtained from Acalypha indica as derivatives namely J7 (Fig: 7) [7— propyloxy-5-hydroxy-2- phenyl- 4H- 1- benzopyran- 4-one] following spectral chemical and physical properties MP: 96 - 980C. IR (KBr) vmax 1661, 3436 ran'1. 1H NMR (400 MHz, DMSOd6) (δ) 8.1-8.2 (2H, m, H-2', 6'), 7.6 -7.7 (3H, m, H-3', 4', 5'), 7.1 (IH, s, H-8), 6.8(1H5 s, H-3), 6.4 (IH, s, H-6), 12.8 (IH5 s, OH-5), 4.1 (2H5Hi, 1"), 1.7-1.8 (4H,m, 2"), 0.9-1.0 ( 3H,t, 3").
Analogs from chrysin obtained from Acalypha indica as derivatives namely JS (Fig: 8) [ (2- phenyl-4H-l-benzopyran-4-one— 7 yloxy) acetic acid ethyl ester] following spectral chemical and physical properties
MP: 110-1120C. IR (KBr) vmax 1769 , 1671cm4. 1HNMR (400 MHz, CDCl3) (δ) 8.0-8.1 (2H5 m5 H-2', 6'), 7.6 -7.7 (2H, m, H-3', 5'), 7.2 (IH, s5 H-4'), 6.7 (IH, s, H-8), 6.6 (IH, s, H- 3), 6.4 (IH5 s, H-6), 4.6 (2H, s, H-I"), 4.19 (3H5 q, H-3'"), 1.19 (2H5 1, H-4'")
Analogs from chrysin obtained from Acalypha indica as derivatives namely J9 (Fig:9) [7- allyloxy-2-phenyl-4H-l-benzopyran-4-one] following spectral chemical and physical properties
MP 960C IR (KBr) vmax 1671 cm'1. 1H NMR (400 MHz, CDCl3) (δ) 7.9-8.0 (2H, d, H-2', 6'), 7.8 -7.7 (3H. m, H-3', 4', 5'), 7.3 (IH, s, H-8), 6.55 (IH, s, H-S)5 6.4 (IH, s, H-6), 12.75(1H, s, OH-5), 4.55 (2Hd5H-I"), 5.8-6.1 (IH, m, H-2'").5.37-522 (2H,m,H-3"0
Analogs from chrysin obtained from Acalypha indica as derivatives namely JlO (Fig: 10) [1 (7-oxy-2-phenyl-4H-l-benzopyran-4-one) acetamide] following spectral chemical* and physical properties
MP 2700C. IR (KBr) vmax 3490, 1704 , 1671cm-1. 1B. NMR (400 MHz, DMS0-d6) (δ) 7.9- 8.0 (2H, d, H-2', 6'), 7.65 -7.75 (2H, m, H-3',5'), 7.5 (IH, S, H-4'), 7.30 (IH, s, H-8), 7.0 (IH, s, H-3), 7.2 (IH, s, H-6), 8.2 (IH5 d, NH).
Analogs from chrysin obtained from Acalypha indica as derivatives namely Jl 1 (Fig:l 1) [ (2-phenyl- 4H-l-benzopyran-4-one-7 yloxy) acetic acid hydrazide] following spectral chemical and physical properties MP242-243°C. IR (KBr) V1113x 3435, 3273, 1672 cm4. 1H NMR (400 MHz, DMS0-d6) (δ) 7.7 -7.75 (2H, d, H-2', 6'), 7.2 -7.5 (3H, m, H-3', 5',4'), β.% (IH, s, H-8), 6.5 (IH, s, H-3), 6.4 (IH, s, H-6), 9.3 (IH, s, NH), 4.3-4.5 (2H, d, NH2), 3.25 (2H, s, 1'").
Analogs from chrysin obtained from Acalypha indica as derivatives namely J12 (Fig: 12) [ 7-benzoyloxy— 2— phenyl- 4H-1- benzopyran— 4-one] following spectral chemical and physical properties
MP: 250-2510C. IR (KBr) vmax 1671, 1615 cm4.1HNMR(400 MHz, DMS0-d6) (δ) 7.82-
7.95 (3H, m, H-2", 6", H-5), 7.4 - 7.7 (1OH, m, H-2',6', 3', 4', 5',3",4", 5",.H-8, H-6 ), 6.64
(IH, s, H-8), 7.05 (IH, s, H-3). 13C NMR (50 MHz, DMSO d6) δ 176 (C-2), 103.0 (C-3), .194 (C-4), 116.2 (C-5), 127.4 (C-6), 116 (C-7), 162 (C-8), 115.5 (C-9), 158 (C-10), 136.4
(C-I' ), 127.4 (C-2' , C-6' ), 128.5 (C-4'), 128.5 (C-3',5' ), 164 (COO), 132 (C-I" ), 129.8 (C-
2" , C-6" ), 134.3 (C-4'), 129 (C-3',5' ), EIMS: M+ 343.3.
Analogs from chrysin obtained from Acalypha indica as derivatives namely Jl 3 (Fig: 13) [ 5,7-diacetoxy -2-phenyl-4H-l- benzopyran-4-one] following spectral chemical and physical properties
MP 3280C IR (KBr) vmax 1650, 1760 cm4. 1H NMR (400 MHz, CDCl3) (δ) 7.8-7.9 (2H, d,
H-2'5 6'), 7.55-7.45 (2H, m, H-3', 5'), 7.35(lH,s, H-4'), 7.1 (IH, s, H-8), 6.9 (IH, s, H-3), 6.4
(IH, s, H-6) 2.35 (6H3 d. OCOCH3).
Analogs from chrysin obtained from Acalypha indica as derivatives namely J14 (Fig: 14) [5-hydroxy ,7 methoxy -2-phenyl-4H~l- benzopyran-4-one] following spectral chemical and physical properties
MP: 2820C. (KBr) vmax 1654 cm"1. 1H NMR (400 MHz, DMSO d6) (δ) 7.8-7.9 (2H, d, H-2', 6'), 7.55-7,45 (2H, m, H-3', 5'), 7.25 (IH, s, H-4'), 6.9 (IH, s, H-8), 6.7 (IH, s, H-3), 6.4 (IH, s, H-6), 3.9 - 4.0 (6H, d, OCH3). EIMS: M+ 283
Analogs from chrysin obtained from Acalypha indica as derivatives namely J15 (Fig:15) [7- methoxy-5-acetoxy-2-phenyl- 4H-l-benzopyran-4-one] following spectral chemical and physical properties MP: 2800C. (KBr) vmax 1760, 1640 cm"1. 1H NMR (400 MHz, DMSO d6) (δ) 7.8-7.9 (2H, d, H-2', 6'), 7.55-7.45 (3H, m, H-3', 4', 5'), 7.2 (IH, s, H-8), 6.9 (IH, s, H-3), 6.7 (IH, s, H-6), 3,9 (3H, s, OCH3), 2.45 (3H, s , OCOCH3).
Applications, administration and the formulations: These antigastric ulcer molecule(s) may be administrated by any suitable conventional method prevalent in pharmaceutical practice for the treatment of gastrointestinal toxicity, control gastric pH and reduction ulcers risk factors in GI toxicity, and also in disease condition such as inflammation, stress conditions, NSAID therapy requiring inhibition of gastric acid output, formation of mucosal lining, elevate the gastric acid pH for prevention and treatment of diseases mentioned and not mentioned in this invention. The potent antiulcer J 12 molecule in this invention augment defensive factors to protect the gastric mucosal from injury and for aggressive factors of gastric lesions its contributions in lesser, which provides as a good mucosal protectant and finds itself not in any other major aggressive biochemical process that are interlinked in one and another thus presents itself as more protectant than blocker. Application as the case of antigastric ulcer molecules may preferably be taken orally and potentiate the mechanism of action and hence impart better therapeutic action. The antigastric ulcer molecules present in pharmaceutical preparation in this invention may be formulated with any of the suitable pharmaceutically acceptable additive, carrier, vehicle, food preparations etc., suitable for human application. The materials should be selected such that they should not interfere with the potency and the property of the mixture or the molecule but materials that can add to or improve the activity, may be preferred and may be decided by the conventional art and the skills available in formulary.
Effect re dose- Effective dose level and duration of drug administration may be decided by the skill of ordinary art in order to bring therapeutic parameter of the disease under consideration under the control. The actual rate, amount of applications, and the time of administration may vary depending upon the disease condition and severity and may be irrespective of the concentration and duration as described in the examples of this invention.
In an embodiment of the present invention, the compound of general Foπ ila 1 is use for anti-ulcer activity, wherein the said compound comprising substituted chrysin or substituted 7-hydroxy flavone analogs,
Figure imgf000016_0001
Formula 1 wherein R' is selected from the group consisting of alkyl, alkenyl, alkoyl, benzoyl, hydrogen carboxymethyl, acetamide and R" is selected from the group consisting of H, OH, alkoxy, acetoxy and propenyl.
In still another embodiment of the present invention, the compound of general formula 1 are selected from the group comprising: i. [7- allyloxy-5-hydroxy-2-phenyl-4H-l -benzopyran-4-one] - (J4) ii. [ 7-benzoyloxy-2-phenyl- 4H-1 -benzopyran-4-one] - (J12) Further, in another embodiment of the present invention, the compound is administered through oral. route at a dose ranging between lmg/lcg-25mg/kg body weight. In another embodiment of the present invention, the compound is effective in treating gastrointestinal toxicity diseases induced by NSAIDs medication such as aspirin, stress, alcohol consumption and related conditions administered at a dose ranging between lmg/kg- 25mg/kg body weight.
In yet another embodiment of the present invention, the compound Jl 2 provides ulcer inhibition at a range of 47.06 - 87.75% induced by aspirin ulceration at a dose ranging between lmg/kg-25mg/kg body weight.
In another embodiment of the present invention, the compound Jl 2 provides glandular ulcer inhibition at a range of 51.21-95.86 % induced by cold stress ulceration at a dose ranging between lmg/kg~25mg/kg body weight.
In still another embodiment of the present invention, the compound J12 provides ulcer inhibition at a range of 57.63- 96.45 % induced by ethanol ulceration at a dose ranging between lmg/kg-25mg/kg body weight.
Further in another embodiment of the present invention, the compound provides elevation of pH in the range of 3.2-3.9 in ethanol induced ulceration at a dose ranging between Img/kg- 25mg/kg body weight
In another embodiment of the present invention, the compound provides reduction in acidity level in the range of 31.66-46.33mEq in ethanol induced ulceration at a dose ranging between lmg/kg-25mg/kg body weight.
In still another embodiment of the present invention, the compound provides reduction in gastric juice secretion at a range of 3-5 ml in ethanol induced ulceration at a dose ranging between lmg/kg-25mg/kg body weight.
In yet another embodiment of the present invention, the compound provides ulcer inhibition in the range of 40.48-92% induced by pylorus ligated ulceration at a dose ranging between lmg/kg-25mg/kg body weight.
In another embodiment of the present invention, the compound provides elevation of pH in the range of 3-4.4 in pylorus ligated induced ulceration at a dose ranging between lmg/kg- 25mg/kg body weight.
In still another embodiment of the present invention, the compound provides reduction in acidity level in the range of 48.33-31.33mEq in pylorus ligated induced ulceration- at a dose ranging between lmg/kg-25mg/kg body weight.
In another embodiment of the present invention, the compound provides reduction in gastric juice secretion at a range 1-2 ml in pylorus ligated induced ulceration at a dose ranging between lmg/kg-25mg/kg body weight.
In further embodiment, the present invention provides a pharmaceutical composition comprising compound of general formula 1, optionally along with a pharmaceutically acceptable carrier, additives, vehicle, and food preparation, useful for anti-ulcer activity.
In another embodiment of the present invention, the composition is administered through oral route at a dose ranging between l-25mg/kg body weight.
In still another embodiment, the present invention provides a method of treating ulcer in a subject wherein the said method comprising the steps of administering to the subject a compound of general formula 1 or a pharmaceutical composition.
In further embodiment of the present invention, the method comprises the steps of administering to the subject the compound or the pharmaceutical composition through oral route at a dose ranging between l-25mg/kg body weight.
In another embodiment of the present invention, the method is effective in treating gastrointestinal toxicity diseases induced by NSAIDs medication such as aspirin, stress, consumption of alcohol and related conditions.
The following examples are given by way of illustration of the present invention and should • not he construed to limit the scope of the present invention. Example 1:
Experimental protocol: process of isolation of Chrysin.
The dried powdered whole plant ( 5 Kg) was first defatted with petrol , then extracted successively with chloroform and ethanol in a soxhlet apparatus. The ethanol extract was partition with n-Butanol and this on concentration obtained brown semisolid n-butanol extract. This extract (100 g) was chromatographed over silica gel (60-120 mesh). The column was successively eluted with 20 % ethyl acetate in hexane to afford Chrysin. The yield of Chrysin is around 310 mg. Further elution of the column with 2% methanol in chloroform afforded compound I, the yield of compound I is around 5g. Further elution of the column with 3%methanol in chloroform afforded compound I, the yield of compound II is around 7g.
Further derivatives of Chrysin so produced are ■ τithesized as described in "Detailed Description of the invention".
Example 2: Experimental method for gastric ulcer The compounds taken under, study for antigastric ulcer screening by four different models were selected using experimental albino rats:
1. Aspirin induced gastric ulceration
2. Pylorus ligated gastric ulceration
3. Ethanol induced gastric ulceration 4. Stress induced gastric ulceration
The commercially available drug ranitidine (sigma), Omeprazole (sigma) and sucralfate (Merck) were used as reference standard in experimental models. The Tween- 80 (SD fine chemicals) was used as vehicle for the administration of the drug, which is used as control. The results obtained are presented in the following tables. 2.1 Acetyl salicylic acid induced ulcer:
The antiulcer activity of the compounds under taken was studied. The animals were divided into 18 groups of 6 animals each. Group 1 received the vehicle Tween 80 (1%, 1 ml) which served as the control. Group 2 received ranitidine at a dose of 50-m.g/kg body weight, which served as standard for comparison. Group 3 to 18 received Jl to J16 compounds respectively at a dose of 25 mg / kg body weight. Rats were administered per orally with a daily dose of the compounds and the drug ranitidine for a period of five days and then fasted for 24 hours. The narcotizing agent acetyl salicylic acid (aspirin) at a dose of 200 mg /kg body weight was administered as a suspension in tween-80 (1%), 30rnin after the drug administration each day. All drugs were administered orally on the 6th day after the last administration of the drugs and the ulcer inducing agent aspirin, the rats were killed by cervical dislocation and their stomach were opened along the greater curvature and washed with luke warm saline and examined under a dissecting microscope. The ulcer index was calculated for each stomach. The results are tabulated below in table no. 1.
Table No. 1 ulcer protective effect of ehrysin analogs in acetyl salicyclic acid (aspirin) induced gastric lesions.
Figure imgf000020_0001
Figure imgf000021_0001
Significance levels: *P<0.05, **P<0;01 when compared with the control values
2.2 Cold restraint induced ulcers
The antiulcer activity of the compounds was studied. The .animals were divided into 19 groups of 6 animals each. Group 1 received the vehicle Tween 80 (1%, 1 ml) which served as the control. Group 2 received ranitidine at a dose of 50-mg/kg body weight, which served as standard for comparison. Group 3 to 19 received Jl to Jl€ compounds respectively at a dose of 25 mg / kg body weight. Animals were deprived of food 48 hours before the experiment. The water was allowed for free access. Rats were administered per orally with compounds and the drug ranitidine. The water was removed 1 hour before restraint and exposed to a temperature of 40C for 2 hours. Two hours after stress, the animals were sacrificed. The stressed animals were opened along the greater curvature and the severity of gastric ulcer was assessed in terms of mean ulcer index. Results are tabulated below in table no.2.
Table No.2 Effect of chrysin analogs on gastric ulceration and mucosal mast cell count in stress rats.
Figure imgf000021_0002
Figure imgf000022_0001
Significance levels: *P<0.05, **P<0.01 when compared with the control values
2.3 Ethanol induced ulcers
The animals were divided into 20 groups of 6 animals each. Animals were deprived of food for 48 hours but had free access to water. Group 1 received the vehicle Tween 80 (1%, 1 ml) which served as the control. Group 2 received ranitidine at a dose of 50-mg/kg-body weight, which served as standard for comparison. Group 3 received Omeprazole at a dose of 30- mg/kg body weight, which served as standard for comparison and administered i.p. Group 4 received sucralfate at a dose of 400-mg/kg body weight, which served as standard for comparison. Group 5 to 16 received Jl to J12 compounds respectively at a dose of 25 mg / kg body weight and group 17 to 20 received Jl 3 to J20 compounds respectively at a dose of 50 mg/kg body weight. Lesions were induced 1 hour after ethanol challenge animals. The stomach was ligated at the pylorus under ether anesthesia. 4hours after pylorus ligation, -the animals were sacrificed and the contents drained and centrifuges at 5000 rpm for lOminutes. Aliquots of supernatant were used for determination of total acid by titrating with 0.01N NaOH using Topfer's reagent and phenolphthalein indicators. Results are tabulated below in table no.3
Table 3 Ulcer protective effect «f chrysin analogs in ethanol induced gastric ulcers
Figure imgf000023_0001
Significance levels: *P<0.05, **P<0.01 when compare .vith the control values
2.4 Pylorus ligated ulcers The animals were divided into 18 groups of 6 animals each. Animals were deprived of food for 4£
99 9 000154
hours but had free access to water. Group 1 received the vehicle Tween 80 (1%, 1 ml) which served as the control. Group 2 received ranitidine at a dose of 50-mg/kg-body weight, which served as standard for comparison. Group 3 to 14 received Jl to J12 compounds respectively at a dose of 25 mg / kg body weight and group 15 to 18 received Jl 3 to Jl 6 compounds respectively at a dose of 50 mg / kg body weight. After one hour of administration of drug the stomach was ligated at the pylorus under ether anesthesia. 4hours after pylorus ligation, the animals were sacrificed and the contents drained and centrifuges at 5000 rpm for lOminutes. Aliquots of supernatant were used for determination of total acid by titrating with 0.01N NaOH using topfers reagent and phenolphthalein indicators. Results are tabulated below in table no.4
Table. 4: Ulcer protective effects of chrysin and flavone analogs in pylorus ligated model
Figure imgf000024_0001
Ii
Figure imgf000025_0001
Significance levels: *P<0.05, **P<0.01 when compared with the control values
2.5 Acetyl salicylic acid induced ulcer:
The antiulcer activity of the compound J12 was studied. The animals were divided into 7 groups of 6 animals each. Group 1 received the vehicle Tween 80 (1%, 1 ml) which served as the control. Group 1 received ranitidine at a dose of 50-mg/kg-body weight, which served as standard for comparison. Group 3 to 7 received J12 compound at a dose of 25, 10,-5, 2, 1 mg / kg body weight respectively. Rats were administered per orally with a daily dose of the compounds and the drug ranitidine to respective groups for a period of five days and then fasted for 24 hours. The narcotizing agent acetyl salicylic acid (aspirin) at a dose of 200 mg /kg body weight was administered as a suspension in tween-80 (1%), 30min after the drug administration each day. All drugs were administered orally on the 6th day after the last administration of the drugs and the ulcer inducing agent aspirin, the rats were killed by cervical dislocation and their stomach were opened along the greater curvature and washed with luke warm saline and examined under a dissecting microscope. The ulcer index was calculated for each stomach. Results are tabulated below in table no. 5 Table 5 Ulcer protective effect of JK compound in acetyl salicyclic acid induced gastric lesions.
Figure imgf000025_0002
Figure imgf000026_0001
Significance levels: *P<0.05, **P<0.01 when compared with the control valuer 2.6 Cold restraint induced ulcers
The antiulcer activity of the compound Jl 2 was studied. The animals were divided into 8 groups of 6 animals each. Group 1 received the vehicle Tween 80 (1%, 1 ml) which served as the control. Group 2 received ranitidine at a dose of 50-mg/kg body weight, group 3 received diazepam at a dose of lmg/kg body weight, which served as standard for comparison. Group 4 to 8 received J12 compound at a dose of 25, 10, 5, 2 and 1 mg / kg body weight respectively. Animals were deprived of food 48 hours before the experiment. The water was allowed for free access. Rats were administered per orally with compounds and the drug ranitidine. The water was removed 1 hour before restraint and exposed to a temperature of 40C for 2 hours. Two hours after, stress, the animals were sacrificed. The stressed animals were opened along the greater curvature and the severity of gastric ulcer was assessed in terms of mean ulcer index. Results are tabulated below in table no. 6 Table 6 Effect of J12 on gastric ulceration and mucosal mast cell count in stress
Figure imgf000026_0002
Significance levels: *P<0.05, **P<0.01 when compared with the control values
2.7 Ethaπol induced ulcers
The animals λvere divided into 10 groups of 6 animals each. Animals were deprived of food for 48 hours but had free access to water. Group 1 received the vehicle Tween 80 (1%, 1 ml) which served as the control. Group 2 received ranitidine at a dose of 50-mg/kg-body weight, which served as standard for comparison. Group 3 received Omeprazole at a dose of 30- mg/kg body weight, which served as standard for comparison. Group 4 received sucralfate at a .dose of 400-mg/kg body weight, which served as standard for comparison. Group 5 to 10 received J12 compound at a dose of 25, 10, 5, 3, 2 and 1 mg / kg body weight respectively. Lesions were induced 1 hour after ethanol challenge animals. The stomach was ligated at the pylorus under ether anesthesia. 4hours after pylorus ligation, the animals were sacrificed and the contents drained and centrifuges at 5000 rpm for lOminutes. Aliquots of supernatant were used for determination of total acid by titrating with 0.01N NaOH using topfers reagent and phenolphthalein indicators. Results are tabulated below in table no. 7 Table 7 Ulcer protective effect of J12 in ethanol induced gastric ulcers
Figure imgf000027_0001
Significance levels: *P<0.05, **P<0.01 when compared with the control values
2.8 Pylorus ligated ulcers
The animals were divided into 7 groups of 6 animals each. Animals were deprived of food for 48 hours but had free access to water. Group 1 received the vehicle Tween 80 (1%, 1 ml) which served as the control. Group 2 received ranitidine at a dose of 50-mg/kg-body weight, which served as standard for comparison. Group 3 to 7 received Jl 2 compound at a dose of 25, 10, 5, 2 and 1 mg / kg body weight respectively. After one hour of administration of drug the stomach was ligated at the pylorus under ether anesthesia. 4hours after pylorus ligation, the animals were sacrificed and the contents drained and centrifuges at 5000 rpm for lOminutes. Aliquots of supernatant were used for determination of total acid by titrating with 0.0 IN NaOH using topfers reagent and phenolphthalein indicators. Results are tabulated below in table no. 8 Table 8 Ulcer Protective effect of J12 in pylorus ligated gastric ulcer model
Figure imgf000028_0001
Significance levels: *P<0.05, **P<0.01 when compared with the control values
Advantages of the invention;
Ulcers are thought to be imbalances in gastric offensive and defensive mucosal factors. While acid and pepsin make up the offensive factors, the defensive factors include mucin secretion, mucosal glycoprotein, cell proliferation etc. To regain the balance, different therapeutic agents are used to inhibit the gastric acid secretion or to boost the mucosal defense mechanisms by increasing mucus production, stabilizing the surface epithelial cells. The number of lesions present on the gastric mucosa is indicative of the severity of the ulcer disease.
When gastric mucosal barriers inflamed or disrupted by acid, ethanol or aspirin, stress deals to damage the cells and exacerbation of hemorrhagic ulcer. Thus, in ethanol induced, aspirin induced, pylorus ligated, stress induced models an attempt has been made to study the antiulcer potential of synthesized chrysin derivatives.
In the present study, J12 (benzoyloxy group substitution in 7-hydroxy flavone) at a dose level of 5mg/kg and J4 (allyloxy group substitution in chrysin) at dose level 25mg/kg found to provide gastro-protection following oral administration in aspirin induced model, pylorus ligation, ethanol induced and stress induced models induced in experimental rats for antiulcer potential of peptic ulcer. In present study, the standard ranitidine at dose of 50mg/kg body weight, omeprazole at dose of 30mg/kg body weight and sucralfate at dose of 400mg/kg body weight are taken for comparison. All three standard have shown good percentage of protection, ranitidine and omeprazole treated groups are comparable to each other, sucralfate treated group are higher.
The dose level of Jl 2 and J4 compounds are effective in protection against gastric ulcers far lower than the standard drugs dose levels. Gastric mucosal damages induced in different experimental ulcer models have different mechanisms. A favorable result here would indicate, it could provide protection from NSAID- induced darn^e, ethanol ulceration, stress induced ulceration, pylorus ligation on oral administration. Thus at lower dose 5 (Jl 2) and 25 (J4) mg/kg body weight this compound are effective against gastric ulcer induced by different ways, probably this can be efficient drug for the protection of gastric ulcer caused due to various factors, low family income, old age, smoking, lower educational attainment, ethnicity, increase gastric acids output, Helicobacter pylori infection, NSAIDs and stress that act as significant and independent basic risk factors in PUD risk factors. Gastric or peptic ulcer constitutes a major disease that affects human gastrointestinal tract and major health problem both in term of morbidity and motility. The common clinical features of peptic ulcer are hyperacid secretion and ulcer formation in the stomach and duodenal part of the intestine.
Za

Claims

We claim:
1. Use of a compound of general formula 1, useful for anti-ulcer activity, wherein the said compound comprising substituted chrysin or substituted 7-hydroxy flavone analogs,
Figure imgf000030_0001
Formula 1 wherein R' is selected from the group consisting of alkyl, alkenyl, alkoyl,benzoyl, hydrogen, carboxyfnethyl, acetamide and R" is selected from the group consisting of H, OH, alkoxy, acetoxy and propenyl.
2. Use of the compound as claimed in claim 1, wherein the representative compounds of the general formula 1 are selected from the group comprising: i. [7- allyloxy-5-hydroxy-2-phenyl-^H-l-benzopyran-4-one] - (J4) ii. [ 7-benzoyloxy— 2-phenyl- 4H— 1— benzopyran—4-one] - (J12)
3. Use of the compound as claimed in claim 1, wherein the said compound is administered through oral route at a dose ranging between lmg/kg-25mg/kg body weight.
4. Use of the compound as claimed in claim 1, wherein the said compound is effective in treating gastrointestinal toxicity diseases induced by NSAIDs medication such as aspirin, stress, alcohol consumption and related conditions administered at a dose ranging between lmg/kg-25mg/kg body weight.
5. Use of. the compound as claimed in claim 2, wherein [ 7-benzoyloxy-2-phenyl- 4H- l-benzopyran-4-one] provides ulcer inhibition at a range of 47.06-87.75% induced by aspirin ulceration at a dose ranging between lmg/kg-25mg/kg body weight.
6. Use of the compound as claimed in claim 2, wherein [ 7-benzoyloxy-2-phenyl- 4H- 5 1— benzopyran—4-one] provides glandular ulcer index inhibition at a range of 51.21 -
95.86 % induced by cold stress ulceration at a dose ranging between lmg/kg- 2Smg/kg body weight
7. Use of "the compound as claimed in claim 2, wherein [ 7-benzoyloxy-2-phenyl- 4H- 10 1— benzopyran—4-one] provides ulcer inhibition at a range of 57.63-96.45 % induced by ethanol ulceration at a dose ranging between lmg/kg-25mg/kg body weight
8. Use of the compound as claimed in claim 1, wherein the said compoun- >rovides elevation of pH in the range of 3.2-3.9 in ethanol induced ulceration at a dose
15 ranging between 1 mg/kg-25mg/kg body weight.
9. Use of the compound as claimed in claim 1, wherein the said compound provides reduction in acidity level in the range of 46.33-31.66 mEq in ethanol induced ulceration at a dose ranging between lmg/kg-25mg/kg body weight.
20
10. Use of the compound as claimed in claim 1, wherein the said compound provides reduction in gastric juice secretion at a range of 3.12-4.26 ml in ethanol induced ulceration at a dose ranging between lmg/kg-25mg/kg body weight.
25 11. Use of the compound as claimed in claim I5 wherein the said compound provides ulcer inhibition in the range of 40-92% induced by pylorus ligated ulceration at a dose ranging between lmg/kg-25mg/kg body weight.
12. Use of the compound as claimed in claim 1, wherein the said compound provides 30. elevation of pH in the range of 3-4.4 in pylorus ligated induced ulceration at a dose ranging between lmg/kg-25mg/kg body weight.
13. Use of the compound as claimed in claim 1, wherein the said compound provides reduction in acidity level in the range of 48.33-31.33 mEq in pylorus ligated induced ulceration at a dose ranging between lrng/kg-25mg/kg body weight.
14. Use of the compound as claimed in claim 1, wherein the said compound provides reduction in gastric juice secretion at a range of 1.9-1.8 ml in pylorus ligated induced ulceration at a dose Tanging between lmg/kg-25mg/kg body weight.
15. A pharmaceutical composition comprising compound of general formula 1, optionally along with a pharmaceutically acceptable carrier, additives, vehicle, food preparation, useful for anti-ulcer activity.
16. The pharmaceutical composition as claimed in claim 15, wherein the said composition is administered through oral route at a dose ranging between l-25mg/kg body weight.
17. A method of treating ulcer in a subject wherein the said method comprising the steps of administering to the subject a compound of general formula 1 as claimed in claiml or a pharmaceutical composition as claimed in claim 15.
18. The method of treating ulcer in a subject as claimed in claim 17, wherein the said method comprises the steps of administering to the subject the compound or the pharmaceutical composition through oral route at a dose ranging between l-25mg/kg body weight.
19. The method of treating ulcer in a subject as claimed in claim 17, wherein the said method is effective in treating gastrointestinal toxicity diseases induced by NSAIDs medication such as aspirin, stress, consumption of alcohol and related conditions.
PCT/IN2009/000154 2008-03-05 2009-03-05 Anti-ulcer activity of flavone analogs WO2009110008A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IN533DE2008 2008-03-05
IN533/DEL/2008 2008-03-05

Publications (1)

Publication Number Publication Date
WO2009110008A1 true WO2009110008A1 (en) 2009-09-11

Family

ID=40846942

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IN2009/000154 WO2009110008A1 (en) 2008-03-05 2009-03-05 Anti-ulcer activity of flavone analogs

Country Status (1)

Country Link
WO (1) WO2009110008A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH035424A (en) * 1989-06-01 1991-01-11 Kuraray Co Ltd Anti-ulcer agent containing flavonoids as active ingredient
US5399584A (en) * 1992-05-05 1995-03-21 The Procter & Gamble Company Use of flavone derivatives for gastroprotection
WO1998004541A1 (en) * 1996-07-25 1998-02-05 Dong A Pharmaceutical Co., Ltd. Gastroprotective flavone/flavanone compounds with therapeutic effect on inflammatory bowel disease
WO2005011671A1 (en) * 2003-08-05 2005-02-10 Lifetree Biotech Co., Ltd. Use of flavonoids for preventing and treating gastritis and gastric ulcer
WO2007080484A2 (en) * 2006-01-09 2007-07-19 Council Of Scientific & Industrial Research Flavonoids isolated from oroxylum indicum for treatment of gastrointestinal toxicity, associated symptoms and ulcers

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH035424A (en) * 1989-06-01 1991-01-11 Kuraray Co Ltd Anti-ulcer agent containing flavonoids as active ingredient
US5399584A (en) * 1992-05-05 1995-03-21 The Procter & Gamble Company Use of flavone derivatives for gastroprotection
WO1998004541A1 (en) * 1996-07-25 1998-02-05 Dong A Pharmaceutical Co., Ltd. Gastroprotective flavone/flavanone compounds with therapeutic effect on inflammatory bowel disease
WO2005011671A1 (en) * 2003-08-05 2005-02-10 Lifetree Biotech Co., Ltd. Use of flavonoids for preventing and treating gastritis and gastric ulcer
WO2007080484A2 (en) * 2006-01-09 2007-07-19 Council Of Scientific & Industrial Research Flavonoids isolated from oroxylum indicum for treatment of gastrointestinal toxicity, associated symptoms and ulcers

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
ARES J J ET AL: "Synthesis and biological evaluation of substituted flavones as gastroprotective agents.", JOURNAL OF MEDICINAL CHEMISTRY, vol. 38, no. 25, 8 December 1995 (1995-12-08), pages 4937 - 4943, XP002537211, ISSN: 0022-2623 *
KHANDHAR MAITREYI ET AL: "Antiulcer activity of the root bark of Oroxylum indicum against experimentalgastric ulcers", PHARMACEUTICAL BIOLOGY, vol. 44, no. 5, 1 July 2006 (2006-07-01), pages 363 - 370, XP009086903, ISSN: 1388-0209 *
TEWTRAKUL ET AL: "Anti-allergic activity of compounds from Kaempferia parviflora", JOURNAL OF ETHNOPHARMACOLOGY, vol. 116, no. 1, 6 November 2007 (2007-11-06), pages 191 - 193, XP022437358, ISSN: 0378-8741 *
TOMBOLA F ET AL: "Plant polyphenols inhibit VacA, a toxin secreted by the gastric pathogen Helicobacter pylori", FEBS LETTERS, vol. 543, no. 1-3, 22 May 2003 (2003-05-22), pages 184 - 189, XP004425058, ISSN: 0014-5793 *

Similar Documents

Publication Publication Date Title
US20110059913A1 (en) Compositions and methods for the treatment of gastrointestinal indications
US4244968A (en) Treatment of arthritis and substances for use in such treatment
EP2186792B1 (en) 2-(a-hydroxypentyl) benzoate and its preparation and use
US8853261B2 (en) Nutraceutical composition from Garcinia mangostana
TWI385178B (en) New diosmetin compounds, a process for their preparation and pharmaceutical compositions containing them
JPH0314286B2 (en)
JP3114895B2 (en) Pharmaceutical preparations for the treatment of inflammatory diseases
AU2005282468A1 (en) Improved GHB compositions
AU2004312445B2 (en) Pharmaceutical compositions comprising an extract of Euphorbia prostrata
JPS6150977A (en) Novel condensed benz(thio)amide, preparation thereof and pharmaceutical containing same as active constituent
EP0732329B1 (en) Chroman derivative
US5481043A (en) Phenone compounds, manufacturing method and pharmaceutical preparations containing them
WO2009110008A1 (en) Anti-ulcer activity of flavone analogs
WO2020125478A1 (en) Drug compound, preparation therefor, and application thereof
CN110563688B (en) Benzopyran compounds with anti-complement activity and application thereof
JPH06509110A (en) Methods and compositions for treating gastric and duodenal disorders and novel PGE↓2 derivatives
US20060198905A1 (en) Pharmaceutical compositions comprising an extract of euphorbia prostrata
US4447437A (en) Pharmaceutical composition and method for treatment of peptic ulcer
KR100508494B1 (en) Analgesic composition
KR100516647B1 (en) Hypoglycemic composition
JP4025817B2 (en) New flavan compounds
SU1600627A3 (en) Method of producing 2-substituted 1-napthols
JPH02250827A (en) Antibacterial agent, anti-inflammatory drug and external medicine for cutis
CN117942329A (en) Application of compound in preparation of medicine for treating and/or preventing cerebral apoplexy
WO1993019066A1 (en) Imidazopyridine derivative and medicine

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: 09717413

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09717413

Country of ref document: EP

Kind code of ref document: A1