Methods and pharmaceutical compositions for treating asthma or parkinson's disease

文档序号:638816 发布日期:2021-05-11 浏览:15次 中文

阅读说明:本技术 用于治疗哮喘或帕金森病的方法和药物组合物 (Methods and pharmaceutical compositions for treating asthma or parkinson's disease ) 是由 康仁哲 J·朴 于 2019-09-05 设计创作,主要内容包括:本发明涉及用于治疗哮喘的方法和包含氯胍的组合物。本发明还涉及用于治疗帕金森病的方法和包含氯胍的组合物。(The present invention relates to methods and compositions comprising proguanil for treating asthma. The invention also relates to methods and compositions comprising proguanil for treating parkinson's disease.)

1. A method of treating asthma in a subject in need thereof, the method comprising administering to the subject a pharmaceutical composition comprising a compound represented by formula 1:

[ formula 1]

2. The method of claim 1, wherein the composition further comprises a compound represented by formula 2:

[ formula 2]

3. The method of claim 1 or 2, wherein the asthma is allergen-induced asthma, virus-induced asthma, cold-induced asthma, pollution-induced asthma and/or exercise-induced asthma.

4. The method of any one of claims 1-3, wherein the composition is administered orally to the individual.

5. The method of any one of claims 1-4, wherein the composition comprises 0.001mg to 100mg of each compound per kilogram body weight of the individual.

6. A pharmaceutical composition comprising a compound represented by formula 1 or a pharmaceutically acceptable salt thereof, a compound represented by formula 2 or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient:

[ formula 1]

And

[ formula 2]

7. The composition of claim 6, wherein the composition is suitable for oral administration.

8. The composition of claim 6 or 7, wherein the composition comprises 1mg to 1000mg of each compound.

9. A method of treating parkinson's disease in a subject in need thereof, the method comprising administering to the subject a pharmaceutical composition comprising a compound represented by formula 1, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient:

[ formula 1]

10. The method of claim 9, wherein the composition further comprises a compound represented by formula 2:

[ formula 2]

11. The method of claim 9 or 10, wherein the composition is administered orally to the subject.

12. The method of any one of claims 9-11, wherein the composition comprises 0.001mg to 100mg of each compound per kilogram body weight of the individual.

Technical Field

The present disclosure relates to methods and compositions comprising proguanil for the treatment of asthma. The present disclosure also relates to methods and compositions comprising proguanil for the treatment of parkinson's disease.

Background

Asthma is a chronic inflammatory disease of the lower respiratory tract characterized by airway hyperresponsiveness and mucus obstruction. Bronchial asthma is the most common chronic disease affecting children and young adults. Strong evidence suggests the presence of a genetic component in asthma (Bleecker et al, am.j.respir.crit.care.med.,156: S113-6 (1997); Kauffmann et al, Chest,121(3 Supp.: 27S (2002)). A variety of environmental factors are also known to regulate clinical expression of asthma and asthma-associated phenotypes: bronchial hyperreactivity, atrophy, and IgE elevation (Koppelman et al, Eur. Resp.J.,13:2-4 (1999); Cookson, Nature,25: B5-11 (1999); Holloway, Clin. exp. allergy,29:1023-1032 (1999)).

Pharmacological analogs of cortisol (e.g., prednisone) have been used clinically since 1948 and remain the standard of care for the treatment of various inflammatory diseases, including asthma. These Glucocorticoids (GC) reduce the pathological inflammation of vital importance to Asthma and they are believed to control clinical Asthma symptoms through their anti-inflammatory action (Expert Panel Report 3(EPR-3): Guidelines for the Diagnosis and Management of asset-Summary Report 2007.J Allergy Clin. Immunol.2007,120: S94-138). There remains a need for improved asthma treatments.

Parkinson's disease is a disorder of voluntary movement in which muscles become stiff and sluggish, movement becomes awkward and difficult, and uncontrolled rhythmic twitching of muscle groups produces characteristic tremors or tremors. The disorder is thought to be caused by degeneration of pre-synaptic dopaminergic neurons in the brain. During neuronal activity, inadequate release of the chemical transmitter dopamine thus leads to parkinsonism symptoms.

The most widely used treatment for parkinsonism is the administration of levodopa (L-DOPA), a precursor of dopamine, which acts indirectly by replacing the missing dopamine. However, there are also disadvantages to using levodopa, for example, patients often suffer from side effects such as dyskinesia and switch effects, and levodopa must be administered in combination with a peripheral dopa decarboxylase inhibitor such as carbidopa or benzaseride. These inhibitors prevent peripheral degradation of levodopa to dopamine, thus enabling more drug to enter the brain and limiting peripheral side effects. Such treatment improves the quality of life of the patient, but does not prevent the development of the disease. In addition, such treatment is associated with a number of side effects, including nausea, vomiting, abdominal distension, and psychiatric side effects (e.g., toxic confusion states, paranoia, and hallucinations). There remains a need for improved treatments for parkinson's disease.

Brief description of the invention

Disclosed herein is a method of treating asthma in a subject in need thereof, the method comprising administering to the subject a pharmaceutical composition comprising a compound represented by formula 1, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient:

[ formula 1]

In some embodiments of the method, the composition further comprises a compound represented by formula 2:

[ formula 2]

Asthma may be, but is not limited to, allergen-induced asthma, virus-induced asthma, cold-induced asthma, pollution-induced asthma, and/or exercise-induced asthma.

In some embodiments of the method of treating asthma, the composition is administered to the individual orally. In some embodiments of the method of treating asthma, the composition comprises 0.001mg to 100mg of each compound per kilogram of body weight of the individual.

Also disclosed herein is a pharmaceutical composition comprising a compound represented by formula 1 or a pharmaceutically acceptable salt thereof, a compound represented by formula 2 or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient:

[ formula 1]

And

[ formula 2]

In some embodiments, the composition is suitable for oral administration to an individual. In some embodiments, the composition comprises 1mg to 1000mg of each compound.

Also disclosed herein is a method of treating parkinson's disease in a subject in need thereof, the method comprising administering to the subject a pharmaceutical composition comprising a compound represented by formula 1, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient:

[ formula 1]

In some embodiments of the method, the composition further comprises a compound represented by formula 2:

[ formula 2]

In some embodiments of the method of treating parkinson's disease, the composition is administered to the individual orally. In some embodiments of the method of treating parkinson's disease, the composition comprises 0.001mg to 100mg of each compound per kilogram body weight of the individual.

Brief Description of Drawings

Fig. 1. Effect of IPS-07004 on survival of a549 lung cancer cells.

Fig. 2. Effect of IPS-07004 on LPS-induced expression of IL-6 and IL-8 in A549 lung cancer cells.

Fig. 3. Effect of IPS-07004 on IL-6 expression in IL-1 β -induced A549 lung cancer cells.

Fig. 4. Effect of IPS-07004 on IL-8 expression in IL-1 β -induced A549 lung cancer cells.

Fig. 5. Effects of IPS-07004 and montelukast combination treatment on LPS-induced IL-6 and IL-8 expression in A549 lung cancer cells.

Fig. 6. Effect of IPS-07004 and montelukast combination treatment on IL-1 β -induced expression of IL-6 and IL-8 in a549 lung cancer cells.

Fig. 7. Inhibition of TSLP expression in mouse and HMC-1 cells by IPS-07004.

Fig. 8. Inhibition of mast cell number and serum IgE levels by IPS-07004.

Fig. 9. Effect of IPS-07005 on human Caspase-1 activity in HMC-1 cells.

Fig. 10A and 10B. Effect of IPS-07005 on PMA-induced expression of IL-6 and IL-8 in HMC-1 cells.

FIGS. 11A and 11B. Effect of combination treatment with IPS-07005 on IL-1 β -induced IL-6 and IL-8 expression in A549 lung cancer cells.

Fig. 12. Inhibition of OVA-specific IgE expression by IPS-07005.

Fig. 13A and 13B. IPS-07005 inhibition of total cell number and eosinophil number in bronchoalveolar lavage fluid (BAL). FIG. 13C. Histological image of eosinophils.

Fig. 14. Effect of IPS-07005 on OVA-induced changes in lung histology as determined by H & E staining (100 ×).

Fig. 15A and 15B. Airway histology images stained with PAS for goblet cell visualization.

Fig. 16A and 16B. IPS-07005 inhibition of IL17A and CXCL1 expression in BAL fluid.

Fig. 17A and 17B. IPS-07005 inhibits the expression of CCL3 and CCL2 in BAL liquid.

Fig. 18. Effect of IPS-07004 on Parkinson's Disease (PD) mouse behavior.

Fig. 19. Effect of IPS-07005 on tyrosine hydroxylase expression in the striatum and substantia nigra in PD brain.

Fig. 20. Effect of IPS-07005 on IL-1 β expression in PD brain.

Detailed Description

Proguanil, also known as chlorogenamide, is a prodrug that is converted by the liver to its active metabolite, cycloproguanil, which is a dihydrofolate reductase (DHFR) inhibitor. The present disclosure provides a composition comprising a compound represented by formula 1 or a pharmaceutically acceptable salt thereof:

[ formula 1]

The composition may further comprise montelukast (trade name SINGLULAIR) (a compound represented by formula 2) or a pharmaceutically acceptable salt thereof:

[ formula 2]

Montelukast is a leukotriene receptor antagonist (LTRA) used for maintenance treatment of asthma and relief of seasonal allergic symptoms. Montelukast is CysLT1An antagonist; it blocks leukotriene D4 (and secondary ligands LTC4 and LTE4) by binding to leukotriene D4 on cysteinyl leukotriene receptor CysLT in lung and bronchi1The function of (1). This reduces bronchoconstriction by leukotrienes and results in less inflammation.

Montelukast is used for a number of conditions including asthma, exercise-induced bronchospasm, allergic rhinitis, primary dysmenorrhea (i.e., dysmenorrhea not associated with a known etiology; see dysmenorrhea etiology) and urticaria. Montelukast is mainly used as a supplementary therapy for adults, in addition to inhaled corticosteroids, if the corticosteroid alone does not bring about the desired effect. It is also used to prevent allergic reactions and asthma outbreaks during intravenous administration of immunoglobulins.

The compositions disclosed herein may comprise proguanil, proguanil and montelukast, or proguanil and other anti-asthma agents, such as zileuton, zafirlukast, terbutaline sulfate, and albuterol. The compositions disclosed herein may also contain proguanil, montelukast and other agents useful in the treatment of parkinson's disease, such as levodopa, carbidopa, and/or benzaseride.

Pharmaceutically acceptable salts of the compounds disclosed herein include salts derived from: inorganic bases such as lithium (Li), sodium (Na), potassium (K), calcium (Ca), magnesium (Mg), iron (Fe), copper (Cu), zinc (Zn), and manganese (Mn); derived from organic bases such as N, N' -diacetylethylenediamine, glucosamine, triethylamine, chlorine, hydroxide, dicyclohexylamine, metformin, benzylamine, trialkylamine, thiamine, and equivalents thereof; chiral bases such as alkylanilines, glycinols, phenylglycinols and equivalents thereof; natural amino acid salts such as glycine, alanine, valine, leucine, isoleucine, norleucine, tyrosine, cystine, cysteine, methionine, proline, hydroxyproline, histidine, ornithine, lysine, arginine, serine, and equivalents thereof; containing alkyl sulfates such as alkyl halides, MeI and (Me)2SO4Quaternary ammonium salts of the compounds of the present invention and equivalents thereof; artificial amino acids such as D-isomer, substituted amino acids, etc.; guanidine, guanine, substituted by a group selected from: nitro, amino, alkyl, alkenyl and alkynyl groups, ammonium or substituted ammonium and aluminium salts. Salts may include acid addition salts, and examples of suitable salts include sulfate, nitrate, phosphate, perchlorate, borate, hydrohalide, acetate, tartrate, maleate, citrate, fumarate, succinate, palmitate, methanesulfonate, benzoate, salicylate, benzenesulfonate, ascorbate, glycerophosphate, ketoglutarate, and equivalents thereof. Pharmaceutically acceptable solvent compounds include crystallization solvents such as hydroxides or alcohols.

In addition to salt forms, the present disclosure provides compounds in prodrug form. Prodrugs of the compounds described herein are those compounds that readily undergo chemical changes under physiological conditions to provide the compounds of the present disclosure. In addition, prodrugs can be converted to the compounds of the present disclosure by chemical or biochemical methods in an ex vivo environment. For example, a prodrug can be slowly converted to a compound of the present disclosure when placed in a transdermal patch reservoir along with a suitable enzyme or chemical agent. Prodrugs are often useful because, in some cases, they are easier to administer than the parent drug. For example, they may be bioavailable by oral administration whereas the parent drug is not. The prodrug may also have a higher solubility in the pharmaceutical composition than the parent drug. Various prodrug derivatives are known in the art, such as those that rely on hydrolytic cleavage or oxidative activation of the prodrug. Non-limiting examples of prodrugs are compounds of the present disclosure that are administered as esters ("prodrugs") but are then metabolically hydrolyzed to carboxylic acids, i.e., the active entities. Additional examples include peptidyl derivatives of the compounds of the present disclosure.

Certain compounds of the present disclosure may exist in unsolvated as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are intended to be encompassed within the scope of the present disclosure. Certain compounds of the present disclosure may exist in polycrystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated by the present disclosure and are intended to be within the scope of the present disclosure.

Certain compounds of the present disclosure have asymmetric carbon atoms (optical centers) or double bonds; racemates, enantiomers, diastereomers, geometric isomers and individual isomers are all intended to be included within the scope of the present disclosure. These isomers can be resolved or asymmetrically synthesized using conventional methods to render the isomers "optically pure," i.e., substantially free of their other isomers. For example, if a particular enantiomer of a compound of the present disclosure is desired, it may be prepared by asymmetric synthesis, or by derivatization with a chiral auxiliary, wherein the resulting diastereomeric mixture is separated and the auxiliary group is cleaved to provide the pure desired enantiomer. Alternatively, where the molecule contains a basic functional group such as an amino group or an acidic functional group such as a carboxyl group, diastereomeric salts are formed with an appropriate optically active acid or base, and the diastereomers thus formed are then resolved by fractional crystallization or chromatographic methods well known in the art, followed by recovery of the pure enantiomers.

Composition comprising a metal oxide and a metal oxide

Also disclosed herein is a pharmaceutical composition comprising a compound represented by formula 1 or a pharmaceutically acceptable salt thereof, a compound represented by formula 2 or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient:

[ formula 1]

And

[ formula 2]

In some embodiments, the composition is suitable for oral administration to an individual. In some embodiments, the composition comprises 1mg to 1000mg of each compound.

In some aspects, the present disclosure provides pharmaceutical compositions suitable for pharmaceutical use comprising one or more compounds of the present disclosure and a pharmaceutically acceptable excipient or carrier. The compositions disclosed herein may comprise proguanil, proguanil and montelukast, or proguanil and other anti-asthma agents, such as zileuton, zafirlukast, terbutaline sulfate, and albuterol. The compositions disclosed herein may also contain proguanil, montelukast and other agents useful in the treatment of parkinson's disease, such as levodopa, carbidopa, and/or benzaseride.

As used herein, "pharmaceutically acceptable" or "pharmacologically acceptable" refers to molecular entities and compositions that do not produce adverse, allergic, or other untoward reactions when administered to an animal or human, as appropriate.

The term "pharmaceutically acceptable excipient or carrier" includes one or more inert excipients including starches, polyols, granulating agents, microcrystalline cellulose, diluents, lubricants, binders, disintegrating agents and the like, as well as any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. The invention relates to the use thereof in therapeutic compositions, except insofar as any conventional media or agent is incompatible with the active ingredient. Supplementary active ingredients may also be incorporated into the composition. "pharmaceutically acceptable excipients" also include controlled release means.

The formulations can improve one or more pharmacokinetic properties (e.g., oral bioavailability, membrane permeability) of a compound or combination of compounds of the present disclosure (referred to herein as an active ingredient).

The composition, shape and type of dosage form may generally vary depending on its application. For example, a dosage form suitable for mucosal administration may comprise a lower amount of the active ingredient than a dosage form suitable for oral administration for treating the same disease. These aspects of the invention will be apparent to those of ordinary skill in the art (ref: Remington's Pharmaceutical Sciences (1990) 18 th edition, Mack Publishing, Easton PA).

Typical pharmaceutical compositions and dosage forms include one or more excipients. Suitable excipients will be apparent to those of ordinary skill in the pharmaceutical art, and the present disclosure is not limited to the examples of suitable excipients described herein.

Whether a particular excipient is suitable for use in a pharmaceutical composition or dosage form depends on various factors well known in the art, including, but not limited to, the method of formulating the formulation to be administered to a patient. For example, a dosage form for oral administration, such as a tablet, may include excipients that are not suitable for use in a formulation for non-oral administration.

Pharmaceutical compositions include those suitable for aerosol, pulmonary, inhalation, oral, parenteral (including subcutaneous, intradermal, intramuscular, intravenous and intraarticular), rectal and topical (including dermal, buccal, sublingual and intraocular) administration. The most suitable route may depend on the condition and disorder of the recipient. The compositions may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Generally, the compositions are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired composition.

The compositions of the present disclosure may also optionally contain other therapeutic ingredients, anti-caking agents, preservatives, sweeteners, colorants, flavorants, desiccants, shaping agents, dyes, and the like. Of course, any such optional ingredients must be compatible with the compounds of the present disclosure to ensure stability of the composition.

The active agent, alone or in combination with other suitable components, may be administered by the pulmonary route using several techniques including, but not limited to, intratracheal instillation (delivery of the solution into the lungs via a syringe), intratracheal liposome delivery, insufflation (administration of the powder composition into the lungs via a syringe or any other similar device), and aerosol inhalation. Compositions suitable for inhalation by the pulmonary route include sterile solutions for nebulization comprising a therapeutically effective amount of the compound dissolved in saline solution, and optionally containing a preservative such as benzalkonium chloride or chlorobutanol, and aerosol compositions comprising a therapeutically effective amount dissolved or suspended in a suitable propellant. Aerosols (e.g., jet or ultrasonic nebulizers, Metered Dose Inhalers (MDIs), and Dry Powder Inhalers (DPIs)) may also be used for intranasal applications. Aerosols may conveniently be presented in unit dosage form and prepared by any of the methods well known in the art of pharmacy. Aerosol compositions are stable dispersions or suspensions of solid materials and liquid droplets in a gaseous medium and can be placed in pressurized acceptable propellants, such as hydrofluoroalkanes (HFA, i.e., HFA-134a and HFA-227, or mixtures thereof), dichlorodifluoromethane (or other chlorofluorocarbon propellants, such as mixtures of propellants 11,12 and/or 114), propane, nitrogen, and the like. Pulmonary compositions may include penetration enhancers such as fatty acids and sugars, chelating agents, enzyme inhibitors (e.g. protease inhibitors), adjuvants (e.g. glycocholate, lipopeptide, span 85 and nafamostat), preservatives (e.g. benzalkonium chloride or chlorobutanol) and ethanol (typically up to 5% but possibly up to 20% by weight). Ethanol is typically included in aerosol compositions because it can improve the function of the metering valve and in some cases also improve the stability of the dispersion. The pulmonary composition may also include surfactants including, but not limited to, bile salts and those described in U.S. patent No. 6,524,557 and references therein. Surfactants described in us patent 6,524,557, such as C8-C16 fatty acid salts, bile salts, phospholipids or alkyl sugars, are advantageous because some of them are reported to also enhance absorption of the compounds in the composition.

Also suitable are dry powder compositions comprising a therapeutically effective amount of the active compound in admixture with a suitable carrier, suitable for use with a dry powder inhaler. Absorption enhancers that may be added to the dry powder compositions of the present disclosure include those described in U.S. patent 6,632,456. WO02/080884 describes novel methods for modifying the surface of powders. Aerosol compositions may include U.S. Pat. No. 5,230,884, U.S. Pat. No. 5,292,499, WO017/8694, WO01/78696, US2003019437, US20030165436, and WO96/40089 (which includes vegetable oils). Sustained release compositions suitable for inhalation are described in US20010036481A1, US20030232019A1 and US20040018243A1 and WO01/13891, WO02/067902, WO03/072080 and WO 03/079885. Pulmonary compositions comprising microparticles are described in WO03/015750, US20030008013 and WO 00/00176. Pulmonary compositions comprising a stable glassy powder are described in US20020141945 and US patent No. 6,309,671. Other aerosol compositions are described in EP 1338272a1, WO90/09781, U.S. patent 5,348,730, U.S. patent 6,436,367, WO91/04011 and U.S. patent 6,294,153 as well as U.S. patent 6,290,987, which describe a liposome-based composition that can be administered by aerosol or other means. Powder compositions for inhalation are described in US20030053960 and WO 01/60341. The agent may be administered intranasally as described in US 20010038824.

Although the pulmonary route is advantageous in most cases, there may be other situations where other routes of administration may be advantageous. For example, oral administration may be desirable. In this regard, it is contemplated to use compositions in which the compounds are releasably encapsulated by modified amino acids, as described in U.S. patent 5,811,127. One may also consider administration in implantable sustained release dosage forms, as described in US 20040115236.

The pharmaceutical compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs. Compositions for oral use may be prepared according to any method known in the art for preparing pharmaceutical compositions. Such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with other non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. These excipients may be, for example, inert diluents such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, such as corn starch or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc. Tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate may be employed. They may also be identified by U.S. Pat. nos. 4,256,108; 4,160,452, respectively; and 4,265,874 to form osmotic therapeutic tablets for controlled release.

Tablets may be prepared by compression or moulding, optionally together with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, lubricant, surfactant or dispersing agent. Molded tablets may be prepared by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide sustained, delayed or controlled release of the active ingredient therein.

Compositions for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin, or olive oil.

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

Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Examples of suitable dispersing or wetting agents and suspending agents are those already mentioned above. Other excipients, for example sweetening, flavoring and coloring agents, may also be present.

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

The pharmaceutical compositions and methods of the present disclosure may further comprise other therapeutically active compounds for the treatment of asthma, allergic diseases, and inflammatory conditions, as described herein. In many cases, compositions comprising a compound disclosed herein and an alternative active agent have an additive or synergistic effect when administered.

Pharmaceutical compositions for administration of the compounds of the present disclosure may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art. All methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. Generally, pharmaceutical compositions are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired composition. In pharmaceutical compositions, the active object compound is present in an amount sufficient to produce the desired effect on the process or condition of the disease.

Application method

The term "subject" or "patient" is defined herein to include animals, such as mammals, including, but not limited to, primates (e.g., human or non-human), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, and the like. In some embodiments, the individual is a human.

As used herein, the term "preventing" refers to a method of delaying or precluding the onset of a disease and/or its attendant symptoms, arresting or reducing the risk of an individual acquiring a disease.

As used herein, the term "treating" is intended to include alleviating, reducing or eliminating a disease or disorder and/or its attendant symptoms as well as alleviating, reducing or eradicating the cause of the disease itself.

The term "therapeutically effective amount" means the amount of the subject compound that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician. The term "therapeutically effective amount" includes an amount of a compound that, when administered, is sufficient to prevent the development of, or alleviate to some extent, one or more of the symptoms of the condition or disorder being treated. The therapeutically effective amount will vary depending on the compound, the disease and its severity and the age, weight, etc., of the mammal to be treated.

Disclosed herein is a method of treating asthma in a subject in need thereof, the method comprising administering to the subject a pharmaceutical composition comprising a compound represented by formula 1, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient:

[ formula 1]

Also disclosed herein is a method of treating asthma in a subject in need thereof, the method comprising administering to the subject a pharmaceutical composition comprising a compound represented by formula 1 or a pharmaceutically acceptable salt thereof, a compound represented by formula 2 or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient:

[ formula 1]

And

[ formula 2]

Also disclosed herein are methods of treating asthma in a subject in need thereof, comprising administering to the subject a pharmaceutical composition comprising proguanil and an additional anti-asthma agent, such as zileuton, zafirlukast, terbutaline sulfate, and albuterol. Methods of preventing and/or treating asthma include allergen-induced asthma, virus-induced asthma, cold-induced asthma, pollution-induced asthma and/or exercise-induced asthma.

In some embodiments of the method of treating asthma, the composition is administered to the individual orally. In some embodiments of the method of treating asthma, the composition comprises 0.001mg to 100mg of each compound per kilogram of body weight of the individual.

Also disclosed herein is a method of treating parkinson's disease in a subject in need thereof, the method comprising administering to the subject a pharmaceutical composition comprising a compound represented by formula 1, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient:

[ formula 1]

Also disclosed herein is a method of treating parkinson's disease in a subject in need thereof, the method comprising administering to the subject a pharmaceutical composition comprising a compound represented by formula 1 or a pharmaceutically acceptable salt thereof, a compound represented by formula 2 or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient:

[ formula 1]

And

[ formula 2]

In some embodiments of the methods of treating parkinson's disease, the composition is administered to the subject orally. In some embodiments of the method of treating parkinson's disease, the composition comprises 0.001mg to 100mg of each compound per kilogram body weight of the individual.

Depending on the disease to be treated and the condition of the individual, the compounds of the present disclosure may be administered by aerosol, pulmonary, inhalation, oral, parenteral (e.g., intramuscular, intraperitoneal, intravenous, ICV, intracisternal injection or infusion, subcutaneous injection or implant), inhalation, nasal, vaginal, rectal, sublingual, or topical (e.g., transdermal, topical) administration routes, and may be formulated, alone or together, in suitable dosage unit compositions containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles appropriate for each route of administration. The present disclosure also relates to administering a compound of the present disclosure in a depot composition, wherein the active ingredient is released over a defined period of time.

The dosage range of the active ingredient or a pharmaceutically acceptable salt thereof for an adult human is generally from about 0.005 mg/day to about 10 g/day, from about 1 mg/day to about 1,000 mg/day, from about 10 mg/day to about 750 m/day, from about 50 mg/day to about 500 mg/day or from about 75 mg/day to about 350 mg/day. A dosage form provided in discrete units, in tablets or other forms, may conveniently contain an amount of a compound of the disclosure which is effective at such a dose, or as multiple doses thereof, for example, units containing from 5mg to 500mg, usually from about 10mg to 200 mg. The precise amount of the compound administered to the patient will be at the responsibility of the attending physician. However, the dosage employed will depend upon a number of factors, including the age and sex of the patient, the exact condition being treated and its severity.

In the treatment or prevention of asthma, a suitable dosage level is typically about 0.001 to 100mg per kg body weight of the patient per day, which may be administered in single or multiple doses. Dosage levels may be from about 0.01 mg/kg/day to about 75 mg/kg/day or from about 0.05 mg/kg/day to about 20 mg/kg/day. Suitable dosage levels may be from about 0.01 mg/kg/day to about 50 mg/kg/day, from about 0.05 mg/kg/day to about 20 mg/kg/day, or from about 0.1 mg/kg/day to about 10 mg/kg/day. Doses within this range may be 0.01 mg/kg/day to 0.1 mg/kg/day, 0.1 mg/kg/day to 1 mg/kg/day, 1 mg/kg/day to 10 mg/kg/day. For oral administration, the compositions may be provided in the form of tablets containing from 1mg to 1000mg of the active ingredient, such as but not limited to from 1mg, 5mg, 10mg, 15mg, 20mg, 25mg, 50mg, 75mg, 100mg, 150mg, 200mg, 250mg, 300mg, 400mg, 500mg, 600mg, 750mg, 800mg, 900mg and 1000mg of the active ingredient, for symptomatic dose adjustment in the patient to be treated. The compounds may be administered on a regimen of 1 to 4 times per day, for example once, twice, three times or four times per day.

Similarly, in the treatment or prevention of Parkinson's disease, suitable dosage levels are typically from about 0.001 to 100mg/kg patient body weight/day, which can be administered in single or multiple doses. Dosage levels may be from about 0.01 mg/kg/day to about 75 mg/kg/day or from about 0.05 mg/kg/day to about 20 mg/kg/day. Suitable dosage levels may be from about 0.01 mg/kg/day to about 50 mg/kg/day, from about 0.05 mg/kg/day to about 20 mg/kg/day, or from about 0.1 mg/kg/day to about 10 mg/kg/day. Doses within this range may be 0.01 mg/kg/day to 0.1 mg/kg/day, 0.1 mg/kg/day to 1 mg/kg/day, 1 mg/kg/day to 10 mg/kg/day. For oral administration, the compositions may be provided in the form of tablets containing from 1mg to 1000mg of the active ingredient, such as, but not limited to, 1mg, 5mg, 10mg, 15mg, 20mg, 25mg, 50mg, 75mg, 100mg, 150mg, 200mg, 250mg, 300mg, 400mg, 500mg, 600mg, 750mg, 800mg, 900mg, and 1000mg of the active ingredient, for symptomatic dose adjustment of the patient to be treated. The compounds may be administered on a regimen of 1 to 4 times per day, for example once, twice, three times or four times per day.

In some embodiments, the dose of proguanil or montelukast is about 0.05 mg/day to about 1000 mg/day, including any and all variations within this range, such as about 0.1 mg/day to about 1000 mg/day, about 1 mg/day to about 1000 mg/day, about 2.5 mg/day to about 100 mg/day, about 5 mg/day to about 50 mg/day, about 10 mg/day to about 750 mg/day, about 50 mg/day to about 500 mg/day, or about 75 mg/day to about 350 mg/day.

In some embodiments, montelukast is provided in the form of tablets, chewable tablets, and granules for oral ingestion. Montelukast may be taken once daily with or without food. Montelukast can be administered as montelukast sodium, where 5.2mg montelukast sodium is equivalent to 5mg montelukast.

It will be understood, however, that the specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.

The compounds of the present disclosure may be combined or used in combination with other active agents that may be useful in the treatment, prevention, inhibition, or amelioration of the diseases or conditions for which the compounds of the present disclosure are useful.

These other active agents or drugs can be administered by the route and amount commonly used therefor, either simultaneously or sequentially with the compounds of the present disclosure. When the compounds of the present disclosure are used simultaneously with one or more other drugs, pharmaceutical compositions containing the other drugs in addition to the compounds of the present disclosure are preferred. Accordingly, the pharmaceutical compositions of the present disclosure include those that contain one or more other active ingredients or therapeutic agents in addition to the compounds of the present disclosure.

The weight ratio of the compound of the present disclosure to the second active ingredient can vary and will depend on the effective dose of each ingredient. Generally, an effective dose of each will be used. Thus, for example, when a compound of the present disclosure is combined with an NSAID, the weight ratio of the compound of the present disclosure to NSAID is typically from about 1000:1 to about 1:1000, or from about 200:1 to about 1: 200. Combinations of the compounds of the present disclosure and other active ingredients will generally also be within the foregoing ranges, but in each case, an effective dose of each active ingredient should be used.

Hereinafter, the present invention will be described in further detail with reference to the following non-limiting examples. However, the examples are provided for illustrative purposes only and are not intended to limit the scope of the present invention.

Examples

Example 1. In vitro cell viability assay (MTT).

IPS-07004 (proguanil) was purchased from Key Organics (London, UK).

A549 lung cancer cell (1x 10)3Per well) were seeded in 96-well plates for 16 hours. After 24 hours of starvation. Cells were pre-treated with LPS or IL-1b for 4 hours. Cells were then treated with IPS-07004 in a dose-dependent manner for 24 hours. After 48 hours of incubation, MTT [3- (4, 5-dimethylthiazol-2-yl) -2, 5-diphenyl-tetrazolium bromide was added to each well in a total volume of 1/10]The solution (5mg/ml) was incubated at 37 ℃ for 2 hours.

To examine the survival of a549 cells treated with IPS-07004, an in vitro cell survival assay was performed using MTT. IPS-07004 had little or no effect on viability when cells were induced by LPS (lipopolysaccharide) and IL-1 β (interleukin-1 β) (FIG. 1).

Example 2. Enzyme-linked immunosorbent assay (ELISA) for IL-6 and IL-8.

A549 cells (1X 10)5Per well) were seeded in 96-well plates for 16 hours. After 24 hours of starvation, cells were pretreated with LPS or IL-1. beta. for 4 hours. Cells were then treated with IPS-07004 in a dose-dependent manner for 24 hours. Cytokine levels (IL-6 and IL-8) in the medium were analyzed according to the manufacturer's instructions (BD Biosciences Pharmingen, San Diego, Calif., USA).

To assess whether IPS-07004 inhibits the expression of IL-6 and IL-8, which play a critical role in asthma pathology, an ELISA for cytokines in A549 cells was used.

IPS-07004 reduced the LPS (FIG. 2) and IL-1 β (FIGS. 3 and 4) induced expression levels of IL-6 and IL-8 in A549 cells in a dose-dependent manner. These data indicate that IPS-07004 can be used to treat asthma.

Example 3. Effect of IPS-07004 and montelukast combination treatment on LPS or IL-1 β induced expression of IL-6 and IL-8 in a549 lung cancer cells.

To test the synergistic effect of IPS-07004 in combination with montelukast, an anti-asthmatic, on anti-asthmatic function, an assay for cytokine expression in a549 cells was performed.

The combination of IPS-07004 with montelukast was shown to be more effective in inhibiting the expression of IL-6 and IL-8 in A549 cells induced by LPS (FIG. 5) and IL-1 β (FIG. 6) compared to treatment with montelukast alone. These data indicate that combination treatment of IPS-07004 with montelukast can be used to treat asthma.

Example 4. Compound characteristics of IPS-07005

Indication of asthma

Caspase-1 inhibitors from the NLRP3 inflammasome

Leading material IPS-07004

Comprises IPS-0700410 mg/kg (mpk) + Montelukast 1.0mpk

Stage of development phase I

FIG. 7 shows the inhibitory effect of IPS-07004 on caspase-1 activity and TSLP (thymic stromal lymphopoietin) expression in HMC-1 cells and DNCB (2, 4-dinitrochlorobenzene) mice.

FIG. 8 shows the inhibitory effect of IPS-07004 on mast cell number and serum IgE levels.

Example 5. Inhibition of asthma by IPS-07005-in vitro cell-based assays

Results

Using an assay based on HMC-1 (human mast cell line) cells, IPS-07005((IPS-07004+ montelukast) inhibits the activity of caspase-1, a key component of the inflammatory corpuscle, to inhibit the conversion of Pro-TSLP, IL-1 β, and IL-18 to active forms, as shown in FIG. 9.

IPS-07004 inhibits caspase-1 activity in HMC-1 cells in a dose-dependent manner alone, with IPS-07005 providing a synergistic effect.

FIG. 10 shows the effect of IPS-07005 on IL-6 and IL-8 expression in PMA (phorbol 12-myristate 13-acetate) and A23187 (calcium ionophore (C1)) induced HMC-1 cells. IPS-07005 inhibited the expression of IL-6 and IL-8 in HMC-1 cells compared to IPS-07004, montelukast alone, or dexamethasone.

To assess whether IPS-07005 inhibited the expression of IL-6 and IL-8, which play a critical role in asthma pathology, an ELISA for cytokines in A549 lung cancer cells was used. IPS-07005 reduced the IL-1 β -induced expression levels of IL-6 and IL-8 in A549 cells in a dose-dependent manner (FIG. 11). IPS-07005 inhibited the expression of IL 6 and IL-8 in A549 cells (human lung cancer cells) compared to IPS-07004, montelukast alone, or dexamethasone.

These data indicate that IPS-07005 is suitable for use in the treatment of asthma.

Example 6. Inhibition of asthma by IPS-07005-in vivo animal data

FIG. 12 shows the inhibitory effect of IPS-07005 on OVA-specific IgE expression. IPS-07005(IPS-0700410mpk + montelukast 1mpk) showed a significant reduction in OVA-induced IgE levels in bronchoalveolar lavage fluid (BALF).

FIGS. 13A and 13B show the inhibitory effect of IPS-07005 on total cell number and eosinophil number in bronchoalveolar lavage (BAL) fluid. FIG. 13C shows that the number of eosinophils in OVA-challenged cells was reduced when treated with IPS-07004 or IPS-07005. Analysis of inflammatory cells in BALF samples showed that OVA sensitization significantly increased the total cell number. However, treatment with IPS-07005 reduced the total number of cells. In particular, administration of IPS-07005 also significantly reduced the total cell number and eosinophil number in BALF in the corresponding group compared to OVA challenge group (fig. 13A and 13B).

FIG. 14 shows the effect of IPS-07005 on OVA-induced changes in lung histology as determined by H & E staining (100X). Fig. 14 shows histological images of airways stained with PAS for visualization of goblet cells. Lung sections were stained with PAS (periodic acid-Schiff) to assess their goblet cell proliferation levels. We observed a clear difference in the size of the purple regions (i.e. the lung tissue regions stained with PAS) in the three groups. We noted goblet cell proliferation and mucus overproduction in OVA-stimulated mouse bronchial passages. However, we noted that the numbers of goblet cells in IPS-07005 and high dose montelukast treated groups were significantly lower than in OVA challenged groups (fig. 15A and 15B).

FIG. 16 shows the inhibitory effect of IPS-07005 on the expression of IL17A and CXCL1 in BAL fluid. FIG. 17 shows the inhibitory effect of IPS-07005 on the expression of CCL3 and CCL2 in BAL fluid.

To determine the anti-asthmatic effect of IPS-07005 on cytokine levels in asthmatic mice, the production of IL-17A, CXCL1, CCL2 and CCL3 in BAL fluid was measured. Exposure to OVA caused a significant increase in IL-17A, CXCL1, CCL2 and CCL3 in BAL fluid compared to normal group of mice (fig. 16 and 17). However, in OVA-exposed mice in the presence of IPS-07005, the level of IL-17A, CXCL1, which plays an important role in neutrophil infiltration, was reduced in BAL fluid after OVA exposure (FIG. 16).

This result strongly suggests that IPS-07005 is capable of inhibiting neutrophilic asthma, a medically unmet need for the treatment of asthma. IPS-07005 reduced the levels of CCL2 and CCL3, which play a key role in macrophage activity in BAL fluid (fig. 17). It was shown to be more effective in reducing the expression of these cytokines compared to IPS-07004 or montelukast. The inhibitory effect of IPS-07005 on the expression of these cytokines is due to the synergistic effect of IPS-07004 and montelukast. Taken together, these findings demonstrate that the anti-inflammatory effects of IPS-07005 are mediated by the modulation of a variety of inflammatory factors.

Method

25mg of Ovalbumin (OVA) and 1mg of aluminum hydroxide hydrate (AlOH3) were dissolved in 300mL of a sterile saline solution and sensitized by intraperitoneal administration on days 0 and 21. At the end of the observation period, all live animals were anesthetized with isoflurane and blood was collected from the vena cava/abdominal aorta. The lungs were removed and washed with sterile saline solution. 250mL of sterile saline was filled into a 1mL syringe, and the injection and recovery were repeated three times. The washed solution was gently centrifuged (300rpm, 10 min), and the supernatant was frozen and used for analysis of CXCL1(ab216951, Abcam PLC) IL-17A (ab199081, Abcam PLC) and CCL3(ab100726, Abcam PLC). The pellet was resuspended in the same amount of sterile saline solution and used for eosinophil and total cell counts. The collected blood was left at room temperature for 30 minutes and centrifuged at 3,000rpm for 20 minutes. Sera were isolated for total IgE and Ova-specific IgE analysis. Histopathological examination was performed using formalin-fixed lung tissue. Lung tissue extracted at the time of dissection was placed in cassettes and immersed in 10% neutral buffered formalin (10% NBF) at least 20 times the tissue volume. After fixation, the tissue was cut to a thickness of about 4mm, and paraffin blocks were prepared after general tissue processing and embedding. Then, the sections were cut to 4 μm thickness with a rotary microtome, and hematoxylin & eosin (H & E) staining and PAS staining were performed.

Example 7. Inhibition of parkinson's disease

Method

For 1-methyl-4-phenyl-1, 2,3, 6-tetrahydropyridine (MPTP) poisoning, mice received four intraperitoneal injections of MPTP-HCl (20mg/kg, free base in saline; Sigma-Aldrich, St. Louis, Mo) in PBS at 2 hour intervals. Twelve hours after the last MPTP injection, MPTP-injected mice received either IPS-07004, montelukast, or IPS-07005 once a day by oral administration.

To investigate the effect of IPS-07004 on the abnormal behaviour of PD mice, pole climbing tests (T-turn and T-LA times) were performed. PD was induced in mice treated with MPTP (20mg/kg, I.P) and administered to mice IPS-07004 at a dose of 10mg/kg, and then a pole climbing test was performed in the mice, which can measure bradykinesia, which is a hallmark of Parkinson's disease. T turn the time for a black mouse to turn down from the top of the pole was measured. As shown in FIG. 18, IPS-07004 reduced T-turn and T-LA times compared to MPTP-treated control and levodopa-treated groups.

To determine the anti-PD function of IPS-07004 and IPS-07005 in PD mice, we analyzed the expression of Tyrosine Hydroxylase (TH) in the striatum and substantia nigra in PD brain. Immunohistochemical analysis in brain showed a decrease in TH levels in IPS-07004 and 07005 treated mice compared to MPTP treated control and levodopa treated groups, as shown in fig. 19.

To assess whether the anti-PD function of IPS-07004 and 07005 was due to inflammation inhibition in the PD brain, an analysis of cytokine expression was performed. IPS-07004 and IPS-07005 reduced the expression of IL-1b in PD mice, as shown in FIG. 20.

The foregoing description of the specific embodiments will so fully reveal the general nature of the invention that others can, by applying knowledge within the skill of the art, readily modify and/or adapt for various applications such specific embodiments without departing from the general concept of the present invention. Therefore, such adaptations and modifications are intended to be within the meaning and range of equivalents of the disclosed embodiments, based on the teaching and guidance presented herein. It is to be understood that the phraseology or terminology herein is for the purpose of description and not of limitation, such that the terminology or phraseology of the present specification is to be interpreted by the skilled artisan in light of the teachings and guidance.

The breadth and scope of the present invention should not be limited by any of the above-described exemplary embodiments, but should be defined only in accordance with the following claims and their equivalents.

All of the various aspects, embodiments and options described herein can be combined in any and all variations.

All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.

39页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:H3R反向激动剂用于治疗与帕金森病(PD)相关的日间过度嗜睡的用途

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!