WO2009063202A2 - Use of crth2 antagonist compounds - Google Patents

Use of crth2 antagonist compounds Download PDF

Info

Publication number
WO2009063202A2
WO2009063202A2 PCT/GB2008/003824 GB2008003824W WO2009063202A2 WO 2009063202 A2 WO2009063202 A2 WO 2009063202A2 GB 2008003824 W GB2008003824 W GB 2008003824W WO 2009063202 A2 WO2009063202 A2 WO 2009063202A2
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
fluoro
acetic acid
indol
compound
Prior art date
Application number
PCT/GB2008/003824
Other languages
French (fr)
Other versions
WO2009063202A3 (en
Inventor
Michael George Hunter
Eric Roy Pettipher
Colin Michael Perkins
Mark Anthony Payton
Luzheng Xue
Original Assignee
Oxagen Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Oxagen Limited filed Critical Oxagen Limited
Priority to JP2010532660A priority Critical patent/JP2011503045A/en
Priority to DE08851028T priority patent/DE08851028T1/en
Priority to EP08851028A priority patent/EP2219645A2/en
Publication of WO2009063202A2 publication Critical patent/WO2009063202A2/en
Publication of WO2009063202A3 publication Critical patent/WO2009063202A3/en
Priority to US12/779,638 priority patent/US20110124683A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • A61K31/405Indole-alkanecarboxylic acids; Derivatives thereof, e.g. tryptophan, indomethacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/4161,2-Diazoles condensed with carbocyclic ring systems, e.g. indazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/498Pyrazines or piperazines ortho- and peri-condensed with carbocyclic ring systems, e.g. quinoxaline, phenazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/14Decongestants or antiallergics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to the use of CRTH2 antagonist compounds for the pulsed treatment of allergic conditions.
  • Allergic conditions are becoming more common throughout the developed world with as much as 10% of the world's population being affected by one or more of allergic asthma, allergic rhinitis, atopic dermatitis and other allergic conditions. Numerous classes of compound have been suggested for the treatment of these conditions, with one of the more recent developments being the use of CRTH2 antagonists, which inhibit the action of agonists such as prostaglandin D 2 (PGD 2 ) acting at the CRTH2 receptor.
  • PPD 2 prostaglandin D 2
  • PGD 2 is an eicosanoid, a class of chemical mediator synthesised by cells in response to local tissue damage, normal stimuli or hormonal stimuli or via cellular activation pathways. Eicosanoids bind to specific cell surface receptors on a wide variety of tissues throughout the body and mediate various effects in these tissues. PGD 2 is known to be produced by mast cells, macrophages and Th2 lymphocytes and has been detected in high concentrations in the airways of asthmatic patients challenged with antigen (Murray et al, (1986), N. Engl. J. Med. 315: 800-804). Instillation of PGD 2 into airways can provoke many features of the asthmatic response including bronchoconstriction (Hardy et al, (1984) N.
  • the first receptor specific for PGD 2 to be discovered was the DPi receptor which is linked to elevation of the intracellular levels of cAMP.
  • PGD 2 is thought to mediate much of its proinflammatory activity through interaction with a G protein- coupled receptor termed CRTH2 (chemoattractant receptor-homologous molecule expressed on Th2 cells) which is expressed by Th2 lymphocytes, eosinophils and basophils (Hirai et al, (2001) J. Exp. Med. 193: 255-261, and EPO851O3O and EP-A- 1211513 and Bauer et al, EP-A-1170594).
  • CRTH2 chemoattractant receptor-homologous molecule expressed on Th2 cells
  • the selective DP agonist BW245C does not promote migration of Th2 lymphocytes or eosinophils (Hirai et al, 2001; Gervais et al, (2001) J. Allergy Clin. Immunol. 108: 982-988).
  • antagonising PGD 2 at the CRTH2 receptor is an attractive approach to treat the inflammatory component of Th2-dependent allergic diseases such as asthma (including allergic asthma), food allergies, acute and chronic urticaria, perennial allergic rhinitis, seasonal allergic rhinitis, atopic dermatitis, contact hypersensitivity (including contact dermatitis) and conjunctivitis, especially allergic conjunctivitis.
  • Documents which discuss the use of CRTH2 antagonists for the treatment of Th2- dependent allergic diseases include WO-A-03/066046, WO-A-03/066047, WO-A- 03/097042, WO-A-03/097598, WO-A-03/101981, WO-A-03/101961, WO-A- 2004/007451, WO-A-2005/019171, WO-A-2005/094816, WO-A-2005/044260, WO-A-2005/040112, WO-A-2005/040114 and WO2006/095183.
  • ramatroban which has been shown to reduce allergic inflammation in the guinea pig nasal mucosa (Narita et al, Im Arch Allergy Immunol 1996;109: 161-166), mouse airways (Nagai et al, Prostaglandins 1995;50:75-87) and mouse skin (Takeshita et al, Int. Immunol. 2004,16:947-959).
  • Ramatroban has also shown to reduce symptoms of perennial allergic rhinitis in human subjects (Terada et al, Allergol. Int. 1998; 47: 59-67).
  • CRTH2 antagonists are effective in treating allergic asthma, allergic rhinitis, allergic conjunctivitis, atopic dermatitis and other allergic conditions, the inventors have now made the surprising discovery that these compounds have an effect which is much longer lasting than previously thought and are thus useful preventing or reducing the symptoms of the allergic condition for a prolonged period of time after dosing.
  • R 1 is C 1 -C 6 alkyl
  • R 2 is halogen
  • R 3 is aryl or heteroaryl optionally substituted with one or more substituents selected from halo, OH, CN, R 6 , COR 6 , CH 2 R 6 , OR 6 , SR 6 , SO 2 R 6 or SO 2 YR 6 ;
  • R 6 is C 1 -C 6 alkyl, C 3 -Cg cycloalkyl, heterocyclyl, aryl or heteroaryl, any of which may optionally be substituted with one or more substituents selected from halo, OH, CN, NO 2 , C 1 -C 6 alkyl or 0(C 1 -C 6 alkyl); and Y is NH or a straight or branched C 1 -C 4 alkylene chain;
  • R 4 is H or C 1 -C 4 alkyl
  • m is 1 or 2;
  • n is 1-4;
  • X is OR 7 or N(R 7 ) 2 ;
  • R 7 is hydrogen or methyl
  • R 8 is C 1 -C 18 alkyl
  • the treatment comprises at least one treatment cycle which comprises: a. a first period during which the compound of general formula (I) is administered to the patient; and b. a second period of at least seven days during which the compound of general formula (I) is administered to the patient in a reduced amount; wherein the allergen is present or suspected to be present throughout the treatment and wherein the compound of general formula (I) retains at least 50% of its effectiveness during the second period of the treatment cycle.
  • the invention arises from observations made in the course of a double blind crossover study of a CRTH2 antagonist of general formula (I) in human subjects with allergic rhinitis.
  • the study compared two groups of patients. The first patient group was treated daily with the CRTH2 antagonist for 8 consecutive days, then had no treatment for 3 weeks, then had daily treatment with placebo for 8 consecutive days. The second patient group was treated with placebo for 8 consecutive days, then had no treatment for 3 weeks, then had daily treatment with the CRTH2 antagonist of general formula (I) for 8 consecutive days.
  • the study showed that in the first treatment period, patients treated with the CRTH2 antagonist of general formula (I) displayed fewer symptoms of allergic rhinitis than patients treated with a placebo and this result was expected by the inventors.
  • the first patient group which was being treated with placebo, still retained a reduction in symptoms, the results being comparable with those of the second patient group which was being treated with the CRTH2 antagonist of general formula (I) at that time.
  • These patients also displayed fewer symptoms than they had displayed in an initial screening before treatment with either CRTH2 antagonist or placebo.
  • CRTH2 antagonists such as the compounds of general formula (I) as defined above have a prolonged effect on the allergic response and that, therefore, these compounds are of use in a pulsed method for the treatment of an allergic condition which is induced by the presence of an allergen to which the patient is sensitive, the method comprising administering to a patient over at least one treatment cycle a CRTH2 antagonist compound of general formula (I), wherein the said treatment cycle comprises: a. a first period during which a compound of general formula (I) is administered to the patient; and b.
  • the invention also provides the use of a CRTH2 antagonist compound of general formula (I) in the preparation of an agent for the treatment of an allergic condition which is induced by the presence of an allergen to which the patient is sensitive, wherein the treatment comprises at least one treatment cycle which comprises: a. a first period during which the compound of general formula (I) is administered to the patient; and b.
  • CRTH2 antagonist compounds such as the compounds of general formula (I) in the treatment of allergic conditions is not completely clear but the inventors speculate that it may be related to a previously unknown effect of CRTH2 antagonists on Th2 cells.
  • PGD 2 plays an important role to play in Th2 cell survival by preventing the apoptosis of Th2 cells and their subsequent clearance from the allergic tissue.
  • CRTH2 antagonists are able to block the effect of CRTH2 and induce apoptosis of Th2 cells.
  • Th2 lymphocytes take part in the cascade leading to an allergic reaction as described by various authors, including Pettipher et al, Nature Reviews, 6 (April 2007), 313-325.
  • the immune response in allergy begins with the presentation of allergens to antigen presenting cells which internalise the antigens and cause them to be expressed on the cell surface. This initiates a cascade of events leading to the production of IgE, which binds to high affinity receptors on mast cells leaving its allergen-specific site available for interaction with allergen.
  • mast cell-bound IgE Upon re-exposure to the same allergen, cross-linking of mast cell-bound IgE initiates the production of a number of factors, including PGD 2 by the mast cells, which in turn leads to the recruitment and activation of Th2 cells via the interaction of PGD 2 with the CRTH2 receptor.
  • the activated Th2 cells produce various cytokines including IL4, IL5 and IL13.
  • IL4 causes the production of further IgE by B cells.
  • the inventors have postulated that the apoptotic effect of CRTH2 antagonists on Th2 lymphocytes interrupts the cascade by promoting apoptosis and clearance of Th2 cells which leads to a reduction in the levels of the Th2 cytokines IL4, IL5 or IL13.
  • the reduction in IL4 and IL 13 production would lead to a decrease in the production of IgE by the B-lymphocytes. Therefore it appears that the immune system would not be able to respond to an allergen by increased production of IgE. This would lead to a significantly decreased immune response to an allergen until such time as the Th2 cell population is replenished.
  • C 1 -C 6 alkyl refers to a straight or branched saturated hydrocarbon chain having one to six carbon atoms and optionally substituted with one or more halo substituents or with one or more C 3 -O 7 cycloalkyl groups.
  • Examples include methyl, ethyl, n-propyl, isopropyl, t-butyl, n-hexyl, trifluoromethyl, 2-chloroethyl, methylenecyclopropyl, methylenecyclobutyl, methylenecyclobutyl and methylenecyclopentyl.
  • C 1 -C 4 alkyl and “C 1 -C 18 alkyl” have similar meanings except that they contain from one to four and from one to eighteen carbon atoms respectively.
  • C 3 -C 7 cycloalkyl refers to a saturated 3 to 7 membered carbocyclic ring. Examples of such groups include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
  • C 1 -C 4 alkylene in the context of the present specification refers to a disubstituted straight or branched saturated hydrocarbon chain having one to four carbon atoms.
  • halo refers to fluoro, chloro, bromo or iodo.
  • aryl in the context of the present specification refers to an aromatic ring system having from 5 to 14 ring carbon atoms and containing up to three rings. Examples of aryl groups are benzene and naphthalene.
  • heteroaryl in the context of the specification refers to a ring system with aromatic character having from 5 to 14 ring atoms, at least one of which is a heteroatom selected from N, O and S, and containing up to three rings. Where a heteroaryl group contains more than one ring, not all rings must be fully aromatic in character. Rings which are not fully aromatic may be substituted with one or more oxo groups.
  • heteroaryl groups include pyrrole, thiophene, thiazole, pyridine, pyrimidine, indole, benzofuran, benzimidazole, tetrahydroquinoline, indoline, quinoline, isoquinoline, quinoxaline, imidazo[l,2-a]pyridine, pyrazolo[l,5- a]pyridine, 2,3-dihydro-l-benzothiopyrane and 2,3-dihydro-l ⁇ 6 -benzothiopyran-l,l- dione.
  • heterocyclyl in the context of the specification refers to a saturated ring system having from 4 to 8 ring atoms, at least one of which is a heteroatom selected from N, O and S and which may be optionally substituted by one or more oxo groups.
  • heterocyclyl groups include azetidinyl, piperidinyl; tetrahydrofuranyl, tetrahydropyranyl, dioxanyl, thiomorpholinyl, 1,1-dioxo-l ⁇ - thiomorpholinyl, morpholinyl, pyrrolyl, piperizinyl, azepanyl, 1,4-diazepanyl, 1,4- oxazepanyl and azocanyl.
  • Appropriate pharmaceutically and veterinarily acceptable salts of the compounds of general formula (I) include basic addition salts such as sodium, potassium, calcium, aluminium, zinc, magnesium and other metal salts as well as choline, diethanolamine, ethanolamine, ethyl diamine, megulmine and other well known basic addition salts as summarised in J. Med. Chem., 50, 6665-6672 (2007) and/or known to those skilled in the art.
  • pharmaceutically or veterinarily acceptable salts may also include salts of organic acids, especially carboxylic acids, including but not limited to acetate, trifluoroacetate, lactate, gluconate, citrate, tartrate, maleate, malate, pantothenate, adipate, alginate, aspartate, benzoate, butyrate, digluconate, cyclopentanate, glucoheptanate, glycerophosphate, oxalate, heptanoate, hexanoate, fumarate, nicotinate, pamoate, pectinate, 3-phenylpropionate, picrate, pivalate, proprionate, tartrate, lactobionate, pivolate, camphorate, undecanoate and succinate, organic sulfonic acids such as methanesulfonate, ethanesulfonate, 2-hydroxyethane sulfonate, camphorsulfonate, 2-naphthalate,
  • Salts which are not pharmaceutically or veterinarily acceptable may still be valuable as intermediates.
  • Prodrugs are any covalently bonded compounds which release the active parent drug according to general formula (I) in vivo.
  • X is OR 7 or N(R 7 ) 2 ;
  • R 7 is hydrogen or methyl
  • R 8 is C 1 -C 18 alkyl.
  • a chiral centre or another form of isomeric centre is present in a compound of the present invention, all forms of such isomer or isomers, including enantiomers and diastereoisomers, are intended to be covered herein.
  • Compounds of the invention containing a chiral centre may be used as a racemic mixture, an enantiomerically enriched mixture, or the racemic mixture may be separated using well-known techniques and an individual enantiomer may be used alone.
  • administered in combination with refers to the co-administration of a CRTH2 antagonist with another drug wherein the administration may be simultaneous, sequential or separate.
  • allergic condition refers to a condition in which the atopic state of a patient is increased above that of subject who does not have an allergic condition. This may be characterised by the presence in the circulation or tissues of a patient of increased numbers of cells reactive to allergens, and by the presence of IgE which binds specifically to an allergen to which the patient is sensitive.
  • the IgE may be circulating or bound, for example to the surface of mast cells and basophils.
  • references herein to a patient being "sensitive" to an allergen mean that when the patient is exposed to the allergen, an allergic response is induced such that the patient will display symptoms of inflammation arising from the production of substances such as histamine, prostaglandin D 2 and tryptase by mast cells and basophils when the allergen binds to IgE expressed on their surfaces.
  • References to "the treatment of an allergic condition” refer to the reduction or removal of the inflammatory symptoms which arise from the exposure of a patient to an allergen to which the patient is sensitive.
  • references herein to an antigen being present or suspected to be present thoughout the treatment cycle are intended to mean that the antigen may be present in the patient's environment either constantly or intermittently and in varying amounts.
  • the pollen to which they are sensitive may be present in the environment throughout the hay fever season but the amount of pollen in the environment may vary depending on the weather, the time of day and other conditions.
  • a patient may suspect that the allergen is present in the environment, even if no tests are carried out to confirm this.
  • a patient would conventionally take medication as a precautionary measure if it was suspected that pollen may be present, as might occur, for example, during the hay fever season.
  • the term "effectiveness" as applied to the compounds of general formula (I) refers to the reduction of the allergic response in the patient.
  • the patient will experience a reduction of at least 30% in symptoms, suitably at least 40% and with increasing order of suitability, at least 50%, at least 60%, at least 70% at least 80% and, in the most favourable cases, at least a 90% reduction in symptoms.
  • rhinitis and conjunctivitis assessment may be carried out, for example in a Vienna Challenge Chamber experiment, which is well known to those of skill in this art and in which patients carry out self-assessment and scoring of their symptoms on a scale of 0 to 3.
  • Separate scores may be given for eye symptoms, nasal symptoms (including nasal obstruction, nasal itch, sneeze and rhinorrhea) and other symptoms.
  • the symptom score for each patient is subjective, if a sufficient number of patients is used, the total scores are meaningful.
  • other clinical trials may be designed and the measurement of reduction in symptoms may either be carried out by a subjective method such as self assessment by patients or, alternatively, may be made by an objective measurement.
  • asthma symptoms may be quantified using measurements of lung function such as forced expiratory volume in one second (FEVi) or peak expiratory flow rate (PEF) or using the Juniper quality of life scale.
  • FEVi forced expiratory volume in one second
  • PEF peak expiratory flow rate
  • Juniper quality of life scale The severity of atopic dermatitis symptoms can be assessed using the scoring atopic dermatitis (SCORAD) or six area six sign atopic dermatitis (SASSAD) systems.
  • SCORAD scoring atopic dermatitis
  • SASSAD six area six sign atopic dermatitis
  • the compound of general formula (I) retains at least 50% of its effectiveness, but it is more suitable that the compound of general formula (I) retains at least 60% of its effectiveness and, in increasing order of suitability, 70%, 80% and 90% of its effectiveness.
  • the amount of compound of general formula (I) administered to the patient during the second period of the treatment cycle is reduced compared with the amount administered during the first period.
  • the daily dose of compound of general formula (I) administered during the second period of the treatment cycle will be not greater than 50% of the daily dose administered during the first period. However, it may be less than this, for example not greater than 40%, not greater than 30%, not greater than 20% or not greater than 10% of the daily dose of compound of general formula (I) administered during the first period.
  • administration of the CRTH2 antagonist compound will be discontinued altogether during the second period of the treatment cycle.
  • the daily dose with be zero and no compound of general formula (I) will be administered to the patient. This is a particularly advantageous result as period during which the medication is necessary will be considerably shorter than for conventional medications, which has positive implications both for the general well being of the patient and the cost of the treatment.
  • the second period of the treatment cycle lasts for at least 7 days. However, it may last for longer periods, for example at least 14 days and it has been observed that the drug may retain at least 50% of its effectiveness during second periods lasting at least 21 days, at least 28 days and, in some cases, in excess of 35 days even when the dose of compound is reduced to zero during the second period of the treatment cycle.
  • the first period of the treatment cycle during which the CRTH2 antagonist is administered may be chosen such that it comprises a period of days that provides the maximum biological response to the compound in the patient. In general, this period may be from two to 8 days, during which time the drug may be administered from one to three times a day. It is particularly suitable however that the CRTH2 antagonist is administered to the patient once daily as this helps to increase patient compliance.
  • one treatment cycle may not be sufficient to last for the whole time over which treatment is required and therefore the treatment may include one or more further treatment cycles. This is often the case with conditions such as seasonal allergic rhinitis, where the hay fever season may last for several months.
  • CRTH2 antagonists such as the compounds of general formula (I) are useful in treating any allergic condition but they are particularly useful in the pulsed treatment of conditions such as asthma (including allergic asthma), food allergies, acute and chronic urticaria, perennial allergic rhinitis, seasonal allergic rhinitis, atopic dermatitis, contact hypersensitivity (including contact dermatitis) and conjunctivitis, especially allergic conjunctivitis.
  • the CRTH2 antagonists of general formula (I) are indole- 1 -acetic acid derivatives and are some of the compounds and analogues thereof described in WO2005/044260, WO2005/094816 and WO2006/095183, WO2008/012511 and UK patent application Nos 0800874.0, filed 18 January 2008; 0801132.2, filed 22 January 2008; 0801671.9, filed 30 January 2008; 0801131.4, filed 22 January 2008; 0801672.7, filed 30 January 2008; and 0801674.3, filed 30 January 2008.
  • the indole- 1-acetic acid derivatives of general formula (I) as defined above are described in detail in these documents.
  • the compound of general formula (I) is a CRTH2 antagonist in which R 5 is hydrogen.
  • X is OR 7 or N(R 7 ) 2 ;
  • R 7 is hydrogen or methyl
  • R 8 is Ci-C 18 alkyl.
  • R 1 is fluoro
  • R 2 is C 1 -C 4 alkyl, particularly methyl or ethyl but more especially methyl;
  • R 4 is H or methyl; and
  • R 3 is quinoline, quinoxaline, isoquinoline, thiazole, phenyl, naphthalene, thiophene, pyrrole or pyridine, any of which may optionally be substituted as set out above.
  • R 4 is H.
  • R 3 groups include optionally substituted quinoline, phenyl, naphthalene, thiophene, pyrrole or pyridine.
  • R 3 is quinoline or isoquinoline, it is suitably unsubstituted or substituted with one or more halo substituents, especially fluoro.
  • R 3 is phenyl, naphthalene, thiophene, pyrrole or pyridine, it may optionally have one or more substituents, with particularly suitable substituents including OR 6 , SO 2 R 0 or SO 2 YR 0 ; where R >6 0 and Y are as defined above.
  • R 6 is C 1 -C 6 alkyl, a 4- to 6-membered cycloalkyl group, a 5- or 6-membered heterocyclyl group or phenyl, any of which may be substituted as defined above.
  • R 3 is pyridyl it is most suitably a 3-pyridyl moiety.
  • Y when present, is a CH 2 moiety.
  • R 6 group is generally unsubstituted or substituted with one or more substituents chosen from methyl and halo, particularly chloro or fluoro.
  • group When group may be unsubstituted or substituted with one or more substituents chosen from halo, cyano, C 1 -C 4 alkyl and 0(Ci-C 4 alkyl).
  • Particularly suitable compounds of general formula (I) include: ⁇ 3-[l-(4-Chloro-phenyl)-ethyl]-5-fluoro-2-methyl-indol-l-yl ⁇ -acetic acid;
  • X is OR 7 or N(R 7 ) 2 ;
  • R 7 is hydrogen or methyl
  • R 8 is C 1 -C 18 alkyl.
  • the compounds of general formula (I) may be prepared according to methods set out in the prior art or methods analogous to those set out in the prior art, in particular WO2005/044260, WO2005/094816 and WO2006/095183, WO2008/012511 and UK patent application Nos 0800874.0, 0801132.2, 0801671.9, 0801131.4, 0801672.7 and 0801674.3.
  • the compound of general formula (I) may be combined with one or more additional agent which is of use in the treatment of these allergic conditions.
  • additional agent which is of use in the treatment of these allergic conditions.
  • agents include existing therapies for allergic and other inflammatory diseases including:
  • B 2 adrenoreceptor agonists such as metaproterenol, isoproterenol, isoprenaline, albuterol, salbutamol, formoterol, salmeterol, terbutaline, orciprenaline, bitolterol mesylate ,pirbuterol and indacaterol or methylxanthanines such as theophylline and aminophylline, mast cell stabilisers such as sodium cromoglycate or muscarinic receptor antagonists such as ipratropium and tiotropium; antihistamines, for example histamine H 1 receptor antagonists such as loratadine cetirizine, desloratadine, levocetirizine, fexofenadine, astemizole, azelastine and chlorpheniramine or H 4 receptor antagonists; (X 1 and ⁇ 2 adrenoreceptor agonists such as propylhexedrine phenylep
  • the CRTH2 antagonist of general formula (I) will generally be administered in a pharmaceutical formulation which may be a formulation suitable for oral, rectal, nasal, bronchial (inhaled), topical (including eye drops, buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration and may be prepared by any methods well known in the art of pharmacy.
  • a pharmaceutical formulation which may be a formulation suitable for oral, rectal, nasal, bronchial (inhaled), topical (including eye drops, buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration and may be prepared by any methods well known in the art of pharmacy.
  • compositions for oral, nasal, bronchial or topical administration.
  • the formulation may be prepared by bringing into association the above defined active agent with a carrier.
  • the formulations are prepared by uniformly and intimately bringing into association the active agent with liquid carriers or finely divided solid carriers or both, and then if necessary shaping the product.
  • Formulations for oral administration in the present invention may be presented as: discrete units such as capsules, sachets, tablets, which may be chewable tablets, or lozenges, each containing a predetermined amount of the active agent; as a powder or granules; as fine particles for sprinkling over food; as a solution or a suspension of the active agent in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water in oil liquid emulsion; or as a bolus etc.
  • the term "acceptable carrier” includes vehicles such as common excipients e.g. binding agents, for example syrup, acacia, gelatin, sorbitol, tragacanth, polyvinylpyrrolidone (Povidone), methylcellulose, ethylcellulose, sodium carboxymethylcellulose, hydroxypropylmethylcellulose, sucrose and starch; fillers and carriers, for example corn starch, gelatin, lactose, sucrose, microcrystalline cellulose, kaolin, mannitol, dicalcium phosphate, sodium chloride and alginic acid; and lubricants such as magnesium stearate, sodium stearate and other metallic stearates, glycerol stearate stearic acid, silicone fluid, talc waxes, oils and colloidal silica.
  • Flavouring agents such as peppermint, oil of wintergreen, cherry flavouring and the like can also be used. It may be desirable to
  • a tablet may be made by compression or moulding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine the active agent in a free flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface-active or dispersing agent.
  • Moulded tablets may be made by moulding 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 slow or controlled release of the active agent.
  • compositions suitable for oral administration include lozenges comprising the active agent in a flavoured base, usually sucrose and acacia or tragacanth; pastilles comprising the active agent in an inert base such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active agent in a suitable liquid carrier.
  • CRTH2 antagonists such as compounds of general formula (I) may be made up into a cream, ointment, jelly, solution or suspension etc.
  • Cream or ointment formulations that may be used for the drug are conventional formulations well known in the art, for example, as described in standard text books of pharmaceutics such as the British Pharmacopoeia.
  • CRTH2 antagonists such as compounds of general formula (I) may be made up into an eye drop formulation.
  • Suitable eye drop formulations are also well known in the art and are described in standard text books of pharmaceutics such as the British Pharmacopoeia.
  • CRTH2 antagonists which are intended to reduce or remove the symptoms of conditions such as allergic asthma, allergic conjunctivitis and allergic rhinitis may be administered to the respiratory tract by nasal, bronchial or buccal administration of, for example, aerosols or sprays which can disperse the pharmacological active ingredient in the form of a powder or in the form of drops of a solution or suspension.
  • Pharmaceutical compositions with powder-dispersing properties usually contain, in addition to the active ingredient, a liquid propellant with a boiling point below room temperature and, if desired, adjuncts, such as liquid or solid non-ionic or anionic surfactants and/or diluents.
  • compositions in which the pharmacological active ingredient is in solution contain, in addition to this, a suitable propellant, and furthermore, if necessary, an additional solvent and/or a stabiliser.
  • a suitable propellant instead of the propellant, compressed air can also be used, it being possible for this to be produced as required by means of a suitable compression and expansion device.
  • Parenteral formulations will generally be sterile.
  • the oral dose of the compound will be about 0.01 to 100 mg/kg; so as to achieve a concentration of drug in the plasma at a concentration effective to inhibit PGD 2 at the CRTH2 receptor.
  • the precise amount of a compound of CRTH2 antagonist which is therapeutically effective, and the route by which such compound is best administered, is readily determined by one of ordinary skill in the art by comparing the blood level of the agent to the concentration required to have a therapeutic effect.
  • the concentration of active compound will typically be about 0.1%-1% w/w such that systemic exposure of the CRTH2 antagonist compound is limited.
  • CRTH2 antagonists for the treatment of Th2-dependent allergic diseases include WO-A-03/066046, WO-A- 03/066047, WO-A-03/097042, WO-A-03/097598, WO- A-03/101981, WO-A- 03/101961, WO-A-2004/007451, WO-A-2005/019171, WO-A-2005/054232, WO- A-2004/089884, WO-A-2004/089885, WO-A-2005/018529, WO-A-2006/005909, WO2006/021759, WO-A-2007/039736, WO-A-2007/052023, WO-A-2006/075139, WO-A-2007/068894, WO- A-2007138282,
  • a first period during which the CRTH2 antagonist compound is administered to the patient and b. a second period of at least seven days during which the compound of general formula (I) is administered to the patient in a reduced amount; wherein the allergen is present or suspected to be present throughout the treatment and wherein the CRTH2 antagonist retains at least 50% of its effectiveness during the second period of the treatment cycle.
  • Some particularly suitable compounds are compounds which are similar to the compounds of general formula (I) but in which the moiety -CHR 3 R 4 of general formula (I) is replaced by R 3 , SR 3 , SOR 3 or SO 2 R 3 , where R 3 is as defined for general formula (I).
  • An example compound of this type, [5-Fluoro-3-(4-chloro- benzenesulfonyl)-2-methyl-indol-l-yl]-acetic acid, which falls within the scope of WO 2004/007451 was tested in the apoptosis assay of Example 3 (below) and was found counter the rescuing function of PGD 2 with an IC 50 of 204 ⁇ 160 nM.
  • a CRTH2 antagonist is a compound which binds to the CRTH2 receptor with a Ki of less than 1000 nM in the radioligand binding assay described below and which inhibits the dose-dependent increase in intracellular Ca 2+ mobilisation in CHO/CRTH2 cells treated with PGD 2 with an IC 50 of less than 1000 nM.
  • the CRTH2 antagonist is suitably a compound of general formula (I).
  • Calcium-3 dye was purchased from Molecular Devices (Wokingham, UK). Mono- -poly resolving medium was obtained from Dainippon Pharmaceuticals (Osaka,
  • Macs anti-CD 16 microbeads were from Miltenyi biotec (Bisley, Surrey).
  • ChemoTx plates were purchased from Neuroprobe (Gaithesburg, MD). PoIy-D- lysine coated 96-well plates were obtained from Greiner (Gloucestershire, UK).
  • [ 3 H]PGD 2 was from Amersham Biosciences (Buckinghamshire, UK).
  • [ 3 H]SQ29548 was purchased from Perkin Elmer Life Sciences (Buckinghamshire, UK). All other reagents were obtained from Sigma-Aldrich (Dorset, UK), unless otherwise stated.
  • Cell culture Chinese Hamster Ovary cells were transfected with CRTH2 receptors (CHO/CRTH2) and were maintained in culture in a humidified atmosphere at 37 0 C (5% CO 2 ) in Minimum Essential Medium (MEM) supplemented with 10% foetal bovine serum, 2 raM glutamine, and 1 mg ml '1 active G418. The cells were passaged every 2-3 days.
  • MEM Minimum Essential Medium
  • radioligand binding assay cells were prepared in triple-layer flasks or in 175 cm 2 square flasks (for membrane preparation).
  • calcium mobilisation assay cells were grown in a 96 well plate 24h prior to the assay at a density of 80,000 cells per well.
  • Membranes were prepared either from CHO/CRTH2 cells. CHO cells grown to confluency were washed with PBS and detached using a Versene solution (15 ml per flask). When the cells were grown in 175 cm 2 square flask, they were collected by scrapping in PBS. The cell suspensions were centrifuged (1,700 rpm, 10 min, 4 0 C) and resuspended in 15 ml of buffer (IxHBSS, supplemented with 10 mM HEPES, pH 7.3). Cell suspensions were then homogenised using an Ultra Turrax at setting 4- 6 for 20 s.
  • the homogenate was centrifuged at 1,700 rpm for 10 min and the supernatant was collected and centrifuged at 20,000 rpm for Ih at 4°C. The resulting pellet was resuspended in buffer and stored at -80 0 C in aliquots of 200-500 ⁇ l.
  • the protein concentration was determined by the method of Bradford (1976), using bovine serum albumin as standard.
  • the platelets were washed by centrifugation at 600xg for 10 min and resuspended in ice-cold assay buffer (10 mM Tris-HCl, pH 7.4, 5 mM Glucose, 120 mM NaCl, 10 ⁇ M indomethacin) and directly centrifuged at 20,000 rpm for 30 min at 4 0 C. The resulting pellet was treated as described above.
  • Radioligand binding assays [ 3 H]PGD 2 (160 Ci/mmol) binding experiments were performed on membranes prepared as described above. Assays were performed in a final volume of 100 ⁇ l of buffer (1XHBSS/HEPES 10 mM, pH 7.3). Cell membranes (15 ⁇ g). Cell membranes 15mg were preincubated at room temperature with varying concentration of competing ligand for 15 min. [ 3 H]PGD 2 (mol, final concentration) was then added and the incubation continued for a further one hour at room temperature.
  • the reaction was terminated by the addition of 200 ⁇ l ice-cold assay buffer to each well, followed by rapid filtration through Whatman GF/B glass fibre filters using a Unifilter Cell harvester (PerkinElmer Life Sciences) and six washes of 300 ⁇ l of ice- cold buffer.
  • the Unifilter plates were dried at room temperature for at least Ih and the radioactivity retained on the filters was determined on a Beta Trilux counter (PerkinElmer Life Sciences), following addition of 40 ⁇ l of Optiphase Hi-Safe 3 (Wallac) liquid scintillation. Non specific binding was defined in the presence of 10 ⁇ M unlabelled PGD 2 .
  • the CRTH2 antagonists useful in the present invention may be selective for the CRTH2 receptor and in this case will not have high affinity for the DP 1 or TP receptors. Binding to the DPi receptor may be measured in a radioligand binding assay similar to the one described above for measurement of CRTH2 antagonist activity except that in the cell culture phase, the Chinese Hamster Ovary cells were transfected with DP 1 receptors (CHO/DP), rather than with CRTH2 receptors, and the CHO/DP cells were used to prepare the cell membranes.
  • DP 1 receptors CHO/DP
  • Ki at the DP 1 receptor may be at least 5 ⁇ M or even at least 10 ⁇ M.
  • TP receptor radioligand binding may be carried out on membranes prepared from platelets. 15-40 ⁇ g of protein were pre-incubated with varying concentrations of competing ligand for 15 min at room temperature in assay buffer (10 mM Tris-HCl, pH 7.4, 5 mM glucose, 120 mM NaCl, 10 ⁇ M indomethacin). [ 3 H]SQ29548 (38 Ci/mmol, 10 nM final concentration) was then added and the incubation continued for a further 30 min at room temperature.
  • the reaction was terminated by the addition of 200 ⁇ l ice-cold assay buffer to each well, followed by rapid filtration through Whatman GF/C glass fibre filters using a Unif ⁇ lter Cell harvester (PerkinElmer Life Sciences) followed with six washes of 300 ⁇ l of ice-cold buffer.
  • the radioactivity was determined as described for the CRTH2 and DP 1 receptors.
  • Ki at the TP receptor may be at least 5 ⁇ M or even at least 10 ⁇ M.
  • the most suitable compounds for use in the present invention are those in which the binding selectivity is greater than 200 fold for CRTH2 receptor, compared to DP 1 TP receptors.
  • CRTH2 antagonist activity can be measured by the calcium mobilisation assay as follows.
  • Cells were seeded onto poly-D-lysine coated 96-well plates at a density of 80,000 cells per well and incubated at 37°C overnight to allow the cells to adhere. Cells were washed twice with HBSS and incubated for Ih at 37°C in lOO ⁇ l HBSS and lOO ⁇ l calcium-3-dye (Molecular Devices), supplemented with 4mM probenecid. Changes in fluorescence were monitored over a 50s time course with agonist addition at 17s using a Flexstation (Molecular Devices).
  • CRTH2 antagonist compounds inhibit this effect suitably with an IC 50 of less than l ⁇ M, more appropriately less than 500 nM still more suitably less than 100 nM and typically less than 50 nM or even less than 20 nM.
  • FIGURE 1 is a series of plots showing the combined symptom scores of two groups of patients with allergic rhinitis in a test comparing a CRTH2 antagonist with a placebo on Days 2 and 8 of each treatment period.
  • Figure IA shows the combined nasal symptom scores for both patient groups in response to challenge with a CRTH2 antagonist
  • Figure IB shows the combined scores for eye symptoms.
  • the plots show that patients taking the CRTH2 antagonist show reduced nasal and eye symptoms on both Day 2 and Day 8 of the treatment period.
  • FIGURE 2 is a series of plots similar to that of Figure 1 but where the scores for each patient group have been shown separately. The plots also show a screening response, which is the response of the patients to allergen before the beginning of the trial.
  • Figures 2A and 2B show the total nasal symptoms for patient group A compared with the screening response score for the same group of patients.
  • Figure 2A is plot showing the scores for nasal symptoms of patients in Group A on day 2 of each of the treatment periods while Figure 2B shows the nasal symptom scores for the same group of patients on day 8.
  • Patient group A was treated with CRTH2 antagonist for 8 days, then given no treatment for 3 weeks, then treated with placebo for 8 days.
  • Figures 2C and 2D are similar to Figures 2 A and 2B but show the scores for nasal symptoms from patients from patient group B on days 2 and 8 of the treatment periods together with the screening response score for the same group of patients.
  • Patient group B was treated with placebo for 8 days, then given no treatment for 3 weeks, then treated with CRTH2 antagonist for 8 days.
  • FIGURE 2 is a plot showing the anti-apoptotic effect of PGD2 in human Th2 cells.
  • Human Th2 cells were treated with 50U/ml IL-2 or various concentrations of PGD 2 in the absence of DL-2 for 16 hrs. The cells were stained with Annexin V-PE/PI and then analysed by FACSArray flow cytometer.
  • the study was a randomised, double blind, placebo controlled, two way crossover evaluation of Compound 1, given orally for eight days. There was a screening period of one week and a washout period of three weeks between the two treatment periods.
  • Subjects commenced dosing with Compound 1 or placebo on Day 1 (visit 2 or visit 5) of each treatment period of the study. Adverse events, total nasal symptom score and concomitant medications were noted in the CRF. Subjects took each dose (2 capsules in the morning and 2 capsules in the evening) as soon as they had finished breakfast and their evening meal respectively.
  • TNSS Total nasal symptom score
  • Eye symptom score watery eyes, itchy eyes, red eyes
  • each symptom scored on a categorical scale from 0 to 3 pre-challenge, every 15mins from 0 to 6h post-start of challenge
  • Subjects in Group A received a CRTH2 antagonist for 8 days, followed by a three week washout and then received placebo for 8 days. The subjects showed a reduced response to allergen after dosing with a CRTH2 antagonist. This reduced response to allergen was still present on dosing with placebo.
  • Subjects in Group B received placebo for 8 days, followed by a three week washout and then received a CRTH2 antagonist for 8 days.
  • the subjects mounted the expected response to allergen on placebo and showed a reduction in TNSS in response to allergen after dosing with the CRTH2 antagonist.
  • Human Th2 cells were treated with 50U/ml IL-2 or various concentrations of PGD 2 in the absence of IL-2 for 16 hrs. The cells were stained with Annexin V-PE/PI and then analysed by FACSArray flow cytometer. The results are illustrated in Figure 3, which shows that PGD 2 has an anti-apoptotic effect on human Th2 cells.
  • Human Th2 cells were treated in the absence of JL-2 with a medium containing 100 nM PGD 2 and various concentrations of CRTH2 antagonistic compounds (Compounds 1, 2, 3, 4 and 5) for 16 hrs. The cells were stained with Annexin V- PE/PI and then analysed by FACSArray flow cytometer. The IC 50 of Compounds 1 to 5 to the rescuing function of PGD 2 were calculated and found to be less than 10OnM in all cases.

Abstract

The invention relates to compounds of general formula (I) wherein R1, R2, R3, R4 and R5 are as defined herein for the treatment of allergic conditions, wherein the treatment is by pulsed therapy which comprises a first period during which the compound is administered to the patient and a second period of at least seven days during which the compound is administered to the patient in a reduced amount.

Description

USE OF CRTH2 ANTAGONIST COMPOUNDS
The present invention relates to the use of CRTH2 antagonist compounds for the pulsed treatment of allergic conditions.
Allergic conditions are becoming more common throughout the developed world with as much as 10% of the world's population being affected by one or more of allergic asthma, allergic rhinitis, atopic dermatitis and other allergic conditions. Numerous classes of compound have been suggested for the treatment of these conditions, with one of the more recent developments being the use of CRTH2 antagonists, which inhibit the action of agonists such as prostaglandin D2 (PGD2) acting at the CRTH2 receptor.
PGD2 is an eicosanoid, a class of chemical mediator synthesised by cells in response to local tissue damage, normal stimuli or hormonal stimuli or via cellular activation pathways. Eicosanoids bind to specific cell surface receptors on a wide variety of tissues throughout the body and mediate various effects in these tissues. PGD2 is known to be produced by mast cells, macrophages and Th2 lymphocytes and has been detected in high concentrations in the airways of asthmatic patients challenged with antigen (Murray et al, (1986), N. Engl. J. Med. 315: 800-804). Instillation of PGD2 into airways can provoke many features of the asthmatic response including bronchoconstriction (Hardy et al, (1984) N. Engl. J. Med. 311: 209-213; Sampson et al, (1997) Thorax 52: 513-518) and eosinophil accumulation (Emery et al, (1989) J. Appl. Physiol. 67: 959-962).
The potential of PGD2 to induce inflammatory responses has been confirmed by the use of transgenic mice overexpressing human PGD2 synthase which exhibit exaggerated eosinophilic lung inflammation and Th2 cytokine production in response to antigen (Fujitani et al, (2002) /. Immunol. 168: 443-449).
The first receptor specific for PGD2 to be discovered was the DPi receptor which is linked to elevation of the intracellular levels of cAMP. However, PGD2 is thought to mediate much of its proinflammatory activity through interaction with a G protein- coupled receptor termed CRTH2 (chemoattractant receptor-homologous molecule expressed on Th2 cells) which is expressed by Th2 lymphocytes, eosinophils and basophils (Hirai et al, (2001) J. Exp. Med. 193: 255-261, and EPO851O3O and EP-A- 1211513 and Bauer et al, EP-A-1170594). It seems clear that the effect of PGD2 on the activation of Th2 lymphocytes and eosinophils is mediated through CRTH2 since the selective CRTH2 agonists 13,14 dihydro-15-keto-PGD2 (DK-PGD2) and 15R- methyl-PGD2 can elicit this response and the effects of PGD2 are blocked by an anti- CRTH2 antibody (Hirai et al, 2001; Monneret et al, (2003) /. Pharmacol. Exp. Ther. 304: 349-355). In contrast, the selective DP agonist BW245C does not promote migration of Th2 lymphocytes or eosinophils (Hirai et al, 2001; Gervais et al, (2001) J. Allergy Clin. Immunol. 108: 982-988). Based on this evidence, antagonising PGD2 at the CRTH2 receptor is an attractive approach to treat the inflammatory component of Th2-dependent allergic diseases such as asthma (including allergic asthma), food allergies, acute and chronic urticaria, perennial allergic rhinitis, seasonal allergic rhinitis, atopic dermatitis, contact hypersensitivity (including contact dermatitis) and conjunctivitis, especially allergic conjunctivitis.
Documents which discuss the use of CRTH2 antagonists for the treatment of Th2- dependent allergic diseases include WO-A-03/066046, WO-A-03/066047, WO-A- 03/097042, WO-A-03/097598, WO-A-03/101981, WO-A-03/101961, WO-A- 2004/007451, WO-A-2005/019171, WO-A-2005/094816, WO-A-2005/044260, WO-A-2005/040112, WO-A-2005/040114 and WO2006/095183.
Other compounds which are combined CRTH2 and TP receptor antagonists are known and an example of such a compound is ramatroban, which has been shown to reduce allergic inflammation in the guinea pig nasal mucosa (Narita et al, Im Arch Allergy Immunol 1996;109: 161-166), mouse airways (Nagai et al, Prostaglandins 1995;50:75-87) and mouse skin (Takeshita et al, Int. Immunol. 2004,16:947-959). Ramatroban has also shown to reduce symptoms of perennial allergic rhinitis in human subjects (Terada et al, Allergol. Int. 1998; 47: 59-67). Analogues of ramatroban that retain CRTH2 antagonist activity, but which are inactive on TP, are effective in reducing airway eosinophilia and mucus cell hyperplasia in a model of allergic asthma in mice (Uller et al, Respir Res 2007;8: 16). Uller et al concluded that the efficacy in the allergic asthma model resulted from blockade of CRTH2 receptors rather than inhibition of TP.
Although there is ample evidence that CRTH2 antagonists are effective in treating allergic asthma, allergic rhinitis, allergic conjunctivitis, atopic dermatitis and other allergic conditions, the inventors have now made the surprising discovery that these compounds have an effect which is much longer lasting than previously thought and are thus useful preventing or reducing the symptoms of the allergic condition for a prolonged period of time after dosing.
Therefore, in a first aspect of the invention there is provided a compound of general formula (I):
Figure imgf000004_0001
(I) wherein
R1 is C1-C6 alkyl; R2 is halogen; and R3 is aryl or heteroaryl optionally substituted with one or more substituents selected from halo, OH, CN, R6, COR6, CH2R6, OR6, SR6, SO2R6 or SO2YR6;
R6 is C1-C6 alkyl, C3-Cg cycloalkyl, heterocyclyl, aryl or heteroaryl, any of which may optionally be substituted with one or more substituents selected from halo, OH, CN, NO2, C1-C6 alkyl or 0(C1-C6 alkyl); and Y is NH or a straight or branched C1-C4 alkylene chain;
R4 is H or C1-C4 alkyl; R5 is hydrogen, C1-C6 alkyl, aryl, (CH2)mOC(=O)C1-C6alkyl, ((CH2)mO)nCH2CH2X, (CH2)mN(R7)2 or CH((CH2)mO(C=O)R8)2; m is 1 or 2; n is 1-4; X is OR7 or N(R7)2;
R7 is hydrogen or methyl;
R8 is C1-C18 alkyl;
or a pharmaceutically acceptable salt, hydrate, solvate, or complex thereof; for the treatment of an allergic condition which is induced by the presence of an allergen to which the patient is sensitive, wherein the treatment comprises at least one treatment cycle which comprises: a. a first period during which the compound of general formula (I) is administered to the patient; and b. a second period of at least seven days during which the compound of general formula (I) is administered to the patient in a reduced amount; wherein the allergen is present or suspected to be present throughout the treatment and wherein the compound of general formula (I) retains at least 50% of its effectiveness during the second period of the treatment cycle.
This is particularly advantageous for conditions such as allergic rhinitis, allergic conjunctivitis or allergic asthma where during the period in which the patient suffers symptoms, medication conventionally has to be taken at least once a day, and often more frequently, in order to suppress those symptoms. In the present invention, treatment is taken as normal during the first period of the treatment cycle but during the second period a greatly reduced dosage of the compound of general formula (I) can be taken and in some cases, the dosage may be reduced to zero during the second period of the treatment cycle. This means that in the case of a condition such as allergic rhinitis where the hay fever season lasts for several weeks or even months, it may only be necessary for the patient to take medication for a small proportion of that season. The invention arises from observations made in the course of a double blind crossover study of a CRTH2 antagonist of general formula (I) in human subjects with allergic rhinitis. The study compared two groups of patients. The first patient group was treated daily with the CRTH2 antagonist for 8 consecutive days, then had no treatment for 3 weeks, then had daily treatment with placebo for 8 consecutive days. The second patient group was treated with placebo for 8 consecutive days, then had no treatment for 3 weeks, then had daily treatment with the CRTH2 antagonist of general formula (I) for 8 consecutive days. The study showed that in the first treatment period, patients treated with the CRTH2 antagonist of general formula (I) displayed fewer symptoms of allergic rhinitis than patients treated with a placebo and this result was expected by the inventors. Surprisingly, however, in the second treatment period, the first patient group, which was being treated with placebo, still retained a reduction in symptoms, the results being comparable with those of the second patient group which was being treated with the CRTH2 antagonist of general formula (I) at that time. These patients also displayed fewer symptoms than they had displayed in an initial screening before treatment with either CRTH2 antagonist or placebo.
It therefore appears that CRTH2 antagonists such as the compounds of general formula (I) as defined above have a prolonged effect on the allergic response and that, therefore, these compounds are of use in a pulsed method for the treatment of an allergic condition which is induced by the presence of an allergen to which the patient is sensitive, the method comprising administering to a patient over at least one treatment cycle a CRTH2 antagonist compound of general formula (I), wherein the said treatment cycle comprises: a. a first period during which a compound of general formula (I) is administered to the patient; and b. a second period of at least seven days during which the compound of general formula (I) is administered to the patient in a reduced amount; wherein the allergen is present or suspected to be present throughout the treatment and wherein the compound of general formula (I) retains at least 50% of its effectiveness during the second period of the treatment cycle. The invention also provides the use of a CRTH2 antagonist compound of general formula (I) in the preparation of an agent for the treatment of an allergic condition which is induced by the presence of an allergen to which the patient is sensitive, wherein the treatment comprises at least one treatment cycle which comprises: a. a first period during which the compound of general formula (I) is administered to the patient; and b. a second period of at least seven days during which the compound of general formula (I) is administered to the patient in a reduced amount; wherein the allergen is present or suspected to be present throughout the treatment and wherein the wherein the compound of general formula (I) retains at least 50% of its effectiveness during the second period of the treatment cycle.
The reason for the prolonged effect of CRTH2 antagonist compounds such as the compounds of general formula (I) in the treatment of allergic conditions is not completely clear but the inventors speculate that it may be related to a previously unknown effect of CRTH2 antagonists on Th2 cells.
The inventors have discovered that PGD2 plays an important role to play in Th2 cell survival by preventing the apoptosis of Th2 cells and their subsequent clearance from the allergic tissue. In addition, they have discovered that CRTH2 antagonists are able to block the effect of CRTH2 and induce apoptosis of Th2 cells.
The inventors believe that it is possible that the prolonged effect may be a result of the effect of CRTH2 antagonists on apoptosis of Th2 cells.
It is known that Th2 lymphocytes take part in the cascade leading to an allergic reaction as described by various authors, including Pettipher et al, Nature Reviews, 6 (April 2007), 313-325. The immune response in allergy begins with the presentation of allergens to antigen presenting cells which internalise the antigens and cause them to be expressed on the cell surface. This initiates a cascade of events leading to the production of IgE, which binds to high affinity receptors on mast cells leaving its allergen-specific site available for interaction with allergen. Upon re-exposure to the same allergen, cross-linking of mast cell-bound IgE initiates the production of a number of factors, including PGD2 by the mast cells, which in turn leads to the recruitment and activation of Th2 cells via the interaction of PGD2 with the CRTH2 receptor. The activated Th2 cells produce various cytokines including IL4, IL5 and IL13. IL4, in turn, causes the production of further IgE by B cells.
The inventors have postulated that the apoptotic effect of CRTH2 antagonists on Th2 lymphocytes interrupts the cascade by promoting apoptosis and clearance of Th2 cells which leads to a reduction in the levels of the Th2 cytokines IL4, IL5 or IL13. The reduction in IL4 and IL 13 production would lead to a decrease in the production of IgE by the B-lymphocytes. Therefore it appears that the immune system would not be able to respond to an allergen by increased production of IgE. This would lead to a significantly decreased immune response to an allergen until such time as the Th2 cell population is replenished.
Also, the reduction in IL5 would lead to reduced eosinophil migration and the reduction in IL13 to reduced mucus production. This combination of factors would also reduce the symptoms of allergy.
However, it should be noted that the long lasting effect of CRTH2 antagonists such as the compounds of general formula (I) in reducing the symptoms of an allergic condition has been shown to be present in vivo, even if this rationalisation proves to be incorrect.
In the present specification "C1-C6 alkyl" refers to a straight or branched saturated hydrocarbon chain having one to six carbon atoms and optionally substituted with one or more halo substituents or with one or more C3-O7 cycloalkyl groups. Examples include methyl, ethyl, n-propyl, isopropyl, t-butyl, n-hexyl, trifluoromethyl, 2-chloroethyl, methylenecyclopropyl, methylenecyclobutyl, methylenecyclobutyl and methylenecyclopentyl. "C1-C4 alkyl" and "C1-C18 alkyl" have similar meanings except that they contain from one to four and from one to eighteen carbon atoms respectively. C3-C7 cycloalkyl refers to a saturated 3 to 7 membered carbocyclic ring. Examples of such groups include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
The term "C1-C4 alkylene" in the context of the present specification refers to a disubstituted straight or branched saturated hydrocarbon chain having one to four carbon atoms.
In the present specification, "halo" refers to fluoro, chloro, bromo or iodo.
The term "aryl" in the context of the present specification refers to an aromatic ring system having from 5 to 14 ring carbon atoms and containing up to three rings. Examples of aryl groups are benzene and naphthalene.
The term "heteroaryl" in the context of the specification refers to a ring system with aromatic character having from 5 to 14 ring atoms, at least one of which is a heteroatom selected from N, O and S, and containing up to three rings. Where a heteroaryl group contains more than one ring, not all rings must be fully aromatic in character. Rings which are not fully aromatic may be substituted with one or more oxo groups. Examples of heteroaryl groups include pyrrole, thiophene, thiazole, pyridine, pyrimidine, indole, benzofuran, benzimidazole, tetrahydroquinoline, indoline, quinoline, isoquinoline, quinoxaline, imidazo[l,2-a]pyridine, pyrazolo[l,5- a]pyridine, 2,3-dihydro-l-benzothiopyrane and 2,3-dihydro-lλ6-benzothiopyran-l,l- dione.
The term "heterocyclyl" in the context of the specification refers to a saturated ring system having from 4 to 8 ring atoms, at least one of which is a heteroatom selected from N, O and S and which may be optionally substituted by one or more oxo groups. Examples of heterocyclyl groups include azetidinyl, piperidinyl; tetrahydrofuranyl, tetrahydropyranyl, dioxanyl, thiomorpholinyl, 1,1-dioxo-lλό- thiomorpholinyl, morpholinyl, pyrrolyl, piperizinyl, azepanyl, 1,4-diazepanyl, 1,4- oxazepanyl and azocanyl.
Appropriate pharmaceutically and veterinarily acceptable salts of the compounds of general formula (I) include basic addition salts such as sodium, potassium, calcium, aluminium, zinc, magnesium and other metal salts as well as choline, diethanolamine, ethanolamine, ethyl diamine, megulmine and other well known basic addition salts as summarised in J. Med. Chem., 50, 6665-6672 (2007) and/or known to those skilled in the art.
Where appropriate, pharmaceutically or veterinarily acceptable salts may also include salts of organic acids, especially carboxylic acids, including but not limited to acetate, trifluoroacetate, lactate, gluconate, citrate, tartrate, maleate, malate, pantothenate, adipate, alginate, aspartate, benzoate, butyrate, digluconate, cyclopentanate, glucoheptanate, glycerophosphate, oxalate, heptanoate, hexanoate, fumarate, nicotinate, pamoate, pectinate, 3-phenylpropionate, picrate, pivalate, proprionate, tartrate, lactobionate, pivolate, camphorate, undecanoate and succinate, organic sulfonic acids such as methanesulfonate, ethanesulfonate, 2-hydroxyethane sulfonate, camphorsulfonate, 2-naphthalenesulfonate, benzenesulfonate, p- chlorobenzenesulfonate and p-toluenesulfonate; and inorganic acids such as hydrochloride, hydrobromide, hydroiodide, sulfate, bisulfate, hemisulfate, thiocyanate, persulfate, phosphoric and sulfonic acids.
Salts which are not pharmaceutically or veterinarily acceptable may still be valuable as intermediates.
The compounds of general formula (I) in which R5 is hydrogen are active as CRTH2 antagonists.
Prodrugs are any covalently bonded compounds which release the active parent drug according to general formula (I) in vivo. Examples of prodrugs include the compounds of general formula (I) in which R5 is C1-C6 alkyl, aryl, (CH2)mOC(=O)CrC6alkyl, ((CH2)mO)nCH2CH2X, (CH2)mN(R7)2 or
CH((CH2)mO(C=O)R8)2; where m is 1 or 2; n is 1-4;
X is OR7 or N(R7)2;
R7 is hydrogen or methyl; and
R8 is C1-C18 alkyl.
If a chiral centre or another form of isomeric centre is present in a compound of the present invention, all forms of such isomer or isomers, including enantiomers and diastereoisomers, are intended to be covered herein. Compounds of the invention containing a chiral centre may be used as a racemic mixture, an enantiomerically enriched mixture, or the racemic mixture may be separated using well-known techniques and an individual enantiomer may be used alone.
The term "administered in combination with" refers to the co-administration of a CRTH2 antagonist with another drug wherein the administration may be simultaneous, sequential or separate.
In the context of the present invention, the term "allergic condition" refers to a condition in which the atopic state of a patient is increased above that of subject who does not have an allergic condition. This may be characterised by the presence in the circulation or tissues of a patient of increased numbers of cells reactive to allergens, and by the presence of IgE which binds specifically to an allergen to which the patient is sensitive. The IgE may be circulating or bound, for example to the surface of mast cells and basophils.
References herein to a patient being "sensitive" to an allergen mean that when the patient is exposed to the allergen, an allergic response is induced such that the patient will display symptoms of inflammation arising from the production of substances such as histamine, prostaglandin D2 and tryptase by mast cells and basophils when the allergen binds to IgE expressed on their surfaces. References to "the treatment of an allergic condition" refer to the reduction or removal of the inflammatory symptoms which arise from the exposure of a patient to an allergen to which the patient is sensitive.
References herein to an antigen being present or suspected to be present thoughout the treatment cycle are intended to mean that the antigen may be present in the patient's environment either constantly or intermittently and in varying amounts. For example, when patients suffer from allergic rhinitis, the pollen to which they are sensitive may be present in the environment throughout the hay fever season but the amount of pollen in the environment may vary depending on the weather, the time of day and other conditions. In some cases, a patient may suspect that the allergen is present in the environment, even if no tests are carried out to confirm this. Again, in the case of allergic rhinitis, a patient would conventionally take medication as a precautionary measure if it was suspected that pollen may be present, as might occur, for example, during the hay fever season. However, with the present invention, it would not be necessary for a patient to take a full dose of the compound of general formula (I) during the second period of the treatment cycle and often no compound of general formula (I) needs to be administered during this second period.
The term "effectiveness" as applied to the compounds of general formula (I) refers to the reduction of the allergic response in the patient. In general, during the first period of the treatment cycle, the patient will experience a reduction of at least 30% in symptoms, suitably at least 40% and with increasing order of suitability, at least 50%, at least 60%, at least 70% at least 80% and, in the most favourable cases, at least a 90% reduction in symptoms.
In the case of allergic rhinitis and conjunctivitis, assessment may be carried out, for example in a Vienna Challenge Chamber experiment, which is well known to those of skill in this art and in which patients carry out self-assessment and scoring of their symptoms on a scale of 0 to 3. Separate scores may be given for eye symptoms, nasal symptoms (including nasal obstruction, nasal itch, sneeze and rhinorrhea) and other symptoms. Although the symptom score for each patient is subjective, if a sufficient number of patients is used, the total scores are meaningful. For other allergic conditions, other clinical trials may be designed and the measurement of reduction in symptoms may either be carried out by a subjective method such as self assessment by patients or, alternatively, may be made by an objective measurement. For example, asthma symptoms may be quantified using measurements of lung function such as forced expiratory volume in one second (FEVi) or peak expiratory flow rate (PEF) or using the Juniper quality of life scale. The severity of atopic dermatitis symptoms can be assessed using the scoring atopic dermatitis (SCORAD) or six area six sign atopic dermatitis (SASSAD) systems.
As set out above, during the second period of the treatment cycle, the compound of general formula (I) retains at least 50% of its effectiveness, but it is more suitable that the compound of general formula (I) retains at least 60% of its effectiveness and, in increasing order of suitability, 70%, 80% and 90% of its effectiveness.
As already noted, the amount of compound of general formula (I) administered to the patient during the second period of the treatment cycle is reduced compared with the amount administered during the first period. In general, the daily dose of compound of general formula (I) administered during the second period of the treatment cycle will be not greater than 50% of the daily dose administered during the first period. However, it may be less than this, for example not greater than 40%, not greater than 30%, not greater than 20% or not greater than 10% of the daily dose of compound of general formula (I) administered during the first period. In many cases administration of the CRTH2 antagonist compound will be discontinued altogether during the second period of the treatment cycle. Thus, in the second period of the treatment cycle, the daily dose with be zero and no compound of general formula (I) will be administered to the patient. This is a particularly advantageous result as period during which the medication is necessary will be considerably shorter than for conventional medications, which has positive implications both for the general well being of the patient and the cost of the treatment.
The second period of the treatment cycle lasts for at least 7 days. However, it may last for longer periods, for example at least 14 days and it has been observed that the drug may retain at least 50% of its effectiveness during second periods lasting at least 21 days, at least 28 days and, in some cases, in excess of 35 days even when the dose of compound is reduced to zero during the second period of the treatment cycle.
The first period of the treatment cycle during which the CRTH2 antagonist is administered may be chosen such that it comprises a period of days that provides the maximum biological response to the compound in the patient. In general, this period may be from two to 8 days, during which time the drug may be administered from one to three times a day. It is particularly suitable however that the CRTH2 antagonist is administered to the patient once daily as this helps to increase patient compliance.
In some cases, one treatment cycle may not be sufficient to last for the whole time over which treatment is required and therefore the treatment may include one or more further treatment cycles. This is often the case with conditions such as seasonal allergic rhinitis, where the hay fever season may last for several months.
CRTH2 antagonists such as the compounds of general formula (I) are useful in treating any allergic condition but they are particularly useful in the pulsed treatment of conditions such as asthma (including allergic asthma), food allergies, acute and chronic urticaria, perennial allergic rhinitis, seasonal allergic rhinitis, atopic dermatitis, contact hypersensitivity (including contact dermatitis) and conjunctivitis, especially allergic conjunctivitis.
The CRTH2 antagonists of general formula (I) are indole- 1 -acetic acid derivatives and are some of the compounds and analogues thereof described in WO2005/044260, WO2005/094816 and WO2006/095183, WO2008/012511 and UK patent application Nos 0800874.0, filed 18 January 2008; 0801132.2, filed 22 January 2008; 0801671.9, filed 30 January 2008; 0801131.4, filed 22 January 2008; 0801672.7, filed 30 January 2008; and 0801674.3, filed 30 January 2008. The indole- 1-acetic acid derivatives of general formula (I) as defined above are described in detail in these documents.
In one embodiment of the invention, the compound of general formula (I) is a CRTH2 antagonist in which R5 is hydrogen.
In an alternative embodiment of the invention, the compound of general formula (I) is a prodrug for a CRTH2 antagonist and R5 is C1-C6 alkyl, aryl, (CH2)mOC(=O)Ci- Qjalkyl, ((CH2)mO)nCH2CH2X, (CH2)mN(R7)2 or CH((CH2)mO(C=O)R8)2; where m is 1 or 2; n is 1-4;
X is OR7 or N(R7)2;
R7 is hydrogen or methyl; and
R8 is Ci-C18 alkyl.
In suitable compounds of general formula (I), independently or in any combination: R1 is fluoro;
R2 is C1-C4 alkyl, particularly methyl or ethyl but more especially methyl; R4 is H or methyl; and R3 is quinoline, quinoxaline, isoquinoline, thiazole, phenyl, naphthalene, thiophene, pyrrole or pyridine, any of which may optionally be substituted as set out above.
In particularly suitable compounds, R4 is H.
More typical R3 groups include optionally substituted quinoline, phenyl, naphthalene, thiophene, pyrrole or pyridine.
When R3 is quinoline or isoquinoline, it is suitably unsubstituted or substituted with one or more halo substituents, especially fluoro.
When R3 is phenyl, naphthalene, thiophene, pyrrole or pyridine, it may optionally have one or more substituents, with particularly suitable substituents including OR6, SO2R0 or SO2YR0; where R >60 and Y are as defined above.
Typically, in this case, R6 is C1-C6 alkyl, a 4- to 6-membered cycloalkyl group, a 5- or 6-membered heterocyclyl group or phenyl, any of which may be substituted as defined above.
When R3 is pyridyl it is most suitably a 3-pyridyl moiety.
In more active compounds, Y, when present, is a CH2 moiety.
When R3 is substituted with SO2R6 or SO2YR6, the R6 group is generally unsubstituted or substituted with one or more substituents chosen from methyl and halo, particularly chloro or fluoro.
When
Figure imgf000016_0001
group may be unsubstituted or substituted with one or more substituents chosen from halo, cyano, C1-C4 alkyl and 0(Ci-C4 alkyl).
Particularly suitable compounds of general formula (I) include: {3-[l-(4-Chloro-phenyl)-ethyl]-5-fluoro-2-methyl-indol-l-yl}-acetic acid;
{5-Fluoro-2-methyl-3-[l-(4-trifluoromethyl-phenyl)-ethyl]-indol-l-yl} -acetic acid;
{3-[l-(4-tert-Butyl-phenyl)-ethyl]-5-fluoro-2-methyl-indol-l-yl}-acetic acid;
{5-Fluoro-3-[l-(4-methanesulfonyl-phenyl)-ethyl]-2-methyl-indol-l-yl}-acetic acid;
[5-Fluoro-2-methyl-3-(l-naphthalen-2-yl-ethyl)-indol-l-yl]-acetic acid; (5-Fluoro-2-methyl-3-quinolin-2-ylmethyl-indol-l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-naphthalen-2-ylmethyl-indol-l-yl)-acetic acid;
[5-Fluoro-3-(8-hydroxyquinolin-2-ylmethyl)-2-methyl-indol-l-yl]-acetic acid;
[5-Fluoro-2-methyl-3-(quinoxalin-2-ylmethyl)indol- 1 -yl]-acetic acid;
[5-Fluoro-3-(4-methoxy-benzyl)-2-methyl-indol-l-yl]-acetic acid; [5-Fluoro-2-methyl-3-( 1 ,3-thiazol-2-ylmethyl)indol- l-yl]-acetic acid;
[3-(4-Chloro-benzyl)-5-fluoro-2-methyl-indol- l-yl]-acetic acid;
[5-Fluoro-2-methyl-3-(4-trifluorornethyl-benzyl)-indol- 1 -yl]-acetic acid; [5-Fluoro-2-methyl-3-(4-tert-butyl-benzyl)-indol-l-yl]-acetic acid;
{5-Fluoro-2-methyl-3-[(4-phenylphenyl)methyl]indol-l-yl}-acetic acid;
[5-Fluoro-3-(4-methanesulfonyl-benzyl)-2-methyl-indol-l-yl]-acetic acid;
{5-Fluoro-3-[(6-fluoroquinolin-2-yl)methyl]-2-methylindol-l-yl}-acetic acid; (2-Methyl-3-quinolin-2-ylmethyl-indol-l-yl)-acetic acid;
(5-Chloro-2-methyl-3-quinolin-2-ylmethyl-indol-l-yl)-acetic acid;
(3-{[l-(Benzenesulfonyl)pyrrol-2-yl]methyl}-5-fluoro-2-methylindol-l-yl)-acetic acid;
[5-Fluoro-2-methyl-3-({ l-[(4-methylbenzene)sulfonyl]pyrrol-2-yl}methyl)indol-l- yl]-acetic acid;
[3-({ l-[(2,4-Difluorobenzene)sulfonyl]pyrrol-2-yl}methyl)-5-fluoro-2-methylindol-
1-yl] -acetic acid;
(3- { [2-(Benzenesulfonyl)phenyl]methyl } -5-fluoro-2-methylindol- 1 -yl)-acetic acid;
[3-({2-[(4-Chlorobenzene)sulfonyl]phenyl}methyl)-5-fluoro-2-methylindol-l-yl]- acetic acid;
[5-Fluoro-3-({2-[(4-fluorobenzene)sulfonyl]phenyl}methyl)-2-methylindol-l-yl]- acetic acid;
(3-{[2-(Benzenesulfonyl)pyridin-3-yl]methyl}-5-fluoro-2-methylindol-l-yl)-acetic acid; [5-Fluoro-3-({2-[(4-fluorobenzene)sulfonyl]pyridin-3-yl}methyl)-2-methylindol-l- yl] -acetic acid;
[3-({2-[(4-Chlorobenzene)sulfonyl]pyridin-3-yl}methyl)-5-fluoro-2-methylindol-l- yl] -acetic acid;
2-(3-(4-(Benzylsulfonyl)benzyl)-5-fluoro-2-methyl-indol-l-yl)-acetic acid; 2-(3-(4-(4-Chlorobenzylsulfonyl)benzyl)-5-fluoro-2-methyl-indol-l-yl)-acetic acid;
2-(3-(3-(Benzylsulfonyl)benzyl)-5-fluoro-2-methyl-indol-l-yl)-acetic acid;
2-(5-Fluoro-3-(3-(4-fluorobenzylsulfonyl)benzyl)-2-methyl-indol- 1 -yl)-acetic acid;
2-(3-(2-(Benzylsulfonyl)benzyl)-5-fluoro-2-methyl-indol-l-yl)-acetic acid;
2-(3-(4-(4-Fluorobenzylsulfonyl)benzyl)-5-fluoro-2-methyl-indol- 1 -yl)-acetic acid; 2-(3-(2-(Cyclohexylsulfonyl)benzyl)-5-fluoro-2-methyl-indol-l-yl)-acetic acid;
2-(5-Fluoro-2-methyl-3-(2-(piperidin-l-ylsulfonyl)benzyl)-indol-l-yl)-acetic acid;
2-(3-(2-(Cyclopentylsulfonyl)benzyl)-5-fluoro-2-methyl-indol-l-yl)-acetic acid; 2-(5-Fluoro-2-methyl-3-(3-(piperidin-l-ylsulfonyl)benzyl)-indol-l-yl)-acetic acid;
2-(5-Fluoro-2-methyl-3-(2-(pyrrolidin-l-ylsulfonyl)benzyl)-indol-l-yl)-acetic acid;
2-(3-(4-(Cyclohexylsulfonyl)benzyl)-5-fluoro-2-methyl-indol-l-yl)-acetic acid;
2-(3-(4-(Cyclopentylsulfonyl)benzyl)-5-fluoro-2-methyl-indol-l-yl)-acetic acid; 2-(3-(2-(Cyclobutylsulfonyl)benzyl)-5-fluoro-2-methyl-indol-l-yl)-acetic acid;
2-(5-Fluoro-2-methyl-3-(3-(pyrrolidin-l-ylsulfonyl)benzyl)-indol-l-yl)-acetic acid acid;
2-(5-Fluoro-2-methyl-3-(4-(piperidin-l-ylsulfonyl)benzyl)-indol-l-yl)-acetic acid;
[5-Fluoro-2-methyl-3-(2-phenoxybenzyl)-indol-l-yl]-acetic acid; [5-Fluoro-2-methyl-3-(2-(4-methoxyphenoxy)benzyl)-indol-l-yl]-acetic acid;
[5-Fluoro-2-methyl-3-(2-(4-methylphenoxy)benzyl)-indol-l-yl]-acetic acid;
[5-Fluoro-2-methyl-3-(2-(2,4-dichlorophenoxy)benzyl)-indol- 1 -yl] -acetic acid;
[5-Fluoro-2-methyl-3-(2-(4-fluorophenoxy)benzyl)-indol-l-yl]-acetic acid;
[5-Fluoro-2-methyl-3-(2-(3,4-difluorophenoxy)benzyl)-indol-l-yl]-acetic acid; [5-Fluoro-2-methyl-3-(2-(4-cyanophenoxy)benzyl)-indol-l-yl]-acetic acid;
[5-Fluoro-2-methyl-3-(2-(4-chlorophenoxy)benzyl)-indol-l-yl]-acetic acid;
[5-Fluoro-2-methyl-3-(2-(2-cyanophenoxy)benzyl)-indol-l-yl]-acetic acid;
(5-Fluoro-2-methyl-3-{[2-(4-methylphenoxy)pyridin-3-yl]methyl}indol-l-yl)-acetic acid; {5-Fluoro-3-[(3-methanesulfonylnaphthalen-2-yl)methyl]-2-methylindol-l-yl}-acetic acid;
{5-Fluoro-3-[(l-methanesulfonylnaphthalen-2-yl)methyl]-2-methylindol-l-yl}-acetic acid;
{S-Fluoro-S-tCo-methanesulfonylnaphthalen^-y^methy^^-methylindol-l-ylj-acetic acid;
[5-Fluoro-2-methyl-3-(quinolin-3-ylmethyl)indol-l-yl]-acetic acid;
[5-Fluoro-2-methyl-3-(quinoxalin-6-ylmethyl)indol-l-yl]-acetic acid;
[S-Fluoro-l-methyl-S-Cquinolin-T-ylmethy^indol-l-yll-acetic acid;
{5-Fluoro-3-[(6-methanesulfonylquinolin-2-yl)methyl]-2-methylindol-l-yl}-acetic acid;
{ 5-Fluoro-3-[(4-methanesulfonylquinolin-2-yl)methyl] -2-methylindol- 1 -yl } -acetic acid; (5-Fluoro-2-methyl-3-{pyrazolo[l,5-a]pyridin-3-ylmethyl}indol-l-yl)-acetic acid;
(5-Fluoro-3- { imidazo[ 1 ,2-a]pyridin-2-ylmethyl } -2-methylindol- 1 -yl)-acetic acid;
(5-Fluoro-2-methyl-3-{ [2-(methylsulfanyl)phenyl]methyl }indol-l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-{ [3-(methylsulfanyl)phenyl]methyl }indol-l-yl)-acetic acid; (5-Fluoro-2-methyl-3-{ [4-(ethylsulfanyl)phenyl] methyl }indol-l-yl)-acetic acid(3-
{ [4-(Ethylsulfanyl)phenyl]methyl }-5-fluoro-2-methylindol- l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-{[4-(n-propylsulfanyl)phenyl]methyl}indol-l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-{[4-(i-propylsulfanyl)phenyl]methyl}indol-l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-{ [4-(t-butylsulfanyl)phenyl]methyl }indol-l-yl)-acetic acid; (5-Fluoro-2-methyl-3-{[4-(pentan-3-ylsulfanyl)phenyl]methyl}indol-l-yl)-acetic acid;
[3-({4-[(Cyclopropylmethyl)sulfanyl]phenyl}methyl)-5-fluoro-2-methylindol-l-yl]- acetic acid;
{3-[(4,4-Dimethyl-2,3-dihydro-l-benzothiopyran-6-yl)methyl]-5-fluoro-2- methylindol- 1 -yl } -acetic acid;
(3-{[2-(Ethanesulfonyl)phenyl]methyl}-5-fluoro-2-methylindol-l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-{[2-(propane-l-sulfonyl)phenyl]methyl}indol-l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-{[2-(propane-2-sulfonyl)phenyl]methyl}indol-l-yl)-acetic acid;
(3-{ [2-(Butane-l-sulfonyl)phenyl]methyl }-5-fluoro-2-methylindol-l-yl)-acetic acid;
(3-{[2-(Butane-2-sulfonyl)phenyl]methyl}-5-fluoro-2-methylindol-l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-{[2-(2-methylpropane-2-sulfonyl)phenyl]methyl}indol-l-yl)- acetic acid; (5-Fluoro-2-methyl-3-{[2-(pentane-l-sulfonyl)phenyl]methyl}indol-l-yl)-acetic acid;
(3-{[2-(Cyclopropylmethane)sulfonylphenyl]methyl}-5-fluoro-2-methylindol-l-yl)- acetic acid;
(5-Fluoro-2-methyl-3-{[2-(propylsulfamoyl)phenyl]methyl}indol-l-yl)-acetic acid; (3-{ [2-(Butylsulfamoyl)phenyl]methyl}-5-fluoro-2-methylindol-l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-{[3-(propylsulfamoyl)phenyl]methyl}indol-l-yl)-acetic acid;
(3- { [3-(Butylsulf amoyl)phenyl]methyl } -5-fluoro-2-methylindol- 1 -yl)-acetic acid; (5-Fluoro-2-methyl-3-{ [4-(trifluoromethane)sulfonylphenyl]methyl}indol-l-yl)- acetic acid;
(3-{ [4-(Ethanesulfonyl)phenyl]methyl }-5-fluoro-2-methylindol-l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-{ [4-(propane-l-sulfonyl)phenyl]methyl}indol-l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-{[4-(propane-2-sulfonyl)phenyl]methyl}indol-l-yl)-acetic acid;
(3-{[4-(Butane-l-sulfonyl)phenyl]methyl}-5-fluoro-2-methylindol-l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-{ [4-(2-methylpropane-2-sulfonyl)phenyl]methyl}indol-l-yl)- acetic acid;
(5-Fluoro-2-methyl-3-{[4-(pentane-l-sulfonyl)phenyl]methyl}indol-l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-{[4-(pentan-3-ylsulfonyl)phenyl]methyl}indol-l-yl)-acetic acid; [3-( { 4-[(Cyclopropylmethyl)sulfonyl]phenyl } methyl)-5-fluoro-2-methylindol- 1 -yl] - acetic acid;
(5-Fluoro-2-methyl-3-{ [4-(propylsulfamoyl)phenyl]methyl}indol-l-yl)-acetic acid;
(3-{ [4-(Butylsulfamoyl)phenyl]methyl }-5-fluoro-2-methylindol-l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-{ [4-(trifluoromethoxy)phenyl]methyl }indol-l-yl)-acetic acid; (5-Fluoro-3- { [4-methanesulfonyl-3-(trifluoromethyl)phenyl]methyl } -2-methylindol- l-yl)-acetic acid;
(5-Fluoro-3- { [4-methanesulfonyl-3-(trifluoromethoxy)phenyl]methyl } -2- methylindol-l-yl)-acetic acid;
{5-Fluoro-3-[(5-methanesulfonylthiophen-2-yl)methyl]-2-methylindol-l-yl}-acetic acid;
{3-[(4)4-dimethyl-l,l-dioxo-2,3-dihydro-lλ6-benzothiopyran-6-yl)methyl]-5-fluoro-
2-methylindol-l-yl} -acetic acid;
[3-({ l-[(4-Chlorobenzene)sulfonyl]pyrrol-2-yl}methyl)-5-fluoro-2-methylindol-lyl]- acetic acid; [5-Fluoro-3-({ l-[(4-fluorobenzene)sulfonyl]pyrrol-2-yl}methyl)-2-methylindol-l- yl]-acetic acid;
[5-Fluoro-3-({ l-[(4-methoxybenzene)sulfonyl]pyrrol-2-yl}methyl)-2-methylindol-l- yl]-acetic acid;
{3-[l-(2,4-Dichloro-benzenesulfonyl)pyrrol-2-ylmethyl]-5-fluoro-2-methyl-indol-l- yl} -acetic acid;
[5-Fluoro-3-({ l-[(4-methanesulfonylbenzene)sulfonyl]pyrrol-2-yl}methyl)-2- methylindol-1-yl] -acetic acid;
{5-Fluoro-2-methyl-3-[(2-phenylphenyl)methyl]indol-l-yl}-acetic acid;
(3-{ [l-(Benzenesulfonyl)indol-2-yl]methyl}-5-fluoro-2-methylindol-l-yl)-acetic acid;
(3-{ [2-(4-Chlorophenyl)phenyl]methyl }-5-fluoro-2-methylindol-l-yl)-acetic acid; (5-Fluoro-2-methyl-3-{ [2-(4-methylphenyl)phenyl]methyl}indol-l-yl)-acetic acid;
{5-Fluoro-2-methyl-3-[(3-phenoxyphenyl)methyl]indol-l-yl} -acetic acid;
[5-Fluoro-3-({4-[(4-fluorophenyl)carbonyl]-l-methylpyrrol-2-yl}methyl)-2- methylindol-l-yl]-acetic acid;
{ 5-Fluoro-2-methyl-3-[(6- { [3-(trifluoromethyl)phenyl]methyl }pyridin-3- yl)methyl]indol-l-yl}-acetic acid;
{5-Fluoro-2-methyl-3-[(3-phenoxythiophen-2-yl)methyl]indol-l-yl} -acetic acid;
(3-{[2-(Benzenesulfonyl)-l,3-thiazol-5-yl]methyl}-5-fluoro-2-methylindol-l-yl)- acetic acid;
{3-[(l-Benzylpyrazol-4-yl)methyl]-5-fluoro-2-methylindol-l-yl}-acetic acid; (3- { [5-(4-Chlorophenoxy)- 1 -methyl-3-(trifluoromethyl)pyrazol-4-yl]methyl } -5- fluoro-2-methylindol-l-yl)-acetic acid;
[3-({5-[(4-Chlorobenzene)sulfonyl]furan-2-yl}methyl)-5-fluoro-2-methylindol-l-yl]- acetic acid;
[3-({5-[(4-Chlorobenzene)sulfonyl]thiophen-2-yl}methyl)-5-fluoro-2-methylindol-l- yl]-acetic acid;
[3-({3-[(4-Chlorobenzene)sulfonyl]thiophen-2-yl}methyl)-5-fluoro-2-methylindol-l- yl]-acetic acid;
{3-[(2-Benzylphenyl)methyl]-5-fluoro-2-methylindol-l-yl}-acetic acid;
or the C-C6 alkyl, aryl, (CH2)mOC(=O)Ci-C6alkyl, ((CH2)mO)nCH2CH2X, (CH2)mN(R7)2 or CH((CH2)mO(C=O)R8)2 esters of any of the above; wherein m is 1 or 2; n is 1-4;
X is OR7 or N(R7)2;
R7 is hydrogen or methyl;
R8 is C1-C18 alkyl.
The compounds of general formula (I) may be prepared according to methods set out in the prior art or methods analogous to those set out in the prior art, in particular WO2005/044260, WO2005/094816 and WO2006/095183, WO2008/012511 and UK patent application Nos 0800874.0, 0801132.2, 0801671.9, 0801131.4, 0801672.7 and 0801674.3.
The compound of general formula (I) may be combined with one or more additional agent which is of use in the treatment of these allergic conditions. Examples of such agents include existing therapies for allergic and other inflammatory diseases including:
Suplatast tosylate and similar compounds;
B2 adrenoreceptor agonists such as metaproterenol, isoproterenol, isoprenaline, albuterol, salbutamol, formoterol, salmeterol, terbutaline, orciprenaline, bitolterol mesylate ,pirbuterol and indacaterol or methylxanthanines such as theophylline and aminophylline, mast cell stabilisers such as sodium cromoglycate or muscarinic receptor antagonists such as ipratropium and tiotropium; antihistamines, for example histamine H1 receptor antagonists such as loratadine cetirizine, desloratadine, levocetirizine, fexofenadine, astemizole, azelastine and chlorpheniramine or H4 receptor antagonists; (X1 and α2 adrenoreceptor agonists such as propylhexedrine phenylephrine, phenylpropanolamine, pseudoephedrine, naphazoline hydrochloride, oxymetazoline hydrochloride, tetrahydrozoline hydrochloride, xylometazoline hydrochloride and ethylnorepinephrine hydrochloride; corticosteroids such as prednisone, prednisolone, flunisolide, triamcinolone acetonide, beclomethasone dipropionate, budesonide, fluticasone propionate, fluticasone furoate, mometasone furoate and ciclesonide; allergen immunotherapy such as Grazax. The CRTH2 antagonist of general formula (I) will generally be administered in a pharmaceutical formulation which may be a formulation suitable for oral, rectal, nasal, bronchial (inhaled), topical (including eye drops, buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration and may be prepared by any methods well known in the art of pharmacy.
The route of administration will depend upon the condition to be treated but preferred compositions are formulated for oral, nasal, bronchial or topical administration.
The formulation may be prepared by bringing into association the above defined active agent with a carrier. In general, the formulations are prepared by uniformly and intimately bringing into association the active agent with liquid carriers or finely divided solid carriers or both, and then if necessary shaping the product.
Formulations for oral administration in the present invention may be presented as: discrete units such as capsules, sachets, tablets, which may be chewable tablets, or lozenges, each containing a predetermined amount of the active agent; as a powder or granules; as fine particles for sprinkling over food; as a solution or a suspension of the active agent in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water in oil liquid emulsion; or as a bolus etc.
For compositions for oral administration (e.g. tablets and capsules), the term "acceptable carrier" includes vehicles such as common excipients e.g. binding agents, for example syrup, acacia, gelatin, sorbitol, tragacanth, polyvinylpyrrolidone (Povidone), methylcellulose, ethylcellulose, sodium carboxymethylcellulose, hydroxypropylmethylcellulose, sucrose and starch; fillers and carriers, for example corn starch, gelatin, lactose, sucrose, microcrystalline cellulose, kaolin, mannitol, dicalcium phosphate, sodium chloride and alginic acid; and lubricants such as magnesium stearate, sodium stearate and other metallic stearates, glycerol stearate stearic acid, silicone fluid, talc waxes, oils and colloidal silica. Flavouring agents such as peppermint, oil of wintergreen, cherry flavouring and the like can also be used. It may be desirable to add a colouring agent to make the dosage form readily identifiable. Tablets may also be coated by methods well known in the art.
A tablet may be made by compression or moulding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active agent in a free flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface-active or dispersing agent. Moulded tablets may be made by moulding 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 slow or controlled release of the active agent.
Other formulations suitable for oral administration include lozenges comprising the active agent in a flavoured base, usually sucrose and acacia or tragacanth; pastilles comprising the active agent in an inert base such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active agent in a suitable liquid carrier.
For topical application to the skin, CRTH2 antagonists such as compounds of general formula (I) may be made up into a cream, ointment, jelly, solution or suspension etc. Cream or ointment formulations that may be used for the drug are conventional formulations well known in the art, for example, as described in standard text books of pharmaceutics such as the British Pharmacopoeia.
For topical application to the eye, CRTH2 antagonists such as compounds of general formula (I) may be made up into an eye drop formulation. Suitable eye drop formulations are also well known in the art and are described in standard text books of pharmaceutics such as the British Pharmacopoeia.
CRTH2 antagonists which are intended to reduce or remove the symptoms of conditions such as allergic asthma, allergic conjunctivitis and allergic rhinitis may be administered to the respiratory tract by nasal, bronchial or buccal administration of, for example, aerosols or sprays which can disperse the pharmacological active ingredient in the form of a powder or in the form of drops of a solution or suspension. Pharmaceutical compositions with powder-dispersing properties usually contain, in addition to the active ingredient, a liquid propellant with a boiling point below room temperature and, if desired, adjuncts, such as liquid or solid non-ionic or anionic surfactants and/or diluents. Pharmaceutical compositions in which the pharmacological active ingredient is in solution contain, in addition to this, a suitable propellant, and furthermore, if necessary, an additional solvent and/or a stabiliser. Instead of the propellant, compressed air can also be used, it being possible for this to be produced as required by means of a suitable compression and expansion device.
Parenteral formulations will generally be sterile.
Typically, the oral dose of the compound will be about 0.01 to 100 mg/kg; so as to achieve a concentration of drug in the plasma at a concentration effective to inhibit PGD2 at the CRTH2 receptor. The precise amount of a compound of CRTH2 antagonist which is therapeutically effective, and the route by which such compound is best administered, is readily determined by one of ordinary skill in the art by comparing the blood level of the agent to the concentration required to have a therapeutic effect.
In formulations for topical administration, such as eye drops, creams or ointments, the concentration of active compound will typically be about 0.1%-1% w/w such that systemic exposure of the CRTH2 antagonist compound is limited.
Since the mechanism of action appears to be that described above, the prolonged effect on the inflammatory symptoms of allergic conditions will observed with any CRTH2 antagonist. Documents which discuss the use of CRTH2 antagonists for the treatment of Th2-dependent allergic diseases include WO-A-03/066046, WO-A- 03/066047, WO-A-03/097042, WO-A-03/097598, WO- A-03/101981, WO-A- 03/101961, WO-A-2004/007451, WO-A-2005/019171, WO-A-2005/054232, WO- A-2004/089884, WO-A-2004/089885, WO-A-2005/018529, WO-A-2006/005909, WO2006/021759, WO-A-2007/039736, WO-A-2007/052023, WO-A-2006/075139, WO-A-2007/068894, WO- A-2007138282, WO-A-2008/119917, WO-A- 2008/113965, WO-A-2008/074966, WO-A-2008/078069, WO-A-2007/144625, WO-A-2007/028999, WO- A-2007/031747, WO-A-2006/136859, WO-A- 2006/111560, WO-A-2005/094816, WO-A-2005/040112, WO-A-2005/040114, WO-A-2004/096777, WO-A-2005/ 123731, WO-A-2006/125784, WO-A- 2007/045867, WO-A-2006/034419, WO-A-2006/036994, WO-A-2007/022501, WO-A-2004/106302, WO-A-2004/032848, WO-A-2005/100321, WO-A- 2006/091674, WO-A-2004/058164, WO-A-2005/007094, WO-A-2007/036743, WO-2004/035543, WO-A-2007/062797, WO-A-2007/062773, WO-A-2007/062678, WO-A-2007/062677, WO-A-2005/ 116001, WO-A-2005/115382, WO-A- 2005/115374, WO-A-2006/111560, WO-A-2006/037982, WO-A-2006/056752, WO-A-2007/039741, WO-A-2005/073234, WO-A-2005/105727, WO-A- 2006/063763, WO-A-2006/125593 and WO-A-2006/125596, and the compounds described in these documents are useful for the treatment of an allergic condition which is induced by the presence of an allergen to which the patient is sensitive, wherein the treatment comprises at least one treatment cycle which comprises: a. a first period during which the CRTH2 antagonist compound is administered to the patient; and b. a second period of at least seven days during which the compound of general formula (I) is administered to the patient in a reduced amount; wherein the allergen is present or suspected to be present throughout the treatment and wherein the CRTH2 antagonist retains at least 50% of its effectiveness during the second period of the treatment cycle.
Some particularly suitable compounds are compounds which are similar to the compounds of general formula (I) but in which the moiety -CHR3R4 of general formula (I) is replaced by R3, SR3, SOR3 or SO2R3, where R3 is as defined for general formula (I). An example compound of this type, [5-Fluoro-3-(4-chloro- benzenesulfonyl)-2-methyl-indol-l-yl]-acetic acid, which falls within the scope of WO 2004/007451 was tested in the apoptosis assay of Example 3 (below) and was found counter the rescuing function of PGD2 with an IC50 of 204±160 nM.
In the context of the present invention, a CRTH2 antagonist is a compound which binds to the CRTH2 receptor with a Ki of less than 1000 nM in the radioligand binding assay described below and which inhibits the dose-dependent increase in intracellular Ca2+ mobilisation in CHO/CRTH2 cells treated with PGD2 with an IC50 of less than 1000 nM. Although any compound with CRTH2 antagonist activity would be suitable for use in desensitising a subject to allergens, the CRTH2 antagonist is suitably a compound of general formula (I).
Radioligand Binding Assay for Measurement of Binding to CRTH2 Receptor
Materials
Calcium-3 dye was purchased from Molecular Devices (Wokingham, UK). Mono- -poly resolving medium was obtained from Dainippon Pharmaceuticals (Osaka,
Japan). Macs anti-CD 16 microbeads were from Miltenyi biotec (Bisley, Surrey).
ChemoTx plates were purchased from Neuroprobe (Gaithesburg, MD). PoIy-D- lysine coated 96-well plates were obtained from Greiner (Gloucestershire, UK).
[3H]PGD2 was from Amersham Biosciences (Buckinghamshire, UK). [3H]SQ29548 was purchased from Perkin Elmer Life Sciences (Buckinghamshire, UK). All other reagents were obtained from Sigma-Aldrich (Dorset, UK), unless otherwise stated.
Methods Cell culture Chinese Hamster Ovary cells were transfected with CRTH2 receptors (CHO/CRTH2) and were maintained in culture in a humidified atmosphere at 370C (5% CO2) in Minimum Essential Medium (MEM) supplemented with 10% foetal bovine serum, 2 raM glutamine, and 1 mg ml'1 active G418. The cells were passaged every 2-3 days. For radioligand binding assay, cells were prepared in triple-layer flasks or in 175 cm2 square flasks (for membrane preparation). For calcium mobilisation assay, cells were grown in a 96 well plate 24h prior to the assay at a density of 80,000 cells per well.
Preparation of cell membranes
Membranes were prepared either from CHO/CRTH2 cells. CHO cells grown to confluency were washed with PBS and detached using a Versene solution (15 ml per flask). When the cells were grown in 175 cm2 square flask, they were collected by scrapping in PBS. The cell suspensions were centrifuged (1,700 rpm, 10 min, 40C) and resuspended in 15 ml of buffer (IxHBSS, supplemented with 10 mM HEPES, pH 7.3). Cell suspensions were then homogenised using an Ultra Turrax at setting 4- 6 for 20 s. The homogenate was centrifuged at 1,700 rpm for 10 min and the supernatant was collected and centrifuged at 20,000 rpm for Ih at 4°C. The resulting pellet was resuspended in buffer and stored at -800C in aliquots of 200-500 μl. The protein concentration was determined by the method of Bradford (1976), using bovine serum albumin as standard. The platelets were washed by centrifugation at 600xg for 10 min and resuspended in ice-cold assay buffer (10 mM Tris-HCl, pH 7.4, 5 mM Glucose, 120 mM NaCl, 10 μM indomethacin) and directly centrifuged at 20,000 rpm for 30 min at 40C. The resulting pellet was treated as described above.
Radioligand binding assays [3H]PGD2 (160 Ci/mmol) binding experiments were performed on membranes prepared as described above. Assays were performed in a final volume of 100 μl of buffer (1XHBSS/HEPES 10 mM, pH 7.3). Cell membranes (15μg). Cell membranes 15mg were preincubated at room temperature with varying concentration of competing ligand for 15 min. [3H]PGD2 (mol, final concentration) was then added and the incubation continued for a further one hour at room temperature. The reaction was terminated by the addition of 200 μl ice-cold assay buffer to each well, followed by rapid filtration through Whatman GF/B glass fibre filters using a Unifilter Cell harvester (PerkinElmer Life Sciences) and six washes of 300 μl of ice- cold buffer. The Unifilter plates were dried at room temperature for at least Ih and the radioactivity retained on the filters was determined on a Beta Trilux counter (PerkinElmer Life Sciences), following addition of 40 μl of Optiphase Hi-Safe 3 (Wallac) liquid scintillation. Non specific binding was defined in the presence of 10 μM unlabelled PGD2.
This assay is also described in Example 3 of WO 2005/044260. More suitable, compounds which are CRTH2 antagonists have a Ki of less than 100 nM and typically less than 50 nM or even less than 20 nM.
The CRTH2 antagonists useful in the present invention may be selective for the CRTH2 receptor and in this case will not have high affinity for the DP1 or TP receptors. Binding to the DPi receptor may be measured in a radioligand binding assay similar to the one described above for measurement of CRTH2 antagonist activity except that in the cell culture phase, the Chinese Hamster Ovary cells were transfected with DP1 receptors (CHO/DP), rather than with CRTH2 receptors, and the CHO/DP cells were used to prepare the cell membranes.
Compounds which do not bind to the DPi receptor have a Ki of lμM or greater in this assay. Indeed, for some compounds the Ki at the DP1 receptor may be at least 5μM or even at least 10 μM.
TP receptor radioligand binding may be carried out on membranes prepared from platelets. 15-40 μg of protein were pre-incubated with varying concentrations of competing ligand for 15 min at room temperature in assay buffer (10 mM Tris-HCl, pH 7.4, 5 mM glucose, 120 mM NaCl, 10 μM indomethacin). [3H]SQ29548 (38 Ci/mmol, 10 nM final concentration) was then added and the incubation continued for a further 30 min at room temperature. The reaction was terminated by the addition of 200 μl ice-cold assay buffer to each well, followed by rapid filtration through Whatman GF/C glass fibre filters using a Unifϊlter Cell harvester (PerkinElmer Life Sciences) followed with six washes of 300 μl of ice-cold buffer. The radioactivity was determined as described for the CRTH2 and DP1 receptors.
Compounds useful in the present invention which do not bind to the TP receptor have a Ki of lμM or greater in this assay. Indeed, for some compounds the Ki at the TP receptor may be at least 5μM or even at least 10 μM.
Among the most suitable compounds for use in the present invention are those in which the binding selectivity is greater than 200 fold for CRTH2 receptor, compared to DP1 TP receptors.
CRTH2 antagonist activity can be measured by the calcium mobilisation assay as follows.
Calcium Mobilisation Assay For Measurement of CRTH2 Antagonist Activity
Cells were seeded onto poly-D-lysine coated 96-well plates at a density of 80,000 cells per well and incubated at 37°C overnight to allow the cells to adhere. Cells were washed twice with HBSS and incubated for Ih at 37°C in lOOμl HBSS and lOOμl calcium-3-dye (Molecular Devices), supplemented with 4mM probenecid. Changes in fluorescence were monitored over a 50s time course with agonist addition at 17s using a Flexstation (Molecular Devices).
Effect ofCRTH2 agonists on calcium mobilisation in CHO-CRTH2 cells PPGGDD22 ccaauusseedd aa ddoossee--ddeeppeennddeenntt iinnccrreeaassee iinn iinnttrraaccellular Ca2+ mobilisation in CHO/CRTH2 cells, with an EC50 = 2.4 ± 0.5nM (n=3).
CRTH2 antagonist compounds inhibit this effect suitably with an IC50 of less than lμM, more appropriately less than 500 nM still more suitably less than 100 nM and typically less than 50 nM or even less than 20 nM.
The invention will now be described in greater detail with reference to the Examples and to the drawings in which:
FIGURE 1 is a series of plots showing the combined symptom scores of two groups of patients with allergic rhinitis in a test comparing a CRTH2 antagonist with a placebo on Days 2 and 8 of each treatment period. Figure IA shows the combined nasal symptom scores for both patient groups in response to challenge with a CRTH2 antagonist, while Figure IB shows the combined scores for eye symptoms. The plots show that patients taking the CRTH2 antagonist show reduced nasal and eye symptoms on both Day 2 and Day 8 of the treatment period.
FIGURE 2 is a series of plots similar to that of Figure 1 but where the scores for each patient group have been shown separately. The plots also show a screening response, which is the response of the patients to allergen before the beginning of the trial. Figures 2A and 2B show the total nasal symptoms for patient group A compared with the screening response score for the same group of patients. Figure 2A is plot showing the scores for nasal symptoms of patients in Group A on day 2 of each of the treatment periods while Figure 2B shows the nasal symptom scores for the same group of patients on day 8. Patient group A was treated with CRTH2 antagonist for 8 days, then given no treatment for 3 weeks, then treated with placebo for 8 days.
Figures 2C and 2D are similar to Figures 2 A and 2B but show the scores for nasal symptoms from patients from patient group B on days 2 and 8 of the treatment periods together with the screening response score for the same group of patients. Patient group B was treated with placebo for 8 days, then given no treatment for 3 weeks, then treated with CRTH2 antagonist for 8 days.
FIGURE 2 is a plot showing the anti-apoptotic effect of PGD2 in human Th2 cells. Human Th2 cells were treated with 50U/ml IL-2 or various concentrations of PGD2 in the absence of DL-2 for 16 hrs. The cells were stained with Annexin V-PE/PI and then analysed by FACSArray flow cytometer.
In the Examples, the following test compounds were used.
Compound 1 (5-Fluoro-2-methyl-3-quinolin-2-ylmethyl-indol- 1 -yl)-acetic acid
Compound 2 (3- { [2-(Benzenesulfonyl)phenyl]methyl } -5-fluoro-2-methylindol- 1 - yl)-acetic acid
Figure imgf000032_0001
Example 1 - Vienna Challenge Chamber Study
Study design
The study was a randomised, double blind, placebo controlled, two way crossover evaluation of Compound 1, given orally for eight days. There was a screening period of one week and a washout period of three weeks between the two treatment periods.
There was a follow up one week after the last dose of study drug. The group of patients who received the study drug for the first treatment period and placebo for the second was designated group A, while the group of patients who received placebo for the first treatment period and the study drug for the second treatment period was designated group B.
Treatment plan and methods
The subjects underwent a complete screening assessment to determine a baseline response to allergens. This screening assessment took place one week prior to the start of dosing.
• Subjects commenced dosing with Compound 1 or placebo on Day 1 (visit 2 or visit 5) of each treatment period of the study. Adverse events, total nasal symptom score and concomitant medications were noted in the CRF. Subjects took each dose (2 capsules in the morning and 2 capsules in the evening) as soon as they had finished breakfast and their evening meal respectively.
Subjects reported back to the clinic on Day 2 of each treatment period for a 6 hour allergen challenge. The following measurements were obtained: • Total nasal symptom score (TNSS) (obstruction, rhinorrhoea, itch, sneeze) with each symptom scored on a categorical scale from 0 to 3 pre-challenge, every 15 mins from 0 to 6h post-start of challenge
• Eye symptom score (watery eyes, itchy eyes, red eyes) with each symptom scored on a categorical scale from 0 to 3 pre-challenge, every 15mins from 0 to 6h post-start of challenge
• Other symptoms (cough, itchy throat, itchy ears) with each symptom scored on a categorical scale from 0 to 3 pre-challenge and every 15mins from 0 to 6h post-start of challenge
Subjects reported back to the clinic on Day 8 of each treatment period for a 6 hour allergen challenge and the measurements obtained on Day 2 were repeated.
A final follow-up visit was conducted one after the last dose of test article in Treatment Period 2.
Results
Subjects in Group A received a CRTH2 antagonist for 8 days, followed by a three week washout and then received placebo for 8 days. The subjects showed a reduced response to allergen after dosing with a CRTH2 antagonist. This reduced response to allergen was still present on dosing with placebo. Subjects in Group B received placebo for 8 days, followed by a three week washout and then received a CRTH2 antagonist for 8 days.
The results of the experiment are shown in Figures 1 and 2. When the results for the two groups of patients were combined, subjects show a 23% reduction in total nasal symptom score after 2 days of dosing with a CRTH2 antagonist compared to placebo. This reduction is increased to 37% after 8 days of dosing (Figure IA). The total eye symptom score was also reduced in patients treated with the CRTH2 antagonist - subjects show a 45% reduction in total eye symptom score after 2 days of dosing with a CRTH2 antagonist compared to placebo. This reduction is increased to 61% after 8 days of dosing (Figure IB). The figures for the individual patient groups were then obtained. Figures 2A and 2B shows that the total nasal symptom scores for patients in Group A on days 2 and 8 respectively of each study period compared with the screening response for these patients obtained at the beginning of the trial. Figures 2C and 2D show the equivalent data for Group B.
The subjects mounted the expected response to allergen on placebo and showed a reduction in TNSS in response to allergen after dosing with the CRTH2 antagonist.
The results show that the patients in Group B, who were treated firstly with placebo and then with Compound 1, responded as expected. Thus, as can be seen from Figures 2C and 2D, patients treated with placebo showed the same response as the baseline screening response obtained before the beginning of the trial.
However, for patients in Group A, the results were different. As shown in Figures 2 A and 2B, patients treated with placebo 3 to 4 weeks after the last dose of Compound 1 still had a nasal symptom score which was considerably lower than the baseline value obtained before the beginning of the trial. This indicates that Compound 1 has a long-lasting effect in the patients in Group A and that this effect lasts for more than 4 weeks after the last treatment with Compound 1.
The results demonstrate that in this experiment in the Vienna Challenge Chamber, a reduction of 36% in Total Nasal Symptom Score (0-6hr) remains 22 days after the last dose of a CRTH2 antagonist and a reduction of 38% in Total Nasal Symptom Score (0-6hr) remains 28 days after the last dose of a CRTH2 antagonist.
Example 2 - Apoptosis Challenge Test
Human Th2 cells were treated with 50U/ml IL-2 or various concentrations of PGD2 in the absence of IL-2 for 16 hrs. The cells were stained with Annexin V-PE/PI and then analysed by FACSArray flow cytometer. The results are illustrated in Figure 3, which shows that PGD2 has an anti-apoptotic effect on human Th2 cells. Human Th2 cells were treated in the absence of JL-2 with a medium containing 100 nM PGD2 and various concentrations of CRTH2 antagonistic compounds (Compounds 1, 2, 3, 4 and 5) for 16 hrs. The cells were stained with Annexin V- PE/PI and then analysed by FACSArray flow cytometer. The IC50 of Compounds 1 to 5 to the rescuing function of PGD2 were calculated and found to be less than 10OnM in all cases.
The results of this experiment show that while PGD2 has an anti-aptotic effect on Th2 cells, the CRTH2 antagonists have a pro-apoptotic effect.

Claims

1. A compound of general formula (I):
Figure imgf000036_0001
(I) wherein
R1 is Ci-C6 alkyl; R2 is halogen; and
R3 is aryl or heteroaryl optionally substituted with one or more substituents selected from halo, OH, CN, R6, COR6, CH2R6, OR6, SR6, SO2R6 or SO2YR6;
R6 is Ci-C6 alkyl, C3-C8 cycloalkyl, heterocyclyl, aryl or heteroaryl, any of which may optionally be substituted with one or more substituents selected from halo, OH, CN, NO2, C1-C6 alkyl or O(CrC6 alkyl); and Y is NH or a straight or branched C1-C4 alkylene chain; R4 is H or Ci-C4 alkyl;
R5 is hydrogen, Ci-C6 alkyl, aryl, (CH2)mOC(=O)Ci-C6alkyl, ((CH2)mO)nCH2CH2X, (CH2)mN(R7)2 or CH((CH2)mO(C=O)R8)2; m is 1 or 2; n is 1-4; X is OR7 or N(R7)2;
R7 is hydrogen or methyl; R8 is Ci-Ci8 alkyl;
or a pharmaceutically acceptable salt, hydrate, solvate, or complex thereof; for the treatment of an allergic condition which is induced by the presence of an allergen to which the patient is sensitive, wherein the treatment comprises at least one treatment cycle which comprises: a. a first period during which the compound of general formula (I) is administered to the patient; and b. a second period of at least seven days during which the compound of general formula (I) is administered to the patient in a reduced amount; wherein the allergen is present or suspected to be present throughout the treatment and wherein the compound of general formula (I) retains at least 50% of its effectiveness during the second period of the treatment cycle.
2. A pulsed method for the treatment of an allergic condition which is induced by the presence of an allergen to which a patient is sensitive, the method comprising administering to the patient over at least one treatment cycle a CRTH2 antagonist compound of general formula (I) as defined in claim 1, wherein the said treatment cycle comprises: a. a first period during which a compound of general formula (I) as defined in claim 1 is administered to the patient; and b. a second period of at least seven days during which the compound of general formula (I) as defined in claim 1 is administered to the patient in a reduced amount; wherein the allergen is present or suspected to be present throughout the treatment and wherein the compound of general formula (I) as defined in claim 1 retains at least 50% of its effectiveness during the second period of the treatment cycle.
3. The use of a CRTH2 antagonist compound of general formula (I) as defined in claim 1 in the preparation of an agent for the treatment of an allergic condition which is induced by the presence of an allergen to which a patient is sensitive, wherein the treatment comprises at least one treatment cycle which comprises: a. a first period during which the compound of general formula (I) as defined in claim 1 is administered to the patient; and b. a second period of at least seven days during which the compound of general formula (I) as defined in claim 1 is administered to the patient in a reduced amount; wherein the allergen is present or suspected to be present thoughout the treatment and wherein the wherein the compound of general formula (I) as defined in claim 1 retains at least 50% of its effectiveness during the second period of the treatment cycle.
4. A compound as claimed in claim 1, a method as claimed in claim 2 or the use as claimed in claim 3, wherein during the first period of the treatment cycle the patient experiences a reduction in symptoms of at least 50%.
5. A compound, a method or a use as claimed in any one of claims 1 to 4, wherein during the second period of the treatment cycle, the compound retains at 80% of its effectiveness.
6. A compound, a method or a use as claimed in any one of claims 1 to 5, wherein during the second period of the treatment cycle, administration of the
CRTH2 antagonist compound is discontinued.
7. A compound, a method or a use as claimed in any one of claims 1 to 6, wherein the second period of the treatment cycle lasts for at least 28 days.
8. A compound, a method or a use as claimed in any one of claims 1 to 7, wherein the first period of the treatment cycle is from two to 8 days.
9. A compound, a method or a use as claimed in any one of claims 1 to 8, wherein, in the first period of the treatment cycle, the compound is administered once daily.
10. A compound, a method or a use as claimed in any one of claims 1 to 9, further including one or more additional treatment cycles.
11. A compound, a method or a use as claimed in any one of claims 1 to 10, wherein the allergic condition is asthma (including allergic asthma), food allergies, acute and chronic urticaria, perennial allergic rhinitis, seasonal allergic rhinitis, atopic dermatitis, contact hypersensitivity (including contact dermatitis) or conjunctivitis, especially allergic conjunctivitis.
12. A compound, use or method as claimed in any one of claims 1 to 11, wherein, in the compound of general formula (I), R5 is hydrogen.
13. A compound, use or method as claimed in any one of claims 1 to 11, wherein, in the compound of general formula (I), R5 is C1-C6 alkyl, aryl, (CH2)mOC(=O)C1- C6alkyl, ((CH2)mO)nCH2CH2X, (CH2)mN(R7)2 or CH((CH2)raO(C=O)R8)2; where m is 1 or 2; n is 1-4;
X is OR7 or N(R7)2;
R7 is hydrogen or methyl; and R8 is Ci-Ci8 alkyl.
14. A compound, use or method as claimed in any one of claims 1 to 13, wherein in the compound of general formula (I), independently or in any combination:
R1 is fluoro; R2 is Ci-C4 alkyl, particularly methyl or ethyl but more especially methyl; R4 is H or methyl; and
R3 is quinoline, quinoxaline, isoquinoline, thiazole, phenyl, naphthalene, thiophene, pyrrole or pyridine, any of which may optionally be substituted as set out in claim 1.
15. A compound, use or method as claimed in claim 14 wherein, in the compound of general formula (I), R4 is H.
16. A compound, use or method as claimed in any one of claims 1 to 15 wherein, in the compound of general formula (I), R3 is optionally substituted quinoline, phenyl, naphthalene, thiophene, pyrrole or pyridine.
17. A compound, use or method as claimed in claim 16 wherein, in the compound of general formula (I), R3 is quinoline or isoquinoline, which is unsubstituted or substituted with one or more halo substituents.
18. A compound, use or method as claimed in claim 16, wherein, in the compound of general formula (I), R3 is phenyl, naphthalene, thiophene, pyrrole or pyridine, optionally substituted with one or more substituents selected from OR6, SO2R6 or SO2YR6; where R6 and Y are as defined in claim 1.
19. A compound, use or method as claimed in claim 18 wherein, in the compound of general formula (I), R6 is C1-C6 alkyl, a 4- to 6-membered cycloalkyl group, a 5- or 6-membered heterocyclyl group or phenyl, any of which may be substituted as defined in claim 1.
20. A compound, use or method as claimed in claim 16 wherein, in the compound of general formula (I), R3 is a 3-pyridyl moiety.
21. A compound, use or method as claimed in any one of claims 1 to 20 wherein R3 is substituted with SO2YR6, wherein Y is a CH2 moiety.
22 A compound, use or method as claimed in any one of claims 1 to 21, wherein R3 is substituted with SO2R6 or SO2YR6, wherein the R6 group is unsubstituted or substituted with one or more substituents chosen from methyl and halo.
23. A compound, use or method as claimed in any one of claims 1 to 20, wherein R3 is substituted with OR6, wherein the R6 group is unsubstituted or substituted with one or more substituents chosen from halo, cyano, C1-C4 alkyl and 0(Ci-C4 alkyl).
24. A compound, use or method as claimed in any one of claims 1 to 10, wherein the compound of general formula (I) is: { 3-[ 1 -(4-Chloro-phenyl)-ethyl]-5-fluoro-2-methyl-indol- 1-yl }-acetic acid;
{5-Fluoro-2-methyl-3-[l-(4-trifluoromethyl-phenyl)-ethyl]-indol-l-yl}-acetic acid; {3-[l-(4-tert-Butyl-phenyl)-ethyl]-5-fluoro-2-methyl-indol-l-yl}-acetic acid; {5-Fluoro-3-[l-(4-methanesulfonyl-phenyl)-ethyl]-2-methyl-indol-l-yl}-acetic acid;
[5-Fluoro-2-methyl-3-(l-naphthalen-2-yl-ethyl)-indol-l-yl]-acetic acid;
(5-Fluoro-2-methyl-3-quinolin-2-ylmethyl-indol-l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-naphthalen-2-ylmethyl-indol-l-yl)-acetic acid; [5-Fluoro-3-(8-hydroxyquinolin-2-ylmethyl)-2-methyl-indol-l-yl]-acetic acid;
[5-Fluoro-2-methyl-3-(quinoxalin-2-ylmethyl)indol-l-yl]-acetic acid;
[5-Fluoro-3-(4-methoxy-benzyl)-2-methyl-indol-l-yl]-acetic acid;
[5-Fluoro-2-methyl-3-(l,3-thiazol-2-ylmethyl)indol-l-yl]-acetic acid;
[3-(4-Chloro-benzyl)-5-fluoro-2-methyl-indol-l-yl]-acetic acid; [5-Fluoro-2-methyl-3-(4-trifluoromethyl-benzyl)-indol-l-yl]-acetic acid;
[5-Fluoro-2-methyl-3-(4-tert-butyl-benzyl)-indol-l-yl]-acetic acid;
{5-Fluoro-2-methyl-3-[(4-phenylphenyl)methyl]indol-l-yl}-acetic acid;
[5-Fluoro-3-(4-methanesulfonyl-benzyl)-2-methyl-indol-l-yl]-acetic acid;
{ 5-Fluoro-3-[(6-fluoroquinolin-2-yl)methyl]-2-methylindol-l-yl }-acetic acid; (2-Methyl-3-quinolin-2-ylmethyl-indol-l-yl)-acetic acid;
(5-Chloro-2-methyl-3-quinolin-2-ylmethyl-indol-l-yl)-acetic acid;
(3-{[l-(Benzenesulfonyl)pyrrol-2-yl]methyl}-5-fluoro-2-methylindol-l-yl)-acetic acid;
[5-Fluoro-2-methyl-3-({ l-[(4-methylbenzene)sulfonyl]pyrrol-2-yl}methyl)indol-l- yl]-acetic acid;
[3-({ l-[(2,4-Difluorobenzene)sulfonyl]pyrrol-2-yl}methyl)-5-fluoro-2-methylindol-
1-yl] -acetic acid;
(3-{[2-(Benzenesulfonyl)phenyl]methyl}-5-fluoro-2-methylindol-l-yl)-acetic acid;
[3-({ 2-[(4-Chlorobenzene)sulfonyl]phenyl }methyl)-5-fluoro-2-methylindol- 1-yl]- acetic acid;
[5-Fluoro-3-({2-[(4-fluorobenzene)sulfonyl]phenyl}methyl)-2-methylindol-l-yl]- acetic acid;
(3-{[2-(Benzenesulfonyl)pyridin-3-yl]methyl}-5-fluoro-2-methylindol-l-yl)-acetic acid; [5-Fluoro-3-({2-[(4-fluorobenzene)sulfonyl]pyridin-3-yl}methyl)-2-methylindol-l- yl] -acetic acid;
[3-({2-[(4-Chlorobenzene)sulfonyl]pyridin-3-yl}methyl)-5-fluoro-2-methylindol-l- yl]-acetic acid;
2-(3-(4-(Benzylsulfonyl)benzyl)-5-fluoro-2-methyl-indol-l-yl)-acetic acid;
2-(3-(4-(4-Chlorobenzylsulfonyl)benzyl)-5-fluoro-2-methyl-indol-l-yl)-acetic acid;
2-(3-(3-(Benzylsulfonyl)benzyl)-5-fluoro-2-methyl-indol-l-yl)-acetic acid; 2-(5-Fluoro-3-(3-(4-fluorobenzylsulfonyl)benzyl)-2-methyl-indol-l-yl)-acetic acid;
2-(3-(2-(Benzylsulfonyl)benzyl)-5-fluoro-2-methyl-indol-l-yl)-acetic acid;
2-(3-(4-(4-Fluorobenzylsulfonyl)benzyl)-5-fluoro-2-methyl-indol-l-yl)-acetic acid;
2-(3-(2-(Cyclohexylsulfonyl)benzyl)-5-fluoro-2-methyl-indol-l-yl)-acetic acid;
2-(5-Fluoro-2-methyl-3-(2-(piperidin-l-ylsulfonyl)benzyl)-indol-l-yl)-acetic acid; 2-(3-(2-(Cyclopentylsulfonyl)benzyl)-5-fluoro-2-methyl-indol-l-yl)-acetic acid;
2-(5-Fluoro-2-methyl-3-(3-(piperidin-l-ylsulfonyl)benzyl)-indol-l-yl)-acetic acid;
2-(5-Fluoro-2-methyl-3-(2-(pyrrolidin-l-ylsulfonyl)benzyl)-indol-l-yl)-acetic acid;
2-(3-(4-(Cyclohexylsulfonyl)benzyl)-5-fluoro-2-methyl-indol-l-yl)-acetic acid;
2-(3-(4-(Cyclopentylsulfonyl)benzyl)-5-fluoro-2-methyl-indol-l-yl)-acetic acid; 2-(3-(2-(Cyclobutylsulfonyl)benzyl)-5-fluoro-2-methyl-indol-l-yl)-acetic acid;
2-(5-Fluoro-2-methyl-3-(3-(pyrrolidin-l-ylsulfonyl)benzyl)-indol-l-yl)-acetic acid acid;
2-(5-Fluoro-2-methyl-3-(4-(piperidin-l-ylsulfonyl)benzyl)-indol-l-yl)-acetic acid;
[5-Fluoro-2-methyl-3-(2-phenoxybenzyl)-indol-l-yl]-acetic acid; [5-Fluoro-2-methyl-3-(2-(4-methoxyphenoxy)benzyl)-indol-l-yl]-acetic acid;
[5-Fluoro-2-methyl-3-(2-(4-methylphenoxy)benzyl)-indol-l-yl]-acetic acid;
[5-Fluoro-2-methyl-3-(2-(2,4-dichlorophenoxy)benzyl)-indol-l-yl]-acetic acid;
[5-Fluoro-2-methyl-3-(2-(4-fluorophenoxy)benzyl)-indol-l-yl]-acetic acid;
[5-Fluoro-2-methyl-3-(2-(3,4-difluorophenoxy)benzyl)-indol-l-yl]-acetic acid; [5-Fluoro-2-methyl-3-(2-(4-cyanophenoxy)benzyl)-indol-l-yl]-acetic acid;
[5-Fluoro-2-methyl-3-(2-(4-chlorophenoxy)benzyl)-indol-l-yl]-acetic acid;
[5-Fluoro-2-methyl-3-(2-(2-cyanophenoxy)benzyl)-indol-l-yl]-acetic acid;
(5-Fluoro-2-methyl-3- { [2-(4-methylphenoxy)pyridin-3-yl]methyl } indol- 1 -yl)-acetic acid; { 5-Fluoro-3-[(3-methanesulfonylnaphthalen-2-yl)methyl]-2-methylindol-l-yl }-acetic acid;
{5-Fluoro-3-[(l-methanesulfonylnaphthalen-2-yl)methyl]-2-methylindol-l-yl}-acetic acid;
{5-Fluoro-3-[(6-methanesulfonylnaphthalen-2-yl)methyl]-2-methylindol-l-yl}-acetic acid;
[5-Fluoro-2-methyl-3-(quinolin-3-ylmethyl)indol-l-yl]-acetic acid; [5-Fluoro-2-methyl-3-(quinoxalin-6-ylmethyl)indol-l-yl]-acetic acid;
[5-Fluoro-2-methyl-3-(quinolin-7-ylmethyl)indol-l-yl]-acetic acid;
{5-Fluoro-3-[(6-methanesulfonylquinolin-2-yl)methyl]-2-methylindol-l-yl}-acetic acid;
{ 5-Fluoro-3-[(4-methanesulfonylquinolin-2-yl)methyl] -2-methylindol- 1 -yl } -acetic acid;
(5-Fluoro-2-methyl-3-{pyrazolo[l,5-a]pyridin-3-ylmethyl}indol-l-yl)-acetic acid;
(5-Fluoro-3- { imidazo[ 1 ,2-a]pyridin-2-ylmethyl } -2-methylindol- l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-{ [2-(methylsulfanyl)phenyl]methyl}indol-l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-{ [3-(methylsulfanyl)phenyl]methyl }indol-l-yl)-acetic acid; (5-Fluoro-2-methyl-3- { [4-(ethylsulfanyl)phenyl]methyl } indol- 1 -yl)-acetic acid(3-
{ [4-(Ethylsulfanyl)phenyl]methyl}-5-fluoro-2-methylindol-l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-{ [4-(n-propylsulfanyl)phenyl]methyl}indol-l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-{ [4-(i-propylsulfanyl)phenyl]methyl}indol-l-yl)-acetic acid;
(5-Ruoro-2-methyl-3-{ [4-(t-butylsulfanyl)phenyl]methyl } indol- l-yl)-acetic acid; (5-Fluoro-2-methyl-3-{ [4-(pentan-3-ylsulfanyl)phenyl]methyl } indol- l-yl)-acetic acid;
[3-({4-[(Cyclopropylmethyl)sulfanyl]phenyl}methyl)-5-fluoro-2-methylindol-l-yl]- acetic acid;
{3-[(4,4-Dimethyl-2,3-dihydro-l-benzothiopyran-6-yl)methyl]-5-fluoro-2- methylindol- 1 -yl } -acetic acid;
(3-{ [2-(Ethanesulfonyl)phenyl]methyl }-5-fluoro-2-methylindol-l-yl)-acetic acid;
(5-Fluoro-2-methyl-3- { [2-(propane- l-sulfonyl)phenyl]methyl }indol- l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-{[2-(propane-2-sulfonyl)phenyl]methyl}indol-l-yl)-acetic acid;
(3-{[2-(Butane-l-sulfonyl)phenyl]methyl}-5-fluoro-2-methylindol-l-yl)-acetic acid;
(3-{[2-(Butane-2-sulfonyl)phenyl]methyl}-5-fluoro-2-methylindol-l-yl)-acetic acid; (5-Fluoro-2-methyl-3-{[2-(2-methylpropane-2-sulfonyl)phenyl]methyl}indol-l-yl)- acetic acid;
(5-Fluoro-2-methyl-3-{[2-(pentane-l-sulfonyl)phenyl]methyl}indol-l-yl)-acetic acid; (3-{ [2-(Cyclopropylmethane)sulfonylphenyl]methyl }-5-fluoro-2-methylindol-l-yl)- acetic acid;
(5-Fluoro-2-methyl-3-{ [2-(propylsulfamoyl)phenyl]methyl }indol-l-yl)-acetic acid;
(3-{[2-(Butylsulfamoyl)phenyl]methyl}-5-fluoro-2-methylindol-l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-{ [3-(propylsulfamoyl)phenyl]methyl }indol-l-yl)-acetic acid; (3-{ [3-(Butylsulfamoyl)phenyl]methyl }-5-fluoro-2-methylindol-l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-{[4-(trifluoromethane)sulfonylphenyl]methyl}indol-l-yl)- acetic acid;
(3-{[4-(Ethanesulfonyl)phenyl]methyl}-5-fluoro-2-methylindol-l-yl)-acetic acid;
(S-Fluoro^-methyl-S-l^-Cpropane-l-sulfonyOphenylJmethylJindol-l-yO-acetic acid;
(5-Fluoro-2-methyl-3-{[4-(propane-2-sulfonyl)phenyl]methyl}indol-l-yl)-acetic acid;
(3-{[4-(Butane-l-sulfonyl)phenyl]methyl}-5-fluoro-2-methylindol-l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-{[4-(2-methylpropane-2-sulfonyl)phenyl]methyl}indol-l-yl)- acetic acid;
(5-Fluoro-2-methyl-3-{[4-(pentane-l-sulfonyl)phenyl]methyl}indol-l-yl)-acetic acid;
(5-Fluoro-2-methyl-3-{[4-(pentan-3-ylsulfonyl)phenyl]methyl}indol-l-yl)-acetic acid; [3-({4-[(Cyclopropylmethyl)sulfonyl]phenyl}methyl)-5-fluoro-2-methylindol-l-yl]- acetic acid;
(5-Fluoro-2-methyl-3-{ [4-(propylsulfamoyl)phenyl]methyl }indol-l-yl)-acetic acid;
(S-IK-CButylsulfamoy^phenyljmethylJ-S-fluoro^-methylindol-l-yO-acetic acid;
(5-Fluoro-2-methyl-3-{[4-(trifluoromethoxy)phenyl]methyl}indol-l-yl)-acetic acid; (5-Fluoro-3-{[4-methanesulfonyl-3-(trifluoromethyl)phenyl]methyl}-2-methylindol- l-yl)-acetic acid;
(5-Fluoro-3- { [4-methanesulfonyl-3-(trifluoromethoxy)phenyl]methyl } -2- methylindol-l-yl)-acetic acid;
{ 5 -Fluoro-3 - [(5 -methanesulfonylthiophen-2-yl)methyl] -2-methylindol- 1 -yl } -acetic acid;
{ 3-[(4,4-dimethyl- 1 , l-dioxo-2,3-dihydro- lλ6-benzothiopyran-6-yl)methyl]-5-fluoro- 2-methylindol- 1-yl} -acetic acid;
[3-({ l-[(4-Chlorobenzene)sulfonyl]pyrrol-2-yl}methyl)-5-fluoro-2-methylindol-lyl]- acetic acid;
[5-Fluoro-3-({ l-[(4-fluorobenzene)sulfonyl]pyrrol-2-yl}methyl)-2-methylindol-l- yl]-acetic acid; [5-Fluoro-3-({ l-[(4-methoxybenzene)sulfonyl]pyrrol-2-yl }methyl)-2-methylindol-l- yl]-acetic acid;
{3-[l-(2,4-Dichloro-benzenesulfonyl)pyrrol-2-ylmethyl]-5-fluoro-2-methyl-indol-l- yl} -acetic acid;
[5-Fluoro-3-({ l-[(4-methanesulfonylbenzene)sulfonyl]pyrrol-2-yl}methyl)-2- methylindol-l-yl]-acetic acid;
{5-Fluoro-2-methyl-3-[(2-phenylphenyl)methyl]indol-l-yl}-acetic acid;
(3- { [ l-(Benzenesulfonyl)indol-2-yl]methyl } -5-fluoro-2-methylindol- 1 -yl)-acetic acid;
(3- { [2-(4-Chlorophenyl)phenyl]methyl } -5-fluoro-2-methylindol- 1 -yl)-acetic acid; (5-Fluoro-2-methyl-3-{ [2-(4-methylphenyl)phenyl]methyl}indol-l-yl)-acetic acid;
{5-Fluoro-2-methyl-3-[(3-phenoxyphenyl)methyl]indol-l-yl}-acetic acid;
[5-Fluoro-3-({4-[(4-fluorophenyl)carbonyl]-l-methylpyrrol-2-yl}methyl)-2- methylindol- 1-yl] -acetic acid;
{ 5-Fluoro-2-methyl-3-[(6-{ [3-(trifluoromethyl)phenyl]methyl }pyridin-3- yl)methyl]indol- 1-yl} -acetic acid;
{ 5-Fluoro-2-methyl-3-[(3-phenoxythiophen-2-yl)methyl]indol-l-yl }-acetic acid;
(3-{[2-(Benzenesulfonyl)-l,3-thiazol-5-yl]methyl}-5-fluoro-2-methylindol-l-yl)- acetic acid;
{3-[(l-Benzylpyrazol-4-yl)methyl]-5-fluoro-2-methylindol-l-yl}-acetic acid; (3-{[5-(4-Chlorophenoxy)-l-methyl-3-(trifluoromethyl)pyrazol-4-yl]methyl}-5- fluoro-2-methylindol-l-yl)-acetic acid;
[3-({5-[(4-Chlorobenzene)sulfonyl]furan-2-yl}methyl)-5-fluoro-2-methylindol-l-yl]- acetic acid;
[3-({5-[(4-Chlorobenzene)sulfonyl]thiophen-2-yl}methyl)-5-fluoro-2-methylindol-l- yl] -acetic acid;
[3-({3-[(4-Chlorobenzene)sulfonyl]thiophen-2-yl}methyl)-5-fluoro-2-methylindol-l- yl] -acetic acid;
{ 3-[(2-Benzylphenyl)methyl]-5-fluoro-2-methylindol-l-yl }-acetic acid;
or the C1-C6 alkyl, aryl, (CH2)mOC(=O)C1-C6alkyl, ((CH2)mO)nCH2CH2X, (CH2)mN(R7)2 or CH((CH2)raO(C=O)R8)2 esters of any of the above; wherein m is 1 or 2; n is 1-4;
X is OR7 or N(R7)2;
R7 is hydrogen or methyl;
R8 is Ci-Ci8 alkyl.
25. A compound, use or method as claimed in any one of claims 1 to 24, wherein the compound of general formula (I) is administered in combination with one or more additional agent which is of use in the treatment of these allergic conditions.
26. A compound, use or method as claimed in claim 25, wherein the additional agent is selected from:
Suplatast tosylate and similar compounds;
B2 adrenoreceptor agonists such as metaproterenol, isoproterenol, isoprenaline, albuterol, salbutamol, formoterol, salmeterol, terbutaline, orciprenaline, bitolterol mesylate ,pirbuterol and indacaterol or methylxanthanines such as theophylline and aminophylline, mast cell stabilisers such as sodium cromoglycate or muscarinic receptor antagonists such as ipratropium and tiotropium; antihistamines, for example histamine Hi receptor antagonists such as loratadine cetirizine, desloratadine, levocetirizine, fexofenadine, astemizole, azelastine and chlorpheniramine or H4 receptor antagonists;
(X1 and α2 adrenoreceptor agonists such as propylhexedrine phenylephrine, phenylpropanolamine, pseudoephedrine, naphazoline hydrochloride, oxymetazoline hydrochloride, tetrahydrozoline hydrochloride, xylometazoline hydrochloride and ethylnorepinephrine hydrochloride; corticosteroids such as prednisone, prednisolone, flunisolide, triamcinolone acetonide, beclomethasone dipropionate, budesonide, fluticasone propionate, fluticasone furoate, mometasone furoate and ciclesonide; allergen immunotherapy such as Grazax.
27. A compound, use or method as claimed in any one of the preceding claims, wherein the compound of general formula (I) is administered via the oral, nasal, bronchial or topical route.
PCT/GB2008/003824 2007-11-13 2008-11-13 Use of crth2 antagonist compounds WO2009063202A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2010532660A JP2011503045A (en) 2007-11-13 2008-11-13 Use of CRTH2 antagonistic compounds
DE08851028T DE08851028T1 (en) 2007-11-13 2008-11-13 USE OF CRTH2 ANTAGONIST COMPOUNDS
EP08851028A EP2219645A2 (en) 2007-11-13 2008-11-13 Use of crth2 antagonist compounds
US12/779,638 US20110124683A1 (en) 2007-11-13 2010-05-13 Use of CRTH2 Antagonist Compounds

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB0722203.7 2007-11-13
GBGB0722203.7A GB0722203D0 (en) 2007-11-13 2007-11-13 Use of CRTH2 antagonist compounds

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2008/003843 Continuation-In-Part WO2009063215A2 (en) 2007-11-13 2008-11-13 Use of crth2 antagonist compounds

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/779,638 Continuation-In-Part US20110124683A1 (en) 2007-11-13 2010-05-13 Use of CRTH2 Antagonist Compounds

Publications (2)

Publication Number Publication Date
WO2009063202A2 true WO2009063202A2 (en) 2009-05-22
WO2009063202A3 WO2009063202A3 (en) 2009-08-27

Family

ID=38858584

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2008/003824 WO2009063202A2 (en) 2007-11-13 2008-11-13 Use of crth2 antagonist compounds

Country Status (6)

Country Link
EP (1) EP2219645A2 (en)
JP (1) JP2011503045A (en)
DE (1) DE08851028T1 (en)
ES (1) ES2353055T1 (en)
GB (1) GB0722203D0 (en)
WO (1) WO2009063202A2 (en)

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7750027B2 (en) 2008-01-18 2010-07-06 Oxagen Limited Compounds having CRTH2 antagonist activity
EP2240444A1 (en) * 2008-01-22 2010-10-20 Oxagen Limited Compounds having crth2 antagonist activity
EP2265581A1 (en) * 2008-01-22 2010-12-29 Oxagen Limited Compounds having crth2 antagonist activity
US8034826B2 (en) 2008-11-06 2011-10-11 Panmira Pharmaceuticals, Llc Cycloalkane[B]azaindole antagonists of prostaglandin D2 receptors
US8049015B2 (en) 2008-09-29 2011-11-01 Panmira Pharmaceuticals, Llc Heteroaryl antagonists of prostaglandin D2 receptors
US8071807B2 (en) 2008-07-03 2011-12-06 Panmira Pharmaceuticals, Llc Antagonists of prostaglandin D2 receptors
US8242145B2 (en) 2008-02-14 2012-08-14 Panmira Pharmaceuticals, Llc Cyclic diaryl ether compounds as antagonists of prostaglandin D2 receptors
WO2012119841A1 (en) 2011-03-07 2012-09-13 Oxagen Limited Amorphous (5-fluoro-2-methyl-3-quinolin-2-ylmethyl-indol-1-yl)-acetic acid
US8338484B2 (en) 2008-02-01 2012-12-25 Panmira Pharmaceuticals, Llc N,N-disubstituted aminoalkylbiphenyl antagonists of prostaglandin D2 receptors
US8383654B2 (en) 2008-11-17 2013-02-26 Panmira Pharmaceuticals, Llc Heterocyclic antagonists of prostaglandin D2 receptors
US8426449B2 (en) 2008-04-02 2013-04-23 Panmira Pharmaceuticals, Llc Aminoalkylphenyl antagonists of prostaglandin D2 receptors
CN103086943A (en) * 2011-11-04 2013-05-08 山东亨利医药科技有限责任公司 Indoles derivative functioning as CRTH2 receptor antagonist
US8501959B2 (en) 2008-06-24 2013-08-06 Panmira Pharmaceuticals, Llc Cycloalkane[B]indole antagonists of prostaglandin D2 receptors
US8697869B2 (en) 2010-03-22 2014-04-15 Actelion Pharmaceuticals Ltd. 3-(heteroaryl-amino)-1,2,3,4-tetrahydro-9H-carbazole derivatives and their use as prostaglandin D2 receptor modulators
US8785393B2 (en) 2009-07-31 2014-07-22 Panmira Pharmaceuticals, Llc Ophthalmic pharmaceutical compositions of DP2 receptor antagonists
US8815917B2 (en) 2009-08-05 2014-08-26 Panmira Pharmaceuticals, Llc DP2 antagonist and uses thereof
US9096595B2 (en) 2011-04-14 2015-08-04 Actelion Pharmaceuticals Ltd 7-(heteroaryl-amino)-6,7,8,9-tetrahydropyrido[1,2-a]indol acetic acid derivatives and their use as prostaglandin D2 receptor modulators
RU2589709C2 (en) * 2010-12-23 2016-07-10 Мерк Шарп Энд Домэ Корп. Quinoxalines and azaquinoxalines as crth2 receptor modulators
US9688624B2 (en) 2010-01-06 2017-06-27 Brickell Biotech, Inc. DP2 antagonist and uses thereof
US9850241B2 (en) 2014-03-18 2017-12-26 Idorsia Pharmaceuticals Ltd Azaindole acetic acid derivatives and their use as prostaglandin D2 receptor modulators
US9879006B2 (en) 2014-03-17 2018-01-30 Idorsia Pharmaceuticals Ltd Azaindole acetic acid derivatives and their use as prostaglandin D2 receptor modulators
US9951042B2 (en) 2014-05-02 2018-04-24 Atopix Therapeutics Limited Polymorphic form of [5-fluoro-3-({2-[(4-fluorobenzene) sulfonyl] pyridin-3-yl}methyl)-2-methylindol-1-yl]-acetic acid
US10011584B2 (en) 2014-05-02 2018-07-03 Atopix Therapeutics Limited Polymorphic form of [5-fluoro-3-({2-[(4-fluorobenzene) sulfonyl]pyridin-3-yl}methyl)-2-methylindol-1-yl]-acetic acid
US10351560B2 (en) 2015-09-15 2019-07-16 Idorsia Pharmaceuticals Ltd Crystalline forms
RU2756270C2 (en) * 2016-07-21 2021-09-29 Чиа Тай Тянцин Фармасьютикал Груп Ко., Лтд. Indole derivative used as crth2 inhibitor

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2229358B1 (en) * 2007-12-14 2011-03-23 Pulmagen Therapeutics (Asthma) Limited Indoles and their therapeutic use
DK2250161T3 (en) * 2008-01-18 2014-01-27 Atopix Therapeutics Ltd Compounds with CRTH2 antagonist activity
WO2017104728A1 (en) * 2015-12-16 2017-06-22 国立大学法人東京大学 Medicine for treating food allergy

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005044260A1 (en) * 2003-10-23 2005-05-19 Oxagen Limited Use of crth2 antagonist compounds in therapy
WO2006092579A1 (en) * 2005-03-01 2006-09-08 Oxagen Limited Microcrystalline ( 5-fluoro-2-methyl-3-quinolin-2-ylmethyl-indol-1-yl) acetic acid
WO2006095183A1 (en) * 2005-03-11 2006-09-14 Oxagen Limited 1-acetic acid-indole derivatives with pgd2 antagonist activity
WO2007107772A1 (en) * 2006-03-22 2007-09-27 Oxagen Limited Salts with crth2 antagonist activity
WO2008012511A1 (en) * 2006-07-22 2008-01-31 Oxagen Limited Compounds having crth2 antagonist activity

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005044260A1 (en) * 2003-10-23 2005-05-19 Oxagen Limited Use of crth2 antagonist compounds in therapy
WO2006092579A1 (en) * 2005-03-01 2006-09-08 Oxagen Limited Microcrystalline ( 5-fluoro-2-methyl-3-quinolin-2-ylmethyl-indol-1-yl) acetic acid
WO2006095183A1 (en) * 2005-03-11 2006-09-14 Oxagen Limited 1-acetic acid-indole derivatives with pgd2 antagonist activity
WO2007107772A1 (en) * 2006-03-22 2007-09-27 Oxagen Limited Salts with crth2 antagonist activity
WO2008012511A1 (en) * 2006-07-22 2008-01-31 Oxagen Limited Compounds having crth2 antagonist activity

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
ANONYMOUS: "View of NCT00290381 on 2006_02_10" CLINICALTRIALS.GOV ARCHIVE, [Online] 10 February 2006 (2006-02-10), XP002534990 Retrieved from the Internet: URL:http://clinicaltrials.gov/archive/NCT00290381/2006_02_10> *
ANONYMOUS: "View of NCT00697281 on 2008_06_12" CLINICAL TRIALS.GOV ARCHIVE, 12 June 2008 (2008-06-12), XP002535051 Retrieved from the Internet: URL:http://clinicaltrials.gov/archive/NCT00697281/2008_06_12> *
See also references of EP2219645A2 *

Cited By (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7919512B2 (en) 2008-01-18 2011-04-05 Oxagen Limited Compounds having CRTH2 antagonist activity
US7750027B2 (en) 2008-01-18 2010-07-06 Oxagen Limited Compounds having CRTH2 antagonist activity
US8563536B2 (en) 2008-01-18 2013-10-22 Atopix Therapeutics Limited Compounds having CRTH2 antagonist activity
US8536158B2 (en) 2008-01-18 2013-09-17 Atopix Therapeutics Limited Compounds having CRTH2 antagonist activity
US8980927B2 (en) 2008-01-18 2015-03-17 Atopix Therapeutics Limited Compounds having CRTH2 antagonist activity
EP2240444A1 (en) * 2008-01-22 2010-10-20 Oxagen Limited Compounds having crth2 antagonist activity
EP2265581A1 (en) * 2008-01-22 2010-12-29 Oxagen Limited Compounds having crth2 antagonist activity
US8362044B2 (en) 2008-02-01 2013-01-29 Panmira Pharmaceuticals, Llc N,N-disubstituted aminoalkylbiphenyl antagonists of prostaglandin D2 receptors
US8338484B2 (en) 2008-02-01 2012-12-25 Panmira Pharmaceuticals, Llc N,N-disubstituted aminoalkylbiphenyl antagonists of prostaglandin D2 receptors
US8242145B2 (en) 2008-02-14 2012-08-14 Panmira Pharmaceuticals, Llc Cyclic diaryl ether compounds as antagonists of prostaglandin D2 receptors
US8426449B2 (en) 2008-04-02 2013-04-23 Panmira Pharmaceuticals, Llc Aminoalkylphenyl antagonists of prostaglandin D2 receptors
US8501959B2 (en) 2008-06-24 2013-08-06 Panmira Pharmaceuticals, Llc Cycloalkane[B]indole antagonists of prostaglandin D2 receptors
US8247602B2 (en) 2008-07-03 2012-08-21 Panmira Pharmaceuticals, Llc Antagonists of prostaglandin D2 receptors
US8071807B2 (en) 2008-07-03 2011-12-06 Panmira Pharmaceuticals, Llc Antagonists of prostaglandin D2 receptors
US8049015B2 (en) 2008-09-29 2011-11-01 Panmira Pharmaceuticals, Llc Heteroaryl antagonists of prostaglandin D2 receptors
US8034826B2 (en) 2008-11-06 2011-10-11 Panmira Pharmaceuticals, Llc Cycloalkane[B]azaindole antagonists of prostaglandin D2 receptors
US8383654B2 (en) 2008-11-17 2013-02-26 Panmira Pharmaceuticals, Llc Heterocyclic antagonists of prostaglandin D2 receptors
US8785393B2 (en) 2009-07-31 2014-07-22 Panmira Pharmaceuticals, Llc Ophthalmic pharmaceutical compositions of DP2 receptor antagonists
US8815917B2 (en) 2009-08-05 2014-08-26 Panmira Pharmaceuticals, Llc DP2 antagonist and uses thereof
US9688624B2 (en) 2010-01-06 2017-06-27 Brickell Biotech, Inc. DP2 antagonist and uses thereof
US8697869B2 (en) 2010-03-22 2014-04-15 Actelion Pharmaceuticals Ltd. 3-(heteroaryl-amino)-1,2,3,4-tetrahydro-9H-carbazole derivatives and their use as prostaglandin D2 receptor modulators
RU2589709C2 (en) * 2010-12-23 2016-07-10 Мерк Шарп Энд Домэ Корп. Quinoxalines and azaquinoxalines as crth2 receptor modulators
WO2012119841A1 (en) 2011-03-07 2012-09-13 Oxagen Limited Amorphous (5-fluoro-2-methyl-3-quinolin-2-ylmethyl-indol-1-yl)-acetic acid
US8980918B2 (en) 2011-03-07 2015-03-17 Atopix Therapeutics Limited Amorphous (5-fluoro-2-methyl-3-quinolin-2-ylmethyl-indol-1-yl)-acetic acid
EP3345897A1 (en) 2011-03-07 2018-07-11 Atopix Therapeutics Limited Amorphous (5-fluoro-2-methyl-3-quinolin-2-ylmethyl-indol-1-yl)-acetic acid
US8703956B2 (en) 2011-03-07 2014-04-22 Atopix Therapeutics Limited Amorphous (5-fluoro-2-methyl-3-quinolin-2-ylmethyl-indol-1-yl)-acetic acid
US9096595B2 (en) 2011-04-14 2015-08-04 Actelion Pharmaceuticals Ltd 7-(heteroaryl-amino)-6,7,8,9-tetrahydropyrido[1,2-a]indol acetic acid derivatives and their use as prostaglandin D2 receptor modulators
CN103086943B (en) * 2011-11-04 2015-04-15 山东亨利医药科技有限责任公司 Indoles derivative functioning as CRTH2 receptor antagonist
CN103086943A (en) * 2011-11-04 2013-05-08 山东亨利医药科技有限责任公司 Indoles derivative functioning as CRTH2 receptor antagonist
US9879006B2 (en) 2014-03-17 2018-01-30 Idorsia Pharmaceuticals Ltd Azaindole acetic acid derivatives and their use as prostaglandin D2 receptor modulators
US10301309B2 (en) 2014-03-17 2019-05-28 Idorsia Pharmaceuticals Ltd Azaindole acetic acid derivatives and their use as prostaglandin D2 receptor modulators
US9850241B2 (en) 2014-03-18 2017-12-26 Idorsia Pharmaceuticals Ltd Azaindole acetic acid derivatives and their use as prostaglandin D2 receptor modulators
US9951042B2 (en) 2014-05-02 2018-04-24 Atopix Therapeutics Limited Polymorphic form of [5-fluoro-3-({2-[(4-fluorobenzene) sulfonyl] pyridin-3-yl}methyl)-2-methylindol-1-yl]-acetic acid
US10011584B2 (en) 2014-05-02 2018-07-03 Atopix Therapeutics Limited Polymorphic form of [5-fluoro-3-({2-[(4-fluorobenzene) sulfonyl]pyridin-3-yl}methyl)-2-methylindol-1-yl]-acetic acid
US10351560B2 (en) 2015-09-15 2019-07-16 Idorsia Pharmaceuticals Ltd Crystalline forms
RU2756270C2 (en) * 2016-07-21 2021-09-29 Чиа Тай Тянцин Фармасьютикал Груп Ко., Лтд. Indole derivative used as crth2 inhibitor

Also Published As

Publication number Publication date
EP2219645A2 (en) 2010-08-25
GB0722203D0 (en) 2007-12-19
DE08851028T1 (en) 2011-02-24
WO2009063202A3 (en) 2009-08-27
ES2353055T1 (en) 2011-02-25
JP2011503045A (en) 2011-01-27

Similar Documents

Publication Publication Date Title
EP2219645A2 (en) Use of crth2 antagonist compounds
WO2009063215A2 (en) Use of crth2 antagonist compounds
US20110124683A1 (en) Use of CRTH2 Antagonist Compounds
EP2250161B1 (en) Compounds having crth2 antagonist activity
US20130052190A1 (en) CRTH2 Antagonists for Treatment of Eosinophilic Diseases and Conditions
RU2412934C2 (en) Application of crth2 antagonists in therapy
US7750027B2 (en) Compounds having CRTH2 antagonist activity
JP5932848B2 (en) Amorphous (5-fluoro-2-methyl-3-quinolin-2-ylmethyl-indol-1-yl) -acetic acid
US20130224151A1 (en) Use of FAAH Inhibitors for Treating Abdominal, Visceral and Pelvic Pain
JP2008514637A (en) CRTH2, COX-2 and FAAH modulators
JP2007533725A (en) Neuropathic pain treatment method using CRTH2 receptor antagonist
EP1988898A2 (en) Pharmaceutical compositions for the treatment of attention deficit hyperactivity disorder comprising flibanserin
KR20200053639A (en) Pharmaceutical composition comprising glutarimide derivatives and use thereof in the treatment of eosinophilic diseases
JP2023030014A (en) Inhibitor of thiol isomerases and use thereof
US20180256534A1 (en) Ketamine and cytochrome p 450 inhibitor combinations
EA015483B1 (en) Use of a p38 kinase inhibitor for treating psychiatric disorders
TW201625253A (en) Pgd2-antagonist-containing medicine for treatment of symptoms associated with allergic diseases
JP2008127365A (en) Prophylactic and/or therapeutic agent of allergic disease
JP2002524507A (en) New composition
US20050113433A1 (en) Compositions of a phenyl acetic acid cyclooxygenase-2 selective inhibitor and a cholinergic agent for the treatment of reduced blood flow or trauma to the central nervous system
JPWO2007102571A1 (en) Functional brain disease treatment

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: 08851028

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2010532660

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2008851028

Country of ref document: EP