WO2018162562A1 - Use of an ep4 antagonist for the treatment of inflammatory pain - Google Patents

Use of an ep4 antagonist for the treatment of inflammatory pain Download PDF

Info

Publication number
WO2018162562A1
WO2018162562A1 PCT/EP2018/055615 EP2018055615W WO2018162562A1 WO 2018162562 A1 WO2018162562 A1 WO 2018162562A1 EP 2018055615 W EP2018055615 W EP 2018055615W WO 2018162562 A1 WO2018162562 A1 WO 2018162562A1
Authority
WO
WIPO (PCT)
Prior art keywords
inhibitors
compound
pain
treatment
inflammatory pain
Prior art date
Application number
PCT/EP2018/055615
Other languages
French (fr)
Inventor
Jens Nagel
Stefan BÄURLE
Maik Obendorf
Markus Koch
Gernot Langer
Anne-Marie GODINHO-COELHO
Original Assignee
Bayer Pharma Aktiengesellschaft
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 Bayer Pharma Aktiengesellschaft filed Critical Bayer Pharma Aktiengesellschaft
Publication of WO2018162562A1 publication Critical patent/WO2018162562A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic 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
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P23/00Anaesthetics

Definitions

  • the present invention relates to the use of an EP4 antagonist for the treatment and/or prophylaxis of inflammatory pain and/or other pathological conditions related to increased activation of EP4 receptors by prostaglandin E2 (PGE2).
  • PGE2 prostaglandin E2
  • inflammatory pain is one form of nociceptive pain which is typically accompanied by an immune response and mediated by pro-inflammatory molecules which are released by tissue due to a trigger which endangers or destroys cell integrity or signals endangered cell integrity.
  • activated nociceptors peripheral nerve endings of the pain pathway
  • non-neural cells that reside within or infiltrate into the injured/endangered area (including mast cells, basophils, platelets, macrophages, neutrophils, endothelial cells, keratinocytes, and fibroblasts).
  • these factors represent a wide array of signaling molecules, including neurotransmitters, peptides (substance P, CGRP, bradykinin), eicosinoids and related lipids like prostaglandins which include the endogenous activators of the EP4 receptor, thromboxanes, leukotrienes, endocannabinoids, neurotrophins, cytokines, and chemokines, as well as extracellular proteases and protons.
  • Nociceptors express one or more cell-surface receptors capable of recognizing and responding to each of these pro- inflammatory or pro-algesic agents.
  • Inflammatory pain of varied origins is associated but not limited to osteoarthritis, rheumatoid arthritis, rheumatic disease, tenosynovitis, gout, ankylosing spondylitis, endometriosis, injuries, various skin diseases and bursitis.
  • NSAID nonsteroidal anti-inflammatory drugs
  • PGE2 endogenously formed prostaglandin E2
  • EP4 belongs to the family of membrane- bound G-protein coupled receptors (GPCR) and is mainly provided with a Gs coupling, which after activation leads to an accumulation of the intracellular signal molecule cAMP.
  • GPCR membrane- bound G-protein coupled receptors
  • the expression of the receptor was detected on peripheral nerve endings of nociceptors, on macrophages and neutrophils. For these cell types, great importance was demonstrated in connection with inflammatory pain. It is assumed that the local inflammation of the endometriotic lesions makes a significant contribution to the genesis of the pain symptoms observed (Stratton & Berkley 2010; Giudice 2010).
  • EP4 antagonists of different structural classes have been described, Thus in WO2005/0121508, WO2005102389 and WO2005/105733 (Pfizer), for example, N-benzylarylamides, N- benzylheteroarylamides and [(1 H-benzimidazol-1-yl)phenylethyl]aryl- and [(1 H-benzimidazol- 1 -yl)phenylethyl]heteroarylsulphonylcarbamates are described for use in the case of pain, inflammation, osteoarthritis and rheumatoid arthritis.
  • Pfizer also describes in WO2002032422, WO2002032900 and WO2003086371 structures that include generic benzimidazoles.
  • Thiophene-A/-benzylamides in WO2008017164 and WO2009020588, indole-N-benzylamides in WO2007121578 and N- ⁇ [(6,8-dihydro-7H-pyrrolo[3,4-g]quinolin-7- yl)-aryl]methyl ⁇ sulphonylamides in WO2008104055 are addressed for nearly the same indication spectrum by Merck-Frosst.
  • WO2004067524 Pulsagene Laboratories
  • furan derivatives for the treatment of headache and migraine are described.
  • EP2172447 (Astellas Pharma) claims generically in a very broad manner compounds that can consist of two heterocycles connected directly to one another, for the indications renal insufficiency and diabetic nephropathy.
  • the underlying problem of the present invention therefore lies in the provision of medication for treatment of inflammatory pain and/or other pathological conditions related to increased activation of EP4 receptors by prostaglandin E2 (PGE2).
  • PGE2 prostaglandin E2
  • the compound showed after p.o. administration robust effects in in vivo studies in mice and rats.
  • the compound attenuated inflammatory pain and edema formation in various settings.
  • the compound can be employed for long term treatment (fig. 5).
  • the suppression of PGE2 induced proliferation of uterine tissue was shown in vitro and in vivo in mice. Therefore the use of compound 1 for the treatment of inflammatory pain and/or other pathological conditions related to increased activation of EP4 receptors by prostaglandin E2 (PGE2) provides a solution for the underliying problem of the invention.
  • PGE2 prostaglandin E2
  • This figure shows the binding charactistics of compound 1 .
  • Experiments were performed to show that the binding is antagonistic (Part a), agonistic (part B) or inverse agonistic (Part C).
  • Part a antagonistic
  • Part B agonistic
  • Part C inverse agonistic
  • the results show that compound 1 is a highly potent, competitive human EP4 receptor antagonist with inverse agonist activity.
  • Figure 2 Effect of Compound 1 on CFA induced pain.
  • Compound 1 was given in increasing concentrations ranging from 0.2 to 25 mg/kg. The figure clearly shows that compound 1 reduced inflammatory pain significantly and dose-dependently ( * p ⁇ 0.05, ** p ⁇ 0.01 , *** p ⁇ 0.005, **** p ⁇ 0.001 , ANOVA followed by post hoc test (Dunnett) vs vehicle).
  • Compound 1 was given in following concentrations: 1 mg/kg, 5 mg/kg and 25 mg/kg.
  • Figure 4 shows that compound 1 significantly reduced PGE2 induced pain ( * p ⁇ 0.01 vs vehicle, analysed by 1 -way ANOVA followed by Dunnett ' s post hoc test).
  • Figure 5 represents the effect of compound 1 on cell proliferation.
  • Compound 1 100 nM was given with (crossed bar) or without (vertical hatched bar) 10 nM PGE2, PGE2 was also given alone (horizontal hatched bar).
  • PGE2 was also given alone (horizontal hatched bar).
  • the total cell number did not increase when compared with the group which contained PGE2 alone.
  • Compound 1 together with PGE2 also did not affect the total cell number.
  • Figure 6 demonstrates that stabilized PGE2, applied directly in the cavity of the uterus, induced proliferation in uterine tissue in aenesthetized mice.
  • Compound 1 was given in concentrations ranging from 0.2 to 25 mg/kg; compound 1 dose-dependently suppressed PGE2-induced cell number increase in uterine tissue ( * p ⁇ 0.05 and ** p ⁇ 0.01 compared to 2 mg/kg dm-PGE2 group, 3 outliers following Grubbs test removed, analysed by 1 -way ANOVA followed by Dunnett's post hoc test).
  • Figure 7 Effect of compound 1 in endomet iosis dyspareunia model (rat, vaginal distension
  • Figure 7 shows the effect of compound 1 in an endometriosis dyspareunia model. Open bars represent the vehicle group, hatched bars represent the compound group, and crossed bars show the effect of ibuprofen.
  • Compound 1 decreased vaginal hyperalgesia in a rat endometriosis model. + stands for: cumulative number of contraction per 0.1 ml distension and ++ for: different vehicle, these were excluded from statistical analysis
  • the present invention is based on the discovery that compound 1 is highly potent in the treatment of inflammatory pain and/or other pathological conditions related to increased activation of EP4 receptors by prostaglandin E2 (PGE2). Therefore subject matter of the present invention is directed to the use of compound 1 for treatment of inflammatory pain and/or other pathological conditions related to increased activation of EP4 receptors by prostaglandin E2 (PGE2).
  • Inflammatory pain inflammatory pain is selected from a group comprising but not limited to ankylosing spondylitis, or pain associated with endometriosis, post operative pain, pain related to skin burn injuries and osteoarthritis.
  • the therapeutically active dose is dependent on the body weight, administration route, individual behaviour, and the type of preparation and time or interval at which administration takes place.
  • a typical dose range for a woman of 70 kg body weight is between 1-500 mg/day, preferably between 5 and 30 mg/day.
  • a further subject of the present invention relates to medicaments containing compound 1 according to the invention and at least one or more other active substances, in particular for the treatment and/or prophylaxis of endometriosis.
  • Suitable combination active substances that may be mentioned by way of example and preferably are: selective oestrogen receptor modulators (SERMs), oestrogen receptor (ER) antagonists, aromatase inhibitors, 17 ⁇ -HSD1 inhibitors, steroid sulphatase (STS) inhibitors, GnRH agonists and antagonists, kisspeptin receptor (KISSR) antagonists, selective androgen receptor modulators (SAR s), androgens, 5 ⁇ -reductase inhibitors, selective progesterone receptor modulators (SPRMs), gestagens, antigestagens, oral contraceptives, inhibitors of mitogen activated protein (MAP) kinase and inhibitors of MAP kinases (Mkk3/6, Mek1/2, Erk1/2), inhibitor
  • Compound 1 can be employed for the treatment of inflammatory pain, both after oral as well as parenteral administration.
  • Inflammatory pain includes forms of acute and chronic pain, including chronic pelvic pain, endometriosis associated pain, as well as pain associated with angina, or pain of varied origins (including but not limited to pain associated with osteoarthritis, rheumatoid arthritis, rheumatic disease, tenosynovitis, gout, bursitis, post operative pain, pain related to skin burn injuries, and ankylosing spondylitis. Therefore subject matter of the present invention is also the use of compound 1 for the treatment of inflammatory pain, whereby the inflammatory pain is selected from a group comprising but not limited to ankylosing spondylitis, or pain associated with endometriosis, post operative pain, pain related to skin burn injuries and osteoarthritis
  • Compound 1 according to the invention can act systemically and/or locally.
  • it can be administered in a suitable manner, such as e.g. orally, parenterally, pulmonarily, nasally, sublingually, lingually, buccally, rectally, dermally, transdermally, conjunctivally, otically or as an implant or stent.
  • the compound according to the invention can be administered in suitable administration forms.
  • the dosage of compound 1 in these preparations should be 0.01 % - 20 %, in order to achieve an adequate pharmacological action.
  • the dosage of the active substances can vary, depending on administration route, age and weight of the patient, nature and severity of the disease to be treated and similar factors.
  • the treatment can be carried out by means of individual doses or by a plurality of doses over a relatively long period, between 1 -500 mg/day, preferably between 5 and 30 mg/day, where the dose can be given as an individual dose to be administered once or subdivided into 2 or more daily doses.
  • surface-active excipients such as salts of the bile acids or animal or vegetable phospholipids can also be used, but also mixtures thereof and liposomes or their constituents.
  • administration forms functioning according to the prior art, releasing compound 1 to be used according to the invention rapidly and/or in modified form, which contain compound 1 according to the invention in crystalline and/or amorphized and/or dissolved form, are suitable, such as, e.g., tablets (non-coated or coated tablets, for example, having enteric or slowly dissolving or insoluble coatings, which control the release of the compound to be used according to the invention), tablets or films/wafers disintegrating rapidly in the oral cavity, films/lyophilizates, capsules (for example, hard or soft gelatine capsules), coated tablets, granules, pellets, powders, emulsions, suspensions, aerosols or solutions.
  • tablets non-coated or coated tablets, for example, having enteric or slowly dissolving or insoluble coatings, which control the release of the compound to be used according to the invention
  • tablets or films/wafers disintegrating rapidly in the oral cavity, films/lyophilizates, capsules (for example, hard or soft gelatin
  • Parenteral administration can take place circumventing an absorption step (e.g. intravenously, intraarterially, intracardially, intraspinaliy or intralumbarly) or with inclusion of an absorption (e.g. intramuscularly, subcutaneously, intracutaneously, percutaneously or intraperitoneally).
  • an absorption step e.g. intravenously, intraarterially, intracardially, intraspinaliy or intralumbarly
  • an absorption e.g. intramuscularly, subcutaneously, intracutaneously, percutaneously or intraperitoneally.
  • suitable administration forms are, inter alia, injection and infusion preparations in the form of solutions, suspensions, emulsions, lyophilizates or sterile powders.
  • compositions for inhalation e.g., pharmaceutical forms for inhalation (inter alia powder inhalers, nebulizers), nose drops, solutions or sprays, tablets to be administered lingually, sublingually or buccally, films/wafers or capsules, suppositories, ear or eye preparations, tinctures, vaginal capsules and suppositories, tampons, intrauterine pessaries, aqueous suspensions (lotions, shaking mixtures), lipophilic suspensions, crystal suspensions, aqueous and oily injection solutions, depot preparations, ointments, fatty ointments, gels, creams, transdermal therapeutic systems (e.g. patches), milk, pastes, foams, dusting powders, implants, intrauterine spirals, vaginal rings or stents are suitable.
  • pharmaceutical forms for inhalation inter alia powder inhalers, nebulizers
  • nose drops solutions or sprays
  • Oral or parenteral administration is preferred, in particular oral and intravenous administration.
  • Topic application is also an option.
  • Compound 1 to be used according to the invention can be converted into the administration forms mentioned. This can take place in a manner known per se by mixing with inert, nontoxic, pharmaceutically suitable excipients.
  • excipients include, inter alia, carrier substances (for example, microcrystalline cellulose, lactose, mannitol), solvents (e.g. liquid polyethylene glycols), emulsifiers and dispersants or wetting agents (for example sodium dodecylsulphate, polyoxysorbitan oleate), binding agents (for example polyvinylpyrrolidone), synthetic and natural polymers (for example albumin), stabilizers (e.g. antioxidants such as, for example, ascorbic acid), colourants (e.g. inorganic pigments such as, for example, iron oxides) and taste and/or odour corrigents.
  • carrier substances for example, microcrystalline cellulose, lactose, mannitol
  • solvents e.g. liquid polyethylene glycols
  • a further subject of the present invention is a medicament that contains compound 1 according to the invention, customarily together with one or more inert, non-toxic, pharmaceutically suitable excipients, and their use for the previously mentioned purposes.
  • the amount per day is approximately 0.01 to 100 mg/kg of body weight.
  • the amount of compound 1 to be administered varies within a wide range and can cover any effective amount.
  • a typical dose range for a woman of 70 kg body weight is between 1 - 500 mg/day, preferably between 5 and 30 mg/dayNevertheless, it can optionally be necessary to deviate from the amounts mentioned, namely, depending on body weight, administration route, individual behaviour towards the active substance, nature of the preparation and time or interval at which administration takes place. Thus, in some cases it can be adequate to manage with less than the aforementioned minimum amount, while in other cases the upper limit mentioned must be exceeded. In the case of the administration of relatively large amounts, it can be advisable to divide these into a number of individual doses over the course of the day.
  • the subject matter of the present invention is also directed to a method for alleviating pathological conditions associated with PGE2 induced increased EP4 receptors activation by antagonising the activation of EP4 receptors by compound 1.
  • Binding of PGE2 to the Gas coupled human EP4-receptor leads to the stimualtion of adenylate cyclase activity and the formation of cAMP.
  • the amount of cAMP produced following agonist stimulation is detected with the help of a competition assay based on a fluorescently labelled cAMP tracer (cAMP-d2) and an Eu-cryptate labelled anti-CAMP antibody.
  • Maximum signal obtained via fluorescence resonance energy transfer; FRET
  • FRET fluorescence resonance energy transfer
  • An appropriate plate reader (RubyStar, PheraStar, ViewLux) is used to induce and measure FRET emissions at 620 and 665nm following excitation at 337 nm. Any signal-decrease shows activation of the GPCR (i.e. change in well-Ratio; defined as 665nm / 620nm * 10000). Antagonists result in signal increase.
  • Table 1 represents the steps of assay ready cells from frozen cell stocks.
  • hEP4-R stably transfected into HEK cells (clone hEP4-C1 cells), Read plates: Poiystyroi; 384well, SV white, Greiner # 784075, Dimethylsulphoxide, DMSO: Sigma-Aldrich # D-2650, 3-lsobutyl-1-methylxanthine, IBMX: Sigma-Aldrich # 1-7018, cAMP HTRF-Assay Kit: Cisbio International 62AM6PEJ high range
  • Step 1 Reconstitution of both, cAMP-d2 & anti-CAMP cryptate with water (5ml_) according to suppliers manual.
  • Step 2 Further 1 :39 dilution with Conjugate & Lysis Buffer.
  • Conjugate & Lysisbuffer prepared according to cAMP kit protocol (Cisbio).
  • CFA Complete Freund's Adjuvant
  • Endpoint 1 Dynamic weight bearing (DWB)
  • the automated DWB device (Bioseb, Boulogne, France) consists of a Plexiglas enclosure (22x22x30cm), a floor sensor composed of captors in order to detect pressure variation and a camera placed above the enclosure in order to validate the animal position during analysis (Robinson et al., 2012; Tetreault et al., 201 1 ).
  • the rats were able to move freely in the enclosure during 5 min while the acquisition data were transmitted to a computer.
  • the animals were not acclimated to the enclosure before the test.
  • Endpoint 2 Paw weight analysis (edema)
  • the paw was removed from the dead animal at the ankle joint and weight of the paw was noted.
  • Figure 4 shows that PGE2 induced pain is mediated by EP4 receptors.
  • Compound 1 significantly reduced PGE2 induced pain dose dependendly.
  • Cells (Cell culture: EM42 cells (Desai et al., 1994, Fert. Steril. 61 : 760) were cultivated one passage before experiment in test media (DMEM w/o phenolred, 1 % Pen/strep, 1 % L- glutamine plus indicated concentration of FCS [e.g. :1 %])
  • Indicated number of cells (e.g. 1000) was plated in wells of 96well plates (Perkin Elmer #6005688). Substances were added as indicated in tetraplicates, inhibitors were added first. PGE2 was given twice daily. For incubation without PGE2 the same amount of media was added twice daily. Incubation of MTPs for 4 days at 37°C and 5% CO? was followed by a CellTiter Glo measurement at day 4.
  • Preparation of cell number calibration curve from cells in test media Cells were counted for calibration and plated in duplicates in rows 1 and 2 and incubated for two hours. Celltiter Glo measurement was performed as described by vendor (Promega #G7571 ). Briefly, MTPs were adjusted to room temperature for 30 minutes. 100 ⁇ ! celltiter reagent was added, followed by mixing and incubation for 10 minutes. The luminescence was measured of with POLARstar (bmg). Measurements of relative light units (RLU) and calculation of cell number equivalent was performed by employing linear part of calibration curve.
  • RLU relative light units
  • EXAMPLE 6 Effect of compound 1 in endometriosis dyspareunia model (rat, vaginal distension
  • VMR viscero motor response
  • VD vaginal distension
  • Compound 1 decreased vaginal hyperalgesia versus vehicle-treated animals. The effect maintained in the treatment free period of week 6. Therefore compound 1 is suitable for long term treatment of the aforementioned diseases. This might point to a disease modyfing component induced by the EP4 receptor antagonism exceeding pure analgesic activity. Interestingly, daily dosed compound 1 showed a more pronounced effect than acute treatment with one analgesic dose of ibuprofenoilected to measure the VMR related to the distension.

Abstract

The present invention relates the use of compound 1 for the treatment of inflammatory pain.

Description

Use of an EP4 antagonist for the treatment of inflammatory pain
The present invention relates to the use of an EP4 antagonist for the treatment and/or prophylaxis of inflammatory pain and/or other pathological conditions related to increased activation of EP4 receptors by prostaglandin E2 (PGE2).
BACKGROUND OF THE INVENTION
According to Basbaum (Basbaum et al (2009). Cell 139, 267-284) inflammatory pain is one form of nociceptive pain which is typically accompanied by an immune response and mediated by pro-inflammatory molecules which are released by tissue due to a trigger which endangers or destroys cell integrity or signals endangered cell integrity. These conditions lead to accumulation of endogenous factors released from activated nociceptors (peripheral nerve endings of the pain pathway) or non-neural cells that reside within or infiltrate into the injured/endangered area (including mast cells, basophils, platelets, macrophages, neutrophils, endothelial cells, keratinocytes, and fibroblasts). Collectively, these factors, referred to as the "inflammatory soup," represent a wide array of signaling molecules, including neurotransmitters, peptides (substance P, CGRP, bradykinin), eicosinoids and related lipids like prostaglandins which include the endogenous activators of the EP4 receptor, thromboxanes, leukotrienes, endocannabinoids, neurotrophins, cytokines, and chemokines, as well as extracellular proteases and protons. Nociceptors express one or more cell-surface receptors capable of recognizing and responding to each of these pro- inflammatory or pro-algesic agents. Such interactions enhance excitability of the nerve fiber, thereby heightening its sensitivity to temperature or touch leading finally to hyperalgesia. Ongoing or frequently occurring inflammatory events lead in many cases to chronic inflammatory pain which severely affecting the quality of life of patients. Inflammatory pain of varied origins is associated but not limited to osteoarthritis, rheumatoid arthritis, rheumatic disease, tenosynovitis, gout, ankylosing spondylitis, endometriosis, injuries, various skin diseases and bursitis.
Currently inflammatory pain can be treated with nonsteroidal anti-inflammatory drugs (NSAID). However, due to the risk of side-effects under long term treatment this treatment option exists only for a short term treatment employing doses as low as possible. Combining all available treatment modalities, recent data confirm a significant degree of unmet medical need since up to 70% of treated patients have persistent symptoms like chronic pelvic pain that are not managed (Bayer market research, 2009, 21.700 women in 8 countries). Therefore, new and long-term treatment options with reduced side-effects and high efficacy are urgently needed. The receptor EP4 (PTGR4) is one of the 4 human receptors that are activated by endogenously formed prostaglandin E2 (PGE2). EP4 belongs to the family of membrane- bound G-protein coupled receptors (GPCR) and is mainly provided with a Gs coupling, which after activation leads to an accumulation of the intracellular signal molecule cAMP. The expression of the receptor was detected on peripheral nerve endings of nociceptors, on macrophages and neutrophils. For these cell types, great importance was demonstrated in connection with inflammatory pain. It is assumed that the local inflammation of the endometriotic lesions makes a significant contribution to the genesis of the pain symptoms observed (Stratton & Berkley 2010; Giudice 2010).
Up to now, no EP4 antagonist has been licensed as a medicament. However, EP4 antagonists of different structural classes have been described, Thus in WO2005/0121508, WO2005102389 and WO2005/105733 (Pfizer), for example, N-benzylarylamides, N- benzylheteroarylamides and [(1 H-benzimidazol-1-yl)phenylethyl]aryl- and [(1 H-benzimidazol- 1 -yl)phenylethyl]heteroarylsulphonylcarbamates are described for use in the case of pain, inflammation, osteoarthritis and rheumatoid arthritis. Pfizer also describes in WO2002032422, WO2002032900 and WO2003086371 structures that include generic benzimidazoles. Thiophene-A/-benzylamides in WO2008017164 and WO2009020588, indole-N-benzylamides in WO2007121578 and N-{[(6,8-dihydro-7H-pyrrolo[3,4-g]quinolin-7- yl)-aryl]methyl}sulphonylamides in WO2008104055 are addressed for nearly the same indication spectrum by Merck-Frosst. In WO2004067524 (Pharmagene Laboratories), furan derivatives for the treatment of headache and migraine are described.
EP2172447 (Astellas Pharma) claims generically in a very broad manner compounds that can consist of two heterocycles connected directly to one another, for the indications renal insufficiency and diabetic nephropathy.
In WO2014/086739 (Bayer Pharma) novel benzimidazole-5-carboxylic acid derivatives and their use for treatment of endometriosis are disclosed. The disclosure of this application is incorporated herewith in its entirety.
As mentioned above there is an urgent need for medicaments which are effective in the treatment of inflammatory pain and/or other pathological conditions related to increased activation of EP4 receptors by prostaglandin E2 (PGE2), which do not have the disadvantages of the prior art.
The underlying problem of the present invention therefore lies in the provision of medication for treatment of inflammatory pain and/or other pathological conditions related to increased activation of EP4 receptors by prostaglandin E2 (PGE2). SUMMARY OF THE INVENTION
It was found that compound 1 (2-(9-Ethyl-6-methyl-9H-carbazol-3-yl)-1-(2-methoxyethyl)-4- methyl-1 H-benzimidazole-5-carboxylic acid)
Figure imgf000004_0001
Compound 1
was highly active in several models of pain. It could be demonstrated that compound 1 was identified as a highly potent, competitive human EP4 receptor antagonist with inverse agonist activity. Secondary in vitro assays revealed a high functional potency on EP4 receptors of mouse, rat and cynomolgus.
The compound showed after p.o. administration robust effects in in vivo studies in mice and rats. The compound attenuated inflammatory pain and edema formation in various settings. The compound can be employed for long term treatment (fig. 5). The suppression of PGE2 induced proliferation of uterine tissue was shown in vitro and in vivo in mice. Therefore the use of compound 1 for the treatment of inflammatory pain and/or other pathological conditions related to increased activation of EP4 receptors by prostaglandin E2 (PGE2) provides a solution for the underliying problem of the invention.
DESCRIPTION OF THE FIGURES Figure 1 : Characterization of binding
This figure shows the binding charactistics of compound 1 . Experiments were performed to show that the binding is antagonistic (Part a), agonistic (part B) or inverse agonistic (Part C). The results show that compound 1 is a highly potent, competitive human EP4 receptor antagonist with inverse agonist activity.
Figure 2: Effect of Compound 1 on CFA induced pain.
Compound 1 was given in increasing concentrations ranging from 0.2 to 25 mg/kg. The figure clearly shows that compound 1 reduced inflammatory pain significantly and dose- dependently (*p<0.05, **p<0.01 , ***p<0.005, ****p<0.001 , ANOVA followed by post hoc test (Dunnett) vs vehicle).
Figure 3: Effect of Compound 1 on CFA induced edema
Compound 1 was given in increasing concentrations ranging from 0.2 to 25 mg/kg. The figure clearly shows that compound 1 reduced edema significantly and dose-dependently (**p<0.01 and ****p<0.001 , ANOVA followed by post hoc test (Dunnett) vs vehicle).
Figure 4: Effect of Compound 1 on PGE2 induced pain
Compound 1 was given in following concentrations: 1 mg/kg, 5 mg/kg and 25 mg/kg. Figure 4 shows that compound 1 significantly reduced PGE2 induced pain (*p<0.01 vs vehicle, analysed by 1 -way ANOVA followed by Dunnett's post hoc test).
Figure 5: Effect of Compound 1 on PGE2 induced proliferation in vitro
Figure 5 represents the effect of compound 1 on cell proliferation. Compound 1 (100 nM) was given with (crossed bar) or without (vertical hatched bar) 10 nM PGE2, PGE2 was also given alone (horizontal hatched bar). In the presence of compound 1 the total cell number did not increase when compared with the group which contained PGE2 alone. Compound 1 together with PGE2 also did not affect the total cell number.
Figure 6: Effect of Compound 1 on PGE2 induced proliferation
Figure 6 demonstrates that stabilized PGE2, applied directly in the cavity of the uterus, induced proliferation in uterine tissue in aenesthetized mice. Compound 1 was given in concentrations ranging from 0.2 to 25 mg/kg; compound 1 dose-dependently suppressed PGE2-induced cell number increase in uterine tissue (*p<0.05 and **p<0.01 compared to 2 mg/kg dm-PGE2 group, 3 outliers following Grubbs test removed, analysed by 1 -way ANOVA followed by Dunnett's post hoc test). Figure 7: Effect of compound 1 in endomet iosis dyspareunia model (rat, vaginal distension
Figure 7 shows the effect of compound 1 in an endometriosis dyspareunia model. Open bars represent the vehicle group, hatched bars represent the compound group, and crossed bars show the effect of ibuprofen. Compound 1 decreased vaginal hyperalgesia in a rat endometriosis model. + stands for: cumulative number of contraction per 0.1 ml distension and ++ for: different vehicle, these were excluded from statistical analysis
**p<0.01 *p<0.05 t-test vehicle vs compound 1.
DETAILED DESCRIPTION OF THE INVENTION
The present invention is based on the discovery that compound 1 is highly potent in the treatment of inflammatory pain and/or other pathological conditions related to increased activation of EP4 receptors by prostaglandin E2 (PGE2). Therefore subject matter of the present invention is directed to the use of compound 1 for treatment of inflammatory pain and/or other pathological conditions related to increased activation of EP4 receptors by prostaglandin E2 (PGE2). Inflammatory pain inflammatory pain is selected from a group comprising but not limited to ankylosing spondylitis, or pain associated with endometriosis, post operative pain, pain related to skin burn injuries and osteoarthritis.
DEFINITION
The therapeutically active dose is dependent on the body weight, administration route, individual behaviour, and the type of preparation and time or interval at which administration takes place. A typical dose range for a woman of 70 kg body weight is between 1-500 mg/day, preferably between 5 and 30 mg/day.
A further subject of the present invention relates to medicaments containing compound 1 according to the invention and at least one or more other active substances, in particular for the treatment and/or prophylaxis of endometriosis. Suitable combination active substances that may be mentioned by way of example and preferably are: selective oestrogen receptor modulators (SERMs), oestrogen receptor (ER) antagonists, aromatase inhibitors, 17· -HSD1 inhibitors, steroid sulphatase (STS) inhibitors, GnRH agonists and antagonists, kisspeptin receptor (KISSR) antagonists, selective androgen receptor modulators (SAR s), androgens, 5· -reductase inhibitors, selective progesterone receptor modulators (SPRMs), gestagens, antigestagens, oral contraceptives, inhibitors of mitogen activated protein (MAP) kinase and inhibitors of MAP kinases (Mkk3/6, Mek1/2, Erk1/2), inhibitors of protein kinases B (ΡΚΒ· /· /· ; Akt1 /2/3), inhibitors of phosphoinositide-3 kinase (PI3K), inhibitors of cyclin- dependent kinase (CDK1/2), Inhibitors of the hypoxia-induced signal pathway (HIF1 alpha inhibitors, activators of the prolylhydroxylases), histone deacetylase (HDAC) inhibitors, prostaglandin F receptor (FP) (PTGFR) antagonists, neurokinin 1 receptor antagonists, paracetamol, selective COX2 inhibitors and/or non-selective COX1/COX2 inhibitors.
Compound 1 can be employed for the treatment of inflammatory pain, both after oral as well as parenteral administration.
Inflammatory pain includes forms of acute and chronic pain, including chronic pelvic pain, endometriosis associated pain, as well as pain associated with angina, or pain of varied origins (including but not limited to pain associated with osteoarthritis, rheumatoid arthritis, rheumatic disease, tenosynovitis, gout, bursitis, post operative pain, pain related to skin burn injuries, and ankylosing spondylitis. Therefore subject matter of the present invention is also the use of compound 1 for the treatment of inflammatory pain, whereby the inflammatory pain is selected from a group comprising but not limited to ankylosing spondylitis, or pain associated with endometriosis, post operative pain, pain related to skin burn injuries and osteoarthritis
Compound 1 according to the invention can act systemically and/or locally. For this purpose, it can be administered in a suitable manner, such as e.g. orally, parenterally, pulmonarily, nasally, sublingually, lingually, buccally, rectally, dermally, transdermally, conjunctivally, otically or as an implant or stent.
For these administration routes, the compound according to the invention can be administered in suitable administration forms.
The dosage of compound 1 in these preparations should be 0.01 % - 20 %, in order to achieve an adequate pharmacological action.
The dosage of the active substances can vary, depending on administration route, age and weight of the patient, nature and severity of the disease to be treated and similar factors. The treatment can be carried out by means of individual doses or by a plurality of doses over a relatively long period, between 1 -500 mg/day, preferably between 5 and 30 mg/day, where the dose can be given as an individual dose to be administered once or subdivided into 2 or more daily doses.
As carrier systems, surface-active excipients such as salts of the bile acids or animal or vegetable phospholipids can also be used, but also mixtures thereof and liposomes or their constituents.
The formulations and administration forms described above are likewise the subject of the present invention.
If, in addition to the compound according to the invention, further active substances are contained, these can be formulated in a common administration form or optionally also administered as a combination preparation.
For oral administration, administration forms functioning according to the prior art, releasing compound 1 to be used according to the invention rapidly and/or in modified form, which contain compound 1 according to the invention in crystalline and/or amorphized and/or dissolved form, are suitable, such as, e.g., tablets (non-coated or coated tablets, for example, having enteric or slowly dissolving or insoluble coatings, which control the release of the compound to be used according to the invention), tablets or films/wafers disintegrating rapidly in the oral cavity, films/lyophilizates, capsules (for example, hard or soft gelatine capsules), coated tablets, granules, pellets, powders, emulsions, suspensions, aerosols or solutions.
Parenteral administration can take place circumventing an absorption step (e.g. intravenously, intraarterially, intracardially, intraspinaliy or intralumbarly) or with inclusion of an absorption (e.g. intramuscularly, subcutaneously, intracutaneously, percutaneously or intraperitoneally). For parenteral administration, suitable administration forms are, inter alia, injection and infusion preparations in the form of solutions, suspensions, emulsions, lyophilizates or sterile powders.
For the other administration routes, e.g., pharmaceutical forms for inhalation (inter alia powder inhalers, nebulizers), nose drops, solutions or sprays, tablets to be administered lingually, sublingually or buccally, films/wafers or capsules, suppositories, ear or eye preparations, tinctures, vaginal capsules and suppositories, tampons, intrauterine pessaries, aqueous suspensions (lotions, shaking mixtures), lipophilic suspensions, crystal suspensions, aqueous and oily injection solutions, depot preparations, ointments, fatty ointments, gels, creams, transdermal therapeutic systems (e.g. patches), milk, pastes, foams, dusting powders, implants, intrauterine spirals, vaginal rings or stents are suitable.
Oral or parenteral administration is preferred, in particular oral and intravenous administration. Topic application is also an option.
Compound 1 to be used according to the invention can be converted into the administration forms mentioned. This can take place in a manner known per se by mixing with inert, nontoxic, pharmaceutically suitable excipients. These excipients include, inter alia, carrier substances (for example, microcrystalline cellulose, lactose, mannitol), solvents (e.g. liquid polyethylene glycols), emulsifiers and dispersants or wetting agents (for example sodium dodecylsulphate, polyoxysorbitan oleate), binding agents (for example polyvinylpyrrolidone), synthetic and natural polymers (for example albumin), stabilizers (e.g. antioxidants such as, for example, ascorbic acid), colourants (e.g. inorganic pigments such as, for example, iron oxides) and taste and/or odour corrigents.
A further subject of the present invention is a medicament that contains compound 1 according to the invention, customarily together with one or more inert, non-toxic, pharmaceutically suitable excipients, and their use for the previously mentioned purposes.
In the case of oral administration, the amount per day is approximately 0.01 to 100 mg/kg of body weight. The amount of compound 1 to be administered varies within a wide range and can cover any effective amount. A typical dose range for a woman of 70 kg body weight is between 1 - 500 mg/day, preferably between 5 and 30 mg/dayNevertheless, it can optionally be necessary to deviate from the amounts mentioned, namely, depending on body weight, administration route, individual behaviour towards the active substance, nature of the preparation and time or interval at which administration takes place. Thus, in some cases it can be adequate to manage with less than the aforementioned minimum amount, while in other cases the upper limit mentioned must be exceeded. In the case of the administration of relatively large amounts, it can be advisable to divide these into a number of individual doses over the course of the day.
The percentages in the following tests and examples are, if not stated otherwise, percentages by weight; parts are parts by weight. Solvent ratios, dilution ratios and concentrations of liquid/liquid solutions in each case relate to the volume.
The subject matter of the present invention is also directed to a method for alleviating pathological conditions associated with PGE2 induced increased EP4 receptors activation by antagonising the activation of EP4 receptors by compound 1.
EXAMPLES
EXAMPLE 1 : Synthesis of compound 1 :
The synthesis of the intermediates as well as of compound 1 is disclosed in WO2014/086739 (example 141 ). The disclosure of this application is incorporated herewith in its entirety Briefly, in analogy to Example 4/Variant B of WO2014/086739, methyl 2-(9-ethyl-6-methyl- 9H-carbazol-3-yl)-1 -(2-methoxyethyl)-4-methyl-1 H-benzimidazole-5-carboxylate was first prepared from 1.0 g (4.2 mmol) of methyl 3-amino-4-[(2-methoxyethyl)amino]-2- methylbenzoate and 1.0 g (4.2 mmol) of 9-ethyl-6-methyl-9H-carbazole-3-carbaldehyde (CAS No 122060-05-3).
1 H-NMR (400 MHz, DMSO-d6), · [ppm] = 1 .35 (t, 3H), 2.50 (s, 3H), 2.89 (s, 3H), 3.12 (s, 3H), 3.69 (t, 2H), 3.87 (s, 3H), 4.49 (q, 2H), 4.55 (t, 2H), 7.35 (dd, 1 H), 7.58 (t, 2H), 7.75 (d, 1 H), 7.83 (d, 1 H), 7.90 (dd, 1 H), 8.06 (s, 1 H), 8.59 (d, 1 H).
This was subsequently reacted analogously to Example 2 of WO2014/086739 to give 2-(9- ethyl-6-methyl-9H-carbazol-3-yl)-1 -(2-methoxyethyl)-4-methyl-1 H-benzimidazole-5-carboxylic acid. (920 mg, 50%).
1 H-NMR (300 MHz, DMSO-d6), · [ppm] = 1 .35 (t, 3H), 2.49 (s, 3H), 2.89 (s, 3H), 3.12 (s, 3H), 3.69 (t, 2H), 4.43 - 4.59 (m, 4H), 7.34 (dd, 1 H), 7.56 (m, 2H), 7.75 (d, 1 H), 7.81 - 7.93 (m, 2H), 8.07 (s, 1 H), 8.59 (d, 1 H), 12.25 - 12.95 (br., 1 H).
EXAMPLE 2: Binding experiments to determine mode of action
Model description I:
A. Principle of detection:
Binding of PGE2 to the Gas coupled human EP4-receptor leads to the stimualtion of adenylate cyclase activity and the formation of cAMP. The amount of cAMP produced following agonist stimulation is detected with the help of a competition assay based on a fluorescently labelled cAMP tracer (cAMP-d2) and an Eu-cryptate labelled anti-CAMP antibody. Maximum signal (obtained via fluorescence resonance energy transfer; FRET) is obtained in the absence of cellular cAMP. Upon cell lysis, any cAMP produced (and accumulated in the presence of IBMX) competes for binding of the tracer to the antibody. An appropriate plate reader (RubyStar, PheraStar, ViewLux) is used to induce and measure FRET emissions at 620 and 665nm following excitation at 337 nm. Any signal-decrease shows activation of the GPCR (i.e. change in well-Ratio; defined as 665nm / 620nm*10000). Antagonists result in signal increase.
B. Standard antagonist assay protocol: Four microliters of a cell suspension containing 625 cells/μΙ (2500 cells/well; by this time also containing the cAMP-D2 tracer) are added to a testplate already containing the test compounds (50 nl; 100 % DMSO). Following a 20 minute pre-incubation at room temperature two microliters of a 3XEC80 PGE2 agonist solution are added per well and the plate is incubated in the presence of a 1XEC80 concentration of agonist for 60 minutes (total volume: 6 μΙ). Finally the whole reaction is stopped via the addition of 2 μΙ of lysisbuffer also containing the Eu-cryptate labelled anti-CAMP antibody (Final volume: 8 μΙ). Another 20 minutes later the cell lysate containing plate is transferred to TR-FRET compatible reader in order to quantify the results (see model description II for a more detailed description of all generic steps involved). (See figure 1 A)
C. Standard assay protocol:
Four microliters of a cell suspension containing 625 cells/μΙ (2500 cells/well; by this time also containing the cAMP-D2 tracer) are added to a testplate already containing the test compounds (50 nl; 100 % DMSO). Following a 20 minute preincubation at room temperature two microliters of cell medium solution are added per well and the plate incubated for another 60 minutes (total volume: 6 μΙ). Finally the whole reaction is stopped via the addition of 2 μΙ of lysisbuffer also containing the Eu-cryptate labelled anti-CAMP antibody (Final volume: 8 μΙ). Another 20 minutes later the cell lysate containing plate is transferred to TR-FRET compatible reader in order to quantify the results (see model description II for a more detailed description of all generic steps involved). (See figure 1 B).
Model description II:
Test for Inverse Agonism: STANDARD ASSAY PROTOCOL
(10 000 cells/well - Target Details: PTGER4, NM_00958; NP_00949).
1 . Principle of detection:
Constitutive hEP4-R activity results the formation of cAMP even in the absence of agonist and these basal cAMP levels are detected when using 10 000 hEP4-R positive HEK293 cells per well. Upon cell lysis, any cAMP produced by now competes with a fluorescent cAMP tracer (cAMP- d2) for binding to an Europium labelled anti-cAMP antibody. Thus, a maximum fluorescence resonance energy transfer (FRET) signal is obtained in the absence of cellular cAMP. Following excitation at 337 nm any reduction of the FRET induced emission of the cAMP-d2 tracer at 665nm is indicative of cAMP production. A second FRET induced emission at 620nm (resulting from the Eu-labelled anti cAMP antibody) is used for well internal referencing. As inverse agonists inhibit basal receptor activity they should result in a signal increase. (Measurement instrument: RubiStar; read-out: well-Ratio defined as 665nm / 620nm * 10000). (see figure 1 C) 2. Preparation of assay ready cells from frozen cells stocks:
Table 1 represents the steps of assay ready cells from frozen cell stocks.
Figure imgf000013_0001
3. Materials:
Cells: hEP4-R stably transfected into HEK cells (clone hEP4-C1 cells), Read plates: Poiystyroi; 384well, SV white, Greiner # 784075, Dimethylsulphoxide, DMSO: Sigma-Aldrich # D-2650, 3-lsobutyl-1-methylxanthine, IBMX: Sigma-Aldrich # 1-7018, cAMP HTRF-Assay Kit: Cisbio International 62AM6PEJ high range
3.1 . Preparation of cAMP-HTRF detection reagents:
Step 1 : Reconstitution of both, cAMP-d2 & anti-CAMP cryptate with water (5ml_) according to suppliers manual.
Step 2: Further 1 :39 dilution with Conjugate & Lysis Buffer. Conjugate & Lysisbuffer prepared according to cAMP kit protocol (Cisbio).
The binding experiments revealed that compound 1 is a highly potent, competitive human EP4 receptor antagonist with inverse agonist activity. IN VIVO CHARACTERIZATION
EXAMPLE 3: Effect of compound 1 on CFA induced pain and edema (rat)
Model description:
Female SD rats were treated with compound 1 or respective vehicle. 1 h later all animals got an intraplantar injection of 50 · L of 100% Complete Freund's Adjuvant (CFA) under isoflurane anesthesia. CFA injection leads within hours to a robust edema accompanied by hyperalgesia and allodynia affecting the treated paw.
Endpoint 1 : Dynamic weight bearing (DWB)
DWB analyses the weight distribution of an animal on the four paws and is therefore able to give evidence for pain-related relief posture and could be used as a method to measure spontaneous pain reaction in the freely moving animal (Gruen et al. 2014). The automated DWB device (Bioseb, Boulogne, France) consists of a Plexiglas enclosure (22x22x30cm), a floor sensor composed of captors in order to detect pressure variation and a camera placed above the enclosure in order to validate the animal position during analysis (Robinson et al., 2012; Tetreault et al., 201 1 ). The rats were able to move freely in the enclosure during 5 min while the acquisition data were transmitted to a computer. The animals were not acclimated to the enclosure before the test.
Endpoint 2: Paw weight analysis (edema)
The paw was removed from the dead animal at the ankle joint and weight of the paw was noted.
Group (1 ): Vehicle: group (2): compound 1 , 0.2 mg/kg; group (3) compound 1 , 1 mg/kg: group (4): compound 1 , 5 mg/kg; group (5): compound 1 , 25 mg/kg Time scheme see Figure 8
Both, figure 2 and 3 demonstrate that compound 1 reduced inflammatory pain and edema formation significantly and dose-dependently.
EXAMPLE 4: Effect of COMPOUND 1 on PGE2 induced pain
Model description:
After the naive baseline values for paw withdrawal thresholds were obtained (day -1 ), animals were assigned to 5 groups (incl. morphine control) and received test article or vehicle administration (day 0, T=0). Approximately 60 minutes after the treatment (day 0, T=60 min), inflammatory pain was induced by a single injection of 10 μg dmPGE2 into the plantar surface of the left hind paw. Morphine served as the positive control for the study and was administered 30 minutes prior to dmPGE2 injection (day 0, T=30 min). Paw withdrawal thresholds were then measured 30 minutes after dmPGE2 injection (day 0, T=90 min). All animals were euthanized on day 0 following the behavioral assessment and plasma was collected from animals treated with test articles. Compound 1 was given at concentrations of 0.2 (dottet bar), 1 (horizontal hatched bar) and 5 mg/kg (vertical hatched bar).
Figure 4 shows that PGE2 induced pain is mediated by EP4 receptors. Compound 1 significantly reduced PGE2 induced pain dose dependendly.
EXAMPLE 5: Inhibition of PGE2 induced proliferation,
Model description:
Cell-number assay:
Cells (Cell culture: EM42 cells (Desai et al., 1994, Fert. Steril. 61 : 760) were cultivated one passage before experiment in test media (DMEM w/o phenolred, 1 % Pen/strep, 1 % L- glutamine plus indicated concentration of FCS [e.g. :1 %])
Indicated number of cells (e.g. 1000) was plated in wells of 96well plates (Perkin Elmer #6005688). Substances were added as indicated in tetraplicates, inhibitors were added first. PGE2 was given twice daily. For incubation without PGE2 the same amount of media was added twice daily. Incubation of MTPs for 4 days at 37°C and 5% CO? was followed by a CellTiter Glo measurement at day 4.
Preparation of cell number calibration curve from cells in test media: Cells were counted for calibration and plated in duplicates in rows 1 and 2 and incubated for two hours. Celltiter Glo measurement was performed as described by vendor (Promega #G7571 ). Briefly, MTPs were adjusted to room temperature for 30 minutes. 100μ! celltiter reagent was added, followed by mixing and incubation for 10 minutes. The luminescence was measured of with POLARstar (bmg). Measurements of relative light units (RLU) and calculation of cell number equivalent was performed by employing linear part of calibration curve.
Compound 1 (100 nM) was given with (crossed bar) or without (vertical hatched bar) 10 nM PGE2, PGE2 was also given alone (horizontal hatched bar). In the presence of compound 1 the total cell number did not increase when compared with the group which contained PGE2 alone. Compound 1 together with PGE2 also did not affect the total cell number.
EXAMPLE 6: Effect of compound 1 in endometriosis dyspareunia model (rat, vaginal distension
Model description:
Female SD rats underwent endometriosis surgery in the estrus phase. 4 uterine fragments were sewn onto the mesenteric artery arc (closed to small intestine) and few other fragments (>2) were sewn onto the distal colonic wall. In parallel electrodes for later EMG recordings were implanted in the abdominal wall to allow recording of the visceral motor response.
After training viscero motor response (VMR) / vaginal distension (VD) testing was performed 5 and 6 weeks post-surgery in the proestrus phase. For vaginal distension session, the uninflated balloon was lubricated and inserted into the mid-vaginal canal, located so, that it would not touch the cervix even when inflated. Inflating the balloon with different volumes of water using a computer-controlled pump distended the vaginal canal. The pressure produced by each volume of distention (corrected for compliance characteristics of the balloon) was measured through a small-volume pressure transducer. In parallel EMG recordings were collected to measure the VMR related to the distension.
Compound 1 (5 mg/kg, p.o.) was administered daily starting from week 3 to week 5 (group 2, N=19). Group 1 (N=20) received vehicle (20 % DMSO / 20 % PEG400 / 60 % water) in the same time period (5 ml/kg, p.o.), Group 3 (N=18) received I bu prof en (100 mg/kg, p.o.). Animals of group 3 were treated once 2 hours prior VMR, VD was at week 5 and week 6).
Compound 1 decreased vaginal hyperalgesia versus vehicle-treated animals. The effect maintained in the treatment free period of week 6. Therefore compound 1 is suitable for long term treatment of the aforementioned diseases. This might point to a disease modyfing component induced by the EP4 receptor antagonism exceeding pure analgesic activity. Interestingly, daily dosed compound 1 showed a more pronounced effect than acute treatment with one analgesic dose of ibuprofenoilected to measure the VMR related to the distension.

Claims

1 . Use of compound 1 for the treatment of inflammatory pain and/or pathological conditions related to increased activation of EP4 receptors by prostaglandin E2 (PGE2).
2. Use of compound 1 for the treatment of inflammatory pain.
3. Use according to Claim 2, whereby the inflammatory pain is selected from a group comprising but not limited to ankylosing spondylitis, or pain associated with endometriosis, post operative pain, pain related to skin burn injuries and osteoarthritis.
4. Use of compound 1 for the preparation of a medicament for treatment of inflammatory pain.
5. Medicament comprising compound 1 in combination with one or more further active compounds, especially with selective oestrogen receptor modulators (SERMs), oestrogen receptor (ER) antagonists, aromatase inhibitors, 17-· -HSD1 inhibitors, steroid sulphatase (STS) inhibitors, GnRH agonists and antagonists, kisspeptin receptor (KISSR) antagonists, selective androgen receptor modulators (SARMs), androgens, 5· -reductase inhibitors, selective progesterone receptor modulators (SPRMs), gestagens, antigestagens, oral contraceptives, inhibitors of mitogen- activated protein (MAP) kinases and inhibitors of the MAP kinases (Mkk3/6, Mek1/2, Erk1/2), inhibitors of protein kinase B (ΡΚΒ· /· /· ; Akt1/2/3), inhibitors of phosphoinositide 3-kinase (PI3K), inhibitors of cyclin-dependent kinase (CDK1/2), inhibitors of the hypoxia-induced signalling pathway (HIFI alpha inhibitors, activators of prolylhydroxylases), histone deacetylase (HDAC) inhibitors, prostaglandin F receptor (FP) (PTGFR) antagonists, neurokinin 1 receptor antagonists, paracetamol, selective COX2 inhibitors and/or non-selective COX1/COX2 inhibitors. Medicament comprising compound 1 in combination with an inert, nontoxic, pharmaceutically suitable auxiliary.
6. Medicament according to Claim 4 or 5 for the treatment of inflammatory pain.
7. Use of compound 1 in the form of a pharmaceutical preparation for enteral, parenteral, vaginal, intrauterine, topical and oral administration.
8. Method for alleviating pathological conditions associated with PGE2 induced increased EP4 receptors activation by antagonising the activation of EP4 receptors by compound 1 .
PCT/EP2018/055615 2017-03-10 2018-03-07 Use of an ep4 antagonist for the treatment of inflammatory pain WO2018162562A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP17160290 2017-03-10
EP17160290.7 2017-03-10

Publications (1)

Publication Number Publication Date
WO2018162562A1 true WO2018162562A1 (en) 2018-09-13

Family

ID=58266489

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2018/055615 WO2018162562A1 (en) 2017-03-10 2018-03-07 Use of an ep4 antagonist for the treatment of inflammatory pain

Country Status (2)

Country Link
TW (1) TW201836604A (en)
WO (1) WO2018162562A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019038156A1 (en) * 2017-08-22 2019-02-28 Bayer Pharma Aktiengesellschaft Use of an ep4 antagonist for the treatment of arthritis

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002032900A2 (en) 2000-10-19 2002-04-25 Pfizer Pharmaceuticals Inc. Aryl or heteroaryl fused imidazole compounds as anti-inflammatory and analgesic agents
WO2003086371A2 (en) 2002-04-12 2003-10-23 Pfizer Japan Inc. Use of ep4 receptor ligands in the treatment of il-6 involved diseases
WO2004067524A1 (en) 2003-01-29 2004-08-12 Pharmagene Laboratories Limited Ep4 receptor antagonists
WO2005102389A2 (en) 2004-04-20 2005-11-03 Pfizer Products Inc. Combinations comprising alpha-2-delta ligands and ep4 receptor antagonists
WO2005105733A1 (en) 2004-05-04 2005-11-10 Pfizer Japan Inc. Ortho substituted aryl or heteroaryl amide compounds
WO2005121508A2 (en) 2004-06-02 2005-12-22 Sunpower, Inc. Method and apparatus for forming a heat exchanger
WO2007121578A1 (en) 2006-04-24 2007-11-01 Merck Frosst Canada Ltd. Indole amide derivatives as ep4 receptor antagonists
WO2008017164A1 (en) 2006-08-11 2008-02-14 Merck Frosst Canada Ltd. Thiophenecarboxamide derivatives as ep4 receptor ligands
WO2008104055A1 (en) 2007-02-26 2008-09-04 Merck Frosst Canada Ltd. Indole and indoline cyclopropyl amide derivatives as ep4 receptor antagonists
WO2009020588A1 (en) 2007-08-09 2009-02-12 Merck & Co., Inc. Process for making thiophene carboxamide derivative
EP2172447A1 (en) 2007-07-03 2010-04-07 Astellas Pharma Inc. Amide compound
WO2014086739A1 (en) 2012-12-06 2014-06-12 Bayer Pharma Aktiengesellschaft Novel benzimidazole derivatives as ep4 antagonists
US20160318905A1 (en) * 2013-12-19 2016-11-03 Bayer Pharma Aktiengesellschaft Novel benzimidazole derivatives as EP4 ligands

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002032900A2 (en) 2000-10-19 2002-04-25 Pfizer Pharmaceuticals Inc. Aryl or heteroaryl fused imidazole compounds as anti-inflammatory and analgesic agents
WO2002032422A2 (en) 2000-10-19 2002-04-25 Pfizer Pharmaceuticals Inc. Ep4 receptor inhibitors to treat rheumatoid arthritis
WO2003086371A2 (en) 2002-04-12 2003-10-23 Pfizer Japan Inc. Use of ep4 receptor ligands in the treatment of il-6 involved diseases
WO2004067524A1 (en) 2003-01-29 2004-08-12 Pharmagene Laboratories Limited Ep4 receptor antagonists
WO2005102389A2 (en) 2004-04-20 2005-11-03 Pfizer Products Inc. Combinations comprising alpha-2-delta ligands and ep4 receptor antagonists
WO2005105733A1 (en) 2004-05-04 2005-11-10 Pfizer Japan Inc. Ortho substituted aryl or heteroaryl amide compounds
WO2005121508A2 (en) 2004-06-02 2005-12-22 Sunpower, Inc. Method and apparatus for forming a heat exchanger
WO2007121578A1 (en) 2006-04-24 2007-11-01 Merck Frosst Canada Ltd. Indole amide derivatives as ep4 receptor antagonists
WO2008017164A1 (en) 2006-08-11 2008-02-14 Merck Frosst Canada Ltd. Thiophenecarboxamide derivatives as ep4 receptor ligands
WO2008104055A1 (en) 2007-02-26 2008-09-04 Merck Frosst Canada Ltd. Indole and indoline cyclopropyl amide derivatives as ep4 receptor antagonists
EP2172447A1 (en) 2007-07-03 2010-04-07 Astellas Pharma Inc. Amide compound
WO2009020588A1 (en) 2007-08-09 2009-02-12 Merck & Co., Inc. Process for making thiophene carboxamide derivative
WO2014086739A1 (en) 2012-12-06 2014-06-12 Bayer Pharma Aktiengesellschaft Novel benzimidazole derivatives as ep4 antagonists
US20160214977A1 (en) * 2012-12-06 2016-07-28 Bayer Pharma Aktiengesellschaft Novel benzimidazole derivatives as ep4 antagonists
US20160318905A1 (en) * 2013-12-19 2016-11-03 Bayer Pharma Aktiengesellschaft Novel benzimidazole derivatives as EP4 ligands

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BASBAUM ET AL., CELL, vol. 139, 2009, pages 267 - 284
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 122060-05-3

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019038156A1 (en) * 2017-08-22 2019-02-28 Bayer Pharma Aktiengesellschaft Use of an ep4 antagonist for the treatment of arthritis

Also Published As

Publication number Publication date
TW201836604A (en) 2018-10-16

Similar Documents

Publication Publication Date Title
US9708311B2 (en) Benzimidazole derivatives as EP4 antagonists
US10730856B2 (en) Benzimidazole derivatives as EP4 ligands
US20080254106A1 (en) Use Of Pirlindole For The Treatment Of Diseases Which Are Characterized By Proliferation Of T-Lymphocytes And/Or Hyperproliferation Of Keratinocytes In Particular Atopic Dermatitis And Psoriasis
JP4152186B2 (en) GABA enhancer in the treatment of diseases associated with reduced neurosteroid activity
CN104470941A (en) Targeted therapeutics
PT1149579E (en) Use of an estrogen agonist/antagonist for treating female sexual dysfunction
DE102011002934A1 (en) CB2 agonists for the treatment and prevention of endometriosis
AU2018280742B2 (en) Gonadotropin-releasing hormone antagonist dosing regimens for the treatment of endometriosis
WO2016007616A1 (en) Methods for treating neurologic disorders
Sanna et al. Dopamine D2-like receptor agonists induce penile erection in male rats: differential role of D2, D3 and D4 receptors in the paraventricular nucleus of the hypothalamus
JP2013504531A (en) Substituted (heteroarylmethyl) thiohydantoins as anticancer drugs
AU2020325655A1 (en) Compositions and methods for the treatment of estrogen-dependent disorders
WO2018162562A1 (en) Use of an ep4 antagonist for the treatment of inflammatory pain
WO2019038156A1 (en) Use of an ep4 antagonist for the treatment of arthritis
EP4243829A1 (en) Methods of administering relugolix
AU2019373349A1 (en) Compositions and methods for the treatment of adenomyosis and rectovaginal endometriosis
JP2022500445A (en) GABAA receptor ligand
JP2005523910A (en) Combination of aldosterone receptor antagonist and bile acid sequestrant
WO2018011015A1 (en) Low-dosed oral dosage forms for treatment of diseases
TWI835716B (en) Tetrahydrocyclopenta[b]indole compounds and phosphodiesterase inhibitors for the treatment of the signs and symptoms of bph
US20230285347A1 (en) Preterm Labour with Prostaglandin E2 Receptor Agonists
IT201800005721A1 (en) NEW COMPOSITIONS FOR THE TREATMENT OF UTERINE LEIOMIOMIS
BR112019018700A2 (en) pharmaceutical compositions and their uses
US20030207854A1 (en) Antiprogestins with partial agonist activity
Clément et al. Selective antagonism of dopamine D3 receptor specifically inhibits the expulsion phase of ejaculation in anaesthetised male rats

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: 18709009

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18709009

Country of ref document: EP

Kind code of ref document: A1