WO2018046685A1 - Quinoline derivatives for treatment of inflammatory skin diseases - Google Patents

Quinoline derivatives for treatment of inflammatory skin diseases Download PDF

Info

Publication number
WO2018046685A1
WO2018046685A1 PCT/EP2017/072626 EP2017072626W WO2018046685A1 WO 2018046685 A1 WO2018046685 A1 WO 2018046685A1 EP 2017072626 W EP2017072626 W EP 2017072626W WO 2018046685 A1 WO2018046685 A1 WO 2018046685A1
Authority
WO
WIPO (PCT)
Prior art keywords
compounds
fluoro
skin diseases
inflammatory skin
psoriasis
Prior art date
Application number
PCT/EP2017/072626
Other languages
French (fr)
Inventor
Markus Berger
Ekkehard May
Claudia Giesen
Kaweh SHAKERY
Heike Schäcke
Kristin KOWAL
Original Assignee
Bayer Aktiengesellschaft
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 Aktiengesellschaft, Bayer Pharma Aktiengesellschaft filed Critical Bayer Aktiengesellschaft
Publication of WO2018046685A1 publication Critical patent/WO2018046685A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/38Nitrogen atoms
    • 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
    • A61K31/47042-Quinolinones, e.g. carbostyril
    • 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
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics

Definitions

  • the present invention relates to compounds for use in the topical treatment of inflammatory skin diseases such as psoriasis, atopic dermatitis, allergic contact dermatitis, wherein the said compounds are proprietary non-steroidal selective glucocorticoid receptor agonists (SEGRAs).
  • SEGRAs selective glucocorticoid receptor agonists
  • Glucocorticoids are commonly viewed as the most active topical compounds for the treatment of inflammatory skin diseases, but while they are well established in the treatment of psoriasis, atopic dermatitis and allergic contact dermatitis, their use is limited by their potential to induce a number of severe and sometimes irreversible local and/or systemic side effects (Hengge UR et al, 2006). Activation and repression of genes are mediated by different molecular mechanisms of the glucocorticoid receptor ("transactivation” and "transrepression”). This differentiation has provided a working model to search for novel synthetic glucocorticoid receptor- ligands that have a better therapeutic index than marketed glucocorticoids.
  • non-steroidal anti-inflammatory agents of the following general formula
  • the present invention relates to compounds according to general formula (I) for use in the topical treatment of T-cell mediated inflammatory skin diseases,
  • R 1 and R 2 represent independently from each other, a hydrogen atom, a hydroxyl group, a halogen atom, an optionally substituted (Ci-Cio)-alkyl group, an optionally substituted (Ci-Cio)-alkoxy group, a (Ci-Cio)-alkylthio group, a (Ci-C5)-perfluoroalkyl group, a cyano group or a nitro group, or
  • R 1 and R 2 represent independently from each other, NR 6 R 7 , wherein R 6 und R 7 are, independently from each other, a hydrogen atom, Ci-Cs-alkyl or -(CO)-(Ci-C5)-alkyl, R 3 represents for a hydrogen atom, a hydroxyl group, a halogen atom, a cyano group, an optionally substituted (Ci-Cio)-alkyl group, a (Ci-Cio)-alkoxy group, a (C1-C10)- alkylthio group or a (Ci-C5)-perfluoroalkyl group,
  • R 4 represents a hydrogen atom, a halogen atom, a hydroxyl group, a (Ci-C5)-alkyl group, a (Ci-C5)-alkoxy group, a (Ci-C5)-alkylthio group, a (Ci-C5)-perfluoroalkyl group, a cyano group, a nitro group, -NR 6 R 7 , -COOR 9 , -(CO)NR 6 R 7 or a (C1-C5- alkylen)-0-(CO)-(Ci-C5)-alkyl group, wherein R 6 and R 7 have the same meaning as defined above and R 9 represents a (Ci-Cio)-alkyl group or a (Ci-Cio)-alkoxy group, R 5 represents a group selected from the group consisting of -(Ci-Cio)-alkyl, which might be fully or partially substituted by a halogen atom,
  • R 8 represents an aryl group, which might be substituted by 1 to 3 hydroxy groups, halogen, Ci-Cs-alkyl, Ci-Cs-alkoxy, cyano group, CF 3 , nitro, -COO(Ci-C5-alkyl) or - C(0)OCH 2 -phenyl or a heteroaryl group, whereby the heteroaryl group might comprise 1 to 3 heteroatoms, which optionally are substituted by 1 to 3 alkyl groups, hydroxy, halogen, cyano or Ci-Cs-alkoxy groups,
  • Compound of formula I-A is known from WO 2009/065503 A (ex. 5 as racemate) and can be isolated from racemic form by chiral HPLC, Chiralpak IC 5 ⁇ with eluent hexane/ethanol (4 : 1).
  • Compound of formula I-B is known from WO 2009/065503 A (ex. 7 as racemate) and can be iso- lated from racemic form by chiral HPLC, Chiralpak IC 5 ⁇ with eluent hexane/ethanol (4 : 1).
  • Compound of formula I-C is known from WO 2009/065503 A (ex. 3 as racemate) and can be isolated from racemic form by chiral HPLC, Chiralpak IC 5 ⁇ with eluent hexane/ethanol (4 : 1).
  • Fluorescence polarization allows the examination of molecular interactions by monitoring changes in the apparent size of fluorescently-labeled compounds. To determine the binding of ligands to a specific receptor the fluorescence polarization technology makes use of a fiuorescent ligand for a specific receptor.
  • the technology is based on the observation that small fluorescent ligands after excitation with plane polarized light emit largely depolarized light as they tumble rapidly in the test solution, while large complexes of fluorescent ligands bound to their receptors rotate significantly slower. Accordingly, the emitted light of these complexes maintains the polarization of the exited light as it remains in the same plane as the excitation energy.
  • the high polarization values of fluo- rescent ligand receptor binding complexes are decreased by the addition of increasing concentrations of non-fluorescently-labeled ligands which replace the fiuorescent ligand.
  • the commercially available fluorescence polarization assay from PanVera/Invitrogen was used.
  • This kit contains the reagents necessary to perform a competition assay to assess the affinity of test compounds for the human glucocorticoid receptor.
  • Recombinant human glucocorticoid receptor together with a stabilizing peptide is added to a fixed concentration (1.0E-9 mo 1/1) of a fluorescent glucocorticoid (FluormoneTM GS Red) ligand in the presence of different concentrations (1.6E-8, 8.0E-8, 4.0E-7, 2.0E-6, 1.0E-5 mo 1/1) of the test compounds in microwell plates.
  • the plates are incubated for 2h at room temperature in the dark.
  • the presence of effective competitors for the GR binding site prevents the formation of GS Red/GR complexes resulting in a decrease of the polarization value.
  • the polarization values are directly determined in the microplate.
  • the shift in polarization values in the presence of test compounds is measured with an Analyst plate reader (LJL Biosystems/Molecular Devices Corporation) (excitation wavelength 530nm, emission wavelength 590 nM).
  • the change in fluorescence polarization with increasing concentrations of non- fluorescent ligand is plotted versus the log of the ligand
  • the IC50 values were calculated using the Microsoft ® Excel-compatible XLfit ® Four Parameter curve fitting tool.
  • the compounds (I-C) , (I-A) and (I-B) see below) are able to replace the fluorescently-labeled ligand in a dose dependent manner. Similar to the reference compound, Clobetasol, with an IC50 value of 6.0E-9 mol/1, the compounds (I-C), (I-A), and (I-B), were also able to replace the fluorescent ligand in a potent manner with low IC50 values of 4.2E-9 mol/1, 3.1E-9 mol/1 and 7.1E-9 mol/1. This clearly shows that the compounds (I-C) , (I-A) and (I-B) have a very high affinity to the human glucocorticoid receptor (see Table 1).
  • the selectivity of the compounds of general formula (I) towards the glucocorticoid receptor has been demonstrated in further steroid hormone receptor binding assays.
  • the binding of the com- pounds of the general formula (I) to the progesterone receptor (PR), the androgen receptor (AR), the estrogen-alpha (ER-a) and the estrogen-beta (ER- ⁇ ) receptor was examined in the presence of a stabilizing peptide with the fluorescence polarization technology.
  • FluomoneTM PL Red (1.0E-9 mol/) was used as fluorescent ligand for the human PR, FluomoneTM AL Green (5.0E-10 mol/1) as fluorescent ligand for the rat AR, and FluomoneTMEL Red (5.0E-10 mol/1) as fluorescent ligand for the ER-a and the ER- ⁇ .
  • the fluorescently- labeled ligand receptor mixtures were incubated in micro well plates at room temperature in the dark for 1-6 h (PR), 4-6 h (AR), and 1-5 h (ER-a and ER ) in the presence of different concentrations (1.6E-8, 8.0E-8, 4.0E-7, 2.0E-6, l .OE-5 mol/1) of the compounds (I-A) and (I-B). At the end of the incubation period the polarization values are directly determined in the microplate.
  • the shift in polarization values in the presence of test com- pounds is measured with an Analyst plate reader (LJL Biosystems/Molecular Devices Corporation) (for FluomoneTM PL Red and EL Red: excitation wavelength 530nm, emission wavelength 590 nM; for FluomoneTM AL Green: excitation wavelength 485 nm, emission wavelength 530 nM).
  • the change in fluorescence polarization with increasing concentrations of non- fluorescent ligand is plotted versus the log of the ligand concentration.
  • the IC50 values were calculated using the Microsoft ® Excel-compatible XLfit ® Four Parameter curve fitting tool.
  • the binding of the compound (I-A) and (I-B) to the human mineralocorticoid receptor (MR) was assessed using a ligand competition binding scintillation proximity assay (SPA).
  • SPA ligand competition binding scintillation proximity assay
  • High Five cells an insect cell line, were transfected with the human mineralocorticoid receptor ligand binding domain (NR3C2; aa729-964) with an N-terminal maltose binding protein (MBP) tag.
  • MBP maltose binding protein
  • the compounds (I-A) and (I-B) do either not bind to the receptors of other steroid hormones or show only at very high concentrations a weak binding to the other steroid hormone receptors. Similar to the glucocorticoid Clobetasol, the compound (I-A) does not bind to the human estrogen-alpha receptor, the human estrogen-beta receptor, and the rat androgen receptor. 50% inhibition of the ⁇ -estradiol and the testosterone bind- ing, respectively, were even not reached in the very high concentration range of ⁇ 1.OE-5 mol/1.
  • the compound (I-A) is also only able to bind very weakly to the human mineralocorticoid receptor. At the very high concentration of 1.2E-6 mol/1 the aldosterone binding is inhibited by 50%, which is an even by a factor of 5 weaker binding compared to Clobetasol.
  • the comparison of the IC50 values for the binding of (I-A) to the human glucocorticoid and the human progesterone receptor shows that it binds by a factor of 20 weaker to the human progesterone receptor: similar to Clobetasol the compound (I-A) inhibits the progesterone binding only at a concentration of 6.1E-8 mol/1 by 50%.
  • the compound of (I-B) does also not bind to the rat androgen receptor. It exerts compared to the IC50 value for the GR binding (IC50: 7.1E-9 mol/1) by a factor of -100 weaker binding to the human mineralocorticoid receptor (IC50: 7.7E-7 mol/1) and by a factor of 15 weaker binding to the human progesterone receptor (IC50: 1.1E-7 mol/1) (see also Table 2).
  • Table 2 Table 2:
  • hGR human glucocorticoid receptor
  • hPR human progesterone receptor
  • RAR rat androgen receptor
  • hMR human mineralocorticoid receptor
  • hERa human estrogen-alpha receptor
  • hERB human estrogen-beta receptor
  • glucocorticoid receptor is characterized by a ligand binding domain (LBD), which specifically recognizes and binds natural and artificial glucocorticoids and by a highly conserved DNA-binding motif (DBD) which recognizes specific DNA sequences, the so-called glucocorticoid response elements (GREs), which regulate gene transcription and lead thereby to the activation or suppression of a distinct subset of genes.
  • LBD ligand binding domain
  • DBD DNA-binding motif
  • GREs glucocorticoid response elements
  • the ligand binding domain of the glucocorticoid receptor as well as that of other members of the steroid hormone receptor family can be fused to the DNA binding domains of other transcription factors of different species like yeast using recombinant DNA technologies.
  • a suited DNA binding domain is that of the yeast GAL4 transcription factor which is involved in transcriptional control of the galactose metabolism in yeast.
  • the upstream activating sequences (UAS) which are recognized by the GAL4 transcription factor are not present in the mammalian genome.
  • GAL4 upstream activating sequences fused to a reporter gene like luciferase into the genome of a mammalian cell such as the CHO (Chinese Hamster Ovary) cells, allows the so-called transactivation of the luciferase gene by the GAL4 transcription factor.
  • the additional fusion of the ligand binding domain of the glucocor- ticoid receptor leads to a chimera which transactivates the luciferase reporter gene upon binding of glucocorticoids.
  • the expression of luciferase is measured by the addition of its substrate, luciferin. Luciferase converts luciferin to oxyluciferin.
  • the modified CHO cells were incubated at room temperature for 5-6h with either the compound (I-A), or the glucocorticoids Clobetasol or Dexamethasone, at 8 different concentrations in the range of 1.0E-9 to 1.0E-6 mo 1/1. After the incubation period the luciferase activity was determined using the Lumibox Video Camera system. Calculation of the EC50 values, the ligand concentration which leads to a half-maximum luciferase expression, was carried out with the GraphPad PRISM tool, a curve fitting program on the basis of non-linear regression.
  • the compound (I-A) was able to induce luciferase expression in a potent and dose dependent way.
  • the compound (I-A) achieved a maximum activation of the luciferase expression (-100% efficacy) which is comparable to the maximum activation induced by the reference glucocorticoids, Clobetasol, and Dexamethasone,
  • the comparison of the EC50 values showed that the compound (I-A) is with an EC50 of 5.2E-9 mo 1/1 as potent as Dexamethasone (EC50: 6.0E-9 mo 1/1) and slightly less potent than Clobetasol (EC50: 1.1E-9 mo 1/1) (see also Table 3).
  • Table 3 Table 3:
  • transfected CHO cell system was also used to check whether compound of the general formula (I) were able to exert antagonistic activities mediated via the GR.
  • transfected CHO cells were incubated with increasing concentrations of the compound (I-A) (1.0E-9 - 1.0E-5 mol/) in the presence of a constant concentration of Dexamethasone (6.0E-9 mol/1). It could be demonstrated that the Dexamethasone-induced half maximum luciferase expres- sion was not attenuated by the addition of increasing concentrations of the compound (I-A). Accordingly, the compound (I-A) was not able to exert antagonistic activities in the physiological concentration range.
  • the object of the present invention is to provide compounds for use in the topical treatment of T- cell mediated inflammatory skin diseases such as psoriasis, atopic dermatitis or allergic contact dermatitis in adults, adolescents, children and infants.
  • Compounds according to the present invention for use in the topical treatment of T-cell mediated inflammatory skin diseases are identified to achieve anti-inflammatory effects by attenuating the activity of cells belonging to the immune system after topical administration. At the same time these compounds should be characterized by a clear dissociation between highly beneficial antiinflammatory activities and undesired side effects.
  • the compounds according to the present invention are characterized by an optimized benefit-risk- ratio which means that their therapeutic effects are not or to an only minor extent accompanied by side effects (Schaecke H et al, 2004; Schaecke H et al, 2006; Schaecke H et al, 2007; Stahn et al, 2007).
  • the compounds according to the present invention represent effective and safe topical treatment alternatives especially for inflammatory skin diseases.
  • Considerable safety concerning the use of such compounds is provided, in particular when such compounds are administered to patients who are susceptible to glucocorticoid side effects, e.g. children and infants.
  • compounds like e.g. (I-D) known form WO 2009/065503 which despite of potent binding to the Glucocorticoid receptor excerted extremely weak inhibition of T-cell mediated activities. This is a clear indicator that compounds like e.g. (I-D) are not able to induce any antiinflammatory or immunomodulatory activities in T-cell mediated skin diseases.
  • the present inventors had to carefully select , develop suitable assays in order to identify suitable compounds for T-cell mediated skin diseases.
  • the present invention relates to compounds according to general formula (I) for use in the topical treatment of T-cell mediated inflammatory skin diseases,
  • R 1 and R 2 represent independently from each other, a hydrogen atom, a hydroxyl group, a halogen atom, an optionally substituted (Ci-Cio)-alkyl group, an optionally substituted (Ci-Cio)-alkoxy group, a (Ci-Cio)-alkylthio group, a (Ci-C5)-perfluoroalkyl group, a cyano group or a nitro group, or
  • R 1 and R 2 represent independently from each other, NR 6 R 7 , wherein R 6 und R 7 are, independently from each other, a hydrogen atom, Ci-Cs-alkyl or -(CO)-(Ci-C5)-alkyl, R represents for a hydrogen atom, a hydroxyl group, a halogen atom, a cyano group, a optionally substituted (Ci-Cio)-alkyl group, a (Ci-Cio)-alkoxy group, a (Ci-Cio)- alkylthio group or a (Ci-C5)-perfluoroalkyl group,
  • R 6 and R 7 have the same meaning as defined above and R 9 represents a (Ci-Cio)-alkyl group or a (Ci-Cio)-alkoxy group, represents for a group, selected from the group consisting of -(Ci-Cio)-alkyl, which might be fully or partially substituted by a halogen atom,
  • R 8 represents for an aryl group, which might be substituted by 1 to 3 hydroxy groups, halogen, Ci-Cs-alkyl, Ci-Cs-alkoxy, cyano group, CF 3 , nitro, -COO(Ci-C5-alkyl) or -C(0)OCH 2 -phenyl or a heteroaryl group, whereby the heteroaryl group might comprise 1 to 3 heteroatoms, which optionally are substituted by 1 to 3 alkyl groups, hydroxy, halogen, cyano or Ci-Cs-alkoxy groups,
  • One aspect of the invention relates to compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases, characterized in that the said compounds are non-steroidal selective glucocorticoid receptor agonists.
  • Another aspect of the invention relates to compounds for use in the topical treatment of T-cell me- diated inflammatory skin diseases, characterized in that the said compounds are able to inhibit the proliferation of stimulated human lymphocytes in a potent and dose-dependent manner, demonstrated in a suited in vitro test system which made use of compound doses ranging between 1.0E- 12 and l .OE-6 mo 1/1 and showed potent inhibition of lymphocyte proliferation in the nanomolar range.
  • Still another aspect of the invention relates to compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases, characterized in that the said compounds are able to inhibit the release of cytokines such as IL-2, IL-4, and IL-23 which induce the proliferation of certain subsets of T cells associated with inflammatory reactions.
  • cytokines such as IL-2, IL-4, and IL-23 which induce the proliferation of certain subsets of T cells associated with inflammatory reactions.
  • a further aspect of the invention relates to compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases, characterized in that the said compounds are able to inhibit the release of cytokines including IL-12, TNF-a or IFN- ⁇ , which exert immuno stimulatory and proinflammatory effects.
  • cytokines including IL-12, TNF-a or IFN- ⁇
  • Another aspect of the invention relates to compounds for use in the topical treatment of inflammatory skin diseases, characterized in that the inhibition of cytokine release and related proliferation of T cells is involved in the induction and perpetuation of psoriasis.
  • Still another aspect of the invention relates to compounds for use in the topical treatment of inflammatory skin diseases, wherein the said inflammatory skin diseases include psoriasis.
  • a further aspect of the invention relates to compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases, wherein the said skin disease, psoriasis, includes the sub- classes psoriasis vulgaris, plaque-type psoriasis, and pustular psoriasis.
  • Another aspect of the invention relates to compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases, wherein the said T-cell mediated inflammatory skin diseases include atopic dermatitis and allergic contact dermatitis.
  • Still another aspect of the invention relates to compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases, characterized in that the said compounds are able to inhibit T-cell induced inflammatory skin reactions in rodent models such as DNFB-induced or TMA- induced allergic contact dermatitis reactions.
  • Another aspect of the invention relates to compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases, characterized in that the said compounds are characterized ei- ther by a lack of or by a reduced level of side effects typically caused by topical glucocorticoids.
  • Still another aspect of the invention relates to compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases, characterized in that the said compounds are marked by a lack of or by a reduced level of side effects typically caused by topical glucocorticoids, which include skin atrophy, hypothalamic-pituitary-adrenal (HP A) axis suppression, thymus and spleen atrophy or growth retardation.
  • topical glucocorticoids include skin atrophy, hypothalamic-pituitary-adrenal (HP A) axis suppression, thymus and spleen atrophy or growth retardation.
  • Another aspect of the invention relates to compounds for use in the topical treatment of T-cell me- diated inflammatory skin diseases, characterized in that the said compounds are used for T-cell mediated inflammatory skin diseases such as psoriasis, atopic dermatitis and allergic contact dermatitis in adults, adolescents, children and infants.
  • T-cell mediated inflammatory skin diseases such as psoriasis, atopic dermatitis and allergic contact dermatitis in adults, adolescents, children and infants.
  • Still another aspect of the invention relates to compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases, wherein the said compounds do not have side effects or have a reduced level of side effects typically caused by topical glucocorticoids and can be used especially for inflammatory skin diseases in children and infants.
  • Another aspect of the invention relates to compounds for use in the topical treatment of inflammatory skin diseases, wherein the following compounds are particularly preferred:
  • T-cell mediated inflammatory skin diseases such as psoriasis, atopic dermatitis, and allergic contact dermatitis and compounds for use in topical treatment of these skin diseases are described in detail.
  • Psoriasis is a common chronic dermatosis and one of the most prevalent chronic inflammatory diseases, affecting approximately 2% of the population in western industrialized countries. The disease often persists for life and is regarded as very troublesome, leading to notable impact on a sufferer's quality of life. A reappraisal of the disease in recent years, which was also triggered by the high economic impact the disease has, led to the discovery of similarities with other inflammatory diseases, leading to psoriasis now being regarded as a model for other chronic inflammatory diseases (Nestle FO et al, 2009). Further, a broad spectrum of associated diseases has been identified in recent years. Psoriasis can occur rather early in life (ca.
  • Psoriasis Typ 1 often comes with a positive family history for psoriasis, and is prominently associated with a number of genetic factors: so called "Psoriasis susceptibility loci" are located on different chromosomes (PSORS 1-9) and involve HLA-Cw6 (PSORS- 1, located at 6p21.3) and other genes important for inflammatory reactions, like TNF-a and IL-23 receptor variants, as well as various other factors (Nestle FO et al, 2009).
  • Typ 2 psoriasis often shows no positive family history, and is less prominently associated with genetic factors. Often, these cases are less severely affected, but show a very chronic disease course with stationary plaques at predilection sites.
  • Psoriasis also shows a number of frequently occurring comorbidities: 1 out of 5 patients develops the so-called psoriatic arthritis, a seronegative spondyloarthropathy. Typical for this condition is an asymmetric involvement of small joints of the hands and feet, but also some large joints (e.g. knee or ankle) may be affected. Spondylitis and sacroiliitis may also be observed. Early recognition and treatment of psoriatic arthritis are mandatory as around 5% of these patients will develop a severe, mutilating (destructive) arthritis, leading to permanent disability.
  • psoriasis Further comorbidities that may occur with psoriasis are other immune-mediated inflammatory diseases like rheumatoid arthritis and Crohn's disease.
  • the metabolic syndrome and the single components defining it hyperertonia, obesity, hyper lipidemia and diabetic-prone metabolic states
  • hyper lipidemia and diabetic-prone metabolic states
  • psoriasis requires a management that includes long-term therapy as well as an awareness regarding potentially occurring co-morbidities (Levine D et al, 2009).
  • psoriasis can be further categorized into psoriasis vulgaris, or plaque-type psoriasis, and pustular psoriasis.
  • Psoriasis vulgaris plaque-type psoriasis
  • Erythro squamous plaques red skin elevation with white or silvery scaling
  • Predilection sites are the extensor sites of the extremities (e.g.
  • Deviations from this scheme are subtypes of psoriasis vulgaris that are defined by the anatomical region of affected skin, like palmoplantar psoriasis (involving palms of the hands and soles of the feet), psoriasis capitis (involving the scalp), psoriasis inversa and psoriasis intertriginosa (involving axillae, groin, submammar areas, skin folds (e.g. obese abdomen)).
  • the erythro squamous plaques may be stable for a long time with a limited amount of body surface area involved, while in moderate or severe forms a sudden extension of affected body area takes place.
  • the range of disease activity pattern thus involves a stable disease state as well as an undulating activity, where phases of stable disease activity (or even without any observable skin affection) take turns with phases of extensive involvement of the body surface area, up to an involvement of the complete integument (erythroderma, psoriasis involving the whole skin).
  • Guttate psoriasis with small erythro squamous plaques (up to the size of a coin) being organized in an exanthema- like pattern over wide areas of the body, also belongs into this group and is sometimes seen after infec- tions, leading to the hypothesis that this form of psoriasis is triggered by immunological pathways triggered by e.g. a bacterial infection.
  • Pustular psoriasis is a severe exudative inflammatory variant of psoriasis defined by the occurrence of pustules. Located on erythematous plaques, the pustules may also become confluent - resulting in larger pus-filled vesicles and bullae. The pustules contain pus that is sterile, as opposed to pus- tules known from other dermato logical diseases (e.g. folliculitis) were pathogens like Staphylococcus aureus are involved.
  • folliculitis e.g. folliculitis
  • the sterile pustules in this case are a symptom of the excessive influx of granulocytes from blood into outer skin layers, triggered by the psoriasis pathophysiology.
  • This disease form is generally seen as an expression of high psoriatic disease activity and may occur as inde- pendent psoriasis subtype, or may be observed as an expression of high disease activity in a patient previously suffering from psoriasis vulgaris (psoriasis cum pustulatione).
  • TLR To 11- like-receptors
  • keratinocytes of the skin produce antimicrobial substances that also exert immune-mo dulatory effects: Dendritic cells are attracted to migrate into the skin and are activated, while a subsequent interaction of dendritic cells and T-cells leads to the expansion of certain lymphocyte subpopula- tions in the skin. The transport of T cells from the dermis into the epidermis is a key event in psoriasis.
  • Interleukin (IL) 12 is secreted by activated dendritic cells and leads to an expansion of the number of Thl -cells, a subset of T-cells, with the subsequent production of other pro -inflammatory cytokine.
  • IL-12 is a heterodimeric cytokine encoded by two separate genes, coding for the subunits IL-12p35 and IL-12p40.
  • the subunit IL-12p40 is also shared with IL-23 subunit that forms, together with IL-23pl9, the proinflammatory cytokine Interleukin 23, another heterodimeric proinflammatory cytokine.
  • IL-23 propels the number of activated Thl 7 cells in the skin, promoting IL-17, IL-22, IL-26 and TNF-a (Biedermann T et al, 2004).
  • the resulting pro -inflammatory milieu leads to a complex secondary, self-sustaining reaction with pro -inflammatory activation of keratinocytes as well as endothelial cells, leading to the influx of more dendritic cells, lymphocytes and neutrophilic granulocytes (sterile micro-abscesses).
  • topical corticosteroids are still the cornerstone of treatment for the majority of patients with psoriasis, according to the American Academy of Dermatology's Guidelines of care for the management of psoriasis (Menter A et al, 2009).
  • Atopic dermatitis (AD, synonyms: neurodermitis, atopic eczema, prurigo besnier, endogenous ec- zema) is an inflammatory, chronically relapsing, non-contagious and intensely pruritic dermatosis characterized by epidermal inflammation, itching, dry skin (with fine scaling) and exudation (in acute lesions). It is one of the most common skin diseases with a prevalence of 2-3% in the adult population, and up to 20% in the pediatric population, with an increasing prevalence in western countries over three decades (Leung DYM et al, 2004). AD usually follows a relapsing course and is often associated with elevated serum immunoglobulin (IgE) levels.
  • IgE serum immunoglobulin
  • AD Alzheimer's disease
  • atopic diseases like asthma bronchiale, rhinitis allergica, conjunctivitis allergica, and/or atopic Dermatitis. These diseases are also often associated with the occurrence of AD in a patient (Leung DYM et al, 2004).
  • AD The diagnosis of AD is made clinically and is based on historical features, morphology and distribution of skin lesions, and associated clinical signs.
  • Formal sets of criteria have been developed by various groups to aid in classification.
  • One of the most recognized sets of diagnostic criteria is the 1980 Hanifin and Rajka criteria, requiring that 3 of 4 major criteria and 3 of 23 minor criteria must be met to make the diagnosis (Hanifin JM and Rajka G, 1980).
  • Predilection sites for the eczema- tous efflorescences of AD are the face, neck and the flexural folds of the extremities. Although predilection sites exist, the skin lesions appear without clear borders, and practically every site of the body could be involved - up to an involvement of the whole integument (erythroderma).
  • the clinical pattern of AD also varies with age.
  • the disease may start on the scalp, thereafter spreads to the face and extensor surfaces of the arms and legs of toddlers, sometimes showing extensive ooz- ing and crusting.
  • the typical preferential pattern develops with eczematous involvement of flexures, neck and hands.
  • This is accompanied by dry skin and skin barrier dysfunction reflected by an increased transepidermal water loss and greater irritant skin response even involving nonlesional skin.
  • Lichenification is a result of scratching and rubbing. Most frequently in adults this may result in the prurigo type of AD with predominant excoriated nodular lesions. Exacerbations often start as increased itch without visible skin lesions. This is then followed by erythema, papules, and infiltration in acute skin lesions.
  • Chronic AD skin lesions have undergone tissue remodeling caused by chronic inflammation.
  • IL-4, IL-5, and IL-13 mR A-expressing cells When compared to normal skin or uninvolved skin of patients suffering from atopic dermatitis, acute skin lesions in atopic dermatitis has a significantly greater number of IL-4, IL-5, and IL-13 mR A-expressing cells.
  • Management comprises a disease adapted treatment combining adjuvant basic therapy (emollient use) and anti- inflammatory measurements. While in very severe cases a systemic treatment with drugs (e.g. systemic glucocorticoids, ciclosporin) or UV light may be indicated for a limited period of time, topical glucocorticoids are the mainstay of the treatment also AD, with their use being well established. However, there are considerable safety concerns associated with their use, particularly when they are applied continuously and/or in patients of young age who are even more susceptible to glucocorticoid side effects, be it local cutaneous side effects (e.g.
  • Allergic contact dermatitis is regarded as a classical example of a cell mediated hypersensitivity reaction.
  • This common inflammatory skin disease develops as a result of xenobiotic chemicals penetrating into the skin, chemically reacting with proteins, eventually resulting in a hapten- specific immune response. It is because of this well-defined localized immune response that the allergic signs and symptoms that are characteristic for the disease occur: skin redness, edema, warmth and pruritus (Gober MD et al, 2008).
  • the diagnosis is confirmed by diagnostic patch testing, a clinically useful test that reiterates the elicitation phase of Allergic contact dermatitis.
  • Allergic contact dermatitis is a distinct disease entity, with well-defined mechanisms of initiation, amplification, plateau phase and disease resolution (Gober MD et al, 2008). Although most environmental agents are too large to penetrate into the skin through the stratum corneum, some are of sufficiently low molecular weight to penetrate through this barrier. These molecules can be derived from naturally occurring substances, such as urushiol found in the resin of poison ivy, synthetic compounds and heavy metal ions. These compounds often are regarded as haptens, thus not being eligible to cause an allergic reaction on their own.
  • haptens For a sensitization reaction to occur, it is required that haptens interact with endogenous compounds (i.e. proteins) within the skin. Such a sensitization reaction has been referred to as immune recognition of 'altered self (Gober MD et al, 2008). That is, chemical alteration of self- molecules by xenobiotic haptens renders such self-molecules antigenic, in that this newly generated antigen (the hapten-modified self-molecule) can elicit a specific immune response.
  • endogenous compounds i.e. proteins
  • the mechanisms of allergic contact dermatitis involve a cascade of complex immune-mediated processes made up of two distinct phases in response to exposure to environmental chemicals, 1) the induction phase (also known as afferent or primary) and 2) the elicitation phase (also known as efferent or secondary phase).
  • haptens applied to the skin interact with cellular proteins to form hapten-protein complexes, the antigenic moiety recognized by the immune system.
  • These complexes are engulfed by antigen-presenting cells, such as dendritic cells, and presented in the context of MHC class II. This activates antigen-specific T-cells, which proliferate into memory T-cells.
  • NK T-cells are activated, leading to the release of cytokines including IL- 2, TNF- ⁇ and IL-4.
  • B-cells also become activated and release circulating IgM.
  • IgM interacts with the hapten-protein complex to induce complement activation, leading to the release of various inflammatory and chemotactic factors from mast cells and endothelial cells. Consequently antigen-specific CD8T-cells migrate to the site of hapten application and interact with local antigen-presenting cells, resulting in the clinical manifestations of allergic contact dermatitis.
  • the mixed lymphocytic infiltrate that can be observed consequently is the result of inflammatory cytokines, as well as cell-mediated cytotoxicity (Gober MD et al, 2008).
  • topical glucocorticoids are the mainstay of the treatment of allergic contact dermatitis, with their use being well established.
  • the present invention relates to compounds according to general formula (I) for use in the topical treatment of inflammatory skin diseases
  • R 1 , R 2 or R 3 according to formula (I) are different from hydrogen atom. In yet a further aspect, all three of R 1 , R 2 or R 3 according to formula (I) are different from hydrogen.
  • the alkyl groups of the compounds of formula (I) have 1-5 carbon atoms.
  • alkyl groups of the compounds of formula (I) have 1-3 carbon atoms.
  • the quinolon ring of formula (I) can be substituted by a group R 4 , selected from the group consisting of halogen, hydroxy, (Ci-C5)-alkyl, (Ci-C5)alkoxy, (Ci-C5)-alkylthio, (Ci-C5)-perfluoroalkyl, cyano group, nitro group, -NR 6 R 7 , -COOR 9 , (CO)NR 6 R 7 or a (Ci-C 5 -alkylene)-0-(CO)-(Ci-C 5 )- alkyl group, preferably R 4 is selected from the group Ci-C3-alkyl, Ci-C3-alkoxy, hydroxy, halogen.
  • R 4 is selected from the group hydrogen, Ci-C3-alkyl, halogen, hydroxy, preferably from hydrogen or halogen, more preferably from hydrogen, chlorine or fluorine.
  • Still another aspect of the invention are compounds according to formula (I), wherein R 4 is hydrogen or fluorine.
  • R 4 is fluorine. More particularly preferred compounds according to formula (I), wherein R 4 is a 7-fluoro- substituent or hydrogen and at least one of R 1 , R 2 or R 3 is selected from chlorine, fluorine, meth- oxy, hydroxy, R 5 is selected from S-CH2-CH3, -O-CH2-CH3, -S-CH3, -O-CH3-, N(CH 3 ) 2 , -OH and - CI.
  • R 4 is a 7- fluoro-substituent or hydrogen and at least one of R 1 , R 2 or R 3 is selected from chlorine, fluorine, methoxy, R 5 is selected from S-CH2-CH3, -O-CH2-CH3, -S-CH 3 , -O-CH3-, -OH and -CI.
  • R 9 is (Ci- Cio)-alkyl group or a (Ci-Cio)-alkoxy group.
  • Preferred aspect of the invention includes the subcombinations of all the residues as disclosed in the examples.
  • One aspect of the invention are compounds of general formula (I), wherein the phenyl group is substituted with 1-3 of the same or different substituents R 1 , R 2 or R 3 ,
  • R 1 and R 2 represent independently from each other, a hydrogen atom, a hydroxyl group, a halogen atom, a optionally substituted (Ci-Cio)-alkyl group, a optionally substituted (Ci-Cio)-alkoxy group, a (Ci-Cio)-alkylthio group, a (Ci-C5)-perfluoroalkyl group, a cyano group or a nitro group, or
  • R 6 und R 7 are, independently from each other, a hydrogen atom, Ci-Cs-alkyl or -(CO)-(Ci-C5)-alkyl
  • R 3 represents for a hydrogen atom, a hydroxyl group, a halogen atom, a cyano group, a optionally substituted (Ci-Cio)-alkyl group, a (Ci-Cio)-alkoxy group, a (C1-C10)- alkylthio group or a (Ci-C5)-perfluoroalkyl group,
  • any other phenyl group may be substituted by a group selected from Ci-C3-alkoxy, hydroxy, and halogen, in particular methoxy, hydroxy, fluorine, chlorine, or bromine.
  • R 5 of compounds according to general formula (I) is selected from -(Ci-Cio)-alkyl, which may be optionally partially or completely halogenated, -(C2-C10)- alkenyl, -(C 2 -Cio)-alkynyl, -(C3-C 7 )-cycloalkyl-(Ci-C8)-alkyl, -(C3-C 7 )-cycloalkyl-(C 2 -C8)-alkenyl, -(C3-C 7 )-cycloalkyl-(Ci-C8)-alkynyl, heterocyclyl-(Ci-C8)-alkyl, heterocyclyl-(C 2 -C8)-alkenyl, heterocyclyl-(C 2 -C8)-alkynyl-R 8 , R 8 -(Ci-C 8 )alkyl, R 8 -(C 2 -C8)alkyl, R 8 -(
  • R 5 of compounds according to general formula (I) (I) is se- lected from the group consisting of -(Ci-Cio)-alkyl, which may be optionally partially or completely halogenated, -(C 2 -Cio)-alkenyl, -(C 2 -Cio)-alkynyl, -(C 3 -C 7 )-cycloalkyl-(Ci-C8)alkyl, -(C 3 -C 7 )- cycloalkyl-(C 2 -C8)-alkenyl, -S-(Ci-Cio)-alkyl, -S0 2 -(Ci-Cio)-alkyl-S-R 8 , -CN, -Hal, -0-(Ci- Cio)alkyl, -NR 6 R 7 , (wherein R 6 and R 7 have the meaning defined above), and -OH with the exception of -
  • R 5 is most preferably selected from -OH, -S-CH 3 , -S-CH 2 -CH 3 , -O- CH 3 , -0-CH 2 -CH 3 or N(CH 3 ) 2 .
  • R 5 of compounds according to general formula (I) is selected from the group consisting of -(C 3 -C7)-cycloalkyl-(Ci-C8)-alkyl, -(C 3 -Cv)-cycloalkyl-(C 2 -C8)- alkenyl, -(C 3 -Cv)-cycloalkyl-(Ci-C8)-alkynyl, heterocyclyl-(Ci-C8)-alkyl, heterocyclyl-(C 2 -C8)- alkenyl, heterocyclyl-(C 2 -C8)-alkynyl-R 8 , R 8 -(Ci-C 8 )alkyl, R 8 -(C 2 -C 8 )alkenyl, R 8 -(C 2 -C8)alkynyl, - S-(Ci-Cio)-alkyl, -S0 2 -(Ci-Cio)-alkyl, -S
  • R 5 of compounds according to general formula (I) is selected from the group consisting of -(C 3 -C7)-cycloalkyl-(Ci-C8)-alkyl, -(C 3 -Cv)-cycloalkyl-(C 2 -C8)- alkenyl, -S-(Ci-Cio)-alkyl, -S0 2 -(Ci-Cio)-alkyl, -CN, -Hal, -0-(Ci-Cio)-alkyl, -NR 6 R 7 , (wherein R 6 and R 7 have the meaning defined above), -OH; preferably R 5 is -OH, -CI, -S-CH 3 , -S-CH 2 -CH 3 , -S- CH 2 -CH 2 -CH 3 , -0-CH 3 , -0-CH 2 -CH 3 , -0-CH 2 -CH 2 -CH 3 , N
  • R 5 of compounds according to general formula (I) is selected from the group consisting of -R 8 , -S-(Ci-Cio)-alkyl, -S0 2 -(Ci-Cio)-alkyl, -S-R 8 , -S0 2 -R 8 , -CN, -hal,
  • R 5 of compounds according to general formula (I) is selected from the group consisting of -S-(Ci-Cio)-alkyl, -S0 2 -(Ci-Cio)-alkyl, -CN, -hal, -0-(Ci-Cio)-alkyl, -
  • R 5 of compounds according to general formula (I) is selected from the group consisting of -S-(Ci-Cio)-alkyl, -S0 2 -(Ci-Cio)-alkyl, -NR 6 R 7 , (wherein R 6 and R 7 have the meaning defined above), and -OH.
  • R 5 of compounds according to general formula (I) is -(Ci- Cio)-alkyl or -(C 2 -Cio)-alkenyl.
  • R 5 is not -(Ci-Cio)-alkyl or -(C 2 -Cio)-alkenyl and from R 1 , R 2 or R 3 at least two are different from hydrogen or R 1 , R 2 or R 3 all are different from hydrogen and R 4 is halogen.
  • One aspect of the present invention are compounds of general formula (I), wherein
  • R 1 and R 2 represent independently from each other, a hydrogen atom, a hydroxyl group, a hal- ogen atom, a optionally substituted (Ci-Cio)-alkyl group, a optionally substituted
  • R 1 and R 2 represent independently from each other, NR 6 R 7 , wherein R 6 und R 7 are, independently from each other, a hydrogen atom, -Ci-Cs-alkyl or -(CO)-(Ci-C5)-alkyl,
  • R 3 represents for a hydrogen atom, a hydroxyl group, a halogen atom, a cyano group, a optionally substituted (Ci-Cio)-alkyl group, a (Ci-Cio)-alkoxy group, a (Ci-Cio)- alkylthio group or a (Ci-C5)-perfluoroalkyl group
  • R 4 represents for a hydrogen atom, a hydroxyl group, or a halogen atom
  • R 5 represents for a group, selected from the group consisting of -(Ci-Cio)-alkyl, which might be fully or partially substituted by a halogen atom,
  • R 8 represents for an aryl group, which might be substituted by 1 to 3 hydroxy groups, halogen, Ci-Cs-alkyl, Ci-Cs-alkoxy, cyano group, a heteroaryl group, whereby the heteroaryl group might comprise 1 to 3 heteroatoms, which optionally are substitut- ed by 1 to 3 alkyl groups, hydroxy, halogen, cyano group or Ci-Cs-alkoxy groups, and salts, solvates or salts of solvates thereof.
  • R 1 and R 2 independently represent, a hydrogen atom, hydroxy group, a halogen atom, an op- tionally substituted -(Ci-Cio) alkyl, an optionally substituted -(Ci-Cio)-alkoxy, (Ci-
  • R 6 and R 7 independently represents hydrogen atom, Ci-Cs-alkyl or -(CO)-(Ci-C5)-alkyl,
  • R 3 represents a hydrogen atom, a hydroxy group, a halogen atom, a cyano group, an optionally substituted -(Ci-Cio)-alkyl group, a -(Ci-Cio)-alkoxy group or a (C1-C5) perfluoroalkyl group,
  • R 4 represents a hydrogen atom, Ci-C 3 -alkyl, Ci-C 3 -alkoxy, a hydroxy group or a halogen atom,
  • R 5 represents a group selected from -(Ci-Cio)-alkyl, which are the optionally partially or completely halogenated is, -(C 2 -Cio)-alkenyl, -(C 2 -Cio)-alkynyl, -(C3-C7)- cycloalkyl-(Ci-C 8 )-alkyl, -(C 3 -C7)-cycloalkyl-(C 2 -C8)-alkenyl, -S-(Ci-Cio)-alkyl, -
  • R ⁇ ⁇ and R 3 independently represent hydrogen, fluorine, chlorine, bromine, a cyano group, a methoxy group, an ethoxy group or a hydroxy group,
  • R 4 represents hydrogen, Ci-C3-alkyl or halogen
  • R 5 represents a hydroxy group, chlorine, -S-CH 3 , -C-CH2-CH3, -CH-CH 2 -CH 2 -CH 3 ,
  • R ! , R 2 and R 3 independently represent hydrogen, fluorine, chlorine, bromine, a cyano group, a methoxy group, an ethoxy group or a hydroxy group,
  • R 4 represents hydrogen, Ci-C 3 alkyl or halogen
  • R 5 represents a hydroxy group, chlorine, -S-CH 3 , -S-CH 2 -CH 3 , -S-CH 2 -CH 2 -CH 3 , -O-
  • R ! , R 2 and R 3 independently represent hydrogen, fluorine, chlorine, bromine, a cyano group, a methoxy group, an ethoxy group or a hydroxyl group,
  • R 4 represents hydrogen, Ci-C 3 -alkyl or halogen
  • R 5 represents a hydroxy group, chlorine, -S-CH 3 , -S-CH 2 -CH 3 , -S-CH 2 -CH 2 -CH 3 , -
  • R 1 and R 2 independently represent hydrogen, fluorine, chlorine, a methoxy group or a hydroxy group
  • R 3 represents hydrogen, fluorine, chlorine, or methoxy group
  • R 4 represents hydrogen or fluorine
  • R 5 represents a hydroxy group, chlorine, -S-CH3, -S-CH2-CH3, -O-CH3, -O-CH2-
  • R 1 and R 2 independently represent hydrogen, fluorine, chlorine or a methoxy group
  • R 3 represents hydrogen, fluorine, chlorine, or methoxy group
  • R 4 represents hydrogen or fluorine
  • R 5 represents hydroxy group, chlorine, -S-CH 3 , -S-CH 2 -CH 3 , -0-CH 3 , -0-CH 2 -CH 3 , hydrogen atom, -CH 3 , or -CH 2 -CH 3 .
  • R 1 and R 2 independently represent fluorine, chlorine or a methoxy group
  • R 3 represents fluorine, chlorine, or methoxy group
  • R 4 represents hydrogen or fluorine
  • R 5 represents hydrogen atom, -CH 3 , or -CH 2 -CH 3 .
  • R 1 represent fluorine or chlorine
  • R 2 represent fluorine or chlorine
  • R 3 represents methoxy group
  • R 4 represents fluorine
  • R 5 represents hydrogen, chlorine, -S-CH 3 , -S-CH 2 -CH 3 , -0-CH 3 , -0-CH 2 -CH 3 .
  • R 1 represent fluorine or chlorine
  • R 2 represent fluorine or chlorine
  • R 3 represents methoxy group
  • R 4 represents fluorine
  • R 5 represents hydrogen atom, -S-CH 3 , -S-CH2-CH3, -0-CH 3 , -0-CH 2 -CH 3 ,
  • R 1 represent ortho-substitution, selected from fluorine or chlorine
  • R 2 represent meta-substitution, selected from fluorine or chlorine
  • R 3 represents para-substituted methoxy group
  • R 4 represents fluorine
  • R 5 represents hydrogen atom, -S-CH 3 , -S-CH 2 -CH 3 , -0-CH 3 , -0-CH 2 -CH 3 ,
  • R 1 represent ortho-substituted chlorine
  • R 2 represent meta-substituted fluorine
  • R 3 represents para-substituted methoxy group
  • R 4 represents fluorine
  • R 5 represents -S-CH 2 -CH 3 , or -0-CH 3 ,
  • the specific compounds may be in enantiomerically pure form and their salts, solvates or salts of solvates.
  • alkyl refers to a straight or branched, substituted or unsubsti- tuted chain.
  • propyl comprises n-propyl and iso-propyl
  • butyl com- prises n-butyl
  • iso-butyl and tert-butyl iso-butyl and tert-butyl.
  • the alkyl groups can be straight-chain or branched and stand e.g. for a methyl, ethyl, n-propyl, iso- propyl, n-butyl, iso-butyl, tert-butyl or n-pentyl group, or a 2,2-dimethylpropyl, 2-methylbutyl or 3- methylbutyl group.
  • a methyl or ethyl group is preferred. They can optionally be substituted by 1-3 hydroxy groups, cyano group, halogen, 1-3 Ci-Cs-alkoxy groups, and/or 1-3 -COO(Ci-Cio-alkyl or benzyl) groups. Preferred is hydroxy group.
  • the total number of substituents depends on the number of carbon atoms of the chain. Usually the number of substituents does not exceed the number of carbon atoms except for halogen which leads at a maximum number of substituents to e.g. perfluo- rated alkyl groups.
  • Ci-C3-alkyl group For a partially or completely fluorinated Ci-C3-alkyl group, the following partially or completely fluorinated groups are considered: fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, 1 , 1- difluoro ethyl, 1 ,2-difluoroethyl, 1 , 1 , 1-trifluoro ethyl, tetrafluoro ethyl, and pentafluoroethyl. Of the latter, the trifluoromethyl group or the pentafluoroethyl group is preferred.
  • the Ci-C5-alkoxy groups in R 1 , R 2 or R 3 and R 3 can be straight-chain or branched and stand for a methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, iso-butoxy, tert-butoxy or n-pentoxy, 2,2- dimethylpropoxy, 2-methylbutoxy or 3-methylbutoxy group.
  • a methoxy or ethoxy group is preferred. They can optionally be substituted by Ci-Cs-alkyl groups, cyano group or halogen.
  • the Ci-C5-alkylthio groups can be straight-chain or branched and stand for a methylthio, ethylthio, n-propylthio, iso-propylthio, n-butylthio, iso-butylthio, tert-butylthio or n-pentylthio, 2,2- dimethylpropylthio, 2-methylbutylthio or 3-methylbutylthio group.
  • a methylthio or ethylthio group is preferred.
  • halogen atom, hal or halogen means a fluorine, chlorine, bromine or iodine atom. Preferred is a fluorine, chlorine or bromine atom.
  • the NR 6 R 7 group includes, for example, -NH 2 , -N(H)CH 3 , -N(CH 3 ) 2 , -N(H)(CO)CH 3 , - N(CH 3 )(CO)CH 3 , -N[(CO)CH 3 ] 2 , -N(H)C0 2 CH 3 , -N(CH 3 )C0 2 CH 3 , or -N(C0 2 CH 3 ) 2 .
  • C 2 -C8-alkenyl is a straight or branched, substituted or unsubstituted, chain including isomers having an E- or Z-configurated double bond such as e.g. vinyl, propen-l-yl, propen-2-yl (Al- lyl), but-l-en-l-yl, but-l-en-2-yl, but-2-en-l-yl, but-2-en-2-yl, 2-methyl-prop-2-en-l-yl, 2-methyl- prop-l-en-l-yl, but-l-en-3-yl, but-3-en-l-yl.
  • alkenyl means alkenylene such as e.g. vinylene, propen-l-ylene, propen-2-ylene (Allylen), but-l-en-l-ylene, but-l-en-2-ylene, but-2-en-l-ylene, but-2-en-2-ylene, 2-methyl-prop- 2-en-l-ylene, 2-methyl-prop-l-en-l-ylene, but-l-en-3-ylen, but-3-en-l-ylene.
  • alkenylene such as e.g. vinylene, propen-l-ylene, propen-2-ylene (Allylen), but-l-en-l-ylene, but-l-en-2-ylene, but-2-en-l-ylene, but-2-en-2-ylene, 2-methyl-prop- 2-en-l-ylene, 2-methyl-prop-l-en-l-ylene, but-l-en-3-ylen, but-3-en-l-ylene.
  • C 2 -C8-alkynyl stands for a straight or branched chain e,g, -C ⁇ CH, -CH 2 -C ⁇ CH, -C ⁇ C- CH 3 , -CH(CH 3 )-C ⁇ CH, -C ⁇ C-CH 2 (CH 3 ), -C(CH 3 ) 2 -C ⁇ CH, -C ⁇ C-CH(CH 3 ) 2 , -CH(CH 3 )-C ⁇ C- CH 3 , -CH 2 -C ⁇ C-CH 2 (CH 3 ) or, if the alkynyl residue is placed between two other moieties the term alkynyl means alkynylene such as e.g.
  • C 3 -C7-cycloalkyl means a substituted or unsubstituted group selected from cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl.
  • the possible substitutents may be selected from hydroxy, halogen, (Ci-C 5 )-alkyl, (Ci-Cs)-alkoxy, NR 4 R 5 , COO(Ci-C 5 )-alkyl, CHO, cyano.
  • C 3 -C7-cycloalkyl-(Ci-Cio)-alkyl- means e.g. -(CH 2 )-cycloalkyl, -(C 2 H4)-cycloalkyl, - (C 3 H6)-cycloalkyl, -(C4H8)-cycloalkyl, -(C6Hio)-cycloalkyl whereby the cycloylkyl stand for e.g. cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl.
  • heterocyclyl means e.g. piperidinyl-, morpholinyl-, thiomorpholinyl-, piperazinyl-, tetra- hydro fur any 1-, tetrahydrothienyl-, imidazolidinyl- or pyrrolidinyl- whereby the heterocyclyl group may be bound via any possible ring atom.
  • the heterocyclyl group may be substituted by Ci-Cs-alkyl (optionally substituted), hydroxy-, Ci- C 5 -alkoxy-, NR 4 R 5 -, halogen, cyano-, COOR 8 -, CHO-. If possible these substitutens may also be bound to one of the free nitrogen atoms if any. N-oxides are also included in the definition.
  • heterocyclyl-(Ci-Cio)-alkenyl- means an alkylene group as defined above which is connected to the heterocyclyl group which also is already defined above.
  • heterocyclyl-(C 2 -C8)-alkenyl- means an alkylenylene group as defined above which is connected to the heterocyclyl group which also is already defined above.
  • aryl in the sense of the invention means aromatic or partially aromatic carbocyclic rings having 6 to 14 carbon atoms, e.g. phenyl and which may also may have a condensed a second or third ring such as e.g. napthyl or anthranyl. Further examples are phenyl, naphthyl, tetralinyl, an- thranyl, benzoxazinone, dihydroindolone, indanyl, and indenyl.
  • the aryl groups may be substituted at any position leading to a stable molecule by one or several substitutents, e.g. 1-3 substitutents, such as e.g. hydroxy, halogen, Ci-Cs-alkyl, Ci-Cs-alkoxy, cy- ano, -CF 3 , nitro, -COO(Ci-C5-alkyl or benzyl) or a heteroaryl group, preferably by 1-3 Ci-Cs-alkyl groups, hydroxyl, halogen, cyano or Ci-Cs-alkoxy.
  • the optionally substituted phenyl group is one aspect of the invention. Yet another aspect are the compounds of formula 1 whereby R 8 is not phenyl.
  • heteroaryl means an aromatic ring system having 1-3 heteroatoms selected from nitrogen, oxygen or sulfur, for five membered rings the maximum number of heteroatoms is three whereby only two oxygen or sulfur atoms are allowed provided that these two are not directly bound to each other. Possible heteroaryl rings are e.g.
  • the compounds according to formula (I) can exist in stereo isomeric forms such as enantiomers of diastereo isomers depending on their structure and residues as defined in Formula (I). In one aspect of the invention therefore all these enantiomers, diastereoisomers or mixtures thereof are encom- passed.
  • the isolation of enantiomerically or diastereomerically pure isomers can be done by methods of the state of the art, e.g. using column chromatography with a chiral solid phase.
  • Salts in the sense of the present invention are not only physiologically unobjectable salts but also salts which might be objectable for pharmaceutical use but which are useful e.g. during the process of isolation or purification.
  • physiologically unobjectable salts includes addition salts of mineral acids, carbonic acids, sulfonic acids, e.g. salts of hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, toluolsulfonic acid, benzenesulfonic acid, naphthalinesulfonic acid, acetic acid, trifluoro acetic acid, propionic acid, lactic acid, tartaric acid, malic acid, citric acid, fumaric acid, pivalic acid, maleic acid, succinic acid and benzoic acid.
  • mineral acids e.g. salts of hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, toluolsulfonic acid, benzenesulfonic acid, naphthalinesul
  • physiologically unobjectable salts includes salts of commonly suitable bases, e.g. salts of alkalimetall (e.g. sodium- and potassium salts), alkaline earth salts (e.g. calcium- and magnesium salts) and ammonium salts, derivatized from NH3 or organic amines with 1 to 16 carbon atoms, e.g.
  • alkalimetall e.g. sodium- and potassium salts
  • alkaline earth salts e.g. calcium- and magnesium salts
  • ammonium salts derivatized from NH3 or organic amines with 1 to 16 carbon atoms, e.g.
  • ethylamine, diethylamine, triethylamine, ethyldiisopropylamine monoethanola- mine, diethanolamine, triethanolamine, dicyclohexylamine, dimethylaminoethanol, prokaine, dibenzylamine, N-methylmorpholin, arginin, lysin, ethylendiamine and N-methylpiperidin.
  • Solvates in the sense of the invention are such forms of the compounds of the present combinations which build complexes by coordination of solvent molecules in a liquid or a solid phase. Hydrates are special forms of a solvate wherein water molecules are coordinated.
  • Salts in the sense of the present invention are not only physiologically unobjectable salts but also salts which might be objectable for pharmaceutical use but which are useful e.g. during the process of isolation or purification.
  • Benzaldehydes of type (II) can be condensed with substituted aminoquinolones of type (III) to imines of type (IV) using Lewis acids, preferably titanium alcoholates Ti(OR) 4 wherein R is Ci-C 4 - alkyl, such as e.g. tetraethyl orthotitanate or tetra tert. butyl orthotitanate and/or acidic conditions, e.g. organic acids such as acetic acid as reagents.
  • Suitable solvents are e.g. toluene, 1 ,4-dioxane or mixtures thereof. step b)
  • Imines of type (IV) are treated at low temperatures of -80° to -100° C. with the lithiated epoxide (V) to yield compounds of type (VI).
  • Suitable solvents are e.g. tetrahydrofurane, hexane, diethy- lether or mixtures thereof.
  • the epoxides (VI) can be opened by nucleophiles of type R 5 -Met to deliver compound (I).
  • Met means metal and includes alkalimetals e.g. sodium or lithium, alkaline earth metals such as e.g.
  • R 5 -Met e.g. alkyl- cuprates, vinylcuprates, thioles, allylsilanes, vinylsilanes, vinylstannanes, grignard compounds whereby R 5 is defined as in claim 1, which react in the presence of Lewis acids like e.g. BF 3 or AlMe3, AICI3.
  • Suitable solvents are e.g. diethylether, dimethylformamide, tetrahydroiurane.
  • the epoxides (VI) can also be opened directly by cyanides, amines, alcoholates, thioalcoholates, halogenides and even water or CS2CO3/H2O in the presence of bases or strong protic acids.
  • Suitable bases in the sense of the invention are e.g. CS2CO3, K2CO3 or NaOH.
  • Suitable strong protic acids are e.g. HCIO4, HC1 or HBr.
  • Methoxymethylamides of type (VII) are treated at low temperatures of -80° to -100° C. with the lithiated epoxide (V) to yield compounds of type (VIII).
  • Compounds of formula (VII) are commercially available or can be synthesized according to Branca et al, Chimia 49, 10; 1995, 381-385. step b)
  • VIII The epoxides (VIII) can be opened by nucleophils of type R 5 -Met to deliver compound (IX). Possible nucleophiles are alkylcuprates, vinylcuprates, thioles, allylsilanes, vinylsilanes, vinylstan- nanes, grignard compounds, in the presence of Lewis acids like BF3 or AUVfe, AICI3, or directly by cyanides, amines, alcohols, thioalcohols, halogenides and water in the presence of bases or strong protic acids. step c)
  • Ketones of type (IX) can be condensed with substituted aminoquinolones of type (III) to imines and subsequently or simultaneously reduced to the aminoalcohol I by a reductive amination using complex hydrides like e.g. NaBH 4 or LiAlH 4 (Katritzky et al. J. Org. Chem. 1995, 60, 7631-7640) or hydrogen in the presence of catalytic amounts of palladium or platinum or by application of an asymmetric organo cat lytic transfer hydrogenation (List et al. Angew. Chem. 2005, 117, 7590- 7593).
  • complex hydrides like e.g. NaBH 4 or LiAlH 4 (Katritzky et al. J. Org. Chem. 1995, 60, 7631-7640) or hydrogen in the presence of catalytic amounts of palladium or platinum or by application of an asymmetric organo cat lytic transfer hydrogenation (List et al. Angew. Chem.
  • intermediates of formula VI, VIII, IX can be purified at the step when they are obtained or compounds of formula I can be purified to obtain enantiomerically or diastereomerically pure end products after the complete reaction cascade. Examples for methods for obtaining enantiopure (enantiomerically pure) compounds are described below.
  • the separation of optical isomers can be performed by separation of one or more of the intermediates and/or separation of the end products. Usually separation of intermediates and separation of end products are alternatives as long as no racemisation had taken place during the production process.
  • the compounds according to the invention are present as racemic mixtures, they can be separated into pure, optically active forms according to the methods of racemate separation that are familiar to one skilled in the art.
  • the racemic mixtures can be separated by chromatography on an even optically active carrier material (CHIRALPAK AD ® ) into the pure isomers.
  • CHIRALPAK AD ® even optically active carrier material
  • chiral auxiliary agents as optically pure acids.
  • the free hydroxy group is esterified to yield a racemic compound of general formula (I) with an optically active acid and to separate the diastereoisomeric esters that are obtained by fractionated crystallization or by chroma- tography, and to saponify the separated esters in each case to the optically pure isomers.
  • one aspect of the invention is the process of obtaining compounds of formula (I) in diastere- omerically pure form, optionally using chromatography with columns containing chiral material or using chiral auxiliary agents.
  • Each of the intermediates of the synthesis of the compounds of formula (I) are one aspect of the present invention as well as especially their use for the synthesis of the compounds of formula (I).
  • a specific aspect of the invention are the concrete intermediates as used for the synthesis of the compounds of the examples, either as racemate or in their enantiomerically (having one chiral center) or diastereomerically (having two chiral centers) pure form.
  • Compound of formula I-A is known from WO 2009/065503 A (ex. 5 as racemate) and can be isolated from racemic form by chiral HPLC, Chiralpak IC 5 ⁇ with eluent hexane/ethanol (4 : 1), it might also be prepared by enantiomeric selective synthesis.
  • Compound of formula I-B is known from WO 2009/065503 A (ex. 7 as racemate) and can be isolated from racemic form by chiral HPLC, Chiralpak IC 5 ⁇ with eluent hexane/ethanol (4 : 1).
  • Compound of formula I-C is known from WO 2009/065503 A (ex. 3 as racemate) and can be isolated from racemic form by chiral HPLC, Chiralpak IC 5 ⁇ with eluent hexane/ethanol (4 : 1). Examples and experimental data
  • the compounds of general formula (I) for use in the topical treatment of T cell mediated inflamma- tory skin diseases according to the invention exert potent anti-inflammatory and immune- modulatory effects in a broad scope of in vitro activities which are related to T-cell mediated skin diseases such as psoriasis, atopic dermatitis or allergic contact dermatitis.
  • Compounds of the general formula (I) are able to inhibit the proliferation of stimulated human lymphocytes in a potent, dose-dependent manner.
  • the mixed leukocyte reaction is an ex vivo cellular immune assay examining lymphocyte proliferation in response to allogenic leukocytes (i.e. leukocytes sufficiently dissimilar in genotype to interact antigenically). It represents a standard in vitro assay for T cell immunity (McDevitt 2000), which is also well suited to determine immunomodulatory effects of added compounds.
  • Peripheral blood mononuclear cells (PBMCs) from two healthy donors were separated from citrate- anticoagulated whole blood. Untreated PBMCs from one donor were mixed with mitomycin C- treated allogeneic PBMC (stimulator cells) from the other donor at a ratio 1 :2 and cultured at 37°C with 5 % C0 2 .
  • the mixture of untreated responder and mitomycin-treated allogeneic stimulator PBMC (ratio 1 :2) were seeded onto 96-well plates (1.5 x 10 5 cells/well) and were incubated without test compounds (positive control showing the maximum achievable lymphocyte proliferation after stimulation by allogenic leucocytes) and with increasing concentrations of the compounds of the general formula (I) (concentration range: l .OE-12 to l .OE-6 mo 1/1).
  • [ 3 H] -thymidine was added for the last 6 hours of the 5 -day culture. Scintillation ⁇ -counting was used for determination of [ 3 H] -thymidine incorporation into the cell nucleus of the lymphocytes as a measure for alloreactive lymphocyte proliferation of responder cells.
  • the dose response usually forms a sigmoidal curve if the re- sponse (y-axis; here the magnitude of the lymphocyte proliferation) is depicted as a function of the increasing inhibitor concentration (x-axis; here: the increasing concentrations of the compounds of the general formula (I)), which serves to determine potency and efficacy of the individual compounds.
  • Efficacy is defined in this experimental setting as the measure of the maximum inhibition of lymphocyte proliferation which a compound can achieve).
  • the potency of a compound refers to the amount (concentration) of compound required to achieve a defined biological effect (in this case a defined inhibition of the lymphocyte proliferation).
  • a defined biological effect in this case a defined inhibition of the lymphocyte proliferation.
  • IC50 value which is the concentration of an inhibitor where the response is reduced by half (in this case: 50% inhibition of the lymphocyte proliferation). The smaller the compound concentration required for 50% inhibition, the more potent is the compound. All tested compounds of the general formula (I) are able to exert a dose-dependent inhibition of the proliferation of stimulated lymphocytes.
  • the compound (I-A) has been tested in parallel with two reference compounds, the very potent topical glucocorticoid, Clobetasol and the weaker topical glucocorticoid, Dexamethasone, which are examined for their inhibitory effects in the same concentration range as the compounds of the general formula (I).
  • the compound (I-A) (IC50: 1.91E-9 mol/1) is slightly less potent than Clobetasol (IC50: 8.0E-10 mol/1), but clearly more potent than Dexamethasone (IC50: 3.2E-8 mol/1) with similar efficacy for all three compounds (see also Table 4).
  • the compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases according to the present invention are able to inhibit the antigen-induced proliferation human lymphocytes in a mixed leukocyte reaction. In other words, the compounds show significant immunomodulatory activity.
  • Compounds of the general formula (I) are able to inhibit the release of cytokines which are related to the proliferation of a T-cell subset, the T-helper (Th) cells, related to the induction and perpetua- tion of psoriasis.
  • the manifestation of psoriasis is orchestrated by two subsets of pro -inflammatory CD4-positive T helper lymphocytes, the T helper (Th)17 and the Thl cells (Ghoreschi K et al, 2007).
  • the Thl7 cells a novel T-cell subset, have been implicated in the pathogenesis of psoriasis, while the Thl cells mainly contribute to the perpetuation of the inflammatory reaction in psoriasis.
  • the cytokine IL-23 stimulates the survival and proliferation of Thl 7 cells, and thus serves as a key cytokine regulator for this disease (Fitch E et al, 2007).
  • the cytokine IL-12 produced by activated monocytes/macrophages and dendritic cells, is the dominant factor promoting Thl cell polarization.
  • TNF-a produced by multiple cells, including activated Thl cells, does not induce proliferation of Thl or Thl7 cells, it is nevertheless a very prominent pro -inflammatory cytokine which can also be correlated to the pathophysiology of psoriasis (Victor FC et al, 2003). Accordingly, the attenuation of IL-23, IL-12 and TNF-a, should exert beneficial therapeutic effects in psoriasis (Numerof RP & Asadullah K, 2006).
  • PBMCs peripheral blood mononuclear cell
  • endotoxin LPS bacteria-derived lipopolysaccharides, which induce a strong immune response in animal and human cells
  • PBMC peripheral blood mononuclear cells
  • cytokine concentrations in supernatants of untreated cells and cells treated with increasing concentrations of test compounds were determined using a specific enzyme- linked immunosorbent assay (ELISA; an antibody-based detection system for the cytokines; TNFa- Biosource, IL-12p40 - R&D Systems) and a chemiluminescence assay (I GEN; suited for the sensi- tive detection of different ELISA reaction products).
  • ELISA enzyme- linked immunosorbent assay
  • I GEN chemiluminescence assay
  • the dose response relation also forms a sigmoidal curve if the response (y- axis; here the magnitude of the TNF-a, as well as the IL-12p40 release) is depicted as a function of the increasing inhibitor concentration (x-axis; here: the increasing concentrations of the compounds of the general formula (I)), which serves to determine potency and efficacy of the individual compounds.
  • Efficacy is defined in this experimental setting as the measure of the maximum inhibition of TNF-a or IL-12p40 release which a compound can achieve).
  • the potency of a compound refers to the amount (concentration) of compound required to achieve a defined biological effect (in this case a defined inhibition of the TNF-a or IL-12p40 release).
  • the IC50 value which is the concentration of an inhibitor where the response is reduced by half (in this case: 50% inhibition of the TNF-a or IL-12p40 release), is used for the comparison of the potencies of different compounds. The smaller the compound concentration required for 50% inhibition, the more potent is the compound.
  • the compound (I-A) (IL12p40 IC50: 7.5E-10 mol/1; TNF-a IC50: 6.5E-10 mol/1) is less potent than Clobetasol (IL12p40 IC50: 1.4E-10 mol/1; TNF-a IC50: 1.2E-10 mol/1), but more potent than Dexamethasone (IL12p40 IC50: 4.7E-9 mol/1; TNF-a IC50: 6.1E-9 mol/1) with similar efficacy for all three compounds.
  • Table 5 Table 5:
  • the experimental data also show that said compounds are also able to inhibit potently and effectively the secretion of TNF-a, which is a potent pro-inflammatory cytokine related to the pathophysiology of psoriasis. Accordingly, it seems reasonable to suppose that at least the compounds (I-C), (I-A) and (I-B) are able to exert beneficial therapeutic effects in psoriasis.
  • Compounds of the general formula (I) are able to inhibit the release of cytokines which are related to the proliferation of T-helper (Th) cells related to the induction and perpetuation of atopic derma- titis and allergic contact dermatitis.
  • atopic dermatitis is mainly caused by the cytokine IL-4 which initiates the proliferation of the Th2 subsets of pro-inflammatory CD4-positive T helper (Th) lymphocytes. These cells play a key role in the early phase of atopic diseases (Biedermann T et al, 2004).
  • Th T helper
  • IL-12 produced by infiltrating pro -inflammatory dendritic epidermal cells, macrophages and eosinophils, triggers a partial switch to a Thl -type cytokine milieu associated mainly with increased IFN- ⁇ expression (Leung DYM, 2000).
  • IL-4 seems also to contribute to the inflammatory reaction in allergic contact dermatitis
  • IL-2 the growth factor of Thl cytokines
  • IFN- ⁇ the growth factor of Thl cytokines
  • the plant lectin, phytohemagglutinine (PHA) acts as a T-cell mitogen, which does not only induce the proliferation of these cells but also induces the secretion of IL-2, IL-4 and IFN- ⁇ if it is incubated with human PBMCs.
  • This assay was used to determine the inhibitory activity of compounds of the general Formula (I) on the IL-2, 11-4 and IFN- ⁇ release.
  • PBMC were seeded onto 96-well plates (2.5 x 10 5 cells/well) and incubated for 24 h with 10 ⁇ / ⁇ PHA without (positive control showing the maximum achievable cytokine release) or with increasing concentrations of compounds of the general Formula (I) (concentration range: 10 "12 to 10 "6 mo 1/1).
  • cytokine concentrations in supernatants of untreated cells and cells 5 treated with increasing concentrations of test compounds were determined using a specific enzyme- linked immunosorbent assay kits from Biosource (ELISA; an antibody-based detection system for the cytokines) and chemiluminescence for the sensitive detection of different ELISA reaction products.
  • ELISA enzyme- linked immunosorbent assay kits from Biosource
  • chemiluminescence for the sensitive detection of different ELISA reaction products.
  • the dose response relation also forms a sigmoidal curve if the response (y- axis; here the magnitude of the IL-2, IL-4 or IFN- ⁇ release) is depicted as a function of the increasing inhibitor concentration (x-axis; here: the increasing concentrations of the compounds of the general formula (I)), which serves to determine potency and efficacy of the individual compounds I 15 the same way as it was done for the TNF-a or IL-12p40 release.
  • the compound of 20 example 8 is significantly less potent with an IL4 IC50 of 3.29E-8 mol/1, an IL-2 IC50 of 7.04E-08 mol/1 and an IFN- ⁇ IC50 of 3.31E-8.
  • Compounds (I-C), (I-A), and (I-B) were also very effective by inducing complete inhibition of the release of all cytokines at higher concentrations, while the compound (I-D) is significantly less effective as it did not even reach complete inhibition even at the highest tested concentration of 10 "6 mol/1.
  • the compound (I-A) (IL-4 IC50: 4.0E-10 mol/1; IL-2 IC50: 8.8E-10 mol/1, IFN- ⁇ IC50: 7.0E-10 mol/1) is less potent than Clobetasol (IL-4 IC50: 7.0E-11 30 mol/1; IL-2 IC50: 1.4E-10 mol/1, IFN- ⁇ IC50: 1.2E-10 mol/1), but more potent than Dexamethasone (IL-4 IC50: 2.3E-9 mo 1/1; IL-2 IC50: 4.6E-9 mo 1/1, IFN- ⁇ IC50: 3.8E-9 mo 1/1) with similar efficacy for all three compounds.
  • the compound (I-A) is also able to inhibit T-cell mediated inflammatory skin reactions in mouse and rat models in a potent and effective manner.
  • mice or rats a contact allergen such as dinitrochlorobenzene (DNCB) or dinitrofluorobenzene (DNFB) induces about 24 hours after challenge a Thl dominated response (Hayashi M et al, 2001).
  • DNCB dinitrochlorobenzene
  • DNFB dinitrofluorobenzene
  • the chal- lenge of the skin with DNFB leads to the recruitment of DNFB-specific T-cells to the skin which produce Thl cytokines, including IFN- ⁇ , IL-12 and IL-2 while the levels of Th2 cytokines are comparatively low (Takeshita K et al, 2004).
  • NMRI outbred mice or Wistar rats were sensitized on the shaved right flank with 0.5 % DNFB at day 0 and day 1. On day 5, the animals were challenged by topical application of 0.3 % DNFB onto the dorsal side of both ears.
  • Different dosages of the compound (I-A) (0.0001%, 0.001%, 0.01% and 0.1% w/vol) or vehicle were topically co-applied with the allergen challenge.
  • edema formation measured as increase in ear weight of ear punch biopsies of the challenged animals compared to the ear weight of ear punch biopsies of non-challenged animals
  • pro-inflammatory granulocyte infiltration into the ear skin measured by an increase of the granulocyte enzyme peroxidase in ear homogenates compared to the enzyme content in non-challenged ears.
  • aqueous supernatants of homogenized ear punch biopsies were incubated for 30 min with a peroxidase sensitive chromogen, Tetramethylbenzidine (TMB) dihy- drochloride.
  • TMB Tetramethylbenzidine
  • the extent of the peroxidase concentration in each sample was photometrically deter- mined. Changes in the optical density were monitored at 450 nm at 25 °C against the mixture of all solutions without the added sample homogenate. Absolute extinction numbers were used to express peroxidase activity. Both parameters were determined in animals sacrificed 24h after challenge.
  • the extent of the inflammatory reaction was characterized by the compound dose, which exerts almost complete, 80% (ED80) inhibition of the inflammatory skin reaction.
  • the compound (I-A) was able to inhibit the DNFB-induced allergic dermatitis reaction in a potent, dose dependent manner.
  • the dose response usually also forms a sigmoidal curve if the response (y-axis; here the magnitude of either edema formation or granulocyte infiltration measured via peroxidase activity) is depicted as a function of the increasing inhibitor dosages (x-axis; here: the increasing dosages [%] of the compound (I-A)), which serves to determine potency and efficacy of the given compounds.
  • the potency of a compound refers to the amount (dosage) of compound required to achieve a defined biological effect (in this case a defined inhibition of edema formation of granulocyte infiltration).
  • ED80 the Effective Dose which achieves 80% inhibition of the symptoms. The lower the compound dosage required for 80%> inhibition, the more potent is the compound.
  • mice Edema formation in mice was almost completely inhibited by the compound (I-A) at the very low dosage of 0.017%.
  • the compound (I-A) inhibits the DNFB-induced edema formation in mice by a factor of 6 more potent compared to that in rats (Rat Edema Formation - ED80: 0.097%).
  • the almost complete inhibition of the granulocyte infiltration in the ear skin of mice did only differ from that of rats by a factor of ⁇ 2 (Granulocyte Infiltration ED80: Mouse: 0.100% vs. Rat: 0.190%) (for all ED80 values see also table 7).
  • the compound (I-A) has been tested in parallel with a reference compound, the very potent topical glucocorticoid Clobetasol, which was examined for its inhibitory effects in the same concentration range as the compound (I-A). In rats the compound (I-A) did not differ in its inhibition of the DNFB-induced edema formation from that of Clobetasol (Rat Edema Formation - Compound (I- A): ED80: 0.097% vs.
  • Clobetasol ED80: 0.085%
  • Clobetasol inhibited the granulocyte infiltration in rats slightly more potent by a factor of ⁇ 3 (Rat Granulocyte Infiltration - Compound (I-A): ED80: 0.190% vs.
  • Clobetasol inhibited the DNFB-induced edema formation in mice with a factor of ⁇ 2 slightly less potent than the compound (I-A) (Mouse Edema Formation - Compound (I-A): ED80: 0.017% vs.
  • Clobetasol ED80: 0.040%), while both compounds attenuated the granulocyte infiltration in mice at a higher, but similar dosage (Mouse Granulocyte Infiltration - Compound (I-A): ED80: 0.100% vs. Clobetasol ED80: 0.107%) (for all ED80 values see also table 8).
  • TMA trimellitic anhydride
  • TDI tolylene 2,4-diisocyanate
  • mice or Wistar rats were sensitized by 5% TMA (mice: day 0; rats: day 0 and 4) or 1% TDI (rats: day 0) onto the shaved right flank.
  • the allergic contact dermatitis reaction was induced on day 5 by challenging the animals with a single application of 10% TMA or 1.5% TDI onto the dorsal side of both ears.
  • mice Edema formation of the TMA-induced reactions in mice is reached at an even lower dosage (Mouse Edema Formation - ED100: 0.001%), while the TMA-induced granulocyte infiltration was not yet reached at the highest tested dosage of 0.01% (for all ED80 / ED 100 values see also table 9).
  • ED Effective Dosage
  • the compound (I-A) has been tested in parallel with a reference compound, the very potent topical glucocorticoid Clobetasol, which was examined for its inhibitory effects in the same concentration range as the compound (I-A).
  • Compound (I-A) and Clobetasol reached almost similar, very low ED80 values for edema formation in the TMA-induced reaction in rats and mice (Mouse & Rat TMA-induced edema formation - Compound of Example 5 ED80: 0.00015% and 0.00052%, respectively vs. Clobetasol ED80: 0.00060% and 0.00077%, respectively).
  • the compound (I-A) is not only able to reduce Thl and Th2 cytokine release in human lymphocytes in vitro, but is also able to attenuate Thl and Th2-mediated inflammatory skin reaction in rodents in a very potent and effective manner.
  • the compounds for use in the topical treatment of inflammatory skin diseases according to the present invention and especially the compounds (I-A) are able to attenuate or even completely inhibit T-cell mediated inflammatory skin reactions in indications such as psoriasis, atopic dermatitis and allergic contact dermatitis.
  • the suitable dose varies and depends on, for example, the active strength of the compound of general formula I, the host, the type of administration, and the type and severity of the conditions that are to be treated, as well as the use as a prophylactic agent or therapeutic agent.
  • the invention provides:
  • a pharmaceutical composition for treating a dermatological diseases comprises one of the compounds according to the invention or mixture thereof and at least one pharmaceutical adjuvant and/or vehicle.
  • the daily doses comprise a range of 1 ⁇ g to 100,000 ⁇ g of the compound according to the invention per kg of body weight.
  • a recommended daily dose lies in the range of 1 ⁇ g to 100,000 ⁇ g per kg of body weight.
  • Preferred is a dose of 10 to 30,000 ⁇ g per kg of body weight, and more preferred is a dose of 10 to 10,000 ⁇ g per kg of body weight.
  • this dose is suitably administered several times daily.
  • individual doses can be given that are significantly above the above-mentioned doses.
  • This invention also relates to pharmaceutical compositions containing one of the forms of the compound of the formula (I) or a mixture thereof.
  • compositions may be utilized to achieve the desired pharmacological effect by administration to a patient in need thereof.
  • a patient for the purpose of this invention, is a mammal, including a human, in need of treatment for the particular condition or disease. Therefore, the present invention includes pharmaceutical compositions which are comprised of a pharmaceutically acceptable carrier and a pharmaceutically effective amount of one of the forms of the compound of the formula (I) or a mixture thereof.
  • a pharmaceutically acceptable carrier is any carrier which is relatively non-toxic and innocuous to a patient at concentrations consistent with effective activity of the active ingredient so that any side effects ascribable to the carrier do not vitiate the beneficial effects of the active ingredient.
  • a pharmaceutically effective amount of compound is that amount which produces a result or exerts an influence on the particular condition being treated.
  • the forms of the compound of the formula (I) of the present invention can be administered with pharmaceuti- cally-acceptable carriers well known in the art using any effective conventional dosage unit forms, including immediate, slow and timed release preparations, orally, parenterally, topically, nasally, ophthalmically, optically, sublingually, rectally, vaginally, and the like.
  • the form of the compound of the formula (I) of the present invention can be formulated into solid or liquid preparations such as solid dispersion, capsules, pills, tablets, troches, lozenges, melts, powders, solutions, suspensions, or emulsions, and may be prepared according to methods known to the art for the manufacture of pharmaceutical compositions.
  • the solid unit dosage forms can be a capsule which can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers such as lactose, sucrose, cal- cium phosphate, and corn starch.
  • the form of the compound of the formula (I) of the present invention may be tableted with conventional tablet bases such as lactose, sucrose and cornstarch in combination with binders such as acacia, corn starch or gelatin, disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch, alginic acid, corn starch, and guar gum, gum tragacanth, acacia, lubricants intended to improve the flow of tab- let granulation and to prevent the adhesion of tablet material to the surfaces of the tablet dies and punches, for example talc, stearic acid, or magnesium, calcium or zinc stearate, dyes, colouring agents, and flavouring agents such as peppermint, oil of wintergreen, or cherry flavouring, intended to enhance the aesthetic qualities of the tablets and make them more acceptable to the patient.
  • conventional tablet bases such as lactose, sucrose and cornstarch in combination with binders such as acacia, corn starch or gelatin, dis
  • Suitable excipients for use in oral liquid dosage forms include dicalcium phosphate and diluents such as water and alcohols, for example, ethanol, benzyl alcohol, and polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent or emulsifying agent.
  • Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance tablets, pills or capsules may be coated with shellac, sugar or both.
  • Dispersible powders and granules are suitable for the preparation of an aqueous suspension. They provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example those sweetening, flavouring and colouring agents described above, may also be present.
  • formulations in gels, ointments, fatty ointments, creams, pastes, powders, milk and tinctures are possible.
  • the dosage of the compounds of general formula I should be 0.01%-20% in these preparations to achieve a sufficient pharmacological action.
  • compositions of the invention can also contain other conventional pharmaceutically acceptable compounding ingredients, generally referred to as carriers or diluents, as necessary or desired. Conventional procedures for preparing such compositions in appropriate dosage forms can be utilized.
  • the multi-phase gel system comprises an exterior lipid matrix and an interior phase coagulated by means of polymer, which is obtained by
  • the multi-phase gel system described above which is preferably provided for application and infiltrate of hardly soluble at high concentration and/or hydrolysis-sensitive active ingredient in combination with the substance class of the compounds of the formula (I) in the Modification I, Modification II and/or Monohydrate, has unexpected advantageous properties.
  • the developed multi-phase gel system with the compound of the formula (I) in the Modification I, Modification II and/or Monohydrate is characterized by an excellent storage-stability over a period of at least 24 months.
  • the physical stability surprisingly outperforms the conventionally ointments and those parallel-developed corresponding cream formulations containing the same active ingredient.
  • the developed system is characterized by an excellent skin-compatibility. This is especially surprising, because the formulation does not contain insubstantial proportions of solvents such as propylene glycol and propylene carbonate. In studies on homo sapiens, the compatibility was identically good or even better than those of parallel-developed cream formulations containing a significantly small proportion of solvents.
  • the formulation expectedly exhibits a highly dermal availability in in vitro model in comparison to conventional systems.
  • a multi-phase gel system is referred to as a system is formed of two or more phases.
  • a preferred system has a lipid phase which contains skin-compatible lipids.
  • the lipids are selected from petrolatum, paraffin, beeswax, silicone oils and mixtures thereof.
  • a system is further preferred which contains polymers such as cellulose derivatives, acrylate polymers and their derivatives or mixture.
  • the cellulose derivative is hydroxypropyl cellulose.
  • the acrylate polymer is cross-linked acrylate.
  • a system is further preferred that swellable polymers or polymer mixtures are swelled by means of OH-group-containing swelling agents.
  • the swelling agent is multivalent aliphatic alcohols (polyols) having a chain length of up to 3 carbon atoms or mixtures thereof.
  • the polyol is propylene glycol. It is also preferred that the swelling agent further comprises carbonic acid diester or mixtures of carbonic acid diesters.
  • the carbonic acid diester is propylene carbonate.
  • the system is exceptionally preferred that the polymer phase contains the active ingredients. It is also preferred that the lipid phase and the polymer phase contain different active ingredients.
  • a preferred system further comprises one or more additives which are useful for a topically applicable composition.
  • Another object of the present invention is the application of the above-mentioned system for preparing a pharmaceutical composition for utilization on skin, the mucous membranes and/or on wound surfaces.
  • Preferred is the application for manufacturing a human or veterinary medical product, also use for humans and for animals.
  • Waterless solvents according to the invention may contain up to 5% water.
  • ethanol used in the present invention may contain up to 4.5% water (azeotrope).
  • the polymers which are swellable through OH group, may be selected from acrylate polymers or the mixtures thereof.
  • acrylate polymers or the mixtures thereof.
  • Several exemplary products from firm Noveon Inc. are referred to as: Carbopol 934 NF, Carbopol 934P NF, Carbopol 940 NF, Carbopol 97 IP NF, Carbo-pin 71G NF, Carbopol 974P NF, Carbopol 980 NF, Carbopol 981 NF, Carbopol 1342 NF, Carbopol 5984 EP, Pemulen TR-1 NF, Pemulen TR -2 NF, Noveon AA-1 USP, Noveon CA-1 USP and Noveon CA-2 USP.
  • short chain alcohols refer to mono- to tri valent aliphatic alcohols having up to five carbon atoms.
  • step b) for producing the dispersing polymer phase can be performed by conventional mixing systems and homogenizers.
  • composition according to the present invention may comprise further one or more useful additives for application as a topically applicable composition.
  • useful additives may be selected, for example, from dyes, odorous substances, preservatives and absorption-supporting agents.
  • the effective dosage of the compounds of this invention can readily be determined for treatment of each desired indication.
  • the amount of the active ingredient to be administered in the treatment of one of these conditions can vary widely according to such considerations as the particular compound and dosage unit employed, the mode of administration, the period of treatment, the age and sex of the patient treated, and the nature and extent of the condition treated.
  • the total amount of the active ingredient to be administered will generally range from about 0.0001 mg/kg to about 20 mg/kg, and preferably from about 0.001 mg/kg to about 2 mg/kg body weight per day.
  • a unit dosage may contain from about 0.05 mg to about 150 mg of active ingredient, and can be administered one or more times per day.
  • the specific initial and continuing dosage regimen for each patient will vary according to the nature and severity of the condition as determined by the attending diagnostician, the activity of the specific compound employed, the age and general condition of the patient, time of administration, route of administration, rate of excretion of the drug, drug combinations, and the like.
  • the desired mode of treatment and number of doses of a compound of the present invention or a pharmaceutically acceptable salt or ester or composition thereof can be ascertained by those skilled in the art using conventional treatment tests.
  • the compound of Formula I-A is also able to reduce psoriatic symptoms in the psoriasis plaque test, a standardized proof-of-concept model, which represents a useful method for predicting treat- ment efficacy in psoriasis in the early clinical stage of development.
  • the psoriasis plaque test was developed initially to compare simultaneously the anti- psoriatic activity of different topical glucocorticoids and the impact of different topical formulation types on their performance in a limited number of psoriasis patients (Dumas and Scholz, 1972, Katz M et al, 2000).
  • the psoriasis plaque test has been further modified and repre- sents now a valuable proof-of-concept model for the intra- individual comparison between different topical psoriasis treatments, which is also characterized by a low risk of adverse events as only a limited amount of the study drugs is applied to the psoriatic lesions (Wozel G, 2006, Queille- Roussel et al. 2013).
  • test field size 3 cm 2
  • the exact localiza- tion of each test field was confirmed at each visit using a stencil made from transparent foil during the test field determination prior to the first treatment.
  • the distance between two test fields was at least 2cm.
  • the anti-psoriatic activity was determined by grading the intensity of symptoms by the investigator's assessment. Recently skin thickening together with the typical in- flammatory cellular infiltration, which represent very prominent clinical signs of psoriasis (hereafter defined as "infiltrate thickness"), is mainly determined with 22 MHz sonography. This method represents a more sensitive efficacy measurement for the PPT compared to the clinical grading. Changes in skin thickness combined with the extent of inflammatory infiltration are depicted by an increase or decrease of an echo-poor band. Depending on the treatment success the size of the echo-poor band should increasingly decrease during the treatment period.
  • LS Least Square Method derived from Regression Analysis makes use of the best fit which minimizes the sum of squared residuals
  • the pairwise comparison over the treatment period of 29 day also shows that 0.1% compound of Formula I-A in the water-free multiphase system exerts - with a mean difference of the infiltrate thickness of 132 ⁇ compared to Clobetasol - the most potent anti-psoriatic activity, followed by the lower concentration, 0.01%, in the water- free multiphase system, with a difference of 170 ⁇ compared to the reference.
  • Both concentrations of compound of Formula I-A in the lipophilic cream formulation type were weaker with differences of infiltrate thickness of 182 ⁇ and 239 ⁇ compared to the reference.
  • topical glucocorticoids can not only induce local side effects such as skin atrophy, but can also frequently affect the Hypothalamic-pituitary-adrenal (HP A) axis and can cause disorders such as iatrogenic Cushing's syndrome, adrenal insufficiency, thymus atrophy and growth retardation or even growth inhibition during long-term therapy (Robertson DB and Maibach HI, 1982, Schaecke H et al, 2002).
  • the compound (I-A) differs from topical glucocorticoids as it is characterized by a significantly reduced or even absent potential to induce glucocorti- coid-related adverse events at therapeutically active dosages.
  • Skin atrophy i.e. the decrease in skin thickness
  • Juvenile hairless (hr/fir) rats represent an especially suited animal model for skin atrophy.
  • the skin fold thickness on day 25 compared to the skin fold thickness of day 1 (before the initiation of the treatment) served as an easily determined parameter for the impact of the topical treatment on skin thickness.
  • Skin breaking strength examined on day 25 with skin of sacrificed rats served as a sensi- tive parameter for the effects of the topical treatment on collagen fibers disintegration or degradation which also contributed to skin atrophy.
  • the compound (I-A) had also a more limited effect on the collagen disintegration than Clobetasol, as it reduced the skin breaking strength after 25 day of daily application to a lower extent compared to Clobetasol.
  • Table 12 The compound (I-A) had also a more limited effect on the collagen disintegration than Clobetasol, as it reduced the skin breaking strength after 25 day of daily application to a lower extent compared to Clobetasol.
  • ED80 Effective Dosage inducing 80% inhibition of Croton-oil induced
  • ED 100 Effective Dosage inducing complete inhibition of Croton-oil induced skin inflammation in rats.
  • the impact of the topical treatment on the growth of juvenile rats was monitored by periodically weighing the animals over time. Daily topical application of therapeutically active dosages of the compound (I-A) for 25 days had no impact on the growth of juvenile rats. The increase in animal weight did not differ between animals treated with the compound (I-A) and vehicle-treated rats over a period of the 25 days, while rats treated with therapeutically active dosages of Clobetasol showed significant growth retardation.
  • the daily application of the Clobetasol dosage which is able to induce complete inhibition of an inflammatory skin reaction after a single administration, vented rat weight gain during the 25 days treatment period completely (see also Table 13).
  • ED80 Effective Dosage inducing 80% inhibition of Croton-oil induced skin inflammation in rats
  • ED 100 Effective Dosage inducing complete inhibition of Croton-oil induced skin inflammation in rats.
  • the thymus which is the central organ for the constitution of the adaptive immune system, is a very glucocorticoid-sensitive organ. It responds to glucocorticoid exposure by rapid apoptotic death which might have fatal consequences on the adaptive immune system and might also lead to the induction of autoimmunity. Accordingly, thymus reduction is also an excellent indicator for systemic side effects of glucocorticoid-receptor ligands.
  • Glucocorticoids are also able to induce apoptosis of T and B lymphocytes, which can be measured by the weight reduction of secondary lymphatic organs such as the spleen. Suppression of the hypothalamic-pituitary-adrenal (HP A) axis is one of the most important side- effects of glucocorticoids as it affects the central neuro-endocrine circuit which is responsible for numerous physiological processes. Once endogenous Cortisol is replaced by exogenous glucocorticoids, Cortisol production, or production of precursors or inducers, is down-regulated. This is asso- ciated with a degeneration of the cortisol-producing organ, the adrenal gland.
  • HP A hypothalamic-pituitary-adrenal
  • HPA hypothalamic-pituitary-adrenal axis
  • ED80 Effective Dosage inducing 80% inhibition of Croton-oil induced
  • ED 100 Effective Dosage inducing complete inhibition of Croton-oil induced skin inflammation in rats.
  • the compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases according to the present invention have a reduced potential to induce unwanted effects on rodent skin after topical administration compared to the very potent topical glucocorticoid Clobeta- sol. This was demonstrated by a more limited effect on skin thickness and breaking strength than Clobetasol. Furthermore, the compounds for use in the topical treatment of inflammatory skin diseases according to the present invention do not reduce thymus, spleen, adrenal gland weight, while the topical glucocorticoid Clobetsol significantly reduced the weight of these immune organs. These observations clearly indicate an improved side effect profile of the said compounds compared to conventional topical glucocorticoids.
  • Hayashi M et al Assessment of Preferential Thl or Th2 Induction by Low-Molecular- Weight Compounds Using a Reverse Transcription-Polymerase Chain Reaction Method: Comparison of Two Mouse Strains, C57BL/6 and Balb/c. Toxicol. & Applied Pharmacol 177: 38-45, 2001.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Dermatology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention is directed to compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases such as psoriasis, atopic dermatitis, allergic contact dermatitis, wherein the said compounds are proprietary non-steroidal selective glucocorticoid receptor agonists according to general formula (I).

Description

QUINOLINE DERIVATIVES FOR TREATMENT OF INFLAMMATORY SKIN
DISEASES
Description
Technical field
The present invention relates to compounds for use in the topical treatment of inflammatory skin diseases such as psoriasis, atopic dermatitis, allergic contact dermatitis, wherein the said compounds are proprietary non-steroidal selective glucocorticoid receptor agonists (SEGRAs).
Background art
Glucocorticoids are commonly viewed as the most active topical compounds for the treatment of inflammatory skin diseases, but while they are well established in the treatment of psoriasis, atopic dermatitis and allergic contact dermatitis, their use is limited by their potential to induce a number of severe and sometimes irreversible local and/or systemic side effects (Hengge UR et al, 2006). Activation and repression of genes are mediated by different molecular mechanisms of the glucocorticoid receptor ("transactivation" and "transrepression"). This differentiation has provided a working model to search for novel synthetic glucocorticoid receptor- ligands that have a better therapeutic index than marketed glucocorticoids.
From the prior art of WO 2009/065503 Al (EP 2 234 979 Al), non-steroidal anti-inflammatory agents of the following general formula
Figure imgf000002_0001
are known, however the anti-inflammatory / immunomodulatory activities of these compounds on components of the immune system are not tested In particular, the effects by the use of these compounds for treatment of T-cell mediated inflammatory skin diseases is not established. Surprisingly compounds like e.g. (I-D), known form WO 2009/065503 which despite of potent binding to the Glucocorticoid receptor excerted extremely weak inhibition of T-cell mediated activities as shown by the present inventors. This is a clear indicator that compounds like e.g. (I-D) are not able to induce any anti-inflammatory or immunomodulatory activities in T-cell mediated skin diseases.
Therefore, compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases are still desired, wherein the said compounds are proprietary non-steroidal selective glucocorticoid receptor agonists (SEGRAs).
Considerable safety in the use of such compounds should be ensured, in particular when such com- pounds are administered to patients susceptible to glucocorticoid side effects, e.g. children and infants.
The present invention relates to compounds according to general formula (I) for use in the topical treatment of T-cell mediated inflammatory skin diseases,
Figure imgf000003_0001
formula (I) wherein
R1 and R2 represent independently from each other, a hydrogen atom, a hydroxyl group, a halogen atom, an optionally substituted (Ci-Cio)-alkyl group, an optionally substituted (Ci-Cio)-alkoxy group, a (Ci-Cio)-alkylthio group, a (Ci-C5)-perfluoroalkyl group, a cyano group or a nitro group, or
R1 and R2 stand together for a group, which is selected from the group consisting of -0-(CH2)p-0-, -0-(CH2)p-CH2-, -0-CH=CH-, -(CH2)p+2-, -NH-(CH2)p+i-, -N(Ci- C3-Alkyl)-(CH2)p+i-, and -NH-N=CH-, wherein p = 1 or 2 and the terminal oxygen atoms and/or carbon atoms and/or nitrogen atoms are connected to each other by neighbour ring carbon atoms, or
R1 and R2 represent independently from each other, NR6R7, wherein R6 und R7 are, independently from each other, a hydrogen atom, Ci-Cs-alkyl or -(CO)-(Ci-C5)-alkyl, R3 represents for a hydrogen atom, a hydroxyl group, a halogen atom, a cyano group, an optionally substituted (Ci-Cio)-alkyl group, a (Ci-Cio)-alkoxy group, a (C1-C10)- alkylthio group or a (Ci-C5)-perfluoroalkyl group,
R4 represents a hydrogen atom, a halogen atom, a hydroxyl group, a (Ci-C5)-alkyl group, a (Ci-C5)-alkoxy group, a (Ci-C5)-alkylthio group, a (Ci-C5)-perfluoroalkyl group, a cyano group, a nitro group, -NR6R7, -COOR9, -(CO)NR6R7 or a (C1-C5- alkylen)-0-(CO)-(Ci-C5)-alkyl group, wherein R6 and R7 have the same meaning as defined above and R9 represents a (Ci-Cio)-alkyl group or a (Ci-Cio)-alkoxy group, R5 represents a group selected from the group consisting of -(Ci-Cio)-alkyl, which might be fully or partially substituted by a halogen atom,
-(C2-C10)-alkenyl,
-(C2-Cio)-alkynyl,
-(C3-C7)-cycloalkyl-(Ci-C8)-alkyl,
-(C3-C7)-cycloalkyl-(Ci-C8)-alkyenyl,
-(C3-C7)-cycloalkyl-(C2-C8)-alkynyl,
heterocyclyl-(Ci-C8)-alkyl,
heterocyclyl-(Ci-C8)-alkenyl,
heterocyclyl-(C2-C8)-alkynyl,
-R8,
R8-(Ci-C8)-alkyl,
R8-(C2-C8)-alkenyl,
R8-(C2-C8)-alkynyl,
-S-(Ci-Cio)-alkyl,
-S02-(Ci-Cio)-alkyl
-S-R8,
-S02-R8,
-CN
-Hal, -0-(Ci-Cio)-alkyl,
-NR6R7, wherein R6 und R7 have the same meaning as defined above,
-O-R8,
-OH,
with the exception of -CH(CH3)2 or -C(CH3)=CH2
R8 represents an aryl group, which might be substituted by 1 to 3 hydroxy groups, halogen, Ci-Cs-alkyl, Ci-Cs-alkoxy, cyano group, CF3, nitro, -COO(Ci-C5-alkyl) or - C(0)OCH2-phenyl or a heteroaryl group, whereby the heteroaryl group might comprise 1 to 3 heteroatoms, which optionally are substituted by 1 to 3 alkyl groups, hydroxy, halogen, cyano or Ci-Cs-alkoxy groups,
and their salts, solvates or salts of solvates,
especially a compound of formula (I) selected from the group consisting of
5- {(l S,2R)[l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- ({methylsulfanyl}methyl)propyl]amino}-7-fluoro-lH-quinolin-2-one, 5- {[(l S,2S)-l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2-
(methoxymethyl)propyl]amino}-7-fluoro-lH-quinolin-2-one,
5- {(l S,2S)[l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2-
(hydroxymethyl)propyl]amino}-7-fluoro-lH-quinolin-2-one,
5- {[l-(5-chloro-3-fluoro-2-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- (hydroxymethyl)propyl]amino}-7-fluoro-lH-quinolin-2-one,
5- {[l-(2-chloro-3-fluoro-4-hydroxyphenyl)-3,3,3-trifluoro-2-hydroxy-2-
(hydroxymethyl)propyl]amino}-7-fluoro-lH-quinolin-2-one.
and their salts, solvates or salts of solvates,
Compounds of example (I-C) , (I-A) and (I-B), (I-D) and (I-E) according to formula (I) used for assays in the context of the present invention are provided below:
Compound (I-C): 5- {(l S,2R)[l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- ({methylsulfanyl}methyl)propyl]amino}-7-fluoro-lH-quinolin-2-one (I-C)
Compound (I-A) : 5- { [( 1 S,2S)- 1 -(2-chloro-3-fluoro-4-methoxyphenyl)-3 ,3 ,3-trifluoro-2-hydroxy-2- (methoxymethyl)propyl]amino}-7-fluoro-lH-quinolin-2-one (I-A) Compound (I-B): 5-{(lS,2S)[l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- (hydroxyrnethyl)propyl]amino}-7-fluoro-lH-quinolin-2-one (I-B)
Compound (I-D) : 5- { [ 1 -(5-chloro-3-fluoro-2-methoxyphenyl)-3 ,3 ,3-trifluoro-2-hydroxy-2- (hydroxymethyl)propyl]amino}-7-fluoro-lH-quinolin-2-one (I-D)
Compound (I-E): 5-{[l-(2-chloro-3-fluoro-4-hydroxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- (hydroxymethyl)propyl]amino}-7-fluoro-lH-quinolin-2-one (I-E)
Compound of formula I-A is known from WO 2009/065503 A (ex. 5 as racemate) and can be isolated from racemic form by chiral HPLC, Chiralpak IC 5 μιη with eluent hexane/ethanol (4 : 1). Compound of formula I-B is known from WO 2009/065503 A (ex. 7 as racemate) and can be iso- lated from racemic form by chiral HPLC, Chiralpak IC 5 μιη with eluent hexane/ethanol (4 : 1).
Compound of formula I-C is known from WO 2009/065503 A (ex. 3 as racemate) and can be isolated from racemic form by chiral HPLC, Chiralpak IC 5 μιη with eluent hexane/ethanol (4 : 1).
Compound of formula I-D is known from WO 2009/065503 A (ex. 8 as racemate) Compound of formula I-E is known from WO 2009/065503 A (ex. 14 as racemate)
Selective glucocorticoid receptor agonists
To demonstrate that the compounds of the general formula (I), preferably selected from the group consisting of
5-{(lS,2R)[l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- ({methylsulfanyl}methyl)propyl]amino}-7-fluoro-lH-quinolin-2-one,
5- { [( 1 S,2S)- 1 -(2-chloro-3-fluoro-4-methoxyphenyl)-3 ,3 ,3-trifluoro-2-hydroxy-2- (methoxymethyl)propyl]amino}-7-fluoro-lH-quinolin-2-one,
5-{(lS,2S)[l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- (hydroxymethyl)propyl]amino}-7-fluoro-lH-quinolin-2-one,
5-{[l-(5-chloro-3-fluoro-2-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- (hydroxymethyl)propyl]amino}-7-fluoro-lH-quinolin-2-one,
5-{[l-(2-chloro-3-fluoro-4-hydroxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- (hydroxymethyl)propyl]amino}-7-fluoro-lH-quinolin-2-one. act as selective glucocorticoid receptor agonists, a limited set of in vitro experiments were used: Fluorescence polarization allows the examination of molecular interactions by monitoring changes in the apparent size of fluorescently-labeled compounds. To determine the binding of ligands to a specific receptor the fluorescence polarization technology makes use of a fiuorescent ligand for a specific receptor. The technology is based on the observation that small fluorescent ligands after excitation with plane polarized light emit largely depolarized light as they tumble rapidly in the test solution, while large complexes of fluorescent ligands bound to their receptors rotate significantly slower. Accordingly, the emitted light of these complexes maintains the polarization of the exited light as it remains in the same plane as the excitation energy. The high polarization values of fluo- rescent ligand receptor binding complexes are decreased by the addition of increasing concentrations of non-fluorescently-labeled ligands which replace the fiuorescent ligand. These competitive binding studies are used to determine the concentration of a ligand of interest, in this case, of compounds of the general formula (I), required to reduce the specific binding of a fluorescent reference compound by 50% (inhibitory concentration of 50% or IC50 value). The lower the IC50 value of a ligand, the higher is its in vitro receptor binding affinity, since a lower concentration of the ligand is required to compete with the high affinity fluorescent reference for the receptor sites.
For the determination of the receptor binding of the compounds of the general formula (I) the commercially available fluorescence polarization assay from PanVera/Invitrogen was used. This kit contains the reagents necessary to perform a competition assay to assess the affinity of test compounds for the human glucocorticoid receptor. Recombinant human glucocorticoid receptor together with a stabilizing peptide is added to a fixed concentration (1.0E-9 mo 1/1) of a fluorescent glucocorticoid (Fluormone™ GS Red) ligand in the presence of different concentrations (1.6E-8, 8.0E-8, 4.0E-7, 2.0E-6, 1.0E-5 mo 1/1) of the test compounds in microwell plates. The plates are incubated for 2h at room temperature in the dark. The presence of effective competitors for the GR binding site prevents the formation of GS Red/GR complexes resulting in a decrease of the polarization value. At the end of the incubation period the polarization values are directly determined in the microplate. The shift in polarization values in the presence of test compounds is measured with an Analyst plate reader (LJL Biosystems/Molecular Devices Corporation) (excitation wavelength 530nm, emission wavelength 590 nM). The change in fluorescence polarization with increasing concentrations of non- fluorescent ligand is plotted versus the log of the ligand The IC50 values were calculated using the Microsoft® Excel-compatible XLfit® Four Parameter curve fitting tool. The compounds (I-C) , (I-A) and (I-B) see below) are able to replace the fluorescently-labeled ligand in a dose dependent manner. Similar to the reference compound, Clobetasol, with an IC50 value of 6.0E-9 mol/1, the compounds (I-C), (I-A), and (I-B), were also able to replace the fluorescent ligand in a potent manner with low IC50 values of 4.2E-9 mol/1, 3.1E-9 mol/1 and 7.1E-9 mol/1. This clearly shows that the compounds (I-C) , (I-A) and (I-B) have a very high affinity to the human glucocorticoid receptor (see Table 1).
Figure imgf000008_0001
Figure imgf000008_0002
Figure imgf000008_0003
Table 1 :
Figure imgf000009_0001
The selectivity of the compounds of general formula (I) towards the glucocorticoid receptor has been demonstrated in further steroid hormone receptor binding assays. The binding of the com- pounds of the general formula (I) to the progesterone receptor (PR), the androgen receptor (AR), the estrogen-alpha (ER-a) and the estrogen-beta (ER-β) receptor was examined in the presence of a stabilizing peptide with the fluorescence polarization technology. Fluomone™ PL Red (1.0E-9 mol/) was used as fluorescent ligand for the human PR, Fluomone™ AL Green (5.0E-10 mol/1) as fluorescent ligand for the rat AR, and Fluomone™EL Red (5.0E-10 mol/1) as fluorescent ligand for the ER-a and the ER-β. The fluorescently- labeled ligand receptor mixtures were incubated in micro well plates at room temperature in the dark for 1-6 h (PR), 4-6 h (AR), and 1-5 h (ER-a and ER ) in the presence of different concentrations (1.6E-8, 8.0E-8, 4.0E-7, 2.0E-6, l .OE-5 mol/1) of the compounds (I-A) and (I-B). At the end of the incubation period the polarization values are directly determined in the microplate. The shift in polarization values in the presence of test com- pounds is measured with an Analyst plate reader (LJL Biosystems/Molecular Devices Corporation) (for Fluomone™ PL Red and EL Red: excitation wavelength 530nm, emission wavelength 590 nM; for Fluomone™ AL Green: excitation wavelength 485 nm, emission wavelength 530 nM). The change in fluorescence polarization with increasing concentrations of non- fluorescent ligand is plotted versus the log of the ligand concentration. The IC50 values were calculated using the Microsoft® Excel-compatible XLfit® Four Parameter curve fitting tool.
The binding of the compound (I-A) and (I-B) to the human mineralocorticoid receptor (MR) was assessed using a ligand competition binding scintillation proximity assay (SPA). To this end, High Five cells, an insect cell line, were transfected with the human mineralocorticoid receptor ligand binding domain (NR3C2; aa729-964) with an N-terminal maltose binding protein (MBP) tag. Different concentrations (8.0E-9 mol/1, 4.0E-8 mol/1, 2.0E-7 mol/1 and 1.5E-6 mol/1) of the compound (I-A) were incubated with tagged MR binding domain-transfected High Five cell lysates and radio- actively- labeled, 3H-aldosterone (5.0E-9 mol/1) for one hour at room temperature. Thereafter anti- rabbit SPA polystyrene beads and rabbit anti-MBP antibodies were added. After a further incubation for 8 hours at room temperature the radioactive signal was detected with a LeadSeeker imaging system (GE Healthcare). The change in radioactive signal with increasing concentrations of the compound (I-A) is plotted versus the log of the ligand concentration. The IC50 values were calculated using the Microsoft® Excel-compatible XLfit® Four Parameter curve fitting tool.
In contrast to the potent binding to the glucocorticoid receptor, the compounds (I-A) and (I-B) do either not bind to the receptors of other steroid hormones or show only at very high concentrations a weak binding to the other steroid hormone receptors. Similar to the glucocorticoid Clobetasol, the compound (I-A) does not bind to the human estrogen-alpha receptor, the human estrogen-beta receptor, and the rat androgen receptor. 50% inhibition of the β-estradiol and the testosterone bind- ing, respectively, were even not reached in the very high concentration range of ~1.OE-5 mol/1. The compound (I-A) is also only able to bind very weakly to the human mineralocorticoid receptor. At the very high concentration of 1.2E-6 mol/1 the aldosterone binding is inhibited by 50%, which is an even by a factor of 5 weaker binding compared to Clobetasol. The comparison of the IC50 values for the binding of (I-A) to the human glucocorticoid and the human progesterone receptor shows that it binds by a factor of 20 weaker to the human progesterone receptor: similar to Clobetasol the compound (I-A) inhibits the progesterone binding only at a concentration of 6.1E-8 mol/1 by 50%. The compound of (I-B) does also not bind to the rat androgen receptor. It exerts compared to the IC50 value for the GR binding (IC50: 7.1E-9 mol/1) by a factor of -100 weaker binding to the human mineralocorticoid receptor (IC50: 7.7E-7 mol/1) and by a factor of 15 weaker binding to the human progesterone receptor (IC50: 1.1E-7 mol/1) (see also Table 2). Table 2:
Figure imgf000011_0001
hGR = human glucocorticoid receptor, hPR = human progesterone receptor, RAR = rat androgen receptor,
hMR = human mineralocorticoid receptor, hERa = human estrogen-alpha receptor, hERB = human estrogen-beta receptor
In summary, derived from the lacking or very weak binding of the compounds (I-A) and (I-B) to the human progesterone, mineralocorticoid, and estrogen receptor-alpha and -beta, as well as to the rat androgen receptor, it can be concluded that the compounds of the general formula (I) with their potent affinity to the glucocorticoid receptor are characterized by high binding selectivity towards this steroid hormone receptor.
Similar to other members of the steroid hormone receptor subfamily the glucocorticoid receptor is characterized by a ligand binding domain (LBD), which specifically recognizes and binds natural and artificial glucocorticoids and by a highly conserved DNA-binding motif (DBD) which recognizes specific DNA sequences, the so-called glucocorticoid response elements (GREs), which regulate gene transcription and lead thereby to the activation or suppression of a distinct subset of genes.
To clarify in an early and simple test system whether the compounds of the general formula (I) induce agonistic or antagonistic activities by their binding to the ligand binding domain of the glu- cocorticoid receptor, an artificial reporting system was used. The ligand binding domain of the glucocorticoid receptor as well as that of other members of the steroid hormone receptor family can be fused to the DNA binding domains of other transcription factors of different species like yeast using recombinant DNA technologies. A suited DNA binding domain is that of the yeast GAL4 transcription factor which is involved in transcriptional control of the galactose metabolism in yeast. The upstream activating sequences (UAS) which are recognized by the GAL4 transcription factor are not present in the mammalian genome. Thus, the introduction of GAL4 upstream activating sequences fused to a reporter gene like luciferase into the genome of a mammalian cell such as the CHO (Chinese Hamster Ovary) cells, allows the so-called transactivation of the luciferase gene by the GAL4 transcription factor. The additional fusion of the ligand binding domain of the glucocor- ticoid receptor leads to a chimera which transactivates the luciferase reporter gene upon binding of glucocorticoids. The expression of luciferase is measured by the addition of its substrate, luciferin. Luciferase converts luciferin to oxyluciferin. This process leads to the emission of light which is measured with a sensitive luminometer. The modified CHO cells were incubated at room temperature for 5-6h with either the compound (I-A), or the glucocorticoids Clobetasol or Dexamethasone, at 8 different concentrations in the range of 1.0E-9 to 1.0E-6 mo 1/1. After the incubation period the luciferase activity was determined using the Lumibox Video Camera system. Calculation of the EC50 values, the ligand concentration which leads to a half-maximum luciferase expression, was carried out with the GraphPad PRISM tool, a curve fitting program on the basis of non-linear regression. Via binding to the GR binding domain expressed in the transfected CHO cells, the compound (I-A) was able to induce luciferase expression in a potent and dose dependent way. The compound (I-A) achieved a maximum activation of the luciferase expression (-100% efficacy) which is comparable to the maximum activation induced by the reference glucocorticoids, Clobetasol, and Dexamethasone, The comparison of the EC50 values showed that the compound (I-A) is with an EC50 of 5.2E-9 mo 1/1 as potent as Dexamethasone (EC50: 6.0E-9 mo 1/1) and slightly less potent than Clobetasol (EC50: 1.1E-9 mo 1/1) (see also Table 3). Table 3:
Figure imgf000013_0001
These results clearly show that the compound (I-A) does not only bind to the glucocorticoid receptor but is also able to exert potent agonistic activities in the same concentration range as glucocorticoids.
To be on the safe side, the transfected CHO cell system was also used to check whether compound of the general formula (I) were able to exert antagonistic activities mediated via the GR. To this end transfected CHO cells were incubated with increasing concentrations of the compound (I-A) (1.0E-9 - 1.0E-5 mol/) in the presence of a constant concentration of Dexamethasone (6.0E-9 mol/1). It could be demonstrated that the Dexamethasone-induced half maximum luciferase expres- sion was not attenuated by the addition of increasing concentrations of the compound (I-A). Accordingly, the compound (I-A) was not able to exert antagonistic activities in the physiological concentration range. Only at unphysio logically high concentrations (>3.0E-6 mol/1) the luciferase signal was reduced, but this is more seen as a reflection of cell damage caused by the unphysio logical concentration of the compound (I-A), than as an expression of its antagonistic activity.
In a similar experimental setting using CHO cells transfected with the progesterone binding domain it was examined whether the compound (I-A), which bind weakly to the progesterone receptor (IC50: 6.1E-8 mol/) is able to exert agonistic activities mediated via the progesterone receptor binding. Via binding to the PR binding domain expressed in the transfected CHO cells, the com- pound (I-A) was only weakly able to induce luciferase expression. In comparison to progesterone the compound (I-A) even at the very high concentration of 1.0E-6 mol/1 was only able to induce a half maximum (50%) luciferase activation. The comparison of the EC50 values also showed that the compound (I-A) is with an EC50 of 2.5E-7 mol/1 significantly less potent than the reference progesterone (EC50: 3.2E-8 mol/1). With the transfected CHO cell system it could be clearly demonstrated that compounds of the general formula (I) exert potent GR-mediated agonistic activities. With the compound (I-A) the high selectivity toward the glucocorticoid receptor could again be demonstrated. Summary of the invention
Problem to be solved by the invention
The object of the present invention is to provide compounds for use in the topical treatment of T- cell mediated inflammatory skin diseases such as psoriasis, atopic dermatitis or allergic contact dermatitis in adults, adolescents, children and infants.
Compounds according to the present invention for use in the topical treatment of T-cell mediated inflammatory skin diseases are identified to achieve anti-inflammatory effects by attenuating the activity of cells belonging to the immune system after topical administration. At the same time these compounds should be characterized by a clear dissociation between highly beneficial antiinflammatory activities and undesired side effects.
Advantageous Effect of the Invention
The compounds according to the present invention are characterized by an optimized benefit-risk- ratio which means that their therapeutic effects are not or to an only minor extent accompanied by side effects (Schaecke H et al, 2004; Schaecke H et al, 2006; Schaecke H et al, 2007; Stahn et al, 2007). This represents a clear difference to the class of topical glucocorticoids where desired anti- inflammatory and immunosuppressive effects are often accompanied by severe or even irreversible side effects (e.g., diabetes mellitus, Cushing's syndrome, osteoporosis, skin atrophy, or glaucoma (Schaecke H et al. 2004 and Schaecke H et al. 2006). Accordingly, the compounds according to the present invention represent effective and safe topical treatment alternatives especially for inflammatory skin diseases. Considerable safety concerning the use of such compounds is provided, in particular when such compounds are administered to patients who are susceptible to glucocorticoid side effects, e.g. children and infants. Surprisingly compounds like e.g. (I-D), known form WO 2009/065503 which despite of potent binding to the Glucocorticoid receptor excerted extremely weak inhibition of T-cell mediated activities. This is a clear indicator that compounds like e.g. (I-D) are not able to induce any antiinflammatory or immunomodulatory activities in T-cell mediated skin diseases.
The present inventors had to carefully select , develop suitable assays in order to identify suitable compounds for T-cell mediated skin diseases.
Means for solving the problem
The present invention relates to compounds according to general formula (I) for use in the topical treatment of T-cell mediated inflammatory skin diseases,
Formula (I)
Figure imgf000015_0001
wherein
R1 and R2 represent independently from each other, a hydrogen atom, a hydroxyl group, a halogen atom, an optionally substituted (Ci-Cio)-alkyl group, an optionally substituted (Ci-Cio)-alkoxy group, a (Ci-Cio)-alkylthio group, a (Ci-C5)-perfluoroalkyl group, a cyano group or a nitro group, or
R1 and R2 stand together for a group, which is selected from the group consisting of -0-(CH2)p-0-, -0-(CH2)p-CH2-, -0-CH=CH-, -(CH2)p+2-, -NH-(CH2)p+i-, -N(Ci- C3-Alkyl)-(CH2)p+i-, and -NH-N=CH-, wherein p = 1 or 2 and the terminal oxygen atoms and/or carbon atoms and/or nitrogen atoms are connected to each other by neighbour ring carbon atoms, or
R1 and R2 represent independently from each other, NR6R7, wherein R6 und R7 are, independently from each other, a hydrogen atom, Ci-Cs-alkyl or -(CO)-(Ci-C5)-alkyl, R represents for a hydrogen atom, a hydroxyl group, a halogen atom, a cyano group, a optionally substituted (Ci-Cio)-alkyl group, a (Ci-Cio)-alkoxy group, a (Ci-Cio)- alkylthio group or a (Ci-C5)-perfluoroalkyl group,
represents for a hydrogen atom, a halogen atom, a hydroxyl group, a (Ci-C5)-alkyl group, a (Ci-C5)-alkoxy group, a (Ci-C5)-alkylthio group, a (Ci-C5)-perfluoroalkyl group, a cyano group, a nitro group, -NR6R7, -COOR9, -(CO)NR6R7 or a (C1-C5- alkylen)-0-(CO)-(Ci-C5)-alkyl group, wherein R6 and R7 have the same meaning as defined above and R9 represents a (Ci-Cio)-alkyl group or a (Ci-Cio)-alkoxy group, represents for a group, selected from the group consisting of -(Ci-Cio)-alkyl, which might be fully or partially substituted by a halogen atom,
-(C2-C10)-alkenyl,
-(C2-Cio)-alkynyl,
-(C3-C7)-cycloalkyl-(Ci-C8)-alkyl,
-(C3-C7)-cycloalkyl-(Ci-C8)-alkyenyl,
-(C3-C7)-cycloalkyl-(C2-C8)-alkynyl,
heterocyclyl-(Ci-C8)-alkyl,
heterocyclyl-(Ci-C8)-alkenyl,
heterocyclyl-(C2-C8)-alkynyl,
-R8,
R8-(Ci-C8)-alkyl,
R8-(C2-C8)-alkenyl,
R8-(C2-C8)-alkynyl,
-S-(Ci-Cio)-alkyl,
-S02-(Ci-Cio)-alkyl
-S-R8,
-S02-R8,
-CN
-Hal,
-0-(Ci-Cio)-alkyl,
-NR6R7, wherein R6 und R7 have the same meaning as defined above,
-O-R8,
-OH,
with the exception of -CH(CH3)2 or -C(CH3)=CH2 R8 represents for an aryl group, which might be substituted by 1 to 3 hydroxy groups, halogen, Ci-Cs-alkyl, Ci-Cs-alkoxy, cyano group, CF3, nitro, -COO(Ci-C5-alkyl) or -C(0)OCH2-phenyl or a heteroaryl group, whereby the heteroaryl group might comprise 1 to 3 heteroatoms, which optionally are substituted by 1 to 3 alkyl groups, hydroxy, halogen, cyano or Ci-Cs-alkoxy groups,
and their salts, solvates or salts of solvates
especially a compound of formula (I) selected from the group consisting of
5- {(l S,2R)[l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- ({methylsulfanyl}methyl)propyl]amino}-7-fluoro-lH-quinolin-2-one, 5- {[(l S,2S)-l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2-
(methoxymethyl)propyl]amino}-7-fluoro-lH-quinolin-2-one,
5- {(l S,2S)[l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2-
(hydroxymethyl)propyl]amino}-7-fluoro-lH-quinolin-2-one,
5- {[l-(5-chloro-3-fluoro-2-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- (hydro xymethyl)propyl] amino } -7-fluoro- lH-quinolin-2-one,
5- {[l-(2-chloro-3-fluoro-4-hydroxyphenyl)-3,3,3-trifluoro-2-hydroxy-2-
(hydroxymethyl)propyl] amino } -7-fluoro- lH-quinolin-2-one..
Preferably
5- { [( 1 S,2S)- 1 -(2-chloro-3-fluoro-4-methoxyphenyl)-3 ,3 ,3-trifluoro-2-hydroxy-2- (methoxymethyl)propyl] amino} -7-fluoro- lH-quinolin-2-one,
One aspect of the invention relates to compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases, characterized in that the said compounds are non-steroidal selective glucocorticoid receptor agonists.
Another aspect of the invention relates to compounds for use in the topical treatment of T-cell me- diated inflammatory skin diseases, characterized in that the said compounds are able to inhibit the proliferation of stimulated human lymphocytes in a potent and dose-dependent manner, demonstrated in a suited in vitro test system which made use of compound doses ranging between 1.0E- 12 and l .OE-6 mo 1/1 and showed potent inhibition of lymphocyte proliferation in the nanomolar range.
Still another aspect of the invention relates to compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases, characterized in that the said compounds are able to inhibit the release of cytokines such as IL-2, IL-4, and IL-23 which induce the proliferation of certain subsets of T cells associated with inflammatory reactions.
A further aspect of the invention relates to compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases, characterized in that the said compounds are able to inhibit the release of cytokines including IL-12, TNF-a or IFN-γ, which exert immuno stimulatory and proinflammatory effects.
Another aspect of the invention relates to compounds for use in the topical treatment of inflammatory skin diseases, characterized in that the inhibition of cytokine release and related proliferation of T cells is involved in the induction and perpetuation of psoriasis.
Still another aspect of the invention relates to compounds for use in the topical treatment of inflammatory skin diseases, wherein the said inflammatory skin diseases include psoriasis.
A further aspect of the invention relates to compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases, wherein the said skin disease, psoriasis, includes the sub- classes psoriasis vulgaris, plaque-type psoriasis, and pustular psoriasis.
Another aspect of the invention relates to compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases, wherein the said T-cell mediated inflammatory skin diseases include atopic dermatitis and allergic contact dermatitis.
Still another aspect of the invention relates to compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases, characterized in that the said compounds are able to inhibit T-cell induced inflammatory skin reactions in rodent models such as DNFB-induced or TMA- induced allergic contact dermatitis reactions.
Another aspect of the invention relates to compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases, characterized in that the said compounds are characterized ei- ther by a lack of or by a reduced level of side effects typically caused by topical glucocorticoids.
Still another aspect of the invention relates to compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases, characterized in that the said compounds are marked by a lack of or by a reduced level of side effects typically caused by topical glucocorticoids, which include skin atrophy, hypothalamic-pituitary-adrenal (HP A) axis suppression, thymus and spleen atrophy or growth retardation.
Another aspect of the invention relates to compounds for use in the topical treatment of T-cell me- diated inflammatory skin diseases, characterized in that the said compounds are used for T-cell mediated inflammatory skin diseases such as psoriasis, atopic dermatitis and allergic contact dermatitis in adults, adolescents, children and infants.
Still another aspect of the invention relates to compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases, wherein the said compounds do not have side effects or have a reduced level of side effects typically caused by topical glucocorticoids and can be used especially for inflammatory skin diseases in children and infants.
Another aspect of the invention relates to compounds for use in the topical treatment of inflammatory skin diseases, wherein the following compounds are particularly preferred:
5-{[(lS,2S)-l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifiuoro-2-hydroxy-2- (methoxymethyl)propyl]amino}-7-fluoro-lH-quinolin-2-one (I-A)
Figure imgf000019_0001
!-A
5-{(lS,2S)[l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifiuoro-2-hydroxy-2- (hydroxymethyl)propyl]amino}-7-fluoro-lH-quinolin-2-one (I-B)
Figure imgf000020_0001
5- {(l S,2R)[l-(2-chloro-3-fiuoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- ({methylsulfanyl}methyl)propyl]amino}-7-fluoro-lH-quinolin-2-one (I-C)
Figure imgf000020_0002
l-C In the following section, the T-cell mediated inflammatory skin diseases such as psoriasis, atopic dermatitis, and allergic contact dermatitis and compounds for use in topical treatment of these skin diseases are described in detail.
Detailed description of disease types Psoriasis
Psoriasis is a common chronic dermatosis and one of the most prevalent chronic inflammatory diseases, affecting approximately 2% of the population in western industrialized countries. The disease often persists for life and is regarded as very troublesome, leading to notable impact on a sufferer's quality of life. A reappraisal of the disease in recent years, which was also triggered by the high economic impact the disease has, led to the discovery of similarities with other inflammatory diseases, leading to psoriasis now being regarded as a model for other chronic inflammatory diseases (Nestle FO et al, 2009). Further, a broad spectrum of associated diseases has been identified in recent years. Psoriasis can occur rather early in life (ca. 70% of cases, starting prior to 40 years of age) and is then often referred to as Psoriasis Typ 1, or rather late in life, as Psoriasis Typ 2 (ca. 30% of cases, starting after 40 years of age). Typ 1 Psoriasis often comes with a positive family history for psoriasis, and is prominently associated with a number of genetic factors: so called "Psoriasis susceptibility loci" are located on different chromosomes (PSORS 1-9) and involve HLA-Cw6 (PSORS- 1, located at 6p21.3) and other genes important for inflammatory reactions, like TNF-a and IL-23 receptor variants, as well as various other factors (Nestle FO et al, 2009). Typ 2 psoriasis often shows no positive family history, and is less prominently associated with genetic factors. Often, these cases are less severely affected, but show a very chronic disease course with stationary plaques at predilection sites.
Psoriasis also shows a number of frequently occurring comorbidities: 1 out of 5 patients develops the so-called psoriatic arthritis, a seronegative spondyloarthropathy. Typical for this condition is an asymmetric involvement of small joints of the hands and feet, but also some large joints (e.g. knee or ankle) may be affected. Spondylitis and sacroiliitis may also be observed. Early recognition and treatment of psoriatic arthritis are mandatory as around 5% of these patients will develop a severe, mutilating (destructive) arthritis, leading to permanent disability. Further comorbidities that may occur with psoriasis are other immune-mediated inflammatory diseases like rheumatoid arthritis and Crohn's disease. In addition, the metabolic syndrome and the single components defining it (hypertonia, obesity, hyper lipidemia and diabetic-prone metabolic states) are observed 2 to 3 times more often in a psoriasis-affected population as compared to the overall population. This leads to elevated risks regarding conditions that are promoted by these factors, like coronary heart disease, myocardial infarction and stroke. Being a life-long condition, psoriasis requires a management that includes long-term therapy as well as an awareness regarding potentially occurring co-morbidities (Levine D et al, 2009).
Clinically, psoriasis can be further categorized into psoriasis vulgaris, or plaque-type psoriasis, and pustular psoriasis. Psoriasis vulgaris (plaque-type psoriasis) is the most common form of psoriasis, affecting up to 90%> of patients (Nestle FO et al, 2009). Erythro squamous plaques (red skin elevation with white or silvery scaling) with well-defined borders are the characteristic skin efflo- rescence of this disease type. Predilection sites are the extensor sites of the extremities (e.g. knee and elbow), along with the lower back, the scalp, the umbilical region and the genitoanal region. Deviations from this scheme are subtypes of psoriasis vulgaris that are defined by the anatomical region of affected skin, like palmoplantar psoriasis (involving palms of the hands and soles of the feet), psoriasis capitis (involving the scalp), psoriasis inversa and psoriasis intertriginosa (involving axillae, groin, submammar areas, skin folds (e.g. obese abdomen)). In mild forms, the erythro squamous plaques may be stable for a long time with a limited amount of body surface area involved, while in moderate or severe forms a sudden extension of affected body area takes place. The range of disease activity pattern thus involves a stable disease state as well as an undulating activity, where phases of stable disease activity (or even without any observable skin affection) take turns with phases of extensive involvement of the body surface area, up to an involvement of the complete integument (erythroderma, psoriasis involving the whole skin). Guttate psoriasis, with small erythro squamous plaques (up to the size of a coin) being organized in an exanthema- like pattern over wide areas of the body, also belongs into this group and is sometimes seen after infec- tions, leading to the hypothesis that this form of psoriasis is triggered by immunological pathways triggered by e.g. a bacterial infection.
Pustular psoriasis is a severe exudative inflammatory variant of psoriasis defined by the occurrence of pustules. Located on erythematous plaques, the pustules may also become confluent - resulting in larger pus-filled vesicles and bullae. The pustules contain pus that is sterile, as opposed to pus- tules known from other dermato logical diseases (e.g. folliculitis) were pathogens like Staphylococcus aureus are involved. In contrast to a pustule resulting from an adequate immunological reaction to an infection, the sterile pustules in this case are a symptom of the excessive influx of granulocytes from blood into outer skin layers, triggered by the psoriasis pathophysiology. This disease form is generally seen as an expression of high psoriatic disease activity and may occur as inde- pendent psoriasis subtype, or may be observed as an expression of high disease activity in a patient previously suffering from psoriasis vulgaris (psoriasis cum pustulatione). It can be seen in a defined, circumscribed areas, or as general pattern involving the whole integument (psoriasis pustu- losa generalisata, type "von Zumbusch"). It often is accompanied by general malaise, fever, myalgia and arthralgia. There are reports that associate this disease form with a sudden stop of a system- ic therapy with corticosteroids. A special form of this disease is the Psoriasis pustulosa palmoplantaris Type Barber-Konigsbeck, involving solely the palmae of the hands and plantae of the feet, triggering difficulties to walk and/or appropriately using the hands.
Elements of the innate as well as the adaptive immune system play crucial roles in the pathophysiology of psoriasis (Nestle FO et al, 2009). Mediated by activated To 11- like-receptors (TLR), keratinocytes of the skin produce antimicrobial substances that also exert immune-mo dulatory effects: Dendritic cells are attracted to migrate into the skin and are activated, while a subsequent interaction of dendritic cells and T-cells leads to the expansion of certain lymphocyte subpopula- tions in the skin. The transport of T cells from the dermis into the epidermis is a key event in psoriasis. It is controlled by the interaction of αΐ βΐ integrin on T-cells with collagen IV in the basement membrane of the psoriatic epidermis. Blocking of this interaction inhibits the development of psoriasis in clinically relevant models of psoriasis. Interleukin (IL) 12 is secreted by activated dendritic cells and leads to an expansion of the number of Thl -cells, a subset of T-cells, with the subsequent production of other pro -inflammatory cytokine. IL-12 is a heterodimeric cytokine encoded by two separate genes, coding for the subunits IL-12p35 and IL-12p40. The subunit IL-12p40 is also shared with IL-23 subunit that forms, together with IL-23pl9, the proinflammatory cytokine Interleukin 23, another heterodimeric proinflammatory cytokine. IL-23 propels the number of activated Thl 7 cells in the skin, promoting IL-17, IL-22, IL-26 and TNF-a (Biedermann T et al, 2004). The resulting pro -inflammatory milieu leads to a complex secondary, self-sustaining reaction with pro -inflammatory activation of keratinocytes as well as endothelial cells, leading to the influx of more dendritic cells, lymphocytes and neutrophilic granulocytes (sterile micro-abscesses). Finally, by interaction with keratinocytes and influence on their reproduction pattern, the clinically observable signs of psoriasis appear (erythema and plaque formation through inflammatory infiltration, scaling as sign of hyperproliferation of keratinocytes with disturbed cell differentiation).
There is a growing armamentarium of systemic treatment options for psoriasis, including metho- trexat, ciclosporin, fumaric acid esters, as well as UV light therapies. Pathogenesis-based approaches include TNF-a and Interleukin- 12p40 antagonists. However, these treatment options are foreseen for moderate to severely affected psoriasis patients, who represent only a minority of all patients suffering from the disease: Approximately 80% of patients affected with psoriasis have mild to moderate disease, and the majority of these patients are treated with topical agents. Despite a list of traditional topical treatment options (salicylic acid, anthralin, coal tar) and other available anti-psoriatic agents (vitamin D analogues, tazarotene, tacrolimus, pimecrolimus), it can be concluded that topical corticosteroids are still the cornerstone of treatment for the majority of patients with psoriasis, according to the American Academy of Dermatology's Guidelines of care for the management of psoriasis (Menter A et al, 2009). While it has been demonstrated for certain sys- temic drugs that countering of the activities of the proinflammatory cytokines IL-12p40 and TNF-a shows good anti-psoriatic activity, a topical therapy directed against the proinflammatory biological effects of these cytokines would be of special interest in the treatment of psoriasis.
Atopic Dermatitis
Atopic dermatitis (AD, synonyms: neurodermitis, atopic eczema, prurigo besnier, endogenous ec- zema) is an inflammatory, chronically relapsing, non-contagious and intensely pruritic dermatosis characterized by epidermal inflammation, itching, dry skin (with fine scaling) and exudation (in acute lesions). It is one of the most common skin diseases with a prevalence of 2-3% in the adult population, and up to 20% in the pediatric population, with an increasing prevalence in western countries over three decades (Leung DYM et al, 2004). AD usually follows a relapsing course and is often associated with elevated serum immunoglobulin (IgE) levels. According to current understanding, the pathophysiology of AD is the product of a complex interaction between various susceptibility genes, host environments, infectious agents, defects in skin barrier function, and immunologic responses. Most often, there is a positive family history for atopic diseases like asthma bronchiale, rhinitis allergica, conjunctivitis allergica, and/or atopic Dermatitis. These diseases are also often associated with the occurrence of AD in a patient (Leung DYM et al, 2004).
The diagnosis of AD is made clinically and is based on historical features, morphology and distribution of skin lesions, and associated clinical signs. Formal sets of criteria have been developed by various groups to aid in classification. One of the most recognized sets of diagnostic criteria is the 1980 Hanifin and Rajka criteria, requiring that 3 of 4 major criteria and 3 of 23 minor criteria must be met to make the diagnosis (Hanifin JM and Rajka G, 1980). Predilection sites for the eczema- tous efflorescences of AD are the face, neck and the flexural folds of the extremities. Although predilection sites exist, the skin lesions appear without clear borders, and practically every site of the body could be involved - up to an involvement of the whole integument (erythroderma). The clinical pattern of AD also varies with age. The disease may start on the scalp, thereafter spreads to the face and extensor surfaces of the arms and legs of toddlers, sometimes showing extensive ooz- ing and crusting. Later on, the typical preferential pattern develops with eczematous involvement of flexures, neck and hands. This is accompanied by dry skin and skin barrier dysfunction reflected by an increased transepidermal water loss and greater irritant skin response even involving nonlesional skin. Lichenification is a result of scratching and rubbing. Most frequently in adults this may result in the prurigo type of AD with predominant excoriated nodular lesions. Exacerbations often start as increased itch without visible skin lesions. This is then followed by erythema, papules, and infiltration in acute skin lesions. Chronic AD skin lesions have undergone tissue remodeling caused by chronic inflammation.
When compared to normal skin or uninvolved skin of patients suffering from atopic dermatitis, acute skin lesions in atopic dermatitis has a significantly greater number of IL-4, IL-5, and IL-13 mR A-expressing cells. Dendritic cells in lesional and, to a lesser extent, in nonlesional skin of patients suffering from the disease bear IgE molecules and together with mast cells contribute to the development of a mixed lymphoctic infiltration reaction, including Th2 cells, a subset of T- cells, expressing IL-4 and IL-13 mR A (Leung DYM et al, 2004). It is thought that after an initial phase with predominantly IL-4 producing Th2 cells, a subsequent phase begins that is characterized by IFN-γ producing Thl cells. This switch is thought to be initiated by the local production of IL-12 from infiltrating eosinophils and/or dendritic cells. Activated T-cells expressing Fas ligand have also been shown to induce keratinocyte apoptosis contributing to the spongiosis found in acute AD (Leung DYM et al, 2004).
For atopic dermatitis, there is no cure available at present time. Management comprises a disease adapted treatment combining adjuvant basic therapy (emollient use) and anti- inflammatory measurements. While in very severe cases a systemic treatment with drugs (e.g. systemic glucocorticoids, ciclosporin) or UV light may be indicated for a limited period of time, topical glucocorticoids are the mainstay of the treatment also AD, with their use being well established. However, there are considerable safety concerns associated with their use, particularly when they are applied continuously and/or in patients of young age who are even more susceptible to glucocorticoid side effects, be it local cutaneous side effects (e.g. skin atrophy, telangiectasias, hypopigmentation) or systemic effects (Hypothalamic-pituitary-adrenal axis (HP A) axis suppression, growth retardation, Cushing syndrome) Therefore, alternative, safer therapies with equal efficacy are needed (Ong PY, 2009). Allergic Contact Dermatitis
Allergic contact dermatitis is regarded as a classical example of a cell mediated hypersensitivity reaction. This common inflammatory skin disease develops as a result of xenobiotic chemicals penetrating into the skin, chemically reacting with proteins, eventually resulting in a hapten- specific immune response. It is because of this well-defined localized immune response that the allergic signs and symptoms that are characteristic for the disease occur: skin redness, edema, warmth and pruritus (Gober MD et al, 2008). The diagnosis is confirmed by diagnostic patch testing, a clinically useful test that reiterates the elicitation phase of Allergic contact dermatitis. The afferent phase of the disease develops gradually over time as a result of repeated, low-grade expo- sures of patients to the offending chemicals. Allergic contact dermatitis is a distinct disease entity, with well-defined mechanisms of initiation, amplification, plateau phase and disease resolution (Gober MD et al, 2008). Although most environmental agents are too large to penetrate into the skin through the stratum corneum, some are of sufficiently low molecular weight to penetrate through this barrier. These molecules can be derived from naturally occurring substances, such as urushiol found in the resin of poison ivy, synthetic compounds and heavy metal ions. These compounds often are regarded as haptens, thus not being eligible to cause an allergic reaction on their own. For a sensitization reaction to occur, it is required that haptens interact with endogenous compounds (i.e. proteins) within the skin. Such a sensitization reaction has been referred to as immune recognition of 'altered self (Gober MD et al, 2008). That is, chemical alteration of self- molecules by xenobiotic haptens renders such self-molecules antigenic, in that this newly generated antigen (the hapten-modified self-molecule) can elicit a specific immune response.
The mechanisms of allergic contact dermatitis involve a cascade of complex immune-mediated processes made up of two distinct phases in response to exposure to environmental chemicals, 1) the induction phase (also known as afferent or primary) and 2) the elicitation phase (also known as efferent or secondary phase).
During the afferent phase of allergic contact dermatitis, haptens applied to the skin interact with cellular proteins to form hapten-protein complexes, the antigenic moiety recognized by the immune system. These complexes are engulfed by antigen-presenting cells, such as dendritic cells, and presented in the context of MHC class II. This activates antigen-specific T-cells, which proliferate into memory T-cells. Further, NK T-cells are activated, leading to the release of cytokines including IL- 2, TNF-α and IL-4. In the presence of IL-4 and antigen, B-cells also become activated and release circulating IgM.
During the elicitation phase, IgM interacts with the hapten-protein complex to induce complement activation, leading to the release of various inflammatory and chemotactic factors from mast cells and endothelial cells. Consequently antigen-specific CD8T-cells migrate to the site of hapten application and interact with local antigen-presenting cells, resulting in the clinical manifestations of allergic contact dermatitis. The mixed lymphocytic infiltrate that can be observed consequently is the result of inflammatory cytokines, as well as cell-mediated cytotoxicity (Gober MD et al, 2008). Apart from the mandatory avoidance of antigens, topical glucocorticoids are the mainstay of the treatment of allergic contact dermatitis, with their use being well established. However, there are considerable safety concerns associated with their use, particularly when they are applied in patients who are susceptible to glucocorticoid side effects (e.g. due to young age), be it local cutaneous side effects (e.g. skin atrophy, telangiectasias, hypopigmentation) or systemic effects (HPA axis suppression, growth retardation, Cushing syndrome). Therefore, compounds with an optimized benefit-risk-ratio compared to marketed glucocorticoids are thought to be treatment alternatives for this indication.
Compounds
The present invention relates to compounds according to general formula (I) for use in the topical treatment of inflammatory skin diseases,
Formula (I)
Figure imgf000027_0001
The preferred compounds with definition of substitutions are disclosed as follows:
Compounds of general Formula (I), wherein at least one of R1, R2 or R3 is different from hydrogen are one preferred embodiment of the invention.
In another aspect, two of R1, R2 or R3 according to formula (I) are different from hydrogen atom. In yet a further aspect, all three of R1, R2 or R3 according to formula (I) are different from hydrogen.
In one aspect of the invention the alkyl groups of the compounds of formula (I) have 1-5 carbon atoms.
In another aspect the alkyl groups of the compounds of formula (I) have 1-3 carbon atoms.
The quinolon ring of formula (I) can be substituted by a group R4, selected from the group consisting of halogen, hydroxy, (Ci-C5)-alkyl, (Ci-C5)alkoxy, (Ci-C5)-alkylthio, (Ci-C5)-perfluoroalkyl, cyano group, nitro group, -NR6R7, -COOR9, (CO)NR6R7or a (Ci-C5-alkylene)-0-(CO)-(Ci-C5)- alkyl group, preferably R4 is selected from the group Ci-C3-alkyl, Ci-C3-alkoxy, hydroxy, halogen. In another aspect of the invention, R4 is selected from the group hydrogen, Ci-C3-alkyl, halogen, hydroxy, preferably from hydrogen or halogen, more preferably from hydrogen, chlorine or fluorine.
Still another aspect of the invention are compounds according to formula (I), wherein R4 is hydrogen or fluorine.
Yet another aspect of the invention are compounds according to formula (I), wherein R4 is fluorine. More particularly preferred compounds according to formula (I), wherein R4 is a 7-fluoro- substituent or hydrogen and at least one of R1, R2 or R3 is selected from chlorine, fluorine, meth- oxy, hydroxy, R5 is selected from S-CH2-CH3, -O-CH2-CH3, -S-CH3, -O-CH3-, N(CH3)2, -OH and - CI.
In another aspect of the invention are compounds according to formula (I), wherein R4 is a 7- fluoro-substituent or hydrogen and at least one of R1, R2 or R3 is selected from chlorine, fluorine, methoxy, R5 is selected from S-CH2-CH3, -O-CH2-CH3, -S-CH3, -O-CH3-, -OH and -CI.
In still another aspect of the invention are compounds according to formula (I), wherein R9 is (Ci- Cio)-alkyl group or a (Ci-Cio)-alkoxy group.
Preferred aspect of the invention includes the subcombinations of all the residues as disclosed in the examples.
One aspect of the invention are compounds of general formula (I), wherein the phenyl group is substituted with 1-3 of the same or different substituents R1, R2 or R3,
wherein
R1 and R2 represent independently from each other, a hydrogen atom, a hydroxyl group, a halogen atom, a optionally substituted (Ci-Cio)-alkyl group, a optionally substituted (Ci-Cio)-alkoxy group, a (Ci-Cio)-alkylthio group, a (Ci-C5)-perfluoroalkyl group, a cyano group or a nitro group, or
R1 and R2 stand together for a group, which is selected from the group consisting of -0-(CH2)p-0-, -0-(CH2)p-CH2-, -0-CH=CH-, -(CH2)P+2-, -NH-(CH2)P+i, -N(Ci- C3-Alkyl)-(CH2)P+i, and -NH-N=CH-, wherein p = 1 or 2 and the terminal oxygen atoms and/or carbon atoms and/or nitrogen atoms are connected to each other by neighbour ring carbon atoms, or
represent independently from each other, NR6R7, wherein R6 und R7 are, independently from each other, a hydrogen atom, Ci-Cs-alkyl or -(CO)-(Ci-C5)-alkyl, R3 represents for a hydrogen atom, a hydroxyl group, a halogen atom, a cyano group, a optionally substituted (Ci-Cio)-alkyl group, a (Ci-Cio)-alkoxy group, a (C1-C10)- alkylthio group or a (Ci-C5)-perfluoroalkyl group,
In another aspect of the invention, any other phenyl group may be substituted by a group selected from Ci-C3-alkoxy, hydroxy, and halogen, in particular methoxy, hydroxy, fluorine, chlorine, or bromine.
In a further aspect of the invention, R5 of compounds according to general formula (I) is selected from -(Ci-Cio)-alkyl, which may be optionally partially or completely halogenated, -(C2-C10)- alkenyl, -(C2-Cio)-alkynyl, -(C3-C7)-cycloalkyl-(Ci-C8)-alkyl, -(C3-C7)-cycloalkyl-(C2-C8)-alkenyl, -(C3-C7)-cycloalkyl-(Ci-C8)-alkynyl, heterocyclyl-(Ci-C8)-alkyl, heterocyclyl-(C2-C8)-alkenyl, heterocyclyl-(C2-C8)-alkynyl-R8, R8-(Ci-C8)alkyl, R8-(C2-C8)alkenyl, R8-(C2-C8)alkynyl, -S-(Ci- Cio)-alkyl, -S02-(Ci-Cio)-alkyl-S-R8, -SO2-R8, -CN, -Hal, -0-(Ci-Cio)alkyl, -NR6R7, (wherein R6 and R7 have the meaning defined above), -O-R8 and -OH with the exception of -CH(CH3)2, or - C(CH3)=CH2.
In yet another aspect of the invention, R5 of compounds according to general formula (I) (I) is se- lected from the group consisting of -(Ci-Cio)-alkyl, which may be optionally partially or completely halogenated, -(C2-Cio)-alkenyl, -(C2-Cio)-alkynyl, -(C3-C7)-cycloalkyl-(Ci-C8)alkyl, -(C3-C7)- cycloalkyl-(C2-C8)-alkenyl, -S-(Ci-Cio)-alkyl, -S02-(Ci-Cio)-alkyl-S-R8, -CN, -Hal, -0-(Ci- Cio)alkyl, -NR6R7, (wherein R6 and R7 have the meaning defined above), and -OH with the exception of -CH(CH3)2, or -C(CH3)=CH2. Preferably R5 is -OH, -CI, -S-CH3, -S-CH2-CH3, -S-CH2- CH2-CH3, -O-CH3, -0-CH2-CH3, -0-CH2-CH2-CH3, N(CH3)2, NHCH3 with the exception of - CH(CH3)2, or -C(CH3)=CH2. R5 is most preferably selected from -OH, -S-CH3, -S-CH2-CH3, -O- CH3, -0-CH2-CH3 or N(CH3)2.
In another aspect of the invention, R5 of compounds according to general formula (I) is selected from the group consisting of -(C3-C7)-cycloalkyl-(Ci-C8)-alkyl, -(C3-Cv)-cycloalkyl-(C2-C8)- alkenyl, -(C3-Cv)-cycloalkyl-(Ci-C8)-alkynyl, heterocyclyl-(Ci-C8)-alkyl, heterocyclyl-(C2-C8)- alkenyl, heterocyclyl-(C2-C8)-alkynyl-R8, R8-(Ci-C8)alkyl, R8-(C2-C8)alkenyl, R8-(C2-C8)alkynyl, - S-(Ci-Cio)-alkyl, -S02-(Ci-Cio)-alkyl-S-R8, -S02-R8, -CN, -hal, -0-(Ci-Cio)alkyl, -NR6R7, (wherein R6 and R7 have the meaning defined above), -O-R8 and -OH.
In yet another aspect of the invention, R5 of compounds according to general formula (I) is selected from the group consisting of -(C3-C7)-cycloalkyl-(Ci-C8)-alkyl, -(C3-Cv)-cycloalkyl-(C2-C8)- alkenyl, -S-(Ci-Cio)-alkyl, -S02-(Ci-Cio)-alkyl, -CN, -Hal, -0-(Ci-Cio)-alkyl, -NR6R7, (wherein R6 and R7 have the meaning defined above), -OH; preferably R5 is -OH, -CI, -S-CH3, -S-CH2-CH3, -S- CH2-CH2-CH3, -0-CH3, -0-CH2-CH3, -0-CH2-CH2-CH3, N(CH3)2, -NHCH3, R5 is most preferably -OH, -S-CH3, -S-CH2-CH3, -0-CH3, -0-CH2-CH3 and N(CH3)2.
In yet another aspect of the invention, R5 of compounds according to general formula (I) is selected from the group consisting of -R8, -S-(Ci-Cio)-alkyl, -S02-(Ci-Cio)-alkyl, -S-R8, -S02-R8, -CN, -hal,
-0-(Ci-Cio)-alkyl, -NR6R7, (wherein R6 and R7 have the meaning defined above), -O-R8 and -OH.
In another aspect of the invention, R5 of compounds according to general formula (I) is selected from the group consisting of -S-(Ci-Cio)-alkyl, -S02-(Ci-Cio)-alkyl, -CN, -hal, -0-(Ci-Cio)-alkyl, -
NR6R7, (wherein R6 and R7 have the meaning defined above), and -OH.
In another aspect of the invention, R5 of compounds according to general formula (I) is selected from the group consisting of -S-(Ci-Cio)-alkyl, -S02-(Ci-Cio)-alkyl, -NR6R7, (wherein R6 and R7 have the meaning defined above), and -OH.
In still another aspect of the invention, R5 of compounds according to general formula (I) is -(Ci- Cio)-alkyl or -(C2-Cio)-alkenyl.
Another aspect of the present invention are compounds of general formula (I) as above defined, wherein R5 is not -(Ci-Cio)-alkyl or -(C2-Cio)-alkenyl and from R1, R2 or R3 at least two are different from hydrogen or R1, R2 or R3 all are different from hydrogen and R4 is halogen.
One aspect of the present invention are compounds of general formula (I), wherein
R1 and R2 represent independently from each other, a hydrogen atom, a hydroxyl group, a hal- ogen atom, a optionally substituted (Ci-Cio)-alkyl group, a optionally substituted
(Ci-Cio)-alkoxy group, a (Ci-Cio)-alkylthio group, a (Ci-C5)-perfluoroalkyl group, a cyano group or a nitro group, or
R1 and R2 stand together for a group, which is selected from the group consisting of -0-(CH2)p-0-, -0-(CH2)p-CH2-, -0-CH=CH-, -(CH2)p+2-, -NH-(CH2)p+i, -N(Ci- C3-Alkyl)-(CH2)p+i, and -NH-N=CH-, wherein p = 1 or 2 and the terminal oxygen atoms and/or carbon atoms and/or nitrogen atoms are connected to each other by neighbour ring carbon atoms, or
R1 and R2 represent independently from each other, NR6R7, wherein R6 und R7 are, independently from each other, a hydrogen atom, -Ci-Cs-alkyl or -(CO)-(Ci-C5)-alkyl,
R3 represents for a hydrogen atom, a hydroxyl group, a halogen atom, a cyano group, a optionally substituted (Ci-Cio)-alkyl group, a (Ci-Cio)-alkoxy group, a (Ci-Cio)- alkylthio group or a (Ci-C5)-perfluoroalkyl group,R4 represents for a hydrogen atom, a hydroxyl group, or a halogen atom, R5 represents for a group, selected from the group consisting of -(Ci-Cio)-alkyl, which might be fully or partially substituted by a halogen atom,
-(C2_Cio)-alkenyl,
-(C2-Cio)-alkynyl,
-(C3-C7)-cycloalkyl-(Ci-C8)-alkyl,
-(C3-C7)-cycloalkyl-(Ci-C8)-alkyenyl,
-(C3-C7)-cycloalkyl-(C2-C8)-alkynyl,
heterocyclyl-(Ci-C8)-alkyl,
heterocyclyl-(Ci-C8)-alkenyl,
heterocyclyl-(C2-C8)-alkynyl,
-R8,
R8-(Ci-C8)-alkyl,
R8-(C2-C8)-alkenyl,
R8-(C2-C8)-alkynyl,
-S-(Ci-Cio)-alkyl,
-S02-(Ci-Cio)-alkyl
-S-R8,
-S02-R8,
-CN -hal,
-0-(Ci-Cio)-alkyl,
-NR6R7, wherein R6 und R7 have the same meaning as defined above, -O-R8,
-OH,
with the exception of -CH(CH3)2 or -C(CH3)=CH2
R8 represents for an aryl group, which might be substituted by 1 to 3 hydroxy groups, halogen, Ci-Cs-alkyl, Ci-Cs-alkoxy, cyano group, a heteroaryl group, whereby the heteroaryl group might comprise 1 to 3 heteroatoms, which optionally are substitut- ed by 1 to 3 alkyl groups, hydroxy, halogen, cyano group or Ci-Cs-alkoxy groups, and salts, solvates or salts of solvates thereof.
According to the present invention, compounds according to formula (I) with the following definitions of the functional groups are preferred:
R1 and R2, independently represent, a hydrogen atom, hydroxy group, a halogen atom, an op- tionally substituted -(Ci-Cio) alkyl, an optionally substituted -(Ci-Cio)-alkoxy, (Ci-
C5) perfluoroalkyl group, a cyano group, or NR6R7, wherein R6 and R7, independently represents hydrogen atom, Ci-Cs-alkyl or -(CO)-(Ci-C5)-alkyl,
R3 represents a hydrogen atom, a hydroxy group, a halogen atom, a cyano group, an optionally substituted -(Ci-Cio)-alkyl group, a -(Ci-Cio)-alkoxy group or a (C1-C5) perfluoroalkyl group,
R4 represents a hydrogen atom, Ci-C3-alkyl, Ci-C3-alkoxy, a hydroxy group or a halogen atom,
R5 represents a group selected from -(Ci-Cio)-alkyl, which are the optionally partially or completely halogenated is, -(C2-Cio)-alkenyl, -(C2-Cio)-alkynyl, -(C3-C7)- cycloalkyl-(Ci-C8)-alkyl, -(C3-C7)-cycloalkyl-(C2-C8)-alkenyl, -S-(Ci-Cio)-alkyl, -
S02-(Ci-Cio) alkyl, -CN, -hal , -O-(Ci-Cio) alkyl, -NR6R7, wherein R6, R7 have the above-mentioned meaning, -OH, with the exception of -CH(CH3)2 or -C(CH3)=CH2, and their salts, solvates or salts of solvates.
According to the present invention, compounds according to Formula (I) with following definitions of the functional groups are further preferred: R^ ^ and R3 independently represent hydrogen, fluorine, chlorine, bromine, a cyano group, a methoxy group, an ethoxy group or a hydroxy group,
R4 represents hydrogen, Ci-C3-alkyl or halogen,
R5 represents a hydroxy group, chlorine, -S-CH3, -C-CH2-CH3, -CH-CH2-CH2-CH3,
-0-CH3 or -0-CH2-CH3, -0-CH2-CH2-CH3, -N-(CH3)2 or -N-(CH2-CH3)2, hydrogen atom, -CH3, or -CH2-CH3.
and salts, solvates or salts of solvates thereof.
According to the present invention, compounds according to formula (I) with the following definitions of the functional groups are further preferred:
R!, R2 and R3 independently represent hydrogen, fluorine, chlorine, bromine, a cyano group, a methoxy group, an ethoxy group or a hydroxy group,
R4 represents hydrogen, Ci-C3 alkyl or halogen,
R5 represents a hydroxy group, chlorine, -S-CH3, -S-CH2-CH3, -S-CH2-CH2-CH3, -O-
CH3 , -0-CH2-CH3, -0-CH2-CH2-CH3, -N(CH3)2, hydrogen atom, -CH3, or -CH2- CH3. and their salts, solvates or salts of solvates.
According to the present invention, compounds according to formula (I) with the following definitions of the functional groups are further preferred:
R!, R2 and R3 independently represent hydrogen, fluorine, chlorine, bromine, a cyano group, a methoxy group, an ethoxy group or a hydroxyl group,
R4 represents hydrogen, Ci-C3-alkyl or halogen,
R5 represents a hydroxy group, chlorine, -S-CH3, -S-CH2-CH3, -S-CH2-CH2-CH3, -
0-CH3, -0-CH2-CH3 or -0-CH2-CH2-CH3, hydrogen atom, -CH3, or -CH2-CH3. and their salts, solvates or salts of solvates.
According to the present invention, compounds according to Formula (I) with the following definitions of the functional groups are further preferred:
R1 and R2 independently represent hydrogen, fluorine, chlorine, a methoxy group or a hydroxy group,
R3 represents hydrogen, fluorine, chlorine, or methoxy group,
R4 represents hydrogen or fluorine, R5 represents a hydroxy group, chlorine, -S-CH3, -S-CH2-CH3, -O-CH3, -O-CH2-
CH3 or N(CH3)2, hydrogen atom, -CH3, or -CH2-CH3.
and their salts, solvates or salts of solvates.
According to the present invention, compounds according to formula (I) with the following defini- tions of the functional groups are further preferred:
R1 and R2 independently represent hydrogen, fluorine, chlorine or a methoxy group,
R3 represents hydrogen, fluorine, chlorine, or methoxy group,
R4 represents hydrogen or fluorine,
R5 represents hydroxy group, chlorine, -S-CH3, -S-CH2-CH3, -0-CH3, -0-CH2-CH3, hydrogen atom, -CH3, or -CH2-CH3.
and their salts, solvates or salts of solvates.
According to the present invention, compounds according to Formula (I) with the following definitions of the functional groups are particularly preferred:
R1 and R2 independently represent fluorine, chlorine or a methoxy group,
R3 represents fluorine, chlorine, or methoxy group,
R4 represents hydrogen or fluorine,
R5 represents hydrogen atom, -CH3, or -CH2-CH3.
and their salts, solvates or salts of solvates.
According to the present invention, compounds according to formula (I) with the following defini- tions of the functional groups are particularly preferred:
R1 represent fluorine or chlorine,
R2 represent fluorine or chlorine,
R3 represents methoxy group,
R4 represents fluorine,
R5 represents hydrogen, chlorine, -S-CH3, -S-CH2-CH3, -0-CH3, -0-CH2-CH3.
and their salts, solvates or salts of solvates.
According to the present invention, compounds according to formula (I) with the following definitions of the functional groups are more preferred:
R1 represent fluorine or chlorine,
R2 represent fluorine or chlorine,
R3 represents methoxy group, R4 represents fluorine,
R5 represents hydrogen atom, -S-CH3, -S-CH2-CH3, -0-CH3, -0-CH2-CH3,
and their salts, solvates or salts of solvates.
According to the present invention, compounds according to Formula (I) with the following defini- tions of the functional groups are much more preferred:
R1 represent ortho-substitution, selected from fluorine or chlorine,
R2 represent meta-substitution, selected from fluorine or chlorine,
R3 represents para-substituted methoxy group,
R4 represents fluorine,
R5 represents hydrogen atom, -S-CH3, -S-CH2-CH3, -0-CH3, -0-CH2-CH3,
and their salts, solvates or salts of solvates.
According to the present invention, compounds according to formula (I) with the following definitions of the functional groups are particularly more preferred:
R1 represent ortho-substituted chlorine,
R2 represent meta-substituted fluorine,
R3 represents para-substituted methoxy group,
R4 represents fluorine,
R5 represents -S-CH2-CH3, or -0-CH3,
and their salts, solvates or salts of solvates.
The specific compounds may be in enantiomerically pure form and their salts, solvates or salts of solvates.
The following compounds are preferred:
5-{[l-(2-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2-
([methylsulfanyl]methyl)propyl]amino}-lH-quinolin-2-one,
5-{[2-([ethylsulfanyl]methyl)-l-(2-fluoro-4-methoxyphenyl)-3,3,3- trifluoro-2-hydroxypropyl] amino } - 1 H-quino lin-2-one,
5-{[l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2-
([methylsulfanyl]methyl)propyl]amino}-7-fluoro-lH-quinolin-2-one,
5-{[l-(2-chloro-3-fluoro-4-methoxyphenyl)-2-([ethylsulfanyl]methyl)- 3 ,3 ,3-trifluoro-2-hydroxypropyl] amino } -7-f uoro- 1 H-quino lin-2-one,
5-{[l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- (methoxymethyl)propyl] amino } -7-fluoro- 1 H-quinolin-2-one,
5-{[l-(2-chloro-3-fluoro-4-methoxyphenyl)-2-(ethoxymethyl)-3,3,3- trifluoro-2-hydroxypropyl] amino} -7-fluoro- lH-quinolin-2-one,
5-{[l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- (hydro xymethyl)propyl] amino } -7-fluoro- 1 H-quinolin-2-one,
5-{[l-(5-chloro-3-fluoro-2-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- (hydroxymethyl)-propyl] amino } -7-fluoro- 1 H-quinolin-2-one,
5-{[l-(5-chloro-3-fluoro-2-methoxyphenyl)-2-(chloromethyl)-3,3,3- trifluoro-2-hydroxypropyl] amino} -7-fluoro- lH-quinolin-2-one,
5-{[3,3,3-trif uoro-2-hydroxy-2-([methoxymethyl)-l-phenylpropyl]amino}- 1 H-quino lin- 1 -one,
5-{[l-(2-chloro-3-fluoro-4-methoxyphenyl)-2-(diaminomethyl)-3,3,3-trifluoro-2- hydroxypropyl] amino} -7-fluoro- 1 H-quino lin-2-one,
5-{[l-(4-chloro-3-fluoro-2-methoxyphenyl)-3,3,3-trifluoro-2- hydro xy-2-(methoxymethyl)propyl] amino } -7-fluoro- 1 H-quino lin-2-one, 5-{[l-(2-chloro-3-fluoro-4-methoxyphenyl)-2-(ethoxymethyl)-3,3,3- trifluoro-2-hydroxypropyl] amino} -7-fluoro- 1 H-quino lin-2-one,
5-{[l-(2-chloro-3-fluoro-4-hydroxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- (hydroxymethyl)propyl] amino } -7-fluoro- 1 H-quino lin-2-one,
and their salts, solvates or salts of solvates.
The following compounds are further preferred:
5-{[l-(2-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- ([methylsulfanyl]methyl)propyl]amino}-lH-quinolin-2-one,
5-{[2-([ethylsulfanyl]methyl)-l-(2-fluoro-4-methoxyphenyl)-3,3,3- trifluoro-2-hydroxypropyl] amino } - 1 H-quino lin-2-one,
5-{[l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- ([methylsulfanyl]methyl)propyl]amino}-7-fluoro-lH-quinolin-2-one,
5-{[l-(2-chloro-3-fluoro-4-methoxyphenyl)-2-([ethylsulfanyl]methyl)- 3 ,3 ,3-trifluoro-2-hydroxypropyl] amino } -7-fluoro- 1 H-quino lin-2-one,
5-{[l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- (methoxymethyl)propyl] amino } -7-fluoro- 1 H-quino lin-2-one, 5-{[l-(2-chloro-3-fluoro-4-methoxyphenyl)-2-(ethoxyrnethyl)-3,3,3- trifluoro-2-hydroxypropyl]amino}-7-fluoro-lH-quinolin-2-one,
5-{[l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifiuoro-2-hydroxy-2- (hydroxymethyl)propyl] amino } -7-fluoro- 1 H-quinolin-2-one,
5-{[l-(5-chloro-3-fluoro-2-methoxyphenyl)-3,3,3-trifiuoro-2-hydroxy-2- (hydroxymethyl)-propyl] amino } -7-fluoro- 1 H-quinolin-2-one,
5-{[l-(5-chloro-3-fluoro-2-methoxyphenyl)-2-(chloromethyl)-3,3,3- trifluoro-2-hydroxypropyl] amino} -7-fluoro- lH-quinolin-2-one,
5-{[3,3,3-trif uoro-2-hydroxy-2-([methoxymethyl)-l-phenylpropyl]amino}- lH-quinolin-l-one,
and their salts, solvates or salts of solvates.
The following compounds are further preferred:
5-{(lS,2R)[l-(2-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- ([methylsulfanyl]methyl)propyl]amino}-lH-quinolin-2-one,
5-{(lS, 2R)[2-([ethylsulfanyl]methyl)-l-(2-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2- hydroxypropyl] amino } - 1 H-quino lin-2-one,
5-{(lS,2R)[l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2- hydroxy-2-([methylsulfanyl]methyl)propyl] amino } -7-fluoro- 1 H-quino lin-2-one, 5-{(lS,2R)[l-(2-chloro-3-fluoro-4-methoxyphenyl)-2-([ethylsulfanyl]methyl)- 3 ,3 ,3-trifluoro-2-hydroxypropyl] amino } -7-fluoro- 1 H-quino lin-2-one,
5-{(lS,2S)[l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2- hydroxy-2-(methoxymethyl)propyl] amino } -7-fluoro- 1 H-quino lin-2-one,
5-{(lS,2S)[l-(2-chloro-3-fluoro-4-methoxyphenyl)-2-(ethoxymethyl)- 3 ,3 ,3-trifluoro-2-hydroxypropyl] amino } -7-fluoro- 1 H-quino lin-2-one,
5-{(lS,2S)[l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2- hydroxy-2-(hydroxymethyl)propyl] amino} -7-fluoro- 1 H-quino lin-2-one,
5-{(lS,2S)[l-(5-chloro-3-fluoro-2-methoxyphenyl)-3,3,3-trifluoro-2- hydroxy-2-(hydroxymethyl)propyl] amino} -7-fluoro- 1 H-quino lin-2-one,
5- {(1 S,2R)[ 1 -(5-chloro-3-fluoro-2-methoxyphenyl)-2-(chloromethyl)- 3 ,3 ,3-trifluoro-2-hydroxypropyl] amino } -7-fluoro- 1 H-quino lin-2-one,
5-{(lS,2S)[3,3,3-trifluoro-2-hydroxy-2-([methoxymethyl)-l- phenylpropyl] amino } - 1 H-quino lin- 1 -one,
5-{[(lS,2R)[l-(2-chloro-3-fluoro-4-methoxyphenyl)-2-(diaminomethyl)-3,3,3-trifluoro-2- hydroxypropyl]amino}-7-fluoro-lH-quinolin-2-one,
5-{(lS,2S)[l-(2-chloro-3-fluoro-4-hydroxyphenyl)-3,3,3-trifiuoro-2-hydroxy-2- (hydro xymethyl)propyl] amino } -7-fluoro- 1 H-quino lin-2-one,
and their salts, solvates or salts of solvates.
The following compounds are further preferred:
5-{(lS,2R)[l-(2-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- ([methylsulfanyl]methyl)propyl]amino}-lH-quinolin-2-one,
5-{(lS, 2R)[2-([ethylsulfanyl]methyl)-l-(2-fiuoro-4-methoxyphenyl)-3,3,3-trifluoro-2- hydroxypropyl] amino } - 1 H-quino lin-2-one,
5-{(lS,2R)[l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2- hydroxy-2-([methylsulfanyl]methyl)propyl] amino } -7-fluoro- 1 H-quino lin-2-one,
5-{(lS,2R)[l-(2-chloro-3-fluoro-4-methoxyphenyl)-2- ([ethylsulfanyljmethyl)- 3 ,3 ,3-trifluoro-2-hydroxypropyl] amino } -7-fluoro- 1 H-quino lin-2-one,
5-{(lS,2S)[l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2- hydroxy-2-(methoxymethyl)propyl] amino } -7-fluoro- 1 H-quino lin-2-one,
5-{(lS,2S)[l-(2-chloro-3-fluoro-4-methoxyphenyl)-2-(ethoxymethyl)- 3 ,3 ,3-trifluoro-2-hydroxypropyl] amino } -7-fluoro- 1 H-quino lin-2-one,
5-{(lS,2S)[l-(2-chloro-3-fiuoro-4-methoxyphenyl)-3,3,3-trifluoro-2- hydroxy-2-(hydroxymethyl)propyl] amino} -7-fluoro- 1 H-quino lin-2-one,
5-{(lS,2S)[l-(5-chloro-3-fluoro-2-methoxyphenyl)-3,3,3-trifluoro-2- hydroxy-2-(hydroxymethyl)propyl] amino} -7-fluoro- 1 H-quino lin-2-one,
5- {(1 S,2R)[ 1 -(5-chloro-3-fluoro-2-methoxyphenyl)-2-(chloromethyl)- 3 ,3 ,3-trifluoro-2-hydroxypropyl] amino } -7-fluoro- 1 H-quino lin-2-one,
5-{(lS,2S)[3,3,3-trifluoro-2-hydroxy-2-([methoxymethyl)-l- phenylpropyl] amino } - 1 H-quino lin- 1 -one,
and their salts, solvates or salts of solvates.
In the present invention, the following compounds according to formula (I) are preferred: 5-{[(lS,2S)-l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- (methoxymethyl)propyl] amino} -7-fluoro- lH-quinolin-2-one (I -A)
Figure imgf000039_0001
5 - {( 1 S ,2S) [ 1 -(2-chloro-3 -fluoro-4-methoxyphenyl)-3 ,3 ,3 -trifluoro-2-hydroxy-2- (hydroxymethyl)propyl]amino}-7-fluoro-lH-quinolin-2-one (I-B)
Figure imgf000039_0002
I-B
5-{(lS,2R)[l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- ({methylsulfanyl}methyl)propyl]amino}-7-fluoro-lH-quinolin-2-one (I-C)
Figure imgf000040_0001
I-C
The compound according to formula (I), 5-{[(lS,2S)-l-(2-chloro-3-fluoro-4-methoxyphenyl)- 3,3,3-trifluoro-2-hydroxy-2-(methoxymethyl)propyl]amino}-7-fluoro-lH-quinolin-2-one (I-A) is
Figure imgf000040_0002
!-A
more preferred.
Definitions of substitutions
Unless otherwise notifed, the term "alkyl" refers to a straight or branched, substituted or unsubsti- tuted chain. For example, the term propyl comprises n-propyl and iso-propyl, the term butyl com- prises n-butyl, iso-butyl and tert-butyl.
The alkyl groups can be straight-chain or branched and stand e.g. for a methyl, ethyl, n-propyl, iso- propyl, n-butyl, iso-butyl, tert-butyl or n-pentyl group, or a 2,2-dimethylpropyl, 2-methylbutyl or 3- methylbutyl group. A methyl or ethyl group is preferred. They can optionally be substituted by 1-3 hydroxy groups, cyano group, halogen, 1-3 Ci-Cs-alkoxy groups, and/or 1-3 -COO(Ci-Cio-alkyl or benzyl) groups. Preferred is hydroxy group. The total number of substituents depends on the number of carbon atoms of the chain. Usually the number of substituents does not exceed the number of carbon atoms except for halogen which leads at a maximum number of substituents to e.g. perfluo- rated alkyl groups.
For a partially or completely fluorinated Ci-C3-alkyl group, the following partially or completely fluorinated groups are considered: fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl, 1 , 1- difluoro ethyl, 1 ,2-difluoroethyl, 1 , 1 , 1-trifluoro ethyl, tetrafluoro ethyl, and pentafluoroethyl. Of the latter, the trifluoromethyl group or the pentafluoroethyl group is preferred.
The Ci-C5-alkoxy groups in R1, R2 or R3 and R3 can be straight-chain or branched and stand for a methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, iso-butoxy, tert-butoxy or n-pentoxy, 2,2- dimethylpropoxy, 2-methylbutoxy or 3-methylbutoxy group. A methoxy or ethoxy group is preferred. They can optionally be substituted by Ci-Cs-alkyl groups, cyano group or halogen.
The Ci-C5-alkylthio groups can be straight-chain or branched and stand for a methylthio, ethylthio, n-propylthio, iso-propylthio, n-butylthio, iso-butylthio, tert-butylthio or n-pentylthio, 2,2- dimethylpropylthio, 2-methylbutylthio or 3-methylbutylthio group. A methylthio or ethylthio group is preferred.
The term halogen atom, hal or halogen means a fluorine, chlorine, bromine or iodine atom. Preferred is a fluorine, chlorine or bromine atom.
The NR6R7 group includes, for example, -NH2, -N(H)CH3, -N(CH3)2, -N(H)(CO)CH3, - N(CH3)(CO)CH3, -N[(CO)CH3]2, -N(H)C02CH3, -N(CH3)C02CH3, or -N(C02CH3)2.
The term C2-C8-alkenyl is a straight or branched, substituted or unsubstituted, chain including isomers having an E- or Z-configurated double bond such as e.g. vinyl, propen-l-yl, propen-2-yl (Al- lyl), but-l-en-l-yl, but-l-en-2-yl, but-2-en-l-yl, but-2-en-2-yl, 2-methyl-prop-2-en-l-yl, 2-methyl- prop-l-en-l-yl, but-l-en-3-yl, but-3-en-l-yl. If the alkenyl residue is placed between two other moieties the term alkenyl means alkenylene such as e.g. vinylene, propen-l-ylene, propen-2-ylene (Allylen), but-l-en-l-ylene, but-l-en-2-ylene, but-2-en-l-ylene, but-2-en-2-ylene, 2-methyl-prop- 2-en-l-ylene, 2-methyl-prop-l-en-l-ylene, but-l-en-3-ylen, but-3-en-l-ylene.
The term C2-C8-alkynyl stands for a straight or branched chain e,g, -C≡CH, -CH2-C≡CH, -C≡C- CH3, -CH(CH3)-C≡CH, -C≡C-CH2(CH3), -C(CH3)2-C≡CH, -C≡C-CH(CH3)2, -CH(CH3)-C≡C- CH3, -CH2-C≡C-CH2(CH3) or, if the alkynyl residue is placed between two other moieties the term alkynyl means alkynylene such as e.g. -C≡C-, -CH2-C≡C-, -C≡C-CH2-, -CH(CH3)-C≡C-, -C≡C- CH(CH3)-, -C(CH3)2-C≡C-, -C≡C-C-(CH3)2-, -CH(CH3)-C≡C-CH2-, -CH2-C≡C-CH(CH3)-.
The term C3-C7-cycloalkyl means a substituted or unsubstituted group selected from cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl. The possible substitutents may be selected from hydroxy, halogen, (Ci-C5)-alkyl, (Ci-Cs)-alkoxy, NR4R5, COO(Ci-C5)-alkyl, CHO, cyano.
The term C3-C7-cycloalkyl-(Ci-Cio)-alkyl- means e.g. -(CH2)-cycloalkyl, -(C2H4)-cycloalkyl, - (C3H6)-cycloalkyl, -(C4H8)-cycloalkyl, -(C6Hio)-cycloalkyl whereby the cycloylkyl stand for e.g. cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl.
The term C3-C7-cycloalkyl-(C2-C8)-alkenyl means e.g. -(CH=CH)-cycloalkyl, -[C(CH3)=CH]- cycloalkyl, -[CH=C(CH3)]-cycloalkyl, -(CH=CH-CH2)-cycloalkyl, -(CH2-CH=CH)-cycloalkyl, - (CH=CH-CH2-CH2)-cycloalkyl, -(CH2-CH=CH-CH2)-cycloalkyl, -(CH2-CH2-CH=CH)-cycloalkyl, -(C(CH3)=CH-CH2)-cycloalkyl, -(CH=C(CH3)-CH2)-cycloalkyl whereby the term cycloalkyl is defined above.
The term heterocyclyl means e.g. piperidinyl-, morpholinyl-, thiomorpholinyl-, piperazinyl-, tetra- hydro fur any 1-, tetrahydrothienyl-, imidazolidinyl- or pyrrolidinyl- whereby the heterocyclyl group may be bound via any possible ring atom.
The heterocyclyl group may be substituted by Ci-Cs-alkyl (optionally substituted), hydroxy-, Ci- C5-alkoxy-, NR4R5-, halogen, cyano-, COOR8-, CHO-. If possible these substitutens may also be bound to one of the free nitrogen atoms if any. N-oxides are also included in the definition.
The term heterocyclyl-(Ci-Cio)-alkenyl- means an alkylene group as defined above which is connected to the heterocyclyl group which also is already defined above.
The term heterocyclyl-(C2-C8)-alkenyl- means an alkylenylene group as defined above which is connected to the heterocyclyl group which also is already defined above.
The term aryl in the sense of the invention means aromatic or partially aromatic carbocyclic rings having 6 to 14 carbon atoms, e.g. phenyl and which may also may have a condensed a second or third ring such as e.g. napthyl or anthranyl. Further examples are phenyl, naphthyl, tetralinyl, an- thranyl, benzoxazinone, dihydroindolone, indanyl, and indenyl.
The aryl groups may be substituted at any position leading to a stable molecule by one or several substitutents, e.g. 1-3 substitutents, such as e.g. hydroxy, halogen, Ci-Cs-alkyl, Ci-Cs-alkoxy, cy- ano, -CF3, nitro, -COO(Ci-C5-alkyl or benzyl) or a heteroaryl group, preferably by 1-3 Ci-Cs-alkyl groups, hydroxyl, halogen, cyano or Ci-Cs-alkoxy. The optionally substituted phenyl group is one aspect of the invention. Yet another aspect are the compounds of formula 1 whereby R8 is not phenyl.
The term heteroaryl means an aromatic ring system having 1-3 heteroatoms selected from nitrogen, oxygen or sulfur, for five membered rings the maximum number of heteroatoms is three whereby only two oxygen or sulfur atoms are allowed provided that these two are not directly bound to each other. Possible heteroaryl rings are e.g. thienyl, furanyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, benzofuranyl, benzothienyl, benzothiazol, benzoxazolyl, benzimidazolyl, indazolyl, indolyl, isoindolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, azaindolizinyl-, benzopyridyl, benzopyridazinyl, benzopyrimidi- nyl, benzopyrazinyl, benzotriazinyl, quinolyl, isoquinolyl, phthalidyl-, thiophthalidyl, indolonyl-, dihydroindolonyl-, iso indolonyl-, dihydroiso indolonyl-, benzofuranyl- or benzimidazolyl.
The compounds according to formula (I) can exist in stereo isomeric forms such as enantiomers of diastereo isomers depending on their structure and residues as defined in Formula (I). In one aspect of the invention therefore all these enantiomers, diastereoisomers or mixtures thereof are encom- passed. The isolation of enantiomerically or diastereomerically pure isomers can be done by methods of the state of the art, e.g. using column chromatography with a chiral solid phase.
Should it be possible that the compounds according to formula (I) also exist in tautomeric forms these are also an aspect of the present invention.
In one aspect of the invention all compounds defined in formula (I) as well as their salts, solvates and solvates of salts are encompassed, especially the salts, solvates and salts of solvates of the compounds disclosed in the examples are one aspect of the invention as long as the disclosed compounds themselves are not already salts, solvates or solvates of the salts.
Salts in the sense of the present invention are not only physiologically unobjectable salts but also salts which might be objectable for pharmaceutical use but which are useful e.g. during the process of isolation or purification.
The term physiologically unobjectable salts includes addition salts of mineral acids, carbonic acids, sulfonic acids, e.g. salts of hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, toluolsulfonic acid, benzenesulfonic acid, naphthalinesulfonic acid, acetic acid, trifluoro acetic acid, propionic acid, lactic acid, tartaric acid, malic acid, citric acid, fumaric acid, pivalic acid, maleic acid, succinic acid and benzoic acid. In addition the term physiologically unobjectable salts includes salts of commonly suitable bases, e.g. salts of alkalimetall (e.g. sodium- and potassium salts), alkaline earth salts (e.g. calcium- and magnesium salts) and ammonium salts, derivatized from NH3 or organic amines with 1 to 16 carbon atoms, e.g. ethylamine, diethylamine, triethylamine, ethyldiisopropylamine, monoethanola- mine, diethanolamine, triethanolamine, dicyclohexylamine, dimethylaminoethanol, prokaine, dibenzylamine, N-methylmorpholin, arginin, lysin, ethylendiamine and N-methylpiperidin.
Solvates in the sense of the invention are such forms of the compounds of the present combinations which build complexes by coordination of solvent molecules in a liquid or a solid phase. Hydrates are special forms of a solvate wherein water molecules are coordinated.
Salts in the sense of the present invention are not only physiologically unobjectable salts but also salts which might be objectable for pharmaceutical use but which are useful e.g. during the process of isolation or purification.
The compounds according to formula (I) can be synthesized by means of the following synthetic methods, procedure A and procedure B:
Procedure A:
step a)
Figure imgf000044_0001
(II) (IV)
Benzaldehydes of type (II) can be condensed with substituted aminoquinolones of type (III) to imines of type (IV) using Lewis acids, preferably titanium alcoholates Ti(OR)4 wherein R is Ci-C4- alkyl, such as e.g. tetraethyl orthotitanate or tetra tert. butyl orthotitanate and/or acidic conditions, e.g. organic acids such as acetic acid as reagents. Suitable solvents are e.g. toluene, 1 ,4-dioxane or mixtures thereof. step b)
Figure imgf000045_0001
(VI)
Imines of type (IV) are treated at low temperatures of -80° to -100° C. with the lithiated epoxide (V) to yield compounds of type (VI). Suitable solvents are e.g. tetrahydrofurane, hexane, diethy- lether or mixtures thereof. The epoxides (VI) can be opened by nucleophiles of type R5-Met to deliver compound (I). Met means metal and includes alkalimetals e.g. sodium or lithium, alkaline earth metals such as e.g. magnesium, caesium; aluminium, copper, silicon or tin (Sn) which bind the nucleophilic residue R5 of R5-Met depending on their valence and according to the knowledge of a person with ordinary skill. The resulting possible nucleophilic reagents R5-Met are e.g. alkyl- cuprates, vinylcuprates, thioles, allylsilanes, vinylsilanes, vinylstannanes, grignard compounds whereby R5 is defined as in claim 1, which react in the presence of Lewis acids like e.g. BF3 or AlMe3, AICI3. Suitable solvents are e.g. diethylether, dimethylformamide, tetrahydroiurane. The epoxides (VI) can also be opened directly by cyanides, amines, alcoholates, thioalcoholates, halogenides and even water or CS2CO3/H2O in the presence of bases or strong protic acids.
Suitable bases in the sense of the invention are e.g. CS2CO3, K2CO3 or NaOH.
Suitable strong protic acids are e.g. HCIO4, HC1 or HBr.
Procedure B:
step a)
Figure imgf000046_0001
(VII) (VIII)
Methoxymethylamides of type (VII) are treated at low temperatures of -80° to -100° C. with the lithiated epoxide (V) to yield compounds of type (VIII). Compounds of formula (VII) are commercially available or can be synthesized according to Branca et al, Chimia 49, 10; 1995, 381-385. step b)
Figure imgf000046_0002
(VII I) (IX) The epoxides (VIII) can be opened by nucleophils of type R5-Met to deliver compound (IX). Possible nucleophiles are alkylcuprates, vinylcuprates, thioles, allylsilanes, vinylsilanes, vinylstan- nanes, grignard compounds, in the presence of Lewis acids like BF3 or AUVfe, AICI3, or directly by cyanides, amines, alcohols, thioalcohols, halogenides and water in the presence of bases or strong protic acids. step c)
Figure imgf000047_0001
(IX) (I)
Ketones of type (IX) can be condensed with substituted aminoquinolones of type (III) to imines and subsequently or simultaneously reduced to the aminoalcohol I by a reductive amination using complex hydrides like e.g. NaBH4 or LiAlH4 (Katritzky et al. J. Org. Chem. 1995, 60, 7631-7640) or hydrogen in the presence of catalytic amounts of palladium or platinum or by application of an asymmetric organo cat lytic transfer hydrogenation (List et al. Angew. Chem. 2005, 117, 7590- 7593).
These procedures described above can be perfomed enantioselectively by use of enantiopure epoxide of formula (V) to yield enantiopure compounds of formula (VI), (VIII), (IX) and (I). The last reductive step of b) can be performed in a diastereoselective manner to yield enantiopure compound I when enantiopure compound IX is used as starting material.
Alternatively during the process of the production of the compounds of formula (I) at different stages purification for obtaining enantiomerically or diastereomerically pure intermediates may be performed e.g. intermediates of formula VI, VIII, IX can be purified at the step when they are obtained or compounds of formula I can be purified to obtain enantiomerically or diastereomerically pure end products after the complete reaction cascade. Examples for methods for obtaining enantiopure (enantiomerically pure) compounds are described below. The separation of optical isomers can be performed by separation of one or more of the intermediates and/or separation of the end products. Usually separation of intermediates and separation of end products are alternatives as long as no racemisation had taken place during the production process.
If the compounds according to the invention are present as racemic mixtures, they can be separated into pure, optically active forms according to the methods of racemate separation that are familiar to one skilled in the art. For example, the racemic mixtures can be separated by chromatography on an even optically active carrier material (CHIRALPAK AD®) into the pure isomers. It is also possible to use chiral auxiliary agents as optically pure acids. For that purpose the free hydroxy group is esterified to yield a racemic compound of general formula (I) with an optically active acid and to separate the diastereoisomeric esters that are obtained by fractionated crystallization or by chroma- tography, and to saponify the separated esters in each case to the optically pure isomers. As an optically active acid, for example, mandelic acid, camphorsulfonic acid or tartaric acid can be used. Thus one aspect of the invention is the process of obtaining compounds of formula (I) in diastere- omerically pure form, optionally using chromatography with columns containing chiral material or using chiral auxiliary agents.
Each of the intermediates of the synthesis of the compounds of formula (I) are one aspect of the present invention as well as especially their use for the synthesis of the compounds of formula (I). A specific aspect of the invention are the concrete intermediates as used for the synthesis of the compounds of the examples, either as racemate or in their enantiomerically (having one chiral center) or diastereomerically (having two chiral centers) pure form. Compound of formula I-A is known from WO 2009/065503 A (ex. 5 as racemate) and can be isolated from racemic form by chiral HPLC, Chiralpak IC 5 μιη with eluent hexane/ethanol (4 : 1), it might also be prepared by enantiomeric selective synthesis.
Compound of formula I-B is known from WO 2009/065503 A (ex. 7 as racemate) and can be isolated from racemic form by chiral HPLC, Chiralpak IC 5 μιη with eluent hexane/ethanol (4 : 1). Compound of formula I-C is known from WO 2009/065503 A (ex. 3 as racemate) and can be isolated from racemic form by chiral HPLC, Chiralpak IC 5 μιη with eluent hexane/ethanol (4 : 1). Examples and experimental data
Biological tests
Impact on T-cell mediated activities
The compounds of general formula (I) for use in the topical treatment of T cell mediated inflamma- tory skin diseases according to the invention exert potent anti-inflammatory and immune- modulatory effects in a broad scope of in vitro activities which are related to T-cell mediated skin diseases such as psoriasis, atopic dermatitis or allergic contact dermatitis.
Inhibition of the mixed leukocyte reaction
Compounds of the general formula (I) are able to inhibit the proliferation of stimulated human lymphocytes in a potent, dose-dependent manner.
The mixed leukocyte reaction (MLR) is an ex vivo cellular immune assay examining lymphocyte proliferation in response to allogenic leukocytes (i.e. leukocytes sufficiently dissimilar in genotype to interact antigenically). It represents a standard in vitro assay for T cell immunity (McDevitt 2000), which is also well suited to determine immunomodulatory effects of added compounds. Peripheral blood mononuclear cells (PBMCs) from two healthy donors were separated from citrate- anticoagulated whole blood. Untreated PBMCs from one donor were mixed with mitomycin C- treated allogeneic PBMC (stimulator cells) from the other donor at a ratio 1 :2 and cultured at 37°C with 5 % C02. To study the immunomodulatory activity of the test compounds, the mixture of untreated responder and mitomycin-treated allogeneic stimulator PBMC (ratio 1 :2) were seeded onto 96-well plates (1.5 x 105 cells/well) and were incubated without test compounds (positive control showing the maximum achievable lymphocyte proliferation after stimulation by allogenic leucocytes) and with increasing concentrations of the compounds of the general formula (I) (concentration range: l .OE-12 to l .OE-6 mo 1/1). [3H] -thymidine was added for the last 6 hours of the 5 -day culture. Scintillation β-counting was used for determination of [3H] -thymidine incorporation into the cell nucleus of the lymphocytes as a measure for alloreactive lymphocyte proliferation of responder cells.
Mixed leukocyte culture of allogeneic responder and stimulator cells led to a significant increase of responder lymphocyte proliferation which was inhibited in a dose dependent manner by the compounds of the general formula (I). The dose response usually forms a sigmoidal curve if the re- sponse (y-axis; here the magnitude of the lymphocyte proliferation) is depicted as a function of the increasing inhibitor concentration (x-axis; here: the increasing concentrations of the compounds of the general formula (I)), which serves to determine potency and efficacy of the individual compounds. Efficacy is defined in this experimental setting as the measure of the maximum inhibition of lymphocyte proliferation which a compound can achieve). The potency of a compound refers to the amount (concentration) of compound required to achieve a defined biological effect (in this case a defined inhibition of the lymphocyte proliferation). Considering the usually sigmoidal shape of a dose-response curve, persons skilled in the art frequently use for the comparison of potencies of different compounds the IC50 value, which is the concentration of an inhibitor where the response is reduced by half (in this case: 50% inhibition of the lymphocyte proliferation). The smaller the compound concentration required for 50% inhibition, the more potent is the compound. All tested compounds of the general formula (I) are able to exert a dose-dependent inhibition of the proliferation of stimulated lymphocytes. All tested compounds of the general Formula (I) are also able to completely inhibit the responder lymphocyte proliferation at high concentrations which means that they are able to achieve -100% efficacy (full inhibition of lymphocyte proliferation). However, the compounds differ in their potencies. Compounds (I-C) and (I-A) exert with IC50 values in the low nanomolar range (4.26E-10 and 1.91E-9 mol/1) significantly more potent inhibitory effects than the compound (I-D) (IC50: 8.34E-8 mol/1). The compound (I-A) has been tested in parallel with two reference compounds, the very potent topical glucocorticoid, Clobetasol and the weaker topical glucocorticoid, Dexamethasone, which are examined for their inhibitory effects in the same concentration range as the compounds of the general formula (I). The compound (I-A) (IC50: 1.91E-9 mol/1) is slightly less potent than Clobetasol (IC50: 8.0E-10 mol/1), but clearly more potent than Dexamethasone (IC50: 3.2E-8 mol/1) with similar efficacy for all three compounds (see also Table 4).
Table 4:
Figure imgf000051_0001
These results indicate that the compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases according to the present invention are able to inhibit the antigen-induced proliferation human lymphocytes in a mixed leukocyte reaction. In other words, the compounds show significant immunomodulatory activity.
Inhibition of psoriasis-inducing and -maintaining cytokines in isolated human PBMCs
Compounds of the general formula (I) are able to inhibit the release of cytokines which are related to the proliferation of a T-cell subset, the T-helper (Th) cells, related to the induction and perpetua- tion of psoriasis.
The manifestation of psoriasis is orchestrated by two subsets of pro -inflammatory CD4-positive T helper lymphocytes, the T helper (Th)17 and the Thl cells (Ghoreschi K et al, 2007). The Thl7 cells, a novel T-cell subset, have been implicated in the pathogenesis of psoriasis, while the Thl cells mainly contribute to the perpetuation of the inflammatory reaction in psoriasis. The cytokine IL-23 stimulates the survival and proliferation of Thl 7 cells, and thus serves as a key cytokine regulator for this disease (Fitch E et al, 2007). The cytokine IL-12, produced by activated monocytes/macrophages and dendritic cells, is the dominant factor promoting Thl cell polarization. Although TNF-a, produced by multiple cells, including activated Thl cells, does not induce proliferation of Thl or Thl7 cells, it is nevertheless a very prominent pro -inflammatory cytokine which can also be correlated to the pathophysiology of psoriasis (Victor FC et al, 2003). Accordingly, the attenuation of IL-23, IL-12 and TNF-a, should exert beneficial therapeutic effects in psoriasis (Numerof RP & Asadullah K, 2006).
The incubation of isolated human peripheral blood mononuclear cell (PBMCs; a mixture of lymphocytes and monocytes) with the endotoxin LPS (bacteria-derived lipopolysaccharides, which induce a strong immune response in animal and human cells) induces the release of TNF-a and IL- 12p40, the common sub-chain of IL-12 and IL-23 heterodimers which can be inhibited by compounds of the general Formula (I).
PBMC were seeded onto 96-well plates (2.5 x 105 cells/well) and were incubated for 24h with 10 ng/ ml LPS without (positive control showing the maximum achievable cytokine release) or with increasing concentrations of compounds of the general Formula (I) (concentration range: 10"12 to 10"6 mo 1/1). After 24 hours cytokine concentrations in supernatants of untreated cells and cells treated with increasing concentrations of test compounds were determined using a specific enzyme- linked immunosorbent assay (ELISA; an antibody-based detection system for the cytokines; TNFa- Biosource, IL-12p40 - R&D Systems) and a chemiluminescence assay (I GEN; suited for the sensi- tive detection of different ELISA reaction products).
The LPS-stimulation of PBMC led to the secretion of TNF-a, as well as IL-12p40, which could be inhibited in a potent and dose dependent manner by the compounds of the general Formula (I). In this experimental setting the dose response relation also forms a sigmoidal curve if the response (y- axis; here the magnitude of the TNF-a, as well as the IL-12p40 release) is depicted as a function of the increasing inhibitor concentration (x-axis; here: the increasing concentrations of the compounds of the general formula (I)), which serves to determine potency and efficacy of the individual compounds. Efficacy is defined in this experimental setting as the measure of the maximum inhibition of TNF-a or IL-12p40 release which a compound can achieve). The potency of a compound refers to the amount (concentration) of compound required to achieve a defined biological effect (in this case a defined inhibition of the TNF-a or IL-12p40 release). The IC50 value, which is the concentration of an inhibitor where the response is reduced by half (in this case: 50% inhibition of the TNF-a or IL-12p40 release), is used for the comparison of the potencies of different compounds. The smaller the compound concentration required for 50% inhibition, the more potent is the compound.
With the exception of (I-D), which was only able to attenuate the TNF-a secretion weakly even at high concentrations, all test compounds exerted almost complete inhibition of the cytokine release. The compounds (I-C), (I-A) and (I-B) inhibited the secretion of IL-12p40 (IC50 values: 3.94E-10 - 7.21E-10 mol/1) as well as the TNF-a release (IC50 values: 6.21E-10 - 1.01E-9 mol/1) very potently with IC50 values the sub-nanomolar range (see table 2), while the compound (I-D) and (I-E) were less potent by inhibiting the IL12p40 secretion at significantly higher concentrations (IC50 values: 3.41E-08 mol/1 and 1.42E-8 mol/1, respectively). The compound (I-D) exerted also a less potent inhibition of the TNF-a secretion (IC50: 6.84E-8 mol/1) (see also Table 5).
In a separate experiment the compound (I-A) has been tested in parallel with two reference compounds, the very potent topical glucocorticoid, Clobetasol and the weaker topical glucocorticoid, Dexamethasone, which are examined for their inhibitory effects in the same concentration range as the compounds of the general Formula (I). The compound (I-A) (IL12p40 IC50: 7.5E-10 mol/1; TNF-a IC50: 6.5E-10 mol/1) is less potent than Clobetasol (IL12p40 IC50: 1.4E-10 mol/1; TNF-a IC50: 1.2E-10 mol/1), but more potent than Dexamethasone (IL12p40 IC50: 4.7E-9 mol/1; TNF-a IC50: 6.1E-9 mol/1) with similar efficacy for all three compounds. Table 5:
Inhibif ion of LPS-ii lduced Inhibit tion of LPS-ii lduced
IL- 12p40 Secret ion T NF-aSecretic in
in human PBM [C in human PBM [C
Formula
IC50: IC50 IC50:
IC50
Comparison Efficacy Comparison Efficacy
[mol/1]
of Potency [mol/1] of Potency
I-C 3,94E-10 1,0 98% 6.21E-10 1,0 105%
I-A 7.17E-10 1,8 97% 7,90E-10 1,3 98%
I-B 7.21E-10 1,8 98% 1.01E-09 1,6 99%
I-D 3,41E-08 87 12% 6,84E-08 110 113%
I-E 1.42E-08 36 92% These experimental data clearly show that the compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases according to the present invention are able to inhibit potently and effectively the secretion of IL-12p40 in human PBMCs, which is the common sub-chain of IL- 23 and IL-12, the essential growth factors for Thl7 and Thl cells. Based on this, it can be derived that the compounds are able to inhibit the proliferation of these T-cells which are related to the induction and perpetuation of psoriasis. Moreover, the experimental data also show that said compounds are also able to inhibit potently and effectively the secretion of TNF-a, which is a potent pro-inflammatory cytokine related to the pathophysiology of psoriasis. Accordingly, it seems reasonable to suppose that at least the compounds (I-C), (I-A) and (I-B) are able to exert beneficial therapeutic effects in psoriasis.
Inhibition of major atopic dermatitis- and allergic contact dermatitis-inducing and maintaining Cytokines
Compounds of the general formula (I) are able to inhibit the release of cytokines which are related to the proliferation of T-helper (Th) cells related to the induction and perpetuation of atopic derma- titis and allergic contact dermatitis.
The manifestation of atopic dermatitis is mainly caused by the cytokine IL-4 which initiates the proliferation of the Th2 subsets of pro-inflammatory CD4-positive T helper (Th) lymphocytes. These cells play a key role in the early phase of atopic diseases (Biedermann T et al, 2004). In chronic atopic dermatitis skin lesions significantly lower levels of IL-4 can be measured compared to acute lesions. IL-12 produced by infiltrating pro -inflammatory dendritic epidermal cells, macrophages and eosinophils, triggers a partial switch to a Thl -type cytokine milieu associated mainly with increased IFN-γ expression (Leung DYM, 2000).
Although IL-4 seems also to contribute to the inflammatory reaction in allergic contact dermatitis, IL-2, the growth factor of Thl cytokines, and IFN-γ play a more prominent role in the initiation and perpetuation of allergic contact dermatitis (Kimber I, 2002).
The plant lectin, phytohemagglutinine (PHA) acts as a T-cell mitogen, which does not only induce the proliferation of these cells but also induces the secretion of IL-2, IL-4 and IFN-γ if it is incubated with human PBMCs. This assay was used to determine the inhibitory activity of compounds of the general Formula (I) on the IL-2, 11-4 and IFN-γ release. PBMC were seeded onto 96-well plates (2.5 x 105 cells/well) and incubated for 24 h with 10 μ /ηιΕ PHA without (positive control showing the maximum achievable cytokine release) or with increasing concentrations of compounds of the general Formula (I) (concentration range: 10"12 to 10"6 mo 1/1). After 24 hours cytokine concentrations in supernatants of untreated cells and cells 5 treated with increasing concentrations of test compounds were determined using a specific enzyme- linked immunosorbent assay kits from Biosource (ELISA; an antibody-based detection system for the cytokines) and chemiluminescence for the sensitive detection of different ELISA reaction products.
The PHA-stimulation of PBMC led to the secretion of IL-2, IL-4 and IFN-γ which could be inhibit) ited in a potent and dose dependent manner by the compounds of the general Formula (I). In this experimental setting the dose response relation also forms a sigmoidal curve if the response (y- axis; here the magnitude of the IL-2, IL-4 or IFN-γ release) is depicted as a function of the increasing inhibitor concentration (x-axis; here: the increasing concentrations of the compounds of the general formula (I)), which serves to determine potency and efficacy of the individual compounds I 15 the same way as it was done for the TNF-a or IL-12p40 release.
Compounds of formula (I-C), (I-A) and (I-B) were able to inhibit the PHA-stimulated cytokine secretion very potently with IC50 values in the subnano molar concentration range (IL4 IC50 range: l . lOE-10 mol/1 - 6.14E-10 mol/1; IL-2 IC50 range: 2.95E-10 mol/1 - 1.22E-9 mol/1; IFN-γ IC50 range: 4.81E-10 mol/1 - 6.48E-10 mol/1; Table 6 for more details). In contrast, the compound of 20 example 8 is significantly less potent with an IL4 IC50 of 3.29E-8 mol/1, an IL-2 IC50 of 7.04E-08 mol/1 and an IFN-γ IC50 of 3.31E-8. Compounds (I-C), (I-A), and (I-B) were also very effective by inducing complete inhibition of the release of all cytokines at higher concentrations, while the compound (I-D) is significantly less effective as it did not even reach complete inhibition even at the highest tested concentration of 10"6 mol/1.
25 In a separate experiment the compound (I-A) has been tested in parallel with two reference compounds, the very potent topical glucocorticoid, Clobetasol and the weaker topical glucocorticoid, Dexamethasone, which are examined for their inhibitory effects in the same concentration range as the compounds of the general Formula (I). The compound (I-A) (IL-4 IC50: 4.0E-10 mol/1; IL-2 IC50: 8.8E-10 mol/1, IFN-γ IC50: 7.0E-10 mol/1) is less potent than Clobetasol (IL-4 IC50: 7.0E-11 30 mol/1; IL-2 IC50: 1.4E-10 mol/1, IFN-γ IC50: 1.2E-10 mol/1), but more potent than Dexamethasone (IL-4 IC50: 2.3E-9 mo 1/1; IL-2 IC50: 4.6E-9 mo 1/1, IFN-γ IC50: 3.8E-9 mo 1/1) with similar efficacy for all three compounds.
Table 6:
Figure imgf000056_0001
The in vitro experiments in isolated human PBMCs clearly show that the compounds (I-C), (I-A) and (I-B) are able to inhibit in a potent and effective manner the release of the cytokines IL-2, IL-4 and IFN-γ, which are involved in the proliferation of Thl and Th2 lymphocytes and in the perpetuation of the inflammatory reaction in T-cell mediated skin diseases such as psoriasis, atopic dermatitis and allergic contact dermatitis.
Since the proliferation of TH2 cells related to the induction and perpetuation of atopic dermatitis and the proliferation of THl cells to allergic contact dermatitis, it is suggested that the compounds (I-C), (I-A) and (I-B) could exert beneficial therapeutic effects in atopic dermatitis and allergic contact dermatitis.
Surprisingly compounds like e.g. (I-D), which despite of potent binding to the Glucocorticoid re- ceptor excerted extremely weak inhibition of T-cell mediated activities. This is a clear indicator that compounds like e.g. (I-D) are not able to induce any anti-inflammatory or immunomodulatory activities in T-cell mediated skin diseases.
Impact on T-helper cell-dominated inflammatory skin reactions in rodent models
The compound (I-A) is also able to inhibit T-cell mediated inflammatory skin reactions in mouse and rat models in a potent and effective manner.
It is well established that in rodents different forms of chemical allergens provoke qualitatively distinct immune responses. Thus, the topical exposure of mice or rats to a contact allergen such as dinitrochlorobenzene (DNCB) or dinitrofluorobenzene (DNFB) induces about 24 hours after challenge a Thl dominated response (Hayashi M et al, 2001). In DNFB-sensitized rodents, the chal- lenge of the skin with DNFB leads to the recruitment of DNFB-specific T-cells to the skin which produce Thl cytokines, including IFN-γ, IL-12 and IL-2 while the levels of Th2 cytokines are comparatively low (Takeshita K et al, 2004).
NMRI outbred mice or Wistar rats were sensitized on the shaved right flank with 0.5 % DNFB at day 0 and day 1. On day 5, the animals were challenged by topical application of 0.3 % DNFB onto the dorsal side of both ears. Different dosages of the compound (I-A) (0.0001%, 0.001%, 0.01% and 0.1% w/vol) or vehicle were topically co-applied with the allergen challenge. As parameters for the antigen-induced, T-cell mediated skin inflammations, two parameters were used: edema formation, measured as increase in ear weight of ear punch biopsies of the challenged animals compared to the ear weight of ear punch biopsies of non-challenged animals, and the extent of pro- inflammatory granulocyte infiltration into the ear skin, measured by an increase of the granulocyte enzyme peroxidase in ear homogenates compared to the enzyme content in non-challenged ears. To determine the peroxidase content, aqueous supernatants of homogenized ear punch biopsies were incubated for 30 min with a peroxidase sensitive chromogen, Tetramethylbenzidine (TMB) dihy- drochloride. The extent of the peroxidase concentration in each sample was photometrically deter- mined. Changes in the optical density were monitored at 450 nm at 25 °C against the mixture of all solutions without the added sample homogenate. Absolute extinction numbers were used to express peroxidase activity. Both parameters were determined in animals sacrificed 24h after challenge. The extent of the inflammatory reaction was characterized by the compound dose, which exerts almost complete, 80% (ED80) inhibition of the inflammatory skin reaction.
The compound (I-A) was able to inhibit the DNFB-induced allergic dermatitis reaction in a potent, dose dependent manner. In in vivo experiments the dose response usually also forms a sigmoidal curve if the response (y-axis; here the magnitude of either edema formation or granulocyte infiltration measured via peroxidase activity) is depicted as a function of the increasing inhibitor dosages (x-axis; here: the increasing dosages [%] of the compound (I-A)), which serves to determine potency and efficacy of the given compounds. The potency of a compound refers to the amount (dosage) of compound required to achieve a defined biological effect (in this case a defined inhibition of edema formation of granulocyte infiltration).
To determine the inhibitory effect of anti-inflammatory compounds the difference between the respective mean value of the positive controls and the mean value of the vehicle controls was set to 100% and the percentile change by the test substance was estimated:
mean value treated group - mean value positive group
% change = x 100
mean value positive group - mean value control group
Persons skilled in the art frequently use for the comparison of potencies of different compounds in in vivo experiments the dosage which achieves almost complete inhibition of the symptoms the
ED80 (ED80 = the Effective Dose which achieves 80% inhibition of the symptoms). The lower the compound dosage required for 80%> inhibition, the more potent is the compound.
Formula used for calculation of ED80:
Cone. 1 + (Cone. 2 -Conc. l) * ((80%) - inhibition 1)
ED80 =
(inhibition 2 -inhibition 1 parameters:
Lower data point adjacent to 80% inhibition = Cone. 1, inhibition 1 (%)
Upper data point adjacent to 80% inhibition = Cone. 2, inhibition 2 (%)
Edema formation in mice was almost completely inhibited by the compound (I-A) at the very low dosage of 0.017%. The compound (I-A) inhibits the DNFB-induced edema formation in mice by a factor of 6 more potent compared to that in rats (Rat Edema Formation - ED80: 0.097%). The almost complete inhibition of the granulocyte infiltration in the ear skin of mice did only differ from that of rats by a factor of ~2 (Granulocyte Infiltration ED80: Mouse: 0.100% vs. Rat: 0.190%) (for all ED80 values see also table 7).
Table 7: Compound of Formula I-A: Inhibition of Th 1 -dominated Allergic Contact
Dermatitis
ED80: ED80:
80% Inhibition 80% Inhibition
Contact Allergen Species
of Edema of Granulocyte
Formation Infiltration
Dinitrofluorobenzene
Mouse 0.17% 0.10%
(DNFB)
Dinitrofluorobenzene
Rat 0.10% 0.19%
(DNFB)
ED = Effective Dosage
The compound (I-A) has been tested in parallel with a reference compound, the very potent topical glucocorticoid Clobetasol, which was examined for its inhibitory effects in the same concentration range as the compound (I-A). In rats the compound (I-A) did not differ in its inhibition of the DNFB-induced edema formation from that of Clobetasol (Rat Edema Formation - Compound (I- A): ED80: 0.097% vs. Clobetasol: ED80: 0.085%), while Clobetasol inhibited the granulocyte infiltration in rats slightly more potent by a factor of ~3 (Rat Granulocyte Infiltration - Compound (I-A): ED80: 0.190% vs. Clobetasol ED80: 0.073%). Clobetasol inhibited the DNFB-induced edema formation in mice with a factor of ~2 slightly less potent than the compound (I-A) (Mouse Edema Formation - Compound (I-A): ED80: 0.017% vs. Clobetasol: ED80: 0.040%), while both compounds attenuated the granulocyte infiltration in mice at a higher, but similar dosage (Mouse Granulocyte Infiltration - Compound (I-A): ED80: 0.100% vs. Clobetasol ED80: 0.107%) (for all ED80 values see also table 8).
Table 8:
Figure imgf000060_0001
ED = Effective Dosage
In summary, both, the compound (I-A), as well as Clobetasol were able to almost completely inhibit the symptoms of the Thl cell mediated, DNFB-induced allergic contact dermatitis in rats and mice at dosages lower than 0.1%. The compound (I-A) does not differ in this animal model in its potency and efficacy from Clobetasol.
The topical exposure of rodent skin to chemical respiratory allergens such trimellitic anhydride (TMA) or tolylene 2,4-diisocyanate (TDI) induces about 24 hours after challenge a Th2 dominated response with draining lymph node cells producing high levels of Th2 cytokines, such as IL-4 or IL-5, but only comparatively low levels of IFN-γ (Kimber et al, 2002).
NMRI outbred mice or Wistar rats were sensitized by 5% TMA (mice: day 0; rats: day 0 and 4) or 1% TDI (rats: day 0) onto the shaved right flank. The allergic contact dermatitis reaction was induced on day 5 by challenging the animals with a single application of 10% TMA or 1.5% TDI onto the dorsal side of both ears. Five different dosages of the compound (I-A) and vehicle were topically co-applied with the allergen challenge in all three experiments (Dosages of the compound (I-A) - Mouse TMA and Rat TDI: 0.00001%, 0.0001%, 0.001%, and 0.01%; Rat TMA: 0.0001%, 0.001%, 0.01%, and 0.1%). After 24 h, animals were sacrificed to determine ear weight and perox- idase activity from ear homogenates as parameters for edema and granulocyte infiltration. The experimental procedure, the determination of the peroxidase activity in ear punch biopsies, the calculation of the percentile change by the test substance as well as the assessment of the effective dosages was carried out in the same way as described for the DNFB-induced allergic contact dermati- tis.
The compound (I-A) was able to inhibit the TMA-induced allergic dermatitis reaction in mice and rats and the TDI-induced reaction in rats in a very potent, dose dependent manner. Edema formation as well as granulocyte infiltration in the ear skin of both species was almost completely inhibited (ED80 = 80% inhibition) at very low topical dosages ranging between 0.00015%) and 0.00860%. Complete inhibition (ED 100) of edema formation and granulocyte infiltration were achieved in rats already at the low dosage of 0.01% for both, the TMA-, as well as the TDI-induced reaction. The edema formation of the TMA-induced reactions in mice is reached at an even lower dosage (Mouse Edema Formation - ED100: 0.001%), while the TMA-induced granulocyte infiltration was not yet reached at the highest tested dosage of 0.01% (for all ED80 / ED 100 values see also table 9).
Table 9:
Figure imgf000061_0001
ED = Effective Dosage The compound (I-A) has been tested in parallel with a reference compound, the very potent topical glucocorticoid Clobetasol, which was examined for its inhibitory effects in the same concentration range as the compound (I-A). Compound (I-A) and Clobetasol reached almost similar, very low ED80 values for edema formation in the TMA-induced reaction in rats and mice (Mouse & Rat TMA-induced edema formation - Compound of Example 5 ED80: 0.00015% and 0.00052%, respectively vs. Clobetasol ED80: 0.00060% and 0.00077%, respectively). The inhibition of the granulocyte infiltration in rats did also not differ between the compound (I-A) and Clobetasol. However, while ED80 values in rats were reached at very low dosages (Rat TMA-induced granulocyte infiltration- Compound (I-A) ED80: 0.00049% vs. Clobetasol ED80: 0.00092%), higher dosages by a factor of ~8 were needed to reach ED80 in mice (Mouse TMA-induced granulocyte infiltration- Compound (I-A) ED80: 0.00860% vs. Clobetasol ED80: 0.00610%) (all ED80 values are also listed in Table 10).
Table 10:
Figure imgf000062_0001
ED = Effective Dosage In summary, both, the compound (I-A), as well as Clobetasol were able to completely inhibit the symptoms of the Th2-mediated TMA- or TDI-induced allergic contact dermatitis in rats and mice at dosages lower than 0.1%. The compound (I-A) does not differ in this animal model in its potency and efficacy from Clobetasol.
Overall, the compound (I-A) is not only able to reduce Thl and Th2 cytokine release in human lymphocytes in vitro, but is also able to attenuate Thl and Th2-mediated inflammatory skin reaction in rodents in a very potent and effective manner.
Derived from the in vitro characterization in human T cell and supported by the inhibition of T cell mediated inflammatory skin reactions in rodents, it can be assumed that the compounds for use in the topical treatment of inflammatory skin diseases according to the present invention and especially the compounds (I-A) are able to attenuate or even completely inhibit T-cell mediated inflammatory skin reactions in indications such as psoriasis, atopic dermatitis and allergic contact dermatitis.
For the therapeutic actions in the above-mentioned pathologic conditions, the suitable dose varies and depends on, for example, the active strength of the compound of general formula I, the host, the type of administration, and the type and severity of the conditions that are to be treated, as well as the use as a prophylactic agent or therapeutic agent.
In addition, the invention provides:
(i) The use of one of the compounds of formula I according to the invention or mixture thereof for the production of a medication for treating a dermato logical disease; (ii) A process for treating a dermatological diseases, said process comprises an
administration of an amount of the compound according to the invention, wherein the amount suppresses the disease and wherein the amount of compound is given to a patient who requires such a medication;
(iii) A pharmaceutical composition for treating a dermatological diseases, said treatment comprises one of the compounds according to the invention or mixture thereof and at least one pharmaceutical adjuvant and/or vehicle.
In general, satisfactory results can be expected in animals when the daily doses comprise a range of 1 μg to 100,000 μg of the compound according to the invention per kg of body weight. In the case of larger mammals, for example the human, a recommended daily dose lies in the range of 1 μg to 100,000 μg per kg of body weight. Preferred is a dose of 10 to 30,000 μg per kg of body weight, and more preferred is a dose of 10 to 10,000 μg per kg of body weight. For example, this dose is suitably administered several times daily. For treating acute shock (e.g., anaphylactic shock), individual doses can be given that are significantly above the above-mentioned doses. Pharmaceutical compositions:
This invention also relates to pharmaceutical compositions containing one of the forms of the compound of the formula (I) or a mixture thereof.
Respective mixtures with regard to the amounts of the Modification I, Modification II, and Mono- hydrate are already described above.
These compositions may be utilized to achieve the desired pharmacological effect by administration to a patient in need thereof. A patient, for the purpose of this invention, is a mammal, including a human, in need of treatment for the particular condition or disease. Therefore, the present invention includes pharmaceutical compositions which are comprised of a pharmaceutically acceptable carrier and a pharmaceutically effective amount of one of the forms of the compound of the formula (I) or a mixture thereof. A pharmaceutically acceptable carrier is any carrier which is relatively non-toxic and innocuous to a patient at concentrations consistent with effective activity of the active ingredient so that any side effects ascribable to the carrier do not vitiate the beneficial effects of the active ingredient. A pharmaceutically effective amount of compound is that amount which produces a result or exerts an influence on the particular condition being treated. The forms of the compound of the formula (I) of the present invention can be administered with pharmaceuti- cally-acceptable carriers well known in the art using any effective conventional dosage unit forms, including immediate, slow and timed release preparations, orally, parenterally, topically, nasally, ophthalmically, optically, sublingually, rectally, vaginally, and the like.
For oral administration, the form of the compound of the formula (I) of the present invention can be formulated into solid or liquid preparations such as solid dispersion, capsules, pills, tablets, troches, lozenges, melts, powders, solutions, suspensions, or emulsions, and may be prepared according to methods known to the art for the manufacture of pharmaceutical compositions. The solid unit dosage forms can be a capsule which can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers such as lactose, sucrose, cal- cium phosphate, and corn starch.
In another embodiment, the form of the compound of the formula (I) of the present invention may be tableted with conventional tablet bases such as lactose, sucrose and cornstarch in combination with binders such as acacia, corn starch or gelatin, disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch, alginic acid, corn starch, and guar gum, gum tragacanth, acacia, lubricants intended to improve the flow of tab- let granulation and to prevent the adhesion of tablet material to the surfaces of the tablet dies and punches, for example talc, stearic acid, or magnesium, calcium or zinc stearate, dyes, colouring agents, and flavouring agents such as peppermint, oil of wintergreen, or cherry flavouring, intended to enhance the aesthetic qualities of the tablets and make them more acceptable to the patient. Suitable excipients for use in oral liquid dosage forms include dicalcium phosphate and diluents such as water and alcohols, for example, ethanol, benzyl alcohol, and polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent or emulsifying agent. Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance tablets, pills or capsules may be coated with shellac, sugar or both.
Dispersible powders and granules are suitable for the preparation of an aqueous suspension. They provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example those sweetening, flavouring and colouring agents described above, may also be present.
For topical application, formulations in gels, ointments, fatty ointments, creams, pastes, powders, milk and tinctures are possible. The dosage of the compounds of general formula I should be 0.01%-20% in these preparations to achieve a sufficient pharmacological action.
The compositions of the invention can also contain other conventional pharmaceutically acceptable compounding ingredients, generally referred to as carriers or diluents, as necessary or desired. Conventional procedures for preparing such compositions in appropriate dosage forms can be utilized.
In the following, a preferred formulation being a waterless multi-phase gel system is described. The multi-phase gel system comprises an exterior lipid matrix and an interior phase coagulated by means of polymer, which is obtained by
a) melting the lipid phase to form a liquid lipid phase,
b) mixing and homogenizing swellable polymer or polymer mixture to form a dispersing pol- ymer phase,
c) dissolving the compound of the formula (I) in the Modification I, Modification II and/or Monohydrate in the polymer phase,
d) combining the polymer phase with the liquid lipid phase and homogenizing the phases and, e) stirring the phase mixture to form a solid gel-like mixture structure of the overall system.
It is surprising that the multi-phase gel system described above, which is preferably provided for application and infiltrate of hardly soluble at high concentration and/or hydrolysis-sensitive active ingredient in combination with the substance class of the compounds of the formula (I) in the Modification I, Modification II and/or Monohydrate, has unexpected advantageous properties.
So the developed multi-phase gel system with the compound of the formula (I) in the Modification I, Modification II and/or Monohydrate is characterized by an excellent storage-stability over a period of at least 24 months. The physical stability surprisingly outperforms the conventionally ointments and those parallel-developed corresponding cream formulations containing the same active ingredient.
In addition, the developed system is characterized by an excellent skin-compatibility. This is especially surprising, because the formulation does not contain insubstantial proportions of solvents such as propylene glycol and propylene carbonate. In studies on homo sapiens, the compatibility was identically good or even better than those of parallel-developed cream formulations containing a significantly small proportion of solvents.
In addition, the formulation expectedly exhibits a highly dermal availability in in vitro model in comparison to conventional systems.
In terms of the present invention, a multi-phase gel system is referred to as a system is formed of two or more phases.
A preferred system has a lipid phase which contains skin-compatible lipids.
It is particularly preferred that the lipids are selected from petrolatum, paraffin, beeswax, silicone oils and mixtures thereof.
A system is further preferred which contains polymers such as cellulose derivatives, acrylate polymers and their derivatives or mixture.
Particularly preferred is also that the cellulose derivative is hydroxypropyl cellulose.
In particular, it is also preferred that the acrylate polymer is cross-linked acrylate.
A system is further preferred that swellable polymers or polymer mixtures are swelled by means of OH-group-containing swelling agents.
It is preferred that the swelling agent is multivalent aliphatic alcohols (polyols) having a chain length of up to 3 carbon atoms or mixtures thereof.
In addition, according to the present invention, it is preferred that the polyol is propylene glycol. It is also preferred that the swelling agent further comprises carbonic acid diester or mixtures of carbonic acid diesters.
Here is particularly preferred that the carbonic acid diester is propylene carbonate.
The system is exceptionally preferred that the polymer phase contains the active ingredients. It is also preferred that the lipid phase and the polymer phase contain different active ingredients. A preferred system further comprises one or more additives which are useful for a topically applicable composition.
Another object of the present invention is the application of the above-mentioned system for preparing a pharmaceutical composition for utilization on skin, the mucous membranes and/or on wound surfaces.
Preferred is the application for manufacturing a human or veterinary medical product, also use for humans and for animals.
Moreover, a process for preparation of a waterless multi-phase gel system consisting of exterior lipid matrix and an interior phase coagulated by means of polymer, wherein the following steps are carried out is also provided:
a) melting the lipid phase to form a liquid lipid phase,
b) mixing and homogenizing swellable polymers or polymer mixtures to form a dispersing polymer phase,
c) dissolving the compound of the formula (I) in the Modification I, Modification II and/or Monohydrate in the polymer phase,
d) combining the polymer phase with the liquid lipid phase and homogenizing the phases e) stirring the phase mixture until forming the system of a solid gel-like mixed structure. In terms of waterless, up to 1% water may be present in the composition. Waterless solvents according to the invention may contain up to 5% water. For example, ethanol used in the present invention may contain up to 4.5% water (azeotrope).
The polymers, which are swellable through OH group, may be selected from acrylate polymers or the mixtures thereof. Several exemplary products from firm Noveon Inc. are referred to as: Carbopol 934 NF, Carbopol 934P NF, Carbopol 940 NF, Carbopol 97 IP NF, Carbo-pin 71G NF, Carbopol 974P NF, Carbopol 980 NF, Carbopol 981 NF, Carbopol 1342 NF, Carbopol 5984 EP, Pemulen TR-1 NF, Pemulen TR -2 NF, Noveon AA-1 USP, Noveon CA-1 USP and Noveon CA-2 USP.
In the context of the present invention, the term of short chain alcohols refer to mono- to tri valent aliphatic alcohols having up to five carbon atoms.
All process steps for producing the compositions according to the present invention can be carried out by means of techniques, which are familiar to the average person skilled in the art. The mixing and homogenizing the components in step b) for producing the dispersing polymer phase can be performed by conventional mixing systems and homogenizers.
The adjustment of the viscosity of the prepared polymer phase can be done for example by heating. In addition to the already-specified ingredients, composition according to the present invention may comprise further one or more useful additives for application as a topically applicable composition. These additives may be selected, for example, from dyes, odorous substances, preservatives and absorption-supporting agents.
Dosage of the pharmaceutical compositions of the present invention:
Based upon standard laboratory techniques known to evaluate compounds useful for the treatment of disorders, by standard pharmacological assays for the determination of treatment of the conditions identified above in mammals, and by comparison of these results with the results of known medica- ments that are used to treat these conditions, the effective dosage of the compounds of this invention can readily be determined for treatment of each desired indication. The amount of the active ingredient to be administered in the treatment of one of these conditions can vary widely according to such considerations as the particular compound and dosage unit employed, the mode of administration, the period of treatment, the age and sex of the patient treated, and the nature and extent of the condition treated.
The total amount of the active ingredient to be administered will generally range from about 0.0001 mg/kg to about 20 mg/kg, and preferably from about 0.001 mg/kg to about 2 mg/kg body weight per day. A unit dosage may contain from about 0.05 mg to about 150 mg of active ingredient, and can be administered one or more times per day.
Of course the specific initial and continuing dosage regimen for each patient will vary according to the nature and severity of the condition as determined by the attending diagnostician, the activity of the specific compound employed, the age and general condition of the patient, time of administration, route of administration, rate of excretion of the drug, drug combinations, and the like. The desired mode of treatment and number of doses of a compound of the present invention or a pharmaceutically acceptable salt or ester or composition thereof can be ascertained by those skilled in the art using conventional treatment tests.
The weight data in the tests and examples which follow are, unless stated otherwise, percentages by weight; parts are parts by weight. Solvent ratios, dilution ratios and concentration data of liquid/liquid solutions are based on each case on the volume.
Topical formulations for in- vivo studies
multi-Phase-Gel system containing 0.1% 5-{[(lS,2S)-l-(2-Chloro-3-fluoro-4-methoxyphenyl)- 3 ,3 J-trifluoro-2-hydroxy-2-(methoxymethyl)propyl]amino}-7-fluoro-lH-quinolin-2-on (= compound of formula I-A (formulation A) component weight.- %
5-{[(lS,2S)-l-(2-Chloro-3-fiuoro-4- methoxyphenyl)-3 ,3 ,3-trifluoro-2- hydroxy-2- 0.1
(methoxymethyl)propyl] amino } -7- fluoro-lH-quinolin-2-on
Carbomer Copolymer
0.21
(Pemulen TR-l/TR-2)
Hydro xypropylcellulo se
0.056
(Klucel HF)
Propylencarbonat 6.9
Propylenglykol 6.067
Natriumhydroxid
0.85
(2% in Propylenglykol)
WeiBes Vaselin 63.817
Paraffinol, dickflussig 10.0
Cyclomethicon 3.0
Bee wax (Cera Alba) 2.0
Hartparaffin 7.0 lipophilic cream containing - 0.1% compound of formula I- A (formulation B
Figure imgf000071_0001
multi-Phase-Gel System containing 0.01% 5-{[(lS,2S)-l-(2-Chloro-3-fluoro-4-methoxyphenyl)- 3 ,3 ,3-trifluoro-2-hydroxy-2-(methoxymethyl)propyl]amino} -7-fluoro- lH-quinolin-2-on (= com- pound of formula I-A) (formulation E)
component weight-%
5- { 1 S,2S) [ 1 -(2-Chloro-3-fiuoro-4- methoxyphenyl)-3 ,3 ,3-trifluoro-2- hydroxy-2- 0.01
(methoxymethyl)propyl] amino } -7- fluoro-lH-quinolin-2-on
Carbomer Copolymer
0.21
(Pemulen TR-l/TR-2) Hydro xypropylcellulo se
0.056
(Klucel HF)
Propylencarbonat 6.9
Propylenglykol 6.067
Natriumhydroxid
0.85
(2% in Propylenglykol)
WeiBes Vaselin 63.907
Paraffinol, dickflussig 10.0
Cyclomethicon 3.0
Bee wax (Cera Alba) 2.0
Hartparaffin 7.0
Lipophilic cream containing 0.01% compound of formula I- A (formulation F
Figure imgf000072_0001
Impact on psoriatic Symptoms in the open Psoriasis Plaque Test
The compound of Formula I-A is also able to reduce psoriatic symptoms in the psoriasis plaque test, a standardized proof-of-concept model, which represents a useful method for predicting treat- ment efficacy in psoriasis in the early clinical stage of development.
The psoriasis plaque test (PPT) was developed initially to compare simultaneously the anti- psoriatic activity of different topical glucocorticoids and the impact of different topical formulation types on their performance in a limited number of psoriasis patients (Dumas and Scholz, 1972, Katz M et al, 2000). In the meantime the psoriasis plaque test has been further modified and repre- sents now a valuable proof-of-concept model for the intra- individual comparison between different topical psoriasis treatments, which is also characterized by a low risk of adverse events as only a limited amount of the study drugs is applied to the psoriatic lesions (Wozel G, 2006, Queille- Roussel et al. 2013).
While most PPTs were carried out in an occlusive study design, the anti-psoriatic activity of com- pound of Formula I-A has been assessed in an open manner to be able to distinguish between different types of formulations in a situation which depicts the real clinical situation more closely than the occlusive design.
In a double-blind, reference-controlled, single center, open PPT in 22 symptomatic adult volunteers with stable, untreated plaque psoriasis the efficacy of compound of Formula I-A at two different concentrations, 0.1% and 0.01%, in two different topical formulation types, a water- free multiphase system and a lipophilic cream, was compared to the efficacy of commercially available 0.05% Clobetasol Propionate in a conventional ointment formulation type.
On day 1 (baseline) one test field per treatment (field size: 3 cm2) was defined on the previously determined comparatively large target plaque of each symptomatic volunteer. The exact localiza- tion of each test field was confirmed at each visit using a stencil made from transparent foil during the test field determination prior to the first treatment. The distance between two test fields was at least 2cm. 10 μΐ of each of the five treatments, 0.1% compound of Formula I-A in a water- free multiphase system (formulation A), 0.01% compound of Formula I-A in a water- free multiphase system (formulation E), 0.1% compound of Formula I-A in a lipophilic cream (formulation B), 0.01% compound of Formula I-A in a lipophilic cream (formulation F) and 0.05% Clobetasol Propionate in a conventional ointment formulation type (reference), was applied to its pre-defined test field. Application of the treatments was performed by study personnel once daily on 6 days per week for 4 weeks.
In the early PPT models the anti-psoriatic activity was determined by grading the intensity of symptoms by the investigator's assessment. Recently skin thickening together with the typical in- flammatory cellular infiltration, which represent very prominent clinical signs of psoriasis (hereafter defined as "infiltrate thickness"), is mainly determined with 22 MHz sonography. This method represents a more sensitive efficacy measurement for the PPT compared to the clinical grading. Changes in skin thickness combined with the extent of inflammatory infiltration are depicted by an increase or decrease of an echo-poor band. Depending on the treatment success the size of the echo-poor band should increasingly decrease during the treatment period.
The mean absolute values (± SD) for infiltrate thickness at day 1, 4, 8, 11, 15, 18, 22 and 29 of the treatment period as well as the infiltrate thickness at day 43 and 57 of the post-treatment period are graphically depicted in Figure 1. It can be derived from Figure 1, that the compound of Formula I- A and the potent topical glucocorticoid, Clobetasol, reduce infiltrate thickness to an increasing ex- tent over the treatment period of 29 days.
The reduction of psoriatic symptoms by compound of Formula I-A is under the given circumstances less prominent compared to Clobetasol: at day 29 the reduction of infiltrate thickness caused by 0.1% compound of Formula I-A in the water-free multiphase system reaches -60% of the reduction caused by 0.05% Clobetasol propionate in a conventional ointment formulation type. Although only two doses were examined, dose dependency can be observed: the higher concentration (0.1 %) of the compound of Formula I-A reaches in the water- free multiphase system -60% and in the lipophilic cream -40% of the reduction of infiltrate thickness which is reached by the Clobetasol reference, while the 10-times lower concentration of 0.01% leads only to -50% reduction in the water-free multiphase system and to 20% reduction in the lipophilic cream compared to the reduction obtained by the reference. The type of the formulation seems also to influence the antipsoriatic activity of compound of Formula I-A: the lower 0.01% concentration of Compound of Formula I-A in the water- free multiphase system was still able to reduced infiltrate thickness at day 29 by -50% compared to Clobetasol Propionate in the ointment formulation type, while the lower 0.01% concentration of compound of Formula I-A in the lipophilic cream was significantly weaker with only 20% reduction. Results of the statistical testing of the pairwise comparison of the change in infiltrate thickness over the treatment period are shown in Table 11. Statistical tests were 2-sided, Confidence Intervals (CI) and p-values were determined via the 2-factorial analysis of variance. Table 11 :
Figure imgf000075_0001
LS = Least Square Method derived from Regression Analysis makes use of the best fit which minimizes the sum of squared residuals,
i.e. the difference between the observed value, and the fitted value provided by this model).
CI = Confidence Interval
The pairwise comparison over the treatment period of 29 day also shows that 0.1% compound of Formula I-A in the water-free multiphase system exerts - with a mean difference of the infiltrate thickness of 132 μιη compared to Clobetasol - the most potent anti-psoriatic activity, followed by the lower concentration, 0.01%, in the water- free multiphase system, with a difference of 170 μιη compared to the reference. Both concentrations of compound of Formula I-A in the lipophilic cream formulation type were weaker with differences of infiltrate thickness of 182 μιη and 239 μιη compared to the reference.
In summary, by using the established Psoriasis Plaque Test model it was possible to demonstrate that the compound of Formula I-A is able to reduce increasingly psoriatic symptoms over a daily treatment period of 29 days in patients with stable plaque psoriasis. Derived from this early clinical proof-of-concept it can be deduced that compound of Formula I-A exerts meaningful therapeutic effects in the T-cell mediated skin disease psoriasis. Improved side effect profile compared to the topical glucocorticoid Clobetasol
At therapeutically active dosages topical glucocorticoids can not only induce local side effects such as skin atrophy, but can also frequently affect the Hypothalamic-pituitary-adrenal (HP A) axis and can cause disorders such as iatrogenic Cushing's syndrome, adrenal insufficiency, thymus atrophy and growth retardation or even growth inhibition during long-term therapy (Robertson DB and Maibach HI, 1982, Schaecke H et al, 2002). The compound (I-A) differs from topical glucocorticoids as it is characterized by a significantly reduced or even absent potential to induce glucocorti- coid-related adverse events at therapeutically active dosages.
As not only anti-inflammatory and immunomodulatory activities of topical glucocorticoids but also local as well as systemic side effects can be ascertained in animal models, the compound (I-A) was examined for its potential to induce various glucocorticoid-related side effects. As a reference compound the very potent glucocorticoid Clobetasol was chosen.
Skin atrophy (i.e. the decrease in skin thickness) which is characterized by thin, fragile and transparent skin, occurs frequently as a cutaneous complication of the topical application of very potent glucocorticoids such as Clobetasol. Juvenile hairless (hr/fir) rats represent an especially suited animal model for skin atrophy. After daily topical applications of test compounds for 25 days, the skin fold thickness on day 25 compared to the skin fold thickness of day 1 (before the initiation of the treatment) served as an easily determined parameter for the impact of the topical treatment on skin thickness. Skin breaking strength examined on day 25 with skin of sacrificed rats served as a sensi- tive parameter for the effects of the topical treatment on collagen fibers disintegration or degradation which also contributed to skin atrophy.
In comparison to vehicle-treated skin, the once daily treatment of rat skin for 25 days with dosages of the compound (I-A), which are able to reduce the Croton-oil induced skin inflammation (J. Exp. Med. 1995, 182, 99-108) in rats by 80% or 100%, induced only a limited reduction in skin thick- ness (-31% and -41%, respectively), while Clobetasol at equi-effective dosages induced a more pronounced reduction of skin thickness (-49% and -54%, respectively) (see also Table 12).
The compound (I-A) had also a more limited effect on the collagen disintegration than Clobetasol, as it reduced the skin breaking strength after 25 day of daily application to a lower extent compared to Clobetasol. Table 12:
Figure imgf000077_0001
ED80 = Effective Dosage inducing 80% inhibition of Croton-oil induced
skin inflammation in rats,
ED 100 = Effective Dosage inducing complete inhibition of Croton-oil induced skin inflammation in rats.
The impact of the topical treatment on the growth of juvenile rats was monitored by periodically weighing the animals over time. Daily topical application of therapeutically active dosages of the compound (I-A) for 25 days had no impact on the growth of juvenile rats. The increase in animal weight did not differ between animals treated with the compound (I-A) and vehicle-treated rats over a period of the 25 days, while rats treated with therapeutically active dosages of Clobetasol showed significant growth retardation. The daily application of the Clobetasol dosage which is able to induce complete inhibition of an inflammatory skin reaction after a single administration, vented rat weight gain during the 25 days treatment period completely (see also Table 13).
Table 13 :
Figure imgf000078_0001
ED80 = Effective Dosage inducing 80% inhibition of Croton-oil induced skin inflammation in rats,
ED 100 = Effective Dosage inducing complete inhibition of Croton-oil induced skin inflammation in rats.
The thymus, which is the central organ for the constitution of the adaptive immune system, is a very glucocorticoid-sensitive organ. It responds to glucocorticoid exposure by rapid apoptotic death which might have fatal consequences on the adaptive immune system and might also lead to the induction of autoimmunity. Accordingly, thymus reduction is also an excellent indicator for systemic side effects of glucocorticoid-receptor ligands.
Glucocorticoids are also able to induce apoptosis of T and B lymphocytes, which can be measured by the weight reduction of secondary lymphatic organs such as the spleen. Suppression of the hypothalamic-pituitary-adrenal (HP A) axis is one of the most important side- effects of glucocorticoids as it affects the central neuro-endocrine circuit which is responsible for numerous physiological processes. Once endogenous Cortisol is replaced by exogenous glucocorticoids, Cortisol production, or production of precursors or inducers, is down-regulated. This is asso- ciated with a degeneration of the cortisol-producing organ, the adrenal gland.
Similar to the repeated topical application of vehicle, the daily topical application of therapeutically active dosages of the compound (I-A) for 25 days did neither reduce thymus, nor spleen, nor adrenal gland weight. On the contrary, the topical application of therapeutically active dosages of Clobetasol induced a significant reduction of the weight of the immune organs and the adrenal glands (see also table 14).
Table 14:
Impact on Organs of the adaptive Immune System and the HP A- Axis:
Comparison between Formula I-A and Clobetasol
Reduction of Reduction of Reduction of Thymus Weight Spleen Weight Adrenal Weight
Compound Compound Compound
Parameter
of of of
Clobetasol Clobetasol Clobetasol
Formula I- Formula I- Formula I- A A A
Once daily application
ofED80 for 21 days
No ReducNo ReducNo Reduc(ED80I_A: 0.0062%) -49% -16% -19% tion tion tion
(ED80ciobetasoi:
0.0044%)
Once daily application
of ED 100 for 21 days
No ReducNo ReducNo Reduc(ED100I-A: 0.01%) -81% -35% -40% tion tion tion
(ED l OOdobetasoi:
0.01%) HPA = hypothalamic-pituitary-adrenal axis,
ED80 = Effective Dosage inducing 80% inhibition of Croton-oil induced
skin inflammation in rats,
ED 100 = Effective Dosage inducing complete inhibition of Croton-oil induced skin inflammation in rats.
In summary, the compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases according to the present invention have a reduced potential to induce unwanted effects on rodent skin after topical administration compared to the very potent topical glucocorticoid Clobeta- sol. This was demonstrated by a more limited effect on skin thickness and breaking strength than Clobetasol. Furthermore, the compounds for use in the topical treatment of inflammatory skin diseases according to the present invention do not reduce thymus, spleen, adrenal gland weight, while the topical glucocorticoid Clobetsol significantly reduced the weight of these immune organs. These observations clearly indicate an improved side effect profile of the said compounds compared to conventional topical glucocorticoids.
Literature:
Biedermann T et al: TH1 and TH2 Lymphocyte Development and Regulation of TH Cell- Mediated Immune Responses of the Skin. J. Investig. Dermatol. Symp. 9: 5-14, 2004.
Dumas KJ and Scholz JR. The psoriasis bio-assay for topical corticosteroid activity. Acta Derm Venereol 52: 43-48, 1972
Fitch E et al: Pathophysiology of Psoriasis: Recent Advances on IL-23 and Thl7 Cytokines. Curr. Rheumatol. Rep 9(6): 461-467, 2007.
Ghoreschi K et al: Immunopathogenesis and role of T cells in psoriasis. Clin. Dermatol. 25:574- 580, 2007.
Gober MD & Gaspari AA: Allergic Contact Dermatitis. Curr Dir Autoimmun 10: 1-26, 2008.
Hanifin JM and Rajka G.: Diagnostic Features of Atopic Dermatitis. Acta. Dermatovener (Stockholm) Suppl. 92: 44-47, 1980.
Hayashi M et al: Assessment of Preferential Thl or Th2 Induction by Low-Molecular- Weight Compounds Using a Reverse Transcription-Polymerase Chain Reaction Method: Comparison of Two Mouse Strains, C57BL/6 and Balb/c. Toxicol. & Applied Pharmacol 177: 38-45, 2001.
Hengge UR et al: Adverse effects of topical glucocorticosteroids. J. Am. Acad. Dermatol. 54: 1- 15, 2006.
Katz M et al. Scholtz-Dumas psoriasis small plaque bioassay. J Dermatol Treatmentl l : 15-19, 2000.
Leung DYM et al: New insights into atopic dermatitis. J. Clin. Invest. 113: 654-657, 2004
McDevitt HO: Discovering the role of the Major Histocomatibility Complex in the immune response. Annu. Rev. Immunol. 18: 1-17, 2000.
Nestle FO et al. Mechanisms of Disease: Psoriasis, N. Engl. J. Med. 361(5):496-509, 2009.
Numerof RP and Asadullah K: Cytokine and Anti-Cytokine Therapies for Psoriasis and Atopic Dermatitis. Biodrugs 20(2): 93-103, 2006.
Ong PY: Emerging drugs for atopic dermatitis. Expert Opinion Emerg. Drugs 14(1): 165-179, 2000.
Queille-Roussel C et al. Use of a psoriasis plaque test in the development of a gel formulation of of calcipotriol and betamethasone dipropionate for scalp psoriasis. J Dermatol Treatment 24: 250-254, 2013. Robertson DB and Maibach HI: Topical Corticosteroids. Int. J. Dermatol. 21 : 59-67, 1982.
Schaecke H et al: Mechanisms involved in the side effects of glucocorticoids. Pharmacology & Therapeutics 96: 23-43, 2002.
Schaecke H et al: Dissociation of transactivation from transrepression by a selective glucocorti- coid receptor agonist leads to separation of therapeutic effects from side effects. PNAS 101(1): 227-232, 2004.
Schaecke H et al: Insight into the molecular mechanisms of glucocorticoid receptor action promotes identification of novel ligands with an improved therapeutic index. Exp. Dermatol. 15: 565- 573, 2006.
Schaecke H et al: Selective glucocorticoid receptor agonists (SEGRAs): Novel ligands with an improved therapeutic index. Mol. Endocrinol. 275: 101-117, 2007.
Stahn C et al: Molecular mechanisms of glucocorticoid action and selective glucocorticoid receptor agonists. Mol. Cellular Endocrinol. 275: 71-78, 2007.
Takeshita K et al: Essential role of MHC II-independent CD4+ T cells, IL-4 and STAT6 in contact hypersensitivity induced by fluorescein isothiocyanate in the mouse. Int. Immunol. 16(5): 685-695, 2004.
Victor FC et al: Changing Paradigms in Dermatology: Tumor Necrosis Factor Alpha (TNF-) Blockade in Psoriasis and Psoriatic Arthritis. Clinics in Dermatology 21 : 392-397, 2003.
Wozel G. Is the psoriasis plaque test still relevant in the age of biologies? Hautarzt 57: 672-678, 2006.

Claims

Claims
Compounds according to general Formula (I) for use in the topical treatment of T-cell diated inflammatory skin diseases,
Formula (I)
Figure imgf000083_0001
selected from the group consisting of
5- { [( 1 S,2S)- 1 -(2-chloro-3-fluoro-4-methoxyphenyl)-3 ,3 ,3-trifluoro-2-hydroxy-2- (methoxymethyl)propyl]amino}-7-fluoro-lH-quinolin-2-one (I -A)
Figure imgf000083_0002
-A
5-{(lS,2S)[l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- (hydroxymethyl)propyl]amino}-7-fluoro-lH-quinolin-2-one (I-B)
Figure imgf000084_0001
l-D
5-{(lS,2R)[l-(2-chloro-3-fluoro-4-methoxyphenyl)-3,3,3-trifluoro-2-hydroxy-2- ({methylsulfanyl}methyl)propyl]amino}-7-fluoro-lH-quinolin-2-one (I-C)
Figure imgf000084_0002
l-C and their salts, solvates or salts of solvates.
2. Compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases according to claim 1, characterized in that the said compounds are nonsteroidal selective glucocorticoid receptor agonists.
3. Compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases according to claims 1 or 2, characterized in that the said compounds are able to inhibit the proliferation of stimulated human lymphocytes, demonstrated in a suited in vitro test system which made use of compound doses ranging between 10"6 and 10"12 mol/L and showed potent inhibition of lymphocyte proliferation in the nanomolar range.
Compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases according to any of claims 1 to 3, characterized in that the said compounds are able to inhibit the release of cytokines such as IL-2, IL-4, and IL-23 which induce the proliferation of certain sub-sets of T cells associated with inflammatory reactions.
Compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases according to any of claims 1 to 4, characterized in that the said compounds are able to inhibit the release of cytokines including IL-12, TNF-a or IFN-γ, which exert immuno stimulatory and pro -inflammatory effects.
Compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases according to any of claims 1 to 5, characterized in that the inhibition of cytokine release and related proliferation of T cells is involved in the induction and perpetuation of psoriasis.
Compounds for use in the topical treatment of inflammatory skin diseases according to any of claims 1 to 6, wherein the said inflammatory skin diseases include psoriasis.
Compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases according to claims 1 to 7, wherein the said skin disease, psoriasis, includes the subclasses psoriasis vulgaris, plaque-type psoriasis, and pustular psoriasis.
Compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases according to any of claims 1 to 5, wherein the said inflammatory skin diseases include atopic dermatitis and allergic contact dermatitis.
Compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases according to any of claims 1 to 5, characterized in that the said compounds are able to inhibit T-cell induced inflammatory skin reactions in rodent models such as DNFB-induced or TMA-induced allergic contact dermatitis reactions.
Compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases according to any of claims 1 to 10, characterized in that the said compounds are characterized either by a lack of or by a reduced level of side effects typically caused by topical glucocorticoids.
Compounds for use in the topical treatment of T-cell mediated inflammatory skin diseases according to claim 11 , characterized in that the said compounds are marked by a lack of or by a reduced level of side effects typically caused by topical glucocorticoids, which include skin atrophy, Hypothalamic-pituitary-adrenal (HP A) axis suppression, thymus and spleen atrophy or growth retardation.
13. Compounds for use in the topical treatment of inflammatory skin diseases according to any of claims 1 to 12, characterized in that the said compounds are used for inflammatory skin diseases such as psoriasis, atopic dermatitis and allergic contact dermatitis in adults, adolescents, children and infants.
14. Compounds for use in the topical treatment of inflammatory skin diseases according to any of claims 1 to 13, wherein the said compounds do not have side effects or have a reduced level of side effects typically caused by topical glucocorticoids and can be used especially for inflammatory skin diseases in children and infants.
PCT/EP2017/072626 2016-09-08 2017-09-08 Quinoline derivatives for treatment of inflammatory skin diseases WO2018046685A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP16187755.0 2016-09-08
EP16187755 2016-09-08

Publications (1)

Publication Number Publication Date
WO2018046685A1 true WO2018046685A1 (en) 2018-03-15

Family

ID=56893806

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2017/072626 WO2018046685A1 (en) 2016-09-08 2017-09-08 Quinoline derivatives for treatment of inflammatory skin diseases

Country Status (1)

Country Link
WO (1) WO2018046685A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020152193A1 (en) 2019-01-22 2020-07-30 Akribes Biomedical Gmbh Selective glucocorticoid receptor modifiers for treating impaired skin wound healing

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009065503A1 (en) 2007-11-22 2009-05-28 Bayer Schering Pharma Aktiengesellschaft 5-[(3,3,3-trifluoro-2-hydroxy-1-arylpropyl)amino]-1h-quinolin-2-ones, a process for their production and their use as anti-inflammatory agents
WO2010009814A1 (en) * 2008-07-21 2010-01-28 Bayer Schering Pharma Aktiengesellschaft 5-[ (3,3,3-trifluoro-2-hydroxy-1-arylpropyl) amino]-1-arylquinolin-2-ones, a process for their production and their use as anti-inflammatory agents

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009065503A1 (en) 2007-11-22 2009-05-28 Bayer Schering Pharma Aktiengesellschaft 5-[(3,3,3-trifluoro-2-hydroxy-1-arylpropyl)amino]-1h-quinolin-2-ones, a process for their production and their use as anti-inflammatory agents
EP2234979A1 (en) 2007-11-22 2010-10-06 Bayer Schering Pharma Aktiengesellschaft 5-ý(3,3,3-trifluoro-2-hydroxy-1-arylpropyl)amino¨-1h-quinolin-2-ones, a process for their production and their use as anti-inflammatory agents
WO2010009814A1 (en) * 2008-07-21 2010-01-28 Bayer Schering Pharma Aktiengesellschaft 5-[ (3,3,3-trifluoro-2-hydroxy-1-arylpropyl) amino]-1-arylquinolin-2-ones, a process for their production and their use as anti-inflammatory agents

Non-Patent Citations (29)

* Cited by examiner, † Cited by third party
Title
BIEDERMANN T ET AL.: "TH1 and TH2 Lymphocyte Development and Regulation of TH Cell-Mediated Immune Responses of the Skin. J", INVESTIG. DERMATOL. SYMP., vol. 9, 2004, pages 5 - 14, XP009119970, DOI: doi:10.1111/j.1087-0024.2004.00829.x
BRANCA ET AL., CHIMIA, vol. 49, no. 10, 1995, pages 381 - 385
DUMAS KJ; SCHOLZ JR: "The psoriasis bio-assay for topical corticosteroid activity", ACTA DERM VENEREOL, vol. 52, 1972, pages 43 - 48
FITCH E ET AL.: "Pathophysiology of Psoriasis: Recent Advances on IL-23 and Thl7 Cytokines", CURR. RHEUMATOL. REP, vol. 9, no. 6, 2007, pages 461 - 467, XP008089113, DOI: doi:10.1007/s11926-007-0075-1
GHORESCHI K ET AL.: "Immunopathogenesis and role of T cells in psoriasis", CLIN. DERMATOL., vol. 25, 2007, pages 574 - 580, XP022342594, DOI: doi:10.1016/j.clindermatol.2007.08.012
GOBER MD; GASPARI AA, ALLERGIC CONTACT DERMATITIS. CURR DIR AUTOIMMUN, vol. 10, 2008, pages 1 - 26
HANIFIN JM; RAJKA G: "Diagnostic Features of Atopic Dermatitis", ACTA. DERMATOVENER, vol. 92, 1980, pages 44 - 47
HAYASHI M ET AL.: "Assessment of Preferential Thl or Th2 Induction by Low-Molecular-Weight Compounds Using a Reverse Transcription-Polymerase Chain Reaction Method: Comparison of Two Mouse Strains, C57BL/6 and Balb/c", TOXICOL. & APPLIED PHARMACOL, vol. 177, 2001, pages 38 - 45
HENGGE UR ET AL.: "Adverse effects of topical glucocorticosteroids", J. AM. ACAD. DERMATOL., vol. 54, 2006, pages 1 - 15, XP005219461, DOI: doi:10.1016/j.jaad.2005.01.010
J. EXP. MED., vol. 182, 1995, pages 99 - 108
KATRITZKY ET AL., J. ORG. CHEM., vol. 60, 1995, pages 7631 - 7640
KATZ M ET AL.: "Scholtz-Dumas psoriasis small plaque bioassay", J DERMATOL TREATMENTLL, vol. 15-19, 2000
LEUNG DYM ET AL.: "New insights into atopic dermatitis", J. CLIN. INVEST., vol. 113, 2004, pages 654 - 657
LIST ET AL., ANGEW. CHEM., vol. 117, 2005, pages 7590 - 7593
MCDEVITT HO: "Discovering the role of the Major Histocomatibility Complex in the immune response", ANNU. REV. IMMUNOL., vol. 18, 2000, pages 1 - 17
NESTLE FO ET AL.: "Mechanisms of Disease: Psoriasis", N. ENGL. J. MED., vol. 361, no. 5, 2009, pages 496 - 509
NUMEROF RP; ASADULLAH K: "Cytokine and Anti-Cytokine Therapies for Psoriasis and Atopic Dermatitis", BIODRUGS, vol. 20, no. 2, 2006, pages 93 - 103
ONG PY: "Emerging drugs for atopic dermatitis", EXPERT OPINION EMERG. DRUGS, vol. 14, no. 1, 2000, pages 165 - 179, XP009171986, DOI: doi:10.1517/14728210902721248
QUEILLE-ROUSSEL C ET AL.: "Use of a psoriasis plaque test in the development of a gel formulation of of calcipotriol and betamethasone dipropionate for scalp psoriasis", J DERMATOL TREATMENT, vol. 24, 2013, pages 250 - 254
ROBERTSON DB; MAIBACH HI: "Topical Corticosteroids", INT. J. DERMATOL., vol. 21, 1982, pages 59 - 67
SAVERIA PASTORE ET AL: "Redox Imbalance in T Cell-Mediated Skin Diseases", MEDIATORS OF INFLAMMATION., vol. 2010, 1 January 2010 (2010-01-01), GB, pages 1 - 9, XP055412332, ISSN: 0962-9351, DOI: 10.1155/2010/861949 *
SCHAECKE H ET AL.: "Dissociation of transactivation from transrepression by a selective glucocorticoid receptor agonist leads to separation of therapeutic effects from side effects", PNAS, vol. 101, no. 1, 2004, pages 227 - 232, XP002392303, DOI: doi:10.1073/pnas.0300372101
SCHAECKE H ET AL.: "Insight into the molecular mechanisms of glucocorticoid receptor action promotes identification of novel ligands with an improved therapeutic index", EXP. DERMATOL., vol. 15, 2006, pages 565 - 573
SCHAECKE H ET AL.: "Mechanisms involved in the side effects of glucocorticoids", PHARMACOLOGY & THERAPEUTICS, vol. 96, 2002, pages 23 - 43, XP002715366, DOI: doi:10.1016/S0163-7258(02)00297-8
SCHAECKE H ET AL.: "Selective glucocorticoid receptor agonists (SEGRAs): Novel ligands with an improved therapeutic index", MOL. ENDOCRINOL., vol. 275, 2007, pages 101 - 117
STAHN C ET AL.: "Molecular mechanisms of glucocorticoid action and selective glucocorticoid receptor agonists", MOL. CELLULAR ENDOCRINOL., vol. 275, 2007, pages 71 - 78, XP022206555, DOI: doi:10.1016/j.mce.2007.05.019
TAKESHITA K ET AL.: "Essential role of MHC II-independent CD4+ T cells, IL-4 and STAT6 in contact hypersensitivity induced by fluorescein isothiocyanate in the mouse", INT. IMMUNOL., vol. 16, no. 5, 2004, pages 685 - 695
VICTOR FC ET AL.: "Changing Paradigms in Dermatology: Tumor Necrosis Factor Alpha (TNF-) Blockade in Psoriasis and Psoriatic Arthritis", CLINICS IN DERMATOLOGY, vol. 21, 2003, pages 392 - 397
WOZEL G: "Is the psoriasis plaque test still relevant in the age of biologies?", HAUTARZT, vol. 57, 2006, pages 672 - 678, XP019393183, DOI: doi:10.1007/s00105-006-1182-z

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020152193A1 (en) 2019-01-22 2020-07-30 Akribes Biomedical Gmbh Selective glucocorticoid receptor modifiers for treating impaired skin wound healing

Similar Documents

Publication Publication Date Title
JP6254138B2 (en) Use of PUFA to treat skin inflammation
CN107709290A (en) Selective androgen receptor degradation agent (SARD) part and its application method
CN102408429A (en) Condensed imidazolo derivatives for the inhibition of aldosterone synthase and aromatase
US11827605B2 (en) Isoquinoline compounds and their use in treating AhR imbalance
KR20180095070A (en) Polymorphic crystalline forms of obetic acid
JPWO2003082820A1 (en) (1-Indanone)-(1,2,3,6-tetrahydropyridine) derivative
JPS62145019A (en) Anti-inflammatory agent
WO2018046685A1 (en) Quinoline derivatives for treatment of inflammatory skin diseases
Fieser et al. Action of Methylmagnesium Iodide on Cholesterol α-Oxide and 6-Ketocholestanol
JPS60209582A (en) Synthesis of ddb and method of pharmaceutical manufacture
JP2024510778A (en) 1,3-substituted cyclobutyl derivatives and uses thereof
US20220117949A1 (en) Lta4h inhibitors for the treatment of hidradenitis suppurativa
US20180118707A1 (en) Beta-naphthoisoflavones, compositions containing, and uses of, same
WO2021242758A1 (en) Improved nitro-fatty acid oral dose regimens
TW201829387A (en) Morphinan derivatives and compositions comprising the same for treating autoimmune, inflammation or infection related disorders
JP2019131563A (en) Tetrahydro-isohumulone derivatives, and methods of making and using the same
Kadaba New compounds: Convenient selective esterification of aromatic carboxylic acids bearing other reactive groups using a boron trifluoride etherate—alcohol reagent
KR20150026640A (en) Novel pseudoceramide derivatives and pharmaceutical or cosmetic composition containing
US3384662A (en) 1, 1-diphenyl-1-methoxy-3-benzylaminopropane and the salts thereof
Fan et al. An emerging principle: selective nuclear receptor modulators
TW201102372A (en) Indane analogs and use as pharmaceutical agents and process of making
JP5689191B1 (en) Process for producing 4-ethylidene-3,4-dihydro-2H-pyrrole
CA3217096A1 (en) Contraceptive compounds and methods
CA1108158A (en) N-(1'allyl 2'-pyrrolidylmethyl) 2,3-dimethoxy 5- sulfamoyl benzamide
JP2021522338A (en) Preparation method of strong thiazole compound, pharmaceutical preparation and its use

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: 17761545

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: 17761545

Country of ref document: EP

Kind code of ref document: A1