Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
NOVEL TETRAZOLES
Document Type and Number:
WIPO Patent Application WO/2021/074198
Kind Code:
A1
Abstract:
This invention relates to tetrazoles and their use as inhibitors of TRPA1 activity, pharmaceutical compositions containing the same, and methods of using the same as agents for treatment and/or prevention of fibrotic diseases, inflammatory and auto-immune diseases and CNS-related diseases.

Inventors:
FLECK MARTIN (DE)
BINDER FLORIAN (DE)
DAHMANN GEORG (DE)
HEHN JOERG (DE)
LESSEL UTA (DE)
WILLWACHER JENS (DE)
Application Number:
PCT/EP2020/078856
Publication Date:
April 22, 2021
Filing Date:
October 14, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BOEHRINGER INGELHEIM INT (DE)
International Classes:
A61K31/522; A61K31/53; A61P13/00; A61P17/00; A61P29/00
Domestic Patent References:
WO2017060488A12017-04-13
WO2015155306A12015-10-15
Other References:
LAURIE B. SCHENKEL ET AL: "Optimization of a Novel Quinazolinone-Based Series of Transient Receptor Potential A1 (TRPA1) Antagonists Demonstrating Potent in Vivo Activity", JOURNAL OF MEDICINAL CHEMISTRY, vol. 59, no. 6, 24 March 2016 (2016-03-24), US, pages 2794 - 2809, XP055373392, ISSN: 0022-2623, DOI: 10.1021/acs.jmedchem.6b00039
Attorney, Agent or Firm:
SIMON, Elke, Anna, Maria et al. (DE)
Download PDF:
Claims:
CLAIMS

1. A compound selected from the group consisting of

2. A salt, particularly a pharmaceutically acceptable salt of a compound according to claim 1

3. A pharmaceutical composition comprising at least one compound according to claim 1 or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable excipient.

4. The compound according to claim 1, or a pharmaceutically acceptable salt thereof for use as a medicament.

5. The compound according to claim 1, or a pharmaceutically acceptable salt thereof for the treatment or prevention of inflammatory airway diseases or fibrotic diseases or cough.

6. The compound according to claim 1, or a pharmaceutically acceptable salt thereof for the treatment or prevention of idiopathic lung disease (IPF) or cough.

Description:
Novel Tetrazoles

TECHNICAL FIELD

This invention relates to tetrazoles and their use as inhibitors of TRPA1 activity, pharma ceutical compositions containing the same, and methods of using the same as agents for treatment and/or prevention of fibrotic diseases, inflammatory and auto-immune diseases and CNS-related diseases.

BACKGROUND INFORMATION

Transient receptor potential channels (TRP channels) are a group of voltage-gated ion channels located mostly on the plasma membrane of numerous mammalian cell types. There are approximately 30 structurally related TRP channels sorted into groups: TRP A, TRPC, TRPM, TRPML, TRPN, TRPP and TRPV. Transient receptor potential cation channel, subfamily A, member 1 (TRPAl), also known as transient receptor potential ankyrin 1, is the only member of the TRPA gene subfamily. Structurally, TRPA channels are characterized by multiple N-terminal ankyrin repeats (~14 in the N-terminus of human TRPAl) that gives rise to the “A” for ankyrin designation (Montell, 2005).

TRPAl is highly expressed in the plasma membrane of sensory neurons in the dorsal root and nodose ganglia that serve both skin and lung, as well as in small intestine, colon, pan creas, skeletal muscle, heart, brain, bladder and lymphocytes (https://www.proteinat- las.org/) as well as in human lung fibroblasts.

TRPAl is best known as a sensor for environmental irritants giving rise to somatosensory modalities such as pain, cold and itch. TRPAl is activated by a number of reactive, elec trophilic stimuli (e.g. allyl isothiocyanate, reactive oxygen species), as well as non-reactive compounds (e.g. icilin), implicated in cough associated with asthma, chronic pulmonary obstructive disease (COPD), idiopathic pulmonary fibrosis (IPF) or post-viral cough or for chronic idiopathic cough as well as cough in sensitive patients. (Song and Chang, 2015; Grace and Belvisi, 2011). TRPAl inhibitors are useful in the treatment of IPF in which cough is highly prevalent because of the link between cough and lung injury, based on studies showing cough-induced elevation of TGF-b (Xie et al., 2009; Froese et al., 2016; Tschumperlin et al., 2003; Yamamoto et al., 2002; Ahamed et al., 2008). TRPA1 antago nists inhibit calcium signaling triggered by cough triggers such as cigarette smoke extract (CSE) oxidative stress, inflammatory mediator release and downregulated antioxidant gene expression (Lin et al., 2015; Wang et al., 2019). TRPA1 antagonists are effective in studies of atopic dermatitis (Oh et al., 2013; Wilson et al., 2013), contact dermatitis (Liu et al.,

2013), psoriasis-associated itch (Wilson et al., 2013) and IL-31 -dependent itch (Cevikbas et al., 2014). A human TRPA1 gain-of-function has been associated with familial episodic pain syndrome (Kremeyer et al., 2010). A TRPA1 antagonist was effective in a behavioral model of migraine-related allodynia (Edelmayer et al., 2012). TRPA1 is selectively in- creased in trigeminal ganglia innervating injured teeth when compared to TRPA1 expres sion in trigeminal ganglia innervating healthy teeth (Haas et al., 2011). Several anaesthet ics are known to be TRPA1 agonists, including isoflurane (Matta et al., 2008) providing rationale for TRPA1 inhibitors for the relief of post-surgical pain. TRPA1 knockout mice and wild type mice treated with a TRPA1 antagonist showed anxiolytic- and antidepres- sant-like phenotypes (de Moura et al., 2014). TRPA1 inhibitors are expected to have bene fit in the treatment of diabetic neuropathy based on studies showing a mechanistic link of inverse regulation between AMPK and TRPA1 (Hiyama et al., 2018; Koivisto and Pertovaara, 2013; Wang et al., 2018). TRPA1 knockout mice exhibit smaller myocardial infarct sizes compared to wild type mice (Conklin et al., 2019). TRPA1 knockout and pharmacological intervention inhibited TNBS-induced colitis in mice (Engel et al., 2011). In a mouse brain ischaemia model, TRPA1 knock-out and TRPA1 antagonists reduce mye lin damage (Hamilton et al., 2016). Urate crystals and joint inflammation are reduced in TRPA1 knockout mice in a monosodium urate mouse model of gout (Moilanen et al., 2015). TRPA1 deletion in rats ameliorated joint inflammation and hyperalgesia in a rat model of acute gout flares (Trevisan et al., 2014). Activation of TRPA1 elicits an inflam matory response in osteoarthritic chondrocytes (Nummenmaa et al., 2016). TRPA1 inhibi tion and genetic deletion reduces inflammatory mediators in osteoarthritic mouse chondro cytes and murine cartilage (Nummenmaa et al., 2016). Finally, TRPA1 knockout mice ex hibited improvements in weight bearing on the osteoarthritic limb in an MIA-evoked knee swelling model (Horvath et al., 2016). TRPA1 is differentially expressed in the in the blad der epithelium rats (Du et al., 2007) and of patients with bladder outlet obstruction (Du et al., 2008). TRPA1 receptor modulation attenuates bladder overactivity in a rat model of spinal cord injury (Andrade et al., 2011) and intrathecal administration of TRPAl antago nists attenuate cyclophosphamide-induced cystitis in rats with hyper-reflexia micturition (Chen et al., 2016). It is therefore desirable to provide potent TRPAl inhibitors.

TRPAl inhibitors of various structural classes are reviewed in S. Skerratt, Progress in Me dicinal Chemistry, 2017, Volume 56, 81-115 and in D. Preti, G. Saponaro, A. Szallasi, Pharm. Pat. Anal. (2015) 4 (2), 75-94.

W02017/060488 discloses compounds that are antagonists of TRPAl, having the general ized structural formula

The TRPAl activity of Examples 28 and 29 bearing a tetrazolyl ring therein is not dis- closed.

L. Schenkel, etal ., J. Med. Chem. 2016, 59, 2794-2809 discloses quinazolinone-based TRPAl antagonists including compounds of the generalized structural formula of which compound 31, wherein R is OH, is disclosed as having an antagonistic TRPA1 activity of IC50 58 nM in a FLIPR assay and having an intrinsic clearance in human liver microsomes of <14 μL/min/kg.

DETAILED DESCRIPTION OF THE INVENTION The present invention provides novel thienopyrimidinones that are surprisingly potent in hibitors of TRPA1 (Assay A), further characterised by improved stability in human liver microsomes (Assay B) improved stability in human hepatocytes (Assay C).

Compounds of the present invention differ structurally from examples 28 and 29 in WO20 17/060488 in that they contain a hydroxyl substituent attached to the benzylic car bon adjacent to the para-chl orophenyl group; with some compounds additionally having differing bicyclic moieties attached via the methylene linker to the tetrazolyl ring. Com pounds of the present invention additionally differ structurally from example 31 in L. Schenkel, et al ., J. Med. Chem. 2016, 59, 2794-2809, in that they bear a tetrazolyl ring. These structural differences unexpectedly lead to a favourable combination of (i) inhibition of TRPA1, (ii) stability in human liver microsomes, (iii) stability in human hepatocytes, and (iv) fewer unwanted human metabolites.

Compounds of the invention are thus superior to those disclosed in the prior art in terms of the combination of the following parameters:

-potency as inhibitors of TRPA1

-stability in human liver microsomes

-stability in human hepatocytes

-giving rise to fewer unwanted human metabolites

Stability in human liver microsomes refers to the susceptibility of compounds to biotrans formation in the context of selecting and/or designing drugs with favorable pharmacoki netic properties as a first screening step. The primary site of metabolism for many drugs is the liver. Human liver microsomes contain the cytochrome P450s (CYPs), and thus repre sent a model system for studying phase I drug metabolism in vitro. Enhanced stability in human liver microsomes is associated with several advantages, including increased bioa vailability and adequate half-life, which can enable lower and less frequent dosing of pa tients. Thus, enhanced stability in human liver microsomes is a favorable characteristic for compounds that are to be used for drugs. Therefore, compounds of the present invention in addition to being able to inhibit TRPA1 are expected to have a favorable in vivo clearance and thus the desired duration of action in humans. Stability in human hepatocytes refers to the susceptibility of compounds to biotransfor mation in the context of selecting and/or designing drugs with favorable pharmacokinetic properties. The primary site of metabolism for many drugs is the liver. Human hepatocytes contain the cytochrome P450s (CYPs) and other drug metabolizing enzymes, and thus rep- resent a model system for studying drug metabolism in vitro. (Importantly, in contrast to liver microsomes assay, the hepatocytes assay covers also phase II biotransformations as well as liver-specific transporter-mediated processes, and therefore represents a more com plete system for drug metabolism studies). Enhanced stability in human hepatocytes is as sociated with several advantages, including increased bioavailability and adequate half- life, which can enable lower and less frequent dosing of patients. Thus, enhanced stability in human hepatocytes is a favorable characteristic for compounds that are to be used for drugs.

USED TERMS AND DEFINITIONS

Terms not specifically defined herein should be given the meanings that would be given to them by one of skill in the art in light of the disclosure and the context. As used in the specification, however, unless specified to the contrary, the following terms have the meaning indicated and the following conventions are adhered to.

The term tetrazolyl refers to the radical of the following ring

Unless specifically indicated, throughout the specification and the appended claims, a given chemical formula or name shall encompass tautomers and all stereo, optical and geo- metrical isomers (e.g. enantiomers, diastereomers, E/Z isomers etc.) and racemates thereof as well as mixtures in different proportions of the separate enantiomers, mixtures of dia stereomers, or mixtures of any of the foregoing forms where such isomers and enantiomers exist, as well as salts, including pharmaceutically acceptable salts thereof and solvates thereof such as for instance hydrates including solvates of the free compounds or solvates of a salt of the compound. In general, substantially pure stereoisomers can be obtained according to synthetic princi ples known to a person skilled in the field, e.g. by separation of corresponding mixtures, by using stereochemically pure starting materials and/or by stereoselective synthesis. It is known in the art how to prepare optically active forms, such as by resolution of racemic forms or by synthesis, e.g. starting from optically active starting materials and/or by using chiral reagents.

Enantiomerically pure compounds of this invention or intermediates may be prepared via asymmetric synthesis, for example by preparation and subsequent separation of appropriate diastereomeric compounds or intermediates which can be separated by known methods (e.g. by chromatographic separation or crystallization) and/or by using chiral reagents, such as chiral starting materials, chiral catalysts or chiral auxiliaries.

Further, it is known to the person skilled in the art how to prepare enantiomerically pure compounds from the corresponding racemic mixtures, such as by chromatographic separa tion of the corresponding racemic mixtures on chiral stationary phases; or by resolution of a racemic mixture using an appropriate resolving agent, e.g. by means of diastereomeric salt formation of the racemic compound with optically active acids or bases, subsequent resolution of the salts and release of the desired compound from the salt; or by derivatiza- tion of the corresponding racemic compounds with optically active chiral auxiliary rea gents, subsequent diastereomer separation and removal of the chiral auxiliary group; or by kinetic resolution of a racemate (e.g. by enzymatic resolution); by enantioselective crystal lization from a conglomerate of enantiomorphous crystals under suitable conditions; or by (fractional) crystallization from a suitable solvent in the presence of an optically active chi ral auxiliary.

The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use without excessive toxicity, irritation, allergic response, or other problem or complication, and commensurate with a reasonable benefit/risk ratio.

As used herein, "pharmaceutically acceptable salt" refers to derivatives of the disclosed compounds wherein the parent compound forms a salt or a complex with an acid or a base. Examples of acids forming a pharmaceutically acceptable salt with a parent compound containing a basic moiety include mineral or organic acids such as benzenesulfonic acid, benzoic acid, citric acid, ethanesulfonic acid, fumaric acid, gentisic acid, hydrobromic acid, hydrochloric acid, maleic acid, malic acid, malonic acid, mandelic acid, methanesul- fonic acid, 4-methyl-benzenesulfonic acid, phosphoric acid, salicylic acid, succinic acid, sulfuric acid and tartaric acid.

Examples for cations and bases forming a pharmaceutically acceptable salt with a parent compound containing an acidic moiety include Na + , K + , Ca 2+ , Mg 2+ , NH 4 + , L-arginine, 2,2’-iminobisethanol, L-lysine, N-methyl-D-glucamine or tris(hydroxymethyl)-amino- methane. The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound that contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a sufficient amount of the appropriate base or acid in water or in an organic diluent like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile, or a mixture thereof.

Salts of other acids than those mentioned above which for example are useful for purifying or isolating the compounds of the present invention (e.g. trifluoroacetate salts,) also com prise a part of the present invention.

BIOLOGICAL ASSAYS

Assay A: TRPA1 assay

The activity of the compounds of the invention may be demonstrated using the following in vitro TRPA1 cell assay:

Method:

A human HEK293 cell line over-expressing the human TRPA1 ion channel (Perkin Elmer, Product No. AX-004-PCL) was used as a test system for compound efficacy and potency. Compound activity was determined by measuring the effect of compounds on intracellular calcium concentration induced by AITC (Allylisothiocyanat) agonism in a FLIPRtetra sys tem (Molecular Devices).

Cell culture:

The cells were obtained as frozen cells in cryo-vials and stored until use at -150°C.

Cells were grown in culture medium (MEM/EBSS medium with 10% FCS and 0.4mg/ML Geneticin). It is important that density does not exceed 90% confluence. For sub-culturing cells were detached from flasks by Versene. At the day before the assay, cells were de tached, washed twice with medium (MEM/EBSS medium with 10% FCS) and 20000 cells in 20 μl/well were seeded to Poly D-Lysin biocoated 384-well plates (black, clear bottom, Cat.356697) from Corning. Plates were incubated for 24 hours at 37°C/5% CO2 before use in the assay. Compound preparation

The test compounds were dissolved in 100 % DMSO at a concentration of 10 mM and in a first step diluted in DMSO to a concentration of 5 mM, followed by serial dilution steps in 100% DMSO. Dilution factor and number of dilution steps may vary according to needs. Typically 8 different concentrations by 1:5 dilutions were prepared, further intermediate dilutions (1 :20) of the substances were carried out with HBSS/HEPES buffer

(lxHEPES, Cat.14065 from Gibco, 20mM HEPES, Cat. 83264 from SIGMA, 0.1% BSA Cat.11926 from Invitrogen, pH 7,4

FLIPR assay: At the assay day cells were washed 3x with assay puffer, 20 μL buffer remained in the wells after washing. 10 μL Ca6 kit (Cat.R8191 MolecularDevices) loading buffer in HBSS/HEPES was added to the cells and the plates were incubated with lid for 120 minutes at 37°/5% C02. 10 μL of compound or controls in HBSS/HEPES buffer/5% DMSO from the intermediate dilution plate were carefully added to the wells. Lumines- cence (indicating the calcium influx or release) was read on the FLIPRtetra device for 10 minutes to monitor the compound induced effects (e.g. agonism). Finally 10 μL of the ago nist AITC 50mM dissolved in HBSS/HEPES buffer/0,05% DMSO (final concentration 10 pM) was added to the wells followed by an additional read on the FLIPRtetra device for 10 minutes. The area under the signal curve (AUC) after AITC addition was used for IC50/%inhibition calculations

Data evaluation and calculation:

Each assay microtiter plate contained wells with vehicle (1% DMSO) controls instead of compound as controls for AITC induced luminescence (100 %CTL; high controls) and wells with vehicle controls without AITC as controls for non-specific changes in lumines cence (0 %CTL; low controls).

The analysis of the data was performed by the calculation of the area under signal curve of the individual wells. Based on this values the % value for the measurement of each sub stance concentration was calculated (AUC(sample) - AUC(low))*100/(AUC(high) - AUC(low))using MegaLab software (in house development). The IC50 values were calcu lated from the % control values using MegaLab software. Calculation: a = low value, d = high value; x = cone M; c=IC50 M; b = hill; y = % Ctrl

Table 1 : Biological data for compounds of the invention as obtained in Assay A

Table 2: Biological data for prior art compounds (examples 28 and 29 in W02017/060488) as obtained in Assay A.

Table 3: Biological data for prior art compounds (example 31 in L. Schenkel, et al. , J. Med. Chem. 2016, 59, 2794-2809) as obtained in Assay A. Assay B: Microsomal clearance: The metabolic degradation of the test compound is assayed at 37°C with pooled liver mi- crosomes. The final incubation volume of 100 μl per time point contains TRIS buffer pH 7.6 at RT (0.1 M), magnesium chloride (5 mM), microsomal protein (1 mg/ml) and the test compound at a final concentration of 1 mM. Following a short preincubation period at 37°C, the reactions are initiated by addition of beta-nicotinamide adenine dinucleotide phosphate, reduced form (NADPH, 1 mM) and ter minated by transferring an aliquot into solvent after different time points (0, 5, 15, 30, 60 min). Additionally, the NADPH-independent degradation is monitored in incubations with out NADPH, terminated at the last time point. The [%] remaining test compound after NADPH independent incubation is reflected by the parameter c(control) (metabolic stabil ity). The quenched incubations are pelleted by centrifugation (10000 g, 5 min).

An aliquot of the supernatant is assayed by LC-MS/MS for the amount of parent com pound. The half-life (tl/2 INVITRO) is determined by the slope of the semilogarithmic plot of the concentration-time profile.

The intrinsic clearance (CL INTRINSIC) is calculated by considering the amount of pro tein in the incubation:

CL INTRINSIC [μl/min/mg protein] = (Ln 2 / (half-life [min] * protein content [mg/ml])) * 1000

CL INTRIN SIC INVI V O [ml/min/kg] = (CL INTRINSIC [μL/min/mg protein] x MPPGL [mg protein/g liver] x liver factor [g/kg body weight]) / 1000

Qh [%] = CL [ml/min/kg] / hepatic blood flow [ml/min/kg])

Hepatocellularity, human: 120xl0e6 cells / g liver Liver factor, human: 25.7 g / kg bodyweight Blood flow, human: 21 ml/(min x kg) Table 4: Biological data for compounds of the invention as obtained in Assay B

Table 5: Biological data for prior art compounds (examples 28 and 29 in W02017/060488) as obtained in Assay B.

Table 6: Biological data for prior art compounds (example 31 in L. Schenkel, et al. , J. Med. Chem. 2016, 59, 2794-2809) as obtained in Assay B.

Assay C: Hepatocyte clearance The metabolic degradation of the test compound is assayed in a hepatocyte suspension.

Hepatocytes (cryopreserved) are incubated in. Dulbecco's modified eagle medium (supple mented with 3.5μg glucagon/5 OOmL, 2.5mg insulin/500mL and 3.75mg/500mL hydrocor- tison) containing 5% or 50% species serum.

Following a 30 min preincubation in an incubator (37°C, 10% CO2) 5 μl of test compound solution (80 mM; from 2mM in DMSO stock solution diluted 1 :25 with medium) are added into 395 μl hepatocyte suspension (cell density in the range 0.25-5 Mio cells/mL depend ing on the species, typically 1 Mio cells/mL; final concentration of test compound ImM, final DMSO concentration 0.05%). The cells are incubated for six hours (incubator, orbital shaker) and samples (25 μl) are taken at 0, 0.5, 1, 2, 4 and 6 hours. Samples are transferred into acetonitrile and pelleted by centrifugation (5 min). The supernatant is transferred to a new 96-deepwell plate, evapo rated under nitrogen and resuspended. Decline of parent compound is analyzed by HPLC-MS/MS

CLint is calculated as follows CL_INTRINSIC = Dose / AUC = (CO/CD) / (AUD + clast/k) x 1000/60. CO: initial concentration in the incubation [mM], CD: cell density of vi tal cells [10e6cells/mL], AUD: area under the data [mM x h], clast: concentration of last data point [pM], k: slope of the regression line for parent decline [h-1] The calculated in vitro hepatic intrinsic clearance can be scaled up to the intrinsic in vivo hepatic Clearance and used to predict hepatic in vivo blood clearance (CL) by the use of a liver model (well stirred model).

CL INTRIN SIC INVI V O [ml/min/kg] = (CL_INTRINSIC [μL/min/10e6cells] x hepato- cellularity [10e6 cells/g liver] x liver factor [g/kg bodyweight]) / 1000

CL [ml/min/kg] = CL_INTRINSIC_INVI VO [ml/min/kg] x hepatic blood flow [ml/min/kg] / (CL_INTRINSIC _INVI VO [ml/min/kg] + hepatic blood flow [ml/min/kg]) Qh [%] = CL [ml/min/kg] / hepatic blood flow [ml/min/kg])

Hepatocellularity, human: 120xl0e6 cells / g liver Liver factor, human: 25.7 g / kg bodyweight Blood flow, human: 21 ml/(min x kg)

Table 7: Biological data for compounds of the invention as obtained in Assay C Table 8: Biological data for prior art compounds (examples 28 and 29 in W02017/060488) as obtained in Assay C. Table 9: Biological data for prior art compounds (example 31 in L. Schenkel, et al. , J. Med. Chem. 2016, 59, 2794-2809) as obtained in Assay C.

Assay D: Metabolism in human hepatocytes in vitro The metabolic pathway of a test compound is investigated using primary human hepatocytes in suspension. After recovery from cryopreservation, human hepatocytes are incubated in Dulbecco's modified eagle medium containing 5% human serum and supplemented with 3.5 μg glucagon/500ml, 2.5mg insulin/500ml and 3.75mg/500ml hydrocortisone.

Following a 30 min preincubation in a cell culture incubator (37°C, 10% CO2), test com- pound solution is spiked into the hepatocyte suspension to obtain a final cell density of 1.0* 10 6 to 4.0* 10 6 cells/ml (depending on the metabolic turnover rate of the compound ob served with primary human hepatocytes), a final test compound concentration of 10 mM, and a final DMSO concentration of 0.05%.

The cells are incubated for six hours in a cell culture incubator on a horizontal shaker, and samples are removed from the incubation after 0, 0.5, 1, 2, 4 or 6 hours, depending on the metabolic turnover rate. Samples are quenched with acetonitrile and pelleted by centrifuga tion. The supernatant is transferred to a 96-deepwell plate, evaporated under nitrogen and resuspended prior to bioanalysis by liquid chromatography-high resolution mass spectrom etry for identification of putative metabolites. Table 10: Major metabolite observed for Example 5 of the invention as obtained by Assay D:

The integrity of tetrazole ring was not affected in any of the major described metabolites, based on tentative LC-MS n structure elucidation.

METHOD OF TREATMENT

The present invention is directed to compounds of general formula 1 which are useful in the prevention and/or treatment of a disease and/or condition associated with or modulated by TRPA1 activity, including but not limited to the treatment and/or prevention of fibrotic disease, inflammatory and immunoregulatory disorders, respiratory or gastrointestinal dis- eases or complaints, ophthalmic diseases, inflammatory diseases of the joints and inflam matory diseases of the nasopharynx, eyes, and skin. Said disorders, diseases and com plaints include cough, idiopathic pulmonary fibrosis, other pulmonary interstitial diseases and other fibrotic, asthma or allergic diseases, eosinophilic diseases, chronic obstructive pulmonary disease, as well as autoimmune pathologies, such as rheumatoid arthritis and atherosclerosis, pain and neurological disorders, such as depression.

The compounds of general formula 1 are useful for the prevention and/or treatment of:

(1) Cough such as chronic idiopathic cough or chronic refractory cough, cough associated with asthma, COPD and lung cancer and post-viral cough.

(2) Pulmonary fibrotic diseases such as pneumonitis or interstitial pneumonitis associated with collagenosis, e.g. lupus erythematodes, systemic scleroderma, rheumatoid arthritis, polymyositis and dermatomysitis, idiopathic interstitial pneumonias, such as pulmonary lung fibrosis (IPF), non-specific interstitial pneumonia, respiratory bronchiolitis associated interstitial lung disease, desquamative interstitial pneumonia, cryptogenic orgainizing pneumonia, acute interstitial pneumonia and lymphocytic interstitial pneumonia, lymangi- oleiomyomatosis, pulmonary alveolar proteinosis, Langerhan’s cell histiocytosis, pleural parenchymal fibroelastosis, interstitial lung diseases of known cause, such as interstitial pneumonitis as a result of occupational exposures such as asbestosis, silicosis, miners lung (coal dust), farmers lung (hay and mould), Pidgeon fanciers lung (birds) or other occupa tional airbourne triggers such as metal dust or mycobacteria, or as a result of treatment such as radiation, methotrexate, amiodarone, nitrofurantoin or chemotherapeutics, or for granulomatous disease, such as granulomatosis with polyangitis, Churg-Strauss syndrome, sarcoidosis, hypersensitivity pneumonitis, or interstitial pneumonitis caused by different origins, e.g. aspiration, inhalation of toxic gases, vapors, bronchitis or pneumonitis or inter stitial pneumonitis caused by heart failure, X-rays, radiation, chemotherapy, M. boeck or sarcoidosis, granulomatosis, cystic fibrosis or mucoviscidosis, or alpha- 1 -antitrypsin defi ciency.

(3) Other fibrotic diseases such as hepatic bridging fibrosis, liver cirrhosis, non-alcoholic steatohepatitis (NASH), atrial fibrosis, endomyocardial fibrosis, old myocardial infarction, glial scar, arterial stiffness, arthrofibrosis, Dupuytren's contracture, keloid, sclero derma/systemic sclerosis, mediastinal fibrosis, myelofibrosis, Peyronie's disease, nephro genic systemic fibrosis, retroperitoneal fibrosis, adhesive capsulitis.

(4) Inflammatory, auto-immune or allergic diseases and conditions such as allergic or non- allergic rhinitis or sinusitis, chronic sinusitis or rhinitis, nasal polyposis, chronic rhinosi- nusitis, acute rhinosinusitis, asthma, pediatric asthma, allergic bronchitis, alveolitis, hyper reactive airways, allergic conjunctivitis, bronchiectasis, adult respiratory distress syn drome, bronchial and pulmonary edema, bronchitis or pneumonitis, eosinophilic cellulites (e.g., Well's syndrome), eosinophilic pneumonias (e.g., Loeffler's syndrome, chronic eosin ophilic pneumonia), eosinophilic fasciitis (e. g., Shulman's syndrome), delayed-type hyper sensitivity, non-allergic asthma; exercise induced bronchoconstriction; chronic obstructive pulmonary disease (COPD), acute bronchitis, chronic bronchitis, cough, pulmonary em physema; systemic anaphylaxis or hypersensitivity responses, drug allergies (e.g., to peni cillin, cephalosporin), eosinophiliamyalgia syndrome due to the ingestion of contaminated tryptophane, insect sting allergies; autoimmune diseases, such as rheumatoid arthritis, Graves' disease, Sjogren's syndrome psoriatic arthritis, multiple sclerosis, systemic lupus erythematosus, myasthenia gravis, immune thrombocytopenia (adult ITP, neonatal throm bocytopenia, pediatric ITP), immune hemolytic anemia (auto-immune and drug induced), Evans syndrome (platelet and red cell immune cytopaenias), Rh disease of the newborn, Goodpasture's syndrome (anti-GBM disease), Celiac, autoimmune cardio-myopathy juve nile onset diabetes; glomerulonephritis, autoimmune thyroiditis, Behcet's disease; graft re jection (e.g., in transplantation), including allograft rejection or graftversus-host disease; inflammatory bowel diseases, such as Crohn's disease and ulcerative colitis; spondyloar thropathies; scleroderma; psoriasis (including T-cell mediated psoriasis) and inflammatory dermatoses such as an dermatitis, eczema, atopic dermatitis, allergic contact dermatitis, ur ticaria; vasculitis (e. g., necrotizing, cutaneous, and hypersensitivity vasculitis); erythema nodosum; eosinophilic myositis, eosinophilic fasciitis, cancers with leukocyte infiltration of the skin or organs; ophthalmic diseases such as age related macular degeneration, dia betic retinopathy and diabetic macular edema, keratitis, eosinophilic keratitis, keratocon junctivitis, vernal keratoconjunctivitis, scarring, anterior segment scarring, blepharitis, ble pharoconjunctivitis, bullous disorders, cicatricial pemphigoid, conjunctival melanoma, pa- pillary conjunctivitis, dry eye, episcleritis, glaucoma, gliosis, Granuloma annulare, Graves' ophthalmopathy, intraocular melanoma, Pinguecula, proliferative vitreoretinopathy, pter ygia, scleritis, uveitis, acute gout flares, gout or osteoarthritis.

(5) Pain such as chronic idiopathic pain syndrome, neuropathic pain, dysesthesia, allo- dynia, migraine, dental pain and post-surgical pain (6) Depression, anxiousness, diabetic neuropathy and bladder disorders such as bladder outlet obstruction, overactive bladder, cystitis; myocardial reperfusion injury or brain is chaemia injury.

Accordingly, the present invention relates to a compound of general formula 1 for use as a medicament. Furthermore, the present invention relates to the use of a compound of general formula 1 for the treatment and/or prevention of a disease and/or condition associated with or modu lated by TRPA1 activity.

Furthermore, the present invention relates to the use of a compound of general formula 1 for the treatment and/or prevention of fibrotic disease, inflammatory and immunoregula- tory disorders, respiratory or gastrointestinal diseases or complaints, ophthalmic diseases, inflammatory diseases of the joints and inflammatory diseases of the nasopharynx, eyes, and skin. Said disorders, diseases and complaints include cough, idiopathic pulmonary fi brosis, other pulmonary interstitial diseases and other fibrotic, asthma or allergic diseases, eosinophilic diseases, chronic obstructive pulmonary disease, as well as autoimmune pa- thologies, such as rheumatoid arthritis and atherosclerosis.

Furthermore, the present invention relates to the use of a compound of general formula 1 for the treatment and/or prevention of: (1) Cough such as chronic idiopathic cough or chronic refractory cough, cough associated with asthma, COPD and lung cancer and post-viral cough.

(2) Pulmonary fibrotic diseases such as pneumonitis or interstitial pneumonitis associated with collagenosis, e.g. lupus erythematodes, systemic scleroderma, rheumatoid arthritis, polymyositis and dermatomysitis, idiopathic interstitial pneumonias, such as pulmonary lung fibrosis (IPF), non-specific interstitial pneumonia, respiratory bronchiolitis associated interstitial lung disease, desquamative interstitial pneumonia, cryptogenic orgainizing pneumonia, acute interstitial pneumonia and lymphocytic interstitial pneumonia, lymangi- oleiomyomatosis, pulmonary alveolar proteinosis, Langerhan’s cell histiocytosis, pleural parenchymal fibroelastosis, interstitial lung diseases of known cause, such as interstitial pneumonitis as a result of occupational exposures such as asbestosis, silicosis, miners lung (coal dust), farmers lung (hay and mould), Pidgeon fanciers lung (birds) or other occupa tional airbourne triggers such as metal dust or mycobacteria, or as a result of treatment such as radiation, methotrexate, amiodarone, nitrofurantoin or chemotherapeutics, or for granulomatous disease, such as granulomatosis with polyangitis, Churg-Strauss syndrome, sarcoidosis, hypersensitivity pneumonitis, or interstitial pneumonitis caused by different origins, e.g. aspiration, inhalation of toxic gases, vapors, bronchitis or pneumonitis or inter stitial pneumonitis caused by heart failure, X-rays, radiation, chemotherapy, M. boeck or sarcoidosis, granulomatosis, cystic fibrosis or mucoviscidosis, or alpha- 1 -antitrypsin defi ciency.

(3) Other fibrotic diseases such as heaptic bridging fibrosis, liver cirrhosis, non-alcoholic steatohepatitis (NASH), atrial fibrosis, endomyocardial fibrosis, old myocardial infarction, glial scar, arterial stiffness, arthrofibrosis, Dupuytren's contracture, keloid, sclero derma/systemic sclerosis, mediastinal fibrosis, myelofibrosis, Peyronie's disease, nephro genic systemic fibrosis, retroperitoneal fibrosis, adhesive capsulitis.

(4) Inflammatory, auto-immune or allergic diseases and conditions such as allergic or non- allergic rhinitis or sinusitis, chronic sinusitis or rhinitis, nasal polyposis, chronic rhinosi- nusitis, acute rhinosinusitis, asthma, pediatric asthma, allergic bronchitis, alveolitis, hyper reactive airways, allergic conjunctivitis, bronchiectasis, adult respiratory distress syn drome, bronchial and pulmonary edema, bronchitis or pneumonitis, eosinophilic cellulites (e.g., Well's syndrome), eosinophilic pneumonias (e.g., Loeffler's syndrome, chronic eosin ophilic pneumonia), eosinophilic fasciitis (e. g., Shulman's syndrome), delayed-type hyper sensitivity, non-allergic asthma; exercise induced bronchoconstriction; chronic obstructive pulmonary disease (COPD), acute bronchitis, chronic bronchitis, cough, pulmonary em physema; systemic anaphylaxis or hypersensitivity responses, drug allergies (e.g., to peni cillin, cephalosporin), eosinophiliamyalgia syndrome due to the ingestion of contaminated tryptophane, insect sting allergies; autoimmune diseases, such as rheumatoid arthritis, Graves' disease, Sjogren's syndrome psoriatic arthritis, multiple sclerosis, systemic lupus erythematosus, myasthenia gravis, immune thrombocytopenia (adult ITP, neonatal throm bocytopenia, pediatric ITP), immune hemolytic anemia (auto-immune and drug induced), Evans syndrome (platelet and red cell immune cytopaenias), Rh disease of the newborn, Goodpasture's syndrome (anti-GBM disease), Celiac, autoimmune cardio-myopathy juve nile onset diabetes; glomerulonephritis, autoimmune thyroiditis, Behcet's disease; graft re jection (e.g., in transplantation), including allograft rejection or graftversus-host disease; inflammatory bowel diseases, such as Crohn's disease and ulcerative colitis; spondyloar thropathies; scleroderma; psoriasis (including T-cell mediated psoriasis) and inflammatory dermatoses such as an dermatitis, eczema, atopic dermatitis, allergic contact dermatitis, ur ticaria; vasculitis (e. g., necrotizing, cutaneous, and hypersensitivity vasculitis); erythema nodosum; eosinophilic myositis, eosinophilic fasciitis, cancers with leukocyte infiltration of the skin or organs; ophthalmic diseases such as age related macular degeneration, dia betic retinopathy and diabetic macular edema, keratitis, eosinophilic keratitis, keratocon junctivitis, vernal keratoconjunctivitis, scarring, anterior segment scarring, blepharitis, ble pharoconjunctivitis, bullous disorders, cicatricial pemphigoid, conjunctival melanoma, pa pillary conjunctivitis, dry eye, episcleritis, glaucoma, gliosis, Granuloma annulare, Graves' ophthalmopathy, intraocular melanoma, Pinguecula, proliferative vitreoretinopathy, pter ygia, scleritis, uveitis, acute gout flares, gout or osteoarthritis.

(5) Pain such as chronic idiopathic pain syndrome, neuropathic pain, dysesthesia, allo- dynia, migraine, dental pain and post-surgical pain.

(6) Depression, anxiousness, diabetic neuropathy and bladder disorders such as bladder outlet obstruction, overactive bladder, cystitis; myocardial reperfusion injury or brain is chaemia injury.

In a further aspect the present invention relates to a compound of general formula 1 for use in the treatment and/or prevention of above mentioned diseases and conditions.

In a further aspect the present invention relates to the use of a compound of general for mula 1 for the preparation of a medicament for the treatment and/or prevention of above mentioned diseases and conditions. In a further aspect of the present invention the present invention relates to methods for the treatment or prevention of above mentioned diseases and conditions, which method com prises the administration of an effective amount of a compound of general formula 1 to a human being.

COMBINATION THERAPY

The compounds of the invention may further be combined with one or more, preferably one additional therapeutic agent. According to one embodiment the additional therapeutic agent is selected from the group of therapeutic agents useful in the treatment of diseases or conditions described hereinbefore, in particular associated with fibrotic diseases, inflam matory and immunoregulatory disorders, respiratory or gastrointestinal diseases or com plaints, inflammatory diseases of the joints or of the nasopharynx, eyes, and skin or condi tions such as for example cough, idiopathic pulmonary fibrosis, other pulmonary intersti- rial diseases, asthma or allergic diseases, eosinophilic diseases, chronic obstructive pulmo nary disease, atopic dermatitis as well as autoimmune pathologies, such as rheumatoid ar thritis and atherosclerosis, or therapeutic agents useful for the treatment of ophthalmic dis eases, pain and depression.

Additional therapeutic agents that are suitable for such combinations include in particular those, which, for example, potentiate the therapeutic effect of one or more active sub stances with respect to one of the indications mentioned and/or allow the dosage of one or more active substances to be reduced.

Therefore, a compound of the invention may be combined with one or more additional therapeutic agents selected from the group consisting of antifibrotic agents, anti-tussive agents, anti-inflammatory agents, anti-atopic dermatitis agents, analgesics, anti-convul- sants, anxiolytics, sedatives, skeletal muscle relaxants or anti-depressants.

Antifibrotic agents are for example nintedanib, pirfenidone, phosphodiesterase-IV (PDE4) inhibitors such as roflumilast, autotaxin inhibitors such as GLPG-1690 or BBT-877; con nective tissue growth factor (CTGF) blocking antibodies such as Pamrevlumab; B-cell ac- tivating factor receptor (BAFF-R) blocking antibodies such as Lanalumab; alpha-V/beta-6 blocking inhibitors such as BG-0001 l/STX-100, recombinant pentraxin-2 (PTX-2) such as PRM-151; c-Jun N-terminal kinase (INK) inhibitors such as CC-90001; galectin-3 inhibi tors such as TD-139; G-protein coupled receptor 84 (GPR84) inhibitors such as GLPG- 1205; G-protein coupled receptor 84/ G-protein coupled receptor 40 dual inhibitors such as PBI-4050; Rho Associated Coiled-Coil Containing Protein Kinase 2 (ROCK2) inhibitors such as KD-025; heat shock protein 47 (HSP47) small interfering RNA such as BMS- 986263/ND-L02-S0201; Wnt pathway inhibitor such as SM-04646; LD4 / PDE3/4 inhibi- tors such as Tipelukast; recombinant immuno-modulatory domains of histidyl tRNA syn thetase (HARS) such as ATYR-1923; prostaglandin synthase inhibitors such as ZL-2102 / SAR-191801; 15-hydroxy-eicosapentaenoic acid (15-HEPE e.g. DS-102); Lysyl Oxidase Like 2 (LOXL2) inhibitors such as PAT-1251, PXS-5382/PXS-5338; phosphoinositide 3- kinases (PI3K)/ mammalian target of rapamycin (mTOR) dual inhibitors such as HEC- 68498; calpain inhibitors such as BLD-2660; mitogen-activated protein kinase kinase ki nase (MAP3K19) inhibitors such as MG-S-2525; chitinase inhibitors such as OATD-01; mitogen-activated protein kinase-activated protein kinase 2 (MAPKAPK2) inhibitors such as MMI-0100; transforming growth factor beta 1 (TGF-betal) small interfering RNA such as TRK250/BNC-1021; or lysophosphatidic acid receptor antagonists such as BMS- 986278.

Anti-tussive agents are, for example, purinoceptor 3 (P2X3) receptor antagonists such as gefapixant, S-600918, BAY-1817080, or BLU-5937; neurokinin 1 (NK-1) receptor antago nist such as Orvepitant, Aprepitant; nicotinic acetylcholine receptor alpha 7 subunit stimu lator such as ATA-lOl/bradanicline; codeine, gabapentin, pregablin, or azithromycin. Anti-inflammatory agents are, for example, corticosteroids such as prednisolone or dexa- methasone; cyclo-oxygenase-2 (COX2) inhibitors such as celecoxib, rofecoxib, parecoxib, valdecoxib, deracoxib, etoricoxib or lumiracoxib; prostaglandin E2 antagonists; leukotri- ene B4 antagonists; leukotriene D4 antagonists such as monteleukast; 5 -lipoxygenase in hibitors; or other nonsteroidal anti-inflammatory agents (NSAIDs) such as aspirin, diclo- fenac, diflunisal, etodolac, ibuprofen or indomethacin.

Anti-atopic dermatitis agents are, for example, cyclosporin, methotrexate, mycophenolate mofetil, azathioprine, phosphodiesterase inhibitors (e.g. apremilast, crisaborole), Janus As sociated Kinase (JAK) inhibitors (e.g. tofacitinib), neutralizing antibodies against IL-4/IL- 13 (e.g. dupilamab), IL-13 (e.g. lebrikizumab, tralokinumab) and IL-31 (nemolizumab). Analgesics are, for example, of the opioid type, such as morphine, oxymorphine, levopa- nol, oxycodon, propoxyphene, nalmefene, fentanyl, hydrocondon, hydromorphone, meripidine, methadone, nalorphine, naloxone, naltrexone, buprenorphine, butorphanol, nal buphine, pentazocine; or of the non-opioid type, such as acetophenamine. Anti-depressants are, for example, tricyclic anti-depressants such as amitriptyline, clomi pramine, despramine, doxepin, desipramine, imipramine, nortriptyline; selective serotonin reuptake inhibitor anti-depressants (SSRIs) such as fluoxetine, paroxetine, sertraline, cital- opram, escitalopram; norepinephrine reuptake inhibitor anti-depressants (SNRIs) such as maprotiline, lofepramine, mirtazapine, oxaprotiline, fezolamine, tomoxetine, mianserin, buproprion, hydroxybuproprion, nomifensine, viloxazine; dual serotonin-norepinephrine reuptake inhibitor anti-depressants (SNRIs) such as duloxetine, venlafaxine, desvenlafax- ine, levomilnacipran; atypical antidepressants such as trazodone, mirtazapine, vortioxetine, vilazodone, bupropion; or monoamine oxidase inhibitor anti-depressantss (MAOIs) such as tranylcypromine, phenelzine, or isocarboxazid.

Anxiolytics are, for example, benzodiazepines such as alprazolam, bromazepam, chlordi- azepoxide, clonazepam, clorazepate, diazepam, flurazepam, lorazepam, oxazepam, temaze pam, triazolam, or tofisopam; or they are nonbenzodiazepine hypnoticssuch as eszopi- clone, zaleplon, zolpidem, or zopiclone; or they are carbamates e.g. meprobamate, cari- soprodol, tybamate, or lorbamate; or they are antihistamines such as hydroxyzine, chlor pheniramine or diphenhydramine.

Sedatives are, for example, barbiturate sedatives, such as amobarbital, aprobarbital, buta- barbital, butabital, mephobarbital, metharbital, methohexital, pentobarbital, secobarbital, talbutal, theamylal, or thiopental; or they are non-barbiturate sedatives such as glute- thimide, meprobamate, methaqualone or dichloalphenazone.

Skeletal muscle relaxants are, for example, baclofen, meprobamate, carisoprodol, cyclo- benzaprine, metaxalone, methocarbamol, tizanidine, chlorzoxazone or orphenadrine.

Other suitable combination partners are inhibitors of Acetylcholinesterase inhibitors such as donepezil; 5-HT-3 anatgonists such as ondansetron; metabotropic glutamate receptor an tagonists; antiarrhythmics such as mexiletine or phenytoin; or NMD A receptor antagonists. Further suitable combination partners are incontinence medications, for example, anticho linergics such as oxybutynin, tolterodine, darifenacin, fesoterodine, solifenacin or tro- spium; or they are bladder muscle relaxants such as mirabegron; or they are alpha blockers such as tamsulosin, alfuzosin, silodosin, doxazosin or terazosin.

The dosage for the combination partners mentioned above is usually 1/5 of the lowest dose normally recommended up to 1/1 of the normally recommended dose.

Therefore, in another aspect, this invention relates to the use of a compound according to the invention in combination with one or more additional therapeutic agents described hereinbefore and hereinafter for the treatment of diseases or conditions which may be af fected or which are mediated by TRPA1, in particular diseases or conditions as described hereinbefore and hereinafter.

In a further aspect this invention relates to a method for treating a disease or condition which can be influenced by the inhibition of TRPA1 in a patient that includes the step of administering to the patient in need of such treatment a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof in combination with a therapeutically effective amount of one or more additional therapeutic agents.

In a further aspect this invention relates to the use of a compound of formula (I) or a phar- maceutically acceptable salt thereof in combination with one or more additional therapeu tic agents for the treatment of diseases or conditions which can be influenced by the inhibi tion of TRPA1 in a patient in need thereof.

In yet another aspect the present invention relates a method for the treatment of a disease or condition mediated by TRPA1 activity in a patient that includes the step of administer- ing to the patient, preferably a human, in need of such treatment a therapeutically effective amount of a compound of the present invention in combination with a therapeutically ef fective amount of one or more additional therapeutic agents described in hereinbefore and hereinafter.

The use of the compound according to the invention in combination with the additional therapeutic agent may take place simultaneously or at staggered times.

The compound according to the invention and the one or more additional therapeutic agents may both be present together in one formulation, for example a tablet or capsule, or separately in two identical or different formulations, for example as a so-called kit-of-parts. Consequently, in another aspect, this invention relates to a pharmaceutical composition that comprises a compound according to the invention and one or more additional thera peutic agents described hereinbefore and hereinafter, optionally together with one or more inert carriers and/or diluents.

In yet another aspect the present invention relates to the use of a compound according to the invention in a cough-measuring device. Other features and advantages of the present invention will become apparent from the fol lowing more detailed examples which illustrate, by way of example, the principles of the invention. PREPARATION

The compounds according to the invention and their intermediates may be obtained using methods of synthesis which are known to the one skilled in the art and described in the lit erature of organic synthesis for example using methods described in “Comprehensive Or- ganic Transformations”, 2nd Edition, Richard C. Larock, John Wiley & Sons, 2010, and “March’s Advanced Organic Chemistry”, 7th Edition, Michael B. Smith, John Wiley & Sons, 2013. Preferably the compounds are obtained analogously to the methods of prepara tion explained more fully hereinafter, in particular as described in the experimental section. In some cases the sequence adopted in carrying out the reaction schemes may be varied Variants of these reactions that are known to the skilled artisan but are not described in de tail herein may also be used. The general processes for preparing the compounds according to the invention will become apparent to the skilled man on studying the schemes that fol low. Starting compounds are commercially available or may be prepared by methods that are described in the literature or herein, or may be prepared in an analogous or similar manner. Before the reaction is carried out, any corresponding functional groups in the starting compounds may be protected using conventional protecting groups. These protect ing groups may be cleaved again at a suitable stage within the reaction sequence using methods familiar to the skilled man and described in the literature for example in “Protect ing Groups”, 3rd Edition, Philip J. Kocienski, Thieme, 2005, and “Protective Groups in Organic Synthesis”, 4th Edition, Peter G. M. Wuts, Theodora W. Greene, John Wiley & Sons, 2006. The terms "ambient temperature" and "room temperature" are used inter changeably and designate a temperature of about 20 °C, e.g. between 19 and 24 °C.

Abbreviations:

Preparation of Starting Compounds Intermediate I

Intermediate 1.1 l-(4-bromophenyl)-2-[5-(chloromethyl)-2H-l,2,3,4-tetrazol-2- yl]ethan-l-one

To 1.00 g (8.44 mmol) of 5-(chloromethyl)-2H-l,2,3,4-tetrazole and 2.17 g (9.28 mmol) 4- chlorophenacyl bromide in 15 mL DMA are added 1.63 g (11.8 mmol) K2CO3. The reaction mixture is stirred at RT for 30 min and before the mixture is filtered. The solution is diluted with water and sat. aq. NaCl-solution and is extracted three times with EtOAc. The combined organic phase is washed with water, dried over Na 2 SO 4 , filtered over activated charcoal and the solvent is removed in vacuo. The residue is purified by column chromatography (silica gel; CH/EtOAc, C 10 H 8 CI 2 N 4 O (M = 271.1 g/mol)

ESI-MS: 271 [M+H] +

R t (HPLC): 1.01 min (method B) Intermediate II

Intermediate II.1

(lR)-l-(4-chlorophenyl)-2-[5-(chloromethyl)-2H-l,2,3,4-te trazol-2-yl]ethan-l-ol

1.30 g (4.80 mmol) l-(4-chlorophenyl)-2-[5-(chloromethyl)-2H-l,2,3,4-tetrazol-2 -yl]ethan- 1-one (intermediate 1.1) are added to 20 mL ACN under inert atmosphere. 11.9 mg (0.02 mmol) Chloro([(lS,2S)-(-)-2-amino-l,2-diphenylethyl](4-toluenesulf onyl)amido)(mesity- lene)ruthenium (II) (CAS 174813-81-1) are added before 0.72 mL (1.73 mmol) formic acid triethylamine complex (5:2) are added dropwise. After stirring at RT for 3 h the solvent is removed in vacuo. To the remaining crude mixture is added water and this mixture is ex- tracted with EtOAc. The organic layers are combined, dried over Na 2 S0 4 and activated char coal, filtered and the solvent is removed in vacuo.

C 10 H 10 CI 2 N 4 O (M = 273.1 g/mol) ESI-MS: 273 [M+H] +

R t (HPLC): 0.96 min (method B)

Intermediate III Intermediate III.1

Ethyl 4-amino-2-methanesulfonyl-l,3-thiazole-5-carboxylate

20.0 g (0.092 mol) Ethyl 4-amino-2-(methylsulfanyl)-l,3-thiazole-5-carboxylate (CAS: 39736-29-3) are added to 500 mL DCM while stirring. The mixture was cooled to 0°C and 35.0 g (0.142 mol) 3-chloroperoxybenzoic acid (70%) are added portionwise keeping the temperature of the reaction mixture below 24°C. The mixture is stirred at ambient tempera ture overnight and diluted with 300 mL DCM before 10.0 g (0.041 mol) 3-chloroperoxyben- zoic acid (70%) are added and the mixture is stirred for another 2.5 h at ambient temperature. Another 10 g (0.041 mmol) is added an the reaction mixture is stirred for 2 h. The mixture is then quenched by addition of saturated Na 2 SO 3 solution and stirred for 10 min. The or ganic phase is separated and washed with 2 M aqueous Na 2 CO 3 solution (2x) and brine. The organic extract is then dried over MgSO 4 , filtered and concentrated to yield the desired prod uct.

C7H10N2O4S2 (M = 250.3 g/mol)

ESI-MS: 251 [M+H] + R t (HPLC): 0.96 min (method E)

Intermediate III.2

Ethyl 4-amino- 1 ,3 -thiazole-5-carboxylate To 7.0 g (28.0 mmol) Ethyl 4-amino-2-methanesulfonyl-l,3-thiazole-5-carboxylate is added EtOH (100 mL) and the mixture is cooled to 0 °C. 3.14 g (83.9 mmol) sodium borohydride is added portionwise and the reaction mixture is stirred for 3 h at ambient temperature. The reaction mixture is concentrated and water and EtOAc is added to the residue. The organic phase is separated, washed with saturated aqueous NaHCO3 solution, dried over MgSO4, filtered and concentrated to give the desired product. C6H8N2O2S (M = 172.2 g/mol)

ESI-MS: 173 [M+H] +

R t (HPLC): 0.77 min (method B)

Intermediate III.3 6H,7H-[l,3]thiazolo[4,5-d]pyrimidin-7-one

To 250 mg (1.45 mmol) ethyl 4-amino-l,3-thiazole-5-carboxylate inEtOH (5.0 mL) is added 650 mg (6.2 mmol) formamidinium acetate. The reaction mixture is heated to 80 °C and stirred at this temperature for 16 h. After cooling to ambient temperature, 605 mg (5.8 mmol) formamidinium acetate is added and the reaction mixture is heated to 80 °C and stirred at this temperature for 5 h. It is further heated to 110 °C and stirred at this temperature for another 16 h. After cooling to ambient temperature, the mixture is purified by preparative HPLC (X-Bridge C18, ACN/H20 containing 0.1% TFA gradient) to yield the desired prod uct. C 5 H 3 N 3 OS (M = 153.2 g/mol)

ESI-MS: 154 [M+H] +

R t (HPLC): 0.12 min (method A) Intermediate IV

Intermediate IV.1 l-Methyl-lH,6H,7H-imidazo[4,5-d]pyridazin-7-one hydrobromide salt

To a mixture of 5.3 g (23.1 mmol) 2-bromo-l -methyl- lH,6H,7H-imidazo[4,5-d]pyridazin- 7-one in 75 mL MeOH is added 200 mg Pd/C (10%) in an autocleave reactor. The space above the reaction mixture is evacuated and charged with 50 psi of hydrogen pressure. The reaction mixture is stirred at ambient temperature under 50 psi of hydrogen pressure over night. After removing the hydrogen and backfilling with nitrogen, the reaction mixture is diluted with 300 mL warm water and filtered. The filtrate is concentrated to dryness to yield the desired product.

C 6 H 6 N 4 O (M = 231.1 g/mol) ESI-MS: 11 [M-HBr+H] +

R t (HPLC): 0.12 min (method C)

Intermediate V

Intermediate V.1

2-(7-methyl-6-oxo-6,7-dihydro-lH-purin-l-yl)acetonitrile

To a solution of 0.750 g (4.99 mmol) of 7-methyl-6,7-dihydro-lH-purin-6-one in 15 mL DMF is added 2.07 g (15.0 mmol) K2CO3 and the mixture is stirred for 30 min at ambient temperature. 0.418 mL (5.99 mmol) Bromoacetonitrile is then added and the reaction mix ture is stirred at ambient temperature overnight. The mixture is filtered and the obtained filtrate is concentrated. The precipitate is suspended in di -isopropyl ether and the remaining solid is collected by filtration. It is further washed with cold di -isopropyl ether and dried at 50 °C under vacuum to yield the desired product. C8H7N5O (M = 189.7 g/mol)

ESI-MS: 190 [M+H] +

R t (HPLC): 0.18 min (method B) Intermediate V.2

7-Methyl- 1-[(2H-1, 2, 3, 4-tetrazol-5-yl)methyl]-6,7-dihydro-lH-purin-6-one To a mixture of 1.27 g (6.71 mmol) of 2-(7-methyl-6-oxo-6,7-dihydro-lH-purin-l-yl)ace- tonitrile in 15 mL of DMF is added 0.480 g (7.39 mmol) of sodium azide and 0.395 g (7.39 mmol) of ammonium chloride. The reaction mixture is heated to 95 °C and stirred at this temperature overnight. After cooling to ambient temperature, the precipitate is collected by filtration and washed with MeOH. The obtained solid is dried under vacuum at 50 °C to yield the desired product.

C8H8N8O (M = 232.2 g/mol)

ESI-MS: 233 [M+H] +

R t (HPLC): 0.17 min (method B)

Intermediate V.3 l-({2-[2-(4-Chlorophenyl)-2-oxoethyl]-2H-l,2,3,4-tetrazol-5- yl}methyl)-7-methyl-6,7- dihydro- 1 H-purin-6-one To a solution of 111 mg (0.474 mmol) of 4-chlorophenacyl bromide in 5 mL ACN is added 100 mg (0.431 mmol) of 7-methyl-l-[(2H-l,2,3,4-tetrazol-5-yl)methyl]-6,7-dihydro-lH -pu- rin-6-one and 83.3 mg (0.603 mmol) K2CO3. The reaction mixture is stirred at 70 °C over night. After cooling, 2.5 mL of DMA is added and the mixture is allowed to stir for another 14 h at 70 °C. After cooling to ambient temperature, the reaction mixture is filtered, concen trated and purified twice by preparative HPLC (ACN/H20/TFA gradient) to yield the de sired product.

C16H13CIN8O2 (M = 384.8 g/mol)

ESI-MS: 385 [M+H] +

R t (HPLC): 0.82 min (method B)

Preparation of Final Compounds

Example 1 (general route) 3-({2-[(2R)-2-(4-chlorophenyl)-2-hydroxyethyl]-2H-l,2,3,4-te trazol-5-yl}methyl)-5-me- thyl-4-oxo-3H,4H-thieno[2,3-d]pyrimidine-6-carboxamide

50.0 mg (0.331 mmol) of l-methyl-lH,6H,7H-[l,2,3]triazolo[4,5-d]pyrimidin-7-one are added to 1.0 mL DMF. Then, 137 mg (0.993 mmol) K2CO3 and 108.4 g (0.397 mmol) of intermediate III are added and the mixture is stirred at 50 °C for 4 h. After cooling to RT, the mixture is diluted with acetonitrile and methanol, filtered and purified by preparative HPLC (ACN/H2O/TFA) to yield the desired product.

C15H14CIN9O2 (M = 387.8 g/mol)

ESI-MS: 388 [M+H] + R t (HPLC): 0.43 min (method B)

1 HNMR (400 MHz, DMSO-d 6 ) d ppm 4.36 (s, 3H), 4.73 - 4.83 (m, 2H), 5.12 (dd, J=7A, 5.4 Hz, 1H), 5.55 (s, 2H), 5.61 - 6.38 (br s, 1H), 7.31 - 7.43 (m, 4H), 8.61 (s, 1H).

The following compounds are prepared according to the general procedure (example 1) de- scribed above:

Analytical data for the compounds described in the table above:

Example 9 l-({2-[(2R)-2-(4-chlorophenyl)-2-hydroxyethyl]-2H-l,2,3,4-te trazol-5-yl}methyl)-7- methyl-6, 7-dihydro-lH-purin-6-one

To 30 mg (0.078 mmol) of intermediate V.3 in MeCN (1.0 mL) is added under inert atmosphere 0.194 mg (0.32 μmol) Chloro([(lS,2S)-(-)-2-amino-l,2-diphenylethyl](4- toluenesulfonyl)amido)(mesitylene)ruthenium (II) (CAS 174813-81-1) followed by dropwise addition of 12 μL (0.028 mmol) formic acid trimethylamine complex (5:2). The reaction mixture is stirred at ambient temperature for 3h. The mixture is concentrated and the residue diluted with water. The mixture is extracted with EtOAc (2x). The combined organic extracts are dried over MgS04, filtered and purified by preparative HPLC. C16H15ClN8O2 (M = 386.8 g/mol)

ESI-MS: 387 [M+H] + R t (HPLC): 0.55 min (method G)

1 H NMR (400 MHz, DMSO-d 6 ) d ppm 3.96 (s, 3H), 4.75 - 4.82 (m, 2H), 5.12 (dd, J=7.4, 5.4 Hz, 1H), 5.52 (s, 2H), 7.34 - 7.42 (m, 4H), 8.25 (d, J=0.4 Hz, 1H), 8.47 (s, 1H). Analytical HPLC methods Method A

Analytical column: XBridge BEH C18 2.1 x 30 mm, 1.7 μm; column temperature: 60°C

Method B

Analytical column: Stable Bond (Agilent) 1.8 μm; 3.0 x 30 mm; column temperature:

60°C

Method C

Analytical column: Sunfire (Waters) 2.5 μm; 3.0 x 30 mm; column temperature: 60°C

Method D

Analytical column: XBridge C18 3.0 x 30 mm_2.5 μm (Waters); column temperature:

60 °C

Method E

Analytical column: XBridge C18 3.0 x 30 mm_2.5 μm ;column temperature: 60 °C

Method F

Analytical column: XSelect HSS PFP 2.1 x 30 mm, 1.8 μm; column temperature:

60 °C

Method G

Analytical column: Sunfire C18, 3.0 x 30 mm, 2.5 μm; column temperature: 60 °C




 
Previous Patent: THIENOPYRIMIDONES AS TRPA1 INHIBITORS

Next Patent: TOOTHED PLATE