US20210107919A1 - Novel Thienopyrimidones - Google Patents

Novel Thienopyrimidones Download PDF

Info

Publication number
US20210107919A1
US20210107919A1 US17/071,092 US202017071092A US2021107919A1 US 20210107919 A1 US20210107919 A1 US 20210107919A1 US 202017071092 A US202017071092 A US 202017071092A US 2021107919 A1 US2021107919 A1 US 2021107919A1
Authority
US
United States
Prior art keywords
compound
alkyl
group
pharmaceutically acceptable
formula
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US17/071,092
Other languages
English (en)
Inventor
Martin Thomas FLECK
Florian Paul Christian Binder
Georg Dahmann
Joerg P. HEHN
Annekatrin Charlotte HEIMANN
Uta Friederike LESSEL
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Boehringer Ingelheim International GmbH
Original Assignee
Boehringer Ingelheim International GmbH
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 Boehringer Ingelheim International GmbH filed Critical Boehringer Ingelheim International GmbH
Publication of US20210107919A1 publication Critical patent/US20210107919A1/en
Priority to US17/463,942 priority Critical patent/US11578080B2/en
Assigned to BOEHRINGER INGELHEIM INTERNATIONAL GMBH reassignment BOEHRINGER INGELHEIM INTERNATIONAL GMBH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BOEHRINGER INGELHEIM PHARMA GMBH & CO. KG
Assigned to BOEHRINGER INGELHEIM PHARMA GMBH & CO. KG reassignment BOEHRINGER INGELHEIM PHARMA GMBH & CO. KG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HEHN, JOERG P., BINDER, FLORIAN PAUL CHRISTIAN, FLECK, MARTIN THOMAS, Willwacher, Jens, LESSEL, UTA FRIEDERIKE, DAHMANN, GEORG, HEIMANN, Annekatrin Charlotte
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/14Antitussive agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/12Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D495/14Ortho-condensed systems

Definitions

  • This invention relates to thienopyrimidinones 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.
  • Transient receptor potential 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: TRPA, TRPC, TRPM, TRPML, TRPN, TRPP and TRPV.
  • Transient receptor potential cation channel, subfamily A, member 1 (TRPA1) also known as transient receptor potential ankyrin 1
  • TRPA channels are characterized by multiple N-terminal ankyrin repeats ( ⁇ 14 in the N-terminus of human TRPA1) that gives rise to the “A” for ankyrin designation (Montell, 2005).
  • TRPA1 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, pancreas, skeletal muscle, heart, brain, bladder and lymphocytes (https://www.proteinatlas.org/) as well as in human lung fibroblasts.
  • TRPA1 is best known as a sensor for environmental irritants giving rise to somatosensory modalities such as pain, cold and itch.
  • TRPA1 is activated by a number of reactive, electrophilic 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.
  • COPD chronic pulmonary obstructive disease
  • IPF idiopathic pulmonary fibrosis
  • post-viral cough or for chronic idiopathic cough as well as cough in sensitive patients.
  • TRPA1 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- ⁇ (Xie et al., 2009; Froese et al., 2016; Tschumperlin et al., 2003; Yamamoto et al., 2002; Ahamed et al., 2008).
  • TRPA1 antagonists 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).
  • CSE cigarette smoke extract
  • 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 increased in trigeminal ganglia innervating injured teeth when compared to TRPA1 expression in trigeminal ganglia innervating healthy teeth (Haas et al., 2011).
  • 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 antidepressant-like phenotypes (de Moura et al., 2014).
  • TRPA1 inhibitors are expected to have benefit 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).
  • TRPA1 knock-out and TRPA1 antagonists reduce myelin damage (Hamilton et al., 2016).
  • TRPA1 knockout mice 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 inflammatory response in osteoarthritic chondrocytes (Nummenmaa et al., 2016).
  • TRPA1 inhibition and genetic deletion reduces inflammatory mediators in osteoarthritic mouse chondrocytes and murine cartilage (Nummenmaa et al., 2016).
  • TRPA1 knockout mice exhibited 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 bladder 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 TRPA1 antagonists attenuate cyclophosphamide-induced cystitis in rats with hyper-reflexia micturition (Chen et al., 2016).
  • TRPA1 inhibitors of various structural classes are reviewed in S. Skerratt, Progress in Medicinal Chemistry, 2017, Volume 56, 81-115 and in D. Preti, G. Saponaro, A. Szallasi, Pharm. Pat. Anal. (2015) 4 (2), 75-94.
  • WO2017/060488 discloses compounds that are antagonists of TRPA1, having the generalized structural formula
  • the present invention provides novel thienopyrimidinones that are surprisingly potent inhibitors of TRPA1 (Assay A), further characterised by
  • Compounds of the present invention differ structurally from examples 28 and 29 in WO2017/060488 in that they contain a thienopyrimidinone core with amido substituents as well as substituents adjacent to a secondary aliphatic alcohol.
  • Compounds 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.
  • Stability in human liver microsomes refers to the susceptibility of compounds to biotransformation in the context of selecting and/or designing drugs with favorable pharmacokinetic 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 represent a model system for studying phase I drug metabolism in vitro
  • CYPs cytochrome P450s
  • Enhanced stability in human liver microsomes is associated with several advantages, including increased bioavailability and adequate half-life, which can enable lower and less frequent dosing of patients.
  • 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 biotransformation 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 reps resent a model system for studying drug metabolism in vitro.
  • CYPs cytochrome P450s
  • hepatocytes assay covers also phase II biotransformations as well as liver-specific transporter-mediated processes, and therefore represents a more complete system for drug metabolism studies.
  • Enhanced stability in human hepatocytes is associated with several advantages, including increased bioavailability and adequate half-life, which can enable lower and less frequent dosing of patients.
  • enhanced stability in human hepatocytes is a favorable characteristic for compounds that are to be used for drugs.
  • the present invention provides novel compounds according to formula (I)
  • A is selected from the group consisting of phenyl, naphthyl, thiophenyl, benzothiophenyl or benzofuranyl, optionally substituted with one or two members of the group consisting of H, F, Cl, Br, C 1-4 -alkyl, F 1-3 -fluoro-C 1-4 -alkyl, CN, OCH 3 , cyclopropyl, and cyclobutyl,
  • A is selected from
  • R 1 is selected from H, C 1-4 -alkyl, F 1-3 -fluoro-C 1-4 -alkyl, C 1-4 -alkyl-OH or C 1-4 -alkyl-CN;
  • R 2 is selected from C 1-2 -alkyl or Cl
  • R 1 and R 2 are each CH 2 joined via a bond forming a 6-membered ring;
  • R 3 is selected from H, or C 1-4 -alkyl.
  • A is selected from the group consisting of phenyl, thiophenyl or benzofuranyl, optionally substituted with one or two members of the group consisting of H, F, Cl, Br, C 1-4 -alkyl, F 1-3 -fluoro-C 1-4 -alkyl, CN, OCH 3 , cyclopropyl, and cyclobutyl,
  • A is selected from
  • A is selected from the group consisting of phenyl, thiophenyl or benzofuranyl, optionally substituted with one or two members of the group consisting of H, F, Cl, Br, C 1-4 -alkyl, F 1-3 -fluoro-C 1-4 -alkyl, CN, OCH 3 , cyclopropyl, and cyclobutyl,
  • A is selected from
  • Another embodiment of the present invention relates to a compound of formula (I), wherein A is selected from the group consisting of phenyl, thiophenyl or benzofuranyl, optionally substituted with one or two members of the group consisting of H, F, Br, Cl, and CH 3 ,
  • A is selected from
  • Another embodiment of the present invention relates to a compound of formula (I), wherein A is selected from the group consisting of phenyl, thiophenyl or benzofuranyl, optionally substituted with one or two members of the group consisting of H, F, Br, Cl, and —CH 3 ,
  • A is selected from
  • Another embodiment of the present invention relates to a compound of formula (I), wherein A is selected from the group consisting of
  • Another embodiment of the present invention relates to a compound of formula (I), wherein R 1 is selected from the group consisting of H and C 1-4 -alkyl; and substituents A and R 2 are defined as in any of the preceding embodiments.
  • Another embodiment of the present invention relates to a compound of formula (I), wherein R 1 is selected from the group consisting of H and H 3 C; and substituents A and R 2 are defined as in any of the preceding embodiments.
  • Another embodiment of the present invention relates to a compound of formula (I), wherein R 2 is H 3 C or Cl; and substituents A and R 1 are defined as in any of the preceding embodiments.
  • Another embodiment of the present invention relates to a compound of formula (I), wherein R 3 is H or H 3 C; and substituents A, R 1 and R 2 are defined as in any of the preceding embodiments.
  • C 1-6 -alkyl means an alkyl group or radical having 1 to 6 carbon atoms.
  • groups like HO, H 2 N, (O)S, (O) 2 S, NC (cyano), HOOC, F 3 C or the like the skilled artisan can see the radical attachment point(s) to the molecule from the free valences of the group itself.
  • aryl-C 1-3 -alkyl means an aryl group which is bound to a C 1-3 -alkyl-group, the latter of which is bound to the core or to the group to which the substituent is attached.
  • the numeration of the atoms of a substituent starts with the atom that is closest to the core or to the group to which the substituent is attached.
  • 3-carboxypropyl-group represents the following substituent:
  • the asterisk may be used in sub-formulas to indicate the bond that is connected to the core molecule as defined.
  • C 1-n -alkyl wherein n is an integer selected from 2, 3, 4 or 5, either alone or in combination with another radical denotes an acyclic, saturated, branched or linear hydrocarbon radical with 1 to n C atoms.
  • C 1-5 -alkyl embraces the radicals H 3 C—, H 3 C—CH 2 —, H 3 C—CH 2 —CH 2 —, H 3 C—CH(CH 3 )—, H 3 C—CH 2 —CH 2 —CH 2 —, H 3 C—CH 2 —CH(CH 3 )—, H 3 C—CH(CH 3 )—CH 2 —, H 3 C—C(CH 3 ) 2 —, H 3 C—CH 2 —CH 2 —CH 2 —CH 2 —, H 3 C—CH 2 —CH 2 —CH(CH 3 )—, H 3 C—CH 2 —CH(CH 3 )—CH 2 —, H 3 C—CH(CH 3 )—CH 2 —, H
  • F 1-m -fluoro-C 1-n -alkyl wherein m is an integer selected from 2 or 3, and n is an integer selected from 2, 3, 4 or 5, denotes an C 1-n -alkyl group as defined hereinbefore wherein one or more hydrogen atoms are replaced by 1-m fluorine.
  • F 1-3 -fluoro-C 1-2 -alkyl embraces the radicals FH 2 C, F 2 HC, F 3 C, FH 2 C—H 2 C, F 2 HC—H 2 C, F 3 CH 2 C, FH 2 C—FHC, FH 2 C—F 2 C, F 2 HC—FHC, H 3 C—FHC, and H 3 C—F 2 C.
  • phenyl refers to the radical of the following ring
  • naphthyl refers to the radical of the following ring
  • thiophenyl refers to the radical of the following ring
  • thienopyrimidone refers to the radical of the following ring
  • benzothiophenyl refers to the radical of the following ring
  • benzofuranyl refers to the radical of the following ring
  • cyclopropyl refers to the radical of the following ring
  • cyclobutyl refers to the radical of the following ring
  • tetrazolyl refers to the radical of the following ring
  • substituted means that any one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valence is not exceeded, and that the substitution results in a stable compound.
  • a given chemical formula or name shall encompass tautomers and all stereo, optical and geometrical 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 diastereomers, 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.
  • substantially pure stereoisomers can be obtained according to synthetic principles 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.
  • phrases “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.
  • 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.
  • 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, methanesulfonic acid, 4-methyl-benzenesulfonic acid, phosphoric acid, salicylic acid, succinic acid, sulfuric acid and tartaric acid.
  • 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, methanesulfonic acid, 4-methyl-benzenesulfonic acid, phosphoric acid, salicylic acid, succinic acid, sulfur
  • 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)-aminomethane.
  • 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.
  • 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 comprise a part of the present invention.
  • 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 system (Molecular Devices).
  • 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 0.4 mg/ML Geneticin). It is important that density does not exceed 90% confluence.
  • MEM/EBSS medium with 10% FCS and 0.4 mg/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 detached, washed twice with medium (MEM/EBSS medium with 10% FCS) and 20000 cells in 20 ⁇ l/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.
  • 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 (1 ⁇ HEPES, Cat. 14065 from Gibco, 20 mM HEPES, Cat. 83264 from SIGMA, 0.1% BSA Cat. 11926 from Invitrogen, pH 7.4
  • 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 luminescence (0% CTL; low controls).
  • the metabolic degradation of the test compound is assayed at 37° C. with pooled liver microsomes.
  • the final incubation volume of 100 ⁇ l 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 ⁇ M.
  • the reactions are initiated by addition of beta-nicotinamide adenine dinucleotide phosphate, reduced form (NADPH, 1 mM) and terminated 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 without NADPH, terminated at the last time point. The [%] remaining test compound after NADPH independent incubation is reflected by the parameter c(control) (metabolic stability). The quenched incubations are pelleted by centrifugation (10000 g, 5 min).
  • CL_INTRINSIC The intrinsic clearance (CL_INTRINSIC) is calculated by considering the amount of protein in the incubation:
  • Hepatocellularity human: 120 ⁇ 10e6 cells/g liver
  • Liver factor, human 25.7 g/kg bodyweight
  • the metabolic degradation of the test compound is assayed in a hepatocyte suspension.
  • Hepatocytes (cryopreserved) are incubated in.
  • Dulbecco's modified eagle medium supplemented with 3.5 ⁇ g glucagon/500 mL, 2.5 mg insulin/500 mL and 3.75 mg/500 mL hydrocortison) containing 5% or 50% species serum.
  • test compound solution 80 ⁇ M; from 2 mM in DMSO stock solution diluted 1:25 with medium
  • 395 ⁇ l hepatocyte suspension cell density in the range 0.25-5 Mio cells/mL depending on the species, typically 1 Mio cells/mL; final concentration of test compound 1 ⁇ M, final DMSO concentration 0.05%).
  • the cells are incubated for six hours (incubator, orbital shaker) and samples (25 ⁇ l) 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, evaporated under nitrogen and resuspended.
  • CO initial concentration in the incubation [ ⁇ M]
  • CD cell density of vital cells [10e6 cells/mL]
  • AUD area under the data [ ⁇ M ⁇ h]
  • clast concentration of last data point [ ⁇ M]
  • 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 [ml/min/kg] CL_INTRINSIC_INVIVO [ml/min/kg] ⁇ hepatic blood flow [ml/min/kg]/(CL_INTRINSIC_INVIVO [ml/min/kg]+hepatic blood flow [ml/min/kg])
  • Hepatocellularity human: 120 ⁇ 10e6 cells/g liver
  • Liver factor, human 25.7 g/kg bodyweight
  • 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 glucagon/500 ml, 2.5 mg insulin/500 ml and 3.75 mg/500 ml hydrocortisone.
  • test compound 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 observed with primary human hepatocytes), a final test compound concentration of 10 ⁇ M, 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 centrifugation. The supernatant is transferred to a 96-deepwell plate, evaporated under nitrogen and resuspended prior to bioanalysis by liquid chromatography-high resolution mass spectrometry for identification of putative metabolites.
  • 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 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 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.
  • Cough such as chronic idiopathic cough or chronic refractory cough, cough associated with asthma, COPD and lung cancer and post-viral cough.
  • 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, lymangioleiomyomatosis, 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
  • 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, scleroderma/systemic sclerosis, mediastinal fibrosis, myelofibrosis, Peyronie's disease, nephrogenic systemic fibrosis, retroperitoneal fibrosis, adhesive capsulitis.
  • NASH non-alcoholic steatohepatitis
  • Inflammatory, auto-immune or allergic diseases and conditions such as allergic or non-allergic rhinitis or sinusitis, chronic sinusitis or rhinitis, nasal polyposis, chronic rhinosinusitis, acute rhinosinusitis, asthma, pediatric asthma, allergic bronchitis, alveolitis, hyperreactive airways, allergic conjunctivitis, bronchiectasis, adult respiratory distress syndrome, bronchial and pulmonary edema, bronchitis or pneumonitis, eosinophilic cellulites (e.g., Well's syndrome), eosinophilic pneumonias (e.g., Loeffler's syndrome, chronic eosinophilic pneumonia), eosinophilic fasciitis (e.g., Shulman's syndrome), delayed-type hypersensitivity, non-allergic asthma; exercise induced bronchoconstriction; chronic obstructive pulmonary disease (COPD), acute bronchitis, chronic bron
  • Pain such as chronic idiopathic pain syndrome, neuropathic pain, dysesthesia, allodynia, migraine, dental pain and post-surgical pain.
  • the present invention relates to a compound of general formula 1 for use as a medicament.
  • 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 modulated by TRPA1 activity.
  • 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 immunoregulatory 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 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.
  • the present invention relates to the use of a compound of general formula 1 for the treatment and/or prevention of:
  • Cough such as chronic idiopathic cough or chronic refractory cough, cough associated with asthma, COPD and lung cancer and post-viral cough.
  • 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, lymangioleiomyomatosis, 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
  • 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, scleroderma/systemic sclerosis, mediastinal fibrosis, myelofibrosis, Peyronie's disease, nephrogenic systemic fibrosis, retroperitoneal fibrosis, adhesive capsulitis.
  • NASH non-alcoholic steatohepatitis
  • atrial fibrosis endomyocardial fibrosis
  • endomyocardial fibrosis old myocardial infarction
  • glial scar old myocardial infarction
  • glial scar glial scar
  • arterial stiffness arthrofibrosis
  • Inflammatory, auto-immune or allergic diseases and conditions such as allergic or non-allergic rhinitis or sinusitis, chronic sinusitis or rhinitis, nasal polyposis, chronic rhinosinusitis, acute rhinosinusitis, asthma, pediatric asthma, allergic bronchitis, alveolitis, hyperreactive airways, allergic conjunctivitis, bronchiectasis, adult respiratory distress syndrome, bronchial and pulmonary edema, bronchitis or pneumonitis, eosinophilic cellulites (e.g., Well's syndrome), eosinophilic pneumonias (e.g., Loeffler's syndrome, chronic eosinophilic pneumonia), eosinophilic fasciitis (e.g., Shulman's syndrome), delayed-type hypersensitivity, non-allergic asthma; exercise induced bronchoconstriction; chronic obstructive pulmonary disease (COPD), acute bronchitis, chronic bron
  • Pain such as chronic idiopathic pain syndrome, neuropathic pain, dysesthesia, allodynia, migraine, dental pain and post-surgical pain.
  • 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.
  • the present invention relates to the use of a compound of general formula 1 for the preparation of a medicament for the treatment and/or prevention of above mentioned diseases and conditions.
  • the present invention relates to methods for the treatment or prevention of above mentioned diseases and conditions, which method comprises the administration of an effective amount of a compound of general formula 1 to a human being.
  • the compounds of the invention may further be combined with one or more, preferably one additional therapeutic agent.
  • 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, inflammatory and immunoregulatory disorders, respiratory or gastrointestinal diseases or complaints, inflammatory diseases of the joints or of the nasopharynx, eyes, and skin or conditions such as for example cough, idiopathic pulmonary fibrosis, other pulmonary interstitial diseases, asthma or allergic diseases, eosinophilic diseases, chronic obstructive pulmonary disease, atopic dermatitis as well as autoimmune pathologies, such as rheumatoid arthritis and atherosclerosis, or therapeutic agents useful for the treatment of ophthalmic diseases, 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 substances with respect to one of the indications mentioned and/or allow the dosage of one or more active substances to be reduced.
  • 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-convulsants, anxiolytics, sedatives, skeletal muscle relaxants or anti-depressants.
  • additional therapeutic agents selected from the group consisting of antifibrotic agents, anti-tussive agents, anti-inflammatory agents, anti-atopic dermatitis agents, analgesics, anti-convulsants, 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; connective tissue growth factor (CTGF) blocking antibodies such as Pamrevlumab; B-cell activating factor receptor (BAFF-R) blocking antibodies such as Lanalumab; alpha-V/beta-6 blocking inhibitors such as BG-00011/STX-100, recombinant pentraxin-2 (PTX-2) such as PRM-151; c-Jun N-terminal kinase (JNK) inhibitors such as CC-90001; galectin-3 inhibitors 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 Kin
  • 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 kinase (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-beta1) small interfering RNA such as TRK250/BNC-1021; or lysophosphatidic acid receptor antagonists such as BMS-986278.
  • LXL2 Lysyl Oxidase Like 2
  • 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 antagonist such as Orvepitant, Aprepitant; nicotinic acetylcholine receptor alpha 7 subunit stimulator such as ATA-101/bradanicline; codeine, gabapentin, pregablin, or azithromycin.
  • Anti-inflammatory agents are, for example, corticosteroids such as prednisolone or dexamethasone; cyclo-oxygenase-2 (COX2) inhibitors such as celecoxib, rofecoxib, parecoxib, valdecoxib, deracoxib, etoricoxib or lumiracoxib; prostaglandin E2 antagonists; leukotriene B4 antagonists; leukotriene D4 antagonists such as monteleukast; 5-lipoxygenase inhibitors; or other nonsteroidal anti-inflammatory agents (NSAIDs) such as aspirin, diclofenac, diflunisal, etodolac, ibuprofen or indomethacin.
  • COX2 cyclo-oxygenase-2
  • Anti-atopic dermatitis agents are, for example, cyclosporin, methotrexate, mycophenolate mofetil, azathioprine, phosphodiesterase inhibitors (e.g. apremilast, crisaborole), Janus Associated 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).
  • cyclosporin methotrexate
  • mycophenolate mofetil e.g. apremilast, crisaborole
  • Janus Associated Kinase (JAK) inhibitors e.g. tofacitinib
  • neutralizing antibodies against IL-4/IL-13 e.g. dupilamab
  • IL-13 e.g. lebriki
  • Analgesics are, for example, of the opioid type, such as morphine, oxymorphine, levopanol, oxycodon, propoxyphene, nalmefene, fentanyl, hydrocondon, hydromorphone, meripidine, methadone, nalorphine, naloxone, naltrexone, buprenorphine, butorphanol, nalbuphine, pentazocine; or of the non-opioid type, such as acetophenamine
  • Anti-depressants are, for example, tricyclic anti-depressants such as amitriptyline, clomipramine, despramine, doxepin, desipramine, imipramine, nortriptyline; selective serotonin reuptake inhibitor anti-depressants (SSRIs) such as fluoxetine, paroxetine, sertraline, citalopram, escitalopram; norepinephrine reuptake inhibitor anti
  • Anxiolytics are, for example, benzodiazepines such as alprazolam, bromazepam, chlordiazepoxide, clonazepam, clorazepate, diazepam, flurazepam, lorazepam, oxazepam, temazepam, triazolam, or tofisopam; or they are nonbenzodiazepine hypnoticssuch as eszopiclone, zaleplon, zolpidem, or zopiclone; or they are carbamates e.g. meprobamate, carisoprodol, tybamate, or lorbamate; or they are antihistamines such as hydroxyzine, chlorpheniramine or diphenhydramine.
  • benzodiazepines such as alprazolam, bromazepam, chlordiazepoxide, clonazepam, clorazepate,
  • Sedatives are, for example, barbiturate sedatives, such as amobarbital, aprobarbital, butabarbital, butabital, mephobarbital, metharbital, methohexital, pentobarbital, secobarbital, talbutal, theamylal, or thiopental; or they are non-barbiturate sedatives such as glutethimide, meprobamate, methaqualone or dichloalphenazone.
  • barbiturate sedatives such as amobarbital, aprobarbital, butabarbital, butabital, mephobarbital, metharbital, methohexital, pentobarbital, secobarbital, talbutal, theamylal, or thiopental
  • non-barbiturate sedatives such as glutethimide, meprobamate, methaqualone or dichloalphenazone.
  • Skeletal muscle relaxants are, for example, baclofen, meprobamate, carisoprodol, cyclobenzaprine, metaxalone, methocarbamol, tizanidine, chlorzoxazone or orphenadrine.
  • Acetylcholinesterase inhibitors such as donepezil; 5-HT-3 anatgonists such as ondansetron; metabotropic glutamate receptor antagonists; antiarrhythmics such as mexiletine or phenytoin; or NMDA receptor antagonists.
  • incontinence medications for example, antichos linergics such as oxybutynin, tolterodine, darifenacin, fesoterodine, solifenacin or trospium; or they are bladder muscle relaxants such as mirabegron; or they are alpha blockers such as tamsulosin, alfuzosin, silodosin, doxazosin or terazosin.
  • antichos linergics such as oxybutynin, tolterodine, darifenacin, fesoterodine, solifenacin or trospium
  • bladder muscle relaxants such as mirabegron
  • 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.
  • 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 affected or which are mediated by TRPA1, in particular diseases or conditions as described hereinbefore and hereinafter.
  • 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.
  • this invention relates to the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof in combination with one or more additional therapeutic agents for the treatment of diseases or conditions which can be influenced by the inhibition of TRPA1 in a patient in need thereof.
  • 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 administering 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 effective amount of one or more additional therapeutic agents described in hereinbefore and hereinafter.
  • this invention relates to a pharmaceutical composition that comprises a compound according to the invention and one or more additional therapeutic agents described hereinbefore and hereinafter, optionally together with one or more inert carriers and/or diluents.
  • the present invention relates to the use of a compound according to the invention in a cough-measuring device.
  • the compounds according to the present 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 literature of organic synthesis.
  • the compounds are obtained in analogous fashion to the methods of preparation explained more fully hereinafter, in particular as described in the experimental section.
  • the order in carrying out the reaction steps may be varied. Variants of the reaction methods that are known to the one skilled in the art but not described in detail here may also be used.
  • the compounds according to the invention are prepared by the methods of synthesis described hereinafter in which the substituents of the general formulae have the meanings given hereinbefore. These methods are intended as an illustration of the invention without restricting its subject matter and the scope of the compounds claimed to these examples. Where the preparation of starting compounds is not described, they are commercially obtainable or may be prepared analogously to known compounds or methods described herein. Substances described in the literature are prepared according to the published methods of synthesis.
  • chloromethyltetrazole is N-alkylated with an appropriate aromatic or heteroaromatic acetophenone derivative carrying a leaving group “LG” (e.g. Cl or Br) alpha to the carbonyl group.
  • LG e.g. Cl or Br
  • the reaction can typically be run at ambient temperature or 50° C. in the presence of a base (e.g. K 2 CO 3 ).
  • the carbonyl group of side chain (A) can be reduced in an enantioselective fashion by using appropriate catalytic systems using a transition metal complex (of e.g. Ru or IR) in combination with a chiral ligand (e.g.
  • Tricyclic thienopyrimidone compounds may be prepared as shown in Scheme II below.
  • a suitable thieno-tetrahydropyridine precursor is converted with an appropriate reagent such as formamidine, formamide or a salt thereof in a suitable solvent (e.g. EtOH) at elevated temperatures (e.g. 100° C.) into the tricyclic thieno-pyrimidone compound (C).
  • the tetrahydropyridine core is then oxidized using a suitable oxidant (e.g. KMnO 4 ) and is accelerated by the presence of a chelating reagent (e.g. 18-crown-6).
  • a suitable oxidant e.g. KMnO 4
  • a chelating reagent e.g. 18-crown-6
  • This reaction is typically performed in a non-polar solvent (e.g. DCM) and run preferentially at ambient temperature.
  • R 3 resembles a protecting group (e.g. BOC)
  • this group can be removed using suitable conditions for deprotection (e.g.
  • 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 literature of organic synthesis for example using methods described in “Comprehensive Organic 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.
  • the compounds are obtained analogously to the methods of preparation explained more fully hereinafter, in particular as described in the experimental section.
  • 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 des tail herein may also be used.
  • any corresponding functional groups in the starting compounds may be protected using conventional protecting groups. These protecting 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 “Protecting 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 interchangeably and designate a temperature of about 20° C., e.g. between 19 and 24° C.
  • example XII.1 To 45.0 mg (0.195 mmol) of example XII.1 in 1.0 mL DMF is added 81.7 mg (0.215 mmol) 1-[Bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate, 74.8 ⁇ L (0.429 mmol) diisopropylethylamine and 1.17 mL (0.5 M, 0.585 mmol) of a solution of ammonia in THF. The reaction mixture is stirred at ambient temperature for 18 h. The reaction mixture is then purified by preparative HPLC (H 2 O/ACN/NH 3 gradient) to yield the desired product.
  • preparative HPLC H 2 O/ACN/NH 3 gradient
  • Analytical column Stable Bond (Agilent) 1.8 ⁇ m; 3.0 ⁇ 30 mm; column temperature: 60° C.
  • Analytical column Sunfire (Waters) 2.5 ⁇ m; 3.0 ⁇ 30 mm; column temperature: 60° C.
  • Analytical column Zorbax StableBond C18 (Agilent) 1.8 ⁇ m; 2.1 ⁇ 30 mm; column temperature: 60° C.
  • Analytical column Sunfire C18 (Waters) 2.5 ⁇ m; 3.0 ⁇ 30 mm; column temperature: 60° C.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (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)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pulmonology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)
US17/071,092 2019-10-15 2020-10-15 Novel Thienopyrimidones Abandoned US20210107919A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/463,942 US11578080B2 (en) 2019-10-15 2021-09-01 Thienopyrimidones

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP19203173 2019-10-15
EP19203173.0 2019-10-15

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/463,942 Continuation US11578080B2 (en) 2019-10-15 2021-09-01 Thienopyrimidones

Publications (1)

Publication Number Publication Date
US20210107919A1 true US20210107919A1 (en) 2021-04-15

Family

ID=68289874

Family Applications (2)

Application Number Title Priority Date Filing Date
US17/071,092 Abandoned US20210107919A1 (en) 2019-10-15 2020-10-15 Novel Thienopyrimidones
US17/463,942 Active US11578080B2 (en) 2019-10-15 2021-09-01 Thienopyrimidones

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/463,942 Active US11578080B2 (en) 2019-10-15 2021-09-01 Thienopyrimidones

Country Status (14)

Country Link
US (2) US20210107919A1 (es)
EP (1) EP4045144A1 (es)
JP (1) JP7354438B2 (es)
KR (1) KR20220084127A (es)
CN (1) CN114945408B (es)
AR (1) AR120221A1 (es)
AU (1) AU2020366678A1 (es)
BR (1) BR112022003729A2 (es)
CA (1) CA3153115A1 (es)
CL (1) CL2022000757A1 (es)
IL (1) IL292095A (es)
MX (1) MX2022004564A (es)
TW (1) TW202128705A (es)
WO (1) WO2021074197A1 (es)

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7754709B2 (en) * 2003-06-10 2010-07-13 Solvay Pharmaceuticals Bv Tetracyclic thiophenepyrimidinone compounds as inhibitors of 17β hydroxysteroid dehydrogenase compounds
MY159059A (en) * 2010-12-20 2016-12-15 Glenmark Pharmaceuticals Sa 2-amino-4-arylthiazole compounds as trpa1 antagonists
MX2013014679A (es) * 2011-06-13 2014-07-14 Glenmark Pharmaceuticals Sa Tratamiento de trastornos respiratorios usando antagonistas de trpa1.
EP3180345B1 (en) * 2014-08-11 2018-10-10 Hydra Biosciences, Inc. Thieno- and furo[2,3-d]pyrimidine-2,4[1h,3h]-dione derivatives as trpc5 modulators for the treatment of neuropsychiatric disorders
WO2016081649A1 (en) * 2014-11-18 2016-05-26 Emory University Thieno[2,3-d]pyrimidin-4-one derivatives as nmdar modulators and uses related thereto
WO2016133888A1 (en) * 2015-02-16 2016-08-25 Biota Pharmaceuticals, Inc. Compounds for treating respiratory syncytial virus infections
WO2017060488A1 (en) * 2015-10-09 2017-04-13 Almirall, S.A. New trpa1 antagonists

Also Published As

Publication number Publication date
US20210395267A1 (en) 2021-12-23
AR120221A1 (es) 2022-02-02
JP7354438B2 (ja) 2023-10-02
MX2022004564A (es) 2022-05-06
CN114945408A (zh) 2022-08-26
EP4045144A1 (en) 2022-08-24
US11578080B2 (en) 2023-02-14
BR112022003729A2 (pt) 2022-05-31
TW202128705A (zh) 2021-08-01
IL292095A (en) 2022-06-01
CL2022000757A1 (es) 2023-01-20
KR20220084127A (ko) 2022-06-21
WO2021074197A1 (en) 2021-04-22
CN114945408B (zh) 2024-04-16
JP2022551956A (ja) 2022-12-14
AU2020366678A1 (en) 2022-03-31
CA3153115A1 (en) 2021-04-22

Similar Documents

Publication Publication Date Title
US11858921B2 (en) Uracil derivatives as TRPA1 inhibitors
US11884652B2 (en) Tetrazole derivatives as TRPA1 inhibitors
US11884681B2 (en) 3H,4H,5H,6H,7H-pyrimido[4,5-b][1,4]oxazine-4,6-dione derivatives as TRPA1 inhibitors
US11891403B2 (en) Tetrazole derivatives as TRPA1 inhibitors
EP4045053B1 (en) Novel tetrazoles
US11578080B2 (en) Thienopyrimidones
US11136336B2 (en) Thienopyrimidones
US11878981B2 (en) Substituted 1,2,4-oxadiazoles as TRPA1 inhibitors
US11661427B2 (en) Tetrazole derivatives as TRPA1 inhibitors
US11661430B2 (en) Tetrazole derivatives as TRPA1 inhibitors
EP4228756B1 (en) Tetrazole derivatives as trpa1 inhibitors
US20220340590A1 (en) 3h,4h-thieno[2,3-d]pyrimidin-4-one derivatives as trpa1 inhibitors

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO PAY ISSUE FEE

AS Assignment

Owner name: BOEHRINGER INGELHEIM INTERNATIONAL GMBH, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:BOEHRINGER INGELHEIM PHARMA GMBH & CO. KG;REEL/FRAME:058843/0229

Effective date: 20210921

Owner name: BOEHRINGER INGELHEIM PHARMA GMBH & CO. KG, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HEHN, JOERG P.;DAHMANN, GEORG;WILLWACHER, JENS;AND OTHERS;SIGNING DATES FROM 20210824 TO 20210916;REEL/FRAME:058843/0096