WO2014207213A1 - Novel inhibitors of protein kinase c epsilon signaling - Google Patents

Novel inhibitors of protein kinase c epsilon signaling Download PDF

Info

Publication number
WO2014207213A1
WO2014207213A1 PCT/EP2014/063723 EP2014063723W WO2014207213A1 WO 2014207213 A1 WO2014207213 A1 WO 2014207213A1 EP 2014063723 W EP2014063723 W EP 2014063723W WO 2014207213 A1 WO2014207213 A1 WO 2014207213A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
compound
cancer
diabetic
disorder
Prior art date
Application number
PCT/EP2014/063723
Other languages
French (fr)
Inventor
Johann Hofmann
Thierry Langer
Johannes KIRCHMAIR
Florian RECHFELD
Peter Gruber
Markus BÖHLER
Georg HECHENBERGER
Dorota GARCZARCZYK
Maria N. Preobrazhenskaya
Gennady B. LAPA
Original Assignee
Medizinische Universität Innsbruck
Universität Innsbruck
Gause Institute Of New Antibiotics
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 Medizinische Universität Innsbruck, Universität Innsbruck, Gause Institute Of New Antibiotics filed Critical Medizinische Universität Innsbruck
Publication of WO2014207213A1 publication Critical patent/WO2014207213A1/en

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
    • C07D495/12Heterocyclic 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4743Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having sulfur as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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

Definitions

  • Novel inhibitors of protein kinase C epsilon signaling Novel inhibitors of protein kinase C epsilon signaling
  • the present invention relates to novel inhibitors of protein kinase C epsilon (PKCE) signaling, including in particular the compounds of formula (I) as described and defined herein, pharmaceutical compositions comprising these inhibitors, and their use in the treatment or prevention of disorders such as, e.g., a cardiovascular disorder, cardiac hypertrophy, heart failure, anxiety, pain, chronic pain, migraine, an allergy, an inflammatory disorder, an autoimmune disorder, diabetes, diabetic complications, diabetic retinopathy, diabetic nephropathy, diabetic cardiomyopathy, diabetic neuropathy, cancer, metastatic cancer, drug-resistant cancer, stomach cancer, lung cancer, thyroid cancer, colon cancer, breast cancer, a neurological disorder, Alzheimer's disease, Parkinson's disease, bipolar disorder, stroke, alopecia, or alcoholism.
  • PKCE protein kinase C epsilon
  • PKC Protein kinase C
  • the PKC isozymes can be classified into three groups: i) the conventional isozymes ⁇ , ⁇ , ⁇ , and ⁇ ; ii) the novel isozymes ⁇ , ⁇ , ⁇ , and ⁇ ; and iii) the atypical isozymes ⁇ / ⁇ (mouse/human) and ⁇ .
  • PKC isozymes seem to play important roles in the activation of signal transduction pathways leading to synaptic transmissions, the activation of ion fluxes, secretion, proliferation, cell cycle control, differentiation and tumorigenesis.
  • PKC isozymes are described, e.g. , in: Mochly-Rosen et al., 2012; Sanchez-Bautista et al., 2013; Bohler, 2006; US 2009/0124553; and US 6,376,467.
  • the PKCE isozyme was reported to participate in neoplastic transformation (Gorin et al., 2009), cardiac hypertrophy (Pass et al., 2001 ), protection from ischemic insult (Pass et al., 2001 ; Johnson et al., 1996), nociceptor function (Cesare et al., 1999), macrophage activation (Castrillo et al. , 2001 ), diabetes (Ikeda et al., 2001 ) and in alcohol consumption (Choi et al., 2002).
  • a PKCe isozyme-specific inhibitor would have pharmaceutical potential, e.g., for the intervention in stroke, Alzheimer's disease or pain (Akita, 2008; Shirai et al., 2008).
  • kinase inhibitors interact with the ATP-binding site, which is well conserved among different kinase families, and is even more so within a family. This poses a serious hurdle for the development of isozyme-specific inhibitors, as there are approximately 518 kinases encoded by the human genome (Manning et al., 2002). Although several selective kinase inhibitors have been reported initially, later it turned out that they also inhibit other targets. For example, the marketed drug imatinib mesylate (Gleevec/Glivec) was developed as an inhibitor of the oncoprotein Bcr-Abl.
  • PKCp-selective inhibitors such as ruboxistaurine (Fedorov et al., 2007; Nakamura et al., 2010) and enzastaurine (Chen et al., 2008) are in clinical trials for diabetic retinopathy and cancer, respectively.
  • Rottlerin was described as a specific inhibitor of PKC5.
  • additional modes of action have been observed by now (Soltoff, 2007).
  • Small molecule inhibitors of protein-protein interactions have previously been shown to be useful for pharmacological purposes (Beeley, 2000; Arkin et al., 2004).
  • the present invention provides the compounds of formula (I) as described herein below and in the claims.
  • Structurally related compounds have also been disclosed in: Sanam et al. , 2010; US 201 1 /0077250; WO 2013/033037; CN 102757447 A; Pillai et al., 2010; US 2013/0129677; RU 2371444 C1 ; WO 92/03427; and CAS registry nos. 338433-13-9 and 333330-1 1 -3.
  • the barbituric acid derivative BAS 02104951 has furthermore been disclosed as an inhibitor of PKCe signal transduction in Gruber et al., 201 1 .
  • the present invention provides a compound of the following formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof, for use as a medicament:
  • n is an integer of 0 to 4 (i.e. , 0, 1 , 2, 3 or 4), and each R 1 is independently selected from C 1-4 alkyl, C 2 remedy 4 alkenyl, C 2 . 4 alkynyl, -OH, -0(C 1-4 alkyl), -0(C-,. 4 alkyl)-OH, -0(d_ 4 alkyl)-0(C 1-4 alkyl), -SH, -S(C 1-4 alkyl), -S(C 1-4 alkyl)-SH, or -S(C,. 4 alkyl)-S(d.
  • n is 2, 3 or 4
  • two groups R 1 attached to adjacent carbon atoms are mutually linked to form a group -0-CH 2 -0-, -0-CH 2 -CH 2 -0- or -0-CH 2 -CH 2 -CH 2 -0-, while the further group(s) R ⁇ if present (i.e. , if n is 3 or 4), is/are independently selected from C,. 4 alkyl, C 2 .
  • n is an integer of 1 to 4 (i.e. , 1 , 2, 3 or 4)
  • one group R is selected from -0(C 1 -4 alkyl), -OH, -0(C 1-4 alkyl)-OH, or -0(d_ 4 alky!-0(d_ 4 alkyl)
  • the remaining group(s) R 1 if present (i.e., if n is 2, 3 or 4), is/are independently selected from d_ 4 alkyl, C 2-4 alkenyl, C 2-4 aikynyl, -OH, -0(Ci_ 4 alkyl), -0(C 1-4 alkyl)-OH, -0(C 1 -4 alkyl)-0(d_ 4 alkyl), -SH, -S(C 1-4 alkyl), -S(C 1-4 alkyl)-SH, or -S(C 1-4 alkyl)-S(C 1 -4 alkyl), -NH
  • n 2 aikyl(C - alkyl), halogen, -CF 3 , or -CN; or, alternatively, n is 2, and the two groups R 1 are attached to adjacent carbon atoms (preferably the same two carbon atoms as in the compounds 1a and 1 b shown below) and are mutually linked to form a group -0-CH 2 -0-, -0-CH 2 -CH 2 -0- or -0-CH 2 -CH 2 -CH 2 -0-.
  • n is 1 and R 1 is selected from -0(C -4 alkyl), -OH, -0(C 1-4 alkyI)-OH, or -0(C 1-4 alkyl)-0(d_ 4 alkyl), wherein it is particularly preferred that R 1 is selected from -OCH 3 or -OCH 2 CH 3 ; or, alternatively, n is 2 and the two groups R 1 are attached to adjacent carbon atoms (preferably the same two carbon atoms as in the compounds 1 a and 1 b shown below) and are mutually linked to form a group -0-CH 2 -0- or -0-CH 2 -CH 2 -0-.
  • R 2 is selected from hydrogen, C 1-4 alkyl, C 2 . 4 alkenyl, C 2 . 4 aikynyl, -OH, -0(C 1 -4 alkyl), -SH, -S(d_ 4 alkyl), -NH 2 , -NH(C 1-4 alkyl), -N(C 1-4 alkyl)(d. 4 alkyl), halogen, -CF 3 , or -CN.
  • R 2 is hydrogen.
  • R 3 is selected from -NH 2 , -NH(C 1-4 alkyl), -N(d.
  • R 3 is -NH 2 , -NH(d_ 4 alkyl), -OH, or -SH. More preferably, R 3 is -NH 2 .
  • X is selected from S, O, N(H), or N(d. 4 alkyl).
  • X is S or O. More preferably, X is S.
  • L is -(CH 2 )i-4-, wherein one -CH 2 - unit comprised in said -(CH 2 ) -4 - is replaced by a group selected from -CO-NH-, -CO-N(d. 4 alkyl)-, -NH-CO-, -N(d_ 4 alkyl)-CO-, -0-, -CO-, -NH-, -N(d_ 4 alkyl)-, -S-, -SO-, or -S0 2 -.
  • L is -(CH 2 ) 1-3 -, wherein one -CH 2 - unit comprised in said -(CH 2 ) 1-3 - is replaced by a group selected from -CO-NH-, -CO-N(d. 4 alkyl)-, -NH-CO-, or -N(d. 4 alkyl)-CO-. More preferably, L is directionally selected from -CO- NH-, -CO-N(d. 4 alkyl)-, -NH-CO-, or -N(d. 4 alkyl)-CO-.
  • L is -CO-NH- or -CO-N(d_ 4 alkyl)-, wherein the nitrogen atom comprised in said -CO-NH- or said -CO- N(d- 4 alkyl)- is attached to ring A.
  • L is -CO-NH-, wherein the nitrogen atom comprised in said -CO-NH- is attached to ring A.
  • A is aryl or heteroaryl, wherein said aryl or said heteroaryl is optionally substituted with one or more groups (such as, e.g., one, two, three or four groups; preferably, one or two groups; more preferably, one group) independently selected from d prison 4 alkyl, C 2-4 alkenyl, C 2 . 4 alkynyl, -OH, -0(C 1-4 alkyl), -SH, -S(d. 4 alkyl), -NH 2 , -NH(d. 4 alkyl), -N(d. 4 alkyl)(C 1-4 alkyl), halogen, -CF 3 , or -CN.
  • groups such as, e.g., one, two, three or four groups; preferably, one or two groups; more preferably, one group
  • groups such as, e.g., one, two, three or four groups; preferably, one or two groups; more preferably, one group
  • groups such as, e
  • Said aryl is preferably phenyl or naphthyl, and more preferably said aryl is phenyl.
  • Said heteroaryl is preferably a heteroaryl having 5 or 6 ring atoms, wherein 1 , 2 or 3 of said ring atoms are heteroatoms independently selected from oxygen, sulfur or nitrogen, and the remaining ones of said ring atoms are carbon atoms, and further wherein said heteroaryl having 5 or 6 ring atoms is optionally fused to a benzene ring.
  • said heteroaryl is selected from thiazolyl (e.g., 1 ,3-thiazol-2-yl), furanyl, thiophenyl (i.e., thienyl), pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, furazanyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, benzothiazolyl (e.g., 1 ,3-benzothiazol-2-yl), benzofuranyl, benzothiophenyl, benzopyrrolyl, benzoimidazolyl, benzopyrazolyl, benzoxazolyl, benzisoxazolyl, benzofurazanyl, benzopyridinyl, benzopyrimidinyl, benzopyrazinyl, or benzopyridazinyl.
  • thiazolyl e.g
  • said heteroaryl is thiazolyl (e.g. , 1 ,3-thiazol-2-yl) or benzothiazolyl (e.g. , 1 ,3-benzothiazol-2-yl).
  • said heteroaryl is benzothiazolyl, particularly 1 .3-benzothiazol-2-yl.
  • A is aryl or heteroaryl, including any of the aforementioned preferred aryl or heteroaryl groups, wherein said aryl or heteroaryl is unsubstituted (i.e., is not substituted with any groups other than R 4 ).
  • A is phenyl, 1 ,3-thiazol-2-yl or 1 ,3-benzothiazol-2-yl, wherein said phenyl, said 1 ,3-thiazol-2-yl or said 1 ,3-benzothiazol-2-yl is optionally substituted with one or more (e.g., one, two or three) groups independently selected from C 1-4 alkyl, C 2 , 4 alkenyl, C 2 diligent 4 alkynyl, -OH, -0(d. 4 alkyl), -SH, -S(C 1-4 alkyl), -NH 2 , -NH(d_ 4 alkyl), -N(C 1-4 alkyl)(d.
  • A is phenyl, 1 ,3-thiazo!-2-yl or 1 ,3-benzothiazol-2-yl, wherein said phenyl, said 1 ,3-thiazol-2-yl or said 1 ,3-benzothiazol-2-yl is unsubstituted (i.e., is not substituted with any groups other than R 4 ). More preferably, A is phenyl which is optionally substituted with one or more (e.g., one, two or three) groups independently selected from d cycle alkyl, C 2-4 alkenyl, C 2 .
  • R 4 is selected from -CO-(C 1-4 alkyl), -CHO, -0(d. 4 alkyl), -OH, -0-CO-(C 1-4 alkyl), -0-CO-0(C 1-4 alkyl), -CO-0(d_ 4 alkyl), -COOH, -CO-NH 2 , -CO-NH-(C 1-4 alkyl), -CO-N(C 1-4 alkyl)(d. 4 alkyl), -0-CO-NH 2 , -0-CO-NH-(C 1-4 alkyl), -0-CO-N(d_ 4 alkyl)(d_ 4 alkyl), -NH-CO-(d.
  • R 4 is selected from -CO-(d. 4 alkyl), -CHO, -0(C 1-4 alkyl), -OH, -0-CO-(C 1-4 alkyl), -0-CO-0(C 1-4 alkyl), -CO-0(C 1-4 alkyl), -COOH, -CO-NH 2 , -CO-NH-(C 1-4 alkyl), -CO-N(C 1-4 alkyl)(C 1-4 alkyl), -0-CO-NH 2 , -0-CO-NH-(C 1-4 alkyl), -0-CO-N(C 1-4 alkyl)(C 1-4 alkyl), -NH-CO-(C 1-4 alkyl), -N(d.
  • R 4 alkyl), -CHO, -0(C 1-4 alkyl), or -OH. Even more preferably, R 4 is -CO-CH 3 , -CO-CH 2 CH 3 , -OCH 3 , or -OCH 2 CH 3 .
  • is a double bond. Accordingly, it is preferred that the compound of formula (I) has the following structure:
  • Particularly preferred compounds of formula (I) are the compounds 1a to 1t shown below as well as pharmaceutically acceptable salts, solvates and prodrugs of each one of these compounds:
  • PLCe141 (also referred to as “PKCe141 ”)
  • PLCe16 PLCe16
  • PLCe129 PLC 15
  • PLCe133 PLCe133
  • PLCe140 PLCe140
  • PLCe2140 PLC 15
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I) as described and defined herein, or a pharmaceutically acceptable salt, solvate or prodrug thereof, in combination with a pharmaceutically acceptable excipient.
  • the compounds according to the present invention have been found to inhibit the interaction of PKCe with its adaptor protein RACK2, as also shown in Examples 2 and 3.
  • Compound 1 b which is an exemplary compound of formula (I) has been demonstrated to inhibit the PKCe/RACK2 interaction in vitro with an IC 50 of 5.9 ⁇ (see Example 2) and to inhibit the phosphorylation of the PKCe-downstream target Elk-1 in HeLa cells with an IC 50 of 1 1.2 ⁇ (see Example 3).
  • a large number of further exemplary compounds of formula (I) has likewise been found to inhibit PKCe signaling (see Example 2).
  • Compound 1 a for example, has been shown to inhibit the PKCe/RACK2 interaction in vitro with an IC 50 of 4.1 ⁇ .
  • the compounds of the present invention are particularly advantageous as they allow the isozyme-specific inhibition of PKCe signaling. Accordingly, it has been demonstrated in Example 4 that compound 1 b inhibits the TPA-induced translocation of PKCE - but not that of PKC5 - from the cytosol to the membrane and, furthermore, compound 1 b has been shown to inhibit the PKCe-induced migration of HeLa cells into a gap (see Example 5).
  • the compounds of the present invention thus fulfill all criteria of an inhibitor of PKCe signal transduction, such as inhibition of Elk-1 phosphorylation, inhibition of PKCe translocation to the membrane and inhibition of cell migration.
  • the compounds of the invention have been shown not to inhibit cell proliferation (see Example 5), which indicates a lack of toxicity and confirms the suitability of these compounds as medicaments.
  • the compounds according to the invention, and in particular the compounds of formula (I) can thus be used for the therapeutic intervention in diseases/disorders in which PKCe signaling is implicated (particularly diseases/disorders associated with an increased activity or a hyperactivity of PKCe signaling), such as, e.g., cardiovascular disorders (e.g., cardiac hypertrophy, hypertrophic cardiomyopathy, or heart failure (including, e.g., congestive heart failure)), anxiety, pain (e.g., chronic pain), migraine, allergies, inflammatory disorders, autoimmune disorders, diabetes (including also insulin resistance), diabetic complications (such as, e.g., diabetic retinopathy, diabetic nephropathy, diabetic cardiomyopathy, or diabetic neuropathy), cancer (such as, e.g., stomach cancer, lung cancer, thyroid cancer, colon cancer, breast cancer; including
  • the present invention provides a compound of formula (I) as described and defined herein, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition comprising any of the aforementioned entities and a pharmaceutically acceptable excipient, for use in the treatment or prevention of cardiovascular disorders (e.g., cardiac hypertrophy, hypertrophic cardiomyopathy, or heart failure (including, e.g., congestive heart failure)), anxiety, pain (e.g., chronic pain), migraine, allergies, inflammatory disorders, autoimmune disorders, diabetes (including also insulin resistance), diabetic complications (such as, e.g.
  • cardiovascular disorders e.g., cardiac hypertrophy, hypertrophic cardiomyopathy, or heart failure (including, e.g., congestive heart failure)
  • anxiety e.g., chronic pain
  • migraine e.g., allergies, inflammatory disorders, autoimmune disorders, diabetes (including also insulin resistance), diabetic complications (such as, e.g.
  • diabetic retinopathy diabetic nephropathy
  • diabetic cardiomyopathy or diabetic neuropathy
  • cancer such as, e.g., stomach cancer, lung cancer, thyroid cancer, colon cancer, breast cancer; including also metastatic cancer, metastasis, drug-resistant cancer, or multidrug-resistant cancer
  • neurological disorders e.g., Alzheimer's disease, Parkinson's disease, bipolar disorder, or stroke; including also neurodegenerative disorders
  • alopecia e.g., Alzheimer's disease, Parkinson's disease, bipolar disorder, or stroke; including also neurodegenerative disorders
  • the invention likewise relates to the use of the compounds provided herein, including the compounds of formula (I) or pharmaceutically acceptable salts, solvates or prodrugs thereof, in the preparation of a medicament for the treatment or prevention of cardiovascular disorders (e.g., cardiac hypertrophy, hypertrophic cardiomyopathy, or heart failure (including, e.g., congestive heart failure)), anxiety, pain (e.g., chronic pain), migraine, allergies, inflammatory disorders, autoimmune disorders, diabetes (including also insulin resistance), diabetic complications (such as, e.g., diabetic retinopathy, diabetic nephropathy, diabetic cardiomyopathy, or diabetic neuropathy), cancer (such as, e.g., stomach cancer, lung cancer, thyroid cancer, colon cancer, breast cancer; including also metastatic cancer, metastasis, drug-resistant cancer, or multidrug-resistant cancer), neurological disorders (e.g., Alzheimer's disease, Parkinson's disease, bipolar disorder, or stroke; including also neurodegenerative disorders), alopecia, or alcoholism
  • the invention also provides a method of treating or preventing a disorder or condition selected from the group consisting of cardiovascular disorders (e.g., cardiac hypertrophy, hypertrophic cardiomyopathy, or heart failure (including, e.g., congestive heart failure)), anxiety, pain (e.g., chronic pain), migraine, allergies, inflammatory disorders, autoimmune disorders, diabetes (including also insulin resistance), diabetic complications (such as, e.g., diabetic retinopathy, diabetic nephropathy, diabetic cardiomyopathy, or diabetic neuropathy), cancer (such as, e.g., stomach cancer, lung cancer, thyroid cancer, colon cancer, breast cancer; including also metastatic cancer, metastasis, drug-resistant cancer, or multidrug-resistant cancer), neurological disorders (e.g., Alzheimer's disease, Parkinson's disease, bipolar disorder, or stroke; including also neurodegenerative disorders), alopecia, and alcoholism, the method comprising the administration of a compound according to the invention, particularly a compound of formula (I) or a pharmaceutical
  • Protein-protein interfaces such as the PKCe/RACK2 interface
  • PKCe/RACK2 interface Protein-protein interfaces, such as the PKCe/RACK2 interface
  • Iigand-binding sites due to their particular physicochemical properties (extensive, hydrophobic, surface-exposed interfaces).
  • Attempts to tackle protein-protein interactions have been reported in recent years but experience is still quite limited and experimental aspects are challenging as well (Rechfeid et al., 201 1 ).
  • the EAVSLKPT peptide derived from the binding site of PKCe to RACK2 and used for molecular modeling in the context of the present invention (see Example 1 ), inhibits this interaction with an IC 5C of 1 .02 ⁇ .
  • a peptidomimetic of EAVSLKPT such as compound 1 b (PKCe141 ) with an IC 50 of 5.9 ⁇ may be close to the optimum already.
  • aurothiomalate which prevents the interaction of PKCi and its adaptor protein Par6, is active in the range of 10 ⁇ (Erdogan et al., 2006). While a compound with an IC 50 in the nanomolar range would be preferable, this is not essential for clinical application.
  • the ribonucleotide reductase inhibitor hydroxyurea inhibits ribonucleotide reductase with an IC 50 of 37.2 ⁇ (Easmon et al., 2001 ) and is used in the clinic as an anticancer agent.
  • IC 50 37.2 ⁇
  • many clinically relevant kinase inhibitors which are active at nanomolar concentrations in vitro have cellular IC 50 values closer to the micromolar range due to the higher physiological concentrations of ATP relative to those typically used for in vitro assays.
  • the invention further relates to novel compounds, namely the compounds 1 a, 1j, 1 k, 11, 1 m, 1 n, 1 o, 1 p, 1 q, 1 r, 1s and 1t having the structures shown above, as well as pharmaceutically acceptable salts, solvates and prodrugs of each one of these compounds.
  • novel compounds namely the compounds 1 a, 1j, 1 k, 11, 1 m, 1 n, 1 o, 1 p, 1 q, 1 r, 1s and 1t having the structures shown above, as well as pharmaceutically acceptable salts, solvates and prodrugs of each one of these compounds.
  • These compounds as provided in the context of the present invention are particularly useful as medicaments, e.g., for the treatment or prevention of diseases/disorders in which PKCE signaling is implicated, as described herein above.
  • the present invention also relates to the compounds shown in Table 1 below, including in particular the compounds PKCe22, PKCe39, PKCe40, PKCe48, PKCe49, PKCe52, PKCe67, PKCe73, PKCe76, PKCe86, PKCe87, PKCe89, PKCe95, PKCe96, PKCe97, PKCe106, PKCe107, PKCe109, PKCe1 14, PKCe125, PKCe143, PKCe145, PKCe2019, PKCe2020, PKCe2021 , PKCe2051 , PKCe2052, PKCe2053, PKCe2054, PKCe2088, PKCe2089, PKCe2090, and PKCe2091 , as well as pharmaceutically acceptable salts, solvates and prodrugs of each one of these compounds.
  • the invention further relates to the use of these compounds as medicaments, including their use in the treatment or prevention of diseases/disorders in which PKCe signaling is implicated, such as the specific diseases/disorders described herein above.
  • the compounds of the present invention including the compounds of formula (I), can be prepared by methods known in the field of synthetic chemistry.
  • the compounds of the invention may be prepared in accordance with or in analogy to the synthetic routes described in Example 8.
  • alkyl refers to a monovalent saturated aliphatic (i.e. non-aromatic) acyclic hydrocarbon group (i.e., a group consisting of carbon atoms and hydrogen atoms), which may be linear or branched and does not comprise any carbon-to- carbon double bond or any carbon-to-carbon triple bond.
  • C 1-4 alkyl denotes an alkyl group having 1 to 4 carbon atoms.
  • Preferred exemplary alkyl groups are methyl, ethyl, propyl, or butyl.
  • alkenyl refers to a monovalent unsaturated aliphatic acyclic hydrocarbon group, which may be linear or branched and comprises at least one carbon-to- carbon double bond while it does not comprise any carbon-to-carbon triple bond.
  • C 2 -4 alkenyl denotes an alkenyl group having 2 to 4 carbon atoms.
  • Preferred exemplary alkenyl groups are ethenyl, propenyl, or butenyl.
  • alkynyl refers to a monovalent unsaturated aliphatic acyclic hydrocarbon group, which may be linear or branched and comprises at least one carbon-to- carbon triple bond and optionally one or more carbon-to-carbon double bonds.
  • C 2 -4 alkynyl denotes an alkynyl group having 2 to 4 carbon atoms.
  • Preferred exemplary alkynyl groups are ethynyl, propynyl, or butynyl.
  • aryl refers to a monovalent aromatic hydrocarbon group, including bridged ring and/or fused ring systems, containing at least one aromatic ring.
  • Aryl may, for example, refer to phenyl, naphthyl or anthracenyl.
  • heteroaryl refers to a monovalent aromatic ring group, which may be a monocyclic ring group or a bridged ring and/or fused ring system (e.g., a bicyclic ring system), said aromatic ring group comprising one or more (such as, e.g., one, two, or three) ring heteroatoms independently selected from O, S, or N, wherein the aromatic ring group may, e.g., have 5 to 14 (particularly 5 or 6) ring atoms.
  • heteroaryl groups include thiazolyl (e.g., 1 ,3-thiazol-2-yl), furanyl, thiophenyl (i.e., thienyl), pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, furazanyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, benzothiazolyl (e.g., 1 ,3-benzothiazol-2-yl), benzofuranyl, benzothiophenyl, benzopyrrolyl, benzoimidazolyl, benzopyrazolyl, benzoxazolyl, benzisoxazolyl, benzofurazanyl, benzopyridinyl, benzopyrimidinyl, benzopyrazinyl, or benzopyridazinyl.
  • thiazolyl e.g.,
  • heterocycloalkyi refers to a 3 to 10 atom ring or ring system containing one or more (e.g., one, two or three) ring heteroatoms independently selected from O, S, or N.
  • Heterocycloalkyi may, for example, refer to tetrahydrofuranyl, piperidinyl, piperazinyl, aziridinyl, azetidinyl, pyrrolidinyl, imidazolidinyl, morpholinyl (e.g., morpholin-4- yl), pyrazolidinyl, tetrahydrothienyl, octahydroquinolinyl, octahydroisoquinolinyl, oxazolidinyl, or isoxazolidinyl.
  • halogen refers to fluoro (-F), chloro (-CI), bromo (-Br) or iodo (-I).
  • the scope of the invention embraces all pharmaceutically acceptable salt forms of the compounds provided herein, including in particular the compounds of formula (I), which may be formed, e.g., by protonation of an atom carrying an electron lone pair which is susceptible to protonation, such as an amino group, with an inorganic or organic acid, or as a salt of a carboxylic acid group with a physiologically acceptable cation as they are well-known in the art.
  • Exemplary base addition salts comprise, for example, alkali metal salts such as sodium or potassium salts; alkaline earth metal salts such as calcium or magnesium salts; ammonium salts; aliphatic amine salts such as trimethylamine, triethylamine, dicyclohexylamine, ethanolamine, diethanolamine, triethanolamine, procaine salts, meglumine salts, or ethylenediamine salts; aralkyl amine salts such as N,N- dibenzylethylenediamine salts, benethamine salts; heterocyclic aromatic amine salts such as pyridine salts, picoline salts, quinoline salts or isoquinoline salts; quaternary ammonium salts such as tetramethylammonium salts, tetraethylammonium salts, benzyltrimethylammonium salts, benzyltriethylammonium salts, benzyltributylammonium salts,
  • Exemplary acid addition salts comprise, for example, mineral acid salts such as hydrochloride, hydrobromide, hydroiodide, sulfate salts, nitrate salts, phosphate salts (such as, e.g., phosphate, hydrogenphosphate, or dihydrogenphosphate salts), carbonate salts, hydrogencarbonate salts or perchlorate salts; organic acid salts such as acetate, propionate, butyrate, pentanoate, hexanoate, heptanoate, octanoate, cyclopentanepropionate, undecanoate, lactate, maleate, oxalate, fumarate, tartrate, malate, citrate, nicotinate, benzoate, salicylate or ascorbate salts; sulfonate salts such as methanesulfonate, ethanesulfonate, 2-hydroxyethanesulfonate, benzenesulfonate
  • the scope of the invention embraces the compounds provided herein, including in particular the compounds of formula (I), in any solvated form, including, e.g., solvates with water, for example hydrates, or with organic solvents such as, e.g., methanol, ethanol or acetonitrile, i.e., as a methanolate, ethanolate or acetonitrilate, respectively, or in the form of any polymorph.
  • the formulae in the present specification are intended to cover all possible stereoisomers, including enantiomers and diastereomers, of the indicated compounds.
  • all stereoisomers of the compounds of the present invention are contemplated as part of the present invention, either in admixture or in pure or substantially pure form.
  • the scope of the compounds according to the invention embraces all of the possible stereoisomers and their mixtures, it particularly embraces the racemic forms and the isolated optical isomers.
  • the racemic forms can be resolved by physical methods, such as, e.g., fractional crystallization, separation or crystallization of diastereomeric derivatives or separation by chiral column chromatography.
  • the individual optical isomers can be obtained from the racemates using conventional methods, such as, e.g., salt formation with an optically active acid followed by crystallization.
  • the present invention also embraces all possible tautomers of the compounds provided herein, including the compounds of formula (I).
  • Pharmaceutically acceptable prodrugs of the compounds according to the present invention are derivatives which have chemically or metabolically cleavable groups and become, by solvolysis or under physiological conditions, the compounds of the invention which are pharmaceutically active in vivo.
  • Prodrugs of compounds according to the the present invention may be formed in a conventional manner with a functional group of the compounds such as, e.g., with an amino, hydroxy or carboxy group.
  • the prodrug derivative form often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, Bundgaard, H., Design of Prodrugs, pp.
  • Prodrugs include acid derivatives well known to the person skilled in the art, such as, for example, esters prepared by reaction of the parent acidic compound with a suitable alcohol, or amides prepared by reaction of the parent acid compound with a suitable amine.
  • esters prepared by reaction of the parent acidic compound with a suitable alcohol
  • amides prepared by reaction of the parent acid compound with a suitable amine.
  • a compound of the present invention has a carboxyl group
  • an ester derivative prepared by reacting the carboxyl group with a suitable alcohol or an amide derivative prepared by reacting the carboxyl group with a suitable amine is exemplified as a prodrug.
  • An especially preferred ester derivative as a prodrug is methylester, ethylester, n- propylester, isopropylester, n-butylester, isobutylester, tert-butylester, morpholinoethylester, ⁇ , ⁇ -diethyiglycolamidoester or a-acetoxyethylester.
  • a compound of the present invention has a hydroxy group
  • an acyloxy derivative prepared by reacting the hydroxyl group with a suitable acylhalide or a suitable acid anhydride is exemplified as a prodrug.
  • an amide derivative prepared by reacting the amino group with a suitable acid halide or a suitable mixed anhydride is exemplified as a prodrug.
  • the compounds described herein may be administered as compounds per se or may be formulated as medicaments.
  • the medicaments/pharmaceutical compositions may comprise one or more pharmaceutically acceptable excipients, such as carriers, diluents, fillers, disintegrants, lubricating agents, binders, colorants, pigments, stabilizers, preservatives, antioxidants, or solubility enhancers.
  • the pharmaceutical compositions may comprise one or more solubility enhancers, such as, e.g., poly(ethylene glycol), including poly(ethylene glycol) having a molecular weight in the range of about 200 to about 5,000 Da, ethylene glycol, propylene glycol, non-ionic surfactants, tyloxapol, polysorbate 80, macrogol-15-hydroxystearate, phospholipids, lecithin, dimyristoyl phosphatidylcholine, dipalmitoyl phosphatidylcholine, distearoyl phosphatidylcholine, cyclodextrins, hydroxyethyl-p-cyclodextrin, hydroxypropyl- ⁇ - cyclodextrin, hydroxyethyl-y-cyclodextrin, hydroxypropyl-v-cyclodextrin, dihydroxypropyl- ⁇ - cyclodextrin, glucosyl-a-cyclodextrin,
  • the pharmaceutical compositions can be formulated by techniques known to the person skilled in the art, such as the techniques published in Remington's Pharmaceutical Sciences, 20 th Edition.
  • the pharmaceutical compositions can be formulated as dosage forms for oral, parenteral, such as intramuscular, intravenous, subcutaneous, intradermal, intraarterial, intracardial, rectal, nasal, topical, aerosol or vaginal administration.
  • Dosage forms for oral administration include coated and uncoated tablets, soft gelatin capsules, hard gelatin capsules, lozenges, troches, solutions, emulsions, suspensions, syrups, elixirs, powders and granules for reconstitution, dispersible powders and granules, medicated gums, chewing tablets and effervescent tablets.
  • Dosage forms for parenteral administration include solutions, emulsions, suspensions, dispersions and powders and granules for reconstitution. Emulsions are a preferred dosage form for parenteral administration.
  • Dosage forms for rectal and vaginal administration include suppositories and ovula.
  • Dosage forms for nasal administration can be administered via inhalation and insufflation, for example by a metered inhaler.
  • Dosage forms for topical administration include creams, gels, ointments, salves, patches and transdermal delivery systems.
  • the compounds according to the invention in particular the compounds of formula (I), or the above described pharmaceutical compositions comprising one or more compounds of formula (I) may be administered to a subject by any convenient route of administration, whether systemically/peripherally or at the site of desired action, including but not limited to one or more of: oral (e.g., as a tablet, capsule, or as an ingestible solution), topical (e.g., transdermal, intranasal, ocular, buccal, and sublingual), parenteral (e.g., using injection techniques or infusion techniques, and including, for example, by injection, e.g., subcutaneous, intradermal, intramuscular, intravenous, intraarterial, intracardiac, intrathecal, intraspinal, intracapsular, subcapsular, intraorbital, intraperitoneal, intratracheal, subcuticular, intraarticular, subarachnoid, or intrasternal by, e.g., implant of a depot, for example, subcutaneously or
  • examples of such administration include one or more of: intravenously, intraarterially, intraperitoneal ⁇ , intrathecally, intraventricular ⁇ , intraurethrally, intrasternally, intracardially, intracranially, intramuscularly or subcutaneously administering the compounds or pharmaceutical compositions, and/or by using infusion techniques.
  • parenteral administration the compounds are best used in the form of a sterile aqueous solution which may contain other substances, for example, enough salts or glucose to make the solution isotonic with blood.
  • the aqueous solutions should be suitably buffered (preferably to a pH of from 3 to 9), if necessary.
  • the preparation of suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well known to those skilled in the art.
  • Said compounds or pharmaceutical compositions can also be administered orally in the form of tablets, capsules, ovules, elixirs, solutions or suspensions, which may contain flavoring or coloring agents, for immediate-, delayed-, modified-, sustained-, pulsed- or controlled-release applications.
  • the tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycolate, croscarmellose sodium and certain complex silicates, and granulation binders such as polyvinylpyrrolidone, hydroxypropylmethylcellulose (HPMC), hydroxypropylcellulose (HPC), sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, stearic acid, glyceryl behenate and talc may be included. Solid compositions of a similar type may also be employed as fillers in gelatin capsules.
  • excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine
  • disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glyco
  • Preferred excipients in this regard include lactose, starch, a cellulose, or high molecular weight polyethylene glycols.
  • the agent may be combined with various sweetening or flavoring agents, coloring matter or dyes, with emulsifying and/or suspending agents and with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof.
  • said compounds or pharmaceutical compositions can be administered in the form of a suppository or pessary, or it may be applied topically In the form of a gel, hydrogel, lotion, solution, cream, ointment or dusting powder.
  • the compounds of the present invention may also be dermally or transdermally administered, for example, by the use of a skin patch.
  • sustained-release compositions include semi-permeable polymer matrices in the form of shaped articles, e.g., films, or microcapsules.
  • Sustained-release matrices include, e.g., polylactides (U.S. Pat. No. 3,773,919), copolymers of L-glutamic acid and gamma-ethyl-L-glutamate (Sidman, U. et al., Biopolymers 22:547-556 (1983)), poly(2-hydroxyethyl methacrylate) (R. Langer et al., J. Biomed. Mater. Res.
  • Sustained-release pharmaceutical compositions also include liposomally entrapped compounds.
  • Liposomes containing a compound of the present invention can be prepared by methods known in the art, such as, e.g., the methods described in any one of: DE3218121 ; Epstein et al., Proc. Natl. Acad. Sci. (USA) 82:3688-3692 (1985); Hwang et al., Proc. Natl. Acad. Sci.
  • Said compounds or pharmaceutical compositions may also be administered by the pulmonary route, rectal routes, or the ocular route.
  • they can be formulated as micronized suspensions in isotonic, pH adjusted, sterile saline, or, preferably, as solutions in isotonic, pH adjusted, sterile saline, optionally in combination with a preservative such as a benzalkonium chloride.
  • they may be formulated in an ointment such as petrolatum.
  • dry powder formulations of the compounds of the present invention including the compounds of formula (I), for pulmonary administration, particularly inhalation.
  • Such dry powders may be prepared by spray drying under conditions which result in a substantially amorphous glassy or a substantially crystalline bioactive powder.
  • dry powders of the compounds of the present invention can be made according to the emulsification/spray drying process disclosed in WO 99/16419 or WO 01/85136. Spray drying of solution formulations of the compounds of the present invention is carried out, for example, as described generally in the "Spray Drying Handbook", 5th ed., K. Masters, John Wiley & Sons, Inc. , NY, NY (1991 ), and in WO 97/41833 or WO 03/05341 1 .
  • said compounds or pharmaceutical compositions can be formulated as a suitable ointment containing the active compound suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, emulsifying wax and water.
  • they can be formulated as a suitable lotion or cream, suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, a polyethylene glycol, liquid paraffin, polysorbate 60, cetyl esters wax, 2-octyldodecanol, benzyl alcohol and water.
  • a proposed, yet non-limiting dose of the compounds according to the invention, particularly the compounds of formula (I), for oral administration to a human may be 0.05 to 2500 mg, preferably 0.1 mg to 1000 mg, of the active ingredient per unit dose.
  • the unit dose may be administered, for example, 1 to 3 times per day.
  • the unit dose may also be administered 1 to 7 times per week, e.g., with not more than one administration per day. It will be appreciated that it may be necessary to make routine variations to the dosage depending on the age and weight of the patient/subject as well as the severity of the condition to be treated. The precise dose and also the route of administration will ultimately be at the discretion of the attendant physician or veterinarian.
  • the subject or patient may be an animal (e.g., a non-human animal), a vertebrate animal, a mammal, a rodent (e.g., a guinea pig, a hamster, a rat, a mouse), a murine (e.g., a mouse), a canine (e.g., a dog), a feline (e.g., a cat), a porcine (e.g., a pig), an equine (e.g., a horse), a primate, a simian (e.g., a monkey or ape), a monkey (e.g., a marmoset, a baboon), an ape (e.g., a gorilla, chimpanzee, orang-utan, gibbon), or a human.
  • an animal e.g., a non-human animal
  • a vertebrate animal e.g.,
  • Non-limiting examples of agronomically important animals are sheep, cattle and pigs, while, for example, cats and dogs may be considered as economically important animals.
  • the subject/patient is a mammal; more preferably, the subject/patient is a human or a non-human mammal (such as, e.g., a guinea pig, a hamster, a rat, a mouse, a rabbit, a dog, a cat, a horse, a monkey, an ape, a marmoset, a baboon, a gorilla, a chimpanzee, an orang-utan, a gibbon, a sheep, cattle, or a pig); even more preferably, the subject/patient is a human.
  • a non-human mammal such as, e.g., a guinea pig, a hamster, a rat, a mouse, a rabbit, a dog, a cat, a horse
  • Treatment of a disorder or disease is well known in the art.
  • Treatment of a disorder or disease implies that a disorder or disease is suspected or has been diagnosed in a patient/subject.
  • a patient/subject suspected of suffering from a disorder or disease typically shows specific clinical and/or pathological symptoms which a skilled person can easily attribute to a specific pathological condition (i.e., diagnose a disorder or disease).
  • Treatment of a disorder or disease may, for example, lead to a halt in the progression of the disorder or disease (e.g., no deterioration of symptoms) or a delay in the progression of the disorder or disease (in case the halt in progression is of a transient nature only).
  • Treatment of a disorder or disease may also lead to a partial response (e.g., amelioration of symptoms) or complete response (e.g., disappearance of symptoms) of the subject/patient suffering from the disorder or disease.
  • “Amelioration” of a disorder or disease may, for example, lead to a halt in the progression of the disorder or disease or a delay in the progression of the disorder or disease. Such a partial or complete response may be followed by a relapse. It is to be understood that a subject/patient may experience a broad range of responses to a treatment (e.g., the exemplary responses as described herein above).
  • Treatment of a disorder or disease may, inter alia, comprise curative treatment (preferably leading to a complete response and eventually to healing of the disorder or disease) and palliative treatment (including symptomatic relief).
  • a patient/subject suspected of being prone to suffer from a disorder or disease as defined herein may, in particular, benefit from a prevention of the disorder or disease.
  • the subject/patient may have a susceptibility or predisposition for a disorder or disease, including but not limited to hereditary predisposition.
  • Such a predisposition can be determined by standard assays, using, for example, genetic markers or phenotypic indicators. It is to be understood that a disorder or disease to be prevented in accordance with the present invention has not been diagnosed or cannot be diagnosed in the patient/subject (for example, the patient/subject does not show any clinical or pathological symptoms).
  • prevention comprises the use of compounds of the present invention before any clinical and/or pathological symptoms are diagnosed or determined or can be diagnosed or determined by the attending physician.
  • a number of documents including patent applications, scientific literature and manufacturers' manuals are cited. The disclosure of these documents, while not considered relevant for the patentability of this invention, is herewith incorporated by reference in its entirety. More specifically, all referenced documents are incorporated by reference to the same extent as if each individual document was specifically and individually indicated to be incorporated by reference. The present invention particularly relates to the following items: 1.
  • n is an integer of 0 to 4, and each R 1 is independently selected from C 2 . 4 alkyl, C 2 . 4 alkenyl, C 2 . 4 alkynyl, -OH, -0(d. 4 alkyl), -0(d. 4 alkyl)-OH, -0(C 1-4 alkyl)-0(C 1-4 alkyl), -SH, -S(C 1-4 alkyl), -S(C V4 alkyl)-SH, or -S(d. 4 alkyl)-S(C 1-4 alkyl), -NH 2 , -NH(C 1-4 alkyl), -N(C,. 4 alkyl)(C 1-4 alkyl), halogen, -CF 3 , or -CN;
  • n 2, 3 or 4
  • two groups R 1 attached to adjacent carbon atoms are mutually linked to form a group -0-CH 2 -0-, -0-CH 2 -CH 2 -0- or -0-CH 2 -CH 2 -CH 2 -0-, while the further group(s) R 1 , if present, is/are independently selected from C 1 .4 alkyl, C 2 . 4 alkenyl, C 2 . 4 alkynyl, -OH, -0(C -4 alkyl), -0(d_ 4 alkyl)-OH, -0(d. 4 alkyl)-0(d.
  • R 2 is selected from hydrogen, C 1-4 alkyl, C 2 . 4 alkenyl, C 2 . 4 alkynyl, -OH, -0(d. 4 alkyl), -SH, -S(d. 4 alkyl), -NH 2 , -NH(d. 4 alkyl), -N(d. 4 alkyl)(d. 4 alkyl), halogen, -CF 3 , or -CN;
  • R 3 is selected from -NH 2 , -NH(d. 4 alkyl), -N(C 1-4 alkyl)(C 1-4 alkyl), -OH, -0(Ci. 4 alkyl), -SH, -S(d_ 4 alkyl), or hydrogen;
  • X is selected from S, O, N(H), or N(dminister 4 alkyl);
  • L is -(CH 2 )i-4-, wherein one -CH 2 - unit comprised in said -(CH 2 )i-4- is replaced by a group selected from -CO-NH-, -CO-N(d).
  • A is aryl or heteroaryl, wherein said aryl or said heteroaryl is optionally substituted with one or more groups independently selected from d- 4 alkyl, C 2 .4 alkenyl, C 2 . 4 alkynyl, -OH, -0(C 1-4 alkyl), -SH, -S(C 1-4 alkyl), -NH 2 , -NH(C 1-4 alkyl), -N(d.
  • R 4 is selected from -CO-(C 1-4 alkyl), -CHO, -0(d- 4 alkyl), -OH, -0-CO-(C 1-4 alkyl), -0-CO-0(C 1-4 alkyl), -CO-0(d. 4 alkyl), -COOH, -CO-NH 2 , -CO-NH-(d. 4 alkyl), -CO-N(d_ 4 alkyl)(d- 4 alkyl), -0-CO-NH 2 , -0-CO-NH-(d.
  • n is an integer of 1 to 4, one group R 1 is selected from -0(d. 4 alkyl), -OH, -0(C 1-4 alkyl)-OH, or -0(C 1-4 alkyl)-0(C 1-4 alkyl), and the remaining group(s) R 1 , if present, is/are independently selected from C 2 - 4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, -OH, -0(C 1-4 alkyl), -0(C 1-4 alkyl)-OH, -0(C 1-4 alkyl)-0(C 1 . 4 alkyl), -SH, -S(d.
  • n 2, 3 or 4
  • two groups R 1 attached to adjacent carbon atoms are mutually linked to form a group -0-CH 2 -0-, -0-CH 2 -CH 2 -0- or -0-CH 2 -CH 2 -CH 2 -0-
  • the further group(s) R 1 if present, is/are independently selected from C 1-4 alkyl, C 2 _ 4 alkenyl, C 2-4 alkynyl, -OH, -0(C 1-4 alkyl), -0(d_ 4 alkyl)-OH, -0(d.
  • A is phenyl, 1 ,3-thiazol-2-yl or 1 ,3-benzothiazol-2-yl, wherein said phenyl, said 1 ,3-thiazol-2-yl or said 1 ,3-benzothiazol-2-yl is optionally substituted with one or more groups independently selected from C -4 alkyl, C 2 _ 4 alkenyl, C 2 _ 4 alkynyl, -OH, -0(d. alkyl), -SH, -S(d_ 4 alkyl), -NH 2 , -NH(C 1-4 alkyl), -N(C,. 4 alkyl)(d.
  • R 4 is selected from -CO-(Ci_4 alkyl), -CHO, -0(C 1-4 alkyl), -OH, -CO-0(C 1-4 alkyl), -COOH, C 1-4 alkyl, -(C 1-4 alkyl)-CO-(d.
  • a pharmaceutical composition comprising a compound as defined in any of Items 1 to 10 or a pharmaceutically acceptable salt, solvate or prodrug thereof, and a pharmaceutically acceptable excipient, for use in the treatment or prevention of a cardiovascular disorder, cardiac hypertrophy, heart failure, anxiety, pain, chronic pain, migraine, an allergy, an inflammatory disorder, an autoimmune disorder, diabetes, diabetic complications, diabetic retinopathy, diabetic nephropathy, diabetic cardiomyopathy, diabetic neuropathy, cancer, metastatic cancer, drug-resistant cancer, stomach cancer, lung cancer, thyroid cancer, colon cancer, breast cancer, bipolar disorder, stroke, alopecia, or alcoholism.
  • a pharmaceutical composition comprising a compound as defined in item 16 or a pharmaceutically acceptable salt, solvate or prodrug thereof, and a pharmaceutically acceptable excipient.
  • FIG. 1 Structure-based modeling of the mimic of the PKCe protein fragment EAVSLKPT (see Example 1 ).
  • A X-ray structure of PKCe (PDB 1 gmi). The protein backbone is illustrated as cartoon, while the EAVSLKPT is highlighted as sticks in a light grey box.
  • B Extracted protein fragment EAVSLKPT in its conformation observed in the X-ray structure. Hydrogen bond donor and acceptor features (dark grey spheres) as well as the hydrophobic interaction area (light grey sphere) are indicated for the pharmacophore model that retrieved compound 1 c (PKCe16) as a hit.
  • FIG. 2 Effects of compounds 1 a (PKCe2138) and 1 b (PKCe141 ) in vitro and in vivo (see Examples 2 and 3).
  • Compound 1 b (referred to as “compound 141 ") prevents the in vitro interaction of ⁇ with RACK2 in a dose-dependent manner.
  • Compound 1 b (PKCe141 ) inhibits the phosphorylation of Elk-1 in PathDetect HeLa HLR cells. Luciferase activity following activation of Elk-1 is shown. Data shown are the means (+/-SD) of 3 independent experiments.
  • C Compound 1 b (PKCe141 ) does not prevent the in vitro interaction between ⁇ and RACK1 .
  • E Compound 1 a (PKCe2138) prevents RACK2-binding to PKCs in vitro in a dose-dependent manner.
  • F Inhibition of RACK2-binding to PKC in vitro by EAVSLKPT which was tagged with seven arginines. A scrambled octapeptide tagged with seven arginines was used as control.
  • FIG. 3 PKCe induces phosphorylation of Elk-1 .
  • A In HeLa cells, a doxycycline-inducible constitutively active PKCe (Xuan et al., 2005) leads to phosphorylation of Elk-1 . The cells were left untreated or induced with doxycycline (2 pg/ml) for 24 hours. Additional stimulation with TPA (50 nM) was performed for 10 minutes. Phosphorylation of Elk-1 was detected with a phospho-specific antibody against the Ser383 residue of Elk-1 . GAPDH was used as loading control.
  • B Densitometric analysis of Western blots.
  • FIG. 4 Effect of compound 1 b (PKCe141 ) on PKCe and PKC5 translocation (see Example 4).
  • IGF!- R and GAPDH were used as loading controls for the membrane and the cytosolic fraction, respectively.
  • the means of three independent experiments were scanned and shown in a graph (+/-SD).
  • FIG. 5 Effect of compound 1 b (PKCe141 ) on PKC isozymes. 50 ⁇ compound 1 b were used in a PKC assay as described in Examples 4 and 6.
  • FIG. 6 Cell proliferation following treatment with compound 1 b (PKCe141 ). Cell proliferation was determined as described in Example 5. The means (+/-SD) of three independent experiments, in which three samples were taken within each experiment, are shown.
  • Figure 7 Effect of compound 1 b (PKCe141 ) on PKCe translocation by immunofluorescence. PC-3 cells were employed for these experiments. TPA induces translocation of PKCe to the plasma membrane. Compound 1 b inhibits this TPA-induced translocation of PKCe. Experiments 1 , 2 and 3 are three independent experiments.
  • FIG 8 PKCe-induced migration of HeLa cells into a gap. The expression of constitutively active PKCe was induced by doxycycline for 24 hours after a scratch was made into monolayer cells with a pipette tip.
  • Figure 9 Inhibition of angiogenesis in vitro (see Example 7).
  • Figure 10 1 H-NMR spectrum (A) and mass spectrum (B) of compound 1a (PKCe2138).
  • a pharmacophore model was derived from the PKCe protein fragment EAVSLKPT using its conformation observed in the crystal structure of PDB entry 1 gmi (Figs. 1A and 1 B). Catalyst (version 4.1 1 , Accelrys inc., San Diego, CA, USA) was employed for pharmacophore modeling and subsequent screening of commercial molecular libraries.
  • the pharmacophore model consists of three hydrogen bond donor/acceptor features and one hydrophobic feature. The latter attempts to resemble the hydrophobic area presented to RACK2 by Val16, while the hydrogen bond donor/acceptor features characterize the side chain properties of Ser17, Lys19, and Glu14.
  • PKCe141 (N-(3-acetylphenyl)- 9-amino-2,3-dihydro-1 ,4-dioxino[2,3-g]thieno[2,3-b]quinoline-8-carboxamide; Asinex ID: ASN 05545158; CAS 602293-00-5) was identified as the strongest disruptor/inhibitor of the PKCe/RACK2 interaction.
  • This screening process was followed by custom synthesis and testing of a series of further compounds related to compound 1 b (PKCe2019 to PKCe2145).
  • PKCe/RACK2 in vitro binding assay Recombinant RACK2 tagged with maltose-binding protein (RACK2-MBP) was purified on columns with amylose resin (New England Biolabs) as described by the manufacturer. The recombinant protein was analyzed by Coomassie Blue staining and Western blotting after SDS-PAGE. Aliquots were stored in liquid nitrogen. The interaction between PKCe and RACK2 was measured using an ELISA-based assay.
  • 96-well EIA/RIA high binding plates (Costar) were coated with 100 ng recombinant PKCe (ProQinase) in buffer A (20 mM Tris-HCI/100 m NaCI, pH 7.5) at 4°C on a shaker with gentle agitation overnight. The plate was washed twice with 225 ⁇ l/well buffer A. After blocking of unspecific binding sites with 225 ⁇ sterile-filtered 3 % bovine serum albumin (BSA; Sigma) in buffer A at room temperature for 3 h, the plate was washed twice with 225 ⁇ of this buffer.
  • PKCe ProQinase
  • buffer A 20 mM Tris-HCI/100 m NaCI, pH 7.5
  • BSA bovine serum albumin
  • PKCe was left untreated or activated by addition of 60 pg/ml phosphatidylserine (Sigma) and 100 nM TPA (Sigma) in a volume of 50 ⁇ buffer A for 10 min at 30°C.
  • Recombinant purified RACK2-MBP was either left untreated or incubated with EAVSLKPT-R7 or compound 1 b (PKCe141 ) at room temperature for 30 min in a final volume of 50 ⁇ buffer A.
  • 500 ng RACK2-MBP was added to untreated or activated PKCe for 1 hour at room temperature for binding. The plate was washed twice with 225 ⁇ buffer A.
  • 100 ⁇ RACK2-specific rabbit anti-RACK2 polyclonal antibody (Prof. F.
  • PKCpil/RACK1 in vitro binding assay This assay was similar to the PKCE/RACK2 binding assay described above. 6-his-tagged RACK1 was cloned into a pET-30a(+) vector (Novagen) and purified with Ni-NTA Agarose (Qiagen). Final elution was performed with 500 nM imidazole in elution buffer (20 mM Tris-HCI/300 mM NaCI, 20% glycerol, pH 7.5). The integrity of purified recombinant protein was analyzed by Coomassie Blue staining and Western blotting.
  • RACK1 200 ng RACK1 was coated onto 96-well EIA/RIA high binding plates (Costar) at 4°C on a shaker with gentle agitation overnight.
  • Compound 1 b (PKCe141 ) was added at room temperature for 30 min.
  • the RACK1 - coated plates with and without compound 1 b were incubated for 1 h with activated ⁇ .
  • the peptide EAVSLKPT-RRRRRRR was used as a control. Seven arginines were added to EAVSLKPT to increase internalization of the peptide into intact cells in in vivo experiments. ICso for the inhibition of the PKCe/RACK2 interaction by EAVSLKPT-RRRRRRR in this assay was 1.02 ⁇ (Fig. 2F). It has been shown previously that ⁇ interacts with the adaptor protein RACK1 (Ron et al., 1999). Therefore, it was investigated whether compound 1 b can also prevent the PKCpil/RACK1 interaction. As shown in Fig. 2C, compound 1 b does not prevent the PKCpil/RACK1 interaction, indicating specificity of this compound for the PKCe/RACK2 interaction.
  • compounds of formula (I) such as, e.g., compounds 1 a and 1 b, selectively inhibit PKCe signaling by preventing the interaction between PKCe and its adaptor protein RACK2.
  • the compounds of the invention are therefore suitable as therapeutic agents for the treatment or prevention of diseases/disorders related to PKCe signaling, as also described herein above.
  • Table 1 Inhibition of PKCe signaling.
  • the inhibition of the PKCE/RACK2 interaction is expressed as a percentage of untreated control. Accordingly, a value of, e.g., 37.7% denotes an inhibition of the PKCE/RACK2 interaction to 37.7% of the untreated control (which is set to 100%).
  • Compounds having a percentage inhibition of 100% or greater were not found to inhibit PKCe signaling in this assay.
  • PKCE is situated in the signal transduction cascade upstream of Raf-1 (Xuan et al., 2005).
  • active PKCE leads to phosphorylation of the transcription factor Elk-1 (see Fig. 3).
  • compound 1 b PLCe141
  • HLR PathDetect HeLa Luciferase
  • HeLa HLR and PC-3 cells were seeded at -10,000 cells per well in 96-well plates. After 4 h various concentrations of compound 1 b (PKCe141 ) were added and left for 72 h. Cell proliferation was determined by the SRB-assay (Skehan et al., 1990). Elk-1 phosphorylation: Elk-1 phosphorylation was determined with the PathDetect System (Agilent). PathDetect HeLa HLR-ELK-1 cells contain a luciferase reporter cassette and express a unique, stably integrated, irans-acting fusion protein.
  • the fusion protein consists of the activation domain of the Elk-1 transcriptional activator (Rao et al., 1989; Price et al., 1995; Marais et al. , 1993), that is fused to the yeast GAL4 DBD (residues 1-147).
  • the transcriptional activator domain of Elk-1 is activated.
  • 200,000 PathDetect HeLa HLR-Elk-1 cells per well were seeded in a 6-well plate and grown for 24 hours. Cells were washed with phosphate buffered saline and starved for 16 hours in starvation medium (DMEM-containing 0.5% fetal bovine serum and 1 % glutamine). Compounds were added in DMEM for 30 min.
  • the membranes were incubated with rabbit polyclonal IgG antibodies for detection of PKCe (Santa Cruz Biotechnology, dilution 1 :2,000), of PKCe phosphoSer 729 (Millipore, dilution 1 :1 ,000) and of PKC5 (Santa Cruz; dilution 1 :1 ,000).
  • rabbit polyclonal IgG antibodies for detection of PKCe (Santa Cruz Biotechnology, dilution 1 :2,000), of PKCe phosphoSer 729 (Millipore, dilution 1 :1 ,000) and of PKC5 (Santa Cruz; dilution 1 :1 ,000).
  • an IGFI- ⁇ rabbit polyclonal IgG antibody Santa Cruz Biotechnology; dilution 1 : 1 ,000
  • secondary antibodies peroxidase-conjugated AffiniPure Goat Anti-Rabbit IgG Jackson Immuno Research Laboratories, dilution
  • GAPDH (Chemicon; 1 : 10,000) was used as loading control for the cytosolic fraction.
  • a peroxidase-conjugated secondary antibody (AffiniPure Goat Anti-Mouse IgG; Jackson Immuno Research Laboratories, dilution 1 :20,000) was employed for detection.
  • In vitro PKC assay 150 ng of recombinant purified PKC isozyme (see Fig.
  • kinase assay-mix 50 ⁇ of the kinase assay-mix are added onto phosphocellulose sheets (Whatman) in a 6-well plate. The sheets were washed 3 times with 1.5% H 3 P0 4 and twice with distilled water. The phosphocellulose sheets are transferred to scintillation vials, 4 ml of scintillation fluid (Ultima Gold, Perkin-Elmer) were added and counted in a ⁇ -counter.
  • scintillation vials 4 ml of scintillation fluid (Ultima Gold, Perkin-Elmer) were added and counted in a ⁇ -counter.
  • the effect of compound 1 b (PKCe141) on the different PKC isozymes is shown in Fig. 5.
  • compound 1 b partially inhibits the cellular translocation of activated and phosphorylated PKCe to the membrane fraction.
  • PKC5 shows a high degree of homology with PKCe. Therefore, the influence of compound 1 b on PKC5 was investigated. As shown in Fig. 4, compound 1 b does not decrease PKC5 in the membrane. A similar result was obtained with immunocytochemistry (Fig. 7). Short-term treatment with TPA led to an increase of PKCe in the plasma membrane. Compound 1 b prevented the TPA-induced PKCe translocation to the plasma membrane, as shown in Fig. 7.
  • compound 1 b exhibits the features of an inhibitor of PKCe signaling in vitro and also in intact cells. A major question is whether such an inhibitor affects cell proliferation or, in other words, whether it is toxic. Therefore, compound 1 b was tested for inhibition of cell proliferation in HeLa-HLR and PC-3 cells. These cells were employed for Elk-1 phosphorylation and for the PKCe translocation experiments described above.
  • PC-3 cells were grown on glass coverslips coated with poly-L-lysine (Sigma). After treatment with compound 1 b (PKCe141 ) for 30 min and with 100 nM TPA for 5 min, the cells were rinsed twice with phosphate buffered saline (PBS) and fixed with filter sterilized 4% (w/v) paraformaldehyde/4% sucrose (w/v) (both from Sigma) in PBS at room temperature for 10 min. Subsequently the cells were washed three times with PBS and permeabilized with 0.2% Triton X-100/0.2% IgG-free BSA in PBS at room temperature for 10 min.
  • PBS phosphate buffered saline
  • sucrose w/v
  • the dish was washed with PBS to remove the cells and further incubated with 2 doxycyclin and 25 ⁇ compound 1 b (PKCe141 ) for 24 hours. Controls were left either untreated or were incubated with 2 pg/ml doxycyclin and DMSO. Migration into the scratch was observed with an Olympus microscope.
  • In vitro PKC assay 150 ng of recombinant purified PKC isozyme (see Fig. 5) were combined with 10 ⁇ 10 x kinase assay buffer (200 mM Tris-HCI , pH 7.5, 200 mM MgCI 2 ), phosphatidylserine (final 10 ⁇ ), TPA (final 1 ⁇ ) and substrate peptide RFARKGSLRQKNV (Alexis) (final 50 ⁇ ) in a volume of 100 ⁇ , 90 ⁇ l/we 11 were pipetted in a 96-well plate and incubated for 1 min at 30°C.
  • 10 ⁇ 10 x kinase assay buffer 200 mM Tris-HCI , pH 7.5, 200 mM MgCI 2
  • phosphatidylserine final 10 ⁇
  • TPA final 1 ⁇
  • substrate peptide RFARKGSLRQKNV Alexis
  • Table 2 Profile of kinase inhibition by 25 ⁇ compound 1 b (PKCe141 ). 109 different protein kinases were tested for their inhibition by compound 1 b (PKCe141 ). The five kinases inhibited most are shown in the table. All other kinases were affected less. Screening was performed by the National Centre for Protein Kinase Profiling, Division of Signal Transduction Therapy, University of Dundee. The data is portrayed as mean % activity and standard deviation (SD) of assay duplicates.
  • SD standard deviation
  • Compound 1 b (PKCe141 ), however, affected these kinases less than the PKCE/RACK2 interaction. As shown in Fig. 2A, 25 ⁇ compound 1 b (PKCe141 ) inhibited the PKCE/RACK2 interaction to approximately 10% of untreated controls, whereas 25 ⁇ of compound 1 b inhibited the most affected kinase RSK2 to 27%, as shown in Table 2 above. All other kinases were less affected. This level of selectivity is quite acceptable.
  • Example 7 Inhibition of angiogenesis Compounds according to the invention were tested for their effect on angiogenesis in vitro in a spheroid sprouting assay and in a chicken egg assay.
  • Spheroid sprouting assay (Korff et al., 1 999): Human umbilical vein endothelial (HUVEC) spheroids where generated overnight in hanging-drop culture consisting of 400 cells in EBM-2 medium, 2% FCS and 20% methylcellulose (Sigma Biochemicals). Spheroids were embedded in collagen type I from rat tail (Becton Dickinson) and stimulated with 50 ng/ml VEGF (Sigma Biochemicals) in the presence or absence of 5 ml solution containing the corresponding test compound (see Fig. 9A). Sprouts were analyzed by inverted transmission-microscopy (Zeiss Axiovert 200 M) and documented by digital imaging (Axiovision Software, Zeiss). The cumulative sprout length (CSL) was analyzed after printing of high quality pictures and counting by two independent blinded observers.
  • HUVEC Human umbilical vein endothelial
  • Chicken egg assay Eggs from hen are incubated at 37°C for 3 days, opened and incubated for further 7 days. On day ten the growth factor VEGF and compound 1 b (PKCe141 ) are added at the concentrations indicated in Fig. 9B. After further incubation for 5 days angiogenesis is observed by microscope.
  • Acetanilide (2) A solution of amine 1 (1 eq) in pyridine was cooled to 5°C and acetanhydride (1 .25 eq) was added dropwise with stirring. The mixture was stirred for 45 min at 60°C and poured into ice-water and stirred for 30 min at 0-10°C. The precipitate was filtered off, washed with water (100 ml) and dried.
  • Chloroquinolinecarbaldehyde (3) in phosphoryl chloride Dimethylformamide (9.13 g, 9.6 ml, 0.125 mol) was cooled to 0°C in a flask equipped with a drying tube and phosphoryl chloride (53.7 g, 32 2 ml, 0.35 mol) was added dropwise with stirring. To this solution was added the acetanilide 2 (0.05 mol) and after 5 min the solution was heated to 75°C for 12- 16 h. The reaction mixture was poured into ice-water (300 ml) and stirred for 30 min at 0- 10°C. The chloroquinolinecarbaldehyde 3 was filtered off, washed with water (100 ml) and dried.
  • the chloroquinolinecarbaldehyde 3 was crystallized with ethylacetate.
  • Hydroxylimine (A) Hydroxylamine hydrochloride (1 .25 eq) was added to the suspension of chloroquinolinecarbaldehyde 3 in ethanol and this mixture was refluxed for 6 h. The hydroxylimine 4 was filtered off, washed with ethanol and dried.
  • reaction mixture was poured into ice-water and stirred for 30 min at 0-10°C.
  • nitrile 5 was filtered off, washed with water and dried.
  • Raw material was purified by flash-chromatography on silica gel with chloroform and hexane (4: 1 ).
  • 2- Thio-3-cyano-quinoline (6) A mixture of 2-chloro-3-cyano-quinoline 5 (1 eq) and thiourea (3 eq) in propanol-1 was heated under reflux for 6 h, then this mixture was cooled and sodium hydroxide (10% 10 eq) was added. The reaction mixture was cooled, acidified with concentrated HCI and the solid product was filtered off and dried.
  • PLCe2024 Thieno[2,3-b]quinoline-2-carboxylanilide (PKCe2024): A mixture of the thieno[2,3- b]quinoline-2-carboxylate 8 (1 eq) and PyBOP (1 .3 eq) in DMF was stirred for 15 min, then appropriate aniline (1 .25 eq), catalytic amount of 4-dimethylaminopyridine (DMAP) and Et 3 N (2 eq) was added. The reaction mixture was stirred overnight at 50°C than it was poured into ice-water and precipitate was filtered off. Raw material was heated under reflux with methanol-DMF (3:2) and filtered off and dried.
  • DMAP 4-dimethylaminopyridine
  • the anilide PKCe2024 was purified by flash- chromatography with CHCI 3 -methanol (9: 1 ).
  • the compounds 1 m and 1 n according to the invention i.e., PKCe2023 and PKCe2024. were prepared in this manner.
  • the compounds PKCe2048 to PKCe2054 were prepared as described in Lapa et ai., 2012.
  • PKCe2053 3-Chloro-N-(7-methoxy-1 H-pyrazolo[3,4-b]quinolin-3-yl)benzamide. Yield 74%.
  • 1 H-NMR, DMSO-D6, ⁇ : 3.94 (3H, s, CH 3 ), 7.1 1 (1 H, dd, J 2.2, 9.2, H-6), 7.29 (1 H, d, J 2.2, H-8), 7.61 (1 H, t, J 8.0, H-5'), 7.71 (1 H, d, J 8.0, H-6'), 8.02 (1 H, d, J 9.2, H-5), 8.07 (1 H, d, J 8.0, H-4'), 8.17 (1 H, s, H-2'), 8.90 (1 H, s, H-4), 1 1.27 (1 H, s, NH), 13.02 (1 H, s, HN-C 0). MW Calc. for C 18 H 13 CIN 4 0 2 352.0727. Found in ESI-ms 353.0708

Abstract

The present invention relates to novel inhibitors of protein kinase C epsilon (PKCε) signaling, including in particular the compounds of formula (I) as described and defined herein, pharmaceutical compositions comprising these inhibitors, and their use in the treatment or prevention of disorders such as, e.g., a cardiovascular disorder, cardiac hypertrophy, heart failure, anxiety, pain, chronic pain, migraine, an allergy, an inflammatory disorder, an autoimmune disorder, diabetes, diabetic complications, diabetic retinopathy, diabetic nephropathy, diabetic cardiomyopathy, diabetic neuropathy, cancer, metastatic cancer, drug-resistant cancer, stomach cancer, lung cancer, thyroid cancer, colon cancer, breast cancer, a neurological disorder, Alzheimer's disease, Parkinson's disease, bipolar disorder, stroke, alopecia, or alcoholism.

Description

Novel inhibitors of protein kinase C epsilon signaling
The present invention relates to novel inhibitors of protein kinase C epsilon (PKCE) signaling, including in particular the compounds of formula (I) as described and defined herein, pharmaceutical compositions comprising these inhibitors, and their use in the treatment or prevention of disorders such as, e.g., a cardiovascular disorder, cardiac hypertrophy, heart failure, anxiety, pain, chronic pain, migraine, an allergy, an inflammatory disorder, an autoimmune disorder, diabetes, diabetic complications, diabetic retinopathy, diabetic nephropathy, diabetic cardiomyopathy, diabetic neuropathy, cancer, metastatic cancer, drug-resistant cancer, stomach cancer, lung cancer, thyroid cancer, colon cancer, breast cancer, a neurological disorder, Alzheimer's disease, Parkinson's disease, bipolar disorder, stroke, alopecia, or alcoholism. Protein kinase C (PKC) is a family of serine/threonine-specific protein kinases. The PKC isozymes can be classified into three groups: i) the conventional isozymes α, βΙ, βΙΙ, and γ; ii) the novel isozymes δ, ε, Θ, and η; and iii) the atypical isozymes λ/ι (mouse/human) and ζ. PKC isozymes seem to play important roles in the activation of signal transduction pathways leading to synaptic transmissions, the activation of ion fluxes, secretion, proliferation, cell cycle control, differentiation and tumorigenesis. Because of their role in a complex network of signal transduction pathways, different isoforms have divergent, sometimes opposite roles within a given biological process. However, no simple, unique function can be assigned to a given PKC isozyme. Modulators of PKC isozymes are described, e.g. , in: Mochly-Rosen et al., 2012; Sanchez-Bautista et al., 2013; Bohler, 2006; US 2009/0124553; and US 6,376,467.
The PKCE isozyme was reported to participate in neoplastic transformation (Gorin et al., 2009), cardiac hypertrophy (Pass et al., 2001 ), protection from ischemic insult (Pass et al., 2001 ; Johnson et al., 1996), nociceptor function (Cesare et al., 1999), macrophage activation (Castrillo et al. , 2001 ), diabetes (Ikeda et al., 2001 ) and in alcohol consumption (Choi et al., 2002). A PKCe isozyme-specific inhibitor would have pharmaceutical potential, e.g., for the intervention in stroke, Alzheimer's disease or pain (Akita, 2008; Shirai et al., 2008).
Typically, kinase inhibitors interact with the ATP-binding site, which is well conserved among different kinase families, and is even more so within a family. This poses a serious hurdle for the development of isozyme-specific inhibitors, as there are approximately 518 kinases encoded by the human genome (Manning et al., 2002). Although several selective kinase inhibitors have been reported initially, later it turned out that they also inhibit other targets. For example, the marketed drug imatinib mesylate (Gleevec/Glivec) was developed as an inhibitor of the oncoprotein Bcr-Abl. However, it has turned out to inhibit also other tyrosine kinases such as Kit and platelet-derived growth factor receptor, as well as non- kinase targets. Similarly, relatively unspecific inhibitors initially intended as PKCp-selective inhibitors, such as ruboxistaurine (Fedorov et al., 2007; Nakamura et al., 2010) and enzastaurine (Chen et al., 2008) are in clinical trials for diabetic retinopathy and cancer, respectively. Rottlerin was described as a specific inhibitor of PKC5. However, also for this compound, additional modes of action have been observed by now (Soltoff, 2007). Due to the high degree of conservation among the ATP-binding sites in PKC isozymes, it is very difficult to develop monospecific inhibitors of a particular PKC isozyme. While monospecificity is not a prerequisite for a successful drug, it would be highly desirable as secondary activities create ambiguity in the interpretation of biochemical, pharmacological, and clinical results.
In the context of the present invention, a different approach was employed to specifically target PKCe signaling. Rather than interacting with the ATP-binding site of PKCE, the signal transduction of PKCE was targeted by preventing the binding of PKCe to its adaptor protein, the PKCe-specific receptor of activated C-kinase 2 (RACK2, β ΟΡ) (Csukai et al., 1997; Schechtman et al., 2001 ). It has been shown that adaptor proteins bind and translocate activated PKC isozymes to subcellular sites in close proximity to their substrates (Mochly- Rosen et al., 1991 ). One example of an inhibitor of such protein-protein interactions is aurothiomalate, which interferes with the interaction between PKCi and its adaptor molecule Par6. The compound blocks oncogenic PKCi signaling and growth of human lung cancer cells (Erdogan et al., 2006). The adaptor protein RACK2 binds to activated PKCe. The peptide EAVSLKPT, corresponding to amino acids 14 to 21 of PKCe, is known to selectively inhibit the translocation of PKCe by binding to RACK2, but not that of other PKC isozymes (Johnson et al. , 1996; Gray et al., 1997). It has also been shown that the PKCe antagonist EAVSLKPT inhibits protection from hypoxia-induced cell death of cardiac myocytes (Gray et al. , 1997).
It is thus an object of the present invention to provide a small molecule peptidomimetic of EAVSLKPT that prevents the PKCe/RACK2 interaction and, hence, selectively inhibits PKC signaling. Small molecule inhibitors of protein-protein interactions have previously been shown to be useful for pharmacological purposes (Beeley, 2000; Arkin et al., 2004). It is also an object of the invention to provide an isozyme-specific cell-permeable small- molecule inhibitor of PKCs signal transduction, which would be highly useful in therapy, particularly for the treatment or prevention of cancer, stroke, Alzheimer's disease, pain, cardiac hypertrophy, and alcoholism (Shirai et al. , 2008), and also as a tool for further research on ΡΚΟε. As a solution to the above-mentioned problems, the present invention provides the compounds of formula (I) as described herein below and in the claims. Structurally related compounds have also been disclosed in: Sanam et al. , 2010; US 201 1 /0077250; WO 2013/033037; CN 102757447 A; Pillai et al., 2010; US 2013/0129677; RU 2371444 C1 ; WO 92/03427; and CAS registry nos. 338433-13-9 and 333330-1 1 -3. The barbituric acid derivative BAS 02104951 has furthermore been disclosed as an inhibitor of PKCe signal transduction in Gruber et al., 201 1 . Accordingly, the present invention provides a compound of the following formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof, for use as a medicament:
Figure imgf000004_0001
(I)
In formula (I), n is an integer of 0 to 4 (i.e. , 0, 1 , 2, 3 or 4), and each R1 is independently selected from C1-4 alkyl, C24 alkenyl, C2.4 alkynyl, -OH, -0(C1-4 alkyl), -0(C-,.4 alkyl)-OH, -0(d_4 alkyl)-0(C1-4 alkyl), -SH, -S(C1-4 alkyl), -S(C1-4 alkyl)-SH, or -S(C,.4 alkyl)-S(d.4 alkyl), -NH2, -NH(C1-4 alkyl), -N(d.4 alkylXC^ alkyl), halogen, -CF3, or -CN. Alternatively, n is 2, 3 or 4, and two groups R1 attached to adjacent carbon atoms are mutually linked to form a group -0-CH2-0-, -0-CH2-CH2-0- or -0-CH2-CH2-CH2-0-, while the further group(s) R\ if present (i.e. , if n is 3 or 4), is/are independently selected from C,.4 alkyl, C2.4 alkenyl, C2-4 aikynyl, -OH, -0(C1-4 alkyl), -0(C1-4 alkyl)-OH, -0(C1-4 aIkyl)-0(C1-4 alkyl), -SH, -S(C1-4 alkyl), -S(C1-4 alkyl)-SH, or -S(C1-4 alkyl)-S(C1-4 alkyl), -NH2l -NH(d_4 alkyl), -N(C1-4 alkyl)(C _4 alkyl), halogen, -CF3, or -CN. It is to be understood that, if n is 0, the condensed phenyl ring moiety (to which R1 would be attached) is unsubstituted, i.e. is substituted with hydrogen.
Preferably, n is an integer of 1 to 4 (i.e. , 1 , 2, 3 or 4), one group R is selected from -0(C1 -4 alkyl), -OH, -0(C1-4 alkyl)-OH, or -0(d_4 alky!)-0(d_4 alkyl), and the remaining group(s) R1, if present (i.e., if n is 2, 3 or 4), is/are independently selected from d_4 alkyl, C2-4 alkenyl, C2-4 aikynyl, -OH, -0(Ci_4 alkyl), -0(C1-4 alkyl)-OH, -0(C1 -4 alkyl)-0(d_4 alkyl), -SH, -S(C1-4 alkyl), -S(C1-4 alkyl)-SH, or -S(C1-4 alkyl)-S(C1 -4 alkyl), -NH2, -NH(C1-4 alkyl), -N(C1-4 alkyl)(C1-4 alkyl), halogen, -CF3, or -CN; or, alternatively, n is 2, 3 or 4, and two groups R1 attached to adjacent carbon atoms are mutually linked to form a group -0-CH2-0-, -0-CH2-CH2-0- or -0-CH2-CH2-CH2-0-, while the further group(s) R , if present (i.e., if n is 3 or 4), is/are independently selected from d„4 alkyl, C2_4 alkenyl, C2-4 aikynyl, -OH, -0(C -4 alkyl), -0(d.4 alkyl)-OH, -0(d_4 alkyl)-0(d.4 alkyl), -SH, -S(d_4 alkyl), -S(d.4 alkyl)-SH, or -S(C1-4 alkyl)-S(d.4 alkyl), -NH2, -NH(C1-4 alkyl), -N(C1-4 alkyl)(d_4 alkyl), halogen, -CF3, or -CN. More preferably, n is 1 or 2, one group R1 is selected from -0(d„ alkyl), -OH, -0(Ci.4 alkyl)-OH, or -0(C -4 alkyl)-0(C1-4 alkyl), and the remaining group R , if present (i.e. , if n is 2), is selected from C1-4 alkyl, C2.4 alkenyl, C2.4 aikynyl, -OH, -0(C1-4 alkyl), -SH, -S(C1-4 alkyl), -NH2, -NH(C1-4 alkyl), -N(d.4 aikyl)(C - alkyl), halogen, -CF3, or -CN; or, alternatively, n is 2, and the two groups R1 are attached to adjacent carbon atoms (preferably the same two carbon atoms as in the compounds 1a and 1 b shown below) and are mutually linked to form a group -0-CH2-0-, -0-CH2-CH2-0- or -0-CH2-CH2-CH2-0-.
Even more preferably, n is 1 and R1 is selected from -0(C -4 alkyl), -OH, -0(C1-4 alkyI)-OH, or -0(C1-4 alkyl)-0(d_4 alkyl), wherein it is particularly preferred that R1 is selected from -OCH3 or -OCH2CH3; or, alternatively, n is 2 and the two groups R1 are attached to adjacent carbon atoms (preferably the same two carbon atoms as in the compounds 1 a and 1 b shown below) and are mutually linked to form a group -0-CH2-0- or -0-CH2-CH2-0-.
R2 is selected from hydrogen, C1-4 alkyl, C2.4 alkenyl, C2.4 aikynyl, -OH, -0(C1 -4 alkyl), -SH, -S(d_4 alkyl), -NH2, -NH(C1-4 alkyl), -N(C1-4 alkyl)(d.4 alkyl), halogen, -CF3, or -CN. Preferably, R2 is hydrogen. R3 is selected from -NH2, -NH(C1-4 alkyl), -N(d.4 alkyl)(C1-4 alkyl), -OH, -0(C1-4 alkyl), -SH, -S(d.4 alkyl), or hydrogen. Preferably, R3 is -NH2, -NH(d_4 alkyl), -OH, or -SH. More preferably, R3 is -NH2.
X is selected from S, O, N(H), or N(d.4 alkyl). Preferably, X is S or O. More preferably, X is S.
L is -(CH2)i-4-, wherein one -CH2- unit comprised in said -(CH2) -4- is replaced by a group selected from -CO-NH-, -CO-N(d.4 alkyl)-, -NH-CO-, -N(d_4 alkyl)-CO-, -0-, -CO-, -NH-, -N(d_4 alkyl)-, -S-, -SO-, or -S02-. Preferably, L is -(CH2)1-3-, wherein one -CH2- unit comprised in said -(CH2)1-3- is replaced by a group selected from -CO-NH-, -CO-N(d.4 alkyl)-, -NH-CO-, or -N(d.4 alkyl)-CO-. More preferably, L is directionally selected from -CO- NH-, -CO-N(d.4 alkyl)-, -NH-CO-, or -N(d.4 alkyl)-CO-. Even more preferably, L is -CO-NH- or -CO-N(d_4 alkyl)-, wherein the nitrogen atom comprised in said -CO-NH- or said -CO- N(d-4 alkyl)- is attached to ring A. Most preferably, L is -CO-NH-, wherein the nitrogen atom comprised in said -CO-NH- is attached to ring A.
A is aryl or heteroaryl, wherein said aryl or said heteroaryl is optionally substituted with one or more groups (such as, e.g., one, two, three or four groups; preferably, one or two groups; more preferably, one group) independently selected from d„4 alkyl, C2-4 alkenyl, C2.4 alkynyl, -OH, -0(C1-4 alkyl), -SH, -S(d.4 alkyl), -NH2, -NH(d.4 alkyl), -N(d.4 alkyl)(C1-4 alkyl), halogen, -CF3, or -CN. Said aryl is preferably phenyl or naphthyl, and more preferably said aryl is phenyl. Said heteroaryl is preferably a heteroaryl having 5 or 6 ring atoms, wherein 1 , 2 or 3 of said ring atoms are heteroatoms independently selected from oxygen, sulfur or nitrogen, and the remaining ones of said ring atoms are carbon atoms, and further wherein said heteroaryl having 5 or 6 ring atoms is optionally fused to a benzene ring. More preferably, said heteroaryl is selected from thiazolyl (e.g., 1 ,3-thiazol-2-yl), furanyl, thiophenyl (i.e., thienyl), pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, furazanyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, benzothiazolyl (e.g., 1 ,3-benzothiazol-2-yl), benzofuranyl, benzothiophenyl, benzopyrrolyl, benzoimidazolyl, benzopyrazolyl, benzoxazolyl, benzisoxazolyl, benzofurazanyl, benzopyridinyl, benzopyrimidinyl, benzopyrazinyl, or benzopyridazinyl. Even more preferably, said heteroaryl is thiazolyl (e.g. , 1 ,3-thiazol-2-yl) or benzothiazolyl (e.g. , 1 ,3-benzothiazol-2-yl). Most preferably, said heteroaryl is benzothiazolyl, particularly 1 .3-benzothiazol-2-yl. It is furthermore preferred that A is aryl or heteroaryl, including any of the aforementioned preferred aryl or heteroaryl groups, wherein said aryl or heteroaryl is unsubstituted (i.e., is not substituted with any groups other than R4).
It is preferred that A is phenyl, 1 ,3-thiazol-2-yl or 1 ,3-benzothiazol-2-yl, wherein said phenyl, said 1 ,3-thiazol-2-yl or said 1 ,3-benzothiazol-2-yl is optionally substituted with one or more (e.g., one, two or three) groups independently selected from C1-4 alkyl, C2,4 alkenyl, C24 alkynyl, -OH, -0(d.4 alkyl), -SH, -S(C1-4 alkyl), -NH2, -NH(d_4 alkyl), -N(C1-4 alkyl)(d.4 alkyl), halogen, -CF3, or -CN, preferably selected independently from d.4 alkyl, -OH, -0(d.4 alkyl), -NH2, -NH(C1-4 alkyl), -N(C1-4 alkyl)(C1-4 alkyl), halogen, -CF3, or -CN, more preferably selected independently from C1-4 alkyl, -OH, -0(C1-4 alkyl), or halogen (e.g., -F, -CI, -Br, or -I). It is furthermore preferred that A is phenyl, 1 ,3-thiazo!-2-yl or 1 ,3-benzothiazol-2-yl, wherein said phenyl, said 1 ,3-thiazol-2-yl or said 1 ,3-benzothiazol-2-yl is unsubstituted (i.e., is not substituted with any groups other than R4). More preferably, A is phenyl which is optionally substituted with one or more (e.g., one, two or three) groups independently selected from d„ alkyl, C2-4 alkenyl, C2.4 alkynyl, -OH, -0(0,-4 alkyl), -SH, -S(C1-4 alkyl), -NH2, -NH(C1-4 alkyl), -N(d.4 alkyl)(d_4 alkyl), halogen, -CF3, or -CN, particularly from .4 alkyl, -OH, -0(C1-4 alkyl), -NH2, -NH(C1-4 alkyl), -N(C1-4 alkyl)(C1-4 alkyl), halogen, -CF3, or -CN. Most preferably, A is phenyl, i.e. unsubstituted phenyl (not substituted with any groups other than R4).
R4 is selected from -CO-(C1-4 alkyl), -CHO, -0(d.4 alkyl), -OH, -0-CO-(C1-4 alkyl), -0-CO-0(C1-4 alkyl), -CO-0(d_4 alkyl), -COOH, -CO-NH2, -CO-NH-(C1-4 alkyl), -CO-N(C1-4 alkyl)(d.4 alkyl), -0-CO-NH2, -0-CO-NH-(C1-4 alkyl), -0-CO-N(d_4 alkyl)(d_4 alkyl), -NH-CO-(d.4 alkyl), -N(C1-4 alkyl)-CO-(C1-4 alkyl), -NH-CO-0(C1-4 alkyl), -N(C1-4 alkyl)-CO-0(d.4 alkyl), -NH2, -NH(d_4 alkyl), -N(C1-4 alky!)(C1-4 alkyl), d_4 alkyl, C2.4 alkenyl, C2_4 alkynyl, -(C1-4 alkyl)-CO-(d.4 alkyl), -(C1-4 alkyl)-CHO, -(C1-4 alkyl)-0(d_4 alkyl), -(C-,.4 alkyl)-OH (e.g., -CH(-CH3)-OH), -(d„4 aikyI)-0-CO-(d_4 alkyl), -(d_4 alkyl)-0-CO-0(C1_4 alkyl), -(C1-4 alkyl)-CO-0(C1-4 alkyl), -(d_4 alkyl)-COOH, -(C1-4 alkyl)-CO-NH2, -(d_4 alkyl)-CO-NH-(C1-4 alkyl), -(C1-4 alkyl)-CO-N(d_4 alkyl)(d_4 alkyl), -(d.4 alkyl)-0-CO-NH2, -(C,.4 alkyl)-0-CO-NH-(d.4 alkyl), -(C1-4 alkyl)-0-CO-N(d_4 alkyl)(C1-4 alkyl), -(d_4 alkyl)-NH-CO-(d.4 alkyl), -(d_4 alkyl)-N(d_4 alkyl)-CO-(C1-4 alkyl), -(C1-4 alkyl)-NH-CO-0(d.4 alkyl), -(C1-4 alkyl)-N(d_4 alkyl)-CO-0(C1-4 alkyl), -(d.4 alkyl)-NH2, -(d.4 alkyl)-NH(d.4 alkyl), -(C1-4 alkyl)-N(d_4 alkyl)(d_4 alkyl), -(C1-4 alkyl)=N-OH (e.g., -C(-CH3)=N-OH), -(d.4 alkyl)=N-0(d.4 alkyl) (e.g., -C(-CH3)=N-0(C1-4 alkyl)), -(d_4 alkyl)=N-0-(d.4 alkyl)-COOH (e.g., -C(-CH3)=N-0-CH2-COOH), -(d.4 alkyl)=N-0-(d_4 alkyl)-CO-0(d.4 alkyl) (e.g., -C(-CH3)=N-0-(d„4 alkyl)-CO-0(d_4 alky))), halogen (e.g., -F, -CI, -Br, or -I), -CF3, -CN, heterocycloalkyl (e.g., morpholinyl), or hydrogen.
Preferably, R4 is selected from -CO-(d.4 alkyl), -CHO, -0(C1-4 alkyl), -OH, -0-CO-(C1-4 alkyl), -0-CO-0(C1-4 alkyl), -CO-0(C1-4 alkyl), -COOH, -CO-NH2, -CO-NH-(C1-4 alkyl), -CO-N(C1-4 alkyl)(C1-4 alkyl), -0-CO-NH2, -0-CO-NH-(C1-4 alkyl), -0-CO-N(C1-4 alkyl)(C1-4 alkyl), -NH-CO-(C1-4 alkyl), -N(d.4 alkyl)-CO-(d_4 alkyl), -NH-CO-0(d.4 alkyl), -N(d_4 alkyl)-CO-0(C1-4 alkyl), -NH2, -NH(C1-4 alkyl), -N(d_4 alkyl)(C1-4 alkyl), C1-4 alkyl, C2-4 alkenyl, d-4 alkynyl, -(d_4 alkyl)-CO-(C1-4 alkyl), -(C1-4 alkyl)-CHO, -(C1-4 alkyl)-0(d-4 alkyl), -(C1-4 alkyl)-OH (e.g., -CH(-CH3)-OH), -(C1-4 alkyl)-0-C0-(d_4 alkyl), -(d.4 alkyl)-0-CO-0(C1-4 alkyl), -(C^ alkyl)-CO-0(C1-4 alkyl), -(C1-4 alkyl)-COOH, -(C1-4 alkyl)-CO-NH2, -(C1-4 alkyl)-CO-NH-(C1-4 alkyl), -(C1-4 alkyl)-CO-N(C1-4 alkyl)(C1-4 alkyl), -(d_4 alkyl)-0-CO-NH2, -(C1-4 alkyl)-0-CO-NH-(C1-4 alkyl), -(C1-4 a!kyl)-0-CO-N(C1-4 alkyl)(d.4 alkyl), -(C1-4 alkyl)-NH-CO-(d_4 alkyl), -(C1-4 alkyl)-N(C1-4 alkyl)-CO-(C1-4 alkyl), -(d_4 a!kyl)-NH-CO-0(C1-4 alkyl), -(C1-4 alkyl)-N(C1-4 alkyl)-CO-0(d.4 alkyl), -(C1-4 alkyl)-NH2, -(C1-4 alkyl)-NH(d-4 alkyl), -(d_4 alkyl)-N(C1-4 alkyl)(C1-4 alkyl), -(C1-4 alkyl)=N-OH (e.g., -C(-CH3)=N-OH), -(d-4 alkyl)=N-0(C1_4 alkyl) (e.g., -C(-CH3)=N-0(d_4 alkyl)), -(d_4 alkyi)=N-0-(C1-4 alkyl)-COOH (e.g., -C(-CH3)=N-0-CH2-COOH), -(d.4 alkyl)=N-0-(d_4 alkyl)-CO-0(d.4 alkyl) (e.g., -C(-CH3)=N-0-(d_4 alkyl)-CO-0(d.4 alkyl)), halogen (e.g., -F, -CI, -Br, or -I), -CF3, -CN, or heterocycloalkyl (e.g., morpholinyl). More preferably, R4 is selected from -CO-(d.4 alkyl), -CHO, -0(d.4 alkyl), -OH, -CO-0(C1-4 alkyl), -COOH, C1-4 alkyl, -(d_4 alkyl)-CO-(d.4 alkyl), -(C1-4 alkyl)-CHO, -(C1-4 alkyl)-0(d_4 alkyl), -(C1-4 alkyl)-OH, -(d.4 alkyl)-CO-0(d_4 alkyl), -(C1-4 alkyl)-COOH, -C(-CH3)=N-OH, -C(-CH3)=N- 0(C1-4 alkyl), -C(-CH3)=N-0-(C1-4 alkyl)-COOH, -C(-CH3)=N-0-(C1-4 alkyl)-CO-0(d.4 alkyl), halogen, -CF3, -CN, or heterocycloalkyl (e.g., morpholinyl). More preferably, R4 is selected from -CO-(d.4 alkyl), -CHO, -0(C1-4 alkyl), or -OH. Even more preferably, R4 is -CO-CH3, -CO-CH2CH3, -OCH3, or -OCH2CH3.
The bond = in formula (I) is a double bond or a single bond. Preferably, = is a double bond. Accordingly, it is preferred that the compound of formula (I) has the following structure:
Figure imgf000009_0001
Particularly preferred compounds of formula (I) are the compounds 1a to 1t shown below as well as pharmaceutically acceptable salts, solvates and prodrugs of each one of these compounds:
Figure imgf000009_0002
1a
(also referred to as "PKCe2138")
Figure imgf000009_0003
1 b
(also referred to as "PKCe141 ")
Figure imgf000009_0004
(also referred to as "PKCe16")
Figure imgf000010_0001
ig
15 (also referred to as "PKCe129")
Figure imgf000011_0001
1h
(also referred to as "PKCe133")
Figure imgf000011_0002
1i
(also referred to as "PKCe140")
Figure imgf000011_0003
10 1j
(also referred to as "PKCe2139")
Figure imgf000011_0004
1k
15 (also referred to as "PKCe2140")
Figure imgf000012_0001
15 (also referred to as "PKCe2141")
Figure imgf000013_0001
15 (also referred to as "PKCe2145")
Figure imgf000014_0001
1t
(also referred to as "PKCe2092") The present invention also provides a pharmaceutical composition comprising a compound of formula (I) as described and defined herein, or a pharmaceutically acceptable salt, solvate or prodrug thereof, in combination with a pharmaceutically acceptable excipient.
The compounds according to the present invention, including in particular the compounds of formula (I), have been found to inhibit the interaction of PKCe with its adaptor protein RACK2, as also shown in Examples 2 and 3. Compound 1 b, for instance, which is an exemplary compound of formula (I) has been demonstrated to inhibit the PKCe/RACK2 interaction in vitro with an IC50 of 5.9 μΜ (see Example 2) and to inhibit the phosphorylation of the PKCe-downstream target Elk-1 in HeLa cells with an IC50 of 1 1.2 μΜ (see Example 3). A large number of further exemplary compounds of formula (I) has likewise been found to inhibit PKCe signaling (see Example 2). Compound 1 a, for example, has been shown to inhibit the PKCe/RACK2 interaction in vitro with an IC50 of 4.1 μΜ.
The compounds of the present invention are particularly advantageous as they allow the isozyme-specific inhibition of PKCe signaling. Accordingly, it has been demonstrated in Example 4 that compound 1 b inhibits the TPA-induced translocation of PKCE - but not that of PKC5 - from the cytosol to the membrane and, furthermore, compound 1 b has been shown to inhibit the PKCe-induced migration of HeLa cells into a gap (see Example 5). The compounds of the present invention thus fulfill all criteria of an inhibitor of PKCe signal transduction, such as inhibition of Elk-1 phosphorylation, inhibition of PKCe translocation to the membrane and inhibition of cell migration. Moreover, the compounds of the invention have been shown not to inhibit cell proliferation (see Example 5), which indicates a lack of toxicity and confirms the suitability of these compounds as medicaments. The compounds according to the invention, and in particular the compounds of formula (I), can thus be used for the therapeutic intervention in diseases/disorders in which PKCe signaling is implicated (particularly diseases/disorders associated with an increased activity or a hyperactivity of PKCe signaling), such as, e.g., cardiovascular disorders (e.g., cardiac hypertrophy, hypertrophic cardiomyopathy, or heart failure (including, e.g., congestive heart failure)), anxiety, pain (e.g., chronic pain), migraine, allergies, inflammatory disorders, autoimmune disorders, diabetes (including also insulin resistance), diabetic complications (such as, e.g., diabetic retinopathy, diabetic nephropathy, diabetic cardiomyopathy, or diabetic neuropathy), cancer (such as, e.g., stomach cancer, lung cancer, thyroid cancer, colon cancer, breast cancer; including also metastatic cancer, metastasis, drug-resistant cancer, or multidrug-resistant cancer), neurological disorders (e.g., Alzheimer's disease, Parkinson's disease, bipolar disorder, or stroke; including also neurodegenerative disorders), alopecia, or alcoholism (Shirai et al., 2008; Pass et al., 2001 ; Reichling et al. , 2009; Galeotti et al., 2013; Takeishi et al., 2000; Takahashi et al., 2000; Gorin et al., 2009; Akita, 2008; Ferreira et al., 2010; Choi et al., 2002; Johnson et al., 1996; Ikeda et al., 2001 ; Mochly-Rosen et al., 2012 and references cited therein).
Accordingly, without being bound by theory, the present invention provides a compound of formula (I) as described and defined herein, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition comprising any of the aforementioned entities and a pharmaceutically acceptable excipient, for use in the treatment or prevention of cardiovascular disorders (e.g., cardiac hypertrophy, hypertrophic cardiomyopathy, or heart failure (including, e.g., congestive heart failure)), anxiety, pain (e.g., chronic pain), migraine, allergies, inflammatory disorders, autoimmune disorders, diabetes (including also insulin resistance), diabetic complications (such as, e.g. , diabetic retinopathy, diabetic nephropathy, diabetic cardiomyopathy, or diabetic neuropathy), cancer (such as, e.g., stomach cancer, lung cancer, thyroid cancer, colon cancer, breast cancer; including also metastatic cancer, metastasis, drug-resistant cancer, or multidrug-resistant cancer), neurological disorders (e.g., Alzheimer's disease, Parkinson's disease, bipolar disorder, or stroke; including also neurodegenerative disorders), alopecia, or alcoholism.
The invention likewise relates to the use of the compounds provided herein, including the compounds of formula (I) or pharmaceutically acceptable salts, solvates or prodrugs thereof, in the preparation of a medicament for the treatment or prevention of cardiovascular disorders (e.g., cardiac hypertrophy, hypertrophic cardiomyopathy, or heart failure (including, e.g., congestive heart failure)), anxiety, pain (e.g., chronic pain), migraine, allergies, inflammatory disorders, autoimmune disorders, diabetes (including also insulin resistance), diabetic complications (such as, e.g., diabetic retinopathy, diabetic nephropathy, diabetic cardiomyopathy, or diabetic neuropathy), cancer (such as, e.g., stomach cancer, lung cancer, thyroid cancer, colon cancer, breast cancer; including also metastatic cancer, metastasis, drug-resistant cancer, or multidrug-resistant cancer), neurological disorders (e.g., Alzheimer's disease, Parkinson's disease, bipolar disorder, or stroke; including also neurodegenerative disorders), alopecia, or alcoholism.
The invention also provides a method of treating or preventing a disorder or condition selected from the group consisting of cardiovascular disorders (e.g., cardiac hypertrophy, hypertrophic cardiomyopathy, or heart failure (including, e.g., congestive heart failure)), anxiety, pain (e.g., chronic pain), migraine, allergies, inflammatory disorders, autoimmune disorders, diabetes (including also insulin resistance), diabetic complications (such as, e.g., diabetic retinopathy, diabetic nephropathy, diabetic cardiomyopathy, or diabetic neuropathy), cancer (such as, e.g., stomach cancer, lung cancer, thyroid cancer, colon cancer, breast cancer; including also metastatic cancer, metastasis, drug-resistant cancer, or multidrug-resistant cancer), neurological disorders (e.g., Alzheimer's disease, Parkinson's disease, bipolar disorder, or stroke; including also neurodegenerative disorders), alopecia, and alcoholism, the method comprising the administration of a compound according to the invention, particularly a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition comprising any of the aforementioned entities and a pharmaceutically acceptable excipient, to a subject (e.g., a human) in need thereof. Protein-protein interfaces, such as the PKCe/RACK2 interface, are much more difficult to target than classical Iigand-binding sites, due to their particular physicochemical properties (extensive, hydrophobic, surface-exposed interfaces). Attempts to tackle protein-protein interactions have been reported in recent years but experience is still quite limited and experimental aspects are challenging as well (Rechfeid et al., 201 1 ). The EAVSLKPT peptide, derived from the binding site of PKCe to RACK2 and used for molecular modeling in the context of the present invention (see Example 1 ), inhibits this interaction with an IC5C of 1 .02 μΜ. Therefore, a peptidomimetic of EAVSLKPT such as compound 1 b (PKCe141 ) with an IC50 of 5.9 μΜ may be close to the optimum already. Also aurothiomalate, which prevents the interaction of PKCi and its adaptor protein Par6, is active in the range of 10 μΜ (Erdogan et al., 2006). While a compound with an IC50 in the nanomolar range would be preferable, this is not essential for clinical application. For example, the ribonucleotide reductase inhibitor hydroxyurea inhibits ribonucleotide reductase with an IC50 of 37.2 μΜ (Easmon et al., 2001 ) and is used in the clinic as an anticancer agent. Furthermore, many clinically relevant kinase inhibitors which are active at nanomolar concentrations in vitro have cellular IC50 values closer to the micromolar range due to the higher physiological concentrations of ATP relative to those typically used for in vitro assays.
The invention further relates to novel compounds, namely the compounds 1 a, 1j, 1 k, 11, 1 m, 1 n, 1 o, 1 p, 1 q, 1 r, 1s and 1t having the structures shown above, as well as pharmaceutically acceptable salts, solvates and prodrugs of each one of these compounds. These compounds as provided in the context of the present invention are particularly useful as medicaments, e.g., for the treatment or prevention of diseases/disorders in which PKCE signaling is implicated, as described herein above.
Moreover, the present invention also relates to the compounds shown in Table 1 below, including in particular the compounds PKCe22, PKCe39, PKCe40, PKCe48, PKCe49, PKCe52, PKCe67, PKCe73, PKCe76, PKCe86, PKCe87, PKCe89, PKCe95, PKCe96, PKCe97, PKCe106, PKCe107, PKCe109, PKCe1 14, PKCe125, PKCe143, PKCe145, PKCe2019, PKCe2020, PKCe2021 , PKCe2051 , PKCe2052, PKCe2053, PKCe2054, PKCe2088, PKCe2089, PKCe2090, and PKCe2091 , as well as pharmaceutically acceptable salts, solvates and prodrugs of each one of these compounds. The invention further relates to the use of these compounds as medicaments, including their use in the treatment or prevention of diseases/disorders in which PKCe signaling is implicated, such as the specific diseases/disorders described herein above. The compounds of the present invention, including the compounds of formula (I), can be prepared by methods known in the field of synthetic chemistry. For example, the compounds of the invention may be prepared in accordance with or in analogy to the synthetic routes described in Example 8. Moreover, certain compounds of formula (I), such as compound 1 b (Asinex ID: ASN 05545158; CAS 602293-00-5) and compound 1 c (Asinex ID: ASN 02538754; CAS 552819-38-2), are also commercially available, e.g., from Asinex Ltd., Moscow, Russia.
As used herein, the term "alkyl" refers to a monovalent saturated aliphatic (i.e. non-aromatic) acyclic hydrocarbon group (i.e., a group consisting of carbon atoms and hydrogen atoms), which may be linear or branched and does not comprise any carbon-to- carbon double bond or any carbon-to-carbon triple bond. The term "C1-4 alkyl" denotes an alkyl group having 1 to 4 carbon atoms. Preferred exemplary alkyl groups are methyl, ethyl, propyl, or butyl.
As used herein, the term "alkenyl" refers to a monovalent unsaturated aliphatic acyclic hydrocarbon group, which may be linear or branched and comprises at least one carbon-to- carbon double bond while it does not comprise any carbon-to-carbon triple bond. The term "C2-4 alkenyl" denotes an alkenyl group having 2 to 4 carbon atoms. Preferred exemplary alkenyl groups are ethenyl, propenyl, or butenyl. As used herein, the term "alkynyl" refers to a monovalent unsaturated aliphatic acyclic hydrocarbon group, which may be linear or branched and comprises at least one carbon-to- carbon triple bond and optionally one or more carbon-to-carbon double bonds. The term "C2-4 alkynyl" denotes an alkynyl group having 2 to 4 carbon atoms. Preferred exemplary alkynyl groups are ethynyl, propynyl, or butynyl.
As used herein, the term "aryl" refers to a monovalent aromatic hydrocarbon group, including bridged ring and/or fused ring systems, containing at least one aromatic ring. "Aryl" may, for example, refer to phenyl, naphthyl or anthracenyl. As used herein, the term "heteroaryl" refers to a monovalent aromatic ring group, which may be a monocyclic ring group or a bridged ring and/or fused ring system (e.g., a bicyclic ring system), said aromatic ring group comprising one or more (such as, e.g., one, two, or three) ring heteroatoms independently selected from O, S, or N, wherein the aromatic ring group may, e.g., have 5 to 14 (particularly 5 or 6) ring atoms. Non-limiting examples of "heteroaryl" groups include thiazolyl (e.g., 1 ,3-thiazol-2-yl), furanyl, thiophenyl (i.e., thienyl), pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, furazanyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, benzothiazolyl (e.g., 1 ,3-benzothiazol-2-yl), benzofuranyl, benzothiophenyl, benzopyrrolyl, benzoimidazolyl, benzopyrazolyl, benzoxazolyl, benzisoxazolyl, benzofurazanyl, benzopyridinyl, benzopyrimidinyl, benzopyrazinyl, or benzopyridazinyl.
As used herein, the term "heterocycloalkyi" refers to a 3 to 10 atom ring or ring system containing one or more (e.g., one, two or three) ring heteroatoms independently selected from O, S, or N. "Heterocycloalkyi" may, for example, refer to tetrahydrofuranyl, piperidinyl, piperazinyl, aziridinyl, azetidinyl, pyrrolidinyl, imidazolidinyl, morpholinyl (e.g., morpholin-4- yl), pyrazolidinyl, tetrahydrothienyl, octahydroquinolinyl, octahydroisoquinolinyl, oxazolidinyl, or isoxazolidinyl. As used herein, the term "halogen" refers to fluoro (-F), chloro (-CI), bromo (-Br) or iodo (-I).
The scope of the invention embraces all pharmaceutically acceptable salt forms of the compounds provided herein, including in particular the compounds of formula (I), which may be formed, e.g., by protonation of an atom carrying an electron lone pair which is susceptible to protonation, such as an amino group, with an inorganic or organic acid, or as a salt of a carboxylic acid group with a physiologically acceptable cation as they are well-known in the art. Exemplary base addition salts comprise, for example, alkali metal salts such as sodium or potassium salts; alkaline earth metal salts such as calcium or magnesium salts; ammonium salts; aliphatic amine salts such as trimethylamine, triethylamine, dicyclohexylamine, ethanolamine, diethanolamine, triethanolamine, procaine salts, meglumine salts, or ethylenediamine salts; aralkyl amine salts such as N,N- dibenzylethylenediamine salts, benethamine salts; heterocyclic aromatic amine salts such as pyridine salts, picoline salts, quinoline salts or isoquinoline salts; quaternary ammonium salts such as tetramethylammonium salts, tetraethylammonium salts, benzyltrimethylammonium salts, benzyltriethylammonium salts, benzyltributylammonium salts, methyltrioctylammonium salts or tetrabutylammonium salts; and basic amino acid salts such as arginine salts or lysine salts. Exemplary acid addition salts comprise, for example, mineral acid salts such as hydrochloride, hydrobromide, hydroiodide, sulfate salts, nitrate salts, phosphate salts (such as, e.g., phosphate, hydrogenphosphate, or dihydrogenphosphate salts), carbonate salts, hydrogencarbonate salts or perchlorate salts; organic acid salts such as acetate, propionate, butyrate, pentanoate, hexanoate, heptanoate, octanoate, cyclopentanepropionate, undecanoate, lactate, maleate, oxalate, fumarate, tartrate, malate, citrate, nicotinate, benzoate, salicylate or ascorbate salts; sulfonate salts such as methanesulfonate, ethanesulfonate, 2-hydroxyethanesulfonate, benzenesulfonate, p-toluenesulfonate (tosylate), 2-naphthalenesulfonate, 3- phenylsulfonate, or camphorsulfonate salts; and acidic amino acid salts such as aspartate or glutamate salts.
Moreover, the scope of the invention embraces the compounds provided herein, including in particular the compounds of formula (I), in any solvated form, including, e.g., solvates with water, for example hydrates, or with organic solvents such as, e.g., methanol, ethanol or acetonitrile, i.e., as a methanolate, ethanolate or acetonitrilate, respectively, or in the form of any polymorph. Furthermore, the formulae in the present specification are intended to cover all possible stereoisomers, including enantiomers and diastereomers, of the indicated compounds. Thus, all stereoisomers of the compounds of the present invention, including the compounds of formula (I), are contemplated as part of the present invention, either in admixture or in pure or substantially pure form. The scope of the compounds according to the invention embraces all of the possible stereoisomers and their mixtures, it particularly embraces the racemic forms and the isolated optical isomers. The racemic forms can be resolved by physical methods, such as, e.g., fractional crystallization, separation or crystallization of diastereomeric derivatives or separation by chiral column chromatography. The individual optical isomers can be obtained from the racemates using conventional methods, such as, e.g., salt formation with an optically active acid followed by crystallization. The present invention also embraces all possible tautomers of the compounds provided herein, including the compounds of formula (I). Pharmaceutically acceptable prodrugs of the compounds according to the present invention are derivatives which have chemically or metabolically cleavable groups and become, by solvolysis or under physiological conditions, the compounds of the invention which are pharmaceutically active in vivo. Prodrugs of compounds according to the the present invention may be formed in a conventional manner with a functional group of the compounds such as, e.g., with an amino, hydroxy or carboxy group. The prodrug derivative form often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, Bundgaard, H., Design of Prodrugs, pp. 7-9, 21 -24, Elsevier, Amsterdam 1985). Prodrugs include acid derivatives well known to the person skilled in the art, such as, for example, esters prepared by reaction of the parent acidic compound with a suitable alcohol, or amides prepared by reaction of the parent acid compound with a suitable amine. When a compound of the present invention has a carboxyl group, an ester derivative prepared by reacting the carboxyl group with a suitable alcohol or an amide derivative prepared by reacting the carboxyl group with a suitable amine is exemplified as a prodrug. An especially preferred ester derivative as a prodrug is methylester, ethylester, n- propylester, isopropylester, n-butylester, isobutylester, tert-butylester, morpholinoethylester, Ν,Ν-diethyiglycolamidoester or a-acetoxyethylester. When a compound of the present invention has a hydroxy group, an acyloxy derivative prepared by reacting the hydroxyl group with a suitable acylhalide or a suitable acid anhydride is exemplified as a prodrug. An especially preferred acyloxy derivative as a prodrug is -OC(=0)-CH3, -OC(=0)-C2H5, -OC(=0)-(tert-Bu), -OC(=0)-C15H31 , -OC(=0)-(m-COONa-Ph), -OC(=0)-CH2CH2COONa, -0(C=0)-CH(NH2)CH3 or -OC(=0)-CH2-N(CH3)2. When a compound of the present invention has an amino group, an amide derivative prepared by reacting the amino group with a suitable acid halide or a suitable mixed anhydride is exemplified as a prodrug. An especially preferred amide derivative as a prodrug is -NHC(=0)-(CH2)20CH3 or -NHC(=0)- CH(NH2)CH3.
The compounds described herein may be administered as compounds per se or may be formulated as medicaments. The medicaments/pharmaceutical compositions may comprise one or more pharmaceutically acceptable excipients, such as carriers, diluents, fillers, disintegrants, lubricating agents, binders, colorants, pigments, stabilizers, preservatives, antioxidants, or solubility enhancers.
In particular, the pharmaceutical compositions may comprise one or more solubility enhancers, such as, e.g., poly(ethylene glycol), including poly(ethylene glycol) having a molecular weight in the range of about 200 to about 5,000 Da, ethylene glycol, propylene glycol, non-ionic surfactants, tyloxapol, polysorbate 80, macrogol-15-hydroxystearate, phospholipids, lecithin, dimyristoyl phosphatidylcholine, dipalmitoyl phosphatidylcholine, distearoyl phosphatidylcholine, cyclodextrins, hydroxyethyl-p-cyclodextrin, hydroxypropyl-β- cyclodextrin, hydroxyethyl-y-cyclodextrin, hydroxypropyl-v-cyclodextrin, dihydroxypropyl-β- cyclodextrin, glucosyl-a-cyclodextrin, glucosyl-p-cyclodextrin, diglucosyl- -cyclodextrin, maltosyl-a-cyclodextrin, maltosyl-3-cyclodextrin, maltosyl-y-cyclodextrin, maltotriosyl-β- cyclodextrin, maltotriosyl-y-cyclodextrin, dimaltosyl-P-cyclodextrin, methyl- -cyclodextrin, carboxyalkyl thioethers, hydroxy-propyl methylcellulose, hydroxypropylcellulose, polyvinylpyrrolidone, vinyl acetate copolymers, vinyl pyrrolidone, sodium lauryl sulfate, dioctyl sodium sulfosuccinate, or any combination thereof.
The pharmaceutical compositions can be formulated by techniques known to the person skilled in the art, such as the techniques published in Remington's Pharmaceutical Sciences, 20th Edition. The pharmaceutical compositions can be formulated as dosage forms for oral, parenteral, such as intramuscular, intravenous, subcutaneous, intradermal, intraarterial, intracardial, rectal, nasal, topical, aerosol or vaginal administration. Dosage forms for oral administration include coated and uncoated tablets, soft gelatin capsules, hard gelatin capsules, lozenges, troches, solutions, emulsions, suspensions, syrups, elixirs, powders and granules for reconstitution, dispersible powders and granules, medicated gums, chewing tablets and effervescent tablets. Dosage forms for parenteral administration include solutions, emulsions, suspensions, dispersions and powders and granules for reconstitution. Emulsions are a preferred dosage form for parenteral administration. Dosage forms for rectal and vaginal administration include suppositories and ovula. Dosage forms for nasal administration can be administered via inhalation and insufflation, for example by a metered inhaler. Dosage forms for topical administration include creams, gels, ointments, salves, patches and transdermal delivery systems.
The compounds according to the invention, in particular the compounds of formula (I), or the above described pharmaceutical compositions comprising one or more compounds of formula (I) may be administered to a subject by any convenient route of administration, whether systemically/peripherally or at the site of desired action, including but not limited to one or more of: oral (e.g., as a tablet, capsule, or as an ingestible solution), topical (e.g., transdermal, intranasal, ocular, buccal, and sublingual), parenteral (e.g., using injection techniques or infusion techniques, and including, for example, by injection, e.g., subcutaneous, intradermal, intramuscular, intravenous, intraarterial, intracardiac, intrathecal, intraspinal, intracapsular, subcapsular, intraorbital, intraperitoneal, intratracheal, subcuticular, intraarticular, subarachnoid, or intrasternal by, e.g., implant of a depot, for example, subcutaneously or intramuscularly), pulmonary (e.g., by inhalation or insufflation therapy using, e.g., an aerosol, e.g., through mouth or nose), gastrointestinal, intrauterine, intraocular, subcutaneous, ophthalmic (including intravitreal or intracameral), rectal, and vaginal.
If said compounds or pharmaceutical compositions are administered parenterally, then examples of such administration include one or more of: intravenously, intraarterially, intraperitoneal^, intrathecally, intraventricular^, intraurethrally, intrasternally, intracardially, intracranially, intramuscularly or subcutaneously administering the compounds or pharmaceutical compositions, and/or by using infusion techniques. For parenteral administration, the compounds are best used in the form of a sterile aqueous solution which may contain other substances, for example, enough salts or glucose to make the solution isotonic with blood. The aqueous solutions should be suitably buffered (preferably to a pH of from 3 to 9), if necessary. The preparation of suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well known to those skilled in the art.
Said compounds or pharmaceutical compositions can also be administered orally in the form of tablets, capsules, ovules, elixirs, solutions or suspensions, which may contain flavoring or coloring agents, for immediate-, delayed-, modified-, sustained-, pulsed- or controlled-release applications. The tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycolate, croscarmellose sodium and certain complex silicates, and granulation binders such as polyvinylpyrrolidone, hydroxypropylmethylcellulose (HPMC), hydroxypropylcellulose (HPC), sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, stearic acid, glyceryl behenate and talc may be included. Solid compositions of a similar type may also be employed as fillers in gelatin capsules. Preferred excipients in this regard include lactose, starch, a cellulose, or high molecular weight polyethylene glycols. For aqueous suspensions and/or elixirs, the agent may be combined with various sweetening or flavoring agents, coloring matter or dyes, with emulsifying and/or suspending agents and with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof. Alternatively, said compounds or pharmaceutical compositions can be administered in the form of a suppository or pessary, or it may be applied topically In the form of a gel, hydrogel, lotion, solution, cream, ointment or dusting powder. The compounds of the present invention may also be dermally or transdermally administered, for example, by the use of a skin patch.
Said compounds or pharmaceutical compositions may also be administered by sustained release systems. Suitable examples of sustained-release compositions include semi-permeable polymer matrices in the form of shaped articles, e.g., films, or microcapsules. Sustained-release matrices include, e.g., polylactides (U.S. Pat. No. 3,773,919), copolymers of L-glutamic acid and gamma-ethyl-L-glutamate (Sidman, U. et al., Biopolymers 22:547-556 (1983)), poly(2-hydroxyethyl methacrylate) (R. Langer et al., J. Biomed. Mater. Res. 15:167-277 (1981 ), and R. Langer, Chem. Tech. 12:98-105 (1982)), ethylene vinyl acetate (R. Langer et al., Id.) or poly-D-(-)-3-hydroxybutyric acid (EP133988). Sustained-release pharmaceutical compositions also include liposomally entrapped compounds. Liposomes containing a compound of the present invention can be prepared by methods known in the art, such as, e.g., the methods described in any one of: DE3218121 ; Epstein et al., Proc. Natl. Acad. Sci. (USA) 82:3688-3692 (1985); Hwang et al., Proc. Natl. Acad. Sci. (USA) 77:4030-4034 (1980); EP0052322; EP0036676; EP088046; EP0143949; EP0142641 ; Japanese Pat. Appl. 83-1 18008; U.S. Pat. Nos. 4,485.045 and 4,544,545; and EP0102324. Said compounds or pharmaceutical compositions may also be administered by the pulmonary route, rectal routes, or the ocular route. For ophthalmic use, they can be formulated as micronized suspensions in isotonic, pH adjusted, sterile saline, or, preferably, as solutions in isotonic, pH adjusted, sterile saline, optionally in combination with a preservative such as a benzalkonium chloride. Alternatively, they may be formulated in an ointment such as petrolatum.
It is also envisaged to prepare dry powder formulations of the compounds of the present invention, including the compounds of formula (I), for pulmonary administration, particularly inhalation. Such dry powders may be prepared by spray drying under conditions which result in a substantially amorphous glassy or a substantially crystalline bioactive powder. Accordingly, dry powders of the compounds of the present invention can be made according to the emulsification/spray drying process disclosed in WO 99/16419 or WO 01/85136. Spray drying of solution formulations of the compounds of the present invention is carried out, for example, as described generally in the "Spray Drying Handbook", 5th ed., K. Masters, John Wiley & Sons, Inc. , NY, NY (1991 ), and in WO 97/41833 or WO 03/05341 1 .
For topical application to the skin, said compounds or pharmaceutical compositions can be formulated as a suitable ointment containing the active compound suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, emulsifying wax and water. Alternatively, they can be formulated as a suitable lotion or cream, suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, a polyethylene glycol, liquid paraffin, polysorbate 60, cetyl esters wax, 2-octyldodecanol, benzyl alcohol and water.
Typically, a physician will determine the actual dosage which will be most suitable for an individual subject. The specific dose level and frequency of dosage for any particular individual subject may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the individual subject undergoing therapy. A proposed, yet non-limiting dose of the compounds according to the invention, particularly the compounds of formula (I), for oral administration to a human (of approximately 70 kg body weight) may be 0.05 to 2500 mg, preferably 0.1 mg to 1000 mg, of the active ingredient per unit dose. The unit dose may be administered, for example, 1 to 3 times per day. The unit dose may also be administered 1 to 7 times per week, e.g., with not more than one administration per day. It will be appreciated that it may be necessary to make routine variations to the dosage depending on the age and weight of the patient/subject as well as the severity of the condition to be treated. The precise dose and also the route of administration will ultimately be at the discretion of the attendant physician or veterinarian.
The subject or patient, such as the subject in need of treatment or prevention, may be an animal (e.g., a non-human animal), a vertebrate animal, a mammal, a rodent (e.g., a guinea pig, a hamster, a rat, a mouse), a murine (e.g., a mouse), a canine (e.g., a dog), a feline (e.g., a cat), a porcine (e.g., a pig), an equine (e.g., a horse), a primate, a simian (e.g., a monkey or ape), a monkey (e.g., a marmoset, a baboon), an ape (e.g., a gorilla, chimpanzee, orang-utan, gibbon), or a human. The meaning of the terms "eukaryote", "animal", "mammal", etc. is well known in the art and can, for example, be deduced from Wehner und Gehring (1995; Thieme Verlag). In the context of this invention, it is particularly envisaged that animals are to be treated which are economically, agronomically or scientifically important. Scientifically important organisms include, but are not limited to, mice, rats, and rabbits. Lower organisms such as, e.g., fruit flies like Drosophila melagonaster and nematodes like Caenorhabditis elegans may also be used in scientific approaches. Non-limiting examples of agronomically important animals are sheep, cattle and pigs, while, for example, cats and dogs may be considered as economically important animals. Preferably, the subject/patient is a mammal; more preferably, the subject/patient is a human or a non-human mammal (such as, e.g., a guinea pig, a hamster, a rat, a mouse, a rabbit, a dog, a cat, a horse, a monkey, an ape, a marmoset, a baboon, a gorilla, a chimpanzee, an orang-utan, a gibbon, a sheep, cattle, or a pig); even more preferably, the subject/patient is a human.
The term "treatment of a disorder or disease" as used herein is well known in the art. "Treatment of a disorder or disease" implies that a disorder or disease is suspected or has been diagnosed in a patient/subject. A patient/subject suspected of suffering from a disorder or disease typically shows specific clinical and/or pathological symptoms which a skilled person can easily attribute to a specific pathological condition (i.e., diagnose a disorder or disease). "Treatment of a disorder or disease" may, for example, lead to a halt in the progression of the disorder or disease (e.g., no deterioration of symptoms) or a delay in the progression of the disorder or disease (in case the halt in progression is of a transient nature only). "Treatment of a disorder or disease" may also lead to a partial response (e.g., amelioration of symptoms) or complete response (e.g., disappearance of symptoms) of the subject/patient suffering from the disorder or disease. "Amelioration" of a disorder or disease may, for example, lead to a halt in the progression of the disorder or disease or a delay in the progression of the disorder or disease. Such a partial or complete response may be followed by a relapse. It is to be understood that a subject/patient may experience a broad range of responses to a treatment (e.g., the exemplary responses as described herein above).
Treatment of a disorder or disease may, inter alia, comprise curative treatment (preferably leading to a complete response and eventually to healing of the disorder or disease) and palliative treatment (including symptomatic relief).
Also the term "prevention of a disorder or disease" as used herein is well known in the art. For example, a patient/subject suspected of being prone to suffer from a disorder or disease as defined herein may, in particular, benefit from a prevention of the disorder or disease. The subject/patient may have a susceptibility or predisposition for a disorder or disease, including but not limited to hereditary predisposition. Such a predisposition can be determined by standard assays, using, for example, genetic markers or phenotypic indicators. It is to be understood that a disorder or disease to be prevented in accordance with the present invention has not been diagnosed or cannot be diagnosed in the patient/subject (for example, the patient/subject does not show any clinical or pathological symptoms). Thus, the term "prevention" comprises the use of compounds of the present invention before any clinical and/or pathological symptoms are diagnosed or determined or can be diagnosed or determined by the attending physician. In this specification, a number of documents including patent applications, scientific literature and manufacturers' manuals are cited. The disclosure of these documents, while not considered relevant for the patentability of this invention, is herewith incorporated by reference in its entirety. More specifically, all referenced documents are incorporated by reference to the same extent as if each individual document was specifically and individually indicated to be incorporated by reference. The present invention particularly relates to the following items: 1. A compound of the following formula (I)
Figure imgf000027_0001
(I) wherein: n is an integer of 0 to 4, and each R1 is independently selected from C2.4 alkyl, C2.4 alkenyl, C2.4 alkynyl, -OH, -0(d.4 alkyl), -0(d.4 alkyl)-OH, -0(C1-4 alkyl)-0(C1-4 alkyl), -SH, -S(C1-4 alkyl), -S(CV4 alkyl)-SH, or -S(d.4 alkyl)-S(C1-4 alkyl), -NH2, -NH(C1-4 alkyl), -N(C,.4 alkyl)(C1-4 alkyl), halogen, -CF3, or -CN;
or, alternatively, n is 2, 3 or 4, and two groups R1 attached to adjacent carbon atoms are mutually linked to form a group -0-CH2-0-, -0-CH2-CH2-0- or -0-CH2-CH2-CH2-0-, while the further group(s) R1, if present, is/are independently selected from C1.4 alkyl, C2.4 alkenyl, C2.4 alkynyl, -OH, -0(C -4 alkyl), -0(d_4 alkyl)-OH, -0(d.4 alkyl)-0(d.4 alkyl), -SH, -S(C1-4 alkyl), -S(d_4 alkyl)-SH, or -S(d_4 alkyl)-S(d.4 alkyl), -NH2, -NH(C1-4 alkyl), -N(d-4 aikyl)(Ci_4 alkyl), halogen, -CF3, or -CN;
R2 is selected from hydrogen, C1-4 alkyl, C2.4 alkenyl, C2.4 alkynyl, -OH, -0(d.4 alkyl), -SH, -S(d.4 alkyl), -NH2, -NH(d.4 alkyl), -N(d.4 alkyl)(d.4 alkyl), halogen, -CF3, or -CN;
R3 is selected from -NH2, -NH(d.4 alkyl), -N(C1-4 alkyl)(C1-4 alkyl), -OH, -0(Ci.4 alkyl), -SH, -S(d_4 alkyl), or hydrogen;
X is selected from S, O, N(H), or N(d„4 alkyl); L is -(CH2)i-4-, wherein one -CH2- unit comprised in said -(CH2)i-4- is replaced by a group selected from -CO-NH-, -CO-N(d.4 alkyl)-, -NH-CO-, -N(d-4 alkyl)-CO-, -0-, -CO-, -NH-, -N(d_4 alkyl)-, -S-, -SO-, or -S02-; A is aryl or heteroaryl, wherein said aryl or said heteroaryl is optionally substituted with one or more groups independently selected from d-4 alkyl, C2.4 alkenyl, C2.4 alkynyl, -OH, -0(C1-4 alkyl), -SH, -S(C1-4 alkyl), -NH2, -NH(C1-4 alkyl), -N(d.4 alkyl)(C1-4 alkyl), halogen, -CF3, or -CN; R4 is selected from -CO-(C1-4 alkyl), -CHO, -0(d-4 alkyl), -OH, -0-CO-(C1-4 alkyl), -0-CO-0(C1-4 alkyl), -CO-0(d.4 alkyl), -COOH, -CO-NH2, -CO-NH-(d.4 alkyl), -CO-N(d_4 alkyl)(d-4 alkyl), -0-CO-NH2, -0-CO-NH-(d.4 alkyl), -0-CO-N(C1-4 alkyl)(d_4 alkyl), -NH-CO-(C1-4 alkyl), -N(d-4 alkyl)-CO-(C1-4 alkyl), -NH-CO-0(d.4 alkyl), -N(d_4 alkyl)-CO-0(d_4 alkyl), -NH2, -NH(d.4 alkyl), -N(d_4 alkyl)(C1-4 alkyl), d.4 alkyl, C2.4 alkenyl,
C2.4 alkynyl, -(d_4 alkyl)-C0-(d.4 alkyl), -(d.4 alkyl)-CHO, -(d-4 alkyl)-0(d.4 alkyl), -(C1-4 alkyl)-OH, -(C1-4 alkyl)-0-CO-(d_4 alkyl), -(d„4 alkyl)-0-CO-0(d.4 alkyl), -(d.4 alkyl)-CO-0(d.4 alkyl), -(d.4 alkyl)-COOH, -(d.4 alkyl)-CO-NH2, -(d_4 alkyl)-CO-NH-(d.4 alkyl), -(d.4 alkyl)-CO-N(d_4 alkyl)(d-4 alkyl), -(d_4 alkyl)-0-CO-NH2, -(d.4 alkyl)-0-CO-NH-(d.4 alkyl),
-(C1-4 alkyl)-0-CO-N(d.4 alkyl)(d.4 alkyl), -(d_4 alkyl)-NH-CO-(d_4 alkyl), -(d-4 alkyl)-N(d.4 alkyl)-CO-(d.4 alkyl), -(C1-4 alkyl)-NH-CO-0(C1-4 alkyl), -(d.4 alkyl)-N(d.4 alkyl)-CO-0(d_4 alkyl), -(C,.4 alkyl)-NH2, -(d_4 alkyl)-NH(d.4 alkyl), -(d_4 alkyl)-N(d.4 alkyl)(C1-4 alkyl), -(d_4 alkyl)=N-OH, -(d.4 alkyl)=N-0(d.4 alkyl), -(C1-4 alkyl)=N-0-(d.4 alkyl)-COOH, -(d_4 alkyl)=N-0-(d_4 alkyl)-CO-0(d_4 alkyl), halogen, -CF3, -CN, heterocycloalkyl, or hydrogen; and
^= is a double bond or a single bond; or a pharmaceutically acceptable salt, solvate or prodrug thereof for use in the treatment or prevention of a cardiovascular disorder, cardiac hypertrophy, heart failure, anxiety, pain, chronic pain, migraine, an allergy, an inflammatory disorder, an autoimmune disorder, diabetes, diabetic complications, diabetic retinopathy, diabetic nephropathy, diabetic cardiomyopathy, diabetic neuropathy, cancer, metastatic cancer, drug-resistant cancer, stomach cancer, lung cancer, thyroid cancer, colon cancer, breast cancer, bipolar disorder, stroke, alopecia, or alcoholism.
The compound for use according to item 1 , wherein n is an integer of 1 to 4, one group R1 is selected from -0(d.4 alkyl), -OH, -0(C1-4 alkyl)-OH, or -0(C1-4 alkyl)-0(C1-4 alkyl), and the remaining group(s) R1, if present, is/are independently selected from C2-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, -OH, -0(C1-4 alkyl), -0(C1-4 alkyl)-OH, -0(C1-4 alkyl)-0(C1.4 alkyl), -SH, -S(d.4 alkyl), -S(C1-4 alkyl)-SH, -S(C1-4 alkyl)-S(C1-4 alkyl), -NH2, -NH(C1-4 alkyl), -N(d.4 aikyi)(C1-4 alkyl), halogen, -CF3, or -CN.
The compound for use according to item 1 , wherein n is 2, 3 or 4, and two groups R1 attached to adjacent carbon atoms are mutually linked to form a group -0-CH2-0-, -0-CH2-CH2-0- or -0-CH2-CH2-CH2-0-, while the further group(s) R1, if present, is/are independently selected from C1-4 alkyl, C2_4 alkenyl, C2-4 alkynyl, -OH, -0(C1-4 alkyl), -0(d_4 alkyl)-OH, -0(d.4 alkyl)-0(C1-4 alkyl), -SH, -S(C1-4 alkyl), -S(C1-4 alkyl)-SH, -S(C1-4 alkyl)-S(d.4 alkyl), -NH2, -NH(C1-4 alkyl), -N(d.4 alkyl)(d-4 alkyl), halogen, -CF3, or -CN.
The compound for use according to any of items 1 to 3, wherein R3 is -NH2. The compound for use according to any of items 1 to 4, wherein X is S. The compound for use according to any of items 1 to 5, wherein L is -CO-NH- or -CO-N(d_4 alkyl)-, wherein the nitrogen atom comprised in said -CO-NH- or said -CO-N(d_4 alkyl)- is attached to ring A. The compound for use according to any of items 1 to 6, wherein A is phenyl, 1 ,3-thiazol-2-yl or 1 ,3-benzothiazol-2-yl, wherein said phenyl, said 1 ,3-thiazol-2-yl or said 1 ,3-benzothiazol-2-yl is optionally substituted with one or more groups independently selected from C -4 alkyl, C2_4 alkenyl, C2_4 alkynyl, -OH, -0(d. alkyl), -SH, -S(d_4 alkyl), -NH2, -NH(C1-4 alkyl), -N(C,.4 alkyl)(d.4 alkyl), halogen, -CF3, or -CN. The compound for use according to any of items 1 to 7, wherein R4 is selected from -CO-(Ci_4 alkyl), -CHO, -0(C1-4 alkyl), -OH, -CO-0(C1-4 alkyl), -COOH, C1-4 alkyl, -(C1-4 alkyl)-CO-(d.4 alkyl), -(C1-4 alkyl)-CHO, -(d-4 alkyl)-0(C1-4 alkyl), -(C1-4 alkyl)-OH, -(C1-4 alkyl)-CO-0(d-4 alkyl), -(C1-4 alkyl)-COOH, -C(-CH3)=N-OH, -C(-CH3)=N-0(C1.4 alkyl), -C(-CH3)=N-0-(C1-4 alkyl)-COOH, -C(-CH3)=N-0-(C1-4 alkyl)-CO-0(d-4 alkyl), halogen, -CF3, -CN, or heterocycloalkyl.
The compound for use according to any of items 1 to 8, wherein X is S, and further wherein L is -CO-NH- wherein the nitrogen atom comprised in said -CO-NH- is attached to ring A.
The compound for use according to item 1 , wherein said compound is a compound of one of the formulae 1a to 1t, or a pharmaceutically acceptable salt, solvate or prodrug thereof:
A pharmaceutical composition comprising a compound as defined in any of Items 1 to 10 or a pharmaceutically acceptable salt, solvate or prodrug thereof, and a pharmaceutically acceptable excipient, for use in the treatment or prevention of a cardiovascular disorder, cardiac hypertrophy, heart failure, anxiety, pain, chronic pain, migraine, an allergy, an inflammatory disorder, an autoimmune disorder, diabetes, diabetic complications, diabetic retinopathy, diabetic nephropathy, diabetic cardiomyopathy, diabetic neuropathy, cancer, metastatic cancer, drug-resistant cancer, stomach cancer, lung cancer, thyroid cancer, colon cancer, breast cancer, bipolar disorder, stroke, alopecia, or alcoholism.
A method of treating or preventing a disorder or condition selected from the group consisting of a cardiovascular disorder, cardiac hypertrophy, heart failure, anxiety, pain, chronic pain, migraine, an allergy, an inflammatory disorder, an autoimmune disorder, diabetes, diabetic complications, diabetic retinopathy, diabetic nephropathy, diabetic cardiomyopathy, diabetic neuropathy, cancer, metastatic cancer, drug-resistant cancer, stomach cancer, lung cancer, thyroid cancer, colon cancer, breast cancer, bipolar disorder, stroke, alopecia, and alcoholism, the method comprising the administration of a compound as defined in any of items 1 to 10 or a pharmaceutically acceptable salt, solvate or prodrug thereof, or the pharmaceutical composition of item 1 1 . to a subject in need thereof. 13. The compound for use according to any of items 1 to 10, or the pharmaceutical composition for use according to item 1 1 , or the method of item 12, wherein said compound or said pharmaceutical composition is to be administered by any one of: an oral route; topical route, including by transdermal, intranasal, ocular, buccal, or sublingual route; parenteral route using injection techniques or infusion techniques, including by subcutaneous, intradermal, intramuscular, intravenous, intraarterial, intracardiac, intrathecal, intraspinal, intracapsular, subcapsular, intraorbital, intraperitoneal, intratracheal, subcuticular, intraarticular, subarachnoid, intrasternal, intraventricular, intraurethral, or intracranial route; pulmonary route, including by inhalation or insufflation therapy; gastrointestinal route; intrauterine route; intraocular route; subcutaneous route; ophthalmic route, including by intravitreal, or intracameral route; rectal route; or vaginal route.
14. The method of item 12 or 13, wherein said subject is a human.
15. A compound having one of the formulae 1a or 1j to it or a pharmaceutically acceptable salt, solvate or prodrug thereof.
16. A compound having one of the formulae 1a, 1d, 1 g, 1 h or 1j to 1t or a pharmaceutically acceptable salt, solvate or prodrug thereof for use as a medicament.
17. A pharmaceutical composition comprising a compound as defined in item 16 or a pharmaceutically acceptable salt, solvate or prodrug thereof, and a pharmaceutically acceptable excipient.
The invention is also described by the following illustrative figures. The appended figures show:
Figure 1 : Structure-based modeling of the mimic of the PKCe protein fragment EAVSLKPT (see Example 1 ). (A) X-ray structure of PKCe (PDB 1 gmi). The protein backbone is illustrated as cartoon, while the EAVSLKPT is highlighted as sticks in a light grey box. (B) Extracted protein fragment EAVSLKPT in its conformation observed in the X-ray structure. Hydrogen bond donor and acceptor features (dark grey spheres) as well as the hydrophobic interaction area (light grey sphere) are indicated for the pharmacophore model that retrieved compound 1 c (PKCe16) as a hit. Figure 2: Effects of compounds 1 a (PKCe2138) and 1 b (PKCe141 ) in vitro and in vivo (see Examples 2 and 3). (A) Compound 1 b (referred to as "compound 141 ") prevents the in vitro interaction of ΡΚΟε with RACK2 in a dose-dependent manner. (B) Compound 1 b (PKCe141 ) inhibits the phosphorylation of Elk-1 in PathDetect HeLa HLR cells. Luciferase activity following activation of Elk-1 is shown. Data shown are the means (+/-SD) of 3 independent experiments. (C) Compound 1 b (PKCe141 ) does not prevent the in vitro interaction between ΡΚΟβΙΙ and RACK1 . (D) Compound 1 a (PKCe2138) inhibits the phosphorylation of Elk-1 . (E) Compound 1 a (PKCe2138) prevents RACK2-binding to PKCs in vitro in a dose-dependent manner. (F) Inhibition of RACK2-binding to PKC in vitro by EAVSLKPT which was tagged with seven arginines. A scrambled octapeptide tagged with seven arginines was used as control.
Figure 3: PKCe induces phosphorylation of Elk-1 . (A) In HeLa cells, a doxycycline-inducible constitutively active PKCe (Xuan et al., 2005) leads to phosphorylation of Elk-1 . The cells were left untreated or induced with doxycycline (2 pg/ml) for 24 hours. Additional stimulation with TPA (50 nM) was performed for 10 minutes. Phosphorylation of Elk-1 was detected with a phospho-specific antibody against the Ser383 residue of Elk-1 . GAPDH was used as loading control. (B) Densitometric analysis of Western blots. Elk-1 phosphorylation was normalized to the GAPDH loading control and data are expressed relative to untreated cells (control). Bar graphs represent quantitation of three independent experiments (+/- S.E.). Dox = doxycycline. Fold of control is calculated from - Dox -TPA.
Figure 4: Effect of compound 1 b (PKCe141 ) on PKCe and PKC5 translocation (see Example 4). IGF!- R and GAPDH were used as loading controls for the membrane and the cytosolic fraction, respectively. The means of three independent experiments were scanned and shown in a graph (+/-SD).
Figure 5: Effect of compound 1 b (PKCe141 ) on PKC isozymes. 50 μΜ compound 1 b were used in a PKC assay as described in Examples 4 and 6.
Figure 6: Cell proliferation following treatment with compound 1 b (PKCe141 ). Cell proliferation was determined as described in Example 5. The means (+/-SD) of three independent experiments, in which three samples were taken within each experiment, are shown. Figure 7: Effect of compound 1 b (PKCe141 ) on PKCe translocation by immunofluorescence. PC-3 cells were employed for these experiments. TPA induces translocation of PKCe to the plasma membrane. Compound 1 b inhibits this TPA-induced translocation of PKCe. Experiments 1 , 2 and 3 are three independent experiments.
Figure 8: PKCe-induced migration of HeLa cells into a gap. The expression of constitutively active PKCe was induced by doxycycline for 24 hours after a scratch was made into monolayer cells with a pipette tip. Figure 9: Inhibition of angiogenesis in vitro (see Example 7). (A) Spheroid sprouting assay. Bortez = bortezomib; DMF = dimethyl fumarate; 141 = compound 1 b (PKCe141 ); 2138 = compound 1a (PKCe2138). (B) Chicken egg assay. 141 = compound 1 b (PKCe141 ).
Figure 10: 1H-NMR spectrum (A) and mass spectrum (B) of compound 1a (PKCe2138).
The invention will now be described by reference to the following examples which are merely illustrative and are not to be construed as a limitation of the scope of the present invention. EXAMPLES
Compound 1 c (PKCe16; 3-amino-N-(6-ethoxy-2-benzothiazolyl)-7-methoxy-thieno[2,3- b]quinoline-2-carboxamide; Asinex ID: ASN 02538754; CAS 552819-38-2) and compound 1 b (PKCe141 , N-(3-acetyiphenyl)-9-amino-2,3-dihydro-1 ,4-dioxino[2,3-g]thieno[2,3- b]quinoline-8-carboxamide; Asinex ID: ASN 05545158, CAS 602293-00-5) were purchased from Asinex Ltd, Moscow, Russia. 25 mM stock solutions were prepared in DMSO. Identity and purity (>95%) of compound 1 b were determined using thin-layer chromatography and LC-MS (liquid chromatography-mass spectrometry). IC50 values were determined with CalcuSyn (Biosoft Cambridge, UK).
Example 1 : Molecular Modeling
A pharmacophore model was derived from the PKCe protein fragment EAVSLKPT using its conformation observed in the crystal structure of PDB entry 1 gmi (Figs. 1A and 1 B). Catalyst (version 4.1 1 , Accelrys inc., San Diego, CA, USA) was employed for pharmacophore modeling and subsequent screening of commercial molecular libraries. The pharmacophore model consists of three hydrogen bond donor/acceptor features and one hydrophobic feature. The latter attempts to resemble the hydrophobic area presented to RACK2 by Val16, while the hydrogen bond donor/acceptor features characterize the side chain properties of Ser17, Lys19, and Glu14. The rationale behind selecting those four features was their solvent exposure and, hence, their likelihood to be involved in the interaction with RACK2. In order to account (as far as possible) for protein flexibility, without becoming overly unspecific, the tolerance spheres of the pharmacophore feature and projection points were set to 1.5 and 2.0 Angstroms, respectively. Screening of the Asinex Gold and Platinum libraries (Asinex Ltd, Moscow, Russia) resulted in a total of 468 identified candidate molecules (hit structures). The hit list was further refined according to the geometric fit value of the candidate molecules to the pharmacophore model (using the Catalyst-internal scoring function) and by visual inspection. Sixteen of the most promising candidate molecules were purchased from Asinex and one of them, i.e. compound 1 c (PKCe16; 3-amino-N-(6-ethoxy-2-benzothiazolyl)-7- methoxy-thieno[2,3-b]quinoline-2-carboxamide; Asinex ID: ASN 02538754; CAS 552819- 38-2), was found to significantly disrupt the PKCe/RACK2 interaction (see also Example 2 below). Compound 1 c (PKCe16) was subsequently used as a template for the search of structurally related compounds available from commercial molecular libraries. During this iterative optimization process (PKCe22 to PKCe145), compound 1 b (PKCe141 ; (N-(3-acetylphenyl)- 9-amino-2,3-dihydro-1 ,4-dioxino[2,3-g]thieno[2,3-b]quinoline-8-carboxamide; Asinex ID: ASN 05545158; CAS 602293-00-5) was identified as the strongest disruptor/inhibitor of the PKCe/RACK2 interaction. This screening process was followed by custom synthesis and testing of a series of further compounds related to compound 1 b (PKCe2019 to PKCe2145).
Example 2: Inhibition of PKCe/RACK2 interaction in vitro
PKCe/RACK2 in vitro binding assay: Recombinant RACK2 tagged with maltose-binding protein (RACK2-MBP) was purified on columns with amylose resin (New England Biolabs) as described by the manufacturer. The recombinant protein was analyzed by Coomassie Blue staining and Western blotting after SDS-PAGE. Aliquots were stored in liquid nitrogen. The interaction between PKCe and RACK2 was measured using an ELISA-based assay. 96-well EIA/RIA high binding plates (Costar) were coated with 100 ng recombinant PKCe (ProQinase) in buffer A (20 mM Tris-HCI/100 m NaCI, pH 7.5) at 4°C on a shaker with gentle agitation overnight. The plate was washed twice with 225 μ l/well buffer A. After blocking of unspecific binding sites with 225 μΙ sterile-filtered 3 % bovine serum albumin (BSA; Sigma) in buffer A at room temperature for 3 h, the plate was washed twice with 225 μΙ of this buffer. PKCe was left untreated or activated by addition of 60 pg/ml phosphatidylserine (Sigma) and 100 nM TPA (Sigma) in a volume of 50 μΙ buffer A for 10 min at 30°C. Recombinant purified RACK2-MBP was either left untreated or incubated with EAVSLKPT-R7 or compound 1 b (PKCe141 ) at room temperature for 30 min in a final volume of 50 μΙ buffer A. 500 ng RACK2-MBP was added to untreated or activated PKCe for 1 hour at room temperature for binding. The plate was washed twice with 225 μΙ buffer A. 100 μΙ RACK2-specific rabbit anti-RACK2 polyclonal antibody (Prof. F. Wieland, University of Heidelberg, Germany) diluted 1 :20,000 in 3 % BSA/buffer A was added for 1 h at room temperature. The plate was subsequently washed 3 times with 225 μΙ buffer A and a goat anti-rabbit HRP-conjugated IgG (Santa Cruz Biotechnology) diluted 1 :20.000 in 100 μΙ 3 % BSA/buffer A was added for 1 h at room temperature. After 3 washes with 225 μΙ buffer A, 100 μΙ of ABTS substrate (0.5 mg/ml) diluted in ABTS buffer (Roche) was added and the plate was incubated in the dark for 30- 80 min. Color development was measured on a plate reader at a wavelength of 420 nm. Compound 1a (PKCe2138) was tested in a similar manner.
PKCpil/RACK1 in vitro binding assay: This assay was similar to the PKCE/RACK2 binding assay described above. 6-his-tagged RACK1 was cloned into a pET-30a(+) vector (Novagen) and purified with Ni-NTA Agarose (Qiagen). Final elution was performed with 500 nM imidazole in elution buffer (20 mM Tris-HCI/300 mM NaCI, 20% glycerol, pH 7.5). The integrity of purified recombinant protein was analyzed by Coomassie Blue staining and Western blotting. 200 ng RACK1 was coated onto 96-well EIA/RIA high binding plates (Costar) at 4°C on a shaker with gentle agitation overnight. Compound 1 b (PKCe141 ) was added at room temperature for 30 min. 500 ng of recombinant GST-tagged ΡΚΟβΙΙ (ProQinase) was activated with CaCI2, phosphatidylserine and TPA for 10 min. The RACK1 - coated plates with and without compound 1 b were incubated for 1 h with activated ΡΚΟβΙΙ. The interaction was determined with a primary rabbit anti-GST antibody (Santa Cruz) and corresponding HRP-conjugated goat anti-rabbit HRP conjugated secondary antibody (Santa Cruz) as described above for the PKCe/RACK2 interaction. Results: Compound 1 b (PKCe141 ) led to a dose-dependent inhibition of the PKCe/RACK2 interaction and exhibited an IC50 of 5.9 μΜ, as also shown in Fig. 2A. Corresponding results were obtained with compound 1a (PKCe2138) which was likewise found to inhibit the PKCe/RACK2 interaction in a dose-dependent manner (see Fig. 2E), with an IC50 of 4.1 μΜ. The peptide EAVSLKPT-RRRRRRR was used as a control. Seven arginines were added to EAVSLKPT to increase internalization of the peptide into intact cells in in vivo experiments. ICso for the inhibition of the PKCe/RACK2 interaction by EAVSLKPT-RRRRRRR in this assay was 1.02 μΜ (Fig. 2F). It has been shown previously that ΡΚΟβΙΙ interacts with the adaptor protein RACK1 (Ron et al., 1999). Therefore, it was investigated whether compound 1 b can also prevent the PKCpil/RACK1 interaction. As shown in Fig. 2C, compound 1 b does not prevent the PKCpil/RACK1 interaction, indicating specificity of this compound for the PKCe/RACK2 interaction.
These results indicate that compounds of formula (I), such as, e.g., compounds 1 a and 1 b, selectively inhibit PKCe signaling by preventing the interaction between PKCe and its adaptor protein RACK2. The compounds of the invention are therefore suitable as therapeutic agents for the treatment or prevention of diseases/disorders related to PKCe signaling, as also described herein above.
Further compounds according to the invention, including further compounds of formula (I), have been tested in the above-described PKCe/RACK2 in vitro binding assay and have likewise been found to inhibit PKCs signaling, as shown in the following Table 1 :
Compound Inhibition (% of SD SEM n
untreated control)
Mean
Compound 1 c (PKCe16) 37.7 24.9 14.4 12
PKCe22 93.3 15.7 3.9 12
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Compound 1d (PKCe119) All 8.9 2.2 16
Figure imgf000040_0001
Compound 1e (PKCe123) 32.4 8.7 2.2 16
PKCe125 48.7 11.5 2.9 16
Compound 1f (PKCe127) 60.8 13.2 3.3 16
NH,
Compound 1g (PKCe129) 60.5 13.0 3.3 16
29.6 9.9 2.5 16
32.9 7.3 2.1 12
Figure imgf000040_0002
Compound 1 b (PKCe141 ) 13.3 6.1 0.7 80
(at a concentration
of 25 μΜ)
23.6 10.2 1.5 48
(at a concentration
of 10 μΜ)
H.C
PKCe143 34.4 5.4 1.5 12
47.3 6.2 1 .8 12
82.1 14.2 4.1 12
Figure imgf000041_0001
90.4 23.2 6.7 12
Figure imgf000041_0002
PKCe2021 69.9 12.5 3.6 12
"~-0 u
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Compound 1 r (PKCe2144) 58.0 7.3 3.7 4
Figure imgf000046_0001
Compound 1s (PKCe2145) 57.6 19.6 9.8 4
Figure imgf000046_0002
Table 1 : Inhibition of PKCe signaling. The inhibition of the PKCE/RACK2 interaction is expressed as a percentage of untreated control. Accordingly, a value of, e.g., 37.7% denotes an inhibition of the PKCE/RACK2 interaction to 37.7% of the untreated control (which is set to 100%). Compounds having a percentage inhibition of 100% or greater were not found to inhibit PKCe signaling in this assay. The compounds PKCe16 to PKCe145 were employed at a concentration of 25 μΜ, while the compounds PKCe2019 to PKCe2145 were tested at a concentration of 10 μΜ; compound 1 b (PKCe141 ) was tested at both 25 μΜ and 10 μΜ. SD = standard deviation; SEM = standard error of the mean; n = number of samples tested.
Example 3: Inhibition of PKCe-induced Elk-1 phosphorylation in vivo
PKCE is situated in the signal transduction cascade upstream of Raf-1 (Xuan et al., 2005). In a HeLa cell line containing a doxycycline-inducible constitutively active PKCE (Garczarczyk et al., 2009), active PKCE leads to phosphorylation of the transcription factor Elk-1 (see Fig. 3). In order to obtain information whether compound 1 b (PKCe141 ) is able to prevent the PKCE/RACK2 interaction in intact cells, compound 1 b was tested for inhibition of Elk-1 phosphorylation in a PathDetect HeLa Luciferase (HLR) frans-reporting HeLa cell line, as described in the following. In these cells, activation of PKC by 12-O-tetradecanoyl phorbol-13-acetate (TPA) leads to the expression of luciferase. Compound 1 a (PKCe2138) was tested in a similar manner. Cells and cell proliferation: HeLa cells containing a tetracycline/doxycycline-inducible constitutively active PKCE were described previously (Garczarczyk et al,, 2009). A Path Detect HeLa HLR cell line was obtained from Agilent. PC-3 prostate adenocarcinoma cells were obtained from Dr. Helmut Klocker, Department of Urology, Innsbruck Medical University. For cell proliferation HeLa HLR and PC-3 cells were seeded at -10,000 cells per well in 96-well plates. After 4 h various concentrations of compound 1 b (PKCe141 ) were added and left for 72 h. Cell proliferation was determined by the SRB-assay (Skehan et al., 1990). Elk-1 phosphorylation: Elk-1 phosphorylation was determined with the PathDetect System (Agilent). PathDetect HeLa HLR-ELK-1 cells contain a luciferase reporter cassette and express a unique, stably integrated, irans-acting fusion protein. The fusion protein consists of the activation domain of the Elk-1 transcriptional activator (Rao et al., 1989; Price et al., 1995; Marais et al. , 1993), that is fused to the yeast GAL4 DBD (residues 1-147). The transcriptional activator domain of Elk-1 is activated. 200,000 PathDetect HeLa HLR-Elk-1 cells per well were seeded in a 6-well plate and grown for 24 hours. Cells were washed with phosphate buffered saline and starved for 16 hours in starvation medium (DMEM-containing 0.5% fetal bovine serum and 1 % glutamine). Compounds were added in DMEM for 30 min. Following treatment with 50 nM TPA for 5 min the cells were washed twice with phosphate buffered saline and incubated for 4 h in starvation medium and compound 1 b (PKCe141 ). 200 μΙ lysis buffer as described by the manufacturer was added. The plates were shaken intensively at 4°C for 20 min. Lysates were collected and centrifuged at 1 1 ,000 x g at 4°C for 2 min and used immediately for luciferase activity measurement. Protein concentration was determined according to Bradford (Bradford, 1976) and 20 μg of each sample was transferred to a white, opaque 96-well plate. 150 μΙ luciferase assay buffer as described by the manufacturer was injected and light emission from the reaction was measured for 3 seconds after a delay time of 2 sec. Relative light units were measured with a 1450 Microbeta Wallac Jet Luminometer (Perkin-Elmer). Results: Compound 1 b (PKCe141 ) inhibited the phosphorylation of Elk-1 in a dose dependent manner with an IC60 of 1 1.2 μΜ in intact cells, as also shown in Fig. 2B. Compound 1a (PKCe2138) was likewise found to inhibit the phosphorylation of Elk-1 (see Fig. 2D). These results confirm that compounds of formula (I), such as compounds 1 a and 1 b, inhibit PKCe signaling in vivo and can thus be used as therapeutic agents in the intervention of diseases/disorders in which PKCe signaling is implicated, as described herein above. Example 4: Effect on ΡΚΟε translocation Upon activation, PKCe associates with RACK2 and is translocated from the cytosolic to the membrane fraction (Brodie et al., 1999; Fenton et al., 2009). It was investigated whether compound 1 b (PKCe141 ) also inhibits the translocation of PKCe to the membrane.
Cell fractionation and Western blotting: For PKCe translocation PC-3 cells were starved for 16 h, treated with compound 1 b (PKCe141 ) for 30 min, stimulated with 50 nM TPA for 5 min, lysed and fractionated with a CNMCS/CNM compartmental protein extraction kit (BioChain Institute). Western Blotting was performed by a standard procedure as described by Garczarczyk et al., 2009 and Garczarczyk et al., 2010. Cytosolic and membrane fractions were loaded onto SDS gels and transferred to an Immobilon membrane (Millipore). The membranes were incubated with rabbit polyclonal IgG antibodies for detection of PKCe (Santa Cruz Biotechnology, dilution 1 :2,000), of PKCe phosphoSer729 (Millipore, dilution 1 :1 ,000) and of PKC5 (Santa Cruz; dilution 1 :1 ,000). For the loading control and as marker for the membrane fraction, an IGFI-βΡν rabbit polyclonal IgG antibody (Santa Cruz Biotechnology; dilution 1 : 1 ,000) and as secondary antibodies peroxidase-conjugated AffiniPure Goat Anti-Rabbit IgG (Jackson Immuno Research Laboratories, dilution 1 :20,000) were used. GAPDH (Chemicon; 1 : 10,000) was used as loading control for the cytosolic fraction. A peroxidase-conjugated secondary antibody (AffiniPure Goat Anti-Mouse IgG; Jackson Immuno Research Laboratories, dilution 1 :20,000) was employed for detection. In vitro PKC assay: 150 ng of recombinant purified PKC isozyme (see Fig. 5) were combined with 10 μΙ 10 x kinase assay buffer (200 mM Tris-HCI , pH 7.5, 200 mM MgCI2), phosphatidylserine (final 10 μΜ), TPA (final 1 μΜ) and substrate peptide RFARKGSLRQKNV (Alexis) (final 50 μΜ) in a volume of 100 μΙ. 90 μΙ/well were pipetted in a 96-well plate and incubated for 1 min at 30°C. 10 μΙ ATP mix containing 0.4 μΙ of 10 mM ATP 1 μθϊ γ-33Ρ-ΑΤΡ (PerkinElmer). After another incubation step for 10 min at 30°C the plate was transferred on ice to stop the kinase reaction. 50 μΙ of the kinase assay-mix are added onto phosphocellulose sheets (Whatman) in a 6-well plate. The sheets were washed 3 times with 1.5% H3P04 and twice with distilled water. The phosphocellulose sheets are transferred to scintillation vials, 4 ml of scintillation fluid (Ultima Gold, Perkin-Elmer) were added and counted in a β-counter. The effect of compound 1 b (PKCe141) on the different PKC isozymes is shown in Fig. 5. Results: In PC-3 prostate adenocarcinoma cells, treatment with TPA led to an increase of PKCe in the membrane fraction. Accordingly, the amount of PKCe in the cytosol decreased (Fig. 4). Furthermore, activated PKCe is phosphorylated at Ser729. TPA increased the phosphorylated form of PKCe. Compound 1 b (PKCe141 ) does not inhibit PKCe in vitro, as also shown in Fig. 5. Treatment of cells with compound 1 b similarly did not reduce Ser729- phosphorylated PKCe at the membrane (Fig. 4). This result illustrates that PKCe is phosphorylated following treatment with TPA, and compound 1 b does not prevent this phosphorylation. However, compound 1 b partially inhibits the cellular translocation of activated and phosphorylated PKCe to the membrane fraction. PKC5 shows a high degree of homology with PKCe. Therefore, the influence of compound 1 b on PKC5 was investigated. As shown in Fig. 4, compound 1 b does not decrease PKC5 in the membrane. A similar result was obtained with immunocytochemistry (Fig. 7). Short-term treatment with TPA led to an increase of PKCe in the plasma membrane. Compound 1 b prevented the TPA-induced PKCe translocation to the plasma membrane, as shown in Fig. 7.
Compound 1 b has thus been demonstrated to inhibit the TPA-induced translocation of PKCe, but not that of PKC5, from the cytosol to the membrane. These results indicate that compounds of formula (I), such as compound 1 b, allow for an isozyme-specific inhibition of PKCe signaling, which makes them particularly advantageous as therapeutic agents in the treatment of prevention of diseases/disorders associated with PKCe signaling.
Example 5: Effects on cell proliferation and cell migration
As shown above, compound 1 b (PKCe141 ) exhibits the features of an inhibitor of PKCe signaling in vitro and also in intact cells. A major question is whether such an inhibitor affects cell proliferation or, in other words, whether it is toxic. Therefore, compound 1 b was tested for inhibition of cell proliferation in HeLa-HLR and PC-3 cells. These cells were employed for Elk-1 phosphorylation and for the PKCe translocation experiments described above.
Immunofluorescence: PC-3 cells were grown on glass coverslips coated with poly-L-lysine (Sigma). After treatment with compound 1 b (PKCe141 ) for 30 min and with 100 nM TPA for 5 min, the cells were rinsed twice with phosphate buffered saline (PBS) and fixed with filter sterilized 4% (w/v) paraformaldehyde/4% sucrose (w/v) (both from Sigma) in PBS at room temperature for 10 min. Subsequently the cells were washed three times with PBS and permeabilized with 0.2% Triton X-100/0.2% IgG-free BSA in PBS at room temperature for 10 min. After blocking with 5% normal goat serum diluted in PBS as described above for 30 min, cells were incubated with the primary antibodies for PKCE (Santa Cruz; diluted 1 :500) in 0.2% Triton X-100/0.2% IgG-free BSA in PBS at 4°C overnight. Subsequently, the cells were washed three times with the same buffer and incubated with the labeled secondary antibody (Alexa Fluor, Invitrogen, 1 :4,000) at room temperature for 1 h. After three more washes with 0.2% Triton X-100/0.2% IgG-free BSA in PBS, cells were mounted with Mowiol (Sigma) and images were taken with an Olympus BX 50 optical microscope (Olympus).
Cell migration: For cell migration and motility, a scratch migration assay described by Cha et al. , 1996 was employed. In a tissue culture dish, with logarithmically growing HeLa cells containing a doxycycline-inducible constitutively active PKCe, a migration gap of approximately 1 mm was created by introducing a 'scratch' to the adherent layer of cultured cells using a sterile Gilson 200 μΙ pipette tip. The scratch was administered by hand with sufficient pressure to remove adherent cells from the polystyrene substrate, but without causing a physical damage to the polystyrene surface. The dish was washed with PBS to remove the cells and further incubated with 2
Figure imgf000050_0001
doxycyclin and 25 μΜ compound 1 b (PKCe141 ) for 24 hours. Controls were left either untreated or were incubated with 2 pg/ml doxycyclin and DMSO. Migration into the scratch was observed with an Olympus microscope.
Results: As shown in Fig. 6, in both of the cell lines even 50 μΜ of compound 1 b (PKCe141 ) did not show any inhibition of cell proliferation. It has been shown previously that PKCe does not increase cell proliferation. However, it increases cell migration (Akita, 2008; Garczarczyk et al. , 2009) and it is associated with metastatic spread and invasiveness of human cancer cells (Stensman et al. , 2008). Therefore, it was investigated whether PKCE inhibits migration. As shown in Fig. 8, untreated HeLa cells containing a constitutively active doxycycline-inducible PKCe showed only low migration into a scratch made with a pipette tip on a tissue culture dish. If the scratch was made into the monolayer cells and the expression of constitutively active PKCe was induced by doxycycline after 24 hours a significant part of the scratch was covered with cells. PKCe increased migration of cells into a gap and compound 1 b (PKCe141 ) indeed inhibited this PKCe-induced migration (Fig. 8), which indicates its potential as an inhibitor of metastasis. Moreover, these data also indicate that compound 1 b does not exhibit toxicity, which is a particularly favorable characteristic for therapeutic application as an inhibitor of PKCe signaling and, accordingly, as a medicament to treat diseases such as mycardial hypertrophy (Takeishi et al. , 2000; Ferreira et al., 2010), diabetes (Ikeda et al. , 2001 ), stroke, Alzheimer's disease or pain (Akita, 2008; Shirai et al., 2008).
Example 6: Inhibition of kinases
In earlier studies it was found that the barbituric acid derivative PKCe97 (BAS 02104951 ) prevented the PKCe/RACK2 interaction (IC50 = 28.5 μΜ). In addition, this compound also inhibited PKCn and PKCe directly (Gruber et al. , 201 1 ). Compound 1 b (PKCe141 ) was therefore tested for its inhibition of PKC isozymes.
In vitro PKC assay: 150 ng of recombinant purified PKC isozyme (see Fig. 5) were combined with 10 μΙ 10 x kinase assay buffer (200 mM Tris-HCI , pH 7.5, 200 mM MgCI2), phosphatidylserine (final 10 μΜ), TPA (final 1 μΜ) and substrate peptide RFARKGSLRQKNV (Alexis) (final 50 μΜ) in a volume of 100 μΙ, 90 μ l/we 11 were pipetted in a 96-well plate and incubated for 1 min at 30°C. 10 μΙ ATP mix containing 0.4 μΙ of 10 mM ATP 1 pCi Y-33P-ATP (PerkinElmer). After another incubation step for 10 min at 30°C the plate was transferred on ice to stop the kinase reaction. 50 μΙ of the kinase assay-mix are added onto phosphocellulose sheets (Whatman) in a 6-well plate. The sheets were washed 3 times with 1 .5% H3P04 and twice with distilled water. The phosphocellulose sheets are transferred to scintillation vials, 4 ml of scintillation fluid (Ultima Gold, Perkin-Elmer) were added and counted in a β-counter. The effect of compound 1 b (PKCe141 ) on the different PKC isozymes is shown in Fig. 5. As shown therein, 50 μΜ of compound 1 b (PKCe141 ) inhibited several PKC isozymes to approximately 65% of controls. If compared to the inhibition of the PKCE/RACK2 interaction (IC50 = 5.90 μΜ), however, this is not a major effect. Furthermore, a screen of 109 kinases showed that many kinases are not or only slightly affected by compound 1 b (PKCe141 ). ERK1 , NUAK1 , PIM3, BTK and RSK2 are the kinases inhibited most by compound 1 b, as also shown below in Table 2. 25 μ PKCe141
% of control SD
Control 100 0
ERK1 55 5
RSK2 27 1
NIJA 1 49 6
PIM3 46 4
BTK 42 1
Table 2: Profile of kinase inhibition by 25 μ compound 1 b (PKCe141 ). 109 different protein kinases were tested for their inhibition by compound 1 b (PKCe141 ). The five kinases inhibited most are shown in the table. All other kinases were affected less. Screening was performed by the National Centre for Protein Kinase Profiling, Division of Signal Transduction Therapy, University of Dundee. The data is portrayed as mean % activity and standard deviation (SD) of assay duplicates.
Compound 1 b (PKCe141 ), however, affected these kinases less than the PKCE/RACK2 interaction. As shown in Fig. 2A, 25 μΜ compound 1 b (PKCe141 ) inhibited the PKCE/RACK2 interaction to approximately 10% of untreated controls, whereas 25 μΜ of compound 1 b inhibited the most affected kinase RSK2 to 27%, as shown in Table 2 above. All other kinases were less affected. This level of selectivity is quite acceptable.
Example 7: Inhibition of angiogenesis Compounds according to the invention were tested for their effect on angiogenesis in vitro in a spheroid sprouting assay and in a chicken egg assay.
Spheroid sprouting assay (Korff et al., 1 999): Human umbilical vein endothelial (HUVEC) spheroids where generated overnight in hanging-drop culture consisting of 400 cells in EBM-2 medium, 2% FCS and 20% methylcellulose (Sigma Biochemicals). Spheroids were embedded in collagen type I from rat tail (Becton Dickinson) and stimulated with 50 ng/ml VEGF (Sigma Biochemicals) in the presence or absence of 5 ml solution containing the corresponding test compound (see Fig. 9A). Sprouts were analyzed by inverted transmission-microscopy (Zeiss Axiovert 200 M) and documented by digital imaging (Axiovision Software, Zeiss). The cumulative sprout length (CSL) was analyzed after printing of high quality pictures and counting by two independent blinded observers.
Chicken egg assay: Eggs from hen are incubated at 37°C for 3 days, opened and incubated for further 7 days. On day ten the growth factor VEGF and compound 1 b (PKCe141 ) are added at the concentrations indicated in Fig. 9B. After further incubation for 5 days angiogenesis is observed by microscope.
Results: As shown in Fig. 9A, compound 1a (PKCe2138) was found to considerably reduce VEGF-induced sprouting from the HUVEC spheroids at a concentration of only 250 nM. Compound 1 b (PKCe141 ) was further tested in a chicken egg angiogenesis assay. In this assay, blood vessels were observed after the addition of VEGF to the eggs but not after the combined addition of VEGF and compound 1 b (see arrows in Fig. 9B). This data indicates that compounds according to the invention, including compounds 1a and 1 b, inhibit angiogenesis and can thus be particularly useful, e.g., in the treatment or prevention of cancer.
Example 8: Preparation of compounds according to the invention
Compounds 1a, 1j, 1 k, 1o, 1 p, 1 q, 1 r, and 1s (PKCe2138 to PKCe2145) were prepared as illustrated in Scheme 1 with reference to compound 1j (PKCe2139) and as further described in the following.
Figure imgf000054_0001
Figure imgf000054_0002
Scheme 1 : Synthesis of compounds 1j (PKCe2139) and PKCe2020.
Ac20 = acetanhydride; Py = pyridine; DMF = dimethylformamide; POCI3 = phosphoryl chloride; NH2OH = hydroxylamine; EtOH = ethano!; SOCI2 = thionyi chloride; NH2CSNH2 = thiourea; 1 -PrOH = propanol-1 ; MeONa = sodium methylate; MeOH = methanol; AcONa = sodium acetate.
Acetanilide (2): A solution of amine 1 (1 eq) in pyridine was cooled to 5°C and acetanhydride (1 .25 eq) was added dropwise with stirring. The mixture was stirred for 45 min at 60°C and poured into ice-water and stirred for 30 min at 0-10°C. The precipitate was filtered off, washed with water (100 ml) and dried.
Chloroquinolinecarbaldehyde (3) in phosphoryl chloride: Dimethylformamide (9.13 g, 9.6 ml, 0.125 mol) was cooled to 0°C in a flask equipped with a drying tube and phosphoryl chloride (53.7 g, 32 2 ml, 0.35 mol) was added dropwise with stirring. To this solution was added the acetanilide 2 (0.05 mol) and after 5 min the solution was heated to 75°C for 12- 16 h. The reaction mixture was poured into ice-water (300 ml) and stirred for 30 min at 0- 10°C. The chloroquinolinecarbaldehyde 3 was filtered off, washed with water (100 ml) and dried. The chloroquinolinecarbaldehyde 3 was crystallized with ethylacetate. Hydroxylimine (A): Hydroxylamine hydrochloride (1 .25 eq) was added to the suspension of chloroquinolinecarbaldehyde 3 in ethanol and this mixture was refluxed for 6 h. The hydroxylimine 4 was filtered off, washed with ethanol and dried. Chloro-3-cyano-quinoline (5): The mixture of the hydroxylimine 4 (1 eq) in DMF was cooled to 0°C in a flask equipped with a drying tube and thionyl chloride (1 .5 eq) was added dropwise with stirring and cooling for 10-15 min. Then this mixture was stirred overnight at room temperature. The reaction mixture was poured into ice-water and stirred for 30 min at 0-10°C. The nitrile 5 was filtered off, washed with water and dried. Raw material was purified by flash-chromatography on silica gel with chloroform and hexane (4: 1 ).
2- Thio-3-cyano-quinoline (6): A mixture of 2-chloro-3-cyano-quinoline 5 (1 eq) and thiourea (3 eq) in propanol-1 was heated under reflux for 6 h, then this mixture was cooled and sodium hydroxide (10% 10 eq) was added. The reaction mixture was cooled, acidified with concentrated HCI and the solid product was filtered off and dried.
3- Amino-thieno[2,3-b]quinoline-2-carboxylamide (PKCe2139): A mixture of the 2-thio-3- cyano-quinoline 6 (1 eq), the respective halo compound (1 .25 eq) and sodium methylate (3 eq) in absolute methanol was refluxed for 1 h. The formed solid was collected by filtration, washed several times with methanol and finally with water and recrystallized from suitable solvent (DMF-methanol). The compounds 1 a, 1j, 1 k, 1 o, 1 p, 1 q, 1 r, and 1 s according to the invention (i.e. , PKCe2138 to PKCe2145) were prepared in this manner.
The identity of compound 1 a (PKCe2138) was confirmed by H-NMR and mass spectrometry, as also shown in Figs. 10A and 10B.
Compound 1 a (P Ce2138): 1 H-NMR, DMSO-D6, δ: 2.53 (3H, s, CH3), 4.41 (1 H, dd, J 3, 3, -CH2-CH2-), 7.44 (2H, s, H-5, H-8), 7.62 (2H, s, NH2), 7.91 (4H, dd, J 3, 3, arom), 8.89 (1 H, s, H-4), 9.68 (1 H, s, HN-C=0). MW Calc. for C22H17N304S 419.0938. Found in ESI-ms 420.1047 (M+H) 100%
2- [(3-Cyanoquinolin-2-yl)thio]-A/-phenylacetamide (PKCe2020): A mixture of the 2-thio-
3- cyano-quinoline 6 (1 eq), the respective halo compound (1 .25 eq) and fused sodium acetate (3 eq) in absolute ethanol was refluxed for 3 h. The formed solid was collected by filtration, washed several times with methanol and finally with water and recrystallized from suitable solvent (DMF-methanol). Compounds such as PKCe2019, PKCe2020, PKCe2021 , PKCe2088 and PKCe2089 were prepared in this manner.
Compounds 1m and 1 n (i.e., PKCe2023 and PKCe2024) were prepared as described in the following Scheme 2 with reference to compound 1 n (PKCe2024).
Figure imgf000056_0001
Scheme 2: Synthesis of compound 1 n (PKCe2024). NaH = sodium hydride; DMF dimethyl-formamide; PyBOP = benzotriazol-1 - oxytripyrrolidinophosphonium hexafluorophosphate; Et3N = triethylamine.
Thieno[2,3-b]quinoline-2-carboxylate (8): A mixture of the quinoline 3 (1 eq; see Scheme 1) with 2-mercaptoacetic acid ethyl ester (1.5 eq) in dry DMF was cooled and NaH (60% in oil , 5 eq) was added. This mixture was stirred for 30 min at 10°C and then 2 h at 60°C. After that 25% solution of NaOH (10 eq) was added dropwise and the mixture was heated under reflux for 2 h. The reaction mixture was cooled and extracted 3 times by benzene. Water layer was acidified with concentrated HCI and the solid product was filtered off and dried. Thieno[2,3-b]quinoline-2-carboxylanilide (PKCe2024): A mixture of the thieno[2,3- b]quinoline-2-carboxylate 8 (1 eq) and PyBOP (1 .3 eq) in DMF was stirred for 15 min, then appropriate aniline (1 .25 eq), catalytic amount of 4-dimethylaminopyridine (DMAP) and Et3N (2 eq) was added. The reaction mixture was stirred overnight at 50°C than it was poured into ice-water and precipitate was filtered off. Raw material was heated under reflux with methanol-DMF (3:2) and filtered off and dried. The anilide PKCe2024 was purified by flash- chromatography with CHCI3-methanol (9: 1 ). The compounds 1 m and 1 n according to the invention (i.e., PKCe2023 and PKCe2024) were prepared in this manner.
The compounds PKCe2048 to PKCe2054 were prepared as described in Lapa et ai., 2012.
PKCe2053: 3-Chloro-N-(7-methoxy-1 H-pyrazolo[3,4-b]quinolin-3-yl)benzamide. Yield 74%. 1 H-NMR, DMSO-D6, δ: 3.94 (3H, s, CH3), 7.1 1 (1 H, dd, J 2.2, 9.2, H-6), 7.29 (1 H, d, J 2.2, H-8), 7.61 (1 H, t, J 8.0, H-5'), 7.71 (1 H, d, J 8.0, H-6'), 8.02 (1 H, d, J 9.2, H-5), 8.07 (1 H, d, J 8.0, H-4'), 8.17 (1 H, s, H-2'), 8.90 (1 H, s, H-4), 1 1.27 (1 H, s, NH), 13.02 (1 H, s, HN-C=0). MW Calc. for C18H13CIN402 352.0727. Found in ESI-ms 353.0708 (M+H) 100%, 355.0773 (M+H) 30%.
REFERENCES
Akita, Y. FEBSJ. 275, 3995-4004 (2008).
Arkin, M.R. et al. Nature Rev Drug Discovery 3, 301 -317 (2004).
Beeley, N.R. Drug Discov. Today 5, 354-363 (2000).
Bohler, M. Dissertation at the University of Innsbruck. 1-208 (2006).
Bradford, M.M. Anal. Biochem. 72, 248-254 (1976).
Brodie, C. et al. Cell Growth Differ. 10, 183-191 (1999).
Castrillo, A. et al. J. Exp. Med. 194, 1231-1242 (2001 ).
Cesare, P. et al. Neuron 23, 6 7-624 (1999).
Cha, D. et al. J. Invest. Dermatol. 106, 590-597, (1996).
Chen, Y.B. et al. Expert Opin. Investig. Drugs 7, 939-944 (2008).
Choi, D.S. et al. J. Neurosci. 22, 9905-991 1 (2002).
Csukai, M. et al. J. Biol. Chem. 272, 29200-29206 (1997).
Easmon, J. et al. Int. J. Cancer 94, 89-96 (2001 ).
Erdogan, E. et al. J. Biol. Chem. 281 , 28450-28459 (2006).
Fedorov, O. et al. Proc. Natl. Acad. Sci. USA 104, 20523-2058 (2007).
Fenton, R.A. et al. Am. J. Physiol. Heart Circ. Physiol. 297, H718-H725 (2009).
Ferreira, J.C. et al. J. Mol. Cell. Cardiol, print ahead (2010).
Galeotti, N. et al. Neurotherapeutics 10, 329-339 (2013).
Garczarczyk, D. et al. Cell Signal. 21 , 745-752 (2009).
Garczarczyk, D. et al. Chem. Biol. Interact. 185, 25-32 (2010).
Gorin, M A. et al. Mol. Cancer 8, 9 (2009). Gray, M.O. et al. J. Biol. Chem. 272, 30945-30951 (1997).
Gruber, P. et al. J. Biochem. 149, 331-336 (201 1 ).
Ikeda, Y. et al. Diabetes 50, 584-92 (2001 ).
Johnson, J.A. et al. J. Biol. Chem. 271 , 24962-24966 (1996).
Korff, T. et al. J. Cell Sci. 112, 3249-3258 (1999).
Lapa, GB. et al. J. Enzyme Inhib. Med. Chem., epub ahead of print (2012). Manning, G. et al. Science 298, 1912-1934 (2002).
Marais, R. et al. Cell 73, 381-393 (1993).
Mochly-Rosen, D. et al. Proc. Natl. Acad. Sci. USA 88, 3997-4000 (1991). Mochly-Rosen, D. et al. Nat. Rev. Drug Discov. 11 , 937-957 (2012).
Nakamura, S. et al. Exp. Eye Res. 90, 137-145 (2010).
Nicholls, A. et al. J. Comput.-Aided Mol. Des. 19, 661 -686 (2005).
Pass, J.M. et al. Am. J. Physiol. Heart Circ. Physiol. 280, H946-955 (2001 ).
Perletti, G. et al. Biochem. Biophys. Res. Commun. 221 , 688-691 (1996). Pillai, A.D. et al. Mol Pharmacol. 77, 724-733 (2010).
Price, M.A. et al. EMBOJ. 14, 2589-2601 (1995).
Rao, V.N. et al. Science 244, 66-70 (1989).
Rechfeld, F. et al. Curr. Topics Med. Chem. 11 , 1305-1319 (201 1 ).
Reichling, DB. et al. Trends Neurosci. 32, 61 1 -618 (2009).
Ron, D. et al. J. Biol. Chem. 274, 27039-27046 (1999).
Sanam, R. et al. J Mol Graph Model. 28, 472-477 (2010).
Sanchez-Bautista, S. et al. Recent Pat. DNA Gene Seq. 7, 74-81 (2013).
Schechtman, D. et al. Oncogene 20, 6339-6347 (2001 ).
Shirai, Y. et al. FEBSJ. 275, 3988-3994 (2008).
Skehan, P. et al. J. Natl. Cancer Inst. 82, 1 107-1 1 12 (1990).
Soltoff, S.P. Trends Pharmacol. Sci. 28, 453-458 (2007).
Stensman, H. et al. BMC Cancer B, 365 (2008).
Takahashi, T. et al. Skin Pharmacol. Appl. Skin Physiol. 13, 133-142 (2000). Takeishi, Y. et al. Circ. Res. 86, 1218-1823 (2000).
Xuan, Y.T. et al. Circulation 112, 971 -978 (2005).

Claims

Claims
1. A compound of the following formula (I)
Figure imgf000059_0001
(I) wherein: n is an integer of 0 to 4, and each R1 is independently selected from C2-4 alkyl, C2-4 alkenyl, C2.4 alkynyl, -OH, -0(C1-4 alkyl), -0(d„4 alkyl)-OH, -0(C1-4 alkyl)-0(d_4 alkyl), -SH, -S(d.4 alkyl), -S(C1-4 alkyl)-SH, or -S(d.4 alkyl)-S(d.4 alkyl), -NH2, -NH(C1-4 alkyl), -N(C1-4 alky!)(C1-4 alkyl), halogen, -CF3, or -CN; or, alternatively, n is 2, 3 or 4, and two groups R1 attached to adjacent carbon atoms are mutually linked to form a group -0-CH2-0-, -0-CH2-CH2-0- or -0-CH2-CH2-CH2-0-, while the further group(s) R1, if present, is/are independently selected from C1-4 alkyl, C2. alkenyl, C2.4 alkynyl, -OH, -0(d-4 alkyl), -0(d_4 alkyl)-OH, -0(C1-4 alkyl)-0(d.4 alkyl), -SH, -S(d„4 alkyl), -S(d.4 alkyl)-SH, or -S(d_4 alkyl)-S(C1-4 alkyl), -NH2, -NH(C1-4 alkyl), -N(C1-4 alkyl)(d_4 alkyl), halogen, -CF3, or -CN;
R2 is selected from hydrogen, d_4 alkyl, C2.4 alkenyl, C2.4 alkynyl, -OH, -0(C -4 alkyl), -SH, -S(d.4 alkyl), -NH2, -NH(C1-4 alkyl), -N(d.4 alkyl)(d_4 aikyl), halogen, -CF3, or -CN;
R3 is selected from -NH2, -NH(d_4 alkyl), -N(C1-4 alkyl)(d.4 alkyl), -OH, -0(d_4 alkyl), -SH, -S(d_4 alkyl), or hydrogen; X is selected from S, O, N(H), or N(C1-4 alkyl);
L is -(CH2)i-4-, wherein one -CH2- unit comprised in said -(CH2)i-4- is replaced by a group selected from -CO-NH-, -C0-N(d-4 alkyl)-, -NH-CO-, -N(C1-4 alkyl)- CO-, -0-, -CO-, -NH-, -N(d-4 alkyl)-, -S-, -SO-, or -S02-;
A is aryl or heteroaryl, wherein said aryl or said heteroaryl is optionally substituted with one or more groups independently selected from C1-4 alkyl, C2.4 alkenyl, C2-4 alkynyl, -OH, -0(d-4 alkyl), -SH, -S(d-4 alkyl), -NH2, -NH(d-4 alkyl), -N(C1-4 alkyl)(d.4 alkyl), halogen, -CF3, or -CN;
R4 is selected from -CO-(C1-4 alkyl), -CHO, -0(d-4 alkyl), -OH, -0-CO-(C1-4 alkyl), -0-CO-0(C1-4 alkyl), -C0-0(d-4 alkyl), -COOH, -CO-NH2, -CO-NH-(C1-4 alkyl), -CO-N(d_4 alkyl)(C1-4 alkyl), -0-CO-NH2, -0-CO-NH-(C1-4 alkyl), -0-CO-N(C1-4 alkyl)(d.4 alkyl), -NH-CO-(C1-4 alkyl), -N(d-4 alkyl)-CO-(C1-4 alkyl), -NH-CO-0(C1-4 alkyl), -N(d.4 alkyl)-CO-0(d.4 alkyl), -NH2, -NH(C1-4 alkyl), -N(d-4 alkyi)(C -4 alkyl), d.4 alkyl, C2-4 alkenyl, C2-4 alkynyl, -(C1-4 alkyl)-CO-(d_4 alkyl), -(C1-4 alkyl)-CHO, -(C1-4 alkyl)-0(C1-4 alkyl), -(C1-4 alkyl)-OH, -(C1-4 alkyl)-0-CO-(d-4 alkyl), -(C1-4 alkyl)-0-CO-0(C1-4 alkyl), -(d-4 alkyl)-CO-0(d.4 alkyl), -(d.4 alkyl)-COOH, -(C1-4 alkyl)-CO-NH2, -(C1-4 alkyl)-CO-NH-(C1-4 alkyl), -(d_4 alkyl)-CO-N(d_4 alky!)(C1-4 alkyl), -(d_4 alkyl)-0-CO-NH2, -(C1-4 alkyl)-0-CO-NH-(C^ alkyl), -(C1-4 alkyl)-0-CO-N(C1-4 alkyl)(d.4 alkyl), -(C1-4 alkyl)-NH-CO-(C1-4 alkyl), -(d_4 alkyl)-N(C1-4 alkyl)-CO-(d_4 alkyl), -(C1-4 alkyl)-NH-CO-0(d.4 alkyl), -(d_4 alkyl)-N(d.4 alkyl)-C0-0(d_4 alkyl), -(C1-4 alkyl)-NH2, -(d.4 alkyl)-NH(C1-4 alkyl), -(C1-4 alkyl)-N(d_4 alkyl)(d.4 alkyl), -(C1-4 alkyl)=N-OH, -(C1-4 alkyl)=N-0(d_4 alkyl), -(d.4
Figure imgf000060_0001
alkyl)-COOH, -(C1- alkyl)=N-0-(d.4 alkyl)-CO-0(d.4 alkyl), halogen, -CF3, -CN, heterocycloalkyl, or hydrogen; and is a double bond or a single bond; or a pharmaceutically acceptable salt, solvate or prodrug thereof for use in the treatment or prevention of a cardiovascular disorder, cardiac hypertrophy, heart failure, anxiety, pain, chronic pain, migraine, an allergy, an inflammatory disorder, an autoimmune disorder, diabetes, diabetic complications, diabetic retinopathy, diabetic nephropathy, diabetic cardiomyopathy, diabetic neuropathy, cancer, metastatic cancer, drug-resistant cancer, stomach cancer, lung cancer, thyroid cancer, colon cancer, breast cancer, bipolar disorder, stroke, alopecia, or alcoholism.
2. The compound for use according to claim 1 , wherein n is an integer of 1 to 4, one group R1 is selected from -0(C1-4 alkyl), -OH, -0(d.4 alkyl)-OH, or -0(d_4 alkyl)-0(Ci_4 alkyl), and the remaining group(s) R , if present, is/are independently selected from C2-4 alkyl, C2.4 alkenyl, C2-4 alkynyl, -OH, -0(C1-4 alkyl), -0(d_4 alkyl)-OH, -0(C1-4 alkyl)-0(C1-4 alkyl), -SH, -S(C1-4 alkyl), -S(C1-4 alkyl)-SH, -S(C1-4 alkyl)-S(C1 -4 alkyl), -NH2, -NH(C1-4 alkyl), -N(d_4 alkyl)(C1-4 alkyl), halogen, -CF3, or -CN.
3. The compound for use according to claim 1 , wherein n is 2, 3 or 4, and two groups R1 attached to adjacent carbon atoms are mutually linked to form a group -0-CH2-0-, -0-CH2-CH2-0- or -0-CH2-CH2-CH2-0-, while the further group(s) R1 , if present, is/are independently selected from C1- alkyl, C24 alkenyl, C2.4 alkynyl, -OH, -0(C1-4 alkyl), -0(d.4 alkyl)-OH, -0(d_4 alkyl)-0(C1 -4 alkyl), -SH, -S(d„4 alkyl), -S(d_4 alkyl)-SH, -S(d.4 alkyl)-S(d-4 alkyl), -NH2, -NH(C1-4 alkyl), -N(C1-4 alkyl)(d_4 alkyl), halogen, -CF3, or -CN.
4. The compound for use according to any of claims 1 to 3, wherein R3 is -NH2.
5. The compound for use according to any of claims 1 to 4, wherein X is S.
6. The compound for use according to any of claims 1 to 5, wherein L is -CO-NH- or -CO-N(d.4 alkyl)-, wherein the nitrogen atom comprised in said -CO-NH- or said -CO-N(d_4 alkyl)- is attached to ring A.
7. The compound for use according to any of claims 1 to 6, wherein A is phenyl, 1 ,3-thiazol-2-yl or 1 ,3-benzothiazol-2-yl, wherein said phenyl, said 1 ,3-thiazol-2-yl or said 1 ,3-benzothiazol-2-yl is optionally substituted with one or more groups independently selected from d.4 alkyl, C2_4 alkenyl, C2.4 alkynyl, -OH, -0(d.4 alkyl), -SH, -S(d-4 alkyl), -NH2, -NH(C1-4 alkyl), -N(d„4 alkyl)(d.4 alkyl), halogen, -CF3, or -CN.
8. The compound for use according to any of claims 1 to 7, wherein R4 is selected from -CO-(d.4 alkyl), -CHO, -0(C1-4 alkyl), -OH, -CO-0(d-4 alkyl), -COOH, d-4 alkyl, -(C1-4 alkyl)-CO-(d-4 alkyl), -(d-4 alkyl)-CHO, -(d-4 alkyl)-0(d-4 alkyl), -(d.4 alkyl)-OH, -(d-4 alkyl)-CO-0(C1-4 alkyl), -(d_4 alkyl)-COOH, -C(-CH3)=N-OH, -C(-CH3)=N-0(d.4 alkyl), -C(-CH3)=N-0-(d-4 alkyl)-COOH, -C(-CH3)=N-0-(C1-4 alkyI)-CO-0(d-4 alkyl), halogen, -CF3, -CN, or heterocycloalkyl.
9. The compound for use according to any of claims 1 to 8, wherein X is S, and further wherein L is -CO-NH- wherein the nitrogen atom comprised in said -CO-NH- is attached to ring A.
10. The compound for use according to claim 1 , wherein said compound is a compound of one of the following formulae 1 a to 1t, or a pharmaceutically acceptable salt, solvate or prodrug thereof:
Figure imgf000062_0001
1 b
Figure imgf000062_0002
1 c
Figure imgf000063_0001
Figure imgf000063_0002
Figure imgf000063_0003
Figure imgf000063_0004
Figure imgf000064_0001
Figure imgf000064_0002
Figure imgf000064_0003
Figure imgf000064_0004
Figure imgf000065_0001
11
Figure imgf000065_0002
1m
Figure imgf000065_0003
1n
Figure imgf000065_0004
1o
Figure imgf000066_0001
Figure imgf000066_0002
Figure imgf000066_0003
Figure imgf000066_0004
Figure imgf000067_0001
1t
1 1 . A pharmaceutical composition comprising a compound as defined in any of claims 1 to 10 or a pharmaceutically acceptable salt, solvate or prodrug thereof, and a pharmaceutically acceptable excipient, for use in the treatment or prevention of a cardiovascular disorder, cardiac hypertrophy, heart failure, anxiety, pain, chronic pain, migraine, an allergy, an inflammatory disorder, an autoimmune disorder, diabetes, diabetic complications, diabetic retinopathy, diabetic nephropathy, diabetic cardiomyopathy, diabetic neuropathy, cancer, metastatic cancer, drug-resistant cancer, stomach cancer, lung cancer, thyroid cancer, colon cancer, breast cancer, bipolar disorder, stroke, alopecia, or alcoholism.
12. A method of treating or preventing a disorder or condition selected from the group consisting of a cardiovascular disorder, cardiac hypertrophy, heart failure, anxiety, pain, chronic pain, migraine, an allergy, an inflammatory disorder, an autoimmune disorder, diabetes, diabetic complications, diabetic retinopathy, diabetic nephropathy, diabetic cardiomyopathy, diabetic neuropathy, cancer, metastatic cancer, drug- resistant cancer, stomach cancer, lung cancer, thyroid cancer, colon cancer, breast cancer, bipolar disorder, stroke, alopecia, and alcoholism, the method comprising the administration of a compound as defined in any of claims 1 to 10 or a pharmaceutically acceptable salt, solvate or prodrug thereof, or the pharmaceutical composition of claim 1 1 , to a subject in need thereof.
13. The compound for use according to any of claims 1 to 10, or the pharmaceutical composition for use according to claim 1 1 , or the method of claim 12, wherein said compound or said pharmaceutical composition is to be administered by any one of: an oral route; topical route, including by transdermal, intranasal, ocular, buccal, or sublingual route; parenteral route using injection techniques or infusion techniques, including by subcutaneous, intradermal, intramuscular, intravenous, intraarterial, intracardiac, intrathecal, intraspinal, intracapsular, subcapsular, intraorbital, intraperitoneal, intratracheal, subcuticular, intraarticular, subarachnoid, intrasternal, intraventricular, intra urethra I, or intracranial route; pulmonary route, including by inhalation or insufflation therapy; gastrointestinal route; intrauterine route; intraocular route; subcutaneous route; ophthalmic route, including by intravitreal, or intracameral route; rectal route; or vaginal route.
14. The method of claim 12 or 13, wherein said subject is a human.
15. A compound having one of the following formulae 1a or 1j to 1t:
Figure imgf000068_0001
1 k
Figure imgf000069_0001
1m
Figure imgf000069_0002
1n
Figure imgf000069_0003
1o
Figure imgf000070_0001
Figure imgf000070_0002
Figure imgf000070_0003
Figure imgf000070_0004
Figure imgf000071_0001
1t
or a pharmaceutically acceptable salt, solvate or prodrug thereof.
16. A compound having one of the following formulae 1a, 1 d, 1g, 1 h or 1j to 1t:
Figure imgf000071_0002
1a
Figure imgf000071_0003
1d
Figure imgf000071_0004
1g
Figure imgf000072_0001
Figure imgf000073_0001
1m
Figure imgf000073_0002
1n
Figure imgf000073_0003
1p
Figure imgf000074_0001
Figure imgf000074_0002
Figure imgf000074_0003
Figure imgf000074_0004
17. A pharmaceutical composition comprising a compound as defined in claim 16 or a pharmaceutically acceptable salt, solvate or prodrug thereof, and a pharmaceutically acceptable excipient.
PCT/EP2014/063723 2013-06-28 2014-06-27 Novel inhibitors of protein kinase c epsilon signaling WO2014207213A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP13174286.8 2013-06-28
EP13174286 2013-06-28

Publications (1)

Publication Number Publication Date
WO2014207213A1 true WO2014207213A1 (en) 2014-12-31

Family

ID=48698944

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2014/063723 WO2014207213A1 (en) 2013-06-28 2014-06-27 Novel inhibitors of protein kinase c epsilon signaling

Country Status (1)

Country Link
WO (1) WO2014207213A1 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017217439A1 (en) * 2016-06-14 2017-12-21 国立大学法人東京大学 THIENO[2,3-b]PYRIDINE DERIVATIVE AND QUINOLINE DERIVATIVE, AND USE THEREOF
CN109438347A (en) * 2018-12-19 2019-03-08 药大制药有限公司 A kind of cyano quinolines class IDO1 inhibitor, preparation method and application
WO2019067955A1 (en) * 2017-09-29 2019-04-04 The Regents Of The University Of California Compositions and methods for regulating let-7 micro rna targets
WO2019182944A1 (en) 2018-03-23 2019-09-26 Allcron Pharma Inc. Novel kinase inhibitors exhibiting anti-cancer activity and their method of use
WO2020080960A1 (en) * 2018-10-19 2020-04-23 Auckland Uniservices Limited Compounds for treating diabetes and/or related conditions
CN111072652A (en) * 2018-10-19 2020-04-28 暨南大学 Compounds for the treatment of diabetes and/or related disorders
US11213513B2 (en) 2017-02-24 2022-01-04 The Regents Of The University Of California Compositions and methods for promoting hair growth with Mpc1 inhibitors
US11312714B2 (en) 2017-06-30 2022-04-26 The Regents Of The University Of California Compositions and methods for modulating hair growth
WO2022162034A1 (en) * 2021-01-29 2022-08-04 F. Hoffmann-La Roche Ag Pyrazoleamide derivatives

Citations (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
EP0036676A1 (en) 1978-03-24 1981-09-30 The Regents Of The University Of California Method of making uniformly sized liposomes and liposomes so made
EP0052322A2 (en) 1980-11-10 1982-05-26 Gersonde, Klaus, Prof. Dr. Method of preparing lipid vesicles by ultrasonic treatment, the use of this method and apparatus for its application
JPS58118008A (en) 1982-01-06 1983-07-13 Nec Corp Data processor
EP0088046A2 (en) 1982-02-17 1983-09-07 Ciba-Geigy Ag Lipids in the aqueous phase
DE3218121A1 (en) 1982-05-14 1983-11-17 Leskovar, Peter, Dr.-Ing., 8000 München Pharmaceutical compositions for tumour treatment
EP0102324A2 (en) 1982-07-29 1984-03-07 Ciba-Geigy Ag Lipids and surfactants in an aqueous medium
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
EP0133988A2 (en) 1983-08-02 1985-03-13 Hoechst Aktiengesellschaft Regulating peptide-containing pharmaceutical preparations with retarded release, and process for their preparation
EP0142641A2 (en) 1983-09-26 1985-05-29 Udo Dr. Ehrenfeld Means and product for the diagnosis and therapy of tumours and for the treatment of weaknesses of the cellular and humoral immune system
EP0143949A1 (en) 1983-11-01 1985-06-12 TERUMO KABUSHIKI KAISHA trading as TERUMO CORPORATION Pharmaceutical composition containing urokinase
US4544545A (en) 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
WO1992003427A1 (en) 1990-08-17 1992-03-05 Yoshitomi Pharmaceutical Industries, Ltd. Ketone compound and remedy for osteoporosis
WO1997041833A1 (en) 1996-05-08 1997-11-13 Inhale Therapeutic Systems Dispersible macromolecule compositions and methods for their preparation and use
WO1999016419A1 (en) 1997-09-29 1999-04-08 Inhale Therapeutic Systems, Inc. Perforated microparticles and methods of use
WO2001085136A2 (en) 2000-05-10 2001-11-15 Alliance Pharmaceutical Corporation Phospholipid-based powders for drug delivery
US6376467B1 (en) 1998-10-09 2002-04-23 The Regents Of The University Of California Use of inhibitors of protein kinase C epsilon to treat pain
WO2003053411A1 (en) 2001-12-19 2003-07-03 Nektar Therapeutics Pulmonary delivery of aminoglycosides
US20090124553A1 (en) 2007-05-04 2009-05-14 Mochly-Rosen Daria D Suppression of inflammation associated with transplantation using an epsilon PKC inhibitor
RU2371444C1 (en) 2008-01-24 2009-10-27 Андрей Александрович Иващенко FURO- AND THIENO[2,3-b]-QUINOLINE-2-CARBOXAMIDES, METHOD OF PRODUCTION AND ANTITUBERCULOUS ACTIVITY
US20110077250A1 (en) 2009-06-26 2011-03-31 Ryder Sean Compounds for modulating rna binding proteins and uses therefor
CN102757447A (en) 2011-07-05 2012-10-31 北京大学 Urea transporter inhibitors, and preparation method and application thereof
WO2013033037A2 (en) 2011-08-26 2013-03-07 The Regents Of The University Of California Novel antiprion compounds
US20130129677A1 (en) 2009-02-27 2013-05-23 Siga Technologies, Inc. Thienopyridine Derivatives for the Treatment and Prevention of Dengue Virus Infections
WO2013123071A1 (en) * 2012-02-13 2013-08-22 Cleave Biosciences, Inc. Methods and compositions for jamm protease inhibition
WO2014116859A1 (en) * 2013-01-23 2014-07-31 The University Of Chicago Methods and compositions for inhibiting human copper trafficking proteins atox1 and ccs

Patent Citations (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
EP0036676A1 (en) 1978-03-24 1981-09-30 The Regents Of The University Of California Method of making uniformly sized liposomes and liposomes so made
EP0052322A2 (en) 1980-11-10 1982-05-26 Gersonde, Klaus, Prof. Dr. Method of preparing lipid vesicles by ultrasonic treatment, the use of this method and apparatus for its application
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
JPS58118008A (en) 1982-01-06 1983-07-13 Nec Corp Data processor
EP0088046A2 (en) 1982-02-17 1983-09-07 Ciba-Geigy Ag Lipids in the aqueous phase
DE3218121A1 (en) 1982-05-14 1983-11-17 Leskovar, Peter, Dr.-Ing., 8000 München Pharmaceutical compositions for tumour treatment
EP0102324A2 (en) 1982-07-29 1984-03-07 Ciba-Geigy Ag Lipids and surfactants in an aqueous medium
US4544545A (en) 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
EP0133988A2 (en) 1983-08-02 1985-03-13 Hoechst Aktiengesellschaft Regulating peptide-containing pharmaceutical preparations with retarded release, and process for their preparation
EP0142641A2 (en) 1983-09-26 1985-05-29 Udo Dr. Ehrenfeld Means and product for the diagnosis and therapy of tumours and for the treatment of weaknesses of the cellular and humoral immune system
EP0143949A1 (en) 1983-11-01 1985-06-12 TERUMO KABUSHIKI KAISHA trading as TERUMO CORPORATION Pharmaceutical composition containing urokinase
WO1992003427A1 (en) 1990-08-17 1992-03-05 Yoshitomi Pharmaceutical Industries, Ltd. Ketone compound and remedy for osteoporosis
WO1997041833A1 (en) 1996-05-08 1997-11-13 Inhale Therapeutic Systems Dispersible macromolecule compositions and methods for their preparation and use
WO1999016419A1 (en) 1997-09-29 1999-04-08 Inhale Therapeutic Systems, Inc. Perforated microparticles and methods of use
US6376467B1 (en) 1998-10-09 2002-04-23 The Regents Of The University Of California Use of inhibitors of protein kinase C epsilon to treat pain
WO2001085136A2 (en) 2000-05-10 2001-11-15 Alliance Pharmaceutical Corporation Phospholipid-based powders for drug delivery
WO2003053411A1 (en) 2001-12-19 2003-07-03 Nektar Therapeutics Pulmonary delivery of aminoglycosides
US20090124553A1 (en) 2007-05-04 2009-05-14 Mochly-Rosen Daria D Suppression of inflammation associated with transplantation using an epsilon PKC inhibitor
RU2371444C1 (en) 2008-01-24 2009-10-27 Андрей Александрович Иващенко FURO- AND THIENO[2,3-b]-QUINOLINE-2-CARBOXAMIDES, METHOD OF PRODUCTION AND ANTITUBERCULOUS ACTIVITY
US20130129677A1 (en) 2009-02-27 2013-05-23 Siga Technologies, Inc. Thienopyridine Derivatives for the Treatment and Prevention of Dengue Virus Infections
US20110077250A1 (en) 2009-06-26 2011-03-31 Ryder Sean Compounds for modulating rna binding proteins and uses therefor
CN102757447A (en) 2011-07-05 2012-10-31 北京大学 Urea transporter inhibitors, and preparation method and application thereof
WO2013033037A2 (en) 2011-08-26 2013-03-07 The Regents Of The University Of California Novel antiprion compounds
WO2013123071A1 (en) * 2012-02-13 2013-08-22 Cleave Biosciences, Inc. Methods and compositions for jamm protease inhibition
WO2014116859A1 (en) * 2013-01-23 2014-07-31 The University Of Chicago Methods and compositions for inhibiting human copper trafficking proteins atox1 and ccs

Non-Patent Citations (65)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences"
A. D. PILLAI ET AL: "A Cell-Based High-Throughput Screen Validates the Plasmodial Surface Anion Channel As an Antimalarial Target", MOLECULAR PHARMACOLOGY, vol. 77, no. 5, 1 May 2010 (2010-05-01), pages 724 - 733, XP055022753, ISSN: 0026-895X, DOI: 10.1124/mol.109.062711 *
AKITA, Y., FEBSJ., vol. 275, 2008, pages 3995 - 4004
ARKIN, M.R. ET AL., NATURE REV DRUG DISCOVERY, vol. 3, 2004, pages 301 - 317
B6HLER, M., DISSERTATION AT THE UNIVERSITY OF INNSBRUCK, 2006, pages 1 - 208
BEELEY, N.R., DRUG DISCOV. TODAY, vol. 5, 2000, pages 354 - 363
BRADFORD, M.M., ANAL. BIOCHEM., vol. 72, 1976, pages 248 - 254
BRODIE, C. ET AL., CELL GROWTH DIFFER., vol. 10, 1999, pages 183 - 191
BUNDGAARD, H.: "Design of Prodrugs", 1985, ELSEVIER, pages: 7 - 9,21-24
CASTRILLO, A. ET AL., J. EXP. MED., vol. 194, 2001, pages 1231 - 1242
CESARE, P. ET AL., NEURON, vol. 23, 1999, pages 617 - 624
CHA, D. ET AL., J. INVEST. DERMATOL., vol. 106, 1996, pages 5 0 - 597
CHEN, Y.B. ET AL., EXPERT OPIN. INVESTIG. DRUGS, vol. 7, 2008, pages 939 - 944
CHOI, D.S. ET AL., J. NEUROSCI., vol. 22, 2002, pages 9905 - 9911
CSUKAI, M. ET AL., J. BIOL. CHEM., vol. 272, 1997, pages 29200 - 29206
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 25 May 2001 (2001-05-25), CHEMICAL LIBRARY SUPPLIER: ASINEX, XP002715019, retrieved from STN accession no. 338433-13-9 Database accession no. 338433-13-9 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 27 April 2001 (2001-04-27), CHEMICAL LIBRARY SUPPLIER: ASINEX, XP002715020, retrieved from STN accession no. 333330-11-3 Database accession no. 333330-11-3 *
EASMON, J. ET AL., INT. J. CANCER, vol. 94, 2001, pages 89 - 96
EPSTEIN ET AL., PROC. NATL. ACAD. SCI. (USA, vol. 82, 1985, pages 3688 - 3692
ERDOGAN, E. ET AL., J. BIOL. CHEM., vol. 281, 2006, pages 28450 - 28459
FEDOROV, O. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 104, 2007, pages 20523 - 2058
FENTON, R.A. ET AL., AM. J. PHYSIOL. HEART CIRC. PHYSIOL., vol. 297, 2009, pages H718 - H725
FERREIRA, J.C. ET AL., J. MOL. CELL. CARDIOL, 2010
FLORIAN RECHFELD ET AL: "Thienoquinolines as Novel Disruptors of the PKC[epsilon]/RACK2 Protein-Protein Interaction", JOURNAL OF MEDICINAL CHEMISTRY, vol. 57, no. 8, 24 April 2014 (2014-04-24), pages 3235 - 3246, XP055143375, ISSN: 0022-2623, DOI: 10.1021/jm401605c *
GALEOTTI, N. ET AL., NEUROTHERAPEUTICS, vol. 10, 2013, pages 329 - 339
GARCZARCZYK, D. ET AL., CELL SIGNAL., vol. 21, 2009, pages 745 - 752
GARCZARCZYK, D. ET AL., CHEM. BIOL. INTERACT., vol. 185, 2010, pages 25 - 32
GORIN, M.A. ET AL., MOL. CANCER, vol. 8, 2009, pages 9
GRAY, M.O. ET AL., J. BIOL. CHEM., vol. 272, 1997, pages 30945 - 30951
GRUBER, P. ET AL., J. BIOCHEM., vol. 149, 2011, pages 331 - 336
HWANG ET AL., PROC. NATL. ACAD. SCI. (USA, vol. 77, 1980, pages 4030 - 4034
IKEDA, Y. ET AL., DIABETES, vol. 50, 2001, pages 584 - 92
JOHNSON, J.A. ET AL., J. BIOI. CHEM., vol. 271, 1996, pages 24962 - 24966
K. MASTERS: "Spray Drying Handbook", 1991, JOHN WILEY & SONS, INC.
KORFF, T. ET AL., J. CELL SCI., vol. 112, 1999, pages 3249 - 3258
LAPA, GB ET AL., J. ENZYME INHIB. MED. CHEM., 2012
MANNING, G. ET AL., SCIENCE, vol. 298, 2002, pages 1912 - 1934
MARAIS, R. ET AL., CELL, vol. 73, 1993, pages 381 - 393
MOCHLY-ROSEN, D. ET AL., NAT. REV. DRUG DISCOV., vol. 11, 2012, pages 937 - 957
MOCHLY-ROSEN, D. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 88, 1991, pages 3997 - 4000
NAKAMURA, S. ET AL., EXP. EYE RES., vol. 90, 2010, pages 137 - 145
NICHOLLS, A. ET AL., J. COMPUT.-AIDED MOL. DES., vol. 19, 2005, pages 661 - 686
P. GRUBER ET AL: "Barbituric acid derivative BAS 02104951 inhibits PKC , PKC , PKC /RACK2 interaction, Elk-1 phosphorylation in HeLa and PKC and translocation in PC3 cells following TPA-induction", JOURNAL OF BIOCHEMISTRY, vol. 149, no. 3, 1 March 2011 (2011-03-01), pages 331 - 336, XP055084145, ISSN: 0021-924X, DOI: 10.1093/jb/mvq147 *
PASS, J.M. ET AL., AM. J. PHYSIOL. HEART CIRC. PHYSIOL., vol. 280, 2001, pages H946 - 955
PERLETTI, G. ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 221, 1996, pages 688 - 691
PILLAI, A.D. ET AL., MOL PHARMACOL., vol. 77, 2010, pages 724 - 733
PRICE, M.A. ET AL., EMBOJ., vol. 14, 1995, pages 2589 - 2601
R. LANGER ET AL., J. BIOMED. MATER. RES., vol. 15, 1981, pages 167 - 277
R. LANGER, CHEM. TECH., vol. 12, 1982, pages 98 - 105
RAO, V.N. ET AL., SCIENCE, vol. 244, 1989, pages 66 - 70
RECHFELD, F. ET AL., CURR. TOPICS MED. CHEM., vol. 11, 2011, pages 1305 - 1319
REICHLING, DB ET AL., TRENDS NEUROSCI., vol. 32, 2009, pages 611 - 618
RON, D. ET AL., J. BIOL. CHEM., vol. 274, 1999, pages 27039 - 27046
SANAM R ET AL: "Combined pharmacophore and structure-guided studies to identify diverse HSP90 inhibitors", JOURNAL OF MOLECULAR GRAPHICS AND MODELLING, ELSEVIER SCIENCE, NEW YORK, NY, US, vol. 28, no. 6, 26 February 2010 (2010-02-26), pages 472 - 477, XP026875379, ISSN: 1093-3263, [retrieved on 20091124] *
SANAM, R. ET AL., J MOL GRAPH MODEL., vol. 28, 2010, pages 472 - 477
SANCHEZ-BAUTISTA, S. ET AL., RECENT PAT. DNA GENE, vol. 7, 2013, pages 74 - 81
SCHECHTMAN, D. ET AL., ONCOGENE, vol. 20, 2001, pages 6339 - 6347
SHIRAI, Y. ET AL., FEBSJ., vol. 275, 2008, pages 3988 - 3994
SIDMAN, U. ET AL., BIOPOLYMERS, vol. 22, 1983, pages 547 - 556
SKEHAN, P. ET AL., J. NATL. CANCER INST., vol. 82, 1990, pages 1107 - 1112
SOLTOFF, S.P., TRENDS PHARMACOL. SCI., vol. 28, 2007, pages 453 - 458
STENSMAN, H. ET AL., BMC CANCER, vol. 8, 2008, pages 365
TAKAHASHI, T. ET AL., SKIN PHARMACOL. APPL. SKIN PHYSIOL., vol. 13, 2000, pages 133 - 142
TAKEISHI, Y. ET AL., CIRC. RES., vol. 86, 2000, pages 1218 - 1823
XUAN, Y.T. ET AL., CIRCULATION, vol. 112, 2005, pages 971 - 978

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP7041877B2 (en) 2016-06-14 2022-03-25 国立大学法人 東京大学 Thieno [2,3-b] pyridine and quinoline derivatives and their use
JPWO2017217439A1 (en) * 2016-06-14 2019-04-11 国立大学法人 東京大学 Thieno [2,3-b] pyridine and quinoline derivatives and their use
WO2017217439A1 (en) * 2016-06-14 2017-12-21 国立大学法人東京大学 THIENO[2,3-b]PYRIDINE DERIVATIVE AND QUINOLINE DERIVATIVE, AND USE THEREOF
US10689394B2 (en) 2016-06-14 2020-06-23 The University Of Tokyo Thieno[2,3-b]pyridine derivative, quinoline derivative, and use thereof
US11213513B2 (en) 2017-02-24 2022-01-04 The Regents Of The University Of California Compositions and methods for promoting hair growth with Mpc1 inhibitors
US11787804B2 (en) 2017-06-30 2023-10-17 The Regents Of The University Of California Compositions and methods for modulating hair growth
US11472804B2 (en) 2017-06-30 2022-10-18 The Regents Of The University Of California Compositions and methods for modulating hair growth
US11312714B2 (en) 2017-06-30 2022-04-26 The Regents Of The University Of California Compositions and methods for modulating hair growth
WO2019067955A1 (en) * 2017-09-29 2019-04-04 The Regents Of The University Of California Compositions and methods for regulating let-7 micro rna targets
WO2019182944A1 (en) 2018-03-23 2019-09-26 Allcron Pharma Inc. Novel kinase inhibitors exhibiting anti-cancer activity and their method of use
US11697654B2 (en) 2018-03-23 2023-07-11 Allcron Pharma Inc. Kinase inhibitors exhibiting anti-cancer activity and their method of use
CN111072652A (en) * 2018-10-19 2020-04-28 暨南大学 Compounds for the treatment of diabetes and/or related disorders
WO2020080960A1 (en) * 2018-10-19 2020-04-23 Auckland Uniservices Limited Compounds for treating diabetes and/or related conditions
CN111072652B (en) * 2018-10-19 2023-05-23 暨南大学 Compounds for the treatment of diabetes and/or related disorders
CN109438347B (en) * 2018-12-19 2022-09-06 药大制药有限公司 Cyanoquinoline IDO1 inhibitor, and preparation method and application thereof
CN109438347A (en) * 2018-12-19 2019-03-08 药大制药有限公司 A kind of cyano quinolines class IDO1 inhibitor, preparation method and application
WO2022162034A1 (en) * 2021-01-29 2022-08-04 F. Hoffmann-La Roche Ag Pyrazoleamide derivatives

Similar Documents

Publication Publication Date Title
WO2014207213A1 (en) Novel inhibitors of protein kinase c epsilon signaling
US10696642B2 (en) TEAD transcription factor autopalmitoylation inhibitors
KR102587175B1 (en) Inhibitors of human ezh2, and methods of use thereof
US20080021063A1 (en) Compositions and methods for modulating sirtuin activity
JP6991993B2 (en) How to treat hyperalgesia
JP5937247B2 (en) Compositions and methods for treating neoplasms, inflammatory diseases, and other disorders
US20090259044A1 (en) Compositions and methods for modulating sirtuin activity
JP2010530736A (en) Inhibitors of cell death induced by thapsigargin.
MX2012013255A (en) Compositions and methods for treating neoplasia, inflammatory disease and other disorders.
JP2008521831A (en) PPAR active compounds
WO1994026260A1 (en) METHODS AND COMPOUNDS FOR INHIBITING CELL PROLIFERATIVE DISORDERS CHARACTERIZED BY ABNORMAL abl ACTIVITY
EP3148967B1 (en) Sulfonamide compounds and their use as stat5 inhibitors
KR20100010894A (en) Medical composition containing a dyrk-inhibiting compound
BR112021007142A2 (en) flavagline derivatives for inhibition of kras oncogene activation
US9206115B2 (en) ATGListatin and pharmaceutical composition comprising the same
WO2019089577A1 (en) Small molecule inhibitors of cancer stem cells and mesenchymal cancer types
KR101438532B1 (en) Pharmaceutical compositions for pregnosing or treating degenerative brain disease and method for screening the same
Wu et al. The development of a novel transforming growth factor-β (TGF-β) inhibitor that disrupts ligand-receptor interactions
US10526322B2 (en) Inhibitors of kidney-type glutaminase, GLS-1
WO2021213518A1 (en) Compound for preventing or treating lipid metabolism-related diseases
AU2015308438B2 (en) Novel chromone oxime derivative and its use as allosteric modulator of metabotropic glutamate receptors
JP2019521964A (en) Adrenergic receptor modulating compounds and methods of use thereof
EP2948433B1 (en) Atglistatin as lipase inhibitor
Kumar et al. Dual targeted 2-Benzylideneindanone pendant hydroxamic acid group exhibits selective HDAC6 inhibition along with tubulin stabilization effect
CN109134433A (en) It is a kind of inhibit ROCK compound and its application

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14744288

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14744288

Country of ref document: EP

Kind code of ref document: A1