WO2019121670A1 - 4,5-dihydro-1h-thieno[2,3-g]indazolyl derivatives having multimodal activity against pain - Google Patents

4,5-dihydro-1h-thieno[2,3-g]indazolyl derivatives having multimodal activity against pain Download PDF

Info

Publication number
WO2019121670A1
WO2019121670A1 PCT/EP2018/085479 EP2018085479W WO2019121670A1 WO 2019121670 A1 WO2019121670 A1 WO 2019121670A1 EP 2018085479 W EP2018085479 W EP 2018085479W WO 2019121670 A1 WO2019121670 A1 WO 2019121670A1
Authority
WO
WIPO (PCT)
Prior art keywords
thieno
dihydro
substituted
dimethylamino
indazol
Prior art date
Application number
PCT/EP2018/085479
Other languages
French (fr)
Inventor
Lourdes GARRIGA-SANAHUJA
Rosalía PASCUAL-RAMON
Carmen ALMANSA-ROSALES
José-Luís DÍAZ-FERNÁNDEZ
Original Assignee
Esteve Pharmaceuticals, S.A.
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 Esteve Pharmaceuticals, S.A. filed Critical Esteve Pharmaceuticals, S.A.
Publication of WO2019121670A1 publication Critical patent/WO2019121670A1/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
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids

Definitions

  • the present invention relates to compounds having dual pharmacological activity towards both the a 2 d subunit of the voltage-gated calcium channel, and the m-opioid receptor (MOR or mu-opioid receptor), and more particularly to 4,5-Dihydro-1 H- thieno[2,3-g]indazolyl derivatives having this pharmacological activity, to processes of preparation of such compounds, to pharmaceutical compositions comprising them, and to their use in therapy, in particular for the treatment of pain.
  • MOR m-opioid receptor
  • NSAIDs non-steroidal anti-inflammatory drugs
  • opioid agonists opioid agonists
  • calcium channel blockers and antidepressants
  • antidepressants but they are much less than optimal regarding their safety ratio. All of them show limited efficacy and a range of secondary effects that preclude their use, especially in chronic settings.
  • Voltage-gated calcium channels are required for many key functions in the body. Different subtypes of voltage-gated calcium channels have been described (Zamponi et al., Pharmacol. Rev. 2015 67:821 -70).
  • the VGCC are assembled through interactions of different subunits, namely oti (Ca v ai), b (Ca v p) a 2 d (Ca v a 2 3) and g (Ca v y).
  • the oti subunits are the key porous forming units of the channel complex, being responsible for the Ca 2+ conduction and generation of Ca 2+ influx.
  • VGCC can be subdivided into low voltage- activated T-type (Ca v 3.1 , Ca v 3.2, and Ca v 3.3), and high voltage-activated L- (Ca v 1 .1 through Ca v 1 .4), N-(Ca v 2.2), P/Q-(Ca v 2.1 ), and R-(Ca v 2.3) types, depending on the channel forming Cava subunits.
  • Current therapeutic agents include drugs targeting L-type Ca v 1 .2 calcium channels, particularly 1 ,4-dihydropyridines, which are widely used in the treatment of hypertension.
  • T-type (Ca v 3) channels are the target of ethosuximide, widely used in absence epilepsy.
  • Ziconotide a peptide blocker of N-type (Ca v 2.2) calcium channels, has been approved as a treatment of intractable pain. (Perret and Luo, 2009, supra, Vink and Alewood, Br J Pharmacol. 2012 167:970-89.).
  • the Ca v 1 and Ca v 2 subfamilies contain an auxiliary a d subunit, which is the therapeutic target of the gabapentinoid drugs of value in certain epilepsies and chronic neuropathic pain.
  • a d subunit which encoded by a unique gene and all possessing splice variants.
  • Each a d protein is encoded by a single messenger RNA and is posttranslationally cleaved and then linked by disulfide bonds.
  • Four genes encoding a d subunits have now been cloned. a 2 d-1 was initially cloned from skeletal muscle and shows a fairly ubiquitous distribution.
  • the a 2 d-2 and a 2 d-3 subunits were subsequently cloned from brain.
  • the most recently identified subunit, a 2 d-4 is largely nonneuronal.
  • the human a 2 d-4 protein sequence shares 30, 32 and 61 % identity with the human a 2 d-1 , a 2 d-2 and a 2 d-3 subunits, respectively.
  • the gene structure of all a 2 d subunits is similar. All a 2 d subunits show several splice variants (Davies et al., Trends Pharmacol Sci. 2007 28:220-8.; Dolphin AC, Nat Rev Neurosci. 2012 13:542-55., Biochim Biophys Acta. 2013 1828:1541 -9.).
  • the Ca v a 2 6-1 subunit may play an important role in neuropathic pain development (Perret and Luo, 2009, supra ; Vink and Alewood, 2012, supra).
  • Biochemical data have indicated a significant Ca v a 2 6-1 , but not Ca v a 2 6-2, subunit upregulation in the spinal dorsal horn, and DRG (dorsal root ganglia) after nerve injury that correlates with neuropathic pain development.
  • the Ca v ot 2 5-1 subunit (and the Ca v ot 2 5-2, but not Ca v ot 2 5-3 and Ca v ot 2 5-4, subunits) is the binding site for gabapentin which has anti-allodynic/ hyperalgesic properties in patients and animal models.
  • injury-induced Ca v ot 2 5-1 expression correlates with neuropathic pain development and maintenance, and various calcium channels are known to contribute to spinal synaptic neurotransmission and DRG neuron excitability
  • injury-induced Ca v ot 2 5-1 subunit upregulation may contribute to the initiation and maintenance of neuropathic pain by altering the properties and/or distribution of VGCC in the subpopulation of DRG neurons and their central terminals, therefore modulating excitability and/or synaptic neuroplasticity in the dorsal horn.
  • Intrathecal antisense oligonucleotides against the Ca v ot 2 5-1 subunit can block nerve injury-induced Ca v ot 2 5-1 upregulation and prevent the onset of allodynia and reserve established allodynia.
  • the 0, 2 6 subunits of VGCC form the binding site for gabapentin and pregabalin, which are structural derivatives of the inhibitory neurotransmitter GABA although they do not bind to GABAA, GABAB, or benzodiazepine receptors, or alter GABA regulation in animal brain preparations.
  • the binding of gabapentin and pregabalin to the Ca v ot 2 5 subunit results in a reduction in the calcium-dependent release of multiple neurotransmitters, leading to efficacy and tolerability for neuropathic pain management.
  • Gabapentinoids may also reduce excitability by inhibiting synaptogenesis (Perret and Luo, 2009, supra, Vink and Alewood, 2012, supra, Zamponi et al., 2015, supra).
  • MOR m-opioid receptor
  • MOR agonists are not optimal for the treatment of chronic pain as indicated by the diminished effectiveness of morphine against chronic pain conditions. This is especially proven for the chronic pain conditions of neuropathic or inflammatory origin, in comparison to its high potency against acute pain.
  • the finding that chronic pain can lead to MOR down-regulation may offer a molecular basis for the relative lack of efficacy of morphine in long-term treatment settings [Dickenson, A.H., Suzuki, R. Opioids in neuropathic pain: Clues from animal studies. Eur J Pain 9, 1 13-6 (2005)].
  • prolonged treatment with morphine may result in tolerance to its analgesic effects, most likely due to treatment-induced MOR down-regulation, internalization and other regulatory mechanisms.
  • long-term treatment can result in substantial increases in dosing in order to maintain a clinically satisfactory pain relief, but the narrow therapeutic window of MOR agonists finally results in unacceptable side effects and poor patient compliance.
  • Polypharmacology is a phenomenon in which a drug binds multiple rather than a single target with significant affinity.
  • the effect of polypharmacology on therapy can be positive (effective therapy) and/or negative (side effects). Positive and/or negative effects can be caused by binding to the same or different subsets of targets; binding to some targets may have no effect.
  • Multi-component drugs or multi-targeting drugs can overcome toxicity and other side effects associated with high doses of single drugs by countering biological compensation, allowing reduced dosage of each compound or accessing context-specific multitarget mechanisms. Because multitarget mechanisms require their targets to be available for coordinated action, one would expect synergies to occur in a narrower range of cellular phenotypes given differential expression of the drug targets than would the activities of single agents.
  • multi-targeting drugs may produce concerted pharmacological intervention of multiple targets and signaling pathways that drive pain. Because they actually make use of biological complexity, multi- targeting (or multi-component drugs) approaches are among the most promising avenues toward treating multifactorial diseases such as pain (Gilron et al., Lancet Neurol. 2013 Nov;12(1 1 ):1084-95.). In fact, positive synergistic interaction for several compounds, including analgesics, has been described (Schroder et al., J Pharmacol Exp Ther. 201 1 ; 337:312-20. Erratum in: J Pharmacol Exp Ther.
  • An alternative strategy for multitarget therapy is to design a single compound with selective polypharmacology (multi-targeting drug). It has been shown that many approved drugs act on multiple targets. Dosing with a single compound may have advantages over a drug combination in terms of equitable pharmacokinetics and biodistribution. Indeed, troughs in drug exposure due to incompatible pharmacokinetics between components of a combination therapy may create a low-dose window of opportunity where a reduced selection pressure can lead to drug resistance. In terms of drug registration, approval of a single compound acting on multiple targets faces significantly lower regulatory barriers than approval of a combination of new drugs (Hopkins, 2008, supra).
  • the present application relates to the advantages of having dual activity, for m- receptor and the a 2 d-1 subunit of voltage-gated calcium channels, in the same molecule to treat chronic pain.
  • the present invention relates to compounds having a complementary dual mechanism of action (m-receptor agonist and blocker of the a 2 d subunit, in particular the a 2 d-1 subunit, of voltage-gated calcium channels) which implies a better profile of tolerability than the strong opioids (morphine, oxycodone, fentanyl etc) and/or better efficacy and tolerability than gabapentinoids (pregabalin and gabapentin).
  • m-receptor agonist and blocker of the a 2 d subunit, in particular the a 2 d-1 subunit, of voltage-gated calcium channels which implies a better profile of tolerability than the strong opioids (morphine, oxycodone, fentanyl etc) and/or better efficacy and tolerability than gabapentinoids (pregabalin and gabapentin).
  • Pain is multimodal in nature, since in nearly all pain states several mediators, signaling pathways and molecular mechanisms are implicated. Consequently, monomodal therapies fail to provide complete pain relief.
  • therapies are a common clinical practice and many efforts are directed to assess the best combination of available drugs in clinical studies (Mao, J., Gold, M.S., Backonja, M.; 201 1 ; J. Pain; 12; 157-166). Accordingly, there is still a need to find compounds that have an alternative or improved pharmacological activity in the treatment of pain, being both effective and showing the desired selectivity, and having good“drugability” properties, i.e. good pharmaceutical properties related to administration, distribution, metabolism and excretion.
  • the inventors have found a series of compounds showing dual pharmacological activity towards both the a 2 d subunit, in particular the a 2 d-1 subunit, of the voltage-gated calcium channel, and the m-opioid receptor (MOR or mu-opioid receptor) resulting in an innovative, effective and alternative solution for the treatment of pain.
  • MOR m-opioid receptor
  • the present invention offers a solution by combining in a single compound binding to two different targets relevant for the treatment of pain. This was mainly achieved by providing the compounds according to the invention that bind both to the m-opioid receptor and to the a 2 d subunit, in particular the a 2 d-1 subunit, of the voltage-gated calcium channel.
  • the inventors have found a series of compounds, encompassed by formula (I), that show a dual pharmacological activity towards both the a 2 d subunit, in particular the a 2 d-1 subunit, of the voltage-gated calcium channel, and the m-opioid receptor thus solving the above problem of identifying alternative or improved pain treatments by offering such dual compounds.
  • the main object of the invention is directed to a compound having a dual activity binding to the a 2 d subunit, in particular the a 2 d-1 subunit, of the voltage-gated calcium channel and the m-opioid receptor.
  • This compound can be used in the treatment of pain.
  • the invention is directed in a main aspect to a compound of formula (I), wherein Ri, Rr R 2 , Rs, R6, W 3 , W 4 , X, Y and A are as defined below in the detailed description.
  • a further aspect of the invention refers to the processes for preparation of compounds of formula (I).
  • a still further aspect of the invention refers to the use of intermediate compounds for the preparation of a compound of formula (I).
  • composition comprising a compound of formula (I).
  • the invention is directed to a family of compounds which have a dual pharmacological activity towards both the a 2 d subunit, in particular the a 2 d-1 subunit, of the voltage-gated calcium channel, and the m-opioid receptor and to their use in the treatment of pain and related disorders.
  • the applicant has surprisingly found that the problem of providing a new effective and alternative for treating pain and pain related disorders can be solved by using a multimodal balanced analgesic approach combining two different synergistic activities in a single drug (i.e., dual ligands which are bifunctional and bind to m-opioid receptor and to a2d subunit, in particular the a2d-1 subunit, of the voltage-gated calcium channel), thereby enhancing through the a2d blockade without increasing the undesirable side effects.
  • This supports the therapeutic value of a dual agent, whereby the a2d binding component acts as an intrinsic adjuvant of the MOR binding component.
  • a dual compound that possess binding to both the m-opioid receptor and to the a2d subunit of the voltage-gated calcium channel shows a highly valuable therapeutic potential by achieving an outstanding analgesia (enhanced in respect to the potency of the opioid component alone) with a reduced side-effect profile (safety margin increased compared to that of the opioid component alone) versus existing opioid therapies.
  • the dual compounds according to the present invention would in addition show one or more the following functionalities: blockade of the a2d subunit, in particular the a2d-1 subunit, of the voltage-gated calcium channel and m-opioid receptor agonism
  • An antagonist blocks or dampens agonist-mediated responses.
  • Known subfunctionalities are neutral antagonists or inverse agonists.
  • An agonist increases the activity of the receptor above its basal level.
  • Known subfunctionalities are full agonists, or partial agonists.
  • the two mechanisms complement each other since MOR agonists are only marginally effective in the treatment of neuropathic pain, while the blockers of the a2d subunit, in particular the a2d-1 subunit, of voltage-gated calcium channels show outstanding effects in preclinical neuropathic pain models.
  • the a2d component in particular the a2d-1 component, adds unique analgesic actions in opioid-resistant pain.
  • the dual approach has clear advantages over MOR agonists in the treatment of chronic pain as lower and better tolerated doses would be needed based on the potentiation of analgesia but not of the adverse events of MOR agonists.
  • a further advantage of using designed multiple ligands is a lower risk of drug-drug interactions compared to cocktails or multi-component drugs, thus involving simpler pharmacokinetics and less variability among patients. Additionally, this approach may improve patient compliance and broaden the therapeutic application in relation to monomechanistic drugs, by addressing more complex aetiologies.
  • the present invention is directed to a compound of formula (I):
  • W3 and W 4 are S and the other is C;
  • Y is -C(O)-, -[C(RaRa')]n-, -CRa(CN)- or -CR a (CONRa'Ra ")-.
  • R a , R a ⁇ and R a” are independently selected from hydrogen, halogen, substituted or unsubstituted Ci- 6 alkyl, substituted or unsubstituted C2-6 alkenyl, and substituted or unsubstituted C2-6 alkynyl; and n is 0, 1 , 2 or 3;
  • X is selected from a bond, -[C(RbRb )] -, -[C(RbRb )] C(0)[C(Rb"Rb )]q- > substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl-C(O) and substituted or unsubstituted aryl- alkyl, wherein each R b , R b ⁇ , R b - and R b - are independently selected from hydrogen, halogen, substituted or unsubstituted Ci- 6 alkyl, substituted or unsubstituted C 2-6 alkenyl, and substituted or unsubstituted C2-6 alkynyl; and p is 0, 1 , 2 or 3; q is 0, 1 , 2 or 3;
  • Group A is a substituted or unsubstituted 4 to 10 membered mono or bicyclic heterocyclyl containing one nitrogen atom and optionally a second heteroatom selected from N and O, wherein A is attached to Y through a nitrogen atom; each R1 and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl, substituted or unsubstituted C2-6 alkynyl, substituted or unsubstituted alkyl-cycloalkyl; N R4R4', C(0)OR 4 and substituted or unsubstituted haloalkyl; wherein each R 4 and R 4' are independently selected from hydrogen, halogen, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl, substituted or unsubstituted C 2-6 alkynyl, and C(0)OR 4 ⁇
  • R 5 is selected from hydrogen, halogen, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl, substituted or unsubstituted C 2-6 alkynyl, CN, -C(0)OR 3 and -S(0) 2 0R 3' ; wherein each R 3 and Ry are independently selected from hydrogen, halogen, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl, and substituted or unsubstituted C 2-6 alkynyl;
  • R 6 is selected from hydrogen, halogen, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2-6 alkynyl;
  • R 2 is a group of the following formula:
  • Wi is selected from C and N; each Rg, Rg ⁇ , Rg- and Rg- are independently selected from hydrogen, substituted or unsubstituted Ci- 6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2-6 alkynyl; alternatively, Rg and Rg ⁇ and/or Rg- and Rg- taken together with the carbon atom to which they are attached may form a substituted or unsubstituted cycloalkyl; alternatively, Rg and Rg ⁇ and/or Rg- and Rg- taken together with the carbon atom to which they are attached may form a carbonyl group; each R10, Rio ⁇ , R10” and Rio- are independently selected from hydrogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2-6 alkynyl; alternatively, R10, Rio ⁇ and/or Rio ⁇
  • R 7 is selected from substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted aryl and substituted or unsubstituted aromatic heterocyclyl;
  • Rs is selected from hydrogen, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl and substituted or unsubstituted C 2 - e alkynyl;
  • R 7 and Rs taken together with -N(Rs')-[CH 2 ] t - atoms to which they are attached form a substituted or unsubstituted heterocyclyl optionally fused with a substituted or unsubstituted aryl group; wherein the compound of formula (I) is optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compounds of the invention represented by the above described formula (I) may include enantiomers depending on the presence of chiral centres or isomers depending on the presence of multiple bonds.
  • the single isomers, enantiomers or diastereoisomers and mixtures thereof fall within the scope of the present invention.
  • these compounds according to the invention are optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt or solvate thereof.
  • alkyl is understood as meaning a straight or branched hydrocarbon chain radical containing no unsaturation, and which is attached to the rest of the molecule by a single bond. It may be unsubstituted or substituted once or several times. It encompasses e.g. -CH3 and -CH2-CH3.
  • Ci-2-alkyl represents C1 - or C2-alkyl
  • Ci-3-alkyl represents C1 -, C2- or C3-alkyl
  • Ci- 4 -alkyl represents C1 -, C2, C3- or C4-alkyl
  • Ci-5-alkyl represents C1 -, C2-, C3-, C4-, or C5-alkyl
  • Ci-6-alkyl represents C1 -, C2-, C3-, C4-, C5- or C6-alkyl.
  • alkyl radicals include among others methyl, ethyl, propyl, methylethyl, butyl, 1 -methylpropyl, 2-methylpropyl, 1 ,1 - dimethylethyl, pentyl, 1 ,1 -dimethylpropyl, 1 ,2-dimethylpropyl, 2,2-dimethylpropyl, hexyl, 1 -methylpentyl. If substituted by cycloalkyl, it corresponds to a“cycloalkylalkyl” radical, such as cyclopropylmethyl.
  • alkyl is understood in the context of this invention Ci- 6 -alkyl like methyl, ethyl, propyl, butyl, pentyl, or hexyl; and more preferably is C1-4- alkyl like methyl, ethyl, propyl or butyl.
  • the alkenyl radicals are preferably vinyl (ethenyl), allyl (2-propenyl).
  • alkenyl is C2-6-alkenyl like ethylene, propylene, butylene, pentylene, or hexylene; or is C2-4-alkenyl, like ethylene, propylene, or butylenes.
  • Alkynyl is understood as meaning a straight or branched hydrocarbon chain radical containing at least two carbon atoms and at least one carbon-carbon triple bond, and which is attached to the rest of the molecule by a single bond. It may be unsubstituted or substituted once or several times. It encompasses groups like e.g. -C ⁇ C-CHs (1 - propynyl).
  • alkynyl in the context of this invention is C2-6-alkynyl like ethyne, propyne, butyene, pentyne, or hexyne; or is C2-4-alkynyl like ethyne, propyne or butyene.
  • alkyl also in aryl-alkyl, alkylheterocyclyl or alkylcycloalkyl
  • substituted in the context of this invention is understood as meaning replacement of at least one hydrogen radical on a carbon atom by halogen, -OR’, -SR’, -SOR’, -SO 2 R’, -OR’, -CN, -COR’, -COOR’, -NR’R”, -CONR’R”, haloalkyl, haloalkoxy or -OC 1-6 alkyl wherein each of the R’ and R” groups is independently selected from the group consisting of hydrogen, OH, NO 2 , NH 2 , SH, CN, halogen, -COH, -COalkyl, -COOH and C 1-6 alkyl.
  • the alkyl, alkenyl or alkynyl as defined in R 1 - R 10” if substituted, is substituted with one or more substituent/s selected from -OR’, halogen, -CN, haloalkyl, haloalkoxy and -NR’R”; wherein R, R’ and R” are independently selected from hydrogen, unsubstituted C 1-6 alkyl, unsubstituted C 2-6 alkenyl, and unsubstituted C 2-6 alkynyl;
  • More than one replacement on the same molecule and also on the same carbon atom is possible with the same or different substituents.
  • This includes for example 3 hydrogens being replaced on the same C atom, as in the case of CF 3 , or at different places of the same molecule, as in the case of e.g. -CH(OH)-CH CH-CHCl2.
  • haloalkyl is understood as meaning an alkyl being substituted once or several times by a halogen (selected from F, Cl, Br, I). It encompasses e.g. -CH2CI, -CH2F, -CHC , -CHF2, -CCI3, -CF3 and -CH2-CHCI2.
  • haloalkyl is understood in the context of this invention as halogen-substituted Ci- 4 -alkyl representing halogen substituted C1 -, C2-, C3- or C4-alkyl.
  • the halogen- substituted alkyl radicals are thus preferably methyl, ethyl, propyl, and butyl.
  • Preferred examples include -CH 2 CI, -CH 2 F, -CH 2 -CH 2 F, -CH 2 -CHF 2 , -CHCb, -CHF 2 , and -CF 3 .
  • haloalkoxy is understood as meaning an -O-alkyl being substituted once or several times by a halogen (selected from F, Cl, Br, I). It encompasses e.g. -OCH2CI, -OCH2F, -OCHCb, -OCHF2, -OCCI3, -OCF3 and -OCH2- CHCb.
  • haloalkoxy is understood in the context of this invention as halogen- substituted -OCi- 4 -alkyl representing halogen substituted C1 -, C2-, C3- or C4-alkoxy.
  • the halogen-substituted O-alkyl radicals are thus preferably O-methyl, O-ethyl, O-propyl, and O-butyl.
  • Preferred examples include -OCH 2 CI, -OCH 2 F, -OCHCb, -OCHF 2 , and - OCF3.
  • cycloalkyl is understood as meaning saturated and unsaturated (but not aromatic) cyclic hydrocarbons (without a heteroatom in the ring), which can be unsubstituted or once or several times substituted.
  • Preferred cycloalkyls are C3-4-cycloalkyl representing C3- or C4-cycloalkyl, C3-5-cycloalkyl representing C3-, C4- or C5-cycloalkyl, C3-6-cycloalkyl representing C3-, C4-, C5- or C6-cycloalkyl, C3-7- cycloalkyl representing C3-, C4-, C5-, C6- or C7-cycloalkyl, C3-8-cycloalkyl representing C3-, C4-, C5-, C6-, C7- or C8-cycloalkyl, C4-5-cycloalkyl representing C4- or C5- cycloalkyl, C4-6-cycloalkyl
  • Examples are cyclopropyl, 2-methylcyclopropyl, cyclopropylmethyl, cyclobutyl, cyclopentyl, cyclopentylmethyl, cyclohexyl, cycloheptyl, cyclooctyl, and also adamantyl.
  • cycloalkyl is C3-8-cycloalkyl like cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, or cyclooctyl; or is C3-7-cycloalkyl like cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl; or is C3-6-cycloalkyl like cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl, especially cyclopentyl or cyclohexyl.
  • Aryl is understood as meaning 6 to 18 membered mono or polycyclic ring systems with at least one aromatic ring but without heteroatoms even in only one of the rings. Examples are phenyl, naphthyl, fluoranthenyl, fluorenyl, tetralinyl or indanyl, 9H-fluorenyl or anthracenyl radicals, which can be unsubstituted or once or several times substituted. Most preferably aryl is understood in the context of this invention as phenyl, naphthyl or anthracenyl, more preferably the aryl is phenyl.
  • a heterocyclyl radical or group (also called heterocyclyl hereinafter) is understood as meaning 4 to 18, preferably 5 to 18, membered mono or polycyclic heterocyclic ring systems, with at least one saturated or unsaturated ring which contains one or more heteroatoms selected from the group consisting of nitrogen, oxygen and/or sulfur in the ring.
  • a heterocyclic group can also be substituted once or several times.
  • Examples include non-aromatic heterocyclyls such as tetrahydropyran, oxazepane, morpholine, piperidine, pyrrolidine as well as heteroaryls such as furan, benzofuran, thiophene, benzothiophene, pyrrole, pyridine, pyrimidine, pyrazine, quinoline, isoquinoline, phthalazine, thiazole, benzothiazole, indole, benzotriazole, carbazole and quinazoline.
  • non-aromatic heterocyclyls such as tetrahydropyran, oxazepane, morpholine, piperidine, pyrrolidine as well as heteroaryls such as furan, benzofuran, thiophene, benzothiophene, pyrrole, pyridine, pyrimidine, pyrazine, quinoline, isoquinoline, phthalazine, thiazole,
  • heteroaryl (being equivalent to heteroaromatic radicals or aromatic heterocyclyls) is an aromatic 5 to 18 membered mono or polycyclic heterocyclic ring system of one or more rings of which at least one aromatic ring contains one or more heteroatoms from the group consisting of nitrogen, oxygen and/or sulfur in the ring; preferably is a 5 to 18 membered mono or polycyclic aromatic heterocyclic ring system of one or two rings of which at least one aromatic ring contains one or more heteroatoms selected from the group consisting of nitrogen, oxygen and/or sulfur in the ring, more preferably it is selected from furan, benzofuran, thiophene, benzothiophene, pyrrole, pyridine, pyrimidine, pyrazine, quinoline, isoquinoline, phthalazine, benzothiazole, indole, benzotriazole, carbazole,
  • the non-aromatic heterocyclyl is a 4 to 18, preferably 5 to 18, membered mono or polycyclic heterocyclic ring system of one or more rings of which at least one ring - with this (or these) ring(s) then not being aromatic - contains one or more heteroatoms from the group consisting of nitrogen, oxygen and/or sulfur in the ring; preferably it is a 5 to 18 membered mono or polycyclic heterocyclic ring system of one or two rings of which one or both rings - with this one or two rings then not being aromatic - contain/s one or more heteroatoms selected from the group consisting of nitrogen, oxygen and/or sulfur in the ring, more preferably it is selected from oxazepam, pyrrolidine, piperidine, piperazine, tetrahydropyran, morpholine, indoline, oxopyrrolidine, benzodioxane, especially is piperazine, benzodioxane, morpholine, te
  • heterocyclyl is defined as a 4 to 18 membered mono or polycyclic heterocyclic ring system of one or more saturated or unsaturated rings of which at least one ring contains one or more heteroatoms selected from the group consisting of nitrogen, oxygen and/or sulfur in the ring.
  • it is a 5 to 18 membered mono or polycyclic heterocyclic ring system of one or two saturated or unsaturated rings of which at least one ring contains one or more heteroatoms selected from the group consisting of nitrogen, oxygen and/or sulfur in the ring. More preferably, it is a 5 to 10 membered mono or bicyclic heterocyclyl ring system containing one nitrogen atom and optionally a second heteroatom selected from nitrogen and oxygen.
  • said heterocyclyl is a substituted mono or bicyclic heterocyclyl ring system.
  • heterocyclyls include oxazepam, pyrrolidine, imidazole, oxadiazole, tetrazole, pyridine, pyrimidine, piperidine, piperazine, benzofuran, benzimidazole, indazole, benzodiazole, thiazole, benzothiazole, tetrahydropyran, morpholine, indoline, furan, triazole, isoxazole, pyrazole, thiophene, benzothiophene, pyrrole, pyrazine, pyrrolo[2,3b]pyridine, quinoline, isoquinoline, tetrahydroisoquinoline, phthalazine, benzo-1 ,2,5-thiadiazole, indole, benzotriazole, benzoxazole ox
  • the ring system is defined first as an aromatic heterocyclyl (heteroaryl) if at least one aromatic ring contains a heteroatom. If no aromatic ring contains a heteroatom, then the ring system is defined as a non-aromatic heterocyclyl if at least one non-aromatic ring contains a heteroatom. If no non-aromatic ring contains a heteroatom, then the ring system is defined as an aryl if it contains at least one aryl cycle. If no aryl is present, then the ring system is defined as a cycloalkyl if at least one non- aromatic cyclic hydrocarbon is present.
  • aryl-alkyl is understood as meaning an aryl group (see above) being connected to another atom through a Ci- 6 -alkyl (see above) which may be branched or linear and is unsubstituted or substituted once or several times.
  • aryl-alkyl is understood as meaning an aryl group (see above) being connected to another atom through 1 to 4 (-CH2-) groups.
  • alkylheterocyclyl is understood as meaning an heterocyclyl group (see above) being connected to another atom through a Ci- 6 -alkyl (see above) which may be branched or linear and is unsubstituted or substituted once or several times.
  • alkylheterocyclyl is understood as meaning an heterocyclyl group (see above) being connected to another atom through 1 to 4 (-CH2-) groups.
  • alkylcycloalkyl is understood as meaning an cycloalkyl group (see above) being connected to another atom through a Ci- 6 -alkyl (see above) which may be branched or linear and is unsubstituted or substituted once or several times.
  • alkylcycloalkyl is understood as meaning a cycloalkyl group (see above) being connected to another atom through 1 to 4 (-CH2-) groups.
  • the aryl is a monocyclic aryl. More preferably the aryl is a 6 or 7 membered monocyclic aryl. Even more preferably the aryl is a 6 membered monocyclic aryl, preferably phenyl.
  • the heteroaryl is a monocyclic heteroaryl. More preferably the heteroaryl is a 5, 6 or 7 membered monocyclic heteroaryl. Even more preferably the heteroaryl is a 5 or 6 membered monocyclic heteroaryl.
  • the non-aromatic heterocyclyl is a monocyclic non-aromatic heterocyclyl. More preferably the non-aromatic heterocyclyl is a 4, 5, 6 or 7 membered monocyclic non-aromatic heterocyclyl. Even more preferably the non-aromatic heterocyclyl is a 5 or 6 membered monocyclic non-aromatic heterocyclyl. In another preferred embodiment, said non-aromatic heterocyclyl is a bicyclic non-aromatic heterocyclyl.
  • the cycloalkyl is a monocyclic cycloalkyl. More preferably the cycloalkyl is a 3, 4, 5, 6, 7 or 8 membered monocyclic cycloalkyl. Even more preferably the cycloalkyl is a 3, 4, 5 or 6 membered monocyclic cycloalkyl.
  • a ring system is a system consisting of at least one ring of connected atoms but including also systems in which two or more rings of connected atoms are joined with“joined” meaning that the respective rings are sharing one (like a spiro structure), two or more atoms being a member or members of both joined rings.
  • leaving group means a molecular fragment that departs with a pair of electrons in heterolytic bond cleavage.
  • Leaving groups can be anions or neutral molecules. Common anionic leaving groups are halides such as CI-, Br-, and I-, and sulfonate esters, such as tosylate (TsO-), mesylate, nosylate or triflate.
  • salt is to be understood as meaning any form of the active compound used according to the invention in which it assumes an ionic form or is charged and is coupled with a counter-ion (a cation or anion) or is in solution.
  • a counter-ion a cation or anion
  • complexes of the active compound with other molecules and ions in particular complexes via ionic interactions.
  • the definition particularly includes physiologically acceptable salts, this term must be understood as equivalent to “pharmacologically acceptable salts”.
  • physiologically acceptable salt means in the context of this invention any salt that is physiologically tolerated (most of the time meaning not being toxic- especially lacking toxicity caused by the counter-ion) if used appropriately for a treatment especially if used on or applied to humans and/or mammals.
  • physiologically acceptable salts can be formed with cations or bases and in the context of this invention is understood as meaning salts of at least one of the compounds used according to the invention - usually a (deprotonated) acid - as an anion with at least one, preferably inorganic, cation which is physiologically tolerated - especially if used on humans and/or mammals.
  • the salts of the alkali metals and alkaline earth metals are particularly preferred, and also those with NH 4 , but in particular (mono)- or (di)sodium, (mono)- or (di)potassium, magnesium or calcium salts.
  • Physiologically acceptable salts can also be formed with anions or acids and in the context of this invention is understood as meaning salts of at least one of the compounds used according to the invention as the cation with at least one anion which are physiologically tolerated - especially if used on humans and/or mammals.
  • the salt formed with a physiologically tolerated acid that is to say salts of the particular active compound with inorganic or organic acids which are physiologically tolerated - especially if used on humans and/or mammals.
  • physiologically tolerated salts of particular acids are salts of: hydrochloric acid, hydrobromic acid, sulfuric acid, methanesulfonic acid, formic acid, acetic acid, oxalic acid, succinic acid, malic acid, tartaric acid, mandelic acid, fumaric acid, lactic acid or citric acid.
  • the compounds of the invention may be present in crystalline form or in the form of free compounds like a free base or acid.
  • solvate any compound that is a solvate of a compound according to the invention like a compound according to formula (I) defined above is understood to be also covered by the scope of the invention. Methods of solvation are generally known within the art. Suitable solvates are pharmaceutically acceptable solvates.
  • the term “solvate” according to this invention is to be understood as meaning any form of the active compound according to the invention in which this compound has attached to it via non- covalent binding another molecule (most likely a polar solvent). Especially preferred examples include hydrates and alcoholates, like methanolates or ethanolates.
  • prodrug is used in its broadest sense and encompasses those derivatives that are converted in vivo to the compounds of the invention. Such derivatives would readily occur to those skilled in the art, and include, depending on the functional groups present in the molecule and without limitation, the following derivatives of the present compounds: esters, amino acid esters, phosphate esters, metal salts sulfonate esters, carbamates, and amides. Examples of well known methods of producing a prodrug of a given acting compound are known to those skilled in the art and can be found e.g. in Krogsgaard-Larsen et al.“Textbook of Drug design and Discovery” Taylor & Francis (April 2002).
  • the compounds of the invention are also meant to include compounds which differ only in the presence of one or more isotopically enriched atoms.
  • compounds having the present structures except for the replacement of a hydrogen by a deuterium or tritium, or the replacement of a carbon by 13 C- or 14 C- enriched carbon or of a nitrogen by 15 N-enriched nitrogen are within the scope of this invention.
  • the compounds of formula (I) as well as their salts or solvates are preferably in pharmaceutically acceptable or substantially pure form.
  • pharmaceutically acceptable pure form is meant, inter alia, having a pharmaceutically acceptable level of purity excluding normal pharmaceutical additives such as diluents and carriers, and including no material considered toxic at normal dosage levels.
  • Purity levels for the drug substance are preferably above 50%, more preferably above 70%, most preferably above 90%. In a preferred embodiment it is above 95% of the compound of formula (I), or of its salts. This applies also to its solvates or prodrugs.
  • the compound of formula (I) is a compound wherein
  • W3 is S and W 4 is C;
  • Y is -C(O)-, -[C(RaRa')]n-, -CRa(CN)- or -CR a (CONRa'Ra ")-.
  • R a , R a ⁇ and R a” are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl, and substituted or unsubstituted C 2-6 alkynyl; and n is 0, 1 , 2 or 3;
  • X is selected from a bond, -[C(RbRb )] -, -[C(RbRb )] C(0)[C(Rb"Rb )]q- > substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl-C(O), substituted or unsubstituted aryl- alkyl, wherein each R b , R b ⁇ , R b - and R b - are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl, and substituted or unsubstituted C 2-6 alkynyl; and p is 0, 1 , 2 or 3; q is 0, 1 , 2 or 3;
  • Group A is a substituted or unsubstituted 5 to 10 membered mono or bicyclic heterocyclyl containing one nitrogen atom and optionally a second heteroatom selected from N and O, wherein A is attached to Y through a nitrogen atom; each R1 and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl, substituted or unsubstituted C 2-6 alkynyl, substituted or unsubstituted alkyl-cycloalkyl; NR 4 R 4' , C(0)0R 4 and substituted or unsubstituted haloalkyl; wherein each R 4 and R 4' are independently selected from hydrogen, halogen, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl, substituted or unsubstituted C 2-6 alkynyl, and C(0)0R
  • R 5 is selected from hydrogen, halogen, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl, substituted or unsubstituted C 2-6 alkynyl, CN, -C(0)0R 3 and -S(0) 2 0R 3' ; wherein each R 3 and R 3 ⁇ are independently selected from hydrogen, halogen, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl, and substituted or unsubstituted C 2-6 alkynyl;
  • R 6 is selected from hydrogen, halogen, substituted or unsubstituted C 1-6 alkyl (tert-butyl), substituted or unsubstituted C 2-6 alkenyl, substituted or unsubstituted C 2-6 alkynyl,
  • R 2 is a group of the following formula
  • Wi is selected from C or N; each Rg, Rg ⁇ , Rg- and Rg- are independently selected from hydrogen, substituted or unsubstituted Ci-6 alkyl, substituted or unsubstituted C 2-6 alkenyl and substituted or unsubstituted C2- 6 alkynyl; alternatively, Rg and Rg ⁇ and/or Rg- and Rg- taken together with the carbon atom to which they are attached may form a substituted or unsubstituted cycloalkyl; alternatively, Rg and Rg ⁇ and/or Rg- and Rg- taken together with the carbon atom to which they are attached may form a carbonyl group; each R10, Rio ⁇ , R10” and Rio- are independently selected from hydrogen, substituted or unsubstituted C1- 6 alkyl, substituted or unsubstituted C2- 6 alkenyl and substituted or unsubstituted C2- 6 alkynyl; alternatively, R1 0 , R-io
  • R 7 is selected from substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted aryl and substituted or unsubstituted aromatic heterocyclyl;
  • Rs is selected from hydrogen, substituted or unsubstituted C1- 6 alkyl, substituted or unsubstituted C2- 6 alkenyl and substituted or unsubstituted C2- e alkynyl;
  • R 7 and Rs taken together with -N(Rs )-[CH2]t- atoms to which they are attached form a substituted or unsubstituted heterocyclyl optionally fused with a substituted or unsubstituted aryl group; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound according to the invention of formula (I) is a compound wherein
  • Y is -C(O)-, -[C(R a R a )] n -, -CR a (CN)- or -CR a (CONR a R a )-, wherein each R a , R a ⁇ and R a - are independently selected from hydrogen, halogen, substituted or unsubstituted Ci- 6 alkyl, substituted or unsubstituted C2-6 alkenyl, and substituted or unsubstituted C2-6 alkynyl; and n is 0, 1 , 2 or 3; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound according to the invention of formula (I) is a compound wherein
  • Y is -C(O)-, -[C(R a R a )] n -, -CR a (CN)- or -CR a (CONR a R a )-, wherein each R a , R a ⁇ and R a - are hydrogen, n is 0, 1 or 2; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound according to the invention of formula (I) is a compound wherein
  • Y is -C(O)- optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound according to the invention of formula (I) is a compound wherein
  • X is selected from a bond, -[C(RbRb )] P -, -[C(RbRb )] P C(0)[C(Rb"Rb )] q -, unsubstituted aryl, preferably phenyl, unsubstituted aryl-C(O), preferably phenyl-C(O) and unsubstituted aryl-alkyl, preferably phenyl-alkyl, and wherein each Rb, Rb ⁇ , Rb- and Rb- are hydrogen, and p is 0, 1 or 2; q is 0, 1 or 2; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound according to the invention of formula (I) is a compound wherein
  • -X-R 2 is selected from -[C(RbRb)] P -R2, -[C(RbRb)] P C(0)[C(Rb"Rb )] q -R2, -aryl-R 2, preferably -phenyl-R 2 and -aryl-C(0)-R 2 , preferably -phenyl-C(0)-R 2 , -aryl-alkyl-R 2 , preferably -phenyl-alkyl-R 2 , wherein each Rb, Rb ⁇ , Rb- and Rb- are hydrogen; and p is 0, 1 or 2; q is 0, 1 or 2; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof
  • the compound according to the invention of formula (I) is a compound wherein group A is selected from: wherein b is 0, 1 or 2; and
  • W 2 is selected from O, C and N; each R1 and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C1- 6 alkyl, substituted or unsubstituted C2- 6 alkenyl, substituted or unsubstituted C2- 6 alkynyl, substituted or unsubstituted alkyl-cycloalkyl; NR4R4', - C(0)0R 4 - and substituted or unsubstituted haloalkyl; wherein each R 4 and R 4' are independently selected from hydrogen, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl, substituted or unsubstituted C 2-6 alkynyl, and -C(0)OR 4 ⁇ -; wherein R 4 " is substituted or unsubstituted C 1-6 alkyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastere
  • the compound according to the invention of formula (I) is a compound wherein when W 2 is N and R 1 or Rr is NR 4 R 4' ; then NR 4 R 4' is not atacthed to W 2 .
  • the compound according to the invention of formula (I) is a compound wherein each R 1 and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted alkyl-cycloalkyl; NR 4 R 4' , and substituted or unsubstituted haloalkyl; wherein each R 4 and R 4 ⁇ are independently selected from hydrogen and substituted or unsubstituted C 1-6 alkyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers
  • the alkyl, alkenyl or alkynyl, also in alkyl-cycloalkyl and haloalkyl defined in R1 or Rr, if substituted, is substituted with one or more substituent/s selected from -O R51 , halogen, and -NRsiRsr; wherein each of R51 or R 5 r is independently selected from hydrogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2-6 alkynyl. In a more preferred embodiment, each of R51 orR 5 r is selected from hydrogen and unsubstituted C1-6 alkyl, more preferably methyl.
  • the compound according to the invention of formula (I) is a compound wherein group A is
  • b is 1 or 2;
  • W 2 is N; and each R 1 and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C 1-6 alkyl, unsubstituted alkyl-cycloalkyl; preferably alkyl-cyclopropyl, and substituted or unsubstituted haloalkyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound according to the invention of formula (I) is a compound wherein group A is
  • W 2 is N; and each R1 and Rr are independently selected from hydrogen, halogen, preferably fluor, substituted or unsubstituted C1-6 alkyl, preferably methyl, substituted or unsubstituted alkyl-cycloalkyl; preferably alkyl-cyclopropyl, and substituted or unsubstituted haloalkyl; preferably -CH 2 F, -CH 2 -CH 2 F, -CH 2 -CHF 2 , or -CHF 2 .
  • halogen preferably fluor, substituted or unsubstituted C1-6 alkyl, preferably methyl, substituted or unsubstituted alkyl-cycloalkyl; preferably alkyl-cyclopropyl, and substituted or unsubstituted haloalkyl; preferably -CH 2 F, -CH 2 -CH 2 F, -CH 2 -CHF 2 , or -CHF 2 .
  • the compound according to the invention of formula (I) is a compound wherein group A is
  • R1 is hydrogen; and Rr is substituted or unsubstituted Ci-6 alkyl, and wherein Rr is directly attached to W 2 .
  • the compound according to the invention of formula (I) is a compound wherein group A is
  • each Ri and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C alkyl; and substituted or unsubstituted haloalkyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound according to the invention of formula (I) is a compound wherein group A is
  • each Ri and Rr are independently selected from hydrogen and unsubstituted C1- 6 alkyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound according to the invention of formula (I) is a compound wherein group A is each Ri and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C alkyl; and substituted or unsubstituted haloalkyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • group A is each Ri and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C alkyl; and substituted or unsubstituted haloalkyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers,
  • the compound according to the invention of formula (I) is a compound wherein group A is
  • each Ri and Rr are independently selected from hydrogen and unsubstituted C1- 6 alkyl. optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound according to the invention of formula (I) is a compound wherein R 2 is a group of the following formula
  • R7 is selected from substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted aryl and substituted or unsubstituted aromatic heterocyclyl; Re is selected from hydrogen and substituted or unsubstituted C1-6 alkyl;
  • Re ⁇ is selected from hydrogen and substituted or unsubstituted C1-6 alkyl; or
  • the compound according to the invention of formula (I) is a compound wherein R 2 is a group of the following formula: wherein m is 1 ; r is 1 ; t is 0;
  • Wi is C; each Rg, Rg ⁇ , Rg- and Rg- are hydrogen; each R10, Rio ⁇ , Rio- and Rio- are hydrogen;
  • R 7 is substituted or unsubstituted aryl; preferably phenyl;
  • R 2 is a group of the following formula
  • Wi is N; each Rg, Rg ⁇ , Rg- and Rg- are hydrogen; each Rio, Rio ⁇ , Rio” and Rio- are hydrogen;
  • R 7 is substituted or unsubstituted aryl; preferably phenyl;
  • Rs is substituted or unsubstituted C 1-6 alkyl; preferably methyl, and
  • Rs ⁇ is substituted or unsubstituted C 1-6 alkyl; preferably methyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound according to the invention of formula (I) is a compound wherein R 7 is selected from substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted aryl, preferably phenyl, and substituted or unsubstituted aromatic heterocyclyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the alkyl, aryl and aromatic heterocyclyl as defined in R 7 if substituted, is substituted with one or more substituent/s selected from -OR 71 and halogen, wherein
  • R 71 is selected from hydrogen, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl and substituted or unsubstituted C 2-6 alkynyl. In a more preferred embodiment, R 71 is selected from hydrogen and unsubstituted C 1-6 alkyl, preferably C 1-3 alkyl, more preferably methyl.
  • the compound according to the invention of formula (I) is a compound wherein R 7 is C 1-6 alkyl; more preferably C 1-3 alkyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound according to the invention of formula (I) is a compound wherein R 7 is substituted or unsubstituted aryl; more preferably, R 7 is phenyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the aryl as defined in R 7 if substituted, is substituted with one or more substituent/s selected from -OR 7 I and halogen, preferably fluor, wherein
  • R 7 I is selected from hydrogen, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl and substituted or unsubstituted C 2-6 alkynyl. In a more preferred embodiment, R 7 I is selected from hydrogen and unsubstituted C 1-6 alkyl, preferably C 1-3 alkyl, more preferably methyl.
  • the compound according to the invention of formula (I) is a compound wherein Re is selected from hydrogen, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl and substituted or unsubstituted C 2-6 alkynyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • Re is selected from hydrogen, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl and substituted or unsubstituted C 2-6 alkynyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in
  • the C 1-6 alkyl as defined in Re if substituted, is substituted with one or more substituent/s selected from substituted or unsubstituted C 1-6 alkyl, preferably unsubstituted C 1-3 alkyl, and halogen, preferably fluor.
  • the compound according to the invention of formula (I) is a compound wherein Re ⁇ is selected from hydrogen, substituted or unsubstituted Ci-6 alkyl, substituted or unsubstituted C 2-6 alkenyl and substituted or unsubstituted C 2-6 alkynyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • Re ⁇ is selected from hydrogen, substituted or unsubstituted Ci-6 alkyl, substituted or unsubstituted C 2-6 alkenyl and substituted or unsubstituted C 2-6 alkynyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in
  • the C 1-6 alkyl as defined in Re ⁇ if substituted, is substituted with one or more substituent/s selected from substituted or unsubstituted C 1-6 alkyl, preferably unsubstituted C 1-3 alkyl, and halogen, preferably fluor.
  • the compound according to the invention of formula (I) is a compound wherein R 7 and Rs taken together with -N(Rs')-[CH 2 ] t - atoms to which they are attached may form a substituted or unsubstituted heterocyclyl optionally fused with a substituted or unsubstituted aryl group; more preferably said heterocyclyl is an unsubstituted heterocyclyl fused with an unsubstituted aryl group; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound according to the invention of formula (I) is a compound wherein R 7 and Rs taken together with -N(Rs')-[CH 2 ] t - atoms to which are attached is
  • Wi and Rs ⁇ are as defined above, preferably Wi is N and Rs ⁇ is Ci-6 alkyl, preferably methyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound according to the invention of formula (I) is a compound wherein when Wi is N then X is not a bond.
  • the compound of the invention according to formula (I) is a compound, wherein: one of W 3 and W 4 is S and the other is C; preferably, W 3 is S and W 4 is C;
  • Y is -C(O)-, -[C(RaRa')]n-, -CRa(CN)- or -CRa(CONRa'Ra' ')-, wherein each R a , R a ⁇ and R a - are hydrogen and n is 0, 1 or 2; and/or
  • X is selected from a bond, -[C(RbRb )] P -, -[C(RbRb )] P C(0)[C(Rb”Rb )]q- > unsubstituted aryl, preferably phenyl, unsubstituted aryl-C(O), preferably phenyl-C(O) and unsubstituted aryl-alkyl, preferably phenyl-alkyl, and wherein each R b , R b ⁇ , R b - and R b - are hydrogen, p is 0, 1 or 2; q is 0, 1 or 2; and/or group A is selected from:
  • W 2 is selected from O, C and N; each R1 and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted alkyl-cycloalkyl; NR4R4', -C(0)OR 4 - and substituted or unsubstituted haloalkyl; wherein each R 4 and R 4' are independently selected from hydrogen, substituted or unsubstituted C 1-6 alkyl and -C(0)OR 4 ⁇ -; wherein R 4 " is substituted or unsubstituted C 1-6 alkyl; and/or
  • R 5 is selected from hydrogen, halogen, substituted or unsubstituted C 1-6 alkyl, CN, - C(0)0R3 and -S(0)20R3 ⁇ ; wherein each R 3 and R 3' are hydrogen; and/or
  • R 6 is selected from hydrogen, halogen and substituted or unsubstituted C 1-6 alkyl; and/or
  • Wi is C or N; each Rg, Rg ⁇ , Rg- and Rg- are hydrogen; each R10, Rio ⁇ , R10” and Rio- are hydrogen; R7 is selected from substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted aryl, preferably phenyl, and substituted or unsubstituted aromatic heterocyclyl;
  • Re is selected from hydrogen and substituted or unsubstituted C1-6 alkyl
  • Re is selected from hydrogen and substituted or unsubstituted C1-6 alkyl; or
  • R7 and Re taken together with -N(R8')-[CH2] t - atoms to which they are attached may form a substituted or unsubstituted heterocyclyl optionally fused with a substituted or unsubstituted aryl group; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound of the invention according to formula (I) is a compound, wherein:
  • W3 is S and W 4 is C;
  • Y is -C(O)-, -[C(RaRa')]n-, -CRa(CN)- or -CR a (CONRa'Ra ")-. wherein each R a , R a ⁇ and R a - are hydrogen and n is 0, 1 or 2; and/or
  • X is selected from a bond, -[C(RbRb )] P -, -[C(RbRb )] P C(0)[C(Rb”Rb )]q- > unsubstituted aryl, preferably phenyl, unsubstituted aryl-C(O), preferably phenyl-C(O) and unsubstituted aryl-alkyl, preferably phenyl-alkyl, and wherein each R b , R b ⁇ , R b - and R b - are hydrogen, p is 0, 1 or 2; q is 0, 1 or 2; and/or group A is selected from: wherein b is 0, 1 or 2; and
  • W2 is selected from O, C and N; each Ri and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted alkyl-cycloalkyl; NR 4 R 4' , -C(0)0R 4 - and substituted or unsubstituted haloalkyl; wherein each R4 and R4' are independently selected from hydrogen, substituted or unsubstituted C1-6 alkyl and -C(0)0R 4 ⁇ -; wherein R4" is substituted or unsubstituted C1-6 alkyl; and/or
  • R5 is selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, CN, - C(0)0R3 and -S(0)20R3 ⁇ ; wherein each R3 and R3' are hydrogen; and/or
  • R 6 is selected from hydrogen, halogen and substituted or unsubstituted C1-6 alkyl; and/or
  • R7 is selected from substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted aryl, preferably phenyl, and substituted or unsubstituted aromatic heterocyclyl;
  • Re is selected from hydrogen and substituted or unsubstituted C1-6 alkyl
  • Re is selected from hydrogen and substituted or unsubstituted C1-6 alkyl; or
  • R7 and Re taken together with -N(R8')-[CH2] t - atoms to which they are attached may form a substituted or unsubstituted heterocyclyl optionally fused with a substituted or unsubstituted aryl group; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound is a compound, wherein in R a , R a and R a - as defined in any of the embodiments of the present invention, the C 1-6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2-methylpropyl; and/or
  • the C 2-6 -alkenyl is preferably selected from ethylene, propylene, butylene, pentylene, hexylene, isopropylene and isobutylene; and/or
  • the C 2-6 -alkynyl is preferably selected from ethyne, propyne, butyne, pentyne, hexyne, isopropyne and isobutyne; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound is a compound, wherein in R b , R b ⁇ , R b - and R b - as defined in any of the embodiments of the present invention, the C 1-6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl, and 2-methylpropyl; and/or
  • the C 2-6 -alkenyl is preferably selected from ethylene, propylene, butylene, pentylene, hexylene, isopropylene and isobutylene; and/or
  • the C 2-6 -alkynyl is preferably selected from ethyne, propyne, butyne, pentyne, hexyne, isopropyne and isobutyne; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound is a compound, wherein in Ri and Rr as defined in any of the embodiments of the present invention, the C 1-6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl, and 2-methylpropyl; and/or
  • the C 2-6 -alkenyl is preferably selected from ethylene, propylene, butylene, pentylene, hexylene, isopropylene and isobutylene; and/or
  • the C 2-6 -alkynyl is preferably selected from ethyne, propyne, butyne, pentyne, hexyne, isopropyne and isobutyne; and/or
  • the cycloalkyl is C 3-8 cycloalkyl like cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, or cyclooctyl; preferably is C 3-7 cycloalkyl like cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl; more preferably from C 3-6 cycloalkyl like cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl; more preferably the cycloalkyl is cyclopropyl; and/or
  • the haloalkyl is selected from -CH 2 CI, -CH 2 F, -CHC , -CHF 2 , -CCI 3 , -CF 3 and -CH2-CHCI2; more preferably the haloalkyl is -CHF2 or -CF3;
  • stereoisomers optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound is a compound, wherein in R 4 and R 4 as defined in any of the embodiments of the present invention, the C 1-6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl, and 2-methylpropyl; and/or
  • the C2-6 -alkenyl is preferably selected from ethylene, propylene, butylene, pentylene, hexylene, isopropylene and isobutylene; and/or
  • the C2-6 -alkynyl is preferably selected from ethyne, propyne, butyne, pentyne, hexyne, isopropyne and isobutyne; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound is a compound, wherein in R 5 as defined in any of the embodiments of the present invention, the alkyl is preferably selected from C1-6 alkyl; more preferably, the Ci- 6 alkyl is selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2- methylpropyl; and/or
  • the C2-6 -alkenyl is preferably selected from ethylene, propylene, butylene, pentylene, hexylene, isopropylene and isobutylene; and/or
  • the C2-6 -alkynyl is preferably selected from ethyne, propyne, butyne, pentyne, hexyne, isopropyne and isobutyne;
  • -C(0)0R 3 or -S(0) 2 0R 3' are preferably -C(0)0H and -S(0) 2 0H; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound is a compound, wherein in R 6 as defined in any of the embodiments of the present invention, the alkyl is preferably selected from C1-6 alkyl; more preferably, the Ci- 6 alkyl is selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2- methylpropyl; and/or
  • the C2-6 -alkenyl is preferably selected from ethylene, propylene, butylene, pentylene, hexylene, isopropylene and isobutylene; and/or
  • the C2-6 -alkynyl is preferably selected from ethyne, propyne, butyne, pentyne, hexyne, isopropyne and isobutyne;
  • stereoisomers optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound is a compound, wherein in R 7 as defined in any of the embodiments of the present invention, the alkyl is preferably selected from C1-6 alkyl; more preferably, the C1-6 alkyl is selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2- methylpropyl; and/or
  • the aryl is selected from phenyl, naphtyl, or anthracenyl; preferably is napthyl and phenyl; and/or
  • the heterocyclyl is a heterocyclic ring system of one or more saturated or unsaturated rings of which at least one ring contains one or more heteroatoms from the group consisting of nitrogen, oxygen and/or sulfur in the ring; preferably is a heterocyclic ring system of one or two saturated or unsaturated rings of which at least one ring contains one or more heteroatoms from the group consisting of nitrogen, oxygen and/or sulfur in the ring, more preferably is selected from oxazepan, pyrrolidine, imidazole, oxadiazole, tetrazole, pyridine, pyrimidine, piperidine, piperazine, benzofuran, benzimidazole, indazole, benzothiazole, benzodiazole, thiazole, benzothiazole, tetrahydropyrane, morpholine, indoline, furan, triazole, isoxazole, pyrazole, thiophene, benzothiophene,
  • stereoisomers optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound is a compound, wherein in Re and Re ⁇ as defined in any of the embodiments of the present invention, the alkyl is preferably selected from C1-6 alkyl; more preferably, the Ci- 6 alkyl is selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2- methylpropyl; and/or
  • the C2-6 -alkenyl is preferably selected from ethylene, propylene, butylene, pentylene, hexylene, isopropylene and isobutylene; and/or
  • the C2-6 -alkynyl is preferably selected from ethyne, propyne, butyne, pentyne, hexyne, isopropyne and isobutyne;
  • stereoisomers optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound is a compound, wherein in Rg, Rg ⁇ , Rg- and Rg- as defined in any of the embodiments of the present invention, the alkyl is preferably selected from C 1-6 alkyl; more preferably, the Ci- 6 alkyl is selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2- methylpropyl; and/or
  • the C 2-6 -alkenyl is preferably selected from ethylene, propylene, butylene, pentylene, hexylene, isopropylene and isobutylene; and/or
  • the C 2-6 -alkynyl is preferably selected from ethyne, propyne, butyne, pentyne, hexyne, isopropyne and isobutyne;
  • stereoisomers optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound is a compound, wherein in R1 0 , R-io ⁇ , R-io- and Rio- as defined in any of the embodiments of the present invention, the alkyl is preferably selected from C 1-6 alkyl; more preferably, the C 1-6 alkyl is selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2- methylpropyl; and/or
  • the C 2-6 -alkenyl is preferably selected from ethylene, propylene, butylene, pentylene, hexylene, isopropylene and isobutylene; and/or
  • the C 2-6 -alkynyl is preferably selected from ethyne, propyne, butyne, pentyne, hexyne, isopropyne and isobutyne;
  • stereoisomers optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound is a compound, wherein in R a , R a and R a - as defined in any of the embodiments of the present invention, the C 1-6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2-methylpropyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound is a compound, wherein in R b , R b ⁇ , R b - and R b - as defined in any of the embodiments of the present invention, the C 1-6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2-methylpropyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound is a compound, wherein in Ri and Rr as defined in any of the embodiments of the present invention, the C 1-6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2-methylpropyl; and/or
  • the cycloalkyl is C 3-8 cycloalkyl like cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, or cyclooctyl; preferably is C 3-7 cycloalkyl like cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl; more preferably from C 3-6 cycloalkyl like cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl; more preferably the cycloalkyl is cyclopropyl; and/or
  • the haloalkyl is selected from -CH 2 CI, -CH 2 F, -CHC , -CHF 2 , -CCI 3 , -CF 3 and -CH2-CHCI2; more preferably the haloalkyl is -CHF2 or -CF3;
  • stereoisomers optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound is a compound, wherein in R 4 and R 4 as defined in any of the embodiments of the present invention, the C 1-6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2-methylpropyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the C 1-6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2-methylpropyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or
  • the compound is a compound, wherein in R 5 as defined in any of the embodiments of the present invention, the alkyl is preferably selected from C 1-6 alkyl; more preferably, the C 1-6 alkyl is selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2- methylpropyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the alkyl is preferably selected from C 1-6 alkyl; more preferably, the C 1-6 alkyl is selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2- methylprop
  • the compound is a compound, wherein in R 6 as defined in any of the embodiments of the present invention, the alkyl is preferably selected from C alkyl; more preferably, the Ci- 6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2-methylpropyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the alkyl is preferably selected from C alkyl; more preferably, the Ci- 6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2-methylpropyl
  • the compound is a compound, wherein in Re and Re ⁇ as defined in any of the embodiments of the present invention, the alkyl is preferably selected from Ci- 6 alkyl; more preferably, the Ci- 6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2-methylpropyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the alkyl is preferably selected from Ci- 6 alkyl; more preferably, the Ci- 6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl
  • the compound is a compound, wherein in Rg, Rg ⁇ , Rg- and Rg- as defined in any of the embodiments of the present invention, the alkyl is preferably selected from Ci- 6 alkyl; more preferably, the Ci- 6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2-methylpropyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the alkyl is preferably selected from Ci- 6 alkyl; more preferably, the Ci- 6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl,
  • the compound is a compound, wherein in R-io, R-io ⁇ , R-io- and Rio” as defined in any of the embodiments of the present invention, the alkyl is preferably selected from C alkyl; more preferably, the C alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2-methylpropyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the alkyl is preferably selected from C alkyl; more preferably, the C alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexy
  • the compound is a compound, wherein one of W 3 and W 4 is S and the other is C;
  • Y is -C(O)-, -[C(RaRa')]n-, -CRa(CN)- or -CR a (CONRa'Ra ")-. wherein each R a , R a ⁇ and R a - are hydrogen and n is 0, 1 or 2; and
  • X is selected from a bond, -[C(RbRb )] P -, -[C(RbRb )] P C(0)[C(Rb”Rb )]q- > unsubstituted aryl, preferably phenyl, unsubstituted aryl-C(O), preferably phenyl-C(O) and unsubstituted aryl-alkyl, preferably phenyl-alkyl, and wherein each Rb, Rb ⁇ , Rb- and Rb- are hydrogen; p is 0, 1 or 2; q is 0, 1 or 2; and group A is selected from: wherein b is 0, 1 or 2; and
  • W 2 is selected from O, C and N; each Ri and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted alkyl-cycloalkyl; NR 4 R 4' , -C(0)0R 4 - and substituted or unsubstituted haloalkyl; and wherein each R 4 and R 4' are independently selected from hydrogen, substituted or unsubstituted C 1-6 alkyl, , and -C(0)0R 4 ⁇ -; wherein R 4 " is substituted or unsubstituted C 1-6 alkyl;
  • R 5 is selected from hydrogen, halogen, substituted or unsubstituted C 1-6 alkyl, CN, - C(0)0R3; -S(0)20R3 ⁇ ; wherein each R 3 and R 3' are hydrogen,
  • R 6 is selected from hydrogen, halogen and substituted or unsubstituted C 1-6 alkyl
  • R 2 is a group of the following formula:
  • Wi is C or N; each Rg, Rg ⁇ , Rg- and Rg- are hydrogen; each R10, Rio ⁇ , R10” and Rio- are hydrogen;
  • R 7 is selected from substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted aryl, preferably phenyl, and substituted or unsubstituted aromatic heterocyclyl;
  • Re is selected from hydrogen and substituted or unsubstituted C 1-6 alkyl
  • Re is selected from hydrogen and substituted or unsubstituted C 1-6 alkyl; or
  • R 7 and Re taken together with -N(R 8' )-[CH 2 ] t - atoms to which they are attached may form a substituted or unsubstituted heterocyclyl optionally fused with a substituted or unsubstituted aryl group; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound is a compound, wherein
  • W 3 is S and W 4 is C;
  • Y is -C(O)-, -[C(R a R a )] n -, -CR a (CN)- or -CR a (CONR a R a )-, wherein each R a , R a ⁇ and R a - are hydrogen and n is 0, 1 or 2; and X is selected from a bond, -[C(RbRb )] P -, -[C(RbRb )] P C(0)[C(Rb"Rb )] q -, unsubstituted aryl, preferably phenyl, unsubstituted aryl-C(O), preferably phenyl-C(O) and unsubstituted aryl-alkyl, preferably phenyl-alkyl, and wherein each Rb, Rb ⁇ , Rb- and Rb- are hydrogen; p is 0, 1 or 2; q is 0, 1 or 2; and group A is selected
  • W 2 is selected from O, C and N; each Ri and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted alkyl-cycloalkyl; NR4R4', -C(0)OR 4 - and substituted or unsubstituted haloalkyl; and wherein each R4 and R4' are independently selected from hydrogen, substituted or unsubstituted C1-6 alkyl, , and -C(0)OR 4 ⁇ -; wherein R4" is substituted or unsubstituted C1-6 alkyl;
  • R5 is selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, CN, - C(0)OR3; -S(0) 2 0R 3' ; wherein each R 3 and Ry are hydrogen,
  • R 6 is selected from hydrogen, halogen and substituted or unsubstituted C1-6 alkyl
  • R2 is a group of the following formula:
  • Wi is C or N; each Rg, Rg ⁇ , Rg- and Rg- are hydrogen; each Rio, Rio ⁇ , Rio” and Rio- are hydrogen;
  • R7 is selected from substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted aryl, preferably phenyl, and substituted or unsubstituted aromatic heterocyclyl;
  • Re is selected from hydrogen and substituted or unsubstituted C1-6 alkyl
  • Re is selected from hydrogen and substituted or unsubstituted C1-6 alkyl
  • R7 and Re taken together with -N(R8')-[CH2] t - atoms to which they are attached may form a substituted or unsubstituted heterocyclyl optionally fused with a substituted or unsubstituted aryl group; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound is a compound, wherein
  • W3 is S and W 4 is C;
  • Y is -C(O)-, -[C(R a Ra )]n-, -CR a (CN)- or -CR a (CONR a R a )-, wherein each R a , R a ⁇ and R a - are hydrogen and n is 0, 1 or 2; and X is selected from a bond, -[C(RbRb )] P -, -[C(RbRb )] P C(0)[C(Rb”Rb )]q- > phenyl, preferably phenyl-C(O) and phenyl-alkyl, and wherein each R b , R b ⁇ , R b - and R b - are hydrogen, p is 0, 1 or 2; q is 0, 1 or 2; and group A is:
  • W 2 is selected from O, C and N; and each R1 and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C alkyl, substituted or unsubstituted alkyl-cycloalkyl; NR4R4', -C(0)OR 4 - and substituted or unsubstituted haloalkyl; wherein each R 4 and R 4 ⁇ are independently selected from hydrogen, substituted or unsubstituted C 1-6 alkyl, , and -C(0)0R 4” -; wherein R 4 - is substituted or unsubstituted C 1-6 alkyl; and R 5 is selected from hydrogen, halogen, substituted or unsubstituted C 1-6 alkyl, CN, - C(0)0R3 and -S(0)20R3 ⁇ ; wherein each R 3 and R 3' are hydrogen; and
  • R 6 is selected from hydrogen and substituted or unsubstituted C 1-6 alkyl
  • R 2 is a group of the following formula
  • Wi is C or N; each Rg, Rg ⁇ , Rg- and Rg- are hydrogen; each R10, Rio ⁇ , R10” and Rio- are hydrogen;
  • R 7 is selected from substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted aryl, preferably phenyl, and substituted or unsubstituted aromatic heterocyclyl; Re is selected from hydrogen and substituted or unsubstituted Ci- 6 alkyl;
  • Re is selected from hydrogen and substituted or unsubstituted Ci- 6 alkyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound is a compound, wherein
  • W 3 is S and W 4 is C;
  • Y is -C(O)-
  • X is selected from a bond and -[C(R b R b )] P -, wherein each R b , R b ⁇ are hydrogen and p is 1 or 2; and group A is:
  • each R1 and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted alkyl-cycloalkyl; NR4R4', -C(0)0R 4 - and substituted or unsubstituted haloalkyl; and each R 4 and R 4 ⁇ are independently selected from hydrogen, substituted or unsubstituted C1-6 alkyl, , and -C(0)0R 4” -; wherein R 4 - is substituted or unsubstituted Ci- 6 alkyl; and
  • R5 is selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, CN, - C(0)0R 3 and -S(0) 2 0R 3 ⁇ ; wherein each R3 and R3' are hydrogen; and
  • R 6 is selected from hydrogen and substituted or unsubstituted C1-6 alkyl
  • R2 is a group of the following formula
  • Wi is C; each Rg, Rg ⁇ , Rg- and Rg- are hydrogen; each R10, Rio ⁇ , R10” and Rio- are hydrogen;
  • R7 is substituted or unsubstituted aryl, preferably phenyl
  • Re is selected from hydrogen and substituted or unsubstituted Ci- 6 alkyl
  • Re is selected from hydrogen and substituted or unsubstituted C1-6 alkyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the halogen is fluorine or chlorine.
  • stereoisomers optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
  • the compound of formula (I) is selected from:
  • the obtained reaction products may, if desired, be purified by conventional methods, such as crystallization and chromatography. Where the processes described below for the preparation of compounds of the invention give rise to mixtures of stereoisomers, these isomers may be separated by conventional techniques such as preparative chromatography.
  • the compounds may be prepared in racemic form, or individual enantiomers may be prepared either by enantiospecific synthesis or by resolution.
  • One preferred pharmaceutically acceptable form of a compound of the invention is the crystalline form, including such form in pharmaceutical composition.
  • the additional ionic and solvent moieties must also be non-toxic.
  • the compounds of the invention may present different polymorphic forms, it is intended that the invention encompasses all such forms.
  • the compounds of formula (I) can be obtained by following the methods described below. As it will be obvious to one skilled in the art, the exact method used to prepare a given compound may vary depending on its chemical structure.
  • Method A represents a first process for synthesizing compounds according to formula
  • the invention refers to a process for the preparation of a compound of formula (I)
  • Ri, Rr, R 2 , R 5 , R6, W 3 , W 4 , X, Y and A have the same meaning as indicated before and Z is selected from OH, Cl, N(OMe)Me and H.
  • W 3 is S and W 4 is C.
  • Z is selected from OH and Cl.
  • the reaction is carried out using a suitable coupling reagent such as N- (3-dimethylaminopropyl)-/V'-ethylcarbodiimide (EDC), dicyclohexylcarbodiimide (DCC), /V-[(dimethylamino)-1 H- 1 ,2,3-triazolo-[4,5-b]pyridin-1 -ylmethylene]-/V- methylmethanaminium hexafluorophosphate N- oxide (HATU) or L/,L/,L/',L/'-tetramethyl- 0-(1H- benzotriazol-1 -yl)uronium hexafluorophosphate (HBTU), optionally in the presence of 1 -hydroxybenzotriazole, optionally in the presence of an organic base such as /V-methylmorpholine or /V,/V-diisopropylethylamine, in a suitable solvent such as
  • the reaction is carried out in a suitable solvent, such as dichloromethane, tetrahydrofuran, ethyl acetate or ethyl acetate-water mixtures; in the presence of an organic base such as triethylamine or /V,/V-diisopropylethylamine or an inorganic base such as K 2 CO 3 ; and at a suitable temperature, preferably comprised between 0 °C and room temperature.
  • a suitable solvent such as dichloromethane, tetrahydrofuran, ethyl acetate or ethyl acetate-water mixtures
  • an organic base such as triethylamine or /V,/V-diisopropylethylamine or an inorganic base such as K 2 CO 3
  • an activating agent such as 4-dimethylaminopyridine can be used.
  • a suitable reagent such as alane
  • a suitable solvent such as tetrahydrofuran
  • a suitable reagent such as sodium hydroxide and hydrogen peroxide
  • a compound of formula V, where Z represents O-alkyl can be prepared by treating a compound of formula II with a suitable hydrazine of formula III, as shown in Scheme 1 , in the presence of a suitable solvent, such as ethanol, at a suitable temperature, preferably heating.
  • a suitable solvent such as ethanol
  • a compound of formula V, where Z represents OH can be prepared by reacting a compound of formula V, where Z represents O-alkyl, in basic media, such as sodium hydroxide in the presence of a suitable solvent, such as water, at a suitable temperature, between room temperature and 100 °C and optionally under microwave heating.
  • basic media such as sodium hydroxide
  • a suitable solvent such as water
  • a compound of formula V, where Z represents OH may be converted to a compound of formula V, where Z represents a chlorine atom, using thionyl chloride, in a suitable solvent, such as pyridine, at a suitable temperature, such as room temperature
  • a compound of formula V, where Z represents hydrogen can be prepared by reduction of a compound of formula V, where Z represents O-alkyl, using a suitable reagent, such as didisobutylaluminiumhydride in the presence of a suitable solvent, such as tetrahydrofuran at a suitable temperature, such as between -10 °C and room temperature.
  • a suitable reagent such as didisobutylaluminiumhydride
  • a suitable solvent such as tetrahydrofuran at a suitable temperature, such as between -10 °C and room temperature.
  • a compound of formula V, where Z represents hydrogen can be prepared using a two-step procedure that involves conversion of a compound of formula V, where Z represents OH, to a compound of formula V where Z represents N(OMe)Me, which is then reduced.
  • a compound of formula V, where Z represents N(OMe)Me can be prepared by treating a compound of formula V, where Z represents OH, with N,O-dimethylhydroxylamine using a suitable coupling reagent such as /V-(3-dimethylaminopropyl)-/V'- ethylcarbodiimide (EDO), dicyclohexylcarbodiimide (DCC), /V-[(dimethylamino)-1 /-/- 1 ,2,3-triazolo-[4,5-b] pyridin-1 -ylmethylene]-/V-methylmethanaminium
  • a suitable coupling reagent such as /V-(3-dimethylaminopropyl)-/V'- ethylcarbodiimide (EDO), dicyclohexylcarbodiimide (DCC), /V-[(dimethylamino)-1 /-/- 1 ,2,3-
  • HATU hexafluorophosphate N- oxide
  • HBTU hexafluorophosphate N- oxide
  • 1- hydroxybenzotriazole optionally in the presence of 1- hydroxybenzotriazole
  • an organic base such as N- methylmorpholine or /V,/V-diisopropylethylamine
  • a suitable solvent such as dichloromethane or dimethylformamide
  • a compound of formula V, where Z represents hydrogen can be prepared by reduction of a compound of formula V, where Z represents N(OMe)Me using a suitable reagent, such as lithiumaluminium hydride in the presence of a suitable solvent, such as tetrahydrofuran at a suitable temperature, such as 0 °C.
  • a suitable reagent such as lithiumaluminium hydride
  • a suitable solvent such as tetrahydrofuran at a suitable temperature, such as 0 °C.
  • the compound of the invention can be produced according to the process explained below.
  • another aspect of the present invention relates to a process for the preparation of a compound of formula (I)
  • R1, Rr, R 2 , Rs, R6, W3, W 4 , X, Y and A have the same meaning as indicated before; and Z is OH or O-alkyl; and b) treating the compound obtained in step a) with a compound of formula XI
  • W 3 is S and W 4 is C.
  • W 4 is C.
  • G is a B(OH)2 group and the reaction of XII with XI is carried out in the presence of a copper salt, such as Cu(OAC)2, in a suitable solvent, such as dichloromethane, at a suitable temperature, such as room temperature.
  • a copper salt such as Cu(OAC)2
  • a suitable solvent such as dichloromethane
  • G is a leaving group and the alkylation reaction may be carried out under alkylation conditions, in a suitable solvent, such as dimethylacetamide, isopropanol, ethanol or acetonitrile; optionally in the presence of an organic base such as triethylamine or diisopropylethylamine or an inorganic base such as K2CO 3 or CS2CO 3 ; at a suitable temperature comprised between room temperature and the reflux temperature, preferably heating, or alternatively, the reactions can be carried out in a microwave reactor.
  • a suitable solvent such as dimethylacetamide, isopropanol, ethanol or acetonitrile
  • an organic base such as triethylamine or diisopropylethylamine or an inorganic base such as K2CO 3 or CS2CO 3
  • the reactions can be carried out in a microwave reactor.
  • Method C represents a third process for synthesizing compounds according to formula
  • the invention refers to a process for the preparation of a compound of formula (I)
  • step b) treating the compound obtained in step a) with R2H; wherein Z is O-alkyl, OH or a leaving group (LG).
  • W3 is S and W 4 is C.
  • a compound of formula XVI, where M represents OH, may be converted to a compound of formula XVII, where M represents a leaving group, such as chlorine.
  • the reaction may be carried out using thionyl chloride in a suitable solvent, such as dichloromethane, at a suitable temperature, such as room temperature.
  • the compound of formula I, wherein Y represents -C(O)- may be obtained by reaction of a compound of formula XVII with a suitable reagent of formula XVIII using alkylation conditions such as the described above.
  • the compounds of formula II may be prepared, as described in Scheme 3, by reacting a compound of formula XIX with a compound of formula XX, where Z is O-alkyl, in the presence of a base such as lithiumhexamethyldisilazane in a suitable solvent such as tetrahydrofuran at a suitable temperature, such as room temperature.
  • a base such as lithiumhexamethyldisilazane
  • a suitable solvent such as tetrahydrofuran
  • the invention refers to the use of a compound selected from
  • R 2 , Rs, R 6 , W 3 , W 4 and X have the same meaning as indicated above and Z is O-alkyl, OH; H or Cl for the manufacture of a compound a compound according to the invention as described above according to formula (I).
  • W 3 is S and W 4 is C.
  • Certain compounds of the present invention can also be obtained starting from other compounds of formula (I) by appropriate conversion reactions of functional groups, in one or several steps, using well-known reactions in organic chemistry under standard experimental conditions.
  • some of these conversions include the reduction of a double bond from a tetrahydropyridine to a piperidine derivative or the reductive amination of an amino group with an aldehyde or ketone, or alternatively the reaction of an amino group with an alkylating agent, to prepare a further substituted amino group.
  • a compound of formula (I) that shows chirality can also be obtained by resolution of a racemic compound of formula (I) either by chiral preparative HPLC or by crystallization of a diastereomeric salt or co-crystal.
  • the resolution step can be carried out at a previous stage, using any suitable intermediate.
  • Another aspect of the invention refers to a pharmaceutical composition which comprises a compound according to the invention as described above according to formula (I) or a pharmaceutically acceptable salt thereof, prodrug, solvate or steroisomer thereof, and a pharmaceutically acceptable carrier, adjuvant or vehicle.
  • the present invention thus provides pharmaceutical compositions comprising a compound of this invention, or a pharmaceutically acceptable salt, prodrug, solvate or stereoisomers thereof together with a pharmaceutically acceptable carrier, adjuvant, or vehicle, for administration to a patient.
  • compositions include any solid (tablets, pills, capsules, granules etc.) or liquid (solutions, suspensions or emulsions) composition for oral, topical or parenteral administration.
  • the pharmaceutical compositions are in oral form, either solid or liquid.
  • Suitable dose forms for oral administration may be tablets, capsules, syrops or solutions and may contain conventional excipients known in the art such as binding agents, for example syrup, acacia, gelatin, sorbitol, tragacanth, or polyvinylpyrrolidone; fillers, for example lactose, sugar, maize starch, calcium phosphate, sorbitol or glycine; tabletting lubricants, for example magnesium stearate; disintegrants, for example starch, polyvinylpyrrolidone, sodium starch glycollate or microcrystalline cellulose; or pharmaceutically acceptable wetting agents such as sodium lauryl sulfate.
  • binding agents for example syrup, acacia, gelatin, sorbitol, tragacanth, or polyvinylpyrrolidone
  • fillers for example lactose, sugar, maize starch, calcium phosphate, sorbitol or
  • the solid oral compositions may be prepared by conventional methods of blending, filling or tabletting. Repeated blending operations may be used to distribute the active agent throughout those compositions employing large quantities of fillers. Such operations are conventional in the art.
  • the tablets may for example be prepared by wet or dry granulation and optionally coated according to methods well known in normal pharmaceutical practice, in particular with an enteric coating.
  • compositions may also be adapted for parenteral administration, such as sterile solutions, suspensions or lyophilized products in the apropriate unit dosage form.
  • Adequate excipients can be used, such as bulking agents, buffering agents or surfactants.
  • Administration of the compounds or compositions of the present invention may be by any suitable method, such as intravenous infusion, oral preparations, and intraperitoneal and intravenous administration. Oral administration is preferred because of the convenience for the patient and the chronic character of the diseases to be treated.
  • an effective administered amount of a compound of the invention will depend on the relative efficacy of the compound chosen, the severity of the disorder being treated and the weight of the sufferer.
  • active compounds will typically be administered once or more times a day for example 1 , 2, 3 or 4 times daily, with typical total daily doses in the range of from 0.1 to 1000 mg/kg/day.
  • the compounds and compositions of this invention may be used with other drugs to provide a combination therapy.
  • the other drugs may form part of the same composition, or be provided as a separate composition for administration at the same time or at different time.
  • Another aspect of the invention refers to a compound of formula (I) as described above, or a pharmaceutical acceptable salt or isomer thereof for use in therapy.
  • Another aspect of the invention refers to a compound of formula I, or a pharmaceutically acceptable salt or isomer thereof, for use in the treatment or prophylaxis of pain.
  • the pain is medium to severe pain, visceral pain, chronic pain, cancer pain, migraine, inflammatory pain, acute pain or neuropathic pain, allodynia or hyperalgesia. This may include mechanical allodynia or thermal hyperalgesia.
  • Another aspect of the invention refers to the use of a compound of the invention in the manufacture of a medicament for the treatment or prophylaxis of pain.
  • the pain is selected from medium to severe pain, visceral pain, chronic pain, cancer pain, migraine, inflammatory pain, acute pain or neuropathic pain, allodynia or hyperalgesia, also preferably including mechanical allodynia or thermal hyperalgesia.
  • Another aspect of this invention relates to a method of treating or preventing pain which method comprises administering to a patient in need of such a treatment or prevention a therapeutically effective amount of a compound as above defined or a pharmaceutical composition thereof.
  • a compound as above defined or a pharmaceutical composition thereof is administered to a patient in need of such a treatment or prevention a therapeutically effective amount of a compound as above defined or a pharmaceutical composition thereof.
  • the pain syndromes that can be treated or prevented are medium to severe pain, visceral pain, chronic pain, cancer pain, migraine, inflammatory pain, acute pain or neuropathic pain, allodynia or hyperalgesia, whereas this could also include mechanical allodynia or thermal hyperalgesia.
  • DIPEA N,N-Diisopropylethylamine
  • Quantitative Rt Retention time rt: Room temperature Sat: Saturated TFA: T rifluoroacetic acid
  • THF Tetrahydrofuran
  • TEA EtsN
  • TOSMIC Toluenesulfonylmethyl isocyanide
  • Wt Weight
  • Method A Column Acquity UPLC BEH C18 2.1x50 mm, 1.7 mm; flow rate 0.61 mL/min; A: NH4HCO3 10 mM; B: ACN; Gradient: 0.3 min 98% A, 98% to 5% A in 2.52 min, isocratic 5% > A 1.02 min.
  • Method B Column Zodiac C18 50x4.6 mm 3 pm; flow rate 0.6 mL/min; A: 0.1 % formic acid in water; B: ACN; Gradient 95% to 50% A in 3 min, 50% to 5% > A in 2 min isocratic 5% A 2 min.
  • Method C Column Acquity UPLC BEH C18 2.1x50 mm, 1.1 pm; flow rate 0.80 mL/min; A: NH4HCO3 10 mM; B: ACN; Gradient: 0.3 min 90% A, 90% to 5% A in 2.7 min, isocratic 5% A 0.7 min.
  • Method D Column Aquity UPLC BEH C18 2.1 x 50 mm, 1.7 pm, flow rate 0.61 mL/min; A: NH4HCO3 10 mM, B: ACN, C: MeOH + 0.1% formic acid; gradient 0.3 min 98% A, 98% A to 0:95:5 A:B:C in 2.7 min; 0:95:5 A:B:C to 100%o B in 0.1 min; isocratic 2 min 100% B.
  • Method E Column Acquity UPLC BEH C18 2.1x50 mm, 1.7 pm; flow rate 0.61 mL/min; A: NH4HCO3 10 mM pH 10.6; B: ACN; Gradient: 0.3 min 98% > A, 98% > to 0% > A in 2.7 min, isocratic 0% > A 2 min.
  • Method F Column Acquity UPLC BEH C18 2.1x50 mm, 1.7 pm; flow rate 0.5 mL/min; A: NH4HCO3 10mM; B: ACN; Gradient: 90% > A to 5% > A in 4 min, 1 min in 5% > A, 5% > A to 90% > A in 0.1 min, 1.9 min in 90% > A
  • Method G Column Acquity UPLC BEH C18 2.1x50 mm, 1.7 pm; flow rate 0.80 mL/min; A: ACONH4 10 mM; B: ACN; Gradient: 0.3 min 90% > A, 90% > to 5% > A in 2.7 min, isocratic 5%o A 0.7 min.
  • Step a 8-(Dimethylamino)-1 ,4-dioxaspiro[4.5]decane-8-carbonitrile.
  • Step b N,N-Dimethyl-8-phenyl-1 ,4-dioxaspiro[4.5]decan-8-amine.
  • THF 100 ml.
  • Grignard solution of bromophenyl (245.64 g, 1 .36 mol) was added dropwise at 0 °C.
  • the reaction mixture was stirred at rt for 18 h and then quenched with sat aq NH4CI, extracted with EtOAc and washed with sat aq NaCI.
  • the combined organic layers were dried over anh Na2S0 4 , filtered and evaporated under reduced pressure.
  • the crude product was purified by flash chromatography silica gel, gradient CHC o CHC iMeOH (9:1 ) to give the title compound (70 g, Yield: 59%).
  • Step c 4-(Dimethylamino)-4-phenylcyclohexanone.
  • step b) To a stirred solution of the compound obtained in step b) (70 g, 0.27 mol) in MeOH (50 ml_), cone. HCI (350 ml.) was added drop wise at 0 °C. The reaction mixture was stirred at rt for 16 h. The reaction was diluted with H2O and washed with EtOAc. The aqueous layer was basified with NaOH 8 N and the product was extracted with CHCI3. The combined organic layers were washed with H2O and sat aq NaCI and dried over anh Na 2 S0 4 , filtered and evaporated to dryness to give the title compound (40 g, Yield: 68%).
  • Step d tert-Butyl 2-(4-(dimethylamino)-4-phenylcyclohexyl)hydrazinecarboxylate.
  • step c) To a stirred solution of the compound obtained in step c) (40 g, 0.184 mol) in MeOH (100 ml_), tert-butyl hydrazinecarboxylate (36.49 g, 276.49 mmol) in MeOH (400 ml.) and acetic acid (0.52 ml_, 9.22 mmol) was added at 0 °C. Then NaCNBHs (23.29 g, 0.37 mol) was added portion wise at 0 °C and the reaction mixture was stirred at rt for 16 h. The reaction mixture was evaporated and diluted with DCM, washed with H2O and sat aq NaCI.
  • Step e di-tert-Butyl 1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)hydrazine-1 ,2- dicarboxylate.
  • step d) To a stirred solution of the compound obtained in step d) (17.5 g, 52.39 mmol) in anhydrous DCM (200 ml_), TEA (20.92 g, 157.17 mmol) and B0C2O (13.70 ml_, 62.86 mmol) were added at 0 °C and the reaction mixture was stirred for 16 h at rt. The reaction mixture was poured into ice cold H2O and extracted with DCM. The combined organic layer were dried over anh Na 2 S0 4 , filtered and evaporated under reduced pressure. The crude product was purified by flash chromatography, silica gel, gradient Hexane to Hexane:EtOAc (7:3) to give the title compound (6.0 g, Yield: 26%).
  • step e To a stirred solution of the compound obtained in step e) (2.5 g, 5.96 mmol), in MeOH (25 ml_), cone. HCI (1.08 g, 29.83 mmol) was added dropwise at 0 °C. The reaction mixture was allowed to stir at rt for 16 h. The mixture was evaporated, co-distilled with toluene (3x 50 ml.) and dried under vacuum to give the title compound. (1.75 g, Yield: 89%).
  • Step a 4-(Dimethylamino)-4-phenylcyclohexanol.
  • step a) To a solution of the compound obtained in step a) (6.5 g; 29.7 mmol) in anh DCM (100 ml_), TEA (8.4 ml_, 60.3 mmol) was added dropwise at rt and the reaction mixture was cooled to 0 °C. Then DMAP (0.2 g, 1.6 mmol) and p-toluensulfonyl chloride (6.22 g, 32.6 mmol) were added at 0 °C and the reaction mixture was stirred at rt for 48 h. The reaction mixture was neutralized with sat aq NaHCCh and the product was extracted with DCM.
  • Step a (1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexanecarbonitrile.
  • Step b (1 r,4r)-Methyl 4-(dimethylamino)-4-phenylcyclohexanecarboxylate.
  • step a) To a solution of the compound obtained in step a) (3 g, 13.1 mmol) in MeOH (30 mL), TMSCI (33.6 ml, 263.1 mmol) was added at rt. The reaction mixture was stirred at rt for 20 h. Then sat aq NaHCOs was added dropwise at 0 °C, and the product was extracted with DCM and washed with sat aq NaCI (1x 10 mL). The organic layer was dried over any Na2S0 4 , filtered and evaporated under reduced pressure to give the title compound (2.9 g, Yield: 85%). Step c.
  • step c) To a stirred solution of the compound obtained in step c) (2.5 g, 10.7 mmol) in anhydrous DCM (30 ml_), TEA (4.5 ml_, 32.2 mmol) and DMAP (130 mg, 1.1 mmol) were added at 0 °C. Then p-toluensulfonyl chloride (2.45 g, 12.9 mmol) was added dropwise at 0 °C, and the reaction mixture was stirred at rt for 4 h. The reaction mixture was quenched with H2O (30 ml.) and extracted with DCM. . The organic layer was washed with sat aq NaCI, dried over anh Na 2 S0 4, filtered and evaporated under vacuum. The crude product was purified by flash chromatography silica gel, gradient DCM to DCM:MeOH (95:5), to give the title compound (2.3 g, Yield: 56%).
  • Step a Methyl 2-(4-(dimethylamino)-4-phenylcyclohexylidene)acetate.
  • DMF (12 ml_) methyl 2-(dimethoxyphosphoryl)acetate
  • potassium 2-methylpropan-2-olate 387.3 mg, 3.4 mmol
  • the organic layer were combined and dried over anh Na 2 S0 4 and the solvent was removed under vacuum to give the title compound (617.4 mg, Yield: 96%).
  • Step b 2-(4-(Dimethylamino)-4-phenylcyclohexylidene)ethanol.
  • LiAlhU 1 M in Et 2 0, 4.78 mL, 4.8 mmol
  • Et 2 0 6 mL
  • the reaction was quenched with ice and the product was extracted with EtOAc and Et 2 0.
  • the combined organic layers were dried over anh Na 2 S0 4, and the solvent was removed under vacuum to give the title compound (211.5 mg, Yield: 90%).
  • Step c 2-(4-(Dimethylamino)-4-phenylcyclohexyl)ethanol.
  • step c) A solution of the compound obtained in step c) (11 1.5 mg, 0.45 mmol) in anhDCM (6 mL), TEA (90 m ⁇ , 0.63 mmol) and DMAP (3.6 mg, 0.03 mmol) were added and the mixture was cooled at 0 °C.
  • p-Toluenesulfonyl chloride (103.1 mg, 0.54 mmol) was added and reaction mixture was allowed to reach rt and was stirred for 4 h. The reaction was quenched with water and the product was extracted with DCM. The combined organic layers were dried over anh Na 2 S0 4, and the solvent was removed under vacuum, to give the title compound (107mg, Yield: 59%).
  • Example 1 (1 -((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone.
  • Step a Ethyl 2-oxo-2-(4-oxo-4,5,6,7-tetrahydrobenzo[b]thiophen-5-yl)acetate.
  • Step b Ethyl 1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazole-3-carboxylate.
  • Step a) To a solution of the compound obtained in step a) (1 .1 g, 4.36 mmol) in EtOH (35 ml.) under argon atmosphere, Intermediate 1 (1 .12 g, 3.54 mmol) was added portion wise and the reaction was heated at 65 °C for 2.5 h. The mixture was cooled at room temperature and a solid precipitated. The solid was filtered and washed with EtOH to give the title compound (1.59 g, Yield: 96%).
  • Step c. 1 -((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazole-3-carboxylic acid.
  • step b) To a solution of the compound obtained in step b) (1.5 g, 3.4 mmol) in EtOH (15 ml.) under argon atmosphere, NaOH (680 mg, 17 mmol) was added and the reaction was heated at 85 °C for 3 h. The reaction was quenched with HCI (2 M in Et 2 0, 8.5 ml.) and solvents were evaporated under vacuum. The crude product was dissolved in DCM:water (1 :1 , 40 ml.) and the remaining solid was filtered, washed with water and dried under vacuum, to give the title compound (1.44 g, Yield: 85%).
  • step c) To a solution of the compound obtained in step c) (1 .23 g, 2.9 mmol) in anh DCM:DMF (1 :1 , 10 ml.) under argon atmosphere, EDC-HCI (1 .1 14 g, 5.81 mmol), HOBt (890.01 mg, 5.81 mmol) and TEA (1.21 ml_, 8.72 mmol) were added and the reaction was stirred at rt for 10 min. Then 1 -methylpiperazine (0.483 ml_, 4.36 mmol) was added dropwise and the reaction was stirred at rt overnight. Solvents were evaporated under vacuum and the crude product was dissolved in DCM and washed with H2O. The organic layer was dried over anh Na2S0 4 , filtered and concentrated to dryness to give the title compound (1 .035 g, Yield: 71 %).
  • Example 77 (1 -((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-1 H-thieno[2,3-g]indazol- 3-yl)(4-methylpiperazin-1 -yl)methanone.
  • DDQ 135.2 mg, 0.6 mmol
  • More DDQ was added (67.6 mg, 0.3 mmol) and the reaction mixture was heated at 100 °C for 1 .5 h.
  • Example 78 (7-Bromo-1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone.
  • step a) Starting from the compound obtained in step a) (300 mg, 1.29 mmol) and following the experimental procedure described in step a) of Example 1 , the title compound was obtained (425.8 mg, Yield: quantitative).
  • Step c Ethyl 7-bromo-1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazole-3-carboxylate.
  • step b) Starting from the compound obtained in step b) (425.8 mg, 1.29 mmol) and following the experimental procedure described in step b) of Example 1 , the title compound was obtained (607.0 mg, Yield: 89 %).
  • Step d 7-Bromo-1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazole-3-carboxylic acid.
  • step c) Starting from the compound obtained in step c) (607.0 mg, 1.15 mmol) and following the experimental procedure described in step c) of Example 1 , the title compound was obtained (574.7 mg, Yield: quantitative).
  • step d) Starting from the compound obtained in step d) (291.4 mg, 0.582 mmols) and following the experimental procedure described in step d) of Example 1 , the title compound was obtained (309.0 mg, Yield: 91%).
  • Example 79 1 -((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-3-(4-methylpiperazine-1 - carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazole-7-carbonitrile.
  • Example 78 To a solution of the compound obtained in Example 78 (50 mg, 0.09 mmol) in anhydrous DMF (1 ml.) under argon atmosphere, Zn(CN) 2 (10.1 mg, 0.09 mmol) and Pd(PPh3)4 (9.92 mg, 0.02 mmol) were added and the reaction mixture was heated under microwave irradiation (100 °C, 150 W, 45 min). The reaction was quenched with 2 N HCI and extracted with EtOAc. The organic layer was dried with anh Na 2 S0 4 and the solvent was removed under vacuum. The crude product was purified by flash chromatography silica gel, gradient DCM to MeOH, to give the title compound (10 mg, Yield: 22%).
  • Example 80 1-((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-3-(4-methylpiperazine-1- carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazole-7-carboxylic acid.
  • Example 81 1-((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-3-(4-methylpiperazine-1- carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazole-7-sulfonic acid.
  • Example 2 To a solution of the compound obtained in Example 1 (62.5 mg, 0.12 mmol) in anhydrous DCM (2 ml_), sulphuric acid (48.7 mg, 0.5 mmol) and acetic anhydride (152 mg, 1.5 mmol) were added drop wise at -10 °C. The mixture was stirred at rt for 16 h. The solvent was removed under vacuum and the crude product was purified by flash chromatography, silica gel gradient DCM to DCM:MeOH (85:15), to give the title compound (68.3 mg, Yield: 94%).
  • Example 82 (1-((1 r,4r)-4-((2-Fluoroethyl)methylamino)-4-phenylcyclohexyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone.
  • Example 84 (1 -((1 r,4r)-4-((2,2-Difluoroethyl)(methyl)amino)-4-phenylcyclohexyl)-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1 -yl)methanone.
  • Example 83 Starting from the compound obtained in Example 83 (86.2 mg, 0.155 mmol) and following the experimental procedure described in Example 77 the title compound was obtained (6 mg, Yield: 7%).
  • Example 85 (1 ,4-Diazepan-1 -yl)(1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)methanone.
  • Example 4 To a solution of the compound obtained in Example 4 (200 mg, 0.33 mmol) in anh DCM (2.7 ml_), TFA (345 mI_) was added drop wise at 0 °C and the reaction mixture was stirred overnight at rt. The mixture was neutralised with 20% aq NaOH, diluted with DCM and washed with sat aq NaCI. The organic layer was dried over anh Na 2 S0 4 , filtered and evaporated to dryness, to give the title compound (45 mg, Yield: 27%).
  • Example 97 (1 -((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-(2-fluoroethyl)piperazin-1-yl)methanone.
  • Example 99 (2-((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-2H- thieno[2,3-g]indazol-3-yl)(4-ethylpiperazin-1-yl)methanone.
  • Step a Ethyl 2-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-2H- thieno[3,2-e]isoindole-3-carboxylate.
  • Step b 2-((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-2H-thieno[3,2- e]isoindole-3-carboxylic acid.
  • step a) Starting from the product obtained in step a) (80 mg, 0.179 mmol) and following the experimental procedure described in step c) of Example 1 , the title compound was obtained (55 mg, Yield: 72%).
  • step b) Starting from the product obtained in step b) (55 mg, 0.128 mmol) and following the experimental procedure described in step d) of Example 1 , the title compound was obtained (57 mg, Yield: 87%).
  • Example 102 2-(1 -((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)-2-(4-methylpiperazin-1-yl)acetonitrile.
  • Step a 1-((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-N-methoxy-N-methyl-4,5- dihydro-1 H-thieno[2,3-g]indazole-3-carboxamide.
  • Step b 1-((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazole-3-carbaldehyde.
  • step a) To a solution of the compound obtained in step a) (66 mg, 0.142 mmols) in anh THF (2ml_) cooled at 0 °C, LiAII-U (1 M in THF, 0.17 ml_, 0.17 mmols) was added drop wise and the mixture was stirred for 30 min. The reaction was quenched with 10 % HCI and the mixture was neutralized with aq NaHCCh sat solution and extracted with EtOAc. The organic layer was dried over anh Na2S0 4 , filtered and concentrated to dryness, to give the title compound (53.6 mg, Yield: 93%).
  • Step c Title compound.
  • hhCbMeOH (1 :1 , 8 ml_)
  • Na2S2C>3 (17.1 mg, 0.09 mmol) was added and the mixture was stirred vigorously at rt for 2 h.
  • Methylpiperazine (18.02 mg, 0.18 mmol) was added, the mixture was stirred for 1 h and then KCN (23.4 mg, 0.36 mmol) was added and the mixture was stirred for 48 h more.
  • MeOH was removed under vacuum and the crude product was extracted with EtOAc, washed with sat NaCI and water. The organic layer was dried over anh Na 2 S0 4 , filtered and concentrated to dryness to obtain the title compound (77.0 mg, Yield: 93%).
  • Example 103 2-(1 -((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)-2-(4-methylpiperazin-1-yl)acetamide.
  • Example 102 To a solution of the compound obtained in Example 102 (48 mg, 0.093 mmol) in MeOH/DMSO (3:1.5, 4.5 ml_), NaOH (2.5 M, 74 mI_, 0.187 mmol) and H 2 0 2 (30%, 48 mI_, 0.466 mmol) were added and the mixture was stirred at rt for 30 min. Solvents were removed under vacuum and the residue was extracted with EtOAc). The organic layer was washed with H2O, dried over anh Na 2 S0 4 and filtered. The solvent was removed under vacuum to give the title compound (49.62 mg, Yield: quantitative).
  • Example 104 (1 r,4r)-4-(8-(tert-Butyl)-3-((4-methylpiperazin-1 -yl)methyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-1-yl)-N,N-dimethyl-1-phenylcyclohexanamine.
  • TFA 0.689 ml_, 9.01 mmol
  • Example 105 N,N-Dimethyl-1-(4-(3-((4-methylpiperazin-1-yl)methyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-1-yl)phenyl)-4-phenylpiperidin-4-amine.
  • Example 38 (195.5 mg, 0.337 mmols) was added at 0 °C and the mixture was stirred at 0 °C for 1.5 h.
  • the reaction mixture was quenched by drop wise addition of water (5ml_) and the mixture was extracted with EtOAc.
  • the combined organic layers were dried over anh Na 2 S0 4 , filtered and evaporated under reduced pressure.
  • the crude product was purified by flash chromatography AI 2 O 3 , using gradient DCM to DCM:MeOH (7:3), to give the title compound (108 mg, Yield: 57%).
  • Example. 107 (1-((4-(Dimethylamino)-4-phenylcyclohexyl)methyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone.
  • Step a Ethyl 4,5-dihydro-1 H-thieno[2,3-g]indazole-3-carboxylate.
  • Step b 4,5-Dihydro-1 H-thieno[2,3-g]indazole-3-carboxylic acid.
  • step a) Starting from the product obtained in step a) (955.2 mg, 3,85 mmol) and following the experimental procedure described in step c) of Example 1 , the title compound was obtained (727 mg, Yield: 86%).
  • Step c (4,5-Dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone.
  • the title compound was obtained (126 mg, Yield: 63%).
  • step c) To a solution of the compound obtained in step c) (60 mg, 0.2 mmol) in DMA (2 ml_), Intermediate 2 (99.2 mg, 0.24 mmol) and CS2CO3 (97 mg, 0.3 mmol) were added, and the mixture was heated at 100 °C for 2 h. The solvent was removed under vacuum and EtOAc was added. The organic layer was washed with water and brine and dried over anh Na 2 S0 4 and the solvent was removed under vacuum. The crude product was purified by flash chromatography S1O2, using gradient DCM to MeOH, to give the title compound (31 mg, Yield: 30%).
  • Example 108 (1 -(2-((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)ethyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1 -yl)methanone.
  • Example 109 (1 -(2-(4-(3-Hydroxyphenyl)-4-(methylamino)piperidin-1 -yl)ethyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1 -yl)methanone.
  • Step a Ethyl 1 -(2-hydroxyethyl)-4,5-dihydro-1 H-thieno[2,3-g]indazole-3-carboxylate.
  • Step b 1 -(2-Hydroxyethyl)-4,5-dihydro-1 H-thieno[2,3-g]indazole-3-carboxylic acid.
  • step a) Starting from the product obtained in step a) (5.87 g, 20.08 mmol) and following the experimental procedure described in step c) of Example 1 , the title compound was obtained (4.35 g, Yield: 82%).
  • Step c (1 -(2-Hydroxyethyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin- 1 -yl)methanone.
  • step b) Starting from the product obtained in step b) (1.5 g, 5.67 mmol) and following the experimental procedure described in step d) of Example 1 , the title compound was obtained (830 mg, Yield: 42%).
  • Step d (1 -(2-Chloroethyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin- 1 -yl)methanone.
  • step c) To a solution of the compound obtained in step c) (155 mg, 0.447 mmol) in anh DCM (25 ml.) under argon atmosphere, thionyl chloride (99.8 mI_, 1 .34 mmol) was added dropwise at 0 °C and the reaction mixture was stirred at rt overnight. The crude mixture was diluted with DCM and washed with H2O. The organic layer was dried over anh Na 2 S0 4 , filtered and concentrated to dryness to give the title compound (152 mg, Yield: 93%).
  • Example 118 1 -(4-(Dimethylamino)-4-phenylpiperidin-1 -yl)-2-(3-(4-methylpiperazine-1 - carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1 -yl)ethanone.
  • Step a 2-Chloro-1-(4-(dimethylamino)-4-phenylpiperidin-1-yl)ethanone.
  • step a) To a solution of the compound obtained in step a) in DMA (15 ml_), the product obtained in step c) (147.0 mg, 0.486 mmol) of Example 106 was added. Following the experimental procedure described in step d) of the same example, the title compound was obtained (60.5 mg, Yield: 23%).
  • Example 151 (1 -((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(3-(dimethylamino)azetidin-1 -yl)methanone.
  • Example 142 (30 mg, 0.061 mmol) dissolved in DCE (2 ml_), formaldehyde (17 mI, 0.613 mmol) and NaBH(OAc) 3 (39 mg, 0.184mmol) were added and the reaction mixture was irradiated at MW at 150 W for 5 min at 120 °C.
  • the reaction mixture was dissolved in DCM and washed three times with H2O. The organic layer was dried over Na2S0 4 filtered and evaporated under vacuum to give the title compound (32 mg, Yield: quant).
  • Example 152 1 -(4-(Methylamino)-4-phenylpiperidin-1 -yl)-2-(3-(4-methylpiperazine- 1 -carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1 -yl)ethanone.
  • Transfected CHO-K1 cell membranes (20 pg) were incubated with [ 3 H]-DAMGO (1 nM) in assay buffer containing Tris-HCI 50 mM, MgCI 2 5 mM at pH 7.4. NBS (non-specific binding) was measured by adding 10 mM Naloxone. The binding of the test compound was measured at five different concentrations. Plates were incubated at 27 °C for 60 min. After the incubation period, the reaction mixture was then transferred to Multiscreen HTS, FC plates (Millipore), filtered and plates were washed 3 times with ice-cold 10 mM Tris-HCI (pH 7.4). Filters were dried and counted at approximately 40% efficiency in a MicroBeta scintillation counter (Perkin-Elmer) using EcoScint liquid scintillation cocktail.
  • MicroBeta scintillation counter Perkin-Elmer
  • This invention is aimed at providing a series of compounds which act as dual ligands of the a 2 d subunit of voltage-gated calcium channels and the m-opioid receptor it is a very preferred embodiment in which the compounds are selected which act as dual ligands of the a 2 d subunit of voltage-gated calcium channels and the m-opioid receptor and especially compounds which have a binding expressed as K, responding to the following scales:
  • K ⁇ (a 2 d-1 ) is preferably ⁇ 10000 nM, more preferably ⁇ 5000 nM, or even more preferably ⁇ 500 nM.
  • K,(m) is preferably ⁇ 1000 nM, more preferably ⁇ 500 nM, even more preferably ⁇ 100 nM.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Pain & Pain Management (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)

Abstract

wherein the meanings for the various substituents are as disclosed in the description, having dual pharmacological activity towards both the α2δ subunit, in particular the α2δ-1 subunit, of the voltage-gated calcium channel and the μ-opioid receptor, to processes of preparation of such compounds, to pharmaceutical compositions comprising them, and to their use in therapy, in particular for the treatment of pain.

Description

4,5-DIHYDRO-1 H-THIENO[2,3-G]INDAZOLYL DERIVATIVES HAVING
MULTIMODAL ACTIVITY AGAINST PAIN
FIELD OF THE INVENTION
The present invention relates to compounds having dual pharmacological activity towards both the a2d subunit of the voltage-gated calcium channel, and the m-opioid receptor (MOR or mu-opioid receptor), and more particularly to 4,5-Dihydro-1 H- thieno[2,3-g]indazolyl derivatives having this pharmacological activity, to processes of preparation of such compounds, to pharmaceutical compositions comprising them, and to their use in therapy, in particular for the treatment of pain.
BACKGROUND OF THE INVENTION
The adequate management of pain constitutes an important challenge, since currently available treatments provide in many cases only modest improvements, leaving many patients unrelieved (Turk, D.C., Wilson, H.D., Cahana, A.; 201 1 ; Lancet ; 377; 2226- 2235). Pain affects a big portion of the population with an estimated prevalence of 20 % and its incidence, particularly in the case of chronic pain, is increasing due to the population ageing. Additionally, pain is clearly related to comorbidities, such as depression, anxiety and insomnia, which lead to important productivity losses and socio- economical burden (Goldberg, D.S., McGee, S.J.; 201 1 ; BMC Public Health ; 1 1 ; 770). Existing pain therapies include non-steroidal anti-inflammatory drugs (NSAIDs), opioid agonists, calcium channel blockers and antidepressants, but they are much less than optimal regarding their safety ratio. All of them show limited efficacy and a range of secondary effects that preclude their use, especially in chronic settings.
Voltage-gated calcium channels (VGCC) are required for many key functions in the body. Different subtypes of voltage-gated calcium channels have been described (Zamponi et al., Pharmacol. Rev. 2015 67:821 -70). The VGCC are assembled through interactions of different subunits, namely oti (Cavai), b (Cavp) a2d (Cava23) and g (Cavy). The oti subunits are the key porous forming units of the channel complex, being responsible for the Ca2+ conduction and generation of Ca2+ influx. The a2d, b, and g subunits are auxiliary, although very important for the regulation of the channel, since they increase the expression of the oti subunits in the plasma membrane as well as modulate their function, resulting in functional diversity in different cell types. Based on their physiological and pharmacological properties, VGCC can be subdivided into low voltage- activated T-type (Cav3.1 , Cav3.2, and Cav3.3), and high voltage-activated L- (Cav1 .1 through Cav1 .4), N-(Cav2.2), P/Q-(Cav2.1 ), and R-(Cav2.3) types, depending on the channel forming Cava subunits. All of these five subclasses are found in the central and peripheral nervous systems. Regulation of intracellular calcium through activation of these VGCC plays obligatory roles in: 1 ) neurotransmitter release, 2) membrane depolarization and hyperpolarization, 3) enzyme activation and inactivation, and 4) gene regulation (Perret and Luo, Neurotherapeutics. 2009 6:679-92; Zamponi et al., 2015 supra ; Neumaier et al., Prog. Neurobiol. 2015 129:1 -36.). A large body of data has clearly indicated that VGCC are implicated in mediating various disease states including pain processing. Drugs interacting with the different calcium channel subtypes and subunits have been developed. Current therapeutic agents include drugs targeting L-type Cav1 .2 calcium channels, particularly 1 ,4-dihydropyridines, which are widely used in the treatment of hypertension. T-type (Cav3) channels are the target of ethosuximide, widely used in absence epilepsy. Ziconotide, a peptide blocker of N-type (Cav2.2) calcium channels, has been approved as a treatment of intractable pain. (Perret and Luo, 2009, supra, Vink and Alewood, Br J Pharmacol. 2012 167:970-89.).
The Cav1 and Cav2 subfamilies contain an auxiliary a d subunit, which is the therapeutic target of the gabapentinoid drugs of value in certain epilepsies and chronic neuropathic pain. To date, there are four known a d subunits, each encoded by a unique gene and all possessing splice variants. Each a d protein is encoded by a single messenger RNA and is posttranslationally cleaved and then linked by disulfide bonds. Four genes encoding a d subunits have now been cloned. a2d-1 was initially cloned from skeletal muscle and shows a fairly ubiquitous distribution. The a2d-2 and a2d-3 subunits were subsequently cloned from brain. The most recently identified subunit, a2d-4, is largely nonneuronal. The human a2d-4 protein sequence shares 30, 32 and 61 % identity with the human a2d-1 , a2d-2 and a2d-3 subunits, respectively. The gene structure of all a2d subunits is similar. All a2d subunits show several splice variants (Davies et al., Trends Pharmacol Sci. 2007 28:220-8.; Dolphin AC, Nat Rev Neurosci. 2012 13:542-55., Biochim Biophys Acta. 2013 1828:1541 -9.).
The Cava26-1 subunit may play an important role in neuropathic pain development (Perret and Luo, 2009, supra ; Vink and Alewood, 2012, supra). Biochemical data have indicated a significant Cava26-1 , but not Cava26-2, subunit upregulation in the spinal dorsal horn, and DRG (dorsal root ganglia) after nerve injury that correlates with neuropathic pain development. In addition, blocking axonal transport of injury-induced DRG Cavot25-1 subunit to the central presynaptic terminals diminishes tactile allodynia in nerve injured animals, suggesting that elevated DRG Cavot25-1 subunit contributes to neuropathic allodynia.
The Cavot25-1 subunit (and the Cavot25-2, but not Cavot25-3 and Cavot25-4, subunits) is the binding site for gabapentin which has anti-allodynic/ hyperalgesic properties in patients and animal models. Because injury-induced Cavot25-1 expression correlates with neuropathic pain development and maintenance, and various calcium channels are known to contribute to spinal synaptic neurotransmission and DRG neuron excitability, injury-induced Cavot25-1 subunit upregulation may contribute to the initiation and maintenance of neuropathic pain by altering the properties and/or distribution of VGCC in the subpopulation of DRG neurons and their central terminals, therefore modulating excitability and/or synaptic neuroplasticity in the dorsal horn. Intrathecal antisense oligonucleotides against the Cavot25-1 subunit can block nerve injury-induced Cavot25-1 upregulation and prevent the onset of allodynia and reserve established allodynia.
As mentioned above, the 0,26 subunits of VGCC form the binding site for gabapentin and pregabalin, which are structural derivatives of the inhibitory neurotransmitter GABA although they do not bind to GABAA, GABAB, or benzodiazepine receptors, or alter GABA regulation in animal brain preparations. The binding of gabapentin and pregabalin to the Cavot25 subunit results in a reduction in the calcium-dependent release of multiple neurotransmitters, leading to efficacy and tolerability for neuropathic pain management. Gabapentinoids may also reduce excitability by inhibiting synaptogenesis (Perret and Luo, 2009, supra, Vink and Alewood, 2012, supra, Zamponi et al., 2015, supra).
As mentioned before, there are few available therapeutic classes for the treatment of pain, and opioids are among the most effective, especially when addressing severe pain states. They act through three different types of opioid receptors (mu, kappa and gamma) which are transmembrane G-protein coupled receptors (GPCRs). Still, the main analgesic action is attributed to the activation of the m-opioid receptor (MOR). However, the general administration of MOR agonists is limited due to their important side effects, such as constipation, respiratory depression, tolerance, emesis and physical dependence [Meldrum, M.L. (Ed.). Opioids and Pain Relief: A Historical Perspective. Progress in Pain Research and Management, Vol 25. IASP Press, Seattle, 2003]. Additionally, MOR agonists are not optimal for the treatment of chronic pain as indicated by the diminished effectiveness of morphine against chronic pain conditions. This is especially proven for the chronic pain conditions of neuropathic or inflammatory origin, in comparison to its high potency against acute pain. The finding that chronic pain can lead to MOR down-regulation may offer a molecular basis for the relative lack of efficacy of morphine in long-term treatment settings [Dickenson, A.H., Suzuki, R. Opioids in neuropathic pain: Clues from animal studies. Eur J Pain 9, 1 13-6 (2005)]. Moreover, prolonged treatment with morphine may result in tolerance to its analgesic effects, most likely due to treatment-induced MOR down-regulation, internalization and other regulatory mechanisms. As a consequence, long-term treatment can result in substantial increases in dosing in order to maintain a clinically satisfactory pain relief, but the narrow therapeutic window of MOR agonists finally results in unacceptable side effects and poor patient compliance.
Polypharmacology is a phenomenon in which a drug binds multiple rather than a single target with significant affinity. The effect of polypharmacology on therapy can be positive (effective therapy) and/or negative (side effects). Positive and/or negative effects can be caused by binding to the same or different subsets of targets; binding to some targets may have no effect. Multi-component drugs or multi-targeting drugs can overcome toxicity and other side effects associated with high doses of single drugs by countering biological compensation, allowing reduced dosage of each compound or accessing context-specific multitarget mechanisms. Because multitarget mechanisms require their targets to be available for coordinated action, one would expect synergies to occur in a narrower range of cellular phenotypes given differential expression of the drug targets than would the activities of single agents. In fact, it has been experimentally demonstrated that synergistic drug combinations are generally more specific to particular cellular contexts than are single agent activities, such selectivity is achieved through differential expression of the drugs’ targets in cell types associated with therapeutic, but not toxic, effects (Lehar et al., Nat Biotechnol 2009; 27: 659-666.).
In the case of chronic pain, which is a multifactorial disease, multi-targeting drugs may produce concerted pharmacological intervention of multiple targets and signaling pathways that drive pain. Because they actually make use of biological complexity, multi- targeting (or multi-component drugs) approaches are among the most promising avenues toward treating multifactorial diseases such as pain (Gilron et al., Lancet Neurol. 2013 Nov;12(1 1 ):1084-95.). In fact, positive synergistic interaction for several compounds, including analgesics, has been described (Schroder et al., J Pharmacol Exp Ther. 201 1 ; 337:312-20. Erratum in: J Pharmacol Exp Ther. 2012; 342:232; Zhang et al., Cell Death Dis. 2014; 5:e1 138.; Gilron et al., 2013, supra). Given the significant differences in pharmacokinetics, metabolisms and bioavailability, reformulation of drug combinations (multi-component drugs) is challenging. Further, two drugs that are generally safe when dosed individually cannot be assumed to be safe in combination. In addition to the possibility of adverse drug-drug interactions, if the theory of network pharmacology indicates that an effect on phenotype may derive from hitting multiple targets, then that combined phenotypic perturbation may be efficacious or deleterious. The major challenge to both drug combination strategies is the regulatory requirement for each individual drug to be shown to be safe as an individual agent and in combination (Hopkins, Nat Chem Biol. 2008; 4:682-90.).
An alternative strategy for multitarget therapy is to design a single compound with selective polypharmacology (multi-targeting drug). It has been shown that many approved drugs act on multiple targets. Dosing with a single compound may have advantages over a drug combination in terms of equitable pharmacokinetics and biodistribution. Indeed, troughs in drug exposure due to incompatible pharmacokinetics between components of a combination therapy may create a low-dose window of opportunity where a reduced selection pressure can lead to drug resistance. In terms of drug registration, approval of a single compound acting on multiple targets faces significantly lower regulatory barriers than approval of a combination of new drugs (Hopkins, 2008, supra).
Thus, the present application, relates to the advantages of having dual activity, for m- receptor and the a2d-1 subunit of voltage-gated calcium channels, in the same molecule to treat chronic pain.
In this way, the present invention relates to compounds having a complementary dual mechanism of action (m-receptor agonist and blocker of the a2d subunit, in particular the a2d-1 subunit, of voltage-gated calcium channels) which implies a better profile of tolerability than the strong opioids (morphine, oxycodone, fentanyl etc) and/or better efficacy and tolerability than gabapentinoids (pregabalin and gabapentin).
Pain is multimodal in nature, since in nearly all pain states several mediators, signaling pathways and molecular mechanisms are implicated. Consequently, monomodal therapies fail to provide complete pain relief. Currently, combining existing therapies is a common clinical practice and many efforts are directed to assess the best combination of available drugs in clinical studies (Mao, J., Gold, M.S., Backonja, M.; 201 1 ; J. Pain; 12; 157-166). Accordingly, there is still a need to find compounds that have an alternative or improved pharmacological activity in the treatment of pain, being both effective and showing the desired selectivity, and having good“drugability” properties, i.e. good pharmaceutical properties related to administration, distribution, metabolism and excretion. The inventors have found a series of compounds showing dual pharmacological activity towards both the a2d subunit, in particular the a2d-1 subunit, of the voltage-gated calcium channel, and the m-opioid receptor (MOR or mu-opioid receptor) resulting in an innovative, effective and alternative solution for the treatment of pain.
In view of the existing results of the currently available therapies and clinical practices, the present invention offers a solution by combining in a single compound binding to two different targets relevant for the treatment of pain. This was mainly achieved by providing the compounds according to the invention that bind both to the m-opioid receptor and to the a2d subunit, in particular the a2d-1 subunit, of the voltage-gated calcium channel.
SUMMARY OF THE INVENTION
The inventors have found a series of compounds, encompassed by formula (I), that show a dual pharmacological activity towards both the a2d subunit, in particular the a2d-1 subunit, of the voltage-gated calcium channel, and the m-opioid receptor thus solving the above problem of identifying alternative or improved pain treatments by offering such dual compounds.
The main object of the invention is directed to a compound having a dual activity binding to the a2d subunit, in particular the a2d-1 subunit, of the voltage-gated calcium channel and the m-opioid receptor. This compound can be used in the treatment of pain.
The invention is directed in a main aspect to a compound of formula (I),
Figure imgf000008_0001
wherein Ri, Rr R2, Rs, R6, W3, W4, X, Y and A are as defined below in the detailed description. A further aspect of the invention refers to the processes for preparation of compounds of formula (I).
A still further aspect of the invention refers to the use of intermediate compounds for the preparation of a compound of formula (I).
It is also an aspect of the invention a pharmaceutical composition comprising a compound of formula (I).
Finally, it is an aspect of the invention a compound of formula (I) for use in therapy and more particularly for the treatment of pain and pain related conditions.
DETAILED DESCRIPTION OF THE INVENTION The invention is directed to a family of compounds which have a dual pharmacological activity towards both the a2d subunit, in particular the a2d-1 subunit, of the voltage-gated calcium channel, and the m-opioid receptor and to their use in the treatment of pain and related disorders.
The applicant has surprisingly found that the problem of providing a new effective and alternative for treating pain and pain related disorders can be solved by using a multimodal balanced analgesic approach combining two different synergistic activities in a single drug (i.e., dual ligands which are bifunctional and bind to m-opioid receptor and to a2d subunit, in particular the a2d-1 subunit, of the voltage-gated calcium channel), thereby enhancing through the a2d blockade without increasing the undesirable side effects. This supports the therapeutic value of a dual agent, whereby the a2d binding component acts as an intrinsic adjuvant of the MOR binding component.
A dual compound that possess binding to both the m-opioid receptor and to the a2d subunit of the voltage-gated calcium channel shows a highly valuable therapeutic potential by achieving an outstanding analgesia (enhanced in respect to the potency of the opioid component alone) with a reduced side-effect profile (safety margin increased compared to that of the opioid component alone) versus existing opioid therapies.
Advantageously, the dual compounds according to the present invention would in addition show one or more the following functionalities: blockade of the a2d subunit, in particular the a2d-1 subunit, of the voltage-gated calcium channel and m-opioid receptor agonism
It has to be noted, though, that functionalities“antagonism” and“agonism” are also sub- divided in their effect into subfunctionalities like partial agonism or inverse agonism. Accordingly, the functionalities of the compound should be considered within a relatively broad bandwidth.
An antagonist blocks or dampens agonist-mediated responses. Known subfunctionalities are neutral antagonists or inverse agonists.
An agonist increases the activity of the receptor above its basal level. Known subfunctionalities are full agonists, or partial agonists.
In addition, the two mechanisms complement each other since MOR agonists are only marginally effective in the treatment of neuropathic pain, while the blockers of the a2d subunit, in particular the a2d-1 subunit, of voltage-gated calcium channels show outstanding effects in preclinical neuropathic pain models. Thus, the a2d component, in particular the a2d-1 component, adds unique analgesic actions in opioid-resistant pain. Finally, the dual approach has clear advantages over MOR agonists in the treatment of chronic pain as lower and better tolerated doses would be needed based on the potentiation of analgesia but not of the adverse events of MOR agonists. A further advantage of using designed multiple ligands is a lower risk of drug-drug interactions compared to cocktails or multi-component drugs, thus involving simpler pharmacokinetics and less variability among patients. Additionally, this approach may improve patient compliance and broaden the therapeutic application in relation to monomechanistic drugs, by addressing more complex aetiologies.
In a first aspect, the present invention is directed to a compound of formula (I):
Figure imgf000010_0001
wherein one of W3 and W4 is S and the other is C;
Y is -C(O)-, -[C(RaRa')]n-, -CRa(CN)- or -CRa(CONRa'Ra ")-. wherein each Ra, Ra· and Ra” are independently selected from hydrogen, halogen, substituted or unsubstituted Ci-6 alkyl, substituted or unsubstituted C2-6 alkenyl, and substituted or unsubstituted C2-6 alkynyl; and n is 0, 1 , 2 or 3;
X is selected from a bond, -[C(RbRb )] -, -[C(RbRb )] C(0)[C(Rb"Rb )]q-> substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl-C(O) and substituted or unsubstituted aryl- alkyl, wherein each Rb, Rb·, Rb- and Rb- are independently selected from hydrogen, halogen, substituted or unsubstituted Ci-6 alkyl, substituted or unsubstituted C2-6 alkenyl, and substituted or unsubstituted C2-6 alkynyl; and p is 0, 1 , 2 or 3; q is 0, 1 , 2 or 3;
Group A is a substituted or unsubstituted 4 to 10 membered mono or bicyclic heterocyclyl containing one nitrogen atom and optionally a second heteroatom selected from N and O, wherein A is attached to Y through a nitrogen atom; each R1 and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl, substituted or unsubstituted C2-6 alkynyl, substituted or unsubstituted alkyl-cycloalkyl; N R4R4', C(0)OR4 and substituted or unsubstituted haloalkyl; wherein each R4 and R4' are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl, substituted or unsubstituted C2-6 alkynyl, and C(0)OR4·; wherein R4 " is substituted or unsubstituted C1-6 alkyl;
R5 is selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6alkenyl, substituted or unsubstituted C2-6 alkynyl, CN, -C(0)OR3 and -S(0)20R3' ; wherein each R3 and Ry are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl, and substituted or unsubstituted C2-6 alkynyl;
R6 is selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2-6 alkynyl;
R2 is a group of the following formula:
Figure imgf000012_0001
wherein m is 0, 1 or 2; r is 0, 1 or 2; t is 0, 1 , 2 or 3;
Wi is selected from C and N; each Rg, Rg·, Rg- and Rg- are independently selected from hydrogen, substituted or unsubstituted Ci-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2-6 alkynyl; alternatively, Rg and Rg· and/or Rg- and Rg- taken together with the carbon atom to which they are attached may form a substituted or unsubstituted cycloalkyl; alternatively, Rg and Rg· and/or Rg- and Rg- taken together with the carbon atom to which they are attached may form a carbonyl group; each R10, Rio·, R10” and Rio- are independently selected from hydrogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2-6 alkynyl; alternatively, R10, Rio· and/or Rio ·, R10- taken together with the carbon atom to which they are attached may form a substituted or unsubstituted cycloalkyl; alternatively, Rio and Rio· and/or Rio- and Rio- taken together with the carbon atom to which they are attached may form a carbonyl group;
R7 is selected from substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted aryl and substituted or unsubstituted aromatic heterocyclyl;
Rs is selected from hydrogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2- e alkynyl;
Rs· is selected from hydrogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2- e alkynyl; alternatively, R7 and Rs taken together with -N(Rs')-[CH2]t- atoms to which they are attached form a substituted or unsubstituted heterocyclyl optionally fused with a substituted or unsubstituted aryl group; wherein the compound of formula (I) is optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
The compounds of the invention represented by the above described formula (I) may include enantiomers depending on the presence of chiral centres or isomers depending on the presence of multiple bonds. The single isomers, enantiomers or diastereoisomers and mixtures thereof fall within the scope of the present invention.
In another embodiment, these compounds according to the invention are optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt or solvate thereof.
For the sake of clarity the expression“a compound according to formula (I), wherein R1, Rr R2, Rs, R6, W3, W4, X, Y and A are as defined herein in the detailed description” would (just like the expression“a compound of formula (I) as defined in any one of claims 1 to 8 found in the claims) refer to“a compound according to formula (I)”, wherein the definitions of the respective substituents R1 etc. (also from the cited claims) are applied. For clarity purposes, all groups and definitions described in the present description and referring to compounds of formula (I), also apply to all intermediates of synthesis.
In addition, and for clarity purposes, it should further be understood that naturally if t is 0, the nitrogen is still present in R2.
In the context of this invention, alkyl is understood as meaning a straight or branched hydrocarbon chain radical containing no unsaturation, and which is attached to the rest of the molecule by a single bond. It may be unsubstituted or substituted once or several times. It encompasses e.g. -CH3 and -CH2-CH3. In these radicals, Ci-2-alkyl represents C1 - or C2-alkyl, Ci-3-alkyl represents C1 -, C2- or C3-alkyl, Ci-4-alkyl represents C1 -, C2, C3- or C4-alkyl, Ci-5-alkyl represents C1 -, C2-, C3-, C4-, or C5-alkyl and Ci-6-alkyl represents C1 -, C2-, C3-, C4-, C5- or C6-alkyl. Examples of alkyl radicals include among others methyl, ethyl, propyl, methylethyl, butyl, 1 -methylpropyl, 2-methylpropyl, 1 ,1 - dimethylethyl, pentyl, 1 ,1 -dimethylpropyl, 1 ,2-dimethylpropyl, 2,2-dimethylpropyl, hexyl, 1 -methylpentyl. If substituted by cycloalkyl, it corresponds to a“cycloalkylalkyl” radical, such as cyclopropylmethyl. If substituted by aryl, it corresponds to an "arylalkyl" radical, such as benzyl, benzhydryl or phenethyl. If substituted by heterocyclyl, it corresponds to a“heterocyclylalkyl” radical. Preferably alkyl is understood in the context of this invention Ci-6-alkyl like methyl, ethyl, propyl, butyl, pentyl, or hexyl; and more preferably is C1-4- alkyl like methyl, ethyl, propyl or butyl.
Alkenyl is understood as meaning straight or branched hydrocarbon chain radical containing at least two carbon atoms and at least one unsaturation, and which is attached to the rest of the molecule by a single bond. It may be unsubstituted or substituted once or several times. It encompasses groups like e.g. -CH=CH-CH3. The alkenyl radicals are preferably vinyl (ethenyl), allyl (2-propenyl). Preferably in the context of this invention alkenyl is C2-6-alkenyl like ethylene, propylene, butylene, pentylene, or hexylene; or is C2-4-alkenyl, like ethylene, propylene, or butylenes.
Alkynyl is understood as meaning a straight or branched hydrocarbon chain radical containing at least two carbon atoms and at least one carbon-carbon triple bond, and which is attached to the rest of the molecule by a single bond. It may be unsubstituted or substituted once or several times. It encompasses groups like e.g. -C^C-CHs (1 - propynyl). Preferably alkynyl in the context of this invention is C2-6-alkynyl like ethyne, propyne, butyene, pentyne, or hexyne; or is C2-4-alkynyl like ethyne, propyne or butyene. In connection with alkyl (also in aryl-alkyl, alkylheterocyclyl or alkylcycloalkyl), alkenyl, alkynyl and O-alkyl - unless defined otherwise - the term substituted in the context of this invention is understood as meaning replacement of at least one hydrogen radical on a carbon atom by halogen, -OR’, -SR’, -SOR’, -SO2R’, -OR’, -CN, -COR’, -COOR’, -NR’R”, -CONR’R”, haloalkyl, haloalkoxy or -OC1-6 alkyl wherein each of the R’ and R” groups is independently selected from the group consisting of hydrogen, OH, NO2, NH2, SH, CN, halogen, -COH, -COalkyl, -COOH and C1-6 alkyl.
In a particular embodiment of the invention, the alkyl, alkenyl or alkynyl as defined in R1- R10” if substituted, is substituted with one or more substituent/s selected from -OR’, halogen, -CN, haloalkyl, haloalkoxy and -NR’R”; wherein R, R’ and R” are independently selected from hydrogen, unsubstituted C1-6 alkyl, unsubstituted C2-6 alkenyl, and unsubstituted C2-6 alkynyl;
More than one replacement on the same molecule and also on the same carbon atom is possible with the same or different substituents. This includes for example 3 hydrogens being replaced on the same C atom, as in the case of CF3, or at different places of the same molecule, as in the case of e.g. -CH(OH)-CH=CH-CHCl2.
In the context of this invention haloalkyl is understood as meaning an alkyl being substituted once or several times by a halogen (selected from F, Cl, Br, I). It encompasses e.g. -CH2CI, -CH2F, -CHC , -CHF2, -CCI3, -CF3 and -CH2-CHCI2. Preferably haloalkyl is understood in the context of this invention as halogen-substituted Ci-4-alkyl representing halogen substituted C1 -, C2-, C3- or C4-alkyl. The halogen- substituted alkyl radicals are thus preferably methyl, ethyl, propyl, and butyl. Preferred examples include -CH2CI, -CH2F, -CH2-CH2F, -CH2-CHF2, -CHCb, -CHF2, and -CF3.
In the context of this invention haloalkoxy is understood as meaning an -O-alkyl being substituted once or several times by a halogen (selected from F, Cl, Br, I). It encompasses e.g. -OCH2CI, -OCH2F, -OCHCb, -OCHF2, -OCCI3, -OCF3 and -OCH2- CHCb. Preferably haloalkoxy is understood in the context of this invention as halogen- substituted -OCi-4-alkyl representing halogen substituted C1 -, C2-, C3- or C4-alkoxy. The halogen-substituted O-alkyl radicals are thus preferably O-methyl, O-ethyl, O-propyl, and O-butyl. Preferred examples include -OCH2CI, -OCH2F, -OCHCb, -OCHF2, and - OCF3.
In the context of this invention cycloalkyl is understood as meaning saturated and unsaturated (but not aromatic) cyclic hydrocarbons (without a heteroatom in the ring), which can be unsubstituted or once or several times substituted. Preferred cycloalkyls are C3-4-cycloalkyl representing C3- or C4-cycloalkyl, C3-5-cycloalkyl representing C3-, C4- or C5-cycloalkyl, C3-6-cycloalkyl representing C3-, C4-, C5- or C6-cycloalkyl, C3-7- cycloalkyl representing C3-, C4-, C5-, C6- or C7-cycloalkyl, C3-8-cycloalkyl representing C3-, C4-, C5-, C6-, C7- or C8-cycloalkyl, C4-5-cycloalkyl representing C4- or C5- cycloalkyl, C4-6-cycloalkyl representing C4-, C5- or C6-cycloalkyl, C4-7-cycloalkyl representing C4-, C5-, C6- or C7-cycloalkyl, C5-6-cycloalkyl representing C5- or C6- cycloalkyl and C5-7-cycloalkyl representing C5-, C6- or C7-cycloalkyl. Examples are cyclopropyl, 2-methylcyclopropyl, cyclopropylmethyl, cyclobutyl, cyclopentyl, cyclopentylmethyl, cyclohexyl, cycloheptyl, cyclooctyl, and also adamantyl. Preferably in the context of this invention cycloalkyl is C3-8-cycloalkyl like cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, or cyclooctyl; or is C3-7-cycloalkyl like cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl; or is C3-6-cycloalkyl like cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl, especially cyclopentyl or cyclohexyl.
Aryl is understood as meaning 6 to 18 membered mono or polycyclic ring systems with at least one aromatic ring but without heteroatoms even in only one of the rings. Examples are phenyl, naphthyl, fluoranthenyl, fluorenyl, tetralinyl or indanyl, 9H-fluorenyl or anthracenyl radicals, which can be unsubstituted or once or several times substituted. Most preferably aryl is understood in the context of this invention as phenyl, naphthyl or anthracenyl, more preferably the aryl is phenyl.
A heterocyclyl radical or group (also called heterocyclyl hereinafter) is understood as meaning 4 to 18, preferably 5 to 18, membered mono or polycyclic heterocyclic ring systems, with at least one saturated or unsaturated ring which contains one or more heteroatoms selected from the group consisting of nitrogen, oxygen and/or sulfur in the ring. A heterocyclic group can also be substituted once or several times.
Examples include non-aromatic heterocyclyls such as tetrahydropyran, oxazepane, morpholine, piperidine, pyrrolidine as well as heteroaryls such as furan, benzofuran, thiophene, benzothiophene, pyrrole, pyridine, pyrimidine, pyrazine, quinoline, isoquinoline, phthalazine, thiazole, benzothiazole, indole, benzotriazole, carbazole and quinazoline.
Subgroups inside the heterocyclyls as understood herein include heteroaryls and non- aromatic heterocyclyls. the heteroaryl (being equivalent to heteroaromatic radicals or aromatic heterocyclyls) is an aromatic 5 to 18 membered mono or polycyclic heterocyclic ring system of one or more rings of which at least one aromatic ring contains one or more heteroatoms from the group consisting of nitrogen, oxygen and/or sulfur in the ring; preferably is a 5 to 18 membered mono or polycyclic aromatic heterocyclic ring system of one or two rings of which at least one aromatic ring contains one or more heteroatoms selected from the group consisting of nitrogen, oxygen and/or sulfur in the ring, more preferably it is selected from furan, benzofuran, thiophene, benzothiophene, pyrrole, pyridine, pyrimidine, pyrazine, quinoline, isoquinoline, phthalazine, benzothiazole, indole, benzotriazole, carbazole, quinazoline, thiazole, imidazole, pyrazole, oxazole, thiophene and benzimidazole;
the non-aromatic heterocyclyl is a 4 to 18, preferably 5 to 18, membered mono or polycyclic heterocyclic ring system of one or more rings of which at least one ring - with this (or these) ring(s) then not being aromatic - contains one or more heteroatoms from the group consisting of nitrogen, oxygen and/or sulfur in the ring; preferably it is a 5 to 18 membered mono or polycyclic heterocyclic ring system of one or two rings of which one or both rings - with this one or two rings then not being aromatic - contain/s one or more heteroatoms selected from the group consisting of nitrogen, oxygen and/or sulfur in the ring, more preferably it is selected from oxazepam, pyrrolidine, piperidine, piperazine, tetrahydropyran, morpholine, indoline, oxopyrrolidine, benzodioxane, especially is piperazine, benzodioxane, morpholine, tetrahydropyran, piperidine, oxopyrrolidine and pyrrolidine.
Preferably, in the context of this invention heterocyclyl is defined as a 4 to 18 membered mono or polycyclic heterocyclic ring system of one or more saturated or unsaturated rings of which at least one ring contains one or more heteroatoms selected from the group consisting of nitrogen, oxygen and/or sulfur in the ring. Preferably it is a 5 to 18 membered mono or polycyclic heterocyclic ring system of one or two saturated or unsaturated rings of which at least one ring contains one or more heteroatoms selected from the group consisting of nitrogen, oxygen and/or sulfur in the ring. More preferably, it is a 5 to 10 membered mono or bicyclic heterocyclyl ring system containing one nitrogen atom and optionally a second heteroatom selected from nitrogen and oxygen. In another preferred embodiment of the invention, said heterocyclyl is a substituted mono or bicyclic heterocyclyl ring system. Preferred examples of heterocyclyls include oxazepam, pyrrolidine, imidazole, oxadiazole, tetrazole, pyridine, pyrimidine, piperidine, piperazine, benzofuran, benzimidazole, indazole, benzodiazole, thiazole, benzothiazole, tetrahydropyran, morpholine, indoline, furan, triazole, isoxazole, pyrazole, thiophene, benzothiophene, pyrrole, pyrazine, pyrrolo[2,3b]pyridine, quinoline, isoquinoline, tetrahydroisoquinoline, phthalazine, benzo-1 ,2,5-thiadiazole, indole, benzotriazole, benzoxazole oxopyrrolidine, pyrimidine, benzodioxolane, benzodioxane, carbazole and quinazoline, especially is pyridine, piperazine, pyrazine, indazole, benzodioxane, thiazole, benzothiazole, morpholine, tetrahydropyran, pyrazole, imidazole, piperidine, thiophene, indole, benzimidazole, pyrrolo[2,3b]pyridine, benzoxazole, oxopyrrolidine, pyrimidine, oxazepane and pyrrolidine. In the context of this invention oxopyrrolidine is understood as meaning pyrrolidin-2-one.
In connection with aromatic heterocyclyls (heteroaryls), non-aromatic heterocyclyls, aryls and cycloalkyls, when a ring system falls within two or more of the above cycle definitions simultaneously, then the ring system is defined first as an aromatic heterocyclyl (heteroaryl) if at least one aromatic ring contains a heteroatom. If no aromatic ring contains a heteroatom, then the ring system is defined as a non-aromatic heterocyclyl if at least one non-aromatic ring contains a heteroatom. If no non-aromatic ring contains a heteroatom, then the ring system is defined as an aryl if it contains at least one aryl cycle. If no aryl is present, then the ring system is defined as a cycloalkyl if at least one non- aromatic cyclic hydrocarbon is present.
In the context of this invention aryl-alkyl is understood as meaning an aryl group (see above) being connected to another atom through a Ci-6-alkyl (see above) which may be branched or linear and is unsubstituted or substituted once or several times. Preferably aryl-alkyl is understood as meaning an aryl group (see above) being connected to another atom through 1 to 4 (-CH2-) groups.
In the context of this invention alkylheterocyclyl is understood as meaning an heterocyclyl group (see above) being connected to another atom through a Ci-6-alkyl (see above) which may be branched or linear and is unsubstituted or substituted once or several times. Preferably alkylheterocyclyl is understood as meaning an heterocyclyl group (see above) being connected to another atom through 1 to 4 (-CH2-) groups.
In the context of this invention alkylcycloalkyl is understood as meaning an cycloalkyl group (see above) being connected to another atom through a Ci-6-alkyl (see above) which may be branched or linear and is unsubstituted or substituted once or several times. Preferably alkylcycloalkyl is understood as meaning a cycloalkyl group (see above) being connected to another atom through 1 to 4 (-CH2-) groups.
Preferably, the aryl is a monocyclic aryl. More preferably the aryl is a 6 or 7 membered monocyclic aryl. Even more preferably the aryl is a 6 membered monocyclic aryl, preferably phenyl.
Preferably, the heteroaryl is a monocyclic heteroaryl. More preferably the heteroaryl is a 5, 6 or 7 membered monocyclic heteroaryl. Even more preferably the heteroaryl is a 5 or 6 membered monocyclic heteroaryl.
Preferably, the non-aromatic heterocyclyl is a monocyclic non-aromatic heterocyclyl. More preferably the non-aromatic heterocyclyl is a 4, 5, 6 or 7 membered monocyclic non-aromatic heterocyclyl. Even more preferably the non-aromatic heterocyclyl is a 5 or 6 membered monocyclic non-aromatic heterocyclyl. In another preferred embodiment, said non-aromatic heterocyclyl is a bicyclic non-aromatic heterocyclyl.
Preferably, the cycloalkyl is a monocyclic cycloalkyl. More preferably the cycloalkyl is a 3, 4, 5, 6, 7 or 8 membered monocyclic cycloalkyl. Even more preferably the cycloalkyl is a 3, 4, 5 or 6 membered monocyclic cycloalkyl.
A ring system is a system consisting of at least one ring of connected atoms but including also systems in which two or more rings of connected atoms are joined with“joined” meaning that the respective rings are sharing one (like a spiro structure), two or more atoms being a member or members of both joined rings.
The term “leaving group” means a molecular fragment that departs with a pair of electrons in heterolytic bond cleavage. Leaving groups can be anions or neutral molecules. Common anionic leaving groups are halides such as CI-, Br-, and I-, and sulfonate esters, such as tosylate (TsO-), mesylate, nosylate or triflate.
The term“salt” is to be understood as meaning any form of the active compound used according to the invention in which it assumes an ionic form or is charged and is coupled with a counter-ion (a cation or anion) or is in solution. By this are also to be understood complexes of the active compound with other molecules and ions, in particular complexes via ionic interactions. The definition particularly includes physiologically acceptable salts, this term must be understood as equivalent to “pharmacologically acceptable salts”.
The term“physiologically acceptable salt” means in the context of this invention any salt that is physiologically tolerated (most of the time meaning not being toxic- especially lacking toxicity caused by the counter-ion) if used appropriately for a treatment especially if used on or applied to humans and/or mammals.
These physiologically acceptable salts can be formed with cations or bases and in the context of this invention is understood as meaning salts of at least one of the compounds used according to the invention - usually a (deprotonated) acid - as an anion with at least one, preferably inorganic, cation which is physiologically tolerated - especially if used on humans and/or mammals. The salts of the alkali metals and alkaline earth metals are particularly preferred, and also those with NH4, but in particular (mono)- or (di)sodium, (mono)- or (di)potassium, magnesium or calcium salts.
Physiologically acceptable salts can also be formed with anions or acids and in the context of this invention is understood as meaning salts of at least one of the compounds used according to the invention as the cation with at least one anion which are physiologically tolerated - especially if used on humans and/or mammals. By this it is understood in particular, in the context of this invention, the salt formed with a physiologically tolerated acid, that is to say salts of the particular active compound with inorganic or organic acids which are physiologically tolerated - especially if used on humans and/or mammals. Examples of physiologically tolerated salts of particular acids are salts of: hydrochloric acid, hydrobromic acid, sulfuric acid, methanesulfonic acid, formic acid, acetic acid, oxalic acid, succinic acid, malic acid, tartaric acid, mandelic acid, fumaric acid, lactic acid or citric acid.
The compounds of the invention may be present in crystalline form or in the form of free compounds like a free base or acid.
Any compound that is a solvate of a compound according to the invention like a compound according to formula (I) defined above is understood to be also covered by the scope of the invention. Methods of solvation are generally known within the art. Suitable solvates are pharmaceutically acceptable solvates. The term “solvate” according to this invention is to be understood as meaning any form of the active compound according to the invention in which this compound has attached to it via non- covalent binding another molecule (most likely a polar solvent). Especially preferred examples include hydrates and alcoholates, like methanolates or ethanolates.
Any compound that is a prodrug of a compound according to the invention like a compound according to formula (I) defined above is understood to be also covered by the scope of the invention. The term “prodrug” is used in its broadest sense and encompasses those derivatives that are converted in vivo to the compounds of the invention. Such derivatives would readily occur to those skilled in the art, and include, depending on the functional groups present in the molecule and without limitation, the following derivatives of the present compounds: esters, amino acid esters, phosphate esters, metal salts sulfonate esters, carbamates, and amides. Examples of well known methods of producing a prodrug of a given acting compound are known to those skilled in the art and can be found e.g. in Krogsgaard-Larsen et al.“Textbook of Drug design and Discovery” Taylor & Francis (April 2002).
Any compound that is an N-oxide of a compound according to the invention like a compound according to formula (I) defined above is understood to be also covered by the scope of the invention.
Unless otherwise stated, the compounds of the invention are also meant to include compounds which differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures except for the replacement of a hydrogen by a deuterium or tritium, or the replacement of a carbon by 13C- or 14C- enriched carbon or of a nitrogen by 15N-enriched nitrogen are within the scope of this invention.
The compounds of formula (I) as well as their salts or solvates are preferably in pharmaceutically acceptable or substantially pure form. By pharmaceutically acceptable pure form is meant, inter alia, having a pharmaceutically acceptable level of purity excluding normal pharmaceutical additives such as diluents and carriers, and including no material considered toxic at normal dosage levels. Purity levels for the drug substance are preferably above 50%, more preferably above 70%, most preferably above 90%. In a preferred embodiment it is above 95% of the compound of formula (I), or of its salts. This applies also to its solvates or prodrugs.
All the groups above mentioned that can be substituted or unsubstituted may be substituted at one or more available positions by one or more suitable groups such as OR’, =0, SR’, SOR’, S02R’, OS02R’, OSOSR’, N02, NHR’, N(R’)2, =N-R’, N(R’)COR’, N(COR’)2, N(R’)S02R’, N(R’)C(=NR’)N(R’)R’, N3, CN, halogen, COR’, COOR’, OCOR’, OCOOR’, OCONHR’, OCON(R’)2, CONHR’, CON(R’)2, CON(R’)OR\ C0N(R’)S02R\ PO(OR’)2, PO(OR’)R’, PO(OR’)(N(R’)R’), CI-I2 alkyl, C3-10 cycloalkyl, C2-i2 alkenyl, C2-i2 alkynyl, aryl, and heterocyclic group, wherein each of the R’ groups is independently selected from the group consisting of hydrogen, OH, N02, NH2, SH, CN, halogen, COH, COalkyl, COOH , CM2 alkyl, C3-10 cycloalkyl, C2-i2 alkenyl, C2-i2 alkynyl, aryl and heterocyclic group. Where such groups are themselves substituted, the substituents may be chosen from the foregoing list.
In a particular embodiment of the invention, the compound of formula (I) is a compound wherein
W3 is S and W4 is C;
Y is -C(O)-, -[C(RaRa')]n-, -CRa(CN)- or -CRa(CONRa'Ra ")-. wherein each Ra, Ra· and Ra” are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl, and substituted or unsubstituted C2-6 alkynyl; and n is 0, 1 , 2 or 3;
X is selected from a bond, -[C(RbRb )] -, -[C(RbRb )] C(0)[C(Rb"Rb )]q-> substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl-C(O), substituted or unsubstituted aryl- alkyl, wherein each Rb, Rb·, Rb- and Rb- are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl, and substituted or unsubstituted C2-6 alkynyl; and p is 0, 1 , 2 or 3; q is 0, 1 , 2 or 3;
Group A is a substituted or unsubstituted 5 to 10 membered mono or bicyclic heterocyclyl containing one nitrogen atom and optionally a second heteroatom selected from N and O, wherein A is attached to Y through a nitrogen atom; each R1 and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl, substituted or unsubstituted C2-6 alkynyl, substituted or unsubstituted alkyl-cycloalkyl; NR4R4', C(0)0R4 and substituted or unsubstituted haloalkyl; wherein each R4 and R4' are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl, substituted or unsubstituted C2-6 alkynyl, and C(0)0R4·; wherein R4 " is substituted or unsubstituted C1-6 alkyl.
R5 is selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6alkenyl, substituted or unsubstituted C2-6 alkynyl, CN, -C(0)0R3 and -S(0)20R3' ; wherein each R3 and R3· are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl, and substituted or unsubstituted C2-6 alkynyl;
R6 is selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl (tert-butyl), substituted or unsubstituted C2-6 alkenyl, substituted or unsubstituted C2-6 alkynyl,
R2 is a group of the following formula
Figure imgf000023_0001
wherein m is 0, 1 or 2; r is 0, 1 or 2; t is 0, 1 , 2 or 3;
Wi is selected from C or N; each Rg, Rg·, Rg- and Rg- are independently selected from hydrogen, substituted or unsubstituted Ci-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2-6 alkynyl; alternatively, Rg and Rg· and/or Rg- and Rg- taken together with the carbon atom to which they are attached may form a substituted or unsubstituted cycloalkyl; alternatively, Rg and Rg· and/or Rg- and Rg- taken together with the carbon atom to which they are attached may form a carbonyl group; each R10, Rio·, R10” and Rio- are independently selected from hydrogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2-6 alkynyl; alternatively, R10, R-io· and/or Rio ·, R-io- taken together with the carbon atom to which they are attached may form a substituted or unsubstituted cycloalkyl; alternatively, R10 and R-io· and/or Rio- and R-io- taken together with the carbon atom to which they are attached may form a carbonyl group;
R7 is selected from substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted aryl and substituted or unsubstituted aromatic heterocyclyl;
Rs is selected from hydrogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2- e alkynyl;
Rs· is selected from hydrogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2- e alkynyl; alternatively, R7 and Rs taken together with -N(Rs )-[CH2]t- atoms to which they are attached form a substituted or unsubstituted heterocyclyl optionally fused with a substituted or unsubstituted aryl group; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In a particular embodiment of the invention, the compound according to the invention of formula (I) is a compound wherein
Y is -C(O)-, -[C(RaRa )]n-, -CRa(CN)- or -CRa(CONRa Ra )-, wherein each Ra, Ra· and Ra- are independently selected from hydrogen, halogen, substituted or unsubstituted Ci-6 alkyl, substituted or unsubstituted C2-6 alkenyl, and substituted or unsubstituted C2-6 alkynyl; and n is 0, 1 , 2 or 3; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In a preferred embodiment, the compound according to the invention of formula (I) is a compound wherein
Y is -C(O)-, -[C(RaRa )]n-, -CRa(CN)- or -CRa(CONRa Ra )-, wherein each Ra, Ra· and Ra- are hydrogen, n is 0, 1 or 2; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another particular embodiment, the compound according to the invention of formula (I) is a compound wherein
Y is -C(O)- optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In a particular embodiment of the invention, the compound according to the invention of formula (I) is a compound wherein
X is selected from a bond, -[C(RbRb )]P-, -[C(RbRb )]PC(0)[C(Rb"Rb )]q-, unsubstituted aryl, preferably phenyl, unsubstituted aryl-C(O), preferably phenyl-C(O) and unsubstituted aryl-alkyl, preferably phenyl-alkyl, and wherein each Rb, Rb·, Rb- and Rb- are hydrogen, and p is 0, 1 or 2; q is 0, 1 or 2; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In a more particular embodiment of the invention, the compound according to the invention of formula (I) is a compound wherein
-X-R2 is selected from -[C(RbRb)]P-R2, -[C(RbRb)]PC(0)[C(Rb"Rb )]q-R2, -aryl-R2, preferably -phenyl-R2 and -aryl-C(0)-R2, preferably -phenyl-C(0)-R2, -aryl-alkyl-R2, preferably -phenyl-alkyl-R2, wherein each Rb, Rb·, Rb- and Rb- are hydrogen; and p is 0, 1 or 2; q is 0, 1 or 2; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another particular embodiment of the invention, the compound according to the invention of formula (I) is a compound wherein group A is selected from:
Figure imgf000027_0001
wherein b is 0, 1 or 2; and
W2 is selected from O, C and N; each R1 and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl, substituted or unsubstituted C2-6 alkynyl, substituted or unsubstituted alkyl-cycloalkyl; NR4R4', - C(0)0R4- and substituted or unsubstituted haloalkyl; wherein each R4 and R4' are independently selected from hydrogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl, substituted or unsubstituted C2-6 alkynyl, and -C(0)OR4·-; wherein R4 " is substituted or unsubstituted C1-6 alkyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In a preferred embodiment, the compound according to the invention of formula (I) is a compound wherein when W2 is N and R1 or Rr is NR4R4'; then NR4R4' is not atacthed to W2. In a preferred embodiment, the compound according to the invention of formula (I) is a compound wherein each R1 and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted alkyl-cycloalkyl; NR4R4', and substituted or unsubstituted haloalkyl; wherein each R4 and R4· are independently selected from hydrogen and substituted or unsubstituted C1-6 alkyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In a preferred embodiment, the alkyl, alkenyl or alkynyl, also in alkyl-cycloalkyl and haloalkyl defined in R1 or Rr, if substituted, is substituted with one or more substituent/s selected from -O R51 , halogen, and -NRsiRsr; wherein each of R51 or R5r is independently selected from hydrogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2-6 alkynyl. In a more preferred embodiment, each of R51 orR5r is selected from hydrogen and unsubstituted C1-6 alkyl, more preferably methyl.
In another particular embodiment, the compound according to the invention of formula (I) is a compound wherein group A is
Figure imgf000028_0001
wherein b is 1 or 2;
W2 is N; and each R1 and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, unsubstituted alkyl-cycloalkyl; preferably alkyl-cyclopropyl, and substituted or unsubstituted haloalkyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof. In a preferred embodiment, the compound according to the invention of formula (I) is a compound wherein group A is
Figure imgf000029_0001
wherein b is 1 ;
W2 is N; and each R1 and Rr are independently selected from hydrogen, halogen, preferably fluor, substituted or unsubstituted C1-6 alkyl, preferably methyl, substituted or unsubstituted alkyl-cycloalkyl; preferably alkyl-cyclopropyl, and substituted or unsubstituted haloalkyl; preferably -CH2F, -CH2-CH2F, -CH2-CHF2, or -CHF2. optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof. In a more preferred embodiment, the compound according to the invention of formula (I) is a compound wherein group A is
Figure imgf000029_0002
wherein b is 1 ; W2 is N;
R1 is hydrogen; and Rr is substituted or unsubstituted Ci-6 alkyl, and wherein Rr is directly attached to W2.
In another particular embodiment, the compound according to the invention of formula (I) is a compound wherein group A is
Figure imgf000030_0001
each Ri and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C alkyl; and substituted or unsubstituted haloalkyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In a preferred embodiment, the compound according to the invention of formula (I) is a compound wherein group A is
Figure imgf000030_0002
each Ri and Rr are independently selected from hydrogen and unsubstituted C1-6 alkyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another particular embodiment, the compound according to the invention of formula (I) is a compound wherein group A is
Figure imgf000031_0001
each Ri and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C alkyl; and substituted or unsubstituted haloalkyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In a preferred embodiment, the compound according to the invention of formula (I) is a compound wherein group A is
Figure imgf000031_0002
each Ri and Rr are independently selected from hydrogen and unsubstituted C1-6 alkyl. optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In a particular embodiment of the invention, the compound according to the invention of formula (I) is a compound wherein R2 is a group of the following formula
Figure imgf000032_0001
wherein m is 1 ; r is 1 ; t is 0 or 1 ; Wi is C or N; each Rg, Rg·, Rg- and Rg- are hydrogen; each Rio, Rio·, Rio- and Rio- are hydrogen;
R7 is selected from substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted aryl and substituted or unsubstituted aromatic heterocyclyl; Re is selected from hydrogen and substituted or unsubstituted C1-6 alkyl;
Re· is selected from hydrogen and substituted or unsubstituted C1-6 alkyl; or
R7 and Re taken together with -N(R8')-[CH2]t- atoms to which they are attached form a substituted or unsubstituted heterocyclyl optionally fused with a substituted or unsubstituted aryl group; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof. In another particular embodiment, the compound according to the invention of formula (I) is a compound wherein R2 is a group of the following formula:
Figure imgf000033_0001
wherein m is 1 ; r is 1 ; t is 0;
Wi is C; each Rg, Rg·, Rg- and Rg- are hydrogen; each R10, Rio·, Rio- and Rio- are hydrogen;
R7 is substituted or unsubstituted aryl; preferably phenyl;
Rs is substituted or unsubstituted C1-6 alkyl; preferably methyl, and Rs· is substituted or unsubstituted C1-6 alkyl; preferably methyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof. In another particular embodiment, the compound according to the invention of formula (I) is a compound wherein R2 is a group of the following formula
Figure imgf000033_0002
(R2) wherein m is 1 ; r is 1 ; t is 0;
Wi is N; each Rg, Rg·, Rg- and Rg- are hydrogen; each Rio, Rio·, Rio” and Rio- are hydrogen;
R7 is substituted or unsubstituted aryl; preferably phenyl;
Rs is substituted or unsubstituted C1-6 alkyl; preferably methyl, and
Rs· is substituted or unsubstituted C1-6 alkyl; preferably methyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In a particular embodiment, the compound according to the invention of formula (I) is a compound wherein R7 is selected from substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted aryl, preferably phenyl, and substituted or unsubstituted aromatic heterocyclyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another particular embodiment, the alkyl, aryl and aromatic heterocyclyl as defined in R7, if substituted, is substituted with one or more substituent/s selected from -OR71 and halogen, wherein
R71 is selected from hydrogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2-6 alkynyl. In a more preferred embodiment, R71 is selected from hydrogen and unsubstituted C1-6 alkyl, preferably C1-3 alkyl, more preferably methyl. In a preferred embodiment, the compound according to the invention of formula (I) is a compound wherein R7 is C1-6 alkyl; more preferably C1-3 alkyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In a preferred embodiment, the compound according to the invention of formula (I) is a compound wherein R7 is substituted or unsubstituted aryl; more preferably, R7 is phenyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In a particular embodiment, the aryl as defined in R7, if substituted, is substituted with one or more substituent/s selected from -OR7I and halogen, preferably fluor, wherein
R7I is selected from hydrogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2-6 alkynyl. In a more preferred embodiment, R7I is selected from hydrogen and unsubstituted C1-6 alkyl, preferably C1-3 alkyl, more preferably methyl.
In a preferred embodiment, the compound according to the invention of formula (I) is a compound wherein Re is selected from hydrogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2-6 alkynyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In a particular embodiment, the C1-6 alkyl as defined in Re, if substituted, is substituted with one or more substituent/s selected from substituted or unsubstituted C1-6 alkyl, preferably unsubstituted C1-3 alkyl, and halogen, preferably fluor. In a preferred embodiment, the compound according to the invention of formula (I) is a compound wherein Re· is selected from hydrogen, substituted or unsubstituted Ci-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2-6 alkynyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In a particular embodiment, the C1-6 alkyl as defined in Re·, if substituted, is substituted with one or more substituent/s selected from substituted or unsubstituted C1-6 alkyl, preferably unsubstituted C1-3 alkyl, and halogen, preferably fluor.
In a preferred embodiment, the compound according to the invention of formula (I) is a compound wherein R7 and Rs taken together with -N(Rs')-[CH2]t- atoms to which they are attached may form a substituted or unsubstituted heterocyclyl optionally fused with a substituted or unsubstituted aryl group; more preferably said heterocyclyl is an unsubstituted heterocyclyl fused with an unsubstituted aryl group; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In a more preferred embodiment, the compound according to the invention of formula (I) is a compound wherein R7 and Rs taken together with -N(Rs')-[CH2]t- atoms to which are attached is
Figure imgf000036_0001
wherein Wi and Rs· are as defined above, preferably Wi is N and Rs· is Ci-6 alkyl, preferably methyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In a particular embodiment, the compound according to the invention of formula (I) is a compound wherein when Wi is N then X is not a bond. In another preferred embodiment, the compound of the invention according to formula (I) is a compound, wherein: one of W3 and W4 is S and the other is C; preferably, W3 is S and W4 is C;
and/or Y is -C(O)-, -[C(RaRa')]n-, -CRa(CN)- or -CRa(CONRa'Ra' ')-, wherein each Ra, Ra· and Ra- are hydrogen and n is 0, 1 or 2; and/or
X is selected from a bond, -[C(RbRb )]P-, -[C(RbRb )]PC(0)[C(Rb”Rb )]q-> unsubstituted aryl, preferably phenyl, unsubstituted aryl-C(O), preferably phenyl-C(O) and unsubstituted aryl-alkyl, preferably phenyl-alkyl, and wherein each Rb, Rb·, Rb- and Rb- are hydrogen, p is 0, 1 or 2; q is 0, 1 or 2; and/or group A is selected from:
Figure imgf000037_0001
wherein b is 0, 1 or 2; and
W2 is selected from O, C and N; each R1 and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted alkyl-cycloalkyl; NR4R4', -C(0)OR4- and substituted or unsubstituted haloalkyl; wherein each R4 and R4' are independently selected from hydrogen, substituted or unsubstituted C1-6 alkyl and -C(0)OR4·-; wherein R4 " is substituted or unsubstituted C1-6 alkyl; and/or
R5 is selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, CN, - C(0)0R3 and -S(0)20R3· ; wherein each R3 and R3' are hydrogen; and/or
R6 is selected from hydrogen, halogen and substituted or unsubstituted C1-6 alkyl; and/or
R2 IS:
Figure imgf000038_0001
wherein m is 1 ; r is 1 ; t is 0 or 1 ;
Wi is C or N; each Rg, Rg·, Rg- and Rg- are hydrogen; each R10, Rio·, R10” and Rio- are hydrogen; R7 is selected from substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted aryl, preferably phenyl, and substituted or unsubstituted aromatic heterocyclyl;
Re is selected from hydrogen and substituted or unsubstituted C1-6 alkyl;
Re’ is selected from hydrogen and substituted or unsubstituted C1-6 alkyl; or
R7 and Re taken together with -N(R8')-[CH2]t- atoms to which they are attached may form a substituted or unsubstituted heterocyclyl optionally fused with a substituted or unsubstituted aryl group; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another preferred embodiment, the compound of the invention according to formula (I) is a compound, wherein:
W3 is S and W4 is C;
and/or
Y is -C(O)-, -[C(RaRa')]n-, -CRa(CN)- or -CRa(CONRa'Ra ")-. wherein each Ra, Ra· and Ra- are hydrogen and n is 0, 1 or 2; and/or
X is selected from a bond, -[C(RbRb )]P-, -[C(RbRb )]PC(0)[C(Rb”Rb )]q-> unsubstituted aryl, preferably phenyl, unsubstituted aryl-C(O), preferably phenyl-C(O) and unsubstituted aryl-alkyl, preferably phenyl-alkyl, and wherein each Rb, Rb·, Rb- and Rb- are hydrogen, p is 0, 1 or 2; q is 0, 1 or 2; and/or group A is selected from:
Figure imgf000040_0001
wherein b is 0, 1 or 2; and
W2 is selected from O, C and N; each Ri and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted alkyl-cycloalkyl; NR4R4', -C(0)0R4- and substituted or unsubstituted haloalkyl; wherein each R4 and R4' are independently selected from hydrogen, substituted or unsubstituted C1-6 alkyl and -C(0)0R4·-; wherein R4" is substituted or unsubstituted C1-6 alkyl; and/or
R5 is selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, CN, - C(0)0R3 and -S(0)20R3· ; wherein each R3 and R3' are hydrogen; and/or
R6 is selected from hydrogen, halogen and substituted or unsubstituted C1-6 alkyl; and/or
R2 IS:
Figure imgf000041_0001
wherein m is 1 ; r is 1 ; t is 0 or 1 ; Wi is C or N; each Rg, Rg·, Rg- and Rg- are hydrogen; each Rio, Rio·, Rio- and Rio- are hydrogen;
R7 is selected from substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted aryl, preferably phenyl, and substituted or unsubstituted aromatic heterocyclyl;
Re is selected from hydrogen and substituted or unsubstituted C1-6 alkyl;
Re’ is selected from hydrogen and substituted or unsubstituted C1-6 alkyl; or
R7 and Re taken together with -N(R8')-[CH2]t- atoms to which they are attached may form a substituted or unsubstituted heterocyclyl optionally fused with a substituted or unsubstituted aryl group; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof. In another preferred embodiment of the invention according to formula (I) the compound is a compound, wherein in Ra, Ra and Ra- as defined in any of the embodiments of the present invention, the C1-6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2-methylpropyl; and/or
the C2-6 -alkenyl is preferably selected from ethylene, propylene, butylene, pentylene, hexylene, isopropylene and isobutylene; and/or
the C2-6 -alkynyl is preferably selected from ethyne, propyne, butyne, pentyne, hexyne, isopropyne and isobutyne; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another preferred embodiment of the invention according to formula (I) the compound is a compound, wherein in Rb, Rb·, Rb- and Rb- as defined in any of the embodiments of the present invention, the C1-6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl, and 2-methylpropyl; and/or
the C2-6 -alkenyl is preferably selected from ethylene, propylene, butylene, pentylene, hexylene, isopropylene and isobutylene; and/or
the C2-6 -alkynyl is preferably selected from ethyne, propyne, butyne, pentyne, hexyne, isopropyne and isobutyne; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another preferred embodiment of the invention according to formula (I) the compound is a compound, wherein in Ri and Rr as defined in any of the embodiments of the present invention, the C1-6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl, and 2-methylpropyl; and/or
the C2-6 -alkenyl is preferably selected from ethylene, propylene, butylene, pentylene, hexylene, isopropylene and isobutylene; and/or
the C2-6 -alkynyl is preferably selected from ethyne, propyne, butyne, pentyne, hexyne, isopropyne and isobutyne; and/or
the cycloalkyl is C3-8 cycloalkyl like cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, or cyclooctyl; preferably is C3-7 cycloalkyl like cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl; more preferably from C3-6 cycloalkyl like cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl; more preferably the cycloalkyl is cyclopropyl; and/or
the haloalkyl is selected from -CH2CI, -CH2F, -CHC , -CHF2, -CCI3, -CF3 and -CH2-CHCI2; more preferably the haloalkyl is -CHF2 or -CF3;
optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another preferred embodiment of the invention according to formula (I) the compound is a compound, wherein in R4and R4 as defined in any of the embodiments of the present invention, the C1-6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl, and 2-methylpropyl; and/or
the C2-6 -alkenyl is preferably selected from ethylene, propylene, butylene, pentylene, hexylene, isopropylene and isobutylene; and/or
the C2-6 -alkynyl is preferably selected from ethyne, propyne, butyne, pentyne, hexyne, isopropyne and isobutyne; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another preferred embodiment of the invention according to formula (I) the compound is a compound, wherein in R5 as defined in any of the embodiments of the present invention, the alkyl is preferably selected from C1-6 alkyl; more preferably, the Ci-6 alkyl is selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2- methylpropyl; and/or
the C2-6 -alkenyl is preferably selected from ethylene, propylene, butylene, pentylene, hexylene, isopropylene and isobutylene; and/or
the C2-6 -alkynyl is preferably selected from ethyne, propyne, butyne, pentyne, hexyne, isopropyne and isobutyne;
and/or
-C(0)0R3 or -S(0)20R3' are preferably -C(0)0H and -S(0)20H; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another preferred embodiment of the invention according to formula (I) the compound is a compound, wherein in R6 as defined in any of the embodiments of the present invention, the alkyl is preferably selected from C1-6 alkyl; more preferably, the Ci-6 alkyl is selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2- methylpropyl; and/or
the C2-6 -alkenyl is preferably selected from ethylene, propylene, butylene, pentylene, hexylene, isopropylene and isobutylene; and/or
the C2-6 -alkynyl is preferably selected from ethyne, propyne, butyne, pentyne, hexyne, isopropyne and isobutyne;
optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another preferred embodiment of the invention according to formula (I) the compound is a compound, wherein in R7 as defined in any of the embodiments of the present invention, the alkyl is preferably selected from C1-6 alkyl; more preferably, the C1-6 alkyl is selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2- methylpropyl; and/or
the aryl is selected from phenyl, naphtyl, or anthracenyl; preferably is napthyl and phenyl; and/or
the heterocyclyl is a heterocyclic ring system of one or more saturated or unsaturated rings of which at least one ring contains one or more heteroatoms from the group consisting of nitrogen, oxygen and/or sulfur in the ring; preferably is a heterocyclic ring system of one or two saturated or unsaturated rings of which at least one ring contains one or more heteroatoms from the group consisting of nitrogen, oxygen and/or sulfur in the ring, more preferably is selected from oxazepan, pyrrolidine, imidazole, oxadiazole, tetrazole, pyridine, pyrimidine, piperidine, piperazine, benzofuran, benzimidazole, indazole, benzothiazole, benzodiazole, thiazole, benzothiazole, tetrahydropyrane, morpholine, indoline, furan, triazole, isoxazole, pyrazole, thiophene, benzothiophene, pyrrole, pyrazine, pyrrolo[2,3b]pyridine, quinoline, isoquinoline, phthalazine, benzo-1 ,2,5- thiadiazole, indole, benzotriazole, benzoxazole oxopyrrolidine, pyrimidine, benzodioxolane, benzodioxane, carbazole and quinazoline; more preferably is piperazine;
optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another preferred embodiment of the invention according to formula (I) the compound is a compound, wherein in Re and Re· as defined in any of the embodiments of the present invention, the alkyl is preferably selected from C1-6 alkyl; more preferably, the Ci-6 alkyl is selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2- methylpropyl; and/or
the C2-6 -alkenyl is preferably selected from ethylene, propylene, butylene, pentylene, hexylene, isopropylene and isobutylene; and/or
the C2-6 -alkynyl is preferably selected from ethyne, propyne, butyne, pentyne, hexyne, isopropyne and isobutyne;
optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof. In another preferred embodiment of the invention according to formula (I) the compound is a compound, wherein in Rg, Rg·, Rg- and Rg- as defined in any of the embodiments of the present invention, the alkyl is preferably selected from C1-6 alkyl; more preferably, the Ci-6 alkyl is selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2- methylpropyl; and/or
the C2-6 -alkenyl is preferably selected from ethylene, propylene, butylene, pentylene, hexylene, isopropylene and isobutylene; and/or
the C2-6 -alkynyl is preferably selected from ethyne, propyne, butyne, pentyne, hexyne, isopropyne and isobutyne;
optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another preferred embodiment of the invention according to formula (I) the compound is a compound, wherein in R10, R-io·, R-io- and Rio- as defined in any of the embodiments of the present invention, the alkyl is preferably selected from C1-6 alkyl; more preferably, the C1-6 alkyl is selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2- methylpropyl; and/or
the C2-6 -alkenyl is preferably selected from ethylene, propylene, butylene, pentylene, hexylene, isopropylene and isobutylene; and/or
the C2-6 -alkynyl is preferably selected from ethyne, propyne, butyne, pentyne, hexyne, isopropyne and isobutyne;
optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another preferred embodiment of the invention according to formula (I) the compound is a compound, wherein in Ra, Ra and Ra- as defined in any of the embodiments of the present invention, the C1-6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2-methylpropyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another preferred embodiment of the invention according to formula (I) the compound is a compound, wherein in Rb, Rb·, Rb- and Rb- as defined in any of the embodiments of the present invention, the C1-6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2-methylpropyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another preferred embodiment of the invention according to formula (I) the compound is a compound, wherein in Ri and Rr as defined in any of the embodiments of the present invention, the C1-6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2-methylpropyl; and/or
the cycloalkyl is C3-8 cycloalkyl like cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, or cyclooctyl; preferably is C3-7 cycloalkyl like cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl; more preferably from C3-6 cycloalkyl like cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl; more preferably the cycloalkyl is cyclopropyl; and/or
the haloalkyl is selected from -CH2CI, -CH2F, -CHC , -CHF2, -CCI3, -CF3 and -CH2-CHCI2; more preferably the haloalkyl is -CHF2 or -CF3;
optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another preferred embodiment of the invention according to formula (I) the compound is a compound, wherein in R4and R4 as defined in any of the embodiments of the present invention, the C1-6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2-methylpropyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another preferred embodiment of the invention according to formula (I) the compound is a compound, wherein in R5 as defined in any of the embodiments of the present invention, the alkyl is preferably selected from C1-6 alkyl; more preferably, the C1-6 alkyl is selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2- methylpropyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another preferred embodiment of the invention according to formula (I) the compound is a compound, wherein in R6 as defined in any of the embodiments of the present invention, the alkyl is preferably selected from C alkyl; more preferably, the Ci-6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2-methylpropyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another preferred embodiment of the invention according to formula (I) the compound is a compound, wherein in Re and Re· as defined in any of the embodiments of the present invention, the alkyl is preferably selected from Ci-6 alkyl; more preferably, the Ci-6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2-methylpropyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another preferred embodiment of the invention according to formula (I) the compound is a compound, wherein in Rg, Rg·, Rg- and Rg- as defined in any of the embodiments of the present invention, the alkyl is preferably selected from Ci-6 alkyl; more preferably, the Ci-6 alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2-methylpropyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another preferred embodiment of the invention according to formula (I) the compound is a compound, wherein in R-io, R-io·, R-io- and Rio” as defined in any of the embodiments of the present invention, the alkyl is preferably selected from C alkyl; more preferably, the C alkyl is preferably selected from methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl and 2-methylpropyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another preferred embodiment of the invention according to formula (I) the compound is a compound, wherein one of W3 and W4 is S and the other is C;
and
Y is -C(O)-, -[C(RaRa')]n-, -CRa(CN)- or -CRa(CONRa'Ra ")-. wherein each Ra, Ra· and Ra- are hydrogen and n is 0, 1 or 2; and
X is selected from a bond, -[C(RbRb )]P-, -[C(RbRb )]PC(0)[C(Rb”Rb )]q-> unsubstituted aryl, preferably phenyl, unsubstituted aryl-C(O), preferably phenyl-C(O) and unsubstituted aryl-alkyl, preferably phenyl-alkyl, and wherein each Rb, Rb·, Rb- and Rb- are hydrogen; p is 0, 1 or 2; q is 0, 1 or 2; and group A is selected from:
Figure imgf000052_0001
wherein b is 0, 1 or 2; and
W2 is selected from O, C and N; each Ri and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted alkyl-cycloalkyl; NR4R4', -C(0)0R4- and substituted or unsubstituted haloalkyl; and wherein each R4 and R4' are independently selected from hydrogen, substituted or unsubstituted C1-6 alkyl, , and -C(0)0R4·-; wherein R4 " is substituted or unsubstituted C1-6 alkyl;
R5 is selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, CN, - C(0)0R3; -S(0)20R3· ; wherein each R3 and R3' are hydrogen,
R6 is selected from hydrogen, halogen and substituted or unsubstituted C1-6 alkyl; and
R2 is a group of the following formula:
Figure imgf000052_0002
m is 1 ; r is 1 ; t is 0 or 1 ;
Wi is C or N; each Rg, Rg·, Rg- and Rg- are hydrogen; each R10, Rio·, R10” and Rio- are hydrogen;
R7 is selected from substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted aryl, preferably phenyl, and substituted or unsubstituted aromatic heterocyclyl;
Re is selected from hydrogen and substituted or unsubstituted C1-6 alkyl;
Re’ is selected from hydrogen and substituted or unsubstituted C1-6 alkyl; or
R7 and Re taken together with -N(R8')-[CH2]t- atoms to which they are attached may form a substituted or unsubstituted heterocyclyl optionally fused with a substituted or unsubstituted aryl group; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another preferred embodiment of the invention according to formula (I) the compound is a compound, wherein
W3 is S and W4 is C;
and
Y is -C(O)-, -[C(RaRa )]n-, -CRa(CN)- or -CRa(CONRa Ra )-, wherein each Ra, Ra· and Ra- are hydrogen and n is 0, 1 or 2; and X is selected from a bond, -[C(RbRb )]P-, -[C(RbRb )]PC(0)[C(Rb"Rb )]q-, unsubstituted aryl, preferably phenyl, unsubstituted aryl-C(O), preferably phenyl-C(O) and unsubstituted aryl-alkyl, preferably phenyl-alkyl, and wherein each Rb, Rb·, Rb- and Rb- are hydrogen; p is 0, 1 or 2; q is 0, 1 or 2; and group A is selected from:
Figure imgf000054_0001
wherein b is 0, 1 or 2; and
W2 is selected from O, C and N; each Ri and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted alkyl-cycloalkyl; NR4R4', -C(0)OR4- and substituted or unsubstituted haloalkyl; and wherein each R4 and R4' are independently selected from hydrogen, substituted or unsubstituted C1-6 alkyl, , and -C(0)OR4·-; wherein R4" is substituted or unsubstituted C1-6 alkyl;
R5 is selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, CN, - C(0)OR3; -S(0)20R3' ; wherein each R3 and Ry are hydrogen,
R6 is selected from hydrogen, halogen and substituted or unsubstituted C1-6 alkyl; and
R2 is a group of the following formula:
Figure imgf000055_0001
m is 1 ; r is 1 ; t is 0 or 1 ;
Wi is C or N; each Rg, Rg·, Rg- and Rg- are hydrogen; each Rio, Rio·, Rio” and Rio- are hydrogen;
R7 is selected from substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted aryl, preferably phenyl, and substituted or unsubstituted aromatic heterocyclyl;
Re is selected from hydrogen and substituted or unsubstituted C1-6 alkyl;
Re’ is selected from hydrogen and substituted or unsubstituted C1-6 alkyl;
R7 and Re taken together with -N(R8')-[CH2]t- atoms to which they are attached may form a substituted or unsubstituted heterocyclyl optionally fused with a substituted or unsubstituted aryl group; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof. In another preferred embodiment of the invention according to formula (I) the compound is a compound, wherein
W3 is S and W4 is C;
and
Y is -C(O)-, -[C(RaRa )]n-, -CRa(CN)- or -CRa(CONRa Ra )-, wherein each Ra, Ra· and Ra- are hydrogen and n is 0, 1 or 2; and X is selected from a bond, -[C(RbRb )]P-, -[C(RbRb )]PC(0)[C(Rb”Rb )]q-> phenyl, preferably phenyl-C(O) and phenyl-alkyl, and wherein each Rb, Rb·, Rb- and Rb- are hydrogen, p is 0, 1 or 2; q is 0, 1 or 2; and group A is:
Figure imgf000056_0001
wherein b is 0, 1 or 2; and
W2 is selected from O, C and N; and each R1 and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C alkyl, substituted or unsubstituted alkyl-cycloalkyl; NR4R4', -C(0)OR4- and substituted or unsubstituted haloalkyl; wherein each R4 and R4· are independently selected from hydrogen, substituted or unsubstituted C1-6 alkyl, , and -C(0)0R4”-; wherein R4- is substituted or unsubstituted C1-6 alkyl; and R5 is selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, CN, - C(0)0R3 and -S(0)20R3· ; wherein each R3 and R3' are hydrogen; and
R6 is selected from hydrogen and substituted or unsubstituted C1-6 alkyl; and
R2 is a group of the following formula
Figure imgf000057_0001
wherein m is 1 ; r is 1 ; t is 0 or 1 ;
Wi is C or N; each Rg, Rg·, Rg- and Rg- are hydrogen; each R10, Rio·, R10” and Rio- are hydrogen;
R7 is selected from substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted aryl, preferably phenyl, and substituted or unsubstituted aromatic heterocyclyl; Re is selected from hydrogen and substituted or unsubstituted Ci-6 alkyl;
Re’ is selected from hydrogen and substituted or unsubstituted Ci-6 alkyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In another preferred embodiment of the invention according to formula (I) the compound is a compound, wherein
W3 is S and W4 is C; and
Y is -C(O)-; and
X is selected from a bond and -[C(RbRb )]P-, wherein each Rb, Rb· are hydrogen and p is 1 or 2; and group A is:
Figure imgf000058_0001
wherein b is 1 and W2 is N; each R1 and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted alkyl-cycloalkyl; NR4R4', -C(0)0R4- and substituted or unsubstituted haloalkyl; and each R4 and R4· are independently selected from hydrogen, substituted or unsubstituted C1-6 alkyl, , and -C(0)0R4”-; wherein R4- is substituted or unsubstituted Ci-6 alkyl; and
R5 is selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, CN, - C(0)0R3 and -S(0)20R3· ; wherein each R3 and R3' are hydrogen; and
R6 is selected from hydrogen and substituted or unsubstituted C1-6 alkyl; and
R2 is a group of the following formula
Figure imgf000059_0001
wherein m is 1 ; r is 1 ; t is 0;
Wi is C; each Rg, Rg·, Rg- and Rg- are hydrogen; each R10, Rio·, R10” and Rio- are hydrogen;
R7 is substituted or unsubstituted aryl, preferably phenyl;
Re is selected from hydrogen and substituted or unsubstituted Ci-6 alkyl; and
Re’ is selected from hydrogen and substituted or unsubstituted C1-6 alkyl; optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof. In a particular embodiment of the compound according to the invention of formula (I) the halogen is fluorine or chlorine. optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
In a preferred further embodiment, the compound of formula (I) is selected from:
[1] (1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[2] (1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(morpholino)methanone;
[3] (1-((1s,4s)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[4] tert-butyl 4-(1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazole-3-carbonyl)-1 ,4-diazepane-1-carboxylate;
[5] (1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(4-methyl-1 ,4-diazepan-1-yl)methanone;
[6] (1-((1 r,4R)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)((3S,5R)-3,5-dimethylpiperazin-1-yl)methanone;
[7] (1-((1 r,4r)-4-(dimethylamino)-4-(3-hydroxyphenyl)cyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[8] (1-((1s,4s)-4-(dimethylamino)-4-(3-fluorophenyl)cyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[9] (1-((1s,4s)-4-(dimethylamino)-4-(4-fluorophenyl)cyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone; [10] (1-((1s,4s)-4-(dimethylamino)-4-(3-methoxyphenyl)cyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[11] (1-((1s,4s)-4-(dimethylamino)-4-(4-methoxyphenyl)cyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[12] (1-((1 r,4r)-4-(dimethylamino)-4-(3-fluorophenyl)cyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[13] (1-((1 r,4r)-4-(dimethylamino)-4-(4-fluorophenyl)cyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[14] (1-((1 r,4r)-4-(dimethylamino)-4-(3-methoxyphenyl)cyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[15] (1-((1s,4s)-4-(dimethylamino)-4-(3-hydroxyphenyl)cyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[16] (1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(4-ethylpiperazin-1-yl)methanone;
[17] (1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(piperidin-1-yl)methanone;
[18] (1-((1 r,4r)-4-(methylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[19] (1-((1s,4s)-4-(dimethylamino)-4-(2-methoxyphenyl)cyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[20] (S)-tert-butyl 4-(1-((1 r,4S)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazole-3-carbonyl)-3-methylpiperazine-1-carboxylate;
[21] (R)-tert-butyl 4-(1-((1 r,4R)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazole-3-carbonyl)-3-methylpiperazine-1-carboxylate;
[22] (1-((1s,4s)-4-(methylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[23] (1-((1 r,4r)-4-(dimethylamino)-4-(4-methoxyphenyl)cyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone; [24] tert-butyl 5-(1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazole-3-carbonyl)hexahydropyrrolo[3,4-c]pyrrole-2(1 H)-carboxylate;
[25] tert-butyl 6-(1-((1 r,4r)-4-(dimethylannino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazole-3-carbonyl)-2,6-diazaspiro[3.4]octane-2-carboxylate;
[26] (1-((1 r,4r)-4-((dimethylannino)nnethyl)-4-(3-nnethoxyphenyl)cyclohexyl)-4, 5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-nnethylpiperazin-1-yl)nnethanone;
[27] (1-((1s,4s)-4-((dimethylamino)nnethyl)-4-(3-nnethoxyphenyl)cyclohexyl)-4, 5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-nnethylpiperazin-1-yl)nnethanone;
[28] tert-butyl 4-(1-((1 r,4r)-4-(dimethylannino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazole-3-carbonyl)-6,6-difluoro-1 ,4-diazepane-1-carboxylate;
[29] (1-((1 r,4R)-4-(dinnethylannino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)((R)-3-(hydroxymethyl)-4-nnethylpiperazin-1-yl)nnethanone;
[30] (1-((1 r,4S)-4-(dimethylannino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)((S)-3-(hydroxynnethyl)-4-nnethylpiperazin-1-yl)nnethanone;
[31] tert-butyl 2-(difluoromethyl)-4-(1-((1 r,4r)-4-(dinnethylannino)-4-phenylcyclohexyl)-4,5- dihydro-1 H-thieno[2,3-g]indazole-3-carbonyl)piperazine-1-carboxylate;
[32] (4-(cyclopropylmethyl)piperazin-1-yl)(1-((1 r,4r)-4-(dinnethylannino)-4- phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)methanone;
[33] (4,6-dimethyl-1 ,4-diazepan-1-yl)(1-((1 r,4r)-4-(dinnethylannino)-4-phenylcyclohexyl)- 4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)nnethanone;
[34] (4,7-dimethyl-1 ,4-diazepan-1-yl)(1-((1 r,4r)-4-(dinnethylannino)-4-phenylcyclohexyl)- 4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)nnethanone;
[35] (1-((1 r,4S)-4-(dimethylannino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)((S)-3-(dinnethylannino)piperidin-1-yl)nnethanone;
[36] (1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(2-methyl-2,6-diazaspiro[3.4]octan-6-yl)nnethanone;
[37] (1-(4-(4-(dimethylannino)-4-(3-hydroxyphenyl)piperidin-1-yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methyl-1 ,4-diazepan-1-yl)nnethanone; [38] (1-(4-(4-(dimethylamino)-4-phenylpiperidin-1-yl)phenyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[39] (1-(3-(4-(dimethylamino)-4-phenylpiperidin-1-yl)phenyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[40] tert-butyl (1-(1-(4-(4-(dimethylamino)-4-phenylpiperidin-1-yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazole-3-carbonyl)pyrrolidin-3-yl)(methyl)carbamate;
[41] (1-(4-(4-(dimethylamino)-4-ethylpiperidin-1-yl)phenyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[42] (1-(4-(2-methyl-3,4-dihydro-2H-spiro[isoquinoline-1 ,4'-piperidin]-1 '-yl)phenyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[43] (1-(4-(4-(dimethylamino)-4-(3-hydroxyphenyl)piperidin-1-yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[44] (1-(4-(4-(dimethylamino)-4-(3-methoxyphenyl)piperidin-1-yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[45] (1-(4-(4-(methylamino)-4-phenylpiperidin-1-yl)phenyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[46] (1-(4-(4-(3-hydroxyphenyl)-4-(methylamino)piperidin-1-yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[47] (1-(3-(4-(methylamino)-4-phenylpiperidin-1-yl)phenyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[48] (1-(3-(2-methyl-3,4-dihydro-2H-spiro[isoquinoline-1 ,4'-piperidin]-1 '-yl)phenyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[49] (1-(3-(4-(dimethylamino)-4-(3-methoxyphenyl)piperidin-1-yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[50] (1-(3-(4-(dimethylamino)-4-(3-hydroxyphenyl)piperidin-1-yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[51] (1-(4-(4-((dimethylamino)methyl)-4-phenylpiperidin-1-yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone; [52] (1-(4-(3-(dimethylamino)-3-phenylpiperidin-1-yl)phenyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[53] (1-(3-(4-((dimethylamino)methyl)-4-phenylpiperidin-1-yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[54] (1-(3-(4-((dimethylamino)methyl)-4-(3-hydroxyphenyl)piperidin-1-yl)phenyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[55] (1-(4-(4-(dimethylamino)-4-phenylpiperidin-1-yl)phenyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(4-methyl-1 ,4-diazepan-1-yl)methanone;
[56] (1-(3-(4-(dimethylamino)-4-(pyridin-2-yl)piperidin-1-yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[57] (1-(3-(4-(dimethylamino)-4-(pyridin-2-yl)piperidin-1-yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[58] (1-(4-(4-(dimethylamino)-4-(pyridin-2-yl)piperidin-1-yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[59] (1-(4-(4-(dimethylamino)-4-(4-methoxyphenyl)piperidin-1-yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[60] (1-(4-(4-(dimethylamino)-4-(2-methoxyphenyl)piperidin-1-yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[61] (1-(4-(4-(dimethylamino)-4-(3-fluorophenyl)piperidin-1-yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[62] (1-(4-(4-(dimethylamino)-4-(4-fluorophenyl)piperidin-1-yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[63] (1-(3-((4-(dimethylamino)-4-(3-hydroxyphenyl)piperidin-1-yl)methyl)phenyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[64] (1-(4-((4-(dimethylamino)-4-phenylpiperidin-1-yl)methyl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[65] (1-(3-((4-(dimethylamino)-4-phenylpiperidin-1-yl)methyl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone; [66] (1-(4-((4-(dimethylamino)-4-(3-hydroxyphenyl)piperidin-1-yl)methyl)phenyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[67] (4-(dimethylamino)-4-(3-hydroxyphenyl)piperidin-1-yl)(3-(3-(4-methylpiperazine-1- carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1-yl)phenyl)methanone;
[68] (4-(dimethylamino)-4-phenylpiperidin-1-yl)(4-(3-(4-methylpiperazine-1-carbonyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-1-yl)phenyl)methanone;
[69] (4-(dimethylamino)-4-phenylpiperidin-1-yl)(3-(3-(4-methylpiperazine-1-carbonyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-1-yl)phenyl)methanone;
[70] (2-methyl-3,4-dihydro-2H-spiro[isoquinoline-1 ,4'-piperidin]-1 '-yl)(4-(3-(4- methylpiperazine-1-carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1- yl)phenyl)methanone;
[71] (2-methyl-3,4-dihydro-2H-spiro[isoquinoline-1 ,4'-piperidin]-1 '-yl)(3-(3-(4- methylpiperazine-1-carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1- yl)phenyl)methanone;
[72] (4-(dimethylamino)-4-(3-methoxyphenyl)piperidin-1-yl)(4-(3-(4-methylpiperazine-1- carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1-yl)phenyl)methanone;
[73] (4-(methylamino)-4-phenylpiperidin-1-yl)(4-(3-(4-methylpiperazine-1-carbonyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-1-yl)phenyl)methanone;
[74] (4-(dimethylamino)-4-(3-hydroxyphenyl)piperidin-1-yl)(4-(3-(4-methylpiperazine-1- carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1-yl)phenyl)methanone;
[75] (4-(dimethylamino)-4-(3-methoxyphenyl)piperidin-1-yl)(3-(3-(4-methylpiperazine-1- carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1-yl)phenyl)methanone;
[76] (4-(methylamino)-4-phenylpiperidin-1-yl)(3-(3-(4-methylpiperazine-1-carbonyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-1-yl)phenyl)methanone;
[77] (1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-1 H-thieno[2,3-g]indazol-3-yl)(4- methylpiperazin-1-yl)methanone;
[78] (7-Bromo-1-((1 r,4r)-4-(dimethylannino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[79] 1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-3-(4-methylpiperazine-1-carbonyl)- 4,5-dihydro-1 H-thieno[2,3-g]indazole-7-carbonitrile; [80] 1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-3-(4-methylpiperazine-1 -carbonyl)-
4,5-dihydro-1 H-thieno[2,3-g]indazole-7-carboxylic acid;
[81] 1-((1 r,4r)-4-(dimethylannino)-4-phenylcyclohexyl)-3-(4-nnethylpiperazine-1-carbonyl)-
4.5-dihydro-1 H-thieno[2,3-g]indazole-7-sulfonic acid;
[82] (1-((1 r,4r)-4-((2-fluoroethyl)nnethylannino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-nnethylpiperazin-1-yl)nnethanone;
[83] (1-((1 r,4r)-4-((2,2-difluoroethyl)(nnethyl)annino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-nnethylpiperazin-1-yl)nnethanone;
[84] (1-((1 r,4r)-4-((2,2-difluoroethyl)(nnethyl)annino)-4-phenylcyclohexyl)-1 H-thieno[2,3- g]indazol-3-yl)(4-nnethylpiperazin-1-yl)nnethanone;
[85] (1 ,4-diazepan-1-yl)(1-((1 r,4r)-4-(dimethylannino)-4-phenylcyclohexyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)methanone;
[86] (1-((1 r,4r)-4-(dinnethylannino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(piperazin-1-yl)nnethanone;
[87] (1-((1 r,4S)-4-(dinnethylannino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)((S)-2-nnethylpiperazin-1-yl)nnethanone;
[88] (1-((1 r,4r)-4-(dinnethylannino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(hexahydropyrrolo[3,4-c]pyrrol-2(1 H)-yl)nnethanone;
[89] (1-((1 r,4r)-4-(dinnethylannino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(2,6-diazaspiro[3.4]octan-6-yl)nnethanone;
[90] (6,6-difluoro-1 ,4-diazepan-1-yl)(1-((1 r,4r)-4-(dinnethylannino)-4-phenylcyclohexyl)-
4.5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)methanone;
[91] (3-(difluoromethyl)piperazin-1-yl)(1-((1 r,4r)-4-(dinnethylannino)-4-phenylcyclohexyl)-
4.5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)nnethanone;
[92] (1-(4-(4-(dinnethylannino)-4-phenylpiperidin-1-yl)phenyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(3-(nnethylannino)pyrrolidin-1-yl)nnethanone;
[93] (1-((1 r,4R)-4-(dinnethylannino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)((R)-2-nnethylpiperazin-1-yl)nnethanone; [94] (7-bromo-1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(piperazin-1-yl)methanone;
[95] (1-((1 r,4R)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)((R)-3-(hydroxymethyl)piperazin-1-yl)methanone;
[96] (1-((1 r,4S)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)((S)-3-(hydroxymethyl)piperazin-1-yl)methanone;
[97] (1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(4-(2-fluoroethyl)piperazin-1-yl)methanone;
[98] (4-(2,2-difluoroethyl)piperazin-1-yl)(1-((1 r,4r)-4-(dimethylannino)-4- phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)methanone;
[99] (2-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-2H-thieno[2,3- g]indazol-3-yl)(4-ethylpiperazin-1-yl)methanone;
[100] (2-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-2H-thieno[2,3- g]indazol-3-yl)(4-methyl-1 ,4-diazepan-1-yl)methanone;
[101] (2-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-2H-thieno[2,3- g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[102] 2-(1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)-2-(4-methylpiperazin-1-yl)acetonitrile;
[103] 2-(1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)-2-(4-methylpiperazin-1-yl)acetamide;
[104] (1 r,4r)-4-(8-(tert-butyl)-3-((4-methylpiperazin-1-yl)methyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-1-yl)-N,N-dimethyl-1-phenylcyclohexanamine;
[105] N,N-dimethyl-1-(4-(3-((4-methylpiperazin-1-yl)methyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-1-yl)phenyl)-4-phenylpiperidin-4-amine;
[106] (1 r,4r)-N,N-dimethyl-4-(3-((4-methylpiperazin-1-yl)methyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-1-yl)-1-phenylcyclohexanamine;
[107] (1-((4-(dimethylamino)-4-phenylcyclohexyl)methyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone; [108] (1-(2-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)ethyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[109] (1-(2-(4-(3-hydroxyphenyl)-4-(methylamino)piperidin-1-yl)ethyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[1 10] (1-(2-(4-(dimethylamino)-4-(3-hydroxyphenyl)piperidin-1-yl)ethyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[1 1 1] (1-(2-(4-((dimethylamino)methyl)-4-(3-hydroxyphenyl)piperidin-1-yl)ethyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[1 12] (1-(2-(4-(methylamino)-4-phenylpiperidin-1-yl)ethyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[1 13] (2-(2-(4-(dimethylamino)-4-(3-hydroxyphenyl)piperidin-1-yl)ethyl)-4,5-dihydro-2H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[1 14] (1-(2-(4-(dimethylamino)-4-(4-fluorophenyl)piperidin-1-yl)ethyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[1 15] (1-(2-(4-(dimethylamino)-4-(3-fluorophenyl)piperidin-1-yl)ethyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[1 16] (1-(2-(4-((dimethylamino)methyl)-4-phenylpiperidin-1-yl)ethyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[1 17] (1-(2-(4-(dimethylamino)-4-phenylpiperidin-1-yl)ethyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[1 18] 1-(4-(dimethylamino)-4-phenylpiperidin-1-yl)-2-(3-(4-methylpiperazine-1-carbonyl)- 4,5-dihydro-1 H-thieno[2,3-g]indazol-1-yl)ethanone;
[1 19] 1-(4-(dimethylamino)-4-(3-fluorophenyl)piperidin-1-yl)-2-(3-(4-methylpiperazine-1- carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1-yl)ethanone;
[120] 1-(4-(dimethylamino)-4-(4-fluorophenyl)piperidin-1-yl)-2-(3-(4-methylpiperazine-1- carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1-yl)ethanone;
[121] 1-(4-(dimethylamino)-4-(4-methoxyphenyl)piperidin-1-yl)-2-(3-(4-methylpiperazine-1- carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1-yl)ethanone; [122] 1-(4-(dimethylamino)-4-(3-methoxyphenyl)piperidin-1-yl)-2-(3-(4-methylpiperazine-1- carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1-yl)ethanone;
[123] 1-(4-((dimethylamino)methyl)-4-phenylpiperidin-1-yl)-2-(3-(4-methylpiperazine-1- carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1-yl)ethanone;
[124] 1-(4-(dimethylamino)-4-(2-methoxyphenyl)piperidin-1-yl)-2-(3-(4-methylpiperazine-1- carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1-yl)ethanone;
[125] 1-(4-(diethylamino)-4-phenylpiperidin-1-yl)-2-(3-(4-methylpiperazine-1-carbonyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-1-yl)ethanone;
[126] 1-(4-(dimethylamino)-4-(3-hydroxyphenyl)piperidin-1-yl)-2-(3-(4-methylpiperazine-1- carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1-yl)ethanone;
[127] 1-(4-((dimethylamino)methyl)-4-(3-hydroxyphenyl)piperidin-1-yl)-2-(3-(4- methylpiperazine-1-carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1-yl)ethanone; tert-butyl 4-(1-((1 r,4r)-4-(dimethylannino)-4-(4-nnethoxyphenyl)cyclohexyl)-4,5- dihydro-1 H-thieno[2,3-g]indazole-3-carbonyl)piperazine-1-carboxylate; tert-butyl (1-(1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazole-3-carbonyl)azetidin-3-yl)(methyl)carbamate;
((S)-4-benzyl-3-methylpiperazin-1-yl)(1-((1 r,4S)-4-(dinnethylannino)-4- phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)nnethanone;
((R)-4-benzyl-3-methylpiperazin-1-yl)(1-((1 r,4R)-4-(dimethylannino)-4- phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)methanone;
(1-((1 r,4R)-4-(dinnethylannino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)((R)-3-methylpiperazin-1-yl)nnethanone;
(1-((1 r,4S)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)((S)-3-methylpiperazin-1-yl)methanone; 1-tert-butyl 2-methyl 4-(1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-
[134]
dihydro-1 H-thieno[2,3-g]indazole-3-carbonyl)piperazine-1 ,2-dicarboxylate;
(1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-1 ,4-
[136] dihydrothieno[3',2':4,5]cyclopenta[1 ,2-c]pyrazol-3-yl)(4-methylpiperazin-1- yl)methanone;
(1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-1 ,4-
[137] dihydrothieno[2',3':4,5]cyclopenta[1 ,2-c]pyrazol-3-yl)(4-methylpiperazin-1- yl)methanone; tert-butyl ((S)-1-(1-((1 r,4S)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-
[138]
thieno[2,3-g]indazole-3-carbonyl)piperidin-3-yl)carbamate;
(1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[3,2-
[139]
g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[140] (1-((1 r,4r)-4-(dimethylamino)-4-(4-methoxyphenyl)cyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(piperazin-1-yl)methanone;
[141] (1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(3-(methylamino)azetidin-1-yl)methanone;
[142] 4-(1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazole-3-carbonyl)piperazine-2-carboxylic acid;
[143] methyl 4-(1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazole-3-carbonyl)piperazine-2-carboxylate;
[144] (R)-4-(1-((1 r,4R)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazole-3-carbonyl)-2-(hydroxymethyl)piperazine-1 -carboxylic acid;
[145] ((S)-3-aminopiperidin-1-yl)(1-((1 r,4S)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)methanone;
[146] N-((1s,4s)-4-(dimethylamino)-4-phenylcyclohexyl)-2-(3-(4-methylpiperazine-1- carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1-yl)acetamide; [147] N-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-2-(3-(4-methylpiperazine-1- carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1-yl)acetannide;
[148] (1-(2-(4-(dimethylamino)-4-(4-methoxyphenyl)piperidin-1-yl)ethyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[149] (1-(2-(4-(dimethylamino)-4-(4-fluorophenyl)piperidin-1-yl)ethyl)-1 H-thieno[2,3- g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[150] (1-((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(3-(dimethylamino)azetidin-1-yl)methanone; and
[151] 1-(4-(Methylamino)-4-phenylpiperidin-1-yl)-2-(3-(4-methylpiperazine-1-carbonyl)- 4,5-dihydro-1 H-thieno[2,3-g]indazol-1-yl)ethanone, optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof. A preferred aspect of the invention is also a process for obtaining a compound of formula (I) as described above. Several procedures have been developed for obtaining all the compounds of the invention, and the procedures will be explained below in methods A- C.
The obtained reaction products may, if desired, be purified by conventional methods, such as crystallization and chromatography. Where the processes described below for the preparation of compounds of the invention give rise to mixtures of stereoisomers, these isomers may be separated by conventional techniques such as preparative chromatography. The compounds may be prepared in racemic form, or individual enantiomers may be prepared either by enantiospecific synthesis or by resolution. One preferred pharmaceutically acceptable form of a compound of the invention is the crystalline form, including such form in pharmaceutical composition. In the case of salts and also solvates of the compounds of the invention the additional ionic and solvent moieties must also be non-toxic. The compounds of the invention may present different polymorphic forms, it is intended that the invention encompasses all such forms. The compounds of formula (I) can be obtained by following the methods described below. As it will be obvious to one skilled in the art, the exact method used to prepare a given compound may vary depending on its chemical structure.
METHOD A
Method A represents a first process for synthesizing compounds according to formula
In this sense, in another aspect, the invention refers to a process for the preparation of a compound of formula (I)
Figure imgf000072_0001
said process comprising treating a compound of formula (VII)
Figure imgf000072_0002
with a compound of formula (V):
Figure imgf000072_0003
(V)
wherein
Ri, Rr, R2, R5, R6, W3, W4, X, Y and A have the same meaning as indicated before and Z is selected from OH, Cl, N(OMe)Me and H.
In a particular embodiment of the method according to the invention, W3 is S and W4 is C.
In a particular embodiment of the method according to the invention, Z is selected from OH and Cl.
In a particular embodiment of the method according to the invention, the compound of formula (la) wherein Y= -C(O)-, can be prepared by treating a compound of formula V, where Z represents OH or a chlorine atom, with an amine of formula VII (see Scheme 1 )·
When Z is OH, the reaction is carried out using a suitable coupling reagent such as N- (3-dimethylaminopropyl)-/V'-ethylcarbodiimide (EDC), dicyclohexylcarbodiimide (DCC), /V-[(dimethylamino)-1 H- 1 ,2,3-triazolo-[4,5-b]pyridin-1 -ylmethylene]-/V- methylmethanaminium hexafluorophosphate N- oxide (HATU) or L/,L/,L/',L/'-tetramethyl- 0-(1H- benzotriazol-1 -yl)uronium hexafluorophosphate (HBTU), optionally in the presence of 1 -hydroxybenzotriazole, optionally in the presence of an organic base such as /V-methylmorpholine or /V,/V-diisopropylethylamine, in a suitable solvent such as dichloromethane or dimethylformamide, and at a suitable temperature, preferably at room temperature. In another particular embodiment, when Z is a chlorine atom, the reaction is carried out in a suitable solvent, such as dichloromethane, tetrahydrofuran, ethyl acetate or ethyl acetate-water mixtures; in the presence of an organic base such as triethylamine or /V,/V-diisopropylethylamine or an inorganic base such as K2CO3; and at a suitable temperature, preferably comprised between 0 °C and room temperature. Additionally, an activating agent such as 4-dimethylaminopyridine can be used.
In another particular embodiment, a compound of formula (lb) wherein Y=-[C(RaRa )]n, more particularly wherein Y=CH2, can be prepared by reduction of a compound of formula (la) using a suitable reagent, such as alane in the presence of a suitable solvent, such as tetrahydrofuran at a suitable temperature, such as between -10 °C and room temperature. In another particular embodiment of the method according to the invention, a compound of formula (lc) wherein Y= -CRaRa (CN)-, in particular -CH2(CN)-, can be prepared by reaction between a compound of formula V, where Z represents hydrogen, and an amine of formula VII in the presence of potassium cyanide, using a suitable solvent, such as mixtures of hhCbMeOH at a suitable temperature, such as room temperature.
In a particular embodiment of the method according to the invention, a compound of formula (Id) wherein Y= -CRa(CONRa Ra”)-, more particularly Y=CH2-(CONH2), can be prepared by hydrolisis of a compound of formula (lc) wherein Y=-CH2(CN)-, using a suitable reagent, such as sodium hydroxide and hydrogen peroxide in the presence of a suitable solvent, such as mixtures of MeOH:DMSO at a suitable temperature, such as room temperature.
A compound of formula V, where Z represents O-alkyl, can be prepared by treating a compound of formula II with a suitable hydrazine of formula III, as shown in Scheme 1 , in the presence of a suitable solvent, such as ethanol, at a suitable temperature, preferably heating.
A compound of formula V, where Z represents OH, can be prepared by reacting a compound of formula V, where Z represents O-alkyl, in basic media, such as sodium hydroxide in the presence of a suitable solvent, such as water, at a suitable temperature, between room temperature and 100 °C and optionally under microwave heating.
Alternatively, a compound of formula V, where Z represents OH, may be converted to a compound of formula V, where Z represents a chlorine atom, using thionyl chloride, in a suitable solvent, such as pyridine, at a suitable temperature, such as room temperature
A compound of formula V, where Z represents hydrogen, can be prepared by reduction of a compound of formula V, where Z represents O-alkyl, using a suitable reagent, such as didisobutylaluminiumhydride in the presence of a suitable solvent, such as tetrahydrofuran at a suitable temperature, such as between -10 °C and room temperature.
Alternatively, a compound of formula V, where Z represents hydrogen, can be prepared using a two-step procedure that involves conversion of a compound of formula V, where Z represents OH, to a compound of formula V where Z represents N(OMe)Me, which is then reduced. The following conditions apply: A compound of formula V, where Z represents N(OMe)Me, can be prepared by treating a compound of formula V, where Z represents OH, with N,O-dimethylhydroxylamine using a suitable coupling reagent such as /V-(3-dimethylaminopropyl)-/V'- ethylcarbodiimide (EDO), dicyclohexylcarbodiimide (DCC), /V-[(dimethylamino)-1 /-/- 1 ,2,3-triazolo-[4,5-b] pyridin-1 -ylmethylene]-/V-methylmethanaminium
hexafluorophosphate N- oxide (HATU) or /V,/V,/V',/V'-tetramethyl-0-( //-/-benzotriazol-1- yl)uronium hexafluorophosphate (HBTU), optionally in the presence of 1- hydroxybenzotriazole, optionally in the presence of an organic base such as N- methylmorpholine or /V,/V-diisopropylethylamine, in a suitable solvent such as dichloromethane or dimethylformamide, and at a suitable temperature, preferably at room temperature.
A compound of formula V, where Z represents hydrogen, can be prepared by reduction of a compound of formula V, where Z represents N(OMe)Me using a suitable reagent, such as lithiumaluminium hydride in the presence of a suitable solvent, such as tetrahydrofuran at a suitable temperature, such as 0 °C.
The reactions steps are shown in Scheme 1 below in more detail:
Figure imgf000075_0001
Scheme 1 METHOD B
As it is shown in Sheme 1 , alternatively, the compound of the invention can be produced according to the process explained below. Thus, in another aspect of the present invention relates to a process for the preparation of a compound of formula (I)
Figure imgf000076_0001
wherein Ri, Rr, R2, Rs, R6, W3, W4, X, Y and A have the same meaning as indicated before, said process comprising a) treating a compound of formula (VII)
Figure imgf000076_0002
with a compound of formula (VIII):
Figure imgf000076_0003
wherein
R1, Rr, R2, Rs, R6, W3, W4, X, Y and A have the same meaning as indicated before; and Z is OH or O-alkyl; and b) treating the compound obtained in step a) with a compound of formula XI
R2-X-G
(XI) wherein R2, and X have the same meaning as indicated before; and G is
B(OH)2 when X is aryl, and G is a leaving group when X is alkylene.
In a particular embodiment of the method according to the invention, W3 is S and W4 is C. Depending on the nature of X, different meanings of G and different reaction conditions apply:
When X is aryl, G is a B(OH)2 group and the reaction of XII with XI is carried out in the presence of a copper salt, such as Cu(OAC)2, in a suitable solvent, such as dichloromethane, at a suitable temperature, such as room temperature. When X is alkylene, G is a leaving group and the alkylation reaction may be carried out under alkylation conditions, in a suitable solvent, such as dimethylacetamide, isopropanol, ethanol or acetonitrile; optionally in the presence of an organic base such as triethylamine or diisopropylethylamine or an inorganic base such as K2CO3 or CS2CO3; at a suitable temperature comprised between room temperature and the reflux temperature, preferably heating, or alternatively, the reactions can be carried out in a microwave reactor.
As an alternative procedure, these reaction conditions with a compound of formula XI may be applied to compounds of formula VIII, wherein Z represents O-alkyl or OH, to give, respectively, compounds of formula V, wherein Z represents O-alkyl or OH. METHOD C
Method C represents a third process for synthesizing compounds according to formula
(I).
Thus, in another aspect, the invention refers to a process for the preparation of a compound of formula (I)
Figure imgf000078_0001
wherein Ri, Rr, R2, Rs, R6, W3, W4, X, and A have the same meaning as as indicated before and Y represents -C(O)- said process comprising: a) treating a compound of formula (XIV)
with a compound of formul
Figure imgf000078_0002
Figure imgf000078_0003
b) treating the compound obtained in step a) with R2H; wherein Z is O-alkyl, OH or a leaving group (LG).
In a particular embodiment of the method according to the invention, W3 is S and W4 is C. A compound of formula XIV may be transformed to a compound of formula XVI as despeicted in Scheme 2 using the conditions described above for the preparation of compound of formula I wherein Y= -C(O)-.
Figure imgf000079_0001
Scheme 2
A compound of formula XVI, where M represents OH, may be converted to a compound of formula XVII, where M represents a leaving group, such as chlorine. The reaction may be carried out using thionyl chloride in a suitable solvent, such as dichloromethane, at a suitable temperature, such as room temperature.
The compound of formula I, wherein Y represents -C(O)- may be obtained by reaction of a compound of formula XVII with a suitable reagent of formula XVIII using alkylation conditions such as the described above.
The compounds of formula II may be prepared, as described in Scheme 3, by reacting a compound of formula XIX with a compound of formula XX, where Z is O-alkyl, in the presence of a base such as lithiumhexamethyldisilazane in a suitable solvent such as tetrahydrofuran at a suitable temperature, such as room temperature.
Figure imgf000079_0002
Scheme 3 The compounds of formula III, VII, XI, XIII, XVIII, XIX and XX used in the methods and schemes disclosed above are commercially available or can be synthesized following common procedures described in the literature and exemplified in the synthesis of some intermediates.
In another aspect, the invention refers to the use of a compound selected from
Figure imgf000080_0001
wherein R2, Rs, R6, W3, W4 and X have the same meaning as indicated above and Z is O-alkyl, OH; H or Cl for the manufacture of a compound a compound according to the invention as described above according to formula (I). In a particular embodiment of the invention, W3 is S and W4 is C.
Certain compounds of the present invention can also be obtained starting from other compounds of formula (I) by appropriate conversion reactions of functional groups, in one or several steps, using well-known reactions in organic chemistry under standard experimental conditions. As a way of example, some of these conversions include the reduction of a double bond from a tetrahydropyridine to a piperidine derivative or the reductive amination of an amino group with an aldehyde or ketone, or alternatively the reaction of an amino group with an alkylating agent, to prepare a further substituted amino group.
In addition, a compound of formula (I) that shows chirality can also be obtained by resolution of a racemic compound of formula (I) either by chiral preparative HPLC or by crystallization of a diastereomeric salt or co-crystal. Alternatively, the resolution step can be carried out at a previous stage, using any suitable intermediate.
Another aspect of the invention refers to a pharmaceutical composition which comprises a compound according to the invention as described above according to formula (I) or a pharmaceutically acceptable salt thereof, prodrug, solvate or steroisomer thereof, and a pharmaceutically acceptable carrier, adjuvant or vehicle. The present invention thus provides pharmaceutical compositions comprising a compound of this invention, or a pharmaceutically acceptable salt, prodrug, solvate or stereoisomers thereof together with a pharmaceutically acceptable carrier, adjuvant, or vehicle, for administration to a patient.
Examples of pharmaceutical compositions include any solid (tablets, pills, capsules, granules etc.) or liquid (solutions, suspensions or emulsions) composition for oral, topical or parenteral administration.
In a preferred embodiment the pharmaceutical compositions are in oral form, either solid or liquid. Suitable dose forms for oral administration may be tablets, capsules, syrops or solutions and may contain conventional excipients known in the art such as binding agents, for example syrup, acacia, gelatin, sorbitol, tragacanth, or polyvinylpyrrolidone; fillers, for example lactose, sugar, maize starch, calcium phosphate, sorbitol or glycine; tabletting lubricants, for example magnesium stearate; disintegrants, for example starch, polyvinylpyrrolidone, sodium starch glycollate or microcrystalline cellulose; or pharmaceutically acceptable wetting agents such as sodium lauryl sulfate.
The solid oral compositions may be prepared by conventional methods of blending, filling or tabletting. Repeated blending operations may be used to distribute the active agent throughout those compositions employing large quantities of fillers. Such operations are conventional in the art. The tablets may for example be prepared by wet or dry granulation and optionally coated according to methods well known in normal pharmaceutical practice, in particular with an enteric coating.
The pharmaceutical compositions may also be adapted for parenteral administration, such as sterile solutions, suspensions or lyophilized products in the apropriate unit dosage form. Adequate excipients can be used, such as bulking agents, buffering agents or surfactants.
The mentioned formulations will be prepared using standard methods such as those described or referred to in the Spanish and US Pharmacopoeias and similar reference texts.
Administration of the compounds or compositions of the present invention may be by any suitable method, such as intravenous infusion, oral preparations, and intraperitoneal and intravenous administration. Oral administration is preferred because of the convenience for the patient and the chronic character of the diseases to be treated.
Generally an effective administered amount of a compound of the invention will depend on the relative efficacy of the compound chosen, the severity of the disorder being treated and the weight of the sufferer. However, active compounds will typically be administered once or more times a day for example 1 , 2, 3 or 4 times daily, with typical total daily doses in the range of from 0.1 to 1000 mg/kg/day.
The compounds and compositions of this invention may be used with other drugs to provide a combination therapy. The other drugs may form part of the same composition, or be provided as a separate composition for administration at the same time or at different time.
Another aspect of the invention refers to a compound of formula (I) as described above, or a pharmaceutical acceptable salt or isomer thereof for use in therapy.
Another aspect of the invention refers to a compound of formula I, or a pharmaceutically acceptable salt or isomer thereof, for use in the treatment or prophylaxis of pain. Preferably, the pain is medium to severe pain, visceral pain, chronic pain, cancer pain, migraine, inflammatory pain, acute pain or neuropathic pain, allodynia or hyperalgesia. This may include mechanical allodynia or thermal hyperalgesia.
Another aspect of the invention refers to the use of a compound of the invention in the manufacture of a medicament for the treatment or prophylaxis of pain. In a preferred embodiment the pain is selected from medium to severe pain, visceral pain, chronic pain, cancer pain, migraine, inflammatory pain, acute pain or neuropathic pain, allodynia or hyperalgesia, also preferably including mechanical allodynia or thermal hyperalgesia.
Another aspect of this invention relates to a method of treating or preventing pain which method comprises administering to a patient in need of such a treatment or prevention a therapeutically effective amount of a compound as above defined or a pharmaceutical composition thereof. Among the pain syndromes that can be treated or prevented are medium to severe pain, visceral pain, chronic pain, cancer pain, migraine, inflammatory pain, acute pain or neuropathic pain, allodynia or hyperalgesia, whereas this could also include mechanical allodynia or thermal hyperalgesia.
The present invention is illustrated below with the aid of examples. These illustrations are given solely by way of example and do not limit the general spirit of the present invention.
EXAMPLES
The following abbreviations are used in the intermediates and examples: ACN: Acetonitrile
Anh: Anhydrous Aq: Aqueous
BOC2O: Di-tert-butyl dicarbonate tBuOH: tert-Butanol Cone: Concentrated DCE: Dichloroethane DCM: Dichloromethane
DDQ: 4,5-Dichloro-3,6-dioxocyclohexa-1 ,4-diene-1 ,2-dicarbonitrile DEA: Diethylamine
DIPEA: N,N-Diisopropylethylamine
DMA: Dimethylacetamide
DMAP: 4-Dimethylaminopyridine
DMF: Dimethylformamide EDC-HCI: N-(3-Dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride
EtOAc: Ethyl acetate Et20: Diethyl ether EtOH: Ethanol h: Hour/s HATU: (1-[Bis(dimethylamino)methylene]-1 H-1 ,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate)
HFIP: Hexafluoroisopropanol
HOBt: Hydroxybenzotriazole HPLC: High-performance liquid chromatography
HRMS: High-resolution mass spectrometry MeOH: Methanol MS: Mass spectrometry Min: Minutes
Quant: Quantitative Rt: Retention time rt: Room temperature Sat: Saturated TFA: T rifluoroacetic acid
THF: Tetrahydrofuran TEA: EtsN, Triethylamine TOSMIC: Toluenesulfonylmethyl isocyanide Wt: Weight
The following methods were used to generate the HPLC or HPLC-MS data:
Method A: Column Acquity UPLC BEH C18 2.1x50 mm, 1.7 mm; flow rate 0.61 mL/min; A: NH4HCO3 10 mM; B: ACN; Gradient: 0.3 min 98% A, 98% to 5% A in 2.52 min, isocratic 5%> A 1.02 min. Method B: Column Zodiac C18 50x4.6 mm 3 pm; flow rate 0.6 mL/min; A: 0.1 % formic acid in water; B: ACN; Gradient 95% to 50% A in 3 min, 50% to 5%> A in 2 min isocratic 5% A 2 min.
Method C: Column Acquity UPLC BEH C18 2.1x50 mm, 1.1 pm; flow rate 0.80 mL/min; A: NH4HCO3 10 mM; B: ACN; Gradient: 0.3 min 90% A, 90% to 5% A in 2.7 min, isocratic 5% A 0.7 min. Method D: Column Aquity UPLC BEH C18 2.1 x 50 mm, 1.7 pm, flow rate 0.61 mL/min; A: NH4HCO3 10 mM, B: ACN, C: MeOH + 0.1% formic acid; gradient 0.3 min 98% A, 98% A to 0:95:5 A:B:C in 2.7 min; 0:95:5 A:B:C to 100%o B in 0.1 min; isocratic 2 min 100% B. Method E: Column Acquity UPLC BEH C18 2.1x50 mm, 1.7 pm; flow rate 0.61 mL/min; A: NH4HCO3 10 mM pH 10.6; B: ACN; Gradient: 0.3 min 98%> A, 98%> to 0%> A in 2.7 min, isocratic 0%> A 2 min.
Method F: Column Acquity UPLC BEH C18 2.1x50 mm, 1.7 pm; flow rate 0.5 mL/min; A: NH4HCO3 10mM; B: ACN; Gradient: 90%> A to 5%> A in 4 min, 1 min in 5%> A, 5%> A to 90%> A in 0.1 min, 1.9 min in 90%> A
Method G: Column Acquity UPLC BEH C18 2.1x50 mm, 1.7 pm; flow rate 0.80 mL/min; A: ACONH4 10 mM; B: ACN; Gradient: 0.3 min 90%> A, 90%> to 5%> A in 2.7 min, isocratic 5%o A 0.7 min.
Synthesis of Intermediates
Figure imgf000085_0001
Intermediate 1. (1 r,4r)-4-Hydrazinyl-N,N-dimethyl-1-phenylcyclohexanamine
trihydrochloride.
Step a. 8-(Dimethylamino)-1 ,4-dioxaspiro[4.5]decane-8-carbonitrile.
To a stirred solution of fe/f-butyl 4-oxopiperidine-1-carboxylate (300 g, 1.51 mol) in EtOH (1200 ml.) and H2O (600 ml.) at rt, dimethylamine (40% Wt in H2O, 1700 mL, 15 mol) was added drop wise followed by the addition of KCN (244 g, 3.7 mol). The reaction mixture was stirred for 4 days at rt and extracted with DCM. The combined organic layers were washed with H2O and sat aq NaCI, dried over anh Na2S04, filtered and evaporated to dryness to give the title compound (350 g, Yield: 89%).
Step b. N,N-Dimethyl-8-phenyl-1 ,4-dioxaspiro[4.5]decan-8-amine. To a stirred solution of the compound obtained in step a) (95 g 0.45 mol) in THF (100 ml.) freshly prepared Grignard solution of bromophenyl (245.64 g, 1 .36 mol) was added dropwise at 0 °C. The reaction mixture was stirred at rt for 18 h and then quenched with sat aq NH4CI, extracted with EtOAc and washed with sat aq NaCI. The combined organic layers were dried over anh Na2S04, filtered and evaporated under reduced pressure. The crude product was purified by flash chromatography silica gel, gradient CHC o CHC iMeOH (9:1 ) to give the title compound (70 g, Yield: 59%).
Step c. 4-(Dimethylamino)-4-phenylcyclohexanone.
To a stirred solution of the compound obtained in step b) (70 g, 0.27 mol) in MeOH (50 ml_), cone. HCI (350 ml.) was added drop wise at 0 °C. The reaction mixture was stirred at rt for 16 h. The reaction was diluted with H2O and washed with EtOAc. The aqueous layer was basified with NaOH 8 N and the product was extracted with CHCI3. The combined organic layers were washed with H2O and sat aq NaCI and dried over anh Na2S04, filtered and evaporated to dryness to give the title compound (40 g, Yield: 68%).
Step d. tert-Butyl 2-(4-(dimethylamino)-4-phenylcyclohexyl)hydrazinecarboxylate.
To a stirred solution of the compound obtained in step c) (40 g, 0.184 mol) in MeOH (100 ml_), tert-butyl hydrazinecarboxylate (36.49 g, 276.49 mmol) in MeOH (400 ml.) and acetic acid (0.52 ml_, 9.22 mmol) was added at 0 °C. Then NaCNBHs (23.29 g, 0.37 mol) was added portion wise at 0 °C and the reaction mixture was stirred at rt for 16 h. The reaction mixture was evaporated and diluted with DCM, washed with H2O and sat aq NaCI. The combined organic layers were dried over anh Na2S04, filtered and evaporated under reduced pressure. The crude product was purified by flash chromatography, silica gel, gradient DCM to DCM:MeOH (95:5) to give the title compound (17.5 g, Yield: 28 % yield).
Step e. di-tert-Butyl 1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)hydrazine-1 ,2- dicarboxylate.
To a stirred solution of the compound obtained in step d) (17.5 g, 52.39 mmol) in anhydrous DCM (200 ml_), TEA (20.92 g, 157.17 mmol) and B0C2O (13.70 ml_, 62.86 mmol) were added at 0 °C and the reaction mixture was stirred for 16 h at rt. The reaction mixture was poured into ice cold H2O and extracted with DCM. The combined organic layer were dried over anh Na2S04, filtered and evaporated under reduced pressure. The crude product was purified by flash chromatography, silica gel, gradient Hexane to Hexane:EtOAc (7:3) to give the title compound (6.0 g, Yield: 26%).
Step f. Title compound.
To a stirred solution of the compound obtained in step e) (2.5 g, 5.96 mmol), in MeOH (25 ml_), cone. HCI (1.08 g, 29.83 mmol) was added dropwise at 0 °C. The reaction mixture was allowed to stir at rt for 16 h. The mixture was evaporated, co-distilled with toluene (3x 50 ml.) and dried under vacuum to give the title compound. (1.75 g, Yield: 89%).
Figure imgf000087_0001
Intemediate 2. (1 s,4s)-4-(Dimethylamino)-4-phenylcyclohexyl 4- methylbenzenesulfonate.
Step a. 4-(Dimethylamino)-4-phenylcyclohexanol.
To a solution of the compound obtained in step c) of Intermediate 1 (7.5 g, 34.6 mmol) in MeOH (75 ml_), sodium borohydride (3.9 g, 9.1 mmol) was added portion wise at 0 °C. The reaction mixture was stirred at rt for 2 h. Sat aq NH4CI was added dropwise at 0 °C and the product was extracted with DCM. The combined organic layers were dried over anh Na2S04, filtered and evaporated under vacuum to give the title compound (6.5 g, Yield: 86 %). Step b. Title compound.
To a solution of the compound obtained in step a) (6.5 g; 29.7 mmol) in anh DCM (100 ml_), TEA (8.4 ml_, 60.3 mmol) was added dropwise at rt and the reaction mixture was cooled to 0 °C. Then DMAP (0.2 g, 1.6 mmol) and p-toluensulfonyl chloride (6.22 g, 32.6 mmol) were added at 0 °C and the reaction mixture was stirred at rt for 48 h. The reaction mixture was neutralized with sat aq NaHCCh and the product was extracted with DCM. The combined organic layers were washed with H2O and sat aq NaCI, dried over anh Na2S04, filtered and evaporated under vacuum. The crude product was purified by flash chromatography silica gel, gradient petroleum ether to petroleum ethenEtOAc (70:30), to give the title compound (3.4 g, Yield: 31 %).
Figure imgf000088_0001
Intemediate 3. ((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)methyl 4- methylbenzenesulfonate.
Step a. (1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexanecarbonitrile.
To a solution of the compound obtained in step c) of Intermediate 1 (30 g, 138.2 mmol) in anhydrous THF (1200 mL), TOSMIC (40.4 g, 207.4 mmol) in THF (900 ml.) was added drop wise at 0 °C. Then, MeOH (8.84 g, 276.4 mmol) and KO‘Bu (46.5 g, 414.7 mmol) were added at 0 °C. The reaction mixture was stirred at 0 °C for 45 min and at rt for 16 h. The mixture was extracted with EtOAc, washed with FhO and sat aqueous NaCI. The combined organic layers were dried over anh Na2S04, filtered and evaporated under vacuum. The crude product was purified by flash chromatography neutral alumina, gradient petroleum ether to petroleum ethenEtOAc (96:4), to give the title compound (1 r,4r, 7.5 g, Yield: 24%) and its (1 s,4s)-isomer (7.0g, Yield: 22%).
Step b. (1 r,4r)-Methyl 4-(dimethylamino)-4-phenylcyclohexanecarboxylate.
To a solution of the compound obtained in step a) (3 g, 13.1 mmol) in MeOH (30 mL), TMSCI (33.6 ml, 263.1 mmol) was added at rt. The reaction mixture was stirred at rt for 20 h. Then sat aq NaHCOs was added dropwise at 0 °C, and the product was extracted with DCM and washed with sat aq NaCI (1x 10 mL). The organic layer was dried over any Na2S04, filtered and evaporated under reduced pressure to give the title compound (2.9 g, Yield: 85%). Step c. ((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)methanol. To a solution of the compound obtained in step b) (3 g, 1 1.5 mmol) in THF (50 ml_), LiAII-U (655 mg, 17.2 mmol) was added portion wise at 0 °C and the mixture was stirred at rt for 3 h. The reaction mixture was diluted with EtOAc (60 ml.) and then sat aq Na2S04 was added dropwise at 0 °C. The organic layer was washed with sat aq NaCI and dried over anh Na2S04, filtered, and evaporated under vacuum to give the title compound (2.5 g, Yield: 93%).
Step d. Title compound.
To a stirred solution of the compound obtained in step c) (2.5 g, 10.7 mmol) in anhydrous DCM (30 ml_), TEA (4.5 ml_, 32.2 mmol) and DMAP (130 mg, 1.1 mmol) were added at 0 °C. Then p-toluensulfonyl chloride (2.45 g, 12.9 mmol) was added dropwise at 0 °C, and the reaction mixture was stirred at rt for 4 h. The reaction mixture was quenched with H2O (30 ml.) and extracted with DCM. . The organic layer was washed with sat aq NaCI, dried over anh Na2S04, filtered and evaporated under vacuum. The crude product was purified by flash chromatography silica gel, gradient DCM to DCM:MeOH (95:5), to give the title compound (2.3 g, Yield: 56%).
Figure imgf000089_0001
Intermediate 4. 2-(4-(Dimethylamino)-4-phenylcyclohexyl)ethyl 4- methylbenzenesulfonate.
Step a. Methyl 2-(4-(dimethylamino)-4-phenylcyclohexylidene)acetate. To a stirred solution of 4-(dimethylamino)-4-phenylcyclohexanone (500 mg, 2.3 mmol) in DMF (12 ml_), methyl 2-(dimethoxyphosphoryl)acetate (628.5 mg, 3.4 mmol) and potassium 2-methylpropan-2-olate (387.3 mg, 3.4 mmol) were added and the mixture was heated at 60 °C for 4 h. Some ice was added and the product was extracted with Et2<D. The organic layer were combined and dried over anh Na2S04 and the solvent was removed under vacuum to give the title compound (617.4 mg, Yield: 96%).
Step b. 2-(4-(Dimethylamino)-4-phenylcyclohexylidene)ethanol. To a stirred solution of LiAlhU (1 M in Et20, 4.78 mL, 4.8 mmol) cooled at -70 0 C, the compound obtained in step a) (261.1 mg, 0.95 mmol) in Et20 (6 mL) was added and the mixture was stirred at -70 °C for 2 h. The reaction was quenched with ice and the product was extracted with EtOAc and Et20. The combined organic layers were dried over anh Na2S04, and the solvent was removed under vacuum to give the title compound (211.5 mg, Yield: 90%).
Step c. 2-(4-(Dimethylamino)-4-phenylcyclohexyl)ethanol.
A solution of the compound obtained in step b) in MeOH (10 mL), was reacted in a continuous flow hydrogenation reactor (Thales Nano H-Cube) using a catalyst cartridge (Pd/C 5%, 30 mm, 25 °C, 30 bars of H2, 1 mL/ min flow rate). The collected fractions were evaporated under reduced pressure to give the title compound (1 11.5 mg, Yield: 89%).
Step d. Title compound.
A solution of the compound obtained in step c) (11 1.5 mg, 0.45 mmol) in anhDCM (6 mL), TEA (90 mί, 0.63 mmol) and DMAP (3.6 mg, 0.03 mmol) were added and the mixture was cooled at 0 °C. p-Toluenesulfonyl chloride (103.1 mg, 0.54 mmol) was added and reaction mixture was allowed to reach rt and was stirred for 4 h. The reaction was quenched with water and the product was extracted with DCM. The combined organic layers were dried over anh Na2S04, and the solvent was removed under vacuum, to give the title compound (107mg, Yield: 59%).
Synthesis of Examples
Figure imgf000090_0001
Example 1. (1 -((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone. Step a. Ethyl 2-oxo-2-(4-oxo-4,5,6,7-tetrahydrobenzo[b]thiophen-5-yl)acetate. To a solution of 6,7-dihydrobenzo[b]thiophen-4(5H)-one (3 g, 19.71 mmol) in anh THF (60 ml.) under argon atmosphere, lithium bis(trimethylsilyl)amide (1 M in THF, 29.56 ml_, 29.56 mmol) was added dropwise at rt. Then diethyl oxalate (4.28 ml_, 31.53 mmol) was added and the reaction mixture was stirred at rt overnight. The reaction crude was diluted with EtOAc and washed with H2O. The aqueous layer was acidified to pH:2 with 2 N HCI and extracted with EtOAc. The combined organic layers were dried over anh Na2S04 and concentrated to dryness to give the title compound (4.40 g, Yield: 89%).
Step b. Ethyl 1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazole-3-carboxylate.
To a solution of the compound obtained in step a) (1 .1 g, 4.36 mmol) in EtOH (35 ml.) under argon atmosphere, Intermediate 1 (1 .12 g, 3.54 mmol) was added portion wise and the reaction was heated at 65 °C for 2.5 h. The mixture was cooled at room temperature and a solid precipitated. The solid was filtered and washed with EtOH to give the title compound (1.59 g, Yield: 96%).
Step c. 1 -((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazole-3-carboxylic acid.
To a solution of the compound obtained in step b) (1.5 g, 3.4 mmol) in EtOH (15 ml.) under argon atmosphere, NaOH (680 mg, 17 mmol) was added and the reaction was heated at 85 °C for 3 h. The reaction was quenched with HCI (2 M in Et20, 8.5 ml.) and solvents were evaporated under vacuum. The crude product was dissolved in DCM:water (1 :1 , 40 ml.) and the remaining solid was filtered, washed with water and dried under vacuum, to give the title compound (1.44 g, Yield: 85%).
Step d. Title compound.
To a solution of the compound obtained in step c) (1 .23 g, 2.9 mmol) in anh DCM:DMF (1 :1 , 10 ml.) under argon atmosphere, EDC-HCI (1 .1 14 g, 5.81 mmol), HOBt (890.01 mg, 5.81 mmol) and TEA (1.21 ml_, 8.72 mmol) were added and the reaction was stirred at rt for 10 min. Then 1 -methylpiperazine (0.483 ml_, 4.36 mmol) was added dropwise and the reaction was stirred at rt overnight. Solvents were evaporated under vacuum and the crude product was dissolved in DCM and washed with H2O. The organic layer was dried over anh Na2S04, filtered and concentrated to dryness to give the title compound (1 .035 g, Yield: 71 %).
HPLC-MS (D): Rt, 1.93 min; ESI+-MS m/z: 504.2 This method was used for the preparation of Examples 2-76 using suitable starting materials and intermediates.
Examples X:
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Example 77. (1 -((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-1 H-thieno[2,3-g]indazol- 3-yl)(4-methylpiperazin-1 -yl)methanone. To a solution of the compound obtained in Example 1 (150 mg, 0.3 mmol) in anhydrous dioxane (5 ml.) under argon atmosphere, DDQ (135.2 mg, 0.6 mmol) was added and the reaction mixture was heated at 100 °C for 1 h. More DDQ was added (67.6 mg, 0.3 mmol) and the reaction mixture was heated at 100 °C for 1 .5 h. After cooling to rt the solvent was removed under vacuum. EtOAc was added and the organic layer was washed with Na2C03 and brine, dried over anh Na2S04 and the solvent was removed. The crude product was purified by flash chromatography silica gel, gradient DCM to MeOH, to give the title compound (8 mg, Yield: 5%).
HPLC-MS (D): Rt, 1.99 min; ESI+-MS m/z: (502.3)
Figure imgf000103_0002
Example 78. (7-Bromo-1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone.
Step a. 2-bromo-6,7-dihydrobenzo[b]thiophen-4(5H)-one.
To a solution of 6,7-dihydrobenzo[b]thiophen-4(5H)-one (1.5 g, 9.85 mmol) in AcOH: H2O (20 ml_, 1 :1 ), bromine (1.57 g, 9.85 mmol) was added dropwise at 0 °C. After 20 min, the reaction mixture was warmed at rt and stirred for 18 h. The reaction was quenched at 0 °C by addition of 1 M NaOH until basic pH. The precipitated formed was collected by filtration, washed with water and dried in a vacuum oven at 50 °C to give the title compound (2.08 g, Yield: 91%). Step b. Ethyl 2-(2-bromo-4-oxo-4,5,6,7-tetrahydrobenzo[b]thiophen-5-yl)-2-oxoacetate.
Starting from the compound obtained in step a) (300 mg, 1.29 mmol) and following the experimental procedure described in step a) of Example 1 , the title compound was obtained (425.8 mg, Yield: quantitative).
Step c. Ethyl 7-bromo-1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazole-3-carboxylate.
Starting from the compound obtained in step b) (425.8 mg, 1.29 mmol) and following the experimental procedure described in step b) of Example 1 , the title compound was obtained (607.0 mg, Yield: 89 %).
Step d. 7-Bromo-1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazole-3-carboxylic acid.
Starting from the compound obtained in step c) (607.0 mg, 1.15 mmol) and following the experimental procedure described in step c) of Example 1 , the title compound was obtained (574.7 mg, Yield: quantitative).
Step e. Title compound.
Starting from the compound obtained in step d) (291.4 mg, 0.582 mmols) and following the experimental procedure described in step d) of Example 1 , the title compound was obtained (309.0 mg, Yield: 91%).
HPLC-MS (D): Rt, 2.28 min; ESI+-MS m/z: 582.1 )
Figure imgf000105_0001
Example 79. 1 -((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-3-(4-methylpiperazine-1 - carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazole-7-carbonitrile.
To a solution of the compound obtained in Example 78 (50 mg, 0.09 mmol) in anhydrous DMF (1 ml.) under argon atmosphere, Zn(CN)2 (10.1 mg, 0.09 mmol) and Pd(PPh3)4 (9.92 mg, 0.02 mmol) were added and the reaction mixture was heated under microwave irradiation (100 °C, 150 W, 45 min). The reaction was quenched with 2 N HCI and extracted with EtOAc. The organic layer was dried with anh Na2S04 and the solvent was removed under vacuum. The crude product was purified by flash chromatography silica gel, gradient DCM to MeOH, to give the title compound (10 mg, Yield: 22%).
HPLC-MS (D): Rt, 2.28 min; ESI+-MS m/z: 470.2 (M-45)
Figure imgf000105_0002
Example 80. 1-((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-3-(4-methylpiperazine-1- carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazole-7-carboxylic acid.
To a solution of the compound obtained in Example 79 (27 mg, 0.05 mmol) in EtOH (0.5 ml_), NaOH (2.5 M, 0.41 ml_, 1.02 mmol) and H2O2 (30%, 78 mI_, 0.77 mmol) were added and the mixture was stirred at 90 °C for 16 h. The solvent was removed under vacuum and the crude product was purified by flash chromatography C18, gradient NH4HCC>3 (10 mM) to ACN to give the title compound (6 mg, Yield: 22%). HPLC-MS (D): Rt, 1.22 min; ESI+-MS m/z: 548.2
Figure imgf000106_0001
Example 81. 1-((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-3-(4-methylpiperazine-1- carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazole-7-sulfonic acid.
To a solution of the compound obtained in Example 1 (62.5 mg, 0.12 mmol) in anhydrous DCM (2 ml_), sulphuric acid (48.7 mg, 0.5 mmol) and acetic anhydride (152 mg, 1.5 mmol) were added drop wise at -10 °C. The mixture was stirred at rt for 16 h. The solvent was removed under vacuum and the crude product was purified by flash chromatography, silica gel gradient DCM to DCM:MeOH (85:15), to give the title compound (68.3 mg, Yield: 94%).
HPLC-MS (D): Rt, 1.93 min; ESI+-MS m/z: 504.2
Figure imgf000106_0002
Example 82. (1-((1 r,4r)-4-((2-Fluoroethyl)methylamino)-4-phenylcyclohexyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone.
A solution of the compound obtained in Example 18 (60 mg, 0.123 mmol) in ACN (5 ml_), 2-methoxyethyl trifluoromethanesulfonate (255.0 mg, 1.23 mmol) and TEA (37.2 mg, 0.368 mmol) were added and the reaction mixture was heated at 100 °C overnight. The solvent was removed under vacuum and EtOAc was added. The organic layer was washed with NaHCC>3, dried over anh Na2S04 and filtered. The solvent was removed to dryness and the crude product was purified by flash chromatography, gradient DCM to MeOH (1% NH3), to give the title compound (2 mg, Yield: 3%).
HPLC-MS (D): Rt, 2.26 min; ESI+-MS m/z: 536.3
This method was used for the preparation of Example 83 using suitable starting materials:
Figure imgf000107_0002
Figure imgf000107_0001
Example 84. (1 -((1 r,4r)-4-((2,2-Difluoroethyl)(methyl)amino)-4-phenylcyclohexyl)-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1 -yl)methanone.
Starting from the compound obtained in Example 83 (86.2 mg, 0.155 mmol) and following the experimental procedure described in Example 77 the title compound was obtained (6 mg, Yield: 7%).
HPLC-MS (D): Rt, 2.46 min; ESI+-MS m/z: 552.3
Figure imgf000108_0001
Example 85. (1 ,4-Diazepan-1 -yl)(1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)methanone.
To a solution of the compound obtained in Example 4 (200 mg, 0.33 mmol) in anh DCM (2.7 ml_), TFA (345 mI_) was added drop wise at 0 °C and the reaction mixture was stirred overnight at rt. The mixture was neutralised with 20% aq NaOH, diluted with DCM and washed with sat aq NaCI. The organic layer was dried over anh Na2S04, filtered and evaporated to dryness, to give the title compound (45 mg, Yield: 27%).
HPLC-MS (D): Rt, 1.77 min; ESI+-MS m/z: 504.2 This method was used for the preparation of Examples 86-96 using suitable starting materials:
Figure imgf000108_0002
Figure imgf000109_0002
Figure imgf000109_0001
Example 97. (1 -((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-(2-fluoroethyl)piperazin-1-yl)methanone.
A sealed tube was charged with a solution of the compound obtained in Example 86 (40 mg, 0.082 mmol) in anh ACN (2 ml_). DIPEA (20.9 mI_, 0.12 mmol) and 1-fluoro-2- iodoethane (22.9 mI_, 0.1 mmol) were added drop wise and the reaction mixture was irradiated under microwaves (120 °C, 150 W, 1 h). The reaction mixture was diluted with DCM and washed with H2O. The organic layer was dried over anh Na2S04, filtered and evaporated under vacuum. The crude product was purified by flash chromatography silica gel, gradient DCM to DCM:MeOH (98:2), to give the title compound (20 mg, Yield: 45%).
HPLC-MS (D): Rt, 2.08 min; ESI+-MS m/z: 536.3
This method was used for the preparation of Example 98 using suitable starting materials:
Figure imgf000110_0002
Figure imgf000110_0001
Example 99. (2-((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-2H- thieno[2,3-g]indazol-3-yl)(4-ethylpiperazin-1-yl)methanone.
Step a. Ethyl 2-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-2H- thieno[3,2-e]isoindole-3-carboxylate. To a solution of the compound obtained in step b) of Example 1 (320 mg, 1.16 mmol) in DMA (1.5 ml.) under argon atmosphere, Intermediate 2 (519 mg, 1.392 mmol) and CS2CO3 (567 mg, 1.74 mmol) were added and the reaction was heated at 100 °C for 2 h. The crude mixture was diluted with EtOAc and washed with H2O. The organic layer was dried over anh Na2S04, filtered and concentrated to dryness. The crude product was purified by flash chromatography silica gel, gradient DCM to DCM:MeOH (8:2), to give the title compound (80.4 mg, Yield: 15%).
Step b. 2-((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-2H-thieno[3,2- e]isoindole-3-carboxylic acid.
Starting from the product obtained in step a) (80 mg, 0.179 mmol) and following the experimental procedure described in step c) of Example 1 , the title compound was obtained (55 mg, Yield: 72%).
Step c. Title compound.
Starting from the product obtained in step b) (55 mg, 0.128 mmol) and following the experimental procedure described in step d) of Example 1 , the title compound was obtained (57 mg, Yield: 87%).
HPLC-MS (E): Rt, 2.66 min; ESI+-MS m/z: 473.2
This method was used for the preparation of Examples 100-101 using suitable starting materials:
Figure imgf000111_0001
Figure imgf000112_0002
Figure imgf000112_0001
Example 102. 2-(1 -((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)-2-(4-methylpiperazin-1-yl)acetonitrile. Step a. 1-((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-N-methoxy-N-methyl-4,5- dihydro-1 H-thieno[2,3-g]indazole-3-carboxamide.
Starting from the compound obtained in step c) of Example 1 (180 mg, 0.427 mmols) and following the experimental procedure described in step d) of the same example, the title compound was obtained (172.63 mg, Yield: 87%). Step b. 1-((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazole-3-carbaldehyde.
To a solution of the compound obtained in step a) (66 mg, 0.142 mmols) in anh THF (2ml_) cooled at 0 °C, LiAII-U (1 M in THF, 0.17 ml_, 0.17 mmols) was added drop wise and the mixture was stirred for 30 min. The reaction was quenched with 10 % HCI and the mixture was neutralized with aq NaHCCh sat solution and extracted with EtOAc. The organic layer was dried over anh Na2S04, filtered and concentrated to dryness, to give the title compound (53.6 mg, Yield: 93%).
Step c. Title compound. To a solution of the compound obtained in step b) (73 mg, 0.18 mmol) in hhCbMeOH (1 :1 , 8 ml_), Na2S2C>3 (17.1 mg, 0.09 mmol) was added and the mixture was stirred vigorously at rt for 2 h. Methylpiperazine (18.02 mg, 0.18 mmol) was added, the mixture was stirred for 1 h and then KCN (23.4 mg, 0.36 mmol) was added and the mixture was stirred for 48 h more. MeOH was removed under vacuum and the crude product was extracted with EtOAc, washed with sat NaCI and water. The organic layer was dried over anh Na2S04, filtered and concentrated to dryness to obtain the title compound (77.0 mg, Yield: 93%).
HPLC-MS (D): Rt, 2.28 min; ESI+-MS m/z: 470.2 (M-45)
Figure imgf000113_0001
Example 103. 2-(1 -((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)-2-(4-methylpiperazin-1-yl)acetamide.
To a solution of the compound obtained in Example 102 (48 mg, 0.093 mmol) in MeOH/DMSO (3:1.5, 4.5 ml_), NaOH (2.5 M, 74 mI_, 0.187 mmol) and H202 (30%, 48 mI_, 0.466 mmol) were added and the mixture was stirred at rt for 30 min. Solvents were removed under vacuum and the residue was extracted with EtOAc). The organic layer was washed with H2O, dried over anh Na2S04 and filtered. The solvent was removed under vacuum to give the title compound (49.62 mg, Yield: quantitative).
HPLC-MS (D): Rt, 1.7 min; ESI+-MS m/z: 533.2
Figure imgf000113_0002
Example 104. (1 r,4r)-4-(8-(tert-Butyl)-3-((4-methylpiperazin-1 -yl)methyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-1-yl)-N,N-dimethyl-1-phenylcyclohexanamine. To a stirred solution of the compound obtained in Example 103 (48.0 mg, 0.090 mmol) in HhOdBuOH (1.6 ml_, 1 :1 ), TFA (0.689 ml_, 9.01 mmol) was added and the mixture was heated at 100 °C for 6 days. The solvent was removed under vacuum, EtOAc (5 ml.) was added and the organic layer was washed with NaHCC>3 aq sat sol. The organic layer was dried over anh Na2S04 and the solvent was removed under vacuum. The crude product was purified by flash chromatography, silica gel, gradient DCM to DCM:MeOH (30:70), to give the title compound. (10 mg, Yield: 20%).
HPLC-MS (D): Rt, 2.75 min; ESI+-MS m/z: 546.2
Figure imgf000114_0001
Example 105. N,N-Dimethyl-1-(4-(3-((4-methylpiperazin-1-yl)methyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-1-yl)phenyl)-4-phenylpiperidin-4-amine.
Step a. Aluminium trihydride.
A vigorously stirred solution of LiAIH4 (1 M in THF, 25 ml_, 25 mmol) was cooled at -78 °C. Sulfuric acid (666.3 mI; 12.5 mmol) was added drop wise and the mixture was stirred at -78 °C for 1.5 h. The mixture was used in the next step.
Step b. Title compound.
To a stirred solution of freshly prepared alane in step a) (1.7 ml_, 1.7 mmols), Example 38 (195.5 mg, 0.337 mmols) was added at 0 °C and the mixture was stirred at 0 °C for 1.5 h. The reaction mixture was quenched by drop wise addition of water (5ml_) and the mixture was extracted with EtOAc. The combined organic layers were dried over anh Na2S04, filtered and evaporated under reduced pressure. The crude product was purified by flash chromatography AI2O3, using gradient DCM to DCM:MeOH (7:3), to give the title compound (108 mg, Yield: 57%). HPLC-MS (A): Rt, 2.41 min; ESI+-MS m/z: 567.3
This method was used for the preparation of Example using suitable starting materials:
Figure imgf000115_0002
Figure imgf000115_0001
Example. 107 (1-((4-(Dimethylamino)-4-phenylcyclohexyl)methyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone.
Step a. Ethyl 4,5-dihydro-1 H-thieno[2,3-g]indazole-3-carboxylate.
To a solution of the compound obtained in step a) of Example 1 (1.0 g, 3.96 mmol) in EtOH (15 ml.) under argon atmosphere, hydrazine hydrate (218.27 mg, 4.36 mmol) was added drop wise. Following the experimental procedure described in step c) of Example 1 , the title compound was obtained (955.2 mg, Yield: 97%).
Step b. 4,5-Dihydro-1 H-thieno[2,3-g]indazole-3-carboxylic acid.
Starting from the product obtained in step a) (955.2 mg, 3,85 mmol) and following the experimental procedure described in step c) of Example 1 , the title compound was obtained (727 mg, Yield: 86%).
Step c. (4,5-Dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone. Starting from the product obtained in step b) (167 mg, 0.67 mmol) and following the experimental procedure described in step d) of Example 1 , the title compound was obtained (126 mg, Yield: 63%).
Step d. Title compound.
To a solution of the compound obtained in step c) (60 mg, 0.2 mmol) in DMA (2 ml_), Intermediate 2 (99.2 mg, 0.24 mmol) and CS2CO3 (97 mg, 0.3 mmol) were added, and the mixture was heated at 100 °C for 2 h. The solvent was removed under vacuum and EtOAc was added. The organic layer was washed with water and brine and dried over anh Na2S04and the solvent was removed under vacuum. The crude product was purified by flash chromatography S1O2, using gradient DCM to MeOH, to give the title compound (31 mg, Yield: 30%).
HPLC-MS (C): Rt, 2.33 min; ESI+-MS m/z: 518.1
Figure imgf000116_0001
Example 108. (1 -(2-((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)ethyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1 -yl)methanone.
Alkylation of the product obtained in step c) of Example 106 (84 mg, 0.278 mmol) with intermediate 4 and following the experimental procedure described in step d) of the same example, provided the title compound (38.1 mg, Yield: 26%).
HPLC-MS (D): Rt, 1.97 min; ESI+-MS m/z: 532.3.
Figure imgf000116_0002
Example 109. (1 -(2-(4-(3-Hydroxyphenyl)-4-(methylamino)piperidin-1 -yl)ethyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1 -yl)methanone.
Step a. Ethyl 1 -(2-hydroxyethyl)-4,5-dihydro-1 H-thieno[2,3-g]indazole-3-carboxylate.
To a solution of the compound obtained in step a) of Example 1 (8 g, 32.89 mmol) in HFIP (55 ml.) under argon atmosphere, 2-hydrazinylethanol (3.34 ml_, 49.35 mmol) was added drop wise and the reaction was stirred at rt for 1 .5 h. The solid product was filtered, washed with HFIP and dried under vacuum to give the title compound (8 g, Yield: 83%).
Step b. 1 -(2-Hydroxyethyl)-4,5-dihydro-1 H-thieno[2,3-g]indazole-3-carboxylic acid.
Starting from the product obtained in step a) (5.87 g, 20.08 mmol) and following the experimental procedure described in step c) of Example 1 , the title compound was obtained (4.35 g, Yield: 82%).
Step c. (1 -(2-Hydroxyethyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin- 1 -yl)methanone.
Starting from the product obtained in step b) (1.5 g, 5.67 mmol) and following the experimental procedure described in step d) of Example 1 , the title compound was obtained (830 mg, Yield: 42%).
Step d. (1 -(2-Chloroethyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin- 1 -yl)methanone.
To a solution of the compound obtained in step c) (155 mg, 0.447 mmol) in anh DCM (25 ml.) under argon atmosphere, thionyl chloride (99.8 mI_, 1 .34 mmol) was added dropwise at 0 °C and the reaction mixture was stirred at rt overnight. The crude mixture was diluted with DCM and washed with H2O. The organic layer was dried over anh Na2S04, filtered and concentrated to dryness to give the title compound (152 mg, Yield: 93%).
Step e. Title compound.
A sealed tube was charged with the compound obtained in step d) (80 mg, 0.219 mmol), DMF (2 ml_), DIPEA (190 mI_, 1 .096 mmol), Kl (36.9 mg, 0.219 mmol) and 3-(4- (methylamino)piperidin-4-yl)phenol (76.9 mg, 0.373 mmol). The reaction mixture was irradiated under microwaves at 150 W for 2 h at 100 °C. EtOAc was added and the mixture was washed three times with H2O. The organic layer was dried over Na2S04 filtered and evaporated under vacuum. The crude product was purified by flash chromatography silica gel, gradient DCM to DCM:MeOH (85:15), to give the title compound (33 mg, Yield: 28%).
HPLC-MS (D): Rt, 1.53 min; ESI+-MS m/z: 535.1
This method was used for the preparation of Examples 109-1 16 using suitable starting materials:
Figure imgf000118_0001
Figure imgf000119_0002
Figure imgf000119_0001
Example 118. 1 -(4-(Dimethylamino)-4-phenylpiperidin-1 -yl)-2-(3-(4-methylpiperazine-1 - carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1 -yl)ethanone.
Step a. 2-Chloro-1-(4-(dimethylamino)-4-phenylpiperidin-1-yl)ethanone.
To a solution of N,N-dimethyl-4-phenylpiperidin-4-amine (491 mg, 1.771 mmol) in anh DCM (30 ml_), 2-chloroacetyl chloride (200 mg, 1.771 mmol) and TEA (0.44 ml_, 3.19 mmol) were added at 0 °C. The mixture was stirred for 3 h and the solvent was removed under vacuum. The crude product was purified by flash chromatography AI2O3, gradient cyclohexane to AcOEt, to give the title compound (256.3 mg, Yield: 62%).
Step b. Title compound.
To a solution of the compound obtained in step a) in DMA (15 ml_), the product obtained in step c) (147.0 mg, 0.486 mmol) of Example 106 was added. Following the experimental procedure described in step d) of the same example, the title compound was obtained (60.5 mg, Yield: 23%).
HPLC-MS (D): Rt, 1.81 min; ESI+-MS m/z: 547.3
This method was used for the preparation of Examples 117-127 using suitable starting materials:
Figure imgf000120_0001
Figure imgf000121_0001
Following the experimental procedure described in Example 1 , and using suitable starting materials, the compounds of Examples 128-140 were prepared.
Figure imgf000121_0002
Figure imgf000122_0001
Following the experimental procedure described in Example 85 and using suitable starting materials, the compounds of Examples 141-146 were prepared.
Figure imgf000123_0001
Following the experimental procedure described in Example 118 and using suitable starting materials, the compounds of Examples 147-148 were prepared.
Figure imgf000124_0001
Following the experimental procedure described in Example 109 and using suitable starting materials, the compounds of Examples 149-150 were prepared.
Figure imgf000124_0002
Figure imgf000125_0001
Example 151. (1 -((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(3-(dimethylamino)azetidin-1 -yl)methanone.
A sealed MW tube was charged with Example 142 (30 mg, 0.061 mmol) dissolved in DCE (2 ml_), formaldehyde (17 mI, 0.613 mmol) and NaBH(OAc)3 (39 mg, 0.184mmol) were added and the reaction mixture was irradiated at MW at 150 W for 5 min at 120 °C. The reaction mixture was dissolved in DCM and washed three times with H2O. The organic layer was dried over Na2S04 filtered and evaporated under vacuum to give the title compound (32 mg, Yield: quant).
HPLC-MS (D): Rt, 2.03 min; ESI+-MS m/z: 549.3.
Figure imgf000125_0002
Example 152. 1 -(4-(Methylamino)-4-phenylpiperidin-1 -yl)-2-(3-(4-methylpiperazine- 1 -carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1 -yl)ethanone.
Following the experimental procedure described in Example 77, and using suitable starting materials, the title compound was prepared.
HPLC-MS (G): Rt, 2.23 min; ESI+-MS m/z: 532.7.
PHARMACOLOGICAL STUDY
Human a2d-1 subunit of Cav2.2 calcium channel assay
Human a,2d-1 enriched membranes (2.5 pg) were incubated with 15 nM of radiolabeled [3H]-Gabapentin in assay buffer containing Hepes-KOH 10mM, pH 7.4. NSB (non specific binding) was measured by adding 10 mM pregabalin. The binding of the test compound was measured at five different concentrations. After 60 min incubation at 27 °C, binding reaction was terminated by filtering through Multiscreen GF/C (Millipore) presoaked in 0.5 % polyethyleneimine in Vacuum Manifold Station, followed by 3 washes with ice-cold filtration buffer containing 50 mM Tris-HCI, pH 7.4. Filter plates were dried at 60 °C for 1 h and scintillation cocktail (30 mI_) were added to each well before radioactivity reading. Readings were performed in a Trilux 1450 Microbeta radioactive counter (Perkin Elmer).
Human u-opioid receptor radioligand assay
Transfected CHO-K1 cell membranes (20 pg) were incubated with [3H]-DAMGO (1 nM) in assay buffer containing Tris-HCI 50 mM, MgCI2 5 mM at pH 7.4. NBS (non-specific binding) was measured by adding 10 mM Naloxone. The binding of the test compound was measured at five different concentrations. Plates were incubated at 27 °C for 60 min. After the incubation period, the reaction mixture was then transferred to Multiscreen HTS, FC plates (Millipore), filtered and plates were washed 3 times with ice-cold 10 mM Tris-HCI (pH 7.4). Filters were dried and counted at approximately 40% efficiency in a MicroBeta scintillation counter (Perkin-Elmer) using EcoScint liquid scintillation cocktail.
Results:
This invention is aimed at providing a series of compounds which act as dual ligands of the a2d subunit of voltage-gated calcium channels and the m-opioid receptor it is a very preferred embodiment in which the compounds are selected which act as dual ligands of the a2d subunit of voltage-gated calcium channels and the m-opioid receptor and especially compounds which have a binding expressed as K, responding to the following scales:
Kί(a2d-1 ) is preferably < 10000 nM, more preferably < 5000 nM, or even more preferably < 500 nM.
K,(m) is preferably < 1000 nM, more preferably < 500 nM, even more preferably < 100 nM.
The following scale has been adopted for representing the binding to the a2d-1 subunit of voltage-gated calcium channels expressed as K,:
+ Kΐ(a2d-1) >= 5000 nM ++ 500nM <= Kί(a2d-1) <5000 nM
+++ Kί(a2d-1) <500 nM
The following scale has been adopted for representing the binding to m-opioid receptor expressed as K,: + Ki (m) >= 500 nM
++ 100 nM <= K,(m) < 500 nM
+++ Kΐ(m) < 100 nM
All compounds prepared in the present application exhibit binding to the a2d-1 subunit of voltage-gated calcium channels and the m-opioid receptor, in particular the following binding results are shown:
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001

Claims

1. A compound of formula (I):
Figure imgf000131_0001
wherein one of W3 and W4 is S and the other is C;
Y is -C(O)-, -[C(RaRa')]n-, -CRa(CN)- or -CRa(CONRa'Ra ")-. wherein each Ra, Ra· and Ra” are independently selected from hydrogen, halogen, substituted or unsubstituted Ci-6 alkyl, substituted or unsubstituted C2-6 alkenyl, and substituted or unsubstituted C2-6 alkynyl; and n is 0, 1 , 2 or 3;
X is selected from a bond, -[C(RbRb )] -, -[C(RbRb )] C(0)[C(Rb"Rb )]q-> substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl-C(O), substituted or unsubstituted aryl- alkyl, wherein each Rb, Rb·, Rb- and Rb- are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl, and substituted or unsubstituted C2-6 alkynyl; and p is 0, 1 , 2 or 3; q is 0, 1 , 2 or 3;
Group A is a substituted or unsubstituted 4 to 10 membered mono or bicyclic heterocyclyl containing one nitrogen atom and optionally a second heteroatom selected from N and O, wherein A is attached to Y through a nitrogen atom; each Ri and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted Ci-6 alkyl, substituted or unsubstituted C2-6 alkenyl, substituted or unsubstituted C2-6 alkynyl, substituted or unsubstituted alkyl-cycloalkyl; NR4R4', C(0)0R4 and substituted or unsubstituted haloalkyl; wherein each R4 and R4' are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl, substituted or unsubstituted C2-6 alkynyl, and C(0)OR4·; wherein R4 " is substituted or unsubstituted C1-6 alkyl.
R5 is selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6alkenyl, substituted or unsubstituted C2-6 alkynyl, CN, -C(0)OR3 and -S(0)20R3' ; wherein each R3 and Ry are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl, and substituted or unsubstituted C2-6 alkynyl; R6 is selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl (tert-butyl), substituted or unsubstituted C2-6 alkenyl, substituted or unsubstituted C2-6 alkynyl,
R2 is a group of the following formula
Figure imgf000132_0001
(R2) wherein m is 0, 1 or 2; r is 0, 1 or 2; t is 0, 1 , 2 or 3;
Wi is selected from C or N; each Rg, Rg·, Rg- and Rg- are independently selected from hydrogen, substituted or unsubstituted Ci-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2-6 alkynyl; alternatively, Rg and Rg· and/or Rg- and Rg- taken together with the carbon atom to which they are attached may form a substituted or unsubstituted cycloalkyl; alternatively, Rg and Rg· and/or Rg- and Rg- taken together with the carbon atom to which they are attached may form a carbonyl group; each R10, Rio·, R10” and R10- are independently selected from hydrogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2-6 alkynyl; alternatively, R10, R-io· and/or Rio ·, R-io- taken together with the carbon atom to which they are attached may form a substituted or unsubstituted cycloalkyl; alternatively, R10 and R-io· and/or Rio- and Rio- taken together with the carbon atom to which they are attached may form a carbonyl group;
R7 is selected from substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted aryl and substituted or unsubstituted aromatic heterocyclyl;
Rs is selected from hydrogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2- e alkynyl; Rs· is selected from hydrogen, substituted or unsubstituted Ci-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2- e alkynyl; alternatively, R7 and Rs taken together with -N(Rs )-[CH2]t- atoms to which they are attached form a substituted or unsubstituted heterocyclyl optionally fused with a substituted or unsubstituted aryl group; wherein the compound of formula (I) is optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
2. Compound according to claim 1 wherein
W3 is S and W4 is C;
Y is -C(O)-, -[C(RaRa')]n-, -CRa(CN)- or -CRa(CONRa'Ra ")-. wherein each Ra, Ra· and Ra” are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl, and substituted or unsubstituted C2-6 alkynyl; and n is 0, 1 , 2 or 3;
X is selected from a bond, -[C(RbRb )] -, -[C(RbRb )] C(0)[C(Rb"Rb )]q-> substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl-C(O), substituted or unsubstituted aryl- alkyl, wherein each Rb, Rb·, Rb- and Rb- are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl, and substituted or unsubstituted C2-6 alkynyl; and p is 0, 1 , 2 or 3; q is 0, 1 , 2 or 3;
Group A is a substituted or unsubstituted 5 to 10 membered mono or bicyclic heterocyclyl containing one nitrogen atom and optionally a second heteroatom selected from N and O, wherein A is attached to Y through a nitrogen atom; each Ri and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted Ci-6 alkyl, substituted or unsubstituted C2-6 alkenyl, substituted or unsubstituted C2-6 alkynyl, substituted or unsubstituted alkyl-cycloalkyl; NR4R4', C(0)0R4 and substituted or unsubstituted haloalkyl; wherein each R4 and R4' are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl, substituted or unsubstituted C2-6 alkynyl, and C(0)0R4·; wherein R4 " is substituted or unsubstituted C1-6 alkyl.
R5 is selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6alkenyl, substituted or unsubstituted C2-6 alkynyl, CN, -C(0)0R3 and -S(0)20R3' ; wherein each R3 and R3' are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl, and substituted or unsubstituted C2-6 alkynyl; R6 is selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl (tert-butyl), substituted or unsubstituted C2-6 alkenyl, substituted or unsubstituted C2-6 alkynyl,
R2 is a group of the following formula
Figure imgf000135_0001
wherein m is 0, 1 or 2; r is 0, 1 or 2; t is 0, 1 , 2 or 3; Wi is selected from C or N; each Rg, Rg·, Rg- and Rg- are independently selected from hydrogen, substituted or unsubstituted Ci-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2-6 alkynyl; alternatively, Rg and Rg· and/or Rg- and Rg- taken together with the carbon atom to which they are attached may form a substituted or unsubstituted cycloalkyl; alternatively, Rg and Rg· and/or Rg- and Rg- taken together with the carbon atom to which they are attached may form a carbonyl group; each R10, Rio·, R10” and Rio- are independently selected from hydrogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2-6 alkynyl; alternatively, R10, R-io· and/or Rio ·, R-io- taken together with the carbon atom to which they are attached may form a substituted or unsubstituted cycloalkyl; alternatively, R10 and R-io· and/or Rio- and R-io- taken together with the carbon atom to which they are attached may form a carbonyl group;
R7 is selected from substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted aryl and substituted or unsubstituted aromatic heterocyclyl;
Rs is selected from hydrogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2- e alkynyl;
Rs· is selected from hydrogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl and substituted or unsubstituted C2- e alkynyl; alternatively, R7 and Rs taken together with -N(Rs )-[CH2]t- atoms to which they are attached form a substituted or unsubstituted heterocyclyl optionally fused with a substituted or unsubstituted aryl group; wherein the compound of formula (I) is optionally in form of one of the stereoisomers, preferably enantiomers or diastereomers, a racemate or in form of a mixture of at least two of the stereoisomers, preferably enantiomers and/or diastereomers, in any mixing ratio, or a corresponding salt thereof, or a corresponding solvate thereof.
3. Compound according to claim 1 or 2 wherein group A is selected from:
Figure imgf000137_0001
wherein b is 0, 1 or 2; W2 is selected from O, C and N; wherein each R1 and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl, substituted or unsubstituted C2-6alkynyl, substituted or unsubstituted alkyl-cycloalkyl; N R4R4', C(0)OR4 and substituted or unsubstituted haloalkyl; wherein each R4 and R4' are independently selected from hydrogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl, substituted or unsubstituted C2-6 alkynyl, and C(0)OR4·; wherein R4" is substituted or unsubstituted C1-6 alkyl.
4. Compound according to claim 3, wherein A is
Figure imgf000137_0002
wherein b is 1 or 2;
W2 is N; wherein each Ri and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted alkyl-cycloalkyl; and substituted or unsubstituted haloalkyl.
5. Compound according to claim 4, wherein b is 1 ;
W2 is N; Ri is hydrogen and Rr is substituted or unsubstituted Ci-6 alkyl, and wherein Rr is directly attached to W2.
6. Compound according to any one of claims 1 to 5, wherein each Ri and Rr are independently selected from hydrogen, halogen, substituted or unsubstituted Ci-6 alkyl, substituted or unsubstituted alkyl-cycloalkyl; NR4R4', and substituted or unsubstituted haloalkyl; wherein each R4 and R4' are independently selected from hydrogen and substituted or unsubstituted C1-6 alkyl.
7. Compound according to any one of claims 1 to 6, wherein R2 is a group of the following formula:
Figure imgf000138_0001
(R2) wherein m is 1 ; r is 1 ; t is 0; Wi is C; each Rg, Rg·, Rg- and Rg- are hydrogen; each Rio, Rio·, Rio” and Rio- are hydrogen;
R7 is substituted or unsubstituted aryl;
Rs is substituted or unsubstituted C1-6 alkyl; and Rs· is substituted or unsubstituted C1-6 alkyl. 8. Compound according to any one of claims 1 to 7, wherein said compound is selected from:
[1] (1 -((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1 -yl)methanone;
[2] (1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(morpholino)methanone;
[3] (1 -((1 s,4s)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1 -yl)methanone;
[4] tert-butyl 4-(1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazole-3-carbonyl)-1 ,4-diazepane-1 -carboxylate;
[5] (1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(4-methyl-1 ,4-diazepan-1 -yl)methanone;
[6] (1 -((1 r,4R)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)((3S,5R)-3,5-dimethylpiperazin-1 -yl)methanone;
[7] (1 -((1 r,4r)-4-(dimethylamino)-4-(3-hydroxyphenyl)cyclohexyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1 -yl)methanone; [8] (1 -((1 s,4s)-4-(dimethylamino)-4-(3-fluorophenyl)cyclohexyl)-4, 5-dihydro- 1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[9] (1 -((1 s,4s)-4-(dimethylamino)-4-(4-fluorophenyl)cyclohexyl)-4, 5-dihydro- 1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[10] (1-((1s,4s)-4-(dimethylamino)-4-(3-methoxyphenyl)cyclohexyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[11] (1-((1s,4s)-4-(dimethylamino)-4-(4-methoxyphenyl)cyclohexyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[12] (1-((1 r,4r)-4-(dimethylamino)-4-(3-fluorophenyl)cyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[13] (1-((1 r,4r)-4-(dimethylamino)-4-(4-fluorophenyl)cyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[14] (1-((1 r,4r)-4-(dimethylamino)-4-(3-methoxyphenyl)cyclohexyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[15] (1-((1s,4s)-4-(dimethylamino)-4-(3-hydroxyphenyl)cyclohexyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[16] (1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(4-ethylpiperazin-1-yl)methanone;
[17] (1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(piperidin-1-yl)methanone;
[18] (1-((1 r,4r)-4-(methylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[19] (1-((1s,4s)-4-(dimethylamino)-4-(2-methoxyphenyl)cyclohexyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[20] (S)-tert-butyl 4-(1 -((1 r,4S)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5- dihydro-1 H-thieno[2,3-g]indazole-3-carbonyl)-3-methylpiperazine-1- carboxylate; [21] (R)-tert-butyl 4-(1-((1 r,4R)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5- dihydro-1 H-thieno[2,3-g]indazole-3-carbonyl)-3-methylpiperazine-1- carboxylate;
[22] (1 -((1 s,4s)-4-(methylamino)-4-phenylcyclohexyl)-4, 5-dihydro- 1 H-thieno[2,3- g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[23] (1 -((1 r,4r)-4-(dimethylamino)-4-(4-methoxyphenyl)cyclohexyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[24] tert-butyl 5-(1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-
1 H-thieno[2,3-g]indazole-3-carbonyl)hexahydropyrrolo[3,4-c]pyrrole-2(1 H)- carboxylate;
[25] tert-butyl 6-(1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-
1 H-thieno[2,3-g]indazole-3-carbonyl)-2,6-diazaspiro[3.4]octane-2- carboxylate;
[26] (1 -((1 r,4r)-4-((dimethylamino)methyl)-4-(3-methoxyphenyl)cyclohexyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[27] (1 -((1 s,4s)-4-((dimethylamino)methyl)-4-(3-methoxyphenyl)cyclohexyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[28] tert-butyl 4-(1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-
1 H-thieno[2,3-g]indazole-3-carbonyl)-6,6-difluoro-1 ,4-diazepane-1 - carboxylate;
[29] (1 -((1 r,4R)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)((R)-3-(hydroxymethyl)-4-methylpiperazin-1- yl)methanone;
[30] (1 -((1 r,4S)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)((S)-3-(hydroxymethyl)-4-methylpiperazin-1- yl)methanone;
[31] tert-butyl 2-(difluoromethyl)-4-(1-((1 r,4r)-4-(dimethylamino)-4- phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3-g]indazole-3- carbonyl)piperazine-1 -carboxylate; [32] (4-(cyclopropylmethyl)piperazin-1 -yl)(1 -((1 r,4r)-4-(dimethylamino)-4- phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)methanone;
[33] (4,6-dimethyl-1 ,4-diazepan-1 -yl)(1 -((1 r,4r)-4-(dimethylamino)-4- phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)methanone;
[34] (4,7-dimethyl-1 ,4-diazepan-1 -yl)(1 -((1 r,4r)-4-(dimethylamino)-4- phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)methanone;
[35] (1 -((1 r,4S)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)((S)-3-(dimethylamino)piperidin-1-yl)methanone;
[36] (1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(2-methyl-2,6-diazaspiro[3.4]octan-6-yl)methanone;
[37] (1 -(4-(4-(dimethylamino)-4-(3-hydroxyphenyl)piperidin-1 -yl)phenyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methyl-1 ,4-diazepan-1 - yl)methanone;
[38] (1 -(4-(4-(dimethylamino)-4-phenylpiperidin-1 -yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[39] (1 -(3-(4-(dimethylamino)-4-phenylpiperidin-1 -yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[40] tert-butyl (1 -(1 -(4-(4-(dimethylamino)-4-phenylpiperidin-1 -yl)phenyl)-4,5- dihydro-1 H-thieno[2,3-g]indazole-3-carbonyl)pyrrolidin-3- yl)(methyl)carbamate;
[41] (1-(4-(4-(dimethylamino)-4-ethylpiperidin-1-yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[42] (1 -(4-(2-methyl-3,4-dihydro-2H-spiro[isoquinoline-1 ,4'-piperidin]-1 '- yl)phenyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1- yl)methanone;
[43] (1 -(4-(4-(dimethylamino)-4-(3-hydroxyphenyl)piperidin-1 -yl)phenyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone; [44] (1 -(4-(4-(dimethylamino)-4-(3-methoxyphenyl)piperidin-1 -yl)phenyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[45] (1 -(4-(4-(methylamino)-4-phenylpiperidin-1 -yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[46] (1 -(4-(4-(3-hydroxyphenyl)-4-(methylamino)piperidin-1 -yl)phenyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[47] (1 -(3-(4-(methylamino)-4-phenylpiperidin-1 -yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[48] (1 -(3-(2-methyl-3,4-dihydro-2H-spiro[isoquinoline-1 ,4'-piperidin]-1 '- yl)phenyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1- yl)methanone;
[49] (1 -(3-(4-(dimethylamino)-4-(3-methoxyphenyl)piperidin-1 -yl)phenyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[50] (1 -(3-(4-(dimethylamino)-4-(3-hydroxyphenyl)piperidin-1 -yl)phenyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[51] (1-(4-(4-((dimethylamino)methyl)-4-phenylpiperidin-1-yl)phenyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[52] (1 -(4-(3-(dimethylamino)-3-phenylpiperidin-1 -yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[53] (1 -(3-(4-((dimethylamino)methyl)-4-phenylpiperidin-1 -yl)phenyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[54] (1 -(3-(4-((dimethylamino)methyl)-4-(3-hydroxyphenyl)piperidin-1 -yl)phenyl)- 4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[55] (1 -(4-(4-(dimethylamino)-4-phenylpiperidin-1 -yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methyl-1 ,4-diazepan-1-yl)methanone;
[56] (1-(3-(4-(dimethylamino)-4-(pyridin-2-yl)piperidin-1-yl)phenyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone; [57] (1-(3-(4-(dimethylamino)-4-(pyndin-2-yl)piperidin-1-yl)phenyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[58] (1-(4-(4-(dimethylamino)-4-(pyridin-2-yl)piperidin-1-yl)phenyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[59] (1 -(4-(4-(dimethylamino)-4-(4-methoxyphenyl)piperidin-1 -yl)phenyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[60] (1 -(4-(4-(dimethylamino)-4-(2-methoxyphenyl)piperidin-1 -yl)phenyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[61] (1-(4-(4-(dimethylamino)-4-(3-fluorophenyl)piperidin-1-yl)phenyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[62] (1 -(4-(4-(dimethylamino)-4-(4-fluorophenyl)piperidin-1 -yl)phenyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[63] (1 -(3-((4-(dimethylamino)-4-(3-hydroxyphenyl)piperidin-1 -yl)methyl)phenyl)- 4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[64] (1 -(4-((4-(dimethylamino)-4-phenylpiperidin-1 -yl)methyl)phenyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[65] (1 -(3-((4-(dimethylamino)-4-phenylpiperidin-1 -yl)methyl)phenyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[66] (1 -(4-((4-(dimethylamino)-4-(3-hydroxyphenyl)piperidin-1 -yl)methyl)phenyl)- 4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[67] (4-(dimethylamino)-4-(3-hydroxyphenyl)piperidin-1-yl)(3-(3-(4- methylpiperazine-1 -carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1 - yl)phenyl)methanone;
[68] (4-(dimethylamino)-4-phenylpiperidin-1 -yl)(4-(3-(4-methylpiperazine-1 - carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1-yl)phenyl)methanone;
[69] (4-(dimethylamino)-4-phenylpiperidin-1 -yl)(3-(3-(4-methylpiperazine-1 - carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1-yl)phenyl)methanone; [70] (2-methyl-3,4-dihydro-2H-spiro[isoquinoline-1 ,4'-piperidin]-1 '-yl)(4-(3-(4- methylpiperazine-1 -carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1 - yl)phenyl)methanone;
[71] (2-methyl-3,4-dihydro-2H-spiro[isoquinoline-1 ,4'-piperidin]-1 '-yl)(3-(3-(4- methylpiperazine-1 -carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1 - yl)phenyl)methanone;
[72] (4-(dimethylamino)-4-(3-methoxyphenyl)piperidin-1-yl)(4-(3-(4- methylpiperazine-1 -carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1 - yl)phenyl)methanone;
[73] (4-(methylamino)-4-phenylpiperidin-1 -yl)(4-(3-(4-methylpiperazine-1 - carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1-yl)phenyl)methanone;
[74] (4-(dimethylamino)-4-(3-hydroxyphenyl)piperidin-1-yl)(4-(3-(4- methylpiperazine-1 -carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1 - yl)phenyl)methanone;
[75] (4-(dimethylamino)-4-(3-methoxyphenyl)piperidin-1-yl)(3-(3-(4- methylpiperazine-1 -carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1 - yl)phenyl)methanone;
[76] (4-(methylamino)-4-phenylpiperidin-1 -yl)(3-(3-(4-methylpiperazine-1 - carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1-yl)phenyl)methanone;
[77] (1 -((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-1 H-thieno[2,3-g]indazol-3- yl)(4-methylpiperazin-1-yl)methanone;
[78] (7-Bromo-1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[79] 1 -((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-3-(4-methylpiperazine-1 - carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazole-7-carbonitrile;
[80] 1 -((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-3-(4-methylpiperazine-1 - carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazole-7-carboxylic acid; [81] 1-((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-3-(4-methylpiperazine-1- carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazole-7-sulfonic acid;
[82] (1 -((1 r,4r)-4-((2-Fluoroethyl)methylamino)-4-phenylcyclohexyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[83] (1 -((1 r,4r)-4-((2,2-difluoroethyl)(methyl)amino)-4-phenylcyclohexyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[84] (1 -((1 r,4r)-4-((2,2-Difluoroethyl)(methyl)amino)-4-phenylcyclohexyl)-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[85] (1 ,4-Diazepan-1 -yl)(1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)methanone;
[86] (1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(piperazin-1-yl)methanone;
[87] (1 -((1 r,4S)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)((S)-2-methylpiperazin-1-yl)methanone;
[88] (1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(hexahydropyrrolo[3,4-c]pyrrol-2(1 H)-yl)methanone;
[89] (1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(2,6-diazaspiro[3.4]octan-6-yl)methanone;
[90] (6,6-difluoro-1 ,4-diazepan-1 -yl)(1 -((1 r,4r)-4-(dimethylamino)-4- phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)methanone;
[91] (3-(difluoromethyl)piperazin-1-yl)(1-((1 r,4r)-4-(dimethylamino)-4- phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)methanone;
[92] (1 -(4-(4-(dimethylamino)-4-phenylpiperidin-1 -yl)phenyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(3-(methylamino)pyrrolidin-1-yl)methanone;
[93] (1 -((1 r,4R)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)((R)-2-methylpiperazin-1-yl)methanone; [94] (7-bromo-1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(piperazin-1-yl)methanone;
[95] (1 -((1 r,4R)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)((R)-3-(hydroxymethyl)piperazin-1-yl)methanone;
[96] (1 -((1 r,4S)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)((S)-3-(hydroxymethyl)piperazin-1-yl)methanone;
[97] (1 -((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-(2-fluoroethyl)piperazin-1-yl)methanone;
[98] (4-(2,2-difluoroethyl)piperazin-1 -yl)(1 -((1 r,4r)-4-(dimethylamino)-4- phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)methanone;
[99] (2-((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-2H- thieno[2,3-g]indazol-3-yl)(4-ethylpiperazin-1-yl)methanone;
[100] (2-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-2H-thieno[2,3- g]indazol-3-yl)(4-methyl-1 ,4-diazepan-1 -yl)methanone;
[101] (2-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-2H-thieno[2,3- g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[102] 2-(1 -((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)-2-(4-methylpiperazin-1-yl)acetonitrile;
[103] 2-(1 -((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)-2-(4-methylpiperazin-1-yl)acetamide;
[104] (1 r,4r)-4-(8-(tert-Butyl)-3-((4-methylpiperazin-1 -yl)methyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-1-yl)-N,N-dimethyl-1-phenylcyclohexanamine;
[105] N,N-Dimethyl-1-(4-(3-((4-methylpiperazin-1-yl)methyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-1-yl)phenyl)-4-phenylpiperidin-4-amine;
[106] (1 r,4r)-N,N-dimethyl-4-(3-((4-methylpiperazin-1-yl)methyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-1 -yl)-1 -phenylcyclohexanamine; [107] (1 -((4-(Dimethylamino)-4-phenylcyclohexyl)methyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1 -yl)methanone;
[108] (1 -(2-((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)ethyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1 -yl)methanone;
[109] (1 -(2-(4-(3-Hydroxyphenyl)-4-(methylamino)piperidin-1 -y I )eth y I )-4 , 5-d i hyd ro- 1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1 -yl)methanone;
[1 10] (1 -(2-(4-(dimethylamino)-4-(3-hydroxyphenyl)piperidin-1 -yl)ethyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1 -yl)methanone;
[1 11] (1 -(2-(4-((dimethylamino)methyl)-4-(3-hydroxyphenyl)piperidin-1 -yl)ethyl)- 4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1 -yl)methanone;
[1 12] (1 -(2-(4-(methylamino)-4-phenylpiperidin-1 -yl)ethyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1 -yl)methanone;
[1 13] (2-(2-(4-(dimethylamino)-4-(3-hydroxyphenyl)piperidin-1 -yl)ethyl)-4,5- dihydro-2H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1 -yl)methanone;
[1 14] (1 -(2-(4-(dimethylamino)-4-(4-fluorophenyl)piperidin-1 -yl)ethyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1 -yl)methanone;
[1 15] (1 -(2-(4-(dimethylamino)-4-(3-fluorophenyl)piperidin-1 -yl)ethyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1 -yl)methanone;
[1 16] (1 -(2-(4-((dimethylamino)methyl)-4-phenylpiperidin-1 -yl)ethyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1 -yl)methanone;
[1 17] (1 -(2-(4-(dimethylamino)-4-phenylpiperidin-1 -yl)ethyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1 -yl)methanone;
[1 18] 1 -(4-(Dimethylamino)-4-phenylpiperidin-1 -yl)-2-(3-(4-methylpiperazine-1 - carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1 -yl)ethanone;
[1 19] 1 -(4-(dimethylamino)-4-(3-fluorophenyl)piperidin-1-yl)-2-(3-(4- methylpiperazine-1 -carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1 - yl)ethanone; [120] 1 -(4-(dimethylamino)-4-(4-fluorophenyl)piperidin-1-yl)-2-(3-(4- methylpiperazine-1 -carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1 - yl)ethanone;
[121] 1 -(4-(dimethylamino)-4-(4-methoxyphenyl)piperidin-1 -yl)-2-(3-(4- methylpiperazine-1 -carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1 - yl)ethanone;
[122] 1 -(4-(dimethylamino)-4-(3-methoxyphenyl)piperidin-1 -yl)-2-(3-(4- methylpiperazine-1 -carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1 - yl)ethanone;
[123] 1 -(4-((dimethylamino)methyl)-4-phenylpiperidin-1 -yl)-2-(3-(4- methylpiperazine-1 -carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1 - yl)ethanone;
[124] 1 -(4-(dimethylamino)-4-(2-methoxyphenyl)piperidin-1 -yl)-2-(3-(4- methylpiperazine-1 -carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1 - yl)ethanone;
[125] 1 -(4-(diethylamino)-4-phenylpiperidin-1 -yl)-2-(3-(4-methylpiperazine-1 - carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1 -yl)ethanone;
[126] 1 -(4-(dimethylamino)-4-(3-hydroxyphenyl)piperidin-1 -yl)-2-(3-(4- methylpiperazine-1 -carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1 - yl)ethanone;
[127] 1 -(4-((dimethylamino)methyl)-4-(3-hydroxyphenyl)piperidin-1 -yl)-2-(3-(4- methylpiperazine-1 -carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1 - yl)ethanone; tert-butyl 4-(1 -((1 r,4r)-4-(dimethylamino)-4-(4-methoxyphenyl)cyclohexyl)-
[128]
4,5-dihydro-1 H-thieno[2,3-g]indazole-3-carbonyl)piperazine-1 -carboxylate; tert-butyl (1 -(1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazole-3-carbonyl)azetidin-3-yl)(methyl)carbamate; ((S)-4-benzyl-3-methylpiperazin-1 -yl)(1 -((1 r,4S)-4-(dimethylamino)-4-
[130]
phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)methanone;
((R)-4-benzyl-3-methylpiperazin-1 -yl)(1 -((1 r,4R)-4-(dimethylamino)-4-
[131 ]
phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)methanone;
(1 -((1 r,4R)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-
[132]
thieno[2,3-g]indazol-3-yl)((R)-3-methylpiperazin-1 -yl)methanone;
(1 -((1 r,4S)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-
[133]
thieno[2,3-g]indazol-3-yl)((S)-3-methylpiperazin-1 -yl)methanone;
1 -tert-butyl 2-methyl 4-(1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5- dihydro-1 H-thieno[2,3-g]indazole-3-carbonyl)piperazine-1 ,2-dicarboxylate;
(R)-tert-butyl 4-(1 -(4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-
[136] thieno[2,3-g]indazole-3-carbonyl)-2-(hydroxymethyl)piperazine-1 - carboxylate;
(1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-1 ,4-
[137] dihydrothieno[3',2':4,5]cyclopenta[1 ,2-c]pyrazol-3-yl)(4-methylpiperazin-1 - yl)methanone;
(1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-1 ,4-
[138] dihydrothieno[2',3':4,5]cyclopenta[1 ,2-c]pyrazol-3-yl)(4-methylpiperazin-1 - yl)methanone; tert-butyl ((S)-1 -(1 -((1 r,4S)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-
[139]
dihydro-1 H-thieno[2,3-g]indazole-3-carbonyl)piperidin-3-yl)carbamate;
(1 -((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[3,2-
[140]
g]indazol-3-yl)(4-methylpiperazin-1 -yl)methanone; [141] (1-((1 r,4r)-4-(dimethylamino)-4-(4-methoxyphenyl)cyclohexyl)-4,5-dihydro- 1 H-thieno[2,3-g]indazol-3-yl)(piperazin-1 -yl)methanone;
[142] (1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3- g]indazol-3-yl)(3-(methylamino)azetidin-1-yl)methanone;
[143] 4-(1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazole-3-carbonyl)piperazine-2-carboxylic acid;
[144] methyl 4-(1-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazole-3-carbonyl)piperazine-2-carboxylate;
[145] (R)-4-(1-((1 r,4R)-4-(dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazole-3-carbonyl)-2-(hydroxymethyl)piperazine-1 -carboxylic acid;
[146] ((S)-3-aminopiperidin-1-yl)(1-((1 r,4S)-4-(dimethylamino)-4- phenylcyclohexyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-3-yl)methanone;
[147] N-((1s,4s)-4-(dimethylamino)-4-phenylcyclohexyl)-2-(3-(4-methylpiperazine- 1 -carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1 -yl)acetamide;
[148] N-((1 r,4r)-4-(dimethylamino)-4-phenylcyclohexyl)-2-(3-(4-methylpiperazine- 1 -carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1 -yl)acetamide;
[149] (1-(2-(4-(dimethylamino)-4-(4-methoxyphenyl)piperidin-1-yl)ethyl)-4,5- dihydro-1 H-thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[150] (1 -(2-(4-(dimethylamino)-4-(4-fluorophenyl)piperidin-1 -yl)ethyl)-1 H- thieno[2,3-g]indazol-3-yl)(4-methylpiperazin-1-yl)methanone;
[151] (1-((1 r,4r)-4-(Dimethylamino)-4-phenylcyclohexyl)-4,5-dihydro-1 H- thieno[2,3-g]indazol-3-yl)(3-(dimethylamino)azetidin-1-yl)methanone; and
[152] 1 -(4-(Methylamino)-4-phenylpiperidin-1 -yl)-2-(3-(4-methylpiperazine-1 - carbonyl)-4,5-dihydro-1 H-thieno[2,3-g]indazol-1 -yl)ethanone.
9. Process for the preparation of a compound of formula (I) according to claim 1 :
Figure imgf000152_0001
wherein R1, Rr, R2, Rs, R6, W3, W4, X, Y and A have the same meaning as indicated in any one of claims 1 to 7, said process comprising treating a compound of formula (VII)
Figure imgf000152_0002
with a compound of formula (V):
Figure imgf000152_0003
wherein
Ri, Rr, R2, R5, R6, W3, W4, X, Y and A have the same meaning as indicated in any one of claims 1 to 7; and Z is selected from OH, Cl, N(OMe)Me and H.
10. Process according to claim 9, wherein W3 is S and W4 is C.
1 1. Process according to any one of claims 9 or 10 wherein Z is OH or Cl. 12. Process for the preparation of a compound of formula (I) according to claim 1 :
Figure imgf000153_0001
wherein Ri, Rr, R2, Rs, R6, W3, W4, X, Y and A have the same meaning as indicated in any one of claims 1 to 7, said process comprising a) treating a compound of formula (VII)
Figure imgf000153_0002
with a compound of formula (VIII):
Figure imgf000153_0003
wherein Ri, Rr, R2, R5, R6, W3, W4, X, Y and A have the same meaning as indicated in any one of claims 1 to 7; and Z is OH or O-alkyl; and b) treating the compound obtained in step a) with a compound of formula XI g R2 X~G
(XI) wherein R2, and X have the same meaning as indicated in any one of claims 1 to 7; and G is B(OH)2 when X is aryl, and G is a leaving group when X is0 alkylene.
13. Process according to claim 12, wherein W3 is S and W4 is C.
14. Process for the preparation of a compound of formula (I) according to claim 1
Figure imgf000154_0001
wherein R1, Rr, R2, R5, R6, W3, W4, X and A have the same meaning as indicated in any one of claims 1 to 7 and Y represents -C(O)-, said process comprising: 0
a) treating a compound of formula (XIV)
Figure imgf000154_0002
(XIV) with a compound of formula (VII)
Figure imgf000155_0001
b) treating the compound obtained in step a) with R2H; wherein Z is O-alkyl, OH or a leaving group.
15. Process according to claim 14, wherein W3 is S and W4 is C.
16. Use of a compound selected from
Figure imgf000155_0002
wherein R2, Rs, R6, W3, W4, and X have the same meaning as indicated in any one of claims 1 to 7 and Z is O-alkyl, OH; H or Cl for the manufacture of a compound according to claim 1.
17. A pharmaceutical composition which comprises a compound of formula (I) as defined in any of claims 1 to 8 or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, adjuvant or vehicle.
18. A compound as defined in any of claims 1 to 8 or a pharmaceutical salt thereof for use in therapy.
19. A compound as defined in any of claims 1 to 8 or a pharmaceutical salt thereof for use in the treatment or prophylaxis of pain, specially medium to severe pain, visceral pain, chronic pain, cancer pain, migraine, inflammatory pain, acute pain or neuropathic pain, allodynia or hyperalgesia.
PCT/EP2018/085479 2017-12-20 2018-12-18 4,5-dihydro-1h-thieno[2,3-g]indazolyl derivatives having multimodal activity against pain WO2019121670A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP17382870.8 2017-12-20
EP17382870 2017-12-20

Publications (1)

Publication Number Publication Date
WO2019121670A1 true WO2019121670A1 (en) 2019-06-27

Family

ID=60813786

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2018/085479 WO2019121670A1 (en) 2017-12-20 2018-12-18 4,5-dihydro-1h-thieno[2,3-g]indazolyl derivatives having multimodal activity against pain

Country Status (1)

Country Link
WO (1) WO2019121670A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3858439A1 (en) * 2020-02-03 2021-08-04 Esteve Pharmaceuticals, S.A. Amide derivatives having multimodal activity against pain
EP3858810A1 (en) * 2020-02-03 2021-08-04 Esteve Pharmaceuticals, S.A. Dialkylaminoarylcycloalkylamide derivatives having multimodal activity against pain
EP3858438A1 (en) * 2020-02-03 2021-08-04 Esteve Pharmaceuticals, S.A. 3,4-dihydro-2h-spiro[isoquinoline-1,4'-piperidin]amide derivatives having multimodal activity against pain

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2230243A1 (en) * 2009-02-25 2010-09-22 Neuroscienze Pharmaness S.C. A R.L. Pharmaceutical compounds
WO2017178510A1 (en) * 2016-04-12 2017-10-19 Laboratorios Del Dr. Esteve, S.A. Arylamide derivatives having multimodal activity against pain

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2230243A1 (en) * 2009-02-25 2010-09-22 Neuroscienze Pharmaness S.C. A R.L. Pharmaceutical compounds
WO2017178510A1 (en) * 2016-04-12 2017-10-19 Laboratorios Del Dr. Esteve, S.A. Arylamide derivatives having multimodal activity against pain

Non-Patent Citations (19)

* Cited by examiner, † Cited by third party
Title
"Progress in Pain Research and Management", vol. 25, 2003, IASP PRESS, article "Opioids and Pain Relief: A Historical Perspective"
BIOCHIM BIOPHYS ACTA, vol. 1828, 2013, pages 1541 - 9
DAVIES ET AL., TRENDS PHARMACOL SCI., vol. 28, 2007, pages 220 - 8
DICKENSON, A.H.; SUZUKI, R.: "Opioids in neuropathic pain: Clues from animal studies", EUR J PAIN, vol. 9, 2005, pages 113 - 6, XP004767581, DOI: doi:10.1016/j.ejpain.2004.05.004
DOLPHIN AC, NAT REV NEUROSCI, vol. 13, 2012, pages 542 - 55
ERRATUM IN: J PHARMACOL EXP THER., vol. 342, 2012, pages 232
GILRON ET AL., LANCET NEUROL, vol. 12, no. 11, November 2013 (2013-11-01), pages 1084 - 95
GOLDBERG, D.S.; MCGEE, S.J., BMC PUBLIC HEALTH, vol. 11, 2011, pages 770
HOPKINS, NAT CHEM BIOL., vol. 4, 2008, pages 682 - 90
KROGSGAARD-LARSEN ET AL.: "Textbook of Drug design and Discovery", April 2002, TAYLOR & FRANCIS
LEHAR ET AL., NAT BIOTECHNOL, vol. 27, 2009, pages 659 - 666
NEUMAIER ET AL., PROG. NEUROBIOL., vol. 129, 2015, pages 1 - 36
PERRET; LUO, NEUROTHERAPEUTICS, vol. 6, 2009, pages 679 - 92
SCHRODER ET AL., J PHARMACOL EXP THER., vol. 337, 2011, pages 312 - 20
TURK, D.C.; WILSON, H.D.; CAHANA, A., LANCET, vol. 377, 2011, pages 2226 - 2235
VINK; ALEWOOD, BIOCHEMICAL, 2012
VINK; ALEWOOD, BR J PHARMACOL., vol. 167, 2012, pages 970 - 89
ZAMPONI ET AL., PHARMACOL. REV., vol. 67, 2015, pages 821 - 70
ZHANG ET AL., CELL DEATH DIS., vol. 5, 2014, pages e1138

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3858439A1 (en) * 2020-02-03 2021-08-04 Esteve Pharmaceuticals, S.A. Amide derivatives having multimodal activity against pain
EP3858810A1 (en) * 2020-02-03 2021-08-04 Esteve Pharmaceuticals, S.A. Dialkylaminoarylcycloalkylamide derivatives having multimodal activity against pain
EP3858438A1 (en) * 2020-02-03 2021-08-04 Esteve Pharmaceuticals, S.A. 3,4-dihydro-2h-spiro[isoquinoline-1,4'-piperidin]amide derivatives having multimodal activity against pain

Similar Documents

Publication Publication Date Title
EP3442960A1 (en) Arylamide derivatives having multimodal activity against pain
EP3288941B1 (en) Spiro-isoquinoline-3,4&#39;-piperidine compounds having activity against pain
WO2019121670A1 (en) 4,5-dihydro-1h-thieno[2,3-g]indazolyl derivatives having multimodal activity against pain
EP3377500B1 (en) Oxadiazaspiro compounds for the treatment of drug abuse and addiction
AU2021280991A1 (en) Pyrazolo(1,5-a)pyrimidine derivatives having multimodal activity against pain
US20220380345A1 (en) Homopiperazinyl and homopiperidinyl quinazolin-4(3h)-one derivatives having multimodal activity against pain
WO2020089397A1 (en) Substituted quinazolin-4(3h)-one derivatives having multimodal activity against pain
WO2020089400A1 (en) Piperazinyl and piperidinyl quinazolin-4(3h)-one derivatives having activity against pain
WO2018100048A1 (en) 2-phenyl-2h-pyrazolo[3,4-d]pyridazine derivatives having activity against pain
EP3630768A1 (en) (hetero)arylalkylamino-pyrazolopyridazine derivatives having multimodal activity against pain
AU2016343551A1 (en) Oxa-diazaspiro compounds having activity against pain
WO2020016315A1 (en) Fused dihydroindazole derivatives having multimodal activity against pain
WO2017067665A1 (en) Substituted morpholine derivatives having activity against pain
WO2019180188A1 (en) Aminopropoxyphenyl and benzyl 1-oxa-4,9-diazaspiroundecane derivatives having multimodal activity against pain
EP3630765A1 (en) Substituted pyrrolidinyl and piperidinyl pyrazolopyridazine derivatives having multimodal activity against pain
JP6891386B2 (en) Oxa-azaspiro compounds that are active against pain
WO2020115045A1 (en) Substituted 1h-benzo[d]imidazole derivatives having multimodal activity against pain and pain related conditions
WO2020089263A1 (en) Optionally nitrogenated isoquinolin-1(2h)-ones and 1h-isochromen- 1-ones for treating pain and pain related conditions
JP2018531267A6 (en) Oxa-azaspiro compounds having activity against pain
WO2018153546A1 (en) Tetrahydropyran and tetrahydrothiopyran methanone derivatives having multimodal activity against pain
WO2019149919A1 (en) Aminopropoxypiperidinylamido derivatives having multimodal activity against pain

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: 18816117

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: 18816117

Country of ref document: EP

Kind code of ref document: A1