Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
NITROGEN CONTAINING BICYCLIC DERIVATIVES FOR TREATING PAIN AND PAIN RELATED CONDITIONS
Document Type and Number:
WIPO Patent Application WO/2018/115069
Kind Code:
A1
Abstract:
The present invention relates to new compounds of formula (I): showing great affinity and activity towards the subunit α2δ of voltage-gated calcium channels (VGCC),especially the α2δ-1 subunit of voltage-gated calcium channels or dual activity towards the subunit α2δ of voltage-gated calcium channels (VGCC), especially the α2δ-1subunit of voltage-gated calcium channels, and the noradrenaline transporter (NET).

Inventors:
ALMANSA-ROSALES CARMEN (ES)
VIRGILI-BERNADÓ MARINA (ES)
ALONSO-XALMA MÓNICA (ES)
Application Number:
PCT/EP2017/083724
Publication Date:
June 28, 2018
Filing Date:
December 20, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ESTEVE LABOR DR (ES)
International Classes:
C07D409/12; A61K31/4709; A61K31/4725; A61K31/502; A61K31/517; A61P25/00; A61P29/00; C07D215/14; C07D217/02; C07D237/30
Domestic Patent References:
WO2004043931A12004-05-27
WO2016154027A12016-09-29
Foreign References:
US5023269A1991-06-11
US20030158185A12003-08-21
Other References:
F.P. BYMASTER ET AL: "Duloxetine (Cymbalta(TM)), a dual inhibitor of serotonin and norepinephrine reuptake", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 13, no. 24, 1 December 2003 (2003-12-01), AMSTERDAM, NL, pages 4477 - 4480, XP055340775, ISSN: 0960-894X, DOI: 10.1016/j.bmcl.2003.08.079
TURK, D.C.; WILSON, H.D.; CAHANA, A., LANCET, vol. 377, 2011, pages 2226 - 2235
GOLDBERG, D.S.; MCGEE, S.J., BMC PUBLIC HEALTH, vol. 11, 2011, pages 770
ZAMPONI ET AL., PHARMACOL. REV., vol. 67, 2015, pages 821 - 870
PERRET; LUO, NEUROTHERAPEUTICS, vol. 6, 2009, pages 679 - 692
NEUMAIER ET AL., PROG. NEUROBIOL., vol. 129, 2015, pages 1 - 36
VINK; ALEWOOD, BRITISH J. PHARMACOL., vol. 167, 2012, pages 970 - 989
DAVIES ET AL., TRENDS PHARMACOL. SCI., vol. 28, 2007, pages 220 - 228
DOLPHIN,A.C., NAT. REV. NEUROSCI., vol. 13, 2012, pages 542 - 555
DOLPHIN,A.C., BIOCHIM. BIOPHYS. ACTA, vol. 1828, 2013, pages 1541 - 1549
MASON,S.T., PROG. NEUROBIOL., vol. 16, 1981, pages 263 - 303
KASPER ET AL., EXPERT OPIN. PHARMACOTHER., vol. 1, 2000, pages 771 - 782
OSSIPOV ET AL., J. CLIN. INVEST., vol. 120, 2010, pages 3779 - 3787
WANG ET AL., J. PAIN, vol. 14, 2013, pages 845 - 853
CHABOT-DORE ET AL., NEUROPHARMACOLOGY, vol. 99, 2015, pages 285 - 300
FAIRBANKS ET AL., PHARMACOL. THER., vol. 123, 2009, pages 224 - 238
MOCHIZUCKI,D., PSYCHOPHARMACOL, vol. 1, 2004, pages S15 - S19
HARTRICK,C.T., EXPERT OPIN. INVESTIG. DRUGS, vol. 21, 2012, pages 1827 - 1834
LEHAR ET AL., NAT. BIOTECHNOL., vol. 27, 2009, pages 659 - 666
GILRON ET AL., LANCET NEUROL., vol. 12, no. 11, 2013, pages 1084 - 1095
SCHRODER ET AL., J. PHARMACOL. EXP. THER., vol. 337, 2011, pages 312 - 320
ZHANG ET AL., CELL DEATH DIS., vol. 5, 2014, pages e1138
HOPKINS, A.L., NAT. CHEM. BIOL., vol. 4, 2008, pages 682 - 690
NICOLSON ET AL., HARV. REV. PSYCHIATRY, vol. 17, 2009, pages 407 - 420
TANABE ET AL., J. NEUROOSCI. RES., 2008
HAYASHIDA,K., EUR. J. PHARMACOL., vol. 598, 2008, pages 21 - 26
MIRANDA, H.F. ET AL., J. OROFAC. PAIN, vol. 27, 2013, pages 361 - 366
PHARMACOLOGY, vol. 95, 2015, pages 59 - 64
FRAMPTON, J.E., CNS DRUGS, vol. 28, 2014, pages 835 - 854
HAJOS, M. ET AL., CNS DRUG REV., vol. 10, 2004, pages 23 - 44
KROGSGAARD-LARSEN ET AL.: "Textbook of Drug design and Discovery", April 2002, TAYLOR & FRANCIS
ANGEW. CHEM. INT. ED., vol. 43, 2004, pages 2816
ANGEW. CHEM. INT. ED., vol. 44, 2005, pages 1687
ANGEW. CHEM. INT. ED., vol. 54, 2015, pages 2260
"IASP, Classification of chronic pain", 2002, IASP PRESS, pages: 210
Attorney, Agent or Firm:
CARPINTERO LOPEZ, Francisco (ES)
Download PDF:
Claims:
A compound of general formula (I):

wherein

n is 0 or 1 ;

Z is one of the following moieties

where

the dotted line represents an optional double bond;

R4 and R4' independently represent a hydrogen atom; a halogen, a branched or unbranched Ci-6 alkyl radical; or a branched or unbranched Ci-6 haloalkyl;

A, B, D and E independently from one another represents -N-; -NH-; -CH-; -CH2- or -C(O)-;

with the proviso that at least one of A, B, D or E is -N- or -NH-; and

with the proviso that when n is 0, Z does not represent a quinoline or isoquinoline; Ri is selected from an optionally substituted 5 to 9 membered aryl group; an optionally substituted 5 to 9 membered heteroaryl group having at least one heteroatom selected from the group of N, O or S; or an optionally substituted C3-9 heterocycloalkyl group having at least one heteroatom selected from the group of N, O or S;

R2 and R3 independently represent a hydrogen atom or a branched or unbranched C1-6 alkyl radical; or a pharmaceutically acceptable salt, isomer, prodrug or solvate thereof.

2. A compound according to claim 1 where Ri represents a benzene, a thiophene, a thiazole, a pyridine or a tetrahydropyran all of them optionally substituted by at least one substituent selected from halogen, C1-6 alkyl, Ci-6-alkoxy, Ci-6-haloalkoxy, C1-6- haloalkyl or a hydroxyl group.

3. A compound according to claim 1 where Ri represents a benzene or a tiophene.

4. A compound according to claim 1 where Z is selected from:

wherein R4 and R4> independently represent a hydrogen atom; a halogen, a branched or unbranched Ci-6 alkyl radical; or a branched or unbranched Ci-6 haloalkyl.

5. A compound according to claim 1 having one of the following formula (la), (lb), (Ia1 ), (Ia2), (Ib1 ) or (Ib2):

6. A compound according to claim 1 selected from the following list :

[1 ] /V-methyl-3-((1 -methyl-1 ,2,3,4-tetrahydroquinolin-5-yl)oxy)-3-(thiophen-2- yl)propan-1 -amine;

[2] /V-methyl-3-((2-methyl-1 ,2,3,4-tetrahydroisoquinolin-5-yl)oxy)-3-(thiophen-2- yl)propan-1 -amine;

[3] 3-((2-(2!2-Difluoroethyl)-1 !2!3!4-tetrahydroisoquinolin-5-yl)oxy)-/V-methyl-3- (thiophen-2-yl)propan-1 -amine;

[4] (R)-/V-methyl-3-(phthalazin-5-yloxy)-3-(thiophen-2-yl)propan-1 -amine;

[5] (S)-/V-methyl-3-(phthalazin-5-yloxy)-3-(thiophen-2-yl)propan-1 -amine;

[6] /V-methyl-3-(phthalazin-5-yloxy)-3-(thiophen-2-yl)propan-1 -amine;

[7] 3-Methyl-5-(3-(methylamino)-1 -(thiophen-2-yl)propoxy)quinazolin-4(3/-/)-one;

[8] 3-Methyl-8-(3-(methylamino)-1 -(thiophen-2-yl)propoxy)quinazolin-4(3/-/)-one;

[9] 2-Methyl-5-(3-(methylamino)-1 -(thiophen-2-yl)propoxy)isoquinolin-1 (2/-/)-one; [10] /V-methyl-3-(quinazolin-5-yloxy)-3-(thiophen-2-yl)propan-1 -amine;

[1 1 ] /V-ethyl-3-phenyl-3-(phthalazin-5-yloxy)propan-1 -amine;

[12] /V-ethyl-3-(phthalazin-5-yloxy)-3-(thiophen-2-yl)propan-1 -amine;

[13] (R)-/V-ethyl-3-phenyl-3-(phthalazin-5-yloxy)propan-1 -amine;

[14] (R)-/V-ethyl-3-(phthalazin-5-yloxy)-3-(thiophen-2-yl)propan-1 -amine;

[15] (S)-3-methyl-5-(3-(methylamino)-1 -(thiophen-2-yl)propoxy)quinazolin-

4(3H)-one;

[16] (R)-3-methyl-5-(3-(methylamino)-1 -(thiophen-2-yl)propoxy)quinazolin- 4(3H)-one;

[17] 3-(lsoquinolin-5-ylmethoxy)-/V-methyl-3-phenylpropan-1 -amine;

[18] 3-(lsoquinolin-8-ylmethoxy)-/V-methyl-3-phenylpropan-1 -amine;

[19] /V-methyl-3-phenyl-3-(quinolin-5-ylmethoxy)propan-1 -amine;

[20] 3-(lsoquinolin-8-ylmethoxy)-/V-methyl-3-(thiophen-2-yl)propan-1 -amine;

[21 ] 3-(lsoquinolin-5-ylmethoxy)-/V-methyl-3-(thiophen-2-yl)propan-1 -amine;

[22] /V-methyl-3-(quinolin-5-ylmethoxy)-3-(thiophen-2-yl)propan-1 -amine;

[23] fS)-/V-methyl-3-((1 -methyl-1 ,2,3,4-tetrahydroquinolin-5-yl)oxy)-3-(thiophen-

2-yl)propan-1 -amine;

[24] (R)-/V-methyl-3-((1 -methyl-1 ,2,3,4-tetrahydroquinolin-5-yl)oxy)-3-(thiophen- 2-yl)propan-1 -amine;

[25] (S)-/V-methyl-3-((2-methyl-1 !2,3,4-tetrahydroisoquinolin-5-yl)oxy)-3- (thiophen-2-yl)propan-1 -amine;

[26] (R)-/V-methyl-3-((2-methyl-1 !2,3,4-tetrahydroisoquinolin-5-yl)oxy)-3- (thiophen-2-yl)propan-1 -amine;

[27] /V-ethyl-3-((2-methyl-1 !2,3,4-tetrahydroisoquinolin-5-yl)oxy)-3-(thiophen-2- yl)propan-1 -amine;

[28] (S)-1 -methyl-5-(3-(methylamino)-1 -(thiophen-2-yl)propoxy)quinolin-2(1 H)- one;

[29] (R)-1 -methyl-5-(3-(methylamino)-1 -(thiophen-2-yl)propoxy)quinolin-2(1 H)- one;

[30] (R)-5-(3-(ethylamino)-1 -(thiophen-2-yl)propoxy)-3-methylquinazolin-4(3/-/)- one;

[31 ] (R)-5-(3-(ethylamino)-1 -(thiophen-2-yl)propoxy)-2-methylisoquinolin-1 (2/-/)- one;

[32] (R)-8-(3-(ethylamino)-1 -(thiophen-2-yl)propoxy)-3-methylquinazolin-4(3/-/)- one;

[33] (S)-/V-methyl-3-(quinazolin-5-yloxy)-3-(thiophen-2-yl)propan-1 -amine; [34] (S)-8-(3-(ethylamino)-1 -(thiophen-2-yl)propoxy)-3-methylquinazolin-4(3/-/)- one;

[35] (S)-5-(3-(ethylamino)-1 -(thiophen-2-yl)propoxy)-2-methylisoquinolin-1 (2/-/)- one;

[36] (S)-5-(3-(ethylamino)-1 -(thiophen-2-yl)propoxy)-3-methylquinazolin-4(3/-/)- one;

[37] (R)-/V-methyl-3-(quinazolin-5-yloxy)-3-(thiophen-2-yl)propan-1 -amine;

[38] (S)-/V-ethyl-3-(quinazolin-5-yloxy)-3-(thiophen-2-yl)propan-1 -amine;

[39] (R)-/V-ethyl-3-(quinazolin-5-yloxy)-3-(thiophen-2-yl)propan-1 -amine;

[40] (R)-/V-ethyl-3-(quinazolin-5-yloxy)-3-(thiophen-3-yl)propan-1 -amine;

[41 ] (R)-8-(3-(ethylamino)-1 -(thiophen-3-yl)propoxy)-3-methylquinazolin-4(3/-/)- one;

[42] (R)-5-(3-(ethylamino)-1 -(thiophen-3-yl)propoxy)-2-methylisoquinolin-1 (2/-/)- one;

[43] (R)-3-methyl-5-(3-(methylamino)-1 -(thiophen-3-yl)propoxy)quinazolin- 4(3H)-one;

[44] (/?)-8-(3-(ethylamino)-1 -(thiophen-2-yl)propoxy)-3,6-dimethylquinazolin- 4(3H)-one and

[45] (R)-7-fluoro-3-methyl-5-(3-(methylamino)-1 -(thiophen-2- yl)propoxy)quinazolin-4(3/-/)-one;

a pharmaceutically acceptable salt, prodrug or solvate thereof.

7. A compound according to claim 1 having general formula (Ic):

(Ic) wherein Ri , R2, R3, A, B and n are as defined in claim 1 with the proviso that at least one of A or B represents a -N(R)- where R can be a hydrogen or a branched or unbranched Ci-6 alkyl radical; or a branched or unbranched Ci-6 haloalkyl.

8. A compound according to claim 7 selected from :

[1] /V-methyl-3-((1 -methyl-1 ,2,3, 4-tetrahydroquinolin-5-yl)oxy)-3-(thiophen-2-yl)propan- 1 -amine and

[3] 3-((2-(2,2-Difluoroethyl)-1 ,2,3,4-tetrahydroisoquinolin-5-yl)oxy)-/V-methyl-3- (thiophen-2-yl)propan-1 -amine;

or a pharmaceutically acceptable salt, prodrug or solvate thereof.

9. Process for the preparation of a compound of formula (la):

(la) comprising:

A) the reaction of a compound of formula (II):

(II) with a compound of formula (Ilia) or (1Mb):

Z-OH or Z-X

(Ilia) (1Mb)

wherein Ri , R2, R3 and Z are as defined in claim 1 and X represents a halogen, or

B) the reaction of a compound of formula (Va):

(Va)

with a compound of formula (VI):

HNR2R3

(VI)

wherein Ri , R2, R3 and Z are as defined in claim 1 and LG represents a leaving group, such as chloro, bromo, iodo, mesylate, tosylate, nosylate or triflate.

10. Process for the preparation of a compound of general formula (lb):

comprising the reaction between a compound of formula (II):

(II)

and a compound of formula (III c):

wherein Ri , R2, R3 and Z are as defined in claim 1 and LG represents a leaving group, such as chloro, bromo, iodo, mesylate, tosylate, nosylate or triflate.

1 1 . A compound according to any of claims 1 to 8 for use as a medicament.

12. A compound according to any of claims 1 to 8, for use in the treatment and/or prophylaxis of diseases and/or disorders mediated by the subunit α2δ, especially α2δ-1 subunit of voltage-gated calcium channels and/or noradrenaline transporter (NET). 13. A compound for use according to claim 12, where the disease or disorder is pain, depression, anxiety and attention-deficit-/hyperactivity disorder (ADHD)

14. A compound for use according to claim 13, where pain is medium to severe pain, visceral pain, chronic pain, cancer pain, migraine, inflammatory pain, acute pain neuropathic pain, allodynia or hyperalgesia, including mechanical allodynia or thermal hyperalgesia.

15. A pharmaceutical composition comprising a compound of general formula (I) according to any of claims 1 to 8 or a pharmaceutically acceptable salt, isomer, prodrug or solvate thereof, and at least a pharmaceutically acceptable carrier, additive, adjuvant or vehicle.

Description:
NITROGEN CONTAINING BICYCLIC DERIVATIVES FOR TREATING PAIN AND

PAIN RELATED CONDITIONS

FIELD OF THE INVENTION

The present invention relates to new compounds that show great affinity and activity towards the subunit α2δ of voltage-gated calcium channels (VGCC), especially the α2δ- 1 subunit of voltage-gated calcium channels or dual activity towards the subunit α2δ of voltage-gated calcium channels (VGCC), especially the α2δ-1 subunit of voltage-gated calcium channels, and the noradrenaline transporter (NET). The invention is also related to the process for the preparation of said compounds as well as to compositions comprising them, and to their use as medicaments.

BACKGROUND OF THE INVENTION

The adequate management of pain represents 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.; 2011 ; 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 correlated to comorbidities, such as depression, anxiety and insomnia, which leads to important productivity losses and socio-economical burden (Goldberg, D.S., McGee, S.J.; 2011 ; 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 -870). The VGCC are assembled through interactions of different subunits, namely a1 (Caval ), β (CavP) α2δ (Cava26) and γ (Ca v y). The a1 subunits are the key porous forming units of the channel complex, being responsible for Ca 2+ conduction and generation of Ca 2+ influx. The α2δ, β, and γ subunits are auxiliary, although they are very important for the regulation of the channel since they increase the expression of a1 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 (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. 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; 679-692; Zamponi et al., 2015; 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.

The Ca v 1 and Ca v 2 subfamilies contain an auxiliary α2δ subunit which is the therapeutic target of the gabapentinoid drugs of value in certain epilepsies and chronic neuropathic pain (Perret and Luo, 2009; Vink and Alewood; British J. Pharmacol.; 2012; 167; 970- 989). To date, there are four known α2δ subunits, each encoded by a unique gene and all possessing splice variants. Each α2δ protein is encoded by a single messenger RNA and is post-translationally cleaved and then linked by disulfide bonds. Four genes encoding α2δ subunits have now been cloned. α2δ-1 was initially cloned from skeletal muscle and shows a fairly ubiquitous distribution. The α2δ-2 and α2δ-3 subunits were subsequently cloned from brain. The most recently identified subunit, α2δ-4, is largely non-neuronal. The human α2δ-4 protein sequence shares 30, 32 and 61 % identity with the human α2δ-1 , α2δ-2 and α2δ-3 subunits, respectively. The gene structure of all α2δ subunits is similar. All α2δ subunits show several splice variants (Davies et al.; Trends Pharmacol. Sci.; 2007; 28; 220-228; Dolphin,A.C.; Nat. Rev. Neurosci.; 2012; 13; 542- 555; Dolphin,A.C.; Biochim. Biophys. Acta; 2013; 1828; 1541 -1549).

The Ca v a26-1 subunit may play an important role in neuropathic pain development (Perret and Luo, 2009; Vink and Alewood, 2012). Biochemical data have indicated a significant Ca v a28-1 , but not Ca v a28-2, 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 Ca v a,25-1 subunit to the central presynaptic terminals diminishes tactile allodynia in nerve injured animals, suggesting that elevated DRG Ca v a28-1 subunit contributes to neuropathic allodynia.

The Ca v a28-1 subunit (and the Ca v a28-2, but not Ca v a28-3 and Ca v a28-4, subunits) is the binding site for gabapentin which has anti-allodynic/hyperalgesic properties in patients and animal models. Because injury-induced Ca v a28-1 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 a28-1 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 a28-1 subunit can block nerve injury-induced Ca v a28-1 upregulation and prevent the onset of allodynia and reserve established allodynia.

As above mentioned, the α2δ 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 a28-1 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; Vink and Alewood, 2012, Zamponi et al., 2015).

Thus, the present invention relates to compounds with inhibitory effect towards α2δ subunits of voltage-gated calcium channels, preferably towards α2δ-1 subunit of voltage- gated calcium channels.

It is also known that Noradrenaline (NA), also called norepinephrine, functions in the human brain and body as a hormone and neurotransmitter. Noradrenaline exerts many effects and mediates a number of functions in living organisms. The effects of noradrenaline are mediated by two distinct super-families of receptors, named alpha- and beta-adrenoceptors. They are further divided into subgroups exhibiting specific roles in modulating behavior and cognition of animals. The release of the neurotransmitter noradrenaline throughout the mammalian brain is important for modulating attention, arousal, and cognition during many behaviors (Mason, ST.; Prog. Neurobiol.; 1981 ; 16; 263-303). The noradrenaline transporter (NET, SLC6A2) is a monoamine transporter mostly expressed in the peripheral and central nervous systems. NET recycles primarily NA, but also serotonin and dopamine, from synaptic spaces into presynaptic neurons. NET is a target of drugs treating a variety of mood and behavioral disorders, such as depression, anxiety, and attention-deficit hyperactivity disorder (ADHD). Many of these drugs inhibit the uptake of NA into the presynaptic cells through NET. These drugs therefore increase the availability of NA for binding to postsynaptic receptors that regulate adrenergic neurotransmission. NET inhibitors can be specific. For example, the ADHD drug atomoxetine is a NA reuptake inhibitor (NRI) that is highly selective for NET. Reboxetine was the first NRI of a new antidepressant class (Kasper et al.; Expert Opin. Pharmacother.; 2000; 1 ; 771 -782). Some NET inhibitors also bind multiple targets, increasing their efficacy as well as their potential patient population.

Endogenous, descending noradrenergic fibers impose analgesic control over spinal afferent circuitry mediating the transmission of pain signals (Ossipov et al.; J. Clin. Invest.; 2010; 120; 3779-3787). Alterations in multiple aspects of noradrenergic pain processing have been reported, especially in neuropathic pain states (Ossipov et a., 2010; Wang et al.; J. Pain; 2013; 14; 845-853). Numerous studies have demonstrated that activation of spinal a2-adrenergic receptors exerts a strong antinociceptive effect. Spinal clonidine blocked thermal and capsaicin-induced pain in healthy human volunteers (Ossipov et a., 2010). Noradrenergic reuptake inhibitors have been used for the treatment of chronic pain for decades: most notably the tricyclic antidepressants, amitriptyline, and nortriptyline. Once released from the presynaptic neuron, NA typically has a short-lived effect, as much of it is rapidly transported back into the nerve terminal. In blocking the reuptake of NA back into the presynaptic neurons, more neurotransmitter remains for a longer period of time and is therefore available for interaction with pre- and postsynaptic ot2-adrenergic receptors (AR). Tricyclic antidepressants and other NA reuptake inhibitors enhance the antinociceptive effect of opioids by increasing the availability of spinal NA. The c^A-AR subtype is necessary for spinal adrenergic analgesia and synergy with opioids for most agonist combinations in both animal and humans (Chabot-Dore et al.; Neuropharmacology; 2015; 99; 285-300). A selective upregulation of spinal NET in a rat model of neuropathic pain with concurrent downregulation of serotonin transporters has been shown (Fairbanks et al.; Pharmacol. Ther.; 2009; 123; 224-238). Inhibitors of NA reuptake such as nisoxetine, nortriptyline and maprotiline and dual inhibitors of the noradrenaline and serotonin reuptake such as imipramine and milnacipran produce potent anti-nociceptive effects in the formalin model of tonic pain. Neuropathic pain resulting from the chronic constriction injury of the sciatic nerve was prevented by the dual uptake inhibitor, venlafaxine. In the spinal nerve ligation model, amitriptyline, a non-selective serotonin and noradrenaline reuptake blocker, the preferential noradrenaline reuptake inhibitor, desipramine and the selective serotonin and noradrenaline reuptake inhibitors, milnacipran and duloxetine, produce a decrease in pain sensitivity whereas the selective serotonin reuptake inhibitor, fluoxetine, is ineffective (Mochizucki,D.; Psychopharmacol.; 2004; Supplm. 1 ; S15-S19; Hartrick,C.T.; Expert Opin. Investig. Drugs; 2012; 21 ; 1827-1834). A number of nonselective investigational agents focused on noradrenergic mechanisms with the potential for additive or even synergistic interaction between multiple mechanisms of action is being developed (Hartrick, 2012).

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; 12(1 1 ); 1084-1095). In fact, positive synergistic interaction for several compounds, including analgesics, has been described (Schroder et al; J. Pharmacol. Exp. Ther.; 201 1 ; 337; 312-320; Zhang et al.; Cell Death Dis.; 2014; 5; e1 138; Gilron et al., 2013). Given the significant differences in pharmacokinetics, metabolism 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, A.L.; Nat. Chem. Biol.; 2008; 4; 682-690). 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).

Thus, in a preferred embodiment, the compounds of the present invention having affinity for α2δ subunits of voltage-gated calcium channels, preferably towards the α2δ-1 subunit of voltage-gated calcium channels, additionally have inhibitory effect towards the noradrenaline transporter (NET) and are, thus, more effective to treat chronic pain.

There are two potentially important interactions between NET and α2δ-1 inhibition: 1 ) synergism in analgesia, thus reducing the risk of specific side effects; and 2) inhibition of pain-related affective comorbidities such as anxiety and/or depressive like behaviors (Nicolson et al.; Harv. Rev. Psychiatry; 2009; 17; 407-420).

1 ) Preclinical research has demonstrated that gabapentinoids attenuated pain- related behaviors through supraspinal activation of the descending noradrenergic system (Tanabe et al.; J. Neuroosci. Res.; 2008; Hayashida,K.; Eur. J. Pharmacol.; 2008; 598; 21 -26). In consequence, the α2δ-1 -related analgesia mediated by NA-induced activation of spinal (^-adrenergic receptors can be potentiated by the inhibition of the NET. Some evidence from combination studies in preclinical models of neuropathic pain exist. Oral duloxetine with gabapentin was additive to reduce hypersensitivity induced by nerve injury in rats

(Hayashida; 2008). The combination of gabapentin and nortriptyline was synergic in mice submitted to orofacial pain and to the peripheral nerve injury model (Miranda, H.F. et al.; J. Orofac. Pain; 2013; 27; 361 -366; Pharmacology; 2015; 95; 59-64).

2) Drug modulation of NET and α2δ-1 has been shown to produce antidepressant and anti-anxiety effects respectively (Frampton, J.E.; CNS Drugs; 2014; 28; 835- 854; Hajos, M. et al.; CNS Drug Rev.; 2004; 10; 23-44). In consequence, a dual drug that inhibited the NET and α2δ-1 subunit of VGCC may also stabilize pain- related mood impairments by acting directly on both physical pain and the possible mood alterations.

SUMMARY OF THE INVENTION The present invention discloses novel compounds with great affinity to the α2δ subunit of voltage-gated calcium channels, more specifically to the α2δ-1 subunit, and which in preferred embodiments also have inhibitory effect towards noradrenaline transporter (NET), thus resulting in a dual activity for treating pain and pain related disorders. The main object of the present invention is related to compounds of general formula (I):

wherein

n is 0 or 1 ;

Z is one of the following moieties

where

the dotted line represents an optional double bond;

R 4 and R 4 > independently represent a hydrogen atom; a halogen, a branched or unbranched Ci-6 alkyl radical; or a branched or unbranched Ci-6 haloalkyl;

A, B, D and E independently from one another represents -N-; -NH-; -CH-; -CH2- or -C(O)-;

with the proviso that at least one of A, B, D or E is -N- or -NH-; and

with the proviso that when n is 0, Z does not represent a quinoline or isoquinoline;

Ri is selected from an optionally substituted 5 to 9 membered aryl group; an optionally substituted 5 to 9 membered heteroaryl group having at least one heteroatom selected from the group of N, O or S; or an optionally substituted C3-9 heterocycoalkyl group having at least one heteroatom selected from the group of N, O or S;

R2 and R3 independently represent a hydrogen atom or a branched or unbranched C1-6 alkyl radical; or a pharmaceutically acceptable salt, isomer, prodrug or solvate thereof.

It is also an object of the invention different processes for the preparation of compounds of formula (I). Another object of the invention refers to the use of such compounds of general formula (I) for the treatment and/or prophylaxis of α2δ-1 mediated disorders and more preferably for the treatment and/or prophylaxis of disorders mediated by the α2δ-1 subunit of voltage-gated calcium channels and/or noradrenaline transporter (NET). The compounds of the present invention are particularly suited for the treatment of pain, specially neuropathic pain, and pain related or pain derived conditions.

It is also an object of the invention pharmaceutical compositions comprising one or more compounds of general formula (I) with at least one pharmaceutically acceptable excipient. The pharmaceutical compositions in accordance with the invention can be adapted in order to be administered by any route of administration, be it orally or parenterally, such as pulmonarily, nasally, rectally and/or intravenously. Therefore, the formulation in accordance with the invention may be adapted for topical or systemic application, particularly for dermal, subcutaneous, intramuscular, intra-articular, intraperitoneal, pulmonary, buccal, sublingual, nasal, percutaneous, vaginal, oral or parenteral application.

DETAILED DESCRIPTION OF THE INVENTION

The invention first relates to compounds of general formula (I)

(I)

wherein

n is 0 or 1 ;

Z is one of the following moieties

where

the dotted line represents an optional double bond;

R 4 and R 4 > independently represent a hydrogen atom; a halogen, a branched or unbranched Ci-6 alkyl radical; or a branched or unbranched Ci-6 haloalkyl;

A, B, D and E independently from one another represents -N-; -NH-; -CH-; - CH 2 - or -C(O)-;

with the proviso that at least one of A, B, D or E is -N- or -NH-; and

with the proviso that when n is 0, Z does not represent a quinoline or isoquinoline;

Ri is selected from an optionally substituted 5 to 9 membered aryl group, an optionally substituted 5 to 9 membered heteroaryl group having at least one heteroatom selected from the group of N, O or S; or an optionally substituted C3-9 heterocycoalkyl group having at least one heteroatom selected from the group of N, O or S;

R2 and R3 independently represent a hydrogen atom or a branched or unbranched C1-6 alkyl radical;

or a pharmaceutically acceptable salt, isomer, prodrug or solvate thereof.

Unless otherwise stated, the compounds of the invention are also meant to include isotopically-labelled forms i.e. 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 at least one hydrogen atom by a deuterium or tritium, or the replacement of at least one carbon by 13 C- or 14 C-enriched carbon, or the replacement of at least one nitrogen by 15 N-enriched nitrogen are within the scope of this invention. The compounds of formula (I) or their salts or solvates are preferably in pharmaceutically acceptable or substantially pure form. By pharmaceutically acceptable 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, solvates or prodrugs.

"Halogen" or "halo" as referred in the present invention represent fluorine, chlorine, bromine or iodine. When the term "halo" is combined with other substituents, such as for instance "Ci-6 haloalkyl" or "Ci-6 haloalkoxy" it means that the alkyl or alkoxy radical can respectively contain at least one halogen atom.

A leaving group is a group that in a heterolytic bond cleavage keeps the electron pair of the bond. Suitable leaving groups are well known in the art and include CI, Br, I and -O- SO2 , wherein R is F, Ci-4-alkyl, Ci-4-haloalkyl, or optionally substituted phenyl. The preferred leaving groups are CI, Br, I, tosylate, mesylate, nosylate, triflate, nonaflate and fluorosulphonate.

"C1-6 alkyl", as referred to in the present invention, are saturated aliphatic radicals. They may be linear or branched and are optionally substituted. Ci-6-alkyl as expressed in the present invention means an alkyl radical of 1 , 2, 3, 4, 5 or 6 carbon atoms. Preferred alkyl radicals according to the present invention include but are not restricted to methyl, ethyl, propyl, n-propyl, isopropyl, butyl, n-butyl, tert-butyl, isobutyl, sec-butyl, 1 - methylpropyl, 2-methylpropyl, 1 ,1 -dimethylethyl, pentyl, n-pentyl, 1 ,1 -dimethylpropyl, 1 ,2-dimethylpropyl, 2,2-dimethylpropyl, hexyl, 1 -methylpentyl. The most preferred alkyl radical are C1-4 alkyl, such as methyl, ethyl, propyl, n-propyl, isopropyl, butyl, n-butyl, tert- butyl, isobutyl, sec-butyl, 1 -methylpropyl, 2-methylpropyl or 1 ,1 -dimethylethyl. Alkyl radicals, as defined in the present invention, are optionally mono-or polysubstituted by substitutents independently selected from a halogen, Ci-6-alkoxy, Ci-6-alkyl, C1-6- haloalkoxy, Ci-6-haloalkyl, trihaloalkyl or a hydroxyl group.

"C3-6 Cycloalkyl" as referred to in the present invention, is understood as meaning saturated and unsaturated (but not aromatic), cyclic hydrocarbons having from 3 to 6 carbon atoms which can optionally be unsubstituted, mono- or polysubstituted. Examples for cycloalkyl radical preferably include but are not restricted to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl. Cycloalkyl radicals, as defined in the present invention, are optionally mono-or polysubstituted by substitutents independently selected from a halogen, Ci-6-alkyl, Ci-6-alkoxy, Ci-6-haloalkoxy, Ci-6-haloalkyl, trihaloalkyl or a hydroxyl group.

A cycloalkylalkyl group/radical C1-6, as defined in the present invention, comprises a linear or branched, optionally at least mono-substituted alkyl chain of 1 to 6 atoms which is bonded to a cycloalklyl group, as defined above. The cycloalkylalkyl radical is bonded to the molecule through the alkyl chain. A preferred cycloalkylalkyl group/radical is a cyclopropylmethyl group or a cyclopentylpropyl group, wherein the alkyl chain is optionally branched or substituted. Preferred substituents for cycloalkylalkyl group/radical, according to the present invention, are independently selected from a halogen, Ci-6-alkyl, Ci-6-alkoxy, Ci-6-haloalkoxy, Ci-6-haloalkyl, trihaloalkyl or a hydroxyl group.

"Heterocycloalkyi" as referred to in the present invention, are understood as meaning saturated and unsaturated (but not aromatic), generally 5 or 6 membered cyclic hydrocarbons which can optionally be unsubstituted, mono- or polysubstituted and which have at least one heteroatom in their structure selected from N, O or S. Examples for heterocycloalkyi radical preferably include but are not restricted to pyrroline, pyrrolidine, pyrazoline, aziridine, azetidine, tetrahydropyrrole, oxirane, oxetane, dioxetane, tetrahydropyran, tetrahydrofuran, dioxane, dioxolane, oxazolidine, piperidine, piperazine, morpholine, azepane or diazepane. Heterocycloalkyi radicals, as defined in the present invention, are optionally mono-or polysubstituted by substitutents independently selected from a halogen, Ci-6-alkyl, Ci-6-alkoxy, Ci-6-haloalkoxy, Ci-6- haloalkyl, trihaloalkyl or a hydroxyl group. More preferably heterocycloalkyi in the context of the present invention are 5 or 6-membered ring systems optionally at least monosubstituted.

A heterocycloalkylalkyl group/radical C1-6, as defined in the present invention, comprises a linear or branched, optionally at least mono-substituted alkyl chain of 1 to 6 atoms which is bonded to a cycloalklyl group, as defined above. The heterocycloalkylalkyl radical is bonded to the molecule through the alkyl chain. A preferred heterocycloalkylalkyl group/radical is a piperidinethyl group or a piperazinylmethyl group, wherein the alkyl chain is optionally branched or substituted. Preferred substituents for cycloalkylalkyl group/radical, according to the present invention, are independently selected from a halogen, Ci-6-alkyl, Ci-6-alkoxy, Ci-6-haloalkoxy, Ci-6-haloalkyl, trihaloalkyl or a hydroxyl group. "Aryl" as referred to in the present invention, is understood as meaning ring systems with at least one aromatic ring but without heteroatoms even in only one of the rings. These aryl radicals may optionally be mono-or polysubstituted by substitutents independently selected from a halogen, Ci-6-alkyl, Ci-6-alkoxy, Ci-6-haloalkoxy, Ci-6-haloalkyl, nitro or a hydroxyl group. Preferred examples of aryl radicals include but are not restricted to phenyl, naphthyl, fluoranthenyl, fluorenyl, tetralinyl, indanyl or anthracenyl radicals, which may optionally be mono- or polysubstituted, if not defined otherwise. More preferably aryl in the context of the present invention are 5 or 6-membered ring systems optionally at least monosubstituted. An arylalkyi radical C1-6, as defined in the present invention, comprises a linear or branched, optionally at least mono-substituted alkyl chain of 1 to 6 carbon atoms which is bonded to an aryl group, as defined above. The arylalkyi radical is bonded to the molecule through the alkyl chain. A preferred arylalkyi radical is a benzyl group or a phenetyl group, wherein the alkyl chain is optionally branched or substituted. Preferred substituents for arylalkyi radicals, according to the present invention, are independently selected from a halogen, Ci-6-alkyl, Ci-6-alkoxy, Ci-6-haloalkoxy, Ci-6-haloalkyl, trihaloalkyl or a hydroxyl group.

"Heteroaryl" as referred to in the present invention, is understood as meaning heterocyclic ring systems which have at least one aromatic ring and may optionally contain one or more heteroatoms from the group consisting of N, O or S and may optionally be mono-or polysubstituted by substituents independently selected from a halogen, Ci-6-alkyl, Ci-6-alkoxy, Ci-6-haloalkoxy, Ci-6-haloalkyl trihaloalkyl or a hydroxyl group. Preferred examples of heteroaryls include but are not restricted to furan, benzofuran, pyrrole, pyridine, pyrimidine, pyridazine, pyrazine, tiophene, quinoline, isoquinoline, phthalazine, triazole, pyrazole, isoxazole, indole, benzotriazole, benzodioxolane, benzodioxane, benzimidazole, carbazole and quinazoline. More preferably heteroaryl in the context of the present invention are 5 or 6-membered ring systems optionally at least monosubstituted.

Heteroarylalkyl group/radical Ci-6 as defined in the present invention, comprises a linear or branched, optionally at least mono-substituted alkyl chain of 1 to 6 carbon atoms which is bonded to an heteroaryl group, as defined above. The heteroarylalkyl radical is bonded to the molecule through the alkyl chain. A preferred heteroarylalkyl radical is a piridinylmethyl group, wherein the alkyl chain is optionally branched or substituted. Preferred substituents for heteroarylalkyl radicals, according to the present invention, are independently selected from a halogen, Ci-6-alkyl, Ci-6-alkoxy, Ci-6-haloalkoxy, Ci-6- haloalkyl, trihaloalkyl or a hydroxyl group.

"Heterocyclic ring" or "heterocyclic system", as defined in the present invention, comprise any saturated, unsaturated or aromatic carbocyclic ring systems which are optionally at least mono-substituted and which contain at least one heteroatom as ring member. Preferred heteroatoms for these heterocyclyl groups are N, S or O. Preferred substituents for heterocyclyl radicals, according to the present invention, are F, CI, Br, I, Nhb, SH, OH, SO2, CF3, carboxy, amido, cyano, carbamyl, nitro, phenyl, benzyl, - SO2NH2, C1-6 alkyl and/or Ci-e-alkoxy.

The term "C1-3 alkylene" is understood as meaning a divalent alkyl group like -CH2- or - CH2-CH2- or -CH2-CH2-CH2-. An "alkylene" may also be unsaturated

The term "condensed" according to the present invention means that a ring or ring- system is attached to another ring or ring-system, whereby the terms "annulated" or "annelated" are also used by those skilled in the art to designate this kind of attachment.

The term "ring system" according to the present invention refers to ring systems comprising saturated, unsaturated or aromatic carbocyclic ring systems which contain optionally at least one heteroatom as ring member and which are optionally at least mono-substituted. Said ring systems may be condensed to other carbocyclic ring systems such as aryl groups, heteroaryl groups, cycloalkyl groups, etc. The term "salt" is to be understood as meaning any form of the active compound according to the invention in which this 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 which are complexed via ionic interactions. The definition particularly includes physiologically acceptable salts, this term must be understood as equivalent to "pharmacologically acceptable salts". The term "pharmaceutically acceptable salts" in the context of this invention means any salt that is tolerated physiologically (normally meaning that it is not toxic, particularly as a result of the counter-ion) when used in an appropriate manner for a treatment, particularly applied or used in humans and/or mammals. These physiologically acceptable salts may be formed with cations or bases and, in the context of this invention, are understood to be salts formed by at least one compound used in accordance with the invention - normally an acid (deprotonated) - such as an anion and at least one physiologically tolerated cation, preferably inorganic, particularly when used on humans and/or mammals. Salts with alkali and alkali earth metals are particularly preferred, as well as those formed with ammonium cations (NhV). Preferred salts are those formed with (mono) or (di)sodium, (mono) or (di)potassium, magnesium or calcium. These physiologically acceptable salts may also be formed with anions or acids and, in the context of this invention, are understood as being salts formed by at least one compound used in accordance with the invention - normally protonated, for example in nitrogen - such as a cation and at least one physiologically tolerated anion, particularly when used on humans and/or mammals. This definition specifically includes in the context of this invention a salt formed by a physiologically tolerated acid, i.e. salts of a specific active compound with physiologically tolerated organic or inorganic acids - particularly when used on humans and/or mammals. Examples of this type of salts are those formed with hydrochloric acid, hydrobromic acid, sulphuric 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 term "solvate" 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 including hydrates and alcoholates, e.g. methanolate.

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 compounds of the invention: 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 a prodrug of a compound of formula (I) is within the scope of the invention. Particularly favored prodrugs are those that increase the bioavailability of the compounds of this invention when such compounds are administered to a patient (e.g., by allowing an orally administered compound to be more readily absorbed into the blood) or which enhance delivery of the parent compound to a biological compartment (e.g., the brain or lymphatic system) relative to the parent species.

In a particular and preferred embodiment of the invention, Ri represents a benzene, a thiophene, a thiazole, a pyridine or a tetrahydropyran. These groups may optionally be substituted by at least one substituent selected from halogen, Ci-6-alkoxy, Ci-6- haloalkoxy, Ci-6-haloalkyl or a hydroxyl group. The benzene, thiophene, thiazole, pyridine or tetrahydropyran group can be attached to the main structure through different points of attachment. For instance, when Ri represents thiophene this might be a 2- thiophene or 3-thiophene, when it represents thiazole it may represent a 2-thiazole, a 4- thiazole or a 5-thiazole, when it represents a pyridine it may represent a 2-pyridine, 3- pyridine or 4-pyridine or when it represents tetrahydropyran it may represent 2- tetrahydropyran, 3-tetrahydropyran or 4-tetrahydropyran. In a particularly prefered embodiment Ri represents a benzene or a thiophene, preferably unsubstituted although optionally substituted by at least one substituent selected from halogen, Ci-6 alkyl, Ci-6-alkoxy, Ci-6-haloalkoxy, Ci-6-haloalkyl or a hydroxyl group. In another particular and preferred embodiment of the invention, Z is selected from:

which can be optionally substituted by at least one substituent selected from a halogen, a branched or unbranched Ci-6 alkyl radical or by a branched or unbranched Ci-6 haloalkyl.

In a still more particularly preferred embodiment Z is selected from:

where R 4 represents a branched or unbranched Ci-6 alkyl radical or a branched or unbranched Ci-6 haloalkyl, and R 4 ' represents a hydrogen atom; a halogen, a branched or unbranched C1-6 alkyl radical; or a branched or unbranched Ci-6 haloalkyl.

Another particular embodiment of the invention is that where Z is represented by:

where A, B, D, E and R 4 are as defined before. A preferred embodiment of the invention is represented by compounds of formula (I):

(I)

wherein

n is 0 or 1 ;

Z is selected from

wherein R 4 represents a hydrogen atom; a branched or unbranched Ci-6 alkyl radical; or a branched or unbranched Ci-6 haloalkyl and R 4 ' represents a hydrogen atom; a halogen, a branched or unbranched Ci-6 alkyl radical; or a branched or unbranched Ci-6 haloalkyl.

Ri represents a benzene; a thiophene; a thiazole; a pyridine or a tetrahydropyran, preferably a benzene or a thiophene, all of them optionally substituted by at least one substituent selected from selected from halogen, Ci-6 alkyl, Ci-6-alkoxy, Ci-6-haloalkoxy, Ci-6-haloalkyl or a hydroxyl group, and

R2 and R3 independently represent a hydrogen atom or a branched or unbranched C1-6 alkyl radical; or a pharmaceutically acceptable salt, isomer, prodrug or solvate thereof.

Another preferred embodiment of the invention is represented by compounds of formula (I):

(I)

wherein

n is 0 or 1 ;

Z is selected from

wherein R 4 represents a hydrogen atom; a branched or unbranched Ci-6 alkyl radical; or a branched or unbranched Ci-6 haloalkyl and R 4 ' represents a hydrogen atom;

Ri represents a benzene; a thiophene; a thiazole; a pyridine or a tetrahydropyran, preferably a benzene or a thiophene, all of them optionally substituted by at least one substituent selected from selected from halogen, Ci-6 alkyl, Ci-6-alkoxy, Ci-6-haloalkoxy, Ci-6-haloalkyl or a hydroxyl group, and

R2 and R3 independently represent a hydrogen atom or a branched or unbranched C1-6 alkyl radical; or a pharmaceutically acceptable salt, isomer, prodrug or solvate thereof. Another particular embodiment of compounds of general formula (I) is represented by compounds of general formula (la):

wherein Ri , R2, R3 and Z have the same meaning as defined before.

Another particular embodiment of of compounds of general formula (I) is represented by compounds of general formula (lb):

(lb)

wherein Ri , R2, R3 and Z have the same meaning as defined before.

Still more particular embodiments falling within general formula (la) and (lb) respectively are compounds of formula (Ia1 ), (Ia2), (Ib1 ) or (Ib2):

The compounds of the present invention represented by the above described formula (I), (la), (lb), (Ia1 ), (Ia2), (Ib1 ) or (Ib2) may include enantiomers depending on the presence of chiral centers or isomers depending on the presence of double bonds (e.g. Z, E). The single isomers, enantiomers or diastereoisomers and mixtures thereof fall within the scope of the present invention.

Among all the compounds described in the general formula (I), the following compounds are preferred for showing an intense inhibitory effect towards subunit α2δ-1 of voltage- gated calcium channels (VGCC):

[1 ] /V-methyl-3-((1 -methyl-1 ,2,3,4-tetrahydroquinolin-5-yl)oxy)-3-(thiophen-2- yl)propan-1 -amine;

[2] /V-methyl-3-((2-methyl-1 ,2,3,4-tetrahydroisoquinolin-5-yl)oxy)-3-(thiophen-2- yl)propan-1 -amine;

[3] 3-((2-(2,2-Difluoroethyl)-1 ,2,3,4-tetrahydroisoquinolin-5-yl)oxy)-/V-methyl-3- (thiophen-2-yl)propan-1 -amine;

[4] (R)-/V-methyl-3-(phthalazin-5-yloxy)-3-(thiophen-2-yl)propan -1 -amine;

[5] (S)-/V-methyl-3-(phthalazin-5-yloxy)-3-(thiophen-2-yl)propan -1 -amine;

[6] /V-methyl-3-(phthalazin-5-yloxy)-3-(thiophen-2-yl)propan-1 -amine; [7] 3-Methyl-5-(3-(methylamino)-1 -(thiophen-2-yl)propoxy)quinazolin-4(3/-/)-one;

[8] 3-Methyl-8-(3-(methylamino)-1 -(thiophen-2-yl)propoxy)quinazolin-4(3/-/)-one;

[9] 2-Methyl-5-(3-(methylamino)-1 -(thiophen-2-yl)propoxy)isoquinolin-1 (2/-/)-one;

[10] /V-methyl-3-(quinazolin-5-yloxy)-3-(thiophen-2-yl)propan-1 -amine;

[1 1 ] /V-ethyl-3-phenyl-3-(phthalazin-5-yloxy)propan-1 -amine;

[12] /V-ethyl-3-(phthalazin-5-yloxy)-3-(thiophen-2-yl)propan-1 -amine;

[13] (R)-/V-ethyl-3-phenyl-3-(phthalazin-5-yloxy)propan-1 -amine;

[14] (R)-/V-ethyl-3-(phthalazin-5-yloxy)-3-(thiophen-2-yl)propan- 1 -amine;

[15] (S)-3-methyl-5-(3-(methylamino)-1 -(thiophen-2-yl)propoxy)quinazolin-

4(3H)-one;

[16] (R)-3-methyl-5-(3-(methylamino)-1 -(thiophen-2-yl)propoxy)quinazolin- 4(3H)-one;

[17] 3-(lsoquinolin-5-ylmethoxy)-/V-methyl-3-phenylpropan-1 -amine;

[18] 3-(lsoquinolin-8-ylmethoxy)-/V-methyl-3-phenylpropan-1 -amine;

[19] /V-methyl-3-phenyl-3-(quinolin-5-ylmethoxy)propan-1 -amine;

[20] 3-(lsoquinolin-8-ylmethoxy)-/V-methyl-3-(thiophen-2-yl)propa n-1 -amine;

[21 ] 3-(lsoquinolin-5-ylmethoxy)-/V-methyl-3-(thiophen-2-yl)propa n-1 -amine;

[22] /V-methyl-3-(quinolin-5-ylmethoxy)-3-(thiophen-2-yl)propan-1 -amine;

[23] fS)-/V-methyl-3-((1 -methyl-1 ,2,3,4-tetrahydroquinolin-5-yl)oxy)-3-(thiophen-

2-yl)propan-1 -amine;

[24] (R)-/V-methyl-3-((1 -methyl-1 ,2,3,4-tetrahydroquinolin-5-yl)oxy)-3-(thiophen- 2-yl)propan-1 -amine;

[25] (S)-/V-methyl-3-((2-methyl-1 ! 2,3,4-tetrahydroisoquinolin-5-yl)oxy)-3- (thiophen-2-yl)propan-1 -amine;

[26] (R)-/V-methyl-3-((2-methyl-1 ! 2,3,4-tetrahydroisoquinolin-5-yl)oxy)-3- (thiophen-2-yl)propan-1 -amine;

[27] /V-ethyl-3-((2-methyl-1 ! 2,3,4-tetrahydroisoquinolin-5-yl)oxy)-3-(thiophen-2- yl)propan-1 -amine;

[28] (S)-1 -methyl-5-(3-(methylamino)-1 -(thiophen-2-yl)propoxy)quinolin-2(1 H)- one;

[29] (R)-1 -methyl-5-(3-(methylamino)-1 -(thiophen-2-yl)propoxy)quinolin-2(1 H)- one;

[30] (R)-5-(3-(ethylamino)-1 -(thiophen-2-yl)propoxy)-3-methylquinazolin-4(3/-/)- one;

[31 ] (R)-5-(3-(ethylamino)-1 -(thiophen-2-yl)propoxy)-2-methylisoquinolin-1 (2/-/)- one; [32] (R)-8-(3-(ethylamino)-1 -(thiophen-2-yl)propoxy)-3-methylquinazolin-4(3 -/)- one;

[33] (S)-/V-methyl-3-(quinazolin-5-yloxy)-3-(thiophen-2-yl)propan -1 -amine;

[34] (S)-8-(3-(ethylamino)-1 -(thiophen-2-yl)propoxy)-3-methylquinazolin-4(3/-/)- one;

[35] (S)-5-(3-(ethylamino)-1 -(thiophen-2-yl)propoxy)-2-methylisoquinolin-1 (2/-/)- one;

[36] (S)-5-(3-(ethylamino)-1 -(thiophen-2-yl)propoxy)-3-methylquinazolin-4(3/-/)- one;

[37] (R)-/V-methyl-3-(quinazolin-5-yloxy)-3-(thiophen-2-yl)propan -1 -amine;

[38] (S)-/V-ethyl-3-(quinazolin-5-yloxy)-3-(thiophen-2-yl)propan- 1 -amine;

[39] (R)-/V-ethyl-3-(quinazolin-5-yloxy)-3-(thiophen-2-yl)propan- 1 -amine;

[40] (R)-/V-ethyl-3-(quinazolin-5-yloxy)-3-(thiophen-3-yl)propan- 1 -amine;

[41 ] (R)-8-(3-(ethylamino)-1 -(thiophen-3-yl)propoxy)-3-methylquinazolin-4(3/-/)- one;

[42] (R)-5-(3-(ethylamino)-1 -(thiophen-3-yl)propoxy)-2-methylisoquinolin-1 (2/-/)- one;

[43] (R)-3-methyl-5-(3-(methylamino)-1 -(thiophen-3-yl)propoxy)quinazolin- 4(3H)-one;

[44] (/?)-8-(3-(ethylamino)-1 -(thiophen-2-yl)propoxy)-3,6-dimethylquinazolin- 4(3H)-one and

[45] (R)-7-fluoro-3-methyl-5-(3-(methylamino)-1 -(thiophen-2- yl)propoxy)quinazolin-4(3/-/)-one; or a pharmaceutically acceptable salt, prodrug or solvate thereof.

Among compounds of general formula (I) some subgroups of compounds have shown in addition a dual affinity towards subunit α2δ-1 of voltage-gated calcium channels (VGCC) and the noradrenaline transporter (NET). These compounds having dual affinity represent the preferred embodiments of the invention and are represented by formula (lc):

(lc) wherein Ri, R2, R3, A, B and n are as defined before with the proviso that at least one of A or B represents a -N(R)- where R can be a hydrogen or a branched or unbranched Ci- 6 alkyl radical; or a branched or unbranched Ci-6 haloalkyl.

The preferred compounds of the invention showing dual inhibitory effect towards subunit α2δ-1 of voltage-gated calcium channels (VGCC) and noradrenaline transporter (NET) are selected from the following group: [1] /V-methyl-3-((1 -methyl-1 ,2,3,4-tetrahydroquinolin-5-yl)oxy)-3-(thiophen-2-yl)propan- 1 -amine and

[3] 3-((2-(2 ! 2-Difluoroethyl)-1 ! 2 ! 3 ! 4-tetrahydroisoquinolin-5-yl)oxy)-/V-methyl-3- (thiophen-2-yl)propan-1 -amine; or a pharmaceutically acceptable salt, prodrug or solvate thereof.

In another aspect, the invention refers to the processes for obtaining the compounds of general formula (I). Several procedures have been developed for obtaining all the compounds of the invention, and the procedures will be explained below in methods A and B.

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. If there are chiral centers the compounds may be prepared in racemic form, or individual enantiomers may be prepared either by enantiospecific synthesis or by resolution. METHOD A

Method A represents a first process for synthesizing compounds according to general formula (I). Method A allows for the preparation of compounds of general formula (la) that is compounds of formula (I) where n is 0.

In this sense, a process is described for the preparation of a compound of general formula (la):

(la) comprising:

A) the reaction of a compound of formula (II):

(II) with a compound of formula (Ilia) or (1Mb):

Z-OH or Z-X

(Ilia) (1Mb) wherein Ri , R2, R3 and Z are as defined before and X represents a halogen, or B) the reaction of a compound of formula (Va):

(Va)

with a compound of formula (VI):

HNR 2 R 3

(VI) wherein Ri , R2, R3 and Z are as defined before and LG represents a leaving group, such as chloro, bromo, iodo, mesylate, tosylate, nosylate or triflate.

Different reaction conditions are applied when the compounds of formula (la) are synthesized through the reaction of a compound of formula (II) with either a compound of formula (Ilia) or a compound of formula (lllb): a) When a hydroxy compound of formula (Ilia) is used, the reaction is carried out under conventional Mitsunobu conditions by treating an alcohol of formula (II) with a compound of formula (Ilia) preferably in the presence of an azo compound such as 1 ,1 '-(azodicarbonyl)dipiperidine (ADDP), diisopropylazodicarboxylate (DIAD) or diethyl azodicarboxylate (DEAD) and a phosphine such as tributylphosphine or triphenylphoshine. The Mitsunobu reaction is carried out in a suitable solvent, such as toluene or tetrahydrofuran; 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. b) When a halo compound of formula (lllb) is used, the reaction is carried out under conventional aromatic nucleophilic substitution conditions by treating an alcohol of formula (II) with a compound of formula (lllb) wherein X represents halogen (preferably fluoro), in the presence of a strong base such as sodium hydride or potassium ie f-butoxide. The reaction is carried out in a suitable solvent, such as a polar aprotic solvent, preferably dimethylformamide, dimethylacetamide or dimethylsulfoxide; 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. Alternatively, when X is bromo or iodo, the compound of formula (lllb) can be introduced under cross- coupling conditions, using a Pd or Cu catalyst and a suitable ligand. Alternatively and, as explained above, a compound of formula (la) can be obtained by reaction of a compound of formula (Va) with an amine of formula (VI). The alkylation reaction is carried out in a suitable solvent, such as ethanol, dimethylformamide, dimethylsulfoxide or acetonitrile, preferably ethanol; optionally in the presence of a base such as K2CO3 or triethylamine; 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. Additionally, an activating agent such as sodium iodide or potassium iodide can be used.

METHOD B

Method B represents a process for synthesizing compounds according to general formula (lb), namely compounds of general formula (I) where n is 1 .

In this sense, a process is described for the preparation of a compound of general formula (lb):

comprising the reaction between a compound of formula (II):

and a compound of formula (III c): wherein Ri , F¾, R3 and Z are as defined before and LG represents a leaving group, such as chloro, bromo, iodo, mesylate, tosylate, nosylate or triflate.

The reaction between a compound of formula (I I) with an alkylating agent of formula (I II c) is preferably carried out in the presence of a strong base such as sodium hydride or potassium ie f-butoxide. The alkylation reaction is carried out in a suitable solvent, such as tetrahydrofuran or dimethylformamide, 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. Additionally, an activating agent such as sodium iodide or a phase transfer catalyst such as tetrabutylammonium iodide can be used.

Both methods A and B use compound (I I) as starting material. The amino group can be either introduced in compound of formula (I I) at an early stage, that is, before the reaction for producing compounds of formula (la) and (lb) or later in the synthesis by reaction of a precursor compound of formula (ll)-LG or (Va) wherein LG represents a leaving group (such as chloro, bromo, iodo, mesylate, tosylate, nosylate or triflate) with an amine of formula (VI) to render a compound of formula (II) or (la), respectively, as shown in Scheme 1 below. The alkylation reaction is carried out in a suitable solvent, such as ethanol, dimethylformamide, dimethylsulfoxide or acetonitrile, preferably ethanol; optionally in the presence of a base such as K2CO3 or triethylamine; 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. Additionally, an activating agent such as sodium iodide or potassium iodide can be used.

The different reactions of methods A and B as well as the reactions for preparing the intermediate compounds for such reactions are depicted in scheme 1 :

Scheme 1

(II) Y=NR 2 R 3 ^ HN R R

H O Y (ll)-LG Y=LG J ΗΝ * 2 * 3

(la) n=0 Y=NR 2 R 3 (lb) n=1 Y=NR 2 R 3

H N R 2 R 3 ( VA ) N =0 Y =| _ ( 3

(VI)

wherein Z can be

The intermediate compound (II) which is the basic reagent for producing compounds of formula (I), according to methods A and B, can be prepared, as shown in scheme 1 above by the reduction of a keto compound of formula (IV) following conventional procedures described in the literature. As a way of example, the reduction can be performed using a hydride source such as sodium borohydride in a suitable solvent such as methanol, ethanol or tetrahydrofuran or lithium aluminium hydride in a suitable solvent such as tetrahydrofuran or diethyl ether, at a suitable temperature, preferably comprised between 0 °C and room temperature.

Alternatively, the reduction can be carried out by hydrogenation under hydrogen atmosphere and metal catalysis, preferably by the use of palladium over charcoal or Nickel-Raney as catalysts, in a suitable solvent such as methanol, ethanol or ethyl acetate. In addition, the reduction can be performed under asymmetric hydrogenation conditions using a rhodium catalyst to render chiral compounds of formula II in enantiopure form, following procedures described in the literature (i.e. Angew. Chem. Int. Ed. 2004, 43, 2816; Angew. Chem. Int. Ed. 2005, 44, 1687; Angew. Chem. Int. Ed. 2015, 54, 2260) A compound of formula (Va) can be synthesized from a compound of formula (ll)-LG by reaction with a compound of formula (Ilia), following the conditions described above for the preparation of a compound of formula (la) from a compound of formula (II) and a compound of formula (Ilia).

The compounds of general formula (Ilia), (1Mb), (III c) and (VI) are commercially available or can be prepared by conventional methods described in the bibliography.

The compounds of formula (ll)-LG are commercially available or can be obtained from a compound of formula (IV)-LG following the reduction conditions described above, (Step 1 ), preferably using a hydride source. In addition, the reduction can be performed under asymmetric conditions described in the literature to render chiral compounds of formula (ll-LG) in enantiopure form. As a way of example, the chiral reduction can be performed using a hydride source such as borane-tetrahydrofuran complex or borane-dimethyl sulfide complex, in the presence of a Corey-Bakshi-Shibata oxazaborolidine catalyst, in a suitable solvent such as tetrahydrofuran or toluene, at a suitable temperature, preferably comprised between 0 °C and room temperature.

In turn, compounds of formula (IV) and (IV)-LG are commercially available or can be synthesized following procedures described in the literature. As a way of example, some routes of synthesis are described in Schemes 2 and 3 below. In addition, a compound of formula (IV) can be prepared from a compound of formula (IV)-LG and an amine of formula (VI) following the conditions described above for the synthesis of a compound of formula (la) from a compound of formula (Va).

The preparation of compounds of general formula (IV) can be performed following several methods described in the literature. As a way of example, two routes of synthesis are described in Scheme 2:

Scheme 2:

wherein Ri , R2 and R3 have the meanings as defined above for a compound of formula

(I)- Following Route A, the treatment of a compound of formula (VIII) with a strong base such as butyl lithium to generate the corresponding organometallic reagent and subsequent condensation with a Weinreb amide of formula (VII), in a suitable solvent such as tetrahydrofuran, renders a compound of formula (IV). Alternatively, the compounds of formula (IV) can be prepared through a Mannich reaction by condensation of an acetyl compound of formula (IX) with an amine of formula (VI) and a formaldehyde source such as paraformaldehyde, preferably in the presence of an acid such as hydrochloric acid, in a suitable solvent such as ethanol or isopropanol, at a suitable temperature, preferably heating.

The preparation of compounds of general formula (IV)-LG can be performed following several methods described in the literature. As a way of example, a route of synthesis is described in Scheme 3:

Scheme 3:

(X) (XI) (IV)-LG wherein Ri has the meaning as defined above for a compound of formula (I), LG represents a leaving group (preferably chloro, bromo or iodo) and X represents halogen (preferably chloro or bromo). The Friedel-Crafts acylation of an heteroaryl compound of formula (X) with an acid halide of formula (XI) in the presence of a Lewis acid such as aluminum trichloride renders a compound of formula (IV)-LG. The reaction is carried out in a suitable solvent, such as dichloromethane or dichloroethane; at a suitable temperature comprised between 0 °C and the reflux temperature.

The compounds of general formula (VI), (VII), (VIII), (IX), (X) and (XI) are commercially available or can be prepared by conventional methods described in the bibliography.

Moreover, 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.

In some of the processes described above it may be necessary to protect the reactive or labile groups present with suitable protecting groups, such as for example Boc (tert- butoxycarbonyl), Teoc (2-(trimethylsilyl)ethoxycarbonyl) or benzyl for the protection of amino groups. The procedures for the introduction and removal of these protecting groups are well known in the art and can be found thoroughly described in the literature.

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.

Compounds of general formula (I) for which R3 contains a Protecting Group (PG), such as Boc or 2-(trimethylsilyl)ethylcarbamate), can be used as intermediates useful for the preparation of other compounds of general formula (I) as defined above.

Another particular aspect is represented by the intermediate compounds used for the preparation of compounds of general formula (I).

In a particular embodiment, these intermediate compounds are selected from:

• 8-Fluoro-3-methylquinazolin-4(3/-/)-one; • 5-Fluoro-2-methylisoquinolin-1 (2/-/)-one;

• 2-(2,2-Difluoroethyl)-1 ,2,3,4-tetrahydroisoquinolin-5-ol;

• (R)-3-(methylamino)-1 -(thiophen-2-yl)propan-1 -ol;

• (7?)-3-(ethylamino)-1 -(thiophen-2-yl)propan-1 -ol;

· 3-(Ethylamino)-1 -(thiophen-2-yl)propan-1 -ol;

• 3-(Ethylamino)-1 -phenylpropan-1 -ol;

• (R)-3-(ethylamino)-1 -phenylpropan-1 -ol;

• (R)-3-(Methylamino)-1 -(thiophen-3-yl)propan-1 -ol;

• (S)-3-(ethylamino)-1 -(thiophen-2-yl)propan-1 -ol;

· (R)-3-(ethylamino)-1 -(thiophen-3-yl)propan-1 -ol;

• 8-Fluoro-3,6-dimethylquinazolin-4(3/-/)-one; and

• 5,7-Difluoro-3-methylquinazolin-4(3/-/)-one.

Turning to another aspect, the invention also relates to the therapeutic use of the compounds of general formula (I). As mentioned above, compounds of general formula (I) show a strong affinity to subunit α2δ and, more preferably, to α2δ-1 subunit of voltage-gated calcium channels. In a more preferred embodiment of the invention compounds of general formula (I) show a strong affinity both to subunit α2δ and, more preferably, to α2δ-1 subunit of voltage-gated calcium channels, as well as to noradrenaline transporter (NET) and can behave as agonists, antagonists, inverse agonists, partial antagonists or partial agonists thereof. Therefore, compounds of general formula (I) are useful as medicaments. They are suitable for the treatment and/or prophylaxis of diseases and/or disorders mediated by the subunit α2δ, especially α2δ-1 subunit of voltage-gated calcium channels and/or noradrenaline transporter (NET). In this sense, compounds of formula (I) are suitable for the treatment and/or prophylaxis of pain, depression anxiety and attention-deficit-/hyperactivity disorder (ADHD).

The compounds of formula (I) are especially suited for the treatment of pain, especially medium to severe pain, visceral pain, chronic pain, cancer pain, migraine, inflammatory pain, acute pain neuropathic pain, allodynia or hyperalgesia, including mechanical allodynia or thermal hyperalgesia. PAIN is defined by the International Association for the Study of Pain (IASP) as "an unpleasant sensory and emotional experience associated with actual or potential tissue damage, or described in terms of such damage (IASP, Classification of chronic pain, 2nd Edition, IASP Press (2002), 210). Even though pain is always subjective its causes or syndromes can be classified.

In a preferred embodiment compounds of the invention are used for the treatment and/or prophylaxis of allodynia and more specifically mechanical or thermal allodynia. In another preferred embodiment compounds of the invention are used for the treatment and/or prophylaxis of hyperalgesia.

In yet another preferred embodiment compounds of the invention are used for the treatment and/or prophylaxis of neuropathic pain and more specifically for the treatment and/or prophylaxis of hyperpathia.

A related aspect of the invention refers to the use of compounds of formula (I) for the manufacture of a medicament for the treatment and/or prophylaxis of disorders and diseases mediated by the subunit α2δ, especially α2δ-1 subunit of voltage-gated calcium channels and/or noradrenaline transporter (NET), as explained before.

Another related aspect of the invention refers to a method for the treatment and/or prophylaxis of disorders and diseases mediated by the subunit α2δ, especially α2δ-1 subunit of voltage-gated calcium channels and/or noradrenaline transporter (NET), as explained before comprising the administration of a therapeutically effective amount of a compound of general formula (I) to a subject in need thereof.

Another aspect of the invention is a pharmaceutical composition, which comprises at least a compound of general formula (I) or a pharmaceutically acceptable salt, prodrug, isomer or solvate thereof, and at least a pharmaceutically acceptable carrier, additive, adjuvant or vehicle.

The pharmaceutical composition of the invention can be formulated as a medicament in different pharmaceutical forms comprising at least a compound binding to the subunit α2δ, especially α2δ-1 subunit of voltage-gated calcium channels and/or noradrenaline transporter (NET) and optionally at least one further active substance and/or optionally at least one auxiliary substance.

Preferably, the composition is suitable for oral or parenteral administration, more preferably for oral, intravenous, intraperitoneal, intramuscular, subcutaneous, intrathekal, rectal, transdermal, transmucosal or nasal administration.

The composition of the invention can be formulated for oral administration in any form preferably selected from the group consisting of tablets, dragees, capsules, pills, chewing gums, powders, drops, gels, juices, syrups, solutions and suspensions. The composition of the present invention for oral administration may also be in the form of multiparticulates, preferably microparticles, microtablets, pellets or granules, optionally compressed into a tablet, filled into a capsule or suspended in a suitable liquid. Suitable liquids are known to those skilled in the art.

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. The daily dosage for humans and animals may vary depending on factors that have their basis in the respective species or other factors, such as age, sex, weight or degree of illness and so forth. The daily dosage for humans may preferably be in the range from 1 to 2000, preferably 1 to 1500, more preferably 1 to 1000 milligrams of active substance to be administered during one or several intakes per day.

The following examples are merely illustrative of certain embodiments of the invention and cannot be considered as restricting it in any way.

EXAMPLES

In the next preparation examples the synthesis of both intermediate derivatives as well as compounds according to the invention are disclosed.

The following abbreviations are used in the examples: ACN: acetonitrile

ADDP: 1 ,1 '-(azodicarbonyl)dipiperidine

Anh: anhydrous

Boc: ie f-butoxycarbonyl

Cone: concentrated

DCM: dichloromethane

DEA: diethylamine

DIPEA: Λ/,/V-diisopropylethylamine

DMA: dimethylacetamide

DME: 1 ,2-dimethoxyethane

DMF: dimethylformamide

Eq: equivalent/s

Et 2 0; diethyl ether

EtOAc; ethyl acetate

EtOH: ethanol

EX: example

h: hour/s HPLC: high performance liquid chromatography

INT: intermediate

2-Me-CBS-oxazaborolidine: 5,5-diphenyl-2-methyl-3,4-propano-1 ,3,2-oxazaborolidine (Corey-Bakshi-Shibata oxazaborolidine catalyst)

MeOH: methanol

MS: mass spectrometry

Min.: minutes

Quant: quantitative

Ret.: retention

r.t: room temperature

Sat: saturated

TFA: trifluoroacetic acid

THF: tetrahydrofuran

Wt: weight

The following methods were used to determine the HPLC-MS spectra: Method A

Column Xbridge C18 XP 30 x 4.6 mm, 2.5um

Temperature: 40°C

Flow: 2.0 mL/min

Gradient: NH4HCO3 pH 8 : ACN (95:5)— 0.5min— (95:5)— 6.5min— (0:100)— 1 min— (0:100)

Sample dissolved approx. 1 mg/ mL in NH4HCO3 PH 8/ ACN

Method B

Column: Gemini-NX 30 x 4.6 mm, 3um

Temperature: 40°C

Flow: 2.0 mL/min

Gradient: NH4HCO3 pH 8 : ACN (95:5)— 0.5min— (95:5)— 6.5min— (0:100)— 1 min— (0:100)

Sample dissolved approx. 1 mg/ mL in NH4HCO3 PH 8/ ACN Method C

Column: Kinetex EVO 50 x 4.6 mm, 2.6um

Temperature:40°C

Flow: 2.0 mL/min Gradient: NH4HCO3 pH 8 : ACN (95:5)— 0.5min— (95:5)— 6.5min— (0:100)— 1 min— (0:100)

Sample dissolved approx. 1 mg/ mL in NH4HCO3 PH 8/ ACN Method D

Column: Eclipse XDB-C18 4.6x150 mm, 5 μηι;

Flow: 1 mL/min

Gradient: H 2 0 (0.05% TFA): ACN (95:5)— 7min— (5:95)— 5min— (5:95) Method E

Column: Kinetex EVO 50 x 4.6 mm, 2.6um

Temperature:40°C

Flow: 1 .5 mL/min

Gradient: NH4HCO3 pH 8 : ACN (95:5)— 0.5min— (95:5)— 6.5min— (0:100)— 2min— (0:100)

Sample dissolved approx. 1 mg/ mL in NH4HCO3 PH 8/ ACN

Synthesis of Intermediates

Intermediate 1 : 8-Fluoro-3-methylquinazolin-4(3H)-

StepL 2-Amino-3-fluorobenzamide: To a solution of 2-amino-3-fluorobenzoic acid (1 g, 6. 5 mmol) in DMF (3 mL), 1 H-benzo[d][1 ,2,3]triazol-1 -ol (1 .13 g, 8.4 mmol), Λ/-(3- dimethylaminopropyl)-/V'-ethylcarbodiimide (1.2 g, 7.7 mmol), NH4CI (1.45 g, 21 .1 mmol) and DIPEA (4.7 mL, 27.1 mmol) were added. The reaction mixture was stirred at r.t. overnight. Water was then added and it was extracted with EtOAc. The combined organic phases were washed with brine, dried over MgSC and concentrated under vacuum. The crude product was slurried in DCM (6.5 mL). The solids were filtered, washed with cold DCM and dried under vacuum to afford the title compound (444 mg, 45% yield)

Step 2. Title compound: A mixture of the product obtained in Step 1 (444 mg, 2.8 mmol) and dimethylformamide dimethylacetal (1 g, 8.6 mmol) in DMF (14 mL) was heated at 150 °C in a sealed tube for 1 .5 h. The solvent was distilled off and the residue (which contained a mixture of the title compound and 8-fluoroquinazolin-4(3/-/)-one) was purified by flash chromatography, silica gel, gradient DCM to MeOH:DCM (1 :4) to give the title compound (104 mg, 20% yield).

Intermediate 2: 5-Fluoro-2-methylisoquinolin-1 (2H)-one

To a solution of 5-fluoroisoquinolin-1 (2/-/)-one (60 mg, 0.37 mmol) in DMF (2 ml_), NaH (60% dispersion in mineral oil, 19 mg, 0.48 mmol) was added. After stirring at r.t. for 1 h, iodomethane (0.028 ml_, 0.44 mmol) was added and the reaction mixture was stirred at r.t. overnight. Water was added and it was extracted with EtOAc. The combined organic phases were washed with water and brine, dried over MgSC>4 and concentrated under vacuum to afford the title compound (66 mg, quant, yield).

Intermediate 3: 2-(2,2-Difluoroethyl)-1 ,2,3,4-tetrahydroisoquinolin-5-ol

To a solution of 1 ,2,3,4-tetrahydroisoquinolin-5-ol (0.95 g, 2.39 mmol) and acetic acid (0.28 ml_, 4.78 mmol) in DCM (16 ml_), 2,2-difluoroacetaldehyde (0.16 M solution in DCM, 15 ml_, 2.39 mmol) and sodium triacetoxyborohydride (1 g, 4.78 mmol) were added. After stirring at r.t. for 24 h, additional 2,2-difluoroacetaldehyde (0.16 M solution in DCM, 15 mL) and sodium triacetoxyborohydride (1 g) were added and the reaction mixture was again stirred at r.t. overnight. NaHCC>3 sat. solution was added and it was extracted with DCM. The combined organic phases were dried over MgS0 4 and concentrated to dryness. The crude product was purified by flash chromatography, silica gel, gradient DCM to MeOH:DCM (1 :4) to give the title compound (371 mg, 73% yield).

Intermediate 4: (/?)-3-(Methylamino)-1 -(thiophen-2-yl)propan-1 -ol

A solution of (R)-3-chloro-1 -(thiophen-2-yl)propan-1 -ol (4.7 g, 26.9 mmol) and methylamine (33%wt in EtOH, 33 mL, 269 mmol) in EtOH (20 mL) was heated in a sealed tube at 90 °C overnight. The solvent was evaporated to dryness and the residue was re- dissolved in DCM. The organic phase was washed with 1 N NaOH, dried over MgSC>4 and concentrated to dryness. The crude product (3.9 g) was slurried in a mixture of methylcyclohexane-toluene (3:1 v/v, 22 mL) and heated at 60 °C for 1 h. Then, it was allowed to cool down and it was stirred at r.t. for 1 h. The solids were filtered, washed with methylcyclohexane and dried under vacuum to obtain the title compound (2.9 g, 62% yield).

This method was used for the preparation of Intermediates 5-8 using suitable starting materials:

(1 ) Final crystallization was not performed

Intermediate 9: ( ?)-3-(Methylamino)-1 -(thiophen-3-yl)propan-1 -ol

Step 1. ( ?)-3-Chloro-1 -(thiophen-3-yl)propan-1 -ol: To a solution of (S)-2-Me-CBS- oxazaborolidine (0.635 g, 2.3 mmol) in dry THF (42 mL), borane dimethyl sulfide complex (2.17 mL, 22.9 mmol) was added dropwise at r.t. After stirring for 10 min, a solution of 3- chloro-1 -(thiophen-3-yl)propan-1 -one (prepared according to the procedure described in US2003/0158185 Example 74 steps a-c) (2 g, 1 1.45 mmol) in dry THF (80 mL) was added during 45 min. The reaction mixture was stirred at r.t. for 1 h. Then, it was concentrated to dryness. The residue was dissolved in Et.20 and it was washed with NH4CI sat. solution. The organic phase was dried over MgSC>4 and concentrated under vacuum. The crude product was purified by flash chromatography, silica gel, gradient Cyclohexane/EtOAc 100:0 to Cyclohexane/EtOAc 0:100, to give the title compound (0.982 g, 48% yield).

Step 2. Title compound: Starting from the product obtained in Step 1 and following the experimental procedure described for the preparation of Intermediate 4, the title compound was obtained (842 mg, 88% yield).

This method was used for the preparation of Intermediates 10-1 1 using suitable starting materials:

Intermediate 12: 8-Fluoro-3,6-dimethylquinazolin-4(3H)-one

Step 1. 2-Amino-5-bromo-3-fluorobenzamide: A solution of 2-amino-5-bromo-3- fluorobenzoic acid (0.5 g, 2.14 mmol) and 1 ,1 '-carbonyldiimidazole (0.346 g, 2.14 mmol) in anhydrous THF (15 mL) was heated to reflux for 30 min. After cooling to r.t, ammonia (32 wt% aq. solution, 10 mL, 3.8 mmol) was added and the reaction was stirred at r.t. overnight. The solvent was evaporated to dryness, the residue was slurried in water and the precipitated solids were filtered, washed with water and dried under vacuum to afford the title compound (349 mg, 70% yield).

Step 2. 6-Bromo-8-fluoro-3-methylquinazolin-4(3H)-one: Following the experimental procedure described in Step 2 of Intermediate 1 , starting from the product obtained in Step 1 , the title compound was obtained (280 mg, 72% yield).

Step 3. Title compound: A mixture of the product obtained in Step 2 (170 mg, 0.66 mmol), K2CO3 (0.59 mg, 4.3 mmol), 2,4,6-trimethyl-1 ,3,5,2,4,6-trioxatriborinane (0.1 mL, 0.73 mmol) and dichloro[1 ,1 '-bis(diphenylphosphino)ferrocene]palladium (II) CH2CI2 adduct (484 mg, 0.66 mmol) in DME (3.5 mL), under an Ar atmosphere, was heated at 120 °C for 1 h in a MW oven. The reaction mixture was filtered through a pad of Celite and it was washed with EtOAc. The filtrate was concentrated to dryness and the residue was purified by by flash chromatography, silica gel, gradient DCM to MeOH:DCM (1 :4) to give the title compound (79 mg, 40% yield).

Intermediate 13: 5,7-Difluoro-3-methylquinazolin-4(3H)-one

Following the procedure described for the synthesis of intermediate 2 but using 5,7- difluoroquinazolin-4(3/-/)-one as starting material, the title compound was obtained.

Synthesis of Examples

Example 1 : yV-Methyl-3-((1 -methyl-1 ,2,3,4-tetrahydroquinolin-5-yl)oxy)-3- (thiophen-2-yl)propan-1 -amine

Step 1. 5-(3-Chloro-1 -(thiophen-2-yl)propoxy)-1 -methyl-1 ,2,3,4- tetrahydro quinoline: To a solution of 3-chloro-1 -(thiophen-2-yl)propan-1 -ol (0.39 g, 2.21 mmol), tributylphosphine (0.66 mL, 2.65 mmol) and 1 -methyl-1 ,2,3,4-tetrahydroquinolin-5-ol (0.36 g, 2.21 mmol) in toluene (15 mL), ADDP (0.67 g, 2.65 mmol) was added. The mixture was stirred at 100 °C overnight. The suspension was filtered through a pad of Celite that was washed with toluene and the filtrate was concentrated under vacuum. The crude product was used in the next step without further purification (1.38 g, overweight, quantitative yield assumed).

Step 2. Title compound: In a sealed tube, a mixture of the product obtained in Step 1 and methylamine (33 wt% in EtOH, 5.3 mL, 42.4 mmol) was heated at 100 °C overnight. Then, it was concentrated to dryness. The residue was dissolved in DCM (10 mL) and it was washed with 1 N NaOH solution and brine. The organic phase was dried over MgSC and concentrated under vacuum. The crude product was purified by flash chromatography, silica gel, gradient DCM to MeOH:DCM (1 :4) to give the title compound (22 mg, 3% yield, 2 steps). HPLC retention time (method C): 4.21 min; MS: 317.1 (M+H).

This method was used for the preparation of Examples 2-3 using suitable starting materials:

Ret HPLC

EX Structure Chemical name MS

time

(M+H) Method (min)

/

/V-methyl-3-((2-methyl- 1 ,2,3,4-

2 tetrahydroisoquinolin-5- 3.19 317.1 C

yl)oxy)-3-(thiophen-2- yl)propan-1-amine

HN— 3-((2-(2,2-difluoroethyl)-

Only

1 ,2,3,4- fragment

tetrahydroisoquinolin-5-

3.96 ation m/z

yl)oxy)-/V-methyl-3-

214.1

(thiophen-2-yl)propan-1- observed

amine

HN—

Example 4: ( ?)-yV-Methyl-3-(phth (thiophen-2-yl)propan-1 -amine

To a solution of Intermediate 4 (75 mg, 0.44 mmol) in anhydrous DMA (3 mL) cooled at 0 °C, NaH (60% dispersion in mineral oil, 44 mg, 1.10 mmol) was added portionwise under a nitrogen atmosphere. After stirring for 30 min at 0 °C, a solution of 5- fluorophthalazine (65 mg, 0.44 mmol) in anhydrous DMA (2 mL) was added and the reaction mixture was heated at 50 °C for 1 .5 h. It was then cooled to 0-5 °C and then water was added. The aqueous phase was extracted with DCM and the combined organic phases were dried over MgS0 4 and evaporated to dryness. The crude product was purified by flash chromatography, silica gel, gradient DCM to MeOH:DCM (1 :4) to give the title compound (34 mg, 49% yield).

HPLC retention time (method A): 2.44 min; MS: 300.1 (M+H).

This method was used for the preparation of Examples 5-16 using suitable starting materials:

Example 17: 3-(lsoquinolin-5-ylmethoxy)-yV-methyl-3-phenylpropan-1 -amine

Step 1. ferf-Butyl (3-(isoquinolin-5-ylmethoxy)-3-phenylpropyl)(methyl) carbamate: To a suspension of NaH (70 mg, 60% dispersion in mineral oil, 1 .74 mmol) in THF (2 mL) cooled at 0 °C, a solution of ie/f-butyl (3-hydroxy-3- phenylpropyl)(methyl)carbamate (181 mg, 0.7 mmol) in THF (9 mL) was added. The reaction mixture was stirred at r.t. for 60 min and then a solution of 5- (chloromethyl)isoquinoline (145 mg, 0.82 mmol) in THF (5 mL) and tetrabutylammonium iodide (22 mg, 0.6 mmol) were added at 0 °C. The reaction mixture was heated at 45 °C for 24 h. NH4CI sat. solution was added and it was extracted with DCM. The organic phase was dried over Na2S0 4 , filtered and concentrated under vacuum. The residue was purified by flash chromatography, silica gel, gradient Hexane to Hexane:EtOAc 1.5:1 , to afford the title compound (147 mg, 53% yield).

Step 2. Title compound: To a solution of the product obtained in Step 1 (54 mg, 0.13 mmol) in 1 ,4-dioxane (0.3 ml_), HCI (4 M solution in 1 ,4-dioxane, 0.47 ml_, 1.9 mmol) was added and the mixture was stirred at r.t. for 35 min. The reaction mixture was concentrated under vacuum, 10% Na2CC>3 aqueous solution was added and it was extracted with DCM. The organic phase was dried over Na2S0 4 , filtered and concentrated to dryness to afford the title compound (34 mg, 84% yield).

HPLC retention time (method D): 4.35 min; MS: 307.4 (M+H).

This method was used for the preparation of Examples 18-19 using suitable starting materials:

Example 20: 3-(lsoquinolin-8-ylmethoxy) V-methyl-3-(thiophen-2-yl)propan-1 amine

Step 1. ferf-Butyl (3-(isoquinolin-8-ylmethoxy)-3-(thiophen-2-yl)propyl)(methyl ) carbamate: ferf-Butyl (3-hydroxy-3-(thiophen-2-yl)propyl)(methyl)carbamate was reacted with 8-(chloromethyl)isoquinoline following the conditions described in Example 17 Step 1 , to afford the title compound (71 % yield).

Step 2. Title compound: In a round bottomed flask, ZnBr2 (350 mg, 1 .5 mmol) was dried under vacuum at 200 °C for 4 h and then it was allowed to cool. Once the solid reached r.t., a solution of the compound obtained in Step 1 (125 mg, 0.3 mmol) in DCM (3 mL) was added and the mixture was stirred at r.t. under an argon atmosphere for 24 h. Water was added and the mixture was stirred for 1 h. The phases were separated and the aqueous phase was extracted with DCM. The organic phase was dried over Na2S0 4 , filtered and concentrated under vacuum. The residue was purified by flash chromatography, silica gel, gradient DCM to MeOH:DCM (1 :1.5) to afford the title compound (1 1 mg, 12% yield).

HPLC retention time (method D): 4.26 min; MS: 313.1 (M+H). This method was used for the preparation of Examples 21 -22 using suitable starting materials:

Ret time MS HPLC

EX Structure Chemical name

(min) (M+H) Method

3-(isoquinolin-5-

O ylmethoxy)-/V-methyl-

21 4.25 313.1 D

3-(thiophen-2- yl)propan-1-amine

/V-methyl-3-(quinolin-5- o ylmethoxy)-3-

22 4.26 313.1 D

(thiophen-2-yl)propan- 1 -amine

N Examples 23 and 24: (S)-yV-methyl-3-((1 -methyl-1 ,2,3,4-tetrahydroquinolin-5- yl)oxy)-3-(thiophen-2-yl)propan-1 -amine and (/?)-/V-methyl-3-((1 -methyl-1 ,2,3,4- tetrahydroquinolin-5-yl)oxy)-3-(thiophen-2-yl)propan-1 -amine

Starting from Example 1 , a chiral preparative HPLC separation (column: Chiralpak IC; temperature: ambient; flow: 2.4 mL/min; eluent: n-Heptane/(EtOH + 0.2% DEA) 90/10 v/v) was carried out to give the title compounds.

Examples 25 and 26: (S)-yV-methyl-3-((2-methyl-1 ,2,3,4-tetrahydroisoquinolin-5- yl)oxy)-3-(thiophen-2-yl)propan-1 -amine and (/?)-/V-methyl-3-((2-methyl-1 ,2,3,4- tetrahydroisoquinolin-5-yl)oxy)-3-(thiophen-2-yl)propan-1 -amine

Starting from Example 2, a chiral preparative HPLC separation (column: Chiralpak IC; temperature: ambient; flow: 1 1 mL/min; eluent: n-Heptane/(IPA + 0.33% DEA) 70/30 v/v) was carried out to give the title compounds.

Following the method described for the preparation of Example 1 but using suitable starting materials, Example 27 was obtained:

Ret time MS HPLC

EX Structure Chemical name

(min) (M+H) Method /V-ethyl-3-((2-methyl-

1 ,2,3,4-

27 tetrahydroisoquinolin- 3,83 331 ,2 E

5-yl)oxy)-3-(thiophen-

2-yl)propan-1 -amine

H

Following the method described for the preparation of Example 4 but using suitable starting materials, Examples 28-45 were obtained:

(R)-8-(3-(ethylamino)-

1 -(thiophen-3-

41 yl)propoxy)-3- 3.52 344.1 E

methylquinazolin-

4(3H)-one< 1 '

(R)-5-(3-(ethylamino)-

1 -(thiophen-3-

42 yl)propoxy)-2- 3.49 343.1 E

0 methylisoquinolin-

1 (2H)-one< 1 '

$ Η

(R)-3-methyl-5-(3-

(methylamino)-l-

43 (thiophen-3- 3.44 330.1 E

yl)propoxy)quinazolin-

4(3H)-one< 1 '

(R)-8-(3-(ethylamino)-

1 -(thiophen-2-

44 yl)propoxy)-3,6- 3.97 358.1 E

ο dimethylquinazolin-

4(3H)-one< 1 '

(R)-7-fluoro-3-methyl-

5-(3-(methylamino)-1 -

45 (thiophen-2- 4.00 348.1 E

0

yl)propoxy)quinazolin-

4(3H)-one< 1 '

Η

( ) The reaction was carried out using 'BuOK as base and DMSO as solvent.

Pharmacological Data

- Binding assay to human οϋδ-Ι subunit of Cav2.2 calcium channel.

Human α,2δ-1 enriched membranes (2.5 g) 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 μΜ pregabalin. The binding of the test compound was measured at five different concentrations. After 60 min of incubation at 27 °C, the 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 30 μΙ of scintillation cocktail were added to each well before radioactivity reading.

Readings were performed in a Trilux 1450 Microbeta radioactive counter (Perkin Elmer).

- Binding assay to human norepinephrine transporter (NET).

Human norepinephrine transporter (NET) enriched membranes (5 μg) were incubated with 5 nM of radiolabeled [3H]-Nisoxetin in assay buffer containing 50 mM Tris-HCI, 120 mM NaCI, 5 mM KCI, pH 7.4.

NSB (non specific binding) was measured by adding 10μΜ desipramine. The binding of the test compound was measured at five different concentrations. After 60 min incubation at 4 °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, 0.9% NaCI, pH 7.4.

Filter plates were dried at 60 °C for 1 h and 30 μΙ of scintillation cocktail were added to each well before radioactivity reading. Readings were performed in a Trilux 1450 Microbeta radioactive counter (Perkin Elmer).

The following scale has been adopted for representing the binding to the α2δ1 receptor expressed as Ki:

+ Κί-α2δ1 >= 3000 nM

++ 500nM < Κί-α2δ1 <3000 nM

+++ 100nM < Κί-α2δ1 <500 nM

++++ Κί-α2δ1 <100 nM For the dual compounds and regarding the NET receptor, the following scale has been adopted for representing the binding expressed as Ki:

+ Ki-NET >= 1000 nM

++ 500nM < Ki-NET <1000 nM

+++ 100nM < Ki-NET <500 nM

++++ Ki-NET <100 nM The results of the binding for α2δ receptor are shown in Table 1 :

Table 1

EXAMPLE Ki(nM)

alpha2delta

Hum

1 +++

2 ++++

3 +++

4 +++

5 +

6 ++++

7 +++

8 +++

9 +++

10 +++

11 ++

12 +++

13 ++

14 ++++

15 +

16 ++++

17 +

18 +

19 +

20 +

21 +

22 +

23 ++

24 +++

25 +++

26 ++++

27 ++++ 28 ++

29 +++

30 ++++

31 ++++

32 +++

33 +++

34 +

35 ++

36 +

37 ++++

38 ++

39 +++

40 ++++

41 ++++

42 ++++

43 +++

44 +++

45 +++

The binding results for the α2δ receptor and the NET receptor for the dual compounds are shown in Table 2:

Table 2

EXAMPLE Ki(nM) NET Ki(nM)

Hum alpha2delta

Hum

1 ++ +++

3 ++ +++