Summary
The histamine H3 receptor (H3R) is an important modulator of numerous central control mechanisms. Novel lead optimizations for H3R antagonists/inverse agonists involved studies of structure–activity relationships, cross‐affinities, and pharmacokinetic properties of promising ligands. Blockade of inhibitory histamine H3 autoreceptors reinforces histaminergic transmission, while antagonism of H3 heteroreceptors accelerates the corticolimbic liberation of acetylcholine, norepinephrine, glutamate, dopamine, serotonin and gamma‐aminobutyric acid (GABA). The H3R positioned at numerous neurotransmission crossroads indicates therapeutic applications of small‐molecule H3R modulators in a number of psychiatric and neurodegenerative diseases with various clinical candidates available. Dual target drugs displaying H3R antagonism/inverse agonism with inhibition of acetylcholine esterase (AChE), histamine N‐methyltransferase (HMT), or serotonin transporter (SERT) are novel class of procognitive agents. Main chemical diversities, pharmacophores, and pharmacological profiles of procognitive agents acting as H3R antagonists/inverse agonists and dual H3R antagonists/inverse agonists with inhibiting activity on AChE, HMT, or SERT are highlighted here.
Keywords: AChE, Histamine H3 receptor, HMT, Pharmacophore, Procognitive, SERT
Introduction
Histamine, 2‐(1H‐imidazol‐4‐yl)ethanamine, is a biogenic amine implicated in various physiological and pathophysiological processes. It is a chemical mediator in immediate allergic response, inflammatory and immunomodulatory processes, and gastric acid secretion and also acts as a neurotransmitter in the central nervous system (CNS) 1. Histamine is synthesized from L‐histidine by cytoplasmatic L‐histidine decarboxylase enzyme (HDC, E.C. 4.1.1.22) in the histaminergic neurons, stored in the vesicles and then released from the axon terminals 1, 2. In the brain, histamine is mainly inactivated by neuronal histamine N‐methyltransferase enzyme (HMT, E.C. 2.1.1.8) 2, 3 in the nearest glia cells 2 or by diamine oxidase (DAO, EC 1.4.3.6) into imidazole acetic acid (IAA), which acts as γ‐aminobutyric acid (GABA)A receptor agonist 4. Histamine mediates its main biological activities by interaction with four distinct histamine receptor subtypes (H1R–H4R). All histamine receptor subtypes belong to the class A family of G protein‐coupled receptors (GPCRs) 1, 3. Although H1R and H2R antagonists are crucial drugs in therapy of allergy and ulcer, respectively, the H3R and H4R antagonists are in clinical or discovery phase. The recently discovered H4 receptors are involved in immunomodulatory and inflammatory processes 4, 5, 6, 7, while H3 receptors play a central role in neurotransmission of histamine and control release of many other neurotransmitters 8, 9. The H3R is pharmacologically discovered in 1983 as histamine presynaptic autoreceptor 3. The H3 autoreceptors are involved in a negative feedback modulation of histamine synthesis and inhibition of its release from histaminergic neurons 3. The histaminergic neurons are mainly located in the posterior hypothalamus and extensively projected into all major areas of the CNS of humans and rodents (Figure 1) 7, 8, 9. The histamine H3 receptor is not only expressed on histaminergic neurons, but also on neighboring aminergic neurons, as a heteroreceptor. (In)Activation of H3 heteroreceptor modulates the release of acetylcholine in cerebral cortex, hippocampus, nucleus accumbens, nucleus basalic magnocellularis, and striatum 10, 11, 12, 13; noradrenaline in cerebral cortex, hippocampus, hypothalamus, locus coeruleus, and para‐ventricular nucleus 14, 15; dopamine in striatum 16, 17, 18, 19, 20; GABA in cerebral cortex, striatum, substantia nigra, and ventral tegmental area 21, 22, 23; and serotonin in cerebral cortex, hypothalamus, substantia nigra (part reticulate), and striatum 24, 25, 26. While this extensive list is not complete, it clearly demonstrates the wide range of H3R‐mediated neuromodulation. Also, the H3 receptor is indirectly involved in regulation of other neurotransmitter systems by modulating levels of histamine, which then activates postsynaptic receptors (Figure 1). Studies on the frontocortical histaminergic pathways proved that blockade of negative feedback mechanism of presynaptic H3 autoreceptors augments histaminergic transmission, while antagonism of H3 heteroreceptors facilitates the corticolimbic liberation of acetylcholine, noradrenaline, dopamine, glutamate, GABA, and serotonin 27, 28, 29, 30. Recent pharmacological and (pre)clinical studies confirmed that the H3R antagonists/inverse agonists are effective in the treatment of a sleep disorders (narcolepsy), cognitive impairment, pain/itch, stroke, depression, schizophrenia, Alzheimer's disease (AD), attention deficit hyperactivity disorder (ADHD), dementia, and neurodegenerative disorders 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41.
Figure 1.

Physiological role of H3R in central nervous system (CNS): HA, histamine; ACh, acetylcholine; NT, neurotransmitters (e.g., noradrenaline, serotonin, dopamine, GABA); TMN, tuberomammillary nucleus; HMT, histamine N‐methyltransferase; DAO, diamine oxidase; NMH, N τ‐methyl‐histamine; IAA, imidazole acetic acid; IP3, inositol trisphosphate; DAG, diacetyl glycerol; cAMP, cyclic adenosine triphosphate; ctx, cerebral cortex, hip, hippocampus; hyp, hypothalamus; na, nucleus accumbens; str, striatum; gp, globus pallidus; amg, amygdala.
Molecular aspects and signaling of the H3R 42, 43, 44, 45, structure–activity relationships of H3R antagonists/inverse agonists 35, 46, 47, 48, 49, 50, 51, 52, 53, 54, and H3R antagonists/inverse agonists pharmacology have recently been discussed in several comprehensive reviews 4, 32, 34, 36, 52, 55, 56, 57, 58.
The location of H3R at the neurotransmission crossroads indicated potential therapeutic applications of ligands that are better refined to therapeutic needs using a less selective pharmacological profile by multiple targeting. In this respect, dual H3R antagonism and inhibitory potency on acetylcholine esterase (AChE), catabolic HMT, or serotonin transporter (SERT) have been described as novel classes of procognitive agents for treatment of wide range of central disorders. In this manuscript, pharmacophores of H3R antagonists/inverse agonists and H3R antagonists/inverse agonists with dual HMT or SERT inhibiting activity are reviewed. The influence of structural variations in peripheral scaffolds on potency and the pharmacokinetic profile of drug candidates are discussed.
Histamin H3R Antagonists/Inverse Agonists
Early synthetic H3R ligands were imidazole derivatives, as analogs of histamine 59, 60, showing in numerous cases some affinity at other aminergic receptors, species selectivity, low oral bioavailability, or rapid metabolism. Similarly, first‐generation H3R antagonists, thioperamide (1), clobenpropite (2), ciproxifan (3), and FUB181 (4) (Figure 2), were derivatives of 4‐monosubstituted‐imidazole 47, 59, 61, 62, 63, 64. The imidazole‐containing H3R ligands (1–4, Figure 2) have displayed low H3R selectivity and intensive hepatic metabolism 47, 65.
Figure 2.

Early synthetic hH 3R antagonists/inverse agonists (1–4). Basic scaffold of the hH 3R antagonists/inverse agonists pharmacophore is marked in blue color.
The vast majority of H3R antagonists were pharmacologically classified as inverse agonists because of their negative effects on the intrinsic activity of the H3R with mostly high levels of constitutive activity 45, 66, 67. Nearly all H3R antagonists/inverse agonists have structurally similar basic moiety with an aliphatic tertiary amine substituted by a linking alkyl chain 68. This basic skeletal moiety is crucial for ionic interactions between protonated ammonium functionality of the H3R antagonists/inverse agonists and Asp114 in transmembrane domain III of the H3R 69. Due to the risks of low selectivity, rapid metabolism, and cytochrome P450 interactions of imidazole‐containing H3R ligands (1–4, Figure 2), this moiety was biosterically replaced with pyrrolidine (5–8, Figure 3, 20, 21 Figure 4), piperidine (9, 10, Figure 3, 18–20, Figure 4), azepane (11, 22 Figure 4), piperazine (12, 13, Figure 4), diazepane (14, 15, Figure 4), tetrahydrobenzo[d]azepine, and related systems (16, 17, Figure 4) that have been investigated. The imidazole exchange resulted in a significant increase in affinities at human H3R (hH3R) and with optimized substitution pattern in improvement of pharmacokinetic properties in various lead compounds (5–22, Figures 3 and 4) 53, 54, 70, 71. Recently developed hH3R antagonists/inverse agonists have demonstrated efficacy in diverse preclinical models of cognitive deficits, suggesting the potential value of the ligands as cognition enhancers in many CNS diseases 72.
Figure 3.

The hH 3R antagonists/inverse agonists for cognition with pyrrolidine/piperidine ring as a basic moiety (4–10). Basic scaffold of the hH 3R antagonists/inverse agonists pharmacophore is marked in blue color.
Figure 4.

The hH 3R antagonists/inverse agonists for cognition with azepane/piperazine/diazepane as a basic moiety (11–22). Basic scaffold of the hH 3R antagonists/inverse agonists pharmacophore is marked in blue color.
The highly potent and selective benzofuran/methylpyrrolidine hH3R antagonist ABT‐239 (5, Figure 3) was in preclinical development for the potential treatment of cognitive disorders and schizophrenia 32, 52 but was not advanced for clinical evaluation due to hERG channel affinity and QT prolongation in monkey 55. ABT‐239 displayed an acceptable rat pharmacokinetic profile (53% oral BA, t 1/2 = 5.3 h) and CNS penetration (B/P ratio >20). The naphthalene/methylpyrrolidine derivative Abbott (6, Figure 3) also showed strong hH3R antagonistic activity, good rat pharmacokinetic properties (55% oral BA, t 1/2 = 5.5 h), but significantly higher CNS penetration (B/P ratio = 72) than the methylpyrrolidine/benzofuran analog ABT‐239 (5, Figure 3) 73. Furthermore, phenoxypropyldimethylpyrrolidine derivatives, such as iradabisant (7, Figure 3), demonstrated potent hH3R antagonistic activity, acceptable rat pharmacokinetic profile (83% oral BA) and CNS penetration (B/P ratio = 2.3), and favorable behavioral effects in studies using several preclinical rodent models of centrally occurring disorders such as schizophrenia, AD, and ADHD 74. A recent preclinical study on ABT‐239 showed that H3R antagonism may deliver symptomatic treatment in AD and also possesses disease‐modifying efficacy 75. Very recently developed 5‐{4‐[3‐(R)‐2‐methylpyrrolidin‐1‐yl)propoxy]phenyl}‐2‐pyridin‐2‐yl‐2H‐pyridazin‐3‐one showed high affinity for hH3R (pKi(hH3R): 8.8) with very high selectivity over the other histamine receptor subtypes (Table 1). The compound displayed excellent drug‐like properties, such as satisfactory pharmacokinetic properties (oral BA = 78% rat, 92% dog, 96% monkey, t 1/2 = 4.5 h rat, 0.8 h dog, 6.5 h monkey), low binding to human plasma proteins, weakly inhibited cytochrome P450 isoforms, and acceptable brain exposure (B/P ratio = 1.1 rat). The ligand showed potent H3R antagonistic activity and enhancement of cognitive function in rat models, but further development was discontinued because of detected genotoxicity 76.
Table 1.
Pharmacodynamic and pharmacokinetic properties for hH3R antagonists with procognitive effects
| Compound | Name | pKi (hH3R) | Oral BA [%] | C(Brain)/C(Plasma) ratio | t 1/2 [h] | Therapeutic application | Clinical development |
|---|---|---|---|---|---|---|---|
| 5 | ABT‐239 | 9.4 | 53 | >20 | 5.3 | Cognitive disorders, schizophrenia | |
| 6 | Abbott | 9.6 | 55 | 72 | 5.5 | Cognitive disorders | |
| 7 | Iradabisant | 9.3 | 83 | 2.6 | Cognitive disorders | Phase I | |
| 8 | PF‐03654746 | 8.6 | 26 | 2.1 | Cognitive disorders, allergy | Terminated in Phase II | |
| 9 | Pitolisant | 7.1 | 84 | 2.0 | Cognitive disorders, narcolepsy | Phase III | |
| 10 | JNJ‐5207852 | 9.2 | 85–107 | >13 | Cognitive disorders, narcolepsy | ||
| 15 | GSK‐334429 | 9.5 | 91 | 2.1 | Cognitive disorders, neuropathic pain | ||
| 16 | GSK‐189254 | 9.9 | 83 | 1.6 | Cognitive disorders, narcolepsy, neuropathic pain | Terminated in Phase II | |
| 21 | ABT‐288 | 8.7 | 37 | 1.5 | 1.3 | Cognitive deficit in AD and schizophrenia | Failed to show efficacy in Phase II AD trials |
| 22 | GSK‐239512 | 10.0 | 51 | 1.8 | 1.2 | Cognitive deficit in AD | Tolerated in Phase II with positive effects on attention/memory in AD |
| AZD‐5213 | 9.3 | Cognitive deficit in AD | Phase II for Tourette's syndrome and neuropathic pain |
Values are obtained for rat species.
AD, Alzheimer's disease.
Other benzene/pyrrolidine derivatives such as PF‐03654746 (8, Figure 3), showed good hH3R antagonistic activity, modest oral bioavailability (26%), acceptable CNS penetration (B/P ratio = 2.1), and procognitive performance in rodents 77. Compound 8 has successfully completed human safety and efficacy studies in adults with ADHD 54, 77. However, PF‐03654746 has failed to show efficacy superior to standard of care in clinical studies of excessive daytime sleepiness associated with narcolepsy (EDS), cognitive impairment in schizophrenia (CIAS), AD, ADHD, and allergic rhinitis (AR) [http://www.ncats.nih.gov/files/PF-03654746.pdf], and clinical development was halted.
Exchange of imidazole ring in FUB 181 (4, Figure 2) resulted in formation of the piperidine analog pitolisant (9, formerly known as BF2.649 and tiprolisant, Figure 3), which showed strong hH3R antagonistic potency and good rat pharmacokinetic properties (84% oral BA, t 1/2 = 2 h) 78. Pitolisant, which has been shown to have some precognitive effects, is currently in late stage of clinical development on therapy in Parkinson's disease and narcolepsy as one of most promising H3R inverse agonist 54, 71, 79. Recently obtained results in an early Phase II study of the human photosensitivity model of epilepsy suggest that pitolisant might be helpful in chronic seizure treatment of both partial and generalized epilepsies 80.
Other phenoxypropylpiperidine preclinical candidate JNJ‐5207852 (10, Figure 3) displayed high and selective hH3R antagonism, high oral bioavailability (>85 %), but rather long half‐life time (t 1/2 > 13 h) and very high risk for phospholipidosis development during long‐term therapy 35, 81. The observed risk of phospholipidosis was assumably attributed to its dibasic and cationic amphiphilic nature 55, 81. Compound 10 showed in vivo efficacy in rodent arousal models and thus represents a new pharmacological tool for the treatment of conditions associated with EDS 82. Phenoxypropylazepane derivative (11, Figure 4) displayed potent and selective antagonistic/inverse agonistic activity on hH3R 54, 74, while phenoxypropylpiperazine analog (12, Figure 4) showed modest hH3R antagonism, high selectivity over H1 and H2 receptors, good oral bioavailability, and procognitive effects in rodent models 83. In addition, phenoxypropylpiperazine derivative (13, S38761‐1, Figure 4) displayed potent and selective antagonistic/inverse agonistic activity on hH3R with possible therapeutic application as procognitive agent 54, 84. Also, phenoxypropyldiazepane analog (14, A‐320436, Figure 4) showed good binding to hH3R, but poor brain penetration and lack of procognitive activity in vivo 85. An improved pharmacokinetic profile (91% oral BA, t 1/2: 2.1 h) was achieved with the novel diazepine derivative (15, GSK‐334429, Figure 4) that demonstrated positive effects in rat models for cognitive disorders, neuropathic pain, and memory impairment 86. Use of the tetrahydrobenzo[d]azepine system as basic moiety of the hH3R antagonist/inverse agonist resulted in development of the clinical candidate for cognitive disorders [16, GSK‐189254, Figure 4]. GSK‐189254 showed potent and selective hH3R antagonistic activity, good pharmacokinetic profile (83% oral BA, t 1/2: 1.6 h), CNS penetration, and favorable procognitive activity in preclinical models of cognitive impairment, neuropathic pain, and narcolepsy 49, 87. Compound 16 displayed promising preclinical results in cognition models 88, but the development of this compound was terminated in Phase II 87, 89. The tetrahydrobenzo[d]azepine and phenoxypropylpiperidine moieties have been combined by GlaxoSmithKline in dibasic antagonist (17, Figure 4), which provide potential flip‐flop binding modes in hH3R 90. The cyclobutylpiperidinyloxy side chain was used as a moiety in a series represented by potent and selective hH3R inverse agonists (18 and 19, Figure 4) 91, 92. A process patent has been applied for compound 18 93, whereas 19 was optimized to overcome adrenergic activity and hERG liability of lead compounds 92. Dibasic biphenyl series of hH3R antagonists, characterized by a biphenyl core and two basic centers, is exemplified by very potent hH3R antagonist (20, Figure 4) 90. The high binding affinity shown by these biaryl compounds supporting the hypothesis that the two regions described within the hH3 receptor binding site can be simultaneously occupied by an dibasic biphenyl hH3R ligand. A related multimodal binding mode is possible for several other dibasic hH3R antagonists, such as 10 and 17.
The human H3R antagonists AZD‐5213 (structure undisclosed) 94 and SAR‐110894 (structure undisclosed) 95 are in Phase I clinical studies for AD (NCT‐ID: NCT01548287 and NCT01266525, respectively) with AZD‐5213 advancing to Phase II for Tourette's syndrome (http://clinicaltrials.gov/ct2/show/NCT01904773) and neuropathic pain (http://clinicaltrials.gov/ct2/show/NCT01928381). AZD‐5213 displayed efficacy in rodent behavioral models of cognition and in humans was safe and well tolerated following repeated doses with a short half‐life 94.
The hH3R antagonists ABT‐288 (21, Figure 4) and GSK‐239512 (22, Figure 4) are in Phase II trials for AD (NCT‐ID: NCT01018875 and NCT01009255, respectively). Human H3R antagonist ABT‐288 (pKi(hH3R = 8.7)) enhances the release of acetylcholine and dopamine in rat prefrontal cortex and shows efficacy in several animal cognition models. ABT‐288 demonstrated good pharmacokinetics (37% oral BA, t 1/2: 1.3 h) and CNS penetration (B/P ratio = 1.5) in rat models, a safe preclinical profile in rodent and dog studies and has advanced to clinical studies for the treatment of cognitive deficits in AD and schizophrenia 89, 96. However, a recent report indicated that while safe and well tolerated in humans, ABT‐288 failed to show procognitive effects in AD and schizophrenic patients 97, 98. The GSK‐239512 (pKi(hH3R = 10.0) displayed acceptable pharmacokinetics (51% oral BA, t 1/2: 1.2 h) and CNS penetration (B/P ratio = 1.8) in rats 99 and in patients with AD, exhibited evidence for positive effects on attention and memory 100. Furthermore, for hH3R inverse agonist, MK‐0249 (structure undisclosed) 101 has been completed Phase II study for AD (NCT‐ID NCT00420420).
Taking into account the beneficial cognitive effects reported for numerous H3R antagonists/inverse agonists, an additive or maybe synergistic effect can be speculated with the combination of additional procognitive pharmacological properties in one molecule.
Dual Properties of H3R Antagonists with Procognitive Effect
Modern medicinal chemistry has applied multitarget directed ligands (MTDLs) design to find multipotent compounds effective in treatment of CNS disorders such as AD, PD, ADHD, schizophrenia, and depression 102, 103. In the last few years, multiple‐targeted compounds have been designed using natural products as lead resources that have shown multiple pharmacological activities in CNS diseases 104. Novel dual‐activity hH3R antagonists/inverse agonists concomitantly interact with at least one further target, such as AChE, HMT, SERT, dopamine D2‐like receptors (D2 and D3), (5‐HT3), and monoamine oxidase 102, 103, 105. Dual‐activity hH3R antagonists with additional acetylcholinesterase inhibitory activity have been identified that lead to cognitive synergy with hH3R antagonism, as both pharmacological actions are able to enhance cholinergic neurotransmission. Based on the structure of acetylcholinesterase inhibitor tacrine, ligands with dual hH3R antagonistic/inverse agonistic and AChE inhibitory activity (23, FUB‐833, Figure 5) 106 were designed. Indeed, this compound class showed additional inhibition of HMT and butyrylcholinesterase (BuChE), simultaneously targeting four different procognitive pathways 106. Based on the dual‐acting tetrahydroaminoacridine hybrids 106, 1,1′‐octa‐, nona‐, and decamethylene‐bis‐piperidine derivatives and novel tetrahydroaminoacridine‐piperidine hybrids were developed that displayed potent and selective hH3R antagonism with remarkable anticholinesterase activity 107.
Figure 5.

Dual‐activity hH 3R antagonists/inverse agonists with procognitive effect (23–28). Basic scaffold of the hH 3R antagonists/inverse agonists pharmacophore is marked in blue color.
The hybrid compounds, such as ligand with (sub)nanomolar activities on the both hH3R and HMT targets (24, Figure 5), could increase intersynaptic histamine levels in the CNS and lead to beneficial procognitive effects in psychiatric and neurodegenerative diseases 33, 106, 108, 109, 110, 111. Multitarget ligands displaying dual hH3R antagonistic/inverse agonistic and HMT‐inhibiting properties are able to greatly enhance histaminergic neurotransmission via hH3 autoreceptors blockade and reduced catabolic rate for histamine inactivation via HMT inhibition 106, 110, 111. Also, group of dibasic dual hH3R antagonists/inverse agonists‐AChE inhibitors (25, 26, Figure 5) have been proposed for therapy of AD 112, 113. Merging the piperidinylpropoxy phenyl hH3R pharmacophore into heterocyclic moiety of AChE inhibitors yielded reversible and competitive AChE inhibitors with balanced nanomolar affinities at both targets (27, Figure 5) and high selectivity over the other histamine receptor subtypes 114. Based on the hH3R antagonists/inverse agonists pharmacophore, the AChE inhibitor BYYT‐25 that possesses β‐secretase (BACE 1) inhibitor activity, a set of quinoxaline‐based compounds acting on hH3R/AChE/BACE 1 were designed as novel MTDLs for treatment of AD. The most effective compound (28, Figure 5) displayed high selectivity over the other histamine receptor subtypes 115.
Results obtained from our previous theoretical studies, pharmacophore modeling, design 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, synthesis, and biological evaluation of novel compounds 33, 106, 108, 109, 110, 111 should be further used in discovery of novel dual‐activity hH3R antagonists/inverse agonists with procognitive effect.
Many clinical studies report that patients with depression often also suffer cognitive impairment 132. Selective serotonin reuptake inhibitors (SSRIs) are the most frequently prescribed antidepressant drugs, but they do not improve cognitive function of depressive patients 133, 134. Therefore, drugs displaying dual hH3R antagonistic/inverse agonistic and serotonin reuptake inhibiting properties may represent novel multitarget agents for the improved treatment of depressive states 135.
The combination of potent and selective hH3R antagonists/inverse agonists 1‐[4‐(3‐piperidin‐1‐ylpropoxy)benzyl]piperidine with tetrahydroisoquinoline‐derived SERT inhibitors gave potent dual hH3R antagonists/inverse agonists SERT inhibitors. Structural modification of the hH3R pharmacophore in the pendant amine portion of the molecule did not show significant influence on the SERT affinity of the compounds while binding affinity at hH3R varied significantly. Potent compounds were obtained by introducing morpholine and substituted piperidines (29, 30, Figure 6). Compounds 29 and 30 were also tested for functional activity at the hH3R and were found to be potent antagonists (pA 2 = 8.6 and 9.6, respectively). Further examination in the 5‐hydroxy‐tryptophan (5‐HTP)‐induced head twitch model in mice and in blood–brain barrier (BBB) experiments demonstrated a good correlation of behavioral pharmacology of 29 and 30 compounds with their brain concentration 136. The compounds with the optimized hH3R pharmacophore (29, 30, Figure 6) were used for further optimization of the SERT pharmacophore by changing at the 4‐aryl position. The modification in the aryl ring of the compound 30, such as replacement of the 4‐methoxy group or monosubstitution at 2 or 3 position, had little effect on the affinity for the hH3R. Also, for several compounds, characterization of enantiomerically pure products was performed. Compounds with 4‐thiomethyl substituent (31, 32 (JNJ‐28583867), Figure 6) or 4‐methoxy substituent (33, Figure 6) were selected as ligands with high SERT affinity. Screening the 31, 32, and 33 for affinity at 50 receptors, ion channels and transporters showed that 31 and 33 have moderate selectivity for the SERT over human norepinephrine transporter (NET) and the dopamine transporter (DAT), while 32 displayed 30‐fold selectivity for the SERT over the NET and DAT. The highest bioavailability in rat and high exposure in the brain after oral administration have been shown for 31 and 32. Following subcutaneous administration, compounds 32 and 33 showed 100% receptor occupancy in the rat brain at both SERT and hH3R 137. An in vivo functional activity of 32 at the hH3R was confirmed using the blockade of imetit‐induced drinking model. In the mouse tail suspension tests, this compound exhibited antidepressant‐like effect. The 32 was very effective at suppressing REM sleep from the dose of 1 mg/kg onward 138.
Figure 6.

Ligands with dual hH 3 antagonistic/inverse agonistic and serotonin transporter (SERT) inhibiting properties for cognition (29–40). Basic scaffold of the hH 3R antagonists/inverse agonists pharmacophore is marked in blue color.
Attachment of a piperidinylpropoxy side chain to the pyrrolidino‐tetrahydroisoquinoline scaffold also gave potent dual hH3R antagonists/inverse agonists SERT inhibitors (34, Figure 6). Substitutions in the 3 or 4 position of aryl ring provided compounds with higher affinity for SERT than their 2‐substituted counterparts. Both electron‐donating and electron‐withdrawing groups were tolerated with exceptions including two compounds (35, 36, Figure 6) of special interest, as these moieties are found in selective SSRIs citalopram (4‐CN) and fluoxetine (4‐CF3). The most potent pyrrolidino‐tetrahydroisoquinolines dual hH3R antagonists/inverse agonists‐SERT inhibitor 34 exhibited moderate bioavailability, relatively high brain concentrations and beneficial pharmacological effects in both 5‐hydroxytryptophan potentiated (5‐HTP) head twitch model and microdialysis experiments 139. Combination of 4‐aryl‐2,6‐tetrahydronaphtyridine scaffold and various hH3R pharmacophores resulted in the potent dual hH3R antagonists/inverse agonists SERT inhibitors. (37, Figure 6). In this group, substitution in the 3 or 4 position of aryl moiety was obligatory for SERT activity. The position of nitrogen on the tetrahydronaphtyridine core was also important for hH3R antagonistic and SERT inhibiting activity. The 4‐aryl‐2,5‐tetrahydronaphtyridines demonstrated significantly lower affinity for both targets indicating that 2,5‐tetrahydronaphtyridine core is not favored particularly with respect to SERT affinity 140.
Dibasic hH3R antagonists with serotonin reuptake inhibition were designed utilizing structure of the fluoxetine, known as SSRI. Starting from the known hH3R pharmacophore blueprint consisting of two basic functional groups attached to the central lipophilic core that contains an aromatic ring, fluoxetine structure was combined. Type and position of the hH3R side chain and substitution on the phenoxy ring significantly affected the affinity for SERT. The most potent ligands with highest hH3R and SERT affinity (38, Figure 6) were obtained by the introduction of the 3‐piperidinylpropoxy side chain in the para position of the phenyl ring (unsubstituted phenyl ring of fluoxetine). Replacement of this side chain with other hH3R components led to the derivatives with higher hH3R affinity but lower SERT inhibiting activity. All examined 3‐piperidinylpropoxy derivatives possess very high hH3R antagonistic activity, while SERT affinity is affected by type and position of substituents in phenyl ring 141. The 5‐ethynyl‐2‐aryloxybenzylamine‐based dual hH3R antagonists/inverse agonists SERT inhibitors were designed modifying the structure of compound 30 with goal to decrease complexity of ligands and improve their physical properties associated with good absorption and distribution into the brain. When position C‐3 of tetrahydroisoquinoline ring of 30 is removed, resulted structure was benzyl amine compounds. Besides, replacement of C‐4 position of tetrahydroisoquinoline ring by oxygen led to the ligands without optical activity. Combination of resulting aryloxybenzyl amine structure having SERT affinity with piperidine alkyne‐based hH3R pharmacophore gave compounds with high affinity for both hH3R and SERT (39, Figure 6). Substitution in 4 position of piperidine with fluorine or piperidine replacement by morpholine (40, Figure 6) resulted in decrease in affinity for hH3R and retained high SERT affinity. The 40 have good selectivity over human NET and DAT 142.
For few dibasic dual hH3R antagonists/inverse agonists SERT inhibitors, 38–40 could be proposed flip‐flop binding modes in hH3R 90.
Conclusion
Based on preclinical data, human H3R antagonists/inverse agonists display beneficial procognitive and arousal effects for the potential treatment of a broad spectrum of centrally occurring disorders such as AD, PD, ADHD, schizophrenia, and depression. This wide spectrum of potential therapeutic indications of hH3R antagonists/inverse agonists defined the hH3R as a highly significant drug target. The pharmacophore of hH3R antagonists/inverse agonists consists of a basic moiety linked by an alkyl spacer to an aromatic and then polar scaffold. As many hH3R antagonists/inverse agonists have failed in the first or second phase of clinical research, further lead optimization and pharmacokinetic screening as well as an increased understanding of the role of H3Rs in human CNS diseases are warranted significant steps in early‐stage H3R antagonist/inverse agonist drug discovery. Drugs displaying dual hH3R blockade and inhibition of AChE, HMT, or SERT represent a novel class of agents potentially capable of additively or synergistically augmenting the procognitive effects of hH3R antagonism.
Conflict of Interest
The authors declare no conflict of interest.
Disclosure
One of the authors (HS) is co‐inventor of pitolisant.
Acknowledgment
KN, SF, and DA acknowledge project supported by the Ministry of Education and Science of the Republic of Serbia, Contract #172033. Further supports by Else Kröner‐Fresenius‐Stiftung, Translational Research Innovation – Pharma (TRIP), Fraunhofer‐Projektgruppe für Translationale Medizin und Pharmakologie (TMP) (to HS), and the European COST Actions BM1007, CM1103, and CM1207 are also kindly acknowledged.
References
- 1. Schwartz JC, Arrang JM, Garbarg M, Pollard H, Ruat M. Histaminergic transmission in the mammalian brain. Physiol Rev 1991;71:1–51. [DOI] [PubMed] [Google Scholar]
- 2. Brown RE, Stevens DR, Haas HL. The physiology of brain histamine. Prog Neurobiol 2001;63:637–672. [DOI] [PubMed] [Google Scholar]
- 3. Arrang JM, Garbarg M, Schwartz JC. Auto‐inhibition of brain histamine release mediated by a novel class (H3) of histamine receptor. Nature 1983;302:832–837. [DOI] [PubMed] [Google Scholar]
- 4. Haas HL, Sergeeva OA, Selbach O. Histamine in the nervous system. Physiol Rev 2008;88:1183–1241. [DOI] [PubMed] [Google Scholar]
- 5. Panula P, Yang HY, Costa E. Histamine‐containing neurons in the rat hypothalamus. Proc Natl Acad Sci USA 1984;81:2572–2576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Walter M, Kottke T, Stark H. Perspective: The histamine H4 receptor: Targeting inflammatory diseases. Eur J Pharmacol 2011;668:1–5. [DOI] [PubMed] [Google Scholar]
- 7. Kiss R, KeserüPrime GM. Histamine H4 receptor ligands and their potential therapeutic applications: An update. Expert Opin Ther Pat 2012;22:205–221. [DOI] [PubMed] [Google Scholar]
- 8. Watanabe T, Taguchi Y, Shiosaka S, et al. Distribution of the histaminergic neuron system in the central nervous system of rats: A fluorescent immunohistochemical analysis with histidine decarboxylase as a marker. Brain Res 1984;295:13–25. [DOI] [PubMed] [Google Scholar]
- 9. Pillot C, Heron A, Cochois V, et al. A detailed mapping of the histamine H(3) receptor and its gene transcripts in rat brain. Neuroscience 2002;114:173–193. [DOI] [PubMed] [Google Scholar]
- 10. Cecchi M, Giorgetti M, Bacciottini L, Giovannini MG, Blandina P. Increase of acetylcholine release from cortex of freely moving rats by administration of histamine into the nucleus basalis magnocellularis. Inflamm Res 1998;47:S32–S33. [DOI] [PubMed] [Google Scholar]
- 11. Philippu A, Prast H. Importance of histamine in modulatory processes, locomotion and memory. Behav Brain Res 2001;124:151–159. [DOI] [PubMed] [Google Scholar]
- 12. Kraus MM, Fischer H, Tran MH, Philippu A, Prast H. Modulation of acetylcholine release by histamine in the nucleus accumbens. Inflamm Res 2001;50:S74–S75. [DOI] [PubMed] [Google Scholar]
- 13. Blandina P, Efoudebe M, Cenni G, Mannaioni P, Passani MB. Acetylcholine, histamine, and cognition: Two sides of the same coin. Learn Mem 2004;11:1–8. [DOI] [PubMed] [Google Scholar]
- 14. Schlicker E, Fink K, Hinterthaner M, Gothert M. Inhibition of noradrenaline release in the rat brain cortex via presynaptic H3 receptors. Naunyn Schmiedebergs Arch Pharmacol 1989;340:633–638. [DOI] [PubMed] [Google Scholar]
- 15. Korotkova TM, Sergeeva OA, Ponomarenko AA, Haas HL. Histamine excites noradrenergic neurons in locus coeruleus in rats. Neuropharmacology 2005;49:129–134. [DOI] [PubMed] [Google Scholar]
- 16. Schlicker E, Fink K, Detzner M, Gothert M. Histamine inhibits dopamine release in the mouse striatum via presynaptic H3 receptors. J Neural Transm Gen Sect 1993;93:1–10. [DOI] [PubMed] [Google Scholar]
- 17. Molina‐Hernández A, Nuñez A, Arias‐Montaño JA. Histamine H3‐receptor activation inhibits dopamine synthesis in rat striatum. NeuroReport 2000;11:163–166. [DOI] [PubMed] [Google Scholar]
- 18. Sánchez‐Lemus E, Arias‐Montaño JA. Histamine H3 receptor activation inhibits dopamine D1 receptor‐induced cAMP accumulation in rat striatal slices. Neurosci Lett 2004;364:179–184. [DOI] [PubMed] [Google Scholar]
- 19. Arias‐Montaño JA, Floran B, Garcia M, Aceves J, Young JM. Histamine H 3 receptor‐mediated inhibition of depolarization‐induced, dopamine D 1 receptor‐dependent release of [3H]‐γ‐aminobutyric acid from rat striatal slices. Br J Pharmacol 2001;133:165–171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Ferrada C, Ferré S, Casadó V, et al. Interactions between histamine H3 and dopamine D2 receptors and the implications for striatal function. Neuropharmacology 2008;55:190–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Yamamoto Y, Mochizuki T, Okakura‐Mochizuki K, Uno A, Yamatodani A. Thioperamide, a histamine H3 receptor antagonist, increases GABA release from the rat hypothalamus. Methods Find Exp Clin Pharmacol 1997;19:289–298. [PubMed] [Google Scholar]
- 22. Korotkova TM, Haas HL, Brown RE. Histamine excites GABAergic cells in the rat substantia nigra and ventral tegmental area in vitro. Neurosci Lett 2002;320:133–136. [DOI] [PubMed] [Google Scholar]
- 23. Bergquist F, Ruthven A, Ludwig M, Dutia MB. Histaminergic and glycinergic modulation of GABA release in the vestibular nuclei of normal and labyrinthectomised rats. J Physiol 2006;577:857–868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Fink K, Schlicker E, Neise A, Gothert M. Veratridine and other depolarizing agents counteract the inhibitory effect of Mg2+ ions on N‐methyl‐D‐aspartate (NMDA)‐induced noradrenaline release in vitro. Naunyn Schmiedebergs Arch Pharmacol 1990;342:513–519. [DOI] [PubMed] [Google Scholar]
- 25. Blandina P, Munari L, Provensi G, Passani MB. Histamine neurons in the tuberomamillary nucleus: A whole center or distinct subpopulations? Front Syst Neurosci 2012, Article number 33, and references herein. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Tiligada E, Kyriakidis K, Chazot PL, Passani MB. Histamine pharmacology and new CNS drug targets. CNS Neurosci Ther 2011;17:620–628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Humbert‐Claude M, Morisset S, Gbahou F, Arrang JM. Histamine H3 and dopamine D2 receptor‐mediated [35S]GTPγ[S] binding in rat striatum: Evidence for additive effects but lack of interactions. Biochem Pharmacol 2007;73:1172–1181. [DOI] [PubMed] [Google Scholar]
- 28. Garduno‐Torres B, Trevino M, Gutierrez R, Arias‐Montano JA. Presynaptic histamine H3 receptors regulate glutamate, but not GABA release in rat thalamus. Neuropharmacology 2007;52:527–535. [DOI] [PubMed] [Google Scholar]
- 29. Dai H, Fu Q, Shen Y, et al. The histamine H3 receptor antagonist clobenpropit enhances GABA release to protect against NMDA induced excitotoxicity through the cAMP/protein kinase A pathway in cultured cortical neurons. Eur J Pharmacol 2007;563:117–123. [DOI] [PubMed] [Google Scholar]
- 30. Threlfell S, Cragg SJ, Imre K, Turi GF, Coen CW, Greenfield SA. HistamineH3 receptors inhibit serotonin release in substantia nigra pars reticulata. J Neurosci 2004;24:8704–8710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Gemkow MJ, Davenport AJ, Harich S, Ellenbroek BA, Cesura A, Hallett D. The histamine H3 receptor as a therapeutic drug target for CNS disorders. Drug Discovery Today 2009;14:509–515. [DOI] [PubMed] [Google Scholar]
- 32. Esbenshade TA, Browman KE, Bitner RS, Strakhova M, Cowart MD, Brioni JD. The histamine H3 receptor: An attractive target for the treatment of cognitive disorders. Br J Pharmacol 2008;154:1166–1181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Sander K, Kottke T, Stark H. Histamine H3 receptor antagonists go to clinics. Biol Pharm Bull 2008;31:2163–2181. [DOI] [PubMed] [Google Scholar]
- 34. Stocking EM, Letavic MA. Histamine H3 antagonists as wake‐promoting and pro‐cognitive agents. Curr Top Med Chem 2008;8:988–1002. [DOI] [PubMed] [Google Scholar]
- 35. Letavic MA, Barbier AJ, Dvorak CA, Carruthers NI. Recent medicinal chemistry of the histamine H3 receptor. Prog Med Chem 2006;44:181–206. [DOI] [PubMed] [Google Scholar]
- 36. Celanire S, Wijtmans M, Talaga P, Leurs R, de Esch IJP. Keynote review: Histamine H3 receptor antagonists reach out for the clinic. Drug Discovery Today 2005;10:1613–1627. [DOI] [PubMed] [Google Scholar]
- 37. Letavic MA, Aluisio L, Atack JR, et al. Pre‐clinical characterization of aryloxypyridine amides as histamine H3 receptor antagonists: Identification of candidates for clinical development. Bioorg Med Chem Lett 2010;20:4210–4214. [DOI] [PubMed] [Google Scholar]
- 38. Hancock AA, Fox GB. Perspectives on cognitive domains, H3 receptor ligands and neurological disease. Expert Opin Investig Drugs 2004;13:1237–1248. [DOI] [PubMed] [Google Scholar]
- 39. Passani MB, Lin JS, Hancock A, Crochet S, Blandina P. The histamine H3 receptor as a novel therapeutic target for cognitive and sleep disorders. Trends Pharmacol Sci 2004;25:618–625. [DOI] [PubMed] [Google Scholar]
- 40. Vohora D. Histamine‐selective H3 receptor ligands and cognitive function: An overview. IDrugs 2004;7:667–673. [PubMed] [Google Scholar]
- 41. Fox MD, Snyder AZ, Vincent JL, Corbetta M, Van Essen DC, Raichie M.E. The human brain is intrinsically organized into dynamic, anticorrelated functional networks. Proc Natl Acad Sci USA 2005;102:9673–9678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Haas HL, Panula P. The role of histamine and the tuberomamillary nucleus in the nervous system. Nat Rev Neurosci 2003;4:121–130. [DOI] [PubMed] [Google Scholar]
- 43. Leurs R, Bakker RA, Timmerman H, de Esch IJ. The histamine H3 receptor: From gene cloning to H3 receptor drugs. Nat Rev Drug Discov 2005;4:107–120. [DOI] [PubMed] [Google Scholar]
- 44. Bongers G, Bakker RA, Leurs R. Molecular aspects of the histamine H3 receptor. Biochem Pharmacol 2007;73:1195–1204. [DOI] [PubMed] [Google Scholar]
- 45. Arrang JM, Morisset S, Gbahou F. Constitutive activity of the histamine H3 receptor. Trends Pharmacol Sci 2007;28:350–357. [DOI] [PubMed] [Google Scholar]
- 46. De Esch IJ, Belzar KJ. Histamine H3 receptor agonists. Mini Rev Med Chem 2004;4:955–963. [DOI] [PubMed] [Google Scholar]
- 47. Stark H, Kathmann M, Schlicker E, Schunack W, Schlegel B, Sippl W. Medicinal chemical and pharmacological aspects of imidazole‐containing histamine H3 receptor antagonists. Mini Rev Med Chem 2004;4:965–977. [DOI] [PubMed] [Google Scholar]
- 48. Mikó T, Ligneau X, Pertz HH, et al. Novel nonimidazole histamine H3 receptor antagonists: 1‐(4‐(phenoxymethyl)benzyl)piperidines and related compounds. J Med Chem 2003;46:1523–1530. [DOI] [PubMed] [Google Scholar]
- 49. Wijtmans M, Leurs R, de Esch I. Histamine H3 receptor ligands break ground in a remarkable plethora of therapeutic areas. Expert Opin Investig Drugs 2007;16:967–985. [DOI] [PubMed] [Google Scholar]
- 50. Berlin M, Boyce CW. Recent advances in the development of histamine H3 antagonists. Expert Opin Ther Pat 2007;17:675–687. [DOI] [PubMed] [Google Scholar]
- 51. Hudkins R, Raddatz R. Chapter 4 Recent advances in drug discovery of histamine H3 antagonists. Annu Rep Med Chem 2007;42:49–62. [Google Scholar]
- 52. Esbenshade TA, Fox GB, Cowart MD. Histamine H3 receptor antagonists: Preclinical promise for treating obesity and cognitive disorders. Mol Interv 2006;6:77–88. [DOI] [PubMed] [Google Scholar]
- 53. Berlin M, Boyce CW, de Lera Ruiz M. Histamine H3 receptor as a drug discovery target. J Med Chem 2011;54:26–53, and references herein. [DOI] [PubMed] [Google Scholar]
- 54. Wingen K, Stark H. Scaffold variations in amine warhead of histamine H3 receptor antagonists. Drug Discov Today Technol 2013;10:e483–e489, and references herein. [DOI] [PubMed] [Google Scholar]
- 55. Hancock AA. The challenge of drug discovery of a GPCR target: Analysis of preclinical pharmacology of histamine H3 antagonists/inverse agonists. Biochem Pharmacol 2006;71:1103–1113. [DOI] [PubMed] [Google Scholar]
- 56. Arias‐Montaño JA. Histamine H3 receptors and their role in basal ganglia physiology and pathophysiology. Sign Transduct 2007;7:364–371. [Google Scholar]
- 57. Leurs R, Vischer HF, Wijtmans M, De Esch IJP. En route to new blockbuster anti‐histamines: Surveying the offspring of the expanding histamine receptor family. Trends Pharmacol Sci 2011;32:250–257. [DOI] [PubMed] [Google Scholar]
- 58. Kuhne S, Wijtmans M, Lim HD, Leurs R, De Esch IJP. Several down, a few to go: Histamine H3 receptor ligands making the final push towards the market? Expert Opin Investig Drugs 2011;20:1629–1948. [DOI] [PubMed] [Google Scholar]
- 59. Arrang JM, Garbarg M, Lancelot JC, et al. Highly potent and selective ligands for histamine H3 receptors. Nature 1987;327:117–123. [DOI] [PubMed] [Google Scholar]
- 60. Garbarg M, Arrang JM, Rouleau A, et al. S‐[2‐(4‐Imidazolyl)ethyl]isothiourea, a highly specific and potent histamine H3 receptor agonist. J Pharmacol Exp Ther 1992;263:304–310. [PubMed] [Google Scholar]
- 61. Van der Goot H, Schepers MJP, Sterk GJ, Timmerman H. Isothiourea analogues of histamine as potent agonists or antagonists of the histamine H3 receptor. Eur J Med Chem 1992;27:511–517. [Google Scholar]
- 62. Ligneau X, Lin JS, Vanni‐Mercier G, et al. Neurochemical and behavioral effects of ciproxifan, a potent histamine H3‐receptor antagonist. J Pharmacol Exp Ther 1998;287:658–666. [PubMed] [Google Scholar]
- 63. Stark H, Hüls A, Ligneau X, et al. Development of FUB 181, a selective histamine H3‐receptor antagonist of high oral in vivo potency with 4‐(ω‐(arylalkyloxy)alkyl)‐1H‐imidazole structure. Arch Pharm (Weinheim) 1998;331:211–218. [DOI] [PubMed] [Google Scholar]
- 64. Onodera K, Miyazaki S, Imaizumi M, Stark H, Schunack W. Improvement by FUB 181, a novel histamine H3‐receptor antagonist, of learning and memory in the elevated plus‐maze test in mice. Naunyn Schmiedebergs Arch Pharmacol 1998;357:508–513. [DOI] [PubMed] [Google Scholar]
- 65. Stephanos J. Drug‐protein interactions: Two‐site binding of heterocyclic ligands to a monomeric hemoglobin. J Inorg Biochem 1996;62:155–169. [DOI] [PubMed] [Google Scholar]
- 66. Bongers G, Sallmen T, Passani MB, et al. The Akt/GSK‐3β axis as a new signaling pathway of the histamine H3 receptor. J Neurochem 2007;103:248–258. [DOI] [PubMed] [Google Scholar]
- 67. Greasley PJ, Clapham JC. Inverse agonism or neutral antagonism at G‐protein coupled receptors: A medicinal chemistry challenge worth pursuing? Eur J Pharmacol 2006;553:1–9. [DOI] [PubMed] [Google Scholar]
- 68. Walter M, Stark H. Histamine receptor subtypes: A century of rational drug design. Front Biosci (Schol Ed) 2012;S4:461–488. [DOI] [PubMed] [Google Scholar]
- 69. Schlegel B, Laggner C, Meier R, et al. Generation of a homology model of the human histamine H3 receptor for ligand docking and pharmacophore‐based screening. J Comput Aided Mol Des 2007;21:437–453. [DOI] [PubMed] [Google Scholar]
- 70. Celanire S, Lebon F, Stark H. Drug discovery: From hits to clinical candidates In: Vohora D, editor. The third histamine receptor: Selective ligands as potential therapeutic agents in CNS disorders. CRC Press, Taylor & Francis CRC Press Inc.: CRS Press, 2009;103–165. [Google Scholar]
- 71. Schwartz JC, Lecomte JM. Treatment of Parkinson's disease obstructive sleep apnea, dementia with Lewy bodies, vascular dementia with non‐imidazole alkylamines histamine H3‐ receptor ligands, U.S. Patent 2006; EP1707203.
- 72. Raddatz R, Tao M, Hudkins RL. Histamine H3 antagonists for treatment of cognitive deficits in CNS diseases. Curr Top Med Chem 2010;10:153–169. [DOI] [PubMed] [Google Scholar]
- 73. Black LA., Nersesian DL, Sharma P, et al. 4‐[6‐(2‐Aminoethyl)naphthalen‐2‐yl]benzonitriles are potent histamine H3 receptor antagonists with high CNS penetration. Bioorg Med Chem Lett 2007;17:1443–1446. [DOI] [PubMed] [Google Scholar]
- 74. Raddatz R, Hudkins RL, Mathiasen JR, et al. CEP‐26401 (irdabisant), a potent and selective histamine H3 receptor antagonist/inverse agonist with cognition‐enhancing and wake‐promoting activities. J Pharmacol Exp Ther 2012;340:124–133. [DOI] [PubMed] [Google Scholar]
- 75. Bitner RS, Markosyan S, Nikkel AL, Brioni JD. In‐vivo histamine H3 receptor antagonism activates cellular signaling suggestive of symptomatic and disease modifying efficacy in Alzheimer's disease. Neuropharmacology 2011;60:460–466. [DOI] [PubMed] [Google Scholar]
- 76. Tao M, Aimone LD, Huang Z, et al. Optimization of 5‐pyridazin‐3‐one phenoxypropylamines as potent, selective histamine H3 receptor antagonists with potent cognition enhancing activity. J Med Chem 2012;55:414–423. [DOI] [PubMed] [Google Scholar]
- 77. Wager TT, Pettersen BA, Schmidt AW, et al. Discovery of two clinical histamine H3 receptor antagonists: Trans ‐ N ‐ethyl‐3‐fluoro‐3‐[3‐fluoro‐4‐(pyrrolidinylmethyl)phenyl] cyclobutanecarboxamide (PF‐03654746) and trans ‐3‐fluoro‐3‐[3‐fluoro‐4‐ (pyrrolidin‐1‐ylmethyl)phenyl]‐ N ‐(2‐methylpropyl)cyclobutanecarboxamide (PF‐03654764). J Med Chem 2011;54:7602–7620. [DOI] [PubMed] [Google Scholar]
- 78. Ligneau X, Perrin D, Landais L, et al. BF2.649 [1‐{3‐[3‐(4‐Chlorophenyl)propoxy]propyl}piperidine, hydrochloride], a nonimidazole inverse agonist/antagonist at the human histamine H3 receptor: Preclinical pharmacology. J Pharmacol Exp Ther 2007;320:365–375. [DOI] [PubMed] [Google Scholar]
- 79. Łazewska D, Kieć‐Kononowicz K. New developments around histamine H3 receptor antagonists/inverse agonists: A patent review (2010‐present). Expert Opin Ther Pat 2014;24:89–111. [DOI] [PubMed] [Google Scholar]
- 80. Trenité DKN, Parain D, Genton P, Masnou P, Schwartz JC, Hirsch E. Efficacy of the histamine 3 receptor (H3R) antagonist pitolisant (formerly known as tiprolisant; BF2.649) in epilepsy: Dose‐dependent effects in the human photosensitivity model. Epilepsy Behav 2013;28:66–70. [DOI] [PubMed] [Google Scholar]
- 81. Bonaventure P, Letavic M, Dugovic C, et al. Histamine H3 receptor antagonists: From target identification to drug leads. Biochem Pharmacol 2007;73:1084–1096. [DOI] [PubMed] [Google Scholar]
- 82. Barbier AJ, Berridge C, Dugovic C, et al. Acute wake‐promoting actions of JNJ‐5207852, a novel, diamine‐based H3 antagonist. Br J Pharmacol 2004;143:649–661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Fox GB, Pan JB, Radek RJ, et al. Two novel and selective nonimidazole H3 receptor antagonists A‐304121 and A‐317920: II. In vivo behavioral and neurophysiological characterization. J Pharmacol Exp Ther 2003;305:897–908. [DOI] [PubMed] [Google Scholar]
- 84. Diaz JAM, Escribano BA, Martin UPE, Romanach MF. Cyclopropylmethanone derivatives, the preparation and therapeutic use thereof. Patent WO2005037810 (04/28/2005).
- 85. Curtis MP, Dwight W, Pratt J, et al. D‐Amino acid homopiperazine amides: Discovery of A‐320436, a potent and selective non‐imidazole histamine H3‐receptor antagonist. Arch Pharm 2004;337:219–229. [DOI] [PubMed] [Google Scholar]
- 86. Medhurst AD, Briggs MA, Bruton G, et al. Structurally novel histamine H3 receptor antagonists GSK207040 and GSK334429 improve scopolamine‐induced memory impairment and capsaicin‐induced secondary allodynia in rats. Biochem Pharmacol 2007;73:1182–1194. [DOI] [PubMed] [Google Scholar]
- 87. Investigational Drugs Database (IDdb), Thomson pharma partnering. http://partnering.thomson-pharma.com (accessed August 2009). [Google Scholar]
- 88. Medhurst AD, Atkins AR, Beresford IJ, et al. GSK189254, a novel H3 receptor antagonist that binds to histamine H3 receptors in Alzheimer's disease brain and improves cognitive performance in preclinical models. J Pharmacol Exp Ther 2007;321:1032–1045. [DOI] [PubMed] [Google Scholar]
- 89. Brioni JD, Esbenshade TA, Garrison TR, Bitner SR, Cowart MD. Discovery of histamine H3 antagonists for the treatment of cognitive disorders and Alzheimer's disease. J Pharmacol Exp Ther 2011;336:38–46. [DOI] [PubMed] [Google Scholar]
- 90. Bordi F, Rivara S, Dallaturca E, et al. Dibasic biphenyl H3 receptor antagonists: Steric tolerance for a lipophilic side chain. Eur J Med Chem 2012;48:214–230. [DOI] [PubMed] [Google Scholar]
- 91. Nagase T, Mizutani T, Sekino E, et al. Synthesis and evaluation of structurally constrained quinazolinone derivatives as potent and selective histamine H3 receptor inverse agonists. J Med Chem 2008;51:6889–6901. [DOI] [PubMed] [Google Scholar]
- 92. Mizutani T, Nagase T, Ito S, et al. Development of novel 2‐[4‐(aminoalkoxy)phenyl]‐4(3H)‐quinazolinone derivatives as potent and selective histamine H3 receptor inverse agonists. Bioorg Med Chem Lett 2008;18:6041–6045. [DOI] [PubMed] [Google Scholar]
- 93. Nagase T, Sato N, Kii S, Sato K, Tsuritani T, Sawada N. Process for production of 4(3H)‐quinazolinone derivative. PCT Int Appl 2006; WO 2006132424. [Google Scholar]
- 94. Jucaite A, Takano A, Boström E, et al. AZD5213: A novel histamine H3 receptor antagonist permitting high daytime and low nocturnal H3 receptor occupancy, a PET study in human subjects. Int J Neuropsychopharmacol 2013;16:1231–1239. [DOI] [PubMed] [Google Scholar]
- 95. Griebel G, Pichat P, Pruniaux MP, et al. SAR110894, a potent histamine H3‐receptor antagonist, displays procognitive effects in rodents. Pharmacol Biochem Behav 2012;102:203–214. [DOI] [PubMed] [Google Scholar]
- 96. Esbenshade TA, Browman KE, Miller TR, et al. Pharmacological properties and procognitive effects of ABT‐288, a potent and selective histamine H3 receptor antagonist. J Pharmacol Exp Ther 2012;343:233–245. [DOI] [PubMed] [Google Scholar]
- 97. Haig G, Meier A, Pritchett Y, Hall C, Gault L, Lenz R. Evaluation of the efficacy and safety of the H3 antagonist ABT‐288 in mild‐to‐moderate Alzheimer's disease. Alzheimers Dement 2012;8:P601–P602. [Google Scholar]
- 98. Haig GM, Bain E, Robieson W, Othman AA, Baker J, Lenz RA. A randomized trial of the efficacy and safety of the H3 antagonist ABT‐288 in cognitive impairment associated with schizophrenia. Schizophr Bull 2014. [Epub ahead of print] DOI: 10.1093/schbul/sbt240 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Wilson DM., Apps J, Bailey N, et al. Identification of clinical candidates from the benzazepine class of histamine H3 receptor antagonists. Bioorg Med Chem Lett 2013;23:6890–6896. [DOI] [PubMed] [Google Scholar]
- 100. Nathan PJ, Boardley R, Scott N, et al. The safety, tolerability, pharmacokinetics and cognitive effects of GSK239512, a selective histamine H3 receptor antagonist in patients with mild to moderate Alzheimer's disease: A preliminary investigation. Curr Alzheimer Res 2013;10:240–251. [DOI] [PubMed] [Google Scholar]
- 101. Herring WJ, Wilens TE, Adler LA. Randomized controlled study of the histamine H3 inverse agonist MK‐0249 in adult attention‐deficit/hyperactivity disorder. J Clin Psychiatry 2012;73:e891–e898. [DOI] [PubMed] [Google Scholar]
- 102. Decker M. Recent advances in the development of hybrid molecules/designed multiple compounds with antiamnesic properties. Mini Rev Med Chem 2007;7:221–229. [DOI] [PubMed] [Google Scholar]
- 103. Bajda M, Guzior N, Ignasik M, Malawska B. Multi‐target‐directed ligands in Alzheimer's disease treatment. Curr Med Chem 2011;18:4949–4975. [DOI] [PubMed] [Google Scholar]
- 104. Chen X, Decker M. Multi‐target compounds acting in the central nervous system designed from natural products. Curr Med Chem 2013;20:1673–1685. [DOI] [PubMed] [Google Scholar]
- 105. Coburg Y, Kottke T, Weizel L, Ligneau X, Stark H. Potential utility of histamine H3 receptor antagonist pharmacophore in antipsychotics serotonin receptors. Bioorg Med Chem Lett 2009;19:538–542. [DOI] [PubMed] [Google Scholar]
- 106. Petroianu G, Arafat K, Sasse BC, Stark H. Multiple enzyme inhibitions by histamine H3 receptor antagonists as potential procognitive agents. Pharmazie 2006;61:179–182. [PubMed] [Google Scholar]
- 107. Incerti M, Flammini L, Saccani F, et al. Dual‐acting drugs: An in vitro study of nonimidazole histamine H3 receptor antagonists combining anticholinesterase activity. ChemMedChem 2010;5:1143–1149. [DOI] [PubMed] [Google Scholar]
- 108. Apelt J, Ligneau X, Pertz H, et al. Development of a new class of non‐imidazole histamine H(3) receptor ligands with combined inhibitory histamine N‐methyltransferase activity. J Med Chem 2002;45:1128–1141. [DOI] [PubMed] [Google Scholar]
- 109. Grassmann S, Apelt J, Sippl W, et al. Imidazole derivatives as a novel class of hybrid compounds with inhibitory histamine N‐methyltransferase potencies and histamine H3 receptor affinities. Bioorg Med Chem 2003;11:2163–2174. [DOI] [PubMed] [Google Scholar]
- 110. Grassmann S, Apelt J, Ligneau X, et al. Search for histamine H(3) receptor ligands with combined inhibitory potency at histamine N‐methyltransferase: Omega‐piperidinoalkanamine derivatives. Arch Pharm (Weinheim) 2004;337:533–545. [DOI] [PubMed] [Google Scholar]
- 111. Apelt J, Grassmann S, Ligneau X, et al. Search for histamine H3 receptor antagonists with combined inhibitory potency at Ntau‐methyltransferase: Ether derivatives. Pharmazie 2005;60:97–106. [PubMed] [Google Scholar]
- 112. Bembenek SD, Keith JM, Letavic MA, et al. Lead identification of acetylcholinesterase inhibitors‐histamine H3 receptor antagonists from molecular modeling. Bioorg Med Chem 2008;16:2968–2973. [DOI] [PubMed] [Google Scholar]
- 113. Morini G, Comini M, Rivara M, et al. Synthesis and structure‐activity relationships for biphenyl H3 receptor antagonists with moderate anti‐cholinesterase activity. Bioorg Med Chem 2008;16:9911–9924. [DOI] [PubMed] [Google Scholar]
- 114. Darras FH, Pockes S, Huang G, et al. Synthesis, biological evaluation, and computational studies of tri‐ and tetracyclic nitrogen‐bridgehead compounds as potent dual‐acting AChE inhibitors and hH3 receptor antagonists. ACS Chem Neurosci 2014;5:225–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Huang W, Tang L, Shi Y, et al. Searching for the multi‐target‐directed ligands against Alzheimer's disease: Discovery of quinoxaline‐based hybrid compounds with AChE, H3R and BACE 1 inhibitory activities. Bioorg Med Chem 2011;19:7158–7167. [DOI] [PubMed] [Google Scholar]
- 116. Nikolic K. Theoretical study of phenolic antioxidants properties in reaction with oxygen‐centered radicals. THEOCHEM 2006;774:95–105. [Google Scholar]
- 117. Nikolic K. QSAR study of aromatic organochalcogens with glutathione peroxidase – like antioxidant activity. QSAR Comb Sci 2007;26:358–367. [Google Scholar]
- 118. Nikolic K. QSAR study of α‐tocopherol derivatives with chemotherapeutic activity against human breast cancer cells. THEOCHEM 2007;809:137–143. [Google Scholar]
- 119. Nikolic K. Design and QSAR study of analogs of alpha‐tocopherol with enhanced antiproliferative activity against human breast adenocarcinoma cells. J Mol Graph Model 2008;26:868–873. [DOI] [PubMed] [Google Scholar]
- 120. Nikolic K, Filipic S, Agbaba D. QSAR study of Imidazoline antihypertensive drugs. Bioorg Med Chem 2008;16:7134–7140. [DOI] [PubMed] [Google Scholar]
- 121. Nikolic K, Filipic S, Agbaba D. QSAR study of selective I1‐imidazoline receptor ligands. SAR QSAR Environ Res 2009;20:133–144. [DOI] [PubMed] [Google Scholar]
- 122. Nikolic K, Agbaba D. Design and QSAR study of analogs of γ‐tocotrienol with enhanced antiproliferative activity against human breast cancer cells. J Mol Graph Model 2009;27:777–783. [DOI] [PubMed] [Google Scholar]
- 123. Nikolic K, Agbaba D. QSAR study and design of novel organoselenium and α‐tocopherol derivatives with enhanced chemotherapeutic activity. Lett Drug Des Discov 2009;6:228–235. [Google Scholar]
- 124. Nikolic K, Agbaba D. Prediction of hepatic microsomal intrinsic clearance and human clearance values for drugs. J Mol Graph Model 2009;28:245–252. [DOI] [PubMed] [Google Scholar]
- 125. Nikolic K, Filipic S, Agbaba D. Multi‐Target QSAR and docking study of steroids binding to corticosteroid‐binding globulin and sex hormone‐binding globulin. Curr Comput Aided Drug Des 2012;8:296–308. [DOI] [PubMed] [Google Scholar]
- 126. Nikolic K, Agbaba D. Pharmacophore development and SAR studies of imidazoline receptor ligands. Mini Rev Med Chem 2012;12:1542–1555. [DOI] [PubMed] [Google Scholar]
- 127. Nikolic K, Veljkovic N, Gemovic B, Srdic‐Rajic T, Agbaba D. Imidazoline‐1 receptor ligands as apoptotic agents: Pharmacophore modeling and virtual docking study. Comb Chem High Throughput Screen 2013;16:298–319. [DOI] [PubMed] [Google Scholar]
- 128. Ivkovic BM, Nikolic K, Ilic BB, et al. Phenylpropiophenone derivatives as potential anticancer agents: Synthesis, biological evaluation and quantitative structure‐activity relationship study. Eur J Med Chem 2013;63:239–255. [DOI] [PubMed] [Google Scholar]
- 129. Filipic B, Nikolic K, Filipic S, et al. Identifying the CmbT substrates specificity by using a quantitative structure–activity relationship (QSAR) study. J Taiwan Inst Chem Eng 2014;45:764–771. [Google Scholar]
- 130. Nikolic K, Filipic S, Smoliński A, Kaliszan R, Agbaba D. Partial least square and hierarchical clustering in ADMET modeling: Prediction of blood – brain barrier permeation of α‐adrenergic and imidazoline receptor ligands. J Pharm Sci 2013;16:622–647. [DOI] [PubMed] [Google Scholar]
- 131. Chen PY, Tsai CT, Ou CY, et al. Computational analysis of novel drugs designed for use as acetylcholinesterase inhibitors and histamine H3 receptor antagonists for Alzheimer's disease by docking, scoring and de novo evolution. Mol Med Rep 2012;5:1043–1048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Quraishi S, Frangou S. Neuropsychology of bipolar disorder: A review. J Affect Disord 2002;72:209–226. [DOI] [PubMed] [Google Scholar]
- 133. Nierenberg AA, Keefe BR, Leslie VC, et al. Residual symptoms in depressed patients who respond acutely to fluoxetine. J Clin Psychiatry 1999;60:221–225. [DOI] [PubMed] [Google Scholar]
- 134. Fava GA, Fabbri S, Sonino N. Residual symptoms in depression: An emerging therapeutic target. Prog Neuropsychopharmacol Biol Psychiatry 2002;26:1019–1027. [DOI] [PubMed] [Google Scholar]
- 135. Milan M. Multi‐target strategies for the improved treatment of depressive states: Conceptual foundations and neuronal substrates, drug discovery and therapeutic application. Pharmacol Ther 2006;110:135–370. [DOI] [PubMed] [Google Scholar]
- 136. Keith JM, Gomez LA, Letavic MA, et al. Dual serotonin transporter/histamine H3 ligands: Optimization of the H3 pharmacophore. Bioorg Med Chem Lett 2007;17:702–706. [DOI] [PubMed] [Google Scholar]
- 137. Letavic MA, Keith JM, Jablonowski JA, et al. Novel tetrahydroisoquinolines are histamine H3 antagonists and serotonin reuptake inhibitors. Bioorg Med Chem Lett 2007;17:1047–1051. [DOI] [PubMed] [Google Scholar]
- 138. Barbier AJ, Aluisio L, Lord B, et al. Pharmacological characterization of JNJ‐28583867, a histamine H3 receptor antagonist and serotonin reuptake inhibitor. Eur J Pharmacol 2007;576:43–54. [DOI] [PubMed] [Google Scholar]
- 139. Keith JM, Gomez LA, Wolin RL, et al. Pyrrolidino‐tetrahydroisoquinolines as potent dual H3 antagonist and serotonin transporter inhibitors. Bioorg Med Chem Lett 2007;17:2603–2607. [DOI] [PubMed] [Google Scholar]
- 140. Letavic MA, Keith JM, Ly KS, et al. Novel naphthyridines are histamine H3 antagonists and serotonin reuptake transporter inhibitors. Bioorg Med Chem Lett 2007;17:2566–2569. [DOI] [PubMed] [Google Scholar]
- 141. Stocking EM, Miller JM, Barbier AJ, et al. Synthesis and biological evaluation of diamine‐based histamine H3 antagonists with serotonin reuptake inhibitor activity. Bioorg Med Chem Lett 2007;17:3130–3135. [DOI] [PubMed] [Google Scholar]
- 142. Letavic MA, Stocking EM, Barbier AJ, et al. Benzylamine histamine H3 antagonists and serotonin reuptake inhibitors. Bioorg Med Chem Lett 2007;17:4799–4803. [DOI] [PubMed] [Google Scholar]
