Gilberto Alves-MS-MRMC

12 downloads 0 Views 2MB Size Report
Mariana Matias1,2, Samuel Silvestre1,2, Amílcar Falcão2,3 and Gilberto Alves1,2* ...... [179] Rosini M, Simoni E, Bartolini M, Cavalli A, Ceccarini L, Pascu N,.
Send Orders for Reprints to [email protected] Mini-Reviews in Medicinal Chemistry, 2017, 17, 000-000

1

REVIEW ARTICLE

Recent Highlights on Molecular Hybrids Potentially Useful in Central Nervous System Disorders Mariana Matias1,2, Samuel Silvestre1,2, Amílcar Falcão2,3 and Gilberto Alves1,2* 1

CICS-UBI – Health Sciences Research Centre, University of Beira Interior, Rua Marquês d’Ávila e Bolama, 6201-001 Covilhã, Portugal; 2CNC – Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; 3Pharmacology Department, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal

ARTICLE HISTORY Received: January 02, 2016 Revised: May 26, 2016 Accepted: October 06, 2016 DOI: 10.2174/13895575176661611111101 21

Abstract: Molecular hybridization is a recent strategy based on the covalent fusion of two or more pharmacophores to create a single molecule with multiple mechanisms of action, which represents an encouraging approach in the development of new drugs with potential therapeutic application in several pathologies. This review provides a comprehensive perspective of the most relevant advances in the development of hybrid molecules acting in the central nervous system. For instance, several opioid hybrids based on endogenous opioid peptides (e.g. enkephalins, deltorphins and endomorphins) have been developed, and γ-aminobutyric acid (GABA) agonists have also been designed for neuropathic pain control. In addition, a number of hybrid compounds have also been synthesized and evaluated for their anticonvulsant activity and neurotoxicity, which may be further developed as potential antiepileptic drugs. Moreover, several hybrid compounds have also been designed for the treatment of neurodegenerative diseases focusing primarily on Alzheimer’s disease by targeting the cholinergic neurotransmission, as acetylcholinesterase inhibitors, and the amyloid β-protein deposition. There are also studies addressing hybrid compounds including an antioxidant moiety, which can be potentially useful in Alzheimer’s and Parkinson’s diseases and other neurodegenerative disorders. Additionally, other research works have also shown promising hybrid molecules for depression, autism and cocaine addiction. Thus, the development of molecular hybrid compounds seems to be a promising strategy in the discovery of novel therapeutic drugs.

Keywords: Alzheimer’s disease, central nervous system, epilepsy, molecular hybridization, pain, Parkinson’s disease. 1. INTRODUCTION Central nervous system (CNS) disorders comprise multiple diseases the symptoms of which range from cognitive impairment to maniac behavior or depression, and they affect millions of people worldwide. In developed countries due to the improvement of living conditions, longer life expectancy and consequent population ageing, the occurrence of CNS diseases is likely to increase in the next years, often accompanied by disability and decline in quality of life [1, 2]. These factors coupled with the diverse mechanisms and heterogeneous pathogenesis of this kind of diseases have emphasized the need to develop new and more effective therapies [3-5]. Most of the current single-target directed drugs have proven to be ineffective to achieve the therapeutic goals [6] and, therefore, novel strategies have *Address correspondence to this author at the Faculty of Health Sciences, University of Beira Interior, CICS-UBI – Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; Tel: +351 275 329002; Fax: +351 275 329099; E-mail: [email protected] 1389-5575/17 $58.00+.00

been used to develop new CNS-active drugs based on singlemolecular compounds that simultaneously influence multiple targets (multi-target directed drugs) [7]. These molecular compounds can be produced involving genetic strategies [8, 9], as well as by total or partial chemical synthesis [10]. A recent approach in medicinal chemistry that represents an encouraging research field in the development of new drugs is the design and preparation of molecular hybrids [11]. Hybrid molecules are constituted by two or more pharmacophores, natural or unnatural, which are covalently linked, forming a single compound capable to have one or multiple pharmacological actions. The combination of structural moieties allows obtaining molecules with potential dual activity, as a result of the union of distinct pharmacophores that can act simultaneously on the same or different pharmacological targets [12-15]. Indeed, these pharmacological agents are proving to be more efficacious, economical and safer than the single-target directed drugs [5, 14]. These molecules are usually classified in three different categories (A, B and C); the category A concerns a single © 2017 Bentham Science Publishers

2

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

target and both entities of the hybrid molecule are able to interact with the target; in the category B, the two entities of the hybrid molecule act independently on two different and nonrelated targets; and in the category C, both entities of the hybrid molecule act at the same time on two connected targets [14]. Moreover, the design of a hybrid molecule has some restrictions: (i) the molecules must contain two or more pharmacophores that exert biological actions via different binding sites; (ii) to be of clinical value the biological properties must be present in the same concentration range; and (iii) hybrid molecules must be resistant to the metabolic elimination processes, avoiding, after administration, the regeneration of the original drugs [16]. The hybridization approach has been applied in several fields, for example, to destroy microorganisms like bacteria and fungi that are becoming resistant to the common drugs used to treat infections [17-21]. For instance, a major problem concerning the drug resistance is the rapid emergence of multidrug-resistant malarial parasites that can be fatal to humans [22-24]. In this context, hybrid molecules to combat malaria have been synthesized with success and some of them have already been tested in clinical trials [25]. Also, hybridization techniques have been applied in the development of molecules useful against different kinds of cancers such as hormone-dependent breast cancer [26, 27], oropharyngeal, cervical and colorectal cancer [28] and others [29-33]. Some of these compounds have shown potent antiproliferative activities, promoting apoptosis in cancer cells and reducing toxicity against normal cells [34-36]. The design of hybrid molecules as potential cardiovascular drugs is also under development, for instance, to control hypertension and cardiovascular impairment and to prevent oxidative vascular damage [37]. Other research areas employing hybridization strategies include diabetes [38-40], cystic fibrosis [41], inflammation [42-45], chronic obstructive pulmonary disease [46], human immunodeficiency virus [47], gastrointestinal diseases [48], obesity [49], osteoporosis [50] and skin disorders [51]. There are also studies reporting the use of hybrid drugs with multiple biological properties [52], namely, molecules that can act simultaneously against malaria and human immunodeficiency virus [15] or those with antimicrobial and antitumor activities [53-56]. Specifically, this review is focused on and summarizes recent developments on molecular hybrid entities directed to the treatment of CNS-related disorders, in particular in those with interest for pain management, epilepsy, neurodegenerative diseases (e.g., Alzheimer’s disease and Parkinson’s disease) and other disorders. 2. PAIN MANAGEMENT Pain is an unpleasant sensory and emotional experience caused by the conscious awareness of a noxious stimulus. The perception of the intensity of pain is dependent on two pathways: the interactions between nociceptive and nonnociceptive impulses in the ascending system and the activation of the descending pain-inhibitory system [57]. Due to the clinical relevance of pain, great efforts have been invested in the characterization of peripheral and central mechanisms involved in the induction of nociception.

Matias et al.

Furthermore, there is an increasing awareness of the importance of cognitive/affective pain components [58, 59]. Acute and chronic pain can lead to a significant disability with social and economic implications [60]; these types of pain may have a variety of etiologies ranging from tissue injury caused by trauma or chronic diseases to health conditions in which there is no direct evidence of cause (e.g. fibromyalgia) [61]. As pain is always a subjective sensation, its inadequate assessment is frequently identified as one of the most significant barriers to effective pain treatment [62]. The choice of suitable analgesic drugs usually depends on the kind of pain (acute or chronic), intensity, duration of pain stimuli and etiology. Among the clinically available analgesic drugs, opioids are widely used for the management of chronic and acute pain. The clinical usefulness of opioids is, however, hampered by their side effects that can have a negative impact on the patients’ quality of life [63, 64]. Additionally, a diverse group of drugs that are often used for other clinical conditions may also have a great analgesic effectiveness as pain adjuvant therapies. For instance, several tricyclic antidepressants and antiepileptic drugs (AEDs) [65, 66] are widely used for the management of neuropathic pain; indeed, neuropathic pain is a type of chronic pain that leads to damage or other dysfunctions in the central and peripheral nervous systems and it is often refractory to the treatment with opioids, non-steroidal anti-inflammatory drugs and acetaminophen [67, 68]. Hence, due to the lack of satisfactory pharmacotherapeutic options available for chronic pain, this topic has gained an increasing interest in the context of drug research [69]. 2.1. Opioid/Opiate Analgesics The opioid system is the major endogenous pathway capable of modulation of pain signal transmission and perception. In this context, endogenous opioid peptides as enkephalins, endorphins, deltorphins and endomorphins are implicated in a variety of processes in the central and peripheral nervous systems, including the modulation of reward, pain and emotion [70]. The biological activities of these opioid peptides are mediated through three major opioid receptor types (µ, δ and κ) and the distinct molecular structure of these peptides has an impact on their selectivity for the different receptors and consequently on their activity profile [71, 72]. However, the clinical utility of naturally occurring opioid peptides is limited because of their poor bioavailability, susceptibility to enzymatic degradation and incapacity to penetrate the blood-brain barrier (BBB) [73, 74]. A promising approach to obtain a selective agonist by adopting a different conformation and increasing the enzymatic stability and activity can be the cyclization of linear active peptides. An example of this strategy was represented in a study by Ciszewska et al. [72] which investigated diverse cyclic opioid structures with different ring sizes. These peptides were evaluated in the µ receptorrepresentative guinea-pig ileum and δ receptor-representative mouse vas deferens assays, and it was shown that the hybridization through the extension of the enkephalin sequence at its C-terminus by a deltorphin fragment (Fig. 1) [75, 76] resulted in molecules with different receptor

Highlights on Molecular Hybrids for CNS Disorders

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

selectivity, favoring the interaction with the δ receptor and decreasing the affinity for µ receptor. Among the studied structures, the most active was peptide 1 (Fig. 1), in which an 18-membered ring was formed by incorporation of sidechain amino groups of D-lysine and diaminopimelic acid [72]. OH

H2N

N H

O

Other endogenous opioids largely studied are endomorphins 1 and 2 (Fig. 2) because they are highly potent and selective µ-opioid receptor agonist neuropeptides [77, 78]. Considering the fact that substance P (Fig. 2) is capable of intensifying opioid-mediated analgesia in low doses, Varamini et al. [79] conjugated the N-terminal amino acids of endomorphin-1 with the C-terminal fragments of substance P. A lipoamino acid was also attached to the Nterminal of endomorphin-1/substance P hybrid peptides to enhance their resistance against enzymatic hydrolysis and lipophilicity, consequently improving the passive diffusion of the peptides across biological membranes such as the BBB. In this study, the metabolic stability and membrane permeability were evaluated using the Caco-2 cell line. Among the compounds evaluated, structure 2 (Fig. 2) showed a relatively high µ-opioid receptor binding affinity and selectivity, and an improved membrane permeability and metabolic stability when compared with the parent peptides. In other research work, Mollica and co-workers [80] linked the endogenous opioid endomorphin-2 with a synthetic opioid peptide DAMGO (Fig. 2), being both structural moieties selective for the µ receptors. In this study, compound 3 (Fig. 2) with two N-methyl residues had the highest affinity and selectivity toward the µ-opioid receptors (in vitro receptor binding assay), a relevant activity in the µ receptor-representative guinea-pig ileum bioassay and also revealed to be the most potent structure in antinociceptive tests. This study confirmed that N-methylation is not only related with a relevant metabolic stability but it is also significantly associated with the bioactive conformation of endomorphine-2 analogues and with their interaction with the µ-opioid receptors.

S

O

H N

O

H N

OH

N H

O

O

Met-enkephalin

OH O

H N

H2N O

CH3

O

H N

N H

O

O

H N

N H

O

O

NH2

N H

O

OH Deltorphin I

OH O

H N

H2N

N H m NH

O O

N H

H N

m= 4

O HN

O H N

O

O

O

H N

N H

H N

N H

O

3

NH2 O

1

Fig. (1). Chemical structures of the analgesic peptide 1 and the endogenous opioids met-enkephalin and deltorphin I [72, 75, 76].

Another research work involved the synthesis of new hybrid molecules incorporating the fentanyl skeleton and a guanidine moiety from agmatine (Fig. 3) linked through an

NH HO

HO

HN

NH2 N O

NH

HN

NH2

NH2

N

O O

O

O

NH2

O O

NH O Endomorphin-2

Endomorphin-1

Arg-Pro-Lys-Pro-Gln-Gln-Phe-Phe-Gly-Leu-Met-NH2 Substance P

HO

NH2 O

H N

O

O N H

N O

OH

N H

DAMGO

H2N HO

NH

C8H17

O

HN

NH N O

NH O 2

O O

HO NH Phe-Gly-Leu-Met-NH2

Bn

NH2 N O

N

N O O

NH OH

O 3

Fig. (2). Chemical structures of compounds 2 and 3 (endomorphine-1 and endomorphine-2 analogs, respectively) and their precursors [7880].

4

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

Matias et al.

alkyl or an aromatic spacer [81]. This molecule series was developed based on the fact that fentanyl is a potent µ-opioid receptor agonist frequently used in the treatment of moderate to severe pain [82] and some evidence suggests that agmatine attenuates neuropathic pain in rats [83]. This study revealed that the presence of the guanidine and the Nphenethyl piperidyl propanamide core was necessary for these hybrid compounds to bind to I2-imidazoline binding site and µ-opioid receptors, both involved in analgesia mechanisms. Compound 4 (Fig. 3), which has six methylene units linking the two pharmacophores, showed to be a good µ-opioid agonist in binding studies; however, in in vivo assays, it did not produce significant analgesia [81].

O H N

N H2N N

NH2 NH

antiallodynic and antihyperalgesic activities. In this study, it was observed that compound 5a (Fig. 4) was the most active in the maximal electroshock seizure (MES) model and in the subcutaneous pentylenetetrazol (scPTZ) test, and revealed no neurotoxicity in the rotarod test. Compounds were also evaluated in two acute nociceptive mouse models, the acetic acid-induced writhing test and the formalin test. In these assays compound 5b (Fig. 4) was the most potent, leading to a 99% inhibition in acetic acid-induced writhing test. In the formalin test the most active chemical entities were the compounds 5c [for phase 1 (0-5 min), with 75% inhibition] and 5d [for phase 2 (10-30 min), with 80% inhibition] (Fig. 4). To assess the antihyperalgesic and antiallodynic activities two peripheral neuropathic pain models were also used: the chronic constriction injury and the L5 spinal nerve ligation; compound 5d completely reversed the spontaneous pain response in both models. These results suggested that the 5aryl substituted 1,2,4-triazole derivatives were the most interesting compounds due to their antihyperalgesic and antiallodynic properties [84].

Agmatine

Fentanyl

3. EPILEPSY O N

H N 6

NH NH2

N 4

Fig. (3). Chemical structures of fentanyl, agmatine and analgesic compound 4 [81]. OH

H2N

HN O

O

N R2

N

GABA

R1 Cl 5a: R1=

5b: R1=

R2= (CH2)3NH2

Br

R2= (CH2)3NH2 O

5c: R1= (CH2)3COOH

R2=

N O

5d: R1= (CH2)3COOH

R2=

Fig. (4). Chemical structure of GABA and examples of compounds studied for the control of neuropathic pain [84].

2.2. Analgesic Adjuvant Therapy One of the most important approaches to control chronic neuropathic pain involves the activation of γ-aminobutyric acid (GABA) (Fig. 4) mediated inhibitory neuronal pathways. Thus, as 1,2,4-triazole derivatives also showed antinociceptive activity, Yogeeswari et al. [84] synthesized GABA derivatives including the 1,2,4-triazol-2H-one nucleus and evaluated their anticonvulsant, peripheral analgesic,

Epilepsy is one of the most common serious neurological disorders [85] and affects approximately 50 million people worldwide [86, 87]. This disorder is characterized by recurrent seizures of cerebral origin, presenting episodes of sensory, motor or autonomic phenomena with or without loss of consciousness. The etiology of seizures is complex and remains uncertain and they can be caused by any type of brain pathology [88] or by oxidative injury [89]. Seizures may also arise by a consequence of interactions between predisposing genetic disturbances and pathological-induced changes [88]. The two most important neurotransmitters involved in the regulation of brain neuronal activity are glutamate (an excitatory neurotransmitter) and GABA (an inhibitory neurotransmitter). In fact, changes in the ratio of concentrations/activities of these neurotransmitters can either contribute to increase or decrease the propensity for seizures [90]. In this context, it has been documented that the reduction of GABAergic neuronal activity plays an important role in a number of neurological disorders, including epilepsy [91-93]. Unfortunately, brain GABA levels cannot be efficiently increased by peripheral administration of exogenous GABA because it does not cross the BBB due to its high hydrophilicity. However, other therapeutic strategies have been explored to potentiate the GABAergic neurotransmission; for instance, some of the currently available AEDs promote GABAergic neurotransmission through different ways such as the inhibition of GABA reuptake (e.g. tiagabine), the inhibition of GABA-transaminase (e.g. vigabatrin) and the allosteric modulation of GABA receptors (e.g. stiripentol) [94]. In fact, one approach used for the development of new AED candidates is based on the mechanism of action, making possible the development of GABA-mimetic drugs [95]. Several studies have also been performed focusing on glycine derivatives with potential anticonvulsant activity by interfering with glutamate levels [96, 97]. Moreover, the ion channels also have a critical role in the function of the CNS, where they are involved in the

Highlights on Molecular Hybrids for CNS Disorders

generation and conduction of nerve impulses by asserting control over the voltage potential across the plasma membrane [98]. Actually, voltage-gated sodium channels have been the molecular targets for several important commonly used drug classes, including hydantoin-related AEDs [99, 100]. Therefore, several studies involving the use of the phenytoin pharmacophore to prepare more potent hydantoin compounds, with less side effects and higher potency for inactivated sodium channels have been published [101, 102]. Over the years, great efforts have been made in the development of clinically effective drugs for the management of epileptic seizures. As previously stated, AEDs are used not only in the treatment of epilepsy but also in other clinical conditions such as analgesic adjuvant therapy. However, this group of drugs is highly susceptible to clinically significant drug-drug interactions [103], and is also associated with undesirable side-effects that can be life threatening [104]. In addition, 30% of the patients with epilepsy continue to have seizures despite the use of appropriately selected AEDs [105]. In view of these facts, there is a clear need to continue developing new AEDs with greater efficacy, namely against pharmacoresistant epilepsy, and/or improved tolerability profiles [101, 106, 107]. Hence, in this section several molecules developed through hybridization strategies are highlighted, which are divided according to the most studied pharmacophores including some of those found in drugs currently used in the clinical practice. 3.1. GABA Hybrids Riluzole (Fig. 5) [108], a functionalized benzothiazole derivative, is an inhibitor of sodium channels and it also inhibits the glutamate release, displaying anticonvulsant and neuroprotective properties [109]. Considering the relevance of this structural moiety, a study reported the incorporation of GABA-like pharmacophores on the benzothiazole nucleus to develop new molecules with synergistic anticonvulsant effects. Among the compounds prepared, the most promising was the N-(6-methoxybenzothiazol-2-yl)-4-oxo-4-phenylbutanamide, 6 (Fig. 5) with the median toxic dose (TD50) of 347.6 mg/kg, and with the median effective doses (ED50) of 40.96 mg/kg and 85.16 mg/kg in the MES and scPTZ tests, respectively. In addition, neither relevant hepatotoxic nor significant CNS depressive effects were observed with this compound [108]. Other studies have demonstrated the promising anticonvulsant activity exhibited by several phthalimide derivatives [110]. In this context, a relevant example was the work carried out by Ragavendran et al. [111], in which amide and acid hydrazone derivatives of phthalimido-GABA were developed by combining the pharmacophoric features of phthalimide, GABA and anilide, and phthalimide, GABA and hydrazones, respectively. Then, the anticonvulsant activity/potency of these two groups of hybrid compounds was evaluated in the MES, scPTZ, subcutaneous strychnine and intraperitoneal picrotoxin-induced seizure threshold tests, and their acute neurotoxicity was assessed through the rotarod assay. Based on the observed results, the authors found that phthalimide-GABA-anilide hybrids were more

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

5

effective than the corresponding phthalimide-GABAhydrazone hybrids, and the compound 7 (Fig. 5) evidenced the most potent anticonvulsant activity [111]. In another research study, supported by the fact that ameltolide (Fig. 5) [112] possesses potent anticonvulsant properties [113], a series of ameltolide-GABA-amides hybrids were developed and synthesized. In this series, the most potent derivative was the compound 8 (Fig. 5), which showed to be active in MES, subcutaneous strychnine and intraperitoneal picrotoxin-induced seizure threshold tests; moreover, at the anticonvulsant dose no neurotoxicity was evidenced when a direct comparison of doses with the standard AEDs was performed. Nevertheless, in these circumstances, a more reliable comparison between the compounds shall be performed using the respective protective index (TD50/ED50) instead of making a direct comparison of doses. Anyway, such as intended, the compounds synthesized had an increased lipophilicity when compared with the GABA and their pharmacological activity profiles suggest their BBB penetration [114].

F

F

O

O

S

F

N

N H

NH2 H2N

Ameltolide

Riluzole

O

O

H N O

S

H N

N

N

O

O

O

6

7

H N O

H N

N H

O 8

Fig. (5). Chemical structure of GABA hybrids and their precursors as well as riluzole and ameltolide [108, 111, 112, 114].

3.2. Benzodiazepine Hybrids Benzodiazepines (e.g. diazepam, Fig. 6) are widely used for CNS disorders, having different and varied therapeutic applications including sedation, sleep induction, reduction of anxiety and anticonvulsant effect [115]. The anticonvulsant activity of several benzodiazepines has been related with their high affinity binding to an accessory site (the “benzodiazepine binding site”) on the GABAA receptor chloride ionophore complex, thus facilitating GABA binding and enhancing the GABAergic neurotransmission and, therefore, limiting the sustained repetitive depolarization of the involved neurons [116, 117]. As the thiazolidine ring has also been associated with anticonvulsant properties, a combination of this nucleus and 1,5-benzodiazepines using molecular hybridization has been reported [118]. Hence, several compounds were synthesized and their anticonvulsant activity was evaluated against MES and isoniazid hydrazone induced seizures, whereas the impairment of motor activity was assessed using an actophotometer and a rotarod

6

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

Matias et al.

apparatus. In this series, compounds 9a and 9b (Fig. 6) evidenced the highest anticonvulsant activity. From the pharmacological data obtained it was concluded that small polar and electron rich groups in the phenyl ring directly bound to the benzodiazepine nucleus significantly contribute to the anticonvulsant activity. In addition, these compounds also showed interesting neuroprotective effects [118].

Cl

R

O

N

effective AEDs, Tripathi et al. [123] designed and synthesized a novel group of compounds combining aryl thiosemicarbazides with 4-(aryloxy)phenyl semicarbazones (Fig. 7). The most active compounds in this group were 10a that presented 100% protection in the 6 Hz test (a psychomotor seizure test), and 10b (Fig. 7) that exhibited anticonvulsant activity in both MES and 6 Hz tests. None of the compounds showed neurotoxicity at the highest administered dose (300 mg/kg). Taking into account these findings, it can be concluded that the introduction of halogens in the aryl ring appears to generate more active anticonvulsant compounds than the corresponding unsubstituted derivatives. Docking study results also suggested that these compounds exhibited good binding properties to the glutamate, GABAA delta and GABAA alpha-1 receptors [123]. Pandeya and co-workers [124] also linked semicarbazones with an isatin pharmacophore (Fig. 7) whose anticonvulsant activity has been studied [125]. All hybrids were evaluated in vivo by MES, scPTZ and subcutaneous strychnine and the most active compounds were examined for oral activity in rats at a dose of 30 mg/kg. Within this group, compound 11 (Fig. 7) exhibited interesting anticonvulsant activity in the MES test, with 50% protection at 0.5 h post dose and 100% protection after 2 h, without evidence of neurotoxicity [124]. This hybrid possesses a chlorine, which was associated to an improvement of the anticonvulsant activity when compared with other compounds without this substituent. More recently, Kumar et al. [126] synthesized 5,7-dibromoisatin semicarbazones and compounds 12a, 12b and 12c (Fig. 7) revealed a significant anticonvulsant activity (12a showed protection at the dose of 100 mg/kg at 4 h and compounds 12b and 12c showed activity at the dose of 30 mg/kg at 0.5 h in MES test). These hybrids were also tested for their CNS depressant effects by Porsolt’s force swim pool test and

H N

N

S N

N

O

NH

H N

Diazepam O

S 9a: R= 2-OH 9b: R= 4-(OCH3)

Fig. (6). Chemical structures of diazepam and some thiazolidinebenzodiazepine hybrids [115, 118].

3.3. Semicarbazones Hybrids Aryl thiosemicarbazides (Fig. 7) are a new class of compounds with reported anticonvulsant activity. These compounds do not contain the dicarboxamide group in their molecular structure, which is associated with toxic effects and is found in conventional AEDs such as barbiturates, oxazolidinones and phenytoin [119]. In addition, the structurally related semicarbazones also display interesting anticonvulsant activity [120-122]. In the search for more H N

R

H N

NH2

O O

N

Ar

O N H

O

S

4-(Aryloxy)phenyl semicarbazones

Aryl thiosemicarbazide

H N

R1

H N

H2N

H N

Isatin

H N

O O

N

N NH

S

R2

10a: R1= F 10b: R1= Br

Br

O N H

Cl R2= Cl R2 = Br

11

Br O Br R2 R1

N

O

NH NH

N O

N

N H

N H

N

H N

O

R3 13

12a: R1= Cl 12b: R1= F 12c: R1= F

R2= H R2= Cl R2= Cl

R3= H R3= H R3= CH3

Fig. (7). Chemical structures of the hybrids synthesized containing the semicarbazone moiety [123, 124, 126, 127].

Highlights on Molecular Hybrids for CNS Disorders

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

exhibited respectively an increase of 29.05, 51.24 and 32.95% in the immobility time. Similarly to a previous study, compound 12a with a p-chloro in its structure was found to be the most active entity in the CNS depression study [126]. Another research work presented a series of substituted N-(3-methylpyridin-2-yl) semicarbazones [127] and the compounds with a pyridine ring also possess anticonvulsant activity [128]. Among the compounds synthesized, compound 13 (Fig. 7) exhibited anticonvulsant protection after 0.5 h in the MES test (100 mg/kg), and in the subcutaneous strychnine-induced seizure model after 0.5 h and 4 h (100 mg/kg), and at 300 mg/kg in the subcutaneous picrotoxin seizure threshold test after 0.5 h and 4 h, without apparent neurotoxicity at the anticonvulsant doses [127]. 3.4. Pyrrolidone Hybrids The pyrrolidone nucleus is a cyclic imide system and constitutes the basic pharmacophore of several AEDs (e.g., levetiracetam and ethosuximide, Fig. 8). Taking this into account, the combination of this pharmacophore with a pyridine ring was described by Siddiqui et al. [129] aiming the development of new hybrid drugs. In this study, compounds 14a and 14b (Fig. 8) demonstrated comparable anticonvulsant activity to the standard drugs, with ED50 values of 13.4 and 18.6 mg/kg in the MES test and ED50 values of 86.1 and 271.6 mg/kg in the scPTZ test, respectively. None of these compounds exhibited any evidence of neurotoxicity and hepatotoxicity. Accordingly to the information discussed above for compounds 10a, 11, 12a, 12b and 12c, which also contain chloro atoms in their structure, it is not surprising that the structure-activity relationship studies conducted with pyrrolidone hybrids have also evidenced that compounds with a chlorinated substituent at fourth position of the pyridyl moiety are associated to a higher potency [129]. O

R1

N H2N O Levetiracetam

N

O N

O

R2

CN OH

HN

O O

Ethosuximide

14a: R1= Cl 14b: R1= NO2

R2= Cl R2= H

Fig. (8). Chemical structures of levetiracetam and ethosuximide containing pyrrolidone nucleus and pyridinyl-pyrrolidone hybrids [129].

3.5. Quinazolin-4(3H)-one Hybrids The discovery of methaqualone (Fig. 9) represented a relevant landmark in the field of synthetic anticonvulsant agents [130]. This compound contains the quinazolin-4(3H)one nucleus, a heterocyclic ring that is important for its anticonvulsant activity [131, 132]. In this context, using the

7

molecular hybridization strategy, Kumar et al. [133] prepared several molecules combining quinazolin-4-(3H)one (Fig. 9) with 4-(aryloxy)phenyl semicarbazones (Fig. 7) because in previous studies the (aryloxy)aryl semicarbazones showed evidence of anticonvulsant activity [134, 135]. Of the prepared hybrids, the most active was compound 15 (Fig. 9), which showed 100% protection in the 6 Hz test at the dose of 100 mg/kg and no neurotoxicity at the highest administered dose (300 mg/kg). Structure-activity studies evidenced, for example, that the methyl substituent at the position 2 of the quinazolin-4(3H)-one nucleus in methaqualone can be replaced by a n-propyl or even a phenyl group, also leading to more active CNS agents. As the previously described hybrids 10a and 10b (Fig. 7), docking studies also predicted that these compounds exhibited good binding properties with glutamate, GABA A delta and GABA A alpha-1 receptors [133]. In addition, the benzothiazole moiety was also incorporated in the quinazolin-4(3H)-one nucleus to obtain single molecules with dual activity. In this series, 16a (Fig. 9) showed significant anticonvulsant activity against the tonic extensor component in the MES test and 16b (Fig. 9) showed activity against the clonic phase of scPTZ-induced seizures. None of these compounds demonstrated any evidence of neurotoxicity or hepatotoxicity [136]. In another study, Malik and co-workers [137] designed and synthesized different molecules also linking the quinazolin-4(3H)-one nucleus and the benzothiazole moiety. The most potent compounds in this group were 17a and 17b (Fig. 9), having significant anticonvulsant potential and minimal toxicity compared to the standard drugs (phenytoin and ethosuximide) in the MES and scPTZ tests and rotarod assay after oral administration in rats at the doses of 30 mg/kg and 50 mg/kg, respectively. In fact, there is evidence that these compounds have slow but sustained absorption and prolonged duration of action. In the scPTZ test, compound 17b displayed an ED50 value of 510.5 µmol/kg and a TD50>2150 µmol/kg, thus determining a very promising protective index (TD50/ED50 >4.2) in comparison to that found with the standard drug ethosuximide (TD50/ED50 >3.0). On the other hand, in the 6-Hz screen test compound 17a revealed to be more active than 17b. Both compounds possess the unsubstituted quinazoline ring that seems to be favorable for their anticonvulsant potential. The presence of electron withdrawing groups like chloro (17a) and trifluoromethoxy (17b) on the benzothiazole ring is associated with a significant anticonvulsant activity and minimal toxicity when compared to reference drugs. These results are probably due to the involvement of GABA pathways as the GABA concentration increased in the rat brain at 2 h (92.1 µg/100 mg of tissue) and 7 days (111 µg/100 mg of tissue) after the treatment (compound 17a), and by antagonizing AMPA mediated excitatory neurotransmission as it was proved against AMPA-induced seizures (ED50 = 37.9 µmol/kg for clonic phase and ED50 = 28.5 µmol/kg for tonic phase) (compound 17b). In addition, both compounds did not lead to any significant increase or malfunctioning of hepatic enzymes in comparison with reference drugs [137]. In another recent research work, the quinazolinone nucleus was the basis of the anticonvulsant activity and the introduction of the benzyloxy tetrazole

8

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

Matias et al. O N N Methaqualone

R O

O N

Cl O

N

Br

S N

N

N

N 16a: R= H 16b: R= OCH3

15

R O

S N

O N H

N N H

S

N

N

O 17a: R= 6-Cl 17b: R= 6-OF3

N

O

O

19

O N N N N X

N

N

N

NH2

18a: X= CO 18b: X= SO2

N N N N

Fig. (9). Chemical structures of methaqualone and hybrid compounds containing the quinazolin-4(3H)-one nucleus [133, 136-138].

moiety as the core fragment synergized this activity. Within these hybrids, compounds 18a, 18b and 19 (Fig. 9) showed a significantly active profile at 0.5 h post-dose against electrically induced seizures in mice treated intraperitoneally with the dose of 30 mg/kg. Compound 18b also showed 100% of anticonvulsant protection in rats submitted to MES test at the same post-dose time point. These interesting activities have been associated not only to the heterocycles but also to the presence of free carbonyl (18a) and sulphonyl groups (18b). An amino fragment at the para-position of the benzene moiety in the compounds 18a and 18b also seems to be relevant. In addition, the 4-pyridinyl analogue 19 established protection against the scPTZ-induced seizures at the maximum dose of 300 mg/kg at 0.5 h, occurring a loss in activity after 4.0 h. All of these compounds seemed to be devoid of neurotoxicity, even at the maximum dose studied [138].

thiosemicarbazide derivative) and 21 (1,3,4-oxadiazole derivative) (Fig. 10) showed the highest protection (80%) in the scPTZ test in the dose of 100 mg/kg, which was equipotent to phenytoin. None of the compounds showed significant neurotoxicity and 20a was found to be devoid of sedative effects [102].  

3.6. Phenytoin Hybrids

Neurodegenerative diseases, such as Alzheimer’s, Parkinson’s and Huntington’s diseases and amyotrophic lateral sclerosis, are a group of pathologies characterized by a progressive and specific loss of determined brain cell populations [142]. The incidence of these diseases increase with ageing, affecting thereby more and more people worldwide [143]. These diseases lead to a considerable deterioration of the patients’ quality of life, having a high socio-economic impact and therefore represent enormous challenges to the scientific community [144]. Alzheimer’s and Parkinson’s diseases are the main neurodegenerative disorders addressed in this review because their high

Phenytoin (Fig. 10), a classic drug commonly used to control epilepsy, has been frequently considered as a starting point for the development of several new anticonvulsant drugs [139]. Using the hybridization approach, phenytoin was combined with several structural moieties associated with anticonvulsant properties such as thiosemicarbazide, 1,3,4-oxadiazole, 1,3,4-thiadiazole and 1,2,4-triazole rings. Among the synthesized compounds, only the phenyl substituted thiosemicarbazide derivative 20a (Fig. 10) revealed an anticonvulsant protection superior to phenytoin in the MES test at the dose of 30 mg/kg. Compounds 20b (a

A combined phenytoin-lidocaine structure was also developed as a novel sodium channel inhibitor [140], because both parent drugs inhibit the voltage-gated sodium channels that are involved in the pharmacological mechanisms to treat CNS-related disorders such as epilepsy, chronic pain and autism [141]. The compound 22 (Fig. 10) was considered to be the best phenytoin derivative, which presented a value of 71 ± 0.30% of inhibition in the [3H]-Batrachotoxin-B test [140]. 4. NEURODEGENERATIVE DISEASES

Highlights on Molecular Hybrids for CNS Disorders

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

9

O O

NH N

HN

N H

O

Lidocaine

Phenytoin

O O

O

O N

HN O

N H

H N

NHR S

N N

N

SH

NH C7H15

O

HN

20a: R= C6H5 20b: R= 4-OCH3C6H4

HN

O O

N H

O 21

N

22

Fig. (10). Chemical structures of phenytoin, lidocaine and some of their hybrids [102, 140].

incidence justifies an urgent need to find effective therapies to control them. However, after the following two sections, hybrid compounds that were designed to combat neurodegenerative diseases in general are also presented, which target basic pathological mechanisms underlying all of them (e.g. oxidative stress). 4.1. Alzheimer’s Disease Alzheimer’s disease (AD) is the most common agerelated neurodegenerative disorder and is characterized by progressive memory loss, language impairment, personality changes and decline in intellectual ability, which worsens as it progresses and eventually leads to death [145, 146]. The complex and multifaceted pathophysiology of this disease, which involves numerous pathways, hinders the development of satisfactory therapies. The etiology of AD includes deficiency in cholinergic neurotransmission, abnormal extracellular accumulation of amyloid β-protein (Aβ), changes in other neurotransmitter systems such as glutamatergic, serotonergic and dopaminergic neurons, and also the involvement of inflammatory, oxidative and hormonal pathways among others [147-149]. Up to date, an effective treatment to prevent the progression of AD is still unknown and the currently approved drugs such as acetylcholinesterase (AChE) inhibitors (tacrine, donepezil, rivastigmine and galantamine) and the N-methyl-D-aspartate (NMDA) antagonist memantine only seem to act as palliative therapeutic approaches to temporary ameliorating the cognitive impairment [146, 150]. The complexity and multiple etiologies of AD can explain the difficulty to obtain desirable therapeutic outcomes with single-target therapeutic strategies, which makes the choice of molecules with dual mechanistic activity one approach potentially more effective [150]. One of these strategies is the molecular hybridization that has been explored mainly using the tacrine pharmacophore to develop new drug candidates able to prevent and stop this neurodegenerative disease. In this review are presented several series of hybrids grouped according to the drugs that are currently being used to manage AD (e.g. tacrine and donepezil) and other important natural and synthetic pharmacophores.

4.2. Tacrine Hybrids Tacrine (Fig. 11) was the first approved cholinesterase inhibitor by the Food and Drug Administration (FDA) for the treatment of AD and it reversibly inhibits this enzyme. Despite its side effects (e.g. hepatotoxicity, gastrointestinal disturbances and hypotension), the search of tacrine-related compounds such as dimmers and hybrids is still of high interest because this compound is the most potent AChE inhibitor in clinical use [151-154]. One example is the combination of tacrine and donepezil, which will be discussed in the next section. Other recent example is the arrangement of methylenedioxybenzene or di- or trimethoxybenzene moieties with tacrine using a long chain linker [155]. This research work was based on the knowledge of the pharmacological properties of tacrine as well as on the fact that compounds containing a methylenedioxybenzene moiety revealed significant activity in inhibiting the selfinduced Aβ aggregation and the AChE enzyme. Within these developed structures, it was observed that compound 23a (Fig. 12), having a methylenedioxybenzene group and an eight-methylene chain linking the two aromatic units, was the most potent AChE inhibitor (IC50 = 7.98 ± 0.12 nM). Compound 23b (Fig. 12), with a trimethoxy substituted benzene moiety and six methylenic linker, exhibited the strongest butyrylcholinesterase (BuChE) inhibition (IC50 = 2.59 ± 0.14 nM); this compound (23b) has three methoxy groups on benzene ring, which seem to be important to BuChE inhibition. Compounds 23c and 23d (Fig. 12) were the most potent inhibitors of the self-mediated aggregation of Aβ42, the most amyloidogenic form of amyloid protein produced in the brain of patients with AD (67.13 ± 5.57% and 68.49 ± 3.63%, respectively) [155]. Considering that oxoisoaporphine synthetic derivatives have shown high AChE inhibitory activity and higher selectivity for AChE over BuChE [156], the combination of these compounds with tacrine was also evaluated. Within this group of compounds, it was found that the heterodimer 24a (Fig. 13) exhibited an interesting AChE inhibitory activity (IC50 = 3.4 ± 0.2 nM) when compared with tacrine, and compound 24b (Fig. 13) showed the highest BuChE inhibition (IC50 = 21.1 ± 1.0 nM). All the compounds in this series revealed a good inhibitory potency on the self-induced

10

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

Matias et al.

Aβ aggregation, and the derivative 24c (85.8 ± 2.2%) was the most active (Fig. 13). Compound 24a also showed to be potent as inhibitor of the AChE-induced Aβ aggregation (83.3 ± 1.2%). The capacity of the compounds to enter the CNS was also assessed using a parallel artificial membrane permeability assay for the BBB (PAMPA-BBB) and it was estimated that compound 24b could cross the BBB while compound 24c had low potential to access the brain [157]. NH2

N Tacrine

Fig. (11). Structure of tacrine [211]. HN

R1

n-2 N H

23a: R1= OCH2O 23b: R1= OCH3 23c: R1= OCH2O 23d: R1= OCH2O

R2 O

N

R2= OCH2O R2= OCH3 R2= OCH2O R2= OCH2O

n= 8 n= 6 n= 6 n= 9

Fig. (12). Structure of tacrine-metoxylated benzene derivatives with anti-Alzheimer activity [155]. N H N N

H N m2

24a: n= 2 m1= 2 m2= 2 24b: n= 2 m1= 3 m2= 3 24c: n= 2 m1= 1 m2= 4

O n

N H

O

m1

Fig. (13). Structure of tacrine-oxoisoaporphine derivatives [157].

Ferulic acid (Fig. 14) is one of the dominant natural phenolic acids and it has shown potent antioxidant properties [158]. Thus, Fang et al. [159] connected the tacrine and the ferulic acid by an alkylenediamine chain aiming to obtain multi-potent anti-Alzheimer drug candidates. In this series, compound 25 (Fig. 14) exhibited the highest inhibitory activity towards both AChE and BuChE, having a reversible and non-competitive inhibitory action for AChE and a reversible but competitive inhibitory action for BuChE. This hybrid also showed relevant antioxidant activity that depends not only on the ferulic acid substructure, but also on the chain length of the connecting linker [159]. The same research group developed a trihybrid based on tacrine-ferulic acid-nitric oxide with potential for the treatment of AD. As compared to the analog lacking the nitrate group, it became evident that the introduction of this group plays a role in the inhibition of electrophorus electricus AChE and equine serum BuChE. In fact, compound 26a (Fig. 14) showed a good inhibition for AChE (IC50 = 3.7 ± 0.6 nM) and BuChE (IC50 = 1.4 ± 0.4 nM) [160]. In addition, the trihybrid 26b (Fig. 14) seems to be very interesting because it can bind to the catalytic (CAS) and peripheral (PAS) sites of the AChE and BuChE enzymes in kinetic studies. This compound exhibited a moderate vasorelaxation in vitro probably due the release of nitric oxide, a better performance in improving

the scopolamine-induced cognition impairment in mice and less hepatotoxicity comparatively to tacrine. However, this compound had no antioxidant activity in the 1,1-diphenyl-2picrylhydrazyl (DPPH) assay, possibly due to the absence of free phenolic hydroxyl groups of the ferulic acid pharmacophore [160]. Additionally, caffeic acid (Fig. 14), an analog of the ferulic acid and a potent antioxidant, also plays an important role in neuroprotection and cytoprotection against oxidative stress [161-163]. Thus, using the hybridization approach, caffeic acid was also bound to tacrine or 6-chlorotacrine and the developed derivatives were biologically evaluated. The compound 27 (Fig. 14) was the most active hybrid as AChE inhibitor, binding to both CAS and PAS of the enzyme. In fact, the chloro-substituted tetrahydroacridine fragment was associated with an increased affinity toward AChE and may also increase the selectivity. These hybrid compounds also exhibited a radical scavenging activity higher than that observed with the individual constituents, as well as interesting inhibitory activities on the self- and AChE-induced Aβ aggregation [164]. Considering that some non-steroidal anti-inflammatory drugs may improve cognition and delay the progression of AD due to their anti-inflammatory and/or antiamyloidogenic activities [165], in some studies tacrinemefenamic acid and tacrine-flurbiprofen hybrids were developed (Fig. 15) [166, 167]. In fact, Bornstein and collaborators [168] by linking tacrine with mefenamic acid obtained an interesting series of compounds with dual activity in the inhibition of CAS and PAS of AChE and the compound 28 (Fig. 15) was the most important of the series with low nanomolar IC50 values under standard conditions (IC50 = 0.418 ± 0.025 nM) and in the presence of reactive oxygen species (ROS)- (IC50 = 0.009 ± 0.003 nM) [168]. In another recent work, a series of trihybrids composed by tacrine, flurbiprofen and nitrate was also prepared as potential therapeutic agent for AD [169]. The results revealed that the length of the alkyl connecting nitrate moiety and the bivalent hybrid (composed by tacrine and flurbiprofen) could significantly influence the AChE inhibitory activity. In fact, it was observed that when the length increases the activity decreases, and a two methylenic spacer was considered to be the optimal length. However, in this study, the most potent compound as AChE inhibitor was the hybrid 29 (Fig. 15), which exhibited an IC50 value (9.1 nM) higher than that observed for tacrine. The kinetic study of cholinesterase inhibition showed that this hybrid compound competes with acetylcholine for the same binding site (CAS) of the enzyme. Additionally, structure 29 afforded a moderate release of nitric oxide and exhibited a moderate blood vessel relaxative activity (28.6%), significant inhibitory effects on the Aβ formation, reducing 17% the levels of Aβ40, and also improved memory impairment in mice. Another important factor favorable to this compound comparatively to tacrine is its better safety profile exhibited in hepatotoxicity assays [170]. In another recent research work, Mao et al. [171] described the design, synthesis and evaluation of a series of o-hydroxyl and o-amino benzylamine-tacrine and o-hydroxy benzylamine-(7-chlorotacrine) hybrids. It was observed that

Highlights on Molecular Hybrids for CNS Disorders

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0 O

O

O HO

OH

HO

OH

HO Ferulic acid

Caffeic acid

O HN

O

nN

HN

H OH

n= 5

O

N

mN

H O

26a: m= 6 26b: m= 3

O

n ONO

2

O

N

25

HN

11

n= 4 n= 3

OH

nN

H

n= 3

OH N

Cl 27

Fig. (14). Structure of antioxidants ferulic and caffeic acids and their derivatives combined with tacrine [159, 160, 164]. HO

OH

O H N

O

F Mefenamic acid

Flurbiprofen

O

F O HN

H N

nN

O

O HN

H N

H

nN

H

n= 6

n= 7

Cl

ONO2

N

N 28

29

Fig. (15). Structure of non-steroidal anti-inflammatory drugs mefenamic acid and flurbiprofen and their hybrids with tacrine [168, 170].

the structures combining tacrine and substituted o-amino benzaldehyde were better AChE inhibitors than the corresponding hybrids of salicylaldehyde and tacrine. The pharmacological evaluation of these compounds evidenced that the AChE inhibitory potency is closely related to the length of the alkylene chain of the two blocks and the potency generally increases as the number of methylene groups increases. Moreover, the presence of substituted aromatic amino groups also seems to have a favorable effect on the AChE inhibitory activity. Accordingly, from these two series, compound 30a (o-amino benzylamine-tacrine hybrid) (Fig. 16), with a 9-carbon spacer linking the two pharmacophores, exhibited the greatest inhibitory potency towards human AChE (IC50 = 3.52 ± 0.057 nM) and compound 30b (o-hydroxyl benzylamine-tacrine hybrid) (Fig. 16), with an 8-carbon linker, had a good inhibitory potency too; these two derivatives also demonstrated a relevant BuChE inhibitory activity. In contrast to the AChE inhibitory potency, the antioxidant activity of these hybrid compounds decreased as the length of the carbon spacer increased and it was observed that substituted amino groups seem to have an unfavorable effect on the antioxidant properties. Moreover, compounds 30a and 30b had good

inhibitory properties against the Aβ aggregation and also good biometal-chelating ability [171]. R HN

nN H

N 30a: R= N(CH3)2 30b: R= OH

n= 9 n= 8

Fig. (16). Structure of o-amino benzylamine-tacrine and o-hydroxyl benzylamine-tacrine hybrids [171].

Tacripyrines are another relevant class of hybrid structures, which were synthesized from tacrine and nimodipine [172]. The last compound is a drug composed by a 1,4-dihydropyridine moiety that selectively blocks L-type voltage-dependent calcium channels [173, 174]. The nimodipine pharmacophore (Fig. 17) may be important in this context namely because it has been shown that cellular

12

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

Matias et al.

O2N O

O

O

OH

O

O

HN

H N

O

O O

N H

Carvedilol

Nimodipine

R X H N

N R1

S

O

NH2

N H

EtO2C

R2

N H

Phenylthiazole derivative

N

31a: X= CH 31b: X= N

O

H N

S

N H

O

n

N H

N

n= 4

S

nN

H

N

N H

N

N H

N

n= 1 n= 4

N

n= 3

O

H HO

34

N H

n

O

33a: R= H 33b: R= OCH3

N O

R= 4'-OCH3 R= H

H N

N

R

32

HN

N

N H

Cl 35

Fig. (17). Chemical structures of nimodipine, carvedilol and phenylthiazole derivatives and some of their tacrine hybrids [172, 178-180].

calcium dysfunction is also involved in pathogenesis of AD [175-177]. In this study, compound 31a (Fig. 17), one of the most potent hybrids in human AChE inhibition (IC50 = 105 ± 15 nM), was found to be a weak inhibitor of the self-induced Aβ42 aggregation (34.9 ± 5.4%). Molecular modeling studies suggested that the binding of the compound 31a to the AChE PAS mainly involves the R-enantiomer. On the other hand, compound 31b (Fig. 17) was found to be the most potent hybrid as calcium-channel blocker (49% inhibition), whose activity was similar to that obtained with nimodipine. Both compounds were able to cross the BBB and afforded a good degree of neuroprotection when compared to tacrine [172]. Knowing that the phenylthiazole nucleus (Fig. 17) could inhibit tau protein aggregation and may also lead to selfinduced Aβ aggregation blockade, a new series of phenylthiazole–tacrine hybrids was also synthesized. Among them, compound 32 (Fig. 17), with a spacer chain length of ten atoms (no counting N atoms), displayed the highest AChE inhibitory potency binding simultaneously to the CAS and the PAS of AChE, and compound 33a (Fig. 17) showed the highest potency and selectivity for BuChE. In addition, compound 33b (Fig. 17) effectively prevented the selfinduced Aβ42 aggregation, but had no relevant blockage effects on calcium-channels [178]. In another study, the carbazole moiety of carvedilol (Fig. 17), a vasodilating βblocker with antioxidant properties, was linked to the chloro-

substituted tetrahydroacridine moiety of 6-chlorotacrine. Within this group it was observed that compound 34 (Fig. 17), having a three-methylene spacer, showed to be long enough to allow a proper interaction with both sites of the AChE. It also exhibited in vitro inhibitory activity against the self-induced Aβ aggregation (36.0 ± 2.3%) and AChEmediated aggregation (57.7 ± 6.1%). In addition, it revealed a relevant antagonistic effect on the NMDA receptors and was able to protect neuronal cells against the ROS generation evoked by oxidative stress [179]. Additionally, Thiratmatrakul and co-workers [180] also described new tacrine-carbazole hybrids potentially useful as multifunctional anti-Alzheimer agents. Among the compounds synthesized, structure 35 (Fig. 17), with a 5-methylene linker, exhibited the most potent inhibitory activity and selectivity toward electric eel AChE (IC50 = 0.48 ± 0.14 µM) as well as relevant antioxidant activity. The high AChE inhibition produced by this compound was related with its methoxyl group at the 7-position of the carbazole moiety. On the other hand, the presence of this group decreased the BuChE inhibitory activity. Docking studies also indicated that the derivative 35 can bind to both CAS and PAS of the AChE. Furthermore, this compound had a potent antioxidant activity, protecting cell damage against oxidative stress induced by hydrogen peroxide, and had protective effect against Aβ1-42 toxicity in a cell model at the concentration of

Highlights on Molecular Hybrids for CNS Disorders

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

100 µM. Moreover, this compound exhibited an ability to improve both short- and long-term memory deficits in mice induced by scopolamine through enhancement of cholinergic signaling [180]. Considering that the benzothiazole moiety is present in compounds that interfere with Aβ peptides [181], Keri et al. [182] conjugated tacrine with this pharmacophore through different suitable linkers. From the pharmacological evaluation studies, compound 36a (Fig. 18), with a five carbon linker, appeared to be the strongest AChE inhibitor with an IC50 value of 0.34 ± 0.1 µM, which is quite similar to the IC50 value obtained for tacrine. Structure-activity studies evidenced that the presence of a simple phenyl group attached to the benzothiazole moiety is associated with the highest inhibitory capacity. Indeed, molecular modeling studies predicted the AChE dual-binding site of this compound (36a). Concerning the self-induced Aβ42 aggregation inhibition, compound 36b (Fig. 18) presented the highest activity (61.3% inhibition). None of these compounds exhibited significant antioxidant activity, possibly because of the absence of phenolic hydroxyl groups [182]. Cl

H N

O

O N H

5

N

X

S

n

N

N N H

8N

H

O N

36a: X= Ph 36b: X= PhCH2

37a: R= Cl 37b: R= H

n= 0 n= 2

R H N

N

H N 9

OH

N 38

Fig. (18). Chemical structures of hybrids synthesized from tacrine and benzothiazole and 5-phenylpyrano[3,2-c]quinoline moieties [182-184].

New hybrid compounds were also developed connecting a 5-phenylpyrano[3,2-c]quinoline moiety with 6-chlorotacrine through an oligomethylene linker containing an amido group at a variable position. The most potent human AChE inhibitor was compound 37a (Fig. 18) with an IC50 value of 7.03 ± 0.3 nM. Molecular modeling and kinetic studies suggested that this compound is able to bind the PAS and the CAS of human AChE. On the other hand, the Aβ antiaggregating effect seems to depend on the position of the amido group within the linker and one of the most potent compounds in the AChE-induced and self-induced Aβ aggregation was compound 37b (45.7 ± 0.3 and 47.3 ± 8.8%, respectively) (Fig. 18). This fact was mainly associated to its two-methylene linker between the amido group and the phenylpirano[3,2-c]quinoline moiety that facilitates the

13

formation of hydrogen bonds with Aβ leading to an increased affinity. This compound was also the most potent β-secretase inhibitor (78% inhibition) and both compounds (37a and 37b) appeared to be able of penetrating the BBB [183]. More recently, the efficacy of a tacrine-8hydroxyquinoline derivative 38 (Fig. 18) was evaluated using in vitro and in vivo models. This compound showed neuroprotective effects in neuronal and astrocytic cell cultures, had anti-amyloid effects in the APP/Ps1 mouse (an AD model), and had also neuroprotective effects in wildtype mice submitted to domoic acid-induced toxicity (an experimental model of neurodegeneration) [184]. A very recent investigational study involved the design and development of tacrine-2,4,5-triphenyl-1H-imidazole hybrids (tacrine-lophine hybrids) as inhibitors of AChE and BuChE. Taking into account all the compounds prepared, the most active as AChE inhibitor was hybrid 39 (IC50 = 5.87 ± 0.3 nM; Fig. 19), which bears an octane chain between the tacrine and lophine moieties (Fig. 19). This result points out that this linker will allow an optimal interaction between the aromatic fragments of the compound and the CAS and PAS of AChE. In addition, it was also observed that a chloride substituent in the para-position of the imidazolic 2-phenyl group is associated with an increased inhibitory activity against BuChE [185]. More studies reported the combination of tacrine with other different natural pharmacophores. Examples of the last compounds include berberine or huprine derivatives that are subsequently discussed. Additionally, the preparation of hybrids linking tacrine derivatives to rhein was also reported [186]. Rhein (Fig. 20) is the main component of rhubarb, a traditional Chinese herb, which has different pharmacological effects such as antioxidant activity and probably positive effects on AD [187]. It has also hepatoprotective effects [188] that can be useful to reduce the above referred liver toxicity caused by tacrine. Among the compounds synthesized, the hybrid 40a (Fig. 20) possessed the most potent inhibitory activity against electric eel AChE (IC50 = 22.0±1.5 nM) and 40b (Fig. 20) showed the highest inhibition against equine serum BuChE (IC50 = 11.0 ± 0.7). On the other hand, compound 40c (Fig. 20) exhibited the strongest inhibition of the AChE-induced Aβ aggregation (70.2% inhibition). Additionally, this last compound showed to be a good metal chelator by effectively chelating Cu2+ and Fe2+ and demonstrated to be safer than tacrine in hepatotoxicity studies [186]. Tacrine was also linked to flavonoids aiming to obtain multi-target molecules against AD (Fig. 20). In this series, structure 41a was the most potent inhibitor for electric eel AChE (IC50 = 8.4 ± 0.8 nM) while 41b showed the strongest inhibition to equine serum BuChE (IC50 = 25.8 ± 0.8 nM). Compound 41c, with a hydroxyl group at the 5position of flavonoid moiety and without the ring at the 2position, was the most potent inhibitor of the Aβ selfinduced aggregation. Structure-activity relationship studies evidenced that an electron-donating group at this position seems to be beneficial to the Aβ self-aggregation inhibitory activity. Compounds 41c and 41d may have metal-chelating ability since they have the 5-hydroxy-4-keto site and, thus, they can serve as metal chelators in treating AD [189].

14

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

H N

[191] that synthesized a series of new hybrids; among these compounds, structures 44a and 44b (Fig. 22) showed better values than the parent drugs in inhibiting human AChE and BChE and in the AChE-induced Aβ40 aggregation [191].

N

N

HN

N

Matias et al.

n-3

n= 8

N

Lophine

Cl 39

Fig. (19). Structures of lophine and the tacrine-lophine hybrid 39 [185]. OH

O

O

OH HN OH

O

nN

H m

O

R

OH

N

Rhein

HN

O

O

40a: R= Cl 40b: R= H 40c: R= H

n= 6 n= 6 n= 6

m= 2 m= 3 m= 2

O

O

R2

OH

N N

N 41a: R1= OCH3 41b: R1= H 41c: R1= OH 41d: R1= OH

R2= C5H6 R2= C5H6 R2= C5H6 R2 = H

R1

O

Fig. (20). Structures of rhein and hybrids developed from the linkage of tacrine with natural compounds such as rhein and flavonoids [186, 189]. O O O

N Donepezil

Fig. (21). Structure of donepezil [190].

4.3. Donepezil Hybrids Donepezil (Fig. 21) is another drug extensively used in the development of hybrid compounds with potential to treat AD. As indicated above, Alonso et al. [190] described a new series of donepezil-tacrine hybrids, combining tacrine (Fig. 11), 6-chlorotacrine or acridine with the indanone (or phthalimide, a similar group) moiety of donepezil (Fig. 21). Within this series, compounds 42 and 43 (Fig. 22), bearing the phthalimide moiety and a long chain containing respectively ten and nine atoms linking the two heterocyclic systems, exhibited an interesting AChE inhibitory activity (IC50 = 2.8 nM and IC50 = 2.4 nM, respectively) mediated by binding simultaneously to the PAS and CAS of the enzyme [190]. Afterwards, the combination of these two potent molecules was also studied by Camps and collaborators

The compound AP2238 (Fig. 23) was the first published structure designed to bind to both the CAS and PAS of human AChE [192]. Thus, considering its interesting properties, Rizzo et al. [193] described several compounds linking the indanone core of donepezil with the phenyl-Nmethylbenzylamino moiety of AP2238 through a double bond. Instead of the indanone system, an analogous tetralone substituent was also evaluated in this study. An interesting enzyme inhibitory activity was observed when a methoxy group was bound at the position 6 of the tetralone ring, probably because this substituent appeared to be crucial to the interaction of the hybrid compound with the enzyme. The replacement of this methoxy substituent by a pentoxy substituent carrying different amines, e.g. diethylamine (45a) or piperidine 45b (Fig. 23) resulted in a relevant increase in the activity. In fact, both compounds were the most active of this series as AChE inhibitors (IC50 = 0.056 ± 0.003 µM and IC50 = 0.052 ± 0.002 µM, respectively); they also showed a high human BuChE inhibitory activity, but evidenced lower selectivity than donepezil and AP2238. In addition, both compounds 45a and 45b were also able to inhibit the AChEinduced Aβ aggregation (26.4 ± 1.1% and 26.9 ± 3.4%, respectively) and exhibited some degree of inhibitory activity against the self-aggregation of Aβ42 peptide (42.9 ± 0.8% and 48.3 ± 0.9%, respectively), while the reference compounds were completely ineffective [193]. There is also evidence suggesting that the use of monoamine oxidase (MAO) inhibitors might be a valuable approach for the treatment of AD; indeed, MAO inhibitors can lead to a reduction in the formation of neurotoxic products and consequently to a reduction of neuronal damage [194]. Thus, a new strategy appeared combining the benzylpiperidine moiety of donepezil and the indolyl propargylamino moiety of N-[(5-benzyloxy)-1-methyl-1Hindol-2-yl)methyl]-N-methylprop-2-yn-1-amine (Fig. 24), a MAO inhibitor. Among the developed compounds, structure 46a (Fig. 24) was found to be the most promising hybrid revealing to be a potent inhibitor of both MAO-A (IC50 = 5.2 ± 1.1 nM) and MAO-B (IC50 = 43 ± 8.0 nM) and also a moderately potent inhibitor of AChE (IC50 = 0.38 ± 0.05 µM) and BuChE (IC50 = 1.7 ± 0.2 µM). These results pointed out the relevant role played by the 1-benzylpiperidin-4-yl moiety existing in the structure of donepezil and in these hybrids in the AChE inhibition. Moreover, compound 46a was considered a moderate inhibitor of the self-induced Aβ aggregation and prevents the human AChE-induced Aβ aggregation with an inhibitory potency similar to donepezil and significantly higher than tacrine [195]. Additionally, Bautista-Aguilera and co-workers [196] intending to develop donepezil indolyl based amines, amides and carboxylic acid derivatives, linked the N-benzylpiperidine moiety of donepezil with the indole nucleus of compound 46a by a three methylene carbon chain and evaluated their activity in inhibiting the cholinesterase and MAO. In this study, structure 46b (Fig. 24) showed to be a promising drug

Highlights on Molecular Hybrids for CNS Disorders

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

O N

N H

N

N H

15

O O N

N

N H

O

N

5

O Cl

43

42

X

O O

N NH

44a: X= O

R= Cl

44b: X= H,H

R= H

R N

Fig. (22). Structures of donepezil-tacrine hybrids [190, 191]. O

O

O

O R (H2C)5 O

O

N

N AP2238

45a: -NEt2 45b:

N

Fig. (23). Structures of the compound AP2238 and the hybrids incorporating donepezil pharmacophore [193]. R N

O

N

N O

N

BnN

N-[(5-benzyloxy)-1-methyl-1H-indol-2-yl)methyl]-N-methylprop-2-yn-1-amine

46a: R= CH3 46b: R= H

Fig. (24). Structures of N-[(5-benzyloxy)-1-methyl-1H-indol-2-yl)methyl]-N-methylprop-2-yn-1-amine and their hybrids with donepezil [195, 196].

candidate with a potent and selective electrophorus electricus AChE (IC50 = 0.19 ± 0.01 µM) and human MAOA (IC50 = 0.0055 ± 0.0014 µM) inhibition. This compound also displayed moderate equine serum BuChE (IC50 = 0.83 ± 0.16 µM) and human MAO-B (IC50 = 0.15 ± 0.031 µM) inhibitory activities. Docking studies predicted that its linear conformation allows to span both the CAS and PAS site of electrophorus electricus AChE, contributing to its superior binding. It was also observed that the propargylamine group is likely to be an important feature for these derivatives in order to exhibit both AChE- and BuChE-inhibitory activities. For MAO enzymes, molecular modeling studies suggested that the selectivity of compound 46b to MAO-A vs MAO-B is likely to be due to the orientation of their propargylamine and phenyl moieties in the interaction with the enzymes [196]. In another work, new hybrids were designed by combining the N-benzylpiperidine moiety of donepezil with an anti-Alzheimer 2-aminopyridine system, and these two

structural moieties were connected by appropriate polymethylene binding spacers. Among these, compound 47a (Fig. 25), with a hydrogen at the C4 of the pyridine ring, was found to be the most potent and selective hybrid in inhibiting the human AChE (IC50 = 0.0094 ± 0.0004 µM) and BuChE (IC50 = 6.6 ± 0.7 µM). This compound is able to bind simultaneously to the CAS and PAS of AChE and it can also cross the BBB by passive diffusion [197]. The N-benzylpiperidine moiety of donepezil was also linked with 2-chloropyridine heterocyclic ring systems through a polymethylene linker. Within this group, compound 47b (Fig. 25), bearing again a hydrogen at C4, was the most active, being almost equipotent with donepezil as AChE inhibitor (IC50 = 0.013 ± 0.002 µM) and 4-fold less active as BuChE inhibitor (IC50 = 8.13 ± 0.41 µM). Moreover, a permeability study using a BBB-PAMPA assay also indicated that this compound would probably cross the BBB by passive diffusion and docking studies performed suggested that the compound possesses the optimal spacer length that allows a strong interaction with human AChE [198].

16

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

NC N

O S

R

O

5

NBn

N H NH

47a: n= 3 47b: n= 4

O

R=NH2 R= Cl

49

Fig. (25). Chemical structures of compounds synthesized from Nbenzylpiperidine moiety of donepezil and 2-aminopyridine/2chloropyridine pharmacophores [197, 198].

Fig. (27). Chemical structure of the hybrid 49 constituted by Lglutamic acid, N-benzylpiperidine moiety and a lipophilic α-hexyl ester [202].

Hu et al. [199] also developed a new series of hybrids using a multimethylene linker to connect the dimethoxyindanone unit of donepezil with the hupyridone fragment of huperzine A (Fig. 26); indeed, huperzine A is a novel lycopodium alkaloid that has shown to be a potent, reversible, selective and well-tolerated AChE inhibitor [200, 201]. The diastereomeric mixture (RS,S)-48 (Fig. 26), with a tetramethylene linker, exhibited the strongest AChE inhibition of this series of compounds, having an IC50 value of 9 nM, and no relevant BuChE inhibitory effect was observed [199].

O NH H2N

O

Cl

CN

nN H

BnN

Matias et al.

H N

O

O

In another study, three active groups were joined to Lglutamic acid as a suitable biological linker. The three pharmacophoric groups were a ω-situated N-benzylpiperidine moiety able to bind the CAS of AChE, a N-protecting group capable of interacting with the PAS in order to inhibit Aβ aggregation, and a lipophilic α-hexyl ester important for the crossing of the BBB. The prepared compounds showed good inhibitory activity against human AChE and BuChE, particularly compound 49 (IC50 = 0.10 ± 0.01 µM and IC50 = 0.07 ± 0.01 µM, respectively) (Fig. 27), and were able to bind to the PAS of AChE and consequently inhibit the Aβ fibril formation promoted by this enzyme. The entry in the brain was studied using a PAMPA-BBB assay and the results suggested that the molecules could cross the BBB, probably by passive diffusion. The compounds were also found to be neuroprotectants against both exogenous and mitochondrial ROS [202].

O

O (H2C)4

Huperzine A

NH

48

4.4. Berberine Hybrids

Fig. (26). Chemical structures of huperzine A and its hybrid compound linked to donepezil moiety [199].

Berberine (Fig. 28), a natural isoquinoline alkaloid, has evidenced potential to treat AD [203-205]. Considering that

O

Cl N

O

O O Berberine

OH

O

Br N

O

50a: R=

O

O n

O

n= 4

R 50b: R=

OH

n= 3

O

N

H N

N

O

N

n

O

O

Br

O

S O 51a: n= 3 51b: n= 6

Fig. (28). Chemical structures of berberine hybrids [209, 210].

N

O 52

Highlights on Molecular Hybrids for CNS Disorders

phenol derivatives as ferulic acid (Fig. 14) and melatonin have a potent antioxidant activity [206-208], Jiang et al. [209] prepared a series of hybrids combining these pharmacophores in the search of multifunctional agents for AD. From all prepared structures, the berberine-pyrocatechol 50a (Fig. 28) was found to be a potent AChE inhibitor (IC50 = 0.123 ± 0.003 µM) and a strong BuChE inhibitor (IC50 = 2.09 ± 0.14 µM) and the hydroquinone berberine hybrid 50b (Fig. 28) presented the highest inhibition of the Aβ aggregation (92%). In an antioxidant activity assay, most of the berberine-benzenediols showed higher activity than berberine-ferulic acid and berberine-melatonin hybrids, probably due to the presence of a phenolic hydroxyl moiety, and compound 50b, possessing a hydroxyl at the paraposition on the benzene ring, had the strongest antioxidant activity (9.54 Trolox equivalents) [209]. Additionally, Huang et al. [210] designed, synthesized and evaluated several berberine-phenyl-benzoheterocyclic and tacrine-phenyl-benzoheterocyclic hybrids as multifunctional anti-Alzheimer agents. Structure-activity relationship studies demonstrated that the potency for AChE inhibition was closely related to the length of the alkylene chain and a 3carbon spacer seem to be the most effective in both series. Accordingly, compound 51a (Fig. 28), combining tacrine and phenyl-benzothiazole linked by a 3-carbon spacer chain, was found to be the most potent AChE inhibitor, with an IC50 value of 0.017 ± 0.002 µM. Hence, it was observed that tacrine-phenyl-benzoheterocyclic hybrids were more potent as AChE inhibitors than berberine-phenyl-benzoheterocyclic hybrids. Compound 51b (Fig. 28) was the most potent as BuChE inhibitor (IC50 = 0.082 ± 0.007 µM). In addition, all the berberine-phenyl-benzoheterocyclic hybrids exhibited greater Aβ42 aggregation inhibitory activities than curcumin and berberine, and within this group the compound 52 (Fig. 28) was the most potent (IC50 = 3.61 ± 0.09 µM) [210]. 4.5. Huprine Hybrids In order to allow the simultaneous interaction with both binding sites of the AChE enzyme, tacrine was connected to huprine Y (Fig. 29), a natural compound with one of the highest affinities for the active site of AChE already reported [211]. Taking into account the data of this study, it was found that the introduction of a protonatable amino group in the linker resulted in an additional increase in the AChE inhibitory activity, probably due to an extra interaction with the AChE gorge as a third recognition site. An unsubstituted tacrine unit 53 (Fig. 29) together with an adequate tether length appeared to have a good inhibitory activity against human AChE. In addition, the presence of a protonatable amino group in the linker seemed to have a detrimental effect on the inhibitory activity of the BuChE, thus explaining the selective effect of these compounds [212]. The series of huprine-tacrine heterodimers continues being explored to find hybrids that have a potential diseasemodifying role in the treatment of AD [213]. Viayna and collaborators [214] attached two pharmacophores belonging to two natural compounds in order to prepare a family of rhein-huprine hybrids. Among others, these authors obtained an interesting racemic hybrid

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

17

54 (Fig. 29) that was extensively studied. In in vitro assays, it showed to be a potent drug candidate as inhibitor of the human AChE, being the (-)-54 isomer (IC50 = 2.39 ± 0.17 nM) much more potent than the (+)-54 (IC50 = 2930 ± 285 nM). On the other hand, (+)-54 (IC50 = 265 ± 21 nM) was two-fold more potent than (-)-54 (IC50 = 513 ± 58 nM) in inhibiting the BuChE. Contrary to what was observed for the human AChE and BuChE inhibition, in the self-induced Aβ aggregation and β-secretase inhibitory activity (IC50 = 120 ± 90 nM) no significant differences were found between the (+)- and (−)-54 isomers. Ex vivo studies in brain slices of C57bl6 mice conducted with the potential leads (+)- and (−)54 revealed that the compounds efficiently protected against the synaptic failure induced by acute treatments with Aβ42 oligomers. Moreover, they exerted a protective effect on synaptic proteins related with the stability of the synapses and its plasticity in the hippocampus. Additionally, in vivo studies in APP-PS1 transgenic mice treated intraperitoneally for 4 weeks with (+)- and (−)-54 revealed that they were able to reduce the levels of hippocampal total soluble Aβ and to increase the levels of mature amyloid precursor protein [214].

NH2 N Huprine Y

Cl HO O HO

HN HN

O

N

HN HN

8

N

9

O

N Cl 53

Cl 54

Fig. (29). Structures of huprine Y and their analogues [212, 214].

4.6. Choline Hybrids Knowing that phenolic compounds such as caffeic acid (Fig. 14), rosmarinic acid (Fig. 30) and trolox have antioxidant properties, hybrids conjugating the choline group of acetylcholine (Fig. 30) with these antioxidant compounds were also developed. In this group, compound 55, the 3,4dimethoxycinnamic choline ester (Fig. 30), was the strongest AChE inhibitor (IC50 = 7.3 ± 0.8 µM). The rosmarinyl- and trolox-choline derivatives showed a much lower AChE inhibitory activity than that found for caffeic acid analogues, possibly due to the bulkiness and/or the hydroxyl groups of the first two structural groups that may form unfavorable interactions with the enzyme, whereas the methoxy groups in caffeic acid derivatives can form favorable interactions. Regarding antioxidant activity, compound 56 (Fig. 30) appeared to be the strongest antioxidant with an EC50 value

18

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

Matias et al.

of 4.3 ± 0.2 µM. This fact is probably due to the presence of phenolic hydroxyl groups, which have an important role in the ROS-scavenging action of polyphenols [215]. O HO

O

OH

O

HO

OH

N OH Rosmarinic acid OH

Choline

O HO

O O O

O

N

O

O

N

O

HO OH

56

55

OH

Fig. (30). Chemical structures of phenolic-choline hybrids and their precursors [215].

4.7. Parkinson’s Disease Parkinson’s disease (PD) is the second most common neurodegenerative disorder [216] and it is characterized by loss of dopamine neurons in substantia nigra pars compacta and later in the ventral tegmental area. Possible causes for the neurodegeneration include oxidative stress, misfolded proteins, inflammation, mitochondrial and ubiquitinproteasome dysfunction and impaired protection against potentially harmful substances [217-219]. Some of the symptoms associated with PD involve rigidity, bradykinesia, tremor and postural instability along with cognitive and psychiatric complications [220, 221]. The dopamine precursor L-DOPA (levopoda) has been widely used in the treatment of PD. However, among other disadvantages, the L-DOPA side effects limit its therapeutic use [222]. Over the years, several strategies have been studied to enhance

L-DOPA chemical stability and water or lipid solubility, and also to reduce its susceptibility to enzymatic degradation [223, 224]. For the treatment of PD other options have also been explored such as the use of indirect dopaminergic agents, like selective MAO-B and cathecol-O-methyltransferase inhibitors, and anticholinergic and antiglutamatergic agents [222, 225]. Nevertheless, among the different therapeutic agents developed, direct dopamine agonist therapy still plays an important role in the treatment of this disease. Although the drug therapy of PD seems to be more successful compared to that of AD, the therapeutic approaches currently available do not prevent the degenerative process and consequently the progression of the disease [226]. In this context, D3 receptor appears to be a promising target for the treatment of PD and several therapeutic approaches have been developed towards this dopamine receptor [222]. An example is given by Dutta et al. [227] that, based on the fact that some aminotetralins and disubstituted piperazine derivatives have already demonstrated high selectivity for D3 receptors [228, 229], linked these two pharmacophores as well as some bioisosteric analogues. A preliminary study showed that the length of the linker connecting the aminotetralin moiety with the piperazine fragment plays an important role in the selectivity to D3 receptors. Indeed, a two methylene linker chain was associated with a higher selectivity and potency for the D 3 receptors in comparison with D2 receptors [227]. In a further study by the same research group, several hybrid compounds were also prepared based on the structure of the D3 agonists pramipexole and 7-hydroxy-2-(dipropylamino) tetralin (7OH-DPAT) (Fig. 31) [230]; the results of binding assays indicated that (±)-57 (Fig. 31) has high potency for the D 3 receptor and it was the most selective compound. In addition, the isomer (-)-58 (Fig. 31) presented higher D3 binding potency and selectivity compared to the corresponding (+)-58 isomer. The isomer (-)-58 was also evaluated in vivo in 6hydroxydopamine induced unilaterally lesioned rats and showed high activity and higher duration of action comparatively to apomorphine, the reference compound [231].

N H2N

HO

S

N H Pramipexole

N

N

N 7-OH-DPAT

N

N

H2N S

N

HO

N

57

58 N

N

59a: R=

H2N S

N

N

N R

59b: R=

Fig. (31). Chemical structures of the PD hybrids and their precursors [231, 232].

N

N

Highlights on Molecular Hybrids for CNS Disorders

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

The synthesis and evaluation of D3 agonist hybrids with antioxidant activity and potential interest in PD continues to be reported [232]. In addition to D2 and D3 receptors binding assays using HEK-293 cells, the functional activity of some of these compounds in stimulating GTPγS binding was determined in CHO cells expressing human D2 receptors and in AtT-20 cells expressing human D3 receptors. Taking into account the results of all assays, compounds 59a and 59b (Fig. 31) were pointed out as the most interesting hybrids in this work. In fact, compound 59a, an isoquinoline-1-yl derivative, showed a relevant and preferential agonist activity for D3 receptors (Ki = 2.23 nM). In contrast, compound 59b, a quinoline-5-yl derivative, displayed high binding affinity for both D2 and D3 receptors (Ki = 57.7 nM and Ki = 1.21 nM, respectively) despite appreciable selectivity for D3 over D2 receptors. By means of a DPPH radical scavenging assay, these two compounds also exhibited potent and dosedependent radical scavenging activity (IC50 = 45.67 ± 6.89 µM for 59a and IC50 = 39.67 ± 6.23 µM for 59b). Moreover, both compounds showed high activity in two PD animal models, with compound 59b being the most potent in the reserpinized rat model, whereas compound 59a was the most active in the neurotoxin 6-hydroxydopamine induced unilaterally lesioned rat model [232]. 4.8. Other Hybrids for Neurodegenerative Diseases

the

Treatment

of

The oxidative stress is involved in the ageing process and contributes to the pathological changes associated with the initiation and progression of neurodegenerative disorders such as AD, PD, amyotrophic lateral sclerosis and Huntington’s disease [233, 234]. For this reason and as previously referred, several research works have been performed aiming the development of hybrid structures containing antioxidant pharmacophores potentially useful in all these neurodegenerative conditions. A relevant example is the work of Yoo et al. [235] in which a series of cinnamoyl ketoamide hybrids were synthesized combining antioxidants and calpain inhibitors. Calpain is a calciumactivated cysteine protease typically associated with cellular necrosis and, in some neurological disorders, it was demonstrated that calpains are overactivated originating serious cell death. Thus, the inhibition of this enzyme has been considered one of the strategies to treat neurodegenerative diseases [236-238]. Among the prepared hybrids, compound 60 (Fig. 32) was the most potent as µ-calpain inhibitor (IC50 = 0.13 µM) and also exhibited strong antioxidant activities in DPPH, superoxide anion radical scavenging and lipid peroxidation inhibition assays [235]. In addition, in order to obtain compounds with dual inhibition of lipid peroxidation and cellular calpain, Auvin et al. [239] prepared hybrid structures formed from the 2-hydroxy-tetrahydrofuran group, a masked aldehyde selected as a calpain pharmacophore, linked to a different antioxidants [239, 240]. In another study intending to obtain potent antioxidant compounds, Koufaki et al. [241] synthesized hybrids containing the chroman moiety of vitamin E and a catechol group and evaluated the influence on antioxidant activity of catechol groups at positions 2 or 5 of the chroman nucleus. The interesting compound 61 (Fig. 33) protected against

19

hydrogen peroxide-induced cellular damage (IC50 = 1 ± 0.1 µM), possibly due to its iron chelating properties, and compound 62 (Fig. 33) possessed a strong neuroprotective activity in glutamate-induced cell death of HT22 cells (EC50 = 0.93 ± 0.19 µM) that could be related not only to their antioxidant activity but also to their capacity to interfere with other cell signaling cascades implicated in oxytosis [241]. In later studies, it was combined the chroman nucleus of vitamin E and catechol derivatives, linked by heterocyclic five-membered rings such as 1,2,4-oxadiazole, 1,3,4oxadiazole, 1,2,3-triazole, tetrazole and isoxazole. Among the 2-substituted chroman analogues, compound 63 (Fig. 33), in which a 3,4-dimethoxyphenyl moiety is directly attached to the 1,2,4-oxadiazole ring, evidenced the highest antioxidant activity in the protection of glutamate-challenged HT22 cells (antiproliferative MTT assay: EC50 = 254 ± 65 nM) in the series of 2-substituted chroman analogues. In the same assay and concerning the 5-substituted chroman analogues, the isoxazole derivative 64 (Fig. 33) exhibited the strongest antioxidant activity, protecting from glutamateinduced oxidative cell destruction (antiproliferative MTT assay: EC50 = 245 ± 38 nM); however, this compound was cytotoxic, probably due to the presence of the chatecol moiety, while the triazole analogue 65 (Fig. 33) in spite of being less effective as antioxidant (EC50 = 801 ± 229 nM) was non-cytotoxic in all tested concentrations [242].

O HO

O N H

O

HO 60

N H

O

OH

Fig. (32). Chemical structures of the hybrid 60 having a strong antioxidant activity and µ-calpain inhibition, both important to the treatment of neurodegenerative diseases [235].

As previously mentioned, the oxidative deamination mediated by human MAOs produces highly reactive peroxides that may contribute to the oxidative damage observed in some neurodegenerative disorders [243, 244]. Considering this fact, a hybrid scaffold was developed combining the hydrazine moiety of iproniazid (Fig. 34), a known irreversible MAO inhibitor [245], and the thiazole nucleus of glitazone drugs (e.g. pioglitazone or rosiglitazone, Fig. 34), whose neuroprotective properties are also related to their ability to selectively inhibit the MAO-B [246]. Despite none of these compounds tested were able to inhibit the human MAO-A, the resulting hybrid structures demonstrated to be highly selective as human MAO-B inhibitors, with compounds 66a (IC50 = 350.03 ± 26.12 nM) and 66b (IC50 = 851.32 ± 64.78 nM) (Fig. 34) being the most potent ones [247]. 5. OTHER CNS DISEASES Depression is a CNS disorder usually related with a dysfunction of the central serotonergic neurotransmission and, therefore, the selective serotonin reuptake inhibitors

20

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

Matias et al.

OH O

OH

HO

NH

HO

O

OH

O

OH

O

N

O

O

O N

O 63

62

61 OH OH

OH

N O

N N N

OH

HO HO O O 64

65

Fig. (33). Chemical structures with antioxidant activity incorporating chroman moiety of vitamin E and a catechol group [241, 242]. O

N

N H

O

H N

O

HN O

Iproniazid

N S Pioglitazone COOEt

O

O

HN O

N N

N

N

R2 R1

S Rosiglitazone

N H

S S

66a: R1= CH3

R 2=

66b: R1= H

R 2=

N H

Fig. (34). Chemical structures of hybrids developed from hydrazine moiety of iproniazid and thiazole nucleus of glitazone drugs and these precursors [247].

appeared to be one of the most important class of antidepressant agents [248]. In this regard, Orús et al. [249] described the development of new hybrid structures with affinity for both the 5-HT1A receptor and the serotonin (5-HT) transporter. This study was based on coupling of structural moieties related to inhibition of serotonin reuptake, such as benzo[b]-thiophene derivatives to arylpiperazines, typical 5-HT1A receptor ligands. All of these compounds showed high affinity for both targets and structure-activity relationship analysis showed that the presence of a heteroatom in the aryl group bound to piperazine ring and its position appears to be critical for the affinity of 5-HT1A receptors, with the compounds derived from 8-quinolyl showing the best results [249]. More recently, a new drug named vilazodone, 5-{4-[4-(5-cyano-3-indolyl)-butyl]-1piperazinyl}2benzofuran-2-carboxamide, 67 (Fig. 35), was approved by the FDA for the treatment of major depressive disorder in adults [250, 251]. Interestingly, this drug is a hybrid with activity as selective serotonin reuptake inhibitor and as 5-HT1A receptor agonist, which resulted of the combination of a 5-cyano-indole-butyl-amine and

2-carboxamidebenzofurane-5-yl-piperazine moieties with a four-carbon-saturated linker between the indole and piperazine rings. This chain originated the optimal configuration for the high 5-HT1A receptor affinity and the introduction of the cyano group in 5-position on the indole ring increased serotonin transporter affinity [252, 253]. Cocaine addiction represents a public health problem and still does not exist pharmacotherapy approved that can lessen its abuse [254]. In a study intending to develop useful drugs for treating cocaine addiction, it was reported the preparation and pharmacologic evaluation of a series of (bisarylmethoxy)butylpiperidine hybrids having dual activity at the dopamine transporter and dopamine/serotonin receptor sites. Within all structures studied, compound 68 (Fig. 36) showed the highest affinity for dopamine transporter and it was observed that in this series, when a large halogen atom (Br) was introduced at the 4-position of the aromatic ring bound to piperidine, the selectivity and binding potency to this transporter were improved. This compound also showed a moderate dopamine D2/D3 receptor antagonism and

Highlights on Molecular Hybrids for CNS Disorders

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

produced a significant increase in spontaneous locomotor activity in mice with an ED50 value of 2.42 mg/kg. Compound 69 (Fig. 36) evidenced to be the strongest binding ligand at the dopamine transporter (Ki = 1.32 ± 0.49 nM) and also showed moderate activity for the D2/D 3 receptor sites [255].

21

(Fig. 38) with a three carbon linker between the imidazole and the heterocyclic moiety, showing, in fact, high H 3 receptor affinity (Ki = 4.1 nM in [125I]Iodoproxyfan binding assay for human H3 receptors stably expressed in CHO cells) and high histamine N-methyltransferase inhibitory activity (IC50 = 0.024 ± 0.004 µM in histamine N-methyltransferase assay on isolated enzyme from rat kidney) [260].

O

NC

CONH2

O

N

O

O

N H

N

F 3C

67 70

Fig. (35). Chemical structure of vilazodone [252].

71 O

F

F

F

O

F N

N

N H

N

O 4

O

N

O

N

N H

N

F3C

HN

O

O

4 N Cl

OH

72

N

O NH

Br 68

Cl

Fig. (37). Chemical structures of the proposed hybrids as antiautism drug candidates [257].

69

Fig. (36). Chemical structures of the compounds synthesized to combat cocaine addiction [255].

Autism is a neurodevelopmental disorder that causes lifelong impairment and the serotonergic system may underlie the etiology of this disorder [256]. Using the tethering technique, a library of virtual ‘hybrid’ molecules incorporating 5-HT reuptake inhibitors and antagonists of the 5-HT1B/1D auto-receptors was created as anti-autism drug candidates. Then, proposed compounds with high fit values were selected for further synthesis and subsequent in vitro biological evaluation. Examples of these proposed compounds include the chemical entities 70, 71 and 72 (Fig. 37), and it was observed that the preliminary in vitro data are promising and consistent with the prediction studies [257]. Finally, the histamine H3 receptor has received particular attention over the past decade as a promising therapeutic target in a large variety of CNS diseases, such as neurodegenerative and psychiatric disorders. This has happened because this receptor can regulate the release of other important neurotransmitters such as acetylcholine, dopamine, norepinephrine and serotonin, which are involved in the pathogenesis of CNS diseases [258, 259]. In this context, a novel series of imidazole-containing compounds was synthesized and evaluated combining structural characteristics of both H3 receptor antagonists and histamine N-methyltransferase inhibitors [260]. In the development of these structures it was considered that potent H3 receptor antagonists containing the imidazole ring [261] and some heterocycles (e.g. tacrine) are also associated to Nmethyltransferase inhibition [262]. The most potent of these hybrid compounds was the aminoquinoline derivative 73

N N

N H N H

73

Fig. (38). Chemical structure of the hybrid with H3 receptor affinity and histamine N-methyltransferase inhibitory activity [260].

6. CONCLUSION The disorders that affect the CNS such as pain, epilepsy and neurodegenerative diseases have a complex and multifaceted pathogenesis involving several biochemical pathways. Over the years, due to their relevance, the molecular and cellular mechanisms underlying these CNS diseases are emerging, along with potential therapeutic targets. In this context, the development of new disease models is offering increasing opportunities to assess the potential of novel drug candidates. A relatively recent and increasingly relevant medicinal chemistry strategy for the design of therapeutic molecules with a multifunctional nature and potential application in these disorders is the hybridization approach. In fact, molecules consisting of two or more pharmacophores that are able to act through different action mechanisms are being developed aiming to find compounds with increased potencies and reduced side effects as well as improved pharmacokinetic profiles. In fact, an increasing number of studies are demonstrating that several hybrid molecules are revealing high efficacy and low toxicity in in vitro and in vivo models. In this review some of

22

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

these research works are discussed in order to highlight and compare new drug candidates that can improve the therapeutic outcomes in patients suffering from CNS-related diseases. At this point, it should be noted that the majority of the research studies poses a great emphasis on the most potent compounds; however, it is important to recognize that a greater potency does not necessarily mean a higher therapeutic efficacy. Hence, it is essential to be aware that after the synthesis of a new drug candidate, multiple simple in vitro tests and more complex in vivo assays in laboratory animals and also in man are required to provide evidence about the potential therapeutic interest of the compound. In order to achieve central analgesia, opioids based essentially on endogenous peptides that have natural analgesic power as well as opiates incorporating the fentanyl structure were developed. Additionally, new hybrid compounds have been studied to control neuropathic pain and epileptic seizures. In the last case, different and important nucleus included in drugs largely used in the clinic are linked to obtain interesting new anticonvulsant drug candidates. However, in the context of epilepsy, given the scarcity of knowledge regarding the mechanisms of action of the clinically available AEDs and also the gold-standard seizure models used to identify and evaluate anticonvulsant compounds (usually non-mechanistic models), the discovery of AEDs drugs is still not commonly based on specific targets in the 21st century, but rather in their potential to avoid seizures. For this reason, it is necessary to promote a better understanding of the complex mechanisms underlying epileptogenesis in order to progress in epilepsy drug research. The hybrid strategy has also been applied in the Table 1.

Matias et al.

search for new pharmacological alternatives to treat neurodegenerative diseases. In this scope, the development of anti-Alzheimer hybrid molecules are being extensively studied, mainly involving hybrids of tacrine, the first FDA approved cholinesterase inhibitor for the treatment of this disease. In this case, the anti-Alzheimer therapies, which are currently available in the market seem to have a specific target and this could be related to the ineffectiveness of the treatment of this multifactorial disease. Indeed, the multitarget drug design concept continues to be explored in other neurologic and psychiatric diseases, and vilazodone is a prominent example of a hybrid molecule approved by FDA for the treatment of major depressive disorder in adults. To summarize the information referred, accordingly with the CNS disorders discussed, a table was elaborated associating the pharmacophores explored with their putative mechanisms of action (Table 1). In this context, it is important to take into consideration that often the pharmacophores (frequently heterocycle rings) display many biological activities and, for this reason, a wide variety of mechanisms of action, known or unknown, could be involved and interconnected to allow that they possess different pharmacological and therapeutic properties. In conclusion, due to the emergent need of new therapeutic compounds for the treatment of complex CNS diseases, it is clear that the hybrid approach can afford molecules that can be safer and more effective and even more economical than classical drugs. Thus, in the near future, it is expected an increase in the number of potential multi-target drugs developed through this novel medicinal chemistry approach.

The pharmacophores and their main putative mechanisms of action accordingly with the central nervous system disorder in which they were evaluated. The pharmacophores are underlined.

Compound

Pharmacophores (main putative mechanism of action)

Ref.

1

Enkephalin (δ and µ opioid receptors agonism) + deltorphin (δ opioid receptor agonism)

[72]

2

Endomorphin-1 (µ opioid receptor agonism) + substance P (opioid system)

[79]

3

Endomorphine-2 (opioid receptor agonism) + DAMGO (µ opioid receptor agonism)

[80]

4

Fentanyl (µ opioid receptor agonism) + agmatine (imidazoline binding site interaction)

[81]

5a-5d

GABA (GABA agonism) + 1,2,4-triazole (P2X7 antagonism, σ opioid receptor inhibition)

[84]

6

GABA (GABA agonism) + benzothiazole (excitatory neurotransmission modulation)

[108]

7

Phthalimide (voltage-dependent sodium channel blockage) + GABA (GABA agonism) + anilide from ameltolide (voltagedependent sodium channel blockage)

[110, 111]

8

2,6-Dimethylphenyl amino from ameltolide (voltage-dependent sodium channel blockage) + GABA (GABA agonism)

[114]

9a-9b

1,5-Benzodiazepine (GABAA receptor agonism) + thiazolidine (tricarboxylic acid cycle oxidation inhibition)

[118, 263]

10a-10b

Aryl thiosemicarbazide (voltage-dependent sodium channel blockage) + 4-(aryloxy)phenyl semicarbazone (voltagedependent sodium channel blockage)

[123, 264]

11

Semicarbazone (voltage-dependent sodium channel blockage) + isatin (voltage-dependent sodium channel blockage)

[124]

Pain

Epilepsy

Highlights on Molecular Hybrids for CNS Disorders

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

23

(Table 1) Contd….

Compound

Pharmacophores (main putative mechanism of action)

Ref.

12a-12c

Semicarbazone (voltage-dependent sodium channel blockage) + isatin (voltage-dependent sodium channel blockage)

[126]

13

N-(3-methylpyridin-2-yl) semicarbazones (voltage-dependent sodium channel blockage) + pyridine (potassium channel blockage, excitatory neurotransmission modulation)

[127, 265]

14a-14b

Pyrrolidone (GABA mechanism) + pyridine (potassium channel blockage, excitatory neurotransmission modulation)

[129, 266]

15

Quinazolin-4-(3H)-one (GABAA receptor agonism) + 4-(aryloxy)phenyl semicarbazones (voltage-dependent sodium channel blockage)

[133]

16a-16b

Quinazolin-4-(3H)-one (GABAA receptor agonism) + benzothiazole (excitatory neurotransmission modulation)

[136]

17a-17b

Quinazolin-4-(3H)-one (GABAA receptor agonism) + benzothiazole (excitatory neurotransmission modulation)

[137]

18a-18b

Quinazolin-4-(3H)-one (GABAA receptor agonism) + tetrazole (GABA degradation inhibition)

[138, 267]

19

Quinazolin-4-(3H)-one (GABAA receptor agonism) + tetrazole (GABA degradation inhibition) + pyridine (potassium channel blockage, excitatory neurotransmission modulation)

[138]

20a-20b

Phenytoin (voltage-gated sodium channels blockage) + thiosemicarbazide (voltage-dependent sodium channel blockage)

[102]

21

Phenytoin (voltage-gated sodium channels blockage) + 1,3,4-oxadiazole (voltage-gated sodium channels blockage)

[102, 268]

22

Phenytoin (voltage-gated sodium channels blockage) + lidocaine (voltage-gated sodium channels blockage)

[140]

Alzheimer’s disease 23a-23d

Tacrine (AChE inhibition) + methylenedioxybenzene (self-induced Aβ aggregation inhibition)

[155]

24a-24c

Tacrine (AChE inhibition) + oxoisoaporphine (AChE inhibition)

[157]

25

Tacrine (AChE inhibition) + ferulic acid (antioxidant)

[159]

26a-26b

Tacrine (AChE inhibition) + ferulic acid (antioxidant) + nitric oxide (cerebral blood flow regulation)

[160]

27

6-Chlorotacrine (AChE inhibition) + caffeic acid (antioxidant)

[164]

28

6-Chlorotacrine (AChE inhibition) + mefenamic acid (antioxidant)

[168]

29

Tacrine (AChE inhibition) + flurbiprofen (γ-secretase inhibition) + nitric oxide (cerebral blood flow regulation)

[170]

30a

Tacrine (AChE inhibition) + o-amino benzylamine (metal chelation)

[171]

30b

Tacrine (AChE inhibition) + o-hydroxyl benzylamine (metal chelation)

[171]

31a-31b

Tacrine (AChE inhibition) + nimodipine (L-type voltage-dependent calcium channels blockage)

[172]

32, 33a-33-b

Tacrine (AChE inhibition) + phenylthiazole (tau protein aggregation inhibition)

[178]

34

6-Chlorotacrine (AChE inhibition) + carbazole from carvedilol (vasodilatation, antioxidant)

[179]

35

Tacrine (AChE inhibition) + carbazole (antioxidant)

[180]

36a-36b

Tacrine (AChE inhibition) + benzothiazole (Aβ-aggregation inhibition)

[182]

37a-37b

6-Chlorotacrine (AChE inhibition) + 5-phenylpyrano[3,2-c]quinolone (AChE peripheral site inhibition)

[183]

38

Tacrine (AChE inhibition) + 8-hydroxyquinoline (metal chelation, Aβ plaque load reduction)

[184]

39

Tacrine (AChE inhibition) + lophine (H3 receptor antagonism)

[185, 269]

40a

6-Chlorotacrine (AChE inhibition) + rhein (antioxidant)

[186]

40b-40c

Tacrine (AChE inhibition) + rhein (antioxidant)

41a-41d

Tacrine (AChE inhibition) + flavonoids (AChE inhibition, Aβ fibril formation inhibition, antioxidant, metal-chelation)

[189]

42

6-Chlorotacrine (AChE inhibition) + indanone from donepezil (AChE inhibition)

[190]

43

Tacrine (AChE inhibition) + indanone from donepezil (AChE inhibition)

[190]

24

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

Matias et al.

(Table 1) Contd….

Compound

Pharmacophores (main putative mechanism of action)

Ref.

44a

6-Chlorotacrine (AChE inhibition) + donepezil (AChE inhibition)

[191]

44b

Tacrine (AChE inhibition) + donepezil (AChE inhibition)

[191]

45a-45b

Indanone from donepezil (AChE inhibition) + phenyl-N-methylbenzylamino from AP2238 (AChE inhibition)

[193]

46a

Benzylpiperidine from donepezil (AChE inhibition) + indolyl propargylamino from N-[(5-benzyloxy)-1-methyl-1H-indol2-yl)methyl]-N-methylprop-2-yn-1-amine (MAO inhibition)

[195]

46b

Benzylpiperidine from donepezil (AChE inhibition) + indole from compound 46a (AChE-induced Aβ aggregation inhibition)

[196]

47a-47b

Benzylpiperidine from donepezil (AChE inhibition) + 2-chloropyridine-3,5-dicarbonitrile (AChE inhibition)

[198]

48

Dimethoxyindanone from donepezil (AChE inhibition) + hupyridone from huperzine A (AChE inhibition)

[199]

49

Benzylpiperidine from donepezil (AChE inhibition) + N-protecting group (AChE interaction) + lipophilic α-hexyl ester (lipophilicity)

[202]

50a-50b

Berberine (antioxidant) + benzenediol (antioxidant)

[209]

51a-51b

Tacrine (AChE inhibition) + phenyl-benzothiazole (Aβ interaction)

[210, 270]

52

Berberine (antioxidant) + phenyl-benzothiazole (Aβ interaction)

[210]

53

Tacrine (AChE inhibition) + huprine Y (AChE inhibition)

[212]

54

Rhein (antioxidant) + huprine Y (AChE inhibition)

[214]

55, 56

Rosmarinic acid (antioxidant) + choline from acetylcholine (AChE interaction)

[215]

Parkinson’s disease 57

Pramipexole (D3 receptor agonism) + arylpiperazine (D3 receptor agonism)

[231]

58

7-Hydroxy-2-(dipropylamino)tetralin (D3 receptor agonism) + arylpiperazine (D3 receptor agonism)

[231]

59a-59b

Aminotetraline (D3 receptor agonism) + piperazine (D3 receptor agonism)

[232]

General neurodegenerative diseases 60

Caffeic acid (antioxidant) + MDL 28170 (calpain inhibition)

[235]

61, 62

Chroman from vitamin E (antioxidant) + catechol (antioxidant)

[241]

63

Chroman from vitamin E (antioxidant) + catechol (antioxidant) + 1,2,4-oxadiazole (Aβ interaction)

[242]

64

Chroman from vitamin E (antioxidant) + catechol (antioxidant) + isoxazole (Aβ interaction)

[242]

65

Chroman from vitamin E (antioxidant) + catechol (antioxidant) + triazole (Aβ interaction)

[242]

66a-66b

Hydrazine from iproniazid (MAO inhibition) + thiazole from glitazone (MAO-B inhibition)

[247]

5-Cyano-indole-butyl-amine (serotonin reuptake inhibition) + 2-carboxamidebenzofurane-5-yl-piperazine (5-HT1A receptor agonism)

[252, 253]

Depression 67

Cocaine addiction 68

(Bisarylmethoxy)ethyl (dopamine transportation) + 4-aryl-4-piperidinol from bromperidol (dopamine D2/D3 antagonism)

[255]

69

(Bisarylmethoxy)ethyl (dopamine transportation) + pimozide (dopamine antagonism)

[255]

Benzyloxy halogenated benzene from fluoxetine (serotonin reuptake inhibition) + N-(4-methoxy-phenyl)amide (5-HT1B/1D autoreceptors antagonism)

[257]

Autism 70, 71

Highlights on Molecular Hybrids for CNS Disorders

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

25

(Table 1) Contd….

Compound

Pharmacophores (main putative mechanism of action)

Ref.

72

Phenylquinoxaline from sertraline (serotonin reuptake inhibition) + N-(4-methoxy-phenyl)amide and 4-methylpiperazine from GR-127935 (5-HT1B/1D autoreceptors antagonism)

[257]

Neurodegenerative and psychiatric disorders 73

Imidazole (H3 receptor antagonism) + aminoquinoline from amodiaquine (histamine N-methyltransferase inhibition)

LIST OF ABBREVIATIONS 5-HT

= Serotonin

7-OH-DPAT = 7-Hydroxy-2-(dipropylamino)tetralin

POPH–QREN, which is co-funded by FSE and MEC. The authors also acknowledge the support provided by COMPETE program through the strategic project Pest-OE/ SAU/UI0709/2014.

AChE

= Acetylcholinesterase

AD

= Alzheimer’s disease

AED

= Antiepileptic drug



= Amyloid β-protein

BBB

= Blood-brain barrier

BuChE

= Butyrylcholinesterase

CAS

= Catalytic anionic site

CNS

= Central nervous system

DPPH

= 1,1-Diphenyl-2-picrylhydrazyl

ED50

=

FDA

= Food and Drug Administration

GABA

= γ-aminobutyric acid

IC50

= Median inhibitory concentration

MAO

= Monoamine oxidase

MES

= Electroshock seizures model

[9]

NMDA

= N-methyl D-aspartate

[10]

PAMPA

= Parallel artificial membrane permeability assay

PAS

= Peripheral anionic site

PD

= Parkinson’s diseases

ROS

= Reactive oxygen species

scPTZ

= Subcutaneous pentylenetetrazole

TD50

=

Median effective dose

Median toxic dose

REFERENCES   [1] [2]

[3] [4] [5] [6] [7] [8]

[11]

[12]

[13]

CONFLICT OF INTEREST

[14]

The author(s) confirm that this article content has no conflict of interest.

[15]

ACKNOWLEDGEMENTS

[16]

The authors are grateful to Fundação para a Ciência e a Tecnologia (Lisbon, Portugal) for the PhD fellowship of Mariana Matias (SFHR/BD/85279/2012), involving the

[260]

Han G, Sun J, Wang J, Bai Z, Song F, Lei H. Genomics in Neurological Disorders. Genomics, proteomics & bioinformatics 2014. Heemskerk J, Farkas R, Kaufmann P. Neuroscience networking, linking discovery to drugs. Neuropsychopharmacology, official publication of the American College of Neuropsychopharmacology 2012, 37, 287-9. Villoslada P, Moreno B, Melero I, Pablos JL, Martino G, Uccelli A, et al. Immunotherapy for neurological diseases. Clinical immunology 2008, 128, 294-305. Chang PK, Verbich D, McKinney RA. AMPA receptors as drug targets in neurological disease--advantages, caveats, and future outlook. The European journal of neuroscience 2012, 35, 1908-16. Bansal Y, Silakari O. Multifunctional compounds, smart molecules for multifactorial diseases. Eur J Med Chem 2014, 76, 31-42. Morphy R, Rankovic Z. Fragments, network biology and designing multiple ligands. Drug discovery today 2007, 12, 156-60. Korcsmaros T, Szalay MS, Bode C, Kovacs IA, Csermely P. How to design multi-target drugs. Expert opinion on drug discovery 2007, 2, 799-808. Mar JC, Matigian NA, Mackay-Sim A, Mellick GD, Sue CM, Silburn PA, et al. Variance of gene expression identifies altered network constraints in neurological disease. PLoS genetics 2011, 7, e1002207. Horisberger MA, Di Marco S. Interferon-alpha hybrids. Pharmacology & therapeutics 1995, 66, 507-34. Wang Z, Silverman RB. Syntheses and evaluation of fluorinated conformationally restricted analogues of GABA as potential inhibitors of GABA aminotransferase. Bioorganic & medicinal chemistry 2006, 14, 2242-52. Carreiras MC, Mendes E, Perry MJ, Francisco AP, MarcoContelles J. The multifactorial nature of Alzheimer's disease for developing potential therapeutics. Current topics in medicinal chemistry 2013, 13, 1745-70. Singh P, Kaur M, Verma P. Design, synthesis and anticancer activities of hybrids of indole and barbituric acids--identification of highly promising leads. Bioorganic & medicinal chemistry letters 2009, 19, 3054-8. Corson TW, Aberle N, Crews CM. Design and Applications of Bifunctional Small Molecules, Why Two Heads Are Better Than One. ACS chemical biology 2008, 3, 677-92. Meunier B. Hybrid molecules with a dual mode of action, dream or reality? Accounts of chemical research 2008, 41, 69-77. Aminake MN, Mahajan A, Kumar V, Hans R, Wiesner L, Taylor D, et al. Synthesis and evaluation of hybrid drugs for a potential HIV/AIDS-malaria combination therapy. Bioorganic & medicinal chemistry 2012, 20, 5277-89. Christiaans JAM, Timmerman H. Cardiovascular hybrid drugs, Combination of more than one pharmacological property in one single molecule. European Journal of Pharmaceutical Sciences 1996, 4, 1-22.

26 [17]

[18]

[19]

[20]

[21]

[22]

[23]

[24]

[25] [26]

[27]

[28] [29] [30]

[31]

[32]

[33] [34]

[35]

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0 Desai N, Pandya D, Joshi V, Rajpara K, Vaghani H, Satodiya H. Synthesis, characterization and antimicrobial screening of hybrid molecules containing benzimidazole-pyrazole and pyridine nucleus. Med Chem Res 2012, 21, 4463-72. Konter J, Mollmann U, Lehmann J. NO-donors. Part 17, Synthesis and antimicrobial activity of novel ketoconazole-NO-donor hybrid compounds. Bioorganic & medicinal chemistry 2008, 16, 8294300. Samala G, Devi PB, Nallangi R, Sridevi JP, Saxena S, Yogeeswari P, et al. Development of novel tetrahydrothieno[2,3-c]pyridine-3carboxamide based Mycobacterium tuberculosis pantothenate synthetase inhibitors, molecular hybridization from known antimycobacterial leads. Bioorganic & medicinal chemistry 2014, 22, 1938-47. Addla D, Jallapally A, Gurram D, Yogeeswari P, Sriram D, Kantevari S. Rational design, synthesis and antitubercular evaluation of novel 2-(trifluoromethyl)phenothiazine-[1,2,3]triazole hybrids. Bioorganic & medicinal chemistry letters 2014, 24, 233-6. Kumar L, Lal N, Kumar V, Sarswat A, Jangir S, Bala V, et al. Azole-carbodithioate hybrids as vaginal anti-Candida contraceptive agents, design, synthesis and docking studies. Eur J Med Chem 2013, 70, 68-77. Kumar A, Srivastava K, Kumar SR, Puri SK, Chauhan PM. Synthesis of new 4-aminoquinolines and quinoline-acridine hybrids as antimalarial agents. Bioorganic & medicinal chemistry letters 2010, 20, 7059-63. Sashidhara KV, Avula SR, Palnati GR, Singh SV, Srivastava K, Puri SK, et al. Synthesis and in vitro evaluation of new chloroquine-chalcone hybrids against chloroquine-resistant strain of Plasmodium falciparum. Bioorganic & medicinal chemistry letters 2012, 22, 5455-9. Singh K, Kaur H, Smith P, de Kock C, Chibale K, Balzarini J. Quinoline-pyrimidine hybrids, synthesis, antiplasmodial activity, SAR, and mode of action studies. Journal of medicinal chemistry 2014, 57, 435-48. Biot C, Nosten F, Fraisse L, Ter-Minassian D, Khalife J, Dive D. The antimalarial ferroquine, from bench to clinic. Parasite 2011, 18, 207-14. Gupta A, Saha P, Descoteaux C, Leblanc V, Asselin E, Berube G. Design, synthesis and biological evaluation of estradiolchlorambucil hybrids as anticancer agents. Bioorganic & medicinal chemistry letters 2010, 20, 1614-8. Mallavadhani UV, Vanga NR, Jeengar MK, Naidu VG. Synthesis of novel ring-A fused hybrids of oleanolic acid with capabilities to arrest cell cycle and induce apoptosis in breast cancer cells. Eur J Med Chem 2014, 74, 398-404. Pendekal MS, Tegginamat PK. Hybrid drug delivery system for oropharyngeal, cervical and colorectal cancer - in vitro and in vivo evaluation. Saudi Pharm J 2013, 21, 177-86. Shen L, Yang X, Yang B, He Q, Hu Y. Novel hybrids from lamellarin D and combretastatin A 4 as cytotoxic agents. Eur J Med Chem 2010, 45, 11-8. Das BC, Mahalingam SM, Panda L, Wang B, Campbell P, Evans T. Design and Synthesis of Potential New Apoptosis Agents, Hybrid Compounds Containing Perillyl Alcohol and New Constrained Retinoids. Tetrahedron letters 2010, 51, 1462-6. Kumar R, Gupta L, Pal P, Khan S, Singh N, Katiyar SB, et al. Synthesis and cytotoxicity evaluation of (tetrahydro-betacarboline)-1,3,5-triazine hybrids as anticancer agents. Eur J Med Chem 2010, 45, 2265-76. Bose DS, Idrees M, Todewale IK, Jakka NM, Rao JV. Hybrids of privileged structures benzothiazoles and pyrrolo[2,1c][1,4]benzodiazepin-5-one, and diversity-oriented synthesis of benzothiazoles. Eur J Med Chem 2012, 50, 27-38. Dhorajiya BD, Ibrahim AS, Badria FA, Dholakiya BZ. Design and synthesis of novel nucleobase-based barbiturate derivatives as potential anticancer agents. Med Chem Res 2014, 23, 839-47. Adsule S, Banerjee S, Ahmed F, Padhye S, Sarkar FH. Hybrid anticancer agents, isothiocyanate-progesterone conjugates as chemotherapeutic agents and insights into their cytotoxicities. Bioorganic & medicinal chemistry letters 2010, 20, 1247-51. Belluti F, Fontana G, Dal Bo L, Carenini N, Giommarelli C, Zunino F. Design, synthesis and anticancer activities of stilbenecoumarin hybrid compounds, Identification of novel proapoptotic agents. Bioorganic & medicinal chemistry 2010, 18, 3543-50.

Matias et al. [36]

[37]

[38]

[39]

[40]

[41]

[42] [43]

[44]

[45]

[46]

[47]

[48]

[49]

[50]

[51]

[52]

Solomon VR, Hu C, Lee H. Design and synthesis of anti-breast cancer agents from 4-piperazinylquinoline, a hybrid pharmacophore approach. Bioorganic & medicinal chemistry 2010, 18, 1563-72. Garcia G, Serrano I, Sanchez-Alonso P, Rodriguez-Puyol M, Alajarin R, Griera M, et al. New losartan-hydrocaffeic acid hybrids as antihypertensive-antioxidant dual drugs, Ester, amide and amine linkers. Eur J Med Chem 2012, 50, 90-101. Calderone V, Rapposelli S, Martelli A, Digiacomo M, Testai L, Torri S, et al. NO-glibenclamide derivatives, prototypes of a new class of nitric oxide-releasing anti-diabetic drugs. Bioorganic & medicinal chemistry 2009, 17, 5426-32. Zhang L, Chen X, Liu J, Zhu Q, Leng Y, Luo X, et al. Discovery of novel dual-action antidiabetic agents that inhibit glycogen phosphorylase and activate glucokinase. Eur J Med Chem 2012, 58, 624-39. Hidalgo-Figueroa S, Ramirez-Espinosa JJ, Estrada-Soto S, Almanza-Perez JC, Roman-Ramos R, Alarcon-Aguilar FJ, et al. Discovery of thiazolidine-2,4-dione/biphenylcarbonitrile hybrid as dual PPAR alpha/gamma modulator with antidiabetic effect, in vitro, in silico and in vivo approaches. Chemical biology & drug design 2013, 81, 474-83. Mills AD, Yoo C, Butler JD, Yang B, Verkman AS, Kurth MJ. Design and synthesis of a hybrid potentiator-corrector agonist of the cystic fibrosis mutant protein DeltaF508-CFTR. Bioorganic & medicinal chemistry letters 2010, 20, 87-91. Bubenyak M, Noszal B, Koczian K, Takacs M, Beni S, Hermecz I, et al. Bioisosteric hybrids of two anti-inflammatory agents, rutaecarpine and piroxicam. Tetrahedron letters 2008, 49, 5711-3. Detsi A, Bouloumbasi D, Prousis KC, Koufaki M, Athanasellis G, Melagraki G, et al. Design and synthesis of novel quinolinone-3aminoamides and their alpha-lipoic acid adducts as antioxidant and anti-inflammatory agents. Journal of medicinal chemistry 2007, 50, 2450-8. Almasirad A, Mousavi Z, Tajik M, Assarzadeh MJ, Shafiee A. Synthesis, analgesic and anti-inflammatory activities of new methyl-imidazolyl-1,3,4-oxadiazoles and 1,2,4-triazoles. Daru , journal of Faculty of Pharmacy, Tehran University of Medical Sciences 2014, 22, 22. Capim SL, Goncalves GM, Dos Santos GC, Marinho BG, Vasconcellos ML. High analgesic and anti-inflammatory in vivo activities of six new hybrids NSAIAs tetrahydropyran derivatives. Bioorganic & medicinal chemistry 2013, 21, 6003-10. Liu A, Huang L, Wang Z, Luo Z, Mao F, Shan W, et al. Hybrids consisting of the pharmacophores of salmeterol and roflumilast or phthalazinone, dual beta(2)-adrenoceptor agonists-PDE4 inhibitors for the treatment of COPD. Bioorganic & medicinal chemistry letters 2013, 23, 1548-52. Zeng ZS, He QQ, Liang YH, Feng XQ, Chen FE, De Clercq E, et al. Hybrid diarylbenzopyrimidine non-nucleoside reverse transcriptase inhibitors as promising new leads for improved antiHIV-1 chemotherapy. Bioorganic & medicinal chemistry 2010, 18, 5039-47. Theoduloz C, Bravo I, Pertino MW, Schmeda-Hirschmann G. Diterpenylquinone hybrids, synthesis and assessment of gastroprotective mechanisms of action in human cells. Molecules 2013, 18, 11044-66. Kinfe HH, Belay YH, Joseph JS, Mukwevho E. Evaluation of the Influence of thiosemicarbazone-triazole hybrids on genes implicated in lipid oxidation and accumulation as potential antiobesity agents. Bioorganic & medicinal chemistry letters 2013, 23, 5275-8. Sashidhara KV, Modukuri RK, Choudhary D, Bhaskara Rao K, Kumar M, Khedgikar V, et al. Synthesis and evaluation of new coumarin-pyridine hybrids with promising anti-osteoporotic activities. Eur J Med Chem 2013, 70, 802-10. Kim S, Kim Y, Kong Y, Kim H, Kang J. Synthesis and in vitro biological activity of retinyl polyhydroxybenzoates, novel hybrid retinoid derivatives. Bioorganic & medicinal chemistry letters 2009, 19, 508-12. Abuo-Rahma Gel D, Abdel-Aziz M, Beshr EA, Ali TF. 1,2,4Triazole/oxime hybrids as new strategy for nitric oxide donors, Synthesis, anti-inflammatory, ulceroginicity and antiproliferative activities. Eur J Med Chem 2014, 71, 185-98.

Highlights on Molecular Hybrids for CNS Disorders [53]

[54]

[55]

[56]

[57] [58] [59]

[60] [61]

[62]

[63] [64] [65]

[66] [67] [68] [69] [70]

[71] [72]

[73] [74]

Lombard MC, N'Da DD, Breytenbach JC, Kolesnikova NI, Tran Van Ba C, Wein S, et al. Antimalarial and anticancer activities of artemisinin-quinoline hybrid-dimers and pharmacokinetic properties in mice. European journal of pharmaceutical sciences , official journal of the European Federation for Pharmaceutical Sciences 2012, 47, 834-41. Jones M, Mercer AE, Stocks PA, La Pensee LJ, Cosstick R, Park BK, et al. Antitumour and antimalarial activity of artemisininacridine hybrids. Bioorganic & medicinal chemistry letters 2009, 19, 2033-7. Sangani CB, Makawana JA, Zhang X, Teraiya SB, Lin L, Zhu HL. Design, synthesis and molecular modeling of pyrazole-quinolinepyridine hybrids as a new class of antimicrobial and anticancer agents. Eur J Med Chem 2014, 76, 549-57. Reiter C, Capci Karagoz A, Frohlich T, Klein V, Zeino M, Viertel K, et al. Synthesis and study of cytotoxic activity of 1,2,4-trioxaneand egonol-derived hybrid molecules against Plasmodium falciparum and multidrug-resistant human leukemia cells. Eur J Med Chem 2014, 75, 403-12. Ripamonti CI. Pain management. Annals of oncology , official journal of the European Society for Medical Oncology / ESMO 2012, 23 Suppl 10, x294-301. Millan MJ. Descending control of pain. Progress in neurobiology 2002, 66, 355-474. Phyomaung PP, Dubowitz J, Cicuttini FM, Fernando S, Wluka AE, Raaijmaakers P, et al. Are depression, anxiety and poor mental health risk factors for knee pain? A systematic review. BMC musculoskeletal disorders 2014, 15. Shah S, Mehta V. Controversies and advances in non-steroidal antiinflammatory drug (NSAID) analgesia in chronic pain management. Postgraduate medical journal 2012, 88, 73-8. Castori M, Morlino S, Celletti C, Ghibellini G, Bruschini M, Grammatico P, et al. Re-writing the natural history of pain and related symptoms in the joint hypermobility syndrome/EhlersDanlos syndrome, hypermobility type. American journal of medical genetics Part A 2013, 161A, 2989-3004. Bozimowski G. Patient perceptions of pain management therapy, a comparison of real-time assessment of patient education and satisfaction and registered nurse perceptions. Pain management nursing , official journal of the American Society of Pain Management Nurses 2012, 13, 186-93. Ketwaroo GA, Cheng V, Lembo A. Opioid-induced bowel dysfunction. Current gastroenterology reports 2013, 15, 344. Law PY, Reggio PH, Loh HH. Opioid receptors, toward separation of analgesic from undesirable effects. Trends Biochem Sci 2013, 38, 275-82. Mellbye A, Svendsen K, Borchgrevink PC, Skurtveit S, Fredheim OM. Concomitant medication among persistent opioid users with chronic non-malignant pain. Acta anaesthesiologica Scandinavica 2012, 56, 1267-76. Portenoy RK. Current pharmacotherapy of chronic pain. Journal of pain and symptom management 2000, 19, S16-20. Jongen JLM, Hans G, Benzon HT, Huygen F, Hartrick CT. Neuropathic Pain and Pharmacological Treatment. Pain Practice 2014, 14, 283-95. Dworkin RH, O'Connor AB, Kent J, Mackey SC, Raja SN, Stacey BR, et al. Interventional management of neuropathic pain, NeuPSIG recommendations. Pain 2013, 154, 2249-61. Kalliomaki J, Miller F, Kagedal M, Karlsten R. Early phase drug development for treatment of chronic pain--options for clinical trial and program design. Contemporary clinical trials 2012, 33, 689-99. Sobczak M, Salaga M, Storr MA, Fichna J. Physiology, signaling, and pharmacology of opioid receptors and their ligands in the gastrointestinal tract, current concepts and future perspectives. J Gastroenterol 2014, 49, 24-45. Bodnar RJ. Endogenous opiates and behavior, 2012. Peptides 2013, 50, 55-95. Ciszewska M, Ruszczynska K, Oleszczuk M, Chung NN, Witkowska E, Schiller PW, et al. Cyclic enkephalin-deltorphin hybrids containing a carbonyl bridge, structure and opioid activity. Acta biochimica Polonica 2011, 58, 225-30. Tomboly C, Peter A, Toth G. In vitro quantitative study of the degradation of endomorphins. Peptides 2002, 23, 1573-80. Mollica A, Pinnen F, Stefanucci A, Feliciani F, Campestre C, Mannina L, et al. The cis-4-amino-L-proline residue as a scaffold

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

[75] [76]

[77]

[78]

[79]

[80] [81]

[82] [83]

[84]

[85] [86] [87]

[88] [89]

[90]

[91] [92]

27

for the synthesis of cyclic and linear endomorphin-2 analogues. Journal of medicinal chemistry 2012, 55, 3027-35. Gredicak M, Supek F, Kralj M, Majer Z, Hollosi M, Smuc T, et al. Computational structure-activity study directs synthesis of novel antitumor enkephalin analogs. Amino acids 2010, 38, 1185-91. Trzeciak-Karlikowska K, Bujacz A, Ciesielski W, Bujacz GD, Potrzebowski MJ. The influence of the stereochemistry of alanine residue on the solid state conformation and crystal packing of opioid peptides containing D-Ala or L-Ala in message domain-XRD and NMR study. The journal of physical chemistry B 2011, 115, 9910-9. Lesma G, Salvadori S, Airaghi F, Bojnik E, Borsodi A, Recca T, et al. Synthesis, pharmacological evaluation and conformational investigation of endomorphin-2 hybrid analogues. Molecular diversity 2013, 17, 19-31. Varamini P, Mansfeld FM, Blanchfield JT, Wyse BD, Smith MT, Toth I. Synthesis and Biological Evaluation of an Orally Active Glycosylated Endomorphin-1. Journal of medicinal chemistry 2012, 55, 5859-67. Varamini P, Hussein WM, Mansfeld FM, Toth I. Synthesis, biological activity and structure-activity relationship of endomorphin-1/substance P derivatives. Bioorganic & medicinal chemistry 2012, 20, 6335-43. Mollica A, Costante R, Stefanucci A, Pinnen F, Luisi G, Pieretti S, et al. Hybrid peptides endomorphin-2/DAMGO, design, synthesis and biological evaluation. Eur J Med Chem 2013, 68, 167-77. Dardonville C, Fernandez-Fernandez C, Gibbons SL, Ryan GJ, Jagerovic N, Gabilondo AM, et al. Synthesis and pharmacological studies of new hybrid derivatives of fentanyl active at the muopioid receptor and I2-imidazoline binding sites. Bioorganic & medicinal chemistry 2006, 14, 6570-80. Vardanyan RS, Hruby VJ. Fentanyl-related compounds and derivatives, current status and future prospects for pharmaceutical applications. Future Med Chem 2014, 6, 385-412. Kotagale NR, Shirbhate SH, Shukla P, Ugale RR. Agmatine attenuates neuropathic pain in sciatic nerve ligated rats, Modulation by hippocampal sigma receptors. European journal of pharmacology 2013, 714, 424-31. Yogeeswari P, Patel SK, Reddy IV, Semwal A, Sharma M, Gangadhar M, et al. GABA derivatives for the treatment of epilepsy and neuropathic pain, A synthetic integration of GABA in 1, 2, 4–Triazolo–2H–one nucleus. Biomedicine & Aging Pathology 2012, 2, 31-40. Ebrahimi H, Shafa M, Hakimzadeh Asl S. Prevalence of active epilepsy in Kerman, Iran, a house based survey. Acta neurologica Taiwanica 2012, 21, 115-24. Dulac O, Plecko B, Gataullina S, Wolf NI. Occasional seizures, epilepsy, and inborn errors of metabolism. Lancet neurology 2014, 13, 727-39. Bidwell J, Khuwatsamrit T, Askew B, Ehrenberg JA, Helmers S. Seizure reporting technologies for epilepsy treatment, A review of clinical information needs and supporting technologies. Seizure 2015, 32, 109-17. Sridharan R. Epidemiology of epilepsy. Curr Sci India 2002, 82, 664-70. Azam F, El-Gnidi BA, Alkskas IA. Combating oxidative stress in epilepsy, design, synthesis, quantum chemical studies and anticonvulsant evaluation of 1-(substituted benzylidene/ ethylidene)-4-(naphthalen-1-yl)semicarbazides. Eur J Med Chem 2010, 45, 2817-26. Rowley NM, Madsen KK, Schousboe A, Steve White H. Glutamate and GABA synthesis, release, transport and metabolism as targets for seizure control. Neurochemistry international 2012, 61, 546-58. Hawker DD, Silverman RB. Synthesis and evaluation of novel heteroaromatic substrates of GABA aminotransferase. Bioorganic & medicinal chemistry 2012, 20, 5763-73. Syvanen S, Labots M, Tagawa Y, Eriksson J, Windhorst AD, Lammertsma AA, et al. Altered GABAA receptor density and unaltered blood-brain barrier transport in a kainate model of epilepsy, an in vivo study using 11C-flumazenil and PET. Journal of nuclear medicine , official publication, Society of Nuclear Medicine 2012, 53, 1974-83.

28 [93] [94] [95] [96]

[97] [98] [99] [100] [101]

[102] [103] [104] [105] [106]

[107]

[108] [109] [110]

[111]

[112]

[113]

[114]

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0 Hines RM, Davies PA, Moss SJ, Maguire J. Functional regulation of GABAA receptors in nervous system pathologies. Current opinion in neurobiology 2012, 22, 552-8. Krasowski MD, McMillin GA. Advances in anti-epileptic drug testing. Clinica chimica acta, international journal of clinical chemistry 2014, 436, 224-36. Miziak B, Chroscinska-Krawczyk M, Blaszczyk B, Radzik I, Czuczwar SJ. Novel approaches to anticonvulsant drug discovery. Expert opinion on drug discovery 2013, 8, 1415-27. Lambert DM, Scriba GKE, Poupaert JH, Dumont P. Anticonvulsant activity of ester- and amide-type lipid conjugates of glycine and N-benzyloxycarbonylglycine. European Journal of Pharmaceutical Sciences 1996, 4, 159-66. Sussan S, Dagan A, Blotnik S, Bialer M. The structural requirements for the design of antiepileptic-glycine derivatives. Epilepsy research 1999, 34, 207-20. Yogeeswari P, Ragavendran JV, Thirumurugan R, Saxena A, Sriram D. Ion channels as important targets for antiepileptic drug design. Current drug targets 2004, 5, 589-602. Lipkind GM, Fozzard HA. Molecular model of anticonvulsant drug binding to the voltage-gated sodium channel inner pore. Molecular pharmacology 2010, 78, 631-8. Abdelsayed M, Sokolov S. Voltage-gated sodium channels Pharmaceutical targets via anticonvulsants to treat epileptic syndromes. Channels 2013, 7, 146-52. Lenkowski PW, Batts TW, Smith MD, Ko SH, Jones PJ, Taylor CH, et al. A pharmacophore derived phenytoin analogue with increased affinity for slow inactivated sodium channels exhibits a desired anticonvulsant profile. Neuropharmacology 2007, 52, 1044-54. Botros S, Khalil NA, Naguib BH, El-Dash Y. Synthesis and anticonvulsant activity of new phenytoin derivatives. Eur J Med Chem 2013, 60, 57-63. Johannessen SI, Landmark CJ. Antiepileptic drug interactions principles and clinical implications. Current neuropharmacology 2010, 8, 254-67. Kerr MP. The impact of epilepsy on patients' lives. Acta neurologica Scandinavica 2012, 126, 1-9. Banach M, Czuczwar SJ, Borowicz KK. Statins - Are they anticonvulsant? Pharmacological Reports 2014, 66, 521-8. Obniska J, Chlebek I, Kaminski K, Bojarski AJ, Satala G. Synthesis, anticonvulsant activity and 5-HT1A/5-HT7 receptors affinity of 1-[(4-arylpiperazin-1-yl)-propyl]-succinimides. Pharmacological reports , PR 2012, 64, 326-35. Jain SV, Bhadoriya KS, Bari SB, Sahu NK, Ghate M. Discovery of potent anticonvulsant ligands as dual NMDA and AMPA receptors antagonists by molecular modelling studies. Med Chem Res 2012, 21, 3465-84. Hassan MZ, Khan SA, Amir M. Design, synthesis and evaluation of N-(substituted benzothiazol-2-yl)amides as anticonvulsant and neuroprotective. Eur J Med Chem 2012, 58, 206-13. Wilson JR, Fehlings MG. Riluzole for Acute Traumatic Spinal Cord Injury, A Promising Neuroprotective Treatment Strategy. World Neurosurg 2014, 81, 825-9. Vamecq J, Bac P, Herrenknecht C, Maurois P, Delcourt P, Stables JP. Synthesis and anticonvulsant and neurotoxic properties of substituted N-phenyl derivatives of the phthalimide pharmacophore. Journal of medicinal chemistry 2000, 43, 1311-9. Ragavendran JV, Sriram D, Patel SK, Reddy IV, Bharathwajan N, Stables J, et al. Design and synthesis of anticonvulsants from a combined phthalimide-GABA-anilide and hydrazone pharmacophore. Eur J Med Chem 2007, 42, 146-51. Philip AE, Poupaert JH, Chevé G, Muccioli G, Lambert D, McCurdy CR. Structure–activity relationship of phenytoinergic antiepileptic drugs related to ameltolide. Med Chem Res 2007, 16, 130-5. Vamecq J, Lambert D, Poupaert JH, Masereel B, Stables JP. Anticonvulsant activity and interactions with neuronal voltagedependent sodium channel of analogues of ameltolide. Journal of medicinal chemistry 1998, 41, 3307-13. Yogeeswari P, Sriram D, Sahitya P, Ragavendran JV, Ranganadh V. Synthesis and anticonvulsant activity of 4-(2-(2,6dimethylphenylamino)-2-oxoethylamino)-N(substituted)butanamides, a pharmacophoric hybrid approach. Bioorganic & medicinal chemistry letters 2007, 17, 3712-5.

Matias et al. [115] [116] [117] [118]

[119]

[120]

[121]

[122]

[123]

[124]

[125] [126] [127]

[128]

[129]

[130] [131] [132] [133]

[134]

Szatkowska P, Koba M, Koslinski P, Wandas J, Baczek T. Analytical methods for determination of benzodiazepines. A short review. Cent Eur J Chem 2014, 12, 994-1007. Deeb TZ, Maguire J, Moss SJ. Possible alterations in GABAA receptor signaling that underlie benzodiazepine-resistant seizures. Epilepsia 2012, 53 Suppl 9, 79-88. Rundfeldt C, Loscher W. The Pharmacology of Imepitoin, The First Partial Benzodiazepine Receptor Agonist Developed for the Treatment of Epilepsy. Cns Drugs 2014, 28, 29-43. Ghogare JG, Bhandari SV, Bothara KG, Madgulkar AR, Parashar GA, Sonawane BG, et al. Design, synthesis and pharmacological screening of potential anticonvulsant agents using hybrid approach. Eur J Med Chem 2010, 45, 857-63. Kulandasamy R, Adhikari AV, Taranalli A, Venkataswamy T. New Hydrazides and Thiosemicarbazides Derived from Ethylenedioxythiophene as Potential Anticonvulsants. Phosphorus Sulfur 2010, 185, 1358-68. Karki SS, Bahaduria VS, Rana V, Kumar S, Subbaro PG, Das U, et al. 1-Arylmethyl-2,3-dioxo-2,3-dihydroindole thiosemicarbazones as leads for developing cytotoxins and anticonvulsants. Journal of enzyme inhibition and medicinal chemistry 2009, 24, 537-44. Kshirsagar A, Toraskar MP, Kulkarni VM, Dhanashire S, Kadam V. Microwave assisted synthesis of potential antiinfective and anticonvulsant thiosemicarbazones. Int J Chem Tech Res 2009, 1, 696-701. Mozaffari S, Ghasemi S, Baher H, Khademi H, Amini M, Sakhteman A, et al. Synthesis and evaluation of some novel methylene-bridged aryl semicarbazones as potential anticonvulsant agents. Med Chem Res 2012, 21, 3797-808. Tripathi L, Kumar P, Singh R, Stables JP. Design, synthesis and anticonvulsant evaluation of novel N-(4-substituted phenyl)-2-[4(substituted) benzylidene]-hydrazinecarbothio amides. Eur J Med Chem 2012, 47, 153-66. Pandeya SN, Raja AS, Stables JR. Synthesis of isatin semicarbazones as novel anticonvulsants - role of hydrogen bonding. Journal of Pharmacy and Pharmaceutical Sciences 2002, 5, 275-80. Xie C, Tang LM, Li FN, Guan LP, Pan CY, Wang SH. Structurebased design, synthesis, and anticonvulsant activity of isatin-1-Nphenylacetamide derivatives. Med Chem Res 2014, 23, 2161-8. Kumar D, Sharma VK, Kumar R, Singh T, Singh H, Singh AD, et al. Design, Synthesis and Anticonvulsant Activity of Some New 5,7-Dibromoisatin Semicarbazone Derivatives. Excli J 2013, 12. Mehta S, Pavana RK, Yogeeswari P, Sriram D, Stables J. Heteroaryl-substuted semicarbazones, synthesis and anticonvulsant activity of N-(3-methylpyridin-2-yl)-substituted semicarbazones. J Heterocyclic Chem 2006, 43, 1287-93. Subudhi BB, Panda PK, Swain SP, Sarangi P. Synthesis, Characterization and Anticonvulsant Activity Evaluation of Some 1,4-Dihydropyridines and 3,5-(Substituted)Oxycarbonyl-1,4Dihydro-2,6-Dimethyl-N-[2-(4-Sulfamoylphenylamino)-Acetyl]-4(Substituted)Pyridines. Acta poloniae pharmaceutica 2009, 66, 147-53. Siddiqui N, Ahsan W, Alam MS, Ali R, Srivastava K. Design, synthesis and evaluation of anticonvulsant activity of pyridinylpyrrolidones, a pharmacophore hybrid approach. Archiv der Pharmazie 2012, 345, 185-94. El-Azab AS, Eltahir KE. Synthesis and anticonvulsant evaluation of some new 2,3,8-trisubstituted-4(3H)-quinazoline derivatives. Bioorganic & medicinal chemistry letters 2012, 22, 327-33. Amir M, Ali I, Hassan MZ. Design and synthesis of some new quinazolin-4-(3H)-ones as anticonvulsant and antidepressant agents. Archives of pharmacal research 2013, 36, 61-8. Alagarsamy V, Saravanan G. Synthesis and anticonvulsant activity of novel quinazolin-4(3H)-one derived pyrazole analogs. Med Chem Res 2013, 22, 1711-22. Kumar P, Shrivastava B, Pandeya SN, Stables JP. Design, synthesis and potential 6 Hz psychomotor seizure test activity of some novel 2-(substituted)-3-{[substituted]amino}quinazolin-4(3H)-one. Eur J Med Chem 2011, 46, 1006-18. Pandeya SN, Yogeeswari P, Stables JP. Synthesis and anticonvulsant activity of 4-bromophenyl substituted aryl semicarbazones. European Journal of Medicinal Chemistry 2000, 35, 879-86.

Highlights on Molecular Hybrids for CNS Disorders [135]

[136] [137]

[138] [139]

[140]

[141]

[142]

[143]

[144] [145] [146] [147]

[148] [149] [150]

[151]

[152]

[153]

[154]

[155]

Yogeeswari P, Sriram D, Veena V, Kavya R, Rakhra K, Ragavendran JV, et al. Synthesis of aryl semicarbazones as potential anticonvulsant agents. Biomedicine & Pharmacotherapy 2005, 59, 51-5. Ugale VG, Patel HM, Wadodkar SG, Bari SB, Shirkhedkar AA, Surana SJ. Quinazolino-benzothiazoles, fused pharmacophores as anticonvulsant agents. Eur J Med Chem 2012, 53, 107-13. Malik S, Bahare RS, Khan SA. Design, synthesis and anticonvulsant evaluation of N-(benzo[d]thiazol-2-ylcarbamoyl)-2methyl-4-oxoquinazoline-3(4H)-carbothioamide derivatives, a hybrid pharmacophore approach. Eur J Med Chem 2013, 67, 1-13. Malik S, Khan SA. Design and evaluation of new hybrid pharmacophore quinazolino-tetrazoles as anticonvulsant strategy. Med Chem Res 2014, 23, 207-23. Trisovic N, Timic T, Divljakovic J, Rogan J, Poleti D, Savic MM, et al. Synthesis, structural and biological characterization of 5phenylhydantoin derivatives as potential anticonvulsant agents. Monatsh Chem 2012, 143, 1451-7. Wang Y, Jones PJ, Batts TW, Landry V, Patel MK, Brown ML. Ligand-based design and synthesis of novel sodium channel blockers from a combined phenytoin-lidocaine pharmacophore. Bioorganic & medicinal chemistry 2009, 17, 7064-72. Eijkelkamp N, Linley JE, Baker MD, Minett MS, Cregg R, Werdehausen R, et al. Neurological perspectives on voltage-gated sodium channels. Brain , a journal of neurology 2012, 135, 2585612. Cacciatore I, Baldassarre L, Fornasari E, Mollica A, Pinnen F. Recent advances in the treatment of neurodegenerative diseases based on GSH delivery systems. Oxidative medicine and cellular longevity 2012, 2012, 240146. de Oliveira DM, Ferreira Lima RM, El-Bacha RS. Brain rust, recent discoveries on the role of oxidative stress in neurodegenerative diseases. Nutritional neuroscience 2012, 15, 94102. Tan L, Yu JT, Tan L. The kynurenine pathway in neurodegenerative diseases, mechanistic and therapeutic considerations. Journal of the neurological sciences 2012, 323, 1-8. Zolezzi JM, Inestrosa NC. Brain metabolite clearance, impact on Alzheimer’s disease. Metabolic brain disease 2014, 1-9. Silva T, Reis J, Teixeira J, Borges F. Alzheimer's disease, enzyme targets and drug discovery struggles, From natural products to drug prototypes. Ageing Res Rev 2014, 15, 116-45. Swomley AM, Forster S, Keeney JT, Triplett J, Zhang Z, Sultana R, et al. Abeta, oxidative stress in Alzheimer disease, Evidence based on proteomics studies. Bba-Mol Basis Dis 2014, 1842, 124857. Walsh R, Rockwood K, Martin E, Darvesh S. Synergistic inhibition of butyrylcholinesterase by galantamine and citalopram. Bba-Gen Subjects 2011, 1810, 1230-5. Doraiswamy PM. Non-cholinergic strategies for treating and preventing Alzheimer's disease. Cns Drugs 2002, 16, 811-24. Zemek F, Drtinova L, Nepovimova E, Sepsova V, Korabecny J, Klimes J, et al. Outcomes of Alzheimer's disease therapy with acetylcholinesterase inhibitors and memantine. Expert opinion on drug safety 2014, 13, 759-74. Romero A, Cacabelos R, Oset-Gasque MJ, Samadi A, MarcoContelles J. Novel tacrine-related drugs as potential candidates for the treatment of Alzheimer's disease. Bioorganic & medicinal chemistry letters 2013, 23, 1916-22. de Aquino RA, Modolo LV, Alves RB, de Fatima A. Synthesis, kinetic studies and molecular modeling of novel tacrine dimers as cholinesterase inhibitors. Organic & biomolecular chemistry 2013, 11, 8395-409. Rizzo S, Bisi A, Bartolini M, Mancini F, Belluti F, Gobbi S, et al. Multi-target strategy to address Alzheimer's disease, design, synthesis and biological evaluation of new tacrine-based dimers. Eur J Med Chem 2011, 46, 4336-43. Di Pietro O, Perez-Areales FJ, Juarez-Jimenez J, Espargaro A, Clos MV, Perez B, et al. Tetrahydrobenzo[h][1,6]naphthyridine-6chlorotacrine hybrids as a new family of anti-Alzheimer agents targeting beta-amyloid, tau, and cholinesterase pathologies. Eur J Med Chem 2014, 84, 107-17. Luo W, Li YP, He Y, Huang SL, Tan JH, Ou TM, et al. Design, synthesis and evaluation of novel tacrine-multialkoxybenzene

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

[156]

[157]

[158] [159]

[160]

[161]

[162]

[163]

[164]

[165] [166]

[167]

[168] [169] [170]

[171]

[172]

[173]

29

hybrids as dual inhibitors for cholinesterases and amyloid beta aggregation. Bioorganic & medicinal chemistry 2011, 19, 763-70. Tang H, Wei YB, Zhang C, Ning FX, Qiao W, Huang SL, et al. Synthesis, biological evaluation and molecular modeling of oxoisoaporphine and oxoaporphine derivatives as new dual inhibitors of acetylcholinesterase/butyrylcholinesterase. Eur J Med Chem 2009, 44, 2523-32. Tang H, Zhao LZ, Zhao HT, Huang SL, Zhong SM, Qin JK, et al. Hybrids of oxoisoaporphine-tacrine congeners, novel acetylcholinesterase and acetylcholinesterase-induced beta-amyloid aggregation inhibitors. Eur J Med Chem 2011, 46, 4970-9. Nabi G, Liu ZQ. Ferulic and Coumaric Acids, Application to Release Oxidative Stress of DNA and Methyl Linoleate. J Food Biochem 2012, 36, 38-45. Fang L, Kraus B, Lehmann J, Heilmann J, Zhang Y, Decker M. Design and synthesis of tacrine-ferulic acid hybrids as multi-potent anti-Alzheimer drug candidates. Bioorganic & medicinal chemistry letters 2008, 18, 2905-9. Chen Y, Sun J, Fang L, Liu M, Peng S, Liao H, et al. Tacrineferulic acid-nitric oxide (NO) donor trihybrids as potent, multifunctional acetyl- and butyrylcholinesterase inhibitors. Journal of medicinal chemistry 2012, 55, 4309-21. Huang Y, Jin M, Pi R, Zhang J, Chen M, Ouyang Y, et al. Protective effects of caffeic acid and caffeic acid phenethyl ester against acrolein-induced neurotoxicity in HT22 mouse hippocampal cells. Neuroscience letters 2013, 535, 146-51. Oboh G, Agunloye OM, Akinyemi AJ, Ademiluyi AO, Adefegha SA. Comparative study on the inhibitory effect of caffeic and chlorogenic acids on key enzymes linked to Alzheimer's disease and some pro-oxidant induced oxidative stress in rats' brain-in vitro. Neurochemical research 2013, 38, 413-9. Virues C, Dominguez Z, Salas M, Navarro RE, Velazquez EF, Cruz S, et al. Host-guest complexation of antioxidative caffeic and ferulic acid amides with a functionalized cyclophane. J Incl Phenom Macro 2012, 74, 407-13. Chao X, He X, Yang Y, Zhou X, Jin M, Liu S, et al. Design, synthesis and pharmacological evaluation of novel tacrine-caffeic acid hybrids as multi-targeted compounds against Alzheimer's disease. Bioorganic & medicinal chemistry letters 2012, 22, 6498502. Weggen S, Rogers M, Eriksen J. NSAIDs, small molecules for prevention of Alzheimer's disease or precursors for future drug development? Trends in pharmacological sciences 2007, 28, 536-43. Joo Y, Kim HS, Woo RS, Park CH, Shin KY, Lee JP, et al. Mefenamic acid shows neuroprotective effects and improves cognitive impairment in in vitro and in vivo Alzheimer's disease models. Molecular pharmacology 2006, 69, 76-84. Carreras I, Mckee AC, Choi JK, Aytan N, Kowall NW, Jenkins BG, et al. R-flurbiprofen improves tau, but not A beta pathology in a triple transgenic model of Alzheimer's disease. Brain research 2013, 1541, 115-27. Bornstein JJ, Eckroat TJ, Houghton JL, Jones CK, Green KD, Garneau-Tsodikova S. Tacrine-mefenamic acid hybrids for inhibition of acetylcholinesterase. MedChemComm 2011, 2, 406-12. Doherty GH. Nitric oxide in neurodegeneration, potential benefits of non-steroidal anti-inflammatories. Neurosci Bull 2011, 27, 36682. Chen Y, Sun J, Huang Z, Liao H, Peng S, Lehmann J, et al. Design, synthesis and evaluation of tacrine-flurbiprofen-nitrate trihybrids as novel anti-Alzheimer's disease agents. Bioorganic & medicinal chemistry 2013, 21, 2462-70. Mao F, Huang L, Luo Z, Liu A, Lu C, Xie Z, et al. O-Hydroxyl- or o-amino benzylamine-tacrine hybrids, multifunctional biometals chelators, antioxidants, and inhibitors of cholinesterase activity and amyloid-beta aggregation. Bioorganic & medicinal chemistry 2012, 20, 5884-92. Marco-Contelles J, Leon R, de los Rios C, Samadi A, Bartolini M, Andrisano V, et al. Tacripyrines, the first tacrine-dihydropyridine hybrids, as multitarget-directed ligands for the treatment of Alzheimer's disease. Journal of medicinal chemistry 2009, 52, 2724-32. Edraki N, Mehdipour AR, Khoshneviszadeh M, Miri R. Dihydropyridines, evaluation of their current and future pharmacological applications. Drug discovery today 2009, 14, 1058-66.

30 [174]

[175]

[176]

[177] [178]

[179]

[180]

[181] [182]

[183]

[184]

[185]

[186]

[187] [188]

[189]

[190]

[191]

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0 Kang S, Cooper G, Dunne SF, Luan CH, James Surmeier D, Silverman RB. Antagonism of L-type Ca2+ channels CaV1.3 and CaV1.2 by 1,4-dihydropyrimidines and 4H-pyrans as dihydropyridine mimics. Bioorganic & medicinal chemistry 2013, 21, 4365-73. Fu H, Li W, Lao Y, Luo J, Lee NT, Kan KK, et al. Bis(7)-tacrine attenuates beta amyloid-induced neuronal apoptosis by regulating L-type calcium channels. Journal of neurochemistry 2006, 98, 1400-10. De Caluwe J, Dupont G. The progression towards Alzheimer's disease described as a bistable switch arising from the positive loop between amyloids and Ca(2+). Journal of theoretical biology 2013, 331, 12-8. Lim D, Iyer A, Ronco V, Grolla AA, Canonico PL, Aronica E, et al. Amyloid beta deregulates astroglial mGluR5-mediated calcium signaling via calcineurin and Nf-kB. Glia 2013, 61, 1134-45. Wang Y, Wang F, Yu JP, Jiang FC, Guan XL, Wang CM, et al. Novel multipotent phenylthiazole-tacrine hybrids for the inhibition of cholinesterase activity, beta-amyloid aggregation and Ca(2)(+) overload. Bioorganic & medicinal chemistry 2012, 20, 6513-22. Rosini M, Simoni E, Bartolini M, Cavalli A, Ceccarini L, Pascu N, et al. Inhibition of acetylcholinesterase, beta-amyloid aggregation, and NMDA receptors in Alzheimer's disease, a promising direction for the multi-target-directed ligands gold rush. Journal of medicinal chemistry 2008, 51, 4381-4. Thiratmatrakul S, Yenjai C, Waiwut P, Vajragupta O, Reubroycharoen P, Tohda M, et al. Synthesis, biological evaluation and molecular modeling study of novel tacrine-carbazole hybrids as potential multifunctional agents for the treatment of Alzheimer's disease. Eur J Med Chem 2014, 75, 21-30. Gan CS, Zhou L, Zhao ZZ, Wang HS. Benzothiazole Schiff-bases as potential imaging agents for beta-amyloid plaques in Alzheimer's disease. Med Chem Res 2013, 22, 4069-74. Keri RS, Quintanova C, Marques SM, Esteves AR, Cardoso SM, Santos MA. Design, synthesis and neuroprotective evaluation of novel tacrine-benzothiazole hybrids as multi-targeted compounds against Alzheimer's disease. Bioorganic & medicinal chemistry 2013, 21, 4559-69. Camps P, Formosa X, Galdeano C, Munoz-Torrero D, Ramirez L, Gomez E, et al. Pyrano[3,2-c]quinoline-6-chlorotacrine hybrids as a novel family of acetylcholinesterase- and beta-amyloid-directed anti-Alzheimer compounds. Journal of medicinal chemistry 2009, 52, 5365-79. Antequera D, Bolos M, Spuch C, Pascual C, Ferrer I, FernandezBachiller MI, et al. Effects of a tacrine-8-hydroxyquinoline hybrid (IQM-622) on Abeta accumulation and cell death, involvement in hippocampal neuronal loss in Alzheimer's disease. Neurobiology of disease 2012, 46, 682-91. da Costa JS, Lopes JP, Russowsky D, Petzhold CL, Borges AC, Ceschi MA, et al. Synthesis of tacrine-lophine hybrids via one-pot four component reaction and biological evaluation as acetyl- and butyrylcholinesterase inhibitors. Eur J Med Chem 2013, 62, 55663. Li SY, Jiang N, Xie SS, Wang KD, Wang XB, Kong LY. Design, synthesis and evaluation of novel tacrine-rhein hybrids as multifunctional agents for the treatment of Alzheimer's disease. Organic & biomolecular chemistry 2014, 12, 801-14. Guo MZ, Li XS, Xu HR, Mei ZC, Shen W, Ye XF. Rhein inhibits liver fibrosis induced by carbon tetrachloride in rats. Acta pharmacologica Sinica 2002, 23, 739-44. Zhao YL, Zhou GD, Yang HB, Wang JB, Shan LM, Li RS, et al. Rhein protects against acetaminophen-induced hepatic and renal toxicity. Food and chemical toxicology , an international journal published for the British Industrial Biological Research Association 2011, 49, 1705-10. Li SY, Wang XB, Xie SS, Jiang N, Wang KD, Yao HQ, et al. Multifunctional tacrine-flavonoid hybrids with cholinergic, betaamyloid-reducing, and metal chelating properties for the treatment of Alzheimer's disease. Eur J Med Chem 2013, 69, 632-46. Alonso D, Dorronsoro I, Rubio L, Munoz P, Garcia-Palomero E, Del Monte M, et al. Donepezil-tacrine hybrid related derivatives as new dual binding site inhibitors of AChE. Bioorganic & medicinal chemistry 2005, 13, 6588-97. Camps P, Formosa X, Galdeano C, Gomez T, Munoz-Torrero D, Ramirez L, et al. Tacrine-based dual binding site

Matias et al.

[192]

[193]

[194]

[195]

[196]

[197]

[198]

[199]

[200]

[201]

[202]

[203] [204] [205]

[206] [207]

acetylcholinesterase inhibitors as potential disease-modifying antiAlzheimer drug candidates. Chemico-biological interactions 2010, 187, 411-5. Piazzi L, Rampa A, Bisi A, Gobbi S, Belluti F, Cavalli A, et al. 3(4-[[Benzyl(methyl)amino]methyl]phenyl)-6,7-dimethoxy-2H-2chromenone (AP2238) inhibits both acetylcholinesterase and acetylcholinesterase-induced beta-amyloid aggregation, a dual function lead for Alzheimer's disease therapy. Journal of medicinal chemistry 2003, 46, 2279-82. Rizzo S, Bartolini M, Ceccarini L, Piazzi L, Gobbi S, Cavalli A, et al. Targeting Alzheimer's disease, Novel indanone hybrids bearing a pharmacophoric fragment of AP2238. Bioorganic & medicinal chemistry 2010, 18, 1749-60. Bolea I, Gella A, Unzeta M. Propargylamine-derived multitargetdirected ligands, fighting Alzheimer's disease with monoamine oxidase inhibitors. Journal of neural transmission 2013, 120, 893902. Bolea I, Juarez-Jimenez J, de Los Rios C, Chioua M, Pouplana R, Luque FJ, et al. Synthesis, biological evaluation, and molecular modeling of donepezil and N-[(5-(benzyloxy)-1-methyl-1H-indol2-yl)methyl]-N-methylprop-2-yn-1-amine hybrids as new multipotent cholinesterase/monoamine oxidase inhibitors for the treatment of Alzheimer's disease. Journal of medicinal chemistry 2011, 54, 8251-70. Bautista-Aguilera OM, Esteban G, Bolea I, Nikolic K, Agbaba D, Moraleda I, et al. Design, synthesis, pharmacological evaluation, QSAR analysis, molecular modeling and ADMET of novel donepezil-indolyl hybrids as multipotent cholinesterase/monoamine oxidase inhibitors for the potential treatment of Alzheimer's disease. Eur J Med Chem 2014, 75, 82-95. Samadi A, Estrada M, Perez C, Rodriguez-Franco MI, Iriepa I, Moraleda I, et al. Pyridonepezils, new dual AChE inhibitors as potential drugs for the treatment of Alzheimer's disease, synthesis, biological assessment, and molecular modeling. Eur J Med Chem 2012, 57, 296-301. Samadi A, de la Fuente Revenga M, Perez C, Iriepa I, Moraleda I, Rodriguez-Franco MI, et al. Synthesis, pharmacological assessment, and molecular modeling of 6-chloro-pyridonepezils, new dual AChE inhibitors as potential drugs for the treatment of Alzheimer's disease. Eur J Med Chem 2013, 67, 64-74. Hu Y, Zhang J, Chandrashankra O, Ip FC, Ip NY. Design, synthesis and evaluation of novel heterodimers of donepezil and huperzine fragments as acetylcholinesterase inhibitors. Bioorganic & medicinal chemistry 2013, 21, 676-83. Tao Y, Fang L, Yang Y, Jiang H, Yang H, Zhang H, et al. Quantitative proteomic analysis reveals the neuroprotective effects of huperzine A for amyloid beta treated neuroblastoma N2a cells. Proteomics 2013, 13, 1314-24. Ziemianin A, Ronco C, Dole R, Jean L, Renard PY, Lange CM. Screening of new huprines--inhibitors of acetylcholinesterases by electrospray ionization ion trap mass spectrometry. Journal of pharmaceutical and biomedical analysis 2012, 70, 1-5. Arce MP, Rodriguez-Franco MI, Gonzalez-Munoz GC, Perez C, Lopez B, Villarroya M, et al. Neuroprotective and cholinergic properties of multifunctional glutamic acid derivatives for the treatment of Alzheimer's disease. Journal of medicinal chemistry 2009, 52, 7249-57. Ji HF, Shen L. Molecular basis of inhibitory activities of berberine against pathogenic enzymes in Alzheimer's disease. TheScientificWorldJournal 2012, 2012, 823201. Jung HA, Min BS, Yokozawa T, Lee JH, Kim YS, Choi JS. AntiAlzheimer and antioxidant activities of Coptidis Rhizoma alkaloids. Biological & pharmaceutical bulletin 2009, 32, 1433-8. Zhu F, Qian C. Berberine chloride can ameliorate the spatial memory impairment and increase the expression of interleukin1beta and inducible nitric oxide synthase in the rat model of Alzheimer's disease. BMC neuroscience 2006, 7, 78. Riviere C, Richard T, Vitrac X, Merillon JM, Valls J, Monti JP. New polyphenols active on beta-amyloid aggregation. Bioorganic & medicinal chemistry letters 2008, 18, 828-31. Yan JJ, Cho JY, Kim HS, Kim KL, Jung JS, Huh SO, et al. Protection against beta-amyloid peptide toxicity in vivo with longterm administration of ferulic acid. British journal of pharmacology 2001, 133, 89-96.

Highlights on Molecular Hybrids for CNS Disorders [208]

[209]

[210]

[211]

[212]

[213]

[214]

[215]

[216] [217]

[218]

[219] [220] [221] [222] [223]

[224] [225] [226] [227]

[228]

Tan DX, Manchester LC, Terron MP, Flores LJ, Reiter RJ. One molecule, many derivatives, a never-ending interaction of melatonin with reactive oxygen and nitrogen species? Journal of pineal research 2007, 42, 28-42. Jiang H, Wang X, Huang L, Luo Z, Su T, Ding K, et al. Benzenediol-berberine hybrids, multifunctional agents for Alzheimer's disease. Bioorganic & medicinal chemistry 2011, 19, 7228-35. Huang L, Su T, Shan W, Luo Z, Sun Y, He F, et al. Inhibition of cholinesterase activity and amyloid aggregation by berberinephenyl-benzoheterocyclic and tacrine-phenyl-benzoheterocyclic hybrids. Bioorganic & medicinal chemistry 2012, 20, 3038-48. Alcala Mdel M, Vivas NM, Hospital S, Camps P, Munoz-Torrero D, Badia A. Characterisation of the anticholinesterase activity of two new tacrine-huperzine A hybrids. Neuropharmacology 2003, 44, 749-55. Camps P, Formosa X, Munoz-Torrero D, Petrignet J, Badia A, Clos MV. Synthesis and pharmacological evaluation of huprine-tacrine heterodimers, subnanomolar dual binding site acetylcholinesterase inhibitors. Journal of medicinal chemistry 2005, 48, 1701-4. Galdeano C, Viayna E, Sola I, Formosa X, Camps P, Badia A, et al. Huprine-Tacrine Heterodimers as Anti-Amyloidogenic Compounds of Potential Interest against Alzheimer's and Prion Diseases. Journal of medicinal chemistry 2012, 55, 661-9. Viayna E, Sola I, Bartolini M, De Simone A, Tapia-Rojas C, Serrano FG, et al. Synthesis and multitarget biological profiling of a novel family of rhein derivatives as disease-modifying antiAlzheimer agents. Journal of medicinal chemistry 2014, 57, 254967. Sebestik J, Marques SM, Fale PL, Santos S, Arduino DM, Cardoso SM, et al. Bifunctional phenolic-choline conjugates as antioxidants and acetylcholinesterase inhibitors. Journal of enzyme inhibition and medicinal chemistry 2011, 26, 485-97. Gaki GS, Papavassiliou AG. Oxidative Stress-Induced Signaling Pathways Implicated in the Pathogenesis of Parkinson's Disease. Neuromol Med 2014, 16, 217-30. Westerlund M, Belin AC, Felder MR, Olson L, Galter D. High and complementary expression patterns of alcohol and aldehyde dehydrogenases in the gastrointestinal tract, implications for Parkinson's disease. The FEBS journal 2007, 274, 1212-23. Marklund P, Larsson A, Elgh E, Linder J, Riklund KA, Forsgren L, et al. Temporal dynamics of basal ganglia under-recruitment in Parkinson's disease, transient caudate abnormalities during updating of working memory. Brain , a journal of neurology 2009, 132, 336-46. McCarthy MM, Ching S, Whittington MA, Kopell N. Dynamical changes in neurological diseases and anesthesia. Current opinion in neurobiology 2012, 22, 693-703. Fernandez-Espejo E. Pathogenesis of Parkinson's disease, prospects of neuroprotective and restorative therapies. Molecular neurobiology 2004, 29, 15-30. Aarsland D, Larsen JP, Cummins JL, Laake K. Prevalence and clinical correlates of psychotic symptoms in Parkinson disease, a community-based study. Archives of neurology 1999, 56, 595-601. Brichta L, Greengard P, Flajolet M. Advances in the pharmacological treatment of Parkinson's disease, targeting neurotransmitter systems. Trends Neurosci 2013, 36, 543-54. Dalpiaz A, Cacciari B, Vicentini CB, Bortolotti F, Spalluto G, Federico S, et al. A novel conjugated agent between dopamine and an A2A adenosine receptor antagonist as a potential anti-Parkinson multitarget approach. Molecular pharmaceutics 2012, 9, 591-604. Pinnen F, Cacciatore I, Cornacchia C, Mollica A, Sozio P, Cerasa LS, et al. CNS delivery of L-dopa by a new hybrid glutathionemethionine peptidomimetic prodrug. Amino acids 2012, 42, 261-9. Devos D, Moreau C, Dujardin K, Cabantchik I, Defebvre L, Bordet R. New Pharmacological Options for Treating Advanced Parkinson's Disease. Clinical therapeutics 2013, 35, 1640-52. Molema G, Meijer DK. Drug targeting, organ-specific strategies, Wiley-VCH, 2001. Dutta AK, Fei XS, Reith ME. A novel series of hybrid compounds derived by combining 2-aminotetralin and piperazine fragments, binding activity at D2 and D3 receptors. Bioorganic & medicinal chemistry letters 2002, 12, 619-22. Levesque D, Diaz J, Pilon C, Martres MP, Giros B, Souil E, et al. Identification, characterization, and localization of the dopamine

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

[229]

[230] [231]

[232]

[233] [234]

[235]

[236] [237]

[238] [239] [240]

[241]

[242]

[243]

[244]

[245]

[246]

31

D3 receptor in rat brain using 7-[3H]hydroxy-N,N-di-n-propyl-2aminotetralin. Proceedings of the National Academy of Sciences of the United States of America 1992, 89, 8155-9. Cheung TH, Nolan BC, Hammerslag LR, Weber SM, Durbin JP, Peartree NA, et al. Phenylpiperazine derivatives with selectivity for dopamine D3 receptors modulate cocaine self-administration in rats. Neuropharmacology 2012, 63, 1346-59. Joyce JN, Millan MJ. Dopamine D3 receptor agonists for protection and repair in Parkinson's disease. Current opinion in pharmacology 2007, 7, 100-5. Dutta AK, Venkataraman SK, Fei XS, Kolhatkar R, Zhang S, Reith ME. Synthesis and biological characterization of novel hybrid 7[[2-(4-phenyl-piperazin-1-yl)-ethyl]-propyl-amino]-5,6,7,8tetrahydro-naphthale n-2-ol and their heterocyclic bioisosteric analogues for dopamine D2 and D3 receptors. Bioorganic & medicinal chemistry 2004, 12, 4361-73. Ghosh B, Antonio T, Zhen J, Kharkar P, Reith ME, Dutta AK. Development of (S)-N6-(2-(4-(isoquinolin-1-yl)piperazin-1yl)ethyl)-N6-propyl-4,5,6,7-tetrahydro benzo[d]-thiazole-2,6diamine and its analogue as a D3 receptor preferring agonist, potent in vivo activity in Parkinson's disease animal models. Journal of medicinal chemistry 2010, 53, 1023-37. Marosi K, Bori Z, Hart N, Sarga L, Koltai E, Radak Z, et al. Longterm exercise treatment reduces oxidative stress in the hippocampus of aging rats. Neuroscience 2012, 226, 21-8. Lee DH, Gold R, Linker RA. Mechanisms of Oxidative Damage in Multiple Sclerosis and Neurodegenerative Diseases, Therapeutic Modulation via Fumaric Acid Esters. International journal of molecular sciences 2012, 13, 11783-803. Yoo YJ, Nam DH, Jung SY, Jang JW, Kim HJ, Jin C, et al. Synthesis of cinnamoyl ketoamides as hybrid structures of antioxidants and calpain inhibitors. Bioorganic & medicinal chemistry letters 2011, 21, 2850-4. Kohli V, Madden JF, Bentley RC, Clavien PA. Calpain mediates ischemic injury of the liver through modulation of apoptosis and necrosis. Gastroenterology 1999, 116, 168-78. Liang B, Duan BY, Zhou XP, Gong JX, Luo ZG. Calpain activation promotes BACE1 expression, amyloid precursor protein processing, and amyloid plaque formation in a transgenic mouse model of Alzheimer disease. The Journal of biological chemistry 2010, 285, 27737-44. Lee KY, Lee KS, Jin C, Lee YS. Design and synthesis of calpain inhibitory 6-pyridone 2-carboxamide derivatives. Eur J Med Chem 2009, 44, 1331-4. Auvin S, Pignol B, Navet E, Pons D, Marin JG, Bigg D, et al. Novel dual inhibitors of calpain and lipid peroxidation. Bioorganic & medicinal chemistry letters 2004, 14, 3825-8. Auvin S, Pignol B, Navet E, Troadec M, Carre D, Camara J, et al. Novel dual inhibitors of calpain and lipid peroxidation with enhanced cellular activity. Bioorganic & medicinal chemistry letters 2006, 16, 1586-9. Koufaki M, Theodorou E, Galaris D, Nousis L, Katsanou ES, Alexis MN. Chroman/catechol hybrids, Synthesis and evaluation of their activity against oxidative stress induced cellular damage. Journal of medicinal chemistry 2006, 49, 300-6. Koufaki M, Theodorou E, Alexi X, Alexis MN. Synthesis of a second generation chroman/catechol hybrids and evaluation of their activity in protecting neuronal cells from oxidative stress-induced cell death. Bioorganic & medicinal chemistry 2010, 18, 3898-909. Kennedy BP, Ziegler MG, Alford M, Hansen LA, Thal LJ, Masliah E. Early and persistent alterations in prefrontal cortex MAO A and B in Alzheimer's disease. Journal of neural transmission 2003, 110, 789-801. Aluf Y, Vaya J, Khatib S, Loboda Y, Finberg JP. Selective inhibition of monoamine oxidase A or B reduces striatal oxidative stress in rats with partial depletion of the nigro-striatal dopaminergic pathway. Neuropharmacology 2013, 65, 48-57. Pons C, Dansette PM, Gregeois J, Homberg JC, Billett EE, Mansuy D. Human anti-mitochondria autoantibodies appearing in iproniazid-induced immunoallergic hepatitis recognize human liver monoamine oxidase B. Biochemical and biophysical research communications 1996, 218, 118-24. Binda C, Aldeco M, Geldenhuys WJ, Tortorici M, Mattevi A, Edmondson DE. Molecular Insights into Human Monoamine

32

[247]

[248] [249]

[250] [251]

[252]

[253]

[254] [255]

[256]

[257]

Mini-Reviews in Medicinal Chemistry, 2017, Vol. 17, No. 0

Matias et al.

Oxidase B Inhibition by the Glitazone Antidiabetes Drugs. ACS medicinal chemistry letters 2012, 3, 39-42. Carradori S, D'Ascenzio M, De Monte C, Secci D, Yanez M. Synthesis and selective human monoamine oxidase B inhibition of heterocyclic hybrids based on hydrazine and thiazole scaffolds. Archiv der Pharmazie 2013, 346, 17-22. Blier P, El Mansari M. Serotonin and beyond, therapeutics for major depression. Philos T R Soc B 2013, 368. Orus L, Perez-Silanes S, Oficialdegui AM, Martinez-Esparza J, Del Castillo JC, Mourelle M, et al. Synthesis and molecular modeling of new 1-aryl-3-[4-arylpiperazin-1-yl]-1-propane derivatives with high affinity at the serotonin transporter and at 5-HT(1A) receptors. Journal of medicinal chemistry 2002, 45, 4128-39. Traynor K. Vilazodone approved for major depression. Am J Health Syst Pharm 2011, 68, 366. Bartoszyk GD, Hegenbart R, Ziegler H. EMD 68843, a serotonin reuptake inhibitor with selective presynaptic 5-HT1A receptor agonistic properties. European journal of pharmacology 1997, 322, 147-53. Heinrich T, Bottcher H, Gericke R, Bartoszyk GD, Anzali S, Seyfried CA, et al. Synthesis and structure--activity relationship in a class of indolebutylpiperazines as dual 5-HT(1A) receptor agonists and serotonin reuptake inhibitors. Journal of medicinal chemistry 2004, 47, 4684-92. Heinrich T, Bottcher H, Schiemann K, Holzemann G, Schwarz M, Bartoszyk GD, et al. Dual 5-HT1A agonists and 5-HT re-uptake inhibitors by combination of indole-butyl-amine and chromenonylpiperazine structural elements in a single molecular entity. Bioorganic & medicinal chemistry 2004, 12, 4843-52. O'Brien CP. Anticraving medications for relapse prevention, A possible new class of psychoactive medications. Am J Psychiat 2005, 162, 1423-31. Choi SW, Elmaleh DR, Hanson RN, Shoup TM, Fischman AJ. Novel (bisarylmethoxy)butylpiperidine analogues as neurotransmitter transporter inhibitors with activity at dopamine receptor sites. Bioorganic & medicinal chemistry 2002, 10, 4091102. Harrington RA, Lee LC, Crum RM, Zimmerman AW, HertzPicciotto I. Serotonin hypothesis of autism, implications for selective serotonin reuptake inhibitor use during pregnancy. Autism research, official journal of the International Society for Autism Research 2013, 6, 149-68. Ghoneim OM, Ibrahim DA, El-Deeb IM, Lee SH, Booth RG. A novel potential therapeutic avenue for autism, design, synthesis and pharmacophore generation of SSRIs with dual action. Bioorganic & medicinal chemistry letters 2011, 21, 6714-23.

[258]

[259] [260]

[261] [262]

[263] [264]

[265] [266] [267]

[268] [269] [270]

Vohora D, Bhowmik M. Histamine H3 receptor antagonists/inverse agonists on cognitive and motor processes, relevance to Alzheimer's disease, ADHD, schizophrenia, and drug abuse. Frontiers in systems neuroscience 2012, 6, 72. Ellenbroek BA, Ghiabi B. The other side of the histamine H3 receptor. Trends Neurosci 2014, 37, 191-9. Grassmann S, Apelt J, Sippl W, Ligneau X, Pertz HH, Zhao YH, et al. Imidazole derivatives as a novel class of hybrid compounds with inhibitory histamine N-methyltransferase potencies and histamine hH(3) receptor affinities. Bioorganic & medicinal chemistry 2003, 11, 2163-74. Celanire S, Wijtmans M, Talaga P, Leurs R, de Esch IJP. Histamine H-3 receptor antagonists reach out for the clinic. Drug discovery today 2005, 10, 1613-27. Cumming P, Vincent SR. Inhibition of Histamine-NMethyltransferase (Hnmt) by Fragments of 9-Amino-1,2,3,4Tetrahydroacridine (Tacrine) and by Beta-Carbolines. Biochemical pharmacology 1992, 44, 989-92. Tripathi AC, Gupta SJ, Fatima GN, Sonar PK, Verma A, Saraf SK. 4-Thiazolidinones, the advances continue. Eur J Med Chem 2014, 72, 52-77. Thirumurugan R, Sriram D, Saxena A, Stables J, Yogeeswari P. 2,4-Dimethoxyphenylsemicarbazones with anticonvulsant activity against three animal models of seizures, synthesis and pharmacological evaluation. Bioorganic & medicinal chemistry 2006, 14, 3106-12. Pandey S. Synthesis and characterization of some heterocyclic schiff bases, potential anticonvulsant agents. Med Chem Res 2011, 20, 1091–101. Fasolato C, Bertazzon A, Previero A, Galzigna L. Effect of 2pyrrolidone on the concentration of GABA in rat tissues. Pharmacology 1988, 36, 258-64. Yuan H, Silverman RB. New substrates and inhibitors of gammaaminobutyric acid aminotransferase containing bioisosteres of the carboxylic acid group, design, synthesis, and biological activity. Bioorganic & medicinal chemistry 2006, 14, 1331-8. Rishipathak D, Shirodkar P. Design and Molecular Docking Studies of Some 1,3,4-Oxadiazole Derivatives. Int J Pharm Sci Rev Res 2013, 20, 285-9. De Luca L. Naturally occurring and synthetic imidazoles, their chemistry and their biological activities. Current medicinal chemistry 2006, 13, 1-23. Ahmed T, Gilani AU, Abdollahi M, Daglia M, Nabavi SF, Nabavi SM. Berberine and neurodegeneration, A review of literature. Pharmacological reports, PR 2015, 67, 970-9.

DISCLAIMER: The above article has been published in Epub (ahead of print) on the basis of the materials provided by the author. The Editorial Department reserves the right to make minor modifications for further improvement of the manuscript.

PMID: 27834131