Sie sind auf Seite 1von 11

Send Orders of Reprints at reprints@benthamscience.

net
1662 Current Medicinal Chemistry, 2013, 20, 1662-1672

Rationally Designed Multi-Targeted Agents Against Neurodegenerative


Diseases

W. J. Geldenhuys and C. J. Van der Schyf*

Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH
44272, USA

Abstract: Neurodegenerative diseases are complex disorders with several pathoetiological pathways leading to cell death.
Rationally designed multi-targeted agents, or “multi-targeted designed drugs” (MTDD) show significant promise in pre-
clinical studies as neuroprotective and disease-modifying agents. In this review, we highlight the use of chemical scaffolds
that lend themselves exquisitely to the development of MTDDs in neurodegeneration. Notably, synthetic polycyclic cage
compounds have served as scaffolds for novel voltage-gated calcium channel blockers, NMDA receptor antagonists, and
sigma-receptor ligands – attractive targets in neurodegeneration. In an entirely different approach, compounds containing
the thiazolidinedione moiety (referred to as glitazones) alter mitochondrial function through the mitochondrial protein mi-
toNEET, an attractive new drug target for the treatment of neurodegenerative diseases. The design strategy for yet another
agent, ladostigil, employed the amalgamation of active chemical moieties of the AChE inhibitor rivastigmine, and the
monoamine oxidase-B (MAO-B) inhibitor rasagiline, leading to a single compound that targets both enzymes simultane-
ously. Natural products have also served as design templates for several MTDD design studies. In particular, the stilbene
scaffold has become popular in particular due to the neuroprotective effects of the non-flavonoid natural product resvera-
trol. Recently, stilbene scaffold-based compounds were developed to reduce – through chelation with metal ions that in-
teract with beta-amyloid – both metal-induced beta-amyloid protein aggregation, and ROS generated from this aggregate.
Other subtle modifications of the stilbene motif led to the creation of reversible, non-competitive MAO inhibitors. Finally,
compounds derived from the xanthine scaffold afford neuroprotection in Parkinson’s disease through mechanisms that in-
clude dual adenosine A2A receptor antagonism and MAO-B inhibition.
Keywords: Multifunctional agents, multi-targeted designed drugs, neurodegenerative diseases, drug discovery.

1. INTRODUCTION would be akin to the discovery of antibiotics in the treatment


of bacterial infections.
As our society ages, we are confronted with an increase
in age-related disorders including neurodegenerative diseases What constitutes the major reasons for the fact that no
such as Alzheimer’s and Parkinson’s disease, Lewy body disease-modifying agents have been brought to the clinic to
disease, post-stroke degeneration, and frontotemporal de- date? This simple question may relate to the methods by
mentias among others. Patients who suffer from these neu- which drug discovery endeavors have been practiced in the
rodegenerative diseases, have increased morbidity and mor- last century. Pharmaceutical companies were traditionally
tality, that in turn also affect society through significantly engaged in developing compounds that display a single ac-
increased health care costs [1], negatively impacting the tivity profile at very high potency – a single “magic bullet”
economies of entire countries. Currently, treatment options which would interact with a singular drug target [4, 5]. In
for patients diagnosed with neurodegenerative diseases are contrast - with neurodegeneration - this concept has not met
limited. Disease modifying therapeutics have yet to emerge with expected success such as accomplished, for example, by
from current research endeavors. Essentially, those drugs treating hypertension with calcium channel antagonists [5].
currently approved only afford symptomatic relief from the A contrasting but alternative strategy is proposed that pur-
disease. For example, Parkinson’s disease treatment consists ports to target multiple drug intervention points in complex
of approaches aimed at correcting the loss of fine motor con- diseases such as neurodegeneration to fundamentally alter
trol, but affords little or no mitigation of the progression of disease progression – a “shotgun” effect [5, 6].
the disease. These medications do not alter the rate or extent Over the past few years, several reviews have indicated
of neuronal cell loss [2, 3]. As far as drug discovery in neu- that this approach is leading to a paradigm shift in drug de-
rodegeneration goes, the development of disease-modifying velopment for neurodegenerative diseases [1, 4, 5, 7-9]. Ad-
agents for the treatment of neurodegenerative disorders ditionally, several studies have now been published in which
active drug design techniques were used to construct multi-
ple-ligand or multifunctional drugs. Morphy and Rankovic
*Address correspondence to this author at the Department of Pharmaceuti-
cal Sciences, College of Pharmacy, Northeast Ohio Medical University, tallied the number of reports describing this active discovery
Rootstown, OH 44272, USA; Tel: +1-330-325-6467; Fax: +1-330-325- of designed multiple-ligand drugs (See Fig. 1). Between
5936; E-mail: cvanders@neomed.edu 1994 and 2004 over 300 reports were published in the

1875-533X/13 $58.00+.00 © 2013 Bentham Science Publishers


Rationally Designed Multi-Targeted Agents Current Medicinal Chemistry, 2013, Vol. 20, No. 13 1663

Traditional Drug Design MTDD


Methods

Drug 1 Drug 2

Drug Target 1 Drug Target 1 Drug Target 2 Drug Target 3

Treatment Treatment

Fig. (1). The development of therapeutic agents has traditionally been focused on one drug target, but the complexity of neurodegenerative
disease has necessitated a new approach whereby one chemical agent is designed to act via several disease pathways.

medicinal chemistry literature regarding the design of com- drug targets to prevent or attenuate neuronal cell death [13,
pounds targeted to more than one rational drug target. The 15]. Additionally, several studies have suggested that a com-
diseases covered encompassed many conditions, most nota- bination of VGCC and NMDAR blockers may be beneficial
bly cancer and neurodegeneration. Both cancer and neurode- as a disease-modifying strategy as compared to blocking
generation have several common themes, of which multiple either of these ion channels in isolation [16, 17].
compensatory mechanisms underlie the basis of multifunc-
Memantine is an adamantanamine with a polycyclic cage
tional drug design [10].
structure (Fig. 2), approved for the treatment of moderate to
In neurodegenerative disease, much of the complexity be- severe Alzheimer’s disease (Trade name: Namenda®) [3,
tween linked signaling pathways is still poorly understood. 18]. Memantine is a derivative of the anti-parkinsonian and
In the case of neurodegenerative disorders, the targeting of a antiviral agent amantadine [19], and has been extensively
single disease pathway may therefore not be sufficient to studied over the past few decades in the treatment of neu-
modulate the cell death process due to compensatory mecha- rodegeneration, both for Alzheimer’s and Parkinson’s dis-
nisms that reside outside the influenced of the drug. Several ease [20, 21]. From electrophysiological studies, it was
studies have indicated that the complexity of neurodegenera- shown that memantine has favorable NMDA modulation
tion requires a multifaceted approach [6, 10]. Additionally, activity due to its fast on-off kinetic profile at the receptor.
by using a single chemical moiety, the probability of serious There, in the presence of memantine, the ion channel of the
side effects is lessened, as compared to the classical poly- NMDAR is normalized in ion conductance as opposed to
pharmaceutical approach that requires the use of multiple MK-801 (dizocilpine) or phencyclidine (PCP), both of which
drug molecules in the treatment of patients [1]. block the channel completely – an effect that leads to hallu-
cinations and cell death [20]. From in vivo studies such as
In this review, we will consider select examples of de-
signed multiple ligand drugs that have been evaluated from stroke animal models, it has been suggested that the
combination of memantine (an NMDAR antagonist) and
against several drug targets currently thought to play a role
nimodipine (a VGCC blocker) is more beneficial than the
in the cell death cascades of neurons. Several challenges
respective individual therapies [22]. The putative positive
exist in the development of designed multiple ligand drugs.
effects of this combined therapy led to the development of
These include: protein targets that are key players in the cell
NGP1-01, a polycyclic cage amine, as a dual NMDAR and
death cascade; finding a balance in the affinity for each re-
spective target (i.e. the magnitude of affinity or activity VGCC blocker [23-25].
needs to be pharmacokinetically relevant); for CNS drugs, Prior to the introduction of amantadine in 1966 as a pro-
the ability to traverse the blood-brain barrier (BBB). The phylactic agent against Asian influenza, polycyclic cage
latter issue is of particular importance, because for CNS structures have not been explored for pharmacological prop-
drugs, a large proportion of novel compounds fail to reach erties [19]. The pentacycloundecane (PCU) cage structure
the clinic due to poor CNS penetration [11]. became prominent in the 1980s after the group of Van der
Schyf et al., demonstrated that their PCU-derived lead com-
2. CALCIUM CHANNEL MODULATORS pound, NGP1-01, blocks calcium influx into cells, specifi-
cally the L-type VGCC [25] (Fig. 2). Initially, the structural
Calcium plays an important role in normal neuronal
data of NGP1-01 was interpreted to contain an aza-
physiology [12-14]. However, under pathological conditions,
bridgehead in the cage (Fig. 2), but X-ray crystallography
an over-accumulation of calcium inside neurons may trigger
soon suggested that the cage was indeed oxa-bridged.
cell death signaling cascades. Two major conduits for cal-
cium entry into neuronal cells include voltage gated calcium The PCU is a versatile scaffold that can be used effec-
channels (VGCC) and the N-methyl-D-aspartate receptor/ion tively for the modification of a compound’s lipophilicity
channel (NMDAR). Both these ion channels have been asso- profile, for example to modify BBB permeability. In 2000
ciated with neuronal cell death and have been considered and 2003, the VGCC channel activities of a series of PCU
1664 Current Medicinal Chemistry, 2013, Vol. 20, No. 13 Geldenhuys and Schyf

derivatives of NGP1-01 were evaluated to determine struc- tor for NMDAR blocking has been corroborated by another
ture-activity trends [26, 27]. These studies indicated that study [28].
both size and electrostatic properties were important deter-
An early (1996) study has suggested that the combination
mining factors for activity at the VGCC. Substitution on the
of NMDAR and VGCC blockers may have improved neuro-
aromatic ring of the NGP1-01 structure could be used to in- protective potential in stroke models [22]. Based on this re-
crease VGCC blocking activity, with meta-substituted com-
port, NGP1-01 was evaluated in both a permanent and a
pounds being more active than ortho, and ortho substitution
transient cerebral artery occlusion model of ischemic stroke.
more active than para substitution. Also, the introduction of a
In both these models, NGP1-01 (20 mg/kg) led to a signifi-
methoxy instead of a nitro-group would generally lead to
cant reduction in both the swelling (>80%) and infarct area
increased activity. Notably, increasing the cage size (and
(>40%) of animals treated with NGP1-01 as compared to the
therefore the molecular volume) from a pentacyclic system vehicle controls. Behaviorally, animals treated with NGP1-
to a hexacyclic system, also led to an increase in activity.
01 performed significantly better than the vehicle control
These findings were also apparent in the QSAR studies per-
groups [29, 30]. This neuroprotective activity of NGP1-01
formed on the data set.
may also in part be associated with its ability to also control
NH2
the entry of iron into cells through the VGCC 31, 32. By block-
ing the VGCC, NGP1-01 may also mitigate iron accumula-
tion following ischemic stroke [31, 32].
H Recently, PCU compounds have also been identified as
N
highly potent sigma receptor ligands, a receptor type that has
O been implicated in neurodegeneration [33]. Several of these
compounds have been shown to have nanomolar affinity for
amantadine NGP1-01:
the sigma-1 and sigma-2 receptor [34-40].
NMDA: 2.4 μM
NH2 sigma-1: 1.5 μM
VGCC: 86 μM
3. MAO-INHIBITION AND ADENOSINE A2 (A2A) RE-
DAT: 55 μM CEPTOR ANTAGONISM
Converging lines of investigation have shown a benefi-
cial correlation between coffee drinkers and the decrease in
the incidence of Parkinson’s disease [41], an effect that has
since been attributed to the xanthine compound caffeine (Fig.
OH 3). Pharmacological studies have determined that caffeine is
memantine
N a non-selective adenosine receptor antagonist, and that sev-
eral caffeine derivatives exhibit mild monoamine oxidase
(MAO) B inhibitory activity. Many compounds of dietary
origin, such as those containing the xanthine scaffold of caf-
feine, can easily be modified to design both A2A and MAO-B
OH aza-NGP1-01 inhibitors [42]. The MPTP model of parkinsonism was util-
N
ized to show that the protective activity of caffeine could be
reversed by the addition of an A2A agonist, suggesting that
caffeine derived its neuroprotective activity in part through
inhibition of the A2A receptor [43]. The xanthine scaffold can
TC-1:
F further be modified to create compounds that can act as both
Sigma-1: 10 nM
Sigma-2: 370 nM
A2A antagonists and MAO-B inhibitors [44]. MAO-B activ-
ity is important in the metabolism of dopamine, and inhibi-
Fig. (2). Structures of the polycyclic cage amines described in the tion of the activity of this enzyme leads to symptomatic re-
text. Detailed descriptions of the synthetic routes to NGP1-01 and lief in Parkinson’s patients. The styrylxanthine or caffeinyl
aza-NGP1-01 have been provided earlier [24, 26, 27]. scaffold of compounds such as CSC (Fig. 3) and istradefyl-
line (KW-6002; Fig. 3) can be modified to yield A2A antago-
The NMDAR activity was evaluated for NGP1-01 in two nists [45] that also exhibit MAO-B inhibitory activity. A few
studies. In a microdialysis study by Kiewert et al., NGP1-01 notable SAR trends can be seen for MAO-B inhibition by the
was shown to reach the CNS and block both the NMDAR styrylxanthines [46-49]. Substitution on the aromatic styryl
and VGCCs [23]. As noted for VGCC blocking activity (see ring plays in important role in MAO-B inhibition. Electron
above) SAR studies of the PCU compounds at the NMDA withdrawing groups generally increase activity, with chlorine
receptor also indicated that the size/volume of the PCU is substitution leading this trend. Also, the steric configuration
important for NMDA receptor blocking. NGP1-01 was able of the stilbene is important, since the trans form is favored to
to block the NMDAR in murine synaptoneurosomes with an the cis form, cis istradefylline being inactive. Selectivity and
IC50 of 2.98 μM. This activity profile was similar to that of activity for MAO-B and A2A can also be optimized by in-
memantine under similar experimental conditions (meman- creasing the linker size of the stilbene, as evidenced by a
tine IC50 = 3.05 μM). Again, by increasing the ring size from series of (E,E)-phenylbutadiene caffeinyl derivatives. By
a 5- to a 6-membered ring, the activity was dramatically re- increasing the linker length, these caffeinyl derivatives were
duced (IC50 = 36 μM for the 6 membered NGP1-01 deriva- seen to be very potent (MAO-B and A2A low nM potencies),
tive). This finding that size is an important determining fac- and the selectivity of MAO-B over MAO-A could be in-
Rationally Designed Multi-Targeted Agents Current Medicinal Chemistry, 2013, Vol. 20, No. 13 1665

creased [46-49]. The substrate pocket of MAO-A is smaller generative diseases such as cerebral stroke [52, 53] and Park-
than that of MAO-B, and the (E,E)-phenylbutadiene caf- inson’s disease [54-60] (Fig. 4). Thiazolidinedione (TZD;
feinyl derivatives appear to be too large for a facile fit into Fig. 4) compounds are PPAR-gamma agonists used in the
the substrate pocket of MAO-A. But these compounds fit treatment of type II diabetes [61]. Both pioglitazone and
comfortably into the substrate cavity of MAO-B after span- rosiglitazone have been shown to have protective effect in
ning the entrance cavity [50, 51]. pre-clinical models of Parkinson’s disease, including the
gold standard MPTP and 6-OH-dopamine models [55-59,
O CH3 62]. The neuroprotective activities of TZDs have been sug-
H3C N gested to be unrelated to their PPAR-gamma activity [63].
caffeine N
Of interest was the finding that pioglitazone was beneficial
O N against striatal toxicity of MPTP in a rodent model. We have
N
recently shown that this activity is related to the MAO in-
CH3
hibitory activity of TZDs, pioglitazone was found to be the
CH3 O most potent inhibitor of MAO-B, followed by rosiglitazone,
CH3
and then ciglitazone [64]. Crystallographic studies of piogli-
N
N O CH3
tazone and rosiglitazone in the MAO-B enzyme indicates
istradyfilline (KW-6002)
that the TZD head group binds in the substrate cavity facing
MAO-B: 28 μM O N N O the FAD catalytic site, while the remainder of the molecule
A2A: 2.2 nM CH3 resides in the entrance cavity. The TZD moiety is important
H3C for PPAR-gamma binding, with the tail end of pioglitazone
O
and rosiglitazone binding into a Y-shaped binding pocket
CH3
[65-67]. Taken together, selective compounds can be de-
H3C
N
N Cl signed to bind to MAO-B and PPAR-gamma, either indi-
CSC vidually or simultaneously as multi-targeted ligands.
O N N
MAO-B: 70 nM 5. MAO-B AND METAL CHELATION
CH3
A2A: 54 nM Metals have been suggested to play in important role in
neurodegeneration [32, 68-71]. These elements have the abil-
Fig. (3). Structures of caffeinyl compounds described in the text. ity to generate ROS species which induce cell death, and
may also play a role in the deleterious effects of misfolded
4. MAO-INHIBITION AND PPAR-GAMMA AGO- proteins such as amyloid- in neurodegeneration [69]. For
NISM instance, significant accumulation of iron has been measured
in amyloid plaques as well as in neurofibrillary tangles in
Recently it was shown that peroxisome proliferator- Alzheimer’s disease patients [72]. Several groups have used
activated receptor gamma (PPAR-gamma) agonists such as metal chelation activity as starting point in designing multi-
pioglitazone and rosiglitazone are neuroprotective in several ple ligands. A series of compounds, also based on the stil-
neurodegenerative disease models, suggesting a promising bene scaffold were developed to interact with -amyloid and
basis for the design of multiple ligands targeted at neurode- simultaneously exhibit metal chelating activity [73-75]. By

HO
O
N
CH3
CH3
NH
N N
S O
CH3
O
O O O
TZD

O O O

NH NH NH
S S S

O O O

troglitazone pioglitazone rosiglitazone


MAO-B: 2.07 μM 298 nM 832 nM

PPR-gamma: 780 nM 550 nM 76 nM

Fig. (4). Thiazolidinedione (TZD) anti-diabetic drugs.


1666 Current Medicinal Chemistry, 2013, Vol. 20, No. 13 Geldenhuys and Schyf

chelating with metals in -amyloid, the resulting ROS levels Based on earlier findings that resveratrol, a stilbene, in-
should be reduced, with subsequent mitigation of protein hibits MAO, several stilbene-derived compounds identified
misfolding. A group of compounds was derived from from the metal chelating studies were screened in an MAO-B
thioflavin T (ThT) and 6-iodo-2-(4'-dimethylamino)phenyl- assay [77-79]. L2-b was found to be a facile MAO-B inhibi-
imidazo[1,2]pyridine (IMPY) [73-75] (Fig. 5). These com- tor (IC50 = 52 μM), and the compounds tested showed pre-
pounds served as lead structures for other compounds devel- ferred selectivity towards MAO-B compared to MAO-A
oped subsequently, and have an added bonus in that they are [80]. Substitution on the aromatic ring of the stilbene was
brain permeable, which is of key importance for CNS drug important for activity, with an increase in potency seen in a
delivery [11]. Metal chelating moieties were introduced series H<NH2<NMe2. L2-b was also shown to be a reversi-
based on the clioquinol template which utilizes the place- ble MAO-B inhibitor.
ment of hydroxyl and nitrogen groups in close proximity as a
design for the chelation of metals. One of the compounds, 6. AMYLOID- AND CHOLINESTERASES
L2-b (Fig. 5), was found to chelate with metal ions with rank
The polyamine-quinone memoquin has served as a tem-
potency of Cu2+/Zn2+>Fe2+>>Ca2+. L2-b as well as several of
plate structure for the development of molecules that prevent
the other derivatives tested, were able to prevent metal-
both amyloid- aggregation and act as acetylcholinesterase
induced -amyloid aggregation in a dose-dependent manner,
(AChE) inhibitors [81-91]. Several molecular properties that
and also reduced the resultant ROS to afford protective ef-
fects in cell culture [76]. are relevant to the treatment and prevention of neuronal cell
death in Alzheimer’s disease are present in these compounds,
including the ability to interfere with the processing and ag-
gregation of amyloid- peptides, the formation of reactive
oxygen species (ROS), and the activity of AChE. Memoquin
and several of its derivatives caused a significant decrease in
stilbene
amyloid- peptide- and ROS formation in animal models
[92-94]. Notably, in these studies, these compounds also
N CH3 were reported to facilitate the reversal of behavioral deficits.
N Recently, a novel series of memoquin derivatives was cre-
I
N
CH3 ated by linking the 2,5-diamino-benzoquinone core of these
compounds with motifs seen in known amyloid- binding
IMPY agents, including curcumin, a natural product of dietary ori-
gin [92-94]. In these compounds, weaker AChE inhibitory
potencies were measured, while retaining equipotent anti-A
N activities to that of memoquin. These findings suggested that
NH a balanced biological profile against the combined targets of
CH3
amyloid- and AChE could be achieved.
N
CH3 7. MAO-B AND CHOLINESTERASE INHIBITION

L2-b: One of the success stories in the development of multiple


targeted ligands is the development history of the multifunc-
MAO-B: Ki = 28 nM
tional ligand ladostigil (TV3326; Fig. 6). This compound is
Chelation: 40 μM currently in Phase II clinical trials (www.avphar.com) for the
treatment of mild cognitive impairment and comorbid de-
Fig. (5). “Dual” metal chelating agents and MAO inhibitors. pression seen in Alzheimer’s and Parkinson’s disease. La-

H3C CH3
N

CH3
CH
NH
O O
rasagiline
N CH3
H3C O

rivastigmine H3C
N O CH
NH
CH3

ladostigil

Fig. (6). Combination of rivastigmine (an AChE inhibitor) and rasagiline (an MAO inhibitor) led to the development of ladostigil.
Rationally Designed Multi-Targeted Agents Current Medicinal Chemistry, 2013, Vol. 20, No. 13 1667

dostigil was designed based on the merger of two active mo- heimer’s disease drug [116]. Here, the AChE inhibitor done-
lecular scaffolds that inhibit both AChE and MAO-B. Rivas- pezil (Fig. 8) was used to connect the 1-benzylpiperidine
tigmine (Fig. 6) is an AChE inhibitor which has been shown containing group with a moiety associated with MAO inhibi-
to be beneficial in the treatment of both Alzheimer’s disease tion, N-[(5-benzyloxy-1-methyl-1H-indol-2-yl)methyl]-N-
and Parkinson’s disease [95, 96]. Rasagiline (Fig. 6) is an methylprop-2-yn-1-amine (3). The moiety associated with
irreversible MAO-B inhibitor that has been approved by the AChE inhibition binds into the catalytic cavity of the AChE
FDA in 2006 for the treatment of Parkinson’s disease [97- enzyme, whereas the moiety associated with MAO inhibition
101]. Notably, rasagiline is one of few compounds that have binds into the substrate pocket of MAO. The most potent
been suggested to possess disease-modifying activity in compound (4) of the series was able to inhibit AChE (IC50 =
Parkinson’s disease [102-104]. 350 nM), BuChE (IC50 = 460 nM), MAO-A (IC50 = 5.2 nM)
Ladostigil as a multiple targeted ligand has been created and MAO-B (IC50 = 43 nM).
through a combination of the rasagiline scaffold containing a O
propargylamine moiety, and the carbamate moiety from the H3C
structure of rivastigmine. This combination of activities af-
O
forded ladostigil the ability to be used for symptomatic re-
lief, as well as the potential to act as a disease-modifying H3C
N
agent [104]. Specifically, the purported disease-modifying O

ability of this molecule has been attributed to the propargy-


lamine moiety [105-109]. In addition, some studies have
suggested that the carbamate part of ladostigil is neuropro-
tective via heat shock protein (HSP) activity [110]. Notably,
ladostigil has been shown to have beneficial effects on cog-
nition [109]. In addition, upregulation of ERK 1/2 and PKC,
CH
reduction of hydrogen peroxide, and prevention of the reduc-
tion in mitochondrial membrane potential all contribute to N H3C

the drug’s effects. It has also been shown to potentially act in O N

mitigating the progression of Alzheimer’s disease by increas-


ing the soluble fraction of amyloid precursor protein (APP) N
[106, 111-114]. CH3 4

The coumarin- and chromone-derived structures (Fig. 7;


1 and 2) have been shown to be useful scaffolds that can
accommodate both MAO and AChE inhibitory activities. In CH
a recent study, several related compounds were shown to be H3C
potent noncompetitive inhibitors of both enzymes [115]. O N
SAR studies in the coumarin scaffold indicated a loss of ac-
tivity for AChE to be associated with meta and/or para sub- N
stitution with a halogen moiety, while similar substitution CH3
led to an increase in MAO-B inhibitory activity, similar to
that seen in the styrylxanthines [45].
3
CH3
CH3
Fig. (8). Structures of novel multi-targeted AChE- and MAO-B
inhibitors described in the text. Structure of donepezil shown at the
top of the figure.
H3C O O

1
The donepezil scaffold (Fig. 8) has also led to other multi
targeted drug design programs developing novel compounds
O
for Alzheimer’s disease treatment [117]. The benzylidene-
O indanone scaffold was successfully employed to develop
MAO inhibitors that also prevented amyloid- aggregation,
O CH3 metal chelation, and also possess antioxidant activity. These
compounds, in particular compound 5 (Fig. 9), inhibited
MAO-B at an IC50 = 7.5 μM, inhibited amyloid- aggrega-
2: tion (at >80%), and was able to inhibit Cu-induced amyloid-
MAO-B: pIC50 = 6.90  fibril formation [117].
AChE: pKi = 4.53
8. HISTONE DEACETYLASE INHIBITORS AND PKC
Fig. (7). Coumarin derivatives that have been shown to serve as ACTIVATORS
templates for both MAO and AChE inhibitors.
A novel emerging strategy for the treatment of Alz-
The ability to inhibit both MAO and ChE enzymes was heimer’s disease is the combination of activators of protein
also investigated in the development of another MTDD Alz- kinase C (PKC), and histone deacetylase (HDAC) inhibitors,
1668 Current Medicinal Chemistry, 2013, Vol. 20, No. 13 Geldenhuys and Schyf

as was recently described [118]. The basis for this strategy of we show that there is great promise in developing disease-
designed multiple ligand drugs was the expectation that PKC modifying agents based on the premise of targeting more
kinase activation would likely increase the alpha-secretase than one disease pathway known to be important in neurode-
pathway thereby increasing the soluble fraction of APP [119, generation. In support of the validity of this approach are the
120]. Additionally HDAC inhibition is thought to be poten- promising results emanating from early clinical studies of
tially neuroprotective via the blocking of oxidative stress ladostigil, which is now in clinical trials, and the emergence
[121]. A series of benzolactams was evaluated for both PKC – in preclinical stages of development – of several new com-
activation via soluble APP determination and HDAC inhibi- pounds described in this review.
tory activity via a homocysteine neuronal model of oxidative
stress. The findings indicated that the benzolactams tested CONFLICT OF INTEREST
were nanomolar inhibitors of PKC and were also able to The author(s) confirm that this article content has no con-
show neuroprotection via HDAC inhibition (10 – 20 μM). flict of interest.
Two compounds (one being compound 6; Fig 10) stood out
in that they were equipotent against HDAC and PKC (~10 ACKNOWLEDGEMENT
μM for both drug targets). This finding is significant consid-
ering that one of the major hurdles in multiple ligand design Declared none.
is the ability to align pharmacological potencies at the indi-
vidual drug targets within a similar range [122]. These ben- LIST OF ABBREVIATIONS
zolactams represent a novel group of compounds that could AChE = Acetylcholine esterase
effectively be developed into disease-modifying agents for
BBB = Blood-brain barrier
Alzheimer’s disease.
CNS = Central nervous system
HO
FDA = Federal Drug Administration of the United
States
HO
IMPY = 6-Iodo-2-(4'-dimethylamino)phenyl-
O
imidazo[1,2]pyridine

N
HSP = Heat shock protein
H3C MAO = Monoamine oxidase
CH3 MTDD = Multi-targeted designed drugs
5: NMDAR = N-methyl-D-aspartate receptor
MAO-B: IC50 = 7.50 μM PCU = Pentacycloundecyl
Anti-amyloid-beta: 80.1% at 20 μM PKC = Protein kinase C
ThT = Thioflavin T
Fig. (9). A multifunctional drug that inhibits MAO-B as well as
prevents amyloid- aggregation. TZD = Thiazolidinedione

O VGCC = Voltage gated calcium channel

NH OH REFERENCES
HN
[1] Van der Schyf, C.J., The use of multi-target drugs in the treatment
of neurodegenerative diseases. Expert Rev. Clin. Pharmacol., 2011,
4(3), 293-298.
[2] Piau, A.; Nourhashemi, F.; Hein, C.; Caillaud, C.; Vellas, B.,
Progress in the development of new drugs in Alzheimer's disease.
AcS O J. Nutr. Health Aging, 2011, 15(1), 45-57.
6 [3] Schwarz, S.; Froelich, L.; Burns, A., Pharmacological treatment of
dementia. Curr. Opin. Psychiatry, 2012, 25(6), 542-550.
6: [4] Morphy, R.; Kay, C.; Rankovic, Z., From magic bullets to designed
PKC: Ki = 4.5 nM multiple ligands. Drug Discov. Today, 2004, 9(15), 641-651.
Histone h4 acetylation: 10 μM [5] Roth, B.L.; Sheffler, D.J.; Kroeze, W.K., Magic shotguns versus
magic bullets: selectively non-selective drugs for mood disorders
Fig. (10). Novel HDAC and PKC ligand. and schizophrenia. Nat. Rev. Drug Discov., 2004, 3(4), 353-359.
[6] Pruss, R.M., Phenotypic screening strategies for neurodegenerative
diseases: a pathway to discover novel drug candidates and potential
7. CONCLUSION disease targets or mechanisms. CNS Neurol. Disord. Drug Targets,
2010, 9(6), 693-700.
The complexity of neurodegeneration has resulted in few [7] Youdim, M.B.; Geldenhuys, W.J.; Van der Schyf, C.J., Why
compounds that can be construed as disease-modifying should we use multifunctional neuroprotective and neurorestorative
agents that can be used clinically. Many compounds that are drugs for Parkinson's disease? Parkinsonism Relat. Disord., 2007,
neuroprotective in vitro and in vivo fail in human trials due 13 Suppl 3, S281-291.
[8] Geldenhuys, W.J.; Youdim, M.B.; Carroll, R.T.; Van der Schyf,
to the complexity of the human disease condition not being C.J., The emergence of designed multiple ligands for
fully recapitulated in the animal-based disease models. Here
Rationally Designed Multi-Targeted Agents Current Medicinal Chemistry, 2013, Vol. 20, No. 13 1669

neurodegenerative disorders. Prog. Neurobiol., 2011, 94(4), 347- [31] Gaasch, J.A.; Geldenhuys, W.J.; Lockman, P.R.; Allen, D.D.; Van
359. der Schyf, C.J., Voltage-gated calcium channels provide an
[9] Cavalli, A.; Bolognesi, M.L.; Minarini, A.; Rosini, M.; Tumiatti, alternate route for iron uptake in neuronal cell cultures.
V.; Recanatini, M.; Melchiorre, C., Multi-target-directed ligands to Neurochem. Res., 2007, 32(10), 1686-1693.
combat neurodegenerative diseases. J. Med. Chem., 2008, 51(3), [32] Gaasch, J.A.; Lockman, P.R.; Geldenhuys, W.J.; Allen, D.D.; Van
347-372. der Schyf, C.J., Brain iron toxicity: differential responses of
[10] Plun-Favreau, H.; Lewis, P.A.; Hardy, J.; Martins, L.M.; Wood, astrocytes, neurons, and endothelial cells. Neurochem. Res., 2007,
N.W., Cancer and neurodegeneration: between the devil and the 32(7), 1196-1208.
deep blue sea. PLoS Genet., 2010, 6(12), e1001257. [33] Luedtke, R.R.; Perez, E.; Yang, S.H.; Liu, R.; Vangveravong, S.;
[11] Pardridge, W.M., Blood-brain barrier delivery. Drug Discov. Tu, Z.; Mach, R.H.; Simpkins, J.W., Neuroprotective effects of
Today, 2007, 12(1-2), 54-61. high affinity sigma 1 receptor selective compounds. Brain Res.,
[12] Martin, L.J., Biology of mitochondria in neurodegenerative 2012, 1441, 17-26.
diseases. Prog. Mol. Biol. Transl. Sci., 2012, 107, 355-415. [34] Banister, S.D.; Moussa, I.A.; Jordan, M.J.; Coster, M.J.; Kassiou,
[13] Hurley, M.J.; Dexter, D.T., Voltage-gated calcium channels and M., Oxo-bridged isomers of aza-trishomocubane sigma (sigma)
Parkinson's disease. Pharmacol. Ther., 2012, 133(3), 324-333. receptor ligands: Synthesis, in vitro binding, and molecular
[14] Surmeier, D.J., Calcium, ageing, and neuronal vulnerability in modeling. Bioorg. Med. Chem. Lett., 2010, 20(1), 145-148.
Parkinson's disease. Lancet Neurol., 2007, 6(10), 933-938. [35] Kassiou, M.; Nguyen, V.H.; Knott, R.; Christie, M.; Hambley,
[15] Kemp, J.A.; McKernan, R.M., NMDA receptor pathways as drug T.W., Trishomocubanes, a new class of selective and high affinity
targets. Nat. Neurosci., 2002, 5 Suppl, 1039-1042. ligands for the simga binding site. Bioorg. Med. Chem. Lett., 1996,
[16] Lippert, K.; Welsch, M.; Krieglstein, J., Over-additive protective 6, 595-600.
effect of dizocilpine and NBQX against neuronal damage. Eur. J. [36] Liu, X.; Banister, S.D.; Christie, M.J.; Banati, R.; Meikle, S.;
Pharmacol., 1994, 253(3), 207-213. Coster, M.J.; Kassiou, M., Trishomocubanes: novel sigma ligands
[17] Krieglstein, J.; Lippert, K.; Poch, G., Apparent independent action modulate cocaine-induced behavioural effects. Eur. J. Pharmacol.,
of nimodipine and glutamate antagonists to protect cultured 2007, 555(1), 37-42.
neurons against glutamate-induced damage. Neuropharmacology, [37] Liu, X.; Kassiou, M.; Christie, M.; Hambley, T.W.,
1996, 35(12), 1737-1742. Trishomocubanes: requirements for sigma receptor binding and
[18] Rodda, J.; Carter, J., Cholinesterase inhibitors and memantine for subtype selectivity. Aust. J. Chem., 2001, 54, 157-163.
symptomatic treatment of dementia. BMJ, 2012, 344, e2986. [38] Liu, X.; Mattner, F.; Katsifis, A.; Christie, M.; Kassiou, M.,
[19] Geldenhuys, W.J.; Malan, S.F.; Bloomquist, J.R.; Marchand, A.P.; Influence of trishomocubanes on sigma receptor binding of N-(1-
Van der Schyf, C.J., Pharmacology and structure-activity benzyl-piperidin-4-yl)-4-[123I]iodobenzamide in vivo in the rat
relationships of bioactive polycyclic cage compounds: a focus on brain. Med. Chem., 2005, 1(1), 31-38.
pentacycloundecane derivatives. Med. Res. Rev., 2005, 25(1), 21- [39] Liu, X.; Nuwayhid, S.; Christie, M.J.; Kassiou, M.; Werling, L.L.,
48. Trishomocubanes: novel sigma-receptor ligands modulate
[20] Parsons, C.G.; Danysz, W.; Quack, G., Memantine is a clinically amphetamine-stimulated [3H]dopamine release. Eur. J.
well tolerated N-methyl-D-aspartate (NMDA) receptor antagonist-- Pharmacol., 2001, 422(1-3), 39-45.
a review of preclinical data. Neuropharmacology, 1999, 38(6), 735- [40] Nguyen, V.H.; Mardon, K.; Kassiou, M.; Christie, M.D., In vitro
767. and in vivo characterisation of [3H]ANSTO-14 binding to the
[21] Danysz, W.; Parsons, C.G.; Kornhuber, J.; Schmidt, W.J.; Quack, sigma 1 binding sites. Nucl. Med. Biol., 1999, 26(2), 209-215.
G., Aminoadamantanes as NMDA receptor antagonists and [41] Campdelacreu, J., Parkinson disease and Alzheimer disease:
antiparkinsonian agents--preclinical studies. Neurosci. Biobehav. environmental risk factors. Neurologia, 2012, [Epub ahead of
Rev., 1997, 21(4), 455-468. Print].
[22] Stuiver, B.T.; Douma, B.R.; Bakker, R.; Nyakas, C.; Luiten, P.G., [42] Geldenhuys, W.J.; Bishayee, A.; Darvesh, A.S.; Carroll, R.T.,
In vivo protection against NMDA-induced neurodegeneration by Natural products of dietary origin as lead compounds in virtual
MK-801 and nimodipine: combined therapy and temporal course of screening and drug design. Curr. Pharm. Biotechnol., 2012, 13(1),
protection. Neurodegeneration, 1996, 5(2), 153-159. 117-124.
[23] Kiewert, C.; Hartmann, J.; Stoll, J.; Thekkumkara, T.J.; Van der [43] Chen, J.F.; Xu, K.; Petzer, J.P.; Staal, R.; Xu, Y.H.; Beilstein, M.;
Schyf, C.J.; Klein, J., NGP1-01 is a brain-permeable dual blocker Sonsalla, P.K.; Castagnoli, K.; Castagnoli, N., Jr.; Schwarzschild,
of neuronal voltage- and ligand-operated calcium channels. M.A., Neuroprotection by caffeine and A(2A) adenosine receptor
Neurochem. Res., 2006, 31(3), 395-399. inactivation in a model of Parkinson's disease. J. Neurosci., 2001,
[24] Geldenhuys, W.J.; Malan, S.F.; Bloomquist, J.R.; Van der Schyf, 21(10), RC143.
C.J., Structure-activity relationships of pentacycloundecylamines at [44] Chen, J.F.; Steyn, S.; Staal, R.; Petzer, J.P.; Xu, K.; Van Der Schyf,
the N-methyl-D-aspartate receptor. Bioorg. Med. Chem., 2007, C.J.; Castagnoli, K.; Sonsalla, P.K.; Castagnoli, N., Jr.;
15(3), 1525-1532. Schwarzschild, M.A., 8-(3-Chlorostyryl)caffeine may attenuate
[25] Van der Schyf, C.J.; Squier, G.J.; Coetzee, W.A., Characterization MPTP neurotoxicity through dual actions of monoamine oxidase
of NGP 1-01, an aromatic polycyclic amine, as a calcium inhibition and A2A receptor antagonism. J. Biol. Chem., 2002,
antagonist. Pharmacol. Res. Commun., 1986, 18(5), 407-417. 277(39), 36040-36044.
[26] Malan, S.F.; Dyason, K.; Wagenaar, B.; Van Der Walt, J.J.; Van [45] Petzer, J.P.; Steyn, S.; Castagnoli, K.P.; Chen, J.F.; Schwarzschild,
Der Schyf, C.J., The structure and ion channel activity of 6- M.A.; Van der Schyf, C.J.; Castagnoli, N., Inhibition of
benzylamino-3-hydroxyhexa-cyclo[6.5.0.0(3,7).0(4,12).0(5,10).0 monoamine oxidase B by selective adenosine A2A receptor
(9,13)]tridecane. Arch. Pharm. (Weinheim), 2003, 336(2), 127-133. antagonists. Bioorg. Med. Chem., 2003, 11(7), 1299-1310.
[27] Malan, S.F.; Van der Walt, J.J.; Van der Schyf, C.J., Structure- [46] Muller, C.E.; Geis, U.; Hipp, J.; Schobert, U.; Frobenius, W.;
activity relationships of polycyclic aromatic amines with calcium Pawlowski, M.; Suzuki, F.; Sandoval-Ramirez, J., Synthesis and
channel blocking activity. Arch. Pharm. (Weinheim), 2000, 333(1), structure-activity relationships of 3,7-dimethyl-1-propargylxanthine
10-16. derivatives, A2A-selective adenosine receptor antagonists. J. Med.
[28] Kashiwagi, K.; Masuko, T.; Nguyen, C.D.; Kuno, T.; Tanaka, I.; Chem., 1997, 40(26), 4396-4405.
Igarashi, K.; Williams, K., Channel blockers acting at N-methyl-D- [47] Muller, C.E.; Sauer, R.; Geis, U.; Frobenius, W.; Talik, P.;
aspartate receptors: differential effects of mutations in the vestibule Pawlowski, M., Aza-analogs of 8-styrylxanthines as A2A-adenosine
and ion channel pore. Mol. Pharmacol., 2002, 61(3), 533-545. receptor antagonists. Arch. Pharm. (Weinheim), 1997, 330(6), 181-
[29] Hao, J.; Mdzinarishvili, A.; Abbruscato, T.J.; Klein, J.; 189.
Geldenhuys, W.J.; Van der Schyf, C.J.; Bickel, U., Neuroprotection [48] Pretorius, J.; Malan, S.F.; Castagnoli, N., Jr.; Bergh, J.J.; Petzer,
in mice by NGP1-01 after transient focal brain ischemia. Brain J.P., Dual inhibition of monoamine oxidase B and antagonism of
Res., 2008, 1196, 113-120. the adenosine A(2A) receptor by (E,E)-8-(4-phenylbutadien-1-
[30] Mdzinarishvili, A.; Geldenhuys, W.J.; Abbruscato, T.J.; Bickel, U.; yl)caffeine analogues. Bioorg. Med. Chem., 2008, 16(18), 8676-
Klein, J.; Van der Schyf, C.J., NGP1-01, a lipophilic polycyclic 8684.
cage amine, is neuroprotective in focal ischemia. Neurosci. Lett., [49] van den Berg, D.; Zoellner, K.R.; Ogunrombi, M.O.; Malan, S.F.;
2005, 383(1-2), 49-53. Terre'Blanche, G.; Castagnoli, N., Jr.; Bergh, J.J.; Petzer, J.P.,
1670 Current Medicinal Chemistry, 2013, Vol. 20, No. 13 Geldenhuys and Schyf

Inhibition of monoamine oxidase B by selected benzimidazole and [68] Kwok, J.B., Role of epigenetics in Alzheimer's and Parkinson's
caffeine analogues. Bioorg. Med. Chem., 2007, 15(11), 3692-3702. disease. Epigenomics, 2010, 2(5), 671-682.
[50] Milczek, E.M.; Binda, C.; Rovida, S.; Mattevi, A.; Edmondson, [69] Pithadia, A.S.; Lim, M.H., Metal-associated amyloid-beta species
D.E., The 'gating' residues Ile199 and Tyr326 in human in Alzheimer's disease. Curr. Opin. Chem. Biol., 2012, 16(1-2), 67-
monoamine oxidase B function in substrate and inhibitor 73.
recognition. FEBS J., 2011, 278(24), 4860-4869. [70] Guilarte, T.R., Manganese and Parkinson's disease: a critical
[51] Binda, C.; Mattevi, A.; Edmondson, D.E., Structural properties of review and new findings. Cien Saude Colet, 2011, 16, (11), 4549-
human monoamine oxidases A and B. Int. Rev. Neurobiol., 2011, 4566.
100, 1-11. [71] Lee, D.W.; Andersen, J.K., Iron elevations in the aging
[52] Culman, J.; Nguyen-Ngoc, M.; Glatz, T.; Gohlke, P.; Herdegen, T.; Parkinsonian brain: a consequence of impaired iron homeostasis? J.
Zhao, Y., Treatment of rats with pioglitazone in the reperfusion Neurochem., 2010, 112(2), 332-339.
phase of focal cerebral ischemia: A preclinical stroke trial. Exp. [72] Magaki, S.; Raghavan, R.; Mueller, C.; Oberg, K.C.; Vinters, H.V.;
Neurol., 2012, 238(2), 243-253. Kirsch, W.M., Iron, copper, and iron regulatory protein 2 in
[53] Culman, J.; Zhao, Y.; Gohlke, P.; Herdegen, T., PPAR-gamma: Alzheimer's disease and related dementias. Neurosci. Lett., 2007,
therapeutic target for ischemic stroke. Trends Pharmacol. Sci., 418(1), 72-76.
2007, 28(5), 244-249. [73] Hindo, S.S.; Mancino, A.M.; Braymer, J.J.; Liu, Y.; Vivekanandan,
[54] Carta, A.R.; Pisanu, A., Modulating Microglia Activity with S.; Ramamoorthy, A.; Lim, M.H., Small molecule modulators of
PPAR-gamma Agonists: A Promising Therapy for Parkinson's copper-induced Abeta aggregation. J. Am. Chem. Soc., 2009,
Disease? Neurotox. Res., 2013, 23(2), 112-123. 131(46), 16663-16665.
[55] Breidert, T.; Callebert, J.; Heneka, M.T.; Landreth, G.; Launay, [74] Kung, H.F.; Lee, C.W.; Zhuang, Z.P.; Kung, M.P.; Hou, C.; Plossl,
J.M.; Hirsch, E.C., Protective action of the peroxisome proliferator- K., Novel stilbenes as probes for amyloid plaques. J. Am. Chem.
activated receptor-gamma agonist pioglitazone in a mouse model of Soc., 2001, 123(50), 12740-12741.
Parkinson's disease. J. Neurochem., 2002, 82(3), 615-624. [75] Kung, M.P.; Hou, C.; Zhuang, Z.P.; Zhang, B.; Skovronsky, D.;
[56] Dehmer, T.; Heneka, M.T.; Sastre, M.; Dichgans, J.; Schulz, J.B., Trojanowski, J.Q.; Lee, V.M.; Kung, H.F., IMPY: an improved
Protection by pioglitazone in the MPTP model of Parkinson's thioflavin-T derivative for in vivo labeling of beta-amyloid plaques.
disease correlates with I kappa B alpha induction and block of NF Brain Res., 2002, 956(2), 202-210.
kappa B and iNOS activation. J. Neurochem., 2004, 88(2), 494- [76] Choi, J.S.; Braymer, J.J.; Nanga, R.P.; Ramamoorthy, A.; Lim,
501. M.H., Design of small molecules that target metal-A{beta} species
[57] Kumar, P.; Kaundal, R.K.; More, S.; Sharma, S.S., Beneficial and regulate metal-induced A{beta} aggregation and neurotoxicity.
effects of pioglitazone on cognitive impairment in MPTP model of Proc. Natl. Acad. Sci. U. S. A., 2010, 107(51), 21990-21995.
Parkinson's disease. Behav. Brain Res., 2009, 197(2), 398-403. [77] Yanez, M.; Fraiz, N.; Cano, E.; Orallo, F., Inhibitory effects of cis-
[58] Laloux, C.; Petrault, M.; Lecointe, C.; Devos, D.; Bordet, R., and trans-resveratrol on noradrenaline and 5-hydroxytryptamine
Differential susceptibility to the PPAR-gamma agonist pioglitazone uptake and on monoamine oxidase activity. Biochem. Biophys. Res.
in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and 6- Commun., 2006, 344(2), 688-695.
hydroxydopamine rodent models of Parkinson's disease. [78] Blanchet, J.; Longpre, F.; Bureau, G.; Morissette, M.; DiPaolo, T.;
Pharmacol. Res., 2012, 65(5), 514-522. Bronchti, G.; Martinoli, M.G., Resveratrol, a red wine polyphenol,
[59] Quinn, L.P.; Crook, B.; Hows, M.E.; Vidgeon-Hart, M.; Chapman, protects dopaminergic neurons in MPTP-treated mice. Prog.
H.; Upton, N.; Medhurst, A.D.; Virley, D.J., The PPARgamma Neuropsychopharmacol. Biol. Psychiatry, 2008, 32(5), 1243-1250.
agonist pioglitazone is effective in the MPTP mouse model of [79] Lu, K.T.; Ko, M.C.; Chen, B.Y.; Huang, J.C.; Hsieh, C.W.; Lee,
Parkinson's disease through inhibition of monoamine oxidase B. M.C.; Chiou, R.Y.; Wung, B.S.; Peng, C.H.; Yang, Y.L.,
Br. J. Pharmacol., 2008, 154(1), 226-233. Neuroprotective effects of resveratrol on MPTP-induced neuron
[60] Aviles-Olmos, I.; Limousin, P.; Lees, A.; Foltynie, T., Parkinson's loss mediated by free radical scavenging. J. Agric. Food Chem.,
disease, insulin resistance and novel agents of neuroprotection. 2008, 56(16), 6910-6913.
Brain, 2013, 136(Pt 2): 374-384. [80] Geldenhuys, W.J.; Ko, K.S.; Stinnett, H.; Van der Schyf, C.J.; Lim,
[61] Gillies, P.S.; Dunn, C.J., Pioglitazone. Drugs, 2000, 60(2), 333- M.H., Identification of multifunctional small molecule-based
343; discussion 344-335. reversible monoamine oxidase inhibitors. Med. Chem. Comm.,
[62] Hunter, R.L.; Dragicevic, N.; Seifert, K.; Choi, D.Y.; Liu, M.; Kim, 2011, 2, 1099-1103.
H.C.; Cass, W.A.; Sullivan, P.G.; Bing, G., Inflammation induces [81] Bolognesi, M.L.; Cavalli, A.; Valgimigli, L.; Bartolini, M.; Rosini,
mitochondrial dysfunction and dopaminergic neurodegeneration in M.; Andrisano, V.; Recanatini, M.; Melchiorre, C., Multi-target-
the nigrostriatal system. J. Neurochem., 2007, 100(5), 1375-1386. directed drug design strategy: from a dual binding site
[63] Akar, C.; Kalinin, S.; Gavrilyuk, V.; Spagnolo, A.; Weinber, G.; acetylcholinesterase inhibitor to a trifunctional compound against
Feinstein, D.L., Receptor-independent metabolic effects of Alzheimer's disease. J. Med. Chem., 2007, 50(26), 6446-6449.
thiazolidinediones in astrocytes. Open Cancer Immunol. J., 2010, [82] Bolognesi, M.L.; Banzi, R.; Bartolini, M.; Cavalli, A.; Tarozzi, A.;
3, 36-40. Andrisano, V.; Minarini, A.; Rosini, M.; Tumiatti, V.; Bergamini,
[64] Geldenhuys, W.J.; Darvesh, A.S.; Funk, M.O.; Van der Schyf, C.J.; C.; Fato, R.; Lenaz, G.; Hrelia, P.; Cattaneo, A.; Recanatini, M.;
Carroll, R.T., Identification of novel monoamine oxidase B Melchiorre, C., Novel class of quinone-bearing polyamines as
inhibitors by structure-based virtual screening. Bioorg. Med. Chem. multi-target-directed ligands to combat Alzheimer's disease. J.
Lett., 2010, 20(17), 5295-5298. Med. Chem., 2007, 50(20), 4882-4897.
[65] Cronet, P.; Petersen, J.F.; Folmer, R.; Blomberg, N.; Sjoblom, K.; [83] Bolognesi, M.L.; Cavalli, A.; Bergamini, C.; Fato, R.; Lenaz, G.;
Karlsson, U.; Lindstedt, E.L.; Bamberg, K., Structure of the Rosini, M.; Bartolini, M.; Andrisano, V.; Melchiorre, C., Toward a
PPARalpha and -gamma ligand binding domain in complex with rational design of multitarget-directed antioxidants: merging
AZ 242; ligand selectivity and agonist activation in the PPAR memoquin and lipoic acid molecular frameworks. J. Med. Chem.,
family. Structure, 2001, 9(8), 699-706. 2009, 52(23), 7883-7886.
[66] Montanari, R.; Saccoccia, F.; Scotti, E.; Crestani, M.; Godio, C.; [84] Bolognesi, M.L.; Matera, R.; Minarini, A.; Rosini, M.; Melchiorre,
Gilardi, F.; Loiodice, F.; Fracchiolla, G.; Laghezza, A.; Tortorella, C., Alzheimer's disease: new approaches to drug discovery. Curr.
P.; Lavecchia, A.; Novellino, E.; Mazza, F.; Aschi, M.; Pochetti, Opin. Chem. Biol., 2009, 13(3), 303-308.
G., Crystal structure of the peroxisome proliferator-activated [85] Bolognesi, M.L.; Rosini, M.; Andrisano, V.; Bartolini, M.;
receptor gamma (PPARgamma) ligand binding domain complexed Minarini, A.; Tumiatti, V.; Melchiorre, C., MTDL design strategy
with a novel partial agonist: a new region of the hydrophobic in the context of Alzheimer's disease: from lipocrine to memoquin
pocket could be exploited for drug design. J. Med. Chem., 2008, and beyond. Curr. Pharm. Des., 2009, 15(6), 601-613.
51(24), 7768-7776. [86] Bolognesi, M.L.; Cavalli, A.; Melchiorre, C., Memoquin: a multi-
[67] Gampe, R.T., Jr.; Montana, V.G.; Lambert, M.H.; Miller, A.B.; target-directed ligand as an innovative therapeutic opportunity for
Bledsoe, R.K.; Milburn, M.V.; Kliewer, S.A.; Willson, T.M.; Xu, Alzheimer's disease. Neurotherapeutics, 2009, 6(1), 152-162.
H.E., Asymmetry in the PPARgamma/RXRalpha crystal structure [87] Tumiatti, V.; Milelli, A.; Minarini, A.; Rosini, M.; Bolognesi,
reveals the molecular basis of heterodimerization among nuclear M.L.; Micco, M.; Andrisano, V.; Bartolini, M.; Mancini, F.;
receptors. Mol. Cell, 2000, 5(3), 545-555. Recanatini, M.; Cavalli, A.; Melchiorre, C., Structure-activity
Rationally Designed Multi-Targeted Agents Current Medicinal Chemistry, 2013, Vol. 20, No. 13 1671

relationships of acetylcholinesterase noncovalent inhibitors based [104] Jenner, P.; Langston, J.W., Explaining ADAGIO: a critical review
on a polyamine backbone. 4. Further investigation on the inner of the biological basis for the clinical effects of rasagiline. Mov.
spacer. J. Med. Chem., 2008, 51(22), 7308-7312. Disord., 2011, 26(13), 2316-2323.
[88] Bolognesi, M.L.; Minarini, A.; Rosini, M.; Tumiatti, V.; [105] Bar-Am, O.; Amit, T.; Weinreb, O.; Youdim, M.B.; Mandel, S.,
Melchiorre, C., From dual binding site acetylcholinesterase Propargylamine containing compounds as modulators of
inhibitors to multi-target-directed ligands (MTDLs): a step forward proteolytic cleavage of amyloid-beta protein precursor:
in the treatment of Alzheimer's disease. Mini Rev. Med. Chem., involvement of MAPK and PKC activation. J. Alzheimers Dis.,
2008, 8(10), 960-967. 2010, 21(2), 361-371.
[89] Bolognesi, M.L.; Calonghi, N.; Mangano, C.; Masotti, L.; [106] Bar-Am, O.; Weinreb, O.; Amit, T.; Youdim, M.B., Regulation of
Melchiorre, C., Parallel synthesis and cytotoxicity evaluation of a Bcl-2 family proteins, neurotrophic factors, and APP processing in
polyamine-quinone conjugates library. J. Med. Chem., 2008, the neurorescue activity of propargylamine. FASEB J., 2005,
51(17), 5463-5467. 19(13), 1899-1901.
[90] Rosini, M.; Simoni, E.; Bartolini, M.; Cavalli, A.; Ceccarini, L.; [107] Weinreb, O.; Amit, T.; Bar-Am, O.; Chillag-Talmor, O.; Youdim,
Pascu, N.; McClymont, D.W.; Tarozzi, A.; Bolognesi, M.L.; M.B., Novel neuroprotective mechanism of action of rasagiline is
Minarini, A.; Tumiatti, V.; Andrisano, V.; Mellor, I.R.; Melchiorre, associated with its propargyl moiety: interaction of Bcl-2 family
C., Inhibition of acetylcholinesterase, beta-amyloid aggregation, members with PKC pathway. Ann. N. Y. Acad. Sci., 2005, 1053,
and NMDA receptors in Alzheimer's disease: a promising direction 348-355.
for the multi-target-directed ligands gold rush. J. Med. Chem., [108] Weinreb, O.; Amit, T.; Bar-Am, O.; Sagi, Y.; Mandel, S.; Youdim,
2008, 51(15), 4381-4384. M.B., Involvement of multiple survival signal transduction
[91] Melchiorre, C.; Bolognesi, M.L.; Minarini, A.; Rosini, M.; pathways in the neuroprotective, neurorescue and APP processing
Tumiatti, V., Polyamines in drug discovery: from the universal activity of rasagiline and its propargyl moiety. J. Neural Transm.
template approach to the multitarget-directed ligand design Suppl., 2006, (70), 457-465.
strategy. J. Med. Chem., 2010, 53(16), 5906-5914. [109] Weinstock, M.; Bejar, C.; Wang, R.H.; Poltyrev, T.; Gross, A.;
[92] Bolognesi, M.L.; Bartolini, M.; Tarozzi, A.; Morroni, F.; Lizzi, F.; Finberg, J.P.; Youdim, M.B., TV3326, a novel neuroprotective
Milelli, A.; Minarini, A.; Rosini, M.; Hrelia, P.; Andrisano, V.; drug with cholinesterase and monoamine oxidase inhibitory
Melchiorre, C., Multitargeted drugs discovery: balancing anti- activities for the treatment of Alzheimer's disease. J. Neural.
amyloid and anticholinesterase capacity in a single chemical entity. Transm. Suppl., 2000, (60), 157-169.
Bioorg. Med. Chem. Lett., 2011, 21(9), 2655-2658. [110] Zhou, X.; Patel, A.R.; Perez, F.; Jurivich, D.A.,
[93] Bongarzone, S.; Tran, H.N.; Cavalli, A.; Roberti, M.; Carloni, P.; Acteylcholinesterase inhibitor rivastigmine enhances cellular
Legname, G.; Bolognesi, M.L., Parallel synthesis, evaluation, and defenses in neuronal and macrophage-like cell lines. Transl. Res.,
preliminary structure-activity relationship of 2,5-diamino-1,4- 2009, 153(3), 132-141.
benzoquinones as a novel class of bivalent anti-prion compound. J. [111] Bar-Am, O.; Amit, T.; Youdim, M.B., Aminoindan and
Med. Chem., 2010, 53(22), 8197-8201. hydroxyaminoindan, metabolites of rasagiline and ladostigil,
[94] Simoni, E.; Bergamini, C.; Fato, R.; Tarozzi, A.; Bains, S.; respectively, exert neuroprotective properties in vitro. J.
Motterlini, R.; Cavalli, A.; Bolognesi, M.L.; Minarini, A.; Hrelia, Neurochem., 2007, 103(2), 500-508.
P.; Lenaz, G.; Rosini, M.; Melchiorre, C., Polyamine conjugation [112] Bar-Am, O.; Yogev-Falach, M.; Amit, T.; Sagi, Y.; Youdim, M.B.,
of curcumin analogues toward the discovery of mitochondria- Regulation of protein kinase C by the anti-Parkinson drug, MAO-B
directed neuroprotective agents. J. Med. Chem., 2010, 53(19), inhibitor, rasagiline and its derivatives, in vivo. J. Neurochem.,
7264-7268. 2004, 89(5), 1119-1125.
[95] Rolinski, M.; Fox, C.; Maidment, I.; McShane, R., Cholinesterase [113] Youdim, M.B.; Amit, T.; Bar-Am, O.; Weinstock, M.; Yogev-
inhibitors for dementia with Lewy bodies, Parkinson's disease Falach, M., Amyloid processing and signal transduction properties
dementia and cognitive impairment in Parkinson's disease. of antiparkinson-antialzheimer neuroprotective drugs rasagiline and
Cochrane Database Syst Rev, 2012, 3, CD006504. TV3326. Ann. N. Y. Acad. Sci., 2003, 993, 378-386; discussion
[96] Salomone, S.; Caraci, F.; Leggio, G.M.; Fedotova, J.; Drago, F., 387-393.
New pharmacological strategies for treatment of Alzheimer's [114] Youdim, M.B.; Weinstock, M., Molecular basis of neuroprotective
disease: focus on disease modifying drugs. Br. J. Clin. Pharmacol., activities of rasagiline and the anti-Alzheimer drug TV3326 [(N-
2012, 73(4), 504-517. propargyl-(3R)aminoindan-5-YL)-ethyl methyl carbamate]. Cell.
[97] Hoy, S.M.; Keating, G.M., Rasagiline: a review of its use in the Mol. Neurobiol., 2001, 21(6), 555-573.
treatment of idiopathic Parkinson's disease. Drugs, 2012, 72(5), [115] Bruhlmann, C.; Ooms, F.; Carrupt, P.A.; Testa, B.; Catto, M.;
643-669. Leonetti, F.; Altomare, C.; Carotti, A., Coumarins derivatives as
[98] Zheng, H.; Amit, T.; Bar-Am, O.; Fridkin, M.; Youdim, M.B.; dual inhibitors of acetylcholinesterase and monoamine oxidase. J.
Mandel, S.A., From anti-Parkinson's drug rasagiline to novel Med. Chem., 2001, 44(19), 3195-3198.
multitarget iron chelators with acetylcholinesterase and monoamine [116] Bolea, I.; Juarez-Jimenez, J.; de Los Rios, C.; Chioua, M.;
oxidase inhibitory and neuroprotective properties for Alzheimer's Pouplana, R.; Luque, F.J.; Unzeta, M.; Marco-Contelles, J.;
disease. J. Alzheimers Dis., 2012, 30(1), 1-16. Samadi, A., Synthesis, biological evaluation, and molecular
[99] Weinreb, O.; Amit, T.; Riederer, P.; Youdim, M.B.; Mandel, S.A., modeling of donepezil and N-[(5-(benzyloxy)-1-methyl-1H-indol-
Neuroprotective profile of the multitarget drug rasagiline in 2-yl)methyl]-N-methylprop-2-yn-1-amine hybrids as new
Parkinson's disease. Int. Rev. Neurobiol., 2011, 100, 127-149. multipotent cholinesterase/monoamine oxidase inhibitors for the
[100] Weinreb, O.; Amit, T.; Bar-Am, O.; Youdim, M.B., Rasagiline: a treatment of Alzheimer's disease. J. Med. Chem., 2011, 54(24),
novel anti-Parkinsonian monoamine oxidase-B inhibitor with 8251-8270.
neuroprotective activity. Prog. Neurobiol., 2010, 92(3), 330-344. [117] Huang, L.; Lu, C.; Sun, Y.; Mao, F.; Luo, Z.; Su, T.; Jiang, H.;
[101] Youdim, M.B.; Gross, A.; Finberg, J.P., Rasagiline [N-propargyl- Shan, W.; Li, X., Multi-target-directed Benzylidene-indanone
1R(+)-aminoindan], a selective and potent inhibitor of derivatives: Anti-beta Amyloid (Abeta) aggregation, Antioxidant,
mitochondrial monoamine oxidase B. Br. J. Pharmacol., 2001, Metal chelation and Monoamine oxidase B (MAO-B) Inhibition
132(2), 500-506. Properties Against Alzheimer's disease. J. Med. Chem., 2012,
[102] Lew, M.F., The evidence for disease modification in Parkinson's 55(19), 8483-8492.
disease. Int. J. Neurosci., 2011, 121 Suppl 2, 18-26. [118] Kozikowski, A.P.; Chen, Y.; Subhasish, T.; Lewin, N.E.;
[103] Rascol, O.; Fitzer-Attas, C.J.; Hauser, R.; Jankovic, J.; Lang, A.; Blumberg, P.M.; Zhong, Z.; D'Annibale, M.A.; Wang, W.L.; Shen,
Langston, J.W.; Melamed, E.; Poewe, W.; Stocchi, F.; Tolosa, E.; Y.; Langley, B., Searching for disease modifiers-PKC activation
Eyal, E.; Weiss, Y.M.; Olanow, C.W., A double-blind, delayed- and HDAC inhibition - a dual drug approach to Alzheimer's disease
start trial of rasagiline in Parkinson's disease (the ADAGIO study): that decreases Abeta production while blocking oxidative stress.
prespecified and post-hoc analyses of the need for additional ChemMedChem, 2009, 4(7), 1095-1105.
therapies, changes in UPDRS scores, and non-motor outcomes. [119] Skovronsky, D.M.; Moore, D.B.; Milla, M.E.; Doms, R.W.; Lee,
Lancet Neurol., 2011, 10(5), 415-423. V.M., Protein kinase C-dependent alpha-secretase competes with
beta-secretase for cleavage of amyloid-beta precursor protein in the
trans-golgi network. J. Biol. Chem., 2000, 275(4), 2568-2575.
1672 Current Medicinal Chemistry, 2013, Vol. 20, No. 13 Geldenhuys and Schyf

[120] Jolly-Tornetta, C.; Wolf, B.A., Protein kinase C regulation of [122] Morphy, R.; Rankovic, Z., Designing multiple ligands - medicinal
intracellular and cell surface amyloid precursor protein (APP) chemistry strategies and challenges. Curr. Pharm. Des., 2009,
cleavage in CHO695 cells. Biochemistry, 2000, 39(49), 15282- 15(6), 587-600.
15290. [123] Braymer, J.J.; Detoma, A.S.; Choi, J.S.; Ko, K.S.; Lim, M.H.,
[121] Gray, S.G.; Dangond, F., Rationale for the use of histone Recent Development of Bifunctional Small Molecules to Study
deacetylase inhibitors as a dual therapeutic modality in multiple Metal-Amyloid-beta Species in Alzheimer's Disease. Int. J.
sclerosis. Epigenetics, 2006, 1(2), 67-75. Alzheimers Dis., 2010, 2011, 623051.

Received: January 10, 2012 Revised: November 02, 2012 Accepted: November 02, 2012

Das könnte Ihnen auch gefallen