[Molecular Medicine and Medicinal Chemistry] Alzheimer's Disease: Insights into Low Molecular Weight...

20
6 Inhibitor Design Against Cytotoxic β -Amyloid Species Andrew J. Doig 6.1 Introduction 6.1.1 Alzheimer’s disease and β -amyloid Alzheimer’s disease (AD) is characterized pathologically by abnormally high levels of brain lesions (senile plaques) and neurofibrillary tangles in dead and dying neurons, and by elevated numbers of amyloid deposits in the walls of cerebral blood vessels. The major component of senile plaques is a small peptide of 39–43 amino acids called β-amyloid (Aβ), which readily self-assembles into amyloid fibrils. Longer variants of Aβ are more prone to form amyloid. Aβ is produced through endoproteolysis of the amyloid precursor protein (APP). APP is first cleaved by β-secretase to give the membrane-bound C99 peptide and then by γ -secretase to give Aβ. Compelling evidence indicates that factors that increase the production of Aβ, particularly its more amyloidogenic variants, or that facilitate deposition or inhibit elimination of amyloid deposits cause AD or are risk factors for the disease (Selkoe, 1999). Manchester Interdisciplinary Biocentre, The University of Manchester, 131 Princess Street, Manchester M1 7DN, UK. Phone: +44 161 3064224; Fax: +44 161 3068918; Email: [email protected]. 187 Alzheimer's Disease: Insights into Low Molecular Weight and Cytotoxic Aggregates from In Vitro and Computer Experiments Downloaded from www.worldscientific.com by UNIVERSITY OF QUEENSLAND on 04/30/13. For personal use only.

Transcript of [Molecular Medicine and Medicinal Chemistry] Alzheimer's Disease: Insights into Low Molecular Weight...

November 26, 2012 10:1 9in x 6in Alzheimer’s Disease: Insights Into Low Molecular … b1377-chA6

6Inhibitor Design Against

Cytotoxic β-Amyloid Species

Andrew J. Doig∗

6.1 Introduction

6.1.1 Alzheimer’s disease and β-amyloid

Alzheimer’s disease (AD) is characterized pathologically by abnormallyhigh levels of brain lesions (senile plaques) and neurofibrillary tangles indead and dying neurons, and by elevated numbers of amyloid deposits inthe walls of cerebral blood vessels. The major component of senile plaquesis a small peptide of 39–43 amino acids called β-amyloid (Aβ), whichreadily self-assembles into amyloid fibrils. Longer variants of Aβ are moreprone to form amyloid. Aβ is produced through endoproteolysis of theamyloid precursor protein (APP). APP is first cleaved by β-secretase togive the membrane-bound C99 peptide and then by γ-secretase to give Aβ.Compelling evidence indicates that factors that increase the productionof Aβ, particularly its more amyloidogenic variants, or that facilitatedeposition or inhibit elimination of amyloid deposits cause AD or are riskfactors for the disease (Selkoe, 1999).

∗Manchester Interdisciplinary Biocentre, The University of Manchester, 131 Princess Street,Manchester M1 7DN, UK. Phone: +44 161 3064224; Fax: +44 161 3068918; Email:[email protected].

187

Alz

heim

er's

Dis

ease

: Ins

ight

s in

to L

ow M

olec

ular

Wei

ght a

nd C

ytot

oxic

Agg

rega

tes

from

In

Vitr

o an

d C

ompu

ter

Exp

erim

ents

Dow

nloa

ded

from

ww

w.w

orld

scie

ntif

ic.c

omby

UN

IVE

RSI

TY

OF

QU

EE

NSL

AN

D o

n 04

/30/

13. F

or p

erso

nal u

se o

nly.

November 26, 2012 10:1 9in x 6in Alzheimer’s Disease: Insights Into Low Molecular … b1377-chA6

188 A. J. Doig

6.1.2 β-Amyloid oligomers

Although Aβ aggregation clearly leads to Aβ toxicity, the structures ofaggregated Aβ were not initially studied closely, leading to the assumptionthat it was the readily visualized large fibrils that were toxic. Controversyraged over whether the fibrils are an epiphenomenon linked to disease orwhether fibril formation causes disease (Robinson and Bishop, 2002). Forexample, although many mutations in the Aβ precursor protein gene arelinked to premature onset of AD, the amount of amyloid deposited in thebrain does not correlate with severity. A resolution of this apparent paradoxfrom in vitro and in vivo evidence is that soluble, oligomeric forms of Aβ

have potent neurotoxic activity and are the primary cause of neuronal injuryand cell death occurring in AD (Lambert et al., 1998; Klein et al., 2001; Lesnéet al., 2006; Haass and Selkoe, 2007; Walsh and Selkoe, 2007). The primarytarget of an Aβ-based therapy should therefore be Aβ oligomers, ratherthan other less toxic or non-toxic species.

Evidence for the involvement of soluble, non-fibrillar Aβ in AD hasbeen obtained from synthetic Aβ peptides, cell culture systems that over-express APP, APP transgenic mice, and human cerebrospinal fluid andpostmortem brain (reviewed by Walsh and Selkoe, 2007). Sodium dodecylsulfate-stable dimers and trimers of Aβ were found in human cerebrospinalfluid (Vigo-Pelfrey et al., 1993) and postmortem brains (McLean et al.,1999). Lesné et al. (2006) used the Tg2576 transgenic mouse model tosearch for Aβ aggregates, the presence of which correlates with cognitivedecline, notably in the Morris water maze test, where performance in thesemice is notably different to wild-type from six months. They found thatnonamers and dodecamers first appear at this time and their levels correlatewith memory deficits. Injection of dodecamer into the ventricle of normalpretrained wild-type rats caused a dramatic fall-off in spatial memoryperformance, leading them to conclude that it is the dodecamer (Aβ56∗)that is the primary toxic species of Aβ (Lesné et al., 2006). 7PA2 Chinesehamster ovarian cells express mutant (V717F) human APP, and henceproduce and secrete low nanomolar amounts of Aβ (Podlisny et al., 1995).These have proved to be a valuable source of sodium dodecyl sulfate-stablelow-n Aβ oligomers that migrate in denaturing gels as dimers, trimers,and occasionally tetramers (Walsh et al., 2002). 7PA2 conditioned medium

Alz

heim

er's

Dis

ease

: Ins

ight

s in

to L

ow M

olec

ular

Wei

ght a

nd C

ytot

oxic

Agg

rega

tes

from

In

Vitr

o an

d C

ompu

ter

Exp

erim

ents

Dow

nloa

ded

from

ww

w.w

orld

scie

ntif

ic.c

omby

UN

IVE

RSI

TY

OF

QU

EE

NSL

AN

D o

n 04

/30/

13. F

or p

erso

nal u

se o

nly.

November 26, 2012 10:1 9in x 6in Alzheimer’s Disease: Insights Into Low Molecular … b1377-chA6

Inhibitor Design Against Cytotoxic β-Amyloid Species 189

inhibits hippocampal long-term potentiation (Walsh et al., 2005) andadversely affects performance in the alternating lever cyclic ratio test in rats(Cleary et al., 2005). The number of neuronal dendritic spines was dramati-cally decreased when neurons were grown in the presence of sub-nanomolarconcentrations of cell-derived Aβ oligomers (Shankar et al., 2007).

In this review, I briefly cover different strategies to treat AD, concen-trating on molecules able to inhibit Aβ aggregation and, ideally, affect thestructures or populations of Aβ oligomers. Such molecules can be foundin various ways, such as by screening libraries of compounds. Alternatively,knowledge of Aβ sequence and structure can be used to rationally designinhibitors (most often peptides).

6.2 Alternative Strategies to Treat Alzheimer’s Disease

A range of strategies are possible to prevent key steps within the amyloidcascade that leads to cell death, initiated by the key event of Aβ aggregationand oligomer formation. The main current approaches are briefly describedhere. As Aβ is produced by the cleavage of APP by β- and γ-secretase,inhibitors of either of these enzymes should prevent production ofAβ. Although numerous effective inhibitors of these targets have beendiscovered, there is concern that their inhibition would cause undesirableside-effects. For example, the deletion of β-secretase (β-site of APP-cleaving enzyme (BACE1)) in mice resulted in an increased death rate,reduced size, and alterations in the steady-state inactivation of voltage-gated sodium channels (Dominguez et al., 2005). BACE1 is also involvedwith the control of peripheral nerve myelination (Willem et al., 2006).Presenilin, the catalytic member of the γ-secretase proteolytic complex,has numerous substrates and roles in addition to cleaving APP, notably inreleasing the transcriptionally active domain of the receptor Notch (Parksand Curtis, 2007). A recent report, however, provides encouragementfor the γ-secretase-based strategy by identifying a γ-secretase activatingprotein that increases Aβ production without affecting Notch (He et al.,2010). Hence, inhibition of this protein may decrease Aβ levels without theunwanted side-effects of other γ-secretase inhibitors.

Immunization with Aβ showed exciting results in preventing the onsetof Aβ-related pathology in a transgenic mouse (Schenk et al., 1999),

Alz

heim

er's

Dis

ease

: Ins

ight

s in

to L

ow M

olec

ular

Wei

ght a

nd C

ytot

oxic

Agg

rega

tes

from

In

Vitr

o an

d C

ompu

ter

Exp

erim

ents

Dow

nloa

ded

from

ww

w.w

orld

scie

ntif

ic.c

omby

UN

IVE

RSI

TY

OF

QU

EE

NSL

AN

D o

n 04

/30/

13. F

or p

erso

nal u

se o

nly.

November 26, 2012 10:1 9in x 6in Alzheimer’s Disease: Insights Into Low Molecular … b1377-chA6

190 A. J. Doig

although a subsequent clinical trial had to be halted after a small number ofpatients developed meningoencephalitis (Nicoll et al., 2003). Nevertheless,vaccination remains a promising approach, perhaps using fragments ofAβ or other modifications. Activated microglia, along with a range ofinflammatory mediators,have been identified in association with the lesionsof AD, implying that anti-inflammatory agents such as non-steroidal anti-inflammatory drugs could protect against the disease, a hypothesis that issupported by epidemiological investigations (McGeer and McGeer, 2007).Although the major toxic species in AD is probably Aβ oligomers, and hencethese would be the principal target of an AD drug, it remains unclear why Aβ

oligomers should be toxic. Plausible mechanisms of toxicity include linksto oxidative stress, metal binding, free radical formation (Chauhan andChauhan, 2006), or ion channel formation (Quist et al., 2005), so it maybe possible to interfere with these processes, using reducing agents, metalchelators (Bush, 2002), or ion channel inhibitors. Using stem cells to replacelost neurons is a possibility for the future, although this approach wouldnot remove the toxic material. Current well-established AD drugs, suchas Donepezil (Aricept™), ENA-713 (Exelon™), Galantamine (Reminyl™),Memantine (Namenda™), and Tacrine (Cognex™) are acetylcholinesteraseinhibitors or N-methyl-D-aspartate receptor antagonists. They can onlyalleviate symptoms for a limited time.

Another strategy could be to activate α-secretase, which cleaves APPin the middle of the Aβ sequence. Several intracellular signalling pathwaysactivate α-secretase, such as muscarinic acetylcholine receptor stimulationand mitogen-activated protein (MAP) kinase activation. A disintegrin andmetalloproteinase (ADAM) enzymes are inactivated by tissue inhibitorsof metalloproteinases, and decreased levels or activity of specific tissueinhibitors of metalloproteinases may facilitate α-secretase cleavage (Postina,2008). Side-effects due to multiple roles of these enzymes and pathways areproblematic.

Numerous enzymes, including neprilysin, insulin-degrading enzyme,endothelin-converting enzyme, angiotensin-converting enzyme, matrixmetalloprotease-9, and plasmin are known to be involved in Aβ degradation(Howell et al., 1995; Qiu et al., 1998). Hence, strategies that enhance theactivity or protein levels of these proteases might be effective for thetreatment of AD. For example, the G-protein coupled receptor GPCR

Alz

heim

er's

Dis

ease

: Ins

ight

s in

to L

ow M

olec

ular

Wei

ght a

nd C

ytot

oxic

Agg

rega

tes

from

In

Vitr

o an

d C

ompu

ter

Exp

erim

ents

Dow

nloa

ded

from

ww

w.w

orld

scie

ntif

ic.c

omby

UN

IVE

RSI

TY

OF

QU

EE

NSL

AN

D o

n 04

/30/

13. F

or p

erso

nal u

se o

nly.

November 26, 2012 10:1 9in x 6in Alzheimer’s Disease: Insights Into Low Molecular … b1377-chA6

Inhibitor Design Against Cytotoxic β-Amyloid Species 191

receptor analogue, somatostatin, significantly increased neprilysin activityand hence treatment of primary neurons with somatostatin leads to areduction of Aβ levels (Saito et al., 2005).

In general, one would want to interfere with the first step of a pathwaythat leads to disease. Treatments designed to target steps that followprotein/peptide aggregation are less likely to be effective because they wouldnot prevent the formation of toxic soluble oligomers, which could continueto kill cells. An attractive therapeutic strategy in principle is therefore toinhibit peptide aggregation and oligomer formation, because this appearsto be the first step in the pathogenic process of amyloidosis that is notassociated with some natural biological function. Hence, the main focus ofthis chapter is on molecules designed to bind to cytotoxic Aβ species andinterfere with Aβ aggregation.

6.3 Inhibitors of β-Amyloid Toxicity

6.3.1 Peptides

Many reported inhibitors of Aβ aggregation and toxicity are peptides. Stud-ies in vitro on fragments of amyloidogenic proteins and synthetic peptideshave established that the tendency for a protein to form amyloid is oftenlimited to a short sequence of the full protein, known as a “self-recognitionelement” (SRE) (Madine et al., 2008; Tzotzos and Doig, 2010). SREs formthe core of amyloid fibrils. These amyloidogenic sequences constitute “hotspots” for aggregation of the native protein into amyloid fibrils and areoften the sites of mutations leading to early-onset amyloidosis. In the caseof tau paired helical filaments, which accumulate in the neurofibrillarytangles characteristic of AD and other neurodegenerative diseases, it hasbeen shown that only three residues, VYK, are sufficient for fibril formation(Goux et al., 2004). Short sequences forming the core domains of variousother amyloid fibrils have been identified, amongst which are KLVFFAE forAβ (Balbach et al., 2000), NFGSVQ for medin (Reches and Gazit, 2004),DFNKF for calcitonin (Reches et al., 2002), FGAIL and NFGAIL for isletamyloid polypeptide (Tenidis et al., 2000),VEALYL and LYQLEN for insulin(Sawaya et al., 2007), and VAQKTV for α-synuclein (Madine et al., 2008).It is often possible to delete residues freely on either side of an SRE whileretaining the ability to form amyloid. These SREs can be used as the basis

Alz

heim

er's

Dis

ease

: Ins

ight

s in

to L

ow M

olec

ular

Wei

ght a

nd C

ytot

oxic

Agg

rega

tes

from

In

Vitr

o an

d C

ompu

ter

Exp

erim

ents

Dow

nloa

ded

from

ww

w.w

orld

scie

ntif

ic.c

omby

UN

IVE

RSI

TY

OF

QU

EE

NSL

AN

D o

n 04

/30/

13. F

or p

erso

nal u

se o

nly.

November 26, 2012 10:1 9in x 6in Alzheimer’s Disease: Insights Into Low Molecular … b1377-chA6

192 A. J. Doig

Figure 6.1. Aβ self-recognition element Ac-KLVFF-NH2.

for the rational design of inhibitors of Aβ aggregation and toxicity as theyare already known to bind to the full-length target peptide.

Tjernberg et al. (1996) showed that Aβ(16-20), sequence KLVFF(Fig. 6.1), was able to bind full-length Aβ and prevent its assemblyinto fibrils. Using molecular graphics simulations they hypothesized thatit bound stereospecifically and in an antiparallel conformation to Aβ

(Tjernberg et al., 1997). Despite being shown to form fibrils in isolation,Aβ(16-20) was proposed as being a key region from which a lead compoundcould be created against amyloid, and indeed most groups have made useof this sequence as a starting point. Aβ(16-20) composed entirely of D-amino acids was as effective as its all-L-analogue with the additional benefitof being protease resistant (Tjernberg et al., 1996). Longer peptides thatretain the 16-20 region, namely 15-22, 16-23, and 17-24, can also inhibitAβ aggregation (Matsunaga et al., 2004). RIIGL, based on Aβ(31-34), whichhas the sequence IIGL, can also inhibit fibril formation by Aβ (Fülöpet al., 2004), so may also act as a starting point for modified peptideaggregation inhibitors. Similarly, GVVIA-NH2 and RVVIA-NH2, whichare based on Aβ(38-42), can inhibit Aβ aggregation and toxicity (Hetényiet al., 2002). Schwarzman et al. (2005) screened a combinatorial peptidelibrary for Aβ aggregation inhibition and found several novel peptidesequences that may be similarly useful. Their hits contained consensussequences ++Hy+(Hy/+), ++XX+, (D/E)LVXHy, or +LVLF, where + isa positively charged residue, Hy is a hydrophobic residue, and X is variable.

Alz

heim

er's

Dis

ease

: Ins

ight

s in

to L

ow M

olec

ular

Wei

ght a

nd C

ytot

oxic

Agg

rega

tes

from

In

Vitr

o an

d C

ompu

ter

Exp

erim

ents

Dow

nloa

ded

from

ww

w.w

orld

scie

ntif

ic.c

omby

UN

IVE

RSI

TY

OF

QU

EE

NSL

AN

D o

n 04

/30/

13. F

or p

erso

nal u

se o

nly.

November 26, 2012 10:1 9in x 6in Alzheimer’s Disease: Insights Into Low Molecular … b1377-chA6

Inhibitor Design Against Cytotoxic β-Amyloid Species 193

Key amyloidogenic regions often contain positively charged Lys or Arg, aswell as hydrophobic residues (Tzotzos and Doig, 2010), which is consistentwith these findings. If these amyloidogenic regions of Aβ could be modifiedto retain binding, but then prevent the association of further Aβ molecules,we would have an inhibitor of Aβ aggregation. A number of groups havefollowed this strategy, by modifying Aβ in various ways, as described below.

Soto et al. (1996) designed inhibitors aimed at the core region of Aβ,in this case residues 17-21. Their strategy is based upon substituting keyresidues for prolines in a bid to reduce the β-propensity. It is difficult tounderstand how preventing the adoption of the β conformation presentin aggregated Aβ could still allow binding. Nevertheless, a five-residuepeptide (LPFFD) with the ability to prevent fibril formation was found.All D-analogues were again found to be as effective, but with increasedprotease resistance. These so-called“β-sheet breakers”were then shown notonly to be stable in vivo, but to be small and hydrophobic enough to haveblood–brain barrier permeability (Soto et al., 1998; Poduslo et al., 1999).

Based initially around the 15-25 region of Aβ, Murphy and co-workers (Ghanta et al., 1996; Pallitto et al., 1999; Lowe et al., 2001)designed a peptide with a “recognition element” homologous to the Aβ

peptide, and a “disrupting element”, tagged to the C-terminus, designed tointerfere with the Aβ aggregation. For example, KLVFFKKKK acceleratedAβ aggregation kinetics, altered fibril morphology, and reduced toxicity.The anionic disrupting KLVFFEEEE had similar effects, whereas the neutraldisrupting KLVFFSSSS was ineffective, suggesting that the hydrophilicnature of the disrupting element is critical. Aβ aggregation need notbe blocked to prevent toxicity, as the compounds work by acceleratingaggregation to remove toxic soluble oligomers (Kim and Murphy, 2004).Moss et al. (2003) have disputed some of the results on KLVFFKKKKKK,however, as they found no reduction in Aβ toxicity, as measured by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT), eventhough the molecule did enhance protofibril association. Cairo et al. (2002)used binding to Aβ(10-35) monitored by surface plasmon resonance tomeasure the affinities of peptides to bind to Aβ. The peptides studied wererelated to KLVFF and some again had up to six charged residues at the C-terminus. No pentapeptides were found to be more effective than KLVFF,whereas the addition of up to six positively charged residues was found

Alz

heim

er's

Dis

ease

: Ins

ight

s in

to L

ow M

olec

ular

Wei

ght a

nd C

ytot

oxic

Agg

rega

tes

from

In

Vitr

o an

d C

ompu

ter

Exp

erim

ents

Dow

nloa

ded

from

ww

w.w

orld

scie

ntif

ic.c

omby

UN

IVE

RSI

TY

OF

QU

EE

NSL

AN

D o

n 04

/30/

13. F

or p

erso

nal u

se o

nly.

November 26, 2012 10:1 9in x 6in Alzheimer’s Disease: Insights Into Low Molecular … b1377-chA6

194 A. J. Doig

to favour binding. Converting some of the C-terminal Lys side-chains bymethylation to betaine [(CH3)3N+CH2COO−] increased activity,probablyby specific binding by KLVFF, increasing surface tension, and inducing Aβ

aggregation into a less toxic form (Kim et al., 2003). Further modificationsto the K6 sequence within the KLVFFKKKKKK inhibitor included branchedrather than linear lysine, L-lysine replaced by D-lysine, lysine replacedby diaminopropionic acid, and lysine replaced by arginine. A similarstrategy was employed by Burkoth et al. (1998) who attached a disruptingpolyethylene glycol block to the amyloidogenic fragment Aβ(10-35), givingan inhibitor of Aβ(10-35) fibrillogenesis.

Findeis et al. (1999) analysed many truncated variants of Aβ with a vari-ety of different N-terminal modifications, to establish a small, yet effective,inhibitor of Aβ polymerization. The strategy here is to retain a peptide thatcan bind to Aβ, but with a bulky group, such as a steroid, at its terminusto hinder further Aβ association. The all-D-amino acid peptide cholyl-LVFFA-OH was the most effective inhibitor of Aβ polymerization (Findeiset al., 2001), able to delay the onset of polymerization and the formation ofneurotoxic Aβ species by binding to Aβ within a fibrillar structure.

Several groups are studying the incorporation of N-methyl aminoacids into peptides as inhibitors of amyloidosis. These peptides againcorrespond to a region that is key to the amyloid protein. One side presentsa hydrogen-bonding “complementary” face to the protein, with the otherhaving N-methyl groups in place of backbone NH groups, thus presentinga “blocking” face. The N-methyl group also induces β-structure at theN-methylated site. Substituting a residue with an N-methyl group is similarto substituting with a Pro (above), as both will prevent backbone hydrogenbond donation. An N-methyl group is constrained to a β conformation,however, unlike Pro. Hughes et al. (2000) showed that N-methyl derivativesof Aβ(25-35), an aggregation-prone and toxic fragment of Aβ, were ableto prevent aggregation and inhibit toxicity in PC-12 cells. The location ofthe N-methyl group was critical, with some of the most effective inhibitorsaltering fibril morphology, rather than preventing Aβ(25-35) aggregation,as anticipated. Meredith and co-workers (Gordon et al., 2001; Gordon et al.,2002) investigated N-methylated peptides of a region corresponding to16-22 and later 16-20 of the amyloid “core domain” region. They preventedAβ fibrils from forming and broke down preformed fibrils. It is essential

Alz

heim

er's

Dis

ease

: Ins

ight

s in

to L

ow M

olec

ular

Wei

ght a

nd C

ytot

oxic

Agg

rega

tes

from

In

Vitr

o an

d C

ompu

ter

Exp

erim

ents

Dow

nloa

ded

from

ww

w.w

orld

scie

ntif

ic.c

omby

UN

IVE

RSI

TY

OF

QU

EE

NSL

AN

D o

n 04

/30/

13. F

or p

erso

nal u

se o

nly.

November 26, 2012 10:1 9in x 6in Alzheimer’s Disease: Insights Into Low Molecular … b1377-chA6

Inhibitor Design Against Cytotoxic β-Amyloid Species 195

that multiple N-methylations are on the same side of the peptide when ina β conformation, i.e. all at odd or all at even positions, so that the peptideretains an unblocked binding face. Although most of this work started withKLVFF, Cruz et al. (2004) used N-methylated inhibitors based on Aβ(36-40)(VGGVV). Kokkoni et al. (2006) optimized the structure of an inhibitorof Aβ aggregation and toxicity by starting with KLVFF and varying theside-chain identity, chirality,N-terminal acetylation,C-terminal amidation,and location of N-methyl groups. After four rounds of optimization,the most active compound was found to be D-[(chGly)-(Tyr)-(chGly)-(chGly)-(mLeu)]-NH2 (SEN304; Fig. 6.2), where every group had changedfrom the starting sequence, though retaining the pentapeptide framework.Inhibitors that had the N-methyl group replaced with an NH were highlyinsoluble and often toxic.

PPI-1019 D-(H-[(mL)VFFL]-NH2) is another effective N-methylatedpeptide inhibitor of Aβ aggregation and toxicity (Findeis, 2002). It isfundamentally different from other N-methylated peptides, however, asthe methylation of the unacetylated N-terminus means that an amineis methylated, rather than an amide, which would be the case at otherpositions. A singly methylated amine still has two hydrogen bond donors,so its mode of action may well be different to peptides that have methylatedamides, where hydrogen bond donation is eliminated. N-methylation ofthe N-terminal amine in Aβ(25-35) has no effect, for example, whereasN-methylation at other sites prevents the peptide being amyloidogenic(Hughes et al., 2000).

Figure 6.2. Peptidomimetic Aβ aggregation inhibitor SEN304.

Alz

heim

er's

Dis

ease

: Ins

ight

s in

to L

ow M

olec

ular

Wei

ght a

nd C

ytot

oxic

Agg

rega

tes

from

In

Vitr

o an

d C

ompu

ter

Exp

erim

ents

Dow

nloa

ded

from

ww

w.w

orld

scie

ntif

ic.c

omby

UN

IVE

RSI

TY

OF

QU

EE

NSL

AN

D o

n 04

/30/

13. F

or p

erso

nal u

se o

nly.

November 26, 2012 10:1 9in x 6in Alzheimer’s Disease: Insights Into Low Molecular … b1377-chA6

196 A. J. Doig

The peptide OR2 (RGKLVFFGR-NH2) is again based on KLVFF, butwith Arg-Gly sequences added to each terminus to improve solubility.This inhibited Aβ oligomer formation and reduced toxicity of Aβ inneuroblastoma SH-SY5Y cells (Austen et al., 2008). As a nine-residuepeptide is likely to be readily hydrolysed in vivo, the OR2 peptide wassubsequently converted to a retro-inverso peptide (RI-OR2), where all ofthe natural L-amino acids are replaced with the D-enantiomer, and thepeptide bonds are reversed (Taylor et al., 2010). This retains the side-chaingeometry of the parent peptide, but gains the beneficial effects of usingD-amino acids. RI-OR2 blocked the formation of Aβ oligomers and fibrils,bound to immobilized Aβ(1-42) monomers and fibrils, and reversed thetoxicity of Aβ(1-42) toward SH-SY5Y cells.

The tandem peptide construct Aβ40-1-(Gly)8-Aβ1-40 links two copiesof Aβ and inhibits Aβ fibrillization at low inhibitor : Aβ ratios (Mustafi et al.,2010), although it is doubtful whether such a large unmodified peptidewould be effective in vivo.

The dipeptide NH2-D-Trp-Aib-OH combines a Trp,as aromatic groupsare known to interact with Aβ, α-aminoisobutyric acid, which cannot formβ structure, and D-amino acids. It stabilizes non-toxic small oligomers andinhibits their growth into larger toxic structures. Most encouragingly, itreduces Aβ toxicity in cells, improves cognitive performance in mousemodels for AD, crosses the blood–brain barrier, and has acceptablepharmacokinetic properties (Frydman-Marom et al., 2009).

The peptide backbone can also be modified by replacing an amidegroup with an ester. This will remove the ability to hydrogen bond,but without adding any steric bulk or strongly affecting the backboneconformation, unlike N-substitution. KLVFF with an ester substitution wasalso able to inhibit the aggregation of Aβ and to disassemble preformed Aβ

fibrils (Gordon and Meredith, 2003). However, one would expect an estergroup to be rapidly hydrolysed in vivo.

6.3.2 Small molecules

Numerous organic compounds have been found to inhibit orreduce the aggregation and toxicity of Aβ in vitro. These includenicotine (Salomon et al., 1996; Moore et al., 2004), β-cyclodextrin(Camilleri et al., 1994), hemin and related porphyrins (Howlett

Alz

heim

er's

Dis

ease

: Ins

ight

s in

to L

ow M

olec

ular

Wei

ght a

nd C

ytot

oxic

Agg

rega

tes

from

In

Vitr

o an

d C

ompu

ter

Exp

erim

ents

Dow

nloa

ded

from

ww

w.w

orld

scie

ntif

ic.c

omby

UN

IVE

RSI

TY

OF

QU

EE

NSL

AN

D o

n 04

/30/

13. F

or p

erso

nal u

se o

nly.

November 26, 2012 10:1 9in x 6in Alzheimer’s Disease: Insights Into Low Molecular … b1377-chA6

Inhibitor Design Against Cytotoxic β-Amyloid Species 197

et al., 1997), anthracycline 4′-iodo-4′-deoxydoxorubicin (Merlini et al.,1995), hexadecyl-N-methylpiperidinium bromide (Wood et al., 1996),rifampicin (Tomiyama et al., 1996), (–)-5,8-dihydroxy-3R-methyl-2R-(dipropylamino)-1,2,3,4-tetrahydronapthalene (Parker et al., 2002), typeIV collagen, a molecule that localizes in senile plaques of patients with AD(Kiuchi et al., 2002), melatonin (Pappola et al., 1998), danuomycin (Howlettet al., 1999), glycosaminoglycans (McLaurin et al., 1999), fullerene (Kim andLee, 2003), 3-amino-1-propanesulfonic acid (Alzhemed or Tramiposate)(Aisen et al., 2006), apomorphine derivatives (Lashuel et al., 2002), 3-indole propionic acid (Bendheim et al., 2002), nordihydroguaiaretic acid,tannic acid, and quercetin (Matsuzaki et al., 2007), phenylazo benzenesul-fonamides (Lin et al., 2004), amphiphilic surfactants (Wang et al., 2005),curcumin (Yang et al., 2005), oligomeric acylated aminopyrazoles (Rzepeckiet al., 2004), cyclohexanehexol (inositol) stereoisomers (McLaurin et al.,2000; McLaurin et al., 2006), methylene blue (Necula et al., 2007),polyphenol (–)-epigallocatechin (Ehrnhoefer et al., 2008), and polyphenols(Porat et al., 2006). Salvianolic acid B directly inhibits PC12 cellular toxicityof aged Aβ (Tang and Zhang, 2001). Aβ aggregation can be induced byacetylcholinesterase, which can be inhibited by �9-tetrahydrocannabinol(Eubanks et al., 2006). Bifunctional inhibitors can be created by covalentlylinking an Aβ-binding molecule (Congo red) to a synthetic ligand forthe FK506 binding protein chaperone (Gestwicki et al., 2004). Oligomericacylated aminopyrazoles (e.g. Ampox, Fig. 6.3) with a donor–acceptor–donor hydrogen bond pattern complementary to that of a β-sheet areeffective inhibitors of Aβ aggregation, presumably by binding in the samemanner as a β-sheet peptide (Rzepecki et al., 2004). Caution should beexercised with some of these reports as they are often only based on a smallnumber of biophysical assays. For example, a molecule that interferes withthioflavin T fluorescence, a commonly used reporter for Aβ aggregation,may give artefactual results.

Figure 6.3. Ampox.

Alz

heim

er's

Dis

ease

: Ins

ight

s in

to L

ow M

olec

ular

Wei

ght a

nd C

ytot

oxic

Agg

rega

tes

from

In

Vitr

o an

d C

ompu

ter

Exp

erim

ents

Dow

nloa

ded

from

ww

w.w

orld

scie

ntif

ic.c

omby

UN

IVE

RSI

TY

OF

QU

EE

NSL

AN

D o

n 04

/30/

13. F

or p

erso

nal u

se o

nly.

November 26, 2012 10:1 9in x 6in Alzheimer’s Disease: Insights Into Low Molecular … b1377-chA6

198 A. J. Doig

The structural diversity in these molecules is remarkable, suggestingthat they can act in multiple ways and bind to Aβ at many different sites, incontrast to competitive enzyme inhibitors, for example, which bind to theenzyme active site as they resemble the substrate. For example, curcumindecreases Aβ levels by modulating APP processing, rather than interactingdirectly with Aβ oligomers. It increases the retention of immature APP inthe endoplasmic reticulum (ER) and attenuates APP endocytosis from theplasma membrane (Zhang et al., 2010). Few of these small molecule com-pounds are rationally designed using knowledge of the Aβ target sequenceand structure, as can be done more straightforwardly with peptides.

6.4 Conclusions

Although the rational design of compounds binding to Aβ is attractivein principle, this strategy has been mainly applied with peptides, withthe interesting exception of oligomeric acylated aminopyrazoles (Rzepeckiet al., 2004). Peptides generally make poor drugs, however, for severalreasons. Most drugs have properties that follow the Lipinski “rule-of-five”, namely five or fewer hydrogen bond donors, ten or fewer hydrogenbond acceptors, log P ≤ 5, and a molecular weight less than 500 Da.Most of the peptides or peptidomimetics described here do not meet thesecriteria. In addition, peptides are prone to degradation and do not crossmembranes, making them poor drugs. Some of the modifications discussedabove, however, do considerably improve the molecules’ properties, such asN-methylation, using all-D-peptides and unnatural side-chains.

Current results from drugs that have reached late-stage clinical tri-als continue to disappoint (http://www.alzforum.org/new/newssearch.asp?categoryID=22). New drugs for AD, perhaps using designs based on Aβ,as covered above, therefore continue to be an active and potentially veryvaluable research strategy for this massively important disease.

References

Aisen, P.S., Saumier, D., Briand, R., Laurin, J., Gervais, F., Tremblay, P. andGarceau, D. (2006). A Phase II study targeting amyloid-β with 3APS inmild-to-moderate Alzheimer disease, Neurology, 67, 1757–1763.

Alz

heim

er's

Dis

ease

: Ins

ight

s in

to L

ow M

olec

ular

Wei

ght a

nd C

ytot

oxic

Agg

rega

tes

from

In

Vitr

o an

d C

ompu

ter

Exp

erim

ents

Dow

nloa

ded

from

ww

w.w

orld

scie

ntif

ic.c

omby

UN

IVE

RSI

TY

OF

QU

EE

NSL

AN

D o

n 04

/30/

13. F

or p

erso

nal u

se o

nly.

November 26, 2012 10:1 9in x 6in Alzheimer’s Disease: Insights Into Low Molecular … b1377-chA6

Inhibitor Design Against Cytotoxic β-Amyloid Species 199

Austen, B.M., Paleologou, K.E., Ali, S.A.E., Qureshi, M.M., Allsop, D. and El-Agnaf,O.M.A. (2008). Designing peptide inhibitors for oligomerization and toxicityof Alzheimer’s β-amyloid peptide, Biochemistry, 47, 1984–1992.

Balbach, J.J., Ishii, Y., Antzutkin, O.N., Leapman, R.D., Rizzo, N.W., Dyda, F.,Reed, J. and Tycko, R. (2000). Amyloid fibril formation by Aβ 16-22, a seven-residue fragment of the Alzheimer’s beta-amyloid peptide, and structuralcharacterization by solid state NMR, Biochemistry, 39, 13748–13759.

Bendheim, P.E., Poeggeler, B., Neria, E., Ziv, V., Pappolla, M.A. and Chain, D.G.(2002). Development of indole-3-propionic acid (OXIGON) for Alzheimer’sdisease, J. Mol. Neurosci., 19, 213–217.

Burkoth, T.S., Benzinger, T.L.S., Jones, D.N.M., Hallenga, K., Meredith, S.C.and Lynn, D.G. (1998). C-terminal PEG blocks the irreversible step inβ-amyloid(10-35) fibrillogenesis., J. Am. Chem. Soc., 120, 7655–7656.

Bush, A.I. (2002). Metal complexing agents as therapies for Alzheimer’s disease,Neurobiol. Aging, 23, 1031–1038.

Cairo, C.W., Strzelec, A., Murphy, R.M., Kiessling, L.L. (2002). Affinity-basedinhibition of β-amyloid toxicity, Biochemistry, 41, 8620–8629.

Camilleri, P., Haskins, N.J. and Howlett, D.R. (1994). β-Cyclodextrin interacts withthe Alzheimer amyloid β-A4 peptide, FEBS Lett., 341, 256–258.

Chauhan, V. and Chauhan, A. (2006). Oxidative stress in Alzheimer’s disease,Pathophysiology, 13, 195–208.

Cleary, J.P., Walsh, D.M., Hofmeister, J.J., Shankar, G.M., Kuskowski, M.A.,Selkoe, D.J. and Ashe, K.H. (2005). Natural oligomers of the amyloid-betaprotein specifically disrupt cognitive function, Nat. Neurosci., 8, 79–84.

Cruz, M., Tusell, J.M., Grillo-Bosch, D., Albericio, F., Serratosa, J., Rabanal, F.and Giralt, E. (2004). Inhibition of β-amyloid toxicity by short peptidescontaining N-methyl amino acids, J. Pept. Res., 63, 324–328.

Dominguez, D., Tournoy, J., Hartmann, D., Huth, T., Cryns, K., Deforce, S.,Serneels, L., Camacho, I.E., Marjaux, E., Craessaerts, K., Roebroek, A.J.M.,Schwake, M., D’Hooge, R., Bach, P., Kalinke, U., Moechars, D., Alzheimer, C.,Reiss, K., Saftig, P. and De Strooper, B. (2005). Phenotypic and biochem-ical analyses of BACE1- and BACE2-deficient mice, J. Biol. Chem., 280,30797–30806.

Ehrnhoefer, D.E., Bieschke, J., Boeddrich, A., Herbst, M., Masino, L., Lurz, R., Enge-mann, S., Pastore,A. and Wanker, E.E. (2008). EGCG redirects amyloidogenicpolypeptides into unstructured, off-pathway oligomers, Nat. Struct. Mol.Biol., 15, 558–566.

Eubanks, L.M., Rogers, C.J., Beuscher, A.E., IV, Koob, G.F., Olson, A.J., Dickerson,T.J. and Janda,K.D. (2006). A molecular link between the active component ofmarijuanaandAlzheimer’sdiseasepathology,Mol.Pharmaceut.,3,773–777.

Findeis, M.A. (2002). Peptide inhibitors of β-amyloid aggregation, Curr. TopicsMed. Chem., 2, 417–423.

Alz

heim

er's

Dis

ease

: Ins

ight

s in

to L

ow M

olec

ular

Wei

ght a

nd C

ytot

oxic

Agg

rega

tes

from

In

Vitr

o an

d C

ompu

ter

Exp

erim

ents

Dow

nloa

ded

from

ww

w.w

orld

scie

ntif

ic.c

omby

UN

IVE

RSI

TY

OF

QU

EE

NSL

AN

D o

n 04

/30/

13. F

or p

erso

nal u

se o

nly.

November 26, 2012 10:1 9in x 6in Alzheimer’s Disease: Insights Into Low Molecular … b1377-chA6

200 A. J. Doig

Findeis, M.A., Musso, G.M., Arico-Muendel, C.C., Benjamin, H.W., Hundal, A.M.,Lee, J.-J., Chin, J., Kelley, M.,Wakefield, J., Hayward, N.J. and Molineaux, S.M.(1999). Modified-peptide inhibitors of amyloid β-peptide polymerization,Biochemistry, 38, 6791–6800.

Findeis, M.A., Lee, J.-D., Kelley, M., Wakefield, J.D., Zhang, M.-H., Chin, J.,Kubasek, W. and Molineaux, S.M. (2001). Characterization of cholyl-leu-val-phe-pheala-OH as an inhibitor of amyloid betapeptide polymerization,Amyloid, 8, 231–241.

Frydman-Marom, A., Rechter, M., Shefler, I., Bram, Y., Shalev, D.E. and Gazit, E.(2009). Cognitive-performance recovery of Alzheimer’s disease model miceby modulation of early soluble amyloidal assemblies, Angew. Chem., 121,2015–2020.

Fülöp, L., Zarándi, M., Datki, Z., Soós, K. and Penke, B. (2004). β-Amyloid-derivedpentapeptide RIIGLa inhibits Aβ1-42 aggregation and toxicity, Biochem.Biophys. Res. Commun., 324, 64–69.

Gestwicki, J.E., Crabtree, G.R. and Graef, I.A. (2004). Harnessing chaperones togenerate small-molecule inhibitors of amyloid β aggregation, Science, 306,865–869.

Ghanta, J., Shen, C.L., Kiessling, L.L. and Murphy, R.M. (1996). A strat-egy for designing inhibitors of β-amyloid toxicity, J. Biol. Chem., 271,29525–29528.

Gordon, D.J. and Meredith, S.C. (2003). Probing the role of backbone hydrogenbonding in β-amyloid fibrils with inhibitor peptides containing ester bondsat alternate positions, Biochemistry, 42, 475–485.

Gordon,D.J., Sciaretta,K.L. and Meredith,S.C. (2001). Inhibition of β-amyloid(40)fibrillogenesis and disassembly of β-amyloid(40) fibrils by short β-amyloidcogeners containing N-methyl amino acids at alternate residues, Biochem-istry, 40, 8237–8245.

Gordon, D.J., Tappe, R. and Meredith, S.C. (2002). Design and characterization of amembrane permeable N-methyl amino acid-containing peptide that inhibitsAβ(1-40) fibrillogenesis, J. Pept. Res., 60, 37–55.

Goux, W.J., Kopplin, L., Nguyen, A.D., Leak, K., Rutkofsky, M., Shanmuganandam,V.D., Sharma, D., Inouye, H. and Kirschner, D.A. (2004). The formation ofstraight and twisted filaments from short tau peptides, J. Biol. Chem., 279,26868–26875.

Haass, C. and Selkoe, D.J. (2007). Soluble protein oligomers in neurodegeneration:lessons from the Alzheimer’s amyloid β-peptide, Nat. Rev. Mol. Cell Biol., 8,101–112.

He, G., Luo, W., Li, P., Remmers, C.J., Netzer, W.J., Hendrick, J., Bettayeb, K.,Flajolet,M.,Gorelick,F.,Wennogle,L.P. and Greengard,P. (2010).γ-Secretaseactivating protein is a therapeutic target for Alzheimer’s disease, Nature, 467,95–98.

Alz

heim

er's

Dis

ease

: Ins

ight

s in

to L

ow M

olec

ular

Wei

ght a

nd C

ytot

oxic

Agg

rega

tes

from

In

Vitr

o an

d C

ompu

ter

Exp

erim

ents

Dow

nloa

ded

from

ww

w.w

orld

scie

ntif

ic.c

omby

UN

IVE

RSI

TY

OF

QU

EE

NSL

AN

D o

n 04

/30/

13. F

or p

erso

nal u

se o

nly.

November 26, 2012 10:1 9in x 6in Alzheimer’s Disease: Insights Into Low Molecular … b1377-chA6

Inhibitor Design Against Cytotoxic β-Amyloid Species 201

Hetényi, C., Szabó, Z., Klement, E., Datki, Z., Körtvélyesi, T., Zarándi, M. andPenke, B. (2002). Pentapeptide amides interfere with the aggregation ofβ-amyloid peptide of Alzheimer’s disease, Biochem. Biophys. Res. Commun.,292, 931–936.

Howell, S., Nalbantoglu, J. and Crine, P. (1995). Neutral endopeptidase canhydrolyze β-amyloid(1-40) but shows no effect on β-amyloid precursorprotein-metabolism, Peptides, 16, 647–652.

Howlett, D., Cutler, P., Heales, S. and Camilleri, P. (1997). Hemin and relatedporphyrins inhibit β-amyloid aggregation, FEBS Lett., 417, 249–251.

Howlett, D., George, A., Owen, D., Ward, R. and Markwell, R. (1999). Commonstructural features determine the effectiveness of carvedilol, daunomycinand rolitetracycline as inhibitors of Alzheimer beta-amyloid fibril formation,Biochem. J., 343, 419–423.

Hughes, E., Burke, R.M. and Doig, A.J. (2000). Inhibition of toxicity inβ-amyloid peptide fragment β(25-35) using N-methylated derivatives:a general strategy to prevent amyloid formation, J. Biol. Chem., 275,25109–25115.

Kim, J.E. and Lee, M. (2003). Fullerene inhibits β-amyloid peptide aggregation,Biochem. Biophys. Res. Commun., 303, 576–579.

Kim, J.R. and Murphy, R.M. (2004). Mechanism of accelerated assembly ofβ-amyloid filaments into fibrils by KLVFFK6, Biophys. J., 86, 3194–3203.

Kim, J.R., Gibson, T.J. and Murphy, R.M. (2003). Targeted control of kinetics ofβ-amyloid self-association by surface tension-modifying peptides, J. Biol.Chem., 278, 40730–40735.

Kiuchi, Y., Isobe, Y. and Fukushima, K. (2002). Type IV collagen prevents amyloidβ-protein fibril formation, Life Sci., 70, 1555–1564.

Klein, W.L., Krafft, G.A. and Finch, C.E. (2001). Targeting small Aβ oligomers:the solution to an Alzheimer’s disease conundrum? Trends Neurosci., 24,219–224.

Kokkoni, N., Stott, K., Amijee, H., Mason, J.M. and Doig, A.J. (2006). N-Methylatedpeptide inhibitors of β-amyloid aggregation and toxicity. Optimisation ofinhibitor structure, Biochemistry, 45, 9906–9918.

Lambert, M.P., Barlow, A.K., Chromy, B.A., Edwards, C., Freed, R., Liosatos, M.,Morgan, T.E., Rozovsky, I., Trommer, B., Viola, K.L., Wals, P., Zhang, C.,Finch, C.E., Krafft, G.A. and Klein, W.L. (1998). Diffusible, nonfibrillarligands derived from Aβ1-42 are potent central nervous system neurotoxins,Proc. Nat. Acad. Sci. U.S.A., 95, 6448–6453.

Lashuel, H.A., Hartley, D.M., Balakhaneh, D., Aggarwal, A., Teichberg, S. andCallaway, D.J.E. (2002). New class of inhibitors of amyloid-β fibril formation.Implications for the mechanism of pathogenesis in Alzheimer’s disease,J. Biol. Chem., 277, 42881–42890.

Alz

heim

er's

Dis

ease

: Ins

ight

s in

to L

ow M

olec

ular

Wei

ght a

nd C

ytot

oxic

Agg

rega

tes

from

In

Vitr

o an

d C

ompu

ter

Exp

erim

ents

Dow

nloa

ded

from

ww

w.w

orld

scie

ntif

ic.c

omby

UN

IVE

RSI

TY

OF

QU

EE

NSL

AN

D o

n 04

/30/

13. F

or p

erso

nal u

se o

nly.

November 26, 2012 10:1 9in x 6in Alzheimer’s Disease: Insights Into Low Molecular … b1377-chA6

202 A. J. Doig

Lesné, S., Ming, T.K., Kotilinek, L., Kayed, R., Glabe, C.G., Yang, A., Gallagher, M.and Ashe, K.H. (2006). A specific amyloid-β protein assembly in the brainimpairs memory, Nature, 440, 352–357.

Lin, S.-J., Shiao,Y.-J., Chi, C.-W. and Yang, L.-M. (2004). Aβ Aggregation inhibitors.Part 1: synthesis and biological activity of phenylazo benzenesulfonamides,Bioorg. Med. Chem. Lett., 14, 1173–1176.

Lowe, T.L., Strzelec, A., Kiessling, L.L. and Murphy, R.M. (2001). Structure–function relationships for inhibitors of β-amyloid toxicity containing therecognition sequence KLVFF, Biochemistry, 40, 7882–7889.

Madine, J., Doig, A.J. and Middleton, D.A. (2008). Design of an N-methylatedpeptide inhibitor of α-synuclein aggregation guided by solid-state NMR,J. Am. Chem. Soc., 130, 7873–7881.

Matsunaga, Y., Fujii, A., Awasthi, A., Yokotani, J., Takakura, T. and Yamada, T.(2004). Eight-residue Aβ peptides inhibit the aggregation and enzymaticactivity of Aβ42, Regulat. Peptides, 120, 227–236.

Matsuzaki, K., Noguch, T., Wakabayashi, M., Ikeda, K., Okada, T., Ohashi, Y.,Hoshino, M. and Naiki, H. (2007). Inhibitors of amyloid β-protein aggre-gation mediated by GM1-containing raft-like membranes, Biochim. Biophys.Acta Biomembr., 1768, 122–130.

McGeer, P.L. and McGeer, E.G. (2007). NSAIDs and Alzheimer disease: epidemio-logical, animal model and clinical studies, Neurobiol. Aging, 28, 639–647.

McLaurin, J., Franklin, T., Zhang, X., Deng, J. and Fraser, P.E. (1999). Interactions ofAlzheimer amyloid-beta peptides with glycosaminoglycans effects on fibrilnucleation and growth, Eur. J. Biochem., 266, 1101–1110.

McLaurin, J., Golomb, R., Jurewicz, A., Antel, J.P. and Fraser, P.E. (2000).Inositol stereoisomers stabilize an oligomeric aggregate of Alzheimer amyloidβ peptide and inhibit Aβ induced toxicity, J. Biol. Chem., 275, 18495–18502.

McLaurin, J., Kierstead, M.E., Brown, M.E., Hawkes, C.A., Lambermon, M.H.L.,Phinney, A.L., Darabie, A.A., Cousins, J.E., French, J.E., Lan, M.F., Chen,F., Wong, S.S.N., Mount, H.T.J., Fraser, P.E., Westaway, D. and George-Hyslop, P.S. (2006). Cyclohexanehexol inhibitors of Aβ aggregation preventand reverse Alzheimer phenotype in a mouse model, Nat. Med., 12, 801–808.

McLean, C.A., Cherny, R.A., Fraser, F.W., Fuller, S.J., Smith, M.J., Beyreuther, K.,Bush, A.I. and Masters, C.L. (1999). Soluble pool of Abeta amyloid as adeterminant of severity of neurodegeneration in Alzheimer’s disease, Ann.Neurol., 46, 860–866.

Merlini, G., Ascari, E., Amboldi, N., Belloti, V., Arbustini, E., Perfetti, V., Ferrari, M.,Zorzoli, I., Marinone, M.G. and Garini, P. (1995). Interaction of theanthracycline 4′-iodo-4′-deoxydoxorubicin with amyloid fibrils: inhibitionof amyloidogenesis, Proc. Nat. Acad. Sci. U.S.A., 92, 2959–2963.

Alz

heim

er's

Dis

ease

: Ins

ight

s in

to L

ow M

olec

ular

Wei

ght a

nd C

ytot

oxic

Agg

rega

tes

from

In

Vitr

o an

d C

ompu

ter

Exp

erim

ents

Dow

nloa

ded

from

ww

w.w

orld

scie

ntif

ic.c

omby

UN

IVE

RSI

TY

OF

QU

EE

NSL

AN

D o

n 04

/30/

13. F

or p

erso

nal u

se o

nly.

November 26, 2012 10:1 9in x 6in Alzheimer’s Disease: Insights Into Low Molecular … b1377-chA6

Inhibitor Design Against Cytotoxic β-Amyloid Species 203

Moore, S.A., Huckerby, T.N., Gibson, G.L., Fullwood, N.J., Turnbull, S., Tabner, B.J.,El-Agnaf, O.M.A. and Allsop, D. (2004). Both the D-(+) and L-(−) enan-tiomers of nicotine inhibit Aβ aggregation and cytotoxicity, Biochemistry, 43,819–826.

Moss, M.A., Nichols, M.R., Reed, D.K., Hoh, J.H. and Rosenberry, T.L. (2003).The peptide KLVFF-K(6) promotes β-amyloid(1-40) protofibril growth byassociation but does not alter protofibril effects on cellular reduction of3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT),Mol.Pharmacol., 64, 1160–1168.

Mustafi, S.M., Garai, K., Crick, S.L., Baban, B. and Frieden, C. (2010). Substoichio-metric inhibition of Aβ(1-40) aggregation by a tandem Aβ(40-1-Gly8-1-40)peptide, Biochem. Biophys. Res. Comm., 397, 509–512.

Necula, M., Breydo, L., Milton, S., Kayed, R., van der Veer, W.E., Tone, P. andGlabe, C.G. (2007). Methylene blue inhibits amyloid Aβ oligomerization bypromoting fibrillization, Biochemistry, 46, 8850–8860.

Nicoll, J.A.R., Wilkinson, D., Holmes, C., Steart, P., Markham, H. and Weller, R.O.(2003). Neuropathology of human Alzheimer disease after immunizationwith amyloid-β peptide: a case report, Nat. Med., 9, 448–452.

Pallitto, M.M., Ghanta, J., Heinzelman, P., Kiessling, L.L. and Murphy, R.M.(1999). Recognition sequence design for peptidyl modulators of β-amyloidaggregation and toxicity, Biochemistry, 38, 3570–3578.

Pappola, M., Bozner, P., Soto, C., Shao, H., Robakis, N.K., Zagorski, M.,Frangione, B. and Ghiso, J. (1998). Inhibition of Alzheimer β-fibrillogenesisby melatonin, J. Biol. Chem., 273, 7185–7188.

Parker, M.H., Chen, R., Conway, K.A., Lee, D.H., Luo, C., Boyd, R.E., Nortey,S.O., Ross, T.M., Scott, M.K. and Reitz, A.B. (2002). Synthesis of (–)-5,8-dihydroxy-3R-methyl-2R-(dipropylamino)-1,2,3,4-tetrahydronaphthalene:an inhibitor of β-amyloid(1-42) aggregation, Bioorg. Med. Chem., 10,3565–3569.

Parks, A.L. and Curtis, D. (2007). Presenilin diversifies its portfolio, Trends Genet.,23, 140–150.

Podlisny, M.B., Ostaszewski, B.L., Squazzo, S.L., Koo, E.H., Rydel, R.E., Teplow, D.B.and Selkoe, D.J. (1995). Aggregation of secreted amyloid β-protein into SDS-stable oligomers in cell culture, J. Biol. Chem., 270, 9564–9570.

Poduslo, J.F., Curran, G.L., Kumar, A., Frangione, B. and Soto, C. (1999). β-Sheetbreaker peptide inhibitor of Alzheimer’s amyloidogenesis with increasedblood–brain barrier permeability and resistance to proteolytic degradationin plasma, J. Neurobiol., 39, 371–382.

Porat, Y., Abramowitz, A. and Gazit, E. (2006). Inhibition of amyloid fibrilformation by polyphenols: structural similarity and aromatic interactionsas a common inhibition mechanism, Chem. Biol. Drug Des., 67, 27–37.

Postina, R. (2008). A closer look at α-secretase, Curr. Alzheimer Res., 5, 179–186.

Alz

heim

er's

Dis

ease

: Ins

ight

s in

to L

ow M

olec

ular

Wei

ght a

nd C

ytot

oxic

Agg

rega

tes

from

In

Vitr

o an

d C

ompu

ter

Exp

erim

ents

Dow

nloa

ded

from

ww

w.w

orld

scie

ntif

ic.c

omby

UN

IVE

RSI

TY

OF

QU

EE

NSL

AN

D o

n 04

/30/

13. F

or p

erso

nal u

se o

nly.

November 26, 2012 10:1 9in x 6in Alzheimer’s Disease: Insights Into Low Molecular … b1377-chA6

204 A. J. Doig

Qiu, W.Q., Walsh, D.M., Ye, Z., Vekrellis, K., Zhang, J.M., Podlisny, M.B.,Rosner, M.R., Safavi, A., Hersh, L.B. and Selkoe, D.J. (1998). Insulin-degrading enzyme regulates extracellular levels of amyloid β-protein bydegradation, J. Biol. Chem., 273, 32730–32738.

Quist,A.,Doudevski, I.,Lin,H.,Azimova,R.,Ng,D.,Frangione,B.,Kagan,B.,Ghiso,J. and Lal, R. (2005). Amyloid ion channels: a common structural link forprotein-misfolding disease, Proc. Nat. Acad. Sci. U.S.A., 102, 10427–10432.

Reches, M. and Gazit, E. (2004). Amyloidogenic hexapeptide fragment of medin:homology to functional islet amyloid polypeptide fragments, Amyloid, 11,81–89.

Reches, M., Porat,Y. and Gazit, E. (2002). Amyloid fibril formation by pentapeptideand tetrapeptide fragments of human calcitonin, J. Biol. Chem., 277,35475–35480.

Robinson, S. and Bishop, G. (2002). Aβ as a bioflocculant: implications for theamyloid hypothesis of Alzheimer’s disease, Neurobiol. Aging, 23, 1051–1072.

Rzepecki, P., Nagel-Steger, L., Feuerstein, S., Linne, U., Molt, O., Zadmard, R.,Aschermann, K., Wehner, M., Schrader, T. and Riesner, D. (2004). Preventionof Alzheimer’s disease-associated Aβ aggregation by rationally designednonpeptidic β-sheet ligands, J. Biol. Chem., 279, 47497–47505.

Saito, T., Iwata, N., Tsubuki, S., Takaki, Y., Takano, J., Huang, S.M., Suemoto, T.,Higuchi, M. and Saido, T.C. (2005). Somatostatin regulates brain amyloidbeta peptide Aβ(42) through modulation of proteolytic degradation, NatureMed., 11, 434–439.

Salomon, A.R., Marcinowski, K.J., Friedland, R.P. and Zagorski, M.G. (1996).Nicotine inhibits amyloid formation by the β-peptide, Biochemistry, 35,13568–13578.

Sawaya, M.R., Sambashivan, S., Nelson, R., Ivanova, M.I., Sievers, S.A., Apostol,M.I., Thompson, M.J., Balbirnie, M., Wiltzius, J.J., McFarlane, H.T., Madsen,A.O., Riekel, C. and Eisenberg, D. (2007). Atomic structures of amyloid cross-beta spines reveal varied steric zippers, Nature, 447, 453–457.

Schenk, D., Barbour, R., Dunn, W., Gordon, G., Grajeda, H., Guido, T., Hu, K.,Huang, J., Johnson-Wood, K., Khan, K., Kholodenko, D., Lee, M., Liao, Z.,Lieberburg, I., Motter, R., Mutter, L., Soriano, F., Shopp, G., Vasquez,N., Vandevert, C., Walker, S., Wogulis, M., Yednock, T., Games, D. andSeubert, P. (1999). Immunization with amyloid-β attenuates Alzheimer-disease-like pathology in the PDAPP mouse, Nature, 400, 173–177.

Schwarzman, A.L., Tsiper, M., Gregori, L., Goldgaber, D., Frakowiak, J., Mazur-Kolecka, B., Taraskina, A., Pchelina, S. and Vitek, M.P. (2005). Selection ofpeptides binding to the amyloid β-protein reveals potential inhibitors ofamyloid formation, Amyloid, 12, 199–209.

Selkoe, D.J. (1999). Translating cell biology into therapeutic advances inAlzheimer’s disease, Nature Supp., 399, A23–A31.

Alz

heim

er's

Dis

ease

: Ins

ight

s in

to L

ow M

olec

ular

Wei

ght a

nd C

ytot

oxic

Agg

rega

tes

from

In

Vitr

o an

d C

ompu

ter

Exp

erim

ents

Dow

nloa

ded

from

ww

w.w

orld

scie

ntif

ic.c

omby

UN

IVE

RSI

TY

OF

QU

EE

NSL

AN

D o

n 04

/30/

13. F

or p

erso

nal u

se o

nly.

November 26, 2012 10:1 9in x 6in Alzheimer’s Disease: Insights Into Low Molecular … b1377-chA6

Inhibitor Design Against Cytotoxic β-Amyloid Species 205

Shankar, G.M., Bloodgood, B.L., Townsend, M., Walsh, D.M., Selkoe, D.J. andSabatini, B.L. (2007). Natural oligomers of the Alzheimer amyloid-β proteininduce reversible synapse loss by modulating an NMDA-type glutamatereceptor-dependent signaling pathway, J. Neurosci., 27, 2866–2875.

Soto, C., Kindy, M.S., Baumann, M. and Frangione, B. (1996). Inhibition ofAlzheimer’s amyloidosis by peptides that prevent β-sheet conformation,Biochem. Biophys. Res. Comm., 226, 672–680.

Soto, C., Sigurdsson, E.M., Morelli, L., Kumar, R.A., Castaño, E.M. and Frangione,B. (1998). β-Sheet breaker peptides inhibit fibrillogenesis in a rat brainmodel of amyloidosis: implications for Alzheimer’s therapy, Nature Med., 4,822–826.

Tang, N.K. and Zhang, J.T. (2001). Salvianolic acid B inhibits fibril formationand neurotoxicity of amyloid β-protein in vitro, Acta Pharmacol. Sin., 22,380–384.

Taylor, M., Moore, S., Mayes, J., Parkin, E., Beeg, M., Canovi, M., Gobbi, M., Mann,D.M.A. and Allsop, D. (2010). Development of a proteolytically stable retro-inverso peptide inhibitor of β-amyloid oligomerization as a potential noveltreatment for Alzheimer’s disease, Biochemistry, 49, 3261–3272.

Tenidis, K., Waldner, M., Bernhagen, J., Fischle, W., Bergmann, M., Weber, M.,Merkle, M.L., Voelter, W., Brunner, H. and Kapurniotu, A. (2000). Identifi-cation of a penta- and hexapeptide of islet amyloid polypeptide (IAPP) withamyloidogenic and cytotoxic properties, J. Mol. Biol., 295, 1055–1071.

Tjernberg, L.O., Näslund, J., Lindqvist, F., Johansson, J., Karlström,A.R., Thyberg, J.,Terenius, L. and Nordstedt, C. (1996). Arrest of β-amyloid fibril formationby a pentapeptide ligand, J. Biol. Chem., 271, 8545–8548.

Tjernberg, L.O., Lilliehook, C., Callaway, D.J.E., Naslund, J., Hahne, S., Thyberg, J.,Terenius, L. and Nordstedt, C. (1997). Controlling amyloid β-peptide fibrilformation with protease-stable ligands, J. Biol. Chem., 272, 12601–12605.

Tomiyama, T., Shoji, A., Kataoka, K., Suwa, Y., Asano, S., Kaneko, H. and Endo, N.(1996). Inhibition of amyloid β protein aggregation and neurotoxicity byrifampicin. Its possible function as a hydroxyl radical scavenger, J. Biol.Chem., 271, 6839–6844.

Tzotzos, S. and Doig, A.J. (2010). Amyloidogenic sequences in native proteinstructures, Prot. Sci., 11, 327–348.

Vigo-Pelfrey, C., Lee, D., Keim, P.S., Lieberburg, I. and Schenk, D. (1993).Characterization of beta-amyloid peptide from human cerebrospinal fluid,J. Neurochem., 61, 1965–1968.

Walsh, D.M. and Selkoe, D.J. (2007). Aβ oligomers — a decade of discovery, J.Neurochem., 101, 1172–1184.

Walsh, D.M., Klyubin, I., Fadeeva, J.V., Rowan, M.J. and Selkoe, D.J. (2002).Amyloid-beta oligomers: their production, toxicity and therapeutic inhi-bition, Biochem. Soc. Trans., 30, 552–557.

Alz

heim

er's

Dis

ease

: Ins

ight

s in

to L

ow M

olec

ular

Wei

ght a

nd C

ytot

oxic

Agg

rega

tes

from

In

Vitr

o an

d C

ompu

ter

Exp

erim

ents

Dow

nloa

ded

from

ww

w.w

orld

scie

ntif

ic.c

omby

UN

IVE

RSI

TY

OF

QU

EE

NSL

AN

D o

n 04

/30/

13. F

or p

erso

nal u

se o

nly.

November 26, 2012 10:1 9in x 6in Alzheimer’s Disease: Insights Into Low Molecular … b1377-chA6

206 A. J. Doig

Walsh, D.M., Klyubin, I., Shankar, G.M., Townsend, M., Fadeeva, J.V., Betts, V.,Podlisny, M.B., Cleary, J.P., Ashe, K.H., Rowan, M.J. and Selkoe, D.J. (2005).The role of cell-derived oligomers of A beta in Alzheimer’s disease andavenues for therapeutic intervention, Biochem. Soc. Trans., 33, 1087–1090.

Wang, S.S.-S., Chen, Y.-T. and Chou, S.-W. (2005). Inhibition of amyloid fibrilformation of β-amyloid peptides via the amphiphilic surfactants, Biochim.Biophys. Acta, 1741, 307–313.

Willem, M., Garratt, A.N., Novak, B., Citron, M., Kaufmann, S., Rittger, A.,DeStrooper, B., Saftig, P., Birchmeier, C. and Haass, C. (2006). Controlof peripheral nerve myelination by the β-secretase BACE1, Science, 314,664–666.

Wood, S.J., MacKenzie, L., Maleef, B., Hurle, M.R. and Wetzel, R. (1996). Selectiveinhibition of Aβ fibril formation, J. Biol. Chem., 271, 4086–4092.

Yang, F., Lim, G.P., Begum, A.N., Ubeda, O.J., Simmons, M.R., Ambegaokar, S.S.,Chen, P.P., Kayed, R., Glabe, C.G., Frautschy|, S.A. and Cole, G.M. (2005).Curcumin inhibits formation of amyloid oligomers and fibrils, binds plaques,and reduces amyloid in vivo, J. Biol. Chem., 280, 5892–5901.

Zhang, C., Browne, A., Child, D. and Tanzi, R.E. (2010). Curcumin decreasesamyloid-β peptide levels by attenuating the maturation of amyloid-βprecursor protein, J. Biol. Chem., 285, 28472–28480.

Alz

heim

er's

Dis

ease

: Ins

ight

s in

to L

ow M

olec

ular

Wei

ght a

nd C

ytot

oxic

Agg

rega

tes

from

In

Vitr

o an

d C

ompu

ter

Exp

erim

ents

Dow

nloa

ded

from

ww

w.w

orld

scie

ntif

ic.c

omby

UN

IVE

RSI

TY

OF

QU

EE

NSL

AN

D o

n 04

/30/

13. F

or p

erso

nal u

se o

nly.