112
Journal of Alzheimer’s Disease 33 (2013) 547–658 DOI 10.3233/JAD-2012-121537 IOS Press 547 Review Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes Wolfgang Froestl , Andreas Muhs and Andrea Pfeifer AC Immune SA, EPFL, Lausanne, Switzerland Accepted 30 August 2012 Abstract. Cognitive enhancers (nootropics) are drugs to treat cognition deficits in patients suffering from Alzheimer’s disease, schizophrenia, stroke, attention deficit hyperactivity disorder, or aging. Cognition refers to a capacity for information processing, applying knowledge, and changing preferences. It involves memory, attention, executive functions, perception, language, and psychomotor functions. The term nootropics was coined in 1972 when memory enhancing properties of piracetam were observed in clinical trials. In the meantime, hundreds of drugs have been evaluated in clinical trials or in preclinical experiments. To classify the compounds, a concept is proposed assigning drugs to 19 categories according to their mechanism(s) of action, in particular drugs interacting with receptors, enzymes, ion channels, nerve growth factors, re-uptake transporters, antioxidants, metal chelators, and disease modifying drugs meaning small molecules, vaccines, and monoclonal antibodies interacting with amyloid- and tau. For drugs whose mechanism of action is not known, they are either classified according to structure, e.g., peptides, or their origin, e.g., natural products. This review covers the evolution of research in this field over the last 25 years. Keywords: Alzheimer’s disease, cognitive enhancers, donepezil, enzymes, galantamine, memory, rivastigmine 1. INTRODUCTION As of August 29, 2012, there are 26,677 entries in PubMed under the term cognitive enhancers, 26,680 entries under the term nootropic and 242 entries under the term cognition enhancers. Scifinder lists 5,105 references under the research topic nootropic, 535 references under the term cognitive enhancer, and 9,780 references for cognition enhancers. The Thom- son Reuters Pharma database lists 1,106 drugs as nootropic agents or cognition enhancers and gives zero results under the term cognitive enhancer. The term nootropics was coined by the father of piracetam Cor- neliu Giurgea in 1972/1973 [1, 2], NOOS = mind and TROPEIN = toward. Correspondence to: Dr. Wolfgang Froestl, AC Immune SA, EPFL Quartier de l’innovation building B, CH-1015 Lausanne, Switzerland. Tel.: +41 21 693 91 21; Fax: +41 21 693 91 20; E-mail: [email protected]. Nootropics are drugs to treat cognition deficits, which are most commonly found in patients suffering from Alzheimer’s disease (AD), schizophrenia, stroke, attention deficit hyperactivity disorder, or aging. Mark J. Millan and 24 eminent researchers [3] presented an excellent overview on cognitive dysfunction in psy- chiatric disorders in the February 2012 issue of Nature Reviews Drug Discovery and defined cognition as “a suite of interrelated conscious (and unconscious) men- tal activities, including pre-attentional sensory gating, attention, learning and memory, problem solving, plan- ning, reasoning and judgment, understanding, knowing and representing, creativity, intuition and insight, spon- taneous thought, introspection, as well as mental time travel, self awareness and meta cognition (thinking and knowledge about cognition)”. Since a first review in 1989 on “Families of Cogni- tion Enhancers” by Froestl and Maˆ ıtre [4], substantial progress has been made in the understanding of ISSN 1387-2877/13/$27.50 © 2013 – IOS Press and the authors. All rights reserved

Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

Embed Size (px)

Citation preview

Page 1: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

Journal of Alzheimer’s Disease 33 (2013) 547–658DOI 10.3233/JAD-2012-121537IOS Press

547

Review

Cognitive Enhancers (Nootropics). Part 2:Drugs Interacting with Enzymes

Wolfgang Froestl∗, Andreas Muhs and Andrea PfeiferAC Immune SA, EPFL, Lausanne, Switzerland

Accepted 30 August 2012

Abstract. Cognitive enhancers (nootropics) are drugs to treat cognition deficits in patients suffering from Alzheimer’s disease,schizophrenia, stroke, attention deficit hyperactivity disorder, or aging. Cognition refers to a capacity for information processing,applying knowledge, and changing preferences. It involves memory, attention, executive functions, perception, language, andpsychomotor functions. The term nootropics was coined in 1972 when memory enhancing properties of piracetam were observedin clinical trials. In the meantime, hundreds of drugs have been evaluated in clinical trials or in preclinical experiments. Toclassify the compounds, a concept is proposed assigning drugs to 19 categories according to their mechanism(s) of action, inparticular drugs interacting with receptors, enzymes, ion channels, nerve growth factors, re-uptake transporters, antioxidants,metal chelators, and disease modifying drugs meaning small molecules, vaccines, and monoclonal antibodies interacting withamyloid-� and tau. For drugs whose mechanism of action is not known, they are either classified according to structure, e.g.,peptides, or their origin, e.g., natural products. This review covers the evolution of research in this field over the last 25 years.

Keywords: Alzheimer’s disease, cognitive enhancers, donepezil, enzymes, galantamine, memory, rivastigmine

1. INTRODUCTION

As of August 29, 2012, there are 26,677 entries inPubMed under the term cognitive enhancers, 26,680entries under the term nootropic and 242 entries underthe term cognition enhancers. Scifinder lists 5,105references under the research topic nootropic, 535references under the term cognitive enhancer, and9,780 references for cognition enhancers. The Thom-son Reuters Pharma database lists 1,106 drugs asnootropic agents or cognition enhancers and gives zeroresults under the term cognitive enhancer. The termnootropics was coined by the father of piracetam Cor-neliu Giurgea in 1972/1973 [1, 2], NOOS = mind andTROPEIN = toward.

∗Correspondence to: Dr. Wolfgang Froestl, AC Immune SA,EPFL Quartier de l’innovation building B, CH-1015 Lausanne,Switzerland. Tel.: +41 21 693 91 21; Fax: +41 21 693 91 20; E-mail:[email protected].

Nootropics are drugs to treat cognition deficits,which are most commonly found in patients sufferingfrom Alzheimer’s disease (AD), schizophrenia, stroke,attention deficit hyperactivity disorder, or aging. MarkJ. Millan and 24 eminent researchers [3] presented anexcellent overview on cognitive dysfunction in psy-chiatric disorders in the February 2012 issue of NatureReviews Drug Discovery and defined cognition as “asuite of interrelated conscious (and unconscious) men-tal activities, including pre-attentional sensory gating,attention, learning and memory, problem solving, plan-ning, reasoning and judgment, understanding, knowingand representing, creativity, intuition and insight, spon-taneous thought, introspection, as well as mental timetravel, self awareness and meta cognition (thinking andknowledge about cognition)”.

Since a first review in 1989 on “Families of Cogni-tion Enhancers” by Froestl and Maıtre [4], substantialprogress has been made in the understanding of

ISSN 1387-2877/13/$27.50 © 2013 – IOS Press and the authors. All rights reserved

Page 2: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

548 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

the mechanism(s) of cognitive enhancers. Therefore,we propose a new classification to assign cognitionenhancing drugs to 19 categories:

1. Drugs interacting with Receptors (Part 1)2. Drugs interacting with Enzymes (Part 2)3. Drugs interacting with Cytokines (Part 3)4. Drugs interacting with Gene Expression

(Part 3)5. Drugs interacting with Heat Shock Proteins

(Part 3)6. Drugs interacting with Hormones (Part 3)7. Drugs interacting with Ion Channels (non-

Receptors) (Part 3)8. Drugs interacting with Nerve Growth Factors

(Part 3)9. Drugs interacting with Re-uptake Transporters

(Part 3)10. Drugs interacting with Transcription Factors

(Part 3)11. Antioxidants (Part 3)12. Metal Chelators (Part 3)13. Natural Products (Part 3)14. Nootropics (“Drugs without mechanism”)

(Part 3)15. Peptides (Part 3)16. Drugs preventing amyloid-� aggregation (Part 3)

16.1. Ligands interacting with amyloid-�(Part 3)

16.2. Inhibitors of serum amyloid P componentbinding (Part 3)

16.3. Vaccines against amyloid-� (Part 3)16.4. Antibodies against amyloid-� (Part 3)

17. Drugs interacting with tau (Part 3)17.1. Small molecules preventing tau aggrega-

tion (Part 3)17.2. Ligands interacting with tau (Part 3)17.3. Vaccines against tau (Part 3)17.4. Antibodies against tau (Part 3)

18. Stem Cells (Part 3)19. Miscellaneous (Part 3)

In Part 1, drugs interacting with receptors weredescribed [5]. In Part 2, we describe drugs interact-ing with enzymes and in Part 3, drugs interacting withtargets 3 to 10 and compounds and preparations ofcategories 11 to 19.

2. DRUGS INTERACTING WITH ENZYMES

Researchers have been investigating drugs inter-acting with a wide variety of enzymes in order to

identify valuable cognition enhancers. These enzymesare:

2.1 Drugs interacting with Acetyl- & Butyryl-cholinesterase (AChE & BChE)2.1.1. Drugs inhibiting AChE and other biolog-

ical targets2.1.1.1. Dual AChE inhibitors and

AChE receptor ligands2.1.1.2. Dual AChE and amyloid-�

inhibitors2.1.1.3. Dual AChE inhibitors and

antioxidants2.1.1.4. Dual AChE and �-secretase-1

or �-secretase inhibitors2.1.1.5. Dual AChE inhibitors and cal-

cium channel blockers2.1.1.6. Dual AChE inhibitors and

cannabinoid receptor antago-nists

2.1.1.7. Dual AChE and fatty acidamide hydrolase inhibitors

2.1.1.8. Dual AChE inhibitors and his-tamine H3 receptor antagonists

2.1.1.9. Dual AChE and monoamineoxidase inhibitors

2.1.1.10. Dual AChE inhibitors andmetal chelators

2.1.1.11. Dual AChE inhibitors and N-methyl-D-aspartic acid recep-tor channel blockers

2.1.1.12. Dual AChE inhibitors andplatelet activating factor antag-onists

2.1.1.13. Dual AChE and serotonin trans-porter inhibitors

2.1.1.14. Dual AChE and sigma receptorinhibitors

2.2 Drugs interacting with �-Secretase2.3 Drugs interacting with �-Secretase2.4 Drugs interacting with �-Secretase

2.4.1. �-Secretase Inhibitors2.4.2. �-Secretase Modulators2.4.3. Inhibitors of �-Secretase Activating

Protein2.4.4. Notch Pathway Inhibitors

2.5. Drugs interacting with �-Hexosaminidase2.6. Drugs interacting with 11�-Hydroxysteroid

Dehydrogenase2.7. Drugs interacting with Calpain2.8. Drugs interacting with Carbonic Anhydrase2.9. Drugs interacting with Caspases

Page 3: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 549

2.10. Drugs interacting with Catechol-O-methyl-transferase

2.11. Drugs interacting with Cathepsin2.12. Drugs interacting with Cholesterol 24S-

hydroxylase (CYP46A1)2.13. Drugs interacting with Cyclooxygenase2.14. Drugs interacting with D-Amino Acid

Oxidase2.15. Drugs interacting with Glutaminyl Cyclase2.16. Drugs interacting with Glyceraldehyde-3-

Phosphate Dehydrogenase2.17. Drugs interacting with Glycogen Synthase

Kinase-3�2.18. Drugs interacting with Guanylyl Cyclase2.19. Drugs interacting with Heme Oxygenase2.20. Drugs interacting with Histone Deacetylase2.21. Drugs interacting with HMG-CoA Reductase2.22. Drugs interacting with Insulin-degrading

Enzyme2.23. Drugs interacting with Kinases ( /=GSK-3� and

/= PKC)2.24. Drugs interacting with Kynurenine Mono-

Oxygenase and Kynurenine Transaminase II2.25. Drugs interacting with 5-Lipoxygenase2.26. Drugs interacting with Monoamine Oxidase2.27. Drugs modulating O-linked N-Acetylgluco-

saminidase2.28. Drugs interacting with Peptidyl-prolyl cis-trans

Isomerase D2.29. Drugs interacting with Phosphodiesterases2.30. Drugs interacting with Phospholipases A2 and

D22.31. Drugs interacting with Plasminogen Activator

Inhibitor2.32. Drugs interacting with Poly ADP-Ribose Poly-

merase2.33. Drugs interacting with Prolyl Endo Peptidase2.34. Drugs interacting with Prostaglandin D & E

Synthases2.35. Drugs interacting with Protein Kinase C2.36. Drugs interacting with Protein Tyrosine Phos-

phatase2.37. Drugs interacting with Rac1 GTPase2.38. Drugs interacting with Ras Farnesyl Trans-

ferase2.39. Drugs interacting with S-Adenosylhomo-

cysteine Hydrolase2.40. Drugs interacting with Sirtuin2.41. Drugs interacting with Steroid sulfatase2.42. Drugs interacting with Transglutaminase2.43. Drugs interacting with Ubiquitin Carboxyl-

terminal Hydroxylase (Usp14).

2.1. Drugs interacting with Acetyl- andButyrylcholinesterase

Since the publication of the cholinergic hypothe-sis of AD by Bartus et al. in 1982 [6], tremendousefforts have been undertaken to find either selectiveacetylcholine receptor agonists (described in Part 1)or acetylcholinesterase (AChE) inhibitors [7–15]. Thehistory of the cholinergic hypothesis was traced back[16]. In retrospect, the latter approach (i.e., AChEinhibitors) turned out to be more successful than the bigefforts to find selective acetylcholine receptor agonists.

Tacrine (tetrahydroaminoacridine, Cognex;Warner-Lambert, Pfizer) was approved by the Foodand Drug Administration (FDA) in 1993 and waslaunched in 1995 for the treatment of AD. It potentlyblocks butyrylcholinesterase (BChE) (IC50 = 47 nM)and AChE (IC50 = 190 nM) [17]. Tacrine has a shorthalf-life (2–3 hours) and must be administered 4times a day. The major problem is its severe, thoughreversible, hepatotoxicity [17, 18].

Donepezil (Aricept; E2020; co-developed by Eisaiand Pfizer, Fig. 1) was approved by the FDA in Novem-ber 1996 and was launched in the US in January1997 for the treatment of mild-to moderate AD. InOctober 2006, FDA approval for the treatment ofsevere AD was granted. It potently blocked AChE(IC50 = 23 nM), whereas blockade of BChE was 320times weaker (IC50 = 7,420 nM) [17]. The Japaneseinventors disclosed values of IC50 = 5.7 nM for AChEand IC50 = 7,140 nM for BChE (factor 1250) [19].Donepezil interacts with the active site of AChE and theperipheral anionic site of the enzyme [20]. Its long ter-minal disposition half-life of 70 hours supports once-daily administration [18]. Interestingly, the two enan-tiomers of donepezil racemize rapidly in vivo [21, 22].

The sales for donepezil reported by Eisai for 2011totaled USD 1.853 billion (Thomson Reuters Pharma,update of August 24, 2012).

2,301 publications on donepezil are listed inPubMed (as of August 29, 2012).

For the synthesis and medicinal chemistry ofdonepezil, see [19, 23], for an efficient process forlarge-scale synthesis of donepezil [24, 25], and formolecular modeling of donepezil [26, 27].

Many preclinical and mechanistic reviews have beenpublished [28–37]. Interestingly, donepezil eliminatedthe suppressive effects of amyloid-� (A�)42 on long-term potentiation (LTP) in rat hippocampal slices [38,39]. Synergistic effects of donepezil and the 5-HT4receptor agonist RS-67333 on the object recogni-tion were observed in mice [40]. Pharmacokinetic

Page 4: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

550 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

Fig. 1. Launched acetylcholinesterase inhibitors.

considerations for donepezil dosing strategies werepresented [41].

There is extensive literature available on clinical trialresults; see (in chronological order) [42–66]. Broaderconsiderations of higher doses of donepezil in thetreatment of mild, moderate, and severe AD were com-municated recently [67], in particular on the questionof the case of the 23 mg dose [68]. For the effectsof donepezil on levels of cholinesterases in the cere-brospinal fluid (CSF) of AD patients, see [69].

For safety and tolerability of donepezil, see [70–72],for pharmaco-economics [73–81].

Donepezil significantly improved abilities in dailylives of young adults with Down syndrome [82] andin female Down syndrome patients [83], whereas inchildren with Down syndrome of age 10–17 years,donepezil did not show any benefit versus placebo [84].

Donepezil was used for the treatment of patientswith Parkinson’s disease [85–87] and for the treatmentof memory impairment in multiple sclerosis patients[88].

Many galenical forms have been developed.Donepezil hydrochloride fine granule formulation waslaunched in Japan in September 2001. Donepezil rapid

oral disintegration tablet was launched in Japan in July2004 and in the US in June 2005. Donepezil oral jellyformulation was launched in Japan in December 2009.Donepezil oral sustained release high-dose (23 mg)tablet formulation was launched in the US in August2010. The US NDA donepezil once weekly transder-mal formulation in collaboration with Teikoku Seiyakuwas submitted in June 2010. A dry syrup formulationadministered by suspending the dry syrup in wateris in pre-registration in Japan. A fast dissolving oralfilm strip formulation in collaboration with AppliedPharma Research and Labtec is in clinical evaluation,as is donepezil oral rapid disintegration film in col-laboration with Kyukyu Pharmaceuticals. Donepeziltransdermal patch with Labtec, donepezil hydrochlo-ride transdermal patch with Nitto Denk and Kowa, andNAL-8812, a 3-day patch formulation co-developedwith NAL Pharmaceuticals are in preclinical evalua-tions, as is the Immediate Suspension encapsulation byALRISE Biosystems. NAL-8817 is an orally dissolv-ing film formulation of donepezil using the BIO-FXfast-onset oral cavity drug delivery system technologyin preclinical evaluation (Thomson Reuters Pharma,update of May 31, 2012).

Page 5: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 551

Deuterated donepezil is investigated by the com-pany Deuteria Pharmaceuticals for the potentialtreatment of AD (Thomson Reuters Pharma, updateof August 10, 2012).

Donepezil + memantine (ADS-8704, Arimenda;Adamas Pharmaceuticals), a once-daily fixed dosecombination extended release formulation, is currentlytested in Phase II clinical trials. In May 2012, the FDAapproved Phase III studies in an end-of-Phase II meet-ing. First clinical results were presented [89] (ThomsonReuters Pharma, update of July 5, 2012).

Rivastigmine (Exelon, SDZ-ENA-713; SDZ-212-713; ENA-713; Novartis, Fig. 1) is a potent BChEinhibitor (IC50 = 37 nM). It blocks AChE with anIC50 = 4,150 nM, a factor of 112 times less than BChE[17]. It forms a carbamoylated complex with the active-site serine inactivating it for about 10 hours producinga “pseudo-irreversible” inhibition. For an excellentreview on the neurobiology of BChE, see [90, 91].

Rivastigmine was launched for the treatment of mildto moderate AD in 2000 in the EU and the US and in2010 in Japan. It was approved for the treatment ofmild to moderately severe dementia associated withidiopathic Parkinson’s disease. Rivastigmine transder-mal patch was launched in the EU in 2007 and in Japanin 2011 in collaboration with the Japanese licensee OnoPharmaceuticals.

Sales for rivastigmine for 2011 were USD 1.067billion representing a 6% increase on 2010 (ThomsonReuters Pharma, update of August 24, 2012).

There are currently 1,181 publications on rivastig-mine listed in PubMed (as of August 29, 2012).

Several syntheses of rivastigmine were published[92–96].

Excellent papers on the preclinical characterizationof rivastigmine were communicated [97–102].

Extensive literature exists on the clinical trialresults of rivastigmine in AD (in chronological order)[103–112]. Rivastigmine at doses of up to 12 mg/dayhad useful efficacy in patients with mild-moderatedementia of the Alzheimer type. Reports from largerPhase III studies confirmed these findings. The b.i.d.administration is the more efficacious regimen and hadcomparable tolerability to the t.i.d. regimen.

Rivastigmine was evaluated for the treatment ofdementia in Parkinson’s disease [113–120]. Oralrivastigmine 3–12 mg/day for 24 weeks was signif-icantly more effective than placebo in amelioratingcognitive and functional decline, including attentionaldeficits, in patients with Parkinson’s disease demen-tia. Rivastigmine was also effective in patients withdementia with Lewy bodies. Benefits were seen on tests

of attention, working memory, and episodic secondarymemory [121]. Rivastigmine was successfully appliedfor the treatment of vascular dementia [122–125],for treatment of hallucinations in Creutzfeldt-Jakobdisease [126], and memory deficits induced by elec-troconvulsive therapy [127].

For safety and tolerability of rivastigmine, see [50,74, 128]; for pharmaco-economics [78, 80, 129].

Many reviews exist on results of clinical trialsusing the rivastigmine transdermal patch [130–154]describing enhanced tolerability and significantly lessside effects.

NAL-8822 (NAL Pharmaceuticals) is a one-daypatch formulation of rivastigmine using its Bio-D3transdermal drug delivery system technology in pre-clinical evaluation (Thomson Reuters Pharma, updateof May 31, 2012).

Galantamine (Razadyne, Reminyl, Nivalin;Janssen Pharmaceutica, part of Johnson & Johnson,Shire and Takeda under license from Sanochemia;Fig. 1) is an alkaloid extracted from the bulbs of theAmaryllidaceae family that include daffodils and thecommon snowdrop (Galanthea woronowii [155]). Itblocks AChE (E.C. 3.1.1.7) with an IC50 of 800 nMand BChE (E.C. 3.1.1.8) with an IC50 of 7,320 nM[17]. Galantamine was launched in 2001 for thetreatment of mild to moderate AD in Europe and theUS and in March 2011 in Japan. In 2005, Johnson& Johnson launched an extended release capsuleformulation.

The sales of galantamine in 2011 for Johnson &Johnson were USD 470 million, for Shire USD 40 mil-lion, and for Takeda USD 34 million (Thomson ReutersPharma, update of August 27, 2012).

There are currently 1,602 publications on galan-tamine (and galanthamine) listed in PubMed (as ofAugust 29, 2012).

To a small extent, galantamine is still produced fromsnowdrops [156, 157]. However, chemical syntheseshave taken over. A technical process for an efficientnine-step procedure, which affords (−)-galanthaminein 12% overall yield, was presented by Sanochemia[158]. The synthetic schemes from academic laborato-ries were not scaled up [159–163].

The pharmacological characterization of galan-tamine was described in depth [164–175]. Galan-tamine weakly inhibited hERG channels [176].

Interestingly, galantamine not only blocks AChEand BChE, but also acts as a positive allosteric mod-ulator of nicotinic acetylcholine receptors enhancingthe concentrations of acetylcholine in the brain by asecond mechanism [177–182].

Page 6: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

552 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

The pharmacokinetics of galantamine werereported [183–187].

Many publications described the clinical trial resultsof galanthamine in AD (in chronological order)[188–201 [1]]. Galantamine improved and stabilizedcognitive performance, activities of daily living, andbehavioral symptoms over the course of 6 months,and its efficacy and tolerability are comparable withthose of other AChE inhibitors (rivastigmine anddonepezil). Galantamine proved to be one of the stan-dard first-line medications for mild-to-moderate AD.For galantamine in Parkinson’s disease, see [201];in schizophrenic patients [202]; in vascular dementia[203].

For safety and tolerability, see [72]; for pharmaco-economics [78, 80, 204]. The historical developmentof galantamine as a clinically used drug was tracedback [205].

Johnson & Johnson developed and launched Raza-dyne ER (Reminyl XL), an extended release capsuleformulation, in April 2005 in the US. Patientstreated with the extended release formulation hadbetter overall Alzheimer’s Disease Assessment Scale(ADAS-cog/11) scores than the placebo group after sixmonths (Thomson Reuters Pharma, update of March21, 2012).

NAL-8801 (NAL Pharmaceuticals) is a one daypatch formulation of galantamine using the Bio-D3transdermal drug delivery system technology in pre-clinical evaluation (Thomson Reuters Pharma, updateof May 31, 2012).

Memogain (GLN-1062; Galantos Pharma) is a pro-drug, the benzoyl ester, of galantamine administered asan intranasal formulation. Memogain has a more than15 times higher bioavailability in the brain than thesame dose of galantamine. It is cleaved enzymaticallyto galantamine [206]. It is in Phase I since November2011 (Thomson Reuters Pharma, update of December28, 2011).

Huperzine A (Cerebra; Shanghai Institute of Mate-ria Medica; Fig. 1) is a natural Lycopodium alkaloidfound in extracts from Huperzia serrate [207]. It waslaunched in China for the treatment of AD in 1995. Thedrug is administered in low oral doses of 0.1 mg fourtimes a day. Huperzine A blocked AChE with an IC50of 82 nM and BChE with an IC50 of 74.43 �M [208,209]. (−)-Huperzine A is 35-fold more potent that (+)-Huperzine A (Ki = 6.2 nM and 210 nM, respectively[210]), corroborating earlier Ki values of 8 nM and300 nM [211]. Molecular modeling studies revealedthat huperzine A binds to the bottom of the bindingcavity of AChE with its ammonium group interacting

with Trp-84, Phe-330, Glu-199, and Asp-72 (catalyticsite) and to the opening of the gorge with its ammo-nium group interacting with Trp-279 (peripheral site)[212]. Tyr-337 is essential for inhibition of recombi-nant human AChE by huperzine A [213]. The X-raystructures of Torpedo californica AChE complexedwith (+)-huperzine A and (−)-huperzine B was pre-sented [214].

Currently huperzine A is produced in large scalefrom natural sources, in particular from Phlegmariu-rus squarrosus, a member of the Huperziaceae [215],although several chemical synthesis schemes wereworked out [208, 216–222]. A large-scale syntheticroute resulted from a joint venture by process chemistsof Shasun Pharma (UK and India), Rhodia Pharma(USA), and Debiopharm (Switzerland) [223].

There are currently 358 publications on huperzineA listed in PubMed (as of August 29, 2012).

Excellent reviews on the pharmacology ofhuperzine A have been published (in chronologicalorder) [224–230]. In particular, the neuroprotec-tive effects of huperzine A have been investigatedin great detail, e.g., protective effects against A�-associated neurotoxicity [231–235]. Huperzine A dosedependently increased secretory amyloid-� proteinprecursor (A�PP) release suggesting a modulatoryeffect of huperzine A on �-secretase activity [236,237]. These findings were confirmed independently[238, 239]. This mechanism seems to play viaWnt/�-catenin signaling. Chronic administration ofhuperzine A to double transgenic mice led to anincrease in ADAM10 and a decrease in �-secretase-1and A�PP695 protein levels [240]. Recently huperzineA was also evaluated in triple transgenic mice con-firming the improvement of cognitive performanceand the non-amyloid pathway activation [241].

Reviews on results of clinical trials of huperzine Awere published [242–244]. A Phase II clinical studyin 210 AD patients treated with either placebo, 200 �gbid, or 400 �g bid of huperzine A (70 patients per arm)showed no effect in the 200 �g group, but huperzineA 400 �g bid showed a 2.27 point improvement inADAS-cog at 11 weeks versus a 0.29 decline in theplacebo group (p = 0.001) [245] (Thomson ReutersPharma, update of February 20, 2012).

The company Xel Pharmaceuticals (Draper, Utah) isdeveloping the once-weekly sustained release transder-mal patch formulation XEL-001HP. This formulationis currently tested in a Phase I clinical trial inChina since September 2008. Xel Pharmaceuticalsis also investigating the sustained release topical gelformulation XEL-001HG in preclinical evaluation

Page 7: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 553

(both Thomson Reuters Pharma, update of March 14,2012).

Novartis chemists synthesized novel analogues ofhuperzine A. The program was terminated [246].

WIN-026 (INM-176, KR-WAP-026; WhanIn underlicense from Scigenic & Scigen Harvest) is an AChEinhibitor with antioxidant and anti-A� properties con-taining a 95% extract of Angelica gigas Nakai in thepre-registration phase [247]. In June 2008, a PhaseIII clinical trial for the treatment of AD was initi-ated, which was completed by May 2011. The resultswere submitted to the Korean FDA. Launch is expectedfor the second half of 2012. The memory ameliorat-ing effects of INM-176 in mice were described [248](Thomson Reuters Pharma, update of May 30, 2012).

Posiphen (the (+)-enantiomer of phenserine, QRPharma under license from TorreyPines Therapeutics,Fig. 2) is in Phase II clinical trials for the oral treat-ment of AD since June 2009. It was effective to lowerA� levels in cell cultures and in mice [249]. Interest-ingly, posiphen also blocked the expression of brain�-synuclein [250, 251]. Posiphen is a candidate drug tolower CSF A�PP, A� peptide, and tau levels in humans[252] (Thomson Reuters Pharma, update of July 25,2012).

Shen Er Yang (Changchun Huayang HighTechnology Co Ltd., Shanghai, Fig. 2) is atablet formulation of 1,2,3,4,5,6,7,8-octohydro-9-phenylacetamidoacridine, a dual cholinesteraseinhibitor in Phase II clinical trials since March 2011(Thomson Reuters Pharma, update of April 18, 2012).

Methanesulfonyl fluoride (SeneXta under licensefrom the University of Texas at El Paso, Fig. 2) is anAChE inhibitor in Phase I clinical trials for AD andpost-stroke cognitive impairment. A Phase II clinicaltrial for AD was planned for 2011. For the inhibitionof BChE by methanesulfonyl fluoride, see [253]; forthe inhibition of AChE [254]. Learning and memoryenhancing effects were described [255–258]. A reporton a double-blind, placebo-controlled study of safetyand efficacy in 5 men and 10 women (60–82 years) withMini-Mental State Examination (MMSE) scores of9–24, who had senile dementia of the Alzheimer type,was published. Methanesulfonyl fluoride improvedcognitive performance as measured by the MMSE,ADAS-cog, Global Deterioration Scale, and the Clin-ical Interview Based Impression of Change [259](Thomson Reuters Pharma, update of June 28, 2012).

There are currently many cholinesterase inhibitorsin preclinical evaluation (in alphabetical order):

Bisnorcymserine (QR Pharma, under license fromTorreyPines Therapeutics; Fig. 2) is a dual BChE and

A�PP inhibitor. An IND was cleared by the FDA inDecember 2010. The inhibition of serum BChE bybisnorcymserine was reported [260]. For the crystalstructure of the complex, see [261] (Thomson ReutersPharma, update of July 25, 2012).

Bis-(7)-tacrine (Mayo Foundation; Fig. 2) wasone of the first homodimers reported in the lit-erature with increased potency as AChE inhibitor(IC50 = 0.4 nM with significantly less affinity to BChE(IC50 = 390 nM, selectivity for AChE is a factor of 980)[262]. For a more detailed description of bis(7)-tacrine,see section 2.1.1.5. Dual AChE inhibitors and calciumchannel blockers.

FS-0311 (Shanghai Institute for Materia Medica;Fig. 2) is a bis-huperzine B derivative with similarpotency for the inhibition of AChE as donepezil buthigher potency than huperzines A and B. It antago-nized cognitive deficits induced by scopolamine or bytransient brain ischemia [263].

Huprines (tacrine-huperzine A hybrids, Fig. 2)were presented by scientists of the University ofAutonoma de Barcelona [264–269] and independentlyfrom the University of Hong Kong [270]. French sci-entists also significantly contributed to novel huprinederivatives [271–273].

The best characterized huprines are huprine-X(3-chloro-9-ethyl), huprine-Y (3-chloro-9-methyl,see Fig. 2), and huprine-Z (3-fluoro-9-methyl). Theyare very potent AChE inhibitors with Ki values of0.026 nM for huprine-X and 0.033 nM for huprineY versus 31 nM for tacrine, 1.1 nM for donepezil,and 4.6 nM for huperzine A [267]. Other scientistsmeasured IC50 values of 0.78 nM for (±)-huprine-Y and 4.58 nM for (±)-huprine-Z in human AChEpreparations [274]. (±)-Huprine-X is an agonist atM1 muscarinic acetylcholine receptors (Ki = 338 nM)and at M2 mAChRs (Ki = 4.66 �M) [275]. The bind-ing of huprine-X to Torpedo californica AChE wasresolved in an X-ray structure at 2.1 A resolution[276]. Huprine-X facilitated learning and memory inthe Morris water maze [277]. Chronic treatment ofTg2576 (A�PPswe) mice and of 3xTgAD mice with0.12 �mol/kg huprine-X i.p. for 21 days caused a sig-nificant reduction of insoluble A�40 by 40% (p < 0.05)in the hippocampus of 3xTgAD mice while showingno effect in A�PPswe mice [278]. Additional data onhuprine X in triple transgenic mice were disclosedrecently [241].

Another way to create huperzine A-tacrine hybridsis to link the primary amino group of huperzine A to theprimary amino group of tacrine with methylene groups[279]. The best compounds showed Ki values against

Page 8: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

554 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

Fig. 2. Acetylcholinesterase inhibitors in development or in preclinical evaluation.

human AChE of 6.4 nM versus 0.026 for huprine-X.A group of Spanish and Italian scientists describedhuprine-tacrine heterodimers [280, 281].

Dimerization of an inactive fragment of huperzineA led to bis-(12)-hupyridone (Hong Kong Univer-sity, Fig. 3) displaying an IC50 = 52 nM versus 115 nMfor huperzine A [282]. The tether length of 12-13

methylene groups is consistent with simultaneousbinding to both the catalytic and the peripheral siteof AChE (see Fig. 5). Bis-(12)-hupyridone may alsoact via �7 nicotinic acetylcholine receptors [283].

NP-0336 (Noscira) is a BChE inhibitor in preclinicalevaluation (Thomson Reuters Pharma, update of July19, 2011). The structure was not communicated.

Page 9: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 555

Fig. 3. Acetylcholinesterase inhibitors in preclinical evaluation II.

Fig. 4. Dual acetylcholinesterase and muscarinic M2 ACh receptor blocker.

SPH-1285 (Sanochemia Pharmazeutika, formerlyWaldheim Pharmazeutika; Fig. 3 shows SPH-1286)is a neuroprotective derivative of galantamine for thepotential treatment of glaucoma (Thomson ReutersPharma, update of January 3, 2012).

The most potent AChE inhibitor is the moleculesyn-1 (Fig. 3) with Kd of 77 fM synthesized at theScripps Research Institute linking a tacrine part viaclick chemistry to a phenanthridium unit. The tacrinepart is binding to the catalytic site (see Fig. 5), the

Page 10: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

556 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

Fig. 5. Indole-tacrine heterodimers binding simultaneously to thecatalytic and the peripheral anionic site of acetylcholinesterase[381]. With permission from ACS Publications.

phenanthridium piece is located at the peripheral site(see Fig. 5) [284].

Many new scaffolds for AChE inhibitors werediscovered recently, such as benzoxazolinones [285],indolizinoquinoline [286], isoindoline-1,3-diones[287], pyrrolo isoxazol benzoic acid derivatives [288],pyrrolo thiazoles [289], quinazolines [290], andquinoline derivatives [291] acting as AChE inhibitors.Also tacrin-ferulic acid-nitric oxide (NO) donortrihybrides were presented [292].

Excellent reviews describe quantitative structureactivity relationships (QSAR) [293, 294], virtualscreening techniques [295], and the use of support vec-tor machines for a classification of AChE inhibitors[296]. High throughput screening and molecularmodeling for novel AChE inhibitors was discussed[297].

The development of many AChE inhibitors wasterminated (in alphabetical order) of amiridin (ipi-dacrine hydrochloride; Nikken Chemicals [298–300]),AP-2238 (University of Bologna [301–303]), BZYX(Zhejiang University), CI-1002 (Parke-Davis, nowPfizer), CHF-2822 and CHF-2957 (Chiesi), EN-101(an orally active 20-base antisense oligonucleotidedirected against an AChE sequence; Amarin underlicense from Yissum [304]), eptastigmine (MF-201;

Mediolanum [305–309], ER-127528 (Eisai), F-3796(Pierre Fabre), FR-152558 (Fujisawa), galantamineliposomal (Sanochemia), ganstigmine (CHF-2819;Chiesi [310–315], Hoe-065 (Hoechst, now sanofi[316–318], HP-290 (Hoechst, now sanofi), icopezil(CP-118954; Pfizer [319]), JES-9501 (dehydroevo-diamine hydrochloride, DHED; Jeil Pharmaceuticalsin collaboration with Seoul Univ.), LB-1003 (LifelikeBiomatic), metrifonate (BAY-a-9826, ProMen; Bayer[320–324]; MF-268 and MF-8615 (Mediolanum),MHP-133 (Medical College of Georgia), mimopezil(DEBIO-9902, ZT-1; Debiopharm under license fromthe Shanghai Institute of Materia Medica; a once–dailyprodrug, a Schiff base of huperzine A with 2-hydroxy-3-methoxy-5-chloro-benzaldehyde [319], NP-7557(Nastech [325]), NXX-066 (Astra), P26 (Phy-topharm), P-10358, P-11012, P-11149, and P-11467(Hoechst Marion Roussel, now sanofi), phenserineand (-)-phenserine (Axonyx under license from NIH,Daewoong Pharmaceuticals [326–329], of controlledrelease and transdermal formulations of physostig-mine (Forest Laboratories, Lohmann Therapie Sys-teme [330–332], protexia (PEG-rBChE; PEGylatedrecombinant human butyryl-cholinesterase; PharmA-thene under license from Nexia Biotechnologies; aprotein produced in the milk of goats in a pegylatedformulation; the development as post-exposure therapyfor chemical nerve agent, currently in Phase I trials, isongoing [333, 334]), Ro-46-5934 (Roche; a dual AChEinhibitor and M2 muscarinic acetylcholine receptorantagonist), RS-1439 (Sankyo; a dual AChE and sero-tonin re-uptake inhibitor), S-9977 (Servier; a dualAChE and serotonin re-uptake inhibitor), SDZ-ENX-792 (Sandoz, now Novartis), SM-10888 (Sumitomo[335–337]), SPH-1359 (Sanochemia), stacofylline(S-9977; Servier [338–341]), suronacrine (HP-128;Hoechst, now sanofi [342, 343]), T-82 (SS Pharmaceu-tical, a subsidiary of Boehringer Ingelheim and Arena[344, 345]), tolserine (Axonyx under license fromNIH [346–349], UCB-11056 (UCB), velnacrine (HP-029; Hoechst, now sanofi [350, 351]), of zanapezil(TAK-147; Takeda [352, 353] and of zifrosilone(MDL-73745, Hoechst Marion Roussel, now sanofi[354–356]).

2.1.1. Drugs inhibiting AChE and otherbiological targets

Tremendous efforts have been undertaken to com-bine AChE inhibition and interaction with multipletargets thought to be responsible for AD pathogenesisto create “multi-target-directed ligands” [357–370].

Page 11: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 557

2.1.1.1. Dual AChE inhibitors and AChE receptorligands. Caproctamine (University of Bologna;Fig. 4) is a potent AChE inhibitor (Ki = 104 nM) anda competitive M2 muscarinic acetylcholine receptorantagonist (Kb = 410 nM) [371].

MHP-133 (Medical College of Georgia; Fig. 4) is anAChE inhibitor interacting with M1 muscarinic acetyl-choline, 5-HT4 and I2 imidazoline receptors [372,373].

Ro-46-5934 (Hoffmann-La Roche, Fig. 4) is a dualAChE inhibitor and M2 AChR antagonist for thepotential treatment of AD. The development was dis-continued [374].

An attempt to combine tacrine and xanomeline ledto hybride compounds displaying potent acetyl andBChE inhibition (pIC50’s of 8.2 and selectivity ratioof 1.0). The hybrids did not bind as agonists to theorthosteric muscarinic acetylcholine M1 receptor butas antagonists to an allosteric site of the M1 AChRthereby reducing the amount of acetylcholine [375].

2.1.1.2. Dual AChE and amyloid-β inhibitors. AChEcolocalizes with A� peptide deposits in the brainsof AD patients and promoted A� fibrillogenesis byforming stable AChE-A� complexes. AChE boundthrough its peripheral site to the A� nonamyloidogenicform and acted as a pathological chaperone inducinga conformational transition to the amyloidogenic form[376–380].

One approach is to design ligands binding to boththe catalytic as well as to the peripheral anionic site ofAChE inhibitors (Fig. 5) [381].

After first attempts [301, 302, 371, 382–384] animportant breakthrough was achieved with NP-61.

NP-61 (NP-0361; Noscira, previously Neu-ropharma) is a once-daily dual binding site AChE andA� secretion inhibitor currently in Phase I clinicaltrials for the treatment of AD. The structure wasnot disclosed, but is probably a combination of6-chloro-tacrine (binding to the catalytic binding site)linked via a methylene bridge from the primary amineto the amide nitrogen of indole propionamide bindingat the peripheral binding site [381]. Key biologicaldata on NP-61 were published: inhibition of AChE:IC50 = 20 pM, inhibition of BChE: IC50 = 2.9 nM,inhibition of A�40 aggregation with human AChE inthe thioflavin T test: IC50 = 2 �M, inhibition of A�40aggregation without AChE in the thioflavin T test:49% at 100 �M [385, 386]. NP-61 was able to reversecognitive impairment tested in the Morris water mazeand to reduce plaque load in cortex and hippocampusof 6 month-old human A�PP (Swedish mutation)

transgenic mice treated once daily by oral gavagefor three months (the doses were not communicated)(Thomson Reuters Pharma, update of May 10, 2012).

IDN-5706, a hyperforin derivative, decreased thecontent of AChE associated with different types of A�plaques in 7-month-old double A�PPSWE/PS1 trans-genic mice after treatment with IDN 5706 for 10 weeks[387]. See also Part 3, Chapter 13. Natural Products.

IQM-622 (Neuroscience Group and CSIC, Madrid;Fig. 6) significantly decreased the brain A� depositsafter treatment of A�PP/PS1 mice for four weeks. Itpromoted the degradation of intracellular A� in astro-cytes and protected against A� toxicity in culturedastrocytes and neurons (chemistry: [388]; biology:[389]).

This is an area of research with numerous novelcontributions presenting new piperidinium-type com-pounds [390], new piperidine derivatives [391],donepezil-type inhibitors [392], novel pyrano[3,2-c]quinoline-6-chlorotacrine hybrids [393], pyridinyli-dene hydrazones [394], novel bis(7)-tacrine derivatives[395], hybrid compounds combining donepezil andAP2238 [303], heterodimers of enantiopure huprineX or Y and donepezil-type derivatives [396],2,4-disubstituted pyrimidine derivatives [397, 398],novel triazole-containing berberine derivatives [399],huprine-tacrine heterodimers [400, 401], isaindigo-tone derivatives [402], and oxoisoaporphine-basedinhibitors [403]. Berberine derivatives act as multi-functional agents of antioxidant, inhibitors of AChE,BChE, and A� aggregation [404].

Bisnorcymserine (QR Pharma, under license fromTorreyPines Therapeutics; see Fig. 2) is a dual BChEand A�PP inhibitor.

2.1.1.3. Dual AChE inhibitors and antioxidants.Lipocrine (University of Bologna; Fig. 6) is a com-bination of 6-chlorotacrine with �-lipoic acid [405].It has been shown that �-lipoic acid is an universalantioxidant [406–408], which protected rat corticalneurons against cell death induced by A�. Lipocrineinhibited AChE with an IC50 of 0.25 nM and BChEwith 10.8 nM within 1 minute. It inhibited the aggre-gation of A� with an IC50 of 45 �M and showedsignificant antioxidant activity against formation ofreactive oxygen species in SH-SY5Y cells after treat-ment with tert.-butyl hydroperoxide at 5 �M (p < 0.01),10 �M (p < 0.001), and 50 �M (p < 0.001) [409].

Memoquin (University of Bologna; Fig. 6) is acombination of the polyamine amide caproctamine[371] showing AChE and M2 muscarinic ACh recep-tor blocking properties with a 1,4-benzoquinone

Page 12: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

558 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

Fig. 6. Dual acetylcholinesterase inhibitors with amyloid-� inhibitor, antioxidant, BACE-1 inhibitor, and calcium channel blocker activities.

functionality derived from coenzyme Q or idebenoneas a radical scavenger [366, 410, 411]. It inhibitedAChE with an IC50 of 1.55 nM, human AChE-inducedaggregation of A�40 with an IC50 of 28.3 �M andA�42 self-aggregation with an IC50 of 5.93 �M.NAD(P)H:quinone oxidoreductase 1 (NQO1) is theenzyme responsible for the regeneration and mainte-nance of the CoQ-reduced state. Memoquin is a goodsubstrate for NQO1 with a Km of 12.7 �M. In addition,memoquin also effectively blocked �-secretase-1 withan IC50 of 108 nM. The compound reduced the num-ber of A� plaques in transgenic AD11 mice after oraladministration and rescued behavioral deficits in anobject recognition test in transgenic AD11 mice [412].

Introduction of methyl groups in the �-position ofthe terminal benzylamine moieties (Fig. 6; Memoquin,R = Me, both in (R)-configuration) led to an improvedderivative (inhibition of AChE with IC50 of 0.5 nM;inhibition of human AChE-induced aggregation ofA�40 with IC50 of 9.34 �M [413, 414].

Combination of the 2,5-diamino-benzoquinonescaffold with curcumin derived substituents is anotherway to multitarget-directed ligands [415].

Dual-acting drugs with extremely potent AChEinhibiting activity and antioxidant properties were pre-sented [416]. Hybrides between 6,8-dichlorotacrineand melatonin showed an IC50 of 8 pM againsthuman AChE and potent peroxyl radical absorbance

Page 13: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 559

capacities of 2.5-fold the value of Trolox in the oxygenradical absorbance capacity assay using fluorescein(ORAC-FL). Full experimental details were pro-vided [417]. N-acylamino-phenothiazines are selectiveBChE inhibitors and protected neurons against exoge-nous and mitochondrial free radicals [418, 419]. Acombination of tacrine and ferulic acid was described[420] as were novel piperazine derivatives [421].Berberine derivatives act as multi-functional agents ofantioxidant, inhibitors of AChE, BChE, and A� aggre-gation [422].

2.1.1.4. Dual AChE and β-secretase-1 or γ-secretaseinhibitors. Memoquin (University of Bologna, Fig. 6)is a potent inhibitor of AChE with an IC50 of 1.55 nMand of �-secretase-1 with an IC50 of 108 nM (see pre-vious section 2.1.1.3.).

Coumarin derivatives are also dual human AChEand �-secretase-1 inhibitors [423]. Inhibitions ofAChE with IC50 of 180 nM and of �-secretase-1 withIC50 of 150 nM were measured.

Researchers from the Shanghai Institute of Mate-ria Medica presented a dual inhibitor of AChE:IC50 = 1.83 �M and of �-secretase-1: IC50 = 0.567 �M[424].

A tacrine-chromene derivative with cholinergic,antioxidant, A� reducing, and BACE1 inhibitingproperties showed the following values: AChE:IC50 = 75 nM, BChE: IC50 = 1 nM, �-secretase-1:IC50 = 2.8 �M [425]. Quinoxaline-based hybrid com-pounds with AChE, histamine H3 receptor, and�-secretase-1 inhibitory activities were described[426].

Galantamine-based hybrid molecules with AChE,BChE, and �-secretase inhibiting activities have beenpresented recently [427].

2.1.1.5. Dual AChE inhibitors and calcium chan-nel blockers. Bis(7)-tacrine (Mayo Foundation, seeFig. 2), first synthesized in 1996 [262], is a verypotent AChE inhibitor (IC50 = 0.4 nM) with signifi-cantly less affinity to BChE (IC50 = 390 nM, selectivityfor AChE by a factor of 980). Bis(7)-tacrine wasextensively characterized. It reversed AF64A-induceddeficits in navigational memory in rats [428]. It pro-tected against ischemic injury in primary culturedmouse astrocytes [429]. It reduced hydrogen peroxide-induced injury in rat pheochromocytoma cells [430]. Itshowed inhibitory action on N-methyl-D-aspartic acid(NMDA) receptors with an IC50 of 0.763 �M [431].Bis(7)-tacrine significantly reduced the augmentationof (L-type) high-voltage activated inward calcium

currents induced by A� [432]. For an excellent review,see [433].

ITH-4012 (TK-19; CSIC Madrid; Fig. 6) is anAChE inhibitor with an IC50 of 0.8 �M [434]. It causedan elevation of the cytosolic concentration of Ca2+in fura 2-loaded bovine chromaffine cells from con-centrations of 10 to 250 nM, which was related to theinduction of protein synthesis relevant for cell survival.This cytoprotective action of ITH-4012 was reversedby the protein synthesis inhibitor cycloheximide.

ITH-12118 (CSIC Madrid; Fig. 6), a tacripyrine,combines tacrine and a L-type voltage-dependent cal-cium channel blocker of the nimodipine type. Thesynthetic work was described in [435, 436] and theextensive biological characterization in [437] show-ing that ITH12118 could be a paradigmatic multitargetcompound having selective brain effects with smallerperipheral side effects. Additional chemical and phar-macological studies were published [438].

The development of NP-34 (or NP-04634; Noscira,formerly Neuropharma), a natural product derivedfrom the marine sponge Aplysina cavericola with cal-cium channel blocking and AChE inhibiting propertieswas terminated [439].

2.1.1.6. Dual AChE inhibitors and cannabinoid recep-tor antagonists. CB1 receptor antagonists increasedACh release in certain brain areas including corti-cal regions and the hippocampus [440]. The CB1antagonist scaffolds of diaryl-pyrazolines and diaryl-imidazoles [441, 442] were combined with tacrine.Compound 20 (Solvay, now Abbott, Fig. 7) displayedan IC50 of 316 nM for acetylcholinesterase and a Ki

of 48 nM for CB1 receptors. The Ki for CB2 receptorswas >1 �M [443].

2.1.1.7. Dual AChE & fatty acid amide hydro-lase inhibitors. In regions of A�-enriched neuriticplaques an increased activity of the enzyme fattyacid amide hydrolase (FAAH) was selectively demon-strated [444]. Inhibitors of FAAH may regulateendocannabinoid signaling and may counteract neu-roinflammation. First dual cholinesterase and FAAHinhibitors were synthesized at the University ofBologna [445]. One carbamate derivative (shown inFig. 7) displayed IC50’s of 50 nM for FAAH, 74.9 nMfor human AChE, and 1.57 nM for human BChE.

MIQ-001 (Meta-IQ Aps) is a fatty acid metabolisminhibitor for the potential treatment of AD. A PhaseI healthy volunteer study was performed with dosesfrom 6 to 100 mg/day starting in February 2010. Noside-effects were observed. Meta-IQ generated excel-

Page 14: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

560 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

Fig. 7. Dual acetylcholinesterase & fatty acid amide hydrolase inhibitor, dual acetylcholinesterase inhibitor & cannabinoid, and histamine H3receptor antagonists.

lent preclinical data showing efficacy in restoration ofshort and long term memory in an AD mouse model(Thomson Reuters Pharma, update of June 25, 2012).The structure was not communicated.

The development of AMR-109 (Amarin Corp.,ultra-pure eicosapentanoic acid), a modulator ofomega-3 fatty acids, was terminated. For the neu-roprotective effects of eicosapentenoic acid againstA� induced impairment of LTP and memory, see[446–448]. For a review on polyunsaturated farry acidsas cognitive enhancers see [449].

2.1.1.8. Dual AChE inhibitors and histamine H3 recep-tor antagonists. FUB833 (Freie University of Berlin;Fig. 7) is a potent AChE inhibitor (IC50 = 2.6 nM),BChE inhibitor (IC50 = 8.8 nM), hH3 receptor antag-onist (Ki = 0.33 nM) inhibiting also histamine N-methyltransferase (HMT with IC50 = 48 nM) [450].

University of Parma medicinal chemists describedpotent dual non imidazole histamine H3 receptor antag-onists and AChE inhibitors [451]. Quinoxaline-based

hybrid compounds with AChE, histamine H3 receptor,and �-secretase-1 inhibitory activities were described[452].

2.1.1.9. Dual AChE and monoamine oxidaseinhibitors. Increased monoamine oxidase (MAO) Bactivity was found in AD brains [453]. A combinationof AChE and of MAO B inhibitors in one moleculewas attempted already in 1996 [454, 455]. Thebreakthrough was achieved with Ladostigil.

Ladostigil (TV-3326; Avraham Pharmaceuticalsunder license from Yissum Research Development,a wholly owned company of the Hebrew Universityof Jerusalem; Fig. 8) combines the carbamate moietyof the BChE inhibitor rivastigmine with the propar-gylamine pharmacophore of the MAO-B inhibitorsselegiline and rasagiline. Ladostigil is more potentagainst BChE (IC50 = 1.7 �M) than against AChE(IC50 = 51 �M). It is a central nervous system (CNS)-selective inhibitor of MAO-A and MAO-B withlittle inhibition of liver and small intestine MAO.

Page 15: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 561

Fig. 8. Dual acetylcholinesterase and monoamine oxidase inhibitor or metal chelator properties.

Propargylamines are irreversible inhibitors of MAO,therefore the long-term daily administration results ina cumulative effect. 50 �mol/kg given daily for 7 and14 days inhibited both MAO-A and MAO-B by >60%.80% inhibition of the brain enzymes was achieved witha dose of 75 �mol/kg given once daily for 2 weeks[456]. Ladostigil was extensively characterized [357,457–472].

Ladostigil is currently in Phase IIb clinical trialsin 190 patients in Europe since December 2010 forthe oral treatment of AD. In February 2012 a PhaseII trial was initiated in Israel and Europe in patientssuffering from mild cognitive impairment (Thom-son Reuters Pharma, update of May 18, 2012). Seealso section 2.26. Drugs interacting with MonoamineOxidase.

Spanish medicinal chemists described potent dualcholinesterase and MAO inhibitors [473–475].

2.1.1.10. Dual AChE inhibitors and metal chelators.HLA20A (Technion Haifa; Fig. 8) is a combinationof an 8-hydroxy-quinoline metal chelator, which wascarbamoylated at the 8-hydroxy function to interactwith AChE as does rivastigmine [476, 477]. Inhibitionof AChE was measured with IC50’s of 0.5 �M and forBChE with 42.58 �M. Interaction with AChE readilycleaved the carbamoyl moity to release the chelatorreadily forming complexes with FeSO4 and CuSO4.The prochelator is nontoxic to human SH-SY5Y cellsat 25 or 50 �M. A recent review described the field ingreat detail [478].

Novel indanone derivatives were designed as AChEinhibitors and metal-chelating agents [479].

2.1.1.11. Dual AChE inhibitors and N-methyl-D-aspartic acid receptor channel blockers. Carbacrine(University of Bologna, Fig. 9), a potent AChEinhibitor (IC50 = 2.15 nM), and weak inhibitor ofBChE (IC50 = 296 nM), blocked AChE-induced A�aggregation to 58% at 100 �M and A� self-aggregation to 36% at 50 �M and inhibited NMDAreceptors with an IC50 of 0.74 �M. It is a noncompeti-tive open-channel blocker. In the ORAC test carbacrinewas a good antioxidant (IC50 = 0.07 �M), more potentthan trolox [480].

University of Jena researchers presented bivalent�-carbolines as dual AChE inhibitors and NMDAreceptor blockers such as compound 22b (Fig. 9):IC50 = 0.5 nM for AChE, 5.7 nM for BChE, and 1.4 �Mfor the NMDA receptor, respectively [481].

Bis(7)-tacrine (Mayo Foundation, Fig. 2, see sec-tion 2.1.1.5. Dual AChE inhibitors and calciumchannel blockers) inhibited NMDA receptors with anIC50 of 763 nM [431].

2.1.1.12. Dual AChE inhibitors & platelet acti-vating factor antagonists. PMS-777 (University ofParis 7-Denis Diderot and Shanghai Jiao TongUniversity; Fig. 10), synthesized in 1996 [482,483], is a tetrahydrofuran derivative, which inhibitedAChE (IC50 = 2.48 �M), BChE (IC50 = 4.47 �M), andacted as platelet factor antagonist (IC50 = 12.5 �M)[484–490]. It inhibited A�-induced human neurob-lastoma SH-SY5Y cell apoptosis in a concentrationdependent manner. It decreased reactive oxygenspecies up to 32%, NO production up to 5 times,and TNF� release by 33% (Thomson Reuters Pharma,update of June 20, 2012).

Page 16: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

562 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

Fig. 9. Dual acetylcholinesterase inhibitors and NMDA receptor blocker.

PMS-1339 (University of Paris 7-Denis Diderotand Shanghai Jiao Tong University; Fig. 10) is anovel piperazine derivative, which inhibited AChE(IC50 = 4.4 �M) and BChE (IC50 = 1.09 �M), A�aggregation (recombinant human AChE-induced:IC50 = 45.1 �M), and acted as platelet factor antago-nist (IC50 = 332 nM) [491] (Thomson Reuters Pharma,update of June 20, 2012).

2.1.1.13. Dual AChE and serotonin transporterinhibitors. RS-1259 (BGC-20-1259; BRG underlicense from Daiichi Sankyo; Fig. 10) is a dual inhibitorof AChE (IC50 = 101 nM) and of the serotonin trans-porter (IC50 = 42 nM). It significantly ameliorated theage-related short-term memory impairment at a dose of1 mg/kg in rats 24-25 months of age [492]. At 2 mg/kgRS-1259 was also effective in recovering from memorydeficits. For the design, synthesis and structure-activityrelationships see [493–495]. Its development was ter-minated.

2.1.1.14. Dual AChE and sigma receptor inhibitors.SP-04 (Samaritan Pharmaceuticals; Fig. 10) is aninhibitor of AChE and of sigma-1 receptors. For syn-thesis and biological evaluation, see [496] (ThomsonReuters Pharma, update of March 23, 2011).

The development of igmesine (JO-1784; Jouveinal,now Pfizer) was terminated. See Part 1, Chapter 1.28,Drugs interacting with Sigma receptors.

2.2. Drugs interacting with alpha-secretase

�-Secretase cleaves A�PP at the Lys16-Leu17bond within the A� sequence to generate the sol-uble N-terminal A�PP fragment (sA�PP�) and themembrane bound CTF83 (carboxy-terminal fragmentof 83 residues in length) thus precluding the forma-tion and subsequent deposition of A�. The activationof the �-secretase processing of A�PP as a thera-peutic approach in AD was reviewed [497–501]. Theactivation of �-secretase is controlled by the pro-tein phosphorylation signal transduction pathway ofprotein kinase C (PKC). A review on �-, �-, and�-secretases in AD was published [502].

Etazolate (EHT-0202; SQ-20009; Exonhit; Fig. 10)is an orally active phosphodiesterase (PDE) 4 inhibitorand anxiolytic GABAA receptor modulator. Etazolatedose-dependently increased the secreted levels of non-amyloidogenic sA�PP� in cortical neurons of ratsthus shifting A�PP processing toward the �-secretasepathway [503]. Etazolate improved the memory perfor-mance in aged rats [504]. The results of the first PhaseII 3-month, randomized, placebo-controlled, double-

Page 17: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 563

Fig. 10. Two dual acetylcholinesterase & platelet-activating factor, one acetylcholinesterase & serotonin transporter, one acetylcholinesteraseand sigma-1 receptor inhibitor(s) and an �-secretase activator.

blind study in AD patients were reported [505]. Thedrug was shown to be safe and well tolerated (ThomsonReuters Pharma, update of September 13, 2011).

Bryostatin-1 (Blanchette Rockefeller Neuro-sciences Institute) is a naturally occurring PKCactivator isolated from the Californian marine bry-ozoan Bugula neritina. Bryostatin-1 increased brainlevels of sA�PP� and reduced brain A�40 levels,which may be explained by activation of �-secretase[499, 506]. In February 2012, the Institute waspreparing to initiate clinical trials in neurologicaldisorders. Chemistry and biology of bryostatins wasreviewed [507] (Thomson Reuters Pharma, updateof February 24, 2012). See also section 2.35. Drugsinteracting with Protein Kinase C.

NP-17, NP-21, NPM-01, NPM-05B1, and NPM-05B2 (Noscira) are orally available �-secretaseactivators of marine origin for the potential treatmentof AD (Thomson Reuters Pharma, updates of July 27,2011). The structures were not communicated.

Many compounds shifted A�PP processing towardthe �-secretase pathway in in vitro experiments, such

as muscarinic acetylcholine receptor agonists, MAO-B inhibitors, statins, non-steroidal anti-inflammatorydrugs (NSAIDs), neuropeptides such as PACAP,estrogen, and others [499]. However, the benefit forAD patients in clinical trials was (so far) disappointing.

2.3. Drugs interacting with beta secretase

In the amyloidogenic pathway, A�PP is firstcleaved by �-secretase (BACE1, EC 3.4.23.46 [508,509]) between residue methionine 671 and asparticacid 672 to release a 100 kDa N-terminal fragmentsA�PP and a 12 kDa membrane bound CFT99,which is subsequently cleaved by �-secretase withinthe transmembrane region to form C-terminal A�peptides ranging from 38 to 43 residues. �-secretasewas cloned simultaneously by five groups in 1999 andearly 2000: at Amgen [510, 511]; at Elan [512], atPharmacia & Upjohn, now Pfizer [513], at GSK [514],and at the Oklahoma Medical Research Foundation[515]. It is a 501 amino acid protein with two active

Page 18: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

564 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

site motifs at amino acids 93-96 and 289-292. Itis structurally related to renin, cathepsins D and E,pepsinogens A and C, and the retroviral asparticproteases [516, 517]. The �-secretase-1 gene wasdescribed [518] as was the cell biology, regulation,and inhibition of �-secretase [519]. For the enzymaticactivities of �-secretase-1 and �-secretase-2 (BACE2;EC 3.4.23.45) in AD, see [520]. �-secretase-1 isalso required for myelination and correct bundling ofaxons by Schwann cells [521, 522].

An X-ray crystal structure of �-secretase-1 boundto the octapeptidic inhibitor OM99-2 was published[523, 524]. Apo and inhibitor complex structures of�-secretase-1 at the resolution of 1.75 A were deter-mined [525]. X-ray crystal structures of �-secretase-1bound to a macrocyclic peptidomimetic [526] and toa spiropiperidine iminohydantoin were communicated[527], as was the crystal structure of an active form of�-secretase-1 [528].

�-secretase-1 knockout mice are healthy despitelacking the primary �-secretase activity in the brain[529, 530]. �-secretase-1 null mice are rescued fromA�-dependent hippocampal memory deficits [531].

A rare gene mutation in the A�PP was identifiedin an Icelandic population by scientists of deCodegenetics, which protects against AD and age-relatedcognitive decline. This A673T mutation is directlyadjacent to the cleavage site of �-secretase (methionine671 and aspartic acid 672) [532]. For a commentary,see [533].

In view of the importance of the amyloid hypothesisfor AD, it is not surprising that many pharmaceu-tical companies embarked on medicinal chemistryprograms to discover �-secretase inhibitors (in alpha-betical order): Actelion, Allosterix, ALSP, Amgen,Archer Pharmaceuticals, Astellas, Astex Therapeutics,AstraZeneca, BACE Therapeutics, Boehringer Ingel-heim, Bristol Myers Squibb, CoMentis (company ofProf. Jordan Tang), Critical Outcome Technologies, DeNovo Pharmaceuticals, DuPont Pharma, Elan, EnvoyTherapeutics, Evotec, Genetics Company, Glaxo-SmithKline, IntelliHep, JADO Technologies, Johnson& Johnson, Kyoto University, LG Life Sciences,Ligand Pharmaceuticals, Lilly, Locus Pharmaceuti-cals, Medivir, Merck, NasVax, Noscira, Novartis,Pfizer, ProMediTech, Roche, Samada Research, sanofi,Schering-Plough, Senex Biotechnology, Shionogi,Sibia, Sunesis Pharmaceuticals, Takeda, Trans-TechPharma, Vertex Pharma, Vitae Pharmaceuticals, andWyeth. In addition, there are numerous excellentpapers by competent synthetic organic chemists andmolecular modelers working at universities. The prob-

lems of �-secretase inhibitors are mostly due to theirpeptidomimetic structures displaying high molecularweight, high polar surface area, low oral bioavail-ability, metabolic instability, low blood brain barrierpenetration, and susceptibility to P-glycoprotein trans-port out of the brain.

There are many interesting reviews on the phar-macology and medicinal chemistry of �-secretaseinhibitors (in chronological order): 2001: [534–536]2002: [537–543], 2003: [544–547], 2004: [548–550],2005: [551, 552], 2006: [553–557], 2007: [558–560],2008: [517, 561–563], 2009: [508, 564–569], 2010:[570–574], 2011: [575–578], 2012: [579–581].

LY-2886721 (Eli Lilly) was in two Phase I clini-cal trials since June 2010 (n = 50) and December 2010(n = 60) in the US. The studies were completed inOctober 2010 and in April 2011, respectively. In April2012 a randomized, double-blind, placebo-controlled,Phase I/II study (NCT01561430) was initiated inpatients with mild cognitive impairment due to AD(expected n = 129) in the US and is scheduled to com-plete in December 2013. For medicinal chemistry, see[582–586] (Thomson Reuters Pharma, update of July27, 2012). The structure was not communicated.

CTS-21166 (ASP-1702, ATG-Z1; Astellas Pharmaand CoMentis) is a �-secretase-1 inhibitor in Phase Iclinical trials since June 2007. In 48 volunteers, thedrug was safe and well tolerated (Thomson ReutersPharma, update of July 10, 2012). The structure wasnot communicated.

E-2609 (Eisai) is a �-secretase inhibitor in PhaseI clinical trials since December 2010 in the US inhealthy young and elderly volunteers (n = 48). Thetrial was completed in May 2012. Multiple-ascendingoral doses of E-2609 (25 to 400 mg for 14 days)were studied at a single site in the US in this ongoingtrial in healthy human volunteers of 50 to 85 yearsof age (both genders). The drug was safe and welltolerated with minor adverse effects. Reductions ofA� in CSF and plasma were demonstrated followingboth single and repeated administrations (ThomsonReuters Pharma, update of August 07, 2012). Thestructure was not communicated.

HPP-854 (TTP-854; High Point Pharmaceuticals,a spin-out from TransTech Pharma) is a �-secretaseinhibitor in Phase I clinical trials since November 2009(Thomson Reuters Pharma, update of April 5, 2012).The structure was not communicated.

MK-8931 (SCH-900931; Schering-Plough, nowMerck; Fig. 11) is an inhibitor of �-secretase inPhase I clinical trials since October 2009 (n = 40).In April 2012 data were presented from a two-part,

Page 19: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 565

Fig. 11. Beta-secretase inhibitors.

Page 20: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

566 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

randomized, double-blind, placebo-controlled singlerising-dose study (n = 40). Single doses of MK-8931from 2.5 to 550 mg were well-tolerated [587]. Formedicinal chemistry, see [548, 588–593] (ThomsonReuters Pharma, update of August 13, 2012).

For further documentation on the impressive�-secretase-1 inhibitor research program ongoing atMerck see (in chronological order): [594–621 [2]].

At the occasion of the 244th ACS meeting inPhiladelphia Merck presented a bicyclic iminohete-rocycle compound (Fig. 11) with an optimizedthiadiazine subunit with high affinity for �-secretase.

RG-7129 (Roche from a research collaboration withSiena Biotech) is a small molecule �-secretase (BACE)inhibitor in Phase I development since September2011 (Thomson Reuters Pharma, update of August 8,2012). The structure was not communicated. See alsothe interesting papers [621, 622].

Several �-secretase inhibitors of known structure arein preclinical evaluation (in alphabetical order):

Amgen (presumably AC-3; AMG-0683; Fig. 11)investigated series of 2-aminoquinolines and 2-aminopyridines as �-secretase-1 inhibitors [623].Hydroxy-ethylamine containing �-secretase inhibitorswere described very recently [624, 625] (ThomsonReuters Pharma, update of July 31, 2012).

AZ-13 (Astex Pharmaceuticals and AstraZeneca;Fig. 11) at 50, 75, 100, or 300 mmol/kg achieved aconcentration-dependent decrease in A�40, A�42, andsA�PP levels in brain and plasma of C57BL/6 mice.For medicinal chemistry, see [626–629] (ThomsonReuters Pharma, update of August 2, 2012).

Bristol-Myers Squibb (Fig. 11) is investigating aseries of 7-aza-indole compounds with IC50 values of10 nM against BACE-1 and 800 nM against BACE-2. For medicinal chemistry, see [630–635] (ThomsonReuters Pharma, update of August 8, 2011).

Elan (Fig. 11) presented data on hydroxyethylamine�-secretase-1 inhibitors with IC50 values against�-secretase-1 of 12 nM and an ED50 value of 2.1 nMin a HEK-293 cellular assay. For medicinal chemistry,see [636–643] (Thomson Reuters Pharma, update ofAugust 13, 2012).

Johnson & Johnson (presumably JNJ-715754,Fig. 11) is evaluating compounds from a seriesof aminopiperazines with an IC50 of 40 nM in a�-secretase-1 enzymatic assay, of 28 nM for hA�42,and 25 nM for hA�Total. Many medicinal chem-istry papers were communicated [644–649] (ThomsonReuters Pharma, update of January 18, 2012).

KMI-008, KMI-358, KMI-370, KMI-420,KMI-429, KMI-574, KMI-1027, and KMI-1303

(Kyoto Pharmaceutical University) are potent BACE1inhibitors [650–665]. KMI-429 (Fig. 12) is a�-secretase 1 inhibitor with enhanced cell membranepermeability (IC50 = 3.9 nM). KMI-574 (Fig. 12)is a �-secretase inhibitor produced by substitutingthe side chain of KMI-429 with a bioequivalentmaterial to enhance blood-brain barrier permeability(IC50 = 5.6 nM). KMI-1027 (Fig. 12) is a low molecu-lar weight �-secretase inhibitor with enhanced in vivoenzyme stability and blood-brain barrier permeability(IC50 = 50 nM). KMI-1303 (Fig. 12) is a �-secretase1 inhibitor with higher affinity for active site pockets(IC50 = 9 nM). The structures were obtained fromthe Wako Online Catalog, Research for Alzheimer’sDisease, edition November 2011.

L-655240 (Merck, Fig. 12) is a thromboxane A2antagonist, which was re-discovered as a BACE1inhibitor with IC50 of 4.47 �M by Chinese authors[666].

Novartis (Fig. 11) investigated a series of cyclicsulfoxide hydroxyethylamine �-secretase-1 inhibitors.The compound reduced CSF and brain A�40 levels ina dose-dependent manner and showed good efficacyat a 10 fold lower dose, when co-administered withthe CYP3A4 inhibitor ritonavir in various animal mod-els. For medicinal chemistry, see [667–671]. (ThomsonReuters Pharma, update of July 5, 2011).

TAK-070 (University of Tokyo under license fromTakeda) is a �-secretase and A� aggregation inhibitor(Thomson Reuters Pharma, update of June 8, 2012).The structure was not communicated.

WAY-258131 (Wyeth, now Pfizer; Fig. 11) inhibited�-secretase-1 with an IC50 value of 10 nM and reducedA�40 with an IC50 value of 20 nM. For medicinalchemistry, see [672–689] (Thomson Reuters Pharma,update of August 15, 2012).

Several companies have �-secretase-1 inhibitorsin preclinical development, whose structures werenot communicated (in alphabetical order): Allosterix,BACE Therapeutics, Boehringer Ingelheim, EnvoyTherapeutics [690], Evotec [691, 692], Il DongPharma (Seoul), IntelliHep (heparinoid �-secretaseinhibitors), LG Life Sciences [693], ProMediTech,Samada Research, Vitae Pharmaceuticals in collabo-ration with Boehringer Ingelheim and the TechnicalUniversity of Munich [694, 695].

A dual �-secretase-1 inhibitor and metal chelatorwas described [696].

Dual �7 nicotinic acetylcholine receptor activa-tors and �-secretase-1 inhibitors are investigated ina program at the University of Maryland (ThomsonReuters Pharma, update of February 24, 2012).

Page 21: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 567

Fig. 12. Beta-secretase inhibitors II.

Page 22: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

568 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

JADO Technologies synthesized a membrane-anchored version of a �-secretase transition-stateinhibitor by linking it to a sterol moiety. This inhibitorreduced enzyme activity much more efficiently thandid the free inhibitor in cultures cells and in vivo [697].

BBS-1 BACE inhibitor mAb vaccine (NasVaxunder license from Ramot at Tel Aviv University) isinvestigating a vaccine based on its lead mAb candidateBBS-1 (blocking �-site-1), which inhibits the abilityof �-secretase-1 to cleave A�PP (Thomson ReutersPharma, update of July 30, 2011).

Antibodies directed against the �-secretase cleav-age site of A�PP were described [698, 699].

Brain-targeted BACE1 antibody (Genentech,Roche Holding) is a bi-specific antibody against�-secretase-1 and the transferrin receptor to allowtranscytosis across the blood-brain barrier [700, 701](Thomson Reuters Pharma, update of May 14, 2012).This technique was pioneered by W. M. Pardridge ofUCLA [702–708].

Excellent papers on �-secretase-1 inhibitors fromuniversities were presented (in alphabetical order oftheir location): Mahidol University Bangkok [709,710], University of Darmstadt [711], TU Dresden[712], University of Duisburg-Essen [713], GwangjuInstitute of Science and Technology Korea [714],Linkoping University [715]; University of Liverpool[716, 717], University of Marseille [718], Max-Planck-Institut fur Biochemie Martinsried [719], Universityof Montreal [720–722], Technical University Munich[695], Peking University [723], University of Pisa[724–726], Purdue University [536, 559, 561, 727,728], Sapienza University Rome [729], Shanghai Insti-tute of Materia Medica [730, 731], Shujitsu University[732], Stockholm University [733–735], Universityof South Florida Tampa [736], University of Tokyo[665], SISSA-ISAS Trieste [737], CNRS Valbonne[738, 739], and Uppsala University [740–743].

Excellent papers on theoretical calculations on�-secretase-1 inhibitors were presented (in chrono-logical order) 2001: [744], 2003: [644], 2004: [645,646, 745], 2005: [746, 747], 2006: [748–751], 2007:[752–754], 2008: [755, 756], 2009: [757, 758], 2010:[759–765], 2011: [766–771], 2012: [772–778].

Several companies or institutions seem to have aban-doned the search for novel �-secretase-1 inhibitors,such as Actelion, De Novo, the Genetics Co (Callisto-Gen), Ligand, Locus Pharmaceuticals, Medivir [779],Noscira, and Plexxikon.

The development of allosteric peptide BACE-1inhibitors (Allosterix), of ARC-050 (Archer Phar-maceuticals), AZD-3839 (AstraZeneca [780, 781]),

BACE inhibitors (BACE Therapeutics, Evotec,Samada Research, and ProMediTech/LG Life Sci-ences), GRL-8234 (Oklahoma Medical ResearchFoundation [782]), GSK-188909 (GSK [783–791]),LY-2811376 (Lilly [792]), SIB-1281 (Sibia), of TAK-070 (Takeda [793, 794]), and of TC-1 (Merck underlicense from Sunesis [795, 796]) was terminated.

2.4. Drugs interacting with gamma-secretase

�-secretase is an aspartyl protease that cleavesits substrates within the transmembrane region in aprocess termed regulated-intramembrane-proteolysis(RIP). The enzyme consists of four protein compo-nents: presenilin 1 or 2, which contains the catalyticdomain, nicastrin, Aph-1 (anterior pharynx-1), andPen-2 (presenilin enhancer-2) in a 1 : 1:1 : 1 ratio [797].It is hypothesized that the free N-terminus of a �-secretase substrate first binds to the ectodomain ofnicastrin (a 709 amino acid type 1 membrane glyco-protein), which may facilitate its interaction with thedocking site on presenilin followed by relocation to theactive site on presenilin, where it is cleaved [798–812].�-secretase cleaves A�PP transmembrane domain in aprogressive stepwise manner at the �, ζ, and �-sites,resulting in A� species of varying length. This step-wise cleavage may occur via two different routes. Theyinitiate with the formation of A�49 and A�48 andthen proceed via the cleavage at approximately everythree residues, i.e., every helical turn of the substrate.The two product lines are �49-ζ46-�43-�40 and �48-ζ45-�42. Further cleavage will subsequently generatethe other isoforms A�39, A�38, and A�37, of whichA�38 results from the product line containing A�42[813–816]. The mechanism of �-secretase dysfunctionin familial AD was addressed [817].

�-secretase also cleaves other protein substrates,e.g., Notch, Jagged, and Nectin 1� [818–821].�-secretase cleavage of Notch leads to the release of thesmaller cytosolic fragment NICD (Notch intracellulardomain) important in signal transduction pathways. �-secretase inhibitors that are not Notch-sparing maycause severe gastrointestinal toxicity and interferewith the maturation of B- and T-lymphocytes in mice[822–827]). Note that A�PP interacts with Notchreceptors [828].

The first proof that �-secretase inhibitors reducedA� peptide levels in the brain was published alreadyin 2001 by Elan scientists [829].

In view of the importance of the amyloid hypothesisfor AD, it is not surprising that many pharmaceu-tical companies embarked on medicinal chemistry

Page 23: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 569

programs to discover �-secretase inhibitors andmodulators (in alphabetical order): Archer Pharmaceu-ticals, AstraZeneca, Bristol-Myers Squibb, Cellzome,Eisai, Elan, EnVivo Pharmaceuticals, Harvard Med-ical School, Intra-Cellular Therapies, Janssen, Lilly,Merck, Myriad Genetics, NeuroGenetic Pharmaceuti-cals, Ortho-McNeill, Pfizer, Roche, TorreyPinesThera-peutics and Wyeth (now Pfizer). There are many highlyinteresting publications on medicinal chemistry andpharmacology of �-secretase inhibitors and modu-lators reviewed (in chronological order) 2002: [539,830–834]; 2003: [835]; 2004: [836, 837]; 2005: [838,839]; 2006: [556, 557, 840–843]; 2007: [819]; 2008:[844]; 2009: [809, 845–847]; 2010: [848, 849]; 2011:[577, 850]; 2012: [851, 852].

A review on the molecular modeling and the designof �-secretase inhibitors was presented [607].

Many excellent reviews on the therapeutic poten-tial of �-secretase inhibitors and modulators werepublished [850, 853–863].

2.4.1. Gamma-secretase inhibitorsAvagacestat (BMS-708163; Bristol-Myers Squibb;

Fig. 13) is an orally active �-secretase inhibitor thatselectively inhibited A� production. A Phase II trialin 200 AD patients was initiated in February 2009;a second Phase II trial in 270 prodromal AD (mildcognitive impairment) patients over a period of 104weeks was initiated in May 2009. First reports on thetolerability profile, pharmacokinetic parameter, andpharmacodynamics markers were published [864–866[3]]. A sensitive and selective LC-MS/MS method fordetermination of avagacestat in plasma and CSF waspresented [866]. For medicinal chemistry and phar-macology, see [867, 878] (Thomson Reuters Pharma,update of August 10, 2012).

NIC5-15 (Humanetics under license from theMount Sinai School of Medicine, Fig. 13) is the naturalproduct D-pinitol, which inhibits the formation of A�plaques and inhibits �-secretase. In July 2009 encour-aging Phase IIa data of the study NCT00470418 werereported (Thomson Reuters Pharma, update of January19, 2012).

E-2212 (Eisai), a �-secretase inhibitor, is in PhaseI clinical evaluation since January 2010 (n = 91). Thestructure of E-2212 was not communicated. E-2212replaces the abandoned �-secretase modulator E-2012(Thomson Reuters Pharma, update of March 15, 2011).

(−)-GSI-1 (L-685,458; SCH-900229; Merck;Fig. 13) has an EC50 value of 15 mg/kg in a rat modeland displayed excellent oral pharmacokinetics. GSI-1lowered A�40 plasma concentration. By August 2011,

Phase I trials had begun. For the extensive medicinalchemistry see [879–902] (Thomson Reuters Pharma,update of May 04, 2012).

Several �-secretase inhibitors are currently in pre-clinical evaluation (in alphabetical order):

Harvard Medical School scientists are investigat-ing a series of Notch-sparing �-secretase inhibitors forthe potential treatment of AD (one structure shownin Fig. 13). One of the compounds (AD-1068) has anIC50 value of 9 nM against A�42 and an IC50 value of1.8 �M for Notch. For medicinal chemistry (early workwas performed in part at the University of Tennessee),see [903–911]. An excellent review was published in2010 [848]. It appears that the development was termi-nated recently (Thomson Reuters Pharma, update ofJune 25, 2012).

MRK-560 (SCH-1375975; Schering-Plough, nowMerck; Fig. 13) reduced A� plaque deposition with-out evidence of notch-related pathology in the Tg2576mouse [912–915]. For medicinal chemistry, see [885,916–919, 895, 896, 920–927] (Thomson ReutersPharma, update of November 24, 2011).

NGP-555 (NeuroGenetic Pharmaceuticals, Fig. 14)has an IC50 for A� of 8.3 nM. It reduced levels ofA�42 in brain and plasma at doses up to 100 mg/kg p.o.Chronic treatment with 50 mg/kg for 7 months signifi-cantly reduced the percentage area of plaques in mousebrains. There was no evidence of Notch-mediated GItoxicity [928]. A potential follow-up compound may beNGP-328, whose structure was not disclosed (Thom-son Reuters Pharma, update of June 4, 2012).

Pepscan Therapeutics is investigating a series ofpeptide-based �-secretase inhibitors for the potentialtreatment of AD (Thomson Reuters Pharma, update ofApril 1, 2011). The structures were not communicated.

Pfizer (Fig. 14) presented a novel �-secretaseinhibitor containing a bicyclo-[1.1.1]-pentanyl moietywith an IC50 of 0.178 nM being equipotent to avagace-stat (IC50 = 0.196 nM) [929]. For additional medicinalchemistry papers from Pfizer, see [930–935] (ThomsonReuters Pharma, update of July 30, 2012).

RO-02 (Roche; Fig. 14) inhibited enzyme activ-ity with an A�42 inhibition IC50 value of 500 nM.For medicinal chemistry, see [936] (Thomson ReutersPharma, update of July 20, 2011).

Large �-secretase inhibitor research programs werealso ongoing at Amgen [937] and at Elan, see[938–947].

Excellent papers on �-secretase inhibitors from uni-versities were presented (in chronological order) 2001:[948, 949]; 2004: [950, 951]; 2005: [952]; 2006: [953,954]; 2009: [955, 956]; 2010: [957]; 2012: [958, 959].

Page 24: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

570 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

Fig. 13. �-secretase inhibitors.

Excellent papers on theoretical calculations on�-secretase inhibitors have been presented (in chrono-logical order) [960–967].

Critical Outcome Technologies is investigatingorally available, dual �- and �- secretase inhibitors(Thomson Reuters Pharma, update of June 24,2011).

The development of several �-secretase inhibitorswas terminated, of ARC-069 (Archer Pharmaceu-ticals, Roskamp Institute), begacestat (GSI-953;WAY-210953; PF-5212362; Wyeth, now Pfizer [809,968–974]), BMS-299897 (Bristol-Myers Squibb;[975–978]), BMS-433796 (Bristol-Myers Squibb[869, 979]), E-2012 (Eisai), ELND-006 and ELND-007 (both Elan [944–946, 980, 981]; GSI-136 (Wyeth,now Pfizer [982]), MK-0752 (Merck; in Phase II for thetreatment of cancer, but terminated for the indicationof AD [983]); PF-03084014 (Pfizer; in Phase I for thetreatment of cancer, but terminated for the indicationof AD [984–987]), RO-4929097 (RG-4733; Roche,for the indication breast cancer [988–993]), sema-gacestat (LY-450,139; Eli Lilly [931, 932, 994–1010])and of SR-973 (DuPont/Scios, now Bristol-MyersSquibb).

2.4.2. Gamma-secretase modulatorsA �-secretase modulator interacts with allosteric

binding sites at the protease complex shifting the pro-teolytic processing of A�PP toward higher productionof shorter A� species such as A�38 at the expense ofthe highly toxic A�42 isoform. Impairment of Notchprocessing and signaling was not observed [1011].Reviews on �-secretase modulators were presented (inchronological order) 2005: [1012]; 2006: [556, 557,1013]; 2007: [1014, 1015]; 2008: [833, 1016–1019];2010: [821]; 2011: [1020, 1021]; 2012: [1022–1024].

Tarenflurbil (MPC-7869; (R)-flurbiprofen, Flur-izan, Myriad Genetics, Loma Linda University;Fig. 14) is a COX inhibitor and �-secretase modu-lator that selectively reduced the levels of A�42 invivo [1025, 1026]). The IC50 values for the inhibi-tion of COX-1 are 44 �M, of COX-2 : 123 �M, and ofA�42: 280 �M [1027]. Chronic administration of R-flurbiprofen attenuated learning impairments in trans-genic A�PPswe mice [1028]. Tarenflurbil upregulatednerve growth factor and brain-derived neurotrophicfactor protecting both human neuroblastoma cell linesand primary neurons from cytotoxicity associated withexposure to A�42 or hydrogen peroxide [1029].

Page 25: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 571

Fig. 14. �-secretase inhibitors and modulators.

Tarenflurbil was tested in a Phase II study in 210patients with doses of 400 mg and 800 mg twice perday or placebo for 12 months. Patients with mild AD inthe 800 mg tarenflurbil group had lower rates of declinethan those in the placebo group concerning activitiesof daily living. In patients with moderate AD 800 mgtarenflurbil twice a day had no significant effects onADCS-ADL and ADAS-cog [1030].

Tarenflurbil was tested in a Phase III study in 1,684patients, of which 1,046 completed the trial. Taren-flurbil had no beneficial effect on the co-primaryoutcomes for ADCS-ADL and for ADAS-cog [1031];for commentaries see [1032–1034] (Thomson ReutersPharma, update of August 24, 2012).

With the termination of the development oftarenflurbil, the NO-releasing flurbiprofen derivativeHCT-1026 (NicOx) was also stopped.

CHF-5074 (Chiesi Farmaceutici; Fig. 14) showed aconcentration dependent inhibitory activity on A�42secretion (IC50 = 40 �M). At 300 �M, an inhibitoryeffect was detected on COX-1 (–40%), but not onCOX-2 [1035]. It is devoid of Notch-interfering activi-ties in vitro [1036]. Treatment of 6 months old hA�PPtransgenic mice for 6 months significantly reduced

the number of plaques in cortex and hippocampus,whereas ibuprofen did not reduce A� burden [1037].CHF-5074 treatment of Tg2576 mice from 6 to 15months of age resulted in a significant attenuation ofthe neurogenesis impairment in hippocampus [859,1038]. For the reduction of the accumulation of hyper-phosphorylated tau, see [1039], for the restorationof hippocampal plasticity in Tg2576 mice [1040]and for the effects in a mouse model of scrapie[1041].

CHF-5074 was tested in a double-blind, placebo-controlled Phase I ascending oral dose study in 48healthy males with 200, 400, and 600 mg/day for 14days (n = 12 each per dose level and 12 receivingplacebo). The drug was well tolerated. In March 2011,a Phase II trial was initiated in the US and in Italy(n = 96). Trial completion is expected for July 2012(Thomson Reuters Pharma, update of July 23, 2012).

EVP-0962 (EnVivo Pharmaceuticals) is a �-secretase modulator in Phase I clinical evaluation sinceJune 2011 in healthy volunteers. By June 2012, thePhase I trial was completed (Thomson Reuters Pharma,update of August 14, 2012). Its structure was not com-municated.

Page 26: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

572 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

Several �-secretase modulators are currently in pre-clinical evaluation (in alphabetical order):

Amgen (Fig. 15) presented preclinical data on anovel �-secretase modulator at the occasion of the41st SFN Meeting in Washington in November 2011(Thomson Reuters Pharma, update of December 6,2011).

AZ-4800 (AstraZeneca; Fig. 15) at 75, 150, and300 �mol/kg caused a dose dependent decrease ofbrain A�42 1.5 hours after administration to C57mice (Thomson Reuters Pharma, update of April 1,2011). For medicinal chemistry, see [1042, 1043].AstraZeneca scientists described the detailed phar-macological evaluation of the �-secretase modulatorsAZ3303 and AZ1136 (both Fig. 16) [1044, 1045]. Thesecond-generation �-secretase modulators have differ-ent modes of action regarding Notch processing [1046](Thomson Reuters Pharma, update of July 4, 2012).

BIIB-042 (Biogen Idec; Fig. 15) is a �-secretasemodulator, which significantly reduced brain A�42 lev-els in CF-1 mice and in Fisher rats and plasma A�42levels in cynomolgus monkeys. For medicinal chem-istry, see [1047, 1048] (Thomson Reuters Pharma,update of March 27, 2012).

GSM-2 (Merck UK [1049], Fig. 15), as was inves-tigated by scientists of Astellas [1050], significantlyameliorated memory deficits in Tg2576 mice and didnot affect normal cognition in wild-type mice. In con-trast �-secretase inhibitors avagacestat (BMS-708163)and semagacestat (LY-450139) on subchronic dosingimpaired normal cognition in 3 month old Tg2576mice.

GSM-10h (GSK, Fig. 15) is an orallyactive piperidine-derived �-secretase modulator[1051–1054]. GSK is also investigating �-secretasemodulators derived from pyridazines (Fig. 15 [1055])and from pyridines (Fig. 15) [1056] (ThomsonReuters Pharma, update of January 13, 2012).

Janssen described a novel series of bicyclic hetero-cycles as potent �-secretase modulators [1057].

Merck (Fig. 16) is investigating �-secretase mod-ulators with fused 5,6-bicyclic heterocycles. The bestcompound had an IC50 of 122 nM for the inhibitionof A�42 and exhibited good selectivity over overall�-secretase inhibition (IC50 ratio of 38 fold). In rats,the compound reduced A�42 in the CSF by 26%after 3 hours at 30 mg/kg p.o. Medicinal chemistrywas described [1058–1063]. Important mechanisticinsights were contributed [1064] (Thomson ReutersPharma, update of August 15, 2012).

Merck (Fig. 16) presented a new series of triazoleanalogues, which inhibited A�42 and A�40 with IC50

values of 0.2 and 2.06 �M, respectively, and showeda hERG interaction of IC50 > 10 �M [1065, 1066].In a third series, Merck scientists presented pyridoneand pyridazone heterocycles as �-secretase modulators[1067].

Pfizer medicinal chemists presented novel dihy-drobenzofuran amides as orally bioavailable, centrallyactive �-secretase modulators [1068] (ThomsonReuters Pharma, update of July 30, 2012).

Roche presented novel �-secretase modulators at theoccasion of the Alzheimer’s Association InternationalConference in Paris, France, July 16–21, 2011. Onecompound (Fig. 16) had an A�42 EC50 value of 0.1 �Mand demonstrated dose-dependent efficacy in trans-genic A�PPswe mice. CSF A�42 levels were reducedin non-transgenic rats for longer than 8 hours. A goodpharmacokinetic profile was seen (Thomson ReutersPharma, update of July 27, 2012).

SPI-014, SPI-1802, SPI-1810, and SPI-1865(Satori Pharmaceuticals, Figure 16) are �-secretasemodulators reducing the levels of A�42 without alter-ing total A� levels in cell lines and in animal modelsof AD (Thomson Reuters Pharma, update of July 31,2012). The structures were not communicated.

TorreyPines Therapeutics presented a novel class ofaminothiazoles as �-secretase modulators. Compound3 (Fig. 16) displayed an IC50 for A�42 of 5 nM [1069].

Excellent papers on �-secretase modulators fromuniversities were presented from the TU Darmstadt[1070–1074], the University of Duesseldorf [1075],Emory University [1076], from the Mayo Clinic[1077], the Memorial Sloan-Kettering Cancer Center[1078–1080], the University of Munich [1081, 1082],and the University of Tokyo [1083].

Excellent papers on theoretical calculations on�-secretase modulators were presented from the Uni-versity of Duesseldorf [1084], the MassachusettsGeneral Hospital [1085–1087], and Vanderbilt Univer-sity [1088].

Dual modulators of �-secretase and peroxisomeproliferator-activated receptor � (PPAR�) werepresented to produce compounds with IC50 (A�42) of5.1 �M and EC50 (PPAR�) of 6.6 �M [1089, 1090].

The development of JNJ-4018677 and JNJ-42601572 (Janssen, formerly Ortho-McNeil Pharma-ceuticals under license from Cellzome) [1091] wasdiscontinued.

2.4.3. Inhibitors of gamma-secretase activatingprotein

�-secretase activating protein (GSAP) interactsboth with �-secretase and its substrate, the A�PP

Page 27: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 573

Fig. 15. �-secretase modulators.

Page 28: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

574 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

Fig. 16. �-secretase modulators and a Notch pathway inhibitor.

carboxy-terminal fragment (A�PP-CTF). ReducingGSAP concentrations in cell lines decreased A�concentrations, hence GSAP may be a new thera-peutic target for AD [1092]. For a commentary, see[1093]. The immunohistochemical characterization of

�-secretase activation protein expression in AD brainswas described [1094]; the purification and characteri-zation of human GSAP [1095].

IC-200155 (Intra-Cellular Therapies) is a com-pound that decreases A� levels in the brain by

Page 29: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 575

Fig. 17. One 11� hydroxysteroid dehydrogenase inhibitor, three calpain and one cathepsin B inhibitor(s).

interacting with GSAP (Thomson Reuters Pharma,update of February 24, 2012). The structure was notcommunicated.

2.4.4. Notch pathway inhibitorsAGT-0031 (AxoGlia Therapeutics; Fig. 16) is an

inhibitor of the Notch pathway, which is involved inastrocyte differentiation and inflammatory activationfor the potential treatment of multiple sclerosis andAD (Thomson Reuters Pharma, update of February29, 2012).

2.5. Drugs interacting with beta hexosaminidase

The first report of short-term memory deficits in amurine model of lysosomal storage diseases includingSandhoff disease was described [1096].

Amicus Therapeutics is investigating smallmolecule pharmacological chaperones that bind to andactivate the lysosomal enzyme beta-hexosaminidasefor the potential oral treatment of AD. Thesecompounds lowered the levels of GM2 and GM3gangliosides associated with A� aggregation (Thom-

son Reuters Pharma, update of March 28, 2012). Nostructures were communicated.

2.6. Drugs interacting with 11 betahydroxysteroid dehydrogenase

The topic 11�-hydroxysteroid dehydrogenase type1 (11�-HSD-1), brain atrophy, and cognitive declinewas reviewed [1097, 1098]. 11�-HSD1 expression wasenhanced in the aged mouse hippocampus and causedmemory impairment [1099]. Enhanced hippocampalLTP and spatial learning was found in 11�-HSD1knockout mice [1100, 1101]. Partial deficiency orshort term inhibition of 11�-HSD1 improved cognitivefunction in aged mice [1102, 1103]). Inhibition of 11�-hydroxysteroid dehydrogenase by carbenoxolone (100mg three times per day) improved cognitive functionin elderly men [1104].

ABT-384 (Abbott Laboratories) is in a double-blind safety/efficacy Phase II trial since May 2010 inpatients (n = 260) with mild to moderate AD in the UK.Despite being safe, well tolerated and tested at dosesassociated with full inhibition of brain 11�-HSD-1activity, it did not induce symptomatic improvement in

Page 30: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

576 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

mild to moderate AD during the 12-week trial period.Therefore the trial was terminated (Thomson ReutersPharma, update of July 18, 2012). The structure ofABT-384 was not communicated.

KR-1-2 (Korea Research Institute of ChemicalTechnology) suppressed cortisol by inhibiting 11�-hydroxysteroid dehydrogenase type 1. The drug isintended for the treatment of glaucoma and demen-tia (Thomson Reuters Pharma, update of February 27,2012).

UE-1961, UE-2811 (Fig. 17) and UE-2343 (Univ.of Edinburgh in collaboration with Argenta Discovery)are inhibitors of 11�-hydroxysteroid dehydrogenase1 (11�-HSD1) for the potential treatment of age-related cognitive disorders, such as memory loss[1102]. UE-2343 showed cognition enhancement inaged transgenic Tg2576 mice after one month of treat-ment in a passive avoidance test and a significantreduction of their A� plaque load (Thomson ReutersPharma, update of March 15, 2012). Another classof 11�-HSD1 inhibitors, i.e., 1,5-substituted 1H tetra-zoles, was described [1105].

2.7. Drugs interacting with calpain

The mechanistic involvement of the calpain-calpastatin system in AD neuropathology wasreviewed [1106]. Calpain inhibitors for the treatmentof AD were discussed [1107, 1108]. Over-activatedcalpain caused a down-regulation of cAMP-dependentprotein kinase in AD brain [1109]. Neuronal over-expression of the endogenous inhibitor of calpains,calpastatin, in a mouse model of AD reduced A�pathology [1110].

A-705253 (Abbott, formerly Knoll; Fig. 17) isan orally active calpain inhibitor for the potentialtreatment of AD. A-705253 significantly reducedcholinergic neurodegeneration caused by A� in adose-dependent manner in rats [1111–1117]. It alsoprevented stress-induced tau hyperphosphorylation invitro and in vivo [1118, 1119] (Thomson ReutersPharma, update of May 16, 2011).

ABT-957 (Abbott) is a calpain inhibitor for thepotential treatment of neurological disorders includ-ing AD (Thomson Reuters Pharma, update of March2, 2012). The structure was not communicated.

AK-295 (CX-295, Vasolex; AxoTect under licensefrom Cortex Pharmaceuticals using technologylicensed from Alkermes, Fig. 17) attenuated motor andcognitive deficits following experimental brain injuryin the rat [1120] (Thomson Reuters Pharma, update ofFebruary 9, 2012).

SNJ-1945 (Senju Pharmaceutical; Fig. 17) is apotent calpain inhibitor for the potential oral treat-ment of age-related macular degeneration, retinopathy,and optic neuritis. Preclinical data were reported[1121–1124]. Medicinal chemistry papers were con-tributed [1125–1128] (Thomson Reuters Pharma,update of April 18, 2012).

The development of calpain inhibitors BDA-410 (Mitsubishi-Tokyo Pharmaceuticals), CEP-3122(Cephalon, now Teva), EP-475 (University of Ten-nessee), MDL-28170 (Hoechst Marion Roussel, nowsanofi [1129–1131]), and of TH-3501 (University ofVirginia) was terminated.

2.8. Drugs interacting with carbonic anhydrase

Carbonic anhydrase dysfunction impaired cognitionand was associated with mental retardation, AD, andaging [1132].

The Blanchette Rockefeller Neurosciences Insti-tute is investigating phenylalanine compounds actingas carbonic anhydrase activators for the potentialtreatment of attention deficit disorders and memoryproblems in AD (Thomson Reuters Pharma, update ofFebruary 16, 2012).

2.9. Drugs interacting with caspases

Caspases, or cysteine-aspartic proteases or cysteine-dependent aspartate-directed proteases, are a family ofcysteine proteases that play essential roles in apoptosis(programmed cell death), necrosis, and inflammation.For a review on caspase-6 and neurodegeneration,see [1133]; for the role of caspases in AD, see[1134–1138].

NWL-117 (New World Laboratories) is an irre-versible inhibitor of active caspase-6 for thepotential treatment of AD (Thomson Reuters Pharma,update of July 18, 2012). The structure was notcommunicated.

2.10. Drugs interacting withCatechol-O-methyltransferase (COMT)

The topic COMT, cognition, and psychosis wasreviewed [1139–1141]. The potential role of COMTinhibitors for the treatment of cognitive deficits asso-ciated with schizophrenia was described [1142, 1143].

Tolcapone (Tasmar; Roche, launched in 1997)improved cognition and cortical information process-ing in normal human subjects [1144]. Tolcapone andEntacapone (Comtan; Orion in collaboration with

Page 31: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 577

Novartis launched in 1999) blocked fibril formation of�-synuclein and A� and protected against A�-inducedtoxicity [1145].

The company Cerecor is investigating COMTinhibitors for the potential treatment of schizophreniaincluding cognition (Thomson Reuters Pharma, updateof April 12, 2012). Structures were not communicated.

2.11. Drugs interacting with cathepsin

Reduction of cathepsin B in transgenic miceexpressing human wild-type A�PP resulted in sig-nificantly decreased brain A� [1146, 1147]. Highercathepsin B levels were found in plasma of AD patientscompared to healthy controls [1148].

Aloxistatin (E-64d; AB-007; ALP-496; ALSP;Fig. 17) is an inhibitor of cathepsin B for the poten-tial treatment of AD and traumatic brain injury[1149–1152]. Aloxistatin was previously evaluated forthe treatment of Duchenne muscular dystrophy byTaisho Pharmaceuticals, but the development was ter-minated (Thomson Reuters Pharma, update of June 29,2012).

Acetyl-L-leucyl-L-valyl-L-lysinal (ALSP) is a cys-teine protease inhibitor, which caused a significantreduction in brain A� and �-secretase activity byapproximately 50% after i.c.v. administration of0.06 mg/g brain weight/day for 28 days [1153] (Thom-son Reuters Pharma, update of June 28, 2012).

The development of CEL-5A (University of Cali-fornia at Irvine; a cathepsin D inhibitor) and cystatinC (New York University/Nathan Kline Institute, a cys-teine protease inhibitor) was terminated.

2.12. Drugs interacting with cholesterol24S-hydroxylase (CYP46A1)

Abnormal induction of the cholesterol-catabolicenzyme CYP46 in glial cells of AD patients wasalready recognized in 2001 by researchers at theKarolinska Institute [1154], followed by [1155–1157].It was shown that polymorphism in the cholesterol24S-hydroxylase gene (CYP46A1) associated withthe ApoE�3 allele increased the risk of AD andof mild cognitive impairment [1158, 1159]. Simi-lar results were obtained in elderly Chinese subjects[1160, 1161]. CYP46A1 functional promotor haplo-types decipher genetic susceptibility to AD [1162].Reviews were published [1163, 1164]. The crystalstructure of the enzyme was elucidated [1165]. ThecDNA cloning was described by scientists from the

University of Texas [1166], who generated also KOmice [1167].

AAV-CYP46A1 (INSERM in collaboration withsanofi) is an adeno-associated virus gene ther-apy encoding cholesterol 24-hydroxylase (CYP46A1gene) for the potential injectable treatment of AD.Reduced A� peptides, amyloid deposits, and trimericoligomers were observed in A�PP23 mice. Signifi-cant improvements in cognitive function assessed bythe Morris water maze were also seen in Tau22 mice[1168] (Thomson Reuters Pharma, update of June 6,2012). See also Part 3, Chapter 4 Drugs interactingwith Gene Expression.

2.13. Drugs interacting with cyclooxygenase(COX)

COX (EC 1.14.99.1) is an enzyme that is responsi-ble for the formation of prostaglandins, prostacyclin,and thromboxane. NSAIDs exert their effects throughinhibition of COX. Oligomers of A�1-42 induced theactivation of COX-2 in astrocytes [1169].

Epidemiological studies have documented a reducedprevalence of AD among users of NSAIDs [1170,1171, 1026, 1172–1178]).

In the Rotterdam study, 6,989 subjects 55 yearsof age or older were analyzed for the associa-tion of NSAIDs use and AD. The relative riskof AD was 0.95 in subjects with short-term use(1 month) of NSAIDs, 0.62 in subjects withintermediate-term use (1 to 24 months), and 0.20in those with long-term use (more than 24 months)[1179]. Several clinical trials using indomethacin,diclofenac/misoprostol, naproxen, nimesulide, cele-coxib, and rofecoxib showed no benefit for ADpatients, reviewed by [1177]. The ADAPT researchgroup could not show a prevention of AD dementiain a clinical trial using naproxen and celecoxib [1180,1181].

The NSAIDs ibuprofen, indomethacin, and sulindacsulfide preferentially decreased the highly amyloido-genic A�42 peptide [1172]. This effect was notmediated by inhibition of COX, but by an allostericmodulation of �-secretase [1064] and by a change inpresenilin 1 conformation [1182]. A list of compoundsdecreasing the formation of A�42 peptide comprisingdiclofenac, (R)-flurbiprofen, fenoprofen, ibuprofen,indomethacin, meclofenamic acid. and sulindac waspresented [1183]. NSAIDs dose-dependently destabi-lized preformed A� fibrils in vitro [1184, 1185]. Theseeffects were also shown in transgenic mice in vivo[1025, 1177].

Page 32: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

578 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

A�42-lowering NSAIDs constitute the foundingmembers of the new class of �-secretase modulators[1186], vide supra section 2.4.2.

2.14. Drugs interacting with D-amino acidoxidase

The therapeutic potential of D-amino acid oxidase(DAAO) inhibitors was described [1187]. Several sus-ceptibility genes for psychiatric disorders have beenidentified, among others G72, which is the D-aminoacid oxidase activator [1188–1191].

AS-2651816-00 (Astellas Pharma) is a novel DAAOinhibitor for the potential treatment of schizophreniawith an IC50 of 1.5 nM against human DAAO (Thom-son Reuters Pharma, update of August 24, 2012). Itis a 3-hydroxy-pyridazin-4(1H)-one, but the precisestructure was not communicated.

Eisai following the acquisition of MGI Pharmawas investigating DAAO inhibitors to enhance theactivity of D-serine. The compound 6-chlorobenzo[d]isoxazol-3-ol (CBIO) showed an IC50 of 200 nM. Itsdevelopment was terminated. Also the development ofDAAO inhibitors of Merck and Pfizer and of SEP-227900 (Sunovion) was terminated.

2.15. Drugs interacting with Glutaminyl Cyclase

Glutaminyl cyclase (QC) is an enzyme that catalyzesthe formation of pyroglutamic peptides or proteinsfrom a N-terminal glutamine residue. Human QC canperform in parallel the conversion of a N-terminal glu-tamate of a peptide to a pyroglutamated peptide. WhenA� is cleaved between amino acids 2 and 3, a pep-tide with a N-terminal glutamate is formed, which iscyclized by QC to the N-terminal pyroglutamate. Thistransformation renders the molecule more hydropho-bic and increases its aggregation velocity [1192–1197].Hence a QC inhibitor could prevent the formationof this species [1198–1204]. Secondary glutaminyl-cyclase inhibitor interactions were presented [4].

PBD-150 (Probiodrug; Fig. 18) is a potent inhibitorof human glutaminyl cyclase (IC50 = 17 nM) andtotally inhibits formation of A�3,11-(pE)40,42 in cellculture at 1 �M (Thomson Reuters Pharma, update ofFebruary 24, 2012).

A second generation of compounds was evaluated,in which the 1-propyl imidazole residue was replacedby the more rigid benzimidazole.

PQ-912 (Probiodrug) is a potent QC inhibitor inPhase I clinical studies with single- and multipleascending dose, blinded, placebo-controlled, random-

ized in healthy volunteers (n = 100). First resultsshowed that oral administration of PQ-912 was safeand well tolerated with relevant therapeutic levels inblood and CSF (Thomson Reuters Pharma, update ofJanuary 4, 2012). The structure of PQ-912 was notcommunicated.

2.16. Drugs interacting withglyceraldehyde-3-phosphate dehydrogenase

A proteomics analysis of the AD hippocampalproteome showed an increase of the levels of glyc-eraldehyde 3-phosphate dehydrogenase [1205]. Adecrease of the activity of cerebral glyceraldehyde-3-phosphate dehydrogenase in different animal modelsof AD was observed [1206].

Omigapil (SNT-317, TCH-346, CGP-3466; San-thera under license from Novartis; Fig. 18) is an orallybioavailable glyceraldehyde 3-phosphate dehydroge-nase modulator with anti-apoptotic properties for thetreatment of Parkinson’s disease and amyotrophic lat-eral sclerosis in Phase I clinical trials [1207–1221](Thomson Reuters Pharma, update of April 11,2012).

2.17. Drugs interacting with glycogen synthasekinase-3β (GSK-3β)

GSK-3� (EC 2.7.11.26, 47 kDa) is a ser-ine/threonine protein kinase ubiquitously expressedand involved in many cellular signaling pathwaysplaying a key role in the pathogenesis of AD. Itis probably the link between A� and tau patholo-gies. A� activates GSK-3� through impairment ofphosphatidyl-inositol-3/Akt signaling. A�-activatedGSK-3� induced hyperphosphorylation of tau, neu-rofibrillary tangle formation, neuronal death, andsynaptic loss (all found in AD brains). GSK-3�induced memory deficits in vivo. Crucial is thephosphorylation at serine 9. The cited papers arelisted in chronological order: 1999: [1222]; 2002:[1223–1225]; 2004: [1226–1228]; 2005: [1229]; 2006:[1230–1232]; 2007: [1233, 1234]; 2008: [1235–1240];2009: [1241–1243]; 2010: [1244, 1245]; 2011:[1246–1252]; 2012: [1253–1260].

GSK-3� was crystallized and the crystal structurewas resolved [1261, 1262]. 3D-QSAR studies werereported [1263, 1264]. Ligand-based virtual screeningwas elucidated [1265]. Scoring functions were applied[1266].

Tideglusib (NP-12, NP-031112; Nypta; Noscira,previously known as Neuropharma; Fig. 18) is the

Page 33: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 579

Fig. 18. A glyceraldehyde 3-phosphate dehydrogenase modulator, a glutaminyl cyclase inhibitor, 10 glycogen synthase-3� and one HDACinhibitor.

Page 34: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

580 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

lead compound of a series of orally active thiadia-zolidinediones, potent inhibitors of GSK-3 [1267]. APhase IIb clinical trial in AD patients was initiated inSpain, Germany, Finland, and the UK in April 2011.In January 2012, randomization of patients (n = 308)was completed in the double-blind trial. Preliminaryresults are expected by the end of 2012. In Decem-ber 2009, a Phase II trial for progressive supranuclearpalsy was initiated in the US and in Europe. Enrollmentwas completed in October 2010 (n = 125). The drugwas granted orphan status for progressive supranuclearpalsy in November 2009. First positive results of aclinical pilot study were reported [1268]. (ThomsonReuters Pharma, update of July 23, 2012).

There are currently many GSK-3� inhibitors in pre-clinical evaluation (in alphabetical order):

AX-9839 (ActivX Biosciences, Kyorin Pharmaceu-ticals, Fig. 18) is an inhibitor of GSK-3� for thepotential treatment of diabetes, AD, and various CNSdisorders [1269] (Thomson Reuters Pharma, update ofApril 18, 2012).

CG-9, CG-701338, CG-701446, and CG-701448(Crystal Genomics) are lead compounds from a seriesof GSK-3 inhibitors for the potential treatment of can-cer, AD, and schizophrenia (Thomson Reuters Pharma,update of May 02, 2012). The structures were not com-municated.

CP-70949 (Pfizer; Fig. 18) is a selective inhibitorof GSK-3� in preclinical evaluation [1270] (ThomsonReuters Pharma, update of July 11, 2011).

Dual GSK-3�/casein kinase 2 modulators (Uni-versity of Illinois at Chicago) are investigated for thepotential treatment of Alzheimer’s disease. (ThomsonReuters Pharma, update of September 20, 2012). Struc-tures were not communicated.

GSK-3� inhibitors (Pfizer, Fig. 18) investigated aseries of oxazole derivatives. The shown compoundhad an IC50 of 5 nM and a more than 100-fold selec-tivity against all other kinases and met safety criteria(Thomson Reuters Pharma, update of Aug. 28, 2012).

JI-7263 (Jeil Pharmaceuticals) is the lead compoundfrom a series of GSK-3� inhibitors for the poten-tial treatment of AD and amyotrophic lateral sclerosis(Thomson Reuters Pharma, update of May 7, 2012).The structure was not communicated.

Mitsubishi-Tanabe (Fig. 18) presented a potentGSK-3� inhibitor at the AIMECS 11 meeting in Tokyowith an IC50 of 12 nM (Thomson Reuters Pharma,update of January 24, 2012).

NNI-362 (NNI-AD; NeuroNascent) is a multikinaseand GSK-3� modulator inhibiting several tau phos-phorylation sites. Other orally active compounds that

stimulate neuron formation are NNI-X01, NNI-C, andNNI-251 (Thomson Reuters Pharma, update of July19, 2012). The structures were not communicated.

NP-101020 (Noscira) is a GSK-3� inhibitor for thepotential treatment of AD (Thomson Reuters Pharma,update of January 12, 2012). The structure was notcommunicated.

SN-2127 (AstraZeneca, Fig. 18) is one of severalGSK-3� inhibitors in evaluation by AstraZeneca asare SN-2568, SN-3728, and others (Thomson ReutersPharma, update of April 4, 2012).

Takeda (Fig. 18) presented a very potent GSK-3�inhibitor with IC50 of 2 nM. After oral administra-tion a significant reduction of tau phosphorylation wasobserved in aged JNPL3 mice [1271–1273]; Thom-son Reuters Pharma, update of January 04, 2012). Thecompound MMBO (Fig. 18) decreased tau phosphory-lation and ameliorated cognitive deficits in a transgenicmodel of AD [1274]).

TWS-119 (Scripps Research Institute; Fig. 18) isa compound that can induce neurogenesis in murineembryonic stem cells. The target of TWS-119 wasshown to be GSK-3� by affinity-based and biochemi-cal methods [1275].

VP1.15 (CSIC Madrid and University of Toronto,Fig. 18) is a dual GSK-3 and PDE7 inhibitor acting asan antipsychotic and cognitive enhancer in C57BL/6Jmice [1276, 1277].

Extensive programs to identify GSK-3� inhibitorshave been ongoing at AstraZeneca [1226, 1278–1280],at ChemDiv [1281], at GSK (e.g., SB-415286 andothers [1282–1287]), at GVK Biosciences [1288], atJohnson & Johnson [1289–1292], at Kemia [1293], atLilly [1294–1296], at Roche [1297], at Vertex [1298]),and at Yuyu in collaboration with CrystalGenomics[1299].

Potent GSK-3� inhibitors were described byresearchers at universities (in alphabetical order oftheir location): University of Athens [1300], UniversityAutonoma de Barcelona [1301], Technical Universityof Braunschweig [1302], University of Caen [1303,1304], University of Illinois at Chicago [1305–1310],Technical University of Darmstadt [1311, 1312, 1312],Martin-Luther University Halle [1313], University ofLa Rochelle [1314], University of Leuven [1231,1239–1241], CSIC Madrid [1315–1318], Universityof Louisiana at Monroe [1319], CNRS/Institut CurieOrsay [1320], Peking University [1321], Indian Insti-tute of Technology Roorkee [1322], Korea Institute ofScience and Technology Seoul [1323], Fudan Univer-sity Shanghai [1324], and the University of Talca, Chile[1325].

Page 35: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 581

SAR studies on GSK-3� inhibitors were publishedby researchers at the Technical University Darmstadt,University of Naples, University of Leuven, and TelAviv University [1326].

The Broad Institute is investigating ATP noncompet-itive and allosteric inhibitors of GSK-3� (ThomsonReuters Pharma update of June 3, 2011).

DM-204 (DiaMedica following its acquisition ofSanomune) is a monoclonal antibody that inhibitsGSK-3� (Thomson Reuters Pharma update of June 15,2012).

The development of AR-A014418 (AstraZeneca)was terminated [1226, 1278–1280] as was NP-103(Noscira) and UDA-680 (SAR-502250; MitsubishiPharma and sanofi). Also the development ofPropentofylline (HWA 285; Hoechst Werke Albert,now sanofi) for AD and for vascular dementia has beenterminated after results of the 72 week propentofyllinelong-term use (PLUS) clinical trial showed no treat-ment differences between the propentofylline-treatedgroup and the placebo-treated group [1327–1331].Propentofylline attenuated tau hyperphosphorylationby reducing the activated form of GSK-3� and byincreasing the inactivated form of GSK-3� [1332].Propentofylline is also a potent inhibitor of PDEs(see section 2.29.). The development of SAN-161(Sanomune, a subsidiary of DiaMedica), SB-216763(SmithKline Beecham, now GSK [1333], and the XD-4000 series (Xcellsyz, Lonza group) was terminated.

2.18. Drugs interacting with guanylyl cyclase

Soluble guanyl cyclase may be required foremotional learning and for both reference andworking memory [1334]. A selective irreversibleinhibitor of soluble guanyl cyclase, i.e., 1H-[1,2,4]-oxadiazole[4,3-a]-quinoxaline-1-one at doses of 5, 10,and 20 mg/kg impaired retention for the passive avoid-ance task in rats. Activation of soluble guanyl cyclaseand cGMP formation in the brain represents one ele-ment of effective neuroprotective pathways mediatedby NO [1335]. The design and synthesis of nomethi-azoles as NO-chimeras for neurodegenerative therapywas described [1336].

sGC-1016 (sGC Pharma) is a sustained-release NOmimetic for the potential treatment of AD in Phase Iclinical trial, which was completed in July 2012 show-ing high bioavailability (Thomson Reuters Pharma,update of July 17, 2012). The structure was not com-municated.

GT-715 and GT-1061 (Cita NeuroPharmaceuticals,Vernalis) were prototypes of engineered NO mimetics,

which activated soluble guanylyl cyclase and increasedcGMP formation. They improved task acquisition incognitively impaired animals and in the 6-OHDAmodel of Parkinson’s disease [1337–1341]. The devel-opment of both compounds was terminated.

2.19. Drugs interacting with heme oxygenase

Glial heme oxygenase-1 expression in AD and mildcognitive impairment was reviewed [1342–1346].

OB-28 (Osta Biotechnologies) is a hemeoxygenase-1 inhibitor for the potential injectabletreatment of AD showing statistically significantimprovement in behavioral deficits in double trans-genic (A�PPSWE/PS1dE9) mice (n = 103) aftertreatment with 15 or 30 mg/kg/day for 4 months(Thomson Reuters Pharma update of August 6, 2012).The structure of OB-28 was not communicated.

2.20. Drugs interacting with histone deacetylase(HDAC)

An in depth overview shows how to target “the cor-rect” HDAC(s) to treat cognitive disorders [1347]. Theroles of HDAC inhibition in neurodegenerative condi-tions were discussed [1348]. The role of HDAC6 inAlzheimer’s disease was discussed [1349]. The loss ofHDAC5 impaired memory function [1350].

EVP-0334 (EnVivo under license from Methyl-Gene) is developing the orally active HDAC inhibitoras a cognition enhancer for the potential treatmentof AD. A Phase I clinical trial was started in May2009 and finished in May 2010. Phase II trial areunderway since November 2011. Medicinal chemistrypapers were published [1351, 1352] (Thomson ReutersPharma, update of August 10, 2012).

4-Phenylbutyrate (Digna Biotech SL) amelioratedcognitive deficits and reduced tau pathology in anAD mouse model [1353–1355], reduced A� plaques,and rescued cognitive behavior in AD transgenic mice[1356, 1357]. The drug is in Phase I clinical trials forthe potential treatment of AD since April 2010 (Thom-son Reuters Pharma update of May 30, 2012).

Other HDAC inhibitors are currently in preclinicalevaluation (in alphabetical order):

CHDI-390576 and CHDI-00381817 (HDAC4inhibitors) are investigated by the CHDI Foundation incollaboration with BioFocus DPI (Thomson ReutersPharma update of August 10, 2012). The structureswere not disclosed.

Crebinostat (Harvard Medical School, Fig. 18)is a novel HDAC inhibitor and enhancer of CREB-

Page 36: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

582 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

regulated transcription and modulator of chromatin-mediated neuroplasticity [1358].

KAR-3010, KAR-3084, and KAR-3166 (KarusTherapeutics) are HDAC6 inhibitors for the poten-tial oral treatment of immune and inflammatorydisease including AD (Thomson Reuters Pharmaupdate of July 05, 2012). The structures were notdisclosed.

LB-201 and LB-205 (Lixte Biotechnology) aredrugs that target HDAC and prevent the degradationand restore the activity of glucocerebrosidase for thepotential treatment of neurological disorders such asGaucher’s disease [1359] (Thomson Reuters Pharmaupdate of April 05, 2012). The structures were notdisclosed.

Dual PKC activators and HDAC inhibitors weredescribed [1360].

The development of EHT-0205 (ExonHit) wasterminated.

2.21. Drugs interacting with HMG-CoA reductase

The enzyme 3-hydroxy-3-methyl-glutaryl-CoAreductase (EC 1.1.1.88) is the rate-controlling enzymeof the mevalonate pathway, which produces choles-terol and other isoprenoids. It is the target of thestatins (or HMG-CoA reductase inhibitors). In alarge study comprising 9,844 participants at agesfrom 40 to 45 years the risk of AD was evaluated inrelation to cholesterol levels. The analyses revealedthat cholesterol levels >220 mg/dL were a significantrisk factor for AD [1361]. See also the review [1362].

A lower prevalence of probable AD in a cohort ofpatients taking lovastatin or pravastatin between 60and 73% (p < 0.001) lower than the total patient popu-lation or compared with patients taking other medica-tions typically used for the treatment of hypertension orcardiovascular disease was found [1363–1367]. Excel-lent reviews were provided [1368–1375]. The linkbetween altered cholesterol metabolism and AD wasdescribed [1376]. Statins, risk of dementia, and cogni-tive function were discussed [1377].

Statins can shift A�PP processing to the non-amyloidogenic �-secretase pathway, e.g., simvastatin,which in addition prevented neuroinflammation andoxidative stress [1378, 1379], atorvastatin, which inaddition prevented neuroinflammation [1380, 1381]and lovastatin, which in addition stimulated the upreg-ulation of �7 nicotinic acetylcholine receptors [1382,1383]. A�40 blocked cerebral perivascular sympa-thetic �7-nAChRs, which was prevented by statinssuch as mevastatin and lovastatin [1384, 1385].

Pitavastatin attenuated celecoxib- and streptozotocin-induced experimental dementia [1386]. Atovastatinand pitavastatin reduced senile plaque and phospho-rylated tau in aged A�PP mice [1387, 1388].

These promising biological results in experimentalanimals were not confirmed in extensive clinical trials.Only a slight reduction of cognitive decline in a largestudy including 3,334 participants was found [1389].A post hoc analysis from three double-blind, placebo-controlled, clinical trials of galantamine in patientswith AD was done [1390]. Patients were divided intofour treatment groups, statin plus galantamine (n = 42),statin alone (n = 50), galantamine alone (n = 614),and placebo (n = 619). Galantamine was associatedwith a significant beneficial effect on cognitive sta-tus (p < 0.001). The use of statins did not produce asignificant beneficial effect (p = 0.083).

A proof-of-concept randomized controlled trial ofAtorvastatin (Lipitor, Pfizer, launched in 1996) indi-cated a trend for symptomatic benefit in mild tomoderate AD patients [1391–1393]. But the largeLEAD (Lipitor’s effect in Alzheimer’s Dementia)study in 640 patients, who were randomized to atorvas-tatin 80 mg/day or placebo for 72 weeks followed bya double-blind, 8-week atorvastatin withdrawal phasedid not show a significant benefit for cognition asmeasured by ADAS-cog (p = 0.26) or for global func-tion as measured by ADCS-Clinical Global Impressionof Change (p = 0.73) compared with placebo [1394,1395]).

Pravastatin (Pravachol, Mevalotin; DaiichiSankyo, launched in 1989) was evaluated in PROS-PER (Prospective Study of Pravastatin in the Elderlyat Risk) in 5,804 participants at six different timepoints. No difference in cognitive decline was foundin patients treated with pravastatin compared toplacebo (p > 0.05). During a three year follow-upperiod no effect on cognitive decline was observed[1396].

Simvastatin (Zocor, MK-0733; Merck launchedin 1989) was evaluated in a 406 patient Phase IIIstudy conducted by the National Institute of Aging.Patients were treated with 20 mg simvastatin per dayfor six weeks and with 40 mg/day simvastatin foran additional 18 months. Simvastatin lowered lipidlevels, but had no effect on change of ADAS-cogscore or on secondary outcome measures [1397–1403].Chronic simvastatin treatment rescued cognitive func-tion in a transgenic mouse model of AD and enhancedLTP in the CA1 region of the hippocampus in slicesfrom C57BL/56 mice via inhibition of farnesylation[1404–1406].

Page 37: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 583

Fig. 19. One HMG-CoA-reductase, 7 kinase and one kynurenine aminotransferase II inhibitor(s).

NST-0037 (Neuron BioPharma; Fig. 19) is a nat-ural small molecule HMG-CoA-reductase inhibitor,which also has neuroprotective, antioxidant, andanti-convulsive activity. The synthesis was described

[1407]. Potential follow-up compounds are NST-0005and NST-0060, whereas the development of NST-0021was terminated (Thomson Reuters Pharma, update ofJune 13, 2012).

Page 38: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

584 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

2.22. Drugs interacting with insulin-degradingenzyme

The role of insulin-degrading enzyme in AD wasdiscussed [1408, 1409]. The expression and functionalprofiling of insulin-degrading enzyme in elderly andAD patients was reported [1410] as was the character-ization in human serum [1411].

The company Inventram is investigating insulin-degrading enzyme activators for the potential treatmentof type 2 diabetes and AD (Thomson Reuters Pharma,update of April 20, 2012). No structures were commu-nicated.

2.23. Drugs interacting with Kinases ( /= GSK-3β

and /= PKC)

Advances in the development of kinase inhibitortherapeutics for AD were presented [1412]. Note-worthy is that acetyl-L-carnitine (ST-200, Nicetile,Zibren; launched by Sigma-Tau in Italy in 1985 andin South Korea in 1995 as a memory enhancer) ame-liorated spatial memory deficits induced by inhibitionof phosphoinositol-3-kinase and PKC [1413]. Alsoclioquinol seems to operate in a similar way [1414].

Masitinib (AB-1010, AB Science; Fig. 19) is aninhibitor of c-kit, Lyn and PDGF-R kinases. It wasevaluated in a Phase II clinical trial as an adjuncttherapy to cholinesterase inhibitor and/or memantinein 26 patients with mild-to-moderate AD versusplacebo (n = 8). The masitinib treated patients showedimprovements in MMSE scores, the ADAS-Cog andthe Alzheimer’s Disease Cooperative Study Activitiesof Daily Living Inventory with statistical significancebetween treatment arms at week 12 and/or week24 (p = 0.016 and 0.030, respectively) [1415]. InSeptember 2010, EMA approved a pivotal Phase IIItrial in 300 AD patients to assess safety and efficacy of6 mg/kg/day masitinib over 24 weeks. The biologicalcharacterization was described [1416] (ThomsonReuters Pharma, update of August 10, 2012). Numer-ous other kinase inhibitor therapeutics are currentlyin preclinical evaluation (in alphabetical order):

AIK-2, AIK-2a, AIK-2c, and AIK-21 (AllinkyBiopharma) are non-ATP-competitive MAP kinaseinhibitors for the potential treatment of cerebrovascu-lar stroke and AD (Thomson Reuters Pharma, updateof October 28, 2011). The structures were not commu-nicated.

Beta carboline alkaloids (Translational GenomicsResearch Institute) including harmol (Fig. 19) inhibitdual specificity tyrosine phosphorylation regulated

kinase 1A (DYRK1A) and tau phosphorylation(IC50 = 90 nM). (Thomson Reuters Pharma, update ofJune 8, 2012).

Cadeprin (group S8A serine protease) antagonistsare investigated by researchers from the Universitiesof Strathclyde and Glasgow [1417] (Thomson ReutersPharma, update of May 3, 2012).

Casein kinase 1δ inhibitors (Proteome Sciences),such as PS-110 and PS-278, are investigated for thepotential treatment of AD. (Thomson Reuters Pharma,update of July 2, 2012). Structures were not commu-nicated.

CDK5/p25 inhibitors (Pfizer) are investigated forthe potential treatment of AD. A novel pyrrolo[3,2-b]pyridine derivative (Fig. 19) displayed low nMpotency. (Thomson Reuters Pharma, update of April14, 2011).

CZC-25146 (GSK, following its acquisition ofCellzome) is leucine-rich repeat kinase 2 (LRRK2)inhibitor for the potential treatment of Parkinson’s dis-ease. [1418] (Thomson Reuters Pharma, update of June1, 2012).

Dasatinib (Bristol-Myers Squibb, launched 2006), aSrc tyrosine kinase inhibitor, attenuated A� associatedmicrogliosis in a murine model of AD [1419].

DYRK-1 alpha protein kinase inhibitors (ArrienPharmaceuticals) are evaluated for the potentialtreatment of Alzheimer’s disease and Down syndrome.(Thomson Reuters Pharma, update of April 24, 2012).Structures were not communicated.

DYRK-1 alpha protein kinase inhibitors (CarnaBiosciences) is a program in the stage of lead optimiza-tion. (Thomson Reuters Pharma, update of March 16,2012). Structures were not communicated.

FRAX-120, FRAX-355, and FRAX-486 (Afraxis)showed IC50 values of 23, 58, and 14 nM for p21-activated kinase. The drugs have a potential for thetreatment of fragile X syndrome, autism, schizophre-nia, and AD (Thomson Reuters Pharma, update ofAugust 14, 2012). Structures were not communicated.

G-2019S (Zenobia Pharmaceuticals in collabora-tion with Johns Hopkins University) is a LRRK2inhibitor for the potential treatment of Parkinson’s dis-ease (Thomson Reuters Pharma, update of March 13,2012). The structure was not communicated.

Hydroxy-fasudil (Fig. 19, Asahi Kasei PharmaCorp.), a rho kinase (ROCK) inhibitor, improvedspatial learning and working memory in rats ina water radial-arm maze [1420]. ROCK inhibitionled to an increase of Rac1 activity and to anactivation of PKCζ which in turn phosphorylatedKIBRA involved in the formation of memory [1421].

Page 39: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 585

ROCK inhibition prevented tau hyperphosphorylation[1422].

KIBRA pathway modulators (Amnestix Inc., awholly owned subsidiary of SYGNIS Pharma) offer anew genetic link to cognition that may benefit patientssuffering from AD. For the association of KIBRA andlate onset AD, see [1423], for enhancement of cog-nition in anxiety disorders [1424] (Thomson ReutersPharma, update of August 14, 2012).

LDN-22684 (Sirtris Pharmaceuticals) is a LRRK2inhibitor for the potential treatment of Parkinson’s dis-ease [1425] (Thomson Reuters Pharma, update of June17, 2011). The structure was not communicated.

LRRK2 inhibitors (Arrien Pharmaceuticals) areinvestigated for the potential oral treatment ofParkinson’s disease (Thomson Reuters Pharma,update of May 18, 2012). The structures were notcommunicated.

LRRK2 inhibitors (Genentech, Fig. 19) are inves-tigated for the potential treatment of Parkinson’sdisease. The medicinal chemistry was described[1426]. (Thomson Reuters Pharma, update of June 26,2012). The lead structure was communicated.

LRRK2 inhibitors (Genosco, Oscotec Inc.) areinvestigated for the potential treatment of Parkinson’sdisease (Thomson Reuters Pharma, update of April 20,2012). The structures were not communicated.

LRRK2 inhibitors (Oncodesign Biotechnology)are investigated for the potential treatment of cancerand CNS diseases including Parkinson’s disease(Thomson Reuters Pharma, update of April 2, 2012).The structures were not communicated.

LRRK2 inhibitors (Origenis GmbH) are investi-gated for the potential treatment of Parkinson’s disease(Thomson Reuters Pharma, update of May 11, 2012).The structures were not communicated.

LRRK2 inhibitors (Pfizer) are investigated for thepotential treatment of Parkinson’s disease (ThomsonReuters Pharma, update of June 20, 2012). The struc-tures were not communicated.

LRRK2 inhibitors (Vernalis/Lundbeck) are inves-tigated for the potential treatment of Parkinson’sdisease (Thomson Reuters Pharma, update of May 02,2012). The structures were not communicated.

MARK inhibitors (microtubule affinity regulat-ing kinase; Medical Research Council Technology,MRCT) are investigated for the inhibition of tau phos-phorylation (Thomson Reuters Pharma, update of June1, 2012).

MARK3 inhibitors (microtubule affinity regulat-ing kinase 3, CDC25-associated kinase-1; Merck) arein preclinical evaluation for the potential treatment of

AD. The pyrazolo[1,5-a]pyridine compound (Fig. 19)showed t1/2’s in rat, dog, and Rhesus monkey of 3.5,2.3, and 1.4 hours, respectively (Thomson ReutersPharma, update of April 14, 2011).

Minokine (MW01-2-069A-SRM; NorthwesternUniversity; Fig. 19) is a p38 MAP kinase inhibitor forthe potential oral treatment of neurodegenerative dis-eases including AD (Thomson Reuters Pharma, updateof April 27, 2011).

NNI-351 (NNI-depression, NNI-DS, Neu-roNascent; Fig. 19) is an orally active compoundpreventing stress-induced neurogenesis reductionvia inhibition of dual specificity tyrosine-phosphorylation-regulated kinase 1 alpha (DYRK-1�)protein (Thomson Reuters Pharma, update of March22, 2012).

P-005 (NB Health Laboratory) is a small moleculehighly selective kinase inhibitor for the potential treat-ment of AD (Thomson Reuters Pharma, update ofDecember 23, 2011). The structure was not commu-nicated.

Protein kinase inhibitors (ManRos Therapeutics)are evaluated as AD medication (Thomson ReutersPharma, update of February 24, 2011). The structureswere not communicated.

SEL-103 (Selvita Life Sciences Solutions) is a pro-gram in collaboration with Orion Corporation, EspooFinland, to investigate novel, orally bioavailable andhighly selective small molecules for the potentialtreatment of multiple cognitive disorders includingAD (Thomson Reuters Pharma, update of July 4,2012).

SEL-141 (Selvita Life Sciences Solutions) is a pro-gram targeting kinases involved in tau phosphorylationfor the potential treatment of AD, Down syndrome, andcognitive disorders. Focus is on DYRK1A (ThomsonReuters Pharma, update of July 4, 2012). Structureswere not communicated.

Sorafenib, a small molecular inhibitor of tyro-sine protein kinases (VEGFR and PDGFR) and ofraf restored working memory in A�PPSWE transgenicmice [1427].

TTT-3002 (TauTaTis) is a multi-targeted kinaseinhibitor that can inhibit the LRRK2 gene and tumorgrowth for the potential treatment of Parkinson’s dis-ease, AD, and cancer. (Thomson Reuters Pharma,update of January 25, 2012). The structure was notcommunicated.

URMC-099-C (Califia, the University of NebraskaMedical Center and the University of Rochester) is aninhibitor of mixed lineage kinase-3 for the treatmentof HIV-associated neurocognitive disorder (Thomson

Page 40: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

586 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

Reuters Pharma, update of June 5, 2012). The structurewas not communicated.

Inhibitors of the protein kinase c-raf1, such as GW-5074 and ZM-336372, protected cortical cells againstA� toxicity [1428].

The development of AIKb2 (Allinky Biopharma;a MAP kinase inhibitor), BA-1001 (BioAxone; arho kinase inhibitor [1429]), GW-5074 (GSK, a c-raf1 inhibitor), ORS-1006 (Arrien Pharmaceuticals, aLRRK2 inhibitor), Pan-MLK inhibitors (Cephalon,now Teva), PLX-3397 (Plexxikon, a subsidiary of Dai-ichi Sankyo; an oral small-molecule dual Fms/kit andFlt3-ITD inhibitor; a Phase II trial for the treatmentof acute myelogenous leukemia continues), RS-4073(Daiichi Sankyo; a potentiator of TrkA and Mapkinase activation), SB-203580 (SmithKline Beecham,now GSK; a protein kinase and cytokine suppressionbinding protein (CSBP)/p38 inhibitor [1430], SCIO-323 (Scios, a p38 kinase inhibitor), SRN-003-556(SIRENADE Pharmaceuticals; an ERK2 inhibitor),and of ZK-808762 (University of Gottingen; aserine protease and Factor Xa inhibitor) wasterminated.

2.24. Drugs interacting with kynureninemono-oxygenase and kynureninetransaminase II

The kynurenine metabolism in AD was described[1431]. Reduction of kynurenic acid formationenhanced hippocampal plasticity and cognitivebehavior [1432]. Crystal structure-based selective tar-geting of kynurenine aminotransferase II for cognitiveenhancement was discussed [1433].

CHDI-003940246 and CHDI-00340246 (Evotec incollaboration with the CHDI Foundation) are kynure-nine mono-oxygenase inhibitors for the potentialtreatment of Huntington’s disease (Thomson ReutersPharma, update of January 10, 2012). The structureswere not communicated.

PF-04859989 (Pfizer, Fig. 19) is an inhibitor ofkynurenine (oxoglutarate) aminotransferase II for thepotential treatment of schizophrenia [1434] (ThomsonReuters Pharma, update of March 21, 2012).

2.25. Drugs interacting with 5-lipoxygenase

The enzyme 5-lipoxygenase (5-LO) catalyzes theconversion of arachidonic acid to 5-hydroxy-peroxy-eicosatetraenoic acid (5-HPETE) and subsequentlyto 5-hydroxy-eicosatetraenoic acid (5-HETE), whichare metabolized to different leukotrienes [1435]. The

group of Domenico Pratico found that A� depositionin brains of Tg2576 mice lacking 5-LO was reducedby 64 to 80%, suggesting that pharmacological inhibi-tion of 5-LO could provide a novel therapy for AD[1436]. Additional investigations showed that 5-LOregulated the formation of A� by activating the cAMP-response element binding protein (CREB), which inturn increased transcription of the �-secretase complex[1437, 1438].

In a Letter to the Editor of Annals of Neurology,Kenji Hashimoto [1439] suggested to test minocy-cline, which protected PC12 cells against NMDA-induced injury via inhibiting 5-lipoxygenase activation[1440].

Minocycline (Wyeth, now Pfizer and Takedalaunched in 1999) is a second generation tetracy-cline that effectively crosses the blood brain barrier.It improved cognitive impairment in AD models[1441]. It provided protection against A�25-35 inducedalterations of the somatostatin signaling pathway[1442, 1443]. It reduced microglial activation [1444,1445] and A�-derived neuroinflammation [1446].It recovered MTT-formazan exocytosis impaired byA� [1447]. Minocycline inhibited �-secretase-1 ina transgenic model of AD-like amyloid pathology[1448]. It reduced the development of abnormal tauspecies in models of AD [1449–1451]. Minocyclineimproved negative symptoms in patients with earlyschizophrenia [1452].

A new class of 5-lipoxygenase inhibitors was com-municated recently [1453].

2.26. Drugs interacting with monoamine oxidase

Recent efforts of medicinal chemists to exploreligands targeting MOAs were reviewed [1454]. Apatent-related survey on new MOA inhibitors was pub-lished [1455]. The changes of MAO A and B in ADwere elucidated [1456] as was the platelet MAO Bactivity in dementia [1457].

Ladostigil (TV-3326; Avraham Pharmaceuticalsunder license from Yissum Research Development, awholly owned company of the Hebrew University ofJerusalem; see Fig. 8) is a dual acetylcholine esteraseand MAO inhibitor currently in Phase II clinical tri-als in 190 patients in Europe since December 2010(Thomson Reuters Pharma, update of May 18, 2012).See section 2.1.1.9. Dual AChE and MAO inhibitors.

RG-1577 (RO-4602522; EVT-302; Roche underlicense from Evotec) is an orally active, selective,and reversible MAO-B inhibitor. The company wasplanning to start a Phase II trial in patients with AD

Page 41: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 587

in the second half of 2012 (Thomson Reuters Pharma,update of August 8, 2012). Its structure was notcommunicated.

OG-45 (Oryzon Genomics) is a dual MAO-Binhibitor and lysine specific demethylase-1 inhibitor.(Thomson Reuters Pharma, update of April 16, 2012).Its structure was not communicated.

VAR-10200 (HLA-20; Varinel; Fig. 20) is a dualiron-chelating agent and MAO-B inhibitor for thepotential treatment of age-related macular degener-ation [1458] (Thomson Reuters Pharma, update ofFebruary 24, 2012).

VAR-10300 (M-30; Varinel, Technion and theWeizmann Institute of Science; Fig. 20) combinesthe iron-chelating properties of 8-hydroxy-quinolinewith the MAO inhibitor moiety of rasaglinine [470,1459–1465] (Thomson Reuters Pharma, update of May15, 2012).

Rasagiline (TVP-1012; (R)-enantiomer; Azilect;marketed by Teva and Lundbeck; Fig. 20) proved to bea very valuable drug for the treatment of Parkinson’sdisease with sales in 2011 of USD 290 million reportedby Teva and USD 221 million reported by Lundbeck.Rasagiline was extensively characterized (in chrono-logical order) [1466–1478]. The effects of rasagiline oncognitive deficits in cognitively-impaired Parkinson’sdisease patients were evaluated [1479]. A�PP process-ing properties of rasagiline were described [1480].Rasagiline improved learning and memory in younghealthy rats [1481]. In July 2004 Eisai initiated PhaseII clinical trials in the US for the treatment of AD, lateralso in combination with donepezil. In March 2008development for the indication AD was discontinued(Thomson Reuters Pharma, update August 29, 2012).

Safinamide (FCE-26743; NM-1015, PNU-151774E; Zambon under license from Newron, whoacquired the rights from Pharmacia and Upjohn,now Pfizer; Fig. 20) is a potent MAO B inhibitor(IC50 = 98 nM; MAO A: IC50 = 580 �M, selectivityindex: 5,918 for the potential treatment of Parkinson’sdisease and AD [1482]. The first synthesis wasdescribed in 1998 [1483] followed by the solid-phasesynthesis [1482]. The enantiomeric separation ofsafinamide was disclosed [1484]. The biologicalcharacterization was described in detail [1485–1487].Several reviews were published [1488–1490]. PhaseIII trials in early Parkinson’s disease patients beganin June 2004. An expert opinion on safinamide inParkinson’s disease was published [1491]. A PhaseIII trial in AD patients was abandoned in late 2007(Thomson Reuters Pharma, update of August 21,2012).

The development of many other MAO inhibitorsfor the indication AD was terminated, of bifemelane(MCI-2016; SON-216; Sosei under license from Mit-subishi [1492, 1493]), EVT-301 (Evotec under licensefrom Roche), EXP-631 (Bristol-Myers Squibb),4-fluoroselegiline (Chinoin, now sanofi), HT-1067(Helicon Therapeutics), indantadol (Vernalis underlicense from Chiesi), lazabemide (Ro 19-6327; Roche[1494–1496]), milacemide (GD Searle, now Pfizer[1497–1499]), NW-1772 (Newron Pharmaceuticals),PF-9601N (University of Autonoma de Barcelona),Ro-41-1049 (Roche), SL-25.1188 and SL-34.0026(both sanofi) and of YY-125 (Yuyu Inc.).

2.27. Drugs modulating O-linkedN-acetylglucosaminidase (O-GlcNAcase;OGA)

O-GlcNAcase is the enzyme that plays a role inthe removal of N-acetylglucosamine groups from ser-ine and threonine residues in both the nucleus andthe cytoplasm of cells [1500, 1501]. O-GlcNAcasemay compete with phosphorylation of the same ser-ine or threonine residues. Studies to elucidate thebinding mode were carried out by [1502, 1503].O-GlcNAcylation directly regulates core componentsof the pluripotency network [1504].

Alectos Therapeutics/Merck are investigatingO-GlcNAcase modulators (Thomson Reuters Pharmaupdates of July 5, 2012).

GlcNAcstatin (University of Dundee in collabo-ration with Aquapharm Biodiscovery; Fig. 20) is amarine natural product for the potential treatment ofAD [1505, 1506]. For an efficient and versatile synthe-sis of GlcNAcstatin derivatives, see [1507] (ThomsonReuters Pharma, update of January 23, 2012).

NButGT (Seoul National University; Fig. 20),a specific inhibitor of O-GlcNAcase, reduced A�production by lowering �-secretase activity both invitro and in vivo. O-GlcNAcylation takes place at theS708 residue of nicastrin, a component of �-secretase[1508] = [5]. The synthesis was described by chemistsof the Simon Fraser University [6].

SEG-4 (Summit Corporation) inhibited OGA withIC50 and Ki values of 10 and 72 nM, respectively. Thecompound significantly reduced tau phosphorylation(Thomson Reuters Pharma, update of August 2, 2012).The structure was not communicated.

Thiamet-G (Simon Fraser University) is apotent inhibitor of human O-GlcNAcase (Ki = 21 nM;Fig. 20) and efficiently reduced phosphorylation oftau at Thr231, Ser396, and Ser422 in both rat cor-

Page 42: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

588 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

Fig. 20. Four monoamine oxidase and three O-linked N-acetylglucosaminidase inhibitors.

tex and hippocampus [1509–1512]. For commentariessee [1513, 1514]. Acute thiamet G treatment led toa decrease in tau phosphorylation at Thr181, Thr212,Ser214, Ser262/Ser356, Ser404, and Ser409 and anincrease in tau phosphorylation at Ser199, Ser202,Ser396, and Ser422 in mouse brain, probably via stim-ulation of GSK-3� activity [1515].

2.28. Drugs interacting with peptidyl-prolylcis-trans isomerase D

Peptidyl-prolyl cis-trans isomerase D or CyclophilinD (Cyp D; EC 5.2.1.8. Isomerase) is implicated in celldeath pathways. Blockade of Cyp D could be a potenttherapeutic strategy for degenerative disorders such asAD, ischemia, and multiple sclerosis [1516]. SolubleA� is also found in mitochondria, where it interactswith Cyp D, a component of the mitochondrial per-meability transition pore. Interference with the normalfunctions of this protein resulted in disruption of cellhomeostasis and ultimately cell death [1517].

The Hsp90-associated cis-trans peptidyl-prolylisomerase-FK506 binding protein 51 (FKBP51) wasrecently found to co-localize with the microtubule-associated protein tau in neurons and physically

interact with tau in brain tissues from humans, whodied from AD [1518].

PIN1 is a peptidyl-prolyl isomerase, which catalyzescis-trans isomerization of peptide bonds N-terminalto specific phospho-serine/threonine-proline motifs.PIN1 isomerizes phosphorylated tau and thus restoresthe ability of phosphorylated tau to bind microtubules,and eventually promote dephosphorylation of tau byPP2A phosphatase [1519].

Scynexis is investigating a series of Cyp D inhibitorsfor the potential treatment of muscle injury, ischemia,reperfusion injury, trauma, and neurodegenerative dis-ease (Thomson Reuters Pharma, update of June 28,2011).

2.29. Drugs interacting with phosphodiesterases

Several excellent reviews on PDE inhibition andcognition enhancement were published [1520–1524].Based on the expression of PDE mRNA in thehuman brain, it was suggested that PDE1 and PDE10inhibitors are strong candidates for the developmentof cognition enhancers. Chronic PDE type 2 inhi-bition with BAY60-7550 improved memory in theA�PPswe/PS1dE9 mouse model of AD [1525]. The

Page 43: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 589

medicinal chemistry of PDE4D allosteric modulators[1526], of PDE4 inhibitors [1527], of PDE5 inhibitors[1528, 1529], and of PDE10A inhibitors was reviewed[1530, 1531].

Etazolate (EHT-202; SQ-20009; Exonhit; seeFig. 10) is an orally active PDE4 inhibitor and anxi-olytic GABAA receptor modulator, which shifts A�PPprocessing toward the �-secretase pathway [503, 504,1532–1536]. The results of the first Phase II 3-month,randomized, placebo-controlled, double-blind study inAD patients was reported [505] (Thomson ReutersPharma, update of September 13, 2011). See also sec-tion 2.2. Drugs interacting with �-secretase.

PF-02545920 (MP-10; Pfizer; Fig. 21) is a PDE10Ainhibitor in a randomized, double-blind, placebo-controlled Phase II clinical trial in patients withacute schizophrenia (n = 260) since October 2010.PF-02545920 is also evaluated for the treatment ofHuntington’s disease [1531, 1537–1540]. (ThomsonReuters Pharma, update of July 19, 2012).

Lu-AF11167 (Lundbeck) is an inhibitor of a brain-expressed PDE enzyme for the potential treatment ofAD, Huntington’s chorea, and schizophrenia in a PhaseI clinical trial since March 2011 (Thomson ReutersPharma, update of August 8, 2012). The structure wasnot communicated.

There are several PDE inhibitors in preclinical eval-uation (in alphabetical order):

AMG-7980 (Amgen, Fig. 21) is a novel PDE10Ainhibitor with a Ki of 0.94 nM useful as tritiatedligand to measure PDE10A target occupancy in ratbrain [1541, 1542].

AVE-8112 (Aventis, now sanofi and Michael J.Fox Foundation) is a PDE4 inhibitor for the poten-tial treatment of Parkinson’s disease. In April 2012,a US Phase Ib study was planned (Thomson ReutersPharma, update of May 30, 2012). The structure wasnot communicated.

BCA-909 (BCR-909; Boehringer Ingelheim underlicense from biocrea GmbH) is a PDE2 inhibitor forthe potential treatment of mild cognitive impairmentin schizophrenia and AD (Thomson Reuters Pharma,update of May 30, 2012). The structure was notcommunicated.

Cilostazol (Otsuka), a selective PDE3 inhibitor, pro-tected against A�1-40-induced suppression of viabilityand neurite elongation [1543].

Dual PDE10/PDE 2 inhibitors (biocrea GmbH)are investigated for the potential treatment ofschizophrenia and AD (Thomson Reuters Pharma,update of January 18, 2012). Structures were notcommunicated.

GEBR-7b (University of Genoa; Fig. 21) is a novelPDE4D selective inhibitor that improves memory inrodents at non-emetic doses [1544] (Thomson ReutersPharma, update of December 21, 2011).

ITI-002A, IC-200214, and ITI-214 (Takedaunder license of Intra-Cellular Therapies) are PDE1inhibitors for the potential oral treatment of cogni-tive disorders associated with schizophrenia (ThomsonReuters Pharma, update of July 30, 2012). The struc-tures were not communicated.

OMS-182410 (Omeros) is a PDE10 inhibitor forthe potential treatment of schizophrenia (ThomsonReuters Pharma, update of July 19, 2012). The struc-ture was not communicated.

PDE2A inhibitor (Pfizer, Fig. 21) is evaluated forthe potential treatment of cognitive impairment asso-ciated with schizophrenia. In March 2012, a PDE2Ainhibitor with an IC50 of 4 nM was presented at the243rd ACS Meeting in San Diego (Thomson ReutersPharma, update of August 24, 2012).

Allosteric PDE4 inhibitors (Tetra Discovery Part-ners; West Virginia University) are evaluated forthe potential treatment of mild cognitive impairment(Thomson Reuters Pharma, update of July 5, 2012).Structures were not communicated.

PDE7 inhibitors (Omeros) are a potential newtreatment of Parkinson’s disease (Thomson ReutersPharma, update of May 23, 2012). The structures werenot communicated.

A PDE9 inhibitor (Pfizer; Fig. 21) for the potentialtreatment of AD was found to penetrate mouse brainand had a half-life of 6.8 hour in dog (Thomson ReutersPharma, update of April 14, 2011).

A PDE10 inhibitor (biocrea, a spin-out from BioTieTherapeutics following its acquisition of elbion,Fig. 21) is investigated for the potential treatmentof Huntington’s disease. (Thomson Reuters Pharma,update of May 29, 2012).

18F-PF-0430 (Pfizer, Fig. 21) is a PDE2-selectiveCNS PET ligand for the potential diagnosis ofAlzheimer’s disease. (Thomson Reuters Pharma,update of October 03, 2012).

PF-999 (Pfizer, Fig. 21) is a PDE2A inhibitor for thepotential treatment of cognitive impairment associatedwith schizophrenia. (Thomson Reuters Pharma, updateof September 6, 2012).

Sildenafil (Pfizer, launched as Viagra in 1998) andTadalafil (Lilly launched as Cialis in 2003), potentPDE5 inhibitors, restored cognitive function withoutaffecting A� burden in a mouse model of AD. Tadalafilreversed cognitive dysfunction in a mouse model of AD[1545, 1546].

Page 44: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

590 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

Fig. 21. Ten phosphodiesterase inhibitors and a phospholipase A2 inhibitor.

THPP-1 (Merck, Fig. 21) is a potent, orally bioavail-able PDE10A inhibitor, which improved episodic-likememory in rats and executive function in Rhesus mon-keys [1547].

VP1.15 (CSIC Madrid and University of Toronto,Fig. 18) is a dual GSK-3 and PDE7 inhibitor acting as

an antipsychotic and cognitive enhancer in C57BL/6Jmice [1276, 1277].

The development of many PDE inhibitors wasterminated, of D-159687 and DG-071 (deCODEgenetics, now DGI Resolution), denbufylline (BRL-30892; SmithKline Beecham, now GSK [1548]),

Page 45: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 591

HT-0712 (IPL-455903; Helicon under license fromOrexo; a PDE4 inhibitor, Fig. 21 [1549, 1550]), IC-041(Intra-Cellular Therapies), KS-505a (Kyowa HakkoKirin), of MEM-1018, MEM-1091, MEM-1414(R-1533) and MEM-1917 (Memory Pharmaceu-ticals, now Roche), MK-0952 (Merck, a selectivePDE4 inhibitor [1551]), NVP-ABE171 (Novartis[1552–1554]), PF-4181366 and PF-04447943 (Pfizer;a PDE9A inhibitor [1555, 1556]), PQ-10 (Pfizer),propentofylline (HWA 285; Hoechst Werke Albert,now sanofi [1327–1329, 1331, 1332, 1557, 1558]),R-1627 (Roche), rolipram (Meiji Seika under licensefrom Bayer Schering Pharma [1521, 1559–1566]),V-11294A (Napp Pharmaceutical Group) andYF-012403 (Columbia University, a PDE5 inhibitor).

2.30. Drugs interacting with Phospholipase A2and D2

Phospholipase A2 reduction ameliorated cognitivedeficits in a mouse model of AD [1567–1569]. Thesefindings were confirmed by genetic phospholipase A2and D2 ablation studies [1570–1572].

Rilapladib (SB-659032; GSK using a technol-ogy licensed from Human Genome Science; Fig. 21)is a lipoprotein-associated phospholipase A2 (LP-PLA2) inhibitor for the potential oral treatment ofatherosclerosis and AD. In October 2011 a random-ized, double-blind, placebo-controlled Phase II studywas initiated in 120 AD patients with evidence ofcardiovascular disease in Europe. The study was sched-uled to complete in November 2012 (Thomson ReutersPharma, update of June 26, 2012).

Icosapent ethyl (AMR-101, SC-111, LAX-101;MND-21; Epadel; ethyl-eicosapentaenoic acid;Mochida Pharmaceutical and Amarin under licensefrom Scotia Holdings), a phospholipase A2 inhibitor,was launched in Japan for the treatment of arte-riosclerosis obliterans. The drug was in Phase IIaclinical trials for the treatment of AD by Mochida,which were completed in May 2007 and in Phase IIaclinical trials for the treatment of memory disordersin age-associated memory impairment in the UK byAmarin, which started in May 2008. By 2011 thedevelopment for age-associated memory impairmentwas discontinued (Thomson Reuters Pharma, updateof August 29, 2012).

2.31. Drugs interacting with plasminogenactivator inhibitor (PAI)

PAI-1 (serpin E1) is a serine protease inhibitorthat functions as the principal inhibitor of tissue

plasminogen activator (tPA) and urokinase, the acti-vators of plasminogen and hence fibrinolysis [1573].Elevated serum PA1-1 was associated with a highrisk for cognitive dysfunction [1574]. PAI-1 promotedsynaptogenesis and protected against A�1-42-inducedneurotoxicity [7]. PAI-1 was also related to lowerspeed and visuomotor coordination in elderly subjects[1575]. PAI-1 may be a useful marker for vasculardementia [1576]. The plasmin system of AD patientswas discussed [1577].

IMD-4482 (Institute of Medicinal MolecularDesign, IMMD) is an inhibitor of PAI-1 for the poten-tial treatment of fibrotic diseases. The indication ADis no longer followed up (Thomson Reuters Pharma,update of January 19, 2012).

The development of the PAI aleplasinin (PAZ-417;Wyeth, now Pfizer) was terminated.

2.32. Drugs interacting with poly ADP-ribosepolymerase (PARP)

Cognitive impairment can be prevented by PARP-1 inhibitors administered after hypoglycemia [1578,1579]. Cognitive and motor deficits were reduced inmice deficient in PARP-1 [1580].

E-7016 (Eisai following the acquisition of MGIPharma, formerly Guilford; presumed to be GPI-21016under license from Johns Hopkins University, Fig. 22)is an orally active, brain-penetrable, PARP PAR syn-thase inhibitor currently in Phase II clinical trialsfor the treatment of stage III/IV melanoma patients[1581]. The indications AD and stroke were abandoned(Thomson Reuters Pharma, update of June 15,2012).

MP-124 (Mitsubishi Tanabe Pharma) in a PARPinhibitor for the potential treatment of acute ischemicstroke in Phase I clinical trials since December 2008(Thomson Reuters Pharma, update of February 04,2011). The structure was not communicated.

AL-309 (Allon Therapeutics), a peptide, is a neu-roprotective agent and PARP stimulator in preclinicalstudies (Thomson Reuters Pharma, update of May 9,2012).

2.33. Drugs interacting with prolyl endo peptidase

Several reviews on prolyl endo peptidase, alsoknown as prolyl oligopeptidase, as a potential targetfor the treatment of cognitive disorders were published[1582–1584]. Sustained efforts over many years havegone into the discovery and characterization of potentprolyl endopeptidase inhibitors for the treatment of

Page 46: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

592 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

memory disorders [1585]. Potent prolyl endopeptidaseinhibitors, such as KYP-2047 and JTP-4819, did notincrease striatal dopamine, acetylcholine, neurotensinand substance P levels [1586–1588].

Subchronic administration of rosmarinic acid, anatural prolyl oligopeptidase inhibitor, enhanced cog-nitive performances [1589].

The development of many prolyl endopeptidaseinhibitors was terminated, of eurystatin A (Bristol-Myers Squibb [1590–1593]), JTP-4819 (JapanTobacco [1594–1597]), KYP-2047 (Univ. of Kuopioand Finncovery [1598]), ONO-1603 (Ono Pharmaceu-ticals [1599, 1600]), S-17092 (Servier [1601–1605])and S-19825 (Servier [1606]), SUAM-1221 (Chinoinand sanofi [1598, 1607–1610]), Y-29794 (Yoshit-omi, now Mitsubishi Pharma Corp. [1611, 1612]),Z-321 (Zeria Pharmaceutical [1613–1615]) and ZTTA[1616].

2.34. Drugs interacting with prostaglandin D & Esynthases

Reviews on the biochemical, structural, genetic,physiological, and patho-physiological features oflipocalin-type prostaglandin D synthase were dis-closed [1617, 1618]. The three prostaglandin Esynthases were described [1619] as was prostaglandinE2 synthase inhibition as a therapeutic target [1620]and the COX-1 mediated prostaglandin E2 elevationand contextual memory impairment [1621]. Deletionof microsomal prostaglandin E synthase-1 protectedneuronal cells from the cytotoxic effects of A�[1622].

HF-0220 (7-beta-hydroxy-epiandrosterone; New-ron Pharmaceuticals; Fig. 22) is a cytoprotectivesteroid, which stimulated prostaglandin D synthasefor the potential treatment of AD. A Phase IIa mul-ticenter, double-blind, placebo-controlled, biomarkertrial (NCT00357357) in AD patients was initiated inApril 2007. The randomized, double-blind, placebo-controlled pilot study enrolled 42 patients in the UK,Sweden, and India. Subjects received 1 to 220 mg/dayof the drug and were allowed to continue with theircurrent AD treatment. Results were reported in Octo-ber 2008. HF-0220 was well tolerated at all doses(Thomson Reuters Pharma, update of August 21,2012).

AAD-2004 (GNT Pharma, formerly Neurotech andUS subsidiary AmKor, Fig. 22) is a prostaglandin Esynthase-1 inhibitor for the treatment of AD, Parkin-son’s disease, and amyotrophic lateral sclerosis. APhase I study was initiated in April 2010. By December

2011, the Phase Ia single ascending dose trial had beensuccessfully completed. In April 2012, it was reportedthat a Phase Ib multiple ascending-dose trial to eval-uate safety and biomarker profiles was planned forthat year (Thomson Reuters Pharma, update of May 4,2012).

The development of an analogue of AAD-2004, i.e.,NG-2006 (GNT Pharma) was terminated.

2.35. Drugs interacting with protein kinase C

Activation of PKC� prevents synaptic loss, A� ele-vation, and cognitive deficits in AD transgenic mice[1623, 1624]. The pharmacology of PKC activatorswas extensively reviewed [1625–1629].

APH-0703 (Aphios Corp.), a potent PKC activa-tor, enhanced the generation of non-amyloidogenic,soluble A�PP in fibroblasts from AD patients. Itreduced brain amyloid plaques (A�40 and A�42) indouble-transgenic mice. The drug, formulated usingAphios’s hydrophobic-based and SFS-PNS polymernanospheres technologies, is in Phase II clinical trialssince May 2010 (Thomson Reuters Pharma, update ofJuly 5, 2012). The structure was not communicated.Potent activators of PKC are indirect activators of �-secretase [1630–1632].

Bryostatin-1 (Blanchette Rockefeller Neuro-sciences Institute) is a naturally occurring PKCactivator isolated from the Californian marinebryozoan Bugula neritina. In February 2012, theInstitute was preparing to initiate clinical trials inneurological disorders. Dual effects of bryostatin-1on spatial memory and depression were described[1633–1635]. Postischemic PKC activation to rescuelong-term memory was investigated [1636, 1637]. Thechemistry and biology of bryostatins was reviewed[504] (Thomson Reuters Pharma, update of February24, 2012). See also Section 2.2. Drugs interactingwith �-secretase.

DCP-LA (Hyogo College of Medicine; Fig. 22)is an activator of PKC� for the potential treatmentof stroke and cognitive disorder (Thomson ReutersPharma, update of May 25, 2012).

PKC epsilon activators (Blanchette RockefellerNeurosciences Institute) are evaluated for the poten-tial treatment of cognitive disorder (Thomson ReutersPharma, update of February 17, 2012). The structureswere not communicated.

The development of FR-236924 (Fujisawa, nowAstellas; an activator of PKC� [1638–1641]) and ofK-252c (Kyowa Hakko Kogyo; a PKC inhibitor) wasterminated.

Page 47: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 593

Fig. 22. A poly ADP-ribose polymerase inhibitor, prostaglandin D and E modulators, a protein kinase C � activator, a Rac1 GTPase inhibitorand a ras farnesyl transferase inhibitor.

2.36. Drugs interacting with protein tyrosinephosphatase

The role of striatal-enriched protein tyrosine phos-phatase in cognition was described [1642] as was theinvolvement of PTPN5, the gene encoding the striatal-enriched protein tyrosine phosphatase in schizophreniaand cognition [1643]. A genome-wide associationstudy in 700 schizophrenic patients found that threeintronic SNPs in the protein tyrosine phosphatasereceptor type O were associated with learning andmemory [1644]. Knockout mice revealed a role forprotein tyrosine phosphatase in cognition [1645].

LDN-33960 (Yale University) is a striatal-enrichedprotein tyrosine phosphatase inhibitor, whichincreased the phosphorylation of NMDA receptorsand of ERK in a dose dependent manner. The drugrestored memory in a novel object recognition test intriple transgenic mice. It appears that the developmentof LDN-33960 was terminated (Thomson ReutersPharma, update of June 19, 2012).

2.37. Drugs interacting with Rac1 GTPase

The role of Rac1 GTPase in cognitive impairmentfollowing cerebral ischemia in the rat was described[1646]. The modulation of synaptic function by Rac1may be a possible link to fragile X syndrome pathology[1647, 1648].

Cytotoxic Necrotizing Factor 1, a modulatorof Rho GTPases including Rac, Rho, and Cdc42subfamilies improved object recognition in mice[1649].

Sanquinarinium chloride (ExonHit; Fig. 22) is anselective Rac1/1b GTPase nucleotide binding inhibitorwith IC50’s of 58 �M and 4.6 �M for Rac1 and Rac1b,respectively in nucleotide binding inhibition assays inpreclinical development (Thomson Reuters Pharma,update of July 20, 2011).

The development of EHT-1864 (EHT-206, EHT-101; ExonHit), an orally active small molecule Rac1GTPase inhibitor capable of crossing the blood brainbarrier was discontinued [1650–1652].

Page 48: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

594 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

Fig. 23. A S-adenosylhomocysteine hydrolase and a sirtuin inhibitor, a sirtuin activator, a steroid sulfatase and a transglutaminase 2 inhibitor.

2.38. Drugs interacting with Ras FarnesylTransferase

Inhibiting farnesylation, but not geranylgeranyla-tion, replicated the enhancement of LTP caused bysimvastatin via the activation of Akt [1653].

LNK-754 (OSI-754, CP-609754; AstraZenecafollowing the acquisition of Link Medicine’s neuro-science assets under license from OSI Pharmaceu-ticals, a subsidiary of Astellas Pharma, and Pfizer;Fig. 22) is an orally active inhibitor of ras farnesyltransferase. Two Phase I clinical trials were initiatedin the US, one in May 2009 in 40 healthy elderlysubjects and one in November 2009 in 110 healthyelderly subjects and in patients with mild AD. Treat-ment of transgenic mice at 6 months of age for 3 monthsshowed a clear reduction of plaques caused by either�-synuclein or A� [1654] (Thomson Reuters Pharma,update of July 13, 2012).

2.39. Drugs interacting withS-adenosylhomocysteine hydrolase

Homocysteine potentiated A� neurotoxicity [1655].The S-adenosyl homocysteine hydrolase inhibitor

3-deaza-adenosine prevented cognitive impairmentfollowing folate and vitamin E deprivation in mice[1656].

L-002259713 (Merck, Fig. 23) is a potent inhibitorof S-adenosylhomocysteine hydrolase for the potentialtreatment of AD. The development was discontinued(Thomson Reuters Pharma, update of June 19,2012).

2.40. Drugs interacting with sirtuin

The neuronal protection by sirtuins in AD wasdescribed [1657] as were details on the sirtuin pathwayin aging and AD [1658].

Resveratrol (Fig. 23) is a natural phyto compound,which activates Sirtuin-1 [1659–1664]. It reducedA� accumulation [1665–1668]. It remodeled solu-ble oligomers and fibrils of A� into off-pathwayconformers [1669]. Resveratrol is not a direct acti-vator of SIRT1 enzyme activity [1670]. Resveratrolimproved memory deficits in mice fed a high-fat diet[1671]. Subchronic oral toxicity and cardiovascularsafety pharmacology studies were carried out [1672].The biosynthesis of resveratrol in yeast and in mam-malian cells was worked out [1673]. The Georgetown

Page 49: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 595

University Medical Center started at Phase II, ran-domized, double-blind, placebo-controlled study inpatients with mild to moderate AD (expected n = 120)in the US in May 2012 (NCT015048549). A novelapplication in Huntington’s disease was discussedrecently [1674] (Thomson Reuters Pharma, update ofJune 22, 2012).

Selisistat (SEN-196, EX-527, SEN-0014196; SienaBiotech under license from Elixir Pharmaceuticals;Fig. 23) is a sirtuin-1 inhibitor for the potentialtreatment of Huntington’s disease in Phase II tri-als (PADDINGTON, NCT01485965) in Europe sinceApril 2011 (Thomson Reuters Pharma, update of May29, 2012).

INDUS-815C (Indus Biotech) is a natural NAD-dependent deacetylase sirtuin-2 (SIRT-2) inhibitorfor the potential treatment of Huntington’s disease.In addition, the drug is evaluated for the potentialtreatment of age-related macular degeneration andretinopathy (Thomson Reuters Pharma, updates ofAugust 19, 2011 and August 16, 2011, respectively).The structure was not communicated.

2.41. Drugs interacting with steroid sulfatase

Steroid sulfatase is a potential modifier of cog-nition in attention deficit hyperactivity disorder[1675].

DU-14 (Dusquesne University, Fig. 23) is asteroid sulfatase inhibitor for the potential enhance-ment of cognitive function via enhanced levels ofplasma dehydro-epiandrosterone and brain acetyl-choline [1676–1679]. Its development was terminated.

2.42. Drugs interacting with transglutaminase(TG2)

Tissue transglutaminase catalyzes protein cross-linking, an important molecular process in AD.Accumulation of insoluble proteins with isopeptidebonds, products of tissue transaminase activity, cor-related with cognitive impairment [1680].

CHDI-00339864 (Fig. 23) and CHDI-00316226(Evotec in collaboration with the CHDI Founda-tion) are selective TG2 inhibitors (IC50 = 7 nM forTG2 for CHDI-00316226) for the potential treat-ment of Huntington’s disease. It displayed an 85fold greater selectivity for TG2 over Factor XIIIA(Thomson Reuters Pharma, update of February 10,2012).

2.43. Drugs interacting with ubiquitincarboxyl-terminal hydroxylase (Usp14)

Inhibitors of this enzyme could accelerate the degra-dation of neurodegenerative disease-related proteins,such as tau, TDP-43, and ataxin-3. Changes in hip-pocampal synaptic transmission were observed inUsp14-deficient mice [1681].

Usp14 inhibitors (Proteostasis Therapeutics underlicense from Harvard University) are investigated forthe potential treatment of neurodegenerative diseases(Thomson Reuters Pharma, update of August 15,2012). Structures were not communicated.

3. CONCLUSION

With the launch of donepezil (Aricept) in 1996,rivastigmine (Exelon) in 2000, and galantamine(Reminyl) in 2001 (all inhibitors of AChE andBChE), three valuable medications are available forthe treatment of patients with mild to moderate AD.Huperzine A was launched in China in 1995, but thefour times a day administration makes it less attractive,a problem which will be solved by a transdermal patchcurrently in Phase I evaluation as XEL-001HP by XelPharmaceuticals.

The development of ten Phase III compounds testedfor the indication AD was terminated, of the AChEinhibitors amiridin (Nikken), eptastigmine (Medi-olanum), metrifonate (Bayer), phenserine (Anonyx),and velnacrine (HMR, now sanofi), of the �-secretaseinhibitors begacestat (Pfizer) and semagacestat (Lilly),the �-secretase modulator tarenflurbil (Myriad Genet-ics), and of the MAO inhibitors rasagiline (Teva) andsafinamide (Merck Serono).

Currently there is one compound interacting withenzymes in the pre-registration phase (WIN-026,WhanIN, an AChE inhibitor), one compound inPhase III trials (masitinib, AB Science, a kinaseinhibitor), and 15 drugs in Phase II evaluation(posiphen and Shen Er Yang, two AChE inhibitors),ladostigil (a dual AChE and MAO inhibitor), etazolate(an �-secretase activator), avagacestat and NIC5-15 (�-secretase inhibitors), CHF-5074 (�-secretasemodulator), tideglusib (GSK-3� inhibitor), EVP-0334 (HDAC inhibitor), RG-1577 (MAO-B inhibitor),PF-02545920 (PDE10A inhibitor), rilapladib (PLA2inhibitor), HF-0220 (prostaglandin D synthase stim-ulator), APH-0703 (PKC activator), and selisistat(sirtuin-1 inhibitor).

It may be that positive results will be obtained fromthe masitinib Phase III trials within the next two to

Page 50: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

596 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

three years, whereas results from the Phase II trialswill be available only within the next five to six years.

DISCLOSURE STATEMENT

Authors’ disclosures available online (http://www.j-alz.com/disclosures/view.php?id=1508).

REFERENCES

[1] Giurgea C (1972) [Pharmacology of integrative activity ofthe brain. Attempt at nootropic concept in psychopharma-cology]. Actual Pharmacol (Paris) 25, 115-156.

[2] Giurgea C (1973) The “nootropic” approach to the pharma-cology of the integrative activity of the brain. Cond Reflex8, 108-115.

[3] Millan MJ, Agid Y, Brune M, Bullmore ET, Carter CS,Clayton NS, Connor R, Davis S, Deakin B, DeRubeisRJ, Dubois B, Geyer MA, Goodwin GM, Gorwood P, JayTM, Joels M, Mansuy IM, Meyer-Lindenberg A, MurphyD, Rolls E, Saletu B, Spedding M, Sweeney J, WhittingtonM, Young LJ (2012) Cognitive dysfunction in psychi-atric disorders: Characteristics, causes and the quest forimproved therapy. Nat Rev Drug Discov 11, 141-168.

[4] Froestl W, Maitre L (1989) The families of cognitionenhancers. Pharmacopsychiatry 22(Suppl 2), 54-100.

[5] Froestl W, Muhs A, Pfeifer A (2012) Cognitive enhancers(nootropics). Part 1: Drugs interacting with receptors.J Alzheimers Dis, doi: 10.3233/JAD-2012-121186 [Epublahead of print].

[6] Bartus RT, Dean RL III, Beer B, Lippa AS (1982) Thecholinergic hypothesis of geriatric memory dysfunction.Science 217, 408-414.

[7] Musial A, Bajda M, Malawska B (2007) Recent develop-ments in cholinesterases inhibitors for Alzheimer’s diseasetreatment. Curr Med Chem 14, 2654-2679.

[8] Pepeu G, Giovannini MG (2009) Cholinesterase inhibitorsand beyond. Curr Alzheimer Res 6, 86-96.

[9] Pepeu G, Giovannini MG (2010) Cholinesterase inhibitorsand memory. Chem Biol Interact 187, 403-408.

[10] Dumas JA, Newhouse PA (2011) The cholinergic hypoth-esis of cognitive aging revisited again: Cholinergicfunctional compensation. Pharmacol Biochem Behav 99,254-261.

[11] Schliebs R, Arendt T (2011) The cholinergic system inaging and neuronal degeneration. Behav Brain Res 221,555-563.

[12] Terry AV Jr, Callahan PM, Hall B, Webster SJ (2011)Alzheimer’s disease and age-related memory decline (pre-clinical). Pharmacol Biochem Behav 99, 190-210.

[13] Speck-Planche A, Luan F, Cordeiro MN (2012) Dis-covery of anti-Alzheimer agents: Current ligand-basedapproaches toward the design of acetylcholinesteraseinhibitors. Mini Rev Med Chem 12, 583-591.

[14] Mehta M, Adem A, Sabbagh M (2012) New acetyl-cholinesterase inhibitors for Alzheimer’s disease. Int JAlzheimers Dis 2012, 728983.

[15] Pohanka M (2012) Acetylcholinesterase inhibitors: Apatent review (2008-present). Expert Opin Ther Pat 22,871-886.

[16] Contestabile A (2011) The history of the cholinergichypothesis. Behav Brain Res 221, 334-340.

[17] Colombres M, Sagal JP, Inestrosa NC (2004) An overviewof the current and novel drugs for Alzheimer’s disease withparticular reference to anti-cholinesterase compounds.Curr Pharm Des 10, 3121-3130.

[18] Grutzendler J, Morris JC (2001) Cholinesterase inhibitorsfor Alzheimer’s disease. Drugs 61, 41-52.

[19] Sugimoto H, Iimura Y, Yamanishi Y, Yamatsu K (1995)Synthesis and structure-activity relationships of acetyl-cholinesterase inhibitors: 1-benzyl-4-[(5,6-dimethoxy-1-oxoindan-2-yl)methyl]piperidine hydrochloride andrelated compounds. J Med Chem 38, 4821-4829.

[20] Kryger G, Silman I, Sussman JL (1998) Three-dimensional structure of a complex of E2020 withacetylcholinesterase from Torpedo californica. J PhysiolParis 92, 191-194.

[21] Haginaka J, Seyama C (1992) Determination ofenantiomers of 1-benzyl-4-[(5,6-dimethoxy-1-indanon)-2-yl]methylpiperidine hydrochloride (E2020), a centrallyacting acetylcholine esterase inhibitor, in plasma by liquidchromatography with fluorometric detection. J Chro-matogr 577, 95-102.

[22] Matsui K, Oda Y, Ohe H, Tanaka S, Asakawa N(1995) Direct determination of E2020 enantiomers inplasma by liquid chromatography-mass spectrometry andcolumn-switching techniques. J Chromatogr A 694, 209-218.

[23] Sugimoto H (2008) The new approach in development ofanti-Alzheimer’s disease drugs via the cholinergic hypoth-esis. Chem Biol Interact 175, 204-208.

[24] Rao RJ, Raol AK, Murthy YL (2007) Efficient and indus-trially viable synthesis of donepezil. Synth Commun 37,2847-2853.

[25] Dubey SK, Kharbanda M, Dubey SK, Mathela CS (2010)A new commercially viable synthetic route for donepezilhydrochloride: Anti-Alzheimer’s drug. Chem Pharm Bull(Tokyo) 58, 1157-1160.

[26] Inoue A, Kawai T, Wakita M, Iimura Y, SugimotoH, Kawakami Y (1996) The simulated binding of(+/−)-2,3-dihydro-5,6-dimethoxy-2-[[1-(phenylmethyl)-4-piperidinyl]meth yl] -1H-inden-1-one hydrochloride(E2020) and related inhibitors to free and acylated acetyl-cholinesterases and corresponding structure-activityanalyses. J Med Chem 39, 4460-4470.

[27] Kawakami Y, Inoue A, Kawai T, Wakita M, Sugimoto H,Hopfinger AJ (1996) The rationale for E2020 as a potentacetylcholinesterase inhibitor. Bioorg Med Chem 4, 1429-1446.

[28] Bryson HM, Benfield P (1997) Donepezil. Drugs Aging10, 234-239.

[29] Heydorn WE (1997) Donepezil (E2020): A new acetyl-cholinesterase inhibitor. Review of its pharmacology,pharmacokinetics, and utility in the treatment ofAlzheimer’s disease. Expert Opin Investig Drugs 6, 1527-1535.

[30] Wilkinson DG (1999) The pharmacology of donepezil: Anew treatment of Alzheimer’s disease. Expert Opin Phar-macother 1, 121-135.

[31] Wilkinson DG, Francis PT, Schwam E, Payne-Parrish J(2004) Cholinesterase inhibitors used in the treatment ofAlzheimer’s disease: The relationship between pharma-cological effects and clinical efficacy. Drugs Aging 21,453-478.

[32] Sugimoto H, Yamanishi Y, Iimura Y, Kawakami Y(2000) Donepezil hydrochloride (E2020) and other acetyl-cholinesterase inhibitors. Curr Med Chem 7, 303-339.

Page 51: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 597

[33] Sugimoto H (2001) Donepezil hydrochloride: A treatmentdrug for Alzheimer’s disease. Chem Rec 1, 63-73.

[34] Shigeta M, Homma A (2001) Donepezil for Alzheimer’sdisease: Pharmacodynamic, pharmacokinetic, and clinicalprofiles. CNS Drug Rev 7, 353-368.

[35] Seltzer B (2005) Donepezil: A review. Expert Opin DrugMetab Toxicol 1, 527-536.

[36] Seltzer B (2007) Donepezil: An update. Expert Opin Phar-macother 8, 1011-1023.

[37] Jacobson SA, Sabbagh MN (2008) Donepezil: Poten-tial neuroprotective and disease-modifying effects. ExpertOpin Drug Metab Toxicol 4, 1363-1369.

[38] Kapai NA, Solntseva EI, Skrebitskii VG (2010) Donepezileliminates suppressive effects of beta-amyloid peptide(1–42) on long-term potentiation in the hippocampus. BullExp Biol Med 149, 33-36.

[39] Kapai NA, Bukanova JV, Solntseva EI, Skrebitsky VG(2012) Donepezil in a narrow concentration range aug-ments control and impaired by beta-amyloid peptidehippocampal LTP in NMDAR-independent manner. CellMol Neurobiol 32, 219-226.

[40] Freret T, Bouet V, Quiedeville A, Nee G, DallemagneP, Rochais C, Boulouard M (2012) Synergistic effect ofacetylcholinesterase inhibition (donepezil) and 5-HT(4)receptor activation (RS67333) on object recognition inmice. Behav Brain Res 230, 304-308.

[41] Gomolin IH, Smith C, Jeitner TM (2011) Donepezil dosingstrategies: Pharmacokinetic considerations. J Am Med DirAssoc 12, 606-608.

[42] Doody RS (1999) Clinical profile of donepezil in the treat-ment of Alzheimer’s disease. Gerontology 45(Suppl 1),23-32.

[43] Doody RS, Dunn JK, Clark CM, Farlow M, Foster NL,Liao T, Gonzales N, Lai E, Massman P (2001) Chronicdonepezil treatment is associated with slowed cognitivedecline in Alzheimer’s disease. Dement Geriatr CognDisord 12, 295-300.

[44] Doody RS (2003) Current treatments for Alzheimer’s dis-ease: Cholinesterase inhibitors. J Clin Psychiatry 64(Suppl9), 11-17.

[45] Dooley M, Lamb HM (2000) Donepezil: A review of itsuse in Alzheimer’s disease. Drugs Aging 16, 199-226.

[46] Meyer JS, Chowdhury MH, Xu G, Li YS, Quach M (2002)Donepezil treatment of vascular dementia. Ann N Y AcadSci 977, 482-486.

[47] Roman GC, Rogers SJ (2004) Donepezil: A clinical reviewof current and emerging indications. Expert Opin Pharma-cother 5, 161-180.

[48] Whitehead A, Perdomo C, Pratt RD, Birks J, Wilcock GK,Evans JG (2004) Donepezil for the symptomatic treatmentof patients with mild to moderate Alzheimer’s disease: Ameta-analysis of individual patient data from randomisedcontrolled trials. Int J Geriatr Psychiatry 19, 624-633.

[49] Takeda A, Loveman E, Clegg A, Kirby J, Picot J, Payne E,Green C (2006) A systematic review of the clinical effec-tiveness of donepezil, rivastigmine and galantamine oncognition, quality of life and adverse events in Alzheimer’sdisease. Int J Geriatr Psychiatry 21, 17-28.

[50] Hansen RA, Gartlehner G, Webb AP, Morgan LC,Moore CG, Jonas DE (2008) Efficacy and safety ofdonepezil, galantamine, and rivastigmine for the treatmentof Alzheimer’s disease: A systematic review and meta-analysis. Clin Interv Aging 3, 211-225.

[51] Raina P, Santaguida P, Ismaila A, Patterson C, CowanD, Levine M, Booker L, Oremus M (2008) Effectiveness

of cholinesterase inhibitors and memantine for treatingdementia: Evidence review for a clinical practice guide-line. Ann Intern Med 148, 379-397.

[52] Sabbagh MN, Richardson S, Relkin N (2008) Disease-modifying approaches to Alzheimer’s disease: Chal-lenges and opportunities-Lessons from donepezil therapy.Alzheimers Dement 4, S109-S118.

[53] Rodda J, Morgan S, Walker Z (2009) Are cholinesteraseinhibitors effective in the management of the behav-ioral and psychological symptoms of dementia inAlzheimer’s disease? A systematic review of randomized,placebo-controlled trials of donepezil, rivastigmine andgalantamine. Int Psychogeriatr 21, 813-824.

[54] Tsuno N (2009) Donepezil in the treatment of patients withAlzheimer’s disease. Expert Rev Neurother 9, 591-598.

[55] Winblad B, Black SE, Homma A, Schwam EM, Moline M,Xu Y, Perdomo CA, Swartz J, Albert K (2009) Donepeziltreatment in severe Alzheimer’s disease: A pooled analysisof three clinical trials. Curr Med Res Opin 25, 2577-2587.

[56] Cummings J, Jones R, Wilkinson D, Lopez O, GauthierS, Waldemar G, Zhang R, Xu Y, Sun Y, Richardson S,Mackell J (2010) Effect of donepezil on cognition in severeAlzheimer’s disease: A pooled data analysis. J AlzheimersDis 21, 843-851.

[57] Rosenblatt A, Gao J, Mackell J, Richardson S (2010)Efficacy and safety of donepezil in patients withAlzheimer’s disease in assisted living facilities. Am JAlzheimers Dis Other Demen 25, 483-489.

[58] Santoro A, Siviero P, Minicuci N, Bellavista E, MishtoM, Olivieri F, Marchegiani F, Chiamenti AM, Benussi L,Ghidoni R, Nacmias B, Bagnoli S, Ginestroni A, ScarpinoO, Feraco E, Gianni W, Cruciani G, Paganelli R, Di IA,Scognamiglio M, Grimaldi LM, Gabelli C, Sorbi S, BinettiG, Crepaldi G, Franceschi C (2010) Effects of donepezil,galantamine and rivastigmine in 938 Italian patients withAlzheimer’s disease: A prospective, observational study.CNS Drugs 24, 163-176.

[59] Lockhart IA, Orme ME, Mitchell SA (2011) The efficacyof licensed-indication use of donepezil and memantinemonotherapies for treating behavioural and psychologi-cal symptoms of dementia in patients with Alzheimer’sdisease: Systematic review and meta-analysis. DementGeriatr Cogn Dis Extra 1, 212-227.

[60] Sabbagh M, Cummings J (2011) Progressive cholinergicdecline in Alzheimer’s Disease: Consideration fortreatment with donepezil 23 mg in patients with moderateto severe symptomatology. BMC Neurol 11, 21.

[61] Waldemar G, Gauthier S, Jones R, Wilkinson D, Cum-mings J, Lopez O, Zhang R, Xu Y, Sun Y, Knox S,Richardson S, Mackell J (2011) Effect of donepezil onemergence of apathy in mild to moderate Alzheimer’s dis-ease. Int J Geriatr Psychiatry 26, 150-157.

[62] Bottini G, Berlingeri M, Basilico S, Passoni S, Danelli L,Colombo N, Sberna M, Franceschi M, Sterzi R, Paulesu E(2012) GOOD or BAD responder? Behavioural and neu-roanatomical markers of clinical response to donepezil indementia. Behav Neurol 25, 61-72.

[63] Doody RS, Geldmacher DS, Farlow MR, Sun Y, MolineM, Mackell J (2012) Efficacy and safety of donepezil23 mg versus donepezil 10 mg for moderate-to-severeAlzheimer’s disease: A subgroup analysis in patientsalready taking or not taking concomitant memantine.Dement Geriatr Cogn Disord 33, 164-173.

[64] Howard R, McShane R, Lindesay J, Ritchie C, BaldwinA, Barber R, Burns A, Dening T, Findlay D, Holmes

Page 52: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

598 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

C, Hughes A, Jacoby R, Jones R, Jones R, McKeithI, Macharouthu A, O’Brien J, Passmore P, Sheehan B,Juszczak E, Katona C, Hills R, Knapp M, Ballard C, BrownR, Banerjee S, Onions C, Griffin M, Adams J, Gray R,Johnson T, Bentham P, Phillips P (2012) Donepezil andmemantine for moderate-to-severe Alzheimer’s disease. NEngl J Med 366, 893-903.

[65] Burke D (2012) Donepezil or memantine improved cogni-tive functioning in moderate-to-severe Alzheimer disease.Ann Intern Med 156, JC6-JC10.

[66] Rodda J, Carter J (2012) Cholinesterase inhibitors andmemantine for symptomatic treatment of dementia. BMJ344, e2986.

[67] Berk C, Sabbagh M (2012) Broader considerations ofhigher doses of donepezil in the treatment of mild, moder-ate, and severe Alzheimer’s disease. Int J Alzheimers Dis2012, 707468.

[68] Schwartz LM, Woloshin S (2012) How the FDA forgot theevidence: The case of donepezil 23 mg. BMJ 344, e1086.

[69] Darreh-Shori T, Soininen H (2010) Effects ofcholinesterase inhibitors on the activities and pro-tein levels of cholinesterases in the cerebrospinal fluidof patients with Alzheimer’s disease: A review of recentclinical studies. Curr Alzheimer Res 7, 67-73.

[70] Jackson S, Ham RJ, Wilkinson D (2004) The safety and tol-erability of donepezil in patients with Alzheimer’s disease.Br J Clin Pharmacol 58(Suppl 1), 1-8.

[71] Doody RS, Corey-Bloom J, Zhang R, Li H, Ieni J,Schindler R (2008) Safety and tolerability of donepezilat doses up to 20 mg/day: Results from a pilot study inpatients with Alzheimer’s disease. Drugs Aging 25, 163-174.

[72] Lockhart IA, Mitchell SA, Kelly S (2009) Safety and tol-erability of donepezil, rivastigmine and galantamine forpatients with Alzheimer’s disease: Systematic review ofthe ‘real-world’ evidence. Dement Geriatr Cogn Disord28, 389-403.

[73] Foster RH, Plosker GL (1999) Donepezil. Pharmacoeco-nomic implications of therapy. Pharmacoeconomics 16,99-114.

[74] Wolfson C, Oremus M, Shukla V, Momoli F, Demers L,Perrault A, Moride Y (2002) Donepezil and rivastigminein the treatment of Alzheimer’s disease: A best-evidencesynthesis of the published data on their efficacy and cost-effectiveness. Clin Ther 24, 862-886.

[75] Wimo A, Winblad B, Engedal K, Soininen H, VerheyF, Waldemar G, Wetterholm AL, Mastey V, HaglundA, Zhang R, Miceli R, Chin W, Subbiah P (2003) Aneconomic evaluation of donepezil in mild to moderateAlzheimer’s disease: Results of a 1-year, double-blind,randomized trial. Dement Geriatr Cogn Disord 15,44-54.

[76] Cappell J, Herrmann N, Cornish S, Lanctot KL (2010) Thepharmacoeconomics of cognitive enhancers in moderate tosevere Alzheimer’s disease. CNS Drugs 24, 909-927.

[77] Getsios D, Blume S, Ishak KJ, Maclaine GD (2010) Costeffectiveness of donepezil in the treatment of mild tomoderate Alzheimer’s disease: A UK evaluation usingdiscrete-event simulation. Pharmacoeconomics 28, 411-427.

[78] Bond M, Rogers G, Peters J, Anderson R, Hoyle M, MinersA, Moxham T, Davis S, Thokala P, Wailoo A, Jeffreys M,Hyde C (2012) The effectiveness and cost-effectivenessof donepezil, galantamine, rivastigmine and memantinefor the treatment of Alzheimer’s disease (review of Tech-

nology Appraisal No. 111): A systematic review andeconomic model. Health Technol Assess 16, 1-470.

[79] Versijpt J (2012) Pharmacoeconomics of Alzheimer’s dis-ease (AD) treatment with cholinesterase inhibitors. ActaNeurol Belg 112, 141-145.

[80] Tricco AC, Soobiah C, Lillie E, Perrier L, Chen MH,Hemmelgarn B, Majumdar SR, Straus SE (2012) Use ofcognitive enhancers for mild cognitive impairment: Pro-tocol for a systematic review and network meta-analysis.Syst Rev 1, 25.

[81] Tricco AC, Vandervaart S, Soobiah C, Lillie E, PerrierL, Straus SE (2012) Efficacy of cognitive enhancers forAlzheimer’s disease Protocol for a systematic review andnetwork meta-analysis. Syst Rev 1, 31.

[82] Kishnani PS, Sommer BR, Handen BL, Seltzer B, CaponeGT, Spiridigliozzi GA, Heller JH, Richardson S, McRaeT (2009) The efficacy, safety, and tolerability of donepezilfor the treatment of young adults with Down syndrome.Am J Med Genet A 149A 1641-1654.

[83] Kondoh T, Kanno A, Itoh H, Nakashima M, Honda R,Kojima M, Noguchi M, Nakane H, Nozaki H, Sasaki H,Nagai T, Kosaki R, Kakee N, Okuyama T, Fukuda M,Ikeda M, Shibata Y, Moriuchi H (2011) Donepezil sig-nificantly improves abilities in daily lives of female Downsyndrome patients with severe cognitive impairment: A 24-week randomized, double-blind, placebo-controlled trial.Int J Psychiatry Med 41, 71-89.

[84] Kishnani PS, Heller JH, Spiridigliozzi GA, Lott I, EscobarL, Richardson S, Zhang R, McRae T (2010) Donepezil fortreatment of cognitive dysfunction in children with Downsyndrome aged 10-17. Am J Med Genet A 152A 3028-3035.

[85] Ravina B, Putt M, Siderowf A, Farrar JT, Gillespie M,Crawley A, Fernandez HH, Trieschmann MM, ReichweinS, Simuni T (2005) Donepezil for dementia in Parkinson’sdisease: A randomised, double blind, placebo controlled,crossover study. J Neurol Neurosurg Psychiatry 76, 934-939.

[86] Rowan E, McKeith IG, Saxby BK, O’Brien JT, Burn D,Mosimann U, Newby J, Daniel S, Sanders J, Wesnes K(2007) Effects of donepezil on central processing speedand attentional measures in Parkinson’s disease withdementia and dementia with Lewy bodies. Dement GeriatrCogn Disord 23, 161-167.

[87] van Laar T, De Deyn PP, Aarsland D, Barone P, Galvin JE(2011) Effects of cholinesterase inhibitors in Parkinson’sdisease dementia: A review of clinical data. CNS NeurosciTher 17, 428-441.

[88] Krupp LB, Christodoulou C, Melville P, Scherl WF, PaiLY, Muenz LR, He D, Benedict RH, Goodman A, RizviS, Schwid SR, Weinstock-Guttman B, Westervelt HJ,Wishart H (2011) Multicenter randomized clinical trial ofdonepezil for memory impairment in multiple sclerosis.Neurology 76, 1500-1507.

[89] Tariot PN, Farlow MR, Grossberg GT, Graham SM,McDonald S, Gergel I (2004) Memantine treatment inpatients with moderate to severe Alzheimer disease alreadyreceiving donepezil: A randomized controlled trial. JAMA291, 317-324.

[90] Bar-On P, Millard CB, Harel M, Dvir H, Enz A, SussmanJL, Silman I (2002) Kinetic and structural studies on theinteraction of cholinesterases with the anti-Alzheimer drugrivastigmine. Biochemistry 41, 3555-3564.

[91] Darvesh S, Hopkins DA, Geula C (2003) Neurobiology ofbutyrylcholinesterase. Nat Rev Neurosci 4, 131-138.

Page 53: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 599

[92] Amstutz R, Enz A, Marzi M, Boelsterli J, WalkinshawM (1990) Cyclische phenylcarbamate des miotin-typs undihre wirkung auf die acetylcholinesterase. Helv Chim Acta73, 739-753.

[93] Boezio AA, Pytkowicz J, Cote A, Charette AB (2003)Asymmetric, catalytic synthesis of alpha-chiral aminesusing a novel bis(phosphine) monoxide chiral ligand.J Am Chem Soc 125, 14260-14261.

[94] Hu M, Zhang FL, Xie MH (2009) Novel convenient syn-thesis of rivastigmine. Synth Commun 39, 1527-1533.

[95] Mangas-Sanchez J, Rodriguez-Mata M, Busto E, Gotor-Fernandez V, Gotor V (2009) Chemoenzymatic synthesisof rivastigmine based on lipase-catalyzed processes. J OrgChem 74, 5304-5310.

[96] Han K, Kim C, Park J, Kim MJ (2010) Chemoenzymaticsynthesis of rivastigmine via dynamic kinetic resolution asa key step. J Org Chem 75, 3105-3108.

[97] Enz A, Boddeke H, Gray J, Spiegel R (1991) Pharmaco-logic and clinicopharmacologic properties of SDZ ENA713, a centrally selective acetylcholinesterase inhibitor.Ann N Y Acad Sci 640, 272-275.

[98] Enz A, Amstutz R, Boddeke H, Gmelin G, Malanowski J(1993) Brain selective inhibition of acetylcholinesterase:A novel approach to therapy for Alzheimer’s disease. ProgBrain Res 98, 431-438.

[99] Weinstock M, Razin M, Chorev M, Enz A (1994)Pharmacological evaluation of phenyl-carbamates asCNS-selective acetylcholinesterase inhibitors. J NeuralTransm Suppl 43, 219-225.

[100] Gottwald MD, Rozanski RI (1999) Rivastigmine, a brain-region selective acetylcholinesterase inhibitor for treatingAlzheimer’s disease: Review and current status. ExpertOpin Investig Drugs 8, 1673-1682.

[101] Williams BR, Nazarians A, Gill MA (2003) A review ofrivastigmine: A reversible cholinesterase inhibitor. ClinTher 25, 1634-1653.

[102] Bailey JA, Ray B, Greig NH, Lahiri DK (2011) Rivastig-mine lowers Abeta and increases sAPPalpha levels, whichparallel elevated synaptic markers and metabolic activ-ity in degenerating primary rat neurons. PLoS One 6,e21954.

[103] Polinsky RJ (1998) Clinical pharmacology of rivastig-mine: A new-generation acetylcholinesterase inhibitor forthe treatment of Alzheimer’s disease. Clin Ther 20, 634-647.

[104] Spencer CM, Noble S (1998) Rivastigmine. A reviewof its use in Alzheimer’s disease. Drugs Aging 13, 391-411.

[105] Forette F, Anand R, Gharabawi G (1999) A phase II studyin patients with Alzheimer’s disease to assess the prelimi-nary efficacy and maximum tolerated dose of rivastigmine(Exelon). Eur J Neurol 6, 423-429.

[106] Robert P (2002) Understanding and managing behaviouralsymptoms in Alzheimer’s disease and related dementias:Focus on rivastigmine. Curr Med Res Opin 18, 156-171.

[107] Farlow MR (2003) Update on rivastigmine. Neurologist 9,230-234.

[108] Gabelli C (2003) Rivastigmine: An update on therapeuticefficacy in Alzheimer’s disease and other conditions. CurrMed Res Opin 19, 69-82.

[109] Finkel SI (2004) Effects of rivastigmine on behavioraland psychological symptoms of dementia in Alzheimer’sdisease. Clin Ther 26, 980-990.

[110] Desai AK, Grossberg GT (2005) Rivastigmine forAlzheimer’s disease. Expert Rev Neurother 5, 563-580.

[111] Onor ML, Trevisiol M, Aguglia E (2007) Rivastigminein the treatment of Alzheimer’s disease: An update. ClinInterv Aging 2, 17-32.

[112] Trebbastoni A, Gilio F, D’Antonio F, Cambieri C,Ceccanti M, de LC, Inghilleri M (2012) Chronic treatmentwith rivastigmine in patients with Alzheimer’s disease:A study on primary motor cortex excitability tested by 5Hz-repetitive transcranial magnetic stimulation. Clin Neu-rophysiol 123, 902-909.

[113] Emre M, Cummings JL, Lane RM (2007) Rivastig-mine in dementia associated with Parkinson’s diseaseand Alzheimer’s disease: Similarities and differences.J Alzheimers Dis 11, 509-519.

[114] Wesnes KA, McKeith I, Edgar C, Emre M, Lane R (2005)Benefits of rivastigmine on attention in dementia associ-ated with Parkinson disease. Neurology 65, 1654-1656.

[115] Wesnes K (2007) Rivastigmine tartrate with a focuson dementia associated with Parkinson’s disease. DrugsToday (Barc ) 43, 349-359.

[116] Siddiqui MA, Wagstaff AJ (2006) Rivastigmine: InParkinson’s disease dementia. CNS Drugs 20, 739-747.

[117] Siddiqui MA, Wagstaff AJ (2007) Rivastigmine inParkinson’s disease dementia: Profile report. Drugs Aging24, 255-259.

[118] Lalli S, Albanese A (2008) Rivastigmine in Parkin-son’s disease dementia. Expert Rev Neurother 8, 1181-1188.

[119] Almaraz AC, Driver-Dunckley ED, Woodruff BK, WellikKE, Caselli RJ, Demaerschalk BM, Adler CH, CavinessJN, Wingerchuk DM (2009) Efficacy of rivastigmine forcognitive symptoms in Parkinson disease with dementia.Neurologist 15, 234-237.

[120] Chitnis S, Rao J (2009) Rivastigmine in Parkinson’sdisease dementia. Expert Opin Drug Metab Toxicol 5,941-955.

[121] Wesnes KA, McKeith IG, Ferrara R, Emre M, del ST,Spano PF, Cicin-Sain A, Anand R, Spiegel R (2002)Effects of rivastigmine on cognitive function in demen-tia with lewy bodies: A randomised placebo-controlledinternational study using the cognitive drug researchcomputerised assessment system. Dement Geriatr CognDisord 13, 183-192.

[122] Vincent S, Lane R (2003) Rivastigmine in vascular demen-tia. Int Psychogeriatr 15(Suppl 1), 201-205.

[123] Moretti R, Torre P, Antonello RM, Cazzato G, Bava A(2004) Rivastigmine in vascular dementia. Expert OpinPharmacother 5, 1399-1410.

[124] Craig D, Birks J (2005) Rivastigmine for vascularcognitive impairment. Cochrane Database Syst Rev 2,CD004744.

[125] Roman GC (2005) Rivastigmine for subcortical vasculardementia. Expert Rev Neurother 5, 309-313.

[126] Chapuis C, Casez O, Lagrange E, Bedouch P, BessonG (2012) Hallucinations treated with rivastigmine inCreutzfeldt-Jakob disease. Fundam Clin Pharmacol 26,212-214.

[127] Stryjer R, Ophir D, Bar F, Spivak B, Weizman A, StrousRD (2012) Rivastigmine treatment for the preventionof electroconvulsive therapy-induced memory deficits inpatients with schizophrenia 1. Clin Neuropharmacol 35,161-164.

[128] Darreh-Shori T, Jelic V (2010) Safety and tolerability oftransdermal and oral rivastigmine in Alzheimer’s diseaseand Parkinson’s disease dementia. Expert Opin Drug Saf9, 167-176.

Page 54: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

600 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

[129] Lamb HM, Goa KL (2001) Rivastigmine. A pharma-coeconomic review of its use in Alzheimer’s disease.Pharmacoeconomics 19, 303-318.

[130] Cummings J, Winblad B (2007) A rivastigmine patchfor the treatment of Alzheimer’s disease and Parkin-son’s disease dementia. Expert Rev Neurother 7, 1457-1463.

[131] Yang LP, Keating GM (2007) Rivastigmine transdermalpatch: In the treatment of dementia of the Alzheimer’stype. CNS Drugs 21, 957-965.

[132] Winblad B, Machado JC (2008) Use of rivastigmine trans-dermal patch in the treatment of Alzheimer’s disease.Expert Opin Drug Deliv 5, 1377-1386.

[133] Kurz A, Farlow M, Lefevre G (2009) Pharmacokinetics ofa novel transdermal rivastigmine patch for the treatment ofAlzheimer’s disease: A review. Int J Clin Pract 63, 799-805.

[134] Cummings JL, Ferris SH, Farlow MR, Olin JT, Meng X(2010) Effects of rivastigmine transdermal patch and cap-sule on aspects of clinical global impression of changein Alzheimer’s disease: A retrospective analysis. DementGeriatr Cogn Disord 29, 406-412.

[135] Emre M, Bernabei R, Blesa R, Bullock R, CunhaL, Daniels H, Dziadulewicz E, Forstl H, Frolich L,Gabryelewicz T, Levin O, Lindesay J, Martinez-Lage P,Monsch A, Tsolaki M, van LT (2010) Drug profile: Trans-dermal rivastigmine patch in the treatment of Alzheimerdisease. CNS Neurosci Ther 16, 246-253.

[136] Farlow MR, Alva G, Meng X, Olin JT (2010) A 25-week,open-label trial investigating rivastigmine transdermalpatches with concomitant memantine in mild-to-moderateAlzheimer’s disease: A post hoc analysis. Curr Med ResOpin 26, 263-269.

[137] Farlow MR, Grossberg GT, Meng X, Olin J, SomogyiM (2011) Rivastigmine transdermal patch and capsulein Alzheimer’s disease: Influence of disease stage onresponse to therapy. Int J Geriatr Psychiatry 26, 1236-1243.

[138] Farlow MR, Grossberg GT, Meng X, Olin J, SomogyiM (2011) Rivastigmine transdermal patch and capsulein Alzheimer’s disease: Influence of disease stage onresponse to therapy. Int J Geriatr Psychiatry 26, 1236-1243.

[139] Grossberg GT, Sadowsky C, Olin JT (2010) Rivastigminetransdermal system for the treatment of mild to moderateAlzheimer’s disease. Int J Clin Pract 64, 651-660.

[140] Grossberg GT, Schmitt FA, Meng X, Tekin S, Olin J (2010)Reviews: Effects of transdermal rivastigmine on ADAS-cog items in mild-to-moderate Alzheimer’s disease. Am JAlzheimers Dis Other Demen 25, 627-633.

[141] Grossberg GT, Olin JT, Somogyi M, Meng X (2011) Doseeffects associated with rivastigmine transdermal patch inpatients with mild-to-moderate Alzheimer’s disease. Int JClin Pract 65, 465-471.

[142] Sadowsky CH, Dengiz A, Meng X, Olin JT (2010)Switching from oral donepezil to rivastigmine transdermalpatch in Alzheimer’s disease: 20-week extension phaseresults. Prim Care Companion J Clin Psychiatry 12, pii:PCC.09m00852.

[143] Sadowsky CH, Grossberg GT, Somogyi M, Meng X (2011)Predictors of sustained response to rivastigmine in patientswith Alzheimer’s disease: A retrospective analysis. PrimCare Companion CNS Disord 13, pii: PCC.10m01101

[144] Alva G, Grossberg GT, Schmitt FA, Meng X, Olin JT(2011) Efficacy of rivastigmine transdermal patch on activ-

ities of daily living: Item responder analyses. Int J GeriatrPsychiatry 26, 356-363.

[145] Articus K, Baier M, Tracik F, Kuhn F, Preuss UW, KurzA (2011) A 24-week, multicentre, open evaluation of theclinical effectiveness of the rivastigmine patch in patientswith probable Alzheimer’s disease. Int J Clin Pract 65,790-796.

[146] Choi SH, Park KW, Na DL, Han HJ, Kim EJ, Shim YS,Lee JH (2011) Tolerability and efficacy of memantine add-on therapy to rivastigmine transdermal patches in mild tomoderate Alzheimer’s disease: A multicenter, randomized,open-label, parallel-group study. Curr Med Res Opin 27,1375-1383.

[147] Dhillon S (2011) Spotlight on rivastigmine transdermalpatchdagger: In dementia of the Alzheimer’s type. DrugsAging 28, 927-930.

[148] Dhillon S (2011) Rivastigmine transdermal patch: Areview of its use in the management of dementia of theAlzheimer’s type. Drugs 71, 1209-1231.

[149] Greenspoon J, Herrmann N, Adam DN (2011) Trans-dermal rivastigmine: Management of cutaneous adverseevents and review of the literature. CNS Drugs 25, 575-583.

[150] Han HJ, Lee JJ, Park SA, Park HY, Kim JE, ShimYS, Shim DS, Kim EJ, Yoon SJ, Choi SH (2011) Effi-cacy and safety of switching from oral cholinesteraseinhibitors to the rivastigmine transdermal patch in patientswith probable Alzheimer’s disease. J Clin Neurol 7, 137-142.

[151] Nagy B, Brennan A, Brandtmuller A, Thomas SK, SullivanSD, Akehurst R (2011) Assessing the cost-effectivenessof the rivastigmine transdermal patch for Alzheimer’s dis-ease in the UK using. Int J Geriatr Psychiatry 26, 483-494.

[152] Weintraub D, Somogyi M, Meng X (2011) Rivastigminein Alzheimer’s disease and Parkinson’s disease dementia:An ADAS-Cog factor analysis. Am J Alzheimers Dis OtherDemen 26, 443-449.

[153] Seibert J, Tracik F, Articus K, Spittler S (2012) Effec-tiveness and tolerability of transdermal rivastigmine in thetreatment of Alzheimer’s disease in daily practice. Neu-ropsychiatr Dis Treat 8, 141-147.

[154] Tian H, Abouzaid S, Chen W, Kahler KH, Kim E(2012) Patient adherence to transdermal rivastigmineafter switching from oral donepezil: A retrospectiveclaims database study. Alzheimer Dis Assoc Disord, doi:10.1097/WAD.0b013e318266fb02 [Epubl ahead of print].

[155] Raskind MA (2003) Update on Alzheimer drugs (galan-tamine). Neurologist 9, 235-240.

[156] Stanilova MI, Molle ED, Yanev SG (2010) Galanthamineproduction by Leucojum aestivum cultures in vitro. Alka-loids Chem Biol 68, 167-270.

[157] Ivanov I, Georgiev V, Georgiev M, Ilieva M, Pavlov A(2011) Galanthamine and related alkaloids productionby Leucojum aestivum L. shoot culture using a tempo-rary immersion technology. Appl Biochem Biotechnol 163,268-277.

[158] Kuenburg B, Czollner L, Frohlich J, Jordis U (1999) Devel-opment of a pilot scale process for the anti-Alzheimer drug(-)-galanthamine using large-scale phenolic oxidative cou-pling and crystallization-induced chiral conversion. OrgProcess Res Development 3, 425-431.

[159] Trost BM, Tang W, Toste FD (2005) Divergent enantios-elective synthesis of (-)-galanthamine and (-)-morphine. JAm Chem Soc 127, 14785-14803.

Page 55: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 601

[160] Satcharoen V, McLean NJ, Kemp SC, Camp NP, Brown RC(2007) Stereocontrolled synthesis of (−)-galanthamine.Org Lett 9, 1867-1869.

[161] Tanimoto H, Kato T, Chida N (2007) Total synthesis of (+)-galanthamine starting from D-glucose. Tetrahedron Lett48, 6267-6270.

[162] Reddy JM, Kumar KV, Raju V, Bhaskar BV, HimabinduV, Bhattacharya A, Sundaram A, Banerjee R, ReddyGM, Bandichhor R (2008) Alternative total synthesis of(-)-galanthamine hydrobromide. Synth Commun 38, 2138-2149.

[163] Chang JH, Kang HU, Jung IH, Cho CG (2010) Totalsynthesis of (+/−)-galanthamine via a C3-selective Stillecoupling and IMDA cycloaddition cascade of 3,5-dibromo-2-pyrone. Org Lett 12, 2016-2018.

[164] Harvey AL (1995) The pharmacology of galanthamine andits analogues. Pharmacol Ther 68, 113-128.

[165] Fulton B, Benfield P (1996) Galanthamine. Drugs Aging9, 60-65.

[166] Blesa R (2000) Galantamine: Therapeutic effects beyondcognition. Dement Geriatr Cogn Disord 11(Suppl 1), 28-34.

[167] Sramek JJ, Frackiewicz EJ, Cutler NR (2000) Reviewof the acetylcholinesterase inhibitor galanthamine. ExpertOpin Investig Drugs 9, 2393-2402.

[168] Pearson VE (2001) Galantamine: A new alzheimerdrug with a past life. Ann Pharmacother 35, 1406-1413.

[169] Marco L, do Carmo Carreiras M (2006) Galanthamine, anatural product for the treatment of Alzheimer’s disease.Recent Pat CNS Drug Discov 1, 105-111.

[170] Marco-Contelles J, do Carmo Carreiras M, Rodriguez C,Villarroya M, Garcia AG (2006) Synthesis and pharma-cology of galantamine. Chem Rev 106, 116-133.

[171] Van Dam D, De Deyn PP (2006) Cognitive evaluation ofdisease-modifying efficacy of galantamine and memantinein the APP23 model. Eur Neuropsychopharmacol 16, 59-69.

[172] Villarroya M, Garcia AG, Marco-Contelles J, Lopez MG(2007) An update on the pharmacology of galantamine.Expert Opin Investig Drugs 16, 1987-1998.

[173] Matharu B, Gibson G, Parsons R, Huckerby TN, MooreSA, Cooper LJ, Millichamp R, Allsop D, Austen B (2009)Galantamine inhibits beta-amyloid aggregation and cyto-toxicity. J Neurol Sci 280, 49-58.

[174] Prvulovic D, Hampel H, Pantel J (2010) Galantamine forAlzheimer’s disease. Expert Opin Drug Metab Toxicol 6,345-354.

[175] Ago Y, Koda K, Takuma K, Matsuda T (2011) Phar-macological aspects of the acetylcholinesterase inhibitorgalantamine. J Pharmacol Sci 116, 6-17.

[176] Vigneault P, Bourgault S, Kaddar N, Caillier B, PiloteS, Patoine D, Simard C, Drolet B (2012) Galantamine(Reminyl(R)) delays cardiac ventricular repolarization andprolongs the QT interval by blocking the HERG current.Eur J Pharmacol 681, 68-74.

[177] Maelicke A (2000) Allosteric modulation of nicotinicreceptors as a treatment strategy for Alzheimer’s disease.Dement Geriatr Cogn Disord 11(Suppl 1), 11-18.

[178] Maelicke A, Schrattenholz A, Samochocki M, Radina M,Albuquerque EX (2000) Allosterically potentiating lig-ands of nicotinic receptors as a treatment strategy forAlzheimer’s disease. Behav Brain Res 113, 199-206.

[179] Maelicke A, Samochocki M, Jostock R, Fehrenbacher A,Ludwig J, Albuquerque EX, Zerlin M (2001) Allosteric

sensitization of nicotinic receptors by galantamine, a newtreatment strategy for Alzheimer’s disease. Biol Psychiatry49, 279-288.

[180] Coyle J, Kershaw P (2001) Galantamine, a cholinesteraseinhibitor that allosterically modulates nicotinic receptors:Effects on the course of Alzheimer’s disease. Biol Psychi-atry 49, 289-299.

[181] Woodruff-Pak DS, Lander C, Geerts H (2002) Nicotiniccholinergic modulation: Galantamine as a prototype. CNSDrug Rev 8, 405-426.

[182] Alexander KS, Wu HQ, Schwarcz R, Bruno JP (2012)Acute elevations of brain kynurenic acid impair cognitiveflexibility: Normalization by the alpha7 positive modu-lator galantamine. Psychopharmacology (Berl) 220, 627-637.

[183] Bickel U, Thomsen T, Weber W, Fischer JP, Bachus R, NitzM, Kewitz H (1991) Pharmacokinetics of galanthamine inhumans and corresponding cholinesterase inhibition. ClinPharmacol Ther 50, 420-428.

[184] Farlow MR (2003) Clinical pharmacokinetics of galan-tamine. Clin Pharmacokinet 42, 1383-1392.

[185] Monbaliu J, Verhaeghe T, Willems B, Bode W, Lavri-jsen K, Meuldermans W (2003) Pharmacokinetics ofgalantamine, a cholinesterase inhibitor, in several animalspecies. Arzneimittelforschung 53, 486-495.

[186] van Beijsterveldt L, Geerts R, Verhaeghe T, WillemsB, Bode W, Lavrijsen K, Meuldermans W (2004) Phar-macokinetics and tissue distribution of galantamine andgalantamine-related radioactivity after single intravenousand oral administration in the rat. Arzneimittelforschung54, 85-94.

[187] Huang F, Fu Y (2010) A review of clinical phar-macokinetics and pharmacodynamics of galantamine, areversible acetylcholinesterase inhibitor for the treatmentof Alzheimer’s disease, in healthy subjects and patients.Curr Clin Pharmacol 5, 115-124.

[188] Rainer M (1997) Galanthamine in Alzheimer’s disease. Anew alternative to tacrine? CNS Drugs 7, 89-97.

[189] Scott LJ , Goa KL (2000) Galantamine: A review of its usein Alzheimer’s disease. Drugs 60, 1095-1122.

[190] Tariot P (2001) Current status and new developmentswith galantamine in the treatment of Alzheimer’s disease.Expert Opin Pharmacother 2, 2027-2049.

[191] Erkinjuntti T (2002) Broad therapeutic benefits in patientswith probable vascular dementia or Alzheimer’s diseasewith cerebrovascular disease after treatment with galan-tamine. Eur J Neurol 9, 545.

[192] Kurz A (2002) A novel treatment for patients withAlzheimer’s disease and with vascular dementia. AnnN Y Acad Sci 977, 476-481.

[193] Lilienfeld S (2002) Galantamine–a novel cholinergic drugwith a unique dual mode of action for the treatment ofpatients with Alzheimer’s disease. CNS Drug Rev 8, 159-176.

[194] Corey-Bloom J (2003) Galantamine: A review of its usein Alzheimer’s disease and vascular dementia. Int J ClinPract 57, 219-223.

[195] Zarotsky V, Sramek JJ, Cutler NR (2003) Galantaminehydrobromide: An agent for Alzheimer’s disease. Am JHealth Syst Pharm 60, 446-452.

[196] Bullock R (2004) Galantamine: Use in Alzheimer’s dis-ease and related disorders. Expert Rev Neurother 4,153-163.

[197] Robinson DM, Plosker GL (2006) Galantamine extendedrelease. CNS Drugs 20, 673-681.

Page 56: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

602 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

[198] Seltzer B (2010) Galantamine-ER for the treatment ofmild-to-moderate Alzheimer’s disease. Clin Interv Aging5, 1-6.

[199] Kavanagh S, Gaudig M, Van Baelen B, Adami M, DelgadoA, Guzman C, Jedenius E, Schauble B (2011) Galantamineand behavior in Alzheimer disease: Analysis of four trials.Acta Neurol Scand 124, 302-308.

[200] Wallin AK, Wattmo C, Minthon L (2011) Galantaminetreatment in Alzheimer’s disease: Response and long-termoutcome in a routine clinical setting. Neuropsychiatr DisTreat 7, 565-576.

[201] Aarsland D, Hutchinson M, Larsen JP (2003) Cognitive,psychiatric and motor response to galantamine in Parkin-son’s disease with dementia. Int J Geriatr Psychiatry 18,937-941.

[201] [1]Scarpini E, Bruno G, Zappala G, Adami M, Richarz U,Gaudig M, Jacobs A, Schauble B (2011) Cessation versuscontinuation of galantamine treatment after 12 months oftherapy in patients with Alzheimer’s disease: a random-ized, double blind, placebo controlled withdrawal trial. JAlzheimers Dis 26, 211-220.

[202] Bora E, Veznedaroglu B, Kayahan B (2005) The effectof galantamine added to clozapine on cognition of fivepatients with schizophrenia. Clin Neuropharmacol 28,139-141.

[203] Craig D, Birks J (2006) Galantamine for vascular cognitiveimpairment. Cochrane Database Syst Rev, CD004746.

[204] Caro JJ, Salas M, Ward A, Getsios D, Mehnert A(2002) Economic analysis of galantamine, a cholinesteraseinhibitor, in the treatment of patients with mild to moderateAlzheimer’s disease in the Netherlands. Dement GeriatrCogn Disord 14, 84-89.

[205] Heinrich M (2010) Galanthamine from Galanthus andother Amaryllidaceae–chemistry and biology based on tra-ditional use. Alkaloids Chem Biol 68, 157-165.

[206] Maelicke A, Hoeffle-Maas A, Ludwig J, Maus A, Samo-chocki M, Jordis U, Koepke AK (2010) Memogainis a galantamine pro-drug having dramatically reducedadverse effects and enhanced efficacy. J Mol Neurosci 40,135-137.

[207] Hirasawa Y, Kobayashi J, Morita H (2009) TheLycopodium alkaloids. Heterocycles 77, 679-729.

[208] Bai DL, Tang XC, He XC (2000) Huperzine A, a poten-tial therapeutic agent for treatment of Alzheimer’s disease.Curr Med Chem 7, 355-374.

[209] Wang R, Yan H, Tang XC (2006) Progress in studies ofhuperzine A, a natural cholinesterase inhibitor from Chi-nese herbal medicine. Acta Pharmacol Sin 27, 1-26.

[210] Saxena A, Qian N, Kovach IM, Kozikowski AP, PangYP, Vellom DC, Radic Z, Quinn D, Taylor P, Doctor BP(1994) Identification of amino acid residues involved inthe binding of Huperzine A to cholinesterases. Protein Sci3, 1770-1778.

[211] McKinney M, Miller JH, Yamada F, Tuckmantel W,Kozikowski AP (1991) Potencies and stereoselectivitiesof enantiomers of huperzine A for inhibition of rat cor-tical acetylcholinesterase. Eur J Pharmacol 203, 303-305.

[212] Pang YP, Kozikowski AP (1994) Prediction of the bindingsites of huperzine A in acetylcholinesterase by dockingstudies. J Comput Aided Mol Des 8, 669-681.

[213] Ashani Y, Grunwald J, Kronman C, Velan B, ShaffermanA (1994) Role of tyrosine 337 in the binding of huperzineA to the active site of human acetylcholinesterase. MolPharmacol 45, 555-560.

[214] Dvir H, Jiang HL, Wong DM, Harel M, Chetrit M, HeXC, Jin GY, Yu GL, Tang XC, Silman I, Bai DL, Suss-man JL (2002) X-ray structures of Torpedo californicaacetylcholinesterase complexed with (+)-huperzine A and(−)-huperzine B: Structural evidence for an active siterearrangement. Biochemistry 41, 10810-10818.

[215] Ma X, Gang DR (2008) In vitro production of huperzineA, a promising drug candidate for Alzheimer’s disease.Phytochemistry 69, 2022-2028.

[216] Xia Y, Kozikowski AP (1989) A practical synthesis of theChinese nootropic agent Huperzine A: A possible lead inthe treatment of Alzheimer’s disease. J Amer Chem Soc111, 4116-4117.

[217] Campiani G, Sun LQ, Kozikowski AP, Aagaard P,McKinney M (1993) A palladium-catalyzed route toHuperzine A and its analogues and their anticholesteraseactivity. J Org Chem 58, 7660-7669.

[218] Kaneko S, Yoshino T, Katoh T, Terashima S (1998) Syn-thetic studies of huperzine A and its fluorinated analogs.1. Novel asymmetric syntheses of an enantiomeric pair ofHuperazine A. Tetrahedron 54, 5471-5484.

[219] Kelly SA, Foricher Y, Mann J, Bentley JM (2003) Aconvergent approach to huperzine A and analogues. OrgBiomol Chem 1, 2865-2876.

[220] Lucey C, Kelly SA, Mann J (2007) A concise and conver-gent (formal) total synthesis of huperzine A. Org BiomolChem 5, 301-306.

[221] Koshiba T, Yokoshima S, Fukuyama T (2009) Total syn-thesis of (−)-huperzine A. Org Lett 11, 5354-5356.

[222] Ding R, Sun BF, Lin GQ (2012) An efficient total synthesisof (−)-huperzine A. Org Lett 14, 4446-4449.

[223] Tudhope SR, Bellamy JA, Ball A, Rajasekar D, Azadi-Ardakani M, Meera HS, Gnanadeepam JM, Saiganesh R,Gibson F, He L, Behrens CH, Underiner G, Marturt J, FavreN (2012) Development of a large-scale synthetic route tomanufacture (−)-Huperzine A. Org Process Res Dev 16,635-642.

[224] Cheng DH, Ren H, Tang XC (1996) Huperzine A, a novelpromising acetylcholinesterase inhibitor. Neuroreport 8,97-101.

[225] Zangara A (2003) The psychopharmacology of huperzineA: An alkaloid with cognitive enhancing and neuroprotec-tive properties of interest in the treatment of Alzheimer’sdisease. Pharmacol Biochem Behav 75, 675-686.

[226] Wang R, Yan H, Tang XC (2006) Progress in studies ofhuperzine A, a natural cholinesterase inhibitor from Chi-nese herbal medicine. Acta Pharmacol Sin 27, 1-26.

[227] Zhang HY, Tang XC (2006) Neuroprotective effects ofhuperzine A: New therapeutic targets for neurodegener-ative disease. Trends Pharmacol Sci 27, 619-625.

[228] Zhang HY, Zheng CY, Yan H, Wang ZF, Tang LL, Gao X,Tang XC (2008) Potential therapeutic targets of huperzineA for Alzheimer’s disease and vascular dementia. ChemBiol Interact 175, 396-402.

[229] Zhang HY, Yan H, Tang XC (2008) Non-cholinergiceffects of huperzine A: Beyond inhibition of acetyl-cholinesterase. Cell Mol Neurobiol 28, 173-183.

[230] Ha GT, Wong RK, Zhang Y (2011) Huperzine a as potentialtreatment of Alzheimer’s disease: An assessment on chem-istry, pharmacology, and clinical studies. Chem Biodivers8, 1189-1204.

[231] Wang R, Zhang HY, Tang XC (2001) Huperzine A atten-uates cognitive dysfunction and neuronal degenerationcaused by beta-amyloid protein-(1-40) in rat 20. Eur JPharmacol 421, 149-156.

Page 57: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 603

[232] Xiao XQ, Zhang HY, Tang XC (2002) Huperzine Aattenuates amyloid beta-peptide fragment 25-35-inducedapoptosis in rat cortical neurons via inhibiting reactive oxy-gen species formation and caspase-3 activation. J NeurosciRes 67, 30-36.

[233] Zhang HY, Liang YQ, Tang XC, He XC, Bai DL (2002)Stereoselectivities of enantiomers of huperzine A in pro-tection against beta-amyloid(25-35)-induced injury inPC12 and NG108-15 cells and cholinesterase inhibitionin mice. Neurosci Lett 317, 143-146.

[234] Gao X, Tang XC (2006) Huperzine A attenuates mitochon-drial dysfunction in beta-amyloid-treated PC12 cells byreducing oxygen free radicals accumulation and improv-ing mitochondrial energy metabolism. J Neurosci Res 83,1048-1057.

[235] Gao X, Zheng CY, Yang L, Tang XC, Zhang HY (2009)Huperzine A protects isolated rat brain mitochondriaagainst beta-amyloid peptide. Free Radic Biol Med 46,1454-1462.

[236] Zhang HY, Yan H, Tang XC (2004) Huperzine A enhancesthe level of secretory amyloid precursor protein and proteinkinase C-alpha in intracerebroventricular beta-amyloid-(1-40) infused rats and human embryonic kidney 293 Swedishmutant cells. Neurosci Lett 360, 21-24.

[237] Yan H, Zhang HY, Tang XC (2007) Involvement of M1-muscarinic acetylcholine receptors, protein kinase C andmitogen-activated protein kinase in the effect of huperzineA on secretory amyloid precursor protein-alpha. Neurore-port 18, 689-692.

[238] Peng Y, Jiang L, Lee DY, Schachter SC, Ma Z, LemereCA (2006) Effects of huperzine A on amyloid precursorprotein processing and beta-amyloid generation in humanembryonic kidney 293 APP Swedish mutant cells. J Neu-rosci Res 84, 903-911.

[239] Peng Y, Lee DY, Jiang L, Ma Z, Schachter SC, LemereCA (2007) Huperzine A regulates amyloid precursor pro-tein processing via protein kinase C and mitogen-activatedprotein kinase pathways in neuroblastoma SK-N-SH cellsover-expressing wild type human amyloid precursor pro-tein 695. Neuroscience 150, 386-395.

[240] Wang CY, Zheng W, Wang T, Xie JW, Wang SL,Zhao BL, Teng WP, Wang ZY (2011) Huperzine Aactivates Wnt/beta-catenin signaling and enhances thenonamyloidogenic pathway in an Alzheimer transgenicmouse model. Neuropsychopharmacology 36, 1073-1089.

[241] Ratia M, Gimenez-Llort L, Camps P, Munoz-TorreroD, Perez B, Clos MV, Badia A (2012) Huprine X andhuperzine A improve cognition and regulate some neu-rochemical processes related with alzheimer’s disease intriple transgenic mice (3xTg-AD). Neurodegener Dis, doi:10.1159/000336427 [Epubl ahead of print].

[242] Little JT, Walsh S, Aisen PS (2008) An update on huperzineA as a treatment for Alzheimer’s disease. Expert OpinInvestig Drugs 17, 209-215.

[243] Desilets AR, Gickas JJ, Dunican KC (2009) Role ofhuperzine a in the treatment of Alzheimer’s disease. AnnPharmacother 43, 514-518.

[244] Wang BS, Wang H, Wei ZH, Song YY, Zhang L, Chen HZ(2009) Efficacy and safety of natural acetylcholinesteraseinhibitor huperzine A in the treatment of Alzheimer’sdisease: An updated meta-analysis. J Neural Transm 116,457-465.

[245] Rafii MS, Walsh S, Little JT, Behan K, Reynolds B, WardC, Jin S, Thomas R, Aisen PS (2011) A phase II trial

of huperzine A in mild to moderate Alzheimer disease.Neurology 76, 1389-1394.

[246] Hogenauer K, Baumann K, Enz A, Mulzer J (2001) Syn-thesis and acetylcholinesterase inhibition of 5-desaminohuperzine A derivatives. Bioorg Med Chem Lett 11, 2627-2630.

[247] Park SJ, Jung HJ, Son MS, Jung JM, Kim DH, Jung IH,Cho YB, Lee EH, Ryu JH (2012) Neuroprotective effectsof INM-176 against lipopolysaccharide-induced neuronalinjury. Pharmacol Biochem Behav 101, 427-433.

[248] Park SJ, Jung JM, Lee HE, Lee YW, Kim DH, Kim JM,Hong JG, Lee CH, Jung IH, Cho YB, Jang DS, Ryu JH(2012) The memory ameliorating effects of INM-176, anethanolic extract of Angelica gigas, against scopolamine-or Abeta(1-42)-induced cognitive dysfunction in mice. JEthnopharmacol 143, 611-620.

[249] Lahiri DK, Chen D, Maloney B, Holloway HW, Yu QS,Utsuki T, Giordano T, Sambamurti K, Greig NH (2007)The experimental Alzheimer’s disease drug posiphen [(+)-phenserine] lowers amyloid-beta peptide levels in cellculture and mice. J Pharmacol Exp Ther 320, 386-396.

[250] Rogers JT, Mikkilineni S, Cantuti-Castelvetri I, Smith DH,Huang X, Bandyopadhyay S, Cahill CM, MaccecchiniML, Lahiri DK, Greig NH (2011) The alpha-synuclein5’untranslated region targeted translation blockers: Anti-alpha synuclein efficacy of cardiac glycosides andPosiphen. J Neural Transm 118, 493-507.

[251] Mikkilineni S, Cantuti-Castelvetri I, Cahill CM, BalliedierA, Greig NH, Rogers JT (2012) The anticholinesterasephenserine and its enantiomer posiphen as 5′untranslated-region-directed translation blockers of the Parkinson’salpha synuclein expression. Parkinsons Dis 2012,142372.

[252] Maccecchini ML, Chang MY, Pan C, John V, ZetterbergH, Greig NH (2012) Posiphen as a candidate drug to lowerCSF amyloid precursor protein, amyloid-beta peptide andtau levels: Target engagement, tolerability and pharma-cokinetics in humans. J Neurol Neurosurg Psychiatry 83,894-902.

[253] Pavlic M (1970) The inhibition of butyrylcholinesteraseby methanesulfonyl fluoride. Biochim Biophys Acta 198,389-391.

[254] Krupka RM (1974) Methanesulfonyl fluoride inactiva-tion of acetylcholinesterase in the presence of substratesand reversible inhibitors. Biochim Biophys Acta 370,197-207.

[255] Malin DH, Toups PJ, Osgood LD, Fowler DE, HunterCL, Arcangeli KR, Moss DE (1991) Methanesulfonyl flu-oride enhances one-trial reward learning in mid-aged rats.Neurobiol Aging 12, 181-183.

[256] Malin DH, Plotner RE, Radulescu SJ, Ferebee RN, LakeJR, Negrete PG, Schaefer PJ, Crothers MK, Moss DE(1993) Chronic methanesulfonyl fluoride enhances one-trial per day reward learning in aged rats. Neurobiol Aging14, 393-395.

[257] Palacios-Esquivel RL, Pacheco G, Moss DE (1993)Methanesulfonyl fluoride (MSF) blocks scopolamine-induced amnesia in rats. Neurobiol Aging 14, 93-96.

[258] Borlongan CV, Sumaya IC, Moss DE (2005) Methanesul-fonyl fluoride, an acetylcholinesterase inhibitor, attenuatessimple learning and memory deficits in ischemic rats.Brain Res 1038, 50-58.

[259] Moss DE, Berlanga P, Hagan MM, Sandoval H, Ishida C(1999) Methanesulfonyl fluoride (MSF): A double-blind,placebo-controlled study of safety and efficacy in the treat-

Page 58: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

604 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

ment of senile dementia of the Alzheimer type. AlzheimerDis Assoc Disord 13, 20-25.

[260] Kamal MA, Klein P, Yu QS, Tweedie D, Li Y, HollowayHW, Greig NH (2006) Kinetics of human serum butyryl-cholinesterase and its inhibition by a novel experimentalAlzheimer therapeutic, bisnorcymserine. J Alzheimers Dis10, 43-51.

[261] Bartolucci C, Stojan J, Yu QS, Greig NH, Lamba D (2012)Kinetics of Torpedo californica acetylcholinesterase inhi-bition by bisnorcymserine and crystal structure of thecomplex with its leaving group. Biochem J 444, 269-277.

[262] Pang YP, Quiram P, Jelacic T, Hong F, Brimijoin S(1996) Highly potent, selective, and low cost bis-tetrahydroaminacrine inhibitors of acetylcholinesterase.Steps toward novel drugs for treating Alzheimer’s disease.J Biol Chem 271, 23646-23649.

[263] Wang ZF, Yan J, Fu Y, Tang XC, Feng S, He XC, BaiDL (2008) Pharmacodynamic study of FS-0311: A novelhighly potent, selective acetylcholinesterase inhibitor. CellMol Neurobiol 28, 245-261.

[264] Badia A, Banos JE, Camps P, Contreras J, Gorbig DM,Munoz-Torrero D, Simon M, Vivas NM (1998) Syn-thesis and evaluation of tacrine-huperzine A hybrids asacetylcholinesterase inhibitors of potential interest for thetreatment of Alzheimer’s disease. Bioorg Med Chem 6,427-440.

[265] Camps P, El AR, Gorbig DM, Morral J, Munoz-Torrero D,Badia A, Eladi BJ, Vivas NM, Barril X, Orozco M, LuqueFJ (1999) Synthesis, in vitro pharmacology, and molecularmodeling of very potent tacrine-huperzine A hybrids asacetylcholinesterase inhibitors of potential interest for thetreatment of Alzheimer’s disease. J Med Chem 42, 3227-3242.

[266] Camps P, Cusack B, Mallender WD, El Achab RE, MorralJ, Munoz-Torrero D, Rosenberry TL (2000) Huprine X isa novel high-affinity inhibitor of acetylcholinesterase thatis of interest for treatment of Alzheimer’s disease. MolPharmacol 57, 409-417.

[267] Camps P, El AR, Morral J, Munoz-Torrero D, BadiaA, Banos JE, Vivas NM, Barril X, Orozco M, LuqueFJ (2000) New tacrine-huperzine A hybrids (huprines):Highly potent tight-binding acetylcholinesterase inhibitorsof interest for the treatment of Alzheimer’s disease. J MedChem 43, 4657-4666.

[268] Camps P, Gomez E, Munoz-Torrero D, Badia A, VivasNM, Barril X, Orozco M, Luque FJ (2001) Synthesis,in vitro pharmacology, and molecular modeling of syn-huprines as acetylcholinesterase inhibitors. J Med Chem44, 4733-4736.

[269] Camps P, Munoz-Torrero D (2001) Tacrine-huperzine Ahybrids (huprines): A new class of highly potent andselective acetylcholinesterase inhibitors of interest for thetreatment of Alzheimer’s disease. Mini Rev Med Chem 1,163-174.

[270] Carlier PR, Du DM, Han Y, Liu J, Pang YP (1999)Potent, easily synthesized huperzine A-tacrine hybridacetylcholinesterase inhibitors. Bioorg Med Chem Lett 9,2335-2338.

[271] Ronco C, Sorin G, Nachon F, Foucault R, Jean L,Romieu A, Renard PY (2009) Synthesis and structure-activity relationship of Huprine derivatives as humanacetylcholinesterase inhibitors. Bioorg Med Chem 17,4523-4536.

[272] Ronco C, Foucault R, Gillon E, Bohn P, Nachon F,Jean L, Renard PY (2011) New huprine derivatives

functionalized at position 9 as highly potent acetyl-cholinesterase inhibitors. ChemMedChem 6, 876-888.

[273] Ronco C, Carletti E, Colletier JP, Weik M, Nachon F,Jean L, Renard PY (2012) Huprine derivatives as sub-nanomolar human acetylcholinesterase inhibitors: Fromrational design to validation by X-ray crystallography.ChemMedChem 7, 400-405.

[274] Alcala MM, Vivas NM, Hospital S, Camps P, Munoz-Torrero D, Badia A (2003) Characterisation of theanticholinesterase activity of two new tacrine-huperzineA hybrids. Neuropharmacology 44, 749-755.

[275] Roman S, Vivas NM, Badia A, Clos MV (2002) Interactionof a new potent anticholinesterasic compound (+/-)huprineX with muscarinic receptors in rat brain. Neurosci Lett 325,103-106.

[276] Dvir H, Wong DM, Harel M, Barril X, Orozco M, LuqueFJ, Munoz-Torrero D, Camps P, Rosenberry TL, SilmanI, Sussman JL (2002) 3D structure of Torpedo califor-nica acetylcholinesterase complexed with huprine X at 2.1A resolution: Kinetic and molecular dynamic correlates.Biochemistry 41, 2970-2981.

[277] Ratia M, Gimenez-Llort L, Camps P, Munoz-Torrero D,Clos MV, Badia A (2010) Behavioural effects and regula-tion of PKCalpha and MAPK by huprine X in middle agedmice. Pharmacol Biochem Behav 95, 485-493.

[278] Hedberg MM, Clos MV, Ratia M, Gonzalez D, Lithner CU,Camps P, Munoz-Torrero D, Badia A, Gimenez-Llort L,Nordberg A (2010) Effect of huprine X on beta-amyloid,synaptophysin and alpha7 neuronal nicotinic acetylcholinereceptors in the brain of 3xTg-AD and APPswe transgenicmice. Neurodegener Dis 7, 379-388.

[279] Gemma S, Gabellieri E, Huleatt P, Fattorusso C, BorrielloM, Catalanotti B, Butini S, De AM, Novellino E, Nacci V,Belinskaya T, Saxena A, Campiani G (2006) Discovery ofhuperzine A-tacrine hybrids as potent inhibitors of humancholinesterases targeting their midgorge recognition sites.J Med Chem 49, 3421-3425.

[280] Galdeano C, Viayna E, Sola I, Formosa X, Camps P, BadiaA, Clos MV, Relat J, Ratia M, Bartolini M, Mancini F,Andrisano V, Salmona M, Minguillon C, Gonzalez-MunozGC, Rodriguez-Franco MI, Bidon-Chanal A, Luque FJ,Munoz-Torrero D (2012) Huprine-tacrine heterodimersas anti-amyloidogenic compounds of potential interestagainst Alzheimer’s and prion diseases. J Med Chem 55,661-669.

[281] Munoz-Torrero D, Pera M, Relat J, Ratia M, Galdeano C,Viayna E, Sola I, Formosa X, Camps P, Badia A, ClosMV (2012) Expanding the multipotent profile of huprine-tacrine heterodimers as disease-modifying anti-Alzheimeragents. Neurodegener Dis 10, 96-99.

[282] Carlier PR, Du DM, Han YF, Liu J, Perola E, WilliamsID, Pang YP (2000) Dimerization of an inactive fragmentof huperzine a produces a drug with twice the potency ofthe natural product. Angew Chem Int Ed Engl 39, 1775-1777.

[283] Cui W, Cui GZ, Li W, Zhang Z, Hu S, Mak S, ZhangH, Carlier PR, Choi CL, Wong YT, Lee SM, Han Y(2011) Bis(12)-hupyridone, a novel multifunctional dimer,promotes neuronal differentiation more potently than itsmonomeric natural analog huperzine A possibly throughalpha7 nAChR. Brain Res 1401, 10-17.

[284] Lewis WG, Green LG, Grynszpan F, Radic Z, Carlier PR,Taylor P, Finn MG, Sharpless KB (2002) Click chemistryin situ: acetylcholinesterase as a reaction vessel for theselective assembly of a femtomolar inhibitor from an array

Page 59: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 605

of building blocks. Angew Chem Int Ed Engl 41, 1053-1057.

[285] Szymanski P, Janik A, Zurek E, Mikiciuk-Olasik E(2011) Design, synthesis and biological evaluation of new2-benzoxazolinone derivatives as potential cholinesteraseinhibitors for therapy of Alzheimer’s disease. Pharmazie66, 399-403.

[286] Wu ZP, Wu XW, Shen T, Li YP, Cheng X, Gu LQ, HuangZS, An LK (2012) Synthesis and acetylcholinesteraseand butyrylcholinesterase inhibitory activities of 7-alkoxylsubstituted indolizinoquinoline-5,12-dione derivatives.Arch Pharm (Weinheim) 345, 175-184.

[287] Ignasik M, Bajda M, Guzior N, Prinz M, HolzgrabeU, Malawska B (2012) Design, synthesis and evaluationof novel 2-(aminoalkyl)-isoindoline-1,3-dione derivativesas dual-binding site acetylcholinesterase inhibitors. ArchPharm (Weinheim) 345, 509-516.

[288] Anand P, Singh B (2012) Synthesis and evaluation of novel4-[(3H,3aH,6aH)-3-phenyl)-4,6-dioxo-2-phenyldihydro-2H-pyrrolo[3,4-d]isoxazol-5(3H,6H,6aH)-yl]benzoicacid derivatives as potent acetylcholinesterase inhibitorsand anti-amnestic agents. Bioorg Med Chem 20, 521-530.

[289] Ashraf AM, Ismail R, Choon TS, Kumar RS, OsmanH, Arumugam N, Almansour AI, Elumalai K, Singh A(2012) AChE inhibitor: A regio- and stereo-selective1,3-dipolar cycloaddition for the synthesis of novelsubstituted 5,6-dimethoxy spiro[5.3′]-oxindole-spiro-[6.3′′]-2,3-dihydro-1H-inden-1′′-one-7-(substitutedaryl)-tetrahydro-1H-pyrrolo[1,2-c][1,3]thiazole 1. BioorgMed Chem Lett 22, 508-511.

[290] Li Z, Wang B, Hou JQ, Huang SL, Ou TM, TanJH, An LK, Li D, Gu LQ, Huang ZS (2012) 2-(2-indolyl-)-4(3H)-quinazolines derivates as new inhibitorsof AChE: Design, synthesis, biological evaluation andmolecular modelling. J Enzyme Inhib Med Chem, doi:10.3109/14756366.2012.663363 [Epubl ahead of print].

[291] Piplani P, Rani A, Saihgal R, Sharma M (2011) Syn-thesis and pharmacological evaluation of some quinolinederivatives as potential cognition enhancers. Arzneimit-telforschung 61, 373-378.

[292] Chen Y, Sun J, Fang L, Liu M, Peng S, Liao H, LehmannJ, Zhang Y (2012) Tacrine-ferulic acid-nitric oxide (NO)donor trihybrids as potent, multifunctional acetyl- andbutyrylcholinesterase inhibitors. J Med Chem 55, 4309-4321.

[293] Wonga KY, Duchowicz PR, Mercader AG, Castro EA(2012) QSAR applications during last decade on inhibitorsof acetylcholinesterase in Alzheimer’s disease. Mini RevMed Chem, [Epubl ahead of print].

[294] Yan A, Wang K (2012) Quantitative structure and bioac-tivity relationship study on human acetylcholinesteraseinhibitors. Bioorg Med Chem Lett 22, 3336-3342.

[295] Chen Y, Fang L, Peng S, Liao H, Lehmann J, Zhang Y(2012) Discovery of a novel acetylcholinesterase inhibitorby structure-based virtual screening techniques. BioorgMed Chem Lett 22, 3181-3187.

[296] Wang K, Hu X, Wang Z, Yan A (2012) Classification ofacetylcholinesterase inhibitors and decoys by a supportvector machine. Comb Chem High Throughput Screen 15,492-502.

[297] Berg L, Andersson CD, Artursson E, Hornberg A, Tune-malm AK, Linusson A, Ekstrom F (2011) Targetingacetylcholinesterase: Identification of chemical leads byhigh throughput screening, structure determination andmolecular modeling. PLoS One 6, e26039.

[298] Kojima J, Nakajima K, Ochiai M, Nakayama K (1997)Effects of NIK-247 on cholinesterase and scopolamine-induced amnesia. Methods Find Exp Clin Pharmacol 19,245-251.

[299] Kojima J, Onodera K (1998) Effects of NIK-247 andtacrine on muscarinic receptor subtypes in rats. Gen Phar-macol 30, 537-541.

[300] Kojima J, Onodera K (1998) NIK-247 induces long-termpotentiation of synaptic transmission in the CA1 region ofrat hippocampal slices through M2 muscarinic receptors.Gen Pharmacol 31, 297-300.

[301] Piazzi L, Rampa A, Bisi A, Gobbi S, Belluti F, CavalliA, Bartolini M, Andrisano V, Valenti P, Recanatini M(2003) 3-(4-[[Benzyl(methyl)amino]methyl]phenyl)-6,7-dimethoxy-2H-2-chromenone (AP2238) inhibits bothacetylcholinesterase and acetylcholinesterase-inducedbeta-amyloid aggregation: A dual function lead forAlzheimer’s disease therapy. J Med Chem 46, 2279-2282.

[302] Piazzi L, Cavalli A, Belluti F, Bisi A, Gobbi S,Rizzo S, Bartolini M, Andrisano V, Recanatini M,Rampa A (2007) Extensive SAR and computationalstudies of 3-{4-[(benzylmethylamino)methyl]phenyl}-6,7-dimethoxy-2H-2-chromenone (AP2238) derivatives. JMed Chem 50, 4250-4254.

[303] Rizzo S, Bartolini M, Ceccarini L, Piazzi L, Gobbi S,Cavalli A, Recanatini M, Andrisano V, Rampa A (2010)Targeting Alzheimer’s disease: Novel indanone hybridsbearing a pharmacophoric fragment of AP2238. BioorgMed Chem 18, 1749-1760.

[304] Bayes M, Rabasseda X, Prous JR (2003) Gateways to clin-ical trials. Methods Find Exp Clin Pharmacol 25, 565-597.

[305] Imbimbo BP, Martelli P, Troetel WM, Lucchelli F, LuccaU, Thal LJ (1999) Efficacy and safety of eptastigmine forthe treatment of patients with Alzheimer’s disease. Neu-rology 52, 700-708.

[306] Imbimbo BP, Troetel WM, Martelli P, Lucchelli F(2000) A 6-month, double-blind, placebo-controlled trialof eptastigmine in Alzheimer’s disease. Dement GeriatrCogn Disord 11, 17-24.

[307] Braida D, Ottonello F, Sala M (2000) Eptastigmineimproves eight-arm radial maze performance in aged rats.Pharmacol Res 42, 299-304.

[308] Braida D, Ottonello F, Sala M (2000) Eptastigmine restoresthe aged rat’s normal cortical spectral power pattern. Phar-macol Res 42, 495-500.

[309] Braida D, Sala M (2001) Eptastigmine: Ten years ofpharmacology, toxicology, pharmacokinetic, and clinicalstudies. CNS Drug Rev 7, 369-386.

[310] Trabace L, Cassano T, Loverre A, Steardo L, CuomoV (2002) CHF2819: Pharmacological profile of a novelacetylcholinesterase inhibitor. CNS Drug Rev 8, 53-69.

[311] Trabace L, Cassano T, Colaianna M, Castrignano S,Giustino A, Amoroso S, Steardo L, Cuomo V (2007) Neu-rochemical and neurobehavioral effects of ganstigmine(CHF2819), a novel acetylcholinesterase inhibitor, in ratprefrontal cortex: An in vivo study. Pharmacol Res 56,288-294.

[312] Jhee SS, Fabbri L, Piccinno A, Monici P, Moran S, Zarot-sky V, Tan EY, Frackiewicz EJ, Shiovitz T (2003) Firstclinical evaluation of ganstigmine in patients with probableAlzheimer’s disease. Clin Neuropharmacol 26, 164-169.

[313] Mazzucchelli M, Porrello E, Villetti G, Pietra C, GovoniS, Racchi M (2003) Characterization of the effect ofganstigmine (CHF2819) on amyloid precursor protein

Page 60: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

606 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

metabolism in SH-SY5Y neuroblastoma cells. J NeuralTransm 110, 935-947.

[314] Windisch M, Hutter-Paier B, Jerkovic L, Imbimbo B,Villetti G (2003) The protective effect of ganstigmineagainst amyloid beta 25-35 neurotoxicity on chickencortical neurons is independent from the cholinesteraseinhibition. Neurosci Lett 341, 181-184.

[315] Bartolucci C, Siotto M, Ghidini E, Amari G, BolzoniPT, Racchi M, Villetti G, Delcanale M, Lamba D (2006)Structural determinants of Torpedo californica acetyl-cholinesterase inhibition by the novel and orally activecarbamate based anti-alzheimer drug ganstigmine (CHF-2819). J Med Chem 49, 5051-5058.

[316] Hock FJ, Gerhards HJ, Wiemer G, Stechl J, Ruger W,Urbach H (1989) Effects of the novel compound, Hoe065, upon impaired learning and memory in rodents. EurJ Pharmacol 171, 79-85.

[317] Wiemer G, Becker R, Gerhards H, Hock F, Stechl J, RugerW (1989) Effects of Hoe 065, a compound structurallyrelated to inhibitors of angiotensin converting enzyme, onacetylcholine metabolism in rat brain. Eur J Pharmacol166, 31-39.

[318] Grupp LA, Chow SY (1991) Effects of the novel com-pound Hoe 065, a central enhancer of cholinergic activity,on voluntary alcohol consumption in rats. Brain Res Bull26, 617-619.

[319] Liston DR, Nielsen JA, Villalobos A, Chapin D, Jones SB,Hubbard ST, Shalaby IA, Ramirez A, Nason D, White WF(2004) Pharmacology of selective acetylcholinesteraseinhibitors: Implications for use in Alzheimer’s disease. EurJ Pharmacol 486, 9-17.

[320] Giacobini E (1997) Metrifonate. A viewpoint. DrugsAging 11, 497.

[321] Schneider LS, Giacobini E (1999) Metrifonate: Acholinesterase inhbitor for Alzheimer’s disease therapy.CNS Drug Rev 5, 13-26.

[322] Schneider LS (2000) Metrifonate for Alzheimer’s diseasepatients. J Clin Psychiatry 61, 218-219.

[323] Farlow MR, Cyrus PA (2000) Metrifonate therapy inAlzheimer’s disease: A pooled analysis of four ran-domized, double-blind, placebo-controlled trials. DementGeriatr Cogn Disord 11, 202-211.

[324] Ormrod D, Spencer C (2000) Metrifonate. A review of itsuse in Alzheimer’s disease. CNS Drugs 13, 443-467.

[325] Giacobini E (2004) Cholinesterase inhibitors: New rolesand therapeutic alternatives. Pharmacol Res 50, 433-440.

[326] Thatte U (2005) Phenserine Axonyx. Curr Opin InvestigDrugs 6, 729-739.

[327] Klein J (2007) Phenserine. Expert Opin Investig Drugs 16,1087-1097.

[328] Kadir A, Andreasen N, Almkvist O, Wall A, Forsberg A,Engler H, Hagman G, Larksater M, Winblad B, ZetterbergH, Blennow K, Langstrom B, Nordberg A (2008) Effectof phenserine treatment on brain functional activity andamyloid in Alzheimer’s disease. Ann Neurol 63, 621-631.

[329] Winblad B, Giacobini E, Frolich L, Friedhoff LT, BruinsmaG, Becker RE, Greig NH (2010) Phenserine efficacy inAlzheimer’s disease. J Alzheimers Dis 22, 1201-1208.

[330] Benech H, Vincenti M, Fouchart F, Pruvost A, Vienet R,Istin M, Grognet JM (1998) Development and in vivoassessment of a transdermal system for physostigmine.Methods Find Exp Clin Pharmacol 20, 489-498.

[331] Moller H-J, Hampel H, Hegerl U, Schmitt W, WalterK (1999) Double-blind, randomized, placebo-controlledclinical trial on the efficacy and tolerability of a physostig-

mine patch in patients with senile dementia of theAlzheimer type. Pharmacopsychiatry 32, 99-106.

[332] Kim JH, Lee CH, Choi HK (2002) Transdermal deliveryof physostigmine: Effects of enhancers and pressure-sensitive adhesives. Drug Dev Ind Pharm 28, 833-839.

[333] Cerasoli DM, Griffiths EM, Doctor BP, Saxena A, FedorkoJM, Greig NH, Yu QS, Huang Y, Wilgus H, Karatzas CN,Koplovitz I, Lenz DE (2005) In vitro and in vivo char-acterization of recombinant human butyrylcholinesterase(Protexia) as a potential nerve agent bioscavenger. ChemBiol Interact 157-158 363-365.

[334] Mumford H, Troyer JK (2011) Post-exposure therapy withrecombinant human BuChE following percutaneous VXchallenge in guinea-pigs. Toxicol Lett 206, 29-34.

[335] Natori K, Okazaki Y, Irie T, Katsube J (1990) Pharmaco-logical and biochemical assessment of SM-10888, a novelcholinesterase inhibitor. Jpn J Pharmacol 53, 145-155.

[336] Okazaki Y, Natori K, Irie T, Katsube J (1990) Effect of anovel CNS-selective cholinesterase inhibitor, SM-10888,on habituation and passive avoidance responses in mice.Jpn J Pharmacol 53, 211-220.

[337] Xu M, Nakamura Y, Yamamoto T, Natori K, Irie T, UtsumiH, Kato T (1991) Determination of basal acetylcholinerelease in vivo by rat brain dialysis with a U-shaped can-nula: Effect of SM-10888, a putative therapeutic drug forAlzheimer’s disease. Neurosci Lett 123, 179-182.

[338] Schroeder H, Dumont I, Boyet S, Mocaer E, NehligA (1992) Effects of the acute administration of a newtrimethylxanthine derivative, S 9977-2, on local cerebralblood flow and glucose utilization in the rat. Eur J Phar-macol 220, 217-229.

[339] Trudeau LE, Fossier P, Baux G, Tauc L (1992) Xanthinederivatives IBMX and S-9977-2 potentiate transmissionat an Aplysia central cholinergic synapse. Brain Res 586,78-85.

[340] Boulanger CM, Hughes H, Bond RA, Rafla E, VanhouttePM (1993) Effects of S9977 on adrenergic neurotransmis-sion. Gen Pharmacol 24, 429-434.

[341] Fernandez-Novoa L, Alvarez XA, tolle-Sarbach S, GuezD, Zas R, Polo E, Cacabelos R (1996) Effects of S-9977-2on histamine levels in rats. Methods Find Exp Clin Phar-macol 18, 197-203.

[342] Braga MF, Harvey AL, Rowan EG (1991) Effects oftacrine, velnacrine (HP029), suronacrine (HP128), and3,4-diaminopyridine on skeletal neuromuscular transmis-sion in vitro. Br J Pharmacol 102, 909-915.

[343] Huff FJ, Antuono P, Murphy M, Beyer J, Dobson C (1991)Potential clinical use of an adrenergic/cholinergic agent(HP 128) in the treatment of Alzheimer’s disease. Ann N YAcad Sci 640, 263-267.

[344] Isoma K, Ishikawa M, Ohta M, Ogawa Y, Hasegawa H,Kohda T, Kamei J (2002) Effects of T-82, a new quino-line derivative, on cholinesterase activity and extracellularacetylcholine concentration in rat brain. Jpn J Pharmacol88, 206-212.

[345] Isomae K, Morimoto S, Hasegawa H, Morita K, KameiJ (2003) Effects of T-82, a novel acetylcholinesteraseinhibitor, on impaired learning and memory in passiveavoidance task in rats. Eur J Pharmacol 465, 97-103.

[346] Yu Q, Holloway HW, Utsuki T, Brossi A, Greig NH (1999)Synthesis of novel phenserine-based-selective inhibitorsof butyrylcholinesterase for Alzheimer’s disease. J MedChem 42, 1855-1861.

[347] Yu QS, Holloway HW, Luo W, Lahiri DK, Brossi A,Greig NH (2010) Long-acting anticholinesterases for

Page 61: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 607

myasthenia gravis: Synthesis and activities of quater-nary phenylcarbamates of neostigmine, pyridostigmineand physostigmine. Bioorg Med Chem 18, 4687-4693.

[348] Kamal MA, Greig NH, Alhomida AS, Al-Jafari AA (2000)Kinetics of human acetylcholinesterase inhibition by thenovel experimental Alzheimer therapeutic agent, tolserine.Biochem Pharmacol 60, 561-570.

[349] Hartmann J, Kiewert C, Duysen EG, Lockridge O, GreigNH, Klein J (2007) Excessive hippocampal acetylcholinelevels in acetylcholinesterase-deficient mice are moder-ated by butyrylcholinesterase activity. J Neurochem 100,1421-1429.

[350] Zemlan FP (1996) Velnacrine for the treatment ofAlzheimer’s disease: A double-blind, placebo-controlledtrial. The mentane study group. J Neural Transm 103,1105-1116.

[351] Zemlan FP, Keys M, Richter RW, Strub RL (1996)Double-blind placebo-controlled study of velnacrine inAlzheimer’s disease. Life Sci 58, 1823-1832.

[352] Hatip-Al-Khatib I, Takashi A, Egashira N, Iwasaki K,Fujiwara M (2004) Comparison of the effect of TAK-147(zanapezil) and E-2020 (donepezil) on extracellular acetyl-choline level and blood flow in the ventral hippocampusof freely moving rats. Brain Res 1012, 169-176.

[353] Hatip-Al-Khatib I, Iwasaki K, Yoshimitsu Y, Arai T,Egashira N, Mishima K, Ikeda T, Fujiwara M (2005) Effectof oral administration of zanapezil (TAK-147) for 21 dayson acetylcholine and monoamines levels in the ventral hip-pocampus of freely moving rats. Br J Pharmacol 145,1035-1044.

[354] Hornsperger JM, Collard JN, Heydt JG, Giacobini E,Funes S, Dow J, Schirlin D (1994) Trimethylsilylated tri-fluoromethyl ketones, a novel class of acetylcholinesteraseinhibitors: Biochemical and pharmacological profile ofMDL 73,745. Biochem Soc Trans 22, 758-763.

[355] Cutler NR, Seifert RD, Schleman MM, Sramek JJ, Szyl-leyko OJ, Howard DR, Barchowsky A, Wardle TS, BrassEP (1995) Acetylcholinesterase inhibition by zifrosilone:Pharmacokinetics and pharmacodynamics. Clin Pharma-col Ther 58, 54-61.

[356] Zhu XD, Giacobini E, Hornsperger JM (1995) Effect ofMDL 73,745 on acetylcholine and biogenic amine levelsin rat cortex. Eur J Pharmacol 276, 93-99.

[357] Youdim MB, Buccafusco JJ (2005) Multi-functional drugsfor various CNS targets in the treatment of neurodegener-ative disorders. Trends Pharmacol Sci 26, 27-35.

[358] del Monte-Millan M, Garcia-Palomero E, Valenzuela R,Usan P, de AC, Munoz-Ruiz P, Rubio L, Dorronsoro I,Martinez A, Medina M (2006) Dual binding site acetyl-cholinesterase inhibitors: Potential new disease-modifyingagents for AD. J Mol Neurosci 30, 85-88.

[359] Martinez A, Castro A (2006) Novel cholinesteraseinhibitors as future effective drugs for the treatment ofAlzheimer’s disease. Expert Opin Investig Drugs 15,1-12.

[360] Munoz-Torrero D, Camps P (2006) Dimeric and hybridanti-Alzheimer drug candidates. Curr Med Chem 13, 399-422.

[361] Munoz-Torrero D (2008) Acetylcholinesterase inhibitorsas disease-modifying therapies for Alzheimer’s disease.Curr Med Chem 15, 2433-2455.

[362] Cavalli A, Bolognesi ML, Minarini A, Rosini M, TumiattiV, Recanatini M, Melchiorre C (2008) Multi-target-directed ligands to combat neurodegenerative diseases. JMed Chem 51, 347-372.

[363] Bolognesi ML, Matera R, Minarini A, Rosini M, Mel-chiorre C (2009) Alzheimer’s disease: New approaches todrug discovery. Curr Opin Chem Biol 13, 303-308.

[364] Bolognesi ML, Simoni E, Rosini M, Minarini A, Tumi-atti V, Melchiorre C (2011) Multitarget-directed ligands:Innovative chemical probes and therapeutic tools againstAlzheimer’s disease. Curr Top Med Chem 11, 2797-2806.

[365] Buccafusco JJ (2009) Multifunctional receptor-directeddrugs for disorders of the central nervous system. Neu-rotherapeutics 6, 4-13.

[366] Melchiorre C, Bolognesi ML, Minarini A, Rosini M, Tumi-atti V (2010) Polyamines in drug discovery: From theuniversal template approach to the multitarget-directed lig-and design strategy. J Med Chem 53, 5906-5914.

[367] Leon R, Garcia AG, Marco-Contelles J (2011) Recentadvances in the multitarget-directed ligands approach forthe treatment of Alzheimer’s disease. Med Res Rev, doi:10.1002/med.20248 [Epubl ahead of print].

[368] Marco-Contelles J, Soriano E (2011) The medicinal chem-istry of hybrid-based drugs targeting multiple sites ofaction. Curr Top Med Chem 11, 2714-2715.

[369] Bajda M, Guzior N, Ignasik M, Malawska B (2011) Multi-target-directed ligands in Alzheimer’s disease treatment.Curr Med Chem 18, 4949-4975.

[370] Rampa A, Belluti F, Gobbi S, Bisi A (2011) Hybrid-basedmulti-target ligands for the treatment of Alzheimer’s dis-ease. Curr Top Med Chem 11, 2716-2730.

[371] Melchiorre C, Andrisano V, Bolognesi ML, Budriesi R,Cavalli A, Cavrini V, Rosini M, Tumiatti V, Recana-tini M (1998) Acetylcholinesterase noncovalent inhibitorsbased on a polyamine backbone for potential use againstAlzheimer’s disease. J Med Chem 41, 4186-4189.

[372] Starks KM, Ortega-Vilain AC, Buccafusco JJ, PowersJC (1996) Novel pyridinium derivatives as inhibitors foracetylcholinesterase. J Enzyme Inhib 10, 27-45.

[373] Buccafusco JJ, Powers JC, Hernandez MA, PrendergastMA, Terry AV, Jonnala RR (2007) MHP-133, a drug withmultiple CNS targets: Potential for neuroprotection andenhanced cognition. Neurochem Res 32, 1224-1237.

[374] Borroni E, Damsma G, Giovacchini C, Mutel V, Jakob-Rotne R, Da PM (1994) A novel acetylcholinesteraseinhibitor, Ro 46-5934, which interacts with muscarinic M2receptors. Biochem Soc Trans 22, 755-758.

[375] Fang L, Jumpertz S, Zhang Y, Appenroth D, Fleck C,Mohr K, Trankle C, Decker M (2010) Hybrid moleculesfrom xanomeline and tacrine: Enhanced tacrine actions oncholinesterases and muscarinic M1 receptors. J Med Chem53, 2094-2103.

[376] Inestrosa NC, Alvarez A, Perez CA, Moreno RD, VicenteM, Linker C, Casanueva OI, Soto C, Garrido J (1996)Acetylcholinesterase accelerates assembly of amyloid-beta-peptides into Alzheimer’s fibrils: Possible role of theperipheral site of the enzyme. Neuron 16, 881-891.

[377] Alvarez A, Opazo C, Alarcon R, Garrido J, Inestrosa NC(1997) Acetylcholinesterase promotes the aggregation ofamyloid-beta-peptide fragments by forming a complexwith the growing fibrils. J Mol Biol 272, 348-361.

[378] Alvarez A, Alarcon R, Opazo C, Campos EO, MunozFJ, Calderon FH, Dajas F, Gentry MK, Doctor BP, DeMello FG, Inestrosa NC (1998) Stable complexes involv-ing acetylcholinesterase and amyloid-beta peptide changethe biochemical properties of the enzyme and increasethe neurotoxicity of Alzheimer’s fibrils. J Neurosci 18,3213-3223.

Page 62: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

608 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

[379] De Ferrari GV, Canales MA, Shin I, Weiner LM, SilmanI, Inestrosa NC (2001) A structural motif of acetyl-cholinesterase that promotes amyloid beta-peptide fibrilformation. Biochemistry 40, 10447-10457.

[380] Bartolini M, Bertucci C, Cavrini V, Andrisano V (2003)beta-Amyloid aggregation induced by human acetyl-cholinesterase: Inhibition studies. Biochem Pharmacol 65,407-416.

[381] Munoz-Ruiz P, Rubio L, Garcia-Palomero E, DorronsoroI, del Monte-Millan M, Valenzuela R, Usan P, de AC, Bar-tolini M, Andrisano V, Bidon-Chanal A, Orozco M, LuqueFJ, Medina M, Martinez A (2005) Design, synthesis,and biological evaluation of dual binding site acetyl-cholinesterase inhibitors: New disease-modifying agentsfor Alzheimer’s disease. J Med Chem 48, 7223-7233.

[382] Castro A, Martinez A (2001) Peripheral and dual bind-ing site acetylcholinesterase inhibitors: Implications intreatment of Alzheimer’s disease. Mini Rev Med Chem 1,267-272.

[383] Belluti F, Rampa A, Piazzi L, Bisi A, Gobbi S, BartoliniM, Andrisano V, Cavalli A, Recanatini M, Valenti P (2005)Cholinesterase inhibitors: Xanthostigmine derivativesblocking the acetylcholinesterase-induced beta-amyloidaggregation. J Med Chem 48, 4444-4456.

[384] Camps P, Formosa X, Munoz-Torrero D, Petrignet J, BadiaA, Clos MV (2005) Synthesis and pharmacological evalu-ation of huprine-tacrine heterodimers: Subnanomolar dualbinding site acetylcholinesterase inhibitors. J Med Chem48, 1701-1704.

[385] del Monte-Millan M, Garcia-Palomero E, Valenzuela R,Usan P, de AC, Munoz-Ruiz P, Rubio L, Dorronsoro I,Martinez A, Medina M (2006) Dual binding site acetyl-cholinesterase inhibitors: Potential new disease-modifyingagents for AD. J Mol Neurosci 30, 85-88.

[386] Garcia-Palomero E, Munoz P, Usan P, Garcia P, DelgadoE, De AC, Valenzuela R, Rubio L, Medina M, MartinezA (2008) Potent beta-amyloid modulators. NeurodegenerDis 5, 153-156.

[387] Carvajal FJ, Inestrosa NC (2011) Interactions of AChEwith Abeta aggregates in Alzheimer’s brain: Therapeuticrelevance of IDN 5706. Front Mol Neurosci 4, 19.

[388] Fernandez-Bachiller MI, Perez C, Gonzalez-Munoz GC,Conde S, Lopez MG, Villarroya M, Garcia AG, Rodriguez-Franco MI (2010) Novel tacrine-8-hydroxyquinolinehybrids as multifunctional agents for the treatment ofAlzheimer’s disease, with neuroprotective, cholinergic,antioxidant, and copper-complexing properties. J MedChem 53, 4927-4937.

[389] Antequera D, Bolos M, Spuch C, Pascual C, Ferrer I,Fernandez-Bachiller MI, Rodriguez-Franco MI, Carro E(2012) Effects of a tacrine-8-hydroxyquinoline hybrid(IQM-622) on Abeta accumulation and cell death: Involve-ment in hippocampal neuronal loss in Alzheimer’s disease.Neurobiol Dis 46, 682-691.

[390] Kapkova P, Alptuzun V, Frey P, Erciyas E, Holzgrabe U(2006) Search for dual function inhibitors for Alzheimer’sdisease: Synthesis and biological activity of acetyl-cholinesterase inhibitors of pyridinium-type and theirAbeta fibril formation inhibition capacity. Bioorg MedChem 14, 472-478.

[391] Kwon YE, Park JY, No KT, Shin JH, Lee SK, Eun JS,Yang JH, Shin TY, Kim DK, Chae BS, Leem JY, KimKH (2007) Synthesis, in vitro assay, and molecular mod-eling of new piperidine derivatives having dual inhibitorypotency against acetylcholinesterase and Abeta1-42 aggre-

gation for Alzheimer’s disease therapeutics. Bioorg MedChem 15, 6596-6607.

[392] Camps P, Formosa X, Galdeano C, Gomez T, Munoz-Torrero D, Scarpellini M, Viayna E, Badia A, Clos MV,Camins A, Pallas M, Bartolini M, Mancini F, AndrisanoV, Estelrich J, Lizondo M, Bidon-Chanal A, Luque FJ(2008) Novel donepezil-based inhibitors of acetyl- andbutyrylcholinesterase and acetylcholinesterase-inducedbeta-amyloid aggregation. J Med Chem 51, 3588-3598.

[393] Camps P, Formosa X, Galdeano C, Munoz-Torrero D,Ramirez L, Gomez E, Isambert N, Lavilla R, Badia A,Clos MV, Bartolini M, Mancini F, Andrisano V, ArceMP, Rodriguez-Franco MI, Huertas O, Dafni T, Luque FJ(2009) Pyrano[3,2-c]quinoline-6-chlorotacrine hybrids asa novel family of acetylcholinesterase- and beta-amyloid-directed anti-Alzheimer compounds. J Med Chem 52,5365-5379.

[394] Alptuzun V, Prinz M, Horr V, Scheiber J, RadackiK, Fallarero A, Vuorela P, Engels B, BraunschweigH, Erciyas E, Holzgrabe U (2010) Interaction of(benzylidene-hydrazono)-1,4-dihydropyridines with beta-amyloid, acetylcholine, and butyrylcholine esterases.Bioorg Med Chem 18, 2049-2059.

[395] Bolognesi ML, Bartolini M, Mancini F, Chiriano G, Cec-carini L, Rosini M, Milelli A, Tumiatti V, AndrisanoV, Melchiorre C (2010) Bis(7)-tacrine derivatives asmultitarget-directed ligands: Focus on anticholinesteraseand antiamyloid activities. ChemMedChem 5, 1215-1220.

[396] Viayna E, Gomez T, Galdeano C, Ramirez L, Ratia M,Badia A, Clos MV, Verdaguer E, Junyent F, Camins A,Pallas M, Bartolini M, Mancini F, Andrisano V, Arce MP,Rodriguez-Franco MI, Bidon-Chanal A, Luque FJ, CampsP, Munoz-Torrero D (2010) Novel huprine derivatives withinhibitory activity toward beta-amyloid aggregation andformation as disease-modifying anti-Alzheimer drug can-didates. ChemMedChem 5, 1855-1870.

[397] Mohamed T, Zhao X, Habib LK, Yang J, Rao PP(2011) Design, synthesis and structure-activity rela-tionship (SAR) studies of 2,4-disubstituted pyrimidinederivatives: Dual activity as cholinesterase and Abeta-aggregation inhibitors. Bioorg Med Chem 19, 2269-2281.

[398] Mohamed T, Yeung JC, Rao PP (2011) Develop-ment of 2-substituted-N-(naphth-1-ylmethyl) and N-benzhydrylpyrimidin-4-amines as dual cholinesterase andAbeta-aggregation inhibitors: Synthesis and biologicalevaluation. Bioorg Med Chem Lett 21, 5881-5887.

[399] Shi A, Huang L, Lu C, He F, Li X (2011) Synthe-sis, biological evaluation and molecular modeling ofnovel triazole-containing berberine derivatives as acetyl-cholinesterase and beta-amyloid aggregation inhibitors.Bioorg Med Chem 19, 2298-2305.

[400] Galdeano C, Viayna E, Sola I, Formosa X, Camps P, BadiaA, Clos MV, Relat. J, Ratia M, Bartolini M, Mancini F,Andrisano V, Salmona M, Minguillon C, Gonzalez-MunozGC, Rodriguez-Franco MI, Bidon-Chanal A, Luque FJ,Munoz-Torrero D (2012) Huprine-tacrine heterodimersas anti-amyloidogenic compounds of potential interestagainst Alzheimer’s and prion diseases. J Med Chem 55,661-669.

[401] Munoz-Torrero D, Pera M, Relat J, Ratia M, Galdeano C,Viayna E, Sola I, Formosa X, Camps P, Badia A, ClosMV (2012) Expanding the multipotent profile of huprine-tacrine heterodimers as disease-modifying anti-Alzheimeragents. Neurodegener Dis 10, 96-99.

Page 63: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 609

[402] Yan JW, Li YP, Ye WJ, Chen SB, Hou JQ, Tan JH, Ou TM,Li D, Gu LQ, Huang ZS (2012) Design, synthesis and eval-uation of isaindigotone derivatives as dual inhibitors foracetylcholinesterase and amyloid beta aggregation. BioorgMed Chem 20, 2527-2534.

[403] Tang H, Zhao HT, Zhong SM, Wang ZY, Chen ZF, Liang H(2012) Novel oxoisoaporphine-based inhibitors of acetyl-and butyrylcholinesterase and acetylcholinesterase-induced beta-amyloid aggregation. Bioorg Med ChemLett 22, 2257-2261.

[404] Shan WJ, Huang L, Zhou Q, Meng FC, Li XS (2011)Synthesis, biological evaluation of 9-N-substituted berber-ine derivatives as multi-functional agents of antioxidant,inhibitors of acetylcholinesterase, butyrylcholinesteraseand amyloid-beta aggregation. Eur J Med Chem 46, 5885-5893.

[405] Rosini M, Andrisano V, Bartolini M, Bolognesi ML, HreliaP, Minarini A, Tarozzi A, Melchiorre C (2005) Rationalapproach to discover multipotent anti-Alzheimer drugs.J Med Chem 48, 360-363.

[406] Packer L (1998) alpha-Lipoic acid: A metabolic antioxi-dant which regulates NF-kappa B signal transduction andprotects against oxidative injury. Drug Metab Rev 30, 245-275.

[407] Holmquist L, Stuchbury G, Berbaum K, Muscat S, YoungS, Hager K, Engel J, Munch G (2007) Lipoic acid as a noveltreatment for Alzheimer’s disease and related dementias.Pharmacol Ther 113, 154-164.

[408] Maczurek A, Hager K, Kenklies M, Sharman M, Mar-tins R, Engel J, Carlson DA, Munch G (2008) Lipoic acidas an anti-inflammatory and neuroprotective treatment forAlzheimer’s disease. Adv Drug Deliv Rev 60, 1463-1470.

[409] Zhang L, Xing GQ, Barker JL, Chang Y, Maric D, Ma W,Li BS, Rubinow DR (2001) Alpha-lipoic acid protects ratcortical neurons against cell death induced by amyloid andhydrogen peroxide through the Akt signalling pathway.Neurosci Lett 312, 125-128.

[410] Cavalli A, Bolognesi ML, Capsoni S, Andrisano V,Bartolini M, Margotti E, Cattaneo A, Recanatini M,Melchiorre C (2007) A small molecule targeting the mul-tifactorial nature of Alzheimer’s disease. Angew Chem IntEd Engl 46, 3689-3692.

[411] Bolognesi ML, Cavalli A, Melchiorre C (2009) Mem-oquin: A multi-target-directed ligand as an innovativetherapeutic opportunity for Alzheimer’s disease. Neu-rotherapeutics 6, 152-162.

[412] Bolognesi ML, Cavalli A, Melchiorre C (2009) Mem-oquin: A multi-target-directed ligand as an innovativetherapeutic opportunity for Alzheimer’s disease. Neu-rotherapeutics 6, 152-162.

[413] Bolognesi ML, Bartolini M, Rosini M, Andrisano V,Melchiorre C (2009) Structure-activity relationships ofmemoquin: Influence of the chain chirality in the multi-target mechanism of action. Bioorg Med Chem Lett 19,4312-4315.

[414] Bolognesi ML, Chiriano G, Bartolini M, Mancini F,Bottegoni G, Maestri V, Czvitkovich S, Windisch M,Cavalli A, Minarini A, Rosini M, Tumiatti V, Andrisano V,Melchiorre C (2011) Synthesis of monomeric derivativesto probe memoquin’s bivalent interactions. J Med Chem54, 8299-8304.

[415] Bolognesi ML, Bartolini M, Tarozzi A, Morroni F, LizziF, Milelli A, Minarini A, Rosini M, Hrelia P, Andrisano V,Melchiorre C (2011) Multitargeted drugs discovery: Bal-ancing anti-amyloid and anticholinesterase capacity in a

single chemical entity. Bioorg Med Chem Lett 21, 2655-2658.

[416] Rodriguez-Franco MI, Fernandez-Bachiller MI, PerezC, Hernandez-Ledesma B, Bartolome B (2006) Noveltacrine-melatonin hybrids as dual-acting drugs forAlzheimer disease, with improved acetylcholinesteraseinhibitory and antioxidant properties. J Med Chem 49,459-462.

[417] Fernandez-Bachiller MI, Perez C, Campillo NE, PaezJA, Gonzalez-Munoz GC, Usan P, Garcia-Palomero E,Lopez MG, Villarroya M, Garcia AG, Martinez A,Rodriguez-Franco MI (2009) Tacrine-melatonin hybridsas multifunctional agents for Alzheimer’s disease, withcholinergic, antioxidant, and neuroprotective properties.ChemMedChem 4, 828-841.

[418] Gonzalez-Munoz GC, Arce MP, Lopez B, Perez C,Villarroya M, Lopez MG, Garcia AG, Conde S,Rodriguez-Franco MI (2010) Old phenothiazine anddibenzothiadiazepine derivatives for tomorrow’s neuro-protective therapies against neurodegenerative diseases.Eur J Med Chem 45, 6152-6158.

[419] Gonzalez-Munoz GC, Arce MP, Lopez B, PerezC, Romero A, del BL, Martin-de-Saavedra MD,Egea J, Leon R, Villarroya M, Lopez MG,Garcia AG, Conde S, Rodriguez-Franco MI (2011)N-acylaminophenothiazines: Neuroprotective agentsdisplaying multifunctional activities for a potentialtreatment of Alzheimer’s disease. Eur J Med Chem 46,2224-2235.

[420] Fang L, Kraus B, Lehmann J, Heilmann J, Zhang Y, DeckerM (2008) Design and synthesis of tacrine-ferulic acidhybrids as multi-potent anti-Alzheimer drug candidates.Bioorg Med Chem Lett 18, 2905-2909.

[421] Salga SM, Ali HM, Abdullah MA, Abdelwahab SI,Wai LK, Buckle MJ, Sukumaran SD, Hadi AH (2011)Synthesis, characterization, acetylcholinesterase inhibi-tion, molecular modeling and antioxidant activitiesof some novel Schiff bases derived from 1-(2-ketoiminoethyl)piperazines. Molecules 16, 9316-9330.

[422] Shan WJ, Huang L, Zhou Q, Meng FC, Li XS (2011)Synthesis, biological evaluation of 9-N-substituted berber-ine derivatives as multi-functional agents of antioxidant,inhibitors of acetylcholinesterase, butyrylcholinesteraseand amyloid-beta aggregation. Eur J Med Chem 46, 5885-5893.

[423] Piazzi L, Cavalli A, Colizzi F, Belluti F, Bartolini M,Mancini F, Recanatini M, Andrisano V, Rampa A (2008)Multi-target-directed coumarin derivatives: hAChE andBACE1 inhibitors as potential anti-Alzheimer compounds.Bioorg Med Chem Lett 18, 423-426.

[424] Zhu Y, Xiao K, Ma L, Xiong B, Fu Y, Yu H, Wang W, WangX, Hu D, Peng H, Li J, Gong Q, Chai Q, Tang X, ZhangH, Li J, Shen J (2009) Design, synthesis and biologicalevaluation of novel dual inhibitors of acetylcholinesteraseand beta-secretase. Bioorg Med Chem 17, 1600-1613.

[425] Fernandez-Bachiller MI, Perez C, Monjas L, RademannJ, Rodriguez-Franco MI (2012) New tacrine-4-oxo-4H-chromene hybrids as multifunctional agents for thetreatment of Alzheimer’s disease, with cholinergic, antiox-idant, and beta-amyloid-reducing properties. J Med Chem55, 1303-1317.

[426] Huang W, Tang L, Shi Y, Huang S, Xu L, Sheng R,Wu P, Li J, Zhou N, Hu Y (2011) Searching for theMulti-Target-Directed Ligands against Alzheimer’s dis-ease: Discovery of quinoxaline-based hybrid compounds

Page 64: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

610 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

with AChE, H(3)R and BACE 1 inhibitory activities.Bioorg Med Chem 19, 7158-7167.

[427] Vezenkov L, Sevalle J, Danalev D, Ivanov T, BakalovaA, Georgieva M, Checler F (2012) Galantamine-basedhybrid molecules with acetylcholinesterase, butyryl-cholinesterase and gamma-secretase inhibition activities.Curr Alzheimer Res 9, 600-605.

[428] Liu J, Ho W, Lee NT, Carlier PR, Pang Y, Han Y (2000)Bis(7)-tacrine, a novel acetylcholinesterase inhibitor,reverses AF64A-induced deficits in navigational memoryin rats. Neurosci Lett 282, 165-168.

[429] Wu DC, Xiao XQ, Ng AK, Chen PM, Chung W, LeeNT, Carlier PR, Pang YP, Yu AC, Han YF (2000) Pro-tection against ischemic injury in primary cultured mouseastrocytes by bis(7)-tacrine, a novel acetylcholinesteraseinhibitor [corrected]. Neurosci Lett 288, 95-98.

[430] Xiao XQ, Lee NT, Carlier PR, Pang Y, Han YF(2000) Bis(7)-tacrine, a promising anti-Alzheimer’s agent,reduces hydrogen peroxide-induced injury in rat pheochro-mocytoma cells: Comparison with tacrine. Neurosci Lett290, 197-200.

[431] Li W, Pi R, Chan HH, Fu H, Lee NT, Tsang HW, Pu Y,Chang DC, Li C, Luo J, Xiong K, Li Z, Xue H, Car-lier PR, Pang Y, Tsim KW, Li M, Han Y (2005) Noveldimeric acetylcholinesterase inhibitor bis7-tacrine, but notdonepezil, prevents glutamate-induced neuronal apoptosisby blocking N-methyl-D-aspartate receptors. J Biol Chem280, 18179-18188.

[432] Fu H, Li W, Lao Y, Luo J, Lee NT, Kan KK, Tsang HW,Tsim KW, Pang Y, Li Z, Chang DC, Li M, Han Y (2006)Bis(7)-tacrine attenuates beta amyloid-induced neuronalapoptosis by regulating L-type calcium channels. J Neu-rochem 98, 1400-1410.

[433] Li W, Mak M, Jiang H, Wang Q, Pang Y, Chen K, HanY (2009) Novel anti-Alzheimer’s dimer Bis(7)-cognitin:Cellular and molecular mechanisms of neuroprotec-tion through multiple targets. Neurotherapeutics 6, 187-201.

[434] Orozco C, de Los RC, Arias E, Leon R, Gar-cia AG, Marco JL, Villarroya M, Lopez MG (2004)ITH4012 (ethyl 5-amino-6,7,8,9-tetrahydro-2-methyl-4-phenylbenzol[1,8]naphthyridine-3-car boxylate), a novelacetylcholinesterase inhibitor with “calcium promotor”and neuroprotective properties. J Pharmacol Exp Ther310, 987-994.

[435] Marco-Contelles J, Leon R, de Los RC, Guglietta A,Terencio J, Lopez MG, Garcia AG, Villarroya M (2006)Novel multipotent tacrine-dihydropyridine hybrids withimproved acetylcholinesterase inhibitory and neuropro-tective activities as potential drugs for the treatment ofAlzheimer’s disease. J Med Chem 49, 7607-7610.

[436] Marco-Contelles J, Leon R, de Los RC, Samadi A, Bar-tolini M, Andrisano V, Huertas O, Barril X, Luque FJ,Rodriguez-Franco MI, Lopez B, Lopez MG, Garcia AG,Carreiras MC, Villarroya M (2009) Tacripyrines, the firsttacrine-dihydropyridine hybrids, as multitarget-directedligands for the treatment of Alzheimer’s disease. J MedChem 52, 2724-2732.

[437] Pereira JD, Caricati-Neto A, Miranda-Ferreira R, SmailiSS, Godinho RO, de Los RC, Leon R, Villaroya M,Samadi A, Marco-Contelles J, Jurkiewicz NH, GarciaAG, Jurkiewicz A (2011) Effects of novel tacripyrinesITH12117 and ITH12118 on rat vas deferens contrac-tions, calcium transients and cholinesterase activity. EurJ Pharmacol 660, 411-419.

[438] Bartolini M, Pistolozzi M, Andrisano V, Egea J, LopezMG, Iriepa I, Moraleda I, Galvez E, Marco-ContellesJ, Samadi A (2011) Chemical and pharmacologicalstudies on enantiomerically pure p-methoxytacripyrines,promising multi-target-directed ligands for the treat-ment of Alzheimer’s disease. ChemMedChem 6, 1990-1997.

[439] Valero T, del BL, Egea J, Canas N, Martinez A, GarciaAG, Villarroya M, Lopez MG (2009) NP04634 preventscell damage caused by calcium overload and mitochondrialdisruption in bovine chromaffin cells. Eur J Pharmacol607, 47-53.

[440] Degroot A, Kofalvi A, Wade MR, Davis RJ, Rodrigues RJ,Rebola N, Cunha RA, Nomikos GG (2006) CB1 receptorantagonism increases hippocampal acetylcholine release:Site and mechanism of action. Mol Pharmacol 70, 1236-1245.

[441] Lange JH, Coolen HK, van Stuivenberg HH, Dijks-man JA, Herremans AH, Ronken E, Keizer HG, TipkerK, McCreary AC, Veerman W, Wals HC, Stork B,Verveer PC, den Hartog AP, de Jong NM, Adolfs TJ,Hoogendoorn J, Kruse CG (2004) Synthesis, biologi-cal properties, and molecular modeling investigations ofnovel 3,4-diarylpyrazolines as potent and selective CB(1)cannabinoid receptor antagonists. J Med Chem 47, 627-643.

[442] Lange JH, van Stuivenberg HH, Coolen HK, AdolfsTJ, McCreary AC, Keizer HG, Wals HC, Veerman W,Borst AJ, de LW, Verveer PC, Kruse CG (2005) Bioisos-teric replacements of the pyrazole moiety of rimonabant:Synthesis, biological properties, and molecular modelinginvestigations of thiazoles, triazoles, and imidazoles aspotent and selective CB1 cannabinoid receptor antago-nists. J Med Chem 48, 1823-1838.

[443] Lange JH, Coolen HK, van der Neut MA, Borst AJ, StorkB, Verveer PC, Kruse CG (2010) Design, synthesis, bio-logical properties, and molecular modeling investigationsof novel tacrine derivatives with a combination of acetyl-cholinesterase inhibition and cannabinoid CB1 receptorantagonism. J Med Chem 53, 1338-1346.

[444] Benito C, Nunez E, Tolon RM, Carrier EJ, Rabano A,Hillard CJ, Romero J (2003) Cannabinoid CB2 receptorsand fatty acid amide hydrolase are selectively overex-pressed in neuritic plaque-associated glia in Alzheimer’sdisease brains. J Neurosci 23, 11136-11141.

[445] Rampa A, Bartolini M, Bisi A, Belluti F, Gobbi S,Andrisano V, Ligresti A, Di Marzo V (2012) The first dualChE/FAAH inhibitors: New perspectives for Alzheimer’sdisease? ACS Med Chem Lett 3, 182-186.

[446] Lynch AM, Loane DJ, Minogue AM, Clarke RM, Kil-roy D, Nally RE, Roche OJ, O’Connell F, Lynch MA(2007) Eicosapentaenoic acid confers neuroprotection inthe amyloid-beta challenged aged hippocampus. Neuro-biol Aging 28, 845-855.

[447] Minogue AM, Lynch AM, Loane DJ, Herron CE, LynchMA (2007) Modulation of amyloid-beta-induced andage-associated changes in rat hippocampus by eicosapen-taenoic acid. J Neurochem 103, 914-926.

[448] Hashimoto M, Hossain S, Tanabe Y, Kawashima A, HaradaT, Yano T, Mizuguchi K, Shido O (2009) The protectiveeffect of dietary eicosapentaenoic acid against impairmentof spatial cognition learning ability in rats infused withamyloid beta(1-40). J Nutr Biochem 20, 965-973.

[449] Yehuda S (2012) Polyunsaturated fatty acids as putativecognitive enhancers. Med Hypotheses 79, 456-461.

Page 65: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 611

[450] Petroianu G, Arafat K, Sasse BC, Stark H (2006) Multipleenzyme inhibitions by histamine H3 receptor antago-nists as potential procognitive agents. Pharmazie 61, 179-182.

[451] Incerti M, Flammini L, Saccani F, Morini G, CominiM, Coruzzi M, Barocelli E, Ballabeni V, Bertoni S(2010) Dual-acting drugs: An in vitro study of non-imidazole histamine H3 receptor antagonists combininganticholinesterase activity. ChemMedChem 5, 1143-1149.

[452] Huang W, Tang L, Shi Y, Huang S, Xu L, Sheng R,Wu P, Li J, Zhou N, Hu Y (2011) Searching for theMulti-Target-Directed Ligands against Alzheimer’s dis-ease: Discovery of quinoxaline-based hybrid compoundswith AChE, H(3)R and BACE 1 inhibitory activities.Bioorg Med Chem 19, 7158-7167.

[453] Saura J, Luque JM, Cesura AM, Da PM, Chan-PalayV, Huber G, Loffler J, Richards JG (1994) Increasedmonoamine oxidase B activity in plaque-associated astro-cytes of Alzheimer brains revealed by quantitative enzymeradioautography. Neuroscience 62, 15-30.

[454] Fink DM, Palermo MG, Bores GM, Huger FP, Kurys BE,Merriman MC, Olsen GE, Petko W, O’Malley GJ (1996)Imino 1,2,3,4-tetrahydrocylopent [b]indole carbamates asdual inhibitors of acetylcholinesterase and monoamineoxidase. Bioorg Med Chem Lett 6, 625-630.

[455] Bruhlmann C, Ooms F, Carrupt PA, Testa B, Catto M,Leonetti F, Altomare C, Carotti A (2001) Coumarinsderivatives as dual inhibitors of acetylcholinesterase andmonoamine oxidase. J Med Chem 44, 3195-3198.

[456] Weinstock M, Bejar C, Wang RH, Poltyrev T, Gross A,Finberg JP, Youdim MB (2000) TV3326, a novel neuropro-tective drug with cholinesterase and monoamine oxidaseinhibitory activities for the treatment of Alzheimer’s dis-ease. J Neural Transm Suppl 60, 157-169.

[457] Weinstock M, Kirschbaum-Slager N, Lazarovici P, BejarC, Youdim MB, Shoham S (2001) Neuroprotective effectsof novel cholinesterase inhibitors derived from rasagilineas potential anti-Alzheimer drugs. Ann N Y Acad Sci 939,148-161.

[458] Weinstock M, Luques L, Poltyrev T, Bejar C, Shoham S(2011) Ladostigil prevents age-related glial activation andspatial memory deficits in rats. Neurobiol Aging 32, 1069-1078.

[459] Sterling J, Herzig Y, Goren T, Finkelstein N, Lerner D,Goldenberg W, Miskolczi I, Molnar S, Rantal F, Tamas T,Toth G, Zagyva A, Zekany A, Finberg J, Lavian G, GrossA, Friedman R, Razin M, Huang W, Krais B, Chorev M,Youdim MB, Weinstock M (2002) Novel dual inhibitorsof AChE and MAO derived from hydroxy aminoindanand phenethylamine as potential treatment for Alzheimer’sdisease. J Med Chem 45, 5260-5279.

[460] Sagi Y, Weinstock M, Youdim MB (2003) Attenuation ofMPTP-induced dopaminergic neurotoxicity by TV3326, acholinesterase-monoamine oxidase inhibitor. J Neurochem86, 290-297.

[461] Youdim MB, Amit T, Bar-Am O, Weinstock M,Yogev-Falach M (2003) Amyloid processing and sig-nal transduction properties of antiparkinson-antialzheimerneuroprotective drugs rasagiline and TV3326. Ann N YAcad Sci 993, 378-386.

[462] Youdim MB, Amit T, Bar-Am O, Weinreb O, Yogev-Falach M (2006) Implications of co-morbidity for etiologyand treatment of neurodegenerative diseases with multi-functional neuroprotective-neurorescue drugs; ladostigil.Neurotox Res 10, 181-192.

[463] Youdim MB (2006) The path from anti Parkinson drugselegiline and rasagiline to multifunctional neuropro-tective anti Alzheimer drugs ladostigil and m30. CurrAlzheimer Res 3, 541-550.

[464] Poltyrev T, Gorodetsky E, Bejar C, Schorer-ApelbaumD, Weinstock M (2005) Effect of chronic treatment withladostigil (TV-3326) on anxiogenic and depressive-likebehaviour and on activity of the hypothalamic-pituitary-adrenal axis in male and female prenatally stressed rats.Psychopharmacology (Berl) 181, 118-125.

[465] Yogev-Falach M, Bar-Am O, Amit T, Weinreb O, YoudimMB (2006) A multifunctional, neuroprotective drug,ladostigil (TV3326), regulates holo-APP translation andprocessing. FASEB J 20, 2177-2179.

[466] Bar-Am O, Weinreb O, Amit T, Youdim MB (2009) Thenovel cholinesterase-monoamine oxidase inhibitor andantioxidant, ladostigil, confers neuroprotection in neu-roblastoma cells and aged rats. J Mol Neurosci 37, 135-145.

[467] Bar-Am O, Amit T, Weinreb O, Youdim MB, Mandel S(2010) Propargylamine containing compounds as mod-ulators of proteolytic cleavage of amyloid-beta proteinprecursor: Involvement of MAPK and PKC activation.J Alzheimers Dis 21, 361-371.

[468] Weinreb O, Amit T, Bar-Am O, Yogev-Falach M, YoudimMB (2008) The neuroprotective mechanism of action ofthe multimodal drug ladostigil. Front Biosci 13, 5131-5137.

[469] Weinreb O, Bar-Am O, Amit T, Drigues N, Sagi Y,Youdim MB (2008) The neuroprotective effect of ladostigilagainst hydrogen peroxide-mediated cytotoxicity. ChemBiol Interact 175, 318-326.

[470] Weinreb O, Mandel S, Bar-Am O, Yogev-Falach M,vramovich-Tirosh Y, Amit T, Youdim MB (2009) Mul-tifunctional neuroprotective derivatives of rasagiline asanti-Alzheimer’s disease drugs. Neurotherapeutics 6, 163-174.

[471] Weinreb O, Amit T, Bar-Am O, Youdim MB (2011) Anovel anti-Alzheimer’s disease drug, ladostigil neuropro-tective, multimodal brain-selective monoamine oxidaseand cholinesterase inhibitor. Int Rev Neurobiol 100, 191-215.

[472] Weinreb O, Amit T, Bar-Am O, Youdim MB (2012)Ladostigil: A novel multimodal neuroprotective drug withcholinesterase and brain-selective monoamine oxidaseinhibitory activities for Alzheimer’s disease treatment.Curr Drug Targets 13, 483-494.

[473] Samadi A, Chioua M, Bolea I, de Los RC, Iriepa I,Moraleda I, Bastida A, Esteban G, Unzeta M, GalvezE, Marco-Contelles J (2011) Synthesis, biological assess-ment and molecular modeling of new multipotent MAOand cholinesterase inhibitors as potential drugs for thetreatment of Alzheimer’s disease. Eur J Med Chem 46,4665-4668.

[474] Samadi A, de Los RC, Bolea I, Chioua M, Iriepa I,Moraleda I, Bartolini M, Andrisano V, Galvez E, ValderasC, Unzeta M, Marco-Contelles J (2012) MultipotentMAO and cholinesterase inhibitors for the treatment ofAlzheimer’s disease: Synthesis, pharmacological analy-sis and molecular modeling of heterocyclic substitutedalkyl and cycloalkyl propargyl amine. Eur J Med Chem52, 251-262.

[475] Bolea I, Juarez-Jimenez J, de los RC, Chioua M,Pouplana R, Luque FJ, Unzeta M, Marco-Contelles J,Samadi A (2011) Synthesis, biological evaluation, and

Page 66: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

612 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

molecular modeling of donepezil and N-[(5-(benzyloxy)-1-methyl-1H-indol-2-yl)methyl]-N-methylprop-2-yn-1-amine hybrids as new multipotent cholinesterase/monoamine oxidase inhibitors for the treatment ofAlzheimer’s disease. J Med Chem 54, 8251-8270.

[476] Zheng H, Weiner LM, Bar-Am O, Epsztejn S, CabantchikZI, Warshawsky A, Youdim MB, Fridkin M (2005)Design, synthesis, and evaluation of novel bifunctionaliron-chelators as potential agents for neuroprotection inAlzheimer’s, Parkinson’s, and other neurodegenerativediseases. Bioorg Med Chem 13, 773-783.

[477] Zheng H, Youdim MB, Fridkin M (2009) Site-activatedmultifunctional chelator with acetylcholinesterase andneuroprotective-neurorestorative moieties for Alzheimer’stherapy. J Med Chem 52, 4095-4098.

[478] Zheng H, Fridkin M, Youdim MB (2012) Novel chela-tors targeting cell cycle arrest, acetylcholinesterase, andmonoamine oxidase for Alzheimer’s therapy. Curr DrugTargets 13, 1089-1106.

[479] Meng FC, Mao F, Shan WJ, Qin F, Huang L, Li XS (2012)Design, synthesis, and evaluation of indanone deriva-tives as acetylcholinesterase inhibitors and metal-chelatingagents. Bioorg Med Chem Lett 22, 4462-4466.

[480] Rosini M, Simoni E, Bartolini M, Cavalli A, Cecca-rini L, Pascu N, McClymont DW, Tarozzi A, BolognesiML, Minarini A, Tumiatti V, Andrisano V, Mellor IR,Melchiorre C (2008) Inhibition of acetylcholinesterase,beta-amyloid aggregation, and NMDA receptors inAlzheimer’s disease: A promising direction for the multi-target-directed ligands gold rush. J Med Chem 51,4381-4384.

[481] Rook Y, Schmidtke KU, Gaube F, Schepmann D, WunschB, Heilmann J, Lehmann J, Winckler T (2010) Bivalentbeta-carbolines as potential multitarget anti-Alzheimeragents. J Med Chem 53, 3611-3617.

[482] Le Texier L, Favre E, Redeuilh C, Blavet N, BellahseneT, Dive G, Pirotzky E, Godfroid JJ (1996) Structure-activity relationships in platelet-activating factor (PAF).7. Tetrahydrofuran derivatives as dual PAF antagonistsand acetylcholinesterase inhibitors. Synthesis and PAF-antagonistic activity. J Lipid Mediat Cell Signal 13,189-205.

[483] Le Texier L, Favre E, Ronzani N, Massicot F, BlavetN, Pirotzky E, Godfroid JJ (1996) Structure-activityrelationships in platelet-activating factor (PAF). 8.Tetrahydrofuran derivatives as dual PAF antagonists andacetylcholinesterase inhibitors: Anti-acetylcholinesteraseactivity and comparative SAR. J Lipid Mediat Cell Signal13, 207-222.

[484] Ezoulin MJ, Li J, Wu G, Dong CZ, Ombetta JE, ChenHZ, Massicot F, Heymans F (2005) Differential effect ofPMS777, a new type of acetylcholinesterase inhibitor, andgalanthamine on oxidative injury induced in human neu-roblastoma SK-N-SH cells. Neurosci Lett 389, 61-65.

[485] Ezoulin MJ, Dong CZ, Liu Z, Li J, Chen HZ, Hey-mans F, Lelievre L, Ombetta JE, Massicot F (2006)Study of PMS777, a new type of acetylcholinesteraseinhibitor, in human HepG2 cells. Comparison with tacrineand galanthamine on oxidative stress and mitochondrialimpairment. Toxicol In Vitro 20, 824-831.

[486] Ezoulin MJ, Liu Z, Dutertre-Catella H, Wu G, Dong CZ,Heymans F, Ombetta JE, Rat P, Massicot F (2007) Anew acetylcholinesterase inhibitor with anti-PAF activitymodulates oxidative stress and pro-inflammatory media-tors release in stimulated RAW 264.7 macrophage cells.

Comparison with tacrine. Int Immunopharmacol 7, 1685-1694.

[487] Li J, Huang H, Miezan Ezoulin JM, Gao XL, Massicot F,Dong CZ, Heymans F, Chen HZ (2007) Pharmacologicalprofile of PMS777, a new AChE inhibitor with PAF antag-onistic activity. Int J Neuropsychopharmacol 10, 21-29.

[488] Li J, Shao B, Zhu L, Cui Y, Dong C, Miezan Ezoulin JM,Gao X, Ren Q, Heymans F, Chen H (2008) PMS777, a bis-interacting ligand for PAF receptor antagonism and AChEinhibition, attenuates PAF-induced neurocytotoxicity inSH-SY5Y cells. Cell Mol Neurobiol 28, 125-136.

[489] Li J, Hu J, Shao B, Zhou W, Cui Y, Dong C, Ezoulin JM,Zhu X, Ding W, Heymans F, Chen H (2009) Protectionof PMS777, a new AChE inhibitor with PAF antagonism,against amyloid-beta-induced neuronal apoptosis and neu-roinflammation. Cell Mol Neurobiol 29, 589-595.

[490] Yang HQ, Sun ZK, Zhao YX, Pan J, Ba MW, Lu GQ,Ding JQ, Chen HZ, Chen SD (2009) PMS777, a newcholinesterase inhibitor with anti-platelet activated factoractivity, regulates amyloid precursor protein processing invitro. Neurochem Res 34, 528-535.

[491] Miezan Ezoulin JM, Shao BY, Xia Z, Xie Q, Li J, CuiYY, Wang H, Dong CZ, Zhao YX, Massicot F, Qiu ZB,Heymans F, Chen HZ (2009) Novel piperazine derivativePMS1339 exhibits tri-functional properties and cognitiveimprovement in mice. Int J Neuropsychopharmacol 12,1409-1419.

[492] Abe Y, Aoyagi A, Hara T, Abe K, Yamazaki R, KumagaeY, Naruto S, Koyama K, Marumoto S, Tago K, Toda N,Takami K, Yamada N, Ori M, Kogen H, Kaneko T (2003)Pharmacological characterization of RS-1259, an orallyactive dual inhibitor of acetylcholinesterase and serotonintransporter, in rodents: Possible treatment of Alzheimer’sdisease. J Pharmacol Sci 93, 95-105.

[493] Toda N, Tago K, Marumoto S, Takami K, Ori M, YamadaN, Koyama K, Naruto S, Abe K, Yamazaki R, Hara T,Aoyagi A, Abe Y, Kaneko T, Kogen H (2003) Design, syn-thesis and structure-activity relationships of dual inhibitorsof acetylcholinesterase and serotonin transporter as poten-tial agents for Alzheimer’s disease. Bioorg Med Chem 11,1935-1955.

[494] Toda N, Tago K, Marumoto S, Takami K, Ori M, YamadaN, Koyama K, Naruto S, Abe K, Yamazaki R, Hara T, Aoy-agi A, Abe Y, Kaneko T, Kogen H (2003) A conformationalrestriction approach to the development of dual inhibitorsof acetylcholinesterase and serotonin transporter as poten-tial agents for Alzheimer’s disease. Bioorg Med Chem 11,4389-4415.

[495] Toda N, Kaneko T, Kogen H (2010) Development of anefficient therapeutic agent for Alzheimer’s disease: Designand synthesis of dual inhibitors of acetylcholinesterase andserotonin transporter. Chem Pharm Bull (Tokyo) 58, 273-287.

[496] Lecanu L, Tillement L, McCourty A, RammouzG, Yao W, Greeson J, Papadopoulos V (2010)Dimethyl-carbamic acid 2,3-Bis-Dimethylcarbamoyloxy-6-(4-Ethyl-Piperazine-1-Carbonyl)-phenyl ester: A novelmulti-target therapeutic approach to neuroprotection. MedChem 6, 123-140.

[497] Panza F, Solfrizzi V, Frisardi V, Capurso C, D’Introno A,Colacicco AM, Vendemiale G, Capurso A, Imbimbo BP(2009) Disease-modifying approach to the treatment ofAlzheimer’s disease: From alpha-secretase activators togamma-secretase inhibitors and modulators. Drugs Aging26, 537-555.

Page 67: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 613

[498] Vincent B, Govitrapong P (2011) Activation of the alpha-secretase processing of AbetaPP as a therapeutic approachin Alzheimer’s disease. J Alzheimers Dis 24(Suppl 2), 75-94.

[499] Frisardi V, Solfrizzi V, Imbimbo PB, Capurso C, D’IntronoA, Colacicco AM, Vendemiale G, Seripa D, Pilotto A,Capurso A, Panza F (2010) Towards disease-modifyingtreatment of Alzheimer’s disease: Drugs targeting beta-amyloid. Curr Alzheimer Res 7, 40-55.

[500] Endres K, Fahrenholz F (2012) Regulation of alpha-secretase ADAM10 expression and activity. Exp Brain Res217, 343-352.

[501] Postina R (2012) Activation of alpha-secretase cleavage.J Neurochem 120(Suppl 1), 46-54.

[502] Epis R, Marcello E, Gardoni F, Luca MD (2012) Alpha,beta-and gamma-secretases in alzheimer’s disease. FrontBiosci (Schol Ed) 4, 1126-1150.

[503] Marcade M, Bourdin J, Loiseau N, Peillon H, RayerA, Drouin D, Schweighoffer F, Desire L (2008) Etazo-late, a neuroprotective drug linking GABA(A) receptorpharmacology to amyloid precursor protein processing.J Neurochem 106, 392-404.

[504] Drott J, Desire L, Drouin D, Pando M, Haun F (2010)Etazolate improves performance in a foraging and homingtask in aged rats. Eur J Pharmacol 634, 95-100.

[505] Vellas B, Sol O, Snyder PJ, Ousset PJ, Haddad R, MaurinM, Lemarie JC, Desire L, Pando MP (2011) EHT0202in Alzheimer’s disease: A 3-month, randomized, placebo-controlled, double-blind study. Curr Alzheimer Res 8, 203-212.

[506] Panza F, Solfrizzi V, Frisardi V, Imbimbo BP, CapursoC, D’Introno A, Colacicco AM, Seripa D, Vendemiale G,Capurso A, Pilotto A (2009) Beyond the neurotransmitter-focused approach in treating Alzheimer’s disease: Drugstargeting beta-amyloid and tau protein. Aging Clin Exp Res21, 386-406.

[507] Ruan BF, Zhu HL (2012) The chemistry and biology ofthe bryostatins: Potential PKC inhibitors in clinical devel-opment. Curr Med Chem 19, 2652-2664.

[508] Vassar R, Kovacs DM, Yan R, Wong PC (2009) Thebeta-secretase enzyme BACE in health and Alzheimer’sdisease: Regulation, cell biology, function, and therapeuticpotential. J Neurosci 29, 12787-12794.

[509] Vassar R, Kandalepas PC (2011) The beta-secretaseenzyme BACE1 as a therapeutic target for Alzheimer’sdisease. Alzheimers Res Ther 3, 20.

[510] Vassar R, Bennett BD, Babu-Khan S, Kahn S, MendiazEA, Denis P, Teplow DB, Ross S, Amarante P, Loeloff R,Luo Y, Fisher S, Fuller J, Edenson S, Lile J, JarosinskiMA, Biere AL, Curran E, Burgess T, Louis JC, CollinsF, Treanor J, Rogers G, Citron M (1999) Beta-secretasecleavage of Alzheimer’s amyloid precursor protein by thetransmembrane aspartic protease BACE. Science 286, 735-741.

[511] Haniu M, Denis P, Young Y, Mendiaz EA, Fuller J, Hui JO,Bennett BD, Kahn S, Ross S, Burgess T, Katta V, Rogers G,Vassar R, Citron M (2000) Characterization of Alzheimer’sbeta -secretase protein BACE. A pepsin family memberwith unusual properties. J Biol Chem 275, 21099-21106.

[512] Sinha S, Anderson JP, Barbour R, Basi GS, CaccavelloR, Davis D, Doan M, Dovey HF, Frigon N, Hong J,Jacobson-Croak K, Jewett N, Keim P, Knops J, Lieber-burg I, Power M, Tan H, Tatsuno G, Tung J, Schenk D,Seubert P, Suomensaari SM, Wang S, Walker D, Zhao J,McConlogue L, John V (1999) Purification and cloning

of amyloid precursor protein beta-secretase from humanbrain. Nature 402, 537-540.

[513] Yan R, Bienkowski MJ, Shuck ME, Miao H, Tory MC,Pauley AM, Brashier JR, Stratman NC, Mathews WR,Buhl AE, Carter DB, Tomasselli AG, Parodi LA, Heinrik-son RL, Gurney ME (1999) Membrane-anchored aspartylprotease with Alzheimer’s disease beta-secretase activity.Nature 402, 533-537.

[514] Hussain I, Powell D, Howlett DR, Tew DG, Meek TD,Chapman C, Gloger IS, Murphy KE, Southan CD, RyanDM, Smith TS, Simmons DL, Walsh FS, Dingwall C,Christie G (1999) Identification of a novel aspartic protease(Asp 2) as beta-secretase. Mol Cell Neurosci 14, 419-427.

[515] Hong L, Koelsch G, Lin X, Wu S, Terzyan S, Ghosh AK,Zhang XC, Tang J (2000) Structure of the protease domainof memapsin 2 (beta-secretase) complexed with inhibitor.Science 290, 150-153.

[516] Citron M (2004) Beta-secretase inhibition for the treatmentof Alzheimer’s disease–promise and challenge. TrendsPharmacol Sci 25, 92-97.

[517] Venugopal C, Demos CM, Rao KS, Pappolla MA,Sambamurti K (2008) Beta-secretase: Structure, function,and evolution. CNS Neurol Disord Drug Targets 7, 278-294.

[518] Murphy T, Yip A, Brayne C, Easton D, Evans JG, XuerebJ, Cairns N, Esiri MM, Rubinsztein DC (2001) The BACEgene: Genomic structure and candidate gene study in late-onset Alzheimer’s disease. Neuroreport 12, 631-634.

[519] Hunt CE, Turner AJ (2009) Cell biology, regulation andinhibition of beta-secretase (BACE-1). FEBS J 276, 1845-1859.

[520] Ahmed RR, Holler CJ, Webb RL, Li F, Beckett TL,Murphy MP (2010) BACE1 and BACE2 enzymatic activi-ties in Alzheimer’s disease. J Neurochem 112, 1045-1053.

[521] Willem M, Garratt AN, Novak B, Citron M, Kaufmann S,Rittger A, DeStrooper B, Saftig P, Birchmeier C, HaassC (2006) Control of peripheral nerve myelination by thebeta-secretase BACE1. Science 314, 664-666.

[522] Willem M, Lammich S, Haass C (2009) Function, regula-tion and therapeutic properties of beta-secretase (BACE1).Semin Cell Dev Biol 20, 175-182.

[523] Hong L, Koelsch G, Lin X, Wu S, Terzyan S, Ghosh AK,Zhang XC, Tang J (2000) Structure of the protease domainof memapsin 2 (beta-secretase) complexed with inhibitor.Science 290, 150-153.

[524] Hong L, Turner RT III, Koelsch G, Shin D, GhoshAK, Tang J (2002) Crystal structure of memapsin 2(beta-secretase) in complex with an inhibitor OM00-3.Biochemistry 41, 10963-10967.

[525] Patel S, Vuillard L, Cleasby A, Murray CW, Yon J (2004)Apo and inhibitor complex structures of BACE (beta-secretase). J Mol Biol 343, 407-416.

[526] Rojo I, Martin JA, Broughton H, Timm D, Erickson J, YangHC, McCarthy JR (2006) Macrocyclic peptidomimeticinhibitors of beta-secretase (BACE): First X-ray structureof a macrocyclic peptidomimetic-BACE complex. BioorgMed Chem Lett 16, 191-195.

[527] Barrow JC, Stauffer SR, Rittle KE, Ngo PL, Yang Z,Selnick HG, Graham SL, Munshi S, McGaughey GB,Holloway MK, Simon AJ, Price EA, SankaranarayananS, Colussi D, Tugusheva K, Lai MT, Espeseth AS, Xu M,Huang Q, Wolfe A, Pietrak B, Zuck P, Levorse DA, HazudaD, Vacca JP (2008) Discovery and X-ray crystallographicanalysis of a spiropiperidine iminohydantoin inhibitor ofbeta-secretase. J Med Chem 51, 6259-6262.

Page 68: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

614 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

[528] Shimizu H, Tosaki A, Kaneko K, Hisano T, Sakurai T,Nukina N (2008) Crystal structure of an active form ofBACE1, an enzyme responsible for amyloid beta proteinproduction. Mol Cell Biol 28, 3663-3671.

[529] Roberds SL, Anderson J, Basi G, Bienkowski MJ,Branstetter DG, Chen KS, Freedman SB, Frigon NL,Games D, Hu K, Johnson-Wood K, Kappenman KE,Kawabe TT, Kola I, Kuehn R, Lee M, Liu W, Motter R,Nichols NF, Power M, Robertson DW, Schenk D, SchoorM, Shopp GM, Shuck ME, Sinha S, Svensson KA, Tat-suno G, Tintrup H, Wijsman J, Wright S, McConlogue L(2001) BACE knockout mice are healthy despite lackingthe primary beta-secretase activity in brain: Implicationsfor Alzheimer’s disease therapeutics. Hum Mol Genet 10,1317-1324.

[530] Harrison SM, Harper AJ, Hawkins J, Duddy G, Grau E,Pugh PL, Winter PH, Shilliam CS, Hughes ZA, DawsonLA, Gonzalez MI, Upton N, Pangalos MN, Dingwall C(2003) BACE1 (beta-secretase) transgenic and knockoutmice: Identification of neurochemical deficits and behav-ioral changes. Mol Cell Neurosci 24, 646-655.

[531] Ohno M, Sametsky EA, Younkin LH, Oakley H, YounkinSG, Citron M, Vassar R, Disterhoft JF (2004) BACE1deficiency rescues memory deficits and cholinergic dys-function in a mouse model of Alzheimer’s disease. Neuron41, 27-33.

[532] Jonsson T, Atwal JK, Steinberg S, Snaedal J, JonssonPV, Bjornsson S, Stefansson H, Sulem P, Gudbjartsson D,Maloney J, Hoyte K, Gustafson A, Liu Y, Lu Y, BhangaleT, Graham RR, Huttenlocher J, Bjornsdottir G, AndreassenOA, Jonsson EG, Palotie A, Behrens TW, MagnussonOT, Kong A, Thorsteinsdottir U, Watts RJ, Stefansson K(2012) A mutation in APP protects against Alzheimer’sdisease and age-related cognitive decline. Nature 488,96-99.

[533] Callaway E (2012) Gene mutation defends againstAlzheimer’s disease. Nature 487, 153.

[534] Dingwall C (2001) Spotlight on BACE: The secretases astargets for treatment in Alzheimer disease. J Clin Invest108, 1243-1246.

[535] Vassar R (2001) The beta-secretase, BACE: A prime drugtarget for Alzheimer’s disease. J Mol Neurosci 17, 157-170.

[536] Ghosh AK, Bilcer G, Harwood C, Kawahama R, Shin D,Hussain KA, Hong L, Loy JA, Nguyen C, Koelsch G,Ermolieff J, Tang J (2001) Structure-based design: Potentinhibitors of human brain memapsin 2 (beta-secretase).J Med Chem 44, 2865-2868.

[537] Citron M (2002) Beta-secretase as a target for the treatmentof Alzheimer’s disease. J Neurosci Res 70, 373-379.

[538] Citron M (2002) Emerging Alzheimer’s disease therapies:Inhibition of beta-secretase. Neurobiol Aging 23, 1017-1022.

[539] Dewachter I, Van Leuven F (2002) Secretases as targetsfor the treatment of Alzheimer’s disease: The prospects.Lancet Neurol 1, 409-416.

[540] Ghosh AK, Hong L, Tang J (2002) Beta-secretase as atherapeutic target for inhibitor drugs. Curr Med Chem 9,1135-1144.

[541] Hong L, Turner RT III, Koelsch G, Ghosh AK, Tang J(2002) Memapsin 2 (beta-secretase) as a therapeutic target.Biochem Soc Trans 30, 530-534.

[542] Roggo S (2002) Inhibition of BACE, a promising approachto Alzheimer’s disease therapy. Curr Top Med Chem 2,359-370.

[543] Vassar R (2002) Beta-secretase (BACE) as a drug target forAlzheimer’s disease. Adv Drug Deliv Rev 54, 1589-1602.

[544] Conway KA, Baxter EW, Felsenstein KM, Reitz AB(2003) Emerging beta-amyloid therapies for the treatmentof Alzheimer’s disease. Curr Pharm Des 9, 427-447.

[545] John V, Beck JP, Bienkowski MJ, Sinha S, HeinriksonRL (2003) Human beta-secretase (BACE) and BACEinhibitors. J Med Chem 46, 4625-4630.

[546] Koelsch G, Turner RT III, Hong L, Ghosh AK, Tang J(2003) Memapsin 2, a drug target for Alzheimer’s disease.Biochem Soc Symp 70, 213-220.

[547] Tang J, Ghosh AK, Hong L, Koelsch G, Turner RT III,Chang W (2003) Study of memapsin 2 (beta-secretase) andstrategy of inhibitor design. J Mol Neurosci 20, 299-304.

[548] Cumming JN, Iserloh U, Kennedy ME (2004) Design anddevelopment of BACE-1 inhibitors. Curr Opin Drug Dis-cov Devel 7, 536-556.

[549] Hussain I (2004) The potential for BACE1 inhibitors inthe treatment of Alzheimer’s disease. IDrugs 7, 653-658.

[550] Vassar R (2004) BACE1: The beta-secretase enzyme inAlzheimer’s disease. J Mol Neurosci 23, 105-114.

[551] Thompson LA, Bronson JJ, Zusi FC (2005) Progress inthe discovery of BACE inhibitors. Curr Pharm Des 11,3383-3404.

[552] Vassar R (2005) beta-Secretase, APP and Abeta inAlzheimer’s disease. Subcell Biochem 38, 79-103.

[553] Durham TB, Shepherd TA (2006) Progress toward the dis-covery and development of efficacious BACE inhibitors.Curr Opin Drug Discov Devel 9, 776-791.

[554] Guo T, Hobbs DW (2006) Development of BACE1inhibitors for Alzheimer’s disease. Curr Med Chem 13,1811-1829.

[555] John V (2006) Human beta-secretase (BACE) and BACEinhibitors: Progress report. Curr Top Med Chem 6, 569-578.

[556] Schmidt B, Baumann S, Braun HA, Larbig G (2006)Inhibitors and modulators of beta- and gamma-secretase.Curr Top Med Chem 6, 377-392.

[557] Schmidt B, Baumann S, Narlawar R, Braun HA, LarbigG (2006) Modulators and inhibitors of gamma- and beta-secretases. Neurodegener Dis 3, 290-297.

[558] Evin G, Kenche VB (2007) BACE inhibitors as poten-tial therapeutics for Alzheimer’s disease. Recent Pat CNSDrug Discov 2, 188-199.

[559] Ghosh AK, Kumaragurubaran N, Hong L, Kulkarni SS, XuX, Chang W, Weerasena V, Turner R, Koelsch G, BilcerG, Tang J (2007) Design, synthesis, and X-ray structure ofpotent memapsin 2 (beta-secretase) inhibitors with isoph-thalamide derivatives as the P2-P3-ligands. J Med Chem50, 2399-2407.

[560] Ghosh AK, Bilcer G, Hong L, Koelsch G, Tang J (2007)Memapsin 2 (beta-secretase) inhibitor drug, between fan-tasy and reality. Curr Alzheimer Res 4, 418-422.

[561] Ghosh AK, Kumaragurubaran N, Hong L, Kulkarni S, XuX, Miller HB, Reddy DS, Weerasena V, Turner R, ChangW, Koelsch G, Tang J (2008) Potent memapsin 2 (beta-secretase) inhibitors: Design, synthesis, protein-ligand X-ray structure, and in vivo evaluation. Bioorg Med ChemLett 18, 1031-1036.

[562] Ghosh AK, Kumaragurubaran N, Hong L, Koelsh G, TangJ (2008) Memapsin 2 (beta-secretase) inhibitors: Drugdevelopment. Curr Alzheimer Res 5, 121-131.

[563] Ghosh AK, Gemma S, Tang J (2008) beta-Secretase asa therapeutic target for Alzheimer’s disease. Neurothera-peutics 5, 399-408.

Page 69: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 615

[564] Albert JS (2009) Progress in the development of beta-secretase inhibitors for Alzheimer’s disease. Prog MedChem 48, 133-161.

[565] Ghosh AK (2009) Harnessing nature’s insight: Designof aspartyl protease inhibitors from treatment of drug-resistant HIV to Alzheimer’s disease. J Med Chem 52,2163-2176.

[566] Huang WH, Sheng R, Hu YZ (2009) Progress in thedevelopment of nonpeptidomimetic BACE 1 inhibitors forAlzheimer’s disease. Curr Med Chem 16, 1806-1820.

[567] Silvestri R (2009) Boom in the development of non-peptidic beta-secretase (BACE1) inhibitors for thetreatment of Alzheimer’s disease. Med Res Rev 29, 295-338.

[568] Stachel SJ (2009) Progress toward the development of aviable BACE-1 inhibitor. Drug Dev Res 70, 101-110.

[569] Tomita T (2009) Secretase inhibitors and modulators forAlzheimer’s disease treatment. Expert Rev Neurother 9,661-679.

[570] De Strooper B, Vassar R, Golde T (2010) The secretases:Enzymes with therapeutic potential in Alzheimer disease.Nat Rev Neurol 6, 99-107.

[571] Klaver DW, Wilce MC, Cui H, Hung AC, Gasperini R,Foa L, Small DH (2010) Is BACE1 a suitable therapeutictarget for the treatment of Alzheimer’s disease? Currentstrategies and future directions. Biol Chem 391, 849-859.

[572] Luo X, Yan R (2010) Inhibition of BACE1 for therapeuticuse in Alzheimer’s disease. Int J Clin Exp Pathol 3, 618-628.

[573] Marks N, Berg MJ (2010) BACE and gamma-secretasecharacterization and their sorting as therapeutic targets toreduce amyloidogenesis. Neurochem Res 35, 181-210.

[574] Evin G, Barakat A, Masters CL (2010) BACE: Therapeutictarget and potential biomarker for Alzheimer’s disease. IntJ Biochem Cell Biol 42, 1923-1926.

[575] Evin G, Lessene G, Wilkins S (2011) BACE inhibitors aspotential drugs for the treatment of Alzheimer’s disease:Focus on bioactivity. Recent Pat CNS Drug Discov 6, 91-106.

[576] Mancini F, De Simone A, Andrisano V (2011) Beta-secretase as a target for Alzheimer’s disease drugdiscovery: An overview of in vitro methods for character-ization of inhibitors. Anal Bioanal Chem 400, 1979-1996.

[577] Woo HN, Baik SH, Park JS, Gwon AR, Yang S, YunYK, Jo DG (2011) Secretases as therapeutic targets forAlzheimer’s disease. Biochem Biophys Res Commun 404,10-15.

[578] Tresadern G, Delgado F, Delgado O, Gijsen H, Macdon-ald GJ, Moechars D, Rombouts F, Alexander R, SpurlinoJ, Van GM, Vega JA, Trabanco AA (2011) Rational designand synthesis of aminopiperazinones as beta-secretase(BACE) inhibitors. Bioorg Med Chem Lett 21, 7255-7260.

[579] Ghosh AK, Brindisi M, Tang J (2012) Developing beta-secretase inhibitors for treatment of Alzheimer’s disease.J Neurochem 120(Suppl 1), 71-83.

[580] Kandalepas PC, Vassar R (2012) Identification and biologyof beta-secretase. J Neurochem 120(Suppl 1), 55-61.

[581] Zhu Z (2012) Iminoheterocycle as a druggable motif:BACE1 inhibitors and beyond. Trends Pharmacol Sci 33,233-240.

[582] Hu J, Cwi CL, Smiley DL, Timm D, Erickson JA, McGeeJE, Yang HC, Mendel D, May PC, Shapiro M, McCarthyJR (2003) Design and synthesis of statine-containingBACE inhibitors. Bioorg Med Chem Lett 13, 4335-4339.

[583] Chen SH, Lamar J, Guo D, Kohn T, Yang HC, McGee J,Timm D, Erickson J, Yip Y, May P, McCarthy J (2004)P3 cap modified Phe*-Ala series BACE inhibitors. BioorgMed Chem Lett 14, 245-250.

[584] Lamar J, Hu J, Bueno AB, Yang HC, Guo D, Copp JD,McGee J, Gitter B, Timm D, May P, McCarthy J, Chen SH(2004) Phe*-Ala-based pentapeptide mimetics are BACEinhibitors: P2 and P3 SAR. Bioorg Med Chem Lett 14,239-243.

[585] Vidal P, Timm D, Broughton H, Chen SH, Martin JA,Rivera-Sagredo A, McCarthy JR, Shapiro MJ, Espinosa JF(2005) Preorganization of the hydroxyethylene dipeptideisostere: The preferred conformation in solution resem-bles the conformation bound to BACE. J Med Chem 48,7623-7627.

[586] May PC, Dean RA, Lowe SL, Martenyi F, Sheehan SM,Boggs LN, Monk SA, Mathes BM, Mergott DJ, Wat-son BM, Stout SL, Timm DE, Smith LE, Gonzales CR,Nakano M, Jhee SS, Yen M, Ereshefsky L, Lindstrom TD,Calligaro DO, Cocke PJ, Greg HD, Friedrich S, Citron M,Audia JE (2011) Robust central reduction of amyloid-betain humans with an orally available, non-peptidic beta-secretase inhibitor. J Neurosci 31, 16507-16516.

[587] Burt J (2010) American Chemical Society - 240th nationalmeeting - chemistry for preventing and combating disease:Part 1. IDrugs 13, 669-672.

[588] Cumming JN, Le TX, Babu S, Carroll C, Chen X, FavreauL, Gaspari P, Guo T, Hobbs DW, Huang Y, Iserloh U,Kennedy ME, Kuvelkar R, Li G, Lowrie J, McHugh NA,Ozgur L, Pan J, Parker EM, Saionz K, Stamford AW,Strickland C, Tadesse D, Voigt J, Wang L, Wu Y, Zhang L,Zhang Q (2008) Rational design of novel, potent piperazi-none and imidazolidinone BACE1 inhibitors. Bioorg MedChem Lett 18, 3236-3241.

[589] Iserloh U, Wu Y, Cumming JN, Pan J, Wang LY, Stam-ford AW, Kennedy ME, Kuvelkar R, Chen X, ParkerEM, Strickland C, Voigt J (2008) Potent pyrrolidine- andpiperidine-based BACE-1 inhibitors. Bioorg Med ChemLett 18, 414-417.

[590] Iserloh U, Pan J, Stamford AW, Kennedy ME, Zhang Q,Zhang L, Parker EM, McHugh NA, Favreau L, Strick-land C, Voigt J (2008) Discovery of an orally efficaceous4-phenoxypyrrolidine-based BACE-1 inhibitor. BioorgMed Chem Lett 18, 418-422.

[591] Wang YS, Strickland C, Voigt JH, Kennedy ME, BeyerBM, Senior MM, Smith EM, Nechuta TL, Madison VS,Czarniecki M, McKittrick BA, Stamford AW, Parker EM,Hunter JC, Greenlee WJ, Wyss DF (2010) Applicationof fragment-based NMR screening, X-ray crystallogra-phy, structure-based design, and focused chemical librarydesign to identify novel microM leads for the develop-ment of nM BACE-1 (beta-site APP cleaving enzyme 1)inhibitors. J Med Chem 53, 942-950.

[592] Zhu Z, Sun ZY, Ye Y, Voigt J, Strickland C, Smith EM,Cumming J, Wang L, Wong J, Wang YS, Wyss DF, ChenX, Kuvelkar R, Kennedy ME, Favreau L, Parker E, McKit-trick BA, Stamford A, Czarniecki M, Greenlee W, HunterJC (2010) Discovery of cyclic acylguanidines as highlypotent and selective beta-site amyloid cleaving enzyme(BACE) inhibitors: Part I–inhibitor design and validation.J Med Chem 53, 951-965.

[593] Cumming JN, Smith EM, Wang L, Misiaszek J, DurkinJ, Pan J, Iserloh U, Wu Y, Zhu Z, Strickland C, Voigt J,Chen X, Kennedy ME, Kuvelkar R, Hyde LA, Cox K,Favreau L, Czarniecki MF, Greenlee WJ, McKittrick BA,

Page 70: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

616 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

Parker EM, Stamford AW (2012) Structure based designof iminohydantoin BACE1 inhibitors: Identification of anorally available, centrally active BACE1 inhibitor. BioorgMed Chem Lett 22, 2444-2449.

[594] Brady SF, Singh S, Crouthamel MC, Holloway MK,Coburn CA, Garsky VM, Bogusky M, Pennington MW,Vacca JP, Hazuda D, Lai MT (2004) Rational designand synthesis of selective BACE-1 inhibitors. Bioorg MedChem Lett 14, 601-604.

[595] Coburn CA, Stachel SJ, Li YM, Rush DM, Steele TG,Chen-Dodson E, Holloway MK, Xu M, Huang Q, Lai MT,DiMuzio J, Crouthamel MC, Shi XP, Sardana V, ChenZ, Munshi S, Kuo L, Makara GM, Annis DA, TadikondaPK, Nash HM, Vacca JP, Wang T (2004) Identificationof a small molecule nonpeptide active site beta-secretaseinhibitor that displays a nontraditional binding mode foraspartyl proteases. J Med Chem 47, 6117-6119.

[596] Coburn CA, Stachel SJ, Jones KG, Steele TG, Rush DM,DiMuzio J, Pietrak BL, Lai MT, Huang Q, Lineberger J, JinL, Munshi S, Katharine HM, Espeseth A, Simon A, HazudaD, Graham SL, Vacca JP (2006) BACE-1 inhibition by aseries of psi [CH2NH] reduced amide isosteres. BioorgMed Chem Lett 16, 3635-3638.

[597] Stachel SJ, Coburn CA, Steele TG, Jones KG, Loutzen-hiser EF, Gregro AR, Rajapakse HA, Lai MT, CrouthamelMC, Xu M, Tugusheva K, Lineberger JE, Pietrak BL, Espe-seth AS, Shi XP, Chen-Dodson E, Holloway MK, MunshiS, Simon AJ, Kuo L, Vacca JP (2004) Structure-baseddesign of potent and selective cell-permeable inhibitors ofhuman beta-secretase (BACE-1). J Med Chem 47, 6447-6450.

[598] Stachel SJ, Coburn CA, Steele TG, Crouthamel MC,Pietrak BL, Lai MT, Holloway MK, Munshi SK, Gra-ham SL, Vacca JP (2006) Conformationally biased P3amide replacements of beta-secretase inhibitors. BioorgMed Chem Lett 16, 641-644.

[599] Stachel SJ, Coburn CA, Sankaranarayanan S, Price EA,Wu G, Crouthamel M, Pietrak BL, Huang Q, LinebergerJ, Espeseth AS, Jin L, Ellis J, Holloway MK, MunshiS, Allison T, Hazuda D, Simon AJ, Graham SL, VaccaJP (2006) Macrocyclic inhibitors of beta-secretase: Func-tional activity in an animal model. J Med Chem 49, 6147-6150.

[600] Stachel SJ, Coburn CA, Rush D, Jones KL, Zhu H,Rajapakse H, Graham SL, Simon A, Katharine HM, Alli-son TJ, Munshi SK, Espeseth AS, Zuck P, Colussi D,Wolfe A, Pietrak BL, Lai MT, Vacca JP (2009) Discoveryof aminoheterocycles as a novel beta-secretase inhibitorclass: pH dependence on binding activity part 1. BioorgMed Chem Lett 19, 2977-2980.

[601] Espeseth AS, Xu M, Huang Q, Coburn CA, Jones KL,Ferrer M, Zuck PD, Strulovici B, Price EA, Wu G, WolfeAL, Lineberger JE, Sardana M, Tugusheva K, Pietrak BL,Crouthamel MC, Lai MT, Dodson EC, Bazzo R, Shi XP,Simon AJ, Li Y, Hazuda DJ (2005) Compounds that bindAPP and inhibit Abeta processing in vitro suggest a novelapproach to Alzheimer disease therapeutics. J Biol Chem280, 17792-17797.

[602] Rajapakse HA, Nantermet PG, Selnick HG, Munshi S,McGaughey GB, Lindsley SR, Young MB, Lai MT, Espe-seth AS, Shi XP, Colussi D, Pietrak B, Crouthamel MC,Tugusheva K, Huang Q, Xu M, Simon AJ, Kuo L, HazudaDJ, Graham S, Vacca JP (2006) Discovery of oxadiazoyltertiary carbinamine inhibitors of beta-secretase (BACE-1). J Med Chem 49, 7270-7273.

[603] Rajapakse HA, Nantermet PG, Selnick HG, Barrow JC,McGaughey GB, Munshi S, Lindsley SR, Young MB, NgoPL, Holloway MK, Lai MT, Espeseth AS, Shi XP, ColussiD, Pietrak B, Crouthamel MC, Tugusheva K, Huang Q,Xu M, Simon AJ, Kuo L, Hazuda DJ, Graham S, VaccaJP (2010) SAR of tertiary carbinamine derived BACE1inhibitors: Role of aspartate ligand amine pKa in enzymeinhibition. Bioorg Med Chem Lett 20, 1885-1889.

[604] Barrow JC, Rittle KE, Ngo PL, Selnick HG, GrahamSL, Pitzenberger SM, McGaughey GB, Colussi D, LaiMT, Huang Q, Tugusheva K, Espeseth AS, Simon AJ,Munshi SK, Vacca JP (2007) Design and synthesis of2,3,5-substituted imidazolidin-4-one inhibitors of BACE-1. ChemMedChem 2, 995-999.

[605] Hills ID, Vacca JP (2007) Progress toward a practicalBACE-1 inhibitor. Curr Opin Drug Discov Devel 10, 383-391.

[606] Holloway KM, McGaughey GB, Coburn CA, Stachel SJ,Jones KG, Stanton EL, Gregro AR, Lai MT, CrouthamelMC, Pietrak BL, Munshi SK (2007) Evaluating scor-ing functions for docking and designing beta-secretaseinhibitors. Bioorg Med Chem Lett 17, 823-827.

[607] Holloway KM, Hunt P, McGaughey GB (2009) Structureand modeling in the design of �-and �-secretase inhibitors.Drug Dev Res 70, 70-93.

[608] Lindsley SR, Moore KP, Rajapakse HA, Selnick HG,Young MB, Zhu H, Munshi S, Kuo L, McGaugheyGB, Colussi D, Crouthamel MC, Lai MT, Pietrak B,Price EA, Sankaranarayanan S, Simon AJ, Seabrook GR,Hazuda DJ, Pudvah NT, Hochman JH, Graham SL, VaccaJP, Nantermet PG (2007) Design, synthesis, and SARof macrocyclic tertiary carbinamine BACE-1 inhibitors.Bioorg Med Chem Lett 17, 4057-4061.

[609] McGaughey GB, Colussi D, Graham SL, Lai MT, MunshiSK, Nantermet PG, Pietrak B, Rajapakse HA, Selnick HG,Stauffer SR, Holloway MK (2007) Beta-secretase (BACE-1) inhibitors: Accounting for 10s loop flexibility usingrigid active sites. Bioorg Med Chem Lett 17, 1117-1121.

[610] Moore KP, Zhu H, Rajapakse HA, McGaughey GB,Colussi D, Price EA, Sankaranarayanan S, Simon AJ,Pudvah NT, Hochman JH, Allison T, Munshi SK,Graham SL, Vacca JP, Nantermet PG (2007) Strategiestoward improving the brain penetration of macrocyclic ter-tiary carbinamine BACE-1 inhibitors. Bioorg Med ChemLett 17, 5831-5835.

[611] Stauffer SR, Stanton MG, Gregro AR, Steinbeiser MA,Shaffer JR, Nantermet PG, Barrow JC, Rittle KE,Collusi D, Espeseth AS, Lai MT, Pietrak BL, HollowayMK, McGaughey GB, Munshi SK, Hochman JH, SimonAJ, Selnick HG, Graham SL, Vacca JP (2007) Discoveryand SAR of isonicotinamide BACE-1 inhibitors that bindbeta-secretase in a N-terminal 10s-loop down conforma-tion. Bioorg Med Chem Lett 17, 1788-1792.

[612] Sankaranarayanan S, Price EA, Wu G, Crouthamel MC,Shi XP, Tugusheva K, Tyler KX, Kahana J, Ellis J, Jin L,Steele T, Stachel S, Coburn C, Simon AJ (2008) In vivobeta-secretase 1 inhibition leads to brain Abeta loweringand increased alpha-secretase processing of amyloid pre-cursor protein without effect on neuregulin-1. J PharmacolExp Ther 324, 957-969.

[613] Sankaranarayanan S, Holahan MA, Colussi D, CrouthamelMC, Devanarayan V, Ellis J, Espeseth A, Gates AT, Gra-ham SL, Gregro AR, Hazuda D, Hochman JH, HollowayK, Jin L, Kahana J, Lai MT, Lineberger J, McGaughey G,Moore KP, Nantermet P, Pietrak B, Price EA, Rajapakse

Page 71: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 617

H, Stauffer S, Steinbeiser MA, Seabrook G, Selnick HG,Shi XP, Stanton MG, Swestock J, Tugusheva K, TylerKX, Vacca JP, Wong J, Wu G, Xu M, Cook JJ, SimonAJ (2009) First demonstration of cerebrospinal fluid andplasma A beta lowering with oral administration of a beta-site amyloid precursor protein-cleaving enzyme 1 inhibitorin nonhuman primates. J Pharmacol Exp Ther 328, 131-140.

[614] Hills ID, Holloway MK, de LP, Nomland A, Zhu H,Rajapakse H, Allison TJ, Munshi SK, Colussi D, PietrakBL, Toolan D, Haugabook SJ, Graham SL, Stachel SJ(2009) A conformational constraint improves a beta-secretase inhibitor but for an unexpected reason. BioorgMed Chem Lett 19, 4993-4995.

[615] Nantermet PG, Rajapakse HA, Stanton MG, Stauffer SR,Barrow JC, Gregro AR, Moore KP, Steinbeiser MA,Swestock J, Selnick HG, Graham SL, McGaughey GB,Colussi D, Lai MT, Sankaranarayanan S, Simon AJ,Munshi S, Cook JJ, Holahan MA, Michener MS, VaccaJP (2009) Evolution of tertiary carbinamine BACE-1inhibitors: Abeta reduction in rhesus CSF upon oral dos-ing. ChemMedChem 4, 37-40.

[616] Steele TG, Hills ID, Nomland AA, de LP, Allison T,McGaughey G, Colussi D, Tugusheva K, Haugabook SJ,Espeseth AS, Zuck P, Graham SL, Stachel SJ (2009) Iden-tification of a small molecule beta-secretase inhibitor thatbinds without catalytic aspartate engagement. Bioorg MedChem Lett 19, 17-20.

[617] Cumming J, Babu S, Huang Y, Carrol C, Chen X, FavreauL, Greenlee W, Guo T, Kennedy M, Kuvelkar R, Le T, Li G,McHugh N, Orth P, Ozgur L, Parker E, Saionz K, StamfordA, Strickland C, Tadesse D, Voigt J, Zhang L, Zhang Q(2010) Piperazine sulfonamide BACE1 inhibitors: Design,synthesis, and in vivo characterization 4. Bioorg Med ChemLett 20, 2837-2842.

[618] Zhu H, Young MB, Nantermet PG, Graham SL, Colussi D,Lai MT, Pietrak B, Price EA, Sankaranarayanan S, Shi XP,Tugusheva K, Holahan MA, Michener MS, Cook JJ, SimonA, Hazuda DJ, Vacca JP, Rajapakse HA (2010) Rapid P1SAR of brain penetrant tertiary carbinamine derived BACEinhibitors. Bioorg Med Chem Lett 20, 1779-1782.

[619] Wyss DF, Wang YS, Eaton HL, Strickland C, Voigt JH, ZhuZ, Stamford AW (2012) Combining NMR and x-ray crys-tallography in fragment-based drug discovery: Discoveryof highly potent and selective BACE-1 inhibitors. Top CurrChem 317, 83-114.

[620] Stachel SJ, Steele TG, Petrocchi A, Haugabook SJ,McGaughey G, Katharine HM, Allison T, Munshi S,Zuck P, Colussi D, Tugasheva K, Wolfe A, GrahamSL, Vacca JP (2012) Discovery of pyrrolidine-basedbeta-secretase inhibitors: Lead advancement through con-formational design for maintenance of ligand bindingefficiency. Bioorg Med Chem Lett 22, 240-244.

[621] Gruninger-Leitch F, Schlatter D, Kung E, Nelbock P,Dobeli H (2002) Substrate and inhibitor profile of BACE(beta-secretase) and comparison with other mammalianaspartic proteases. J Biol Chem 277, 4687-4693.

[621] [2]Mandal M, Zhu Z, Cumming JN, Liu X, StricklandC, Mazzola RD, Caldwell JP, Leach P, Grzelak M, HydeL, Zhang Q, Terracina G, Zhang L, Chen X, KuvelkarR, Kennedy ME, Favreau L, Cox K, Orth P, Buevich A,Voigt JH, Wang H, Kazakevich I, McKittrick BA, GreenleeW, Parker EM, Stamford AW (2012) Design and vali-dation of bicyclic iminopyrimidinones as beta amyloidcleaving enzyme-1 (BACE1) inhibitors - Conformational

constraint to favor a bioactive conformation. J Med Chem,doi: 10.1021/jm301039c-

[622] Kuglstatter A, Stahl M, Peters JU, Huber W, Stihle M,Schlatter D, Benz J, Ruf A, Roth D, Enderle T, HennigM (2008) Tyramine fragment binding to BACE-1. BioorgMed Chem Lett 18, 1304-1307.

[623] Cheng Y, Judd TC, Bartberger MD, Brown J, Chen K, Fre-meau RT Jr, Hickman D, Hitchcock SA, Jordan B, Li V,Lopez P, Louie SW, Luo Y, Michelsen K, Nixey T, Pow-ers TS, Rattan C, Sickmier EA, St JD Jr, Wahl RC, WenPH, Wood S (2011) From fragment screening to in vivoefficacy: Optimization of a series of 2-aminoquinolinesas potent inhibitors of beta-site amyloid precursor proteincleaving enzyme 1 (BACE1). J Med Chem 54, 5836-5857.

[624] Dineen TA, Weiss MM, Williamson T, Acton P, Babu-Khan S, Bartberger MD, Brown J, Chen K, Cheng Y,Citron M, Croghan MD, Dunn RT, Esmay J, Graceffa RF,Harried SD, Hickman D, Hitchcock SA, Horne DB, HuangH, Imbeah-Ampiah R, Judd T, Kaller MR, Kreiman CR,La DS, Li V, Lopez P, Louie S, Monenschein H, NguyenTT, Pennington LD, San MT, Sickmier EA, Vargas HM,Wahl RC, Wen PH, Whittington DA, Wood S, Xue Q, YangBH, Patel VF, Zhong W (2012) Design and synthesis ofpotent, orally efficacious hydroxyethylamine derived beta-site amyloid precursor protein cleaving enzyme (BACE1)inhibitors. J Med Chem, doi: 10.1021/jm300118s [Epublahead of print].

[625] Weiss MM, Williamson T, Babu-Khan S, Bartberger MD,Brown J, Chen K, Cheng Y, Citron M, Croghan MD,Dineen TA, Esmay J, Graceffa RF, Harried S, HickmanD, Hitchcock SA, Horne DB, Huang H, Imbeah-AmpiahR, Judd T, Kaller MR, Kreiman CR, La DS, Li V, LopezP, Louie S, Monenschein H, Nguyen TT, Pennington LD,Rattan C, San MT, Sickmier EA, Wahl RC, Wen PH, WoodS, Xue Q, Yang BH, Patel VF, Zhong W (2012) Design andpreparation of a potent series of hydroxyethylamine con-taining beta-secretase inhibitors that demonstrate robustreduction of central beta-amyloid. J Med Chem, doi:10.1021/jm300119p [Epubl ahead of print].

[626] Congreve M, Aharony D, Albert J, Callaghan O,Campbell J, Carr RA, Chessari G, Cowan S, EdwardsPD, Frederickson M, McMenamin R, Murray CW, PatelS, Wallis N (2007) Application of fragment screening byX-ray crystallography to the discovery of aminopyridinesas inhibitors of beta-secretase. J Med Chem 50, 1124-1132.

[627] Edwards PD, Albert JS, Sylvester M, Aharony D, AndisikD, Callaghan O, Campbell JB, Carr RA, Chessari G,Congreve M, Frederickson M, Folmer RH, Geschwind-ner S, Koether G, Kolmodin K, Krumrine J, Mauger RC,Murray CW, Olsson LL, Patel S, Spear N, Tian G (2007)Application of fragment-based lead generation to the dis-covery of novel, cyclic amidine beta-secretase inhibitorswith nanomolar potency, cellular activity, and high ligandefficiency. J Med Chem 50, 5912-5925.

[628] Geschwindner S, Olsson LL, Albert JS, Deinum J,Edwards PD, de BT, Folmer RH (2007) Discovery of anovel warhead against beta-secretase through fragment-based lead generation. J Med Chem 50, 5903-5911.

[629] Murray CW, Callaghan O, Chessari G, Cleasby A,Congreve M, Frederickson M, Hartshorn MJ, McMenaminR, Patel S, Wallis N (2007) Application of fragment screen-ing by X-ray crystallography to beta-secretase. J MedChem 50, 1116-1123.

[630] Kornacker MG, Lai Z, Witmer M, Ma J, Hendrick J, LeeVG, Riexinger DJ, Mapelli C, Metzler W, Copeland RA

Page 72: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

618 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

(2005) An inhibitor binding pocket distinct from the cat-alytic active site on human beta-APP cleaving enzyme.Biochemistry 44, 11567-11573.

[631] Iben LG, Kopcho L, Marcinkeviciene J, Zheng C, Thomp-son LA, Albright CF, Toyn JH (2008) [3H]BMS-599240–anovel tritiated ligand for the characterization of BACE1inhibitors. Eur J Pharmacol 593, 10-15.

[632] Wu YJ, Zhang Y, Good AC, Burton CR, Toyn JH, AlbrightCF, Macor JE, Thompson LA (2009) Synthesis andSAR of hydroxyethylamine based phenylcarboxyamidesas inhibitors of BACE. Bioorg Med Chem Lett 19, 2654-2660.

[633] Marcin LR, Higgins MA, Zusi FC, Zhang Y, DeeMF, Parker MF, Muckelbauer JK, Camac DM, MorinPE, Ramamurthy V, Tebben AJ, Lentz KA, Grace JE,Marcinkeviciene JA, Kopcho LM, Burton CR, BartenDM, Toyn JH, Meredith JE, Albright CF, Bronson JJ,Macor JE, Thompson LA (2011) Synthesis and SAR ofindole-and 7-azaindole-1,3-dicarboxamide hydroxyethy-lamine inhibitors of BACE-1. Bioorg Med Chem Lett 21,537-541.

[634] Thompson LA, Shi J, Decicco CP, Tebben AJ, Olson RE,Boy KM, Guernon JM, Good AC, Liauw A, Zheng C,Copeland RA, Combs AP, Trainor GL, Camac DM, Muck-elbauer JK, Lentz KA, Grace JE, Burton CR, Toyn JH,Barten DM, Marcinkeviciene J, Meredith JE, Albright CF,Macor JE (2011) Synthesis and in vivo evaluation of cyclicdiaminopropane BACE-1 inhibitors. Bioorg Med ChemLett 21, 6909-6915.

[635] Boy KM, Guernon JM, Shi J, Toyn JH, MeredithJE, Barten DM, Burton CR, Albright CF, Marcinkevi-ciene J, Good AC, Tebben AJ, Muckelbauer JK, CamacDM, Lentz KA, Bronson JJ, Olson RE, Macor JE,Thompson LA (2011) Monosubstituted gamma-lactamand conformationally constrained 1,3-diaminopropan-2-ol transition-state isostere inhibitors of beta-secretase(BACE). Bioorg Med Chem Lett 21, 6916-6924.

[636] Tung JS, Davis DL, Anderson JP, Walker DE, MamoS, Jewett N, Hom RK, Sinha S, Thorsett ED, John V(2002) Design of substrate-based inhibitors of human beta-secretase. J Med Chem 45, 259-262.

[637] Hom RK, Fang LY, Mamo S, Tung JS, Guinn AC, WalkerDE, Davis DL, Gailunas AF, Thorsett ED, Sinha S, KnopsJE, Jewett NE, Anderson JP, John V (2003) Design andsynthesis of statine-based cell-permeable peptidomimeticinhibitors of human beta-secretase. J Med Chem 46, 1799-1802.

[638] Hom RK, Gailunas AF, Mamo S, Fang LY, Tung JS, WalkerDE, Davis D, Thorsett ED, Jewett NE, Moon JB, JohnV (2004) Design and synthesis of hydroxyethylene-basedpeptidomimetic inhibitors of human beta-secretase. J MedChem 47, 158-164.

[639] Maillard MC, Hom RK, Benson TE, Moon JB, MamoS, Bienkowski M, Tomasselli AG, Woods DD, PrinceDB, Paddock DJ, Emmons TL, Tucker JA, Dappen MS,Brogley L, Thorsett ED, Jewett N, Sinha S, John V(2007) Design, synthesis, and crystal structure of hydrox-yethyl secondary amine-based peptidomimetic inhibitorsof human beta-secretase. J Med Chem 50, 776-781.

[640] Sealy JM, Truong AP, Tso L, Probst GD, Aquino J, HomRK, Jagodzinska BM, Dressen D, Wone DW, Brogley L,John V, Tung JS, Pleiss MA, Tucker JA, Konradi AW, Dap-pen MS, Toth G, Pan H, Ruslim L, Miller J, Bova MP, SinhaS, Quinn KP, Sauer JM (2009) Design and synthesis ofcell potent BACE-1 inhibitors: Structure-activity relation-

ship of P1′ substituents. Bioorg Med Chem Lett 19, 6386-6391.

[641] Probst GD, Bowers S, Sealy JM, Stupi B, Dressen D,Jagodzinska BM, Aquino J, Gailunas A, Truong AP, Tso L,Xu YZ, Hom RK, John V, Tung JS, Pleiss MA, Tucker JA,Konradi AW, Sham HL, Jagodzinski J, Toth G, Brecht E,Yao N, Pan H, Lin M, Artis DR, Ruslim L, Bova MP, SinhaS, Yednock TA, Gauby S, Zmolek W, Quinn KP, Sauer JM(2010) Design and synthesis of hydroxyethylamine (HEA)BACE-1 inhibitors: Structure-activity relationship of thearyl region. Bioorg Med Chem Lett 20, 6034-6039.

[642] Truong AP, Probst GD, Aquino J, Fang L, Brogley L,Sealy JM, Hom RK, Tucker JA, John V, Tung JS, PleissMA, Konradi AW, Sham HL, Dappen MS, Toth G, YaoN, Brecht E, Pan H, Artis DR, Ruslim L, Bova MP,Sinha S, Yednock TA, Zmolek W, Quinn KP, Sauer JM(2010) Improving the permeability of the hydroxyethy-lamine BACE-1 inhibitors: Structure-activity relationshipof P2’ substituents. Bioorg Med Chem Lett 20, 4789-4794.

[643] Truong AP, Toth G, Probst GD, Sealy JM, Bowers S, WoneDW, Dressen D, Hom RK, Konradi AW, Sham HL, Wu J,Peterson BT, Ruslim L, Bova MP, Kholodenko D, Mot-ter RN, Bard F, Santiago P, Ni H, Chian D, Soriano F,Cole T, Brigham EF, Wong K, Zmolek W, Goldbach E,Samant B, Chen L, Zhang H, Nakamura DF, Quinn KP,Yednock TA, Sauer JM (2010) Design of an orally effi-cacious hydroxyethylamine (HEA) BACE-1 inhibitor ina preclinical animal model. Bioorg Med Chem Lett 20,6231-6236.

[644] Tounge BA, Reynolds CH (2003) Calculation of the bind-ing affinity of beta-secretase inhibitors using the linearinteraction energy method. J Med Chem 46, 2074-2082.

[645] Rajamani R, Reynolds CH (2004) Modeling the protona-tion states of the catalytic aspartates in beta-secretase. JMed Chem 47, 5159-5166.

[646] Rajamani R, Reynolds CH (2004) Modeling the bindingaffinities of beta-secretase inhibitors: Application to sub-site specificity. Bioorg Med Chem Lett 14, 4843-4846.

[647] Tounge BA, Rajamani R, Baxter EW, Reitz AB, ReynoldsCH (2006) Linear interaction energy models for beta-secretase (BACE) inhibitors: Role of van der Waals,electrostatic, and continuum-solvation terms. J Mol GraphModel 24, 475-484.

[648] Baxter EW, Conway KA, Kennis L, Bischoff F, MerckenMH, Winter HL, Reynolds CH, Tounge BA, Luo C, ScottMK, Huang Y, Braeken M, Pieters SM, Berthelot DJ,Masure S, Bruinzeel WD, Jordan AD, Parker MH, BoydRE, Qu J, Alexander RS, Brenneman DE, Reitz AB (2007)2-Amino-3,4-dihydroquinazolines as inhibitors of BACE-1 (beta-site APP cleaving enzyme): Use of structure baseddesign to convert a micromolar hit into a nanomolar lead.J Med Chem 50, 4261-4264.

[649] Huang Y, Strobel ED, Ho CY, Reynolds CH, Conway KA,Piesvaux JA, Brenneman DE, Yohrling GJ, Moore AH,Rosenthal D, Alexander RS, Tounge BA, Mercken M, Van-dermeeren M, Parker MH, Reitz AB, Baxter EW (2010)Macrocyclic BACE inhibitors: Optimization of a micro-molar hit to nanomolar leads. Bioorg Med Chem Lett 20,3158-3160.

[650] Shuto D, Kasai S, Kimura T, Liu P, Hidaka K, HamadaT, Shibakawa S, Hayashi Y, Hattori C, Szabo B, IshiuraS, Kiso Y (2003) KMI-008, a novel beta-secretaseinhibitor containing a hydroxymethylcarbonyl isostere as atransition-state mimic: Design and synthesis of substrate-based octapeptides. Bioorg Med Chem Lett 13, 4273-4276.

Page 73: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 619

[651] Kimura T, Shuto D, Kasai S, Liu P, Hidaka K, HamadaT, Hayashi Y, Hattori C, Asai M, Kitazume S, SaidoTC, Ishiura S, Kiso Y (2004) KMI-358 and KMI-370,highly potent and small-sized BACE1 inhibitors con-taining phenylnorstatine. Bioorg Med Chem Lett 14,1527-1531.

[652] Kimura T, Shuto D, Hamada Y, Igawa N, Kasai S, LiuP, Hidaka K, Hamada T, Hayashi Y, Kiso Y (2005)Design and synthesis of highly active Alzheimer’s beta-secretase (BACE1) inhibitors, KMI-420 and KMI-429,with enhanced chemical stability. Bioorg Med Chem Lett15, 211-215.

[653] Kimura T, Hamada Y, Stochaj M, Ikari H, NagamineA, Abdel-Rahman H, Igawa N, Hidaka K, Nguyen JT,Saito K, Hayashi Y, Kiso Y (2006) Design and synthe-sis of potent beta-secretase (BACE1) inhibitors with P1′carboxylic acid bioisosteres. Bioorg Med Chem Lett 16,2380-2386.

[654] Hamada Y, Igawa N, Ikari H, Ziora Z, Nguyen JT, YamaniA, Hidaka K, Kimura T, Saito K, Hayashi Y, Ebina M,Ishiura S, Kiso Y (2006) beta-Secretase inhibitors: Modi-fication at the P4 position and improvement of inhibitoryactivity in cultured cells. Bioorg Med Chem Lett 16, 4354-4359.

[655] Hamada Y, Ohta H, Miyamoto N, Yamaguchi R, YamaniA, Hidaka K, Kimura T, Saito K, Hayashi Y, Ishiura S,Kiso Y (2008) Novel non-peptidic and small-sized BACE1inhibitors. Bioorg Med Chem Lett 18, 1643-1647.

[656] Hamada Y, Abdel-Rahman H, Yamani A, Nguyen JT,Stochaj M, Hidaka K, Kimura T, Hayashi Y, Saito K,Ishiura S, Kiso Y (2008) BACE1 inhibitors: Optimiza-tion by replacing the P1′ residue with non-acidic moiety.Bioorg Med Chem Lett 18, 1649-1653.

[657] Hamada Y, Ohta H, Miyamoto N, Sarma D, Hamada T,Nakanishi T, Yamasaki M, Yamani A, Ishiura S, KisoY (2009) Significance of interactions of BACE1-Arg235with its ligands and design of BACE1 inhibitors with P2pyridine scaffold. Bioorg Med Chem Lett 19, 2435-2439.

[658] Hamada Y, Tagad HD, Nishimura Y, Ishiura S, Kiso Y(2011) Tripeptidic BACE1 inhibitors devised by in-silicoconformational structure-based design. Bioorg Med ChemLett 22, 1130-1135.

[659] Ziora Z, Kimura T, Kiso Y (2006) Small-sized BACE1inhibitors. Drugs Future 31, 53-63.

[660] Ziora Z, Kasai S, Hidaka K, Nagamine A, Kimura T,Hayashi Y, Kiso Y (2007) Design and synthesis ofBACE1 inhibitors containing a novel norstatine derivative(2R,3R)-3-amino-2-hydroxy-4-(phenylthio)butyric acid.Bioorg Med Chem Lett 17, 1629-1633.

[661] Kakizawa T, Hidaka K, Hamada D, Yamaguchi R, UemuraT, Kitamura H, Tagad HD, Hamada T, Ziora Z, HamadaY, Kimura T, Kiso Y (2010) Tetrapeptides, as small-sizedpeptidic inhibitors; synthesis and their inhibitory activityagainst BACE1. J Pept Sci 16, 257-262.

[662] Tagad HD, Hamada Y, Nguyen JT, Hamada T, bdel-Rahman H, Yamani A, Nagamine A, Ikari H, Igawa N,Hidaka K, Sohma Y, Kimura T, Kiso Y (2010) Designof pentapeptidic BACE1 inhibitors with carboxylic acidbioisosteres at P1’ and P4 positions. Bioorg Med Chem18, 3175-3186.

[663] Tagad HD, Hamada Y, Nguyen JT, Hidaka K,Hamada T, Sohma Y, Kimura T, Kiso Y (2011)Structure-guided design and synthesis of P1′ position1-phenylcycloalkylamine-derived pentapeptidic BACE1inhibitors. Bioorg Med Chem 19, 5238-5246.

[664] Ebina M, Futai E, Tanabe C, Sasagawa N, Kiso Y, IshiuraS (2009) Inhibition by KMI-574 leads to dislocaliza-tion of BACE1 from lipid rafts. J Neurosci Res 87, 360-368.

[665] Asai M, Hattori C, Iwata N, Saido TC, Sasagawa N,Szabo B, Hashimoto Y, Maruyama K, Tanuma S, Kiso Y,Ishiura S (2006) The novel beta-secretase inhibitor KMI-429 reduces amyloid beta peptide production in amyloidprecursor protein transgenic and wild-type mice. J Neu-rochem 96, 533-540.

[666] Lu Q, Chen WY, Zhu ZY, Chen J, Xu YC, Kaewpet M,Rukachaisirikul V, Chen LL, Shen X (2012) L655,240, act-ing as a competitive BACE1 inhibitor, efficiently decreasesbeta-amyloid peptide production in HEK293-APPswecells. Acta Pharmacol Sin, doi: 10.1038/aps.2012.74[Epubl ahead of print].

[667] Machauer R, Veenstra S, Rondeau JM, Tintelnot-BlomleyM, Betschart C, Neumann U, Paganetti P (2009) Structure-based design and synthesis of macrocyclic peptidomimeticbeta-secretase (BACE-1) inhibitors. Bioorg Med ChemLett 19, 1361-1365.

[668] Machauer R, Laumen K, Veenstra S, Rondeau JM,Tintelnot-Blomley M, Betschart C, Jaton AL, DesrayaudS, Staufenbiel M, Rabe S, Paganetti P, Neumann U (2009)Macrocyclic peptidomimetic beta-secretase (BACE-1)inhibitors with activity in vivo. Bioorg Med Chem Lett 19,1366-1370.

[669] Lerchner A, Machauer R, Betschart C, Veenstra S, RueegerH, McCarthy C, Tintelnot-Blomley M, Jaton AL, Rabe S,Desrayaud S, Enz A, Staufenbiel M, Paganetti P, RondeauJM, Neumann U (2010) Macrocyclic BACE-1 inhibitorsacutely reduce Abeta in brain after po application. BioorgMed Chem Lett 20, 603-607.

[670] Rueeger H, Rondeau JM, McCarthy C, Mobitz H,Tintelnot-Blomley M, Neumann U, Desrayaud S (2011)Structure based design, synthesis and SAR of cyclichydroxyethylamine (HEA) BACE-1 inhibitors. BioorgMed Chem Lett 21, 1942-1947.

[671] Rueeger H, Lueoend R, Rogel O, Rondeau JM, MobitzH, Machauer R, Jacobson L, Staufenbiel M, DesrayaudS, Neumann U (2012) Discovery of cyclic sulfonehydroxyethylamines as potent and selective beta-site APP-cleaving enzyme 1 (BACE1) Inhibitors: Structure-baseddesign and in vivo reduction of amyloid beta-peptides. JMed Chem 55, 3364-3386.

[672] Hu B, Fan KY, Bridges K, Chopra R, Lovering F, ColeD, Zhou P, Ellingboe J, Jin G, Cowling R, Bard J (2004)Synthesis and SAR of bis-statine based peptides as BACE1 inhibitors. Bioorg Med Chem Lett 14, 3457-3460.

[673] Bridges KG, Chopra R, Lin L, Svenson K, Tam A, JinG, Cowling R, Lovering F, Akopian TN, Blasio-SmithE, Annis-Freeman B, Marvell TH, LaVallie ER, ZollnerRS, Bard J, Somers WS, Stahl ML, Kriz RW (2006) Anovel approach to identifying beta-secretase inhibitors:Bis-statine peptide mimetics discovered using structureand spot synthesis. Peptides 27, 1877-1885.

[674] Cole DC, Manas ES, Stock JR, Condon JS, Jennings LD,Aulabaugh A, Chopra R, Cowling R, Ellingboe JW, FanKY, Harrison BL, Hu Y, Jacobsen S, Jin G, Lin L, Lover-ing FE, Malamas MS, Stahl ML, Strand J, Sukhdeo MN,Svenson K, Turner MJ, Wagner E, Wu J, Zhou P, Bard J(2006) Acylguanidines as small-molecule beta-secretaseinhibitors. J Med Chem 49, 6158-6161.

[675] Cole DC, Stock JR, Chopra R, Cowling R, Ellingboe JW,Fan KY, Harrison BL, Hu Y, Jacobsen S, Jennings LD,

Page 74: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

620 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

Jin G, Lohse PA, Malamas MS, Manas ES, Moore WJ,O’Donnell MM, Olland AM, Robichaud AJ, Svenson K,Wu J, Wagner E, Bard J (2008) Acylguanidine inhibitorsof beta-secretase: Optimization of the pyrrole ring sub-stituents extending into the S1 and S3 substrate bindingpockets. Bioorg Med Chem Lett 18, 1063-1066.

[676] Fobare WF, Solvibile WR, Robichaud AJ, Malamas MS,Manas E, Turner J, Hu Y, Wagner E, Chopra R, Cowling R,Jin G, Bard J (2007) Thiophene substituted acylguanidinesas BACE1 inhibitors. Bioorg Med Chem Lett 17, 5353-5356.

[677] Freskos JN, Fobian YM, Benson TE, Bienkowski MJ,Brown DL, Emmons TL, Heintz R, Laborde A, McDon-ald JJ, Mischke BV, Molyneaux JM, Moon JB, MullinsPB, Bryan PD, Paddock DJ, Tomasselli AG, WinterrowdG (2007) Design of potent inhibitors of human beta-secretase. Part 1. Bioorg Med Chem Lett 17, 73-77.

[678] Freskos JN, Fobian YM, Benson TE, Moon JB,Bienkowski MJ, Brown DL, Emmons TL, Heintz R,Laborde A, McDonald JJ, Mischke BV, Molyneaux JM,Mullins PB, Bryan PD, Paddock DJ, Tomasselli AG,Winterrowd G (2007) Design of potent inhibitors ofhuman beta-secretase. Part 2. Bioorg Med Chem Lett 17,78-81.

[679] Kortum SW, Benson TE, Bienkowski MJ, EmmonsTL, Prince DB, Paddock DJ, Tomasselli AG, MoonJB, Laborde A, TenBrink RE (2007) Potent and selec-tive isophthalamide S2 hydroxyethylamine inhibitors ofBACE1. Bioorg Med Chem Lett 17, 3378-3383.

[680] Jennings LD, Cole DC, Stock JR, Sukhdeo MN, EllingboeJW, Cowling R, Jin G, Manas ES, Fan KY, Malamas MS,Harrison BL, Jacobsen S, Chopra R, Lohse PA, MooreWJ, O’Donnell MM, Hu Y, Robichaud AJ, Turner MJ,Wagner E, Bard J (2008) Acylguanidine inhibitors of beta-secretase: Optimization of the pyrrole ring substituentsextending into the S1′ substrate binding pocket. BioorgMed Chem Lett 18, 767-771.

[681] Malamas MS, Erdei J, Gunawan I, Barnes K, Johnson M,Hui Y, Turner J, Hu Y, Wagner E, Fan K, Olland A, BardJ, Robichaud AJ (2009) Aminoimidazoles as potent andselective human beta-secretase (BACE1) inhibitors. J MedChem 52, 6314-6323.

[682] Malamas MS, Erdei J, Gunawan I, Turner J, Hu Y, WagnerE, Fan K, Chopra R, Olland A, Bard J, Jacobsen S, MagoldaRL, Pangalos M, Robichaud AJ (2010) Design and syn-thesis of 5,5′-disubstituted aminohydantoins as potent andselective human beta-secretase (BACE1) inhibitors. J MedChem 53, 1146-1158.

[683] Malamas MS, Barnes K, Johnson M, Hui Y, Zhou P, TurnerJ, Hu Y, Wagner E, Fan K, Chopra R, Olland A, Bard J, Pan-galos M, Reinhart P, Robichaud AJ (2010) Di-substitutedpyridinyl aminohydantoins as potent and highly selectivehuman beta-secretase (BACE1) inhibitors. Bioorg MedChem 18, 630-639.

[684] Malamas MS, Barnes K, Hui Y, Johnson M, LoveringF, Condon J, Fobare W, Solvibile W, Turner J, Hu Y,Manas ES, Fan K, Olland A, Chopra R, Bard J, Panga-los MN, Reinhart P, Robichaud AJ (2010) Novel pyrrolyl2-aminopyridines as potent and selective human beta-secretase (BACE1) inhibitors. Bioorg Med Chem Lett 20,2068-2073.

[685] Malamas MS, Robichaud A, Erdei J, Quagliato D, Solvi-bile W, Zhou P, Morris K, Turner J, Wagner E, Fan K,Olland A, Jacobsen S, Reinhart P, Riddell D, Pangalos M(2010) Design and synthesis of aminohydantoins as potent

and selective human beta-secretase (BACE1) inhibitorswith enhanced brain permeability. Bioorg Med Chem Lett20, 6597-6605.

[686] Malamas MS, Erdei J, Gunawan I, Barnes K, Hui Y, John-son M, Robichaud A, Zhou P, Yan Y, Solvibile W, TurnerJ, Fan KY, Chopra R, Bard J, Pangalos MN (2011) Newpyrazolyl and thienyl aminohydantoins as potent BACE1inhibitors: Exploring the S2′ region. Bioorg Med ChemLett 21, 5164-5170.

[687] Nowak P, Cole DC, Aulabaugh A, Bard J, Chopra R, Cowl-ing R, Fan KY, Hu B, Jacobsen S, Jani M, Jin G, LoMC, Malamas MS, Manas ES, Narasimhan R, ReinhartP, Robichaud AJ, Stock JR, Subrath J, Svenson K, TurnerJ, Wagner E, Zhou P, Ellingboe JW (2010) Discovery andinitial optimization of 5,5′-disubstituted aminohydantoinsas potent beta-secretase (BACE1) inhibitors. Bioorg MedChem Lett 20, 632-635.

[688] Zhou P, Li Y, Fan Y, Wang Z, Chopra R, Olland A, Hu Y,Magolda RL, Pangalos M, Reinhart PH, Turner MJ, BardJ, Malamas MS, Robichaud AJ (2010) Pyridinyl amino-hydantoins as small molecule BACE1 inhibitors. BioorgMed Chem Lett 20, 2326-2329.

[689] Efremov IV, Vajdos FF, Borzilleri K, Capetta S, Dorff PH,Dutra JK, Mansour M, Chen H, Goldstein SW, Noell S,Oborski CE, O’Connell TN, O’Sullivan TJ, Pandit J, WangH, Wei B, Withka JM, McColl A (2012) Discovery andoptimization of a novel spiro-pyrrolidine inhibitors of beta-secretase (BACE1) through fragment based drug design.J Med Chem, doi: 10.1021/jm201715d [Epubl ahead ofprint].

[690] Monenschein H, Horne DB, Bartberger MD, HitchcockSA, Nguyen TT, Patel VF, Pennington LD, Zhong W(2012) Structure guided P1′ modifications of HEA derivedbeta-secretase inhibitors for the treatment of Alzheimer’sdisease. Bioorg Med Chem Lett 22, 3607-3611.

[691] Godemann R, Madden J, Kramer J, Smith M, Fritz U,Hesterkamp T, Barker J, Hoppner S, Hallett D, Cesura A,Ebneth A, Kemp J (2009) Fragment-based discovery ofBACE1 inhibitors using functional assays. Biochemistry48, 10743-10751.

[692] Madden J, Dod JR, Godemann R, Kraemer J, SmithM, Biniszkiewicz M, Hallett DJ, Barker J, Dyekjaer JD,Hesterkamp T (2010) Fragment-based discovery and opti-mization of BACE1 inhibitors. Bioorg Med Chem Lett 20,5329-5333.

[693] Park H, Min K, Kwak HS, Koo KD, Lim D, Seo SW, ChoiJU, Platt B, Choi DY (2008) Synthesis, SAR, and X-raystructure of human BACE-1 inhibitors with cyclic ureaderivatives. Bioorg Med Chem Lett 18, 2900-2904.

[694] Manzenrieder F, Frank AO, Huber T, Dorner-Ciossek C,Kessler H (2007) Synthesis and biological evaluation ofphosphino dipeptide isostere inhibitor of human beta-secretase (BACE1). Bioorg Med Chem 15, 4136-4143.

[695] Huber T, Manzenrieder F, Kuttruff CA, Dorner-CiossekC, Kessler H (2009) Prolonged stability by cyclization:Macrocyclic phosphino dipeptide isostere inhibitors ofbeta-secretase (BACE1). Bioorg Med Chem Lett 19, 4427-4431.

[696] Huang W, Lv D, Yu H, Sheng R, Kim SC, Wu P, Luo K,Li J, Hu Y (2010) Dual-target-directed 1,3-diphenylureaderivatives: BACE 1 inhibitor and metal chelator againstAlzheimer’s disease. Bioorg Med Chem 18, 5610-5615.

[697] Rajendran L, Schneider A, Schlechtingen G, Weidlich S,Ries J, Braxmeier T, Schwille P, Schulz JB, SchroederC, Simons M, Jennings G, Knolker HJ, Simons K (2008)

Page 75: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 621

Efficient inhibition of the Alzheimer’s disease beta-secretase by membrane targeting. Science 320, 520-523.

[698] Arbel M, Yacoby I, Solomon B (2005) Inhibition ofamyloid precursor protein processing by beta-secretasethrough site-directed antibodies. Proc Natl Acad Sci U S A102, 7718-7723.

[699] Boddapati S, Levites Y, Sierks MR (2011) Inhibiting beta-secretase activity in Alzheimer’s disease cell models withsingle-chain antibodies specifically targeting APP. J MolBiol 405, 436-447.

[700] Atwal JK, Chen Y, Chiu C, Mortensen DL, Meilandt WJ,Liu Y, Heise CE, Hoyte K, Luk W, Lu Y, Peng K, Wu P,Rouge L, Zhang Y, Lazarus RA, Scearce-Levie K, WangW, Wu Y, Tessier-Lavigne M, Watts RJ (2011) A ther-apeutic antibody targeting BACE1 inhibits amyloid-betaproduction in vivo. Sci Transl Med 3, 84ra43.

[701] Yu YJ, Zhang Y, Kenrick M, Hoyte K, Luk W, Lu Y, AtwalJ, Elliott JM, Prabhu S, Watts RJ, Dennis MS (2011) Boost-ing brain uptake of a therapeutic antibody by reducing itsaffinity for a transcytosis target. Sci Transl Med 3, 84ra44.

[702] Boado RJ, Zhang Y, Wang Y, Pardridge WM (2009) Engi-neering and expression of a chimeric transferrin receptormonoclonal antibody for blood-brain barrier delivery inthe mouse. Biotechnol Bioeng 102, 1251-1258.

[703] Boado RJ, Zhou QH, Lu JZ, Hui EK, Pardridge WM (2010)Pharmacokinetics and brain uptake of a genetically engi-neered bifunctional fusion antibody targeting the mousetransferrin receptor. Mol Pharm 7, 237-244.

[704] Zhou QH, Fu A, Boado RJ, Hui EK, Lu JZ, PardridgeWM (2011) Receptor-mediated abeta amyloid antibodytargeting to Alzheimer’s disease mouse brain. Mol Pharm8, 280-285.

[705] Zhou QH, Boado RJ, Hui EK, Lu JZ, Pardridge WM (2011)Chronic dosing of mice with a transferrin receptor mon-oclonal antibody-glial-derived neurotrophic factor fusionprotein. Drug Metab Dispos 39, 1149-1154.

[706] Zhou QH, Boado RJ, Pardridge WM (2012) Selectiveplasma pharmacokinetics and brain uptake in the mouseof enzyme fusion proteins derived from species-specificreceptor-targeted antibodies. J Drug Target 20, 715-719.

[707] Pardridge WM, Boado RJ (2012) Reengineering biophar-maceuticals for targeted delivery across the blood-brainbarrier. Methods Enzymol 503, 269-292.

[708] Boado RJ, Pardridge WM (2011) The Trojan Horse Lipo-some Technology for Nonviral Gene Transfer across theBlood-Brain Barrier. J Drug Deliv 2011, 296151.

[709] Jiaranaikulwanitch J, Boonyarat C, Fokin VV, VajraguptaO (2010) Triazolyl tryptoline derivatives as beta-secretaseinhibitors. Bioorg Med Chem Lett 20, 6572-6576.

[710] Jiaranaikulwanitch J, Govitrapong P, Fokin VV, VajraguptaO (2012) From BACE1 inhibitor to multifunctional-ity of tryptoline and tryptamine triazole derivatives forAlzheimer’s disease. Molecules 17, 8312-8333.

[711] Larbig G, Schmidt B (2006) Synthesis of tetramic andtetronic acids as beta-secretase inhibitors. J Comb Chem8, 480-490.

[712] Linning P, Haussmann U, Beyer I, Weidlich S, Schieb H,Wiltfang J, Klafki HW, Knolker HJ (2012) Optimisationof BACE1 inhibition of tripartite structures by modifica-tion of membrane anchors, spacers and pharmacophores- development of potential agents for the treatmentof Alzheimer’s disease. Org Biomol Chem 10, 8216-8235.

[713] Schieb H, Weidlich S, Schlechtingen G, Linning P, Jen-nings G, Gruner M, Wiltfang J, Klafki HW, Knolker

HJ (2010) Structural design, solid-phase synthesis andactivity of membrane-anchored beta-secretase inhibitorson Abeta generation from wild-type and Swedish-mutantAPP. Chemistry 16, 14412-14423.

[714] Choi SJ, Cho JH, Im I, Lee SD, Jang JY, Oh YM, JungYK, Jeon ES, Kim YC (2010) Design and synthesis of1,4-dihydropyridine derivatives as BACE-1 inhibitors. EurJ Med Chem 45, 2578-2590.

[715] Back M, Nyhlen J, Kvarnstrom I, Appelgren S, BorkakotiN, Jansson K, Lindberg J, Nystrom S, Hallberg A, Rosen-quist S, Samuelsson B (2008) Design, synthesis and SARof potent statine-based BACE-1 inhibitors: Exploration ofP1 phenoxy and benzyloxy residues. Bioorg Med Chem16, 9471-9486.

[716] Patey SJ, Edwards EA, Yates EA, Turnbull JE (2006) Hep-arin derivatives as inhibitors of BACE-1, the Alzheimer’sbeta-secretase, with reduced activity against factor Xa andother proteases. J Med Chem 49, 6129-6132.

[717] Patey SJ, Edwards EA, Yates EA, Turnbull JE (2008)Engineered heparins: Novel beta-secretase inhibitors aspotential Alzheimer’s disease therapeutics. NeurodegenerDis 5, 197-199.

[718] Laras Y, Garino C, Dessolin J, Weck C, Moret V, Rol-land A, Kraus JL (2009) New N(4)-substituted piperazinenaphthamide derivatives as BACE-1 inhibitors. J EnzymeInhib Med Chem 24, 181-187.

[719] Barazza A, Gotz M, Cadamuro SA, Goettig P, Willem M,Steuber H, Kohler T, Jestel A, Reinemer P, Renner C, BodeW, Moroder L (2007) Macrocyclic statine-based inhibitorsof BACE-1. Chembiochem 8, 2078-2091.

[720] Hanessian S, Yun H, Hou Y, Yang G, BayrakdarianM, Therrien E, Moitessier N, Roggo S, Veenstra S,Tintelnot-Blomley M, Rondeau JM, Ostermeier C, StraussA, Ramage P, Paganetti P, Neumann U, Betschart C(2005) Structure-based design, synthesis, and memapsin 2(BACE) inhibitory activity of carbocyclic and heterocyclicpeptidomimetics. J Med Chem 48, 5175-5190.

[721] Hanessian S, Yang G, Rondeau JM, Neumann U,Betschart C, Tintelnot-Blomley M (2006) Structure-baseddesign and synthesis of macroheterocyclic peptidomimeticinhibitors of the aspartic protease beta-site amyloid pre-cursor protein cleaving enzyme (BACE). J Med Chem 49,4544-4567.

[722] Hanessian S, Shao Z, Betschart C, Rondeau JM, NeumannU, Tintelnot-Blomley M (2010) Structure-based designand synthesis of novel P2/P3 modified, non-peptidic beta-secretase (BACE-1) inhibitors. Bioorg Med Chem Lett 20,1924-1927.

[723] Niu Y, Wang Y, Zou X, Yang X, Ma C, Lu Y, Zhou B, YuanY, Du G, Xu P (2010) Synthesis and preliminary evaluationof peptidomimetic inhibitors of human beta-secretase. EurJ Med Chem 45, 2089-2094.

[724] Asso V, Ghilardi E, Bertini S, Digiacomo M, Granchi C,Minutolo F, Rapposelli S, Bortolato A, Moro S, MacchiaM (2008) alpha-Naphthylaminopropan-2-ol Derivatives asBACE1 Inhibitors. ChemMedChem 3, 1530-1534.

[725] Bertini S, Ghilardi E, Asso V, Granchi C, Minutolo F,Pineschi M, Di B, V, Bortolato A, Moro S, Saba A, MacchiaM (2010) BACE1 inhibitory activities of enantiomericallypure, variously substituted N-(3-(4-benzhydrylpiperazin-1-yl)-2-hydroxypropyl) arylsulfonamides. Bioorg MedChem 18, 7991-7996.

[726] Bertini S, Asso V, Ghilardi E, Granchi C, Manera C, Min-utolo F, Saccomanni G, Bortolato A, Mason J, Moro S,Macchia M (2011) Carbazole-containing arylcarboxam-

Page 76: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

622 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

ides as BACE1 inhibitors. Bioorg Med Chem Lett 21,6657-6661.

[727] Ghosh AK, Devasamudram T, Hong L, DeZutter C, XuX, Weerasena V, Koelsch G, Bilcer G, Tang J (2005)Structure-based design of cycloamide-urethane-derivednovel inhibitors of human brain memapsin 2 (beta-secretase). Bioorg Med Chem Lett 15, 15-20.

[728] Ghosh AK, Kumaragurubaran N, Hong L, Lei H, Hus-sain KA, Liu CF, Devasamudram T, Weerasena V, TurnerR, Koelsch G, Bilcer G, Tang J (2006) Design, synthesisand X-ray structure of protein-ligand complexes: Impor-tant insight into selectivity of memapsin 2 (beta-secretase)inhibitors. J Am Chem Soc 128, 5310-5311.

[729] La Regina G, Piscitelli F, Silvestri R (2009) Syntheticstrategies of nonpeptidic b-secretase (BACE1) inhibitors.J Heterocyclic Chem 46, 10-17.

[730] Xiao K, Li X, Li J, Ma L, Hu B, Yu H, Fu Y, Wang R, MaZ, Qiu B, Li J, Hu D, Wang X, Shen J (2006) Design, syn-thesis, and evaluation of Leu*Ala hydroxyethylene-basednon-peptide beta-secretase (BACE) inhibitors. Bioorg MedChem 14, 4535-4551.

[731] Zhu YP, Xiao K, Yu HP, Ma LP, Xiong B, Zhang HY,Wang X, Li JY, Li J, Shen JK (2009) Discovery ofpotent beta-secretase (bace-1) inhibitors by the synthe-sis of isophthalamide-containing hybrids. Acta PharmacolSin 30, 259-269.

[732] Sasaki H, Miki K, Kinoshita K, Koyama K, Juliawaty LD,Achmad SA, Hakim EH, Kaneda M, Takahashi K (2010)beta-Secretase (BACE-1) inhibitory effect of biflavonoids.Bioorg Med Chem Lett 20, 4558-4560.

[733] Bjorklund C, Adolfsson H, Jansson K, Lindberg J, VrangL, Hallberg A, Rosenquist S, Samuelsson B (2010) Dis-covery of potent BACE-1 inhibitors containing a newhydroxyethylene (HE) scaffold: Exploration of P1′ alkoxyresidues and an aminoethylene (AE) central core. BioorgMed Chem 18, 1711-1723.

[734] Bjorklund C, Oscarson S, Benkestock K, Borkakoti N,Jansson K, Lindberg J, Vrang L, Hallberg A, Rosenquist A,Samuelsson B (2010) Design and synthesis of potent andselective BACE-1 inhibitors. J Med Chem 53, 1458-1464.

[735] Meredith JA, Bjorklund C, Adolfsson H, Jansson K, Hall-berg A, Rosenquist A, Samuelsson B (2010) P2′-truncatedBACE-1 inhibitors with a novel hydroxethylene-like core.Eur J Med Chem 45, 542-554.

[736] Kumar AB, Anderson JM, Melendez AL, Manetsch R(2012) Synthesis and structure-activity relationship stud-ies of 1,3-disubstituted 2-propanols as BACE-1 inhibitors.Bioorg Med Chem Lett 22, 4740-4744.

[737] Chiriano G, De SA, Mancini F, Perez DI, Cavalli A,Bolognesi ML, Legname G, Martinez A, Andrisano V,Carloni P, Roberti M (2012) A small chemical libraryof 2-aminoimidazole derivatives as BACE-1 inhibitors:Structure-based design, synthesis, and biological evalu-ation. Eur J Med Chem 48, 206-213.

[738] Andrau D, Dumanchin-Njock C, Ayral E, Vizzavona J,Farzan M, Boisbrun M, Fulcrand P, Hernandez JF, Mar-tinez J, Lefranc-Jullien S, Checler F (2003) BACE1- andBACE2-expressing human cells: Characterization of beta-amyloid precursor protein-derived catabolites, design of anovel fluorimetric assay, and identification of new in vitroinhibitors. J Biol Chem 278, 25859-25866.

[739] Lefranc-Jullien S, Lisowski V, Hernandez JF, Martinez J,Checler F (2005) Design and characterization of a newcell-permeant inhibitor of the beta-secretase BACE1. Br JPharmacol 145, 228-235.

[740] Russo F, Wangsell F, Savmarker J, Jacobsson M, Larhed M(2009) Synthesis and evaluation of a new class of tertiaryalcohol based BACE-1 inhibitors. Tetrahedron 65, 10047-10059.

[741] Wangsell F, Russo F, Savmarker J, Rosenquist A, Samuels-son B, Larhed M (2009) Design and synthesis of BACE-1inhibitors utilizing a tertiary hydroxyl motif as the transi-tion state mimic. Bioorg Med Chem Lett 19, 4711-4714.

[742] Wangsell F, Gustafsson K, Kvarnstrom I, Borkakoti N,Edlund M, Jansson K, Lindberg J, Hallberg A, Rosen-quist A, Samuelsson B (2010) Synthesis of potent BACE-1inhibitors incorporating a hydroxyethylene isostere as cen-tral core. Eur J Med Chem 45, 870-882.

[743] Wangsell F, Nordeman P, Savmarker J, Emanuelsson R,Jansson K, Lindberg J, Rosenquist S, Samuelsson B,Larhed M (2011) Investigation of alpha-phenylnorstatineand alpha-benzylnorstatine as transition state isosteremotifs in the search for new BACE-1 inhibitors. BioorgMed Chem 19, 145-155.

[744] Marcinkeviciene J, Luo Y, Graciani NR, Combs AP,Copeland RA (2001) Mechanism of inhibition of beta-siteamyloid precursor protein-cleaving enzyme (BACE) by astatine-based peptide. J Biol Chem 276, 23790-23794.

[745] Xiong B, Huang XQ, Shen LL, Shen JH, Luo XM, Shen X,Jiang HL, Chen KX (2004) Conformational flexibility ofbeta-secretase: Molecular dynamics simulation and essen-tial dynamics analysis. Acta Pharmacol Sin 25, 705-713.

[746] Huang D, Luthi U, Kolb P, Edler K, Cecchini M, AudetatS, Barberis A, Caflisch A (2005) Discovery of cell-permeable non-peptide inhibitors of beta-secretase byhigh-throughput docking and continuum electrostatics cal-culations. J Med Chem 48, 5108-5111.

[747] Polgar T, Keseru GM (2005) Virtual screening for beta-secretase (BACE1) inhibitors reveals the importance ofprotonation states at Asp32 and Asp228. J Med Chem 48,3749-3755.

[748] Huang D, Luthi U, Kolb P, Cecchini M, Barberis A,Caflisch A (2006) In silico discovery of beta-secretaseinhibitors. J Am Chem Soc 128, 5436-5443.

[749] Moitessier N, Therrien E, Hanessian S (2006) A methodfor induced-fit docking, scoring, and ranking of flexibleligands. Application to peptidic and pseudopeptidic beta-secretase (BACE 1) inhibitors. J Med Chem 49, 5885-5894.

[750] Polgar T, Keseru GM (2006) Ensemble docking into flexi-ble active sites. Critical evaluation of FlexE against JNK-3and beta-secretase. J Chem Inf Model 46, 1795-1805.

[751] Yu N, Hayik SA, Wang B, Liao N, Reynolds CH, MerzKM Jr (2006) Assigning the protonation states of the keyaspartates in beta-Secretase using QM/MM X-ray struc-ture refinement. J Chem Theory Comput 2, 1057-1069.

[752] English NJ (2007) Calculation of binding affinities ofHIV-1 RT and beta-secretase inhibitors using the linearinteraction energy method with explicit and continuumsolvation approaches. J Mol Model 13, 1081-1097.

[753] Limongelli V, Marinelli L, Cosconati S, Braun HA,Schmidt B, Novellino E (2007) Ensemble-dockingapproach on BACE-1: Pharmacophore perception andguidelines for drug design. ChemMedChem 2, 667-678.

[754] Villaverde MC, Gonzalez-Louro L, Sussman F (2007) Thesearch for drug leads targeted to the beta-secretase: Anexample of the roles of computer assisted approaches indrug discovery. Curr Top Med Chem 7, 980-990.

[755] Fujimoto T, Matsushita Y, Gouda H, Yamaotsu N, Hirono S(2008) In silico multi-filter screening approaches for devel-

Page 77: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 623

oping novel beta-secretase inhibitors. Bioorg Med ChemLett 18, 2771-2775.

[756] Huang W, Yu H, Sheng R, Li J, Hu Y (2008)Identification of pharmacophore model, synthesis andbiological evaluation of N-phenyl-1-arylamide andN-phenylbenzenesulfonamide derivatives as BACE 1inhibitors. Bioorg Med Chem 16, 10190-10197.

[757] Chirapu SR, Pachaiyappan B, Nural HF, Cheng X, YuanH, Lankin DC, Abdul-Hay SO, Thatcher GR, Shen Y,Kozikowski AP, Petukhov PA (2009) Molecular model-ing, synthesis, and activity studies of novel biaryl andfused-ring BACE1 inhibitors. Bioorg Med Chem Lett 19,264-274.

[758] Rizzi L, Vaiana N, Sagui F, Genesio E, Pilli E, PorcariV, Romeo S (2009) Design, synthesis and docking studiesof Hydroxyethylamine and Hydroxyethylsulfide BACE-1inhibitors. Protein Pept Lett 16, 86-90.

[759] Al-Nadaf A, Abu SG, Taha MO (2010) Elaborate ligand-based pharmacophore exploration and QSAR analysisguide the synthesis of novel pyridinium-based potentbeta-secretase inhibitory leads. Bioorg Med Chem 18,3088-3115.

[760] Gutierrez LJ, Enriz RD, Baldoni HA (2010) Structuraland thermodynamic characteristics of the exosite bind-ing pocket on the human BACE1: A molecular modelingapproach. J Phys Chem A 114, 10261-10269.

[761] Liu S, Zhou LH, Wang HQ, Yao ZB (2010) Super-imposing the 27 crystal protein/inhibitor complexes ofbeta-secretase to calculate the binding affinities by the lin-ear interaction energy method. Bioorg Med Chem Lett 20,6533-6537.

[762] Pandey A, Mungalpara J, Mohan CG (2010) Compara-tive molecular field analysis and comparative molecularsimilarity indices analysis of hydroxyethylamine deriva-tives as selective human BACE-1 inhibitor. Mol Divers 14,39-49.

[763] Salum LB, Valadares NF (2010) Fragment-guidedapproach to incorporating structural information into aCoMFA study: BACE-1 as an example. J Comput AidedMol Des 24, 803-817.

[764] Xu W, Chen G, Zhu W, Zuo Z (2010) Molecular dockingand structure-activity relationship studies on benzothia-zole based non-peptidic BACE-1 inhibitors. Bioorg MedChem Lett 20, 6203-6207.

[765] Xu W, Chen G, Zhu W, Zuo Z (2010) Identification ofa sub-micromolar, non-peptide inhibitor of beta-secretasewith low neural cytotoxicity through in silico screening 7.Bioorg Med Chem Lett 20, 5763-5766.

[766] Gueto-Tettay C, Drosos JC, Vivas-Reyes R (2011) Quan-tum mechanics study of the hydroxyethylamines-BACE-1active site interaction energies. J Comput Aided Mol Des25, 583-597.

[767] John S, Thangapandian S, Sakkiah S, Lee KW (2011)Potent BACE-1 inhibitor design using pharmacophoremodeling, in silico screening and molecular docking stud-ies. BMC Bioinformatics 12 (Suppl 1), S28.

[768] Sussman F, Otero JM, Villaverde MC, Castro M,Dominguez JL, Gonzalez-Louro L, Estevez RJ, Estevez JC(2011) On a possible neutral charge state for the catalyticdyad in beta-secretase when bound to hydroxyethylenetransition state analogue inhibitors. J Med Chem 54, 3081-3085.

[769] Nino H, Garcia-Pintos I, Rodriguez-Borges JE, Escobar-Cubiella M, Garcia-Mera X, Prado-Prado F (2011) Reviewof synthesis, biological assay and QSAR studies of beta-

secretase inhibitors. Curr Comput Aided Drug Des 7, 263-275.

[770] Al-Tel TH, Semreen MH, Al-Qawasmeh RA, Schmidt MF,El-Awadi R, Ardah M, Zaarour R, Rao SN, El-AgnafO (2011) Design, synthesis, and qualitative structure-activity evaluations of novel beta-secretase inhibitors aspotential Alzheimer’s drug leads. J Med Chem 54, 8373-8385.

[771] Chiriano G, Sartini A, Mancini F, Andrisano V, BolognesiML, Roberti M, Recanatini M, Carloni P, Cavalli A (2011)Sequential virtual screening approach to the identificationof small organic molecules as potential BACE-1 inhibitors.Chem Biol Drug Des 77, 268-271.

[772] Liu S, Fu R, Zhou LH, Chen SP (2012) Application ofconsensus scoring and principal component analysis forvirtual screening against beta-secretase (BACE-1). PLoSOne 7, e38086.

[773] Razzaghi-Asl N, Ebadi A, Edraki N, Mehdipour A, Sha-habipour S, Miri R (2012) Response surface methodologyin docking study of small molecule BACE-1 inhibitors.J Mol Model, doi: 10.1007/S00894-012-1424-1 [Epublahead of print].

[774] Barman A, Prabhakar R (2012) Protonation states of thecatalytic dyad of beta-secretase (BACE1) in the presenceof chemically diverse inhibitors: A molecular dockingstudy. J Chem Inf Model 52, 1275-1287.

[775] Niu Y, Ma C, Jin H, Xu F, Gao H, Liu P, Li Y, Wang C,Yang G, Xu P (2012) The discovery of novel beta-secretaseinhibitors: Pharmacophore modeling, virtual screening,and docking studies. Chem Biol Drug Des 79, 972-980.

[776] Nino H, Rodriguez-Borges JE, Garcia-Mera X, Prado-Prado F (2012) Review of synthesis, assay, and predictionof beta and gamma-secretase inhibitors. Curr Top MedChem 12, 828-844.

[777] Xu Y, Li MJ, Greenblatt H, Chen W, Paz A, Dym O, PelegY, Chen T, Shen X, He J, Jiang H, Silman I, Sussman JL(2012) Flexibility of the flap in the active site of BACE1as revealed by crystal structures and molecular dynamicssimulations. Acta Crystallogr D Biol Crystallogr 68, 13-25.

[778] Liu S, Fu R, Cheng X, Chen SP, Zhou LH (2012) Explor-ing the binding of BACE-1 inhibitors using comparativebinding energy analysis (COMBINE). BMC Struct Biol12, 21.

[779] Back M, Nyhlen J, Kvarnstrom I, Appelgren S, BorkakotiN, Jansson K, Lindberg J, Nystrom S, Hallberg A, Rosen-quist S, Samuelsson B (2008) Design, synthesis and SARof potent statine-based BACE-1 inhibitors: Exploration ofP1 phenoxy and benzyloxy residues. Bioorg Med Chem16, 9471-9486.

[780] Swahn BM, Holenz J, Kihlstrom J, Kolmodin K, Lind-strom J, Plobeck N, Rotticci D, Sehgelmeble F, SundstromM, Berg S, Falting J, Georgievska B, Gustavsson S, Neelis-sen J, Ek M, Olsson LL, Berg S (2012) Aminoimidazolesas BACE-1 inhibitors: The challenge to achieve in vivobrain efficacy. Bioorg Med Chem Lett 22, 1854-1859.

[781] Swahn BM, Kolmodin K, Karlstrom S, von BS, SodermanP, Holenz J, Berg S, Lindstrom J, Sundstrom M, Turek D,Kihlstrom J, Slivo C, Andersson L, Pyring D, Rotticci D,Ohberg L, Kers A, Bogar K, Bergh M, Olsson LL, JansonJ, Eketjall S, Georgievska B, Jeppsson F, Falting J (2012)Design and synthesis of beta-secretase (BACE1) inhibitorswith in vivo brain reduction of beta-amyloid peptides.J Med Chem, doi: 10.1021/jm3009025 [Epubl ahead ofprint].

Page 78: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

624 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

[782] Chang WP, Huang X, Downs D, Cirrito JR, Koelsch G,Holtzman DM, Ghosh AK, Tang J (2011) Beta-secretaseinhibitor GRL-8234 rescues age-related cognitive declinein APP transgenic mice. FASEB J 25, 775-784.

[783] Charrier N, Clarke B, Cutler L, Demont E, Dingwall C,Dunsdon R, East P, Hawkins J, Howes C, Hussain I, Jef-frey P, Maile G, Matico R, Mosley J, Naylor A, O’BrienA, Redshaw S, Rowland P, Soleil V, Smith KJ, SweitzerS, Theobald P, Vesey D, Walter DS, Wayne G (2008) Sec-ond generation of hydroxyethylamine BACE-1 inhibitors:Optimizing potency and oral bioavailability. J Med Chem51, 3313-3317.

[784] Charrier N, Clarke B, Cutler L, Demont E, Dingwall C,Dunsdon R, Hawkins J, Howes C, Hubbard J, HussainI, Maile G, Matico R, Mosley J, Naylor A, O’Brien A,Redshaw S, Rowland P, Soleil V, Smith KJ, Sweitzer S,Theobald P, Vesey D, Walter DS, Wayne G (2009) Sec-ond generation of BACE-1 inhibitors. Part 1: The need forimproved pharmacokinetics. Bioorg Med Chem Lett 19,3664-3668.

[785] Charrier N, Clarke B, Demont E, Dingwall C, DunsdonR, Hawkins J, Hubbard J, Hussain I, Maile G, MaticoR, Mosley J, Naylor A, O’Brien A, Redshaw S, RowlandP, Soleil V, Smith KJ, Sweitzer S, Theobald P, Vesey D,Walter DS, Wayne G (2009) Second generation of BACE-1 inhibitors part 2: Optimisation of the non-prime sidesubstituent. Bioorg Med Chem Lett 19, 3669-3673.

[786] Charrier N, Clarke B, Cutler L, Demont E, Dingwall C,Dunsdon R, Hawkins J, Howes C, Hubbard J, HussainI, Maile G, Matico R, Mosley J, Naylor A, O’Brien A,Redshaw S, Rowland P, Soleil V, Smith KJ, Sweitzer S,Theobald P, Vesey D, Walter DS, Wayne G (2009) Sec-ond generation of BACE-1 inhibitors part 3: Towards nonhydroxyethylamine transition state mimetics. Bioorg MedChem Lett 19, 3674-3678.

[787] Hussain I, Hawkins J, Harrison D, Hille C, Wayne G, Cut-ler L, Buck T, Walter D, Demont E, Howes C, Naylor A,Jeffrey P, Gonzalez MI, Dingwall C, Michel A, RedshawS, Davis JB (2007) Oral administration of a potent andselective non-peptidic BACE-1 inhibitor decreases beta-cleavage of amyloid precursor protein and amyloid-betaproduction in vivo. J Neurochem 100, 802-809.

[788] Beswick P, Charrier N, Clarke B, Demont E, Dingwall C,Dunsdon R, Faller A, Gleave R, Hawkins J, Hussain I,Johnson CN, MacPherson D, Maile G, Matico R, MilnerP, Mosley J, Naylor A, O’Brien A, Redshaw S, RiddellD, Rowland P, Skidmore J, Soleil V, Smith KJ, StanwayS, Stemp G, Stuart A, Sweitzer S, Theobald P, Vesey D,Walter DS, Ward J, Wayne G (2008) BACE-1 inhibitorspart 3: Identification of hydroxy ethylamines (HEAs) withnanomolar potency in cells. Bioorg Med Chem Lett 18,1022-1026.

[789] Clarke B, Demont E, Dingwall C, Dunsdon R, Faller A,Hawkins J, Hussain I, MacPherson D, Maile G, Matico R,Milner P, Mosley J, Naylor A, O’Brien A, Redshaw S, Rid-dell D, Rowland P, Soleil V, Smith KJ, Stanway S, StempG, Sweitzer S, Theobald P, Vesey D, Walter DS, Ward J,Wayne G (2008) BACE-1 inhibitors part 1: Identificationof novel hydroxy ethylamines (HEAs). Bioorg Med ChemLett 18, 1011-1016.

[790] Clarke B, Demont E, Dingwall C, Dunsdon R, Faller A,Hawkins J, Hussain I, MacPherson D, Maile G, MaticoR, Milner P, Mosley J, Naylor A, O’Brien A, RedshawS, Riddell D, Rowland P, Soleil V, Smith KJ, Stanway S,Stemp G, Sweitzer S, Theobald P, Vesey D, Walter DS,

Ward J, Wayne G (2008) BACE-1 inhibitors part 2: Iden-tification of hydroxy ethylamines (HEAs) with reducedpeptidic character. Bioorg Med Chem Lett 18, 1017-1021.

[791] Clarke B, Cutler L, Demont E, Dingwall C, DunsdonR, Hawkins J, Howes C, Hussain I, Maile G, Matico R,Mosley J, Naylor A, O’Brien A, Redshaw S, RowlandP, Soleil V, Smith KJ, Sweitzer S, Theobald P, Vesey D,Walter DS, Wayne G (2010) BACE-1 hydroxyethylamineinhibitors using novel edge-to-face interaction with Arg-296. Bioorg Med Chem Lett 20, 4639-4644.

[792] May PC, Dean RA, Lowe SL, Martenyi F, Sheehan SM,Boggs LN, Monk SA, Mathes BM, Mergott DJ, WatsonBM, Stout SL, Timm DE, Smith LE, Gonzales CR, NakanoM, Jhee SS, Yen M, Ereshefsky L, Lindstrom TD, Cal-ligaro DO, Cocke PJ, Greg HD, Friedrich S, Citron M,Audia JE (2011) Robust central reduction of amyloid-betain humans with an orally available, non-peptidic beta-secretase inhibitor. J Neurosci 31, 16507-16516.

[793] Fukumoto H, Takahashi H, Tarui N, Matsui J, Tomita T,Hirode M, Sagayama M, Maeda R, Kawamoto M, HiraiK, Terauchi J, Sakura Y, Kakihana M, Kato K, Iwatsubo T,Miyamoto M (2010) A noncompetitive BACE1 inhibitorTAK-070 ameliorates Abeta pathology and behavioraldeficits in a mouse model of Alzheimer’s disease. J Neu-rosci 30, 11157-11166.

[794] Takahashi H, Fukumoto H, Maeda R, Terauchi J, KatoK, Miyamoto M (2010) Ameliorative effects of a non-competitive BACE1 inhibitor TAK-070 on Abeta peptidelevels and impaired learning behavior in aged rats. BrainRes 1361, 146-156.

[795] Yang W, Lu W, Lu Y, Zhong M, Sun J, Thomas AE, Wilkin-son JM, Fucini RV, Lam M, Randal M, Shi XP, JacobsJW, McDowell RS, Gordon EM, Ballinger MD (2006)Aminoethylenes: A tetrahedral intermediate isostere yield-ing potent inhibitors of the aspartyl protease BACE-1.J Med Chem 49, 839-842.

[796] Yang W, Fucini RV, Fahr BT, Randal M, Lind KE, LamMB, Lu W, Lu Y, Cary DR, Romanowski MJ, Colussi D,Pietrak B, Allison TJ, Munshi SK, Penny DM, Pham P, SunJ, Thomas AE, Wilkinson JM, Jacobs JW, McDowell RS,Ballinger MD (2009) Fragment-based discovery of non-peptidic BACE-1 inhibitors using tethering. Biochemistry48, 4488-4496.

[797] Sato T, Diehl TS, Narayanan S, Funamoto S, Ihara Y, DeSB, Steiner H, Haass C, Wolfe MS (2007) Active gamma-secretase complexes contain only one of each component.J Biol Chem 282, 33985-33993.

[798] Haass C, Steiner H (2002) Alzheimer disease gamma-secretase: A complex story of GxGD-type presenilinproteases. Trends Cell Biol 12, 556-562.

[799] Sisodia SS, St George-Hyslop PH (2002) gamma-Secretase, Notch, Abeta and Alzheimer’s disease: Wheredo the presenilins fit in? Nat Rev Neurosci 3, 281-290.

[800] Tsai JY, Wolfe MS, Xia W (2002) The search for gamma-secretase and development of inhibitors. Curr Med Chem9, 1087-1106.

[801] Knappenberger KS, Tian G, Ye X, Sobotka-BrinerC, Ghanekar SV, Greenberg BD, Scott CW (2004)Mechanism of gamma-secretase cleavage activation: Isgamma-secretase regulated through autoinhibition involv-ing the presenilin-1 exon 9 loop?. Biochemistry 43,6208-6218.

[802] Steiner H (2004) Uncovering gamma-secretase. CurrAlzheimer Res 1, 175-181.

Page 79: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 625

[803] Steiner H, Than M, Bode W, Haass C (2006) Pore-formingscissors? A first structural glimpse of gamma-secretase.Trends Biochem Sci 31, 491-493.

[804] Steiner H (2008) The catalytic core of gamma-secretase:Presenilin revisited. Curr Alzheimer Res 5, 147-157.

[805] Steiner H, Winkler E, Haass C (2008) Chemical cross-linking provides a model of the gamma-secretase complexsubunit architecture and evidence for close proximity ofthe C-terminal fragment of presenilin with APH-1. J BiolChem 283, 34677-34686.

[806] Lazarov VK, Fraering PC, Ye W, Wolfe MS, Selkoe DJ,Li H (2006) Electron microscopic structure of purified,active gamma-secretase reveals an aqueous intramem-brane chamber and two pores. Proc Natl Acad Sci U S A103, 6889-6894.

[807] Sato C, Morohashi Y, Tomita T, Iwatsubo T (2006) Struc-ture of the catalytic pore of gamma-secretase probed bythe accessibility of substituted cysteines. J Neurosci 26,12081-12088.

[808] Sato C, Takagi S, Tomita T, Iwatsubo T (2008) TheC-terminal PAL motif and transmembrane domain 9 ofpresenilin 1 are involved in the formation of the cat-alytic pore of the gamma-secretase. J Neurosci 28, 6264-6271.

[809] Kreft AF, Martone R, Porte A (2009) Recent advances inthe identification of gamma-secretase inhibitors to clini-cally test the Abeta oligomer hypothesis of Alzheimer’sdisease. J Med Chem 52, 6169-6188.

[810] Krishnaswamy S, Verdile G, Groth D, Kanyenda L, Mar-tins RN (2009) The structure and function of Alzheimer’sgamma secretase enzyme complex. Crit Rev Clin Lab Sci46, 282-301.

[811] Takagi S, Tominaga A, Sato C, Tomita T, IwatsuboT (2010) Participation of transmembrane domain 1 ofpresenilin 1 in the catalytic pore structure of the gamma-secretase. J Neurosci 30, 15943-15950.

[812] Wolfe MS (2010) Structure, mechanism and inhibition ofgamma-secretase and presenilin-like proteases. Biol Chem391, 839-847.

[813] Page RM, Baumann K, Tomioka M, Perez-Revuelta BI,Fukumori A, Jacobsen H, Flohr A, Luebbers T, OzmenL, Steiner H, Haass C (2008) Generation of Abeta38and Abeta42 is independently and differentially affectedby familial Alzheimer disease-associated presenilin muta-tions and gamma-secretase modulation. J Biol Chem 283,677-683.

[814] Page RM, Gutsmiedl A, Fukumori A, Winkler E, Haass C,Steiner H (2010) Beta-amyloid precursor protein mutantsrespond to gamma-secretase modulators. J Biol Chem 285,17798-17810.

[815] Takami M, Nagashima Y, Sano Y, Ishihara S, Morishima-Kawashima M, Funamoto S, Ihara Y (2009) gamma-Secretase: Successive tripeptide and tetrapeptide releasefrom the transmembrane domain of beta-carboxyl terminalfragment. J Neurosci 29, 13042-13052.

[816] Tolia A, De Strooper B (2009) Structure and function ofgamma-secretase. Semin Cell Dev Biol 20, 211-218.

[817] Chavez-Gutierrez L, Bammens L, Benilova I, VandersteenA, Benurwar M, Borgers M, Lismont S, Zhou L, Van CS,Esselmann H, Wiltfang J, Serneels L, Karran E, Gijsen H,Schymkowitz J, Rousseau F, Broersen K, De Strooper B(2012) The mechanism of gamma-Secretase dysfunctionin familial Alzheimer disease. EMBO J 31, 2261-2274.

[818] De Strooper B, Annaert W, Cupers P, Saftig P, CraessaertsK, Mumm JS, Schroeter EH, Schrijvers V, Wolfe MS, Ray

WJ, Goate A, Kopan R (1999) A presenilin-1-dependentgamma-secretase-like protease mediates release of Notchintracellular domain. Nature 398, 518-522.

[819] Lundkvist J, Naslund J (2007) Gamma-secretase: A com-plex target for Alzheimer’s disease. Curr Opin Pharmacol7, 112-118.

[820] Lleo A (2008) Activity of gamma-secretase on substratesother than APP. Curr Top Med Chem 8, 9-16.

[821] Oehlrich D, Berthelot DJ, Gijsen HJ (2011) gamma-secretase modulators as potential disease modifyinganti-Alzheimer’s drugs. J Med Chem 54, 669-698.

[822] Searfoss GH, Jordan WH, Calligaro DO, Galbreath EJ,Schirtzinger LM, Berridge BR, Gao H, Higgins MA, MayPC, Ryan TP (2003) Adipsin, a biomarker of gastroin-testinal toxicity mediated by a functional gamma-secretaseinhibitor. J Biol Chem 278, 46107-46116.

[823] Milano J, McKay J, Dagenais C, Foster-Brown L, PognanF, Gadient R, Jacobs RT, Zacco A, Greenberg B, CiaccioPJ (2004) Modulation of notch processing by gamma-secretase inhibitors causes intestinal goblet cell metaplasiaand induction of genes known to specify gut secretorylineage differentiation. Toxicol Sci 82, 341-358.

[824] Wong GT, Manfra D, Poulet FM, Zhang Q, Josien H, BaraT, Engstrom L, Pinzon-Ortiz M, Fine JS, Lee HJ, ZhangL, Higgins GA, Parker EM (2004) Chronic treatment withthe gamma-secretase inhibitor LY-411,575 inhibits beta-amyloid peptide production and alters lymphopoiesis andintestinal cell differentiation. J Biol Chem 279, 12876-12882.

[825] Grosveld GC (2009) Gamma-secretase inhibitors: Notchso bad. Nat Med 15, 20-21.

[826] Rao SS, O’Neil J, Liberator CD, Hardwick JS, Dai X,Zhang T, Tyminski E, Yuan J, Kohl NE, Richon VM, Vander Ploeg LH, Carroll PM, Draetta GF, Look AT, StrackPR, Winter CG (2009) Inhibition of NOTCH signalingby gamma secretase inhibitor engages the RB pathwayand elicits cell cycle exit in T-cell acute lymphoblasticleukemia cells. Cancer Res 69, 3060-3068.

[827] Jorissen E, De Strooper B (2010) Gamma-secretase andthe intramembrane proteolysis of Notch. Curr Top DevBiol 92, 201-230.

[828] Oh SY, Ellenstein A, Chen CD, Hinman JD, Berg EA,Costello CE, Yamin R, Neve RL, Abraham CR (2005)Amyloid precursor protein interacts with notch receptors.J Neurosci Res 82, 32-42.

[829] Dovey HF, John V, Anderson JP, Chen LZ, de Saint AP,Fang LY, Freedman SB, Folmer B, Goldbach E, Hol-sztynska EJ, Hu KL, Johnson-Wood KL, Kennedy SL,Kholodenko D, Knops JE, Latimer LH, Lee M, Liao Z,Lieberburg IM, Motter RN, Mutter LC, Nietz J, Quinn KP,Sacchi KL, Seubert PA, Shopp GM, Thorsett ED, TungJS, Wu J, Yang S, Yin CT, Schenk DB, May PC, Alt-stiel LD, Bender MH, Boggs LN, Britton TC, ClemensJC, Czilli DL, Dieckman-McGinty DK, Droste JJ, FusonKS, Gitter BD, Hyslop PA, Johnstone EM, Li WY, Lit-tle SP, Mabry TE, Miller FD, Audia JE (2001) Functionalgamma-secretase inhibitors reduce beta-amyloid peptidelevels in brain. J Neurochem 76, 173-181.

[830] Beher D, Shearman MS (2002) Gamma-secretase inhibi-tion. Biochem Soc Trans 30, 534-537.

[831] Josien H (2002) Recent advances in the development ofgamma-secretase inhibitors. Curr Opin Drug Discov Devel5, 513-525.

[832] Rochette MJ, Murphy MP (2002) Gamma-secretase: Sub-strates and inhibitors. Mol Neurobiol 26, 81-95.

Page 80: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

626 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

[833] Wolfe MS, Esler WP, Das C (2002) Continuing strate-gies for inhibiting Alzheimer’s gamma-secretase. J MolNeurosci 19, 83-87.

[834] Xu M, Lai MT, Huang Q, Muzio-Mower J, Castro JL,Harrison T, Nadin A, Neduvelil JG, Shearman MS, ShaferJA, Gardell SJ, Li YM (2002) gamma-Secretase: Charac-terization and implication for Alzheimer disease therapy.Neurobiol Aging 23, 1023-1030.

[835] Harrison T, Beher D (2003) gamma-Secretase inhibitors–from molecular probes to new therapeutics? Prog MedChem 41, 99-127.

[836] Harrison T, Churcher I, Beher D (2004) gamma-Secretaseas a target for drug intervention in Alzheimer’s disease.Curr Opin Drug Discov Devel 7, 709-719.

[837] Marjaux E, De Strooper B (2004) Gamma-Secretaseinhibitors: Still in the running as Alzheimer’s therapeutics.Drug Discov Today Therap Strategies 1, 1-6.

[838] Churcher I, Beher D (2005) Gamma-secretase as a thera-peutic target for the treatment of Alzheimer’s disease. CurrPharm Des 11, 3363-3382.

[839] Pollack SJ, Lewis H (2005) Secretase inhibitors forAlzheimer’s disease: Challenges of a promiscuous pro-tease. Curr Opin Investig Drugs 6, 35-47.

[840] Barten DM, Meredith JE Jr, Zaczek R, Houston JG,Albright CF (2006) Gamma-secretase inhibitors forAlzheimer’s disease: Balancing efficacy and toxicity.Drugs R D 7, 87-97.

[841] Evin G, Sernee MF, Masters CL (2006) Inhibitionof gamma-secretase as a therapeutic intervention forAlzheimer’s disease: Prospects, limitations and strategies.CNS Drugs 20, 351-372.

[842] Tomita T, Iwatsubo T (2006) gamma-secretase as a ther-apeutic target for treatment of Alzheimer’s disease. CurrPharm Des 12, 661-670.

[843] Ziani-Cherif C, Mostefa-Kara B, Brixi-Gormat FZ(2006) Gamma-secretase as a pharmacological target inAlzheimer disease research: When, why and how? CurrPharm Des 12, 4313-4335.

[844] Olson RE, Albright CF (2008) Recent progress in themedicinal chemistry of gamma-secretase inhibitors. CurrTop Med Chem 8, 17-33.

[845] Jakob-Roetne R, Jacobsen H (2009) Alzheimer’s disease:From pathology to therapeutic approaches. Angew ChemInt Ed Engl 4, 3030-3059.

[846] Wu WL, Zhang L (2009) Gamma-Secretase inhibitors forthe treatment of Alzheimer’s disease. Drug Dev Res 70,94-100.

[847] Woo HN, Park JS, Gwon AR, Arumugam TV, Jo DG(2009) Alzheimer’s disease and Notch signaling. BiochemBiophys Res Commun 390, 1093-1097.

[848] Augelli-Szafran CE, Wei HX, Lu D, Zhang J, Gu Y,Yang T, Osenkowski P, Ye W, Wolfe MS (2010) Dis-covery of notch-sparing gamma-secretase inhibitors. CurrAlzheimer Res 7, 207-209.

[849] Guardia-Laguarta C, Pera M, Lleo A (2010) gamma-Secretase as a therapeutic target in Alzheimer’s disease.Curr Drug Targets 11, 506-517.

[850] Lleo A, Saura CA (2011) gamma-secretase substrates andtheir implications for drug development in Alzheimer’sdisease. Curr Top Med Chem 11, 1513-1527.

[851] Wolfe MS (2012) gamma-Secretase inhibitors and mod-ulators for Alzheimer’s disease. J Neurochem 120(Suppl1), 89-98.

[852] Barthet G, Georgakopoulos A, Robakis NK (2012)Cellular mechanisms of gamma-secretase substrate selec-

tion, processing and toxicity. Prog Neurobiol 98, 166-175.

[853] Imbimbo BP (2008) Therapeutic potential of gamma-secretase inhibitors and modulators. Curr Top Med Chem8, 54-61.

[854] Wolfe MS (2008) Gamma-secretase inhibition and mod-ulation for Alzheimer’s disease. Curr Alzheimer Res 5,158-164.

[855] Wolfe MS (2008) Inhibition and modulation of gamma-Secretase for Alzheimer’s disease. Neurotherapeutics 5,391-398.

[856] Tomita T (2009) Secretase inhibitors and modulators forAlzheimer’s disease treatment. Expert Rev Neurother 9,661-679.

[857] Bergmans BA, De Strooper B (2010) gamma-secretases:From cell biology to therapeutic strategies. Lancet Neurol9, 215-226.

[858] Ross JS, Imbimbo BP (2010) Are gamma-secretaseinhibitors detrimental for Alzheimer’s disease patients?.J Alzheimers Dis 22, 401-404.

[859] Imbimbo BP, Giardina GA (2011) gamma-secretaseinhibitors and modulators for the treatment of Alzheimer’sdisease: Disappointments and hopes. Curr Top Med Chem11, 1555-1570.

[860] Imbimbo BP, Panza F, Frisardi V, Solfrizzi V, D’OnofrioG, Logroscino G, Seripa D, Pilotto A (2011) Therapeuticintervention for Alzheimer’s disease with gamma-secretase inhibitors: Still a viable option? Expert OpinInvestig Drugs 20, 325-341.

[861] Panza F, Frisardi V, Solfrizzi V, Imbimbo BP, LogroscinoG, Santamato A, Greco A, Seripa D, Pilotto A (2011)Interacting with gamma-secretase for treating Alzheimer’sdisease: From inhibition to modulation. Curr Med Chem18, 5430-5447.

[862] Sambamurti K, Greig NH, Utsuki T, Barnwell EL, SharmaE, Mazell C, Bhat NR, Kindy MS, Lahiri DK, Pappolla MA(2011) Targets for AD treatment: Conflicting messagesfrom gamma-secretase inhibitors. J Neurochem 117, 359-374.

[863] D’Onofrio G, Panza F, Frisardi V, Solfrizzi V, ImbimboBP, Paroni G, Cascavilla L, Seripa D, Pilotto A(2012) Advances in the identification of gamma-secretaseinhibitors for the treatment of Alzheimer’s disease. ExpertOpin Drug Discov 7, 19-37.

[864] Tong G, Wang JS, Sverdlov O, Huang SP, Slemmon R,Croop R, Castaneda L, Gu H, Wong O, Li H, BermanRM, Smith C, Albright CF, Dockens RC (2012) Mul-ticenter, randomized, double-blind, placebo-controlled,single-ascending dose study of the oral gamma-secretaseinhibitor BMS-708163 (Avagacestat): Tolerability pro-file, pharmacokinetic parameters, and pharmacodynamicmarkers. Clin Ther 34, 654-667.

[865] Coric V, van Dyck CH, Salloway S, Andreasen N, BrodyM, Richter RW, Soininen H, Thein S, Shiovitz T, PilcherG, Colby S, Rollin L, Dockens R, Pachai C, PorteliusE, Andreasson U, Blennow K, Soares H, Albright C,Feldman HH, Berman RM (2012) Safety and tolerabilityof the gamma-secretase inhibitor avagacestat in a Phase 2study of mild to moderate Alzheimer disease. Arch Neu-rol, doi: 10.1001/archneurol.2012.2194 [Epubl ahead ofprint].

[866] Gu H, Deng Y, Wang J, Aubry AF, Arnold ME(2010) Development and validation of sensitive andselective LC-MS/MS methods for the determina-tion of BMS-708163, a gamma-secretase inhibitor,

Page 81: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 627

in plasma and cerebrospinal fluid using deprotonatedor formate adduct ions as precursor ions. J Chro-matogr B Analyt Technol Biomed Life Sci 878, 2319-2326.

[866] [3]Dockens R, Wang JS, Castaneda L, Sverdlov O, HuangSP, Slemmon R, Gu H, Wong O, Li H, Berman RM, SmithC, Albright CF, Tong G (2012) A Placebo-Controlled,Multiple Ascending Dose Study to Evaluate the Safety,Pharmacokinetics and Pharmacodynamics of Avagacestat(BMS-708163) in Healthy Young and Elderly Subjects.Clin Pharmacokinet 51, 681-693.

[867] Prasad CV, Wallace OB, Noonan JW, Sloan CP, Lau W,Vig S, Parker MF, Smith DW, Hansel SB, Polson CT,Barten DM, Felsenstein KM, Roberts SB (2004) Hydrox-ytriamides as potent gamma-secretase inhibitors. BioorgMed Chem Lett 14, 1917-1921.

[868] Prasad CV, Vig S, Smith DW, Gao Q, Polson CT, Corsa JA,Guss VL, Loo A, Barten DM, Zheng M, Felsenstein KM,Roberts SB (2004) 2,3-Benzodiazepin-1,4-diones as pep-tidomimetic inhibitors of gamma-secretase. Bioorg MedChem Lett 14, 3535-3538.

[869] Prasad CV, Zheng M, Vig S, Bergstrom C, Smith DW, GaoQ, Yeola S, Polson CT, Corsa JA, Guss VL, Loo A, WangJ, Sleczka BG, Dangler C, Robertson BJ, Hendrick JP,Roberts SB, Barten DM (2007) Discovery of (S)-2-((S)-2-(3,5-difluorophenyl)-2-hydroxyacetamido)-N-((S,Z)-3-methyl-4-oxo-4,5-dihydro-3H-benzo[d][1,2]diazepin-5-yl) propanamide (BMS-433796): A gamma-secretaseinhibitor with Abeta lowering activity in a transgenicmouse model of Alzheimer’s disease. Bioorg Med ChemLett 17, 4006-4011.

[870] Yan XX, Li T, Rominger CM, Prakash SR, Wong PC,Olson RE, Zaczek R, Li YW (2004) Binding sites ofgamma-secretase inhibitors in rodent brain: Distribution,postnatal development, and effect of deafferentation. JNeurosci 24, 2942-2952.

[871] Thompson LA, Liauw AY, Ramanjulu MM, Kasireddy-Polam P, Mercer SE, Maduskuie TP, Glicksman M, RoachAH, Meredith JE, Liu RQ, Combs AP, Higaki JN, CordellB, Seiffert D, Zaczek RC, Robertson DW, Olson RE(2006) Synthesis and evaluation of succinoyl-caprolactamgamma-secretase inhibitors. Bioorg Med Chem Lett 16,2357-2363.

[872] Parker MF, Barten DM, Bergstrom CP, Bronson JJ, CorsaJA, Deshpande MS, Felsenstein KM, Guss VL, HanselSB, Johnson G, Keavy DJ, Lau WY, Mock J, PrasadCV, Polson CT, Sloan CP, Smith DW, Wallace OB,Wang HH, Williams A, Zheng M (2007) N-(5-chloro-2-(hydroxymethyl)-N-alkyl-arylsulfonamides as gamma-secretase inhibitors. Bioorg Med Chem Lett 17, 4432-4436.

[873] Parker MF, Bronson JJ, Barten DM, Corsa JA, Du W,Felsenstein KM, Guss VL, Izzarelli D, Loo A, McEl-hone KE, Marcin LR, Padmanabha R, Pak R, Polson CT,Toyn JH, Varma S, Wang J, Wong V, Zheng M, RobertsSB (2007) Amino-caprolactam derivatives as gamma-secretase inhibitors. Bioorg Med Chem Lett 17, 5790-5795.

[874] Yang MG, Shi JL, Modi DP, Wells J, Cochran BM, WolfMA, Thompson LA, Ramanjulu MM, Roach AH, ZaczekR, Robertson DW, Wexler RR, Olson RE (2007) Designand synthesis of benzoazepinone-derived cyclic mal-onamides and aminoamides as potent gamma-secretaseinhibitors. Bioorg Med Chem Lett 17, 3910-3915.

[875] Bergstrom CP, Sloan CP, Wang HH, Parker MF, Smith DW,Zheng M, Hansel SB, Polson CT, Barber LE, Bursuker I,Guss VL, Corsa JA, Barten DM, Felsenstein KM, Roberts

SB (2008) Nitrogen-appended N-alkylsulfonamides asinhibitors of gamma-secretase. Bioorg Med Chem Lett 18,175-178.

[876] Bergstrom CP, Sloan CP, Lau WY, Smith DW, ZhengM, Hansel SB, Polson CT, Corsa JA, Barten DM,Felsenstein KM, Roberts SB (2008) Carbamate-appendedN-alkylsulfonamides as inhibitors of gamma-secretase.Bioorg Med Chem Lett 18, 464-468.

[877] Gillman KW, Starrett JE, Parker MF, Xie K, BronsonJJ, Marcin LR, McElhone KE, Bergstrom CP, Mate RA,Williams R, Meredith JE, Burton CR, Barten DM, ToynJH, Roberts SB, Lentz KA, Houston JG, Zaczek R,Albright CF, Decicco CP, Macor JE, Olson RE (2010) Dis-covery and evaluation of BMS-708163, a potent, selectiveand orally bioavailable gamma-secretase inhibitor. ACSMed Chem Lett 1, 120-124.

[878] Panza F, Frisardi V, Imbimbo BP, Capurso C, LogroscinoG, Sancarlo D, Seripa D, Vendemiale G, Pilotto A, Sol-frizzi V (2010) Review: Gamma-Secretase inhibitors forthe treatment of Alzheimer’s disease: The current state.CNS Neurosci Ther 16, 272-284.

[879] Shearman MS, Beher D, Clarke EE, Lewis HD, Harrison T,Hunt P, Nadin A, Smith AL, Stevenson G, Castro JL (2000)L-685,458, an aspartyl protease transition state mimic, is apotent inhibitor of amyloid beta-protein precursor gamma-secretase activity. Biochemistry 39, 8698-8704.

[880] Nadin A, Sanchez Lopez JM, Neduvelil JG, ThomasSR (2001) A stereocontrolled synthesis of 2R-benzyl-5S-tert-butoxycarbonyl-amino-4R-(tert- butyld-imethylsilanyloxy)-6-phenyl-hexanoic acid (Phe-Phehydroxy-ethylene dipeptide isostere). Tetrahedron 57,1861-1864.

[881] Nadin A, Owens AP, Castro JL, Harrison T, ShearmanMS (2003) Synthesis and gamma-secretase activity ofAPP substrate-based hydroxyethylene dipeptide isosteres.Bioorg Med Chem Lett 13, 37-41.

[882] Xu M, Lai MT, Huang Q, Muzio-Mower J, Castro JL,Harrison T, Nadin A, Neduvelil JG, Shearman MS, ShaferJA, Gardell SJ, Li YM (2002) gamma-Secretase: Charac-terization and implication for Alzheimer disease therapy.Neurobiol Aging 23, 1023-1030.

[883] Churcher I, Ashton K, Butcher JW, Clarke EE, HarrisonT, Lewis HD, Owens AP, Teall MR, Williams S, WrigleyJD (2003) A new series of potent benzodiazepine gamma-secretase inhibitors. Bioorg Med Chem Lett 13, 179-183.

[884] Churcher I, Williams S, Kerrad S, Harrison T, Castro JL,Shearman MS, Lewis HD, Clarke EE, Wrigley JD, BeherD, Tang YS, Liu W (2003) Design and synthesis of highlypotent benzodiazepine gamma-secretase inhibitors: Prepa-ration of (2S,3R)-3-(3,4-difluorophenyl)-2-(4-fluoro-phenyl)-4-hydroxy-N-((3S)-1-methyl-2-oxo-5-phenyl-2,3-dihydro-1H-benzo[e][1,4]-diazepin-3-yl)butyramideby use of an asymmetric Ireland-Claisen rearrangement.J Med Chem 46, 2275-2278.

[885] Churcher I, Beher D, Best JD, Castro JL, Clarke EE,Gentry A, Harrison T, Hitzel L, Kay E, Kerrad S, LewisHD, Morentin-Gutierrez P, Mortishire-Smith R, OakleyPJ, Reilly M, Shaw DE, Shearman MS, Teall MR, WilliamsS, Wrigley JD (2006) 4-substituted cyclohexyl sulfones aspotent, orally active gamma-secretase inhibitors. BioorgMed Chem Lett 16, 280-284.

[886] Scott JP, Lieberman DR, Beureux OM, Brands KM, DaviesAJ, Gibson AW, Hammond DC, McWilliams CJ, StewartGW, Wilson RD, Dolling UH (2007) A practical synthesis

Page 82: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

628 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

of a gamma-secretase inhibitor. J Org Chem 72, 4149-4155.

[887] Owens AP, Nadin A, Talbot AC, Clarke EE, HarrisonT, Lewis HD, Reilly M, Wrigley JD, Castro JL (2003)High affinity, bioavailable 3-amino-1,4-benzodiazepine-based gamma-secretase inhibitors. Bioorg Med Chem Lett13, 4143-4145.

[888] Lewis SJ, Smith AL, Neduvelil JG, Stevenson GI, LindonMJ, Jones AB, Shearman MS, Beher D, Clarke E, BestJD, Peachey JE, Harrison T, Castro JL (2005) A novelseries of potent gamma-secretase inhibitors based on abenzobicyclo[4.2.1]nonane core. Bioorg Med Chem Lett15, 373-378.

[889] Sparey T, Beher D, Best J, Biba M, Castro JL, ClarkeE, Hannam J, Harrison T, Lewis H, Madin A, ShearmanM, Sohal B, Tsou N, Welch C, Wrigley J (2005) Cyclicsulfamide gamma-secretase inhibitors. Bioorg Med ChemLett 15, 4212-4216.

[890] Sparey T, Clarke E, Hannam J, Harrison T, Madin A,Shearman M, Sohal B (2008) Sulfonamide derivatives ofbridgehead substituted bicyclo[4.2.1]nonanes as gamma-secretase inhibitors. Bioorg Med Chem Lett 18, 375-379.

[891] Jelley RA, Elliott J, Gibson KR, Harrison T, BeherD, Clarke EE, Lewis HD, Shearman M, Wrigley JD(2006) 3-Substituted gem-cyclohexane sulfone basedgamma-secretase inhibitors for Alzheimer’s disease: Con-formational analysis and biological activity. Bioorg MedChem Lett 16, 3839-3842.

[892] Scott JP, Oliver SF, Brands KM, Brewer SE, DaviesAJ, Gibb AD, Hands D, Keen SP, Sheen FJ, ReamerRA, Wilson RD, Dolling UH (2006) Practical asymmet-ric synthesis of a gamma-secretase inhibitor exploitingsubstrate-controlled intramolecular nitrile oxide-olefincycloaddition. J Org Chem 71, 3086-3092.

[893] Shaw D, Best J, Dinnell K, Nadin A, Shearman M, PattisonC, Peachey J, Reilly M, Williams B, Wrigley J, Harrison T(2006) 3,4-Fused cyclohexyl sulfones as gamma-secretaseinhibitors. Bioorg Med Chem Lett 16, 3073-3077.

[894] Davies AJ, Scott JP, Bishop BC, Brands KM, BrewerSE, Dasilva JO, Dormer PG, Dolling UH, Gibb AD,Hammond DC, Lieberman DR, Palucki M, Payack JF(2007) A novel crystallization-induced diastereomerictransformation based on a reversible carbon-sulfur bondformation. Application to the synthesis of a gamma-secretase inhibitor. J Org Chem 72, 4864-4871.

[895] Josien H, Bara T, Rajagopalan M, Asberom T, Clader JW,Favreau L, Greenlee WJ, Hyde LA, Nomeir AA, ParkerEM, Pissarnitski DA, Song L, Wong GT, Zhang L, ZhangQ, Zhao Z (2007) Small conformationally restricted piperi-dine N-arylsulfonamides as orally active gamma-secretaseinhibitors. Bioorg Med Chem Lett 17, 5330-5335.

[896] Josien H, Bara T, Rajagopalan M, Clader JW, GreenleeWJ, Favreau L, Hyde LA, Nomeir AA, Parker EM, SongL, Zhang L, Zhang Q (2009) Novel orally active morpho-line N-arylsulfonamides gamma-secretase inhibitors withlow CYP 3A4 liability. Bioorg Med Chem Lett 19, 6032-6037.

[897] Keown LE, Collins I, Cooper LC, Harrison T, Madin A,Mistry J, Reilly M, Shaimi M, Welch CJ, Clarke EE, LewisHD, Wrigley JD, Best JD, Murray F, Shearman MS (2009)Novel orally bioavailable gamma-secretase inhibitorswith excellent in vivo activity. J Med Chem 52, 3441-3444.

[898] Sasikumar TK, Qiang L, Burnett DA, Cole D, Xu R, Li H,Greenlee WJ, Clader. J, Zhang L, Hyde L (2010) Tricyclic

sulfones as orally active gamma-secretase inhibitors: Syn-thesis and structure-activity relationship studies. BioorgMed Chem Lett 20, 3632-3635.

[899] Sasikumar TK, Burnett DA, Asberom T, Wu WL, BennettC, Cole D, Xu R, Greenlee WJ, Clader J, Zhang L, HydeL (2010) Tetracyclic sulfones as potent gamma-secretaseinhibitors: Synthesis and structure-activity relationshipstudies. Bioorg Med Chem Lett 20, 3645-3648.

[900] Li H, Xu R, Cole D, Clader JW, Greenlee WJ, Nomeir AA,Song L, Zhang L (2010) Design, synthesis, and structure-activity relationship studies of N-arylsulfonyl morpholinesas gamma-secretase inhibitors. Bioorg Med Chem Lett 20,6606-6609.

[901] Lee J, Song L, Terracina G, Bara T, Josien H, Asberom T,Sasikumar TK, Burnett DA, Clader J, Parker EM, ZhangL (2011) Identification of presenilin 1-selective gamma-secretase inhibitors with reconstituted gamma-secretasecomplexes. Biochemistry 50, 4973-4980.

[902] Wu WL, Domalski M, Burnett DA, Josien H, Bara T,Rajagopalan M, Xu R, Clader J, Greenlee WJ, Brun-skill A, Hyde LA, Del Vecchio RA, Cohen-Williams ME,Song L, Lee J, Terracina G, Zhang Q, Nomeir A, ParkerEM, Zhang L (2012) Discovery of SCH-900229, a potentpresenilin 1 selective gamma-secretase inhibitor for thetreatment of Alzheimer’s disease. ACS Med Chem Lett,doi: 10.1021/ml300044f [Epubl ahead of print].

[903] Wolfe MS, Citron M, Diehl TS, Xia W, Donkor IO,Selkoe DJ (1998) A substrate-based difluoro ketoneselectively inhibits Alzheimer’s gamma-secretase activity.J Med Chem 41, 6-9.

[904] Wolfe MS, Xia W, Moore CL, Leatherwood DD,Ostaszewski B, Rahmati T, Donkor IO, Selkoe DJ(1999) Peptidomimetic probes and molecular model-ing suggest that Alzheimer’s gamma-secretase is anintramembrane-cleaving aspartyl protease 132. Biochem-istry 38, 4720-4727.

[905] Esler WP, Kimberly WT, Ostaszewski BL, Diehl TS,Moore CL, Tsai JY, Rahmati T, Xia W, Selkoe DJ, WolfeMS (2000) Transition-state analogue inhibitors of gamma-secretase bind directly to presenilin-1. Nat Cell Biol 2,428-434.

[906] Moore CL, Leatherwood DD, Diehl TS, Selkoe DJ, WolfeMS (2000) Difluoro ketone peptidomimetics suggest alarge S1 pocket for Alzheimer’s gamma-secretase: Impli-cations for inhibitor design. J Med Chem 43, 3434-3442.

[907] Moore CL, Diehl TS, Selkoe DJ, Wolfe MS (2000)Toward the characterization and identification of gamma-secretases using transition-state analogue inhibitors. AnnN Y Acad Sci 920, 197-205.

[908] Das C, Berezovska O, Diehl TS, Genet C, Buldyrev I,Tsai JY, Hyman BT, Wolfe MS (2003) Designed helicalpeptides inhibit an intramembrane protease. J Am ChemSoc 125, 11794-11795.

[909] Kornilova AY, Das C, Wolfe MS (2003) Differential effectsof inhibitors on the gamma-secretase complex. Mechanis-tic implications. J Biol Chem 278, 16470-16473.

[910] Bakshi P, Wolfe MS (2004) Stereochemical analysis of(hydroxyethyl)urea peptidomimetic inhibitors of gamma-secretase. J Med Chem 47, 6485-6489.

[911] Bihel F, Das C, Bowman MJ, Wolfe MS (2004) Discoveryof a Subnanomolar helical D-tridecapeptide inhibitor ofgamma-secretase. J Med Chem 47, 3931-3933.

[912] Best JD, Jay MT, Otu F, Churcher I, ReillyM, Morentin-Gutierrez P, Pattison C, HarrisonT, Shearman MS, Atack JR (2006) In vivo

Page 83: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 629

characterization of Abeta(40) changes in brain andcerebrospinal fluid using the novel gamma-secretaseinhibitor N-[cis-4-[(4-chlorophenyl)sulfonyl]-4-(2,5-difluorophenyl)cyclohexyl]-1,1,1-trifluoromethane-sulfonamide (MRK-560) in the rat. J Pharmacol ExpTher 317, 786-790.

[913] Best JD, Smith DW, Reilly MA, O’Donnell R, LewisHD, Ellis S, Wilkie N, Rosahl TW, Laroque PA,Boussiquet-Leroux C, Churcher I, Atack JR, HarrisonT, Shearman MS (2007) The novel gamma secretaseinhibitor N-[cis-4-[(4-chlorophenyl)sulfonyl]-4-(2,5-difluorophenyl)cyclohexyl]-1,1, 1-trifluoromethane-sulfonamide (MRK-560) reduces amyloid plaquedeposition without evidence of notch-related pathology inthe Tg2576 mouse. J Pharmacol Exp Ther 320, 552-558.

[914] Townsend M, Qu Y, Gray A, Wu Z, Seto T, Hutton M,Shearman MS, Middleton RE (2010) Oral treatment witha gamma-secretase inhibitor improves long-term potentia-tion in a mouse model of Alzheimer’s disease. J PharmacolExp Ther 333, 110-119.

[915] Lee J, Song L, Terracina G, Bara T, Josien H, Asberom T,Sasikumar TK, Burnett DA, Clader J, Parker EM, ZhangL (2011) Identification of presenilin 1-selective gamma-secretase inhibitors with reconstituted gamma-secretasecomplexes. Biochemistry 50, 4973-4980.

[916] Teall M, Oakley P, Harrison T, Shaw D, Kay E, Elliott J,Gerhard U, Castro JL, Shearman M, Ball RG, Tsou NN(2005) Aryl sulfones: A new class of gamma-secretaseinhibitors. Bioorg Med Chem Lett 15, 2685-2688.

[917] Asberom T, Bara TA, Clader JW, Greenlee WJ, GuzikHS, Josien HB, Li W, Parker EM, Pissarnitski DA, SongL, Zhang L, Zhao Z (2007) Tetrahydroquinoline sulfon-amides as gamma-secretase inhibitors. Bioorg Med ChemLett 17, 205-207.

[918] Asberom T, Zhao Z, Bara TA, Clader JW, Greenlee WJ,Hyde LA, Josien HB, Li W, McPhail AT, Nomeir AA,Parker EM, Rajagopalan M, Song L, Wong GT, Zhang L,Zhang Q, Pissarnitski DA (2007) Discovery of gamma-secretase inhibitors efficacious in a transgenic animalmodel of Alzheimer’s disease. Bioorg Med Chem Lett 17,511-516.

[919] Guo T, Gu H, Hobbs DW, Rokosz LL, Stauffer TM, JacobB, Clader JW (2007) Design, synthesis, and evaluationof tetrahydroquinoline and pyrrolidine sulfonamide car-bamates as gamma-secretase inhibitors. Bioorg Med ChemLett 17, 3010-3013.

[920] Li H, Asberom T, Bara TA, Clader JW, Greenlee WJ,Josien HB, McBriar MD, Nomeir A, Pissarnitski DA,Rajagopalan M, Xu R, Zhao Z, Song L, Zhang L(2007) Discovery of 2,4,6-trisubstituted N-arylsulfonylpiperidines as gamma-secretase inhibitors. Bioorg MedChem Lett 17, 6290-6294.

[921] Pissarnitski DA, Asberom T, Bara TA, Buevich AV, CladerJW, Greenlee WJ, Guzik HS, Josien HB, Li W, McE-wan M, McKittrick BA, Nechuta TL, Parker EM, SinningL, Smith EM, Song L, Vaccaro HA, Voigt JH, Zhang L,Zhang Q, Zhao Z (2007) 2,6-Disubstituted N-arylsulfonylpiperidines as gamma-secretase inhibitors. Bioorg MedChem Lett 17, 57-62.

[922] McBriar MD, Clader JW, Chu I, Del Vecchio RA, FavreauL, Greenlee WJ, Hyde LA, Nomeir AA, Parker EM, Pissar-nitski DA, Song L, Zhang L, Zhao Z (2008) Discovery ofamide and heteroaryl isosteres as carbamate replacementsin a series of orally active gamma-secretase inhibitors.Bioorg Med Chem Lett 18, 215-219.

[923] Vaccaro HA, Zhao Z, Clader JW, Song L, Terracina G,Zhang L, Pissarnitski DA (2008) Solution-phase paral-lel synthesis of carbamates as gamma-secretase inhibitors.J Comb Chem 10, 56-62.

[924] Xu R, Cole D, Asberom T, Bara T, Bennett C, Burnett DA,Clader J, Domalski M, Greenlee W, Hyde L, Josien H,Li H, McBriar M, McKittrick B, McPhail AT, PissarnitskiD, Qiang L, Rajagopalan M, Sasikumar T, Su J, Tang H,Wu WL, Zhang L, Zhao Z (2010) Design and synthesisof tricyclic sulfones as gamma-secretase inhibitors withgreatly reduced Notch toxicity. Bioorg Med Chem Lett 20,2591-2596.

[925] Su J, Tang H, McKittrick BA, Xu R, Clader JW, Green-lee WJ, Hyde L, Zhang L (2011) Synthesis and SARstudy of tricyclic sulfones as gamma-secretase inhibitors:C-6 and C-8 positions. Bioorg Med Chem Lett 21, 3447-3451.

[926] Sun ZY, Asberom T, Bara T, Bennett C, Burnett D, ChuI, Clader J, Cohen-Williams M, Cole D, Czarniecki M,Durkin J, Gallo G, Greenlee W, Josien H, Huang X, HydeL, Jones N, Kazakevich I, Li H, Liu X, Lee J, Maccoss M,Mandal MB, McCracken T, Nomeir A, Mazzola R, PalaniA, Parker EM, Pissarnitski DA, Qin J, Song L, TerracinaG, Vicarel M, Voigt J, Xu R, Zhang L, Zhang Q, Zhao Z,Zhu X, Zhu Z (2012) Cyclic hydroxyamidines as amideisosteres: Discovery of oxadiazolines and oxadiazines aspotent and highly efficacious gamma-secretase modulatorsin vivo. J Med Chem 55, 489-502.

[927] Close J, Heidebrecht R Jr, Hendrix J, Li C, Munoz B,Surdi L, Kattar S, Tempest P, Moses P, Geng X, HughesB, Smotrov N, Moxham C, Chapnick J, Kariv I, Nikov G,Burke JE, Deshmukh S, Jeliazkova-Mecheva V, Leach JK,Diaz D, Xu L, Yang Z, Kwei G, Moy L, Shah S, Tanga F,Kenefic C, Savage D, Shearman M, Ball RG, McNevin MJ,Markarewicz A, Miller T (2012) Lead optimization of 4,4-biaryl piperidine amides as gamma-secretase inhibitors.Bioorg Med Chem Lett 22, 3203-3207.

[928] Burt J (2010) American Chemical Society - 240th nationalmeeting - chemistry for preventing and combating disease:Part 1. IDrugs 13, 669-672.

[929] Stepan AF, Subramanyam C, Efremov IV, Dutra JK,O’Sullivan TJ, Dirico KJ, McDonald WS, Won A, DorffPH, Nolan CE, Becker SL, Pustilnik LR, Riddell DR,Kauffman GW, Kormos BL, Zhang L, Lu Y, Capetta SH,Green ME, Karki K, Sibley E, Atchison KP, Hallgren AJ,Oborski CE, Robshaw AE, Sneed B, O’Donnell CJ (2012)Application of the bicyclo[1.1.1]pentane motif as a non-classical phenyl ring bioisostere in the design of a potentand orally active gamma-secretase inhibitor. J Med Chem55, 3414-3424.

[930] Lanz TA, Himes CS, Pallante G, Adams L, Yamazaki S,Amore B, Merchant KM (2003) The gamma-secretaseinhibitor N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester reduces A beta levels in vivoin plasma and cerebrospinal fluid in young (plaque-free)and aged (plaque-bearing) Tg2576 mice. J PharmacolExp Ther 305, 864-871.

[931] Lanz TA, Hosley JD, Adams WJ, Merchant KM (2004)Studies of Abeta pharmacodynamics in the brain, cere-brospinal fluid, and plasma in young (plaque-free) Tg2576mice using the gamma-secretase inhibitor N2-[(2S)-2-(3,5- difluorophenyl)-2-hydroxyethanoyl]-N1-[(7S)-5-methyl-6-oxo -6,7-dihydro-5H-dibenzo[b,d]azepin-7-yl]-L-alaninamide (LY-411575). J Pharmacol Exp Ther 309,49-55.

Page 84: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

630 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

[932] Lanz TA, Karmilowicz MJ, Wood KM, Pozdnyakov N,Du P, Piotrowski MA, Brown TM, Nolan CE, RichterKE, Finley JE, Fei Q, Ebbinghaus CF, Chen YL, Sprack-lin DK, Tate B, Geoghegan KF, Lau LF, Auperin DD,Schachter JB (2006) Concentration-dependent modulationof amyloid-beta in vivo and in vitro using the gamma-secretase inhibitor, LY-450139. J Pharmacol Exp Ther319, 924-933.

[933] Chen YL, Cherry K, Corman ML, Ebbinghaus CF, Gam-lath CB, Liston D, Martin BA, Oborski CE, SahaganBG (2007) Thiazole-diamides as potent gamma-secretaseinhibitors. Bioorg Med Chem Lett 17, 5518-5522.

[934] Brodney MA, Auperin DD, Becker SL, Bronk BS, BrownTM, Coffman KJ, Finley JE, Hicks CD, Karmilowicz MJ,Lanz TA, Liston D, Liu X, Martin BA, Nelson RB, NolanCE, Oborski CE, Parker CP, Richter KE, Pozdnyakov N,Sahagan BG, Schachter JB, Sokolowski SA, Tate B, WoodDE, Wood KM, Van Deusen JW, Zhang L (2011) Design,synthesis, and in vivo characterization of a novel series oftetralin amino imidazoles as gamma-secretase inhibitors:Discovery of PF-3084014. Bioorg Med Chem Lett 21,2637-2640.

[935] Stepan AF, Karki K, McDonald WS, Dorff PH, DutraJK, Dirico KJ, Won A, Subramanyam C, Efremov IV,O’Donnell CJ, Nolan CE, Becker SL, Pustilnik LR, SneedB, Sun H, Lu Y, Robshaw AE, Riddell D, O’SullivanTJ, Sibley E, Capetta S, Atchison K, Hallgren AJ,Miller E, Wood A, Obach RS (2011) Metabolism-directeddesign of oxetane-containing arylsulfonamide derivativesas gamma-secretase inhibitors. J Med Chem 54, 7772-7783.

[936] Lubbers T, Flohr A, Jolidon S, vid-Pierson P, Jacobsen H,Ozmen L, Baumann K (2011) Aminothiazoles as gamma-secretase modulators. Bioorg Med Chem Lett 21, 6554-6558.

[937] Rishton GM, Retz DM, Tempest PA, Novotny J, Kahn S,Treanor JJ, Lile JD, Citron M (2000) Fenchylamine sul-fonamide inhibitors of amyloid beta peptide productionby the gamma-secretase proteolytic pathway: Potentialsmall-molecule therapeutic agents for the treatment ofAlzheimer’s disease. J Med Chem 43, 2297-2299.

[938] Garofalo AW (2008) Patents targeting g-secretase inhi-bition and modulation for the treatment of Alzheimer’sdisease: 2004–2008. Expert Opin Ther Patents 18, 693-703.

[939] Zhao B, Yu M, Neitzel M, Marugg J, Jagodzinski J, LeeM, Hu K, Schenk D, Yednock T, Basi G (2008) Identifi-cation of gamma-secretase inhibitor potency determinantson presenilin. J Biol Chem 283, 2927-2938.

[940] Bowers S, Probst GD, Truong AP, Hom RK, Konradi AW,Sham HL, Garofalo AW, Wong K, Goldbach E, QuinnKP, Sauer JM, Wallace W, Nguyen L, Hemphill SS, BovaMP, Basi GS (2009) N-Bridged bicyclic sulfonamides asinhibitors of gamma-secretase. Bioorg Med Chem Lett 19,6952-6956.

[941] Truong AP, Aubele DL, Probst GD, Neitzel ML, SemkoCM, Bowers S, Dressen D, Hom RK, Konradi AW,Sham HL, Garofalo AW, Keim PS, Wu J, Dappen MS,Wong K, Goldbach E, Quinn KP, Sauer JM, BrighamEF, Wallace W, Nguyen L, Hemphill SS, Bova MP,Bard F, Yednock TA, Basi G (2009) Design, synthe-sis, and structure-activity relationship of novel orallyefficacious pyrazole/sulfonamide based dihydroquinolinegamma-secretase inhibitors. Bioorg Med Chem Lett 19,4920-4923.

[942] Basi GS, Hemphill S, Brigham EF, Liao A, Aubele DL,Baker J, Barbour R, Bova M, Chen XH, Dappen MS,Eichenbaum T, Goldbach E, Hawkinson J, Lawler-HerboldR, Hu K, Hui T, Jagodzinski JJ, Keim PS, Kholodenko D,Latimer LH, Lee M, Marugg J, Mattson MN, McCauley S,Miller JL, Motter R, Mutter L, Neitzel ML, Ni H, NguyenL, Quinn K, Ruslim L, Semko CM, Shapiro P, Smith J,Soriano F, Szoke B, Tanaka K, Tang P, Tucker JA, Ye XM,Yu M, Wu J, Xu YZ, Garofalo AW, Sauer JM, KonradiAW, Ness D, Shopp G, Pleiss MA, Freedman SB, SchenkD (2010) Amyloid precursor protein selective gamma-secretase inhibitors for treatment of Alzheimer’s disease.Alzheimers Res Ther 2, 36.

[943] Mattson MN, Neitzel ML, Quincy DA, Semko CM, Garo-falo AW, Keim PS, Konradi AW, Pleiss MA, Sham HL,Brigham EF, Goldbach EG, Zhang H, Sauer JM, BasiGS (2010) Discovery of sulfonamide-pyrazole gamma-secretase inhibitors. Bioorg Med Chem Lett 20, 2148-2150.

[944] Ye XM, Konradi AW, Smith J, Xu YZ, Dressen D, Garo-falo AW, Marugg J, Sham HL, Truong AP, JagodzinskiJ, Pleiss M, Zhang H, Goldbach E, Sauer JM, BrighamE, Bova M, Basi GS (2010) Discovery of a novelsulfonamide-pyrazolopiperidine series as potent and effi-cacious gamma-secretase inhibitors. Bioorg Med ChemLett 20, 2195-2199.

[945] Ye XM, Konradi AW, Smith J, Aubele DL, Garofalo AW,Marugg J, Neitzel ML, Semko CM, Sham HL, Sun M,Truong AP, Wu J, Zhang H, Goldbach E, Sauer JM,Brigham EF, Bova M, Basi GS (2010) Discovery of a novelsulfonamide-pyrazolopiperidine series as potent and effi-cacious gamma-secretase inhibitors (Part II). Bioorg MedChem Lett 20, 3502-3506.

[946] Aubele DL, Truong AP, Dressen DB, Probst GD, Bow-ers S, Mattson MN, Semko CM, Sun M, Garofalo AW,Konradi AW, Sham HL, Zmolek W, Wong K, Gold-bach E, Quinn KP, Sauer JM, Brigham EF, WallaceW, Nguyen L, Bova MP, Hemphill SS, Basi G (2011)Design, synthesis and structure-activity relationship ofnovel[3.3.1] bicyclic sulfonamide-pyrazoles as potentgamma-secretase inhibitors. Bioorg Med Chem Lett 21,5791-5794.

[947] Neitzel ML, Aubele DL, Marugg JL, Jagodzinski JJ, Kon-radi AW, Pleiss MA, Szoke B, Zmolek W, Goldbach E,Quinn KP, Sauer JM, Brigham EF, Wallace W, Bova MP,Hemphill S, Basi G (2011) Amino-caprolactam gamma-secretase inhibitors showing potential for the treatmentof Alzheimer’s disease. Bioorg Med Chem Lett 21, 3715-3720.

[948] Petit A, Bihel F, ves da CC, Pourquie O, Checler F,Kraus JL (2001) New protease inhibitors prevent gamma-secretase-mediated production of Abeta40/42 withoutaffecting Notch cleavage. Nat Cell Biol 3, 507-511.

[949] Petit A, Pasini A, ves da CC, Ayral E, Hernandez JF,Dumanchin-Njock C, Phiel CJ, Marambaud P, Wilk S,Farzan M, Fulcrand P, Martinez J, Andrau D, CheclerF (2003) JLK isocoumarin inhibitors: Selective gamma-secretase inhibitors that do not interfere with notchpathway in vitro or in vivo. J Neurosci Res 74, 370-377.

[950] Chun J, Yin YI, Yang G, Tarassishin L, Li YM (2004)Stereoselective synthesis of photoreactive peptidomimeticgamma-secretase inhibitors. J Org Chem 69, 7344-7347.

[951] Kan T, Tominari Y, Rikimaru K, Morohashi Y, Natsug-ari H, Tomita T, Iwatsubo T, Fukuyama T (2004) Parallelsynthesis of DAPT derivatives and their gamma-secretase-

Page 85: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 631

inhibitory activity. Bioorg Med Chem Lett 14, 1983-1985.

[952] Checler F, Alves da Costa C, Ayral E, Andrau D,Dumanchin C, Farzan M, Hernandez JF, Martinez J,Lefranc-Jullien S, Marambaud P, Pasini A, Petit A, PhielC, Robert P, St George-Hyslop P, Wilk S (2005) JLKinhibitors: Isocoumarin compounds as putative probesto selectively target the gamma-secretase pathway. CurrAlzheimer Res 2, 327-334.

[953] Fuwa H, Hiromoto K, Takahashi Y, Yokoshima S, Kan T,Fukuyama T, Iwatsubo T, Tomita T, Natsugari H (2006)Synthesis of biotinylated photoaffinity probes based onarylsulfonamide gamma-secretase inhibitors. Bioorg MedChem Lett 16, 4184-4189.

[954] Morohashi Y, Kan T, Tominari Y, Fuwa H, Okamura Y,Watanabe N, Sato C, Natsugari H, Fukuyama T, Iwat-subo T, Tomita T (2006) C-terminal fragment of presenilinis the molecular target of a dipeptidic gamma-secretase-specific inhibitor DAPT (N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester). J Biol Chem 281,14670-14676.

[955] Yang G, Yin YI, Chun J, Shelton CC, Ouerfelli O, Li YM(2009) Stereo-controlled synthesis of novel photoreactivegamma-secretase inhibitors. Bioorg Med Chem Lett 19,922-925.

[956] Yokoshima S, Abe Y, Watanabe N, Kita Y, Kan T, Iwat-subo T, Tomita T, Fukuyama T (2009) Development ofphotoaffinity probes for gamma-secretase equipped witha nitrobenzenesulfonamide-type cleavable linker. BioorgMed Chem Lett 19, 6869-6871.

[957] Hottecke N, Liebeck M, Baumann K, Schubenel R,Winkler E, Steiner H, Schmidt B (2010) Inhibition ofgamma-secretase by the CK1 inhibitor IC261 does notdepend on CK1delta. Bioorg Med Chem Lett 20, 2958-2963.

[958] Adeniji AO, Wells RM, Adejare A (2012) Syntheses andin-vitro evaluation of novel adamantane based gamma-secretase inhibitors. Curr Med Chem 19, 2458-2471.

[959] Nino H, Rodriguez-Borges JE, Garcia-Mera X, Prado-Prado F (2012) Review of synthesis, assay, and predictionof beta and gamma-secretase inhibitors. Curr Top MedChem 12, 828-844.

[960] Gundersen E, Fan K, Haas K, Huryn D, Steven JJ, Kreft A,Martone R, Mayer S, Sonnenberg-Reines J, Sun SC, ZhouH (2005) Molecular-modeling based design, synthesis,and activity of substituted piperidines as gamma-secretaseinhibitors. Bioorg Med Chem Lett 15, 1891-1894.

[961] Ravi Keerti A, Ashok KB, Parthasarathy T, Uma V (2005)QSAR studies-potent benzodiazepine gamma-secretaseinhibitors. Bioorg Med Chem 13, 1873-1878.

[962] Clarke EE, Churcher I, Ellis S, Wrigley JD, Lewis HD,Harrison T, Shearman MS, Beher D (2006) Intra- or inter-complex binding to the gamma-secretase enzyme. A modelto differentiate inhibitor classes. J Biol Chem 281, 31279-31289.

[963] Rishton GM, LaBonte K, Williams AJ, Kassam K,Kolovanov E (2006) Computational approaches to the pre-diction of blood-brain barrier permeability: A comparativeanalysis of central nervous system drugs versus secretaseinhibitors for Alzheimer’s disease. Curr Opin Drug DiscovDevel 9, 303-313.

[964] Sammi T, Silakari O, Ravikumar M (2008) Three-dimensional quantitative structure-activity relationshipmodeling of gamma-secretase inhibitors using molecularfield analysis. Chem Biol Drug Des 71, 155-166.

[965] Sammi T, Silakari O, Ravikumar M (2009) Three-dimensional quantitative structure-activity relationship(3D-QSAR) studies of various benzodiazepine analoguesof gamma-secretase inhibitors. J Mol Model 15, 343-348.

[966] Yang XG, Lv W, Chen YZ, Xue Y (2010) In silicoprediction and screening of gamma-secretase inhibitorsby molecular descriptors and machine learning methods.J Comput Chem 31, 1249-1258.

[967] Das R, Nachbar RB, Edelstein-Keshet L, Saltzman JS,Wiener MC, Bagchi A, Bailey J, Coombs D, SimonAJ, Hargreaves RJ, Cook JJ (2011) Modeling effect ofa gamma-secretase inhibitor on amyloid-beta dynamicsreveals significant role of an amyloid clearance mecha-nism. Bull Math Biol 73, 230-247.

[968] Comery TA, Martone RL, Aschmies S, Atchison KP,Diamantidis G, Gong X, Zhou H, Kreft AF, Panga-los MN, Sonnenberg-Reines J, Jacobsen JS, MarquisKL (2005) Acute gamma-secretase inhibition improvescontextual fear conditioning in the Tg2576 mouse modelof Alzheimer’s disease. J Neurosci 25, 8898-8902.

[969] Kreft A, Harrison B, Aschmies S, Atchison K, CasebierD, Cole DC, Diamantidis G, Ellingboe J, Hauze D, HuY, Huryn D, Jin M, Kubrak D, Lu P, Lundquist J, MannC, Martone R, Moore W, Oganesian A, Porte A, RiddellDR, Sonnenberg-Reines J, Stock JR, Sun SC, Wagner E,Woller K, Xu Z, Zhou H, Steven JJ (2008) Discovery of anovel series of Notch-sparing gamma-secretase inhibitors.Bioorg Med Chem Lett 18, 4232-4236.

[970] Mayer SC, Kreft AF, Harrison B, bou-Gharbia M, AntaneM, Aschmies S, Atchison K, Chlenov M, Cole DC, Com-ery T, Diamantidis G, Ellingboe J, Fan K, Galante R,Gonzales C, Ho DM, Hoke ME, Hu Y, Huryn D, Jain U, JinM, Kremer K, Kubrak D, Lin M, Lu P, Magolda R, MartoneR, Moore W, Oganesian A, Pangalos MN, Porte A, Rein-hart P, Resnick L, Riddell DR, Sonnenberg-Reines J, StockJR, Sun SC, Wagner E, Wang T, Woller K, Xu Z, ZaleskaMM, Zeldis J, Zhang M, Zhou H, Jacobsen JS (2008) Dis-covery of begacestat, a Notch-1-sparing gamma-secretaseinhibitor for the treatment of Alzheimer’s disease. J MedChem 51, 7348-7351.

[971] Cole DC, Stock JR, Kreft AF, Antane M, AschmiesSH, Atchison KP, Casebier DS, Comery TA, Diaman-tidis G, Ellingboe JW, Harrison BL, Hu Y, Jin M,Kubrak DM, Lu P, Mann CW, Martone RL, MooreWJ, Oganesian A, Riddell DR, Sonnenberg-Reines J,Sun SC, Wagner E, Wang Z, Woller KR, Xu Z,Zhou H, Jacobsen JS (2009) (S)-N-(5-Chlorothiophene-2-sulfonyl)-beta,beta-diethylalaninol a Notch-1-sparinggamma-secretase inhibitor. Bioorg Med Chem Lett 19,926-929.

[972] Martone RL, Zhou H, Atchison K, Comery T, Xu JZ,Huang X, Gong X, Jin M, Kreft A, Harrison B, Mayer SC,Aschmies S, Gonzales C, Zaleska MM, Riddell DR, Wag-ner E, Lu P, Sun SC, Sonnenberg-Reines J, Oganesian A,Adkins K, Leach MW, Clarke DW, Huryn D, bou-GharbiaM, Magolda R, Bard J, Frick G, Raje S, Forlow SB, BallietC, Burczynski ME, Reinhart PH, Wan HI, Pangalos MN,Jacobsen JS (2009) Begacestat (GSI-953): A novel, selec-tive thiophene sulfonamide inhibitor of amyloid precursorprotein gamma-secretase for the treatment of Alzheimer’sdisease. J Pharmacol Exp Ther 331, 598-608.

[973] Pu J, Kreft AF, Aschmies SH, Atchison KP, Berkowitz J,Caggiano TJ, Chlenov M, Diamantidis G, Harrison BL, HuY, Huryn D, Steven JJ, Jin M, Lipinski K, Lu P, MartoneRL, Morris K, Sonnenberg-Reines J, Riddell DR, Sabalski

Page 86: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

632 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

J, Sun SC, Wagner E, Wang Y, Xu Z, Zhou H, ResnickL (2009) Synthesis and structure-activity relationship of anovel series of heterocyclic sulfonamide gamma-secretaseinhibitors. Bioorg Med Chem 17, 4708-4717.

[974] Hopkins CR (2012) ACS Chemical Neurosciencemolecule spotlight on Begacestat (GSI-953). ACS ChemNeurosci 3, 3-4.

[975] Anderson JJ, Holtz G, Baskin PP, Turner M, Rowe B,Wang B, Kounnas MZ, Lamb BT, Barten D, Felsen-stein K, McDonald I, Srinivasan K, Munoz B, WagnerSL (2005) Reductions in beta-amyloid concentrations invivo by the gamma-secretase inhibitors BMS-289948 andBMS-299897. Biochem Pharmacol 69, 689-698.

[976] Barten DM, Guss VL, Corsa JA, Loo A, Hansel SB, ZhengM, Munoz B, Srinivasan K, Wang B, Robertson BJ, Pol-son CT, Wang J, Roberts SB, Hendrick JP, Anderson JJ,Loy JK, Denton R, Verdoorn TA, Smith DW, FelsensteinKM (2005) Dynamics of {beta}-amyloid reductions inbrain, cerebrospinal fluid, and plasma of {beta}-amyloidprecursor protein transgenic mice treated with a {gamma}-secretase inhibitor. J Pharmacol Exp Ther 312, 635-643.

[977] Zhang D, Hanson R, Roongta V, Dischino DD, Gao Q,Sloan CP, Polson C, Keavy D, Zheng M, Mitroka J, YeolaS (2006) In vitro and in vivo metabolism of a gamma-secretase inhibitor BMS-299897 and generation of activemetabolites in milligram quantities with a microbial biore-actor. Curr Drug Metab 7, 883-896.

[978] Zheng M, Wang J, Lubinski J, Flint OP, Krishna R, Yao M,Pursley JM, Thakur A, Boulton DW, Santone KS, BartenDM, Anderson JJ, Felsenstein KM, Hansel SB (2009)Studies on the pharmacokinetics and metabolism of agamma-secretase inhibitor BMS-299897, and exploratoryinvestigation of CYP enzyme induction. Xenobiotica 39,544-555.

[979] Goldstein ME, Cao Y, Fiedler T, Toyn J, Iben L, BartenDM, Pierdomenico M, Corsa J, Prasad CV, Olson RE, LiYW, Zaczek R, Albright CF (2007) Ex vivo occupancyof gamma-secretase inhibitors correlates with brain beta-amyloid peptide reduction in Tg2576 mice. J PharmacolExp Ther 323, 102-108.

[980] Quinn K, Gullapalli RP, Merisko-Liversidge E, Gold-bach E, Wong A, Liversidge GG, Hoffman W, SauerJM, Bullock J, Tonn G (2012) A formulation strategyfor gamma secretase inhibitor ELND006, a BCS class IIcompound: Development of a nanosuspension formula-tion with improved oral bioavailability and reduced foodeffects in dogs. J Pharm Sci 101, 1462-1474.

[981] Quinn K, Gullapalli RP, Merisko-Liversidge E, Gold-bach E, Wong A, Liversidge GG, Hoffman W, SauerJM, Bullock J, Tonn G (2012) A formulation strategyfor gamma secretase inhibitor ELND006, a BCS class IIcompound: Development of a nanosuspension formula-tion with improved oral bioavailability and reduced foodeffects in dogs. J Pharm Sci 101, 1462-1474.

[982] Plentz R, Park JS, Rhim AD, Abravanel D, Hezel AF,Sharma SV, Gurumurthy S, Deshpande V, Kenific C,Settleman J, Majumder PK, Stanger BZ, Bardeesy N(2009) Inhibition of gamma-secretase activity inhibitstumor progression in a mouse model of pancreatic ductaladenocarcinoma. Gastroenterology 136, 1741-1749.

[983] Bai F, Tagen M, Colotta C, Miller L, Fouladi M, StewartCF (2010) Determination of the gamma-secretase inhibitorMK-0752 in human plasma by online extraction and elec-trospray tandem mass spectrometry (HTLC-ESI-MS/MS).

J Chromatogr B Analyt Technol Biomed Life Sci 878, 2348-2352.

[984] Lanz TA, Wood KM, Richter KE, Nolan CE, Becker SL,Pozdnyakov N, Martin BA, Du P, Oborski CE, Wood DE,Brown TM, Finley JE, Sokolowski SA, Hicks CD, Coff-man KJ, Geoghegan KF, Brodney MA, Liston D, Tate B(2010) Pharmacodynamics and pharmacokinetics of thegamma-secretase inhibitor PF-3084014. J Pharmacol ExpTher 334, 269-277.

[985] Wei P, Walls M, Qiu M, Ding R, Denlinger RH, Wong A,Tsaparikos K, Jani JP, Hosea N, Sands M, Randolph S,Smeal T (2010) Evaluation of selective gamma-secretaseinhibitor PF-03084014 for its antitumor efficacy and gas-trointestinal safety to guide optimal clinical trial design.Mol Cancer Ther 9, 1618-1628.

[986] Brodney MA, Auperin DD, Becker SL, Bronk BS, BrownTM, Coffman KJ, Finley JE, Hicks CD, Karmilowicz MJ,Lanz TA, Liston D, Liu X, Martin BA, Nelson RB, NolanCE, Oborski CE, Parker CP, Richter KE, Pozdnyakov N,Sahagan BG, Schachter JB, Sokolowski SA, Tate B, VanDeusen JW, Wood DE, Wood KM (2011) Diamide amino-imidazoles: A novel series of gamma-secretase inhibitorsfor the treatment of Alzheimer’s disease. Bioorg MedChem Lett 21, 2631-2636.

[987] Samon JB, Castillo-Martin M, Hadler M, mbesi-ImpiobatoA, Paietta E, Racevskis J, Wiernik PH, Rowe JM,Jakubczak J, Randolph S, Cordon-Cardo C, FerrandoAA (2012) Preclinical Analysis of the gamma-secretaseinhibitor PF-03084014 in combination with glucocorti-coids in T-cell acute lymphoblastic leukemia. Mol CancerTher 11, 1565-1575.

[988] Peters JU, Galley G, Jacobsen H, Czech C, vid-PiersonP, Kitas EA, Ozmen L (2007) Novel orally active,dibenzazepinone-based gamma-secretase inhibitors.Bioorg Med Chem Lett 17, 5918-5923.

[989] Kitas EA, Galley G, Jakob-Roetne R, Flohr A, WostlW, Mauser H, Alker AM, Czech C, Ozmen L, vid-Pierson P, Reinhardt D, Jacobsen H (2008) Substituted2-oxo-azepane derivatives are potent, orally active gamma-secretase inhibitors. Bioorg Med Chem Lett 18, 304-308.

[990] Hoffmann-Emery F, Jakob-Roetne R, Flohr A, Bliss F,Reents R (2009) Improved synthesis of (S)-7-amino-5H,7H-dibenzo[b,d]azepin-6-one, a bulding block forg-secretase inhibitors. Tetrahedron Lett 50, 6380-6382.

[991] Luistro L, He W, Smith M, Packman K, Vilenchik M, Car-vajal D, Roberts J, Cai J, Berkofsky-Fessler W, Hilton H,Linn M, Flohr A, Jakob-Rotne R, Jacobsen H, Glenn K,Heimbrook D, Boylan JF (2009) Preclinical profile of apotent gamma-secretase inhibitor targeting notch signal-ing with in vivo efficacy and pharmacodynamic properties.Cancer Res 69, 7672-7680.

[992] He W, Luistro L, Carvajal D, Smith M, Nevins T, Yin X,Cai J, Higgins B, Kolinsky K, Rizzo C, Packman K, Heim-brook D, Boylan JF (2011) High tumor levels of IL6 andIL8 abrogate preclinical efficacy of the gamma-secretaseinhibitor, RO4929097. Mol Oncol 5, 292-301.

[993] Wu J, Wiegand R, LoRusso P, Li J (2011) Validationand implementation of a liquid chromatography/tandemmass spectrometry assay for quantitation of the total andunbound RO4929097, a gamma-secretase inhibitor target-ing Notch signaling, in human plasma. J Chromatogr BAnalyt Technol Biomed Life Sci 879, 1537-1543.

[994] Wong GT, Manfra D, Poulet FM, Zhang Q, Josien H, BaraT, Engstrom L, Pinzon-Ortiz M, Fine JS, Lee HJ, ZhangL, Higgins GA, Parker EM (2004) Chronic treatment with

Page 87: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 633

the gamma-secretase inhibitor LY-411,575 inhibits beta-amyloid peptide production and alters lymphopoiesis andintestinal cell differentiation. J Biol Chem 279, 12876-12882.

[995] Best JD, Jay MT, Otu F, Ma J, Nadin A, EllisS, Lewis HD, Pattison C, Reilly M, Harrison T,Shearman MS, Williamson TL, Atack JR (2005)Quantitative measurement of changes in amyloid-beta(40) in the rat brain and cerebrospinal fluidfollowing treatment with the gamma-secretaseinhibitor LY-411575[N2-[(2S)-2-(3,5-difluorophenyl)-2-hydroxyethanoyl]-N1-[(7S)-5-methyl-6-ox o-6,7-dihydro-5H-dibenzo[b,d]azepin-7-yl]-L-alaninamide].J Pharmacol Exp Ther 313, 902-908.

[996] Siemers E, Skinner M, Dean RA, Gonzales C, SatterwhiteJ, Farlow M, Ness D, May PC (2005) Safety, tolerability,and changes in amyloid beta concentrations after adminis-tration of a gamma-secretase inhibitor in volunteers. ClinNeuropharmacol 28, 126-132.

[997] Siemers ER, Quinn JF, Kaye J, Farlow MR, PorsteinssonA, Tariot P, Zoulnouni P, Galvin JE, Holtzman DM, Knop-man DS, Satterwhite J, Gonzales C, Dean RA, May PC(2006) Effects of a gamma-secretase inhibitor in a random-ized study of patients with Alzheimer disease. Neurology66, 602-604.

[998] Siemers ER, Dean RA, Friedrich S, Ferguson-Sells L,Gonzales C, Farlow MR, May PC (2007) Safety, tol-erability, and effects on plasma and cerebrospinal fluidamyloid-beta after inhibition of gamma-secretase. ClinNeuropharmacol 30, 317-325.

[999] Hyde LA, McHugh NA, Chen J, Zhang Q, Manfra D,Nomeir AA, Josien H, Bara T, Clader JW, Zhang L, ParkerEM, Higgins GA (2006) Studies to investigate the in vivotherapeutic window of the gamma-secretase inhibitorN2-[(2S)-2-(3,5-difluorophenyl)-2-hydroxyethanoyl]-N1-[(7S)-5-methyl-6-oxo -6,7-dihydro-5H-dibenzo[b,d]azepin-7-yl]-L-alaninamide (LY411,575) in the CRND8mouse. J Pharmacol Exp Ther 319, 1133-1143.

[1000] Fauq AH, Simpson K, Maharvi GM, Golde T, Das P (2007)A multigram chemical synthesis of the gamma-secretaseinhibitor LY411575 and its diastereoisomers. Bioorg MedChem Lett 17, 6392-6395.

[1001] Fleisher AS, Raman R, Siemers ER, Becerra L, Clark CM,Dean RA, Farlow MR, Galvin JE, Peskind ER, QuinnJF, Sherzai A, Sowell BB, Aisen PS, Thal LJ (2008)Phase 2 safety trial targeting amyloid beta production witha gamma-secretase inhibitor in Alzheimer disease. ArchNeurol 65, 1031-1038.

[1002] Bateman RJ, Siemers ER, Mawuenyega KG, Wen G,Browning KR, Sigurdson WC, Yarasheski KE, FriedrichSW, DeMattos RB, May PC, Paul SM, Holtzman DM(2009) A gamma-secretase inhibitor decreases amyloid-beta production in the central nervous system. Ann Neurol66, 48-54.

[1003] Henley DB, May PC, Dean RA, Siemers ER (2009)Development of semagacestat (LY450139), a func-tional gamma-secretase inhibitor, for the treatment ofAlzheimer’s disease. Expert Opin Pharmacother 10, 1657-1664.

[1004] Imbimbo BP, Peretto I (2009) Semagacestat, a gamma-secretase inhibitor for the potential treatment ofAlzheimer’s disease. Curr Opin Investig Drugs 10, 721-730.

[1005] Cummings J (2010) What can be inferred from theinterruption of the semagacestat trial for treatment

of Alzheimer’s disease? Biol Psychiatry 68, 876-878.

[1006] Portelius E, Dean RA, Gustavsson MK, Andreasson U,Zetterberg H, Siemers E, Blennow K (2010) A novel Abetaisoform pattern in CSF reflects gamma-secretase inhibitionin Alzheimer disease. Alzheimers Res Ther 2, 7.

[1007] Yi P, Hadden C, Kulanthaivel P, Calvert N, Annes W,Brown T, Barbuch RJ, Chaudhary A, yan-Oshodi MA,Ring BJ (2010) Disposition and metabolism of semagace-stat, a gamma-secretase inhibitor, in humans. Drug MetabDispos 38, 554-565.

[1008] Schor NF (2011) What the halted phase III gamma-secretase inhibitor trial may (or may not) be telling us.Ann Neurol 69, 237-239.

[1009] Willis BA, Zhang W, yan-Oshodi M, Lowe SL, AnnesWF, Sirois PJ, Friedrich S, de la PA (2012) Semagacestatpharmacokinetics are not significantly affected by formu-lation, food, or time of dosing in healthy participants.J Clin Pharmacol 52, 904-913.

[1010] Zhang W, Ayan-Oshodi M, Willis BA, Annes W, Hall SD,Chiesa J, Seger M (2012) QT effect of semagacestat at ther-apeutic and supratherapeutic doses. Int J Clin PharmacolTher 50, 290-299.

[1011] Wolfe MS (2006) The gamma-secretase complex:Membrane-embedded proteolytic ensemble. Biochemistry45, 7931-7939.

[1012] Aisen PS (2005) The development of anti-amyloid ther-apy for Alzheimer’s disease: From secretase modulatorsto polymerisation inhibitors. CNS Drugs 19, 989-996.

[1013] Czirr E, Weggen S (2006) Gamma-secretase modulationwith Abeta42-lowering nonsteroidal anti-inflammatorydrugs and derived compounds. Neurodegener Dis 3, 298-304.

[1014] Pissarnitski D (2007) Advances in gamma-secretase mod-ulation. Curr Opin Drug Discov Devel 10, 392-402.

[1015] Wolfe MS (2007) gamma-Secretase modulators. CurrAlzheimer Res 4, 571-573.

[1016] Beher D (2008) Gamma-secretase modulation and itspromise for Alzheimer’s disease: A rationale for drug dis-covery. Curr Top Med Chem 8, 34-37.

[1017] Evin G (2008) Gamma-secretase modulators: Hopes andsetbacks for the future of Alzheimer’s treatment. ExpertRev Neurother 8, 1611-1613.

[1018] Peretto I, La Porta E (2008) Gamma-secretase modula-tion and its promise for Alzheimer’s disease: A medicinalchemistry perspective. Curr Top Med Chem 8, 38-46.

[1019] Yang T, Arslanova D, Gu Y, Augelli-Szafran C, Xia W(2008) Quantification of gamma-secretase modulation dif-ferentiates inhibitor compound selectivity between twosubstrates Notch and amyloid precursor protein. Mol Brain1, 15.

[1020] Bulic B, Ness J, Hahn S, Rennhack A, Jumpertz T, WeggenS (2011) Chemical biology, molecular mechanism andclinical perspective of gamma-secretase modulators inAlzheimer’s disease. Curr Neuropharmacol 9, 598-622.

[1021] Pettersson M, Kauffman GW, am Ende CW, Patel NC, StiffC, Tran TP, Johnson DS (2011) Novel gamma-secretasemodulators: A review of patents from 2008 to 2010. ExpertOpin Ther Pat 21, 205-226.

[1022] Jumpertz T, Rennhack A, Ness J, Baches S, Pietrzik CU,Bulic B, Weggen S (2012) Presenilin is the molecular targetof acidic gamma-secretase modulators in living cells. PLoSOne 7, e30484.

[1023] Wagner SL, Tanzi RE, Mobley WC, Galasko D (2012)Potential use of gamma-secretase modulators in the

Page 88: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

634 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

treatment of Alzheimer disease. Arch Neurol, doi:10.1001/archneurol.2012.540

[1024] Xia W, Wong ST, Hanlon E, Morin P (2012) gamma-secretase modulator in Alzheimer’s disease: Shifting theend. J Alzheimers Dis 31, 685-696.

[1025] Eriksen JL, Sagi SA, Smith TE, Weggen S, Das P, McLen-don DC, Ozols VV, Jessing KW, Zavitz KH, Koo EH,Golde TE (2003) NSAIDs and enantiomers of flurbiprofentarget gamma-secretase and lower Abeta 42 in vivo. J ClinInvest 112, 440-449.

[1026] Weggen S, Eriksen JL, Sagi SA, Pietrzik CU, Ozols V,Fauq A, Golde TE, Koo EH (2003) Evidence that non-steroidal anti-inflammatory drugs decrease amyloid beta42 production by direct modulation of gamma-secretaseactivity. J Biol Chem 278, 31831-31837.

[1027] Peretto I, Radaelli S, Parini C, Zandi M, Raveglia LF,Dondio G, Fontanella L, Misiano P, Bigogno C, Rizzi A,Riccardi B, Biscaioli M, Marchetti S, Puccini P, CatinellaS, Rondelli I, Cenacchi V, Bolzoni PT, Caruso P, VillettiG, Facchinetti F, Del GE, Moretto N, Imbimbo BP (2005)Synthesis and biological activity of flurbiprofen analoguesas selective inhibitors of beta-amyloid(1)(-)(42) secretion.J Med Chem 48, 5705-5720.

[1028] Kukar T, Prescott S, Eriksen JL, Holloway V, Murphy MP,Koo EH, Golde TE, Nicolle MM (2007) Chronic adminis-tration of R-flurbiprofen attenuates learning impairmentsin transgenic amyloid precursor protein mice. BMC Neu-rosci 8, 54.

[1029] Zhao X, Rebeck GW, Hoe HS, Andrews PM (2008) Taren-flurbil protection from cytotoxicity is associated with anupregulation of neurotrophins. J Alzheimers Dis 15, 397-407.

[1030] Wilcock GK, Black SE, Hendrix SB, Zavitz KH, SwabbEA, Laughlin MA (2008) Efficacy and safety of tarenflur-bil in mild to moderate Alzheimer’s disease: A randomisedphase II trial. Lancet Neurol 7, 483-493.

[1031] Green RC, Schneider LS, Amato DA, Beelen AP, WilcockG, Swabb EA, Zavitz KH (2009) Effect of tarenflurbil oncognitive decline and activities of daily living in patientswith mild Alzheimer disease: A randomized controlledtrial. JAMA 302, 2557-2564.

[1032] Imbimbo BP (2009) Why did tarenflurbil fail inAlzheimer’s disease? J Alzheimers Dis 17, 757-760.

[1033] Sano M (2010) Tarenflurbil: Mechanisms and myths. ArchNeurol 67, 750-752.

[1034] Vellas B (2010) Tarenflurbil for Alzheimer’s disease:A “shot on goal” that missed. Lancet Neurol 9, 235-237.

[1035] Imbimbo BP, Del GE, Cenacchi V, Volta R, Villetti G,Facchinetti F, Riccardi B, Puccini P, Moretto N, Grassi F,Ottonello S, Leon A (2007) In vitro and in vivo profiling ofCHF5022 and CHF5074 Two beta-amyloid1-42 loweringagents. Pharmacol Res 55, 318-328.

[1036] Imbimbo BP, Del GE, Colavito D, D’Arrigo A, Dalle CM,Villetti G, Facchinetti F, Volta R, Pietrini V, Baroc MF,Serneels L, De SB, Leon A (2007) 1-(3′,4′-Dichloro-2-fluoro[1,1′-biphenyl]-4-yl)-cyclopropanecarboxylic acid(CHF5074), a novel gamma-secretase modulator, reducesbrain beta-amyloid pathology in a transgenic mouse modelof Alzheimer’s disease without causing peripheral toxicity.J Pharmacol Exp Ther 323, 822-830.

[1037] Imbimbo BP, Hutter-Paier B, Villetti G, Facchinetti F,Cenacchi V, Volta R, Lanzillotta A, Pizzi M, WindischM (2009) CHF5074, a novel gamma-secretase modula-tor, attenuates brain beta-amyloid pathology and learning

deficit in a mouse model of Alzheimer’s disease. Br JPharmacol 156, 982-993.

[1038] Imbimbo BP, Giardino L, Sivilia S, Giuliani A, GusciglioM, Pietrini V, Del GE, D’Arrigo A, Leon A, Villetti G,Calza L (2010) CHF5074, a novel gamma-secretase mod-ulator, restores hippocampal neurogenesis potential andreverses contextual memory deficit in a transgenic mousemodel of Alzheimer’s disease. J Alzheimers Dis 20, 159-173.

[1039] Lanzillotta A, Sarnico I, Benarese M, Branca C, BaigueraC, Hutter-Paier B, Windisch M, Spano P, Imbimbo BP,Pizzi M (2011) The gamma-secretase modulator CHF5074reduces the accumulation of native hyperphosphorylatedtau in a transgenic mouse model of Alzheimer’s disease.J Mol Neurosci 45, 22-31.

[1040] Balducci C, Mehdawy B, Mare L, Giuliani A, LorenziniL, Sivilia S, Giardino L, Calza L, Lanzillotta A, Sarnico I,Pizzi M, Usiello A, Viscomi AR, Ottonello S, Villetti G,Imbimbo BP, Nistico G, Forloni G, Nistico R (2011) Thegamma-secretase modulator CHF5074 restores memoryand hippocampal synaptic plasticity in plaque-free Tg2576mice. J Alzheimers Dis 24, 799-816.

[1041] Poli G, Corda E, Lucchini B, Puricelli M, Martino PA,Dall’ara P, Villetti G, Bareggi SR, Corona C, CostassaEV, Gazzuola P, Iulini B, Mazza M, Acutis P, MantegazzaP, Casalone C, Imbimbo BP (2012) Therapeutic effect ofCHF5074, a new gamma-secretase modulator, in a mousemodel of scrapie. Prion 6, 62-72.

[1042] Tian G, Sobotka-Briner CD, Zysk J, Liu X, Birr C,Sylvester MA, Edwards PD, Scott CD, Greenberg BD(2002) Linear non-competitive inhibition of solubilizedhuman gamma-secretase by pepstatin A methylester,L685458, sulfonamides, and benzodiazepines. J BiolChem 277, 31499-31505.

[1043] Tian G, Ghanekar SV, Aharony D, Shenvi AB, Jacobs RT,Liu X, Greenberg BD (2003) The mechanism of gamma-secretase: Multiple inhibitor binding sites for transitionstate analogs and small molecule inhibitors. J Biol Chem278, 28968-28975.

[1044] Borgegard T, Jureus A, Olsson F, Rosqvist S, SabirshA, Rotticci D, Paulsen K, Klintenberg R, Yan H, Wald-man M, Stromberg K, Nord J, Johansson J, Regner A,Parpal S, Malinowsky D, Radesater AC, Li T, Singh R,Eriksson H, Lundkvist J (2012) First and second gen-eration gamma-secretase modulators (GSMs) modulateamyloid-beta (Abeta) peptide production through differentmechanisms. J Biol Chem 287, 11810-11819.

[1045] Sehgelmeble F, Janson J, Ray C, Rosqvist S, Gustavsson S,Nilsson LI, Minidis A, Holenz J, Rotticci D, Lundkvist J,Arvidsson PI (2012) Sulfonimidamides as sulfonamidesbioisosteres: Rational evaluation through synthetic, invitro, and in vivo studies with gamma-secretase inhibitors.ChemMedChem 7, 396-399.

[1046] Wanngren J, Ottervald J, Parpal S, Portelius E, StrombergK, Borgegard T, Klintenberg R, Jureus A, BlomqvistJ, Blennow K, Zetterberg H, Lundkvist J, RosqvistS, Karlstrom H (2012) Second generation gamma-secretase modulators exhibit different modulation of Notchbeta and Abeta production. J Biol Chem 287, 32640-32650.

[1047] Peng HR, Talreja T, Xin ZL, Cuergvo JH, Kumaravek G,Humora MJ, Xu L, Rohde E, Gan L, Jung MY, SahckettMN, Chollate S, Dunah AW, Snodgrass-belt PA, ArnoldHM, Taveras AG, Rhodes KJ, Scannevin RH (2011)Discovery of BIIB042, a potent, selective, and orally

Page 89: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 635

bioavailable gamma-secretase modulator. ACS Med ChemLett 2, 786-791.

[1048] Xin Z, Peng H, Zhang A, Talreja T, Kumaravel G, XuL, Rohde E, Jung MY, Shackett MN, Kocisko D, Chol-late S, Dunah AW, Snodgrass-Belt PA, Arnold HM,Taveras AG, Rhodes KJ, Scannevin RH (2011) Discoveryof 4-aminomethylphenylacetic acids as gamma-secretasemodulators via a scaffold design approach. Bioorg MedChem Lett 21, 7277-7280.

[1049] Garcia Y, Hannam JC, Harrison T, Hamblett CL HJKJ,Madin A, Ridgill MP, Seward E (2007) Piperidines andrelated compounds for treatment of Alzheimer’s disease.WO/2007-125364. Prior. 26.04.2006 for Merck Sharp &Dohme Ltd. UK.

[1050] Mitani Y, Yarimizu J, Saita K, Uchino H, Akashiba H,Shitaka Y, Ni K, Matsuoka N (2012) Differential effectsbetween gamma-secretase inhibitors and modulatorson cognitive function in amyloid precursor protein-transgenic and nontransgenic mice. J Neurosci 32, 2037-2050.

[1051] Hall A, Elliott RL, Giblin GM, Hussain I, Musgrave J,Naylor A, Sasse R, Smith B (2010) Piperidine-derivedgamma-secretase modulators. Bioorg Med Chem Lett 20,1306-1311.

[1052] Hussain I, Harrison DC, Hawkins J, Chapman T, Mar-shall I, Facci L, Ahmed S, Brackenborough K, SkaperSD, Mead TL, Smith BB, Giblin GM, Hall A, Gonza-lez MI, Richardson JC (2011) TASTPM mice expressingamyloid precursor protein and presenilin-1 mutant trans-genes are sensitive to gamma-secretase modulation andamyloid-beta lowering by GSM-10h. Neurodegener Dis8, 15-24.

[1053] Hawkins J, Harrison DC, Ahmed S, Davis RP, ChapmanT, Marshall I, Smith B, Mead TL, Medhurst A, GiblinGM, Hall A, Gonzalez MI, Richardson J, Hussain I (2011)Dynamics of Abeta42 reduction in plasma, CSF and brainof rats treated with the gamma-secretase modulator, GSM-10h. Neurodegener Dis 8, 455-464.

[1054] Li T, Huang Y, Jin S, Ye L, Rong N, Yang X, Ding Y, ChengZ, Zhang J, Wan Z, Harrison DC, Hussain I, Hall A, LeeDH, Lau LF, Matsuoka Y (2012) Gamma-secretase mod-ulators do not induce Abeta-rebound and accumulation ofbeta-C-terminal fragment. J Neurochem 121, 277-286.

[1055] Wan Z, Hall A, Jin Y, Xiang JN, Yang E, Eatherton A,Smith B, Yang G, Yu H, Wang J, Ye L, Lau LF, YangT, Mitchell W, Cai W, Zhang X, Sang Y, Wang Y, TongZ, Cheng Z, Hussain I, Elliott JD, Matsuoka Y (2011)Pyridazine-derived gamma-secretase modulators. BioorgMed Chem Lett 21, 4016-4019.

[1056] Wan Z, Hall A, Sang Y, Xiang JN, Yang E, Smith B, Har-rison DC, Yang G, Yu H, Price HS, Wang J, Hawkins J,Lau LF, Johnson MR, Li T, Zhao W, Mitchell WL, Su X,Zhang X, Zhou Y, Jin Y, Tong Z, Cheng Z, Hussain I, ElliottJD, Matsuoka Y (2011) Pyridine-derived gamma-secretasemodulators. Bioorg Med Chem Lett 21, 4832-4835.

[1057] Bischoff F, Berthelot D, De Cleyn M, Macdonald G, MinneG, Oehlrich D, Pieters S, Surkyn M, Trabanco AA, Tre-sadern G, Van Brandt S, Velter I, Zaja M, Borghys H,Masungi C, Mercken M, Gijsen HJ (2012) Design andsynthesis of a novel series of bicyclic heterocycles aspotent gamma-secretase modulators. J Med Chem, doi:10.1021/jm201710f [Epubl ahead of print].

[1058] Caldwell JP, Bennett CE, McCracken TM, Mazzola RD,Bara T, Buevich A, Burnett DA, Chu I, Cohen-Williams M,Josein H, Hyde L, Lee J, McKittrick B, Song L, Terracina

G, Voigt J, Zhang L, Zhu Z (2010) Iminoheterocycles asgamma-secretase modulators. Bioorg Med Chem Lett 20,5380-5384.

[1059] Rivkin A, Ahearn SP, Chichetti SM, Kim YR, Li C, Rose-nau A, Kattar SD, Jung J, Shah S, Hughes BL, CrispinoJL, Middleton RE, Szewczak AA, Munoz B, ShearmanMS (2010) Piperazinyl pyrimidine derivatives as potentgamma-secretase modulators. Bioorg Med Chem Lett 20,1269-1271.

[1060] Rivkin A, Ahearn SP, Chichetti SM, Hamblett CL, Gar-cia Y, Martinez M, Hubbs JL, Reutershan MH, DanielsMH, Siliphaivanh P, Otte KM, Li C, Rosenau A, SurdiLM, Jung J, Hughes BL, Crispino JL, Nikov GN, Middle-ton RE, Moxham CM, Szewczak AA, Shah S, Moy LY,Kenific CM, Tanga F, Cruz JC, Andrade P, Angagaw MH,Shomer NH, Miller T, Munoz B, Shearman MS (2010)Purine derivatives as potent gamma-secretase modulators.Bioorg Med Chem Lett 20, 2279-2282.

[1061] Stanton MG, Hubbs J, Sloman D, Hamblett C, Andrade P,Angagaw M, Bi G, Black RM, Crispino J, Cruz JC, Fan E,Farris G, Hughes BL, Kenific CM, Middleton RE, NikovG, Sajonz P, Shah S, Shomer N, Szewczak AA, Tanga F,Tudge MT, Shearman M, Munoz B (2010) Fluorinatedpiperidine acetic acids as gamma-secretase modulators.Bioorg Med Chem Lett 20, 755-758.

[1062] Fischer C, Shah S, Hughes BL, Nikov GN, Crispino JL,Middleton RE, Szewczak AA, Munoz B, Shearman MS(2011) Quinazolinones as gamma-secretase modulators.Bioorg Med Chem Lett 21, 773-776.

[1063] Qin J, Dhondi P, Huang X, Mandal M, Zhao Z, PissarnitskiD, Zhou W, Aslanian R, Zhu Z, Greenlee W, Clader J,Zhang L, Cohen-Williams M, Jones N, Hyde L, PalaniA (2011) Discovery of fused 5,6-bicyclic heterocycles asgamma-secretase modulators. Bioorg Med Chem Lett 21,664-669.

[1064] Beher D, Clarke EE, Wrigley JD, Martin AC, NadinA, Churcher I, Shearman MS (2004) Selected non-steroidal anti-inflammatory drugs and their derivativestarget gamma-secretase at a novel site. Evidence foran allosteric mechanism. J Biol Chem 279, 43419-43426.

[1065] Fischer C, Zultanski SL, Zhou H, Methot JL, Brown WC,Mampreian DM, Schell AJ, Shah S, Nuthall H, Hughes BL,Smotrov N, Kenific CM, Cruz JC, Walker D, BouthilletteM, Nikov GN, Savage DF, Jeliazkova-Mecheva VV, DiazD, Szewczak AA, Bays N, Middleton RE, Munoz B, Shear-man MS (2011) Triazoles as gamma-secretase modulators.Bioorg Med Chem Lett 21, 4083-4087.

[1066] Fischer C, Zultanski SL, Zhou H, Methot JL, Shah S,Nuthall H, Hughes BL, Smotrov N, Hill A, SzewczakAA, Moxham CM, Bays N, Middleton RE, Munoz B,Shearman MS (2012) Triazoloamides as potent gamma-secretase modulators with reduced hERG liability. BioorgMed Chem Lett 22, 3140-3146.

[1067] Huang X, Aslanian R, Zhou W, Zhu X, Qin J, GreenleeW, Zhu Z, Zhang L, Hyde L, Chu I, Coen-Willaims M,Palani A (2010) The discovery of pyridone and pyridazoneheterocycles as gamma-secretase modulators. ACS MedChem Lett 1, 184-187.

[1068] Pettersson M, Johnson DS, Subramanyam C, Bales KR,am Ende CW, Fish BA, Green ME, Kauffman GW, LiraR, Mullins PB, Navaratnam T, Sakya SM, Stiff CM, TranTP, Vetelino BC, Xie L, Zhang L, Pustilnik LR, Wood KM,O’Donnell CJ (2012) Design and synthesis of dihydroben-zofuran amides as orally bioavailable, centrally active

Page 90: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

636 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

gamma-secretase modulators. Bioorg Med Chem Lett 22,2906-2911.

[1069] Kounnas MZ, Danks AM, Cheng S, Tyree C, AckermanE, Zhang X, Ahn K, Nguyen P, Comer D, Mao L, YuC, Pleynet D, Digregorio PJ, Velicelebi G, StaudermanKA, Comer WT, Mobley WC, Li YM, Sisodia SS, TanziRE, Wagner SL (2010) Modulation of gamma-secretasereduces beta-amyloid deposition in a transgenic mousemodel of Alzheimer’s disease. Neuron 67, 769-780.

[1070] Narlawar R, Perez Revuelta BI, Haass C, SteinerH, Schmidt B, Baumann K (2006) Scaffold of thecyclooxygenase-2 (COX-2) inhibitor carprofen providesAlzheimer gamma-secretase modulators. J Med Chem 49,7588-7591.

[1071] Narlawar R, Perez Revuelta BI, Baumann K, SchubenelR, Haass C, Steiner H, Schmidt B (2007) N-Substitutedcarbazolyloxyacetic acids modulate Alzheimer associatedgamma-secretase. Bioorg Med Chem Lett 17, 176-182.

[1072] Narlawar R, Baumann K, Czech C, Schmidt B (2007) Con-version of the LXR-agonist TO-901317–from inverse tonormal modulation of gamma-secretase by addition of acarboxylic acid and a lipophilic anchor. Bioorg Med ChemLett 17, 5428-5431.

[1073] Baumann S, Hottecke N, Schubenel R, Baumann K,Schmidt B (2009) NSAID-derived gamma-secretase mod-ulators. Part III: Membrane anchoring. Bioorg Med ChemLett 19, 6986-6990.

[1074] Zall A, Kieser D, Hottecke N, Naumann EC,Thomaszewski B, Schneider K, Steinbacher DT,Schubenel R, Masur S, Baumann K, Schmidt B (2011)NSAID-derived gamma-secretase modulation requiresan acidic moiety on the carbazole scaffold. Bioorg MedChem 19, 4903-4909.

[1075] Hahn S, Bruning T, Ness J, Czirr E, Baches S, GijsenH, Korth C, Pietrzik CU, Bulic B, Weggen S (2011)Presenilin-1 but not amyloid precursor protein mutationspresent in mouse models of Alzheimer’s disease attenuatethe response of cultured cells to gamma-secretase modula-tors regardless of their potency and structure. J Neurochem116, 385-395.

[1076] Kukar TL, Ladd TB, Robertson P, Pintchovski SA, MooreB, Bann MA, Ren Z, Jansen-West K, Malphrus K, EggertS, Maruyama H, Cottrell BA, Das P, Basi GS, Koo EH,Golde TE (2011) Lysine 624 of the amyloid precursor pro-tein (APP) is a critical determinant of amyloid beta peptidelength: Support for a sequential model of gamma-secretaseintramembrane proteolysis and regulation by the amyloidbeta precursor protein (APP) juxtamembrane region. J BiolChem 286, 39804-39812.

[1077] Kukar TL, Ladd TB, Bann MA, Fraering PC, NarlawarR, Maharvi GM, Healy B, Chapman R, Welzel AT, PriceRW, Moore B, Rangachari V, Cusack B, Eriksen J, Jansen-West K, Verbeeck C, Yager D, Eckman C, Ye W, SagiS, Cottrell BA, Torpey J, Rosenberry TL, Fauq A, WolfeMS, Schmidt B, Walsh DM, Koo EH, Golde TE (2008)Substrate-targeting gamma-secretase modulators. Nature453, 925-929.

[1078] Crump CJ, Johnson DS, Li YM (2011) Target of gamma-secretase modulators, presenilin marks the spot. EMBO J30, 4696-4698.

[1079] Crump CJ, Fish BA, Castro SV, Chau DM, Gertsik N, AhnK, Stiff C, Pozdnyakov N, Bales KR, Johnson DS, Li YM(2011) Piperidine acetic acid based gamma-secretase mod-ulators directly bind to Presenilin-1. ACS Chem Neurosci2, 705-710.

[1080] Crump CJ, am Ende CW, Ballard TE, Pozdnyakov N, Pet-tersson M, Chau DM, Bales KR, Li YM, Johnson DS(2012) Development of clickable active site-directed pho-toaffinity probes for gamma-secretase. Bioorg Med ChemLett 22, 2997-3000.

[1081] Ebke A, Luebbers T, Fukumori A, Shirotani K, Haass C,Baumann K, Steiner H (2011) Novel gamma-secretaseenzyme modulators directly target presenilin protein.J Biol Chem 286, 37181-37186.

[1082] Kretner B, Fukumori A, Gutsmiedl A, Page RM, LuebbersT, Galley G, Baumann K, Haass C, Steiner H (2011) Atten-uated Abeta42 responses to low potency gamma-secretasemodulators can be overcome for many pathogenic pre-senilin mutants by second-generation compounds. J BiolChem 286, 15240-15251.

[1083] Ohki Y, Higo T, Uemura K, Shimada N, Osawa S, Bere-zovska O, Yokoshima S, Fukuyama T, Tomita T, IwatsuboT (2011) Phenylpiperidine-type gamma-secretase modu-lators target the transmembrane domain 1 of presenilin 1.EMBO J 30, 4815-4824.

[1084] Zettl H, Ness J, Hahnke V, Beher D, Jumpertz T,Saric A, Baumann K, Pietrzik CU, Bulic B, Schnei-der G, Weggen S (2012) Discovery of gamma-secretasemodulators with a novel activity profile by text-based virtual screening. ACS Chem Biol 7, 1488-1495.

[1085] Uemura K, Lill CM, Li X, Peters JA, Ivanov A, FanZ, DeStrooper B, Bacskai BJ, Hyman BT, BerezovskaO (2009) Allosteric modulation of PS1/gamma-secretaseconformation correlates with amyloid beta(42/40) ratio.PLoS One 4, e7893.

[1086] Uemura K, Farner KC, Hashimoto T, Nasser-Ghodsi N,Wolfe MS, Koo EH, Hyman BT, Berezovska O (2010)Substrate docking to gamma-secretase allows access ofgamma-secretase modulators to an allosteric site. NatCommun 1, 130.

[1087] Uemura K, Farner KC, Nasser-Ghodsi N, Jones P, Bere-zovska O (2011) Reciprocal relationship between APPpositioning relative to the membrane and PS1 conforma-tion. Mol Neurodegener 6, 15.

[1088] Barrett PJ, Sanders CR, Kaufman SA, Michelsen K, JordanJB (2011) NSAID-based gamma-secretase modulators donot bind to the amyloid-beta polypeptide. Biochemistry 50,10328-10342.

[1089] Hieke M, Ness J, Steri R, Dittrich M, Greiner C,Werz O, Baumann K, Schubert-Zsilavecz M, WeggenS, Zettl H (2010) Design, synthesis, and biologicalevaluation of a novel class of gamma-secretase modula-tors with PPARgamma activity. J Med Chem 53, 4691-4700.

[1090] Hieke M, Ness J, Steri R, Greiner C, Werz O, Schubert-Zsilavecz M, Weggen S, Zettl H (2011) SAR studiesof acidic dual gamma-secretase/PPARgamma modulators.Bioorg Med Chem 19, 5372-5382.

[1091] Van Broeck B, Chen JM, Treton G, Desmidt M, HopfC, Ramsden N, Karran E, Mercken M, Rowley A (2011)Chronic treatment with a novel gamma-secretase modula-tor, JNJ-40418677, inhibits amyloid plaque formation ina mouse model of Alzheimer’s disease. Br J Pharmacol163, 375-389.

[1092] He G, Luo W, Li P, Remmers C, Netzer WJ, Hendrick J,Bettayeb K, Flajolet M, Gorelick F, Wennogle LP, Green-gard P (2010) Gamma-secretase activating protein is atherapeutic target for Alzheimer’s disease. Nature 467,95-98.

Page 91: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 637

[1093] St George-Hyslop P, Schmitt-Ulms G (2010) Alzheimer’sdisease: Selectively tuning gamma-secretase. Nature 467,36-37.

[1094] Satoh J, Tabunoki H, Ishida T, Saito Y, Arima K(2012) Immunohistochemical characterization of gamma-secretase activating protein expression in Alzheimer’sdisease brains. Neuropathol Appl Neurobiol 38, 132-141.

[1095] Deatherage CL, Hadziselimovic A, Sanders CR (2012)Purification and characterization of the human gamma-secretase activating protein. Biochemistry 51, 5153-5159.

[1096] Gulinello M, Chen F, Dobrenis K (2008) Early deficits inmotor coordination and cognitive dysfunction in a mousemodel of the neurodegenerative lysosomal storage disor-der, Sandhoff disease. Behav Brain Res 193, 315-319.

[1097] Chapman KE, Seckl JR (2008) 11beta-HSD1, inflam-mation, metabolic disease and age-related cognitive(dys)function. Neurochem Res 33, 624-636.

[1098] Maclullich AM, Ferguson KJ, Reid LM, Deary IJ,Starr JM, Wardlaw JM, Walker BR, Andrew R, SecklJR (2012) 11beta-hydroxysteroid dehydrogenase type 1,brain atrophy and cognitive decline. Neurobiol Aging 33,207-208.

[1099] Holmes MC, Carter RN, Noble J, Chitnis S, DutiaA, Paterson JM, Mullins JJ, Seckl JR, Yau JL (2010)11beta-hydroxysteroid dehydrogenase type 1 expressionis increased in the aged mouse hippocampus and parietalcortex and causes memory impairments. J Neurosci 30,6916-6920.

[1100] Yau JL, McNair KM, Noble J, Brownstein D, Hibberd C,Morton N, Mullins JJ, Morris RG, Cobb S, Seckl JR (2007)Enhanced hippocampal long-term potentiation and spa-tial learning in aged 11beta-hydroxysteroid dehydrogenasetype 1 knock-out mice. J Neurosci 27, 10487-10496.

[1101] Yau JL, Noble J, Seckl JR (2011) 11beta-hydroxysteroiddehydrogenase type 1 deficiency prevents memory deficitswith aging by switching from glucocorticoid receptorto mineralocorticoid receptor-mediated cognitive control.J Neurosci 31, 4188-4193.

[1102] Sooy K, Webster SP, Noble J, Binnie M, Walker BR,Seckl JR, Yau JL (2010) Partial deficiency or short-terminhibition of 11beta-hydroxysteroid dehydrogenase type 1improves cognitive function in aging mice. J Neurosci 30,13867-13872.

[1103] Mohler EG, Browman KE, Roderwald VA, Cronin EA,Markosyan S, Scott BR, Strakhova MI, Drescher KU,Hornberger W, Rohde JJ, Brune ME, Jacobson PB, RueterLE (2011) Acute inhibition of 11beta-hydroxysteroiddehydrogenase type-1 improves memory in rodent modelsof cognition. J Neurosci 31, 5406-5413.

[1104] Sandeep TC, Yau JL, MacLullich AM, Noble J, DearyIJ, Walker BR, Seckl JR (2004) 11Beta-hydroxysteroiddehydrogenase inhibition improves cognitive function inhealthy elderly men and type 2 diabetics. Proc Natl AcadSci U S A 101, 6734-6739.

[1105] Webster SP, Binnie M, McConnell KM, Sooy K, WardP, Greaney MF, Vinter A, Pallin TD, Dyke HJ, Gill MI,Warner I, Seckl JR, Walker BR (2010) Modulation of11beta-hydroxysteroid dehydrogenase type 1 activity by1,5-substituted 1H-tetrazoles. Bioorg Med Chem Lett 20,3265-3271.

[1106] Higuchi M, Iwata N, Matsuba Y, Takano J, Suemoto T,Maeda J, Ji B, Ono M, Staufenbiel M, Suhara T, Saido TC(2012) Mechanistic involvement of the calpain-calpastatinsystem in Alzheimer neuropathology. FASEB J 26, 1204-1217.

[1107] Battaglia F, Trinchese F, Liu S, Walter S, Nixon RA,Arancio O (2003) Calpain inhibitors, a treatment forAlzheimer’s disease: Position paper. J Mol Neurosci 20,357-362.

[1108] Getz GS (2012) Calpain inhibition as a potential treatmentof Alzheimer’s disease. Am J Pathol 181, 388-391.

[1109] Liang Z, Liu F, Grundke-Iqbal I, Iqbal K, Gong CX (2007)Down-regulation of cAMP-dependent protein kinase byover-activated calpain in Alzheimer disease brain. J Neu-rochem 103, 2462-2470.

[1110] Morales-Corraliza J, Berger JD, Mazzella MJ, Veeranna,Neubert TA, Ghiso J, Rao MV, Staufenbiel M, NixonRA, Mathews PM (2012) Calpastatin modulates APP pro-cessing in the brains of beta-amyloid depositing but notwild-type mice. Neurobiol Aging 33, 1125-1128.

[1111] Neuhof C, Fabiunke V, Deibele K, Speth M, MollerA, Lubisch W, Fritz H, Tillmanns H, Neuhof H (2004)Reduction of myocardial infarction by calpain inhibitorsA-705239 and A-705253 in isolated perfused rabbit hearts.Biol Chem 385, 1077-1082.

[1112] Neuhof C, Fabiunk V, Speth M, Moller A, Fritz F, Till-manns H, Neuhof H, Erdogan A (2008) Reduction ofmyocardial infarction by postischemic administration ofthe calpain inhibitor A-705253 in comparison to theNa(+)/H(+) exchange inhibitor Cariporide in isolated per-fused rabbit hearts. Biol Chem 389, 1505-1512.

[1113] Nangle MR, Cotter MA, Cameron NE (2006) The cal-pain inhibitor, A-705253, corrects penile nitrergic nervedysfunction in diabetic mice. Eur J Pharmacol 538, 148-153.

[1114] Nimmrich V, Szabo R, Nyakas C, Granic I, ReymannKG, Schroder UH, Gross G, Schoemaker H, Wicke K,Moller A, Luiten P (2008) Inhibition of calpain prevents N-methyl-D-aspartate-induced degeneration of the nucleusbasalis and associated behavioral dysfunction. J Pharma-col Exp Ther 327, 343-352.

[1115] Nimmrich V, Reymann KG, Strassburger M, Schoder UH,Gross G, Hahn A, Schoemaker H, Wicke K, Moller A(2010) Inhibition of calpain prevents NMDA-induced celldeath and beta-amyloid-induced synaptic dysfunction inhippocampal slice cultures. Br J Pharmacol 159, 1523-1531.

[1116] Sinjoanu RC, Kleinschmidt S, Bitner RS, Brioni JD,Moeller A, Ferreira A (2008) The novel calpain inhibitorA-705253 potently inhibits oligomeric beta-amyloid-induced dynamin 1 and tau cleavage in hippocampalneurons. Neurochem Int 53, 79-88.

[1117] Granic I, Nyakas C, Luiten PG, Eisel UL, Halmy LG,Gross G, Schoemaker H, Moller A, Nimmrich V (2010)Calpain inhibition prevents amyloid-beta-induced neu-rodegeneration and associated behavioral dysfunction inrats. Neuropharmacology 59, 334-342.

[1118] Nikkel AL, Martino B, Markosyan S, Brederson JD,Medeiros R, Moeller A, Bitner RS (2012) The novelcalpain inhibitor A-705253 prevents stress-induced tauhyperphosphorylation in vitro and in vivo. Neuropharma-cology 63, 606-612.

[1119] Medeiros R, Kitazawa M, Chabrier MA, Cheng D,Baglietto-Vargas D, Kling A, Moeller A, Green KN,Laferla FM (2012) Calpain inhibitor A-705253 miti-gates Alzheimer’s disease-like pathology and cognitivedecline in aged 3xTgAD mice. Am J Pathol 181, 616-625.

[1120] Saatman KE, Murai H, Bartus RT, Smith DH, Hayward NJ,Perri BR, McIntosh TK (1996) Calpain inhibitor AK295

Page 92: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

638 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

attenuates motor and cognitive deficits following experi-mental brain injury in the rat. Proc Natl Acad Sci U S A93, 3428-3433.

[1121] Oka T, Walkup RD, Tamada Y, Nakajima E, TochigiA, Shearer TR, Azuma M (2006) Amelioration ofretinal degeneration and proteolysis in acute ocu-lar hypertensive rats by calpain inhibitor ((1S)-1-((((1S)-1-benzyl-3-cyclopropylamino-2,3-di-oxopropyl)amino)carbonyl)-3-me thylbutyl)carbamic acid 5-methoxy-3-oxapentyl ester. Neuroscience 141, 2139-2145.

[1122] Shimazawa M, Suemori S, Inokuchi Y, MatsunagaN, Nakajima Y, Oka T, Yamamoto T, Hara H(2010) A novel calpain inhibitor, ((1S)-1-((((1S)-1-Benzyl-3-cyclopropylamino-2,3-di-oxopropyl)amino)carbonyl)-3-me thylbutyl)carbamic acid 5-methoxy-3-oxapentyl ester (SNJ-1945), reduces murine retinal celldeath in vitro and in vivo. J Pharmacol Exp Ther 332,380-387.

[1123] Koumura A, Nonaka Y, Hyakkoku K, Oka T, ShimazawaM, Hozumi I, Inuzuka T, Hara H (2008) A novel calpaininhibitor, ((1S)-1((((1S)-1-benzyl-3-cyclopropylamino-2,3-di-oxopropyl)amino)carbonyl)-3-met hylbutyl)carbamic acid 5-methoxy-3-oxapentyl ester, protectsneuronal cells from cerebral ischemia-induced damage inmice. Neuroscience 157, 309-318.

[1124] Shirasaki Y, Yamaguchi M, Miyashita H (2006) Retinalpenetration of calpain inhibitors in rats after oral adminis-tration. J Ocul Pharmacol Ther 22, 417-424.

[1125] Shirasaki Y, Nakamura M, Yamaguchi M, Miyashita H,Sakai O, Inoue J (2006) Exploration of orally availablecalpain inhibitors 2: Peptidyl hemiacetal derivatives. J MedChem 49, 3926-3932.

[1126] Shirasaki Y, Miyashita H, Yamaguchi M (2006) Explo-ration of orally available calpain inhibitors. Part 3:Dipeptidyl alpha-ketoamide derivatives containing pyri-dine moiety. Bioorg Med Chem 14, 5691-5698.

[1127] Shirasaki Y, Miyashita H, Yamaguchi M, Inoue J,Nakamura M (2005) Exploration of orally available cal-pain inhibitors: Peptidyl alpha-ketoamides containing anamphiphile at P3 site. Bioorg Med Chem 13, 4473-4484.

[1128] Shirasaki Y, Takahashi H, Yamaguchi M, Inoue J (2008)Molecular design to enhance the penetration into the retinavia ocular instillation. Bioorg Med Chem Lett 18, 5174-5177.

[1129] Ennes-Vidal V, Menna-Barreto RF, Santos AL, Bran-quinha MH, vila-Levy CM (2010) Effects of the calpaininhibitor MDL28170 on the clinically relevant forms ofTrypanosoma cruzi in vitro. J Antimicrob Chemother 65,1395-1398.

[1130] Ennes-Vidal V, Menna-Barreto RF, Santos AL, Bran-quinha MH, vila-Levy CM (2011) MDL28170, a calpaininhibitor, affects Trypanosoma cruzi metacyclogene-sis, ultrastructure and attachment to Rhodnius prolixusmidgut. PLoS One 6, e18371.

[1131] Bi SH, Jin ZX, Zhang JY, Chen T, Zhang SL, Yang Y, DuanWX, Yi DH, Zhou JJ, Ren J (2012) Calpain inhibitor MDL28170 protects against the Ca(2+) paradox in rat hearts.Clin Exp Pharmacol Physiol 39, 385-392.

[1132] Sun MK, Alkon DL (2002) Carbonic anhydrase gatingof attention: Memory therapy and enhancement. TrendsPharmacol Sci 23, 83-89.

[1133] Graham RK, Ehrnhoefer DE, Hayden MR (2011) Caspase-6 and neurodegeneration. Trends Neurosci 34, 646-656.

[1134] Rohn TT (2010) The role of caspases in Alzheimer’s dis-ease; potential novel therapeutic opportunities. Apoptosis15, 1403-1409.

[1135] Rohn TT, Head E (2009) Caspases as therapeutic targetsin Alzheimer’s disease: Is it time to “cut” to the chase? IntJ Clin Exp Pathol 2, 108-118.

[1136] Rohn TT (2008) Caspase-cleaved TAR DNA-bindingprotein-43 is a major pathological finding in Alzheimer’sdisease. Brain Res 1228, 189-198.

[1137] Rohn TT, Head E (2008) Caspase activation in Alzheimer’sdisease: Early to rise and late to bed. Rev Neurosci 19,383-393.

[1138] Castro RE, Santos MM, Gloria PM, Ribeiro CJ, FerreiraDM, Xavier JM, Moreira R, Rodrigues CM (2010) Celldeath targets and potential modulators in Alzheimer’s dis-ease. Curr Pharm Des 16, 2851-2864.

[1139] Savitz J, Solms M, Ramesar R (2006) The molecular genet-ics of cognition: Dopamine, COMT and BDNF. GenesBrain Behav 5, 311-328.

[1140] Tunbridge EM, Harrison PJ, Weinberger DR (2006)Catechol-o-methyltransferase, cognition, and psychosis:Val158Met and beyond. Biol Psychiatry 60, 141-151.

[1141] Dickinson D, Elvevag B (2009) Genes, cognition and brainthrough a COMT lens. Neuroscience 164, 72-87.

[1142] Apud JA, Weinberger DR (2007) Treatment of cognitivedeficits associated with schizophrenia: Potential role ofcatechol-O-methyltransferase inhibitors. CNS Drugs 21,535-557.

[1143] Gray JA, Roth BL (2007) Molecular targets for treatingcognitive dysfunction in schizophrenia. Schizophr Bull 33,1100-1119.

[1144] Apud JA, Mattay V, Chen J, Kolachana BS, CallicottJH, Rasetti R, Alce G, Iudicello JE, Akbar N, Egan MF,Goldberg TE, Weinberger DR (2007) Tolcapone improvescognition and cortical information processing in normalhuman subjects. Neuropsychopharmacology 32, 1011-1020.

[1145] Di Giovanni S, Eleuteri S, Paleologou KE, Yin G, Zweck-stetter M, Carrupt PA, Lashuel HA (2010) Entacapone andtolcapone, two catechol O-methyltransferase inhibitors,block fibril formation of alpha-synuclein and beta-amyloidand protect against amyloid-induced toxicity. J Biol Chem285, 14941-14954.

[1146] Hook V, Hook G, Kindy M (2010) Pharmacogenetic fea-tures of cathepsin B inhibitors that improve memory deficitand reduce beta-amyloid related to Alzheimer’s disease.Biol Chem 391, 861-872.

[1147] Hook V, Funkelstein L, Wegrzyn J, Bark S, Kindy M,Hook G (2012) Cysteine Cathepsins in the secretory vesi-cle produce active peptides: Cathepsin L generates peptideneurotransmitters and cathepsin B produces beta-amyloidof Alzheimer’s disease. Biochim Biophys Acta 1824, 89-104.

[1148] Sundelof J, Sundstrom J, Hansson O, Eriksdotter-Jonhagen M, Giedraitis V, Larsson A, Degerman-Gunnarsson M, Ingelsson M, Minthon L, Blennow K,Kilander L, Basun H, Lannfelt L (2010) Higher cathep-sin B levels in plasma in Alzheimer’s disease compared tohealthy controls. J Alzheimers Dis 22, 1223-1230.

[1149] Hook V, Kindy M, Hook G (2007) Cysteine proteaseinhibitors effectively reduce in vivo levels of brain beta-amyloid related to Alzheimer’s disease. Biol Chem 388,247-252.

[1150] Hook VY, Kindy M, Hook G (2008) Inhibitors of cathepsinB improve memory and reduce beta-amyloid in transgenic

Page 93: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 639

Alzheimer disease mice expressing the wild-type, but notthe Swedish mutant, beta-secretase site of the amyloidprecursor protein. J Biol Chem 283, 7745-7753.

[1151] Hook VY, Kindy M, Reinheckel T, Peters C, HookG (2009) Genetic cathepsin B deficiency reduces beta-amyloid in transgenic mice expressing human wild-typeamyloid precursor protein. Biochem Biophys Res Commun386, 284-288.

[1152] Hook G, Hook V, Kindy M (2011) The cysteine proteaseinhibitor, E64d, reduces brain amyloid-beta and improvesmemory deficits in Alzheimer’s disease animal modelsby inhibiting cathepsin B, but not BACE1, beta-secretaseactivity. J Alzheimers Dis 26, 387-408.

[1153] Hook G, Hook VY, Kindy M (2007) Cysteine proteaseinhibitors reduce brain beta-amyloid and beta-secretaseactivity in vivo and are potential Alzheimer’s disease ther-apeutics. Biol Chem 388, 979-983.

[1154] Bogdanovic N, Bretillon L, Lund EG, Diczfalusy U, Lan-nfelt L, Winblad B, Russell DW, Bjorkhem I (2001) On theturnover of brain cholesterol in patients with Alzheimer’sdisease. Abnormal induction of the cholesterol-catabolicenzyme CYP46 in glial cells. Neurosci Lett 314, 45-48.

[1155] Desai P, DeKosky ST, Kamboh MI (2002) Genetic vari-ation in the cholesterol 24-hydroxylase (CYP46) geneand the risk of Alzheimer’s disease. Neurosci Lett 328,9-12.

[1156] Papassotiropoulos A, Streffer JR, Tsolaki M, Schmid S,Thal D, Nicosia F, Iakovidou V, Maddalena A, Lutjo-hann D, Ghebremedhin E, Hegi T, Pasch T, Traxler M,Bruhl A, Benussi L, Binetti G, Braak H, Nitsch RM, HockC (2003) Increased brain beta-amyloid load, phosphory-lated tau, and risk of Alzheimer disease associated with anintronic CYP46 polymorphism. Arch Neurol 60, 29-35.

[1157] Papassotiropoulos A, Lambert JC, Wavrant-De VF,Wollmer MA, von der KH, Streffer JR, Maddalena A,Huynh KD, Wolleb S, Lutjohann D, Schneider B, Thal DR,Grimaldi LM, Tsolaki M, Kapaki E, Ravid R, KonietzkoU, Hegi T, Pasch T, Jung H, Braak H, Amouyel P, RogaevEI, Hardy J, Hock C, Nitsch RM (2005) Cholesterol 25-hydroxylase on chromosome 10q is a susceptibility genefor sporadic Alzheimer’s disease. Neurodegener Dis 2,233-241.

[1158] Fernandez DPV, Alvarez AM, Fernandez MM, GaldosAL, Gomez BF, Pena JA, Afonso-Sanchez MA, ZarranzImirizaldu JJ, de Pancorbo MM (2006) Polymorphism inthe cholesterol 24S-hydroxylase gene (CYP46A1) asso-ciated with the APOEpsilon3 allele increases the risk ofAlzheimer’s disease and of mild cognitive impairment pro-gressing to Alzheimer’s disease. Dement Geriatr CognDisord 21, 81-87.

[1159] Kolsch H, Lutjohann D, Jessen F, Popp J, Hentschel F,Kelemen P, Schmitz S, Maier W, Heun R (2009) CYP46A1variants influence Alzheimer’s disease risk and braincholesterol metabolism. Eur Psychiatry 24, 183-190.

[1160] Fu BY, Ma SL, Tang NL, Tam CW, Lui VW, Chiu HF,Lam LC (2009) Cholesterol 24-hydroxylase (CYP46A1)polymorphisms are associated with faster cognitive dete-rioration in Chinese older persons: A two-year follow upstudy. Int J Geriatr Psychiatry 24, 921-926.

[1161] Lai CL, Hsu CY, Liou LM, Hsieh HY, Hsieh YH, Liu CK(2011) Effect of cholesterol and CYP46 polymorphism oncognitive event-related potentials. Psychophysiology 48,1572-1577.

[1162] Li Y, Chu LW, Wang B, Yik PY, Huriletemuer, Jin DY,Ma X, Song YQ (2010) CYP46A1 functional promoter

haplotypes decipher genetic susceptibility to Alzheimer’sdisease. J Alzheimers Dis 21, 1311-1323.

[1163] Lutjohann D (2006) Cholesterol metabolism in the brain:Importance of 24S-hydroxylation. Acta Neurol ScandSuppl 185, 33-42.

[1164] Garcia AN, Muniz MT, Souza e Silva HR, da SilvaHA, thayde-Junior L (2009) Cyp46 polymorphisms inAlzheimer’s disease: A review. J Mol Neurosci 39, 342-345.

[1165] Mast N, White MA, Bjorkhem I, Johnson EF, Stout CD,Pikuleva IA (2008) Crystal structures of substrate-boundand substrate-free cytochrome P450 46A1, the principalcholesterol hydroxylase in the brain. Proc Natl Acad SciU S A 105, 9546-9551.

[1166] Lund EG, Guileyardo JM, Russell DW (1999) cDNAcloning of cholesterol 24-hydroxylase, a mediator ofcholesterol homeostasis in the brain. Proc Natl Acad SciU S A 96, 7238-7243.

[1167] Lund EG, Xie C, Kotti T, Turley SD, Dietschy JM, RussellDW (2003) Knockout of the cholesterol 24-hydroxylasegene in mice reveals a brain-specific mechanismof cholesterol turnover. J Biol Chem 278, 22980-22988.

[1168] Hudry E, Van DD, Kulik W, De Deyn PP, Stet FS, Ahouan-sou O, Benraiss A, Delacourte A, Bougneres P, Aubourg P,Cartier N (2010) Adeno-associated virus gene therapy withcholesterol 24-hydroxylase reduces the amyloid pathol-ogy before or after the onset of amyloid plaques in mousemodels of Alzheimer’s disease. Mol Ther 18, 44-53.

[1169] Carrero I, Gonzalo MR, Martin B, Sanz-Anquela JM,revalo-Serrano J, Gonzalo-Ruiz A (2012) Oligomers ofbeta-amyloid protein (Abeta1-42) induce the activationof cyclooxygenase-2 in astrocytes via an interaction withinterleukin-1beta, tumour necrosis factor-alpha, and anuclear factor kappa-B mechanism in the rat brain. ExpNeurol 236, 215-227.

[1170] McGeer PL, McGeer E, Rogers J, Sibley J (1990) Anti-inflammatory drugs and Alzheimer disease. Lancet 335,1037.

[1171] Pasinetti GM, Aisen PS (1998) Cyclooxygenase-2 expres-sion is increased in frontal cortex of Alzheimer’s diseasebrain. Neuroscience 87, 319-324.

[1172] Weggen S, Eriksen JL, Das P, Sagi SA, Wang R, PietrzikCU, Findlay KA, Smith TE, Murphy MP, Bulter T, KangDE, Marquez-Sterling N, Golde TE, Koo EH (2001) Asubset of NSAIDs lower amyloidogenic Abeta42 indepen-dently of cyclooxygenase activity. Nature 414, 212-216.

[1173] Weggen S, Rogers M, Eriksen J (2007) NSAIDs: Smallmolecules for prevention of Alzheimer’s disease or pre-cursors for future drug development? Trends PharmacolSci 28, 536-543.

[1174] Aisen PS (2002) Evaluation of selective COX-2 inhibitorsfor the treatment of Alzheimer’s disease. J Pain SymptomManage 23, S35-S40.

[1175] Golde TE, Erikson JL, Weggen S, Sagi SA, Koo EH (2002)Nonsteroidal antiinflammatory drugs as therapeutic agentsfor Alzheimer’s disease. Drug Dev Res 56, 415-420.

[1176] Pasinetti GM (2002) From epidemiology to therapeu-tic trials with anti-inflammatory drugs in Alzheimer’sdisease: The role of NSAIDs and cyclooxygenase in beta-amyloidosis and clinical dementia. J Alzheimers Dis 4,435-445.

[1177] McGeer PL, McGeer EG (2007) NSAIDs and Alzheimerdisease: Epidemiological, animal model and clinical stud-ies. Neurobiol Aging 28, 639-647.

Page 94: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

640 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

[1178] Szekely CA, Zandi PP (2010) Non-steroidal anti-inflammatory drugs and Alzheimer’s disease: Theepidemiological evidence. CNS Neurol Disord Drug Tar-gets 9, 132-139.

[1179] in ‘t Veld, Ruitenberg A, Hofman A, Launer LJ, vanDuijn CM, Stijnen T, Breteler MM, Stricker BH (2001)Nonsteroidal antiinflammatory drugs and the risk ofAlzheimer’s disease. N Engl J Med 345, 1515-1521.

[1180] Lyketsos CG, Breitner JC, Green RC, Martin BK, MeinertC, Piantadosi S, Sabbagh M (2007) Naproxen and cele-coxib do not prevent AD in early results from a randomizedcontrolled trial. Neurology 68, 1800-1808.

[1181] Leoutsakos JM, Muthen BO, Breitner JC, Lyketsos CG(2012) Effects of non-steroidal anti-inflammatory drugtreatments on cognitive decline vary by phase of pre-clinical Alzheimer disease: Findings from the randomizedcontrolled Alzheimer’s Disease Anti-inflammatory Pre-vention Trial. Int J Geriatr Psychiatry 27, 364-374.

[1182] Lleo A, Berezovska O, Herl L, Raju S, Deng A, BacskaiBJ, Frosch MP, Irizarry M, Hyman BT (2004) Nonsteroidalanti-inflammatory drugs lower Abeta42 and change prese-nilin 1 conformation. Nat Med 10, 1065-1066.

[1183] Gasparini L, Ongini E, Wenk G (2004) Non-steroidal anti-inflammatory drugs (NSAIDs) in Alzheimer’s disease: Oldand new mechanisms of action. J Neurochem 91, 521-536.

[1184] Hirohata M, Ono K, Naiki H, Yamada M (2005)Non-steroidal anti-inflammatory drugs have anti-amyloidogenic effects for Alzheimer’s beta-amyloidfibrils in vitro. Neuropharmacology 49, 1088-1099.

[1185] Hirohata M, Ono K, Yamada M (2008) Non-steroidal anti-inflammatory drugs as anti-amyloidogenic compounds.Curr Pharm Des 14, 3280-3294.

[1186] Leuchtenberger S, Beher D, Weggen S (2006) Selectivemodulation of Abeta42 production in Alzheimer’s disease:Non-steroidal anti-inflammatory drugs and beyond. CurrPharm Des 12, 4337-4355.

[1187] Smith SM, Uslaner JM, Hutson PH (2010) The therapeuticpotential of D-Amino Acid Oxidase (DAAO) inhibitors.Open Med Chem J 4, 3-9.

[1188] Jansen A, Krach S, Krug A, Markov V, Eggermann T,Zerres K, Thimm M, Nothen MM, Treutlein J, RietschelM, Kircher T (2009) Effect of the G72 (DAOA) putativerisk haplotype on cognitive functions in healthy subjects.BMC Psychiatry 9, 60.

[1189] Jansen A, Krach S, Krug A, Markov V, Eggermann T,Zerres K, Stocker T, Shah NJ, Nothen MM, Treutlein J,Rietschel M, Kircher T (2009) A putative high risk diplo-type of the G72 gene is in healthy individuals associatedwith better performance in working memory functions andaltered brain activity in the medial temporal lobe. Neuroim-age 45, 1002-1008.

[1190] Jansen A, Krach S, Krug A, Markov V, Thimm M, PaulusFM, Zerres K, Stocker T, Shah NJ, Nothen MM, Treut-lein J, Rietschel M, Kircher T (2010) The effect of G72genotype on neural correlates of memory encoding andretrieval. Neuroimage 53, 1001-1006.

[1191] Otte DM, Sommersberg B, Kudin A, Guerrero C,Albayram O, Filiou MD, Frisch P, Yilmaz O, Drews E,Turck CW, Bilkei-Gorzo A, Kunz WS, Beck H, ZimmerA (2011) N-acetyl cysteine treatment rescues cognitivedeficits induced by mitochondrial dysfunction in G72/G30transgenic mice. Neuropsychopharmacology 36, 2233-2243.

[1192] Schlenzig D, Manhart S, Cinar Y, Kleinschmidt M, HauseG, Willbold D, Funke SA, Schilling S, Demuth HU

(2009) Pyroglutamate formation influences solubility andamyloidogenicity of amyloid peptides. Biochemistry 48,7072-7078.

[1193] Schlenzig D, Ronicke R, Cynis H, Ludwig HH, Scheel E,Reymann K, Saido T, Hause G, Schilling S, Demuth HU(2012) N-Terminal pyroglutamate formation of Abeta38and Abeta40 enforces oligomer formation and potency todisrupt hippocampal long-term potentiation. J Neurochem121, 774-784.

[1194] Morawski M, Hartlage-Rubsamen M, Jager C, Waniek A,Schilling S, Schwab C, McGeer PL, Arendt T, Demuth HU,Rossner S (2010) Distinct glutaminyl cyclase expressionin Edinger-Westphal nucleus, locus coeruleus and nucleusbasalis Meynert contributes to pGlu-Abeta pathology inAlzheimer’s disease. Acta Neuropathol 120, 195-207.

[1195] Hartlage-Rubsamen M, Morawski M, Waniek A, Jager C,Zeitschel U, Koch B, Cynis H, Schilling S, Schliebs R,Demuth HU, Rossner S (2011) Glutaminyl cyclase con-tributes to the formation of focal and diffuse pyroglutamate(pGlu)-Abeta deposits in hippocampus via distinct cellularmechanisms. Acta Neuropathol 121, 705-719.

[1196] Jawhar S, Wirths O, Schilling S, Graubner S, Demuth HU,Bayer TA (2011) Overexpression of glutaminyl cyclase,the enzyme responsible for pyroglutamate Abeta forma-tion, induces behavioral deficits, and glutaminyl cyclaseknock-out rescues the behavioral phenotype in 5XFADmice. J Biol Chem 286, 4454-4460.

[1197] Jawhar S, Wirths O, Bayer TA (2011) Pyroglutamateamyloid-beta (Abeta): A hatchet man in Alzheimer dis-ease. J Biol Chem 286, 38825-38832.

[1198] Schilling S, Niestroj AJ, Rahfeld JU, Hoffmann T, Wer-mann M, Zunkel K, Wasternack C, Demuth HU (2003)Identification of human glutaminyl cyclase as a metal-loenzyme. Potent inhibition by imidazole derivatives andheterocyclic chelators. J Biol Chem 278, 49773-49779.

[1199] Schilling S, Lauber T, Schaupp M, Manhart S, Scheel E,Bohm G, Demuth HU (2006) On the seeding and oligomer-ization of pGlu-amyloid peptides (in vitro). Biochemistry45, 12393-12399.

[1200] Schilling S, Zeitschel U, Hoffmann T, Heiser U, FranckeM, Kehlen A, Holzer M, Hutter-Paier B, Prokesch M,Windisch M, Jagla W, Schlenzig D, Lindner C, RudolphT, Reuter G, Cynis H, Montag D, Demuth HU, Rossner S(2008) Glutaminyl cyclase inhibition attenuates pyroglu-tamate Abeta and Alzheimer’s disease-like pathology. NatMed 14, 1106-1111.

[1201] Schilling S, Appl T, Hoffmann T, Cynis H, SchulzK, Jagla W, Friedrich D, Wermann M, Buchholz M,Heiser U, von HS, Demuth HU (2008) Inhibition ofglutaminyl cyclase prevents pGlu-Abeta formation afterintracortical/hippocampal microinjection in vivo/in situ. JNeurochem 106, 1225-1236.

[1202] Cynis H, Schilling S, Bodnar M, Hoffmann T, Heiser U,Saido TC, Demuth HU (2006) Inhibition of glutaminylcyclase alters pyroglutamate formation in mammaliancells. Biochim Biophys Acta 1764, 1618-1625.

[1203] Cynis H, Scheel E, Saido TC, Schilling S, Demuth HU(2008) Amyloidogenic processing of amyloid precursorprotein: Evidence of a pivotal role of glutaminyl cyclase ingeneration of pyroglutamate-modified amyloid-beta. Bio-chemistry 47, 7405-7413.

[1204] Nussbaum JM, Schilling S, Cynis H, Silva A, SwansonE, Wangsanut T, Tayler K, Wiltgen B, Hatami A, RonickeR, Reymann K, Hutter-Paier B, Alexandru A, Jagla W,Graubner S, Glabe CG, Demuth HU, Bloom GS (2012)

Page 95: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 641

Prion-like behaviour and tau-dependent cytotoxicity ofpyroglutamylated amyloid-beta. Nature 485, 651-655.

[1204] [4]Koch B, Buchholz M, Wermann M, Heiser U, SchillingS, Demuth HU (2012) Probing Secondary Glutaminyl-Cyclase (QC)-Inhibitor Interactions. Chem Biol Drug Des,doi: 10.1111/cbdd.12046.-

[1205] Sultana R, Boyd-Kimball D, Cai J, Pierce WM, Klein JB,Merchant M, Butterfield DA (2007) Proteomics analy-sis of the Alzheimer’s disease hippocampal proteome. JAlzheimers Dis 11, 153-164.

[1206] Shalova IN, Cechalova K, Rehakova Z, Dimitrova P,Ognibene E, Caprioli A, Schmalhausen EV, MuronetzVI, Saso L (2007) Decrease of dehydrogenase activityof cerebral glyceraldehyde-3-phosphate dehydrogenase indifferent animal models of Alzheimer’s disease. BiochimBiophys Acta 1770, 826-832.

[1207] Kragten E, Lalande I, Zimmermann K, Roggo S, SchindlerP, Muller D, van OJ, Waldmeier P, Furst P (1998)Glyceraldehyde-3-phosphate dehydrogenase, the putativetarget of the antiapoptotic compounds CGP 3466 and R-(-)-deprenyl. J Biol Chem 273, 5821-5828.

[1208] Zimmermann K, Roggo S, Kragten E, Furst P, WaldmeierP (1998) Synthesis of tools for target identification of theanti-apoptotic compound CGP 3466; Part I. Bioorg MedChem Lett 8, 1195-1200.

[1209] Andringa G, Cools AR (2000) The neuroprotective effectsof CGP 3466B in the best in vivo model of Parkinson’sdisease, the bilaterally MPTP-treated rhesus monkey. JNeural Transm Suppl 215-225.

[1210] Andringa G, van Oosten RV, Unger W, Hafmans TG,Veening J, Stoof JC, Cools AR (2000) Systemic admin-istration of the propargylamine CGP 3466B preventsbehavioural and morphological deficits in rats with 6-hydroxydopamine-induced lesions in the substantia nigra.Eur J Neurosci 12, 3033-3043.

[1211] Andringa G, Eshuis S, Perentes E, Maguire RP, Roth D,Ibrahim M, Leenders KL, Cools AR (2003) TCH346 pre-vents motor symptoms and loss of striatal FDOPA uptakein bilaterally MPTP-treated primates. Neurobiol Dis 14,205-217.

[1212] Sagot Y, Toni N, Perrelet D, Lurot S, King B, Rixner H,Mattenberger L, Waldmeier PC, Kato AC (2000) An orallyactive anti-apoptotic molecule (CGP 3466B) preservesmitochondria and enhances survival in an animal modelof motoneuron disease. Br J Pharmacol 131, 721-728.

[1213] Waldmeier PC, Spooren WP, Hengerer B (2000) CGP 3466protects dopaminergic neurons in lesion models of Parkin-son’s disease. Naunyn Schmiedebergs Arch Pharmacol362, 526-537.

[1214] Waldmeier PC, Boulton AA, Cools AR, Kato AC, TattonWG (2000) Neurorescuing effects of the GAPDH ligandCGP 3466B. J Neural Transm Suppl 197-214.

[1215] Waldmeier PC (2003) Prospects for antiapoptotic drugtherapy of neurodegenerative diseases. Prog Neuropsy-chopharmacol Biol Psychiatry 27, 303-321.

[1216] Matarredona ER, Meyer M, Seiler RW, Widmer HR (2003)CGP 3466 increases survival of cultured fetal dopaminer-gic neurons. Restor Neurol Neurosci 21, 29-37.

[1217] Hara MR, Thomas B, Cascio MB, Bae BI, HesterLD, Dawson VL, Dawson TM, Sawa A, Snyder SH(2006) Neuroprotection by pharmacologic blockade of theGAPDH death cascade. Proc Natl Acad Sci U S A 103,3887-3889.

[1218] Olanow CW, Schapira AH, LeWitt PA, Kieburtz K, SauerD, Olivieri G, Pohlmann H, Hubble J (2006) TCH346 as

a neuroprotective drug in Parkinson’s disease: A double-blind, randomised, controlled trial. Lancet Neurol 5, 1013-1020.

[1219] Miller R, Bradley W, Cudkowicz M, Hubble J, MeiningerV, Mitsumoto H, Moore D, Pohlmann H, Sauer D, Silani V,Strong M, Swash M, Vernotica E (2007) Phase II/III ran-domized trial of TCH346 in patients with ALS. Neurology69, 776-784.

[1220] Erb M, Meinen S, Barzaghi P, Sumanovski LT, Courdier-Fruh I, Ruegg MA, Meier T (2009) Omigapil amelioratesthe pathology of muscle dystrophy caused by laminin-alpha2 deficiency. J Pharmacol Exp Ther 331, 787-795.

[1221] Meinen S, Lin S, Thurnherr R, Erb M, Meier T, Ruegg MA(2011) Apoptosis inhibitors and mini-agrin have additivebenefits in congenital muscular dystrophy mice. EMBOMol Med 3, 465-479.

[1222] Munoz-Montano JR, Lim F, Moreno FJ, Avila J, Diaz-NidoJ (1999) Glycogen synthase kinase-3 modulates neuriteoutgrowth in cultured neurons: Possible implications forneurite pathology in Alzheimer’s disease. J Alzheimers Dis1, 361-378.

[1223] Eldar-Finkelman H (2002) Glycogen synthase kinase 3:An emerging therapeutic target. Trends Mol Med 8, 126-132.

[1224] Martinez A, Alonso M, Castro A, Perez C, MorenoFJ (2002) First non-ATP competitive glycogen synthasekinase 3 beta (GSK-3beta) inhibitors: Thiadiazolidinones(TDZD) as potential drugs for the treatment of Alzheimer’sdisease. J Med Chem 45, 1292-1299.

[1225] Martinez A, Castro A, Dorronsoro I, Alonso M (2002)Glycogen synthase kinase 3 (GSK-3) inhibitors as newpromising drugs for diabetes, neurodegeneration, cancer,and inflammation. Med Res Rev 22, 373-384.

[1226] Bhat RV, Budd Haeberlein SL, Avila J (2004) Glycogensynthase kinase 3: A drug target for CNS therapies. JNeurochem 89, 1313-1317.

[1227] Cohen P, Goedert M (2004) GSK3 inhibitors: Develop-ment and therapeutic potential. Nat Rev Drug Discov 3,479-487.

[1228] Meijer L, Flajolet M, Greengard P (2004) Pharmacologicalinhibitors of glycogen synthase kinase 3. Trends Pharma-col Sci 25, 471-480.

[1229] Chin PC, Majdzadeh N, D’Mello SR (2005) Inhibitionof GSK3beta is a common event in neuroprotection bydifferent survival factors. Brain Res Mol Brain Res 137,193-201.

[1230] Huang HC, Klein PS (2006) Multiple roles for glycogensynthase kinase-3 as a drug target in Alzheimer’s disease.Curr Drug Targets 7, 1389-1397.

[1231] Muyllaert D, Terwel D, Borghgraef P, Devijver H,Dewachter I, Van Leuven F (2006) Transgenic mousemodels for Alzheimer’s disease: The role of GSK-3B incombined amyloid and tau-pathology. Rev Neurol (Paris)162, 903-907.

[1232] Takashima A (2006) GSK-3 is essential in the pathogenesisof Alzheimer’s disease. J Alzheimers Dis 9(3 Suppl), 309-317.

[1233] Avila J, Hernandez F (2007) GSK-3 inhibitors forAlzheimer’s disease. Expert Rev Neurother 7, 1527-1533.

[1234] Mazanetz MP, Fischer PM (2007) Untangling tau hyper-phosphorylation in drug design for neurodegenerativediseases. Nat Rev Drug Discov 6, 464-479.

[1235] Hooper C, Killick R, Lovestone S (2008) The GSK3hypothesis of Alzheimer’s disease. J Neurochem 104,1433-1439.

Page 96: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

642 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

[1236] Martinez A (2008) Preclinical efficacy on GSK-3inhibitors: Towards a future generation of powerful drugs.Med Res Rev 28, 773-796.

[1237] Martinez A, Perez DI (2008) GSK-3 inhibitors: A rayof hope for the treatment of Alzheimer’s disease? JAlzheimers Dis 15, 181-191.

[1238] Medina M, Castro A (2008) Glycogen synthase kinase-3 (GSK-3) inhibitors reach the clinic. Curr Opin DrugDiscov Devel 11, 533-543.

[1239] Muyllaert D, Kremer A, Jaworski T, Borghgraef P, Devi-jver H, Croes S, Dewachter I, Van Leuven F (2008)Glycogen synthase kinase-3beta, or a link between amy-loid and tau pathology? Genes Brain Behav 7(Suppl 1),57-66.

[1240] Terwel D, Muyllaert D, Dewachter I, Borghgraef P, CroesS, Devijver H, Van Leuven F (2008) Amyloid activatesGSK-3beta to aggravate neuronal tauopathy in bigenicmice. Am J Pathol 172, 786-798.

[1241] Dewachter I, Ris L, Jaworski T, Seymour CM, KremerA, Borghgraef P, De VH, Godaux E, Van Leuven F(2009) GSK3beta, a centre-staged kinase in neuropsychi-atric disorders, modulates long term memory by inhibitoryphosphorylation at serine-9. Neurobiol Dis 35, 193-200.

[1242] Giese KP (2009) GSK-3: A key player in neurodegenera-tion and memory. IUBMB Life 61, 516-521.

[1243] Zhou XW, Winblad B, Guan Z, Pei JJ (2009) Interactionsbetween glycogen synthase kinase 3beta, protein kinaseB, and protein phosphatase 2A in tau phosphorylation inmouse N2a neuroblastoma cells. J Alzheimers Dis 17, 929-937.

[1244] Avila. J, Wandosell F, Hernandez F (2010) Role of glyco-gen synthase kinase-3 in Alzheimer’s disease pathogenesisand glycogen synthase kinase-3 inhibitors. Expert RevNeurother 10, 703-710.

[1245] Lei P, Ayton S, Bush AI, Adlard PA (2011) GSK-3 inNeurodegenerative Diseases. Int J Alzheimers Dis 2011,189246.

[1246] Martinez A, Gil C, Perez DI (2011) Glycogen synthasekinase 3 inhibitors in the next horizon for Alzheimer’sdisease treatment. Int J Alzheimers Dis 2011, 280502.

[1247] Eldar-Finkelman H, Martinez A (2011) GSK-3 inhibitors:preclinical and clinical focus on CNS. Front Mol Neurosci4, 32.

[1248] Mines MA, Jope RS (2011) Glycogen synthase kinase-3: A promising therapeutic target for fragile x syndrome.Front Mol Neurosci 4, 35.

[1249] Mines MA, Beurel E, Jope RS (2011) Regulation of cellsurvival mechanisms in Alzheimer’s disease by glycogensynthase kinase-3. Int J Alzheimers Dis 2011, 861072.

[1250] Kremer A, Louis JV, Jaworski T, Van Leuven F (2011)GSK3 and Alzheimer’s disease: Facts and fiction. FrontMol Neurosci 4, 17.

[1251] Amar S, Belmaker RH, Agam G (2011) The possibleinvolvement of glycogen synthase kinase-3 (GSK-3) indiabetes, cancer and central nervous system diseases. CurrPharm Des 17, 2264-2277.

[1252] Martin L, Page G, Terro F (2011) Tau phosphorylationand neuronal apoptosis induced by the blockade of PP2Apreferentially involve GSK3beta. Neurochem Int 59, 235-250.

[1253] Avila J, Leon-Espinosa G, Garcia E, Garcia-Escudero V,Hernandez F, Defelipe J (2012) Tau phosphorylation byGSK3 in different conditions. Int J Alzheimers Dis 2012,578373.

[1254] Hernandez F, Lucas JJ, Avila J (2012) GSK3 and tau: Twoconvergence points in Alzheimer’s disease. J AlzheimersDis, doi: 10.3233/JAD-2012-129025 [Epubl ahead ofprint].

[1255] Li XH, Lv BL, Xie JZ, Liu J, Zhou XW, Wang JZ (2012)AGEs induce Alzheimer-like tau pathology and memorydeficit via RAGE-mediated GSK-3 activation. NeurobiolAging 33, 1400-1410.

[1256] Mondragon-Rodriguez S, Perry G, Zhu X, Moreira PI,Williams S (2012) Glycogen synthase kinase 3: A point ofintegration in Alzheimer’s disease and a therapeutic target?Int J Alzheimers Dis 2012, 276803.

[1257] Rada P, Rojo AI, Evrard-Todeschi N, Innamorato NG,Cotte A, Jaworski T, Tobon-Velasco JC, Devijver H,Garcia-Mayoral MF, Van Leuven F, Hayes JD, Bertho G,Cuadrado A (2012) Structural and functional characteriza-tion of Nrf2 degradation by the glycogen synthase kinase3/beta-TrCP axis. Mol Cell Biol 32, 3486-3499.

[1258] Rocha-Souto B, Coma M, Perez-Nievas BG, Scotton TC,Siao M, Sanchez-Ferrer P, Hashimoto T, Fan Z, HudryE, Barroeta I, Sereno L, Rodriguez M, Sanchez MB,Hyman BT, Gomez-Isla T (2012) Activation of glyco-gen synthase kinase-3 beta mediates beta-amyloid inducedneuritic damage in Alzheimer’s disease. Neurobiol Dis 45,425-437.

[1259] Cai Z, Zhao Y, Zhao B (2012) Roles of glycogen synthasekinase 3 in Alzheimer’s disease. Curr Alzheimer Res, inpress. [Epubl ahead of print].

[1260] Parr C, Carzaniga R, Gentleman S, Van Leuven F,Walter J, Sastre M (2012) GSK3 inhibition promoteslysosomal biogenesis and the autophagic degradation ofthe amyloid-beta precursor protein. Mol Cell Biol, doi:10.1128/MCB.00930-12 [Epubl ahead of print].

[1261] Dajani R, Fraser E, Roe SM, Young N, Good V, DaleTC, Pearl LH (2001) Crystal structure of glycogen syn-thase kinase 3 beta: Structural basis for phosphate-primedsubstrate specificity and autoinhibition. Cell 105, 721-732.

[1262] Ter Haar E, Coll JT, Austen DA, Hsiao HM, SwensonL, Jain J (2001) Structure of GSK3beta reveals a primedphosphorylation mechanism. Nat Struct Biol 8, 593-596.

[1263] Martinez A, Alonso M, Castro A, Dorronsoro I, Gelpi JL,Luque FJ, Perez C, Moreno FJ (2005) SAR and 3D-QSARstudies on thiadiazolidinone derivatives: Exploration ofstructural requirements for glycogen synthase kinase 3inhibitors. J Med Chem 48, 7103-7112.

[1264] Akhtar M, Bharatam PV (2012) 3D-QSAR and moleculardocking studies on 3-anilino-4-arylmaleimide derivativesas glycogen synthase kinase-3beta inhibitors. Chem BiolDrug Des 79, 560-571.

[1265] Karpov PV, Osolodkin DI, Baskin II, Palyulin VA, ZefirovNS (2011) One-class classification as a novel method ofligand-based virtual screening: The case of glycogen syn-thase kinase 3beta inhibitors. Bioorg Med Chem Lett 21,6728-6731.

[1266] Osolodkin DI, Palyulin VA, Zefirov NS (2011) Structure-based virtual screening of glycogen synthase kinase 3betainhibitors: Analysis of scoring functions applied to largetrue actives and decoy sets. Chem Biol Drug Des 78, 378-390.

[1267] Castro A, Encinas A, Gil C, Brase S, Porcal W, PerezC, Moreno FJ, Martinez A (2008) Non-ATP competitiveglycogen synthase kinase 3beta (GSK-3beta) inhibitors:Study of structural requirements for thiadiazolidinonederivatives. Bioorg Med Chem 16, 495-510.

Page 97: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 643

[1268] del Ser T, Steinwachs KC, Gertz HJ, Andres MV, Gomez-Carrillo B, Medina M, Vericat JA, Redondo P, Fleet D,Leon T (2012) Treatment of Alzheimer’s disease with theGSK-3 inhibitor tideglusib: A pilot study. J AlzheimersDis, doi: 10.3233/JAD-2012-120805 [Epubl ahead ofprint].

[1269] Cociorva OM, Li B, Nomanbhoy T, Li Q, Nakamura A,Nakamura K, Nomura M, Okada K, Seto S, Yumoto K,Liyanage M, Zhang MC, Aban A, Leen B, SzardeningsAK, Rosenblum JS, Kozarich JW, Kohno Y, Shreder KR(2011) Synthesis and structure-activity relationship of 4-quinolone-3-carboxylic acid based inhibitors of glycogensynthase kinase-3beta. Bioorg Med Chem Lett 21, 5948-5951.

[1270] Coffman K, Brodney M, Cook J, Lanyon L, PanditJ, Sakya S, Schachter J, Tseng-Lovering E, WesselM (2011) 6-amino-4-(pyrimidin-4-yl)pyridones: Novelglycogen synthase kinase-3beta inhibitors. Bioorg MedChem Lett 21, 1429-1433.

[1271] Saitoh M, Kunitomo J, Kimura E, Hayase Y, Kobayashi H,Uchiyama N, Kawamoto T, Tanaka T, Mol CD, DouganDR, Textor GS, Snell GP, Itoh F (2009) Design, synthe-sis and structure-activity relationships of 1,3,4-oxadiazolederivatives as novel inhibitors of glycogen synthase kinase-3beta. Bioorg Med Chem 17, 2017-2029.

[1272] Saitoh M, Kunitomo J, Kimura E, Iwashita H, Uno Y,Onishi T, Uchiyama N, Kawamoto T, Tanaka T, MolCD, Dougan DR, Textor GP, Snell GP, Takizawa M,Itoh F, Kori M (2009) 2-3-[4-(Alkylsulfinyl)phenyl]-1-benzofuran-5-yl-5-methyl-1,3,4-oxadiazol e derivatives asnovel inhibitors of glycogen synthase kinase-3beta withgood brain permeability. J Med Chem 52, 6270-6286.

[1273] Saitoh M, Kunitomo J, Kimura E, Yamano T, Itoh F, KoriM (2010) Enantioselective synthesis of the novel chiralsulfoxide derivative as a glycogen synthase kinase 3betainhibitor. Chem Pharm Bull (Tokyo) 58, 1252-1254.

[1274] Onishi T, Iwashita H, Uno Y, Kunitomo J, Saitoh M,Kimura E, Fujita H, Uchiyama N, Kori M, Takizawa M(2011) A novel glycogen synthase kinase-3 inhibitor 2-methyl-5-(3-4-[(S )-methylsulfinyl]phenyl-1-benzofuran-5-yl)-1,3,4-oxadiazole decreases tau phosphorylation andameliorates cognitive deficits in a transgenic model ofAlzheimer’s disease. J Neurochem 119, 1330-1340.

[1275] Ding S, Wu TY, Brinker A, Peters EC, Hur W, Gray NS,Schultz PG (2003) Synthetic small molecules that con-trol stem cell fate. Proc Natl Acad Sci U S A 100, 7632-7637.

[1276] Paterniti I, Mazzon E, Gil C, Impellizzeri D, Palomo V,Redondo M, Perez DI, Esposito E, Martinez A, CuzzocreaS (2011) PDE 7 inhibitors: New potential drugs for thetherapy of spinal cord injury. PLoS One 6, e15937.

[1277] Lipina TV, Palomo V, Gil C, Martinez A, Roder JC (2013)Dual inhibitor of PDE7 and GSK-3 - VP1.15 acts asantipsychotic and cognitive enhancer in C57BL/6J mice.Neuropharmacology 61, 205-214.

[1278] Bhat R, Xue Y, Berg S, Hellberg S, Ormo M, NilssonY, Radesater AC, Jerning E, Markgren PO, Borgegard T,Nylof M, Gimenez-Cassina A, Hernandez F, Lucas JJ, az-Nido J, Avila J (2003) Structural insights and biologicaleffects of glycogen synthase kinase 3-specific inhibitorAR-A014418. J Biol Chem 278, 45937-45945.

[1279] Gould TD, Einat H, Bhat R, Manji HK (2004)AR-A014418, a selective GSK-3 inhibitor, producesantidepressant-like effects in the forced swim test. Int JNeuropsychopharmacol 7, 387-390.

[1280] Berg S, Bergh M, Hellberg S, Hogdin K, Lo-Alfredsson Y,Soderman P, von BS, Weigelt T, Ormo M, Xue Y, Tucker J,Neelissen J, Jerning E, Nilsson Y, Bhat R (2012) Discov-ery of novel potent and highly selective glycogen synthasekinase-3beta (GSK3beta) inhibitors for Alzheimer’s dis-ease: Design, synthesis, and characterization of pyrazines.J Med Chem, doi: 10.1021/jm201742m [Epubl ahead ofprint].

[1281] Koryakova AG, Ivanenkov YA, Ryzhova EA, BulanovaEA, Karapetian RN, Mikitas OV, Katrukha EA,Kazey VI, Okun I, Kravchenko DV, Lavrovsky YV,Korzinov OM, Ivachtchenko AV (2008) Novel aryland heteroaryl substituted N-[3-(4-phenylpiperazin-1-yl)propyl]-1,2,4-oxadiazole-5-carboxamides as selectiveGSK-3 inhibitors. Bioorg Med Chem Lett 18, 3661-3666.

[1282] Macaulay K, Hajduch E, Blair AS, Coghlan MP, SmithSA, Hundal HS (2003) Use of lithium and SB-415286to explore the role of glycogen synthase kinase-3 in theregulation of glucose transport and glycogen synthase. EurJ Biochem 270, 3829-3838.

[1283] Pizarro JG, Yeste-Velasco M, Rimbau V, CasadesusG, Smith MA, Pallas M, Folch J, Camins A (2008)Neuroprotective effects of SB-415286 on hydrogenperoxide-induced cell death in B65 rat neuroblastoma cellsand neurons. Int J Dev Neurosci 26, 269-276.

[1284] Pizarro JG, Folch J, Esparza JL, Jordan J, Pallas M, CaminsA (2009) A molecular study of pathways involved in theinhibition of cell proliferation in neuroblastoma B65 cellsby the GSK-3 inhibitors lithium and SB-415286. J CellMol Med 13, 3906-3917.

[1285] Yeste-Velasco M, Folch J, Jimenez A, Rimbau V, Pallas M,Camins A (2008) GSK-3 beta inhibition and preventionof mitochondrial apoptosis inducing factor release are notinvolved in the antioxidant properties of SB-415286. EurJ Pharmacol 588, 239-243.

[1286] Gentile G, Bernasconi G, Pozzan A, Merlo G, MarzoratiP, Bamborough P, Bax B, Bridges A, Brough C, CarterP, Cutler G, Neu M, Takada M (2011) Identification of2-(4-pyridyl)thienopyridinones as GSK-3beta inhibitors.Bioorg Med Chem Lett 21, 4823-4827.

[1287] Gentile G, Merlo G, Pozzan A, Bernasconi G, Bax B, Bam-borough P, Bridges A, Carter P, Neu M, Yao G, BroughC, Cutler G, Coffin A, Belyanskaya S (2012) 5-Aryl-4-carboxamide-1,3-oxazoles: Potent and selective GSK-3inhibitors. Bioorg Med Chem Lett 22, 1989-1994.

[1288] Vadivelan S, Sinha BN, Tajne S, Jagarlapudi SA (2009)Fragment and knowledge-based design of selective GSK-3beta inhibitors using virtual screening models. Eur J MedChem 44, 2361-2371.

[1289] Kuo GH, Prouty C, Deangelis A, Shen L, O’Neill DJ,Shah C, Connolly PJ, Murray WV, Conway BR, CheungP, Westover L, Xu JZ, Look RA, Demarest KT, EmanuelS, Middleton SA, Jolliffe L, Beavers MP, Chen X (2003)Synthesis and discovery of macrocyclic polyoxygenatedbis-7-azaindolylmaleimides as a novel series of potent andhighly selective glycogen synthase kinase-3beta inhibitors.J Med Chem 46, 4021-4031.

[1290] O’Neill DJ, Shen L, Prouty C, Conway BR, Westover L,Xu JZ, Zhang HC, Maryanoff BE, Murray WV, DemarestKT, Kuo GH (2004) Design, synthesis, and biologicalevaluation of novel 7-azaindolyl-heteroaryl-maleimidesas potent and selective glycogen synthase kinase-3beta(GSK-3beta) inhibitors. Bioorg Med Chem 12, 3167-3185.

Page 98: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

644 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

[1291] Shen L, Prouty C, Conway BR, Westover L, Xu JZ,Look RA, Chen X, Beavers MP, Roberts J, Mur-ray WV, Demarest KT, Kuo GH (2004) Synthesisand biological evaluation of novel macrocyclic bis-7-azaindolylmaleimides as potent and highly selectiveglycogen synthase kinase-3 beta (GSK-3 beta) inhibitors.Bioorg Med Chem 12, 1239-1255.

[1292] Zhang HC, Ye H, Conway BR, Derian CK, Addo MF,Kuo GH, Hecker LR, Croll DR, Li J, Westover L, Xu JZ,Look R, Demarest KT, ndrade-Gordon P, Damiano BP,Maryanoff BE (2004) 3-(7-Azaindolyl)-4-arylmaleimidesas potent, selective inhibitors of glycogen synthase kinase-3. Bioorg Med Chem Lett 14, 3245-3250.

[1293] Lum C, Kahl J, Kessler L, Kucharski J, Lundstrom J, MillerS, Nakanishi H, Pei Y, Pryor K, Roberts E, Sebo L, Sulli-van R, Urban J, Wang Z (2008) 2,5-Diaminopyrimidinesand 3,5-disubstituted azapurines as inhibitors of glycogensynthase kinase-3 (GSK-3). Bioorg Med Chem Lett 18,3578-3581.

[1294] Engler TA, Henry JR, Malhotra S, Cunningham B,Furness K, Brozinick J, Burkholder TP, Clay MP, Clay-ton J, Diefenbacher C, Hawkins E, Iversen PW, LiY, Lindstrom TD, Marquart AL, McLean J, MendelD, Misener E, Briere D, O’toole JC, Porter WJ,Queener S, Reel JK, Owens RA, Brier RA, Eessalu TE,Wagner JR, Campbell RM, Vaughn R (2004) Substi-tuted 3-imidazo[1,2-a]pyridin-3-yl- 4-(1,2,3,4-tetrahydro-[1,4]diazepino-[6,7,1-hi]indol-7-yl)pyrrole-2,5-dion es ashighly selective and potent inhibitors of glycogen synthasekinase-3. J Med Chem 47, 3934-3937.

[1295] Engler TA, Malhotra S, Burkholder TP, Henry JR, MendelD, Porter WJ, Furness K, Diefenbacher C, Marquart A,Reel JK, Li Y, Clayton J, Cunningham B, McLean J,O’toole JC, Brozinick J, Hawkins E, Misener E, BriereD, Brier RA, Wagner JR, Campbell RM, Anderson BD,Vaughn R, Bennett DB, Meier TI, Cook JA (2005) Thedevelopment of potent and selective bisarylmaleimideGSK3 inhibitors. Bioorg Med Chem Lett 15, 899-903.

[1296] Magnus NA, Ley CP, Pollock PM, Wepsiec JP (2010)Pictet-Spengler based synthesis of a bisarylmaleimideglycogen synthase kinase-3 inhibitor. Org Lett 12, 3700-3703.

[1297] Gong L, Hirschfeld D, Tan YC, Heather HJ, Peltz G, AvnurZ, Dunten P (2010) Discovery of potent and bioavailableGSK-3beta inhibitors. Bioorg Med Chem Lett 20, 1693-1696.

[1298] Arnost M, Pierce A, Ter HE, Lauffer D, Madden J, TannerK, Green J (2010) 3-Aryl-4-(arylhydrazono)-1H-pyrazol-5-ones: Highly ligand efficient and potent inhibitors ofGSK3beta. Bioorg Med Chem Lett 20, 1661-1664.

[1299] Shin D, Lee SC, Heo YS, Lee WY, Cho YS, Kim YE, HyunYL, Cho JM, Lee YS, Ro S (2007) Design and synthe-sis of 7-hydroxy-1H-benzoimidazole derivatives as novelinhibitors of glycogen synthase kinase-3beta. Bioorg MedChem Lett 17, 5686-5689.

[1300] Polychronopoulos P, Magiatis P, Skaltsounis AL, Myri-anthopoulos V, Mikros E, Tarricone A, Musacchio A, RoeSM, Pearl L, Leost M, Greengard P, Meijer L (2004)Structural basis for the synthesis of indirubins as potentand selective inhibitors of glycogen synthase kinase-3 andcyclin-dependent kinases. J Med Chem 47, 935-946.

[1301] Sereno L, Coma M, Rodriguez M, Sanchez-Ferrer P,Sanchez MB, Gich I, Agullo JM, Perez M, Avila J,Guardia-Laguarta C, Clarimon J, Lleo A, Gomez-Isla T(2009) A novel GSK-3beta inhibitor reduces Alzheimer’s

pathology and rescues neuronal loss in vivo. Neurobiol Dis35, 359-367.

[1302] Stukenbrock H, Mussmann R, Geese M, Ferandin Y,Lozach O, Lemcke T, Kegel S, Lomow A, Burk U,Dohrmann C, Meijer L, Austen M, Kunick C (2008) 9-cyano-1-azapaullone (cazpaullone), a glycogen synthasekinase-3 (GSK-3) inhibitor activating pancreatic beta cellprotection and replication. J Med Chem 51, 2196-2207.

[1303] Rochais C, Lescot E, Lisowski V, Lepailleur A, SantosJS, Bureau R, Dallemagne P, Meijer L, Rault S (2004)Synthesis and biological evaluation of thienopyrrolizines,a new family of CDK/GSK-3 inhibitors. J Enzyme InhibMed Chem 19, 585-593.

[1304] Rochais C, Duc NV, Lescot E, Sopkova-de Oliveira SJ,Bureau R, Meijer L, Dallemagne P, Rault S (2009) Synthe-sis of new dipyrrolo- and furopyrrolopyrazinones relatedto tripentones and their biological evaluation as potentialkinases (CDKs1-5, GSK-3) inhibitors. Eur J Med Chem44, 708-716.

[1305] Kozikowski AP, Gaisina IN, Petukhov PA, Sridhar J,King LT, Blond SY, Duka T, Rusnak M, Sidhu A (2006)Highly potent and specific GSK-3beta inhibitors that blocktau phosphorylation and decrease alpha-synuclein pro-tein expression in a cellular model of Parkinson’s disease.ChemMedChem 1, 256-266.

[1306] Kozikowski AP, Gaisina IN, Yuan H, Petukhov PA,Blond SY, Fedolak A, Caldarone B, McGonigle P (2007)Structure-based design leads to the identification of lithiummimetics that block mania-like effects in rodents. possiblenew GSK-3beta therapies for bipolar disorders. J Am ChemSoc 129, 8328-8332.

[1307] Kozikowski AP, Gunosewoyo H, Guo S, Gaisina IN,Walter RL, Ketcherside A, McClung CA, MesecarAD, Caldarone B (2011) Identification of a glycogensynthase kinase-3beta inhibitor that attenuates hyperac-tivity in CLOCK mutant mice. ChemMedChem 6, 1593-1602.

[1308] Gaisina IN, Gallier F, Ougolkov AV, Kim KH, KuromeT, Guo S, Holzle D, Luchini DN, Blond SY, BilladeauDD, Kozikowski AP (2009) From a natural product leadto the identification of potent and selective benzofuran-3-yl-(indol-3-yl)maleimides as glycogen synthase kinase3beta inhibitors that suppress proliferation and survivalof pancreatic cancer cells. J Med Chem 52, 1853-1863.

[1309] Chen W, Gaisina IN, Gunosewoyo H, Malekiani SA, Hana-nia T, Kozikowski AP (2011) Structure-guided design of ahighly selective glycogen synthase kinase-3beta inhibitor:A superior neuroprotective pyrazolone showing antimaniaeffects. ChemMedChem 6, 1587-1592.

[1310] Chen PC, Gaisina IN, El-Khodor BF, Ramboz S, Makhor-tova NR, Rubin LL, Kozikowski AP (2012) Identificationof a maleimide-based glycogen synthase kinase-3 (GSK-3) inhibitor, BIP-135, that prolongs the median survivaltime of Delta7 SMA KO mouse model of spinal muscularatrophy. ACS Chem Neurosci 3, 5-11.

[1311] Lo Monte F, Kramer T, Bolander A, Plotkin B, Eldar-Finkelman H, Fuertes A, Dominguez J, Schmidt B (2011)Synthesis and biological evaluation of glycogen synthasekinase 3 (GSK-3) inhibitors: An fast and atom efficientaccess to 1-aryl-3-benzylureas. Bioorg Med Chem Lett 21,5610-5615.

[1312] Lo Monte F, Kramer T, Gu J, Brodrecht M, Pilakowski J,Fuertes A, Dominguez JM, Plotkin B, Eldar-FinkelmanH, Schmidt B (2012) Structure-based optimization of

Page 99: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 645

oxadiazole-based GSK-3 inhibitors. Eur J Med Chem, doi:10.1016/j.ejmech.2012.06.006 [Epubl ahead of print].

[1313] Voigt B, Krug M, Schachtele C, Totzke F, Hilgeroth A(2008) Probing novel 1-aza-9-oxafluorenes as selectiveGSK-3beta inhibitors. ChemMedChem 3, 120-126.

[1314] Testard A, Loge C, Leger B, Robert JM, Lozach O,Blairvacq M, Meijer L, Thiery V, Besson T (2006)Thiazolo[5,4-f]quinazolin-9-ones, inhibitors of glycogensynthase kinase-3. Bioorg Med Chem Lett 16, 3419-3423.

[1315] Palomo V, Soteras I, Perez DI, Perez C, Gil C, CampilloNE, Martinez A (2011) Exploring the binding sites ofglycogen synthase kinase 3. Identification and characteri-zation of allosteric modulation cavities. J Med Chem 54,8461-8470.

[1316] Palomo V, Perez DI, Perez C, Morales-Garcia JA, SoterasI, Alonso-Gil S, Encinas A, Castro A, Campillo NE,Perez-Castillo A, Gil C, Martinez A (2012) 5-imino-1,2,4-thiadiazoles: First small molecules as substratecompetitive inhibitors of glycogen synthase kinase 3. JMed Chem 55, 1645-1661.

[1317] Perez DI, Palomo V, Perez C, Gil C, Dans PD, LuqueFJ, Conde S, Martinez A (2011) Switching reversibilityto irreversibility in glycogen synthase kinase 3 inhibitors:Clues for specific design of new compounds. J Med Chem54, 4042-4056.

[1318] Perez DI, Pistolozzi M, Palomo V, Redondo M, For-tugno C, Gil C, Felix G, Martinez A, Bertucci C (2012)5-Imino-1,2-4-thiadiazoles and quinazolines derivativesas glycogen synthase kinase 3beta (GSK-3beta) andphosphodiesterase 7 (PDE7) inhibitors: Determination ofblood-brain barrier penetration and binding to humanserum albumin. Eur J Pharm Sci 45, 677-684.

[1319] Khanfar MA, Hill RA, Kaddoumi A, El Sayed KA (2010)Discovery of novel GSK-3beta inhibitors with potent invitro and in vivo activities and excellent brain permeabilityusing combined ligand- and structure-based virtual screen-ing. J Med Chem 53, 8534-8545.

[1320] Ibrahim N, Mouawad L, Legraverend M (2010) Novel 8-arylated purines as inhibitors of glycogen synthase kinase.Eur J Med Chem 45, 3389-3393.

[1321] Zou H, Zhou L, Li Y, Cui Y, Zhong H, Pan Z, Yang Z,Quan J (2010) Benzo[e]isoindole-1,3-diones as potentialinhibitors of glycogen synthase kinase-3 (GSK-3). Syn-thesis, kinase inhibitory activity, zebrafish phenotype, andmodeling of binding mode. J Med Chem 53, 994-1003.

[1322] Sondhi SM, Singh N, Kumar A, Lozach O, Meijer L (2006)Synthesis, anti-inflammatory, analgesic and kinase (CDK-1, CDK-5 and GSK-3) inhibition activity evaluation ofbenzimidazole/benzoxazole derivatives and some Schiff’sbases. Bioorg Med Chem 14, 3758-3765.

[1323] Kim HJ, Choo H, Cho YS, No KT, Pae AN (2008) NovelGSK-3beta inhibitors from sequential virtual screening.Bioorg Med Chem 16, 636-643.

[1324] Zhou X, Zhou J, Li X, Guo C, Fang T, Chen Z (2011)GSK-3beta inhibitors suppressed neuroinflammation in ratcortex by activating autophagy in ischemic brain injury.Biochem Biophys Res Commun 411, 271-275.

[1325] Caballero J, Zilocchi S, Tiznado W, Collina S, Rossi D(2011) Binding studies and quantitative structure-activityrelationship of 3-amino-1H-indazoles as inhibitors ofGSK3beta. Chem Biol Drug Des 78, 631-641.

[1326] Lo Monte F, Kramer T, Gu J, Anumala UR, Marinelli L, LaPietra V, Novellino V, Franco E, Demedts B, Van LeuvenD, Fuertes F, Dominguez A, Plotkin JM, Eldar-Finkelman

B, Schmidt H, B (2012) Identification of glycogen syn-thase kinase-3 inhibitors with a selective sting for glycogensynthase kinase-3alpha. J Med Chem 55, 4407-4424.

[1327] Rother M, Kittner B, Rudolphi K, Rossner M, Labs KH(1996) HWA 285 (propentofylline) – a new compound forthe treatment of both vascular dementia and dementia ofthe Alzheimer type. Ann N Y Acad Sci 777, 404-409.

[1328] Rother M, Erkinjuntti T, Roessner M, Kittner B, Marcus-son J, Karlsson I (1998) Propentofylline in the treatmentof Alzheimer’s disease and vascular dementia: A reviewof phase III trials. Dement Geriatr Cogn Disord 9(Suppl1), 36-43.

[1329] Mielke R, Ghaemi M, Kessler J, Kittner B, Szelies B,Herholz K, Heiss WD (1998) Propentofylline enhancescerebral metabolic response to auditory memory stimula-tion in Alzheimer’s disease. J Neurol Sci 154, 76-82.

[1330] Mielke R, Moller HJ, Erkinjuntti T, Rosenkranz B,Rother M, Kittner B (1998) Propentofylline in the treat-ment of vascular dementia and Alzheimer-type dementia:Overview of phase I and phase II clinical trials. AlzheimerDis Assoc Disord 12(Suppl 2), S29-S35.

[1331] Kittner B (1999) Clinical trials of propentofylline in vas-cular dementia. European/Canadian Propentofylline StudyGroup. Alzheimer Dis Assoc Disord 13(Suppl 3), S166-S171.

[1332] Chauhan NB, Siegel GJ, Feinstein DL (2005)Propentofylline attenuates tau hyperphosphoryla-tion in Alzheimer’s Swedish mutant model Tg2576.Neuropharmacology 48, 93-104.

[1333] Hu S, Begum AN, Jones MR, Oh MS, Beech WK, BeechBH, Yang F, Chen P, Ubeda OJ, Kim PC, Davies P,Ma Q, Cole GM, Frautschy SA (2009) GSK3 inhibitorsshow benefits in an Alzheimer’s disease (AD) model ofneurodegeneration but adverse effects in control animals.Neurobiol Dis 33, 193-206.

[1334] Komsuoglu-Celikyurt I, Gocmez SS, Mutlu O, Gacar N,Aricioglu F, Utkan T (2011) Evidence for the involvementof neuronal nitric oxide synthase and soluble guanylatecyclase on cognitive functions in rats. Life Sci 89, 905-910.

[1335] Thatcher GR, Bennett BM, Reynolds JN (2005) Nitricoxide mimetic molecules as therapeutic agents inAlzheimer’s disease. Curr Alzheimer Res 2, 171-182.

[1336] Qin Z, Luo J, Vandevrede L, Tavassoli E, Fa’ M, Teich AF,Arancio O, Thatcher GR (2012) Design and synthesis ofneuroprotective methylthiazoles and modification as NO-chimeras for neurodegenerative therapy. J Med Chem 55,6784-6801.

[1337] Smith S, Dringenberg HC, Bennett BM, Thatcher GR,Reynolds JN (2000) A novel nitrate ester reverses the cog-nitive impairment caused by scopolamine in the Morriswater maze. Neuroreport 11, 3883-3886.

[1338] Reynolds JN, Bennett BM, Boegman RJ, Jhamandas K,Ratz JD, Zavorin SI, Scutaru D, Dumitrascu A, ThatcherGR (2002) Neuroprotection against ischemic brain injuryconferred by a novel nitrate ester. Bioorg Med Chem Lett12, 2863-2866.

[1339] Thatcher GR, Bennett BM, Dringenberg HC, ReynoldsJN (2004) Novel nitrates as NO mimetics directed atAlzheimer’s disease. J Alzheimers Dis 6, S75-S84.

[1340] Thatcher GR, Bennett BM, Reynolds JN (2006) NOchimeras as therapeutic agents in Alzheimer’s disease.Curr Alzheimer Res 3, 237-245.

[1341] Bennett BM, Reynolds JN, Prusky GT, Douglas RM,Sutherland RJ, Thatcher GR (2007) Cognitive deficits inrats after forebrain cholinergic depletion are reversed by a

Page 100: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

646 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

novel NO mimetic nitrate ester. Neuropsychopharmacol-ogy 32, 505-513.

[1342] Schipper HM, Bennett DA, Liberman A, Bienias JL,Schneider JA, Kelly J, Arvanitakis Z (2006) Glial hemeoxygenase-1 expression in Alzheimer disease and mildcognitive impairment. Neurobiol Aging 27, 252-261.

[1343] Schipper HM (2007) Biomarker potential of hemeoxygenase-1 in Alzheimer’s disease and mild cognitiveimpairment. Biomark Med 1, 375-385.

[1344] Schipper HM (2011) Heme oxygenase-1 in Alzheimer dis-ease: A tribute to Moussa Youdim. J Neural Transm 118,381-387.

[1345] Hascalovici JR, Vaya J, Khatib S, Holcroft CA, ZukorH, Song W, Arvanitakis Z, Bennett DA, Schipper HM(2009) Brain sterol dysregulation in sporadic AD andMCI: Relationship to heme oxygenase-1. J Neurochem110, 1241-1253.

[1346] Hascalovici JR, Song W, Vaya J, Khatib S, FuhrmanB, Aviram M, Schipper HM (2009) Impact of hemeoxygenase-1 on cholesterol synthesis, cholesterol effluxand oxysterol formation in cultured astroglia. J Neurochem108, 72-81.

[1347] Fischer A, Sananbenesi F, Mungenast A, Tsai LH (2010)Targeting the correct HDAC(s) to treat cognitive disorders.Trends Pharmacol Sci 31, 605-617.

[1348] Chuang DM, Leng Y, Marinova Z, Kim HJ, Chiu CT(2009) Multiple roles of HDAC inhibition in neurodegen-erative conditions. Trends Neurosci 32, 591-601.

[1349] Zhang L, Sheng S, Qin C (2012) The role of HDAC6 inAlzheimer’s disease. J Alzheimers Dis, doi: 10.3233/JAD-2012-120727 [Epubl ahead of print].

[1350] Agis-Balboa RC, Pavelka Z, Kerimoglu C, Fischer A(2012) Loss of HDAC5 impairs memory function: Impli-cations for Alzheimer’s disease. J Alzheimers Dis, doi:10.3233/JAD-2012-121009 [Epubl ahead of print].

[1351] Vaisburg A, Bernstein N, Frechette S, Allan M, Abou-Khalil E, Leit S, Moradei O, Bouchain G, Wang J, WooSH, Fournel M, Yan PT, Trachy-Bourget MC, Kalita A,Beaulieu C, Li Z, Macleod AR, Besterman JM, DelormeD (2004) (2-amino-phenyl)-amides of omega-substitutedalkanoic acids as new histone deacetylase inhibitors.Bioorg Med Chem Lett 14, 283-287.

[1352] Vaisburg A, Paquin I, Bernstein N, Frechette S, Gaudette F,Leit S, Moradei O, Raeppel S, Zhou N, Bouchain G, WooSH, Jin Z, Gillespie J, Wang J, Fournel M, Yan PT, Trachy-Bourget MC, Robert MF, Lu A, Yuk J, Rahil J, MacleodAR, Besterman JM, Li Z, Delorme D (2007) N-(2-Amino-phenyl)-4-(heteroarylmethyl)-benzamides as new histonedeacetylase inhibitors. Bioorg Med Chem Lett 17, 6729-6733.

[1353] Ricobaraza A, Cuadrado-Tejedor M, Perez-Mediavilla A,Frechilla D, Del RJ, Garcia-Osta A (2009) Phenylbutyrateameliorates cognitive deficit and reduces tau pathology inan Alzheimer’s disease mouse model. Neuropsychophar-macology 34, 1721-1732.

[1354] Ricobaraza A, Cuadrado-Tejedor M, Garcia-Osta A (2011)Long-term phenylbutyrate administration prevents mem-ory deficits in Tg2576 mice by decreasing Abeta. FrontBiosci (Elite Ed) 3, 1375-1384.

[1355] Ricobaraza A, Cuadrado-Tejedor M, Marco S, Perez-Otano I, Garcia-Osta A (2012) Phenylbutyrate rescuesdendritic spine loss associated with memory deficits ina mouse model of Alzheimer disease. Hippocampus 22,1040-1050.

[1356] Wiley JC, Pettan-Brewer C, Ladiges WC (2011) Phenylbu-tyric acid reduces amyloid plaques and rescues cognitivebehavior in AD transgenic mice. Aging Cell 10, 418-428.

[1357] Cuadrado-Tejedor M, Garcia-Osta A, Ricobaraza A,Oyarzabal J, Franco R (2011) Defining the mechanism ofaction of 4-phenylbutyrate to develop a small-molecule-based therapy for Alzheimer’s disease. Curr Med Chem18, 5545-5553.

[1358] Fass DM, Reis SA, Ghosh B, Hennig KM, Joseph NF, ZhaoWN, Nieland TJ, Guan JS, Groves Kuhnle CE, Tang W,Barker DD, Mazitschek R, Schreiber SL, Tsai LH, Hag-garty SJ (2013) Crebinostat: A novel cognitive enhancerthat inhibits histone deacetylase activity and modulateschromatin-mediated neuroplasticity. Neuropharmacology61, 81-96.

[1359] Lu J, Yang C, Chen M, Ye DY, Lonser RR, Brady RO,Zhuang Z (2011) Histone deacetylase inhibitors preventthe degradation and restore the activity of glucocerebrosi-dase in Gaucher disease. Proc Natl Acad Sci U S A 108,21200-21205.

[1360] Kozikowski AP, Chen Y, Subhasish T, Lewin NE, Blum-berg PM, Zhong Z, D’Annibale MA, Wang WL, Shen Y,Langley B (2009) Searching for disease modifiers-PKCactivation and HDAC inhibition – a dual drug approach toAlzheimer’s disease that decreases Abeta production whileblocking oxidative stress. ChemMedChem 4, 1095-1105.

[1361] Solomon A, Kivipelto M, Wolozin B, Zhou J, WhitmerRA (2009) Midlife serum cholesterol and increased riskof Alzheimer’s and vascular dementia three decades later.Dement Geriatr Cogn Disord 28, 75-80.

[1362] Solomon A, Kivipelto M (2009) Cholesterol-modifyingstrategies for Alzheimer’s disease. Expert Rev Neurother9, 695-709.

[1363] Jick H, Zornberg GL, Jick SS, Seshadri S, Drachman DA(2000) Statins and the risk of dementia. Lancet 356, 1627-1631.

[1364] Wolozin B, Kellman W, Ruosseau P, Celesia GG, SiegelG (2000) Decreased prevalence of Alzheimer diseaseassociated with 3-hydroxy-3-methyglutaryl coenzyme Areductase inhibitors. Arch Neurol 57, 1439-1443.

[1365] Wolozin B, Manger J, Bryant R, Cordy J, Green RC,McKee A (2006) Re-assessing the relationship betweencholesterol, statins and Alzheimer’s disease. Acta NeurolScand Suppl 185, 63-70.

[1366] Rockwood K, Kirkland S, Hogan DB, MacKnight C,Merry H, Verreault R, Wolfson C, McDowell I (2002)Use of lipid-lowering agents, indication bias, and the riskof dementia in community-dwelling elderly people. ArchNeurol 59, 223-227.

[1367] Rockwood K (2006) Epidemiological and clinical trialsevidence about a preventive role for statins in Alzheimer’sdisease. Acta Neurol Scand Suppl 185, 71-77.

[1368] Doraiswamy PM (2002) Non-cholinergic strategies fortreating and preventing Alzheimer’s disease. CNS Drugs16, 811-824.

[1369] Whitfield JF (2006) Can statins put the brakes onAlzheimer’s disease? Expert Opin Investig Drugs 15,1479-1485.

[1370] Sparks DL (2011) Alzheimer disease: Statins in the treat-ment of Alzheimer disease. Nat Rev Neurol 7, 662-663.

[1371] Pac-Soo C, Lloyd DG, Vizcaychipi MP, Ma D (2011)Statins: The role in the treatment and prevention ofAlzheimer’s neurodegeneration. J Alzheimers Dis 27, 1-10.

Page 101: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 647

[1372] Shepardson NE, Shankar GM, Selkoe DJ (2011) Choles-terol level and statin use in Alzheimer disease: I. Review ofepidemiological and preclinical studies. Arch Neurol 68,1239-1244.

[1373] Shepardson NE, Shankar GM, Selkoe DJ (2011) Choles-terol level and statin use in Alzheimer disease: II. Reviewof human trials and recommendations. Arch Neurol 68,1385-1392.

[1374] Pani A, Mandas A, Dessi S (2010) Cholesterol,Alzheimer’s disease, prion disorders: A menage a trois?Curr Drug Targets 11, 1018-1031.

[1375] Sabbagh MN, Sparks DL (2012) Statins to treatAlzheimer’s disease: An incomplete story. Expert RevNeurother 12, 27-30.

[1376] Gamba P, Testa G, Sottero B, Gargiulo S, Poli G, Leonar-duzzi G (2012) The link between altered cholesterolmetabolism and Alzheimer’s disease. Ann N Y Acad Sci1259, 54-64.

[1377] Bettermann K, Arnold AM, Williamson J, Rapp S, SinkK, Toole JF, Carlson MC, Yasar S, Dekosky S, Burke GL(2012) Statins, risk of dementia, and cognitive function:Secondary analysis of the ginkgo evaluation of memorystudy. J Stroke Cerebrovasc Dis 21, 436-444.

[1378] Fassbender K, Simons M, Bergmann C, Stroick M, Lutjo-hann D, Keller P, Runz H, Kuhl S, Bertsch T, von BK,Hennerici M, Beyreuther K, Hartmann T (2001) Simvas-tatin strongly reduces levels of Alzheimer’s disease beta-amyloid peptides Abeta 42 and Abeta 40 in vitro and invivo. Proc Natl Acad Sci U S A 98, 5856-5861.

[1379] Tong XK, Nicolakakis N, Fernandes P, Ongali B, Brouil-lette J, Quirion R, Hamel E (2009) Simvastatin improvescerebrovascular function and counters soluble amyloid-beta, inflammation and oxidative stress in aged APP mice.Neurobiol Dis 35, 406-414.

[1380] Parvathy S, Ehrlich M, Pedrini S, Diaz N, RefoloL, Buxbaum JD, Bogush A, Petanceska S, Gandy S(2004) Atorvastatin-induced activation of Alzheimer’salpha secretase is resistant to standard inhibitors of proteinphosphorylation-regulated ectodomain shedding. J Neu-rochem 90, 1005-1010.

[1381] Piermartiri TC, Figueiredo CP, Rial D, Duarte FS,Bezerra SC, Mancini G, de Bem AF, Prediger RD,Tasca CI (2010) Atorvastatin prevents hippocampal celldeath, neuroinflammation and oxidative stress followingamyloid-beta(1-40) administration in mice: Evidence fordissociation between cognitive deficits and neuronal dam-age. Exp Neurol 226, 274-284.

[1382] Xiu J, Nordberg A, Shan KR, Yu WF, Olsson JM, NordmanT, Mousavi M, Guan ZZ (2005) Lovastatin stimulates up-regulation of alpha7 nicotinic receptors in cultured neuronswithout cholesterol dependency, a mechanism involvingproduction of the alpha-form of secreted amyloid precur-sor protein. J Neurosci Res 82, 531-541.

[1383] Xiu J, Nordberg A, Qi X, Guan ZZ (2006) Influence ofcholesterol and lovastatin on alpha-form of secreted amy-loid precursor protein and expression of alpha7 nicotinicreceptor on astrocytes. Neurochem Int 49, 459-465.

[1384] Si ML, Long C, Yang DI, Chen MF, Lee TJ (2005) Statinsprevent beta-amyloid inhibition of sympathetic alpha7-nAChR-mediated nitrergic neurogenic dilation in porcinebasilar arteries. J Cereb Blood Flow Metab 25, 1573-1585.

[1385] Mozayan M, Lee TJ (2007) Statins prevent cholinesteraseinhibitor blockade of sympathetic alpha7-nAChR-mediated currents in rat superior cervical ganglion

neurons. Am J Physiol Heart Circ Physiol 293,H1737-H1744.

[1386] Sharma B, Singh N, Singh M (2008) Modulationof celecoxib- and streptozotocin-induced experimentaldementia of Alzheimer’s disease by pitavastatin anddonepezil. J Psychopharmacol 22, 162-171.

[1387] Kurata T, Miyazaki K, Kozuki M, Panin VL, MorimotoN, Ohta Y, Nagai M, Ikeda Y, Matsuura T, Abe K (2011)Atorvastatin and pitavastatin improve cognitive functionand reduce senile plaque and phosphorylated tau in agedAPP mice. Brain Res 1371, 161-170.

[1388] Kurata T, Miyazaki K, Kozuki M, Morimoto N, Ohta Y,Ikeda Y, Abe K (2012) Atorvastatin and pitavastatin reducesenile plaques and inflammatory responses in a mousemodel of Alzheimer’s disease. Neurol Res 34, 601-610.

[1389] Bernick C, Katz R, Smith NL, Rapp S, Bhadelia R, CarlsonM, Kuller L (2005) Statins and cognitive function in theelderly: The Cardiovascular Health Study. Neurology 65,1388-1394.

[1390] Winblad B, Jelic V, Kershaw P, Amatniek J (2007)Effects of statins on cognitive function in patients withAlzheimer’s disease in galantamine clinical trials. DrugsAging 24, 57-61.

[1391] Sparks DL, Sabbagh MN, Connor DJ, Lopez J, LaunerLJ, Browne P, Wasser D, Johnson-Traver S, Lochhead J,Ziolwolski C (2005) Atorvastatin for the treatment of mildto moderate Alzheimer disease: Preliminary results. ArchNeurol 62, 753-757.

[1392] Sparks DL, Sabbagh MN, Connor DJ, Lopez J, LaunerLJ, Petanceska S, Browne P, Wassar D, Johnson-TraverS, Lochhead J, Ziolkowski C (2005) Atorvastatin ther-apy lowers circulating cholesterol but not free radicalactivity in advance of identifiable clinical benefit in thetreatment of mild-to-moderate AD. Curr Alzheimer Res 2,343-353.

[1393] Sparks DL, Connor DJ, Sabbagh MN, Petersen RB,Lopez J, Browne P (2006) Circulating cholesterol levels,apolipoprotein E genotype and dementia severity influ-ence the benefit of atorvastatin treatment in Alzheimer’sdisease: Results of the Alzheimer’s Disease Cholesterol-Lowering Treatment (ADCLT) trial. Acta Neurol ScandSuppl 185, 3-7.

[1394] Jones RW, Kivipelto M, Feldman H, Sparks L, DoodyR, Waters DD, Hey-Hadavi J, Breazna A, SchindlerRJ, Ramos H (2008) The Atorvastatin/Donepezil inAlzheimer’s Disease Study (LEADe): Design and baselinecharacteristics. Alzheimers Dement 4, 145-153.

[1395] Feldman HH, Doody RS, Kivipelto M, Sparks DL, WatersDD, Jones RW, Schwam E, Schindler R, Hey-Hadavi J,DeMicco DA, Breazna A (2010) Randomized controlledtrial of atorvastatin in mild to moderate Alzheimer disease:LEADe. Neurology 74, 956-964.

[1396] Trompet S, van VP, de Craen AJ, Jolles J, Buckley BM,Murphy MB, Ford I, Macfarlane PW, Sattar N, Packard CJ,Stott DJ, Shepherd J, Bollen EL, Blauw GJ, Jukema JW,Westendorp RG (2010) Pravastatin and cognitive functionin the elderly. Results of the PROSPER study. J Neurol257, 85-90.

[1397] Sano M, Bell KL, Galasko D, Galvin JE, Thomas RG, vanDyck CH, Aisen PS (2011) A randomized, double-blind,placebo-controlled trial of simvastatin to treat Alzheimerdisease. Neurology 77, 556-563.

[1398] Sjogren M, Gustafsson K, Syversen S, Olsson A, Edman A,Davidsson P, Wallin A, Blennow K (2003) Treatment withsimvastatin in patients with Alzheimer’s disease lowers

Page 102: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

648 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

both alpha- and beta-cleaved amyloid precursor protein.Dement Geriatr Cogn Disord 16, 25-30.

[1399] Hoglund K, Syversen S, Lewczuk P, Wallin A, Wiltfang J,Blennow K (2005) Statin treatment and a disease-specificpattern of beta-amyloid peptides in Alzheimer’s disease.Exp Brain Res 164, 205-214.

[1400] Hoglund K, Thelen KM, Syversen S, Sjogren M, von BK,Wallin A, Vanmechelen E, Vanderstichele H, LutjohannD, Blennow K (2005) The effect of simvastatin treatmenton the amyloid precursor protein and brain cholesterolmetabolism in patients with Alzheimer’s disease. DementGeriatr Cogn Disord 19, 256-265.

[1401] Hoglund K, Wallin A, Blennow K (2006) Effect of statinson beta-amyloid metabolism in humans: Potential impor-tance for the development of senile plaques in Alzheimer’sdisease. Acta Neurol Scand Suppl 185, 87-92.

[1402] Hoglund K, Blennow K (2007) Effect of HMG-CoAreductase inhibitors on beta-amyloid peptide levels: Impli-cations for Alzheimer’s disease. CNS Drugs 21, 449-462.

[1403] Carlsson CM, Gleason CE, Hess TM, Moreland KA,Blazel HM, Koscik RL, Schreiber NT, Johnson SC,Atwood CS, Puglielli L, Hermann BP, McBride PE, SteinJH, Sager MA, Asthana S (2008) Effects of simvastatin oncerebrospinal fluid biomarkers and cognition in middle-aged adults at risk for Alzheimer’s disease. J AlzheimersDis 13, 187-197.

[1404] Mans RA, Chowdhury N, Cao D, McMahon LL, LiL (2010) Simvastatin enhances hippocampal long-termpotentiation in C57BL/6 mice. Neuroscience 166, 435-444.

[1405] Mans RA, McMahon LL, Li L (2012) Simvastatin-mediated enhancement of long-term potentiation is drivenby farnesyl-pyrophosphate depletion and inhibition of far-nesylation. Neuroscience 202, 1-9.

[1406] Mans RA, McMahon LL, Li L (2012) Molecular mecha-nisms for simvastatin-mediated enhancement of synapticplasticity. Mol Neurodegener 7(Suppl 1), O4.

[1407] Campoy S, Sierra S, Suarez B, Ramos MC, Velasco J,Burgos JS, Adrio JL (2010) Semisynthesis of novelmonacolin J derivatives: Hypocholesterolemic and neu-roprotective activities. J Antibiot (Tokyo) 63, 499-505.

[1408] Messier C, Teutenberg K (2005) The role of insulin, insulingrowth factor, and insulin-degrading enzyme in brain agingand Alzheimer’s disease. Neural Plast 12, 311-328.

[1409] Zhao Z, Xiang Z, Haroutunian V, Buxbaum JD, StetkaB, Pasinetti GM (2007) Insulin degrading enzyme activ-ity selectively decreases in the hippocampal formation ofcases at high risk to develop Alzheimer’s disease. Neuro-biol Aging 28, 824-830.

[1410] Wang S, Wang R, Chen L, Bennett DA, Dickson DW,Wang DS (2010) Expression and functional profilingof neprilysin, insulin-degrading enzyme, and endothelin-converting enzyme in prospectively studied elderly andAlzheimer’s brain. J Neurochem 115, 47-57.

[1411] Liu Z, Zhu H, Fang GG, Walsh K, Mwamburi M, WolozinB, bdul-Hay SO, Ikezu T, Leissring MA, Qiu WQ (2012)Characterization of insulin degrading enzyme and otheramyloid-beta degrading proteases in human serum: A rolein Alzheimer’s disease? J Alzheimers Dis 29, 329-340.

[1412] Savage MJ, Gingrich DE (2009) Advances in the devel-opment of kinase inhibitor therapeutics for Alzheimer’sdisease. Drug Dev Res 70, 125-144.

[1413] Jiang X, Tian Q, Wang Y, Zhou XW, Xie JZ, WangJZ, Zhu LQ (2011) Acetyl-L-carnitine ameliorates spatial

memory deficits induced by inhibition of phosphoinositol-3 kinase and protein kinase C. J Neurochem 118, 864-878.

[1414] Filiz G, Caragounis A, Bica L, Du T, Masters CL,Crouch PJ, White AR (2008) Clioquinol inhibitsperoxide-mediated toxicity through up-regulation ofphosphoinositol-3-kinase and inhibition of p53 activity.Int J Biochem Cell Biol 40, 1030-1042.

[1415] Piette F, Belmin J, Vincent H, Schmidt N, Pariel S,Verny M, Marquis C, Mely J, Hugonot-Diener L, KinetJP, Dubreuil P, Moussy A, Hermine O (2011) Masitinibas an adjunct therapy for mild-to-moderate Alzheimer’sdisease: A randomised, placebo-controlled phase 2 trial.Alzheimers Res Ther 3, 16.

[1416] Dubreuil P, Letard S, Ciufolini M, Gros L, Humbert M,Casteran N, Borge L, Hajem B, Lermet A, Sippl W, Vois-set E, Arock M, Auclair C, Leventhal PS, Mansfield CD,Moussy A, Hermine O (2009) Masitinib (AB1010), apotent and selective tyrosine kinase inhibitor targetingKIT. PLoS One 4, e7258.

[1417] MacGregor DG, Mallon AP, Harvey AL, Young L,Nimmo HG, Stone TW (2007) Group S8A serineproteases, including a novel enzyme cadeprin, inducelong-lasting, metabotropic glutamate receptor-dependentsynaptic depression in rat hippocampal slices. Eur J Neu-rosci 26, 1870-1880.

[1418] Ramsden N, Perrin J, Ren Z, Lee BD, Zinn N, Dawson VL,Tam D, Bova M, Lang M, Drewes G, Bantscheff M, BardF, Dawson TM, Hopf C (2011) Chemoproteomics-baseddesign of potent LRRK2-selective lead compounds thatattenuate Parkinson’s disease-related toxicity in humanneurons. ACS Chem Biol 6, 1021-1028.

[1419] Dhawan G, Combs CK (2012) Inhibition of Src kinaseactivity attenuates amyloid associated microgliosis in amurine model of Alzheimer’s disease. J Neuroinflamma-tion 9, 117.

[1420] Huentelman MJ, Stephan DA, Talboom J, CorneveauxJJ, Reiman DM, Gerber JD, Barnes CA, Alexander GE,Reiman EM, Bimonte-Nelson HA (2009) Peripheral deliv-ery of a ROCK inhibitor improves learning and workingmemory. Behav Neurosci 123, 218-223.

[1421] Schneider A, Huentelman MJ, Kremerskothen J, DuningK, Spoelgen R, Nikolich K (2010) KIBRA: A new gatewayto learning and memory? Front Aging Neurosci 2, 4.

[1422] Castro-Alvarez JF, Gutierrez-Vargas J, Darnaudery M,Cardona-Gomez GP (2011) ROCK inhibition prevents tauhyperphosphorylation and p25/CDK5 increase after globalcerebral ischemia. Behav Neurosci 125, 465-472.

[1423] Corneveaux JJ, Liang WS, Reiman EM, Webster JA,Myers AJ, Zismann VL, Joshipura KD, Pearson JV, Hu-Lince D, Craig DW, Coon KD, Dunckley T, Bandy D, LeeW, Chen K, Beach TG, Mastroeni D, Grover A, Ravid R,Sando SB, Aasly JO, Heun R, Jessen F, Kolsch H, RogersJ, Hutton ML, Melquist S, Petersen RC, Alexander GE,Caselli RJ, Papassotiropoulos A, Stephan DA, HuentelmanMJ (2010) Evidence for an association between KIBRAand late-onset Alzheimer’s disease. Neurobiol Aging 31,901-909.

[1424] Hofmann SG, Smits JA, Asnaani A, Gutner CA, Otto MW(2011) Cognitive enhancers for anxiety disorders. Phar-macol Biochem Behav 99, 275-284.

[1425] Liu M, Poulose S, Schuman E, Zaitsev AD, Dobson B,Auerbach K, Seyb K, Cuny GD, Glicksman MA, Stein RL,Yue Z (2010) Development of a mechanism-based high-throughput screen assay for leucine-rich repeat kinase

Page 103: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 649

2–discovery of LRRK2 inhibitors. Anal Biochem 404, 186-192.

[1426] Chen H, Chan BK, Drummond J, Estrada AA, Gunzner-Toste J, Liu X, Liu Y, Moffat J, Shore D, Sweeney ZK, TranT, Wang S, Zhao G, Zhu H, Burdick DJ (2012) Discoveryof selective LRRK2 inhibitors guided by computationalanalysis and molecular modeling. J Med Chem 55, 5536-5545.

[1427] Echeverria V, Burgess S, Gamble-George J, Zeitlin R, LinX, Cao C, Arendash GW (2009) Sorafenib inhibits nuclearfactor kappa B, decreases inducible nitric oxide synthaseand cyclooxygenase-2 expression, and restores work-ing memory in APPswe mice. Neuroscience 162, 1220-1231.

[1428] Echeverria V, Burgess S, Gamble-George J, Arendash GW,Citron BA (2008) Raf inhibition protects cortical cellsagainst beta-amyloid toxicity. Neurosci Lett 444, 92-96.

[1429] Gingras K, Avedissian H, Thouin E, Boulanger V,Essagian C, McKerracher L, Lubell WD (2004)Synthesis and evaluation of 4-(1-aminoalkyl)-N-(4-pyridyl)cyclohexanecarboxamides as Rho kinaseinhibitors and neurite outgrowth promoters. Bioorg MedChem Lett 14, 4931-4934.

[1430] Page TH, Brown A, Timms EM, Foxwell BM, Ray KP(2010) Inhibitors of p38 suppress cytokine production inrheumatoid arthritis synovial membranes: Does variableinhibition of interleukin-6 production limit effectivenessin vivo? Arthritis Rheum 62, 3221-3231.

[1431] Baran H, Jellinger K, Deecke L (1999) Kynureninemetabolism in Alzheimer’s disease. J Neural Transm 106,165-181.

[1432] Potter MC, Elmer GI, Bergeron R, Albuquerque EX,Guidetti P, Wu HQ, Schwarcz R (2010) Reduction ofendogenous kynurenic acid formation enhances extra-cellular glutamate, hippocampal plasticity, and cognitivebehavior. Neuropsychopharmacology 35, 1734-1742.

[1433] Rossi F, Valentina C, Garavaglia S, Sathyasaikumar KV,Schwarcz R, Kojima S, Okuwaki K, Ono S, Kajii Y,Rizzi M (2010) Crystal structure-based selective targetingof the pyridoxal 5′-phosphate dependent enzyme kynure-nine aminotransferase II for cognitive enhancement. J MedChem 53, 5684-5689.

[1434] Dounay AB, Anderson M, Vechle BM, Campbell BM,Claffey MM, Evdokimov A, Evrard E, Fonseca KR, GanXM, Ghosh S, Hayward MM, Horner W, Kim JY, McAllis-ter LA, Pandit J, Paradis V, Parikh VD, Reese MR, Rong S,Salafia MA, Schuyten K, Strick CA, Tuttle JB, Valentine J,Wang H, Zawadzke LE, Verhoest PR (2012) Discovery ofbrain-penetrant, irreversible kynurenine aminotransferaseII inhibitors for schizophrenia. ACS Med Chem Lett 3,187-192.

[1435] Radmark O, Werz O, Steinhilber D, Samuelsson B (2007)5-Lipoxygenase: Regulation of expression and enzymeactivity. Trends Biochem Sci 32, 332-341.

[1436] Firuzi O, Zhuo J, Chinnici CM, Wisniewski T, Pratico D(2008) 5-Lipoxygenase gene disruption reduces amyloid-beta pathology in a mouse model of Alzheimer’s disease.FASEB J 22, 1169-1178.

[1437] Chu J, Pratico D (2011) 5-lipoxygenase as an endogenousmodulator of amyloid beta formation in vivo. Ann Neurol69, 34-46.

[1438] Chu J, Pratico D (2011) Pharmacologic blockade of5-lipoxygenase improves the amyloidotic phenotype of anAlzheimer’s disease transgenic mouse model involvementof gamma-secretase. Am J Pathol 178, 1762-1769.

[1439] Hashimoto K (2011) Can minocycline prevent the onset ofAlzheimer’s disease? Ann Neurol 69, 739-740.

[1440] Song Y, Wei EQ, Zhang WP, Ge QF, Liu JR, Wang ML,Huang XJ, Hu X, Chen Z (2006) Minocycline protectsPC12 cells against NMDA-induced injury via inhibiting5-lipoxygenase activation. Brain Res 1085, 57-67.

[1441] Choi Y, Kim HS, Shin KY, Kim EM, Kim M, Kim HS,Park CH, Jeong YH, Yoo J, Lee JP, Chang KA, KimS, Suh YH (2007) Minocycline attenuates neuronal celldeath and improves cognitive impairment in Alzheimer’sdisease models. Neuropsychopharmacology 32, 2393-2404.

[1442] Burgos-Ramos E, Puebla-Jimenez L, rilla-Ferreiro E(2008) Minocycline provides protection against beta-amyloid(25-35)-induced alterations of the somatostatinsignaling pathway in the rat temporal cortex. Neuroscience154, 1458-1466.

[1443] Burgos-Ramos E, Puebla-Jimenez L, rilla-FerreiroE (2009) Minocycline prevents Abeta(25-35)-inducedreduction of somatostatin and neprilysin content in rattemporal cortex. Life Sci 84, 205-210.

[1444] Seabrook TJ, Jiang L, Maier M, Lemere CA (2006)Minocycline affects microglia activation, Abeta deposi-tion, and behavior in APP-tg mice. Glia 53, 776-782.

[1445] Fan R, Xu F, Previti ML, Davis J, Grande AM, Robin-son JK, Van Nostrand WE (2007) Minocycline reducesmicroglial activation and improves behavioral deficits ina transgenic model of cerebral microvascular amyloid. JNeurosci 27, 3057-3063.

[1446] Parachikova A, Vasilevko V, Cribbs DH, LaFerla FM,Green KN (2010) Reductions in amyloid-beta-derivedneuroinflammation, with minocycline, restore cognitionbut do not significantly affect tau hyperphosphorylation.J Alzheimers Dis 21, 527-542.

[1447] Kreutzmann P, Wolf G, Kupsch K (2010) Minocyclinerecovers MTT-formazan exocytosis impaired by amyloidbeta peptide. Cell Mol Neurobiol 30, 979-984.

[1448] Ferretti MT, Allard S, Partridge V, Ducatenzeiler A, CuelloAC (2012) Minocycline corrects early, pre-plaque neuroin-flammation and inhibits BACE-1 in a transgenic model ofAlzheimer’s disease-like amyloid pathology. J Neuroin-flammation 9, 62.

[1449] Noble W, Garwood C, Stephenson J, Kinsey AM, HangerDP, Anderton BH (2009) Minocycline reduces the devel-opment of abnormal tau species in models of Alzheimer’sdisease. FASEB J 23, 739-750.

[1450] Noble W, Garwood CJ, Hanger DP (2009) Minocycline asa potential therapeutic agent in neurodegenerative disor-ders characterised by protein misfolding. Prion 3, 78-83.

[1451] Garwood CJ, Cooper JD, Hanger DP, Noble W (2010)Anti-inflammatory impact of minocycline in a mousemodel of tauopathy. Front Psychiatr 1, 136.

[1452] Chaudhry IB, Hallak J, Husain N, Minhas F, StirlingJ, Richardson P, Dursun S, Dunn G, Deakin B(2012) Minocycline benefits negative symptoms inearly schizophrenia: A randomised double-blind placebo-controlled clinical trial in patients on standard treatment.J Psychopharmacol 26, 1185-1193.

[1453] Hieke M, Rodl CB, Wisniewska JM, la BE, StarkH, Schubert-Zsilavecz M, Steinhilber D, HofmannB, Proschak E (2012) SAR-study on a newclass of imidazo[1,2-a]pyridine-based inhibitors of5-lipoxygenase. Bioorg Med Chem Lett 22, 1969-1975.

[1454] Pisani L, Catto M, Leonetti F, Nicolotti O, Stefanachi A,Campagna F, Carotti A (2011) Targeting monoamine oxi-

Page 104: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

650 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

dases with multipotent ligands: An emerging strategy inthe search of new drugs against neurodegenerative dis-eases. Curr Med Chem 18, 4568-4587.

[1455] Carradori S, Secci D, Bolasco A, Chimenti P, D’AscenzioM (2012) Patent-related survey on new monoamine oxi-dase inhibitors and their therapeutic potential. Expert OpinTher Pat 22, 759-801.

[1456] Kennedy BP, Ziegler MG, Alford M, Hansen LA, ThalLJ, Masliah E (2003) Early and persistent alterations inprefrontal cortex MAO A and B in Alzheimer’s disease. JNeural Transm 110, 789-801.

[1457] Gotz ME, Fischer P, Gsell W, Riederer P, Streifler M,Simanyi M, Muller F, Danielczyk W (1998) Plateletmonoamine oxidase B activity in dementia. A 4-yearfollow-up. Dement Geriatr Cogn Disord 9, 74-77.

[1458] Zheng H, Weiner LM, Bar-Am O, Epsztejn S, CabantchikZI, Warshawsky A, Youdim MB, Fridkin M (2005)Design, synthesis, and evaluation of novel bifunctionaliron-chelators as potential agents for neuroprotection inAlzheimer’s, Parkinson’s, and other neurodegenerativediseases. Bioorg Med Chem 13, 773-783.

[1459] Gal S, Fridkin M, Amit T, Zheng H, Youdim MB (2006)M30, a novel multifunctional neuroprotective drug withpotent iron chelating and brain selective monoamineoxidase-ab inhibitory activity for Parkinson’s disease. JNeural Transm Suppl 70, 447-456.

[1460] Youdim MB (2006) The path from anti Parkinson drugselegiline and rasagiline to multifunctional neuropro-tective anti Alzheimer drugs ladostigil and m30. CurrAlzheimer Res 3, 541-550.

[1461] Avramovich-Tirosh Y, Amit T, Bar-Am O, Zheng H,Fridkin M, Youdim MB (2007) Therapeutic targets andpotential of the novel brain- permeable multifunctionaliron chelator-monoamine oxidase inhibitor drug, M-30,for the treatment of Alzheimer’s disease. J Neurochem 100,490-502.

[1462] Avramovich-Tirosh Y, Reznichenko L, Mit T, Zheng H,Fridkin M, Weinreb O, Mandel S, Youdim MB (2007) Neu-rorescue activity, APP regulation and amyloid-beta peptidereduction by novel multi-functional brain permeable iron-chelating- antioxidants, M-30 and green tea polyphenol,EGCG. Curr Alzheimer Res 4, 403-411.

[1463] Weinreb O, Mandel S, Bar-Am O, Amit T (2011)Iron-chelating backbone coupled with monoamine oxi-dase inhibitory moiety as novel pluripotential therapeuticagents for Alzheimer’s disease: A tribute to MoussaYoudim. J Neural Transm 118, 479-492.

[1464] Johnson S, Tazik S, Lu D, Johnson C, Youdim MB, WangJ, Rajkowska G, Ou XM (2010) The new inhibitor ofmonoamine oxidase, M30, has a neuroprotective effectagainst dexamethasone-induced brain cell apoptosis. FrontNeurosci 4, 180.

[1465] Zheng H, Fridkin M, Youdim MB (2010) Site-activatedchelators derived from anti-Parkinson drug rasagiline as apotential safer and more effective approach to the treatmentof Alzheimer’s disease. Neurochem Res 35, 2117-2123.

[1466] Sterling J, Veinberg A, Lerner D, Goldenberg W, LevyR, Youdim M, Finberg J (1998) (R)(+)-N-propargyl-1-aminoindan (rasagiline) and derivatives: Highly selectiveand potent inhibitors of monoamine oxidase B. J NeuralTransm Suppl 52, 301-305.

[1467] Youdim MB, Bar AO, Yogev-Falach M, Weinreb O,Maruyama W, Naoi M, Amit T (2005) Rasagiline:Neurodegeneration, neuroprotection, and mitochondrialpermeability transition. J Neurosci Res 79, 172-179.

[1468] Oldfield V, Keating GM, Perry CM (2007) Rasagiline: Areview of its use in the management of Parkinson’s disease.Drugs 67, 1725-1747.

[1469] Oldfield V, Keating GM, Perry CM (2008) Spotlight onrasagiline in Parkinson’s disease. CNS Drugs 22, 83-86.

[1470] Olanow CW, Hauser RA, Jankovic J, Langston W, Lang A,Poewe W, Tolosa E, Stocchi F, Melamed E, Eyal E, RascolO (2008) A randomized, double-blind, placebo-controlled,delayed start study to assess rasagiline as a disease modi-fying therapy in Parkinson’s disease (the ADAGIO study):Rationale, design, and baseline characteristics. Mov Dis-ord 23, 2194-2201.

[1471] Olanow CW, Rascol O, Hauser R, Feigin PD, JankovicJ, Lang A, Langston W, Melamed E, Poewe W, StocchiF, Tolosa E (2009) A double-blind, delayed-start trial ofrasagiline in Parkinson’s disease. N Engl J Med 361, 1268-1278.

[1472] Mehta SH, Morgan JC, Sethi KD (2010) Does rasagilinehave a disease-modifying effect on Parkinson’s disease?Curr Neurol Neurosci Rep 10, 413-416.

[1473] Chahine LM, Stern MB (2011) Rasagiline in Parkinson’sdisease. Int Rev Neurobiol 100, 151-168.

[1474] Perez-Lloret S, Rascol O (2011) Safety of rasagiline forthe treatment of Parkinson’s disease. Expert Opin DrugSaf 10, 633-643.

[1475] Rascol O, Fitzer-Attas CJ, Hauser R, Jankovic J, Lang A,Langston JW, Melamed E, Poewe W, Stocchi F, Tolosa E,Eyal E, Weiss YM, Olanow CW (2011) A double-blind,delayed-start trial of rasagiline in Parkinson’s disease (theADAGIO study): Prespecified and post-hoc analyses of theneed for additional therapies, changes in UPDRS scores,and non-motor outcomes. Lancet Neurol 10, 415-423.

[1476] Silver DE, Buck PO (2011) Determining the efficacy ofrasagiline in reducing bradykinesia among Parkinson’sdisease patients: A review. Int J Neurosci 121, 485-489.

[1477] Tolosa E, Stern MB (2012) Efficacy, safety and tolerabilityof rasagiline as adjunctive therapy in elderly patients withParkinson’s disease. Eur J Neurol 19, 258-264.

[1478] Weinreb O, Amit T, Riederer P, Youdim MB, MandelSA (2011) Neuroprotective profile of the multitarget drugrasagiline in Parkinson’s disease. Int Rev Neurobiol 100,127-149.

[1479] Hanagasi HA, Gurvit H, Unsalan P, Horozoglu H, TuncerN, Feyzioglu A, Gunal DI, Yener GG, Cakmur R, SahinHA, Emre M (2011) The effects of rasagiline on cognitivedeficits in Parkinson’s disease patients without dementia:A randomized, double-blind, placebo-controlled, multi-center study. Mov Disord 26, 1851-1858.

[1480] Youdim MB, Maruyama W, Naoi M (2005) Neurophar-macological, neuroprotective and amyloid precursorprocessing properties of selective MAO-B inhibitorantiparkinsonian drug, rasagiline. Drugs Today (Barc ) 41,369-391.

[1481] Wong FK, Lee SH, Atcha Z, Ong AB, Pemberton DJ, ChenWS (2010) Rasagiline improves learning and memory inyoung healthy rats. Behav Pharmacol 21, 278-282.

[1482] Leonetti F, Capaldi C, Pisani L, Nicolotti O, MuncipintoG, Stefanachi A, Cellamare S, Caccia C, Carotti A (2007)Solid-phase synthesis and insights into structure-activityrelationships of safinamide analogues as potent and selec-tive inhibitors of type B monoamine oxidase. J Med Chem50, 4909-4916.

[1483] Pevarello P, Bonsignori A, Dostert P, Heidempergher F,Pinciroli V, Colombo M, McArthur RA, Salvati P, Post C,Fariello RG, Varasi M (1998) Synthesis and anticonvulsant

Page 105: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 651

activity of a new class of 2-[(arylalky)amino]alkanamidederivatives. J Med Chem 41, 579-590.

[1484] Zhang K, Xue N, Shi X, Liu W, Meng J, Du Y (2011)A validated chiral liquid chromatographic method for theenantiomeric separation of safinamide mesilate, a newanti-Parkinson drug. J Pharm Biomed Anal 55, 220-224.

[1485] Strolin Benedetti MS, Marrari P, Colombo M, Castelli MG,Arand M, Oesch F, Dostert P (1994) The anticonvulsantFCE 26743 is a selective and short-acting MAO-B inhibitordevoid of inducing properties towards cytochrome P450-dependent testosterone hydroxylation in mice and rats. JPharm Pharmacol 46, 814-819.

[1486] Strolin Benedetti M, Tocchetti P, Rocchetti M, MartignoniM, Marrari P, Poggesi I, Dostert P (1995) Enantioselectiverecognition of two anticonvulsants, FCE 26743 and FCE28073, by MAO, and relationship between MAO-B inhi-bition and FCE 26743 concentrations in rat brain. ProgBrain Res 106, 123-134.

[1487] Fariello RG, McArthur RA, Bonsignori A, Cervini MA,Maj R, Marrari P, Pevarello P, Wolf HH, Woodhead JW,White HS, Varasi M, Salvati P, Post C (1998) Preclinicalevaluation of PNU-151774E as a novel anticonvulsant. JPharmacol Exp Ther 285, 397-403.

[1488] Chazot PL (2001) Safinamide (Newron Pharmaceuticals).Curr Opin Investig Drugs 2, 809-813.

[1489] Chazot PL (2007) Safinamide for the treatment of Parkin-son’s disease, epilepsy and restless legs syndrome. CurrOpin Investig Drugs 8, 570-579.

[1490] Fariello RG (2007) Safinamide. Neurotherapeutics 4, 110-116.

[1491] Onofrj M, Bonanni L, Thomas A (2008) An expert opinionon safinamide in Parkinson’s disease. Expert Opin InvestigDrugs 17, 1115-1125.

[1492] Naoi M, Nomura Y, Ishiki R, Suzuki H, Nagatsu T (1988)4-(O-benzylphenoxy)-N-methylbutylamine (bifemelane)and other 4-(O-benzylphenoxy)-N-methylalkylamines asnew inhibitors of type A and B monoamine oxidase. JNeurochem 50, 243-247.

[1493] Yoshida Y, Nakane A, Morita M, Kudo Y (2006) A noveleffect of bifemelane, a nootropic drug, on intracellularCa2+ levels in rat cerebral astrocytes. J Pharmacol Sci100, 126-132.

[1494] Cesura AM, Galva MD, Imhof R, Kyburz E, Picotti GB,Da PM (1989) [3H]Ro 19-6327: A reversible ligand andaffinity labelling probe for monoamine oxidase-B. Eur JPharmacol 162, 457-465.

[1495] Cesura AM, Borroni E, Gottowik J, Kuhn C, Malherbe P,Martin J, Richards JG (1999) Lazabemide for the treat-ment of Alzheimer’s disease: Rationale and therapeuticperspectives. Adv Neurol 80, 521-528.

[1496] Henriot S, Kuhn C, Kettler R, Da PM (1994) Lazabemide(Ro 19-6327), a reversible and highly sensitive MAO-B inhibitor: Preclinical and clinical findings. J NeuralTransm Suppl 41, 321-325.

[1497] Janssens de Varebeke P, Schallauer E, Rausch WD,Riederer P, Youdim MB (1990) Milacemide, the selec-tive substrate and enzyme-activated specific inhibitor ofmonoamine oxidase B, increases dopamine but not sero-tonin in caudate nucleus of rhesus monkey. Neurochem Int17, 325-329.

[1498] Dysken MW, Mendels J, LeWitt P, Reisberg B, PomaraN, Wood J, Skare S, Fakouhi JD, Herting RL (1992)Milacemide: A placebo-controlled study in senile demen-tia of the Alzheimer type. J Am Geriatr Soc 40, 503-506.

[1499] Finkelstein JE, Hengemihle JM, Ingram DK, Petri HL(1994) Milacemide treatment in mice enhances acquisi-tion of a Morris-type water maze task. Pharmacol BiochemBehav 49, 707-710.

[1500] Dorfmueller HC, van Aalten DM (2010) Screening-baseddiscovery of drug-like O-GlcNAcase inhibitor scaffolds.FEBS Lett 584, 694-700.

[1501] Dorfmueller HC, Borodkin VS, Blair DE, Pathak S,Navratilova I, van Aalten DM (2011) Substrate and prod-uct analogues as human O-GlcNAc transferase inhibitors.Amino Acids 40, 781-792.

[1502] Schimpl M, Schuttelkopf AW, Borodkin VS, van AaltenDM (2010) Human OGA binds substrates in a conservedpeptide recognition groove. Biochem J 432, 1-7.

[1503] Gay LM, Zheng X, van Aalten DM (2011) Molecularrecognition: O-GlcNAc transfer: Size matters. Nat ChemBiol 7, 134-135.

[1504] Jang H, Kim TW, Yoon S, Choi SY, Kang TW, Kim SY,Kwon YW, Cho EJ, Youn HD (2012) O-GlcNAc regulatespluripotency and reprogramming by directly acting on corecomponents of the pluripotency network. Cell Stem Cell11, 62-74.

[1505] Dorfmueller HC, Borodkin VS, Schimpl M, Shepherd SM,Shpiro NA, van Aalten DM (2006) GlcNAcstatin: A pico-molar, selective O-GlcNAcase inhibitor that modulatesintracellular O-glcNAcylation levels. J Am Chem Soc 128,16484-16485.

[1506] Dorfmueller HC, Borodkin VS, Schimpl M, van Aal-ten DM (2009) GlcNAcstatins are nanomolar inhibitorsof human O-GlcNAcase inducing cellular hyper-O-GlcNAcylation. Biochem J 420, 221-227.

[1507] Borodkin VS, van Aalten DM (2010) An effi-cient and versatile synthesis of GlcNAcstatins-potentand selective O-GlcNAcase inhibitors built on thetetrahydroimidazo[1,2-a]pyridine scaffold. Tetrahedron66, 7838-7849.

[1508] Kim C, Nam DW, Park SY, Song H, Hong HS,Boo JH, Jung ES, Kim Y, Baek JY, Kim KS,Cho JW, Mook-Jung I (2012) O-linked beta-N-acetylglucosaminidase inhibitor attenuates beta-amyloidplaque and rescues memory impairment. Neurobiol Aging,doi:10.1016/j.neurobiolaging.2012.03.001 [Epubl aheadof print].

[1508] [5] Kim C, Nam DW, Park SY, Song H, HongHS, Boo JH, Jung ES, Kim Y, Baek JY, KimKS, Cho JW, Mook-Jung I (2012) O-linked beta-N-acetylglucosaminidase inhibitor attenuates beta-amyloidplaque and rescues memory impairment. Neurobiol Aging,doi:10.1016/j.neurobiolaging.2012.03.001

[1508] [6] Macauley MS, Whitworth GE, Debowski AW, Chin D,Vocadlo DJ (2005) O-GlcNAcase uses substrate-assistedcatalysis: kinetic analysis and development of highly selec-tive mechanism-inspired inhibitors. J Biol Chem 280,25313-25322.

[1509] Yuzwa SA, Macauley MS, Heinonen JE, Shan X,Dennis RJ, He Y, Whitworth GE, Stubbs KA, McEach-ern EJ, Davies GJ, Vocadlo DJ (2008) A potentmechanism-inspired O-GlcNAcase inhibitor that blocksphosphorylation of tau in vivo. Nat Chem Biol 4, 483-490.

[1510] Yuzwa SA, Vocadlo DJ (2009) O-GlcNAc modificationand the tauopathies: Insights from chemical biology. CurrAlzheimer Res 6, 451-454.

[1511] Yuzwa SA, Yadav AK, Skorobogatko Y, Clark T, VossellerK, Vocadlo DJ (2011) Mapping O-GlcNAc modification

Page 106: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

652 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

sites on tau and generation of a site-specific O-GlcNAc tauantibody. Amino Acids 40, 857-868.

[1512] Yuzwa SA, Shan X, Macauley MS, Clark T, SkorobogatkoY, Vosseller K, Vocadlo DJ (2012) Increasing O-GlcNAcslows neurodegeneration and stabilizes tau against aggre-gation. Nat Chem Biol 8, 393-399.

[1513] Fischer PM (2008) Turning down tau phosphorylation. NatChem Biol 4, 448-449.

[1514] Lefebvre T (2012) Recall sugars, forget Alzheimer’s.Nature Chem Biol 8, 325-326.

[1515] Yu Y, Zhang L, Li X, Run X, Liang Z, Li Y, LiuY, Lee MH, Grundke-Iqbal I, Iqbal K, Vocadlo DJ,Liu F, Gong CX (2012) Differential effects of an O-GlcNAcase inhibitor on tau phosphorylation. PLoS One 7,e35277.

[1516] Mouri A, Noda Y, Shimizu S, Tsujimoto Y, Nabeshima T(2010) The role of cyclophilin D in learning and memory.Hippocampus 20, 293-304.

[1517] Muirhead KE, Borger E, Aitken L, Conway SJ, Gunn-Moore FJ (2010) The consequences of mitochondrialamyloid beta-peptide in Alzheimer’s disease. Biochem J426, 255-270.

[1518] Koren J III, Jinwal UK, Davey Z, Kiray J, Arulselvam K,Dickey CA (2011) Bending tau into shape: The emerg-ing role of peptidyl-prolyl isomerases in tauopathies. MolNeurobiol 44, 65-70.

[1519] Golde TE (2006) Disease modifying therapy for AD?J Neurochem 99, 689-707.

[1520] Reneerkens OA, Rutten K, Steinbusch HW, Blok-land A, Prickaerts J (2009) Selective phosphodiesteraseinhibitors: A promising target for cognition enhancement.Psychopharmacology (Berl) 202, 419-443.

[1521] Rutten K, Van Donkelaar EL, Ferrington L, BloklandA, Bollen E, Steinbusch HW, Kelly PA, Prickaerts JH(2009) Phosphodiesterase inhibitors enhance object mem-ory independent of cerebral blood flow and glucoseutilization in rats. Neuropsychopharmacology 34, 1914-1925.

[1522] Schmidt CJ (2010) Phosphodiesterase inhibitors as poten-tial cognition enhancing agents. Curr Top Med Chem 10,222-230.

[1523] Blokland A, Menniti FS, Prickaerts J (2012) PDE inhibi-tion and cognition enhancement. Expert Opin Ther Pat 22,349-354.

[1524] Xu Y, Zhang HT, O’Donnell JM (2011) Phosphodi-esterases in the central nervous system: Implications inmood and cognitive disorders. Handb Exp Pharmacol 447-485.

[1525] Sierksma AS, Rutten K, Sydlik S, Rostamian S, Stein-busch HW, van den Hove DL, Prickaerts J (2013) Chronicphosphodiesterase type 2 inhibition improves memory inthe APPswe/PS1dE9 mouse model of Alzheimer’s disease.Neuropharmacology 61, 124-136.

[1526] Burgin AB, Magnusson OT, Singh J, Witte P, Staker BL,Bjornsson JM, Thorsteinsdottir M, Hrafnsdottir S, HagenT, Kiselyov AS, Stewart LJ, Gurney ME (2010) Design ofphosphodiesterase 4D (PDE4D) allosteric modulators forenhancing cognition with improved safety. Nat Biotechnol28, 63-70.

[1527] Ghavami A, Hirst WD, Novak TJ (2006) Selective phos-phodiesterase (PDE)-4 inhibitors: A novel approach totreating memory deficit? Drugs R D 7, 63-71.

[1528] Prickaerts J, Sik A, van Staveren WC, Koopmans G, Stein-busch HW, van der Staay FJ, de Vente J, Blokland A (2004)Phosphodiesterase type 5 inhibition improves early mem-

ory consolidation of object information. Neurochem Int 45,915-928.

[1529] Reneerkens OA, Rutten K, Akkerman S, Blokland A, Shaf-fer CL, Menniti FS, Steinbusch HW, Prickaerts J (2012)Phosphodiesterase type 5 (PDE5) inhibition improvesobject recognition memory: Indications for central andperipheral mechanisms. Neurobiol Learn Mem 97, 370-379.

[1530] Chappie T, Humphrey J, Menniti F, Schmidt C (2009)PDE10A inhibitors: An assessment of the current CNSdrug discovery landscape. Curr Opin Drug Discov Devel12, 458-467.

[1531] Grauer SM, Pulito VL, Navarra RL, Kelly MP, Kelley C,Graf R, Langen B, Logue S, Brennan J, Jiang L, CharychE, Egerland U, Liu F, Marquis KL, Malamas M, HageT, Comery TA, Brandon NJ (2009) Phosphodiesterase10A inhibitor activity in preclinical models of the posi-tive, cognitive, and negative symptoms of schizophrenia.J Pharmacol Exp Ther 331, 574-590.

[1532] Williams M, Risley EA (1979) Enhancement of the bind-ing of 3H-diazepam to rat brain membranes in vitro bySQ 20009, A novel anxiolytic, gamma-aminobutyric acid(GABA) and muscimol. Life Sci 24, 833-841.

[1533] Supavilai P, Karobath M (1980) Interaction of SQ 20009and GABA-like drugs as modulators of benzodiazepinereceptor binding. Eur J Pharmacol 62, 229-233.

[1534] Ticku MK, Davis WC (1982) Molecular interactions ofetazolate with benzodiazepine and picrotoxinin bindingsites. J Neurochem 38, 1180-1182.

[1535] Barnes DM, White WF, Dichter MA (1983) Eta-zolate (SQ20009): Electrophysiology and effects on[3H]flunitrazepam binding in cultured cortical neurons. JNeurosci 3, 762-772.

[1536] Borea PA, Supavilai P, Karobath M (1983) Differentialmodulation of etazolate or pentobarbital enhanced [3H]muscimol binding by benzodiazepine agonists and inverseagonists. Brain Res 280, 383-386.

[1537] Verhoest PR, Chapin DS, Corman M, Fonseca K, HarmsJF, Hou X, Marr ES, Menniti FS, Nelson F, O’Connor R,Pandit J, Proulx-Lafrance C, Schmidt AW, Schmidt CJ,Suiciak JA, Liras S (2009) Discovery of a novel classof phosphodiesterase 10A inhibitors and identificationof clinical candidate 2-[4-(1-methyl-4-pyridin-4-yl-1H-pyrazol-3-yl)-phenoxymethyl]- quinoline (PF-2545920)for the treatment of schizophrenia. J Med Chem 52, 5188-5196.

[1538] Torremans A, Ahnaou A, Van HA, Straetemans R, Geys H,Vanhoof G, Meert TF, Drinkenburg WH (2010) Effects ofphosphodiesterase 10 inhibition on striatal cyclic AMP andperipheral physiology in rats. Acta Neurobiol Exp (Wars )70, 13-19.

[1539] Dedeurwaerdere S, Wintmolders C, Vanhoof G, LangloisX (2011) Patterns of brain glucose metabolism inducedby phosphodiesterase 10A inhibitors in the mouse: Apotential translational biomarker. J Pharmacol Exp Ther339, 210-217.

[1540] Malamas MS, Ni Y, Erdei J, Stange H, Schindler R, LankauHJ, Grunwald C, Fan KY, Parris K, Langen B, Egerland U,Hage T, Marquis KL, Grauer S, Brennan J, Navarra R, GrafR, Harrison BL, Robichaud A, Kronbach T, Pangalos MN,Hoefgen N, Brandon NJ (2011) Highly potent, selective,and orally active phosphodiesterase 10A inhibitors. J MedChem 54, 7621-7638.

[1541] Hu E, Ma J, Biorn C, Lester-Zeiner D, Cho R, Rumfelt S,Kunz RK, Nixey T, Michelsen K, Miller S, Shi J, Wong

Page 107: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 653

J, Hill Della PG, Able J, Talreja S, Hwang DR, Hitch-cock SA, Porter A, Immke D, Allen JR, Treanor J, ChenH (2012) Rapid identification of a novel small moleculephosphodiesterase 10A (PDE10A) tracer. J Med Chem 55,4776-4787.

[1542] Hu E, Kunz RK, Rumfelt S, Chen N, Burli R, Li C,Andrews KL, Zhang J, Chmait S, Kogan J, Lindstrom M,Hitchcock SA, Treanor J (2012) Discovery of potent, selec-tive, and metabolically stable 4-(pyridin-3-yl)cinnolinesas novel phosphodiesterase 10A (PDE10A) inhibitors.Bioorg Med Chem Lett 22, 2262-2265.

[1543] Lee HR, Park SY, Kim HY, Shin HK, Lee WS, RhimBY, Hong KW, Kim CD (2012) Protection by cilostazolagainst amyloid-beta(1-40)-induced suppression of viabil-ity and neurite elongation through activation of CK2alphain HT22 mouse hippocampal cells. J Neurosci Res 90,1566-1576.

[1544] Bruno O, Fedele E, Prickaerts J, Parker LA, Canepa E,Brullo C, Cavallero A, Gardella E, Balbi A, DomenicottiC, Bollen E, Gijselaers HJ, Vanmierlo T, Erb K, LimebeerCL, Argellati F, Marinari UM, Pronzato MA, Ricciarelli R(2011) GEBR-7b, a novel PDE4D selective inhibitor thatimproves memory in rodents at non-emetic doses. Br JPharmacol 164, 2054-2063.

[1545] Cuadrado-Tejedor M, Hervias I, Ricobaraza A, Puerta E,Perez-Roldan JM, Garcia-Barroso C, Franco R, Aguirre N,Garcia-Osta A (2011) Sildenafil restores cognitive func-tion without affecting beta-amyloid burden in a mousemodel of Alzheimer’s disease. Br J Pharmacol 164, 2029-2041.

[1546] Garcia-Barroso C, Ricobaraza A, Pascual-Lucas M, Unc-eta N, Rico AJ, Goicolea MA, Salles J, Lanciego JL,Oyarzabal J, Franco R, Cuadrado-Tejedor M, Garcia-OstaA (2013) Tadalafil crosses the blood-brain barrier andreverses cognitive dysfunction in a mouse model of AD.Neuropharmacology 61, 114-123.

[1547] Smith SM, Uslaner JM, Cox CD, Huszar SL, CannonCE, Vardigan JD, Eddins D, Toolan DM, Kandebo M,Yao L, Raheem IT, Schreier JD, Breslin MJ, ColemanPJ, Renger JJ (2013) The novel phosphodiesterase 10Ainhibitor THPP-1 has antipsychotic-like effects in rat andimproves cognition in rat and rhesus monkey. Neurophar-macology 61, 215-223.

[1548] Treves TA, Korczyn AD (1999) Denbufylline in dementia:A double-blind controlled study. Dement Geriatr CognDisord 10, 505-510.

[1549] MacDonald E, Van der LH, Pocock D, Cole C, ThomasN, VandenBerg PM, Bourtchouladze R, Kleim JA (2007)A novel phosphodiesterase type 4 inhibitor, HT-0712,enhances rehabilitation-dependent motor recovery andcortical reorganization after focal cortical ischemia.Neurorehabil Neural Repair 21, 486-496.

[1550] Giembycz MA (2008) Can the anti-inflammatory poten-tial of PDE4 inhibitors be realized: Guarded optimism orwishful thinking? Br J Pharmacol 155, 288-290.

[1551] Gallant M, Aspiotis R, Day S, Dias R, Dube D, DubeL, Friesen RW, Girard M, Guay D, Hamel P, Huang Z,Lacombe P, Laliberte S, Levesque JF, Liu S, MacDonald D,Mancini J, Nicholson DW, Styhler A, Townson K, WatersK, Young RN, Girard Y (2010) Discovery of MK-0952,a selective PDE4 inhibitor for the treatment of long-termmemory loss and mild cognitive impairment. Bioorg MedChem Lett 20, 6387-6393.

[1552] Hersperger R, Bray-French K, Mazzoni L, Muller T (2000)Palladium-catalyzed cross-coupling reactions for the syn-

thesis of 6, 8-disubstituted 1,7-naphthyridines: A novelclass of potent and selective phosphodiesterase type 4Dinhibitors. J Med Chem 43, 675-682.

[1553] Hersperger R, Dawson J, Mueller T (2002) Synthesisof 4-(8-benzo[1,2,5]oxadiazol-5-yl-[1,7]naphthyridine-6-yl)-benzoic acid: A potent and selective phosphodiesterasetype 4D inhibitor. Bioorg Med Chem Lett 12, 233-235.

[1554] Trifilieff A, Wyss D, Walker C, Mazzoni L, HerspergerR (2002) Pharmacological profile of a novel phos-phodiesterase 4 inhibitor, 4-(8-benzo[1,2,5]oxadiazol-5-yl-[1,7]naphthyridin-6-yl)-benzoic acid (NVP-ABE171),a 1,7-naphthyridine derivative, with anti-inflammatoryactivities. J Pharmacol Exp Ther 301, 241-248.

[1555] Hutson PH, Finger EN, Magliaro BC, Smith SM,Converso A, Sanderson PE, Mullins D, Hyde LA,Eschle BK, Turnbull Z, Sloan H, Guzzi M, ZhangX, Wang A, Rindgen D, Mazzola R, Vivian JA, EddinsD, Uslaner JM, Bednar R, Gambone C, Le-Mair W,Marino MJ, Sachs N, Xu G, Parmentier-Batteur S (2011)The selective phosphodiesterase 9 (PDE9) inhibitor PF-04447943(6-[(3S,4S)-4-methyl-1-(pyrimidin-2-ylmethyl)pyrrolidin-3-yl]-1-(tetrahydro-2H-py ran-4-yl)- 1,5-dihydro-4H-pyrazolo[3,4-d]pyrimidin-4-one) enhancessynaptic plasticity and cognitive function in rodents.Neuropharmacology 61, 665-676.

[1556] Verhoest PR, Fonseca KR, Hou X, Proulx-LafranceC, Corman M, Helal CJ, Claffey MM, Tuttle JB,Coffman KJ, Liu S, Nelson F, Kleiman RJ, Menniti FS,Schmidt CJ, Vanase-Frawley M, Liras S (2012) Designand discovery of 6-[(3S,4S)-4-Methyl-1-(pyrimidin-2-ylmethyl)pyrrolidin-3-yl]-1-(tetrahydro-2H-pyr an-4-yl)-1,5-dihydro-4H-pyrazolo[3,4-d]pyrimidin-4-one (PF-04447943), a selective brain penetrant PDE9A inhibitorfor the treatment of cognitive disorders. J Med Chem, doi:10.1021/jm3007799 [Epubl ahead of print].

[1557] Plaschke K, Martin E, Bardenheuer HJ (1998) Effect ofpropentofylline on hippocampal brain energy state andamyloid precursor protein concentration in a rat modelof cerebral hypoperfusion. J Neural Transm 105, 1065-1077.

[1558] Yamada K, Tanaka T, Senzaki K, Kameyama T, NabeshimaT (1998) Propentofylline improves learning and memorydeficits in rats induced by beta-amyloid protein-(1-40). EurJ Pharmacol 349, 15-22.

[1559] Rutten K, Prickaerts J, Blokland A (2006) Rolipramreverses scopolamine-induced and time-dependent mem-ory deficits in object recognition by different mechanismsof action. Neurobiol Learn Mem 85, 132-138.

[1560] Rutten K, Prickaerts J, Schaenzle G, Rosenbrock H, Blok-land A (2008) Sub-chronic rolipram treatment leads toa persistent improvement in long-term object memory inrats. Neurobiol Learn Mem 90, 569-575.

[1561] Rutten K, Basile JL, Prickaerts J, Blokland A, Vivian JA(2008) Selective PDE inhibitors rolipram and sildenafilimprove object retrieval performance in adult cynomolgusmacaques. Psychopharmacology (Berl) 196, 643-648.

[1562] Rutten K, Wallace TL, Works M, Prickaerts. J, BloklandA, Novak TJ, Santarelli L, Misner DL (2011) Enhancedlong-term depression and impaired reversal learning inphosphodiesterase 4B-knockout (PDE4B(−/−)) mice.Neuropharmacology 61, 138-147.

[1563] de Lima MN, Presti-Torres J, Garcia VA, Guimaraes MR,Scalco FS, Roesler R, Schroder N (2008) Ameliorationof recognition memory impairment associated with ironloading or aging by the type 4-specific phosphodiesterase

Page 108: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

654 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

inhibitor rolipram in rats. Neuropharmacology 55, 788-792.

[1564] Kanes SJ, Tokarczyk J, Siegel SJ, Bilker W, Abel T,Kelly MP (2007) Rolipram: A specific phosphodiesterase4 inhibitor with potential antipsychotic activity. Neuro-science 144, 239-246.

[1565] Roesler R, Schroder N (2011) Cognitive enhancers: Focuson modulatory signaling influencing memory consolida-tion. Pharmacol Biochem Behav 99, 155-163.

[1566] Wang C, Yang XM, Zhuo YY, Zhou H, Lin HB, ChengYF, Xu JP, Zhang HT (2012) The phosphodiesterase-4 inhibitor rolipram reverses Abeta-induced cogni-tive impairment and neuroinflammatory and apoptoticresponses in rats. Int J Neuropsychopharmacol 15, 749-766.

[1567] Sanchez-Mejia RO, Newman JW, Toh S, Yu GQ, Zhou Y,Halabisky B, Cisse M, Scearce-Levie K, Cheng IH, Gan L,Palop JJ, Bonventre JV, Mucke L (2008) Phospholipase A2reduction ameliorates cognitive deficits in a mouse modelof Alzheimer’s disease. Nat Neurosci 11, 1311-1318.

[1568] Sanchez-Mejia RO, Mucke L (2010) Phospholipase A2and arachidonic acid in Alzheimer’s disease. Biochim Bio-phys Acta 1801, 784-790.

[1569] Gentile MT, Reccia MG, Sorrentino PP, Vitale E, Sor-rentino G, Puca AA, Colucci-D’Amato L (2012) Roleof cytosolic calcium-dependent phospholipase A2 inAlzheimer’s disease pathogenesis. Mol Neurobiol 45, 596-604.

[1570] Mancuso DJ, Kotzbauer P, Wozniak DF, Sims HF, JenkinsCM, Guan S, Han X, Yang K, Sun G, Malik I, Cony-ers S, Green KG, Schmidt RE, Gross RW (2009) Geneticablation of calcium-independent phospholipase A2gammaleads to alterations in hippocampal cardiolipin content andmolecular species distribution, mitochondrial degenera-tion, autophagy, and cognitive dysfunction. J Biol Chem284, 35632-35644.

[1571] Oliveira TG, Chan RB, Tian H, Laredo M, Shui G,Staniszewski A, Zhang H, Wang L, Kim TW, Duff KE,Wenk MR, Arancio O, Di PG (2010) Phospholipase d2ablation ameliorates Alzheimer’s disease-linked synapticdysfunction and cognitive deficits. J Neurosci 30, 16419-16428.

[1572] Oliveira TG, Di Pado G (2010) Phospholipase D in brainfunction and Alzheimer’s disease. Biochim Biophys Acta1801, 799-805.

[1573] Cesari M, Pahor M, Incalzi RA (2010) Plasminogenactivator inhibitor-1 (PAI-1): A key factor linking fibri-nolysis and age-related subclinical and clinical conditions.Cardiovasc Ther 28, e72-e91.

[1574] Weng SC, Shu KH, Tang YJ, Sheu WH, Tarng DC, WuMJ, Chen YM, Chuang YW (2012) Progression of cogni-tive dysfunction in elderly chronic kidney disease patientsin a veteran’s institution in central Taiwan: A 3-year lon-gitudinal study. Intern Med 51, 29-35.

[1574] [7]Cho H, Joo Y, Kim S, Woo RS, Lee SH, Kim HS (2012)Plasminogen activator inhibitor-1 promotes synaptogene-sis and protects against Abeta(1-42)-induced neurotoxicityin primary cultured hippocampal neurons. Int J Neurosci,doi:10.3109/00207454.2012.724127-

[1575] Miralbell J, Soriano JJ, Spulber G, Lopez-Cancio E, Are-nillas JF, Bargallo N, Galan A, Barrios MT, Caceres C,Alzamora MT, Pera G, Kivipelto M, Wahlund LO, Dava-los A, Mataro M (2012) Structural brain changes andcognition in relation to markers of vascular dysfunction.Neurobiol Aging 33, 1003-1017.

[1576] Quinn TJ, Gallacher J, Deary IJ, Lowe GD, Fenton C,Stott DJ (2011) Association between circulating hemo-static measures and dementia or cognitive impairment:Systematic review and meta-analyzes. J Thromb Haemost9, 1475-1482.

[1577] Martorana A, Sancesario GM, Esposito Z, Nuccetelli M,Sorge R, Formosa A, Dinallo V, Bernardi G, BernardiniS, Sancesario G (2012) Plasmin system of Alzheimer’sdisease patients: CSF analysis. J Neural Transm 119, 763-769.

[1578] Suh SW, Aoyama K, Chen Y, Garnier P, MatsumoriY, Gum E, Liu J, Swanson RA (2003) Hypoglycemicneuronal death and cognitive impairment are preventedby poly(ADP-ribose) polymerase inhibitors administeredafter hypoglycemia. J Neurosci 23, 10681-10690.

[1579] Suh SW, Aoyama K, Matsumori Y, Liu J, Swanson RA(2005) Pyruvate administered after severe hypoglycemiareduces neuronal death and cognitive impairment. Dia-betes 54, 1452-1458.

[1580] Whalen MJ, Clark RS, Dixon CE, Robichaud P, MarionDW, Vagni V, Graham SH, Virag L, Hasko G, StachlewitzR, Szabo C, Kochanek PM (1999) Reduction of cogni-tive and motor deficits after traumatic brain injury in micedeficient in poly(ADP-ribose) polymerase. J Cereb BloodFlow Metab 19, 835-842.

[1581] Russo AL, Kwon HC, Burgan WE, Carter D, Beam K,Weizheng X, Zhang J, Slusher BS, Chakravarti A, TofilonPJ, Camphausen K (2009) In vitro and in vivo radiosen-sitization of glioblastoma cells by the poly (ADP-ribose)polymerase inhibitor E7016. Clin Cancer Res 15, 607-612.

[1582] Rossner S, Schulz I, Zeitschel U, Schliebs R, Bigl V,Demuth HU (2005) Brain prolyl endopeptidase expressionin aging, APP transgenic mice and Alzheimer’s disease.Neurochem Res 30, 695-702.

[1583] Mannisto PT, Venalainen J, Jalkanen A, Garcia-HorsmanJA (2007) Prolyl oligopeptidase: A potential target for thetreatment of cognitive disorders. Drug News Perspect 20,293-305.

[1584] Garcia-Horsman JA (2011) Prolyl oligopeptidase in brainfunction and dysfunction. CNS Neurol Disord Drug Target10, 296.

[1585] Lopez A, Tarrago T, Giralt E (2011) Low molecular weightinhibitors of prolyl oligopeptidase: A review of com-pounds patented from 2003 to 2010. Expert Opin TherPat 21, 1023-1044.

[1586] Jalkanen AJ, Hakkarainen JJ, Lehtonen M, VenalainenT, Kaariainen TM, Jarho E, Suhonen M, Forsberg MM(2011) Brain pharmacokinetics of two prolyl oligopepti-dase inhibitors, JTP-4819 and KYP-2047, in the rat. BasicClin Pharmacol Toxicol 109, 443-451.

[1587] Jalkanen AJ, Savolainen K, Forsberg MM (2011) Inhibi-tion of prolyl oligopeptidase by KYP-2047 fails to increasethe extracellular neurotensin and substance P levels in ratstriatum. Neurosci Lett 502, 107-111.

[1588] Jalkanen AJ, Piepponen TP, Hakkarainen JJ, De MeesterI, Lambeir AM, Forsberg MM (2012) The effect of pro-lyl oligopeptidase inhibition on extracellular acetylcholineand dopamine levels in the rat striatum. Neurochem Int 60,301-309.

[1589] Park DH, Park SJ, Kim JM, Jung WY, Ryu JH (2010)Subchronic administration of rosmarinic acid, a naturalprolyl oligopeptidase inhibitor, enhances cognitive perfor-mances. Fitoterapia 81, 644-648.

[1590] Kamei H, Ueki T, Obi Y, Fukagawa Y, Oki T (1992)Protective effect of eurystatins A and B, new prolyl

Page 109: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 655

endopeptidase inhibitors, on scopolamine-induced amne-sia in rats. Jpn J Pharmacol 60, 377-380.

[1591] Toda S, Obi Y, Numata K, Hamagishi Y, Tomita K,Komiyama N, Kotake C, Furumai T, Oki T (1992) Eurys-tatins A and B, new prolyl endopeptidase inhibitors. I.Taxonomy, production, isolation and biological activities.J Antibiot (Tokyo) 45, 1573-1579.

[1592] Toda S, Kotake C, Tsuno T, Narita Y, Yamasaki T, KonishiM (1992) Eurystatins A and B, new prolyl endopeptidaseinhibitors. II. Physico-chemical properties and structuredetermination. J Antibiot (Tokyo) 45, 1580-1586.

[1593] Suzuki K, Toda S, Furumai T, Fukagawa Y, Oki T (1994)Eurystatins A and B, new prolyl endopeptidase inhibitors.III. Fermentation and controlled biosynthesis of eurystatinanalogs by Streptomyces eurythermus. J Antibiot (Tokyo)47, 982-991.

[1594] Toide K, Iwamoto Y, Fujiwara T, Abe H (1995) JTP-4819:A novel prolyl endopeptidase inhibitor with potential asa cognitive enhancer. J Pharmacol Exp Ther 274, 1370-1378.

[1595] Toide K, Shinoda M, Iwamoto Y, Fujiwara T, Okamiya K,Uemura A (1997) A novel prolyl endopeptidase inhibitor,JTP-4819, with potential for treating Alzheimer’s disease.Behav Brain Res 83, 147-151.

[1596] Toide K, Shinoda M, Fujiwara T, Iwamoto Y (1997) Effectof a novel prolyl endopeptidase inhibitor, JTP-4819, onspatial memory and central cholinergic neurons in agedrats. Pharmacol Biochem Behav 56, 427-434.

[1597] Toide K, Shinoda M, Miyazaki A (1998) A novel pro-lyl endopeptidase inhibitor, JTP-4819–its behavioral andneurochemical properties for the treatment of Alzheimer’sdisease. Rev Neurosci 9, 17-29.

[1598] Wallen EA, Christiaans JA, Jarho EM, Forsberg MM,Venalainen JI, Mannisto PT, Gynther J (2003) New pro-lyl oligopeptidase inhibitors developed from dicarboxylicacid bis(l-prolyl-pyrrolidine) amides. J Med Chem 46,4543-4551.

[1599] Katsube N, Sunaga K, Chuang DM, Ishitani R (1996)ONO-1603, a potential antidementia drug, shows neuro-protective effects and increases m3-muscarinic receptormRNA levels in differentiating rat cerebellar granule neu-rons. Neurosci Lett 214, 151-154.

[1600] Katsube N, Sunaga K, Aishita H, Chuang DM, Ishitani R(1999) ONO-1603, a potential antidementia drug, delaysage-induced apoptosis and suppresses overexpression ofglyceraldehyde-3-phosphate dehydrogenase in culturedcentral nervous system neurons. J Pharmacol Exp Ther288, 6-13.

[1601] Barelli H, Petit A, Hirsch E, Wilk S, De Nanteuil G, MorainP, Checler F (1999) S 17092-1, a highly potent, specific andcell permeant inhibitor of human proline endopeptidase.Biochem Biophys Res Commun 257, 657-661.

[1602] Morain P, Robin JL, De Nanteuil G, Jochemsen R, HeidetV, Guez D (2000) Pharmacodynamic and pharmacokineticprofile of S 17092, a new orally active prolyl endopeptidaseinhibitor, in elderly healthy volunteers. A phase I study. BrJ Clin Pharmacol 50, 350-359.

[1603] Morain P, Lestage P, De Nanteuil G, Jochemsen R, RobinJL, Guez D, Boyer PA (2002) S 17092: A prolyl endopep-tidase inhibitor as a potential therapeutic drug for memoryimpairment. Preclinical and clinical studies. CNS Drug Rev8, 31-52.

[1604] Morain P, Boeijinga PH, Demazieres A, De Nanteuil G,Luthringer R (2007) Psychotropic profile of S 17092, aprolyl endopeptidase inhibitor, using quantitative EEG in

young healthy volunteers. Neuropsychobiology 55, 176-183.

[1605] Bellemere G, Morain P, Vaudry H, Jegou S (2003) Effectof S 17092, a novel prolyl endopeptidase inhibitor, onsubstance P and alpha-melanocyte-stimulating hormonebreakdown in the rat brain. J Neurochem 84, 919-929.

[1606] Petit A, Barelli H, Morain P, Checler F (2000) Novel pro-line endopeptidase inhibitors do not modify Abeta40/42formation and degradation by human cells expressingwild-type and swedish mutated beta-amyloid precursorprotein. Br J Pharmacol 130, 1613-1617.

[1607] Arai H, Nishioka H, Niwa S, Yamanaka T, Tanaka Y,Yoshinaga K, Kobayashi N, Miura N, Ikeda Y (1993) Syn-thesis of prolyl endopeptidase inhibitors and evaluation oftheir structure-activity relationships: In vitro inhibition ofprolyl endopeptidase from canine brain. Chem Pharm Bull(Tokyo) 41, 1583-1588.

[1608] Tanaka Y, Niwa S, Nishioka H, Yamanaka T, TorizukaM, Yoshinaga K, Kobayashi N, Ikeda Y, Arai H (1994)New potent prolyl endopeptidase inhibitors: Synthesis andstructure-activity relationships of indan and tetralin deriva-tives and their analogues. J Med Chem 37, 2071-2078.

[1609] Portevin B, Benoist A, Remond G, Herve Y, VincentM, Lepagnol J, De Nanteuil G (1996) New prolylendopeptidase inhibitors: In vitro and in vivo activi-ties of azabicyclo[2.2.2]octane, azabicyclo[2.2.1]heptane,and perhydroindole derivatives. J Med Chem 39, 2379-2391.

[1610] Jarho EM, Venalainen JI, Poutiainen S, Leskinen H,Vepsalainen J, Christiaans JA, Forsberg MM, MannistoPT, Wallen EA (2007) 2(S)-(Cycloalk-1-enecarbonyl)-1-(4-phenyl-butanoyl)pyrrolidines and 2(S)-(aroyl)-1-(4-phenylbutanoyl)pyrrolidines as prolyl oligopeptidaseinhibitors. Bioorg Med Chem 15, 2024-2031.

[1611] Nakajima T, Ono Y, Kato A, Maeda J, Ohe T (1992) Y-29794–a non-peptide prolyl endopeptidase inhibitor thatcan penetrate into the brain. Neurosci Lett 141, 156-160.

[1612] Kato A, Fukunari A, Sakai Y, Nakajima T (1997)Prevention of amyloid-like deposition by a selectiveprolyl endopeptidase inhibitor, Y-29794, in senescence-accelerated mouse. J Pharmacol Exp Ther 283, 328-335.

[1613] Miura N, Shibata S, Watanabe S (1995) Increase in the sep-tal vasopressin content by prolyl endopeptidase inhibitorsin rats. Neurosci Lett 196, 128-130.

[1614] Miura N, Shibata S, Watanabe S (1997) Z-321, a pro-lyl endopeptidase inhibitor, augments the potentiation ofsynaptic transmission in rat hippocampal slices. BehavBrain Res 83, 213-216.

[1615] Umemura K, Kondo K, Ikeda Y, Nishimoto M, HiragaY, Yoshida Y, Nakashima M (1999) Pharmacokineticsand safety of Z-321, a novel specific orally active pro-lyl endopeptidase inhibitor, in healthy male volunteers.J Clin Pharmacol 39, 462-470.

[1616] Shishido Y, Furushiro M, Tanabe S, Taniguchi A,Hashimoto S, Yokokura T, Shibata S, Yamamoto T, Watan-abe S (1998) Effect of ZTTA, a prolyl endopeptidaseinhibitor, on memory impairment in a passive avoidancetest of rats with basal forebrain lesions. Pharm Res 15,1907-1910.

[1617] Urade Y, Hayaishi O (2000) Biochemical, structural,genetic, physiological, and pathophysiological features oflipocalin-type prostaglandin D synthase. Biochim BiophysActa 1482, 259-271.

[1618] Urade Y, Hayaishi O (2000) Prostaglandin D synthase:Structure and function. Vitam Horm 58, 89-120.

Page 110: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

656 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

[1619] Hara S, Kamei D, Sasaki Y, Tanemoto A, Nakatani Y,Murakami M (2010) Prostaglandin E synthases: Under-standing their pathophysiological roles through mousegenetic models. Biochimie 92, 651-659.

[1620] Iyer JP, Srivastava PK, Dev R, Dastidar SG, Ray A (2009)Prostaglandin E(2) synthase inhibition as a therapeutictarget. Expert Opin Ther Targets 13, 849-865.

[1621] Matousek SB, Hein AM, Shaftel SS, Olschowka JA,Kyrkanides S, O’Banion MK (2010) Cyclooxygenase-1 mediates prostaglandin E(2) elevation and contextualmemory impairment in a model of sustained hippocampalinterleukin-1beta expression. J Neurochem 114, 247-258.

[1622] Kuroki Y, Sasaki Y, Kamei D, Akitake Y, Takahashi M,Uematsu S, Akira S, Nakatani Y, Kudo I, Hara S (2012)Deletion of microsomal prostaglandin E synthase-1 pro-tects neuronal cells from cytotoxic effects of beta-amyloidpeptide fragment 31-35. Biochem Biophys Res Commun424, 409-413.

[1623] Hongpaisan J, Alkon DL (2007) A structural basis forenhancement of long-term associative memory in singledendritic spines regulated by PKC. Proc Natl Acad Sci US A 104, 19571-19576.

[1624] Hongpaisan J, Sun MK, Alkon DL (2011) PKC epsilonactivation prevents synaptic loss, Abeta elevation, andcognitive deficits in Alzheimer’s disease transgenic mice.J Neurosci 31, 630-643.

[1625] Sun MK, Alkon DL (2006) Protein kinase C pharmacol-ogy: Perspectives on therapeutic potentials as antidementicand cognitive agents. Recent Pat CNS Drug Discov 1, 147-156.

[1626] Sun MK, Alkon DL (2010) Pharmacology of proteinkinase C activators: Cognition-enhancing and antidemen-tic therapeutics. Pharmacol Ther 127, 66-77.

[1627] Sun MK, Alkon DL (2009) Protein kinase C activatorsas synaptogenic and memory therapeutics. Arch Pharm(Weinheim) 342, 689-698.

[1628] Sun MK, Alkon DL (2005) Protein kinase C isozymes:Memory therapeutic potential. Curr Drug Targets CNSNeurol Disord 4, 541-552.

[1629] Govoni S, Amadio M, Battaini F, Pascale A (2010) Senes-cence of the brain: Focus on cognitive kinases. Curr PharmDes 16, 660-671.

[1630] Kozikowski AP, Nowak I, Petukhov PA, EtcheberrigarayR, Mohamed A, Tan M, Lewin N, Hennings H, Pearce LL,Blumberg PM (2003) New amide-bearing benzolactam-based protein kinase C modulators induce enhancedsecretion of the amyloid precursor protein metabolite sAP-Palpha. J Med Chem 46, 364-373.

[1631] Lee J, Kang JH, Han KC, Kim Y, Kim SY, Youn HS, Mook-Jung I, Kim H, Lo Han JH, Ha HJ, Kim YH, Marquez VE,Lewin NE, Pearce LV, Lundberg DJ, Blumberg PM (2006)Branched diacylglycerol-lactones as potent protein kinaseC ligands and alpha-secretase activators. J Med Chem 49,2028-2036.

[1632] Mach UR, Lewin NE, Blumberg PM, Kozikowski AP(2006) Synthesis and pharmacological evaluation of 8- and9-substituted benzolactam-v8 derivatives as potent ligandsfor protein kinase C, a therapeutic target for Alzheimer’sdisease. ChemMedChem 1, 307-314.

[1633] Sun MK, Alkon DL (2005) Dual effects of bryostatin-1on spatial memory and depression. Eur J Pharmacol 512,43-51.

[1634] Sun MK, Alkon DL (2009) Protein kinase C activatorsas synaptogenic and memory therapeutics. Arch Pharm(Weinheim) 342, 689-698.

[1635] Sun MK, Alkon DL (2010) Pharmacology of proteinkinase C activators: Cognition-enhancing and antidemen-tic therapeutics. Pharmacol Ther 127, 66-77.

[1636] Sun MK, Hongpaisan J, Nelson TJ, Alkon DL (2008) Post-stroke neuronal rescue and synaptogenesis mediated invivo by protein kinase C in adult brains. Proc Natl AcadSci U S A 105, 13620-13625.

[1637] Sun MK, Hongpaisan J, Alkon DL (2009) PostischemicPKC activation rescues retrograde and anterograde long-term memory. Proc Natl Acad Sci U S A 106, 14676-14680.

[1638] Tanaka A, Nishizaki T (2003) The newly synthesizedlinoleic acid derivative FR236924 induces a long-lastingfacilitation of hippocampal neurotransmission by target-ing nicotinic acetylcholine receptors. Bioorg Med ChemLett 13, 1037-1040.

[1639] Kanno T, Yaguchi T, Yamamoto S, Yamamoto H, FujikawaH, Nagata T, Tanaka A, Nishizaki T (2005) 8-[2-(2-pentyl-cyclopropylmethyl)-cyclopropyl]-octanoic acidstimulates GABA release from interneurons projecting toCA1 pyramidal neurons in the rat hippocampus via pre-synaptic alpha7 acetylcholine receptors. J Neurochem 95,695-702.

[1640] Kanno T, Yamamoto H, Yaguchi T, Hi R, Mukasa T,Fujikawa H, Nagata T, Yamamoto S, Tanaka A, NishizakiT (2006) The linoleic acid derivative DCP-LA selectivelyactivates PKC-epsilon, possibly binding to the phos-phatidylserine binding site. J Lipid Res 47, 1146-1156.

[1641] Yaguchi T, Nagata T, Mukasa T, Fujikawa H, Yamamoto H,Yamamoto S, Iso H, Tanaka A, Nishizaki T (2006) Linoleicacid derivative DCP-LA improves learning impairment inSAMP8. Neuroreport 17, 105-108.

[1642] Fitzpatrick CJ, Lombroso PJ (2011) The Role ofStriatal-Enriched Protein Tyrosine Phosphatase (STEP) inCognition. Front Neuroanat 5, 47.

[1643] Pelov I, Teltsh O, Greenbaum L, Rigbi A, Kanyas-SarnerK, Lerer B, Lombroso P, Kohn Y (2012) Involvementof PTPN5, the gene encoding the striatal-enriched pro-tein tyrosine phosphatase, in schizophrenia and cognition.Psychiatr Genet 22, 168-176.

[1644] Leblanc M, Kulle B, Sundet K, Agartz I, Melle I, DjurovicS, Frigessi A, Andreassen OA (2012) Genome-wide studyidentifies PTPRO and WDR72 and FOXQ1-SUMO1P1interaction associated with neurocognitive function. J Psy-chiatr Res 46, 271-278.

[1645] Patrignani C, Magnone MC, Tavano P, Ardizzone M,Muzio V, Greco B, Zaratin PF (2008) Knockout micereveal a role for protein tyrosine phosphatase H1 in cog-nition. Behav Brain Funct 4, 36.

[1646] Raz L, Zhang QG, Zhou CF, Han D, Gulati P, YangLC, Yang F, Wang RM, Brann DW (2010) Role ofRac1 GTPase in NADPH oxidase activation and cognitiveimpairment following cerebral ischemia in the rat. PLoSOne 5, e12606.

[1647] Bongmba OY, Martinez LA, Elhardt ME, Butler K, Tejada-Simon MV (2011) Modulation of dendritic spines andsynaptic function by Rac1: A possible link to Fragile Xsyndrome pathology. Brain Res 1399, 79-95.

[1648] Ramakers GJ, Wolfer D, Rosenberger G, KuchenbeckerK, Kreienkamp HJ, Prange-Kiel J, Rune G, Richter K,Langnaese K, Masneuf S, Bosl MR, Fischer KD, KrugersHJ, Lipp HP, van GE, Kutsche K (2012) Dysregulationof Rho GTPases in the alphaPix/Arhgef6 mouse model ofX-linked intellectual disability is paralleled by impairedstructural and synaptic plasticity and cognitive deficits.Hum Mol Genet 21, 268-286.

Page 111: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes 657

[1649] Borrelli S, Musilli M, Martino A, Diana G (2013)Long-lasting efficacy of the cognitive enhancer CytotoxicNecrotizing Factor 1. Neuropharmacology 61, 74-80.

[1650] Desire L, Bourdin J, Loiseau N, Peillon H, Picard V, DeOliveira C, Bachelot F, Leblond B, Taverne T, BeausoleilE, Lacombe S, Drouin D, Schweighoffer F (2005) RAC1inhibition targets amyloid precursor protein processing bygamma-secretase and decreases Abeta production in vitroand in vivo. J Biol Chem 280, 37516-37525.

[1651] Shutes A, Onesto C, Picard V, Leblond B, SchweighofferF, Der CJ (2007) Specificity and mechanism of action ofEHT 1864, a novel small molecule inhibitor of Rac familysmall GTPases. J Biol Chem 282, 35666-35678.

[1652] Onesto C, Shutes A, Picard V, Schweighoffer F, DerCJ (2008) Characterization of EHT 1864, a novel smallmolecule inhibitor of Rac family small GTPases. MethodsEnzymol 439, 111-129.

[1653] Mans RA, McMahon LL, Li L (2012) Simvastatin-mediated enhancement of long-term potentiation is drivenby farnesyl-pyrophosphate depletion and inhibition of far-nesylation. Neuroscience 202, 1-9.

[1654] Gagnon KT (2010) HD Therapeutics - CHDI Fifth AnnualConference. IDrugs 13, 219-223.

[1655] Ho PI, Collins SC, Dhitavat S, Ortiz D, Ashline D, RogersE, Shea TB (2001) Homocysteine potentiates beta-amyloidneurotoxicity: Role of oxidative stress. J Neurochem 78,249-253.

[1656] Shea TB, Ashline D, Ortiz D, Milhalik S, Rogers E(2004) The S-adenosyl homocysteine hydrolase inhibitor3-deaza-adenosine prevents oxidative damage and cogni-tive impairment following folate and vitamin E deprivationin a murine model of age-related, oxidative stress-induced neurodegeneration. Neuromolecular Med 5, 171-180.

[1657] Anekonda TS, Reddy PH (2006) Neuronal protection bysirtuins in Alzheimer’s disease. J Neurochem 96, 305-313.

[1658] Bonda DJ, Lee HG, Camins A, Pallas M, Casadesus G,Smith MA, Zhu X (2011) The sirtuin pathway in age-ing and Alzheimer disease: Mechanistic and therapeuticconsiderations. Lancet Neurol 10, 275-279.

[1659] Baur JA, Sinclair DA (2006) Therapeutic potential ofresveratrol: The in vivo evidence. Nat Rev Drug Discov5, 493-506.

[1660] Baur JA, Pearson KJ, Price NL, Jamieson HA, Lerin C,Kalra A, Prabhu VV, Allard JS, Lopez-Lluch G, Lewis K,Pistell PJ, Poosala S, Becker KG, Boss O, Gwinn D, WangM, Ramaswamy S, Fishbein KW, Spencer RG, Lakatta EG,Le Couteur D, Shaw RJ, Navas P, Puigserver P, IngramDK, de Cabo R, Sinclair DA (2006) Resveratrol improveshealth and survival of mice on a high-calorie diet. Nature444, 337-342.

[1661] Baur JA (2010) Resveratrol, sirtuins, and the promise of aDR mimetic. Mech Ageing Dev 131, 261-269.

[1662] Baur JA, Chen D, Chini EN, Chua K, Cohen HY, de CaboR, Deng C, Dimmeler S, Gius D, Guarente LP, HelfandSL, Imai S, Itoh H, Kadowaki T, Koya D, LeeuwenburghC, McBurney M, Nabeshima Y, Neri C, Oberdoerffer P,Pestell RG, Rogina B, Sadoshima J, Sartorelli V, SerranoM, Sinclair DA, Steegborn C, Tatar M, Tissenbaum HA,Tong Q, Tsubota K, Vaquero A, Verdin E (2010) Dietaryrestriction: Standing up for sirtuins. Science 329, 1012-1013.

[1663] Kennedy DO, Wightman EL, Reay JL, Lietz G, OkelloEJ, Wilde A, Haskell CF (2010) Effects of resveratrol oncerebral blood flow variables and cognitive performance

in humans: A double-blind, placebo-controlled, crossoverinvestigation. Am J Clin Nutr 91, 1590-1597.

[1664] Quideau S, Deffieux D, Pouysegu L (2012) Resveratrolstill has something to say about aging! Angew Chem IntEd Engl 51, 6824-6826.

[1665] Marambaud P, Zhao H, Davies P (2005) Resveratrolpromotes clearance of Alzheimer’s disease amyloid-betapeptides. J Biol Chem 280, 37377-37382.

[1666] Albani D, Polito L, Batelli S, De MS, Fracasso C, MartelliG, Colombo L, Manzoni C, Salmona M, Caccia S, NegroA, Forloni G (2009) The SIRT1 activator resveratrol pro-tects SK-N-BE cells from oxidative stress and againsttoxicity caused by alpha-synuclein or amyloid-beta (1-42)peptide. J Neurochem 110, 1445-1456.

[1667] Albani D, Polito L, Forloni G (2010) Sirtuins as noveltargets for Alzheimer’s disease and other neurodegen-erative disorders: Experimental and genetic evidence.J Alzheimers Dis 19, 11-26.

[1668] Albani D, Polito L, Signorini A, Forloni G (2010)Neuroprotective properties of resveratrol in different neu-rodegenerative disorders. Biofactors 36, 370-376.

[1669] Ladiwala AR, Lin JC, Bale SS, Marcelino-Cruz AM, Bhat-tacharya M, Dordick JS, Tessier PM (2010) Resveratrolselectively remodels soluble oligomers and fibrils of amy-loid Abeta into off-pathway conformers. J Biol Chem 285,24228-24237.

[1670] Beher D, Wu J, Cumine S, Kim KW, Lu SC, AtanganL, Wang M (2009) Resveratrol is not a direct activa-tor of SIRT1 enzyme activity. Chem Biol Drug Des 74,619-624.

[1671] Jeon BT, Jeong EA, Shin HJ, Lee Y, Lee DH, Kim HJ,Kang SS, Cho GJ, Choi WS, Roh GS (2012) Resvera-trol attenuates obesity-associated peripheral and centralinflammation and improves memory deficit in mice fed ahigh-fat diet. Diabetes 61, 1444-1454.

[1672] Johnson WD, Morrissey RL, Usborne AL, KapetanovicI, Crowell JA, Muzzio M, McCormick DL (2011)Subchronic oral toxicity and cardiovascular safety phar-macology studies of resveratrol, a naturally occurringpolyphenol with cancer preventive activity. Food ChemToxicol 49, 3319-3327.

[1673] Zhang Y, Li SZ, Li J, Pan X, Cahoon RE, Jaworski JG,Wang X, Jez JM, Chen F, Yu O (2006) Using unnatu-ral protein fusions to engineer resveratrol biosynthesis inyeast and Mammalian cells. J Am Chem Soc 128, 13030-13031.

[1674] Pasinetti GM, Wang J, Marambaud P, Ferruzzi M, GregorP, Knable LA, Ho L (2011) Neuroprotective and metaboliceffects of resveratrol: Therapeutic implications for Hunt-ington’s disease and other neurodegenerative disorders.Exp Neurol 232, 1-6.

[1675] Stergiakouli E, Langley K, Williams H, Walters J,Williams NM, Suren S, Giegling I, Wilkinson LS, OwenMJ, O’Donovan MC, Rujescu D, Thapar A, Davies W(2011) Steroid sulfatase is a potential modifier of cogni-tion in attention deficit hyperactivity disorder. Genes BrainBehav 10, 334-344.

[1676] Johnson DA, Rhodes ME, Boni RL, Li PK (1997)Chronic steroid sulfatase inhibition by (p-O-sulfamoyl)-N-tetradecanoyl tyramine increases dehydroepiandrosteronesulfate in whole brain. Life Sci 61, L-9.

[1677] Li PK, Rhodes ME, Burke AM, Johnson DA (1997)Memory enhancement mediated by the steroid sulfataseinhibitor (p-O-sulfamoyl)-N-tetradecanoyl tyramine. LifeSci 60, L45-L51.

Page 112: Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with … · tal activities, including pre-attentional sensory gating, attention,learningandmemory,problemsolving,plan-ning,reasoningandjudgment,understanding,knowing

658 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes

[1678] Rhodes ME, Li PK, Burke AM, Johnson DA (1997)Enhanced plasma DHEAS, brain acetylcholine and mem-ory mediated by steroid sulfatase inhibition. Brain Res 773,28-32.

[1679] Chu GH, Peters A, Selcer KW, Li PK (1999) Synthesisand sulfatase inhibitory activities of (E)- and (Z)-4-hydroxytamoxifen sulfamates. Bioorg Med Chem Lett 9,141-144.

[1680] Wang DS, Uchikado H, Bennett DA, Schneider JA, Muf-son EJ, Wu J, Dickson DW (2008) Cognitive performance

correlates with cortical isopeptide immunoreactivity aswell as Alzheimer type pathology. J Alzheimers Dis 13,53-66.

[1681] Walters BJ, Campbell SL, Chen PC, Taylor AP, SchroederDG, Dobrunz LE, Artavanis-Tsakonas K, Ploegh HL, Wil-son JA, Cox GA, Wilson SM (2008) Differential effects ofUsp14 and Uch-L1 on the ubiquitin proteasome systemand synaptic activity. Mol Cell Neurosci 39, 539-548.